Download Vol. 23, No. 4 – Disparities in Oncology

Document related concepts

Prostate-specific antigen wikipedia , lookup

Transcript
cancercontroljournal.org Vol. 23, No. 4, October 2016
H. LEE MOFFITT CANCER CENTER & RESEARCH INSTITUTE, AN NCI COMPREHENSIVE CANCER CENTER
Barriers to Clinical Trial Enrollment in Racial and Ethnic
Minority Patients With Cancer
Racial and Ethnic Differences in the Epidemiology and
Genomics of Lung Cancer
Black Heterogeneity in Cancer Mortality: US-Blacks,
Haitians, and Jamaicans
Genomic Disparities in Breast Cancer Among Latinas
Clinical Considerations of Risk, Incidence, and Outcomes
of Breast Cancer in Sexual Minorities
Construction and Validation of a Multi-Institutional Tissue
Microarray of Invasive Ductal Carcinoma From Racially
and Ethnically Diverse Populations
Determinants of Treatment Delays Among Underserved
Hispanics With Lung and Head and Neck Cancers
Geographical Factors Associated With Health Disparities
in Prostate Cancer
Disparities in Penile Cancer
Chemoprevention in African American Men With
Prostate Cancer
Disparities in Adolescents and Young Adults With Cancer
Tobacco-Related Health Disparities Across the Cancer
Care Continuum
Cancer Control is included in
Index Medicus/MEDLINE
Editorial Board Members
Editor:
Lodovico Balducci, MD
Senior Member
Program Leader, Senior Adult Oncology Program
Moffitt Cancer Center
Deputy Editor:
Julio M. Pow-Sang, MD
Senior Member
Chair, Department of Genitourinary Oncology
Director of Moffitt Robotics Program
Moffitt Cancer Center
Editor Emeritus:
Conor C. Lynch, PhD
Assistant Member
Tumor Biology
Production Team:
Kristen J. Otto, MD
Associate Member
Head and Neck and
Endocrine Oncology
Sherri Damlo
Medical Copy Editor
Michael A. Poch, MD
Assistant Member
Genitourinary Oncology
Jeffery S. Russell, MD, PhD
Assistant Member
Endocrine Tumor Oncology
John Horton, MB, ChB
Professor Emeritus of Medicine & Oncology
Elizabeth M. Sagatys, MD
Assistant Member
Pathology - Clinical
Moffitt Cancer Center
Journal Advisory Committee:
Saïd M. Sebti, PhD
Senior Member
Drug Discovery
Daniel A. Anaya, MD
Associate Member
Gastrointestinal Oncology
Bijal D. Shah, MD
Assistant Member
Malignant Hematology
Aliyah Baluch, MD
Assistant Member
Infectious Diseases
Lubomir Sokol, MD, PhD
Senior Member
Hematology/Oncology
Dung-Tsa Chen, PhD
Associate Member
Biostatistics
Hey Sook Chon, MD
Assistant Member
Gynecological Oncology
Jasreman Dhillon, MD
Assistant Member
Pathology - Anatomic
Jennifer S. Drukteinis, MD
Associate Member
Diagnostic Radiology
Clement K. Gwede, PhD, RN
Associate Member
Health Outcomes and Behavior
Sarah E. Hoffe, MD
Associate Member
Radiation Oncology
John V. Kiluk, MD
Associate Member
Breast Oncology
Richard D. Kim, MD
Associate Member
Gastrointestinal Oncology
Bela Kis, MD, PhD
Assistant Member
Diagnostic Radiology
Rami Komrokji, MD
Associate Member
Malignant Hematology
Hatem H. Soliman, MD
Assistant Member
Breast Oncology
Jonathan R. Strosberg, MD
Assistant Member
Gastrointestinal Oncology
Veronica Nemeth
Editorial Coordinator
Diane McEnaney
Graphic Design Consultant
Associate Editor of
Educational Projects
John M. York, PharmD
Akita Biomedical Consulting
1111 Bailey Drive
Paso Robles, CA 93446
Phone: 805-238-2485
Fax: 949-203-6115
E-mail: [email protected]
For Consumer and
General Advertising
Information:
Veronica Nemeth
Editorial Coordinator
Cancer Control:
Journal of the Moffitt Cancer Center
12902 Magnolia Drive – MBC-JRNL
Tampa, FL 33612
Phone: 813-745-1348
Fax: 813-449-8680
E-mail: [email protected]
Jennifer Swank, PharmD
Clinical Pharmacist Medical Oncology/Internal Medicine
Sarah W. Thirlwell, MSc, MSc(A), RN
Nurse Director
Supportive Care Medicine Program
Eric M. Toloza, MD, PhD
Assistant Member
Thoracic Oncology
Nam D. Tran, MD
Assistant Member
Neuro-Oncology
Jonathan S. Zager, MD
Senior Member
Cutaneous Oncology
Cancer Control is a member of
the Medscape Publishers’ Circle®,
an alliance of leading medical
publishers whose content is
featured on Medscape
(www.medscape.com).
Most issues and supplements of
Cancer Control are available at
cancercontroljournal.org
Cancer Control: Journal of the Moffitt Cancer Center (ISSN 1073-2748) is published by H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612.
Telephone: 813-745-1348. Fax: 813-449-8680. E-mail: [email protected]. Internet address: cancercontroljournal.org. Cancer Control is included in Index Medicus ®/MEDLINE® and EMBASE®/
Excerpta Medica, Thomson Reuters Science Citation Index Expanded (SciSearch®) and Journal Citation Reports/Science Edition. Copyright 2016 by H. Lee Moffitt Cancer Center & Research Institute.
All rights reserved.
Cancer Control: Journal of the Moffitt Cancer Center is a peer-reviewed journal that is published to enhance the knowledge needed by professionals in oncology to help them minimize the
impact of human malignancy. Each issue emphasizes a specific theme relating to the detection or management of cancer. The objectives of Cancer Control are to define the current state of
cancer care, to integrate recently generated information with historical practice patterns, and to enlighten readers through critical reviews, commentaries, and analyses of recent research studies.
Disclaimer: All articles published in this journal, including editorials and letters, represent the opinions of the author(s) and do not necessarily reflect the opinions of the editorial board, the
H. Lee Moffitt Cancer Center & Research Institute, Inc, or the institutions with which the authors are affiliated unless clearly specified. The reader is advised to independently verify the effectiveness of all methods of treatment and the accuracy of all drug names, dosages, and schedules. Dosages and methods of administration of pharmaceutical products may not be those listed in
the package insert and solely reflect the experience of the author(s) and/or clinical investigator(s).
Table of Contents
Letter From the Editor
Better Cancer Treatment for All
322
Lodovico Balducci, MD
Editorial
Highlighting Health Disparities in Racial and Ethnic Minorities and Other Underserved Populations
323
Clement K. Gwede, PhD, RN, Gwendolyn P. Quinn, PhD, and B. Lee Green, PhD
Articles
Barriers to Clinical Trial Enrollment in Racial and Ethnic Minority Patients
With Cancer 327
Lauren M. Hamel, PhD, Louis A. Penner, PhD, Terrance L. Albrecht, PhD, Elisabeth Heath, MD,
Clement K. Gwede, PhD, RN, and Susan Eggly, PhD
Racial and Ethnic Differences in the Epidemiology and Genomics of Lung Cancer
338
Matthew B. Schabath, PhD, W. Douglas Cress, PhD, and Teresita Muñoz-Antonia, PhD
Black Heterogeneity in Cancer Mortality: US-Blacks, Haitians, and Jamaicans
347
Paulo S. Pinheiro, MD, PhD, Karen E. Callahan, MPH, Camille Ragin, PhD, MPH,
Robert W. Hage, MD, PhD, DLO, MBA, Tara Hylton, MPH, and Erin N. Kobetz, PhD, MPH
Genomic Disparities in Breast Cancer Among Latinas
359
Clinical Considerations of Risk, Incidence, and Outcomes of Breast Cancer
in Sexual Minorities
373
Filipa Lynce, MD, Kristi D. Graves, PhD, Lina Jandorf, MA, Charité Ricker, MS,
Eida Castro, PsyD, Laura Moreno, Bianca Augusto, Laura Fejerman, PhD, and
Susan T. Vadaparampil, PhD
Anne E. Mattingly, MD, John V. Kiluk, MD, and M. Catherine Lee, MD
Construction and Validation of a Multi-Institutional Tissue Microarray of Invasive Ductal Carcinoma From Racially and Ethnically Diverse Populations 383
Edward Seijo, MS, Diana Lima, MPH, Egiebade Iriabho, MS, Jonas Almeida, PhD,
Jesus Monico, MPH, MS, Margarita Echeverri, PhD, Sylvia Gutierrez, MD, Idhaliz Flores, PhD,
Ji-Hyun Lee, PhD, Kate Fisher, MA, William E. Grizzle, MD, PhD, Gabriel L. Sica, MD, PhD,
Charles Butler, Chindo Hicks, PhD, Cathy D. Meade, PhD, RN, Stephen Olufemi Sodeke, PhD,
Krzysztof Moroz, MD, Domenico Coppola, MD, and Teresita Muñoz-Antonia, PhD
October 2016, Vol. 23, No. 4
Cancer Control 319
Table of Contents
Articles, continued
Determinants of Treatment Delays Among Underserved Hispanics With Lung and Head and Neck Cancers
390
Evelinn A. Borrayo, PhD, Katie L. Scott, PhD, Ava R. Drennen, PhD, Tiare MacDonald, PhD,
and Jennifer Nguyen, MS
Geographical Factors Associated With Health Disparities in Prostate Cancer
401
Scott M. Gilbert, MD, Julio M. Pow-Sang, MD, and Hong Xiao, PhD
Disparities in Penile Cancer
409
Pranav Sharma, MD, Kamran Zargar-Shoshtari, MD, Curtis A. Pettaway, MD,
Matthew B. Schabath, PhD, Anna R. Giuliano, PhD, and Philippe E. Spiess, MD
Chemoprevention in African American Men With Prostate Cancer
415
Nagi B. Kumar, PhD, RD, Julio M. Pow-Sang, MD, Philippe E. Spiess, MD, Jong Y. Park, PhD,
Ganna Chornokur, PhD, Andrew R. Leone, MD, and Catherine M. Phelan, PhD, MD
Disparities in Adolescents and Young Adults With Cancer
424
Leidy L. Isenalumhe, MD, Olivia Fridgen, MPH, Lynda K. Beaupin, MD,
Gwendolyn P. Quinn, PhD, and Damon R. Reed, MD
Tobacco-Related Health Disparities Across the Cancer Care Continuum 434
Vani Nath Simmons, PhD, Bárbara Piñeiro, PhD, Monica Webb Hooper, PhD,
Jhanelle E. Gray, MD, and Thomas H. Brandon, PhD
Departments
Case Series: Human Metapneumovirus Infection in Immunocompromised Patients
442
Sharmeen Samuel, MD, Sowmya Nanjappa, MBBS, MD, Christopher D. Cooper, MD, and John N. Greene, MD
Special Report: Systematic Review and Case Series Report of Acinar Cell Carcinoma of the Pancreas
446
Special Report: A Single-Institution Study of Demographics and Outcomes of Adult Patients With Multiple Cancers 455
Evan S. Glazer, MD, PhD, Kevin G. Neill, MD, Jessica M. Frakes, MD, Domenico Coppola, MD, Pamela J. Hodul, MD,
Sara E. Hoffe, MD, Jose M. Pimiento, MD, Gregory M. Springett, MD, PhD, and Mokenge P. Malafa, MD, PhD
Thanh P. Ho, MD, Nathan R. Foster, MS, and Aminah Jatoi, MD
320 Cancer Control
October 2016, Vol. 23, No. 4
Table of Contents
Departments, continued
Ten Best Readings Related to Health Disparities in Oncology
461
Peer Reviewers, 2016
463
Index for 2016, Volume 23
465
About the art in this issue:
Bryan Whitney is a photographer based in New York City. His experimental, architectural, and fine art work have been featured in many magazines,
books, and has been exhibited in the United States and Europe. He has been the recipient of a Fulbright lecture grant and currently teaches at the International Center for Photography. To view more of Bryan’s work or to contact him, please visit www.x-rayphotography.com or www.bryanwhitney.com.
Cover: Calla Lillies
Articles:
Cherry Blossoms
Fiddle Head
Cactus
Chrysanthemum
Stag Horn
Proteus
Lily
Orchid
Birds Nest
Rose
Yellow Tulips
Iris
October 2016, Vol. 23, No. 4
Cancer Control 321
Letter From the Editor
Better Cancer Treatment for All
The best scientific and technological advances may reduce cancer-related mortality only when they are fully available to the people at risk. Availability is a multifaceted construct that includes economical and social resources,
education, and cultural adherence. This issue of Cancer Control could not be timelier because it addresses the
obstacles that prevents access by different minority groups to cancer diagnoses and treatments. The articles in this
issue propose practical solutions to this mounting problem.
Minority groups include African Americans, Hispanics, Asian Americans, new immigrants, and gay, lesbians,
and transgender persons, among others. When taken together, these select minority groups represent a sizable
portion of the US population as well as many Americans living with cancer. Given our globalization and the reduced birth rate among white Americans, these minority groups may embody a majority of patients with cancer
in the future. These minority groups deserve to become a main focus of cancer control in our country, because the
rate of cancer-related mortality is highest among them.1
In the accompanying editorial, our guest editors describe the burden of cancer affecting selected minorities
and found that it is largely due to remediable causes, including poor access to screening and early detection, lifestyle choices, cultural beliefs, and limited socioeconomic resources. In addition, they outline some biological issues
that have been largely ignored until recently and instead deserve immediate attention, such as the development of
deadly types of breast cancer in young African American women.
Cancer control in minorities is an ethical and social imperative. In addition to improving the welfare of the
general population, it will likely reduce the cost of medical care and provide a trait d’union among the different
ethnic/racial, religious, and gender-identity groups that represent the demographical richness of our country. It is
not far fetched to assume that the common fight against the suffering related to cancer may cement the alliance of
these different groups and provide a sense of common national identity.
Older patients with cancer represent another group with increased rates of morbidity and mortality due in part to
the inadequate care they receive.2 This group has been the subject of multiple studies and the focus of medical societies,3 but they have not been included in the present review — appropriately in my opinion.
My personal experience with the Middle Eastern Cancer Control group, which was led by a visionary Israeli
surgeon and included representatives of 32 countries in the Middle East, taught me that the common goal to relieve
pain and discomfort caused by cancer may bring peace and cooperation, even in the most war-torn regions of the
world.4,5 The cooperation of different cultures, even those from the poorest countries, is a 2-way street. I learned
how important spirituality can be in the delivery of palliative care, even when the technology is primitive and the
resources are scarce. I wish we could look to our minority patients as an opportunity to learn to provide better cancer treatment to all patients.
Lodovico Balducci, MD
Senior Member, Senior Adult Oncology Program
Editor, Cancer Control
H. Lee Moffitt Cancer Center & Research Institute
Tampa, Florida
[email protected]
References
1. Tehranifar P, Goyal A, Phelan JC, et al. Age at cancer diagnosis,
amenability to medical interventions, and racial/ethnic disparities in cancer
mortality. Cancer Causes Control. 2016;27(4):553-560.
2. Balducci L. Studying cancer treatment in the elderly patient population.
Cancer Control. 2014;21(3):215-20.
3. Wildiers H, Heeren P, Puts M, et al. International Society of Geriatric
Oncology consensus on geriatric assessment in older patients with cancer.
J Clin Oncol. 2014;32(24):2595-2603.
4. Silbermann M, Dweib Khleif A, Balducci L. Healing by cancer. J Clin
Oncol. 2010;28(8):1436-1467.
5. Silbermann M, Epner DE, Charalambous, A et al. Promoting new approaches for cancer care in the Middle East. Ann Oncol. 2013;24(suppl 7):vii5-vii10.
322 Cancer Control
October 2016, Vol. 23, No. 4
Editorial
Highlighting Health Disparities in Racial and Ethnic Minorities
and Other Underserved Populations
More than 50 years ago, Martin Luther King Jr said,
“Of all the forms of inequality, injustice in health care
is the most shocking and inhumane.”1 Yet more than
30 years after the report from Heckler2 in 1985 on the
national state of minority health in the United States,
inequities in cancer care have persisted, widened, and,
in some cases, emerged in new areas.3-7 Disparities in
health status refer to the variation in rates of disease
occurrence and disabilities between populations defined by demographical, socioeconomical, or geographical factors, most of which exist in populations
referred to as minorities and underserved groups.3-5
These groups may be classified by race (eg, black,
Asian), ethnicity (eg, Hispanic, Ashkenazi Jewish),
sexual orientation (eg, gay, lesbian), gender (eg, transgender, gender queer), socioeconomic status (eg, low
income, poor, underserved), or age (eg, seniors, adolescents and young adults [AYAs]).3-7
Evidence suggests that health disparities in the
United States result from a combination of poverty,
limited access to quality care, perceived racism/discrimination, genetics, biology, and environmental
exposure.8,9 A common thread in health disparities
specific to cancer is their multidimensionality and
complex nature: covering the lifespan, across the
continuum of cancer care from prevention to survivorship, and transcending socioecological and transcultural contexts.5,8,9 A growing body of literature
suggests that cancer-related disparities are fueled by
a multifaceted and concurrent interplay of factors that
range from biology and genetics to lifestyle and behavior.5,8-10 As such, intersectionality, a theoretical focus on the interlocking factors of cultural influence
and the multiple identities perceived by patients (eg, a
racial/ethnic minority who also identifies as a sexual
minority), helps explain how these complex realities
exacerbate disparities.11
It is important to consider reducing health disparities in terms of modifiable factors. That is, race
and ethnicity do not change, but certain contextual
and behavioral aspects, including cancer risk, screening behavior, and cancer care needs, can be identified
and addressed. The core of critical interventions requires appropriate attention to the cultural and health
literacy levels of individuals and groups.12-14 Furthermore, as a community of clinicians, researchers, and
educators, we can change the way cancer control and
prevention services are delivered by reducing the perOctober 2016, Vol. 23, No. 4
ceived racism/discrimination and associated stress in
minority and underserved populations. Psychological stress caused by perceived discrimination and stereotyping, poverty, and differential access to care can
produce higher rates of depression and other morbidities that in turn impact quality of life and health
outcomes.13,14
Articles in this issue of Cancer Control examine
multiple aspects of health disparities in a variety of racial/ethnic minorities and other underserved patient
populations. This issue is a collection of articles that
broadly address health disparities in cancer care from
2 perspectives: going beyond race and ethnicity (addressing subethnicity and emerging disparate populations, eg, AYAs, gender and sexual minorities) and
addressing multiple points across the continuum of
cancer control and contextual factors (eg, geography).
These articles explore pathways and mechanisms propelling cancer-related disparities, and they offer summary evidence to address such disparities.
A narrative review of the literature by Dr Hamel
and colleagues examines participation in clinical
trials and takes a close look at the barriers to enrollment in clinical trials, with an emphasis on the under-representation of racial and ethnic minorities in
research studies. The authors identify barriers across
3 levels — system, individual, and interpersonal — and
conclude that many of the interventions utilized to address minority accrual to clinical trials have focused on
a single level. They suggest that successful interventions must be multilevel.
Dr Schabath and fellow researchers review the literature to summarize the rates of incidence, mortality,
and survival among racial and ethnic minorities with
lung cancer. Identifying racial and ethnic differences in
lung cancer will aid in the creation of effective prevention and treatment strategies that in turn may reduce
the overall burden of lung cancer.
Dr Pinheiro and others examine a unique aspect of disparities research by focusing on the subethnic groups within blacks. The authors examined
the mortality rates among blacks in the United States
(eg, blacks, Haitians, Jamaicans) and compared them
with their country of origin. Their research suggests that US-born blacks have the highest risk of
cancer-related mortality when compared with blacks
from other countries.
Dr Lynce and colleagues summarize what is
Cancer Control 323
known about Latinas and breast cancer, noting the exyond racial disparities to include younger age, lower
istence of genetic mutations of high and moderate peneducational and income levels, marital status, lack of
etrance within this ethnic population. They conclude
circumcision, and a number of sexual behaviors as
that reduced awareness and uptake of genetic counselfactors that compound disparities in this disease. This
ing were observed within this group and that tailored
article reminds readers to broaden their view of disinterventions are needed.
parities beyond race and ethnicity.
Dr Mattingly and fellow researchers summarize
Dr Kumar and colleagues tackle a topic fraught
the literature on sexual minority women with breast
with clinical and scientific uncertainties: screencancer, concluding that limited research exists on this
ing, early detection, and prevention of prostate canpopulation primarily due to lack of collection of sexual
cer among high-risk African American men. Their
orientation information in databases.
work examines biological etiology, biomarkers, and
Mr Seijo and coauthors address the lack of banked
pathways, specifically underscoring the need for chetissue among racial and ethnic minorities. Their team
moprevention agents and strategies. This synthesis
reports on an established infrastructure for a biobankidentifies unique potential pathways that drive excess
ing hub in the southeastern United States and Puerto
risk and burden of disease and the potential of green
Rico to create a web-based database and tissue microtea catechins as a chemoprevention agent in African
arrays for breast cancer.
American men diagnosed with high-grade prostatic inDr Borrayo and colleagues examine the determitraepithelial neoplasia.
nants of treatment delays among underserved HisDr Isenalumhe and fellow researchers examine
panics with lung and head and neck cancers. They
the literature on AYAs with cancer for whom survival
report that women experience longer delays than
rates have not improved as they have in younger and
men. Other observed factors associated with longer
older patients. Disparities in this population range
delays include fewer comorbidities, receiving treatfrom the unique biology of the cancer itself to delays
ment at a public facility, and being underserved (eg,
in diagnoses further exacerbated by the psychosouninsured). This article identifies actionable factors
cial needs of this population (eg, reproductive presamenable to intervention to improve outcomes in diservation). Their article focuses on identifying these
advantaged Hispanic patients.
unique needs and the research advancements made
Dr Gilbert and others take an innovative apto improve outcomes and quality of life in AYAs.
proach to addressing racial and ethnic disparities in
Dr Simmons and coauthors attend to racial/ethnic
prostate cancer treatment and outcomes by going
and socioeconomic disparities in tobacco use, one of
beyond the traditional factors of race and socioecothe leading preventable causes of lung cancer–related
nomic factors. Their focus is on the geographic disdeath. Their contribution focuses attention on importribution of disparities (ie, how a place of residence
tant disparities that can occur across the cancer coninteracts with race and socioeconomic factors). Using
tinuum — from prevention to survivorship. They synthe Geographic Information System, they illustrate
thesize the research that has reduced tobacco-related
an example showing locations
in the state of Florida where
disparities in prostate cancer
Race and ethnicity are terms often interchangeably used, although
are the most problematic. The
they describe 2 different but complex concepts. They are often comauthors identify geographical
ingled in describing populations in health care and health disparities
differences in treatment across
research. In addition, the criticism is growing about the shortcomings
racial and socioeconomic straof using these classifications in health research.
ta, thus availing a better tool for
Race is a social classification (not a biological category) that depublic health officials, health
scribes a person’s observable physical characteristics and identity
systems, health care profesbased on phenotypic genetic characteristics such as anatomical or
sionals, and policymakers to
bone structure and skin, hair, or eye color.
use when addressing health
Ethnicity describes the shared identity of a population based on
disparities.
sociocultural factors, linguistic or religious traits, or national heriDr Sharma and coauthors
tage, regardless of race that often shape that group’s world-view, lifeaddress a less-common cancer
style, and behaviors.
and rare disparity in developed
Individuals can be of “mixed race or more than 1 race” and can
nations, penile cancer, in which
have more than 1 ethnicity. Advances in genomic and precision
racial differences exist in both
medicine are helping to elucidate the genetic and biological basis of
rates of disease incidence and
health disparities.
associated survival outcomes.
Their chief findings extend be324 Cancer Control
October 2016, Vol. 23, No. 4
disparities, pointing to the limited successes of both
pharmacotherapy and behavioral counseling for racial
and ethnic minorities, concluding with a critical call for
innovative efforts directed at each point along that continuum. These authors remind us of the critical role of
culture and socioeconomic status in addition to racial
and ethnic status.
The Centers for Disease Control and Prevention
suggests the key to reducing cancer-related inequities
is to increase screening and early detection, promote
healthy behaviors, and expand access to health care for
underserved populations and groups at greatest risk.3
The articles in this issue of Cancer Control help us
identify the ever-expanding definition of what it means
to be “at risk” as well as how minority and underserved
populations are disproportionately affected by an excess burden of cancer. These articles also point us in
the direction of key areas for expanding cancer care
and new research to help us identify and reduce cancer-related inequities. This issue highlights promising
strategies for reducing the cancer-related burden and
improving the quality of life of all patients. A heightened understanding of both differences and similarities across age, sex, gender, race, ethnicity, socioeconomic class, and sexual orientation is essential to the
delivery of equitable cancer care. It is incumbent upon
us to focus on ways to reduce the cancer burden and
disparities in rates of cancer incidence, morbidity, and
mortality so that no one is left behind.
4. National Institutes of Health; National Cancer Institute. About cancer
health disparities. https://www.cancer.gov/about-nci/organization/crchd/
about-health-disparities. Accessed October 3, 2016.
5. Alcaraz K, Sly J, Ashing K, et al. The ConNECT Framework: a model
for advancing behavioral medicine science and practice to foster health
equity. J Behav Med. 2016. Epub ahead of print.
6. Quinn GP, Sanchez JA, Sutton SK et al. Cancer and lesbian,
gay, bisexual, transgender/transsexual, and queer/questioning (LGBTQ)
populations. CA Cancer J Clin. 2015;65(5):384-400.
7. Committee on Lesbian, Gay, Bisexual, and Transgender Health
Issues and Research Gaps and Opportunities; Board on the Health of
Select Populations; Institute of Medicine. The Health of Lesbian, Gay,
Bisexual, and Transgender People: Building a Foundation for Better
Understanding. Washington, DC: National Academies Press; 2011.
8. Freeman HP. Poverty, culture, and social injustice: determinants of
cancer disparities. CA Cancer J Clin. 2004;54(2):72-77.
9. Freeman HP, Chu KC. Determinants of cancer disparities: barriers
to cancer screening, diagnosis, and treatment. Surg Oncol Clin N Am.
2005;14(4):655-669.
10. Dressler WW, Oths KS, Gravlee CC. Race and ethnicity in public
health research: models to explain health disparities. Ann Rev Anthropol.
2005;34:231-252.
11. Hsieh N, Ruther M. Sexual minority health and health risk factors:
Intersection effects of gender, race, and sexual identity. Am J Prev Med.
2016;50(6):746-755.
12. Kagawa-Singer M, Dressler WW, George SM, et al. The Cultural
Framework for Health: An Integrative Approach for Research and Program
Design and Evaluation. Bethesda, MD; National Institutes of Health; 2016.
13. Smedley BD, Stith AY, Nelson AR, eds; Committee on Understanding
and Eliminating Racial and Ethnic Disparities in Health Care. Unequal
Treatment: Confronting Racial and Ethnic Disparities in Health Care.
Washington, DC: National Academies Press; 2003.
14. LaVeist TA, Isaac LA, eds. Race, Ethnicity, and Health: A Public
Health Reader. 2nd ed. San Francisco, CA: Josey-Bass; 2012.
Clement K. Gwede, PhD, RN
Associate Member, Health Outcomes and Behavior Program
H. Lee Moffitt Cancer Center & Research Institute
Tampa, Florida
[email protected]
Gwendolyn P. Quinn, PhD
Senior Member, Health Outcomes and Behavior Program
H. Lee Moffitt Cancer Center & Research Institute
Tampa, Florida
[email protected]
B. Lee Green, PhD
Vice President, Diversity, Public Relations and Strategic
Communications
Senior Member, Health Outcomes and Behavior Program
H. Lee Moffitt Cancer Center & Research Institute
[email protected]
References
1. King ML Jr. Presentation at: Second National Convention of the
Medical Committee for Human Rights; Chicago, IL; March 25, 1966.
2. Heckler MM. Report of the Secretary’s Task Force on Black and
Minority Health. Washington, DC: US Department of Health and Human
Services; US Government Printing Office; 1985.
3. Centers for Disease Control and Prevention. Health disparities in cancer.
http://www.cdc.gov/cancer/dcpc/resources/features/cancerhealthdisparities.
Accessed September 26, 2016.
October 2016, Vol. 23, No. 4
Cancer Control 325
Health Outcomes and Behavior Program
H. Lee Moffitt Cancer Center & Research Institute
The Health Outcomes and Behavior Program contributes to the prevention, detection, and control of
cancer through the study of health-related behaviors, health care practices, and health-related quality of life.
Work toward this goal involves research across the disease spectrum, from prevention and detection through
survivorship or advanced disease. The program’s 4 specific aims are to: (1) understand the determinants of
health behaviors that can lead to the prevention and early detection of cancer and develop effective methods
of promoting those behaviors, (2) understand and improve the quality of life of patients and family members
throughout the disease course, (3) contribute to the evidence base and synthesis of evidence regarding the
delivery of cancer care and clinical outcomes, and (4) understand and intervene upon the social, cultural, and
behavioral determinants of cancer-related health disparities.
Awareness is growing of the disparities that occur across the cancer continuum, including prevention, incidence, diagnosis, treatment, and outcomes. These disparities occur with respect to sex, gender identity, race,
ethnicity, socioeconomic status, and other patient characteristics. To understand and address these disparities,
program members conduct community-based participatory research throughout the catchment area. Program
members have translated their earlier observational research into numerous ongoing, randomized controlled
trials of interventions designed to reduce a range of cancer-related health disparities.
DEPARTMENT CHAIR/ PROGRAM LEADER
Thomas H. Brandon, PhD
VICE DEPARTMENT CHAIR
Susan Thomas Vadaparampil, PhD
Faculty
Daniel A. Anaya, MD
Lodovico Balducci, MD
Alicia Best, PhD
Margaret Booth-Jones, PhD
Ellen M. Daley, PhD
Benjamin Djulbegovic, MD, PhD
Kristine A. Donovan, PhD
David J. Drobes, PhD
David Evans, PhD
Martine Extermann, MD, PhD
Michael Fradley, MD
Scott M. Gilbert, MD
B. Lee Green, PhD
Clement K. Gwede, PhD, RN
Ming Ji, PhD
Heather S. Jim, PhD
Julio C. Jiminez Chavez, MD
Peter A.S. Johnstone, MD
Cecile A. Lengacher, RN, PhD
Dinorah Martinez Tyson, PhD
Susan C. McMillan, PhD, RN
Cathy D. Meade, PhD, RN
Teresita Muñoz-Antonia, PhD
Michael A. Poch, MD
Gwendolyn P. Quinn, PhD
Maija Reblin, PhD
Carmen S. Rodriguez, PhD, ARNP-BC
Richard G. Roetzheim, MD
Lori A. Roscoe, PhD
Julian Sanchez, MD
Vani Nath Simmons, PhD
Brent J. Small, PhD
Marilyn Stern, PhD
Steve K. Sutton, PhD
Tawee Tanvetyanon, MD
Lora M.A. Thompson, PhD
Christine Vinci, PhD
Hsiao-Lan Wang, PhD
For more detailed information about the Health Outcomes and Behavior Program, please visit
www.moffitt.org/research-science/research-programs/health-outcomes-and-behavior
www.moffitt.org
326 Cancer Control
October 2016, Vol. 23, No. 4
Barriers to racial and ethnic minorities
enrolling in clinical trials exist at system,
individual, and interpersonal levels.
Cherry Blossoms. X-Ray image courtesy of Bryan Whitney. www.x-rayphotography.com.
Barriers to Clinical Trial Enrollment in Racial and
Ethnic Minority Patients With Cancer
Lauren M. Hamel, PhD, Louis A. Penner, PhD, Terrance L. Albrecht, PhD, Elisabeth Heath, MD,
Clement K. Gwede, PhD, RN, and Susan Eggly, PhD
Background: Clinical trials that study cancer are essential for testing the safety and effectiveness of promising treatments, but most people with cancer never enroll in a clinical trial — a challenge exemplified in
racial and ethnic minorities. Underenrollment of racial and ethnic minorities reduces the generalizability of
research findings and represents a disparity in access to high-quality health care.
Methods: Using a multilevel model as a framework, potential barriers to trial enrollment of racial and ethnic
minorities were identified at system, individual, and interpersonal levels. Exactly how each level directly or
indirectly contributes to doctor–patient communication was also reviewed. Selected examples of implemented
interventions are included to help address these barriers. We then propose our own evidence-based intervention addressing barriers at the individual and interpersonal levels.
Results: Barriers to enrolling a diverse population of patients in clinical trials are complex and multilevel.
Interventions focused at each level have been relatively successful, but multilevel interventions have the greatest potential for success.
Conclusion: To increase the enrollment of racial and ethnic minorities in clinical trials, future interventions
should address barriers at multiple levels.
Introduction
Clinical trials focused on cancer research are essential for testing the safety and effectiveness of potential
From the Department of Oncology (LMH, LAP, TLA, EH, SE) and Population Studies Disparities Research Program (LMH, LAP, TLA, SE),
Wayne State University, Karmanos Cancer Institute, Detroit, Michigan,
and the Department of Oncologic Sciences/Population Sciences (CKG),
H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida.
Submitted January 26, 2016; accepted May 3, 2016.
Address correspondence to Lauren M. Hamel, PhD, Wayne State
University, Karmanos Cancer Institute, Department of Oncology,
Population Studies Disparities Research Program, 4100 John R
Street, MM03CB, Detroit, MI 48201. E-mail: [email protected]
This work was funded in part by National Institutes of Health/
National Cancer Institute grant no. R01CA200718-01.
No significant relationships exist between the authors and the companies/organizations whose products or services may be referenced
in this article.
October 2016, Vol. 23, No. 4
treatments and translating new knowledge into tangible benefits for patients; they also represent options for
novel therapy for cancer.1,2 However, approximately
2% to 3% of all patients with cancer ever enroll in a trial.3,4 Estimates of the number of trials that fail to meet
scientific objectives because of insufficient accrual
rates range from 22% to 50%.5,6 Low accrual rates jeopardize the ability of researchers to assess the safety and
effectiveness of new approaches to cancer care, wastes
resources, precludes follow-up studies, and reduces
the ability of our clinical research system to translate
research into evidence-based practice.6-8
Underenrollment is an even greater challenge
among racial and ethnic minorities — particularly
African Americans — despite a requirement by the
National Institutes of Health that members of minority
Cancer Control 327
populations be represented in clinical research.1,3,4,9-11
A systematic review compared the proportion of underrepresented minority participants in phase 3 cancer
treatment and prevention clinical trials conducted between the periods 1990 to 2000 and 2001 to 2010.12 In the
treatment studies conducted between 2001 and 2010 that
reported race/ethnicity, the reviewers found that 82.9%
of participants were white, 6.2% were African American, 3.3% were Asian, 2.2% were Hispanic, and 0.1%
were Native American.12 This is in contrast to studies
conducted between 1990 and 2000, in which 89% of
participants were white, 10.5% were African American,
0.4% were Hispanic, and 0.04% were Asian.12 In other
words, even though the proportion of white participants decreased, whites continued to comprise a large
majority of participants in cancer treatment trials, and
the proportion of African American participants decreased between the periods 1990 to 2000 and 2001
to 2010. However, during those same periods, the
proportion of African American participants in clinical prevention trials increased (5.5% from 1990–2000,
11.6% from 2001–2010).12
Although several patient populations are underrepresented in clinical trials, including elderly patients
(≥ 65 years), residents of rural areas, and those with low
socioeconomic status, the current review focuses on
racial and ethnic minority underenrollment for several
reasons.4,10,13-16 Racial and ethnic minorities — particu-
larly African Americans — bear the greatest cancer burden in the United States, so they should be adequately
represented in cancer research.17-19 Under-representation of racial and ethnic minorities also limits the generalizability of research findings.20-22 The Institute of
Medicine has recommended that every individual with
cancer have access to high-quality clinical trials, so we
believe that under-representation of racial and ethnic
minorities represents a disparity in health care.2 Underenrollment of racial and ethnic minorities in clinical trials may therefore contribute to preventable disparities
in treatment outcomes and survival.1,17,23,24
Purpose
The purpose of this paper is to identify and describe
potential barriers to the enrollment of racial and ethnic minorities in clinical trials at the system, individual
(health care professional, patient, and family), and interpersonal levels (eg, doctor–patient relationship).
The paper also describes selected examples of evidence-based interventions already implemented to address some of these barriers at the patient-, physician-,
and doctor–patient interpersonal communication
levels. We take this approach because multilevel interventions, as compared with single-level interventions,
may have the greatest potential to achieve substantial
and sustained change and to produce additive — and
possibly multiplicative — effects.25,26
System Level
Availability of clinical trials
Supporting infrastructure
Cost/payer
Strict eligibility criteria
Lack of community engagement
Individual Level
Health Care Professional
• Awareness of clinical trials
• Perception of the hospital/infrastructure
• Attitude/bias/experience
• Perception of patient’s race/ethnicity
Patient/Family
• Awareness of clinical trials
• Eligibility
• Personal identity: ethnicity, race, religion
• Attitude/bias/experience
(eg, suspicion of medical care)
• Access (eg, transportation)
Interpersonal Level
Perception of the institutional infrastructure
Attitude/perception of each other and care received
Doctor–patient relationship
Disparities
Fig 1. — A multilevel model of factors contributing to disparities in clinical trials.
328 Cancer Control
October 2016, Vol. 23, No. 4
We propose a multilevel model derived from
general systems theory, which posits that organizations are comprised of discrete but related levels
(Fig 1).27 Our multilevel model demonstrates how
system- and individual-level barriers contribute to
interpersonal-level barriers to the enrollment of racial
and ethnic minorities in clinical trials.
System-Level Barriers
Barriers at the level of health care systems and hospitals include the limited number of trials nationally
and regionally available; hospital infrastructures that
lack the resources to support trials; financial costs to
hospitals and patients; restrictive study designs and
eligibility criteria; and lack of community engagement.
Many of these barriers have a disproportionate effect
on minority enrollment because members of minority
populations are more likely to receive care at underresourced hospital systems where few clinical trials
are available, to be underinsured, and to present with
comorbidities that make them ineligible for trials that
may be available.28-30
Available clinical trials supported by a well-resourced and functional infrastructure are critical to the
enrollment of minority patients.31 For example, this infrastructure includes staff dedicated to enrolling, managing, and tracking participants; data collection and
management capabilities; and an efficient Institutional
Review Board. Without a functional infrastructure, efforts at increasing the enrollment of minorities in other
levels are less likely to be successful.31-33 The financial
costs of enrollment to institutions and individual patients affect all patient populations, but they have a
disproportionate impact on minorities compared with
nonminorities. Minorities are more likely to be underinsured, to seek care at under-resourced hospitals,
and to have concerns about the cost of participating
in a clinical trial.29,34 In addition, lack of or inadequate
health insurance acts as a barrier to enrollment in clinical trials for several under-represented populations, including racial and ethnic minorities.29,35
As a way to enhance community engagement, the
infrastructure of clinical trials should exist within a
medical institution that has an established, trusted relationship with the members of the community it serves.36
Building trust within a community requires that an
institution conduct its research in an open and honest
manner that involves a complete and accurate description of the known and potential differences in the risks
and benefits for different racial and ethnic populations.
Ways and means to facilitate and encourage open discussions into research ethics, including the past abuse of
minority patients in research, are needed, as are efforts
to create and sustain partnerships with the community
to share ownership of the research.37-40
In response to system-level barriers, several nationOctober 2016, Vol. 23, No. 4
al, regional, and consortia efforts have implemented
programs to increase enrollment among racial and ethnic minority populations.8,41,42 For example, the Minority-Based Community Clinical Oncology Program of the
National Cancer Institute was designed to increase minority participation in cancer clinical trials by developing outreach efforts at health care institutions that serve
large numbers of minority patients with cancer.32 McCaskill-Stevens et al32 reported that the success of this
program was largely due to system- and hospital-level
factors, such as opening trials that matched the clinical
characteristics of patient populations and developing
relationships with local physicians and cancer advocacy groups that increased their willingness to enroll or
refer minority patients to clinical trials. However, their
success was limited by lack of funding for minority outreach activities, lack of system support to provide staff
and mentoring for minority investigators, and lack of
funding to provide protocol-related drugs and related
services for uninsured patients.32
The Community Oncology Research Program of
the National Cancer Institute is a national network of
health care professionals, investigators, and organizations that conducts cancer-related research across the
United States.43 The program focuses on determining
the reasons for the existence of racial and ethnic disparities in clinical trial participation, as well as ways to
increase the participation of racial and ethnic minorities in clinical trials through numerous interventions.43
Because its inauguration was in 2014, evaluation data
have not yet been published.
In recent years NRG Oncology convened a workshop on the challenges and opportunities of clinical
trial enrollment in an attempt to address the issue of
minority underenrollment in clinical trials.44 Experts
in oncology, including members of the Minority-Based
Community Clinical Oncology Program of the National
Cancer Institute, and a large patient advocacy group
were in attendance.44 Ten themes emerged regarding
future plans and interventions to increase minority enrollment in clinical trials; of these, 8 themes focused on
the system level44:
1. Target recruitment and emphasize the personalized nature of trials as a way to improve minority
enrollment to specialized studies
2. Define study populations at the molecular level
rather than by traditional and less-precise eligibility criteria
3. Make the delivery of cancer care research available at the community and academic levels so
studies are designed to better improve health
outcomes
4. Improve local infrastructure by providing translation and navigation services and research-friendly
electronic medical records
5. Expand and standardize the collection of demoCancer Control 329
graphics and real-time information about accruals
for all patient populations
6. Strengthen the infrastructure of information
technology
7. Improve recruitment, training, and mentorship of
young investigators, particularly those of racial
and ethnic minority backgrounds and those with
an interest in cancer disparities
8. Continue and improve budgetary support from
the US Congress for all clinical cancer research
within the National Cancer Institute
Individual-Level Barriers
Our multilevel model emphasizes individual factors that
may present a barrier to the enrollment of racial and ethnic minorities in clinical trials. The term individuals is
meant to refer to persons directly involved in the clinical
trial enrollment process, including health care professionals, patients, and their family members. Barriers on
a system level can influence how individuals perceive
the enrollment of minority patients in clinical trials and
how they communicate about trials. For example, an investigation of physician perceptions of clinical trials and
trial support available at their institution revealed that
most physicians held a favorable attitude about clinical
trials, both as a source of high-quality patient care and
as a professional benefit to themselves.33 However, approximately two-thirds of practicing oncologists perceived a lack of needed infrastructure to conduct trials.33
This included a low number of trials open and lack of
nonphysician staff to support patient participation in
clinical trials.33 Similarly, the health care system can also
influence patient concerns about the cost of participating in trials and, in turn, their decisions to enroll or not
enroll in a clinical trial.34 Thus, these 2 levels are linked
in terms of whether and how health care professionals
and minority patients discuss clinical trials and whether
minority patients agree to enroll.
Health Care Professionals
Health care professionals play a critical role in facilitating or inhibiting the enrollment of a diverse population
of patients into clinical trials.33,45,46 Several factors are
related to the reluctance of health care professionals to
enroll their patients in trials, regardless of whether or
not they are members of minority groups themselves.
A major barrier is the lack of awareness of available trials: Health care professionals must be made
aware of national, regional, and local trials currently
taking patients and the eligibility criteria for each of
those trials to discuss such enrollment with their patients.45,46 In addition, the attitudes of health care professionals about trials and discussing such trials with
patients and their families present a major barrier to
trial enrollment, and they likely affect the quality of
communication during discussions of clinical trials.33,45
330 Cancer Control
For example, health care professionals may not fully
agree with or understand the scientific value of trials
in general or the details of specific trials; they may feel
they do not have adequate system support; or they may
have concerns about practical issues such as strict protocol designs, patient inconvenience, and added work
for the health care staff.33,45 Some clinicians find it difficult to reconcile the roles of physician and researcher,
or are concerned about unduly influencing patient decisions about enrolling in a clinical trial.47
Some attitudinal factors are specific to the enrollment of minority patients, such as the concern about
harming the therapeutic relationship. For example, a focus-group study involving community physicians found
that some physicians were hesitant to discuss clinical trials with their African American patients because of their
perceptions that African Americans are often mistrustful of physicians and medical institutions.48
An additional attitudinal factor that may deter
health care professionals from discussing trials with
minority patients is implicit bias against members of
minority groups. Prior research has shown that implicit
bias (comprised of conscious and unconscious biases)
among physicians toward members of minority groups
has an impact on clinical interactions with minority patients.49-52 We are unaware of studies that specifically
investigate whether the implicit bias of health care professionals is a barrier to the enrollment of minorities
in clinical trials, but research in other contexts suggests that some physicians may have implicit negative
attitudes about African Americans that may lead them
to believe they will be poor candidates for clinical trials.53-56 For example, research has shown that primary
care physicians tend to trust minority patients less than
they trust white patients.54 Research has also shown
that physicians high in implicit bias may view their
minority patients as less likely to comply with recommended treatment,57 and that physicians’ diagnoses
and treatment of African Americans can be influenced
by racial stereotypes.55 This body of research suggests
that physicians may limit offers for trial enrollment to
those patients they perceive as good study candidates
so that the studies will be conducted in a timely and
efficient manner.58 This may mean physicians are less
likely to offer a clinical trial to patients who are racial
and ethnic minorities than to white patients, and research has shown this to be the case.46,59
Recommendations have been made for evidencebased approaches to reduce the impact of health care
professional bias in clinical interactions.60 One approach is to train health care professionals in the use of
high-quality patient-centered communication, which
requires the development of a positive interpersonal
relationship.61 Patient-centered communication focuses
on patient needs and perspectives, and it does not ignore the racial and ethnic background of the patient,
October 2016, Vol. 23, No. 4
because doing so denies an important part of an individual.61 Results of the NRG Oncology workshop identified the commitment of physicians and study investigators to the enrollment of minorities into clinical trials
as critical to the future of clinical cancer research.44
They also emphasized that physicians must be culturally sensitive and aware of the impact appropriate communication and patient trust have on minority enrollment in clinical trials.44
Several communication curricula are available, but
they include little training on discussing clinical trials
with patients or with specific patient populations.62
One exception is an Australian study, which reported
on a 1-day intensive course for physicians to improve
aspects of their communication related to discussing
clinical trials with patients.45 Results showed that physicians improved in some shared decision-making behaviors and were more likely to describe some of the
key clinical or ethical aspects of the trial.63 However,
we are unaware of any specific training curriculum
that emphasizes ways to communicate about clinical
trials with minority patients.
Patients and Families
Some major barriers to enrolling racial and ethnic minority patients in clinical trials are that patients are often not aware they are eligible for an existing trial; they
are often underinsured and cannot afford the extra expenses related to trial participation; and they may not
meet the clinical criteria due to comorbid conditions or
age restrictions.29,30,64 Similar to health care professionals, patients hold attitudes and beliefs that may also affect their willingness to participate and the way they
communicate during clinic visits in which trials are discussed.65,66 Despite this, research shows that, overall,
minorities are as likely as whites to consent if they are
offered a trial.30,59,67,68 For example, an assessment of
more than 4,000 racially diverse patients with cancer
found no significant association between race and ethnicity and refusal to participate in a clinical trial or lack
of desire to participate in research.30 Similarly, Katz et
al69 implemented the Tuskegee Legacy Project to address and understand if and how the Tuskegee syphilis
experiment impacted the trial recruitment and retention of African Americans into biomedical studies. The
researchers found no difference in self-reported willingness among African Americans to participate in biomedical research.69 A systematic review of the available
research on the factors that influence participation in
clinical trials among African Americans concluded that
the strongest inhibitors of participation were low levels
of knowledge and lack of awareness of clinical trials.66
The most important facilitators of participation were
social support and recommendations from physicians,
family members, and friends.66
Several patient-focused interventions designed to
October 2016, Vol. 23, No. 4
improve knowledge of, attitudes about, and participation in clinical trials among racial and ethnic minority patients has been developed and tested.70,71 A multicenter, randomized trial of a web-based, interactive,
educational tool was designed to increase knowledge,
decrease attitudinal barriers, and improve preparation
for making decisions about clinical trial enrollment
among minority patients with cancer.70 At baseline,
the level of knowledge about clinical trials, attitudinal barriers to participation, and preparation for decision-making among the participants were assessed.70
Prior to their initial visit with an oncologist, patients in
the intervention arm watched a set of brief, individually tailored videos addressing knowledge and attitudinal barriers to clinical trial participation, whereas
those assigned to the control arm received text-based
general information about clinical trials from the National Cancer Institute. The study results showed
that, compared with baseline levels, participants in
the intervention and control arms both showed improved knowledge (control arm: mean [standard
deviation {SD}] = 2.5 [3.1]; intervention arm: mean
[SD] = 3.2 [3.1]; P < .001) and decreased attitudinal
barriers (control arm: mean [SD] = –0.2 [0.4]; intervention arm: mean [SD] = –0.3 [0.5]; P < .001).70 However,
patients in the intervention arm had a significantly
greater increase in knowledge and decrease in attitudinal barriers compared with patients in the control
arm (P < .001).71 All patients had a significant increase
in their preparedness to consider participation in
clinical trials (control arm: mean [SD] = 3.4 [13.5]; intervention arm: mean [SD] = 4.7 [12.8]; P < .001).70
Another intervention specifically addressed the
negative perceptions and attitudes of patients about
clinical trials by assessing the effectiveness of a multimedia, psychoeducational intervention compared with
printed educational materials.71 The intervention was
designed to improve patient attitudes and knowledge
of clinical trials, their ability to effectively make decisions, their receptivity to receiving more information,
and their willingness to participate in clinical trials.71
The researchers observed that study participants who
received the intervention had more positive attitudes
toward clinical trials (mean [SD] = 0.21 [0.01]; P = .02),
and, although the 2 interventions were not developed
and tested with minority patients, the study results
showed no significant difference between intervention by patient demographics.71 Thus, the intervention
could potentially be adapted to specifically benefit racial and ethnic minority patient populations.
Although racial and ethnic minority patients are
overall as likely as white patients to agree to enroll
in a clinical trial, reasons for declining may differ by
racial and ethnic background.30,59,66-68 Minority patients — particularly African Americans — may hold
negative race-related attitudes and beliefs that could diCancer Control 331
rectly and indirectly influence interactions with their
health care professional regarding trials and decisions
about enrollment.38,40,66,72 These attitudes, which have
been derived in large part from issues of racism, uninformed consent, and poor health care for minorities
in the United States, include greater mistrust in medical institutions and health care professionals and a
greater sense of having been the target of discrimination.20,23,29,38,69,72-78 A systematic review of the research
assessing the multiple barriers to minority enrollment
in clinical trials found that mistrust in research and the
medical system was the most common barrier to patients’ willingness to participate in a clinical trial.29
The attitudes of African American patients toward
medical research were assessed using focus groups of
African American adult patients from a single urban
public hospital.72 Results showed that most were in favor of medical research as long as they were not treated as “guinea pigs.”72 The authors also reported that
many of the participants had a limited understanding
of the informed consent process, and many patients assumed the consent form was protecting hospitals and
doctors from any legal responsibility, not to protect patients.72 Another study that assessed and compared the
racial differences in factors that influence patient willingness to enroll in clinical research studies found that
African Americans were less willing to participate in
medical research if they attributed a high importance
to the race of the physician when seeking routine medical care and believed that minorities bear most of the
risks of medical research.40
Interventions designed to increase the recruitment
of minorities in clinical trials are generally focused on
communities, rather than on individuals.32,79 One such
intervention targeted minority recruitment using strategic planning, which included meetings and conferences with key stakeholders and minority organizations as a means to increase minority enrollment.79 The
authors examined institutions that employed these
strategies and those that did not, and they found a significant increase in the rate of minority accrual in institutions that implemented strategic planning compared
with those that did not.79 This effort suggests that the
mistrust of the medical system seen among minority
patients might be addressed with culturally sensitive
and open community engagement.
Other identified patient-level barriers to the enrollment of racial and ethnic minorities in clinical trials
include lack of transportation, inadequate insurance,
lack of childcare, and poor access to health care.42,66
The NRG Oncology workshop also identified direct-topatient communication and advertising as critical for
the future of minority enrollment in clinical trials, emphasizing that this should be done with collaboration
with key stakeholders such as community groups, survivor advocacy groups, and religious institutios.44 The
332 Cancer Control
intersection between the impact of patient attitudes
and access to trial participation illustrates the complex
and multilevel nature of this issue and the need for
more comprehensive interventional strategies.
Interpersonal-Level Barriers
Our model suggests that system- and individual-level
barriers to minority enrollment in clinical trials affect whether and how trials are discussed during
doctor–patient clinical interactions. This suggestion is
based on our work and that of others showing that patient and physician attitudes affect the quality of communication during clinical interactions with African
Americans.80-88 Our model also partly explains why
communication during these interactions is often of a
lower quality and may affect racial and ethnic minorities’ decisions about enrollment.
For example, implicit bias among health care professionals (eg, oncologists) negatively affects communication with minority patients.89 An investigation of the
impact of the implicit racial bias of oncologists on their
clinical interactions with African Americans with cancer revealed that oncologists with higher rates of implicit bias against African Americans had shorter interactions with African Americans than oncologists with
lower levels of implicit bias.90 In addition, oncologists
with higher rates of implicit bias used less patient-centered communication.90 Although this study was not
specifically focused on clinical trials, the findings suggest that implicit bias among health care professionals
may affect clinical interactions with African Americans
who are eligible or express interest in participating in a
clinical trial.90
Racial and ethnic differences in the quality of communication are due to both patient-related communication behaviors (eg, African Americans often ask
fewer questions),82,86 general communication behaviors
among physicians, including amount of patient centeredness, information giving, and shared decisionmaking,61,81,86,91 and communication specific to discussing trials.45,63,92-95 Although few researchers have
specifically focused on discussions with minority patients about enrolling in cancer trials, a single study of
video-recorded interactions was undertaken in which
oncologists discussed clinical trials with whites and African Americans.96 Using linguistic analyses, the interactions with African Americans compared with whites
were found to be shorter, the topic of clinical trials was
less frequently mentioned, and, when clinical trials
were mentioned, less time was spent discussing them.96
Compared with whites, differences were observed in
the discussion of some of the key aspects of clinical trials: Oncologists and African American patients spent
less time discussing the purpose of the trial, its risks
and benefits, and alternatives to participating in the
trial; however, they spent more time discussing the volOctober 2016, Vol. 23, No. 4
untary nature of trials.96 These findings are particularly
problematic because very few oncologists are African
American, and so racially discordant interactions may
be considered the norm for African American patients
with cancer.96,97
Few patient-focused interventions designed to improve doctor–patient discussions of clinical trials exist,
and none of them focus on minority patient populations.98 One study assessed an intervention designed
to improve patient understanding of early-phase clinical trials for which they were eligible and which would
be discussed during a subsequent oncology visit.99 Prior to the visit with the oncologist to discuss the trial,
study participants in the intervention group viewed an
interactive, computer-based presentation about clinical trials that included information about several aspects of clinical trials and urged them to discuss the
risks and benefits of the trial with their oncologist;
those assigned to the control group were provided a
pamphlet that covered similar topics.99 Following the
clinic visit, participants assigned to the intervention
were more likely than the controls to understand that
the purpose of an early-phase trial was related to the
safety and dosing of the drug; however, the majority
of participants in both groups reported that the main
purpose of an early-phase trial was “to see if the drug
works.”99 In addition, those in the intervention group
were also more likely than the control group to believe
that the physician talked to them about joining an early-phase trial because they might benefit from the drug
compared with controls. Brown et al100 developed and
pilot tested another patient-focused intervention that
provided eligible patients with a booklet with trial-related questions they could ask their oncologist during
the clinic visit. Findings suggested that the study patients had high informational needs prior to the visit,
selected most of the questions to ask their oncologist,
and asked many of the questions during the visit.100 Although these interventions were not tested in populations with minority patients, they could potentially be
Individual Level
Health Care Professional
Training to address
negative attitudes and
biases to improve communication about clinical
trials
Patient/Family
Question prompts
to encourage active
patient participation in
clinical interaction
Interpersonal
Level
Team building
adapted for specific populations.
The clinical interaction is based on a relationship,
so focus on the social interaction between patients and
physicians from differing social backgrounds may be
valuable. Research from the field of social psychology
suggests that patients and physicians in racially discordant clinical interactions should create a sense of common purpose, support, and understanding as ways to
reduce bias and increase cooperation and trust.101,102
This approach formed the basis of a “team” intervention in a primary care clinic that succeeded in increasing patient trust and adherence following such clinic
visits.103 Thus, this type of approach may reduce the
impact of racial bias in interactions when clinical trials
could be discussed.
Proposed Multilevel Interventional Model
Our multilevel model and supporting research identify
and describe factors that may inhibit the enrollment of
minorities in clinical trials. Authors have challenged
researchers to move beyond reductionist, single-level
interventions.25,26,104 We have met this challenge by
proposing a multilevel intervention comprised of evidence-based strategies focused at the patient, oncologist, and patient–oncologist interpersonal levels. Although the proposed intervention does not directly
address the system level, addressing these other 2 levels may, in combination, have a greater and longer-lasting impact on increasing enrollment among minority
patients (Fig 2). Ideally, an intervention should address
all levels; however, in reality, it may be difficult for all
interventions to address all of them.
Our proposed intervention builds on previously
published descriptive and interventional research on
individual attitudes and clinical communication by
addressing these 3 levels of barriers. The intervention
places emphasis on the communication about clinical
trials that occurs between patients and physicians in
clinical settings. We place the focus on clinical communication for 3 reasons:
High-quality
communication about
clinical trials
Greater rates of
informed racial
and ethnic minority
participation in
clinical trials
Reduced disparities,
improved cancer
treatments
Fig 2. — Our proposed model for a system-level intervention.
October 2016, Vol. 23, No. 4
Cancer Control 333
• The quality of doctor–patient communication during clinic visits is considered the most central and
proximal influence on a patient’s decision to participate in a clinical trial.
• Health care is transacted through these interpersonal processes among health care organizations,
health care professionals, and patients and their
families.105-107
• The quality of communication during clinical interactions with minority patients is less than that
seen among similar interactions with whites.80-88
However, even multilevel interventions will have limited success if they occur in a vacuum; in other words,
they must be implemented within institutions with the
appropriate infrastructure to support trials that match
the clinical characteristics of an engaged, diverse patient population.
Our proposed multilevel intervention illustrates
some potential ways to influence the attitudes and communication skills of minorities related to engaging in
discussion about clinical trials with oncologists; the attitudes and communication skills of oncologists when
discussing trials with minority patients; and doctor–patient interactions in which trials may be discussed.
The first aspect of the intervention is a patient-focused communication intervention called a questionprompt list. A question-prompt list is a list of questions
related to the physical and psychosocial aspects of a
medical condition that patients may wish to ask their
physician during a medical visit. Question-prompt lists
are designed as a simple, inexpensive way to support
patients in gaining information about their diagnosis
and treatment and improve doctor–patient communication by encouraging patients to actively participate in
their health care (eg, ask questions, state concerns).108-111
Active participation has been shown to influence the
amount of information physicians provide, treatments
they recommend, topics discussed, and patient psychosocial and physical health outcomes.112-116 Questionprompt lists have been developed and tested in several
medical settings, and, given the research showing that
racially discordant interactions are often characterized
by poor-quality communication, it is surprising that
this type of intervention has not been tested in this context.98,100,117,118 We have collaborated with community
members, African American patients and their families,
and oncologists to develop a question-prompt list for
use in the context of racially discordant oncology treatment interactions.119 Preliminary findings from a randomized trial of the question-prompt list show that its
use was feasible and acceptable in this patient population, and it was successful in improving their level of active participation.120
For the purpose of a multilevel intervention relevant to minority enrollment in clinical trials, we suggest adapting a question-prompt list developed for use
334 Cancer Control
in the context of clinical trials, although it has never
been tested among minority patients.100 This questionprompt list includes 33 questions divided into 11 categories (eg, finding out more about the trial, understanding the trial’s purpose and background, understanding
possible risks). Adapting the question-prompt list for
use in a diverse population of patients will require engaging community members and patients to ensure
that it reflects findings from focus groups and other research with individuals representative of the patients
likely to be the recipients of the intervention.121
The second aspect of the intervention is intended
to enhance the patient-focused intervention by improving physician attitudes and communication skills
related to discussing trials with minority patients. The
physician-focused intervention would include 2 components, one focused on communication skills and
the other focused on trial-related attitudes. The communication component would build on prior work on
training physicians to improve physician communication skills related to discussing trials but would also
involve increasing physician knowledge about the fact
that most patients, including racial and ethnic minority
groups, are willing to participate in trials if their physician offers a trial and does so using high-quality, patient-centered communication.45,63,67,92,122
In clinical communication, participants exchange
both informational and relational messages, and the
training intervention would involve skill-building in
both.92 Skill-building in informational communication
would include guidelines for discussing information patients need to make an informed decision about participating in a trial based on international guidelines.123,124
Skill-building in relational communication would include explanations and illustrations of communication
strategies such as using organizing statements, eliciting
questions and concerns by utilizing strategies such as
the ask–tell–ask method, using plain language rather
than technical jargon, assessing understanding by using
the teach-back method, directly acknowledging and empathically responding to questions and concerns, and
using shared–decision-making principles.125-127
The attitudinal component of the physician-focused intervention would occur following the communication component. This aspect of the intervention would be designed to increase the likelihood that
physicians will discuss and offer trials to their minority
patients. Asking people to think about their attitudes
affects both their attitudes and relevant behavior, and
asking people to form a situation-specific plan for a
type of behavior, such as discussing a trial with a patient, increases the likelihood they will engage in the
behavior.128-130 Our proposed intervention translates
that research into an intervention in which physicians
are provided with a brief e-mail message prior to a visit
with a patient potentially eligible for a trial. In the mesOctober 2016, Vol. 23, No. 4
sage, physicians would be asked to rate the scientific
and clinical benefits of offering a trial to minority patients and to consider exactly how they will discuss the
trial with the patient.
The interpersonal aspect, which is the third aspect
of the intervention, involves both patients and physicians. It is based on research that suggests the encouragement of patients and physicians to create a sense of
common purpose, support, and understanding may
assist in reducing bias and increasing cooperation and
trust.101,102 Based on this research, members of our research group tested a team intervention in a primary
care clinic that successfully increased patient trust and
adherence following racially discordant clinic visits.103
A similar team intervention could be provided to physicians who have the ability to offer trials to a diverse
population of patients as well as their patients who are
potentially eligible for an ongoing trial. Patients and oncologists would receive simple instructions about working together as a team to achieve a common goal —
providing high-quality care to treat cancer — and they
would be provided with team items, such as pens and
buttons, with a team logo. This type of intervention may
overcome — or reduce — some of the effects of patient
mistrust of physicians and researchers, physician implicit bias toward minorities, and concerns about harming
the therapeutic relationship with these patients.
Conclusions
Although racial and ethnic disparities in cancer and
other conditions have been documented for decades,
researchers, health care professionals, and policymakers have been unable to eliminate these persistent and
preventable contributors to poor health.17,23 Underenrollment of racial and ethnic minority populations
in clinical trials is a health care disparity that results
from preventable and interlinked policies, practices,
and barriers at the system, individual, and interpersonal levels.23 Although many interventions designed
to increase the clinical trial enrollment of racial and
ethnic minorities and other under-represented groups
have experienced varying levels of success, multilevel
interventions are likely to be the most effective method for increasing enrollment among a well-informed,
diverse population of patients.25 When members of
these groups decide to enroll in a clinical trial based
on a supportive and efficient system-level environment
and high-quality communication with their health care
professionals, medical researchers will be able to move
toward their goal of translating new knowledge into
tangible benefits and providing high-quality cancer
care for all patients.
References
1. Newman LA, Roff NK, Weinberg AD. Cancer clinical trials accrual:
missed opportunities to address disparities and missed opportunities to
October 2016, Vol. 23, No. 4
improve outcomes for all. Ann Surg Oncol. 2008;15(7):1818-1819.
2. Nass SJ, Moses HL, Mendelsohn, eds; Committee on Cancer Clinical
Trials; NCI Cooperative Group Program; Institute of Medicine. A National
Cancer Clinical Trials System for the 21st Century: Reinvigorating the
NCI Cooperative Group Program. Washington, DC: National Academies
Press; 2010.
3. Murthy VH, Krumholz HM, Gross CP. Participation in cancer clinical
trials: race-, sex-, and age-based disparities. JAMA. 2004;291(22):2720-2726.
4. Stewart JH, Bertoni AG, Staten JL, et al. Participation in surgical
oncology clinical trials: gender-, race/ethnicity-, and age-based disparities.
Ann Surg Oncol. 2007;14(12):3328-3334.
5. Cheng SK, Dietrich MS, Dilts DM. A sense of urgency: evaluating the
link between clinical trial development time and the accrual performance of cancer therapy evaluation program (NCI-CTEP) sponsored studies. Clin Cancer
Res. 2010;16(22):5557-5563.
6. Korn EL, Freidlin B, Mooney M, et al. Accrual experience of National
Cancer Institute Cooperative Group phase III trials activated from 2000 to
2007. J Clin Oncol. 2010;28(35):5197-5201.
7. Penner LA, Manning M, Eggly S, et al. Prosocial behavior in cancer
research: patient participation in cancer clinical trials. In: Graziano B,
Schroeder D, eds. Handbook of Prosocial Behavior. Oxford, UK: Oxford
University Press; 2014.
8. Minasian LM, Carpenter WR, Weiner BJ, et al. Translating research
into evidence-based practice: the National Cancer Institute Community
Clinical Oncology Program. Cancer. 2010;116(19):4440-4449.
9. Du W, Gadgeel SM, Simon MS. Predictors of enrollment in lung
cancer clinical trials. Cancer. 2006;106(2):420-425.
10. Al-Refaie WB, Vickers SM, Zhong W, et al. Cancer trials versus the
real world in the United States. Ann Surg. 2011;254(3):438-442.
11. Freedman LS, Simon R, Foulkes MA, et al. Inclusion of women and
minorities in clinical trials and the NIH Revitalization Act of 1993—the
perspective of NIH clinical trialists. Control Clin Trials. 1995;16(5):277-285.
12. Kwiatkowski K, Coe K, Bailar JC, et al. Inclusion of minorities and
women in cancer clinical trials, a decade later: have we improved? Cancer.
2013;119(16):2956-2963.
13. Unger JM, Hershman DL, Albain KS, et al. Patient income level and
cancer clinical trial participation. J Clin Oncol. 2013;31(5):536-542.
14. Baquet CR, Commiskey P, Daniel Mullins C, et al. Recruitment and
participation in clinical trials: socio-demographic, rural/urban, and health
care access predictors. Cancer Detect Prev. 2006;30(1):24-33.
15. Paskett ED, Cooper MR, Stark N, et al. Clinical trial enrollment of
rural patients with cancer. Cancer Pract. 2002;10(1):28-35.
16. Vanderpool RC, Kornfeld J, Mills L, et al. Rural-urban differences
in discussions of cancer treatment clinical trials. Patient Educ Couns.
2011;85(2):e69-e74.
17. Siegel R, Ward E, Brawley O, et al. Cancer statistics, 2011: the
impact of eliminating socio-economic and racial disparities on premature
cancer deaths. CA Cancer J Clin. 2011;61(4):212-236.
18. American Cancer Society. Cancer Facts & Figures 2016. Atlanta, GA:
American Cancer Society. http://www.cancer.org/acs/groups/content/@
research/documents/document/acspc-047079.pdf. Accessed September 13, 2016.
19. American Cancer Society. Cancer Facts & Figures for African Americans
2016-2018. Atlanta, GA: American Cancer Society. http://www.cancer.org/acs/
groups/content/@editorial/documents/document/acspc-047403.pdf. Accessed
September 13, 2016.
20. Banda DR, St. Germain D, McCaskill-Stevens W, et al. A critical review of the enrollment of black patients in cancer clinical trials. ASCO Org.
2012:153-157.
21. Carpenter WR, Tyree S, Wu Y, et al. A surveillance system for monitoring, public reporting, and improving minority access to cancer clinical trials.
Clin Trials. 2012;9(4):426-435.
22. Unger JM, Gralow JR, Albain KS, et al. Patient income level and
cancer clinical trial participation: a prospective survey study. JAMA Oncol.
2016;2(1):137-139.
23. Smedley BD, Stith AY, Nelson AR, eds; Committee on Understanding and Eliminating Racial and Ethnic Disparities in Health Care; Board on
Health Sciences Policy; Institute of Medicine. Unequal Treatment: Confronting Racial and Ethnic Disparities in Health Care. Washington, DC: National
Academies Press; 2003.
24. Bolen S, Tilburt J, Baffi C, et al. Defining “success” in recruitment
of underrepresented populations to cancer clinical trials: moving toward a
more consistent approach. Cancer. 2006;106(6):1197-1204.
25. Taplin SH, Anhang Price R, Edwards HM, et al. Understanding and
influencing multilevel factors across the cancer care continuum. J Natl
Cancer Inst Monogr. 2012;2012(44):2-10.
26. Clauser SB, Taplin SH, Foster MK, et al. Multilevel intervention research: lessons learned and pathways forward. J Natl Cancer Inst Monogr.
2012;2012(44):127-133.
27. Kozlowski SWJ, Klein KJ. A multilevel approach to theory and research in organizations: contextual, temporal, and emergent processes.
In: Klein KJ, Koszlowski SWJ, eds. Multilevel Theory, Research, and Methods
in Organizations: Foundations, Extensions, and New Directions. San
Francisco: Jossey-Bass; 2000.
Cancer Control 335
28. Diehl KM, Green EM, Weinberg A, et al. Features associated with
successful recruitment of diverse patients onto cancer clinical trials: report
from the American College of Surgeons Oncology Group. Ann Surg Oncol.
2011;18(13):3544-3550.
29. Ford JG, Howerton MW, Lai GY, et al. Barriers to recruiting underrepresented populations to cancer clinical trials: a systematic review. Cancer.
2008;112(2):228-242.
30. Langford AT, Resnicow K, Dimond EP, et al. Racial/ethnic differences in clinical trial enrollment, refusal rates, ineligibility, and reasons for
decline among patients at sites in the National Cancer Institute’s Community
Cancer Centers Program. Cancer. 2014;120(6):877-884.
31. Bruner DW, Jones M, Buchanan D, et al. Reducing cancer disparities for minorities: a multidisciplinary research agenda to improve patient
access to health systems, clinical trials, and effective cancer therapy. J Clin
Oncol. 2006;24(14):2209-2215.
32. McCaskill-Stevens W, McKinney MM, Whitman CG, et al. Increasing
minority participation in cancer clinical trials: the minority-based community
clinical oncology program experience. J Clin Oncol. 2005;23(22):5247-5254.
33. Somkin CP, Altschuler A, Ackerson L, et al. Organizational barriers to physician participation in cancer clinical trials. Am J Manag Care.
2005;11(7):413-421.
34. Wong YN, Schluchter MD, Albrecht TL, et al. Financial concerns
about participation in clinical trials among patients with cancer. J Clin Oncol.
2016;34(5):479-487.
35. Sateren WB, Trimble EL, Abrams J, et al. How socio-demographics,
presence of oncology specialists, and hospital cancer programs affect accrual
to cancer treatment trials. J Clin Oncol. 2002;20(8):2109-2117.
36. Farmer DF, Jackson SA, Camacho F, et al. Attitudes of African
American and low socio-economic status white women toward medical
research. Health Care Poor Underserved. 2007;18(1):85-99.
37. Paskett ED, Reeves KW, McLaughlin JM, et al. Recruitment of minority and underserved populations in the United States: the Centers for
Population Health and Health Disparities experience. Contemp Clin Trials.
2008;29(6):847-861.
38. Skloot R. The Immortal Life of Henrietta Lacks. New York: Broadway
Books; 2011.
39. Katz RV, Kegeles SS, Kressin NR, et al. Awareness of the Tuskegee syphilis study and the US presidential apology and their influence
on minority participation in biomedical research. Am J Public Health.
2008;98(6):1137-1142.
40. Shavers VL, Lynch CF, Burmeister LF. Racial differences in factors
that influence the willingness to participate in medical research studies.
Ann Epidemiol. 2002;12(4):248-256.
41. Weiner BJ, Jacobs SR, Minasian LM, et al. Organizational designs
for achieving high treatment trial enrollment: a fuzzy set analysis of the
Community Clinical Oncology Program. J Oncol Pract. 2012;8(5):287-291.
42. Wujcik D, Wolff SN. Recruitment of African Americans to national
oncology clinical trials through a clinical trial shared resource. J Health
Care Poor Underserved. 2010;21(1 suppl):38-50.
43. McCaskill-Stevens W, Lyss AP, Good M, et al. The NCI Community
Oncology Research Program: what every clinician needs to know. Am Soc
Clin Oncol Educ Book. 2013.
44. Brooks SE, Muller CY, Robinson W, et al. Increasing minority enrollment onto clinical trials: practical strategies and challenges emerge from
the NRG Oncology Accrual Workshop. J Oncol Pract. 2015;11(6):486-490.
45. Brown R, Albrecht TL. Enrollment in clinical trials. In: Kissane D,
Bultz BD, Butow PN, et al, eds. Handbook of Communication in Oncology
and Palliative Care. Oxford, UK: Oxford University Press; 2010.
46. Howerton MW, Gibbons MC, Baffi CR, et al. Provider roles in the recruitment of underrepresented populations to cancer clinical trials. Cancer.
2007;109(3):465-476.
47.Henderson GE, Churchill LR, Davis AM, et al. Clinical trials
and medical care: defining the therapeutic misconception. PLoS Med.
2007;4(11):e324.
48. Pinto HA, McCaskill-Stevens W, Wolfe P, et al. Physician perspectives on increasing minorities in cancer clinical trials: an Eastern Cooperative Oncology Group (ECOG) initiative. Ann Epidemiol. 2000;10(8
suppl):S78-S84.
49. Hagiwara N, Penner LA, Gonzalez R, et al. Racial attitudes, physician-patient talk time ratio, and adherence in racially discordant medical
interactions. Soc Sci Med. 2013;87:123-131.
50. Cooper LA, Roter DL, Carson KA, et al. The associations of clinicians’ implicit attitudes about race with medical visit communication and patient
ratings of interpersonal care. Am J Public Health. 2012;102(5):979-987.
51. Blair IV, Steiner JF, Fairclough DL, et al. Clinicians’ implicit ethnic/
racial bias and perceptions of care among black and Latino patients. Ann
Fam Med. 2013;11(1):43-52.
52. Penner LA, Dovidio JF, West TV, et al. Aversive racism and medical interactions with Black patients: a field study. J Exp Soc Psychol.
2010;46(2):436-440.
53. van Ryn M, Burgess D, Malat J, et al. Physicians’ perceptions of
patients’ social and behavioral characteristics and race disparities in treatment recommendations for men with coronary artery disease. Am J Public
Health. 2006;96(2):351-357.
336 Cancer Control
54. Moskowitz D, Thom DH, Guzman D, et al. Is primary care providers’
trust in socially marginalized patients affected by race? J Gen Intern Med.
2011;26(8):846-851.
55. Moskowitz GB, Stone J, Childs A. Implicit stereotyping and medical decisions: unconscious stereotype activation in practitioners’ thoughts
about African Americans. Am J Public Health. 2012;102(5):996-1001.
56. Sabin JA, Greenwald AG. The influence of implicit bias on treatment recommendations for 4 common pediatric conditions: pain, urinary
tract infection, attention deficit hyperactivity disorder, and asthma. Am J
Public Health. 2012;102(5):988-995.
57. Sabin JA, Rivara FP, Greenwald AG. Physician implicit attitudes
and stereotypes about race and quality of medical care. Med Care.
2008;46(7):678-685.
58. Joseph G, Dohan D. Diversity of participants in clinical trials in an
academic medical center: the role of the ‘Good Study Patient?’. Cancer.
2009;115(3):608-615.
59. Simon MS, Du W, Flaherty L, et al. Factors associated with breast
cancer clinical trials participation and enrollment at a large academic medical
center. J Clin Oncol. 2004;22(11):2046-2052.
60. Penner LA, Blair IV, Albrecht TL, et al. Reducing racial health care
disparities: a social psychological analysis. Policy Insights Behav Brain
Sci. 2014;1:204-212.
61. Epstein RM, Street RL Jr. Patient-Centered Communication in Cancer
Care: Promoting Healing and Reducing Suffering. NIH publication no. 07-6225.
Bethesda, MD: National Cancer Institute; 2007.
62. Kissane DW, Bylund CL, Banerjee SC, et al. Communication skills
training for oncology professionals. J Clin Oncol. 2012;30(11):1242-1247.
63. Brown RF, Butow PN, Boyle F, et al. Seeking informed consent to
cancer clinical trials: evaluating the efficacy of doctor communication skills
training. Psychooncology. 2007;16(6):507-516.
64. Gross CP, Herrin J, Wong N, et al. Enrolling older persons in cancer
trials: the effect of socio-demographic, protocol, and recruitment center
characteristics. J Clin Oncol. 2005;23(21):4755-4763.
65. Manne S, Kashy D, Albrecht T, et al. Attitudinal barriers to participation in oncology clinical trials: factor analysis and correlates of barriers. Eur J
Cancer Care (Engl). 2015;24(1):28-38.
66. Rivers D, August EM, Sehovic I, et al. A systematic review of the
factors influencing African Americans’ participation in cancer clinical trials.
Contemp Clin Trials. 2013;35(2):13-32.
67. Wendler D, Kington R, Madans J, et al. Are racial and ethnic minorities less willing to participate in health research? PLoS Med. 2006;3(2):e19.
68. Svensson K, Ramirez OF, Peres F, et al. Socio-economic determinants associated with willingness to participate in medical research among
a diverse population. Contemp Clin Trials. 2012;33(6):1197-1205.
69. Katz RV, Kegeles SS, Kressin NR, et al. The Tuskegee Legacy Project:
willingness of minorities to participate in biomedical research. J Health
Care Poor Underserved. 2006;17(4):698-715.
70. Meropol NJ, Wong YN, Albrecht T, et al. Randomized trial of a
web-based intervention to address barriers to clinical trials. J Clin Oncol.
2016;34(5):469-478.
71. Jacobsen PB, Wells KJ, Meade CD, et al. Effects of a brief multimedia psychoeducational intervention on the attitudes and interest of patients
with cancer regarding clinical trial participation: a multicenter randomized
controlled trial. J Clin Oncol. 2012;30(20):2516-2521.
72. Corbie-Smith G, Thomas SB, Williams MV, et al. Attitudes and beliefs of African Americans toward participation in medical research. J Gen
Intern Med. 1999;14(9):537-546.
73. Scharff DP, Mathews KJ, Jackson P, et al. More than Tuskegee:
understanding mistrust about research participation. J Health Care Poor
Underserved. 2010;21(3):879-897.
74. Dovidio JF, Penner LA, Albrecht TL, et al. Disparities and distrust:
the implications of psychological processes for understanding racial disparities in health and health care. Soc Sci Med. 2008;67(3):478-486.
75. Penner LA, Dovidio JF, Edmondson D, et al. The experience of
discrimination and black-white health disparities in medical care. J Black
Psychol. 2009;35(2):180-203.
76. Halbert CH, Armstrong K, Gandy OH Jr, et al. Racial differences in
trust in health care providers. Arch Intern Med. 2006;166(8):896-901.
77. Halbert CH, Weathers B, Delmoor E, et al. Racial differences in
medical mistrust among men diagnosed with prostate cancer. Cancer.
2009;115(11):2553-2561.
78. Boulware L, Cooper L, Ratner L, et al. Race and trust in the health
care system. Public Health Reports. 2003;118:358-365.
79. Duda C, Mahon I, Chen MH, et al. Impact and costs of targeted
recruitment of minorities to the National Lung Screening Trial. Clin Trials.
2011;8(2):214-223.
80. Jean-Pierre P, Fiscella K, Griggs J, et al. Race/ethnicity-based concerns over understanding cancer diagnosis and treatment plan. J Natl Med
Assoc. 2010;102(3):184-189.
81. Street RL Jr, Gordon H, Haidet P. Physicians’ communication and
perceptions of patients: is it how they look, how they talk, or is it just the
doctor? Soc Sci Med. 2007;65(3):586-598.
82. Eggly S, Harper FW, Penner LA, et al. Variation in question asking
during cancer clinical interactions: a potential source of disparities in access
October 2016, Vol. 23, No. 4
to information. Patient Educ Couns. 2011;82(1):63-68.
83. Cooper LA, Roter DL, Johnson RL, et al. Patient-centered communication, ratings of care, and concordance of patient and physician race.
Ann Intern Med. 2003;139(11):907-915.
84. Johnson RL, Roter D, Powe NR, et al. Patient race/ethnicity and
quality of patient-physician communication during medical visits. Am J
Public Health. 2004;94(12):2084-2090.
85. Gordon HS, Street RL Jr, Sharf BF, et al. Racial differences in trust
and lung cancer patients’ perceptions of physician communication. J Clin
Oncol. 2006;24(6):904-909.
86.Gordon HS, Street RL Jr, Sharf BF, et al. Racial differences
in doctors’ information-giving and patients’ participation. Cancer.
2006;107(6):1313-1320.
87. Siminoff LA, Graham GC, Gordon NH. Cancer communication patterns and the influence of patient characteristics: disparities in informationgiving and affective behaviors. Patient Educ Couns. 2006;62(3):355-360.
88. Song L, Hamilton JB, Moore AD. Patient-healthcare provider communication: perspectives of African American cancer patients. Health Psychol.
2012;31(5):539-547.
89. Sabin J, Nosek BA, Greenwald A, et al. Physicians’ implicit and
explicit attitudes about race by MD race, ethnicity, and gender. J Health
Care Poor Underserved. 2009;20(3):896-913.
90. Penner LA, Dovidio JF, Gonzalez R, et al. The effects of oncologist implicit racial bias in racially discordant oncology interactions. J Clin
Oncol. 2016;34(24):2874-2880.
91. Janz NK, Wren PA, Copeland LA, et al. Patient-physician concordance: preferences, perceptions, and factors influencing the breast cancer
surgical decision. J Clin Oncol. 2004;22(15):3091-3098.
92. Albrecht TL, Eggly SS, Gleason ME, et al. Influence of clinical communication on patients’ decision making on participation in clinical trials. J Clin
Oncol. 2008;26(16):2666-2673.
93. Eggly S, Albrecht TL, Harper FW, et al. Oncologists’ recommendations of clinical trial participation to patients. Patient Educ Couns.
2008;70(1):143-148.
94. Barton E, Eggly S. Ethical or unethical persuasion? The rhetoric of
offers to participate in clinical trials. Writ Commun. 2009;26(3):295-319.
95. Kass N, Taylor H, Fogarty L, et al. Purpose and benefits of early
phase cancer trials: what do oncologists say? What do patients hear? J Empir
Res Hum Res Ethics. 2008;3(3):57-68.
96. Eggly S, Barton E, Winckles A, et al. A disparity of words: racial differences in oncologist-patient communication about clinical trials. Health
Expect. 2015;18(5):1316-1326.
97. Hamel LM, Chapman R, Malloy M, et al. Critical shortage of
African American medical oncologists in the United States. J Clin Oncol.
2015;33(32):3697-3700.
98. Street RL Jr, Slee C, Kalauokalani DK, et al. Improving physicianpatient communication about cancer pain with a tailored education-coaching intervention. Patient Educ Couns. 2010;80(1):42-47.
99. Kass NE, Sugarman J, Medley AM, et al. An intervention to improve cancer patients’ understanding of early-phase clinical trials. IRB.
2009;31(3):1-10.
100. Brown RF, Bylund CL, Li Y, et al. Testing the utility of a cancer clinical
trial specific question prompt list (QPL-CT) during oncology consultations.
Patient Educ Couns. 2012;88(2):311-317.
101. Gaertner S, Dovidio JF. A common ingroup identity: a categorization-based approach for reducing intergroup bias. In: Nelson TD, ed.
Handbook of Prejudice, Stereotyping, and Discrimination. Thousand
Oaks, CA: Sage; 2009.
102. Gaertner S, Dovidio JF, Houlette M. Social categorization. In: Dovidio
JF, Hewstone M, Glick P, eds. Handbook of Prejudice, Stereotyping, and
Discrimination. London: Sage; 2010.
103. Penner LA, Gaertner S, Dovidio JF, et al. A social psychological
approach to improving the outcomes of racially discordant medical interactions. J Gen Intern Med. 2013;28(9):1143-1149.
104. Stange KC, Breslau ES, Dietrich AJ, et al. State-of-the-art and future
directions in multilevel interventions across the cancer control continuum.
J Natl Cancer Inst Monogr. 2012;2012(44):20-31.
105. Albrecht TL, Penner LA, Cline RJ, et al. Studying the process of
clinical communication: issues of context, concepts, and research directions. J Health Commun. 2009;14(suppl 1):47-56.
106. McCormack LA, Treiman K, Rupert D, et al. Measuring patient-centered communication in cancer care: a literature review and the development of a systematic approach. Soc Sci Med. 2011;72(7):1085-1095.
107.Epstein RM, Street RL Jr. Patient-Centered Communication in
Cancer Care: Promoting Healing and Reducing Suffering. NIH publication
no. 07-6225. Bethesda, MD: National Cancer Institute; US Department of
Health and Human Services, National Institutes of Health; 2007.
108. Sansoni JE, Grootemaat P, Duncan C. Question Prompt Lists in health
consultations: a review. Patient Educ Couns. 2015. Epub ahead of print.
109. Brandes K, Linn AJ, Butow PN, et al. The characteristics and
effectiveness of question prompt list interventions in oncology: a systematic review of the literature. Psychooncology. 2015;24(3):245-252.
110. Dimoska A, Tattersall MH, Butow PN, et al. Can a “prompt list” empower
cancer patients to ask relevant questions? Cancer. 2008;113(2):225-237.
October 2016, Vol. 23, No. 4
111. Henselmans I, de Haes HC, Smets EM. Enhancing patient participation in oncology consultations: a best evidence synthesis of patient-targeted interventions. Psychooncology. 2013;22(5):961-977.
112. Dimoska A, Butow PN, Dent E, et al. An examination of the initial
cancer consultation of medical and radiation oncologists using the Cancode
interaction analysis system. Br J Cancer. 2008;98(9):1508-1514.
113. Cegala DJ, Street RL Jr, Clinch CR. The impact of patient participation
on physicians’ information provision during a primary care medical interview.
Health Commun. 2007;21(2):177-185.
114. Street RL, Jr, Tancredi DJ, Slee C, et al. A pathway linking patient
participation in cancer consultations to pain control. Psychooncology.
2014;23(10):1111-1117.
115. Clayton JM, Butow PN, Tattersall MH, et al. Randomized controlled
trial of a prompt list to help advanced cancer patients and their caregivers to ask questions about prognosis and end-of-life care. J Clin Oncol.
2007;25(6):715-723.
116. Greene J, Hibbard JH. Why does patient activation matter? An
examination of the relationships between patient activation and healthrelated outcomes. J Gen Intern Med. 2012;27(5):520-526.
117. Clayton J, Butow P, Tattersall M, et al. Asking questions can help:
development and preliminary evaluation of a question prompt list for palliative care patients. Br J Cancer. 2003;89(11):2069-2077.
118. Cegala DJ, Chisolm DJ, Nwomeh BC. A communication skills intervention for parents of pediatric surgery patients. Patient Educ Couns.
2013;93(1):34-39.
119. Eggly S, Tkatch R, Penner LA, et al. Development of a question
prompt list as a communication intervention to reduce racial disparities in
cancer treatment. J Cancer Educ. 2013;28(2):282-289.
120. Eggly S, Hamel LM, Foster T, et al. Using a question prompt list to
increase patient active participation in racially discordant cancer interactions. Presented at: 13th International Conference on Communication in
Healthcare; October 25–28, 2015; New Orleans, LA.
121. Brown RF, Cadet DL, Houlihan RH, et al. Perceptions of participation in a phase I, II, or III clinical trial among African American patients with
cancer: what do refusers say? J Oncol Pract. 2013;9(6):287-293.
122. Bylund CL, Brown R, Gueguen JA, et al. The implementation and
assessment of a comprehensive communication skills training curriculum
for oncologists. Psychooncology. 2010;19(6):583-593.
123.Wendler D, Grady C. What should research participants understand to understand they are participants in research? Bioethics.
2008;22(4):203-208.
124. Council for International Organizations of Medical Sciences
(CIOMS); World Health Organization. International Ethical Guidelines for
Biomedical Research Involving Human Subjects. Geneva: CIOMS; 2002.
125. Back AL, Arnold RM, Baile WF, et al. Approaching difficult communication tasks in oncology. CA Cancer J Clin. 2005;55(3):164-177.
126. Kemp EC, Floyd MR, McCord-Duncan E, et al. Patients prefer the
method of “tell back-collaborative inquiry” to assess understanding of
medical information. J Am Board Fam Med. 2008;21(1):24-30.
127. Makoul G, Clayman ML. An integrative model of shared decision
making in medical encounters. Patient Educ Couns. 2006;60(3):301-312.
128. Young AI, Fazio RH. Attitude accessibility as a determinant of object construal and evaluation. J Exp Soc Psychol. 2013;49(3):404-418.
129. Gollwitzer P. The volitional benefits of planning. In: Gollwitzer P,
Bargh J, eds. The Psychology of Action: Linking Cognition and Motivation
to Behaviour. New York: Guilford Press; 1996.
130. Gollwitzer PM. Implementation intentions: strong effects of simple
plans. Am Psychologist. 1999;54(7):493-503.
Cancer Control 337
Lung carcinogenesis is a multifactorial
process that can affect racial and
ethnic groups differently.
Fiddle Head. X-Ray image courtesy of Bryan Whitney. www.x-rayphotography.com.
Racial and Ethnic Differences in the Epidemiology and
Genomics of Lung Cancer
Matthew B. Schabath, PhD, W. Douglas Cress, PhD, and Teresita Muñoz-Antonia, PhD
Background: Lung cancer is the most common cancer in the world. In addition to the geographical and
sex-specific differences in the incidence, mortality, and survival rates of lung cancer, growing evidence suggests that racial and ethnic differences exist.
Methods: We reviewed published data related to racial and ethnic differences in lung cancer.
Results: Current knowledge and substantive findings related to racial and ethnic differences in lung cancer
were summarized, focusing on incidence, mortality, survival, cigarette smoking, prevention and early detection, and genomics. Systems-level and health care professional–related issues likely to contribute to specific
racial and ethnic health disparities were also reviewed to provide possible suggestions for future strategies to
reduce the disproportionate burden of lung cancer.
Conclusions: Although lung carcinogenesis is a multifactorial process driven by exogenous exposures, genetic
variations, and an accumulation of somatic genetic events, it appears to have racial and ethnic differences
that in turn impact the observed epidemiological differences in rates of incidence, mortality, and survival.
Introduction
Lung cancer is the most common cancer worldwide.1,2
In 2012, new diagnoses of lung cancer reached approximately 1.8 million worldwide, accounting for 13%
of the global cancer burden. 2 Among men, lung
cancer remains the most common cancer diagnoFrom the Departments of Cancer Epidemiology (MBS), Molecular
Oncology (WDC), and Tumor Biology Program (TM-A), H. Lee Moffitt
Cancer Center & Research Institute, Tampa, Florida.
Submitted February 19, 2016; accepted May 11, 2016.
Address correspondence to Matthew B. Schabath, PhD, Department
of Cancer Epidemiology, Moffitt Cancer Center, 12902 Magnolia Drive,
MRC-CANCONT, Tampa, FL 33612. E-mail: [email protected]
No significant relationships exist between the authors and the companies/
organizations whose products or services may be referenced in this article.
This work is supported in part by the James & Esther King Biomedical
Research Program (5JK06), National Institutes of Health Grant from
the National Cancer Institute (U54-CA 163068), and by a cancer center
support grant at the H. Lee Moffitt Cancer Center & Research Institute
(P30-CA76292).
338 Cancer Control
sis. Approximately 1.2 million cases among men alone
were diagnosed in 2012.2 In the United States, lung
cancer is the second most common cancer in men after
prostate cancer and the second most common cancer
in women after breast cancer.3 In 2016, an estimated
224,390 new cases of lung cancer will be diagnosed.3
Lung cancer is also the leading cause of cancerrelated death in the United States and accounts for more
deaths than prostate, breast, colorectal, and pancreatic
cancers combined.3 In 2016, an estimated 158,080 deaths
related to lung cancer will occur.3 From 2008 to 2012,
the mortality rate among men was 59.8 per 100,000
and 37.8 per 100,000 persons for women.3
Although lung carcinogenesis is a multifactorial
process driven by exogenous exposures (eg, cigarette
smoking), inherited genetic variations, and an accumulation of somatic genetic events, it also appears to
have racial and ethnic differences. However, many of
these observed differences attributed to underlying raOctober 2016, Vol. 23, No. 4
cial and ethnic disparities in lung cancer are not fully
understood. Health disparities can also occur as a result of systems-level issues, such as access to care, insurance, and hospital-level factors.4-6 Other potential
sources of racial and ethnic disparities are derived
from health care professional–related factors, such as
limited cultural sensitivity, stereotyping, and poor doctor–patient relationships.7 Studies have shown that,
even after controlling for these factors, treatment disparities persist, suggesting that patient-specific factors
may also contribute to lung cancer differences.8,9
In addition to the established geographical and
sex-specific differences in the incidence, mortality, and
survival rates of lung cancer, certain racial and ethnic
differences exist.10,11 Although the terms race and
ethnicity are often inconsistently applied, a commonly used definition describes race as a social construct
that incorporates beliefs about language, history, and
culture, forming the basis on which social identity, traditions, and politics are built.12,13 In the United States,
the race classification scheme used in the 2000 census,
which is often used in biomedical research, includes
5 major groups: white, black/African American, Asian,
Native Hawaiian/Pacific Islander, and American Indian/Alaska Native.14 Broadly, this classification scheme
emphasizes the geographical region of origin of an individual’s ancestry. Ethnicity is a broader construct that
takes into consideration cultural tradition, common
history, religion, and, oftentimes, a shared genetic heritage.13 Thus, to avoid confusion, we will use the terms
(race or ethnicity) as reported in the originally cited
work to describe the population studied.
Epidemiology
Incidence
The incidence rates of lung cancer are generally lower
among women (583,000 new lung cancer diagnoses
in 2012).2 The highest incidence rates occur in eastern
and Central Europe as well as eastern Asia; by contrast,
the lowest rates occur in western and Central Africa.2
The geographical variations for incidence and mortality differ between men and women, largely attributed to geographical differences in cigarette smoking
between the 2 sexes.2,15 The highest incidence rates in
women are observed in North America and northern
Europe, whereas the lowest rates are found in western
and Central Africa.2
In the United States, the incidence rates have been
declining in men in the past 20 years and in women
since the mid-2000s: From 2008 to 2012, they annually
decreased by 3.0% among men and 1.9% among women.3 During this time period, the incidence rate among
men was 76.7 per 100,000 and 54.1 per 100,000 persons for women.3
Race and ethnicity are complex social and cultural constructs, but they are also often associated with
October 2016, Vol. 23, No. 4
socioeconomic status. Racial and ethnic differences in
disease burden can reveal specific issues of a particular population or subpopulation. Racial and ethnic differences in rates of incidence, mortality, and survival
of lung cancer are well documented.10,11 The most comprehensive report in the United States is based on data
from the Surveillance, Epidemiology, and End Results
(SEER) program, which revealed that blacks have higher rates of incidence and mortality than any other racial or ethnic group.10
A report from the Centers for Disease Control and
Prevention assessed potential racial/ethnic disparities
and geographical differences in incidence rates of lung
cancer from 1998 to 2006 using data from SEER and
the National Program of Cancer Registries.11 In this
report, the annual incidence rate of lung cancer was
highest among blacks, followed by whites, American
Indians/Alaska Natives, and Asian/Pacific Islanders.11
Hispanics had lower rates of lung cancer incidence
than non-Hispanic whites.11 In the United States, the
highest rate of lung cancer incidence was found in
the South and the lowest rate of incidence was in the
West.11 Among whites, the highest rate of lung cancer
incidence was in the South; the highest incidence rates
among blacks, American Indians/Alaska Natives, and
Hispanics were in the Midwest; and the highest incidence among Asian/Pacific Islanders was in the West.11
The identification of geographical differences in incidence rates among racial and ethnic populations presents novel opportunities for targeted efforts in primary
prevention and early detection.
Survival
Despite considerable improvements in patient survival
during the past several decades for breast and prostate
cancers, few improvements have been seen in survival
rates of lung cancer. Improvements in survival are lacking because the majority of lung cancers are at an advanced stage by the time a diagnosis is made and treatment options are limited. Because of its high mortality
rate, variability in survival is lacking in different world
regions.2 In the United States, the 5-year relative survival rate for all lung cancers is 17%.3
Among all stages of non–small-cell lung cancer
(NSCLC) and small-cell lung cancer, the combined
5-year relative survival rates are 21% and 6%, respectively.3,16 The overall prognosis for NSCLC remains
poor, and prognostic factors associated with poor survival include late-stage diagnosis, current smoking status, advanced age, male sex, poor pulmonary function,
presence of cardiovascular disease, nonsquamous cell
histology, and pneumonectomy.17-21 Among individuals
with small-cell lung cancer, poor prognosis is associated with age older than 70 years, male sex, relapsed
disease, extensive disease, weight loss of more than
10% of total body weight at diagnosis, and poor perCancer Control 339
formance status.22-26 Despite the poor outcomes associated with a diagnosis of lung cancer, the emergence
of immunotherapy and immune checkpoint inhibitors
has demonstrated durable rates of long-term survival
in select patients.27,28 As such, these therapies may lead
to improved survival rates and, in the early stage, possibly curable disease.29
Published data have demonstrated that the survival rates of those with lung cancer also differ based on
race and ethnicity.4,30-33 Blacks are less likely to receive
surgical resection than whites, a finding that likely contributes to data that show blacks have lower rates of
survival for early-stage NSCLC.4,30-32 Using SEER data
linked to Medicare claims data during 1995 and 1999,
Shugarman et al31 reported that blacks were 66% less
likely to receive timely and appropriate treatment than
whites, and black men were least likely to receive resection (22.0% for black men vs 43.7% for white men).
The authors also reported that blacks were 34.0% less
likely to receive timely surgery, chemotherapy, or radiotherapy for stage III disease and were 51.0% less
likely to receive chemotherapy in a timely fashion for
stage IV disease relative to whites.31 Howington et al33
reported that American Indians and Alaskan Natives
have worse survival rates than non-Hispanic whites.
With regard to potential ethnic differences, a study by
Lin et al34 compared rates of stage-appropriate treatment among blacks, Hispanics, and nonminority patients and did not observe treatment-related disparities
among Hispanics. However, the authors did find that
blacks were less likely than nonminorities to receive
stage-appropriate treatment, similar to data reported in
previous studies.5,34-36
(18.8%) than females (14.8%), and nearly 17% of adults
still continue to smoke cigarettes.41,43
By racial and ethnic groups, smoking prevalence
rates were highest among American Indians/Alaska
Natives (29.2%) and multiracial adults (27.9%) and lowest among Asians (9.5%).41,43 For other racial and ethnic groups, smoking prevalence rates were 18.2% for
whites, 17.5% for blacks, and 11.2% for Hispanics.41,43
Although the annual incidence is highest among
blacks compared with any other race, the prevalence
of smoking among blacks is lower than American Indians/Alaska Natives, multiracial adults, and whites.41,43
Moreover, significant differences have been reported
in the association between cigarette smoking and the
risk of lung cancer among 5 ethnic/racial groups.44
Among those who smoked no more than 30 cigarettes
per day, African Americans and Native Hawaiians had
significantly greater risks of lung cancer than the other
groups studied.44 As such, these data provide further
support for ethnic/racial differences, which could be
attributed to innate genetic differences in the smokingassociated risk of lung cancer.
Other established and putative risk factors for
lung cancer include exposure to secondhand smoke
(ie, passive smoke), history of chronic obstructive pulmonary disease, family history of lung cancer, radon
exposure, and occupational exposure (eg, asbestos,
arsenic, diesel exhaust, chromium).19,45,46 Evidence is
conflicting regarding the impact of exogenous hormones on women, diet, and body mass index in association with risk of lung cancer.46-53 Moreover, few
studies have assessed the potential racial and ethnic
differences for such risk factors.50
Risk Factors
Tobacco smoking is the most important and prevalent risk for lung cancer.37-39 Lung cancer is one of the
first chronic diseases to be causally linked to tobacco
smoking, and approximately 90% of lung cancer diagnoses in the United States are attributed to tobacco
smoking.19,20 Cigarette smoke contains more than 7,000
chemicals, including more than 60 established carcinogens and other toxicants associated with chronic diseases.40 Although approximately 1 in 9 individuals who
smoke develops lung cancer, the relative risk of lung
cancer in those who are long-term smokers is estimated to be between 10- and 30-fold higher than that of a
lifetime never-smoker.19
The percentage of cigarette smoking among
adults in the United States and most Western nations
has been steadily declining during the past several decades — from 20.9% in 2005 to 16.8% in 2014 — and
it continues to decline today due to successful tobacco-control efforts.2,41 In the United States, smoking
rates have steadily declined since the 1960s.42 In 2014,
the prevalence of smoking was higher among males
Prevention and Early Detection
340 Cancer Control
Until recently, no screening method was shown to
decrease the mortality rates for NSCLC. The National
Lung Screening Trial (NLST) randomized 53,452 individuals who were former or current cigarette smokers, between the ages of 55 and 74 years, and had a
pack-year smoking history of at least 30 years into either low-dose helical computed tomography (LDCT)
or standard chest radiography — both arms were administered 3 annual screenings (baseline screening
and 2 follow-up screening sessions approximately
12 months apart).54 After a median follow-up period of
6.4 years, a 20% relative reduction in rate of lung cancer mortality was observed for the LDCT group compared with those assigned to standard chest radiography.54 The rate of lung cancer detected via screening
(ie, diagnosed on follow-up screening intervals) accounted for 58% of all LDCT-detected cases of lung
cancer, were 2.7-fold higher in the LDCT arm vs the
chest radiography arm, were associated with a stage
shift from late-stage to more early-stage lung cancer,
and demonstrated improved rates of 5-year survival
October 2016, Vol. 23, No. 4
compared with cancers diagnosed at the baseline prevalence screening.54
An important benefit of early detection is the ability to detect more cases of early-stage lung cancer. A
meta-analysis revealed that the rate of detection of
stage I lung cancer was 70% with LDCT screening compared with 16% when detected during routine care.55
Thus, the incidence of lung cancer detected in screening and then diagnosed as a result of LDCT have better surgical outcomes and improved 5-year survival
rates than prevalence and routine care–detected lung
cancer diagnosed when patients develop symptoms of
late-stage disease.19 Based on the findings from the
NLST, the US Preventive Services Task Force issued a
recommendation in December 2013 for annual screening for lung cancer using LDCT in adults aged 55 to
80 years who have a 30 pack-year smoking history and
currently smoke or have quit within the past 15 years.54,56
Of the 53,452 participants in the NLST, 90.8%
were white (n = 48,549), 4.4% were black (n = 2,376),
1.7% were Asian (n = 895), 2.0% were of other racial groups or reported more than 1 race or ethnic
group (n = 1,062), and 1.7% were Hispanic or Latino
(n = 935).54 Although the NLST did not provide ethnic-/racial-specific results,54 a posthoc analysis by
Tanner et al57 assessed racial differences in outcomes
between blacks and whites who participated in the
NLST. Although demographics associated with improved survival in lung cancer, such as higher education, former smoking status, and fewer comorbidities, were less commonly found among blacks in the
NLST, their analysis revealed that screening with LDCT
reduced mortality in all racial groups, but more so in
blacks (hazard ratio [HR] 0.61 for blacks vs HR 0.86 for
whites).57 Among all racial groups, Tanner et al57 also
noted that study participants who currently smoked
had worse rates of lung cancer–specific mortality;
however, the risk was 2-fold higher in blacks who
were current smokers compared with white current
smokers. In addition, the rate of all-cause mortality
was 1.35 times higher in blacks vs whites, but blacks
screened with LDCT had a statistically significant reduction in all-cause mortality when compared with
whites.57 As such, screening for lung cancer appears
to be beneficial in blacks, but different strategies are
needed to achieve a significant reduction in lung cancer–related mortality in this population.57 Ongoing
disparities in access to screening and follow-up visits
(in the case of abnormal results) underscore the importance of health care institutions to offer screening and
then quickly resolve any abnormal findings.
In a separate posthoc analysis of the NLST, Kumar
et al58 examined racial differences in smoking behaviors among white and black study participants in the
NLST who were current smokers at screening. They
analyzed data from a follow-up survey on 24-hour and
October 2016, Vol. 23, No. 4
7-day cessation attempts, 6-month continuous abstinence from smoking, and the use of smoking cessation
programs and cessation aids 12 months after screening. The authors reported that blacks were more likely
than whites to have 24-hour and 7-day cessation attempts; however, these attempts did not translate to
increased rates of 6-month continuous abstinence
among blacks.58 Specifically, 1 year after screening,
blacks were more likely to report a 24-hour (52.7% for
blacks vs 41.2% for whites; P < .01) or 7-day (33.6% for
blacks vs 27.2% for whites; P < .01) cessation attempt.58
Although no statistically significant racial differences
were found in 6-month continuous abstinence
(5.6% for blacks vs 7.2% for whites), black race was a
statistically significant predictor in multivariable analyses of a higher likelihood of a 24-hour and 7-day cessation attempt.58 Race was not associated with 6-month,
continuous abstinence; by contrast, a positive result
on screening for lung cancer was the only significantly predictor of successful 6-month, continuous abstinence.58 At present, knowledge is lacking about the effectiveness of screening for lung cancer with LDCT in
other racial and ethnic groups.
The data from the NLST demonstrated that screening with LDCT can mitigate lung cancer mortality via
early detection.55 Screening with LDCT appears to be
second only to primary prevention (ie, smoking prevention/cessation) for mitigating lung cancer–related
mortality — and the single remaining option for those
who have already quit smoking. The risk of lung cancer caused by smoking is reduced following smoking
cessation, but this risk always remains elevated compared with those who are lifetime never-smokers.59
Genomics
The most frequently mutated genes in lung cancer are TP53 (53.6%), KRAS (16.1%), STK11 (9.8%),
EGFR (7.2%), KEAP1 (6.6%), and NFE2L2 (4.5%).60 The
frequency of mutations in oncogenes and tumor-suppressor genes differ across racial and ethnic populations and are selectively shown in the Table. Advances
in tumor genomic profiling have resulted in a paradigm shift, whereby types of lung cancer are characterized and classified by genetic alternations in oncogenes and tumor-suppressor genes critical to tumor
growth and survival and can be exploited with specific
targeted agents.61
The focus of this paper is on specific oncogenes
and tumor-suppressor genes where published studies
have assessed potential racial and ethnic differences. Although other genes have also been studied, including
MET, PI3KCA, PTEN, and ROS1, current data are limited
and suggest that no differences exist in these genes.62
Because most types of lung cancer harbor somatic mutations, alterations, or both, the use of targeted
therapy is important to improve outcomes.63 Research
Cancer Control 341
Table. — Estimated Frequencies of Select Mutated Genes
in Lung Cancer
Gene
Frequency, %
EGFR
By region
Asian populations
30.0–40.0
Western populations
10.0–20.0
By country
Argentina
19.3
Colombia
24.8
Costa Rica
26.0–35.3
Mexico
31.2
Panama
24.8
Peru
67.0
KRAS
By region
Asian populations
Western populations
3.8–8.0
18.0–26.0
STK11
By region
Asian populations
3.0–7.0
Western populations
9.0–17.0
BRAF a
Overall
1.0–3.5
PIK3CAa
Overall
1.0–3.0
ALK Fusion
3.0–7.0
By region
Asian populations
2.3–6.7
Western populations
1.0–3.0
a No
published reports have assessed racial and ethnic differences.
has been limited on the impact of race and ethnicity
in targeted therapies. The impact and frequency of tumor mutations are not as well characterized in blacks
and Hispanics/Latinos to the same extent as they are
in Asians and whites.62,64 Although evidence suggests
that racial and ethnicity differences exist in markers
of susceptibility, we have chosen to focus on somatic
mutations because of their clinical implications and
observed racial and ethnic differences, as well as because germline variations in lung cancer have yet to be
deemed clinically actionable.62,65
EGFR
Epidermal growth factor receptor (EGFR) is a transmembrane protein with cytoplasmic kinase activity
that facilitates critical growth-factor signaling from the
extracellular milieu to the cell.66 EGFR is expressed on
the cell surface. Tyrosine kinase inhibitors can be ef342 Cancer Control
fective therapy for patients whose tumors harbor activating mutations in the tyrosine kinase domain
of EGFR.66 Therefore, mutation testing is routinely
performed to identify patients harboring targetable
EGFR mutations, given that selection based on clinical
and pathological characteristics alone is inadequate.
Overall, approximately 10% of patients with NSCLC
in Western countries and 35% of patients in East Asia
have a tumor that exhibits an EGFR mutation.67,68 The
most frequent EGFR mutations occur in exons 18 to 21,
encoding a portion of the EGFR kinase domain; in addition, EGFR mutations are often heterozygous, with
the mutant allele also showing gene amplification.69 Approximately 90% of these mutations are exon 19 deletions or exon 21 L858R point mutations.70 Irrespective
of ethnicity, EGFR mutations are more often found in
females who are never-smokers and have adenocarcinoma histology.67,68 However, EGFR mutations can also be
found in other histological subtypes of NSCLC, and they
can be found in former and current smokers.71-73
In-frame deletions and insertions in exon 19 and
the point mutation L858R in exon 21 are the most common activating EGFR mutations, and their prevalence
is significantly higher in individuals of Japanese, Korean, and Chinese descent than American or European
whites.74 Based on data from the Iressa Pan-Asia Study,
the frequency of activating EGFR mutations in Asian
never-smokers or light smokers with advanced-stage
adenocarcinoma was nearly 60%.75 By contrast, the
rate of frequency was 15% in non-Asian patients based
on data from phase 3 trials of retrospective EGFR mutation testing of archival samples.76,77 One study compared EGFR mutations in individuals of Asian and Russian descent with advanced NSCLC and reported that
the frequency of EGFR mutations in adenocarcinoma
was 49% in East Asians and 18% in Russians, and the
frequencies of EGFR mutations in nonadenocarcinoma lung cancers were 14% and 4%, respectively.78 Several studies have explored whether the frequencies of
EGFR mutations differ by race and ethnicity, and others have compared the frequencies of EGFR mutations
between blacks and whites with NSCLC; however, the
results are conflicting.79-82 Specifically, Yang et al79 and
Leidner et al80 found a lower frequency of EGFR mutations among blacks, whereas Riely et al,81 Cote et al,82
and Reinersman et al83 did not find a statistically significant difference between the 2 groups.
Few studies have addressed the frequency of
mutations in lung cancer among Hispanics/Latinos.
Arrieta et al84 analyzed 1,150 biopsy specimens from
patients with NSCLC who were from Argentina, Colombia, Peru, and Mexico and found that the combined
frequency of EGFR mutations among all 4 countries
was 32.5%. The frequencies of mutations by country
were 19.3% in Argentina, 24.8% in Colombia, 31.2%
in Mexico, and 67.0% in Peru.84 The high frequency of
October 2016, Vol. 23, No. 4
EGFR mutations in Peru could be attributed to Asian
migration.62 The frequency of EGFR mutations ranges
from 26.0% to 35.3% in Costa Ricans and is 24.8% in
Panamanians.85-87 By contrast, a lower frequency of
EGFR mutations has also been reported among Hispanics/Latinos, and other conflicting reports have
found no statistically significant difference between
Hispanics/Latinos and whites.62
KRAS
The RAS family, which includes HRAS, NRAS, and
KRAS, encodes for membrane-bound guanosine triphosphate–binding proteins that regulate cell growth,
differentiation, and apoptosis by interacting with mitogen-activated protein kinase, phosphoinositide 3-kinase,
and signal transducer and activator of transcription cascades.88,89 When aberrantly activated, KRAS is a potent
oncogenic driver when a point mutation occurs at codon
12 or 13 in exon 2 or codon 61 in exon 3.90 These mutations result in impaired guanosine-triphosphatase activity and a constitutive activation of RAS signaling.91
Mutations in KRAS occur frequently in NSCLC — particularly adenocarcinoma (20%–30%) — and less commonly in squamous cell carcinoma (~ 7%).92,93
Although mutationally activated KRAS tumors were
originally identified in 1982,78 no successful treatment
strategies target these tumors,93 and their impact on survival and prognosis in lung cancer is unclear and controversial.94,95 To date, more than 50 studies have evaluated
KRAS mutations on clinical outcomes in lung cancer, the
results of which are varied and inconsistent.94,95 A metaanalysis of 41 studies concluded that KRAS mutations
are associated with a poor prognosis in patients with
NSCLC, and this was particularly true for those with adenocarcinoma and early-stage NSCLC.95
Data have also suggested that the rates of frequency of KRAS mutations differ by race and ethnicity. Specifically, KRAS mutations in the setting of lung cancer
are less common in Asians compared with whites.96,97
The frequency of KRAS mutations ranged from 3.8%
to 8.0% in studies of Chinese study participants with
NSCLC — a rate lower than that seen in whites (range,
18%–26%).65,80,98 Few studies have assessed KRAS mutational status among blacks, and the published data
are inconsistent: 3 studies reported no statistically significant difference in frequency of KRAS mutations between whites and blacks,80,83,99 and some noted that the
frequency in Hispanics may actually be lower.84,100
STK11
STK11 encodes a tumor suppressor located on chromosome 19p13.3 that encodes the serine/threonine
kinase 11 (STK11) protein. The gene is made up of
coding exons 1 to 9 and a final, noncoding exon 10.
STK11 regulates cellular energy metabolism and
cell polarity by initiating adenosine monophosOctober 2016, Vol. 23, No. 4
phate–activated protein kinase (AMPK) and other
members of the AMPK family.101,102 As a multifunctional kinase, STK11 is involved in a broad spectrum of
cellular activity that includes metabolism, polarity, and
epithelial-mesenchymal transition, as well as cell-cycle
regulation, apoptosis, and autophagy.103 Germline mutations in STK11 were first identified in patients with
Peutz-Jeghers syndrome,104 a rare autosomal dominant disorder associated with an increased risk of gastrointestinal and other malignancies.105 Studies have
also reported that STK11 somatic mutations are common in NSCLC: The prevalence of inactivating mutations can occur in more than 50% of cases (range,
0.6%–44.4%), thus revealing an important role of
STK11 in lung tumorigenesis.103,106-112 Previously published data have reported that STK11 inactivating mutations occur in other histology subtypes of lung cancer and include 19% of squamous cell carcinomas, 14%
of large cell carcinomas, and 25% of adenosquamous
carcinomas.103,109,110,113-115
Similar to KRAS and EGFR, ethnic and racial differences appear in STK11 mutations. Studies in Asian
populations (those of Japanese, Korean, and Chinese
descent) have reported lower rates of STK11 mutations
than whites (range, 3%–7%).115-118 This observation is
similar to that seen in KRAS mutations in the setting
of lung cancer, because they frequently co-occur with
STK11. Lung tumors in Western populations harbor a
higher frequency of KRAS mutations compared with
Asian populations.98,119 Asian populations have also
been found to express an STK11 germline F354L polymorphism at a frequence of approximately 10%.117 This
allele has been called a nonfunctional polymorphism
in lung cancer, but it also affects cell polarity maintenance in an AMPK-dependent manner.120 At present, no data have been published on the frequency of
STK11 mutations among blacks and Hispanics/Latinos.
BRAF
BRAF is a proto-oncogene that belongs to a family of
serine-threonine protein kinases that also includes
ARAF and RAF1. Mutant BRAF has been implicated in
the pathogenesis of several cancers.98 The most commonly identified BRAF mutation is V600E, which accounts for 90% of BRAF mutations in melanoma.121 By
contrast, the frequency of BRAF mutations in NSCLC
ranges from 1.0% to 3.5%.122-125 No reports have assessed racial and ethnic differences of BRAF mutations
in patients with lung cancer, likely because this mutation is rare in this population.
PIK3CA
PIK3CA belongs to a gene family of lipid kinases involved in many cellular processes, including cell
growth, proliferation, differentiation, motility, and
survival. PIK3CA is mutated in more than 30% of
Cancer Control 343
colorectal cancer cases.126 By contrast, mutations in
PIK3CA have been found in 1% to 3% of all types of
NSCLC.126,127 Similar to BRAF, no reports have been
published that have assessed racial and ethnic differences of PIK3CA mutations in the setting of lung
cancer, likely because this mutation is rare in this patient population.
ALK
Anaplastic lymphoma kinase is a tyrosine kinase
receptor abnormally expressed by forming a fusion
gene with one of several other genes, by gaining additional gene copies, or by somatic mutations. The
EML4-ALK fusion was first documented in 2007 in
NSCLC as a potentially novel oncogenic-driver mutant
kinase.128 Approximately 3% to 7% of all lung tumors
harbor ALK fusions, and EML4-ALK fusions are usually found in light- or never-smokers and are typically
diagnosed at a young age.128-132
EML4-ALK rearrangements may differ across
different racial groups. The frequency of translocation ranges from between 2.3% and 6.7% among
Asians.133-135 By contrast, EML4-ALK rearrangement is
lower in whites and ranges from 1.0% to 3.0%.128,131,136
One study analyzed a cohort of NSCLC samples collected from Italy and Spain and found that 7.5% of the
samples expressed EML4-ALK transcripts — a finding
more similar to the data seen in Asians than whites.137
Conclusions
Lung cancer is the most common cancer in the world
and the second most common cancer in both men and
women in the United States.1-3 Although lung carcinogenesis is a multifactorial process driven by exogenous
exposure (eg, cigarette smoking), inherited genetic
variations, and an accumulation of somatic genetic
events, it also appears to have racial and ethnic differences. However, many observed racial and ethnic disparities in lung cancer are not fully understood.
The genomic diversity of oncogenes and tumor-suppressor genes across racial and ethnic groups poses
unique but important challenges for therapeutic opportunities to provide personalized medicine. Molecular genomic profiling for specific alterations is necessary to identify patients likely to benefit from targeted
therapy. As such, identifying novel but actionable targetable mutations exclusive to specific racial and ethnic groups is critical to ensure these populations benefit from such therapies.
Even after smoking cessation is successfully accomplished, former smokers remain at significant risk
of developing lung cancer.19 Although effective smoking cessation programs and early cancer detection will
reduce the overall lung cancer burden, improvements
to the access of these modalities and the affordability
of health care are important topics to address.55 Screen344 Cancer Control
ing with low-dose helical computed tomography can
mitigate the risk because it can detect early-stage lung
cancer.54,55 Identifying and addressing cultural factors
may also help improve the implementation of racially
and ethnicity sensitive, population-based interventions. Identifying and eliminating regional and racial
and ethnic differences in lung cancer could contribute
to more effective, preventive treatment strategies to
help reduce the disproportionate burden of lung cancer in the United States.43
References
1. American Cancer Society. Global Cancer Facts & Figures. 3rd ed.
http://www.cancer.org/acs/groups/content/@research/documents/document/acspc-044738.pdf. Accessed September 1, 2016.
2. International Agency for Research on Cancer; World Health
Organization. GLOBOCAN 2012: estimated cancer incidence, mortality and
prevalence worldwide in 2012. http://globocan.iarc.fr/Pages/fact_sheets_
cancer.aspx. Accessed August 29, 2016.
3. American Cancer Society. Cancer Facts & Figures 2016. http://
www.cancer.org/acs/groups/content/@research/documents/document/
acspc-047079.pdf. Accessed August 29, 2016.
4. Farjah F, Wood DE, Yanez ND, 3rd, et al. Racial disparities among
patients with lung cancer who were recommended operative therapy. Arch
Surg. 2009;144(1):14-18.
5. Steele CB, Pisu M, Richardson LC. Urban/rural patterns in receipt
of treatment for non-small cell lung cancer among black and white Medicare
beneficiaries, 2000-2003. J Natl Med Assoc. 2011;103(8):711-718.
6. Esnaola NF, Gebregziabher M, Knott K, et al. Underuse of surgical
resection for localized, non-small cell lung cancer among whites and African
Americans in South Carolina. Ann Thorac Surg. 2008;86(1):220-226.
7. van Ryn M. Research on the provider contribution to race/ethnicity
disparities in medical care. Med Care. 2002;40(1 suppl):I140-I151.
8. Underwood JM, Townsend JS, Tai E, et al. Racial and regional disparities in lung cancer incidence. Cancer. 2012;118(7):1910-1918.
9. Yang R, Cheung MC, Byrne MM, et al. Do racial or socioeconomic
disparities exist in lung cancer treatment? Cancer. 2010;116(10):2437-2447.
10. Howlader N, Noone AM, Krapcho M, eds. SEER Cancer Statistics
Review, 1975-2012. Bethesda, MD: National Cancer Institute. Based on
November 2014 SEER data submission, posted to the SEER web site, April
2015. http://seer.cancer.gov/csr/1975_2012. Accessed June 30, 2016.
11.Centers for Disease Control and Prevention. Racial/ethnic disparities and geographic differences in lung cancer incidence: 38 states
and the District of Columbia, 1998-2006. MMWR Morb Mortal Wkly Rep.
2010;59(44):1434-1438.
12. Sankar P, Cho MK. Genetics. Toward a new vocabulary of human
genetic variation. Science. 2002;298(5597):1337-1338.
13. Witzig R. The medicalization of race: scientific legitimization of a
flawed social construct. Ann Intern Med. 1996;125(8):675-679.
14. Grieco EM, Cassidy RC. Overview of race and Hispanic origin.
Census 2000 brief. https://www.census.gov/prod/2001pubs/c2kbr01-1.pdf.
Accessed August 29, 2016.
15. Islami F, Torre LA, Jemal A. Global trends of lung cancer mortality
and smoking prevalence. Transl Lung Cancer Res. 2015;4(4):327-338.
16. Harris K, Khachaturova I, Azab B, et al. Small cell lung cancer doubling time and its effect on clinical presentation: a concise review. Clin Med
Insights Oncol. 2012;6:199-203.
17. Schabath MB, Thompson ZJ, Gray JE. Temporal trends in demographics and overall survival of non-small-cell lung cancer patients at Moffitt
Cancer Center from 1986 to 2008. Cancer Control. 2014;21(1):51-56.
18. Kachroo S, Tong L, Spitz MR, et al. Trends in prevalence of prognostic factors and survival in lung cancer patients from 1985 to 2004 at a
tertiary care center. Cancer Detect Prev. 2008;32(2):101-108.
19. Dela Cruz CS, Tanoue LT, Matthay RA. Lung cancer: epidemiology,
etiology, and prevention. Clin Chest Med. 2011;32(4):605-644.
20. Birim O, Kappetein AP, van Klaveren RJ, et al. Prognostic factors in
non-small cell lung cancer surgery. Eur J Surg Oncol. 2006;32(1):12-23.
21.National Cancer Institute. SEER Survival Monograph: Cancer
Survival Among Adults: U.S. SEER Program, 1988-2001, Patient and
Tumor Characteristics. NIH pub. no. 076215, 2007. https://surveillance.
cancer.gov/publications/factsheets/SEER_Cancer_Survivor_Fact_Sheet.pdf.
Accessed August 29, 2016.
22. Paesmans M, Sculier JP, Lecomte J, et al. Prognostic factors for
patients with small cell lung carcinoma: analysis of a series of 763 patients
included in 4 consecutive prospective trials with a minimum follow-up of 5 years.
Cancer. 2000;89(3):523-533.
23. Spiegelman D, Maurer LH, Ware JH, et al. Prognostic factors in
October 2016, Vol. 23, No. 4
small-cell carcinoma of the lung: an analysis of 1,521 patients. J Clin Oncol.
1989;7(3):344-354.
24. Albain KS, Crowley JJ, LeBlanc M, et al. Determinants of improved
outcome in small-cell lung cancer: an analysis of the 2,580-patient Southwest
Oncology Group data base. J Clin Oncol. 1990;8(9):1563-1574.
25. Osterlind K, Andersen PK. Prognostic factors in small cell lung cancer: multivariate model based on 778 patients treated with chemotherapy
with or without irradiation. Cancer Res. 1986;46(8):4189-4194.
26. Schabath MB, Nguyen A, Wilson P, et al. Temporal trends from
1986 to 2008 in overall survival of small cell lung cancer patients. Lung Cancer.
2014;86(1):14-21.
27. Santarpia M, Giovannetti E, Rolfo C, et al. Recent developments in
the use of immunotherapy in non-small cell lung cancer. Expert Rev Respir
Med. 2016;10(7):781-798.
28.Tchekmedyian N, Gray JE, Creelan BC, et al. Propelling immunotherapy combinations into the clinic. Oncology (Williston Park).
2015;29(12):pii:214814.
29. Brahmer JR. Harnessing the immune system for the treatment of
non-small-cell lung cancer. J Clin Oncol. 2013;31(8):1021-1028.
30. Margolis ML, Christie JD, Silvestri GA, et al. Racial differences
pertaining to a belief about lung cancer surgery: results of a multicenter
survey. Ann Intern Med. 2003;139(7):558-563.
31. Shugarman LR, Mack K, Sorbero ME, et al. Race and sex differences in the receipt of timely and appropriate lung cancer treatment. Med
Care. 2009;47(7):774-781.
32. Zullig LL, Carpenter WR, Provenzale DT, et al. The association of
race with timeliness of care and survival among Veterans Affairs health
care system patients with late-stage non-small cell lung cancer. Cancer
Manag Res. 2013;5:157-163.
33. Howington JA, Blum MG, Chang AC, et al. Treatment of stage I and
II non-small cell lung cancer: diagnosis and management of lung cancer,
3rd ed: American College of Chest Physicians evidence-based clinical
practice guidelines. Chest. 2013;143(5 suppl):e278S-e313S.
34. Lin JJ, Mhango G, Wall MM, et al. Cultural factors associated with racial disparities in lung cancer care. Ann Am Thorac Soc. 2014;11(4):489-495.
35. Hardy D, Liu CC, Xia R, et al. Racial disparities and treatment
trends in a large cohort of elderly black and white patients with nonsmall
cell lung cancer. Cancer. 2009;115(10):2199-2211.
36. Hardy D, Xia R, Liu CC, et al. Racial disparities and survival for
nonsmall-cell lung cancer in a large cohort of black and white elderly patients.
Cancer. 2009;115(20):4807-4818.
37. US Department of Health and Human Services. The Health Consequences of Smoking: 50 Years Of Progress. A Report of the Surgeon General.
Atlanta, GA: US 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.
38. Carter BD, Freedman ND, Jacobs EJ. Smoking and mortality:
beyond established causes. N Engl J Med. 2015;372(22):2170.
39. Pesch B, Kendzia B, Gustavsson P, et al. Cigarette smoking and lung
cancer: relative risk estimates for the major histological types from a pooled
analysis of case-control studies. Int J Cancer. 2012;131(5):1210-1219.
40. US Department of Health and Human Services. How Tobacco Smoke
Causes Disease: The Biology and Behavioral Basis for Smoking-Attributable
Disease: A Report of the Surgeon General. Atlanta: US 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; 2010.
41. Jamal A, Homa DM, O’Connor E, et al. Current cigarette smoking
among adults: United States, 2005-2014. MMWR Morb Mortal Wkly Rep.
2015;64(44):1233-1240.
42. Centers for Disease Control and Prevention. Cigarette smoking
among adults: United States, 2006. MMWR Morb Mortal Wkly Rep.
2007;56(44):1157-1161.
43. Jamal A, Agaku IT, O’Connor E, et al. Current cigarette smoking
among adults: United States, 2005-2013. MMWR Morb Mortal Wkly Rep.
2014;63(47):1108-1112.
44. Haiman CA, Stram DO, Wilkens LR, et al. Ethnic and racial differences in the smoking-related risk of lung cancer. N Engl J Med.
2006;354(4):333-342.
45. Driscoll T, Nelson DI, Steenland K, et al. The global burden of disease due to occupational carcinogens. Am J Ind Med. 2005;48(6):419-431.
46. Alberg AJ, Brock MV, Ford JG, et al. Epidemiology of lung cancer: diagnosis and management of lung cancer, 3rd ed: American College
of Chest Physicians evidence-based clinical practice guidelines. Chest.
2013;143(5 suppl):e1S-e29S.
47. Brinton LA, Gierach GL, Andaya A, et al. Reproductive and hormonal factors and lung cancer risk in the NIH-AARP Diet and Health Study
cohort. Cancer Epidemiol Biomarkers Prev. 2011;20(5):900-911.
48. Yao Y, Gu X, Zhu J, et al. Hormone replacement therapy in females
can decrease the risk of lung cancer: a meta-analysis. PLoS One.
2013;8(8):e71236.
49. Yang Y, Dong J, Sun K, et al. Obesity and incidence of lung cancer:
a meta-analysis. Int J Cancer. 2013;132(5):1162-1169.
50. Smith L, Brinton LA, Spitz MR, et al. Body mass index and risk of
October 2016, Vol. 23, No. 4
lung cancer among never, former, and current smokers. J Natl Cancer Inst.
2012;104(10):778-789.
51. Wu SH, Liu Z. Soy food consumption and lung cancer risk: a
meta-analysis using a common measure across studies. Nutr Cancer.
2013;65(5):625-632.
52. Wu QJ, Xie L, Zheng W, et al. Cruciferous vegetables consumption and the risk of female lung cancer: a prospective study and a metaanalysis. Ann Oncol. 2013;24(7):1918-1924.
53. Song J, Su H, Wang BL, et al. Fish consumption and lung cancer risk:
systematic review and meta-analysis. Nutr Cancer. 2014;66(4):539-549.
54. 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.
55. Slatore CG, Baumann C, Pappas M, et al. 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.
56. US Preventive Services Task Force. Lung cancer: screening,
December 2013. http://www.uspreventiveservicestaskforce.org/Page/
Document/RecommendationStatementFinal/lung-cancer-screening.
Accessed August 29, 2016.
57.Tanner NT, Gebregziabher M, Hughes Halbert C, et al. Racial
differences in outcomes within the National Lung Screening Trial.
Implications for widespread implementation. Am J Respir Crit Care Med.
2015;192(2):200-208.
58. Kumar P, Gareen IF, Lathan C, et al. Racial differences in tobacco
cessation and treatment usage after lung screening: an examination of the
National Lung Screening Trial. Oncologist. 2016;21(1):40-49.
59. Pinsky PF, Zhu CS, Kramer BS. Lung cancer risk by years since
quitting in 30+ pack year smokers. J Med Screen. 2015;22(3):151-157.
60. Clinical Lung Cancer Genome Project (CLCGP); Network Genomic
Medicine (NGM). A genomics-based classification of human lung tumors.
Sci Transl Med. 2013;5(209):209ra153.
61. Pao W, Girard N. New driver mutations in non-small-cell lung cancer.
Lancet Oncol. 2011;12(2):175-180.
62. Cress WD, Chiappori A, Santiago P, et al. Lung cancer mutations and
use of targeted agents in Hispanics. Rev Recent Clin Trials. 2014;9(4):225-232.
63. Politi K, Herbst RS. Lung cancer in the era of precision medicine. Clin
Cancer Res. 2015;21(10):2213-2220.
64.Bollig-Fischer A, Chen W, Gadgeel SM, et al. Racial diversity
of actionable mutations in non-small cell lung cancer. J Thorac Oncol.
2015;10(2):250-255.
65. Zhou W, Christiani DC. East meets West: ethnic differences in epidemiology and clinical behaviors of lung cancer between East Asians and
Caucasians. Chin J Cancer. 2011;30(5):287-292.
66. da Cunha Santos G, Shepherd FA, et al. EGFR mutations and lung
cancer. Annu Rev Pathol. 2011;6:49-69.
67. Pao W, Miller V, Zakowski M, et al. EGF receptor gene mutations
are common in lung cancers from “never smokers” and are associated with
sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci U S A.
2004;101(36):13306-13311.
68. Paez JG, Janne PA, Lee JC, et al. EGFR mutations in lung cancer:
correlation with clinical response to gefitinib therapy. Science.
2004;304(5676):1497-1500.
69. Soh J, Okumura N, Lockwood WW, et al. Oncogene mutations,
copy number gains and mutant allele specific imbalance (MASI) frequently
occur together in tumor cells. PLoS One. 2009;4(10):e7464.
70. Ladanyi M, Pao W. Lung adenocarcinoma: guiding EGFR-targeted
therapy and beyond. Mod Pathol. 2008;21(suppl 2):S16-S22.
71. Shi Y, Li J, Zhang S, et al. Molecular epidemiology of EGFR mutations in Asian patients with advanced non-small-cell lung cancer of adenocarcinoma histology - mainland China subset analysis of the PIONEER
study. PLoS One. 2015;10(11):e0143515.
72. Dogan S, Shen R, Ang DC, et al. Molecular epidemiology of EGFR
and KRAS mutations in 3,026 lung adenocarcinomas: higher susceptibility of women to smoking-related KRAS-mutant cancers. Clin Cancer Res.
2012;18(22):6169-6177.
73. Varghese AM, Sima CS, Chaft JE, et al. Lungs don’t forget: comparison of the KRAS and EGFR mutation profile and survival of collegiate
smokers and never smokers with advanced lung cancers. J Thorac Oncol.
2013;8(1):123-125.
74. Shigematsu H, Lin L, Takahashi T, et al. Clinical and biological features associated with epidermal growth factor receptor gene mutations in
lung cancers. J Natl Cancer Inst. 2005;97(5):339-346.
75. Yang JC, Wu YL, Chan V, et al. Epidermal growth factor receptor
mutation analysis in previously unanalyzed histology samples and cytology
samples from the phase III Iressa Pan-ASia Study (IPASS). Lung Cancer.
2014;83(2):174-181.
76. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361(10):947-957.
77. Zhu CQ, da Cunha Santos G, Ding K, Sakurada A, et al. Role of
KRAS and EGFR as biomarkers of response to erlotinib in National Cancer Institute of Canada Clinical Trials Group Study BR.21. J Clin Oncol.
2008;26(26):4268-4275.
Cancer Control 345
78. Han B, Tjulandin S, Hagiwara K, et al. Determining the prevalence
of EGFR mutations in Asian and Russian patients (pts) with advanced nonsmall-cell lung cancer (aNSCLC) of adenocarcinoma (ADC) and non-ADC
histology: Ignite Study. Ann Oncol. 2015;26(suppl 1):29-44.
79. Yang SH, Mechanic LE, Yang P, et al. Mutations in the tyrosine kinase domain of the epidermal growth factor receptor in non-small cell lung
cancer. Clin Cancer Res. 2005;11(6):2106-2110.
80. Leidner RS, Fu P, Clifford B, et al. Genetic abnormalities of the
EGFR pathway in African American Patients with non-small-cell lung cancer.
J Clin Oncol. 2009;27(33):5620-5626.
81. Riely GJ, Pao W, Pham D, et al. Clinical course of patients with
non-small cell lung cancer and epidermal growth factor receptor exon 19
and exon 21 mutations treated with gefitinib or erlotinib. Clin Cancer Res.
2006;12(3 pt 1):839-844.
82. Cote ML, Haddad R, Edwards DJ, et al. Frequency and type of
epidermal growth factor receptor mutations in African Americans with
non-small cell lung cancer. J Thorac Oncol. 2011;6(3):627-630.
83. Reinersman JM, Johnson ML, Riely GJ, et al. Frequency of EGFR
and KRAS mutations in lung adenocarcinomas in African Americans.
J Thorac Oncol. 2011;6(1):28-31.
84. Arrieta O, Cardona AF, Federico Bramuglia G, et al. Genotyping
non-small cell lung cancer (NSCLC) in Latin America. J Thorac Oncol.
2011;6(11):1955-1959.
85. Corrales-Rodriguez L, Abbas J, Ramos-Esquivel A, et al. Immunohistochemistry as a predictor of EGFR mutations: a retrospective casecontrol study. J Thorac Oncol. 2014;9(9 suppl 3):S176.
86. Rojas Vigott R, Ugalde Gamboa L, Rojas Solano J. Thoracic oncology clinic: our first 200 cases. San José, Costa Rica. J Thorac Oncol.
2014;9(9 suppl 3):S182.
87. Castillo-Fernandez O, Arauz E, Cabreja A, et al. EGFR mutation and
outcomes in patients with metastatic NSCLC treated with erlotinib at Instituto
Oncologico Nacional, Panama. J Thorac Oncol. 2014;9(9 suppl 3):S189.
88. Riely GJ, Marks J, Pao W. KRAS mutations in non-small cell lung
cancer. Proc Am Thorac Soc. 2009;6(2):201-205.
89. Adjei AA. Blocking oncogenic Ras signaling for cancer therapy.
J Natl Cancer Inst. 2001;93(14):1062-1074.
90. Cox AD, Der CJ. Ras history: the saga continues. Small GTPases.
2010;1(1):2-27.
91. Castellano E, Santos E. Functional specificity of ras isoforms: so
similar but so different. Genes Cancer. 2011;2(3):216-231.
92. Pao W, Girard N. New driver mutations in non-small-cell lung cancer.
Lancet Oncol. 2011;12(2):175-180.
93. Ding L, Getz G, Wheeler DA, et al. Somatic mutations affect key
pathways in lung adenocarcinoma. Nature. 2008;455(7216):1069-1075.
94. Califano R, Landi L, Cappuzzo F. Prognostic and predictive value of KRAS mutations in non-small cell lung cancer. Drugs. 2012;72(suppl 1):28-36.
95. Meng D, Yuan M, Li X, et al. Prognostic value of K-RAS mutations
in patients with non-small cell lung cancer: a systematic review with metaanalysis. Lung Cancer. 2013;81(1):1-10.
96. Riely GJ, Kris MG, Rosenbaum D, et al. Frequency and distinctive
spectrum of KRAS mutations in never smokers with lung adenocarcinoma.
Clin Cancer Res. 2008;14(18):5731-5734.
97. Sun Y, Ren Y, Fang Z, et al. Lung adenocarcinoma from East Asian
never-smokers is a disease largely defined by targetable oncogenic mutant
kinases. J Clin Oncol. 2010;28(30):4616-4620.
98. El-Telbany A, Ma PC. Cancer genes in lung cancer: racial disparities:
are there any? Genes Cancer. 2012;3(7-8):467-480.
99. Bauml J, Mick R, Zhang Y, et al. Frequency of EGFR and KRAS
mutations in patients with non small cell lung cancer by racial background:
do disparities exist? Lung Cancer. 2013;81(3):347-353.
100. McQuitty E, Zhang W, Hendrickson H, et al. Lung adenocarcinoma
biomarker incidence in Hispanic versus non-Hispanic white patients. Arch
Pathol Lab Med. 2014;138(3):390-394.
101. Shaw RJ, Kosmatka M, Bardeesy N, et al. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates
apoptosis in response to energy stress. Proc Natl Acad Sci U S A.
2004;101(10):3329-3335.
102. Cao Y, Li H, Liu H, et al. The serine/threonine kinase LKB1 controls
thymocyte survival through regulation of AMPK activation and Bcl-XL
expression. Cell Res. 2010;20(1):99-108.
103. Gao Y, Ge G, Ji H. LKB1 in lung cancerigenesis: a serine/threonine
kinase as tumor suppressor. Protein Cell. 2011;2(2):99-107.
104. Hemminki A, Markie D, Tomlinson I, et al. A serine/threonine kinase
gene defective in Peutz-Jeghers syndrome. Nature. 1998;391(6663):184-187.
105. Lim W, Hearle N, Shah B, et al. Further observations on LKB1/
STK11 status and cancer risk in Peutz-Jeghers syndrome. Br J Cancer.
2003;89(2):308-313.
106. Sanchez-Cespedes M, Parrella P, Esteller M, et al. Inactivation of
LKB1/STK11 is a common event in adenocarcinomas of the lung. Cancer
Res. 2002;62(13):3659-3662.
107. Shah U, Sharpless NE, Hayes DN. LKB1 and lung cancer: more
than the usual suspects. Cancer Res. 2008;68(10):3562-3565.
108. Sanchez-Cespedes M. The role of LKB1 in lung cancer. Fam Cancer.
2011;10(3):447-453.
346 Cancer Control
109. Matsumoto S, Iwakawa R, Takahashi K, et al. Prevalence and
specificity of LKB1 genetic alterations in lung cancers. Oncogene.
2007;26(40):5911-5918.
110. Ji H, Ramsey MR, Hayes DN, et al. LKB1 modulates lung cancer
differentiation and metastasis. Nature. 2007;448(7155):807-810.
111. O’Neill GM, Seo S, Serebriiskii IG, et al. A new central scaffold for metastasis: parsing HEF1/Cas-L/NEDD9. Cancer Res. 2007;67(19):8975-8979.
112. Gao Y, Xiao Q, Ma H, et al. LKB1 inhibits lung cancer progression
through lysyl oxidase and extracellular matrix remodeling. Proc Natl Acad
Sci U S A. 2010;107(44):18892-18897.
113. Wingo SN, Gallardo TD, Akbay EA, et al. Somatic LKB1 mutations
promote cervical cancer progression. PloS One. 2009;4(4):e5137.
114. Sanchez-Cespedes M. A role for LKB1 gene in human cancer beyond the Peutz-Jeghers syndrome. Oncogene. 2007;26(57):7825-7832.
115. Koivunen JP, Kim J, Lee J, et al. Mutations in the LKB1 tumour suppressor are frequently detected in tumours from Caucasian but not Asian
lung cancer patients. Br J Cancer. 2008;99(2):245-252.
116. Onozato R, Kosaka T, Achiwa H, et al. LKB1 gene mutations in
Japanese lung cancer patients. Cancer Sci. 2007;98(11):1747-1751.
117. Gao B, Sun Y, Zhang J, et al. Spectrum of LKB1, EGFR, and KRAS
mutations in Chinese lung adenocarcinomas. J Thorac Oncol. 2010
Aug;5(8):1130-1135.
118. Sun YH, Ren Y, Fang ZY, et al. Lung adenocarcinoma from East
Asian never-smokers is a disease largely defined by targetable oncogenic
mutant kinases. J Clin Oncol. 2010;28(30):4616-4620.
119. Schabath MB, Welsh EA, Fulp WJ, et al. Differential association of
STK11 and TP53 with KRAS mutation-associated gene expression, proliferation and immune surveillance in lung adenocarcinoma. Oncogene.
2016;35(24):3209-3216.
120. Forcet C, Etienne-Manneville S, Gaude H, et al. Functional analysis
of Peutz-Jeghers mutations reveals that the LKB1 C-terminal region exerts
a crucial role in regulating both the AMPK pathway and the cell polarity.
Hum Mol Genet. 2005;14(10):1283-1292.
121. Sánchez-Torres JM, Viteri S, Molina MA, et al. BRAF mutant nonsmall cell lung cancer and treatment with BRAF inhibitors. Transl Lung
Cancer Res. 2013;2(3):244-250.
122. Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in
human cancer. Nature. 2002;417(6892):949-954.
123. Naoki K, Chen TH, Richards WG, et al. Missense mutations of the BRAF
gene in human lung adenocarcinoma. Cancer Res. 2002;62(23):7001-7003.
124. Brose MS, Volpe P, Feldman M, et al. BRAF and RAS mutations in
human lung cancer and melanoma. Cancer Res. 2002;62(23):6997-7000.
125. Marchetti A, Felicioni L, Malatesta S, et al. Clinical features and
outcome of patients with non-small-cell lung cancer harboring BRAF mutations. J Clin Oncol. 2011;29(26):3574-3579.
126. Samuels Y, Wang Z, Bardelli A, et al. High frequency of mutations
of the PIK3CA gene in human cancers. Science. 2004;304(5670):554.
127. Kawano O, Sasaki H, Endo K, et al. PIK3CA mutation status in
Japanese lung cancer patients. Lung Cancer. 2006;54(2):209-215.
128. Soda M, Choi YL, Enomoto M, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature.
2007;448(7153):561-566.
129. Koivunen JP, Mermel C, Zejnullahu K, et al. EML4-ALK fusion gene
and efficacy of an ALK kinase inhibitor in lung cancer. Clin Cancer Res.
2008;14(13):4275-4283.
130. Wong DW, Leung EL, So KK, et al. The EML4-ALK fusion gene is
involved in various histologic types of lung cancers from nonsmokers with
wild-type EGFR and KRAS. Cancer. 2009;115(8):1723-1733.
131. Inamura K, Takeuchi K, Togashi Y, et al. EML4-ALK fusion is linked
to histological characteristics in a subset of lung cancers. J Thorac Oncol.
2008;3(1):13-17.
132. Inamura K, Takeuchi K, Togashi Y, et al. EML4-ALK lung cancers
are characterized by rare other mutations, a TTF-1 cell lineage, an acinar
histology, and young onset. Mod Pathol. 2009;22(4):508-515.
133. Ren S, Kuang P, Zheng L, et al. Analysis of driver mutations in female non-smoker Asian patients with pulmonary adenocarcinoma. Cell
Biochem Biophys. 2012;64(2):155-160.
134. Li C, Fang R, Sun Y, et al. Spectrum of oncogenic driver mutations
in lung adenocarcinomas from East Asian never smokers. PLoS One.
2011;6(11):e28204.
135. Kim HR, Shim HS, Chung JH, et al. Distinct clinical features and outcomes in never-smokers with nonsmall cell lung cancer who harbor EGFR
or KRAS mutations or ALK rearrangement. Cancer. 2012;118(3):729-739.
136. Shinmura K, Kageyama S, Tao H, et al. EML4-ALK fusion transcripts, but no NPM-, TPM3-, CLTC-, ATIC-, or TFG-ALK fusion transcripts,
in non-small cell lung carcinomas. Lung Cancer. 2008;61(2):163-169.
137. Martelli MP, Sozzi G, Hernandez L, et al. EML4-ALK rearrangement
in non-small cell lung cancer and non-tumor lung tissues. Am J Pathol.
2009;174(2):661-670.
October 2016, Vol. 23, No. 4
We must continue to focus attention
and effort to reduce the disproportionate
burden of cancer among blacks living
in the United States.
Cactus. X-Ray image courtesy of Bryan Whitney. www.x-rayphotography.com.
Black Heterogeneity in Cancer Mortality:
US-Blacks, Haitians, and Jamaicans
Paulo S. Pinheiro, MD, PhD, Karen E. Callahan, MPH, Camille Ragin, PhD, MPH,
Robert W. Hage, MD, PhD, DLO, MBA, Tara Hylton, MPH, and Erin N. Kobetz, PhD, MPH
Introduction: The quantitative intraracial burden of cancer incidence, survival and mortality within black
populations in the United States is virtually unknown.
Methods: We computed cancer mortality rates of US- and Caribbean-born residents of Florida, specifically focusing on black populations (United States, Haiti, Jamaica) and compared them using age-adjusted mortality
ratios obtained from Poisson regression models. We compared the mortality of Haitians and Jamaicans residing
in Florida to populations in their countries of origin using Globocan.
Results: We analyzed 185,113 cancer deaths from 2008 to 2012, of which 20,312 occurred in black populations. The overall risk of death from cancer was 2.1 (95% CI: 1.97–2.17) and 1.6 (95% CI: 1.55–1.71) times
higher for US-born blacks than black Caribbean men and women, respectively (P < .001).
Conclusions: Race alone is not a determinant of cancer mortality. Among all analyzed races and ethnicities,
including Whites and Hispanics, US-born blacks had the highest mortality rates while black Caribbeans had the
lowest. The biggest intraracial difference was observed for lung cancer, for which US-blacks had nearly 4 times
greater mortality risk than black Caribbeans. Migration from the islands of Haiti and Jamaica to Florida resulted in
lower cancer mortality for most cancers including cervical, stomach, and prostate, but increased or stable mortality
for 2 obesity-related cancers, colorectal and endometrial cancers. Mortality results in Florida suggest that US-born
blacks have the highest incidence rate of “aggressive” prostate cancer in the world, rather than Caribbean men.
Introduction
Cancer is the second leading cause of death among
blacks in the United States. In relative terms, black
men and women suffer the largest cancer burden,
with higher mortality and lower survival compared to
all major racial-ethnic groups: whites, Asians, Hispan-
From the School of Community Health Sciences University of Nevada Las Vegas, Las Vegas, Nevada (PSP, KEC), the African-Caribbean
Cancer Consortium (PSP, CR, RWH), College of Public Health, Temple University and Fox Chase Cancer Center, Philadelphia, Pennsylvania (CR), Department of Anatomy, St. George's University, St. George’s, Grenada, West Indies (RWH), Florida Department of Health, Tallahassee, Florida (TH), and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida (ENK).
Submitted November 23, 2015; accepted March 4, 2016.
This document is the unedited authors’ version of a submitted work that was accepted for publication.
Address correspondence to Paulo S. Pinheiro, MD, PhD, University of Nevada Las Vegas, School of Community Health Sciences, AfricanCaribbean Cancer Consortium, 4505 South Maryland Parkway, Las Vegas, NV 89154. E-mail: [email protected]
No significant relationships exist between the authors and the companies/organizations whose products or services may be referenced in this article.
Data provided by the Florida Department of Health, Bureau of Vital Statistics. Any conclusions are the authors’ own and do not necessarily
reflect the opinion of the Florida Department of Health.
Dr Pinheiro is sponsored by 8 P20 GM103440-11 from the National Institute of General Medical Sciences as a NV/INBRE recipient.
October 2016, Vol. 23, No. 4
Cancer Control 347
ics, and American Indians.1,2 Black males also have the
greatest cancer incidence.2 These cancer disparities by
race are the result of a complex and entangled web of
differences in risk factors that directly impact not only
cancer incidence, but also access to and availability of
early detection and timely treatment. In turn, these affect the capacity to successfully combat cancer, thus
impacting cancer survival. Cancer mortality is a result
of the interplay between both incidence and survival.
Assessing each of these 3 components ­— incidence,
survival, and mortality — is critical to understanding
how disparities between populations arise and how
they can be reduced or eliminated.
Black men and women in the United States are often categorized under a single ostensibly homogenous
umbrella: blacks, United States blacks, African Americans, or people of African descent. For researchers, the
use of different denominations for describing populations can lead to confusion. “United States blacks”, commonly used for data collection, may or may not characterize blacks born outside of the United States, and
“African descent” may include North African populations. For well-documented historical reasons, the vast
majority of black people living in the Western hemisphere are genetic admixtures of African, European,
and/or Native American populations, warranting caution when attributing risk to race, a social construct.3
Most importantly, however, the use of one aggregate
term obscures considerable heterogeneity in health outcomes among the black population in the United States,
irrespective of whether these differences are attributable to biological, cultural, or socioeconomic factors.4
Blacks born outside the United States number
3.8 million in the United States, nearly 9% of the black
population.5 According to Census projections, this percentage will reach 17% by 2060.6 Substantial black immigration into the United States is a recent dynamic
following the Immigration America Act of 1963.7 The
majority of black immigrants come from English and
French speaking nations of the Caribbean, led by Jamaica and Haiti, the birthplace of 18% and 15% of United States-black immigrants in 2013.6 To the extent that
health is partially determined by social and cultural
factors, including beliefs on disease etiology, types of
family structure and social support, expectations regarding medical care, as well as cumulative lifetime exposure to cancer risk factors, aggregating these distinct
Caribbean populations together with blacks who have
been in the United States for generations may limit the
ability to detect and address culturally driven determinants of health outcomes among all black heterogeneous sub-populations.4,8,9
Two studies have shown lower overall cancer mortality for blacks born outside of the United States than
for US-born.10,11 Yet a more detailed analysis of the intraracial cancer heterogeneity in black populations
348 Cancer Control
living in the United States is non-existent. Problems
studying this topic derive from limitations in the available incidence and survival data. First, cancer registries
do not collect demographic data on black ancestries.
Secondly, the collection of birthplace is substantially
incomplete in the United States cancer surveillance
programs.12,13 These limitations have impeded unbiased cancer incidence and survival studies of these
populations.14-16 Birthplace reporting on death certificates, however, is nearly complete. Thus, mortality
data can be a valuable source of information for assessing the intraracial cancer heterogeneity among blacks
in the United States.
Here we use cancer mortality data for Florida,
a state with a 19% immigrant and 17% black population,17 to analyze cancer outcome heterogeneity. First,
we compare US-born populations of all races with
Hispanic Caribbean populations and majority-black
Caribbean populations born outside the United States.
Then, we compare US-born blacks with majority-black
Caribbean populations. Within the latter, we look specifically at Haitians, Jamaicans, and other West Indian
populations. Through this analysis we aim to characterize race-specific patterns of cancer mortality, explore the intraracial heterogeneity in blacks, and make
a contribution that will assist health policy makers in
shaping targeted cancer prevention and control efforts
to reduce health inequities in cancer outcomes in the
United States.
Materials and Methods
Cancer Data
Cancer mortality data for 5 years, January 1, 2008
through December 31, 2012, were obtained from the
Florida Department of Health Vital Statistics.18 We considered only resident cases in the state. We analyzed
the most common causes of cancer death: lung and
bronchus, breast, prostate, colorectal, pancreas, corpus uteri (endometrial); and cancers of special interest
in populations born outside the United States: cervix
uteri, liver, stomach; and all-sites-combined, which included all cases of malignant cancers. Cancer site was
coded according to the International Statistical Classification of Diseases 10th revision.
We examined the cancer mortality burden at
2 levels of comparison. First, all Florida populations
regardless of race and based on birthplace alone: USborn populations and the following 2 immigrant Caribbean populations: Hispanic Caribbean, including
Cuba, the Dominican Republic and Puerto Rico, and
majority-black Caribbean including Haiti, Jamaica and
Other West Indies (OWI). Haiti and Jamaica are considered separately as they are the 2 largest non-Hispanic
Caribbean immigrant groups in Florida as well as the
United States.7 The OWI category included the following territories and island nations: Anguilla, Antigua
October 2016, Vol. 23, No. 4
and Barbuda, Aruba, Bahamas, Barbados, Cayman
Islands, Dominica, Grenada, Guadeloupe, Martinique,
Montserrat, Saint Kitts and Nevis, Saint Barthelemy,
Saint Lucia, Saint Martin, Saint Vincent and the Grenadines, the Netherlands Antilles, the British Virgin
Islands, Trinidad and Tobago, Turks and Caicos, and
West Indies not otherwise specified. The vast majority
of immigrants from these nations are black, including
99% of all Haitians immigrants and 96% of all Jamaicans.7 Thus, while recognizing that a small number of
immigrants born in these countries may not be black,
we will for the purposes of this paper refer to these
populations as black Caribbean. To strictly adhere to
the Caribbean definition as a geographical region distinct from the continental Americas, Belize and Guyana,
(although also majority black populations), were excluded from the OWI category.
Second, we examined a subset of the Florida population, based on race for US-born populations and
birthplace for those born in the Caribbean. We compare US-born blacks with the black population born
outside the United States. Caribbean populations combined (Haiti, Jamaica, and OWI), as well as the 2 largest populations, Haitians and Jamaicans. For simplicity
purposes in the current study, the Haitian-born, Jamaicanborn, and those born in other West Indies (OWI), and
Hispanic Caribbean-born populations are referred to
as Haitian, Jamaican, OWI and Hispanic Caribbean.
The designation US-blacks will exclusively refer to USborn blacks.
Non-Hispanic black populations from Africa or
elsewhere outside the United States or the Caribbean
were excluded from this study. These excluded populations are relatively young with a more recent immigration history,6,7 and accounted only for 1% —
­­ approximately 40 deaths annually —
­ of all cancer deaths in
Florida among blacks in 2008–2012.
Population Data
The American Community Survey estimates for 2010
indicated that 30% (795,500) of the total 2.6 million
people of non-Hispanic West Indian ancestry in the
United States, lived in Florida.19 Of the 2.4 million
blacks in Florida, 33% were of Caribbean ancestry,
representing the highest proportion of a Caribbean
black-majority population in any American state.19
Since specific ancestry information is unavailable for
cancer mortality data, we are using birthplace. While
ancestry and birthplace are not exactly overlapping,
the overwhelming majority of the Caribbean population in the United States aged 30 and above was born
outside the United States.6,7 These older age groups are
also the ones where cancer will be an important cause
of death; therefore, birthplace and ancestry are not
likely to result in substantially different mortality numbers. Looking at birthplace alone in Florida in 2010,
October 2016, Vol. 23, No. 4
there were 14.6 million US-born (10.2 million whites,
2.4 million blacks, 2 million other races); 1.3 million
Hispanic Caribbean-born; and 570,000 black Caribbean-born (277,000 Haitians, 196,000 Jamaicans and
97,000 OWIs).19
Population denominators for the State of Florida
were obtained from the 5-year 2008–2012 American
Community Survey using the University of Minnesota
interface,20 thus serving the same years as our mortality data. Population numbers were grouped by birthplace and by gender for all groups, and also by race for
the US-born.
Data Analysis
A very small number of cases of non-Hispanic blacks
with unknown birthplace (n = 145, 0.7% of the total)
were proportionally reassigned by imputation models
stratified by age, sex, and cancer site into the United
States, Haiti, Jamaica, OWI, and all other non-US birthplaces as described elsewhere.21
Cancer mortality rates for 2008–2012 were calculated per 100,000 persons, annualized and age-standardized to the 2000 US Standard Population and the 2000
World Standard Population using eighteen age group
bands, all 5-year except the last, which was 85 and older. We used mortality and population data for a period
of 5 years (2008–2012), accumulating cancer mortality
data for a total of 2.85 million person-years for black Caribbean populations. Corresponding 95% confidence
intervals (CIs) were calculated with gamma intervals
modification.22 Site-specific mortality ratios with 95%
CIs were computed by using Poisson regression adjusted for 5-year age groups starting at age 35, except
for prostate cancer where the groups started at age 50.
Because breast cancer risk factors differ between preand post-menopausal women and these rates differ by
race,23 we used approximate pre- and post-menopausal ages, using 50 as our dividing age, to calculate preand post- menopausal ratios. SAS 9.3 (SAS Institute Inc.,
Cary, NC, USA) was used for data analysis.
The OWI cancer burden in Florida was distributed
between more than 20 different countries and territories of origin, the largest of which were Trinidad and
Tobago at 34%, Bahamas at 22% and Barbados at 9%.
This diverse origin as well as relatively small numbers
makes interpretation of the results difficult. Moreover,
unlike Haiti and Jamaica, some of these countries have
a more diverse racial composition, including Trinidad
and Tobago, with a 38% East Indian population.7 Nonetheless, we present OWI data to demonstrate Caribbean completeness.
Finally, we calculated age-adjusted mortality rates,
adjusted to the 2000 World Standard, for Haitian and
Jamaican immigrant populations and well as US-born
blacks living in Florida.
For comparison purposes, we used the best availCancer Control 349
able data from each of these countries, the 2009-2011 avliver cancers. Both Hispanic and black Caribbean poperage cancer mortality rates from the World Health Orgaulations had lower rates for lung, pancreas and breast
nization Mortality Database24 for Jamaica, and the 2012
cancers when compared to the US-born population.
Globocan25 estimates for Haiti.
This study was approved
by the University of Nevada,
Table 1. — Average Annual Age-Adjusted* Mortality Rates for Selected Cancers
Las Vegas Institutional Review
(per 100,000) by Birthplace: Florida, 2008–2012
Board. A data use agreement
US-Born
Born Outside the United States
was obtained from the Florida
United States
Hispanic Caribbean**
Majority Black
Vital Statistics Department.
All Races
All Races
Caribbean
All Races
Results
A total of 841,054 deaths
were recorded among Florida
residents during 2008–2012;
185,113 were deaths attributed to cancers in US-born and
Caribbean-born populations
(Table 1). There were 16,119
cancer deaths in US-blacks,
and 4,113 among black Caribbeans. People born in Jamaica accounted for 43% of these
deaths, those from Haiti, 38%,
and from OWI countries, 19%.
For 2008–2012 in Florida,
cancer was the leading cause
of death for US-born women
as well as for the black Caribbean populations. It accounted for 27.2% of all deaths
among Jamaicans and 23.7%
among Haitians. Cancer ranked
second only to heart disease
as the leading cause of death
among the US-born male
population as well as males
and females born in Hispanic
Caribbean countries.
In comparison to the total
US-born Florida population,
both black Caribbeans and
Hispanic Caribbeans have
lower all-sites-combined
cancer mortality rates. However, there is considerable
heterogeneity in the rates
between them and by cancer
site (Tables 1-2). For black
Caribbeans, there were significantly higher mortality rates
for stomach, prostate and endometrial cancers than the
US-born; among Hispanic
Caribbeans, mortality rates
were higher for stomach and
350 Cancer Control
Deaths
Rate
(95% CI)
Deaths
Rate
(95% CI)
Deaths
Rate
(95% CI)
Stomach
1,386
3.2
(3.0–3.4)
235
4.7
(4.2–5.5)
97
6.7|
(5.4–8.4)
Colorectal
7,455
16.9
(16.6–17.3)
917
18.2
(17.1–19.5)
206
13.8
(11.9–16.1)
Liver and
Intrahepatic
Bile Duct
3,766
8.4
(8.1–8.7)
476
9.3
(8.5–10.3)
109
6.9
(5.6–8.5)
Pancreas
5,392
12.1
(11.7–12.4)
540
10.6
(9.7–11.6)
128
9.1
(7.5–11.1)
Lung and
Bronchus
28,023
62.3
(61.6–63.1)
2,287
44.7
(42.8–46.6)
317
21.0
(18.6–23.6)
Prostate
7,936
17.9
(17.5–18.3)
943
19.1
(17.9–20.4)
437
35.5
(32.1–39.3)
All combined
89,387
201.7
(200.4–203.1)
8,487
169.4
(165.7–173.2)
2,020
141.4
(134.9–148.2)
Stomach
911
1.7
(1.6–1.8)
173
2.6
(2.2–3.2)
92
4.6
(3.7–5.8)
Colorectal
6,335
11.7
(11.4–12.0)
855
12.4
(11.6–13.4)
243
11.9
(10.4–13.6)
Liver and
Intrahepatic
Bile Duct
1,518
2.8
(2.7–3.0)
260
3.8
(3.3–4.4)
76
3.9
(3.0–5.0)
Pancreas
4,874
8.9
(8.7–9.2)
537
7.6
(7.0–8.4)
129
6.4
(5.4–7.8)
Lung and
Bronchus
22,217
41.9
(41.4–42.5)
1090
16.1
(15.2–17.2)
194
9.8
(8.4–11.4)
Breast
10,819
21.6
(21.2–22.1)
1,100
17.5
(16.4–18.6)
413
19.1
(17.3–21.2)
Premenopausal,
less than 50
1,141
3.9
(3.7–4.1)
100
2.6
(2.1–3.2)
97
4.8
(3.9–6.0)
Postmenopausal,
50 or more
9,678
17.8
(17.5–18.2)
1,000
14.9
(14.0–15.9)
316
14.3
(12.7–16.1)
Cervix Uteri
1,140
2.8
(2.6–3.0)
138
2.6
(2.2–3.2)
68
3.3
(2.5–4.3)
Corpus and
Uterus
(endometrial)
1,975
3.8
(3.6–4.0)
278
4.2
(3.7–4.8)
152
7.3
(6.2–8.7)
All combined
74,241
141.4
(140.4–142.5)
6,890
104.6
(102.1–107.3)
2,093
103.6
(98.7–108.8)
Male
Female
*2000 US Standard Population **Cuba, Dominican Republic and Puerto Rico
October 2016, Vol. 23, No. 4
Table 2. — Average Annual Age-Adjusted* Mortality Rates for Selected Cancers per 100,000 for
US-Born and Blacks Born Outside the United States: Florida, 2008–2012
US-Born
Blacks
Born Outside the United States
Black Caribbean**
All Races
Haitian
All Races
Jamaican
All Races
Other West Indian
(OWI) All Races
Deaths
Rate
(95% CI)
Deaths
Rate
(95% CI)
Deaths
Rate
(95% CI)
Deaths
Rate
(95% CI)
Deaths
Rate
(95% CI)
Stomach
292
9.4
(8.3–10.6)
97
6.7
(5.4–8.4)
45
7.4
(5.2–10.3)
39
6.8
(4.8–11.9)
<15
5.0
(2.4–14.5)
Colorectal
895
28.3
(26.3–30.4)
206
13.8
(11.9–16.1)
83
12.4
(9.7–15.8)
86
14.7
(11.6–20.3)
37
15.4
(10.4–25.7)
Liver and
Intrahepatic
Bile Duct
436
11.0
(9.9–12.2)
109
6.9
(5.6–8.5)
69
9.4
(7.2–12.2)
28
4.6
(3.0–9.6)
<15
4.5
(2.0–14.0)
Pancreas
474
14.7
(13.3–16.2)
128
9.1
(7.5–11.1)
34
5.0
(3.4–7.4)
63
11.5
(8.8–17.0)
31
11.6
(7.5–21.4)
Lung and
Bronchus
2,536
78.9
(75.6–82.3)
317
21.0
(18.6–23.6)
105
17.5
(14.1–21.6)
153
25.1
(21.1–31.2)
59
21.2
(15.6–31.7)
Prostate
1,218
50.6
(47.6–53.7)
437
35.5
(32.1–39.3)
142
29.2
(24.3–34.7)
194
37.8
(32.4–45.2)
101
44.0
(35.0–57.3)
All combined
8,575
273.9
(267.6–280.3)
2,020
141.4
(134.9–148.2)
753
124.5
(115.1–134.6)
883
154.4
(143.9–166.4)
384
148.3
(132.2–167.6)
Stomach
179
4.0
(3.4–4.6)
92
4.6
(3.7–5.8)
42
4.9
(3.5–6.9)
31
3.7
(2.5–6.8)
19
6.4
(3.8–11.8)
Colorectal
809
17.7
(16.5–19.0)
243
11.9
(10.4–13.6)
75
8.6
(6.7–11.1)
122
14.2
(11.8–18.1)
46
13.4
(9.7–19.5)
Liver and
Intrahepatic
Bile Duct
169
3.6
(3.0–4.1)
76
3.9
(3.0–5.0)
37
4.6
(3.2–6.6)
28
3.3
(2.2–6.5)
<15
3.4
(1.6–8.2)
Pancreas
532
11.8
(10.8–12.8)
129
6.4
(5.4–7.8)
53
6.7
(5.0–9.0)
47
5.3
(3.9–8.6)
29
9.3
(6.1–15.0)
Lung and
Bronchus
1,473
32/3
(30.6–34.0)
194
9.8
(8.4–11.4)
66
8.6
(6.6–11.2)
95
11.1
(8.9–14.7)
33
10.3
(7.0–16.1)
Breast
1,514
32.1
(30.4–33.7)
413
19.1
(17.3–21.2)
158
17.0
(14.4–20.2)
187
21.1
(18.2–25.4)
68
19.0
(14.6–25.6)
Premenopausal,
less than 50
298
6.2
(5.5–6.9)
97
4.8
(3.9–6.0)
50
5.2
(3.9–7.1)
38
5.2
(3.7–8.6)
<15
2.6
(1.2–7.3)
Postmenopausal,
50 or more
1,216
25.9
(24.4–27.4)
316
14.3
(12.7–16.1)
108
11.8
(9.6–14.6)
149
15.9
(13.5–19.2)
<68
16.4
(12.4–22.7)
Cervix Uteri
267
5.5
(4.8–6.2)
68
3.3
(2.5–4.3)
36
3.9
(2.7–5.7)
20
2.6
(1.6–5.8)
<15
3.6
(1.8–8.5)
Corpus and
Uterus
(endometrial)
373
8.1
(7.3–9.0)
152
7.3
(6.2–8.7)
49
5.7
(4.2–7.8)
75
8.6
(6.7–12.1)
28
7.6
(5.0–12.8)
All combined
7,544
163.2
(159.5–167.0)
2,093
103.6
(98.7–108.8)
798
95.6
(88.5–103.3)
902
105.4
(98.4–113.4)
393
121.3
(106.8–137.8)
Male
Female
*2000 US Standard Population **Haiti, Jamaica and OWI combined
October 2016, Vol. 23, No. 4
Cancer Control 351
When narrowing the comparisons to Haitian,
Jamaican, and US-black populations in Florida, the
cancer mortality rate differences are surprisingly even
greater (Table 2). Haitians have the lowest mortality rates for all cancers combined, 125 per 100,000
among males and 96 per 100,000 in females, followed
closely by Jamaicans with 154 and 105 per 100,000
among males and females, respectively. Contrasting
with these relatively low rates, US-blacks had high
mortality rates, 274 per 100,000 for males and 163
per 100,000 for females. The risk of cancer death in
US-black males is 2.1 times higher than among black
Caribbean males and 1.6 times higher for black Caribbean females (P < .05). Of all cancers examined, lung
cancer has the greatest risk differential. US-blacks
compared to black Caribbeans have an astonishing
4 times greater risk of lung cancer death among males
and a 3.5 times greater risk among females (Table 3).
The leading cause of cancer mortality among USblack males was lung cancer, at 78.9 per 100,000, followed distantly by prostate cancer at 50.6 per 100.000.
For black Caribbean males the opposite was true: pros-
Table 3. — Mortality Rate Ratios for Selected Cancers:
Florida, 2008–2012
US-Born Blacks in Relation
to Black Caribbean
(Reference Population)
Rate Ratio
95% CI
1.4
(1.13–1.80)
Male
Stomach
Colorectal
2.1
(1.81–2.46)
Liver and Intrahepatic Bile Duct
1.9
(1.55–2.39)
tate cancer mortality was highest, at 35.5 per 100,000,
followed by lung cancer at 21.0 per 100,000 (Table 2).
Across the male populations of US-blacks, Jamaicans,
Haitians, and OWIs, colorectal was the third leading
cause of cancer death, while pancreatic cancer was
fourth, except among Haitians for whom liver cancer
was fourth.
Among US-black women in Florida, the 2 most
common causes of cancer mortality were lung and
breast, almost equal at 32.3 and 32.1 per 100,000 respectively, followed by colorectal and pancreatic cancers. For black Caribbean women, breast cancer was
by far the leading cause of cancer mortality at 19.1 per
100,000, with colorectal cancer second and lung cancer only third (Table 2). Still, US-black women had a
1.7 times greater overall mortality risk for breast cancer than black Caribbean women, although pre-menopausal breast cancer risk was only 1.3 times higher
(Table 3). Endometrial cancer was the fourth leading
cause of cancer death for black Caribbean women. Notably, cervical cancer rates were relatively low across
black Caribbean female populations.
Lastly, the cancer mortality rates of Haitian and
Jamaican populations residing in Florida when compared to the rates in their respective countries of origin
are lower for both males and females for all cancer sites
combined (Table 4). This advantage is most marked for
cervix, stomach, and prostate cancers for both countries, liver cancer for Haitians and female breast for
Jamaicans. On the other hand, for colorectal (male and
female), endometrial, and pancreatic cancers, mortality rates are higher for Haitians and relatively similar
for Jamaicans in Florida in comparison to rates found
in the countries of origin. Also higher in Florida than
in countries of origin are rates for lung cancer among
Haitian males and Jamaican females as well as breast
cancer among Haitian females (Table 4).
Pancreas
1.8
(1.45–2.15)
Lung and Bronchus
4.0
(3.51–4.45)
Prostate
1.4
(1.29–1.61)
Discussion
All combined
2.1
(1.97–2.17)
Stomach
0.9
(0.66–1.09)
Colorectal
1.5
(1.30–1.73)
This is the first study to address intraracial differences in
cancer mortality among persons often categorized under the US-blacks designation. Using birthplace, we differentiate and directly compare black populations born
in and outside the United States, presenting the first
quantification of the cancer burden for black Caribbeans. In Florida, cancer mortality rates for US-blacks are
higher than all analyzed groups. However, black Caribbean populations have the lowest rates, suggesting that
racial health disparities in the United States are attributable to more than socioeconomic status (SES) and health
care access, for which Caribbeans are likely to be at least
equally as disadvantaged as US-blacks.
The major contributor for the relatively low overall cancer mortality in blacks born outside the United
States is the difference in lung cancer rates. This is
most likely a reflection of differences in smoking hab-
Female
Liver and Intrahepatic Bile Duct
1.0
(0.77–1.34)
Pancreas
1.9
(1.55–2.27)
Lung and Bronchus
3.5
(2.99–4.03)
Breast
1.7
(1.48–1.84)
Pre-menopausal, less than 50
1.3
(0.99–1.60)
Post-menopausal, 50 or more
1.8
(1.55–1.99)
Cervix Uteri
1.7
(1.30–2.24)
Corpus and Uterus
(endometrial)
1.1
(0.93–1.35)
All combined
1.6
(1.55–1.71)
*Poisson regression rate ratios adjusted for age group
352 Cancer Control
October 2016, Vol. 23, No. 4
represent a healthier sample
of the population than those
who remain in the country
Haitians
US-Blacks
Jamaicans
US-Blacks
of origin.32 Having said that,
immigrants
also likely carry
In country In Florida In Florida In Country In Florida In Florida
of origin,
of Origin,
with them baseline protective
Haiti
Jamaica
factors inherent to their counMale
tries of origin that are persisStomach
8.7
5.1
6.3
9.6
4.9
6.3
tent in reducing risk, and resistant to the negative effects
Colorectal
4.6
9.2
19.3
9.8
10.7
19.3
of acculturation, as has been
Liver and
7.5
7.4
8.8
3.1
4.0
8.8
shown for Hispanics, particuIntrahepatic
Bile Duct
larly for Mexican Hispanics.21
Pancreas
2.6
3.6
10.3
3.1
7.2
10.3
Caribbean immigrants in the
United States tend to maintain
Lung and
6.6
11.9
55.1
24.1
18.7
55.1
Bronchus
traditional dietary patterns
Prostate
32.3
17.3
28.8
40.1
22.2
28.8
that include less red meat consumption and higher intake of
All-site
88.4
85.6
186.3
131.8
107.4
186.3
combined
whole grains, fruits and vegFemale
etables, which are protective
against colorectal and cancers
Stomach
6.0
3.6
2.6
4.1
2.7
2.6
of the upper gastrointestinal
Colorectal
5.8
6.4
12.3
9.7
9.5
12.3
tract.33-36 Also, fertility rates
Liver and
5.1
3.2
2.6
2.4
2.2
2.6
and breastfeeding patterns
Intrahepatic
Bile Duct
may help explain breast cancer differences.23 Several studPancreas
2.3
4.6
8.2
3.1
3.5
8.2
ies
demonstrated significantly
Lung and
6.2
5.6
23.1
6.0
7.5
23.1
Bronchus
higher odds of breastfeeding
initiation among immigrant
Breast
11.5
13.4
24.0
21.5
16.0
24.0
black women than US-born
Cervix uteri
14.6
3.1
4.4
12.8
2.0
4.4
blacks.37-39 Overall, the lower
Corpus and
1.4
4.2
6.0
6.6
5.9
6.0
rates we found for breast and
Uterus
(endometrial)
colorectal cancers among
black Caribbeans are all the
All-site
80.3
69.2
117.7
99.7
75.0
117.7
combined
more surprising in light of
previous studies document*Average annual age-adjusted to 2000 World Standard.
Country of origin data: Haiti, GLOBOCAN 2012; Jamaica, WHO Mortality Database 200–2011 average.
ing lower rates of mammography and colonoscopy among
blacks born outside the United
its 2 to 4 decades ago. In 1980, the prevalence of smokStates, especially Haitians, in comparison to US-born
ing in Haiti (18% males, 6% females) and Jamaica
blacks.40-43 Additionally, if they are of low SES, non–
(27% males, 7% females) was much lower than in USUS-citizen immigrants are less likely than citizens to
blacks (45% males, 31% female).26 Moreover, studies
have had consistent health insurance.44 In the case of
have documented that blacks born outside the United
the undocumented or recently documented, access is
States are less likely to smoke than their US-born counblocked to any federal assistance programs in Florida
terparts.27,28 The risk factor of smoking may also acexcept limited emergency Medicaid.45
count for some of the differences seen in stomach and
While most intraracial cancer mortality differences
pancreatic cancers, as well as other tobacco-related
in our study seem to be determined by environmental
cancer sites like bladder, larynx, and oral cavity which
factors, some of which are inherently related to place
are included in the all sites combined category.29
of birth, it is important to note that some patterns are
Our lower cancer mortality findings among Caribseemingly more related to race and possibly genetics,
bean blacks may be partially explained by the Healthy
regardless of geography. Endometrial, premenopausImmigrant Effect (HIE), whereby immigrants have betal breast (defined here as below age 50) and prostate
ter overall health outcomes not solely related to observcancer rates reveal specific vulnerabilities. Compared
able socioeconomic factors such as education and into other groups, all black populations (US-blacks,
come.10,30,31 In other words, those who immigrate may
Haitians, and Jamaicans) have higher mortality for
Table 4. — Mortality Rates* (per 100,000) for Selected Cancers for Haitians
and Jamaicans in Country of Origin and in Florida and for US-Blacks: Florida, 2008–2012
October 2016, Vol. 23, No. 4
Cancer Control 353
these 3 cancers. Endometrial cancer is a cancer of the
Western world,46 highly related to obesity,36 whose
prevalence commonly increases among first generation immigrants to the United States.21 Studies have
shown that endometrial cancer survival and mortality
outcomes for black women, even after adjustment for
their higher proportion of hysterectomies, are worse
than all other racial groups.47,48 Black women also have
an elevated risk of more aggressive tumor subtypes
with poorer prognosis.48,49
For premenopausal breast cancer, the finding of
high mortality among all analyzed black populations
relative to the all US races category as well as Hispanic
Caribbeans is intriguing and requires further study.
Given the lack of detailed incidence data for each of
these populations, it is impossible to assess whether
this finding can be attributed to the well-documented
survival disparity between blacks and Whites in the
United States,50 or if it reflects a true increased risk for
premenopausal breast cancer among black women.
For prostate cancer, black African descent has
been well established as a risk factor.1,51,52 Our results
confirm high rates among both US-born and Caribbean-born blacks. However, the significantly higher
rates we find for US-blacks compared to Caribbeanborn blacks is notable, given the lower prostate cancer screening rates for the latter compared to the former.40,53,54 While prostate cancer incidence rates are
challenging to compare globally due to increased use
of prostate-specific antigen (PSA) screening in some
countries,55,56 mortality rates in Florida likely reflect
the incidence of aggressive prostate cancer. Thus, our
study is highly indicative of US-blacks having the highest aggressive prostate cancer incidence in the world,
not Caribbeans as previously suggested.57
Three infection-related cancers ­­— cervical, liver
and stomach cancer — are more commonly found in
developing countries.46 Strangely, the mortality rates
are similar or even higher for US-blacks compared to
the black populations born outside the United States.
For cervical cancer, despite the fact that the pap test
screening rates are higher among US-black women
than Caribbean-born black women23,53,58 which would
predict earlier stages at diagnosis with better prognosis, the mortality rates for cervical cancer are higher
in US-blacks than Caribbean-born black women. For
liver cancer, it is possible that the epidemiology and
patterns of transmission are different between these
populations, resulting in different gender-specific incidence and thus mortality. Hepatitis C, a more common
liver cancer risk factor than hepatitis B in the United
States, is largely transmitted by sharing contaminated
intravenous drug needles.59 The greater differential in
liver cancer between males and females in US-blacks
than in Caribbean blacks may be the result of greater
IV drug use among US-black men than US-black wom354 Cancer Control
en.60 For Caribbeans, Hepatitis B likely accounts for the
high rates, especially among Haitians,61 and would not
result in a marked gender difference. As a result, liver
cancer mortality rates are higher for US black males
but similar between US-black females and black Caribbean females in our study.
Haitians and Jamaicans were clearly distinct in
their cancer mortality patterns, although the precision
of the statistical comparisons may be impacted by the
relatively small size of the populations in Florida.
Haitians had lower all-sites-combined mortality rates
than Jamaicans as well as all other analyzed populations. However, for all 3 infection-related cancers, liver,
cervical and stomach, Haitian mortality rates exceeded
those of Jamaicans. Haitian rates were lower than Jamaicans’ for prostate, breast, endometrial and colorectal
cancers: all cancers commonly associated with a “Western” lifestyle.62 Jamaicans have a longer immigration
history in Florida than Haitians;20 therefore it is possible that these differences reflect acculturation patterns,
predicted by length of time in the United States, and the
extent of assimilation into US culture, which may be
greater for English-speaking Jamaicans. Additionally,
since the country of Jamaica has been more developed
than Haiti for at least the past 25 years,63 the prevalence
of baseline immigrant risk factors such as obesity may
already be more “Westernized”.64
Additional important findings arise from the examination of differences in mortality rates between
Haitians and Jamaicans in their countries of origin
compared to those same populations in Florida. Since
mortality is a function of cancer incidence and survival, changes in mortality rates may reflect a difference in
either or both of these. If the healthcare infrastructure
in Florida is better than in Jamaica and Haiti, we would
expect cancer survival to be higher, and thus mortality rates to be lower among Jamaicans and Haitians in
Florida than in their countries of origin. Despite the
fact that survival data accuracy for blacks born outside
the United States can be problematic,16 some studies
have documented this pattern.65-68 On the other hand,
if Caribbeans living in Florida have higher or even stable cancer mortality rates compared to their counterparts in their countries of origin, then these changes
likely reflect true increases in incidence rather than decreases in survival.
In our study, mortality rates for Haitians and Jamaicans in Florida compared to their countries of origin were predictably lower for all 3 infection-related
cancers, except for liver cancer in male Jamaicans.
Stomach cancer gains are likely attributable to a higher
standard of living in the United States coinciding with
a lower prevalence of infection with Helicobacter
pylori.69,70 Likewise, lower liver cancer mortality rates
in Florida populations compared to their countries of
origin suggest lower incidence, at least in part due to
October 2016, Vol. 23, No. 4
a lower prevalence among US blacks (0.9%) of chronic
hepatitis B compared to prevalence in Jamaica (3.7%)
or Haiti (13.6%).61,71 Gains for cervical cancer were substantial in both Haitian and Jamaican populations in
Florida, with mortality rates reduced to less than 25%
of the rates in their countries of origin. This likely reflects improved access to health care, including screening and radiotherapy treatment, which would impact
incidence and survival, respectively. The same likely
applies to the substantial improvement for female
breast cancer among Jamaicans.
We also found an advantage for prostate cancer for
immigrants living in Florida. Mortality rates among
Haitians and Jamaicans are substantially lower in Florida than in the country of origin. This is not remarkable
given the complexity of prostate cancer control: including screening, computed tomography and bone scans
for staging, surgery, hormone therapy and radiotherapy.
The mortality rates for colorectal cancer in both
sexes and endometrial cancer in women suggest an increased risk in Florida compared to the country of origin. For Haitians, the rates are significantly higher than
in their countries of origin, while for Jamaicans there
is minimal change. Yet, in the United States, access to
colorectal screening if utilized would result in either
averted cancer by removal of precancerous lesions or
earlier cancer diagnosis.72 Moreover, since there is no
knowledge of latent forms of either colorectal or endometrial cancers, which eventually progress to death
if left untreated, access to better health care facilities
should result in improved treatment, prolonged survival, and lower mortality in Florida. Therefore, the lack
of decreases seen in mortality for these cancers must be
rooted in an increase in incidence, which is a worrisome
finding. Unfortunately, a common risk factor for both
cancers is obesity.1 Data at the population level shows a
trend of increasing prevalence of obesity and transition
to a high caloric fatty diet when immigrants settle in the
United States.64,73,74
Some of the results did not follow known or expected patterns. Breast cancer has protective risk factors associated with fertility patterns23 that have been
shown to change with migration to the United States,
including delayed childbearing and a reduction in total number of pregnancies.75,76 In our study, for Haitian
females, breast cancer mortality rates were somewhat
higher in Florida than Haiti. Offsetting a possible gain
in survival due to screening and more access to quality
treatment, the increased mortality among Haitians in
Florida may well derive from an increased incidence of
breast cancer which may be higher due to diminished
fertility and delayed childbearing among other factors.
On the other hand, for Jamaican women, breast cancer
mortality rates were much lower in Florida than in
Jamaica. It is possible that Jamaican women in Florida,
who tend to have higher levels of education,7 may reap
October 2016, Vol. 23, No. 4
the benefit of better health care access. The greater
total baseline fertility rate in Haiti than Jamaica
(3.1 compared to 2.3 children77) as well as higher obesity rates in Jamaica64 may also play a role in explaining
why breast cancer rates in Florida are higher among
Jamaican than Haitian women.
For lung cancer, for Haitian males and Jamaican females, rates were significantly higher in Florida than
Haiti. Lung cancer has a poor survival prognosis regardless of gender, SES, or race, and even location.78
Therefore, this difference would suggest an increase in
incidence rather than a change in survival. Yet among
Jamaican men, lung cancer rates were much lower in
Florida than in Jamaica, suggesting an intriguing decreased incidence. It is possible that these results reflect changes in smoking prevalence when immigrants
settle in the United States.79 Given the importance of
smoking as a risk factor for many cancers, these changes need to be carefully monitored.
Our study has the usual limitations of descriptive
epidemiology. There is a lack of both individual level
risk factor data for all subjects as well as group level
risk factor data specific to Caribbean-born populations
in Florida. We also had some relatively small population sizes, which sometimes resulted in a lack of precision in our mortality rates, particularly for the small
OWI group. We attempted to mitigate this by accumulating 5 years of mortality experience and annualizing
the rates. Another limitation is that Globocan mortality estimates for 2012 for Haitians are modelled, relying on the last date for which real data were available,
which may be several years old.
Our study examines black populations in Florida,
which may not be representative of all black populations in the United States. Additionally, in theory,
our mortality numbers could reflect a “salmon bias”,
whereby small numbers of immigrants who are very ill
return to their home countries of origin to die, which
would artificially lower mortality rates for those born
outside the United States in our study. However, the
magnitude of this out-migration for Hispanics has
been found to be very small,80 and the literature for
this phenomenon among black immigrants in the
United States is non-existent.
Relying on birthplace rather than ancestry may
result in a small degree of misclassification. Haitian,
Jamaican and other West Indian ancestry populations,
which may include second generation Caribbeans,
are larger than the birthplace populations we used for
this study. Some of the people in the US-born blacks
category in our study are quite possibly of Haitian,
Jamaican or OWI ancestry. However, given the trend
of relatively recent immigration for these populations,
this would likely only affect cancer sites with mortality
in relatively younger age groups, such as cervical cancer. Our Florida mortality data provided information
Cancer Control 355
on Haitian ancestry, but not any other Caribbean ancestry. To test for differences between using birthplace
and ancestry, we calculated mortality rates using the
Haitian ancestry information and found that the differences in rates were negligible, thus reducing the likelihood of a threat to validity based on misclassification.
Strengths of our study include the very high completeness (99%) of birthplace information among all
deaths in the state of Florida, allowing for reliable classification of the different populations, which is unfortunately never achieved in cancer incidence data. We
also used detailed denominators based on birthplace
available from the American Community Survey.
Overall, cancer mortality rates among black
Caribbeans are remarkably low. While there has been
much research demonstrating health advantages for
Hispanics, our results point to an even greater health
advantage for black Caribbeans than Hispanic Caribbeans in Florida. More striking is the markedly worse
cancer mortality rates for US-blacks when compared
to Caribbean blacks. The low mortality among Caribbean blacks has the effect of improving the overall US
cancer rates for blacks. Not surprisingly, of the 17 states
with black populations over 1 million, Florida and New
York, states with the highest proportions of black
Caribbean populations, have the lowest cancer mortality rates for blacks in the United States.1
To our knowledge these are the only results available looking at cancer outcomes on a population basis
that analyze US-born blacks separately, directly comparing them to Caribbean-born people residing in the
United States. The mortality rates presented here will
also constitute a baseline for monitoring the cancer
burden among Caribbean populations and help the
fight against cancer across all majority-black Caribbean
nations. Cancer literature is somewhat scarce for the
Caribbean region due to a lack of reliable data. Phillips
et al. used estimates from Globocan 2002 data to characterize cancer incidence and mortality in the Caribbean nations.81 However, in a quickly evolving world,
these data are possibly now outdated. Fortunately, cancer registration in Jamaica and other Caribbean nations
is showing signs of improvement.46
The current study highlights once again the need
to thoroughly address known racial disparities in cancer risk and outcomes in the United States. To analyze
intraracial differences, it is fundamental to have specific and accurate cancer data, including data on immigrant groups. In states with large Caribbean populations, such as Florida and New York, it would be
beneficial to have health data on this population collected methodically as has been done for other racial/
ethnic subgroups in California and New York City. An
active role of the North American Central Cancer Registries Association and the central cancer registries
in Florida and New York would greatly improve the
356 Cancer Control
knowledge of cancer epidemiology among black populations, especially regarding incidence data. Better
understanding of intraracial cancer patterns including
the extent to which acculturation impacts cancer incidence, survival, and mortality among black US immigrants will help in targeted US cancer prevention and
control efforts.
Conclusions
The US-born black population has the highest cancer
mortality risk in the nation — even higher than previously documented when rates for blacks born outside
the United States are calculated separately. Continued
attention and effort is required to reduce the disproportional burden of cancer in US blacks. On the other
hand, a record 3.8 million black immigrants, accounting for 9% of the current black population in the United States,5 are currently low risk populations for most
cancers. It is important to monitor these trends, looking for ways to help all populations acquire protective
factors and resist risk factors for cancer. More studies
into the mechanisms that would avoid an unfavorable
trend towards worse cancer outcomes in Caribbean
blacks are necessary. Our study suggests that obesity,
a known risk factor for many US blacks, may be an
important focus for intervention in Caribbean blacks,
especially among females, as immigrants have higher
colorectal and endometrial cancer mortality rates in
relation to their countries of origin. Targeted screening programs and expanding access to care would also
help these immigrant populations. Concurrently, the
public health community must clarify and address the
reasons for such poor cancer outcomes among USborn blacks so that the interracial gaps can be significantly narrowed.
References
1. American Cancer Society. Cancer Facts & Figures for African
Americans 2013-2014. Atlanta, GA: American Cancer Society; 2013.
2. Kohler BA, Sherman RL, Howlader N, et al. Annual report to the
nation on the status of cancer, 1975-2011, featuring incidence of breast
cancer subtypes by race/ethnicity, poverty, and state. J Natl Cancer Inst.
2015;107(6):djv048.
3. Benn-Torres J, Bonilla C, Robbins CM, et al. Admixture and population stratification in African Caribbean populations. Ann Hum Genet.
2008;72(pt 1):90-98.
4. Arthur CM, Katkin ES. Making a case for the examination of ethnicity
of Blacks in United States Health Research. J Health Care Poor Underserved.
2006;17(1):25-36.
5. Colby SL, Ortman JM. Projections of the Size and Composition of the
U.S. Population: 2014 to 2060, Current Population Reports. Washington, DC:
U.S. Census Bureau; 2014. https://www.census.gov/content/dam/Census/
library/publications/2015/demo/p25-1143.pdf. Accessed July 15, 2016.
6. Anderson M. A Rising Share of the U.S. Black Population is Foreign
Born: 9 Percent are Immigrants; and While Most are From the Caribbean,
Africans Drive Recent Growth. Washington, DC: Pew Research Center;
2015. http://www.pewsocialtrends.org/2015/04/09/a-rising-share-of-the-u-sblack-population-is-foreign-born/. Access July 18, 2016.
7. Thomas KAJ. A Demographic Profile of Black Caribbean Immigrants in the United States. Washington, DC: Migration Policy Institute;
2012. http://www.migrationpolicy.org/research/CBI-demographic-profileblack-caribbean-immigrants. Accessed July 18, 2016.
8. Singh GK, Siahpush M. Ethnic-immigrant differentials in health behaviors, morbidity, and cause-specific mortality in the United States: an
analysis of two national data bases. Hum Biol. 2002;74(1):83-109.
October 2016, Vol. 23, No. 4
9. Consedine NS, Magai C, Spiller R, et al. Breast cancer knowledge
and beliefs in subpopulations of African American and Caribbean women.
Am J Health Behav. 2004;28(3):260-271.
10.Singh GK, Siahpush M. All-cause and cause-specific mortality
of immigrants and native born in the United States. Am J Public Health.
2001;91(3):392-399.
11. Fang J, Madhavan S, Alderman MH. Influence of nativity on cancer
mortality among black New Yorkers. Cancer. 1997;80(1):129-135.
12. Pinheiro PS, Bungum TJ. Country of origin and breast cancer survival. Asia‐Pac J Clin Oncol. 2014;10(3):279.
13. Pinheiro PS, Bungum TJ, Jin H. Limitations in the imputation strategy to handle missing nativity data in the Surveillance, Epidemiology, and
End Results program. Cancer. 2014;120(20):3261-3262.
14. Pinheiro PS, Williams M, Miller EA, et al. Cancer survival among Latinos and the Hispanic Paradox. Cancer Causes Control. 2011;22(4):553-561.
15.Gomez SL, Glaser SL. Quality of cancer registry birthplace
data for Hispanics living in the United States. Cancer Causes Control.
2005;16(6):713-723.
16. Pinheiro PS, Morris CR, Liu L, et al. The impact of follow-up type
and missed deaths on population-based cancer survival studies for
Hispanics and Asians. J Natl Cancer Inst Monogr. 2014;2014(49):210-217.
17. U.S. Census Bureau. QuickFacts: Florida. http://www.census.gov/
quickfacts/table/PST045215/12,00. Accessed July 18, 2016.
18.Bureau of Vital Statistics, Florida Department of Health,
Jacksonville, Florida. Mortality data 2008-2012, received June 2015.
19. U.S. Census Bureau. American Fact Finder. Selected Population
Profile in the United States: 2009-2011 American Community Survey
3-Year Estimates, Table S0201. http://factfinder.census.gov/faces/tableservices/jsf/pages/productview.xhtml?src= bkmk. Accessed July 18, 2016.
20. Ruggles S, Genadek K, Goeken R, Grover J, and Sobek
M. Integrated Public Use Microdata Series: Version 6.0 [Machine-readable database]. American Community Survey 2012 5-Year Estimates.
Minneapolis: University of Minnesota. Accessed June 2015.
21. Pinheiro PS, Sherman RL, Trapido EJ, et al. Cancer incidence in
first generation U.S. Hispanics: Cubans, Mexicans, Puerto Ricans, and
new Latinos. Cancer Epidemiol Biomarkers Prev. 2009;18(8):2162-2169.
22. Tiwari RC, Clegg LX, Zou Z. Efficient interval estimation for ageadjusted cancer rates. Stat Methods Med Res. 2006;15(6):547-569.
23.
American Cancer Society. Breast Cancer Facts & Figures, 20152016. Atlanta, GA: American Cancer Society; 2015.
24. World Health Organization. WHO Mortality Database. http://apps.
who.int/healthinfo/statistics/mortality/whodpms/. Accessed July 19, 2016.
25. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and
mortality worldwide: sources, methods and major patterns in GLOBOCAN
2012. Int J Cancer. 2015;136(5):E359-E386.
26. Ng M, Freeman MK, Fleming TD, et al. Smoking prevalence and cigarette consumption in 187 countries, 1980-2012. JAMA. 2014;311(2):183-192.
27. King G, Polednak AP, Bendel R, et al. Cigarette smoking among native and foreign-born African Americans. Ann Epidemiol. 1999;9(4):236-244.
28. Taylor KL, Kerner JF, Gold KF, et al. Ever vs never smoking among
an urban, multiethnic sample of Haitian-, Caribbean-, and U.S.-born blacks.
Prev Med. 1997;26(6):855-865.
29. Siegel RL, Jacobs EJ, Newton CC, et al. Deaths Due to Cigarette
Smoking for 12 Smoking-Related Cancers in the United States. JAMA
Intern Med. 2015;175(9): 1574-1576.
30. Argeseanu Cunningham S, Ruben JD, Narayan KM. Health of foreignborn people in the United States: A review. Health Place. 2008;14(4):623-635.
31. Singh GK, Hiatt RA. Trends and disparities in socioeconomic and
behavioural characteristics, life expectancy, and cause-specific mortality of
native-born and foreign-born populations in the United States, 1979-2003.
Int J Epidemiol. 2006;35(4):903-919.
32. Kennedy S, McDonald JT, Biddle N. The healthy immigrant effect
and immigrant selection: Evidence from four countries. SEDAP Research
Program. Social and Economic Dimensions of an Aging Population.
SEDAP-II – Canada in the 21st Century: Moving Towards an Older Society.
Research paper No. 164. January-December, 2006. http://socserv.mcmaster.
ca/sedap/p/sedap164.pdf. Accessed July 19, 2016.
33. Aune D, Chan DS, Lau R, et al. Dietary fiber, whole grains, and risk
of colorectal cancer: systematic review and dose-response meta-analysis
of prospective studies. BMJ. 2011;343:d6617.
34.Greenberg MR, Schneider D, Northridge ME, et al. Region of
birth and black diets: the Harlem Household Survey. Am J Public Health.
1998;88(8):1199-1202.
35. Odedina FT, Dagne G, LaRose-Pierre M, et al. Within-group differences between native-born and foreign-born Black men on prostate cancer risk reduction and early detection practices. J Immigr Minor Health.
2011;13(6):996-1004.
36. American Cancer Society. Cancer Facts & Figures 2015. Atlanta, GA:
American Cancer Society; 2015.
37. Borrell LN, Castor D, Conway FP, et al. Influence of nativity
status on breast cancer risk among US black women. J Urban Health.
2006;83(2):211-220.
38. Celi AC, Rich-Edwards JW, Richardson MK, et al. Immigration,
race/ethnicity, and social and economic factors as predictors of breastOctober 2016, Vol. 23, No. 4
feeding initiation. Arch Pediatr Adolesc Med. 2005;159(3):255-260.
39. Singh GK, Kogan MD, Dee DL. Nativity/immigrant status, race/
ethnicity, and socioeconomic determinants of breastfeeding initiation and
duration in the United States, 2003. Pediatrics. 2007;119:suppl 1:S38-S46.
40. Consedine NS, Tuck NL, Ragin CR, et al. Beyond the black box:
a systematic review of breast, prostate, colorectal, and cervical screening
among native and immigrant African-descent Caribbean populations.
J Immigr Minor Health. 2015;17(3):905-924.
41. Gwede CK, William CM, Thomas KB, et al. Exploring disparities
and variability in perceptions and self-reported colorectal cancer screening among three ethnic subgroups of U. S. Blacks. Oncol Nurs Forum.
2010;37(5):581-591.
42.Magai C, Consedine N, Conway F, et al. Diversity matters:
Unique populations of women and breast cancer screening. Cancer.
2004;100(11):2300-2307.
43. Wilcox ML, Acuña JM, de la Vega PR, et al. Factors affecting compliance with colorectal cancer screening among households residing in the
largely Haitian community of Little Haiti, Miami-Dade County, Florida: an
observational study. Medicine (Baltimore). 2015;94(18):e806.
44. Derose KP, Escarce JJ, Lurie N. Immigrants and health care: sources
of vulnerability. Health Aff (Millwood). 2007;26(5):1258-1268.
45.The Florida Health Care Landscape. The Henry J. Kaiser Family
Foundation – The Kaiser Commission on Medicaid and the Uninsured.
November 2013 Fact Sheet. http://kff.org/health-reform/fact-sheet/theflorida-health-care-landscape/. Accessed July 19, 2016.
46. Forman D, Bray F, Brewster DH, et al. Cancer Incidence in Five
Continents, Vol. X. IARC Scientific Publications, No. 164. Lyon, France:
International Agency for Research on Cancer; 2014.
47. Jamison PM, Noone AM, Ries LA, et al. Trends in endometrial cancer incidence by race and histology with a correction for the prevalence of
hysterectomy, SEER 1992 to 2008. Cancer Epidemiol Biomarkers Prev.
2013;22(2):233-241.
48. Cote ML, Ruterbusch JJ, Olson SH, et al. The Growing Burden of
Endometrial Cancer: A Major Racial Disparity Affecting Black Women.
Cancer Epidemiol Biomarkers Prev. 2015;24(9):1407-1415.
49.Felix AS, Weissfeld JL, Stone RA, et al. Factors associated
with Type I and Type II endometrial cancer. Cancer Causes Control.
2010;21(11):1851-1856.
50. Daly B, Olopade OI. A perfect storm: how tumor biology, genomics,
and health care delivery patterns collide to create a racial survival disparity
in breast cancer and proposed interventions for change. CA Cancer J Clin.
2015;65(3):221-238.
51. Blake G, Hanchard B, Mitchell K, et al. Jamaica cancer mortality
statistics, 1999. West Indian Med J. 2002;51(2):64-67.
52. Bunker CH, Patrick AL, Konety BR, et al. High prevalence of screeningdetected prostate cancer among Afro-Caribbeans: the Tobago Prostate
Cancer Survey. Cancer Epidemiol Biomarkers Prev. 2002;11(8):726-729.
53. Consedine NS, Morgenstern AH, Kudadjie-Gyamfi E, et al. Prostate
cancer screening behavior in men from seven ethnic groups: the fear factor.
Cancer Epidemiol Biomarkers Prev. 2006;15(2):228-237.
54. Gonzalez JR, Consedine NS, McKiernan JM, et al. Barriers to the
initiation and maintenance of prostate specific antigen screening in Black
American and Afro-Caribbean men. J Urol. 2008;180(6):2403-2408.
55. Etzioni R, Penson DF, Legler JM, et al. Overdiagnosis due to
prostate-specific antigen screening: lessons from U.S. prostate cancer
incidence trends. J Natl Cancer Inst. 2002;94(13):981-990.
56. Baade PD, Youlden DR, Krnjacki LJ. International epidemiology of
prostate cancer: geographical distribution and secular trends. Mol Nutr
Food Res. 2009;53(2):171-184.
57. Glover FE Jr, Coffey DS, Douglas LL, et al. The epidemiology of
prostate cancer in Jamaica. J Urol. 1998;159(6):1984-1987.
58. Tsui J, Saraiya M, Thompson T, et al. Cervical cancer screening among foreign-born women by birthplace and duration in the United
States. J Womens Health (Larchmt). 2007;16(10):1447-1457.
59. Ryerson AB, Eheman CR, Altekruse SF, et al. Annual report to the
nation on the status of cancer, 1975‐2012, featuring the increasing incidence of liver cancer. Cancer. 2016;122(9):1312-1237.
60. Denniston MM, Jiles RB, Drobeniuc J, et al. Chronic hepatitis C
virus infection in the United States, National Health and Nutrition Examination Survey 2003 to 2010. Ann Intern Med. 2014;160(5):293-300.
61. Schweitzer A, Horn J, Mikolajczyk RT, et al. Estimations of worldwide prevalence of chronic hepatitis B virus infection: a systematic review
of data published between 1965 and 2013. Lancet. 2015;386(10003):
1546-1555.
62. Jemal A, Bray F, Center MM, et al. Global cancer statistics. CA Cancer
J Clin. 2011;61(2):69-90.
63. United Nations Development Programme Human Development
Reports. International Human Development Indicators. http://hdr.undp.
org/en/countries. Accessed July 19, 2016.
64. World Health Organization. Global status report on noncommunicable diseases 2014. Geneva: World Health Organization; 2014
65. Taioli E, Attong-Rogers A, Layne P, et al. Breast cancer survival in
women of African descent living in the US and in the Caribbean: effect of
place of birth. Breast Cancer Res Treat. 2010;122(2):515-520
Cancer Control 357
66. Mutetwa B, Taioli E, Attong-Rogers A, et al. Prostate cancer characteristics and survival in males of African Ancestry according to place
of birth: data from Brooklyn-New York, Guyana, Tobago and Trinidad.
Prostate. 2010;70(10):1102-1109.
67. Ragin C, Mutetwa B, Attong-Rogers A, et al. Geographic and outcome variation among black men diagnosed with prostate cancer. Infect
Agent Cancer. 2011;6(suppl 2):S2.
68. Rebbeck TR, Devesa SS, Chang BL, et al. Global patterns of prostate cancer incidence, aggressiveness, and mortality in men of african descent. Prostate Cancer. 2013;2013:560857.
69. Khalifa MM, Sharaf RR, Aziz RK. Helicobacter pylori: a poor man’s
gut pathogen? Gut Pathog. 2010;2(1):2.
70. Plummer M, Franceschi S, Vignat J, et al. Global burden of gastric
cancer attributable to Helicobacter pylori. Int J Cancer. 2015;136(2):487-490.
71. Wasley A, Kruszon-Moran D, Kuhnert W, et al. The prevalence of
hepatitis B virus infection in the United States in the era of vaccination.
J Infect Dis. 2010;202(2):192-201.
72. DeSantis CE, Siegel RL, Sauer AG, et al. Cancer statistics for
African Americans, 2016: progress and opportunities in reducing racial
disparities. CA Cancer J Clin. 2016;66(4):290-308.
73. Singh GK, Siahpush M, Hiatt RA, et al. Dramatic increases in obesity
and overweight prevalence and body mass index among ethnic-immigrant
and social class groups in the United States, 1976-2008. J Community
Health. 2011;36(1):94-110.
74. Bennett GG, Wolin KY, Okechukwu CA, et al. Nativity and cigarette
smoking among lower income blacks: results from the Healthy Directions
Study. J Immigr Minor Health. 2008;10(4):305-311.
75. Ford K. Duration of residence in the United States and the fertility of
U.S. immigrants. Int Migr Rev. 1990;24(1):34-68.
76. Porter P. “Westernizing” women’s risks? Breast cancer in lowerincome countries. N Engl J Med. 2008;358(3):213-216.
77. TheWorld Bank. Birth rate, crude (per 1,000 people). The World
Bank Group; 2013. http://data.worldbank.org/indicator/SP.DYN.CBRT.IN/
countries. Accessed July 20, 2016.
78. Mariotto AB, Noone AM, Howlader N, et al. Cancer survival: an
overview of measures, uses, and interpretation. J Natl Cancer Inst
Monographs. 2014;2014(49):145-186.
79. Blue L, Fenelon A. Explaining low mortality among US immigrants
relative to native-born Americans: the role of smoking. Int J Epidemiol.
2011;40(3):786-793.
80. Turra CM, Elo IT. The Impact of Salmon Bias on the Hispanic Mortality Advantage: New Evidence from Social Security Data. Popul Res
Policy Rev. 2008;27(5):515-530.
81. Phillips AA, Jacobson JS, Magai C, et al. Cancer incidence and
mortality in the Caribbean. Cancer Invest. 2007;25(6):476-483.
358 Cancer Control
October 2016, Vol. 23, No. 4
There is an urgent need to ensure that
existing genomic research considers
the unique needs of US Latinas with
breast cancer.
Chrysanthemum. X-Ray image courtesy of Bryan Whitney. www.x-rayphotography.com.
Genomic Disparities in Breast Cancer Among Latinas
Filipa Lynce, MD, Kristi D. Graves, PhD, Lina Jandorf, MA, Charité Ricker, MS, Eida Castro, PsyD,
Laura Moreno, Bianca Augusto, Laura Fejerman, PhD, and Susan T. Vadaparampil, PhD
Background: Breast cancer is the most common cancer diagnosed among Latinas in the United States and
the leading cause of cancer-related death among this population. Latinas tend to be diagnosed at a later stage
and have worse prognostic features than their non-Hispanic white counterparts. Genetic and genomic factors
may contribute to observed breast cancer health disparities in Latinas.
Methods: We provide a landscape of our current understanding and the existing gaps that need to be filled
across the cancer prevention and control continuum.
Results: We summarize available data on mutations in high and moderate penetrance genes for inherited
risk of breast cancer and the associated literature on disparities in awareness of and uptake of genetic counseling and testing in Latina populations. We also discuss common genetic polymorphisms and risk of breast
cancer in Latinas. In the treatment setting, we examine tumor genomics and pharmacogenomics in Latina
patients with breast cancer.
Conclusions: As the US population continues to diversify, extending genetic and genomic research into this
underserved and understudied population is critical. By understanding the risk of breast cancer among
ethnically diverse populations, we will be better positioned to make treatment advancements for earlier stages
of cancer, identify more effective and ideally less toxic treatment regimens, and increase rates of survival.
Introduction
An estimated 55 million individuals living in the United States identify as being Hispanic or Latino.1 Latinos
are a culturally and genetically diverse group with origins in Mexico, the Caribbean, Central America, and
South America. In the United States, 64.0% of Latinos
are of Mexican background, 9.6% of Puerto Rican
background, 3.8% of Salvadoran background, 3.7% of
Cuban background, 3.2% of Dominican background,
2.4% of Guatemalan background, and the remainder
From Georgetown Lombardi Comprehensive Cancer Center (FL, KDG), Washington, District of Columbia, Department of Hematology and
Oncology (FL), MedStar Georgetown University Hospital, Washington, District of Columbia, Department of Oncology (KDG), Georgetown
University, Washington, District of Columbia, Department of Oncological Science (LJ), Icahn School of Medicine at Mount Sinai, New York, New York,
Department of Medicine (CR), University of Southern California Norris Comprehensive Cancer Center, Los Angeles, California, Department of
Psychiatry (EC), Ponce Health Sciences University, Ponce, Puerto Rico, Health Outcomes and Behavior Program, H. Lee Moffitt Cancer Center &
Research Institute (LM, BA, STV), Tampa, Florida, and the Department of Medicine (LF), University of California, San Francisco, California.
Submitted April 4, 2016; accepted September 8, 2016.
Address correspondence to Susan T. Vadaparampil, PhD, Health Outcomes and Behavior Program, Moffitt Cancer Center, 12902 Magnolia Drive,
MRC-CANCONT, Tampa, FL 33612. E-mail: [email protected]
No significant relationships exist between the authors and the companies/organizations whose products or services may be referenced in this article.
Funding for this work was supported in part by U54 CA163071 (Ponce Health Sciences University) and U54 CA163068 (Moffitt Cancer Center).
October 2016, Vol. 23, No. 4
Cancer Control 359
are of other origins.2 Although the terms Hispanic
and Latino/Latina are often interchangeably used, we
selected the term Latina for the current manuscript as
we feel it extends beyond spoken language to reflect
both origin and cultural traditions of women from
Latin America.
Breast cancer is the most common cancer diagnosed among Latinas in the United States and is the
leading cause of cancer-related death in this population.3 Although the overall prevalence of breast cancer in Latinas is lower than in non-Latina whites, Latinas tend to be diagnosed at a later stage and have
worse prognostic features (eg, triple-negative disease,
ERBB2 [formerly HER2 or HER2/neu]–positive disease).4 A myriad of socioeconomic and cultural factors contribute to health disparities in breast cancer
among Latinas,5-7 but biological factors — particularly
genomics — remain an important but understudied
consideration.
High and Moderate Penetrance Genes
Approximately 10% to 15% of breast cancer cases are
attributed to inherited gene mutations. 8 Although
multiple genes confer an inherited risk for cancer,9
BRCA mutations are the most prevalent and penetrant
mutations, accounting for the majority of hereditary
types of breast cancer.10 BRCA mutations result in an
increased lifetime risk of breast cancer of up to approximately 60% to 70% and a lifetime ovarian cancer
risk of up to 40%.11-13 Among Latinas, breast cancer
is often diagnosed at younger ages and with worse
prognostic features, including increased rates of triple-negative disease, than their non-Hispanic white
counterparts.3,14-16 Triple-negative disease and premenopausal breast cancer are both clinical characteristics associated with a higher probability of having a
BRCA1 or BRCA2 mutation.17,18
Prevalence of BRCA
The prevalence of BRCA mutations in the general
US population is estimated to be 1 in 400, excluding
women of Ashkenazi Jewish descent in whom the prevalence is 1 in 40.19-21 However, less is known about the
prevalence among racial and ethnic minority groups,
including Latinas as a whole or by subethnicity
based on country of origin. A review examined the
spectrum of BRCA1 and BRCA2 mutations in Latin
America and the Caribbean using studies published
between the years 1994 and 2015.22 Six of the 33 studies were conducted among Latinas living in the United States, with the vast majority of participants drawn
from clinic-based samples of patients of Mexican origin with breast cancer residing in California, Arizona,
and Texas.22 Prevalence estimates of carrying a BRCA
mutation for this US Latina group ranged from 0.7% to
42.0% and varied based on whether cases were select360 Cancer Control
ed or unselected for family history or clinical characteristics (eg, affected vs unaffected, age at diagnosis),
cancer site (eg, breast, ovarian), and type of testing
(eg, inclusion of large rearrangement testing).22 In the
cohorts of unselected patients with breast cancer, the
BRCA mutation prevalence was 1.2% to 4.9%, which
was consistent with expected rates.22
BRCA mutations have also been documented in
all residents of Latin American countries where these
genes have been studied, including Argentina, Brazil, Chile, Colombia, Costa Rica, Cuba, Mexico, Peru,
Puerto Rico, Uruguay, and Venezuela.23-54 Most studies have focused on the spectrum of BRCA mutations.22,55 In a review of BRCA1 and BRCA2 mutations
in persons living in Latin America and the Caribbean,
36% of the 33 studies primarily focused on Mexican
or Mexican American patients.22 Of the Mexican study
population, the mutation prevalence was between
4.3% and 23.0%.22 For other Latina subethnic groups,
the mutation prevalence estimates of each country studied were: Colombia (1.2%–15.6%; 2 studies),
Costa Rica (4.5%; 1 study), Cuba (2.6%; 1 study), Peru
(4.9%; 1 study), Uruguay (17%; 1 study), and Venezuela (17.2%; 1 study).22 These studies provide insight
into areas of future research of BRCA mutation distribution and frequency based on country of origin,
the role of specific founder mutations, the contribution of large genomic rearrangements to the spectrum
of mutations across various Latina subethnic groups,
and the consideration of other non-BRCA genes that
increase the risk of breast cancer.
Although recurrent mutations were identified
within most studies, the specific mutation varied by
study and country.22 BRCA1 185delAG has also been
documented in Latinas across Latin America and the
United States.40,42-44,47,56-58 One of the 3 Jewish founder mutations, BRCA1 185delAG is estimated to have
arisen about 800 years ago or earlier and is believed
to have been introduced into Latin America about
650 years ago.59 When this mutation is identified in
Latinos, haplotype analysis supports that this mutation is of the same origin as the Jewish founder mutation, rather than a separate genetic event.60,61 Pooled
mutation estimates performed by Porchia et al55 found
that BRCA1 185delAG is the second most prevalent
BRCA1 mutation and its frequency is not significantly
different between Mexico and other Latin American
countries (P = .70). However, it is worth noting that
not all Central and South American countries were
represented in their analysis.55
The most common BRCA1 mutation in the same
meta-analysis was deletion of exons 9 to 12.55 This mutation is estimated to have originated nearly 1,500 years
ago near Puebla Mexico.48,58 However, to date, it has
been reported in Mexicans and Mexican Americans
alone.22,32,55,61,62 The contribution of large genomic reOctober 2016, Vol. 23, No. 4
arrangements to BRCA1 in Latin American patients
was evaluated in a study of US Latinas and described
the prevalence of rearrangements by racial and ethnic groups.63 Large rearrangements were significantly more common in individuals who reported Latin
American ancestry, and the prevalence of rearrangements was two-fold higher than in the overall population tested.63 This laboratory-based cohort extracted ethnicity data from genetic testing request forms;
therefore, no data about subethnicity was available.63
However, the 2 most frequent BRCA1 rearrangements
identified in this study were deletion of exons 9 to 12
and deletion of exons 1 and 2, likely reflecting the
underlying US Latino population in whom the majority is of Mexican ancestry.63 In a Puerto Rican study,
BRCA1 deletion of exons 1 to 2 was seen in nearly
20% of study patients positive for BRCA1.49 Because
Puerto Ricans represent the second largest US Latino
group after those of Mexican ancestry, these findings
support utilizing an assay that includes large rearrangements when testing Latinos.49 The study results
also highlight the importance of understanding more
granular aspects of ethnicity, such as country of origin, to ensure that all mutations that contribute significantly are captured.
Dutil et al22 noted that most Latin American studies they reviewed identified a higher proportion of
BRCA1 than BRCA2 mutations, a finding similar to
reports in other populations. However, studies from
4 different countries (Costa Rica,37 Cuba,35 Puerto
Rico,49 Uruguay64) reported more BRCA2 mutations
than those in BRCA1. While these studies may have
been limited by sample size and the mutation-detection strategies and technologies,35,37,49,64 this finding
has been also reported in a single US-based clinical
site and may warrant further exploration.65
The meta-analysis performed by Porchia et al55
identified recurrent BRCA2 mutations across all studies with the following pooled prevalence: H372N
(0.88%; 95% confidence interval [CI]: 0.24–1.92),
E49X (0.38%; 95% CI: 0.13–0.75), and 3492insT (0.32%;
95% CI: 0.24–0.53). BRCA2 3492insT has been identified
in different regions of Spain with a frequency as high
as 2.08%.65-71 Although it is possible that this mutation
was introduced in Latin America by the Spaniards, no
haplotype studies of this specific mutation were identified to confirm a shared ancestry rather than a separate
mutational event.49
BRCA1 and BRCA2 account for the majority of hereditary breast cancer, but other high- and
moderate-risk genes also predispose individuals to
breast cancer, including TP53, PTEN, CDH1, STK11,
CHEK2, PALB2, and ATM, among others.26,27,54,72-80
Limited studies have been performed of non-BRCA
genes in Latina breast-cancer cohorts, leaving much
to be learned about the prevalence and spectrum of
October 2016, Vol. 23, No. 4
mutations in these genes among Latinas with breast
cancer (Table 1).26,27,54,72-80
One exception is the Brazilian founder mutation
in TP53, R337H. Mutations in TP53 cause Li-Fraumeni
syndrome, which is associated with an elevated risk
for a wide spectrum of cancers, including adrenal cortical carcinoma, soft-tissue and bone sarcomas, brain
tumors, and breast cancer.81-83 The overall contribution of TP53 mutations to breast cancer is estimated to
be less than 1%, unless selecting for early-onset breast
cancer.84,85 In studies of women diagnosed with breast
cancer at or before the age of 30 or 35 years, 5% to 8%
had TP53 mutations.81,86-88
TP53 R337H was first identified in individuals
with childhood adrenal cortical carcinomas living
in southern Brazil.89 This mutation occurs in 2.4% to
8.6% of Brazilian women with breast cancer.78,79,89,90 In
a large study, which included 403 patients with breast
cancer diagnosed at 45 years or younger, 12.1% carried the TP53 R337H mutation. Although the mutation was significantly more frequent in younger patients compared with those diagnosed at 55 years or
older (P < .001), 5.1% of the older group carried the
mutation.79 To date, no other populations have been
identified in whom TP53 makes such a significant
contribution to breast cancer. The prevalence of this
mutation in southern Brazil has been estimated to be
approximately 0.3%.91,92 Additional haplotype analyses support the hypothesis that this recurrent mutation is a founder mutation from a shared ancestor.79,93
Historically, the genetic assessment for hereditary
breast cancer involved the formation of a differential diagnosis followed by a syndrome-by-syndrome
evaluation through the sequential testing of genes.
However, the rapid integration of next-generation
sequencing has enabled simultaneous testing of multiple inherited cancer genes, thereby expanding the
use of multigene panels in clinical testing at a reduced
cost.94 This expansion is reflected in the emerging
body of literature on breast cancer focused on multigene panel findings from the research, clinical, and
laboratory settings.95-103
These literature cohorts are predominantly
non-Hispanic whites, with Latinas representing
less than 1.0% to 7.4% of study participants, thus
highlighting another area where future research is
needed.95-97,99-102 One study of 475 patients undergoing
multigene panel testing included 228 Latino patients
(47.6% of the study population), and it reported that
the likelihood of detecting a deleterious mutation was
no different among the ethnic and racial groups represented.96 Of the patients with breast cancer (n = 197),
14.8% (n = 28) carried mutations, and, as expected,
BRCA1 and BRCA2 were the most commonly mutated
genes; however, 16 mutations were identified in other
genes (CDH1 = 4, CHEK2 = 3, MUTYH = 3, PALB2 = 2,
Cancer Control 361
TP53 = 1, RAD50 = 1, RAD51D = 1, BARD1 = 1).96 Of
note, the likelihood of identifying more than 1 variant
of uncertain significance in Latinos was significantly
higher than that of non-Hispanics whites.104 Thus, a
need exists for further research to better classify rare
variants, especially given the under-representation of
Latinos in laboratory and research databases.
Genetic Counseling and Testing
Patient- and Health Care–Related Factors
An important step toward understanding the role of
BRCA and other high- and moderate-risk breast cancer
genes in Latinas is to increase the number of individu-
als who receive genetic counseling and subsequently
elect to undergo testing. However, growing evidence
identifies disparities in awareness of and access to
genetic counseling among Latinas compared with
non-Hispanic white women. Data from health interview surveys from 2000, 2005, and 2010 show that Latinas had the lowest level of awareness about genetic
testing for inherited cancer risk than all of the other
US racial ethnic groups.105-107 Using telephone surveys,
Gammon et al108 studied 63 Latinas and 84 non-Hispanic whites at increased risk for carrying a BRCA1
or BRCA2 mutation, examining their awareness, cognition level, and psychosocial needs related to genetic
Table 1. — Select Non-BRCA Genes Observed in Latina Populations
Study
Country
Cohort
No. of
Patients
Assumpção78
Brazil
Gene
Analysis
Findings
Inclusion
Criteria
123
Family history of
breast cancer
Family history of
ovarian cancer
Sporadic breast cancer
Carraro54
54
Early-onset breast cancer
diagnosis < 30 y
Felix 26
106
HBOC testing
Giacomazzi79
874
Family history of cancer
(group 1)
Consecutive breast cancer
(group 2)
Silva 27
120
HBOC testing
TP53
Site-specific analysis of
TP53 R337H
2.4% of cases carried the
mutation (P = .0442)
BRCA1
BRCA2
CHEK2
TP53
Coding introns/exons of
BRCA1/2, TP53, and
site-specific analysis of
CHEK2 (c.1100delC)
22% carried mutations
mostly in BRCA1/2
2% (n = 1) had TP53
mutation
PCR of each exon of BRCA1
Site-specific analyses of
BRCA2 (c.5946_5946delT;
c.156_157insAlu),
CHEK2 (c.1100delC;
c.444+1G>A; p.I157T),
and TP53 (p.R337H)
2.8% carried mutations
(BRCA = 2, TP53 = 1)
Single-site analysis for
p.R337H
p.R337H identified in
3.4% (group 1) and
8.6% (group 2)
Higher prevalence when
diagnosed ≤ 45 y (12.1%)
vs 55 y (5.1%; P < .001)
Coding introns/exons of
BRCA1/2
Site-specific analysis of
CHEK2 (c.1100delC) and
TP53 (p. R337H)
Array comparative genomic
hybridization for CNVs in
other 14 genes
26% (n = 31) mutations
(BRCA1/2 = 27, CHEK2 = 1,
TP53 = 3)
BRCA1/2
CHEK2
TP53
TP53
ATM
BRCA1/2
BRIP1
CDH1
CDKN2A
CTNNB1–
CHEK2
MLH1
MSH6
NBN
PALB2
PTEN
RAD50
RAD51
TP53
Continued on next page
362 Cancer Control
October 2016, Vol. 23, No. 4
Table 1. — Select Non-BRCA Genes Observed in Latina Populations, continued
Study
GonzálezHormazábal72
Country
Chile
Jara80
Leyton73
Cohort
Gene
Analysis
Findings
No. of
Patients
Inclusion
Criteria
137
(BRCA – = 126,
BRCA+ = 11)
≥ 2 family members with
breast cancer
≥ 2 family members with
ovarian cancer
Family history of male
breast cancer
Early-onset breast cancer
with no family history
ATM
PCR-based analysis of
coding sequence and exon/
intron boundaries of ATM
Analysis of ATM 5557G>A,
IVS38-8T>C, IVS24-9delT
5557G>A, IVS38-8T>C,
IVS24-9delT associated
with elevated risk of breast
cancer if BRCA –
Identification of composite
genotype that confers
3.19-fold risk for breast
cancer
143
(BRCA – = 131,
BRCA+ = 12)
≥ 2 family members with
breast cancer
≥ 2 family members with
ovarian cancer
Family history of male
breast cancer
Early-onset breast cancer
with no family history
RAD51D
PCR-based analysis of
coding sequence and
exon-intron boundaries of
RAD51D
Analysis of RAD51D,
c.135G>C
No mutations detected in
RAD51D
c.135G>C associated with
elevated breast cancer risk
if BRCA –
436
BRCA+
≥ 2 family members with
breast cancer
≥ 2 family members with
ovarian cancer
Single case of early-onset
diagnosed ≤ 50 y
PALB2
Full gene sequencing in
100 “high-risk” cases
Analysis of identified variants
No pathogenic mutations
identified
3 variants identified
(c.1676A>G a , c.2993C>Ta ,
c.1861C>A)
PCR-FLP of 3 specific
mutations (IVS24-9delT,
IVS38-8T>C, 5557G>A)
5557G>A (13%)
IVS24-9delT (21% vs
8% controls; P = .0122)
IVS38-8T>C (< 1%)
CalderónZúñiga74
Mexico
94
Familial breast cancer
Early-onset breast cancer
ATM
Bell76
United
States
362b
Early-onset breast cancer
CHEK2
169 cases diagnosed ≤ 40 y
had sequencing of coding
region of CHEK2
Specific analysis of
1100delC, H143Y, and 8 other
CHEK2 variants/mutations
Data not reported by
ethnicity, but reported
“infrequency” of
c.1100delC among Latinas
Bretsky75
101b
Personal history of breast
cancer
ATM
20 specific ATM missense
mutations or polymorphisms
L546V had modest but
not significant predictor of
risk; almost exclusive to
African American women
(found in 2 Latinas)
Damiola77
158 b
Breast cancer diagnosed
≤ 45 y
MRE11
RAD50
NBN
PCR-based analysis of
coding sequence and exon/
intron boundaries of MRE11,
RAD50, NBN
Data not reported by
ethnicity
MRE11, RAD50, NBN are
intermediate-risk genes
aThese variants play a role in risk of breast cancer.
bLatinas were part of a larger multiethnic cohort.
CNV = copy number variation, FLP = fragment length polymorphism, HBOC = hereditary breast and ovarian carcinoma, PCR = polymerase chain reaction.
counseling and testing. Among those who had not previously undergone genetic counseling (53 of the 120),
Latinas were more unaware than their white counterparts of the availability of testing (56.9% vs 34.8%,
respectively).108 Vadaparampil et al109 reported on a
sample of Latinas with a personal or family history of
breast cancer, all of whom reported an awareness of
genetic risk for breast cancer (ie, family history). However, none of the Latinas had a clear understanding of
October 2016, Vol. 23, No. 4
what genetic testing was and had not received physician referral for genetic testing.109 Findings did differ
based on country of origin — an important area to consider in future work, given the diversity of Hispanic
populations across the United States.109 In another report, Kaplan et al110 reported differences in awareness
of genetic testing by race and ethnicity, such that 19.4%
of Latinas had heard of genetic testing compared with
59.4% of whites, 26.1% of Asian Americans, and 31.0%
Cancer Control 363
of black women.
In a study of more than 2,400 patients completing a family cancer history form, Mays et al111 found
that, overall, despite low levels of initial awareness,
65 patients (2.7%) met criteria for cancer risk assessment; of those, 72.3% expressed interest in receiving
genetic counseling. Furthermore, no differences in interest in genetic services were reported across all racial
and ethnic groups.111 Among 1,536 women with nonmetastatic breast cancer, Jagsi et al112 found that Latinas
had a greater desire for genetic counseling than other
groups (58.8% of Spanish-speaking Latinas; 36.7% of
English-speaking Latinas; 27.1% of non-Latina whites;
and 28.1% of blacks). In addition, Lagos et al113 examined social, cognitive, and cultural variables among Latinas prior to an appointment for genetic counseling.
Fifty low-income, underserved Latinas completed the
assessment, and the results demonstrated their readiness (having the necessary skills for the genetic-counseling process), low fatalism, and high rate of self-efficacy, and social support.113 However, this study was
conducted in women who showed up to their genetic
counseling appointments, thus representing a unique
group of women.113 Vadaparampil et al114 studied a
group of Puerto Rican women (living in Puerto Rico
or central Florida) with a family or personal history of
breast cancer and found that the vast majority of participants said they would undergo genetic testing within
the next 6 months if it was available. Barriers included
the potential physical pain associated with the test.
Uptake of Services
Given lower levels of patient awareness, physician
recommendations may provide a critical approach to
increasing the utilization of genetic counseling and
testing for hereditary risk of breast cancer. However,
available studies suggest a missed clinical opportunity, because both English- and Spanish-speaking
Latina survivors of breast cancer may be more likely
to have unmet needs for discussion with a health care
professional about genetic testing for cancer than
their non-Hispanic white counterparts. For example,
Jagsi et al112 reports that minority patients were the
most likely to express an unmet need for a discussion
about genetic testing, although they also showed a
strong desire for such testing.
Preliminary studies support the uptake of genetic
counseling when services are offered.115-118 One study
of predominantly Latina patients (71.4%) offered genetic counseling at a safety-net hospital found that 88.0%
kept their appointments.115 Another study of women
(69.6% were Latinas) seen in a safety-net hospital setting reported that 96.4% of them underwent BRCA testing when it was recommended to them.116 Once Latinas
were referred, Olaya et al117 found that they are equally
likely as the general population to complete BRCA test364 Cancer Control
ing. Overall, 52% completed genetic testing, and no differences by race and ethnicity were observed.117 Woodson et al118 reported on the utilization of group pretest
genetic counseling in a community clinic made up of
mostly Latinas (62.3%) with breast cancer; the majority
(86.7%) underwent BRCA genetic testing when offered.
Overall, these studies have focused on the delivery of genetics services for cancer to majority Latina cohorts, demonstrating that genetic counseling and testing is likely well-received by Latinas with breast cancer;
however, these studies were all conducted in safety-net
hospitals or in community, low-resource settings and
were aimed at the provision of service to low-income,
uninsured patients.115-118 Although Latinas continue to
be disproportionally uninsured or underinsured, these
study findings might not generalize to other health care
settings. Thus, further studies are needed across various
clinical settings and in a wider representation of Latinas
with breast cancer to better understand the utilization of
genetic testing as well as the barriers for referrals.
Common Genetic Polymorphisms and
Risk of Breast Cancer
Genome-Wide Association Studies
Progress in the discovery of germline genetic polymorphisms associated with breast cancer risk changed pace
when technological advances in genotyping made it
possible to characterize genome-wide genetic variation
at a relatively low cost.119 In 2007, the first breast cancer
genome-wide association studies were published, and
they reported a handful of single nucleotide polymorphisms (SNPs) associated with a modest increase in
risk.120-122 Since then, more than 100 common variants
that either increase risk for or are protective against
developing breast cancer have been discovered and,
including replication efforts, data from more than
120,000 women have been analyzed.120-143 A small proportion of samples included in these major initiatives
are from minority populations in the United States
(eg, Latinas, African Americans),120-143 and the first results of genome-wide association studies of Latinas
with breast cancer were published in 2014.144 This latter study represents important but limited progress,
considering that the sample size was one-tenth of that
available for genome-wide association studies involving women of European origin.120-122,144
Until the first genome-wide association studies of
breast cancer in women of European origin were published, the search for risk-predisposing genetic variants
was focused on finding polymorphisms within genes
that, for known or hypothesized involvement in the biology of the disease, were likely to contribute to breast
cancer risk.120-122 These studies in US Latinas or Latin
American women typically consisted of the replication
of previously associated polymorphisms reported in
Europeans, with few of these studies looking for variaOctober 2016, Vol. 23, No. 4
tion in samples of Latinas before further testing specific
polymorphisms for associations in larger samples.145,146
Compared with the hundreds of genome-wide association studies in non-Hispanic white women, we
identified 13 case-control studies or cohorts that include US Latina or Latin American women.147-160 These
studies include populations of women of no more than
100 and up to approximately 5,000 women of Latin
American origin; combined, the study populations tally approximately 5,000 Latina women with breast cancer and 11,000 Latina healthy controls.147-160
Candidate Gene or Pathway Studies
Multiple breast cancer–association studies of candidate
genes or pathways have been reported for US Latina
and Latin American women during the last 20 years.
Genes or pathways studied have included those related to hormone metabolism, hormone receptors,
hormone coactivators or supressors,145,161-166 growth
factors,146,167-173 matrix metalloproteinases,174,175 inflammation and energy balance,149,176-181 metabolism of xenobiotic compounds and oxidative stress,150,182-184 DNA
repair,151,152,154,186 and angiogenesis.187 Results reported
in these publications should be interpreted with caution, given that approximately 60% of the candidate
gene analyses included did not adjust for genetic ancestry, which is a known confounder in genetic association studies in admixed populations.188,189 In addition,
no associations in candidate gene or pathways studies,
nor any of the interactions with risk factors, genetic ancestry, or tumor characteristics, have been replicated in
independent samples of Latinas.
Replication of Identified Single Nucleotide
Polymorphisms
Few studies have included Latinas and tested the association between SNPs discovered in genome-wide
association studies of breast cancer conducted in samples of European or Asian women.190-195 The first study
genotyped previously reported SNPs in the 2q35 region
and FGFR2, TOX3, and MAP3K1, reporting statistically significant replications for the polymorphisms
in FGFR2 and 2q35.190 Two different studies published
the results of analyses conducted in the same sample of
high-risk families from Chile and healthy controls, testing associations between previously reported variants
in FGFR2, MAP3K1, and TOX3 and the 2q35 and 8q24
regions and breast cancer risk.194,195 They replicated the
associations for FGFR2, MAP3K1, TOX3, and 2q35 but
not for 8q24.194,195 An analysis conducted in a pooled
sample of Latina cases and controls from the FourCorners study, San Francisco Bay Area Breast Cancer
Study, and a study in Mexico, investigated the association between 10 identified polymorphisms in genomewide association studies (in region 2q35 and in or near
RELN, MRPS30, RNF146, FGFR2, TOX3, LSP1, TLR1,
October 2016, Vol. 23, No. 4
MAP3K1, and RAD51L1) and breast cancer risk.192
They replicated associations for the polymorphisms
in RELN, FGFR2, TOX3, and TLR1 and 2q35 and found
heterogeneity by ancestry for the RELN, 2q35, and
TLR1 SNPs.192 A follow-up study reported that the heterogeneity by ancestry for the 2q35 polymorphism was
likely due to the association between genetic ancestry,
use of hormone therapy, and breastfeeding.191 Another analysis of the FGFR2 polymorphism in the Mexican study reported an interaction between the FGFR2
polymorphism and alcohol intake.193 The first genomewide association study of breast cancer in US Latinas
also replicated previous associations, with most of the
SNPs being concordant in terms of direction and magnitude of association with those reported in European
or Asian populations.144 Twenty-three of the 83 variants tested had probability values below .05.144
Ancestry
Admixture mapping leverages the demographical history of admixed populations to find genomic regions
that may carry trait-associated variants.196-204 An admixed population results from the combination of 2 or
more ancestral groups.200 The principle of admixture
mapping is to identify genomic regions in which cases
share more of the same genetic ancestry than either
population-based controls (case-control analysis) or
compared with the average ancestry of the rest of the
genome among cases (case-only analysis).202 This approach has identified risk variants or risk regions for
multiple complex traits, including obesity, hypertension, and cancer.196-199,201,203,204 The incidence of breast
cancer varies across different racial and ethnic groups
in the United States, and Latinas have lower incidence
rates than non-Latina whites but higher rates than
American Indian women.205 Genetic ancestry has also
been associated with breast cancer risk in US Latinas
and Mexican women after adjusting for nongenetic risk
factors, suggesting that a genetic component could be
responsible for the difference in risk.149,206,207 An admixture mapping study in Latinas reported a statistically significant association between a region in the
long arm of chromosome 6 (6q25) near ESR1 and risk
of breast cancer and a suggestive association on chromosome 11.208 Higher Indigenous American ancestry
at chromosome 6q25 was associated with lower risk of
breast cancer.208 This finding was concordant with the
previous reports of lower rates of risk of breast cancer
among Latinas with high American Indian ancestry
compared with women with high European ancestry
after adjusting for possible risk factors such as socioeconomic status, number of full-term pregnancies, and
breast feeding.206-208
One included a discovery phase and replication
in 3 additional studies.144 The study reported genome-wide results that were statistically significant
Cancer Control 365
for 2 linked SNPs 56kb upstream of ESR1 (rs140068132
and rs147157845).144 These SNPs have a frequency
of between 5% and 23% in Latin American populations and are absent in most all other groups.144 The
minor allele was protective, with an associated odds
ratio (OR) of 0.60 (95% CI: 0.53–0.67) per allele and
was more protective for ER-negative disease than for
ER-positive disease (OR for ER-negative disease 0.34;
95% CI: 0.21–0.54).144
Treatment
Tumor Genomics
A growing body of evidence suggests differences in
the tumor biology of breast carcinoma across various races and ethnicities. Several studies have evaluated the prevalence of phenotypic subtypes of breast
cancer in Latinas compared with other population
groups.209-215 Most data have shown a higher proportion of HR-negative disease types among Latinas when
compared with non-Hispanic whites (Table 2).4,209-213
However, those results have not always been concordant, and the differences seen across these studies
could represent small sample sizes, patient age, or unadjusted rates for genetic ancestry. Although studies
based on data from the California Cancer Registry indicated a higher proportion of triple-negative tumors,214
this finding was not confirmed in a Colorado study.209
In a retrospective study performed in Brazil, patients
in the southern regions with a higher percentage of European ancestry and higher socioeconomic status presented with the highest proportion of luminal tumors,
whereas the more aggressive subtypes were seen in
the northern parts of Brazil, an area with a higher African ancestral influence.215
Approximately 15% of breast cancer types over-
express human epidermal growth factor receptor 2
(ERBB2; formerly HER2 or HER2/neu) protein.216 High
levels of ERBB2 expression identify those women who
benefit from treatment with ERBB2-targeted agents,
which have been shown to increase survival in the adjuvant and metastatic settings.217,218 Most studies with
ERBB2-targeted therapies have enrolled majority
populations of non-Hispanic whites, although consistent evidence demonstrates that a higher proportion
of ERBB2-positive tumors exist among Latinas, even after adjusting for other tumor characteristics (eg, grade,
stage, ER status) and breast cancer risk factors (eg, number of children, alcohol consumption).209
Further tumor characterization has been made
possible due to advances in molecular tumor profiling. Oncotype DX (Genomic Health, Redwood City,
CA) is a 21-gene breast cancer assay — known as a
recurrence score — that provides prognostic and predictive information regarding the benefits of adjuvant
chemotherapy in patients with ER-positive tumors.
Use of Oncotype DX is part of several guidelines from
professional medical organizations, including the National Comprehensive Cancer Network, the American
Society of Clinical Oncology, and the European Society
for Medical Oncology.219-221 The characteristics of this
assay and the impact of its results on treatment decisions among Latinas with breast cancer are lacking in
the medical literature. Kalinsky et al222 studied 74 Latinas and 145 non-Hispanic white women matched for
age, disease stage, and nodal status, and they observed
no differences in the overall recurrence score, ER
or PR status, or ERBB2 expression by Oncotype DX.
However, Latinas had a higher expression of CCNB1
and AURKA, 2 genes that are part of the proliferation
score and heavily weighted in the calculation of the
Table 2. — Comparison of ER and ERBB2 Status in Latina, Black, and White Women
Study
No. of Patients
Latina
White
Black
Chlebowski211
N = 3,800
n = 103 Latinas
ER+: 83.0%
ER+: 87.0%
ER+: 71.0%
Dunnwald212
N = 209,276
n = 5,585 Latinas
ER+: 70.2%
ER+: 78.4%
ER+: 60.5%
Hausauer 213
N = 243,906
n = 15,355 Latinas
ER+: 53.2%
(unknown ER status: 29.4%)
ER+: 63.5%
(unknown ER status: 22.3%)
ER+: 46.4%
(unknown ER status: 29.4%)
Hines209
N = 285
n = 69 Latinas
ER+: 63.8%
ERBB2 +: 31.9%
TNBC: 17.4%
ER+: 77.3%
ERBB2 +: 14.3%
TNBC: 15.1%
Li210
N = 124,934
n = 7,219 Latinas
ER+: 68.7%
ER+: 78%
ER+: 53.4%
Parise 4
N = 143,184
n = 24,078 Latinas
ER+: 74.6 %
ERBB2 +: 22.2%
TNBC: 15.9%
ER+: 82.7%
ERBB2 +: 17.2%
TNBC: 11.2%
ER+: 66.5%
ERBB2 +: 20.1%
TNBC: 24.5%
—
TNBC = triple-negative breast cancer.
366 Cancer Control
October 2016, Vol. 23, No. 4
recurrence score.
Multiple trials whose study populations were
mostly comprised of non-Hispanic white women have
shown that use of Oncotype DX affects treatment recommendations and leads to an increase in physician
and patient confidence in treatment decisions.223-226 A
small study of 96 patients with breast cancer treated in
Mexico showed that use of the Oncotype DX changed
treatment decisions for 32% of patients, a finding suggesting that its use has a meaningful impact on recommendations for adjuvant treatment.227 Results from
cost-effectiveness analyses indicated that use of the
Oncotype DX assay was projected to improve rates of
life expectancy when compared with the current standard of care.228
Pharmacogenomics
Several factors cause variations in an individual’s response to drugs, including age, body mass index,
diet, and genetic variation.229-231 SNPs in genes related
to drug-metabolizing enzymes have been recognized
as important determinants of variability to drug response.232 Most studies have not been sufficiently
powered to determine whether specific chemotherapy
agents used to treat breast cancer have different rates
of effectiveness and toxicities based on race or ethnicity.233 However, differences in the metabolism of endocrine therapies have been well documented according
to race and ethnicity.234-237
In a study that evaluated clinical data and blood
samples from patients with breast cancer undergoing
adjuvant tamoxifen therapy, mostly non-Hispanic white
women (68%) and Latinas (26%) had significantly higher
serum levels of tamoxifen and 4-hydroxytamoxifen,
one of the tamoxifen metabolites (P = .02 and P = .007,
respectively).234 In 2 other studies, genetic polymorphisms
in CYP2D6 associated with lower plasma concentrations
of the active metabolites of tamoxifen were described
in Mexican, Puerto Rican, and Spanish patients.235,236
A higher prevalence of this poor metabolizer phenotype has also been observed in non-Hispanic whites.237
In an attempt to clarify whether CYP2D6 allele status
influences outcomes from tamoxifen, investigators assessed data from 2 large prospective trials and found
that CYP2D6 allele status did not predict clinical benefit of adjuvant tamoxifen in terms of risk of recurrence.238,239 Therefore, changes in treatment decisions
based on CYP2D6 allele status alone are not recommended. Differences in the incidence of polymorphisms of the aromatase gene among different ethnic
groups have also been reported and could potentially
lead to different outcomes and toxicities among populations.240 One trial evaluated the benefit of extended
hormonal therapy with an aromatase inhibitor after
5 years of tamoxifen treatment in non-Hispanic whites
(n = 4,708) and minority women (n = 352; 1.5% were
October 2016, Vol. 23, No. 4
Latinas).241 In general, the researchers found that, compared with non-Hispanic whites, minorities had fewer
associated toxicities and no definitive survival benefit
with aromatase inhibitors.241 However, these results
should be cautiously interpreted, because the minority
participants were less adherent to hormonal therapy
and the study was not powered to detect survival benefit in the subgroups.241
Conclusions
Several genetic and genomic factors are related to the
health disparities of breast cancer in Latinas. Increasing our knowledge about the contribution of high- and
moderate-penetrance mutations to the risk of breast
cancer among Latinas overall and for subgroups based
on country of origin is an important priority. Although
genetic counseling and testing for inherited susceptibility to breast cancer has been clinically available for
nearly 20 years, disparities in awareness, referral to
services, and access persist. Therefore, interventions to
address barriers related to low levels of awareness and
lacking physician referrals are critical.105-110
Even though multiple candidate gene studies
have been conducted in this population, only identified variants in genome-wide association studies have
been systematically replicated. Much larger sample
sizes will be required if we expect to discover similar
results in Latinas as would be identified for the European genome in view of their Indigenous American
component. We cannot assume that overlapping variants alone between different ancestral genomes will
be associated with rate of risk, so our efforts should be
focused on reducing research disparities by expanding available resources to include large cohorts and
case-control studies of diverse populations in and outside the United States.
Latinas remain systematically underrepresented
in pharmacogenomics studies and the current studies
were not powered to detect outcome differences. They
are also underrepresented in clinical treatment trials
and other patient-reported outcomes research. Future
research should draw from the few models of success
in prior studies that have recruited sufficient numbers
of Latinas.242-245 Lessons can also be learned from successful examples of recruiting other racial and ethnic
minority patients who have survived breast cancer.246
Elements that appear to bolster success include partnering with community-based organizations that provide services to Latinas and the provision of languageconcurrent clinical care.247-249 The heterogeneity of the
US Latina population must also be considered, because
different cultural influences, levels of awareness of,
and interest in genetic and genomic services appear to
vary by country of origin.109
An urgent need exists to ensure that existing genomic research considers the unique needs of this
Cancer Control 367
Latina population. As the US population continues to
diversify with up to one-third identifying as Hispanics by 2060,1 extending genetic and genomic research
into this underserved and understudied population
will be critical. By understanding the risk of breast cancer among diverse populations, we will be better positioned to make advancements in the number of women diagnosed at earlier stages, identify more effective
and less toxic treatment regimens, and increase rates of
survival. Meeting these goals will contribute to reducing the current health disparities in these patients with
breast cancer.
Acknowledgment: The authors wish to thank
Lindsay Salem for her assistance in the literature review for this manuscript.
References
1. Colby SL, Ortman JM. Projections of the size and composition of the
US population: 2014 to 2060. Curr Pop Rep. 2014:1-13 https://www.census.gov/
content/dam/Census/library/publications/2015/demo/p25-1143.pdf. Accessed
September 14, 2016.
2. Pew Research Center. Statistical portrait of hispanics in the United
States, 2014. http://www.pewhispanic.org/files/2016/04/Statistical-Portrait-ofHispanics-in-the-United-States-2014-final.pdf. Accessed September 14, 2016.
3. American Cancer Society. Cancer Facts & Figures for Hispanics/
Latinos 2015-2017. http://www.cancer.org/acs/groups/content/@research/
documents/document/acspc-046405.pdf. Accessed September 14, 2016.
4. Parise CA, Caggiano V. The Influence of socioeconomic status on
racial/ethnic disparities among the ER/PR/HER2 breast cancer subtypes.
J Cancer Epidemiol. 2015;2015:813456.
5. Quinn GP, McIntyre JQ, Vadaparampil ST. Challenges in recruiting Mexican women for cancer genetics research. J Community Genet.
2011;2(1):43-47.
6. Giuliano AR, Mokuau N, Hughes C, et al. Participation of minorities
in cancer research: the influence of structural, cultural, and linguistic factors.
Ann Epidemiol. 2000;10(8 suppl):S22-S34.
7. Lantz PM, Mujahid M, Schwartz K, et al. The influence of race,
ethnicity, and individual socioeconomic factors on breast cancer stage at
diagnosis. Am J Public Health. 2006;96(12):2173-2178.
8. Ellsworth RE, Decewicz DJ, Shriver CD, et al. Breast cancer in the
personal genomics era. Curr Genomics. 2010;11(3):146-161.
9. Lindor NM, McMaster ML, Lindor CJ, et al. Concise handbook of
familial cancer susceptibility syndromes - second edition. J Natl Cancer
Inst Monogr. 2008(38):1-93.
10. Euhus DM, Robinson L. Genetic predisposition syndromes and
their management. Surg Clin North Am. 2013;93(2):341-362.
11. Chen S, Parmigiani G. Meta-analysis of BRCA1 and BRCA2 penetrance.
J Clin Oncol. 2007;25(11):1329-1333.
12. Antoniou A, Pharoah P, Narod S, et al. Average risks of breast and
ovarian cancer associated with BRCA1 or BRCA2 mutations detected in
case series unselected for family history: a combined analysis of 22 studies.
Am J Hum Genet. 2003;72(5):1117-1130.
13. Litton JK, Ready K, Chen H, et al. Earlier age of onset of BRCA mutationrelated cancers in subsequent generations. Cancer. 2012;118(2):321-325.
14. Miranda PY, Wilkinson AV, Etzel CJ, et al. Policy implications of early
onset breast cancer among Mexican‐origin women. Cancer. 2011;117(2):390-397.
15. Boyle T, McPadden E. Breast cancer presents at an earlier age in Mexican
American women. Breast J. 2004;10(5):462-464.
16. Lara‐Medina F, Pérez‐Sánchez V, Saavedra‐Pérez D, et al. Triple‐
negative breast cancer in Hispanic patients. Cancer. 2011;117(16):3658-3669.
17. Claus EB, Schildkraut JM, Thompson WD, et al. The genetic attributable risk of breast and ovarian cancer. Cancer. 1996;77(11):2318-2324.
18. Hartman AR, Kaldate RR, Sailer LM, et al. Prevalence of BRCA
mutations in an unselected population of triple‐negative breast cancer.
Cancer. 2012;118(11):2787-2795.
19. Anglian Breast Cancer Study Group. Prevalence and penetrance
of BRCA1 and BRCA2 mutations in a population-based series of breast
cancer cases. Br J Cancer. 2000;83(10):1301-1308.
20. Whittemore AS, Gong G, John EM, et al. Prevalence of BRCA1 mutation
carriers among US non-Hispanic Whites. Cancer Epidemi Biomark Prev.
2004;13(12):2078-2083.
21. Struewing JP, Hartge P, Wacholder S, et al. The risk of cancer associated
with specific mutations of BRCA1 and BRCA2 among Ashkenazi Jews. N
Engl J Med. 1997;336(20):1401-1408.
22. Dutil J, Golubeva VA, Pacheco-Torres AL, et al. The spectrum of
368 Cancer Control
BRCA1 and BRCA2 alleles in Latin America and the Caribbean: a clinical
perspective. Breast Cancer Res Treat. 2015;154(3):441-453.
23. Gomes MC, Costa MM, Borojevic R, et al. Prevalence of BRCA1
and BRCA2 mutations in breast cancer patients from Brazil. Breast Cancer
Res Treat. 2006;103(3):349-353.
24. Esteves V, Thuler L, Amêndola L, et al. Prevalence of BRCA1 and
BRCA2 gene mutations in families with medium and high risk of breast and
ovarian cancer in Brazil. Brazl J Med Biol Res. 2009;42(5):453-457.
25. Ewald IP, Izetti P, Vargas FR, et al. Prevalence of the BRCA1 founder mutation c. 5266dupin Brazilian individuals at-risk for the hereditary
breast and ovarian cancer syndrome. Hered Cancer Clin Pract. 2011;9:12.
26. Felix GE, Abe-Sandes C, Machado-Lopes TM, et al. Germline mutations in BRCA1, BRCA2, CHEK2 and TP53 in patients at high-risk for HBOC:
characterizing a Northeast Brazilian population. Human Genome Variation.
2014;1:14012.
27. Silva FC, Lisboa BC, Figueiredo MC, et al. Hereditary breast and
ovarian cancer: assessment of point mutations and copy number variations
in Brazilian patients. BMC Medi Genet. 2014;15(1):1.
28. Gallardo M, Silva A, Rubio L, et al. Incidence of BRCA1 and BRCA2
mutations in 54 Chilean families with breast/ovarian cancer, genotype–
phenotype correlations. Breast Cancer Res Treat. 2006;95(1):81-87.
29. Sanchez A, Faundez P, Carvallo P. Genomic rearrangements of the
BRCA1 gene in Chilean breast cancer families: an MLPA analysis. Breast
Cancer Res Treat. 2011;128(3):845-853.
30.Jara L, Ampuero S, Santibáñez E, et al. BRCA1 and BRCA2
mutations in a South American population. Cancer Genet Cytogenet.
2006;166(1):36-45.
31.Gonzalez-Hormazabal P, Gutierrez-Enriquez S, Gaete D, et al.
Spectrum of BRCA1/2 point mutations and genomic rearrangements in
high-risk breast/ovarian cancer Chilean families. Breast Cancer Res Treat.
2011;126(3):705-716.
32. Torres D, Rashid MU, Seidel-Renkert A, et al. Absence of the
BRCA1 del (exons 9–12) mutation in breast/ovarian cancer families outside
of Mexican Hispanics. Breast Cancer Res Treat. 2009;117(3):679-681.
33. Torres D, Rashid MU, Gil F, et al. High proportion of BRCA1/2
founder mutations in Hispanic breast/ovarian cancer families from Colombia.
Breast Cancer Res Treat. 2006;103(2):225-232.
34. Sanabria MC, Muñoz G, Vargas CI. Mutations in the BRCA1 gene
(185delAG and 5382insC) are not present in any of the 30 breast cancer patients
analyzed from eastern Colombia [in Spanish]. Biomedica. 2009;29(1):61-72.
35. Rodriguez RC, Esperon AA, Ropero R, et al. Prevalence of BRCA1
and BRCA2 mutations in breast cancer patients from Cuba. Familial Cancer.
2008;7(3):275-279.
36. Hernandez J, Llacuachaqui M, Palacio GV, et al. Prevalence of
BRCA1 and BRCA2 mutations in unselected breast cancer patients from
medellín, Colombia. Hered Cancer Clin Pract. 2014;12(11):11.
37. Gutiérrez Espeleta G, Llacuachaqui M, García‐Jiménez L, et al. BRCA1
and BRCA2 mutations among familial breast cancer patients from Costa Rica.
Clin Genet. 2012;82(5):484-488.
38. García-Jiménez L, Gutiérrez-Espeleta G, Narod SA. Descriptive
epidemiology and molecular genetics of hereditary breast cancer in Costa
Rica. Revis Biol Trop. 2012;60(4):1663-1668.
39. Rodríguez AO, Llacuachaqui M, Pardo GG, et al. BRCA1 and BRCA2
mutations among ovarian cancer patients from Colombia. Gynecol Oncol.
2012;124(2):236-243.
40. Ruiz‐Flores P, Sinilnikova OM, Badzioch M, et al. BRCA1 and BRCA2
mutation analysis of early‐onset and familial breast cancer cases in Mexico.
Hum Mutat. 2002;20(6):474-475.
41. Calderón-Garcidueñas AL, Ruiz-Flores P, Cerda-Flores RM, et al. Clinical
follow up of Mexican women with early onset of breast cancer and mutations in
the BRCA1 and BRCA2 genes. Salud Publica Mex. 2005;47(2):110-115.
42. Vaca-Paniagua F, Alvarez-Gomez RM, Fragoso-Ontiveros V, et al.
Full-exon pyrosequencing screening of BRCA germline mutations in Mexican
women with inherited breast and ovarian cancer. PloS One. 2012;7(5):e37432.
43. Vidal-Millan S, Taja-Chayeb L, Gutierrez-Hernandez O, et al. Mutational
analysis of BRCA1 and BRCA2 genes in Mexican breast cancer patients. Eur J
Gynaecol Oncol. 2009;30(5):527-530.
44. Villarreal-Garza C, Weitzel J, Llacuachaqui M, et al. The prevalence
of BRCA1 and BRCA2 mutations among young Mexican women with triplenegative breast cancer. Breast Cancer Res Treat. 2015;150(2):389-394.
45. Villarreal‐Garza C, Alvarez‐Gómez RM, Pérez‐Plasencia C, et al.
Significant clinical impact of recurrent BRCA1 and BRCA2 mutations in
Mexico. Cancer. 2015;121(3):372-378.
46. Nahleh Z, Otoukesh S, Dwivedi AK, et al. Clinical and pathological characteristics of Hispanic BRCA-associated breast cancers in the
American-Mexican border city of El Paso, TX. American journal of cancer
research. 2015;5(1):466.
47. Torres-Mejía G, Royer R, Llacuachaqui M, et al. Recurrent BRCA1 and
BRCA2 mutations in Mexican women with breast cancer. Cancer Epidemi
Biomark Prevent. 2015;24(3):498-505.
48. Abugattas J, Llacuachaqui M, Allende YS, et al. Prevalence of
BRCA1 and BRCA2 mutations in unselected breast cancer patients from
Peru. Clin Genet. 2015;88(4):371-375.
49. Dutil J, Colon-Colon JL, Matta JL, et al. Identification of the prevaOctober 2016, Vol. 23, No. 4
lent BRCA1 and BRCA2 mutations in the female population of Puerto Rico.
Cancer Genet. 2012;205(5):242-248.
50. Delgado La, Fernández G, González A, et al. Hereditary breast cancer
associated with a germline BRCA2 mutation in identical female twins with
similar disease expression. Cancer Genet Cytogenet. 2002;133(1):24-28.
51. Lara K, Consigliere N, Pérez J, et al. BRCA1 and BRCA2 mutations
in breast cancer patients from Venezuela. Biol Res. 2012;45(2):117-130.
52. Solano AR, Aceto GM, Delettieres D, et al. BRCA1 And BRCA2
analysis of Argentinean breast/ovarian cancer patients selected for age
and family history highlights a role for novel mutations of putative southAmerican origin. Springerplus. 2012;1:20.
53. Dufloth RM, Carvalho S, Heinrich JK, et al. Analysis of BRCA1 and
BRCA2 mutations in Brazilian breast cancer patients with positive family
history. Sao Paulo Med J. 2005;123(4):192-197.
54. Carraro DM, Folgueira MA, Lisboa BCG, et al. Comprehensive analysis of BRCA1, BRCA2 and TP53 germline mutation and tumor characterization:
a portrait of early-onset breast cancer in Brazil. PloS One. 2013;8(3):e57581.
55. Porchia LM, Gonzalez Mejia ME, Calderilla-Barbosa L, et al. Common
BRCA1 and BRCA2 butations among Latin American breast cancer subjects: a
meta-analysis. J Carcinogen Mutagen. 2015;2015. http://www.omicsonline.org/
open-access/common-brca1-and-brca2-mutations-among-latin-americanbreast-cancer-subjects-a-metaanalysis-2157-2518-1000228.pdf. Accessed
September 15, 2016.
56. John EM, Miron A, Gong G, et al. Prevalence of pathogenic BRCA1
mutation carriers in 5 US racial/ethnic groups. JAMA. 2007;298(24):2869-2876.
57. Mullineaux LG, Castellano TM, Shaw J, et al. Identification of germline
185delAG BRCA1 mutations in non‐Jewish Americans of Spanish ancestry
from the San Luis Valley, Colorado. Cancer. 2003;98(3):597-602.
58. Weitzel JN, Clague J, Martir-Negron A, et al. Prevalence and type of
BRCA mutations in Hispanics undergoing genetic cancer risk assessment in
the southwestern United States: a report from the Clinical Cancer Genetics
Community Research Network. J Clin Oncol. 2013;31(2):210-216.
59. Laitman Y, Feng B-J, Zamir IM, et al. Haplotype analysis of the
185delAG BRCA1 mutation in ethnically diverse populations. Eur J Hum
Genet. 2013;21(2):212-216.
60. Ah Mew N, Hamel N, Galvez M, et al. Haplotype analysis of a BRCA1:
185delAG mutation in a Chilean family supports its Ashkenazi origins. Clin
Genet. 2002;62(2):151-156.
61. Weitzel JN, Lagos V, Blazer KR, et al. Prevalence of BRCA mutations
and founder effect in high-risk Hispanic families. Cancer Epidemiol Biomark
Prevent. 2005;14(7):1666-1671.
62. Weitzel JN, Lagos VI, Herzog JS, et al. Evidence for common ancestral origin of a recurring BRCA1 genomic rearrangement identified in high-risk
Hispanic families. Cancer Epidemiol Biomark Prevent. 2007;16(8):1615-1620.
63. Judkins T, Rosenthal E, Arnell C, et al. Clinical significance of large
rearrangements in BRCA1 and BRCA2. Cancer. 2012;118(21):5210-5216.
64. Delgado L, Fernández G, Grotiuz G, et al. BRCA1 and BRCA2 germline mutations in Uruguayan breast and breast–ovarian cancer families.
Identification of novel mutations and unclassified variants. Breast Cancer
Res Treat. 2011;128(1):211-218.
65. Esteban Cardeñosa E, Bolufer Gilabert P, de Juan Jimenez I, et al.
Broad BRCA1 and BRCA2 mutational spectrum and high incidence of recurrent
and novel mutations in the eastern Spain population. Breast Cancer Res Treat.
2010;121(1):257-260.
66. Miramar M, Calvo M, Rodriguez A, et al. Genetic analysis of BRCA1
and BRCA2 in breast/ovarian cancer families from Aragon (Spain): two novel
truncating mutations and a large genomic deletion in BRCA1. Breast Cancer
Res Treat. 2008;112(2):353-358.
67. Infante M, Durán M, Esteban-Cardeñosa E, et al. High proportion
of novel mutations of BRCA1 and BRCA2 in breast/ovarian cancer patients
from Castilla-Leon (central Spain). J Hum Genet. 2006;51(7):611-617.
68. de la Hoya M, Osorio A, Godino J, et al. Association between
BRCA1 and BRCA2 mutations and cancer phenotype in Spanish breast/
ovarian cancer families: implications for genetic testing. Int J Cancer.
2002;97(4):466-471.
69. Blay P, Santamaría I, Pitiot AS, et al. Mutational analysis of BRCA1
and BRCA2 in hereditary breast and ovarian cancer families from Asturias
(Northern Spain). BMC Cancer. 2013;13(1):1.
70. Bolufer P, Munárriz B, Santaballa A, et al. BRCA1 and BRCA2
mutations in patients with familial breast cancer [In Spanish]. Med Clin.
2005;124(1):10-12.
71. de Juan Jiménez I, Casado ZG, Suela SP, et al. Novel and recurrent
BRCA1/BRCA2 mutations in early onset and familial breast and ovarian
cancer detected in the Program of Genetic Counseling in Cancer of Valencian
Community (eastern Spain). Relationship of family phenotypes with mutation
prevalence. Fam Cancer. 2013;12(4):767-777.
72. González-Hormazábal P, Bravo T, Blanco R, et al. Association of
common ATM variants with familial breast cancer in a South American
population. BMC Cancer. 2008;8(1):1.
73. Leyton Y, Gonzalez-Hormazabal P, Blanco R, et al. Association of
PALB2 sequence variants with the risk of familial and early-onset breast
cancer in a South-American population. BMC Cancer. 2015;15(1):1.
74. Calderón-Zúñiga Fdel C, Ocampo-Gomez G, Lopez-Marquez FC, et
al. ATM polymorphisms IVS24-9delT, IVS38-8T>C, and 5557G>A in Mexican
October 2016, Vol. 23, No. 4
women with familial and/or early-onset breast cancer. Salud Publica Mex.
2014;56(2):206-212.
75. Bretsky P, Haiman CA, Gilad S, et al. The relationship between
twenty missense ATM variants andb cancer risk the multiethnic cohort.
Cancer Epidemiol Biomark Prevent. 2003;12(8):733-738.
76.Bell DW, Kim SH, Godwin AK, et al. Genetic and functional
analysis of CHEK2 (CHK2) variants in multiethnic cohorts. Int J Cancer.
2007;121(12):2661-2667.
77. Damiola F, Pertesi M, Oliver J, et al. Rare key functional domain
missense substitutions in MRE11A, RAD50, and NBN contribute to breast
cancer susceptibility: results from a breast cancer family registry casecontrol mutation-screening study. Breast Cancer Res. 2014;16(3):R58.
78. Assumpção JG, Seidinger AL, Mastellaro MJ, et al. Association of
the germline TP53 R337H mutation with breast cancer in southern Brazil.
BMC Cancer. 2008;8(1):1.
79. Giacomazzi J, Graudenz MS, Osorio CA, et al. Prevalence of the
TP53 p. R337H mutation in breast cancer patients in Brazil. PloS One.
2014;9(6):e99893.
80. Jara L, Acevedo ML, Blanco R, et al. RAD51 135G> C polymorphism and risk of familial breast cancer in a South American population.
Cancer Genet Cytogenet. 2007;178(1):65-69.
81. Gonzalez KD, Noltner KA, Buzin CH, et al. Beyond Li Fraumeni
Syndrome: clinical characteristics of families with p53 germline mutations.
J Clin Ocol. 2009;27(8):1250-1256.
82. Olivier M, Goldgar DE, Sodha N, et al. Li-Fraumeni and related syndromes correlation between tumor type, family structure, and TP53 genotype. Cancer Res. 2003;63(20):6643-6650.
83. Li FP, Fraumeni JF Jr, Mulvihill JJ, et al. A cancer family syndrome
in twenty-four kindreds. Cancer Res. 1988;48(18):5358-5362.
84. Børresen A-L, Andersen TI, Garber J, et al. Screening for germ line
TP53 mutations in breast cancer patients. Cancer Res. 1992;52(11):3234-3236.
85. Sidransky D, Tokino T, Helzlsouer K, et al. Inherited p53 gene mutations in breast cancer. Cancer research. 1992;52(10):2984-2986.
86. Lalloo F, Varley J, Moran A, et al. BRCA1, BRCA2 and TP53 mutations in very early-onset breast cancer with associated risks to relatives.
Eur J Cancer. 2006;42(8):1143-1150.
87. Ginsburg OM, Akbari MR, Aziz Z, et al. The prevalence of germ-line
TP53 mutations in women diagnosed with breast cancer before age 30.
Fam Cancer. 2009;8(4):563-567.
88. McCuaig JM, Armel SR, Novokmet A, et al. Routine TP53 testing for breast cancer under age 30: ready for prime time? Fam Cancer.
2012;11(4):607-613.
89. Ribeiro RC, Sandrini F, Figueiredo B, et al. An inherited p53 mutation that contributes in a tissue-specific manner to pediatric adrenal cortical
carcinoma. Proceed Natl Acad Sci. 2001;98(16):9330-9335.
90. Gomes M, Kotsopoulos J, de Almeida GL, et al. The R337H mutation
in TP53 and breast cancer in Brazil. Hered Cancer Clin Pract. 2012;10(1):3.
91. Custódio G, Parise GA, Kiesel Filho N, et al. Impact of neonatal
screening and surveillance for the TP53 R337H mutation on early detection
of childhood adrenocortical tumors. J Clin Oncol. 2013;31(20):2619-2626.
92. Palmero EI, Schüler-Faccini L, Caleffi M, et al. Detection of R337H,
a germline TP53 mutation predisposing to multiple cancers, in asymptomatic women participating in a breast cancer screening program in southern
Brazil. Cancer Lett. 2008;261(1):21-25.
93. Garritano S, Gemignani F, Palmero EI, et al. Detailed haplotype
analysis at the TP53 locus in p. R337H mutation carriers in the population of
southern Brazil: evidence for a founder effect. Hum Mutat. 2010;31(2):143-150.
94.Blazer KR, Nehoray B, Solomon I, et al. Next-generation testing for cancer risk: perceptions, experiences, and needs among early
adopters in community healthcare settings. Genet Test Molec Biomark.
2015;19(12):657-665.
95. Maxwell KN, Wubbenhorst B, D’Andrea K, et al. Prevalence of mutations in a panel of breast cancer susceptibility genes in BRCA1/2-negative
patients with early-onset breast cancer. Genet Med. 2014;17(8):630-638.
96. Tung N, Battelli C, Allen B, et al. Frequency of mutations in individuals
with breast cancer referred for BRCA1 and BRCA2 testing using next‐
generation sequencing with a 25‐gene panel. Cancer. 2015;121(1):25-33.
97. Kapoor NS, Curcio LD, Blakemore CA, et al. Multigene panel testing detects equal rates of pathogenic BRCA1/2 mutations and has a higher
diagnostic yield compared to limited BRCA1/2 analysis alone in patients at
risk for hereditary breast cancer. Ann Surg Oncol. 2015;22(10):3282-3288.
98. Li J, Meeks H, Feng B-J, et al. Targeted massively parallel sequencing
of a panel of putative breast cancer susceptibility genes in a large cohort of
multiple-case breast and ovarian cancer families. J Med Genet. 2016;53(1):34-42.
99. Couch FJ, Hart SN, Sharma P, et al. Inherited mutations in 17 breast
cancer susceptibility genes among a large triple-negative breast cancer cohort
unselected for family history of breast cancer. J Clin Oncol. 2015;33(4):304-311.
100. Desmond A, Kurian AW, Gabree M, et al. Clinical actionability of multigene panel testing for hereditary breast and ovarian cancer risk assessment.
JAMA Oncol. 2015;1(7):943-951.
101. Doherty J, Bonadies DC, Matloff ET. Testing for hereditary breast
cancer: panel or targeted testing? Experience from a clinical cancer genetics
practice. J Genet Counsel. 2015;24(4):683-687.
102. Kurian AW, Hare EE, Mills MA, et al. Clinical evaluation of a multiCancer Control 369
ple-gene sequencing panel for hereditary cancer risk assessment. J Clin
Oncol. 2014;32(19):2001-2009.
103. Lincoln SE, Kobayashi Y, Anderson MJ, et al. A systematic comparison
of traditional and multigene panel testing for hereditary breast and ovarian
cancer genes in more than 1000 patients. J Molec Diagnost. 2015;17(5):533-544.
104. Ricker C, Culver JO, Lowstuter K, et al. Increased yield of actionable
mutations using multi-gene panels to assess hereditary cancer susceptibility
in an ethnically diverse clinical cohort. Cancer Genet. 2016; 209(4):130-137.
105. Mai PL, Vadaparampil ST, Breen N, et al. Awareness of cancer
susceptibility genetic testing: the 2000, 2005, and 2010 National Health
Interview Surveys. Am J Prevent Med. 2014;46(5):440-448.
106. Vadaparampil ST, Wideroff L, Breen N, et al. The impact of acculturation
on awareness of genetic testing for increased cancer risk among Hispanics
in the year 2000 National Health Interview Survey. Cancer Epidemiol Biomark
Prevent. 2006;15(4):618-623.
107. Wideroff L, Thomas Vadaparampil S, Breen N, et al. Awareness of
genetic testing for increased cancer risk in the year 2000 National Health
Interview Survey. Pub Health Genom. 2004;6(3):147-156.
108. Gammon AD, Rothwell E, Simmons R, et al. Awareness and preferences regarding BRCA1/2 genetic counseling and testing among Latinas
and non-Latina white women at increased risk for hereditary breast and
ovarian cancer. J Genet Counsel. 2011;20(6):625-638.
109. Vadaparampil ST, McIntyre J, Quinn GP. Awareness, perceptions,
and provider recommendation related to genetic testing for hereditary breast
cancer risk among at-risk Hispanic women: similarities and variations by
sub-ethnicity. J Genet Counsel. 2010;19(6):618-629.
110. Kaplan CP, Haas JS, Pérez-Stable EJ, et al. Breast cancer risk reduction options: awareness, discussion, and use among women from four
ethnic groups. Cancer Epidemiol Biomark Prevent. 2006;15(1):162-166.
111. Mays D, Sharff ME, DeMarco TA, et al. Outcomes of a systemslevel intervention offering breast cancer risk assessments to low-income
underserved women. Fam Cancer. 2012;11(3):493-502.
112. Jagsi R, Griffith KA, Kurian AW, et al. Concerns about cancer risk
and experiences with genetic testing in a diverse population of patients
with breast cancer. J Clin Oncol. 2015;33(14):1584-1591.
113. Lagos VI, Perez MA, Ricker CN, et al. Social‐cognitive aspects of
underserved Latinas preparing to undergo genetic cancer risk assessment for
hereditary breast and ovarian cancer. Psycho‐Oncology. 2008;17(8):774-782.
114. Vadaparampil ST, Quinn GP, Dutil J, et al. A pilot study of knowledge and interest of genetic counseling and testing for hereditary breast
and ovarian cancer syndrome among Puerto Rican women. J Comm Genet.
2011;2(4):211-221.
115. Ricker C, Lagos V, Feldman N, et al. If we build it… will they come?–
establishing a cancer genetics services clinic for an underserved predominantly
Latina cohort. J Genet Counsel. 2006;15(6):505-514.
116. Komenaka IK, Nodora JN, Madlensky L, et al. Participation of lowincome women in genetic cancer risk assessment and BRCA 1/2 testing:
the experience of a safety-net institution. J Comm Genet. 2015:1-7.
117. Olaya W, Esquivel P, Wong JH, et al. Disparities in BRCA testing:
when insurance coverage is not a barrier. Am J Surg. 2009;198(4):562-565.
118. Woodson AH, Profato JL, Rizvi SH, et al. Service delivery model
and experiences in a cancer genetics clinic for an underserved population.
J Health Care Poor Underserv. 2015;26(3):784-791.
119. Macconaill LE, Garraway LA. Clinical implications of the cancer
genome. J Clin Oncol. 2010;28(35):5219-5228.
120. Easton DF, Pooley KA, Dunning AM, et al. Genome-wide association study identifies novel breast cancer susceptibility loci. Nature.
2007;447(7148):1087-1093.
121. Stacey SN, Manolescu A, Sulem P, et al. Common variants on chromosome 5p12 confer susceptibility to estrogen receptor–positive breast
cancer. Nat Genet. 2008;40(6):703-706.
122. Hunter DJ, Kraft P, Jacobs KB, et al. A genome-wide association
study identifies alleles in FGFR2 associated with risk of sporadic postmenopausal breast cancer. Nat Genet. 2007;39(7):870-874.
123. Garcia-Closas M, Couch FJ, Lindstrom S, et al. Genome-wide
association studies identify four ER negative-specific breast cancer risk loci.
Nat Genet. 2013;45(4):392-398.
124. Fletcher O, Johnson N, Orr N, et al. Novel breast cancer susceptibility locus at 9q31. 2: results of a genome-wide association study. J Natl
Cancer Inst. 2011;103(5):425-435.
125. Gold B, Kirchhoff T, Stefanov S, et al. Genome-wide association
study provides evidence for a breast cancer risk locus at 6q22. 33. Proceed
Natl Acad Sci. 2008;105(11):4340-4345.
126. Turnbull C, Ahmed S, Morrison J, et al. Genome-wide association study identifies five new breast cancer susceptibility loci. Nat Genet.
2010;42(6):504-507.
127. Gaudet MM, Kuchenbaecker KB, Vijai J, et al. Identification of a
BRCA2-specific modifier locus at 6p24 related to breast cancer risk. PLoS
Genet. 2013;9(3):e1003173.
128. Long J, Cai Q, Sung H, et al. Genome-wide association study in
east Asians identifies novel susceptibility loci for breast cancer. PLoS
Genet. 2012;8(2):e1002532.
129. Couch FJ, Wang X, McGuffog L, et al. Genome-wide association
study in BRCA1 mutation carriers identifies novel loci associated with
370 Cancer Control
breast and ovarian cancer risk. PLoS Genet. 2013;9(3):e1003212.
130. Li J, Humphreys K, Darabi H, et al. A genome-wide association
scan on estrogen receptor-negative breast cancer. Breast Cancer Res.
2010;12(6):R93.
131. Murabito JM, Rosenberg CL, Finger D, et al. A genome-wide
association study of breast and prostate cancer in the NHLBI’s Framingham
Heart Study. BMC Med Genet. 2007;8(1):1.
132. Antoniou AC, Wang X, Fredericksen ZS, et al. A locus on 19p13
modifies risk of breast cancer in BRCA1 mutation carriers and is associated
with hormone receptor-negative breast cancer in the general population.
Nat Genet. 2010;42(10):885-892.
133. Cai Q, Long J, Lu W, et al. Genome-wide association study identifies
breast cancer risk variant at 10q21. 2: results from the Asia Breast Cancer
Consortium. Hum Molec Genet. 2011;20(24):4991-4999.
134. Kim H-c, Lee J-Y, Sung H, et al. A genome-wide association study
identifies a breast cancer risk variant in ERBB4 at 2q34: results from the
Seoul Breast Cancer Study. Breast Cancer Res. 2012;14(2):1-12.
135. Elgazzar S, Zembutsu H, Takahashi A, et al. A genome-wide association study identifies a genetic variant in the SIAH2 locus associated
with hormonal receptor-positive breast cancer in Japanese. J Hum Genet.
2012;57(12):766-771.
136. Long J, Cai Q, Shu X-O, et al. Identification of a functional genetic
variant at 16q12. 1 for breast cancer risk: results from the Asia Breast Cancer
Consortium. PLoS Genet. 2010;6(6):e1001002.
137. Michailidou K, Beesley J, Lindstrom S, et al. Genome-wide association analysis of more than 120,000 individuals identifies 15 new susceptibility loci for breast cancer. Nat Genet. 2015;47(4):373-380.
138. Thomas G, Jacobs KB, Kraft P, et al. A multistage genome-wide
association study in breast cancer identifies two new risk alleles at 1p11. 2
and 14q24. 1 (RAD51L1). Nat Genet. 2009;41(5):579-584.
139. Siddiq A, Couch FJ, Chen GK, et al. A meta-analysis of genomewide association studies of breast cancer identifies two novel susceptibility
loci at 6q14 and 20q11. Hum Molec Genet. 2012;21(24):5373-5384.
140. Michailidou K, Hall P, Gonzalez-Neira A, et al. Large-scale genotyping identifies 41 new loci associated with breast cancer risk. Nat Genet.
2013;45(4):353-361.
141.Chen F, Chen GK, Stram DO, et al. A genome-wide association study of breast cancer in women of African ancestry. Hum Genet.
2013;132(1):39-48.
142. Haiman CA, Chen GK, Vachon CM, et al. A common variant at
the TERT-CLPTM1L locus is associated with estrogen receptor-negative
breast cancer. Nat Genet. 2011;43(12):1210-1214.
143. Zheng W, Long J, Gao Y-T, et al. Genome-wide association study
identifies a new breast cancer susceptibility locus at 6q25. 1. Nat Genet.
2009;41(3):324-328.
144. Fejerman L, Ahmadiyeh N, Hu D, et al. Genome-wide association
study of breast cancer in Latinas identifies novel protective variants on 6q25.
Nat Comm. 2014;5.
145. Haiman CA, Stram DO, Pike MC, et al. A comprehensive haplotype
analysis of CYP19 and breast cancer risk: the multiethnic cohort. Hum
Molec Genet. 2003;12(20):2679-2692.
146. Cheng I, Penney KL, Stram DO, et al. Haplotype-based association
studies of IGFBP1 and IGFBP3 with prostate and breast cancer risk: the multiethnic cohort. Cancer Epidemiol Biomark Prevent. 2006;15(10):1993-1997.
147. Women’s Health Initiative Study Group. Design of the Women’s
Health Initiative clinical trial and observational study. Control Clin Trials.
1998;19(1):61-109.
148. Hays J, Hunt JR, Hubbell FA, et al. The Women’s Health Initiative
recruitment methods and results. Ann Epidemiol. 2003;13(9):S18-S77.
149. Slattery ML, John EM, Torres-Mejia G, et al. Genetic variation in genes
involved in hormones, inflammation and energetic factors and breast cancer
risk in an admixed population. Carcinogenesis. 2012;33(8):1512-1521.
150. Ramírez-Patiño R, Figuera LE, Puebla-Pérez AM, et al. Intron 4
VNTR (4a/b) polymorphism of the endothelial nitric oxide synthase gene
is associated with breast cancer in Mexican women. J Korean Med Sci.
2013;28(11):1587-1594.
151. Quintero-Ramos A, Gutiérrez-Rubio S, Del Toro-Arreola A, et al.
Association between polymorphisms in the thymidylate synthase gene and risk
of breast cancer in a Mexican population. Genet Mol Res. 2014;13(4):8749-8756.
152. Jara L, Dubois K, Gaete D, et al. Variants in DNA double-strand break
repair genes and risk of familial breast cancer in a South American population.
Breast Cancer Res Treat. 2010;122(3):813-822.
153. Oliveira CB, Cardoso-Filho C, Bossi LS, et al. Association of CYP1A1
A4889G and T6235C polymorphisms with the risk of sporadic breast cancer
in Brazilian women. Clinics. 2015;70(10):680-685.
154. Pérez‐Mayoral J, Pacheco‐Torres AL, Morales L, et al. Genetic polymorphisms in RAD23B and XPC modulate DNA repair capacity and breast cancer
risk in Puerto Rican women. Molec Carcinogen. 2013;52(suppl 1):127-138.
155. Kolonel LN, Henderson BE, Hankin JH, et al. A multiethnic cohort
in Hawaii and Los Angeles: baseline characteristics. Am J Epidemiol.
2000;151(4):346-357.
156. Kolonel LN, Altshuler D, Henderson BE. The multiethnic cohort study:
exploring genes, lifestyle and cancer risk. Nat Rev Cancer. 2004;4(7):519-527.
157. John EM, Phipps AI, Davis A, et al. Migration history, acculturation,
October 2016, Vol. 23, No. 4
and breast cancer risk in Hispanic women. Cancer Epidemiol Biomark
Prevent. 2005;14(12):2905-2913.
158. John EM, Horn-Ross PL, Koo J. Lifetime physical activity and breast
cancer risk in a multiethnic population the san francisco Bay area breast
cancer study. Cancer Epidemiol Biomark Prevent. 2003;12(11):1143-1152.
159. John EM, Hopper JL, Beck JC, et al. The breast cancer family registry: an infrastructure for cooperative multinational, interdisciplinary and
translational studies of the genetic epidemiology of breast cancer. Breast
Cancer Res. 2004;6(4):R375-R389.
160. Slattery ML, Sweeney C, Edwards S, et al. Body size, weight change,
fat distribution and breast cancer risk in Hispanic and non-Hispanic white
women. Breast Cancer Res Treat. 2007;102(1):85-101.
161. Slattery ML, Sweeney C, Herrick J, et al. ESR1, AR, body size, and
breast cancer risk in Hispanic and non-Hispanic white women living in the
southwestern United States. Breast Cancer Res Treat. 2007;105(3):327-335.
162. Gallegos-Arreola MP, Figuera LE, Flores-Ramos LG, et al. Association of the Alu insertion polymorphism in the progesterone receptor
gene with breast cancer in a Mexican population. Arch Med Science.
2015;11(3):551.
163. Haiman CA, Garcia RR, Hsu C, et al. Screening and association
testing of common coding variation in steroid hormone receptor co-activator
and co-repressor genes in relation to breast cancer risk: the multiethnic
cohort. BMC Cancer. 2009;9(1):1.
164. Boone SD, Baumgartner KB, Baumgartner RN, et al. Associations
between CYP19A1 polymorphisms, Native American ancestry, and breast
cancer risk and mortality: the Breast Cancer Health Disparities Study.
Cancer Cause Control. 2014;25(11):1461-1471.
165. Moreno-Galván M, Herrera-González NE, Robles-Pérez V, et al.
Impact of CYP1A1 and COMT genotypes on breast cancer risk in Mexican
women: a pilot study. Int J Biol Mark. 2009;25(3):157-163.
166. Weston A, Pan C, Bleiweiss IJ, et al. CYP17 genotype and breast
cancer risk. Cancer Epidemiol Biomark Prevent. 1998;7(10):941-944.
167. Sarkissyan M, Mishra DK, Wu Y, et al. IGF gene polymorphisms
and breast cancer in African-American and Hispanic women. Int J Oncol.
2011;38(6):1663-1673.
168. Slattery ML, John EM, Stern MC, et al. Associations with growth
factor genes (FGF1, FGF2, PDGFB, FGFR2, NRG2, EGF, ERBB2) with
breast cancer risk and survival: the Breast Cancer Health Disparities study.
Breast Cancer Res Treat. 2013;140(3):587-601.
169. Connor AE, Baumgartner RN, Baumgartner KB, et al. Epidermal
growth factor receptor (EGFR) polymorphisms and breast cancer among
Hispanic and non-Hispanic white women: the Breast Cancer Health Disparities study. Int J Molec Epidemiol Genet. 2013;4(4):235.
170. Boone SD, Baumgartner KB, Baumgartner RN, et al. Associations
between genetic variants in the TGF-β signaling pathway and breast cancer risk among Hispanic and non-Hispanic white women. Breast Cancer
Res Treat. 2013;141(2):287-297.
171. Slattery ML, John EM, Torres‐Mejia G, et al. Genetic variation in
bone morphogenetic proteins and breast cancer risk in hispanic and non‐
hispanic white women: The breast cancer health disparities study. Int J
Cancer. 2013;132(12):2928-2939.
172. Ingles SA, Garcia DG, Wang W, et al. Vitamin D receptor genotype and breast cancer in Latinas (United States). Cancer Cause Control.
2000;11(1):25-30.
173. Connor AE, Baumgartner RN, Baumgartner KB, et al. Associations
between TCF7L2 polymorphisms and risk of breast cancer among Hispanic
and non-Hispanic White women: the Breast Cancer Health Disparities
study. Breast Cancer Res Treat. 2012;136(2):593-602.
174. Slattery ML, John E, Torres-Mejia G, et al. Matrix metalloproteinase
genes are associated with breast cancer risk and survival: the Breast Cancer
Health Disparities study. PloS One. 2013;8(5):e63165.
175. Delgado-Enciso I, Cepeda-Lopez FR, Monrroy-Guizar EA, et al.
Matrix metalloproteinase-2 promoter polymorphism is associated with breast
cancer in a Mexican population. Gynecol Obstet Invest. 2008;65(1):68-72.
176. Slattery ML, Herrick JS, Torres-Mejia G, et al. Genetic variants in
interleukin genes are associated with breast cancer risk and survival in
a genetically admixed population: the Breast Cancer Health Disparities
study. Carcinogenesis. 2014:bgu078.
177. Slattery ML, Lundgreen A, Hines L, et al. Energy homeostasis
genes and breast cancer risk: The influence of ancestry, body size, and
menopausal status, the breast cancer health disparities study. Cancer
Epidemiol. 2015;39(6):1113-1122.
178. Connor A, Baumgartner RN, Kerber RA, et al. ADRB2 G–G haplotype associated with breast cancer risk among Hispanic and non-Hispanic
white women: interaction with type 2 diabetes and obesity. Cancer Cause
Control. 2012;23(10):1653-1663.
179. Connor AE, Baumgartner RN, Baumgartner KB, et al. Associations
between ALOX, COX, and CRP polymorphisms and breast cancer among
Hispanic and non‐Hispanic white women: the breast cancer health disparities study. Molec Carcinogen. 2015;54(12):1541-1553.
180. Kaklamani VG, Hoffmann TJ, Thornton TA, et al. Adiponectin pathway polymorphisms and risk of breast cancer in African Americans and
Hispanics in the Women’s Health Initiative. Breast Cancer Res Treat.
2013;139(2):461-468.
October 2016, Vol. 23, No. 4
181. Flores-Ramos LG, Escoto-De Dios A, Puebla-Pérez A, et al. Association of the tumor necrosis factor-alpha-308G>: a polymorphism with breast
cancer in Mexican women. Genet Mol Res. 2013;12:5680-5693.
182. Pellatt AJ, Wolff RK, John EM, et al. SEPP1 influences breast cancer
risk among women with greater native american ancestry: the breast cancer
health disparities study. PLoS One. 2013;8(11):e80554.
183. Baumgartner KB, Schlierf TJ, Yang D, et al. N-acetyltransferase 2
genotype modification of active cigarette smoking on breast cancer risk
among hispanic and non-hispanic white women. Toxicol Sci. 2009:kfp199.
184. Cardenas-Rodriguez N, Lara-Padilla E, Bandala C, et al. CYP2W1,
CYP4F11 and CYP8A1 polymorphisms and interaction of CYP2W1 genotypes with risk factors in Mexican women with breast cancer. Asian Pac J
Cancer Prevent. 2012;13(3):837-846.
185. Gutierrez-Rubio S, Quintero-Ramos A, Duran-Cardenas A, et al.
1236 C/T and 3435 C/T polymorphisms of the ABCB1 gene in Mexican
breast cancer patients. Genet Mol Res. 2015;14(1):1250-1259.
186. Ramos-Silva A, Figuera L, Soto-Quintana O, et al. Association of the
C677T polymorphism in the methylenetetrahydrofolate reductase gene with
breast cancer in a Mexican population. Genet Mol Res. 2015;14(2):4015.
187. Slattery ML, John EM, Torres‐Mejia G, et al. Angiogenesis genes,
dietary oxidative balance and breast cancer risk and progression: the Breast
Cancer Health Disparities study. Int J Cancer. 2014;134(3):629-644.
188. Tsai H-J, Choudhry S, Naqvi M, et al. Comparison of three methods to
estimate genetic ancestry and control for stratification in genetic association
studies among admixed populations. Hum Genet. 2005;118(3-4):424-433.
189. Tang H, Quertermous T, Rodriguez B, et al. Genetic structure, selfidentified race/ethnicity, and confounding in case-control association studies.
Am J Hum Genet. 2005;76(2):268-275.
190. Slattery ML, Baumgartner KB, Giuliano AR, et al. Replication of five
GWAS-identified loci and breast cancer risk among Hispanic and nonHispanic white women living in the southwestern United States. Breast Cancer
Res Treat. 2011;129(2):531-539.
191. Fejerman L, Stern MC, John EM, et al. Interaction between common breast cancer susceptibility variants, genetic ancestry, and nongenetic risk factors in Hispanic women. Cancer Epidemiol Biomark Prevent.
2015;24(11):1731-1738.
192. Fejerman L, Stern MC, Ziv E, et al. Genetic ancestry modifies the
association between genetic risk variants and breast cancer risk among
Hispanic and non-Hispanic white women. Carcinogenesis. 2013;34(8):1787-1793.
193. Murillo-Zamora E, Moreno-Macías H, Ziv E, et al. Association between
rs2981582 polymorphism in the FGFR2 gene and the risk of breast cancer in
Mexican women. Arch Med Res. 2013;44(6):459-466.
194. Jara L, Gonzalez-Hormazabal P, Cerceño K, et al. Genetic variants in
FGFR2 and MAP3K1 are associated with the risk of familial and early-onset
breast cancer in a South-American population. Breast Cancer Res Treat.
2013;137(2):559-569.
195. Elematore I, Gonzalez-Hormazabal P, Reyes JM, et al. Association of
genetic variants at TOX3, 2q35 and 8q24 with the risk of familial and earlyonset breast cancer in a South-American population. Molec Biol Rep.
2014;41(6):3715-3722.
196. Cheng C-Y, Kao WL, Patterson N, et al. Admixture mapping of
15,280 African Americans identifies obesity susceptibility loci on chromosomes 5 and X. PLoS Genet. 2009;5(5):e1000490.
197. Jia L, Landan G, Pomerantz M, et al. Functional enhancers at the
gene-poor 8q24 cancer-linked locus. PLoS Genet. 2009;5(8):e1000597.
198. Reich D, Nalls MA, Kao WL, et al. Reduced neutrophil count in
people of African descent is due to a regulatory variant in the Duffy antigen
receptor for chemokines gene. PLoS Genet. 2009;5(1):e1000360.
199. Nalls MA, Wilson JG, Patterson NJ, et al. Admixture mapping of white
cell count: genetic locus responsible for lower white blood cell count in the
Health ABC and Jackson Heart studies. Am J Hum Genet. 2008;82(1):81-87.
200.Pritchard JK, Stephens M, Donnelly P. Inference of population
structure using multilocus genotype data. Genetics. 2000;155(2):945-959.
201. Reich D, Patterson N, Ramesh V, et al. Admixture mapping of an
allele affecting interleukin 6 soluble receptor and interleukin 6 levels. Am J
Hum Genet. 2007;80(4):716-726.
202. Winkler CA, Nelson GW, Smith MW. Admixture mapping comes of age.
Ann Rev Genom Hum Genet. 2010;11:65-89.
203. Deo RC, Patterson N, Tandon A, et al. A high-density admixture scan
in 1,670 African Americans with hypertension. PLoS Genet. 2007;3(11):e196.
204. Freedman ML, Haiman CA, Patterson N, et al. Admixture mapping
identifies 8q24 as a prostate cancer risk locus in African-American men.
Proceed Natl Acad Sci. 2006;103(38):14068-14073.
205. Jemal A, Siegel R, Xu J, et al. Cancer statistics, 2010. CA Cancer J
Clin. 2010;60(5):277-300.
206. Fejerman L, John EM, Huntsman S, et al. Genetic ancestry and risk
of breast cancer among US Latinas. Cancer Res. 2008;68(23):9723-9728.
207. Fejerman L, Romieu I, John EM, et al. European ancestry is positively associated with breast cancer risk in Mexican women. Cancer
Epidemiol Biomark Prevent. 2010;19(4):1074-1082.
208. Fejerman L, Chen GK, Eng C, et al. Admixture mapping identifies a
locus on 6q25 associated with breast cancer risk in US Latinas. Hum Molec
Genet. 2012;21(8):1907-1917.
209. Hines LM, Risendal B, Byers T, et al. Ethnic disparities in breast tumor
Cancer Control 371
phenotypic subtypes in Hispanic and non-Hispanic white women. J Women
Health. 2011;20(10):1543-1550.
210. Li CI, Malone KE, Daling JR. Differences in breast cancer stage, treatment, and survival by race and ethnicity. Arch Intern Med. 2003;163(1):49-56.
211. Chlebowski RT, Chen Z, Anderson GL, et al. Ethnicity and breast
cancer: factors influencing differences in incidence and outcome. J Natl
Cancer Inst. 2005;97(6):439-448.
212. Dunnwald LK, Rossing MA, Li CI. Hormone receptor status, tumor
characteristics, and prognosis: a prospective cohort of breast cancer
patients. Breast Cancer Res. 2007;9(1):R6.
213. Hausauer AK, Keegan T, Chang ET, et al. Recent breast cancer
trends among Asian/Pacific Islander, Hispanic, and African-American women
in the US: changes by tumor subtype. Breast Cancer Res. 2007;9(6):R90.
214. Parise CA, Bauer KR, Brown MM, et al. Breast cancer subtypes as
defined by the estrogen receptor (ER), progesterone receptor (PR), and the
human epidermal growth factor receptor 2 (HER2) among women with invasive
breast cancer in California, 1999–2004. Breast J. 2009;15(6):593-602.
215. Carvalho FM, Bacchi LM, Pincerato KM, et al. Geographic differences in the distribution of molecular subtypes of breast cancer in Brazil.
BMC Women Health. 2014;14(1):1.
216. Howlader N, Altekruse SF, Li CI, et al. US incidence of breast cancer subtypes defined by joint hormone receptor and HER2 status. J Natl
Cancer Inst. 2014;106(5):dju055.
217. Swain SM, Baselga J, Kim S-B, et al. Pertuzumab, trastuzumab,
and docetaxel in HER2-positive metastatic breast cancer. N Engl J Med.
2015;372(8):724-734.
218. Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus adjuvant
chemotherapy for operable HER2-positive breast cancer. N Engl J Med.
2005;353(16):1673-1684.
219. Harris LN, Ismaila N, McShane LM, et al; American Society of Clinical
Oncology Clinical Practice Guideline. Use of biomarkers to guide decisions on
adjuvant systemic therapy for women with early-stage invasive breast cancer.
J Clin Oncol. 2016;34(10):1134-1150.
220. Senkus E, Kyriakides S, Penault-Llorca F, et al. Primary breast
cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and
follow-up. Ann Oncol. 2013;24 Suppl 6:vi7-23.
221.National Comprehensive Cancer Network (NCCN). NCCN Clinical
Pratice Guidelines in Oncology: Breast Cancer. v2.2016. https://www.nccn.org/
professionals/physician_gls/pdf/breast.pdf. Accessed September 15, 2016.
222. Kalinsky K, Lim EA, Andreopoulou E, et al. Increased expression of
tumor proliferation genes in Hispanic women with early-stage breast cancer.
Cancer Invest. 2014;32(9):439-444.
223. Eiermann W, Rezai M, Kümmel S, et al. The 21-gene recurrence
score assay impacts adjuvant therapy recommendations for ER-positive,
node-negative and node-positive early breast cancer resulting in a riskadapted change in chemotherapy use. Annals of oncology. 2012:mds512.
224. Partin JF, Mamounas EP. Impact of the 21-gene recurrence score
assay compared with standard clinicopathologic guidelines in adjuvant
therapy selection for node-negative, estrogen receptor-positive breast
cancer. Ann Surg Oncol. 2011;18(12):3399-3406.
225. Geffen D, Abu-Ghanem S, Sion-Vardy N, et al. The impact of the
21-gene recurrence score assay on decision making about adjuvant chemotherapy in early-stage estrogen-receptor-positive breast cancer in an
oncology practice with a unified treatment policy. Ann Oncol. 2011:mdq769.
226. Lo SS, Mumby PB, Norton J, et al. Prospective multicenter study
of the impact of the 21-gene recurrence score assay on medical oncologist and patient adjuvant breast cancer treatment selection. J Clin Oncol.
2010;28(10):1671-1676.
227. Bargallo JE, Lara F, Shaw‐Dulin R, et al. A study of the impact of
the 21‐gene breast cancer assay on the use of adjuvant chemotherapy
in women with breast cancer in a Mexican public hospital. J Surg Oncol.
2015;111(2):203-207.
228. Bargalló-Rocha JE, Lara-Medina F, Pérez-Sánchez V, et al. Costeffectiveness of the 21-gene breast cancer assay in Mexico. Adv Ther.
2015;32(3):239-253.
229. Kalow W, Gunn DR. Some statistical data on atypical cholinesterase of human serum. Ann Hum Genet. 1959;23(3):239-250.
230. Kalow W, Staron N. On distribution and inheritance of atypical
forms of human serum cholinesterase, as indicated by dibucaine numbers.
CA J Biochem Physiol. 1957;35(12):1305-1320.
231. Evans DAP, Manley KA, McKusick VA. Genetic control of isoniazid
metabolism in man. Br Med J. 1960;2(5197):485.
232. Li J, Bluth MH. Pharmacogenomics of drug metabolizing enzymes
and transporters: implications for cancer therapy. Pharmgenomics Pers
Med. 2011;4:11-33.
233.O’Donnell, Dolan ME. Cancer pharmacoethnicity: ethnic differences in susceptibility to the effects of chemotherapy. Clin Cancer Res.
2009;15(15):4806-4814.
234. Grabinski J, Smith L, Chisholm G, et al. Relationship between
CYP2D6 and estrogen receptor alpha polymorphisms on tamoxifen metabolism in adjuvant breast cancer treatment. ASCO Ann Meet Proceed.
2006;24(18 suppl):505.
235. Alcazar-González GA, Calderón-Garcidueñas AL, Garza-Rodríguez
ML, et al. Comparative study of polymorphism frequencies of the CYP2D6,
372 Cancer Control
CYP3A5, CYP2C8 and IL-10 genes in Mexican and Spanish women with
breast cancer. Pharmacogenomics. 2013;14(13):1583-1592.
236. González-Tejera G, Gaedigk A, Corey S. Genetic variants of the
drug-metabolizing enzyme CYP2D6 in Puerto Rican psychiatry patients:
a preliminary report and potential implications for breast cancer patients.
P R Health Sci J. 2010;29(3):299-304.
237. Bernard S, Neville KA, Nguyen AT, et al. Interethnic differences in
genetic polymorphisms of CYP2D6 in the US population: clinical implications.
Oncologist. 2006;11(2):126-135.
238. Cuzick J, Sestak I, Baum M, et al. Effect of anastrozole and tamoxifen
as adjuvant treatment for early-stage breast cancer: 10-year analysis of
the ATAC trial. Lancet Oncol. 2010;11(12):1135-1141.
239. Coates AS, Keshaviah A, Thürlimann B, et al. Five years of letrozole
compared with tamoxifen as initial adjuvant therapy for postmenopausal
women with endocrine-responsive early breast cancer: update of study
BIG 1-98. J Clin Oncol. 2007;25(5):486-492.
240. Ma CX, Adjei AA, Salavaggione OE, et al. Human aromatase: gene
resequencing and functional genomics. Cancer Res. 2005;65(23):11071-11082.
241. Moy B, Tu D, Pater J, et al. Clinical outcomes of ethnic minority women
in MA.17: a trial of letrozole after 5 years of tamoxifen in postmenopausal
women with early stage breast cancer. Ann Oncol. 2006;17(11):1637-1643.
242. Allman R, Dite GS, Hopper JL, et al. SNPs and breast cancer risk
prediction for African American and Hispanic women. Breast Cancer Res
Treat. 2015;154(3):583-589.
243. Parra A, Karnad AB, Thompson IM. Hispanic accrual on randomized
cancer clinical trials: a call to arms. J Clin Oncol. 2014;32(18):1871-1873.
244. Gordon NP, Iribarren C. Health-related characteristics and preferred methods of receiving health education according to dominant language among Latinos aged 25 to 64 in a large Northern California health
plan. BMC Public Health. 2008;8(1):1.
245. Merchant G, Buelna C, Castañeda SF, et al. Accelerometermeasured sedentary time among Hispanic adults: results from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL). Prevent Med
Rep. 2015;2:845-853.
246. Bonner D, Cragun D, Reynolds M, et al. Recruitment of a population-based sample of young black women with breast cancer through a
state cancer Registry. Breast J. 2016;22(2):166-172.
247. Darling M, Gonzalez F, Graves K, et al. Practical tips for establishing
partnerships with academic researchers: a resource guide for communitybased organizations. Prog Community Health Partnersh. 2015;9(2):203-212.
248. Rush CL, Darling M, Elliott MG, et al. Engaging Latina cancer survivors,
their caregivers, and community partners in a randomized controlled trial:
Nueva Vida intervention. Qua Life Res. 2015;24(5):1107-1118.
249. Detz A, Mangione CM, de Jaimes FN, et al. Language concordance, interpersonal care, and diabetes self-care in rural Latino patients.
J Gen Intern Med. 2014;29(12):1650-1656.
October 2016, Vol. 23, No. 4
Various clinical and treatment
factors differently affect sexual
minorities with breast cancer.
Stag Horn. X-Ray image courtesy of Bryan Whitney. www.x-rayphotography.com.
Clinical Considerations of Risk, Incidence, and Outcomes of
Breast Cancer in Sexual Minorities
Anne E. Mattingly, MD, John V. Kiluk, MD, and M. Catherine Lee, MD
Background: Breast cancer is a leading cause of cancer-related mortality in women. Limited research exists on
the impact of sexual orientation on overall risk of and mortality from breast cancer. We sought to summarize the
medical literature on breast cancer in sexual minority women and identify possible disparities in this population.
Methods: A comprehensive literature search was conducted for English-language studies in peer-reviewed medical
journals that referenced breast cancer and sexual minority, lesbian, bisexual, or transgender individuals.
Articles published between January 2000 and November 2015 were included. They were reviewed for relevance
to breast cancer risk stratification, breast cancer mortality, breast reconstruction, and transgender issues.
Results: Behavioral risks, reproductive risks, and risks associated with decreased access to health care may all
affect outcomes for sexual minorities with breast cancer. Limited studies have mixed results regarding mortality
associated with breast cancer in sexual minorities due to an inconsistent reporting of sexual orientation.
Conclusions: Overall, the research examining breast cancer in sexual minority women remains limited. This
finding is likely due to limitations in the reporting of sexual orientation within large databases, thus making
broader-scale research difficult.
Introduction
For American women, breast cancer remains the most
common cancer and second most common cause of
cancer-related mortality.1 In 2016, the estimated incidence of new breast cancer cases will be 246,660, or
29.0% of all new cancer cases, and 72,160 cancer-related deaths are estimated to be due to breast cancer.1
Approximately 1 in 8 women (12.3%) will be diagnosed
with breast cancer during their lifetime, and nearly
From the Comprehensive Breast Program, H. Lee Moffitt Cancer Center
& Research Institute, Tampa, Florida.
Submitted February 19, 2016; accepted May 11, 2016.
Address correspondence to M. Catherine Lee, MD, Comprehensive
Breast Program, Moffitt Cancer Center, 109201 North McKinley Drive,
MKC-BR PROG, Tampa, FL 33612. E-mail: [email protected]
No significant relationships exist between the authors and the companies/organizations whose products or services may be referenced
in this article.
October 2016, Vol. 23, No. 4
3 million women in 2012 were living with breast cancer
in the United States.2 Research has been performed on
racial disparities in the screening, diagnosis, treatment,
and survival rates of breast cancer, as well as disparities
due to insurance status and socioeconomic position.3
Disparity in the risk and mortality rates of breast cancer
are likely to continue until we better understand the underlying reasons for the divergence.3,4
According to US Census data, 1.2 million individuals live in a household with a same-sex partner.5 The
Surveillance, Epidemiology, and End Results (SEER)
Program collects demographical data, including age,
race, sex, income, education, and geographical location, but it does not collect data on sexual orientation.2,4 This fact makes research on the disparities in
breast cancer among women who identify as lesbian,
bisexual, or sexual minority women difficult. Sexual
minority women are infrequently the focus of studCancer Control 373
ies in breast cancer, and controversy exists on whether sexual minority women are at increased risk for
breast cancer compared with women in the general
population.6
Methods
The goal of this article is to provide health care professionals with a discussion framework for their patients
who are sexual minorities. This is a general overview
of the limited published research on breast cancer in
sexual minority patients focusing on clinical considerations. Within the medical literature we identified recent and comprehensive studies to address the following clinical questions:
• Are sexual minority women at increased risk for
developing breast cancer?
• Do sexual minority women have an increased
frequency of breast cancer related to a potential
increase in risk factors?
• Can health care professionals influence patient
behavior to mitigate the risk of breast cancer in
these sexual minority patients?
• Does any identified increased risk lead to a difference in outcomes for sexual minority women,
specifically breast cancer–related mortality?
• How do sexual minority women react to breast
reconstruction after completing their course of
treatment?
• How does the risk of breast cancer change in the
transgender population, specifically when taking
cross-sex hormone therapy?
Risk Factors
Risk factors for breast cancer have been extensively researched, and studies evaluating risk factors for breast
cancer in sexual minority women have been published. However, whether sexual minority women are
at an increased risk of breast cancer is still a controversial topic. For example, Cochran et al7 reviewed data
from 7 surveys of women who have sex with women
and self-reported bisexual women 18 to 75 years of age
and compared their responses to 2 large national surveys of women similar in age (N = 19,000). Case et al8
also examined sexual orientation and general health
risk factors, comparing self-reported sexual minority
women to heterosexual female participants in a prospective cohort of 116,671 registered nurses 25 to
43 years of age who were followed with biennial surveys starting in 1989. Sexual orientation was added to
the surveys in 1995, and demographical information
was collected as well as risk factors for breast cancer, other cancers, and cardiovascular disease. Sexual
orientation was reported by 98% of respondents, of
whom 0.8% identified as lesbian and 0.3% identified as
bisexual — thus making the sexual minority respondents a smaller subset of the larger cohort.8 It is diffi374 Cancer Control
cult to generalize data as all the respondents were premenopausal registered nurses, and their work likely
affected their interaction with the health care system
and may not have been reflective of other sexual minority women.
Breast cancer is much more common in persons
as they age, and a premenopausal cohort does not
give a complete picture of breast cancer either in sexual minority or heterosexual women.1 In 2012, Austin
et al9 used the same prospective cohort data as Case
et al8 to estimate risk of breast cancer in premenopausal women using a modified Rosner–Colditz risk model
and compared sexual minority women with heterosexual women. Similar to the Gail model, which uses
current age, age of menarche, age at first birth, family
history, previous benign results on breast biopsy, history of biopsy with atypia, and race to estimate risk of
breast cancer, the Rosner–Colditz model also includes
subsequent births following first live birth, type and
age of menopause, height, current weight, weight at
18 years of age, and alcohol consumption. Austin et al9
calculated a 1-year breast cancer incidence rate per
100,000 person-years and compared sexual minority
women with the heterosexual group.
However, these studies have limitations. The
self-reported nature of sexual orientation makes objective study inherently difficult. Oftentimes sexual orientation is not included in large databases of breast
cancer research.10 Sexual minority women may be
less likely to disclose sexual orientation to their health
care professional for fear of discrimination.11 As the
stigma of sexual minority status has lessened over the
decades, it is likely that, as the years go on, more respondents than those who initially self-reported would
have come forward as a sexual minority in large national surveys of women. For health care professionals, understanding the risk of breast cancer in sexual
minority women could help them to educate their patients, thereby improving rates of screening for breast
cancer, or to modify behaviors that could have numerous impacts on patient health in general.
From these studies, the risk factors examined can
be categorized as behavioral risks (obesity, alcohol
use, tobacco use), reproductive risks (parity, use of
oral contraceptives), and access to health care (health
insurance status, screening).7-9 A summary of selected
increased risks in sexual minority women are outlined
in the Table.7-9
Behavioral Risks
Obesity is associated with an increased risk of breast
cancer in postmenopausal women and a higher risk of
breast cancer recurrence in premenopausal and postmenopausal women.12 Cochran et al7 observed that 28%
of sexual minority women were classified as obese, suggesting that a significantly larger percentage of sexual
October 2016, Vol. 23, No. 4
minority women were obese than expected (P < .05).
Despite this, sexual minority women were significantly
less likely to consider themselves obese than women in
the general population (P < .05).7 Compared with heterosexuals, lesbians and bisexuals have higher prevalence rates of obesity (50% and 40%, respectively),8 and
both premenopausal groups have a significantly higher
mean body mass index (P < .0001 and P = .0004, respectively).9 These data suggest that sexual minority women
are more likely than heterosexual women to have challenges with obesity, which is a risk factor for breast
cancer as well as other health problems.
Evidence also suggests that smoking can cause
breast cancer.13 Cochran et al7 reported that sexual minority women were more likely to be current or pre-
vious smokers than those in the general population
(P < .05). Case et al8 found that lesbians and bisexuals reported higher rates of former smoking (60% and
50%, respectively), and twice as many sexual minority women were current smokers. Increased smoking
in women, including sexual minority women, likely
increases their risk for breast cancer as well as other
medical conditions.1
Alcohol has been considered a carcinogen to humans and has been shown to increase the risk of many
cancers, including female breast cancer.14 Cochran et
al7 did not note any significant differences in current
alcohol use among sexual minority women compared
with heterosexual women, but they did find an increased prevalence of alcohol-related problems, such
Table. — Comparison of Select Studies Evaluating Risk Factors for Breast Cancer in Sexual Minority Women
Author
Cochran7
Case8
Austin9
Range
1986–1997
1989–1995
1989–2005
Age, y
18–75
25–43
25–42
Type of
participant
Heterosexual women = 19,000
Sexual minority women = 11,886
Heterosexual women = 89,812
Lesbian = 694
Bisexual = 317
Heterosexual women = 87,392
Lesbian = 665
Bisexual = 309
Risk factor
Heterosexual
Sexual
minority
Heterosexual
Lesbian
Bisexual
Heterosexual
Lesbian
Bisexual
Body mass
index
18.3 kg/m2
27.7 kg/m2
(P < .05)
19.8%a
PR: 1.0
28.1%a
PR: 1.5
(95% CI: 1.4–1.7)
28.2%a
PR: 1.4
(95% CI: 1.1–1.6)
22.04 kg/m2b
23.28 kg/m2b
(P < .0001)
23.11 kg/m2b
(P = .0004)
4.0%c
12.4%c
(P < .05)
4.1%d
PR: 1.0
9.2%d
PR: 1.9
(95% CI: 1.5–2.3)
11.2%d
PR: 2.7
(95% CI: 2.1–3.5)
3.56 ge
6.18 ge;
(P < .0001)
5.18 ge;
(P < .0001)
Current
smoker, %
16.1
21.2
(P < .05)
10.6 f
PR: 1.0
18.9 f
PR: 2.0
(95% CI: 1.7–2.3)
20.6 f
PR: 2.2
(95% CI: 1.8–2.7)
Former
smoker, %
20.1
34.0
(P < .05)
23.9 g
PR: 1.0
33.5 g
PR: 1.6
(95% CI: 1.4–1.7)
31.5 g
PR: 1.5
(95% CI: 1.3–1.8)
Nulliparity
Pregnancy h:
66.7%
Live birthi:
56.9%
Pregnancy h:
28.1%
(P < .05)
Live birthi:
16.0%
(P < .05)
22.1%
PR: 1.0
76.5%
PR: 3.3
(95% CI: 2.9–3.6)
49.5%
PR: 2.1
(95% CI: 1.8–2.6)
No. of births:
1.71
No. of births:
0.38
(P < .0001)
No. of births:
0.98
(P < .0001)
History of
OCP use, %
79.7
36.2
(P < .05)
—
—
—
—
—
—
Alcohol
intake
NA
a BMI
≥ 30.0 kg/m2 compared with < 25.0 kg/m2.
BMI premenopause.
c History of self-reported alcohol-related problems.
d Alcohol intake ≥ 15 g/d vs none.
e Alcohol intake/d (g) during premenopause.
f Current smoker vs never smoker.
g Former smoker vs never smoker.
h History of pregnancy.
iHistory of live birth.
BMI = body mass index, CI = confidence interval, NA = not applicable, OCP = oral contraceptive pills, PR = prevalence ratio.
b Mean
October 2016, Vol. 23, No. 4
Cancer Control 375
as history of alcoholism, in sexual minority women
compared with heterosexual women (P < .05). By contrast, 2 other studies found significantly increased rates
of alcohol consumption: Twice as many sexual minority women drank at least 15.0 g of alcohol every day,
and a significantly greater mean daily intake of alcohol during premenopause was seen among lesbians
(P < .0001) and bisexuals (P < .0001) compared with
heterosexual women.8,9 Thus, increased alcohol intake in sexual minority women increases the potential risk of breast cancer as well as other health problems in this population.1
Reproductive Risks
Nulliparity has been associated with an increased risk
of breast cancer, and increased parity can reduce a
woman’s risk of breast cancer, particularly HR-positive breast cancer.15 Cochran et al7 demonstrated that
sexual minority women had lower lifetime rates of
pregnancy and live births when compared against
national data (P < .05 for both). Furthermore, subsequent studies demonstrate that nulliparity is 3 times
more likely in lesbians and twice as likely in bisexual
women, with significantly reduced number of births
in lesbian and bisexual women compared with heterosexual women (P < .0001 for both).8,9 These data
suggest that sexual minority women are significantly
more likely to be nulliparous, thus increasing their
risk of breast cancer.
If sexual minority women did have children, Case
et al8 found that they were twice as likely to have given
birth before 19 years of age, a factor associated with
a decreased risk of breast cancer,16 conferring up to a
50% risk reduction compared with nulliparous women.17 However, Austin et al9 found no significant difference in age at first birth between lesbian and bisexual
women compared with heterosexual women. This is
a confounding factor when attempting to elucidate reproductive risk factors for breast cancer in sexual minority women.
Use of oral contraceptives is associated with a
decreased risk of HR-positive breast cancer.15 Cochran
et al7 noted that sexual minority women reported significantly lower rates of oral contraceptive pill use than the
general female population (P < .05). Although Austin et
al9 did not specifically look at this factor, they did study
age of menarche and found a 20% increase rate of menarche at 11 years of age or younger in lesbian women;
however, subsequent data found no significant difference between lesbian and bisexual women compared
with heterosexual women. Thus, these reproductive
risk factors deserve further study.
Access to Health Care
Health insurance status is important when discussing preventive care such as screening for breast can376 Cancer Control
cer. At the time of publication, most sexual minority
women are unable to marry their partners by law and
may have had impaired access to a partner’s employer health insurance.7 Cochran et al7 indicated lower
prevalence rates of health insurance coverage (P < .05)
among sexual minority women compared with standardized estimates, and sexual minority women had
lower rates of previous mammograms when standardized for demographics across all age groups (P < .05).
Current health insurance coverage was associated with
a positive history for mammography among sexual minority women in their 40s, with 77% of insured sexual
minority women vs 64% of uninsured sexual minority
women having undergone mammography at least once
(P < .05).7 This finding suggests that health insurance
coverage could improve access to screening mammography in sexual minority women, and that sexual minority women are less likely to have health insurance
coverage. However, Case et al8 found no significant
difference between sexual minority women compared
with heterosexual women in regard to history of mammography. Likely, this is an evolving issue. With the recent legalization of same-sex marriage, access to employer-sponsored health care insurance may change
for sexual minority women.
Despite the increased individual risk factors for
breast cancer described in sexual minority women,
no definitive studies have demonstrated an increased
prevalence of breast cancer in sexual minority women.7-9 Using the Rosner–Colditz risk model, Austin
et al9 predicted an incidence rate of breast cancer significantly elevated in both lesbian and bisexual women (P < .0001 for both), and this rate was consistently
predicted across all age groups. Regression analysis
found lesbian and bisexual women were more likely to
be at higher risk than heterosexual women of similar
age, race, geography, and income.
However, sexual minority women have not been
shown to have an increased rate of breast cancer when
compared with the general population. Cochran et al7
were unable to link sexual minority women with an
increased prevalence of breast cancer compared with
US women in general. A self-reported history of breast
cancer among sexual minority women was 0.9%,
which was concordant with expected standardized
estimates, and no statistically significant difference in
self-reported prevalence of breast cancer between the
study sample of sexual minority women and US estimates for women was observed.7 Therefore, sexual minority women may not have an increased incidence or
prevalence rate of breast cancer despite their increased
predicted risk.
Modulating Risk
Based on identified individual risk factors, attempts are
warranted by health care professionals to modify paOctober 2016, Vol. 23, No. 4
tient behavior in the hopes that doing so will improve
the health of sexual minority patients. It is important
to note that many of the risk factors for breast cancer
identified in sexual minority women are modifiable
behavioral risks.7-9,12,14
Bowen et al18 attempted to modulate risk by increasing screening participation in sexual minority women using group counseling. This prospective
study randomized sexual minority women to a counseling (n = 69) or control group (n = 81). The intervention included psychosocial counseling with education,
group discussion, and skills training during 4 weekly
2-hour sessions led by a trained health counselor specializing in sexual minority women. Weekly sessions
had themes that included a personalized breast cancer
risk assessment, discussions of mammography, recommendations for self-examinations and clinical breast
examination, stress management, progressive muscle
relaxation and guided imagery, and social support in
4 main areas (emotional, informational, instrumental,
and companionship) focused on devising strategies for
increasing support where needed.18
At 6 and 24 months, participants were evaluated on
perceived risk of breast cancer, worry of cancer, mental health, “outness,” and identification with the lesbian
community.18 At 24 months, the intervention group reported a higher rate of breast cancer screening in the
past 2 years compared with the control group (87% vs
75%; P < .05). The intervention group also had a significantly higher rate of breast self-examinations at 6
(P < .01) and 24 months (P < .05) than the control group.18
The intervention group changed more over time in
perceived risk (P < .001), cancer worry (P < .001), and
mental health (P < .01).18 Regression analysis showed
identification with the lesbian community and feelings
of outness were predictors of participation in screening
mammography and a feeling of outness was also a significant predictor of performing a breast self-examination.
With education and group counseling structured
for sexual minority women, these researchers demonstrated that trained health counselors can successfully
increase participation of sexual minority women in
breast cancer screening.18 However, it is worth noting
that this study has limitations because it studied 1 volunteer group in a single community, thus making it difficult to generalize to all sexual minority women.
It is encouraging to think that an intensive, demographical directed counseling intervention can improve the participation of sexual minority women in
breast cancer screening.18 However, without a heterosexual cohort, it is difficult to know whether behavior
modification is specific to sexual minority women or
generalizable to others. Although it is not reasonable
to assume that a health care professional would have
the resources to educate his or her sexual minority patients to such a degree that this study entails, improvOctober 2016, Vol. 23, No. 4
ing participation in breast cancer screening for sexual
minority women is important. Sexual minority women
have consistently lower lifetime prevalence rates of
screening mammography than expected across all age
groups, although this is likely confounded by their decreased access to health care.7
Improving the participation rates of breast cancer
screening among sexual minority women are important, but opportunities still exist to modulate other
modifiable high-risk behaviors identified in sexual
minority women, such as alcohol intake, obesity, and
smoking.7-9 Health care professionals should be aware
of these modifiable risk factors associated with breast
cancer and other health problems, as they may present
an educational opportunity to promote healthy behaviors. Clinicians should also be encouraged that intensive education is successful at promoting behavioral
change in sexual minority women and improving their
participation with breast cancer screening.18
Mortality
The goals of reducing risk and modifying behavior
should be to lower the mortality rate associated with
breast cancer and promote healthy living. Cochran
and Mays6 examined the mortality risk of breast cancer among sexual minority women using the National
Health Interview Survey, which is an annual population-based, household interview of married or cohabitating women 18 to 80 years of age that was conducted
between 1997 and 2003. Of the 155,427 respondents,
693 reported having female partners (0.5%).6 Sexual
minority women were younger (P < .001), had higher
levels of education (P < .001) and higher family incomes (P < .05), were less likely to have health insurance (P < .05), and were more likely to be in a cohabitating relationship rather than be married (P < .001).6 The
mortality rate of breast cancer was identified using the
National Death Index: Of the 4,396 identified deaths,
274 (3.1%) were attributed to death from breast cancer.6
Age, race, education, family income, and health insurance coverage were strongly associated with death
from any cause (P < .001 for all).6 After adjusting for
confounding variables, sexual minority women were
not at increased risk of overall mortality; however,
sexual minority women had an elevated risk for breast
cancer–related mortality compared with heterosexual women (P < .05).6 Overall, sexual minority women
were 3 times more likely to die from breast cancer after adjusting for age, although the researchers did not
evaluate the prevalence of breast cancer in the sexual
minority population. The data are limited because they
only include women who were in a defined relationship and, thus, the data cannot be generalized to sexual minority women without partners.
In interpreting potential reasons for this difference in mortality rates, a review of the risk facCancer Control 377
tors previously discussed is important. Is there an
increased rate of incidence or prevalence of breast
cancer not yet identified in sexual minority women
because of reproductive and behavioral risks that
leads to this increased mortality risk of breast cancer?
Perhaps the increased mortality risk of breast cancer
among sexual minority women could be due to later
diagnoses because of decreased access to health care
due in part to lack of insurance or decreased rates of
screening.
Cochran and Mays19 attempted to validate their
findings by reviewing mortality risks associated with
sexual orientation both in women and in men using
data from the General Social Surveys and comparing mortality status using the same National Death
Index. Respondents were interviewed face-to-face
in their household and then were self-administered
questionnaires or computer-assisted self-interviews;
these responses were matched to National Death Index records.19 Sexual orientation was classified based
on lifetime numbers of male or female sexual partners and the gender of those sexual partners in the
year prior to the interview. Of the 17,886 respondents who completed the interview/questionnaire,
853 (4.8%) reported a history of same-sex partners.
Results were then subdivided by race, education, income, geographical region, overall self-rated health
status, and generalized self-reported happiness.
Similar to the previous study, sexual minority women were younger and reported higher education6;
however, in this cohort, sexual minority women had
lower household incomes and, geographically, more
sexual minority women lived in western states.19
All-cause mortality among women was significantly
associated with older age, non-white race, and lower
education (P < .001 for all), but sexual minority status
was not significantly correlated with all-cause mortality or breast cancer, even after multivariate survival analysis (all-cause mortality, P = .72; breast cancer
mortality, P = .27).19 Overall, 9.1% of sexual minority women died from breast cancer vs 4.4% of heterosexual women.19 Thus, Cochran and Mays19 were unable to replicate their 2013 findings,6 and it is unclear
whether sexual minority women have an increased
risk of mortality from breast cancer.
The later study benefited from a much broader
definition of sexual orientation (it included lifetime
partners), which helped capture people not in current same-sex relationships.19 However, sexual orientation remains difficult to objectively measure due
to inherent self-reporting bias in identifying sexual
minority status. Overall, the later study reported
more same-sex partners (4.8% vs 0.5%), but this is
still a relatively low percentage of respondents —
possibly underpowering the study.6,19 Thus, further
research is necessary.
378 Cancer Control
Breast Reconstruction
A unique aspect of the treatment for female breast cancer is the impact that surgery, including breast reconstruction, can potentially have on a patient’s self-identity.20 In general, breast cancer treatment is dictated
by tumor biology, particularly with regard to systemic
therapy; sexual orientation has no impact on chemotherapeutic or hormonal therapy recommendations.1
However, surgery for breast cancer is the cornerstone
of breast cancer management in many American women.1 Decision-making regarding breast reconstruction
in these patients is not well understood, but it has the
potential for disparate care among women.20
In particular, breast reconstruction is scrutinized
because of the presumed heterosexual focus of the
breast, as well as an example of health care access.20
A paucity of literature exists on use of reconstruction
options in sexual minority women. It is possible that
sexual orientation and self-identity could have significant impact on decisions regarding type of oncological surgery and whether or not a patient would desire
reconstruction. Sexual minority women may perceive
that health care professionals are not always considering sexual orientation when making recommendations
for surgery or reconstruction.
The scientific literature on sexual minority women and their decisions to undergo breast reconstruction is limited: A single, retrospective, qualitative study
has explored decisions by sexual minority women for
or against reconstruction.20 A total of 15 self-reported
sexual minority women diagnosed with nonrecurrent
and nonmetastatic breast cancer who completed invasive treatment within the past 5 years completed demographical surveys and a disclosure scale. These participants were separated into breast reconstruction and no
breast-reconstruction groups. Semi-structured interviews covered diagnosis, treatment course, treatment
decision-making, sources of social support, and adjustment; participants identified support persons who were
then interviewed covering similar topics. Comparisons
of demographics, clinical stage, receipt of adjuvant therapy, and time since diagnosis revealed that the groups
were similar. All support persons were female, and the
majority of support persons were partners. Mastectomy
was recommended for some, whereas others opted for
mastectomy to avoid radiotherapy; one-half of participants underwent breast conservation before undergoing
mastectomy.20 Of those in the reconstruction group
(n = 8), 3 had autologous reconstruction, 5 had implant-based reconstruction, 3 received chemotherapy, 3 received radiotherapy, and 6 received hormonal therapy; of those in the no-reconstruction group
(n = 7), 4 received chemotherapy, 2 received radiotherapy, and 4 received hormonal therapy.20
The researchers discovered that breast size was a
consideration among the participants when deciding
October 2016, Vol. 23, No. 4
whether to accept or reject reconstruction.20 Sexual
minority women with small breasts felt more comfortable rejecting reconstruction, whereas sexual minority women with larger breasts feared being unbalanced
and having a heavy prosthesis. Both groups mentioned
the importance of the breast for their self-image but
emphasized physical functioning and body strength
over aesthetics. Sexual minority women emphasized
that their sexual minority status contributed to them
not being defined by the presence of their breasts, and
they noted a discrepancy between their values and
body image and those of mainstream society — this
was especially true when communicating with specialists in breast cancer. Sexual minority women who
decided against reconstruction emphasized strength,
long-term health, and survival; they complained less of
physical problems but did discuss having to adjust to
their changed body shape; they did not voice doubts or
regrets about their choice. Their partners emphasized
body image and their value system as motivators, and
they confirmed the same satisfaction with respect to
body image. They characterized making the decision
jointly, and they reported attraction and desire being
present as before.20
Sexual minority women who underwent reconstruction reported physical issues, including numbness
and limited range of motion; 1 woman reversed her reconstruction completely.20 The group expressed doubt
or regret about their choice and lack of information
about potential complications. Some opted for reconstruction to cover the physical effects of the cancer and
regain a “normal” appearance, although some chose to
forego formal nipple reconstruction.20 Their partners
confirmed that outcomes of the reconstruction had not
been thoroughly explained by the plastic surgeon, and
they echoed doubts about reconstruction; their partners were characterized as more passive in the decision-making process, and these couples displayed discordance in values and body image.20 Women without
a partner at the time of their decision were influenced
by their relationship status, and support persons not in
a partner relationship were more reluctant to express
reconstructive preferences.
Overall, sexual minority women who underwent
reconstruction expressed doubt and regret; by contrast, sexual minority women who elected against reconstruction did not, and this finding was echoed by
their support persons.20 However, this study has limitations, including its small convenience sample, retrospective recall bias, and, thus, low applicability to the
general population.20 This study also has no comparison to a heterosexual control, making it difficult to define the themes as unique to sexual minority women,
but it did outline potential themes that may arise for
sexual minority women as they embark on their decision for or against reconstructive surgery.20
October 2016, Vol. 23, No. 4
It is also worth underscoring that the notion that
women may regret undergoing reconstruction after
mastectomy may not be limited to the sexual minority community. Metcalfe et al21 prospectively followed
patients after mastectomy with and without delayed
breast reconstruction and found that psychosocial
functioning improves over time in patients following
mastectomy. During the long-term follow-up, women
with delayed breast reconstruction had significantly
higher levels of total distress (P = .01), obsessiveness
(P = .03), and cancer-related distress (P = .02) compared with those who underwent mastectomy alone,
although no differences were seen in quality of life between the 2 groups.21 Metcalfe et al22 published another report that demonstrated no difference in body image for women with mastectomy alone compared with
mastectomy and immediate breast reconstruction.
They further found that women with delayed breast reconstruction had higher levels of body stigma (P = .01),
body concerns (P = .002), and concerns regarding the
obviousness of mastectomy-related changes in appearance (“transparency”; P = .001) than mastectomy alone
or mastectomy with immediate reconstruction.22
Another study reported on women who underwent breast-conserving surgery, mastectomy alone,
and mastectomy with reconstruction, and it found
that women who underwent mastectomy with reconstruction had greater mood disturbance and poorer
well-being (P = .002 for both) compared with those
who underwent mastectomy alone.22 This finding
persisted up to 18 months after surgery.
All of these studies suggest the potential for
selection bias for women who undergo breast reconstruction — in particular, delayed breast reconstruction — as pursuing that option because of their underlying distress regarding their body image.20-22 It is
difficult to generalize this information to sexual minority women, but these issues of dissatisfaction may
not be unique to either population. It is important for
health care professionals to understand that sexual
minority women may have a different perspective on
breast reconstruction after mastectomy regardless of
their surgical choice for or against reconstruction.
Boehmer et al22 described sexual minority women
as not being defined by having breasts, emphasizing
body strength and physical functioning over aesthetics,
and “otherness,” described as a discrepancy of values
and body image among sexual minority women when
compared with the general population. Overall, health
care professionals must be aware of a patient’s sexual
orientation and include the patient’s support persons
or partners in discussions regarding treatment. Health
care professionals should also consider how the presence or absence of the breast might affect the patient’s
body image, understanding that sexual minority women may not feel emotionally attached to the physical
Cancer Control 379
presence of their breasts. Health care professionals
should also discuss physical functioning after breast
reconstruction and not limit the discussion to aesthetics alone. Reconstructive options should be clearly and
comprehensively presented, elucidating realistic expectations for patients.
Transgender Persons
The term transgender defines a person whose
self-identity does not conform to gender norms, and
many patients may choose to use medical or surgical
means to live as another gender. The risk of breast cancer is particularly relevant due to cross-sex hormone
therapy for many patients who desire to feminize or
masculinize their appearance.
In general, research on breast cancer in the transgender community is limited, and our understanding
is also limited with regard to how hormonal therapy
affects a person’s risk of breast cancer. Sattari et al23
summarized the cases of male-to-female transgender
patients with breast cancer published in the literature
since 1968. The authors identified 10 cases of breast
cancer in male-to-female transgender persons, all of
whom received long-term cross-sex hormone treatment;
7 of the cases had ER status reported (n = 5 negative,
n = 2 positive).23 Thus, breast cancer in general appears
to be rare among male-to-female transgender persons,
although it is difficult to make any generalizations
about the risks of cross-sex hormone therapy in this
transgender population.
Understanding the risk of breast cancer in femaleto-male transgender persons who take testosterone for
masculinization is important, because it is likely different from the risk of breast cancer in male-to-female
transgender persons. Circulating testosterone is converted to estradiol by aromatase enzyme, and higher
levels of circulating testosterone could increase peripheral estrogen. Shao et al24 described 2 cases of female-to-male transgender persons, both of whom took
testosterone for masculinization. One patient was postmenopausal (aged 48 years) when testosterone was
initiated, was also taking an aromatase inhibitor for
symptoms of endometriosis, and was diagnosed with
breast cancer 5 years later; the other case patient was
premenopausal (aged 21 years) when testosterone
was initiated and then diagnosed with breast cancer
6 years later. Both patients had ER-positive breast cancer, and both tested negative for a deleterious BRCA
mutation.24 These cases are worth noting, but they
do not provide conclusive evidence for an increased
risk of breast cancer in transgender persons receiving
cross-sex hormone therapy.
A study by the US Veterans Health Administration
(VHA) is the largest and most comprehensive research
on breast cancer in the transgender community (as of
publication).25 This research has evaluated the inci380 Cancer Control
dence of breast cancer in transgender men and women
treated within the VHA. Using encounter and prescription data from 1996 to 2013, transgender-related diagnosis and breast cancer events were identified within
the VHA system.25 Medical records were reviewed for
any patients with both a transgender-related diagnosis
and a breast cancer event; cross-sex hormone therapy
prescribed by a VHA physician was classified as androgen/anabolic or estrogen. The incidence rates of breast
cancer were compared with Surveillance, Epidemiology, and End Results data from 2007 to 2011 using overall observed and expected rates.25
Of approximately 6 million veterans seen every
fiscal year, 5,135 had a transgender-related diagnosis,
1,579 were female at birth, and 3,556 were male at birth.25
No statistically significant difference was observed in
breast cancer incidence among women, men, and in the
overall population. For the entire sample, 10 confirmed
cases of breast cancer were found in transgender veterans
(0.2%), 7 in female-to-male veterans, 2 in male-to-female
veterans, and 1 in a natal male veteran with transvestic
fetishism.25 The average age at diagnosis was 63.8 years
and all participating veterans racially self-identified as
non-Hispanic white. Breast cancer surgery was performed in 8 transgender veterans; 4 patients died. Of
those patients not receiving cross-sex hormone therapy, breast cancer was diagnosed in 3 transgender men
and 1 transgender woman. For those patients receiving
cross-sex hormone therapy breast cancers occurred
in 3 transgender women, and 3 cases of female breast
cancer occurred in transgender veterans who received
cross-sex hormone therapy many years following their
diagnosis of breast cancer.25
More than one-half (52%; n = 2,645) of transgender
veterans received at least 1 prescription for cross-sex
hormone therapy, with the average duration of treatment
being 6.5 years and the average age at initial treatment
being 48.9 years.25 None of the males prescribed crosssex hormone therapy developed breast cancer, suggesting that male breast cancer is rare in the transgender
population receiving cross-sex hormone therapy. Breast
cancers occurred in 3 transgender female veterans receiving cross-sex hormone therapy (n = 2 received estrogen products, n = 1 received testosterone products)
prior to diagnosis.25 In general, no significant difference was observed for transgender patients receiving
estrogen, testosterone, or cross-sex hormone therapy
when compared with the overall cohort.24 Transgender
veterans receiving cross-sex hormone therapy had no
increased risk of breast cancer, and the overall incidence of breast cancer in transgender persons was low;
however, late presentations in the 3 male patients with
breast cancer and the high mortality rate seen in transgender veterans are concerning.25
Clinicians should consider breast and chest examinations and possible mammography, if indicated, in this
October 2016, Vol. 23, No. 4
subset of patients. History of chest-contouring surgery
or mastectomy does not preclude the development of
breast cancer, as observed in 1 female-to-male transgender patient in this study.25 This study did not include
gene mutation data because they were not readily available, but it is the largest series of transgender patients
with breast cancer reported in the literature to date.25
Interpretation of the use of cross-sex hormone therapy
may be limited because the study required that at least
1 prescription was obtained through the VHA system
to identify the patient as receiving cross-sex hormone
therapy.24 For example, if a patient received cross-sex
hormone therapy outside of the VHA system, then the
therapy may not have been recorded.25
Typically, male breast cancer is hormonally sensitive (92%).2,26 As data have emerged and are now associating female breast cancer with use of perimenopausal estrogen-based hormone replacement therapy,
concern exists about the potential risks of estrogen
therapy in male-to-female transgender persons.27 However, male breast cancer is rare in the transgender
population.25 At this time, the evidence is insufficient
to support an increased incidence of breast cancer in
transgender male-to-female persons, even when taking cross-sex hormone therapy.
Risk of breast cancer may be different in the female-to-male transgender population compared with
the general population. In female-to-male persons receiving testosterone cross-sex hormone therapy, theoretical concern of breast cancer is extrapolated from
the data on perimenopausal estrogen-based hormone
therapy.27 The theory postulates that the metabolism of
testosterone could increase peripheral estrogen levels,
and, as reported in Shao et al,24 2 such patients both
developed ER-positive breast cancer. However, only
1 female-to-male patient receiving testosterone crosssex hormone therapy in the VHA transgender cohort developed breast cancer.25 Although transgender females
are at an increased risk of breast cancer due to their sex
at birth compared with male-to-female transgender patients, the data are lacking for us to conclude whether
an increased risk of breast cancer exists in transgender
female-to-male patients receiving cross-sex hormone
therapy compared with the general female population.
Moreover, the data do not support an increased incidence of breast cancer in transgender persons receiving
cross-sex hormone therapy, and, in general, breast cancer in transgender persons is rare or under-reported.23
However, the scientific research related to transgender
persons is lacking, making conclusions difficult to draw.
It does highlight the fact that more research is needed.
Conclusions
Breast cancer is the most common cancer and second
most common cause of cancer-related death in American women.1 Given the pervasive nature of breast canOctober 2016, Vol. 23, No. 4
cer, many researchers have looked into specific groups
of women in an effort to identify and reduce disparities
in breast cancer among different populations of women. Numerous racial and socioeconomic disparities
have already been identified, but research on sexual
orientation and disparities in breast cancer is still limited and the data contradictory.3 Some evidence supports that sexual minority women have higher rates
of risk factors associated with breast cancer.6,7 Studies
are conflicting on which risk factors are more common in sexual minority women, but such risk factors
consistently noted to be increased in sexual minority
women include nulliparity, a history of smoking, and
increased alcohol consumption.7-9 Published data also
showed lower rates of health insurance coverage and
decreased rates of breast cancer screening in sexual
minority women.6,7 Despite an increase in risk factors, no clear increased incidence or prevalence rate
for breast cancer in sexual minority women has been
identified,7,8 and research on breast cancer in the transgender population remains limited. Utilizing education and structured support groups for sexual minority women help to improve rates of participation in
breast cancer screening,18 and health care professionals
should consider breast and chest examinations as well
as mammography in transgender patients.
Clinicians should also be aware of the sexual orientation of their patients and the potential association with disparities in breast cancer and differences
in related outcomes. Recent legislation supporting
same-sex marriage may improve the documentation of
sexual orientation, so health care professionals should
be aware of their patient’s sexual orientation and include support persons in treatment discussions, as
needed, to ensure they understand the patient’s goals
while counseling them on treatment options, including breast reconstruction. Consistent documentation of
gender preferences as part of the medical record could
improve the delivery of personalized care. Large-scale
databases should consider adding sexual orientation
to their demographical data to allow for more robust
research. With these considerations in mind, researchers should consider standardizing terminology regarding sexual minority patients to allow for more cohesive
dialogue in the scientific literature.
References
1. American Cancer Society. Cancer Facts & Figures 2016. http://www.
cancer.org/acs/groups/content/@research/documents/document/acspc047079.pdf. Accessed October 3, 2016.
2. National Cancer Institute. Surveillance, Epidemiology, and End Results
Program. SEER Stat Fact Sheets: Female Breast Cancer. http://seer.cancer.
gov/statfacts/html/breast.html. Accessed July 1, 2016.
3. Bigby J, Holmes MD. Disparities across the breast cancer continuum.
Cancer Causes Control. 2005;16(1):35-44.
4. Brown JP, Tracy JK. Lesbians and cancer: an overlooked health
disparity. Cancer Causes Control. 2008; 19(10):1009-1020.
5. Mayer KH, Bradford JB, Makadon MD, et al. Sexual and gender
minority health: what we know and what needs to be done. Am J Pub Health.
2008;98(6):989-995.
Cancer Control 381
6. Cochran SD, Mays VM. Risk of breast cancer mortality among women
cohabiting with same sex partners: findings from the National Health Interview
Survey, 1997-2003. J Womens Health (Larchmt). 2012;21(5):528-533.
7. Cochran SD, Mays VM, Bowen D, et al. Cancer-related risk indicators
and preventative screening behaviors among lesbians and bisexual women.
Am J Public Health. 2001;91(4): 591-597.
8. Case P, Austin SB, Hunter DJ, et al. Sexual orientation, health risk
factors, and physical functioning in the Nurses’ Health Study II. J Womens
Health (Larchmt). 2004;13(9):1033-1047.
9. Austin SB, Pazaris MJ, Rosner B, et al. Application of the RosnerColditz risk prediction model to estimate sexual orientation group disparities
in breast cancer risk in a U.S. cohort of premenopausal women. Cancer
Epidemiol Biomarkers Prev. 2012;21(12):2201-2208.
10. Meads C, Moore D. Breast cancer in lesbians and bisexual women:
a systematic review of incidence, prevalence and risk studies. BMC Public
Health. 2013;13:1127.
11. Hart SL, Bowen DJ. Sexual orientation and intentions to obtain
breast cancer screening. J Womens Health (Larchmt). 2009;18(2):177-185.
12. Jain R, Strickler HD, Fine E, et al. Clinical studies examining the
impact of obesity on breast cancer risk and prognosis. J Mammary Gland
Biol Neoplasia. 2013;18(3-4):257-266.
13. Reynolds, P. Smoking and breast cancer. J Mammary Gland Biol
Neoplasia. 2013;18(1):15-23.
14. Scoccianti C, Lauby-Secretan B, Bello PY, et al. Female breast cancer and alcohol consumption: a review of the literature. Am J Prev Med.
2014;46(3 suppl 1): S16-S25.
15. Anderson KN, Schwab RB, Martinez ME. Reproductive risk factors
and breast cancer subtypes: a review of the literature. Breast Cancer Res
Treat. 2014;144(1):1-10.
16. Hunter DJ, Speigelman D, Adami HO, et al. Non-dietary factors as
risk factors for breast cancer and as effect modifiers of the association of fat
intake and risk of breast cancer. Cancer Causes Control. 1997;8(1):49-56.
17. Kobayashi S, Sugiura H, Ando Y, et al. Reproductive history and
breast cancer risk. Breast Cancer. 2012;19(4):302-308.
18. Bowen DJ, Powers D, Greenlee H. Effects of breast cancer risk counseling for sexual minority women. Health Care Women Int. 2006;27(1):59-74.
19. Cochran SD, Mays VM. Mortality risks among persons reporting
same-sex sexual partners: evidence from the 2008 General Social SurveyNational Death Index data set. Am J Public Health. 2015;105(2): 358-364.
20. Boehmer U, Linde R, Freund KM. Breast reconstruction following
mastectomy for breast cancer: the decisions of sexual minority women.
Plast Reconstr Surg. 2007;119(2):464-472.
21. Metcalfe KA, Zhong T, Narod SA, et al. A prospective study of mastectomy patients with and without delayed breast reconstruction: long-term
psychosocial functioning in the breast cancer survivorship period. J. Surg.
Oncol. 2015;111(3):258-264.
22. Metcalfe KA, Semple J, Quan ML, et al. Changes in psychosocial
functioning 1 year after mastectomy alone, delayed breast reconstruction,
or immediate breast reconstruction. Ann Surg Oncol. 2012;19(1):233-241.
23. Sattari M. Breast cancer in male-to-female transgender patients: a
case for caution. Clin Breast Cancer. 2015;15(1):e67-e69.
24. Shao T, Grossbard ML, Klein P. Breast cancer in female-to-male
transsexuals: two cases with a review of physiology and management. Clin
Breast Cancer. 2011;11(6): 417-419.
25. Brown GR, Jones KT. Incidence of breast cancer in a cohort of 5,135
transgender veterans. Breast Cancer Res Treat. 2015;149(1):191-198.
26. Ruddy KJ, Winer EP. Male breast cancer: risk factors, biology, diagnosis, treatment, and survivorship. Ann Oncol. 2013;24(6):1434-1443.
27. Women’s Health Initiative Study Group. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results
from the Women’s Health Initiative randomized controlled trial. JAMA.
2002;288(3):321-333.
382 Cancer Control
October 2016, Vol. 23, No. 4
A large racially and ethnically diverse
database was created and TMAs
of IDC were constructed to represent
African American, Hispanic, and
non–Hispanic-white patient populations.
Proteus. X-Ray image courtesy of Bryan Whitney. www.x-rayphotography.com.
Construction and Validation of a Multi-Institutional Tissue
Microarray of Invasive Ductal Carcinoma From Racially
and Ethnically Diverse Populations
Edward Seijo, MS, Diana Lima, MPH, Egiebade Iriabho, MS, Jonas Almeida, PhD, Jesus Monico, MPH, MS,
Margarita Echeverri, PhD, Sylvia Gutierrez, MD, Idhaliz Flores, PhD, Ji-Hyun Lee, PhD, Kate Fisher, MA,
William E. Grizzle, MD, PhD, Gabriel L. Sica, MD, PhD, Charles Butler, Chindo Hicks, PhD,
Cathy D. Meade, PhD, RN, Stephen Olufemi Sodeke, PhD, Krzysztof Moroz, MD, Domenico Coppola, MD,
and Teresita Muñoz-Antonia, PhD
Background: The scarcity of tissues from racial and ethnic minorities at biobanks poses a scientific constraint
to research addressing health disparities in minority populations.
Methods: To address this gap, the Minority Biospecimen/Biobanking Geographic Management Program for
region 3 (BMaP-3) established a working infrastructure for a “biobanking” hub in the southeastern United
States and Puerto Rico. Herein we describe the steps taken to build this infrastructure, evaluate the feasibility
of collecting formalin-fixed, paraffin-embedded tissue blocks and associated data from a single cancer type
(breast), and create a web-based database and tissue microarrays (TMAs).
Results: Cancer registry data from 6 partner institutions were collected, representing 12,408 entries from
8,279 unique patients with breast cancer (years 2001–2011). Data were harmonized and merged, and deidentified information was made available online. A TMA was constructed from formalin-fixed, paraffin-embedded
samples of invasive ductal carcinoma (IDC) representing 427 patients with breast cancer (147 African
Americans, 168 Hispanics, and 112 non-Hispanic whites) and was annotated according to biomarker status
and race/ethnicity. Biomarker analysis of the TMA was consistent with the literature.
Conclusions: Contributions from participating institutions have facilitated a robust research tool. TMAs of
IDC have now been released for 5 projects at 5 different institutions.
From the Tissue Core Facility (ES), Departments of Thoracic Oncology (DL), Health Outcomes and Behavior (CM), and Pathology (CD),
Biostatistics and Bioinformatics Core (KF), and Tumor Biology Program (TM-A), H. Lee Moffitt Cancer Center & Research Institute, Tampa,
Florida, Pathology Informatics Core (EI, JA) and Pathology Program for Translational Research in Neoplasia (WEG), University of Alabama at
Birmingham, Birmingham, Alabama, Departments of Pathology (JM) and Medicine and Cancer Institute (CH), University of Mississippi Medical
Center, Jackson, Mississippi, Center for Minority Health and Health Disparities Research and Education, Xavier University of Louisiana (ME)
and Department of Pathology, Tulane University School of Medicine (KM), New Orleans, Louisiana, Department of Anatomy (SG) and Department of Microbiology (IF), Ponce Health Sciences University, Ponce, Puerto Rico, Department of Internal Medicine, University of New Mexico,
Albuquerque, New Mexico (J-H L), Department of Pathology and Laboratory Medicine, Winship Cancer Institute and Emory School of Medicine,
Atlanta, Georgia (GLS, CB), and Tuskegee University National Center for Bioethics in Research and Health Care, Tuskegee, Alabama (SOS).
Submitted March 3, 2016; accepted May 31, 2016.
Address correspondence to Teresita Muñoz-Antonia, PhD, Tumor Biology Program, Moffitt Cancer Center, 12902 Magnolia Drive,
MRC-3 WEST, Tampa, FL 33612. E-mail: [email protected]
This work was supported in part by the Tissue Core Facility and Bioinformatics and Biostatistics Core at Moffitt Cancer Center,
a National Cancer Institute–designated Comprehensive Cancer Center (P30-CA076292).
October 2016, Vol. 23, No. 4
Cancer Control 383
Introduction
The burden of cancer disparities on African American and Hispanic populations has been well documented.1-8 It is our opinion that discoveries in precision
medicine and basic science with the potential to improve the prognosis or treatment of cancer in affected
individuals might be limited by the scarcity of research
biospecimens from racial/ethnic minority populations.
The availability of diverse samples is key to inclusive
precision medicine.9 For example, the triple-negative
breast cancer subtype, defined by the absence of ER,
PR, and ERBB2 (formerly HER2 or HER2/neu) expression, accounts for 10% to 20% of all breast cancer cases,
with an uneven distribution among ethnicities.10 In African American women, triple-negative breast cancer
is nearly twice as prevalent as it is in European Americans, with some studies reporting triple-negative subtypes in up to 40% of all breast cancer cases among
premenopausal African Americans.10,11
Consistent with this observation, epidemiological
studies have shown substantial differences in rates of
breast cancer mortality between racial groups.5,8,12 For
example, premenopausal African American women
have a lower 5-year breast cancer–related survival rate
than European American women (79.0% vs 91.0%, respectively), placing them at a higher risk of mortality
despite an overall lower incidence of breast cancer.12
This disparity may be due to the increased frequency
of triple-negative breast cancer observed in African
American women who typically present at a later disease stage and are more likely to have lymph-node metastases at similar tumor sizes.13
Study findings have revealed that Hispanic women
have a higher prevalence of ER-negative tumors compared with non-Hispanic women (36.2% vs 22.7%;
P = .05) and an unexpectedly high proportion of
ERBB2-positive tumors compared with non-Hispanic women (31.9% vs 14.3%; P < .01).14 These findings,
coupled with more recent data, suggest that considerable molecular differences in cancers may exist in
various ethnic groups.10 Because molecular features
can be used for predicting cancer prognosis and therapeutic response, research focused on understanding
the molecular features of cancers occurring in ethnic
minorities is important.
Purpose
To address the lack of tissue-based studies for ethnic
minorities with cancer, the Biospecimen/Biobanking
Geographic Management Program (BMaP) was initiated by the National Cancer Institute’s Center to Reduce Cancer Health Disparities. BMaP coordinated
regional efforts focused on health disparities in cancer research, training, and care and was organized
into 6 geographical regions within the United States.
BMaP region 3 (BMaP-3), which makes up the south384 Cancer Control
eastern region, consists of Alabama, Florida, Georgia,
Louisiana, Mississippi, and Puerto Rico, and it included
9 partner institutions that created a collaborative network. A key goal for BMaP-3 was to develop a stateof-the-art network that could provide a foundation and
infrastructure to adequately and continuously supply
high-quality, multiethnic, human biospecimens for
cancer research. Since 2015, the National Cancer Institute has reorganized and consolidated the regions into
6 regions, and the institutions previously in region 3
are now, at the time of publication, in Geographical
Management of Cancer Health Disparities region 2.15
In 2014, members of the participating BMaP-3 institutions completed a comprehensive assessment tool
that revealed a diversity of samples by race/ethnicity
and cancer site were available as formalin-fixed, paraffin-embedded (FFPE) tumor samples or fresh-frozen
samples (from 6 of 9 institutions).16 In addition, 5 of the
institutions had a tissue advisory/steering committee,
thus providing the necessary infrastructure and expertise to build a network approach. This provided the basis for the design of a BMaP-3 pilot study to test the
feasibility of developing and implementing a regional,
web-based database of available biospecimens and
collecting FFPE blocks to create unique tissue microarrays (TMAs) for health disparities in cancer research.
This pilot study represents a regional team, science-oriented approach for constructing and validating
multi-institutional breast cancer TMAs from a racially
and ethnically diverse population. Using the construction and validation of a breast cancer TMA as an example, we illustrate how a regional team, science-oriented approach can effectively leverage the tissue and
database resources of multiple institutions to facilitate
tissue-based studies for a minority cohort — a difficult
challenge for single institutions alone.
Materials and Methods
The research conducted in this study was performed
with tissue samples and patient data unlinked from patient identifiers. Approval or waiver of informed consent was obtained by the Institutional Review Boards
of the 6 institutions contributing FFPE biospecimens
and data to support this pilot study.
Collection and Management of Data
Cancer registry coding discrepancies were harmonized
by adopting a standardized coding for each variable
and systematically replacing prior coding with current coding using software R v2.15.2 (R Foundation for
Statistical Computing, Vienna, Austria). Patient cases
were removed from the data sets for any of the following reasons: younger than 18 years of age at diagnosis
(n = 1), tumor marker and site-specific factor did not
agree for either ER or PR status (n = 157), had a Spanish surname and unknown “Spanish Hispanic” variOctober 2016, Vol. 23, No. 4
able (n = 17), or assigned a study identification number
that already existed (n = 1). The original site-specific
factor 16 was captured after the year 2010; therefore,
the site-specific factor was manually derived from the
tumor marker 1 ER assay, tumor marker 2 PR assay,
and ERBB2 variables, with assignments of either HR+/
ERBB2+, HR+/ERBB2–, PR–/ER–/ERBB2+, or PR–/ER–/
ERBB2–. If the original site-specific factor 16 disagreed
with the derived site-specific factor, then the entry was
deemed unreliable and was deleted (n = 19).
Multi-Institutional, Web-Based Database
To ensure no changes were made to the data set originally merged by the biostatistician, the web-based
data set was exported and a comparison of the webbased data set against the original merged data set
was executed. Counts were also verified between
both data sets. The aggregated data were posted on
a website (http://labpages.moffitt.org/bmap3/Cancer%20Database/Cancer_Database.html [access restricted]) in the form of an output of descriptive statistics generated by SPSS (IBM, Armonk, NY). BMaP-3
public access database (PAD) has logging features enabled so that, in the event of improper data modification, the user account responsible, the data that were
changed, and the time of the change can be identified
and corrected in a timely manner. The governance
model for the BMaP-3 PAD consists of 3 hierarchical
user-access levels (administrator, data manager, and
investigator), ranging from full to search access.
Archival Case Selection and Tissue Microarray
Construction
Each archival tissue sample was categorized according
to tumor expression of ER, PR, and ERBB2. For each
case, optimal areas for inclusion as cores in the TMA
were marked on the slides stained with hematoxylin
and eosin by pathologists. When sufficient tumor tissue was available, 3 or fewer 1-mm cores from each selected donor block were punched and put into 8 TMAs
60 × 10 mm in size using Tissue Arrayer (Beecher Instruments, Sun Prairie, WI). Each TMA also included
1 sample of normal breast tissue. Cores from kidney,
spleen, and tonsil, as well as melanoma, human breast
cancer lines (MCF-7 and MDA231), human prostate adenocarcinoma cells (LNCaP), and human ovarian carcinoma
cells (SK-OV-3), were included as reference cells/tissues.
Immunohistochemical Staining and Evaluation
All TMA slides were stained with hematoxylin and
eosin and processed for whole-slide imaging. Each
TMA slide was scanned using ScanScope XT (Aperio,
Vista, CA) with a 20×/0.8 numerical aperture objective
lens at a rate of 10 minutes per slide. Image analyses
for stained TMAs were performed using Nuclear v9.1
(Aperio) to segment nuclei of various intensities. The
October 2016, Vol. 23, No. 4
captured hematoxylin and eosin stained images were
made available to investigators at all participating institutions. The biomarker status obtained from cancer
registry data was validated with immunostains for estrogen receptor (ER), progesterone receptor (PR), and
erb-b2 receptor tyrosine kinase 2 (ERBB2) using the
following antibodies from Ventana Medical Systems
(Tucson, AZ): 790-4324 CONFIRM anti-ER (SP1), 7902223 CONFIRM anti-PR (1E2), and 790-100 PATHWAY
anti-HER2/Neu (4B5). Antigen retrieval and incubation times were optimized for each antibody as follows:
60-minute retrieval for ER and PR antibodies; 30-minute retrieval for ERBB2 antibodies; 32-minute incubation for the ER antibody; and 16-minute incubation for
the PR and ERBB2 antibodies.
The Ventana Benchmark XT (Tucson, AZ) platform
was used for all immunohistochemistry analyses. The
Allred scoring system was used to score the ER and PR
stains.17 ERBB2 stains were scored in accordance with
guidelines from the College of American Pathologists,
which defines ERBB2-positive status by immunohistochemistry as protein overexpression.18 The HR rules
for the categorization of cases were: if either ER or PR is
positive, then HR+; if both are negative, then HR–; and if
both ER and PR are missing, or if one is missing and the
other is negative, then it was labeled as “missing.”
Results
Building an Infrastructure
To ensure equitable governance for issues concerning
human biospecimen research sponsored by BMaP-3, a
Tissue Advisory Board was formed with representatives
from all 9 BMaP-3 institutions. A collaboration agreement, signed by members of each of the 9 partner institutions, allowed data and tissue sharing between the
partner institutions regardless of the ability of any individual institution to contribute samples. Once the infrastructure was established, the goal was to evaluate the
feasibility of collecting FFPE tissue blocks and associated data and creating a web-based database and TMAs.
The first step in this pilot study was to choose a tumor type for the TMA. The BMaP-3 investigators chose
breast cancer as the highest priority to address health
disparities related to cancer in the southeastern United
States and Puerto Rico. This decision was based on a
variety of factors, including unpublished observations
on the scarcity of breast TMAs from diverse populations described in the literature, lack of commercially
available breast TMAs with associated patient data on
race and ethnicity, the observed rates of incidence and
mortality associated with breast cancer at the 6 contributing cancer sites, and the unpublished results
of a multi-institutional survey of researcher needs
for TMAs.16 In addition, researchers specializing in
breast cancer were surveyed to determine what type
of breast cancer TMA is most needed for research. As
Cancer Control 385
shown in Fig 1, 29 of the 61 respondents (47.5%) expressed greatest need for a TMA based on biomarker
status between different racial/ethnic groups, followed
by a TMA based on 1 histological type of breast tissue in different racial/ethnic groups (26%), and a TMA
based on 1 tumor, node, and metastasis pathology
stage group in different racial/ethnic groups (14%).
Collection and Management of Cancer
Registry Data
Deidentified American College of Surgeons cancer registry data from each institution served as the basis for
the web-based database and for the selection of cases
to be included in the TMA. Requests for data from cancer registries at each of the 9 partner institutions specified the following criteria for selecting cases: date range,
2001 to 2011 (to avoid using biospecimens that might
be needed for clinical care), age at diagnosis (≥ 18 years
of age), primary breast cancer only (no metastasis), and
first-course surgery at the institution. In addition, because cases from minority populations were a priority
for inclusion in the TMA, members at partnering institutions were asked to specifically retrieve patient data
from African Americans and Hispanics/Latinos. The
17 data elements requested are described in Table 1.
Cancer registry data sets were retrieved from 6 institutions, and a biostatistician harmonized the data
sets. A total of 12,408 entries from 8,279 unique patients with breast cancer pathology were confirmed
after merging the data sets. Deidentified information
about breast cancer cases collected from the different
cancer registries is available in BMaP-3 PAD (https://
apps.mathbiol.org/bmap [access restricted]).
Archival Case Selection and Tissue Microarray
Construction
The data set was further analyzed for cases with invasive ductal carcinoma (IDC; histology selected by the
Tissue Advisory Board) and then cross-tabulated by
race/ethnicity and biomarker status (triple-negative,
TMA based on 1 TNM stage group
in different racial/ethnic groups
TMA of 1 histological type of breast
tissue in different racial/ethnic groups
TMA based on biomarker status
between different racial/ethnic groups
Other
0
5
10 15 20 25 30 35 40 45 50
HR+/ERBB2 –, HR+/ERBB2+, ER –/PR –/ERBB2+). The
demographical and clinical characteristics of the data
from the breast cancer cases obtained from the 6 institutions are presented in Table 2. Because cancer registry standards for ERBB2 were largely adopted from
2006 onward, many of the archival cases (collected
from 2001 to 2005) were categorized as missing for
ERBB2 status. Therefore, the Tissue Advisory Board
decided that the 3 biomarker categories for selecting
cases would be as follows: (1) triple negative, (2) HR+/
ERBB2–, and (3) ERBB2+ (HR+/ERBB2+ and ER–/PR–/
ERBB2+ grouped together). All unique cases of female
Table 1. — Data Elements and Their Descriptions
Data Element
Study identifier
Unique code used by honest broker for
reidentification
Year first seen
Year of first patient contact (inpatient or
outpatient), with the reporting facility for the
diagnosis of the tumor, its treatment, or both
Site primary
Code for the primary site of the tumor being
reported using ICD-O (2nd or 3rd ed.)
Age at diagnosis
Records the age of the patient in complete
years at his or her last birthday before
diagnosis
Sex
Identifies the sex of the patient
Race
Identifies the primary race of the patient
Spanish/Hispanic
origin
Identifies patients of Spanish or Hispanic
origin
Histology
(ICD-O [3rd ed.])
First 4 digits of the morphology code that
describe the tumor/cell type
Behavior
(ICD-O [3rd ed.])a
Code for the behavior of the tumor being
reported
Year of surgery
Year in which a surgical procedure was
performed
Surgery of primary
site, at hospital
Type of surgery performed to breast aimed at
modifying, controlling, removing, or destroying
cancerous tissue
Systemic/surgery
sequence
Records the sequencing of systemic treatment and surgical procedures given as part
of first-line therapy
Surgery/radiation
sequence
Records the sequencing of radiation and
surgical procedures provided as part of firstline therapy
Collaborative stage,
site-specific factor 16
Combinations of ER, PR, and ERBB2 results
Tumor marker 1
ER assay
Tumor marker 2
PR assay
ERBB2
Interpretation of results (not the same as the
laboratory value) for the ERBB2 test used to
determine the status of ERBB2
TNM, pathology,
stage group
The anatomical extent of disease based
on TNM elements of the tumor following
surgery
Respondents, %
Fig 1. — Results of the breast cancer researcher survey (N = 61). A multiinstitutional, web-based poll was distributed to investigators in region 3
(currently region 2) institutions to assess the research interest regarding
different kinds of breast TMAs. Survey responses were anonymous. As
can be seen, the greatest need was for TMA based on biomarker status
between different racial and ethnic groups.
TMA = tissue microarray, TNM = tumor, node, metastasis.
386 Cancer Control
Description
a Behavior
is the fifth digit of the ICD-O (3rd ed.) morphology code.
ICD-O = International Classification of Diseases for Oncology,
TNM = tumor, node, metastasis.
October 2016, Vol. 23, No. 4
IDC were tabulated according to biomarker status
(site-specific factor 16) and by race/ethnicity (Table 3).
Although a large portion of cases were missing data,
this analysis revealed that, in this cohort, African
American women have a higher incidence of triplenegative breast cancer compared with white women,
and Hispanic women have a higher incidence of HRnegative breast cancer compared with non-Hispanic
women. These data, as well as data on breast cancer
cases not included in the TMA, were incorporated into
the BMaP-3 PAD web-based database.
FFPE blocks were requested from participating
Table 2. — Characteristics of Patients at the
Contributing Institutions (N = 8,279)
Variable
No. of Patients, n (%)
Sex
Male
Female
59 (0.72)
8,219 (99.30)
Race
American Indian
2 (0.02)
Asian Indian
1 (0.01)
Asian Other
Black/African American
1 (0.01)
2,012 (24.30)
Filipino
2 (0.02)
Pacific Islander
3 (0.04)
Vietnamese
3 (0.04)
White
5,904 (71.30)
Other
80 (0.97)
Unknown
271 (3.30)
Ethnicity
institutions, and 427 blocks from 427 unique patients
with breast cancer were obtained. Tumor expressions
of ER, PR, and ERBB2 were annotated based on cancer registry data in the following manner: (1) HR+ (ER+,
PR+, or both) and ERBB2–, (2) HR+ (ER+, PR+, or both)
and ERBB2+, and (3) HR– (ER– and PR–) and ERBB2–.
These 427 FFPEs were used to construct the TMA
blocks, of which 8 blocks were constructed. TMA
slides from each of the blocks were stained for ER, PR,
and ERBB2 to confirm the biomarker status of the samples. Fig 2 shows a sample representation of tumors
from each of the 3 types of breast TMAs (ERBB2+,
ERBB2–, and triple negative) for each stain. Thereafter,
the cancer registry data were reviewed, and 4 samples
originally classified as triple negative were reclassified
as either ERBB2+ or HR+.
Several discrepancies were found between the
cancer registry data and the TMA immunohistochemistry results. Specifically, for all racial/ethnic groups,
the biomarker status obtained from the cancer registry data was 100% in concordance to the staining results for ERBB2– cases. However, this was not true for
the triple-negative cases (86.8% and 85.7% rates of accuracy for African Americans/whites and Hispanics,
respectively), and concordance was much lower in
the case of ERBB2+ staining (43.6% and 30.6% for African Americans/whites and Hispanics, respectively).
This finding could be due to different factors such as
tumor heterogeneity or loss of antigenicity (eg, some
donor blocks were several years old).19 These stains
were scanned and quantified, and links to the images
were made available upon request. Detailed information on the samples included on the TMA is posted on
http://labpages.moffitt.org/bmap3/Cancer%20Database
/Cancer_Database.html.
Cuban
43 (0.52)
Dominican Republic
7 (0.08)
Discussion
Mexican
24 (0.29)
Puerto Rican
248 (3.00)
South/Central American
66 (0.80)
Spanish (not otherwise specified)
368 (4.40)
This pilot study successfully accomplished the following goals:
• Creation of a regional infrastructure with an organizing framework for communications, processes
for memorandums of understanding and Institutional Review Board approval from multiple institutions, and an established Tissue Advisory Board
and related guidelines
• Processes for retrieval and synchronization of data
from multiple cancer registries into a single data set
• Development of a collaborative model for optimizing TMA design considerations
• Development and implementation of a web-based
database of cancer registry cases
All of these accomplishments demonstrate the
overall feasibility of creating a unique, centralized,
public resource for multiethnic biobanking and the
collection of biospecimens. Given the large number
of cases included in the web-based database and the
Other Spanish
9 (0.12)
Non-Spanish
7,370 (89.00)
Spanish surname only
Unknown
2 (0.02)
142 (1.70)
Age, y
< 25
23 (0.28)
25–34
222 (2.70)
35–44
1,126 (13.60)
45–54
2,217 (26.80)
55–64
2,267 (27.40)
65–74
1,588 (19.20)
≥ 75
October 2016, Vol. 23, No. 4
836 (10.10)
Cancer Control 387
Table 3. — Site-Specific Factor 16 for Invasive Ductal Carcinoma by Race and Ethnicity for Women in All Institutions Combined
HR+/ERBB2–
HR+/ERBB2+
PR–/ER–/ERBB2–
PR–/ER–/
ERBB2+
Missing
Totals
African American
169 (15)
58 (5)
130 (11)
33 (3)
755 (66)
1,145
White
585 (18)
91 (3)
163 (5)
43 (2)
2,295 (72)
3,177
Race, n (%)
Other/missing
Totals
99 (36)
15 (6)
58 (21)
26 (9)
77 (28)
275
853 (19)
164 (3)
351 (8)
102 (2)
3,127 (68)
4,597
Ethnicity, n (%)
Hispanic
128 (25)
21 (4)
97 (19)
28 (6)
234 (46)
508
Non-Hispanic
711 (18)
139 (3)
251 (6)
73 (2)
2,835 (71)
4,009
Other/missing
Totals
14 (17)
4 (5)
3 (4)
1 (1)
58 (73)
80
853 (19)
164 (3)
351 (8)
102 (2)
3,127 (68)
4,597
amount of TMAs, this model demonstrates that no institution could have accomplished these outcomes
alone. A primary benefit of this approach is that members of all the partner institutions are eligible to access
the data and tissue resources regardless of their capabilities to contribute to the database or TMA.
The first step in this pilot study was to select a
tumor type for creation of the TMAs. We chose to focus on breast cancer as the highest priority to address
health disparities related to cancer in the southeastern
United States and Puerto Rico for a variety of reasons.16
This decision was based on1,10,13,14,20-22:
• Analysis of the literature on TMAs from patients
with different racial/ethnic backgrounds showing
a scarcity of breast TMAs from diverse populations
• Lack of commercially available breast TMAs that
provide associated data on race and ethnicity
• Observed incidence and mortality rates associated
with breast cancer at each of the 6 contributing
cancer sites by race/ethnicity
• Data from the comprehensive assessment tool on
FFPE biospecimens for 6 cancer types available for
study at the 9 study institutions
• Survey results of researcher needs for TMAs
This pilot study demonstrated the feasibility of collecting cancer registry data for more than 12,000 breast
cancer cases and creating a web-based database. We
were successful in creating a racially/ethnically diverse
TMA from FFPEs representing 427 breast cancer cases.
For validation of the TMAs, we focused on studying
the characteristics of IDC among different racial/ethnic
groups because the investigators from the BMaP3 institutions identified these as an important area of
investigation, given the known differences in incidence
of triple-negative IDC among diverse populations.
The results of the biomarker analysis showed a
higher incidence rate of triple-negative breast cancer
among African American women compared with white
women and a higher incidence rate of HR-negative
388 Cancer Control
Biomarker
Status
Antibodies
ERBB2
PR
ER
Triple
negative
ERBB2+
ERBB2–
Fig 2. — Sample representation of immunostaining according to
biomarker status. TMA slides were immunostained with anti-ER,
anti-PR, and anti-ERBB2 antibodies.
ER = estrogen receptor, ERBB2 = erb-b2 receptor tyrosine kinase 2,
PR = progesterone receptor, TMA = tissue microarray.
breast cancer among Hispanic women compared with
non-Hispanic women. These observations are consistent with the literature.10,14 However, our analysis is
compromised by a large amount of missing data. Hines
et al14 showed that breast cancer among Hispanic women comprises a distinct spectrum of tumor subtypes
compared with non-Hispanic white women. In that
study, a diverse TMA based on biomarkers was created, allowing researchers to study potential molecular
differences in triple-negative breast cancer in different
racial/ethnic groups.14
Although most institutions had compatible bioinformatics systems for the management of biospecimen
data, these systems were used for fresh frozen samples.
No single institution had a direct database for associated FFPE tissue data; thus, cancer registry data and pathology reports were the sources of data for cases likely to have tissues stored at the respective institution.
When needed, data would have had to be abstracted
October 2016, Vol. 23, No. 4
from pathology reports — which was beyond the resources of this project. Our findings suggest that the
idea of synchronizing pathology reports with cancer
registry data should be pursued because pathology reports offer more complete and accurate information
on biomarker data than is currently provided by a cancer registry.
Conclusions
We have created a model for collaboration. Through
the contribution of formalin-fixed, paraffin-embedded blocks from the Minority Biospecimen/Biobanking Geographic Management Program for region
3 (BMaP-3) institutions, this project facilitated the
creation of a robust research tool that would not be
readily available if each institution worked independently. Moreover, each institution benefited from the
collaboration and from the team approach to tackle a
research/clinical issue common to all team members.
It is expected that investigators at the partner institutions will utilize the web-based database on an ongoing basis for data analysis. This database is freely
available with open access for the BMaP-3 investigators. We fully expect that the model and process for
the work described herein could be applied and exported to other multi-institutional networks focused
on a participatory approach to biobanking.
We also demonstrated the feasibility of creating
tissue microarrays from racially and ethnically diverse
populations through the BMaP-3 partnership. This
work was made possible by the infrastructure of this
partnership, and it can serve as a model for other multiinstitutional networks focused on research utilizing
human biospecimens. To date, these tissue microarrays of invasive ductal carcinoma have been released
for 5 projects at 5 different institutions.
Acknowledgments: The authors would like to
thank Ivana Sehovic and Zena Sayegh for managing
and constructing the BMaP-3 TMA blocks. Marithea
Goberville, PhD, provided editorial support.
Substantial support was provided by Region 3
GMaP/BMaP partner institutions: Emory Winship Cancer Center, Moffitt Cancer Center, Morehouse School
of Medicine, Ponce Health Sciences University, Tulane
University, Tuskegee University, University of Alabama
at Birmingham, University of Mississippi Medical Center, and Xavier University of Louisiana. This work was
supported in part by National Cancer Institute grants
3U56 CA118809-14S2; 3U54 CA153509-02S3 and 3U54
CA153509-03S1 (Moffitt Cancer Center).
2011;127(3):729-738.
3. Partridge EE, Fouad MN, Hinton AW, et al. The deep South network
for cancer control: eliminating cancer disparities through community-academic collaboration. Fam Community Health. 2005;28(1):6-19.
4. Ward E, Jemal A, Cokkinides V, et al. Cancer disparities by race/
ethnicity and socioeconomic status. CA Cancer J Clin. 2004;54(2):78-93.
5. Ademuyiwa FO, Edge SB, Erwin DO, et al. Breast cancer racial
disparities: unanswered questions. Cancer Res. 2010;71(3):640-644.
6. Evens AM, Antillón M, Aschebrook-Kilfoy B, et al. Racial disparities
in Hodgkin’s lymphoma: a comprehensive population-based analysis. Ann
Oncol. 2012;23(8):2128-2137.
7. Fedewa SA, Ward EM, Stewart AK, et al. Delays in adjuvant chemotherapy treatment among patients with breast cancer are more likely
in African American and Hispanic populations: a national cohort study
2004-2006. J Clin Oncol. 2010;28(27):4135-4141.
8. Tamimi RM, Hughes ME, et al. Racial and ethnic differences in breast
cancer survival: mediating effect of tumor characteristics and sociodemographic and treatment factors. J Clin Oncol. 2015;33(20):2254-2261.
9. Tian Q, Price ND, Hood L. Systems cancer medicine: towards realization of predictive, preventive, personalized and participatory (P4) medicine.
J Intern Med. 2012;271(2):111-121.
10.Carey LA, Perou CM, Livasy CA, et al. Race, breast cancer
subtypes, and survival in the Carolina Breast Cancer Study. JAMA.
2006;295(21):2492-2502.
11. Kohler BA, Sherman RL, Howlader N, et al. Annual report to the
nation on the status of cancer, 1975-2011, featuring incidence of breast
cancer subtypes by race/ethnicity, poverty, and state. J Natl Cancer Inst.
2015;107(6):djv048.
12. U.S. Department of Health and Human Services; National Institutes
of Health; National Cancer Institute. Surveillance, Epidemiology, and End
Results (SEER) 2010. http://seer.cancer.gov/. Accessed March 24, 2016.
13. McBride R, Hershman D, Tsai WY, et al. Within-stage racial differences in tumor size and number of positive lymph nodes in women with
breast cancer. Cancer. 2007;110(6):1201-1208.
14. Hines LM, Risendal B, Byers T, et al. Ethnic disparities in breast
tumor phenotypic subtypes in Hispanic and non-Hispanic white women.
J Womens Health (Larchmt). 2011;20(10):1543-1550.
15. National Cancer Institute. Geographical Management of Cancer
Health Disparities (GMaP). Updated August 1, 2016. http://www.cancer.
gov/about-nci/organization/crchd/inp/gmap. Accessed August 19, 2016.
16. Wells KJ, Lima DS, Meade CD, et al. Assessing needs and assets
for building a regional network infrastructure to reduce cancer related
health disparities. Eval Program Plann. 2014;44:14-25.
17. Allred DC, Carlson RW, Berry DA, et al. NCCN task force report: estrogen receptor and progesterone receptor testing in breast cancer by immunohistochemistry. J Natl Compr Canc Netw. 2009;(7 suppl 6):S1-S21.
18. Wolff AC, Hammond ME, Schwartz JN, et al. American Society
of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast
cancer. J Clin Oncol. 2007;25(1):118-145.
19. Miller RT, Swanson PE, Wick MR. Fixation and epitope retrieval in
diagnostic immunohistochemistry: a concise review with practical considerations. Appl Immunohistochem Mol Morphol. 2000;8(3):228-235.
20. Wu Y, Sarkissyan M, Elshimali Y, et al. Triple negative breast tumors
in African-American and Hispanic/Latina women are high in CD44+, low in
CD24+, and have loss of PTEN. PLoS One. 2013;8(10):e78259.
21. Lindner R, Sullivan C, Offor O, et al. Molecular phenotypes in triple
negative breast cancer from African American patients suggest targets for
therapy. PLoS One. 2013;8(11):e71915.
22. Winter JL, Stackhouse BL, Russell GB, et al. Measurement of PTEN
expression using tissue microarrays to determine a race-specific prognostic marker in breast cancer. Arch Pathol Lab Med. 2007;131(5):767-772.
References
1. Lisovicz N, Johnson RE, Higginbotham J, et al. The Deep South
Network for cancer control. Building a community infrastructure to reduce
cancer health disparities. Cancer. 2006;107(8 suppl):1971-1979.
2. Ooi SL, Martinez ME, Li CI. Disparities in breast cancer characteristics and outcomes by race/ethnicity. Breast Cancer Res Treat.
October 2016, Vol. 23, No. 4
Cancer Control 389
Data suggest that immediate action
must be taken to improve the time
between diagnosis and treatment
for disadvantaged patients.
Lily. X-Ray image courtesy of Bryan Whitney. www.x-rayphotography.com.
Determinants of Treatment Delays Among Underserved
Hispanics With Lung and Head and Neck Cancers
Evelinn A. Borrayo, PhD, Katie L. Scott, PhD, Ava R. Drennen, PhD, Tiare MacDonald, PhD,
and Jennifer Nguyen, MS
Background: Evidence is lacking to explain the reasons why treatment is delayed among disadvantaged Hispanic patients diagnosed with lung and head and neck cancers. Data indicate that treatment delays beyond
46 days increase the risk of death for individuals with these cancers.
Methods: A mixed-methods design was used to explore determinants of treatment delays by analyzing data
from the medical records of 40 Hispanic patients and data from interviews with 29 Hispanic patients, caregivers, health care professionals, and patient navigators from a safety-net hospital.
Results: Of the 40 Hispanic patients, 35% initiated treatment 46 days or more after being diagnosed, but
women experienced longer delays than men (average of 48 days). Women with few comorbid diseases (≤ 4)
were more likely to experience treatment delays. Institutional-related determinants at publicly funded
hospitals appear to delay treatment for patients at the safety-net hospital, and patient-related determinants common to underserved patients (eg, no health insurance coverage) are likely to further contribute
to these delays.
Conclusions: Delayed treatment is associated with poor outcomes and low rates of survival in patients with
lung and head and neck cancers. Therefore, action should be taken to improve the time between diagnosis
and the initiation of treatment for disadvantaged Hispanic patients.
From the Department of Psychology (EAB, TM, JN), Colorado
State University, Colorado School of Public Health, University
of Colorado, Denver, Colorado, Department of Neurology (KLS),
Spectrum Health Medical Group, Grand Rapids, Michigan, and
the Department of Behavioral Health (ARD), Salud Family Health
Centers, Fort Collins, Colorado.
Submitted May 5, 2016; accepted July 28, 2016.
Address correspondence to Evelinn A. Borrayo, PhD, Department
of Psychology, Colorado State University, 1876 Campus Delivery,
Fort Collins, CO 80523. E-mail: [email protected]
No significant relationships exist between the authors and the
companies/organizations whose products or services may be referenced in this article.
This work was supported by a grant from the National Cancer
Institute at the National Institutes of Health (U01, CA114604). Its
contents are solely the responsibility of the authors and do not
necessarily represent the official views of the National Cancer
Institute.
390 Cancer Control
Introduction
In the United States, lung cancer accounts for more
deaths than any other cancer in both men and women.1 Among Hispanics, lung cancer is the leading
cause of cancer-related death for men and the secondleading cause for women.2 Those with lung cancer are
also at increased risk for developing secondary neoplasms of the head and neck and vice versa.3,4 Lung
and head and neck cancers are largely induced by tobacco use, which accounts for 87% and 70% of lung
cancer–related deaths for men and women, respectively.5 Approximately 75% of head and neck cancers
are attributed to tobacco use, and a 30-fold increased
risk is seen among those who smoke and drink alcohol.1,6 Moreover, those with lung and head and neck
October 2016, Vol. 23, No. 4
cancers who smoke cigarettes and drink alcohol are
at increased risk for developing comorbid conditions,
such as chronic bronchitis, emphysema, and cardiovascular disease.7 Although Hispanics have lower incidences of and mortality rates from lung and head
and neck cancers than non-Hispanic whites, disparities in timely and quality treatment remain for Hispanics — poverty is typically cited as the explaining
factor.1 Mounting evidence demonstrates that worse
outcomes (eg, survival, quality of life) for ethnic minorities with cancer are also best explained by their
low socioeconomic status (SES).1,8 Similarly, inequalities exist in properly treating medically underserved
patients (eg, low-income, uninsured) with lung and
head and neck cancers.9-11
In a systemic review and meta-analysis of socioeconomic inequalities and lung cancer treatment, Forrest et al9 found that lower SES was associated with a
statistically significant reduced likelihood of receiving
any treatment — particularly surgery and chemotherapy. These SES inequalities could not be accounted for
by differences in stage at presentation or by differences
in health care systems (ie, publicly funded vs private
health care systems).9 Choi et al10 conducted a longitudinal study to examine disparities as predictors of
survival among patients with head and neck cancers
and found that low income, low education, and advanced age were significant independent predictors of
poor survival (ie, 5-year overall, cancer-specific, and
disease-free survival rates; P < .001). Similarly, Yang
et al11 found in a population-based study that significant independent predictors of worse survival rates
among patients with lung cancer were older age and
severe poverty. The effects of ethnicity were statistically significant: A higher percentage of Hispanic patients
resided in the highest poverty areas, were less likely to
receive radiotherapy, and had shorter survival rates if
they smoked compared with their non-Hispanic white
counterparts.11
Treatment Initiation
More than 10 years ago, the Institute of Medicine recommended improving the timeliness of treatment by
reducing wait times that can potentially result in harmful delays for patients.12 Timeliness in the treatment of
lung and head and neck cancers is important, because
delaying the start of treatment has been associated
with disease recurrence,13-16 lower rates of survival,17
and poor patient outcomes (eg, distress).18,19
Using the US National Cancer Database, Murphy
et al20 found that the interval from diagnosis to the
start of treatment has been increasing since 1985 for
Americans diagnosed with head and neck cancers.
Their examination of predictors found that patients
with advanced-stage disease who underwent chemoradiotherapy, were treated at academic facilities, and
October 2016, Vol. 23, No. 4
who received transitional care had the greatest increases in time to treatment initiation.20 In a follow-up
study using the same database, Murphy et al17 found
that the time to treatment initiation of 46 to 52 days
was associated with a significant increased risk of
death — a risk that was consistently detrimental beyond 60 days. After examining covariates, the researchers also found that the effect of time to treatment initiation on mortality risk was greater for
early-stage (stage I/II) than advanced-stage disease
(stage III/IV) and for radiotherapy and surgery than
for chemoradiotherapy.17
To assess the impact of delayed treatment for lung
cancer on an international level, Vinas et al21 conducted a systemic review of multinational studies and
found that long delays were predictive of better survival rates than shorter delays, but patients who experienced longer delays had fewer symptoms — possibly
explaining a better prognosis. Instead, short delays in
lung cancer treatment had a negative prognostic meaning because urgent treatment was provided to patients
with more advanced disease.21
Patients with lung and head and neck cancers increasingly experience longer delays between their
diagnoses and subsequent treatment.20,21 Delays are
more prevalent for patients with early-stage disease,
thus increasing their disease progression, their wait
times (if candidates for surgical treatment), and their
level of distress.18,19,22-25 Instead, shorter delays are experienced by patients with more advanced-stage disease for whom treatment is urgently initiated without
clear benefits in their disease progression or rate of
survival.17,21 A paucity of evidence explains what factors drive the treatment delays experienced by patients
with lung and head and neck cancers. Murphy et al20
contend that the delay in the start of treatment is due
to increases in more sophisticated, pretreatment diagnostics, complex multimodal therapies, and transitions
in care for patients treated at academic cancer centers.
However, these factors are not as likely to explain treatment delays for patients treated at community-based
hospitals.20
Community-Based Facilities
Few studies have compared the intervals from diagnosis to treatment among academic and public hospitals that treat head and neck cancers; however, among
those that have, significantly longer treatment delays
have been observed at public hospitals.26,27 In a study
by Yorio et al,26 age, sex, race, and type of insurance
coverage were not significantly associated with a longer time to treatment. The single significant factor they
found for the delay of lung cancer treatment was receiving that care at a public hospital. Patel et al28 suggest that the factors that affect the timely delivery of
cancer treatment are likely institution-related factors
Cancer Control 391
that generally apply to public institutions, including
availability of specialty services, adequacy of equipment, timely pathological analysis, and lack of imaging
and surgical procedures — this is particularly true for
patients with head and neck cancers.
Patient Factors
Less is known about patient-related factors that might
contribute to delays in cancer treatment, but they are
likely related to low SES, limited access to transportation, lack of paid leave from work, language barriers,
comorbid medical conditions, and homelessness.28
Purpose
To our knowledge, no studies have investigated the
factors at the institutional and patient levels that are
likely to contribute to treatment delays among underserved Hispanic patients with lung and head and
neck cancers treated at community-based hospitals,
let alone studies that have focused on underserved
Hispanic patients who experience high rates of morbidity from these cancers. We conducted this exploratory study to better understand the institution- and
the patient-level determinants associated with the
timely initiation of cancer treatment among underserved Hispanic patients diagnosed with lung and
head and neck cancers.
variables that procure or impede a patient’s initiation
of treatment (eg, health insurance, income); and predisposing factors include personal propensities to access treatment (eg, age, sex, language).30,31
Measures
Data were extracted from the medical records of Hispanic patients with a primary diagnosis of lung cancer
or head and neck cancer treated at a “safety-net” hospital in Colorado across a 1-year period. The patient data
included sociodemographic variables (eg, age, sex), diagnosis (eg, cancer type, stages I–IV), cancer treatment
(eg, chemotherapy, surgery), number of comorbid diseases (using the Charlson Comorbidity Index), date
of cancer diagnosis (by diagnostic biopsy), and date
when the patient began any type of cancer treatment.32
The interval between the initial diagnosis and the start
of treatment was calculated as the number of days between the date when patients were diagnosed and the
date when they began their first curative treatment. For
patients diagnosed at the time of resection and surgery
was their first treatment, the number of days was coded as 1 day.
Study Design
A triangulation, mixed-method design (ie, quantitative
and qualitative data linked to the literature) was used
for breadth and depth of understanding and corroboration from multiple angles to determine the initiation
of cancer treatment among the study population.29 We
collected quantitative data from medical records to assess the magnitude of the influence of patient variables
and diagnostic factors on treatment initiation as well
as qualitative research to explore stakeholder perspectives and other influential factors not captured by the
data in the medical records. One-on-one interviews
were also conducted with patients who had lung cancer and head and neck cancers and their health care
professionals (HCPs), while focus-group interviews
were conducted with the caregivers and patient navigators who assisted the study patients.
Data Analysis
For medical record data, a stepwise, multiple regression method was used to evaluate whether predisposing, enabling, and need factors were necessary to predict the range of days between initial diagnosis and
initiation of treatment. Data were centered by converting age, cancer stage at diagnosis, and comorbid diseases into z scores. Regression assumptions and diagnostics were satisfactory, except for homoscedasticity,
which led to the removal of a single influential case.
Ethnographical content analysis was utilized to extract
relevant themes from the semi-structured and focusgroup interviews.33
The goal of the analysis was to identify factors that
detail the delays in receiving cancer treatment from the
perspective of patients, caregivers, HCPs, and patient
navigators. Five analysts were trained and employed
in this study to follow the content analysis methods
of Krippendorff.34 All analysts independently coded
each transcript and then met to corroborate the codes,
broaden the subject categories, and identify quotes to
illustrate the themes.
Theoretical Framework
The Behavioral Model for Vulnerable Populations was
used as the guiding theoretical framework.30,31 The
model proposes that need, enabling, and predisposing factors facilitate or block the use of health services
by vulnerable populations. Need factors include the
vulnerabilities that make patients necessitate medical
treatment (eg, cancer type and stage, comorbid diseases); enabling factors include social or environmental
Participants
The patients included in this study were treated at a
safety-net hospital that serves impoverished community
neighborhoods.
Medical Records: Medical records were reviewed
for 53 Hispanic patients with a primary diagnosis of
lung or head and neck cancer; of those, 40 patients
had medical records of the number of days from diagnosis to treatment. Table 1 displays the characteristics
Methods
392 Cancer Control
October 2016, Vol. 23, No. 4
for all 40 patients.
Patients were an average
of 57 years of age (standard deviation [SD] 16.3; range, 23–88);
57% were diagnosed with head
and neck cancers and 43% were
diagnosed with lung cancer. The
average number of days from
diagnosis to treatment was 40
(SD 33.2; range, 1–141); 65% received treatment in less than 42
days, 18% between 46 and 59
days, and 17% received treatment after more than 60 days.
The number of comorbidities in
the study patients ranged from
2 to 17 conditions (mean [SD]
= 5 [3.84]), with the most common being hypertension (58%),
chronic obstructive pulmonary disease (43%), tobacco use
(40%), and depression (33%).
Men and women were significantly different in their cancer
stage at diagnosis: Men were
more likely to have advanced
cancer (79%) and women were
more likely to have less-advanced cancer (stages I/II; 69%).
Moreover, men were significantly more likely to have received
chemotherapy than women
(65% vs 31%, respectively).
Interviews
In-depth interviews were conducted in 29 participants: 4 with
lung cancer, 5 with head and neck
cancers, 6 caregivers, 7 HCPs,
and 7 patient navigators. The patients interviewed were an average of 62 years of age (SD 9.64;
range, 49–75). Five were men
and 4 were women. Five patients
were diagnosed with head and
neck cancers and 4 with lung
cancer; 3 patients were diagnosed at stage I/II disease and
6 at stage III/IV disease. Of the
6 caregivers, two cared for patients with head and neck cancers and 4 cared for patients with
lung cancer. The ages of the caregivers ranged from 34 to 59 years
(mean [SD] = 46.67 [10.31]). One
was a man and 5 were women.
October 2016, Vol. 23, No. 4
Table 1. — Select Characteristics of Study Patients
Variable
Men
(n = 23)
Women (n
= 17)
All Participants
F
(N = 40)
Statistic
Mean (SD)
Age, y
61 (14.5)
52 (17.6)
57 (16.3)
Diagnosis to treatment delay, dª
34 (23.1)
48 (42.8)
40 (33.2)
1.76
5 (4.4)
4 (2.9)
5 (3.8)
0.51
n (%)
n (%)
n (%)
X²
United States
13 (57)
9 (53)
22 (55)
Latin America
10 (43)
8 (17)
18 (45)
No. of comorbid diseases
2.95
Sociodemographic Characteristics
Place of Birth
0.05
Language
0.06
English
14 (58)
11 (56)
25 (62)
Spanish
9 (42)
6 (44)
15 (38)
9 (39)
7 (41)
16 (40)
14 (61)
10 (59)
24 (60)
Marital Status
Married
Not married (eg, single, widow)
4.36
Insurance Coverage
0.27
Insured (Medicaid/Medicare)
10 (43)
6 (44)
16 (40)
Uninsured (no health insurance)
13 (57)
11 (56)
24 (60)
12 (57)
6 (40)
18 (50)
6,240–13,908
3 (14)
6 (40)
9 (25)
14,248–33,192
6 (29)
3 (20)
9 (25)
Head/neck
11 (48)
12 (71)
23 (57)
Lung
12 (52)
5 (29)
17 (43)
Income, $
< 6,144
4.25
Cancer Diagnosis
Cancer Site
2.07
Cancer Stage at Diagnosis
13.38*
I
4 (17)
11 (69)
II
1 (4)
0 (0)
15 (38)
1 (3)
III
3 (13)
3 (19)
6 (15)
IV
15 (66)
2 (12)
17 (44)
Yes
11 (50)
13 (76)
24 (60)
No
11 (50)
4 (24)
15 (40)
Yes
15 (65)
5 (31)
20 (51)
No
8 (36)
11 (69)
19 (49)
Cancer Treatment
Surgery
2.84
Chemotherapy
4.36**
Radiotherapy
1.12
Yes
16 (73)
9 (56)
25 (66)
No
6 (27)
7 (44)
13 (34)
Numbers in the group categories do not sum to the total number of participants because of
missing data for certain variables.
ªAssessed as the number of days from the time patients received a cancer diagnosis until their
first treatment.
*P < .01.
**P < .05.
Cancer Control 393
Results
Medical Records
Predisposing (ie, age, sex), enabling (ie, health insurance), and need (ie, cancer site, stage at diagnosis, comorbid conditions) variables were entered as
predictors of days from diagnosis to the initiation of
treatment. The prediction model contained 4 of the
6 predictors and was reached in 4 steps with 2 variables removed (ie, age, health insurance). As shown in
Table 2, the model was significant (F [4, 36] = 2.58;
P < .05, R: 0.482; R²: 0.233 [adjusted R²: 0.142]). The
variables of sex and comorbid conditions were significant predictors of the time from diagnosis to treatment
initiation. Compared with men, women were significantly more likely to experience more days between
diagnosis and the start of treatment. Patients with fewer comorbid conditions also experienced significantly
more days from diagnosis to treatment initiation than
those with more comorbidities. Specifically, with each
additional comorbid condition, a patient’s time from
diagnosis to treatment initiation decreased by nearly
17 days.
Sex accounted for 9% of the variance and comorbidities accounted for 17% of the variance of diagnosis to treatment initiation (see Table 2). Inspection of
the structure coefficients suggests that women and
patients with fewer comorbidities experienced significantly more days between their initial diagnosis and
the start of treatment. Although we examined the interaction effect of all the predictors in the model, the
interaction alone between sex and comorbidities was
found to be significant (F [2, 21] = 3.68; P = .04). As seen
in the Fig, compared with men, women with fewer comorbidities experienced the longest delay from their
initial diagnosis to the start of cancer treatment.
Interviews
Overall, HCPs and patient navigators had more years of
experiences to draw upon when discussing how various factors influenced the time from initial diagnosis to
treatment initiation among underserved Hispanic patients with lung and head and neck cancers. Thus, patient and caregiver responses were organized around
the responses of their HCPs and navigators.
Need Factors: All participating HCPs conveyed
that lung and head and neck cancers are usually diagnosed at a later stage of disease, thus decreasing survival rates among patients with these cancers. They
recognized that reducing delays in treatment is impor-
100
Estimated Marginal Mean
(Diagnosis to Treatment, d)
The 7 HCPs included a pulmonologist, medical oncologist, oncology nurse, oncology pharmacist, otolaryngologist, pulmonology surgeon, and radiation
oncologist. Five HCPs identified as being white and
2 self-identified as ethnic minorities; 2 were
women and 5 were men. Their time at the safety-net hospital ranged from 9 months to 10 years (mean
[SD] = 5.46 [3.28]).
Of the 7 patient navigators, 5 were women and
2 were men. Six self-identified as Hispanic and 1 as nonHispanic white. Their employment at the safety-net
hospital ranged 2 to 9 years (mean [SD] = 3.43 [2.57]).
Sex
Men
Women
80
60
40
20
0
0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17
Sum of Charlson Comorbidity Status
Fig. — Interaction between the sexes and number of comorbidities.
Table 2. — Coefficients for Predictors of Diagnosis to Treatment Delay (N = 40)
Model
Unstandardized Coefficient
B Value
Standard Error
Standardized Coefficient
β Level
T test
sr ²
Structured
Coefficient
(Constant)
24.87
9.35
—
2.55
—
—
Sex
24.49
12.30
.369
1.99*
.09
.52
Cancer site
12.42
12.04
.187
1.03
.02
–.21
7.88
6.63
.238
1.19
.03
–.27
–16.53
6.07
–.523
.17
–.77
Cancer stage at diagnosis
Comorbid diseases
–2.73**
R = 0.482; R² = 0.233 (adjusted R² = 0.142). Sex coded as 0 = men, 1 = women. Cancer site coded as 0 = head and neck cancer, 1 = lung cancer.
Cancer diagnosis stage coded as 1 = stage 0/1, 2 = stage II, 3 = stage III, and 4 = stage IV.
*P < .05.
**P < .01.
394 Cancer Control
October 2016, Vol. 23, No. 4
tant to reduce the high rates of morbidity from these
cancers. The pulmonologist said:
Lung cancer is a progressive disease and cancer grows.
My fear with patients is always that this is a particular
cancer that can be treated with surgery and cured. If it
advances to the point where it can no longer be taken
care of in time for surgery, then it’s impossible to cure,
and the only thing we can do is give people radiation or
drugs to slow it down, but not cure it.
The HCPs acknowledged lengthy delays in
treatment, similar to what the pulmonology surgeon
reported:
Well, ultimately, the care is good. It’s just the delay from
one step to the next. They’re getting the appropriate
treatments and procedures done — it’s just the delay [of
8 months].
Delays in treatment were viewed as being influenced by systems-level factors. At the systems level,
most patients and caregivers said that a missed diagnosis of cancer by their primary care physician was the
factor that contributed to receiving delayed treatment.
When 1 caregiver explained the reasons her relative
with head and neck cancer experienced a lengthy delay of treatment, she said:
Just maybe look to see if it was cancer before. Before they
thought it was a blister or fever blister. She had it for a
long time, and it was probably growing.
HCPs identified lack of coordinated care as a systems-level factor that contributed to delays in treatment. Although underserved patients receive affordable care at the safety-net hospital, they recognized
that the process of coordinating these patients’ treatment is difficult. HCPs believed that enrolling for
services was an obstacle to patients because of the
amount of paperwork needed and the requirements
that they must meet before the coordination of these
patients’ treatment can begin. The clinical oncology
pharmacist summarized it as such:
Getting them enrolled is the biggest gap. I think that, if
they can get through that hurdle, everything else goes a
lot smoother [to begin their treatment].
HCPs also reported that the coordination of care
for lung and head and neck cancers is complex and
can delay treatment. Medical care from various departments (eg, medical oncology, surgery, radiation) and
specialty clinics (eg, otolaryngology) must be coordinated prior to the start of treatment as well as during
the various procedures necessary for the treatment of
October 2016, Vol. 23, No. 4
these cancers. The oncology nurse also reported on
the complications of coordinating care among the different specialties:
Oh, yeah, particularly the head and neck patients because they have to go between radiation, medical oncology, and otolaryngology, so their care is more complicated.
The complex coordination of care was also believed to result in negative treatment outcomes, as illustrated by the radiation oncologist:
If the patient at [my institution] had surgery that I’m not
aware of, and the patient comes to me, and really there
was a long gap, a long interval … I actually had quite
a few patients when they had surgery who, when they
came to me, their tumor had recurred already.
Related to lack of care coordination was lack of
communication across departments, among HCPs, and
with patients. Communication was seen as lacking between different constituents and related to treatment
delays and to difficulties tracking patients during follow-up or transitional periods. The pulmonology surgeon said:
I think the gaps — the major ones — are the communication between clinics and providers. So, we have a gap
there that results in delays, and sometimes we don’t have
all the information transmitted. It’s only what was written down in the chart. If you don’t look in the right place,
you might not find it. That’s the biggest issue.
Lack of communication between HCPs and patients was also recognized as contributing to delays
in treatment. The patient navigators stated that HCPs
should better communicate with their patients about
the proper expectations of treatment. Patient navigators believed that when the explanation of the HCP is
only verbally communicated, without written instructions or drawn diagrams, then patients forget the instructions given to them — which, in turn, results in
patients delaying treatment or not adhering to treatment instructions.
Enabling Factors: Patients, caregivers, HCPs, and
patient navigators agreed that the enabling factors that
generally apply to procuring or impeding patient access
to health care were also contributors to delays in cancer treatment. Factors related to low SES mentioned by
the study participants included financial barriers. All the
respondents highlighted that patients at the safety-net
hospital generally lacked the finances to cover basic expenses for housing, food, and transportation, as well as
health insurance or, if they had insurance, the ability to
cover their co-pays. Thus, the access barrier most directCancer Control 395
ly related to low SES was lack of health insurance or of
sufficient health insurance to cover the cancer treatment
(eg, radiotherapy) they needed.
Patients who did have health insurance were, in
general, covered through Medicaid or Medicare; few
had sufficient private health insurance. Instead, a majority of patients, similar to other underserved Hispanic patients, had no health insurance, which significantly impeded their access to treatment. The medical
oncologist summarized it by saying:
People [who] do not have good health coverage have a
hard time accessing the system. There are some things
we cannot do for people who do not have good coverage at [this hospital], eg, positron emission tomography, which is used for staging lung cancer to see how
advanced it is.
Delays in proper cancer staging may then lead to delays in timely treatment for these underserved patients.
Due to the significant financial barriers that disadvantaged Hispanic patients at the safety-net hospital face
to initiate and complete their cancer treatment, statefunded programs are available for select patients, such
as the Colorado Indigent Care Program, or some hospitals can assist them by finding resources that could help
them cover some treatment costs. For example, some
patients received assistance with medications they could
not afford, as mentioned by the oncology nurse:
There is an indigent formulary at this hospital, so that
means certain folks [with no financial resources] come
and get certain drugs.
However, the oncology nurse explained that the
process of finding resources for underserved patients
makes the treatment of these patients difficult:
We have our hands tied in a lot of situations or have a
number of hurdles to jump through to provide the best
care that we can for patients, and we absolutely do everything that we can to make sure we are providing
the best care possible.
The HCPs believed that the process of assisting
patients was also a lengthy process that contributed to
treatment delays.
Although the safety-net hospital staff has administrative difficulties in providing care to this underserved
population, these patients also face the hurdles of navigating the health care system themselves. Patients encountered system difficulties throughout the treatment
process, from scheduling appointments to receiving
timely and proper treatment. Navigation hurdles that
delayed treatment tended to exist at the system level, as
the pulmonology surgeon indicated:
396 Cancer Control
The routine times to get appointments in some clinics
can be pretty long. I think that’s why we see 8-month
delays. It’s not surprising when we see it. We see it
frequently.
Patients with cancer may receive care at a hematology/oncology clinic that functions within the overall
hospital structure, which further contributes to treatment delays, as the oncology nurse explained:
The patient can call and get scheduled into the wrong
clinic. There are issues with having central scheduling that handles all the appointments for all the outpatient clinics, as opposed to having someone dedicated
to oncology who knows the patients, understands how
our clinic works, and why patients need to be seen in a
timely fashion.
At the study hospital, the hematology/oncology
clinic does not offer radiation oncology services but
refers patients to a partnering academic cancer center
when patients require radiotherapy. Navigating between separate treatment clinics was problematic and
resulted in further treatment delays, particularly when
care coordination was lacking, as the radiation oncologist explained:
I just had a patient — he had surgery done, he’s late
already, he’s like close to 4 weeks — so I saw the patient.
I did the treatment planning the same day, so I want to
start the treatment really quick. [Results from] computed tomography were not there [with the patient file from
the safety-net hospital].
Treatment delays that result from difficulties navigating the health care system are also patient-dependent, and they generally originate from patients not
understanding their HCPs’ follow-up instructions or
not understanding how the health care system works.
The pulmonologist made the observation:
Part of the problem is the patients don’t understand
how to get where they need to be. Sometimes they need
help filling out forms to get their tests done in a timely
fashion.
Lack of patient understanding is likely linked to
lack of care coordination at the systems-level, and difficulties navigating the health care system then contribute to delays in treatment, as the pulmonology surgeon
explained:
I think the major barriers are delays related to the process of getting from one clinic to the next in our system.
Some of them may be language-related and some of
them are just the way our system has been set up.
October 2016, Vol. 23, No. 4
Predisposing Factors: Personal, social, cultural,
and psychological propensities were noted as predisposing factors that contribute to a patient’s ability to access cancer treatment. Influential personal factors were
related to patient immigration status and included lack
of language proficiency and lack of legal status.
Most Hispanic patients at the safety-net hospital
spoke only Spanish — a barrier to timely treatment:
The patient showed up late with the tumor recurred. …
I think just for the Spanish patients — I think there are
things … they probably had more difficult than the other
patient population. Well, once, for whatever reason, they
missed the appointment; they had [a] language barrier.
Similarly, patients who lacked documentation of
their legal immigration status encountered problems
accessing treatment care, as the oncology pharmacist
explained:
If they [patients] are undocumented and there is a patient-assistance program available through pharmaceutical companies to provide drugs at low cost or free,
undocumented patients don’t qualify.
Language and legal status barriers impeded access to care, and, if they eventually accessed resources,
their treatment was significantly delayed.
Another predisposing factor to timely treatment is
a patient’s social support system. Patients who have a
caregiver or family members to assist them with navigating the medical system and support them through
treatment are more successful at obtaining timely and
proper treatment, as noted by the otolaryngologist’s report of the important role of a caregiver:
I have one man who lost his larynx from laryngeal
cancer, and his sister — his wicked sister — brings him
in and she’s just a riot, but she keeps on this guy like
you can’t believe. She has him here on time, all the time.
An HCP also emphasized how a social support system can help patients overcome other barriers that imped timely treatment:
My Latino patients who have done well have Englishspeaking families who have been here, and then they
bring mom and dad in.
By contrast, patients without caregivers or family members to care for them have a more difficult
experience:
A lot of our patients have no support system. So, if
they get sick, they have no one to help them. And that’s
tough, really tough.
October 2016, Vol. 23, No. 4
Thus, social support appears to impact patients’
timely access and adherence to treatment.
Lack of health literacy was another predisposing
factors that played an important role in whether Hispanic
men experienced treatment delays. We found that these
underserved patients have difficulties understanding
medical information related to their diagnosis and treatment, and, consequently, do not follow-up with their treatment regimen, as explained by the medical oncologist:
Sometimes patients have no idea that they have to come
back. Even though we tell them we want them back,
they have no idea why. … And also there is lack of understanding from the patients of what is required to be
successful when they’re in treatment.
Misconceptions about cancer treatment were also
reported as stemming from lack of health literacy. For
example, when the pulmonologist was asked to expand on how he believed lack of health literacy contributed to treatment delays, he said:
Well, so, I think one of the things I’ve heard from a lot
of immigrants when we talked about surgery for lung
cancer is that they don’t want to have surgery because
they’ve heard that will cause the cancer to spread.
Other similar diagnosis and treatment misconceptions also appeared to contribute to treatment delays.
Mental health propensities were identified as directly and indirectly influencing a patient’s ability to
initiate treatment. Substance abuse was related to treatment delays, which the medical oncologist explained:
Yeah, well, that’s a problem because, number one, they
sometimes have to actually quit these other complications [before treatment], … and, you know, sometimes
that doesn’t happen.
Similarly, emotional distress (eg, depression, anxiety, fear) affects patients’ ability to participate in treatment. The otolaryngologist illustrated:
People who drink have depression. Uh, either it’s
generated by the alcoholism or it’s preceding the alcoholism — where they drink because they’re depressed.
I would think that’s an impediment [to treatment] that
begins, you know, with the patient itself.
Patients and their caregivers also recognized the
role of emotional distress in the patient’s ability to initiate treatment and attend follow-up visits, as 1 woman
with head and neck cancer explained:
Yup, it was the first [round of] chemotherapy, after we
found out. I missed my first — I was scared — I missed
Cancer Control 397
my first appointment because I was scared. Same with
radiation. I missed the first appointment.
Discussion
Our exploratory study findings help to understand the
systems- and patient-level determinants of treatment
delay among underserved Hispanic patients diagnosed
with lung and head and neck cancers. Such contributing evidence was unavailable prior to this study. The
literature that does exist suggests that socioeconomic
inequalities negatively impact the ability of underserved patients to receive treatment for lung and head
and neck cancers, and, when ethnicity is included, impoverished Hispanic patients are at a particular disadvantage.9-11
A paucity of evidence explains what factors determine the treatment delays experienced by this patient
population, but the available data indicate that the time
interval between the initial diagnosis and the start of
treatment continues to increase and that delays beyond
46 days increase the risk of death.17,20 Treatment delays
among patients with lung and head and neck cancers
occur more often in those with early-stage disease,17,21
placing them at a higher risk for disease progression,22-25 disease recurrence,13-16 increased emotional
distress,18,19 and lower rates of survival.17 Thus, assessing the determinants of treatment delays is important,
because such understanding could lead to interventions that effectively target the factors that influence
treatment inequalities among vulnerable populations,
such as underserved Hispanic patients with lung and
head and neck cancers.
We used the Behavioral Model for Vulnerable
Populations to frame our study design and analysis because it includes domains relevant to understanding
the factors that influence access and the utilization of
health care services among vulnerable populations.30,31
Thus, the Behavioral Model for Vulnerable Populations
guided the choice of variables to include in the prediction model, from the medical records and the questions to ask patients and their caregivers, HCPs, and
patient navigators.
Institutional-Related Factors
The literature and our findings unequivocally point to
the need to decrease the treatment delays for lung and
head and neck cancers because the timely initiation of
treatment is likely to result in improved outcomes and
survival for patients with these types of cancer. In our
study, 35% of patients had treatment delays longer than
46 days, at a point when these patients have a significant increased risk of death.17 Series of institution-level
factors common to publicly funded hospitals appear to
be associated with such treatment delays in the underserved patients of our study. At the safety-net hospital
where we conducted the study, lack of care coordina398 Cancer Control
tion was common, either from enrolling patients for
services to coordinating the specialty treatments they
need — all of which were believed to result in treatment delays.
In our study, communication was lacking across
various clinics, providers, and patients when coordinating care, and it was consistent with the intuitive notion that the multidisciplinary coordination
of care is complex and takes time, particularly if
specialty care services are limited.28 Creating and
encouraging the participation of HCPs in multidisciplinary tumor boards at public hospitals may
help improve communication and contribute to better care coordination among oncologists and other
HCPs. Use of tumor review boards has been associated with improved care and increased rates of survival among patients with cancer, although this association is controversial, and limitations still exist
for those living in rural, low-resource areas, as well
as in other suboptimal settings.35,36 Undergoing curative surgery among patients with early-stage lung
and head and neck cancers is also likely to be improved when their HCPs participate in weekly tumor
board meetings.37
The patients and caregivers interviewed did not
attribute poor treatment outcomes to treatment delays
due to lack of care coordination or lack of communication; rather, they attributed treatment delays to an initial missed diagnosis of cancer by their primary care
physician. They implied that this initial missed diagnosis resulted in a delayed referral to an oncologist. One
meta-analysis found that delayed referrals to specialists
predicted a 3-fold mortality risk for patients with head
and neck cancers, likely resulting from treatment delays and advanced disease.38 In our study, patients and
caregivers believed that, after being diagnosed, the
safety-net hospital provided patients with the care that
they needed and viewed any treatment delays as being
less consequential.
The 7 patient navigators interviewed recognized
that delays in treatment were overall related to lack of
care coordination and miscommunication across hospital clinics. They were particularly concerned that
patients with lung and head and neck cancers need
system-navigation services to decrease delays in treatment, but they noted that patients were not receiving
such services at the hospital.
If the systems-level factors are not addressed,
our findings suggest that treatment delays are likely
to continue — and possibly increase disparities in
treatment outcomes — among underserved Hispanic
patients with lung and head and neck cancers. An
evidence-based solution that can improve communication and care coordination may well be a patientnavigation program for this underserved patient
population.39,40
October 2016, Vol. 23, No. 4
Patient-Related Factors
As is characteristic of other patients at the safety-net
hospital, the Hispanic patients in our study were medically underserved, with the majority of participants
(60%) being uninsured and all of them being poor
(annual income < $36,000).41 Due to lack of variability
in income and health insurance status, this factor did
not contribute to the prediction of treatment delays in
our study. Instead, sex and number of comorbidities
had independent effects on treatment delay, but these
variables were also intertwined. Compared with men
whose treatment was, on average, delayed by 34 days,
disparities in treatment delays were more pronounced
for women whose treatment was delayed by, on average, 48 days (above the critical 46-day delay17) from initial diagnosis to treatment initiation. Moreover, women
with fewer comorbidities (≤ 4) were more likely to have
increased delays in treatment compared with women
who had more comorbidities or men who had a similar
lower number of comorbid conditions. Compared with
men, women were also more likely to be diagnosed
with early-stage disease. Thus, our findings suggest
that healthier women might be at high risk for treatment delays and patients whom safety-net hospital
staff should follow-up more closely to avoid disparities
in poor treatment outcomes,13-16,22-25 distress,18,19 and
low rates of survival.17
Some published data indicate that the time from
first diagnosis to treatment initiation does not significantly vary by comorbidity among patients treated at
academic facilities.17 For patients treated at high-burden, safety-net hospitals, it is possible that a greater
number of comorbidities means that these patients are
better integrated into the system and, consequently,
experience fewer treatment delays. If this is the case,
then we think it appropriate to assume that patients
who receive care for comorbid conditions are likely to
have already enrolled in hospital services, medication
programs, and be adept at navigating the health care
system. Thus, having more than 4 comorbid conditions
could be a proxy for patients having had some institution-level barriers addressed, possibly resulting in a
more timely initiation of cancer treatment.
Lack of legal status in the United States and lack
of English-language proficiency are determinants
of treatment delays that disadvantage medically underserved Hispanic patients, although the effects of
these factors on treatment delays among these patients have not been quantified. As found in our
study, patients with undocumented status are unable to qualify for resources that other underserved
patients are able to access to cover their cancer treatment, and, if they eventually do, their treatment is
likely to be significantly delayed. Similarly, our findings indicate that immigrant Hispanic patients who
do not communicate in English face barriers to acOctober 2016, Vol. 23, No. 4
cessing care that may result in treatment delays. An
adequate social support system that can assist patients with language barriers and to navigate the
health care system could facilitate the ability of patients to obtain proper and timely treatment. A factor
common to underserved patients is the lack of health
literacy that, in this study, was reported to be associated with lack of proper and timely adherence to
recommended treatment.42 For patients who lack adequate social support — particularly those with limited proficiency in English — or adequate health literacy, the presence of language translators or of patient
navigators can help to engage patients and caregivers
during physician visits, thereby increasing their ability to understand and follow the required steps in the
cancer treatment process.39,40
A barrier to individuals with lung and head and
neck cancers is substance abuse, which, in our study,
was anecdotally linked to delays for patients unable to
initiate treatment if they continued to smoke tobacco
or drink alcohol. In particular, substance-abuse problems place underserved Hispanic patients with these
types of cancers at a higher risk for emotional distress
and mortality.1,11,43 Because mental health disparities
are present among underserved Hispanic patients with
cancer, affordable mental health services can assist
them in addressing these problems and improve treatment delays.44
Limitations
This exploratory study used a convenience sample to
explore some possible determinants of treatment delays among underserved Hispanic patients with lung
and head and neck cancers. A randomly selected
sample followed longitudinally would strengthen the
generalizability of the study results to this population
of patients with cancer. The categorization of patients
as Hispanics also limits generalizations to how determinants of treatment delays may differ by other ethnic groups (eg, country of origin, acculturation levels).
Methodologically, we were limited by the data available in the medical records of the study patients, and
we were unable to assess their reliability.
Because the incomes of all of our study participants fell below the poverty level at the beginning
of the study,14 we could not examine the effects of
SES on the number of days from the initial diagnosis to treatment initiation as reported in their medical records. Instead, in-depth interviews with the
HCPs suggested that low SES contributed to treatment delays via the patients’ lack of health insurance. We also could not explore the effects of health
insurance coverage, because most patients were uninsured or lacked proper health insurance through
Medicaid or Medicare. However, the Affordable Care
Act that expands the coverage of public health inCancer Control 399
surance for underserved patients may now improve
their access to timely and proper treatment; however, its effects on cancer disparities remains to be
demonstrated.45
Conclusions
Data suggest that delays in cancer treatment are associated with poor outcomes and lower rates of survival for
patients with lung and head and neck cancers. Thus,
immediate action should be taken to improve the time
between diagnosis and treatment initiation for these
disadvantaged patients.
Future studies should assess the effects of the determinants that our interviewees believed influence
delays in treatment initiation among underserved Hispanic patients with cancer treated at a safety-net hospital. Our findings could be used to guide future research as well as interventions for underserved patient
populations.
Acknowledgment: The authors wish to thank Drs
Estevan Flores, Paula Espinoza, Al Marcus, and Tim
Byers for their generous advice.
References
1. American Cancer Society. Cancer Facts and Figures 2016.
http://w w w.cancer.org/acs/groups/content /@research/documents/
document/acspc-047079.pdf. Accessed September 6, 2016.
2. American Cancer Society. Cancer Facts & Figures for Hispanics/
Latinos: 2015-2017. http://www.cancer.org/acs/groups/content/@research/
documents/document/acspc-046405.pdf. Accessed September 6, 2016.
3.
Ries LAG, Melbert D, Krapcho M, et al, eds. SEER Cancer Statistics
Review, 1975-2005, National Cancer Institute. Bethesda, MD. http://seer.
cancer.gov/csr/1975_2005. Based on November 2007 SEER data submission.
Accessed July 29, 2016.
4. Kuriakose MA, Loree TR, Rubenfeld A, et al. Simultaneously
presenting head and neck and lung cancer: a diagnostic and treatment
dilemma. Laringoscope. 2002;112(1):120-123.
5.
Centers for Disease Control and Prevention. Smoking-attributable
mortality, years of potential life lost, and productivity losses – United
States, 2000-2004. MMWR Morb Mortal Wkly Rep. 2008;57(45):1226-1228.
6.
Blot WJ, McLaughlin JK, Winn DM, et al. Smoking and drinking in
relation to oral and pharyngeal cancer. Cancer Res. 1988;48(11):3282-87.
7.
Centers for Disease Control and Prevention. Cigarette smokingattributable morbidity -- United States, 2000. MMWR Morb Mortal Wkly Rep.
2003;52(35):842-844.
8. Agency for Healthcare Research and Quality (AHRQ). 2014 National
Healthcare Quality & Disparities Report. Rockville, MD: AHRQ; 2015.
http://www.ahrq.gov/sites/default/files/wysiwyg/research/findings/nhqrdr/
nhqdr14/2014nhqdr.pdf. Accessed September 6, 2016.
9. Forrest LF, Adams J, Wareham H, et al. Socioeconomic inequalities
in lung cancer treatment: systematic review and meta-analysis. PLoS Med.
2013;10(2):e1001376.
10.
Choi SH, Terrell JE, Fowler KE, et al. Socioeconomic and other
demographic disparities predicting survival among head and neck cancer
patients. PloS one. 2016;1;11(3):e0149886.
11. Yang R, Cheung MC, Byrne MM, et al. Do racial or socioeconomic
disparities exist in lung cancer treatment? Cancer. 2010;116(10):2437-2447.
12.
Committee on Quality of Health Care in America; Institute of Medicine.
Crossing the Quality Chasm: A New Health System for the 21st Century.
Washington, DC: National Academy Press; 2001.
13. Ang KK, Trotti A, Brown BW, et al. Randomized trial addressing risk
features and time factors of surgery plus radiotherapy in advanced headand-neck cancer. Int J Radiat Oncol Biol Phys. 2001;51(3):571-578.
14. Fortin A, Bairati I, Albert M, et al. Effect of treatment delay on outcome
of patients with early-stage head-and neck carcinoma receiving radical
radiotherapy. Int J Radiat Oncol Biol Phys. 2002;52(4):929-936.
15. Hansen O, Larsen S, Bastholt L, et al. Duration of symptoms:
impact on outcome of radiotherapy in glottis cancer patients. Int J Radiat
Oncol Biol Phys. 2005;61(3):789-794.
16. Chen Z, King W, Pearcey R, et al. The relationship between waiting
time for radiotherapy and clinical outcomes: a systematic review of the
400 Cancer Control
literature. Radiother Oncol. 2008;87(1):3-16.
17.
Murphy CT, Galloway TJ, Handorf EA, et al. Survival impact of
increasing time to treatment initiation for patients with head and neck cancer
in the United States. J Clin Oncol. 2016;34(2):169-178.
18.
Haisfield-Wolfe ME, McGuire DB, Soeken K, et al. Prevalence
and correlates of depression among patients with head and neck cancer:
a systematic review of implications for research. Oncol Nurs Forum.
2009;36(3):E107-E125.
19.Hess CB, Chen AM. Measuring psychosocial functioning in
the radiation oncology clinic: a systematic review. Psychooncology.
2014;23(8):841-854.
20. Murphy CT, Galloway TJ, Handorf EA, et al. Increasing time to treatment initiation for head and neck cancer: an analysis of the National
Cancer Database. Cancer. 2015;121(8):1204-1213.
21. Vinas F, Ben Hassen I, Jabot L, et al. Delays for diagnosis and treatment of lung cancers: a systematic review. Clin Respir J. 2016;10(3):267-271.
22.
Waaijer A, Terhaard CHJ, Dehnad H, et al. Waiting times for radiotherapy: consequences of volume increase for the TCP in oropharyngeal
carcinoma. Radiother Oncol. 2003;66(3):271-276.
23.
Jensen AR, Nellemann HM, Overgaard J. Tumor progression in
waiting time for radiotherapy in head and neck cancer. Radiother Oncol.
2007;84(1)5-10.
24.
Kowalski LP, Carvalho AL. Influence of time delay and clinical upstaging in the prognosis of head and neck cancer. Oral Oncol.
2001;37(1):94-98.
25. Laroche C, Wells F, Coulden R, et al. Improving surgical resection
rate in lung cancer. Thorax. 1998;53(6):445-449.
26. Yorio JT, Xie Y, Yan J, et al. Lung cancer diagnostic and treatment
intervals in the United States: a health care disparity? J Thoracic Oncol.
2009;4(11):1322-1330.
27.
Munck K, Ali MJ, Murr AH, et al. Impact of socioeconomic status
on the diagnosis to treatment interval (DTI) in Waldeyer’s ring carcinoma.
Laryngoscope. 2005;115(7):1283-1287.
28.
Patel UA, Brennan TE. Disparities in head and neck cancer: assessing delay in treatment initiation. Laryngoscope. 2012;122(8):1756-1760.
29.
Creswell JW, Plano Clark VL. Designing and Conducting Mixed
Methods Research. 2nd ed. Thousand Oaks, CA: Sage Publications; 2007.
30.
Gelberg L, Andersen RM, Leake BD. Healthcare access and utilization. The Behavioral Model for Vulnerable Populations: application to
medical care use and outcomes for homeless people. Health Serv Res.
2000;34(6):1273-1302.
31.
Bazargan M, Bazargan SH, Farooq M, et al. Correlates of cervical cancer screening among underserved Hispanic and African American
women. Prev Med. 2004;39(3):465-473.
32. Singh B, Bhaya M, Stern J, et al. Validation of the Charlson
Comorbidity Index in patients with head and neck cancer: a multi‐institutional study. Laryngoscope. 1997;107(11 pt 1):1469-1475.
33.Altheide DL. Ethnographic content analysis. Qualitat Sociol. 1987;
10(1):65-77.
34.
Krippendorf K. Content Analysis: An Introduction to its Methodology.
2nd ed. Beverly Hills, CA: Sage Publications; 2004.
35. Keating NL, Landrum MB, Lamont EB, et al. Tumor boards and the
quality of cancer care. J Natl Cancer Inst. 2013;105(2):113-121.
36. Krasna M, Freeman RK, Petrelli NJ. Re: Tumor boards and the
quality of cancer care. J Natl Cancer Inst. 2013;105(23):1839-1840.
37. Kehl KL, Landrum MB, Kahn KL, et al. Tumor board participation
among physicians caring for patients with lung or colorectal cancer. J Oncol
Pract. 2015;11(3):e267-e278.
38.Seoane J, Takkouche B, Varela-Centelles P, et al. Impact of delay
in diagnosis on survival to head and neck carcinomas: a systematic review
with meta-analysis. Clin Otolaryngol. 2012;37(2):99-106.
39.
Pasket ED, Harrop PJ, Wells KJ. Patient navigation: an update on
the state of science. Cancer. 2011;61(4):237-249.
40.
Raich PC, Whitley EM, Thorland W, et al. Patient navigation improves cancer diagnostic resolution: an individually randomized clinical
trial in an underserved population. Cancer Epidemiol Biomarkers Prev.
2012;21(10):1629-1638.
41.
US Department of Health and Human Services; Office of The
Assistant Secretary for Planning and Evaluation. 2013 poverty guidelines.
Published December 1, 2013. http://aspe.hhs.gov/poverty/13poverty.cfm.
Accessed September 6, 2016.
42. Shaw SJ, Huebner C, Armin J, et al. The role of culture in health
literacy and chronic disease screening and management. J Immigr Minor
Health. 2009;11(6):460-467.
43.Glazier KA. Tobacco-induced diseases: an emphasis on mental
health conditions that may result from cigarette use. N Sch Psychol Bulln.
2010;8(1):56-61.
44.
Luckett T, Goldstein D, Butow PN, et al. Psychological morbidity
and quality of life of ethnic minority patients with cancer: a systematic
review and meta-analysis. Lancet Oncol. 2011;12(13):1240-1248.
45. Moy B, Polite BN, Halpern MT, et al. American Society of Clinical
Oncology policy statement: opportunities in the patient protection and
Affordable Care Act to reduce cancer care disparities. J Clin Oncol.
2011;29(28):3816-3824.
October 2016, Vol. 23, No. 4
Geographical analysis can be used to
respond to select barriers that contribute
to disparities in prostate care.
Orchid. X-Ray image courtesy of Bryan Whitney. www.x-rayphotography.com.
Geographical Factors Associated With Health Disparities
in Prostate Cancer
Scott M. Gilbert, MD, Julio M. Pow-Sang, MD, and Hong Xiao, PhD
Background: Treatment variation in prostate cancer is common, and it is driven by clinical and clinician
factors, patient preferences, availability of resources, and access to physicians and treating facilities. Most
research on treatment disparities in men with prostate cancer has focused on race and socioeconomic factors.
However, the geography of disparities — capturing racial and socioeconomic differences based on where patients live — can provide insight into barriers to care and help identify outlier areas in which access to care,
health resources, or both are more pronounced.
Methods: Research regarding treatment patterns and disparities in prostate cancer using the Geographical
Information System (GIS) was searched. Studies were limited to English-language articles and research focused
on US populations. A total of 43 articles were found; of those, 30 provided information about or used spatial
or geographical analyses to assess and describe differences or disparities in prostate cancer and its treatment.
Two additional GIS resources were included.
Results: The research on geographical and spatial determinants of prostate cancer disparities was reviewed.
We also examined geographical analyses at the state level, focusing on Florida. Overall, we described a geographical framework to disparities that affect men with prostate cancer and reviewed existing published
evidence supporting the interplay of geographical factors and disparities in prostate cancer.
Conclusions: Disparities in prostate cancer are common and persistent, and notable differences in treatment
are observable across racial and socioeconomic strata. Geographical analysis provides additional information about where disparate groups live and also helps to map access to care. This information can be used by
public health officials, health-systems administrators, clinicians, and policymakers to better understand and
respond to geographical barriers that contribute to disparities in care.
From the Departments of Genitourinary Oncology (SMG) and
Health Outcomes and Behavior (SMG, JP), H. Lee Moffitt Cancer
Center & Research Institute, Tampa, Florida, and the Department
of Pharmaceutical Outcomes and Policy (HX), College of Pharmacy, University of Florida, Gainesville, Florida.
Submitted May 2, 2016; accepted September 20, 2016.
Address correspondence to Scott M. Gilbert, MD, Departments
of GU Oncology & Health Outcomes and Behavior, Moffitt Cancer Center, 12902 Magnolia Drive, XCB-GU Program, Tampa, FL
33612-9416. E-mail: [email protected]
No significant relationships exist between the authors and the
companies/organizations whose products or services may be referenced in this article.
October 2016, Vol. 23, No. 4
Introduction
Prostate cancer is the most common nondermatological cancer diagnosed among American men, accounting for approximately 180,000 new cases each year in
the United States.1 Although it is typically characterized by a protracted clinical course, prostate cancer is
also one of the most common causes of cancer-related
death (fourth most common overall following lung,
colorectal, and breast cancers) and accounts for more
than 26,000 deaths related to cancer each year in the
United States alone.2
Cancer Control 401
Although most prostate cancers (80%) are detected at an early stage and 10-year survival rates surpass
90%, 16% of men present with either locally advanced
or metastatic disease.3 Metastatic prostate cancer is
an incurable disease despite advances in treatment.3
Early-stage prostate cancer is also a heterogeneous
disease defined by variable clinical behavior and outcomes.4 Following local treatment, prostate cancer recurs in up to 40% of men with early-stage disease.5,6
An estimated 2.85 million American men are living
with prostate cancer, making this patient group the
second largest group of cancer survivors.3 With health
care expenditures surpassing an estimated $9.8 billion each year, prostate cancer is also one of the most
costly cancers in the United States.7
Although these figures underscore the personal
and public health burdens associated with prostate
cancer, not all men fare equally. Disparities in risk of
aggressive disease and differences in treatment and
outcomes are consistently observed.3-11 For example,
incidence rates among black men (214.5 per 100,000
persons) are significantly higher compared with their
white counterparts (130.0 per 100,000 persons), and
death rates from prostate cancer are more than double
for black men compared with their white counterparts
(46.3 per 100,000 persons vs 19.8/100,000 persons,
respectively).3 Several factors may contribute to observed racial disparities, including biological determinants (differences in genetic polymorphisms related to
testosterone metabolism and the androgen receptor)
differences in health behaviors, and access to health
care resources.8 Beyond racial factors, men with lower
social status and educational level are also more frequently diagnosed with more aggressive, clinically significant forms of prostate cancer than men from higher
socioeconomic backgrounds, and prior research indicates that health seeking and literacy issues, as well as
access to health care resources, may partially explain
why some men have poorer outcomes than others.9-11
Certainly social and demographical factors may be signals for other yet-to-be identified sources that drive divergent outcomes, or they may be principal factors that
explain why some men die from prostate cancer and
others do not. Disparate risk factors and outcomes can
be defined by geographical boundaries, and there may
be a geographical component to health disparities.
Methods
In addition to studying broader reviews of racial and sociodemographic disparities, our search of available literature databases focused on geographical factors associated
with health and outcome disparities, and use of spatial
analyses, including the Geographical Information System (GIS), in disparity research studies related to prostate
cancer. Additional, nonindexed prostate cancer treatment
and outcome reports using the GIS were searched online,
402 Cancer Control
and reports were examined and included if they were related to geographical and health disparities.
Studies were limited to research focusing on US
populations and those published in the English language. A total of 43 publications were identified, 30 of
which were relevant and included in our review.
Results
The geographical and prostate cancer factors found in
selected disparity studies have been summarized in
the Table.12-25
Racial and Sociodemographic Disparities
Disparities in treatment and survival between black and
non-black men have long been observed. Research has
shown that black men are at significantly higher risk of
dying from prostate cancer than white men, they are
less likely to receive curative local therapy for intermediate- and high-risk disease than their white counterparts,
and small but significant differences in 5-year survival
rates have been documented.26,27 Mahal et al27 reported
that black men are 12% more likely to die from prostate
cancer than white men (hazard ratio 1.12; 95% confidence interval [CI]: 1.01–1.25), corresponding to an absolute risk of death of 4.5% for blacks (95% CI: 4.1–4.9)
compared with 3.4% for whites (95% CI: 3.3–3.6). They
also found that black men were 18% less likely to receive
curative treatment than their white counterparts.27 Other studies have also reported lower rates of screening
and appropriate staging rates among black men compared with white men.28,29 Black men less frequently receive curative therapy, they may receive treatment later
after diagnosis, and they are less likely to be treated with
adjuvant therapy than white men.29
Increased risk of mortality has been observed in
black men as well as in men from disadvantaged social, educational, and economic backgrounds: these
men have higher rates of death from prostate cancer,
regardless of race.30 Inequity in mortality related to
prostate cancer is generally ascribed to 1 of 3 factors:
disease factors (eg, differences in biology, risk of more
aggressive disease in some groups), patient factors
(eg, health behaviors), and issues endemic to the health
care system (eg, differences in access to health care facilities and resources, screening and detection practices, treatment variation).31-33
Social, economical, and education disparities may
be linked to significant differences in disease management and outcomes.34-39 Several studies have shown that
men from disadvantaged groups (eg, those with lower
socioeconomic status) are less likely to receive aggressive
local treatment, such as radical surgery or radiotherapy,
than those of higher socioeconomic status.34-37 Screenings are also less commonly performed in poorer and
less-educated communities, possibly leading to diagnoses at a later-stage disease when fewer treatment options
October 2016, Vol. 23, No. 4
Table. — Summary of Select Disparity Studies With Geographical and Prostate Cancer Factors
Study
Geographical Unit
Disease Phase
Key Finding
Aneja12
United States, county
Mortality
3.91%–5.45% reduction in mortality in counties with
≥ 1 radiation oncologist compared with counties lacking
radiation oncologists
Antwi13
Kentucky, Appalachian region
Survival
Black men had lower survival rates compared with white men
(HR 1.41; 95% CI: 1.26–1.65)
Cetnar 14
Wisconsin, census tract
Primary treatment
Likelihood of noncurative treatment no different in rural
compared with urban areas (OR 0.96; 95% CI: 0.52–1.77)
Cobran15
United States, SEER site
Treatment
Intensity-modulated radiotherapy more commonly used in
white than blacks (53% vs 45%; P < .05)
Socioeconomic-, geographic-, and tumor-related factors
explained preponderance of variation
Cook 16
United States, census divisions
Incidence rates
Large geographical differences in black-to-white incidence
rate ratios
DeChello17
Connecticut and Massachusetts,
census tract
Incidence rates
Significant geographical variation observed in age-adjusted
incidence rates among white and black men
Garg18
United States, state
Screening
Significant variation in PSA screening among states
Men living in urban areas have lower odds of screening
(OR 0.8; 95% CI: 0.7–0.9), low physician density, low PSA
screening (OR 0.78; 95% CI: 0.63–0.94)
Goovaerts19
Florida, county
Incidence of late-stage disease
Variation noted in temporal decrease in late-stage diagnosis
by county and race
Gregorio20
Connecticut, census tract
Survival
Spatial scan statistical analysis of adjusted survival times
identified one significant outlier area in the state with
significantly lower survival (death hazard 1.39 > state average; P < .05)
Meliker 21
Michigan, legislative districts
Survival
Higher survival rates observed in white men at state level
without confirmation of racial disparities at smaller
geographical units
Onukwugha 22
United States, county
Follow-up care
Significant variation in follow-up physician visits explained
by geographical factors
Black men living in low socioeconomic and high-crime areas
less likely to receive follow-up care
Wagner 23
Georgia, county
Incidence rates
“Hot spots” identified in several counties in both black and
white men in northern and northwestern-central Georgia
Xiao24
Florida, county and census tract
Incidence of late-stage disease
Late-stage diagnosis related to low census tract median
income and lower educational attainment
Xiao25
Florida, county and census tract
Incidence of late-stage disease
Late-stage diagnosis associated with lack of insurance,
low household income, rural location
CI = confidence interval, HR = hazard ratio, OR = odds ratio, PSA = prostate-specific antigen, SEER = Surveillance, Epidemiology, and End Results.
may be available or cure is less likely.38,39 Level of health
literacy and patient decision-making capacity may also
contribute to socioeconomic disparities,40 a hypothesis
partly supported by research showing that racial disparities appear to be modified by socioeconomic status.41
This suggests that the gap between white and black men
residing in higher-income and higher-level education
neighborhoods is narrower than the gap separating residents from low socioeconomic status regions.
A systematic review analyzing the association between socioeconomic status and risk of prostate cancer–related death reported that most research supports
a significant link between socioeconomic status and
prostate cancer–related mortality, even after adjusting
for factors such as patient comorbidities and differences
October 2016, Vol. 23, No. 4
in treatment.30 In aggregate, these data suggest that low
socioeconomic status increases the risk of prostate cancer–related death in men with prostate cancer, even after accounting for differences in health, stage at diagnosis, and treatment. It is worth noting that socioeconomic
status disparities do not appear to have decreased in
the last 10 years.10 By contrast, average survival rates of
prostate cancer have improved during that same time.3
Geographical Disparities
Examining nationwide treatment trends, Harlan
et al42 noted significant regional variation across definitive treatment modalities among both white and black
men. The age-adjusted proportion of men treated with
prostatectomy ranged from 22.5% in Connecticut to
Cancer Control 403
47.8% in Utah, although use of radiotherapy varied less
across Surveillance, Epidemiology, and End Results
(SEER) cancer registry sites.42 In addition to confirming widespread variation in treatment modality in another SEER cohort, Krupski et al36 also documented
significant racial and socioeconomic variation in care,
with black and low-income men receiving surgery for
localized disease significantly less commonly than
white, higher-income men. The adjusted likelihood
of surgical therapy was less than one-half (odds ratio
[OR] 0.44; 95% CI: 0.41–0.47) in the lowest utilization
region compared with the highest, and the investigators documented that black men were one-half as likely
to be treated with surgery than whites (OR 0.52; 95% CI:
0.48–0.56).36 Regional variation in radiotherapy was
observed: it was typically more common in regions
with low rates of surgery.36 The results of another study
examining ecological correlations between variation in
prostate cancer–related deaths and surrogate measures
of access to medical care (eg, prostate-specific antigen
[PSA] screening uptake, late-stage presentation) suggest that between 10% and 30% of observed variations
in mortality rates may be attributable to access issues.43
Although regional differences in treatment for prostate cancer have been well described, a paucity of research has focused on the underlying determinants of
geographically linked disparities. Access, distance, and
travel time to health care facilities are germane when
compared with concerns related to disparities in treatment and subsequent outcomes among US residents living in rural areas.44 One review examined the differences in incidence and mortality rates between residents of
rural and urban areas.45 Few differences were observed
in incidence rates, particular in the post–PSA screening
era, but the findings of the review suggested that men
living in rural areas are at higher risk of prostate cancer–
related death than those living in urban areas, presumably due to issues related to health care accessability.45
Other research has also shown that patients living in rural areas, regardless of their racial/ethnic background,
are less likely to undergo PSA screening and more likely to present with more advanced disease, so they may
have fewer curative treatment options.43 Another study
found that awareness of and knowledge about prostate
cancer were lower among rural men,46 and another confirmed that screening and detection practices are significantly less common in rural than urban areas.47 Rates of
mortality and survival are also significantly associated
with race/ethnicity, socioeconomic status, and accessibility to health care among men with prostate cancer.48,49
Geographical Information System and Its Role in
Disparity Research
The GIS is a computer system designed to capture, store,
analyze, manage, and display spatially referenced data.
It was developed in the 1960s by Tomlinson and then
404 Cancer Control
further refined and commercialized by Fisher.50 Applying the GIS to visualize and analyze spatial and temporal
disease patterns has become more prevalent in health
care research in recent years.51 Health conditions such
as cardiovascular disease, hepatitis C virus infection,
birth defects, and various cancers have been displayed
in a spatial context to better understand these disease
states.51 By merging health data and geography, the GIS
can also help inform decision-making when allocating
resources. Thus, research involving GIS brings together
several different fields, including medicine, epidemiology, geography, computer sciences, and statistics.50 In
recent years, GIS has started to extend to areas of epidemiological and health services research — focusing on
health disparities — to examine the availability of health
resources, utilization of health care, the geographical
distribution and clustering of cancers, treatment, and
rates of mortality. For example, health-related GIS studies have increased the ability of public health officials
to monitor diseases and to identify areas of need. The
National Cancer Institute has an interactive website that
uses the GIS for cancer control purposes.52
In the field of prostate cancer, research using GIS
approaches has been conducted to investigate diagnoses, management, and treatment outcomes across
different geographical levels, such as counties, census
division/tracts, cancer registry areas, and other aggregate geographical scales.12-25,53-68 Cockburn et al53 used
the GIS to geocode the historical residential addresses
of individuals and measured the association between
ambient exposure to pesticide and incidence rates of
prostate cancer in California. They found a positive relationship between prostate cancer and ambient pesticide exposure in and around homes in intensely agricultural areas.53 Hsu et al56 used a spatial scan statistic
to detect counties with increased mortality between
1980 and 2001 in Texas. DeChello et al17 used the same
method and found geographical variation in incidence
rates of prostate cancer among both white and black
men between 1994 and 1998 in Connecticut and Massachusetts. In a subsequent analysis of the Connecticut data, researchers identified an outlier locale with
a significantly higher mortality rate (adjusted for age,
disease, and race) than other parts of the state. 20
Wagner et al23,62 applied GIS methods to visualize ratios of cancer mortality and incidence and detected
large geographical disparities among blacks in Georgia
compared with national benchmark data. Antwi et al13
found that men with prostate cancer living in the Appalachian region within Kentucky have lower rates of
survival than those living in non-Appalachian regions.
Differences in health resources and treatment patterns have also been examined among men with prostate cancer, and they have been linked to outcomes and
possible quality-of-care issues. Aneja and Yu12 mapped
the density of radiation oncologists for all counties in
October 2016, Vol. 23, No. 4
≥ 17.0% black (top quartile)
30–45
Rural (large and small)
> 45
Late-Stage Diagnosis (%)
0.4
15–30
Bla
2006
0.5
≥ 20% living in poverty (low income)
1982
ck males
0.3
County boundary
0–15
2006
0.1
Travel Time (min)
1982
ales
White m
0.2
≥ High-volume facility
temporal trends for whites and blacks were also observed in rural counties of Florida.19
Another study used boundary analysis and reported significant regional differences in relation to when
a decline of late-stage cases was seen, suggesting that
the introduction of PSA testing and urbanization in several areas of Florida led to fewer late-stage diagnoses
than that seen in nonurbanized areas, where uptake of
screening practices are likely to have been slower.64 Al-
0.0
the United States and found that a higher density rate
of radiation oncologists was associated with lower
rates of prostate cancer–related mortality. Cetnar et al14
compared trends in first-line treatment of prostate cancer in rural and urban areas in Wisconsin and found
that a person’s place of residence was not associated
with treatment choice.
GIS projects in other states have also examined
disparities related to geography. Research involving
men with prostate cancer has shown that the risk of
late-stage prostate cancer diagnoses among black men
living in Florida consistently exceeds that of white Floridians, although this gap has decreased in recent years.24,25
These studies used geographically weighted regression
to account for the geographical variation in racial and
sociodemographic composition, which provides estimates more likely to indicate the actual local impact of
sociodemographic and clinical characteristics on patient
outcomes.24,25 Using multiscale join-point regression
analysis to investigate geographical, temporal, and racial/ethnic disparities in the incidence rates of late-stage
prostate cancer among men living in Florida, researchers
illustrated how the proportion of late-stage incidence can
change over time.19,23 They observed striking geographical and racial disparities within a single state (Fig 1).19,23
Although the percentage of late-stage disease was halved
between 1980 and 2000, nonmetropolitan areas lagged
behind urban parts of the state, suggesting that late-stage
presentation is a larger problem in rural areas compared
with nonrural areas in Florida.19 Larger differences in
Fig 1. — Temporal patterns of late-stage prostate cancer across Florida
counties. Reprinted from Goovaerts P, Xiao H. Geographical, temporal
and racial disparities in late-stage prostate cancer incidence across
Florida: a multiscale joinpoint regression analysis. Int J Health Geogr.
2011;10:63. © Goovaerts and Xiao; licensee BioMed Central Ltd. 2011.
Licensed under the terms of the Creative Commons Attribution License
http://creativecommons.org/licenses/by/2.0.
The Northwest Florida CCC is notable for relatively
large uncovered (> 45 min to high-volume facilities)
rural areas characterized by low income and a high
percentage of black residents. Tallahassee is the
one area covering a relatively significant area of
black residents.
Fig 2. — Service area coverage and average travel times to high-volume prostate cancer treatment facilities in the Northwest Florida CCC area.
Reproduced from reference 71.
CCC = Cancer Control Collaborative.
October 2016, Vol. 23, No. 4
Cancer Control 405
though this gap has narrowed across most Florida counand time to high-volume treatment centers.71 Furtherties and the rate of late-stage diagnoses continues to
more, low income and rural areas of Florida also appear
decrease, some parts of the state still demonstrate perto be characterized by limited resources for prostate
sistent disparity.64 The decline in number of late-stage
cancer care and specialist health care professionals
diagnoses of prostate cancer has been slowest in Flor(eg, urologists, radiation oncologists).71 Treatment faciliida’s Big Bend region.64 In another study, researchers
ties are more sparse in the North-Central and Panhandle
compared the impact of comorbidities on late-stage diregions of Florida, thus resulting in longer travel times
agnoses at the local level with the expected state-level
for residents in those regions that appear to disproporrisk and found significant differences in observed-totionately affect disadvantaged men (Fig 2); by contrast,
expected results in Tallahassee and Pensacola, where
the southeastern and western parts of the state are
the local OR was smaller than expected, and in Palm
characterized by a greater number and higher density
Beach, where the OR was larger than expected.65
of treatment facilities, shorter average travel times
Racial/ethnic differences in how prostate cancer is
(particularly along the coastal areas and in high-income,
treated have also been documented in Florida through
low-minority areas).71 However, low income and black
GIS-based studies.66,67 These results point to the imporcommunities are still located beyond the close access
tance of more granular, local analyses to evaluate potenbands even in those regions of Florida, thus indicating
tial disparities and gaps, although the stability of estimata disproportionate and disparate service coverage for
ed incidence, treatment, and mortality rates can become
disadvantaged men (Fig 3).71
an issue when using small
geographical units (eg, zip
codes, census tracts), parThe Southwest Florida CCC is characterized
ticularly when cases are
by clustering of high-volume facilities and
further stratified by race, soservice coverage areas along the Gulf Coast
cioeconomic status, or both.
with large uncovered inland areas that are
low income and rural.
It is worth noting that empirical research suggests that
the census tract is the most
reliable unit of analysis for
spatial analyses.69,70
Another Florida study
examining racial/ethnic
and sociodemographic disparities focused on access
to health care resources
and differences in statewide treatment patterns.71
The Florida Prostate Cancer
Atlas Project71 chronicled
geographical variation in
the burden of disease (eg,
High-volume facilities
late-stage diagnosis), disCounty boundaries
tribution and availability of
≥ 20% living in poverty (low income)
health care resources relevant to prostate cancer,
≥ 17.0% black (top quartile)
as well as access to those
Rural (large and small)
resources, as assessed by
Travel Time (min)
travel times to treatment
0–15
facilities using cancer registry data from 1998 through
15–30
2007. The results of the
30–45
project demonstrate that
> 45
low-income areas and areas
characterized by a high percentage of black residents
Fig 3. — Service area coverage and average travel times to high-volume prostate cancer treatment facilities
in the Southwest Florida CCC area. Reproduced from reference 71.
were associated with the
CCC = Cancer Control Collaborative.
greatest distance to travel
406 Cancer Control
October 2016, Vol. 23, No. 4
Age-Adjusted Incidence Rate
≥ 15.8% Above state ratio by ≥ 20%
14.5%–15.8% Above state ratio by 10% to < 20%
11.9%–14.4% Within 10% of state ratio
mutable factors that contribute to health disparities.52 Responses to actionable geographical health
information include outreach and awareness campaigns, community-based
health ser vice effor ts,
such as screening events,
organizing providers and
resources into more accessible networks, and creating local infrastructure in
underserved communities
to allow better access and
care to needed health
services.52
References
1. Siegel RL, Miller KD, Jemal A.
Cancer statistics, 2016. CA Cancer
J Clin. 2016;66(1):7-30.
≤ 10.5% Below state ratio by ≥ 20%
2. American Cancer Society. Cancer Facts & Figures 2016. Atlanta, GA:
Suppressed (cases < 10)
American Cancer Society. http://www.
cancer.org/acs/groups/content /@
Unstable rate
re s e arc h /d o c um e nt s /d o c um e nt /
acspc-047079.pdf. Accessed September 21, 2016.
Fig 4. — Proportion of prostate cancer diagnoses at an advanced stage across Florida counties
3. National Cancer Institute. Sur(age-adjusted incidence rates, 1998–2007). Reproduced from reference 71.
veillance, Epidemiology, and End
Results Program. http://seer.cancer.
gov/statfacts/html/prost.html. Accessed
March 31, 2016.
4.Albertsen PC, Hanley JA, Fine J. 20-year outcomes following
Differences in resource accessibility may be asconservative management of clinically localized prostate cancer. JAMA.
sociated with differences in how prostate cancer
2005;293(17):2095-2101.
5. D’Amico AV, Whittington R, Malkowicz B, et al. Biochemical outpresents and how it is treated. The Florida Prostate
come after radical prostatectomy, external beam radiation therapy, or inCancer Atlas Project71 observed an overlap between
terstitial radiation therapy for clinically localized prostate cancer. JAMA.
1998;280(11):969-974.
low health care resource regions and areas in which
6. Freedland SJ, Humphreys EB, Mangold LA, et al. Risk of prostate
the case proportion of late-stage or aggressive discancer-specific mortality following biochemical recurrence after radical
prostatectomy. JAMA. 2005;294(4):433-439.
ease is highest (Fig 4). This geospatial data indicate
7. Roehrborn CG, Black LK. The economic burden of prostate cancer.
significant differences in access to health care, treatBJU Int. 2011;108(6):806-813.
8. Pettaway CA. Racial differences in the androgen/androgen receptor
ment, and outcomes that underlie and contribute to
pathway in prostate cancer. J Natl Med Assoc. 1999;91(12):653-660.
racial/ethnic, rural, and socioeconomic disparities,
9. Ward E, Jemal A, Cokkinides V, et al. Cancer disparities by race/
ethnicity and socioeconomic status. CA Cancer J Clin. 2004;54(2):78-93.
and they potentially point to areas in which public
10. Woods LM, Rachet B, Coleman MP. Origins of socio-economic
health efforts and resources could be directed to adinequalities in cancer survival: a review. Ann Oncol. 2006;17(1):5-19.
11. Bennett CL, Ferreira MR, Davis TC, et al. Relation between literacy,
dress access, awareness, and treatment gaps.71
race, and stage of presentation among low-income patients with prostate
cancer. J Clin Oncol. 1998;16(9):3101-3104.
12. Aneja S, Yu JB. The impact of county-level radiation oncologist
Conclusions
density on prostate cancer mortality in the United States. Prostate Cancer
Geographical variation in the presentation and
Prostatic Dis. 2012;15(4):391-396.
13. Antwi S, Tucker TC, Coker AL, et al. Racial disparities in survival
management of prostate cancer is well documentafter diagnosis of prostate cancer in Kentucky, 2001-2010. Am J Mens
ed. Although biological and disease-related factors
Health. 2013;7(4):306-316.
most likely explain some of this variation, differ 14. Cetnar JP, Hampton JM, Williamson AA, et al. Place of residence
and primary treatment of prostate cancer: examining trends in rural and
ences in the health care system, such as availability
nonrural areas in Wisconsin. Urology. 2013;81(3):540-546.
of and access to treating physicians and treatment
15. Cobran EK, Chen RC, Overman R, et al. Racial differences in diffusion of intensity-modulated radiation therapy for localized prostate cancer.
facilities, are likely to contribute to disparate treatAm J Mens Health. 2015;10(5):399-407.
ment and outcomes.51 Geographical level analy 16. Cook MB, Rosenberg PS, McCarty FA, et al. Racial disparities in
prostate cancer incidence rates by census division in the United States,
ses augment disparities research, providing a lens
1999-2008. Prostate. 2015;75(7):758-763.
through which clinicians, public health officials,
17. DeChello LM, Gregorio DI, Samociuk H. Race-specific geography
of prostate cancer incidence. Int J Health Geogr. 2006;5:59.
and policymakers can identify underserved areas
18. Garg V, Raisch DW, Selig JP, et al. Health disparities in clinical
and make informed decisions about how to address
practice patterns for prostate cancer screening by geographic regions
10.6%–11.8% Below state ratio by 10% to < 20%
October 2016, Vol. 23, No. 4
Cancer Control 407
in the United States: a multilevel modeling analysis. Prostate Cancer
Prostatic Dis. 2013;16(2):193-203.
19. Goovaerts P, Xiao H. Geographical, temporal and racial disparities
in late-stage prostate cancer incidence across Florida: a multiscale joinpoint regression analysis. Int J Health Geogr. 2011;10:63.
20. Gregorio DI, Huang L, DeChello LM, et al. Place of residence effect on
likelihood of surviving prostate cancer. Ann Epidemiol. 2007;17(7):520-524.
21. Meliker JR, Goovaerts P, Jacquez GM, et al. Breast and prostate
cancer survival in Michigan: can geographic analyses assist in understanding racial disparities? Cancer. 2009;115(10):2212-2221.
22. Onukwugha E, Osteen P, Jayasekera J, et al. Racial disparities in
urologist visits among elderly men with prostate cancer: a cohort analysis of patient-related and county of residence-related factors. Cancer.
2014;120(21):3385-3392.
23. Wagner SE, Bauer SE, Bayakly AR, Vena JE. Prostate cancer incidence and tumor severity in Georgia: descriptive epidemiology, racial disparity, and geographic trends. Cancer Cause Control. 2013;24(1):153-166.
24. Xiao H, Gwede CK, Kiros G, et al. Analysis of prostate cancer
incidence using geographic information system and multilevel modeling.
J Natl Med Assoc. 2007;99(3):218-225.
25. Xiao H, Tan F, Goovaerts P. Racial and geographic disparities in latestage prostate cancer diagnosis in Florida. J Health Care Poor Underserv.
2011;22(4 suppl):187-199.
26. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer
J Clin. 2015;65(1):5-29.
27. Mahal BA, Aizer AA, Ziehr DR, et al. Trends in disparate treatment
of African American men with localized prostate cancer across National
Comprehensive Cancer Network risk groups. Urology. 2014;84(2):386-392.
28. Barocas DA, Grubb R III, Black A, et al. Association between race
and follow-up diagnostic care after a positive prostate cancer screening
test in the prostate, lung, colorectal, and ovarian screening trial. Cancer.
2013;119(12):2223-2239.
29. Zeliadt SB, Potosky AL, Etzioni R, et al. Racial disparity in primary
and adjuvant treatment for nonmetastatic prostate cancer: SEER-Medicare
trends 1991 to 1999. Urology. 2004;64(6):1171-1176.
30. Klein J, von den Knesebeck O. Socioeconomic inequalities in prostate
cancer survival: a review of the evidence and explanatory factors. Soc Sci
Med. 2015;142:9-18.
31. Chornokur G, Dalton K, Borysova ME, et al. Disparities at presentation, diagnosis, treatment, and survival in African American men affected
by prostate cancer. Prostate. 2011;71:985-997.
32. Auviven A, Karjalainen S. Possible explanations for social class
differences in cancer patient survival. IARC Sci Publ. 1997;138:377-397.
33. Woods VD, Montgomery SB, Herring RP, et al. Social ecological
predictors of prostate-specific antigen blood test and digital rectal examination in black American men. J Natl Med Assoc. 2006;98(4):492-504.
34. Fairley L, Baker M, Whiteway J, et al. Trends in non-metastatic
prostate cancer management in the Northern and Yorkshire region of
England, 2000-2006. Br J Cancer. 2009;101(11):1839-1845.
35. Hayen A, Smith DP, Patel MI, et al. Patterns of surgical care for
prostate cancer in NSW, 1993-2002: rural/urban and socioeconomic
variation. Aust N Z J Public Health. 2008;32(5):417-420.
36.Krupski TL, Kwan L, Afifi AA, et al. Geographic and socioeconomic variation in the treatment of prostate cancer. J Clin Oncol.
2005;23(31):7881-7888.
37. Lyratzopoulos G, Barbiere JM, Greenberg DC, et al. Population
based time trends and socioeconomic variation in use of radiotherapy and
radical surgery for prostate cancer in the UK region: continuous survey.
BMJ. 2010;240:c1928.
38. Ross LE, Taylor YJ, Howard DL. Trends in prostate-specific antigen
test use, 2000-2005. Public Health Rep. 2011;126(2):228-239.
39. Williams N, Hughes LJ, Turner EL, et al. Prostate-specific antigen
testing rates remain low in UK general practice: a cross-sectional study in
six English cities. BJU Int. 2011;108(9):1402-1408.
40. Yin HS, Dreyer BP, Vivar KL, et al. Perceived barriers to care and
attitudes towards shared decision-making among low socioeconomic status parents: role of health literacy. Acad Pediatr. 2012;12(2):117-124.
41. Ziehr DR, Mahal BA, Aizer AA, et al. Income inequality and treatment of African American men with high-risk prostate cancer. Urol Oncol.
2015;33:18e7-e13.
42. Harlan L, Brawley O, Pommerenke F, et al. Geographic, age, and
racial variation in the treatment of local/regional carcinoma of the prostate.
J Clin Oncol. 1995;13(1):93-100.
43. Jemal A, Ward E, Wu X, et al. Geographic patterns of prostate cancer mortality and variations in access to medical care in the United States.
Cancer Epidemiol Biomark Prevent. 2005;14(3):590-595.
44. Jones AP, Haynes R, Sauerzapf V, et al. Travel times to health
care and survival from cancers in Northern England. Eur J Cancer.
2008;44(2):269-274.
45. Obertova Z, Brown C, Holmes M, et al. Prostate cancer and mortality in rural men – a systematic review of the literature. Rural Remote
Health. 2012;12(2):2039.
46. Pruthi RS, Tornehl C, Gaston K, et al. Impact of race, age, income,
and residence on prostate cancer knowledge, screening behavior, and
408 Cancer Control
health maintenance in siblings of patients with prostate cancer. Eur Urol.
2006;50(1):64-69.
47. Koh HK, Judge CM, Ferrer B, et al. Using public health data systems to understand and eliminate cancer disparities. Cancer Cause Control.
2005;16(1):15-26.
48. Xiao H, Tan F, Goovaerts P, et al. Multilevel factors associated with
overall mortality for men diagnosed with prostate cancer in Florida. Am J
Mens Health. 2014;8(4):316-326.
49. Xiao H, Tan F, Adunlin G, et al. Factors associated with overall survival prostate cancer in Florida: a multilevel analysis. J Health Care Poor
Underserved. 2015;26(1):266-277.
50. Coppock JT, Rhind DW. The history of GIS. In: Longley PA, Goodchild MF, Maguire DJ, et al, eds. Geographical Information Systems.
Hoboken, NJ: John Wiley & Sons; 1991:21-43. http://www.wiley.com/
legacy/wileychi/gis/Volume1/BB1v1_ch2.pdf. Accessed September 21, 2016.
51. Krieger N, Chen JT, Waterman PD, et al. Choosing area based
socioeconomic measures to monitor social inequities in low birth weight
and childhood lead poisoning: The Public Health Disparities Geocoding
Project (US). J Epidemiol Commun Health. 2003;57(3):186-199.
52. National Cancer Institute. Geographic information systems and science for cancer control. https://gis.cancer.gov. Accessed September 21, 2016.
53. Cockburn M, Mills P, Zhang X, et al. Prostate cancer and ambient
pesticide exposure in agriculturally intensive areas in California. Am J
Epidemiol. 2011;173(11):1280-1288.
54. Marusek JC, Cockburn MG, Mills PK, et al. Control selection and
pesticide exposure assessment via GIS in prostate cancer studies. Am J
Prev Med. 2006;30(2 suppl):S109-S116.
55. Cary C, Odisho AY, Cooperberg MR. Variation in prostate cancer
treatment associated with population density of the county of residence.
Prostate Cancer Prostatic Dis. 2016;19(2):174-179.
56. Hsu CE, Mas FS, Miller JA, et al. A spatial-temporal approach to
surveillance of prostate cancer disparities in population subgroups. J Natl
Med Assoc. 2007;99(1):72-80, 85-77.
57. Dean LT, Subramanian SV, Williams DR, et al. Getting black men to
undergo prostate cancer screening: the role of social capital. Am J Mens
Health. 2015;9(5):385-396.
58. Wagner SE, Burch JB, Hussey J, et al. Soil zinc content, groundwater usage, and prostate cancer incidence in South Carolina. Cancer Cause
Control. 2009;20(3):345-353.
59. Fogleman AJ, Mueller GS, Jenkins WD. Does where you live play
an important role in cancer incidence in the U.S.? Am J Cancer Res.
2015;5(7):2314-2319.
60. Lu-Yao G, Albertsen PC, Stanford JL, et al. Screening, treatment,
and prostate cancer mortality in the Seattle area and Connecticut:
fifteen-year follow-up. J Gen Intern Med. 2008;23(11):1809-1814.
61. Mossanen M, Izard J, Wright JL, et al. Identification of underserved
areas for urologic cancer care. Cancer. 2014;120(10):1565-1571.
62. Wagner SE, Hurley DM, Hebert JR, et al. Cancer mortality-toincidence ratios in Georgia: describing racial cancer disparities and
potential geographic determinants. Cancer. 2012;118(16):4032-4045.
63.Wan N, Zhan FB, Cai Z. Socioeconomic disparities in prostate cancer mortality and the impact of geographic scale. South Med J.
2011;104(8):553-559.
64. Goovaerts P, Xiao H. The impact of place and time on the proportion
of late-stage diagnosis: the case of prostate cancer in Florida, 1981-2007.
Spat Spatiotemporal Epidemiol. 2012;3(3):243-253.
65. Goovaerts P, Xiao H, Adunlin G, et al. Geographically-weighted
regression analysis of percentage of late-stage prostate cancer diagnosed
in Florida. Appl Geogr. 2015;62:191-200.
66. Xiao H, Warrick C, Huang Y. Prostate cancer treatment patterns among
racial/ethnic groups in Florida. J Natl Med Assoc. 2009;101(9):936-943.
67. Xiao H, Tan F, Goovaerts P, et al. Factors associated with time-totreatment of prostate cancer in Florida. J Health Care Poor Underserv.
2013;24(4 suppl):132-146.
68.Oliver MN, Smith E, Siadaty M, et al. Spatial analysis of prostate
cancer incidence and race in Virginia, 1990-1999. Am J Prev Med.
2006;30(suppl):S67-S76.
69. Krieger N, Waterman P, Chen JT, et al. Zip code caveat: bias due
to spatiotemporal mismatches between zip codes and US census-defined
geographic areas – the Public Health Disparities Project. Am J Pub Health.
2002;92(7):1100-1102.
70. Gregorio DI, Dechello LM, Samociuk H, et al. Lumping or splitting:
seeking the preferred areal unit for health geography studies. Int J Health
Geogr. 2005;4(1):6.
71. Gilbert SM, Henriksen C, Hutton C, et al. Florida Prostate Cancer
Atlas. https://com-uro-advisory-council.sites.medinfo.ufl.edu/files/2014/07/
Atlas_final_with_cover_final.pdf. Accessed September 21, 2016.
October 2016, Vol. 23, No. 4
Differences in racial/ethnic, sociodemographical, and economic disparities
in penile cancer can influence cancer
control and survival-related outcomes.
Birds Nest. X-Ray image courtesy of Bryan Whitney. www.x-rayphotography.com.
Disparities in Penile Cancer
Pranav Sharma, MD, Kamran Zargar-Shoshtari, MD, Curtis A. Pettaway, MD, Matthew B. Schabath, PhD,
Anna R. Giuliano, PhD, and Philippe E. Spiess, MD
Background: Although penile cancer is a rare malignancy in developed nations, racial and socioeconomic
differences exist in the incidence of the disease and its associated survival-related outcomes.
Methods: A search of the literature was performed for research published between the years 1990 and 2015.
Case reports and non–English-language articles were excluded, instead focusing specifically on large, population-based studies.
Results: The incidence of penile cancer is higher in Hispanic and African American men compared with whites
and Asians. Men with penile cancer also appear to have a distinct epidemiological profile, including lower
educational and income levels, a history of multiple sexual partners and sexually transmitted infections, and
lack of circumcision with the presence of phimosis. African American men presented at a younger age with a
higher stage of disease and worse survival rates when compared with white men. Rates of cancer-specific mortality increased with age, single marital status, and among those living in regions of lower socioeconomic status.
Conclusions: An understanding of sociodemographical differences in the incidence and survival rates of
patients with penile cancer can help advance health care policy changes designed to improve access and
minimize disparities in cancer care for all men alike.
Introduction
Penile cancer is rare in the United States, with an
estimated 2,030 new cases in the United States and
340 related deaths estimated to occur this year.1
From the Departments of Genitourinary Oncology (PS, KZ-S, PES)
and Cancer Epidemiology (MBS, ARG), H. Lee Moffitt Cancer Center
& Research Institute, Tampa, Florida, and the Department of
Urology (CAP), University of Texas MD Anderson Cancer Center,
Houston, Texas.
Submitted January 21, 2016; accepted February 26, 2016.
Address correspondence to Philippe E. Spiess, MD, Department of
Genitourinary Oncology, Moffitt Cancer Center, 12902 Magnolia Drive,
WCB-GU PROG, Tampa, FL 33612. E-mail: [email protected]
Dr Sharma is now affiliated with the Department of Urology at Texas
Tech University Health Sciences Center in Lubbock, Texas.
No significant relationships exist between the authors and the companies/organizations whose products or services may be referenced
in this article.
October 2016, Vol. 23, No. 4
However, its incidence is higher in some countries of
Asia, Africa, and South America, where it represents
10% of all malignancies diagnosed among men.2
Patients with penile cancer appear to have a distinct epidemiological profile consisting of lower income and education levels, poor hygiene, a history of
sexually transmitted infections, phimosis, and multiple
sexual partners.3 Risk factors for penile cancer include
lack of circumcision, presence of phimosis or paraphimosis, poor hygiene, human papillomavirus (HPV) infection, presence of genital warts, chronic inflammatory conditions of the foreskin, glans penis, or penile
shaft (eg, balanitis xerotica obliterans, lichen sclerosis),
presence of smegma (a whitish substance that accumulates underneath the foreskin), tobacco abuse, or history of psoriasis treatment with ultraviolet light.4,5 Tumor
grade and stage, basaloid or sarcomatoid histological
Cancer Control 409
features, and the presence and extent of lymph-node
(LN) involvement in the groin or pelvis have all been
identified as important prognostic factors for penile
cancer in determining long-term, cancer-related survival outcomes.6 In addition to disease-specific characteristics, significant demographical, racial/ethnic, social,
and economic disparities exist with regard to both the
incidence and survival rates of penile cancer.7 This is
especially important considering the diverse patient
population in which this disease presents.2 These findings also highlight the importance of considering these
differences when developing health care policies to
help improve access to standard medical care and reduce the treatment gap in cancer management.
Methods
A search of the medical literature was performed for
research published between January 1990 and December 2015. Case reports and non–English-language articles were excluded, focusing instead on large, population-based studies from national or international
datasets. A total of 54 studies were initially identified,
30 of which are included in this review.
Incidence
The incidence of penile cancer varies within specific racial/ethnic and socioeconomic groups, and a
summary of the reviewed literature analyzing disparities in penile cancer incidence based on large population-based data sets is shown in Table 1.8-10
Hernandez et al8 examined the burden of invasive squamous cell carcinoma (SCC) of the penis in
the United States from 1998 to 2003 based on data
from the Surveillance, Epidemiology, and End Results
Table 1. — Overview of the Disparities in Incidence of Penile
Cancer in Select Population-Based Studies
Study
Colón-López
Data Set
Select Findings
Puerto Rico
Cancer Registry
SEER program
Puerto Rican men had higher
rates of penile cancer than other
groups
Lower socioeconomic status
and educational level associated
with higher incidence rates in
Puerto Rican men
Goodman9
US populationbased registries
Hispanic and African American men
had highest rates of penile cancer
African American men diagnosed
at advanced stage with more nodepositive disease
Highest incidence in the southern
United States
Hernandez8
SEER program
Penile cancer rates higher in
Hispanics and those living in regions
of lower socioeconomic status or
in the southern United States
10
SEER = Surveillance, Epidemiology, and End Results.
410 Cancer Control
(SEER) program of the National Cancer Institute. The
annual, average age-adjusted incidence rate of penile SCC was 0.81 cases per 100,000 men, and rates
steadily increased with age.8 The incidence of penile
SCC was comparable in whites and African Americans,
but it was approximately twofold lower in Asians and
Pacific Islanders.8 Rates among Hispanics were 72%
higher compared with non-Hispanics, which may be
attributable to a decreased prevalence of circumcision in this ethnic group.11 From 1993 to 2002, white
Hispanics had the highest incidence rates of penile
cancer (1.01 per 100,000 men) followed by Alaska Natives/American Indians (0.77 per 100,000 men) and African Americans (0.62 per 100,000 men).11 Incidence of
penile SCC was also elevated in the southern United
States and in regions of lower socioeconomic status.
However, the study had limitations because the education level of participants was not analyzed.
Colón-López et al10 compared the incidence of penile cancer in Puerto Rico with other racial and ethnic
groups in the United States. Puerto Rican men had a
higher incidence of penile cancer than non-Hispanic
white men (standardized rate ratio [SRR] 3.33), nonHispanic African American men (SRR 3.04), and other US Hispanic men (SRR 2.59).10 This difference was
more pronounced for Puerto Rican men younger than
60 years of age when compared with similarly aged US
counterparts.10 Puerto Rican men with the lowest socioeconomic status had a 70% higher incidence rate of
penile cancer than Puerto Rican men with the highest
socioeconomic status (SRR 1.70).10 Less education
(< 12 years of education) was also statistically associated with a higher incidence rate of penile cancer among
Puerto Rican men (SRR 2.18).10 However, the overall
rate of penile cancer in Puerto Rico declined over time.
In other studies, Hispanic and African American
men have been reported to have the highest age-adjusted rates of penile cancer compared with whites,
Asians, or American Indians.9,12 The highest rates of
penile cancer were observed in Hispanic and African
American men older than 85 years of age, but penile
cancer was rare among males younger than 20 years of
age. Analyses to assess temporal trends from 1995 to
2003 revealed a statistically significant decline in the
incidence of penile cancer for African Americans and
whites. Approximately 60% of penile cancers were diagnosed at a localized stage (≤ pT2) among all racial
and ethnic groups, although Hispanic and African
American men tended to be diagnosed at an advanced
stage (≥ pT3) with a higher incidence of node-positive
disease than whites.9 Racial and ethnic differences in
tumor grade were not found. The incidence of penile
cancer was highest in the southern United States and
lowest in the western states. The highest age-adjusted
incidence rate of penile SCC was seen in African American men living in the southern United States, and the
October 2016, Vol. 23, No. 4
lowest rate was found among Asian-Pacific Islanders
living in western states.9 The incidence of primary
malignant penile cancer has decreased significantly over time from 1973 to 2002 based on data from
SEER; however, the incidence of nodal disease has increased over time.13
The overall incidence of penile cancer in the
United Kingdom (England and Wales) is higher than
in Australia and the United States, although the incidence of penile cancer in all 3 countries was stable
over time.14 Although cancer-specific mortality rates
were higher in England and Wales compared with
Australia and the United States, the incidence ratios were similar for all 3 countries.14 In Finland, the
age-adjusted incidence rate of penile cancer has decreased from the 1960s onward.15 Penile cancer was
primarily a disease of elderly men during this time
in Finland, and no systematic geographical variation
or predilection in terms of incidence was observed.15
The most common predisposing risk factors in Finland
were phimosis (44% of cases) and condylomatous
genital lesions (20% of cases).15
Human Papillomavirus Infection
An estimated 40% to 80% of invasive penile cancers
have been linked to HPV infection.16-18 Warty and basaloid penile carcinomas have been associated with
HPV infection, particularly HPV types 16 and 18, more
so than typical SCC or verrucous carcinoma of the penis.19,20 No significant association between HPV status
and pathological tumor stage, grade, or LN status was
found in this study.19
The impact of HPV infection on the prognosis of
patients with penile cancer is still controversial. Lont
et al21 reported an association between high-risk HPV
infection and reduced disease-specific mortality from
penile cancer (hazard ratio [HR] 0.14), whereas Hernandez et al16 observed no association between HPV
status and rate of survival in patients with penile cancer. HPV-associated penile malignancies, similar to
other nonviral penile cancers, have been associated
with lower education level (incidence density ratio
1.32; P = .0002) and income status (incidence density
ratio 1.59; P < .0001).22 Current and former smokers
were significantly more likely to have an α-HPV infection than any other type.23
In a study by Chaux et al3 of 103 Paraguayans with
penile cancer, patients with penile cancer were generally found to dwell in rural or suburban areas (82%),
live below the poverty line (75%), have a lower level of
education (91%), and have a significant smoking history (76%). Phimosis (57%), moderate or poor hygienic
habits (90%), and a history of sexually transmitted infections (74%) were also observed.3 Patients with more
than 10 lifetime female sexual partners were nearly
4 times as likely to present with HPV-positive penile
October 2016, Vol. 23, No. 4
tumors when compared with patients who had fewer
than 6 lifetime female sexual partners.3 However, this
trend was not significant when the number of sexual
partners was adjusted for age at first coitus and past
history of sexually transmitted infections. HPV-positive tumors were found in 36% of cases studied, and
they were characterized by warty or basaloid morphology in addition to a high histological grade in the majority of cases.3
Treatment
The management of penile carcinoma has evolved
in recent years, resulting in a shift in the treatment of
the disease.24 Options for the treatment of localized
disease have expanded to organ-sparing therapy, and
treatment of advanced disease has developed into a
multidisciplinary, team-based approach using multimodal therapies.24 Demographical, racial/ethnic, and
socioeconomic disparities can influence the treatments
offered for patients with penile cancer in different populations (Table 2).25,26
Zhu et al26 used data from SEER to identify individuals diagnosed with penile SCC from 1998 to 2009
and treated with either local tumor excision or partial or total penectomy for their primary tumor. Of the
1,292 eligible patients, 24.2% underwent local tumor
excision for surgical treatment of penile cancer.26
For stage T1 disease, the rates of local tumor excision increased from 29% to 40% within 10 years.26
Following a multivariable analyses, age younger than
65 years, African American descent, tumor size of
less than 3 cm, and pathological stage disease of no
higher than T1 were reported predictors for the use
of local tumor excision as surgical treatment of the
primary penile tumor as part of a penile-sparing approach.26 The authors concluded that penile-sparing
surgery is underutilized in the general population,
and significant age and racial/ethnic disparities exist
in its use.26
Other studies have noted that the majority of paTable 2. — Overview of the Disparities in Treatment for
Penile Cancer in Select Population-Based Studies
Study Data Set
Zhu
Select Findings
25
SEER program
Men < 50 y of age more likely to receive
extensive LN dissection (≥ 8 LNs removed)
during inguinal LN dissection for high-risk
penile cancer
Zhu26
SEER program
More likely to receive penile-sparing approach
for surgical treatment of primary tumor:
Age < 65 y
African American decent
Tumor size < 3 cm
Pathological stage ≤ T1
LN = lymph node, SEER = Surveillance, Epidemiology, and End Results.
Cancer Control 411
tients identified with penile SCC in the SEER data set
were treated with surgical therapy, and a small percentage of patients received radiotherapy alone or as
adjuvant therapy.27,28 No statistically significant sociodemographic or racial/ethnic differences were reported with regard to radiotherapy for penile cancer
due to small sample size.28
Zhu et al25 also assessed the impact of patient
characteristics on the extent of inguinal LN dissection when it is clinically indicated for patients with
high-risk penile SCC. Using SEER data, the authors
identified 393 patients who underwent regional LN dissection for penile cancer; this study cohort was then
stratified into 2 groups, limited LN dissection (< 8 LNs
removed) and extensive LN dissection (≥ 8 LNs removed).25 The median number of dissected LNs in this
series was 15, and 28% of patients underwent limited
LN dissection.25 The prevalence of extensive LN dissection decreased in tandem with increasing age (81% in
men aged < 50 years to 65% in men aged ≥ 70 years).25
On multivariable analysis, age alone was the independent predictor of extensive LN dissection (odds
ratio 0.98; P = .01).25 Inadequate LN dissection with few
LNs removed was observed in some patients with penile cancer, particularly among elderly men, even despite evidence that inguinal lymphadenectomy has a
similar morbidity rate in this age group compared with
men younger than 65 years of age.25,26
Table 3. — Overview of the Disparities in Survival Rates of
Penile Cancer in Select Population-Based Studies
Study
Data
Set
Findings
Hernandez8
SEER
program
Penile SCC mortality elevated in:
African American men
Hispanic men
Elderly (age > 80 y)
Southern US residents
Low-income areas
Less-educated populations
Rippentrop27
SEER
program
African American race independent
predictor of worse cancer-specific survival
Sharma31
NCDB
African American race independent
predictor of worse OS
Private insurance and higher median
income independent predictors of better OS
Thuret 32
SEER
program
Single marital status independent predictor of:
Locally advanced primary tumor
Higher-grade disease
Increased overall mortality
Tyson30
SEER
program
Tumors on penile shaft and overlapping
lesions increased cancer-specific mortality vs preputial lesions
Verhoeven3
SEER
program
5-year cancer-specific survival of penile
cancer in United States decreased from
1998 to 2011
NCDB = National Cancer Database, OS = overall survival,
SEER = Surveillance, Epidemiology, and End Results.
Survival
Factors affecting health insurance coverage and access to early detection and treatment could influence
the cancer-specific mortality rate of penile cancers. A
summary of the literature analyzing disparities in penile cancer survival rates based on select, large population-based data sets is shown in Table 3.3,8,27,30-32
Rippentrop et al27 first described the racial and
socioeconomic disparity in survival from penile carcinoma based on data from SEER. They reported that
African American men presented at a younger age and
with a higher stage of disease compared with other
racial groups. The disease-specific risk of death for
African American men was also nearly twice as high
than for whites (P = .0023), and marriage was associated with improved disease-specific rates of survival in
the localized (P = .0002) and regional stages (P = .001)
of disease.27 Multivariable analysis demonstrated that a
higher stage at diagnosis, age older than 65 years, African American race, and positive LNs were all significant independent predictors of cancer-specific survival
from penile carcinoma.27
Thuret et al32 also used SEER data to evaluate the
association between marital status, tumor stage and
grade, overall mortality, and cancer-specific mortality in patients with penile SCC. Multivariable logistic regression models revealed that unmarried men
412 Cancer Control
had a 1.5-fold higher risk of having locally advanced
(pT3/T4, N+, M+) or higher grade (grade 3/4) disease
at the time of surgery (P < .001) and a 1.3-fold higher
risk of overall mortality (P = .001); however, marital
status had no effect on cancer-specific mortality.32 Age,
socioeconomic status (median family income above
or below the poverty line within the patient’s county
of residence), and race (white vs African American vs
other) did not correlate with rates of locally advanced
or higher-grade disease and cancer-specific mortality.32
However, the study was limited by its small sample size
of African Americans (n = 183).
In a multivariable competing-risk model, Thuret
et al33 studied a population of patients with cN0T1 penile SCC after primary tumor excision and before initial LN dissection and found no statistically significant
association between race and age (≥ 70 vs ≤ 69 years)
for cancer-specific mortality after accounting for
tumor grade. Tyson et al 30 also found no significant association between age and penile SCC-specific mortality rate after adjusting for tumor grade
based on SEER data. Penile tumors on the body of
the penis (HR 1.61) and overlapping lesions (HR
1.79; P = .01) had increased cancer-specific mortality
compared with preputial lesions.30 Furthermore, the
disease-specific 10-year survival rate of those with
October 2016, Vol. 23, No. 4
preputial tumors was 89.4% compared with 78.7% for
tumors at other locations combined (P < .0001).30
Hernandez et al8 analyzed more than 4,900 cases
of histologically confirmed invasive penile SCC in the
United States using SEER data, the Centers for Disease
Control and Prevention’s National Program for Cancer
Registries, and the National Center for Health Statistics. The authors found that the rate of mortality due
to penile SCC increased with age (mortality rate at age
< 50 years: 0.02 vs age > 80 years: 1.80).8 African American men were also diagnosed at a significantly younger age than white or Asian men (median age, 62 vs 68
vs 68 years, respectively) and had a lower incidence-tomortality ratio than white or Asian men (3.44 vs 5.18
vs 5.63, respectively; P < .01 for all).8 This was also true
for Hispanic men compared with non-Hispanic men
(median age, 58 vs 69 years; incidence-to-mortality ratio: 4.61 vs 5.25; P < .01).8 Rate of mortality due to penile SCC was also increased among those men living in
southern states, in regions of lower socioeconomic status, and among those living in areas with lower levels
of educated people.8
Sharma et al 31 used the National Cancer Database (NCDB) to study patients with a diagnosis of
penile SCC from 1998 to 2011, and they found that
African American and Hispanic men were diagnosed at a younger age (< 55 years: 38.3% vs 25.9%
and 51.0% vs 24.5%; P < .01), and African Americans
presented with a higher stage of disease (pT3/T4:
16.6% vs 13.2%; P = .027) and had worse median overall survival (OS) rates (68.6 vs 93.7 months; P < .01).
The estimated median OS rate in the entire cohort
was 91.9 months at a median follow-up period of
44.7 months, and survival did not change over the
study period.31 Hispanic ethnicity was not associated
with all-cause mortality in the penile cancer population.31 Location and education level also did not have
a significant relationship with OS. Patients with private health insurance and a median income of at least
$63,000 based on zip code presented with a lower
stage of disease (pT3/T4: 11.6% vs 14.7%, P = .002;
12.0% vs 14.0%, P = .042) and had better median rates
of OS (163.2 vs 70.8 months, P < .01; 105.3 vs 86.4
months, P = .001).31 Multivariable analysis revealed that
African American race (HR 1.39; 95% confidence interval [CI]: 1.21–1.58; P < .01) was independently associated with worse OS, whereas private insurance (HR 0.79;
95% CI: 0.63–0.98; P = .028) and higher median income
of at least $63,000 (HR 0.82; 95% CI: 0.72–0.93; P = .001)
were independently associated with better OS.31
Colón-López et al10 compared the mortality rates
of penile cancer in Puerto Rican men with other racial and ethnic groups in the United States and evaluated the influence of socioeconomics and educational
level on the rates of mortality observed in Puerto Rico.
From 2000 to 2004, Puerto Rican men had a statistiOctober 2016, Vol. 23, No. 4
cally significant higher mortality rate when compared
with non-Hispanic white men (HR 3.32), non-Hispanic
African American men (HR 2.51), and other US Hispanic men (HR 1.89).10 A higher rate of mortality related
to penile cancer was also observed among Puerto Rican men in the lowest socioeconomic status (HR 1.61)
when compared with Puerto Rican men in the highest
socioeconomic status, although this difference was not
statistically significant.10 An increasing trend in mortality due to penile cancer was observed among men in
Puerto Rico due to a possible convergence of early and
high levels of sexual activity, low circumcision rates,
and high prevalence rates of sexually transmitted infections in Puerto Rico — particularly among the medically underserved.
Paiva et al34 reported an increased cancer-specific mortality rate in patients younger than 40 years of
age at a mean follow-up period of 22.4 months (19%)
compared with patients aged between 40 and 60 years
(11%) and those older than 60 years of age (13%;
P < .05). Younger patients also had an increased likelihood of perineural invasion (26% vs 13%) and infiltrative growth pattern (70% vs 50%) on pathological
review, and they had a higher risk of recurrence
(45% vs 25%; P < .05 for all).34 Study findings from
Sharma et al31 similarly showed an increased prevalence of node-positive disease in patients younger than
55 years of age compared with those older than
75 years (21.5% vs 13.1%; P < .01) but a decreased prevalence of pT3 to T4 disease (12.0% vs 15.4%; P = .007).
Others have also shown younger men (< 50 years) were
more likely to receive more extensive LN dissection
(> 8 LNs removed) than older men (> 70 years;
81% vs 65%; P = .01) with better rates of cancer-specific survival (P = .006).25 These findings remained
true even in the subgroup of study patients with
node-positive penile cancer (P = .01).25 Older age was
an independent predictor of OS in the populationbased study using the NCDB on multivariable analysis
(P < .01), but its relationship with cancer-specific mortality is unknown.31
Large population-based data have also been used
to demonstrate penile cancer survival trends over time.
The NCDB study did not show a correlation between
the year of diagnosis and OS during the period from
1998 to 2011.31 However, Verhoeven et al35 reported
that the 5-year cancer-specific survival rate of patients with penile cancer in the United States has significantly decreased from 72% to 63% based on SEER
data, and they also confirmed a significantly increasing relative risk of death with increasing age (relative
increase risk of death = 1.15; P < .01). Other groups
have also observed an increase in the incidence of
regional-stage disease (pN+) over time in US men that
also paralleled rising age.36 Furthermore, Arya et al37
reported on more than 9,500 men diagnosed with priCancer Control 413
mary penile cancer in England from 1979 to 2009 and
noted that the mortality rates fell by 20% after 1994
(from 0.39 to 0.31 per 100,000 men). The 5-year survival rate they observed also increased from 61.4% to
70.2%, and the 5-year survival rate for men diagnosed
between 2006 and 2010 was 77% for men younger than
60 years of age and 53% for men older than 80 years
of age.37 The 8% difference in 5-year survival rate (66%
vs 74%) between men living in the most affluent vs the
most deprived areas during this time was not statistically significant.37 The authors propose that the 21%
increase in incidence of penile cancer since the 1970s
in England could be explained by changes in sexual
habits, greater exposure to sexually transmitted infections, such as HPV infection, and the decreasing rates
of childhood and neonatal circumcision.37 They also
suggest that the improvement in survival may be due
to advances in medical care, including diagnostic, staging, and surgical techniques.37
Conclusions
Disparities exist in the incidence, treatment, and outcomes of penile carcinoma. Younger patients are often
offered more penile-sparing approaches for primary
tumors but more aggressive treatment such as lymphadenectomy for nodal disease. African American men,
Hispanics, the uninsured, those who are less educated, and those with lower socioeconomic status appear
to have lower rates of survival when compared with
their insured, educated, white, non-Hispanic counterparts. Marital status, number of sexual partners, and
human papillomavirus status also seem to play a role
in prognosis. Although the treatment options and rates
of survival from penile squamous cell carcinoma have
improved during the past several decades, further understanding of the racial/ethnic, socioeconomic, and
age-related differences seen in penile malignancies
may help minimize future disparities in cancer care.
References
1. American Cancer Society. Cancer Facts and Figures 2016. http://
www.cancer.org/acs/groups/content/@research/documents/document/
acspc-047079.pdf. Accessed October 4, 2016.
2. Bleeker MC, Heideman DA, Snijders PJ, et al. Penile cancer: epidemiology, pathogenesis and prevention. World J Urol. 2009;27(2):141-150.
3. Chaux A, Netto GJ, Rodriguez IM, et al. Epidemiologic profile, sexual
history, pathologic features, and human papillomavirus status of 103 patients
with penile carcinoma. World J Urol. 2013;31(4):861-867.
4. Stern RS, Bagheri S, Nichols K, et al. The persistent risk of genital
tumors among men treated with psoralen plus ultraviolet A (PUVA) for psoriasis.
J Am Acad Dermatol. 2002;47(1):33-39.
5. Tsen HF, Morgenstern H, Mack T, et al. Risk factors for penile
cancer: results of a population-based case-control study in Los Angeles
County (United States). Cancer Causes Control. 2001;12(3):267-277.
6. Jayaratna IS, Mitra AP, Schwartz RL, et al. Clinicopathologic characteristics and outcomes of penile cancer treated at tertiary care centers
in the Western United States. Clin Genitourin Cancer. 2014;12(2):138-142.
7. Pow-Sang MR, Ferreira U, Pow-Sang JM, et al. Epidemiology and
natural history of penile cancer. Urology. 2010;76(2 suppl 1):S2-S6.
8. Hernandez BY, Barnholtz-Sloan J, German RR, et al. Burden of invasive squamous cell carcinoma of the penis in the United States, 1998-2003.
Cancer. 2008;113(10 suppl):2883-2891.
9.Goodman MT, Hernandez BY, Shvetsov YB. Demographic and
414 Cancer Control
pathologic differences in the incidence of invasive penile cancer in
the United States, 1995-2003. Cancer Epidemiol Biomarkers Prev.
2007;16(9):1833-1839.
10. Colón-López V, Ortiz AP, Soto-Salgado M, et al. Penile cancer disparities in Puerto Rican men as compared to the United States population.
Int Braz J Urol. 2012;38(6):728-738.
11. Laumann EO, Masi CM, Zuckerman EW. Circumcision in the United
States. Prevalence, prophylactic effects, and sexual practice. JAMA.
1997;277(13):1052-1057.
12. Slopnick EA, Kim SP, Kiechle JE, et al. Racial disparities differ for
African Americans and hispanics in the diagnosis and treatment of penile
cancer. Urology. 2016;96:22-28.
13. Barnholtz-Sloan JS, Maldonado JL, Pow-sang J, et al. Incidence
trends in primary malignant penile cancer. Urol Oncol. 2007;25(5):361-367.
14. Sewell J, Ranasinghe W, De Silva D, et al. Trends in penile cancer:
a comparative study between Australia, England and Wales, and the US.
Springerplus. 2015;4:420.
15. Maiche AG. Epidemiological aspects of cancer of the penis in Finland.
Eur J Cancer Prev. 1992;1(2):153-158.
16. Hernandez BY, Goodman MT, Unger ER, et al. Human papillomavirus
genotype prevalence in invasive penile cancers from a registry-based
United States population. Front Oncol. 2014;4:9.
17. Kirrander P, Kolaric A, Helenius G, et al. Human papillomavirus
prevalence, distribution and correlation to histopathological parameters in a large Swedish cohort of men with penile carcinoma. BJU Int.
2011;108(3):355-359.
18. Miralles-Guri C, Bruni L, Cubilla AL, et al. Human papillomavirus prevalence and type distribution in penile carcinoma. J Clin Pathol.
2009;62(10):870-878.
19. Gregoire L, Cubilla AL, Reuter VE, et al. Preferential association of
human papillomavirus with high-grade histologic variants of penile-invasive
squamous cell carcinoma. J Natl Cancer Inst. 1995;87(22):1705-1709.
20. Rubin MA, Kleter B, Zhou M, et al. Detection and typing of human
papillomavirus DNA in penile carcinoma: evidence for multiple independent
pathways of penile carcinogenesis. Am J Pathol. 2001;159(4):1211-1218.
21. Lont AP, Kroon BK, Horenblas S, et al. Presence of high-risk human
papillomavirus DNA in penile carcinoma predicts favorable outcome in survival.
Int J Cancer. 2006;119(5):1078-1081.
22. Benard VB, Johnson CJ, Thompson TD, et al. Examining the association between socioeconomic status and potential human papillomavirusassociated cancers. Cancer. 2008;113(10 suppl):2910-2918.
23. Sichero L, Pierce Campbell CM, Fulp W, et al. High genital prevalence of cutaneous human papillomavirus DNA on male genital skin: the
HPV Infection in Men Study. BMC Infect Dis. 2014;14:677.
24. Sharma P, Zargar-Shoshtari K, Spiess PE. Current surgical management of penile cancer. Curr Probl Cancer. 2015;39(3):147-157.
25. Zhu Y, Gu CY, Ye DW. Population-based assessment of the number
of lymph nodes removed in the treatment of penile squamous cell carcinoma.
Urol Int. 2014;92(2):186-193.
26. Zhu Y, Gu WJ, Wang HK, et al. Surgical treatment of primary disease
for penile squamous cell carcinoma: a Surveillance, Epidemiology, and
End Results database analysis. Oncol Lett. 2015;10(1):85-92.
27. Rippentrop JM, Joslyn SA, Konety BR. Squamous cell carcinoma
of the penis: evaluation of data from the surveillance, epidemiology, and
end results program. Cancer. 2004;101(6):1357-1363.
28. Burt LM, Shrieve DC, Tward JD. Stage presentation, care patterns,
and treatment outcomes for squamous cell carcinoma of the penis. Int J
Radiat Oncol Biol Phys. 2014;88(1):94-100.
29. Gopman JM, Djajadiningrat RS, Baumgarten AS, et al. Predicting
postoperative complications of inguinal lymph node dissection for penile
cancer in an international multicentre cohort. BJU Int. 2015;116(2):196-201.
30. Tyson MD, Etzioni DA, Wisenbaugh ES, et al. Anatomic site-specific
disparities in survival outcomes for penile squamous cell carcinoma. Urology.
2012;79(4):804-808.
31. Sharma P, Ashouri K, Zargar-Shoshtari K, et al. Racial and economic
disparities in the treatment of penile squamous cell carcinoma: results from
the National Cancer Database. Urol Onc. 2016;34(3):122.
32. Thuret R, Sun M, Budaus L, et al. A population-based analysis of
the effect of marital status on overall and cancer-specific mortality in patients
with squamous cell carcinoma of the penis. Cancer Causes Control.
2013;24(1):71-79.
33. Thuret R, Sun M, Abdollah F, et al. Competing-risks analysis in patients
with T1 squamous cell carcinoma of the penis. BJU Int. 2013;111(4 pt B):E174-E179.
34. Paiva GR, de Oliveira Araújo IB, Athanazio DA, et al. Penile cancer:
impact of age at diagnosis on morphology and prognosis. Int Urol Nephrol.
2015;47(2):295-299.
35. Verhoeven RH, Janssen-Heijnen ML, Saum KU, et al. Populationbased survival of penile cancer patients in Europe and the United States of
America: no improvement since 1990. Eur J Cancer. 2013;49(6):1414-1421.
36. Barnholtz-Sloan JS, Maldonado JL, Pow-Sang J, et al. Incidence
trends in primary malignant penile cancer. Urol Oncol. 2007;25(5):361-367.
37. Arya M, Li R, Pegler K, et al. Long-term trends in incidence, survival
and mortality of primary penile cancer in England. Cancer Causes Control.
2013;24(12):2169-2176.
October 2016, Vol. 23, No. 4
The underlying biology of carcinogenesis
should be addressed in African American
men at high risk for prostate cancer.
Rose. X-Ray image courtesy of Bryan Whitney. www.x-rayphotography.com.
Chemoprevention in African American Men With
Prostate Cancer
Nagi B. Kumar, PhD, RD, Julio M. Pow-Sang, MD, Philippe E. Spiess, MD, Jong Y. Park, PhD,
Ganna Chornokur, PhD, Andrew R. Leone, MD, and Catherine M. Phelan, PhD, MD
Background: Recommendations for cancer screening are uncertain for the early detection or prevention of
prostate cancer in African American men. Thus, chemoprevention strategies are needed to specifically target
African American men.
Methods: The evidence was examined on the biological etiology of disparities in African Americans related
to prostate cancer. Possible chemopreventive agents and biomarkers critical to prostate cancer in African
American men were also studied.
Results: High-grade prostatic intraepithelial neoplasia may be more prevalent in African American men,
even after controlling for age, prostate-specific antigen (PSA) level, abnormal results on digital rectal examination, and prostate volume. Prostate cancer in African American men can lead to the overexpression of
signaling receptors that may mediate increased proliferation, angiogenesis, and decreased apoptosis. Use of
chemopreventive agents may be useful for select populations of men.
Conclusions: Green tea catechins are able to target multiple pathways to address the underlying biology of
prostate carcinogenesis in African American men, so they may be ideal as a chemoprevention agent in these
men diagnosed with high-grade prostatic intraepithelial neoplasia.
From the Cancer Epidemiology (NBK, JYP, GC, CMP) and Genitourinary Oncology Programs (NBK, JMP-S, PES), H. Lee Moffitt Cancer
Center & Research Institute, Tampa, Florida, and the Departments
of Oncologic Sciences (NBK, PES, JYP, GC, CMP), Urology (PES, ARL),
Molecular Medicine (JYP), Cell, Molecular and Microbiology (JYP),
and Epidemiology and Biostatistics (JYP), University of South Florida, Tampa, Florida.
Submitted March 1, 2016; accepted July 6, 2016.
Address correspondence to Nagi B. Kumar, PhD, RD, Department
of Epidemiology, Moffitt Cancer Center, 12902 Magnolia Drive,
MRC-CAN CONT, Tampa, FL 33612. E-mail: [email protected]
No significant relationships exist between the authors and the
companies/organizations whose products or services may be referenced in this article.
The development of this manuscript was supported in part by
the National Cancer Institute (grants nos. 1R21CA179659 and
R01CA128813). The work was made possible by research funding
awarded by the National Institute of Health and the National Cancer
Institute (grant no. R01 CA12060-01A1).
October 2016, Vol. 23, No. 4
Introduction
Despite treatment advances in recent years, prostate
cancer remains the leading cause of cancer-related
death among men in the United States.1 In 2016, the
American Cancer Society estimates that 180,890 new
cases of prostate cancer will be diagnosed in the
United States, and 26,120 men will die from the disease.1 Racial and geographical differences have been
observed, including a 40-fold difference in incidence
rates between low-risk (Chinese men) and high-risk
populations (African American men).2 In the United
States, age-standardized incidence rates for prostate
cancer are 272 per 100,000 men for African Americans
compared with 164 per 100,000 men for whites.2 Compared with white men, African American men with
Cancer Control 415
prostate cancer have more aggressive disease, higher
rates of incidence, are diagnosed at a younger age,
present with more advanced disease at diagnosis, and
have a worse prognosis.3 The mortality rate for African
American men with prostate cancer is 3 times higher
than white men, although the rate decreased in both
groups between 1990 and 2000.2
With disparate rates of prostate cancer as well as
aggressive prostate cancer observed in African American men, the logical approach is to focus efforts on
prostate cancer screening for early detection and prevention.4,5 However, uncertainties still exist about the
overall value of early detection. Although periodic testing for serum concentrations of prostate-specific antigen (PSA) may reduce the mortality of prostate cancer,
PSA testing has been linked to increased diagnoses
and overtreatment of clinically insignificant, potentially indolent tumors that pose little risk of metastasis or
death.5,6 Taking the accumulated evidence into consideration, the US Preventive Services Task Force recommended against PSA-based prostate cancer screening
in asymptomatic men.7 However, these recommendations note that, in African American men, firm conclusions cannot be made to balance the risks and benefits
of such screening because data do not exist to support
a more favorable risk–benefit ratio.7 Two cancer screening trials included a majority of men with European
ancestry, thus largely precluding conclusions specifically pertaining to men of African descent.6,8 Hence,
no specific evidence-based recommendation regarding prostate cancer screening exists for the high risk
population of African American men, underscoring the
need for chemoprevention strategies targeting African
American men.2,3
Biological Etiology of Disparity
The etiology of increased susceptibility of African
American men to prostate cancer has not been elucidated, but it is likely multifactorial involving genetic,
biological, sociocultural, and lifestyle determinants.
The initiation and progression of prostate cancer involves a complex series of events. During progression,
genetic changes and loss of cellular control are observed as cell phenotypes change from normal to dysplasia (prostatic intraepithelial neoplasia), severe dysplasia (high-grade prostatic intraepithelial neoplasia
[HGPIN]), clinically localized disease, and to metastatic
disease.9-12 Observations point to the role of genetic
susceptibility factors in human prostate cancer.9-15 Other than older age, African ancestry and family history
of prostate cancer are well-established, nonmodifiable
risk factors for prostate cancer.9-13 The lifetime risk of
prostate cancer increases 1.5- to 4-fold in men with 1 or
2 first-degree relatives with prostate cancer.14 A Scandinavian study of twins estimated that 42% of the observed rate of prostate cancer susceptibility was associ416 Cancer Control
ated with inherited genetic risk factors.15
Genetic risk factors are also significant at a younger age, and the attributed risk of inherited susceptibility is thought to be as high as 40% among men diagnosed with prostate cancer at 55 years or younger.13
Although genome-wide association studies (GWASs)
and large genetic variation studies have identified
more than 100 prostate cancer risk loci, elucidating
the biological basis for these associations is challenging.16-18 Identified risk loci include the noncoding variants, such as those located in the 8q24 region, as well
as polymorphisms in the gene-coding regions that either alter, or are predicted to alter, protein expression
(eg, HNF1B, TERT, RNASEL, KLK3).16,19-22
HNF1B single-nucleotide polymorphisms
(rs7501939 and rs4430796) have been identified in
GWASs of prostate cancer and are associated with a
risk of prostate cancer in African American men.23-25 In
addition, the CTBP2 single-nucleotide polymorphism
rs4962416 is associated with risk of prostate cancer in
white men.23-25 HNF1B encodes a transcription factor
protein that forms heterodimers with other members
of the HNF1 family and can influence the gene transcription. While it was previously believed that HNF1B
expression is specific to the liver, its transcripts have
been identified in various tissues, including bone marrow, and the pancreas, urinary tract, gastrointestinal
organs, and prostate.26 Mutations in HNF1B cause
type 5 maturity-onset diabetes that may be accompanied by urinary tract disorders.27,28 Some men with
HNF1B mutations have malformations in the reproductive tract, including epididymal cysts, agenesis of the vas
deferens, or infertility due to abnormal spermatozoa.29
Post-GWASs are suggestive of the interaction
between genetic variants and environmental risk
factors, but our understanding of this is still inadequate.30,31 Rs7501939 at HNF1B has been shown to
increase the risk of prostate cancer in African American men who are obese but not in African American
men who are not obese or European American men
of any body weight.23 Although these are preliminary
findings, they suggest that weight-loss interventions
may decrease the risk of prostate cancer in African
American men who are carriers of the high-risk allele
at rs7501939 of HNF1B. Future studies should further
examine this observation.
CTBP2 encodes a transcriptional co-repressor
activated under stress and can mediate the stress-induced migration of tumor cells.32 CTBP2 expression
is detected in the prostate and has been linked to decreased PTEN expression and activation of the phosphatidylinositol 3-kinase pathway, which may support
or promote the growth of prostate cancer.33
Lindquist et al34 reported a higher prevalence rate
of mutations in MUC3A and PRIM2 in African American men with prostate cancer when compared with
October 2016, Vol. 23, No. 4
their white counterparts. MUC3 is involved in cell signaling, growth, and survival; MUC3 and PRIM2 are
also associated with carcinogenesis.35 The different
patterns of somatic mutations observed between racial
groups may help provide a basis for understanding the
genomic contributions to aggressive tumors and associated disparate outcomes in African American men
with prostate cancer.
Evidence is also emerging for racial differences
in markers of prostate tumor subtypes. For example,
gene fusions in TMPRSS2 and ERG are found in prostate tumors.35-43 However, several studies report that
the prevalence of TMPRSS2–ERG fusions in the tumors
of African American men is lower than that observed
in white men.35-43 Thus, future studies must continue to
pursue this research because such racial differences in
marker expression could reveal mechanisms contributing to race disparities.
Differences exist in tumor biology between African
American men and white men and may be attributable to race-specific differences in tumor location
(ie, anterior vs posterior tumors). African Americans
have an increased incidence of prostate cancer (located anterior to the peripheral zone in radical prostatectomy specimens that may potentially have a prognostic significance.44 In a study of 1,245 patients who
underwent radical prostatectomy, the overall tumor
locations were anterior in 14%, posterior in 58%, or
both in 28% of cases.44 The incidence of anterior tumors was higher in African American men compared
with white men, and the rates of positive surgical
margins in anteriorly and posteriorly located tumors
were 60% vs 38% in African American men and 48%
vs 27% in white men, respectively.44
In patients with an abnormal level of serum PSA
and negative findings on sextant prostate biopsies,
biopsy is recommended for the anterior zone of the
prostate.45 In a study of 398 men, of whom 70% were
African American men, most patients had prostate
cancer limited to the peripheral zone or in both the
peripheral and anterior to the peripheral zone.46 For
4% of study patients, prostate cancer was limited to
the anterior to the peripheral zone — 5% of African
Americans vs 2% of those not of African American
descent — but this result was not statistically significant.46 Another study showed a biologically important
relationship between tumor location and molecular subtype, but racial differences in molecular subtypes did not persist when tumors were analyzed by
location.47 The study investigated triple-negative disease and AR signaling. African American men were
more likely to be positive for m-SPINK1+ and have
triple-negative disease than white men.47
Based on this evidence, the racial differences in
location of the prostate cancer do not appear to be
significant, nor are they likely to impact the progOctober 2016, Vol. 23, No. 4
nosis and management of prostate cancer in African
American men.
HGPIN is considered by many to be a premalignant lesion of prostate cancer.48-50 Data from some
studies suggest that the prevalence of HGPIN is greater
in African American men and that subgroups of African American men with HGPIN are more prone to the
development of aggressive, clinically significant cancer.51,52 HGPIN may also be a risk factor for biochemical recurrence following definitive treatment in African
American men.51,52 HGPIN is more prevalent in African American men, even after controlling for age, PSA
level, abnormal findings on digital rectal examination,
and prostate volume.51 Significantly higher prevalence
rates of HGPIN have been reported in African American men aged 40 to 49 years compared with white
men (46% vs 29%, respectively), suggesting that this
early-onset age range may represent the beginning of
a racial disparity related to prostate cancer; this is because more African American men with HGPIN go on
to develop prostate cancer.52 In concordance with these
data, Potts et al53 reported that African American men
were more likely to be diagnosed with prostatic intraepithelial neoplasia compared with white men, even
after adjusting for PSA levels.
In a study to investigate racial differences in tumor
burden (cancer volume, cancer percentage, and tumor
volume–PSA ratio) in a large cohort of men undergoing
radical prostatectomy, African Americans had higher
disease burden (estimated tumor volume, percent of
cancer involvement, estimated tumor volume–PSA
ratio) compared with non–African American men.54
This association was pronounced in low-grade cancer
(Gleason score ≤ 6), thus depicting a complex picture
of relations between race and tumor burden across the
aggressive spectrum of prostate cancer.54 The frequency of HGPIN and the autopsy prevalence of prostate
cancer have been reported to be similar in large studies comparing Asian and Western populations, with
widely differing incidence and mortality rates, suggesting an environmental influence on the expression of
this disease and the possibility of preventing disease
progression from HGPIN to prostate cancer through
pharmacological means.12,48-50,55,56 Targeting precursor lesions (HGPIN) that may display simpler genomic
aberrations relative to early stages of cancer in these
high-risk groups may offer more promise to develop
and test targeted interventions based on the biological
differences reported in prostate carcinogenesis among
African American men. Based on this evidence, African American men with biopsy-proven HGPIN are a
subset of men who can be considered to be at high risk
for prostate cancer and, thus, may be ideal candidates
for chemoprevention interventions.
Although PSA and its kinetics are used to monitor
disease progression, controversy exists regarding the
Cancer Control 417
efficacy of PSA level as a screening tool due to its low
rate of specificity; in addition, PSA levels are increased
in benign prostatic diseases, such as benign prostatic
hyperplasia (PSA concentrations can be ≤ 50% higher
in some cases).57 African American men continue to
have higher PSA levels and Gleason scores than white
men, despite a narrowing of the differences in pathological stage.58 One study reported that African American men with prostate cancer and a Gleason score of 6
produce less PSA than white men.59 African American
and white men had equal serum PSA and PSA masses
despite significantly larger prostates in African American men and all other parameters being the same.60
Another report showed that men with higher levels of
testosterone had higher levels of PSA even after taking
into account other hormones, age, and race/ethnicity.61
While transcription of the PSA gene is transactivated
by the androgen receptor (AR) with bound androgen,
it is unclear whether circulating androgens influence
circulating PSA levels. Men treated for prostate cancer
with androgen-deprivation therapy experience a decline in serum PSA concentration, but this decline may
be due to fewer cancer cells remaining to produce PSA
and reduced testosterone available to all PSA-producing cells.41 Despite these drawbacks related to specificity, serum PSA as a continuous variable — as well
as its doubling time and velocity — has been used in
prostate cancer chemoprevention trials and in clinical
practice to define risk categories.59,62-67
Other biological differences in benign tissue
and prostate tumors have been elucidated in African
American and white men alike. Prostate tumors derived from African American men overexpress signaling receptors that may mediate increased proliferation, including EGFR and AR.68,69 On average, AR has
a shorter polyglutamine repeat in African American
men and, thus, increased AR activity.11 Reports have
indicated decreased apoptosis and increased immunostaining for antiapoptotic protein B-cell lymphoma
2 (BCL2), suggesting that increased expression of
BCL2 may decrease apoptosis and increase rates of
tumor proliferation in African American men.70,71 Metastatic capacity may be higher in prostate tumor cells
in African Americans because of the overexpression
of several metastasis-related genes (AMFR, CXCR4,
CCR7, and MMP9) as well as KI67 and CAV1 in African American men.72-74 Prostate tumors obtained from
African American men may have decreased rates of
tumor suppression.
Decreased TCEAL7 expression has been observed
in tumors from African American men compared with
tumors from white men.75 Reams et al75 observed that
TCEAL7 expression in prostate cancer and in nonmalignant tissue was higher in white men compared with
African American men. Chien et al76 demonstrated
that TCEAL7 also inhibits the growth of ovarian can418 Cancer Control
cer cells, suggesting that TCEAL7 is a tumor-suppressor
gene and that varying expression levels of TCEAL7 between white and African American men may explain
some of the disparities in the growth patterns observed in prostate cancer–derived tumors cells in these
populations. Population studies comparing Japanese
and African Americans have observed that short CAG
repeats are more frequently associated with higher
transactivational function in African Americans, possibly explaining racial differences in the incidence rate
of prostate cancer between the 2 populations.11
Taken together, these data suggest that there may
be functional biological differences in the prostate tumors of African American men that may predispose
them to a biologically more aggressive malignancy.
However, these observations have not been validated
in clinical trials targeting African American men.
Current Approach
Chemoprevention refers to the inhibition of preinvasive and invasive cancer and its progression or
treatments of identifiable precancers.49,77 Chemoprevention efforts require an understanding of the
mechanism of carcinogenesis, including signaling, metabolic, and genetic progression pathways.
New technologies in genomics and proteomics have
spurred this field of research. Use of this knowledge
to develop pharmacological agents, botanicals, and
biologics to reverse or halt the process of carcinogenesis is called chemoprevention.
Agents for chemoprevention include antipromotion and antiprogression agents that prevent the
growth and survival of cells already committed to become malignant.49,77 Finasteride and dutasteride, which
block the conversion of testosterone to dihydrotestosterone, have been evaluated for chemoprevention
of prostate cancer in large phase 3 chemoprevention
trials.78-80 Although these agents significantly reduced
the risk of prostate cancer, their use was also associated with an increased rate of high-grade disease, severely limiting their clinical adoption and underscoring
the need to identify novel chemoprevention agents for
prostate cancer.79
Botanicals influence multiple biochemical and
molecular cascades that inhibit mutagenesis, proliferation, induce apoptosis, and suppress the formation
and growth of human cancers, thus modulating several
hallmarks of carcinogenesis. In addition, these agents
appear promising in their potential to impact the field
of cancer chemoprevention, because they have a significantly superior safety profile than most available
agents.81-88 Several botanicals have been characterized
and used for hundreds of years, although existing challenges and limitations have hampered progress in this
field.89,90 Multiple botanicals have been identified and
appear promising for the chemoprevention of prostate
October 2016, Vol. 23, No. 4
cancer.91-93 However, the slow pace of growth might
be attributed to the regulatory protection of classical
formulations and lack of standardization, quality control, a molecular mechanism-based approach in evaluation, a population-based normal range of biomarkers,
laboratory practices, and few translational scientists
engaged in conducting well-designed trials. However,
several valuable lessons have been learned from previous chemoprevention trials.91,94,95 Critical requirements
for moving botanicals from bench to bedside include
adopting a systematic, molecular-mechanism based
approach and utilizing the same ethical and rigorous
methods such as those used to evaluate other pharmacological agents.
Chemoprevention trials using combinations of
botanicals have demonstrated that synergy between
agents can lead to lower doses, improved rates of efficacy, and fewer or less-severe toxicities.96 An assessment of the end points of chemoprevention trials
whose results have been used to support approval of
an agent for prostate cancer revealed that nearly all
have been approved on the basis of intraepithelial neoplasia.97 Intermediate biomarker end points must be
identified, validated, and obtained using noninvasive
techniques without compromising the safety to men
in chemoprevention trials. To reduce patient burden,
these markers must be obtained from accessible organs and during the normal course of clinical surveillance. Randomized placebo control trials and the longterm follow-up and monitoring of patients and study
participants are critical to meet the requirements of the
US Food and Drug Association and to promote the acceptance of new agents into the marketplace.92,93
Green Tea Catechins
The Table summarizes the clinical, molecular, and biological characteristics relevant to prostate cancer in
African American men and compares them with the
effects of green tea catechins used to modulate those
characteristics.11,51,52,58,59,68-75,98-116
Laboratory studies have identified epigallocatechin gallate (EGCG) as a potent chemopreventive
agent because it affects various key molecular processes in prostate carcinogenesis that help induce
apoptosis and inhibit tumor growth and angiogenesis.117-121 Preclinical studies have demonstrated
chemopreventive efficacy in prostate cancer, showing that it has significant activity on prostate cancer
cells.99,122-129 Phase 1/2 studies have demonstrated the
bioavailability and tolerance of green tea catechin at
various ranges of doses of EGCG.64,93,98,100,130-136
Although several mechanisms exist by which
EGCG may operate in prostate carcinogenesis, EGCG
selectively inhibits the proteasome activity in intact
human prostate cancer cells and accumulates in IκBα
and p27 proteins, thus leading to growth arrest.119-121
High-grade green tea polyphenol extract, which is a
mixture of tea catechins with more than 50% of EGCG,
inhibits the proteasomal chymotrypsin-like activity
with a half maximal inhibitory concentration value
of 7 μM.93 The half maximal inhibitory concentration
value for trypsin-like activity is higher than 100 μM,
Table. — Summary of Relevant Characteristics of Prostate Cancer and the Effects of Green Tea Catechins in African Americans
Characteristic of Prostate Cancer
Effect of Green Tea Catechins
Clinical
Increased frequency of HGPIN51,52
Men with HGPIN had significantly reduced disease progression98,101
Higher PSA levels
Men with HGPIN had lower PSA levels than the control arm98,101
58
Higher Gleason scores at diagnosis59
Men given short-term daily doses had significant decreases in serum PSA levels100
Molecular
Increased Proliferation
Decreased Proliferation
EGFR overexpression68
In vitro models102-104
In vivo models99,105-107
AR overexpression69
Inhibition of EGFR signaling108
Increased androgen-receptor activity11
Reduced androgen-receptor signaling and activity 99,109-111
Decreased Apoptosis
Induced Apoptosis Induction
Increased expression of BCL2 70,71
Various mechanisms (eg, reduced BCL2 expression)112-114
Increased Metastatic Capacity
Inhibition of Invasion and Metastasis
Overexpression of AMFR, CXCR4, CCR7, MMP9, KI67,
and CAV1 72-74
Various mechanisms (eg, MMP9, MMP2 suppression)115
Decreased Tumor Suppression
Reactivation of Silenced Tumor-Suppressor Genes
Downregulation of TCEAL7 75
Restores balanced proliferation116
HGPIN = high-grade prostatic intraepithelial neoplasia, PSA = prostate-specific antigen.
October 2016, Vol. 23, No. 4
Cancer Control 419
demonstrating that high-grade green tea polyphenol
extract preferentially inhibits the proteasomal chymotrypsin-like activities over other activities.93
Kim et al126 investigated the safety and efficacy of
high-grade green tea polyphenol extract in an animal
model. Their goal was to reduce the progression of
prostate adenocarcinoma in transgenic mice. Mice
treated with the green tea extract had significantly
fewer tumors and decreased tumoral sizes compared
with the animals that did not receive the extract. The
high-grade green tea polyphenol extract also significantly inhibited metastasis in the treated mice in a
dose-dependent manner. Therefore, the data suggest
that high-grade green tea polyphenol extract is an effective chemopreventive agent in preventing the progression of prostate cancer to metastasis in a mouse
model.126 These findings provide evidence for the
safety and chemopreventive effect of this green tea
polyphenol extract for preventing metastatic spread
in prostate cancer.
Based on the results of previous studies, a placebocontrolled, randomized clinical trial was conducted
of the high-grade green tea extract in 97 men with HGPIN, atypical small acinar proliferation, or both conditions.64,92,119-121 No differences were observed in the
number of prostate cancer cases. The cumulative rate of
prostate cancer plus atypical small acinar proliferation
among men with HGPIN but without atypical small acinar proliferation at the start of the study was researched.
The data revealed a statistically significant decrease in
this composite end point: 3 of 26 (high-grade green tea
polyphenol extract gorup) vs 10 of 25 (placebo group).126
A decrease in diagnoses of atypical small acinar proliferation was also seen among those receiving the green tea
extract, as was a decrease in serum PSA level, compared
with those assigned to placebo.126
Intake of a standardized, decaffeinated green tea
catechin mixture every day for 1 year accumulated in
plasma and was well tolerated, and researchers saw
that its consumption significantly reduced the cumulative rates of prostate cancer and atypical small acinar
proliferation among men with HGPIN who did not
have atypical small acinar proliferation at baseline.137
Three of the 9 African American men in the study progressed to atypical small acinar proliferation or prostate cancer after 1 year, but no such progression was
seen among those assigned to the high-grade green tea
polyphenol extract arm.137 This disease progression
rate from HGPIN in the placebo arm was higher than
US norms and in white men assigned to the placebo
arm of this trial (20%).62,64,65,137,138 In men with a baseline diagnosis of atypical small acinar proliferation,
the diagnostic rates of cancer at 1 year were similar, regardless of treatment arm assignment or race.137 Overall, a significant increase in plasma EGCG concentration was observed, as was a reduction of serum PSA
420 Cancer Control
and disease progression from HGPIN to atypical small
acinar proliferation or prostate cancer with green tea
catechin treatment. Although these are early observations with small sample sizes, these data are provocative and warrant further evaluation in a well-powered
clinical trial targeting African American men with
HGPIN, because they suggest that green tea catechins
have the potential to decrease the risk of progression
to atypical small acinar proliferation and, subsequently, prostate cancer.137 However, the study was limited
by its small sample size of African American men, so
the researchers were unable to make comparisons
among white men with HGPIN in other blood and tissue-based biomarkers.
Experimental models to address the fundamental molecular pathways of green tea catechin have yet
to be validated in green tea catechin–treated tissue
samples from clinical trials.118 In addition to developing and refining the fundamental pathways of green
tea catechin, future studies evaluating these molecular pathways should define intermediate biomarkers of
prostate carcinogenesis. However, obtaining sufficient
tissue from prostate biopsies from precursor lesions
in prevention trials continues to present a challenge.
Therefore, it will be important to prioritize intermediate biomarkers available for evaluation based on the
robustness of evidence and relevance to the agent, the
potential molecular mechanism, and the patient cohort
and stage of disease targeted in future studies.92,93
Green tea catechins target every major clinical prostate cancer characteristic reported in African
American men (see Table).11,51,52,58,59,68-75,98-116 They may
do this by acting through the biological mechanisms
especially relevant to African American men, including reducing proliferation, inducing apoptosis, and
decreasing tumoral metastatic capacity. Thus, it is our
opinion that green tea catechins are a promising candidate for the chemoprevention of prostate cancer in
African American men.
Conclusions
Prostate cancer in African American men is a major
public health problem with significant morbidity and
mortality rates. A variety of laboratory approaches are
being applied to unravel the molecular pathogenesis of
prostate cancer. African American men with a family
history of prostate cancer, diagnosed with high-grade
prostatic intraepithelial neoplasia prior to age 50 years,
represent an ideal target population for chemoprevention. Green tea catechins have been characterized and
evaluated as agents able to target multiple pathways
that address the underlying biology of prostate carcinogenesis in African American men. Assessing the
efficacy of this intervention might eventually translate into clinical use in high-risk populations. It is our
opinion that the greatest challenge to evaluating promOctober 2016, Vol. 23, No. 4
ising agents for chemoprevention is the recruitment
and retention of African American men in randomized
clinical trials, which will require multiple approaches,
actions, and activities for achieving meaningful advances. The process is a complex and progressive one,
including the formation of trusting partnerships with
target populations and their circles of influence, power
sharing, priority setting, capacity building, education
and training, the transparent sharing of data and information, the development of goals, and insisting on
community involvement in the research.
References
1. American Cancer Society. Cancer Facts and Figures 2016. Atlanta, GA:
2016. http://www.cancer.org/acs/groups/content/@research/documents/
document/acspc-047079.pdf. Accessed October 12, 2016.
2. Centers for Disease Control and Prevention. Prostate cancer rates
by race and ethnicity. Updated June 16, 2016. http://www.cdc.gov/cancer/
prostate/statistics/race.htm. Accessed October 12, 2016.
3. Makridakis NM, Reichardt JK. Molecular epidemiology of hormonemetabolic loci in prostate cancer. Epidemiol Rev. 2001;23(1):24-29.
4. Chornokur G, Dalton K, Borysova ME, et al. Disparities at presentation,
diagnosis, treatment, and survival in African American men, affected by prostate
cancer. Prostate. 2011;71(9):985-997.
5. Powell IJ. Epidemiology and pathophysiology of prostate cancer in
African-American men. J Urol. 2007;177(2):444-449.
6. Ilic D, O’Connor D, Green S, et al. Screening for prostate cancer: an
updated Cochrane systematic review. BJU Int. 2011;107(6):882-891.
7. Moyer VA; US Preventive Services Task Force. Screening for prostate
cancer: U.S. Preventive Services Task Force recommendation statement.
Ann Intern Med. 2012;157(2):120-134.
8. Andriole GL, Crawford ED, Grubb RL, 3rd, et al. Mortality results
from a randomized prostate-cancer screening trial. N Engl J Med.
2009;360(13):1310-1319.
9. Lippman SM, Hong WK. Cancer prevention science and practice.
Cancer Res. 2002;62(18):5119-5125.
10. Montironi R, Mazzucchelli R, Lopez-Beltran A, et al. Mechanisms of
disease: high-grade prostatic intraepithelial neoplasia and other proposed
preneoplastic lesions in the prostate. Nat Clin Pract Urol. 2007;4(6):321-332.
11. Suzuki H, Ueda T, Ichikawa T, et al. Androgen receptor involvement in
the progression of prostate cancer. Endocr Relat Cancer. 2003;10(2):209-216.
12. Zlotta AR, Egawa S, Pushkar D, et al. Prevalence of prostate cancer
on autopsy: cross-sectional study on unscreened Caucasian and Asian
men. J Natl Cancer Inst. 2013;105(14):1050-1058.
13. Wolters T, Roobol MJ, Steyerberg EW, et al. The effect of study
arm on prostate cancer treatment in the large screening trial ERSPC. Int J
Cancer. 2010;126(10):2387-2393.
14. Cunningham GR, Ashton CM, Annegers JF, et al. Familial aggregation of prostate cancer in African-Americans and white Americans. Prostate.
2003;56(4):256-262.
15. Gibbs M, Stanford JL, Jarvik GP, et al. A genomic scan of families
with prostate cancer identifies multiple regions of interest. Am J Hum Genet.
2000;67(1):100-109.
16. Kote-Jarai Z, Olama AA, Giles GG, et al. Seven prostate cancer
susceptibility loci identified by a multi-stage genome-wide association
study. Nat Genet. 2011;43(8):785-791.
17. Eeles RA, Olama AA, Benlloch S, et al. Identification of 23 new
prostate cancer susceptibility loci using the iCOGS custom genotyping array.
Nat Genet. 2013;45(4):385-391, 391e1-391e2.
18. Freedman ML, Monteiro AN, Gayther SA, et al. Principles for the
post-GWAS functional characterization of cancer risk loci. Nat Genet.
2011;43(6):513-518.
19. Haiman CA, Patterson N, Freedman ML, et al. Multiple regions
within 8q24 independently affect risk for prostate cancer. Nat Genet.
2007;39(5):638-644.
20. Stevens VL, Ahn J, Sun J, et al. HNF1B and JAZF1 genes, diabetes,
and prostate cancer risk. Prostate. 2010;70(6):601-607.
21. Shen MM, Abate-Shen C. Molecular genetics of prostate cancer:
new prospects for old challenges. Genes Dev. 2010;24(18):1967-2000.
22. Stegeman S, Amankwah E, Klein K, et al. A large-scale analysis
of genetic variants within putative miRNA binding sites in prostate cancer.
Cancer Discov. 2015;5(4):368-379.
23. Chornokur G, Amankwah EK, Davis SN, et al. Variation in HNF1B
and obesity may influence prostate cancer risk in African American men: a
pilot study. Prostate Cancer. 2013;2013:384594.
24. Eeles RA, Kote-Jarai Z, Giles GG, et al. Multiple newly identified loci
associated with prostate cancer susceptibility. Nat Genet. 2008;40(3):316-321.
October 2016, Vol. 23, No. 4
25. Gudmundsson J, Sulem P, Steinthorsdottir V, et al. Two variants on
chromosome 17 confer prostate cancer risk, and the one in TCF2 protects
against type 2 diabetes. Nat Genet. 2007;39(8):977-983.
26. The Human Protein Atlas website. http://www.proteinatlas.org/
ENSG00000275410-HNF1B/tissue. Accessed October 10, 2016.
27. Bingham C, Hattersley AT. Renal cysts and diabetes syndrome
resulting from mutations in hepatocyte nuclear factor-1𝛽. Nephrol Dial
Transplant. 2004;19:2703-2708.
28. Haldorsen IS, Vesterhus M, Raeder H, et al. Lack of pancreatic
body and tail in HNF1B mutation carriers. Diabet Med. 2008;25(7):782-787.
29. Nakayama, K, Nozu, Y. Goto Y, et al. HNF1B alterations associated
with congenital anomalies of the kidney and urinary tract. Pediatr Nephrol.
201025(6):1073-1079.
30. Cornelis MC, Agrawal A, Cole JW, et al. The Gene, Environment
Association Studies consortium (GENEVA): maximizing the knowledge
obtained from GWAS by collaboration across studies of multiple conditions.
Genet Epidemiol. 2010;34(4):364-372.
31. Han KS, Jeong IG, Joung JY, et al. Prevalence of high-grade prostatic intraepithelial neoplasia in prostate gland of Korean men: comparisons between radical prostatectomy and cystoprostatectomy. Urology.
2007;70(6):1100-1103.
32. Paliwal S, Kovi RC, Nath B, et al. The alternative reading frame
tumor suppressor antagonizes hypoxia-induced cancer cell migration via
interaction with the COOH-terminal binding protein corepressor. Cancer
Res. 2007;67(19):9322-9329.
33. Thomas G, Jacobs KB, Yeager M, et al. Multiple loci identified
in a genome-wide association study of prostate cancer. Nat Genet.
2008;40(3):310-315
34. Lindquist KJ, Paris PL, Hoffmann TJ, et al. Mutational landscape
of aggressive prostate tumors in African American men. Cancer Res.
2016;76(7):1860-1868.
35. Yatsula B, Galvao C, McCrann M, et al. Assessment of F-MuLVinduced tumorigenesis reveals new candidate tumor genes including
Pecam1, St7, and Prim2. Leukemia. 2006;20(1):162-165.
36. Berger MF, Lawrence MS, Demichelis F, et al. The genomic complexity of priary human prostate cancer. Nature. 2011;470:214-220.
37. Baca SC, Prandi D, Lawrence MS, et al. Punctuated evolution of
prostate cancer genomes. Cell. 2013;153(3):666-677.
38. Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature. 2013;500(7463):415-421.
39. Boutros PC, Fraser M, Harding NJ, et al. Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nat Genet.
2015;47(7):736-745.
40. Cooper CS, Eeles R, Wedge DC, et al. Analysis of the genetic
phylogeny of multifocal prostate cancer identifies multiple independent
clonal expansions in neoplastic and morphologically normal prostate
tissue. Nat Genet. 2015;47(4):367-372.
41. Clark J, Merson S, Jhavar S, et al. Diversity of TMPRSS2-ERG
fusion transcripts in the human prostate. Oncogene. 2007;26(18):2667-2673.
42. Kumar-Sinha C, Tomlins SA, Chinnaiyan AM. Recurrent gene fusions
in prostate cancer. Nat Rev Cancer. 2008;8(7):497-511.
43. Mosquera JM, Mehra R, Regan MM, et al. Prevalence of TMPRSS2ERG fusion prostate cancer among men undergoing prostate biopsy in the
United States. Clin Cancer Res. 2009; 15(14):4706-4711.
44. Tiguert R, Gheiler EL, Tefilli MV, et al. Racial differences and prognostic significance of tumor location in radical prostatectomy specimens.
Prostate. 1998;37(4):230-235.
45. Tiguert R, Gheiler EL, Tefilli MV, et al. Racial differences and prognostic significance of tumor location in radical prostatectomy specimens.
Prostate. 1998 37(4):230-235.
46. Feibus AH, Levy J, McCaslin IR, et al. Racial variation in prostate
needle biopsy templates directed anterior to the peripheral zone. Urol Oncol.
2016;34(8):336.e1-6.
47. Faisal FA, Sundi D, Tosoian JJ, et al. Racial variations in prostate
cancer molecular subtypes and androgen receptor signaling reflect anatomic
tumor location. Eur Urol. 2016;70(1):14-17.
48. Epstein JI, Herawi M. Prostate needle biopsies containing prostatic
intraepithelial neoplasia or atypical foci suspicious for carcinoma: implications for patient care. J Urol. 2006;175(3 pt 1):820-834.
49. Kelloff GJ, Lieberman R, Steele VE, et al. Chemoprevention of
prostate cancer: concepts and strategies. Eur Urol. 1999;35(5-6):342-350.
50. Lieberman R. Prostate cancer chemoprevention: strategies for designing efficient clinical trials. Urology. 2001;57(4 suppl 1):224-229.
51. Fowler JE, Jr, Bigler SA, Lynch C, et al. Prospective study of correlations between biopsy-detected high grade prostatic intraepithelial
neoplasia, serum prostate specific antigen concentration, and race. Cancer.
2001;91(7):1291-1296.
52. Powell IJ, Bock CH, Ruterbusch JJ, et al. Evidence supports a faster
growth rate and/or earlier transformation to clinically significant prostate
cancer in black than in white American men, and influences racial progression
and mortality disparity. J Urol. 2010;183(5):1792-1796.
53. Potts JM, Lutz M, Walker E, et al. Trends in PSA, age and prostate
cancer detection among black and white men from 1990-2006 at a tertiary
care center. Cancer. 2010;116(16):3910-3915.
Cancer Control 421
54. Tsivian M, Banez LL, Keto CJ, et al. African-American men with
low-grade prostate cancer have higher tumor burdens: results from the
Duke Prostate Center. Prostate Cancer Prostatic Dis. 2013;16(1):91-94.
55. Chornokur G, Han G, Tanner R, et al. High grade prostate intraepithelial neoplasia (PIN) is a PSA-independent risk factor for prostate
cancer in African American men: results from a pilot study. Cancer Lett.
2013;331(2):154-157.
56. Tan PH, Tan HW, Tan Y, et al. Is high-grade prostatic intraepithelial
neoplasia on needle biopsy different in an Asian population: a clinicopathologic study performed in Singapore. Urology. 2006;68(4):800-803.
57. Martin BJ, Finlay JA, Sterling K, et al. Early detection of prostate
cancer in African-American men through use of multiple biomarkers:
human kallikrein 2 (hK2), prostate-specific antigen (PSA), and free PSA
(fPSA). Prostate Cancer Prostatic Dis. 2004;7(2):132-137.
58. Lilja H, Ulmert D, Vickers AJ. Prostate-specific antigen and
prostate cancer: prediction, detection and monitoring. Nat Rev Cancer.
2008;8(4):268-278.
59. Gee J, Bailey H, Kim K, et al. Phase II open label, multi-center clinical trial
of modulation of intermediate endpoint biomarkers by 1alpha-hydroxyvitamin D2
in patients with clinically localized prostate cancer and high grade pin. Prostate.
2013;73(9):970-978.
60. Kryvenko ON, Balise R, Soodana Prakash N, et al. African-American
men with Gleason score 3+3=6 prostate cancer produce less prostate specific
antigen than Caucasian men: a potential impact on active surveillance. J Urol.
2016;195(2):301-306.
61. Peskoe SB, Joshu CE, Rohrmann S, et al. Circulating total testosterone
and PSA concentrations in a nationally representative sample of men without
a diagnosis of prostate cancer. Prostate. 2015;75(11):1167-1176.
62. Herawi M, Kahane H, Cavallo C, et al. Risk of prostate cancer on
first re-biopsy within 1 year following a diagnosis of high grade prostatic
intraepithelial neoplasia is related to the number of cores sampled. J Urol.
2006;175(1):121-124.
63. Taneja SS, Morton R, Barnette G, et al. Prostate cancer diagnosis
among men with isolated high-grade intraepithelial neoplasia enrolled onto
a 3-year prospective phase III clinical trial of oral toremifene. J Clin Oncol.
2013;31(5):523-529.
64. Kumar NB, Pow-Sang J, Egan KM, et al. Randomized, placebocontrolled trial of green tea catechins for prostate cancer prevention. Cancer
Prev Res (Phila). 2015;8(10):879-887.
65. Amin MM, Jeyaganth S, Fahmy N, et al. Subsequent prostate cancer
detection in patients with prostatic intraepithelial neoplasia or atypical
small acinar proliferation. Can Urol Assoc J. 2007;1(3):245-249.
66. Bryant RJ, Lilja H. Emerging PSA-based tests to improve screening.
Urol Clin North Am. 2014;41(2):267-276.
67. D’Amico AV, Whittington R, Malkowicz SB, et al. Biochemical outcome after radical prostatectomy, external beam radiation therapy, or
interstitial radiation therapy for clinically localized prostate cancer. JAMA.
1998;280(11):969-974.
68. Shuch B, Mikhail M, Satagopan J, et al. Racial disparity of epidermal growth factor receptor expression in prostate cancer. J Clin Oncol.
2004;22(23):4725-4729.
69. Gaston KE, Kim D, Singh S, et al. Racial differences in androgen
receptor protein expression in men with clinically localized prostate cancer.
J Urol. 2003;170(3):990-993.
70. deVere White RW, Deitch AD, Jackson AG, et al. Racial differences in clinically localized prostate cancers of black and white men. J Urol.
1998;159(6):1979-1982.
71. Guo Y, Sigman DB, Borkowski A, et al. Racial differences in prostate
cancer growth: apoptosis and cell proliferation in Caucasian and AfricanAmerican patients. Prostate. 2000;42(2):130-136.
72. Kim HS, Moreira DM, Jayachandran J, et al. Prostate biopsies
from black men express higher levels of aggressive disease biomarkers
than prostate biopsies from white men. Prostate Cancer Prostatic Dis.
2011;14(3):262-265.
73. Wallace TA, Prueitt RL, Yi M, et al. Tumor immunobiological differences in prostate cancer between African-American and European-American
men. Cancer Res. 2008;68(3):927-936.
74. Yang G, Addai J, Ittmann M, et al. Elevated caveolin-1 levels in
African-American versus white-American prostate cancer. Clin Cancer
Res. 2000;6(9):3430-3433.
75. Reams RR, Agrawal D, Davis MB, et al. Microarray comparison
of prostate tumor gene expression in African-American and Caucasian
American males: a pilot project study. Infect Agent Cancer. 2009;4(suppl
1):S3.
76. Chien J, Staub J, Avula R, et al. Epigenetic silencing of TCEAL7
(Bex4) in ovarian cancer. Oncogene. 2005;24(32):5089-5100.
77. Kelloff GJ, Lippman SM, Dannenberg AJ, et al. Progress in chemoprevention drug development: the promise of molecular biomarkers for
prevention of intraepithelial neoplasia and cancer--a plan to move forward.
Clin Cancer Res. 2006;12(12):3661-3697.
78. Andriole GL, Bostwick D, Brawley OW, et al. The effect of dutasteride on the usefulness of prostate specific antigen for the diagnosis of high
grade and clinically relevant prostate cancer in men with a previous negative biopsy: results from the REDUCE study. J Urol. 2011;185(1):126-131.
422 Cancer Control
79. Hamilton RJ, Kahwati LC, Kinsinger LS. Knowledge and use of finasteride for the prevention of prostate cancer. Cancer Epidemiol Biomarkers
Prev. 2010;19(9):2164-2171.
80.Thompson IM, Goodman PJ, Tangen CM, et al. The influence
of finasteride on the development of prostate cancer. N Engl J Med.
2003;349(3):215-224.
81. Aggarwal BB, Prasad S, Reuter S, et al. Identification of novel antiinflammatory agents from Ayurvedic medicine for prevention of chronic
diseases: “reverse pharmacology” and “bedside to bench” approach. Curr
Drug Targets. 2011;12(11):1595-1653.
82. Amin AR, Kucuk O, Khuri FR, et al. Perspectives for cancer prevention
with natural compounds. J Clin Oncol. 2009;27(16):2712-2725.
83. Aravindaram K, Yang NS. Anti-inflammatory plant natural products
for cancer therapy. Planta Med. 2010;76(11):1103-1117.
84. Gogtay NJ, Bhatt HA, Dalvi SS, et al. The use and safety of nonallopathic Indian medicines. Drug Saf. 2002;25(14):1005-1019.
85. Jia L. Cancer complementary and alternative medicine research at
the US National Cancer Institute. Chin J Integr Med. 2012;18(5):325-332.
86. Kannappan R, Gupta SC, Kim JH, et al. Tocotrienols fight cancer by
targeting multiple cell signaling pathways. Genes Nutr. 2012;7(1):43-52.
87. Reddy BS. Chemoprevention of colon cancer by minor dietary constituents and their synthetic analogues. Prev Med. 1996;25(1):48-50.
88. Wargovich MJ, Jimenez A, McKee K, et al. Efficacy of potential chemopreventive agents on rat colon aberrant crypt formation and progression.
Carcinogenesis. 2000;21(6):1149-1155.
89. Minich DM, Bland JS. A review of the clinical efficacy and safety of
cruciferous vegetable phytochemicals. Nutr Rev. 2007;65(6 pt 1):259-267.
90. Scott EN, Gescher AJ, Steward WP, et al. Development of dietary
phytochemical chemopreventive agents: biomarkers and choice of dose
for early clinical trials. Cancer Prev Res (Phila). 2009;2(6):525-530.
91. Lippman SM, Klein EA, Goodman PJ, et al. Effect of selenium and
vitamin E on risk of prostate cancer and other cancers: the Selenium and
Vitamin E Cancer Prevention Trial (SELECT). JAMA. 2009;301(1):39-51.
92. Kumar N, Crocker T, Smith T, et al. Prostate cancer chemoprevention
targeting men with high-grade prostatic intraepithelial neoplasia (HGPIN)
and atypical small acinar proliferation (ASAP): model for trial design and
outcome measures. J Clin Trials. 2012;2(1).
93. Kumar N, Crocker T, Smith T, et al. Prostate cancer chemoprevention
targeting high risk populations: model for trial design and outcome measures.
J Cancer Sci Ther. 2012;2011(S3).
94. The effect of vitamin E and beta carotene on the incidence of lung cancer
and other cancers in male smokers. The Alpha-Tocopherol, Beta Carotene
Cancer Prevention Study Group. N Engl J Med. 1994;330(15):1029-1035.
95. Omenn GS, Goodman GE, Thornquist MD, et al. Risk factors for
lung cancer and for intervention effects in CARET, the Beta-Carotene and
Retinol Efficacy Trial. J Natl Cancer Inst. 1996;88(21):1550-1559.
96. Kakarala M, Brenner DE, Korkaya H, et al. Targeting breast stem
cells with the cancer preventive compounds curcumin and piperine. Breast
Cancer Res Treat. 2010;122(3):777-785.
97. Parnes HL, Brawley OW, Minasian LM, et al. Phase III prostate cancer chemoprevention trials. Recent Results Cancer Res. 2014;202:73-77.
98. Bettuzzi S, Brausi M, Rizzi F, et al. Chemoprevention of human prostate cancer by oral administration of green tea catechins in volunteers with
high-grade prostate intraepithelial neoplasia: a preliminary report from a
one-year proof-of-principle study. Cancer Res. 2006;66(2):1234-1240.
99. Harper CE, Patel BB, Wang J, et al. Epigallocatechin-3-Gallate
suppresses early stage, but not late stage prostate cancer in TRAMP mice:
mechanisms of action. Prostate. 2007;67(14):1576-1589.
100. McLarty J, Bigelow RL, Smith M, et al. Tea polyphenols decrease
serum levels of prostate-specific antigen, hepatocyte growth factor, and
vascular endothelial growth factor in prostate cancer patients and inhibit
production of hepatocyte growth factor and vascular endothelial growth
factor in vitro. Cancer Prev Res (Phila). 2009;2(7):673-682.
101. Kumar NB, Pow-Sang J, Egan KM, et al. Effect of polyphenon E
on progression to prostate cancer after diagnosis of high grade prostatic
intraepithelial neoplasia or atypical small acinar proliferation in African
American men. Presented at: American Association for Cancer Research;
November 9–12, 2014; San Antonio, Texas.
102. Albrecht DS, Clubbs EA, Ferruzzi M, et al. Epigallocatechin-3-gallate
(EGCG) inhibits PC-3 prostate cancer cell proliferation via MEK-independent
ERK1/2 activation. Chem Biol Interact. 2008;171(1):89-95.
103. Paschka AG, Butler R, Young CY. Induction of apoptosis in prostate
cancer cell lines by the green tea component, (-)-epigallocatechin-3-gallate.
Cancer Lett. 1998;130(1-2):1-7.
104. Yu HN, Yin JJ, Shen SR. Growth inhibition of prostate cancer cells
by epigallocatechin gallate in the presence of Cu2+. J Agric Food Chem.
2004;52(3):462-466.
105. McCarthy S, Caporali A, Enkemann S, et al. Green tea catechins
suppress the DNA synthesis marker MCM7 in the TRAMP model of prostate
cancer. Mol Oncol. 2007;1(2):196-204.
106. Nyska A, Suttie A, Bakshi S, et al. Slowing tumorigenic progression
in TRAMP mice and prostatic carcinoma cell lines using natural anti-oxidant
from spinach, NAO--a comparative study of three anti-oxidants. Toxicol
Pathol. 2003;31(1):39-51.
October 2016, Vol. 23, No. 4
107.Sartor L, Pezzato E, Dona M, et al. Prostate carcinoma and
green tea: (-)epigallocatechin-3-gallate inhibits inflammation-triggered
MMP-2 activation and invasion in murine TRAMP model. Int J Cancer.
2004;112(5):823-829.
108. Rahmani AH, Al Shabrmi FM, Allemailem KS, et al. Implications of
Green Tea and Its Constituents in the Prevention of Cancer via the Modulation
of Cell Signalling Pathway. Biomed Res Int. 2015;2015:925640.
109. Chuu CP, Chen RY, Kokontis JM, et al. Suppression of androgen
receptor signaling and prostate specific antigen expression by (-)-epigallocatechin-3-gallate in different progression stages of LNCaP prostate cancer
cells. Cancer Lett. 2009;275(1):86-92.
110. Kao YH, Hiipakka RA, Liao S. Modulation of endocrine systems
and food intake by green tea epigallocatechin gallate. Endocrinology.
2000;141(3):980-987.
111. Ren F, Zhang S, Mitchell SH, et al. Tea polyphenols down-regulate
the expression of the androgen receptor in LNCaP prostate cancer cells.
Oncogene. 2000;19(15):1924-1932.
112. Hastak K, Gupta S, Ahmad N, et al. Role of p53 and NF-kappaB in
epigallocatechin-3-gallate-induced apoptosis of LNCaP cells. Oncogene.
2003;22(31):4851-4859.
113. Siddiqui IA, Adhami VM, Afaq F, et al. Modulation of phosphatidylinositol-3-kinase/protein kinase B- and mitogen-activated protein kinasepathways by tea polyphenols in human prostate cancer cells. J Cell
Biochem. 2004;91(2):232-242.
114. Siddiqui IA, Malik A, Adhami VM, et al. Green tea polyphenol EGCG
sensitizes human prostate carcinoma LNCaP cells to TRAIL-mediated
apoptosis and synergistically inhibits biomarkers associated with angiogenesis and metastasis. Oncogene. 2008;27(14):2055-2063.
115. Kleiner DE, Stetler-Stevenson WG. Matrix metalloproteinases and
metastasis. Cancer Chemother Pharmacol. 1999;43 Suppl:S42-51.
116. Nandakumar V, Vaid M, Katiyar SK. (-)-Epigallocatechin-3-gallate
reactivates silenced tumor suppressor genes, Cip1/p21 and p16INK4a, by
reducing DNA methylation and increasing histones acetylation in human
skin cancer cells. Carcinogenesis. 2011;32(4):537-544.
117. Adhami VM, Ahmad N, Mukhtar H. Molecular targets for green tea
in prostate cancer prevention. J Nutr. 2003;133(7 suppl):2417S-2424S.
118.Connors SK, Chornokur G, Kumar NB. New insights into the
mechanisms of green tea catechins in the chemoprevention of prostate
cancer. Nutr Cancer. 2012;64(1):4-22.
119. Kazi A, Daniel KG, Smith DM, et al. Inhibition of the proteasome
activity, a novel mechanism associated with the tumor cell apoptosisinducing ability of genistein. Biochem Pharmacol. 2003;66(6):965-976.
120. Nam S, Smith DM, Dou QP. Ester bond-containing tea polyphenols
potently inhibit proteasome activity in vitro and in vivo. J Biol Chem.
2001;276(16):13322-13330.
121. Smith DM, Wang Z, Kazi A, et al. Synthetic analogs of green tea
polyphenols as proteasome inhibitors. Mol Med. 2002;8(7):382-392.
122.Adhami VM, Siddiqui IA, Sarfaraz S, et al. Effective prostate
cancer chemopreventive intervention with green tea polyphenols in the
TRAMP model depends on the stage of the disease. Clin Cancer Res.
2009;15(6):1947-1953.
123. Caporali A, Davalli P, Astancolle S, et al. The chemopreventive
action of catechins in the TRAMP mouse model of prostate carcinogenesis is accompanied by clusterin over-expression. Carcinogenesis.
2004;25(11):2217-2224.
124. Gupta S, Hastak K, Ahmad N, et al. Inhibition of prostate carcinogenesis in TRAMP mice by oral infusion of green tea polyphenols. Proc
Natl Acad Sci U S A. 2001;98(18):10350-10355.
125. Khan N, Adhami VM, Mukhtar H. Review: green tea polyphenols in
chemoprevention of prostate cancer: preclinical and clinical studies. Nutr
Cancer. 2009;61(6):836-841.
126. Kim SJ, Amankwah E, Connors S, et al. Safety and chemopreventive
effect of Polyphenon E in preventing early and metastatic progression of
prostate cancer in TRAMP mice. Cancer Prev Res (Phila). 2014;7(4):435-444.
127. Siddiqui IA, Zaman N, Aziz MH, et al. Inhibition of CWR22Rnu1
tumor growth and PSA secretion in athymic nude mice by green and black
teas. Carcinogenesis. 2006;27(4):833-839.
128. Suttie A, Nyska A, Haseman JK, et al. A grading scheme for the
assessment of proliferative lesions of the mouse prostate in the TRAMP
model. Toxicol Pathol. 2003;31(1):31-38.
129. Yang CS, Lambert JD, Hou Z, et al. Molecular targets for the cancer
preventive activity of tea polyphenols. Mol Carcinog. 2006;45(6):431-435.
130. Brausi M, Rizzi F, Bettuzzi S. Chemoprevention of human prostate
cancer by green tea catechins: two years later. A follow-up update. Eur
Urol. 2008;54(2):472-473.
131. Chow HH, Cai Y, Alberts DS, et al. Phase I pharmacokinetic study of
tea polyphenols following single-dose administration of epigallocatechin gallate and polyphenon E. Cancer Epidemiol Biomarkers Prev. 2001;10(1):53-58.
132. Chow HH, Cai Y, Hakim IA, et al. Pharmacokinetics and safety of
green tea polyphenols after multiple-dose administration of epigallocatechin gallate and polyphenon E in healthy individuals. Clin Cancer Res.
2003;9(9):3312-3319.
133. Chow HH, Hakim IA, Vining DR, et al. Effects of dosing condition
on the oral bioavailability of green tea catechins after single-dose
October 2016, Vol. 23, No. 4
administration of polyphenon E in healthy individuals. Clin Cancer Res.
2005;11(12):4627-4633.
134. Jatoi A, Ellison N, Burch PA, et al. A phase II trial of green tea in
the treatment of patients with androgen independent metastatic prostate
carcinoma. Cancer. 2003;97(6):1442-1446.
135. Nguyen MM, Ahmann FR, Nagle RB, et al. Randomized, doubleblind, placebo-controlled trial of polyphenon E in prostate cancer patients
before prostatectomy: evaluation of potential chemopreventive activities.
Cancer Prev Res (Phila). 2012;5(2):290-298.
136. Pisters KM, Newman RA, Coldman B, et al. Phase I trial of oral green tea
extract in adult patients with solid tumors. J Clin Oncol. 2001;19(6):1830-1838.
137. Kumar NB, Pow-Sang J, Spiess PE et al. Effect of polyphenol E
on progression to prostate cancer after diagnosis of high grade prostatic
intraepithelial neoplasia or atypical small acinar proliferation in African
American men: preliminary finds of a phase II randomized, double-blind,
placebo-controlled clinical trial. Presented at: 7th Annual American Association of Cancer Research Science of Cancer Health Disparities in Racial/
Ethnic Minorities and the Medically Underserved; November 9, 2015; San
Antonio, TX.
138. Eskew LA, Bare RL, McCullough DL. Systematic 5 region prostate
biopsy is superior to sextant method for diagnosing carcinoma of the prostate.
J Urol. 1997;157(1):199-202.
Cancer Control 423
Challenges remain in the field of cancer
care for young adults and adolescents.
Yellow Tulips. X-Ray image courtesy of Bryan Whitney. www.x-rayphotography.com.
Disparities in Adolescents and Young Adults With Cancer
Leidy L. Isenalumhe, MD, Olivia Fridgen, MPH, Lynda K. Beaupin, MD, Gwendolyn P. Quinn, PhD,
and Damon R. Reed, MD
Background: Cancer care for adolescents and young adults (AYAs) focuses on the care of patients aged
15 to 39 years. Historically, this group has favorable outcomes based on a preponderance of diagnoses such as
thyroid cancers and Hodgkin lymphoma. Improvements in outcomes among the AYA population have lagged
behind compared with younger and older populations.
Methods: We discuss and review recent progress in AYA patient care and highlight remaining disparities that
exist, including financial disadvantages, need for fertility care, limited clinical trial availability, and other
areas of evolving AYA-focused research.
Results: Survival rates have not improved for this age group as they have for children and older adults. Disparities are present in the AYA population and have contributed to this lack of progress.
Conclusions: Recognizing disparities in the care of AYAs with cancer has led many medical specialty disciplines to improve the lives of these patients through advocacy, education, and resource development. Research
addressing barriers to clinical trial enrollment in this population, quality-of-life issues, and the improvement
of survivorship care is also under way.
Introduction
In 2008, an article published in Cancer Control about
adolescents and young adults (AYAs) addressed this
emerging, complicated, and multifaceted field.1 The
authors concluded that more data were needed on
treatment and survivorship in this patient populaFrom the Adolescent and Young Adult Oncology (LLI, OF, GPQ, DRR)
and Health Outcomes and Behavior Programs (GPQ, DRR), Sarcoma Department (DRR), H. Lee Moffitt Cancer Center & Research
Institute, Tampa, Florida, Department of Pediatric Oncology (LKB),
Roswell Park Cancer Institute, Buffalo, New York, and Morsani College of Medicine (GPQ), University of South Florida, Tampa, Florida.
Submitted February 9, 2016; accepted June 2, 2016.
Address correspondence to Damon R. Reed, MD, Sarcoma Department,
Moffitt Cancer Center, 12902 Magnolia Drive, FOB-1, Tampa, FL 33612.
E-mail: [email protected]
No significant relationships exist between the authors and the companies/organizations whose products or services may be referenced
in this article.
424 Cancer Control
tion to improve outcomes and opined that major academic centers should develop the framework for a
specialized oncology discipline focusing on AYAs.1
Indeed, published research on AYAs has addressed
aspects of their care but has not yet successfully
improved upon the entirety of oncology specific to
AYAs. Programs devoted to AYAs have developed
and specialized their focus to address fertility or psychosocial aspects of care, but no single model has yet
to be universally adopted or standardized as the gold
standard for the care of AYAs with cancer. Difficulties exist when discussing or acknowledging aspects
of AYA care, such as reproductive health and sexuality, which was the theme of the annual conference
of the Critical Mass Young Adult Alliance in 2015.2
These topics were presented and discussed within
the context of multidisciplinary teams.
Disparities are defined by the National Cancer
October 2016, Vol. 23, No. 4
Institute as adverse differences in incidence, outcome, or burden of cancer existing among a population group.3 Although chronological age demarcates
oncology in AYAs, it is this emerging developmental
period of adulthood that brings in other aspects of
disparities. In the context of oncology, AYAs are defined as individuals diagnosed with cancer between
15 and 39 years of age.4 Cancer is the primary cause of
disease-related death in this population.5 In 2011, an
estimated 69,212 AYAs were diagnosed with cancer,
equating to 6 times more than children younger than
14 years of age.6 According to a cancer statistics review
of Surveillance, Epidemiology, and End Results program data from 1975 to 1997, improvement in 5-year
survival rates, as reflected in the average annual percent change in US cancer mortality rates, was significantly lower among individuals 15 to 39 years of age
compared with those aged 1 to 14 years.4 AYAs have
inferior outcomes compared with children and older
adults with the same diagnosis.7-11
Among younger AYAs aged 15 to 24 years, leukemia, lymphoma, central nervous system tumors, sarcomas, and germ cell tumors are the most common
malignancies.6 In AYAs 25 to 39 years of age, leukemia and lymphoma are still common, but the incidence rates of breast cancer, melanoma, and colorectal cancer (CRC) are increased (Fig).6
AYAs may experience economic disparities as
they enter the workforce, often in jobs without benefits.4 In addition, disparities in health education may
exist, because AYAs are inexperienced in navigating
the health system or inquiring about clinical trials.4
They also may experience delays in diagnosis, because malignancies are low on the differential diagnosis for ill patients who are young.12
Presentation, Diagnosis, and Access
to Health Care
Number of Observed Cases
Number of Observed Cases
Health Insurance and Financial Hardship
Cancer diagnosis is typically delayed in AYAs compared with children. The cause is multifactorial and
relates to low suspicion of a malignant diagnosis by
both patient and caretaker, but also is related to inadequate insurance coverage.4 In the United States, those
between the ages of 18 and 24 years and between 25
and 34 years are more likely than any other age group
to be uninsured.13,14 Lack of health insurance in AYAs
is associated with delays in diagnosis, an increased
likelihood of metastatic disease at presentation, and
a decreased likelihood of receiving definitive therapy
compared with those who are insured.15-18 In a study
of more than 39,000 participants aged 20 to 40 years,
health insurance coverage was associated with a decreased likelihood of metastatic disease at presentation; those who were uninsured were more likely to
be male, nonwhite, and unmarried.15 After adjusting
for various factors, health insurance coverage was
also significantly associated with lower estimates of
all-cause mortality.15 In a study looking at the association between health insurance coverage, socioeconomic status, and the risk
of advanced Hodgkin
lymphoma in AYAs, pub40,000
12,000
lic insurance coverage for
35,000
10,000
low-income or individuals
30,000
with medical need was as8,000
25,000
sociated with an increased
risk of advanced disease
20,000
6,000
on presentation compared
15,000
4,000
with patients with private
10,000
insurance.16
2,000
5,000
Financial hardships
are
not uncommon in
0
0
Age
Age
Age
Age
Age
patients
with cancer but
15–19 y 20–24 y
25–29 y 30–34 y 35–39 y
they may be more burBreast
Thyroid
Leukemia and lymphoma
Germ cell
densome among AYAs.19
Melanoma of the skin
Leukemia and lymphoma
Thyroid
CNS
Some AYAs may have inColon and rectum
CNS
Melanoma of the skin
Malignant bone tumors
Cervix
and
uterus
Soft
tissue
and
Kaposi
sarcoma
troductory level jobs with
Other
Soft tissue and Kaposi sarcoma
Germ cell
Other
limited time off and without the ability to take exIncludes testicular cancer.
tended leave, have limited
Includes breast, cervical, colon, and other less-prevalent cancers.
savings, and may have
Includes malignant bone tumors and other less-prevalent cancers.
college debt.19
Fig. — Common types of cancer affecting adolescents and young adults. CNS = central nervous system.
Two-thirds of patients
Reproduced with permission from the National Cancer Institute; Surveillance, Epidemiology, and End Results.
aged
21 to 40 years reportA snapshot of adolescent and young adult cancer. https://www.cancer.gov/research/progress/snapshots
/adolescent-young-adult. Accessed October 5, 2016.
ed that their cancer exa
b
a
c
a
b
c
October 2016, Vol. 23, No. 4
Cancer Control 425
perience negatively affected their financial situation.19
Financial hardship in relation to the costs of cancer
treatment can cause stress, may result in reduced quality of life, and may lead to patients terminating their
therapeutic plan.19
The Affordable Care Act has enabled more than
2.5 million young adults aged 19 to 25 years to gain access to health insurance, although the uninsured rate is
still highest among those aged 15 to 39 years.14,17 After
implementation of the Affordable Care Act Dependent
Coverage Expansion, detection rates increased for early-stage cervical cancer among women aged 21 to 25
years secondary to increases in access to health care.20
Diagnosing cervical cancer in its early stage affords
the oncologist the possibility of using fertility-sparing treatments; thus, the proportion of young women
with cancer able to receive fertility-sparing treatment
increased.20 Further analyses are needed to determine
whether expanded coverage for health insurance is associated with other favorable changes in AYAs.
Treatment
The optimal location of care is a complex topic. Expertise for a given cancer diagnosis may lie within either
pediatric or medical oncologists. Pathways could provide an optimal range of therapeutic options, identify areas for future clinical research for AYAs, and create disease-specific champions for AYAs passionate about the
care of this patient population. Although the treatment
protocol may be similar for certain cancers, others may
vary with respect to outcomes, cost of care, or intensity
of care.21 It is our opinion that a larger pool of expertise
may exist at an adult facility for melanoma or neuroendocrine diseases, but patients with sarcoma most likely
receive similar care at both the adult and pediatric cancer centers due to shared staffing. Care for a patient with
leukemia must involve increasing levels of collaboration
to provide personalized care using an evidence-based
approach to treatment.22
Patients treated in a National Cancer Institute–designated comprehensive cancer center have superior
outcomes compared with those treated at facilities
not designated as such, a fact likely due to the preponderance of trials conducted at these centers.18
Barriers impeding access to National Cancer Institute–designated comprehensive cancer centers include lack of health care insurance, public insurance,
lower socioeconomic status, and African American or
Hispanic race/ethnicity.22 Approximately 28% of patients older than 15 years of age are treated at community centers, not tertiary care centers.23 AYAs are the
least likely of all age groups to be treated at comprehensive cancer centers and, therefore, they may lack
opportunities for multidisciplinary care, tumor board
discussions, subspecialty care (eg, rare tumors), and
the opportunity to enroll in clinical trials.22,24
426 Cancer Control
Clinical Trial Enrollment
The percentage of AYAs enrolled in clinical trials is
lower than that seen in younger or older patients.21,25-29
AYAs who are uninsured, older in age, or treated by
nonpediatric oncologists are less likely than older patients to be enrolled in a clinical trial.25 Even within
institutions that have the same clinical trials, enrollment of AYAs is lower than the enrollment of the pediatric population.28,29 Many obstacles contribute to the
low number of AYAs enrolled in clinical trials, one of
which is lack of knowledge about clinical trials among
AYAs. In 1 study, 62% of AYAs did not know if clinical trials were available for their type of cancer.30 Treatment location is also a factor. A total of 40% of AYAs
are treated at academic centers.31 Even when treated
at a cancer center, not all clinical trials are available.
Some hospitals are not allowed to enroll patients in
Children Oncology Group trials if they are not participating institutions.32
In 2012, the Centers for Disease Control and Prevention convened a group of oncologists who were
experts in the AYA population to examine barriers to
clinical trial enrollment among adolescents. Five issues
were identified as the most important barriers33:
1. Low referral rates of adolescents with cancer to
pediatric cancer centers
2. Limited availability of clinical trials for certain
cancers
3. Physician-related barriers limiting clinical trial
accrual
4. Institutional barriers impeding collaboration
between pediatric and nonpediatric oncologists
on clinical trials
5. Unique psychosocial needs of adolescents with
cancer
Progress has been made by creating AYA programs at select cancer centers and by increasing the
development and availability of clinical trials for
AYA patients.34,35 Although the number of programs
for AYAs with cancer is increasing in the United
States, no formal accreditation, registry, or minimal
standard exists by which to measure these targeted
programs; thus, the structures available and the services offered vary among programs.36 In an attempt
to standardize treatment and services for AYAs with
cancer, various organizations have developed treatment guidelines and recommendations for this patient population (Table 1).4,35-40 In 2016, the National
Comprehensive Cancer Network published updated
treatment guidelines for AYAs with cancer.37 Its recommendations emphasize the need for a comprehensive, multidisciplinary approach to AYA care, including supportive and treatment guidelines that aim to
improve treatment tolerance, compliance, and clinical outcomes.37
Although dedicated efforts to design programs
October 2016, Vol. 23, No. 4
Table 1. — Select Resources for the Care of AYAs
With Cancer
Reference
Comment
Adolescent and
Young Adult
Oncology Progress
Review Group 4
Establish age demarcation of AYA population
Summarize epidemiology
Dictate disparities and unique issues that
affect AYAs
Ferrari35
AYAs require patient-focused multidisciplinary
approach
A need exists for oncologists trained in
AYA oncology
Importance of establishing a physical space
in the institution for AYAs
Coccia39
National Comprehensive Cancer
Network 37
Identify issues specific to AYAs
Explain special considerations related to
cancer management in AYAs
Provide supportive and treatment guidelines
that aim to improve treatment tolerance,
compliance, and clinical outcomes
Discuss long-term consequences
Promote participation in clinical trials
Nass 36
Summarize gaps in cancer care confronted
by AYAs
Describe potential strategies and actions to
improve the quality of care for AYAs
Focus on quality-of-life issues and late effects
Loren40
Discuss fertility preservation with all
patients of reproductive ages with cancer and
discuss with parents/guardians of children
and adolescents
A need exists for proper documentation
of fertility discussion and options provided
to patients
Embryo and oocyte cryopreservation now
standard practice
AYA = adolescent and young adult.
aimed at increasing clinical trial enrollment in AYAs
have shown some promise, improvements are more
likely with sustained, systematic approaches to clinical trial research in the AYA population.41 Efforts to
improve trial enrollment are ongoing, some of which
use multidisciplinary, collaborative oncology and pediatric specialist teams to study diseases such as leukemia and sarcoma.38 Clinical trial registries, advocacy
groups, and efforts to increase collaborations and communication across adult and pediatric cancer centers
are likely to continue progress in increasing clinical
trial enrollment rates in AYAs (Table 2).
Psychosocial Needs and Late Effects
The wide age range of AYA care includes patients still
under the guardianship of their parents to those in
committed relationships and married; thus, the psychosocial needs among this age group widely vary.
Quality of life, adjusting to a cancer diagnosis, searching for meaning or understanding after a diagnosis,
October 2016, Vol. 23, No. 4
and vocational outcomes can be challenging. AYAs
may require psychosocial care throughout the cancer care continuum because, if left unaddressed, such
psychosocial issues may lead to poorer outcomes
than patients with access to age-appropriate resources. During young adulthood, autonomy and self-reliance are prized and contribute to a person’s sense
of accomplishment and maturity.42 AYAs with cancer
may have experienced these shortly before medical
and financial hardships forced them into the care of
their parents, away from the working world or from
pursuing their education and possibly isolating them
from their peers without cancer.36 This isolation can
be reinforced when AYAs see their friends achieving
age-specific milestones, such as graduation, career
success, marriage, and starting a family, because they
may be unable to achieve or experience these same
life events while undergoing treatment or recovering
in survivorship stages.43
Because of these experiences, the coordination of
cancer care while maintaining social connectivity is
paramount to the successful treatment of AYAs.42 Some
of the services AYAs require while undergoing treatment include financial counseling and insurance assistance, pain management, and mental health and peer
support (Table 3). The need for these services is not exclusive to AYAs with cancer; however, the life stage of
AYAs exacerbates these associated needs, particularly
because many AYAs report that these needs remain
unmet.44 AYAs have higher rates of distress at diagnosis than older matched populations, and they maintain
high rates of distress during survivorship.45-47 Smith
et al48 looked at the unmet needs in AYAs and found
that patients who lacked mental health services had
the poorest health-related quality-of-life scores. AYAs
have identified family support, religious beliefs, and
personal encouragement as the strongest sources of
support during a cancer diagnosis and subsequent
treatment.49 Another study reported that patients who
had adequate health care insurance, a good family support system, and were properly informed about preserving their fertility were the most happy with their
cancer care.50 In addition, AYAs who were the most
capable of coping with their diagnosis maintained involvement in their career, school, or social spheres.47
Relationships can shape the lives of AYAs and may
be a helpful source of support throughout the cancer
care continuum. Significant others or life partners may
also play roles in decision-making, potential discourse,
and act as support systems. The key caregiver or health
care proxy assisting in medical decisions may vary
with each AYA, their state of residence, and may not
remain consistent throughout the disease trajectory.
Research suggests that the most important opinions
about the medical care of AYAs may not come from
family members, as a clinical care team may assume;
Cancer Control 427
spouses, and siblings, especially if family members
make special work arrangeClinical Trial Type of Study Eligibility
Study Objective
Identifier
(Age), y
ments and sacrifices to be
more available to patients unProspective
Feasibility and safety of a pediatric regimen in patients
NCT01406756
16–39
phase 2
with ALL treated by a hematologist/oncologist
dergoing treatment.56
Standard- or high-dose combination chemotherapy
Health care professionRandomized
NCT02375204
≥ 14
and stem-cell transplant in males with relapsed or
als
have
cited end-of-life care
phase 3
refractory germ cell tumors
as the most difficult need
Combination chemotherapy in treating newly
NCT01406756
Phase 3
1–31
to meet in the AYA populadiagnosed ALL
tion.57-59 Several studies have
Randomized
Combination chemotherapy in treating nonmetastatic,
NCT01231906
≤ 50
looked
at the best time to
phase 3
extracranial Ewing sarcoma
discuss
advanced
care planRandomized
Combination chemotherapy ± ganitumab in newly
NCT02306161
≤ 50
ning,
suggesting
that
discusphase 2
diagnosed metastatic Ewing sarcoma
sions
should
begin
as
early
NCT02470091
Phase 2
11–49
Use of denosumab for recurrent/refractory osteosarcoma
as the initial diagnosis and
Use of eribulin mesylate in relapse/refractory
NCT02097238
Phase 2
12–49
as late as hospice referral.60-62
osteosarcoma
Research also suggests that
Preoperative chemoradiotherapy or preoperative
Randomized
AYAs who make their final
NCT02180867
≥2
radiotherapy ± pazopanib in nonrhabdomyosarcoma
phase 2/3
soft-tissue sarcoma
wishes known regain more
Comparing ipilimumab to high-dose interferon α-2b
control of their health care
NCT01274338
Phase 3
≥ 12
in high-risk stage 3/4 melanoma after surgery
situation than patients who
Targeted therapy directed by genetic testing in
do not express their wishNCT02465060
Phase 2
≥ 18
advanced refractory solid tumors or lymphomas
es.63,64 In addition, AYAs who
ALL = acute lymphoblastic leukemia, AYA = adolescent and young adult, Ph+ = Philadelphia chromosome positive.
had a written advanced care
planning document reported
higher rates of active coping
Table 3. — Select Resources for Adolescents and Adults With Cancer
and emotional support than
Focus
Organization
Website
those without a written inFertility preservation
Oncofertility Consortium
www.oncofertility.northwestern.edu
structions for advanced care
planning.65 One study found
Financial assistance
SAMFund
www.thesamfund.org
that advanced care planning
Legal assistance
Triage Cancer
www.triagecancer.org
reduced levels of anxiety and
Cancer Care
www.cancercare.org
National support and
familial conflict, because
resources
Critical Mass
www.criticalmass.org
such wishes were discussed
Patient advocacy and
in advance and a health care
Stupid Cancer
www.stupidcancer.org
awareness
surrogate was identified.63
Physician education
Focus Under Forty
www.university.asco.org/focus-under-forty
Caregivers of AYAs also benefitted from the conversation
Program development
Teen Cancer America
www.teencanceramerica.org
about end-of-life planning
Program standardization
Change it Back
www.HCRI.org/programs/change-it-back
as they were able, in times
of great grief, to realize and
carry out the wishes of their
instead, AYAs hold opinions of friends and peers in
loved one.63 Such events would not likely have haphigher regard.51 Doing so may increase levels of stress
pened without guidance for advanced care planning.
or isolate patients in such a way that they feel they are
Some meaningful progress in AYA care has inmaking decisions on their own.52
volved the formative and focused research to identify
Many AYAs are also unsure how or when to share
the psychosocial needs of AYAs and the development
their cancer diagnosis, impaired fertility, and medical
of programs to help address those needs; similarly,
debt, among other issues, with a prospective partner.53
AYA-focused organizations strive to provide financial,
Self-confidence and vulnerability are important aspects
vocational, and insurance assistance to cancer patients,
of any relationship formation, but many AYAs struggle
survivors, and their friends and families (see Table 3).
with negative body issues or low levels of self-esteem,
Progress was also made when the American Acadand report that their experience with cancer has negemy of Pediatrics and the Institute of Medicine estabatively affected their developing romantic relationlished recommendations to include AYAs in their own
ships.53-55 AYAs may also feel burdensome to parents,
end-of-life discussions, stating that such discussions
Table 2. — Select Clinical Trials for Adolescents and Young Adults
428 Cancer Control
October 2016, Vol. 23, No. 4
did not contradict a hopeful, positive outcome.61 The
creation of Five Wishes (Aging With Dignity, Tallahassee, FL) has allowed health care professionals to use
a more age-appropriate advance care planning tool to
respect the goals and priorities of AYAs at the end of
their life.61 This tool has also served as a starting point
for feedback from AYAs and to continue to develop
the most applicable advance care planning for young
adults with cancer and the thoughts they wish to convey to their loved ones.
Fertility and Reproductive Health
A significant psychosocial concern among AYA patients
is loss of fertility. Chemotherapy, radiotherapy, surgery,
and cancer itself can cause temporary or permanent
damage to the reproductive organs and endocrine or
pituitary functioning. The risk of permanent infertility
depends on patient age (younger patients typically fare
better than older patients), type of cancer and its treatment, and the dose and intensity of the treatment.66,67
In addition, females may experience premature ovarian
insufficiency or failure, causing infertility, premature
menopause, or both.67
In the midst of a cancer diagnosis, understanding a
patient’s risk of infertility and possible options for preserving fertility can be daunting and complex. Decisionmaking about infertility and fertility preservation can be
challenging for the AYA patient because multiple barriers exist.67-72 A patient may not have considered his or
her desire for biological children in the future, and such
decisions may become compounded by the stress of a
cancer diagnosis.68 The patient or the oncologist may
feel that a delay in treatment to pursue fertility preservation is unwise, and thus the patient may not have time
to pursue preservation.69,70 The financial costs associated with fertility preservation are often not covered by
health care insurance, thus making fertility preservation
out of reach for many AYA patients.67 Ethical, religious,
and other matters should be considered and may impede the decision-making of an AYA about fertility preservation. Discussions about fertility and sexual health
may be uncomfortable for the AYA patient, particularly
if his or her parents are present. Although the parents
and loved ones of AYAs with cancer typically want to
be helpful and support the patient as much as possible,
studies suggest that they may not be appropriate proxy
decision-makers about fertility on behalf of AYA patients.71,72
It is also worth noting that AYAs with cancer are as
sexually active as their peers without cancer.73-76 Thus,
health care professionals should be aware of intimacy
concerns and changes due to treatment and have tactful and meaningful conversations with AYAs about reported adverse events and solutions, if possible. Equally important is for the oncologist to refer AYA patients
to contraception counseling, because AYAs have been
October 2016, Vol. 23, No. 4
shown to take more sexual risk, such as not using adequate contraceptive measures.77,78 Therefore, contraception should be a key discussion point with patients
and their medical team, because some patients may assume they are infertile due to their treatment and think
they have no need for contraception for pregnancy
purposes or they may engage in unprotected sex to
verify the fact that their infertility has been impaired.79
It is also worth noting that patients receiving active
treatment are more susceptible to sexually transmitted
infections because their immune systems are compromised.77,80 Pregnancy while undergoing cancer treatment can pose risks to the fetus as well as the patient,
and making difficult decisions to terminate a pregnancy may cause increased stress and turmoil for a young
patient already trying to cope with a cancer diagnosis
and treatment.
Discussions of potential threats to fertility with all
patients of childbearing age with newly diagnosed cancer are recommended by several medical organizations,
including as the American Society of Clinical Oncology,40 the National Comprehensive Cancer Network,68
and the American Society for Reproductive Medicine.81
In addition to discussion about the potential impact of
treatment or cancer type on fertility, the American Society of Clinical Oncology and other resources recommend that AYA patients be referred to a reproductive
endocrinologist or infertility specialist to discuss options for fertility preservation as soon as possible after
a diagnosis of cancer is received and prior to the start
of treatment (see Table 1).4,35-40 The established fertility
preservation methods are sperm cryopreservation for
postpubertal males and oocyte and embryo cryopreservation for postpubertal females as well as ovarian transposition for patients undergoing pelvic or abdominal
radiotherapy.37,40 For prepubertal children, fertility preservation options are experimental and include testicular
or ovarian tissue cryopreservation.40
Studies suggest that patients who do not learn
about potential threats to their fertility may experience feelings of remorse and regret during the survivorship period.82-84 Although few patients (particularly females) opt to use fertility-preservation methods,
most patients report satisfaction and appreciation for
learning about this information from their health care
professional.85,86
Progress of Cancer Biology
Though many common diagnoses afflicting the AYA
population overlap with those seen in younger and
older patients, new data have emerged showing differences in disease biology by age.8,87,88
Acute lymphoblastic leukemia (ALL) is the most
common malignancy and leading cause of cancer-related deaths in AYAs.88 Outcomes of AYAs treated for
ALL are worse for those aged 10 years or older.8,88 The
Cancer Control 429
treatment varies: In adult-based protocols, transplants
are more common, whereas pediatric-based protocols
call for intensive therapy, including asparaginase, and
do not rely as heavily on transplants.89-92 A difference
exists in the frequency of cytogenetic abnormalities
that predispose AYAs to worse outcomes, but it may
also serve as a target for improved therapy.8,10,93,94
Opportunities for improved outcomes may also exist in Philadelphia chromosome–positive ALL when
tyrosine kinase inhibitors are added to treatment
(Table 4).7-10,88,93-102 A large prospective trial of 296 patients aged 16 to 39 years demonstrated that treatment
with an intensive pediatric regimen showed significant rates of improvement in event-free survival (66%)
and overall survival (78%) along with manageable toxicities.89,90 Other trials have shown superior outcomes
in AYAs treated in pediatric protocols.89 Such ongoing
collaborations with cooperative groups may lead to
uniform treatment based on biology, with age being
one among many other variables.
Sarcoma is a common type of cancer in AYAs.103-107
Overall, these mesenchymal-derived malignancies of
the soft tissue and bone represent 8% of new diagnoses in patients aged 15 to 39 years.108 Standard chemotherapies for front-line regimens for osteosarcoma,
rhabdomyosarcoma, and Ewing sarcoma have been
established through cooperative group studies.103-107
In these studies, pediatric patients typically comprise
the majority of the groups. Standard therapies for other
soft-tissue sarcomas have been primarily derived from
studies of adult populations, in whom these diagnoses
are more common.109,110 Oftentimes, no clear consensus exists regarding systemic therapies for metastatic,
relapsed, or recurrent disease.108 Some retrospective
reviews of single-institution experiences and cooperative group studies explore the presentation, biology,
and therapeutic variables and how they generally negatively impact the outcomes of AYA patients compared
with younger patients (see Table 4).7-10,88,93-102,111-113 Effort has been made to collaborate through the adultand pediatric-focused cooperative groups and may
serve as a model for future trials.38
Breast cancer has an age-related biological difference that dictates outcomes.99,100,114,115 Breast cancer
is one of the most frequently diagnosed malignancies among women aged 15 to 39 years and makes
up 14% of all cancers diagnosed in AYAs.9 Nearly 7%
of all breast cancers are diagnosed in women younger than 40 years of age.8,99 AYAs have a higher incident rate of more aggressive phenotypes, such as
triple-negative breast cancer, ERBB2 (formerly known
as HER2/neu)-overexpressing cancers, and luminal B cancers.9,99,100,114,115 A low proportion of AYAs
present with luminal A disease, which confers a better prognosis.100 In addition, a higher proportion of
AYAs present with stage III/IV disease.99 AYA pa430 Cancer Control
Table 4. — Select Characteristics and Features of
Cancers in AYA
Type
of Cancer
Description
Leukemia8,10,93-97 Ph+ ALL t(9,22) BCR-ABL
3% of children
3%–25% of AYAs
T-cell leukemia
Age 1–9 y: 12.8%
Age 10–15 y: 16.9%
Age 16–21 y: 24.7%
Chromosome 21 (iAMP21)
Increase incidence in patients aged > 10 y
Hypodiploid
MLL at 11q23
TEL-AML1 translocation (ETV6-RUNX1)
50% in children
10% in AYA
Sarcoma101,102
Ewing sarcoma
More common in AYAs aged ≥ 15 y than children
Increasing age associated with poor prognosis
Osteosarcoma
Poor event-free survival associated with age ≥ 18 y
Rhabdomyosarcoma
Higher prevalence of alveolar subtype in AYAs,
conferring a poorer prognosis
Colorectal
cancer 10
High incidence of microsatellite instability
High incidence of heritable forms of colorectal
cancer (adenomatous polyposis, nonpolyposis
colon cancer)
Breast
cancer 99,100
Higher prevalence of BRAC1/BRAC2 mutations
Present with stage III/IV disease
Higher incidence of aggressive pathology
Triple-negative breast cancer
Age 15–39 y: 22.8%
Age 40–49 y: 14.3%
Age ≥ 50 y: 11.7%
ERBB2-overexpressing cancer
Age 15–39 y: 10.3%
Age 40–49 y: 6.9%
Age ≥ 50 y: 5.9%
Luminal B cancer
Age 15–39 y: 17.9%
Age 40–49 y: 14.9%
Age ≥ 50 y: 10.7%
Melanoma88
Risk factors: congenital melanocytic nevi,
xeroderma pigmentosa, dysplastic nevus, prior
treatment for childhood cancer
Comprehensive staging guidelines not established
for children and adolescents
Metastases to sentinel lymph nodes occur more
frequently in children and AYAs than in adults with
same disease stage
ALL = acute lymphoblastic leukemia,
AYA = adolescent and young adult,
Ph+ = Philadelphia chromosome positive.
October 2016, Vol. 23, No. 4
tients with breast cancer have a high prevalence of
BRCA1/BRCA2 mutations.9,99,100,114,115
CRC is another example of the biological and genetic differences seen in AYAs compared with older adults.
The AYA population has high incidence rates of microsatellite instability and heritable forms of CRC such as
familial adenomatous polyposis and hereditary nonpolyposis colon cancer.10 It may also be true that the disease
biology of young patients with CRC but without inherited syndromes may differ from the type of CRC seen
in older adults.10 Overall, compared with older patients,
AYAs present with more advanced-stage disease at diagnosis and more aggressive tumor features.116
Melanoma can affect individuals at any age, but it
is more common in adults than children.87 Its basic disease biology — numerous mutations and a preponderance of BRAF V600E mutations — appears to hold true
across all ages.117 Metastases to sentinel lymph nodes
occur more frequently in children and AYAs than in
adults with the same stage of disease.88 With the approval of ipilimumab, nivolumab, and other inhibitors
and combination therapies, we question whether AYAs
were adequately represented and enrolled in recent
successful trials.118 Based on the biology of melanoma,
similar treatment outcomes are expected in the AYA
population.118
Remaining Challenges
The management of cancer in AYA patients is a continuous process that has experienced progress in recent
years. We now understand more about what AYA patients need and are poised to learn even more about
possible approaches to improve many aspects of AYA
care. However, doing so requires future research that
emphasizes the biological differences in certain diseases to develop targeted treatments for this population.
Lack of health insurance among this age group
is still a major concern because being uninsured can
potentially delay diagnosis and access to treatment as
well as to the possibility of inhibiting any transition to
surveillance and life-long survivorship care. A large
study looking at the unmet needs of AYA patients reported that 50% of AYA participants had unmet information needs about cancer recurrence, cancer treatments, and long-term adverse events, and up to 50% of
AYA patients had unmet information needs on fertility,
nutrition, and financial support.44
Conclusions
Adolescent and young adult (AYA) patients face disparities based on age that can lead to a delay in diagnosis,
an advanced stage of disease at presentation, suboptimal treatment location, lack of clinical trial availability, and an increased burden of quality survivorship
through the loss of fertility or disruption in social,
personal, or financial growth and stability. These disOctober 2016, Vol. 23, No. 4
parities in AYAs have inspired individuals and institutions to develop programs designed for AYAs to meet
the specific or overall health care needs of this patient
population. In addition, some organizations have advocated for AYA cancer care as a specific discipline
within oncology; to improve fertility education and access to preservation; to address the root causes of the
lack of survival improvement; and to define the essential elements of care for this population. Today we understand more about what AYA patients need, and we
are poised to learn even more about approaches to improve many aspects of care for these patients. We are
optimistic that motivated patients, health care professionals, advocacy organizations, and institutions will
align to develop improved models for the care of AYAs.
References
1. Soliman H, Agresta S. Current issues in adolescent and young
adult cancer survivorship. Cancer Control. 2008;15(1):55-62.
2. Young Adult Cancer Alliance. Critical Mass Annual Conference.
November 4, 2015–November 6, 2015; Chicago, IL. http://criticalmass.org/
the-annual-conference/2015-conference. Accessed October 18, 2016.
3. National Cancer Institute. Cancer health disparities. https://www.
cancer.gov/about-nci/organization/crchd/cancer-health-disparities-fact-sheet.
Accessed October 5, 2016.
4. Adolescent and Young Adult Oncology Program Review Group.
Closing the Gap: Research and Care Imperatives for Adolescents And
Young Adults With Cancer. Bethesda, MD: US Department of Health and
Human Services; National Institutes of Health; National Cancer Institute;
LIVESTRONG Young Adult Alliance; 2006. https://www.cancer.gov/types/
aya/research/ayao-august-2006.pdf. Accessed October 5, 2016.
5. Lewis DR, Seibel NL, Smith AW, et al. Adolescent and Young Adult
Cancer Survival. J Natl Cancer Inst Monogr. 2014;2014(49):228-235.
6. National Cancer Institute; Surveillance, Epidemiology, and End Results. A snapshot of adolescent and young adult cancer. https://www.cancer.gov/research/progress/snapshots/adolescent-young-adult. Accessed
October 5, 2016.
7. Schafer ES, Hunger SP. Optimal therapy for acute lymphoblastic leukemia in adolescents and young adults. Nat Rev Clin Oncol. 2011;8(7):417-424.
8. Bleyer A, Barr R, Hayes-Lattin B, et al. The distinctive biology of cancer
in adolescents and young adults. Nat Rev Cancer. 2008;8(4):288-298.
9. Keegan TH, Press DJ, Tao L, et al. Impact of breast cancer subtypes on 3-year survival among adolescent and young adult women.
Breast Cancer Res. 2013;15(5):R95.
10. Tricoli JV, Seibel NL, Blair DG, et al. Unique characteristics of adolescent and young adult acute lymphoblastic leukemia, breast cancer, and
colon cancer. J Natl Cancer Inst. 2011;103(8):628-635.
11. Zbuk K, Sidebotham EL, Bleyer A, et al. Colorectal cancer in young
adults. Semin Oncol. 2009;36(5):439-450.
12. Martin S, Ulrich C, Munsell M, et al. Delays in cancer diagnosis in underinsured young adults and older adolescents. Oncologist. 2007;12(7):816-824.
13. Grassley CE, ed. Living Without Health Insurance: Hearing Before
the Committee on Finance, US Senate. Darby, PA: Diane Publishing; 2001.
14. Smith JC, Medalia C. Health Insurance Coverage in the United
States: 2013. Washington, DC: US Department of Commerce; Economics
and Statistics Administration; US Census Bureau; 2014. http://www.nber.
org/cps/hi/2014redesign/p60-250.pdf. Accessed October 5, 2016.
15. Aizer AA, Falit B, Mendu ML, et al. Cancer-specific outcomes among
young adults without health insurance. J Clin Oncol. 2014;32(19):2025-2030.
16. Smith EC, Ziogas A, Anton-Culver H. Association between insurance
and socioeconomic status and risk of advanced stage Hodgkin lymphoma
in adolescents and young adults: Hodgkin’s lymphoma in adolescents and
young adults. Cancer. 2012;118(24):6179-6187.
17. Bleyer A, Ulrich C, Martin S. Young adults, cancer, health insurance,
socioeconomic status, and the Patient Protection and Affordable Care Act.
Cancer. 2012;118(24):6018-6021.
18.Rosenberg AR, Kroon L, Chen L, et al. Insurance status and
risk of cancer mortality among adolescents and young adults. Cancer.
2015;121(8):1279-1286.
19. Bellizzi KM, Smith A, Schmidt S, et al. Positive and negative psychosocial impact of being diagnosed with cancer as an adolescent or young adult.
Cancer. 2012;118(20):5155-5162.
20. Robbins AS, Han X, Ward EM, et al. Association between the Affordable Care Act dependent coverage and cervical cancer stage and treatment in
young women. JAMA. 2015;314(20):2189-2191.
Cancer Control 431
21. Shaw PH, Reed DR, Yeager N, et al. Adolescent and young adult
(AYA) oncology in the United States: a specialty in its late adolescence.
J Pediatr Hematol Oncol. 2015;37(3):161-169.
22. Wolfson JA, Sun CL, Wyatt LP, et al. Impact of care at comprehensive
cancer centers on outcome: results from a population-based study. Cancer.
2015;121(21):3885-3893.
23. Yeager ND, Hoshaw-Woodard S, Ruymann FB, et al. Patterns of
care among adolescents with malignancy in Ohio. J Pediatr Hematol Oncol.
2006;28(1):17-22.
24. Wolfson J, Sun CL, Kang T, et al. Impact of treatment site in adolescents and young adults with central nervous system tumors. J Natl Cancer
Inst. 2014;106(8):dju166.
25. Parsons HM, Harlan LC, Seibel NL, et al. Clinical trial participation
and time to treatment among adolescents and young adults with cancer:
does age at diagnosis or insurance make a difference? J Clin Oncol.
2011;29(30):4045-4053.
26. Fern L, Davies S, Eden T, et al. Rates of inclusion of teenagers and
young adults in England into National Cancer Research Network clinical trials:
report from the National Cancer Research Institute (NCRI) Teenage and Young
Adult Clinical Studies Development Group. Br J Cancer. 2008;99(12):1967-1974.
27. McTiernan A. Issues surrounding the participation of adolescents with
cancer in clinical trials in the UK. Eur J Cancer Care (Engl). 2003;12(3):233-239.
28. Shaw PH, Ritchey AK. Different rates of clinical trial enrollment between adolescents and young adults aged 15 to 22 years old and children under 15 years old with cancer at a children’s hospital. J Pediatr Hematol Oncol.
2007;29(12):811-814.
29. Collins CL, Malvar J, Hamilton AS, et al. Case-linked analysis of
clinical trial enrollment among adolescents and young adults at a National
Cancer Institute-designated comprehensive cancer center. Cancer.
2015;121(24):4398-4406.
30. Albritton KH, Coccia P. Influencing referral of adolescents and young
adults with cancer to sites with higher rates of trial enrollment. Pediatrics.
2014;133(suppl 3):S104-S108.
31. Albritton KH, Wiggins CH, Nelson HE, et al. Site of oncologic specialty care for older adolescents in Utah. J Clin Oncol. 2007;25(29):4616-4621.
32.Felgenhauer J, Hooke MC. Regulatory barriers to clinical trial
enrollment of adolescent and young adult oncology patients. Pediatrics.
2014;133(suppl 3):S119-S122.
33. Tai E, Buchanan N, Eliman D, et al. Understanding and addressing
the lack of clinical trial enrollment among adolescents with cancer. Pediatrics.
2014;133(suppl 3):S98-S103.
34. Reed DR, Block RG, Johnson R. Creating an adolescent and young
adult cancer program: lessons learned from pediatric and adult oncology
practice bases. J Natl Compr Canc Netw. 2014;12(10):1409-1415.
35. Ferrari A, Thomas D, Franklin AR, et al. Starting an adolescent and
young adult program: some success stories and some obstacles to overcome.
J Clin Oncol. 2010;28(32):4850-4857.
36. Nass SJ, Beaupin LK, Demark-Wahnefried W, et al. Identifying and
addressing the needs of adolescents and young adults with cancer: summary of an Institute of Medicine workshop. Oncologist. 2015;20(2):186-195.
37. National Comprehensive Cancer Network. Adolescent and Young
Adult (AYA) Oncology. V1.2016. https://www.nccn.org/professionals/
physician_gls/pdf/aya.pdf. Accessed October 5, 2016.
38. Weiss AR, Nichols CR, Freyer DR. Enhancing adolescent and young
adult oncology research within the National Clinical Trials Network: rationale,
progress, and emerging strategies. Semin Oncol. 2015;42(5):740-747.
39. Coccia PF, Altman J, Bhatia S, et al. Adolescent and young adult
oncology. J Natl Compr Canc Netw. 2012;10(9):1112-1150.
40. Loren AW, Mangu PB, Beck LN, et al. Fertility preservation for patients with cancer: American Society of Clinical Oncology clinical practice
guideline update. J Clin Oncol. 2013;31(19):2500-2510.
41. Shaw PH, Boyiadzis M, Tawbi H, et al. Improved clinical trial enrollment in adolescent and young adult (AYA) oncology patients after the
establishment of an AYA oncology program uniting pediatric and medical
oncology divisions. Cancer. 2012;118(14):3614-3617.
42. D’Agostino NM, Penney A, Zebrack B. Providing developmentally
appropriate psychosocial care to adolescent and young adult cancer survivors. Cancer. 2011;117(10 suppl):2329-2334.
43. Wilkins KL, D’Agostino N, Penney AM, et al. Supporting adolescents and young adults with cancer through transitions: position statement
from the Canadian Task Force on Adolescents and Young Adults With
Cancer. J Pediatr Hematol Oncol. 36(7):545-551.
44. Keegan TH, Lichtensztajn DY, Kato I, et al. Unmet adolescent and
young adult cancer survivors information and service needs: a populationbased cancer registry study. J Cancer Surviv. 2012;6(3):239-250.
45. Clerici CA, Massimino M, Casanova M, et al. Psychological referral
and consultation for adolescents and young adults with cancer treated at
pediatric oncology unit. Pediatr Blood Cancer. 2008;51(1):105-109.
46. Sansom-Daly UM, Wakefield CE, Bryant RA, et al. Online groupbased cognitive-behavioural therapy for adolescents and young adults
after cancer treatment: a multicenter randomised controlled trial of Recapture Life-AYA. BMC Cancer. 2012;12(1):339.
47. Zebrack BJ, Corbett V, Embry L, et al. Psychological distress and
unsatisfied need for psychosocial support in adolescent and young adult
432 Cancer Control
cancer patients during the first year following diagnosis. Psychooncology.
2014;23(11):1267-1275.
48. Smith AW, Parsons HM, Kent EE, et al. Unmet support service
needs and health-related quality of life among adolescents and young
adults with cancer: the AYA HOPE study. Front Oncol. 2013;3:75.
49. Kyngäs H, Mikkonen R, Nousiainen EM, et al. Coping with the onset
of cancer: coping strategies and resources of young people with cancer.
Eur J Cancer Care (Engl). 2001;10(1):6-11.
50. Zebrack BJ, Mills J, Weitzman TS. Health and supportive care
needs of young adult cancer patients and survivors. J Cancer Surviv.
2007;1(2):137-145.
51. Zebrack B, Bleyer A, Albritton K, et al. Assessing the health care
needs of adolescent and young adult cancer patients and survivors. Cancer.
2006;107(12):2915-2923.
52. Cacioppo JT, Hawkley LC. Social isolation and health, with an emphasis
on underlying mechanisms. Perspect Biol Med. 2003;46(3 suppl):S39-S52.
53. Murphy D, Klosky JL, Reed DR, et al. The importance of assessing
priorities of reproductive health concerns among adolescent and young
adult patients with cancer. Cancer. 2015;121(15):2529-2536.
54. Nightingale CL, Quinn GP, Shenkman EA, et al. Health-related
quality of life of young adult survivors of childhood cancer: a review of
qualitative studies. J Adolesc Young Adult Oncol. 2011;1(3):124-132.
55. Olsen PR, Harder I. Caring for teenagers and young adults with
cancer: a grounded theory study of network-focused nursing. Eur J Oncol
Nurs. 2011;15(2):152-159.
56. Rechis R; LIVESTRONG Foundation. Session 2: unique risks &
vulnerabilities of AYA patients across the lifespan: AYA survey results.
Published July 2013. https://www.nationalacademies.org/hmd/~/media/Files/
Activity%20Files/Disease/NCPF/2013-JUL-15/2A_Rechis.pdf. Accessed
October 5, 2016.
57. Durall A, Zurakowski D, Wolfe J. Barriers to conducting advance
care discussions for children with life-threatening conditions. Pediatrics.
2012;129(4):e975-e982.
58. Feudtner C. Collaborative communication in pediatric palliative care:
a foundation for problem-solving and decision-making. Pediatr Clin North
Am. 2007;54(5):583-607.
59. Davies B, Sehring SA, Partridge JC, et al. Barriers to palliative care
for children: perceptions of pediatric health care providers. Pediatrics.
2008;121(2):282-288.
60. Glare PA. Early implementation of palliative care can improve patient
outcomes. J Natl Compr Canc Netw. 2013;11(suppl 1):S3-S9.
61. Wiener L, Ballard E, Brennan T, et al. How I wish to be remembered: the use of an advance care planning document in adolescent and
young adult populations. J Palliat Med. 2008;11(10):1309-1313.
62. Otsuka M, Koyama A, Matsuoka H, et al. Early palliative intervention
for patients with advanced cancer. Jpn J Clin Oncol. 2013;43(8):788-794.
63. Lyon ME, Jacobs S, Briggs L, et al. Family-centered advance care
planning for teens with cancer. JAMA Pediatr. 2013;167(5):460.
64. Wicks L, Mitchell A. The adolescent cancer experience: loss of control and benefit finding: Adolescent cancer experience: control and benefit
finding. Eur J Cancer Care (Engl). 2010;19(6):778-785.
65. Loberiza FR, Swore-Fletcher BA, Block SD, et al. Coping styles,
health status and advance care planning in patients with hematologic
malignancies. Leuk Lymphoma. 2011;52(12):2342-2348.
66. Moss JL, Choi AW, Fitzgerald Keeter MK, et al. Male adolescent
fertility preservation. Fertil Steril. 2016;105(2):267-273.
67. Levine JM, Kelvin JF, Quinn GP, et al. Infertility in reproductiveage female cancer survivors: infertility in female cancer survivors. Cancer.
2015;121(10):1532-1539.
68. Johnson RH, Kroon L. Optimizing fertility preservation practices for
adolescent and young adult cancer patients. J Natl Compr Canc Netw.
2013;11(1):71-77.
69. Quinn GP, Vadaparampil ST, Bell-Ellison BA, et al. Patient-physician
communication barriers regarding fertility preservation among newly
diagnosed cancer patients. Soc Sci Med. 2008;66(3):784-789.
70. Quinn GP, Vadaparampil ST, Lee JH, et al. Physician referral for
fertility preservation in oncology patients: a national study of practice
behaviors. J Clin Oncol. 2009;27(35):5952-5957.
71. Crawshaw M. Psychosocial oncofertility issues faced by adolescents and young adults over their lifetime: a review of the research. Hum
Fertil. 2013;16(1):59-63.
72. Quinn GP, Vadaparampil ST. Fertility Preservation and adolescent/
young adult cancer patients: physician communication challenges. J Adolesc
Health. 2009;44(4):394-400.
73. Vadaparampil ST, Clayton H, Quinn GP, et al. Pediatric oncology nurses’ attitudes related to discussing fertility preservation with pediatric cancer
patients and their families. J Pediatr Oncol Nurs. 2007;24(5):255-263.
74. King L, Quinn GP, Vadaparampil ST, et al. Oncology nurses’ perceptions of barriers to discussion of fertility preservation with patients with
cancer. Clin J Oncol Nurs. 2008;12(3):467-476.
75. Clayton H, Quinn GP, Lee JH, et al. Trends in clinical practice and
nurses’ attitudes about fertility preservation for pediatric patients with cancer. Oncol Nurs Forum. 2008;35(2):249-255.
76. Fallowfield L, Saul J, Gilligan B. Teaching senior nurses how to
October 2016, Vol. 23, No. 4
teach communication skills in oncology. Cancer Nurs. 2001;24(3):185-191.
77. Murphy D, Klosky JL, Termuhlen A, et al. The need for reproductive
and sexual health discussions with adolescent and young adult cancer
patients. Contraception. 2013;88(2):215-220.
78. Maslow BS, Morse CB, Schanne A, et al. Contraceptive use and the
role of contraceptive counseling in reproductive-aged women with cancer.
Contraception. 2014;90(1):79-85.
79. Benedict C, Shuk E, Ford JS. Fertility issues in adolescent and
young adult cancer survivors. J Adolesc Young Adult. 2016;5(1):48-57.
80. Laurence V, Gbolade BA, Morgan SJ, et al. Contraception for teenagers and young adults with cancer. Eur J Cancer. 2004;40(18):2705-2716.
81. Ethics Committee of American Society for Reproductive Medicine.
Fertility preservation and reproduction in patients facing gonadotropic
therapies: a committee opinion. Fertil Steril. 2013;100(5):1224-1231.
82. Benedict C, Thom B, Kelvin JF. Young adult female cancer survivors’ decision regret about fertility preservation. J Adolesc Young Adult
Oncol. 2015;4(4):213-218.
83. Crawshaw M, Sloper P. A Qualitative Study of the Experiences of
Teenagers and Young Adults When Faced With Possible or Actual Fertility
Impairment Following Cancer Treatment. York, UK: University of York; 2006.
http://www.york.ac.uk/inst/spru/pubs/pdf/fertilityExecSumm.pdf. Accessed
October 5, 2016.
84. Chapple A, Salinas M, Ziebland S, et al. Fertility issues: the perceptions and experiences of young men recently diagnosed and treated for
cancer. J Adolesc Health. 2007;40(1):69-75.
85. Garvelink MM, ter Kuile MM, Bakker RM, et al. Women’s experiences
with information provision and deciding about fertility preservation in the
Netherlands: “satisfaction in general, but unmet needs.” Health Expect.
2015;18(5):956-968.
86. Letourneau JM, Ebbel EE, Katz PP, et al. Pretreatment fertility
counseling and fertility preservation improve quality of life in reproductive
age women with cancer. Cancer. 2012;118(6):1710-1717.
87. American Cancer Society. Cancer Facts & Figures 2016. http://
www.cancer.org/acs/groups/content/@research/documents/document/
acspc-047079.pdf. Accessed October 18, 2016.
88. Tricoli JV, Blair DG, Anders CK, et al. Biologic and clinical characteristics of adolescent and young adult cancers: acute lymphoblastic leukemia, colorectal cancer, breast cancer, melanoma, and sarcoma. Cancer.
2016;122(7):1017-1028.
89. Curran E, Stock W. How I treat acute lymphoblastic leukemia in
older adolescents and young adults. Blood. 2015;125(24):3702-3710.
90. Stock W, Luger SM, Advani AS, et al. Favorable outcomes for
older adolescents and young adults (AYA) with acute lymphoblastic
leukemia (ALL): early results of U.S. Intergroup trial C10403. Blood.
2014;124(21 suppl):796.
91. Isakoff M, Freyer D, Bleyer A. Young adults with acute lymphoblastic
leukemia treated with a pediatric-inspired regimen do not need a bone
marrow transplant in first remission. Blood. 2013;121(26):5253-5255.
92. Ram R, Wolach O, Vidal L, et al. Adolescents and young adults with
acute lymphoblastic leukemia have a better outcome when treated with
pediatric-inspired regimens: Systematic review and meta-analysis. Am J
Hematol. 2012;87(5):472-478.
93. Perez-Andreu V, Roberts KG, Xu H, et al. A genome-wide association
study of susceptibility to acute lymphoblastic leukemia in adolescents and
young adults. Blood. 2015;125(4):680-686.
94. Mullighan CG, Willman CL. Advances in the biology of acute lymphoblastic leukemia—from genomics to the clinic. J Adolesc Young Adult
Oncol. 2011;1(2):77-86.
95. Bleyer WA, Barr RD, eds. Pediatric Oncology: Cancer in Adolescents
and Young Adults. New York: Springer; 2007.
96. Roberts KG, Li Y, Payne-Turner D, et al. Targetable kinaseactivating lesions in Ph-like acute lymphoblastic leukemia. N Engl J Med.
2014;371(11):1005-1015.
97. Pui CH, Yang JJ, Hunger SP, et al. Childhood acute lymphoblastic leukemia: progress through collaboration. J Clin Oncol. 2015;33(27):2938-2948.
98. Janeway KA, Barkauskas DA, Krailo MD, et al. Outcome for adolescent and young adult patients with osteosarcoma: a report from the Children’s
Oncology Group. Cancer. 2012;118(18):4597-4605.
99. Keegan TH, DeRouen MC, Press DJ, et al. Occurrence of breast
cancer subtypes in adolescent and young adult women. Breast Cancer
Res. 2012;14(2):R55.
100. Assi HA, Khoury KE, Dbouk H, et al. Epidemiology and prognosis
of breast cancer in young women. J Thorac Dis. 2013;5(suppl 1):S2-S8.
101. Janeway KA, Pappo A. Treatment guidelines for gastrointestinal
stromal tumors in children and young adults. J Pediatr Hematol Oncol.
2012;34:S69-S72.
102. Bisogno G, Compostella A, Ferrari A, et al. Rhabdomyosarcoma in
adolescents: a report from the AIEOP Soft Tissue Sarcoma Committee.
Cancer. 2012;118(3):821-827.
103. Marina N, Bielack S, Whelan J, et al. International collaboration is
feasible in trials for rare conditions: the EURAMOS experience. In: Jaffe
N, Bruland OS, Bielack S, eds. Pediatric and Adolescent Osteosarcoma.
Boston: Springer US; 2010.
104.Gorlick R, Janeway K, Lessnick S, et al. Children’s Oncology
October 2016, Vol. 23, No. 4
Group’s 2013 blueprint for research: bone tumors.. Pediatr Blood Cancer.
2013;60(6):1009-1015.
105. Womer RB, West DC, Krailo MD, et al. Randomized controlled trial
of interval-compressed chemotherapy for the treatment of localized
Ewing sarcoma: a report from the Children’s Oncology Group. J Clin Oncol.
2012;30(33):4148-4154.
106.Malempati S, Hawkins DS. Rhabdomyosarcoma: review of the
Children’s Oncology Group (COG) Soft-Tissue Sarcoma Committee experience and rationale for current COG studies. Pediatr Blood Cancer.
2012;59(1):5-10.
107. Weiss A, Gill J, Goldberg J, et al. Advances in therapy for pediatric
sarcomas. Curr Oncol Rep. 2014;16:395.
108. Amankwah EK, Conley AP, Reed DR. Epidemiology and therapies
for metastatic sarcoma. Clin Epidemiol. 2013;5:147-162.
109. Judson I, Verweij J, Gelderblom H, et al. Doxorubicin alone versus
intensified doxorubicin plus ifosfamide for first-line treatment of advanced
or metastatic soft-tissue sarcoma: a randomised controlled phase 3 trial.
Lancet Oncol. 2014;15(4):415-423.
110. van der Graaf WT, Blay JY, Chawla SP, et al. Pazopanib for metastatic soft-tissue sarcoma (PALETTE): a randomised, double-blind, placebocontrolled phase 3 trial. Lancet. 19;379(9829):1879-1886.
111. Streby KA, Ruymann FB, Whiteside S, et al. Rhabdomyosarcoma
in adolescent and young adults: a 25-year review at Nationwide Children’s
Hospital. J Adolesc Young Adult Oncol. 2012;1(4):164-167.
112. Collins M, Wilhelm M, Conyers R, et al. Benefits and adverse
events in younger versus older patients receiving neoadjuvant chemotherapy for osteosarcoma: findings from a meta-analysis. J Clin Oncol.
2013;31(18):2303-2312.
113. Haddox CL, Han G, Anijar L, et al. Osteosarcoma in pediatric patients and young adults: a single institution retrospective review of presentation, therapy, and outcome. Sarcoma. 2014;2014: 402509.
114. Parise CA, Bauer KR, Caggiano V. Variation in breast cancer subtypes with age and race/ethnicity. Crit Rev Oncol Hematol. 2010;76(1):44-52.
115. Gewefel H, Salhia B. Breast cancer in adolescent and young adult
women. Clin Breast Cancer. 2014;14(6):390-395.
116. Teng A, Lee DY, Cai J, Patel SS, et al. Patterns and outcomes
of colorectal cancer in adolescents and young adults. J Surg Res.
2016;205(1):19-27.
117. Lu C, Zhang J, Nagahawatte P. The genomic landscape of childhood and adolescent melanoma. J Invest Dermatol. 2015;135(3):816-823.
118. Sreeraman Kumar R, Messina JL, Sondak VK, et al. Treating melanoma in adolescents and young adults: challenges and solutions. Clin Oncol
Adolesc Young Adults. 2015;5:75-86.
Cancer Control 433
Tobacco-related health disparities
exist at each point of
the cancer care continuum.
Iris. X-Ray image courtesy of Bryan Whitney. www.x-rayphotography.com.
Tobacco-Related Health Disparities Across the
Cancer Care Continuum
Vani Nath Simmons, PhD, Bárbara Piñeiro, PhD, Monica Webb Hooper, PhD, Jhanelle E. Gray, MD,
and Thomas H. Brandon, PhD
Background: Use of tobacco is the leading preventable cause of death in the United States. Racial/ethnic minorities and individuals of low socioeconomic status disproportionately experience tobacco-related disease
and illness. Unique challenges and circumstances exist at each point in the cancer care continuum that may
contribute to the greater cancer burden experienced by these groups.
Methods: We reviewed tobacco-related disparities from cancer prevention to cancer survivorship. We also
describe research that seeks to reduce tobacco-related disparities.
Results: Racial/ethnic minorities and low-income individuals experience unique social and environmental
contextual challenges such as greater environmental cues to smoke and greater levels of perceived stress and
social discrimination. Clinical practice guidelines support the effectiveness of pharmacotherapy and behavioral
counseling for racial and ethnic minorities, yet smoking cessation rates are lower in this group when compared
with non-Hispanic whites. Superior efficacy for culturally adapted interventions has not yet been established.
Conclusions: To reduce health disparities in this population, a comprehensive strategy is needed with efforts directed at each point along the cancer care continuum. Strategies are needed to reduce the impact of
contextual factors such as targeted tobacco marketing and social discrimination on smoking initiation and
maintenance. Future efforts should focus on increasing the use of evidence-based cessation treatment methods
and studying its effectiveness in these populations. Attention must also be focused on improving treatment
outcomes by reducing smoking in diverse racial and ethnic patient populations.
From the Health Outcomes and Behavior Program (VNS, THB)
and Department of Medical Oncology (JEG), H. Lee Moffitt Cancer
Center & Research Institute, Tampa, Florida, Department of
Clinical Psychology and Psychobiology (BP), University of
Santiago de Compostela, Santiago de Compostela, Spain, and the
Department of Psychology (MWH), Sylvester Comprehensive
Cancer Center, University of Miami, Miami, Florida.
Submitted April 13, 2016; accepted July 28, 2016.
Address correspondence to Vani N. Simmons, PhD, Health Outcomes
and Behavior Program, Moffitt Cancer Center, 4115 East Fowler Avenue, Tampa, FL 33617. E-mail: [email protected]
Dr Brandon receives grants and research support from Pfizer and is a
consultant for Voxiva and the Australian General Solicitor. The other
authors report no significant relationships with the companies/organizations whose products or services may be referenced in this article.
434 Cancer Control
Introduction
In the United States, tobacco smoking represents the
leading preventable cause of death.1,2 Nearly one-third
of all cancer-related deaths are attributable to tobacco
use.1-3 The health burden of tobacco use is not equally
distributed: Racial and ethnic minorities who smoke
and have lower levels of education, income, and occupational status disproportionately experience tobacco-related disease burden and illness.4 Examples of
tobacco-related disparities include exposure to tobacco
smoke, current patterns of use and cessation, subsequent health consequences among specific population
October 2016, Vol. 23, No. 4
groups, capacity and infrastructure, and access to resources.4 Attention to tobacco-related health disparities
has been growing, and the number of priority populations has extended beyond racial and ethnic minorities
to include individuals of low socioeconomic status as
well as sexual minorities (lesbian, gay, bisexual, and
transgender).5 In addition, those in the tobacco-related
health disparities field have moved beyond descriptive
studies and have begun to develop and test interventions to help reduce smoking among these vulnerable
populations.6
Although tobacco use contributes to the presence
and exacerbation of health disparities with respect to
health outcomes such as stroke, diabetes, and coronary
heart disease, this paper specifically focuses on the
association between tobacco use and cancer.7 Unique
challenges and circumstances also exist at different
points along the cancer care continuum that contribute
to the larger cancer burden experienced by ethnic/racial
minorities and underserved populations (Fig).3,4,8-27
Cancer Prevention
dition to the potential health outcomes of increased
exposure to second-hand smoke, less-restrictive policies may serve to maintain smoking behavior. Compared with workplaces that do not have smoking restrictions, workplaces that prohibit smoking result in
fewer cigarettes smoked, greater cessation attempts,
and more quitting successes among employees.30 Research also suggests that lack of self-imposed smoking
restrictions in the home contributes to greater maintenance of smoking behavior, whereas the adoption
of smoke-free restrictions results in greater cessation
ability.31 Individuals who smoke, have a lower socioeconomic status, and are African American are less
likely than non-Hispanic whites to impose smoking
bans in the home and automobile; thus, environmental exposure in these settings is an additional barrier
to consider.10-13 Disproportionate exposure to tobacco
smoke is also a public health issue among residents of
multiunit housing properties who may be exposed to
second-hand smoke due to shared air between units
and poor ventiliation.32 Residents of subsidized housing also experience high levels of stress, which can
impact smoking behavior.33
Evidence suggests that cigarette smoking is associated
with increased risks of developing oropharyngeal, laryngeal, esophageal, tracheal, bronchial, lung, stomTargeted Marketing
ach, liver, pancreatic, kidney, ureteral, cervical, bladAnother key factor that influences the initiation and
der, and colorectal cancers as well as acute myeloid
maintenance of tobacco smoking among low-income
leukemia.1,2 Thus, decreasing rates of
tobacco initiation, as well as increasing smoking cessation rates, are imKey Factors Influencing Disparities at Each Point
portant steps for cancer prevention.
To further our understanding of toCancer Prevention
Cancer Diagnosis
Cancer Survivorship
bacco initiation and maintenance
among racial/ethnic minorities and
individuals of lower socioeconomic
Environmental
Multiple negative
status, it is important to consider enAfrican Americans
influences contribute
effects of continued
have highest lung
vironmental and social contextual into smoking, exposure
smoking on cancer
cancer incidence
fluences.9
to second-hand
outcomes 3,22
compared to all
smoke9-13
Environmental Influences
Tobacco-control policies, such as
smoking restrictions, can be effective at reducing tobacco initiation,
use, and related health care costs.2
Despite progress on reducing the
amount of exposure to second-hand
smoke via smoke-free restrictions
in public places, disparities in workplace policies can create or exacerbate tobacco-related disparities in
racial/ethnic minority and underserved populations.28 For example,
those who smoke and are employed
in blue-collar or service occupations
are more likely to work in environments that permit smoking.29 In adOctober 2016, Vol. 23, No. 4
Targeted tobacco
marketing creates
vulnerability14
Perceived stress/
discrimination leads to
more smoking15,16
Validated smoking
cessation options
lacking for minorities20
other racial and ethnic
groups21
Racial and ethnic
minorities more likely
to be diagnosed at
more advanced-stage
disease23
Potential unequal
implementation
of cancer screening
guidelines8,17-19
Low representation
of racially/ethnically
diverse patients
with cancer in
cessation trials26,27
Lack of education,
training, and
infrastructure for
providers to
implement smoking
cessation into
clinical care 4,24,25
Fig. — Tobacco-related health disparities exist across the cancer care continuum.
Cancer Control 435
and racial/ethnic minority populations is targeted tobacco marketing.2 The tobacco industry uses targeted
marketing and promotions to reach ethnic/racial minorities, sexual minorities, and low-income communities.2,14 Examples of these industry efforts include
a greater presence of tobacco billboards in specific
neighborhoods and the targeted marketing of menthol cigarettes.14 In a systematic review, the presence
and quantity of tobacco marketing at tobacco retailers
were examined by neighborhood characteristics such
as race, ethnicity, and socioeconomic status.14 The results suggested that lower-income neighborhoods are
exposed to more tobacco marketing, and the amount
of marketing of menthol products is higher in both urban neighborhoods and communities with larger numbers of African American residents.14 Among neighborhoods with a greater proportion of Hispanic residents,
no observed increase in tobacco marketing was seen
(possibly due to cultural reasons).2,14
Perceived Stress and Social Discrimination
Racial/ethnic minorities and low-income individuals
report higher levels of perceived stress, which is defined by the degree to which individuals perceive circumstances in their life as stressful.15,34 Elevated levels
of stress may can be attributed to several factors such
as lower wages, more daily hassles, and stressful life
events.15,35 Social discrimination is also a commonly
perceived stressor among African Americans and Latinos.34 Perceived levels of stress and social discrimination have been associated with cancer burden and tobacco smoking.2 The prevalence of tobacco smoking is
higher among individuals who perceive greater rates of
discrimination.16 For example, compared with nonminority individuals, lesbian, gay, bisexual, and transgender individuals are more likely to experience stressors
such as discrimination and stigma and have a higher
smoking prevalence rate (between 24% and 45%).36 In
addition, an inverse relationship has been observed
between perceived levels of stress and rates of smoking cessation.37
A primary mechanism through which stress may
influence smoking behavior is via negative-affect management.34 Although tobacco smoking may not actually
reduce stress (except for nicotine withdrawal-related
stress), if those who smoke believe that smoking reduces stress, then that belief will motivate smoking — ie,
smoking is often used as a coping strategy for dealing
with stress, and, as stress increases, so does smoking.38
The relationship between negative affect and smoking
has been well documented.39,40 The greater perceived
level of stress among racial/ethnic minorities and lowincome populations, as well as the robust relationship
between stress and smoking, remains an important factor to assess and consider in the development of targeted interventions for underserved persons who smoke.
436 Cancer Control
Secondary Prevention
To work toward eliminating tobacco-related cancer
disparities, we must add to our understanding of social and environmental factors that influence tobacco
use among underserved populations so that primary
prevention strategies to prevent smoking initiation
may be implemented. Equally important are secondary prevention strategies that seek to develop effective
smoking cessation interventions to help those who
have started smoking. Several evidence-based treatments exist for treating tobacco dependence. Clinical
practice guidelines from the US Public Health Service
summarize these treatments and describe a brief intervention model called the 5 As8,41: ask about tobacco
use, advise to quit smoking, assess interest in quitting
smoking, assist in quitting smoking via counseling
and pharmacotherapy, and arrange follow-up. These
recommendations apply to a broad population of persons who smoke and include those from all racial and
ethnic groups, individuals with low socioeconomic status, those with limited formal education, those who are
hospitalized, and those who identify as lesbian, gay,
bisexual, or transgender.41 Despite the applicability of
these guidelines for diverse populations, the unequal
implementation of these guidelines has been documented.8,17-19 Among those who smoke, Hispanics and
those with low socioeconomic status are less likely to
receive advice and assistance from their health care
professional about smoking cessation compared with
non-Hispanic whites and those with higher socioeconomic status, respectively.17-19 Access to evidence-based
smoking cessation methods is also lacking for individuals with limited or no English proficiency.42 The absence of readily available, Spanish-language, evidencebased materials has been cited as a key barrier to the
ability of primary care physicians to assist with smoking cessation.43
Targeted Smoking Cessation
The US Public Health Service guidelines for evidence-based smoking cessation interventions apply to
all racial and ethnic minorities.8 However, important
differences exist between racial/ethnic minorities and
whites that warrant research on the effectiveness of
smoking cessation interventions.44 The smoking prevalence rate for non-Hispanic whites (18.2%) is comparable with that seen in African Americans (17.5%) but
is lower for Hispanics (11.2%).45 However, smoking
prevalence varies by cultural subgroup; for example,
the smoking prevalence among individuals of Puerto
Rican and Cuban descent exceeds 30%.46 Moreover, racial and ethnic minorities are disproportionately represented in lower-income categories, where smoking
prevalence is highest.45 Several notable differences in
smoking behavior patterns also exist among different
groups. Although smoking prevalences among African
October 2016, Vol. 23, No. 4
Americans and non-Hispanic whites are comparable,
African Americans start smoking at a later age, are
more likely to smoke menthol cigarettes, and smoke
fewer cigarettes per day on average.3,47,48 Despite lower
daily rates of smoking among Hispanics and African
Americans compared with non-Hispanic whites, racial
and ethnic minorities who smoke have greater difficulty quitting smoking.49
Cultural factors and acculturation must be taken
into account, because those who are more acculturated
are more likely to smoke and have lower odds of sustaining cessation.46 Limited interventional research has
also been conducted with American Indians and Alaska
Natives, who have the highest rates of tobacco use — including both ceremonial and commercial tobacco use.5,45
Benowitz et al50,51 also reports that differences
may exist in the genetics and physiology (eg, how
fast nicotine is metabolized) among racial and ethnic
groups that warrant the evaluation of specific smoking cessation interventions among these groups, rather than assuming equivalent rates of efficacy across
groups. This concept is relevant given that most clinical trials for smoking cessation medications include
few minorities who smoke.50
Narrative and meta-analytic reviews of smoking
cessation interventions have been conducted for racial
and ethnic minorities who smoke.44,52-54 One comprehensive and narrative review included 64 studies involving African Americans, Latinos, American Indians,
Asians, and Pacific Islanders who smoked.44 Pharmacotherapy, behavioral interventions, and community
interventions for smoking cessation were examined.
The authors concluded that existing data support the
use of pharmacotherapy treatments for smoking cessation among African Americans, Latinos, and Hispanics who smoke.44 Specifically, nicotine patches,
nicotine nasal sprays, and bupropion were effective in
African Americans who smoke, whereas the nicotine
patch alone was effective for Hispanics who smoke.44
Nicotine gum has not demonstrated efficacy in 3 studies with African Americans who smoke, and trials have
not been conducted with nicotine lozenges, inhalers,
or combination nicotine-replacement therapy.44
Regardless of the outcomes of efficacy trials,
real-world effectiveness and use of pharmacotherapy
must be considered. Qualitative research conducted
by Fu et al55 with minorities who smoke (African Americans, American Indians, and Southeast Asians) revealed
that knowledge is lacking about how pharmacotherapy
works as well as the skepticism regarding the effectiveness of smoking cessation. These beliefs may explain
the low utilization of pharmacotherapy among racial
and ethnic minorities who smoke.56 Further research on
the beliefs and barriers of racial and ethnic differences
regarding pharmacotherapy may aid in the development of strategies to increase use of cessation methods.
October 2016, Vol. 23, No. 4
Tested modalities for behavioral interventions
have included both group and individual counseling.46,54 In a randomized trial, Webb et al57 found
that African Americans who smoked and received
group-based cognitive behavioral therapy to help
them quit smoking had higher cessation rates than
African Americans who smoked and were involved in
a general health education group that controlled for
contact and intensity. In addition to support for group
therapy, researchers concluded that individual counseling (in-person and on the telephone) was effective
for aiding smoking cessation among racial and ethnic
minorities.44
Some research has focused on the development
of culturally adapted smoking cessation interventions
that take into account unique cultural characteristics.44
However, little research has been conducted to evaluate the effectiveness of culturally adapted or culturally
specific interventions compared with generic interventions. Select studies conducted with African Americans who smoke have been mixed.20,58-62 One study
found evidence of a greater level of interest and attention for culturally specific materials, yet readiness to
quit and cessation attempts were greater for the standard materials.58 By contrast, a study conducted with
African Americans who smoke found greater rates of
smoking abstinence at a 12-month follow-up period
among participants who received culturally tailored
materials than those who received standard materials.59 Other studies have also demonstrated the benefits of culturally tailored interventions for Hispanics
who smoke.60,61 Overall, researchers have reported that
study participants are generally accepting of interventional materials that include information about cultural
adaptation, yet additional research is needed to determine whether such cultural tailoring is superior to
standard materials to improve smoking cessation rates
among minorities.20,58,62
Cancer Diagnosis
Higher rates of cancer morbidity and mortality are
evident among racial/ethnic minorities and those with
low socioeconomic status.21 Several pathways exist in
which the relationship between tobacco and cancer
health disparities can present. A higher risk of tobaccorelated cancers among persons who smoke and have
low incomes could, in part, be a function of higher
smoking prevalence rates. Although the overall prevalence of smoking has declined to 16.8%, low-income
individuals who smoke (ie, household income below
the poverty level) have a smoking prevalence rate of
30.4%.63 Other pathways are less clear. Referred to as
the “smoker’s paradox,” African Americans smoke fewer cigarettes per day and are less likely to smoke daily,
yet they have higher rates of lung, esophageal, and oral
cancers than non-Hispanic whites who smoke.64
Cancer Control 437
Disparities in Lung Cancer
Lung cancer is the leading cause of cancer-related
deaths in the United States, and 80% of these deaths
are attributable to cigarette smoking.2 African Americans have the highest lung cancer incidence and mortality rates compared with all other racial and ethnic
groups.3,21 Risk factors for poor cancer outcomes
have been attributed to several factors such as treatment access and lower socioeconomic status, as well
as smoking-related factors.22 The disproportionate
burden in lung cancer risk among African Americans
cannot be explained by a greater number of cigarettes smoked.64 However, a distinguishing smoking behavior among African Americans who smoke,
which may aid in explaining disparate disease outcomes, is that African Americans are more likely to
smoke menthol cigarettes than whites.3 As part of the
2009 Family Smoking Prevention and Tobacco Control Act, the Tobacco Products Scientific Advisory
Committee of the US Food and Drug Administration
was tasked with submitting a report on the impact
of menthol.65 Findings from that report suggest that
little evidence exists to demonstrate that menthol
contributes to greater disease risk; however, menthol
cigarettes may have more addictive qualities than
nonmenthol cigarettes, which could contribute to
low rates of smoking cessation.3,65 Menthol also has
cooling and anesthetic effects that reduce the harshness of cigarette smoke.65 Use of menthol cigarettes
is also associated with increased initiation and progression to cigarette smoking and difficulty quitting
smoking.65 Based on these findings, the Tobacco
Products Scientific Advisory Committee concluded
that no longer selling menthol cigarettes would be
of a public health benefit.65 However, more research
is needed to further examine the potential role that
menthol plays in the unequal disease burden experienced by African Americans.3 Specifically, as noted
by Alexander et al,64 research is needed on examining additional pathways such as the role of taste sensitivity and the effects of menthol on cellular mechanisms and subsequent disease processes.
Role of Beliefs About Lung Cancer
Research suggests that beliefs about lung cancer may
differ by race/ethnicity. Compared with whites, African
Americans have lower perceived notions of risk
for lung cancer and are more likely to doubt the relationship between lung cancer and behavior.66,67 African Americans also have demonstrated greater skepticism regarding surgical treatment for lung cancer.68,69
Moreover, the belief that exposure to air during surgery can cause the tumor to spread and fatalistic beliefs (eg, “If I have lung cancer, I believe it was meant to
be”) may serve as barriers to accepting surgical intervention among racial and ethnic minorities.68,69 These
438 Cancer Control
findings underscore the need to understand perceptions of lung cancer by race/ethnicity to identify potential barriers to smoking cessation and lung cancer
treatment.
Challenges in Screening
Based on evidence from the National Lung Cancer
Screening Trial, low-dose computed tomography for
lung cancer screening provides an opportunity to reduce the rate of lung cancer mortality.70 A concern
raised by Holford et al71 is that the 30 pack-years’ eligibility criteria required by the US Preventive Services Task Force and Centers for Medicare & Medicaid
Services may result in fewer screenings for African
American men who smoke and have fewer pack years.
African Americans have also been found to be more reluctant to get screened for lung cancer due to fear of
the disease.66 Thus, it is possible that unequal implementation of newer screening modalities and heightened fears of disease may broaden cancer disparities.
Head and Neck Cancers
Tobacco use is a major risk factor for the development of head and neck cancers: 75% of all head and
neck cancers are attributable to tobacco and alcohol
use.72 Molina et al23 examined survival rates of head
and neck cancers based on race/ethnicity and socioeconomic status. Their findings suggested that,
compared with non-Hispanic whites, African Americans were diagnosed with head and neck cancers at
a younger age and at more advanced-disease stages.23 In addition, the average survival time for African American patients was significantly lower than
whites (21 vs 40 months).23 Poor survival outcomes in
African Americans were observed despite the lack of
difference in the frequency of smoking. Living in an
area of high poverty was found to be an independent
predictor of worse outcomes.73 Poor cancer outcomes
among patients from impoverished communities has
also been attributed to lack of access to health care,
decreased likelihood of insurance coverage, and advanced disease at presentation.73 Among Hispanics and
Latinos, incidence and mortality rates of head and neck
cancers are lower than both African Americans and
non-Hispanic whites.74
Cancer Survivorship
Once an individual who smokes has been diagnosed
with cancer, he or she may mistakenly believe that it
is too late to quit smoking; however, a growing body
of research suggests that patients with cancer who
continue to smoke are at risk for several adverse
health outcomes.1 The 2014 report of the US Surgeon
General concluded that sufficient evidence has accumulated to infer a causal relationship between individuals with cancer who smoke cigarettes and cancer
October 2016, Vol. 23, No. 4
survivors for both all-cause mortality and cancer-specific mortality.1 Evidence also suggests that continued smoking results in an increased risk for second
primary cancers known to be caused by cigarette
smoking, and, although this evidence is insufficient
to infer a causal relationship, an association does exist between cigarette smoking and risk of cancer recurrence, poor response to treatment, and increased
rates of cancer treatment-related toxicity.1 Because
racial and ethnic minorities are more likely to be diagnosed with late-stage tobacco-related cancers, it
is possible that they could be particularly vulnerable
to the adverse effects of continued smoking. Park
et al22 reported that smoking cessation represents a
modifiable risk factor that could result in better treatment outcomes in African Americans with lung cancer
by improving rates of treatment efficacy and quality of
life. Given the strong associations between continued
smoking and cancer outcomes, organizations such as
the American Society of Clinical Oncology and American Association for Cancer Research have called for
smoking cessation assistance to be provided to cancer
survivors.75 The National Comprehensive Cancer Network also established practice guidelines for smoking
cessation that call for all patients with cancer to receive
evidence-based smoking cessation interventions.76
Implementation of the 5As model for a brief cessation intervention has been suboptimal in the oncology
setting.8,24,25,77 Although the “ask” and “advise” steps
are often implemented, the remaining 3 steps are not
routinely completed.8,24 A review of members of the
American Society of Clinical Oncology reported that
44% of its members surveyed said they discuss medication options for smoking cessation, and 39% said
they provided smoking cessation treatment.24 These
findings coincide with patient reports of low levels
of cessation assistance provided by oncology specialists.77 Because racial and ethnic minorities who smoke
are generally less likely to receive assistance from their
physicians in quitting smoking, it is plausible that this
assistance may also not occur in the oncology setting.18,19 Perceptions of inadequate training in tobacco
cessation interventions and low rates of self-efficacy
in helping patients to quit have been cited barriers to
cessation support by health care professionals.24 These
findings suggest a need for improved education and
training of health care professionals to better convey
the rationale for smoking cessation and to develop
strategies to implement smoking cessation into clinical
care. In the absence of health care professional training, and taking into consideration the constraint of time,
an alternative ask–advise–refer model may be more feasible to implement.25 This model involves asking about
tobacco use, advising the patient to quit, and then referring the patient to existing, evidence-based resources
(eg, national tobacco quit line [800-QUIT-NOW]).
October 2016, Vol. 23, No. 4
Despite the importance of quitting smoking for
individuals with cancer, few clinical trials of smoking
cessation have been conducted in this population. In
a systematic review and meta-analysis, 10 randomized
controlled trials and 3 prospective cohort studies were
evaluated.26 Studies included a range of nonpharmacological (eg, counseling, self-help) and pharmacological (eg, nicotine-replacement therapy) interventions.26
Overall, findings indicated that tobacco cessation interventions in the oncology setting were not significantly
different from usual care, suggesting the need for additional research to enhance smoking cessation among
patients with cancer.26 Although a single smoking cessation trial demonstrated feasibility of recruiting African
American patients, most smoking cessation trials conducted in the oncology setting have had low representation of racially and ethnically diverse patients.26,27 To
our knowledge, cessation interventions are not available
and have not been tested in Spanish-speaking patients.
Thus, as new interventions are developed and tested in
the oncology setting, the inclusion of racial and ethnic
minorities is imperative to extend the generalizability of
findings and reduce tobacco-related cancer disparities.
Conclusions
Racial and ethnic minorities experience shared social
and environmental contextual factors that impact the
initiation and maintenance of smoking, and the potential for tobacco-related health disparities exist at each
point of the oncology care continuum. Thus, a comprehensive strategy for reducing health disparities must
call for future research efforts at each of these points.
Health disparities exist in the incidence
and mortality of the 2 most strongly associated tobacco-related cancers, namely lung and head and neck
cancers.2,3,21,23,72-74 Future research is needed to elucidate the causes of these disparities. As new cancer
screening tools become available, attention must be
paid to prevent the widening of health disparities due
to unequal implementation.
Overall rates of smoking cessation tend to be lower among racial and ethnic minorities.46,49 Culturally
adapted interventions are more acceptable44,46; however, research has not yet supported their effectiveness
when compared with standard materials.55,61 Future research efforts should be directed at increasing the use
of treatment and greater rates of efficacy in these vulnerable populations. Individuals with cancer must also
be provided with cessation assistance and interventions
aimed at helping them quit smoking to improve cancer
outcomes.75,76 Additional education and training are
needed for oncologists so they can begin to understand
the impact of continued smoking for patients as well as
to increase their comfort level for providing smoking
cessation treatment to their patients who need it.25 Due
to the disproportionate disease burden experienced by
Cancer Control 439
African Americans and other underserved populations
who smoke, future trials on smoking cessation interventions must include diverse racial and ethnic patient
populations and those cessation tools should be validated for use in all vulnerable patient populations.3,26,27
References
1. US Department of Health and Human Services. The Health Consequences of Smoking: 50 Years Of Progress. A Report of the Surgeon General. Atlanta, GA: US 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.
2. American Cancer Society. Cancer Facts & Figures 2016. Atlanta, GA:
American Cancer Society. http://www.cancer.org/acs/groups/content/@
research/documents/document/acspc-047079.pdf. Accessed August 19, 2016.
3. American Cancer Society. Cancer Facts & Figures for African Americans
2016-2018. Atlanta, GA: American Cancer Society. http://www.cancer.org/
acs/groups/content/@editorial/documents/document/acspc-047403.pdf.
Accessed August 19, 2016.
4. Fagan P, King G, Lawrence D, et al. Eliminating tobacco-related
health disparities: directions for future research. Am J Public Health.
2004;94(2):211-217.
5. US Surgeon General. Tobacco use among U.S. racial/ethnic minority groups--African Americans, American Indians and Alaska Natives,
Asian Americans and Pacific Islanders, Hispanics. Executive summary.
MMWR Recomm Rep. 1998;47(RR-18):v-xv, 1-16.
6. Okuyemi KS, Reitzel LR, Fagan P. Interventions to reduce tobaccorelated health disparities. Nicotine Tob Res. 2015;17(8):887-891.
7. Garrett BE, Dube SR, Winder C, et al. Cigarette smoking — United
States, 2006-2008 and 2009-2010. MMWR Suppl. 2013;62(3):81-84.
8. Fiore MC, Jaén CR, Baker TB, et al. Clinical Practice Guideline:
Treating Tobacco Use and Dependence: 2008 Update. Rockville, MD:
US Department of Health and Human Services; Public Health Service; 2008.
http://bphc.hrsa.gov/buckets/treatingtobacco.pdf. Accessed August 19, 2016.
9. Garrett BE, Dube SR, Babb S, et al. Addressing the social determinants of health to reduce tobacco-related disparities. Nicotine Tob Res.
2015;17(8):892-897.
10. Honjo K, Tsutsumi A, Kawachi I, et al. What accounts for the relationship between social class and smoking cessation? Results of a path
analysis. Soc Sci Med. 2006;62(2):317-328.
11. Muilenburg Legge J, Latham T, Annang L, et al. The home smoking environment: influence on behaviors and attitudes in a racially diverse
adolescent population. Health Educ Behav. 2009;36(4):777-793.
12. Norman GJ, Ribisl KM, Howard-Pitney B, et al. The relationship
between home smoking bans and exposure to state tobacco control efforts
and smoking behaviors. Am J Health Promot. 2000;15(2):81-88.
13. Mills AL, White MM, Pierce JP, et al. Home smoking bans among
U.S. households with children and smokers. Opportunities for intervention.
Am J Prev Med. 2011;41(6):559-565.
14.Lee JG, Henriksen L, Rose SW, et al. A systematic review of
neighborhood disparities in point-of-sale tobacco marketing. Am J Public
Health. 2015;105(9):e8-e18.
15. Webb Hooper M, Kolar SK. Distress, race/ethnicity and smoking cessation in treatment-seekers: implications for disparity elimination. Addiction.
2015;110(9):1495-1504.
16. Purnell JQ, Peppone LJ, Alcaraz K, et al. Perceived discrimination, psychological distress, and current smoking status: results from the
Behavioral Risk Factor Surveillance System Reactions to Race module,
2004-2008. Am J Public Health. 2012;102(5):844-851.
17. Kruger J, Shaw L, Kahende J, et al. Health care providers’ advice
to quit smoking, National Health Interview Survey, 2000, 2005, and 2010.
Prev Chronic Dis. 2012;9:E130.
18. Lopez-Quintero C, Crum RM, Neumark YD. Racial/ethnic disparities
in report of physician-provided smoking cessation advice: analysis of the 2000
National Health Interview Survey. Am J Public Health. 2006;96(12):2235-2239.
19. Sonnenfeld N, Schappert SM, Lin SX. Racial and ethnic differences
in delivery of tobacco-cessation services. Am J Prev Med. 2009;36(1):21-28.
20. Simmons VN, Quinn G, Litvin EB, et al. Transcreation of validated
smoking relapse-prevention booklets for use with Hispanic populations.
J Health Care Poor Underserved. 2011;22(3):886-893.
21. Howlader N, Noone AM, Krapcho M, et al, eds. SEER cancer
statistics review, 1975-2012. http://seer.cancer.gov/csr/1975_2012.
Based on November 2014 SEER data submission, posted April 2015.
Accessed August 3, 2016.
22.Park ER, Japuntich SJ, Traeger L, et al. Disparities between
blacks and whites in tobacco and lung cancer treatment. Oncologist.
2011;16(10):1428-1434.
23. Molina MA, Cheung MC, Perez EA, et al. African American and
poor patients have a dramatically worse prognosis for head and neck cancer: an examination of 20,915 patients. Cancer. 2008;113(10):2797-2806.
440 Cancer Control
24. Warren GW, Marshall JR, Cummings KM, et al. Addressing tobacco
use in patients with cancer: a survey of American Society of Clinical Oncology members. J Oncol Pract. 2013;9(5):258-262.
25. Schroeder SA. What to do with a patient who smokes. JAMA.
2005;294(4):482-487.
26. Nayan S, Gupta MK, Strychowsky JE, et al. Smoking cessation
interventions and cessation rates in the oncology population: an updated systematic review and meta-analysis. Otolaryngol Head Neck Surg.
2013;149(2):200-211.
27. Martinez E, Tatum KL, Weber DM, et al. Issues related to implementing a smoking cessation clinical trial for cancer patients. Cancer
Causes Control. 2009;20(1):97-104.
28. Moolchan ET, Fagan P, Fernander AF, et al. Addressing tobaccorelated health disparities. Addiction. 2007;102(suppl 2):30-42.
29.Aakko E, Schafer E, Gyarmathy VA, et al. Smoking policies
in manufacturing and assembly workplaces, Wisconsin, 1999. WMJ.
2001;100(3):67-69.
30. Glasgow RE, Cummings KM, Hyland A; Community Intervention
Trial for Smoking Cessation. Relationship of worksite smoking policy to
changes in employee tobacco use: findings from COMMIT. Tob Control.
1997;6(suppl 2):S44-S48.
31. Pizacani BA, Martin DP, Stark MJ, et al. A prospective study of
household smoking bans and subsequent cessation related behaviour:
the role of stage of change. Tob Control. 2004;13(1):23-28.
32. Kraev TA, Adamkiewicz G, Hammond SK, et al. Indoor concentrations
of nicotine in low-income, multi-unit housing: associations with smoking
behaviours and housing characteristics. Tob Control. 2009;18(6):438-444.
33. Andrews JO, Mueller M, Newman SD, et al. The association of individual and neighborhood social cohesion, stressors, and crime on smoking status among African-American women in southeastern US subsidized
housing neighborhoods. J Urban Health. 2014;91(6):1158-1174.
34. Kendzor DE, Businelle MS, Reitzel LR, et al. The influence of discrimination on smoking cessation among Latinos. Drug Alcohol Depend.
2014;136:143-148.
35. Skinner ML, Shirtcliff EA, Haggerty KP, et al. Allostasis model
facilitates understanding race differences in the diurnal cortisol rhythm.
Dev Psychopathol. 2011;23(4):1167-1186.
36. Grady ES, Humfleet GL, Delucchi KL, et al. Smoking cessation
outcomes among sexual and gender minority and nonminority smokers in
extended smoking treatments. Nicotine Tob Res. 2014;16(9):1207-1215.
37. Berg CJ, Thomas JL, Guo H, et al. Predictors of smoking reduction
among Blacks. Nicotine Tob Res. 2010;12(4):423-431.
38. Landrine H, Klonoff EA. Racial segregation and cigarette smoking among blacks: findings at the individual level. Journal Health Psychol.
2000;5(2):211-219.
39. Brandon TH. Negative affect as motivation to smoke. Curr Dir Psychol
Sci. 1994;3(2):33-37.
40. Kassel JD, Stroud LR, Paronis CA. Smoking, stress, and negative
affect: correlation, causation, and context across stages of smoking. Psychol
Bull. 2003;129(2):270-304.
41. Fiore MC, Jaén CR. A clinical blueprint to accelerate the elimination
of tobacco use. JAMA. 2008;299(17):2083-2085.
42. Wetter DW, Mazas C, Daza P, et al. Reaching and treating Spanishspeaking smokers through the National Cancer Institute’s Cancer Information
Service. A randomized controlled trial. Cancer. 2007;109(2 suppl):406-413.
43. Blumenthal DS. Barriers to the provision of smoking cessation services reported by clinicians in underserved communities. J Am Board Fam
Med. 2007;20(3):272-279.
44. Cox LS, Okuyemi K, Choi WS, et al. A review of tobacco use
treatments in U.S. ethnic minority populations. Am J Health Promot.
2011;25(5 suppl):S11-S30.
45. Jamal A, Homa DM, O’Connor E, et al. Current cigarette smoking
among adults - United States, 2005–2014. MMWR Morb Mortal Wkly Rep.
2015;64(44):1233-1240
46. Merzel CR, Isasi CR, Strizich G, et al. Smoking cessation among
U.S. Hispanic/Latino adults: findings from the Hispanic Community Health
Study/Study of Latinos (HCHS/SOL). Prev Med. 2015;81:412-419.
47. Haiman CA, Stram DO, Wilkens LR, et al. Ethnic and racial differences in the smoking-related risk of lung cancer. N Engl J Med.
2006;354(4):333-342.
48. Trinidad DR, Gilpin EA, Lee L, et al. Do the majority of AsianAmerican and African-American smokers start as adults? Am J Prev Med.
2004;26(2):156-158.
49. Irvin Vidrine J, Reitzel LR, Wetter DW. The role of tobacco in cancer
health disparities. Curr Oncol Rep. 2009;11(6):475-481.
50. Benowitz NL. Smoking cessation trials targeted to racial and economic minority groups. JAMA. 2002;288(4):497-499.
51. Benowitz NL, Perez-Stable EJ, Herrera B, et al. Slower metabolism
and reduced intake of nicotine from cigarette smoking in Chinese-Americans. J Natl Cancer Inst. 2002;94(2):108-115.
52. Lawrence D, Graber JE, Mills SL, et al. Smoking cessation interventions in U.S. racial/ethnic minority populations: an assessment of the
literature. Prev Med. 2003;36(2):204-216.
53. Robles GI, Singh-Franco D, Ghin HL. A review of the efficacy of
October 2016, Vol. 23, No. 4
smoking-cessation pharmacotherapies in nonwhite populations. Clin Ther.
2008;30(5):800-812.
54. Webb MS. Treating tobacco dependence among African Americans:
a meta-analytic review. Health Psychol. 2008;27(3 suppl):S271-S282.
55. Fu SS, Burgess D, van Ryn M, et al. Views on smoking cessation
methods in ethnic minority communities: a qualitative investigation. Prev
Med. 2007;44(3):235-240.
56. Levinson AH, Perez-Stable EJ, Espinoza P, et al. Latinos report
less use of pharmaceutical aids when trying to quit smoking. Am J Prev
Med. 2004;26(2):105-111.
57. Webb MS, de Ybarra DR, Baker EA, et al. Cognitive-behavioral
therapy to promote smoking cessation among African American smokers:
a randomized clinical trial. J Consult Clin Psychol. 2010;78(1):24-33.
58. Webb MS. Culturally specific interventions for African American
smokers: an efficacy experiment. J Natl Med Assoc. 2009;101(9):927-935.
59. Orleans CT, Boyd NR, Bingler R, et al. A self-help intervention for
African American smokers: tailoring cancer information service counseling
for a special population. Prev Med. 1998;27(5 pt 2):S61-S70.
60. Nevid JS, Javier RA. Preliminary investigation of a culturally specific smoking cessation intervention for Hispanic smokers. Am J Health
Promot. 1997;11(3):198-207.
61. Rodriguez Esquivel D, Webb Hooper M, Baker EA, et al. Culturally
specific versus standard smoking cessation messages targeting Hispanics: an experiment. Psychol Addict Behav. 2015;29(2):283-289.
62. Simmons VN, Cruz LM, Brandon TH, et al. Translation and adaptation of smoking relapse-prevention materials for pregnant and postpartum
Hispanic women. J Health Commun. 2011;16(1):90-107.
63. Jamal A, Homa DM, O’Connor E, et al. Current cigarette smoking
among adults - United States, 2005-2014. MMWR Morb Mortal Wkly Rep.
2015;64(44):1233-1240.
64. Alexander LA, Trinidad DR, Sakuma KL, et al. Why we must continue
to investigate menthol’s role in the African American smoking paradox.
Nicotine Ton Res. 2016;18(suppl 1):S91-S101.
65. Tobacco Products Scientific Advisory Committee. Menthol Cigarettes
and Public Health: Review of the Scientific Evidence and Recommendations. Bethesda, MD: US Food and Drug Administration; 2011. http://www.
fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/
TobaccoProductsScientificAdvisoryCommittee/UCM269697.pdf. Accessed
August 3, 2016.
66. Lathan CS, Okechukwu C, Drake BF, et al. Racial differences in the
perception of lung cancer: the 2005 Health Information National Trends
Survey. Cancer. 2010;116(8):1981-1986.
67. Park ER, Ostroff JS, Rakowski W, et al. Risk perceptions among
participants undergoing lung cancer screening: baseline results from the
National Lung Screening Trial. Ann Behav Med. 2009;37(3):268-279.
68. George M, Margolis ML. Race and lung cancer surgery--a qualitative analysis of relevant beliefs and management preferences. Oncol Nurs
Forum. 2010;37(6):740-748.
69.Jonnalagadda S, Bergamo C, Lin JJ, et al. Beliefs and attitudes about lung cancer screening among smokers. Lung Cancer.
2012;77(3):526-531.
70. 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.
71. Holford TR, Levy DT, Meza R. Comparison of smoking history patterns among African American and white cohorts in the United States born
1890 to 1990. Nicotine Tob Res. 2016;18(suppl 1):S16-S29.
72. Blot WJ, McLaughlin JK, Winn DM, et al. Smoking and drinking in
relation to oral and pharyngeal cancer. Cancer Res. 1988;48(11):3282-3287.
73. Conway DI, Petticrew M, Marlborough, et al. Socioeconomic inequalities and oral cancer risk: a systematic review and meta-analysis of
case-control studies. Int J Cancer. 2008;122(12):2811-2819.
74. Fagan P, Moolchan ET, Lawrence D, et al. Identifying health disparities across the tobacco continuum. Addiction. 2007;102(suppl 2):5-29.
75. 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(8):1941-1948.
76. National Comprehensive Cancer Network. NCCN Clinical Practice
Guidelines in Oncology: Smoking Cessation. v.2.2015. https://www.nccn.org/
professionals/physician_gls/pdf/smoking.pdf. Accessed August 4, 2016.
77. Simmons VN, Litvin EB, Unrod M, et al. Oncology healthcare providers’ implementation of the 5A’s model of brief intervention for smoking
cessation: patients’ perceptions. Patient Educ Couns. 2012;86(3):414-419.
October 2016, Vol. 23, No. 4
Cancer Control 441
Case Series
Human Metapneumovirus Infection in Immunocompromised Patients
Sharmeen Samuel, MD, Sowmya Nanjappa, MBBS, MD, Christopher D. Cooper, MD, and John N. Greene, MD
Summary: Human metapneumovirus (HMPV) is a pathogen associated with respiratory tract infection and
is related to avian pneumovirus. Typically, children, the elderly, and those who are immunocompromised are
the most susceptible to HMPV infection; however, the virus can infect persons of all ages. In otherwise healthy
individuals, HMPV infection is generally self-limiting, but immunocompromised individuals can develop fatal
complications. We present a case series of 3 severely immunocompromised patients who were infected with
HMPV and describe their clinical course. All 3 patients had acute myeloid leukemia, histories of neutropenic
fever, and prolonged hospitalization stays. This case series highlights the severe sequelae observed in individuals infected with HMPV, particularly among those who are immunocompromised.
Introduction
In 2001, van den Hoogen et al1 identified human
metapneumovirus (HMPV) in children with respiratory tract infection, because they were able to obtain
a genomic sequence of the pathogen via randomly
primed polymerase chain reaction (PCR). The virus was
found to be related to avian pneumovirus, a member
of the Metapneumovirus genus.2 HMPV is an emerging
pathogen associated with both upper and lower respiratory tract infections in all age groups, but it is more
common in children (age < 5 years), in the elderly
(age > 60 years), and in those with compromised immune systems.3,4 Although its formal discovery occurred
15 years ago, HMPV may have been responsible for
respiratory tract infections for at least 60 years. 1
Case Reports
This series presents the cases of 3 immunocompromised patients treated at the H. Lee Moffitt Cancer
Center & Research Institute (Tampa, FL) who were infected with HMPV. A summary of the case series is
found in the Table.
Case 1
A Hispanic woman 47 years of age with acute myeloid
leukemia (AML) had a clinical course complicated by
central nervous system malignancy, bacterial enteritis,
and diffuse alveolar and left-sided retinal hemorrhages. She was taking cladribine, cytarabine, and growthFrom the Departments of Internal Medicine (SS, SN) and Infectious
Diseases (JNG), H. Lee Moffitt Cancer Center & Research Institute,
Tampa, Florida, the Department of Infectious Diseases (CDC), Florida
Hospital, Altamonte Springs, Florida.
Submitted February 17, 2016; accepted May 31, 2016.
Address correspondence to John N. Greene, MD, Department of
Infectious Diseases, Moffitt Cancer Center, 12902 Magnolia Drive,
FOB-3 BMT PROG, Tampa, FL 33612. E-mail: [email protected]
No significant relationships exist between the authors and the companies/organizations whose products or services may be referenced
in this article.
442 Cancer Control
factor therapy for AML and received multiple doses of
intrathecal methotrexate.
Three months later she was readmitted to the hospital for blast crisis and reinduction chemotherapy. During
that admission, she developed bacteremia with vancomycin-resistant enterococcus and Streptococcus mitis.
She was subsequently treated with daptomycin, liposomal amphotericin B, cefepime, and acyclovir.
One month later she developed intermittent hemoptysis and mild dyspnea on ambulation that progressed to
hypoxia (oxygen saturation = 90% on room air). She also
experienced confusion, fever (temperature = 105.5 °F),
bilateral scattered rhonchi, and crackles on auscultation.
Noncontrast computed tomography (CT) of the thorax
was obtained and revealed new focal consolidative infiltrates, including increased peripheral, dense consolidation of the right middle, left upper, and left lower lobes,
widespread ill-defined nodules, and ground-glass infiltrates due to alveolar hemorrhage (Fig).
Bronchoalveolar fluid cultures obtained by bronchoscopy were negative for bacteria and fungi. However,
HMPV was detected by PCR from nasopharynx swab
sampling. Findings on PCR testing of other viral agents
(adenovirus, influenza, parainfluenza, and rhinoviruses)
were negative.
The patient was treated with oral ribavirin (RBV)
600 mg 3 times a day. After 14 days of therapy, her condition clinically improved and oxygen therapy was
discontinued. Repeat CT showed improved multifocal
pneumonia compared with the findings on the initial
imaging study.
Case 2
An African American man 61 years of age with hepatitis B virus infection and non-Hodgkin lymphoma presented with fever and chills. Laboratory work-up studies were ordered and the results revealed pancytopenia.
Findings on bone marrow biopsy were conclusive of
AML. He was treated with cladribine, cytarabine, and
October 2016, Vol. 23, No. 4
Table. — Summary of the Case Series
Case No.
Underlying
Malignancy
Etiology of Immunocompromised
State
Clinical Signs and
Symptoms
Treatment
Outcome
1
AML
CNS
Chemotherapy
Dyspnea
Fever
Hemoptysis
Oral RBV 600 mg
TID
Survived
2
AML
NHL
Chemotherapy
Cough
Dyspnea
Fever
Oral RBV 600 mg
TID
Died
3
AML
Chemotherapy
Cough
Dyspnea
Fever
Oral RBV 600 mg
TID
Died
AML = acute myeloid leukemia, CNS = central nervous system, NHL = non-Hodgkin lymphoma, RBV = ribavirin, TID = 3 times a day.
growth factors and eventually achieved remission.
Several months later, bone marrow biopsy was repeated, the results of which showed that the AML had
relapsed; thus, cycle 1 of azacitidine therapy was initiated. The treatment course was complicated by pneumonia (presumed to be fungal in origin based on findings seen on CT of his chest). The patient was given
daily liposomal amphotericin B to treat pneumonia.
On his hospital readmission for the second cycle of
azacitidine, the patient developed submandibular cellulitis, transaminitis, and became infected with type 1 herpes simplex virus and respiratory syncytial virus. He
was treated with intravenous meropenem every day
for 6 weeks, weekly liposomal amphotericin B, acyclovir, and lamivudine. However, his clinical course was
complicated by neutropenic fever and worsening dyspnea. Blood cultures grew Enterococcus faecium and
multi–drug-resistant S maltophilia. Samples on bronchoscopy were positive for S maltophilia and Candida
glabrata. He was treated with intravenous daptomycin
and minocycline. Blood cultures were repeated and
the results were negative after 6 days.
However, the patient again developed fever (temperature = 102.5 °F), dry cough, and hypoxia (oxygen saturation = 92% on room air). Respiratory viral panel was repeated and HMPV was detected in his nasopharynx swab
specimen. He was treated with oral RBV 600 mg 3 times
a day. The patient died 4 days after the start of therapy.
Case 3
A Hispanic man 54 years of age with relapsed AML
was admitted to the hospital for induction chemotherapy and was treated with cladribine, cytarabine, and
growth-factor therapy. One week later while on neutropenic prophylaxis (ciprofloxacin, voriconazole, and
acyclovir), the patient developed cough, mild dyspnea
(oxygen saturation = 94% on room air), and fever (temperature = 103.0 °F). Although findings on CT of the
chest revealed bilateral patchy and nodular opacities
consistent with an infectious process, sputum and bronOctober 2016, Vol. 23, No. 4
Fig. — Computed tomography of the chest from the patient in case 1.
Findings show focal consolidative infiltrates and dense peripheral consolidation in the left upper and lower lobes.
choalveolar fluid cultures were negative for bacteria and
fungi. However, findings on respiratory viral panel from
nasopharynx sampling were positive for HMPV.
The patient was treated with oral RBV 600 mg
3 times a day. Repeat CT of his chest showed extensive
bilateral ground-glass nodules consistent with bronchiolitis obliterans organizing pneumonia.
His clinical presentation initially improved after oral
RBV therapy, but he presented again with high-grade
fever, tachycardia, and hypotension. He was started on
meropenem, tobramycin, voriconazole, and acyclovir.
Blood cultures were obtained and grew Escherichia
coli. Despite aggressive treatment with antibiotics, the
patient died the following day from septic shock.
Discussion
HMPV is an enveloped virus with a nonsegmented,
negative-sense RNA genome.1 Genetic analysis has elucidated 2 subgroups of the virus that often circulate together in a population.5 Although the virus is related
to avian pneumovirus, HMPV does not cause illness
in birds.4,6 Study results have shown that the inoculaCancer Control 443
tion of HMPV in other nonhuman primates can lead to
significant viral replication restricted to the respiratory
tract with the subsequent development of upper respiratory tract symptoms.7 HMPV is commonly associated
with respiratory tract infection, causing flulike symptoms, bronchiolitis, and pneumonia. HMPV infection
may also be associated with exacerbations of asthma.8
The data available are limited on the association of
HMPV with infections of other organ systems beyond
the respiratory tract. In a case report concerning a previously healthy child 14 months of age who died of encephalitis, HMPV RNA was detected in both brain and
lung tissue, although HMPV antigens went undetected
in these tissues via immunostaining.9
Several studies have demonstrated the presence
of HMPV and absence of other pathogenic organisms
among patients with respiratory disease.8,10 Williams
et al11 examined the nasal-wash specimens from children with lower respiratory tract infections during a
25-year period. A viral cause other than HMPV was
identified in 41% of health care visits among 2,009 otherwise healthy infants and children presenting with
acute symptoms.11 No viral or bacterial cause was identified in 59% of visits; of the available specimens for
which no cause was previously identified, 20% tested
positive for HMPV.11 This finding extrapolates to 12% of
lower respiratory tract infections in their patient population. Thus, Williams et al11 concluded that HMPV infection is an important cause of respiratory tract illness,
particularly during the first few years of life.
In general, transmission of HMPV is through direct or close contact via large-particle aerosols, droplets, and fomites.12 Many nosocomial infections have
been reported in children and adults.13 Case 2 most
likely represents a nosocomial infection, because the
patient in this case was hospitalized for a prolonged
duration prior to developing symptoms. HMPV infection also causes a variable array of clinical symptoms,
including self-limiting cough, low-grade fever, rhinorrhea, severe respiratory failure, and death.14 The incubation period has not yet been defined but is thought
to be 5 to 6 days in most cases.15 Little is known about
the pathogenesis of HMPV, but an integrin alpha v beta
1 receptor has been identified that facilitates infection
of the epithelial cells of the respiratory tract.16
The most common reasons for hospitalization in
children are bronchiolitis and pneumonia. Although
HMPV has been associated with upper respiratory
tract infections (rhinopharyngitis, laryngitis, croup),
severe lower respiratory tract infection has been reported.8 The infection can occur in adults of all ages,
although elderly persons with histories of long hospitalization stays and cardiopulmonary disease are at
greater risk for becoming infected.5 The possible role
of HMPV in the exacerbation of chronic obstructive
pulmonary disease in adults is controversial.17
444 Cancer Control
Respiratory tract infections are a significant source
of morbidity and mortality in immunocompromised
patients, and this is especially true for individuals with
hematological malignancies and in those receiving
hematopoietic stem cell transplantation (HSCT).18 Prospective and retrospective studies of patients receiving
HSCT have provided data on the incidence of upper
and lower respiratory tract infections caused by HMPV
infection and the associated mortality rates.19,20 In these
studies, cohorts of symptomatic and asymptomatic
patients with HSCT were followed for 4 years, and the
virus was identified through PCR testing of nasopharyngeal specimens.19,20 The incidence rates of HMPV
infections in patients receiving HSCT ranged from 3%
to 7% in those with any respiratory disease, whereas
the rate increased to between 27% and 41% among
patients with lower respiratory disease.19,20 The rate of
deaths associated with lower respiratory tract infection
was between 33% and 40%, and the rate of deaths related to overall infections (upper and lower respiratory
tracts) was between 0% and 14%.19 In the retrospective
study, bronchoalveolar lavage specimens were obtained for the work-up of pulmonary infiltrates from
163 patients after receiving HSCT; of these, 3% were
positive for HMPV and 20% of patients died due to respiratory failure.20 These results highlight the potential
severity of HMPV-related pneumonia in recipients of
HSCT. Because many such patients have concurrent
bacterial and fungal infections, describing the exact
clinical features of HMPV infection can be difficult.
HMPV infection has important associations with
respiratory tract illness in patients with hematological malignancies and in those who have received
lung transplants.21,22 A retrospective evaluation of
55 children (age range, 5 months–19 years) with laboratory-confirmed HMPV infection highlighted a number
of associations. HMPV infection was associated with
underlying hematological malignancy (44%), HSCT
(15%), solid tumors (16%), and solid organ transplants
(15%).21 Another report showed that 25% of study patients with hematological disease who tested positive
for HMPV infection also had pneumonia.22 A review
by Kamboj et al23 of 1,899 study patients with cancer
showed that nearly 3% were positive for HMPV; more
children were positive than adults.
To diagnose HMPV infection, reverse transcriptase (RT)-PCR is the most sensitive and commonly used
method available; it detects the virus in nasal and bronchial secretions and is easy to perform.24 Other less-commonly used methods for detecting the virus include direct fluorescent antibody test, serology, and viral culture.
The incidence of HMPV infection increases during
the winter and spring.25 Patients in cases 1 and 2 in our
series were infected with HMPV in June 2015, and the
patient in case 3 was infected in May 2014. Prior infection with HMPV does not protect against reinfection,
October 2016, Vol. 23, No. 4
which can occur throughout life.4
A live recombinant vaccine for HMPV is still in preclinical trials.26 However, we must learn more about its
pathogenesis, genetics, and serological properties to
achieve a safe and effective vaccine against HMPV.
Treatment for HMPV infection is mainly supportive, and no data exist on any antiviral agent shown to
have significant activity against the virus. No guidelines are available for treatment, but some centers use
RBV and intravenous immunoglobulin; however, the
usefulness of RBV in HMPV infection is controversial.27
Some animal models have shown that RBV reduces
viral replication in HMPV-infected mice,28 and use of
RBV to successfully treat the infection in humans has
been previously described.29 Other study data show
that RBV does not reduce mortality in immunocompromised patients and is not effective against the virus.30 Although case 1 in our series is an example of
successful treatment against HMPV using oral RBV,
this finding does not establish a beneficial effect of
RBV against HMPV infection.
Conclusions
Discovered 15 years ago, human metapneumovirus
(HMPV) can cause respiratory tract infections.1 Although data have been published on the prevalence of
the virus in high-risk groups (eg, children < 5 years of
age, persons > 60 years of age with comorbidities [eg,
cardiopulmonary diseases], immunocompromised individuals), important information on the immunological properties and pathogenesis of the virus is still unknown.5 This case series presents the clinical courses
of 3 severely immunocompromised patients infected
with HMPV and describes the impact of the virus on
their disease course. Infection with HMPV can add to
the severity of an underlying malignancy, especially in
immunocompromised patients, and can be a contributing factor toward the death of such patients. Therefore,
it is important to prevent this viral infection in immunocompromised individuals. Measures to control such
infection, including avoiding contact with large-particle
aerosols and droplets, practicing proper hand hygiene,
and, when possible, limiting the visitation of persons
who are ill, can all help to prevent the spread of HMPV.
A need exists for recommendations to guide treatment
for HMPV infection in high-risk patient groups, such as
the severely immunocompromised patients presented
in this case series. Development of an HMPV vaccine is
a reasonable goal for the prevention of this pathogen.
Acknowledgment: We gratefully acknowledge the
help of Peter P. Karpawich, MD, from the Detroit Medical Center in Michigan for reviewing this case series.
References
1. van den Hoogen BG, de Jong JC, Groen J, et al. A newly discovered human pneumovirus isolated from young children with respiratory
October 2016, Vol. 23, No. 4
tract disease. Nat Med. 2001;7(6):719-724.
2. Fauquet CM, Mayo MA, Maniloff J, et al, eds; Virology Division International Union of Microbiological Societies. Virus Taxonomy: Classification
and Nomenclature of Viruses. Eighth Report of the International Committee
on Taxonomy of Viruses. Oxford, UK: Elsevier Academic Press; 2005.
3. Kahn JS. Epidemiology of human metapneumovirus. Clin Microbiol
Rev. 2006;19(3):546-557.
4. Falsey AR, Walsh EE. Viral pneumonia in older adults. Clin Infect
Dis. 2006;42(4): 518-524.
5. Boivin G, Mackay I, SlootsTP, et al. Global genetic diversity of human
metapneumovirus fusion gene. Emerg Infect Dis. 2004;10(6):1154-1157.
6. Njenga MK, Lwamba HM, Seal BS. Metapneumovirus in birds and
humans. Virus Res. 2003;91(2):163-169.
7. Boivin G, DeSerres G, Côté S, et al. Human metapneumovirus infections in hospitalized children. Emerg Infect Dis. 2003;9(6):634-640.
8. Mullins JA, Erdman, DD, Weinberg GA, et al. Human metapneumovirus infection among children hospitalized with acute respiratory illness.
Emerg Infect Dis. 2004;10(4):700-705.
9. Schildgen O, Glatzel T, Geikowski T, et al. Human metapneumovirus RNA in encephalitis patient. Emerg Infect Dis. 2005;11(3):467-470.
10. Williams JV, Wang CK, Yang CF et al. The role of hMPV in upper
respiratory tract infections in children: a 20 year experience. J Infect Dis.
2006;193(3):387-395.
11. Williams JV, Harris PA, Tollefson SJ, et al. Human metapneumovirus and lower respiratory tract disease in otherwise healthy infants and
children. N Engl J Med. 2004;350(5):443-450.
12. Williams JV. Human metapneumovirus: an important cause of respiratory disease in children and adults. Curr Infect Dis Rep. 2005;7(3):204-210.
13. van Dijk NM, Linssen, CF, Bussink M, et al. Critical illness after horizontal nosocomial transmission of human metapneumovirus in the haemotology ward. Neth J Crit Care. 2012;16(1):22-25. http://njcc.nl/sites/default/
files/NJCC%2001%20casereport-vDijk.pdf. Accessed August 24, 2016.
14. Xepapadaki P, Psarras S, Bossios A, et al. Human metapneumovirus as a causative agent of acute bronchiolitis in infants. J Clin Virol.
2004;30(3):267-270.
15. Centers for Disease Control and Prevention. Outbreaks of human
metapneumovirus in two skilled nursing facilities-West Virginia and Idaho,
2011-2012. MMWR Morb Mortal Wkly Rep. 2013;62(46):909-913.
16. Cseke G, Maginnis MS, Cox RG, et al. Integrin alphabeta1 promotes infection by human metapneumovirus. Proc Natl Acad Sci USA.
20093;106(5):1566-1571.
17. Hamelin ME, Côté S, Laforge J, et al. Human metapneumovirus infection in adults with community acquired pneumonia and exacerbation of
chronic obstructive pulmonary disease. Clin Infect Dis. 2005;41(4):498-502.
18. Renaud C, Campbell AP. Changing epidemiology of respiratory
viral infections in hematopoietic cell transplant recipients and solid organ
transplant recipients. Curr Opin Infect Dis. 2011;24(4):333-343.
19. Milano F, Campbell AP, Guthrie KA, et al. Human rhinovirus and
coronavirus detection among allogeneic hematopoietic stem cell transplantation recipients. Blood. 2010;115(10):2088-2094.
20. Englund JA, Boeckh M, Kuypers J, et al. Brief communication: fatal
human metapneumovirus infection in stem cell transplant recipients. Ann
Intern Med. 2006;144(5): 344-349.
21. Chu HY, Renaud C, Ficken E, et al. Respiratory tract infections due
to human metapneumovirus in immunocompromised children. J Pediatric
Infect Dis Soc. 2014;3(4):286-293.
22. Godet C, Le Goff J, Beby-Defaux A, et al. Human metapneumovirus pneumonia in patients with hematological malignancies. J Clin Virol.
2014;61(4):593-596.
23. Kamboj M, Gerbin M, Huang CK, et al. Clinical characterization of
human metapneumovirus infection among patients with cancer. J Infect.
2008;57(6):464-471.
24. Côté S, Abed Y, Boivin G. Comparative evaluation of real-time
PCR assays for detection of human metapneumovirus. J Clin Microbiol.
2003;41(8):3631-3635.
25. Caracciolo S, Minni C, Colombrita D, et al. Human metapneumovirus
infection in young children hospitalized with acute respiratory tract disease:
virologic and clinical features. Pediatr Infect Dis J. 2008;27(5):406-412.
26. Buchholz UJ, Nagashima K, Murphy BR, et al. Live vaccines for human metapneumovirus designed by reverse genetics. Expert Rev Vaccines.
2006;5(5):695-706.
27. Hirsch HH, Martino R, Ward KN, et al. Fourth European Conference
on Infections in Leukaemia (ECIL-4): guidelines for diagnosis and treatment of human respiratory syncytial virus, parainfluenza virus, metapneumovirus, rhinovirus, and coronavirus. Clin Infect Dis. 2013;56(2):258-266.
28. Hamelin ME, Prince GA, Boivin G. Effect of ribavirin and glucocorticoid treatment in a mouse model of human metapneumovirus infection.
Antimicrob Agents Chemother. 2006;50(2):774-777.
29. Kitanovski L, Kopriva S, Pokorn M, et al. Treatment of severe human metapneumovirus pneumonia in an immunocompromised child with
oral ribavirin and IVIG. J Pediatr Hematol Oncol. 2013;35(7):e311-e313.
30. Debur MC, Vidal LR, Stroparo E, et al. Human metapneumovirus
infection in hematopoietic stem cell transplant recipients. Transpl Infect
Dis. 2010;12(2):173-179.
Cancer Control 445
Special Report
Systematic Review and Case Series Report of Acinar Cell
Carcinoma of the Pancreas
Evan S. Glazer, MD, PhD, Kevin G. Neill, MD, Jessica M. Frakes, MD, Domenico Coppola, MD,
Pamela J. Hodul, MD, Sara E. Hoffe, MD, Jose M. Pimiento, MD, Gregory M. Springett, MD, PhD,
and Mokenge P. Malafa, MD, PhD
Background: Acinar cell carcinoma of the pancreas is a rare malignancy representing less than 1% of all
pancreatic malignancies.
Methods: We report on a case series of 21 patients with acinar cell carcinoma of the pancreas treated at a
high-volume quaternary center. A systematic review of the medical literature was performed that described
typical therapeutic management approaches for acinar cell carcinoma of the pancreas and reported on disease
control and survival rates. Data for the case series were obtained from a prospective database.
Results: In our systematic review of 6 articles, study patients had a median age of 61 years, 66% were male,
52% had stage I/II disease, and 55% of lesions were located in the pancreatic head. The rates of median
survival were approximately 47 months after resection with adjuvant therapy, 38 months for nonmetastatic,
locally unresectable disease, and 17 months for metastatic disease treated with chemotherapy. Combination
fluoropyrimidine-based chemotherapy regimens had better rates of disease control than other therapies. Our
case series included 21 study patients, 14 of whom required resection and 7 who had metastatic disease. The
rates of median survival were 40.2 ± 31.9 months in those who underwent surgery and were treated with
adjuvant therapy and 13.8 ± 11.3 months for patients with metastatic disease.
Conclusions: Multidisciplinary treatment for acinar cell carcinoma of the pancreas should be considered due
to the rarity of the disease and its lack of high-level therapeutic data. Progress in the molecular analysis of this
tumor may improve outcomes through the use of personalized therapy based on underlying tumor mutations.
Introduction
Acinar cell carcinoma of the pancreas is a rare exocrine
malignancy representing less than 1% of all pancreatic
neoplasms.1 The exocrine pancreas is primarily composed of ductal cells and acinar cells. Acinar cells are
enzyme-secreting cells that release amylase, lipases,
and proteases. Debate exists regarding whether acinar
cell carcinoma of the pancreas is unique from pancreatic ductal adenocarcinoma2,3; however, acinar cell carcinoma of the pancreas responds to treatment differently than pancreatic ductal adenocarcinoma and its
rates of overall survival are not similar4 — data suggesFrom the Departments of Gastrointestinal Oncology (ESG, PJH,
JMP, MPM), Pathology (KGN, DC, GMS), and Radiation Oncology
(JMF, SEH), H. Lee Moffitt Cancer Center & Research Institute,
Tampa, Florida.
Submitted February 18, 2016; accepted May 31, 2016.
Address correspondence to Mokenge P. Malafa, MD, PhD,
Department of Gastrointestinal Oncology, Moffitt Cancer Center,
12902 Magnolia Drive, FOB-2 GI PROG, Tampa, FL 33612.
E-mail: [email protected]
Dr Glazer is now affiliated with the Division of Surgical Oncology
in the
Department of Surgery at the
University of Tennessee Health
Sciences Center, Memphis, Tennessee.
No significant relationships exist between the authors and the
companies/organizations whose products or services may be referenced in this article.
446 Cancer Control
tive that it is a distinct entity.
The rarity of acinar cell carcinoma of the pancreas, lack of high-quality evidence, and its diagnostic
difficulty have all limited consensus among authors
regarding the most appropriate management of this
disease.5-7 Although patients who have undergone resection for acinar cell carcinoma of the pancreas may
appear to have higher survival rates than patients with
pancreatic ductal adenocarcinoma3,4 — whose median
survival rate is between 4 and 5 years1 — the limited
and selection-biased data make assertions difficult.
Nonsurgical treatment options for patients with
acinar cell carcinoma of the pancreas include chemotherapy and radiotherapy. Use of various chemotherapeutic agents, including capecitabine monotherapy
and folinic acid/fluorouracil/oxaliplatin (FOLFOX),
have been reported in the literature.8,9 Whether radiotherapy improves survival rates in patients with acinar
cell carcinoma of the pancreas remains unclear.10-12
Case reports of the use of radiotherapy to treat acinar
cell carcinoma of the pancreas have noted an association with extended rates of survival.8,13
The patterns of genetic alterations and specific
driver mutations in patients with acinar cell carcinoma of the pancreas are not well described in the
literature. Studies have demonstrated lower rates of
October 2016, Vol. 23, No. 4
EGFR and KRAS mutations in acinar cell carcinoma of
the pancreas than in pancreatic ductal adenocarcinoma.14 Some have suggested that pancreatic ductal adenocarcinoma may develop from acinar cells that undermutate p53 without evidence of developing acinar
cell carcinoma of the pancreas.15 In addition, nearly
one-half of acinar cell carcinoma of the pancreas tumors have alterations in APC.16
Unlike pancreatic ductal adenocarcinoma, KRAS mutation is infrequently a driving force in acinar cell carcinoma of the pancreas; however, chromosomal imbalances
are common in acinar cell carcinoma of the pancreas.14
Whole-exome sequencing has revealed that mutations in
BRCA2 and the FAT family are common in patients with
acinar cell carcinoma of the pancreas, but these mutations are not yet readily targetable.17 p53 expression has
been demonstrated as both a positive and negative prognostic indicator in acinar cell carcinoma of the pancreas.18
More than 70% of tumors of the acinar cell carcinoma
of the pancreatic type demonstrated a decrease or complete loss of DCC expression on immunohistochemistry.14 Bergmann et al14 also suggested that DCC variation
is an early genetic change in acinar cell carcinoma of the
pancreas, whereas others have suggested that it is a late
change in pancreatic ductal adenocarcinoma.19
Purpose
The purpose of this systematic review is to delineate
the most appropriate treatment and expected outcomes in patients diagnosed with acinar cell carcinoma of the pancreas. Our article focuses on adult patients because genetic mutations in childhood tumors
suggest alternative etiologies.
Methods
Literature and Systematic Review
We searched the medical literature for studies describing typical therapeutic management approaches for
acinar cell carcinoma of the pancreas and reported
on disease control and survival rates. Studies were
excluded during the initial search if they investigated
fewer than 4 patients, focused on mixed carcinoma,
were not written in English, did not include clinical
data, were review articles, or were a technical manuscript on a surgical procedure alone. Case reports with
fewer than 4 patients were also excluded due to lack
of ability (too few patients to develop a systematic approach given the rarity of the malignancy). For studies
that included previously published data, the most complete and most recent data were used for our systematic analysis. When studies listed patient-level data for
multiple histologies, only data concerning acinar cell
carcinoma of the pancreas were used. Rates of disease
control were calculated using complete response in
combination with partial response and stable disease
(based on the definition used in each study). No studOctober 2016, Vol. 23, No. 4
ies were identified as having high-level evidence.
A total of 132 manuscripts were identified during the search, and 10 studies were included based
on our inclusion/exclusion criteria.20 Six studies were
then identified and selected for summary analysis of
response rates to chemotherapy and radiotherapy as
well as to estimate the average median overall survival
rate.3,21-25 Four studies contained incomplete data or
nonstandard treatment approaches and were excluded
from the summary data analyses but were included in
the systematic review.
Case Series
We reviewed our records for patients with acinar cell
carcinoma of the pancreas treated between 2004 and
2014 at the H. Lee Moffitt Cancer Center & Research
Institute (Tampa, FL). Demographical, clinical, and
pathological data were obtained from a retrospective
database. Charts were reviewed to confirm data. The
Institutional Review Board at our institution approved
this research.
Statistical Analyses
For the systematic review, summary clinical data from
each study were averaged in a weighted fashion to
determine the overall value for each characteristic
(demographic, treatment, or survival data). STATA statistical software was utilized for analysis (StataCorp,
College Station, Texas).
For the case series, data analyses were also performed with STATA (StataCorp), using χ2, KaplanMeier, and multivariate linear-regression techniques.
Statistical significance was set to α being equal to .05.
Uncertainties are standard deviations of the mean unless otherwise specified.
Results
A total of 68 patients identified from the systematic review had a median age of 61 years, and 66% were men
(Table 1). These results are similar to other nonsystemTable 1. — Characteristics of Patients With Acinar Cell
Carcinoma of the Pancreas in Select Studies
Study
Butturini25
No. of Median
Study
Age, y
Patients
9
Male,
%
Stage Pancreatic
I/II,
Head, %
%
53
67
57
67
39
60
77
46
44
Lowery 24
40
65
73
55
55
Matos 3
17
60
Unknown
59
76
Holen
21
4
63
75
25
62
Seth23
14
57
33
86
64
Current
series
21
64
76
33
66
Seki
22
Cancer Control 447
atic analyses in which the male-to-female patient ratio is at least 2:1.3,18,21-26 In our systematic review, 52%
of patients with acinar cell carcinoma of the pancreas had resectable disease (stage I/II; see Table
1).3,21-25 Nonspecific symptoms, such as nausea, abdominal pain, and weight loss, were observed in most patients.18 We also found that 55% of patients with acinar
cell carcinoma of the pancreas had tumors located in
the pancreatic head. The data derived from the systematic review also demonstrated that most of the lesions
described were generally well circumscribed without
significant peritumoral stroma.3,25
Diagnosis
The diagnosis of acinar cell carcinoma of the pancreas is confirmed by biopsy, such as fine needle aspiration, or resection of the pancreatic tumor — with the
latter being more reliable than the other methods.27
The highly cellular nodules of monotonous tumor
cells have little to no stroma and lack a desmoplastic
response (Fig 1). Therefore, tumor cells may be difficult to identify with fine needle aspiration alone but
reliably seen on a resected specimen.1 Tumor cells
are arranged in solid sheets with spaces between
the acinar structures and small lumens.18 Cytologically, these cells exhibit basal polarization. Fig 1 also
demonstrates that nuclei can range from moderate to
marked atypia and contain a single, prominent nucleolus. In the apical region, the cells contain moderate
to abundant eosinophilic cytoplasm rich in zymogen
granules.
Other groups have utilized immunohistochemistry to confirm the diagnosis, and researchers have
demonstrated that an antibody against a portion of
the B-cell chronic lymphocytic leukemia/lymphoma
10 protein has utility in diagnosing acinar cell carcinoma of the pancreas.18 In addition, immunohistochemistry staining for trypsin and chymotrypsin in
A
the acinar pattern and, typically, lack of staining for
neuroendocrine neoplasm markers (neural cell adhesion molecule, chromogranin, synaptophysin) can
confirm the diagnosis of acinar cell carcinoma of the
pancreas.14
Radiotherapy
Our systematic review revealed a modest response
rate to radiotherapy in patients with localized acinar cell carcinoma of the pancreas. In several studies, a “major response” rate was seen in 25% to 35%
of these patients. 21,23 However, if stable disease
was included in the definition of response (ie,
disease control rate), significantly higher response
rates were seen in highly selected studies when radiotherapy was added (Table 2).3,21-25 Radiotherapy is
often used to “down-stage” or convert a tumor from
being borderline resectable to resectable, and, in general, is provided with both conventional fractionation
and hypofractionated stereotactic body radiotherapy
(SBRT).25 Lack of progression following neoadjuvant
radiotherapy can help select patients who may benefit
from resection.
Chemotherapy
Significant heterogeneity was seen in the systematic
review regarding specific chemotherapeutic protocols; however, most study patients received combination gemcitabine- or fluoropyrimidine-based
chemotherapy protocols. 23,25 Indeed, Holen et al 21
and Lowery et al24 concurred on the favored use of
combination fluoropyrimidine-based therapies and
radiotherapy in patients with acinar cell carcinoma
of the pancreas. However, data regarding disease
control rate (complete response + partial response +
stable disease) are mixed (see Table 2).3,21-25,28 Combination fluoropyrimidine-based chemotherapies
were shown to be associated with higher rates of
B
Fig 1A–B. — (A) Acinar cell carcinoma of the pancreas in a man aged 83 y and (B) pancreatic ductal adenocarcinoma in a woman aged 65 y.
Hematoxylin and eosin stain, ×20.
448 Cancer Control
October 2016, Vol. 23, No. 4
disease control (Table 3).3,21-25 Therefore, FOLFOX or
folinic acid/fluorouracil/irinotecan (FOLFIRI) is usually administered to patients with high performance
status, whereas patients with a lower performance
status usually receive gemcitabine/protein-bound
paclitaxel.3,8,9,21-25
Table 2. — Disease Control Rates of Chemotherapy
and Radiotherapy in Select Series of Acinar Cell Carcinoma
of the Pancreas
Study
Type of Treatment
No. of
Patients
Disease
Control
Rate,a %
Butturini25
Gemcitabine-based
chemotherapy
7
29
Holen21
Fluoropyrimidine-based
chemotherapy
22
41
RT alone
1
100
Fluoropyrimidine-based
chemotherapy
RT
4
100
Lowery 24
Gemcitabine-based
chemotherapy
20
35
Matos3
Mixed chemotherapy
4
50
Mixed chemotherapy
RT
2
100
Gemcitabine-based
chemotherapy
1
0
Fluoropyrimidine-/gemcitabinebased chemotherapy
3
33
Seth23
Mixed chemotherapy
RT
4
100
Current
series
Gemcitabine-based
chemotherapy
6
50
Fluoropyrimidine-based
chemotherapy
6
67
Mixed chemotherapy
RT
5
100
Seki22
Table 4. — Systematic Review Summary of Survival
Differences Between Select Patients With Resectable and
Nonresectable Acinar Cell Carcinoma of the Pancreas
Disease
Status
Metastatic
Primary Treatment
Median Rate
of Overall
Survival, mos
Chemotherapy
17
Unresectable
Chemotherapy and radiotherapy
38
Resectable
Resection and adjuvant therapy
47
Patient Characteristic
No. of Patients (%)
Sex
Male
16 (76)
Female
5 (24)
II
Table 3. — Summary of Select Disease Control Rates
For the Nonsurgical Therapy of Acinar Cell Carcinoma
of the Pancreas
No. of
Patients
Disease
Control
Rate,a %
Any chemotherapy
79
55
Any radiotherapy
16
100
Gemcitabine-based chemotherapy
34
28
Fluoropyrimidine-based chemotherapy
35
60
a Complete response + partial response + stable disease.
Data from references 3 and 21 to 25.
Table 5. — Select Patient Characteristics From Current
Patient Series
American Joint Committee on Cancer Stage
Study patients not receiving chemotherapy are not included.
response + partial response + stable disease.
RT = radiotherapy.
October 2016, Vol. 23, No. 4
Treatment at Our Institution
We identified 21 patients in a case series; 7 patients
had metastatic disease based on imaging at the time
of initial presentation (Table 5). Most patients were
Data from references 3 and 21 to 25.
a Complete
Treatment
Surgery
Overall, surgical resection remains the mainstay of therapy for nonmetastatic disease, although adjuvant therapy is common. Most patients with acinar cell carcinoma
of the pancreas undergo negative margin resection.3,21
From our systematic review, we found that patients who
underwent resection had an overall median survival rate
of approximately 47 months (Table 4).3,21-25 The contribution of adjuvant chemotherapy and radiotherapy has
not been well investigated in the literature. Resection
and adjuvant chemotherapy were associated with the
longest rate of median overall survival (see Table 4).3,21-25
7 (33)
III
5 (24)
IV
9 (43)
Site
Head
8 (38)
Neck/body
7 (33)
Tail
6 (29)
Surgical Intervention
Whipple
6 (29)
Distal pancreatectomy
3 (14)
Total pancreatectomy
2 (10)
Unresectable
3 (14)
No surgery
7 (33)
Mean age ± standard deviation: 63.5 ± 14 y.
Cancer Control 449
100
No surgery
Surgery
P < .001
75
50
25
0
0
20
Survival Probability, %
No surgery
Surgery
7
14
0
8
80
0
3
0
1
0
0
100
75
No RT
RT
P = .008
50
25
0
0
20
40
No RT
RT
16
5
4
4
1
2
C
80
0
1
0
0
Discussion
No chemo
Chemo
P = .09
100
75
50
25
0
0
20
40
60
80
0
1
0
0
Months
No. at Risk
No chemo
Chemo
60
Months
No. at Risk
Survival Probability, %
60
Months
No. at Risk
B
40
4
17
1
7
1
2
Fig 2A–C. — (A) Kaplan-Meier survival estimates based on surgery,
(B) RT, and (C) chemotherapy. Patients with acinar cell carcinoma of the
pancreas had significantly improved rates of survival following surgery
(P < .001) and RT (P = .008) but not chemotherapy (P = .09).
RT = radiotherapy.
men. The average age was 63.5 ± 14.1 years, and 38%
of lesions were located in the pancreatic head. No patients had stage I disease. The median follow-up period was 1.2 years (range, 1 month to 6.5 years), and
62% of study patients were initially diagnosed elsewhere. Of the 21 patients, 14 were candidates for curative resection. Of those, 11 underwent R0 or R1 resections and 3 patients received R2 resection. At last
follow-up, 43% of patients were alive; rates of actual
1- and 5-year survival were 67% and 5%, respectively. All patients who received radiotherapy lived for at
450 Cancer Control
least 13 months after diagnosis.
Of the patients who underwent surgery, 1 patient
also received neoadjuvant chemotherapy and 1 patient
also received neoadjuvant radiotherapy. Eight patients
received adjuvant chemotherapy, and 4 patients received adjuvant radiotherapy. More surgical patients
received chemotherapy than nonsurgical patients
(93% vs 57%; P < .05), with 82% of those receiving a
gemcitabine-based chemotherapy protocol. Five patients (24%) received radiotherapy, and all were surgical
patients: 1 in a neoadjuvant and 4 in adjuvant settings.
Utilizing Kaplan-Meier analysis, we found rates
of survival to be statistically associated with stage of
disease (P = .005), surgery (Fig 2A; log-rank P < .001),
and radiotherapy (Fig 2B; log-rank P = .008), but not
chemotherapy (Fig 2C; log-rank P = .09). Patients who
underwent resection followed by adjuvant radiotherapy had the longest survival (Fig 3). On multivariate
Cox regression analysis, higher disease stage was associated with a shorter rate of survival (hazard ratio
[HR] 6.70, P =.003) and adjuvant radiotherapy was associated with longer survival (HR 0.05, P = .03), but
neither R0 margin nor adjuvant chemotherapy was
statistically associated with improved rates of survival
(0.70 and 0.30, respectively; P = .2 for both).
Few large trials have studied acinar cell carcinoma of
the pancreas because it is such a rare tumor. We were
only able to identify 6 studies that fully met the criteria
for our systematic review. Our institutional experience
with 21 patients is the second largest case series in the
literature. Because acinar cell carcinoma of the pancreas is treated similarly to pancreatic ductal adenocarcinoma, it is interesting to compare the outcomes. Analysis of Surveillance, Epidemiology, and End Results data
Survival Probability, %
Survival Probability, %
A
100
75
No RT
RT
P = .018
50
25
0
0
20
60
80
0
1
0
0
Months
No. at Risk
No RT
RT
40
9
5
4
4
1
2
Fig 3. — Kaplan-Meier survival estimates among patients undergoing
surgery and receiving RT. Surgical candidates with acinar cell carcinoma
of the pancreas had significant improvement in their rates of survival
probability when treated with RT.
RT = radiotherapy.
October 2016, Vol. 23, No. 4
demonstrated higher rates of survival in acinar cell
carcinoma of the pancreas compared with pancreatic
ductal adenocarcinoma for both resected and unresected cases,1 and analysis of the National Cancer Database demonstrated 5-year survival rates for patients
with resected acinar cell carcinoma of the pancreas
of 52%, 40%, 23%, and 17% for stages I, II, III, and IV,
respectively.29 Positive nodal disease was not associated with decreased rates of survival in patients with
acinar cell carcinoma of the pancreas, but patients with
high-grade acinar cell carcinoma of the pancreas had a
2-fold worse rate of 5-year survival (P = .003).29 Regarding margin status, negative microscopic margins were
associated with 2-year median survival rates compared
with 1-year median survival rates for patients with
R1-/R2-positive margins (P < .02).29
Debate exists as to whether pancreatic ductal adenocarcinoma is a radiosensitive tumor, but acinar
cell carcinoma of the pancreas does appear to be radiosensitive based on the multivariate analysis and
Kaplan-Meier survival curves presented here along
with other published data.30 Given the findings here
and in the literature, it appears that, although acinar
cell carcinoma of the pancreas and pancreatic ductal
adenocarcinoma are similar exocrine pancreatic malignancies, they are distinct entities.29,30
This systematic review and case series illuminate
3 important findings. As expected, patients able to tolerate surgery live significantly longer than patients with
unresectable disease.3,21-25 Patients who undergo radiotherapy in combination with surgery also live significantly longer than those not receiving radiotherapy.3,21-25
The biological mechanism of this result is unknown, but
we hypothesize it is because acinar cell carcinoma of the
pancreas is a radiosensitive tumor. This survival advantage of radiotherapy was seen despite 11 of 14 patients
in our case series receiving surgery (R0/R1 resections),
further suggesting that radiotherapy is valuable regardless of margin status. We did not find an association between use of chemotherapy and survival rate. This may
be due to the low number of patients in the nonchemotherapy group, but lack of high-level evidence remains
problematic given the overall poor prognosis of acinar
cell carcinoma of the pancreas.
The cell of origin for acinar cell carcinoma of the
pancreas is presumed to be the acinar cell, whereas
the cell of origin for pancreatic adenocarcinoma is the
duct epithelium.31-33 However, controversy is ongoing
regarding this assumption because it is based on the
phenotype of pancreatic ducts in normal tissue, pancreatic intraepithelial neoplastic lesions, and carcinoma.31 For example, injury to benign cells in the setting
of malignancy (ie, enzyme-mediated cell destruction
due to malignant local duct obstruction) may create a
setting in which malignancy may appear to originate in
a duct cell when in fact it actually originated in an aciOctober 2016, Vol. 23, No. 4
nar cell.33 An alternative pathway for carcinogenesis is
described by KRAS-mediated acinar-ductal metaplasia
and the subsequent development of pancreatic ductal
adenocarcinoma.31,34 In this setting, KRAS is hypothesized as being the driving force for the development of
pancreatic ductal adenocarcinoma, as most commonly
seen in the humans, whereas non–KRAS-mutated exocrine pancreatic carcinomas develop into acinar cell
carcinoma of the pancreas.35-37 Assuming this is true,
then KRAS wild-type status would be a critical finding
in distinguishing acinar cell carcinoma of the pancreas
from pancreatic ductal adenocarcinoma.
Mutation of DCC may also serve as an early genetic change in acinar cell carcinoma of the pancreas;
by contrast, it is a late change in pancreatic ductal adenocarcinoma. Furthermore, the rate of disease control
was higher in patients with acinar cell carcinoma of the
pancreas than in those with pancreatic ductal adenocarcinoma. This finding suggests that acinar cell carcinoma of the pancreas and pancreatic ductal adenocarcinoma are 2 distinct malignancies arising from similar
cells but with important and early points of divergence
(KRAS vs DCC). Other groups have demonstrated that
acinar cell cystadenoma, a benign and rarely premalignant pancreatic lesion, may have genetic alteration
patterns more similar to pancreatic ductal adenocarcinoma than to acinar cell carcinoma of the pancreas,
suggesting that some genetic overlap may be present.38
At our institution, we favor the initial use of
FOLFOX followed by FOLFIRI as second-line treatment
for patients with acinar cell carcinoma of the pancreas and moderate to high performance status (Eastern Cooperative Oncology Group [ECOG] 0–1) either
in the adjuvant setting or as systemic therapy (Fig 4).
Use of folinic acid/fluorouracil/irinotecan/oxaliplatin (FOLFIRINOX) as first-line therapy for metastatic acinar cell carcinoma of the pancreas has been
described in the literature, but whether the benefits of
such an aggressive regimen outweighs the risks is unclear.39 Because rates of disease control are higher with
the use of combination, fluoropyrimidine-based chemotherapies, we also consider FOLFIRINOX in relatively young, very highly active individuals with ECOG
performance status 0 (but rarely seen in our clinical
practice). Among patients with moderate ECOG performance status 1 to 2, gemcitabine combined with
protein-bound paclitaxel is our preferred chemotherapy regimen, with single-agent gemcitabine used for
patients with poor performance status not wishing to
enter hospice or palliative care.
In the neoadjuvant setting, our clinical approach
for pancreatic cancer typically involves treating patients with SBRT who have borderline resectable or
locally advanced disease. Therapy involves 5 days of
fiducial marker–based radiotherapy applied to gross
disease (median dose of 30 Gy and simultaneously
Cancer Control 451
50 Gy to the area of vessel involvement/abutment) to
help facilitate a negative margin resection.40,41 In patients with acinar cell carcinoma of the pancreas, this
general treatment approach could be considered given
the high R0 resection rates and minimal rates of late
toxicity.30,42 Based on data from the National Cancer
Database, R0 resection has been shown to improve
rates of overall survival on multivariate analysis, so
neoadjuvant radiotherapy should be considered when
a likelihood exists of positive margins.29 Because patients with locally advanced tumors are unlikely to be candidates
for resection, care must be taken
to avoid late radiation-associatDiagnosis Made on Biopsy (eg, FNA)
ed toxicities to the surrounding
gastrointestinal organs. As such,
intensity-modulated radiotheraComplete Staging
py can be considered instead of
Local/regional:
SBRT for locally advanced acinar
• Endoscopic ultrasonography to evaluate superior mesenteric vein/artery and portal vein
cell carcinoma of the pancreas.
• CT protocol for the pancreas (cut 3 mm, arterial/portal/venous phase)
Distant:
• CT of the thorax with contrast and PET-CT
Performance status
Resectable
Pancreaticoduodenectomy
or distal pancreatectomy
Borderline
Resectable
Neoadjuvant
chemotherapy +
SBRT +
resectiona
Adjuvant
Chemotherapy
FOLFOX
Adjuvant
Radiotherapy
Positive
margins
SBRT-naïve
Locally
Advanced
Systemic
chemotherapy
FOLFOX
RT
Metastatic
Systemic
chemotherapy
FOLFOX
Progression
Second-Line Therapy
FOLFIRI
Third-Line Therapy
Gemcitabine/protein-bound
paclitaxel
Palliative Care
Fig 4. — Algorithm for the treatment of acinar cell carcinoma of the pancreas. This treatment plan
is based on expert opinion and focuses on the importance of negative-margin resection coupled
with RT.
CT = computed tomography, FNA = fine needle aspiration, FOLFIRI = folinic acid/fluorouracil/irinotecan, FOLFOX = folinic acid/fluorouracil/oxaliplatin, PET = positron emission tomography,
RT = radiotherapy, SBRT = stereotactic body radiotherapy.
aIf no progression on repeat staging.
452 Cancer Control
Limitations
Due to the rarity of this malignancy, we did not identify any
prospective studies or large casecontrol studies to help guide clinical management. Likewise, oncologists are more familiar with the
treatment protocols for pancreatic
ductal adenocarcinoma, because
this disease is more common,
and, as such, these regimens are
often utilized for acinar cell carcinoma of the pancreas despite the
fact that evidence is lacking. The
retrospective nature of this review
also limits our interpretation of
the results. Fundamentally, this
limitation is also due to the rarity
of the malignancy, thus limiting
the initiation of prospective trials. Another important limitation
relates to tumor response rates in
retrospective studies. Oftentimes,
the time to progression is based
on when a patient is scheduled for
his or her next imaging, not a predefined time point, as seen in a
prospective study. As such, unless
a patient is symptomatic, the time
to progression may be partially related to an institutional bias. This
is a difficult to quantify but important limitation.
Future Research
Three critical inquiries must be explored to improve rates of survival
among individuals with acinar cell
carcinoma of the pancreas. Specific mechanisms (ie, gene mutaOctober 2016, Vol. 23, No. 4
tions) that lead to acinar cell carcinoma of the pancreas
must be better elucidated to (theoretically) incorporate
small molecule inhibitors into therapy. As demonstrated by the overall higher survival rates seen in acinar
cell carcinoma of the pancreas (see Table 4), acinar cell
carcinoma of the pancreas may likely be molecularly
driven in a distinct way from pancreatic ductal adenocarcinoma.3,21-25 The radiosensitivity of acinar cell carcinoma of the pancreas must also be investigated and
modulated to optimize therapy. Our data suggest that
acinar cell carcinoma of the pancreas is a radiosensitive tumor. Similar to many other malignancies, active
chemotherapeutics also need identification. Whether
current chemotherapy options add any improved likelihood of long-term survival remains unclear based on
published data.
Conclusions
Acinar cell carcinoma of the pancreas is a rare pancreatic malignancy with a prognosis slightly better than
ductal adenocarcinoma. At Moffitt Cancer Center, our
standard approach is surgical resection followed by adjuvant fluorouracil-based chemotherapy, radiotherapy,
or both (see Fig 4). The utility of chemotherapy is uncertain; however, when it is used, a fluorouracil-based
regimen is often proposed because of its higher rates
of disease control when compared with gemcitabine or
other chemotherapeutic agents. (However, this treatment plan is based on expert opinion and individualized for patient performance status.) Negative microscopic margins are the most important prognostic
factor, because survival for patients with positive microscopic margins is generally as poor as those seen
with positive gross margins.
Multidisciplinary management of pancreatic acinar
cell carcinoma should be considered due to the lack of
high-level data in treating this patient population. In
addition, referral to a quaternary center is appropriate
given the rarity of this malignancy. Despite the rarity
of this disease, further investigation is needed to personalize care to improve long-term survival in these
patients.
Acknowledgments: The authors thank Rasa
Hamilton for editorial support and Kim Murphy for administrative support.
References
1. Wisnoski NC, Townsend CM Jr, Nealon WH, et al. 672 patients with
acinar cell carcinoma of the pancreas: a population-based comparison to
pancreatic adenocarcinoma. Surgery. 2008;144(2):141-148.
2. Kong B, Michalski CW, Erkan M, et al. From tissue turnover to
the cell of origin for pancreatic cancer. Nat Rev Gastroenterol Hepatol.
2011;8(8):467-472.
3. Matos JM, Schmidt CM, Turrini O, et al. Pancreatic acinar cell carcinoma: a multi-institutional study. J Gastrointest Surg. 2009;13(8):1495-1502.
4. Wang Y, Wang S, Zhou X, et al. Acinar cell carcinoma: a report
of 19 cases with a brief review of the literature. World J Surg Oncol.
2016;14(1):172.
5. Basturk O, Adsay V, Hruban RH, et al. Pancreatic acinar cell carcinomas with prominent neuroendocrine differentiation: clinicopathologic
October 2016, Vol. 23, No. 4
analysis of a distinct and diagnostically challenging neoplasm. Modern
Pathol. 2014;27(suppl 2):447A.
6. Bergmann F, Aulmann S, Welsch T, et al. Molecular analysis of
pancreatic acinar cell cystadenomas: Evidence of a non-neoplastic nature.
Oncol Lett. 2014;8(2):852-858.
7. Hartwig W, Denneberg M, Bergmann F, et al. Acinar cell carcinoma
of the pancreas: is resection justified even in limited metastatic disease?
Am J Surg. 2011;202(1):23-27.
8. Lee JL, Kim TW, Chang HM, et al. Locally advanced acinar cell
carcinoma of the pancreas successfully treated by capecitabine and concurrent radiotherapy: report of two cases. Pancreas. 2003;27(1):e18-e22.
9. Chaudhary P, Ranjan G, Chaudhary A, et al. Acinar cell carcinoma:
a rare pancreatic malignancy. Clin Pract. 2013;3(2):e18.
10. Wang T, X SM, Kong D, et al. Effect of ionizing radiation on acinar
morphogenesis of human prostatic epithelial cells under three-dimensional culture conditions. Neoplasma. 2012;59(3):269-281.
11. Tateishi Y, Sasabe E, Ueta E, et al. Ionizing irradiation induces
apoptotic damage of salivary gland acinar cells via NADPH oxidase
1-dependent superoxide generation. Biochem Biophys Res Commun.
2008;366(2):301-307.
12. Peter B, Van Waarde MA, Vissink A, et al. Radiation-induced cell
proliferation in the parotid and submandibular glands of the rat. Radiat
Res. 1994;140(2):257-265.
13. Ogbonna OH, Garcon MC, Syrigos KN, et al. Mixed acinar-neuroendocrine carcinoma of the pancreas with neuroendocrine predominance.
Case Rep Med. 2013;2013:705092.
14. Bergmann F, Aulmann S, Sipos B, et al. Acinar cell carcinomas of
the pancreas: a molecular analysis in a series of 57 cases. Virchows Archiv.
2014;465(6):661-672.
15. Baer R, Cintas C, Dufresne M, et al. Pancreatic cell plasticity and
cancer initiation induced by oncogenic Kras is completely dependent on
wild-type PI 3-kinase p110alpha. Genes Dev. 2014;28(23):2621-2635.
16. Furlan D, Sahnane N, Bernasconi B, et al. APC alterations are frequently involved in the pathogenesis of acinar cell carcinoma of the pancreas, mainly through gene loss and promoter hypermethylation. Virchows
Arch. 2014;464(5):553-564.
17. Furukawa T, Sakamoto H, Takeuchi S, et al. Whole exome sequencing reveals recurrent mutations in BRCA2 and FAT genes in acinar
cell carcinomas of the pancreas. Sci Rep. 2015;5:8829.
18. La Rosa S, Sessa F, Capella C. Acinar cell carcinoma of the pancreas: overview of clinicopathologic features and insights into the molecular pathology. Front Med (Lausanne). 2015;2:41.
19. Hilgers W, Song JJ, Haye M, et al. Homozygous deletions inactivate
DCC, but not MADH4/DPC4/SMAD4, in a subset of pancreatic and biliary
cancers. Genes Chromosomes Cancer. 2000;27(4):353-357.
20. Moher D, Liberati A, Tetzlaff J, et al. Preferred reporting items for
systematic reviews and meta-analyses: the PRISMA statement. PLoS
Med. 2009;6(7):e1000097.
21. Holen KD, Klimstra DS, Hummer A, et al. Clinical characteristics
and outcomes from an institutional series of acinar cell carcinoma of the
pancreas and related tumors. J Clin Oncol. 2002;20(24):4673-4678.
22. Seki Y, Okusaka T, Ikeda M, et al. Four cases of pancreatic acinar
cell carcinoma treated with gemcitabine or S-1 as a single agent. Jpn J Clin
Oncol. 2009;39(11):751-755.
23. Seth AK, Argani P, Campbell KA, et al. Acinar cell carcinoma of the
pancreas: an institutional series of resected patients and review of the current
literature. J Gastrointest Surg. 2008;12(6):1061-1067.
24. Lowery MA, Klimstra DS, Shia J, et al. Acinar cell carcinoma of
the pancreas: new genetic and treatment insights into a rare malignancy.
Oncologist. 2011;16(12):1714-1720.
25. Butturini G, Pisano M, Scarpa A, et al. Aggressive approach to acinar cell carcinoma of the pancreas: a single-institution experience and a
literature review. Langenbecks Arch Surg. 2011;396(3):363-369.
26. Lack EE, Cassady JR, Levey R, et al. Tumors of the exocrine pancreas in children and adolescents. A clinical and pathologic study of eight
cases. Am J Surg Pathol. 1983;7(4):319-327.
27. Kim HJ, Kim YK, Jang KT, et al. Intraductal growing acinar cell carcinoma of the pancreas. Abdominal Imaging. 2013;38(5):1115-1119.
28. Sudo K, Ishihara T, Hirata N, et al. Randomized controlled study
of gemcitabine plus S-1 combination chemotherapy versus gemcitabine
for unresectable pancreatic cancer. Cancer Chemother Pharmacol.
2014;73(2):389-396.
29. Schmidt CM, Matos JM, Bentrem DJ, et al. Acinar cell carcinoma
of the pancreas in the United States: prognostic factors and comparison to
ductal adenocarcinoma. J Gastrointest Surg. 2008;12(12):2078-2086.
30. Moningi S, Dholakia AS, Raman SP, et al. The role of stereotactic
body radiation therapy for pancreatic cancer: a single-institution experience. Ann Surg Oncol. 2015;22(7):2352-2358.
31. Shi C, Hong SM, Lim P, et al. KRAS2 mutations in human pancreatic acinar-ductal metaplastic lesions are limited to those with PanIN: implications for the human pancreatic cancer cell of origin. Mol Cancer Res.
2009;7(2):230-236.
32. Morohoshi T, Held G, Kloppel G. Exocrine pancreatic tumours and
their histological classification. A study based on 167 autopsy and 97 surgiCancer Control 453
cal cases. Histopathology. 1983;7(5):645-661.
33. Bockman DE. Cells of origin of pancreatic cancer: experimental animal tumors related to human pancreas. Cancer. 1981;47(6 suppl):1528-1534.
34. Aichler M, Seiler C, Tost M, et al. Origin of pancreatic ductal adenocarcinoma from atypical flat lesions: a comparative study in transgenic
mice and human tissues. J Pathol. 2012;226(5):723-734.
35. Morris JP IV, Wang SC, Hebrok M. KRAS, Hedgehog, Wnt and the
twisted developmental biology of pancreatic ductal adenocarcinoma. Nat
Rev Cancer. 2010;10(10):683-695.
36. Shi G, DiRenzo D, Qu C, et al. Maintenance of acinar cell organization
is critical to preventing Kras-induced acinar-ductal metaplasia. Oncogene.
2013;32(15):1950-1958.
37. Wood LD, Klimstra DS. Pathology and genetics of pancreatic neoplasms with acinar differentiation. Semin Diagn Pathol. 2014;31(6):491-497.
38. Jones M, Zheng Z, Wang J, et al. Impact of next-generation sequencing on the clinical diagnosis of pancreatic cysts. Gastrointest Endosc.
2016;83(1):140-148.
39. Schempf U, Sipos B, Konig C, et al. FOLFIRINOX as first-line treatment for unresectable acinar cell carcinoma of the pancreas: a case report.
Zeitschrift Gastroenterol. 2014;52(2):200-203.
40. Mellon EA, Hoffe SE, Springett GM, et al. Long-term outcomes of
induction chemotherapy and neoadjuvant stereotactic body radiotherapy
for borderline resectable and locally advanced pancreatic adenocarcinoma.
Acta Oncol. 2015;54(7):979-985.
41. National Comprehensive Cancer Network. NCCN Clinical Practice
Guidelines in Oncology: Pancreatic Adenocarcinoma. v I.2016. https://
www.nccn.org/professionals/physician_gls/pdf/pancreatic.pdf. Accessed
August 9, 2016.
42. Herman JM, Chang DT, Goodman KA, et al. Phase 2 multi-institutional trial evaluating gemcitabine and stereotactic body radiotherapy for
patients with locally advanced unresectable pancreatic adenocarcinoma.
Cancer. 2015;121(7):1128-1137.
454 Cancer Control
October 2016, Vol. 23, No. 4
Special Report
A Single-Institution Study of Demographics and Outcomes of
Adult Patients With Multiple Cancers
Thanh P. Ho, MD, Nathan R. Foster, MS, and Aminah Jatoi, MD
Background: Few studies have described the demographics and outcomes in patients with multiple cancers.
Methods: We reviewed the health records of patients with at least 3 invasive solid-tumor malignancies
(lung, colorectal, pancreatic, prostate, breast, or ovarian) diagnosed between 1995 and 2014 at our institution.
Results: A total of 163 patients were identified. The median age at first cancer diagnosis was 61 years.
Sixteen (10%) had a family history of cancer, and 7 (4%) had a cancer-predisposing syndrome. Most had
early-stage malignancies and underwent a series of surgical resections. The median survival from the first cancer diagnosis was 14 years (95% confidence interval: 12–16). Forty-three percent developed 4 or more cancers.
In univariate analyses, older age, male sex, smoking history, concurrent cancer, and stage IV disease were
associated with worse survival; by contrast, breast cancer as the first cancer diagnosis yielded better survival.
In multivariate analyses, older age, smoking history, and stage IV disease were associated with worse survival.
Conclusions: A small group of patients with no ostensible risk factors develop multiple early-stage cancers, receive
curative therapy (often surgery), and live for years. These patients merit further study to ascertain the broader prevalence rates of individuals with multiple invasive cancers and to better understand factors that contribute to cancer risk.
Introduction
Adult patients with cancer are living longer.1 Zeng et al2
reported on patients with cancer aged 50 to 64 years
at the time of their diagnosis between 2005 and 2009
and observed that, when compared with similar patients diagnosed during an earlier time interval, the
hazard ratios (HRs) for patients with colorectal, breast,
liver, and prostate cancers were 0.57 (95% confidence
interval [CI]: 0.55–0.60), 0.48 (95% CI: 0.45–0.51), 0.61
(95% CI: 0.57–0.69), and 0.32 (95% CI: 0.30–0.36), respectively. These findings suggest that patients with
cancer are living longer and that cancer can, at times,
be viewed as a long-term disease.2
Few studies have focused in general terms on
patients who live for many years following a first diagnosis of cancer and then continue to be diagnosed
with multiple cancers over time.3,4 Previous studies
have described multiple cancers in adult patients with
predisposing syndromes, showing, for example, that
individuals with Lynch syndrome or BRCA1/BRCA2
mutations have a more favorable prognosis after cancer diagnoses.5,6 Others have described patients with
therapy-related malignancies who have worse progFrom the Departments of Medicine (TPH), Health Sciences Research
(NRF), and Oncology (AJ), Mayo Clinic, Rochester, Minnesota.
Submitted March 10, 2016; accepted May 3, 2016.
Address correspondence to Aminah Jatoi, MD, Department of Oncology,
Mayor Clinic, 200 First Street SW, Rochester, Minnesota 55905.
E-mail: [email protected]
No significant relationships exist between the authors and the companies/organizations whose products or services may be referenced
in this article.
October 2016, Vol. 23, No. 4
noses compared with their counterparts who develop
de novo cancers.7,8 However, to our knowledge, these
studies and others have not explored the presence of
multiple cancers in the absence of predisposing syndromes or therapy-related secondary cancers.
Purpose
This study explored demographics and outcomes in
patients who developed multiple cancers. Some patients contend with multiple malignancies throughout their lives, thus displaying a poorly characterized
predisposition to cancer and a need for ongoing cancer surveillance. Reporting on the characteristics and
outcomes of patients with multiple cancers might help
oncologists better counsel patients about prognoses
and realistic treatment goals in the setting of multiple
malignancies.
Methods
Overview
This single-institution study focused on patients
with cancer seen at the Mayo Clinic (Rochester,
MN) from 1995 to 2014, encompassing a 20-year
interval that saw national improvements in cancer
outcomes.2 The Mayo Clinic Tumor Registry, which
relied in part on International Classification of
Diseases codes, pathology reports, and other clinical
sources to input data, was searched for patients who
met study criteria.
Eligible patients had to be diagnosed between
1995 and 2014 with at least 3 invasive malignancies of
at least 1 of the following types: lung, colorectal, panCancer Control 455
creatic, prostate, breast, or ovarian. These malignancies were chosen because they are among the most
common and lethal in the United States and have
5-year relative survival rates of 16%, 59%, 7%, 95%,
85%, and 41%.9 Inclusion criteria were determined a
priori. No major changes occurred in the Mayo Clinic
Tumor Registry or in institutional infrastructure within
the interval above; hence, data retrieval uniformly occurred over time. The Institutional Review Board at the
Mayo Clinic approved this study.
Medical Record Review
Each patient’s health records were reviewed and abstracted for date of birth, sex, date of each cancer diagnosis, stage of each cancer, smoking history, family
history of cancer (cancer in ≥ 2 first-degree relatives),
documentation of genetic predisposition to cancer,
cancer treatment, vital status, and date of death or last
follow-up visit. Because definitions of predisposing
syndromes to cancer have evolved over time to include
clinical criteria and, in some instances, molecular criteria, the methodology congruent with a real-time contemporary standard was used in this study. If a date
was not clearly documented, then a mid-month or midyear date was used. If the health record lacked clear
documentation of an item, then an unknown category
was used.
Data Analyses
Results are reported with means and percentages;
all analyses are considered exploratory. Because patients were heavily selected based on 6 targeted cancer types, this study was not designed to report on
the prevalence rates of patients with multiple cancers.
Kaplan-Meier curves were constructed to assess survival, and data were censored as appropriate. Cox proportional hazard models were used to explore whether
certain clinical variables were associated with survival.
Variables were chosen based on their clinical relevance
to cancer outcomes and based on a desire for parsimony to ensure stability of statistical models; in this
context, age at cancer diagnosis, sex, smoking history,
concurrent cancer diagnosis, stage IV disease (any malignancy), and cancer type at first diagnosis were chosen. HRs and 95% CIs were reported. A 2-tailed P value
of less than .05 was considered statistically significant.
All analyses were performed with JMP Statistical Software (SAS Institute, Carey, NC).
Results
Baseline Demographics
Nine patients were excluded because of lack of clarity
as to whether 1 or more of their cancers represented
a new malignancy, a metastatic site, or recurrence of
a previous cancer. The remaining 163 patients are the
focus of this report.
456 Cancer Control
The median age of the study participants was
61 years (range, 24–87). Sixteen (10%) had cancer in at
least 2 first-degree relatives, and 7 (4%) had a known
syndrome that predisposed them to cancer. Other
patient demographics appear in Table 1.
Cancer Characteristics
Cancer of the breast, colorectum, and lung were the
3 most common cancer types in the study group
(Table 2). Seventy patients (43%) had more than 3 cancers. In 10 patients, the first cancer diagnosis did not
fall within the 1995 to 2014 time interval, but these
patients were eligible for the study because they developed 3 invasive cancers during this interval. The
median interval from the first ever to the second cancer diagnosis was 2.6 years (range, 0–45), and the
median interval from the first ever to the third cancer diagnosis was 6.2 years (range, 0–58; Fig 1). Most
patients had relatively early-stage malignancies, and
the majority also underwent surgical treatment for all
3 cancers. Fewer than one-half of these specific cancers were treated with chemotherapy or radiotherapy.
Table 1. — Characteristics of Study Patients (N = 163)
Characteristic
Study Patients,
n (%)
Median age at first cancer diagnosis, y (range)
61 (24–87)
Sex
Male
Female
75 (46)
88 (54)
Cancer in ≥ 2 first-degree relatives
Yes
No
16 (10)
144 (88)
Genetic syndrome
Yes
No
Unknown
7 (4)
12 (7)
144 (88)
Smoking history
Yes
No
118 (74)
42 (26)
Concurrent cancer
Yes, 1
Yes, 2
No
13 (8)
17 (10)
133 (82)
≥ 4 cancers a
Yes
No
70 (43)
93 (57)
Percentages may not sum to 100% because of rounding or missing data.
aOther cancers included multiple cases of colorectal cancer, multiple
cases of lung cancer, multiple cases of breast cancer, 6 cases of
lymphoma, 5 cases of bladder cancer, 4 cases of melanoma, 4 cases
of prostate cancer, 3 cases of head and neck cancers, 2 cases of small
bowel cancer, 2 cases of ureteral cancer, 2 cases of ovarian cancer, and
1 each of chronic myelogenous and lymphocytic leukemias, and tubal,
esophageal, pancreatic, testicular, and cervical cancers.
October 2016, Vol. 23, No. 4
Table 2. — Cancer Diagnoses Among Study Patients (N = 163)
Diagnosis
Study Patients,
n (%)
Diagnosis
First Cancer
Cancer Type
Breast
Colorectal
Lung
Ovarian
Pancreas
Prostate
Other
Study Patients,
n (%)
Diagnosis
Second Cancer
43 (26)
40 (25)
38 (23)
3 (2)
1 (1)
26 (16)
12 (7)
Cancer Type
Breast
Colorectal
Lung
Ovarian
Pancreas
Prostate
Other
Stage
I
II
III
IV
72 (47)
43 (28)
31 (20)
7 (5)
Surgery
Yes
No
Study Patients,
n (%)
Third Cancer
39 (24)
52 (32)
55 (34)
0
1 (1)
13 (8)
3 (2)
Cancer Type
Breast
Colorectal
Lung
Ovarian
Pancreas
Prostate
Other
28 (17)
44 (27)
78 (48)
1 (1)
3 (2)
6 (4)
3 (2)
Stage
I
II
III
IV
87 (55)
40 (25)
26 (16)
6 (4)
Stage
I
II
III
IV
80 (55)
30 (21)
19 (13)
17 (12)
147 (90)
15 (9)
Surgery
Yes
No
145 (90)
17 (10)
Surgery
Yes
No
126 (78)
36 (22)
Systemic Therapy
Yes
No
69 (43)
93 (57)
Systemic Therapy
Yes
No
64 (40)
97 (60)
Systemic Therapy
Yes
No
62 (39)
99 (61)
Radiotherapy
Yes
No
53 (33)
107 (66)
Radiotherapy
Yes
No
34 (21)
126 (78)
Radiotherapy
Yes
No
42 (26)
117 (73)
A
B
50
60
50
40
40
Years
Years
30
20
30
20
10
10
0
0
No. of Patients
No. of Patients
Fig 1A–B. — Time interval between cancer diagnoses. These bar graphs show time on the vertical axis and each bar is representative of the number of
patients. (A) The interval from the first to the second cancer diagnosis was 2.6 years (range, 0–45 years). (B) The median interval from the first to the
third cancer was 6.2 years (range, 0–58 years).
October 2016, Vol. 23, No. 4
Cancer Control 457
Patients Alive, %
Cancer Outcomes
of cancer, and 2 had a documented genetic predispoA total of 92 patients (56%) had died. The median folsition to cancer. The median age at first cancer dilow-up of patients still living was 11.4 years (range,
agnosis was 64 years (range, 33–80). Thirty-one pa0.3–58.0). In 62 (67%) of the deceased patients, the
tients (74%) were diagnosed with stage I disease. The
cause of death was cancer; in 6 (7%), a nonmalignant
remaining stage distribution included 5 with stage II,
cause was determined; and, in 24 (26%), the cause was
5 with stage III, 1 with stage IV, and 1 whose stage
unknown. The overall median survival for the entire
was unknown. A total of 41 patients (95%) were treatgroup was 14 years from the time of first cancer diaged with surgery for the first cancer, 26 (60%) were
nosis (95% CI: 12–16; Fig 2).
treated with chemotherapy for the first cancer, and
In univariate analyses, older age, male sex, a
25 (58%) were treated with radiotherapy for the first
smoking history, a concurrent
cancer diagnosis, and stage
100
IV disease among any of the
3 malignancies yielded a
statistically significant asso80
ciation with worse survival
(Table 3). Breast cancer as the
first diagnosis of cancer was
60
associated with improved survival. However, in multivariate analyses, older age, smoking history, and any diagnosis
40
of stage IV disease yielded a
statistically significant association with worse survival
20
(see Table 3).
Among the 43 patients
who had breast cancer as
0
their first cancer, all were
0
2
4
6
8
10
12
14
16
18
20
22
24
women. A total of 29 (69%)
Years From First Cancer Diagnosis
had a smoking history (med ian of 35 pack-years),
Fig 2. — The overall median survival was 14 years. The 95% confidence intervals of 12 and 16 years are
5 (12%) had a family history
shown as the parallel dotted lines.
Table 3. — Results of the Cox Proportional Hazards Models
Univariate Analyses
(N = 163, n = 92 deaths)
Variable
HR (95% CI)
P Valueb
Multivariate Analyses
(n = 136, n = 72 deaths)a
P Valueb
HR (95% CI)
Age (1-y increase)
1.06 (1.04–1.09)
< .0001
1.09 (1.06–1.13)
< .0001
Sex (male vs female)
1.99 (1.31–3.04)
.0010
0.92 (0.47–1.82)
.8020
1.79 (1.07–3.19)
.0240
2.53 (1.36–5.04)
.0020
1.98 (1.13–3.28)
.0190
1.54 (0.76–2.90)
.2180
.0240
2.02 (1.09–3.65)
Smoking history (yes vs no)
c
Concurrent cancers (yes vs no)
Stage IV (vs not stage
IV)d
First diagnosis of cancer
Breast
Colorectal
Lung
Other (ref)
1.84 (1.09–3.02)
.0040
0.48 (0.25–0.87)
1.43 (0.83–2.43)
1.06 (0.60–1.83)
—
.0250
.4300
0.75 (0.31–1.84)
0.96 (0.46–1.95)
1.42 (0.69–2.87)
—
aThe
multivariate model included 72 deaths as a result of missing data. Separate models that included select variables above yielded similar findings,
thus demonstrating the stability of the model.
bLikelihood-ratio P value.
c Missing 3 observations and 2 events (n = 160, n = 90 events).
d Missing 24 observations and 18 events (n = 139, n = 74 events).
CI = confidence interval, HR = hazard ratio.
458 Cancer Control
October 2016, Vol. 23, No. 4
cancer. Second cancers within this group included
breast cancer in 25 (58%), colorectal cancer in 9 (21%),
and lung cancer in 9 (21%). Third cancers included
breast cancer in 16 (37%), colorectal cancer in 6 (14%),
lung cancer in 18 (42%), ovarian cancer in 1 (2%), pancreatic cancer in 1 (2%), and another type of cancer in
1 (2%). Twenty-eight patients remain alive at the time
of this report; the rate of overall median survival was
16 years. Among the 15 deceased patients, the cause of
death was related to cancer in 9 cases.
Discussion
This study focused on patients diagnosed with 3 or
more common, invasive, and sometimes lethal malignancies. It was not intended to report on the prevalence
rates of patients with multiple cancers because our eligibility criteria were deliberatively selective; however,
the scenario of multiple cancers appears to be rare, particularly when considering the number of cancer diagnoses every year at our institution — 868 patients were
diagnosed with breast cancer in 2014 at the Mayo Clinic
alone (unpublished data) — and when acknowledging
that cancer remains a major cause of death around the
world.10 We have identified a small cohort of patients
who illustrate this clinical scenario. This study contributes to a small but growing body of literature that
reports on the development of multiple cancers in a
group of patients who have not been specifically selected based on a genetic or otherwise known predisposition for having a higher risk of cancer.4,11
Despite the presumed rarity of this clinical scenario, this study provides important insights. These
patients often develop an early-stage malignancy, undergo surgical resection, and then live for many years
prior to developing a subsequent malignancy that can
be treated in a similar fashion. The median survival for
the study group exceeded 10 years, underscoring the
fact that this subgroup is treated and then re-treated for
a new cancer over time. Thus, a history of more than
1 early-stage cancer should not negatively impact the
management of subsequent early-stage cancers. In addition, although comparisons across data sets are perilous and, although our sample size is too small to allow for formal adjusted comparisons across data sets,
when we examined patients who were first diagnosed
with breast cancer and then developed multiple malignances thereafter, we did not observe major differences in survival based on benchmark data.9 For example,
the latter shows that 88% of patients with breast cancer reached the 5-year mark,9 whereas a survival curve
from patients with breast cancer in our study shows
that more than 90% did the same (data not shown).
Risk factors for early death remain relevant to patients with multiple cancers. Older age, smoking, and
stage IV disease are all associated with poor survival
in patients with multiple cancers. These prognostic facOctober 2016, Vol. 23, No. 4
tors remain important regardless of whether a patient
has been diagnosed with 1 cancer type or several.
More than 40% of the patients in our study were
diagnosed with 4 or more cancers. Although old age
is a risk factor for cancer, the patients in the current
study were not elderly when diagnosed with their first
malignancy (median age, 61 years). Moreover, a small
percentage had a known syndrome that predisposed
them to cancer or a compelling reason to suspect that
their history of recurrent cancers was the result of
prior cancer therapy, although our understanding of
genetic predisposition to cancer has grown over the
years. However, many of these patients were treated
surgically — a therapy not generally known to spawn
secondary cancers. Thus, perhaps other, undiscovered
factors are at play to explain the development of multiple cancers. This potential for uncharacterized risk factors for cancer — including genetic factors — requires
further investigation.
Limitations
The retrospective design of our study is a limitation and
led to missing data in some instances, as is the case in
any retrospective study. However, by using a retrospective design, we were able to assess 20 years of data within a short time frame. Another limitation is our small
sample size, which is the result of our single-institution
study design. However, our single-institution focus enabled us to acquire detailed information on the dates,
cancer stage, cancer therapy, and, in some cases, causes
of death. The single-institution nature of our study may
have also allowed for a greater degree of homogeneity
among data elements, thereby enabling us to identify
conclusions more readily.
Conclusions
Patients diagnosed with multiple cancers may continue
to live for years. To our knowledge, little insight exists
in the literature on the attitudes of health care professionals toward patients with a history of multiple malignancies, and repeated cancer diagnoses followed by
a cumulative history of cancer treatment could conceivably lead to a biased approach to repeat cancer therapy
over time. The findings from this study underscore the
need to continue to try to apply curative therapy to patients with a history of multiple cancers, regardless of
whether these cancers are primary, secondary, or tertiary. These findings also suggest the need to further
investigate the prevalence and risk factors for the development of multiple cancers.
References
1. American Cancer Society. Cancer Facts and Figures 2016.
http://w w w.cancer.org/acs/groups/content /@research/documents/
document/acspc-047079.pdf. Accessed September 2, 2016.
2. Zeng C, Wen W, Morgans AK, et al. Disparities by race, age, and
sex in the improvement of survival for major cancers: results from the NaCancer Control 459
tional Cancer Institute Surveillance, Epidemiology, and End Results (SEER)
program in the United States, 1990 to 2010. JAMA Oncol. 2015;1(1):88-96.
3. Utada M, Ohno Y, Hori M, et al. Incidence of multiple primary
cancers and interval between first and second primary cancers. Cancer Sci.
2014;105(7): 890-896.
4. Xu LL, Gu KS. Clinical retrospective analysis of cases with multiple
primary malignant neoplasms. Genet Mol Res. 2014;13(4):9271-9284.
5. Van den Broek AJ, Schmidt MK, van ’t Veer LJ, et al. Worse breast
cancer prognosis of BRCA1/BRCA2 mutation carriers: what’s the evidence?
A systematic review with meta-analysis. PLoS One. 2015;10(3):e0120189.
6. Gryfe R, Kim H, Hsieh ET, et al. Tumor microsatellite instability and
clinical outcome in young patients with colorectal cancer. N Engl J Med.
2000;342(2):69-77.
7. Travis LB. The epidemiology of second primary cancers. Cancer
Epidemiol Biomark Prev. 2006;15(11):2020-2026.
8. Travis LB, Fosså SD, Schonfeld SJ, et al. Second cancers among
40,576 testicular cancer patients: focus on long-term survivors. J Natl Cancer
Inst. 2005;97(18):1354-1365.
9. US Cancer Statistics Working Group. United States Cancer Statistics:
1999–2013 Incidence and Mortality Web-Based Report. Atlanta, GA:
US Department of Health and Human Services; Centers for Disease Control
and Prevention; National Cancer Institute; 2016. https://nccd.cdc.gov/uscs
/Survival/Relative_Survival_Tables.pdf. Accessed September 2, 2016.
10. World Health Organization. The top 10 causes of death. http://www.
who.int/mediacentre/factsheets/fs310/en/index2.html. Accessed September 13, 2016.
11. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer
J Clin. 2016;66(1):7-30.
460 Cancer Control
October 2016, Vol. 23, No. 4
Ten Best Readings Relating to
Health Disparities in Oncology
Sharma P, Ashouri K, Zargar-Shoshtari K, et al.
Racial and economic disparities in the treatment of penile squamous cell carcinoma: results
from the National Cancer Database. Urol Oncol.
2016;34(3):122.e9-15.
Researchers evaluated overall survival (OS) differences in economics and sociodemographics and
economic differences among 5,412 study patients
with penile squamous cell carcinoma. Survival did
not change over the study period, but the authors
found that black patients had worse median OS and
presented with a more advanced stage of disease.
Those who had median incomes of at least $63,000
and carried private insurance had better median OS
and presented with a lower stage of disease. Thus,
the researchers determined that racial and economic
differences exist in the OS rates of patients with penile
squamous cell carcinoma.
Langford AT, Resnicow K, Dimond EP, et al. Racial/
ethnic differences in clinical trial enrollment, refusal rates, ineligibility, and reasons for decline
among patients at sites in the National Cancer
Institute’s Community Cancer Centers Program.
Cancer. 2014;120(6):877-884.
Racial and ethnic differences were studied among
patients who had enrolled in, refused to enroll, were
ineligible to enroll, or wanted to participate in a clinical trial enrollment. Age 65 years or older, male sex,
and non-Hispanic black race were significantly associated with more medical or physical conditions. Those
who were aged 65 years or older were significantly
more likely to have low clinical trial enrollment; by
contrast, being male was significantly associated with
low refusal rates. The authors concluded that better
management of physical or medical conditions before
and during treatment might increase the pool of patients eligible to enroll in clinical trials.
Spratt DE, Chan T, Waldron L, et al. Racial/ethnic
disparities in genomic sequencing. JAMA Oncol.
2016;2(8):1070-1074.
A retrospective review of individual patient data
was conducted to determine the racial distribution
among samples sequenced within The Cancer Genome Atlas (TCGA) and the number of samples still
needed to detect select mutational frequencies in racial minorities. Of the 5,729 samples analyzed, 77%
were white, 12% were black, 3% were Asian, 3% were
Hispanic, and less than 0.5% were Native Americans.
These percentages, thus, over-represent whites comOctober 2016, Vol. 23, No. 4
pared with the US population and under-represent
Asians and Hispanics. All tumor types from whites
contained enough samples to detect a 10% mutational
frequency; however, group-specific mutations with
a 10% frequency were detectable for black patients
with breast cancer alone. It is probable but not well
understood that ethnic diversity may be related to the
pathogenesis of cancer and may have an impact on
the generalizability of these study findings.
Dutil J, Golubeva VA, Pacheco-Torres AL, et al. The
spectrum of BRCA1 and BRCA2 alleles in Latin
America and the Caribbean: a clinical perspective.
Breast Cancer Res Treat. 2015;154(3):441-453.
A review of the medical literature reporting on
the prevalence and/or spectrum of BRCA1 and BRCA2
variants was conducted. A total of 4,835 individuals
from Latin America, the Caribbean, and Hispanics
from the United States were included, of which 167
unique pathogenic variants were reported. In unselected breast cancer cases, the prevalence ranged
from 1.2% to 27.1%. Some countries presented with
few recurrent pathogenic variants, whereas others
were characterized by diverse, nonrecurrent variants.
The proportion of BRCA pathogenic variants shared
between US Hispanics and Latin American populations was estimated to be 10.4%. Within Latin America
and the Caribbean, 8.2% of the BRCA variants were
present in more than 1 country. Countries with a
high prevalence of BRCA pathogenic variants may
benefit from more aggressive testing strategies, and
testing of recurrent variant panels might present a
cost-effective solution for improving genetic testing
in some countries.
Weitzel JN, Clague J, Martir-Negron A, et al. Prevalence and type of BRCA mutations in Hispanics
undergoing genetic cancer risk assessment in the
southwestern United States: a report from the
Clinical Cancer Genetics Community Research
Network. J Clin Oncol. 2013;31(2):210-216.
Researchers sought to determine the prevalence
and type of BRCA1 and BRCA2 mutations among Hispanics in the southwestern United States and their
potential impact on genetic cancer risk assessment.
Deleterious BRCA mutations were detected in 25% of
746 samples. A total of 11% of 189 samples were large
rearrangement mutations, of which 62% were BRCA1
exon 9 to exon 12 deletions. Nine recurrent mutations
accounted for 53% of the total. Among these, BRCA1
exon 9 to exon 12 deletions appear be a Mexican
Cancer Control 461
founder mutation representing up to 12% of all BRCA1
mutations in select cohorts. The authors conclude that
the high frequency of large rearrangement mutations
warrants screening in every case.
Meyskens FL Jr, Mukhtar H, Rock CL, et al. Cancer
prevention: obstacles, challenges and the road
ahead. J Natl Cancer Inst. 2015;108(2):pii:djv309.
The genetic heterogeneity and complexity of advanced cancers support rationale for interrupting the
carcinogenic process early and target prevention to
reduce the burden of cancer; however, the focus of
cancer prevention should be on persons at high risk
and on primary localized disease, for which screening
and detection should also play a role. The timing and
doses of chemopreventive interventions also affect
response. The intervention may be ineffective if the
target population is very high risk or presents with
preneoplastic lesions and irreversible cellular changes.
The field must begin to focus on targeted organ-site
prevention approaches in patients at high risk. The
authors also suggest that comparative effectiveness research designs and the value of information obtained
from large-scale prevention studies are necessary so
that preventive interventions become a routine part
of cancer management.
Carlson RW, Scavone JL, Koh WJ, et al. NCCN framework for resource stratification: a framework for
providing and improving global quality oncology
care. J Natl Compr Canc Netw. 2016;14(8):961-969.
This article describes the rationale for resourcestratified guidelines and the methodology for developing the National Comprehensive Cancer Network
(NCCN) Framework. Disparities in available resources
for cancer care are enormous, which is one reason
why the NCCN developed a framework for stratifying
its clinical practice for cancer. NCCN also hopes to
help health care systems with varying levels of available resources provide optimal care for patients with
cancer. The framework is modified from a method
developed by the Breast Health Global Initiative. The
NCCN Framework for Resource Stratification identifies
4 resource environments and presents the recommendations in a graphic format that maintains the context
of the NCCN guidelines.
DeSantis CE, Siegel RL, Sauer AG, et al. Cancer
statistics for African Americans, 2016: progress
and opportunities in reducing racial disparities.
CA Cancer J Clin. 2016;66(4):290-308.
Approximately 189,910 new cases of cancer and
69,410 cancer-related deaths will occur among blacks
in 2016. Although blacks continue to have higher rates
of cancer-related mortality than whites, the disparity
has narrowed for all cancers combined in men and
462 Cancer Control
women and for lung and prostate cancers in men.
The racial gap in death rates has widened for breast
cancer in women and remained stable for colorectal
cancer in men. In men, incidence rates from 2003 to
2012 decreased for all cancers combined as well as for
the top 3 cancer sites. In women, overall rates during
the corresponding time period remained unchanged,
reflecting increasing trends in breast cancer combined
with decreasing trends in lung and colorectal cancer
rates. The 5-year relative survival rate is lower for
blacks than whites for most cancers at each stage of
diagnosis. The extent to which these disparities reflect
unequal access to health care vs other factors remains
an active area of research.
Ward E, Jemal A, Cokkinides V, et al. Cancer disparities by race/ethnicity and socioeconomic status.
CA Cancer J Clin. 2004;54(2):78-93.
The authors sought to identify disparities in cancer incidence, mortality, and survival rates in relation to race and ethnicity. For all cancer sites combined, residents of poorer counties had 13% higher
death rates from cancer in men and 3% higher rates
in women when compared with individuals living in
more affluent counties. Among both sexes, the 5-year
survival rate for all cancers combined was 10% lower
among persons who live in poorer rather than more
affluent census tracts. When the census tract poverty
rate was accounted for, African American, American
Indian, Alaskan Native, and Asian/Pacific Islander men
and African American, American Indian, and Alaskan
Native women had lower 5-year survival rates than
non-Hispanic whites. Detailed analyses of select cancers showed large variations in rate of cancer survival
by race and ethnicity.
Quinn GP, Sanchez JA, Sutton SK, et al. Cancer and
lesbian, gay, bisexual, transgender/transsexual,
and queer/questioning (LGBTQ) populations.
CA Cancer J Clin. 2015;65(5):384-400.
The authors review the literature on 7 cancer
sites that may disproportionately affect lesbian, gay,
bisexual, transgender/trans-sexual, and queer/questioning populations. For each cancer site, descriptive statistics, primary prevention, secondary prevention and preclinical disease, tertiary prevention and
late-stage disease, and clinical implications are discussed. The authors also provide an overview of psychosocial factors related to cancer survivorship as well
as strategies for improving access to care.
October 2016, Vol. 23, No. 4
Peer Reviewers, 2016
Peer review is the indispensable and critical element in the evaluation of all manuscripts being considered for
publication in our journal. The oncology professionals who took part in our peer review process this past year are
listed below, and we thank them for their invaluable efforts on behalf of Cancer Control.
— The Editor
External Reviewers
Abate, Getahun, MD, PhD
Saint Louis University
Alberg, Anthony J., PhD, MPH
Medical University of South Carolina
Alcaraz, Kassandra, PhD, MPH
American Cancer Society
Ahmad, Nazeel, MD
James A. Haley Veterans Administration
Medical Center
Allott, Emma H., PhD
University of North Carolina at Chapel Hill
Alrabaa, Sally, MBBS
University of South Florida
Apata, Jummai, MBBS, MPH
Morgan State University
Bao, Renyue, MD
University of Houston
Biermann, Martin, MD
University of Bergen, Norway
Bowen, Deborah J., PhD
University of Washington
Briere, Elizabeth C., MD, MPH
Centers for Disease Control and
Prevention
Brierley, James, D., MB
Princess Margaret Cancer Centre
Bull, Joan M.C., MD
University of Texas Health Science Center at Houston
Chevez-Barrios, Patricia, MD
Houston Methodist
Chowdary, Sajeel, MD
Boca Raton Regional Hospital
Climent, Consuelo, MD
University of Puerto Rico
Colon-Otero, Gerardo, MD
Mayo Clinic
Dagan, Roi, MD
University of Florida
DeGregorio, Richard, MD
Surgical Pathology Laboratories
October 2016, Vol. 23, No. 4
Deneve, Jeremiah L., MD
University of Tennessee Health
Science Center
Dickson, Paxton, MD
University of Tennessee Health
Sciences Center
Drucker, Mitchell D., MD
University of South Florida
Eisbruch, Avraham, MD
University of Michigan
Friedman, Scott, MD
Mount Sinai Hospital
Glas, Martin, MD
Robert Janker Clinic
University of Bonn Medical Center
Glezerman, Ilya G., MD
Memorial Sloan Kettering
Cancer Center
Goovaerts, Pierre, PhD
BioMedware
Hara, Wendy, MD
Stanford University
Harbour, J. William, MD
Bascom Palmer Eye Institute
Hassanein, Ashraf, MD, PhD
Florida Dermatologic Surgery
& Aesthetics Institute
Florida Pathology
Hernandez, Brenda Y., PhD, MPH
University of Hawaii
Jatoi, Aminah, MD
Mayo Clinic
Jones, Lovell A., PhD
Prairie View A&M University College
of Nursing
Lichtman, Stuart, MD
Memorial Sloan Kettering
Luque, John, MD
Medical University of South Carolina
Mahtani, Reshma L., MD
University of Miami Health System
Maluccio, Mary A., MD, MPH
Indiana University School of Medicine
Margo, Curtis, MD
University of South Florida
Margolies, Laurie, MD
Mount Sinai School of Medicine
Montero, Jose, MD
University of South Florida
Moore, Charles E., MD
Emory University School of Medicine
Muro-Cacho, Carlos, MD
James A. Haley Veterans Hospital
Nevidjon, Brenda, MSN, RN
Oncology Nursing Society
Nguyen, Jennifer, PhD, MPH
University of Florida
Oehler, Richard, MD
James A. Haley Veterans
Administration Hospital
Pentz, Rebecca D., PhD
Emory School of Medicine
Phuphanich, Surasak, MD
Cedars-Sinai Medical Center
Razis, Evangelia, MD, PhD
Hygeia Hospital
Rigakos George, MD, MSc
Hygeia Hospital
Kamen, Charles, PhD
University of Rochester Medical Center
Rowe, Christopher, MD
University of Newcastle
Kanji, Jamil, MD
University of Alberta
Rusch, Valerie W., MD
Memorial Sloan Kettering
Cancer Center
Lavand’homme, Patricia, MD, PhD
Université Catholique de Louvain
LeRuhn, Emilie, MD
University Hospital and Oscar
Lambret Center
Saeed, Maythem, MD
University of California at San Francisco
Sheikhattari, Payam, MD, MPH
Morgan State University
Cancer Control 463
Peer Reviewers, 2016
Simmons, Melissa A., MD
Northwestern University
Sioutos, Panayiotis, MD
Hygeia Hospital
Sniffin, Jason, DO
Florida Hospital Orlando
Infectious Disease Consultants
Tang, Haidong, PhD
University of Texas
Southwestern Medical Center
Thompson, Haley, PhD
Barbara Ann Karmanos Cancer Institute
Wayne State University
Tyson, Mark D., MD
Mayo Clinic
Vander Poorten, Vincent, MD
Leuven Cancer Institute
Wayman, Laura L., MD
Vanderbilt University
Yom, Sue S., MD, PhD
University of California at San Francisco
Zhang, Paul J., MD
University of Pennsylvania
Internal Reviewers
(H. Lee Moffitt Cancer Center
& Research Institute)
Ahmad, Nazeel, MD
Arrington, John, MD
Balducci, Lodovico, MD
Caracciolo, Jamie, MD
Caudell, Jimmy J., MD, PhD
Chon, Hye Sook, MD
Clark, Terry J., MScA
Coppola, Domenico, MD
Dessureault, Sophie, MD
Dhillon, Jasreman, MD
Dossett, Lesly, MD
Green, B. Lee, PhD
Greene, John N., MD
Gwede, Clement K., PhD
Harrison, Louis B., MD
Hudson, Janella, PhD
Kumar, Nagi B., PhD
Laber, Damian A., MD
Lancet, Jeffrey, MD
Makanji, Rikesh, MD
Mullinax, John E., MD
Nanjappa, Sowmya, MD
Niell, Bethany, MD, PhD
Otto, Kristen, MD
Pasikhova, Yanina, PharmD
Pow-Sang, Julio M., MD
Quinn, Gwendolyn P., PhD
So, Wonhee, PharmD
Sutton, Steve K., PhD
Trotti, Andy, MD
Wang, Xia, MD
Zager, Jonathan S., MD
464 Cancer Control
October 2016, Vol. 23, No. 4
Index for 2016, Volume 23
Vol. 23, No. 1: Regional Therapeutics
3 Art, Science, and Personalized Cancer Care Through Regional Therapeutics
61Special Report: Genetic Investigation of Uterine Carcinosarcoma: Case Report and Cohort Analysis
Timothy N. Hembree, DO, PhD, Jamie K. Teer, PhD, Ardeshir Hakam, MD, and Alberto A. Chiappori, MD
Kristen Otto, MD, and Jonathan S. Zager, MD
6 Stereotactic Body Radiotherapy for
Recurrent Unresectable Head and Neck
Cancers
Tobin Strom, MD, Christian Wishka, and Jimmy J. Caudell, MD, PhD
12 Developments in Intralesional Therapy for Metastatic Melanoma
67Special Report: Advances in the Endoscopic Diagnosis of Barrett Esophagus
Ashley H. Davis-Yadley, MD, Kevin G. Neill, MD,
Mokenge P. Malafa, MD, and Luis R. Peña, MD
78Special Report: Association of microRNA 21 With Biological Features and Prognosis of Neuroblastoma
Yaodong Zhou, MD, and Bo Sheng, MD
Sarah Sloot, MD, Omar M. Rashid, MD, JD, Amod A. Sarnaik, MD, and Jonathan S. Zager, MD
21Stereotactic Body Radiotherapy in
the Management of Oligometastatic
Disease
Kamran A. Ahmed, MD, and Javier F. Torres-Roca, MD
Vol. 23, No. 2: Ocular Oncology
90Are Ocular and Ocular Adnexal Cancers Overdiagnosed? Historical Perspectives on Diagnosis
30Local Ablation for Solid Tumor Liver
Metastases: Techniques and Treatment
Efficacy
Curtis E. Margo, MD
93Assessing Prognosis in Uveal Melanoma
Joyce Wong, MD, and Amanda Cooper, MD
Zélia M. Corrêa, MD, PhD
36Hyperthermic Intraperitoneal Chemotherapy and Cytoreductive Surgery in the Management of Peritoneal Carcinomatosis
Rahul Rajeev, MBBS, and Kiran K. Turaga, MD
47Disparities Report: Racial Disparity in Time Between First Diagnosis and Initial Treatment of Prostate Cancer
Ballington L. Kinlock, PhD, Roland J. Thorpe Jr, PhD, Daniel L. Howard, PhD, Janice V. Bowie, PhD, Louie E. Ross, PhD, David O. Fakunle, and Thomas A. LaVeist, PhD
52Disparities Report: Exploring the Perceptions of Anal Cancer Screening and Behaviors Among Gay and Bisexual Men Infected
With HIV
Alexis M. Koskan, PhD, Natalie LeBlanc, MPH, and Isabella Rosa-Cunha, MD
99Therapeutic Options for Retinoblastoma
Pia R. Mendoza, MD, and Hans E. Grossniklaus, MD
110 Primary Vitreoretinal Lymphoma:
Management of Isolated Ocular Disease
Matthew T. Witmer, MD
117 Management of Primary Acquired Melanosis, Nevus, and Conjunctival Melanoma
Andrew Kao, MD, Armin Afshar, MD, Michele Bloomer, MD,
and Bertil Damato, MD, PhD
126Sebaceous Carcinoma of the Eyelid
Carlos Prieto-Granada, MD, and Paul Rodriguez-Waitkus, MD, PhD
133Role of Vismodegib in the Management of Advanced Periocular Basal Cell Carcinoma
Kyle F. Cox, MD, and Curtis E. Margo, MD
59Case Report: Hyperuricemia in 2 Patients Receiving Palbociclib for Breast Cancer
David J. Bromberg, MD, Mauricio Valenzuela, MD,
Sowmya Nanjappa, MD, and Smitha Pabbathi, MD
140Extranodal Marginal Zone B-cell Lymphoma of the Ocular Adnexa
October 2016, Vol. 23, No. 4
Jean Guffey Johnson, MD, Lauren A. Terpak, MS, Curtis E. Margo, MD, and Reza Setoodeh, MD
Cancer Control 465
Index for 2016, Volume 23
150Ophthalmic Complications Related to
Chemotherapy in Medically Complex Patients
213Clinical Benefits of Proton Beam Therapy for Tumors of the Skull Base
Lynn E. Harman, MD
157Disparities Report: Disparities Among Minority Women With Breast Cancer Living in
Impoverished Areas of California
Sundus Haji-Jama, Kevin M. Gorey, PhD, Isaac N.
Luginaah, PhD, Guangyong Zou, PhD, Caroline Hamm, MD, and Eric J. Holowaty, MD
163Infectious Disease Report: Bordetella pertussis Infection in Patients With Cancer
Abraham Yacoub, MD, Sowmya Nanjappa, MD,
Tyler Janz, and John N. Greene, MD
167Pharmacy Report: Megestrol Acetate-Induced Adrenal Insufficiency
Sowmya Nanjappa, MD, Christy Thai, PharmD,
Sweta Shah, MD, and Matthew Snyder, PharmD
170 Pathology Report: Presacral Noncommuni-
cating Enteric Duplication Cyst
Shabnam Seydafkan, MD, David Shibata, MD,
Julian Sanchez, MD, Nam D. Tran, MD, Marino Leon, MD, and Domenico Coppola, MD
175 Symposium Report: Second Annual Moffitt Anatomic Pathology Symposium
Vol. 23, No. 3: Head and Neck Cancers
192Maximizing Cures and Preserving Function in Head and Neck Cancers
Louis B. Harrison, MD, and Jimmy J. Caudell, MD, PhD
197Management of Oropharyngeal Cancer in the HPV Era
Arash O. Naghavi, MD, Tobin J. Strom, MD, Kamran A. Ahmed, MD, Michelle I. Echevarria, MD, Yazan A.
Abuodeh, MD, Puja S. Venkat, MD, Jessica M. Frakes, MD,
Louis B. Harrison, MD, Andy M. Trotti, MD, and Jimmy J. Caudell, MD, PhD
208Definitive Radiotherapy for Squamous Cell Carcinoma of the Glottic Larynx
William M. Mendenhall, MD, Roi Dagan, MD, Curtis M. Bryant, MD, Robert J. Amdur, MD, and Anthony A.
Mancuso, MD
466 Cancer Control
Kamran A. Ahmed, MD, Stephanie K. Demetriou,
Mark McDonald, MD, and Peter A.S. Johnstone, MD
220Management of BCC and SCC of the
Head and Neck
Tobin J. Strom, MD, Jimmy J. Caudell, MD, PhD, and Louis B. Harrison, MD
228Treatment of Head and Neck
Paragangliomas
Kenneth Hu, MD, and Mark S. Persky, MD
242Multidisciplinary Management of Salivary Gland Cancers
Matthew J. Mifsud, MD, Jon N. Burton, MD,
Andy M. Trotti, MD, and Tapan A. Padhya, MD
249Pathology Report: Practical Issues for
Retroperitoneal Sarcoma
Vicky Pham, MS, Evita Henderson-Jackson, MD,
Matthew P. Doepker, MD, Jamie T. Caracciolo, MD, Ricardo J. Gonzalez, MD, Mihaela Druta, MD, Yi Ding, MD, and Marilyn M. Bui, MD, PhD
265Infectious Diseases Report: Primary
Gangrenous Cutaneous Mold
Infections in a Patient With Cancer
and Neutropenia
Abraham Yacoub, MD, Kiran K. Soni, Lysenia Mojica, MD,
Jane Mai, MD, Jamie Morano, MD, C. Wayne Cruse, MD, Ramon L. Sandin, MD, Sowmya Nanjappa, MBBS, MD, Chandrashekar Bohra, MBBS, Ganesh Gajanan, MBBS, and John N. Greene, MD
272Case Series: Diffuse Alveolar Hemorrhage
in Acute Myeloid Leukemia
Sowmya Nanjappa, MBBS, MD, Daniel K. Jeong, MD, Manjunath Muddaraju, MD, Katherine Jeong, MD,
Eboné D. Hill, MD, and John N. Greene, MD
278Case Series: Marijuana Smoking in Patients With Leukemia
Sara Khwaja, MD, Abraham Yacoub, MD, Asima Cheema, MD, Nancy Rihana, MD, Robin Russo, MD, Ana Paula Velez, MD, Sowmya Nanjappa, MBBS, MD, Ramon L. Sandin, MD, Chandrashekar Bohra, MBBS, Ganesh Gajanan, MBBS, and John N. Greene, MD
October 2016, Vol. 23, No. 4
Index for 2016, Volume 23
284
Special Report: Platelet-to-Lymphocyte Ratio and Use of NSAIDs During the Perioperative Period as Prognostic Indicators in Patients With NSCLC Undergoing Surgery
Brenda M. Lee, MD, Andrea Rodríguez, MD, Gabriel Mena, MD, Vijaya Gottumukkala, MB, MD, Reza J.
Mehran, MD, David C. Rice, MB, Lei Feng, MS,
Jun Yu, MS, and Juan P. Cata, MD
295Special Report: Resection of the First Rib With Preservation of the T1 Nerve Root in Pancoast Tumors of the Lung
Andreas K. Filis, MD, Lary A. Robinson, MD, and
Frank D. Vrionis, MD, PhD
302Special Report: Feasibility, Preliminary
Efficacy, and Lessons Learned From a Garden-Based Lifestyle Intervention for
Cancer Survivors
Colleen K. Spees, PhD, RDN, Emily B. Hill, Elizabeth M.
Grainger, PhD, RDN, Jackie L. Buell, PhD, RDN,
Susan E. White, PhD, Matthew D. Kleinhenz, PhD, and Steven K. Clinton, MD, PhD
311 Special Report: Hypofractionated Stereotactic
Radiotherapy for Auditory Canal or Middle Ear Cancer
Taro Murai, MD, Shin-Etsu Kamata, MD, Kengo Sato, MD,
Kouki Miura, MD, Mitsuhiro Inoue, Naoki Yokota, MD, Seiji Ohta, MD, Michio Iwabuchi, MD, Hiromitsu Iwata, MD,
and Yuta Shibamoto, MD
Vol. 23, No. 4 Supplement: Lung Cancer
2 Best Practices in Treatment Selection for
Patients With Advanced NSCLC 2
Mark A. Socinski, MD, and Nathan Pennell, MD, PhD
Vol. 23, No. 4: Health Disparities in Oncology
327Barriers to Clinical Trial Enrollment in Racial and Ethnic Minority Patients With Cancer
Lauren M. Hamel, PhD, Louis A. Penner, PhD,
Terrance L. Albrecht, PhD, Elisabeth Heath, MD,
Clement K. Gwede, PhD, RN, and Susan Eggly, PhD
338Racial and Ethnic Differences in the
Epidemiology and Genomics of Lung Cancer
347Black Heterogeneity in Cancer Mortality:
US-Blacks, Haitians, and Jamaicans
Paulo S. Pinheiro, MD, PhD, Karen E. Callahan, MPH, Camille Ragin, PhD, MPH, Robert W. Hage, MD, PhD, DLO, MBA, Tara Hylton, MPH, and Erin N. Kobetz, PhD, MPH
359Genomic Disparities in Breast Cancer Among Latinas
Filipa Lynce, MD, Kristi D. Graves, PhD, Lina Jandorf, MA, Charité Ricker, MS, Eida Castro, PsyD, Laura Moreno, Bianca Augusto, Laura Fejerman, PhD, and Susan T. Vadaparampil, PhD
373Clinical Considerations of Risk, Incidence, and Outcomes of Breast Cancer in Sexual Minorities
Anne E. Mattingly, MD, John V. Kiluk, MD, and
M. Catherine Lee, MD
383Construction and Validation of a Multi Institutional Tissue Microarray of Invasive
Ductal Carcinoma From Racially and Ethnically Diverse Populations
Edward Seijo, MS, Diana Lima, MPH, Egiebade Iriabho, MS, Jonas Almeida, PhD, Jesus Monico, MPH, MS, Margarita Echeverri, PhD, Sylvia Gutierrez, MD, Idhaliz Flores, PhD, Ji-Hyun Lee, PhD, Kate Fisher, MA, William E. Grizzle, MD, PhD, Gabriel L. Sica, MD, PhD, Charles Butler, Chindo Hicks, PhD, Cathy D. Meade, PhD, RN, Stephen Olufemi Sodeke, PhD, Krzysztof Moroz, MD, Domenico Coppola, MD, and Teresita Muñoz-Antonia, PhD
390Determinants of Treatment Delays Among Underserved Hispanics With Lung and Head and Neck Cancers
Evelinn A. Borrayo, PhD, Katie L. Scott, PhD, Ava R. Drennen, PhD, Tiare MacDonald, PhD, and
Jennifer Nguyen, MS
401Geographical Factors Associated With Health Disparities in Prostate Cancer
Scott M. Gilbert, MD, Julio M. Pow-Sang, MD, and Hong Xiao, PhD
409Disparities in Penile Cancer
October 2016, Vol. 23, No. 4
Matthew B. Schabath, PhD, W. Douglas Cress, PhD, and Teresita Muñoz-Antonia, PhD
Pranav Sharma, MD, Kamran Zargar-Shoshtari, MD, Curtis A. Pettaway, MD, Matthew B. Schabath, PhD, Anna R. Giuliano, PhD, and Philippe E. Spiess, MD
Cancer Control 467
Index for 2016, Volume 23
415 Chemoprevention in African American Men With Prostate Cancer
Nagi B. Kumar, PhD, RD, Julio M. Pow-Sang, MD, Philippe E.
Spiess, MD, Jong Y. Park, PhD, Ganna Chornokur, PhD, Andrew R. Leone, MD, and Catherine M. Phelan, PhD, MD
424Disparities in Adolescents and Young Adults With Cancer
Leidy L. Isenalumhe, MD, Olivia Fridgen, MPH, Lynda K.
Beaupin, MD, Gwendolyn P. Quinn, PhD, and Damon R. Reed, MD
434Tobacco-Related Health Disparities Across the Cancer Care Continuum
Vani Nath Simmons, PhD, Bárbara Piñeiro, PhD, Monica Webb Hooper, PhD, Jhanelle E. Gray, MD, and Thomas H. Brandon, PhD
442 Case Series: Human Metapneumovirus
Infection in Immunocompromised Patients
Sharmeen Samuel, MD, Sowmya Nanjappa, MBBS, MD, Christopher D. Cooper, MD, and John N. Greene, MD
446Special Report: Systematic Review and Case Series Report of Acinar Cell Carcinoma of the Pancreas
Evan S. Glazer, MD, PhD, Kevin G. Neill, MD, Jessica M. Frakes, MD, Domenico Coppola, MD, Pamela J. Hodul, MD,
Sara E. Hoffe, MD, Jose M. Pimiento, MD, Gregory M. Springett, MD, PhD, and Mokenge P. Malafa, MD, PhD
455Special Report: A Single-Institution Study of Demographics and Outcomes of Adult Patients With Multiple Cancers
Thanh P. Ho, MD, Nathan R. Foster, MS, and Aminah Jatoi, MD
468 Cancer Control
October 2016, Vol. 23, No. 4
Current Perspectives in
Oncology Nursing
Save the Date! | Call for Abstracts
February 16–17, 2017
Moffitt Cancer Center, Tampa, FL
16th Annual Nursing Conference
Special Event
The 22nd Annual Rhinehart Reception & Lecture | February 15, 2017
Moffitt Cancer Center
Vincent A. Stabile Research Building
12902 Magnolia Drive, Tampa, FL 33612
(
For additional conference information please contact
Gina Woodward at 813-745-1741 or [email protected]
(