Download Vol. 23, No. 2 – Ocular Oncology

Document related concepts

Gene therapy of the human retina wikipedia , lookup

Index of oncology articles wikipedia , lookup

Transcript
cancercontroljournal.org Vol. 23, No. 2, April 2016
H. LEE MOFFITT CANCER CENTER & RESEARCH INSTITUTE, AN NCI COMPREHENSIVE CANCER CENTER
Assessing Prognosis in Uveal Melanoma
Zélia M. Corrêa, MD, PhD
Therapeutic Options for Retinoblastoma
Pia R. Mendoza, MD, and Hans E. Grossniklaus, MD
Primary Vitreoretinal Lymphoma: Management of
Isolated Ocular Disease
Matthew T. Witmer, MD
Management of Primary Acquired Melanosis, Nevus, and
Conjunctival Melanoma
Andrew Kao, MD, Armin Afshar, MD, Michele Bloomer, MD, et al
Sebaceous Carcinoma of the Eyelid
Carlos Prieto-Granada, MD, and Paul Rodriguez-Waitkus, MD, PhD
Role of Vismodegib in the Management of Advanced
Periocular Basal Cell Carcinoma
Kyle F. Cox, MD, and Curtis E. Margo, MD
Extranodal Marginal Zone B-cell Lymphoma of the
Ocular Adnexa
Jean Guffey Johnson, MD, Lauren A. Terpak, MS, Curtis E. Margo, MD, et al
Ophthalmic Complications Related to Chemotherapy in
Medically Complex Patients
Lynn E. Harman, MD
Cancer Control is included in
Index Medicus/MEDLINE
Jacksonville, FL
Permit No. 4390
H LEE MOFFITT CANCER CENTER
& RESEARCH INSTITUTE INC
12902 Magnolia Drive
Tampa FL 33612-9416
PAID
NON-PROFIT ORG
U.S. POSTAGE
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:
John Horton, MB, ChB
Professor Emeritus of Medicine & Oncology
Guest Editor:
Richard D. Kim, MD
Associate Member
Gastrointestinal Oncology
Bela Kis, MD, PhD
Assistant Member
Diagnostic Radiology
Rami Komrokji, MD
Associate Member
Malignant Hematology
Conor C. Lynch, PhD
Assistant Member
Tumor Biology
Kristen J. Otto, MD
Associate Member
Head and Neck and
Endocrine Oncology
Michael A. Poch, MD
Assistant Member
Genitourinary Oncology
Curtis E. Margo, MD
Professor
Departments of Pathology and Cell Biology
and Ophthalmology
Morsani College of Medicine
University of South Florida
Jeffery S. Russell, MD, PhD
Assistant Member
Endocrine Tumor Oncology
Moffitt Cancer Center
Journal Advisory Committee:
Saïd M. Sebti, PhD
Senior Member
Drug Discovery
Daniel A. Anaya, MD
Associate Member
Gastrointestinal Oncology
Aliyah Baluch, MD
Assistant Member
Infectious Diseases
Dung-Tsa Chen, PhD
Associate Member
Biostatistics
Elizabeth M. Sagatys, MD
Assistant Member
Pathology - Clinical
Bijal D. Shah, MD
Assistant Member
Malignant Hematology
Production Team:
Veronica Nemeth
Editorial Coordinator
Sherri Damlo
Medical Copy Editor
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]
Lubomir Sokol, MD, PhD
Senior Member
Hematology/Oncology
Hatem H. Soliman, MD
Assistant Member
Breast Oncology
Hey Sook Chon, MD
Assistant Member
Gynecological Oncology
Jonathan R. Strosberg, MD
Assistant Member
Gastrointestinal Oncology
Jasreman Dhillon, MD
Assistant Member
Pathology - Anatomic
Jennifer Swank, PharmD
Clinical Pharmacist Medical Oncology/Internal Medicine
Jennifer S. Drukteinis, MD
Associate Member
Diagnostic Radiology
Sarah W. Thirlwell, MSc, MSc(A), RN
Nurse Director
Supportive Care Medicine Program
Clement K. Gwede, PhD
Associate Member
Health Outcomes and Behavior
Eric M. Toloza, MD, PhD
Assistant Member
Thoracic Oncology
Sarah E. Hoffe, MD
Associate Member
Radiation Oncology
Nam D. Tran, MD
Assistant Member
Neuro-Oncology
John V. Kiluk, MD
Associate Member
Breast 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).
April 2016, Vol. 23, No. 2
Cancer Control 87
Table of Contents
Editorial
Are Ocular and Ocular Adnexal Cancers Overdiagnosed? Historical Perspectives on Diagnosis
90
Curtis E. Margo, MD
Articles
Assessing Prognosis in Uveal Melanoma
93
Zélia M. Corrêa, MD, PhD
Therapeutic Options for Retinoblastoma
99
Pia R. Mendoza, MD, and Hans E. Grossniklaus, MD
Primary Vitreoretinal Lymphoma: Management of Isolated Ocular Disease
110
Management of Primary Acquired Melanosis, Nevus, and Conjunctival Melanoma
117
Matthew T. Witmer, MD
Andrew Kao, MD, Armin Afshar, MD, Michele Bloomer, MD, and Bertil Damato, MD, PhD
Sebaceous Carcinoma of the Eyelid
126
Role of Vismodegib in the Management of Advanced Periocular
Basal Cell Carcinoma
133
Carlos Prieto-Granada, MD, and Paul Rodriguez-Waitkus, MD, PhD
Kyle F. Cox, MD, and Curtis E. Margo, MD
Extranodal Marginal Zone B-cell Lymphoma of the Ocular Adnexa
140
Jean Guffey Johnson, MD, Lauren A. Terpak, MS, Curtis E. Margo, MD,
and Reza Setoodeh, MD
88 Cancer Control
April 2016, Vol. 23, No. 2
Table of Contents
Articles, continued
Ophthalmic Complications Related to Chemotherapy in Medically Complex Patients
150
Lynn E. Harman, MD
Departments
Disparities Report: Disparities Among Minority Women With Breast Cancer Living in
Impoverished Areas of California
157
Infectious Disease Report: Bordetella pertussis Infection in Patients With Cancer 163
Sundus Haji-Jama, Kevin M. Gorey, PhD, Isaac N. Luginaah, PhD, Guangyong Zou, PhD, Caroline Hamm, MD,
and Eric J. Holowaty, MD
Abraham Yacoub, MD, Sowmya Nanjappa, MD, Tyler Janz, and John N. Greene, MD
Pharmacy Report: Megestrol Acetate-Induced Adrenal Insufficiency
167
Pathology Report: Presacral Noncommunicating Enteric Duplication Cyst
170
Sowmya Nanjappa, MD, Christy Thai, PharmD, Sweta Shah, MD, and Matthew Snyder, PharmD
Shabnam Seydafkan, MD, David Shibata, MD, Julian Sanchez, MD, Nam D. Tran, MD, Marino Leon, MD,
and Domenico Coppola, MD
Symposium Report: Second Annual Moffitt Anatomic Pathology Symposium
175
Ten Best Readings Relating to Ocular Oncology
187
About the art in this issue:
Lynn E. Harman, MD, is an ophthalmologist at the University of South Florida Morsani College of Medicine and the James A. Haley Veterans Affairs
Hospital in Tampa, Florida. She practices general ophthalmology and specializes in the diagnosis and treatment of glaucoma.
Dr Harman is an amateur photographer who enjoys her hobby when traveling. For the works included in this issue, she used a high-dynamicrange imaging technique which expands the breadth of color and luminosity that is typically captured on a single digital image.
Cover: Bernese Alps
Articles:
Virgin River Gorge
Themes at the Shard
Manhattan
Chicago River
Half Dome From Glacier Point
Hohenschwangau Castle
Los Angeles
Point Reyes Beach North
April 2016, Vol. 23, No. 2
Cancer Control 89
Editorial
Are Ocular and Ocular Adnexal Cancers Overdiagnosed?
Historical Perspectives on Diagnosis
Applying the term overdiagnosis to cancer reflects a
conceptual shift in thinking about cancer as a neoplastic proliferation based on histopathological criteria to
a disease that, if left untreated, becomes symptomatic
and results in morbidity or death.1-3 During the past
decade, select cancers — correctly diagnosed histologically as cancer — would have gone undetected
during a patient’s life (ie, remained clinically silent) if
it were not for screening programs. The most robust
evidence for overdiagnosis comes from randomized
clinical trials of cancer screening and observational
studies. For some cancers, the results of randomized
trials have revealed no compensatory decrease in
cause-specific mortality rates among a screened cohort
of patients.1 Observational studies complement these
findings, demonstrating a rapid rise in cancer diagnoses after screening programs are instituted without any
long-term change in cancer-specific mortality rates.3
The notion that early detection will lead to early
treatment and reduced rates of cancer-specific mortality is intuitively appealing. Although this ideal does
occur with some cancers like colon and cervical cancer, this is not universally the case. In industrialized
countries, the overdiagnosis phenomenon involves
mostly prostate, breast, renal, and thyroid cancers.1
Given that these seemingly counterintuitive findings
may be at odds with compelling anecdotes of success,
the discussion about cancer screening has become
controversial.4-7
Overdiagnosis arises because cancer screening uncovers a reservoir of indolent tumors that would have
otherwise gone unnoticed throughout life. Early detection of either nonprogressive or slow-growing cancer
gets caught up in this net, which is larger if precursor lesions and in situ neoplasia are factored into the
screening process. For select patients, death from other
causes will occur during the period of cancer inexpression. Because of tumor latencies and competing risks
of mortality, some patients screened for cancer are subject to the costs and adverse events of treatment without any prospect of benefit.
However, the conundrum of overdiagnosis involves applying this epidemiological perspective to individual patients. At the time of screening diagnosis,
it may be impossible to distinguish persons with indolent, nonprogressive lesions from those with potentially lethal cancers. The problem of overdiagnosis has
surfaced in ophthalmology before but not as the result
90 Cancer Control
of epidemiological sleuthing and certainly not in the
context of screening programs. Claims of overdiagnosis were raised because there was a sense of incongruity between the lesions observed under the microscope
and the commensurate treatments. A few examples illustrating this phenomenon are offered.
Beginning in the late 1930s, Reese8-11 solidified
the idea of conjunctival melanosis as a precursor lesion to conjunctival melanoma. He distinguished it
from congenital melanosis and termed it precancerous
melanosis.8-11 Acknowledging that its progression to
melanoma is often protracted, Reese8-11 estimated that
approximately 17% of cases of precancerous melanosis
become cancerous (ie, conjunctival melanoma) within
5 years and that conjunctiva melanoma had a mortality
rate of at least 40%.Based on clinical and pathological
observations, he favored orbital exenteration for some
early stages of melanosis when it appeared flat.7,9,11 Few
ophthalmologists were as influential in establishing
practice guidelines as Reese, and few pathologists had
much proficiency in interpreting findings on conjunctival biopsies as he did.
By 1966, Zimmerman12 accumulated considerable experience with conjunctival melanosis treated by
radical surgery. He grew concerned that orbital exenteration might not be justified for some cases of precancerous melanosis because, to him, the histological
findings suggested low potential for aggressive growth
or distant metastasis.12 He proposed a method for
evaluating acquired melanosis based on conjunctival
biopsy that stratified risk for melanoma progression.12
This strategy was formally presented by Zimmerman13
about a decade later.
Folberg et al14,15 would later introduce new terminology for conjunctival intraepithelial melanocytic
neoplasia (ie, primary acquired melanosis with and
without atypia) and then test the prognostic capabilities of different histopathological variables. These
studies tempered the role that radical surgery played in
the management of acquired conjunctival melanosis by
introducing a means of gauging the likelihood of progression.14,15
Jakobiec and Silbert16 perceived a similar challenge
with the overdiagnosis of iris melanoma. They used a
retrospective, clinicopathological study to test their hypothesis and then published their results in 1981 with
the provocative title “Are Most Iris `Melanomas’ Really
Nevi?:”16 Just as Zimmerman12,13 had emphasized that
April 2016, Vol. 23, No. 2
the diagnosis of precancerous melanosis inflated the
risk of cancer in the minds of clinicians, Jakobiec and
Silbert16 believed taxonomy obstructing effective communication. Seldom have charges of cancer overdiagnosis been so unambiguously stated.
The dilemma of how best to classify indolent,
melanocytic tumors of the iris did not simply materialize. Inadequacies with the classification of melanocytic tumors of the uveal tract had been known since
the 1960s, which is when it was realized that the original system developed in 1931 by Callender17 did not
include nevus (Table).18-20 In 1978, McClean et al18,19
addressed this oversight by studying 105 spindle A
melanomas of the choroid and ciliary body that had
already been histologically diagnosed by expert consultants. After reviewing each case, they found that
15 cases (14.3%) of spindle A melanomas were actually cytologically benign nevi.18,19 Among this group, no
evidence existed of spread or tumor-related death.18,19
Another 75 tumors (71.4%) were diagnosed as spindle cell melanomas based on cytological features.18,20
They consisted of mixtures of spindle A and B melanocytes, and they had a prognosis similar to those previously reported as spindle B melanoma.18,20 Another
15 tumors (14.3%) contained some proportion of epithelioid melanocytes and had a prognosis similar to a
mixed-cell type.18 These cases were accordingly reclassified as mixed-cell type melanomas.18 The so-called
“classic” spindle A melanocyte fell along a cytological spectrum, and it could be found in both nevi and
spindle melanomas.20 The authors recommended that
choroidal and ciliary body nevus be added to the spectrum of diagnoses and that the distinction between
spindle A and B types of melanomas be dropped
(see Table).18,20
The approach to testing the perception of the
overdiagnoses of cancer and precancer that these investigators took was similar. Based on histopathological observations, they proposed improved means of
predicting clinical behavior and then tested those algorithms against clinical outcomes retrospectively ob-
tained. In each situation, pilot studies supported their
premises of overdiagnosis. However, uncommon cancers with prolonged clinical courses are difficult to
study. Nonetheless, the boundary between indolent
and aggressive neoplasia examined by these investigators must always be scrutinized in order to improve diagnostic precision.
These examples emphasize a critical difference between the overdetection of cancer, related to excessive
cancers found in screening programs, and overdiagnosis, which applies to pathologically verified cancers (or
in situ disease) of an indolent type.3 These overdiagnoses are not the result of misdiagnoses; rather, the diagnoses satisfied the standards for precancerous melanosis, iris melanoma, and spindle A choroidal and ciliary
body melanoma that existed at the time. What was in
dispute was whether the diagnostic categories were
too broad, harboring lesions with little propensity for
growth or metastatic spread and no risk of death.
Epidemiological deduction provides a belated defense against overdiagnosis, but this safeguard is ill
suited for uncommon cancers or ones not subject to
screening programs like those in and around the eye.
However, the legacy of overdiagnosis of ocular and
ocular adnexal cancers may unconsciously linger in
the minds of many clinicians, possibly explaining the
intensity with which ocular oncologists have critiqued
new systems of cancer staging.21 Ever since the seventh
edition of the American Joint Committee on Cancer
Staging System was published,21 a flurry of studies has
assessed the updated algorithms.22-26 Although most
have found that the modifications are based on sound
evidence and are thusly improved, some already anticipate supplementing them with new technologies.27
Establishing the interface of benign and malignant and indolent and aggressive cancers has been
the province of anatomical pathologists for a century,
but this jurisdiction is yielding to molecular “pathologists” who have at their disposal the genetic makeup
of tumors from which to work. The genetic signatures
of primary ocular cancers (eg, retinoblastoma, uveal
Table. — Histological Classification of Choroidal and Ciliary Body Melanomas
Original Grouping17
Spindle A–type melanoma
Modified Classification18-20
Comment
Melanocytic nevus
Callender did not consider benign category
Spindle cell melanoma
Not synonymous with nevus
Spindle B–type melanoma
17
Spindles A and B–type melanocytes commonly
found together
Epithelioid-type melanoma
Epithelioid type
Worse prognosis
Mixed spindle- and epithelioid-type melanoma
Mixed spindle- and epithelioid-type melanoma
Prognosis falls between spindle and
epithelioid types of melanoma
Fascicular pattern melanoma
Spindle cell melanoma
Similar outcome as spindle cell
melanoma
Necrotic melanoma
Necrotic melanoma
Tumors too necrotic to classify
April 2016, Vol. 23, No. 2
Cancer Control 91
melanoma) may forecast risk with greater reliability
and precision than before and also provide insights
into targeted therapies.27,28
In conclusion, timely, accurate diagnosis and the
ability to forecast the behavior of malignancy are the
goals of clinical practice and are central themes running through papers in this issue of Cancer Control.
Clinical decision-making in ocular oncology may continue to rely on histopathological parsing to minimize
overdiagnosis, but predicting the biological behavior
of cancer will increasingly depend on molecular-based
strategies as we look toward the future.
23. Shinder R, Ivan D, Seigler D, et al. Feasibility of using American Joint
Committee on Cancer Classification criteria for staging eyelid carcinomas.
Orbit. 2011;30(5):202-207.
24. Sniegowski MC, Roberts D, Bakhoum M, et al. Ocular adnexal lymphoma: validation of American Joint Committee on Cancer seventh edition
staging guidelines. Br J Ophthamol. 2014;98(9):1265-1260.
25. Graue GF, Finger PT, Maher E, et al. Ocular adnexal lymphoma
staging and treatment: American Joint Committee on Cancer versus Ann
Arbor. Eur J Ophthalmol. 2013;23(3):344-355.
26. Yousef YA, Al-Hussaini M, Mehyar M, et al. Predictive value of TNM
classification, international classification, and Reese-Ellsworth staging of
retinoblastoma for the likelihood of high-risk pathologic features. Retina.
2015;35(9):1863-1869.
27. Kapatai G, Brundler MA, Jenkinson H, et al. Gene expression
profiling identifies different sub-types of retinoblastoma. Br J Cancer.
2013;109(2):512-525.
28. Onken DD, Worley LA, Tuscan MD, et al. An accurate, clinically
feasible multi-gene expression assay for predicting metastasis in uveal
melanoma. J Mol Diagn. 2010;12(4):461-468.
Curtis E. Margo, MD
Department of Pathology and Cell Biology
Department of Ophthalmology
University of South Florida Morsani College of Medicine
Tampa, Florida
[email protected]
References
1. Welch HG, Black WC. Overdiagnosis in cancer. J Natl Cancer Inst.
2010;102(9):605-613.
2. Kramer BS.The science of early detection. Urol Oncol. 2004;22(4):344-347.
3. Bae J-M. Overdiagnosis: epidemiologic concept and estimation.
Epidemiol Health. 2015;37:e2015004.
4. Barry MJ. Screening for prostate cancer--the controversy that
refuses to die. N Engl J Med. 2009;360(13):1351-1354.
5. Wegwarth O, Gigerenzer G. Less or more: overdiagnosis and overtreatment: evaluation of what physicians tell their patients about screening
harms. JAMA Intern Med. 2013;173(22):2086-2087.
6. Heleno B, Thomasen MF, Rodrigues DS, et al. Quantification of
harms in cancer screening trials. literature review. BMJ. 2013:347;f5334.
7. Welch HG, Gorski DH, Albertsen PC. Trends in metastatic breast
and prostate cancer – lessons in cancer dynamics. N Engl J Med.
2015;373(18):1685-1687.
8. Reese AB. Precancerous melanosis and diffuse malignant melanoma of the conjunctiva. Arch Ophthalmol. 1938;19:354-365.
9.Reese AB. Precancerous melanosis and the results malignant
melanoma (cancerous melanoma) of conjunctiva and skin of lids. Arch
Ophthalmol. 1943;29:737-747.
10. Reese AB. Precancerous and cancerous melanosis of the conjunctiva.
Am J Ophthalmol. 1955;39:96-100.
11. Reese AB. Tumors of the Eye. Hagerstown, MD: Harper & Row;
1977: 250-254.
12. Zimmerman LE. Criteria for management of melanosis. Arch
Ophthalmol. 1966;76:307-8.
13. Zimmerman LE. The histogenesis of conjunctival melanomas: the first
Algernon B. In: Jakobiec FA, ed. Ocular and Adnexal Tumors. Birmingham,
AL: Aesculapius; 1977: 600-631.
14. Folberg R, McLean I, Zimmerman LE. Primary acquired melanosis
of the conjunctiva. Hum Pathol. 1985;16(2):129-135.
15. Folberg R, McLean IW. Primary acquired melanosis and melanoma
of the conjunctiva: terminology, classification and biologic behavior. Human
Pathol. 1986;17(7):652-654.
16. Jakobiec FA, Silbert G. Are most iris “melanomas” really nevi? A clinicopathologic study of 189 lesions. Arch Ophthalmol. 1981;99(12):2117-2132.
17. Callender GR. Malignant melanotic tumors of the eye: a study of
histological types in 111 cases. Trans Am Acad Ophthalmol Otolaryngol.
1931;36:131-141.
18. McLean IW, Zimmerman LE, Evans RM. Reappraisal of Callender’s
spindle A type of malignant melanoma of choroid and ciliary body. Am J
Ophthalmol. 1978;86(4):557-564.
19. McLean IW. Prognostic features of uveal melanoma. Ophthalmol
Clin North Am. 1995;8(1):143-153.
20. McLean IW, Foster WD, Zimmerman LE. Modifications of Callender’s
classification of uveal melanoma at the Armed Forces Institute of Pathology.
Am J Ophthalmol. 1983;96(4):502-509.
21. Edge S, Byrd DR, Compton CC, et al, eds. AJCC Cancer Staging
Manual. 7th ed. New York: Springer; 2010.
22. Yousef YA, Finger PT. Predictive value of the seventh edition American
Joint Committee on Cancer staging system for conjunctival melanoma. Arch
Ophthalmol. 2012;130(5):599-606.
92 Cancer Control
April 2016, Vol. 23, No. 2
Genomic, analytical, and sequencing
technologies are a critical step toward
use of targeted therapies for select
patients with high-risk uveal melanoma.
Photo courtesy of Lynn E. Harman, MD. Virgin River Gorge.
Assessing Prognosis in Uveal Melanoma
Zélia M. Corrêa, MD, PhD
Background: Because uveal melanoma is the most common primary malignant intraocular tumor in adults
and carries a significant risk of metastases, which are mostly unresponsive to available systemic therapy,
researchers have been searching for prognostic indicators to identify patients at increased risk for developing
such metastasis.
Methods: The purpose of this study is to describe recent advances in prognostic testing of patients with uveal
melanoma and the impact of these advances on the management of uveal melanoma. The relevant, peerreviewed literature as extracted and then further reviewed for scientific content.
Results: Demographic characteristics, clinical, and histopathological features alone are inadequate for predicting metastatic risk in individual patients with uveal melanoma. Some research has shown that cytogenetic
abnormalities and principally transcriptomic features of tumor cells can independently predict high risk for
uveal melanoma metastatic spread. Gene expression profiling of uveal melanoma cells may be accurate and
biologically informative for molecular prognostication. Methods for detecting chromosomal gains and losses
have predictive value but require additional clinical and cytological information. The latest step in the evolution of molecular testing has been the discovery of major driver mutations for possible use in targeted therapy.
Conclusions: Assay validation, quality control, and interpretation of results are essential for the reliability
and reproducibility of these tests. Although these prognostic tests have improved the ability to identify patients
at increased risk for developing metastasis, their use has not changed the management of uveal melanoma.
However, genomic, analytical, and sequencing technologies will provide a critical step toward useful targeted
therapies for patients with high-risk uveal melanoma.
Introduction
Uveal melanoma is the most common primary, maligFrom the Department of Ophthalmology, College of Medicine,
University of Cincinnati, Cincinnati, Ohio.
Address correspondence to Zélia M. Corrêa, MD, PhD, Department
of Ophthalmology, Medical Arts Building, 222 Piedmont Avenue,
Suite 1500, Cincinnati, OH 45267. E-mail: [email protected]
Submitted August 26, 2015; accepted October 30, 2015.
This work was supported in part by an unrestricted grant from
Research to Prevent Blindness (New York, New York), the Quest for
Vision Fund and Ophthalmology Research and Education Fund
of the University of Cincinnati (Cincinnati, Ohio), and the Mary
Knight Asbury Chair endowment fund from the University of
Cincinnati.
April 2016, Vol. 23, No. 2
nant intraocular tumor in adults and carries a significant risk for metastasis that is typically unresponsive
to available systemic therapy, so researchers have been
searching for prognostic indicators to identify patients
with an increased risk for developing such metastasis.1,2 Previously identified factors indicative of poor
prognosis include older age at presentation, male sex,
large tumor size (basal diameter and thickness), scleral invasion and/or extraocular extension, presence of
ocular or oculodermal melanocytosis, ciliary body involvement, epithelioid cells, high values of the mean
diameter of the 10 largest nucleoli, higher microvascular density, vascular mimicry patterns, tumor-infilCancer Control 93
trating lymphocytes and macrophages, high mitotic
activity, high expression of insulin-like growth factor 1
receptor, and high expression of class 1/2 HLA.2-9 However, none of these factors are accurate enough to be
used across the entire spectrum of uveal melanoma.
The challenge clinicians and researchers face is the
ability to accurately identify high-risk patients so as to
further detect the (micro) metastasizing cells at an early phase — possibly a prerequisite for proper patient
selection in future therapeutic interventions.
Methods
The purpose of this study is to describe the recent advances in prognostic testing of patients with uveal melanoma and how these advances are being applied in
clinical practice. A search of the peer-reviewed literature was performed, and relevant articles were identified and carefully reviewed for scientific content and
relevance. The author critically reviewed these publications and summarized the most up-to-date information
related to prognostic testing for uveal melanoma.
Several reports have been published about the
clinical and histological factors associated with the
development of metastasis in patients with uveal
melanoma, and peer-reviewed publications discussing the prognosis of patients with uveal melanoma
date as early as 1948.10 This review article will focus
on publications since 1990, for which recent advances
in prognostic testing of patients with uveal melanoma
are reported when the implications of a gain or loss of
specific chromosomes in these tumors were first realized.
Results
After relying for several decades on demographical,
clinical, and histopathological features to provide a
prognostic estimate for uveal melanoma, and after realizing our limited rate of accuracy for predicting outcomes in individual patients, the research focus shifted
toward chromosomal and cytogenetic indicators of
metastatic risk in uveal melanoma, following the trend
of other cancers.11,12
Early studies of karyotype analysis used fluorescence in situ hybridization (FISH) and comparative
genomic hybridization (CGH). Through karyotype
analysis, Prescher et al13 discovered that monosomy
3 and increased copies of chromosome 8q were commonly found in uveal melanoma. Thus, the association between uveal melanoma with monosomy 3 and
chromosome 8q gain and the development of metastasis has been known for a long time.14
The loss of 1 chromosome 3 was also proven to be
part of a 2-step mutation mechanism for the inactivation of BAP1, a tumor suppressor gene.15 Concomitant
loss of chromosomes 1p and 3 has a stronger correlation with metastasis.16 The association with chromo94 Cancer Control
some 8q gain was also shown to be less significant
than for monosomy of chromosome 3 and than either
one of them separately.17 Gain of chromosome 8 or acquisition of an isochromosome 8q may be a later event
in the setting of uveal melanoma and is seen in both
low- and high-risk uveal melanoma.17 Gain of isochromosome 8q is more frequently associated with metastasis when it is accompanied by chromosome 8p loss
(so-called isochromosome 8q).16 This frequently occurs in tumors that have lost a copy of chromosome
3, and it is considered an independent prognostic factor of progressive disease.17 Chromosome 6p abnormality was predictive of a more favorable outcome.18
Uveal melanomas with gain of chromosome 6p may
represent a separate group of tumors with an alternative genetic pathway in carcinogenesis, because gain
of chromosome 6p is frequently found in tumors with
disomy 3.19,20 Other chromosomal abnormalities have
also been detected in uveal melanoma; however, they
often lead to contradictory results regarding the prognostic impact.21
Although FISH has been used to detect these abnormal chromosomal gains and losses, it is a low-resolution technique associated with many false-negative
and false-positive results, so its prognostic utility has
been limited.22,23 Schouten et al24 described the multiplex ligation–dependent probe amplification (MLPA)
that can be used for detecting the relative quantities of
as many as 40 different DNA sequences. Array CGH
and MLPA have higher rates of accuracy than FISH but
are limited by intratumoral genetic heterogeneity and
an inability to detect isodisomy 3 (tumor cells are functionally monosomy 3 but have duplicated the diseased
copy of the chromosome).23 Molecular classification
based on gene-expression profiling (GEP) of posterior
uveal melanoma has also been shown to be superior
to FISH and array CGH for detecting monosomy 3 and
other chromosomal abnormalities, as well as the clinicopathological prognostic factors for predicting metastasis in uveal melanoma.25,26
Multiplex Ligation–Dependent Probe
Amplification
MLPA is an assay that analyzes the gain and loss of
chromosomal material.24 Through the reaction, denatured genomic DNA is mixed with probes for the
specific target genes of interest. MLPA is sensitive
and sequence specific for detecting changes in DNA
copy numbers, detecting deletions and amplifications
of single exons. MLPA can be performed on fresh,
snap-frozen, formalin-fixed, paraffin-embedded tissue
samples, although fresh and snap-frozen samples have
been reported to yield more reliable results.27
Use of MLPA for posterior uveal melanoma tissue was described by a group of researchers led by
Damato et al3 and Coupland et al.21 Their initial reApril 2016, Vol. 23, No. 2
search analyzed uveal melanoma tissue samples from
73 patients.3,21 MLPA can detect chromosomal abnormalities that correlated with metastatic death, most
importantly loss of chromosome 3 and gain of chromosomal material on 8q.28 A larger subsequent study
showed that losses of chromosomes 1p and 3 and gain
of chromosome 8q correlated with increased rates of
mortality, whereas gain in chromosome 6p correlated
with improved rates of survival.29 MLPA also provided
prognostic information related to chromosomal aberrations in patients with uveal melanomas, and the authors reported that their results correlated with the
clinicopathological features of the tumors.29 However,
although the results of MLPA provided accurate prognoses in this study, the modality did not offer discriminatory stratification of cases as robust as that seen with
GEP testing.29 In addition, the estimation of metastatic
risk using MLPA requires the clinician to order the test,
carefully interpret its complex report, and add clinicopathological features to improve its rate of accuracy.30
The intratumor genetic heterogeneity of posterior uveal melanoma has also been studied with MLPA using
formalin-fixed, paraffin-embedded tumor tissues.31 Researchers found that 25% of the studied tumors were
homogeneous; consequently, they found that heterogeneity causes equivocal results on MLPA and may impair the rate of accuracy of their results.31
In retrospective studies, MLPA results reportedly
correlate well with the development of metastasis and
the survival rate of patients with posterior uveal melanoma; however, the test has not been prospectively
validated in a multicenter clinical trial.4,28,30 This test is
now commercially available.
Gene Expression Profile
GEP can be used for the rapid detection of the upregulation or down-regulation of select genes in a tissue sample.26 The technique involves isolating RNA
from a tissue sample followed by its conversion to
complementary DNA, whose targets are subsequently hybridized to gene chips, and microarray analysis
is performed. Onken et al26 demonstrated that uveal
melanomas could be divided into 2 distinct prognostic
classes that predict a person’s death from metastasis.
The authors initially identified 62 genes that showed
distinct aberrant expression patterns in the studied
samples.26 When the authors combined their findings
with the clinical outcome of patients, the up-regulation
or down-regulation of specific gene clusters identified
by the GEP assay enabled this stratification scheme to
predict metastatic risk.26 Class 1 tumors generally have
the clinical and pathological features known to be associated with decreased metastatic risk, such as the
presence of spindle cells, whereas class 2 tumors generally have more aggressive clinical and pathological
features such as epithelioid cells.26
April 2016, Vol. 23, No. 2
Subsequently, a multicenter prospective study
was conducted to develop and later validate a 15-gene
polymerase chain reaction–based assay that could discern between class 1 and class 2 tumors.32,33 This multicenter validation ensured that this test would yield
equally reliable results in different settings and that the
results would strongly correlate with rates of survival
and metastasis development.33 Since then, this test has
become commercially available for routine use in clinical practice and has been adopted by several national
and international centers.34
The assay examines the expression patterns of
12 class-discriminating genes identified by the previous analysis and 3 control genes shown to be unchanged in uveal melanomas.26 When compared with
monosomy 3 and the clinicopathological features of
the tumor, GEP demonstrated superior rates of accuracy at predicting the risk of metastatic disease in patients with uveal melanoma.25 Since the development
of the assay, class 1 tumors have been further subdivided in classes 1A and B.34 The 5-year rates of metastatic
risk have been estimated to be approximately 2% for
class 1A tumors, 21% for class 1B tumors, and 72% for
class 2 tumors.35 CGH, FISH, and MLPA provide static
measurements of structural chromosomal changes,
whereas GEP detects a dynamic RNA signature of the
tumor microenvironment.16 This fact may explain why
this test has been shown to be the most robust prognostic test for uveal melanoma available.32-34,36-38
Although subdividing class 1 tumors has been
helpful in stratifying the very-low-risk patients (class
1A tumors) from the medium-risk patients (class 1B tumors), this system lacks discrimination compared with
the prior system of 2 classes.39 PRAME was recently
identified as the most significant predictor of metastasis in patients with class 1 tumors.40 Incorporating this
biomarker should further improve the accuracy rate of
the GEP test.
However, several points must be considered regarding the GEP test, as well as any others looking at
genetic and transcriptomic features of tumors. Most
importantly, any tissue sample can be tested and will
originate a result (even if with a low-confidence rate).
Some authors have misinterpreted the purpose of
GEP, erroneously employing it in nonmelanoma specimens.41,42 It is important to emphasize that MLPA and
GEP are prognostic in nature and not diagnostic. Although some have claimed that use of GNAQ/GNA11
mutations ensures that a tumor is uveal melanoma, at
least 15% of uveal melanomas do not have this mutation.43 Rather, the proper diagnostic approach of any
tumor (including uveal melanoma) is by histopathology or cytopathology.36
Another important fact is that the GEP assay can
be performed in paraffin-fixed tissue, but tumor-cell
procurement for this test is best obtained by fine neeCancer Control 95
dle aspiration biopsy at the time of plaque implantation or immediately following enucleation of the eye
containing the tumor.35 The tumor aspirate is flushed
in an RNA-stabilizing buffer solution and then frozen
until the time the assay is performed.33
An advantage of this test is that it requires a minimal amount of cells to generate a result. A single-center study of 159 tumoral samples obtained via biopsy
showed that the obtained tumor aspirate was insufficient for GEP in a single case (0.6%) compared with
34 cases (21.9%) that had an insufficient aspirate for cytology diagnosis.36 In cases of very small uveal melanoma, a heterogeneous genetic make-up may limit this
result (and likely any other test result) in samples yielded by needle aspiration in select patients.31,44
A study performed during the early development
of the current 15-gene test — before it was commercially available — showed discordance of GEP classification in a few cases of very small uveal melanomas sampled at 2 distinct sites.44 Overall, discordant
results were seen in 7.5% of the tested cases compared
with up to 75% of tested cases for which MLPA findings showed tumor heterogeneity.31 That early study
of GEP concluded that the tumor samples obtained
via biopsy from a single site have a small likelihood of
prognostic misclassification (and more likely in thinner tumors [< 3.5 mm in thickness]); therefore, the
authors recommended taking this information into
account when advising patients with smaller tumors
about their prognosis.44
Newer genomic sequencing technologies have also
allowed us a more rapid understanding of the molecular landscape of posterior uveal melanoma.45 In addition, the GEP assay (and other prognostic tests) for
uveal melanoma have provided opportunities for early
detection in patients at high risk of metastasis, thus significantly changing how patients with uveal melanoma
are clinically monitored.35
Mutational Profiling
Although it is not as useful in the prognostication of
patients, detection of specific mutations in uveal melanoma has the potential to improve therapeutic options
for metastatic disease in the future. Through a search
for mutations in the oncogenic mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase pathway (ERK), Onken et al46 first identified the
GNAQ mutation in approximately 50% of uveal melanoma samples. A mutation in GNAQ was detected in
samples of posterior uveal melanoma at all of the
stages of malignant progression, indicating that such a
mutation may play a role in the initial development of
the tumor.46 Mutations in GNA11 were also sufficient
to induce metastases in a mouse model, so researchers concluded that mutations in GNAQ and GNA11 affect a critical oncogenic signaling cascade to affect the
96 Cancer Control
metastatic potential of tumors.43 However, this finding has been questioned because GNAQ and GNA11
mutations in humans result in a benign nevus unless
accompanied by further genetic events such as BAP1
mutation.15 GNA11 mutations were initially thought to
be more aggressive than GNAQ due to their incidence
being slightly higher in metastatic tumors.43 We now
know this is because GNA11 mutations are slightly
more common in ciliary body melanomas, which have
a higher metastatic rate.43 Thus, it is more likely that
GNA11 is not any more aggressive than GNAQ.
Mutations in GNAQ and GNA11 are mutually exclusive and represent early or initiating events that constitutively activate the MAPK pathway.45 Although these
mutations are sensitive to MAPK, protein kinase C, and
Akt inhibitors, GNAQ and GNA11 remain difficult therapeutic targets.47 Researchers have demonstrated that
the oncogenic activity of mutant GNAQ/GNA11 is mediated at least in part through YAP, which could have
therapeutic potential.47,48
Furthermore, mutations in BAP1, SF3B1, and another driver mutation, EIF1AX, may be largely mutually exclusive, and they occur later in tumor progression.49 BAP1 mutations have been reported to be
strongly associated with metastasis, class 2 GEP, and
older age.50,51 Using multiple regression analysis, BAP1
mutations associated with EIF1AX mutations were also
related to class 1 GEP and the absence of ciliary body
involvement, whereas SF3B1 mutations are associated
with a more favorable outcome and younger age.48-51
BAP1 mutations can arise in the germ line, leading to
a newly described BAP1 familial cancer syndrome.45,52
Researchers and clinicians are realizing that, similar to
other cancers, uveal melanomas comprise a heterogeneous group of diseases, each with distinct molecular
features that might respond best to a specific therapeutic strategy.53
All of these recent discoveries have led to new trials to assess several classes of compounds, including
MAPK/ERK inhibitors,54 protein kinase C,55 histone
deacetylase inhibitors,56 and combination therapy,57in
the adjuvant setting for high-risk patients and in the
setting of advanced disseminated disease. The next
generation of targeted therapy agents is also being introduced into the clinical setting to evaluate the efficacy of these molecular agents in patients with uveal
melanoma.58
Conclusions
Prognostic testing has become an important component in the evaluation of patients with uveal melanoma
to assess metastatic risk. Although gene expression
profiling (GEP) and chromosomal counting methods, such as multiplex ligation–dependent probe amplification, each provide different information about
metastatic risk stratification, clinicians can now offer
April 2016, Vol. 23, No. 2
patients important insight about their prognosis for
metastases. Although the potential for tumor heterogeneity and biopsy sampling error must be further investigated, new developments such as the identification
of PRAME expression via GEP may increase the rate
of accuracy in small tumors tested by GEP. Although
the therapeutic options available for the treatment of
metastatic posterior uveal melanoma are in their infancy, patients continue to express their desire to have
accurate prognostic information about their tumors
in order to plan their lives and to consider enrollment in clinical trials for adjuvant therapy,59 several of
which are now available (NCT02601378, NCT01551459,
NCT01377025, NCT01413191). Many centers are also
revising their surveillance protocols to better suit the
needs of patients.60 Recent advances in genomic sequencing technologies have also increased our knowledge of the molecular landscape of uveal melanoma,
thus leading to clinical trials using targeted molecules
to treat metastatic uveal melanoma. Patients whose increased risk for developing metastasis is detected by
genomic testing may wish to enroll in adjuvant treatment trials or they may wish to choose periodic surveillance testing for the earlier detection of asymptomatic metastatic disease. In addition, clinicians can now
offer treatments that have the potential to extend the
lives of patients with uveal melanoma.60
References
1. Augsburger JJ, Correa ZM, Shaikh AH. Effectiveness of treatments
for metastatic uveal melanoma. Am J Ophthalmol. 2009;148(1):119-127.
2. Augsburger JJ, Gamel JW. Clinical prognostic factors in patients
with posterior uveal malignant melanoma. Cancer. 1990;66(7):1596-1600.
3. Damato B, Eleuteri A, Taktak AF, et al. Estimating prognosis for
survival after treatment of choroidal melanoma. Prog Retin Eye Res.
2011;30(5):285-295.
4. Damato BE, Heimann H, Kalirai H, et al. Age, survival predictors, and metastatic death in patients with choroidal melanoma: tentative evidence of a therapeutic effect on survival. JAMA Ophthalmol.
2014;132(5):605-613.
5. McLean IW, Ainbinder DJ, Gamel JW, et al. Choroidal-ciliary body
melanoma. A multivariate survival analysis of tumor location. Ophthalmology.
1995;102(7):1060-1064.
6.Gamel JW, McCurdy JB, McLean IW. A comparison of prognostic covariates for uveal melanoma. Invest Ophthalmol Vis Sci.
1992;33(6):1919-1922.
7. Folberg R, Rummelt V, Parys-Van Ginderdeuren R, et al. The prognostic value of tumor blood vessel morphology in primary uveal melanoma. Ophthalmology. 1993;100(9):1389-1398.
8. Kivela T, Kujala E. Prognostication in eye cancer: the latest tumor,
node, metastasis classification and beyond. Eye (Lond). 2013;27(2):243-252.
9. Toivonen P, Makitie T, Kujala E, et al. Microcirculation and tumorinfiltrating macrophages in choroidal and ciliary body melanoma and corresponding metastases. Invest Ophthalmol Vis Sci. 2004;45(1):1-6.
10. Benjamin B, Cumings JN, et al. Prognosis in uveal melanoma. Br J
Ophthalmol. 1948;32(10):729-747.
11. Kerbel RS, Cornil I, Theodorescu D. Importance of orthotopic transplantation procedures in assessing the effects of transfected genes on human
tumor growth and metastasis. Cancer Metastasis Rev. 1991;10(3):201-215.
12. Bowden GT, Schneider B, Domann R, et al. Oncogene activation
and tumor suppressor gene inactivation during multistage mouse skin carcinogenesis. Cancer Res. 1994;54(7 Suppl):1882s-1885s.
13. Prescher G, Bornfeld N, Becher R. Nonrandom chromosomal abnormalities in primary uveal melanoma. J Natl Cancer Inst. 1990;82(22):1765-1769.
14. Prescher G, Bornfeld N, Hirche H, et al. Prognostic implications of
monosomy 3 in uveal melanoma. Lancet. 1996;347(9010):1222-1225.
15. Harbour JW, Onken MD, Roberson ED, et al. Frequent mutation of
BAP1 in metastasizing uveal melanomas. Science. 2010;330(6009):1410-1413.
16. Ehlers JP, Worley L, Onken MD, et al. Integrative genomic analysis
April 2016, Vol. 23, No. 2
of aneuploidy in uveal melanoma. Clin Cancer Res. 2008;14(1):115-122.
17. van den Bosch T, van Beek JG, Vaarwater J, et al. Higher percentage of FISH-determined monosomy 3 and 8q amplification in uveal melanoma cells relate to poor patient prognosis. Invest Ophthalmol Vis Sci.
2012;53(6):2668-2674.
18. White VA, Chambers JD, Courtright PD, et al. Correlation of cytogenetic abnormalities with the outcome of patients with uveal melanoma.
Cancer. 1998;83(2):354-359.
19. Aalto Y, Eriksson L, Seregard S, et al. Concomitant loss of chromosome 3 and whole arm losses and gains of chromosome 1, 6, or 8
in metastasizing primary uveal melanoma. Invest Ophthalmol Vis Sci.
2001;42(2):313-317.
20. Gordon KB, Thompson CT, Char DH, et al. Comparative genomic
hybridization in the detection of DNA copy number abnormalities in uveal
melanoma. Cancer Res. 1994;54(17):4764-4768.
21. Coupland SE, Lake SL, Zeschnigk M, et al. Molecular pathology of
uveal melanoma. Eye (Lond). 2013;27(2):230-242.
22.White VA, McNeil BK, Horsman DE. Acquired homozygosity
(isodisomy) of chromosome 3 in uveal melanoma. Cancer Genet Cytogenet.
1998;102(1):40-45.
23. Speicher MR, Prescher G, du Manoir S, et al. Chromosomal gains
and losses in uveal melanomas detected by comparative genomic hybridization. Cancer Res. 1994;54(14):3817-3823.
24. Schouten JP, McElgunn CJ, Waaijer R, et al. Relative quantification of 40 nucleic acid sequences by multiplex ligation-dependent probe
amplification. Nucleic Acids Res. 2002;30(12):e57.
25. Worley LA, Onken MD, Person E, et al. Transcriptomic versus chromosomal prognostic markers and clinical outcome in uveal melanoma.
Clin Cancer Res. 2007;13(5):1466-1471.
26. Onken MD, Worley LA, Ehlers JP, et al. Gene expression profiling
in uveal melanoma reveals two molecular classes and predicts metastatic
death. Cancer Res. 2004;64(20):7205-7209.
27. Lake SL, Kalirai H, Dopierala J, et al. Comparison of formalin-fixed
and snap-frozen samples analyzed by multiplex ligation-dependent probe
amplification for prognostic testing in uveal melanoma. Invest Ophthalmol
Vis Sci. 2012;53(6):2647-2652.
28. Damato B, Dopierala J, Klaasen A, et al. Multiplex ligation-dependent probe amplification of uveal melanoma: correlation with metastatic
death. Invest Ophthalmol Vis Sci. 2009;50(7):3048-3055.
29. Lake SL, Damato BE, Dopierala J, et al. Multiplex ligation-dependent probe amplification analysis of uveal melanoma with extraocular extension demonstrates heterogeneity of gross chromosomal abnormalities.
Invest Ophthalmol Vis Sci. 2011;52(8):5559-5564.
30. Damato B, Dopierala JA, Coupland SE. Genotypic profiling of 452
choroidal melanomas with multiplex ligation-dependent probe amplification. Clin Cancer Res. 2010;16(24):6083-6092.
31. Dopierala J, Damato BE, Lake SL, et al. Genetic heterogeneity in
uveal melanoma assessed by multiplex ligation-dependent probe amplification. Invest Ophthalmol Vis Sci. 2010;51(10):4898-4905.
32. Onken MD, Worley LA, Tuscan MD, et al. An accurate, clinically
feasible multi-gene expression assay for predicting metastasis in uveal
melanoma. J Molec Diag. 2010;12(4):461-468.
33. Onken MD, Worley LA, Char DH, et al. Collaborative Ocular Oncology
Group report number 1: prospective validation of a multi-gene prognostic
assay in uveal melanoma. Ophthalmology. 2012;119(8):1596-1603.
34. Harbour JW, Chen R. The DecisionDx-UM gene expression profile test provides rsk stratification and individualized patient care in uveal
melanoma. PLoS Curr. 2013;5:pii.
35. Harbour JW, Chao DL. A molecular revolution in uveal melanoma: implications for patient care and targeted therapy. Ophthalmology.
2014;121(6):1281-1288.
36. Correa ZM, Augsburger JJ. Sufficiency of FNAB aspirates of
posterior uveal melanoma for cytologic versus GEP classification in 159
patients, and relative prognostic significance of these classifications.
Graefes Arch Clin Exp Ophthalmol. 2014;252(1):131-135.
37. Gill HS, Char DH. Uveal melanoma prognostication: from lesion size
and cell type to molecular class. Can J Ophthalmol. 2012;47(3):246-253.
38. Petrausch U, Martus P, Tonnies H, et al. Significance of gene
expression analysis in uveal melanoma in comparison to standard risk
factors for risk assessment of subsequent metastases. Eye (Lond).
2008;22(8):997-1007.
39. Field MG, Harbour JW. Recent developments in prognostic and predictive testing in uveal melanoma. Curr Opin Ophthalmol. 2014;25(3):234-239.
40.Field MG, Decatur CL, Harbour JW. PRAME as a biomarker
for a new molecular subclass of uveal melanoma. Clin Cancer Res.
2016;22(5):1234-1242.
41. Seider MI, Stewart PJ, Mishra KK, et al. Uveal melanoma gene expression profile test result provided for uveal metastasis. Ophthalmic Surg
Lasers Imaging Retina. 2014;45(5):441-442.
42. Klufas MA, Itty S, McCannel CA, et al. Variable results for uveal
melanoma-specific gene expression profile prognostic test in choroidal
metastasis. JAMA Ophthalmol. 2015;133(9):1073-1076.
43. Van Raamsdonk CD, Griewank KG, Crosby MB, et al. Mutations in
Cancer Control 97
GNA11 in uveal melanoma. N Engl J Med. 2010;363(23):2191-2199.
44. Augsburger JJ, Correa ZM, Augsburger BD. Frequency and implications of discordant gene expression profile class in posterior uveal
melanomas sampled by fine needle aspiration biopsy. Am J Ophthalmol.
2015;159(2):248-256.
45. Harbour JW. Genomic, prognostic, and cell-signaling advances in
uveal melanoma. Am Soc Clin Oncol Educ Book. 2013:388-391.
46.Onken MD, Worley LA, Long MD, et al. Oncogenic mutations
in GNAQ occur early in uveal melanoma. Invest Ophthalmol Vis Sci.
2008;49(12):5230-5234.
47. Field MG, Harbour JW. GNAQ/11 mutations in uveal melanoma: is
YAP the key to targeted therapy? Cancer Cell. 2014;25(6):714-715.
48. Yu FX, Zhang K, Guan KL. YAP as oncotarget in uveal melanoma.
Oncoscience. 2014;1(7):480-481.
49. Decatur CL, Ong E, Garg N, et al. Driver mutations in uveal melanoma: associations with gene expression profile and patient outcomes.
JAMA Ophthalmol. 2016. Epub ahead of print.
50. Harbour JW, Roberson ED, Anbunathan H, et al. Recurrent mutations at codon 625 of the splicing factor SF3B1 in uveal melanoma.
Nat Genet. 2013;45(2):133-135.
51. van Essen TH, van Pelt SI, Versluis M, et al. Prognostic parameters in uveal melanoma and their association with BAP1 expression.
Br J Ophthalmol. 2014;98(12):1738-1743.
52. Njauw CN, Kim I, Piris A, et al. Germline BAP1 inactivation is preferentially associated with metastatic ocular melanoma and cutaneousocular melanoma families. PLoS One. 2012;7(4):e35295.
53. Patel M, Smyth E, Chapman PB, et al. Therapeutic implications
of the emerging molecular biology of uveal melanoma. Clin Cancer Res.
2011;17(8):2087-2100.
54. Templeton IE, Musib L. MEK inhibitors beyond monotherapy: current and future development. Curr Opin Pharmacol. 2015;23:61-67.
55. Cerne JZ, Hartig SM, Hamilton MP, et al. Protein kinase C inhibitors
sensitize GNAQ mutant uveal melanoma cells to ionizing radiation. Invest
Ophthalmol Vis Sci. 2014;55(4):2130-2139.
56. Landreville S, Agapova OA, Matatall KA, et al. Histone deacetylase
inhibitors induce growth arrest and differentiation in uveal melanoma. Clin
Cancer Res. 2012;18(2):408-416.
57. Chen X, Wu Q, Tan L, et al. Combined PKC and MEK inhibition in uveal melanoma with GNAQ and GNA11 mutations. Oncogene.
2014;33(39):4724-4734.
58. Luke JJ, Triozzi PL, McKenna KC, et al. Biology of advanced uveal
melanoma and next steps for clinical therapeutics. Pigment Cell Melanoma
Res. 2015;28(2):135-147.
59. Cook SA, Damato B, Marshall E, et al. Psychological aspects of
cytogenetic testing of uveal melanoma: preliminary findings and directions
for future research. Eye. 2009;23(3):581-585.
60.Shoushtari AN, Carvajal RD. Treatment of uveal melanoma.
Cancer Treat Res. 2016;167:281-293.
98 Cancer Control
April 2016, Vol. 23, No. 2
Various forms of chemotherapy
and targeted therapies have helped
improve rates of survival and
ocular salvage in retinoblastoma.
Photo courtesy of Lynn E. Harman, MD. Themes at the Shard.
Therapeutic Options for Retinoblastoma
Pia R. Mendoza, MD, and Hans E. Grossniklaus, MD
Background: Retinoblastoma is the most common primary intraocular malignancy in children. The management of retinoblastoma is complex and depends on several factors.
Methods: This review provides an update on current and emerging therapeutic options for retinoblastoma.
The medical literature was searched for articles relevant to the management of retinoblastoma. The results of
prospective and retrospective studies on chemotherapy and focal therapy for retinoblastoma are summarized.
Animal models for novel therapeutic agents are also discussed.
Results: Treatment strategies for retinoblastoma involve intravenous chemoreduction, local administration routes
of chemotherapy (eg, intra-arterial, intravitreal), focal therapy for tumor consolidation (eg, photocoagulation,
thermotherapy, cryotherapy, plaque brachytherapy), external beam radiotherapy, and surgical enucleation.
Emerging therapies include alternative chemotherapeutic agents, molecularly targeted therapies, and novel
drug-delivery systems.
Conclusion: In the past 10 years, the management strategy for retinoblastoma has significantly changed, shifting
toward local chemotherapy and away from systemic chemotherapy. Innovations in the field of molecular biology
and the development of targeted therapies have led to improvements in survival rates and ocular salvage for this
disease. However, the need still exists to further assess the long-term effects of such directional changes in therapy.
Introduction
Retinoblastoma is the most common primary intraocular malignancy in children; its incidence rate is 1 in
14,000 to 20,000 livebirths.1 However, compared with
other pediatric malignancies, retinoblastoma is rare,
comprising 3% to 4% of all pediatric cancers.2 It is a
tumor of the developing retina that arises from primitive retinal stem cells or cone precursor cells associated
with inactivation of both alleles of the tumor suppresFrom the Department of Ophthalmology, Emory University School
of Medicine, Atlanta, Georgia.
Address correspondence to Hans E. Grossniklaus, MD, 1365
Clifton Road, Northeast, BT428, Atlanta, GA 30322. E-mail:
[email protected]
Submitted October 14, 2015; accepted January 5, 2016.
No significant relationships exist between the authors and the
companies/organizations whose products or services may be referenced in this article.
April 2016, Vol. 23, No. 2
sor gene RB1. Major advances, such as our understanding of the molecular biology of the tumor and the development of targeted therapy, have improved survival
rates in developed countries.3 In the United States, the
5-year overall survival rate is 97%.4 Much of this success is due to the benefits of effective treatments, including chemotherapy, focal or consolidation therapy,
external beam radiotherapy, and surgical enucleation
(Table 1).5 With early diagnosis and aggressive, multimodal treatment strategies, near-complete cure rates
are possible, and many patients retain functional vision in at least 1 eye. However, in developing nations,
where access to health care is limited, retinoblastoma
can cause blindness and death.6
Among the mainstay treatments for retinoblastoma
are intravenous chemotherapy (involving agents associated with significant toxicities), surgical enucleation,
external beam radiotherapy, or all 3 options, potenCancer Control 99
tially result in disfigurement and secConcepts and Principles of
Table 1. — Therapeutic Options
ondary malignancies. However, in reTreatment
in Retinoblastoma
cent years, an ongoing evolution has
The goal of treatment in retinoblasChemotherapy
taken place, where treatment is being
toma is the elimination of the tumor
Intravenous
geared toward more targeted therapy
while concurrently minimizing coland local medication delivery. Local
lateral injury to other tissues. The
Chemoreduction
delivery increases exposure to intrapriorities in management are (1) preAdjuvant therapy
ocular target areas, reduces overall
venting metastasis, (2) reducing the
Metastatic retinoblastoma
systemic exposure and harmful late
risk of long-term secondary tumors
Intra-arterial
effects, and improves effectiveness
(eg, osteosarcoma, soft-tissue sarcoIntravitreal
and rates of tolerability. The introma), (3) saving the eye, and (4) prePeriocular
duction of intra-arterial and intravitserving vision.11 Caring for a patient
Focal Therapy
real chemotherapy has resulted in a
with retinoblastoma must be a mulchange in treatment algorithms and
tidisciplinary effort involving pediThermotherapy
improved outcomes, with their posatric oncologists, ocular oncologists,
Photocoagulation
sibility of ocular salvage and mainteradiation oncologists, ocular patholCryotherapy
nance of vision.7
ogists, and geneticists to optimize
Plaque brachytherapy
Preclinical models that simulate
treatment outcomes.
External Beam Radiotherapy
human disease are needed for rare
Treatment depends on tumor
Enucleation
childhood cancers such as retinoblasstage, laterality and number of tutoma because not enough patients
mor foci (unifocal, unilateral, mulwith the disease exist for large-scale,
tifocal, or bilateral), the localization
multicenter clinical trials. Despite the lack of clinical
and size of tumors within the eye, presence of vittrials, rates of survival and ocular preservation have
reous seeding, age and health of the child, and the
improved.7 In vitro studies and animal models remain
family’s wishes.12 The type of management depends
instrumental in understanding biological mechanisms
on the tumor classification, which is based on 2 sysand assessing safety and effectiveness of the treatment
tems: the International Classification of Retinoblasoptions for this disease.8-10 Examples of emerging thertoma13 used to predict success of chemoreduction14
apies under investigation for retinoblastoma are alterand the tumor, node, and metastasis classification
native chemotherapeutic agents, molecularly targeted
of the American Joint Committee on Cancer15 for tutherapies, and novel drug-delivery systems.
mor staging (Table 2). Location of the tumor in relaTable 2. — International Classification and General Therapeutic Options for Retinoblastoma
Eye Group
General Feature
Specific Feature (Presence of ≥ 1)
Therapy
A
Small tumor away from
fovea and optic disc
Tumor ≤ 3 mm
Laser photocoagulation
Thermotherapy
Cryotherapy
Plaque brachytherapy
B
Larger tumor
Macular
Juxtapapillary
Subretinal fluid
Tumor > 3 mm
Tumor located < 3 mm from fovea
Tumor located ≤ 1.5 mm from optic disc
Presence of subretinal fluid ≤ 3 mm from tumor margin
Laser photocoagulation
Thermotherapy
Cryotherapy
Plaque brachytherapy
Intravenous/intra-arterial chemoreduction
C
Focal seeds
Presence of subretinal, vitreous seeds, or both located ≤ 3 mm
from main tumor
Intra-arterial chemotherapy
Intravitreal chemotherapy
D
Diffuse seeds
Presence of subretinal, vitreous seeds, or both located > 3 mm
from main tumor
Intra-arterial chemotherapy
Intravitreal chemotherapy
Enucleation
E
Extensive tumor
Tumor occupying > 50% of the globe
Neovascular glaucoma
Opaque media from hemorrhage in anterior chamber, vitreous,
or subretinal space
Invasion of postlaminar optic nerve, choroid (> 2 mm), sclera,
orbit, anterior chamber
Intra-arterial chemotherapy
Enucleation
Adjuvant intravenous chemotherapy
if high-risk histopathological features
present
100 Cancer Control
April 2016, Vol. 23, No. 2
tion to the optic disc and macula is important in the
clinical evaluation because it largely affects the visual prognosis. Involvement of certain tissues in and
around the eye, including the optic nerve, choroid,
iris, sclera, and orbit, increase the likelihood of tumor
metastasis.16
In children, unilateral retinoblastoma is differently managed than bilateral tumors. In general,
unilateral cases are the result of sporadic mutations,
whereas bilateral cases are typically multifocal, hereditary, and occur in patients carrying a germline
RB1 mutation, thus placing them at risk for other malignancies (eg, intracranial neuroblastic tumors [pinealoblastoma in trilateral retinoblastoma]). In general,
unilateral tumors can be managed with focal therapy
alone; chemoreduction, intra-arterial chemotherapy
with focal therapy, or both; or enucleation. If genetic
testing is not performed, then patients with unilateral
disease require close follow-up of the unaffected eye
to exclude bilateral involvement. Alternatively, treatment for bilateral tumors will depend on the extent of
tumor in each eye. In asymmetric disease, an attempt
is made to salvage the eye with less severe disease.
One may consider giving patients systemic intravenous chemoreduction as initial therapy to treat both
eyes as well as to prevent metastasis and associated
secondary tumors or to use local (intra-arterial) chemotherapy with focal treatment.7,17,18
Initial chemotherapy in retinoblastoma can be given either systemically (intravenous) or locally (directly
through the ophthalmic artery/intra-arterial). Chemoreduction is then usually followed by consolidation,
which is achieved by focal therapy (eg, laser photocoagulation, thermotherapy, cryotherapy, plaque
brachytherapy). Focal therapy may be employed as
the only or initial therapy for small retinoblastoma
tumors located away from the fovea and optic nerve
(group A); it is often used for tumor consolidation in
retinoblastoma following chemoreduction for more
advanced (groups B–D) tumors. Surgical removal of
the eye or enucleation is reserved for very advanced
(group E) cases.
Management
Chemotherapy
Intravenous: Systemic chemotherapy generally involves a combination 2-, 3-, or 4-drug regimen delivered through an intravenous catheter. Three classes
of agents are commonly employed: DNA-crosslinking agents (carboplatin, cisplatin), DNA topoisomerase 2 inhibitors (etoposide, topotecan, teniposide) and
Vinca alkaloids (vincristine; Fig 1). The most commonly used regimen is vincristine/etoposide/carboplatin.11
Chemoreduction via systemic chemotherapy was
introduced as a management option for retinoblastoma
in the mid-1990s following preliminary observations
April 2016, Vol. 23, No. 2
that systemic chemotherapy delivered prior to external
beam radiotherapy contributed to tumor control and
ocular salvage.19 A subsequent study on eyes treated
with chemoreduction combined with focal treatments
(cryotherapy, thermotherapy, or plaque radiotherapy)
demonstrated tumor regression and decreased need
for additional external beam radiotherapy and enucleation.20 This was a major advancement in management
because it was evident that satisfactory tumor control
could be attained with systemic chemotherapy while
saving the eye and avoiding the adverse events of external beam radiotherapy.20 Using the International Classification of Retinoblastoma, treatment success was found
in 100% of group A eyes, 93% of group B eyes, 90% of
group C eyes, and 48% group D eyes.14 This launched
the so-called “systemic chemotherapy era” (1990s to
2006); however, with the advent of local routes of administration (intra-arterial and intravitreal), use of systemic chemotherapy has decreased in recent years.14
Intravenous chemotherapy is indicated as initial
therapy for bilateral advanced disease in which attempts are made to salvage both eyes. Many centers
use systemic chemotherapy in bilateral (germline) retinoblastoma for intraocular retinoblastoma control as
well as to prevent metastasis, reduce the likelihood of
the development of pineoblastoma, and to reduce the
long-term risk of secondary cancers.17,18 Systemic chemotherapy is also utilized as adjuvant treatment to prevent metastasis following enucleation in patients with
optic nerve invasion posterior to the lamina cribrosa,
massive choroidal invasion, or any combination of optic nerve and choroidal invasion.21 The treatment regimens used include vincristine/etoposide/carboplatin,
vincristine/doxorubicin/idarubicin/cyclophosphamide,
or hybrid regimens, typically delivered monthly for
6 to 9 months.22 A major obstacle to use of intravenous
chemotherapy in retinoblastoma is the blood–retina
barrier, which limits the entry of some chemotherapeutic agents into the eye, thereby reducing their efficacy.23
Systemic chemotherapy for retinoblastoma is generally
safe and effective, but it is not without adverse events,
such as neurotoxicity, hyponatremia, nephrotoxicity,
ototoxicity, and secondary leukemia. Treatment failure in the form of persistent vitreous seeds, subretinal
seeds, and intraretinal tumors following therapy have
been attributed to the inability of the treatment drugs
to reach the tumor.24 Tumor resistance or unresponsiveness to chemotherapy is thought to occur more often
in well-differentiated tumors, presumably because cells
are not cycling, so they are less likely to respond to
treatment modalities affecting cell division.25
Intra-Arterial: In the late 1980s, melphalan was
found to be effective for the management of retinoblastoma in vitro.26 Yamane et al27 and Suzuki et al28 then
began to treat patients with intracarotid artery melphalan, developing a technique to safely and effectively
Cancer Control 101
Fig 1. — Chemotherapeutic agents used in retinoblastoma. Alkylating agents such as melphalan cause DNA intra–strand-linking and cross-linking,
resulting in DNA damage and disruption of transcription. Topoisomerase inhibitors such as etoposide and topotecan cap free double-strand breaks in
DNA, preventing DNA repair and apoptosis. Platinum-based agents such as carboplatin bind DNA bases, resulting in kinked DNA. Vinca alkaloids such
as vincristine bind tubulin dimers and block mitotic spindle formation.
Reprinted from Grossniklaus HE. Retinoblastoma. Fifty years of progress. The LXXI Edward Jackson Memorial Lecture. Am J Ophthalmol.
2014;158(5):875-891, with permission from Elsevier.
cannulate the ophthalmic artery for chemotherapy infusion. In the United States, intra-arterial chemotherapy (IAC) was initially used by Abramson et al.29 IAC involves transfemoral catheterization with advancement
into the ophthalmic artery, which is where melphalan,
topotecan, or carboplatin is infused. The medication
is slowly delivered for 30 minutes in a pulsatile fashion with care to not occlude the artery and to minimize
reflux into the internal carotid artery. Most patients
receive 3 monthly sessions. Melphalan is the most frequently used agent, with topotecan added if extensive
vitreous seeding is present.
Generally, IAC is employed for unilateral or nongermline retinoblastoma, recurrent tumors following
previous intravenous chemotherapy or plaque radiotherapy, recurrent subretinal seeds involving at least
2 quadrants, recurrent vitreous seeds, or, if the patient
wishes to avoid systemic therapy, enucleation can be
considered if consent is obtained. In a 5-year experience with IAC, complete regression was achieved for
solid tumors in 94%, for subretinal seeds in 95%, and
for vitreous seeds in 87% of study patients.30 In recent
102 Cancer Control
years, globe-salvage results (85%–94% globe salvage
rate in group D tumors with primary IAC) have been
reported in study patients with advanced retinoblastoma.30,31 In some centers, most group D eyes are primarily treated with IAC, and, on occasion, group D
and E eyes are managed with combined intravenous
chemotherapy followed by intra-arterial chemotherapy
— particularly if the patient has advanced bilateral disease or disease in a single eye.7 Fair success has been
reported in group E tumors with total retinal detachment cautiously managed with initial IAC.32,33 Treatment of more advanced disease with initial IAC has
also not been shown to compromise survival rates.7
After adequate reduction is visualized and subretinal
fluid is resolved, complete consolidation with either
thermotherapy or cryotherapy and supplemental intravitreal chemotherapy after IAC are commonly performed. Consolidation is generally performed during
the second or third cycle of chemoreduction.
In infants younger than 3 months who weigh less
than 7 kg, IAC is initially deferred to allow the femoral artery to grow before attempting catheterization.
April 2016, Vol. 23, No. 2
In such cases, patients are treated with 1 to 2 doses of
single-agent carboplatin as bridge therapy before definitive IAC.34
Reports of high rates of efficacy of IAC abound,
but studies also exist on its associated ocular toxicities.35-37 Use of intra-arterial infusion of melphalan
and carboplatin compromise the ocular vasculature
in nonhuman primates and induces inflammation
and leukostasis in human retinal endothelial cells.35,36
Other complications include vitreous hemorrhage,
microemboli to the retina and choroid, myositis,
eyelid edema, orbital congestion with resulting dysmotility, choroidal atrophy, optic atrophy, ophthalmic
artery stenosis, and branch-retinal artery occlusion
resulting in blindnesss.37
A major drawback to this therapy is that only
skilled neurosurgeons and/or interventional radiologists at major cancer centers can treat patients using IAC.38 The therapy is also expensive.39 In developing countries, IAC is not available or is too costly,
so enucleation as initial therapy is still a mainstay of
treatment for advanced retinoblastoma.40 In addition,
a study has shown that initial chemotherapy prior to
enucleation for advanced retinoblastoma can mask
pathological evidence of tumor extension, thus leading to reduced surveillance and the undertreatment of
high-risk disease — thereby increasing the risk of metastatic death.41
Intravitreal: The major question encountered
in the management of retinoblastoma is how to treat
subretinal and vitreous seeds of tumor unresponsive
to laser treatment, conventional chemoreduction therapy, or IAC.24 Treatment of vitreous seeds is challenging due to lack of vasculature in the vitreous. Another
route of administration developed to address this problem is the intravitreal method, which involves injecting
the therapeutic agent into the vitreous cavity of the eye
through the pars plana under aseptic precautions.
Although health care professionals were initially
skeptical of injecting drugs through the sclera into the
vitreous for fear of tumor spread along needle tracks,
techniques for safe and effective intravitreal injections
have been developed. Intraocular pressure is first lowered prior to the injection. A small-volume dose of melphalan, topotecan, or a combination of both is injected
into the eye using a fine needle (30- or 32-gauge) and
the needle is frozen with a cryoprobe as it is withdrawn from the eye to prevent tumor seeding.42 The
first-line indication for intravitreal chemotherapy includes vitreous seeds refractory to standard therapy
and recurrent vitreous seeds after previous therapy.43,44
In an analysis of 57 eyes with viable recurrent vitreous seeds, intravitreal melphalan led to eye salvage
in 51% of eyes after 10 years of follow-up; no cases
of extraocular extension were reported during that
time.45 A systematic review showed that proper techApril 2016, Vol. 23, No. 2
nique does not increase the risk of tumor spread.46
Treatment of retinoblastoma with recurrent vitreous seeds has been reported to have a success rate of
83%.47 The number of injections depends on the response, and 6 injections delivered weekly or biweekly is recommended. Reported adverse events include
transient vitreous hemorrhage, chorioretinal atrophy,
and extraocular tumor spread.42,48,49
Melphalan is the most commonly used drug for intravitreal injections. Although it caused no systemic toxicity in humans or rabbits, melphalan did result in significant retinal toxicity at high doses and lower but safer
doses only achieved incomplete tumor control.42,50
Periocular: Chemotherapeutic agents may be
periocularly injected, either as subconjunctival or subtenon. Periocular injection enables transcleral drug
delivery, using the large surface area of the sclera
and its high permeability to small molecules without
the danger of puncturing the globe.51 Periocular chemotherapy achieves levels within the vitreous rapidly
and at levels 6 to 10 times higher than the intravenous
route.52 In general, periocular injection of chemotherapeutic agents has been used for retinoblastoma control
as an adjunct to systemic chemotherapy and, on occasion, to treat tumor recurrence. Either carboplatin or
topotecan can be employed. The first-line indications
for periocular chemotherapy are bilateral advanced
group D or E eyes in which a higher local dose of chemotherapy is needed, treatment of vitreous seeds, and
recurrent localized tumor. Subtenon chemotherapy using carboplatin can increase tumor control, especially
if it is coupled with intravenous chemoreduction. Subtenon carboplatin showed initial favorable results as
single therapy, but long-term follow-up revealed a high
failure rate as initial treatment; therefore, it should be
combined with other therapeutic approaches.53
Complications of periocular chemotherapy include
orbital and eyelid edema, ecchymosis, orbital fat atrophy, muscle fibrosis leading to strabismus, and optic
atrophy.53-55 Because of its significant toxic effects, periocular chemotherapy is rarely used in current practice.
Focal Therapy
Thermotherapy/Transpupillary Thermotherapy:
This type of focal therapy uses an 810-nm diode laser
directly on the tumor to heat it up to subcoagulation
temperatures of 42 to 60 °C to induce a cytotoxic effect. It is used for local control and for small (< 4.5-mm
base and 2.5-mm thickness) tumors posteriorly located
to the equator of the globe. In cases of macular or juxtapapillary tumors, consolidation should be performed
with caution so as to protect the optic disc and neurosensory papillomacular bundle. The tumor may be observed while on chemoreduction and, if unresponsive,
foveal-sparing thermotherapy may be administered. In
an analysis of 68 macular retinoblastomas treated with
Cancer Control 103
chemoreduction, tumor recurrence occurred in 17% of
those consolidated with foveal-sparing thermotherapy
compared with 35% of those observed without consolidation.56 Complications of transpupillary thermotherapy include iris atrophy, focal cataracts, tumor seeding
into the vitreous, and retinal fibrosis, transition, and
vascular occlusion.
Laser Photocoagulation: This type of therapy
uses an 520-nm argon laser to coagulate the blood
supply of the tumor by generating heat with temperatures in excess of 65 °C within the treatment spot. Direct photocoagulation to the tumor must be avoided.
A second mechanism occurring around the ring of tissue beyond the laser spot is the increase in temperature to a thermotherapeutic range (45–60 oC). In this
region, synergism exists with carboplatin, so this laser
treatment is typically performed within 24 hours of
the intravenous chemotherapy cycle. Small posterior
tumors without seeding respond well to laser photocoagulation. It is not used for tumors impinging on the
fovea, because of the risk of compromising a patient’s
central vision. In such cases, some form of chemotherapy can be used to shrink the tumor prior to the start
of definitive laser treatment. Photocoagulation is also
used to treat tumor-associated retinal neovascularization. Complications include vitreous seeding if the laser power is too high, and retinal fibrosis, traction, and
vascular occlusion.
Cryotherapy: Cryotherapy involves a cryoprobe
by which liquid nitrogen is delivered and directly
applied to the outer surface of the sclera adjacent
to the tumor. It is suitable for small (< 3.5-mm base
and < 2-mm thickness) tumors anteriorly located to the
equator of the globe. Complications of cryotherapy include retinal tears and detachment, proliferative vitreoretinopathy, and chorioretinal atrophy.
Plaque Brachytherapy: This type of treatment
refers to implantation of radioactive material on the
sclera over the base of the tumor. Tissue absorption of
ionizing radiation causes DNA damage and cell death.
Because retinoblastoma has a high rate of proliferating cells, it is quite radiosensitive. Brachytherapy is
used for solitary, medium-sized tumors (6–15 mm and
> 3 mm from the optic disc or fovea) and, if little subretinal fluid is present, plaque radiotherapy can generally be used to achieve tumor control. Iodine-125
and ruthenium-106 isotopes are the most common
source of radiation currently used in brachytherapy
for retinoblastoma to deliver 40 to 45 Gy to the apex
of the tumor. In addition to treatment for retinoblastoma, ocular brachytherapy is commonly used for uveal
melanoma, in which a clinician uses a similar surgical technique of plaque application of suturing to the
episcleral surface but uses a lower dose of radiation.
Complications of plaque brachytherapy include radiation retinopathy and optic neuropathy.18
104 Cancer Control
Consolidation alone may be used in group A and B
tumors. Bilateral cases with small, extrafoveal tumors
can be managed with focal treatments alone. In asymmetric disease, a single eye can be managed with focal
techniques and the other with enucleation or intra-arterial chemotherapy with or without systemic chemotherapy.7 The desired end point is tumor regression in
the form of a completely calcified tumor (type 1 regression) or a flat chorioretinal scar (type 4 regression).
Enucleation
Enucleation remains a favored approach in cases of extensive retinoblastoma with buphthalmos, neovascular
glaucoma, aqueous seeding, and transcleral extension
(advanced group E eyes) — particularly for unilateral
cases with no likelihood of functional vision.6 Surgery
involves careful removal of the eye with minimal globe
trauma to prevent tumor seeding into the orbit, and
then excising a long section of optic nerve for proper
histopathological evaluation.57 Following enucleation,
an orbital implant is placed to provide the socket with
enough volume and allow the 4 rectus muscles to attach for motility. Immediately after enucleation, fresh
tissue should be harvested on a separate tray for genetic studies.
A corollary to enucleation is the adequate histopathological evaluation of globe pathology, because
retinoblastoma with histological features signifying a
high risk of metastasis requires adjuvant intravenous
chemotherapy.58 High-risk features include choroidal
invasion, retrolaminar optic nerve invasion, scleral and
orbital invasion, and anterior chamber invasion. An
analysis of 1,020 study patients with high-risk features
after enucleation showed that 24% of study volunteers
at high risk developed metastasis if not treated with adjuvant chemotherapy, whereas 4% of these study patients had metastasis if treated with chemotherapy.59
The regimen for adjuvant chemotherapy is vincristine/etoposide/carboplatin for 4 to 6 cycles.
External Beam Radiotherapy
External beam radiotherapy can be used to treat eyes
unresponsive to other treatments, those with a tumor
located at the optic nerve resection margin (36 Gy to
orbit and 10 Gy boost to chiasm), and as treatment of
extraocular retinoblastoma such as those extending
through the sclera, orbit, or intracranially (40–45 Gy).18
External beam radiotherapy is rarely used and employed only when necessary because of its subsequent
risks. It is known to cause midfacial hypoplasia in
young patients and increase the risk of development of
soft-tissue sarcomas, brain tumors, osteosarcomas, and
other cancers.60 In patients with bilateral disease, the
incidence of secondary malignancy at 50 years from diagnosis of retinoblastoma is 21% in patients who were
not treated vs 38% for those who were treated with raApril 2016, Vol. 23, No. 2
diation.61-63 If possible, any radiation (including radiography, computed tomography, and external beam)
should be avoided in individuals with heritable retinoblastoma to minimize their lifetime risk of developing
secondary cancers.
Extraocular Therapy
Although their incidence is rare in developed countries, cases of extraocular disease are occasionally
seen in neglected or inadequately treated patients.64,65
Tumors may directly extend through emissary vessels
of the sclera into the orbital soft tissue surrounding
the eye. Treatment of orbital retinoblastoma includes
systemic chemotherapy and external beam radiotherapy (40–45 Gy) with a rate of cure between 60% and
85%.18 Treatment may also extend to the optic nerve
into the brain and meninges with subsequent seeding
of spinal fluid. Most recurrences occur in the central
nervous system and are associated with a low cure
rate; regimens proven to be effective include vincristine, cyclophosphamide, doxorubicin, and platinum as
well as epipodophyllotoxin-based drugs.66 No preclinical or clinical evidence supports the use of intrathecal chemotherapy. Craniospinal irradiation using 25 to
35 Gy is sometimes provided. Retinoblastoma may also
hematogenously spread to the bone marrow, bones,
lungs, or liver. For metastatic disease, multiple-agent,
high-dose chemotherapy and autologous hematopoietic stem cell transplantation can be administered as
rescue therapy.67 The Children’s Oncology Group developed a prospective, multi-institutional, international trial for metastatic retinoblastoma, and data will be
available in early 2016; that study hopes to define differences in site of metastasis and outcome.68
Emerging Therapies
New developments in the treatment of retinoblastoma
are geared toward an ideal system that is affordable
and can easily be used by ophthalmologists or ocular
oncologists — even those in developing nations — and
is locally effective to the main tumor as well as tumor
seeds, causes minimal complications, and can achieve
a sustained treatment effect to retinoblastoma without
spreading tumor cells. A sustained treatment effect is
desired because, even when compounds do reach the
target site, rapid clearance from the eye often results
in short intraocular residence times. In addition, many
patients with retinoblastoma are young children who
oftentimes require sedation to receive intraocular therapy — sustained drug-release mechanisms will enable
continuous medication dosing.
Preclinical models that accurately reflect human
disease are needed for rare childhood cancers such as
retinoblastoma because not enough patients exist for
large-scale multicenter clinical trials.8 Despite this lack
of clinical trials, rates of patient survival and ocular
April 2016, Vol. 23, No. 2
preservation have improved.7 Animal models remain
instrumental in understanding biological mechanisms
and designing targeted therapies for this disease.8-10
Novel treatment options being explored in vitro and
in vivo include alternative chemotherapeutic agents,
molecularly targeted therapies, gene therapy, localized
delivery, and the sustained-release of broad-spectrum
chemotherapy agents.
Alternative Chemotherapeutic Agents
Topotecan, a topoisomerase inhibitor that works
in the G0 phase of the cell cycle, may be used as a
single agent or in combination with other agents for
the treatment of retinoblastoma. Subconjunctival, sustained-release topotecan has been shown to decrease
tumor burden in a transgenic mouse model of retinoblastoma.69 In a similar fashion, periocular injections
of topotecan in fibrin sealant in humans have been
shown to achieve retinoblastoma tumor volume reduction.70 Topotecan can also reduce retinoblastoma
tumor burden when given systemically,71 via IAC,72
or via intravitreal injections.43 When compared with
standard chemotherapeutic agents used to treat retinoblastoma, topotecan avoids the retinal toxicity of
melphalan, the nephrotoxicity of carboplatin, and the
hematotoxicity of etoposide.73 Topotecan is a good
candidate drug for intravitreal injection because it has
minimal retinal toxicity and achieves maximum concentration levels 50 times higher than systemic exposure.73,74 Our laboratory has shown that 20-µg topotecan can provide sustained retinoblastoma cell-line
killing in vitro,75 similar to the clinically demonstrated
retinoblastoma cell-line killing ability with 20-µg intravitreal topotecan.43
Anthracyclines are a class of chemotherapeutic drugs that inhibit topoisomerase and intercalate
DNA. They are used to treat several cancers and are
active against retinoblastoma; however, because of
their limited blood–brain barrier penetration, their
use is typically limited to extraocular and metastatic retinoblastoma. The anthracycline idarubicin has
been shown to be effective against retinoblastoma tumor cells in bone marrow (response rate, 60%), but
sufficient concentrations have not been achieved at
target sites (eg, central nervous system).76 A study to
improve intraocular exposure of the anthracycline
doxorubicin while also reducing systemic exposure
to the drug was successful in maintaining sustained
levels of doxorubicin after the intravitreal injection of
the drug encapsulated in polyhydroxybutyrate microspheres in rabbits.77
Molecularly Targeted Therapy
RB1 was the first tumor suppressor gene identified and
cloned, yet no effective, molecularly targeted cure currently exists for retinoblastoma.78 This could perhaps
Cancer Control 105
be attributed to the variety of unknown secondary mutations and differential expression of other genes aside
from RB1 involved in tumorigenesis, as well as questions on the cellular origin of retinoblastoma.79,80 Significant progress has been made in our understanding
of tumor biology, leading to the discovery and development of small molecules for the treatment of retinoblastoma, including novel, targeted therapeutics such
as inhibitors of the MDMX–p53 response (nutlin-3a),
histone deacetylase (HDAC) inhibitors, and spleen tyrosine kinase (SYK) inhibitors.9,81
Nutlin-3 is an imidazole analogue that plays a role
in the activation of p53, a tumor suppressor protein.
It is a small molecule inhibitor of MDM2 and MDMX
and prevents the association of both these proteins
with p53. Studies have shown that nutlin-3 restores
the p53 pathway in retinoblastoma cells lacking both
retinoblastoma protein and p53 activity, thus inducing
p53-mediated apoptosis.82 Subconjunctival injection of
nutlin-3 in mouse models of retinoblastoma resulted
in reduced tumor burden when used in combination
with topotecan, demonstrating a synergistic effect.83
Nutlin-3 is being studied in phase 1 clinical trials for
the treatment of retinoblastoma.82,83
Although it is not expressed in normal human retinas, SYK was found to be upregulated in retinoblastoma tumor samples and has been shown to be required
for retinoblastoma cell survival.84 Inhibition of SYK
with BAY-61-3606 and R406 caused cell death in retinoblastoma, and an in vivo study of orthotopic, xenograft mice showed that subconjunctival BAY-61-3606
was effective at blocking retinoblastoma cell proliferation.84 In addition, studies of SYK inhibition have also
implicated several downstream signaling molecules as
mediators of SYK survival, including the B-cell chronic
lymphocytic leukemia/lymphoma 2 (BCL2) family of
proteins.85 Myeloid cell leukemia 1, a member of the
BCL2 family, is a potential target because it is upregulated in retinoblastoma and is being developed as a
therapy for other cancers.86
Another class of targeted therapies in phase 1
clinical trials are the inhibitors of HDAC.87 Cells with
elevated E2F1 activity are uniquely sensitive to inhibitors of HDAC through the overexpression of proapoptotic factors. Cells lacking retinoblastoma protein
have increased E2F1 activity, and retinoblastoma-derived cell lines have demonstrated particular sensitivity to apoptosis induced from inhibitors of HDAC.88
In a preclinical trial, inhibitors of HDAC slowed the
growth of retinoblastoma-derived tumors in both
transgenic and xenograft murine models of retinoblastoma, with minimal off-target effects, suggesting
that inhibitors of HDAC may specifically inhibit the
proliferation of retinoblastoma tumor cells and, thus,
have lower systemic toxicities relative to chemotherapeutic agents.89
106 Cancer Control
Gene Therapy
Suicide gene therapy is based on the introduction of a
viral or bacterial gene into tumor cells, thus allowing
the conversion of a nontoxic compound into a lethal
drug to kill the tumor cells. In a phase 1 study, intravitreal injections of an adenovirus vector containing
HSVtk followed by treatment with ganciclovir were
shown to be safe and effective against vitreous seeds.90
Although these results were promising, it may be unlikely that this type of gene therapy could be used as
first-line treatment for retinoblastoma but rather its use
may be better suited as an adjunct to standard therapy
for the treatment of refractory vitreous seeds.90
Local Drug-Delivery Systems
Nanotechnology-based drug-delivery systems for the
treatment of cancer have significantly evolved during
the past decade by enabling options for site-specific
delivery and increased bioavailability.91 Investigations
have been conducted on the potential use of different
materials as intraocular drug carriers, such as biodegradable polyesters, dendrimers, liposomes, mesoporous silica, and gold nanoparticles.91 These particles
can be modified to target specific cells, and they can be
designed to ensure sustained drug release to increase
the therapeutic effectiveness of the drug molecules
they contain.92 Nanoparticle-based systems designed
for the treatment of retinoblastoma have improved
rates of drug delivery to the posterior segment of the
eye and have increased the intravitreal half-life of chemotherapy agents, thus highlighting their potential
in treatment of this cancer.93 The intravitreal delivery
of doxorubicin-loaded, polyester-based microspheres
in the ocular tissue from rabbits exhibited reduced
rates of toxicity to surrounding normal structures.77
Dendrimer nanoparticles containing carboplatin significantly reduced tumor volume compared with free
carboplatin in a retinoblastoma mouse model.94 Polylactic glycolic acid nanoparticles developed to deliver
doxorubicin95 and etoposide96 were tested on Y79 retinoblastoma cell lines and may be potential candidates
for sustained drug-release models.
Gold-based nanoparticles have unique physical properties, including a strong ability to absorb
near-infrared light, which enables the photothermal
destruction of cancer cells.97 Light-activated drug release can be attained using gold nanoparticles conjugated with chemotherapeutic agents.98 For example,
gold nanocages are encapsulated with a smart polymer, wherein the nanocages absorb light converted
to heat, thus causing the collapse of the polymer and
the subsequent release of doxorubicin.99 The light-responsiveness of gold and other photosensitive nanocarriers infers considerable potential for ophthalmic
conditions such as retinoblastoma because of the regular use of lasers in the treatment of retinal disease
April 2016, Vol. 23, No. 2
and the added benefit of facilitating the controlled
timing and location of delivery of therapeutic agents.
Gold nanoparticles can also cross the blood–retina
barrier without significant rates of cytotoxicity.100 The
intravitreal injection of gold-tethered liposomes and
viral-like nanoparticles containing topotecan is being
investigated in a rabbit model of retinoblastoma that
develops vitreous seeds.55,101
Another injectable agent being studied as a delivery system of chemotherapeutic agents for the treatment of retinoblastoma is the biodegradable carrier
fibrin glue.102 Carboplatin103 and topotecan69 released
from fibrin depots have both been shown to retain
their bioactivity against retinoblastoma cells in culture. Carboplatin released from a fibrin depot was also
shown to decrease tumor burden in a transgenic murine model of retinoblastoma cells.104 In another report,
fibrin sealants sustained delivery of carboplatin in ocular tissues compared with carboplatin in plain solution,
which clears rapidly in vivo.105 Promising results have
been reported for topotecan combined with fibrin in
clinical studies.71,106
roidal and retinal concentrations targeted toward the
elimination of subretinal seeds and the prevention of
choroidal invasion, with minimal biodistribution and
toxicity to surrounding tissue. Our laboratory is investigating the suprachoroidal and intravitreal injection of topotecan using microneedles in a rabbit model of retinoblastoma (Fig 2).5
Conclusions
Management of retinoblastoma is individualized according to patient characteristics and preferences, with
the main goals being to save the patient’s life and to
preserve useful vision, if possible. Therapeutic options for retinoblastoma have rapidly evolved in recent
years, with a paradigm shift in standard treatment protocols toward the targeted delivery of chemotherapeutic agents. Increased use of intra-arterial and intravitreal types of chemotherapy with focal consolidation,
along with the reduced use of systemic chemotherapy,
enucleation, and external beam radiotherapy, has contributed to improved rates of survival and globe salvage while also minimizing unwanted treatment-related adverse events. However, these approaches are not
without complications, and a need still exists to weigh
the risks and benefits of treatment and to evaluate their
long-term effects. New therapies being investigated include novel carriers and routes for local drug delivery
as well as molecularly targeted therapies. Our under-
Suprachoroidal Injection
Suprachoroidal injection with microneedles is a route
of targeted drug delivery to the choroid and retina that
poses minimal risk of extraocular tumoral spread, because the vitreous and subretinal spaces are not entered. In a rabbit model, bevacizumab was determined to be
A
B
safe and effective when injected
into the suprachoroidal space;
high concentrations were found
in the posterior segment tissues.107 Microneedles are 0.8- to
1.0-mm long, 32-gauge needles
with a short bevel that penetrate
through the sclera into the supraciliary space. When the microneedle injects compounds
into the supraciliary space, these
C
D
compounds posteriorly spread
into the suprachoroidal space.108
In phase 1 clinical trials, microneedles for suprachoroidal
injections have been used to inject triamcinolone into the suprachoroidal space in patients with
uveitis and macular edema following retinal venous occlusion
(NCT02303184, NCT01789320).
The suprachoroidal delivery of
Fig 2A–D. — Intravitreal injection of topotecan in a rabbit model of retinoblastoma. (A) Vitreous
seeds (asterisks) form in this retinoblastoma rabbit model. (B) A 32-gauge needle is inserted into the
chemotherapeutic agents is a
pars plana. (C) The needle (arrows) is inserted into its hub and topotecan is injected. (D) Vitreous
promising approach for appliseeds have disappeared after 3 weekly injections of 20 µg topotecan.
cations in retinoblastoma, and,
Reprinted from Grossniklaus HE. Retinoblastoma. Fifty years of progress. The LXXI Edward Jackson
ideally, will result in high choMemorial Lecture. Am J Ophthalmol. 2014;158(5):875-891, with permission from Elsevier.
April 2016, Vol. 23, No. 2
Cancer Control 107
standing of tumor biology and how to develop safe and
effective therapy continues to progress, thus helping to
ensure that retinoblastoma will be a curable childhood
cancer worldwide in the future.
References
1. Broaddus E, Topham A, Singh AD. Incidence of retinoblastoma in
the USA: 1975-2004. Br J Ophthalmol. 2009;93(1):21-23.
2. Parkin DM, Stiller CA, Draper GJ, et al. The international incidence
of childhood cancer. Int J Cancer. 1988;42(4):511-520.
3. Tamboli D, Topham A, Singh N, et al. Retinoblastoma: a SEER
dataset evaluation for treatment patterns, survival, and second malignant
neoplasms. Am J Ophthalmol. 2015;160(5):953-958.
4. Broaddus E, Topham A, Singh AD. Survival with retinoblastoma in
the USA: 1975-2004. Br J Ophthalmol. 2009;93(1):24-27.
5. Grossniklaus HE. Retinoblastoma. Fifty years of progress. The LXXI
Edward Jackson Memorial Lecture. Am J Ophthalmol. 2014;158(5):875-891.
6. Canturk S, Qaddoumi I, Khetan V et al. Survival of retinoblastoma
in less-developed countries impact of socioeconomic and health-related
indicators. Br J Ophthalmol. 2010;94(11):1432-1436.
7. Abramson DH, Shields CL, Munier FL, et al. Treatment of retinoblastoma in 2015: agreement and disagreement. JAMA Ophthalmol.
2015;133(11):1341-1347.
8. Mendoza PR, Grossniklaus HE. The biology of retinoblastoma.
Prog Mol Biol Transl Sci. 2015;134:503-516.
9.Pritchard EM, Dyer MA, Guy RK. Progress in small molecule
therapeutics for the treatment of retinoblastoma. Mini Rev Med Chem.
2016;16(6):430-454.
10. Chévez-Barrios P, Hurwitz MY, Louie K et al. Metastatic and nonmetastatic models of retinoblastoma. Am J Pathol. 2000;157(4):1405-1412.
11. Shields CL, Lally SE, Leahey AM, et al. Targeted retinoblastoma
management: when to use intravenous, intra-arterial, periocular, and intravitreal chemotherapy. Curr Opin Ophthalmol. 2014;25(5):374-385.
12. Shields CL, Shields JA. Retinoblastoma management: advances in
enucleation, intravenous chemoreduction, and intra-arterial chemotherapy.
Curr Opin Ophthalmol. 2010;21(3):203-212.
13. Linn Murphree A. Intraocular retinoblastoma: the case for a new
group classification. Ophthalmol Clin North Am. 2005;18(1):41-53.
14. Shields CL, Mashayekhi A, Au AK, et al. The International classification of retinoblastoma predicts chemoreduction success. Ophthalmology.
2006;113(12):2276-2280.
15. Edge S, Byrd DR, Compton CC, et al, eds. AJCC Cancer Staging
Manual. 7th ed. New York: Springer; 2010.
16. Kaliki S, Shields CL, Rojanaporn D, et al. High-risk retinoblastoma
based on international classification of retinoblastoma: analysis of 519 enucleated eyes. Ophthalmology. 2013;120(5):997-1003.
17. Ramasubramanian A, Kytasty C, Meadows AT, et al. Incidence of
pineal gland cyst and pineoblastoma in children with retinoblastoma during
the chemoreduction era. Am J Ophthalmol. 2013;156(4):825-829.
18. Rodriguez-Galindo C, Chantada GL, Haik BG, et al. Treatment of
retinoblastoma: current status and future perspectives. Curr Treat Options
Neurol. 2007;9(4):294-307.
19. Kingston JE, Hungerford JL, Madreperla SA, et al. Results of combined chemotherapy and radiotherapy for advanced intraocular retinoblastoma. Arch Ophthalmol. 1996;114(11):1339-1343.
20. Shields CL, Honavar SG, Meadows AT, et al. Chemoreduction
plus focal therapy for retinoblastoma: factors predictive of need for treatment with external beam radiotherapy or enucleation. Am J Ophthalmol.
2002;133(5):657-664.
21. Sullivan EM, Wilson MW, Billups CA, et al. Pathologic risk-based
adjuvant chemotherapy for unilateral retinoblastoma following enucleation. J Pediatr Hematol Oncol. 2014;36(6):e335-340.
22. Chantada GL, Dunkel IJ, de Davila MT, et al. Retinoblastoma patients
with high risk ocular pathological features: who needs adjuvant therapy? Br J
Ophthalmol. 2004;88(8):1069-1073.
23. Short BG. Safety evaluation of ocular drug delivery formulations:
techniques and practical considerations. Toxicol Pathol. 2008;36(1):49-62.
24. Hwang CK, Aaberg TM Jr, Chevez-Barrios P, et al. Residual intraretinal retinoblastoma after chemoreduction failure. Arch Ophthalmol.
2012;130(2):246-248.
25. Demirci H, Eagle RC Jr, Shields CL, et al. Histopathologic findings in
eyes with retinoblastoma treated only with chemoreduction. Arch Ophthalmol.
2003;121(8):1125-1131.
26. Inomata M, Kaneko A. Chemosensitivity profiles of primary and cultured human retinoblastoma cells in a human tumor clonogenic assay. Jpn J
Cancer Res. 1987;78(8):858-868.
27. Yamane T, Kaneko A, Mohri M. The technique of ophthalmic arterial infusion therapy for patients with intraocular retinoblastoma. Int J Clin
Oncol. 2004;9(2):69-73.
28. Suzuki S, Yamane T, Mohri M, et al. Selective ophthalmic arterial
108 Cancer Control
injection therapy for intraocular retinoblastoma: the long-term prognosis.
Ophthalmology. 2011;118(10):2081-2087.
29. Abramson DH, Dunkel IJ, Brodie SE, et al. A phase I/II study of direct
intraarterial (ophthalmic artery) chemotherapy with melphalan for intraocular
retinoblastoma initial results. Ophthalmology. 2008;115(8):1398-1404.
30. Shields CL, Manjandavida FP, Lally SE, et al. Intra-arterial chemotherapy for retinoblastoma in 70 eyes: outcomes based on the international
classification of retinoblastoma. Ophthalmology. 2014;121(7):1453-1460.
31. Abramson DH, Daniels AB, Marr BP, et al. Intra-arterial chemotherapy
(ophthalmic artery chemosurgery) for group D retinoblastoma. PLoS One.
2016;11(1):e0146582.
32. Shields CL, Kaliki S, Shah SU, et al. Effect of intraarterial chemotherapy on retinoblastoma-induced retinal detachment. Retina. 2012;32(4):799-804.
33. Palioura S, Gobin YP, Brodie SE, et al. Ophthalmic artery chemosurgery for the management of retinoblastoma in eyes with extensive
(>50%) retinal detachment. Pediatr Blood Cancer. 2012;59(5):859-864.
34. Gobin YP, Dunkel IJ, Marr BP, et al. Combined, sequential intravenous and intra-arterial chemotherapy (bridge chemotherapy) for young
infants with retinoblastoma. PLoS One. 2012;7(9):e44322.
35. Tse BC, Steinle JJ, Johnson D, et al. Superselective intraophthalmic artery chemotherapy in a nonhuman primate model: histopathologic
findings. JAMA Ophthalmol. 2013;131(7):903-911.
36. Steinle JJ, Zhang Q, Thompson KE, et al. Intra-ophthalmic artery
chemotherapy triggers vascular toxicity through endothelial cell inflammation and leukostasis. Invest Ophthalmol Vis Sci. 2012;53(4):2439-2445.
37. Vajzovic LM, Murray TG, Aziz-Sultan MA, et al. Supraselective
intra-arterial chemotherapy: evaluation of treatment-related complications
in advanced retinoblastoma. Clin Ophthalmol. 2011;5:171-176.
38. Jabbour P, Chalouhi N, Tjoumakaris S, et al. Pearls and pitfalls
of intraarterial chemotherapy for retinoblastoma. J Neurosurg Pediatr.
2012;10(3):175-181.
39. Aziz HA, Lasenna CE, Vigoda M, et al. Retinoblastoma treatment
burden and economic cost: impact of age at diagnosis and selection of
primary therapy. Clin Ophthalmol. 2012;6:1601-1606.
40. Zanaty M, Barros G, Chalouhi N. Update on intra-arterial chemotherapy for retinoblastoma. Sci World J. 2014;2014:869604.
41. Zhao J, Dimaras H, Massey C, et al. Pre-enucleation chemotherapy
for eyes severely affected by retinoblastoma masks risk of tumor extension
and increases death from metastasis. J Clin Oncol. 2011;29(7):845-851.
42. Munier FL, Soliman S, Moulin AP, et al. Profiling safety of intravitreal injections for retinoblastoma using an anti-reflux procedure and sterilisation of the needle track. Br J Ophthalmol. 2012;96(8):1084-1087.
43. Ghassemi F, Shields CL, Ghadimi H, et al. Combined intravitreal
melphalan and topotecan for refractory or recurrent vitreous seeding from
retinoblastoma. JAMA Ophthalmol. 2014;132(8):936-941.
44.Ghassemi F, Shields CL. Intravitreal melphalan for refractory
or recurrent vitreous seeding from retinoblastoma. Arch Ophthalmol.
2012;130(10):1268-1271.
45. Kaneko A, Suzuki S. Eye-preservation treatment of retinoblastoma
with vitreous seeding. Jpn J Clin Oncol. 2003;33(12):601-607.
46. Smith SJ, Smith BD. Evaluating the risk of extraocular tumour
spread following intravitreal injection therapy for retinoblastoma: a systematic review. Br J Ophthalmol. 2013;97(10):1231-1236.
47. Munier FL, Gaillard MC, Balmer A, et al. Intravitreal chemotherapy
for vitreous seeding in retinoblastoma: Recent advances and perspectives. Saudi J Ophthalmol. 2013;27(3):147-150.
48. Seregard S, Singh AD. Retinoblastoma: direct chemotherapeutic
drug delivery into the vitreous cavity. Br J Ophthalmol. 2012;96(4):473-474.
49. Suzuki S, Aihara Y, Fujiwara M, et al. Intravitreal injection of melphalan for intraocular retinoblastoma. Jpn J Ophthalmol. 2015;59(3):164-172.
50. Francis JH, Schaiquevich P, Buitrago E, et al. Local and systemic
toxicity of intravitreal melphalan for vitreous seeding in retinoblastoma: a
preclinical and clinical study. Ophthalmology. 2014;121(9):1810-1817.
51. Geroski DH, Edelhauser HF. Transscleral drug delivery for posterior
segment disease. Adv Drug Deliv Rev. 2001;52(1):37-48.
52. Hayden BC, Jockovich ME, Murray TG, et al. Pharmacokinetics of
systemic versus focal Carboplatin chemotherapy in the rabbit eye: possible
implication in the treatment of retinoblastoma. Invest Ophthalmol Vis Sci.
2004;45(10):3644-3649.
53. Abramson DH, Frank CM, Dunkel IJ. A phase I/II study of subconjunctival carboplatin for intraocular retinoblastoma. Ophthalmology.
1999;106(10):1947-1950.
54. Mulvihill A, Budning A, Jay V, et al. Ocular motility changes after
subtenon carboplatin chemotherapy for retinoblastoma. Arch Ophthalmol.
2003;121(8):1120-1124.
55. Schmack I, Hubbard GB, Kang SJ, et al. Ischemic necrosis and atrophy of the optic nerve after periocular carboplatin injection for intraocular
retinoblastoma. Am J Ophthalmol. 2006;142(2):310-315.
56. Shields CL, Mashayekhi A, Cater J, et al. Macular retinoblastoma
managed with chemoreduction: analysis of tumor control with or without adjuvant thermotherapy in 68 tumors. Arch Ophthalmol. 2005;123(6):765-773.
57. Schiedler V, Dubovy SR, Murray TG. Snare technique for enucleation of eyes with advanced retinoblastoma. Arch Ophthalmol.
2007;125(5):680-683.
April 2016, Vol. 23, No. 2
58. Sastre X, Chantada GL, Doz F, et al. Proceedings of the consensus
meetings from the International Retinoblastoma Staging Working Group on
the pathology guidelines for the examination of enucleated eyes and evaluation of prognostic risk factors in retinoblastoma. Arch Pathol Lab Med.
2009;133(8):1199-1202.
59. Honavar SG, Singh AD, Shields CL, et al. Postenucleation adjuvant
therapy in high-risk retinoblastoma. Arch Ophthalmol. 2002;120(7):923-931.
60. Imhof SM, Mourits MP, Hofman P, et al. Quantification of orbital and
mid-facial growth retardation after megavoltage external beam irradiation
in children with retinoblastoma. Ophthalmology. 1996;103(2):263-268.
61.Wong FL, Boice JD Jr, Abramson DH, et al. Cancer incidence after retinoblastoma. Radiation dose and sarcoma risk. JAMA.
1997;278(15):1262-1267.
62. Marees T, Moll AC, Imhof SM, et al. Risk of second malignancies in
survivors of retinoblastoma: more than 40 years of follow-up. J Natl Cancer
Inst. 2008;100(24):1771-1779.
63. Wong JR, Morton LM, Tucker MA, et al. Risk of subsequent malignant neoplasms in long-term hereditary retinoblastoma survivors after
chemotherapy and radiotherapy. J Clin Oncol. 2014;32(29):3284-3290.
64. Ali MJ, Honavar SG, Reddy VA. Orbital retinoblastoma: present status
and future challenges - a review. Saudi J Ophthalmol. 2011;25(2):159-167.
65. Truong B, Green AL, Friedrich P, et al. Ethnic, racial, and socioeconomic disparities in retinoblastoma. JAMA Pediatr. 2015;169(12):1096-1104.
66. Radhakrishnan V, Kashyap S, Pushker N, et al. Outcome, pathologic findings, and compliance in orbital retinoblastoma (International Retinoblastoma Staging System stage III) treated with neoadjuvant chemotherapy: a prospective study. Ophthalmology. 2012;119(7):1470-1477.
67. Dunkel IJ, Chan HS, Jubran R, et al. High-dose chemotherapy with
autologous hematopoietic stem cell rescue for stage 4B retinoblastoma.
Pediatr Blood Cancer. 2010;55(1):149-152.
68. Children’s Oncology Group website. ARET0321: phase III study for
children diagnosed with extra-ocular retinoblastoma. https://childrensoncologygroup.org/index.php/aret0321. Accessed April 4, 2016.
69. Tsui JY, Dalgard C, Van Quill KR, et al. Subconjunctival topotecan in
fibrin sealant in the treatment of transgenic murine retinoblastoma. Invest
Ophthalmol Vis Sci. 2008;49(2):490-496.
70. Mallipatna AC, Dimaras H, Chan HS, et al. Periocular topotecan for
intraocular retinoblastoma. Arch Ophthalmol. 2011;129(6):738-745.
71. Chantada GL, Fandino AC, Casak SJ, et al. Activity of topotecan in
retinoblastoma. Ophthalmic Genet. 2004;25(1):37-43.
72. Francis JH, Gobin YP, Dunkel IJ, et al. Carboplatin +/- topotecan
ophthalmic artery chemosurgery for intraocular retinoblastoma. PLoS One.
2013;8(8):e72441.
73. Buitrago E, Del Sole MJ, Torbidoni A, et al. Ocular and systemic
toxicity of intravitreal topotecan in rabbits for potential treatment of retinoblastoma. Exp Eye Res. 2013;108:103-109.
74. Buitrago E, Hocht C, Chantada G, et al. Pharmacokinetic analysis
of topotecan after intra-vitreal injection. Implications for retinoblastoma
treatment. Exp Eye Res. 2010;91(1):9-14.
75. Jijelava K, Kompella U, Durazo S, et al. Evaluation of the efficacy of
topotecan Au-tethered liposomes and AU-011 for the treatment of retinoblastoma in vitro. Invest Ophthamol Visual Sci. 2014. http://iovs.arvojournals.org/article.aspx?articleid=2268458. Accessed March 29, 2016.
76. Chantada GL, Fandino A, Mato G, et al. Phase II window of idarubicin in
children with extraocular retinoblastoma. J Clin Oncol. 1999;17(6):1847-1850.
77. Hu T, Le Q, Wu Z, et al. Determination of doxorubicin in rabbit ocular tissues and pharmacokinetics after intravitreal injection of a single dose
of doxorubicin-loaded poly-beta-hydroxybutyrate microspheres. J Pharm
Biomed Anal. 2007;43(1):263-269.
78. Friend SH, Bernards R, Rogelj S, et al. A human DNA segment with
properties of the gene that predisposes to retinoblastoma and osteosarcoma. Nature. 1986;323(6089):643-646.
79. Corson TW, Gallie BL. One hit, two hits, three hits, more? Genomic
changes in the development of retinoblastoma. Genes Chromosomes
Cancer. 2007;46(7):617-634.
80. Kapatai G, Brundler MA, Jenkinson H, et al. Gene expression
profiling identifies different sub-types of retinoblastoma. Br J Cancer.
2013;109(2):512-525.
81. Sachdeva UM, O’Brien JM. Understanding pRb: toward the necessary development of targeted treatments for retinoblastoma. J Clin Invest.
2012;122(2):425-434.
82. Laurie NA, Donovan SL, Shih CS, et al. Inactivation of the p53 pathway in retinoblastoma. Nature. 2006;444(7115):61-66.
83.Brennan RC, Federico S, Bradley C, et al. Targeting the p53
pathway in retinoblastoma with subconjunctival Nutlin-3a. Cancer Res.
2011;71(12):4205-4213.
84. Zhang J, Benavente CA, McEvoy J, et al. A novel retinoblastoma therapy from genomic and epigenetic analyses. Nature. 2012;481(7381):329-334.
85. Gobessi S, Laurenti L, Longo PG, et al. Inhibition of constitutive and
BCR-induced Syk activation downregulates Mcl-1 and induces apoptosis
in chronic lymphocytic leukemia B cells. Leukemia. 2009;23(4):686-697.
86. Azmi AS, Wang Z, Philip PA, et al. Emerging Bcl-2 inhibitors for the
treatment of cancer. Expert Opin Emerg Drugs. 2011;16(1):59-70.
87. Falkenberg KJ, Johnstone RW. Histone deacetylases and their inApril 2016, Vol. 23, No. 2
hibitors in cancer, neurological diseases and immune disorders. Nat Rev
Drug Discov. 2014;13(9):673-691.
88. Zhao Y, Tan J, Zhuang L, et al. Inhibitors of histone deacetylases target the Rb-E2F1 pathway for apoptosis induction through activation of proapoptotic protein Bim. Proc Natl Acad Sci U S A. 2005;102(44):16090-10695.
89. Dalgard CL, Van Quill KR, O’Brien JM. Evaluation of the in vitro and
in vivo antitumor activity of histone deacetylase inhibitors for the therapy of
retinoblastoma. Clin Cancer Res. 2008;14(10):3113-3123.
90. Chevez-Barrios P, Chintagumpala M, Mieler W, et al. Response
of retinoblastoma with vitreous tumor seeding to adenovirus-mediated delivery of thymidine kinase followed by ganciclovir. J Clin Oncol.
2005;23(31):7927-7935.
91. Bertrand N, Wu J, Xu X,et al. Cancer nanotechnology: the impact of
passive and active targeting in the era of modern cancer biology. Adv Drug
Deliv Rev. 2014;66:2-25.
92. Salmaso S, Caliceti P. Stealth properties to improve therapeutic efficacy of drug nanocarriers. J Drug Deliv. 2013;2013:374252.
93. Bhavsar D, Subramanian K, Sethuraman S, et al. Management of
retinoblastoma: opportunities and challenges. Drug Deliv. 2015:1-9.
94. Kang SJ, Durairaj C, Kompella UB, et al. Subconjunctival nanoparticle carboplatin in the treatment of murine retinoblastoma. Arch Ophthalmol.
2009;127(8):1043-1047.
95. Boddu SH, Jwala J, Chowdhury MR, et al. In vitro evaluation of
a targeted and sustained release system for retinoblastoma cells using
doxorubicin as a model drug. J Ocul Pharmacol Ther. 2010;26(5):459-468.
96. Mitra M, Dilnawaz F, Misra R, et al. Toxicogenomics of nanoparticulate delivery of etoposide: potential impact on nanotechnology in retinoblastoma therapy. Cancer Nanotechnol. 2011;2(1-6):21-36.
97. Choi WI, Sahu A, Kim YH, et al. Photothermal cancer therapy and
imaging based on gold nanorods. Ann Biomed Eng. 2012;40(2):534-546.
98. Menon JU, Jadeja P, Tambe P, et al. Nanomaterials for photo-based
diagnostic and therapeutic applications. Theranostics. 2013;3(3):152-166.
99. Yavuz MS, Cheng Y, Chen J, et al. Gold nanocages covered by
smart polymers for controlled release with near-infrared light. Nat Mater.
2009;8(12):935-939.
100.Kim JH, Kim JH, Kim KW, et al. Intravenously administered
gold nanoparticles pass through the blood-retinal barrier depending on the particle size, and induce no retinal toxicity. Nanotechnology.
2009;20(50):505101.
101. Kang SJ, Grossniklaus HE. Rabbit model of retinoblastoma. J Biomed
Biotechnol. 2011;2011:394730.
102. Martin NE, Kim JW, Abramson DH. Fibrin sealant for retinoblastoma: where are we? J Ocul Pharmacol Ther. 2008;24(5):433-438.
103. Gorodetsky R, Peylan-Ramu N, Reshef A, et al. Interactions of
carboplatin with fibrin(ogen), implications for local slow release chemotherapy. J Control Release. 2005;102(1):235-245.
104. Van Quill KR, Dioguardi PK, Tong CT, et al. Subconjunctival carboplatin in fibrin sealant in the treatment of transgenic murine retinoblastoma.
Ophthalmology. 2005;112(6):1151-1158.
105.Simpson AE, Gilbert JA, Rudnick DE, et al. Transscleral diffusion of carboplatin: an in vitro and in vivo study. Arch Ophthalmol.
2002;120(8):1069-1074.
106. Yousef YA, Halliday W, Chan HS, et al. No ocular motility complications after subtenon topotecan with fibrin sealant for retinoblastoma. Can J
Ophthalmol. 2013;48(6):524-528.
107. Patel SR, Berezovsky DE, McCarey BE, et al. Targeted administration into the suprachoroidal space using a microneedle for drug
delivery to the posterior segment of the eye. Invest Ophthalmol Vis Sci.
2012;53(8):4433-4441.
108. Kim YC, Oh KH, Edelhauser HF, et al. Formulation to target delivery
to the ciliary body and choroid via the suprachoroidal space of the eye using microneedles. Eur J Pharm Biopharm. 2015;95(part B):398-406.
Cancer Control 109
Treatment regimens for
primary vitreoretinal lymphoma
continue to evolve.
Photo courtesy of Lynn E. Harman, MD. Manhattan.
Primary Vitreoretinal Lymphoma: Management of Isolated
Ocular Disease
Matthew T. Witmer MD
Background: The prognosis for patients with primary vitreoretinal is dismal. The close association of primary
vitreoretinal lymphoma with primary central nervous system lymphoma is responsible for high rates of mortality. Traditional treatments consist of systemic chemotherapy and whole-brain radiotherapy. The optimal
approach for the treatment of isolated primary vitreoretinal lymphoma is unclear.
Methods: A review of the relevant medical and scientific literature was performed, focusing on the clinical features of primary vitreoretinal lymphoma and the progress made in the management of isolated ocular disease.
Results: Ocular treatment options for primary vitreoretinal lymphoma have recently expanded with the addition of intravitreal chemotherapeutic agents and localized radiation. Based on several retrospective reports,
a general shift has been made toward local therapy (eg, orbital radiotherapy, intravitreal chemotherapy) for
ocular disease. No prospective, randomized clinical trials yet exist to guide therapy.
Conclusions: Optimal treatment regimens for isolated primary vitreoretinal lymphoma continue to evolve.
Further investigations into novel therapies and protocols are needed to decrease recurrence rates, reduce or
prevent central nervous system involvement, and improve rates of overall survival.
Introduction
Primary vitreoretinal lymphoma (formerly known as
primary intraocular lymphoma or reticulum cell sarcoma) continues to have a poor prognosis.1,2 Between
65% and 90% of patients with primary vitreoretinal
lymphoma will develop primary central nervous system (CNS) lymphoma, a disease that ultimately results
in a high rate of mortality.1,2 Several different treatment modalities can be used for these patients, each
with varying degrees of success. Due to the rarity of
From the Retina Centers of Washington, Rockville, Maryland.
Address correspondence to Matthew T. Witmer, MD,
15020 Shady Grove Road, Suite 302, Rockville, MD 20850.
E-mail: [email protected]
Submitted July 13, 2015; accepted November 27, 2015.
No significant relationship exists between the author and the companies/organizations whose products or services may be referenced in this article.
110 Cancer Control
primary vitreoretinal lymphoma, no prospective, randomized controlled trials exist to guide therapeutic decisions. Consequently, no consensus exists regarding
the optimal treatment regimen.
Pathogenesis
Primary vitreoretinal lymphoma is most often (> 95% of
the time) diffuse large B-cell lymphoma, which is a type
of high-grade malignancy.3 The malignant cells are typically found in the vitreous, retina, or optic nerve head;
however, the anatomical site of origin in many cases is
unknown, as spillover into multiple compartments of
the eye can occur early. Some have suggested a solitary,
systemic, primary source of the malignant cells followed
by intraocular metastases,4 whereas others have suggested that the malignancy has multiple, independent
sites of origin (eg, eye, CNS) that can occasionally develop simultaneously.5 Infiltration of malignant lymphoApril 2016, Vol. 23, No. 2
cytes into the eye or brain from the systemic circulation may be due to the ability of a malignant clone of
cells, which expresses specific cell-surface molecules,
to selectively home in to CNS tissue.6 Signaling molecules, such as chemokines, may direct these cells to the
CNS.7 This theory is supported by evidence of tumor
clones related to primary CNS lymphoma found in the
blood and bone marrow of affected patients.8 The CNS
may provide an attractive microenvironment for these
cells to colonize.
Close association exists between primary vitreoretinal lymphoma and primary CNS lymphoma. Approximately 20% of patients with primary CNS lymphoma develop lymphoma in the eye, and as many as
90% of patients with primary vitreoretinal lymphoma
develop CNS disease.6 The similarities between primary vitreoretinal lymphoma and primary CNS lymphoma include a unique affinity for CNS tissue (whereby
this rarely appears outside the CNS), a worse prognosis
than most extranodal types of non-Hodgkin lymphoma (NHL), and a high response rate to methotrexate.6
Historically, the treatment strategies for the 2 malignancies overlapped due to these similarities and the
frequency with which they clinically coexisted.
Although they are pathologically similar in appearance, evidence suggests that primary vitreoretinal
lymphoma differs from systemic diffuse large B-cell
lymphoma in its origin from mature B cells due to differing frequency of expressed immunoglobulins.9
epithelium (RPE) lesions, or a combination of both.
The sub-RPE lesions may appear as large RPE detachments with classic, creamy, or yellow colors. Others
have described punctate or spiculated sub-RPE deposits, also referred to as a leopard-spot pattern of
pigmentation.2 In more advanced cases, malignant
cells can seed the anterior chamber, where they can
settle in the angle or accumulate on the surface of the
iris or lens. Less often, the tumor infiltrates the optic
nerve head, where it presents as a mass lesion or results in ocular ischemia by interfering with the posterior circulation of the eye.
Diagnosis
Diagnosis can be difficult owing to its insidious onset,
tendency to simulate other conditions, and its relative
rarity.6,10 It is estimated that approximately 380 cases
are diagnosed in the United States each year.6 The median age of diagnosis is 60 years and immunosuppression increases the risk of developing the condition.11,12
Delay in diagnosis, often confounded with prior diagnoses and treatments for uveitis, is common. In a large
series, the median time to diagnosis was 6 months.10
However, delays in diagnosis as long as 24 months are
not unusual.13,14 The diagnosis of primary vitreoretinal
lymphoma is usually based on a cytological examination of a vitreous specimen on biopsy, supplemented
with findings from immunohistochemistry, flow cytometry, gene rearrangement analysis, and assays for
key cytokines.
Vitreous Biopsy
Definitive diagnosis of primary vitreoretinal lymphoma
relies on vitreous biopsy. The decision to perform vitreous biopsy is often made after treatment for chronic
progressive uveitis has failed. An anterior chamber tap
for levels of interleukin (IL) 10 has been proposed to
screen patients for vitreous biopsy.16
However, this approach is controversial because the
performance profile of the laboratory study (eg, positive
and negative predictive probabilities, rates of sensitivity
and specificity) is unclear.16 The findings of the vitreous
samples of patients with primary vitreoretinal lymphoma often show evidence of apoptosis, cellular necrosis,
scavenging macrophages, and reactive inflammatory
cells.17 It is typically recommended that corticosteroids
be withheld prior to vitreous biopsy because they may
alter cellular morphology.18
Clinical Examination
Primary vitreoretinal lymphoma may present as a collection of nonclumped vitreous cells visible upon slit
lamp examination. The vitreous gel may appear hazy
due to dispersed vitreous cells. When primary vitreoretinal lymphoma affects the retina, it can present
with grey- to cream-colored retinal infiltrates (often
found along retinal arterioles), sub-retinal pigment
Cytology
Although cytology (or histology) remains the gold
standard of care, diagnostic sensitivity is not absolute
due to sampling phenomenon, problems with cellular preservation, and difficulties with interpretation.19
Frequently, specimens are obtained with too few cells
or cells that are poorly preserved. Under ideal circumstances, malignant B cells appear unambiguously ab-
April 2016, Vol. 23, No. 2
Ocular Imaging
Findings on autofluorescent imaging will vary according to the location of the lesions. If lymphoma cells accumulate under the RPE, then the RPE may have hyperfluoresecence. However, if the retinal deposits are
anterior to the RPE, then they appear to have hypofluorescence due to blocking.
Spectral-domain optical coherence tomography
can be used to demonstrate an intraretinal tumor,
which may extend through the full-thickness retina,
toward the RPE. Multiple, punctate, sub-RPE deposits
have also been described.15
Tumor deposits on fluorescein angiography will
typically have hypofluorescence on early and late
frames. However, leakage may be present from retinal
veins and staining of retinal arterioles.15
Cancer Control 111
normal with large, irregular nuclei, scanty cytoplasm,
and prominent nucleoli (Figs 1 and 2).4,6
Retinal, biopsy, choroidal biopsy, or both are less
commonly performed than vitreous biopsy because
this procedure has a higher risk of ocular and visual
morbidity.
Intraocular Cytokine Assay
B-cell primary vitreoretinal lymphoma typically presents with elevated IL-10 levels in the eye.16 Lymphoma
cells are thought to produce this cytokine, which acts
as a growth factor. This is in contrast to IL-6, which is
more commonly produced by inflammatory cells and
is characteristic of uveitis.
Vitreous IL-10 levels, in combination with the
IL-10:IL-6 ratio, have been used as a diagnostic test
for lymphoma, although they do not have a 100%
sensitivity rate. 20 Additional studies have verified
the high accuracy rate of cytokine profiling, but they
have also confirmed that negative testing does not
exclude the disease.19,21
Fig 1. — Finding on vitreous biopsy of primary vitreoretinal lymphoma.
The tissue was concentrated and sectioned from a cell block. The section stained with hematoxylin and eosin shows degenerating, moderately large lymphocytes. Many cells are necrotic. Nuclei vary in size and
shape, and their nuclear chromatin is hyperchromatic.
Photo courtesy of Curtis Margo, MD.
Immunohistochemistry
With samples of adequate size, immunohistochemistry
studies for cluster designation20 and κ and λ light chain
restriction can confirm B-cell monoclonality. However,
demonstration of surface light chain on lymphocytes
fixed in formalin can be technically challenging.
Flow Cytometry
Flow cytometry is well suited for the detection of
cell-surface markers if the sample is sufficiently cellular. The ratio of κ:λ light chains may be useful to confirm a monoclonal expansion of lymphocytes.
Polymerase Chain Reaction
Monoclonal gene rearrangements in IGH (B-cell
lymphoma) or TRG (T-cell lymphoma) are strongly inter-related with malignancy.21,22 A technique known as
microdissection can be employed to obtain abnormal
cells for analysis.21
Treatment
No currently agreed upon therapeutic strategy exists
for primary vitreoretinal lymphoma without CNS involvement (ie, isolated eye disease). Because no large
comparative clinical series, or randomized, masked,
clinical trials exist, the optimal treatment of primary
vitreoretinal lymphoma relies on clinical judgment.
Treatment regimens include local therapy, systemic
therapy, or a combination of both. However, it is unclear whether any of the available treatments of isolated ocular primary vitreoretinal lymphoma increase
rates of overall survival.23 Local therapy consists of
intravitreal chemotherapy and ocular radiotherapy.
Systemic therapy includes intravenous chemotherapy,
112 Cancer Control
Fig 2. — Magnified image of a histological specimen from an eye with
primary vitreoretinal lymphoma. The arrow denotes Bruch membrane,
anterior to which malignant cells have invaded the retina.
Photo courtesy of Curtis Margo, MD.
intrathecal chemotherapy, whole-brain radiotherapy,
and peripheral blood stem cell transplantation. Despite
available treatments, which may produce adequate initial responses, recurrence is common, often involving
the CNS.10 The goals of therapy include eradication of
local disease with prevention of local and CNS recurrences as well as minimizing adverse events.
Primary Central Nervous System Lymphoma
Many of the current treatment strategies for primary
vitreoretinal lymphoma are based on established treatments of primary CNS lymphoma due to their high coincidence and presumed shared pathogenesis. Several
of the current treatments for primary vitreoretinal lymApril 2016, Vol. 23, No. 2
phoma first demonstrated their effectiveness in the
treatment of primary CNS lymphoma.
Based on data from older studies in which the
option of solely supportive care was more common,
untreated primary CNS lymphoma was found to
have an extremely poor prognosis (average survival
rate < 3 months).12,24
The survival rate from primary CNS lymphoma
is significantly worse in patients with AIDS.12 Initial
success in the treatment of primary CNS lymphoma
has been achieved through whole brain irradiation,
but disease invariably recurs; the median survival rate
is approximately 12 months and the local recurrence
rate is nearly 90%.25 In 1 report, the 2-year survival
rate was 28%.25
Treatment of NHL metastatic to the brain with
high-dose methotrexate alone was introduced in
1977.26 The subsequent use of high-dose methotrexate alone in patients with primary CNS lymphoma produced a response rate between 38% and 74% and a median overall survival rate of about 2 years.27-29
Through the use of multiple chemotherapeutic
agents, including methotrexate, response rates for primary CNS lymphoma have increased to 71%, with a
median overall survival rate of 50 months.30
The success of these therapies in primary CNS
lymphoma led to their application in primary vitreoretinal lymphoma.
with primary CNS lymphoma or primary vitreoretinal
lymphoma found a 100% response rate when patients
were treated with intravenous methotrexate, vincristine, and thiotepa and intrathecal methotrexate and
arabinofuranosyl cytidine in 21-day cycles without radiation.34 However, recurrence rates were high: 66% of
cases recurred within 4.5 years.34
Experience with systemic chemotherapy and
ASCT is limited.35 One trial demonstrated longer
rates of progression-free survival and overall survival
in patients with relapsed or refractory primary CNS
lymphoma administered ASCT (in combination with
high-dose chemotherapy) compared with those patients who did not receive it.36 Ten of the 43 study patients (23%) had intraocular involvement.36
Several challenges have arisen due to the direct application of these systemic treatments for primary CNS
lymphoma to the management of primary vitreoretinal
lymphoma. One in particular is the blood–ocular barrier, which limits access of systemically administered
medication to the eye. Another is the vitreous: It is normally avascular and will limit chemical diffusion of
specific agents depending on their physical properties.
The vitreous is a prime tissue that harbors lymphoma.
When given systemically, high-dose methotrexate
may achieve tumoricidal levels in the anterior chamber
for up to 74 hours but at inadequate levels in the vitreous fluid.37,38
Systemic Chemotherapy
Systemic therapy for primary vitreoretinal lymphoma
without CNS involvement can include intravenous and
intrathecal chemotherapy and whole-brain radiotherapy. The preferred strategy varies by clinical center.2
Systemic chemotherapy regimens for primary
vitreoretinal lymphoma have included a single agent,
multiple agents, and chemotherapeutic agents plus
autologous stem cell transplantation (ASCT). Prior
single-agent treatment regimens have included
high-dose arabinofuranosyl cytidine, high-dose methotrexate, ifosfamide, and trofosfamide with varying
degrees of success.31-33 Prior studies with intravenous
high-dose methotrexate for the treatment of primary
CNS lymphoma with concurrent primary vitreoretinal
lymphoma achieved response rates that approached
100% in the brain, despite lower response rates of 78%
in the eye.29,32 One trial with either ifosfamide or trofosfamide found a 100% response rate with a mean progression-free survival rate of 18 months and a median
overall survival rate of 32 months.33
Comparing the efficacy of systemic chemotherapy
alone for primary vitreoretinal lymphoma is confounded by the fact that many earlier studies included initial
or simultaneous treatment with radiation; intravenous,
intrathecal, and intravitreal chemotherapy; ASCT; or
all therapies combined. One series of 14 study patients
Local Ocular Disease
Local therapy for primary vitreoretinal lymphoma has
included intravitreal chemotherapy and orbital radiotherapy. Often times, local treatment of ocular disease
is initiated because health care professionals may be
hesitant to treat a patient systemically unless evidence
of CNS disease is present.2
Ocular lymphoma cells are highly sensitive to radiation.39 However, this treatment has not significantly
increased the overall survival rate of these patients.40-42
Local response rates are generally high, but recurrence
is common. When the tumor reappears, additional radiation is considered hazardous due to the cumulative
effects of the radiation on the retina and optic nerve.
The complications of orbital radiation include dry eyes,
cataract, radiation retinopathy, and optic neuropathy.
Although cataracts are surgically treatable, radiation
injury to the retina and optic nerve is often permanent.
Radiation to the orbits usually involves 35 to 40 Gy delivered in 14 to 15 separate doses, including both eyes
in the treatment field.
Experience with intravitreal chemotherapy for primary vitreoretinal lymphoma has included methotrexate and rituximab. Intravitreal methotrexate for intraocular lymphoma is typically administered in a 400-µg
dose in 0.1 uL, every 2 weeks as induction therapy, followed by consolidation and maintenance therapy.5,43,44
April 2016, Vol. 23, No. 2
Cancer Control 113
When administered intravitreally, this dose maintains
therapeutic levels in the eye for at least 5 days.45 The
adverse events of intravitreal methotrexate include epithelial keratopathy, cataract, retinopathy, and hypotony (ie, low intraocular pressure). Treated patients may
also be at risk of tachyphylaxis with recurrent injections.6,43-46 Epithelial keratopathy, which has been reported in up to 58% of eyes,46 may be reduced by paracentesis before the injection.47
No double-masked, randomized clinical trials
exist for use of intravitreal methotrexate. The largest
reported series consists of 44 eyes.5 In that series, remission was achieved after a mean of 6.4 injections
(Table).5,43-46,48-51 The major shortcoming of intravitreal
therapy is that it fails to prevent CNS relapse or disease
in the contralateral eye.
Intravitreal rituximab has demonstrated efficacy
and has the ability to penetrate full-thickness retina.52
It is typically administered on a weekly basis as
1 mg in a 0.1-mL dose.48 Experience with the medication has revealed a high response rate (≤ 100%), but
it also has a high recurrence rate when treatment is
discontinued (≤ 55% within 3 months).49 Recurrences
typically occur within 3 months following treatment
discontinuation.49 Additional courses of rituximab
may cause disease remission; however, the effectiveness of the drug may diminish.49 Grossly visible retinal lesions may resolve with rituximab. Rituximab
has been reported to cause clinical remission in some
patients unresponsive to methotrexate.49 Adverse
events of rituximab include vitritis, elevated intraocular pressure (60%), and iridocyclitis, with mutton-fat
keratic precipitates (35%).49 Despite resolution of ocular lesions with rituximab, 69% of patients in a single
trial developed CNS lymphoma.49
Rituximab has a half-life of about 5 days in the
eye and exerts tumoricidal effects for 3 to 4 weeks.53,54
Combination intravitreal methotrexate/rituximab has
also been used with some benefit and may potentially
decrease the adverse events seen with single-agent
therapy.48
A collaborative study of patients with primary vitreoretinal lymphoma without clinical or radiographic
evidence of CNS involvement from 16 centers (including 9 study patients [11%] with lymphoma in the spinal fluid) found that local therapy did not increase the
risk of brain relapse compared with systemic therapy.10
This retrospective series of 83 immunocompetent
study patients with primary vitreoretinal lymphoma
from 7 countries evaluated 16 different treatment regimens (3 local and 16 extensive).10 The median progression-free survival rate was 29.6 months and the overall median survival rate was 58 months; this rate was
not impacted by the therapeutic regimen.10 Relapse
rates were high (56%); the median time to relapse was
19 months.10 The site of relapse (eg, eyes, brain, other
sites) was also not affected by the treatment regimen,
with most (92%) patients experiencing relapse in the
brain or eyes.10
Another collaborative review lasting 20 years and
covering 78 study patients with primary vitreoretinal
lymphoma without signs of primary CNS lymphoma
from 17 centers found that use of systemic chemotherapy was not associated with a reduced rate of CNS lym-
Table. — Local Therapy With Intravitreal Chemotherapy
Study
Intravitreal
Treatment
No. of Cases
of Primary
Vitreoretinal
Lymphoma
No. of Cases
of Primary
Lymphoma
of the CNS
No. of
Eyes
de Smet45
Methotrexate/thiotepa
1
1
Fishburne 43
Methotrexate
4
2
Frenkel5
Methotrexate
26
Hashida49
Rituximab
Itty48
Kitzmann 50
Ohguro 51
Sen
44
Smith46
Ocular
Response,
% (n)
No. of
Injections
Until
Remission
No. of Eye
Relapses
1
100 (1)
6 (3rd week)
0
18
7
100 (7)
6.7 (mean)
0
13.1 (mean)
13
44
100 (40)
with all data
6.4 (mean)
0
21 (median)
13
0
20
100 (20)
4 (mean)
11/20 (55%)
24.7 (mean)
Rituximab
1
0
2
100 (2)
6 (mean)
2/2 (100%)
11
Rituximab
3
3
5
100 (5)
2 (mean)
0
3.6 (median)
Rituximab
2
1
3
100 (3)
1.67 (mean)
0
2 (mean)
Methotrexate
1
1
1
100 (1),
then
tachyphylaxis
3, followed by
tachyphylaxis
1 (100%)
48
Methotrexate
16
16
26
100 (22)
100 (4) after
pars plana
vitrectomy
8.5 (median)
6/26 (23%)
18.5 (median)
Follow-Up,
mo
CNS = central nervous system.
114 Cancer Control
April 2016, Vol. 23, No. 2
phoma, but it was associated with more severe adverse
events compared with those seen with local treatment
(ocular radiotherapy, ocular chemotherapy, or both).55
Combination Therapy
Combination therapy consists of local and systemic
therapy. Some institutions recommend systemic treatment with high-dose methotrexate plus adjuvant local
radiotherapy,56,57 whereas other centers prefer intravitreal chemotherapy with methotrexate or rituximab
rather than radiotherapy.5,48
One retrospective series of 221 immunocompetent
patients with primary CNS lymphoma and primary vitreoretinal lymphoma found that those who received
dedicated ocular therapy in combination with systemic
treatment had increased progression-free survival rates
compared with those not receiving dedicated ocular
therapy.58 However, no difference was seen in overall
survival rates, and the study did not contain a control
group and involved a variety of treatments.58 Those
who did not receive dedicated ocular therapy were not
at increased risk of developing ocular recurrence.58
Recommendations
No universally agreed-upon therapeutic strategy exists for primary vitreoretinal lymphoma without CNS
involvement, although a common (but not standard)
approach involves systemic therapy for patients with
CNS disease and local therapy for those with primary
vitreoretinal lymphoma confined to the eye.
Recommendations from the International Primary Central Nervous System Lymphoma Collaborative Group and the British Neuro-Oncology Society
differ.6,18 For the treatment of ocular-only primary vitreoretinal lymphoma, the Primary Central Nervous
System Lymphoma Collaborative Group recommends
local therapy for unilateral disease and local therapy,
systemic chemotherapy, or both for those with bilateral
disease.6 Orbital radiotherapy may be the preferred local therapy in patients with bilateral disease, whereas
intravitreal chemotherapy with or without orbital radiotherapy is preferred for unilateral disease.6 In patients with a prior history of radiotherapy, intravitreal
chemotherapy is recommended.6
By contrast, the British Neuro-Oncology Society
recommends systemic chemotherapy with high-dose
methotrexate and ocular radiotherapy to both globes
for isolated primary vitreoretinal lymphoma.18 The
British Neuro-Oncology Society recognized that intravitreal methotrexate was an effective treatment option
for isolated recurrences.18
Conclusions
The search for the optimal treatment for primary vitreoretinal lymphoma without central nervous system
involvement presents a daunting challenge. At the crux
April 2016, Vol. 23, No. 2
of the dilemma is whether local treatment will be effective given the high risk of central nervous system disease and whether early systemic treatment has the potential to improve survival. The differential treatment
response of primary vitreoretinal lymphoma may be
related to different molecular genetic subtypes of the
malignancy, which is a hypothesis that requires further
study.3 Lack of a superior treatment strategy despite a
decade of collective experience suggests that any major impact on reducing relapse rates in central nervous
system and improving outcomes awaits future innovative therapeutic approaches.
References
1. Mochizuki M, Singh AD. Epidemiology and clinical features of intraocular lymphoma. Ocul Immunol Inflamm. 2009;17(2):69-72.
2. Davis JL. Intraocular lymphoma: a clinical perspective. Eye (Lond).
2013;27(2):153-162.
3.Coupland SE. Molecular pathology of lymphoma. Eye (Lond).
2013;27(2):180-189.
4. Coupland SE, Damato B. Understanding intraocular lymphomas.
Clin Experiment Ophthalmol. 2008;36(6):564-578.
5. Frenkel S, Hendler K, Siegal T, et al. Intravitreal methotrexate for
treating vitreoretinal lymphoma: 10 years of experience. Br J Ophthalmol.
2008;92(3):383-388.
6. Chan CC, Rubenstein JL, Coupland SE, et al. Primary vitreoretinal lymphoma: a report from an International Primary Central Nervous System Lymphoma Collaborative Group symposium. Oncologist. 2011;16(11):1589-1599.
7. Falkenhagen KM, Braziel RM, Fraunfelder FW, et al. B-Cells in ocular adnexal lymphoproliferative lesions express B-cell attracting chemokine 1 (CXCL13). Am J Ophthalmol. 2005;140(2):335-337.
8. McCann KJ, Ashton-Key M, Smith K, et al. Primary central nervous
system lymphoma: tumor-related clones exist in the blood and bone marrow
with evidence for separate development. Blood. 2009;113(19):4677-4680.
9. Coupland SE, Loddenkemper C, Smith JR, et al. Expression of immunoglobulin transcription factors in primary intraocular lymphoma and
primary central nervous system lymphoma. Invest Ophthalmol Vis Sci.
2005;46(11):3957-3964.
10. Grimm SA, Pulido JS, Jahnke K, et al. Primary intraocular lymphoma: an International Primary Central Nervous System Lymphoma Collaborative Group Report. Ann Oncol. 2007;18(11):1851-1855.
11. Behin A, Hoang-Xuan K, Carpentier AF, et al. Primary brain tumours
in adults. Lancet. 2003;361(9354):323-331.
12. Coté TR, Manns A, Hardy CR, et al; AIDS/Cancer Study Group.
Epidemiology of brain lymphoma among people with or without acquired
immunodeficiency syndrome. J Natl Cancer Inst. 1996;88(10):675-679.
13. Hoffman PM, McKelvie P, Hall AJ, et al. Intraocular lymphoma: a
series of 14 patients with clinicopathological features and treatment outcomes. Eye (Lond). 2003;17(4):513-521.
14. Jahnke K, Korfel A, Komm J, et al. Intraocular lymphoma 2000–
2005: results of a retrospective multicentre trial. Graefes Arch Clin Exp
Ophthalmol. 2006;244(6):663-669.
15. Fardeau C, Lee CP, Merle-Béral H, et al. Retinal fluorescein, indocyanine green angiography, and optic coherence tomography in non-Hodgkin primary intraocular lymphoma. Am J Ophthalmol. 2009;147(5):886894, 894.e881.
16. Cassoux N, Giron A, Bodaghi B, et al. IL-10 measurement in aqueous humor for screening patients with suspicion of primary intraocular lymphoma. Invest Ophthalmol Vis Sci. 2007;48(7):3253-3259.
17. Coupland SE. Vitreous biopsy: specimen preparation and interpretation. Monogr Clin Cytol. 2012;21:61-71.
18. British Neuro-Oncology Society; National Cancer Action Team.
Rare Brain and CNS Tumours Guidelines: Guidelines on the Diagnosis
and Management of Adult Pineal Area Tumours. Published June 2011.
http://www.bnos.org.uk/wp-content/uploads/2015/08/Guidelines-on-the
-Diagnosis-and-Management.pdf. Accessed April 6, 2016.
19. Kimura K, Usui Y, Goto H; Group JILS. Clinical features and diagnostic significance of the intraocular fluid of 217 patients with intraocular
lymphoma. Jpn J Ophthalmol. 2012;56(4):383-389.
20. Sugita S, Takase H, Sugamoto Y, et al. Diagnosis of intraocular
lymphoma by polymerase chain reaction analysis and cytokine profiling of
the vitreous fluid. Jpn J Ophthalmol. 2009;53(3):209-214.
21. Wang Y, Shen D, Wang VM, et al. Molecular biomarkers for the diagnosis of primary vitreoretinal lymphoma. Int J Mol Sci. 2011;12(9):5684-5697.
22. Shen DF, Zhuang Z, LeHoang P, et al. Utility of microdissection and
polymerase chain reaction for the detection of immunoglobulin gene rearCancer Control 115
rangement and translocation in primary intraocular lymphoma. Ophthalmology.
1998;105(9):1664-1669.
23. Hormigo A, Abrey L, Heinemann MH, et al. Ocular presentation of
primary central nervous system lymphoma: diagnosis and treatment. Br J
Haematol. 2004;126(2):202-208.
24. Jellinger K, Radaskiewicz TH, Slowik F. Primary malignant lymphomas of the central nervous system in man. Acta Neuropathol Suppl.
1975;(suppl 6):95-102.
25. Nelson DF, Martz KL, Bonner H, et al. Non-Hodgkin’s lymphoma of
the brain: can high dose, large volume radiation therapy improve survival?
Report on a prospective trial by the Radiation Therapy Oncology Group
(RTOG): RTOG 8315. Int J Radiat Oncol Biol Phys. 1992;23(1):9-17.
26. Skarin AT, Zuckerman KS, Pitman SW, et al. High-dose methotrexate with folinic acid in the treatment of advanced non-Hodgkin lymphoma
including CNS involvement. Blood. 1977;50(6):1039-1047.
27. Herrlinger U, Schabet M, Brugger W, et al. German Cancer Society
Neuro-Oncology Working Group NOA-03 multicenter trial of single-agent
high-dose methotrexate for primary central nervous system lymphoma.
Ann Neurol. 2002;51(2):247-252.
28. Herrlinger U, Küker W, Uhl M, et al. NOA-03 trial of high-dose methotrexate in primary central nervous system lymphoma: final report. Ann
Neurol. 2005;57(6):843-847.
29. Batchelor T, Carson K, O’Neill A, et al. Treatment of primary CNS
lymphoma with methotrexate and deferred radiotherapy: a report of
NABTT 96-07. J Clin Oncol. 2003;21(6):1044-1049.
30. Pels H, Schmidt-Wolf IG, Glasmacher A, et al. Primary central nervous system lymphoma: results of a pilot and phase II study of systemic
and intraventricular chemotherapy with deferred radiotherapy. J Clin Oncol.
2003;21(24):4489-4495.
31. Baumann MA, Ritch PS, Hande KR, et al. Treatment of intraocular
lymphoma with high-dose Ara-C. Cancer. 1986;57(7):1273-1275.
32. Batchelor TT, Kolak G, Ciordia R, et al. High-dose methotrexate for
intraocular lymphoma. Clin Cancer Res. 2003;9(2):711-715.
33. Jahnke K, Thiel E, Bechrakis NE, et al. Ifosfamide or trofosfamide
in patients with intraocular lymphoma. J Neurooncol. 2009;93(2):213-217.
34. Sandor V, Stark-Vancs V, Pearson D, et al. Phase II trial of chemotherapy alone for primary CNS and intraocular lymphoma. J Clin Oncol.
1998;16(9):3000-3006.
35. Soussain C, Merle-Béral H, Reux I, et al. A single-center study of
11 patients with intraocular lymphoma treated with conventional chemotherapy followed by high-dose chemotherapy and autologous bone marrow transplantation in 5 cases. Leuk Lymphoma. 1996;23(3-4):339-345.
36. Soussain C, Hoang-Xuan K, Taillandier L, et al. Intensive chemotherapy followed by hematopoietic stem-cell rescue for refractory
and recurrent primary CNS and intraocular lymphoma: Société Française de Greffe de Moëlle Osseuse-Thérapie Cellulaire. J Clin Oncol.
2008;26(15):2512-2518.
37. de Smet MD, Stark-Vancs V, Kohler DR, et al. Intraocular levels
of methotrexate after intravenous administration. Am J Ophthalmol.
1996;121(4):442-444.
38. Henson JW, Yang J, Batchelor T. Intraocular methotrexate level after high-dose intravenous infusion. J Clin Oncol. 1999;17(4):1329.
39. Margolis L, Fraser R, Lichter A, et al. The role of radiation therapy in the
management of ocular reticulum cell sarcoma. Cancer. 1980;45(4):688-692.
40. Fine HA, Mayer RJ. Primary central nervous system lymphoma.
Ann Intern Med. 1993;119(11):1093-1104.
41. Ferreri AJ, Blay JY, Reni M, et al. Relevance of intraocular involvement in the management of primary central nervous system lymphomas.
Ann Oncol. 2002;13(4):531-538.
42. Schlegel U, Schmidt-Wolf IG, Deckert M. Primary CNS lymphoma:
clinical presentation, pathological classification, molecular pathogenesis
and treatment. J Neurol Sci. 2000;181(1-2):1-12.
43. Fishburne BC, Wilson DJ, Rosenbaum JT, et al. Intravitreal methotrexate as an adjunctive treatment of intraocular lymphoma. Arch Ophthalmol.
1997;115(9):1152-1156.
44. Sen HN, Chan CC, Byrnes G, et al. Intravitreal methotrexate resistance in a patient with primary intraocular lymphoma. Ocul Immunol
Inflamm. 2008;16(1):29-33.
45. de Smet MD, Vancs VS, Kohler D, et al. Intravitreal chemotherapy
for the treatment of recurrent intraocular lymphoma. Br J Ophthalmol.
1999;83(4):448-451.
46. Smith JR, Rosenbaum JT, Wilson DJ, et al. Role of intravitreal
methotrexate in the management of primary central nervous system lymphoma with ocular involvement. Ophthalmology. 2002;109(9):1709-1716.
47. Hardwig PW, Pulido JS, Bakri SJ. The safety of intraocular methotrexate in silicone-filled eyes. Retina. 2008;28(8):1082-1086.
48.Itty S, Pulido JS. Rituximab for intraocular lymphoma. Retina.
2009;29(2):129-132.
49. Hashida N, Ohguro N, Nishida K. Efficacy and complications of intravitreal rituximab injection for treating primary vitreoretinal lymphoma.
Transl Vis Sci Technol. 2012;1(3):1.
50. Kitzmann AS, Pulido JS, Mohney BG, et al. Intraocular use of rituximab. Eye (Lond). 2007;21(12):1524-1527.
51. Ohguro N, Hashida N, Tano Y. Effect of intravitreous rituximab in116 Cancer Control
jections in patients with recurrent ocular lesions associated with central
nervous system lymphoma. Arch Ophthalmol. 2008;126(7):1002-1003.
52. Pulido JS, Bakri SJ, Valyi-Nagy T, et al. Rituximab penetrates fullthickness retina in contrast to tissue plasminogen activator control. Retina.
2007;27(8):1071-1073.
53. Kim H, Csaky KG, Chan CC, et al. The pharmacokinetics of rituximab following an intravitreal injection. Exp Eye Res. 2006;82(5) 760-766.
54. Chu LC, Eberhart CG, Grossman SA, et al. Epigenetic silencing of multiple genes in primary CNS lymphoma. Int J Cancer. 2006;119(10):2487-2491.
55. Riemens A, Bromberg J, Touitou V, et al. Treatment strategies in
primary vitreoretinal lymphoma: a 17-center European collaborative study.
JAMA Ophthalmol. 2015;133(2):191-197.
56. Berenbom A, Davila RM, Lin HS, et l. Treatment outcomes for primary intraocular lymphoma: implications for external beam radiotherapy.
Eye (Lond). 2007;21(9):1198-1201.
57. Stefanovic A, Davis J, Murray T, et al. Treatment of isolated primary
intraocular lymphoma with high-dose methotrexate-based chemotherapy and
binocular radiation therapy: a single-institution experience. Br J Haematol.
2010;151(1):103-106.
58. Grimm SA, McCannel CA, Omuro AM, et al. Primary CNS lymphoma with intraocular involvement: International PCNSL Collaborative Group
Report. Neurology. 2008;71(17):1355-13601.
April 2016, Vol. 23, No. 2
Many patients with conjunctival melanoma
experience suboptimal outcomes because
of iatrogenic tumor seeding, inadequate
local tumor control, and surgical morbidity.
Photo courtesy of Lynn E. Harman, MD. Chicago River.
Management of Primary Acquired Melanosis, Nevus, and
Conjunctival Melanoma
Andrew Kao, MD, Armin Afshar, MD, Michele Bloomer, MD, and Bertil Damato, MD, PhD
Background: The management of conjunctival melanoma is difficult because of the rarity of the disease,
confusing terminology, high rates of local tumor recurrence, controversies regarding treatment, a poor evidence base, unreliable prognostication, and significant mortality rates.
Methods: The medical literature was reviewed, focusing on treatment and management options for conjunctival
melanoma. Recent trends and developments were summarized with respect to terminology, local treatment,
histology, genetic analysis, prognostication, and systemic treatment, highlighting the scope for research and
possible improvements in patient care.
Results: Histopathological diagnostic terminology for primary acquired melanosis is being superseded by
more explicit terminology, thus differentiating hypermelanosis from conjunctival melanocytic intraepithelial
neoplasia. Topical chemotherapy and increased use of adjunctive radiotherapy have helped improve rates of
local tumor control. Use of exenteration has become rare. Regional and systemic metastases are common in
patients with nonbulbar conjunctival melanoma, although long-term survivors with metastases are growing
in number. Prognostication is mainly based on tumor size and location, but histological and genetic data
into multivariate analyses will soon be incorporated. The role of sentinel lymph-node biopsy continues to be
controversial. Chemotherapy for metastatic disease is being superseded by targeted therapy based on genetic
abnormalities such as BRAF mutations.
Conclusions: The management of conjunctival melanoma requires expert care from an experienced, multidisciplinary team. The goal of therapy is to provide good local tumor control with minimal morbidity,
high-quality pathology, and adequate psychological support. Maximizing patient enrollment in multicenter
clinical trials is likely to strengthen evidence-based decision-making.
Introduction
Conjunctival melanoma is a rare but potentially
From the Department of Ophthalmology, University of California,
San Francisco, California.
Address correspondence to Bertil Damato, MD, PhD, Department of Ophthalmology, 8 Koret Way, San Francisco, CA 94143.
E-mail: [email protected]
Submitted August 15, 2015; accepted November 12, 2015.
No significant relationships exist between the authors and the
companies/organizations whose products or services may be referenced in this article.
April 2016, Vol. 23, No. 2
sight- and life-threatening malignancy of the eye.1,2
It arises from melanocytes of the basal layer of the
conjunctival epithelium and comprises approximately 2% to 7% of ocular melanomas.1-3 The incidence is
approximately 0.24 to 0.8 cases per 1 million and is
increasing.1,2 Risk factors for conjunctival melanoma
are not well understood because of the rarity of the
disease and lack of large population-based studies.1,2
The disease is most common in whites and rare in
Asian/Pacific Islanders.2 Most studies have not shown
any sex predilection.2 Conjunctival melanoma has a
Cancer Control 117
strong association with primary acquired melanosis
(PAM) and conjunctival nevi. Conjunctival melanoma tends to locally recur, seed to distant parts of the
conjunctiva, and systemically metastasize to regional
lymph nodes. Once metastatic disease has occurred,
outcomes are often fatal.
Classification of Conjunctival Pigmented
Lesions
Melanocytic lesions of the conjunctiva are generally
classified as PAM, nevus, and melanoma (Fig 1).
The term PAM tends to be both clinically and
histologically used. Although this term is adequate
for clinical findings, it is not ideal for histological
diagnosis because it encompasses a wide range of
conditions, from benign to malignant, that include
complexion melanosis and PAM with and without
atypia. Furthermore, PAM with atypia tends to be
categorized as mild, moderate, or severe, and is often
inconsistently used. To improve the rate of precision
in the reporting of histological findings, Damato and
Coupland4 recommend distinguishing between hypermelanosis and conjunctival melanocytic intraepithelial neoplasia. In their classification, hypermelanosis refers to the overproduction of melanin from
a normal population of melanocytes, as occurs with
complexion/racial melanosis.4 To make classification
more consistent, Damato and Coupland4 have created a system for scoring the grade of malignancy
from 0 to 10 according to pattern of melanocytic pro-
liferation, extent of vertical spread, and degree of cellular atypia (Table).4
Conjunctival melanocytic intraepithelial neoplasia with a score of 0 would correspond with PAM
without atypia and would indicate an overpopulation of melanocytes showing no cellular atypia and
confined to the basal layer of the epithelium. Higher scores correspond to PAM with atypia, indicating
cellular atypia of melanocytes with invasion into the
more superficial layers of the epithelium but without
breaching the basement membrane of the epithelium. No consensus exists as to how severe PAM/conjunctival melanocytic intraepithelial neoplasia with
atypia should be classified prior to being labeled as
melanoma in situ. Damato and Coupland4 suggest
designating any atypia with a score exceeding 5 as in
situ, which corresponds to confluent proliferation of
atypical melanocytes involving more than 50% of the
thickness of the epithelium. This designation might
improve communication between the pathologist
and clinician, resulting in fewer delays of treatment
due to vagaries in classification. Multicenter studies
are needed to determine whether this 10-point scoring system improves objectivity and clinical care compared with simply categorizing disease as mild, moderate, and severe.
Conventionally, conjunctival melanoma implies
invasion of the substantia propria, even if the word
invasive is omitted. We believe that it would be safe
practice to distinguish between melanoma in situ and
A
B
C
D
E
F
Fig 1A–F. — Clinical features of pigmented conjunctival lesions. (A) Complexion melanosis appearing as diffuse conjunctival pigmentation in a
pigmented individual. (B) Primary acquired melanosis manifesting as a discrete patch of conjunctival pigment. (C) Nevus shown as a slightly elevated pigmented lesion with intraepithelial cysts. (D) Invasive melanoma demonstrated as a pigmented nodule arising from a patch of conjunctival
melanosis. (E) Invasive melanoma of the palpebral conjunctiva. (F) Invasive melanoma with locally disseminated disease, due to prior biopsy at an
outside institution, without use of adjuvant therapy.
118 Cancer Control
April 2016, Vol. 23, No. 2
Table. — Coupland–Damato Scoring System for Conjunctival
Melanocytic Intraepithelial Neoplasia
Feature
Melanocytic
pattern
Vertical spread
of melanocytes
with atypia
Cellular atypia
Total
Description
Score
No melanocytic neoplasia (± hypermelanosis)
0
Basal/lentiginous proliferation or
1
Pagetoid spread (± basal/lentiginous) or
2
Nonconfluent nests (± basal/lentiginous
pagetoid) or
3
Confluent nests (± basal/lentiginous
pagetoid)
4
Basal layer of epithelium
0
< 50% epithelial thickness
1
50%–90% epithelial thickness
2
Total replacement of epithelium
3
Nuclei < basal squamous cells
0
Nuclei ≥ basal squamous cells
1
Cytoplasm < basal squamous cells
0
Cytoplasm ≥ basal squamous cells
1
Nucleolus and mitoses absent
0
Nucleolus ± mitoses present
1
(Must be ≤ 10 points)
Length of specimen invaded by most
severe disease, %
Changes extend to or near lateral
resection edge(s)
Yes/No
Invasive melanoma (if yes, report
elsewhere)
Yes/No
From Damato B, Coupland SE. Conjunctival melanoma and melanosis:
a reappraisal of terminology, classification and staging. Clin Exp Ophthalmol.
2008;36(8):786-795.
ed, or amelanotic; unifocal or multifocal; and with or
without adjacent melanosis.2,6 Amelanotic melanomas
can be white, yellow, pink, or red in color. Feeder vessels are usually present. Hemorrhage is rare. The tumor surface does not keratinize. Most tumors are located in the bulbar conjunctiva, usually involving the
limbus.2 Less commonly, tumors may present on the
palpebral or fornical conjunctiva, plica semilunaris,
or caruncle.2 Regional lymph nodes may be enlarged
if regional metastasis has occurred.7
Differential Diagnosis
Primary acquired melanosis requires incisional biopsy to distinguish diffuse invasive melanoma and
melanoma in situ from hypermelanosis and conjunctival melanocytic intraepithelial neoplasia without
atypia (ie, PAM without atypia). Congenital ocular
melanocytosis is slate-grey in color rather than brown
and involves the sclera alone; the overlying conjunctiva is transparent. Conjunctival nevi tend to have multiple cysts and are not associated with feeder vessels,
except when occurring in children. Squamous cell
carcinoma is usually amelanotic but can be deeply
pigmented, especially in dark-skinned individuals. It
usually presents as frosty keratinization, which does
not occur with melanoma. Typically, lymphoma has a
salmon-pink color, is located in the plica and fornices,
and is bilateral. Rare mimicking lesions can include
pinguecula, pterygium, pyogenic granuloma, deposition of pigmented foreign material, extraocular spread
of uveal melanoma, metastasis from cutaneous melanoma, staphyloma, subconjunctival hematoma, foreign
body, and hematic cyst.8
Clinical Examination
invasive melanoma when discussing pathology. In the
presence of invasive melanoma, it can be difficult or
impossible to histologically distinguish between primary conjunctival melanocytic intraepithelial neoplasia that caused invasive melanoma and pagetoid spread
from the invasive tumor.
Damato and Coupland4 suggest that PAM is limited
to clinical findings and that histological reports should
distinguish between hypermelanosis and melanocytic
intraepithelial neoplasia, numerically scoring the degree of malignancy instead of using vague terms such
as mild, moderate, and severe.
Clinical Features
Melanoma in situ presents as 1 or more brown patches
developing anywhere in the conjunctiva, almost always unilaterally, and becoming more extensive over
time.5 Melanocytic proliferation can become amelanotic, especially after unsuccessful treatment.
Invasive conjunctival melanoma can be nodular,
diffuse, or mixed; deeply pigmented, lightly pigmentApril 2016, Vol. 23, No. 2
Examination of the entire conjunctiva must include the
superior fornix and superior palpebral regions, either
after double eversion of the upper eyelid with a Desmarres retractor or by pinching and lifting the upper
lid away from the bulbar conjunctiva while performing
inspection with a binocular indirect ophthalmoscope
and a 20-D lens. The preauricular, postauricular parotid, submandibular, and cervical lymph nodes are routinely palpated.7
Imaging
Color drawings are valuable for defining tumor size
and extent; Fig 2 can be used as a template for this
purpose. In addition, if performed correctly, color
photography with adequate retraction of the eyelids
is useful for documenting surface features and tumor margins.
Anterior segment ocular coherence tomography
uses light waves to obtain cross-sectional images of
various eye structures, with an axial resolution of up to
5 μm.9,10 PAM/conjunctival melanocytic intraepithelial
Cancer Control 119
neoplasia without atypia appears as normal-thickness
epithelium, with strong hyper-reflectivity of the basal
epithelium, correlating with the basal layer of melanocytes seen on histopathology.9,10 Nevi are hyperreflective, well circumscribed, and often have visible
cysts, with appearances varying according to whether
the lesion is junctional, compound, or subepithelial.10
Invasive melanoma reveals subepithelial disease that
casts a significant shadow (Fig 3).10 Ultrasonographic
Fig 2. — Template for drawing conjunctival tumors that shows the entire
conjunctival surface divided into regions (cornea, limbus, bulbar conjunctiva, fornix, plica, caruncle, palpebral conjunctiva, eyelid skin) and sectors.
A
B
biomicroscopy using transducers between 50 and
100 MHz can measure the thickness of conjunctival
melanomas with reasonable accuracy and may identify tumors greater than 2 mm in thickness so that
sentinel lymph-node biopsy can be planned before
tumor excision.11 High-frequency ultrasonography
can be useful for detecting or excluding suspected
underlying uveal melanoma.
Diagnosis
Histology
Histology by a trained pathologist is required to differentiate between hypermelanosis, conjunctival melanocytic intraepithelial neoplasia, in situ melanoma,
invasive melanoma, and other pathology. Bleaching of
specimens is often required to examine cell morphology. Features of atypical melanocytes include nuclear
pleomorphism, prominent nucleoli, increased eosinophilic cytoplasm, and mitotic figures.
Hypermelanosis is characterized by increased
melanin granules within normal melanocytes in the
basal epithelium. Conjunctival melanocytic intraepithelial neoplasia with a score of 0 consists of an increased number of melanocytes without atypical
features (Fig 4A). Conjunctival melanocytic intraepithelial neoplasia with higher scores is identified by intraepithelial growth of atypical melanocytes, extending up to full thickness, but with no invasion through
to the epithelial basement membrane (Fig 4B). Invasive melanoma is defined by atypical melanocytes
that have invaded the basement membrane into the
substantia propria (Fig 4C).
Histological assessment has been improved due
to the development of immunohistochemistry. Markers for melanocytes include S100, human melanoma
black 45, SRY-box containing gene 10, and Melan-A. A
red chromogen is useful in the presence of a heavily
pigmented lesion.
C
Fig 3A–C. — Ocular coherence tomographical features of conjunctival melanoma. (A) Slit lamp photograph of a raised, reddish, nonpigmented conjunctival lesion suspected to be squamous neoplasia. (B) Ultra high-resolution optical coherence tomography in the area of the black dotted arrow
disclosed a subepithelial lesion (a) beneath a normal thickness epithelium (b). The lesion was casting a significant shadow (c) on the underlying tissue.
Foci of epithelial cleavage (d) and moderate hyper-reflectivity of the epithelium were noted. (C) Histopathological examination confirmed the diagnosis
of melanoma (hematoxylin and eosin stain, ×200).
Reprinted from Shousha MA, Karp CL, Canto AP, et al. Diagnosis of ocular surface lesions using ultra-high-resolution optical coherence tomography.
Ophthalmology. 2013;120(5):883-891. © May 2013, with permission from Elsevier.
120 Cancer Control
April 2016, Vol. 23, No. 2
A
B
C
Fig 4A–C. — Histology of pigmented conjunctival lesions (hematoxylin and eosin stain, ×400). (A) Conjunctival melanocytic intraepithelial neoplasia
without atypia with an increased number of melanocytes within the basal epithelium without atypical features. (B) Melanoma in situ with atypical melanocytes and invasion of superficial layers of epithelium, but without progression through the epithelial basement membrane. (C) Invasive melanoma
with highly atypical melanocytes that fill the epithelium, invading the underlying substantia propria.
Biopsy
Incisional biopsy of nodular melanoma can cause tumor seeding, so it should be performed for PAM alone;
otherwise, excisional biopsy is preferred when possible. The specimen should be placed on a stiff card
or filter paper to prevent scrolling so that oblique histological sections are avoided. Care must be taken to
avoid crush artifact. Testing for BRAF mutations should
be considered at this stage if concern exists for metastatic disease potentially treatable with v-raf murine
sarcoma viral oncogene homolog B1 (BRAF) inhibitors. When a patient is referred to an oncology center
after biopsy, we routinely arrange for our ophthalmic
pathologist to review the histology results.
geons not following the “no touch” technique or who
may not understand the importance of using fresh instruments for closure to avoid seeding.12 After witnessing recurrences in distant parts of the conjunctiva in
such cases, Damato and Coupland12 routinely administer adjunctive topical chemotherapy to all such patients
irrespective of histological findings. Although data are
insufficient for statistical evaluation of this approach,
clinical impressions have been positive.2
Treatment
Adjuvant Therapy
Cryotherapy: Historically, cryotherapy was the only
treatment for PAM/conjunctival melanocytic intraepithelial neoplasia with atypia; in modern times, its use
is advocated by some as adjunctive therapy following local excision. It is delivered using the double or
triple freeze-thaw technique.2 Jakobiec et al13 recommend using a liquid nitrogen probe and a subconjunctival thermocouple to ensure that a temperature
of –20 °C is achieved; however, Damato and Coupland14 reported an unacceptable tumor recurrence
rate in addition to complications such as uveitis, hypotony, macular edema, and squamous metaplasia
of the conjunctiva. This treatment has largely been
superseded by topical chemotherapy. We now perform cryotherapy for residual or recurrent intraepithelial disease alone after at least 2 courses of topical
chemotherapy.
Topical Chemotherapy: This type of treatment is
administered using mitomycin C (MMC), because other
agents such as fluorouracil are less effective. The standard protocol is to prescribe drops 4 times per day for
2 weeks, repeating the treatment after a 2-week rest
period. We prefer a 1-week course of topical chemotherapy, repeated every 4 weeks for 4 cycles, because
doing so causes less pain and inflammation as well
Surgical Excision
The conventional approach to treatment is to excise
nodular conjunctival melanomas with wide safety margins and lamellar scleral dissection, to ensure deep
and lateral clearance, and to administer adjunctive
cryotherapy as a precaution.1 Such extensive surgery
often requires amniotic membrane grafting, which can
result in significant morbidity (eg, corneal scarring,
scleral translucency). Furthermore, significant rates of
local tumor recurrence have been reported, sometimes
exceeding 30%.1,7,12 Growing circumstantial evidence
suggests that such local recurrence is associated with
higher rates of mortality.12
For at least 15 years, the approach was to excise
the tumor with minimal surgical clearance, perform
no lamellar scleral excision, and provide no adjunctive cryotherapy, instead administering adjunctive
radiotherapy for all cases with invasive melanoma,
irrespective of histological evidence of clearance.8
In addition, adjunctive topical chemotherapy is warranted if adjacent intraepithelial neoplasia is present,
whether this consists of primary disease or secondary
pagetoid spread.8
Many patients are referred to an oncology center
after undergoing incisional biopsy or excision by surApril 2016, Vol. 23, No. 2
Primary Exenteration
Primary exenteration has been abandoned as a primary treatment for PAM.5 Such extensive surgery is now
performed for advanced invasive melanoma alone.
Cancer Control 121
as improved patient compliance. Topical steroids are
helpful in reducing the inflammation, and our preference is to use 0.1% fluorometholone because it does
not penetrate the globe. No consensus exists regarding
the ideal dose of MMC. We prescribe 0.03% MMC because we found that lower concentrations were associated with local treatment failure, whereas higher doses
caused limbal stem cell failure in select patients. Some
authors advocate punctual occlusion during topical
chemotherapy2; however, we do not favor this practice,
especially with patients who have tumor recurrence
in the nasolacrimal system. When administering topical chemotherapy, the patient must be provided with
disposable gloves, petroleum jelly to protect the eyelid
skin, a disposal container (to be returned to the hospital with all soiled materials), and detailed oral and written instructions on the storage and administration of
the MMC drops.
Radiotherapy: Radiotherapy can consist of
brachytherapy, which uses isotopes such as strontium,
ruthenium, or iodine, or teletherapy, which uses photons or a proton beam (Fig 5). Adjunctive radiotherapy
after local excision was reported by Collins15 in 1918
and later by Lederman16 in 1958 but it has since fell into
disuse. Damato and Coupland12 reported high rates of
local tumor control with adjunctive ruthenium plaque
radiotherapy, delivering a dose of 100 Gy to a depth
of 1 mm, which involves suturing the plaque over the
target area and removing it after 1 day with topical
anesthesia. They found this to be less labor intensive
than strontium brachytherapy, which requires delivery
for several days.12 Iodine plaques are more bulky, so
they are more likely to be uncomfortable for the patient; however, radiotherapy can be tailored to each individual by adjusting the number and distribution of
the iodine seeds. Such tailoring of the radiation dose
is easier with proton beam radiotherapy, which does
not require surgery but may be inconvenient for the
patient because of increased travel time. Proton beam
radiotherapy may be useful for lesions in the nonbulbar conjunctiva, which cannot easily be treated with
brachytherapy.17 Damato18 has also achieved good re-
A
B
sults with radiotherapy alone in select patients with
unresectable tumors.
Topical Interferon Immunotherapy: This type
of treatment is a relatively recent addition to the therapeutic armamentarium. Although interferon α-2b is
commonly used to treat ocular surface squamous neoplasia and conjunctival papillomas, data are limited on
its use for conjunctival melanoma.19,20 No consensus
exists regarding the most appropriate dosing regimen.
Several case series have shown it to be effective for the
treatment of conjunctival melanoma and conjunctival
melanocytic intraepithelial neoplasia with good rates
of tolerance by patients2; however, further studies are
required to examine its efficacy and safety data in patients with pigmented conjunctival lesions.
Outcomes
Loss of Vision
Loss of can occur as a result of keratitis following tumor excision, cryotherapy, or topical chemotherapy;
hypotony and macular edema can occur following
bulbar cryotherapy; and cataracts can occur following
radiotherapy.
Patient-Reported Outcomes
Outcomes reported by patients such as grittiness, discharge, epiphora, and concerns about appearance are
common, but, to our knowledge, they have not yet
been analyzed in a published study.
Local Recurrence
The rate of local recurrence of PAM/conjunctival melanocytic intraepithelial neoplasia has been reported
to be 27%.21 The reported rate of progression to invasive melanoma following treatment for PAM/conjunctival melanocytic intraepithelial neoplasia ranges
from 13% to 46%.21,22 Recurrent invasive melanoma has
been reported in 8% to 62% of study patients at 5 years
and in approximately 38% to 52% of study patients at
10 years.1,7,12,23 Factors associated with lower rates of
local recurrence include extent of initial disease, the
“no touch” surgical technique, and adjunctive therapy (particularly radiotherapy).12
Rarely, tumor recurrence can intraocularly occur, particularly if
the Bowman membrane has been
surgically disrupted and if adjunctive radiotherapy has not been
administered.24
Fig 5A–B. — Conjunctival melanoma (A) at presentation and (B) after excision with adjunctive proton beam radiotherapy and topical chemotherapy.
122 Cancer Control
Secondary Exenteration
Secondary exenteration was previously reported in 13% of study
patients.25 It has now become rare
due to improved rates of local tumor control.7,23,25,26
April 2016, Vol. 23, No. 2
Metastatic Disease
Metastasis can be regional, systemic, or both. Regional metastases develop by lymphatic spread and
involve the parotid, preauricular, postauricular, and
submandibular nodes.7 Systemic metastases can occur by lymphatic or hematogenous spread. Approximately 20% to 30% of patients develop regional
metastases; of these, 50% subsequently develop systemic metastatic disease.27,28 Conversely, up to 25% of
patients develop systemic metastases alone without
any clinical evidence of regional metastases.7 Treatment for metastatic disease is the same as for cutaneous melanoma and includes cytotoxic chemotherapy,
immunotherapy, and molecularly targeted therapy.29
The 10-year mortality rate from conjunctiva melanoma ranges from 13% to 38%.7,12
Prognostication
Ocular Predictors
Ocular predictors of regional and systemic metastatic
disease — hence mortality — include large basal tumor diameter, increased tumor thickness, and nonbulbar conjunctival involvement (particularly the caruncular tumor location).14 The tumor, node, and metastasis
(TNM) staging system of the American Joint Committee on Cancer (AJCC)30 stages conjunctival melanomas
as: T(is) (intraepithelial melanoma), T1 (bulbar), T2
(nonbulbar, including palpebral, forniceal, or caruncular), T3 (invasion into globe, eyelid, nasolacrimal system, orbit, or sinus), and T4 (invasion into brain). Two
studies have validated the current AJCC staging criteria, and both have demonstrated that higher-staged tumors have a higher risk of local recurrence, local and
distant metastasis, and death.30-32 The current staging
system is currently being revised, for example, to describe the circumferential tumor extent in quadrants irrespective of whether the tumor crosses the horizontal
or vertical meridians.4,33
Regional Nodal Involvement
Regional nodal involvement is a known predictor of
systemic metastasis and this has stimulated interest in
sentinel lymph-node biopsy.27,28,34-38 It involves injecting radionuclide (technetium 99m sulfur colloid) into
the subconjunctival space surrounding the tumor, followed by imaging of the afferent lymphatic system,
and then excising any suspicious lymph nodes, intraoperatively identifying them using a handheld γ probe.
The excised nodes are serially “bread loafed” in 1- to
2-mm slices and examined for malignant cells using
immunohistochemistry. Sentinel lymph-node biopsy
has been advocated in patients with tumors in a nonlimbal location, histological ulceration, and with a
tumor thickness of at least 2 mm.34 This procedure is
controversial because of its unproven value in predicting metastasis and prolonging survival.36 Furthermore,
April 2016, Vol. 23, No. 2
lymph-node resection can result in complications, including scarring and facial nerve (VII) palsy.
Histological Predictors
Histological predictors of metastasis include tumor
thickness exceeding 2 mm, surface ulceration, epithelioid cytomorphology, high mitotic count, and microsatellites, as well as vascular invasion or lymphatic invasion.7,39-43
Genetic Predictors
Genetic predictors of metastasis have not been identified, although BRAF, CDKN1A, RUNX2, MLH1, TIMP2,
MGMT, and ECHS1 have been reported.8 BRAF V600E
mutation occurs in about 50% of cases of conjunctival
melanomas.44 Approximately 50% of such melanomas
respond to systemic BRAF inhibitors, so routine testing
for this mutation occurs in some centers.45
Further Studies
Diagnosis
In theory, hope exists for the external validation and
refinement of the Damato–Coupland scoring system
for conjunctival melanocytic intraepithelial neoplasia4;
however, in practice, the system is limited by the rarity of invasive melanoma in such cases, because most
patients now receive treatment. High-resolution ocular
coherence tomography is now available, so this imaging modality could be used to detect various grades of
conjunctival melanocytic intraepithelial neoplasia, not
only at presentation but also following topical chemotherapy.10 Such imaging could reduce the need for conjunctival biopsies.
Treatment
In view of the morbidity caused by topical MMC, we
avoid such treatment in patients with conjunctival melanocytic intraepithelial neoplasia who have a score of
3 or below, instead preferring long-term surveillance
for such cases. It would be useful to determine how
many such patients develop more severe conjunctival
melanocytic intraepithelial neoplasia, invasive melanoma, or both. With regard to invasive disease, future
randomized trials should study extensive resection and
cryotherapy compared with minimalist resection with
radiotherapy and topical chemotherapy. Such studies should measure both patient-reported and clinical
outcomes (ie, local tumor control, ocular conservation,
vision, metastasis-free survival rates). Instruments for
measuring patient-reported outcomes must be validated in the setting of conjunctival melanoma.
Prognostication
Survival estimates for patients with choroidal melanomas have been improved by mathematical methods
that combine the AJCC staging system with histological
Cancer Control 123
grade of malignancy and genetic tumor type.29,30,46 Similar prognostication methods should be applied to patients with conjunctival melanoma. Further evaluation
of sentinel lymph-node biopsy is also warranted.27,34-38
The search for genetic predictors should continue so
that different subgroups of conjunctival melanoma are
identified, similar to that seen in uveal melanoma.47 It
would be useful to understand why nonbulbar conjunctival melanomas are associated poor rates of
survival — that is, whether this is because of genetic
differences (possibly related to sunlight exposure) or
stromal influences (eg, lymphatic density).
Systemic Adjuvant Therapy
In patients known to be at high risk for metastasis, it
would be worth undertaking randomized trials of
systemic chemotherapy or immunotherapy, similar to
those in progress for cutaneous melanoma. Because of
the rarity of conjunctival melanoma, it may be possible
to enroll patients with conjunctival melanoma in such
trials, subsequently performing subgroup analyses.
Treatment for Metastatic Disease
Similar to those of systemic adjuvant therapy, clinical
trials evaluating treatment for clinical metastases are
limited by the rarity of conjunctival melanomas, so investigators should explore the feasibility of enrolling
these patients in trials studying cutaneous melanoma.
Future Directions
Many patients with conjunctival melanoma experience
suboptimal outcomes because of iatrogenic tumor seeding, inadequate local tumor control, and surgical morbidity. The published evidence base is poor, so a need
exists for clinical trials and basic science research. Because of the rarity of conjunctival melanoma, multicenter studies are needed. Such collaboration is becoming easier with the development in electronic medical
records, automatic extraction of data, and the collaboration of ocular oncology working groups and organizations. Progress should also be encouraged by the
formation of patient advocacy groups, which can help
raise awareness of the importance of holistic care, emphasizing the need for doctor–patient communication,
adequate patient education, informed consent, effective
emotional support, and, if necessary, referral to psychology. The greatest improvement in patient care will occur when ocular oncologists are supported by a skilled
multidisciplinary team and when biopsy or treatment at
nonspecialty centers is no longer considered acceptable.
References
1. Lim LA, Madigan MC, Conway RM. Conjunctival melanoma: a review
of conceptual and treatment advances. Clin Ophthalmol. 2013;6:521-531.
2. Wong JR, Nanji AA, Galor A, et al. Management of conjunctival
malignant melanoma: a review and update. Expert Rev Ophthalmol.
2014;9(3):185-204.
124 Cancer Control
3. Damato BE, Coupland SE. Ocular melanoma. Saudi J Ophthalmol.
2012;26(2):137-144.
4. Damato B, Coupland SE. Conjunctival melanoma and melanosis:
a reappraisal of terminology, classification and staging. Clin Exp Ophthalmol.
2008;36(8):786-795.
5. Reese AB. Precancerous melanosis and malignant melanoma of the
conjunctiva and skin of the lids. Trans Am Ophthalmol Soc. 1942;40:224-232.
6. Shields CL, Shields JA. Conjunctival primary acquired melanosis
and melanoma: tales, fairy tales, and facts. Ophthal Plast Reconstr Surg.
2009;25(3):167-172.
7. Shildkrot Y, Wilson MW. Conjunctival melanoma: pitfalls and dilemmas in management. Curr Opin Ophthalmol. 2010;21(5):380-386.
8. Kenawy N, Lake SL, Coupland SE, et al. Conjunctival melanoma and
melanocytic intra-epithelial neoplasia. Eye (Lond). 2013;27(2):142-152.
9. Nanji AA, Sayyad FE, Galor A, et al. High-resolution optical coherence tomography as an adjunctive tool in the diagnosis of corneal and
conjunctival pathology. Ocul Surf. 2015;13(3):226-235.
10. Shousha MA, Karp CL, Canto AP, et al. Diagnosis of ocular surface lesions using ultra-high-resolution optical coherence tomography.
Ophthalmology. 2013;120(5):883-891.
11. Ho VH, Prager TC, Diwan H, et al. Ultrasound biomicroscopy for estimation of tumor thickness for conjunctival melanoma. J Clin Ultrasound.
2007;35(9):533-537.
12. Damato B, Coupland SE. An audit of conjunctival melanoma treatment in Liverpool. Eye (Lond). 2009;23(4):801-809.
13. Jakobiec FA, Brownstein S, Wilkinson RD, et al. Combined surgery
and cryotherapy for diffuse malignant melanoma of the conjunctiva. Arch
Ophthalmol. 1980;98(8):1390-1396.
14. Damato B, Coupland SE. Management of conjunctival melanoma.
Expert Rev Anticancer Ther. 2009;9(9):1227-1239.
15. Collins ET. Case of epibulbar sarcoma cured by removal and the
application of radium. T Ophthal Soc UK. 1918;38;125-127.
16. Lederman M. Radiotherapy of cancerous and precancerous melanosis. T Ophthal Soc UK. 1958:LXXVIII;147-164.
17. Wuestemeyer H, Sauerwein W, Meller D, et al. Proton radiotherapy
as an alternative to exenteration in the management of extended conjunctival melanoma. Graefes Arch Clin Exp Ophthalmol. 2006;244(4):438-446.
18. Damato B. Melanotic conjunctival lesions. In: Damato B. Ocular
Tumours: Diagnosis and Treatment. Oxford, UK: Butterworth-Heinemann;
2000:21-29.
19. Finger PT, Sedeek RW, Chin KJ. Topical interferon alfa in the treatment of conjunctival melanoma and primary acquired melanosis complex.
Am J Ophthalmol. 2008;145(1):124-129.
20.Herold TR, Hintschich C. Interferon alpha for the treatment of
melanocytic conjunctival lesions. Graefes Arch Clin Exp Ophthalmol.
2010;248(1):111-115.
21. Shields JA, Shields CL, Mashayekhi A, et al. Primary acquired melanosis of the conjunctiva: experience with 311 eyes. Trans Am Ophthalmol
Soc. 2007;105:61-72.
22. Folberg R, McLean IW, Zimmerman LE. Conjunctival melanosis
and melanoma. Ophthalmology. 1984;91(6):673-678.
23. Shields CL, Shields JA, Gündüz K, et al. Conjunctival melanoma:
risk factors for recurrence, exenteration, metastasis, and death in 150 consecutive patients. Arch Ophthalmol. 2000;118(11):1497-1507.
24. Sandinha T, Russell H, Kemp E, et al. Malignant melanoma of the
conjunctiva with intraocular extension: a clinicopathological study of three
cases. Graefes Arch Clin Exp Ophthalmol. 2007;245(3):431-436.
25. Shields JA, Shields CL, Gündüz K, et al. Clinical features predictive
of orbital exenteration for conjunctival melanoma. Ophthal Plast Reconstr
Surg. 2000;16(3):173-178.
26. Paridaens AD, McCartney AC, Minassian DC, et al. Orbital exenteration in 95 cases of primary conjunctival malignant melanoma. Br J
Ophthalmol. 1994;78(7):520-528.
27. Esmaeli B. Regional lymph node assessment for conjunctival melanoma: sentinel lymph node biopsy and positron emission tomography. Br J
Ophthalmol. 2008;92(4):443-445.
28. Tuomaala S, Kivelä T. Metastatic pattern and survival in disseminated conjunctival melanoma: implications for sentinel lymph node biopsy.
Ophthalmology. 2004;111(4):816-821.
29. Bhatia S, Tykodi SS, Lee SM, et al. Systemic therapy of metastatic
melanoma: on the road to cure. Oncology (Williston Park). 2015;29(2):126-135.
30. Edge SB, Byrd DR, Compton CC, et al, ed. AJCC Cancer Staging
Manual. 7th ed. New York: Springer; 2010.
31. Shields CL, Kaliki S, Al-Dahmash SA, et al. American Joint Committee on Cancer (AJCC) clinical classification predicts conjunctival melanoma outcomes. Ophthal Plast Reconstr Surg. 2012;28(5):313-323.
32. Yousef YA, Finger PT. Predictive value of the seventh edition American Joint Committee on Cancer staging system for conjunctival melanoma.
Arch Ophthalmol. 2012;130(5):599-606.
33. Damato B, Coupland SE. Clinical mapping of conjunctival melanomas. Br J Ophthalmol. 2008;92(11):1545-1549.
34. Cohen VM, Tsimpida M, Hungerford JL, et al. Prospective study of
sentinel lymph node biopsy for conjunctival melanoma. Br J Ophthalmol.
April 2016, Vol. 23, No. 2
2013;97(12):1525-1529.
35. Pfeiffer ML, Savar A, Esmaeli B. Sentinel lymph node biopsy for
eyelid and conjunctival tumors: what have we learned in the past decade?
Ophthal Plast Reconstr Surg. 2013;29(1):57-62.
36. Mendoza PR, Grossniklaus HE. Sentinel lymph node biopsy for
eyelid and conjunctival tumors: what is the evidence? Int Ophthalmol Clin.
2015;55(1):123-136.
37. Tuomaala S, Kivelä T. Sentinel lymph node biopsy guidelines for
conjunctival melanoma. Melanoma Res. 2008;18(3):235.
38.Schwarz KA, Davison SP, Crane AE. Sentinel lymph node biopsy in the setting of conjunctival melanoma. Plast Reconstr Surg.
2008;121(4):212e-213e.
39. Savar A, Esmaeli B, Ho H, et al. Conjunctival melanoma: localregional control rates, and impact of high-risk histopathologic features.
J Cutan Pathol. 2011;38(1):18-24.
40. Missotten GS, Keijser S, De Keizer RJet al. Conjunctival melanoma in the Netherlands: a nationwide study. Invest Ophthalmol Vis Sci.
2005;46(1):75-82.
41. Esmaeli B, Roberts D, Ross M, et al. Histologic features of conjunctival melanoma predictive of metastasis and death (an American Ophthalmological thesis). Trans Am Ophthalmol Soc. 2012;110:64-73.
42. Sheng X, Li S, Chi Z, et al. Prognostic factors for conjunctival melanoma: a study in ethnic Chinese patients. Br J Ophthalmol. 2015;99(7):990-996.
43. Heindl LM, Hofmann-Rummelt C, Adler W, et al. Prognostic significance of tumor-associated lymphangiogenesis in malignant melanomas of
the conjunctiva. Ophthalmology. 2011;118(12):2351-2360.
44. Lake SL, Jmor F, Dopierala J, et al. Multiplex ligation-dependent
probe amplification of conjunctival melanoma reveals common BRAF
V600E gene mutation and gene copy number changes. Invest Ophthalmol
Vis Sci. 2011;52(8):5598-5604.
45.Blum ES, Yang J, Komatsubara KM, et al. Clinical management of uveal and conjunctival melanoma. Oncology (Williston Park).
2016;30(1):29-32, 34-43, 48.
46. Damato B, Eleuteri A, Taktak AF, et al. Estimating prognosis for
survival after treatment of choroidal melanoma. Prog Retin Eye Res.
2011;30(5):285-295.
47. Bagger M, Andersen MT, Andersen KK, et al. The prognostic effect of American Joint Committee on Cancer staging and genetic status
in patients with choroidal and ciliary body melanoma. Invest Ophthalmol
Vis Sci. 2014;56(1):438-444.
April 2016, Vol. 23, No. 2
Cancer Control 125
Helpful immunostains for the
differential diagnosis of PSC include EMA,
Ber-Ep4, AR, and adipophilin.
Photo courtesy of Lynn E. Harman, MD. Half Dome From Glacier Point.
Sebaceous Carcinoma of the Eyelid
Carlos Prieto-Granada, MD, and Paul Rodriguez-Waitkus, MD, PhD
Background: Periocular sebaceous carcinoma (PSC) is a rare but aggressive neoplasm that tends to clinically
and histopathologically mimic other conditions. PSC can be challenging to diagnose using histomorphology
alone given its overlap with 2 more common tumors that occur in this area (basal cell carcinoma [BCC] and
squamous cell carcinoma [SCC]). Use of immunohistochemistry can help resolve this differential diagnosis.
Methods: A review of the literature was performed, focusing on the epidemiology, morphology, and immunohistochemical features of PSC.
Results: The most useful immunostains in the differential diagnosis of PSC are epithelial membrane antigen,
Ber-Ep4, androgen receptor (AR), and adipophilin. To discern PSC from BCC, one should use EMA, Ber-Ep4,
AR, and adipophilin, whereas discerning PSC from SCC can be achieved by evaluating AR and adipophilin.
In addition, p53 and ERBB2 (formally known as HER2/neu) are other potentially useful immunohistochemical markers for the differential diagnosis of PSC.
Conclusions: Use of new immunohistochemical techniques, as well as the elucidation of molecular alterations,
such as the presence of ERBB2 amplification, will advance our understanding of PSC.
Introduction
Cutaneous sebaceous carcinoma is a rare disease that
makes up an estimated 0.05% of all cutaneous malignancies.1,2 However, because the eyelid and periocular
regions harbor a high density of sebaceous glands either associated with eyelashes (Zeis) or the tarsal plate
(meibomian), it is not surprising that, when considering all cutaneous sebaceous carcinomas, up to 40% of
these tumors arise in this particular anatomical site.3
In the United States, sebaceous carcinoma represents
From the Departments of Dermatology & Cutaneous Surgery (CPG, PR-W) and Pathology & Cell Biology (PR-W), Morsani College of
Medicine, University of South Florida, Tampa, Florida.
Address correspondence to Carlos Prieto-Granada, MD, University of South Florida, 13330 USF Laurel Drive, Fourth floor, Tampa,
FL 33612. E-mail: [email protected]
Submitted August 15, 2015; accepted December 17, 2015.
No significant relationships exist between the authors and the
companies/organizations whose products or services may be referenced in this article.
126 Cancer Control
5% of all malignant neoplasms arising in the periocular region, followed basal cell carcinoma (BCC), which
makes up 90% of eyelid malignancies.4-6
In general, periocular sebaceous carcinoma (PSC)
presents in the elderly (average of 70 years of age).1-3
Predisposing factors, such as immunosuppression, prior history of radiotherapy to the head and neck, and
Muir–Torre syndrome, occur in a minority of patients.7
PSC can clinically present as a diffuse thickening of the
eyelid, often exhibiting inflammatory features. More
advanced cases are characterized as a discrete, palpebral mass. The clinical presentation of early-stage PSC
has the potential of mimicking non-neoplastic lesions,
notably chalazion, keratoconjunctivitis, and other inflammatory, non-neoplastic processes.5,8,9 Thus, those
with PSC are at an increased risk for delayed diagnosis.
Historically, a diagnosis of PSC was associated with a poor prognosis and a mortality rate that
reached 50%.10,11 However, with increased disease
awareness and the introduction of better diagnostic
April 2016, Vol. 23, No. 2
techniques, rates of survival have improved (mortality rates of 2%–11%).4,11,12 Early identification is crucial
and relies on a high level of clinical suspicion, together
with an accurate histopathological diagnosis that often
depends on small or limited specimens on biopsy.
Histomorphology
When examining the invasive component of PSC at
low magnification, 4 basic morphological patterns
can be recognized: trabecular, lobular, papillary, and
BCC-like (Figs 1 and 2).7 Similar to BCC, a mixture of
2 or more of these patterns can be seen in a single lesion. However, all of the classic features of BCC are
not found, such as peripheral palisading, tumor–stromal cleft artifacts, and mucinous/myxoid spindle cell
stroma. The trabecular pattern of PSC has a tendency
to reveal areas of central comedo-like necrosis. An in
situ component of PSC may be encountered, either as a
standalone lesion as well as adjacent or overlying an invasive lesion (see Figs 1C–F). The intraepithelial lesions
are often bowenoid in appearance and can exhibit exuberant pagetoid spread (see Figs 1D and 1E). They can
also radially spread by undermining the normal conjunctival epithelium (see Fig 1F). Keratinization is not
usually a feature of PSC. The presence of changing keratinization should prompt the health care professional
to consider squamous cell carcinoma (SCC) with sebaceous differentiation rather than PSC.7
In a similar fashion to cutaneous sebaceous carcinoma, cytologically PSC tumors are composed of 3 basic cell types: basaloid, sebaceous/vacuolated cells, and
intermediate forms. These cell types are usually intermixed at varying ratios, which will determine the level
of differentiation of a given tumor, among other factors
(see Figs 2D, 2E, and 2G). Broadly speaking, the percentage of basaloid cells will be inversely correlated
with the level of differentiation, which ranges from welldifferentiated tumors to poorly differentiated and anaplastic/de-differentiated tumors.
Another factor to consider when grading a lesion is
the architecture of the tumor. Well-differentiated PSC tumors are often nodular, whereas poorly differentiated
tumors are more infiltrative. Taking into account all of
these features, including grading, architecture, presence
or absence of an in situ component/pagetoid spread, as
well as presence of spread to distant sites, the following
4-tiered classification has been proposed13: PSC in situ,
low-grade, high-grade infiltrating PSC with or without
pagetoid spread, and PSC with extraocular/extracutaneous spread, including distant metastases.
In addition to its ability to clinically mimic other lesions, PSC can also overlap the histopathology of several
benign and malignant entities encountered in the periocular region. Among the benign processes, the health
care professional should consider sebaceous adenoma,14
follicular adnexal tumors (eg, desmoplastic trichoepithe-
A
B
C
D
E
F
Fig 1A–F. Composite demonstrating different morphological patterns of PSC. (A) Basal cell carcinoma–like type of configuration. H & E, ×40.
(B) Complete with superficial ulceration with the presence of vacuolated cells. H & E, ×200. (C, D) Usual presentation of PSC with extensive intraepidermal involvement exhibiting pagetoid spread. A subjacent main tumor mass is present with tumor cells expanding into the epidermis through the
follicular structures. C: H & E, ×20. D: H & E, ×100 (E) High-power imaging shows the nuclear features of PSC: vesicular chromatin with prominent
yet small nucleoli. H & E, ×200. (F) The radial extension of PSC cells through normal epithelium reveals conjunctival mucosa undermined by tumor
cells. H & E, ×40.
H & E = hematoxylin and eosin, PSC = periocular sebaceous carcinoma.
April 2016, Vol. 23, No. 2
Cancer Control 127
A
B
C
D
E
F
G
H
I
Fig 2A–I. — Panel exhibiting immunohistochemical staining patterns of 3 markers useful for the differential diagnosis of PSC. (A–C) Epithelial
membrane antigen often exhibits patchy but intense membranous and cytoplasmic positivity in PSC. An internal control is present and represented
by normal sebaceous glands (C, arrow). A: H & E, ×20. B: H & E, ×20. C: H & E, ×200. (A, D, E) Although this case exhibits a basal cell carcinoma–like
pattern (D and E: H & E, ×200), (F) Ber-Ep4 is completely negative. F: H & E, ×200. An internal control is present in the germinative portion of an
adjacent follicular structure (arrow). (G) The presence of intermediate cells with lipid vacuoles (arrows) is highlighted with adipophilin. G: H & E,
×400. (H, I) A “vacuolar-type” staining pattern is seen. H: H & E, ×200. I: H & E, ×400.
H & E = hematoxylin and eosin, PSC = periocular sebaceous carcinoma.
lioma),15 trichoadenoma,16 and squamous papilloma.17-19
When considering malignancies, the 2 main possibilities
are BCC — particularly with sebaceous differentiation
— and SCC with sebaceous differentiation (rare). The
latter also includes a basaloid variant. A rare but plausible differential diagnostic possibility — particularly
for poorly differentiated types of PSC — is Merkel cell
carcinoma, which is a malignant neoplasm with a predilection to arise in the head and neck area.20,21 When confronted with in situ PSC with abundant pagetoid spread,
other pagetoid malignancies such as pagetoid SCC in
situ, melanoma in situ, and intraepidermal Merkel cell
carcinoma should be considered.
Morphologically, the nuclear features in PSC tumor cells have a distinctive shape — namely, they
are round, oval, or indented — and contain fine, inkdrop, or smudgy chromatin with 1 or 2 small nucleoli
(see Figs 1E and 2G).7 These features are not found in
128 Cancer Control
Merkel cell carcinoma, which have washed-out, neuroendocrine-type chromatin. By contrast, melanoma
cells often display large nuclei with vesicular chromatin and macronucleoli. In a proportion of poorly differentiated types of PSC (approximately 23%–77%),
morphological assessment will be hampered by the
presence of overlapping features with other entities
included the differential diagnosis.4,22,23 In such situations, immunohistochemical stains may be helpful.
Immunohistochemistry
Among the malignant tumors that arise in the periocular region, the 3 most common are BCC, squamous
cell carcinoma (SCC), and PSC.5,6 Several immunohistochemical markers are useful in the differential diagnosis, including epithelial antigens, as are hormonal
markers and other markers highlighting fat vacuole
production. Immunohistochemical stains will also reApril 2016, Vol. 23, No. 2
flect subjacent molecular alterations. The different expression profiles using the most
useful markers for PSC, BCC,
and SCC are summarized in
the Table.
Table. — Three Commonly Used Immunohistochemical Markers for the Differential
Diagnosis of Periocular Sebaceous Carcinoma
Type of Carcinoma
Epithelial
Membrane
Antigen
Ber-Ep4
Androgen
Receptor
Adipophilin
p53
ERBB2
Periocular sebaceous
+
−/+
+
+
+
+
−
+
+/−
−
−/+
−
+
−
−
−
+/−
−
Basal cell
Epithelial Markers
Squamous cell
Epithelial antigens are tools
that can be used to help in the
differential diagnosis of PSC.
The 2 epithelial markers deemed to be most useful in
the differential diagnosis of PSC, BCC, and SCC are epithelial membrane antigen (EMA) and Ber-Ep4.7,24
Epithelial Membrane Antigen: Also known as
mucin 1, cluster designation 227, and cancer antigen
15-3 (particularly when detected in serum), EMA is a
75-kDa transmembrane glycoprotein coded by the
1q21 locus. It is commonly used as an epithelial marker
in conjunction with cytokeratins. In normal skin and
conjunctiva, EMA is expressed in glandular structures,
including sebocytes (see Fig 2C, arrow), as well as in
squamous epithelium, but not in follicular structures.25
EMA exhibits a membranous and cytoplasmic signal
(see Figs 2B and 2C) and is prominently expressed in
the majority of cases of PSC and SCC, while being predominantly negative in BCC.7,24
Ber-Ep4: Ber-Ep4 is a monoclonal antibody directed toward the epithelial cell adhesion molecule, a
transmembrane glycoprotein coded by the 2p21 locus
and normally expressed at the basolateral membrane of
cells by the majority of epithelial tissues, except in adult
squamous epithelium and by some specific epithelium cell types (eg, hepatocytes).26,27 This marker is expressed in the secretory portion of eccrine glands and
follicular germinative cells in normal skin and eyelids
(see Fig 2F, arrow).28 This antibody exhibits a predominant membranous pattern of staining; it is most useful
in distinguishing BCC from PSC and SCC because it is
frequently positive for Ber-Ep4 (70%–100%).24,28 By contrast, PSC and SCC are negative for Ber-Ep4 in 74% to
94% and 100% of cases, respectively (see Fig 2F).24,28
Cytokeratin: Intermediate filaments known as cytokeratins are expressed in carcinomas (and rarely in a
few mesenchymal tumors). Because cytokeratin is also
expressed in other entities included in the differential
diagnosis of PSC, expression of CAM 5.2 (a cytokeratin
cocktail) is not particularly useful to resolve the differential diagnosis for PSC.29 Cytokeratin 7 is expressed in
a higher percentage in PSC cases (88%) compared with
BCC cases (28%); this is especially true in SCC cases
(9%), so it could be used as an adjunctive tool.24 In addition, the expression of cytokeratin 19 may be helpful in
distinguishing sebaceous carcinomas (17% focally positive) from BCC (64% strongly positive and 14% focally
positive).30 However, given the ubiquity of cytokeratins
April 2016, Vol. 23, No. 2
in epithelial malignancies, the ability of cytokeratins
to discriminate among the common tumors around the
eye is limited.
Thomsen–Friedenreich (T) and its Precursor
(Tn) Antigen: These markers are composed of Oglycosylated, mucin-type complex carbohydrates and
are — in addition to the glycophorin protein system
— part of the MNS blood group system. They are expressed in carcinomas from a visceral origin while being hidden from the immune system via the addition of
covalently linked carbohydrates and sialic acid in the
majority of normal tissues, with the exception of mature sebocytes, the luminal aspect of the sweat glands,
and in the intercellular junctions of the spinous layer of
the epidermis.31,32 A small study specifically testing the
immunohistochemical expression of the Thomsen–
Friedenreich (T) antigen found a strong expression of
this marker in the majority of sebaceous carcinoma
cases (n = 8) tested as well as in the sebaceous adenoma (n = 15) and sebaceoma (n = 9) samples, while
being negative in the all but 1 of the tested BCC cases
(n = 13); only 1 case of BCC was positive for sebaceous
differentiation.13,32 Although no follow-up studies were
found in the literature regarding this marker, one could
consider incorporating T-antigen immunohistochemistry to resolve this differential diagnosis.
Lipid-Processing Markers
Sebaceous carcinomas are related to the production of lipids (mainly triglycerides). Historically, the
health care professional was only able to rely on the
often cumbersome histochemical demonstration of
lipids utilizing Oil red O or Sudan black IV, inconvenient stains that required a frozen tissue sample and
were associated with both poor rates of sensitivity
(40%) and stain fading over time.33 However, another
group of immunohistochemical markers demonstrating lipid production are now available. This group of
markers is composed of 3 lipid droplet–associated proteins comprised of the so-called perilipin, adipophilin,
and TIP47 group (also known as perilipins 1, 2, and 3,
respectively). These molecules are associated with the
phospholipid monolayer membrane surrounding the
triglyceride core of the intracytoplasmic lipid droplets
and are related in the formation, maintenance, modifiCancer Control 129
cation, and involution of such structures.34 All of these
markers appear specific for lipid droplets in normal
tissues as well as in a variety of malignant neoplasms
with lipogenic features.28,35-38 Although adipophilin appears to be the most sensitive marker of the group, perilipin is considered to have superior rates of specificity,
and both immunostains have proven to be of particular utility in the differential diagnosis of PSC.7,23,25,33,39,40
Adipophilin: Also known as adipocyte differentiation–related protein and perilipin 2, adipophilin is a lipid vesicle–associated protein coded by a gene in 9p22.1.
This marker is expressed in normal sebocytes. However,
when using this stain, the health care professional must
be aware of its different staining patterns, including its
diffuse cytoplasmic signal pattern, which is often encountered in well-differentiated areas of PSC tumors; its
vacuolar, vesicular-rimming pattern (see Figs 2G and 2I);
or the presence of both of these, thus highlighting the
intermediate cells of a PSC tumor; and a fine particulate, granular, or dusty pattern (considered nonspecific
and could be found in other tumors [eg, SCC, BCC]). The
health care professional must remember the poorly differentiated areas of PSC tumors will often be lipid-poor
and basaloid cell-rich; thus, they may be predominantly
negative or weakly positive for adipophilin in an overall
patchy pattern.7,23,25,33,39 Adipophilin can be useful when
differentiating PSC that is predominantly immunoreactive, showing either vacuolar or
diffuse patterns, or is predomiA
nantly negative or weakly positive
with a finely granular pattern in
the setting of BCC and SCC.
Perilipin: Perilipin is a transmembranous protein related to
lipid vesicles coded by the 15q26
locus that is expressed in sebocytes. Perilipin appears to be
more specific but less sensitive
than adipophilin.33,39 The different patterns of staining are similar
to those of adipophilin, and the
B
health care professional could use
this marker in cases for which adipophilin was unsatisfactory.
Androgen Receptor
Androgen receptor (AR) is coded
by Xq12 and forms part of a family of nuclear-located, steroid
hormone transcription factors/
receptors that bind testosterone and dihydrotestosterone.
In normal skin and conjunctiva,
AR is predominantly expressed
in follicular and adnexal structures, especially in the sebaceous
130 Cancer Control
glands.25,41,42 Because it is a transcription factor and
a hormone receptor from the steroid class, its immunohistochemical signal is predominantly nuclear (see
Fig 3). This marker can be of particularly help when
differentiating between PSC (predominantly positive,
33%–83%), SCC (predominantly negative, 90%–100%),
and BCC (occasionally negative, 50%–86%).27,28
Underlying Mutations/Genetic Aberrations
p53 Protein: The p53 protein is a transcription factor coded by TP53 (17p13.1), which induces apoptosis
when DNA damage occurs. Approximately 67% of PSC
tumors will harbor missense or nonsense TP53 mutations that are not of the ultraviolet (UV) signature type
(unrelated to UV damage).43 Patients with germline
TP53 mutations (Li–Fraumeni syndrome) frequently
develop PSC, along with other malignancies that characterize the condition.44 Because the gene mutation
rate is quite high in the setting of PSC, many of these
tumors (40%–72%) will show an overexpression (accumulation) of p53 protein on immunohistochemistry
translated by a strong nuclear signal.7,45 By contrast,
this pattern of p53 staining occurs in fewer than 20%
of BCC cases and 50% to 60% in SCC cases.7,46,47 Thus,
some have advocated for use of p53 immunohistochemistry as a diagnostic aid in the differential diagnosis of PSC.7 Both the presence of TP53 mutations and
C
Fig 3A–C. — Pattern of immunohistochemical staining with androgen receptor. (A) In this case,
periocular sebaceous carcinoma exhibited a striking bowenoid appearance predominantly composed of basaloid cells. H & E, ×40. (B, C) Androgen-receptor immunohistochemical stain demonstrated diffuse and strong nuclear positivity in the tumor cells with a weak cytoplasmic blush.
B: H & E, ×100. C: H & E, ×200.
H & E = hematoxylin and eosin.
April 2016, Vol. 23, No. 2
the consequent p53 overexpression are more frequently encountered in PSC than extraocular sebaceous carcinoma and are inversely correlated with the presence
of alterations of the mismatched repair (MMR) proteins
and Muir–Torre syndrome.45
Mismatched Repair Proteins: Assessing the expression of MMR proteins with immunohistochemistry for mutL homolog 1, mutS homolog 2, mutS homolog 6, and PMS1 homolog 2 (MLH1, MSH2, MSH6
and PMS2, MMR system components) in cutaneous
neoplasms — particularly in sebaceous adenoma —
is becoming an easy and convenient way to screen for
Muir–Torre syndrome, a variant of the Lynch syndrome
spectrum.48-50 However, by contrast to the extraocular
forms of sebaceous neoplasms, most PSC tumors appear to be unrelated to loss of MMR expression. Therefore, PSC should be considered as a poor “index lesion”
to screen for Muir–Torre syndrome.45,51,52 Nevertheless,
periocular sebaceous adenoma appears to follow the
same trend as extraocular sebaceous neoplasms in
terms of its relationship with Muir–Torre syndrome.53
ERBB2 (formally known as HER2/neu):
PSC has been shown to exhibit 2+ and 3+ patterns of
ERBB2 (formally known as HER2/neu) immunohistochemical positivity in up to 82% of cases, with 75% of
these cases showing ERBB2 amplifications by fluorescence in situ hybridization.50 Based on the results of a
study reporting on a series of 14 cases, these findings
have diagnostic value because both ERBB2 immunoreactivity of more than 1+ and gene amplification are
infrequently found in mimickers and they provide a
possible therapeutic avenue for PSC.50,54
Adhesion Protein Expression and Epithelial
Mesenchymal Transition–Related Markers: Similar to other types of aggressive carcinomas, such as
lobular carcinoma of the breast, PSC (in particular,
poorly differentiated PSC) has been shown to disrupt
the intercellular and cell stroma adhesion machinery with loss or diminished membranous E-cadherin
and beta-catenin protein expression in 53% to 83%
and 56% to 61% of cases, respectively.55,56 Loss or diminished expression of these markers was correlated
with the presence of the epigenetic promoter methylation of CDH1 (16q22.1).55
Similarly, PSC also appears to overexpress ZEB2 in
up to 68% of cases.57 ZEB2 is a transcription factor gene
closely related to the epithelial to mesenchymal transition phenomenon that also interacts with CDH1-producing gene repression and the subsequent decrease
of E-cadherin expression. The epithelial to mesenchymal transition is an embryonic process to which poorly
differentiated neoplasms regress to enhance their invasiveness and metastatic potential by increased cell
migration. The presence of the immunohistochemical
overexpression of ZEB2, as well as loss of E-cadherin
expression, and CDH1 promoter methylation in the setApril 2016, Vol. 23, No. 2
ting of PSC have been correlated with poor rates of survival.55,57 Loss, diminution, and aberrant E-cadherin expression as well as CDH1 promoter methylation58 can
also be found in both BCC59-62 and SCC,61,63 so these
findings preclude use of ZEB2 as a diagnostic marker
in this setting.
Conclusions
Several markers are differentially expressed in periocular sebaceous carcinoma (PSC) compared with lesions
in the differential diagnosis. A panel of immunohistochemical stains is recommended because different
combinations of immunoprofiles characterize these
entities, of which epithelial membrane antigen (EMA),
Ber-Ep4, androgen receptor (AR), and adipophilin
appear to be the most useful. An optimal stain panel
to discriminate PSC from basal cell carcinoma (BCC)
is presented by EMA, Ber-Ep4, AR, and adipophilin,
while distinguishing PSC from squamous cell carcinoma (SCC) can be achieved by exploring the expressions
of AR and adipophilin.
The 3 most typical immunophenotypes are the
following: (1) PSC is positive for EMA, AR, and adipophilin but negative for Ber-Ep4, (2) SCC shows a predominant immunoprofile of EMA positivity but is negative for AR, Ber-Ep42, and adipophilin, and (3) BCC is
predominantly negative for EMA and adipophilin but
positive for Ber-Ep4. Two other potential immunohistochemical markers that may be useful in the differential diagnosis are p53 and HER2/neu, both of which
will be predominantly expressed in PSC. Mismatched
repair protein immunohistochemical expression aberrations appear to be absent in the majority of cases of
PSC, because they appear to harbor TP53 mutations.
PSC as an index case to screen of Muir–Torre syndrome
is currently not recommended.
The introduction of new immunohistochemical
stains for PSC and the expansion of our knowledge
regarding the underlying molecular aberrations of
PSC, such as the presence of ERBB2 amplifications,
represent an exciting new era in understanding of
this entity. These advancements could facilitate the
early detection of PSC and possibly result in improved
patient outcomes.
References
1. Anderson HL, Joseph AK. A pilot feasibility study of a rare skin
tumor database. Dermatol Surg. 2007;33(6):693-696.
2. Kyllo RL, Brady KL, Hurst EA. Sebaceous carcinoma: review of the
literature. Dermatol Surg. 2015;41(1):1-15.
3. Dasgupta T, Wilson LD, Yu JB. A retrospective review of 1349 cases of sebaceous carcinoma. Cancer. 2009;115(1):158-165.
4.Shields JA, Demirci H, Marr BP, et al. Sebaceous carcinoma
of the eyelids: personal experience with 60 cases. Ophthalmology.
2004;111(12):2151-2157.
5. Shields JA, Demirci H, Marr BP, et al. Sebaceous carcinoma of the
ocular region: a review. Surv Ophthalmol. 2005;50(2):103-122.
6. Shields JA, Shields CL. Sebaceous adenocarcinoma of the eyelid.
Int Ophthalmol Clin. 2009;49(4):45-61.
7. Jakobiec FA, Mendoza PR. Eyelid sebaceous carcinoma: clinicoCancer Control 131
pathologic and multiparametric immunohistochemical analysis that includes adipophilin. Am J Ophthalmol. 2014;157(1):186-208.e182.
8. Buitrago W, Joseph AK. Sebaceous carcinoma: the great masquerader: emgerging concepts in diagnosis and treatment. Dermatol Ther.
2008;21(6):459-466.
9. Older JJ. Eyelid carcinoma--another one of medicine’s imitators.
Ophthalmic Surg. 1989;20(1):73.
10. Boniuk M, Zimmerman LE. Sebaceous carcinoma of the eyelid,
eyebrow, caruncle, and orbit. Trans Am Acad Ophthalmol Otolaryngol.
1968;72(4):619-642.
11. Rao NA, Hidayat AA, McLean IW, et al. Sebaceous carcinomas of
the ocular adnexa: A clinicopathologic study of 104 cases, with five-year
follow-up data. Hum Pathol. 1982;13(2):113-122.
12. Muqit MM, Roberts F, Lee WR, et al. Improved survival rates in
sebaceous carcinoma of the eyelid. Eye (Lond). 2004;18(1):49-53.
13. Hassanein AM. Sebaceous carcinoma and the T-antigen. Semin
Cutan Med Surg. 2004;23(1):62-72.
14. Tok L, Tok OY, Argun M, et al. Corneal limbal sebaceous adenoma.
Cornea. 2014;33(4):425-427.
15. Cabral ES, Cassarino DS. Desmoplastic tricholemmoma of the
eyelid misdiagnosed as sebaceous carcinoma: a potential diagnostic pitfall. J Cutan Pathol. 2007;34(suppl 1):22-25.
16. Lever JF, Servat JJ, Nesi-Eloff F, et al. Trichoadenoma of an eyelid
in an adult mimicking sebaceous cell carcinoma. Ophthal Plast Reconstr
Surg. 2012;28(4):e101-102.
17. Jakobiec FA, Mendoza PR, Colby KA. Clinicopathologic and immunohistochemical studies of conjunctival large cell acanthoma, epidermoid dysplasia, and squamous papilloma. Am J Ophthalmol. 2013;156(4):830-846.
18. Khong JJ, Leibovitch I, Selva D, et al. Sebaceous gland carcinoma of the eyelid presenting as a conjunctival papilloma. Clin Experiment
Ophthalmol. 2005;33(2):197-198.
19. Stagner AM, Jakobiec FA, Chi A, et al. Conjunctival inverted squamous papilloma: a case report with immunohistochemical analysis and
review of the literature. Surv Ophthalmol. 2015;60(3):263-268.
20. Missotten GS, de Wolff-Rouendaal D, de Keizer RJ. Merkel cell
carcinoma of the eyelid review of the literature and report of patients with
Merkel cell carcinoma showing spontaneous regression. Ophthalmology.
2008;115(1):195-201.
21. Slutsky JB, Jones EC. Periocular cutaneous malignancies: a review
of the literature. Dermatol Surg. 2012;38(4):552-569.
22. Doxanas MT, Green WR. Sebaceous gland carcinoma. Review of
40 cases. Arch Ophthalmol. 1984;102(2):245-249.
23. Ostler DA, Prieto VG, Reed JA, et al. Adipophilin expression in sebaceous tumors and other cutaneous lesions with clear cell histology: an
immunohistochemical study of 117 cases. Mod Pathol. 2010;23(4):567-573.
24. Plaza JA, Mackinnon A, Carrillo L, et al. Role of immunohistochemistry in the diagnosis of sebaceous carcinoma: a clinicopathologic and immunohistochemical study. Am J Dermatopathol. 2015;37(11):809-821.
25. Ansai S, Arase S, Kawana S, et al. Immunohistochemical findings
of sebaceous carcinoma and sebaceoma: retrieval of cytokeratin expression by a panel of anti-cytokeratin monoclonal antibodies. J Dermatol.
2011;38(10):951-958.
26. Pai RK, West RB. MOC-31 exhibits superior reactivity compared with
Ber-EP4 in invasive lobular and ductal carcinoma of the breast: a tissue microarray study. Appl Immunohistochem Mol Morphol. 2009;17(3):202-206.
27. Winter MJ, Nagtegaal ID, van Krieken JH, et al. The epithelial cell
adhesion molecule (Ep-CAM) as a morphoregulatory molecule is a tool in
surgical pathology. Am J Pathol. 2003;163(6):2139-2148.
28. Ansai S, Takeichi H, Arase S, et al. Sebaceous carcinoma: an immunohistochemical reappraisal. Am J Dermatopathol. 2011;33(6):579-587.
29. Sramek B, Lisle A, Loy T. Immunohistochemistry in ocular carcinomas. J Cutan Pathol. 2008;35(7):641-646.
30. Heyl J, Mehregan D. Immunolabeling pattern of cytokeratin 19 expression may distinguish sebaceous tumors from basal cell carcinomas.
J Cutan Pathol. 2008;35(1):40-45.
31. Kanitakis J, al-Rifai I, Faure M, et al. Thomsen-Friedenreich and
its precursor (Tn) antigen expression in normal skin and in benign cutaneous tumours: a marker for sebaceous differentiation. Acta Derm Venereol.
1998;78(3):173-176.
32. Hassanein AM, Al-Quran SZ, Kantor GR, et al. Thomsen-Friedenreich
(T) antigen: a possible tool for differentiating sebaceous carcinoma from its
simulators. Appl Immunohistochem Mol Morphol. 2001;9(3):250-254.
33. Muthusamy K, Halbert G, Roberts F. Immunohistochemical staining
for adipophilin, perilipin and TIP47. J Clin Pathol. 2006;59(11):1166-1170.
34. Straub BK, Herpel E, Singer S, et al. Lipid droplet-associated PAT-proteins show frequent and differential expression in neoplastic steatogenesis.
Mod Pathol. 2010;23(3):480-492.
35. Meadows JW, Pitzer B, Brockman DE, et al. Expression and localization of adipophilin and perilipin in human fetal membranes: association
with lipid bodies and enzymes involved in prostaglandin synthesis. J Clin
Endocrinol Metab. 2005;90(4):2344-2350.
36. Mariano FV, dos Santos HT, Azanero WD, et al. Mammary analogue
secretory carcinoma of salivary glands is a lipid-rich tumour, and adipophilin
can be valuable in its identification. Histopathology. 2013;63(4):558-567.
132 Cancer Control
37. Moritani S, Ichihara S, Hasegawa M, et al. Intracytoplasmic lipid accumulation in apocrine carcinoma of the breast evaluated with adipophilin
immunoreactivity: a possible link between apocrine carcinoma and lipidrich carcinoma. Am J Surg Pathol. 2011;35(6):861-867.
38. Hoffman A, Ghadimi MP, Demicco EG, et al. Localized and metastatic myxoid/round cell liposarcoma: clinical and molecular observations.
Cancer. 2013;119(10):1868-1877.
39.Boussahmain C, Mochel MC, Hoang MP. Perilipin and adipophilin expression in sebaceous carcinoma and mimics. Hum Pathol.
2013;44(9):1811-1816.
40. Milman T, Schear MJ, Eagle RC Jr. Diagnostic utility of adipophilin immunostain in periocular carcinomas. Ophthalmology. 2014;121(4):964-971.
41. Ruizeveld de Winter JA, Trapman J, Vermey M, et al. Androgen
receptor expression in human tissues: an immunohistochemical study.
J Histochem Cytochem. 1991;39(7):927-936.
42. Asadi-Amoli F, Khoshnevis F, Haeri H, et al. Comparative examination of androgen receptor reactivity for differential diagnosis of sebaceous
carcinoma from squamous cell and basal cell carcinoma. Am J Clin Pathol.
2010;134(1):22-26.
43. Kiyosaki K, Nakada C, Hijiya N, et al. Analysis of p53 mutations
and the expression of p53 and p21WAF1/CIP1 protein in 15 cases of sebaceous carcinoma of the eyelid. Invest Ophthalmol Vis Sci. 2010;51(1):7-11.
44. Baumuller S, Herwig MC, Mangold E, et al. Sebaceous gland carcinoma of the eyelid masquerading as a cutaneous horn in Li--Fraumeni
syndrome. Br J Ophthalmol. 2011;95(10):1470, 1478.
45. Shalin SC, Sakharpe A, Lyle S, et al. p53 staining correlates with
tumor type and location in sebaceous neoplasms. Am J Dermatopathol.
2012;34(2):129-135; quiz 136-128.
46. Barrett TL, Smith KJ, Hodge JJ, et al. Immunohistochemical nuclear staining for p53, PCNA, and Ki-67 in different histologic variants of basal
cell carcinoma. J Am Acad Dermatol. 1997;37(3 Pt 1):430-437.
47. Athanassiadou AM, Lazaris AC, Patsouris E, et al. Significance of
cyclooxygenase 2, EZH-2 polycomb group and p53 expression in actinic
keratosis and squamous cell carcinomas of the skin. Am J Dermatopathol.
2013;35(4):425-431.
48. Boennelycke M, Thomsen BM, Holck S. Sebaceous neoplasms
and the immunoprofile of mismatch-repair proteins as a screening target
for syndromic cases. Pathol Res Pract. 2015;211(1):78-82.
49. Mathiak M, Rutten A, Mangold E, et al. Loss of DNA mismatch repair proteins in skin tumors from patients with Muir-Torre syndrome and
MSH2 or MLH1 germline mutations: establishment of immunohistochemical analysis as a screening test. Am J Surg Pathol. 2002;26(3):338-343.
50. Plocharczyk EF, Frankel WL, Hampel H, et al. Mismatch repair
protein deficiency is common in sebaceous neoplasms and suggests
the importance of screening for Lynch syndrome. Am J Dermatopathol.
2013;35(2):191-195.
51. Rajan Kd A, Burris C, Iliff N, et al. DNA mismatch repair defects and
microsatellite instability status in periocular sebaceous carcinoma. Am J
Ophthalmol. 2014;157(3):640-647.e641-642.
52. Liau JY, Liao SL, Hsiao CH, et al. Hypermethylation of the CDKN2A gene promoter is a frequent epigenetic change in periocular sebaceous carcinoma and is associated with younger patient age. Hum Pathol.
2014;45(3):533-539.
53. Jagan L, Zoroquiain P, Bravo-Filho V, et al. Sebaceous adenomas of
the eyelid and Muir-Torre Syndrome. Br J Ophthalmol. 2015;99(7):909-913.
54. Kwon MJ, Shin HS, Nam ES, et al. Comparison of HER2 gene amplification and KRAS alteration in eyelid sebaceous carcinomas with that
in other eyelid tumors. Pathol Res Pract. 2015;211(5):349-355.
55. Jayaraj P, Sen S, Sharma A, et al. Epigenetic inactivation of the
E-cadherin gene in eyelid sebaceous gland carcinoma. Br J Dermatol.
2012;167(3):583-590.
56. Li L, Zhang Z, Li B, et al. E-cadherin and beta-catenin expression in
sebaceous eyelid adenocarcinomas. Graefes Arch Clin Exp Ophthalmol.
2011;249(12):1867-1873.
57. Bhardwaj M, Sen S, Sharma A, et al. ZEB2/SIP1 as novel prognostic indicator in eyelid sebaceous gland carcinoma. Hum Pathol. 2015.
58. Chiles MC, Ai L, Zuo C, et al. E-cadherin promoter hypermethylation in preneoplastic and neoplastic skin lesions. Mod Pathol.
2003;16(10):1014-1018.
59. Pizarro A, Benito N, Navarro P, et al. E-cadherin expression in basal
cell carcinoma. Br J Cancer. 1994;69(1):157-162.
60. Pizarro A. E-cadherin expression is frequently reduced in infiltrative
basal cell carcinoma. J Dermatol. 2000;27(12):804-805.
61. Papadavid E, Pignatelli M, Zakynthinos S, et al. Abnormal immunoreactivity of the E-cadherin/catenin (alpha-, beta-, and gamma-) complex
in premalignant and malignant non-melanocytic skin tumours. J Pathol.
2002;196(2):154-162.
62. Papanikolaou S, Bravou V, Gyftopoulos K, et al. ILK expression in
human basal cell carcinoma correlates with epithelial-mesenchymal transition markers and tumour invasion. Histopathology. 2010;56(6):799-809.
63. Koseki S, Aoki T, Ansai S, et al. An immunohistochemical study of
E-cadherin expression in human squamous cell carcinoma of the skin: relationship between decreased expression of E-cadherin in the primary lesion
and regional lymph node metastasis. J Dermatol. 1999;26(7):416-422.
April 2016, Vol. 23, No. 2
The role that vismodegib will
play in advanced periocular
basal cell carcinoma depends on
its postmarketing results.
Photo courtesy of Lynn E. Harman, MD. Hohenschwangau Castle.
Role of Vismodegib in the Management of Advanced Periocular
Basal Cell Carcinoma
Kyle F. Cox, MD, and Curtis E. Margo, MD
Background: Vismodegib is the first selective hedgehog pathway inhibitor approved to treat locally advanced
and metastatic basal cell carcinoma (BCC). Limited information is available concerning its role in managing
advanced BCC around the eye.
Methods: The medical literature was searched for cases of nonsyndromic periocular BCC treated with
vismodegib. Clinical information was abstracted and analyzed. In addition, a review of the pharmacology of
vismodegib, including general effectiveness and safety, was conducted.
Results: Thirty study patients with nonsyndromic periocular BCC treated with vismodegib were found in the
literature. Vismodegib was used in 3 ways: medical therapy, adjuvant therapy prior to surgery or radiotherapy,
and treatment of positive surgical margins. Complete regression was reported in 9 study patients (30%), with
follow-up visits after therapy averaging fewer than 5 months. Four study participants developed squamous
cell carcinoma while receiving treatment.
Conclusions: Too few cases exist to draw any conclusions on the role that vismodegib might play in the management of periocular BCC. In addition, long-term follow-up data are not yet available. Although the objective
response rate of advanced BCC is impressive in study patients receiving vismodegib, well-controlled clinical
studies are needed to determine whether vismodegib has any impact on survival or quality of life.
Introduction
In general, periocular basal cell carcinoma (BCC) is
usually treated with surgical excision and, when indicated, microscopic control of margins. Typically, recurrence rates are low and functional outcomes are good;
however, some tumors because of their size, location,
From the Departments of Ophthalmology (KFC, CEM) and Pathology
and Cell Biology (CEM), University of South Florida Morsani
College of Medicine, Tampa, Florida.
Address correspondence to Kyle F. Cox, MD, Department
of Ophthalmology, Morsani College of Medicine, MDC Box
21, 12901 Bruce B. Downs Boulevard, Tampa, FL 33612.
E-mail: [email protected]
Submitted July 13, 2015; accepted January 5, 2016.
No significant relationships exist between the authors and the
companies/organizations whose products or services may be referenced in this article.
April 2016, Vol. 23, No. 2
or lack of therapeutic response are inoperable.1 In such
cases, the rates of continued morbidity are high and
the rates of cure are low.1
Vismodegib is a selective hedgehog pathway
inhibitor. It was approved by the US Food and Drug
Administration (FDA) in 2012 and the European Medicines Agency in 2013 for the treatment of locally advanced and metastatic types of BCC.2-4 Given the rarity of these conditions and lack of treatment options,
approval was based on noncomparative clinical series
of study participants. Phase 1/2 trials showed favorable primary end point responses for both locally advanced and metastatic BCC.5 In a phase 2 study, 43% of
patients with locally advanced BCC showed partial or
complete responses, whereas 30% of those with metastatic tumor responded to treatment.6
Cancer Control 133
Since the US and European approvals of vismodegib,2-4 case reports and small clinical series have appeared in the literature reflecting how the therapy is
used in clinical practice. Some of these reports involve
periocular (or ocular adnexal) BCC.7-13 Use of vismodegib for BCC around the eye poses unique challenges
and opportunities because conjunctival and orbital involvement may result in blindness and death.1
Hedgehog Pathway
The hedgehog pathway is a signaling system that
helps regulate cell growth and development. Initially
described in Drosophila, the hedgehog pathway controls a range of cellular activities in invertebrates and
vertebrates, starting during embryogenesis. Among
vertebrates, the hedgehog genes encode polypeptides
that, after modification by cholesterol, attach to cell
surfaces where they affect signaling. The functional
receptors for hedgehog are 2 transmembrane proteins
called patched 1 and smoothened. The discovery that
aberrant activity of the pathway was linked to several human tumors spurred investigation into drugs
that could alter various components of the signaling
sequence.14 Interest into the therapeutic potential of
hedgehog pathway inhibitors was stimulated further
when researchers tied the signaling system to diverse
functions such as angiogenesis and potentially metastatic spread.15
BCC is driven by mutations in 1 or more hedgehog genes.16 Typically, these alterations take the form
of a loss-of-function mutation in PTCH1 or an activating mutation in SMO. Nevoid basal cell carcinoma syndrome (also called Gorlin–Goltz syndrome) is caused
by a germline mutation in PTCH1. The mechanism or
mechanisms by which PTCH1 mutations eventually alter downstream transcription factors is an area of active investigation.16
Pharmacology
Vismodegib is the first hedgehog inhibitor approved
for the treatment of BCC.17 It appears to exert its clinical effect by binding to the smoothened receptor protein, preventing nuclear localization of transcription
factors that target gene induction.14,18 This in turn can
promote cellular proliferation, inhibit apoptosis, and
further prolong cell survival through mechanisms
such as angiogenesis. Compared with other small
molecules capable of blocking this receptor, the pharmacokinetic profile of vismodegib was considered
better suited for human use.19 Approximately one-third
of a 150-mg oral dose of vismodegib (the recommended daily dose) is absorbed; more than 99% is bound
to plasma proteins (mostly albumin).20 Vismodegib
is synthetically derived and not extensively metabolized; more than 80% is excreted in feces.20,21 Metabolic oxidation in the liver is incompletely understood.20
134 Cancer Control
After continuous daily dosing, the elimination halflife of vismodegib is 4 days, although the half-life of a
single dose can be up to 12 days.20-22 Its bioavailability
is nonlinear, decreasing with increasing strength and
frequency of administration.20,23
Studies
Phase 1
Although no dose-limiting toxicities were found
among an initial cohort of patients enrolled in an
open-label phase 1 trial, daily doses higher than 150 mg
demonstrated no substantially greater steady-state
plasma concentration.24 Once the maximum tolerated
dose was determined to be 150 mg/day, enrollment
of the phase 1 study expanded to include 68 patients
with advanced/metastatic BCC and other solid tumors.25 In this study, tumor response was only seen
among participants with BCC and medulloblastoma.25
Six participants (9%) experienced life-threatening or
disabling adverse events and 20 participants (28%)
experienced severe adverse events; however, the
majority of adverse events experienced were mild to
moderate in nature (eg, muscles spasms, fatigue, alopecia, dysgenesia, diminished appetite).25 Those with
BCC had an overall response rate of 55%, stable disease was seen in 33%, and the median duration of response was 12.8 months (range, 3.7–26.4 months).25
Five deaths (7%) were reported.25
Phase 2
Two phase 2 clinical trials followed, one of which was
a dual-cohort study of 96 volunteers with locally advanced/metastatic BCC; the other was a randomized
trial involving 41 study patients with nevoid basal
cell carcinoma syndrome.6,26 The objective response
rate for 63 study patients with locally advanced disease was 43%; of those participants, 13 (21%) had a
complete response.6 The median duration of response
was 7.6 months (range, 1–12.9 months), and 34 of the
63 specimens (54%) obtained via biopsy showed no
residual tumor.6 The median duration of drug exposure was approximately 10 months.6 Anatomical locations of individual tumors were not available, so
analysis of advanced periocular BCC could not be
performed.
Each study patient in the dual cohort trial experienced 1 or more adverse event, and 12% of those
resulted in treatment discontinuation.6 Serious adverse events occurred in 26 study patients (27%); of
those, 7 (7%) of those events were grade 5 and fatal.6
The clinical context of the fatalities were incompletely presented, but the authors found no drug-related
culpability because all 7 volunteers had preexisting
comorbidities.6
Vismodegib displayed similar rates of effectiveness
in reducing BCC in persons with nevoid basal cell carApril 2016, Vol. 23, No. 2
cinoma syndrome.6,26 Objective tumor shrinkage was
seen in 65% of treated study patients compared with
11% in those receiving placebo (P = .003), but the rate
of adverse events led to treatment discontinuation in
more than 50% of volunteers (14/26).26
A 12-month update of a phase 2 trial found a 4.7%
increase in objective response rate for study patients
with advanced BCC (from 42.9% to 47.6%).27 The median duration of response in these patients also increased from 7.7 to 9.5 months, and no new safety concerns were reported.27
An interim analysis of an international, open-label trial with safety as its primary end point used data
from 499 study volunteers with advanced/metastatic
BCC.28 Treatment was discontinued in 400 volunteers
(80%), 180 (36%) experienced adverse events, 70 (14%)
had disease progression, and 51 (10%) requested treatment be stopped.28 Overall response rate for those
with advanced BCC was 66.7% (302/453); of the 302
cases of clinical responses, 153 of those were complete
responses.28 Deaths were reported in 31 study volunteers, 21 of which were considered to be due to a treatment-related adverse event.28
An expanded access, open-label multicenter study
of 150 mg vismodegib daily in 119 participants with advanced BCC demonstrated an objective response rate
of 46% in those with locally advanced disease and 31%
in those with metastatic BCC.17 Inclusion criteria was a
tumor diameter of at least 1 cm and either confirmation
of an inoperable tumor or a contraindication to surgery.17 Study patients were followed for 6.5 months17;
during that time, the rate of adverse events paralleled
those previously reported.6 Four grade 4 treatmentemergent adverse events and 2 treatment-related
deaths were reported.17
Treatment Results
The outcomes of periocular tumors are difficult to
identify because site-specific characteristics are not always available in postmarketing clinical series of BCC.
In cases that can be identified as periocular, vismodegib has been used as neoadjuvant therapy prior to
surgery and radiotherapy and as treatment for positive
surgical margins.7-13,17,28,29 We were able to find 30 patients for whom sufficient clinical information could be
abstracted for analysis (Table 1).7-13
Vismodegib was reasonably effective in a series
of 7 study patients with advanced periocular BCC
not amenable to surgery or radiotherapy whose largest tumor, on average, was 3.4 cm in size.7 None had
metastatic disease, but 4 study patients had orbital involvement.7 All study patients had BCC that had previously failed controlled surgical resection.7 The average
duration of treatment with vismodegib was 11 weeks
(range, 4–16 weeks), and the mean duration of followup was 7.3 months (range, 5–10 months).7 Two study
April 2016, Vol. 23, No. 2
patients (29%) demonstrated complete regression,
2 (29%) had more than 50% regression, 2 (29%) had
less than 50% regression, and 1 (14%) had disease progression.7 Six (86%) reported experiencing adverse
events similar to those previously documented (eg, alopecia, muscle cramps).6,7,24-26 Two study patients also
developed squamous cell carcinoma (SCC).7
Demirci et al8 described the results of a study involving 8 patients with periocular and orbital BCC,
one of whom had nevoid basal cell carcinoma syndrome. Of the 7 nonsyndromic study patients, 5 received vismodegib alone for treatment.8 Four obtained
partial response, and, at the time of publication, all
4 were still taking vismodegib.8 One study patient
with complete response was followed for 3 months.8
In another study patient, vismodegib was used as adjuvant therapy for involved surgical margins following
surgery and in another study patient as neoadjuvant
therapy prior to surgery.8 In both of these roles, tumor response was deemed to be complete during observation periods of 11 and 16 months, respectively.8
Adverse events were characterized as comparable in
severity and frequency as those seen in earlier studies.6,7,24-26 Data from 1 patient in this series were also
published in a separate report.7
In a series of 13 study patients with high-risk BCC
for whom vismodegib was used as neoadjuvant therapy prior to surgery, 2 tumors were located around the
eye.9 One tumor with a surface area of 2.8 cm2 had an
86% reduction in its size prior to surgery, and the resected tissue showed histological cure.9 The second tumor with a surface area of 2.0 cm2 regressed 5% and,
when resected, it was still histologically present.9
Vismodegib has also been used as adjuvant
therapy prior to radiotherapy in a man with recurrent BCC of the medial canthus and lower eyelid.10
After receiving 150 mg/day vismodegib for 2 months,
cross-sectional measurements of his tumor decreased
from 6.5 × 7.4 mm to 6.3 × 5.6 mm prior to radiotherapy.10 Following radiotherapy, he was deemed to be disease free at 12 months by magnetic resonance imaging.10
Two other cases of periocular BCC treated with
vismodegib have been reported because the study patients developed SCC (keratoacanthoma types) within
2 and 7 weeks of starting therapy.11 Both secondary
cancers arose in previously documented normal skin.11
Meaningful clinical follow-up beyond the context of
the new tumors was not provided.11
A man aged 84 years with left upper eyelid and orbital BBC was treated with vismodegib and had complete response in 3 months; however, the tumor recurred and progressed after 9 months.12 Vismodegib
was stopped at 18 months and orbital exenteration was
performed the next month.12 The case was published
as an example of secondary resistance.
A retrospective, interventional case series includCancer Control 135
Table 1. — Study Patients With Nonsyndromic Periocular Basal Cell Carcinoma Treated With Vismodegib
Study
Age,
y
Sex
Orbital
Involvement
TNM Stage
Duration
of Therapy,
mo
Response
Intervention
After
Vismodegib
Follow-Up
After Treatment,
mo
Comment
Aasi11
53
M
N
NA
1.75
Complete
—
0
New case of SCC
Ally9
Demirci8
Gill7
Ozgur 13
100
W
N
NA
Unknown
“Regression”
—
Unknown
New case of SCC
39
W
N
NA
3.5
Partial
Surgery
11
—
100
W
N
NA
4
Complete
Surgery
20
—
60
M
Y
T3bN0M0
8
Complete
—
11
Used to treat
positive surgical
margin
60
M
Y
T4N0M0
5
Partial
—
0
—
63
W
Y
T3bN0M0
7
Partial
—
0
—
73
M
N
T3aN0M0
9
Complete
—
3
—
79
M
Y
T3bN0M0
13
Partial
—
0
—
79
M
a
Y
T3bN0N0
5
Complete
Surgery
16
Adjuvant prior to
surgery
86
M
Y
T3bN0M0
7
Partial
—
0
—
—
New case of SCC
43
M
N
NA
1
Complete
—
10
53
60
M
M
N
N
NA
NA
1.75
2.5
Complete
Partial
—
—
8
6
75
M
Y
NA
4
Partial
—
8
—
75
W
Y
NA
4
Progression
—
8
—
91
M
Y
NA
2.25
Partial
—
6
—
—
101
W
Y
NA
4
Partial
—
5
New case of SCC
51
W
N
T4N2cM1
9
Partial
—
0
—
56
M
N
T3bN0M0
18
Complete
—
0
—
58
M
Y
T3bN1M0
7
Partial
—
0
—
64
M
N
T3bN0M0
9
Partial
—
0
—
65
M
N
T4N1M1
18
Progression
—
2
Died of disease
69
W
N
T3bN0M0
10
Partial
—
0
Lost to follow-up
70
M
Y
T3bN0M0
11
Stable
—
0
—
76
M
N
T2N1M1
13
Stable
—
0
—
84
M
N
T3bN0M0
12
Complete
—
0
—
86
M
N
T3bN0M0
11
Partial
—
13
Recurrence after
3 mo
Papastepfanous12
84
M
Y
NA
18
Partial
Exenteration
0
Recurrence after
initial response
Pollom10
70
M
Y
NA
2
Partial
RT
12
Adjuvant prior
to RT
Average
7.6
4.8
Data reported through December 2015. All study patients were white.
from this study patient reported in another publication.
M = man, N = no, NA = not applicable, RT = radiotherapy, SCC = squamous cell carcinoma, TNM = tumor, node, metastasis, W = woman, Y = yes.
a Data
ed 10 study patients with locally advanced BCC, 4 of
whom had metastatic disease.13 Of the 6 study patients
without metastasis, 2 had a complete response.13 Five
would have needed exenteration but instead avoided
surgery with vismodegib treatment, although, at the
time of reporting, the follow-up period was short (or
none at all) after stopping vismodegib.13 One study
136 Cancer Control
patient died of progressive disease.13 The profile of reported adverse events was similar to that previously
described.6,13,24-26
It is worth noting that, of the 30 cases of advanced
eyelid and periocular BCC treated with vismodegib to
date, the average follow-up period after stopping vismodegib is 4.8 months (see Table 1).7-13
April 2016, Vol. 23, No. 2
Inoperable and Locally Advanced Disease
The role that vismodegib might play in the clinical
treatment of inoperable and locally advanced BCC depends on the perception of what these terms actually
mean. Because these conditions have no consensus
definition, a panel sponsored by Roche (Basel, Switzerland) of oncologists, dermatologists, and radiation
oncologists helped to establish the guidelines for these
terms.30 Although the panel addressed the management of periocular BCC, no member of the group was
an ocular oncologist or oculoplastic surgeon.30
The proposed guidelines of inoperability for
use of vismodegib was based on the tumor, node,
and metastasis staging classification of the American
Joint Committee of Cancer for eyelid carcinoma (Table 2).30,31 Consensus was defined as majority opinion
of the 9-member panel.30 Tumors greater than 2 cm in
size that do not invade adjacent ocular or orbital structures (T3a) should be first considered for radiotherapy.27 The panel also recommended that eyelid tumors
of stages T3a (invasive), T3b, and T4 not appropriate for
radical local therapy be assessed for possible systemic
therapy with vismodegib.30 The panel also noted that
additional factors must be considered in determining
the suitability of medical therapy, including general
medical health, severe disfigurement, and loss of function.30 However, the panel’s recommendations were not
ideal because guidelines for inoperable tumors defer to
a judgment of what constitutes “not appropriate for radical local therapy.”30
Any guidelines recommending vismodegib as alternative therapy to surgery must weigh the limited
knowledge of vismodegib against its long-term outcomes. For example, locally advanced BCC involving
the orbit would require surgical exenteration (stage
T3b) for cure.30 In an otherwise healthy patient, this
surgery should confer long-term, tumor-free survival,
albeit with a significant cosmetic cost. However, medical treatment with vismodegib offers an approximately
30% likelihood of complete response and an unknown
risk of recurrence beyond 1 year.28 In addition, the
long-term behavior of regressed BCC once vismodegib
is discontinued is unknown.
compounds that overcome vismodegib resistance in
vitro are under development for clinical use.36
Information is limited about the drug–drug interactions of vismodegib, and the safety of vismodegib in persons with renal and hepatic dysfunction
is unknown.4,5 A man aged 72 years treated with
vismodegib developed cholestatic hepatic dysfunction 1 month after beginning treatment.37 The patient
self-medicated with aspirin and naproxen because of
vismodegib-related myalgias, suggesting a drug interaction may have been involved.37
Ventarola and Silverstein 38 searched the FDA
Adverse Events Reporting System from January
2012 through January 2013 for cases of vismodegib
hepatic toxicity, and they found 65 cases of vismodegib-associated reactions, the most frequent of
which was gastrointestinal distress (34%) followed
by liver toxicity (23%). In 4 cases, vismodegib was
considered the sole agent implicated in the hepatotoxic event.38 However, Ventarola and Silverstein 38
Table 2. — Tumor Staging for Carcinoma of the Eyelid
Description
Comment
TX
Stage
Primary tumor cannot be assessed
—
T0
No evidence of primary tumor
T1
Tumor ≤ 5 mm in greatest dimension
and
Does not invade tarsus or margin
of eyelid
Average horizontal
length of adult eyelid:
~ 3 cm
T2a
Tumor > 5 mm but ≤ 10 mm in
greatest dimension
or
Any tumor than invades tarsus or
involves margin of eyelid
Average vertical
length of upper
tarsus: 10–12 mm
Average vertical
length of lower tarsus:
~ 4 mm
T2b
Tumor > 10 mm but ≤ 20 mm in
greatest dimension
or
Tumor involves entire thickness
of eyelid
April 2016, Vol. 23, No. 2
—
T3a
Tumor > 20 mm in greatest dimension
or
Any tumor than invades adjacent
ocular or orbital structures
or
Any tumor with perineural tumor
invasion
Adjacent ocular
structures include
bulbar conjunctiva
Anterior orbit begins
at orbital septum
T3b
Complete resection of tumor
requires enucleation, exenteration, or
bone resection
Exenteration involves
removal of orbital
contents, including eye
T4
Tumor unresectable due to extensive invasion of ocular, orbital, or
craniofacial structures or brain
Other Observations
Four cases of secondary SCC were reported among
the 19 patients treated for periocular BCC.7,11 Rapidly
growing SCC has also been observed in persons receiving treatment with vismodegib for BCC located
elsewhere.32,33
Some types of BCC do not respond to vismodegib, and some of these failures may reflect mutational resistance, which is a new area of investigation
likely to gain momentum during the postmarketing
period.18,34,35 At least 1 case of periocular BCC with
secondary resistance has been described.12 Novel
—
—
Adapted from Edge S, Byrd DR, Compton CC, et al, eds. AJCC Cancer
Staging Manual. 7th ed. New York: Springer; 2010. Republished with
permission of Springer; permission conveyed through Copyright Clearance Center, Inc.
Cancer Control 137
stressed that inclusion in the FDA database does not
mean the relationship is causal.
Cost
The cost of vismodegib will depend on factors such as
geographical area and markup of intermediate providers.39,40 When made available, the cost of standard daily
treatment in clinical studies has ranged from $7,500 to
9,000 per month.9,20 In a single case series of 12 study
patients treated for periocular BCC, 2 (17%) discontinued treatment because of cost.13
Alternative Therapies
Vismodegib was approved by the FDA and European
Medicines Agency for the treatment of locally advanced and metastatic BCC without the benefit of a
randomized clinical trial because locally advanced and
metastatic BCC are uncommon and evidence of effective alternative therapies is lacking.2,4 Historically, patients may have been treated with agents like cisplatinum, which, although not curative, induce a transient
reduction in tumor size.41,42 Some investigators have
used cisplatinum as neoadjuvant therapy to reduce the
tumor size of periocular BCC prior to surgery,41 whereas others have used cisplatinum/doxorubicin to treat
advanced BCC around the eye with only short-term
benefit.42,43 However, experience with these chemotherapeutic agents is limited in the setting of periocular BCC, and use of these agents has been associated
with serious adverse events.41-44
Treatment for Operable Disease
Although no study has examined use of vismodegib
for operable BCC around eye, a phase 2 multicenter,
open-label trial has studied its use at all skin sites.3
This nonrandomized trial studied 74 volunteers but
was unable to achieve its predefined primary efficacy
end point of histological clearance.3 In addition, of the
37 study patients deemed to have complete clinical
regression (clearance), 12 (32%) had histological evidence of residual BCC.3 Thus, these results may influence the design of future clinical trials studying operable and inoperable tumors.
Conclusions
Vismodegib may be an important treatment option to
patients with locally advanced basal cell carcinoma
(BCC) around the eye. However, the exact role it will
play has not been established. The reviewed literature
suggests that many patients with locally advanced periocular BCC will completely or partially respond when
treated with daily 150 mg vismodegib, but their duration of response is unknown because follow-up times
have been for periods less than 10 months.
Use of vismodegib outside of a clinical trial has
expanded to include the neoadjuvant settings for sur138 Cancer Control
gery and radiotherapy.7,8,10 However, because phase 2
studies of locally advanced and metastatic BCC are not
placebo-controlled trials and follow-up has been limited, it is unclear whether vismodegib prolongs survival
rates or whether it reduces serious rates of morbidity.45
Given that most patients with locally advanced periocular BCC developed the condition over 5 to 15 years
and that clinical follow-up after treatment is brief, judging the role that vismodegib might play in management may be premature.1 When vismodegib is used to
shrink surgical margins, it is uncertain whether these
margins will remain tumor free over time, particularly
after the medication is discontinued. A report from the
US Veterans Affairs concluded in 2013 that the net clinical benefit of vismodegib treatment was minimal and
that its use had a low likelihood of benefit as well as a
low risk of harm.4
Vismodegib is not well tolerated in study patients:
Approximately 40% are unable to continue therapy
due to its adverse events.4 Reports of hepatotoxicity,
particularly in persons taking other medications that
may interact with vismodegib, have also been reported.6,37,38 More information is also needed on the safety
of vismodegib in patients with renal and hepatic impairment. Whether any serious, long-term complications are associated with its use is unknown, and no
evidence exists to suggest that vismodegib improves
rates of survival.
Ever since vismodegib was first approved for clinical use, case reports and small clinical series have appeared in the literature. These studies suffer from
short-term follow-up, lack of control groups, and potentially publication bias, because complications and
therapeutic misadventures tend to be under-reported
after drugs receive market approval.46 Before the therapeutic potential of vismodegib can be realized, larger
studies with appropriate control populations and
long-term follow-up periods are necessary.
References
1. Margo CE, Waltz K. Basal cell carcinoma of the eyelid and periocular
skin. Surv Ophthalmol. 1993;38(2):169-192.
2. Axelson M, Liu K, Jiang X, et al. U.S. Food and Drug Administration
approval: vismodegib for recurrent, locally advanced, or metastatic basal
cell carcinoma. Clin Cancer Res. 2013;19(9):2289-2293.
3. Sofen H, Gross KG, Goldberg LH, et al. A phase II, multicenter,
open-label, 3-cohort trial evaluating the efficacy and safety of vismodegib in
operable basal cell carcinoma. J Am Acad Dermatol. 2015;73(1):99-105.e1.
4. US Department of Veterans Affairs (VA) Pharmacy Benefits Management Services; Medical Advisory Panel; VIAN Pharmacist Executive.
Vismodegib (Erivedge) National Drug Monograph. Washington, DC: US VA;
2013. www.pbm.va.gov/clinicalguidance/drugmonographs/vismodegib_drug
_monograph.docx. Accessed March 22, 2016.
5. Erdem GU, Sendur MA, Ozdemir NY, et al. A comprehensive review
of the role of Hedgehog pathway and vismodegib in the management of
basal cell carcinoma. Curr Med Res Opin. 2015;31(4):743-756.
6. Sekulic A, Migden MR, Oro AE, et al. Efficacy and safety of vismodegib
in advanced basal cell carcinoma. N Engl J Med. 2012;366(23):2171-2179.
7. Gill HS, Moscato EE, Chang AL, et al. Vismodegib for periocular and
orbital basal cell carcinoma. JAMA Ophthalmol. 2013;131(12):1591-1594.
8. Demirci H, Worden F, Nelson CC, et al. Efficacy of vismodegib
(Erivedge) for basal cell carcinoma involving the orbit and periocular area.
Ophthal Plast Reconstr Surg. 2015;31(6):463-466.
April 2016, Vol. 23, No. 2
9. Ally MS, Aasi S, Wysong A, et al. An investigator-initiated openlabel clinical trial of vismodegib as a neoadjuvant to surgery for high risk
basal cell carcinoma. J Am Acad Dermatol. 2014;71(5):904-911.e1.
10. Pollom EL, Bui TT, Chang ALS, et al. Concurrent vismodegib and
radiotherapy for recurrent, advanced basal cell carcinoma. JAMA Dermatol.
2015;151(9):998-1001.
11. Aasi S, Silkiss R, Tang JY, et al. New onset of keratoacanthomas
after vismodegib treatment for locally advanced basal cell carcinomas: a
report of 2 cases. JAMA Dermatol. 2013;149(2):242-243.
12. Papastefanous VP, René C. Secondary resistance to vismodegib
after initial successful treatment of extensive recurrent periocular basal
cell carcinoma with orbital invasion. Ophthal Plast Reconstr Surg. 2015.
Epub ahead of print.
13. Ozgur OK, Yin V, Chou E, et al. Hedgehog pathway inhibition for
locally advanced periocular basal cell carcinoma and basal cell nevus
syndrome. Am J Ophthalmol. 2015;160(2):220-227.e2.
14. Gould SE, Low JA, Marsters JC Jr, et al. Discovery and preclinical
development of vismodegib. Expert Opin Drug Discov. 2014;9(8):969-684.
15. Xin M. Hedgehog inhibitors: a patent review (2013 - present). Expert
Opin Therap Pat. 2015;25(5):549-565.
16. Geeraert P, Williams JS, Brownell I. Targeting the hedgehog pathway
to treat basal cell carcinoma. J Drugs Dermatol. 2013;12(5):519-523.
17. Chang AL, Solomon JA, Hainsworth JD, et al. Expanded access study
of patients with advanced basal cell carcinoma treated with the Hedgehog
pathway inhibitor, vismodegib. J Am Acad Dermatol. 2014;70(1):60-69.
18. Ridky TW, Cotsarelis G. Vismodegib resistance in basal cell carcinoma: not a smooth fit. Cancer Cells. 2015;27(3):315-316.
19.Robarge KD, Brunton SA, Castanedo GM, et al. GDC-0449:
a potent inhibitor of the hedgehog pathway. Bioorg Med Chem Lett.
2009;19(19):5576-5581.
20. Proctor AE, Thompson LA, O’Bryant CL. Vismodegib: an inhibitor
of the hedgehog signaling pathway in the treatment of basal cell carcinoma.
Ann Pharmcother. 2014;48(1):99-106.
21. Graham RA, Lum BL, Morrison G, et al. A single dose mass balance study of the Hedgehog pathway inhibitor vismodegib (GDC-0449)
in humans using accelerator mass spectrometry. Drug Metab Dispos.
2011;39(8):1460-1467.
22. Graham RA, Hop CE, Borin MT, et al. Single and multiple dose intravenous and oral pharmacokinetics of the hedgehog pathway inhibitor vismodegib in healthy female subjects. Br J Clin Pharmacol. 2012;74(5):788-796.
23. Vismodegib [package insert]. South San Francisco, CA: Genentech;
2012.
24.Von Hoff DD, LoRusso PM, Rudin CM, et al. Inhibition of
hedgehog pathway in advanced basal cell carcinoma. N Engl J Med.
2009;361(12);1164-1172.
25. LoRusso PM, Rudin CM, Reddy JC, et al. Phase 1 trial of Hedgehog
pathway inhibitor vismodegib (GDC-0449) in patients with refractory, locally
advanced or metastatic solid tumors. Clin Cancer Res. 2011;17(8):2502-2511.
26. Tang JY, Mackay-Wiggans JM, Aszterbaum M, et al. Inhibiting the
hedgehog pathway in patients with basal cell nevus syndrome. N Engl J
Med. 2012;366(23):2180-2188.
27. Sekulic A, Migden MR, Lewis K, et al; ERIVANCE BCC Investigators.
Pivotal ERIVANCE basal cell carcinoma (BCC) study: 12-month update of
efficacy and safety of vismodegib in advanced BCC. J Am Acad Dermatol.
2015;72(6):1021-1026.
28. Basset-Sequin N, Hauschild A, Grob JJ, et al. Vismodegib in patients
with advanced basal cell carcinoma (STEVIE): a pre-planned interim analysis of an international, open-label trial. Lancet Oncol. 2015;16(6):729-736.
29.Kahana A, Worden FP, Elner VM. Vismodegib as eye-sparing
adjuvant treatment for orbital basal cell carcinoma. JAMA Ophthalmol.
2013;131(10):1364-1366.
30. Peris K, Licitra L, Ascierto PA, et al. Identifying locally advanced
basal cell carcinoma eligible for treatment with vismodegib: an expert panel
consensus. Future Oncology. 2015;11(4):703-712.
31. Edge S, Byrd DR, Compton CC, et al, eds. AJCC Cancer Staging
Manual. 7th ed. New York: Springer; 2010: 523-530.
32. Poulalhon N, Dalle S, Balme B, Thomas L. Fast-growing cutaneous
squamous cell carcinoma in a patient treated with vismodegib. Dermatology.
2015;230(2):101-104.
33. Saintes C, Saint-Jean M, Brocard A, et al. Development of squamous cell carcinoma into basal cell carcinoma under treatment with
vismodegib. J Eur Acad Dermatol Venereol. 2015;29(5):1006-1009.
34. Atwood SX, Li M, Lee A, et al. GLI activation by atypical protein kinase C i/ʎ regulates the growth of basal cell carcinomas. Nature.
2013;28:494(7438):484-488.
35.Sharpe HJ, Pau G, Dijkgraaf GJ, et al. Genomic analysis of
smoothened inhibitor resistance in basal cell carcinoma. Cancer Cell.
2015;9(3):27:327-341.
36. Zhao J, Quan H, Zie C, Lou L. NL-103, a novel dual-targeted inhibitor
of histone deacetylases and hedgehog pathway, effectively overcomes
vismodegib resistance conferred by Smo mutations. Pharmacol Res
Perspect. 2014;2(3):e00043.
37. Ash MM, Jolly PS. Cholestatic hepatic injury associated with
vismodegib, aspirin, naproxen use: a case study and review of vismodegib
April 2016, Vol. 23, No. 2
safety. Int J Dermatol. 2015;54(3):370-374.
38. Ventarola DJ, Silverstein DI. Vismodegib-associated hepatotoxicity: a potential side effect detected in postmarketing surveillance. J Am
Acad Dermatol. 2014;71(2):397-398.
39. Zhang Y, Baicker K, Newhouse JP. Geographic variation in Medicare
drug spending. N Engl J Med. 2010;363(5):405-409.
40. Acosta A, Ciapponi A, Aaserud M, et al. Pharmaceutical policies:
effects of reference pricing, other pricing and purchasing policies.
Cochrane Database Syst Rev. 2014;10:CD005959.
41. Morley M, Finger PT, Perlin M, et al. Cis-platinum for ocular basal
cell carcinoma. Br J Ophthalmol. 1991;75(7):407-410.
42. Luxenberg MN, Guthrie TH Jr. Chemotherapies of eyelid and periorbital tumors. Trans Am Ophthalmol Soc. 1985;83:162-180.
43. Luxenberg MN, Gutherie TH Jr. Chemotherapy of basal cell and squamous cell carcinoma of the eyelids and periorbital tissues. Ophthalmology.
1986;93(4):504-510.
44. Smith CM, Reynard AM, eds. Textbook of Pharmacology. Philadelphia,
PA: WB Saunders; 1992.
45. [No authors listed]. Vismodegib (ERIVEDGE) in basal cell carcinoma:
too many unknowns. Prescrire Int. 2015;24(156):11-14.
46. French DD, Margo CE, Campbell RR. Enhancing post marketing
surveillance: continuing challenges. Br J Clin Pharmacol. 2015;80(4):615-617.
Cancer Control 139
Evidence is insufficient to recommend
chemotherapy, immunotherapy,
or antibiotics as initial treatment for
localized extranodal marginal zone B-cell
lymphoma of the ocular adnexa.
Photo courtesy of Lynn E. Harman, MD. Los Angeles.
Extranodal Marginal Zone B-cell Lymphoma of the
Ocular Adnexa
Jean Guffey Johnson, MD, Lauren A. Terpak, MS, Curtis E. Margo, MD, and Reza Setoodeh, MD
Background: Low-grade B-cell lymphomas located around the eye present unique challenges in diagnosis and
treatment. Extranodal marginal zone B-cell lymphoma is the most common lymphoma of the ocular adnexa
(conjunctiva, orbit, lacrimal gland, and eyelid).
Methods: A systematic search of the relevant literature was performed. Material pertinent to the diagnosis,
prognosis, pathogenesis, and treatment of extranodal marginal zone B-cell lymphoma of the ocular adnexa
was identified, reviewed, and analyzed, focusing on management strategies for primary localized disease.
Results: The primary cause of extranodal marginal zone B-cell lymphoma of the ocular adnexa remains
elusive, although an infectious agent is suspected. Radiotherapy is the most common initial treatment for localized disease. Initial treatment with chemotherapy, immunotherapy, and antibiotics has shown promising
results, but the number of series is limited and controlled trials do not exist.
Conclusions: Although the long-term outcome of localized extranodal marginal zone B-cell lymphoma of
the ocular adnexa is good, optimal treatment remains a goal. The variation in rates of local and systemic relapse among treated stage 1E tumors suggests that critical factors affecting outcomes are not fully understood.
Radiotherapy is the standard of care; at this time, the evidence is insufficient to recommend chemotherapy,
immunotherapy, or antibiotics for initial treatment of extranodal marginal zone B-cell lymphoma localized
to the ocular adnexa. Well-controlled comparative studies are needed.
Introduction
Lymphomas are the most common malignancy of the
From the Department of Pathology (JGJ, RS), James Haley Veterans
Hospital, Departments of Ophthalmology and Pathology and Cell
Biology (CEM), Morsani College of Medicine (LAT), University of
South Florida, Tampa, Florida.
Address correspondence to Curtis E. Margo, MD, Department of
Ophthalmology, MDC Box 21, 12901 Bruce B. Downs Boulevard,
Tampa, FL 33612. E-mail: [email protected]
Submitted July 27, 2015; accepted October 5, 2015.
No significant relationships exist between the authors and the
companies/organizations whose products or services may be referenced in this article.
The authors discuss unlabeled/unapproved uses of doxycycline
and clarithromycin for extranodal marginal zone B-cell lymphoma of the ocular adnexa.
140 Cancer Control
orbit and lacrimal gland, ranking third behind squamous cell carcinoma and melanoma among the malignancies of the conjunctiva.1-3 The eyelid is an uncommon site of primary lymphoma and is more often
secondarily involved when tumors spread from the
conjunctiva or orbit.4,5 The incidence of ocular adnexal
lymphoma has increased in recent decades, primarily
due to an increase in extranodal marginal zone B-cell
lymphoma of the mucosal-associated lymphoid tissue
(MALT) type.1,4,6,7 Most types of lymphoma have been
reported in periocular tissues, of which 95% or more
are B cell in origin; extranodal marginal zone B-cell
lymphoma is the most frequent, making up approximately 70% of cases, followed by follicular lymphoma
and diffuse large B-cell lymphoma.5,8-15 Extranodal
April 2016, Vol. 23, No. 2
marginal zone B-cell lymphoma of the ocular adnexa
shares similar features with extranodal marginal zone
B-cell lymphoma located elsewhere, including general
morphology and immunophenotype, and, presumptively, pathogenesis. Extranodal marginal zone B-cell
lymphoma of the ocular adnexa is a presumed antigendriven neoplasm based on the model of gastric extranodal marginal zone B-cell lymphoma. The putative
antigenic stimulus (or stimuli) is a subject of investigation.
We provide a general overview of extranodal marginal zone B-cell lymphoma of the ocular adnexa, emphasizing key similarities and differences in biological behavior and management with other MALT-type
lymphomas in different locations. Because most cases
of extranodal marginal zone B-cell lymphoma of the
ocular adnexa present as localized disease, this subset
of patients with lymphoma will be the focus of clinical
management in this article. Therapeutic studies were
included if the researchers examined single treatment
protocols for localized extranodal marginal zone B-cell
lymphoma of the ocular adnexa (Ann Arbor stage 1E).
To avoid omitting studies containing valuable clinical
information on the treatment of localized extranodal
marginal zone B-cell lymphoma, inclusion criteria allowed series with up to 15% of cases with disease located at other sites as long as outcomes were not collectively reported.
Diagnosis
The histological features of extranodal marginal zone
B-cell lymphoma are similar to those of MALT-type lymphomas in general. Neoplastic lymphocytes consist of
varying combinations of small cells resembling centrocytes, plasmacytoid lymphocytes, and monocytoid B
cells, with fewer numbers of scattered large cells resembling centroblasts or immunoblasts. This heterogeneous
population of cells is usually found among reactive follicles, some of which may eventually be overrun by lymphoma cells (follicular colonization). Plasma cells are
prominent in some cases, and intranuclear pseudoinclusions (Dutcher bodies) are sometimes noted. Characteristic infiltration of the epithelium by neoplastic lymphocytes (lymphoepithelial lesion) is uncommon, and,
when observed, it is found in the lacrimal gland and
conjunctiva but not the orbit.16 However, collections of
atypical lymphocytes in conjunctival epithelium and lacrimal ducts similar to lymphoepithelial lesions are not
specific for extranodal marginal zone B-cell lymphoma.
They have also been described in reactive processes of
the conjunctiva and lacrimal gland.16 Monocytoid cells
with abundant pale cytoplasm are observed less often in
extranodal marginal zone B-cell lymphoma of the ocular adnexa than other sites.11 Transformation to diffuse
large B-cell lymphoma should be considered when solid or sheet-like proliferations of large cells are encounApril 2016, Vol. 23, No. 2
tered. These larger cells are usually positive for B-cell
chronic lymphocytic leukemia/lymphoma (BCL) 6 and
sometimes cluster designation (CD) 10.17
Given the morphological overlap between reactive
lymphoid hyperplasia and extranodal marginal zone
B-cell lymphoma, ancillary tests are often used to enhance diagnostic certainty. Extranodal marginal zone
B-cell lymphomas are positive for CD20, BCL2, paired
box 5 (PAX5), and CD79A but typically do not express
CD5, CD10, or CD23.16 Fewer than 5% of cases express
CD5.11-14,16 In the setting of CD5 positivity, the differential diagnosis might include mantle cell lymphoma
(positive for cyclin D1, t[11;14] present) and small lymphocytic lymphoma (positive for CD23).14 CD43 expression is less common in extranodal marginal zone
B-cell lymphoma of the ocular adnexa compared with
the salivary glands (12%–25% vs 70%).11,18 Lymphoma
cells typically express immunoglobulin (Ig) M and are
IgD negative; on average, the proliferation index is 15%
by Ki-67 immunostaining.19
The immunohistochemical demonstration of
light-chain restriction using formalin-fixed, paraffinembedded tissue is challenging because detection of
cytoplasmic light chains in nonplasmacytic proliferations is prone to false-negative testing.20-22 An alternative approach to demonstrating light-chain restriction
is flow cytometry, which requires fresh tissue.23,24 Flow
cytometry may also be useful in confirming immunophenotype, particularly when the size of a specimen is
limited.23,24 Recurrent structural genetic abnormalities
have been identified in MALT-type lymphomas, and the
frequency of these alterations vary, depending on anatomical site.25 Although it is not specific, trisomy of chromosome 3 is the most commonly reported cytogenetic
finding among MALT-type lymphomas.25 Among the
common sites of MALT lymphomas, t(14;18)(q32;q21) involving IgH and MALT1 is the most frequently seen in
the ocular adnexal MALTomas.26 This translocation has
been reported in about 25% of cases in some series.26
Clonality of B cells can be documented by polymerase
chain reaction (PCR) of Ig heavy chain.27,28
More centers are employing combinations of immunohistochemistry, flow cytometry, and PCR to diagnosis low-grade lymphoma and relying less on morphology alone.29
Clinical Staging
The approach to staging ocular adnexal lymphoma is
similar to that for lymphoma in general, and typically
includes thorough clinical and laboratory examinations with bone marrow biopsy.30 Although computed tomography (CT) or magnetic resonance imaging
(MRI) with contrast is valuable to determining the extent of local disease of the orbit, eyelid, and paranasal
sinuses, positron emission tomography (PET) may be
superior for the initial staging of ocular adnexal lymCancer Control 141
phoma.31 When compared with CT, use of
PET has upstaged a majority of patients
with ocular adnexal lymphoma.32,33
Evidence suggests that subclinical involvement of the eye can occur in persons
with periocular lymphoma.34,35 In a study
of ocular adnexal lymphomas, 25 study
patients with bilateral involvement had
uveal thickening with ocular B-scan ultrasonography.34 (Intraocular disease might
have been missed if staging were based
on CT alone.34) Although uveal involvement with extranodal marginal zone B-cell
lymphoma has been previously reported,
few studies have documented how often it
goes unrecognized when patients present
with conjunctival and orbital disease.34,35
How uveal involvement might affect prognosis is also unclear.
Roughly two-thirds of ocular adnexal
lymphomas are stage 1E (localized extranodal tumors) using the Ann Arbor system.8,36,37 This disproportionate accumulation of cases in a single stage limits the
discriminatory potential to forecast outcomes. Other shortcomings exist to the Ann
Arbor system for ocular adnexal lymphoma,
including the inability to take into account
multicentric and bilateral tumors, extent of
localized tissue involvement, and precise anatomical location around the eye.38
Application of the tumor, node, metastasis (TNM)–based system incorporating
information on extent of disease within
anatomical compartments of the ocular adnexa, bilateral nature, and information on
regional lymph-node involvement may better predict outcome (Table 1).39-42 An initial
study showed that tumor stage may more
thoroughly describe the extent of disease,
although it does not yet predict rate of relapse or survival.41 Stages N1 to N4 and M1
are associated with worse rates of survival.41 In a multicenter study using the TNM
system, any prognostic utility ascribed to
size and location of ocular adnexal lymphoma was eclipsed by histological classification and type of treatment.42 The capability of the TNM system to predict worse
outcomes appears to reside in its ability to
identify bilateral disease and to delineate
nodal and metastatic involvement at the
time of presentation.39,40
The full potential of the TNM system
may not be realized until it is modified to
include additional biomarkers and when
142 Cancer Control
Table 1. — TNM Staging for Lymphoma of the Ocular Adnexa
Clinical
Stage
TX
Primary Tumor
Pathological
Stage
Lymphoma extent not specified
TX
T0
No evidence of lymphoma
T0
T1
Lymphoma involving the conjunctiva alone without
orbital involvement
T1
T1a
Bulbar conjunctiva only
T1a
T1b
Palpebral conjunctiva ± fornix ± caruncle
T1b
T1c
Extensive conjunctival involvement, both bulbar and
nonbulbar
T1c
T2
Lymphoma with orbital involvement ± any conjunctival involvement
T2
T2a
Anterior orbital involvement (± conjunctival involvement) but no lacrimal gland involvement
T2a
T2b
Anterior orbital involvement (± conjunctival involvement) plus lacrimal involvement
T2b
T2c
Posterior orbital involvement (± conjunctival
involvement ± anterior orbit involvement and ± any
extraocular muscle involvement)
T2c
T2d
Nasolacrimal drainage system involvement
(± conjunctival involvement but not including
nasopharynx)
T2d
T3
Lymphoma with preseptal eyelid involvement ±
orbital involvement ± any conjunctival involvement
T3
T4
Orbital adnexal lymphoma extending beyond orbit to
adjacent structures such as bone and brain
T4
T4a
Involvement of nasopharynx
T4a
T4b
Osseous involvement (including periosteum)
T4b
T4c
Involvement of maxillofacial, ethmoidal, and/or
frontal sinuses
T4c
T4d
Intracranial spread
T4d
Regional Lymph Nodes
NX
Regional lymph nodes cannot be assessed
NX
N0
No evidence of lymph node involvement
N0
N1
Involvement of ipsilateral regional lymph nodes
(preauricular, parotid, submandibular, and cervical)
N1
N2
Involvement of contralateral or bilateral regional
lymph nodes
N2
N3
Involvement of peripheral lymph nodes not draining;
ocular adnexal regional
N3
N4
Involvement of central lymph nodes
N4
M0
No evidence of involvement of other extranodal
sites (no pathologic M0; use clinical M to complete
stage group)
M0
M1a
Noncontiguous involvement of tissues or
organs external to the ocular adnexa (eg, parotid
glands, submandibular gland, lung, liver, spleen,
kidney, breast)
M1a
M1b
Lymphoma involvement of the bone marrow
M1b
M1c
Both M1a and M1b
M1c
Distant Metastasis
TNM = tumor, node, metastasis.
Adapted from Edge S, Byrd DR, Compton CC, et al, eds. AJCC Cancer Staging Manual.
7th ed. New York: Springer; 2010. Republished with permission of Springer; permission conveyed through Copyright Clearance Center, Inc.
April 2016, Vol. 23, No. 2
new imaging technologies can be used to help provide superior metrics in terms of tumor location and
volume.
Pathogenesis
Infectious agents have been suspected in the etiology
of extranodal marginal zone B-cell lymphoma of the
ocular adnexa based on the causative role that Helicobacter pylori infection plays in gastric MALT-type lymphoma.43 The progression of H pylori–induced chronic
gastritis to extranodal marginal zone B-cell lymphoma
is comprehensible both in terms of light microscopy
and molecular changes.43 Unlike malignant transformation of lymphocytes through the direct infection of
lymphotropic viruses like Epstein–Barr virus or human
T-cell leukemia virus type 1, H pylori indirectly acts by
inducing chronic inflammation. This antigen-driven
process, coupled with other oncogenic events, can lead
to the emergence of antigen-independent lymphocyte
proliferation.
Chlamydia psittaci
The search for putative infectious agents for periocular
lymphoma has resulted in conflicting results.44-47 Using
PCR, an Italian study found that 80% of patients with
extranodal marginal zone B-cell lymphoma of the ocular adnexa were exposed to Chlamydia psittaci.46 However, similar investigations from other regions of the
world could not duplicate these results.44,47 In a review
of 11 international studies, the overall rate of Chlamydia positivity in extranodal marginal zone B-cell
lymphoma of the ocular adnexa was 23%, with the vast
majority of all positive cases (90%) occurring in 3 countries.47 In a survey of 423 cases of extranodal marginal
zone B-cell lymphoma of the ocular adnexa, the detection rate of Chlamydia ranged from 0% to 87%, with
seemingly inconsistent geographic variation.44 To control for spurious laboratory results, investigators used a
single standardized procedure for Chlamydia DNA detection among 142 cases of extranodal marginal zone
B-cell lymphoma of the ocular adnexa.45 The results
confirmed a variation in prevalence (eg, 11% in southern China, 47% in Germany); the overall prevalence
of C psittaci DNA was 22%, which was more than
twice the control group (10%; P = .04).45 However,
based on historical prevalence, this result was not
much greater than that established for non-neoplastic
disorders of the orbit.44
Helicobacter pylori
The causal association between H pylori infection
and gastric extranodal marginal zone B-cell lymphoma is well established, so investigators have begun
searching for a link between the bacterium and extranodal marginal zone B-cell lymphoma of the ocular
adnexa.48,49 Chan et al48 described H pylori DNA in 4 of
April 2016, Vol. 23, No. 2
5 cases of conjunctival extranodal marginal zone B-cell
lymphomas using PCR amplification and Southern
blot hybridization. The DNA of H pylori was not found
in normal conjunctiva from the same study patients.48
The same group examined 8 study patients with orbital extranodal marginal zone B-cell lymphoma and
found 1 study patient with the genomic fingerprints of
H pylori.49 C pneumoniae (but not C psittaci or C trachomatis) was identified in another.49
Lee et al 5 0 identi f ied DNA of H pylor i i n
15 of 15 cases of extranodal marginal zone B-cell
lymphoma of the conjunctiva that they examined; no
such discovery was made in the control group (n = 8).
These findings contrast with another study involving
13 cases of extranodal marginal zone B-cell lymphoma of the conjunctiva in which H pylori DNA could not
be detected in any tumor (either using immunohistochemistry or PCR).51
The prevalence of gastric H pylori infection at the
time of initial diagnosis of extranodal marginal zone
B-cell lymphoma varies between where the primary
site of lymphoma is located.52 For example, the prevalence rate of infection was found to be 45% (37 of
83 cases) in extranodal marginal zone B-cell lymphoma of the ocular adnexa compared with 25% (25 of
101 cases) in extranodal marginal zone B-cell lymphoma located elsewhere (but not the stomach).52 The rate
of prevalence was 12% (18 of 156) among control cases
without lymphoma.52 These findings led investigators
to propose another pathogenic mechanism involving
the attraction of circulating lymphomatous cells to the
ophthalmic mucosa: Once around the eye, the lymphocytes would then be transformed under the influence
of additional mitogenic stimuli.52 This study and the
proposed hypothesis has generated need for further
investigation.52,53
Other Infectious Agents
An international study investigating the presence of
viral DNA for herpes simplex virus types 1 and 2 and
adenovirus types 8 and 19 in persons with extranodal
marginal zone B-cell lymphoma of the ocular adnexa
found no association.45 Although the conceptual model
of an infectious etiology is appealing, consistent and
reproducible evidence supporting any specific pathogen has been elusive.54,55
Autoimmunity
The possibility of an autoantigenic stimulus is supported by the association of extranodal marginal zone
B-cell lymphoma of the ocular adnexa with various autoimmune disorders (eg, Sjögren disease, Hashimoto
disease, IgG4-related disorder).56 The connection between Sjögren disease and lymphoma has been known
since the 1970s.57 Although Sjögren disease conveys a
14-fold greater risk of B-cell lymphoma, the cause for
Cancer Control 143
this susceptibility is unclear.57 Several cases of extranodal marginal zone B-cell lymphoma of the ocular adnexa have been reported in persons with IgG4-related
disease — an association that provides little additional
insight into pathogenesis.58
Treatment
A comprehensive review of treatment options for extranodal marginal zone B-cell lymphoma of the ocular adnexa is beyond the scope of this review. Most
therapeutic studies have consisted of retrospective
case series.30,58-79 Comparing clinical outcomes in these
nonrandomized trials is perilous for several reasons.
Clinical series of ocular adnexal lymphoma published
prior to 2000 often lumped extranodal marginal zone
B-cell lymphoma together with other low-grade lymphomas and atypical lymphoid hyperplasia.5 Even
large studies involving 2 or more treatment arms had
limited statistical power to exclude small but meaningful differences in outcome.5,8,15,30 The success of
local therapy is often confounded by supplemental
treatment given to partial responders because clinical
outcomes are collectively reported.5,8,15,30 In addition,
short-term studies (< 5 years) may also not adequately
reflect the biological potential of the disease, particularly in terms of rates of local and systemic recurrence,
and tumor-related mortality.
Radiotherapy
Typically, radiotherapy results in a high rate of local
control that ranges from 85% to 100%, even in the
presence of systemic disease.59,60,62,64,73,75,76,79-83 Since
2000, a total of 9 studies have examined the outcome of radiotherapy for localized extranodal marginal zone B-cell lymphoma of the ocular adnexa
(Table 2).64,70,73,79-84 Some included small numbers
of study patients with systemic disease, and clinical outcomes were measured in ways that prohibit uniform summarization. The majority of the
503 study patients with stage 1E disease achieved
local control.64,70,73,79-84 When reported, complete response (CR) rates ranged from 52% to 93%, whereas
the proportion of 5-year, systemic-free relapse rates
usually exceeded 90%.64,70,73,79-84 Overall, systemic
relapses occurred in 31 study patients (6.2%), ranging from a low rate of 2.2% (median follow-up of
32 months) to a high rate of 16.8% (median follow-up
of 5.9 years).64,70,73,79-84
However, these results must be interpreted with
caution because the methods and rigor used to detect
recurrent disease differed and the protocols employed
to treat partial response and recurrence varied. The
different types of second-line therapies for incomplete
localized responses confound the direct comparison of
clinical series, particularly in terms of rates of systemic
recurrence and tumor-related deaths.
144 Cancer Control
Chemotherapy
Use of chemotherapy for local ocular adnexal disease
has been studied in 2 centers (n = 54); of the patients
studied, 52 had stage 1E disease (Table 3).61,67-69 Among
21 study patients (19 [90%] with stage 1E) treated with
cyclophosphamide, vincristine, and prednisolone, CR
was reported in 16 (76%), local relapse occurred in 5,
and systemic relapse occurred in 2; no deaths were reported during a median follow-up time of 55 months.68
The other trial studied chlorambucil in 33 participants
with stage 1E extranodal marginal zone B-cell lymphoma.69 During a median follow-up of 26 months, CR was
reported in 26, local recurrence in 1, and systemic relapse in 3; 1 tumor-related death was noted.69
Proponents of initial chemotherapy state that this
treatment option eliminates local complications from
radiotherapy, such as dry eyes, cataracts, and skin irritation, and is associated with fewer serious complications.68 Direct comparison with radiotherapy for stage
1E disease is methodologically problematic, because
some reports include small proportions of study patients with more advanced disease.68 The collective
rate of systemic relapse for radiotherapy is 6.2%, which
is less than that currently reported for chemotherapy
(9.3%; see Tables 2 and 3).61,64,69,70,73,79-84
Immunotherapy
Rituximab, a chimeric mouse antihuman CD20 monoclonal antibody, has been used as treatment for lymphoma localized to the ocular adnexa.74 Two reports
involving 12 study patients with localized extranodal marginal zone B-cell lymphoma have been published (see Table 3).61,67-69 In one study, 11 eyes of
10 study patients with ocular adnexal lymphoma were
treated with systemic rituximab.61 CR was reported in
5 (56%; median follow-up of 31 months) without systemic or ocular adverse events, and no tumor-related
deaths were reported.61 The remaining study patients
required radiotherapy.61 Another report involved
5 study patients with extranodal marginal zone B-cell
lymphoma of the ocular adnexa, 3 of whom had stage
1E disease.67 Four of the 5 study patients who initially
responded to treatment locally relapsed.67
To date, rituximab has been reported in relatively
few patients with extranodal marginal zone B-cell lymphoma localized to the ocular adnexa, with seemingly
high rates of inadequate periocular response and local
recurrence (see Table 3).61,67-69 The median follow-up
times (< 31 months) are too short to assess the role of
rituximab as therapy for local disease at this time.61,67-69
Antibiotic Therapy
Because some cases of extranodal marginal zone B-cell
lymphoma of the ocular adnexa have been associated
with C psittaci infection, treatment with doxycycline
has been attempted.47 A meta-analysis of 4 treatment
April 2016, Vol. 23, No. 2
Table 2. — Radiotherapy for Localized Extranodal Marginal Zone B-Cell Lymphoma of the Ocular Adnexaa
Study
Location
Stage
Cases of
Systemic
Relapse
Local
Relapseb
Goda80
89
Conjunctiva: 59
Lacrimal gland: 20
Orbit: 10
1E: 89
Hashimoto70
78
Conjunctiva: 37
Orbit: 29
Lacrimal gland: 12
1E: 78
Le84
31
Conjunctiva: 21
Orbit: 10
1E: 31
Bilateral: 4
None
Nam73
66
Conjunctiva: 29
Orbit: 20
Lacrimal gland: 7
Eyelid: 10
1E: 66
Bilateral: 14
Son82
46
Conjunctiva: 37
Eyelid: 3
Orbit: 6
Suh83
48
Tran81
15
Other
Outcome
Measure
No. of
TumorRelated
Deaths
7-y relapse-free
survival: 64%
3
66 mo
5-y PFS: 86%
10-y OS: 95.3%
None
5
5.9 y
(range,
0.75–20.3)
100% local
control
1
Conjunctiva: 2
Orbit: 0
Lacrimal
gland: 2
Eyelid: 2
3
50 mo
(range,
12–114)
5-y systemic
relapse-free
survival: 95.4%
None
1E: 46
Bilateral: 3
1
1
32 mo
(range,
3–114)
CR with 5-y
relapse-free
survival: 93%
None
“Orbit”
1E: 46
Bilateral: 4
IV: 2
3
None
39–72 mo
10-y relapsefree survival:
93%
1c
24
Conjunctiva: 16
Orbit: 3
Lacrimal gland: 7
Eyelid: 1
1E: 22
Bilateral: 3
IV: 2
2
1
41 mo
(range,
5–137)
PFS: 90%
5-y OS: 100%d
1 case of
high-grade
lymphoma
Uno64
50
Conjunctiva: 29
Orbit: 17
Eyelid: 2
Lacrimal gland: 2
1E: 50
Bilateral: 7
3
3
46 mo
(range,
10–140)
CR: 26
PR: 20
No response: 4
1
Woolf 79
71 + 10 lowgrade lymphomas
Orbit: 56
Conjunctiva: 14
Lacrimal gland: 11
Eyelid: 4
1E: 81
Bilateral: 4
None
Distant: 3
Contralateral
lacrimal gland
(local): 1
4.4 y
(range,
0.2–10.4)
100% local
controle
None
503
7
Median
Follow-Up
5.9 y
Totalf
a
No. of
Study
Patients
499
6 (1.2%) g
Includes series since 2000 with 85% or more localized cases of stage 1E disease.
bLocal relapse defined as any portion of ocular adnexa, including the contralateral side, of initial
cTumor death occurred in 1 study patient with stage IV disease.
dHigh-grade lymphoma death occurred at 71 mo.
e Of the 81 original study patients, 71 (88%) had extranodal marginal zone B-cell lymphoma.
f Data not sufficiently detailed to provide totals for local and systemic relapses.
gPercentage based on study patients with stage 1E disease.
unilateral disease.
CR = complete response, OS = overall survival, PFS = progression-free survival, PR = partial response.
trials taking place prior to 2006 totaling 42 study patients found that 20 responded to oral antibiotics,
20 remained stable, and 2 progressed.47 Although CR
was reported in 8 study patients, objective responses with radiography or slit lamp photography were
available in 3.47
Since 2006, researchers have initiated 3 trials to
study the effect of doxycycline on the management
April 2016, Vol. 23, No. 2
of predominantly primary extranodal marginal zone
B-cell lymphoma localized to the ocular adnexa in
151 study patients (Table 4).76,78,85-87 Because the rate
of tumor response (ie, reduction in size) differs following treatment with doxycycline and radiotherapy
or chemotherapy, it is difficult to directly measure and
compare clinical outcomes.78,85,87 One study reported a
2-year failure-free survival rate of 67%,85 whereas anCancer Control 145
Table 3. — Chemotherapy or Immunotherapy for Localized Extranodal Marginal Zone Lymphoma of the Ocular Adnexaa
Study
Treatment
No. of
Study
Patients
Ben
Simon69
Chlorambucil
33
Orbit: 10
Lacrimal gland: 8
Conjunctiva: 7
Eyelid: 6
Combination: 2
Song68
Cyclophosphamide
Vincristine
Prednisolone
21
Conjunctiva: 6
Orbit: 8
Eyelid: 5
Lacrimal gland: 2
Adnexa (general): 2
Ferreri67
Rituximab
5
Conjunctiva: 2
Orbital: 1
Tuncer 61
Rituximab
9
Conjunctiva: 5
Bilateral: 1
Orbit: 4
Cases
of Local
Relapseb
Cases of
Systemic
Relapse
1E: All
1
3
26 (range,
8–62)
CR: 26
1
1E: 19
Bilateral: 2
IIE: 2
5
2
58 (range,
6–163)
CR: 16
PR: 5
None
1E: 3
IV: 2
4
1
23 (range,
2–4)
1E: All
6
None
31 (range,
10–61)
Location
Stage
Median
FollowUp, mo
Other
Outcome
Measures
No. of
TumorRelated
Deaths
Chemotherapy
Immunotherapy
aIncludes series with ≤ 2 study patients with disease beyond the orbit.
bLocal relapse defined as any portion of ocular adnexa, including the contralateral
CR = complete response, PR = partial response.
None
CR: 5
PR: 4
None
side, of initial unilateral disease.
Table 4. — Antibiotic Therapy or Observation for Extranodal Marginal Zone Lymphoma of the Ocular Adnexaa
Study
Treatment
No. of
Study
Patients
Ferreri85
Doxycycline
27
Conjunctiva: 14
Orbit: 13
1E: 24
IIE: 3
Bilateral: 5
Unable to determine
local from systemic
relapses
Ferreri87
Doxycycline
34
Conjunctiva: 23
Orbit: 14
Lacrimal gland: 5
Conjunctiva/orbit: 5
1E: All
14 failures
Unable to delineate
local from systemic
Han78
Doxycycline
90
Conjunctiva: 74
Orbit: 12
Eyelid: 3
Lacrimal gland: 2
T1N0M0: 62
T2N0–2 M0: 22
T3N0–2 M0: 5
T4N0M0: 1
Matsuo
None other
than biopsy
8
Tanimoto86
None other
than biopsyc
36
Location
Stage
Cases of
Cases
Median
of Local Systemic Follow-Up
Relapseb Relapse
Other
Outcome
Measures
No. of
TumorRelated
Deaths
21 mo
(range,
4–204)
Overall
response: 64%
2-y failure-free
survival: 67%
None
37 mo
(range,
15–62)
5-y PFS: 55%
None
40.5 mo
(range,
8–85)
5-y PFS: 61%
None
5.4 y
(range,
1–11)
7 spontaneously
regressed
None
10.5 y
(range,
0.7–16.7)
OS:
5-y: 94%
10-y: 94%
15-y: 71%
Antibiotic Therapy
31
6
Observation Without Treatment
76
c
Conjunctiva: 8
1E: All
Conjunctiva: 15
Orbit: 19
Lacrimal gland: 2
1E: 36
Bilateral: 5
None
None
15
2
aIncludes series with ≥ 8 5% localized stage 1E extranodal marginal zone B-cell lymphoma since 2000.
bLocal relapse defined as any portion of the ocular adnexa, including the contralateral side, of initial unilateral
c Eight of 13 study patients who declined treatment and are the focus of this review.
2
disease.
OS = overall survival, PFS = progression-free survival.
146 Cancer Control
April 2016, Vol. 23, No. 2
other series reported that the failure-free survival rate
after 5 years was 55%.87 Progressive disease was treated with a variety of traditional protocols, and no tumor-related deaths were reported (see Table 4).76,78,85-87
In that study, patients were excluded from the trial if
they had large tumors or tumors demonstrating rapid
growth.87 Because criteria describing large tumor size
and rapid growth were not provided, an exclusion bias
of unknown clinical importance complicates any interpretation of these results.87
Long-term follow-up in 1 study of 90 patients
initially treated with doxycycline had mixed results.78 Patients enrolled in the clinical trial received
twice-daily oral 100 mg doxycycline for either 3 or
6 weeks and were followed for a mean time of 40.5
months.78 A total of 61% had no progression after 5
years, and 34 study patients who failed doxycycline therapy were successfully managed with chemotherapy, radiotherapy, or both.78 No tumor-related deaths were reported; systemic relapse was
reported in 6 study patients.78 Pretreatment exposure to
C psittaci was not determined.
“Failed to progress” reports are difficult to compare to studies whose benchmark outcomes are either
complete or partial regression, and this is particularly
true for cases of lymphoma with an indolent course.
Lacking or incomplete knowledge of a history of chlamydial infection among patients enrolled in antibiotic
studies further complicates interpretation, because the
global variation in C psittaci infection rate should affect
the response rate to doxycycline. Future trials of antibiotic treatment may be more efficient if their enrollment
is appropriately limited to patients with documented
exposure to C psittaci.
A 6-month trial of a second-line therapeutic agent,
clarithromycin, was reported in a trial of 7 study volunteers who had extranodal marginal zone B-cell lymphoma that failed to respond to doxycycline.88 Partial
response was reported in 4 study patients and stable
disease for varying periods of time was reported in 3;
however, all study patients experienced disease progression (average time to progression, 16 months).88
Observation
Given the indolent nature of extranodal marginal zone
B-cell lymphoma of the ocular adnexa, some investigators have wondered whether observation may be a
viable option, particularly in persons of advanced age
or with other comorbidities that may limit survival.76,86
Two groups reported on the long-term follow-up data
of 44 study patients with localized disease (5 bilateral); the median follow-up periods were 5.4 years76 and
10.5 years.86 Twenty-five (57%) study patients did not
require treatment throughout the course of the study,
and 2 succumbed to progressive lymphoma.76,86 A
total of 17 study patients (39%) progressed; transformaApril 2016, Vol. 23, No. 2
tion to high-grade lymphoma occurred in 1 case.76,86
The majority of tumors arose in conjunctiva (23 [52%]),
7 of which spontaneously regressed.76,86 Therefore, the
study authors concluded that watchful waiting might
be an acceptable option in select patients with unilateral extranodal marginal zone B-cell lymphoma of the
ocular adnexa.86
Assessment of Clinical Outcome
Comparing the treatment of primary lymphoma localized to the ocular adnexa is difficult because some series combine different histological types of lymphoma,
some researchers do not separately analyze for anatomical subgroups (eg, conjunctiva, orbit), and some
report clinical outcomes differently. In addition, not
all studies employ standardized intervals for followup or methods for monitoring outcome (eg, CT, MRI,
PET/CT, PET). Few provide information on primary tumor size, and most employ different salvage therapies
for partial responders.
Tumor size is an important parameter to monitor because the rapidity and completeness with
which a tumor shrinks may correlate with durability of response.89 However, the speed at which tumor
size is reduced may be of less importance in cases of
low-grade lymphoma compared with other neoplasms,
and it may be of less value in terms of assessing antibiotic effectiveness. Use of tumor diameter — not volume — as an outcome variable for extranodal marginal
zone B-cell lymphoma of the ocular adnexa reflects
how difficult it is to obtain reliable volumetric data on
periocular tumors in general.
Measuring the volume or planar dimensions of
periocular tumors presents unique challenges. For example, oftentimes, lymphomas of the ocular adnexa
are irregularly shaped and conform to the contours of
the eye and orbital walls. It might be valuable to apply the Response Evaluation Criteria in Solid Tumors
(RECIST) and RECIST 1.1 to these lesions, but these
criteria have not been independently validated in the
context of low-grade lymphoma of the ocular adnexa.89
One of the few studies that addressed the role of
quantitative, monitored outcomes in the setting of extranodal marginal zone B-cell lymphoma of the ocular adnexa following radiotherapy was performed by
Jung et al.90 These researchers found that the maximum tumor diameter decreased by 50% of its initial
size after 4.7 months of treatment.90 The authors emphasize that partial and total responses are usually
poorly defined in most clinical series — both in terms
of tumor-specific size reductions and in follow-up intervals after treatment.90
Conclusions
Extranodal marginal zone B-cell lymphoma of the ocular adnexa can arise in the setting of reactive lymphoid
Cancer Control 147
hyperplasia, but the underlying cause of this presumed
antigen-driven process is unclear. Given the epidemiological results to date, a more complex interplay of putative infectious agents and immune response is possible.
In terms of treatment options for primary localized extranodal marginal zone B-cell lymphoma of the ocular
adnexa, radiotherapy provides good local control and is
associated with relatively low systemic spread. Studies
assessing the effectiveness of other initial treatment options for localized disease (eg, chemotherapy, immunotherapy, antibiotics) are limited by several weaknesses,
including small study size, enrollment biases, and imbalanced mixtures of anatomical locations (ie, lacrimal
gland, conjunctiva, eyelid, orbit). The evidence is insufficient at this time to recommend an alternative to radiotherapy for primary localized extranodal marginal
zone B-cell lymphoma of the ocular adnexa.
As more optimal therapies are sought and randomized clinical trials remain unlikely, future investigations might consider use of methodologies to
volumetrically measure tumor response, to establish standardized protocols for initial evaluation and
follow-up, and to use equivalent rescue protocols for
partial responses so that different clinical series can be
compared against one another. References
1. Margo CE, Mulla ZD. Malignant tumors of the orbit: analysis of the
Florida Cancer Registry. Ophthalmology. 1998;105(1):185-190.
2. Bonavolontà G, Strianese D, Grassi P, et al. An analysis of 2,480
space-occupying lesions of the orbit from 1976 to 2011. Ophthal Plast
Reconstr Surg. 2013;29(7):79-86.
3. Shields JA, Shields CL, Scartozzi R. Survey of 1264 patients with
orbital tumors and simulating lesions: the 2002 Montgomery Lecture, part
1. Ophthalmology. 2004;111(5):997-1008.
4. Margo CE, Mulla ZD. Malignant tumors of the eyelid: a populationbased study of non-basal cell and non-squamous cell malignant neoplasms. Arch Ophthalmol. 1998;116(2):195-198.
5. Knowles DM, Jakobiec JA. Malignant lymphomas and lymphoid
hyperplasias that occur in the ocular adnexa (orbit, conjunctiva, and eyelids). In: Knowles DM, ed. Neoplastic Hematopathology. Baltimore, MD:
Williams & Wilkins; 1992: 1009-1046.
6.Moslehi, R, Devesa SS, Schairer C, et al. Rapidly increasing incidence of ocular non-Hodgkin lymphoma. J Natl Cancer Inst.
2006;98(13):936-939.
7. Sjö LD, Ralfkiaer E, Prause JU, et al. Increasing incidence of ophthalmic lymphoma in Denmark from 1980 to 2005. Invest Ophthal Vis Sci.
2008;49(8):3283-3288.
8. Jenkins C, Rose GE, Bunce C, et al. Histological features of ocular
adnexal lymphoma (REAL classification) and their association with patient
morbidity and survival. Br J Ophthamol. 2000;84(8):907-913.
9. Meunier J, Lumbroso-le Rouic L, Vincent-Salomon A, et al. Ophthalmologic and intraocular non-Hodgkin’s lymphoma: a large single center study of initial characteristics, natural history and prognostic factors.
Hematol Oncol. 2004;22(4):143-158.
10. Sullivan TJ, Whitehead K, Williamson R, et al. Lymphoproliferative
disease of the ocular adnexa: a clinical and pathologic study with statistical
analysis of 69 patients. Ophthalmic Plast Reconstr Surg. 2005;21(3):177-188.
11. Ferry JA, Fung CY, Zukerberg L, et al. Lymphoma of the ocular adnexa: a study of 353 cases. Am J Surg Pathol. 2007;31(2):170-184.
12. Cho EY, Han JJ, Ree JH, et al. Clinicopathologic analysis of ocular
adnexal lymphomas: extranodal marginal zone B-cell lymphoma constitutes the vast majority of ocular lymphomas among Koreans and affects
younger patients. Am J Hematol. 2003;73(2):87-96.
13. Coupland SE, Hellmich M, Auw-Haedrich C, et al. Prognostic value
of cell-cycle markers in ocular adnexal lymphomas: an assessment of 230
cases. Graefes Arch Clin Exp Ophthalmol. 2004;42(2):130-143.
14. Stefanovic A, Lossos IS. Extranodal marginal zone lymphoma of
the ocular adnexa. Blood. 2009;114(3):505-510.
15. McKelvic PA, McNab A, Francis IC, et al. Ocular adnexal lym148 Cancer Control
phoproliferative disease: a series of 73 cases. Clin Exp Ophthalmol.
2001:29(6):387-393.
16. Lagoo AS, Haggerty C, Kim Y, et al. Morphologic features of 115
lymphomas of the orbit and ocular adnexa categorized according to the
World Health Organization classification: are marginal zone lymphomas in
the orbit mucosa-associated lymphoid type lymphomas? Arch Pathol Lab
Med. 2008;132(9):1405-1416.
17. Harris NL, Jaffe ES, Diebold J, et al. The World Health Organization classification of hematological and lymphoid neoplasms: report of the
Clinical Advisory Committee meeting, Airlie House, Virginia, Winter 1997.
J Clin Oncol. 1999;17(12):3835-3849.
18. Ruiz A, Reischl U, Swerdlow SH, et al. Extranodal marginal zone Bcell lymphomas of the ocular adnexa: multiparameter analysis of 34 cases
including interphase molecular cytogenetics and PCR for Chlamydia psittaci. Am J Surg Path. 2007;31(5):792-802.
19. Isaacson PG, Spencer J. Malignant lymphoma of mucosa‐associated lymphoid tissue. Histopathology. 1987;11(5):445-462.
20. Marshall-Taylor CE, Cartun RW, Mandich D, et al. Immunohistochemical detection of immunoglobulin light chain expression in B-cell nonHodgkin’s lymphoma using formalin-fixed paraffin-embedded tissues and
a heat-induced epitope retrieval technique. Apply Immunohistochem Mol
Morphol. 2002;10(3):258-262.
21. Evans HL. Extranodal small lymphocytic proliferations: a clinicopathologic and immunocytochemical study. Cancer. 1982;49(1):84-96.
22. Ashton-Key M, Diss TC, Isaacson PG, et al. A comparative study of
the value of immunohistochemistry and the polymerase chain reaction in
the diagnosis of follicular lymphoma. Histopathology. 1995;27(6):501-508.
23. Barrena S, Almeida J, Del Carmen Garcia-Macias M, et al. Flow cytometry immunophenotyping of fine-needle aspiration specimens: utility in
the diagnosis of classification of non-Hodgkin lymphomas. Histopathology.
2011;58(5):906-918.
24. Ensani F, Mehravaran S, Irvanlou G, et al. Fine-needle aspiration
cytology and flow cytometic immunophenotying of diagnosis and classification of non-Hodgkin lymphoma in comparison to histopathology. Diagn
Cytopathol. 2012;40(4):305-310.
25. Troppan K, Wenzl K, Neumeister P, et al. Molecular pathogenesis of
MALT lymphoma. Gastroenterol Res Pract. 2015;2015:102656.
26. Streubel B, Simonitsch-Klupp I, Müllauer L, et al. Variable frequencies of MALT lymphoma-associated genetic aberrations in MALT lymphomas of different sites. Leukemia. 2004;18(10):1722-1726.
27. White VA, Gascoyne RD, McNeil BK, et al. Histopathologic findings and frequency of clonality by the polymerase chain reaction in ocular
adnexal lymphoproliferative lesions. Mod Pathol. 1996;9(11):1052-1061.
28. Adachi A, Tamaru J, Kaneko K, et al. No evidence of correlation between BCL10 expression and AP12-MALT1 gene rearrangement in ocular
adnexal MALT lymphoma. Path Intern. 2004;54(1):16-25.
29. Sharara N, Holden JT, Wojno TH, et al. Ocular adnexal lymphoid
proliferations: clinical, histologic, flow cytometric and molecular analysis of
43 cases. Ophthalmology. 2003;110(6):1245-1254.
30.McKelvic PA. Ocular adnexal lymphomas: a review. Adv Anat
Pathol. 2010;17(4):251-261.
31. Gayed I, Eskandari MF, McLaughlin P, et al. Value of positron emission tomography in staging ocular adnexal lymphomas and evaluating their
response to therapy. Ophthalmic Surg Laser Imaging. 2007;38(4):319-325.
32. Sullivan TJ, Valenzuela AA. Imaging features of ocular adnexal lymphoproliferative disease. Eye (Lond). 2006;20(10):1189-1195.
33. Valenzuela AA, Allen C, Grimes D, et al. Positron emission tomography in the detection and staging of ocular adnexal lymphoproliferative
disease. Ophthalmology. 2006;113(12):2331-2337.
34. Portell GA, Aronow ME, Rybicki LA, et al. Clinical characteristics of
95 patients with ocular adnexal and uveal lymphoma: treatment outcomes
in extranodal marginal zone subtype. Clin Lymphoma Myeloma Leuk.
2014;14(3):203-210.
35. Coupland SE, Foss HD, Haydiat AA, et al. Extranodal marginal
zone B cell lymphoma of the uvea: an analysis of 13 cases. J Pathol.
2002;197(3):333-340.
36.Coupland SE, Krause L, Delecluse HJ, et al. Lymphoproliferative lesions of ocular adnexa: analysis of 112 cases. Ophthalmology.
1998;105(8):1430-1441.
37. Decaudin D, de Cremoux P, Vincent-Salomon A, et al. Ocular adnexal
lymphoma: a review of clinicopathologic features and treatment options.
Blood. 2006;108(5):1451-1460.
38. Coupland SE, White VA, Rootman J, et al. A TNM-based clinical staging system of ocular adnexal lymphomas. Arch Pathol Lab Med.
2009;133(8):1262-1267.
39. Edge S, Byrd DR, Compton CC, et al, eds. AJCC Cancer Staging
Manual. 7th ed. New York: Springer; 2010.
40. Rath S. Connors JM, Dolman PJ, et al. Comparison of American
Joint Committee on Cancer TNM-based staging system (7th edition) and
Ann Arbor classification for predicting outcome in ocular adnexal lymphoma.
Orbit. 2014;33(1):23-28.
41. Aronow ME, Portell CA, Rybicki LA, et al. Ocular adnexal lymphoma:
assessment of a tumor-node-metastasis staging system. Ophthalmology.
2013;120(9):1915-1919.
April 2016, Vol. 23, No. 2
42. Graue GF, Finger PT, Maher E, et al. Ocular adnexal lymphoma
staging and treatment: American Joint Committee on Cancer versus Ann
Arbor. Eur J Ophthalmol. 2013;23(3):344-355.
43. Zucca E, Bertoni F, Roggero E, et al. Molecular analysis of the progression from Helicobacter pylori-associated chronic gastritis to mucosaassociated lymphoid-tissue lymphoma of the stomach. N Engl J Med.
1998;338(12):804-810.
44. Decaudin D, Dolcetti R, De Cremoux P, et al. Variable association
between Chlamydophila psittaci infection and ocular adnexal lymphomas:
methodological biases or true geographical variations? Anticancer Drugs.
2008;19(8):761-765.
45. Chanudet E, Zhou Y, Bacon CM, et al. Chlamydia psittaci is variably
associated with ocular adnexa MALT lymphoma in different geographical
regions. J Pathol. 2006;209(3):344-351.
46. Ferreri AJ, Guidoboni M, Ponzoni M, et al. Evidence for an association between Chlamydia psittaci and ocular adnexal lymphomas. J Natl
Cancer Inst. 2004;96(8)586-594.
47.Husain A, Roberts D, Pro B, et al. Meta-analysis of the association between Chlamydia psittaci and ocular adnexal lymphoma
and the response of ocular adnexal lymphoma to antibiotics. Cancer.
2007;110(4):809-815.
48. Chan CC, Smith JA, Shen DF, et al. Helicobacter pylori (H. pylori)
molecular signature in conjunctival mucosa-associated lymphoid tissue
(MALT) lymphoma. Histol Histopathol. 2004;19(4):1219-1226.
49. Chan CC, Shen D, Mochizuki M, et al. Detection of Helicobacter pylori
and Chlamydia pneumoniae genes in primary orbital lymphoma. Trans Am
Ophthalmol Soc. 2006;104:62-70.
50.Lee SB, Yang JW, Kim CS. The association between conjunctival MALT lymphoma and Helicobacter pylori. Br J Ophthalmol.
2008;92(4):534-536.
51. Sjö NC, Foegh P, Juhl BR, et al. Role of Helicobacter pylori in conjunctival mucosa-associated lymphoid tissue lymphoma. Ophthalmology.
2007;114(1):182-186.
52. Decaudin D, Ferroni A, Vincent-Salomon A, et al. Ocular adnexal
lymphoma and Helicobacter pylori gastric infection. Am J Hematol.
2010;85(9):645-649.
53. Coupland SE. A possible new role for Helicobacter pylori in the development of ocular adnexal lymphoma. Am J Hematol. 2010;85(9):641-642.
54. Cohen VM, Sweetenham J, Singh AR. Ocular adnexal lymphoma. What is the evidence for an infectious aetiology? Br J Ophthalmol.
2008;92(4):446-448.
55. Margo CE. From Robert Koch to Bradford Hill. Chronic infection and
the origins of ocular adnexal cancers. Arch Ophthalmol. 2011;129(4):498-500.
56. Gruenberger B, Woehrer S, Troch M, et al. Assessing the role of
hepatitis C, Helicobacter pylori and autoimmunity in MALT lymphoma of
ocular adnexal in 45 Austrian patients. Acta Oncol. 2008;47(3):355-359.
57. Liang Y, Yang Z, Qin B, et al. Primary Sjögren’s syndrome and
malignancy risk: a systematic review and meta-analysis. Ann Rheum Dis.
2014;73(6):1151-1156.
58. Zinzani PL. The many faces of marginal zone lymphoma. Hematology Am Soc Hematol Educ Program. 2012;2012;426-432.
59. Smijanic M, Milosevic R, Antic D, et al. Orbital and ocular adnexal
mucosa-associated lymphoid tissue (MALT) lymphomas: a single-center
10-year experience. Med Oncol. 2013 30(4):722-730.
60. Harada K, Murakami N, Kitaguchi M, et al. Localized ocular adnexal
mucosa-associated lymphoid tissue lymphoma treated with radiation therapy: a long-term outcome in 86 patients with 104 treated eyes. Int J Radiat
Oncol Biol Phys. 2014;88(3):650-654.
61. Tuncer S, Tanyildiz B, Basaran M, et al. Systemic rituximab immunotherapy in the management of primary ocular adnexal lymphoma: single
institution experience. Curr Eye Res. 2015;40(8):780-785.
62. Kiesewetter B, Lukas J, Kuchar A, et al. Clinical features, treatment
and outcome of mucosa-associated lymphoid tissue (MALT) lymphoma of
the ocular adnexa: a single center experience of 60 patients. PLoS One.
2014;9(7):e104004.
63. Martinet S, Ozsahin M, Belkacémi Y, et al. Outcome and prognostic factors in orbital lymphoma: a rare cancer network study on 90 consecutive patients treated with radiotherapy. Int J Rad Oncol Biol Phys.
2003;55(4):892-898.
64. Uno T, Isobe K, Shikama N, et al. Radiotherapy for extranodal
marginal zone B-cell lymphoma of mucosa-associated lymphoid tissue
originating in the ocular adnexa: a multi-institutional, retrospective review
of 50 patients. Cancer. 2003;98(4):865-871.
65. Tanimoto K, Kaneko A, Suzuki S, et al. Primary ocular adnexal
MALT lymphoma: a long-term follow-up study of 114 patients. Jpn J Clin
Oncol. 2007;37(5):337-344.
66. Kao SC, Kau HC, Tsai CC, et al. Lacrimal gland extranodal marginal
zone B-cell lymphoma of MALT-type. Am J Ophthalmol. 2007;143(2):311-315.
67. Ferreri AJ, Ponzoni M, Martinelli G, et al. Rituximab in patients with
mucosa-associated lymphoid tissue type lymphoma of the ocular adnexa.
Haematologica. 2005;90(11):1578-1579.
68. Song E-K, Kim S-Y, Kim TM, et al. Efficacy of chemotherapy as a
first-line treatment in ocular adnexal extranodal marginal zone B-cell lymphoma. Ann Oncol. 2008;19(2):242-246.
April 2016, Vol. 23, No. 2
69. Ben Simon GJ, Cheung N, McKelvie P, et al. Oral chlorambucil for
extranodal marginal zone lymphoma of mucosa-associated lymphoid tissue of the orbit. Ophthalmology. 2006;113(7):1209-1213. 70. Hashimoto N, Sasaki R, Nishimura H, et al. Long-term outcome and
patterns of failure of primary ocular adnexal mucosa-associated lymphoid
tissue lymphoma treated with radiotherapy. Int J Radiat Oncol Biol Phys.
2012;82(4):1509-1514.
71. Paik JS, Cho WK, Lee SE, et al. Ophthalmologic outcomes after
chemotherapy and/or radiotherapy in non-conjunctival ocular adnexal
MALT lymphoma. Ann Hematol. 2012;91(9):1393-1401.
72. Yoon JS, Ma KT, Kim SJ, et al. Prognosis for patients in a Korean
population with ocular adnexal lymphoproliferative lesions. Ophthal Plast
Reconst Surg. 2007;23(2):94-99.
73. Nam H, Ahn YC, Kim YD, et al. Prognostic significance of anatomic
subsites: results of radiation therapy for 66 patients with localized orbital
marginal zone B cell lymphoma. Radiation Oncol. 2009;90(2):236-241.
74. Sullivan TJ, Grimes D, Bunce I. Monoclonal antibody treatment of
orbital lymphoma. Ophthalmic Plast Reconstr Surg. 2004;20(2):103-106.
75. Russell W, Herskovic A, Gessert D, et al. Ocular adnexal MALTomas:
case series of patients treated with primary radiation. Clin Adv Hematol
Oncol. 2013;11(4):209-214.
76. Matsuo T, Yoshino T. Long-term follow-up results of observation or radiation for conjunctival lymphoma. Ophthalmology. 2004;111(6):1233-1237.
77. Lim SH, Kang M, Son J. Extranodal marginal zone B cell lymphoma
of mucosa-associated lymphoid tissue type of the ocular adnexa: retrospective single institution review of 95 patients. Indian J Ophthlamol.
2011;59(4):273-277.
78. Han JJ, Kim TM, Jeon YK, et al. Long-term outcomes of first-line
treatment with doxycycline in patients with previously untreated ocular adnexal marginal zone B cell lymphoma. Ann Hematol. 2015;94(4):575-581.
79. Woolf DK, Kuhan H, Shoffren O, et al. Outcome of primary lymphoma of the ocular adnexal (orbital lymphoma) treated with radiotherapy.
Clin Oncol (R Coll Radiol). 2015;27:(3):153-159.
80. Goda JS, Le LW, Lapperriere NJ, et al. Localized orbital mucosaassociated lymphoid tissue lymphoma managed with primary radiation therapy: efficacy and toxicity. Int J Radiat Oncol Biol Phys. 2011;81(4):e659-e666.
81. Tran KH, Campbell BA, Fua T, et al. Efficacy of low dose radiotherapy for primary orbital marginal zone lymphoma. Leuk Lymphoma.
2013;54(3):491-496.
82. Son SH, Choi BO, Kim, GW, et al. Primary radiation therapy in patients with localized orbital marginal zone B-cell lymphoma of mucosaassociated lymphoid tissue (MALT lymphoma). Int J Radiat Oncol Biol
Phys. 2010;77(1):86-91.
83. Suh CO, Shim SJ, Lee SW, et al Orbital marginal zone B-cell lymphoma of MALT: radiotherapy results and clinical behavior. Radiat Oncol
Biol. 2006;65(1):228-233.
84. Le QT, Eulau SM, George TI, et al. Primary radiotherapy for localized
orbital MALT lymphoma. Int J Radiat Oncol Biol Phys. 2002;52(3):657-663.
85. Ferreri AJ, Ponzoni M, Guidoboni M, et al. Bacteria-eradicating
therapy with doxycycline in ocular adnexal MALT lymphoma: a multicenter
prospective trial. J Natl Cancer Inst. 2006 98(19):1375-1382.
86. Tanimoto K, Kaneko A, Suzuki S, et al. Long-term follow-up results of no initial therapy for ocular adnexal MALT lymphoma. Ann Oncol.
2006;17(1)135-140.
87. Ferreri AJ, Govi S, Pasini E, et al. Chlamydophila psittaci eradication with doxycycline as first-line targeted therapy for ocular adnexae
lymphoma: final results of an international phase II trial. J Clin Oncol.
2012;30(24):2988-2994.
88. Govi S, Dognini GP, Licata G, et al. Six-month oral clarithromycin is
safe and active in extranodal marginal zone B-cell lymphomas: final results
of a single-centre phase II trial. Br J Haematol. 2010:150(2):226-248.
89. Wahl RL, Jacene H, Kasamon Y, et al. From RECIST to PERCIST:
evolving consideration for PET response criteria in solid tumors. J Nucl
Med. 2009;50(suppl 1):122S-50S.
90. Jung SK, Paik JS, Jung SE, et al. Suggestion of response evaluation criteria in patients with ocular adnexal mucosa-associated lymphoid
tissue lymphoma (OAML). Am Hematol. 2015;94(7):1185-1193.
Cancer Control 149
Recognizing ocular complications of
chemotherapy before they result in irreversible
injury involves taking a clinical history
and performing a basic eye examination.
Photo courtesy of Lynn E. Harman, MD. Point Reyes Beach North.
Ophthalmic Complications Related to Chemotherapy in
Medically Complex Patients
Lynn E. Harman, MD
Background: Systemic cancer therapies cause a variety of ophthalmic complications. Mitigating harmful
adverse events involves screening patients at risk for ocular injury and vision loss.
Methods: A review of the relevant literature on the ophthalmic complications of cancer therapy was used to
formulate an approach to screening patients for serious complications presenting at a nonophthalmic specialty center.
Results: Rarely, ocular complications of cancer therapy can occur. Establishing a causal association for any
given agent is complicated because many treatment-related adverse events result in symptoms and ocular
findings indistinguishable from primary eye disorders.
Conclusions: Recognizing potentially serious ocular complications of cancer therapy before they result in
irreversible injury starts with taking a relevant clinical history and performing a basic eye examination,
including assessments of visual acuity and fields. Given the wide range of treatment-related adverse events
and the challenges of diagnosis, the screening process plays an important role in expediting referral to an
ophthalmologic specialist.
Introduction
The majority of published research about the ocular
complications of cancer therapy reviews the adverse
events of individual chemotherapies and targeted
therapies.1-7 Cataloging these adverse events can be approached in several ways. One method is anatomical, in
which the therapeutic agents are listed according to the
parts of the eye or ocular adnexa they affect (eg, conFrom the Department of Ophthalmology, University of South
Florida Morsani College of Medicine, and Section of Ophthalmology,
James Haley Veterans Affairs Hospital, Tampa, Florida.
Address correspondence to Lynn E. Harman, MD, James Haley
Veterans Affairs Hospital, Eye Clinic, 13000 Bruce B. Downs
Boulevard, Tampa, FL 33612. E-mail: [email protected]
Submitted July 14, 2015; accepted October 29, 2015.
No significant relationship exists between the author and the companies/organizations whose products or services may be referenced in this article.
150 Cancer Control
junctiva, lens, retina),1 whereas another method lists
therapies according to the type of injury they cause
(eg, punctate keratitis, sterile uveitis, macular edema).4 It
is also possible to describe ocular toxicities and adverse
events by pharmacological group (eg, plant alkaloids,
antimetabolites, nitrosoureas) or by individual agent.2,3
Such surveys are invaluable when attempting to establish whether causal links exist between a particular ocular condition and a therapeutic agent; however, these inventories do not address clinical questions about how to
manage the onset of a suspected event.
Uncertainty sometimes exists about the seriousness of an ocular symptom or a new finding on an eye
examination if it occurs in patients receiving chemotherapy. For those who are hospitalized or too ill to be
seen at an ophthalmology center, the situation can be
disconcerting. Few adverse drug reactions are characteristic enough to diagnose with absolute certainty on
April 2016, Vol. 23, No. 2
an abbreviated examination, and confirming whether
an adverse event is drug related with a drug rechallenge is usually not an option. Consultative input is desirable even when it may be logistically difficult.
In this article, the term ocular is inclusively used
to describe the eye, visual pathway, and ocular adnexa.
More than 60 distinct ocular-related adverse events
have been associated with systemic chemotherapies
and targeted cancer therapies.1-7 These events vary
from finding asymptomatic deposits in the retina to
cataracts, cranial nerve palsies, to keratoconjunctivitis sicca. Most of these complications have clinical
features that overlap with acquired diseases.1,4,5 Treatment-related adverse events range from being selflimited conditions to permanent vision loss.1,4,5 The
diversity of these complications speaks to the variety
of molecular and cellular mechanisms involved. Thus,
this review will focus on evaluating ocular complaints
occurring in the context of medically complex treatment and when it is difficult for patients to be seen at a
specialty eye clinic.
This paper will also provide a general approach
for screening patients outside the setting of an eye
clinic in order to expedite consultation with an eye
specialist and reduce unnecessary referrals. It will
also address when to anticipate, and possibly preemptively reduce, some common ocular-related adverse events associated with select chemotherapy
agents. Given the vastness of the topic, this paper will
not include complications of radiotherapy, therapies
for primary cancers of the eye and ocular adnexa, or
the ocular manifestations of graft-vs-host disease. Although selected examples of adverse events are cited
to emphasize elements of the ocular history and eye
examination, they are not meant to represent a comprehensive directory of complications.
Incidence
The proportion of patients who experience a serious
adverse event from chemotherapy is low. Most of these
complications are considered rare or reported only as
single case reports.1-7 A 2006 comprehensive review
found that 57 different ocular-related (ie, eye, visual
pathway, ocular adnexal) complications could be attributed to cytotoxic chemotherapies (Table 1).4 A total
of 38% of drug-specific complications were deemed to
be rare and another 46% were identified through case
reports.4 A total of 15% of drug-specific complications
were considered to be somewhat common and 1.4%
were noted to be common.4
The low overall proportion of adverse events comes
with a clinical caveat1-7: The event rate is probably lower than anticipated for “incidental” (or unrelated) findings on routine eye examinations. Common conditions
such as uncorrected presbyopia, refractive errors, dry
eyes, chronic blepharitis, and age-related cataracts can
April 2016, Vol. 23, No. 2
Table 1. — Somewhat Commonly Occurring Adverse Events
of Cytotoxic Chemotherapies
Adverse Event
Drug
Route of
Administration
Arteriovenous shunts
Carmustine
(central nervous system)
Intra-arterial
Blurred vision
Busulfan
Intravenous
5-fluorouracil
Intravenous
5-fluorouracil
Intravenous
Deoxycoformycin
Intravenous
Corneal opacity
Cytosine arabinoside
Intravenous
Cranial nerve palsies
Plant alkaloids
Intravenous
Epiphora
5-fluorouracila
Intravenous
Eye pain
5-fluorouracil
Intravenous
Focal demyelination of
the optic nerve
Carmustine/cisplatin
Intra-arterial
Foreign body sensation
Cytosine arabinoside
Intravenous
5-fluorouracil
Intravenous
Keratitis
Cytosine arabinosidea
Intravenous
Conjunctivitis
5-fluorouracil
Intravenous
Keratoconjunctivitis
sicca
Cyclophosphamide
Intravenous
Busulfan
Intravenous
Macular pigment
Cisplatin
Intravenous
Papilledema
Carmustine
Intra-arterial
Periorbital edema
5-fluorouracil
Intravenous
Methotrexate
Intravenous
Photophobia
5-fluorouracil
Intravenous
Ptosis
Plant alkaloids
Intravenous
Retinal arterial narrowing
Carmustine
Intra-arterial
Retinal hemorrhages
Carmustine
Intra-arterial
aConsidered common.
Adapted from Schmid KE, Kornek GV, Scheithauer W, et al. Update on
ocular complications of systemic cancer chemotherapy. Surv Ophthalmol.
2006;51(1):19-40, with permission from Elsevier.
all cause symptoms that could be mistakenly attributed
to chemotherapy if the clinician has not taken a careful
clinical history and performed a basic eye examination.
Clinicians who do not specialize in ophthalmology are
not typically expected to perform a complete eye examination, nor are they usually familiar with the nuances of
minor, pre-existing eye conditions. This emphasizes the
importance of a clinical history and basic tests of visual
function (with corrective lenses) to screen for potentially
serious problems.
Attributing an ocular complication to a therapeutic agent is a process of exclusion. Some of the most
challenging diagnoses in ophthalmology arise in the
context of cancer treatment; for example, the distinction between sterile drug-related uveitis and infectious
endophthalmitis (typically when a patient is immunocompromised), as well as the distinction between
Cancer Control 151
drug-related cranial nerve palsy and metastatic cancer
or an age-related vascular event.
Complaints
The ophthalmic complications of systemic cancer
therapy can cause some of the same symptoms seen
with primary eye diseases. They can include vision
loss (central, peripheral [or both], night blindness),
abnormal visual perceptions (distorted vision, smoky
vision, double vision, floaters), and all degrees of eye
and periocular pain, among other issues.1-7 The close
temporal relationship of new-onset eye complaints
with the initiation of cancer therapy provides presumptive evidence of a causal link. This chronological correlation, along with the cessation of symptoms
after stopping the offending agent, may be the most
compelling evidence of causation.
Vision Loss/Impairment
Patients who have difficulty describing their visual loss
should be queried as to whether the loss is unilateral or
bilateral, whether it occurs with or without pain, and
whether it was preceded by or associated with other
symptoms. If it was transient, then the clinician should
ask the patient how long it lasted. Asking the patient
to describe the visual experience that surrounded the
sensory phenomenon of vision loss may also be helpful in sorting through diagnostic possibilities.
Table 2 lists clinical inferences typically correlated
with different types of complaints associated with vision loss or impairment.
Double Vision
Acute onset of binocular diplopia requires a neurological evaluation, whereas monocular diplopia does not.
Monocular diplopia is typically caused by an underlying refractive problem. Patients may report double vision as being a shadowing or ghosting of images due
to improperly corrected refractive error. Patients may
not be aware whether they can see 2 images with both
eyes open or with a single eye. Because such a distinction is critical, the clinician must directly question the
patient to confirm and test for true binocular diplopia
in all fields of gaze. If diplopia disappears when either
eye is occluded (ensuring vision is tested in the direction of symptomatic double vision), then double vision
is binocular. New-onset binocular diplopia necessitates
a neurological evaluation.8-15
Examples of chemotherapy agents associated
with cranial nerve palsies or double vision are listed
in Table 3.4,5,8-18
Pain
Characteristics of ocular and periocular pain can provide clues to an underlying disease process. Dry eyes
and mild forms of keratitis may be described as produc152 Cancer Control
ing a “sandy feeling” or foreign-body sensation. Generalized mild eye discomfort, heavy eyelids, and brow
ache associated with near visual tasks are features of asthenopia, which may be secondary to use of outdated
prescription glasses or need for reading glasses or bifocals. Onset of sudden, sharp, stabbing pain with intense
sensitivity to light suggests a corneal epithelial erosion
or corneal abrasion. Typically, 1 drop of topical anesthetic quickly relieves such pain. A deep boring pain
exacerbated by eye movements is suggestive of scleritis. Optic neuritis can also produce pain or discomfort
with eye movements, but it is usually not as severe as
scleritis. Severe, unilateral, ocular and periocular pain
progressing for minutes to hours and associated with ipsilateral blurred vision or halos around lights is a typical
presentation of acute glaucoma. Two symptoms of acute
Table 2. — Clinical Inferences Associated With Symptoms
or Features of Visual Loss
Symptom
Clinical Inference
Colored halos around lights
Corneal edema from elevated intraocular pressure
Decreased vision in dim
illumination
Impaired dark adaption
Vitamin A deficiency
Distortion of straight lines
Macular dysfunction due to edema or
blood beneath or within the retina
Floaters, unilateral or
bilateral
Blood or inflammatory cells in vitreous
Age-related collapse, condensation of
vitreous gel; with photophobia and
eye pain suggests inflammation
Loss of side or peripheral
vision
Branch artery occlusion
Cerebrovascular accident
Retinal detachment
Pain on eye movement
Optic neuritis
Orbital inflammation
Scleritis
Painless “spot” in center
of vision
Macular dysfunction
Optic neuropathy
Photophobia, tender eye
Acute glaucoma
Keratitis
Uveitis
Severe lancet-like pain,
blurred vision
Corneal abrasion
Corneal erosion
Keratitis
Sudden, catastrophic
vision loss
Vascular occlusion of retina or
optic nerve
Transient obscurations
of vision lasting seconds,
usually bilateral
Increased intracranial pressure
Transient visual loss lasting
minutes, binocular
Cardiac arrhythmia
Posterior circulation event
Orthostatic hypotension
Transient visual loss lasting
minutes, monocular
Anterior circulation event
Amaurosis fugax
April 2016, Vol. 23, No. 2
glaucoma, periocular pain and headache, are sometimes
associated with nausea and vomiting.
Terms and phrases frequently used to characterize pain associated with specific ocular conditions are
listed in Table 4.
Table 3. — Chemotherapeutic Agents Associated With
Cranial Nerve Palsies or Diplopiaa
Complication
Select Example
Route of
Administration
Bilateral lateral
rectus palsy
Cytosine arabinoside
and mitoxantrone15
Intravenous
Cavernous sinus
syndrome
Cisplatin14
Intra-arterial
Cranial nerve palsy
Vinca alkaloids10,11
Intravenous
Diplopia
Chlorambucil5
Intravenous
Disturbance in
oculomotor function
5-fluorouracil12
Intravenous
Fibrosis of
extraocular muscles
Carmustine
Nitrosoureas8,13
Intra-arterial
Intravenous
Internuclear
ophthalmoplegia
Methotrexate
Nitrosoureas8,9
Intra-arterial
Intrathecal
Oculomotor nerve
palsy
Interferon α5
Intravenous
Also refer to references 4 and 16 to 18 for a more thorough discussion
on neuro-ophthalmological complications of chemotherapy.
Table 4. — Descriptions of Ocular and Periocular Pain
Description
Clinical Inferencea
Deep boring eye pain worse with
eye movement
Scleritis
Foreign body sensation
Grit or sand in eyes
Dry eyes
Mild keratitis
Mild to moderate pain with eye
movement and vision loss
Optic neuritis
Moderate to severe eye pain
worse with light exposure
Uveitis
Ocular and periocular
discomfort associated
with near visual tasks
Lid heaviness
Tired eyes
Brow ache
Asthenopia
Eye strain
Does not suggest serious
underlying disease
Ocular and periocular pain severity
increasing minutes to hours with
progressive vision loss
Acute glaucoma
Ocular pain in presence of bright
light (photophobia)
After exclusion of known
causes (eg, keratitis, uveitis)
consider “essential” or
idiopathic photophobia
Sudden severe stabbing in 1 eye,
relieved when eye is shut
Recurrent erosion or corneal
abrasion
Pain relief with topical anesthetic
Erosions more common in dry
eyes or eyes with corneal edema
a Clinical
Other Symptoms
Excessive tearing is a common complaint among middle-aged and older individuals with dry eyes (and dry
eye with chronic blepharitis) due to reflex tearing, and
it is often associated with a mild foreign body sensation. Exacerbations can be triggered by air conditioning, low humidity, or use of over-the-counter medications (eg, antihistamines). If the symptoms are not
relieved by treatment with artificial tears, then other
causes of epiphora should be considered. A variety of
chemotherapy agents has been associated with excessive tearing, although the mechanisms triggering excessive or overflow lacrimation differ (Table 5).4,5,7,19-27
Excessive tearing also accompanies disorders of the
ocular surface such as conjunctivitis and keratitis. In
these situations, findings on an eye examination will
usually indicate that the corneal surface lacks its normal luster or that the eye is inflamed.
Because mild photophobia is a common chronic
problem, particularly in certain environmental settings, it is important for the clinician to exclude the
possibility of a pre-existing condition through a careful history. Keratitis and uveitis also commonly cause
photophobia, and these diagnoses must be excluded
before idiopathic (or drug-related) photophobia is a
considered. Doing so requires a slit lamp inspection of
Table 5. — Clinical Inferences Associated With
Excessive Tearing
Relevant History
and Finding
Clinical
Inference
Potential Drug
Implication
Excessive tearing
associated with
mass in medial
canthal region
Consider mechanical
obstruction of nasallacrimal outflow
Swelling in medial
canthal region may or
may not show signs
of inflammation
No drug implications
In context of cancer
treatment, consider
infection or metastatic
tumor
New-onset tearing
associated with
mild red eyes,
ocular discomfort,
or photophobia
Consider secondary
to ocular surface
disorder (keratitis or
conjunctivitis)
Diagnosis of drugrelated keratitis or
conjunctivitis is
process of exclusion
5-fluorouracil19,20
Capecitabine5,21
Cytosine
arabinoside23-25
Deoxycoformycin4
Chorambucil4
Docetaxel26,27
Pre-existing symptoms associated
with itching, or past
“problems” with
eyelids
Consider ocular
allergies, trichiasis,
or abnormalities of
eyelid position
Same medications
that exacerbate dry
eyes can worsen
pre-existing allergy
symptoms
Pre-existing symptoms worse in certain
environments like
air-conditioned
rooms or under fans
Mild foreign body
sensation
Reflex tearing due to
dry eyes
Often exacerbated
by antihistamines
and other common
medications
conditions do not always produce stereotypic pain complaints.
April 2016, Vol. 23, No. 2
Cancer Control 153
the corneal surface (with fluorescein stain) and a magnified examination of the aqueous humor and vitreous cavity for cells and protein transudate. Presence of
photophobia associated with ocular inflammation typically requires referral to an eye specialist.
Eye Examination
Patients unable to be promptly evaluated in a specialty ophthalmology clinic or onsite by an ophthalmologist should have a reliable assessment of visual
function (central and peripheral vision) and a basic
eye examination, which involves a modicum of basic
equipment (Fig).
Central vision can be tested using a near visual
acuity card held at 40 cm. If patients are older than
40 years of age, near vision usually requires bifocals
(for persons using distance correction) or separate
near-reading glasses (unless near sighted). The clinician must individually test each eye. If prescription
glasses are unavailable, then the patient’s uncorrected
vision should be obtained and then tested again using
over-the-counter readers (+1.25 to +3.00 D). For lens
powers greater than +2.50 D, the near visual acuity card
may need to be held closer than 40 cm for best focus.
Confrontation visual field testing is designed to detect gross field defects. The patient should be situated
approximately 1 m from the clinician at equal eye level.
The clinician then presents 1 or 2 fingers in each quadrant of the visual field. While the patient occludes 1 eye
and maintains central fixation with the other, the clinician asks the patient to count the clinician’s fingers,
randomly shifting his or her fingers to include each
sector of the visual field.
In patients complaining of diplopia, the clinician
must confirm their visual experience as binocular
(ie, with both eyes open in each field of gaze). Testing should then be repeated, with the patient closing
each eye so that the clinician can document diplopia
that abates. Typically, moderate and large deviations in
ocular alignment can be detected on casual inspection;
however, small misalignments are difficult to identify,
and they may require special examination techniques.
The Amsler grid is used to screen for macular
disease, and standard color plates (Hardy–Rand–
Ritter or Ishihara) are valuable for the screening of
optic neuropathies. These sensory tests are available in physical prints but are also available online
(Amsler grid: http://development.aao.org/eyecare
/conditions/macular-degeneration/amsler.cfm; color
testing: http://colorvisiontesting.com). Both tests are
presented individually to each eye and take less than
1 minute to complete.
Inspection of periocular tissues, eyelids, and the
front surface of the eyes with good illumination are often sufficient to detect clinically important inflammation during a basic eye examination. Abnormalities of
154 Cancer Control
Fig. — Basic equipment for visual sensory testing includes an Amsler
grid (top left), Ishihara color plates (bottom right), and a near vision
card (bottom left). Visual stimuli are separately presented to each eye,
making sure 1 eye is occluded. Over-the-counter +2.50 glasses (focal
length 40 cm) may be needed for patients older than 40 years who have
no reading glasses or bifocals without myopia.
the cornea can often be appreciated without magnification because the tissue lacks its normal luster and
transparency. If available, 1 drop of sterile fluorescein dye will stain epithelial defects and enhance them
when using a cobalt blue light filter.
A penlight can detect aberrations in the normal
red light reflex seen through the pupil. Deviations are
easier for the clinician to appreciate when they are unilateral and the patient’s opposite eye serves as a control. Although fundamentals of a pupil examination
(eg, size, shape, reactivity to light, presence or absence
of a relative afferent pupillary defect) are beyond the
scope of this article, characterizing pupil function is
helpful when localizing injury to the visual pathway
and for prioritizing special studies.
A penlight may also be useful for detecting small
lesions of the eyelid such as petechial hemorrhages or
abnormal position, but gross swelling or erythema of
the eyelids can often be seen best under general illumination. New-onset proptosis is a rare complication
of chemotherapy.4,14 In a patient with cancer who is
receiving chemotherapy, new-onset proptosis is more
likely to be secondary to hemorrhage (bleeding diathesis or infection) or a rapidly expanding tumor.28-30
Regardless of the cause, new-onset proptosis requires
prompt evaluation. Subtle displacement of the eye in
the orbit can be appreciated by viewing the patient
from several feet away, looking for differences in
symmetry and the relative amounts of visible sclera.
Proptosis can be mimicked by enophthalmos of the
opposite eye (eg, due to metastatic scirrhous breast
carcinoma) or by lid ptosis.
An exception to the penlight detection of important
April 2016, Vol. 23, No. 2
ocular inflammation can occur with uveitis, in which
signs of external inflammation may be subtle. In this
situation, symptoms of pain and photophobia, newonset floaters, or evidence of decreased vision may be
clues to more serious problems.
Clinicians not specializing in ophthalmology are
not typically expected to pursue an eye examination
beyond these tests because doing so usually requires
use of special equipment (slit lamp, tonometer, ophthalmoscope).
Anticipated Complications
Few complications occur with sufficient regularity that
some authorities recommend preventive measures to
mitigate their occurrence (Table 6).4,5,7,19-27,31-37 Examples of such complications include keratitis caused by
cytarabine, ocular irritation from high-dose methotrexate, and canalicular and nasolacrimal duct stenosis after taking docetaxel.19-25,27 Optimal strategies
are not universally agreed upon, but most offer some
benefit.31,33,35,37
Logistical Dilemma
Onset of new visual symptoms or other ocular complaints in patients receiving cancer treatment could
represent an opportunistic infection, metastatic disease, or the coincidental occurrence or exacerbation of an unrelated eye disease. Some ocular or visual complaints arising in medically complex cases or
in high-intensity medical care settings are neither
vision-threatening nor have long-term adverse consequences. Most ocular-related complications of chemotherapy are rare,1-7 so a clinical history and eye
Table 6. — Preemptive Strategies for Common
Ocular-Related Adverse Events of Chemotherapy
Adverse
Event
Strategya
5-fluorouracil
Conjunctivitis
(noninfectious)
Cool compresses
Topical methylcellulose drops
Topical corticosteroids 4,37
Cyclophosphamide
Dry eyes
Artificial tear treatment4,7
Cytosine
arabinoside
Keratitis
Topical corticosteroid drops
started prior to treatment and
tapered33-36
Topical artificial tears 33-36
Agent
Topical 2-deoxycytine drops
prior to therapy 33,36
Docetaxel
Canalicular
fibrosis
Secondary
epiphora
Temporary intubation of
lacrimal puncta in patients
with early symptoms 31
Methotrexate
Keratitis
Photophobia
Artificial tear treatment 32
aThe
references cited discuss the relative merits of therapy and alternative strategies.
April 2016, Vol. 23, No. 2
examination are usually required to distinguish treatment-related adverse events from common disorders
of the eye, as well as to differentiate potentially serious
from relatively unimportant problems. However, when
patients cannot be examined at an eye clinic in a timely
manner because of extenuating circumstances, screening for serious eye complications must be performed
by their primary care physicians and oncologists.
Conclusions
The potential ocular complications with the greatest
likelihood of causing adverse outcomes present with
demonstrable declines in visual function (eg, loss of
central vision, loss of peripheral vision), symptoms
and/or signs of oculomotor nerve palsy (eg, diplopia,
loss of ocular alignment related to cranial nerves III,
IV, or VI), or abnormalities detectable on penlight
examination (eg, loss of red reflex; inflammation or
swelling of the eyelids, conjunctiva, sclera; proptosis).1-7,16-18 Correlating symptoms and basic clinical
findings with previously reported treatment-related
adverse events is the first step in identifying possible
causative drugs.1-7,16-18 Because causal associations with
drug exposures usually involve the exclusion of other
disease processes, it is likely that an ophthalmologic
specialist will need to be involved in the diagnostic
evaluation; however, the urgency of the consultation
will be dictated by the results of screening.
For patients with suspected ocular complications
related to cancer therapy who cannot be quickly seen at
a specialty eye clinic, care must be expedited through
communication with an ophthalmologist and include
relevant ocular history findings, results of vision tests,
and findings from the external eye examination.
References
1. Imperia PS, Lazarus HM, Lass JH. Ocular complications of systemic cancer chemotherapy. Surv Ophthalmol. 1989;34(3):209-230.
2. Renouf DJ, Velazquez-Martin JP, Simpson R, et al. Ocular toxicity
of targeted therapies. J Clin Oncol. 2012;30(26):3277-3286.
3. Liu CY, Francis JH, Brodie SE, et al. Retinal toxicities of cancer
therapy drugs. Biologics, small molecule inhibitors, and chemotherapies.
Retina. 2014;34(7):1261-1280.
4. Schmid KE, Kornek GV, Scheithauer W, Binder S. Update on ocular complications of systemic cancer chemotherapy. Surv Ophthalmol.
2006;51(1)19-40.
5. Al-Tweigeri T, Nabholtz, JM, Mackey JR. Ocular toxicity and cancer
chemotherapy: a review. Cancer. 1996;78(7):1359-1373.
6. Kheir WJ, Sniegowski MC, El-Sawy T, et al. Ophthalmic complications of targeted cancer therapy and recently recognized ophthalmic complications of traditional chemotherapy. Surv Ophthalmol. 2014;59(5):493-502.
7. Hazin R, Abuzetun JY, Daoud YJ, et al. Ocular complications of
cancer therapy: a primer for the ophthalmologist treating cancer patients.
Curr Opin Ophthalmol. 2009;20(4):308-317.
8. Lepore FE, Nissenblatt MJ. Bilateral internuclear ophthalmoplegia
after intrathecal chemotherapy and cranial irradiation. Am J Ophthlamol.
1981;92(6):851-853.
9. Kapp J, Vance R, Parker LJ, et al. Limitations of high dose intraarterial 1,3-bis (2-chloroethyl)-1-nitrosourea (BCNU) chemotherapy for
malignant gliomas. Neurosurgery. 1982;10(6):715-719.
10. Albert DM, Wong VG, Henderson ES. Ocular complications of vincristine therapy. Arch Ophthalmol. 1967;78(6):709-713.
11. Toker E, Yenice O, Oğüt MS. Isolated abducens nerve palsy induced
by vincristine therapy. J AAPOS. 2004;8(1):69-71.
12.Bixenman WW, Nicholis JV, Warwick OH. Oculomotor disturCancer Control 155
bances associated with 5-flurouracil chemotherapy. Am J Ophthalmol.
1977;83(6):789-793.
13. Greenberg HS, Ensminger WD, Chandler WF, et al Intra-arterial
BCNU chemotherapy for treatment of malignant gliomas of the central nervous system. J Neurosurg. 1984;61(3):423-429.
14. Margo CE. Murtagh FR. Ocular and orbital toxicity after intracarotid
cisplatin therapy. Am J Ophthalmol. 1993;116(4):508-509.
15. Ventura GJ, Keating MJ, Castellanos AM, et al. Reversible bilateral
lateral rectus muscle palsy associated with high-dose cytosine arabinoside and mitoxantrone therapy. Cancer. 1986;58(8):1633-1635.
16.Singh P, Singh A. Ocular adverse effects of anti-cancer chemotherapy and targeted therapy. J Can Therap Res. Published 2012.
h t t p : / / w w w. h o a j o n l i n e . c o m / j o u r n a l s / p d f / 2 0 4 9 -7 9 6 2 -1- 5 . p d f .
Accessed March 10, 2016.
17. Nolan CP, DeAngelis LM, Neurologic complications of chemotherapy
and radiation therapy. Continuum. 2015;21(2):429-451.
18. Sioka C, Kyritsis AP. Central and peripheral nervous system toxicity of common chemotherapeutic agents. Cancer Chemother Pharmacol.
2009;63(5):761-767.
19. Agarwal MR, Esmaeli B. Burnstine MA. Squamous metaplasia of
the canaliculi associated with 5-fluorouracil: a clinicopathologic case report.
Ophthalmology. 2002;109(12):2359-2361.
20. Brink HM, Beex LV. Punctal and canalicular stenosis associated
with systemic fluorouracil therapy. Doc Ophthalmol. 1995;90(1):1-6.
21. Fraunfelder FT, Meyer SM. Ocular toxicity of antineoplastic agents.
Ophthalmology. 1983;90(1):1-3.
22. Kende G, Sirkin SR, Thomas PR, et al. Blurring of vision: a previously undescribed complication of cyclophosphamide therapy. Cancer.
1979;44(1):69-71.
23. Barletta JP, Fanous MM, Margo CE. Corneal and conjunctival toxicity
with low-dose cytosine arabinoside. Am J Ophthalmol. 1992;113(5):587‑588.
24. Ritch PS, Hansen RM, Heuer DK. Ocular toxicity from high-dose
cytosine arabinoside. Cancer. 1983;51(3):430-432.
25. Hopen G, Mondino BJ, Johnson BL, et al. Corneal toxicity with
systemic cytarabine. Am J Ophthalmol. 1981;91(4):500-504.
26. Esmaeli B, Burnstine MA, Ahmadi MA, et al. Docetaxel-induced
histologic changes in the lacrimal sac and the nasal mucosa. Ophthal
Plast Reconstr Surg. 2003;19(4):305-308.
27. Esmaeli B, Hidaji L, Adinin RB, et al. Blockage of the lacrimal drainage
apparatus as a side effect of docetaxel therapy. Cancer. 2003;98(3);504-507.
28. Barahimi B, Patel A, Bilyk JR. Orbital metastasis mimicking subperiosteal abscess. Orbit. 2010;29(3):165-167.
29. McNab AA. Nontraumatic orbital hemorrhage. Surv Ophthalmol.
2014;59(2)168-184.
30. Valenzuela AA, Archibald CW, Fleming B, et al. Orbital metastasis:
clinical features, management and outcome. Orbit. 2009;28(2-3):153-159.
31. Leyssens B, Wildiers H, Lobelle JP, et al. A double-blind randomized
phase II study on the efficacy of topical eye treatment in the prevention of
docetaxel-induced dacryostenosis. Ann Oncol. 2010;21(2):419-423.
32. Doroshow JH, Locker GY, Gaasterland DE, et al. Ocular irritation
from high dose methotrexate therapy: pharmacokinetics of drug in the tear
film Cancer. 1981;48(10):2158-2162.
33. Lass JH, Lazarus HM, Reed MD, et al. Topical corticosteroid therapy for corneal toxicity from systemically administered cytarabine. Am J
Ophthalmol. 1982;94(5):617-621.
34. Stentoft J. The toxicity of cytarabine. Drug Saf. 1990;5(1):7-27.
35. Higa GM, Gockerman JP, Hunt AL, et al. The use of prophylactic
eye drops during high-dose cytosine arabinoside therapy. Cancer.
1991;68(8):1691-1693.
36. Lazarus HM, Hartnett, Reed MD, et al. Comparison of the prophylactic effects of 2-deoxycytidine and prednisolone for high-dose intravenous
cytarabine-induced keratitis. Am J Ophthalmol. 1987;105(5):476-480.
37. Loprinzi CL, Wender DB, Veeder MH, et al. Inhibition of 5-fluorouracil-induced ocular irritation by ocular ice packs. Cancer. 1994;74(3):945-948.
156 Cancer Control
April 2016, Vol. 23, No. 2
Disparities 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
Background: Interaction effects of poverty and health care insurance coverage on overall survival rates of
breast cancer among women of color and non-Hispanic white women were explored.
Methods: We analyzed California registry data for 2,024 women of color (black, Hispanic, Asian, Pacific
Islander, American Indian, or other ethnicity) and 4,276 non-Hispanic white women (Anglo-European ancestries and no Hispanic-Latin ethnic backgrounds) diagnosed with breast cancer between the years 1996
and 2000 who were then followed until 2011. The 2000 US census categorized rates of neighborhood poverty.
Health care insurance coverage was either private, Medicare, Medicaid, or none. Cox regression was used to
model rates of survival.
Results: A 3-way interaction between ethnicity, health care insurance coverage, and poverty was observed.
Women of color inadequately insured and living in poor or near-poor neighborhoods in California were the
most disadvantaged. Women of color adequately insured and who lived in such neighborhoods in California were also disadvantaged. The incomes of such women of color were typically lower than the incomes of
non-Hispanic white women.
Conclusions: Women of color with or without insurance coverage are disadvantaged in poor and near-poor
neighborhoods of California. Such women may be less able to bare the indirect, direct, or uncovered costs of
health care for breast cancer treatment.
Background
Prognoses are excellent among women with breast cancer diagnosed early and treated in a timely manner with
evidence-based surgical and adjuvant care.1 The vast
majority of such women will survive for 5 to 10 years
or more with a high quality of life, but racial and ethFrom the Dalla Lana School of Public Health (SH-J, EJH), University of Toronto, Toronto, Ontario, Canada, School of Social Work
(KMG), University of Windsor, Windsor, Ontario, Canada, Departments of Geography (INL), Epidemiology and Biostatistics (GZ), and
Oncology (CH), School of Medicine and Dentistry (CH), Western
University, London, Ontario, Canada, Robarts Research Institute
(GZ), London, Ontario, Canada, and the Department of Oncology
(CH), Windsor Regional Cancer Center, Windsor, Ontario, Canada.
Address correspondence to Kevin M. Gorey, PhD, 401 Sunset Avenue,
Windsor Ontario, Canada, N9B 3P4. E-mail: [email protected]
Submitted August 12, 2015; accepted January 19, 2016.
This study was supported in part with funds from Canadian Institutes of Health Research grant no. 67161-2. This study was also
supported by the California Department of Public Health; National
Cancer Institute’s Surveillance, Epidemiology and End Results Program contracts HHSN261201000140C, HHSN261201000035C, and
HHSN261201000034C; and the Centers for Disease Control and
Prevention’s National Program of Cancer Registries under agreement U58DP003862-01. The funders had no role in study design,
data collection and analysis, decision to publish, or preparation of
the manuscript.
The ideas and opinions expressed herein are those of the authors,
and endorsement by the State of California, the Department of Public Health, the National Cancer Institute, and the Centers for Disease
Control and Prevention or their contractors and subcontractors is
not intended or should be inferred.
April 2016, Vol. 23, No. 2
nic disparities persist.1 Findings from systematic reviews
have found consistent disadvantages in breast cancer
screening, diagnosis, treatment, and survival rates in the
United States among ethnic minority women of color
compared with non-Hispanic white women.2-13 NonHispanic white women have Anglo-European ancestries and no Hispanic-Latin ethnic backgrounds. Women of color represent a diverse population — defined
as black, Hispanic, Asian, Pacific Islander, American
Indian, or other minority ethnicity — and certain subpopulations of Asian and Hispanic American women
even seem to be advantaged on access to breast cancer
care and survival.14,15 However, ethnic minority women
of color who live in poverty or are inadequately insured
tend to be more alike than higher income women of
color and they also tend to be disadvantaged on cancer
care compared with non-Hispanic white women.14,15
This field of research may also be limited by its focus on the main effects of ethnicity, rates of income, and
health care insurance coverage. Access to cancer care
as well as rates of survival may be affected by diverse
sociodemographic and economic factors, possibly in
complex ways.16,17 For example, a 3-way interaction of
ethnicity, health care insurance coverage, and poverty
has been observed among patients with colon cancer,
indicating that the multiplicative disadvantage of being
inadequately insured and living in impoverished areas
was worse for African Americans than for non-Hispanic
Cancer Control 157
white Americans.16 Furthermore, such disadvantages
may be greater for women than for men.17-20
Because select groups of African Americans and
women who live in impoverished areas have fewer capital reserves than their non-Hispanic white American
counterparts, researchers have suggested that these
vulnerable groups may be less able to absorb the indirect, direct, or uncovered costs of cancer care.21 This
suggestion led us to hypothesize a 3-way interaction of
ethnicity, health care insurance coverage, and poverty.
Furthermore, we hypothesize that the interaction will
operate such that the survival disadvantage of women
of color with breast cancer compared with non-Hispanic
white women with breast cancer will be greatest in places where the economic divide between them is greatest.
Methods
Women diagnosed with breast cancer between 1996
and 2000 were randomly selected from 3 socioeconomic strata of the California Cancer Registry and
followed until 2011. Cancer data were joined via US
census tracts to the 2000 US census with strata based
on federal poverty criteria defined as extremely
poor (≥ 30% households poor), poor (5%–29%), and
near-poor neighborhoods (< 5% poor).22-24 Based on
previous analyses, primary health care insurance coverage was defined as adequate (private or Medicare) or
inadequate (Medicaid or none).15,16,18-20
Oversampling of women living in poverty seemed
to be associated with oversampling of women of color.
Approximately one-third of this sample was women
of color (n = 2,024), defined as being black, Hispanic,
Asian, Pacific Islander, American Indian, or other ethnicity; the other two-thirds were non-Hispanic white
women (n = 4,276). Within the study population, women of color were represented as black (28%), Hispanic
(49%), Asian, Pacific Islander, American Indian (21%),
or other minority ethnicity (2%). None of the ethnic
minority subsamples significantly differed from each
other with regard to rates of low income or inadequate
health care insurance coverage.
We used an age- and tumor grade–adjusted Cox
regression model to explore hypotheses about the interacting effects of ethnicity, health care insurance coverage, and level of poverty on the predictive outcomes
of 7-year overall survival (OS) rates (SPSS Statistics for
Windows, v22.0, IBM, Armonk, New York).25 Hazard
ratios (HRs) and 95% confidence intervals (CIs) were
estimated. Prevalence estimates were used to describe
our study population, and survival rates aided interpretation of the observed 3-way interaction. Prevalence
estimates and rates per 100 participants were directly
adjusted for age and grade using the study population
as the standard population and reported as percentages. Standardized prevalence ratios, rate ratios, or
rate differences were then used to assess the practical
158 Cancer Control
significance of discrete comparisons with chi-square
test–based 95% CIs. The median test was used for continuous comparisons of skewed distributions.26 Other
details have been previously reported elsewhere.27,28
The study was reviewed and cleared by the University of Windsor research ethics board.
Results
Study Sample
Table 1 displays study sample descriptions. Women of
color were significantly younger than the non-Hispanic white women. They were also more likely to have
high-grade, poorly or undifferentiated tumors than
non-Hispanic white women. Women of color (56%)
were also more than twice as likely as non-Hispanic
white women (22%) to live in poor neighborhoods (adjusted prevalence ratio = 2.63; 95% CI, 2.46–2.81), and
they were nearly twice as likely to be inadequately in-
Table 1. — Sociodemographic and Clinical
Characteristics of Women With Breast Cancer at Diagnosis:
Unadjusted Percentage Distributions
Variable
Non-Hispanic
White Women
n
Women of
Color
%
n
%
Age,a y
25–44
458
10.7
446
22.0
45–54
874
20.4
512
25.3
55–64
907
21.2
437
21.6
65–74
1,021
23.9
354
17.5
≥ 75
1,016
23.8
275
13.6
<5
1,763
41.2
337
16.6
5–29
1,552
36.3
548
27.1
≥ 30
961
22.5
1139
56.3
Private
2,390
55.9
986
48.7
Medicare
1,298
30.4
476
23.5
Medicaid
167
3.9
298
14.7
Uninsured
421
9.8
264
13.0
Node negative
2,942
68.8
1227
60.6
Node positive
1,334
31.2
797
39.4
Well differentiated
1,052
24.6
279
13.8
Moderately differentiated
1,843
43.1
753
37.2
Poorly or undifferentiated
1,381
32.3
992
49.0
Neighborhood poverty, %
Primary health insurers
Summary stage
Tumor grade
All categorical ethnic group differences were statistically significant
(chi-square test; P < .001).
a Non-Hispanic white women (M = 62.9; SD = 14.1) vs women of color
(M = 56.9; SD = 14.3); 1-way analysis of variance = 246.01; P < .001.
M = mean, SD = standard deviation.
April 2016, Vol. 23, No. 2
sured compared with nonHispanic white women (ie,
uninsured or Medicaid insured; 28% vs 14%; adjusted
prevalence ratio = 1.84; 95%
CI, 1.71–2.05). Therefore,
further analyses were adjusted for age and grade
while testing the effects of
ethnicity, health care insurance coverage, and poverty.
Table 3. — Description of 3-Way Interaction of Ethnicity, Health Insurance, and Poverty on
7-Year Survival Rates Among Women With Breast Cancer
Ethnic Group Living Within
a Location
No. of
Cases of
Breast
Cancer
Rate, %
Rate
Ratioa
95% CI
Difference
in Rate of
Survival,
%
0.90–0.99
4.4
Poor or near-poor neighborhood
and adequately insured
Non-Hispanic white women
Women of color
2,922
76.7
1.00
714
72.3
0.94
Poor or near-poor neighborhood
and inadequately insured
Interaction of Ethnicity
Non-Hispanic white women
393
68.6
1.00
by Health Insurance
Women of color
171
54.5
0.79
0.69–0.91
14.1
Coverage and Poverty
Extremely poor neighborhood
Table 2 displays the survival
analysis. Consistent with
Non-Hispanic white women
961
64.1
1.00
findings from previous reWomen of color
1,139
62.0
0.97
0.91–1.03
2.1
search,14-20 having adequate
All rates were adjusted for age and tumor grade.
a A rate ratio of 1.00 was the baseline.
health care insurance covCI = confidence interval.
erage predicted rates of OS
while living in poverty and
being a woman of color
Women of color and non-Hispanic white women living
predicted rates of mortality. The women of color in our
study were twice as likely to die within 7 years of being
in extremely poor neighborhoods did not significantly
diagnosed with breast cancer than were non-Hispanic
differ on rates of OS, and the effect of health care insurance coverage did not differ by ethnicity. Typical or
white women (HR = 2.28). Significant 2-way interactions
median annual household incomes of women of color
of ethnicity with adequate health care insurance cover($24,050) and non-Hispanic white women ($25,150) were
age and poverty as well as a significant 3-way interacalso similar among those living in high poverty places.
tion were observed (P = .047).
Among women living in lower poverty, poor, or
The 3-way interaction of ethnicity, health care insurnear-poor neighborhoods, women of color with priance coverage, and level of poverty is depicted in Table 3.
vate or Medicare insurance coverage were modestly
disadvantaged on rates of OS compared with their
counterparts (4% rate difference to 6% rate ratio differTable 2. — Cox Regression Effects of Ethnicity,
Health Insurance, and Poverty on 7-Year Mortality Rates
ential). Among these women, median incomes of womAmong Women With Breast Cancer
en of color ($61,700) and non-Hispanic white women
($68,725) differed by more than $7,000 (P < .05). In the
Predictor Variable
HR
95% CI
same
poor or near-poor neighborhoods, women of colMain Effecta
or inadequately insured by Medicaid or those without
Ethnicity (non-Hispanic white women)
health care insurance coverage had an OS disadvanWomen of color
2.28
1.43–3.63
tage when compared with similar non-Hispanic white
Health insurer (uninsured or Medicaid)
women (14% rate difference to 21% rate ratio differenMedicare or private insurance
0.71
0.60–0.85
tial). The incomes of these women of color ($46,425)
were typically about $15,000 lower than those of
Neighborhood poverty (near poor)
non-Hispanic white women ($61,000; P < .05). The
Poor
1.34
1.17–1.53
OS disadvantage among women of color was greatest
Extremely poor
1.90
1.63–2.23
among those living in places where the economic diInteraction Effect
vide between women of color and non-Hispanic white
Ethnicity by health insurer
0.66
0.51–0.85
women was greatest.
Ethnicity by poverty
0.58
0.35–0.96
Ethnicity by health insurer by poverty
1.33
1.00–1.75
All effects were adjusted for all other effects in the regression model
as well as for age and tumor grade.
a Baseline.
CI = confidence interval, HR = hazard ratio.
April 2016, Vol. 23, No. 2
Adjunct Interpretive Findings
Women of color living in poor and near-poor neighborhoods were less likely to be diagnosed early
with node-negative disease (rate ratio = 0.95; 95% CI,
0.90–1.00). When breast-conservation surgery was
Cancer Control 159
the most indicated mode of therapy, women of color
were less likely than their non-Hispanic white women counterparts to receive it (rate ratio = 0.94; 95% CI,
0.88–1.00) or breast reconstruction (rate ratio = 0.44;
95% CI, 0.32–0.60). Women of color were significantly less likely to receive all adjuvant therapies when
they were the most indicated: radiotherapy (rate
ratio = 0.91; 95% CI, 0.84–0.99), chemotherapy (rate ratio = 0.91; 95% CI, 0.83–0.99), or hormone therapy (rate
ratio = 0.87; 95% CI, 0.78–0.97). Women of color were
also more likely to experience long waits for initial surgery (≥ 90 days after diagnosis; rate ratio = 1.48; 95%
CI, 1.15–1.91) and radiotherapy (≥ 120 postoperative
days; rate ratio = 1.28; 95% CI, 1.03–1.59). When these
factors were added to the HR model of OS, the interactions involving ethnicity as well as the main effect of
ethnicity no longer entered the model.
Discussion
The results of this study suggest that an interaction exists between ethnicity, health care insurance coverage,
and poverty on rates of OS among a cohort of women
with breast cancer living in select areas of California.
Our data were able to produce 3 central findings across
these 3 socioeconomic strata.
Among women of color and non-Hispanic white
women living in extremely poor neighborhoods — in
areas where at least 30% of the households were poor
— the rates of OS did not significantly vary, and this
finding is similar to a previous report.23 Regardless of
ethnicity, women living in extremely poor neighborhoods appear to be have similar cancer care and OS
disadvantages.28 The largest rate of OS based on ethnicity was seen among those who were uninsured
or Medicaid-insured and who also lived in neighborhoods where poverty was less prevalent. Women of
color with breast cancer living in such poor or nearpoor neighborhoods were 21% less likely to survive
than their non-Hispanic white women counterparts.
Living in poor or near-poor neighborhoods
proved to have the greatest effect on ethnicity and
income. The difference among women of color and
non-Hispanic white women in annual income was
$14,575. Thus, it may be possible that uninsured women of color are less able to bare the uncovered costs
of care due to a possible inability to cover out-of-pocket expenses; in addition, women of color covered by
Medicaid were also at a disadvantage.29 In the same
lower-poverty neighborhoods, women of color with
private health care insurance or those with Medicare
were 6% less likely to survive than were similarly insured non-Hispanic white women. On average, income
among these women of color was $7,025 less than that
of non-Hispanic white women. Women of color with
private health care insurance coverage may have been
more likely to be covered by so-called “bronze plans”
160 Cancer Control
with high deductibles, whereas women of color with
Medicare coverage may be less able to purchase necessary “Medigap” coverage.30,31
Disadvantages among women of color may also
exist in relation to diagnostic and therapeutic care for
breast cancer due to the possible inadequacy of their
incomes and health care insurance coverage. That is to
say that the effects we observed in this study may not
be racial or ethnic effects per se; rather, they may be
socioeconomic effects. This inference could be interpreted to mean that race or ethnicity does not matter
in this instance. However, we think not for the following reasons. Our findings are similar to those of other
studies, which may have observed only the tip of the
proverbial socioeconomic iceberg — these disparities
may be the result of structural inequalities not only
in health care, but in education, employment, housing, and banking.5-10,32-34 For example, compared with
non-Hispanic white women, women of color are more
likely to live in poverty, to live in deeper poverty, and
be less wealthy.21,35,36 Lacking capital reserves seems to
further disadvantage women of color in many ways, including compounding their inability to purchase adequate health care insurance coverage for breast cancer
care. Race/ethnicity still seems to matter very much in
American health care.37
Limitations
We focused on OS rates rather than cancer-specific
rates of survival. Although vital status and survival
duration are accurate in cancer registries, the underlying cause of death probably is not.38-40 In addition, the
underlying cause of many deaths not coded as being
a cancer-related death can be directly associated with
lack of treatment or with treatment-related complications.41 Therefore, we believe that OS has a higher rate
of accuracy and is a better practical indicator of policy
and of clinical significance.
Our findings could be confounded by comorbid
differences between women of color and non-Hispanic
white women. The California Cancer Registry did not
code comorbidities known to be associated with socioeconomic factors and breast cancer survival.42 However,
women of color and non-Hispanic white women with
similar tumors were compared through mathematical
modeling, matching them to cancer virulence proxy,
grade, and on 2 correlates of other chronic diseases
(age, poverty). Therefore, the 2 groups are quite similar,
making comorbid alternative explanations unlikely.
Our findings about women of color living in California may not be generalizable to all such women in
the United States. Our sample of women of color was
composed of even more diverse subsamples, some of
which were quite small. The majority of the Hispanic
participants were Mexican American (83%) and the remainder had diverse Central or South American or CaApril 2016, Vol. 23, No. 2
ribbean heritages. Asian American participants were
Filipino (31%), Chinese (20%), Japanese (15%), Korean
(6%), Vietnamese (6%), East Indian or Pakistani American (5%), or of another Asian heritage. The consistency
and significance, both statistical and practical, of our
findings, along with our explanation of a coherent
health care insurance theory, bode for convergence.
That is, it seems likely that these findings apply to most
women of color, particularly those with inadequate incomes and health care insurance coverage.
This study was exploratory. Its findings are best
thought of as screened hypotheses. We recommend
that investigators systematically replicate these analyses with much more powerful ethnic group subsamples. Narrative study of the experiences of each ethnic
group would also substantially advance our practical
knowledge.
Conclusions
Women of color living in poor and near-poor neighborhoods of California are disadvantaged in terms of
breast cancer care. It is those neighborhoods where
they may be less able than non-Hispanic white women
to bare the indirect, direct, or uncovered costs of care.
Intersecting structural barriers may exist between
high-quality care for women of color, those who live
in poverty, and those who are uninsured or underinsured. Thus, US policy makers ought to be cognizant
of these factors as they consider future reforms of
health care.
Acknowledgments: We acknowledge the administrative and logistical assistance of Kurt Snipes of the
Cancer Surveillance and Research Branch, California
Department of Public Health. We also acknowledge
the research and technical assistance of Arti ParikhPatel of the California Cancer Registry and Madhan
Balagurusamy, Daniel Edelstein, and Nancy Richter of
the University of Windsor.
References
1. DeSantis C, Ma J, Bryan L, Jemal A. Breast cancer statistics, 2013.
CA Cancer J Clin. 2014;64(1):52-62.
2. 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.
3. Jones CEL, Maben J, Jack RH, et al. A systematic review of barriers to early presentation and diagnosis with breast cancer among black
women. BMJ Open. 2014;4:4076.
4. Alexandraki I, Mooradian AD. Barriers related to mammography use
for breast cancer screening among minority women. J Natl Med Assoc.
2010;102(3):206-218.
5. Lee HY, Vang S. Barriers to cancer screening in among Americans: the
influence of health care accessibility, culture, and cancer literacy. J Commun
Health. 2010;35(3):302-314.
6. Purc-Stephenson RJ, Gorey KM. Lower adherence to screening
mammography guidelines among ethnic minority women in America: a
meta-analytic review. Prev Med. 2008;46(6):479-488.
7. Wells KJ, Roetzheim RG. Health disparities in receipt of screening mammography in Latinas: a critical review of recent literature. Cancer
Control. 2007;14(4):369-379.
8. Wu TY, Bancroft J, Guthrie B. An integrative review on breast cancer
screening practice and correlates among Chinese, Korean, Filipino, and Asian
April 2016, Vol. 23, No. 2
Indian American women. Health Care Women Int. 2005;26(3):225-246.
9. Shavers VL, Brown ML. Racial and ethnic disparities in the receipt
of cancer treatment. J Natl Cancer Inst. 2002;94(5):334-357.
10. Wen KY, Fang CY, Ma GX. Breast cancer experience and survivorship among Asian Americans: a systematic review. J Cancer Surviv.
2014;8(1):94-107.
11. Lopez-Class M, Gomez-Duarte J, Graves K, et al. A contextual
approach to understanding breast cancer survivorship among Latinas.
Psychooncology. 2012;21(2):115-124.
12. McKenzie F, Jeffreys M. Do lifestyle or social factors explain ethnic/
racial inequalities in breast cancer survival? Epidemiol Rev. 2009;31:52-66.
13. Gerend MA, Pai M. Social determinants of black-white disparities
in breast cancer mortality: a review. Cancer Epidemiol Biomarkers Prev.
2008;17(11):2913-2923.
14. Haji-Jama S, Gorey KM, Luginaah IN, et al. Health insurance mediation of the Mexican American non-Hispanic white disparity on early
breast cancer diagnosis. SpringerPlus. 2013;2:285.
15. Richter NL, Gorey KM, Haji-Jama S, et al. Care and survival of
Mexican American women with node negative breast cancer: historical cohort evidence of health insurance and barrio advantages. J Immigr Minor
Health. 2015;17(3):652-659.
16. Gorey KM, Haji-Jama S, Bartfay E, et al. Lack of access to chemotherapy for colon cancer: multiplicative disadvantage of being extremely
poor, inadequately insured and African American. BMC Health Serv Res.
2014;14:133.
17. Levitz NR, Haji-Jama S, Munro T, et al. Multiplicative disadvantage
of being an unmarried and inadequately insured woman living in poverty
with colon cancer: Historical cohort in California. BMC Womens Health.
2015;15:8.
18. Gorey KM, Luginaah IN, Holowaty EJ, et al. Mediation of the effects of living in extremely poor neighborhoods by health insurance: Breast
cancer care and survival in California, 1996 to 2011. Int J Equity Health.
2013;12:6.
19. Gorey KM, Richter NL, Luginaah IN, et al. Breast cancer among
women living in poverty: Better care in Canada than in the United States.
Soc Work Res. 2015;39(2):107-118.
20. Gorey KM, Luginaah IN, Bartfay E, et al. Better colon cancer care
for extremely poor Canadian women compared with American women.
Health Soc Work. 2013;38(4):240-248.
21. Shapiro TM. The Hidden Cost of Being African American: How
Wealth Perpetuates Inequality. New York: Oxford University Press; 2004.
22. US Census Bureau. 2000 Census of Population and Housing in
California: Summary Tape File 3 [CD-ROM]. Washington, DC: US Government Printing Office; 2002.
23. Wilson WJ. The Truly Disadvantaged: The Inner City, The Underclass,
and Public Policy. 2nd ed. Chicago, IL: University of Chicago Press; 2012.
24. Jargowsky PA. Stunning progress, hidden problems: The dramatic
decline of concentrated poverty in the 1990s. In: Berube A, Katz B, Lang
RE, eds. Redefining Urban and Suburban America: Evidence from Census
2000. Washington, DC: Brookings Institution Press; 2005: 137-171.
25. Vittinghoff E, Glidden DV, Shiboski SC, et al. Regression Methods
in Biostatistics: Linear, Logistic, Survival, and Repeated Measures Models.
2nd ed. New York: Springer; 2012.
26. Corder GW, Foreman DI. Nonparametric Statistics for Non-Statisticians:
A Step-by-Step Approach. Hoboken, NJ: Wiley; 2009.
27. Gorey KM, Luginaah IN, Bartfay E, et al. Effects of socioeconomic
status on colon cancer treatment accessibility and survival in Toronto, Ontario, and San Francisco, California, 1996 to 2006. Am J Public Health.
2011;101(1):112-119.
28. Gorey KM, Luginaah IN, Hamm C, et al. Breast cancer care in Canada and the United States: ecological comparisons of extremely impoverished and affluent urban neighborhoods. Health Place. 2010;16(1):156-163.
29.Smolen JR, Thorpe JR, Jr, Bowie JV, et al. Health insurance
and chronic conditions in low income urban whites. J Urban Health.
2014;91(4):637-647.
30.Magge H, Cabral HJ, Kazis LE, et al. Prevalence and predictors of underinsurance among low-income adults. J Gen Intern Med.
2013;28(9):1136-1142.
31. Wharam JF, Ross-Degnan D, Rosenthal MB. The ACA and high-deductible insurance—strategies for sharpening a blunt instrument. New Engl
J Med. 2013;369(16):1481-1484.
32. Maar M, Burchell A, Little J, et al. A qualitative study of provider
perspectives of structural barriers to cervical cancer screening among first
nations women. Womens Health Issues. 2013;23(5):e319-325.
33. Walsemann KM, Gee GC, Ro A. Educational attainment in the
context of social inequality: new directions for research on education and
health. Am Behav Sci. 2013;57(8):1082-1104.
34. Bowleg L. The problem with the phrase women and minorities: Intersectionality-an important theoretical framework for public health. Am J
Public Health. 2012;102(7):1267-1273.
35. Gordon Nembhard J, Chiteji N, eds. Wealth Accumulation and
Communities of Color in the United States: Current Issues. Ann Arbor, MI:
University of Michigan Press; 2006.
Cancer Control 161
36. Conley D. Being Black, Living in the Red: Race, Wealth, and Social
Policy in America. Los Angeles: University of California Press; 1999.
37. West C. Race Matters. Boston, MA: Beacon Press; 1993.
38. Yin D, Morris CR, Bates JH, et al. Effect of misclassified underlying cause of death on survival estimates of colon and rectal cancer. J Natl
Cancer Inst. 2011;103(14):1130-1133.
39. Hall S, Schulze K, Groome P, et al. Using cancer registry data for survival studies: the example of the Ontario Cancer Registry. J Clin Epidemiol.
2006;59(1):67-76.
40. Lenfant C, Freidman L, Thom T. Fifty years of death certificates: the
Framingham Heart Study. Ann Intern Med. 1998;129(12):1066-1067.
41. Brown BW, Brauner C, Minnotte MC. Noncancer deaths in white
adult cancer patients. J Natl Cancer Inst. 1993;85(12):979-987.
42. Maskarinec G, Sen C, Koga K, et al. Ethnic differences in breast cancer survival: status and determinants. Womens Health. 2011;7(6):677-687.
162 Cancer Control
April 2016, Vol. 23, No. 2
Infectious Diseases Report
Bordetella pertussis Infection in Patients With Cancer
Abraham Yacoub, MD, Sowmya Nanjappa, MD, Tyler Janz, and John N. Greene, MD
Summary: We illustrate 2 cases of pneumonia associated with Bordetella pertussis infection in 72-year-old
and 61-year-old patients with cancer receiving myelosuppressive therapy after hematopoietic stem cell transplantation. Bacterial infections are a significant cause of morbidity and mortality in patients with cancer, and
those receiving hematopoietic stem cell transplant, solid organ transplant, or myelosuppressive therapy are at
increased risk. The infection was detected and the 2 patients had good outcomes following azithromycin treatment. Pertussis, also known as whooping cough, is a contagious respiratory illness that has become a public
health challenge due to decreased immunity of the pertussis vaccine. Therefore, it is critical to recognize pertussis
early in the course of the disease.
Introduction
The lung is one of the most frequently involved organs in a variety of complications in the immunocompromised host.1 Among the pulmonary complications
that occur in persons who are immunocompromised,
infection is the most common and is associated with
high rates of morbidity and mortality.1 The most commonly encountered type of infection is bacterial in
origin.2 Before the development of vaccines, Bordetella pertussis infection was a significant threat among
immunocompetent hosts.3 Bordetella vaccination has
significantly reduced the number of infections, but immunity appears to be short lived.4 In addition, a large
portion of the population remains susceptible to infection from B pertussis.5 We present 2 cases of B pertussis
infection that led to pneumonia in patients with cancer.
The medical staff members at our institution were up
to date on their vaccinations, and no secondary cases
of pertussis were reported.
Case Reports
Case 1
A woman aged 72 years with metastatic ovarian cancer who was receiving chemotherapy with intrathecal
methotrexate, gemcitabine, and carboplatin presented
with a 12-day history of productive cough with yelFrom the Division of Infectious Diseases (AY, JNG) and International Medicine (AY), Departments of Internal Medicine (SN) and
Oncologic Sciences (SN), H. Lee Moffitt Cancer Center & Research
Institute, and the Morsani College of Medicine (AY, SM, JNG), Department of Biomedical Sciences (TJ), University of South Florida,
Tampa, Florida.
Submitted January 20, 2016; accepted March 5, 2016.
Address correspondence to John N. Greene, MD, Department of
Infectious Diseases, Moffitt Cancer Center, 12902 Magnolia Drive,
Tampa, FL 33612-9497. 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.
April 2016, Vol. 23, No. 2
low sputum and shortness of breath associated with
the coughing spells. She had tried a cough suppressant
with minimal relief. She denied fever, chills, sweats,
hemoptysis, ill contacts, and recent travel.
Her vital signs were within normal limits, and
findings on physical examination were significant for
crackles heard throughout the lung fields. Her white
blood cell count was 4,910/µL with a normal differential. The patient denied receiving pneumocystis prophylaxis in the past and tested negative for Pneumocystis jiroveci.
The result from a nasopharyngeal swab sent for
a respiratory viral polymerase chain reaction (PCR)
panel was positive for B pertussis. PCR testing was
negative for adenovirus, Chlamydia pneumoniae,
coronavirus, metapneumovirus, rhinovirus, enterovirus, influenza types A and B, Mycoplasma, parainfluenza virus types 1 to 4, and respiratory syncytial virus.
Computed tomography (CT) of the chest showed dependent areas of subpleural consolidation and patchy,
ground-glass opacities (Fig 1).
She was discharged home with a 5-day course of
oral azithromycin (250 mg daily). Repeat PCR testing
1 week later was negative for B pertussis. She reported
improvement of her symptoms 3 weeks later.
Case 2
A man aged 61 years admitted to our hospital with a
5-year history of chronic lymphocytic leukemia and
a matched, unrelated donor stem cell transplantation
12 days prior complained of a 1-day history of nonproductive cough, sore throat, and fatigue. He denied
fever, chills, shortness of breath, hemoptysis, nausea,
vomiting, ill contacts, and recent travel.
His vital signs were all within normal limits. Finding on physical examination was remarkable for coarse
breath sounds heard across the left lung fields. His
white blood cell count was 560 µL, absolute neutrophil
Cancer Control 163
Fig 1. — Computed tomography of the chest showing dependent areas of subpleural consolidation and patchy, ground-glass opacities.
count was 190 µL, and lymphocyte count was 200 µL.
The patient was receiving pneumocystis prophylaxis
and tested negative for P jiroveci.
The result from a nasopharyngeal swab sent for a
respiratory viral PCR panel was positive for B pertussis.
PCR testing was negative for adenovirus, C pneumoniae, coronavirus, metapneumovirus, rhinovirus, enterovirus, influenza types A and B, Mycoplasma, parainfluenza virus types 1 to 4, and respiratory syncytial virus.
Radiography of his chest showed an interval increase
in opacification of the perihilar lungs. CT of his chest
demonstrated patchy, bilateral airspace disease and
ground-glass nodularity, which was greatest in the left
lower lobe and bilateral small pleural effusions (Fig 2).
Rapid strep testing was negative, and blood cultures
did not show any growth after 3 days.
He was treated with a 7-day course of oral azithromycin (250 mg daily) and was followed-up in the clinic. He reported complete resolution of his symptoms
2 weeks later. Repeat PCR testing for B pertussis performed on day 26 was negative.
Discussion
B pertussis, a gram-negative aerobic, pleomorphic
coccobacilli, is known to cause whooping cough,
which is a respiratory tract infection characterized
by paroxysmal cough.6-9 B pertussis is transmitted by
large droplets produced during coughing, sneezing,
or talking. These droplets generally travel fewer than
3 feet and can be deposited on mucosal surfaces of
susceptible individuals.10 B pertussis belongs to the
Bordetella genus, which now includes 9 species, and
B pertussis, B parapertussis, and B bronchiseptica are
the 3 most studied.11
Infection is initiated by the attachment of B pertussis to the cilia of epithelial cells of the upper respi164 Cancer Control
Fig 2. — Computed tomography of the chest demonstrating patchy,
bilateral airspace disease and ground-glass nodularity (greatest in the
left lower lobe).
ratory tract. Evasion of host defenses is facilitated by
adenylate cyclase toxin (CyaA) and pertussis toxin.12
CyaA enters the neutrophils and catalyzes the excessive production of cyclic adenosine monophosphate,
which intoxicates the cells such that phagocytosis
is compromised. Similar to CyaA, pertussis toxin adversely affects phagocytosis and inhibits the migration of lymphocytes and macrophages to areas of infection.12 Local tissue damage of the ciliated epithelial
cells may be due to tracheal cytotoxin, dermonecrotic
toxin, and perhaps CyaA.12
Although B pertussis mainly causes pulmonary
issues, it has been documented to be associated with
otitis media,13 encephalopathy,14 bacteremia,15 hemolytic uremic syndrome,16-18 infantile hypertrophic
pyloric stenosis,19 multiple sclerosis, amyotrophic
April 2016, Vol. 23, No. 2
lateral sclerosis,01 cerebral ataxia, 21 and Wegener
granulomatosis.22
Cough is one of the most common complaints
among patients with B pertussis infection.23 Diagnosis
can be made by PCR assay because it is a rapid, efficient, and specific technique with superior sensitivity
compared with that of culture.24-26
Five other cases have been cited in the literature of
pertussis in patients with cancer and a mix of hematological malignancies and solid tumors (Table).15,27-30 It
is worthy of note that these cases were published prior the advent of PCR assay and several were based on
culture of the organism from extrapulmonary sources
(eg, blood, empyema fluid).15,27-30
The recommended antimicrobial agents for the
treatment or chemoprophylaxis of pertussis in patients
who are immunocompromised or immunocompetent
are azithromycin, clarithromycin, and erythromycin.31
Trimethoprim/sulfamethoxazole can also be used.32
Erythromycin, clarithromycin, and azithromycin are
preferred for the treatment of pertussis in persons who
are at least 1 month of age.31 For infants younger than
1 month of age, azithromycin is preferred for postexposure prophylaxis and treatment because azithromycin has not been associated with infantile hypertrophic
pyloric stenosis, unlike erythromycin.33,34 Azithromycin should be used with caution because it is associ-
ated with prolongation of the QT interval.35 For patients allergic or intolerant to these agents, doxycycline
or levofloxacin is an alternative option (rather than no
treatment).35 If symptoms do not improve after a trial
of macrolides, then resistance should be suspected.36,37
Use of vaccination against B pertussis has been
well studied.38 The diphtheria, tetanus, and acellular
pertussis (DTaP) vaccine is indicated only for children
younger than 7 years of age.38 However, the adult tetanus, diphtheria, and acellular pertussis (Tdap) vaccine
is a 3-dose series with high tetanus and pertussis content that may be more immunogenic in recipients of
hematopoietic stem cell transplantation, so it should
be considered as the initial vaccination for this patient
population, regardless of age.38 Furthermore, Suzuki
et al30 suggested that acellular pertussis vaccine can be
considered for patients after receiving hematopoietic
stem cell transplantation, even if they are adults or older than 7 years of age.
Conclusions
We presented 2 patients who had good outcomes following azithromycin treatment. Due to the advent of
and availability of polymerase chain reaction–based
diagnostic testing, the rate of Bordetella pertussis infection among patients who are immunocompromised
and have cancer is expected to increase.39 Such test-
Table. — Studies of Bordetella pertussis Infection in Patients With Cancer
Study
Age, y
Sex
Malignancy
Symptoms
Source of Culture/
PCR
Treatment
Outcome
Case 1
72
F
Ovarian cancer
Productive cough
Shortness of breath
Pharynx
PCR assay
Azithromycin
Improved
Case 2
61
M
Chronic lymphocytic
leukemia
Nonproductive cough
Sore throat
Fatigue
Pharynx
PCR assay
Azithromycin
Improved
Florax 27
16
M
ALL
Nonproductive cough
Pharynx
PCR
Roxithromycin
Symptoms resolved
after 2 mo of
treatment
MacLean29
—
—
Bronchogenic lung
cancer
—
—
—
—
Senturk 28
64
F
Non–small-cell lung
cancer
Dyspnea
Chest pain
Empyema
PCR assay
Erythromycin 2 mg/d
Improved
Suzuki30
10
M
ALL
Dry cough
Sleep disturbances
Plasma serum
PCR assay
Cephalosporin
Clarithromycin
Poor
Troseid15
63
M
Multiple myeloma
Cough
Hoarseness
Fever
Blood culture
Clarithromycin
Poor
ALL = acute lymphoblastic leukemia, F = female, M = male, PCR = polymerase chain reaction.
April 2016, Vol. 23, No. 2
Cancer Control 165
ing can be easily done via a nasopharyngeal swab.
The literature is scant regarding pertussis in
adults — and this is particularly true among those
with cancer — due to the historical difficulty of accurate diagnostic testing.39
References
1. Oh YW, Effmann EL, Godwin JD. Pulmonary infections in immunocompromised hosts: the importance of correlating the conventional radiologic appearance with the clinical setting. Radiology. 2000;217(3):647-656.
2. Conces DJ Jr. Bacterial pneumonia in immunocompromised patients.
J Thorac Imaging. 1998;13(4):261-270.
3. Hewitt M, Canning BJ. Coughing precipitated by Bordetella pertussis
infection. Lung. 2010;188(suppl 1):S73-S79.
4. Ward JI, Cherry JD, Chang SJ, et al; APERT Study Group. Bordetella pertussis infections in vaccinated and unvaccinated adolescents and
adults, as assessed in a national prospective randomized Acellular Pertussis Vaccine Trial (APERT). Clin Infect Dis. 2006;43(2):151-157.
5. Bloom DE. The value of vaccination. Adv Exp Med Biol. 2011;697:1-8.
6. Shumilla JA, Lacaille V, Hornell TM, et al. Bordetella pertussis infection of primary human monocytes alters HLA-DR expression. Infect Immun.
2004;72(3):1450-1462.
7. Skerry CM, Mahon BP. A live, attenuated Bordetella pertussis vaccine provides long-term protection against virulent challenge in a murine
model. Clin Vaccine Immunol. 2011;18(2):187-193.
8. Greenberg D, Bamberger E, Ben-Shimol S, et al. Pertussis is under
diagnosed in infants hospitalized with lower respiratory tract infection in the
pediatric intensive care unit. Med Sci Monit. 2007;13(11):CR475-CR480.
9. Bettiol S, Thompson MJ, Roberts NW, et al. Symptomatic treatment of the cough in whooping cough. Cochrane Database Syst Rev.
2010;(1):CD003257.
10. Sandora TJ, Gidengil CA, Lee GM. Pertussis vaccination for health
care workers. Clin Microbiol Rev. 2008;21(3):426-434.
11. Guiso N. Bordetella pertussis and pertussis vaccines. Clin Infect
Dis. 2009;49(10):1565-1569.
12. Mattoo S, Cherry JD. Molecular pathogenesis, epidemiology, and
clinical manifestations of respiratory infections due to Bordetella pertussis
and other Bordetella subspecies. Clin Microbiol Rev. 2005;18(2):326-382.
13. Decherd ME, Deskin RW, Rowen JL, et al. Bordetella pertussis
causing otitis media: a case report. Laryngoscope. 2003;113(2):226-227.
14. Halperin SA, Marrie TJ. Pertussis encephalopathy in an adult: case
report and review. Rev Infect Dis. 1991;13(6):1043-1047.
15.Trøseid M, Jonassen TØ, Steinbakk M. Isolation of Bordetella
pertussis in blood culture from a patient with multiple myeloma. J Infect.
2006;52(1):e11-e13.
16. Cohen-Ganelin E, Davidovits M, Amir J, et al. Severe Bordetella
pertussis infection associated with hemolytic uremic syndrome. Isr Med
Assoc J. 2012l;14(7):456-458.
17. Chaturvedi S, Licht C, Langlois V. Hemolytic uremic syndrome caused
by Bordetella pertussis infection. Pediatr Nephrol. 2010;25(7):1361-1364.
18. Pela I, Seracini D, Caprioli A, et al. Hemolytic uremic syndrome
in an infant following Bordetella pertussis infection. Eur J Clin Microbiol
Infect Dis. 2006;25(8):515-517.
19. Morrison W. Infantile hypertrophic pyloric stenosis in infants treated
with azithromycin. Pediatr Infect Dis J. 2007;26(2):186-188.
20. Flore D. Diagnosis and treatment of multiple sclerosis and amyotrophic lateral sclerosis: neuropathies from Bordetella pertussis. Am J
Ther. 2003;10(5):377-379.
21. Setta F, Baecke M, Jacquy J, et al. Cerebellar ataxia following
whooping cough. Clin Neurol Neurosurg. 1999;101(1):56-61.
22. Janda WM, Santos E, Stevens J, et al. Unexpected isolation of Bordetella pertussis from a blood culture. J Clin Microbiol. 1994;32(11):2851-2853.
23. Melo N, Dias AC, Isidoro L, et al. Bordetella pertussis, an agent not
to forget: a case report. Cases J. 2009;2(1):128.
24. van der Zee A, Agterberg C, Peeters M, et al. A clinical validation of
Bordetella pertussis and Bordetella parapertussis polymerase chain reaction: comparison with culture and serology using samples from patients with
suspected whooping cough from a highly immunized population. J Infect
Dis. 1996;174(1):89-96.
25. Birkebaek NH, Heron I, Skjødt K. Bordetella pertussis diagnosed by
polymerase chain reaction. APMIS. 1994;102(4):291-294.
26. Gilberg S, Njamkepo E, Du Chatelet IP, et al. Evidence of Bordetella pertussis infection in adults presenting with persistent cough in
a French area with very high whole-cell vaccine coverage. J Infect Dis.
2002;186(3):415-418.
27. Florax A, Ehlert K, Becker K, et al. Bordetella pertussis respiratory
infection following hematopoietic stem cell transplantation: time for universal vaccination? Bone Marrow Transplant. 2006;38(9):639-640.
28. Senturk E, Sen S, Telli M. Empyema due to Bordetella pertussis in
an adult patient with lung cancer [in English, Spanish]. Arch Bronconeumol.
166 Cancer Control
2012;48(7):263-264.
29. MacLean DW. Bordetella pertussis infection in patients with bronchogenic carcinoma. Lancet. 1981;1(8218):503-504.
30. Suzuki N, Mizue N, Hori T, et al. Pertussis in adolescence after unrelated cord blood transplantation. Bone Marrow Transplant. 2003;32(9):967.
31. Dierig A, Beckmann C, Heininger U. Antibiotic treatment of pertussis:
are 7 days really sufficient? Pediatr Infect Dis J. 2015;34(4):444-445.
32. Altunaiji S, Kukuruzovic R, Curtis N, et al. Antibiotics for whooping
cough (pertussis). Cochrane Database Syst Rev. 2007;(3):CD004404.
33. Cooper WO, Griffin MR, Arbogast P, et al. Very early exposure to
erythromycin and infantile hypertrophic pyloric stenosis. Arch Pediatr Adolesc
Med. 2002;156(7):647-650.
34. Santos N, Oliveira M, Galrinho A, et al. QT interval prolongation
and extreme bradycardia after a single dose of azithromycin [in English,
Portuguese]. Rev Port Cardiol. 2010;29(1):139-142.
35. Flanagan MP. How to manage a pertussis outbreak in your practice.
Fam Pract Manag. 2005;12(7):31-34.
36. Guillot S, Descours G, Gillet Y, et al. Macrolide-resistant Bordetella
pertussis infection in newborn girl, France. Emerg Infect Dis. 2012;18(6):966-968.
37. Lewis K, Saubolle MA, Tenover FC, et al. Pertussis caused by an
erythromycin-resistant strain of Bordetella pertussis. Pediatr Infect Dis J.
1995;14(5):388-391.
38. Rubin LG, Levin MJ, Ljungman P, et al; Infectious Diseases Society of
America. 2013 IDSA clinical practice guideline for vaccination of the immunocompromised host [Erratum appears in Clin Infect Dis. 2014;59(1):144].
Clin Infect Dis. 2014;58(3):e44-e100.
39. McGuiness CB, Hill J, Fonseca E, et al. The disease burden of pertussis in adults 50 years old and older in the United States: a retrospective
study. BMC Infect Dis. 2013;13:32.
April 2016, Vol. 23, No. 2
Pharmacy Report
Megestrol Acetate–Induced Adrenal Insufficiency
Sowmya Nanjappa, MD, Christy Thai, PharmD, Sweta Shah, MD, and Matthew Snyder, PharmD
Summary: A man aged 65 years with metastatic renal cell carcinoma presented for evaluation after a recent
fall. A thorough workup of the case was performed and secondary adrenal insufficiency induced by the administration of megestrol acetate was determined to be the cause. Adrenal insufficiency is a serious disorder that is
a potential adverse event of megestrol acetate, a medication used to help patients with cancer cachexia increase
their appetite and gain weight. This association is not well recognized in clinical practice, so this case highlights
the importance of distinguishing possible endocrine complications induced by the long-term administration or
sudden discontinuation of megestrol acetate.
Introduction
Adrenal insufficiency is a serious complication that is
a potential adverse event of megestrol acetate, a medication used to help patients with cancer cachexia increase their appetite and gain weight.1,2 However, this
association is not well recognized in clinical practice.3
Case Report
A man aged 65 years with hypertension and stage 4
clear cell renal cell carcinoma (RCC) with metastases
to the brain, lungs, bones, and pancreas was admitted for evaluation after a recent fall. His past surgical
history was significant for left nephrectomy and adrenalectomy nearly 20 years ago. After his RCC did not
respond to sunitinib and pazopanib treatment, his chemotherapy regimen was changed about 1 month ago to
twice daily oral axitinib (5 mg). During his admission,
he was hypotensive and his mental status was altered.
Vitals were blood pressure of 71/49 mm Hg, respiratory rate of 18 breaths/minute, heart rate of
101 beats/minute, temperature of 98.8 °F, and oxygen
saturation of 96% on room air. Findings on physical examination were unremarkable, except for tenderness
and swelling in his left lower extremities secondary to
the recent fall. Findings on complete blood count with
a differential and comprehensive metabolic panel were
all within normal limits. Findings following a workup
for sepsis and brain imaging were negative.
After ruling out potential causes, including infecFrom the Departments of Internal Medicine (SN), Pharmacy (CT,
MS), H. Lee Moffitt Cancer Center & Research Institute, and University of South Florida Morsani College of Medicine (SS), Tampa,
Florida.
Submitted February 26, 2016; accepted March 4, 2016.
Address correspondence to Sowmya Nanjappa, MD, Moffitt
Cancer Center, 12902 Magnolia Drive, 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.
April 2016, Vol. 23, No. 2
tion, brain metastases, dehydration, electrolyte imbalances, and hypothyroidism, a workup for adrenal
insufficiency was performed because hypotension
persisted while his mental status improved. A morning serum cortisol level was below 1.0 µg/dL. A cosyntropin stimulation test revealed serum cortisol
levels of 5.9 and 8.9 µg/dL at 30 and 60 minutes, respectively, with a pretest plasma adrenocorticotropic
hormone (ACTH) level below 5 pg/mL, indicating secondary adrenal insufficiency.
A review of the patient’s medication list unveiled
no agents commonly known to induce adrenal insufficiency with one exception. He had recently been
treated with daily megestrol acetate (oral suspension
625 mg) for about 4 weeks prior to his current admission, and this dose had been continued for the
initial portion of this hospitalization. We suspected
that this agent might be the likely culprit, so it was
discontinued.
Oral hydrocortisone 20 mg every morning/10 mg
every evening was initiated for adrenal insufficiency, and his blood pressure significantly improved.
Steroids were discontinued prior to a follow-up appointment 3 months following discharge. Repeat serum cortisol level and ACTH value were 20 µg/dL
and 68 pg/mL, respectively; these results confirmed
resolution of the adrenal insufficiency secondary to
megestrol acetate.
Discussion
Adrenal insufficiency is a serious, potentially
life-threatening condition that stems from primary adrenal failure or secondary adrenal disease owing to an
impaired hypothalamic–pituitary–adrenal (HPA) axis.1
The disorder can chronically cause fatigue, anorexia,
weakness, abdominal pain, and hypotension, whereas
the acute syndrome may induce hypotensive crisis, fever, clouded sensorium, and myopathy.1 Those affected
typically require stress-dose steroids for surgical proCancer Control 167
cedures or severe illness and glucocorticoid replacement on a long-term basis.
Although management of the disorder is relatively straightforward, making an accurate and timely
diagnosis often proves to be challenging in the oncology setting. Patients with cancer frequently experience these symptoms, sometimes as a result of disease
progression, surgery, radiation, chemotherapy, or a
combination of all of these. Oftentimes, cachexia seen
over time with adrenal insufficiency can mimic wasting characteristic of metastatic disease.4 Paraneoplastic
syndromes and malnutrition may cause severe electrolyte abnormalities, clouding, or covering up of the
evidence of the role of adrenal insufficiency in such irregularities.4
In addition to causing adrenal insufficiency–associated symptoms, such as fatigue, weakness, myopathy, and pain, chemotherapy and immunotherapy
agents can directly give rise to the disorder.5-7 The tyrosine kinase inhibitors sunitinib and imatinib and, more
frequently, the checkpoint-blocking immunotherapies
ipilimumab, nivolumab, and pembrolizumab have
been reported to cause adrenal insufficiency.5-7 Given
the limited data available, it will be interesting to see
whether the effects of these agents on adrenal function
extend to other members of their respective classes as
their use continues to increase.
Medications commonly used as supportive care
can also induce adrenal insufficiency.1,7-10 The detrimental impacts of the long-term administration and
abrupt discontinuation of steroids on adrenal function
are well known.1 Use of glucocorticoids is also common in patients with cancer, and these agents play a
critical role in the treatment of various hematological
malignancies, immune-related adverse events,7,8 the relief of symptoms secondary to brain metastases,9 and
pain management.10
Similarly, megestrol acetate, which is a synthetic
progestin with antiestrogenic properties, is often used
to help patients with cancer cachexia increase their appetite and gain weight.2 Secondary to its inherent glucocorticoid-like effect,2 the agent is thought to cause
suppression of the HPA axis at the hypothalamus
through negative feedback, thus resulting in low levels
of serum cortisol and plasma ACTH.11,12
Although stimulating the appetite through the
use of megestrol acetate was useful in a patient like
ours, megestrol acetate has been associated with
thrombosis, hyperglycemia, hypertension, osteoporosis, hypogonadism, and adrenal insufficiency.3 Its
clinical benefit is often outweighed by these potential adverse events, many of which are already at a
heightened risk of occurring in this patient population (eg, thrombosis). Several questions regarding
the length of time needed for megestrol acetate–induced adrenal suppression and subsequent restora168 Cancer Control
tion of HPA axis function after discontinuation of
the drug are still unknown.
Leinung et al13 conducted a trial that initiated 80
mg megestrol acetate 3 times a day in study patients
with normal cortisol and ACTH levels prior to therapy.
After 1 month, both values significantly decreased,
and study participants also displayed a significantly
decreased response to cosyntropin stimulation.13 An
interventional trial found that 6 of 7 study patients
with adrenal suppression recovered normal function
within 2 weeks, with the remaining patient doing so
in 6 weeks (NCT00575029). However, general data are
lacking, so definitive timeframes are uncertain. Nevertheless, our case describes a patient who developed
treatment-related adrenal insufficiency after 1 month
of therapy, but successfully recovered in 3 months after
the medication was discontinued.
Although adrenal insufficiency is a reported potential adverse event of megestrol acetate, it is not well
recognized in clinical practice.3 Adrenal suppression
can stem from abrupt discontinuation of the agent.
However, as in our case, megestrol acetate has been
found to cause this effect in patients actively taking the
agent, thus complicating the diagnosis.2,14,15
Postulated mechanisms for the latter scenario
include a dual agonist–antagonist action (binding to
the glucocorticoid receptor while simultaneously preventing endogenous glucocorticoids from doing so), a
greater tendency to suppress the HPA axis rather than
induce glucocorticoid-like effects, and concurrent
acute stress or illness in a patient whose HPA axis is
already suppressed.11,15 Retrospectively, the patient
was not taking any other medication likely to cause
such an effect.
The origin of our patient’s adrenal insufficiency
was also clouded by numerous factors, including prior
left adrenalectomy, tyrosine kinase inhibitor use, and
metastatic RCC. This case of megestrol-induced adrenal insufficiency was only discovered after extensive
workup and elimination of other possible causes.
Conclusions
Although obtaining and analyzing a patient’s medication history may be challenging, thorough and
accurate assessment of previous and current medication use is a necessity that proved to be pivotal in
this case. Given the potentially severe complications
of adrenal insufficiency, this report underscores the
importance of recognizing possible endocrine complications induced by the long-term administration
or sudden discontinuation of megestrol acetate. However, further study is still needed to draw definitive
conclusions on the effects of megestrol acetate on adrenal function.
Acknowledgment: We thank the patient for his
cooperation throughout the duration of the case.
April 2016, Vol. 23, No. 2
References
1. Bornstein SR. Predisposing factors for adrenal insufficiency. N Engl
J Med. 2009;360(22):2328-2339.
2. Delitala AP, Fanciulli G, Maioli M, et al. Primary symptomatic adrenal insufficiency induced by megestrol acetate. Neth J Med. 2013;71:17-21.
3. Dev R, Del Fabbro E, Bruera E. Association between megestrol acetate treatment and symptomatic adrenal insufficiency with hypogonadism
in male patients with cancer. Cancer. 2007;110(6):1173-1177.
4. Yeung SC, Escalante CP, Gagel RF. Medical Care of Cancer Patients.
Shelton, CT: BC Decker; 2009.
5. Charmandari E, Nicolaides NC, Chrousos GP. Adrenal insufficiency. Lancet. 2014;383(9935):2152-2167.
6. Bilgir O, Kebapcilar L, Bilgir F, et al. Is there any relationship between imatinib mesylate medication and hypothalamic-pituitary-adrenal
axis dysfunction? Int J Clin Pract. 2010;64(1):45-50.
7. Villadolid J, Amin A. Immune checkpoint inhibitors in clinical practice: update on management of immune-related toxicities. Transl Lung
Cancer Res. 2015;4(5):560-575.
8. Sionov RV, Spokoini R, Kfir-Erenfeld S, et al. Mechanisms regulating the susceptibility of hematopoietic malignancies to glucocorticoidinduced apoptosis. Adv Cancer Res. 2008;101:127-248.
9. Ryken TC, McDermott M, Robinson PD, et al. The role of steroids in
the management of brain metastases: a systematic review and evidencebased clinical practice guideline. J Neurooncol. 2010;96(1):103-114.
10. Leppert W, Buss T. The role of corticosteroids in the treatment of
pain in cancer patients. Curr Pain Headache Rep. 2012;16(4):307-313.
11. Mann M, Koller E, Murgo A, et al. Glucocorticoidlike activity of
megestrol. A summary of Food and Drug Administration experience and a
review of the literature. Arch Intern Med. 1997;157(15):1651-1656.
12. Naing KK, Dewar JA, Leese GP. Megestrol acetate therapy and
secondary adrenal suppression. Cancer. 1999;86(6):1044-1049.
13.Leinung MC, Liporace R, Miller CH. Induction of adrenal suppression by megestrol acetate in patients with AIDS. Ann Intern Med.
1995;122(11):843-845.
14.González Villarroel P, Fernández Pérez I, Páramo C, et al. Megestrol
acetate-induced adrenal insufficiency. Clin Transl Oncol. 2008;10(4):235-237.
15. Mehta K, Weiss I, Goldberg MD. Megace mystery: a case of central
adrenal insufficiency. Case Rep Endocrinol. 2015;2015:147265.
April 2016, Vol. 23, No. 2
Cancer Control 169
Pathology Report
Presacral Noncommunicating Enteric Duplication Cyst
Shabnam Seydafkan, MD, David Shibata, MD, Julian Sanchez, MD, Nam D. Tran, MD, Marino Leon, MD,
and Domenico Coppola, MD
Background: Gastrointestinal (GI) tract duplication cysts or enteric duplication cysts are rare congenital
malformations sometimes found on the mesenteric aspect of segments of the alimentary tract. Enteric duplication cysts are lined by normal GI epithelium and may be classified as foregut, mid-gut, and hindgut cysts.
Except in very rare cases of retroperitoneal enteric duplication cysts, these cysts communicate with the GI tract
and share a common blood supply. Concurrent congenital malformations are not uncommon and malignant
transformation within enteric duplication cysts has also been reported.
Methods: We describe a case of a noncommunicating enteric duplication cyst in a 52-year-old woman.
Results: The patient presented with a presacral cystic mass requiring frequent drainage procedures that was
primarily believed to be of neural origin. Upon resection, the lesion contained heterotopic tissue, including
ciliated bronchial epithelium, squamous and transitional epithelia, and pancreatic and gastric tissue. Focal,
low-grade intestinal adenoma was present, but malignancy was not detected in this case.
Conclusion: To our knowledge, this is the sixth reported case of a noncommunicating enteric duplication cyst
in the English medical literature.
Introduction
An enteric duplication cyst is a cystic or tubular congenital malformation located on the mesenteric side of the
gastrointestinal (GI) tract that firmly attaches to or shares
the gut wall. Theories regarding the origin of GI duplications include diverticulization, canalization defects, intrauterine vascular accident, split notochord theory, and
the traction hypothesis between the intestinal endoderm
and the overlying structures.1-4 Enteric duplication cyst is
rare, with a prevalence of 1 out of 4,500 individuals, and
it may originate from anywhere in GI tract.5-7
Enteric duplication cysts are lined with epithelium
similar to that of the adjacent normal GI mucosa and
are classified by location as foregut, midgut, or hindgut.5,8 In order of decreasing frequency, the most common sites of enteric duplication cysts are the ileum,
stomach, and appendix, with the rarest site being the
gastroduodenal region.9,10 Criteria to establish the diagnosis of enteric duplication cyst include the presence of
a double-walled cyst, simulating the normal anatomy
From the Departments of Anatomic Pathology (SS, ML, DC), Gastrointestinal Oncology (DS, JS), Chemical Biology & Molecular
Medicine Program (NDT, DC), Tumor Biology Program (DC),
H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida.
Address correspondence to Domenico Coppola, MD, Department of
Anatomic Pathology, Moffitt Cancer Center, 12902 Magnolia Drive,
Tampa, FL 33612. E-mail: [email protected]
Submitted March 11, 2015; accepted January 15, 2016.
No significant relationships exist between the authors and the companies/organizations whose products or services may be referenced
in this article.
Dr Shibata is now with Department of Surgery at the University of
Tennessee Health Science Center, Memphis, Tennessee.
170 Cancer Control
of the intestine, lined with enteric-type epithelium, and
sharing a common blood supply with its neighboring
alimentary tract.11 Rare subtypes of retroperitoneal enteric duplication cysts do not communicate with the intestine and have their own exclusive blood supply.2,12
A patient’s presenting symptoms depend on the
anatomical location, size, and the inner lining of the
cyst and may include abdominal pain, rectal bleeding,
and hematuria. Enteric duplication cyst may also be incidentally discovered in asymptomatic patients. However, enteric duplication cysts become symptomatic in
a person’s early childhood and present in the newborn
period as an abdominal mass. Few adulthood cases of
enteric duplication cyst have been reported.8,11,13
Intussusception, volvulus, or intestinal obstruction
and, in rare cases, malignant transformation may also
occur.8,14 In more than 50% of cases, other associated
malformations such as vertebral defects and other GI
duplications may be present.10 Ultrasonography, computed tomography (CT), and magnetic resonance imaging (MRI) remain the main diagnostic tools.14
Herein we present a case of enteric duplication
cyst initially believed to be of neural origin. In addition, we provide a review of the literature for similar
cases and discuss the diagnostic challenges presented
by enteric duplication cysts.
Case Report
A 52-year-old woman presented with a history of a cystic presacral mass that was partially and initially resected via an abdominal approach in 2006. She sustained
multiple recurrences, which were all managed by percuApril 2016, Vol. 23, No. 2
taneous image-guided drainage. She was subsequently
referred to our treatment team approximately 7 years after her initial diagnosis with worsening rectal pain.
Findings on both CT and pelvic MRI revealed a
posterior pelvic bilobed cystic mass extending from
the puborectalis posteriorly to the sacrum and coccyx
and displacing the rectum anteriorly (Fig 1). The lesion
also involved part of the levator muscle and had 2 cystic components measuring 3.4 and 3.2 cm, respectively.
The lesion showed high-signal intensity on T1-weighted sequences and somewhat low-signal intensity on
T2-weighted sequences. The lesion did not show suspicious enhancement, and the vagina and bladder were
not involved.
The decision was made to proceed with surgical
resection via a local, transcoccygeal Kraske approach.
To allow for intraoperative identification of rectothecal
communication (in the event of pseudomeningocele),
preoperative lumbar puncture with intrathecal injection of fluorescein dye was performed.
During surgery, a large cystic mass to the left of the
midline crossing anterior to the coccyx in a dumbbell
shape was identified. The mass was adherent to the
posterior rectal wall and appeared to caudally extend to
the level of the external sphincter. It was completely excised by the Kraske approach. The cystic mass had no
communication with the rectum or the thecal sac.
Gross examination revealed an irregular fragment
of fibrous and adipose tissue measuring 7 × 6 × 3 cm
and weighing 77 g. Upon sectioning the specimen,
3 cystic lesions were found, ranging from 3 to 3.5 cm
in their greatest dimensions. Two of the cysts were
previously opened and were connected to each other.
The third cyst was independent and contained a large
amount of tan-brown colored viscous material. The
inner walls of the cysts were pink-tan to dusky red in
color, focally erythematous and hemorrhagic, and averaged less than 0.1 cm in thickness. The fibroadipose
tissue surrounding the cysts was grossly unremarkable.
Routine hematoxylin and eosin stain was done,
and immunohistochemical stains were obtained with a
BenchMark automated platform (Ventana Medical Systems, Tucson, Arizona) using the ultra-view DAB Detection Kit (Ventana) and antibodies cytokeratin (CK)
CK7, CK20, transcription termination factor 1 (TTF1),
p63, smooth muscle myosin heavy chain, and GATA
binding protein 3. A trichrome special stain was also
performed. Immunostain for p16 and synaptophysin
were not contributory. The immunostains for p53 and
Ki67 were also performed.
Fig 1. — Axial magnetic resonance imaging showing a presacral dumbbell shape lesion with high signal on T1-weighted images (T1 FL 2D FS).
A
B
Results
Histopathology examination showed the cysts to be
lined by intestinal-type epithelium with focal adenomatous changes surrounded by a thick muscle coat
(Fig 2). The lesions demonstrated heterotopic tissue,
April 2016, Vol. 23, No. 2
Fig 2A–B. — (A) Histology of the entire cyst at low power (H & E, × 10).
(B) Component of the cyst lined by adenomatous colonic-type epithelium
(H & E, × 100). H & E = hematoxylin and eosin.
Cancer Control 171
including ciliated bronchial epithelium (Fig 3A), squamous and transitional epithelia (Figs 3B and 3C), and
pancreatic and gastric tissue (Fig 3D).
The adenomatous epithelium and the adjacent
intestinal mucosa were positive for CK7 and negative
for CK20. The TTF1 immunostain was negative. The
squamous and urothelial epithelia were positive for
p63 (Fig 4). The inner and outer smooth-muscle layers
surrounding 1 of the cysts was highlighted by smoothmuscle myosin and trichrome special stain. Ki-67 and
p53 immunostains did not support high-grade dysplasia or invasive carcinoma.
Findings after trichrome special stain, which was
used to highlight the layers of muscularis propria, supported the final diagnosis (Fig 5).
of the location and include proximity to the GI tract, a
2-layered coat of well-developed smooth muscle, and a
GI-type epithelium.15,16
Discussion
Enteric duplication cysts are rare congenital lesions
that can originate from anywhere in the GI tract from
the oropharynx to the anus, including the retrorectal
and presacral space.1-4 The key diagnostic features for
enteric duplication cysts remain the same regardless
Fig 4. — p63 immunostain marking the squamous epithelium (immunohistochemistry, × 400).
A
B
C
D
Fig 3A–D. — Heterotopic components within the cyst. (A) Ciliated epithelium (H & E, × 400). (B) Squamous epithelium (H & E, × 200). (C) Mucinous
metaplasia (H & E, × 400). (D) Pancreatic ectopia (H & E, × 200). H & E = hematoxylin and eosin.
172 Cancer Control
April 2016, Vol. 23, No. 2
Fig 5. — Trichrome special stain highlighting the layers of muscularis
propria, supporting the final diagnosis (immunohistochemistry, × 400).
Rectal duplication cysts belong to the family of developmental cysts and usually present as unilocular
cystic masses located in the prerectal space.17 They are
lined with intestinal or respiratory epithelium and are
associated with well-defined muscle layers and myenteric plexus.17
Enteric duplication cysts may have various types
of presenting features.18 For example, enteric duplication cysts found in the duodenum may have communication with the biliary tract or the pancreatic duct.6
Those of gastric origin may involve the pancreas and
may contain heterotopic tissues such as gastric mucosa, pancreatic tissue, or bronchial ciliated epithelium,
or all 3 types, as was observed in our case.19-21 Correlating the radiological findings with the histopathological
features is useful for making the correct diagnosis of
enteric duplication cyst.
In our case, the enteric duplication cysts did not
communicate with the GI tract. Such noncommunicating, isolated enteric duplication cysts have GI
epithelium and a wall similar to that seen in regular
enteric duplication cysts but without an anatomical
association with the alimentary tract. This feature has
previously been reported, but it is rare: Approximately
8 cases of this type have been reported in the English
literature.20 Of the cases reported, 2 were infected
cases of isolated duplication cysts. One was an isolated enteric duplication cyst with features of a classic enterogenous duplication cyst, and 1 case showed
mucinous cystadenoma arising within an isolated duplication cyst of the ileum.20,21
Differential Diagnosis
Clinically, the differential diagnosis of a retrorectal
mass is broad and includes both benign and malignant
entities ranging from inflammatory, congenital or developmental cysts, neurogenic and osseous lesions like
chordoma, or anterior sacral meningocele.16,22
April 2016, Vol. 23, No. 2
Retrorectal developmental cysts include different
types of lesions such as duplication cysts, tailgut cysts,
epidermoid cysts, dermoid cysts, and teratomas. Retrorectal developmental cysts are rare and may be misdiagnosed as a supralevator abscess or a complex anal
fistula.23 A retrorectal mass with a major cystic component is highly suggestive of a developmental disorder,
and the epidermoid, dermoid, and rectal duplications
cysts are more often unilocular.24
On CT, tailgut cysts appear as retrorectal masses
with well-defined smooth borders and uneven attenuation. Calcifications and invasion of surrounding tissues
are uncommon findings for tailgut cysts.25 Tailgut cysts
have a female predominance, whereas enteric duplication cysts are equally present in both sexes.15,16
In general, duplication cysts are unilocular developmental cysts with GI- or respiratory-type epithelium.
The GI epithelium reveals villi, crypts, and glands that
replicate normal gut mucosa. By contrast, tailgut cysts
are usually multicystic or multiloculated, lined with a
variety of stratified squamous, transitional, stratified
columnar, ciliated pseudostratified columnar, and gastric-type mucosa. However, when seen in conjunction
with enteric duplication cysts, the mucosa usually is associated with a regular and continuous 2-layer muscle
coat containing a nerve plexus. This feature is characteristic of duplication cysts and is not seen in tailgut
cysts, where the smooth muscle is disorganized and focally well formed.17,25-30
Cuboidal, transitional, or columnar epithelium can
be seen in enterogenous cysts and tailgut cystic hamartomas. Oftentimes, they are multilocular, and disorganized smooth-muscle fibers can be identified surrounding their thin walls.22
A developmental cyst with stratified squamous
epithelium and skin appendages is suggestive of a dermoid cyst, whereas no skin appendages are seen in the
epidermoid subtype. These cysts lack smooth muscle
in their walls.
Teratomas are also included in the differential diagnosis of developmental retrorectal cysts. Teratomas
contain heterogeneous structures from all 3 embryonic
layers. Mature teratomas manifest as complex, well-circumscribed cystic and solid masses with calcifications
along with epithelial lining, skin adnexa, heterologous
mesenchymal tissues, and neural tissues. Primitive endoderm, mesoderm, and ectoderm are characteristic of
immature teratoma. Germ cell–derived neoplastic cells
are seen in malignant teratomas. Intralesional calcifications, bony destruction of the coccyx and sacrum, and
hypoattenuating fat within the lesions are features of
teratomas and were not seen in our case.22,24,27,28,31-33
Developmental cysts are generally benign but do
have the potential to undergo premalignant and malignant transformations. The presence of adenocarcinoma within a completely isolated duplication cyst
Cancer Control 173
has been reported.34,35 These cases may manifest as
bowel-like cysts with no anatomical and luminal connection with the adjacent GI tract and with a separate
vascular pedicle. Development of adenocarcinomas
and neuroendocrine tumors has also been reported
in rectal tailgut cysts. One-half of all reported cases of
presacral carcinoids are thought to have developed in a
preexisting tailgut cyst or teratoma.29,32,34,36,37
Conclusions
Enteric duplication cysts are rare congenital anomalies
that should be considered in the differential diagnosis
of a retrorectal mass. Careful review of the clinical presentation, radiological features, and gross examination
of specimens and its surrounding tissue will help to
narrow the diagnostic possibilities, while histopathology and immunohistochemistry are critical to establish
the final diagnosis of enteric duplication cyst.
Acknowledgment: We thank Jackie Hattle for
technical assistance during the preparation and submission of the manuscript and the Pathology Histology
Laboratory for the preparation of the stains.
References
1.Mandhan P, Ehsan TM, Al-Sibai S, et al. Noncommunicating
multiple intra-abdominal enteric duplication cysts. Afr J Paediatr Surg.
2014;11(3):276-278.
2.Gümüş M, Kapan M, Gümüş H, et al. Unusual noncommunicating isolated enteric duplication cyst in adults. Gastroenterol Res Pract.
2011;2011:323919.
3. Atalar MH, Cankorkmaz L, Ozer H, et al. A huge duplication cyst of
the ileum. Pol J Radiol. 2013;78(3):70-73.
4. Sheikh MA, Latif T, Shah MA, et al. Ileal duplication cyst causing
recurrent abdominal pain and melena. APSP J Case Rep. 2010;1(1):4.
5. Stern LE, Warner BW. Gastrointestinal duplications. Semin Pediatr
Surg. 2000;9(3):135-140.
6. Lopez-Fernandez S, Hernandez-Martin S, Ramírez M, et al. Pyloroduodenal duplication cysts: Treatment of 11 cases. Eur J Pediatr Surg.
2013;23(4):312-316.
7. Gebesce A, Korkmaz M, Keles E, et al. Importance of the ultrasonography in diagnosis of ileal duplication cyst. Gastroenterol Res Pract.
2013;2013:248625.
8. Blank G, Königsrainer A, Sipos B, et al. Adenocarcinoma arising in
a cystic duplication of the small bowel: case report and review of literature.
World J Surg Oncol. 2012;10:55.
9. Rasool N, Safdar CA, Ahmad A, et al. Enteric duplication in children:
Clinical presentation and outcome. Singapore Med J. 2013;54(6):343-346.
10. Bal HS, Kisku S, Sen S, et al. A retroperitoneal enteric duplication
cyst communicating with the right upper ureter in an infant. BMJ Case Rep.
2014;9:2014.
11. Choi SO, Park WH, Kim SP. Enteric duplications in children--an analysis
of 6 cases. J Korean Med Sci. 1993;8(6):482-487.
12.Ho YC. Total colorectal and terminal ileal duplication presenting as intussusception and intestinal obstruction. World J Gastroenterol.
2012;18(43):6338-6340.
13. Patel MP, Meisheri IV, Waingankar VS, et al. Duplication cyst of the
pylorus--a rare cause of gastric outlet obstruction in the newborn. J Postgrad
Med. 1997;43(2):43-45.
14. Deesomsak M, Aswakul P, Junyangdikul P, et al. Rare adult gastric duplication cyst mimicking a gastrointestinal stromal tumor. World J
Gastroenterol. 2013;19(45):8445-8448.
15. Liaqat N, Latif T, Khan FA, et al. Enteric duplication in children: a
case series. Afr J Paediatr Surg. 2014;11(3):211-214.
16. Joyce EA, Kavanagh DO, Winter DC. A rare cause of low back pain:
Report of a tailgut cyst. Case Rep Med. 2012;2012:623142.
17. Raisolsadat SM, Zabolinejad N, Tabrizian-Namini F, et al. Tailgut cyst in an infant with imperforate anus: a case report. Iran J Pediatr.
2013;23(5):597-600.
18.Glasgow SC, Dietz DW. Retrorectal tumors. Clin Colon Rectal
Surg. 2006;19(2):61-68.
19. Baek SW, Kang HJ, Yoon JY, et al. Clinical study and review of
174 Cancer Control
articles (Korean) about retrorectal developmental cysts in adults. J Korean
Soc Coloproctol. 2011;27(6):303-314.
20. Kim H, Kim JH, Lim JS, et al. MRI findings of rectal submucosal
tumors. Korean J Radiol. 2011;12(4):487-498.
21. Kang JW, Kim SH, Kim KW, et al. Unusual perirenal location of a
tailgut cyst. Korean J Radiol. 2002;3(4):267-270.
22. Jang SH, Jang KS, Song YS, et al. Unusual prerectal location of a
tailgut cyst: a case report. World J Gastroenterol. 2006;12(31):5081-5083
23. Gunkova P, Martinek L, Dostalik J, et al. Laparoscopic approach to
retrorectal cyst. World J Gastroenterol. 2008;14(42):6581-6583.
24.Bathla L, Singh L, Agarwal PN. Retrorectal cystic hamartoma
(tailgut cyst): report of a case and review of literature. Indian J Surg.
2013;75(suppl 1):204-207.
25. Graadt van Roggen JF, Welvaart K, de Roos A, et al. Adenocarcinoma arising within a tailgut cyst: Clinicopathological description and follow up of an unusual case. J Clin Pathol. 1999;52(4):310-312.
26. Cho BC, Kim NK, Lim BJ, et al. A carcinoembryonic antigen-secreting adenocarcinoma arising in tailgut cyst: clinical implications of carcinoembryonic antigen. Yonsei Med J. 2005;46(4):555-561.
27. Alzaraa A, Mousa H, Dickens P, et al. Idiopathic benign retroperitoneal
cyst: a case report. J Med Case Rep. 2008;2:43.
28. Chhabra S, Wise S, Maloney-Patel N, et al. Adenocarcinoma associated with tail gut cyst. J Gastrointest Oncol. 2013;4(1):97-100.
29. Govaerts K, Van Eyken P, Verswijvel G, et al. A bronchogenic cyst,
presenting as a retroperitoneal cystic mass. Rare Tumors. 2012;4(1):e13.
30. Ramakrishna HK. Intestinal duplication. Indian J Surg. 2008;70(6):270-273.
31. Ekbote G, Pokharkar AB, Moon P. A rare case of perforated tubular
ileal duplication in 72-year-old male. Indian J Surg. 2013;75(suppl 1):418-420.
32. De Roeck A, Vervloessem D, Mattelaer C, et al. Isolated enteric
duplication cyst with respiratory epithelium: case report and review of the
literature. Eur J Pediatr Surg. 2008;18(5):337-339.
33. Pins MR, Compton CC, Southern JF, et al. Ciliated enteric duplication cyst presenting as a pancreatic cystic neoplasm: report of a case with
cyst fluid analysis. Clin Chem. 1992;38(8 pt 1):1501-1503.
34. Shin SY, Cho MY, Ryu H, et al. Adenocarcinoma originating from
a completely isolated duplication cyst of the mesentery in an adult. Intest
Res. 2014;12(4):328-332.
35. Okamoto T, Takamizawa S, Yokoi A, et al. Completely isolated alimentary tract duplication in a neonate. Pediatr Surg Int. 2008;24(10):1145-1147.
36. Spada F, Pelosi G, Squadroni M, et al. Neuroendocrine tumour arising inside a retro-rectal tailgut cyst: Report of two cases and a review of the
literature. Ecancermedicalscience. 2011;5:201
37. Liang JJ, Alrawi S, Fuller GN, et al. Carcinoid tumors arising in tailgut cysts may be associated with estrogen receptor status: case report and
review of the literature. Int J Clin Exp Pathol. 2008;1(6):539-543.
April 2016, Vol. 23, No. 2
Symposium Report
Second Annual Moffitt Anatomic Pathology Symposium
Dr Magliocco, chair of the Department of Anatomic Pathology, welcomed a near-capacity audience
made up of pathologists, histotechnologists, researchers, and pathology residents and medical students
at the 2nd Annual Moffitt Anatomic Pathology Symposium: Practicing Pathology in a Changing World
that took place in Clearwater Beach, Florida, on September 26, 2015. Th focus of the symposium was on
the updates of contemporary and emerging information and technology that impact practicing pathologists in an ever-changing world.
Invited speakers included Drs Odze and Schnitt, both from Harvard Medical School in Boston, who
shared their expert knowledge in gastrointestinal (GI) and breast pathology, as well as Theresa B. Burchette,
Lawrence Patton Jr, John J. Shelley, and Steven Westra. Experts from the H. Lee Moffitt Cancer Center &
Research Institute included Drs Magliocco, Bui, Centeno, Coppola, Hussaini, Khalil, Khazia, and Rosa,
as well as Mr Hicks and Ms Balasi, Molina, and Snyder. Drs Minton and Malafa also presented topics in
molecular pathology, digital pathology, and GI and breast cancers. The Ask the Expert Digital Pathology
session was an interactive discussion of real breast and GI pathology cases among the experts and the
audience.
A total of 22 scientific abstracts were proffered by students, residents, fellows, and staff. Dr Altiok,
chair of the research section of the Department of Anatomic Pathology, presented awards to the 3 best
submissions.
Dr Ryzhova and colleagues earned the Best Poster award for their work titled “Mutation Profiling of
High-Grade Serous Ovarian Adenocarcinomas Suggests STK11 and NOTCH1 as Candidate Biomarkers.” This paper reported identification of STK11 mutations as possible biomarkers for platinum-therapy
response and detailed actionable mutations in several patients with ovarian cancer.
The Best Research Project prize was awarded to Dr Dettloff and colleagues for their work “Utility of
a microRNA-Based Assay for Diagnosing Tumor Origin in Soft Tissue Sarcomas.” The investigators
examined the recent experience at Moffitt Cancer Center with noncoding microRNA tumor profiling of
tumors of uncertain origin. They listed several situations in which the technique can be particularly useful, such as the identification of germ-cell tumors. They also recognized pitfalls of the technique, such as a
bias toward sarcoma, possibly representing a biological shift in recurrent or previously treated tumors.
The Best Clinicopathologic Correlation award was won by Dr Liu and colleagues for their poster titled
“Spindle Cell/Sclerosing Rhabdomyosarcoma Is a New and Underdiagnosed Entity: A Clinical and Tissue
Microarray Immunohistochemical Study With Literature Review.” This work emphasized the utility
of using tissue microarray to screen previously encountered cases with antibodies directed against
emerging biomarkers.
Opening Remarks
Anthony M. Magliocco, MD
The esoteric and molecular laboratories in the Department of Pathology have implemented many new
equipment platforms and assays to support the personalized medicine programs at Moffitt Cancer Center.
These include the introduction of new next-generation
sequencing assays for solid tumors and hematological malignancies, the arrival of the NanoString platform (NanoString Technologies, Seattle, Washington)
to enable multiplex gene-expression profiling, and the
performance of the Prosigna assay (NanoString Technologies) for the analysis of early breast cancer on site.
Molecular experts continue to closely work with the
personalized medicine services at Moffitt Cancer Center;
a training fellowship program is also offered in moApril 2016, Vol. 23, No. 2
lecular pathology. Future directions will focus on the
development of new, broader panels and technologies
to monitor circulating tumor cells and free nucleic acid
in patients with cancer (liquid biopsy). The laboratory
also acknowledges the support from the Moffitt Foundation and the Morsani Family.
Summaries of Select Platform Presentations
Progression of Neoplasia in Barrett Esophagus
Robert D. Odze, MD
Barrett esophagus is a result of columnar metaplasia of the esophagus induced by gastroesophageal
reflux disease that, through a progressive sequence of
molecular changes, renders the mucosa at increased
risk for cancer via a metaplasia–dysplasia–carcinoma
sequence. The lecture focused on the diagnostic criteCancer Control 175
ria, controversies, and difficulties with regard to diagnosing and treating dysplasia in patients with Barrett
esophagus. The lecture highlighted areas of new developments and future trends. The risk of malignancy
and the treatment of patients with different types and
grades of dysplasia were discussed along with new
methods of endoscopic therapy and ablation. The roles
of pathologists in evaluating biopsies and endoscopic
mucosal resection specimens were also addressed.
Progression of Neoplasia in Irritable Bowel Disease
Robert D. Odze, MD
Similar to Barrett esophagus, cancer in patients
with irritable bowel disease (IBD) develops via an inflammation–dysplasia–carcinoma sequence. New
developments have been made in regard to the gross
(endoscopic) and microscopic classification of neoplasia in IBD. These new developments were discussed
at length as well as various areas of controversy with
regard to the diagnosis and management of dysplasia in patients with IBD. The criteria and limitations
of dysplasia grading were discussed in detail, as were
areas in need of future research and newly developed
technologies for the treatment of patients with IBD and
dysplastic lesions.
The Continuing Dilemma of Ductal Carcinoma
in Situ
Stuart J. Schnitt, MD
The term ductal carcinoma in situ (DCIS) encompasses a heterogeneous group of lesions that vary in
their clinical presentation, distribution in the breast,
pathological features, genetic and molecular alterations, and their potential to progress to invasive breast
cancer. During the past 30 years, widespread use of
screening mammography has resulted in a 9-fold increase in the incidence of DCIS. As a result, approximately 50,000 new cases of DCIS are diagnosed each
year in the United States, and this lesion accounts for
nearly 20% of breast “cancers” detected by mammography. Treatment of patients with DCIS is aimed at preventing the development of invasive breast cancer, and
this is best thought of as prophylactic management.
Current treatment options include mastectomy, excision, radiotherapy, and excision, any of which may be
accompanied by endocrine therapy. Clinical follow-up
studies have demonstrated that not all patients with
DCIS will progress to invasive breast cancer. However,
after decades of research, it is still not possible based
on clinical and pathological features alone to reliably
identify which patients with DCIS are likely to progress to invasive breast cancer and require aggressive
treatment and, conversely, which patients are unlikely
to progress and whose cancer may be adequately managed by excision alone or perhaps even no treatment
beyond diagnostic biopsy. Although a variety of bio176 Cancer Control
markers, as well as a newer molecular assay, is available to help stratify risk among patients with DCIS,
the clinical value of biomarkers remains to be determined. Newer treatment strategies are assessing the
role of neoadjuvant endocrine therapy and trastuzumab for estrogen receptor–positive and ERBB2 (formerlly HER2 or HER2/neu)–positive DCIS, respectively.
In addition, clinical trials evaluating the role of “active
surveillance” in patients with low-risk DCIS are underway. Furthermore, results from studies of the tumor
microenvironment are providing new insights into the
factors related to the progression of DCIS to invasive
breast cancer. A better understanding of the molecular alterations associated with the progression of DCIS
to invasive breast cancer may lead to new methods to
distinguish those patients with DCIS likely to recur or
progress from those who are not and to the identification of new targets for treatment and prevention.
Clinical Decision-Making Based on Molecular
Pathology
Susan Minton, MD
Genomic testing is now available and used in both
the early- and late-stage settings of breast cancer to
aid in the discussion of therapeutic options for select
patients. With the development and approval of several targeted therapies for the treatment of metastatic
breast cancer, the need is increasing to expand genomic testing to determine the most optimal therapy for patients. The availability of next-generation sequencing
provides more data for the physician to utilize when
making new treatment decisions. Several factors, such
as the particular line of therapy, patient comorbidities,
and previous biological behavior of the cancer, can
help differentiate which test may be more beneficial
in a particular circumstance. Prognostic genomic tests
can enhance treatment decisions for early stage breast
cancer. Several are now available in the early treatment
setting and provide information about possible early
and late risk of recurrence. Some of the advantages and
limitations of the most commonly used tests were reviewed through case discussions.
Evaluation of Postchemotherapy Breast Specimens
Marilin Rosa, MD
Neoadjuvant chemotherapy has become an established treatment modality for locally advanced and
large breast carcinomas. In addition, its use provides
an opportunity to evaluate patient response to chemotherapeutic drugs as well as to potentially change
a patient’s treatment course if satisfactory results are
not being achieved. The pathological evaluation of
postchemotherapy breast specimens is challenging
and requires careful correlation with previous biopsy
results, radiographic imaging (especially pre- and posttreatment tumor sizes), and clinical information. PathApril 2016, Vol. 23, No. 2
ological complete response (pCR) is strongly associated with long-term outcome (disease-free and overall
survival rates); therefore, histological evaluation is the
gold standard for the evaluation of tumor response to
therapy. The definition of pCR in the setting of breast
carcinoma varies among institutions, but it is generally
considered as the absence of residual invasive carcinoma within the breast and the lymph nodes. When
examining a postchemotherapy specimen, the residual
tumor, fibrotic tumor bed, or both should be grossly
identified and mapped in its larger dimensions. The
residual tumor should be generously sampled because
estimations of residual tumor size and cellularity are
necessary. To determine the rate of pCR is necessary in
order to identify chemotherapy changes in the fibrotic
tumor bed, at the site of biopsy, or both. Histological
chemotherapy changes include hyalinized vascular
stroma, stromal fibrosis, edema and elastosis, presence
of macrophages, inflammation, and hemosiderin pigmentation, among others. In the absence of these, additional sampling is required and correlation with clinical and radiological findings is necessary.
Multidisciplinary Gastrointestinal Case Presentation
Kevin Neill, MD, et al
We presented the case of a 33-year-old woman
with a locally advanced, solid pseudopapillary neoplasm (SPN) of the pancreas that later metastasized
to the liver. The presentation had a multidisciplinary
format. Dr Neill began with a brief summary of the
patient’s complete clinical course and the histopathology of solid pseudopapillary neoplasms. Dr Khazanchi
presented a video on the endoscopic ultrasonographical imaging characteristics of solid pseudopapillary
neoplasms. Dr Coppola then presented a comprehensive review of the current scientific literature on the
pathology, molecular alterations, miRNA, and gene
expression profiles of solid pseudopapillary neoplasms. Dr Malafa presented an overview on pancreaticoduodenectomy procedures, surgical management
of solid pseudopapillary neoplasms, and expected surgical outcomes in relation to the patient case featured
in this presentation. Dr Kis closed with a summary of
the chosen interventional radiology treatment course,
which included multiple chemoembolization procedures, chemoradiotherapy, and microwave ablation
surgery.
Incorporating Digital Pathology Into Pathology
Practice
Marilyn M. Bui, MD, PhD
Digital pathology, including telepathology,
whole-slide imaging, and image analysis is gaining
more adoption in pathology practice. This talk discussed the current state of using digital pathology in
surgical pathology practice in the United States based
April 2016, Vol. 23, No. 2
on a literature review, communication with early
adopters, and institutional and personal perspectives.
Helpful resources, updates in technology, best-practice
examples, practical issues using whole-slide imaging,
such as validation, and the guidelines of telepathology
were discussed. This talk also shed light on the bright
future of digital pathology in surgical pathology and its
opportunities in education and research.
Leadership Challenges in Histology
Malissa Snyder
Histotechnologists in leadership roles face many
challenges on a continual basis, and yet they must maintain a strong emphasis on patient care. The histology
laboratory produces high volumes of work and must
also keep quality control while maintaining appropriate turnaround times, which can be achieved by workload measurements and maintaining adequate staffing
levels. This workshop discussed strategies for prioritizing workflow and managing fast turnaround times in
a high-pressure environment. Tips for troubleshooting
common problems, tools for effective communication
between all team members in the laboratory, and tips
for managing difficult personalities were discussed.
Challenges in Immunohistochemistry
Jodi Balasi
With new antibodies emerging and the demand
for these antibodies to be integrated into the pathologist’s diagnosis, histologists are challenged to test and
validate new antibodies into their current immunohistochemistry menu. Whether a histotechnologist is new
to the process or is a seasoned professional, the process always starts with the optimization and validation
of the antibody. This workshop highlighted the immunohistochemistry protocol set up, testing, documentation, and validation of in vitro diagnostic, analyte specific reagent, and research-use only antibodies.
Selected Abstracts Accepted for Poster
Presentations
1. Further Characterization of Circulating Tumor
Cells With User-Defined Antibodies
John Puskas, Gisela Cáceres, Carolyn Loret de Mola,
Mike Gruidl, and Anthony Magliocco
H. Lee Moffitt Cancer Center & Research Institute,
Tampa, Florida
Evaluation of circulating tumor cells (CTCs) is
emerging as an important component of the liquid biopsy and provides important information for oncologists about their patients. CTCs provide a real time
evaluation of current disease state in patients and
provide information of tumor evolution in metastatic
sites that cannot be biopsied. Also, monitoring CTCs
may provide pharmacodynamic information regarding the effect of treatment to cause cell cycle arrest or
Cancer Control 177
dampen signaling pathways. This could be used to
more quickly discontinue futile treatments, enabling a
switch to alternate options or maintain treatments that
appear effective. The most recognizable CTC phenotype is EpCAM+, cytokeratin 8, 18 or 19+, DAPI+, and
CD45- cells and CTCs are measured in the peripheral
blood (PB) using the FDA-approved CellTracks System
for metastatic breast, colorectal and prostate cancers.
To use this technology for other applications the assay requires modification that includes capture antibody selection (CD146, N-cadherin and O-cadherin),
unique cell surface markers (Her2/neu and AR) and
nuclear markers (Ki67) and validation using cell lines,
creating a LDT. The technology is limited by the number of available markers and the ability to measure the
expression level of the protein in the captured cells as
well as the viability which is 24–96 hours. Recently we
have verified the use of HER2/neu, Ki67 and androgen
receptor (AR) user-defined antibodies for further characterization of CTCs and are applying these new assays
for use in clinical trials.
2. Comparing and Contrasting Decalcification
Solutions: Cellular and Nuclear Preservation
Malissa Snyder and Lindsey Martinez
H. Lee Moffitt Cancer Center & Research Institute,
Tampa, Florida
The goal of this project is to preserve nuclear and
cellular detail by finding the best decalcification solution. Preserving antigenicity is also a main goal in
order to achieve the highest quality of immunohistochemical staining. Three decalcification solutions were
used by testing surgical and bone marrow solutions
and comparing the H&E stains and pertinent Immunohistochemistry stains. Three decalcification solutions
were used: nitric acid (Nitrical), formic acid (Formical),
and formic acid with formalin (Formical-4). Six surgical
specimens were tested, and of the six, the Formical-4
yielded the best results. Four bone marrow specimens
were also tested with the three different decalcification solutions, and again, the Formical-4 gave the best
results. The Formical-4 preserved the nuclei and cytoplasmic components of the cells of both specimen
types the best. All of the immunohistochemistry stains
that were tested also yielded the best staining results.
3. Cancer Cell Line Doubling Time Describes
Epithelial-Mesenchymal Transition
Bernadette M. Boac,1 Yin Xiong,1,2 Douglas
C. Marchion,1,2 Forough Abbasi,1,2 Beman R.
Khulpateea,1 E. Clair McClung,1 Stephen H. Bush,1
Sharon E. Robertson,1 Johnathan M. Lancaster,1–3 and
Anthony M. Magliocco1,3,4
1 Department of Women’s Oncology, 2 Chemical Biology and Molecular Medicine Program,
3 Department of Oncologic Sciences, 4 Department
178 Cancer Control
of Anatomic Pathology, H. Lee Moffitt Cancer
Center & Research Institute, Tampa, Florida
Background: Patients with aggressively dividing cancers are generally diagnosed at an advanced
stage of disease, have higher tumor burden, and carry
a poorer prognosis. We sought to identify molecular
signaling pathways associated with fast dividing (high
doubling rate) cancer cells.
Methods: Pathways were identified using SAM
(significance analysis of microarrays) correlation
of Affymetrix HG-U133_Plus_2 expression data and
doubling time in 59 human cancer cell lines encompassing 9 cancer cell types. PCA (principal component analysis) was used to analyze the pathways association with
overall survival using institutional as well as publically
available clinicogenomic datasets containing a total of
2,205 patients with a broad range of human cancers,
including ovarian, breast, lung, leukemia, brain, and colon.
Results: Pearson’s correlation of doubling times
and genomic expression data identified the differential
expression of 3,811 probesets (FDR < 0.05: 1,110 upregulated; 2,701 downregulated) representing 47 molecular signaling pathways (FDR<0.05, fold change > 2)
to be associated with fast dividing cancer cells. Twentytwo pathways (P < 0.05) were found to be associated
with overall survival in 2 or more datasets. The integrins pathway was associated with survival in 5 datasets
comprising of breast, colon, and ovarian cancers.
Conclusion: Several pathways associated with
cancer cell doubling time may influence overall survival and thus be good therapeutic targets for drug
development. Surprisingly, the majority of these pathways are descriptive of epithelial-mesenchymal transition biology.
4. Validation of a Rapid, Sensitive, and Inexpensive
QClamp BRAF Assay for the Detection of Codon
600 Specific Mutations
Ravi Kothapalli,1 James Saller,1 Carolyn Loret de Mola,1
and Anthony Magliocco1,2
1Clinical Testing Development, 2 Department of
Anatomic Pathology, H. Lee Moffitt Cancer Center &
Research Institute, Tampa, Florida
BRAF mutations occur in approximately 8% of all
human cancers and around 50% in melanoma and papillary thyroid carcinoma. Identification of these mutations provides potentially valuable theranostic and
prognostic information in cancer patients. DiaCarta
(Hayward, CA) has developed a QClamp BRAF Codon
Specific Mutation Test in Codon 600 based on xenonucleic acid mediated PCR clamping technology. Validation of this assay is performed prior to implementing it for routine use in clinical laboratory. DNA patient
samples that were selected have a BRAF mutation at
the 600 codon whereas others are negatives. Allelic frequencies of between 50% to 0.156% BRAF, V600E were
April 2016, Vol. 23, No. 2
made by mixing appropriate portions of BRAF mutant
DNA and wild type DNA obtained from Horizon Diagnostic (Cambridge, UK) and used in the assay. DiaCarta
BRAF assay can reliably detect up to 0.156 % allelic frequency. The results of the assay are 100% in agreement
with Nextgen sequencing or pyro sequencing. The
main advantage of this assay is to detect the BRAF
mutation in ultra-low concentrations of DNA (10 ng
of total DNA) as obtained from specimens including
FFPE, liquid biopsies, cytology and tissues samples.
The BRAF V600E mutation may serve as a very sensitive biomarker when the DiaCarta assay is used. Assays
with high analytical sensitivity will likely be required
for effective screening and minimal residual disease
monitoring in liquid biopsy samples, as they can detect mutations in samples of DNA with high contaminating normal DNA background and identification of
rare subclones within heterogeneous tumor samples. 5. Mutation Profiling of High-Grade Serous Ovarian
Adenocarcinomas Suggests STK11 and NOTCH1 as
Candidate Biomarkers
Elena Ryzhova,1 Douglas C. Marchion,2,3 Forough
Abbasi,2,3 Liang Nong,1 Alexandra Gartel, 4 Yin Xiong,2,3
and Anthony Magliocco1,5
1Clinical Testing Development Laboratory,
2Department of Women’s Oncology, 3Chemical Biology and Molecular Medicine, 4Molecular Laboratory,
5Department of Anatomic Pathology, H. Lee Moffitt
Cancer Center & Research Institute, Tampa, Florida
Introduction: The standard treatment of high
grade serous ovarian cancer (HGS-OvCa) with platinum-taxane chemotherapy is followed by the development of chemoresistant disease in a significant proportion of patients. Molecular mechanisms underpinning
chemoresistance remain obscure. In this study we performed mutation profiling of patients with varying responses to chemotherapy to investigate whether platinum-resistant phenotype can be linked to a particular
molecular signature.
Methods: DNA samples from 37 HGS-OvCa patients classified as either complete responders (CR,
n = 18) or incomplete responders (IR, n = 19) were tested in Agena BioScience OncoPanel. The OncoPanel relies on the iPlex-MassArray methodology and permits
parallel screening of 214 mutations in 26 oncogenes
and tumor suppressor.
Results: Frameshift or nonsense mutations in
STK11 were found in 22% of the CR group suggesting a significant association with the disease phenotype (Fisher’s exact test, p=0.046). Further, mutations
in NOTCH1 were identified in 16% samples regardless
of platinum-taxane response status. NOTCH has been
previously associated with certain hematologic and
solid tumors; however it was not described in the context of HGS-OvCa. Lastly, well-established actionable
April 2016, Vol. 23, No. 2
mutations in KRAS, EGFR, and BRAF were detected in
patients demonstrating CR and IR.
Conclusions: We have identified STK11 mutations as possible biomarkers for platinum-therapy
response. Additionally, we have found that NOTCH1
could be a candidate for therapeutic targeting in HGSOvCa. Furthermore, we have identified several HGSOvCa specimens that harbor known actionable mutations. These data suggest that routine multiplexed
mutation profiling of HGS-OvCa patients may facilitate the proper selection of available therapeutic options and/or clinical trials.
6. Development and Validation of a MassArray
Custom GliomaPanel for Testing IDH1 and IDH2
Mutations in a Clinical Setting
Elena Ryzhova,1 Rob Macaulay,2 Liang Nong,1 Ravi
Kothapalli,1 Linda McIntosh,1 Gisela Caceres,1
Alexandra Gartel,3 Janella Lal,3 Shital Gandhi,3 Hema
Liyanage,5 Robin Everts,5 Carolyn Loret de Mola,1
Dahui Qin,3 Peter Forsyth,4 and Anthony Magliocco1,2
1Clinical Testing Development, 2 Department of
Anatomic Pathology, 3Laboratory of Molecular Pathology, 4Department of Neuro Oncology, H. Lee Moffitt
Cancer Center & Research Institute, Tampa, Florida,
5 Applications and Technology, Agena Bioscience,
San Diego, California
Introduction: IDH1 and IDH2 mutations are associated with favorable outcome in low-grade gliomas.
The common methods of IDH1, 2 testing are characterized by either limited spectrum of detected mutations
(IHC) or low sensitivity (Sanger sequencing). To overcome these limitations we developed high throughput,
sensitive and multiplexed GliomaPanel test using Agena Biosciences’ MassARRAY technology.
Methods: GliomaPanel was designed to interrogate all the essential mutations in IDH1 and IDH2. Additionally, it includes 116 hot-spot targets in 17 genes
with potential clinical or research utility. The detection method relies on a single nucleotide extension followed by MALDI-TOF mass spectrometry.
Results: The panel was validated in accordance
with the CAP guidelines and included the total of 78
FFPE diagnostic samples. For the majority of samples,
the mutation status was pre-determined by IHC. The established GliomaPanel accuracy, specificity and sensitivity were 99%, 100%, and 97% respectively. Notably, the
rare IDH1 R132C mutation was detected in several patients that were IDH1 negative in IHC tests. Additionally,
R132C mutation at low frequency (20%) was identified
in one patient, whose IDH1 status was negative in both
IHC and Sanger sequencing. The discrepancies were resolved by Sanger sequencing confirming GliomaPanel
results. Finally, we demonstrated that 70% of DNA samples, which failed NGS testing acceptance criteria, were
accurately processed by GliomaPanel.
Cancer Control 179
Conclusions: GliomaPanel offers cost-effective
high-throughput solution for accurate of IDH1 and
IDH2 profiling. The method detects a broad range of
mutations, is highly sensitive, has relaxed DNA quality
requirements, and represents a valuable alternative to
conventional methods for targeted mutation profiling.
7. Higher Frequency of Isolated PMS2 Loss in
Colorectal Tumors From Colombian Population
Rania Shamekh,1 Mauro Cives,2 Jaime Mejia,3 and
Domenico Coppola4
1Department of Pathology, University of South
Florida, Tampa, Florida, 2Department of GastroIntestinal Oncology, H. Lee Moffitt Cancer Center &
Research Institute, Tampa, Florida, 3Department of
Pathology, Institutode Patologia Mejia Jimenez, Cali,
Colombia, 4Department of Anatomic Pathology, H. Lee
Moffitt Cancer Center & Research Institute, Tampa,
Florida
Colorectal cancer (CRC) is the third most common cancer and the fourth most common cause of
death worldwide. It accounts for over 9% of all cancer. One of the pathogenic factors of CRC is germ line
mutation, leading to alteration and inactivation of the
MMR genes. The aim of the study is to compare the
frequency of alterations in MMR protein expression in
CRC of Caucasian as compared to Colombian patients.
Forty-five Colombians and 48 Caucasians with CRC
were studied. The tumors’ microsatellite instability
(MSI) status was determined in primary CRC by immunohistochemistry using the automated Ventana Ultra.
The combined loss of MLH1 and PMS2 was the most
common alteration in both Colombian (11%; 5 out of
45) and Caucasian (12%; 6 out of 48) CRC patients. Interestingly, the loss of PMS2 expression in the presence
of intact MLH1 was the second most common alteration in Colombians (8%; 4 out of 45) which was never
seen in the Caucasians cohort (P = 0.05). The loss of
MLH1 alone and the combined loss of MSH6 and PMS2
expression was only observed in one out of 45 (2%)
in Colombians but not in Caucasians. The combined
loss of MSH2 and MSH6 was not observed in any of
the patients studied. The current findings highlight the
significant difference in alterations of MMR protein expression in CRC patients from Colombia compared to
Caucasians. These findings are novel and warrant further studies in larger cohorts.
8. Human Lung Cancer Derived Cancer Associated
Fibroblasts Express a Variety of Immunosuppressive Mechanisms
Arjun Rammohandas, David Noyes, Melanie MediavillaVarela, Liliana Lyviv, Dillan Villavisanis, and Scott
Antonia
H. Lee Moffitt Cancer Center & Research Institute,
Tampa, Florida
180 Cancer Control
Immunotherapy has emerged as clinical efficacious in a variety of different cancer types. In particular this modality has been revolutionary in the treatment of lung cancer with anti-PD1/PD-L1 producing
objective tumor responses in approximately 20% of
patients and a dramatic impact on overall survival. As
impactful as this has been, a large number of lung cancer patients do not benefit. This is due to the fact that
there are numerous ways that tumors evade rejection
by T cells, including a number of immunosuppressive
mechanisms that can be operational within the tumor
microenvironment including immune checkpoint ligands, immunosuppressive cytokines and enzymes,
and immunosuppressive cells. One of these is the
cancer associated fibroblast (CAF), though the precise
way that these cells inhibit T-cells has been unknown.
Freshly resected tumors not necessary for pathology
were harvested, mechanically and enzymatically disaggregated and digested cells were plated in complete
RPMI and allowed to adhere and proliferate CAFs. We
have successfully propagated dozens of CAF and tumor infiltrating lymphocyte (TIL) lines and characterized a portion of them so far. Characterization was accomplished with immune-phenotypic characterization
through flow cytometry. ELISA was used to determine
immunosuppression with CAFs and TILs. Cytokine
production was analyzed with multiple analyte profiling. Data were confirmed with qRT-PCR and Western
blot analysis. Here we show that several human lung
cancer derived CAFs constitutively express PD-L1 and
CD73, and can be induced to express MHC class II,
HVEM, IL-6, CXCL8, IDO, and NOS, all of which are
immunosuppressive. These data suggest that a single
manipulation targeting CAFs could influence CAF phenotype and cytokine secretion affecting the tumor microenvironment.
9. Decreased Stromal Expression of Caveolin-1
as a Potential Biomarker for Ovarian Serous
Carcinomas
Daryoush Saeed-Vafa,1 Douglas C. Marchion,2 Susan
M. McCarthy,1 Lu Chen,3 Ardeshir Hakam,1 Emily C.
McClung,2 Robert M. Wenham,2 Sachin M. Apte,2
Dung-Tsa Chen,3 Anthony M. Magliocco,1 and Jenny
Permuth-Wey4,5
Departments of 1Anatomic Pathology, 2Women’s
Oncology, 3Biostatistics and Bioinformatics, 4Cancer
Epidemiology, 5Gastrointestinal Oncology, H. Lee Moffitt
Cancer Center & Research Institute, Tampa, Florida
Background: Previous studies have demonstrated that Caveolin-1 (Cav-1) can act as a tumor suppressor in various malignant neoplasms. It has recently been
shown that there is reduced expression of Cav-1 in the
stroma of malignant tumors as compared to the stroma
of their benign counter parts and that this expression
may be correlated with histological grade, specifically
April 2016, Vol. 23, No. 2
in ovarian serous tumors. To our knowledge this difference has yet to be quantified. This study utilized the
Automated Quantitative Software Analysis (AQUA) system and attempted to quantify the difference in the level
of expression of Cav-1 in the reactive stroma of ovarian
serous carcinomas as compared to stroma from benign
ovaries.
Material/Methods: Cav-1 expression was studied via a multiplex immunofluorescence assay in five
tissue microarrays containing duplicate 1 mm cores
from various grades of ovarian serous carcinomas
and benign ovaries, and was quantified via an AQUA
algorithm specifically designed to target stromal Cav-1
expression.
Results: The study included cores representing
reactive stroma from 117 unique ovarian serous carcinoma cases and 30 cases demonstrating stroma from
benign ovaries. There was a statistically significant decrease in the expression of Cav-1 in the reactive stroma
of ovarian serous carcinomas as compared to the stroma of benign ovaries, with mean AQUA scores, quantifying the amount of Cav-1, of 161.1 and 324.9 respectively (P < .0001).
Conclusion: Our findings demonstrate that decreased stromal Cav-1 expression may be associated
with malignant ovarian serous carcinomas. Studies are
underway to evaluate stromal Cav-1 expression as a
novel prognostic marker and rational therapeutic target for ovarian cancer.
10. Utility of a microRNA-Based Assay for Diagnosing Tumor Origin in Soft Tissue Sarcomas
Jennifer Dettloff, Evita Henderson-Jackson, and
Marilyn M. Bui
H. Lee Moffitt Cancer Center & Research Institute,
Tampa, Florida
Background: An accurate and definitive diagnosis plays a critical role in precision medicine. However,
soft tissue sarcomas frequently present as diagnostic
challenges due to their rarity, diversity and overlapping
with other malignancies, both morphologically and
immunohistochemically. MicroRNA (miRNA) is a master control of post-transcriptional regulation of proteincoding genes, some of which influence oncogenesis.
MiRNA-based testing is an emerging tool in diagnosing
tumor origin. This study is intended to investigate its
utility in diagnosing soft tissue sarcomas.
Materials and Methods: The MCC archives contained 44 cases that had been sent for a commercially
available miRNA-based tumor-of-origin test. Of those 44
cases 33 had sarcoma in the differential diagnosis. The
data included pre-testing working diagnosis, miRNA test
result, and the post-testing multidisciplinary decision.
Results: Of the 33 cases we used clinical, histopathologic and immunohistochemical findings to determine the most likely diagnosis as the gold standard.
April 2016, Vol. 23, No. 2
There were 23/33 cases (69%) that yielded a result of
sarcoma. Of those only 8/23 cases (35%) appeared to
actually be sarcomas. The test appeared especially
weak at differentiating sarcoma from carcinoma/
melanoma, which was the differential in 17 cases. Of
those 17 cases the clinical and pathologic features favored: carcinoma/melanoma in 11/17, sarcoma in 3/17,
and 3/17 were equivocal. The majority of the probable
carcinoma/melanoma cases appeared clinically to be
metastatic. Cytokeratin or EMA expression was seen in
all probable carcinoma cases except one adrenal cortical carcinoma. Seven of 33 tested as mesothelioma,
none of which clinically or pathologically appeared to
be mesothelioma, including one suspected chordoma;
these results were therefore considered uninformative. Three of the 33 cases included a differential diagnosis specifically between a germ cell tumor (GCT)
and a sarcoma. One of these three cases, although the
clinical impression was lymphoma or sarcoma, tested
for seminomatous GCT, which eventually lead to the
correct diagnosis of a seminoma. In the second of
these three cases, a patient with a history of a testicular tumor presented with a new groin mass. The test
confirmed the correct diagnosis of a new primary sarcoma. The third of these cases also carried the differential of GCT due increased HCG clinically; the miRNA
test was helpful in helping rule out that differential.
Four of the 33 cases yielded a test result of lymphoma
(B- or T-cell origin). Two of these were of predicted
dendritic cell origin and the miRNA test was helpful;
one was a medullary carcinoma of the breast and one
was a sarcoma.
11. Incidental Metastatic Medullary Carcinoma of
the Colon Involving an Extraterritorial Adenoma:
Diagnostic Pitfall and Impact on Patient Care
Carolina Dominguez, Cory Porteus, Diana Braswell,
Sherma Zibadi, and Kun Jiang
H. Lee Moffitt Cancer Center & Research Institute,
Tampa, Florida
Context: Microsatellite instability associated
colorectal medullary carcinoma (MC) has been gaining
attention recently due to its impact on clinical management and prognostication. Usually, MC is composed of
sheets of tumor cells with pushing borders and lymphoplasmacytic banding. Rarely does MC present as
a mucinous adenocarcinoma or a pure signet ring cell
carcinoma. It is believed that MC possesses less aggressive properties compared with microsatellite-stable tumors; however, we recently encountered an opposite
example, with lymphovascular invasion and distant
metastasis in MC.
Design: We present a unique case of metastatic
signet ring cell MC, which was incidentally identified
involving a noncontiguous distant adenoma following screening colonoscopy, so as to emphasize these
Cancer Control 181
unique and essential histological features and to caution diagnostic pitfalls.
Results: A 58-year-old man presents for a routine
colonoscopy. A 2-cm adenoma was removed from hepatic flexure, which showed signet ring cell carcinoma,
with loss of MLH1/PMS2 and retained MSH2/MSH6
immunohistochemically. Next generation sequencing
showed no mutation in BRAF, EGFR, and Ras. The cauterized margin was benign. Significantly, biopsy of a
noncontiguous 0.5 cm adenoma (35 cm distal) showed
rare signet ring cells within lymphovascular spaces,
with identical morphology and immunophenotype as
seen in MC. Due to this unexpected finding, patient received chemotherapy, awaiting resection.
Conclusions: MC could present as signet ring call
carcinoma, with multifocal lymphovascular invasion
and aggressive clinical behavior; understanding the
molecular basis of MC and awareness of its histological features and clinical impact are pivotal for reaching the correct diagnosis and for improving patient
management.
12. Metastatic Basal Cell Carcinoma: Recent Moffitt
Cancer Center Experience
T. Mendoza, Farah Khalil, and J. Messina
H. Lee Moffitt Cancer Center & Research Institute,
Tampa, Florida
Introduction: Basal cell carcinoma (BCC) of the
skin has the lowest incidence of metastasis of any epithelial malignancy, even though it is the most common
primary carcinoma.
Methods: Cases of metastatic BCC encountered
in our practice were included. The subtype of primary
BCC was documented on review of the cases. The diagnosis of metastatic BCC was based on standard histologic criteria: histomorphologic confirmation with
primary cutaneous BCC and supportive immunohistochemical studies were performed.
Results: Six cases of metastatic basal cell carcinoma were included for study. The histologic subtypes included: four infiltrative BCCs, one infiltrative BCC with
basosquamous features, and one basosquamous type
BCC. The age of patients at diagnosis of the metastatic BCC ranged from 50 to 77. All were men. The sites
of metastasis included: lung (3 patients), lymph nodes
and lung (2 patients) and bone (1 patient). The time to
metastasis was available in five cases and ranged from
4 years to 20 years; the median time was 7 years. Of
the two patients with lymph node and lung metastases,
one patient had a lymph node metastasis 9 years after
diagnosis of the primary and a lung metastasis 16 years
after diagnosis. The lymph node and lung metastases
in the other patient were found 20 years after diagnosis. Significant therapy and survival information was
available for four patients not recently diagnosed: four
patients were treated in a hedgehog inhibitor trial
182 Cancer Control
(vismodegib). Three of those four also underwent radiation therapy. One patient died of disease progression
at one year, one patient had disease progression at two
years, one patient had stable disease at two years and
one patient was lost to follow-up shortly after therapy.
Parenthetically, one of the six cases was identified as
having a PTCH1 genetic alteration, which is a target of
hedgehog inhibitor therapy.
Conclusion: The metastatic BCC in our experience
preferentially spread to the lung, followed infiltrative
BCCs, and occurred a median of seven years after
diagnosis.
13. Neuroendocrine Tumors (NETs) of the Thymus:
Experience of a Single Cancer Center
Sameer Al Diffalha1 and Farah Khalil1,2
1Department of Pathology and Surgical Oncology
H. Lee Moffitt Cancer Center & Research Institute,
Tampa, Florida, 2University of South Florida Department of Pathology and Cell Biology, Tampa, Florida
Background: Neuroendocrine tumors (NETs)
of the mediastinum and thymus are rare. The term
carcinoid was first presented in 1907 by Oberndorfer.
Rosai and Higa were the first to acknowledge the existence of carcinoid tumors in the thymus.
The median age for thymic NETs is 59 years and
it is more common in males (M: F ratio 3:1). It has an
annual incidence of 0.01/100,000 in the United States
with ~ 400 cases reported in the literature to date. The
prevalence of thymic NET is ∼ 3% of the total number
of NETs at all sites and is estimated to account for approximately 2% to 4% of all anterior mediastinal neoplasms. It may be part of multiple endocrine neoplasia
type 1 syndrome (MEN-1, 5%–15%).
Material and Methods: We searched our database for all thymic neuroendocrine tumors that had
been diagnosed between 1987 and 2015 (August). During this interval, there were 27 patients who had been
diagnosed with thymic neuroendocrine tumor. Three
patients had typical carcinoid tumor and 23 patients
had atypical carcinoid tumor or high-grade neuroendocrine tumor.
Tumorigenesis: The first hypothesis proposes
that the NET tumor cells arise from ectopic tissues and
the other hypothesis suggests that they arise from teratomatous components.
Molecular Biology: NETs have a distinct gene expression from the normal thymus. The development of
NETs may involve different genes, each of which may
be associated with several different abnormalities that
include point mutations, gene deletions, DNA methylation, chromosomal losses and chromosomal gains.
The foregut, mid-gut and hindgut NETs have different
molecular pathways. Foregut NETs have frequent deletions and mutations of MEN1, mid-gut NETs have losses of chromosome 18, 11q and 16q and hindgut NETs
April 2016, Vol. 23, No. 2
express transforming growth factor alpha and the epidermal growth factor receptor.
There are chromosomal imbalances that have
been found by some investigators in thymic tumors,
which include gains of chromosome Xp, 7p, 7q, 11q,
12q and 20q, and losses at 6q, 6p, 4q, 3p, 10q, 11q and
13q. Loss of heterozygosity (LOH) at chromosome
1p has been reported in two thymic neuroendocrine
tumors. Loss of chromosomes 3, 9p21, Y and gain of
chromosome 19p were discovered in one case. Bi Y.F.
et al found evidence of Wnt pathway activation and
consistently elevated level of cyclin D1 in AC tumors
but, c-Myc expression changed only moderately.
Prognosis: Thymic NETs have more aggressive
behavior than their pulmonary equivalents. The most
important prognostic factors with validated significance are: Tumor size, histological grade, paraneoplastic symptoms, Thymic Masaoka staging and surgical
resection. Ki67 proliferation index is the key player in
the stratification of patients with good and bad prognosis in other neuroendocrine tumors. Regional lymph
nodes are often affected and distant metastases are
common.
Management: Surgical resection remains the
typical management for NETs. Surgery followed by
chemotherapy has become standard for metastatic
bronchial and thymic NETs. Tyrosine kinase inhibitors (eg, sunitinib) and TOR inhibitor everolimus have
been stated to show better prognosis in a small series.
Cisplatinum-based regimens have also been of value
in thymic NETs.
SCLC is sensitive to chemotherapy and since the biological characteristics of LCNEC are similar to those of
SCLC, both are treated with chemotherapy regimens.
14. Lymphoepithelioma-Like Hepatocellular Carcinoma Mimicking Hodgkin Lymphoma: Diagnostic
Pitfalls and Impact on Management
Ian Martin, Rania Shamekh, Julie Gibbs, Ardeshir
Hakam, and Kun Jiang
H. Lee Moffitt Cancer Center & Research Institute,
Tampa, Florida
Introduction: Hepatocellular lymphoepitheliomalike carcinoma (LELC) is a unique, poorly differentiated carcinoma featuring dense lymphoid infiltrate surrounding neoplastic hepatocytes. To date, only a few
cases have been reported. The etiological connection
with immunosuppression or Epstein–Barr virus has
not been determined. Morphologically, hepatocellular
LELC mimics certain types of lymphoma, especially
Hodgkin lymphoma, posing diagnostic challenges. As
LELC patients generally have a better prognosis than
those with conventional hepatocellular carcinoma, the
correct diagnosis is crucial for patient care.
Methods: We present a case of hepatocellular
LELC that was misdiagnosed and managed as Hodgkin
April 2016, Vol. 23, No. 2
lymphoma at an outside institution during a 6-month
period. The aim of the study is to emphasize the key
histological and immunophenotypical characteristics
of LELC in an attempt to reinforce its diagnostic features and impact on clinical course.
Results: A 71-year-old male was referred by his
oncologist for a second opinion regarding his liver
mass and hilar adenopathy, which was originally interpreted as Hodgkin’s lymphoma by an outside institution 6 months prior. The liver biopsy was reviewed at
our institution. It showed a poorly differentiated carcinoma with dense lymphocytic infiltrate admixed with
few large and prominent epithelial cells. Notably, no
eosinophils or neutrophils were identified within the
lymphoid infiltrate. The epithelial cells were immunoreactive for AE1/3, HepPar-1, and EMA, indicating
hepatocellular origin. They were negative for CD15,
CD30, and CD45. Importantly, the Ki-67 index was
80%, which is highly suspicious for malignancy. Furthermore, repeat bone marrow biopsy, flow cytometry,
and lymph node biopsies were unremarkable. The diagnosis of hepatocellular LELC was established. The
patient’s clinical management was subsequently adjusted following this diagnosis.
Conclusions: This case demonstrates that hepatocellular LELC could be misinterpreted as a lymphoproliferative disease, potentially derailing clinical management. An astute histological and immunophenotypical
examination plus awareness of this entity’s clinical impact are pivotal for appropriate patient management.
15. A Rare Case of Combined Small Cell Lung
Cancer With Adenocarcinoma and Squamous Cell
Carcinoma
Sameer Al Diffalha1 and Farah Khalil1,2
1Department of Pathology and Surgical Oncology, H. Lee Moffitt Cancer Center & Research Institute,
Tampa, Florida, 2University of South Florida College of
Medicine, Department of Pathology and Cell Biology,
Tampa, Florida
Introduction: Combined small cell lung cancer
(c-SCLC) is relatively rare and occurs in approximately
2% to 14% of all cases of small cell lung cancer (SCLC).
c-SCLC is defined by the World Health Organization
(WHO) as small cell carcinoma combined with an additional component consisting of any non–small-cell
histologic type, including adenocarcinoma, squamous cell carcinoma (SCC), and large cell neuroendocrine carcinoma (LCNEC). Most of the combined
cases are SCLC with either adenocarcinoma or squamous component. Rare cases with up to 3 different
morphologic tumors have been reported, and individual tumors containing up to 4 different morphologic
constituents have been described.
Case Presentation: We present the case of a
78-year-old male with a smoking history of 2 to 3
Cancer Control 183
packs of cigarettes per day for 35 years. The patient
was referred for evaluation of right upper lobe (RUL)
lung mass to Moffitt Cancer Center. The CT scan
showed a 3.0 × 2.3 × 2.5 × cm RUL lung mass with
mildly prominent mediastinal and hilar lymphadenopathy. The patient elected to proceed with right thoracotomy with right upper lobectomy, intercostal muscle
flap, and lymphadenectomy. Microscopic examination
showed combined well-differentiated adenosquamous
carcinoma and small cell carcinoma. Next generation
sequencing did not identify clinically relevant results.
Tumor Genesis, Molecular: The mechanism of
c-SCLC is believed to be the simultaneous appearance
of both SCLC and other histological types, or the partial malignant transformation from non-SCLC. Tyrosine kinase inhibitors (TKIs) are active against different mutations in EGFR. While EGFR mutations are very
rare in SCLC, they are quiet common (about 15%–20%)
in c-SCLC, usually in nonsmoking females with adenocarcinoma component.
Prognosis: In general, patients with c-SCLC have
poor prognosis due to the small cell component. However, c-SCLC has worse outcome due to increased resistance to chemotherapy (CT) and/or radiation therapy
(RT) than “pure” SCLC.
Management: While there are available guidelines for treating SCLC, the optimal treatment for cSCLC, which will improve prognosis has not been adequately determined. Few studies suggest surgery for
very early stage c-SCLC, which might improve the patient’s outcome. However others recommend starting
CT and/or RT and then following with surgical intervention for the residual NSCLC components.
Conclusion: Combined tumors in any organ site
raise interesting biologic questions about the pathogenesis and relationship of the individual components.
Moreover, an accurate understanding of c-SCLCs is of
great practical importance because treatment strategies are significantly different for NSCLC and SCLC.
16. Diagnostic Dilemma: Incidentally Identified
Disseminated Peritoneal Mantle Cell Lymphoma
Mimicking Peritonitis Following Esophagogastrectomy and Splenectomy for an Invasive Esophageal
Adenocarcinoma
Cory Porteus, Carolina Dominguez, Diana Braswell,
Elizabeth M. Sagatys, and Kun Jiang
H. Lee Moffitt Cancer Center & Research Institute,
Tampa, Florida
Introduction: Mantle cell lymphoma (MCL) is an
aggressive subtype of B-cell lymphoma that represents
7% of non-Hodgkin cases, composed of CD5-positive
naive B-cell of mantle zone surrounding germinal
center follicles. MCL is clinically challenging to manage, with an ominous prognosis. MCL involves lymph
nodes, spleen, bone marrow, and gastrointestinal tract
184 Cancer Control
organs. MCL involvement of peritoneal cavity and
omentum is extremely rare, if not unheard of.
Methods: Here we present a novel case of disseminated peritoneal and omental MCL mimicking postsurgery peritonitis after surgical resection of an esophageal tumor.
Results: A 67-year-old gentleman was referred to
our cancer center recently for an esophagogastrectomy
and splenectomy due to esophageal adenocarcinoma
and splenomegaly. Incidental MCL was diagnosed in
the splenectomy. Following the surgeries he developed
high fever and acute abdomen, tentatively managed as
surgery-related peritonitis up to 3 months; he finally
underwent debriding omentectomy. Microscopic examination illustrates numerous perivascular and nodular atypical lymphocytes aggregates with associated fat
necrosis. No metastatic carcinoma was seen. Immunohistochemistry demonstrates that the lymphocytes are
positive for CD20, PAX5, and BCL-1 (cyclin D1). Special
stains and cultures for microorganisms were negative.
These findings indicate diffuse peritoneal involvement
by MCL, ruling out an infectious etiology.
Conclusion: This is the first report of post-surgery
peritoneal dissemination by MCL mimicking peritonitis and omentitis, implicating the surgery as the main
culprit of peritoneal lymphomatosis, suggesting that
peritoneal seeding and lymphomatosis could be potential complication of surgeries in MCL patients. Additionally, our case demonstrates that peritoneal MCL
recurrence could be under recognized in patient(s)
even after curative resection of an unrelated entity; the
underlying mechanism is likely related to wound contamination, peritoneal trauma, postoperative immunosuppression and potentially various surgical routes.
17. Quantification of Focal Oligodendroglial Hyperplasia in a Patient With Epilepsy
H.C. Rutherford,1 J.A.E. Radic,2 R.J.B. Macaulay,1
R.M. Sadler,3 T. Bardouille,4 A. Ross,5 and D.B. Clarke2
1 Department of Anatomic Pathology, H. Lee
Moffitt Cancer Center & Research Institute, Tampa,
Florida, Divisions of 2Neurosurgery and 3Neurology,
University of South Florida, Tampa, Florida, 4Biomedical Translational Imaging Centre, IWK Health Centre,
Halifax, Nova Scotia, Canada, 5Division of Nuclear
Medicine, Department of Diagnostic Radiology, Dalhousie University, Halifax, Nova Scotia, Canada
A 39-year-old female with medically refractory
epilepsy underwent MRI, EEG as well as magneto-encephalography, and fluorodeoxyglucose positron emission tomography (PET). A lesion in the right inferomesial frontal lobe was identified, and subdural electrode
recordings demonstrated seizure onset from this area.
The patient underwent surgical resection of the lesion.
Histopathology revealed focally increased cell density
in the subcortical white matter. Overlying cortex was
April 2016, Vol. 23, No. 2
unremarkable, with no dysmorphic or disorganized
neurons, and no balloon cells to suggest focal cortical
dysplasia (FCD). The supernumerary cells were morphologically similar to oligodendrocytes, and occupied
normally myelinated white matter. Rare mildly reactive
astrocytes were found. There was a gradual tapering
of cell density towards deeper white matter regions.
No perinuclear halo, microcalcification, microcystic
change, nuclear pleomorphism or mitotic activity was
evident, militating against oligodendroglioma (ODG).
There was no pooling of mucinous material or ‘floating’ neuron to suggest dysembryoplastic neuroepithelial tumor (DNT). Immunohistochemistry (IHC) for
IDH1 R13H and BRAF V600E were negative. Quantification of oligodendrocytes in high (3350.5 cells/mm2),
compared to low (1683.6 cells/mm2) density areas in
adjacent normal-appearing white matter was highly
statistically significant (P < .001). A descriptive diagnosis of focal oligodendroglial hyperplasia (ODH) was
eventually proposed. ODH is a poorly characterized
subtype of microdysgenesis distinct from FCD, and a
rare cause of medically refractory epilepsy. No carefully documented case is available in the medical literature for comparison. Histologically, it is important that
ODH is differentiated from ODG and DNT. Because
of the paucity of published cases, the clinical impact
of surgical resection of ODH found within the seizure
onset zone is uncertain. The patient continues to have
1-2 nocturnal partial (focal with dyscognitive features)
seizures per month, but has had no convulsive seizure since her surgery (4.5-year follow-up). This report
quantifies the density of oligodendrocytes within different neighboring portions of ODH and documents
its morphologic and IHC features through analysis of a
right inferomesial frontal lobe lesion.
18. Electronic Atlas of Epithelioid Soft Tissue Tumors: A Pilot for Pathology Atlas “App” Development
Robert P. Seifert1 and Marilyn M. Bui1,2
1Department of Pathology and Cell Biology, University of South Florida Morsani College of Medicine,
Tampa, Florida, 2Department of Anatomic Pathology,
H. Lee Moffitt Cancer Center & Research Institute,
Tampa, Florida
Background: The “Atlas of Pancreatic Pathology
and Cytopathology” apps developed by Johns Hopkins
Pathology allow for accessible, rapid review of pancreatic histology. However, pathology, as a specialty, is
lagging in integrating handheld devices into training
and daily practice. An electronic atlas “app” for handheld devices can correlate clinical and histologic features quickly. The scarcity of pathology apps can be
attributed to lack of time, funding and programming
knowledge. To address this gap, we developed a proofof-concept atlas app encompassing soft tissue tumors
with epithelioid morphology.
April 2016, Vol. 23, No. 2
Methods: A free video game development platform (GameMaker Studio v1.4, YoYo Games, Dundee,
Scotland) was selected because it is export platform
agnostic and requires minimal programming knowledge. This platform has a relatively low entry cost:
$299/export module. Static digital images were retrieved from our institution’s whole slide image database. A gallery user interface was designed within
GameMaker including query and filter functions.
Results: The app currently includes 11 diagnoses
spanning 170 images; each with a brief description
along with expected immunohistochemical staining
patterns. Our app also acts as a template, allowing pathologists with no programming knowledge to create
an atlas of their own. Following a brief tutorial, enduser pathologists may take our framework, add their
own diagnoses and publish their atlas to the web.
Conclusions: With minimal programming knowledge and expense, a multiplatform pathology app can
be developed. This app will be tested in a pathology
residency focus group to further evaluate its efficacy.
This may provide a unique opportunity to enhance pathology education in the absence of resources.
19. Spindle Cell/Sclerosing Rhabdomyosarcoma Is
a New and Underdiagnosed Entity: A Clinical and
Tissue Microarray Immunohistochemical Study
With Literature Review
Yunguang Liu, Maryam Tahmasbi, and Marilyn M. Bui
Department of Anatomic Pathology, H. Lee Moffitt
Cancer Center & Research Institute, Tampa, Florida
Background: In adults rhabdomyosarcoma (RMS)
accounts for less than 2% of all sarcomas. Spindle
cell/sclerosing RMS is a newly recognized variant by
2013 WHO Classification of Tumors of Soft Tissue and
Bone. It accounts for 5% to 10% of RMS with male to
female ratio up to 6:1. One previous study suggested
that spindle cell RMS may have not as rare as previously thought. Recently we encountered a case that was
thought to be leiomyosarcoma of soft tissue. However,
after further review and work up, the diagnosis was
revised to spindle cell/sclerosing RMS. Therefore we
intend to study how often spindle cell/sclerosing RMS
is misdiagnosed for leiomyosarcoma and how to avoid
this diagnostic pitfall.
Materials and Methods: Literature review was
conducted to identify the most sensitive and specific
immunohistochemical (IHC) markers of RMS and the
expression pattern of myogenin and myoD1 in leiomyosarcoma. Subsequent to the index case, 3 cases with previously diagnosed leiomyosarcomas were evaluated in
real time and subjected to myogenin and myoD1 stains.
A tissue microarray (TMA) consisting of 42 cases of previously diagnosed leiomyosarcoma was subjected to
IHC of myogenin (MYF4, 1:20-1:40 dilution, Newcastle,
UK) and myoD1 (EP212, 0.01–1.0 µg/ml, Rocklin, CA).
Cancer Control 185
Results: According to the literature review myogenin and myoD1 are sensitive (97%) and specific
(90% vs 91%) markers for RMS. Most authors agree that
one of these markers is necessary for a diagnosis of
RMS. Leiomyosarcoma rarely expresses myogenin or
myoD1, but co-expression was not reported. In comparing the index case and the three subsequent cases,
two of the three cases were reclassified as RMS (one
spindle cell, one pleomorphic). The leiomyosarcoma
TMA (age range 26-82, mean ± SE 55.8 ± 2.2; male 15,
female 27; GYN origin 9, non-GYN origin 33) revealed
one case expressing myoD1 not myogenin. Repeat
staining on the corresponding whole slide section of
the tumor rendered the same result.
Conclusions: Spindle cell/sclerosing RMS can
be sometimes misdiagnosed as leiomyosarcoma due
to the overlapping histological features and immunohistochemical profile. However, judicious use of myogenin and myoD1 can be helpful to achieve an accurate
and definitive diagnosis. Co-expression of myogenin
and myoD1 is strong confirmation.
save the date
3 RD ANNUAL MOFFITT CANCER CENTER PATHOLOGY SYMPOSIUM
Transforming Discovery
into Practice
A 2.5 day program geared to pathologists,
cancer clinicians, and laboratory
professionals focused on improving
practice by implementation of recent
discoveries in biomarkers, molecular
genomics, laboratory improvements,
and personalized medicine.
Friday, February 3 –
Sunday, February 5, 2017
Sarasota, Florida
186 Cancer Control
April 2016, Vol. 23, No. 2
Ten Best Readings Relating
to Ocular Oncology
Onken MD, Worley LA, Char DH, et al. Collaborative Ocular Oncology Group report number
1: prospective validation of a multi-gene prognostic assay in uveal melanoma. Ophthalmology.
2012;119(8):1596-1603.
The prognostic performance of a gene expression
profiling assay covering 15 genes was assessed in a
prospective, multicenter study. Primary posterior uveal
melanomas were assigned to classes 1 (low metastatic
risk) and 2 (high metastatic risk) prognostic categories. The assay had a high technical success rate and
provided a highly significant improvement in rates
of prognostic accuracy over clinical tumor, node, and
metastasis classification.
Jakobiec FA, Mendoza PR. Eyelid sebaceous carcinoma: clinicopathologic and multiparametric immunohistochemical analysis that includes adipophilin.
Am J Ophthalmol. 2014;157(1):186-208.
This retrospective, clinicopathologic study sought
to review the fine cytopathological features and immunohistochemistry of eyelid sebaceous carcinoma.
The authors concluded that immunohistochemistry
can make a significant contribution to the diagnosis
of sebaceous carcinoma, and that p53 and vesicular
granular adipophilin positivity could reliably supplement routine microscopic diagnosis of infiltrative tumors and could be used in paraffin sections, thereby
obviating the oil red O staining of frozen sections.
Corrêa ZM, Augsburger JJ. Independent prognostic
significance of gene expression profile class and
largest basal diameter of posterior uveal melanomas.
Am J Ophthalmol. 2016;162:20-27.
The authors of a prospective, interventional case
series set out to determine whether select clinical
prognostic factors for metastatic uveal melanoma
were prognostically significant in multivariate models that incorporated a gene expression profile class
of tumor cells. Gene expression profile class was
the strongest prognostic factor for metastatic death,
followed by tumor larger basal diameter, thickness, and intraocular tumor location. This study
also showed that larger basal diameter and gene
expression profile class of tumor were both independent prognostic factors for metastasis and
metastatic-related death.
Plaza JA, Mackinnon A, Carrillo L, et al. Role of
immunohistochemistry in the diagnosis of sebaceous carcinoma: a clinicopathologic and immunohistochemical study. Am J Dermatopathol.
2015;37(11):809-821.
The diagnosis of sebaceous carcinoma, which is
an uncommon malignant epithelial neoplasm with a
predilection for the periocular region, can be difficult
to discern upon the initial presentation of a patient because the condition can clinically and histopathologically resemble other common benign and malignant
epithelial lesions. A diagnosis of sebaceous carcinoma
is made by confirmation of sebaceous differentiation
of neoplastic cells; however, its recognition may be
sometimes difficult and may require ancillary studies. The results of this study suggest that adipophilin
represents a sensitive and reliable marker for diagnosing sebaceous carcinoma. Including various epithelial markers in the panel could also be of help if
adequately used.
Field MG, Decatur CL, Kurtenbach S, et al. PRAME
as an independent biomarker for metastasis in uveal
melanoma. Clin Cancer Res. 2016;22(5):1234-1242.
Some class 1 uveal melanoma tumors are known
to metastasize. Among class 1 tumors, the most significant predictor of metastasis was PRAME messenger
RNA expression. The 5-year actuarial rate of metastasis was 0% for class 1 (PRAME–), 38% for class 1
(PRAME+), and 71% for class 2 tumors. The rate of
median metastasis-free survival for patients with class
1 (PRAME+) tumors was 88 months compared with
32 months for patients with class 2 tumors. PRAME
expression was associated with SF3B1 mutations and
larger tumor diameter. The study findings suggested
that PRAME is an independent prognostic biomarker
in uveal melanoma; therefore, its presence is associated with an increased risk of metastasis in patients
with class 1 or disomy 3 tumors.
April 2016, Vol. 23, No. 2
Riemens A, Bromberg J, Touitou V, et al. Treatment
strategies in primary vitreoretinal lymphoma: a
17-center European collaborative study. JAMA
Ophthalmol. 2015;133(2):191-197.
In this study, researchers evaluated the outcomes
of treatment regimens used for primary vitreoretinal
lymphoma for the prevention of subsequent central
nervous system lymphoma (CNSL). Therapy to prevent
CNSL included ocular radiotherapy, ocular chemotherapy, or both (group A); extensive systemic treatment (group B); and a combination of ocular and
extensive treatment (group C). CNSL developed in
32% of group A, 43% of group B, and 39% of group C.
The 5-year cumulative survival rate was lower in those
with CNSL compared with those without CNSL (68%)
Cancer Control 187
and was similar among all treatment groups. The most
common adverse event was acute renal failure. The
researchers of this study were unable to prove that
use of systemic chemotherapy could prevent CNSL.
Chan CC, Rubenstein JL, Coupland SE, et al. Primary
vitreoretinal lymphoma: a report from an International Primary Central Nervous System Lymphoma Collaborative Group symposium. Oncologist.
2011;16(11):1589-1599.
Primary vitreoretinal lymphoma (PVRL) commonly masquerades as posterior uveitis and has a unique
tropism for the retina and central nervous system
(CNS). More than 15% of patients with primary CNS
lymphoma develop intraocular lymphoma, usually
occurring in the retina, vitreous, or both areas. Conversely, 65% to 90% of patients with PVRL develop
CNS lymphoma. PVRL is most often diagnosed using
both histology to identify lymphoma cells in the vitreous or retina and immunohistochemistry to indicate
monoclonality. Optimal therapy for PVRL has not yet
been defined, but it is sensitive to radiotherapy and
exhibits high responsiveness to intravitreal methotrexate or rituximab. Although systemic chemotherapy
alone can result in high response rates in patients with
PVRL, a high relapse rate is seen in this population.
from follicular lymphoma. The researchers noted that
an undescribed “multifocal RLH” syndrome must be
distinguished from follicular lymphoma. Conjunctival
RLH can usually be managed surgically without radiotherapy, but “multifocal RLH” required systemic treatment in 2 of 3 patients. Follicular lymphoma requires
systemic chemotherapy if discovered beyond stage 1E.
Al-Jamal RT, Cassoux N, Desjardins L, et al. The pediatric choroidal and ciliary body melanoma study:
a survey by the European Ophthalmic Oncology
Group. Ophthalmology. 2016;123(4):898-907.
In this retrospective, multicenter, observational
trial, European researchers collected comprehensive data on choroidal and ciliary body melanoma
in children in order to determine whether children
younger than 18 years of age, those of the male sex,
and those without ciliary body involvement have a
more favorable survival prognosis than young adults
18 to 24 years of age, those of the female sex, and
those with ciliary body involvement. By multivariate
analysis, being a young adult, having a higher tumor,
node, and metastasis stage, and being of the female
sex independently predicted less favorable rates of
survival. Ciliary body involvement and cell type were
not associated with rates of survival.
Grossniklaus HE. Progression of ocular melanoma
metastasis to the liver: the 2012 Zimmerman lecture.
JAMA Ophthalmol. 2013;131(4):462-469.
Grossniklaus evaluated the immunohistochemical
and histological findings of uveal melanoma that metastasizes to the liver. Stage 1 metastases were identified in the sinusoidal spaces of 90% of study patients;
these metastases were avascular and lacked mitotic
activity. Stages 2 and 3 metastases were found in all
study patients. Immunohistochemical stains were positive for S100 or HMB45 in all tumors. Overall, stage 1
metastases outnumbered stage 2 metastases (which
outnumbered stage 3 metastases). The mean vascular
density and mitotic index increased from stage 2 to
stage 3 metastases. The architecture of stage 2 metastases mimicked the surrounding hepatic parenchyma,
whereas stage 3 metastases exhibited either lobular
or portal growth patterns.
Stacy RC, Jakobeic FA, Schoenfield L, et al. Unifocal
and multifocal lymphoid hyperplasia vs follicular
lymphoma of the ocular adnexa. Am J Ophthalmol.
2010;150(3):412-426.
This study was conducted so researchers could
characterize the differentiating histopathological and
immunophenotypic features of reactive lymphoid hyperplasia (RLH) and follicular lymphoma of the ocular
adnexa. Microscopic analysis with immunohistochemical staining can be reliably used to distinguish RLH
188 Cancer Control
April 2016, Vol. 23, No. 2