Alberts Principles and Practice of Ophthalmology Volume II PDF

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 1373

2

Albert: Albert & Jakobiec's Principles &


Practice of Ophthalmology

THIRD EDITION

Daniel M. Albert, MD MS
Chair Emeritus, F. A. Davis Professor and Lorenz F. Zimmerman Professor, Department of
Ophthalmology and Visual Sciences, Retina Research Foundation Emmett A. Humble Distinguished
Director, of the Alice R. McPherson, MD, Eye Research Institute, University of Wisconsin
Medical School, Madison, Wisconsin, USA
Joan W. Miller, MD
Henry Willard Williams Professor of Ophthalmology, Chief and Chair, Department of
Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston,
Massachusetts, USA
Associate Editors:
Dimitri T. Azar, MD
B.A. Field Chair of Ophthalmologic Research, Professor and Head, Department of Ophthalmology
and Visual Sciences, University of Illinois Eye and Ear Infirmary, Chicago, Illinois, USA
Barbara A. Blodi, MD
Associate Professor, Department of Ophthalmology and Visual Sciences, University of Wisconsin
Medical School, Madison, Wisconsin, USA
Managing Editors:
Janet E. Cohan
Administrative Manager, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary,
Harvard Medical School, Boston, Massachusetts, USA
Tracy Perkins, MPH
Administrative Director, Alice R. McPherson, MD Eye Research Institute, University of
Wisconsin Medical School, Madison, Wisconsin, US
DEDICATION

To CLAES H. DOHLMAN
Superb surgeon, mentor, teacher, innovator and friend.
D.M.A & J.W.M
SAUNDERS ELSEVIER
SAUNDERS is an imprint of Elsevier Inc.
? 2000, 1994 by W.B Saunders Company
? 2008, Elsevier Inc. All rights reserved.
First published 2008
First edition 1994
Second edition 2000
Third edition 2008
No part of this publication may be reproduced, stored in a
retrieval system, or transmitted in any form or by any means,
electronic, mechanical, photocopying, recording or otherwise,
without the prior permission of the Publishers. Permissions may
be sought directly from Elsevier's Health Sciences Rights
Department, 1600 John F. Kennedy Boulevard, Suite 1800,
Philadelphia, PA 19103-2899, USA: phone: (+1) 215 239 3804;
fax: (+1) 215 239 3805; or, e-mail:
healthpermissions@elsevier.com. You may also complete your
request on-line via the Elsevier homepage (http://
www.elsevier.com), by selecting ‘Support and contact’ and then
‘Copyright and Permission’.
ISBN: 978-1-4160-0016-7
Notice
Medical knowledge is constantly changing. Standard safety
precautions must be followed, but as new research and clinical
experience broaden our knowledge, changes in treatment and drug
therapy may become necessary or appropriate. Readers are
advised to check the most current product information provided
by the manufacturer of each drug to be administered to verify
the recommended dose, the method and duration of
administration, and contraindications. It is the responsibility
of the practitioner, relying on experience and knowledge of the
patient, to determine dosages and the best treatment for each
individual patient. Neither the Publisher nor the author assume
any liability for any injury and/or damage to persons or
property arising from this publication.
The Publisher
Preface to the 3rd Edition

Do clinicians and trainees really need textbooks anymore? In an medicine, and genetics information has been incorporated into
era of ever-expanding connectivity and immediate access to all sections. Finally, the last section of the textbook headed by
published articles, why would anyone consult a textbook, which Kathy Colby and Nancy Holekamp is a section on Ethics and
by its very nature is incomplete before it is even published? No Professionalism topics that are increasingly important to
doubt these are strange questions coming from the editors of practicing clinicians, and an ACGME requirement for resident
the third edition of the most popular multi-volume ophthalmic training. A concerted effort was made throughout the third
textbook, but they must be asked and answered. Our answer edition to complement the text with diagrams, line drawings
is an unequivocal “yes”! Books like this serve an extremely and color figures. In addition, each chapter contains a key
important function – that of a repository for expert reviews of points section. Overall, the third edition has exceeded the
our current understanding of ophthalmic health and disease. expectations of all of the editors. We were pleased by the
The chapters and sections in Albert and Jakobiec are an enthusiasm of new and returning authors, more than 600 in
important resource for the clinician and student, providing a total, as well as new and returning section editors, and were
comprehensive information base on an extensive list of topics. excited by the teamwork and cooperation shown in upgrading
Of course journal articles continue to be the most useful source and improving this important project. The result is a definitive
of information about new developments in the field but they do textbook in ophthalmology, available in hardcover and by web
not replace books. Constraints on the length of journal articles, access.
inattention to the provenance of the ideas they contain, and an The editorial team has been a wonderful collaboration and
understandable tendency to self-promote the authors’ thesis, the senior editors are very grateful for the prodigious efforts of
limit the value of many “original contributions.” Readers Drs. Dimitri Azar and Barbara Blodi. We were saddened that Dr.
of journal articles forearmed with information found in an Frederick Jakobiec, a co-founder of this project and co-editor on
encyclopedic text can place these articles into perspective. editions 1 and 2, was unable to participate as an editor in the
Thus, the two sources are complimentary. In a very real sense third edition, although still contributing as a co-author. We look
this textbook serves as a springboard to the constantly forward to his return to the ophthalmology community, and we
expanding universe of published scientific literature. can report that Dr. Jakobiec is pleased and supportive of the
What is new in the third edition? The second edition (2002) upcoming 3rd edition of the textbook named for him and Dr.
was a reworking of the very successful first edition (1996) of Albert. All of the editorial team is most appreciative of the
Albert and Jakobiec’s Principles and Practice of Ophthalmology. unstinting and generous support of Elsevier Publishing; in
For the third edition we undertook a critical evaluation of each particular the leadership of the senior editor, Russell Gabbedy,
section and chapter to ensure that topics were well-covered with and the hard work and diligence of Zak Knowles, contributing
minimal redundancy, that new areas of practice and research editor, whose efforts in collecting and coordinating chapters, as
were adequately described, and that topics that were over- well as initial editing of chapters were unsurpassed. The
represented could be substantially shortened or deleted. This managing editors, Tracy Perkins and Janet Cohan, provided
evaluation involved all of the editors (Dan Albert, Joan Miller, important coordination between the authors, section editors,
Barbara Blodi and Dimitri Azar) as well as new and returning editors and publisher, and handled all of their responsibilities
section editors. As an example, under the direction of Dimitri with aplomb. Above all, the contributing authors who wrote the
Azar, we incorporated a new section on refractive surgery that chapters and the section editors who delineated the section
provides the principles of refractive surgery as well as useful content and edited the component chapters deserve the greatest
descriptions of evaluation techniques and procedures. The credit for the superb quality of the textbook.
Oncology section was substantially expanded and revised under We sincerely hope that the third edition of Albert and
the section editorship of Evangelos Gragoudas and Joan Jakobiec’s Principles and Practice in Ophthalmology provides
O’Brien. Pediatrics was also extensively revised by David ophthalmologists and trainees with a gateway into the
Hunter and Monte Mills, and the Pharmacology and Toxicology wonderful science and art of ophthalmology in order to provide
sections were combined and revised under the direction of Mark the best care for our patients, and to continually advance
Abelson. Barbara Blodi and Joan Miller reworked the extensive our field.
retina section, to include current techniques, new diagnostic
modalities (including OCT), and new drug therapies. The Daniel M. Albert and Joan W. Miller
human genome project and modern genetics are revolutionizing

xvii
Preface to the 1st Edition

“INCIPIT.” The medieval scribe would write this Latin word, with an anatomic woodcut (Fig. 2) and then lists in tabular
meaning so it begins, to signal the start of the book he was form various eye conditions, including strabismus, paralysis,
transcribing. It was a dramatic word that conveyed promise of amblyopia, and nictalops. The work uses a distinctly Greco
instruction and delight. In more modern times INCIPIT has Roman terminology, presenting information on the parts of the
been replaced by the PREFACE. It may be the first thing the eye and their affections, including conjunctivitis, ophthalmia,
reader sees, but it is, in fact, the last thing the author writes carcinoma, and “glaucoma.” The book concludes with a remedy
before the book goes to press. I appreciate the opportunity to collection similar to that found in the Büchlin. Most significant
make some personal comments regarding Principles and in the association of Leonhart Fuchs with this book is the fact
Practice of Ophthalmology. that a properly trained and well recognized physican addressed
One of the most exciting things about writing and editing a the subject of ophthalmology.
book in a learned field is that it puts the authors and editors in Julius Hirschberg, the ophthalmic historian, noted that
touch with those who have gone before. Each author shares Fuch’s Alle Kranckheyt, along with the anonymous Büchlin,
with those who have labored in past years and in past centuries apparently influenced Georg Bartisch in his writing of Das Ist
the tasks of assessing the knowledge that exists in his or her Augendienst. This latter work, published in 1583, marked the
field, of determining what is important, and of trying to convey founding of modern ophthalmology. Bartisch (1535–1606) was
it to his or her peers. In the course of the work the author an itinerant barber surgeon but nonetheless a thoughtful and
experiences the same anticipation, angst, and ennui of those skillful surgeon, whose many innovations included the first
who have gone before. He or she can well envision the various procedure for extirpation of the globe for ocular cancer. Bartisch
moments of triumph and despair that all authors and editors proposed standards for the individual who practices eye surgery,
must feel as they organize, review, and revise the accumulating noting that rigorous training and concentration of effort were
manuscripts and reassure, cajole, and make demands of their needed to practice this specialty successfully.
fellow editors, authors, and publisher. By the late 16th century, eye surgery and the treatment of eye
This feeling of solidarity with early writers becomes even disease began to move into the realm of the more formally
more profound when one is a collector and reviewer of books, trained and respected surgeon. This is evidenced by Jacques
and conversant with the history of one’s field. In Ecclesiastes Guillemeau’s Traité des Maladies de L’Oeil, published in 1585.
it is stated, “of the making of books, there is no end” (12:12). Guillemeau (1550–1612) was a pupil of the surgical giant
Indeed, there are more books than any other human artifact on Ambroise Paré, and his book was an epitome of the existing
earth. There is, however, a beginning to the “making of books” knowledge on the subject.
in any given field. The first ophthalmology book to be published The transition from couching of cataracts to the modern
was Benvenuto Grassi’s De Oculis in Florence in 1474. Firmin method of treating cataracts by extraction of the lens, as
Didot in his famous Bibliographical Encyclopedia wrote that introduced by Jacques Daviel in 1753, further defined the skill
Grassus, an Italian physician of the School of Solerno, lived in and training necessary for the care of the eyes. The initiation
the 12th century and was the author of two books, the Ferrara of ophthalmology as a separate specialty within the realm of
Quarto (1474) and the Venetian Folio (1497). Eye care in the medicine and surgery was signaled by the publication of George
15th century was in the hands of itinerant barber surgeons and Joseph Beer’s two volume Lehre von den Augenkrankheiten in
quacks, and a treatise by a learned physician was a remarkable 1813–1817. Beer (1763–1821) founded the first eye hospital in
occurrence. The next book on the eye to appear was an anony- 1786 in Vienna, and his students became famous ophthalmic
mous pamphlet written for the layperson in 1538 and entitled surgeons and professors throughout Europe.
Ein Newes Hochnutzliches Büchlin von Erkantnus der In England, it was not only the demands of cataract surgery
Kranckheyten der Augen. Like Principles and Practice of but also the great pandemic of trachoma following the Napo-
Ophthalmology, the Büchlin stated its intention to provide leonic wars that led to the establishment of ophthalmology as a
highly useful knowledge of eye diseases, the anatomy of the eye, recognized specialty. Benjamin Travers (1783–1858) published
and various remedies. It was illustrated with a fullpage woodcut the earliest treatise in English on diseases of the eye, A Synopsis
of the anatomy of the eye (Fig. 1). At the conclusion of the book, of the Diseases of the Eye, in 1820. In the United States,
the publisher, Vogtherr, promised to bring more and better acceptance of ophthalmology as a specialty had to await the
information to light shortly, and indeed, the next year he description of the ophthalmoscope by Helmholtz in 1851,
published a small book by Leonhart Fuchs (1501–1566) entitled and the additional special skills that using the early primitive
Alle Kranckheyt der Augen. “Augenspiegel” required.
Fuchs, a fervent Hippocratist, was Professor first of Philo- As the complexity of ophthalmology increased and as sub-
sophy and then of Medicine at Ingolstadt, Physician of the specialization began to develop in the 19th century, multi-
Margrave Georg of Brandenburg, and finally Professor at authored books began to appear. This culminated in the
Tübingen for 31 years. Like the earlier Büchlin, his work begins appearance in 1874 of the first volume of the GraefeSaemisch xix
Preface to the 1st Edition

FIGURE 1.
FIGURE 2.

Handbuch. The final volume of this great collective work, of The writing of these two series, the Textbook and the System,
which Alfred Carl Graefe (1830–1899) and Edwin Theodor has occupied all my available time for half a century. I cannot
Saemisch (1833–1909) were editors, appeared in 1880. The deny that its completion brings me relief on the recovery of my
definitive second edition, which for more than a quarter of a freedom, but at the same time it has left some sadness for I have
century remained the most comprehensive and authoritative enjoyed writing it. As Edward Gibbon said on having written
work in the field, appeared in 15 volumes between 1899 and the last line of The Decline and Fall of the Roman Empire:
1918. The great French counterpart to the Graefe Saemisch “A sober melancholy has spread over my mind by the idea
Handbuch was the Encyclopédie Française d’Ophtalmologie, that I have taken everlasting leave of an old and agreeable
which appeared in nine volumes (1903–1910), edited by Octave companion.”
Doin, and filled a similar role for the French speaking Duke Elder adds a final line that I hope will be more àpropos
ophthalmologist. to the present editors and contributors. “At the same time
In 1896, the first of four volumes of Norris and Oliver’s the prayer of Sir Francis Drake on the eve of the attack of the
System of Diseases of the Eye was published in the United Spanish Armada is apposite: ‘Give us to know that it is not the
States. The senior editor, Dr. William Fisher Norris beginning but the continuing of the same until it is entirely
(1839–1901), was the first Clinical Professor of Diseases of finished which yieldeth the true glory.”’ The void that developed
the Eye at the University of Pennsylvania. Charles A. Oliver as the Duke Elder series became outdated has been partially
(1853–1911) was his student. Norris considered the System filled by many fine books, notably Thomas Duane’s excellent 5
to be his monumental work. For each section he chose an volume Clinical Ophthalmology.
outstanding authority in the field, having in the end more than Inspiration to undertake a major work such as this is derived
60 American, British, Dutch, French, and German ophthal- not only from the past books but also from teachers and role
mologists as contributors. Almost 6 years of combined labor on models. For me, this includes Francis Heed Adler, Harold
the part of the editors was needed for completion of the work. G. Scheie, William C. Frayer, David G. Cogan, Ludwig von
In 1913, Casey A. Wood (1856–1942) introduced the first of Sallmann, Alan S. Rabson, Lorenz E. Zimmerman, Frederick C.
his 18 volumes of the American Encyclopedia and Dictionary Blodi, Claes H. Dohlman, and Matthew D. Davis.
of Ophthalmology. The final volume appeared in 1921. Drawn Whereas the inspiration for the present text was derived from
largely from the Graef Saemisch Handbuch and the Encyclo- Duke Elder’s Textbook and System and from teachers and role
pédie Française d’Ophtalmologie, Wood’s Encyclopedia models, learning how to write and organize a book came for
provided information on the whole of ophthalmology through a me from Adler’s Textbook of Ophthalmology, published by
strictly alphabetic sequence of subject headings. W.B. Saunders. This popular textbook for medical students and
The book from which the present work draws inspiration general practitioners was first produced by Dr. Sanford Gifford
is Duke Elder’s Textbook of Ophthalmology (7 volumes; 1932) (1892–1945) in 1938. Francis Heed Adler (1895–1987), after
and particularly the second edition of this work entitled System writing the 6th edition, published in 1962, invited Harold G.
of Ophthalmology (15 volumes, published between 1958 and Scheie (1909–1989), his successor as Chairman of Ophthal-
1976). The System of Ophthalmology was written by Sir mology at the University of Pennsylvania, and myself to take
Stewart Duke Elder (1898–1978) in conjunction with his over authorship. We completely rewrote this book and noted
colleagues at the Institute of Ophthalmology in London. In in the Preface to the 8th edition, published in 1969: “This
1976, when the last of his 15 volumes appeared, Duke Elder book aims to provide the medical student and the practicing
xx wrote in the Preface: physician with a concise and profusely illustrated current text,
Preface to the 1st Edition

organized in a convenient and useable manner, on the eye and textbook or system previously published, in terms of quantity
its disorders. It is hoped that the beginning, or even practicing, and quality and usefulnesss of the pictures.
ophthalmologist may find it of value.” In specific terms, in editing the book we tried to identify
In 1969 it was apparent that even for the intended audience, and eliminate errors in accuracy. We worked to provide as
contributions by individuals expert in the subspecialties of uniform a literary style as is possible in light of the numerous
ophthalmology were required. The book was published in contributors. We attempted to make as consistent as possible
Spanish and Chinese editions and was popular enough to the level of detail presented in the many sections and chapters.
warrant an updated 9th edition, which appeared in 1977. One Related to this, we sought to maintain the length according to
of the high points of this work was interacting with John our agreed upon plan. We tried, as far as possible, to eliminate
Dusseau, the Editor in Chief for the W.B. Saunders Company. repetition and at the same time to prevent gaps in information.
As a 10th edition was contemplated, I became increasingly We worked to direct the location of information into a logical
convinced that what was needed in current ophthalmology was and convenient arrangement. We attempted to separate the
a new, comprehensive, well illustrated set of texts intended basic science chapters to the major extent into the separate
for the practicing ophthalmologist and written by outstanding Basic Sciences volume, but at the same time to integrate basic
authorities in the field. I envisioned a work that in one series of science information with clinical detail in other sections as
volumes would provide all of the basic clinical and scientific needed. These tasks were made challenging by the size of the
information required by practicing ophthalmologists in their work, the number of authors, and the limited options for
everyday work. For more detailed or specialized information, change as material was received close to publishing deadlines.
this work should direct the practitioner to the pertinent journal We believe that these efforts have succeeded in providing
articles or more specialized publications. As time progressed, a ophthalmologists and visual scientists with a useful resource in
plan for this work took shape and received support from the their practices. We shall know in succeeding years the level of
W.B. Saunders Company. this success and hope to have the opportunity to improve all
Memories of the formative stages of the Principles and these aspects as the book is updated and published in future
Practice of Ophthalmology remain vivid: Proposing the project editions. Bacon wrote: “Reading maketh a full man, conference
to Frederick Jakobiec in the cafeteria of the Massachusetts a ready man, and writing an exact man.” He should have added:
Eye and Ear Infirmary in early 1989. Having dinner with Lewis Editing maketh a humble man.
Reines, President and Chief Executive Officer, and Richard I am personally grateful to a number of individuals for
Zorab, Senior Medical Editor, at the Four Seasons Hotel in making this book a reality. Nancy Robinson leads the list. Her
May 1989, where we agreed upon the scope of the work. My intelligent, gracious, and unceasing effort as Managing Editor
excitement as I walked across the Public Garden and down was essential to its successful completion. Mr. Lewis Reines,
Charles Street back to the Infirmary, contemplating the work President of the W.B. Saunders Company, has a profound
we were to undertake. Finalizing the outline for the book in knowledge of publishing and books that makes him a worthy
Henry Allen’s well stocked “faculty lounge” in a dormitory at successor to John Dusseau. Richard Zorab, Senior Medical
Colby College during the Lancaster Course. Meeting with Editor, and Hazel N. Hacker, Developmental Editor, are
members of the Harvard Faculty in the somber setting of the thoroughly professional and supportive individuals with whom
rare book room to recruit the Section Editors. Persuading Nancy it was a pleasure to work. Many of the black and white
Robinson, my able assistant since 1969, to take on the job of illustrations were drawn by Laurel Cook Lhowe and Marcia
Managing Editor. The receipt of our first manuscript from Dr. Williams; Kit Johnson provided many of the anterior segment
David Cogan. photographs. Archival materials were retrieved with the aid
We considered making this work a departmental under- of Richard Wolfe, Curator of Rare Books at the Francis A.
taking, utilizing the faculty and alumni of various Harvard Countway Library of Medicine, and Chris Nims and Kathleen
programs. However, the broad scope of the series required Kennedy of the Howe Library at the Massachusetts Eye and Ear
recruitment of outstanding authors from many institutions. Infirmary.
Once the Section Editors were in place, there was never any The most exciting aspect of writing and editing a work of
doubt in my mind that this work would succeed. The Section this type is that it puts one in touch with the present day
Editors proved a hardworking and dedicated group, and their ophthalmologists and visual scientists as well as physicians
choice of authors reflects their good judgment and persuasive training to be ophthalmologists in the future. We hope that this
abilities. I believe that you will appreciate the scope of book will establish its own tradition of excellence and useful-
knowledge and the erudition. ness and that it will win it a place in the lives of ophthal-
The editorship of this book provided me not only with an mologists today and in the future.
insight into the knowledge and thinking of some of the finest “EXPLICIT,” scribes wrote at the end of every book.
minds in ophthalmology but also with an insight into their EXPLICIT means it has been unfolded. Olmert notes in The
lives. What an overwhelmingly busy group of people! Work was Smithsonian Book of Books, “the unrolling or unfolding of
completed not through intimidation with deadlines but by knowledge is a powerful act because it shifts responsibility from
virtue of their love of ophthalmology and their desire to share writer to reader.... Great books endure because they help us
their knowledge and experience. The talent, commitment, interpret our lives. It’s a personal quest, this grappling with the
persistence, and good humor of the authors are truly what made world and ourselves, and we need all the help we can get.” We
this book a reality. hope that this work will provide such help to the professional
It was our intent to present a work that was at once scholarly lives of ophthalmologists and visual scientists.
and pragmatic, that dealt effectively with the complexities
and subtleties of modern ophthalmology, but that did not DANIEL M. ALBERT, M.D., M.S.
overwhelm the reader. We have worked toward a series of MADISON, WISCONSIN
volumes that contained the relevant basic science information
to sustain and complement the clinical facts. We wanted a
well illustrated set that went beyond the illustrations in any

xxi
List of Contributors

Juan-Carlos Abad MD Lloyd P Aiello MD PhD Ibrahim A Al Jadaan MD


Clinica Oftalmologica de Medellin Director of Beetham Eye Institute Chief
Medellin Section Head of Eye Research Glaucoma Division
Colombia Joslin Diabetes Center King Khaled Eye Specialist Hospital
Beetham Eye Institute Riyadh
Mark B Abelson MD CM FRCS Boston MA Kingdom of Saudi Arabia
Associate Clinical Professor of USA
Ophthalmology Sabah Al-Jastaneiah MD
Massachusetts Eye and Ear Infirmary Levent Akduman MD Consultant Ophthalmologist
Harvard Medical School Assistant Professor of Ophthalmology Anterior Segment and Refractive Surgery
Clinical Senior Scientist Department of Ophthalmology Division
Schepens Eye Research Institute St Louis University School of Medicine King Khaled Eye Specialist Hospital
Boston MA St Louis MO Riyadh
USA USA Kingdom of Saudi Arabia
David H Abramson MD Marissa L Albano MD Calliope E Allen MD
Chief c/o Robert P Murphy Fellow
Ophthalmic Oncology Service The Retina Group of Washington Eye Plastics, Orbital and Cosmetic Surgery
Department of Surgery Fairfax VA Massachusetts Eye & Ear Infirmary
Memorial Sloane Kettering Cancer Center USA Boston MA
New York NY USA
USA Daniel M. Albert MD MS
Chair Emeritus, F. A. Davis Professor and David Allen BSc FRCS FRCOphth
Martin A Acquadro MD Lorenz F. Zimmerman Professor Consultant Ophthamologist
Perioperative Medical Doctor Department of Ophthalmology and Visual Sunderland Eye Infirmary
Director Sciences Sunderland
Department of Anesthesiology and Pain Retina Research Foundation Emmett A. United Kingdom
Caritas Carney Hospital Humble Distinguished Director
Dorchester MA Alice R. McPherson, MD, Eye Research Robert C Allen MD (deceased)
USA Institute Formerly Professor of Ophthalmology and
University of Wisconsin Pharmacology
Anthony P Adamis MD Madison WI Formerly Chairman, Department of
Chief Scientific Officer USA Ophthalmology
Executive Vice President, Research & Virginia Commonwealth University
Development Terry J Alexandrou MD Richmond VA
(OSI) Eyetech Pharmaceuticals Chief Resident USA
New York NY Department of Ophthalmology and Visual
USA Science Albert Alm MD PhD
University of Chicago Professor
Wesley H Adams MD Chicago IL Department of Neuroscience, Ophthalmology
Ophthalmology Resident USA University Hospital
Department of Ophthalmology Uppsala
Wake Forest University Eye Center Eduardo C Alfonso MD Sweden
Winston-Salem NC Professor, Edward W D Norton Chair in
USA Ophthalmology Samar Al-Swailem MD
Medical Director Consultant Ophthalmologist
Natalie A Afshari MD Ocular Microbiology Laboratory Anterior Segment Division
Associate Professor of Ophthalmology Bascom Palmer Eye Institute King Khaled Eye Specialist Hospital
Department of Ophthalmology University of Miami Riyadh
Duke University Eye Center Miami FL Kingdom of Saudi Arabia
Durham NC USA
USA Abigail K Alt BA
Jorge L Alió MD PhD c/o Thaddeus P Dryja MD
Everett Ai MD Professor and Chairman of Ophthalmology, Massachusetts Eye and Ear Infirmary
Director Miguel Hernandez University Harvard Medical School
Retina Unit Medical Director, VISSUM Boston MA
California Pacific Medical Center Instituto Oftalmológico de Alicante USA
San Francisco CA Alicante
USA Michael M Altaweel MD FRCS(C)
Spain Assistant Professor & Co-Director, Fundus
Lloyd M Aiello MD Hassan Alizadeh PhD Photograph Reading Center
Clinical Professor of Medicine Assistant Professor of Ophthalmology Department of Ophthalmology and Visual
Joslin Diabetes Center – Beetham Eye Department of Ophthalmology Science
Institute University of Texas Southwestern Medical University of Wisconsin
Harvard Medical School Center Madison WI
Boston MA Dallas TX USA xxiii
USA USA
List of Contributors

Russell Anderson BA Isabelle Audo MD PhD George B Bartley MD


Medical Writer Ophthalmologist Professor of Ophthalmology
Dry Eye Department Laboratory of Cellular Physiopathology and Mayo Medical School
Ophthalmic Research Associates Retinal Molecules Chief Executive Officer
North Andover MA Faculty of Medicine Mayo Clinic
USA INSERM Jacksonville FL
Université Pierre et Marie Curie USA
Christopher M Andreoli MD Hôpital St Antoine
Ophthalmologist Paris Jason J S Barton MD PhD FRCPC
Department of Ophthalmology France Director of Neuro-Ophthalmology
Massachusetts Eye and Ear Infirmary Professor and Canada Research Chair
Harvard Medical School Gerd U Auffarth Priv-Doz Dr med Neuro-Ophthalmology
Boston MA Research Group Leader VGH Eye Care Center
USA Heidelberg IOL & Refractive Surgery Vancouver BC
Research Group Canada
Sofia Androudi MD Department of Ophthalmology
First Department of Ophthalmology University of Heidelberg Irmgard Behlau MD
Aristotle University of Thessaloniki Heidelberg Department of Ophthalmology
Thessaloniki Germany Massachusetts Eye and Ear Infirmary
Greece Instructor In Medicine, Harvard Medical
Robin K Avery MD School
Leonard P K Ang MD MMed(Ophth) FRCS(Ed) Section Head, Transplant Infectious Disease Boston MA
MRCOphth Department of Infectious Diseases USA
Consultant Cleveland Clinic Foundation
Department of Cataract and Comprehensive Cleveland OH Jose I Belda MD PhD EBO
Ophthalmology USA Chairman
Singapore National Eye Centre Department of Ophthalmology
Singapore Dimitri T Azar MD Hospital de Torrevieja
B A Field Chair of Ophthalmologic Research Alicante
Fahd Anzaar MD Professor and Head, Department of Spain
Research Coordinator Ophthalmology and Visual Sciences
Massachusetts Eye Research and Surgery University of Illinois Eye and Ear Infirmary Jeffrey L Bennett MD PhD
Institute Chicago IL Associate Professor of Neurology &
Cambridge MA USA Ophthalmology
USA Department of Neurology
Ann S Baker MD (deceased) University of Colorado Health Sciences
David J Apple MD Formerly Director of the Infectious Disease Center
Professor of Ophthalmology and Pathology Service Denver CO
Director of Research Pawek-Vallotton Massachusetts Eye and Ear Infirmary USA
University of South Carolina Formerly Associate Professor of
Charleston SC Ophthalmology Timothy J Bennett CRA FOPS
USA Harvard Medical School Ophthalmic Photographer
Boston MA Department of Ophthalmology
Claudia A Arrigg MD MEd Penn State Milton S Hershey Medical Center
Senior Surgeon USA
Hershey PA
Lawrence General Hospital Mark Balles MD USA
Lawrence MA Retina Center of Maine
USA South Portland ME Gregg J Berdy MD FACS
USA Assistant Professor of Clinical Ophthalmology
Pablo Artal PhD & Visual Science
Professor of Optics Scott D Barnes MD Department of Ophthalmology and Visual
Centro de Investigacion en Optica y Fellow, Cornea Service, Massachusetts Eye Science
Nanofisica (CiOyN) and Ear Infirmary and Harvard Medical Washington University School of Medicine
Universidad de Murcia School St Louis MO
Murcia Chief, Ophthalmology and Refractive Surgery USA
Spain Department of Ophthalmology
Womack Army Medical Center Carlo Roberto Bernardino MD FACS
Penny Asbell MD Associate Professor of Ophthalmology
Professor of Ophthalmology Fort Bragg NC
USA Yale University School of Medicine
Department of Ophthalmology New Haven CT
Mount Sinai Medical Center Donald M Barnett MD USA
New York NY Assistant Clinical Professor of Medicine
USA Joslin Diabetes Center Vitaliano Bernardino MD
Beetham Eye Institute Ophthalmologist
George K Asdourian MD Private Practice
Chief, Division of Ophthalmology Harvard Medical School
Boston MA Langhorne PA
University of Massachusetts Memorial USA
Medical Center USA
Worcester MA Neal P Barney MD Eliot L Berson MD
USA Associate Professor of Ophthalmology Director, Electroretinography Service
Department of Ophthalmology and Visual Massachusetts Ear and Eye Infirmary
Neal Atebara MD William F Chatlos Professor of
Ophthalmologist Sciences
University of Wisconsin School of Medicine Ophthalmology
Retina Center of Hawaii Harvard Medical School
Honolulu HI Madison WI
USA Boston MA
USA USA
Pelin Atmaca-Sonmez Fina C Barouch MD
Research Fellow Assistant Professor of Ophthalmology
Department of Ophthalmology Eye Institute
University of Michigan Lahey Clinic Medical Center
Peabody MA
xxiv Ann Arbor MI
USA
USA
List of Contributors

Amitabh Bharadwaj MD Luigi Borrillo MD Alfred Brini MD


Ophthalmologist Retina-Vitreous Associates Inc Emeritus Professor of Ophthalmology
Department of Ophthalmology El Camino Hospital Louis Pasteur University
Wills Eye Hospital Mountain View, CA Strasbourg
Philadelphia PA USA France
USA
Gary E Borodic MD Donald L Budenz MD MPH
Robert Bhisitkul MD PhD Ophthalmologist Associate Professor
Assistant Professor of Clinical Ophthalmology Department of Ophthalmology Epidemiology and Public Health
Department of Ophthalmology Massachusetts Eye and Ear Infirmary Bascom Palmer Eye Institute
UCSF Beckman Vision Center Harvard Medical School Miami FL
San Francisco CA Boston MA USA
USA USA
Angela N Buffenn MD MPH
Ravinder D Bhui BApSc in Elec Eng S Arthur Boruchoff MD Assistant Professor of Clinical Ophthalmology
Senior Medical Student Professor (Retired) Childrens Hospital Los Angeles
Schulich School of Medicine and Dentistry Department of Ophthalmology Department of Ophthalmology
The University of Western Ontario Boston University School of Medicine Los Angeles CA
London ON Boston MA USA
Canada USA
Scott E Burk MD PhD
Jurij Bilyk MD Swaraj Bose MD Ophthalmologist
Attending Surgeon Associate Professor Department of Ophthalmology
Oculoplastic and Orbital Surgery Service Department of Ophthalmology Cincinnati Eye Institute
Wills Eye Institute University of California, Irvine Cincinnati OH
Philadelphia PA Irvine CA USA
USA USA
Salim Butrus MD
Valérie Biousse MD Michael E Boulton PhD Associate Clinical Professor
Associate Professor of Ophthalmology and Director of AMD Center Department of Ophthalmology
Neurology Department of Ophthalmology and Visual George Washington University
Emory Eye Center Sciences Washington DC
Emory University School of Medicine University of Texas Medical Branch USA
Atlanta GA Galveston TX
USA USA David Callanan MD
Vitreoretinal Specialist
Alan C Bird MD FRCS FRCOphth R W Bowman MD Texas Retina Associates
Professor Professor Arlington TX
Department of Clinical Ophthalmology Department of Ophthalmology USA
Moorfields Eye Hospital University of Texas Southwestern Medical
London Center J Douglas Cameron MD
United Kingdom Dallas TX Professor of Ophthalmology
USA Clinical Ophthalmology
Norman Paul Blair MD Scheie Eye Institute
Professor of Ophthalmology, Director of Elizabeth A Bradley MD University of Pennsylvania
Vitreoretinal Service Assistant Professor of Ophthalmology Philadelphia PA
Department of Ophthalmology and Visual Department of Ophthalmology USA
Sciences Mayo Clinic
University of Illinois Rochester MN Louis B Cantor MD
Chicago IL USA Professor of Ophthalmology
USA Department of Ophthalmology
Periklis D Brazitikos MD Indiana University School of Medicine
Barbara A Blodi MD Associate Professor of Ophthalmology Indianapolis IN
Associate Professor, Specialist in Retinal Department of Ophthamology USA
Disease Aristotle University of Thessaloniki
Department of Ophthalmology & Visual Thessaloniki William A Cantore MD
Sciences Greece Associate Professor of Ophthalmology and
University of Wisconsin-Madison Neurology
Madison WI Robert Breeze MD Department of Ophthalmology
USA Professor and Vice Chair Penn State University College of Medicine
Deparment of Neurosurgery Hershey PA
Mark S Blumenkranz MD University of Colorado Health Sciences USA
Professor and Chairman Center
Department of Ophthalmology Aurora CO Jorge Cantu-Dibildox MD
Stanford University School of Medicine USA Centro de Oftalmologia San Jose, S C
Stanford CA Fundación de Ojos Vidaurri, A C
USA Neil M Bressler MD Monterrey NL
Professor of Ophthalmology Mexico
H Culver Boldt MD The Wilmer Eye Institute
Professor of Ophthalmology The Johns Hopkins University School of Victoria Casas MD
Department of Ophthalmology Medicine Research Fellow
University of Iowa Baltimore MD Ocular Surface Research & Education
Iowa City IA USA Foundation
USA Miami FL
Susan B Bressler MD USA
Mark S Borchert MD Professor of Ophthalmology
Associate Professor of Ophthalmology Department of Ophthalmology Miriam Casper MD
Department of Ophthalmology John Hopkins Hospital c/o David J Apple MD
Children’s Hospital Los Angeles Baltimore MD University of South Carolina
Los Angeles CA USA Charleston SC
USA USA
xxv
List of Contributors

Robin J Casten PhD Joe Chen MD Antonio P Ciardella MD


Assistant Professor c/o Keith L Lane MD Chief, Department of Ophthalmology
Department of Psychiatry and Human ORA Clinical Research and Development Denver Health Medical Center
Behaviour North Andover MA Denver CO
Thomas Jefferson University USA USA
Philadelphia PA
USA Julie A Chen MD Mortimer Civan MD
c/o Joan M O’Brien MD Professor of Physiology
Yara P Catoira MD Division of Ophthalmology Department of Physiology
Assistant Professor of Clinical Ophthalmology University of California San Francisco University of Pennsylvania Health System
Department of Ophthalmology Medical Center Philadelphia PA
Indiana University School of Medicine San Francisco CA USA
Indianapolis IN USA
USA Liane Clamen MD
Teresa C Chen MD Harvard Medical School
Jerry Cavallerano OD PhD Assistant Professor of Medicine Boston MA
Assistant to the Director Glaucoma Service USA
Joslin Diabetes Center Massachusetts Eye and Ear Infirmary
Beetham Eye Institute Assistant Professor of Medicine, Harvard John I Clark PhD
Boston MA Medical School Professor, Biological Structure
USA Boston MA School of Medicine
USA University of Washington
Samantha J Chai MD Seattle WA
Medical Resident Zhou Chen PhD USA
Department of Ophthalmology Senior Pharmacologist and Toxicologist
Cullen Eye Institute Center for Drug Evaluation and Research Glenn Cockerham MD
Baylor College of Medicine Food & Drug Administration Clinical Associate Professor
Houston TX Silver Spring MD Department of Ophthalmology
USA USA Stanford University
Stanford CA
Maria R Chalita MD PhD Patricia Chévez-Barrios MD USA
Director of Cornea and Refractive Surgery Clinical Assistant Professor
Department of Ophthalmology Departments of Ophthalmology & Pathology Andre Cohen MD
Federal University of Brazil Baylor College of Medicine and the Texas Ophthalmologist
Sao Paulo Children’s Cancer Center Marietta Eye Consultants
Brazil Houston TX Marietta GA
USA USA
Sherman M Chamberlain MD FACP FACG
Assistant Professor of Medicine Emily Y Chew MD Elisabeth J Cohen MD
Gastroenterology and Hepatology Medical Officer, Division of Biometry and Director Cornea Service, Attending Surgeon,
Medical College of Georgia Epidemiology Wills Eye Hospital
Augusta GA National Eye Institute Professor, Department of Ophthalmology
USA National Institutes of Health Jefferson Medical College of Thomas
Bethseda MD Jefferson University
Audrey S Chan MD USA Philadelphia PA
Cornea and Refractive Surgery Fellow USA
Massachusetts Eye and Ear Infirmary Mark Chiang MBBS
Harvard Medical School Birmingham and Midland Eye Centre Kathryn A Colby MD PhD
Boston MA Birmingham Director, Joint Clinical Research Center
USA United Kingdom Attending Surgeon, Cornea Service
Massachusetts Eye and Ear Infirmary
Chi-Chao Chan MD James Chodosh MD Assistant Professor of Ophthalmology,
Head, Immunopathology Section Professor of Ophthalmology Harvard Medical School
National Eye Institute Department of Ophthalmology Boston MA
National Institutes of Health University of Oklahoma Health Sciences USA
Bethesda MD Center
USA Oklahoma City OK Anne L Coleman MD PhD
USA Professor of Ophthalmology and
Paul Chan MD Epidemiology
Assistant Professor of Ophthalmology Eva-Marie Chong MBBS Departments of Ophthalmology and
New York Presbyterian Physician Epidemiology
Wiell Medical College of Cornell University Department of Ophthalmology Jules Stein Eye Institute
New York NY Arizona Medical Center Los Angeles CA
USA Peoria AZ USA
USA
Matthew J Chapin MD Hanna R Coleman MD
Ophthalmic Research Associates, Inc Denise Chun BS Staff Clinician
North Andover MA Doctoral Candidate in Genetics, Harvard Department of Ophthalmology
USA Medical School New York Presbyterian Hospital
Department of Molecular Biology Columbia University Medical Center
Karen L Chapman MD Massachusetts General Hospital New York NY
University of South Florida Boston MA USA
Sarasota Memorial Hospital USA
Sarasota FL Joseph Colin MD
USA Leo T Chylack Jr MD Professor of Ophthalmology
Director of Research Department of Ophthalmology
Eric Chen MD Center for Ophthalmic Research C H U Morvan
Retina Research Center Brigham & Women’s Hospital Brest
Austin TX Boston MA France
USA USA

xxvi
List of Contributors

J Michael Collier PhD Donald J D’Amico MD Adam G de la Garza MD


Instructor, Harvard Medical School Professor and Chairman Chief Resident, Wake Forest University Eye
Senior Medical Physicist Ophthalmologist-in-Chief Center
Department of Radiation Oncology Department of Ophthalmology Wake Forest University School of Medicine
Massachusetts General Hospital Weill Cornell Medical College Winston-Salem NC
Boston MA New York NY USA
USA USA
Margaret M DeAngelis PhD
Grant M Comer MD Reza Dana MD MSc MPH Instructor of Ophthalmology
Assistant Professor Director, Cornea and Refractive Surgery Massachusetts Eye & Ear Infirmary
Kellogg Eye Center Services Boston MA
University of Michigan Massachusetts Eye and Ear Infirmary USA
Ann Arbor MI Professor, Harvard Medical School
USA Senior Scientist & W Clement Stone Scholar Sheri L DeMartelaere MD
Schepens Eye Research Institute Director of Orbital and Ocular Trauma
M Ronan Conlon MD FRCSC Boston MA Ophthalmology Service
Eye Physician and Surgeon USA Brooke Army Medical Center
Midwest Eye Care Institute Fort Sam Houston TX
Saskatoon SK Aude Danan-Husson MD USA
Canada Service d’ophtalmologie
Centre Hospitalier National d’Ophtalmologie Joseph L Demer MD PhD
Kim E Cooper MD des Quinze-vingts Leonard Apt Professor of Ophthalmology
Associate Professor Paris Departments of Ophthalmology and
Southwest College of Naturopathic Medicine France Neurology
Tempe AR Jules Stein Eye Institute
USA Helen B Danesh-Meyer MBChB MD Los Angeles CA
FRANZCO USA
James J Corbett MD Associate Professor of Ophthalmology
McCarty Professor and Chairman for Department of Ophthalmology Avninder Dhaliwal MD
Neurology University of Auckland Medical School University of Minnesota Medical School
Department of Neurology Auckland Minneapolis MN
University of Mississippi Medical Center New Zealand USA
Jackson MS
USA Ronald P Danis MD J Paul Dieckert MD
Professor of Ophthalmology and Visual Center Director, Division of Ophthalmology
Miguel C Coma MD FEBOphth Science Scott and White Memorial Hospital
Massachusetts Eye Research and Surgery Director, Fundus Photograph Reading Center Temple TX
Institute Department of Ophthalmology and Visual USA
Cambridge MA USA Science
Department of Ophthalmology Diana V Do MD
University of Wisconsin Fellow in Advanced Speciality Training in
Hospital de León, León, Spain Madison WI Medical and Surgical Diseases of the Retina
Marshall N Cyrlin MD USA Assistant Professor of Ophthalmology
Clinical Professor of Biomedical Sciences Jason K Darlington MD The Johns Hopkins University School of
Eye Research Institute Department of Ophthalmology Medicine
Oakland University University of California at Davis The Wilmer Eye Institute
Rochester MN Sacramento CA Baltimore MD
USA USA USA
Linda R Dagi MD Stefanie L Davidson MD Marshall G Doane PhD
Director of Adult Strabismus, Instructor in Assistant Clinical Professor, University of Emeritus Senior Scientist
Ophthalmology Pennsylvania Department of Ophthalmology
Department of Ophthalmology Division of Ophthalmology Schepens Eye Research Institute
Childrens Hospital Childrens Hospital of Philadelphia Harvard Medical School
Boston MA Philadelphia PA Boston MA
USA USA USA
Matthew A Dahlgren MD Janet L Davis MD MA Christopher Dodds MBBS MRCGP FRCA
Fellow, Cornea and Anterior Segement, Associate Professor of Ophthalmology Professor of Anaesthesia
Department of Ophthalmology Division of Ophthalmology Academic Anaesthetic Department
University of Minnesota Medical School University of Miami James Cook University Hospital
Minneapolis MN Miami FL Middlesbrough
USA USA United Kingdom
Timothy J Daley BS Elizabeth A Davis MD FACS Claes H Dohlman MD PhD
University of Wisconsin Hospital and Clinics Adjunct Clinical Assistant Professor, Professor of Ophthalmology, Harvard Medical
Madison WI University of Minnesota School, Chief Emeritus
USA Director, Minnesota Eye, Laser and Surgery Cornea Service
Center Massachusetts Eye and Ear Infirmary
Andrea P Da Mata MD Harvard Medical School
Ocular Immunology and Uveitis Foundation Bloomington MN
USA Boston MA
Massachusetts Eye Research and Surgery USA
Institute Jose J de la Cruz MD
Cambridge MA Cornea Fellow, Department of Guy Donati MD
USA Ophthalmology and Visual Science Chare D’Ensign
University of Illinois at Chicago Department of Pathology
Bertil Damato MD PhD FRCOphth University of Geneva
Professor of Ophthalmology Chicago IL
USA Geneva
Ocular Oncology Service Switzerland
Royal Liverpool University Hospital
Liverpool
United Kingdom xxvii
List of Contributors

Eric D Donnenfeld MD FACS Chiara M Eandi MD Frederick L Ferris III MD


Co-director, Cornea Division The LuEsther T Mertz Retinal Research Director, Division of Epidemiology and
Ophthalmic Consultants of Long Island Fellow Clinical Research
New York NY Manhattan Eye, Ear and Throat Hospital National Eye Institute
USA New York NY National Institutes of Health
USA Bethesda MD
Arlene Drack MD USA
Chief of Ophthalmology, Children’s Hospital Deepak P Edward MD
Associate Professor Professor and Chairman Howard F Fine MD MHSc
Department of Ophthalmology Department of Ophthalmology - Suma Health Vitreoretinal surgical fellow
University of Colorado Health Sciences Systems Vitreous Retina Macula New York
Center Northeastern Ohio University School of New York NY
Aurora CO Medicine USA
USA Akron OH
USA Donald C Fletcher MD
Thaddeus P Dryja MD Medical Director
Director, David C Cogan Eye Pathology Robert A Egan MD Frank Stein & Paul May Center for Low Vision
Laboratory Assistant Professor of Ophthalmology and Rehabilitation
Massachusetts Eye and Ear Infirmary Neurology California Pacific Medical Center
Harvard Medical School Departments of Ophthalmology and Scientist, Smith-Kettlewell Eye Research
Boston MA Neurology Institute
USA Casey Eye Institute San Francisco CA
Portland OR USA
David Dueker MD USA
Professor of Ophthalmology Paul Flikier MD
The Eye Institute David A Eichenbaum MD Farmacia Alvarez, Heredia
Medical College of Wisconsin Associate Director, Centro Medico de la Vision
Milwaukee WI Retina-Vitrous Associates of Florida San Jose
USA St Petersburg FL Costa Rica
USA
Jay S Duker MD Richard P Floyd MD
Director New England Eye Center Susan E Eklund BA Clinical Instructor
Chairman and Professor of Ophthalmology Assistant, Department of Ophthalmology Department of Ophthalmology
Tufts University School of Medicine Children’s Hospital Harvard Medical School
Tufts New England Medical Center Boston MA Boston MA
Boston MA USA USA
USA
Elizabeth C Engle MD Harry W Flynn Jr, MD
Jennifer A Dunbar MD Associate Professor of Neurology Professor, The J Donald M Gass
Director of Pediatric Ophthalmology Harvard Medical School Distinguished Chair of Ophthalmology
Department of Ophthalmology Department of Neurology, Program in Bascom Palmer Eye Institute
Loma Linda University Genomics, Children’s Hospital The University of Miami Miller School of
Loma Linda CA Boston MA Medicine
USA USA Miami FL
USA
James P Dunn MD Kristine Erickson OD PhD
Associate Professor of Ophthalmology Senior Director Clinical Affairs Donald S Fong MD MPH
The Wilmer Eye Institute Unigene Corporation Director, Cinical Trials Research
John Hopkins School of Medicine Boonton NJ Kaiser Permanente Southern California
Baltimore MD USA Pasadena CA
USA USA
Bita Esmaeli MD FACS
William J Dupps Jr, MD PhD Associate Professor of Ophthalmology; Ramon L Font MD
Associate Staff, Ophthalmology and Director of Ophthalmic Plastic and Professor of Pathology and Ophthalmology
Biomedical Engineering Reconstructive and Orbital Surgery The Sarah Campbell Blaffer Chair of
Cole Eye Institute Fellowship Ophthalmology
Cleveland Clinic and Lerner Research Department of Medicine The Neurosensory Center
Institute The University of Texas Houston TX
Cleveland OH Houston TX USA
USA USA
Brian J R Forbes MD PhD
Marlene L Durand MD Aaron Fay MD Assistant Professor of Ophthalmology
Director of Infectious Diseases, Interim Director, Ophthalmic Plastic Surgery Department of Ophthalmology
Massachusetts Eye and Ear Infirmary Massachusetts Eye and Ear Infirmary The Childrens Hospital of Philadelphia
Assistant Professor of Medicine, Harvard Assistant Clinical Professor of Wallingford PA
Medical School; Ophthalmology, Department of USA
Infectious Diseases Unit Ophthalmology, Harvard Medical School
Massachusetts General Hospital Boston MA Rod Foroozan MD
Boston MA USA Assistant Professor of Ophthalmology
USA Department of Ophthalmology
Leonard Feiner MD PhD Baylor College of Medicine
Jonathan J Dutton MD PhD Ophthalmology Department Houston TX
Professor and Vice Chair Montefiore Medical Center USA
Department of Ophthalmology Lawrence NY
University of North Carolina USA Bradley S Foster MD
Chapel Hill NC Assistant Clinical Professor of Ophthalmology
USA Sharon Fekrat MD New England Retina Consultants
Assistant Professor West Springfield MA
Department of Ophthalmology USA
Vitreoretinal Surgery
Duke Eye Center
xxviii Durham NC
USA
List of Contributors

C Stephen Foster MD FACS David Friedman MD James A Garrity MD


Founder and President Assistant Professor Professor of Ophthalmology
The Massachusetts Eye Research Institute Ophthalmology Department Department of Ophthalmology
Clinical Professor of Ophthalmology Wilmer Eye Institute Mayo Clinic
Harvard Medical School John Hopkins University School of Medicine Rochester MN
Cambridge MA Baltimore MD USA
USA USA
Damien Gatinel MD
Jill A Foster MD Deborah I Friedman MD FAAN Assistant Professor
Assistant Clinical Professor Associate Professor of Ophthalmology and Ophthalmology Department
The William H Havener Eye Institute Neurology Fondation Ophtalmologique A de Rothschild
The Ohio State University Departments of Ophthalmology and Paris
Columbus OH Neurology France
USA University of Rochester School of Medicine
and Dentistry Steven J Gedde MD
Gary N Foulks MD FACS Rochester NY Professor of Ophthalmology and Residency
Arthur & Virginia Keeney Professor of USA Program Director
Ophthalmology Department of Ophthalmology
Department of Ophthalmology Ephraim Friedman MD Bascom Palmer Eye Institute
University of Louisville School of Medicine Former Chief, Department of Ophthalmology Miami FL
Louisville KY Massachusetts Eye and Ear Infirmary USA
USA Harvard Medical School Retina Service
Boston MA Craig E Geist MD FACS
Tamara R Fountain MD USA Chairman, Department of Ophthalmology
Associate Professor Associate Professor, Ophthalmology,
Department of Ophthalmology Arthur D Fu MD Neurology, Neurological Surgery
Rush University in Chicago Ophthalmologist Director, Oculoplastics, Orbit, Lacrimal
Northbrook IL Pacific Vision Foundation Director, Neuro-Ophthalmology
USA California Pacific Medical Center The George Washington University
San Francisco CA Washington DC
Gregory M Fox MD USA USA
Clinical Instructor of Ophthalmology
Department of Ophthalmology Anne B Fulton MD Steve Gerber MD
Allegheny University Associate Professor of Ophthalmology and Chairman
Wilmington DE Senior Associate in Ophthalmology Department of Ophthalmology
USA Department of Ophthalmology Memorial Hospital
Children’s Hospital South Bend IN
Thomas F Freddo OD PhD FAAO Boston MA USA
Professor and Director USA
School of Optometry Ramon C Ghanem MD
University of Waterloo Ahmed Galal MD PhD Sadalla Amin Ghanem
Waterloo ON Department of Refractive Surgery Hospital de Olhos
Canada Vissum/Instituto Oftalmologico de Alicante Batista
Alicante Joinville - SC
Sharon F Freedman MD Spain Brazil
Associate Professor of Ophthalmology
Associate Professor of Pediatrics Steven Galetta MD Jon P Gieser MD
Department of Pediatric Ophthalmology & Director, Neuro-Ophthalmology Services Wheaton Eye Clinic
Strabismus Hospital of the University of Pennsylvania Wheaton IL
Duke University Eye Center Philadelphia PA USA
Durham NC USA
USA Michael S Gilmore PhD
Mark Gallardo MD Charles L Schepens Professor of
K Bailey Freund MD Resident Physician Ophthalmology
Retina Specialist Office of Border Health President and Ankeny Director of Research
Vitreous-Retina-Macula Consultants of Texas Tech University Health Sciences Center The Schepens Eye Research Institute
New York El Paso TX Harvard Medical School
New York NY USA Boston MA
USA USA
Brenda Gallie MD FRCS(C)
Thomas R Friberg MD Professor of Ophthalmology Howard V Gimbel MD MPH FRCSC FACS
Professor of Ophthalmology, Professor of Departments of Medical Biophysics and Chair and Professor of The Department of
Bioengineering Molecular and Medical Genetics Ophthalmology
Director of the Retina Service University of Toronto Refractive Surgery, Department of
Departments of Ophthalmology and Head of Cancer Informatics Ophthalmology
Bioengineering University Health Network Loma Linda University
UPMC Eye Center Ontario Cancer Institute Loma Linda CA
Pittsburgh PA Princess Margaret Hospital USA
USA Toronto ON
Canada Ilene K Gipson PhD
Alan H Friedman MD Senior Scientist and Professor of
Department of Ophthalmology Alec Garner MD Ophthalmology
Mount Sinai School of Medicine Head of Department Department of Ophthalmology
New York NY Department of Pathology Schepens Eye Research Institute
USA Institute of Ophthalmology Boston MA
London USA
United Kingdom

xxix
List of Contributors

Tyrone Glover MD David B Granet MD FACS FAAP FAAO Vamsi K Gullapalli MD PhD
Clinical Professor, Ophthalmology Anne F Ratner Professor of Ophthalmology & Resident
Oculoplastic Surgery Pediatrics Department of Ophthalmology and Visual
Kaiser Permanente Director, Pediatric Ophthalmology & Adult Science
Sacramento CA Re-Alignment Services Institute of Ophthalmology and Visual
USA Anne F & Abraham Ratner Children’s Eye Center Science
Shiley Eye Center University of Medicine and Dentistry of
Robert A Goldberg MD FACS University of California, San Diego New Jersey
Associate Professor of Ophthalmology La Jolla CA Newark NJ
Chief, Division of Orbital and Ophthalmic USA USA
Plastic Surgery
Jules Stein Eye Institute Michael J Greaney Padma Gulur MD
Los Angeles CA Senior Clinical Lecturer, Department of Instructor in Anaesthesia, Harvard Medical
USA Ophthalmology, University of Bristol School
Senior Consultant Pain Center Department of Anesthesia and
Mordechai Goldenfeld MD
Bristol Eye Hospital Critical Care
Senior Attending Ophthalmologist
Bristol Massachusetts General Hospital
The Sam Rothberg Glaucoma Centre
United Kingdom Boston MA
Goldschleger Eye Institute
USA
Sheba Medical Center Daniel G Green PhD
Tel-Hashomer Professor Emeritus, Ophthalmology and Jonathan Gunther MD
Israel Visual Sciences Department of Ophthalmology and Visual
Scott M Goldstein MD Professor, Biomedical Engineering Sciences
Clinical Associate The University of Michigan Kellogg Eye University of Wisconsin Medical School
Childrens Hospital of Philadelphia Center Madison WI
Tricounty Eye Physicians & Surgeons Ann Arbor MI USA
Southampton PA USA
Manish Gupta DNB FRCS(Glasg), MRCS(Ed)
USA Franz Grehn Dr h.c. NHS Greater Glasgow and Clyde
Cintia F Gomi MD Professor of Ophthalmology Stobhill and Gartnevel Hospital
Hamilton Glaucoma Center Chairman, Department of Ophthalmology Glasgow
University of California, San Diego, University of Würzburg United Kingdom
La Jolla CA Würzburg
Germany Mayank Gupta
USA
c/o Deepak P Edward MD
Haiyan Gong MD MS PhD Jack V Greiner DO PhD Northeastern Ohio University School of
Research Assistant Professor Instructor of Ophthalmology Medicine
Department of Ophthalmology Schepens Eye Research Institute Akron OH
Boston University School of Medicine Harvard Medical School USA
Boston MA Boston MA
USA USA David R Guyer MD
Clinical Professor
John A Gonzales MD Craig M Greven MD FACS Department of Ophthalmology
Physician Director, Professor and Chairman NYU Medical Center
Immunopathology Section Department of Ophthalmology New York NY
Laboratory of Immunology Wake Forest University Eye Center USA
National Eye Institute Wake Forest University School of Medicine
National Institutes of Health Winston-Salem NC Darin R Haivala MD
Besthesda MD USA Clinical Assistant Professor
USA Department of Ophthalmology
Gregory J Griepentrog MD University of Oklahoma
John Goosey MD Chief Resident Associate Dean A McGee Eye Institute
Director Mayo Clinic Oklahoma City OK
Houston Eye Associates Rochester MN USA
Houston TX USA
USA Julia A Haller MD
Carl Groenewald MD Robert Bond Welch Professor of
Justin L Gottlieb MD
Consultant Vitreoretinal Surgeon Ophthamology
Associate Professor
St Paul’s Eye Unit Wilmer Ophthalmological Institute
Department of Ophthalmology and Visual
Royal Liverpool University Hospital Johns Hopkins Medical Institutions
Sciences
Liverpool Baltimore MD
University of Wisconsin
United Kingdom USA
Madison WI
USA Cynthia L Grosskreutz MD PhD G M Halmagyi MD BSc FACS DCH
Joshua Gould DO Co-Director, Glaucoma Service Professor of Neurology
Physician Massachusetts Eye and Ear Infirmary Department of Neurology
Eye Care Center of New Jersey Associate Professor of Ophthalmology Royal Prince Albert Hospital
Bloomingfield NJ Harvard Medical School Sydney NSW
USA Boston MA Australia
USA
Evangelos S Gragoudas MD Lawrence S Halperin MD FACS
Director, Retina Service Lori Latowski Grover OD Physician
Massachusetts Eye and Ear Infirmary Assistant Professor of Ophthalmology Retina Vitreous Consultants of South Florida
Professor of Ophthalmology, Harvard Medical Department of Ophthalmology Fort Lauderdale FL
School Lions Vision Research and Rehabilitation USA
Boston MA Center
Baltimore MD Islam M Hamdi FRCS MD
USA Magrabi Center
USA
Jeddah
Kingdom of Saudi Arabia

xxx
List of Contributors

Steven R Hamilton MD Yasutaka Hayashida MD PhD Ahmed A Hidayat MD


Clinical Associate Professor of Research Fellow Chief, Ophthalmic Pathology
Ophthalmology and Neurology Ocular Surface Research and Education Armed Forces Institute of Pathology
Department of Ophthalmology and Neurology Foundation Rockville MD
University of Washington Miami FL USA
Neuro-Ophthalmic Associates Northwest USA
Seattle WA Eva Juliet Higginbotham MD
John R Heckenlively MD FRCOpath Professor of Ophthalmology and Chair
USA
Paul R Lichter Professor of Ophthalmic Department of Ophthalmology
Kristin M Hammersmith MD Genetics University of Maryland Medicine
Assistant Surgeon, Cornea Service, Wills Eye Professor of Ophthalmology and Visual Baltimore MD
Hospital Science USA
Instructor, Thomas Jefferson Medical College Kellogg Eye Centre
Wills Eye Institute University of Michigan Tatsuo Hirose MD
Thomas Jefferson University Ann Arbor MI Clinical Professor of Ophthalmology
Philadelphia PA USA Schepens Retina Associates
USA Boston MA
Thomas R Hedges III, MD USA
Dennis P Han MD Director, Neuro-Ophthalmology Service
Jack A and Elaine D Klieger Professor of Co-Director, Electrophysiology Service Allen C Ho MD
Ophthalmology, Vitreoretinal Section Head Director, Neuro-Ophthalmology Fellowship Professor of Ophthalmology
Department of Ophthalmology Program Retina Service
Medical College of Wisconsin New England Eye Center Thomas Jefferson University
Milwaukee WI Boston MA Philadelphia PA
USA USA USA

Ronald M Hansen PhD Alfred D Heggie MD ThucAnh T Ho MD


Instructor Professor Emeritus of Pediatrics Vitreoretinal Fellow
Department of Ophthalmology Departments of Pediatrics, Preventive Illinois Retina Associates
Children’s Hospital and Harvard Medical Medicine, and Obstetrics and Gynecology Rush University Medical Center
School Case Western Reserve University School of Chicago IL
Boston MA Medicine USA
USA Cleveland OH R Nick Hogan MD PhD
J William Harbour MD USA Associate Professor of Ophthalmology
Distinguished Professor of Ophthalmology, Department of Ophthalmology
Katrinka L Heher MD
Cell Biology, and Medicine (Molecular University of Texas South Western Medical
Director, Aesthetic Eyelid & Facial Surgery
Oncology) Center
Director, Oculoplastic & Orbital Surgery
Director, Ocular Oncology Service Dallas TX
Service
Department of Ophthalmology USA
Director, Ophthalmic Plastics and
Washington University School of Medicine Reconstructive Surgery Fellowship Program David E Holck MD
St Louis MO New England Eye Center Director, Oculoplastic, Reconstructive, Orbit,
USA Tufts University School of Medicine and Ocular Oncology Service
Seenu M Hariprasad MD Boston MA Department of Ophthalmology
Assistant Professor and Director of Clinical USA Wilford Hall Medical Center
Research Assistant Professor of Surgery, USUHS
Jeffrey S Heier MD Assistant Professor of Ophthalmology
Chief, Vitreoretinal Service Vitreoretinal Specialist
Department of Ophthalmology and Visual University of Texas
Ophthalmic Consultants of Boston San Antonio TX
Science Boston MA
University of Chicago USA
USA
Chicago IL Nancy M Holekamp MD
USA J Fielding Hejtmancik MD PhD Associate Professor of Clinical
Medical Officer Ophthalmology
Mona Harissi-Dagher MD Ophthalmic Genetics and Visual Function
Assistant in Ophthalmology Department of Ophthalmology and Visual
Branch Science
Massachusetts Eye and Ear Infirmary National Eye Institute
Harvard Medical School Washington University School of Medicine
National Institutes of Health Barnes Retina Institute
Boston MA Bethesda MD
USA St Louis MO
USA USA
Shirin E. Hassan PhD
Bonnie A Henderson MD FACS Peter G Hovland MD PhD
c/o David Friedman
Assistant Clinical Professor Physician
Assistant Professor
Department of Ophthalmology Colorado Retina Associates
Wilmer Eye Institute
Harvard Medical School Denver CO
John Hopkins University School of Medicine
Boston MA USA
Baltimore MD
USA
USA Thomas C Hsu MD
Mark P Hatton MD Peter S Hersh MD FACS Tufts University School of Medicine
Clinical Instructor, Harvard Medical School Professor of Ophthalmology New England Eye Center
Adjunct Clinical Scientist Director, Cornea and Laser Eye Institute - Boston MA
Schepens Eye Research Institute Hersh Vision Group USA
Ophthalmic Consultants of Boston Clinical Professor of Ophthalmology
Chief, Cornea and Refractive Surgery William C Hsu MD
Boston MA Assistant Professor of Medicine
USA University of Medicine and Dentistry New
Jersey Joslin Diabetes Center
Pamela Hawley MS Teaneck NJ Beetham Eye Institute
Genetic Counseling Center USA Harvard Medical School
Children’s Hospital Boston MA
Harvard USA
Boston MA xxxi
USA
List of Contributors

Andrew J W Huang MD MPH Fei Ji PhD Alon Kahana MD PhD


Director of Cornea and Refractive Surgery Research Associate Assistant Professor; Eye Plastics, Orbit and
Department of Ophthalmology Laboratory of Statistical Genetics Facial Cosmetic Surgery
University of Minnesota Rockefeller University Department of Ophthalmology and Visual
Minneapolis MN New York NY Sciences
USA USA Kellogg Eye Center
Ann Arbor MI
Mark S Hughes MD David L Johnson MD USA
Adjunct Assistant Clinical Scientist Clinical Instructor/Vitreoretinal Fellow
The Schepens Eye Research Institute Department of Ophthalmology and Visual Malik Y Kahook MD
Boston MA Sciences Assistant Professor and Director of Clinical
USA University Of Wisconsin Medical School Research
Jennifer Hui MD Madison WI Rocky Mountain Lions Eye Institute
Ophthalmology Resident USA University of Colorado at Denver Health
Department of Ophthalmology Sciences Center
Douglas H Johnson MD (deceased) Aurora CO
Bascom Palmer Eye Institute Formerly Professor of Ophthalmology
Miami FL USA
Department of Ophthalmology
USA Mayo Clinic Elliott Kanner MD PhD
David G Hunter MD PhD Rochester MN Assistant Professor of Ophthalmology
Associate Professor of Ophthalmology, USA Hamilton Eye Institute
Harvard Medical School University of Tennessee Health Science
Mark W Johnson MD Center
Ophthalmologist-in-Chief Professor
Richard Robb Chair in Ophthalmology Memphis TN
Kellogg Eye Center USA
Department of Ophthalmology University of Michigan
Children’s Hospital Boston Ann Arbor MI Kevin Kalwerisky MD
Boston MA USA Department of Otolaryngology, Head & Neck
USA Surgery
R Paul Johnson MD The New York Presbyterian Hospital
Laryssa A Huryn MD Associate Professor of Medicine
Bascom Palmer Eye Institute Weill Medical College of Cornell University
Infectious Diseases Unit New York NY
Miami FL Massachusetts General Hospital
USA USA
Charlestown MA
Deeba Husain MD USA Henry J Kaplan MD
Assistant Professor of Ophthalmology Professor and Chairman
Robert N Johnson MD Department of Ophthalmology and Visual
Retina Service - Dept of Ophthalmology Assistant Clinical Professor of Ophthalmology
Boston University School of Medicine Sciences
Department of Ophthalmology University of Louisville
Boston MA University of California
USA Louisville KY
West Coast Retina Medical Group USA
Robert A Hyndiuk MD San Francisco CA
The Eye Institute USA Ekaterini C Karatza MD
Medical College of Wisconsin Staff Ophthalmologist
Karen M Joos MD PhD Cincinnati Eye Institute
Milwaukee WI Associate Professor
USA Cincinnati OH
Department of Ophthalmology and Visual USA
Michael Ip MD Sciences
Associate Professor of Ophthalmology Vanderbilt University Randy Kardon MD PhD
Department of Ophthalmology and Visual Nashville TN Associate Professor of Ophthalmology
Sciences USA Director of Neuro-ophthalmology
Fundus Photograph Reading Center The University of Iowa Hospitals and Clinics
Nancy C Joyce PhD Iowa City IA
Madison WI Schepens Eye Research Institute
USA USA
Senior Scientist Associate Professor, Harvard
Brian J Jacobs MD Medical School James A Katowitz MD
Assistant Professor of Ophthalmology Boston MA Attending Surgeon
Rush University Medical Center USA Ophthalmology
Chicago IL Childrens Hospital of Philadelphia
J Michael Jumper MD Philadelphia PA
USA Assistant Clinical Professor of Ophthalmology USA
Frederick A Jakobiec MD DSc(Med) University of California
Former Henry Willard Williams Professor and Director, Retina Service William R Katowitz MD
Former Chief of Ophthalmology West Coast Retina Medical Group Department of Ophthalmology
Departments of Pathology and San Francisco CA University of Rochester School of Medicine
Ophthalmology USA and Dentistry
Harvard Medical School Rochester NY
Ula V. Jurkunas MD USA
Boston MA Instructor in Ophthalmology
USA Massachusetts Eye and Ear Infirmary Melanie Kazlas MD
Lee M Jampol MD Clinical Scientist Acting Director; Instructor
Louis Feinberg Professor and Chairman Schepens Eye Research Institute Pediatric Ophthalmology & Strabismus
Department of Ophthalmology Harvard Medical School Massachusetts Eye & Ear Infirmary
Northwestern University Medical School Boston MA Boston MA
Chicago IL USA USA
USA Kelly S Keefe CAPT MC USN
Harold G Jensen PhD Staff Ophthalmic Pathologist
Clinical Project Manager Comprehensive Ophthalmologist
Allergan, Inc Naval Medical Center
Irvine CA San Diego CA
xxxii USA USA
List of Contributors

Lara Kelley MD Jonathan W Kim MD Thomas Kohnen MD


Assistant Professor, Dermatology Physician Professor of Ophthalmology
Harvard Medical School Memorial Sloan-Kettering Cancer Center Deputy Chairman
Beth Israel Deaconess Medical Center New York NY Klinik fur Augenheilkunde
Boston MA USA Johann Wolfgang Goethe University
USA Frankfurt
Rosa Y Kim MD Germany
Charles J Kent MD Physician
Fellowship Training in Ocuplastics and Ocular Vitreoretinal Consultants Takeshi Kojima MD PhD
Pathology Houston TX Research Group for Environmental
Everett & Hurite Ophthalmology Associates USA Conservation Processing
Pittsburgh PA Department of Material Development
USA Stella K Kim MD Takasaki Radiation Chemistry Research
Assistant Professor of Ophthalmology Establishment
Kenneth R Kenyon MD FACS Section of Ophthalmology Japan Atomic Energy Research Institute
Associate Clinical Professor MD Anderson Cancer Center Takasaki-shi
Harvard Medical School; Houston TX Japan
Eye Health Vision Centers USA
North Dartmouth MA Tobias Koller MD
USA Tae-Im Kim MD PhD Refractive Surgeon
Department of Ophthalmology Institute of Ophthalmic and Refractive
Bilal F Khan MD Yonsei University Health System Surgery
Assistant in Ophthalmology Seoul Zurich
Massachusetts Eye and Ear Infirmary South Korea Switzerland
Harvard Medical School
Boston MA Christina M Klais MD David A Kostick MD
USA Retina Fellow Assistant Professor of Ophthalmology
LuEsther T Mertz Retinal Research Center Department of Ophthalmology
Jemshed A Khan MD Manhattan Eye, Ear and Throat Hospital Mayo Clinic College of Medicine
Clinical Professor of Ophthalmology New York NY Jacksonville FL
Kansas University USA USA
Kansas City MO
USA Stephen R Klapper MD FACS Joel A Kraut MD
Ophthalmologist Medical Director
Naheed W Khan PhD Klapper Eyelid & Facial Plastic Surgery Vision Rehabilitation Service
Electrophysiologist Carmel IN Massachusetts Eye and Ear Infirmary
Department of Ophthalmology and Visual USA Harvard Medical School
Sciences Boston MA
W K Kellogg Eye Center Barbara E K Klein MD MPH
USA
University of Michigan Professor of Medicine
Ann Arbor MI Department of Ophthalmology and Visual Chandrasekharan Krishnan MD
USA Sciences Assistant Professor of Ophthalmology
University of Wisconsin Medical School Tufts University School of Medicine
Peng Tee Khaw PhD FRCP FRCS FRCOphth Madison WI Glaucoma and Cataract Service
FIBiol FRCPath FMedSci USA New England Eye Center
Professor of Glaucoma and Ocular Healing Boston MA
and Consultant Ophthalmic Surgeon Guy Kleinmann MD
USA
Biomedical Research Centre (Ophthalmology) Adjunct Assistant Professor of
UCL Institute of Ophthalmology and Ophthalmology Ronald R Krueger MD MSE
Moorfields Eye Hospital Department of Ophthalmology Director of Refractive Surgery, Cleveland
London Herman Eye Center Clinic Foundation, Cleveland, OH, USA
United Kingdom Houston TX Saint Louis University Eye Institute
USA Saint Louis University School of Medicine
Femida Kherani MD FRCSC St Louis MO
Ophthalmic Cosmetic Surgeon Thomas Klink DrMed USA
Heights Laser Centre Scientific Assistant
Burnaby BC Department of Ophthalmology Joseph H Krug Jr, MD
Canada University of Würzburg Assistant Director of Glaucoma Consultation
Würzburg Service
Eva C Kim MD Germany Massachusetts Eye and Ear Infirmary
Fellow in Ocular Inflammation/Uveitis Harvard Medical School
The Proctor Foundation Dino D Klisovic MD Boston MA
University of California San Francisco Department of Ophthalmology USA
San Francisco CA Nationwide Children’s Hospital
USA Midwest Retina Inc Sara Krupsky MD
Columbus OH Goldschleger Eye Institute
Hee Joon Kim MD USA Sheba Medical Center
Resident Tel Hashomer
Department of Ophthalmology and Visual Stephen D Klyce Israel
Science Executive Editor
Department of Ophthalmology Rachel W Kuchtey MD PhD
University of Texas Health Science Center at
Louisiana State University Eye Institute Clinical Ophthalmologist, Glaucoma
Houston
New Orleans LA Vanderbilt University of Ophthalmology &
Houston TX
USA Visual Sciences
USA
Nashville TN
Ivana K Kim MD Tolga Kocaturk MD USA
Instructor of Ophthalmology Department of Ophthalmology
Retina Service Adnan Menderes University Medical School
Massachusetts Eye and Ear Infirmary Aydin
Harvard Medical School Turkey
Boston MA
USA xxxiii
List of Contributors

Ramsay S Kurban MD Jonathan H Lass MD Andrea Leonardi MD


Clinical Assistant Professor Charles I Thomas Professor and Chairman Assistant Professor in Ophthalmology
Department of Dermatology CWRU Department of Ophthalmology and Department of Neuroscience, Ophthalmology
Penn State University Visual Sciences Chairman Unit
Milton S Hershey Medical Center Department of Ophthalmology and Visual University of Padua
Hershey PA Sciences Padua
USA University Hospitals Case Medical Center Italy
Cleveland OH
Paul A Kurz MD USA Simmons Lessell MD
Instructor of Ophthalmology Director, Neuro-Ophthalmology Service
Casey Eye Institute Mary G Lawrence MD MPH Massachusetts Eye and Ear Infirmary
Oregon Health & Science University Associate Professor, Glaucoma, Cataract and Professor, Harvard Medical School
Portland OR Visual Rehabilitation Boston MA
USA Glaucoma Service USA
University of Minnesota Medical School
J R Kuszak PhD Minneapolis MN Leonard A Levin MD PhD
Departments of Ophthalmology and USA Professor of Ophthalmology and Visual
Pathology Sciences, Neurology, and Neurological
Rush University Medical Center Andrew G Lee MD Surgery
Chicago IL Professor of Ophthalmology, Neurology and University of Wisconsin School of Medicine
USA Neurosurgery and Public Health
Departments of Ophthalmology, Neurology Madison WI
Young H Kwon MD PhD and Neurosurgery USA
Associate Professor of Ophthalmology University of Iowa Hospitals Canada Research Chair of Ophthalmology
Department of Ophthalmology Iowa City IA and Visual Sciences
University of Iowa USA University of Montreal
Iowa City IA Montreal QC
USA Carol M Lee MD Canada
Clinical Professor, Department of
Thad A Labbe MD Ophthalmology Grace A Levy-Clarke MD
Glaucoma Specialist NYU Medical Center Fellowship Program Director
Ophthalmologist New York NY Uveitis and Ocular Immunology
Eye Associates of Central Texas USA Laboratory of Immunology
Austin TX National Eye Institute
USA Michael S Lee MD National Institutes of Health
Associate Professor Bethesda MD
Deborah L Lam MD Departments of Ophthalmology, Neurology,
Pacific Northwest Eye Associates USA
and Neurosurgery
Tacoma WA University of Minnesota Julie C Lew MD
USA Minneapolis MN Assistant Clinical Professor
Jeffrey C Lamkin MD USA Suny Downstate Medical Centre
Department of Ophthalmology Department of Ophthalmology
Paul P Lee MD JD Brooklyn NY
Akron City Hospital Professor of Ophthalmology
The Retina Group of NE Ohio Inc USA
Department of Ophthalmology
Akron OH Duke University Eye Center Craig Lewis MD
USA Durham NC Cole Eye Institute
Kathleen A Lamping MD USA Cleveland Clinic
Associate Clinical Professor Cleveland OH
William B Lee MD USA
Department of Ophthalmology Eye Consultant
Case Western Reserve University Eye Consultants of Atlanta Wei Li MD PhD
South Euclid OH Piedmont Hospital Research Fellow
USA Atlanta GA Ocular Surface Center
Anne Marie Lane MPH USA Miami FL
Clinical Research Manager, Retina Service USA
Igal Leibovitch MD
Massachusetts Eye and Ear Infirmary Oculoplastic and Orbital Division Laurence S Lim MBBS
Instructor in Ophthalmology, Harvard Medical Ophthalmology Department Principal Investigator
School Tel-Aviv Medical Center Singapore National Eye Centre
Boston MA Tel-Aviv Singapore
USA Israel
Lyndell L Lim MBBS FRANZCO
Katherine A Lane MD Bradley N Lemke MD FACS Mankiewicz-Zelkin Crock Fellow
Resident, Oculoplastic and Orbital Surgery Clinical Professor of Oculofacial Surgery Centre for Eye Research Australia
Service Department of Ophthalmology and Visual University of Melbourne
Wills Eye Hospital Sciences East Melbourne VIC
Philadelphia PA University of Wisconsin - Madison Australia
USA Madison WI
USA Wee-Kiak Lim FRCOphth FRCS(Ed) MMED
Keith J Lane MD Associate Consultant
Senior Manager, Research and Development Craig A Lemley MD Ocular Inflammation and Immunology
/Preclinical The Eye Institute Singapore National Eye Centre
ORA Clinical Research and Development Medical College of Wisconsin Singapore
North Andover MA Milwaukee WI
USA USA Grant T Liu MD
Neuro-ophthalmologist
Children’s Hospital of Philadelphia
Philadelphia PA
USA

xxxiv
List of Contributors

John I Loewenstein MD Robert E Marc PhD Michael McCrakken


Associate Professor of Ophthalmology Director of Research Clinical Instructor
Retina Service John A Moran Eye Center Department of Ophthalmology
Massachusetts Eye and Ear Infirmary Salt Lake City UT University of Colorado Health Sciences
Harvard Medical School USA Center
Boston MA Denver CO
USA Mellone Marchong USA
Department of Applied Molecular Oncology
McGregor N Lott MD Ontario Cancer Institute - University Health James P McCulley MD
Department of Ophthalmology Network Professor & Chairman of Ophthalmology
Medical College of Georgia Princess Margaret Hospital Department of Ophthalmology
Augusta GA Toronto ON University of Texas Southwestern Medical
USA Canada Center
Dallas TX
Jonathan C Lowry MD Dennis M Marcus MD USA
Ophthalmologist Professor of Clinical Ophthalmology
Morganton Eye Physicians Department of Clinical Ophthalmology John A McDermott
Morganton NC Southeast Retina Center Assistant Clinical Professor of Ophthalmology
USA Augusta GA Department of Ophthalmology
USA New York Eye and Ear Infirmary
David B Lyon MD FACS New York NY
Associate Professor Julie A Mares PhD USA
Department of Ophthalmology Professor
University of Missouri-Kansas City School of Department of Ophthalmology & Visual H Richard McDonald MD
Medicine Sciences Director, San Francisco Retina Foundation
Prairie Village KS WARF Co-Director, Vitreoretinal Fellowship
USA Madison WI California Pacific Retina Center
USA West Coast Retina Medical Group
Robert E Lytle MD San Francisco CA
Ophthalmologist Brian P Marr MD USA
Massachusetts Eye and Ear Infirmary Oncology Service
Harvard Medical School Wills Eye Institute Marguerite B McDonald MD FACS
Boston MA Thomas Jefferson University Ophthalmic Consultants of Long Island
USA Philadelphia PA Lynbrook NY
USA USA
Mathew MacCumber MD PhD
Associate Professor Carlos E Martinez MS MD Peter J McDonnell MD
Associate Chairman of Research Eye Physicians of Long Beach William Holland Wilmer Professor of
Rush University Medical Center Long Beach CA Ophthalmology
Chicago IL USA Director, Wilmer Ophthalmological Institute
USA Johns Hopkins University School of Medicine
Robert W Massof PhD Baltimore MD
Bonnie T Mackool MD MSPH Professor of Ophthalmology, Professor of USA
Director of Dermatology Neuroscience
Consultation Service Director, Lions Vision Research and Robert McGillivray BSEE CLVT
Massachusetts General Hospital Rehabilitation Center Director
Boston MA Wilmer Ophthalmological Institute Low Vision Services
USA Johns Hopkins University School of Medicine The Carroll Center for the Blind
Baltimore MD Low Vision Engineering Consultant
Nalini A Madiwale MD USA Massachusetts Commission for the Blind
Physician Newton MA
Albany-Troy Cataract & Laser Associates Yukihiro Matsumoto USA
Troy NY Research Fellow
USA Ocular Surface Research and Education Craig A McKeown MD
Foundation Associate Professor of Clinical
Francis Mah MD Miami FL Ophthalmology
Assistant Professor of Ophthalmology USA Bascom Palmer Eye Institute
Department of Ophthalmology Miller School of Medicine
University of Pittsburgh Medical Center Cynthia Mattox MD University of Miami
Pittsburgh PA Assistant Professor of Ophthalmology Miami FL
USA Ophthalmology - New England Eye Center USA
Tufts-New England Medical Center
Martin A Mainster PhD MD FRCOphth Boston MA James McLaughlin MD
Fry Endowed Professor and Vice Chairman of USA Medical Writer
Ophthalmology Ophthalmic Research Associates, Inc
Department of Ophthalmology Marlon Maus MD North Andover MA
University of Kansas School of Medicine DrPH Candidate USA
Kansas City MO University of California at Berkeley
USA Berkeley CA W Wynn McMullen MD
USA Vitereoretinal Consultant
Michael H Manning Jr Coastal Eye Associates
c/o Sherman M Chamberlain MD FACP Cathleen M McCabe MD Houston TX
FACG Indiana LASIK Center USA
Medical College of Georgia Fort Wayne IN
Augusta GA USA Shlomo Melamed MD
USA The Sam Rothberg Glaucoma Centre
Steven A McCormick MD Goldschleger Eye Institute
Steven L Mansberger MD MPH Director of Pathology and Laboratory Sheba Medical Center
Associate Scientist Medicine Tel-Hashomer
Devers Eye Institute The New York Eye and Ear Infirmary Israel
Portland OR New York NY
USA USA xxxv
List of Contributors

George Meligonis FRCPath Neil R Miller MD A Linn Murphree MD


Corneoplastic Unit Professor of Ophthalmology, Neurology and Director
Queen Victoria Hospital Neuro-Ophthalmology The Retinoblastoma Centre
East Grinstead Departments of Ophthalmology, Neurology Childrens Hospital of Los Angeles
East Sussex and Neuro-Ophthalmology Los Angeles CA
United Kingdom Wilmer Eye Institute USA
Johns Hopkins Hospital
Efstratios Mendrinos MD Baltimore MD Robert P Murphy MD
Ophthalmic Fellow USA The Retina Group of Washington
Ophthalmic Service Fairfax VA
Geneva University David M Mills MD USA
Geneva Oculofacial Plastic, Reconstructive, and
Switzerland Cosmetic Surgeon Timothy G Murray MD MBA FACS
Nicolitz Eye Consultants Professor of Ophthalmology
Dale R Meyer MD Jacksonville FL Department of Ophthalmology
Director, Ophthalmic Plastic Surgery USA Bascom Palmer Eye Institute
Professor of Ophthalmology Miami FL
Lions Eye Institute Monte D Mills MD USA
Albany Medical Center Chief, Division of Ophthalmology
Albany NY Children’s Hospital of Philadelphia Philip I Murray PhD FRCP FRCS FRCOphth
USA Philadelphia PA Professor of Ophthalmology
USA Academic Unit of Ophthalmology
Catherine B Meyerle MD Birmingham and Midland Eye Centre
Retinal Physician Tatyana Milman MD City Hospital NHS Trust
National Eye Institute Assistant Professor of Ophthalmology Birmingham
National Institutes of Health Co-director, Ophthalmic Pathology Division United Kingdom
Bethesda MD Institute of Ophthalmology and Visual
USA Science Karina Nagao MD
UMDNJ-New Jersey Medical School Harvard Medical School
William F Mieler MD Newark NJ Boston MA
Professor and Chairman USA USA
Department of Ophthalmology and Visual
Science Lylas Mogk MD Jay Neitz PhD
University of Chicago Director R D and Linda Peters Professor
Chicago IL Visual Rehabilitation and Research Center Department of Ophthalmology
USA Henry Ford Health System Medical College of Wisconsin
Livonia MI Milwaukee WI
Michael Migliori MD USA USA
Clinical Associate Professor
The Warren Alpert Medical School Marja Mogk PhD Maureen Neitz PhD
Brown University Assistant Professor of English Richard O Schultz-Ruth A
Providence RI California Lutheran University Works-Ophthalmology Research Professor
USA Los Angeles CA The Eye Institute
USA Medical College of Wisconsin
Martin C Mihm Jr, MD Milwaukee WI
Clinical Professor of Pathology Jordi Monés MD USA
Senior Dermatopathologist Associate Professor of Ophthalmology
The Pigmented Lesion Clinic Institut de la Macula i de la Retina Peter A Netland MD PhD
Massachusetts General Hospital Barcelona Siegal Professor of Ophthalmology, Director
Boston MA Spain of Glaucoma, Academic Vice-Chair
USA Department of Ophthalmology
Robert Montes-Micó OD MPhil Hamilton Eye Institute
Darlene Miller DHSc MPH SM CIC Optica University of Tennessee Health Science
Research Assistant Professor Facultat de Fisica Center
Scientific Director Universidad de Valencia Memphis TN
Abrams Ocular Microbiology Laboratory Valencia USA
Bascom Palmer Eye Institute Spain
Anne Bates Leach Eye Hospital Arthur H Neufeld PhD
Miller School of Medicine Christie L Morse MD Professor of Ophthalmology
University of Miami Concord Eye Care Forsythe Laboratory for the Investigation of
Miami FL Concord NH Aging Retina
USA USA Northwestern University Fienberg School of
Medicine
David Miller MD Asa D Morton MD Chicago IL
Associate Clinical Professor of Ophthamology Eye Care of San Diego/CA Laser Vision, Inc USA
Department of Ophthalmology Escondido CA
Harvard Medical School USA Nancy J Newman MD
Jamaica Plain MA Professor of Ophtalmology and Neurology
Anne Moskowitz OD PhD Neuro-Ophthalmology Unit
USA Research Associate in Ophthalmology Emory Eye Center
Joan W Miller MD Children’s Hospital, Boston Atlanta GA
Henry Willard Williams Professor of Instructor of Ophthalmology USA
Ophthalmology Harvard Medical School
Chief and Chair, Department of Boston MA Eugene W M Ng MD
Ophthalmology USA Eyetech Pharmaceuticals, Inc
Massachusetts Eye and Ear Infirmary New York NY
Shizuo Mukai MD USA
Harvard Medical School Assistant Professor of Ophthalmology
Boston MA Massachusetts Eye and Ear Infirmary
USA Harvard Medical School
Boston MA
xxxvi USA
List of Contributors

Quan Dong Nguyen MD MSc Yen Hoong Ooi MD Louis R Pasquale MD


Assistant Professor of Ophthalmology c/o Douglas Rhee MD Co-Director, Glaucoma Service
Diseases of the Retina and Vitreous, and Massachusetts Eye and Ear Infirmary Assistant Professor of Ophthalmology
Uveitis Harvard Medical School Department of Ophthalmology
Wilmer Eye Institute Boston MA Massachusetts Eye and Ear Infirmary
Johns Hopkins Hospital USA Harvard Medical School
Baltimore MD Boston MA
USA E Mitchel Opremcak MD USA
Clinical Associate Professor
Jerry Y Niederkorn PhD Department of Ophthalmology Neha N Patel MD
Professor of Ophthalmology Ohio State University College of Medicine Resident
Department of Ophthalmology Columbus OH Department of Ophthalmic and Visual
University of Texas Southwestern Medical USA Science
Center University of Chicago
Dallas TX George Ousler BS Chicago IL
USA Director USA
Dry Eye Department
Robert J Noecker MD Ophthalmic Research Associates Sayjal J Patel MD
Vice Chair, Clinical Affairs North Andover MA Wilmer Eye Institute
Eye and Ear Institute USA Baltimore MD
Associate Professor USA
University of Pittsburgh School of Medicine Randall R Ozment MD
Pittsburgh PA Physician Thomas D Patrianakos DO
USA Dublin Eye Associates Attending Physician
Dublin GA Division of Ophthalmology
Robert B Nussenblatt MD MPH USA John H Stroger Hospital of Cook County
Scientific Director and Chief, Laboratory of Chicago IL
Immunology, Intramural Program Samuel Packer MD USA
Section Head, Clinical Immunology Section Professor of Clinical Ophthalmology,
National Eye Institute New York University School of Medicine James R Patrinely MD FACS
National Institutes of Health Chair, Department of Ophthalmology Plastic Eye Surgery Associates PLLC
Bethesda MD North Shore Long Island Jewish Health Houston TX
USA System USA
New York NY
Joan M O’Brien MD USA Deborah Pavan-Langston MD FACS
Professor of Ophthalmology and Pediatrics Associate Professor of Ophthalmology
Director of Ocular Oncology Millicent L Palmer MD Surgeon and Director of Clinical Virology
Division of Ophthalmology Associate Professor, Department of Surgery Massachusetts Eye and Ear Infirmary
University of California San Francisco Creighton University Medical School Harvard School of Medicine
Medical Center Division of Ophthalmology Boston MA
San Francisco CA Creighton University Medical Center USA
USA Omaha NE
USA Eli Peli MSc OD
Paul D O’Brien FRCSI MRCOphth MMedSci Professor of Ophthalmology
Specialist Registrar in Ophthalmology George N Papaliodis MD Harvard Medical School
Royal Victoria Eye and Ear Hospital Instructor in Ophthalmology and Internal Moakley Scholar in Aging Eye Research
Dublin Medicine Schepens Eye Research Institute
Ireland Massachusetts Eye and Ear Infirmary Boston MA
Harvard Medical School USA
Terrence P O’Brien MD Boston MA
Professor of Ophthalmology USA Susan M Pepin MD
Charlotte Breyer Rodgers Distinguished Chair Assistant Professor of Surgery
in Ophthalmology D J John Park MD Section of Ophthalmology
Director of the Refractive Surgery Service Resident Dartmouth Hitchcock Medical Center
Bascom Palmer Eye Institute Department of Plastics and Reconstructive Lebanon NH
Palm Beach FL Surgery USA
USA University of California
Victor L Perez MD
Irvine CA
Assistant Professor
Denis O’Day MD FACS USA
Bascom Palmer Eye Institute
Professor of Ophthalmology
David W Parke II MD University of Miami School of Medicine
Department of Ophthalmology
Edward L Gaylord Professor and Chairman Miami FL
Vanderbilt Eye Institute
Department of Ophthalmology USA
Nashville TN
USA President and CEO Juan J Pérez-Santonja MD PhD
The Dean A McGee Eye Institute Instituto Oftalmológico de Alicante
R Joseph Olk MD Oklahoma City OK Alicante
Bond Eye Associates USA Spain
Peoria IL
USA Cameron F Parsa MD John R Perfect MD
Assistant Professor of Ophthalmology Director, Duke University Mycology Research
Karl R Olsen MD Krieger Children’s Eye Center Unit (DUMRU)
Clinical Assistant Professor of Ophthalmology The Wilmer Eye Institute Division of Infectious Diseases
University of Pittsburgh School of Medicine Baltimore MD Department of Medicine
Retina Vitreous Consultants USA Duke University
Pittsbrugh PA Winston-Salem NC
USA M Andrew Parsons FRCPath
Honorary Consultant in Ophthalmic USA
Sumru Onal MD Pathology
Department of Ophthalmology Academic Unit of Pathology
Marmara University School of Medicine Royal Hallamshire Hospital
Istanbul Sheffield
Turkey United Kingdom xxxvii
List of Contributors

Henry D Perry MD FACS Valerie Purvin MD Elias Reichel MD


Founding Partner Clinical Professor of Ophthalmology & Associate Professor of Ophthalmology
Director: Cornea Division Neurology Vitreoretinal Diseases
Ophthalmic Consultants of Long Island Departments of Ophthalmology and New England Eye Center
Rockville Center NY Neurology Tufts University School of Medicine
USA Indiana Medical Center Boston MA
Indianapolis IN USA
Joram Piatigorsky PhD USA
Chief Martin H Reinke MD
Laboratory of Molecular and Developmental David A Quillen MD Private Practice
Biology George and Barbara Blankenship Professor Southlake TX
National Eye Institute - National Institute of and Chair USA
Health Department of Ophthalmology
Bethesda MD Penn State College of Medicine Douglas Rhee MD
USA Hershey PA Assistant Professor of Ophthalmology
USA Department of Ophthalmology
Dante Pieramici MD Massachusetts Eye and Ear Infirmary
Co-Director Graham E Quinn MD Harvard Medical School
California Retina Consultants Attending Surgeon, Research Fellow Boston MA
Santa Barbara CA Department of Ophthalmology USA
USA The Childrens Hospital of Philadelphia
Philadelphia PA Claudia U Richter MD
Eric A Pierce MD PhD Ophthalmic Consultants of Boston
Assistant Professor of Ophthalmology USA
Boston MA
F.M. Kirby Center for Molecular Melvin D Rabena BSc USA
Ophthalmology Director of Research
Scheie Eye Institute California Retina Consultants Joseph F Rizzo lll MD
University of Pennsylvania School of Medicine Santa Barbara CA Associate Professor of Ophthalmology
Philadelphia PA USA Massachusetts Eye and Ear Infirmary
USA Harvard Medical School
James L Rae PhD Boston MA
Roberto Pineda II MD Professor of Ophthalmology and Physiology USA
Assistant Professor Physiology and Biomedical Engineering
Massachusetts Eye and Ear Infirmary Mayo Clinic Richard M Robb MD
Chief of Ophthalmology, Brigham & Women’s Rochester MN Associate Professor of Ophthalmology
Hospital, Boston USA Harvard Medical School
Assistant Professor, Department of Department of Ophthalmology
Ophthalmology, Harvard Medical School Michael B Raizman MD Children’s Hospital Boston
Boston MA Ophthalmic Consultant Boston MA
USA Ophthalmic Consultants Of Boston USA
Associate Professor of Ophthalmology
Misha L Pless MD Tafts University School of Medicine Anja C Roden MD
Director, Division of General Neurology Boston MA c/o Diva R Salomao MD
Massachusetts General Hospital USA Department of Pathology
Boston MA Mayo Clinic
USA Alessandro Randazzo MD Rochester MN
Department of Ophthalmology USA
Howard D Pomeranz MD PhD Istituto Clinico Humanitas Rozzano
Clinical Associate Professor Milano University I Rand Rodgers MD
Department of Ophthalmology Milan Assistant Clinical Professor, Mount Sinai
North Shore Long Island Jewish Health Italy Medical Center
System Director of Ophthalmic Facial and Plastic
Great Neck NY Narsing A Rao MD Surgery
USA Professor of Ophthalmology and Pathology North Shore University Hospital NYU
Doheny Eye Institute Private Practice
Constantin J Pournaras MD University of California New York NY
Department of Ophthalmology Los Angeles CA USA
Geneva University Hospitals USA
Geneva Merlyn M Rodrigues MD PhD
Switzerland Christopher J Rapuano MD c/o Kelly S Keefe MD
Co-Director Cornea Service Naval Medical Center
William Power MBBCH FRCS FRCOphth Co-Director Professor of Ophthalmology, San Diego CA
Consultant Ophthalmic Surgeon Jefferson Medical College USA
Blackrock Clinic Thomas Jefferson University
Blackrock Co-Director, Cornea Service Yonina Ron MD
Co Dublin Refractive Surgery Department Department of Ophthalmology
Ireland Wills Eye Hospital Rabin Medical Center
Philadelphia PA Beilinson Campus
Manvi Prakash MD Petah Tiqva
Postdoctoral Fellow USA
Israel
Joslin Diabetes Center Sherman W Reeves MD MPH
Beetham Eye Institute Cornea, External Disease and Retractive Geoffrey E Rose DSC MS MRCP FRCS
Harvard Medical School Surgery FRCOphth
Boston MA Minnesota Eye Consultants Consultant Ophthalmic Surgeon
USA Minneapolis MN Adnexal Department
USA Moorfields Eye Hospital
Anita G Prasad MD London
Department of Ophthalmology and Visual Carl D Regillo MD FACS United Kingdom
Sciences Professor of Ophthalmology
Washington University Medical School Wills Eye Hospital
St Louis MO Philadelphia PA
xxxviii USA USA
List of Contributors

Emanuel S Rosen MD FRCS FRCOphth Mark S Ruttum MD Michael A Sandberg PhD


Consultant Ophthalmic Surgeon Professor of Ophthalmology Associate Professor of Ophthalmology
Manchester Central Health Care Authority Head, Pediatric Ophthalmology and Adult Berman-Gund Laboratory
Manchester Strabismus Section Massachusetts Eye and Ear Infirmary
United Kingdom Medical College of Wisconsin Harvard Medical School
Milwaukee WI Boston MA
James T Rosenbaum MD USA USA
Professor of Medicine, Ophthalmology and
Cell Biology Allan R Rutzen MD FACS Virender S Sangwan MD
Chief, Division of Arthritis and Rheumatic Associate Professor of Ophthalmology Head, Cornea and Anterior Segment Services
Diseases Department of Ophthalmology L V Prasad Eye Institute
Director, Uveitis Clinic Casey Eye Institute University of Maryland Hyderabad
Oregon Health and Science University Baltimore MD India
Portland OR USA
Maria A Saornil MD
USA
Edward T Ryan MD Ocular Pathology Unit
Perry Rosenthal MD Director, Tropical & Geographic Medicine Hospital Clinico Universitario
Assistant Clinical Professor of Ophthalmology Center Valladolid
Department of Ophthalmology Massachusetts General Hospital Spain
Boston Foundation for Sight Associate Professor of Medicine Joseph W Sassani MD
Boston MA Harvard Medical School Professor of Ophthalmology and Pathology
USA Assistant Professor Pennsylvania State University
Dept of Immunology and Infectious Diseases Hershey Medical Center
Strutha C Rouse II MD Harvard School of Public Health
Horizon Eye Care Hershey PA
Boston MA USA
Charlotte NC USA
USA Rony R Sayegh MD
Alfredo A Sadun MD PhD Research Fellow
Barry W Rovner MD Thornton Professor of Ophthalmology and
Professor & Medical Director Cornea and Refractive Surgery Service
Neurosurgery Massachusetts Eye and Ear Infirmary
Department of Psychiatry and Human Doheny Eye Institute
Behavior Department of Ophthalmology
Kech School of Medicine Boston MA
Thomas Jefferson University University of California
Philadelphia PA USA
Los Angeles CA
USA USA Andrew P Schachat MD
Malgorzata Rozanowska PhD Vice Chairman for Clinical Affairs
José-Alain Sahel MD Cole Eye Institute
Lecturer Professor of Ophthalmology
School of Optometry and Vision Sciences Cleveland Clinic Foundation
Head, Laboratory of Retinal Pathobiology Cleveland OH
Cardiff University University Louis Pasteur
Cardiff USA
Strasbourg
United Kingdom France Wiley A Schell MD
Michael P Rubin MD Director, Medical Mycology Research Center
Leorey Saligan MD Assistant Professor of Medicine Department
Fellow in Vitreoretinal Diseases and Surgery Nurse Practitioner
Massachusetts Eye and Ear Infirmary, of Medicine
National Eye Institute Division of Infectious Diseases and
Harvard Medical School National Institutes of Health
Boston MA International Health
Bethesda MD Duke University Medical Center
USA USA Durham NC
Peter A D Rubin MD FACS Sarwat Salim MD FACS USA
Eye Plastics Consultant Assistant Clinical Professor of Ophthalmology
Brookline MA Amy C Schefler MD
Yale Eye Center Resident in Ophthalmology
Associate Clinical Professor Yale University School of Medicine
Harvard Medical School Bascom Palmer Eye Institute
New Haven CT Miami FL
USA USA USA
Shimon Rumelt MD John F Salmon MD FRCS FRCOphth
Attending Physician Tina Scheufele MD
Consultant Ophthalmic Surgeon Vitreoretinal Surgeon
Ophthalmology Department The Radcliffe Infirmary
Western Galilee - Nahariya Medical Center Ophthalmic Consultants of Boston
Oxford Eye Hospital Boston MA
Nahariya Oxford
Israel USA
United Kingdom
Anil K Rustgi MD Vivian Schiedler MD
Diva R Salomão MD Oculoplastic and Orbital Surgeon,
Professor of Medicine and Genetics Associate Professor of Pathology
Chief of Gastroenterology Charlottesville, VA
Department of Pathology Ophthalmic Plastic & Reconstructive Surgery
University of Pennsylvania Medical Center Mayo Clinic
Philadelphia PA Fellow
Rochester MN Department of Ophthalmology
USA USA University of Washington
Tina Rutar MD David Sami MD Seattle WA
Resident Division Chief for PSF Ophthalmology USA
Department of Ophthalmology CHOC Children’s Hospital
University of California San Francisco Gretchen Schneider MD
Orange CA Adjunct Assistant Professor in the Genetic
San Francisco CA USA
USA Counseling program
Genetic Counseling Faculty
Brandeis University
Waltham MA
USA
xxxix
List of Contributors

Alison Schroeder BA Irina Serbanescu BA Bradford J Shingleton MD


Laboratory Manager Research Assistant Clinical Professor of
Department of Ophthalmology Division of neurology Ophthalmology, Harvard Medical School
Boston University School of Medicine The Hospital for Sick Children Ophthalmic Consultants of Boston
Boston MA Toronto ON Boston MA
USA Canada USA
Ronald A Schuchard PhD Briar Sexton MD FRCSC John W Shore MD FACS
Director of Rehabilitation Research and Fellow in Neuro-Ophthalmology Texas Oculoplastics Consultants
Development Center VGH Eye Care Center Austin TX
Associate Professor Vancouver BC USA
Department of Neurology Canada
Emory University School of Medicine Lesya M Shuba MD PhD
Atlanta GA Tarek M Shaarawy MD Assistant Professor
USA Chef Department of Ophthalmology & Visual
Clinique d’ophtalmologie Sciences
Joel S Schuman MD Secteur du Glaucome Dalhousie University
Eye and Ear Foundation Professor and Hôpitaux Universitaires de Génève Halifax NS
Chairman Génève Canada
Department of Ophthalmology Switzerland
University of Pittsburgh School of Medicine Guy J Ben Simon MD
Pittsburgh PA Peter Shah BSc (Hons) MBChB FRCOphth Goldschleger Eye Institute
USA Consultant Sheba Medical Center
Birmingham and Midland Eye Centre Tel Hashomer
Ivan R Schwab MD FACS City Hospital Israel
Professor of Ophthalmology Birmingham
Department of Ophthalmology United Kingdom Richard J Simmons MD
University of California at Davis Emeritus Ophthalmic Surgeon
Sacramento CA Aron Shapiro BS Harvard Medical School
USA Director Boston MA
Anti-inflammatory/Anti-infectives Department USA
Adrienne Scott MD Ophthalmic Research Associates
Clinical Associate North Andover MA Michael Simpson
Vitreoretinal Surgery USA c/o David Miller MD
Duke University Eye Center Department of Ophthalmology
Durham NC Savitri Sharma MD MAMS Harvard Medical School
USA Associate Director, Laboratory Services Jamaica Plain MA
L V Prasad Eye Institute USA
Ingrid U Scott MD MPH Bhubaneswar, Orissa
Professor of Ophthalmology and Health India Arun D Singh MD
Evaluation Sciences Director
Department of Ophthalmology Jean Shein MD Department of Ophthalmic Oncology
Penn State College of Medicine Attending Physician Cole Eye Institute and Taussing Cancer Center
Hershey PA Crane Eye Care Hana Kukui Center Cleveland OH
USA Lihue HI USA
USA
Marvin L Sears MD Omah S Singh MD
Professor and Chairman Emeritus Debra J Shetlar MD Director
Department of Ophthalmology and Visual Associate Professor of Ophthalmology New England Eye Center
Science Baylor College of Medicine Beverley MA
Yale University School of Medicine Staff Physician USA
New Haven CT Michael E DeBakey V A Medical Center
Houston TX Karen Sisley BSc PhD
USA Non-Clinical Lecturer Ocular Oncology
USA
Johanna M Seddon MD ScD Academic Unit of Ophthalmology and
Professor of Ophthalmology M Bruce Shields MD Orthoptics
Tufts University School of Medicine Professor of Ophthalmology and Visual University of Sheffield
Director, Ophthalmic Epidemiology and Science Sheffield
Genetics Service Yale Eye Center United Kingdom
New England Eye Center New Haven CT
USA Arthur J Sit MD
Boston MA Assistant Professor of Ophthalmology
USA Carol L Shields MD Mayo Clinic
Theo Seiler MD PhD Professor of Ophthalmology, Thomas Rochester MN
Professor Jefferson Medical College USA
Institut für Refractive und Ophthalmochirurgie Attending Surgeon and Associate Director
David Smerdon FRCSEd FRCOphth
(IROC) Wills Eye Hospital
Consultant Ophthalmologist
Zürich Philadelphia PA
James Cook University Hospital
Switzerland USA
Middlesbrough
Jerry A Shields MD United Kingdom
Robert P Selkin MD
Private Practice Professor of Ophthalmology, Thomas William E Smiddy MD
Plano TX Jefferson University Professor of Ophthalmology
USA Director Department of Ophthalmology
Oncology Services Bascom Palmer Eye Institute
Richard D Semba MD MA MPH Wills Eye Hospital Miami FL
W Richard Green Professor of Philadelphia PA USA
Ophthalmology USA
Wilmer Eye Institute
Baltimore MD
USA
xl
List of Contributors

Ronald E Smith MD Tomy Starck MD Timothy J Sullivan FRANZCO FRACS


Professor and Chair Director Eyelid, Lacrimal and Orbital Clinic
Department of Ophthalmology UltraVision Center Department of Ophthalmology
Keck School of Medicine of USC San Antonio TX Royal Brisbane Hospital
Los Angeles CA USA Herston QLD
USA Australia
Walter J Stark MD
Terry J Smith MD Professor of Ophthalmology Jennifer K Sun MD
Professor and Head Director of the Stark-Mosher Center Lecturer
Division of Molecular Medicine The John Hopkins Hospital, Wilmer Eye Joslin Diabetes Center
David Geffen School of Medicine Institute Beetham Eye Institute
Harbor-UCLA Medical Center Baltimore MD Harvard Medical School
Torrance CA USA Boston MA
USA USA
Joshua D Stein MD MS
Neal G Snebold MD Assistant Professor Janet S Sunness MD
Ophthalmologist Department of Ophthalmology and Visual Medical Director
Massachusetts Eye and Ear Infirmary Sciences Richard E Hoover Rehabilitation Services for
Harvard Medical School Kellogg Eye Center Low Vision and Blindness
Boston MA Ann Arbor MI Greater Baltimore Medical Center
USA USA Baltimore MD
Lucia Sobrin MD USA
Roger F Steinert MD
Instructor of Ophthalmology Professor of Ophthalmology and Biomedical Francis C Sutula MD
Retina and Uvetis Services Engineering Milford Eye Care
Massachusetts Eye and Ear Infirmary Director of Cornea, Refractive and Cataract Milford MA
Harvard Medical School Surgery USA
Boston MA Vice Chair of Clinical Ophthalmology
USA Department of Ophthalmology Nasreen A Syed MD
University of California Irvine Assistant Professor, Ophthalmology and
John A Sorenson MD Pathology
Attenting Surgeon Irvine CA
USA Department of Ophthalmology and Visual
Vitreoretinal Service Sciences
Manhattan Eye, Ear, and Throat Hospital Leon Strauss MD University of Iowa
New York NY Instructor Iowa City IA
USA Wilmer Eye Institute USA
Sarkis H Soukiasian MD John Hopkins University School of Medicine
Baltimore MD Christopher N Ta MD
Director: Cornea and External Disease
USA Associate Professor of Ophthalmology
Director: Ocular Inflammation and Uveitis
Department of Ophthalmology
Lahey Clinic
Barbara W Streeten MD Stanford University
Burlington MA
Professor of Ophthalmology and Pathology Palo Alto CA
USA
State University of New York USA
George L Spaeth MD FRCO FACS Upstate Medical University
Louis Esposito Research Professor of Syracuse NY Hidehiro Takei MD
Ophthalmology USA Staff Pathologist
Jefferson Medical College Department of Pathology
Director of the William & Anna Goldberg J Wayne Streilein MD (deceased) The Methodist Hospital
Glaucoma Service Formerly Senior Scientist, President, Charles Houston TX
Wills Eye Institute L Schepens Professor of Ophthalmology, USA
Philadelphia PA Professor of Dermatology
Formerly Vice Chair for Research, Jonathan H Talamo MD
USA Associate Clinical Professor of
Department of Ophthalmology
Richard F Spaide MD Harvard Medical School Ophthalmology
Associate Clinical Professor of Ophthalmology Boston MA Department of Ophthalmology
Manhattan Eye, Ear, and Throat Hospital USA Harvard Medical School
New York NY Waltham MA
USA James D Strong CRA USA
Senior Ophthalmic Imager
Monika Srivastava MD Department of Ophthalmology Richard R Tamesis MD
Clinical Assistant Professor Penn State Milton S Hershey Medical Center Department of Ophthalmology
Department of Dermatology Hershey PA Loma Linda University Medical Center
New York University USA Loma Linda CA
New York NY USA
USA Ilene K Sugino MS
Director, Ocular Cell Transplantation Madhura Tamhankar MD
Sunil K Srivastava MD Laboratory Associate Professor
Assistant Professor of Ophthalmology Institute of Ophthalmology and Visual Science Department of Ophthalmology
Section of Vitreoretinal Surgery & Disease New Jersey Medical School University of Pennsylvania Medical School
Emory Eye Center Newark NJ Philadelphia PA
Atlanta GA USA USA
USA Kristen J Tarbet MD SACS
Eric B Suhler MD MPH
Alexandros N Stangos MD Chief of Ophthalmology Private Practice
Division of Ophthalmology Portland VA Medical Center Bellevue WA
Department of Clinical Neurosciences Assistant Professor of Ophthalmology and USA
University Hospitals of Geneva Co-director
Geneva Department of Ophthalmology
Switzerland Casey Eye Institute
Portland OR
USA
xli
List of Contributors

Michelle Tarver-Carr MD PhD Gail Torkildsen MD Russell N Van Gelder MD PhD


Assistant, Ocular Immunology Physician Associate Professor of Ophthalmology and
Wilmer Eye Institute Andover Eye Associates Visual Sciences
Departments of Medicine and Epidemiology Andover MA Department of Ophthalmology and Visual
Johns Hopkins University School of Medicine USA Sciences
Baltimore MD Washington University School of Medicine
USA Cynthia A Toth MD St Louis MO
Associate Professor of Ophthalmology and USA
Mark A Terry MD Biomedical Engineering
Director, Corneal Services Duke Eye Center Gregory P Van Stavern MD
Clinical Professor, Department of Durham NC Assitant Professor of Ophthalmology,
Ophthalmology USA Neurology and Nerosurgery
Devers Eye Institute Kresge Eye Institute
Oregon Health Sciences University Elias I Traboulsi MD Wayne State University
Portland OR Professor of Ophthalmology Detroit MI
USA The Cole Eye Institute USA
Cleveland OH
Joseph M Thomas MD USA Deborah K Vander Veen MD
Associate Clinical Professor Assistant Professor
Department of Neurology Michele Trucksis PhD MD Department of Ophthalmology
Case Western Reserve University School of Associate Clinical Professor Children’s Hospital and Harvard Medical
Medicine Harvard Medical School School
Cleveland OH Associate Director Clinical Pharmacology Boston MA
USA Merck & Co. Inc USA
Boston MA
Vance Thompson MD USA Demetrios Vavvas MD PhD
Assistant Professor of Medicine Instructor in Ophthalmology
University of South Dakota School of James C Tsai MD Retina Service Department of Ophthalmology
Medicine Robert R Young Professor and Chairman Massachusetts Eye and Ear Infirmary
Director of Refractive Surgery Department of Ophthalmology and Visual Harvard Medical School
Sioux Valley Clinic Science Boston MA
Vance Thompson Vision Yale University School of Medicine USA
Sioux Falls SD New Haven CT
USA USA David H Verity MA FRC Ophth
Consultant Ophthalmic Surgeon
Jennifer E Thorne MD PhD Julie H Tsai MD Adnexal Departments
Assistant Professor of Ophthalmology Assistant Professor Moorfields Eye Hospital
Division of Ocular Immunology Department of Ophthalmology London
Wilmer Eye Institute University of South Carolina School of United Kingdom
Baltimore MD Medicine
USA Columbia SC Paolo Vinciguerra MD
USA Medical Director
Matthew J Thurtell BSc(Med) MBBS MScMed Studio Oculistico Vincieye SRL
Neuro-Ophthalmology Fellow David T Tse MD FACS Milan
Department of Neurology Professor of Ophthalmology Italy
Royal Prince Albert Hospital Department of Ophthalmology
Sydney NSW Bascom Palmer Eye Institute Paul F Vinger MD
Australia Miami FL Clinical Professor
USA Ophthalmology
David P Tingey MD FRCSC Tufts University School of Medicine
Associate Professor Scheffer C G Tseng MD PhD New England Medical Center
Ivey Eye Institute Research Director Boston MA
London Health Sciences Center Ocular Surface Center USA
London ON Miami FL
Canada USA Nicholas J Volpe MD
Professor of Ophthalmology and Neurology
King W To MD Elmer Y Tu MD Vice Chair and Residency Program Director
Clinical Professor of Ophthalmology Associate Professor of Clinical Department of Ophthalmology
Brown University School of Medicine Ophthalmology PENN Eye Care
Barrington RI Director of the Cornea and External Disease Philadelphia PA
USA Service USA
Department of Ophthalmology
Faisal M Tobaigy MD University of Illinois at Chicago Werner Wackernagel MD
Department of Ophthalmology Chicago IL Physician
Massachusetts Eye and Ear Infirmary and the USA Department of Ophthalmology
Schepens Eye Research Institute Medical University Graz
Harvard Medical School Ira J Udell MD Graz
Boston MA Professor of Ophthalmology Austria
USA Albert Einstein College of Medicine
New York NY Sonal Desai Wadhwa MD
Michael J Tolentino MD USA Assistant Professor of Ophthalmology
Director of Research, Center for Retina and Division of Ophthalmology
Macular Disease Alejandra A Valenzuela MD University of Maryland
Center for Retina and Macular Disease Assistant Professor Baltimore MD
Winter Haven FL Department of Ophthalmology and Visual USA
USA Sciences
Dalhousie University
Melissa G Tong BSc Halifax NS
Department of Medicine Canada
Jefferson Medical College
Philadelphia PA
xlii USA
List of Contributors

Michael D Wagoner MD Scott M Whitcup MD Jules Winokur MD


Professor of Ophthalmology Executive Vice President North Shore Long Island Jewish Health
Department of Ophthalmology and Visual Head of Research and Development System
Sciences Allegran Inc New York NY
University of Iowa Hospitals and Clinics Irvine CA USA
Iowa City IA USA
USA William J Wirostko MD
Valerie A White MD FRCPC Associate Professor of Ophthalmology
Nadia K Waheed MD Professor The Eye Institute
Fellow Department of Pathology & Laboratory Medical College of Milwaukee
Immunology and Uveitis Service Medicine, Milwaukee WI
Department of Ophthalmology University of British Columbia USA
Massachusetts Eye and Ear Infirmary Vancouver General Hospital
Harvard Medical School Vancouver BC Gadi Wollstein MD
Boston MA Canada Assistant Professor and Director
USA Ophthalmic Imaging Research Laboratories
William L White MD The Eye & Ear Institute
David S Walton MD Department of Ophthalmology Dept of Ophthalmology
Clinical Professor of Ophthalmology The Eye Foundation UPMC Eye Center
Harvard Medical School University of Missouri-Kansas City Pittsburgh PA
Boston MA Kansas City MO USA
USA USA
Albert Chak Ming Wong FCOph(HK)
Martin Wand MD Jason Wickens MD FHKAM(Ophth)
Clinical Professor of Ophthalmology Barnes Retina Institute Associate Consultant
University of Connecticut School of Medicine Department of Ophthalmology Caritas Medical Center
Farmington CT Washington University School of Medicine Shamshuipo, Kowloon
USA St Louis MO Hong King
USA China
Jie Jin Wang MMed PhD
Associate Professor of Epidemiology Janey L Wiggs MD PhD Tien Y Wong MBBS MMED (Ophth) FRCSE
Westmead Millennium Institute Associate Professor of Ophthalmology FRANZCO FAFPHM MPH PhD
University of Sydney Massachusetts Eye and Ear Infirmary Professor of Ophthalmology
Sydney NSW Harvard Medical School Department of Ophthalmology & Centre for
Australia Boston MA Eye Research Australia
USA University of Melbourne
Scott M Warden MD East Melbourne VIC
Retina Service Jacob T Wilensky MD Australia
Massachusetts Eye and Ear Infirmary Professor of Ophthalmology
Department of Ophthalmology Director, Glaucoma Service John J Woog MD FACS
Harvard Medical School University of Illinois College of Medicine Associate Professor of Ophthalmology,
Boston MA Chicago IL Ophthalmic Plastic and Reconstructive
USA USA Surgery
Department of Ophthalmology
Lennox Webb FRCOphth FRCS(Ed) Charles P Wilkinson MD Mayo Clinic
Consultant Ophthalmic Surgeon Chairman, Department of Ophthalmology Rochester MN
Royal Alexandra Hospital Greater Baltimore Medical Center USA
Paisley Professor, Department of Ophthalmology
United Kingdom John Hopkins University Michael Wride PhD
Baltimore MD Lecturer
David Weber MD USA School of Optemetry and Vision Sciences
Assistant Professor Cardiff University
Department of Physical Medicine & Patrick D Williams MD Cardiff
Rehabilitation Vitreo Retinal Specialist United Kingdom
Mayo Clinic College of Medicine Texas Retina Associates
Rochester MN Arlington TX Carolyn S Wu MD
USA USA Instructor of Ophthalmology
Harvard Medical School
Daniel Wee MD David J Wilson MD Boston MA
Department of Ophthalmology Associate Professor USA
The Palmetto Health/ University of South Department of Ophthalmology;
Carolina School of Medicine Director, Christensen Eye Pathology Darrell WuDunn MD PhD
Columbia SC Laboratory Associate Professor of Ophthalmology
USA Casey Eye Institute Indiana University School of Medicine
Oregon Health Sciences University Indianapolis IN
Corey B Westerfeld MD Portland OR USA
Research Fellow USA
Department of Ophthalmology Jean Yang MD
Massachusetts Eye and Ear Infirmary M Roy Wilson MD MS Department of Ophthalmology
Harvard Medical School Chancellor North Shore-Long Island Jewish Medical
Boston MA University of Colorado and Health Sciences Center
USA Center Great Neck NY
Denver CO USA
Christopher T Westfall MD USA
Professor of Ophthalmology Lawrence A Yannuzzi MD
Jones Eye Institute & Arkansas Children’s Steven E Wilson MD Vice-Chairman, Department of
Hospital Director of Corneal Research and Professor Ophthalmology
University of Arkansas for Medical Sciences of Ophthalmology Director of Retinal Services
Little Rock AR The Cleveland Clinic Foundation Manhattan Eye, Ear and Throat Hospital
USA Cole Eye Institute New York NY
Cleveland OH USA
USA xliii
List of Contributors

Michael J Yaremchuk MD Jenny Y Yu MD Marco Zarbin MD PhD FACS


Clinical Professor of Surgery Consulting Physician Professor of Ophthalmology and
Harvard Medical School Department of Ophthalmology Neuroscience
Boston MA UPMC Children’s Hospital of Pittsburgh Department of Ophthalmology
USA Pittsburgh PA Institute of Ophthalmology and Visual
USA Science
R Patrick Yeatts MD FACS University of Medicine and Dentistry,
Professor and Vice Chairman Beatrice Y J T Yue PhD New Jersey
Department of Ophthalmology Thanis A Field Professor of Ophthamology Newark NJ
Wake Forest University Eye Center Department of Ophthalmology & Visual USA
Winston-Salem NC Sciences
USA University of Illinois at Chicago Leonidas Zografos MD
Chicago IL Professor and Chairman
Richard W Yee MD USA Jules Gonin Eye Hospital
Medical Director LADARVISION Center Lausanne
Hermann Eye Center Charles M Zacks MD Switzerland
Memorial Hermann Hospital Corneal Specialist
Houston TX Maine Eye Center Christopher I Zoumalan MD
USA Portland ME Resident in Ophthalmology
USA Department of Ophthalmology
Steven Yeh MD Stanford University Medical Center
Clinical Fellow Bruce M Zagelbaum MD FACS Stanford CA
Uveitis and Ocular Immunology Associate Clinical Professor of USA
Laboratory of Immunology Ophthalmology
National Eye Institute New York University School of Medicine
National Institute of Health New York NY
Bethesda MD USA
USA
Maryam Zamani MD
Lucy H Y Young MD PhD FACS Oculoplastic Fellow
Associate Professor London
Massachusetts Eye and Ear Infirmary United Kingdom
Harvard Medical School
Boston MA
USA

xliv
SECTION 10 RETINA AND VITREOUS
Edited by Barbara A. Blodi

CHAPTER

122 Functional Anatomy of the Neural Retina


Robert E. Marc

(previously lumped with ACs) that signal over long distances


Overview with intraretinal axons.8 The molecular determinants of signal
This chapter provides an outline of the organization of the amplification, signaling speed, signal integration, and memory
mammalian retina, with a strong focus on primate vision and in systems associated with synaptic transfers will be reviewed in
the context of a nearly complete cellular catalog and extensive the context of cell-specific associations and canonical (main)
new understanding of the molecular diversity of signaling pathways that encode and decode them.
pathways. The mammalian neural retina, albeit simplified by
Every species has a subtly different retina reflecting its dis-
evolutionary losses in cone-driven pathways, is proving to be
more complex than anticipated, with over 60 classes of neurons,
tinctive evolutionary history. Features that are diagnostically
yielding at least 15 and perhaps 20 different ganglion cell ‘filters’ vertebrate, mammalian, or primate will be noted when they
for the visual world. Elucidating the wiring of these filters for any highlight the special attributes of human vision. This chapter
retina remains a major challenge. Though several canonical reviews the functional anatomy of primate and mammalian
signaling pathways have been mapped (including a novel neural retinas in the context of discoveries that have revolu-
scotopic path), we have not been able to reconstruct the likely tionized our understanding of the retinal cells and their associa-
networks of most of the filters. New discoveries regarding the tions (Box 122.1). The details of most of these discoveries are
molecular mechanisms of red and green cone visual pigment beyond the scope of this chapter but the era of purely descriptive
expression have profound implications for the development of neuroanatomy is past and new molecular, physiological, and
color-selective circuits and color vision. A new ganglion cell class
connective contexts accompany descriptions of cell class. New
has intrinsic phototransduction mediated by a novel
photopigment, melanopsin, and is selectively wired to at least technologies have revolutionized cell visualization and analysis,
three different functional pathways. Finally, the wiring of the including cell specific reporter gene expression,9 ballistic
retina is dynamic and manifests significant connectivity changes labeling,10 and more. This knowledge is more than academic, as
both under postnatal visual drive and retinal degenerations. studies of animal models of human disease have revealed new
dynamics in retinal neuroanatomy.11,12 Neurodegenerative
brain disorders and some slow photoreceptor degenerations
INTRODUCTION similarly involve protein misfolding and proteasome stress.13 In
retina, as in brain, this stress induces anomalous neural rewiring.
The retina evolved to report spatiotemporal and chromatic Ultimately, devising interventions that ameliorate vision
patterns of photons imaged by the eye.1 The plan of the human impairment in macular degeneration, retinitis pigmentosa (RP),
neurosensory retina is generically vertebrate in form and glaucoma, or vasculopathies will require in-depth understand-
development, with key specializations. Photoreceptors form a ing of the molecular nuances of contact specificity, signaling,
discrete photon capture screen roughly similar in scale to a cell form, and cell patterning in the retina.
high-end color imaging chip (Fig. 122.1, panels 1–3).2
Vertebrate retinas contain rods, cones, bipolar cells (BCs), THE EVOLUTIONARY CONTEXT
horizontal cells (HCs), amacrine cells (ACs), association cells
(AxCs), interplexiform cells (IPCs), and ganglion cells (GCs).
Cones, BCs, and HCs connect to each other in the outer
INTRODUCTION
plexiform layer (OPL). BCs, ACs, AxCs, and GCs connect in the Molecular biology has added new rigor to comparative biology.
inner plexiform layer (IPL). It is now clear that the mammalian retina reflects a major
The basic vertical channel is the cone ˜ BC ˜ GC chain. evolutionary reduction in neuronal diversity and a simpler
Vertical channel signals are encoded by vesicular glutamate structure than those of most other vertebrates. The genetics of
release (Fig. 122.2) and decoded by ionotropic or metabotropic that reduction is linked to an array of inherited eye defects.
glutamate receptors (iGluRs, mGluRs) expressed by target Every distinctive mammalian feature has been shaped by an
neurons.3–5 Lateral channels mediate signal comparisons evolutionary bottleneck that occurred over 200 million years
over time, space, or color via feedback and feedforward (MY) ago in the late Triassic/early Jurassic as stem radiations of
signaling. Lateral channels are numerous and include cone ˜ therapsid reptiles gave rise to early mammals (Fig. 122.3). This
HC ˜ target cell transfers in the OPL and many BC ˜ AC ˜ included the collapse of the ancestral mammalian axial
target cell transfers in the IPL. AC signals are largely encoded skeleton, cranium, and visual system.
by vesicular g-aminobutyric acid (GABA) or glycine release This sequence from early amniotes to mammals is the most
(Fig. 122.2) and decoded by cognate receptors expressed on fully documented of the major transitions in vertebrate
target cells.3,6 HC signaling mechanisms remain in debate.7 evolution. The entire skeleton was modified, as was the soft
Signaling in the IPL is also mediated by specialized AxCs anatomy, behavior, and physiology down to the level of cellular 1565
RETINA AND VITREOUS

BOX 122.1 Advances in retinal anatomy in the past


decade
• The assembly of a nearly complete cellular catalog
• The assembly of a primitive catalog of transcriptional
regulators of cell phenotype
• New connectivity implications of primate VP gene evolution
• Neuronal phototransduction in melanopsin-expressing GCs
• Neural plasticity in the developing and mature retina
• Circuitry remodeling in retinal disease
• Extensive molecular understanding of the diversity, function
and cellular dispositions of
• glutamate-gated AMPA, KA, NMDA, and mGluR
receptors
• GABA-activated and glycine-activated receptors
• glutamate, GABA, glycine, and anion transporters
SECTION 3

• calcium channels, CNG channels, other ion channels


• connexins
• Resources of particular value
• Oyster. The Human Eye. 1999 Raven Press. A wonderful,
readable and comprehensive book on functional anatomy
of the human eye
• The Visual Neurosciences Vols 1 and 2 (Chalupa and
Werner, eds), MIT Press. 2004 A detailed treatment of
visual system signaling structures and mechanisms
• Webvision: http://webvision.med.utah.edu. A
comprehensive and dynamic resource.

FIGURE 122.1. Cellular elements of the parafoveal primate retina


(vertical plane, left panel; serial horizontal planes 1–7 at right). metabolism. Many of these changes are demonstrated, either
Schematics of cells are layered onto a computationally enhanced
directly or indirectly, through the fossil record.14
toluidine blue thin section of baboon retina (http://prometheus.med.
utah.edu/imagery.html) taken at ~10° eccentricity. The vascular supply The adoption of a nocturnal, fossorial, and insectivorous
for the sensory retina distal to the ELM is formed by the choroid (CH) niches by early mammals triggered the loss of over half the
and the choriocapillaris (CC), apposed to the basal surface of the RPE. retinal neuron phenotypes still manifested by extant non-
Black cells in the CH are resident melanocytes; RPE cells contain dark mammalians (Tables 122.1 and 122.2). The dominant visual
melanosomes. The vascular supply for the neural retina proximal to the stream was switched from the color-rich nonmammalian
ELM is formed by capillary nets in the horizontal cell layer (HCL), collothalamic stream (retina ˜ tectum ˜ N. rotundus ˜
amacrine cell layer (ACL) and distal GCL. Capillary lumens are marked ectostriatum) to the largely achromatic lemnothalamic stream
with asterisks. Rod and cone outer segments (ros, cos) form a discrete (retina ˜ lateral geniculate nucleus (LGN) ˜ striate cortex).
layer (OSL) and abut the apical face of the RPE. Rod and cone inner
Further, the widespread epithalamic (pineal and parapineal
segments extend to the OPL where they form their synaptic terminals:
rod spherules and cone pedicles. Cones (tinted red, green, and blue)
parietal) pathways disappeared in mammals as functional photo-
contact sets of ON and OFF BCs and the somas of HCs (e.g., H1 HCs), sensitive systems. The following sections summarize the major
while rods (white) contact only rod BCs and the axon terminals of HCs evolutionary revisions of mammalian and primate retinas.
(H1ATs). Cones distal to the ELM are gently tilted in the periphery
proportional to their displacement from the visual axis. It is thought that
they ‘point’ toward the nodal point of the eye at all eccentricities. The THE REEVOLVED MAMMALIAN ROD CIRCUIT
slight curvature is a defect of tissue processing. Cone BCs send their Mammalian rod circuits represent a complex revision of
axons into the IPL to contact specific classes of cone GCs, while rod ancestral scotopic vision. The primary mammalian rod path-
BCs contact rod-specific ACs. The INL in primate retina can be clearly ways are now well understood as the following stream:4,15–18
divided into discrete HC, BC, MC and AC layers (HCL, BCL, MCL,
ACL). The IPL is segmented into sublayers specific for the output rods ˜ rod BC ˜ rod (AII) AC ˜ cone BCs ˜ cone GCs
synapses of OFF cone BCs (sublayer a), ON cone BCs (sublayer b), and
rod BCs (sublayer c). The supporting glial MCs span the neural retina Remarkably, this pathway loops back into cone pathways after
from the ELM to the inner limiting membrane (ILM), sealing the optic traversing the entire retina. It has no known antecedent in
fiber layer (OFL) and GC layer (GCL) from the vitreous. Right: Seven any other vertebrate group. In most nonmammalians, rods
horizontal plane views of the primate retina viewed as 30 mm µ 30 mm and cones share BCs and scotopic paths to the brain are direct:
patches. Panel 1: cone myoids in the foveola at the level ELM. Panel 2: rods ˜ BCs ˜ GCs˜ CNS.19 The mammalian rod AC must
cone and rod myoids at ~10° in the periphery with a Bayer-filter overlay have developed from an extant but still unknown non-
(see text). Panel 3: cone and rod myoids at ~10° in the periphery. Panel mammalian sister cell. The molecular genetics of rod pathway
4: Rod spherules (cyan) interspersed with cone axons (red–yellow).
development,20–22 especially comparative patterns of transcrip-
Panel 5: Cone pedicles (red–yellow) surrounded by processes of rod BC
and HC axon processes. Panel 6: HCL. H1 HCs (yellow); H2 HCs (bright
tional regulation and growth factor signaling across vertebrates,
green); rod BCs (blue, r); MCs (black, m). Panel 7: GCL. Midget GCs is likely the key to discovering how the rod circuit lost direct
(Blue, M), parasol GCs (Cyan, P), starburst ACs (s, yellow). All images access to GCs. That knowledge may be central in learning how
1566 are at the same scale and each square is 30 mm wide. to repair wiring anomalies in retinal degenerations.
© REM 2006.
Functional Anatomy of the Neural Retina

FIGURE 122.2. Visualization of GABA (red), glycine (green), and glutamate (blue) signals in the primate retina. Glutamate signals are enriched in
rods, cones, BCs, and GCs. Glycine signals are preferentially enriched in a subset of ACs with sparse terminals. GABA signals are expressed by
a large subset of ACs with widely distributed terminals. Mixed GABA–glutamate magenta signals are expressed by a subset of BCs in some
primates. MCs express little or no glutamate, GABA or glycine and appear black. Each of these signatures can be resolved in the IPL as synaptic
terminals or processes from each of these cell classes (inset). The image is 0.9 mm wide.
© REM 2005.

CHAPTER 122
Paleozoic Mesozoic Cenozoic
C O S D Cm Cp P Tr J K T
Group Hyperoartia
Lampreys

Group Chondrichthyes Sharks


Skates & Rays
Ratfishes
Sturgeons
Teleosts
Group Osteichthyes Lungfishes
Frogs & Toads
Caecelians
Group Amphibia Salamanders
Turtles
Lizards
Group Reptilia Order Serpentes Snakes
Crocodilians
Group Dinosaura
Class Aves
Avians
Order Primates
Group Mammalia
Mammals
590 505 438 408 360 320 286 248 144 65 0 MY BP

FIGURE 122.3. Spectral mixtures of rods and cones mapped on the evolution of the major vertebrate taxa. Solid lines indicate a continuous
fossil record; dashes indicate gaps. Cone LWS, cone SWS1 and rod RH1 VPs (see text) evolved early and were likely expressed by Cambrian
ancestors of the lampreys (Group Hyperoartia, yellow cone, blue cone, and green rod icons). These pigments were extensively diversified during
the evolution of the Osteichthyes to include LWS cones (red icons), RH2 cones (green icons), SWS1 cones (blue icons), and SWS2 UV cones
(violet icons), as well as rods. This diversity persists in modern descendent groups (Amphibia, Reptilia, Aves). With the evolution of mammals at
200–240 MY BP (million years before present, arrow), the RH2 and SWS2 systems were lost. Roughly 20 MY BP (arrow), trichromatic primates
evolved a new green pigment from the LWS system. C, Cambrian; O, Ordivician; S, Silurian; D, Devonian; Cm, Carboniferous Mississippian; Cp,
Carboniferous Pennsylvanian; P, Permian; Tr, Triassic; J, Jurassic; K, Cretaceous; T, Tertiary.
© REM 2004.

TABLE 122.1. Retinal Attributes of the 7 Vertebrate Classes and Primates

Cl Cl Cl Cl Cl Cl Cl Order
Hyperoartia Chondrichthyes Osteichthyes Amphibia Reptilia Aves Mammalia Primates

Divergence MY < 500 450 417 500 500 500 200 200
Cone classes 3 0–2 ? 3–7 4 3–7 3–7 0–2 2–3
Rod classes 1 1 1 1–2 1–2 1 1 1
Rod pathway Direct Direct Direct Direct Direct Direct Indirect Indirect
Neuronal classes ? ? >120 >100 >100 >100 50–60 50–60
Distinct ACs, or foveas 0 0 0–1 0–1 0–1 0–2 0–1 1
Intraretinal vessels — — — — — — + +
Pineal/parietal organs 1 1 1 2 2 1 0 0
Divergence is the time in millions of years (MY) before present that the taxon is clearly identified in the fossil record. Cone classes are defined by combined opsin
expression and structural phenotype. Neuronal classes are identified by morphology.
–, absent; +, present; ?, uncertain or unknown.
1567
RETINA AND VITREOUS

TABLE 122.2. Neuronal Diversity in Selected Mammals Compared with a Teleost Fish

Goldfish Mouse Cat Rabbit Macaque Human

Cone classes 7 2 2 2 3 3
Cone chromatypes 7 2 2 2 2 2
Cone visual pigments 4 2 2 2 3 3
Horizontal cells 4 1 2 2 3 3
Bipolar cells ~20 ~10 ~12 ~12 ~12 ~12
Amacrine cells >70 ? ? ~30 ? ?
Ganglion cells >20 ? ~20 >15 ? ?

REDUCTION OF THE IMAGE-FORMING CONE spatial patterning, subtleties of shape (in primates) and
COHORT TO TWO TYPES: LWS AND SWS1 connectivity.24–26 Within the past 30–40 MY, a gene duplication
SECTION 3

Old-world primate retinas express three cone classes: red- event resulted in the formation of a tandem head-to-tail array
sensitive, green-sensitive, and blue-sensitive or R, G, B cones of red (lmax ~ 560 nm) and green (lmax ~530 nm) pigment
(Box 122.2). The massive loss of cones and reduction in cone genes on the primate X chromosome.27 Historically, primate red
diversity was the prelude to creating a predominantly nocturnal and green cones have been viewed as the initiators of separate
retina. Some mechanisms must have repressed cone progenitor color channels, with separate connectivities leading to hard-
proliferation and enhanced rod progenitor expansion. Again, wired color-opponent neural assemblies at the GC level. This
this history is not merely of academic interest, since defects in deterministic model is now in doubt; a stochastic process likely
these genes are associated with eye degenerations. In most controls whether a cone stably expresses green or red pigment.28
nonmammalians (>95% of all vertebrate species), color-coding Thus it is difficult to see how pathway-specific genes could be
pathways are constructed from 3 to 6 structurally distinct specifically coupled to a green or red phenotype. All mammals
cone types predominantly expressing one of 3–4 cone visual may express only two image-forming cone chromatypes and
pigment (VP) genes,23 as well as many distinctive neurons that this would reduce the required diversity of retinal neurons and
selectively contact those classes. Each cone is a member of a the number of neuron types.
chromatype: a phenotype complex that includes genes for The full set of cone chromatypes may not yet be in hand.
cone shape, patterning, and connectivity in addition to VP Recently, a sparse, orderly population of novel cones has been
expression. It would not be surprising if red and green cone described in mouse retina that exclusively expresses a vesicular
phenotypes in nonmammalians differed in expressions of tens- glutamate transporter type vGlut2.29 Further, a small set of
to-hundreds of genes. In contrast, primate red and green cone human cones has been shown to express melanopsin, a novel
phenotypes may differ in only one gene: the VP. photosensitive pigment.30
Mammals, including primates, express cone VPs derived
from two primordial gene groups (long-wave (LWS) and short-
wave (SWS1) systems) yielding retinas with two cone chroma- REDUCTION OF NEURONAL DIVERSITY
types: green-yellow and violet-blue absorbing. SWS1 cones Over half of the retinal neuron classes expressed by non-
differ from LWS cones in timing of developmental emergence, mammalians have been lost in the mammalian retina
(Table 122.1), including many cone-driven HC, BC, AC, and
GCs. In addition to expressing more than three cone chroma-
types, some nonmammalians possess as many as four HC,
BOX 122.2 Issues in naming cones >20 BC and >70 AC classes (Table 122.2). Further, there are
• Two schemes have been used: RGB (red, green, blue) and likely at least four classes of IPCs in teleost fishes31 and none
LMS (long-, mid-, and short-wave) of their homologs have been identified in mammals. Most
• The LMS system denotes a cone’s relative spectral peak to teleost IPCs evolved fairly recently; i.e., they are apomorphic.
avoid confusion between perceptual names (red, green, blue) However, glycinergic IPCs are expressed by pre- and post-
and VP absorption peaks (yellow-green, green, violet) osteichthyan vertebrates and are thus ancient.31 But they are
• The LMS system is adequate for mammals, but is awkward absent in mammals. While tools to precisely classify all
for nonmammalian species that express four cone VPs and neuronal classes are still emerging, it is clear that the
up to seven chromatypes mammalian retina represents a reduced set of antecedent
neuron classes that have persisted in other taxa.
• A rich tradition of primate CNS physiology exploits the RGB
terminology
• The LMS system can be confused with VP gene groups: APPARENT LOSS OF NEURAL RENEWAL
primate L and M cones are group LWS cones; fish L and M MECHANISMS
cones are group LWS and Rh2 cones, respectively One of the more remarkable features of many nonmammalian
• Perceptual channels initiated by RGB cones do match color retinas is the persistence of neuronal progenitor cells in the eye
names well throughout life, near the retina and perhaps in the retina. In
• Increment threshold spectral sensitivities of trichromatic fishes, rod progenitor cells are known to be able to migrate far
primates show three peaks: 610 nm (orange-red), 535 nm from the progenitor-rich ciliary marginal zone, a torus some
(green), and 430 nm (blue), each primarily driven by R, G, or 10–20 cells thick interposed between the termination of the
B cones retina and the ciliary epithelium.32 The amphibian marginal
1568 zone is similar and some urodele amphibians can regenerate
Functional Anatomy of the Neural Retina

an entire retina at any point in life. There is also evidence contain four dense capillary nets; two bordering the inner
that new photoreceptors and retinal neurons can replace nuclear layer (INL) and two bordering the ganglion cell layer
damaged patches in mature fish retinas and that glial Müller (GCL).1 The retina is functionally partitioned into outer sensory
cells (MCs) can proliferate and may even become neuroprog- and inner neural layers. The sensory layer is composed of rod
enitor cells. The mammalian retina lacks these robust pro- and cone photoreceptors, surrounded by distal MC processes.
cesses, as far as is known though small clusters of potential Similar to other high-gain sensory systems, the sensory layer is
neuroprogenitor cells have been found at the mammalian separated by the external limiting membrane (ELM) into
retinal margin.33 ionically distinct distal and proximal extracellular compartments.
The ELM is a precise border of macromolecule-impermeant
intermediate-junctions between MCs and photoreceptors (Fig.
NEW RETINAL ENERGETICS AND 122.1). The ELM and the basolateral tight junctions of the RPE
VASCULARIZATION delimit the subretinal space; a dynamic regime transited by high
The success of the mammals was largely due to the emergence fluxes of water, oxygen, bicarbonate, protons, inorganic ions,
of a lightweight vascularized skeleton, a more space-efficient sugars, amino acids, osmolytes, and retinoids. The part of the
cortical expansion, endothermy, and improved respiratory and MC proximal to the ELM is responsible for regulating a similar
vascular systems.14 The mammalian retina is the only truly array of moieties in the neural retina, in addition to critical
vascularized retina.1 However, this bias toward high mito- recycling of carbon skeletons derived from synaptic overflow of
chondrial energy supply engages several risks. The first is poor glutamate and GABA.3 MCs ensheath the entire neural retina,

CHAPTER 122
tolerance of low oxygen tensions. Aquatic species, even those comprising ~30% of the INL in the primate central retina and
that demand high oxygen levels (e.g., salmonids), are also up to 50% in the far periphery.35 This vertical view masks the
remarkably resistant to both hypoxia (a common event in fresh elegant spatial tiling of the retina. At the level of the ELM in the
water environments) and hyperoxia.34 The neural retinas of foveola, an array of ~160 000/mm2 cone myoids sample visual
turtles and tortoises are thick, contain perhaps triple the space with apertures of <2 mm each (Fig. 122.1, panel 1). In the
number of neuronal classes as mammals, have complex near periphery, where cone density drops to ~9000/mm2
chromatic processing and high-acuity fovea-like specializations, (Fig. 122.1, panels 1 and 3) large cone apertures are evenly
but are robust in hypoxic settings. Second, the mammalian distributed in a matrix of rods that have nearly the same density
dependence on high-speed blood gas and metabolite transport as foveolar cones, i.e., 150 000/mm2. The same field is also
via fine capillary networks in the OPL and IPL entails shown with a scale overlay of a Bayer-style filter2 for color-
significant light scattering and demands that MCs play an imaging chips, with a conventional 7.5 mm pixel size (Fig.
active role in retinal homeostasis by investing the endothelial 122.1, panel 2). This fine screen of rod and cone apertures is
layer in the same manner as protoplasmic astrocytes in brain.1 remapped into separate arrays of rod and cone synaptic
Finally, the unique vascularization of the mammalian retina terminals (Fig. 122.1, panels 4 and 5). Because rod spherules are
also exposes it to the danger of renewed angiogenesis. so large (>2 mm diameter), they cannot be packed into a single
plane and, at this eccentricity, are stacked four deep, just distal
THE FUNDAMENTALS OF RETINAL to the layer. Some extracellular matrix mechanism may control
STRUCTURE this precise lamination. Foveolar cones have long axons (fibers
of Henle) that spray out like an aster from the central foveola
and array their pedicles into a ring packed edge-to-edge in a
THE LAYOUT OF THE NEURAL RETINA single tile layer. As the cone density drops, the tile spacing
The retina is designed to do two things; sample the torrent of increases and in the periphery the cone pedicle mosaic roughly
photons in the retinal image plane and edit the neural signals maps onto the myoid mosaic. Just beneath the pedicles, diverse
produced by photoreceptors into several sets of filtered neural classes of HCs and BCs form rough patterns to cover image
images.1 This requires a large number of sensory cells, neurons, space (Fig. 122.1, panel 6), ultimately converging on a complex
glia, and supporting epithelia and vasculature. Like most brain tiling of GCs (Fig. 122.1, panel 7).
nuclei, the retina is a lattice of neurons framed by glia and sealed
from its supporting vasculature (Fig. 122.1). At the outer (distal,
sclerad) retinal margin (Box 122.3), the retinal pigmented BASIC NEURONAL PHENOTYPES
epithelium (RPE) forms coupled, high-resistance basolateral There are two major retinal neuron phenotypes; sensory neurons
barrier between the endothelia of the choroicapillaris and the and multipolar neurons. The sensory neuron phenotype
outer segments of the photoreceptors next to the RPE apical includes rods, cones, and BCs (Fig. 122.1), all of which display
face. At the inner (proximal, vitread) retinal margin, the end feet essential attributes of a polarized epithelium.36 Cells of the
of MCs and sparse astrocytes in the optic fiber layer form an sensory phenotype possess the following distinctive features:
intermediate junction seal between neural retina and vitreous. • Ciliary apical and secretory basal specializations
Between these two seals, vascularized mammalian retinas characteristic of polarized epithelium
• Apical poles specialized as photoreceptor outer
segments or BC dendrites
BOX 122.3 Glossary of orientations • G-protein coupled receptor-mediated transduction (e.g.,
Term Reference Object Use photoreceptors and ON-center BCs)
Sclerad ˜ Vitread Layers of the eye, Global location in • Basal poles specialized as a single axon terminating in a
outside ˜ inside the eye secretory synaptic ending
Outer ˜ Inner Layers of the eye, Fine tissue layering • Synaptic release driven by high-capacity ribbon-assisted
outside ˜ inside vesicle fusion
Distal ˜ Proximal Distance from the Position along a • A glutamatergic phenotype
CNS, far˜ near neural chain
Vertical ˜ Image plane, normal Histologic plane of The multipolar neuron phenotype includes ACs, AxCs, and
Horizontal ˜ parallel view GCs and these display classic projection or local circuit neuron
features: 1569
RETINA AND VITREOUS

• Ovoid somas with weak apical–basal polarization


• One to many primary dendrites arising from one BOX 122.4 Encoding molecules and decoding
hemisphere of the cell molecules
• One or more classical axons in many cell types Many different small transmitter molecules are used to encode
• Both axons and dendrites can form many synaptic sites retinal signals:
• Synaptic release driven by low-capacity conventional • Glutamate is used by photoreceptors and BCs for fast
vesicle fusion vertical channel signaling
• ACs tend to be GABAergic or glycinergic • GABA is used by AC subsets for fast lateral inhibition,
• GCs are predominantly glutamatergic usually within IPL strata
• Glycine is used by AC subsets for fast lateral inhibition,
usually across IPL strata
HCs have no clear homology to either bipolar or multipolar • Acetylcholine (ACh) is used by starburst ACs for fast
cells and been provisionally designated their own phenotype. lateral excitation within IPL strata
Though HCs appear multipolar, express iGuRs, and respond • Dopamine (DA) is used by large AxCs for slow
directly to photoreceptors, they have a number of anomalous modulation events
features: • Nitric oxide is used by many cells and certain AxCs for
• Few or no defined presynaptic sites slow modulation events
• Unresolved synaptic signaling mechanisms • Peptides are produced by both ACs and AxCs for
SECTION 3

• Nonspiking axons that appear longer than their cable modulatory signaling
space constant • Melatonin is produced by photoreceptors in a diurnal
• Direct contact with capillaries pattern
• Unresolved neurotransmitter phenotypes Different macromolecules are used to decode retinal signals and
• Intermediate filament expression characteristic of glia. most cells express more than one:
• All cells express ionotropic (iGluR) and/or metabotropic
(mGluR) glutamate receptors
SIGNALING MECHANISMS • mGluR6 receptors are fast, high-gain, and sign-inverting
Neurons encode their voltage signals as changing rates of group C mGluRs (ON BCs)
neurotransmitter release and decode incoming neurotrans- • AMPA receptors are fast, medium-gain, and sign-
mitter signals via transmembrane receptors. Neurotransmitter conserving iGluRs (OFF BCs, HCs, ACs, GCs)
systems of the vertebrate retina have been extensively • KA receptors fast, high-gain, and sign-conserving iGluRs
reviewed3,6,37 and will be summarized only briefly. Many types (OFF BCs)
of vertebrate sensory neurons use ribbon synapses37 for high • NMDA receptors are slow, low-gain, and sign-conserving
rates of tonic release, while multipolar neurons use conven- iGluRs (some ACs, most GCs)
tional synapses.37 Photoreceptors and BCs are glutamatergic • GABAA ionotropic receptors are fast, low-gain, and sign-
neurons and use ribbon synapses for release. Each photo- inverting (BCs, ACs, GCs)
receptor uses a single specialized presynaptic terminal • GABAB metabotropic receptors are slow, high-gain, and
containing thousands of vesicles and ribbon-associated vesicle sign-inverting (mostly BCs)
fusion sites that mobilize a smaller releasable pool of vesicles.18 • GABAC ionotropic receptors are slow, high-gain, and
Rods contain one or two ribbon sites38 and large primate cones usually sign-inverting (mostly BCs)
contain ~20–50. BCs use branched axon terminals that contain • Gly ionotropic receptors are fast, low-gain, and sign-
similarly large numbers of ribbon synapses. The ribbon is a inverting (BCs, ACs, GCs)
mechanism for collecting vesicles at high density near the • nACh receptors are fast, high-gain, and sign-conserving
membrane fusion active zone and such synapses are capable of (GC subsets)
sustained fusion at 500–2000 vesicles/s.39,40 In general, ACs are • mAch receptors are slow, high-gain, and usually sign-
either GABAergic or glycinergic neurons and use conventional inverting (AC subsets)
synapses for release. Each AC cell contains hundreds of small • DA1 receptors are very slow, low-gain, and usually
presynaptic assemblies ranging in size from 10 to 1000 vesicles increase cAMP levels in many cells
and specialized for low release rates (20–100 vesicles/s).41 GCs • DA2 receptors are very slow, high-gain, and usually
are generally glutamatergic, project to central targets and use decrease cAMP levels in many cells
conventional synapses.1,3 The diversity of coding and signaling • Nitric oxide activates soluble guanyl cyclase and raises
mechanisms is summarized in Boxes 122.4 and 122.5. We are cGMP levels in many cells
still untangling the molecular mechanisms of these processes.
For example, four different glutamate subunit genes (GluR1–4)
produce many posttranslationally modified proteins that can
apparently associate in any stoichiometry, generating AMPA filtered versions of the visual world. Each of these chains
receptors with different glutamate affinities, conductances, represents a distinct class of sampling unit with biases toward
kinetics, and co-protein associations.3 Box 122.5 summarizes various stimulus qualities.
the ionic mechanisms mediated by the major signaling Each retinal neuron collects photoreceptor signals from
pathways in the retina. specific synaptic chains. GCs collect direct signals from cone
˜ BC ˜ GC vertical channels and indirect signals that pass
through lateral channels containing HC or AC elements
THE NEURON SET AND ITS CONNECTIVITY (Fig. 122.4). This collection of vertical and lateral signals is
Box 122.6 summarizes the neuronal classes in the mammalian combined by the target neuron to form its receptive field: a
retina. Signals from rods and cones diverge to at least 10 response waveform Í stimulus map. In the simplest receptive
distinct BC classes (4) and thence into ~15 GC42,43 and ~30 fields, a patch of light generates a single response polarity, such
AC classes.44 Ultimately, photoreceptor signals drive at least as cone responses to small spots. Most BCs and GCs possess
1570 15 distinct synaptic chains of neurons representing different concentric center–surround receptive fields,45 where vertical
Functional Anatomy of the Neural Retina

BOX 122.5 Five major signaling mechanisms BOX 122.6 Retinal neuron phenotypes
• Metabotropic photoreceptor >i ON BC signaling 1. The Sensory Neuron Phenotype (bipolar shape, ribbon-
Glutamate released by photoreceptors binds to mGluR6 and based vesicle fusion)
initiates a G protein signal cascade thought to lead to the 1.1. Photoreceptors (VP family = class)
closure of nonselective cation channels permeant to Na+, K+, 1.1.1. Red/Green Cones (Stochastic LWS VP530 or
Ca2+, and Mg2+. As photoreceptors hyperpolarize, LWS VP560, 1 class)
intrasynaptic glutamate levels drop and unbound mGluR6 1.1.2. Blue Cones (SWS1 VP 410, 1 class)
becomes permissive of cation channel opening, increasing 1.1.3. Rods (RH1 VP499, 1 class)
Dg and depolarizing ON BCs through an inward cation 1.2. Bipolar Cells
current. 1.2.1. ON Bipolar Cells (mGluR6 expression)
1.2.1.1. Rod ON Bipolar Cells (1 class)
• Ionotropic photoreceptor > HC and OFF BC signaling, BCs
1.2.1.2. Blue Cone ON Bipolar Cells (1 class)
> AC and GC signaling
1.2.1.3. RGB Cone ON Bipolar Cells
Glutamate released by photoreceptors or BCs binds to
1.2.1.3.1. Midget ON Bipolar Cells
iGluRs and initiates the opening of nonselective cation
(1 class)
channels, increasing Dg and depolarizing target cells through
1.2.1.3.2. Diffuse ON Bipolar Cells
inward cation currents. As photoreceptors or BCs
(~3 or more classes)

CHAPTER 122
hyperpolarize, intrasynaptic glutamate levels drop, unbound
1.2.2. OFF Bipolar Cells (iGluR expression)
iGluRs gate cation channel closure and hyperpolarize target
1.2.2.1. RG or RGB Cone OFF Bipolar Cells
neurons.
1.2.2.2. Midget OFF Bipolar Cells (1 class)
• Ionotropic AC >i BC, AC, and GC signaling
1.2.2.3. Diffuse OFF Bipolar Cells (~3 or
GABA or glycine release from ACs binds to ionotropic GABA
more classes)
or glycine receptors and initiates the opening of a
2. The Multipolar Neuron Phenotype
nonselective anion channel (Cl– is the prime permeant),
2.1. Projection Cells (axon-bearing, spiking)
increasing Dg and hyperpolarizing target cells through inward
2.1.1. Ganglion Cells (>15 classes)
anion currents. When these currents are large, they constitute
2.1.2. Association Cells (>6 classes)
hyperpolarizing inhibition. Unlike cation conductances, anion
2.2. Local Circuit Neurons
conductances often operate near the chloride equilibrium
2.2.1. Lateral Amacrine Cells (mostly GABAergic,
potential and polarization changes may be small. Even so, Dg
20–25 classes)
may be large, constituting shunting inhibition.
2.2.2. Vertical Amacrine Cells (mostly Glycinergic,
• Metabotropic AC > BC and GC signaling 5–10 classes)
GABA release from ACs binds to metabotropic GABA 3. The Horizontal Cell Phenotype (2–3 classes)
receptors and initiates a G protein signal cascade thought to
lead to (1) a desensitization of the voltage sensitivity of BC
synaptic Ca2+ channels, depressing glutamate release or (2)
opening of voltage-gate K+ channels, increasing Dg and
Rod and cone photoreceptors are complex, polarized sensory
hyperpolarizing target cells through outward K+ currents.
neurons (Fig. 122.6) whose structures and biologies are detailed
• Gap junction signaling in the following chapter. Peripheral primate rods and cones
Gap junctions composed of connexin rafts permit direct sign- are cylindrical cells ~70–80 mm long, each possessing a sensory
conserving current flow between pairs of identical outer segment ~10–30 mm long (species-dependent) and a
(homocellular coupling) or different (heterocellular coupling) larger, neuron-like inner segment. Rods and cones exhibit sig-
neuron classes. Slow signals (dopamine, NO) can modulate nificant functional similarity to fiber optics and ‘guide’ captured
connexin conductance. photons. Primate rod outer segments and the optically active
inner segment portion have a diameter of ~1.5 mm. Large
diurnal or crepuscular mammals such as primates have large
cones with optically active inner segment diameters of
channels drive a center response and lateral channels generate ~5–7 mm, yielding a large photon-capture cross section. In con-
annular surrounds of opposite polarity (Fig. 122.5). More trast, both rods and cones of mouse are small and ~1–2 mm in
complex fields encode time-dependent events. optical diameter. The outer segment is an expansion of the
membrane enclosing single nonmotile cilium to form hundreds
to thousands of rod disks or cone lamellae specialized for
PHOTORECEPTORS – STAGE 1 OF THE photon capture and signal transduction. The cilium provides
VERTICAL CHANNEL microtubule-based bidirectional transport of cytosolic proteins.
The primate retina possesses three image-forming photoreceptor The dominant membrane proteins, opsins, are delivered to
chromatypes. nascent discs and lamellae via a targeted vesicle fusion pathway
• Rods express the RH1 opsin group VP 499, have a surrounding the cilium. Just proximal to the cilium is the
unique rod structural phenotype, and selectively ellipsoid region of the photoreceptor; an array of longitudinally
contact rod BCs and HC axon terminals. oriented mitochondria providing high rates of ATP production
• Blue cones express the SWS1 opsin group VP 420, have a for visual transduction and Na+/K+ ATPase in the inner
subtle but unique blue cone morphology, contact cone segment. Primate cone ellipsoids are ~6–8 mm in diameter and
BCs and HCs, with a strong preference for a blue ~12 mm long (a gross volume of ~340 fL), precisely positioned
ON BC. in a single band centered ~8 mm distal to the ELM in peripheral
• Red and green cones express VP 530 or VP 560 from retina. Primate rod ellipsoids are 20-fold smaller in volume.
the LWS opsin group, are structurally indistinguishable In contrast, both mouse rod and cone ellipsoids are small
from each other, contact cone BCs and HCs, but avoid and similar to primate rods.46 This is an important neuro-
the blue ON BC. anatomical distinction. First, the energetics of mouse cones are 1571
SECTION 3 RETINA AND VITREOUS

FIGURE 122.4. The basic cone-driven elements of the retina. Vertical


channels are formed by cone ˜ BC ˜ GC chains. Cones (C) are FIGURE 122.5. GC center-surround receptive fields. GCs at the left
presynaptic (arrows) to ON BCs via sign-inverting (䊊) mGluR6 view a brief light pulse (white band, elevated profile) in their receptive
receptors, OFF BCs via sign-conserving (䊉) KA receptors, and HCs field centers. This elicits a hyperpolarizing voltage in illuminated cones
via sign-conserving AMPA receptors. ON BCs and OFF BCs drive and connected OFF BCs and HCs via sign-conserving synapses (䊉);
matched ON ACs/GCs and OFF ACs/GCs via sign-conserving AMPA mGluR6 receptors invert (䊊) the signal in ON BCs, producing a
and NMDA receptors. Surround channels are formed by HCs in the depolarization. ON BCs synaptically depolarize ON GCs and elicit
OPL and ACs in the IPL. HCs are coupled into homocellular networks spiking. Concurrently, OFF BCs synaptically hyperpolarize OFF GCs
by gap junctions (resistor symbol) and engage in feedback onto cones and inhibit spiking. GCs at the right view a brief light pulse in their
and feedforward onto BCs. The mechanisms of these feedback receptive field surrounds. Responses from distant cones propagate
schemes are not well understood (see text), but it has been argued decrementally through the HC layer and reach nonilluminated center
that HC ˜ cone signaling could be ephaptic (ⵧ), rather than cones and their connected BCs. Sign-inverting HC ˜ cone and
transmitter-mediated. GABAergic ACs dominate feedback and HC ˜ OFF BC signals lead to a small depolarization in OFF BCs;
feedforward in the IPL via sign-inverting synapses, usually of complex sign-conserving HC ˜ OFF BC signals lead to a small hyperpolarization
receptor composition. AC ˜ BC signaling is dominated by GABAC in ON BCs. Thus HCs create opponent surrounds in BCs. These are
receptors, while AC ˜ GC and AC ˜ AC signaling are dominated by passed directly to GCs, so that surround light, with a small delay (arrows),
GABAA receptors. excites OFF GCs and inhibits ON GCs. Matched ACs mediate the same
© REM 2006. pattern of surround signals in the IPL. In summary, bright centers and
dark surrounds excite ON center cells, while dark centers and bright
surrounds excite OFF center cells.
© REM 2006.
likely specialized for use in crepuscular contexts, rather than
the high bleach rates of the noon-time savannah. Second, as the
mouse model has become a key tool for understanding human
retinal disease, these energetic differences associated with cone containing many thousands of synaptic vesicles, ~50 or more
size may be relevant to interpreting disease progression. Mouse synaptic ribbons,48 multivesicular bodies, and a larger mito-
cones appear quite susceptible to indirect or bystander killing in chondrial volume than rods. The glutamate signal from each
some retinal degenerations,11,47 while human cones seem much ribbon site is sampled by several cone BC and HC dendrites,
more resistant. and each cone pedicle is thus sampled by hundreds of dendrites
The component of most relevance to this chapter is the from 8 to 12 different cell classes.
photoreceptor synaptic terminal (Fig. 122.7). Mammalian rod There is weak electrical coupling mediated by small gap
synaptic terminals are termed spherules and are spheroids of junctions between the pedicles of neighboring cones, between
~2 mm in diameter. Each rod axon terminates in a single spherules of neighboring rods, and between cone pedicles and
spherule containing thousands of synaptic vesicles, one or two rod spherules, mediated by connexin36.49–52 Rod–cone coupling
synaptic ribbons that serve as vesicle tethering stations next to (Fig. 122.8) allows rod signals to enter the cone pathway at high
the vesicle fusion sites of the spherule presynaptic membrane,40 scotopic levels. Coupling also occurs between pairs of cones.
many multivesicular bodies thought to represent synaptic Evidence from the dichromatic ground squirrel show that LWS
vesicle recycling pathways, and apparently one mitochondrion. green and SWS1 blue cones do not form G–B coupled pairs,
The glutamate signal from each rod spherule’s synaptic vesicle whereas G–G pairs are common.53 Similarly, red and green
release is ‘sampled’ by two to four fine rod BC dendritic cones are coupled extensively in macacque.54
processes and one to four lobular HC axon terminal processes.40
Mammalian cone synaptic terminals are termed pedicles,
and they have the shape of an architectural pediment, with a BCS – STAGE 2 OF THE VERTICAL CHANNEL
2 mm axon at the peak widening to a base width of >5 mm in Primates display at least 10 distinct BC classes (Figs 122.9 and
1572 primates. Each cone axon terminates in a single pedicle 122.10).
Functional Anatomy of the Neural Retina

FIGURE 122.6.
Generic
photoreceptors from
vertebrates with large
rod–cone dimorphisms
(e.g., primates). Broad
outer segments,
ellipsoids and myoids
both a larger entrance
pupil and higher
metabolic and protein
synthesis capacity in
cones compared to
rods. Large cone
nuclei are located in a
layer just proximal to
the ELM, while rod
nuclei are diversely
positioned throughout a b

CHAPTER 122
the ONL. Thick cone
and thin rod axons FIGURE 122.7. Electron micrographs of rod and cone synapses with
terminate in unique HC and BC processes. (a) The invaginating synapse of a rod
synaptic terminal spherule. The rod vesicle fusion apparatus is composed of a synaptic
shapes. Rod spherules ribbon that organizes vesicles for release at active zones located on
contain one or two either side of the dense protuberance known as the synaptic ridge.
synaptic ribbons and Opposite each face of the synaptic ridge are HC axon terminal
contact ~1–2 HCs and processes (H). (b) An invaginating synapse of a cone pedicle
4–5 BCs, while cone containing two lateral HC dendrites (H) and the dendrite of an
pedicles contain ~50 invaginating midget ON BC (IMB). The arrowheads show basal
ribbon and contact contacts between the cone pedicle and the dendrites of flat contact
50–100 BC and HC with OFF BCs. Each vesicle is ~25–30 nm in diameter.
processes. From Fawcett DW: Bloom and Fawcett: a textbook of histology. 11th edn.
© REM 2006. Philadelphia: WB Saunders; 1986.

• Blue cone BCs (ON) contact blue cones and express the
mGluR6 receptor
• Rod BCs (ON) contact only rods and express the
mGluR6 receptor

BC cells effectively copy photoreceptor signals, mix cone


channels, and produce the essential functional dichotomy of all
vertebrate retinas: ON and OFF channels.4 ON BCs respond to
light onset with sustained nonspiking depolarizations,45 display
long axons that extend deep into the proximal half of the IPL
(known as sublamina b) and slender invaginating dendrites that
preferentially terminate near the synaptic ribbons of rod and
cone photoreceptors.18,55–58 OFF BCs respond to light onset with
sustained nonspiking hyperpolarizations, display short axons
that extend into the distal half of the IPL (known as sublamina
a) and either short flat-contact or semi-invaginating dendrites
that preferentially terminate further from the synaptic ribbons
of cone photoreceptors (Figs 122.7 and 122.9).59 In general,
cone BCs are thought not to be part of the direct rod-driven path
• dB1 cone BCs (OFF) may contact all cones and express but there is now evidence that nonprimate OFF cone BCs do
KA/AMPA receptors make sparse OFF-like contacts at rod spherules.18,60 This will be
• dB2 cone BCs (OFF) may contact only red/green cones reviewed in more detail later.
and express KA receptors The final patterns and mechanisms of selectivity among BCs
• dB3 cone BCs (OFF) may contact all cone classes and have not been fully resolved, but we have broad models for most
express AMPA receptors classes (Fig. 122.11). For example, it appears that most OFF
• OFF midget BCs contact red, green, or blue cones and BCs will contact any cone class and midget OFF BCs exist for
express KA/AMPA receptors red, green and blue cones.61 The presence of a midget blue
• dB4 cone BCs (ON) seem to contact all cone classes pathway is puzzling, as chromatic aberration should blur blue
and express mGluR6 receptors targets when optimally focused for long-wave targets. It has
• dB5 cone BCs (ON) seem to contact all cone classes been established that the ground squirrel homolog (squirrel type
and express mGluR6 receptors b3 OFF BC) to the primate dB2 BC selectively avoids contact
• dB6 cone BCs (ON) seem to contact all cone classes with blue cones, making it an LWS cone-driven cell.62 This will
and express mGluR6 receptors be presumed to be true for primates as well. As far as is known,
• ON midget BCs contact either red or green cones and other diffuse cone BCs are not selective and contact all cone
express mGluR6 receptors classes. The exceptions are the blue cone ON BC that has 1573
RETINA AND VITREOUS

FIGURE 122.8. Gap junctions between


photoreceptor pairs in the primate retina. (a) A
cone pedicle and a rod spherule coupled by a
punctate junction (arrow). Inset, At higher
magnification with lanthanum tracer in the
intercellular space the junction is characterized
by a focal apposition of the adjoining
extracellular leaflets of the two plasma
membranes. C, cone; R, rod. (b) Freeze fracture
imaging shows that such focal appositions
represent the fortuitous cross section of a linear
array of connexons. (c and d), A pair of cone
pedicles connected by gap junctions (arrows)
that consist of a circular array of connexons.
Scaling for transmission images: each vesicle is
~25–30 nm in diameter. Scaling for freeze
fracture images: each connexon is ~6.5 nm in
diameter.
From Raviola E, Gilula NB: Gap junctions between
photoreceptor cells in the vertebrate retina. Proc Natl
SECTION 3

Acad Sci USA 1973; 70:1677–1681.

a b

c d

FIGURE 122.9. Topologically simplified vesicle


fusion and glutamate diffusion patterns around
a single cone synaptic ribbon (thick black line in
the gray box). Vesicles loaded with high levels
of glutamate (dark gray) fuse with the cone
membrane (EX) at active zones on either side of
the synaptic ribbon and are recovered from
sites displaced from the ribbon (EN). HCs
expressing sign-conserving (+) AMPA receptors
are likely closest to sites of vesicle fusion. ON
BCs expressing sign-inverting (–) mGluR6
receptors are centered under the ribbon but are
slightly displaced from fusion sites. OFF BCs
expressing sign-conserving (+) KA receptors are
most distant from vesicle fusion sites.
Alternatively, HCs and OFF BCs are closest to
cone glutamate transporters that remove
glutamate from the cleft. The glutamate
released into the cleft is progressively cleared
as it diffuses from the ribbon. Thus OFF BC
receptors likely sense a much lower mean
glutamate level.
© REM 2006.

sparse dendrites and avoids all red and green cones26 and, suggesting that their synaptic terminals represent distinct
apparently, midget ON BCs that avoid blue cones.61 The chromatypes. There is no evidence that any postsynaptic cell
mechanisms that guide selective contacts are unknown and can discriminate red and green cones terminals.
could involve paired-cell adhesion and recognition mechanisms The ultrastructural patterns of contacts are specialized by
or competition for synaptic targets, or both. On balance, it is class. Most ON BCs tend to generate slender invaginating
1574 clear that rods and blue cones are either targeted or avoided, dendritic tips that form a narrow adhesion en passant contacts
Functional Anatomy of the Neural Retina

FIGURE 122.10. Three diffuse BC classes from Rhesus monkey retina,


visualized by the Golgi technique. At left, one of the OFF cone BC
classes (diffuse flat cone bipolar or DFCB) makes flat contacts with
cones in the OPL and has a broadly stratified terminal in sublamina a of
the IPL. At center, an ON rod BC (RB) makes invaginating ribbon
contacts with cones in the OPL and has a varicose terminal in sublamina
c of the IPL. At right, one of the ON cone BC classes (diffuse
invaginating cone bipolar or DICB) generates invaginating ribbon
contacts with cones in the OPL and has a narrowly stratified terminal in
sublamina b of the IPL.
From Mariani AP: a diffuse, invaginating cone bipolar cell in primate retina.
J Comp Neurol 1981; 197:661.

FIGURE 122.11. A summary of ten BC classes


and their connections in the trichromatic primate

CHAPTER 122
retina. (Class 1) Three short OFF midget BCs
terminate in the OFF sublayer. (Class 2) Two
long ON midget BCs terminate in the ON
sublayer. (Classes 3–5) Three diffuse OFF cone
BCs with flat contacts terminate in different
levels of the OFF sublayer. Classes dB1 and
dB3 have narrow terminal stratifications and
may contact all cone classes. Class dB2 has a
broadly stratified terminal and may contact only
R and G cones. (Classes 6–8) Three diffuse ON
cone BCs with invaginating contacts terminate
in different levels of the ON sublayer. (Class 9)
Blue BCs contact B cones with invaginating
contacts and terminate deep in the ON
sublayer. (Class 10) Rod BCs contact rods with
invaginating contacts and have the deepest
terminals in the ON sublayer. The circles
indicate representative cone contact patterns
for dB1, dB2, and blue BCs.
© REM 2006.

with cone membrane and terminate in a so-called central ribbons to facilitate high rates of vesicle release. The ribbons are
position (Fig. 122.7) in a triad of processes near the synaptic generally smaller than rod and cone ribbons and quite
ribbons of photoreceptors.48,56–58 This means that ON BC numerous, with each BC having many small synaptic ridges,
mGluR6 receptors are positioned close to the source of vesicle each targeting a pair of postsynaptic AC dendrites or,
fusion and experience a relatively high mean glutamate level, less frequently, a GC–AC pair. BC synaptic terminals tend to be
modulated by cone voltage-dependent increases and decreases (1) lobular and branched in a local cluster of small telodendria
in release and clearance.18 This is consistent with the modest (midget, rod, and blue BCs) with a lateral spread similar to the
glutamate sensitivity of the mGluR6 receptor.3 Conversely, diameter of the soma or (2) filamentous and branched in a
most OFF BCs tend to generate blunt ‘flat’ dendritic tips that pattern resembling the dendritic arbor in shape and extent.
form wide adhesion contacts with the cone plasmalemma and Thus some BCs provide output to only a few target cells and
terminate in a position distant from the synaptic ribbons of preserve a narrow receptive field structure, while others branch
photoreceptors (Figs 122.7 and 122.9). This means that OFF to reach many targets, facilitating a divergence of signals.
BC iGluR receptors are positioned farther from the source of The axon terminals of BCs are the vertical structuring
vesicle fusion, experience a lower mean glutamate level elements of the IPL. OFF BCs terminate in the distal half
modulated by cone voltage-dependent increases and decreases (sublamina a) and ON BCs in the proximal half (sublamina b)
in release and clearance.3 This is consistent with the relatively (classical refs). The late Brian Boycott pointed out that the deep
high glutamate sensitivity of the KA receptor found on subsets proximal IPL where rod BCs drive rod ACs is structurally
of OFF BCs.63–65 On an intermediate scale, some OFF BCs are unique and he proposed it to be sublayer c.66 Each AC and GC
known to invaginate some dendrites much closer to the ribbon thus sends its dendrites to specific levels of the IPL where the
than those that make flat contacts59 and these invaginating OFF specific BCs are sampled. GCs targeting sublamina a or b are
BCs express AMPA receptors.65 The fluctuations in glutamate respectively dominated by OFF or ON BCs, while those whose
levels are also faster near the ribbon and smoothed at flat dendrites span both sublayers typically show mixed ON–OFF
contacts, suggesting that KA and AMPA receptors represent key behavior.45 It has long been common to separate the IPL into
temporal filters for BCs.65 The complex topology of this five sublayers, though there is no biological basis for the prac-
arrangement of processes is represented in a simplified two- tice. Indeed, various immunochemical markers show that the
dimensional form (Fig. 122.9). mammalian IPL can be segmented into no fewer than seven
BC outputs, like those of photoreceptors, are restricted to sublayers and nonmammalian retinas easily possess up to 15
the axon terminal specialization of the basal pole of the cell sublayers.3 The best practice is to specify the level of the IPL by
(Fig. 122.12). Again, like photoreceptors, BCs utilize synaptic setting the ACL/IPL border at 0 and the GCL/IPL border at 100. 1575
RETINA AND VITREOUS

GCS– STAGE 3 OF THE VERTICAL CHANNEL


Mammals display at least 15 distinct GC classes (Figs 122.13
and 122.14).
The diversity of mammalian retinal GCs has been assessed
by Golgi impregnation, dye labeling, transgene expression and
molecular phenotyping strategies.42,43,67–71 The exact numbers
are not known but clearly exceed 15 and may even exceed
20,68,70 even in primates. Exact homologies have not been estab-
lished across mammals, but there are some basic commonalties
of structure. Nearly all GCs have their somas in the GC layer
proper and exhibit a variety of dendritic patterns, ranging from
compact narrow-field to highly branched wide-field, with
laminar patterns ranging from narrowly stratified to bistratified
and ultimately to diffuse laminations. In the primate retina,
several GC classes have been studied described anatomically
and physiologically.
• Midget GCs (OFF) contact OFF midget BCs
SECTION 3

• Midget GCs (ON) contact ON midget BCs

FIGURE 122.12. Cone BC synaptic terminals in the mammalian IPL.


BC ribbon synapses generally target two postsynaptic processes in
dyad synapse. In this instance, both postsynaptic processes are AC
dendrites, one of which (asterisk) makes a conventional reciprocal
feedback synapse (curved arrow) with the BC. Another AC synapses
on the BC the right margin of the terminal. Rb, synaptic ribbon;
primate retina. Inset upper left, A ribbon synapse at higher FIGURE 122.14. A horizontal plane view of medium-field class b ON
magnification showing the pentalaminar ribbon structure, a halo of center (A, B, C) and OFF center (D, E, F) GCs of cat retina visualized
synaptic vesicles, enlarged synaptic cleft, postsynaptic densities in with the Golgi technique. Cells A and D are near the area centralis,
the target cells (arrowheads). Rabbit retina. Inset lower right, a BC ˜ cells B and E from the near periphery, and cells C and F from the
AC ˜ GC feedforward chain. The curved arrows indicate the direction periphery at 9 mm eccentricity (~40°). All are the same class of cell
of signaling. Primate retina. Scaling for transmission images: each with graded field diameters reflecting changes in cone density. Bar =
vesicle is ~25–30 nm in diameter. 100 mm.
From Raviola E, Raviola G: Structure of the synaptic membranes in the inner From Wässle H, Boycott BB, Illing RB: Morphology and mosaic of on and off
plexiform layer of the retina: a freeze fracture study in monkeys and rabbits. beta cells in the cat retina and some functional considerations. Proc R Soc Lond
J Comp Neurol 1982; 209:233–248. [B] 1981; 212:177–195.

FIGURE 122.13. Primate narrow-field midget,


medium field parasol and wide-field garland
GCs visualized with the Golgi technique and
labeled according to Steven Polyak’s 1941
classification. Two different parasol GCs (P)
send their dendritic arbors to the distal OFF and
proximal ON sublayers of the IPL, as do several
midget GCs (unlabeled). A shrub GC (S)
appears to have a small diffusely bistratified
arbor. A garland GC (G) arborizes widely in the
OFF sublayer. The arrowheads denotes the IPL
borders.
Modified from Polyak SL: The retina. Chicago:
1576 University of Chicago Press; 1941.
Functional Anatomy of the Neural Retina

TABLE 122.3. Provisional Functional and Anatomical Assignments of Mammalian GCs

Physiology MacNeil/Marc/Famiglietti Structural Features, Level Primate Homologs


Class (Coupling), Percent

Concentric Brisk Linear


ON sustained X, b G4 / 3 / IIa, 12.2% Medium monostratified, 35–45 ON midget
OFF sustained X, b G4 / 6 / IIb1 (g), 15% Medium monostratified, 55–65 OFF midget
ON transient G? / 1b Medium monostratified, 35–45 ON parasol?
OFF transient G? / 5 (g) Medium monostratified, 35–45 OFF parasol?
Concentric Brisk Nonlinear
ON transient Y, a G11 / 1a / Ia2, 2.8% Wide monostratified, 60–80 ?
OFF transient Y, a G11 / 9 / Ib2 (g), 1.4% Wide monostratified, 30–40 ?
Concentric Sluggish Linear

CHAPTER 122
ON sluggish sustained G? / 2, 4.2% ?
OFF sluggish sustained G? / 8 (g) , 12.2% ?
Concentric Sluggish Nonlinear
ON + OFF sluggish transient G? / 12 (g), 5.2% ?
Complex
Local edge detector G1 / 7 (gG), 5.6% Narrow monostratified, 50 ?
Uniformity detector G6 / 5 (g) or 1b? Medium monostratified, 80 Melanopsin GC ?
Orientation selective G? / 11 (g), 6.6% ?
ON–OFF DS G7 / 1c & 10 (g) Wide bistratified, 25 and 75 ?
ON DS G10 / 4, 7.7% Medium monostratified, 70–80 ?
Blue ON Green OFF G3 ? / ? Medium bistratified, 20 and 80 Blue ON Yellow OFF

• Large bistratified blue ON GCs contact blue BCs and Narrow-field GCs, such as midget GCs (Fig. 122.13), are
OFF bB2-like BCs monostratified cells with medium-sized somas that selectively
• Small bistratified blue ON GCs contact blue BCs and contact the axon terminals of midget BCs, and are ON or OFF
OFF bB2-like BCs cells.45 Such cells typically generate relatively sustained spiking
• Parasol GCs (OFF) contact diffuse OFF BCs (likely dB3) patterns to a maintained light stimulus. In the foveola each
• Parasol GCs (ON) contact diffuse ON BCs (likely dB4) midget GC contacts a single midget BC but midget GCs may
• Inner giant melanopsin GCs contact diffuse ON BCs contact several midget BCs in peripheral retina.72,73 Midget
(likely dB6) GCs project to the dorsal parvocellular (small cell) P layers of
• Outer giant melanopsin GCs contact unknown cell the LGN that in turn project to layer 4Cb of striate visual cortex
classes area V1.74 Projection neurons along parvocellular retina ˜ LGN
˜ cortex stream appear to be key elements in high-acuity
GC populations in cat and rabbit retina have been analyzed vision, and tend to have sustained responses. Their roles in hue
more comprehensively and, based on several recent studies can discrimination have been debated, but it is likely that the
be parsed into at least a dozen clear structural, molecular, and essential information for hue discrimination is embedded in
physiological categories (Table 122.3 and Table 122.4). their signals.1 As they share some morphological features, func-
tions and projections, primate74 and ground squirrel midget
GCs,75 b cells of the cat retina76 and classes 3 (IIb2) and 6 (IIa)
TABLE 122.4. Optical Sampling Across Species GCs of rabbit42,67 are likely homologs. Species differences in GC
morphology are partly due to variations in cone density and
Species Visual Angle in (mm/deg) patterning. In trichromatic primates, midget cells are thought
Human 280–300 to generate the VP 560-driven and VP 530-driven ON and OFF
sustained GCs that may subserve red–green hue discrimi-
Macaque 246 nation. However, just as midget BCs likely do not discriminate
Cat 220 between red and green cones, parafoveal midget GCs that
collect signals from a few midget BCs do not appear to have any
Rabbit 160–180
color selectivity1,77 and lack color opponency.73
Rat 75 Medium to wide-field GCs (Fig. 122.13) are the most com-
Goldfish 60 mon types in most retinas but likely represent mixed functional
classes. One distinctive class in most species is a medium-
Mouse 31 density to sparse population of large somas with very large
Zebrafish 10 (estimated) dendritic arbors, such as parasol GCs in primates and a or
a-like cells in other mammals. These cells sharply stratify their 1577
RETINA AND VITREOUS

dendrites at specific levels of the IPL, suggesting that they HCS – THE LATERAL CHANNEL FOR
preferentially sample from cells such as OFF dB2/3 BCs78 or ON PHOTORECEPTOR ˜ BC SIGNALING
dB4/5 BCs. Parasol GCs preferentially project to the ventral
magnocellular (large cell) or M layers of the LGN, thence to Every GC is a logical device that compares vertical channel
layer 4Ca of striate visual cortex area V179 and are achromatic signals with those from several lateral channels. There are at
or luminance-driven neurons. In cat and rabbit, such cells show least two formal lateral channel topologies in the OPL:
transient responses; brief bursts of spikes to a step of light at • cone1 ˜ HC ˜ cone1 reciprocal feedback (temporal)
light onset (ON cells) or offset (OFF cells).45 However, the • cone1 ˜ HC ˜ cone2 lateral feedback (spatial, spectral)
presumed parasol GCs of primate retina and most LGN mag- • cone ˜ HC ˜ BC lateral feedforward (temporal,
nocellular neurons are rather linear in their responses79 and spatial, spectral)
may not be true a homologs, or may be a re-derived variant
(e.g., progenitor duplication instead of gene duplication). Large These chains partly shape GC responses that encode contrast,
sparse GCs with even larger dendritic arbors are plausibly the color, and spatial timing of natural stimuli. A general
primate a OFF cell.69,70 engineering principle states that each step of high gain
Bistratified GCs arborize in both sublayers of the IPL, giving (photoreceptor or BC ribbon synapses) requires a stabilizing
them the opportunity to capture signals from BCs with opposite negative feedback element.95 The quantitative differences
polarities. There are several known examples, but the attributes between feedback and feedforward are beyond the scope of this
of none are known particularly well. In cat, ON–OFF transient chapter. HCs are the lateral processing elements of the OPL
SECTION 3

cells resemble bistratified a cells. In rabbit, a set of GCs sample and enable BCs to compare direct light signals captured by
from sublamina a and b near the midline of the IPL and photoreceptors they contact and indirect light signals from
generate independent ON and OFF directionally selective (DS) surrounding photoreceptors they do not. Neither the mech-
responses. ON–OFF DS cells are one of the most complex anisms of HC signaling or the relative strengths of the cone ˜
neurons known and, though their receptive field mechanisms HC ˜ cone lateral feedback and the cone ˜ HC ˜ BC lateral
have been studied intensively, the underlying biological basis of feedforward paths are known though several models exist.3
its tuning remains controversial.80 DS cells have not been Topologically, HCs exhibit two forms in mammals; axon-
studied thoroughly in primates, but the ACs most often bearing and axonless.96–99 All HCs contact cones with their
associated with them are present in primates81 and likely dendritic arbors while axon-bearing HCs form large axon
candidates for both monostratified ON DS and bistratified terminal arbors that contact rods (Fig. 122.15).99 Many
ON–OFF DS cells have recently been described in macaque.70 mammalian species display both, while rodents apparently
The most distinctive primate small-field bistratified cell is the develop only a single class of axon-bearing cells.100 No evidence
blue ON bistratified GC that collects inputs from a diffuse OFF exists to support the idea that the axons of mammalian HCs are
BC (likely dB2) and blue ON BCs. Thus the receptive field electrically functional.101 HCs do not generate action potentials
center of this cell is spectrally biphasic, depolarizing and spiking and the somatic and axon terminal fields respectively generate
to blue light and hyperpolarizing to yellow light.82 cone-selective and rod-selective light responses with no
Melanopsin GCs in primate are ‘giant’ GCs that narrowly evidence of signal mixing that can be attributed to the axon.
stratify at the proximal or distal margins of the primate retina.83 Indeed we might consider an axon-bearing HC to be two
Some giant melanopsin GCs appear misplaced or displaced to separate cells that share a single nucleus. The primate retina is
the ACL. Melanopsin GCs (also known as intrinsically photo- more complex than any other mammal as it likely harbors three
sensitive or ipGCs) are present in other mammals,84–86 project HC classes.101–104
widely in the thalamus and pretectum, and partly drive the • H1 HC somatic dendrites contact all cones and lack
pupillary response.87 In primates, these cells appear to be the axons in the rod-free foveola
rare blue OFF/yellow ON (B–/Y+) GCs of the retina.87 In • H1 HC responses indicate dominance by R and G cone
addition to a full range of rod and cone responses, these cells inputs
express melanopsin; a photosensitive G-protein coupled • H1 HC axon terminals contact rods
receptor (GPCR) that uses 11-cis retinaldehyde as a chromo- • H2 HC somatic dendrites appear to contact all cone
phore.88 At high photopic levels, melanopsin directly drives classes, with a blue cone bias
highly sustained spiking activity, partially compensating for • H2 HC axon terminals appear to contact all cones,
the transient nature of cone-driven responses.83 Melanopsin with a blue cone bias
GCs in many species project to the suprachiasmatic nucleus • H3 HC somatic dendrites contact cones, apparently
(SCN) as part of the circadian clock pathway and to the olivary avoiding blue cones
pretectal complex as part of the pupillary response pathway. In • H3 HC axon terminals contact rods
primates they also project to the LGN.83 Evidence is building for
the existing of several subclasses of melanopsin GCs.86 Most H1 and H2 cells are the main HC classes, and H1 cells are
importantly, these cells likely underlie the persistence of photic four times as abundant as H2 cells.105 H3 HCs have been
entrainment of circadian cycles, even after photoreceptor described only in Golgi preparations and have been difficult
degeneration.89,90 to document. But Golgi studies have rarely been incorrect and
GCs are purely postsynaptic neurons from a neurochemical so the search for the H3 cell by molecular means continues.
perspective, and decode BC signals with a mixture of AMPA The somas of primate HCs and mammalian axon-bearing
and NMDA receptor subtypes.3,42,91,92 AC signals are decoded HCs are ovoid and give rise to 8–12 dendrites that ultimately
with a variety of GABA and glycine receptors.6 GC dendrites contact ~12 cones in the foveola and 20 cones in the periphery.
make no presynaptic contacts and are thought to be entirely The axons contact a few hundred rods in primates and many
postsynaptic. However, some GC classes make heterocellular more in cat. Though primate H1 somas appear to contact
gap junctions with ACs, forming specific GC::AC::GC all cones, they clearly have responses dominated by R and G
syncitia.8,42,93,94 Each retinal GC generates a single unmye- cones, with little B input.106–108 Conversely H2 HCs are
linated axon that becomes myelinated in the optic nerve and enriched in blue responses, reflecting their tendency to contact
projects to one or more CNS targets: LGN, SCN, pretectum, a disproportionate number of blue cones.108 Axon terminals
1578 and superior colliculus. have rod-driven responses.
Functional Anatomy of the Neural Retina

nonmammalians. Physiological studies in nonmammalians


imply that there is no feedback path from HCs to rods but there
is to cones. Paradoxically, vesicles similar to classical synaptic
vesicles are more common in HC dendrites contracting rod
spherules than those contacting cone pedicles,111 but there is no
obvious conventional presynaptic assembly and no evidence of
stimulated fast exocytosis or endocytosis in HC dendrites.
Indeed, similar dendritic accumulations of vesicles in brain
neurons appear to be involved in the regulated cytoskeletal
delivery and turnover of postsynaptic AMPA receptors to
dendrites as vesicular cargo.112 HC dendrites have also been
argued to serve as ephaptic feedback devices via patches of
hemijunctions (arrays of half-connexins) through which
currents leak constitutively.7 In fishes, connexin+ hemi-
junctions are very close to the voltage-sensitive Ca2+ channels
that regulate cone vesicle fusion.113 When the HC layer is
hyperpolarized by closure of AMPA receptor channels, the focal
inward current through hemijunctions makes the local

CHAPTER 122
intrasynaptic potential more negative than the adjacent
intracellular cone potential and this relative depolarization is
perhaps sensed by cone Ca2+ channels which begin to open,
increasing transmitter release briefly. This feedback effect
requires no transmitter-dependent or vesicle mechanism, which
nicely explains why many HCs contain no measurable
inhibitory transmitter, yet apparently function quite effectively.
Finally, HCs somas are strongly coupled to each other by
large dendritic gap junctions, so that synaptic currents gener-
ated by cones spread readily in the HC layer.101 In nonprimates
such as rabbits, axonless HCs are more strongly coupled than
the far more abundant axon-bearing HCs.114,115 This generates
two distinct spatial classes of HCs, though the functional
significance of this dichotomy remains unknown. Coupling
efficacy of axon-bearing primate H1 cells coupling resembles
the weak coupling of rabbit axon-bearing coupling.116 H1 and
H2 cells form separate coupled mosaics.105

ACS – LATERAL CHANNELS FOR BC ˜ GC


SIGNALING
• BC ˜ AC ˜ BC reciprocal lateral feedback
FIGURE 122.15. A horizontal plane view of H1 (HI) and H2 (HII) HCs
from the macaque visualized with the Golgi technique. ax, axon. Bar =
• BC1 ˜ AC ˜ BC2 lateral feedback
15 mm. • BC ˜ AC ˜ GC lateral feedforward
Slightly modified from Kolb H, Mariani A, Gallego A: A second type of horizontal • AC ˜ AC concatenated feedforward chains
cell in the monkey retina. J Comp Neurol 1980; 189:31–44. Copyright © 1994
John Wiley & Sons. ACs are the most diverse group of neurons in the retina,8 with
over 70 classes in teleost fishes117 and over 25 classes in
mammals.44 Most ACs, by definition, lack classical axons and
The processes of both HC somas and axon terminals are function as local circuit neurons via dendritic synapses, usually
dendritic in nature. The dendrites are lobular and flank the as negative feedback and feedforward control elements. While a
synaptic ridges of photoreceptors as lateral elements of a precise tally is still uncertain, about two-third of AC classes
synaptic triad (Fig. 122.7).99 The dendrites are thus close to the appear GABAergic, with the rest glycinergic. Other candidate
sites of photoreceptor vesicle fusion. HCs express AMPA AC transmitters often co-localize with GABA.3 ACs exhibit
receptors.3,91,109,110 but the precise locations of the functional diverse morphologies with lateral spread of dendrites ranging
receptor patches are not certain. Since geometric factors such from narrow (<100 mm) through medium (100–200 mm) to
as the spatial coherence of vesicle release and the distribution wide (>200 mm, with some cells > 1 mm) based on a com-
of glutamate transporters along the diffusion path, the precise prehensive study of rabbit ACs.44 Narrow-field cells such as the
receptor position is a key datum. classic glycinergic rod (AII) AC (Fig. 122.16) typifies many
While HCs are clearly postsynaptic to photoreceptors, it has narrowly stratified, bistratified, and diffusely arborized
been extremely difficult to discover their outputs. This issue has classes.15–17,118,119 Most medium-field cells have diffuse arbors
been treated extensively elsewhere,3,18 but classical presynaptic while most wide-field cells such as GABAergic starburst ACs
specializations are missing in HCs. Presence of GABA in some (Fig. 122.17) are narrowly stratified.120,121 Similarly, the wide-
mammalian HCs has led to the view that they must be field type S1 and S1 GABAergic rod ACs (also known as A17,
GABAergic, though compelling demonstrations of that fact are AI, and indoleamine-accumulating ACs), appear to have very
few. Wilson39 provides a comprehensive and insightful review of diffuse arbors but, in reality have most of their inputs and
these issues. However, there are many mammalian HCs that outputs very narrowly stratified within sublayer c at ~90–100
completely lack any evidence of GABA content. Further, there is level of the IPL.68,122–125 Thus there are two broad signaling
no evidence for transporter-mediated export found in motifs of importance; narrowly stratified and usually 1579
SECTION 3 RETINA AND VITREOUS

FIGURE 122.17 A horizontal plane view of a wide-field starburst AC


from the rabbit retina. Note the regular dichotomous branching of its
dendrites and the concentration of endings at the periphery. Visualized
by single-cell dye injection. The dendritic field is ~140 mm µ 200 mm.
From Tauchi M, Masland RH: The shape and arrangement of the cholinergic
neurons in the rabbit retina. Proc R Soc Lond [B] 1984; 223:101–119.

diffuse or bistratified ACs may have similarly diverse patterns


of inputs and outputs. There is an exception to this complex
lamination. The proximal band at level 90–100 contains the
axon terminals of rod BCs and only the arbors of only three
classes of rod ACs; the narrow-field glycinergic AC and two
wide-field GABAergic rod ACs.44,125 This further supports the
designation of this band of IPL as a unique sublayer.66 One
additional feature tends to correlate with signaling within
versus across levels and that is neurotransmitter class. Many
narrowly stratified cells tend to be GABAergic and AC ˜ BC
reciprocal synapses and the BC > AC >i BC chain are almost
exclusively GABAergic.3 Correspondingly, the density of GABA
synapses is extremely high and accounts for ~90% of the
FIGURE 122.16 A narrow-field rod AC (AII) viewed in vertical (image synaptic innervation in the vertebrate IPL (Fig. 122.2). Several
A) and horizontal planes in the OFF (image B) and ON (image C) diffuse or multistratified cells may mediate BC > gly AC >i GC
sublayers of the IPL in rabbit retina. In the OFF sublayer, tortuous fine or BC signaling across strata. Cholinergic signaling is mediated
branches arise from the cell body and primary dendrite and terminate in mammals by the ON and OFF starburst ACs3,8,80,126 which
as large lobular appendages (asterisks). In ON sublayer, the primary
are also GABAergic neurons.3 These cells are narrowly stratified
dendrite gives rise to a conical tuft of arboreal branches that spread
tangentially at the IPL–GCL border and carry small swellings and form two excitatory output systems at levels 20–25 and
(arrowheads). Image A was visualized with the Golgi technique, and 65–75 of the IPL. Their presumed primary targets are DS GCs
images B and C are optical sections visualized by single cell dye in rabbits80 and DS candidate GCs have been described in
injection. The IPL width is ~25 mm. macaque.81 Expression patterns of AC neurotransmitters are
Slightly modified from Dacheux RF, Raviola E: The rod pathway in the rabbit reviewed in detail by Marc3 and Brecha.37
retina: a depolarizing bipolar and amacrine cell. J Neurosci 1986; 6:331–345.

LARGE-SCALE PATTERNING OF RETINAL


GABAergic ACs provide lateral signals within a set of BCs and CELLS
their targets, while diffuse (often glycinergic) ACs provide lateral Vertebrates with large eyes also show large-scale variations in
signals across different sets of BCs and their targets. As there cell density and composition of the neural retina,1 likely reflec-
are at least twice as many AC classes as BC and GC classes, ting the power of niche selection to control global tissue
each BC should thus drive at least two different kinds of ACs. patterning signals (Fig. 122.18). Many species concentrate
But things are clearly more complex than this, as dendrites cones in retinal regions where the optical quality is high (central
from at least ten different ACs arborize at any level of the IPL foveas in primates) or in horizontal streaks that reflect a strong
between 0 and 90. This emphasizes our nearly complete lack of behavioral bias for horizon-related visual transitions (urodele
understanding of the various inputs and outputs for any AC amphibians, chelonian reptiles, lagomorph mammalians). The
across levels except for one archetypal neuron; the glycinergic biophysics of foveal formation are poorly understood, though
rod (AII) AC. This AC is actually a tristratified cell that receives Springer and Hendrickson127 have argued that increased
rod BC input at level 90–100, makes gap junctions with cone intraocular pressure and growth-induced retinal stretch induce
ON cone BCs at levels 55–80 and forms inhibitory glycinergic the primate foveal pit. While these ideas do not easily explain
1580 synapses on cone OFF BCs at levels 10–35.3,16–18 Many other other patterns, especially the deep convexiclivate foveas of
Functional Anatomy of the Neural Retina

FIGURE 122.19. Sampling of image space by photoreceptors. Cone


density (red), rod density (cyan), and cone coverage (yellow) profiles in
the human retina as a function of retinal eccentricity in the equatorial

CHAPTER 122
plane. Temporal retina is left and nasal is right on the abscissa, with a
gap centered on ~14° eccentricity representing the optic nerve head.
The left ordinate is density data replotted from Curcio129 on a square
root scale. The right ordinate is linear fractional coverage: the fraction
of image space captured by cones. Cone density forms a wide
pedestal at ~5000 cones/mm2 with an extremely steep peak in the
central 2° reaching ~160 000 cones/mm2 in the foveola. Rod density
is a broad profile of 90 000–140 000 rods/mm2 that would also peak
at ~160 000 rods/mm2 were it not for a deep declivity formed by their
displacement in the central 2°. The foveola is rod free. Cone myoid
and ellipsoid diameters increase with eccentricity so that cones never
capture less than ~30% of the available image data.

development of transgenic and knockout avians and advanced


quantitative trait locus analyses may offer new ways to
understand the genes that control foveal formation and global
neuron patterning.
The description of human retinal neuron distributions and
patternings in Oyster1 is without peer and will only be sum-
marized briefly. The relative numbers of rods and cones change
radically across human retina (Fig. 122.19), ranging from
~160 000 cones/mm2 at the foveola to 5000 cones/mm2 at
~20° eccentricity.1,129 Rods show a broad profile ranging
from ~90 000 to 140 000 rods/mm2 with shoulders at
~15–20°. But it is clear from the trend that rods would also
FIGURE 122.18. Large-scale spatial variations in cone density. (1) Most peak in the central retina at ~160 000 rods/mm2 were it not for
rodents display weak central elevations in cone density. (2) Carnivores
the fact that cones displace them. This is about the maximum
have strong concentrations of cones and GCs in the central retina.
(3) Prey animals such as rabbits (Lagomorpha) express distinct linear density possible for any photoreceptor, corresponding to a
bands of high cone, BC, AC, and GC density. (4) Diurnal primates spacing of ~2.5 mm (including MC space at the ELM). As cones
express compact, high-density, pure-cone foveas centered in a large develop early and capture the foveola, rods can encroach only
low-density cone field. (5) Avians express the most complex density from the outer margins, leading to a deep declivity in the rod
variations, with a central pure cone region exceeding primate densities profile and a rod-free foveola. In addition to changing density,
and a second moderately high density region specialized for binocular cone size also changes with eccentricity. While this negatively
vision. (6) Many rodents, such as domestic mice, display pure blue affects acuity, it increases cone photon capture. In fact, the
cone ventral fields. (7) Aquatic reptilians such as turtles (Chelonia) coverage factor (CF) of the cone mosaic (the fraction of the
possess very high-density linear streaks (approaching primate
retinal image captured by cones) never drops much below 0.3
densities) enriched in cones with red oil droplets. (8) Primates are
unique among all vertebrates (as far as is known) in having a tritanopic
outside 15°, and smoothly rises to 100% over the foveola.
fovea where the central 15’ arc contains few or no blue cones and a Remarkably, overall cone density in rodents130 can be much
peak of blue density at 0.3–1° eccentricity. (9) Finally, the bifoveate higher than primate peripheral cone density, reaching ~12 000
avians possess differential enhancements of cones, with high red-oil cones/mm2. But as rodent cones and rods are similar in size and
droplet cones dominating the temporal fovea and yellow oil cones cones comprise only 3% of the photoreceptors, their coverage is
dominating the central fovea and most of the peripheral retina. only 0.03. The significance of this is more obvious when we
© REM 2005. normalize sampling for the relative optical sizes of the eyes
(Table 122.4). An image in peripheral human retina that
subtends a circle 1° in diameter covers a patch of 425 cones,
lizards or the dual and differently shaped foveas of avians that whereas the same 1° image in mouse131 covers a patch of only
may involve both local proliferation and cell migration, they eight cones. The statistical danger of generalizing visual losses
offer testable models. Developing genetic models for studying or recoveries in mouse models of retinal degeneration should be
tissue sculpting in large eyes will be challenging,128 although gauged carefully.11 1581
RETINA AND VITREOUS

Relative acuity loss with eccentricity in the human eye is prevents mature blue cone expression in the foveola also may
partially a consequence of cone density decline and partly the regulate patterning of the rods.
increased receptive field sizes of retinal GCs that drive
perception. Indeed, many neuron classes show large changes in
local density over the eye.1 The foveola itself (the central 20 s FINE-SCALE PATTERNING OF RETINAL CELLS
arc of the retina) is composed solely of cones with all other The retina is an assembly of sampling units that cover the image
neurons and cone axons and pedicles displaced to a ring around plane.1 Over 60 classes of neural elements are patterned across
the foveola. However, the corresponding GC and BC density this plane and, if we treat them as tiles, we find them arranged
profiles roughly track cone density. The cortical magnification in different pattern types,1,138 partly quantified by their CFs.
factor (the disproportionate area of cortex devoted to foveal • Packings have no overlaps: Photoreceptor arrays are
vision), reflects a relatively constant volume of cortex captured packings with CF < 1
per GC axon. Visual acuity does not simply follow either cone • Coverings have no gaps: AC arrays are coverings with
density or the density of midget GCs. A mixture of GC CF > 1
sampling units exists at any retinal position. GCs with small • Tilings have no overlaps or gaps: BC and GC arrays
receptive fields can resolve smaller targets, but are not as light resemble tilings with CF ~1
sensitive as those with larger receptive fields. Thus supra- • Mosaics are general patterns of any type, with tile
threshold acuity depends highly on image contrast and mean subtypes.
luminance. Multiple classes of sampling units likely participate Cone arrays are mosaics of three tile subtypes (R, G, B) with
SECTION 3

in setting visual performance. CFs < 1 (Fig. 122.20). CFs for complex, branched cells such
Large-scale chromatic patterning also accompanies these as HCs, ACs, or GCs can be more precisely defined as CF =
variations in cone density. Many species show variations in A µ D, where A is the dendritic or receptive field area (defined
the distributions of blue cones, with many mammals (some as its convex hull or Voronoi domain) of a single cell in a given
rodents and lagomorphs), exhibiting ventral (inferior) fields class and D is the density of cells per unit retinal area. Thus, an
entirely composed of or enriched in blue cones.132–134 No AC with a dendritic field 0.5 mm in diameter and a density of
exploration has been made of connectivity in these regions, but 200 cells/mm2 has a CF of 39; each point in the image is
one might imagine that BCs or HCs that might avoid blue sampled by the receptive fields of 39 ACs of that class. Each
cones elsewhere cannot do so in these regions; or they may kind of retinal neuron has a coverage that reflects the sampling
be excluded from them. Blue cones are not the only variable necessary to create a seamless set of signals. Patterns can be
types. The linear visual streak in turtles, where cone densities very orderly (crystalline), statistically orderly, uniformly dis-
approach primate foveal levels, is disproportionately enriched orderly (random) or statistically clumped. One measure of order
in red cones expressing VP620 and red oil droplets. The tem- within a cell class is the conformity ratio: CR = NND/s, where
poral foveas of some avians are positioned within a red-field NND is the mean nearest-neighbor spacing between cells in
some 4–5 mm in diameter highly enriched in red cones a class and s is the standard deviation of that spacing.139,140 For
expressing VP580 and red oil droplets. Finally, but no less com- large samples, CR ~3 when patterns are statistically orderly
pelling, the tritanopic or blue-blind fovea of the primate foveola and C > 10 when patterns become nearly crystalline, with clear
is a small zone of ~15 min of arc with few or no blue axes of object orientation. The primate peripheral blue cone
cones.24,135,136 Roughly following rods, blue cones increase in pattern can reach CR > 3–5, while patterning near the fovea
frequency until they represent 5–10% of the cones (depending seems random. Human blue cone patterns seem less rigid than
on species), peaking at ~0.3–1°, thereafter following the density those of other primates. Nonmammalians, especially teleost
decline of the cones. The mechanisms controlling fine fishes, can display stunning crystalline mosaics141 and blue
variations are unknown but the molecular control of overall cones in those mosaics have CR values approaching 30
cone differentiation may involve selective fibroblast growth (Fig. 122.18), which is so precise that such images can be used
factor (FGF) receptor expression.137 The mechanism that as optical diffraction masks. No mammalian neuron has

FIGURE 122.20. Fine-scale cone patterns.


(Left) Blue cones in the baboon retina visualized
with a redox probe (135). The nonblue cones
were randomly selected to represent VP560
(red) or VP530 (green) cones. The blue cone CR
is 5.5 in the larger data set (not shown). (Right)
Precise blue cone patterns from flatfish retina
(Pleuronectes sp) associated with a precise,
repeating cone mosaic, with CR = 26.7.
(Left) © REM 2003. (Right) Derived from Engstrom K,
Ahlbert IB: Cone types and cone arrangement in the
retina of some flatfishes. Acta Zool 1963; 44:1–11,
edge filtered and thresholded.

1582
Functional Anatomy of the Neural Retina

BOX 122.7 Multiple pathways for rod signals


• Canonical “Starlight’’ pathways
• Canonical Scotopic OFF pathway
rods >i rod BCs > rod ACs >i OFF cone BCs > OFF
cone GCs
• Canonical Scotopic ON pathway
rods >i rod BCs > rod ACs :: ON cone BCs > ON
cone GCs
• Noncanonical “Moonlight” pathway
• Noncanonical direct OFF pathway
rods > mixed OFF BC > OFF cone GCs
• Noncanonical “Twilight’’ pathways
• Noncanonical OFF coupling pathway
rods :: cones > OFF cone BCs > OFF cone GCs

CHAPTER 122
• Noncanonical ON coupling pathway
rods :: cones >i ON cone BCs > ON cone GCs

by these high-gain canonical scotopic ON and OFF pathways.


The integration of rod signals by rod BCs generates a response
far more sensitive than an individual rod.18 Rod BCs drive
glycinergic rod ACs with AMPA receptors, making the gly-
cinergic rod AC one of the most light-sensitive elements in the
retina.146 Glycinergic rod ACs, or perhaps the aggregate IPL rod
network, likely generate the scotopic threshold potential of
the electroretinogram (ERG).147 At slightly higher brightnesses,
it is thought that a small population of OFF cone BCs behave as
FIGURE 122.21. Three major rod pathways in the mammalian retina. mixed rod–cone BCs (a cell type abundant in nonmam-
Pathway one is initiated by the high-gain rod >i rod BC > glycinergic malians19) and collect a small number of rod inputs.18 These
rod AC chain. Glycinergic rod ACs are fanout devices that pass rod cells may require higher brightnesses and larger rod responses
signals to ON cone BCs via gap junctions (resistor symbol) and to to generate perceptual responses. Why these are segregated to
OFF cone BCs via sign-inverting glycinergic synapses. Pathway 2 is OFF channels remains unclear, and these paths have only been
initiated at higher scotopic brightnesses by direct rod > OFF BC
found in nonprimates so far. However, Li et al148 have shown
signaling. Pathway 3 is initiated by small gap junctions (Fig. 122.8) at
mesopic ranges and mediate signaling directly though cone pedicles. that all rabbit OFF BC classes show some minor rod input,
© REM 2005. though not all individual cells do. Any cell in the equivalent of
class dB2 and dB3 BCs has ~80% chance of contacting a few
rods. Finally, in the mesopic range where both rods and cones
patterning this rigid. Conversely, widely spaced cells such as begin to operate, a ‘twilight’ system allows significant leakage of
dopamine neurons, other AxCs and IPCs142 in many species numerous rod signals into a sparse array of cone pedicles directly.
have apparently random mosaics. This would be expected of
neurons with global signaling modes, mediated more by volume The Achromatic Cone Pathways
diffusion (e.g., dopamine) than by specific cell contacts. Again, As normal human vision seems richly colored, the concept of
CR is of more than academic interest. Though the genetic and abundant achromatic channels seems odd. But sampling units
signaling mechanisms that control spatial patterning are still in retina must measure the spectral dispersion of light reflected
largely unknown,143,144 defects in these pathways may cause from an object as the sum of R+G+B (or at least R+G) signals
serious sensory impairment. so that the visual system can encode both object brightness
and the spectral purity or saturation of a patch of light. This is
one role of the diffuse cone BC system. Put simply, these cells
THE BASIC PATHWAYS OF RETINAL collect summed cone signals and pass them on to parasol GCs
SIGNALING and other wide-field GCs (Fig. 122.22, Box 122.8). However, the
The Basic Rod Pathways pathways for brightness are clearly more complex, as color-
Five discrete variants of three major pathways inject rod signals coded midget systems become noncolor-coded in parafovea and
into the visual system (Fig. 122.21, Box 122.7).18 No known beyond, as they randomly collect signals from midget BCs
GC population exclusively carries rod signals, although it contacting R and G cones.108 There are two likely pathways for
was reported that GC-like biplexiform cells made direct rod cone OFF BCs. Extending concepts gleaned from homologous
contacts with their dendrites.145 This CG has not yet been vali- BCs in ground squirrel retinas, primate class dB1 and dB2 likely
dated to form a distinct population in the mammalian retina. use KA receptors,64–65 which are highly glutamate sensitive,
In pathway 1, the so-called ‘starlight’ circuit, narrow-field gly- explaining the positioning of their dendrites as flat contacts far
cinergic rod (AII) ACs collect signals from several rod BCs and from the ribbon active site. KA receptors are also slightly
redistribute them via gap junctions to ON cone BCs and via slower and more sustained in current responses than AMPA
sign-inverting glycinergic synapses to OFF cone BCs. The same receptors likely used by dB3 cells. The idea that AMPA and KA
brain pathways that carry cone signals process perception of receptors parse the visual world into more and less transient
scotopic signals. At near-threshold levels, detection is mediated temporal events deserves careful analysis. Consistent with 1583
RETINA AND VITREOUS

FIGURE 122.22. Wide-field achromatic signaling


via OFF (left) and ON (right) parasol GCs in
primate retina. Each GC collects signals directly
from a set of diffuse cone BCs in its central
dendritic field and captures sign-inverting signals
from distant BCs via BC > AC >i BC > GC lateral
feedback and BC > AC >i GC lateral feedforward
chains.
© REM 2005.

BOX 122.8 Pathways for diffuse cone BC signals


• Major Diffuse OFF pathways
cones > diffuse OFF KA BCs (dB1, dB3) > OFF cone
GCs
cones > diffuse OFF AMPA BCs (dB2) > OFF cone GCs
• Minor Diffuse OFF pathways
SECTION 3

rods > diffuse OFF BC (mostly dB2, dB3) > OFF cone
GCs
• Diffuse ON pathways
cones > diffuse ON BCs (dB4, 5, 6) > ON cone GC

this, it would appear that large transient GCs sample the


dB3 arbor.79
FIGURE 122.23. Narrow-field chromatic signaling in bistratified blue
The Blue Cone Pathways ON/yellow OFF GCs (left) and monostratified ON and OFF foveal
Blue ON / yellow OFF (B+/Y–) GCs are the only known mam- midget (right) GCs in primate retina. Blue ON signals are captured via
malian retinal cells with spectrally biphasic receptive field blue cone >i blue BC > blue GC chains in the proximal arbor. Yellow
centers.82,108 Many nonmammalians have such color-opponent (R+G) OFF signals are captured via R + G cone > dB2 OFF BC > blue
GC chains. Midget GCs that contact single cones have pure R
GCs, BCs, and HCs.101 Both large and small classes of
(VP560) or pure G (VP530) centers and AC-mediated surrounds driven
bistratified GC sample from level 80 of the IPL to capture by varied mixtures of R+G cones. There is also evidence for blue cone
signals from the unique blue cone BCs and levels 20–30 to > OFF midget BC signaling (not shown).
capture dB2 BC R+G signals (Fig. 122.23, Box 122.9).69 It is © REM 2005.
presumed that both arbors of the B+/Y– GCs express the same
mixture of AMPA and NMDA receptors, but this is not known.
Recent evidence suggests the existence of a midget blue–OFF
BOX 122.9 Pathways for blue cone signals
pathway,61 although how this cell would function in vision is
less clear given the problem of chromatic aberration, where • B+ center/Y- center pathway:
optimal focus on R and G cones would blur images on the B B cones >i blue ON BCs > B+/Y- small and large bistratified
cone mosaic. Blurring is arguably one of the selection pressures GCs
forcing sparse distributions of B cones in all species. Finally, the RG cones > diffuse OFF KA BCs (dB2) > B+/Y- small and
newly described melanopsin GC is a large-field Y+/B– cell,83 large bistratified GCs
though the path by which the B– signals are acquired is not • Blue OFF center/surround pathways
clear. B cones > blue OFF midget BCs > blue OFF midget GC
• B-/Y+ large field pathways
Red–Green Color-Opponent Pathways Pathway for B cones not known for this cell
Since the early 1970s it has been clear that the primate retina
passes an assortment of color opponent signals to the LGN and
that these tend to be grouped into four categories:
• R+ center / G– surround cones or at least R+G cones, that mixture might be present in
• R– center / G+ surround all midget BC surrounds.153 If R and G cones are not
• G+ center / R– surround chromatypes and downstream BCs, ACs, and GCs make
• G– center / R+ surround. opportunistic contacts, how can nearly pure R or G surrounds
appear?149 The answer may partly come from two features of
One physiological view originally held that the surround paths cone patterning. First, the proportions of R and G cones can
of midgets must also be spectrally pure, while other studies vary across individuals (and perhaps species), with human R:G
support spectral mixing by random contacts.73,108,149–151 On values ranging from 16 to 1.154–156 Second, fine-scale patterning
balance, the evidence suggests that cone-specific contacts are of R and G cones is often statistically clustered rather than
not present in the R and G channels of the primate retina. For randomly uniform.156 Thus a single G cone ˜ midget BC ˜
example, though a midget BC ˜ midget GC transfer may report midget GC chain in an R cone-dominated retina may show
the signals of a single R cone, the ACs that comprise the lateral nearly pure R surround signaling. And similarly, that chain in a
elements collect from nearly all adjacent midget BCs,152 so that retina with equal numbers of R and G cones can plausibly be
the composition of the surround might be spectrally mixed found centered in a patch of R cones, since the dendritic fields
1584 (Fig. 122.23, Box 122.10). And if HCs clearly collect from all of ACs associated with midget cells also tend to be small.157
Functional Anatomy of the Neural Retina

BOX 122.10 Pathways for red and green cone signals


• Red or Green ON center pathway:
R or G cones >i midget ON BC > midget ON GC
• Red or Green ON center pathway:
R or G cones > midget OFF BC > midget OFF GC
• Generic surround pathways:
RG cones > H1 HC >i RG cones > …
RG cones > H1 HC > midget ON BC …
RG cones > H1 HC >i midget OFF BC …
R and G midget BCs > small-field ACs >i midget BCs >
midget GC
R and G midget BCs > small-field ACs >i midget GC

BOX 122.11 Increment spectral sensitivity functions in


FIGURE 122.24. The mismatch between primate photopic spectral

CHAPTER 122
trichromatic primates sensitivity profiles and VP absorption functions plotted on a
• The absorption peaks of cones do not predict the daylight normalized log10 sensitivity ordinate (log S) and a linear wavelength
increment threshold spectral sensitivities of primates abscissa (l). Dotted lines from left to right are normalized VP420,
(Fig. 122.24). VP499, VP530, and VP560 absorbance functions. The circles are the
4-day mean increment threshold spectral sensitivity for a rhesus
• No sum of absorptions reproduces the spectral sensitivity
monkey in log S = log (1/Q) for a 2° foveally centered test flash on a
envelope.
10° neutral white 10 000 K background Maxwellian view field. Data
• Cones express VP560 (yellow-green), VP530 (green), or recorded by REM in 1971.There are several mismatches with the
VP420 (violet-blue). pigment curves: (1) The long-l peak is at 610 nm;(2) The mid-l
• Sensitivity peaks are 610 nm (orange-red), 520–530 nm complex is broad; (3) There is a minimum at 580 nm; (4) There is a
minimum at 470 nm; (5) The short-l peak is at 450+ nm; (6) The short-
(green), and 430 nm (blue).
l band is narrow; (7) No sum of VPs matches the spectral sensitivity.
• A number of factors shape the sensitivity channels of the © REM 2005.
eye, but the most important are opponent interactions.
• In the absence of opponent functions, the R sensitivity peak
moves to the VP peak:
If green cones are desensitized by adapting lights, the long- BOX 122.12 Speculative pathways for melanopsin GCs
wave sensitivity peak progressively moves from 610 to • RG Cones >i ON diffuse BCs (dB6?) > inner melanopsin GCs
560 nm.
• RG cones > OFF diffuse KA BCs (dB1?) > GABA ACs
>i outer melanopsin GCs
• B cones >i blue ON BCs > GABA ACs >i inner melanopsin
Given the individual variability in the R/G cone ratio, it is GCs
surprising that many aspects of color vision seem stable across
• B cones > ? > outer melanopsin GCs
individuals, such as the photopic increment threshold spectral
sensitivity (see Box 122.11, Fig. 122.24) and the wavelength of • Melanopsin GCs > LGN > Cx > brightness perception?
unique yellow.158 Other measures that probe the densities of • Melanopsin GCs > SCN > photoentrainment
current generators in the eye (e.g., the ERG spectral sensitivity) • Melanopsin GCs > pretectum > pupillary reflex
show that known variations in the R/G ratio roughly predict ? denotes unknown or uncertain.
spectral peaks.158 Perception does not vary much. This suggests
that there is a normalization mechanism in the visual system,
such as activity-dependent axon sorting; a major mechanism for
organizing sensory fields. Cortical area V2 in macaque shows to flux over many decades83 argues that they may correspond to
strong evidence of such spectral sorting.159 Furthermore, color intrinsic luminosity cells of the visual system.
perception itself shows evidence of highly plastic properties that
tune vision, perhaps regardless of R/G cone ratio.160 FURTHER ELEMENTS OF RETINAL
STRUCTURE
Melanopsin Pathways
The melanopsin pathway is an exciting discovery, but not easy
to understand as these cells integrate signals from rods, cones INTRODUCTION
and their own intrinsic phototransduction. Dacey et al show The retina is a complex, dynamic neural structure. Despite the
that primate melanopsin GCs are also rare Y+/B– cells but that accumulation of apparently precise pathway models, they are
both inner and outer classes of melanopsin GCs show the same incomplete in many ways. The modes and weights of HC
polarity of response.83 The signaling channels for melanopsin signaling are still uncertain. The most common synapses in the
GCs (Box 122.12) include rods and cones via directly glutamate IPL, serial AC ˜ AC elements, have had no formal place in
gated AMPA and/or NMDA receptors. The intrinsic 11-cis any model until recently. Many modes of physiological sig-
retinaldehyde isomerization coupled transduction88 accesses an naling involve widespread targets and multiple sources,
unknown conductance to initiate spiking. Melanopsin GCs including retinal efferents, and their roles in vision is not
appear to be a diverse population morphologically and target the understood. MCs could clearly locally modulate neural efficacy
LGN, SCN, and olivary pretectum. The fact that they project to through ATP-gated channels and regulation of extracellular ion
LGN and give extremely sustained light responses proportional and neurotransmitter levels, but it is not known if they do. 1585
RETINA AND VITREOUS

Visual experience in development influences GC maturation 0–10 of the IPL with medium-field dendritic arbors and fine,
and loss of visual drive in retinal degenerations triggers neural complex axon terminal fields.172 It has long been suspected that
remodeling of the retina. Some GCs are intrinsically most retinas harbor more than one class of TH immunoreactive
photosensitive, as has been seen. This next section will briefly cell173,174 and rodents clearly possess at least two; TH1 cells are
review the imports of some of these topics. typical AxCs and are not GABA immunoreactive, while TH2
cells are more AC-like (lacking axons), often express epitope-
masked or phosphorylation-masked TH immunoreactivity and
SERIAL SYNAPSES AND NESTED FEEDBACK are also GABA immunoreactive.175,176 TH1 AxCs appear to be
Serial AC ˜ AC synapses comprise about two-third of the IPL spontaneously spiking ‘clock-like’ cells that resemble spon-
synapses in nonmammalians161 and a smaller fraction in taneously signaling dopamine neurons in brain.177,178 They
mammals,162 yet they had no established role in any pathway, have some weak excitatory inputs but many inhibitory inputs
until recently. Indeed, AC ˜ AC ˜ AC triplets are far from appear to be under strong GABAergic control.3 In any case, TH1
rare, which offers significant opportunities to shape circuits, cells appear to be activated by photopic stimuli, making them
but why? And how? At the least, this argues that most network signalers of dawn. Dopamine released by vesicular means
models are incomplete. Some evidence indicates that such probably diffuses through the retina to its various targets.171
synapses are part of nested feedback and feedforward paths Virtually every class of retinal cell, including photoreceptors and
• Feedback: BC > AC >i BC MCs has been reported to express dopamine receptors.171
• Nested Feedback: BC > AC >i > AC >i BC Basically, dopamine signals appear to light-adapt the retina by
SECTION 3

• Feedforward: BC > AC >i GC diverse G-protein coupled mechanisms such as HC uncoupling179


• Nested Feedforward: BC > AC >i > AC >i GC and enhancement of GC response speed.180 The scale and scope
of these adaptive signals are under intense study, but it is of
Concatenating sign-inverting paths formally represent positive great interest that retinal dopamine cells also display high levels
feedback, which is potentially destabilizing: AC >i AC >i BC ß of circadian clock gene expression, potentially making them key
AC > BC. Biological reality is more complex. First, the elements for intrinsic retinal photoperiod control.181 This
abundance of serial chains means that positive feedback, if it pathway appears separate from the melanopsin GC path, but it
occurs, does not destabilize vision. Second, these signals are is intriguing that the dendrites of outer melanopsin GCs co-
progressively delayed in time, so their effects do not sum stratify with TH1 cells.
statically. Finally, the gains of inhibitory synapses are typically
fractional (<1), so the net pathway gain becomes even
smaller.161 This suggests that serial synapses could effectively NONCANONICAL SIGNALING MODES
shape many pathways in subtle ways and may be generic to In many ways the retina is a complex neuroendocrine organ
many feedback and feedforward networks. whose operations we understand poorly. We can only offer a
More recently, Hennig et al163 implemented nested AC brief survey of many intrinsic ‘parahormonal’ systems in retina
elements in a comprehensive mathematical model of linear and that depend on nonvesicular or diffusion-based signaling
nonlinear retinal GC signaling, and found that the influence of (e.g., dopamine neurons). Photoreceptors are known to syn-
nesting was weak, consistent with subtle shaping rather than thesize and release melatonin at night, and this signaling is
destabilizing positive feedback. Results from the laboratory of thought to modulate both RPE-mediated photoreceptor outer
the late Ramon Dacheux strongly implicate serial synapses in segment phagocytosis and the activity of dopaminergic
signaling asymmetries that may contribute to the directional neurons.3,182 There is now evidence, in addition to cyclic cons-
biases of DS GCs.80,164 titutive regulation, that excessive melatonin exposure enhances
the sensitivity of photoreceptors to light damage and specifically
regulates the expression of a restricted set of ocular genes.182
EFFERENTS Neurons with the capacity to express high levels of nitric
Efferent pathways from CNS systems are well known in oxide (NO) synthase and generate NO as a signaling molecule
nonmammalians but have been controversial in mammals, have been termed nitrergic.3,8,183 Some of these cells are clearly
though both Ramón y Cajal165 and Polyak166 described them. AxCs.8 By local diffusion across cell membranes, NO binds to
Polyak clearly identified efferent fibers in the optic nerve that the heme core of soluble guanyl cyclase and activates high levels
arborized in the IPL of primate retina166 and separate serotonin- of cGMP production which, in turn, has the capacity to
and histamine-immunoreactive efferents targeted the IPL have modulate gap junction permeability184 as well as open local
now been described.167–170 The roles of these efferents for vision cyclic nucleotide gated cation channels. The regulation of these
remains unknown, but the close association of both systems systems and their light stimulus selectivities are not well
with retinal vessels is provocative. Further, both rod and cone known and it is suspected that many cell classes can produce
ON BCs in baboon express HR3 histamine receptors on their NO beyond the identified nitrergic AxCs.183
dendritic tips, but the relation between neuronal expression of GABA has intermittently been identified in BCs of the
histamine receptors and IPL efferents is uncertain.171 However, mammalian retina, including cat and monkey, with no clear
the discovery of serotonin-immunoreactive efferents gives the mechanism of action.3 Recently it has been suggested that
first compelling physiological drive path for serotonin-receptor a set of OFF cone BCs in cat retina similar to dB1 in monkey are
systems in the retina, as there are no known intrinsic sero- dual GABA/glutamate-releasing neurons.185 The co-stratification
toninergic neurons in the mammalian retina, unlike nonmam- of the terminals of these BCs with the dopaminergic TH1
malians3 which express elegant wide-field GABAergic/ plexus in the distal IPL suggests the possibility of OFF BC >i
serotoninergic ACs. dopamine AC signaling, which would be consistent with the
apparent depolarization of dopamine cells by light, yet
restriction of synaptic inputs to the OFF BC region of the IPL.
THE DOPAMINE PATHWAY GABA-immunoreactive BCs appear more abundant in monkey
Dopamine neurons represent the quintessential AxC and it is not clear, however, that they can all be OFF BCs.
pathway.3,18,171 The dominant TH1 (tyrosine hydroxylase MCs and the optic fiber layer are key positions to modulate
1586 type 1) AxCs, are narrowly stratified cells that arborize at level the signaling of neurons in the retina. Indeed it appears that
Functional Anatomy of the Neural Retina

excitation-induced calcium waves that propagate locally in the revisions of retinal circuitry in three phases.11 In phase 1,
GCL and OFL in the astrocyte and MC network can directly photoreceptor stress triggers the retraction of BC dendrites from
modulate GC excitability.186 Moreover, MCs have now been rod and/or cone synapses. Indeed, the first signs of visual
shown to release ATP, which may directly activate calcium entry impairment in RP are likely to be a result of the phase 1 loss of
via purinergic receptors on vascular pericytes, in turn triggering dendritic compartment in BCs before there is significant loss in
local vasoconstriction.187 Thus retinal activity has the potential photoreceptor signaling. Indeed, given the ability of BCs to
to locally regulate blood flow through MC signal integration. report even small photoreceptor signals, early visual
The scope and strength of such MC signaling is yet unknown. impairment more readily implies defects in synaptic signaling
Peptide-releasing neurons in the mammalian retina include than phototransduction. The diverse genetic types of RP exhibit
different cohorts of wide-field GABAergic ACs that express different modes of photoreceptor loss. If the mode of degener-
neuropeptide Y (NPY), substance P (SP) or vasoactive intestinal ation is cone-sparing, rod BCs attempt to transient capture
peptide (VIP), AxC-like cells expressing somatostatin (SRIF) inputs from surviving cones. If the degeneration is rod–cone
and minor (but not necessarily unimportant) populations lethal, all BCs disassemble their dendritic modules, including
expressing several other neuroactive peptides.37 The detailed their signaling receptors. In phase 2, photoreceptor death leads
relationships between fast neurotransmitter versus slow peptide to loss of the ONL and the development of a MC seal between
secretion signaling from the same cell are simply unclear. But it the remnant RPE (if it survives) or the choroid and the remnant
has long been suspected that peptide-containing vesicles are neural retina. Finally, in phase 3, the retina undergoes a
released by an exocytosis mechanism that requires much more prolonged epoch of revision that involves additional neuronal

CHAPTER 122
calcium entry (and hence stronger depolarization) than fast death, formation of new process fascicles and new ectopic
neurotransmitter vesicle fusion. Even when there is evidence of synaptic microneuromas, and even neuronal migration leading
a possible signaling mode, we have little insight as to the to mixing of the INL and GCL through disrupted zones of the
purpose. For example, SRIF appears excitatory on a seconds-to- IPL.11,193 Synaptogenesis leads to new networks in micro-
minutes timescale in GCs188 and induces an increase in input neuromas, and these networks appear to be random collections
resistance in ON BCs.189 How these events are related remain of opportunistic connections. These generate networks that
unclear. Though it is widely accepted that specific peptides seem optimized for self-excitation rather than visual sig-
likely have circuit-specific modulatory functions, specific roles naling.194 In general, these transformations challenge thera-
in any of the canonical pathways are not known. It is also not peutic intervention windows and repair strategies of all types,
known how far peptides diffuse and how long they persist in the from genetic to bionic, for all forms of retinal degeneration.
extracellular space. Other peptide-like associations have even
less certain functions. The blue cone BCs of primates also SUMMARY AND PERSISTENT QUESTIONS
express cholecystokinin (CCK)-like immunoreactivity26,37 and
CCK does suppress GC activity, but no correspondence has Our understanding of the populations of neurons that make up
been established for this in the canonical blue cone ˜ blue cone a retina has expanded. Clearly, at least 60 and maybe even
BC ˜ B+/Y– GC pathway. And it is not clear that blue cone 80 cell classes are involved, and our catalog is likely to become
BCs actually release bona fide CCK. even more detailed.195 Though most of the canonical pathway
neurons have likely been identified, every pathway is beset with
questions regarding signaling modes and strengths; almost
ACTIVITY-DEPENDENT PLASTICITY, RETINAL every pathway has undefined synaptic partners, especially
REMODELING, AND PHOTORECEPTOR among the AC cohort; and every retina almost certainly harbors
DEGENERATIONS small populations of poorly defined or even yet-unknown
The elegant structure of the mammalian retina can no longer neuron classes.195 Sparseness or difficulty in identification of a
be viewed as static and hard-wired. Indeed, the retina undergoes cell class does not imply relative unimportance. TH1 dopamine
postnatal refinement in synaptic connectivity,190,191 possible AxCs are among the rarest of neurons in the ACL and their
revisions in gene expression in response to visual environments actions are global and powerful. Melanopsin GCs are among the
at maturity, and reactive rewiring when challenged by rarest of GCs and they are essential carriers of photoperiod and
photoreceptor degenerations.11 We are only beginning to pupillary control signals. We still do not have a proven role for
understand the scope of these physical transformations, but it a clearly heterogeneous set of retinal efferent fibers originating
is now clear that adult retinal neurons can revise their patterns in hypothalamus and brainstem.
of synaptic contacts and generate new processes.11 During Even simple issues; such as how neurons choose partners to
postnatal life in rodents, the visual environment influences the contact, remains elusive. This is especially true of the R and G
onset of bouts of spontaneous signaling thought to be required cones of trichromatic primates, which seemed to have evolved
for synaptic maturation144,190 and modulates the segregation of so recently via VP gene duplication that no other gene
the IPL into ON and OFF sublayers, apparently through a expression differences have clearly emerged that would ‘label’
dendritic pruning process.191 While previous research in the them as specific chromatypes for putative R–G opponent neu-
1970s produced contentious views on activity-dependent retinal rons, as did occur in nonmammalians. Conversely, B cones and
maturation,11 many studies can now be revisited in light of rods each express many different genes (beyond VP expression),
modern findings. The IPL is clearly the site of most of these some of which clearly drive formation of selective contacts. But
effects, but it would now be imprudent to exclude the OPL. what are we to make of new patterns of cones that selectively
Recently, Fisher and colleagues have detailed a range of express vGlut2 but are still G cones in mouse,29 or sparse
cellular remodeling phenomena and mechanisms including human cones that express no opsin except melanopsin?30
rapid neurite sprouting, neuronal migration and MC hyper- HCs, the first cells in the outer retina from which
trophy in response to chronic retinal detachment.192 These intracellular recordings were ever made, remain one of the most
plastic abilities of adult neuron cells presaged discoveries enigmatic. Are they truly neurons? How do they signal their
perhaps even more surprising to retinal biologists (but not to targets? Why do they form nearly half of the capillary
CNS biologists); the amazing propensity for retinal neurons to endothelial ensheathment in the mature retina?196 What does
rewire aggressively in response to retinal degenerations.11,193 So the rod-specific axon terminal actually do? Is signaling
far, all known photoreceptor degenerations trigger major transmitter-mediated by GABA or is it ephaptic, or both? 1587
RETINA AND VITREOUS

Why do most HCs express no GABA? Where are the vesicles essential functions, which are likely nonperceptual, likely
and are the vesicles we can see presynaptic or cargo vesicles? involve more types of GCs than the major perceptual
What are the selective roles of H1 and H2 HCs. And where is pathways,1 which in turn demand the bulk of retinal wiring.
the H3 HC array? Further classification and reconstructing the connectivity of
The BC population is clearly settling into 10 defined classes; diverse GC classes remains a key target for the next decade.
a single rod BC class, a single blue cone ON BC class, two In the end, why should such effort be applied to the details
midget cone BC classes (ON and OFF), and at least three classes and the nuances of neuronal form and retinal circuitry? First,
each of diffuse ON cone BCs and diffuse OFF cone BCs. The discoveries based on new molecular imaging tools continue to
canonical rod ˜ rod BC ˜ rod AC ˜ cone BC ˜ cone GC challenge any simplistic model of retinal organization by
pathway has now been augmented with a minor rod ˜ diffuse finding new cells, new contacts and new functions in the retina.
OFF cone BC ˜ OFF cone GC path, owing to very small Second, a range of inherited disorders arise from genes associ-
numbers of rod contacts made by some OFF cone BCs.148 The ated with building the neural retina and those genes represent
stochastic nature of these contacts gives pause (not all OFF BCs both our evolutionary path and mechanisms we must under-
of a given class make them), but they clearly function. The stand if we are ever to make retinal repair a reality. Third, the
discovery of blue OFF midget BCs61 might partly explain the details of wiring and global control, and the scope and speed of
sparse appearance of B–/Y+ neurons in the primate LGN197 disease-triggered rewiring reveal that the effects of many forms
except for the fact that the fields of these cells tend to large and of retinal degenerations once thought to be restricted to the
better match those of melanopsin GCs, whose dendrites likely outer retina actually propagate aggressively into the neural
SECTION 3

never go near the terminals of blue OFF midget BCs. An retina and likely the brain. Learning the rules and molecular
exciting new concept in BC physiology is the idea that AMPA mechanisms underlying postnatal plasticity is required to
receptors and KA receptors are differentially expressed on dB3 realize retinal restoration in diseases and traumas we cannot yet
and dB1/2 OFF cone BCs respectively, perhaps setting up the prevent. Finally, the neural retina remains unbowed. Though
basis of fast (transient) and slow (sustained) BC channels64 and we believe we have disclosed its essences, it is proving to be a
matching GC contacts. If this is a basic format, then one might far richer organ than anticipated.
expect mGluR6-mediated transduction to vary in kinetics across Retinal neuroanatomy is not a static field. The accelerating
classes of diffuse ON cone BCs. Specifically, might not dB4 pace of discoveries augurs major revelations in retinal circuitry
resemble dB3 in being the ‘fast’ cone BC for the ON channel? in the next decade, rather than mere refinements of current
The ACs and the specialized, axon-bearing group we term views. Since the last revision of this chapter, over 1500 papers
association or AxCs (which include polyaxonal cells), remain have been published with reference to cones alone; over 1000
our biggest challenge to understand. The diversity of AC on retinal GCs alone; over 1000 on ACs and BCs combined.
dendritic arbors clearly does not fit a simplistic world of ACs Over half of the references cited herein have been published
monostratified for corresponding BCs44 but does argue for since the year 2000. Many of the references are reviews and
highly circuit-selective functions.195 It is possible that glycine space limitations prevent a traditional historical treatment of
AC systems are biased to signal across BC channels while the literature. It is no longer practical to cite the first instance
GABA AC systems mostly signal within BC channels. The of an idea or discovery (e.g., Ramón y Cajal or Tartuferi), much
signaling of GABA receptors remains complex and likely finely less its most lucid modern declamation or important related
tuned. Each conventional inhibitory synapse access a mixture of papers. It is hoped that readers will use this chapter as a point
receptors or individually pure receptor patches. This fine-scale of departure in a greater scientific and medical adventure.
analysis is just beginning.
The cohort of GCs has been better circumscribed over the
past decade, but has also become far more complex with the
ACKNOWLEDGMENTS
advent of multimodal melanopsin GC signaling and more cell These last lines are the most difficult. On 30 May 2006 Ramon F Dacheux
classes than we have models for. We are closer than ever to passed away: untimely, much beloved and admired. He and Elio Raviola
understanding how DS GCs work, though study of such cells in crafted the modern view of retinal neuroanatomy for Principles and
primates is just beginning. Many classes of GCs clearly have Practice of Ophthalmology a decade ago. It strongly guided my revisions.
Much new science has transpired and I have retained few references.
more to do with the optical ‘plant’ of the eye; guiding fast and
I believe that nothing would have pleased Ray more than the inclusion of
fine eye movements, driving foveation, discriminating self- one of his most vivid recent accomplishments: Dacheux RF, Chimento MF,
movement from world-movement, creating the optic flow field, Amthor FR. Synaptic input to the on-off directionally selective ganglion cell
harmonizing visual drift and vestibular information, etc. These in the rabbit retina. J Comp Neurol 2003; 456:267–278.

REFERENCES
1. Oyster C: The human eye. New York: neurosciences. Cambridge: MIT Press; Imaging neuronal subsets in transgenic
Sinauer; 1999. 2004:320–333. mice expressing multiple spectral
2. Bayer BE: Color imaging array. US patent 6. Slaughter M: Inhibition in the retina. In: variants of GFP. Neuron 2000;
no. 3971065; 1976. Chalupa LM, Werner J, eds. The visual 28:41–51.
3. Marc RE: Retinal neurotransmitters. In: neurosciences. Cambridge: MIT Press; 10. Morgan J, Huckfeldt R, Wong RO: Imaging
Chalupa LM, Werner J, eds. The visual 2004:355–368. techniques in retinal research. Exp Eye Res
neurosciences. Cambridge: MIT Press; 7. Kamermans M, Fahrenfort I: Ephaptic 2005; 80:297–306.
2004:315–330. interactions within a chemical synapse: 11. Marc RE, BW Jones, CB Watt, Strettoi E:
4. Wässle H: Parallel processing in the hemichannel-mediated ephaptic inhibition Neural remodeling in retinal degeneration.
mammalian retina. Nat Rev Neurosci 2004; in the retina. Curr Opin Neurobiol 2004; Prog Retin Eye Res 2003; 22:607–655.
5:747–757. 14:531–541. 12. Marc RE, BW Jones, Watt CB: Retinal
5. Copenhagen D: Excitation in the retina: the 8. Vaney D. Retinal Amacrine Cells. In: remodeling: circuitry revisions triggered by
flow, filtering and molecules of visual Chalupa LM, Werner J, eds. The visual photoreceptor degeneration. In: Pinaud R,
signaling in the glutamatergic pathways neurosciences. Cambridge: MIT Press; Tremere LA, De Weerd P, eds. Plasticity in
from photoreceptors to ganglion cells. In: 2004: 395–409. the visual system: from genes to circuits.
Chalupa LM, Werner J, eds. The visual 9. Feng G, Mellor RH, Bernstein M, et al: Springer; 2005: 33–54.
1588
Functional Anatomy of the Neural Retina

13. Illing ME, Rajan RS, Bence NF, Kopito RR: retina. Invest Ophthalmol Vis Sci 2006; 50. Raviola E, Gilula NB: Intramembrane
A rhodopsin mutant linked to autosomal 47:1636–1641. organization of specialized contacts in the
dominant retinitis pigmentosa is prone to 31. Marc RE: Interplexiform cell connectivity in outer plexiform layer of the retina. A freeze
aggregate and interacts with the ubiquitin the outer retina. In: Archer S, Djamgoz MBA, fracture study in monkeys and rabbits.
proteasome system. J Biol Chem 2002; 37, Vallerga S, eds. Neurobiology of the J Cell Biol 1975; 65:192-222.
34150–34160. vertebrate outer retina. London: Chapman 51. Güldenagel M, Ammermüller J,
14. Carroll R: Vertebrate paleontology and and Hall; 1995:369–393. Feigenspan A, et al: Visual transmission
evolution. New York: W. H. Freeman and 32. Wehman AM, Staub W, Meyers JR, et al: deficits in mice with targeted disruption
company; 1988:698. Genetic dissection of the zebrafish retinal of the gap junction gene connexin36.
15. Famiglietti EV, Kolb H: A bistratified stem-cell compartment. Dev Biol 2005; J Neurosci 2001; 21:6036–6044.
amacrine cell and synaptic circuitry in the 281:53–65. 52. Deans MR, Volgyi B, Goodenough DA,
inner plexiform layer of the retina. Brain 33. Perron M, Harris WA: Retinal stem cells in et al: Connexin36 is essential for
Res 1975; 84:293–300. vertebrates. Bioessays 2000; 22:685–688. transmission of rod-mediated visual signals
16. Strettoi E, Dacheux RF, Raviola E: Synaptic 34. Hoffert JR, Baeyens DA, Fromm PO: The in the mammalian retina. Neuron 2002;
connections of rod bipolar cells in the inner resistance of teleost ocular tissues to 36:703–712.
plexiform layer of the rabbit retina. J Comp oxygen toxicity. Invest Ophthalmol 1973; 53. Li W, DeVries SH: Separate blue and green
Neurol 1990; 295:449–466. 12:858–861. cone networks in the mammalian retina.
17. Strettoi E, Raviola E, Dacheux RF: Synaptic 35. Kalloniatis M, Marc RE, Murry R: Amino Nat Neurosci 2004; 7:751–756.
connections of the narrowfield, bistratified acid signatures in the primate retina. 54. Hornstein EP, Verweij J, Schnapf J:
rod amacrine cell (AII) in the rabbit retina. J Neurosci 1996; 16:6807–6829. Electrical coupling between red and green

CHAPTER 122
J Comp Neurol 1992; 325:152–168. 36. Morgan JL, Dhingra A, Vardi N, Wong RO: in primate retina. Nat Neuroscience 2004;
18. Sterling P: How retinal circuits optimize the Axons and dendrites originate from 7:745–750.
transfer of visual information. In: neuroepithelial-like processes of retinal 55. Nelson R, Famiglietti EV, Kolb H:
Chalupa LM, Werner J, eds. The visual bipolar cells. Nat Neurosci 2006; Intracellular staining reveals different levels
neurosciences. Cambridge: MIT Press; 9:85–92. of stratification for on and offcenter
2004:234–259. 37. Brecha N: Peptide and peptide receptor ganglion cells in cat retina. J Neurophysiol
19. Ishida A, Stell W, Lightfoot D: Rod and expression and function in the vertebrate 1978; 41:472–483.
cone inputs to bipolar cells in goldfish retina. In: Chalupa LM, Werner J, eds. The 56. Missotten L: The ultrastructure of the
retina. J Comp Neurol 1980; visual neurosciences. Cambridge: MIT human retina. Brussels: Editions Arscia
191:315–335. Press; 2004:334–354. Uitgaven; 1965.
20. Graham DR, Overbeek PA, Ash JD: Leukemia 38. Migdale K, Herr S, Klug K, et al: Two ribbon 57. Kolb H: Organization of the outer plexiform
inhibitory factor blocks expression of Crx synaptic units in rod photoreceptors of layer of the primate retina: electron
and Nrl transcription factors to inhibit macaque, human, and cat. J CompNeurol microscopy of Golgi impregnated cells.
photoreceptor differentiation. Invest 2003; 455:100–112. Phil Trans R Soc Lond B 1970;
Ophthalmol Vis Sci 2005; 46:2601–2610. 39. Sterling P, Matthews G: Structure and 258:261–283.
21. Bumsted O’Brien KM, Cheng H, Jiang Y, function of ribbon synapses. Trends 58. Dowling JE, Boycott BB: Organization of
et al: Expression of photoreceptor-specific Neurosci 2005; 28:20–29. the primate retina: electron microscopy.
nuclear receptor NR2E3 in rod 40. Matthews G: Synaptic mechanisms of Proc R Soc Lond B 1966; 166:80–111.
photoreceptors of fetal human retina. Invest bipolar cell terminals. Vision Res 1999; 59. Boycott BB, Hopkins M: Cone synapses of
Ophthalmol Vis Sci 2004; 45:2807–2812. 39:2469–2476. a flat diffuse cone bipolar cell in the
22. Cheng H, Khanna H, Oh ECT, et al: 41. Wilson M. Retinal Synapses. In: Chalupa LM, primate retina. J Neurocytol 1993;
Photoreceptor-specific nuclear receptor Werner J, eds. The visual neurosciences. 22:765–778.
NR2E3 functions as a transcriptional Cambridge: MIT Press; 2004:279–303. 60. Tsukamoto Y, Morigiwa K, Ueda M,
activator in rod photoreceptors. Hum Mol 42. Marc RE, Jones BW: Molecular Sterling P: Microcircuits for night vision in
Genet 2004; 13:1563–1575. phenotyping of retinal ganglion cells. mouse retina. J Neurosci 2001;
23. Register EA, Yokoyama R, Yokoyama S:. J Neurosci 2002; 22:413–427. 21:8616–8623.
Multiple origins of the green-sensitive opsin 43. Rockhill RL, Daly FJ, MacNeil MA, et al: 61. Klug K, Herr S, Tran Ngo I, et al: Macaque
genes in fish. J Mol Evol 1994; 39:268–273. The diversity of ganglion cells in a retina contains an s-cone off midget
24. Bumsted K, Hendrickson A: Distribution mammalian retina. J Neurosci 2002; pathway. J Neurosci 2003; 23:9881–9887.
and development of short-wavelength 22:3831–3843. 62. Li W, DeVries SH: Bipolar cell pathways for
cones differ between Macaca monkey 44. MacNeil MA, Heussy JK, Dacheux RF, et al: color and luminance vision in a dichromatic
and human fovea. J Comp Neurol 1999; The shapes and numbers of amacrine cells: mammalian retina. Nat Neurosci 2006;
403:502–516. matching of photofilled with Golgi-stained 9:669–675.
25. Ahnelt PK, Kolb H, Pflug R: Identification of cells in the rabbit retina and comparison 63. Haverkamp S, Grunert U, Wässle H:
a subtype of cone photoreceptor, likely to with other mammalian species. J Comp Localization of kainate receptors at the
be blue sensitive, in the human retina. Neurol 1999; 413:305–326. cone pedicles of the primate retina.
J Comp Neurol 1987; 255:18–34. 45. Nelson R., Kolb H: On and off pathways in J Comp Neurol 2001; 436:471–486.
26. Kouyama N, Marshak DW: Bipolar cell the vertebrate retina and visual system 64. DeVries SH: Bipolar cells use kainate and
specific for blue cones in the macaque (2004). In: Chalupa LM, Werner J, eds. AMPA receptors to filter visual information
retina. J Neurosci 1992; 12:1233–1252. The visual neurosciences. Cambridge: into separate channels. Neuron 2000;
27. Nathans J: The evolution and physiology of MIT Press; 2004:260–278. 28:847–856.
human color vision: insights from molecular 46. Perkins GA, Ellisman MH, Fox DA: 65. Devries SH, Li W, Saszik S: Parallel
genetic studies of visual pigments. Neuron Three-dimensional analysis of mouse rod processing in two transmitter
1999; 24:299–312. and cone mitochondrial cristae microenvironments at the cone
28. Wang Y, Smallwood PM, Cowan M, et al: architecture: bioenergetic and functional photoreceptor synapse. Neuron 2006;
Mutually exclusive expression of human implications. Mol Vis 2003; 9:60–73. 50:735–748.
red and green visual pigment-reporter 47. Ripps H: Cell death in retinitis pigmentosa: 66. Boycott B, Wässle H: Parallel processing in
transgenes occurs at high frequency in gap junctions and the ‘bystander’ effect. the mammalian retina: the proctor lecture.
murine cone photoreceptors. Proc Natl Exp Eye Res 2002; 74:327–336. Invest Ophthalmol Vis Sci 1999;
Acad Sci USA 1999; 96:5251–5256. 48. Chun MH, Grunert U, Martin PR, Wässle H: 40:1313–1327.
29. Wässle H, Regus-Leidig H, Haverkamp S: The synaptic complex of cones in the fovea 67. Famiglietti EV Jr: Class I and class II
Expression of the vesicular glutamate and in the periphery of the macaque ganglion cells of rabbit retina: a structural
transporter vGluT2 in a subset of cones of monkey retina. Vision Res 1996; basis for X and Y (brisk) cells. J Comp
the mouse retina. J Comp Neurol 2006; 36:3383–3395. Neurol 2004; 478:323–346.
496:544–555. 49. Raviola E, Gilula NB: Gap junctions 68. Kolb H, Nelson R, Mariani A: Amacrine
30. Dkhissi-Benyahya O, Rieux C, Hut RA, between photoreceptor cells in the cells, bipolar cells and ganglion cells of the
Cooper HM: Immunohistochemical vertebrate retina. Proc Natl Acad Sci USA cat retina: a Golgi study. Vision Res 1981;
evidence of a melanopsin cone in human 1973; 70:1677–1681. 21:1081–1114. 1589
RETINA AND VITREOUS

69. Dacey DM, Peterson BB, Robinson FR, 88. Fu Y, Zhong H, Wang MH, et al: Intrinsically 106. Dacheux RF, Raviola E: Physiology of HI
Gamlin PD: Fireworks in the primate retina: photosensitive retinal ganglion cells detect horizontal cells in the primate retina. Proc R
in vitro photodynamics reveals diverse light with a vitamin A-based photopigment, Soc Lond B 1990; 239:213–230.
LGN-projecting ganglion cell types. Neuron melanopsin. Proc Natl Acad Sci USA 2005; 107. Dacey DM, Lee BB, Stafford DK, et al:
2003; 37:15–27. 102:10339–10344. Horizontal cells of the primate retina: cone
70. Yamada ES, Bordt AS, Marshak DW: 89. Freedman MS, Lucas RJ, Soni B, et al: specificity without spectral opponency.
Wide-field ganglion cells in macaque Regulation of mammalian circadian Science 1996; 271:656–659.
retinas. Vis Neurosci 2005; 22:383–393. behavior by non-rod, non-cone, ocular 108. Dacey DM: Parallel pathways for spectral
71. Coombs J, van der List D, Wang GY, photoreceptors. Science 1999; coding in primate retina. Annu Rev
Chalupa LM: Morphological properties of 284:502–504. Neurosci 2000; 23:743–775.
mouse retinal ganglion cells. Neuroscience 90. Silva MM, Albuquerque AM, Araujo JF: 109. Blanco R, de la Villa P: Ionotropic
2006; 140:123–136. Light-dark cycle synchronization of glutamate receptors in isolated horizontal
72. Kolb H, Marshak DW: The midget pathways circadian rhythm in blind primates. cells of the rabbit retina. Eur J Neurosci
of the primate retina. Doc Ophthalmol J Circadian Rhythms 2005; 3:1–5. 1999; 11:867–873.
2003; 106:67–81. 91. Marc RE: 1999 Mapping glutamatergic 110. Haverkamp S, Grunert U, Wässle H: The
73. Diller L, Packer OS, Verweij J, et al: L and drive in the vertebrate retina with a channel synaptic architecture of AMPA receptors at
M cone contributions to the midget and permeant organic cation. J Comp Neurol the cone pedicle of the primate retina.
parasol ganglion cell receptive fields of 1999; 407:47–64. Neurosci 2001; 21:2488–2500.
macaque monkey retina. J Neurosci 2004; 92. Marc RE: Kainate activation of horizontal, 111. Lindberg KA, Fisher SK: Ultrastructural
24:1079–1088. bipolar, amacrine and ganglion cells in the evidence that horizontal cell axon terminals
74. Dowling JE, Boycott BB: Organization of rabbit retina. J Comp Neurol 1999; are presynaptic in the human retina.
SECTION 3

the primate retina: electron microscopy. 407:65–76. J Comp Neurol 1988; 268:281–297.
Proc R Soc Lond B 1966; 166:80–111. 93. Vaney DI, Weiler R: Gap junctions in the 112. Setou M, Seog D-H, Tanaka Y, et al:
75. West RW, Dowling JE: Synapses onto eye: evidence for heteromeric, heterotypic Glutamate-receptor-interacting protein
different morphological types of retinal and mixed-homotypic interactions. GRIP1 directly steers kinesin to dendrites.
ganglion cells. Science 1972; 178:510–512. Brain Res Brain Res Rev 2000; Nature 2002; 417:83–87.
76. Wässle H, Boycott BB, Illing RB: 32:115–120. 113. Kamermans M, Fahrenfort I, Schultz K,
Morphology and mosaic of on and off beta 94. Kenyon GT, Marshak DW: Gap junctions et al: Hemichannel-mediated inhibition in
cells in the cat retina and some functional with amacrine cells provide a feedback the outer retina. Science 2001;
considerations. Proc R Soc Lond [B] 1981; pathway for ganglion cells within the retina. 292:1178–1180.
212:177–195. Proc Biol Sci 1998; 265:919–925. 114. Mills SL, Massey SC: Distribution and
77. Jusuf PR, Martin PR, Grunert U: Random 95. Marc RE: The structure of GABAergic coverage of A- and B-type horizontal cells
wiring in the midget pathway of primate circuits in ectotherm retinas. In: Mize R, stained with Neurobiotin in the rabbit
retina. J Neurosci 2006; 26:3908–3917. Marc RE, Sillito A, eds. GABA in the retina retina. Vis Neurosci 1994; 11:549–560.
78. Jacoby RA, Marshak DW: Synaptic and central visual system. Elsevier: 115. Mills SL, Massey SC: A series of
connections of db3 diffuse bipolar cell Amsterdam; 1992:61–92. biotinylated tracers distinguishes three
axons in macaque retina. J Comp Neurol 96. Dacheux RF, Raviola E: Horizontal cells in types of gap junction in retina. J Neurosci
2000; 416:19–29. the retina of the rabbit. J Neurosci 1982; 2000; 20:8629–8636.
79. Calloway EM: Cell types and local circuits 2:1486–1493. 116. Packer OS, Dacey DM: Synergistic
in primary visual cortex of the macaque 97. Raviola E, Dacheux RF: Variations in center-surround receptive field model of
monkey. In Chalupa LM, Werner J, eds. structure and response properties of monkey H1 horizontal cells. J Vision 2005;
The visual neurosciences. Cambridge: horizontal cells in the retina of the rabbit. 5:1038–1054.
MIT Press; 2004:680–694. Vision Res 1983; 23:1221–1227. 117. Wagner HJ, Wagner E: Amacrine cells in
80. Masland RH: Directional selectivity in 98. Nelson R: Cat cones have rod input: a the retina of a teleost fish, the road (Rutilus
retinal ganglion cells. In Chalupa LM, comparison of the response properties of rutilus): a Golgi stidy on differentiation
Werner J, eds. The visual neurosciences. cones and horizontal cell bodies in the and layering. Phil Trans Roy Soc Lond B
Cambridge: MIT Press; 2004:451–562. retina of the cat. J Comp Neurol 1977; 1988; 321: 263–324.
81. Yamada ES, Dmitrieva N, Keyser KT, et al: 172:109–136. 118. Pourcho RG, Goebel DJ: A combined Golgi
Synaptic connections of starburst amacrine 99. Fisher SK, Boycott BB: Synaptic connexions and autoradiographic study of (3H) glycine
cells and localization of acetylcholine made by horizontal cells within the outer accumulating amacrine cells in the cat
receptors in primate retinas. J Comp plexiform layer of the retina of the cat retina. J Comp Neurol 1985; 233:473–480.
Neurol 2003; 461:76–90. and rabbit. Proc R Soc Lond B 1974; 119. Vaney DI: The morphology and topographic
82. Dacey DM, Lee BB: The blue-ON opponent 186:317–331. distribution of AII amacrine cells in the cat
pathway in primate retina originates from a 100. Peichl L, González-Soriano J: retina. Proc Roy Soc (Lond) 1985;
distinct bistratified ganglion cell type. Morphological types of horizontal cell in 224:475–488.
Nature 1994; 367:731–735. rodent retinae: a comparison of rat, mouse, 120. Tauchi M, Masland RH: The shape and
83. Dacey DM, Liao HW, Peterson BB, et al: gerbil, and guinea pig. Vis Neurosci 1994; arrangement of the cholinergic neurons in
Melanopsin-expressing ganglion cells in 11:501–517. the rabbit retina. Proc R Soc Lond B 1984;
primate retina signal colour and irradiance 101. Perlman I, Kolb H, Nelson R: Anatomy, 223:101–119.
and project to the LGN. Nature 2005; circuitry, and physiology of vertebrate 121. Famiglietti EV Jr: Synaptic organization of
433:749–754. horizontal cells. In Chalupa LM, Werner J, starburst amacrine cells in rabbit retina:
84. Berson DM, Dunn FA, Takao M: eds. The visual neurosciences. Cambridge: analysis of serial thin sections by electron
Phototransduction by retinal ganglion cells MIT Press; 2004:369–394. microscopy and graphic reconstruction.
that set the circadian clock. Science 2002; 102. Boycott BB, Kolb H: The horizontal cells of J Comp Neurol 1991; 309:40–70.
295:1070–1073. the rhesus monkey retina. J Comp Neurol 122. Sandell JH, Masland RH: A system of
85. Hattar S, Lucas RJ, Mrosovsky N, et al: 1973; 148:115–140. indoleamine-accumulating neurons in the
Melanopsin and rod–cone photoreceptive 103. Kolb H, Mariani A, Gallego A: A second rabbit retina. J Neurosci 1986; 6:3331–3347.
systems account for all major accessory type of horizontal cell in the monkey retina. 123. Vaney DI: Morphological identification of
visual functions in mice. Nature 2003; J Comp Neurol 1980; 189:31–44. serotonin-accumulating neurons in the
424:76–81. 104. Kolb H, Fernandez E, Schouten J, et al: living retina. Science 1986; 233:444–446.
86. Tu DC, Zhang D, Demas J, et al: Are there three types of horizontal cell in 124. Sandell JH, Masland RH, Raviola E,
Physiologic diversity and development of the human retina? J Comp Neurol 1994; Dacheux RF: Connections of indoleamine-
intrinsically photosensitive retinal ganglion 343:370–386. accumulating cells in the rabbit retina.
cells. Neuron 2005; 48:987–999. 105. Wässle H, Dacey DM, Haun T, et al: The J Comp Neurol 1989; 283:303–313.
87. Lucas RJ, Hattar S, Takao M, et al: mosaic of horizontal cells in the macaque 125. Li W, Zhang J, Massey SC: Coupling
Diminished pupillary light reflex at high monkey retina: with a comment on pattern of S1 and S2 amacrine cells in
irradiances in melanopsin-knockout mice. biplexiform ganglion cells. Vis Neurosci the rabbit retina. Vis Neurosci 2002;
1590 Science 2003; 299:245–247. 2000; 17:591–608. 19:119–131.
Functional Anatomy of the Neural Retina

126. Rodieck RW, Marshak DW: Spatial density 146. Trexler EB, Li W, Massey SC: Simultaneous selective ganglion cell in the rabbit retina.
and distribution of choline acetyltransferase contribution of two rod pathways to AII J Comp Neurol 2003; 456:267–278.
immunoreactive cells in human, macaque, amacrine and cone bipolar cell light 165. Cajal SR: La rétine des vertébrés. La
and baboon retinas. J Comp Neurol 1992; responses. J Neurophysiol 2005; Cellule 1893; 9:119–257.
321:46–64. 93:1476–1485. 166. Polyak SL: The retina. Chicago: University
127. Springer AD, Hendrickson AE: 147. Saszik SM, Robson JG, Frishman LJ: The of Chicago Press; 1941.
Development of the primate area of high scotopic threshold response of the 167. Gastinger MJ, O’Brien JJ, Larsen NB,
acuity, 3: temporal relationships between dark-adapted electroretinogram of the Marshak DW: Histamine im-munoreactive
pit formation, retinal elongation and cone mouse. J Physiol 2002; 543:899–916. axons in the macaque retina. Invest
packing. Vis Neurosci 2005; 22:171–185. 148. Li W, Keung JW, Massey SC: Direct Ophthalmol Vis Sci 1999; 40:487–495.
128. Hendrickson A, Troilo D, Possin D, synaptic connections between rods and 168. Gastinger MJ, Barber AJ, Khin SA, et al:
Springer A: Development of the neural OFF cone bipolar cells in the rabbit retina. Abnormal centrifugal axons in
retina and its vasculature in the marmoset J Comp Neurol 2004; 474:1–12. streptozotocin-diabetic rat retinas. Invest
Callithrix jacchus. J Comp Neurol 2006; 149. Reid RC, Shapley RM: Spatial structure of Ophthalmol Vis Sci 2001; 42:2679–2685.
497:270–286. cone inputs to receptive fields in primate 169. Gastinger MJ, Bordt AS, Bernal MP,
129. Curcio CA, Sloan KR, Kalina RE, lateral geniculate nucleus. Nature 1992; Marshak DW: Serotonergic retinopetal
Hendrickson AE: Human photoreceptor 356:716–718. axons in the monkey retina. Curr Eye Res
topography. J Comp Neurol 1990; 150. Lee BB, Kremers J, Yeh T: Receptive fields 2005; 30:1089–1095.
292:497–523. of primate retinal ganglion cells studied 170. Gastinger MJ, Barber AJ, Vardi N, Marshak
130. Jeon CJ, Strettoi E, Masland RH: The with a novel technique. Vis Neurosci 1998; DW: Histamine receptors in mammalian

CHAPTER 122
major cell populations of the mouse retina. 15:161–175. retinas. J Comp Neurol 2006; 495:658–667.
J Neurosci 1998; 18:8936–8946. 151. Sun H, Smithson HE, Zaidi Q, Lee BB: 171. Witkovsky P: Dopamine and retinal
131. Remtulla S, Hallett PE: A schematic eye for Specificity of cone inputs to macaque function. Doc Ophthalmol 2004; 108:17–39.
the mouse, and comparisons with the rat. retinal ganglion cells. J Neurophys 2006; 172. Dacey DM: The dopaminergic amacrine
Vis Res 1985; 25:21–31. 95:837–849. cell. J Comp Neurol 1990; 301:461–489.
132. Szel IA, Rohlich P, Caffe AR, et al: Unique 152. Calkins DJ, Sterling P: Absence of 173. Nguyen-Legros J: Morphology and
topographic separation of two spectral spectrally specific lateral inputs to midget distribution of catecholamine neurons in
classes of cones in the retina. J Comp ganglion cells in primate retina. Nature mammalian retina. In: Osborne NN,
Neurol 1992; 325:327–342. 1996; 381:613–615. Chader G, eds. Progress in retinal research.
133. Juliusson B, Bergstrom A, Rohlich P, et al: 153. Dacey D, Packer OS, Diller L, et al: Center Oxford: Pergamon; 1988:113–147.
Complementary cone fields of the rabbit surround receptive field structure of cone 174. Mariani AP, Hokoc JN: Two types of
retina. Invest Ophthalmol Vis Sci 1994; bipolar cells in primate retina. Vision Res tyrosine hydroxylase-immunoreactive
35:811–818. 2000; 40:1801–1811. amacrine cell in the rhesus monkey retina.
134. Lukats A, Szabo A, Rohlich P, et al: 154. Williams DR, Hofer H. Formation and J Comp Neurol 1988; 276:81–91.
Photopigment coexpression in mammals: Acquisition of the Retinal Image. In 175. Oh SJ, Kim IB, Lee EJ, et al:
comparative and developmental aspects. Chalupa LM, Werner J, eds. The visual Immunocytological localization of
Histol Histopathol 2005; 20:551–574. neurosciences. Cambridge: MIT Press; dopamine in the guinea pig retina. Cell
135. Marc RE, Sperling HG: The chromatic 2004: 795–810. Tissue Res 1999; 298:561–565.
organization of primate cones. Science 155. Roorda A, Williams DR: The arrangement of 176. Zhang D-Q, Stone JF, Zhou T, et al:
1977; 196:454–456. the three cone classes in the living human Characterization of genetically labeled
136. Curcio CA, Allen KA, Sloan KR, et al: eye. Nature 1999; 397:520–522. catecholamine neurons in the mouse retina.
Distribution and morphology of human 156. Hofer H, Carroll J, Neitz J, et al: Neuroreport 2004; 15:1761-1765.
cone photoreceptors stained with Organization of the human trichromatic 177. Feigenspan A, Gustincich S, Bean BP,
anti-blue opsin. J Comp Neurol 1991; cone mosaic. J Neurosci 2005; Raviola E: Spontaneous activity of solitary
312:610–624. 25:9669–9679. dopaminergic cells of the retina. J Neurosci
137. Cornish EE, Madigan MC, Natoli R, et al: 157. Kolb H, Dekorver L: Midget ganglion cells 1998; 18:6776–6789.
Gradients of cone differentiation and FGF of the parafovea of the human retina: a 178. Xiao J, Cai Y, Yen J, et al: Voltage-clamp
expression during development of the study by electron microscopy and serial analysis and computational model of
foveal depression in macaque retina. Vis section reconstructions. J Comp Neurol dopaminergic neurons from mouse retina.
Neurosci 2005; 22:447–459. 1991; 303:617–636. Vis Neurosci 2004; 21:835–849.
138. Grünbaum B, Shephard GC: Tilings and 158. Brainard DH, Roorda A, Yamauchi Y, et al: 179. Hampson EC, Vaney DI, Weiler R:
patterns. New York: WH Freeman; Functional consequences of the relative Dopaminergic modulation of gap junction
1987:700. numbers of L and M cones. J Opt Soc Am permeability between amacrine cells in
139. Cook JE. Spatial regularity among retinal A 2000; 17:607–614. mammalian retina. J Neurosci 1992;
neurons. In Chalupa LM, Werner J, eds. 159. Xiao Y, Wang Y, Felleman DJ: A spatially 12:4911–4922.
The visual neurosciences. Cambridge: MIT organized representation of colour in 180. Ishida AT: Retinal ganglion cell excitability.
Press; 2004:463–478. macaque cortical area V2. Nature 2003; In: Chalupa LM, Werner J, eds. The visual
140. Cook JE: Spatial properties of retinal 421:535–539. neurosciences. Cambridge: MIT Press;
mosaics: an empirical evauation of some 160. Neitz J, Carroll J, Yamauchi Y, et al: Color 2004:422–450.
existing measures. Vis Neurosci 1996; perception is mediated by a plastic neural 181. Ruan GX, Zhang DQ, Zhou T, et al:
13:15–30. mechanism that is adjustable in adults. Circadian organization of the mammalian
141. Engstrom K, Ahlbert IB: Cone types and Neuron 2002; 35:783–792. retina. Proc Natl Acad Sci USA. 2006;
cone arrangement in the retina of some 161. Marc RE, Liu WL: Fundamental GABAergic 103:9703–9708.
flatfishes. Acta Zool 1963; 44:1–11. amacrine cell circuitries in the retina: 182. Wiechmann AF: Regulation of gene
142. Kalloniatis M, RE Marc: Interplexiform cells nested feedback, concatenated inhibition, expression by melatonin: a microarray
of the goldfish retina. J Comp Neurol 1990; and axosomatic synapses. J Comp Neurol survey of the rat retina. J Pineal Res 2020;
297:340–358. 2000; 425:560–582. 33:178–185.
143. Reese BE, Galli-Resta L: The role of 162. Dubin MW: The inner plexiform layer of the 183. Eldred WD, Blute TA: Imaging of nitric
tangential dispersion in retinal mosaic vertebrate retina: a quantitative and oxide in the retina. Vision Res 2005;
formation. Prog Ret Eye Res 2002; comparative electron microscopic analysis. 45:3469–3486.
21:153–168. J Comp Neurol 1970; 140:479–505. 184. Mills SL, Massey SC: Differential properties
144. Wong ROL, Godhino L: Development of the 163. Hennig MH, Funke K, Wörgötter F: The of two gap junctional pathways made by
vertebrate retina. In Chalupa LM, Werner J, influence of different retinal subcircuits AII amacrine cells. Nature 1995;
eds. The visual neurosciences. Cambridge: on the nonlinearity of ganglion cell 377:734–737.
MIT Press; 2004:77–94. behavior. J Neurosci 2002; 185. Kao YH, Lassova L, Bar-Yehuda T, et al:
145. Mariani AP: Biplexiform cells: ganglion cells 22:8726–8738. Evidence that certain retinal bipolar cells
of the primate retina that contact photo- 164. Dacheux RF, Chimento MF, Amthor FR: use both glutamate and GABA. J Comp
receptors. Science 1982; 216:1134–1136. Synaptic input to the on-off directionally Neurol 2004; 478:207–218. 1591
RETINA AND VITREOUS

186. Newman EA: Glial modulation of synaptic 190. Tian N, Copenhagen DR: Visual deprivation 194. Marc RE, Jones BW, Watt CB: Retinal
transmission in the retina. Glia 2004; alters development of synaptic function in remodeling: circuitry revisions triggered by
47:268–274. inner retina after eye opening. Neuron photoreceptor degeneration. In: Pinaud, et
187. Kawamura H, Sugiyama T, Wu DM, et al: 2001; 32:439–449. al, eds. Plasticity in the visual system: from
ATP: a vasoactive signal in the 191. Tian N: Visual experience and maturation of genes to circuits. Springer; 2005; 33–54.
pericyte-containing microvasculature of retinal synaptic pathways. Vision Res 2004; 195. Masland RH: The fundamental plan of the
the rat retina. J Physiol 2005; 44:3307–3316. retina. Nat Neurosci 2004; 4:877–886.
551:787–799. 192. Fisher SK, Lewis GP, Linberg KA, Verardo 196. Ochs M, Mayhew TM, Knabe W: To what
188. Zalutsky RA, Miller RF: The physiology of MR: Cellular remodeling in mammalian extent are the retinal capillaries ensheathed
somatostatin in the rabbit retina. J Neurosci retina: results from studies of experimental by muller cells? A stereological study in the
1990; 10:383–393. retinal detachment. Prog Retin Eye Res tree shrew Tupaia belangeri. J Anat 2000;
189. Johnson J, Caravelli ML, Brecha NC: 2005; 24:395–431. 196:453–461.
Somatostatin inhibits calcium influx into rat 193. Jones BW, Marc RE: Retinal remodeling 197. Chatterjee S, Callaway EM: Parallel
rod bipolar cell axonal terminals. Vis during retinal degeneration. Exp Eye Res colour-opponent pathways to primary
Neurosci 2001; 18:101–108. 2005; 81:121–244. visual cortex. Nature 2003; 426:668–671.
SECTION 3

1592
CHAPTER

123 Visual Acuity, Adaptation, and Color Vision


Maureen Neitz, Daniel G. Green, and Jay Neitz

We so readily use our eyes to organize and process information,


making it is easy to ignore the truly remarkable adjustments the
eye makes to enable us to see. When our interest focuses on an
object, such as a colorful bird, both eyes are directed so that an
image of the bird is focused on the foveas of both the retinas.
These two planar projections of the object are encoded and
transformed in the retina into elaborate patterns of neural
activity and relayed to various targets in the brain. As we orient
and move around, the retinal projection of an object, its
distance from us, its spectral content, and its intensity vary, and a
yet we continue to see the object as having the same shape, size,
color, and brightness.
The eye often is compared to a camera, and because each
shares common functions, this can be a useful starting point for
understanding visual processes. The eye and the camera both
have mechanisms for adjusting focus, setting exposure, and
storing an image. Yet the differences between vision and photo-
graphy are probably greater than the similarities. Photography is
a process that captures a permanent record of the variations in
intensity falling on a light-sensitive surface of the film. The
process has similarities only to events that occur in the earliest
stages of visual processing. The information we capture in a
photograph is equivalent to visualizing the pattern of activity
b
occurring at a particular instant in the mosaic of photoreceptors.
Thus, the photograph itself replicates only the simplest aspects
FIGURE 123.1. (a) Examples of visual acuity targets. The smallest
of vision. Without an eye to see it, a photograph is but a feeble detail is indicated by arrows. (b) Spatial relationships that define visual
shadow of the reality around us. When we look at a photograph, angle.
our eyes instantly see the color, form, and shape of objects from
the real world.
The biologic processes transforming the shower of photons 1.0 and 2.0 min–1. In conventional charts, with black patterns
falling on a mosaic of photoreceptors into the stable and invari- of various sizes on a white background, acuity is quantified in a
ant experience of sight have long been of immense scientific slightly different fashion. The letters on this chart have been
interest. This chapter touches on what we have discovered designed with the assumption that normal acuity corresponds
about those processes, the information our eyes make available to being able to resolve 1 min of arc (an acuity of 1.0 min–1).
to us, and the details of the psychophysics of acuity, adaptation, The size of each letter is such that its strokes will subtend
and color. 1 min of arc at a specified distance. The letter sizes can be
thought of as being designated by these distances (Fig. 123.1b).
VISUAL ACUITY This leads to the familiar fractional acuity notation, in which
the numerator of the fraction indicates the viewing distance and
Ordinarily, seeing refers to our ability to recognize forms and the denominator the size of the letter. An observer who from 20
patterns. An essential part of being able to see is the ability to ft away can just recognize the line with letters having strokes of
appreciate the fine detail in a scene. Visual acuity, the ability 1 min has a visual acuity of 20/20, an observer who requires
to resolve fine detail in a pattern, is usually determined by letters twice that size has a vision of 20/40, and so forth.
reducing the size of a test pattern until the smallest detail in the
pattern can just be resolved. Visual acuity can be expressed OPTICS
numerically in terms of the reciprocal of the size of the smallest
resolvable detail. The size is expressed as the angle that the The first stage of visual processing is the formation of an image
detail subtends at the eye of the observer. Figure 123.1a shows of the world on the mosaic of photoreceptors. Good vision
a Snellen letter and two other examples of acuity targets. Using depends on having a high-quality retinal image. Ideally, there is
such targets, visual acuity for normal observers ranges between only a single distance plane where a given object is brought to 1593
RETINA AND VITREOUS

sharpest optical focus. However, because we are tolerant of of receptors in the mosaic of foveal cones is just barely adequate
small amounts of optical blur, objects at a range of distances to reproduce the image of these targets, one can readily
appear to be in sharp focus1. To bring targets closer or farther appreciate that visual acuity is close to the limits set by the
than this into focus, the lens of the eye must change its focal retinal mosaic. Of the two factors, optics and retinal packing,
length through the process of accommodation. When the target which is of primary importance? A direct experimental answer
is moved closer than the range of accommodation, the image to this question has been obtained by using laser-generated
plane falls behind the retina, and fine detail begins to be blurred. interference fringes. Interference fringes, which are not images
Even when an object is in best focus, there is loss of image detail of objects but patterns resulting from the intrinsic wave proper-
owing to both aberrations and diffraction. These degradations in ties of light, are not degraded by the eye’s optics.5 Consequently,
the sharpness of the retinal image are potentially more serious it is possible to produce exceedingly fine high-contrast gratings
than focus errors because they are not correctable with the directly on the retina. Visual acuity is ~50% higher with
ordinary spherical and cylindrical lenses. The word aberration interference fringes (20/ to 20/10).4 Thus, the resolution limit
refers to a failure of the rays originating from a point source to set by the packing density of the cones is similar to, but slightly
be brought to a point focus. Inaccuracies and irregularities in higher than, the limit set by the optics of the eye. As a result,
the shapes of the curved refracting surfaces of the cornea and under ideal conditions, an observer with excellent vision can
lens produce aberrations. Also, there is chromatic aberration just resolve fine detail whose angular subtense approaches that
because the refractive properties of the eye’s dioptrics vary with of a single cone.6
wavelength, and different wavelengths are brought to focus at To transmit to the brain the information about fine detail
SECTION 10

different points. Chromatic difference of focus amounts to a that is available at the level of the photoreceptors, there must be
change of power of ~2 D over the 400–700 nm visible spectrum. at least as one ganglion cell for each cone. For the fovea, where
It has been suggested that for large pupils it is the dominant acuity is highest, there are between two and three ganglion cells
aberration that limits retinal image quality.2 Diffraction that transmitting information to the brain for every cone. A neural
occurs when light waves are abruptly truncated by an edge, such network compares the number of photons absorbed by a cone to
as the edge of the iris, also degrades the retinal image. As the the average number absorbed by its neighbors and projects the
result of interference between light waves and the edge of the result via two ‘private lines’ to the brain in the form of one
pupil, a point, for example, will be imaged onto the retina as a ON- and one OFF-center midget ganglion cell. These midget
fuzzy disk. The angular size of the disk varies inversely with ganglion cells are responsible for transmitting the information
pupil diameter. about fine details in the image.
The exact magnitude of the loss in image quality, resulting The response properties of the ganglion cells represent a first
from focus errors and aberrations, also depends on the size of level of processing of photoreceptor signals into visual percepts.
the pupil. Small pupils make diffraction effects worse; however, Each cone provides input to several types of ganglion cells each
as the pupil becomes smaller, depth of focus increases and a specialized to carry specific information about the visual
small pupil also tends to reduce the deleterious effects of stimulus. As shown in Figure 123.2, when part of the image of
aberrations by limiting the area of the optically imperfect a black horizontal stroke of the Snellen E falls on a cone, its
cornea and lens that are involved in producing the image. Over OFF-center ganglion cell fires, signaling the presence of a dark
the physiologic range of pupil sizes (2 to about 7 mm), the area in the image. When the image of the white background
balance between the effects of diffraction and those of between two strokes of the E falls on a cone, the ON-center
aberrations occurs when the pupil is ~3 mm in diameter, ganglion cell fires, signaling the presence of a light area of the
approximately the size that it tends to achieve under normal image. The ratio of ganglion cells to cones is greater than 2:1 in
bright light conditions.3,4 Under these conditions, the quality of the fovea because foveal cones connect to other types of
the retinal image is quite high and deviates only slightly from ganglion cells that collect signals from larger numbers of cones
an ideal system limited only by diffraction. and carry other types of information. There are ganglion cells
The density of packing in the mosaic of foveal cones is also that transmit the presence of a hue, for example, ‘blueness’, and
an important limiting factor. Each photoreceptor samples the other ganglion cells transmit information responsible for the
local intensity at a point in the retinal image. Consequently, the percept of movement.
size and the density of packing of the receptors must be
adequate if we are to appreciate the fine detail in the retinal RETINAL POSITION
image. The foveal cones are very thin and tightly packed,
making them particularly well suited to encode the fine detail in Visual acuity falls rapidly as the focal point moves away from
the image (Fig. 123.2a). The effect of the mosaic of foveal cones the fovea, as would be expected from the decrease both in
on vision is illustrated in Figure 123.2b. Acuity test patterns density of cone photoreceptors and in the relative number of
close to the limits of resolution have been drawn as a pattern of ganglion cells available to carry information from the retina.
stimulated and unstimulated foveal cones. Because the density The exact shape of the fall in acuity with eccentricity depends

FIGURE 123.2. (a) Section through cone inner


segments at the center of human fovea. The
bar indicates 2 min of arc (10 mm distance on
retina). The particular retina illustrated had the
lowest peak density of the four retinas studied.
(b) 20/20 Snellen letter drawn as a pattern of
stimulated and unstimulated cones.
(a) Reprinted from Curcio CA, Sloan KRJ, Packer O,
et al: Distribution of cones in human and monkey
retina: individual variability and radial asymmetry.
Science 1987; 236:579. Copyright 1987 American
a b Association for the Advancement of Science.
1594
Visual Acuity, Adaptation, and Color Vision

on the type of target used, but acuity falls roughly to half at 1° (1) any stimulus can be considered to be a sum of sinusoidal
and to one-fourth at 5°.7 To separate the optical factors from re- components, and (2) purely sinusoidal patterns are imaged by
tinal factors, one can use interference fringes formed directly on optical systems in a uniquely simple way. That is, the spatial
the retina. With the stimulus near the fovea, the fall in acuity variations in the image of a sinewave target are also sinusoidal,
for interference fringes parallels the fall in cone density.6 Beyond of the same spatial frequency but of reduced contrast. Thus, for
~5° of eccentricity, the rate of decrease is too great for which the any grating, a contrast reduction factor (and sometimes a phase
peripheral cone spacing happens to be the limiting factor. At shift factor) completely describes the object to image trans-
these eccentricities, the fall-off in resolution and the estimated formation. This number as a function of frequency defines the
decrease in spacing between ganglion cells seem to agree optical-transfer function.16 Because any pattern of luminances
reasonably well.8 can be described by a sum of sinewaves in two dimensions,
knowing how sinewaves are imaged (i.e., the transfer function),
CONTRAST it is possible to calculate the image that will be formed by any
arbitrary pattern. That is, the optical-transfer function com-
Although visual acuity is frequently used to characterize an pletely describes the imaging properties of the eye, and using the
individual’s ability to see, there is considerably more to func- transfer function it is possible to quantify the quality of an image
tioning in everyday life than being able to resolve fine detail in and to give a detailed account of how one or another factor
well-illuminated, high-contrast black-and-white patterns. In influences the image quality. Figure 123.3a shows measure-
the real world, one must detect and recognize a variety of targets ments of the optical-transfer function for an observer in good

CHAPTER 123
varying in contrast, size, and shape. focus with a range of pupil sizes. Because at 30 cycles/degree the
In recent years, we have increasingly come to appreciate the bars of the grating are of the same width as the strokes in the
importance of the effects of contrast on visual performance.4,9–15 20/20 letter; the curves show that near the ‘normal’ limit of reso-
To study the effects of contrast, an approach borrowed from lution, the image of a grating is reduced in contrast by ~50%.
optical engineering has been quite useful. The optical-transfer In an attempt to extend the idea of an optical transfer to the
function is frequently used to characterize the imaging abilities processing occurring in the retina and brain, the contrast-
of cameras and television systems. The idea of using the optical- sensitivity function has been evolved. To characterize the eye’s
transfer function stems from two key facts about linear systems: ability to process spatial information, the observer is presented

a b

FIGURE 123.3. (a) Optical-transfer functions for the in-focus eye of a normal observer at several pupil sizes (•, 2 mm; ▼, 2.8 mm; ■, 3.8 mm;
▲, 5.8 mm). (b) Contrast-sensitivity functions for 10 normal observers. Each point plots the contrast at which an observer could just detect the
sinewave grating.
(a) From Campbell FW, Green DG: Optical and retinal factors affecting visual resolution. J Physiol 1965; 181:576. (b) From Green DG: Visual acuity: The influence of
refraction and diffraction and the use of interference fringes. Int Ophthalmol Clin 1978; 18:21. 1595
RETINA AND VITREOUS

with a periodic pattern, usually a sinewave, and adjusts the con-


trast of the target until it can be just detected. The reciprocal of
the threshold contrast defines the contrast sensitivity. Contrast
sensitivity is then plotted as a function of the grating fineness,
with the fineness of the grating expressed in terms of the
number of bars in 1° of visual angle (cycles/degree). The shape
of the contrast-sensitivity function reflects, both the optical-
transfer function of the eye’s dioptric apparatus and the neural
processing of spatial frequency information.
Typical contrast-sensitivity functions are shown in Figure
123.3b. The contrast sensitivity, the reciprocal of the just-
visible contrast, peaks at an intermediate spatial frequency.
Gratings that are coarser or finer than this optimal frequency
require more contrast to be seen. The point where the contrast-
sensitivity function intersects the horizontal axis establishes
the highest spatial frequency that an observer can detect.
Because one cycle of a grating contains a dark and a light bar, a
30 cycles/degree grating has bars that subtend 1 min of arc and
SECTION 10

corresponds in size to the strokes in the 20/20 letter.


FIGURE 123.4. Visual acuity intensity functions under free viewing
Thus, in the same way that the optical-transfer function char-
conditions.
acterizes the imaging properties of a lens, the contrast-sensitivity From Pirenne MH, Denton EJ: Accuracy and sensitivity of the human eye. Nature
function describes a patient’s ability to see. In general, by con- 1952; 170:1039.
sidering the spatial frequency content of targets and knowing an
observer’s ability to process sinewave information at a variety of
spatial frequencies, one obtains information relevant to asses- light intensity increases, there is a changeover from vision
sing an observer’s functional visual capabilities. mediated by rods with high sensitivity but poor resolution to
Contrast sensitivity provides information that supplements vision mediated by cones with intrinsically better resolution.
what one can obtain from acuity measurements alone. For This switch from rods to cones frequently produces a sharp and
example, two patients can have the same acuity and yet have rather abrupt increase in the acuity-intensity relationship.
different middle- and low-frequency contrast sensitivities. The
differences in middle- and low-frequency sensitivity can have EYE MOVEMENT
dramatic effects on the patients’ visual performances. Moreover,
contrast-sensitivity loss not only can influence the detection of The eyes are continuously executing small involuntary move-
larger low-contrast targets but also seems to affect tasks that ments. The pattern of involuntary eye movements is a mixture
one might have thought would require only good acuity. It has of tremor, slow drifts, and rapid saccades. By analogy with a
been reported that patients with only slight losses in acuity but camera, one might think that any movement would be detri-
with losses in contrast sensitivity over a wide range of spatial mental. In fact, eye movements seem to be an essential prere-
frequencies can experience difficulties in reading.17,18 The expla- quisite to normal vision. If the retinal image is stabilized,
nation, in part, is provided by Rubin and Legge’s19 finding that within seconds the visual image fades.23,24 After a minute or so,
peak contrast sensitivity is an important determinant of how only a very blurred, cloudy version of the original scene persists.25
rapidly low-vision observers can read letters. On the basis of These subjective reports of the appearance of stabilized images
these and other studies, it seems likely that contrast-sensitivity might suggest that high-frequency information disappears more
determinations will continue to be useful in assessing func- quickly than coarser detail, but systematic measurements with
tional visual capacities, and in predicting such things as the gratings seem to suggest the reverse.26 The key point is that for
benefit that might be derived from a particular low-vision aid. vision to be possible, there have to be eye movements. The eye
movements move the retinal image, and as a result, the photo-
INTENSITY receptors are subjected to continually changing spatial and
temporal transients.
Our ability to see is strongly dependent on having adequate
light. At the lowest light levels, a few quanta absorbed in an area LIGHT AND DARK ADAPTATION
containing 500 rods can produce a visual sensation. At these
levels, some semblance of form vision is possible, but acuity is We are largely unaware of the great variations in illumination
exceedingly low. Even at an intensity of ~10–5 cd/m2, which is that occur in the real world because the eye light-adapts. By
about a log unit above absolute threshold, visual acuity is only increasing (or decreasing) sensitivity when the environmental
~20/1000.20 As luminance of the target is increased, the ability intensity increases (or decreases), our perception of the visual
to resolve details continuously improves up to luminances of world remains relatively constant. The term light adaptation is
~10 cd/m2, where it reaches a plateau (Fig. 123.4).21 A number used to describe the changes in visual sensitivity produced by
of factors contribute to the dependence on target luminance. steady background lights. An everyday example of this change
With the dimmest targets, vision is mediated by rods 4° or more is the disappearance of the stars with the coming of dawn. This,
from the fovea.22 As stimulus intensity increases, there is move- of course, is not due to a change in the amount of light emitted
ment of fixation from the periphery toward the fovea. At lowest by the stars but rather is a result of the desensitization produced
levels of illumination, where the stimuli are so weak that not by the veil of scattered light in the sky. Dark adaptation is the
every receptor will absorb a quantum, the quantal nature of the recovery of sensitivity with time after exposure to a background
stimulus can severely limit the eye’s ability to appreciate details that reduces sensitivity. Light adaptation works well in the
and contrast. Smaller pupil sizes that accompany higher lumi- natural world because we are attempting to sense objects around
nances can also contribute improved vision because the quality us that are not self-luminous but rather are illuminated from a
1596 of the retinal image with a fully dilated pupil is not optimal. As distant source and seen by reflected light (Box 123.1).
Visual Acuity, Adaptation, and Color Vision

Increment thresholds and dark adaptation are the two classic


adaptation paradigms that have been used to reveal the changes in
sensitivity occurring in the eye. In both, the dependent measure
is usually an observer’s ability to detect a small, briefly presented
test flash. To determine increment thresholds, the small incre-
mental test stimulus is usually presented on a spatially uniform
background field, which is systematically varied in intensity.

BOX 123.1 Adaptation and Weber’s law


If E is the intensity of the illumination locally, and r(x,y) is the
spatial variation in the reflectivity of the objects in a scene, the
equation L = r(x,y)E gives the point-by-point variations in the
luminance from particular region of the scene. The information
extracted by the visual system from the image on the retina is an
impression of r(x,y), which is relatively independent of E. The eye
does this by adjusting its sensitivity according to Weber’s law,
which states that sensitivity varies inversely with E, the intensity

CHAPTER 123
of the illumination. When E increases by, say, a factor of 10, the
retinal gain decreases by this same factor, and consequently the
neural signal, which depends on the product of retinal
illumination by retinal gain, remains unchanged. FIGURE 123.5. Increment threshold functions. The lower limb is for
rods, and the upper limb is for cones.

LIGHT ADAPTATION
In controlled laboratory conditions, one typically finds that DARK ADAPTATION
when a small test flash is added to steady background over a It takes time for the threshold to reach a new equilibrium value
considerable range of background intensities, the intensity of after a background abruptly changes from one intensity to
the just-detectable increment is approximately proportional to another. The recovery of sensitivity in the dark after prior expo-
background intensity. This property, that the ratio of the test sure to a bright stimulus is called dark adaptation. The speed
flash intensity to the background intensity is roughly constant, with which a new equilibrium is reached depends on the direc-
is Weber’s law relationship (Box 123.1). There are, however, tion and magnitude of the change. In general, reduction in
failures at both ends of the scale, when the background is either sensitivity occurs quickly relative to restoration of sensitivity.
very bright or very dim. At the low end, Weber’s law fails when That is, it takes a matter of seconds to adapt to a brighter back-
the background becomes so dim that it no longer affects sen- ground, whereas adjustment to decreases in background inten-
sitivity. At the other extreme, a bright background can overload sity is slower. In particular, the recovery to total darkness may
the system. If the background is sufficiently intense, a new proceed very slowly. After exposure to a bright stimulus, the
phenomenon called saturation occurs. That is, in order to be exact time needed to adjust to a dimmer background depends
seen, the increment needs to be made considerably brighter than on whether rods or cones are being tested and on the intensity
one would predict from Weber’s law.27 In addition, increment of the prior exposure. In the extreme situation, in which one is
threshold curves frequently have a discontinuity. Rod signals plunged from a very bright environment into complete
mediate low-intensity vision, and cones subserve the upper darkness, it can take up to an hour for rod sensitivity to recover
range of intensities. As a result, plots of increment threshold as fully. Measuring the dark-adaptation curve is the standard
a function of intensity yield curves that frequently are divisible method for tracking this recovery process. One plots the thres-
into two distinctly different portions, with a kink in the curve hold as a function of time after the termination of the condi-
marking the changeover from predominantly rod to predomi- tioning light stimulus. The shape of the dark-adaptation curve
nantly cone control of sensitivity (Fig. 123.5). depends on the test stimulus parameters, such as size, color,
The foregoing description of the behavior of the visual system and retinal location, as well as conditioning stimulus para-
during adaptation says little about what is happening in the eye meters, such as its intensity, duration, and color. Fortunately, to
during these changes. Because exposure to light bleaches visual a great degree these multiple factors can be reduced to just two
pigment, the simplest possible mechanism imaginable to account principal determining variables. These are the extent to which
for losses in sensitivity would be a decrease in the potency of the the test light stimulates rods and cones and the quantity of
test probe caused by the direct removal of visual pigment. This, visual pigment that has been bleached by the exposure to the
however, is not the case. Many years ago, Rushton28 showed conditioning stimulus (see Box 123.2).
that a background can be so dim that only a few rods can absorb If the size, color, and retinal location are arranged so that the
photons and yet significantly elevate threshold. Only one rod in test flash stimulates both rods and cones, and significant
50 needs to absorb a photon of light for the threshold of a amounts of rod and cone pigment are bleached by the condi-
stimulus that falls on the receptors not directly affected by the tioning stimulus, then dark adaptation proceeds in two distinct
background to be elevated by a factor of four. Even at back- phases. In the first phase or branch, typically lasting ~5–10 min,
ground levels where rods saturate, only small amounts of visual the threshold is determined by the cones as they recover their
pigment are bleached. sensitivity. The time course of the process parallels the regen-
Cones may be a bit different in this regard. It is not com- eration of cone pigment. Later, the rods recover sufficiently for
pletely clear to what extent backgrounds can desensitize the their thresholds to be lower than those of the cones, and they
photoreceptors themselves. In addition, with cones the adapting mediate threshold, giving rise to a rod branch in the curve.
backgrounds can bleach pigment, and so the depletion of visual Complete recovery of rod sensitivity takes as long as it takes for
pigment may contribute to sensitivity loss in cones at high rod pigment to regenerate. During this time, sensitivity can
light levels. increase by a factor of 10 000 (Fig. 123.6). 1597
RETINA AND VITREOUS

COLOR VISION
Humans value color perception highly as a sensory capacity.
Few of us would accept a black and white television or
monochrome computer monitor in place of color ones even in
exchange for large savings in cost. One reason color is
important to us is that it has a powerful effect on our emotions.
This is presumably, in part, because some components of our
color vision system are evolutionarily ancient, predating other
sensory capacities. Color cues are associated with time of day,
the season, and an organism’s position and orientation in
space. Color can signal the presence of an injury or illness, the
presence and or quality of food, and the identity of a mate. In
our modern world color-coding is extremely important in
transmitting information visually. Objects identified by color
among a large number of distracters can be located nearly
instantly in visual search and color is invaluable in perceptual
grouping and segmenting objects.
SECTION 10

With regard to mechanism, color vision is based on three


FIGURE 123.6. A typical dark-adaptation curve. The first limb of the types of cone photoreceptors which are the basis for all vision
curve reflects recovery of cones, and the second, slower limb reflects with the exception of vision under very dim light conditions,
rod recovery. which is dependent on rods. Information about pattern, lumi-
nance, and color are all extracted from a mosaic of three types
of cone, one class most sensitive to short wavelength light (S),
BOX 123.2 Adaptation and the Dowling–Rushton Law
a second class (M) most sensitive to middle wavelength light and
The Dowling–Rushton Law is the empirical relationship between
a third (L) most sensitive to long wavelength light (Fig. 123.7).
bleached pigment and sensitivity. Rather than being linear it is, to
a good approximation, logarithmic, given by the equation Perception of black, white, and gray, the hues of red, green, blue,
L It = a(1 – r) where It is threshold, r is the proportion of pigment, and yellow and their patterns in the retinal image are extracted
and a is a constant. by different types of ganglion cells each specialized to carry
specific information about the visual stimulus from one mosaic
of cones.

a b

FIGURE 123.7. (a) Illustration of the arrangement of the three types of cones, L-, M-, or S-, which represent classes of photoreceptors that are
primarily sensitive to long wavelength light (L), middle wavelength light (M), and short wavelength light (S) within the visible spectrum. In this
diagram the cones are colored blue, green, or red in order to represent the different photoreceptor classes. S cones represent a minority, ~5% of
the total. The arrangement in a trichromat is very close to random for L and M cone classes. Midget ganglion cells have center surround-
receptive fields with the center derived from a single cone (for example, within the inner ring of the two black concentric circles). The six
adjacent cones are the most important contributors to the surround (for example, the cones within the outer ring of the two black concentric
circles). In a normal trichromat the surrounds of many L or M cones will have some cones of a different type than the center. Thus most midget
ganglion cells have spectrally opponent responses to diffuse red and green lights; they are either excited by red and inhibited by green or vice
versa. (b) Arrangement of cones in a deuteranope, a dichromat, with only two different cone classes L and S. In a dichromat, usually all the
cones in the surround are of the same class as the center input to a midget ganglion cell. Such ganglion cells do not respond to diffuse colored
1598 lights; they are specialized to signal patterns of light and dark on the retina.
Visual Acuity, Adaptation, and Color Vision

A great deal is known about the neural types and their inter- which is the most common of all human single locus genetic
connections in the retina responsible for luminance, color, and disorders. The increasing proportion of mutant cone pigment
form, and there is a growing body of information about the genes is also presumably the root of a growing number of other
higher visual centers; yet, how these operate to give us vision problems and complications of human vision.
remains a fascinating puzzle. Clues toward solving the puzzle
come from information about the evolution and development of INHERITED RED-GREEN COLOR VISION
visual system and about its anatomy and physiology. In contrast DEFICIENCY
to our persisting ignorance about color vision circuitry, the last
20 years has seen an explosion of information about the cone Inherited color vision deficiency occurs at an extraordinarily
photopigments. Many of the long standing questions about these high frequency in human populations although the prevalence
pigments and their role in normal vision and vision disorders varies with ethnicity and race.29 Caucasians exhibit among the
have now been answered. In humans there are three types of highest rates with 7–8% of males affected, and native Fijians
cone photopigment, one for each class of cone (see Box 123.3). have the lowest rate with 0.82% of males affected, while
The L and M cone opsins and the genes that encode them are Japanese and Africans have intermediate rates with 4.17% and
unusually variable, presumably due to the unstable tandem 2.61% of males affected, respectively. Compared to many other
arrangement of the genes and their unique evolutionary history. common inherited recessive disorders, such as cystic fibrosis
Among the mammals, red-green color vision first evolved in a and sickle cell anemia, color vision deficiency is unusual in
primate ancestor. Strong selective pressure favoring trichromatic occurring at an exceptionally high frequency with no compelling

CHAPTER 123
color vision acted on primates in the wild, minimizing the evidence of a strong heterozygote advantage to explain why. For
prevalence of mutant L and M opsin gene arrays despite the example, cystic fibrosis is the most-common life-limiting auto-
extreme instability inherent in tandemly duplicated genes. somal recessive disorder among humans, estimated to occur at
However, in humans, selection against mutant X-linked pig- a rate of about one in 3200 live births.30 Heterozygotes for cystic
ment genes has been relaxed. The variability that has resulted fibrosis are protected against heat- and disease-induced
includes gene arrangements responsible for color blindness, dehydration, thereby providing them with a survival advantage
over the history of human existence. Another example is sickle
cell anemia in which heterozygotes are protected against the
severe pathogenesis of malaria.30 The answer to why color
BOX 123.3 Normal color vision: terms and genetics
vision deficiency is so prevalent in human populations lies in
Normal human color vision is trichromatic, mediated by three
the evolutionary origin and arrangement of the OPN1LW and
well-separated classes of cone photoreceptor commonly referred
to as the blue, green, and red cones. Calling them short-,
OPN1MW genes in the human genome, and on the strength of
middle-, and long-wavelength sensitive, abbreviated S, M, and L natural selection on trichromatic color vision.
cones minimizes confusion that can arise from giving them color Genetic evidence indicates that all vertebrate opsin genes
names. evolved from a common ancestor through a process of gene
Photopigment molecules within each cone are responsible divergence and duplication.31 Most mammals have two types of
for the spectral properties of the cones. Each photopigment cone photoreceptor, one maximally sensitive to short-wave-
molecule is composed of two parts; a protein termed the opsin, length (S) or in some cases ultraviolet (UV) light and another
and an 11-cis-retinal chromophore. maximally sensitive to light in the middle-to-long wavelengths
The official names for the genes encoding the L, M, and S
(M/L).32 The opsin components of the photopigment molecules
cone opsins are OPN1LW, OPN1MW, and OPN1SW respectively.
that determine the spectral properties of the cones are encoded
Both OPN1LW and OPN1MW are on the X-chromosome at
position Xq28, OPN1SW is located on chromosome 7 at 7q32.1. by an autosomal gene in the case of the S or UV opsin, and a
The location of OPN1LW and OPN1MW on the X-chromosome gene on the X-chromosome in the case of M/L opsin. Together,
accounts for the great gender difference in the prevalence of the two cone types form the basis for dichromatic color vision.
color vision deficiencies. In New World primates, trichromatic color vision was acquired
through evolution of allelic diversity in the X-chromosome
opsin gene, which produced variety in spectral sensitivity of the
encoded photopigments.33 Males have only one X-chromosome,
BOX 123.4. Red-green color vision deficiency: but females have two. In female New World monkeys who are
classification and terminology heterozygous at the X-chromosome opsin gene locus,
Red-Green color vision defects fall into two categories X-inactivation segregates expression of the alleles into separate
depending on which cone class does not contribute to color populations of cones, producing three cone types. The heterozy-
vision. The noncontributing cone class is indicated by the gous females have trichromatic color vision,34 indicating that
prefixes: they have all of the components necessary to form fully func-
Protan- for absence of L cone contribution to vision. tional circuits for trichromatic color vision. In Old World
Deutan- for absence of M cone contribution to vision. primates, trichromatic color vision arose via a gene duplication
Further categorization of color vision defects depends on that placed two opsin genes together in tandem on the
whether the remaining color vision is based on two (dichromacy)
X-chromosome.31 X-inactivation cannot segregate expression of
versus three (anomalous trichromacy) spectrally distinct types of
cones. The suffix -opia denotes dichromacy. The suffix -anomaly
tandem OPN1LW and OPN1MW genes into separate popula-
denotes anomalous trichromacy in which two of the cone classes tions of cones; however, a critical enhancer known as the locus
are more similar in spectral sensitivity than the corresponding control region (LCR) was not duplicated along with the opsin
normal cones: gene, thereby limiting the photoreceptor to expressing one opsin
Deuteranopia. color vision mediated by L and S cones. gene at a time. 35–38 Ultimately, in the adult cone photoreceptor,
Protanopia. color vision mediated by M and S cones. only one opsin gene is expressed to the exclusion of all others.39
Protanomaly. color vision mediated by S and two spectrally Tandemly duplicated genes are inherently unstable because
distinct classes of M cone. they are prone to unequal homologous recombination between
Deuteranomaly. color vision mediated by S and two misaligned arrays during meiotic cell division in females, which
spectrally distinct classes of L cones. produces gene rearrangements that underlie inherited color
vision deficiency,31 as illustrated in Figure 123.8. Natural selection 1599
RETINA AND VITREOUS

Parental arrays

Recombinant arrays

Normal Color Vision Proton-type color Deutan-type color


Deutan-type color Deuteranope
vision defect Protanope S L M vision defect vision defect
S M S L
S L L S M M S L L

photoreceptors

Trichromat Dichromat
Dichromat
Dichromat or Dichromat or
Dichromat or
anomalous trichromat anomalous trichromat
anomalous trichromat

a b c

FIGURE 123.8. The variety of normal and defective color vision phenotypes in humans is produced by unequal homologous recombination
during meiotic cell division in females. Red and green arrows represent the OPN1LW and OPN1MW genes, respectively; arrows that are half red
and half green represent chimeric genes produced by a crossover between an OPN1LW and an OPN1MW gene. The color of the arrowhead
indicates whether the gene encodes an L pigment (red) or M pigment (green). (a) A crossover between an OPN1LW and OPN1MW in the
parental arrays produces two recombinant arrays. Only the two 5„ genes (left-most) in the three gene array are expressed and both of these
SECTION 10

encode L-class pigments. The array produces dichromatic color vision if the encoded L-class pigments have identical spectral properties, or it
produces anomalous trichromacy if the pigments differ in spectral properties. (b) A crossover in the region between the genes in one array and
the region downstream of the last gene in another array produces a three gene array that encodes an L and an M pigment and thus will produce
normal color vision. The other recombinant array contains a single gene, which encodes an L opsin and thus produces dichromatic color vision.
(c) Recombination between an OPN1MW in a three gene array and OPN1LW in a two gene array gives rise to two recombinant arrays, both of
which give rise to color vision defects. One array will have two genes, an OPN1LW/OPN1MW hybrid followed by an OPN1MW gene, which if
both genes encode pigments identical in spectral properties will cause dichromacy, or if the encoded pigments differ in spectral poperties, the
array will cause anomalous trichromacy. Likewise the array with an OPN1LW gene followed by an L-pigment encoding chimeric gene will either
cause dichromacy or trichromacy if the encoded pigments are identical or different in spectral properties, respectively.

is expected to virtually eliminate visual pigment gene arrays CAUSES OF COLOR VISION DEFICIENCY
that confer color vision defects; however, if selection is relaxed, A common misconception is that inherited red-green color
then arrays causing color vision defects can accumulate in the vision deficiency is a single entity; it is not. Instead it is a group
population giving rise to an increase in color vision deficient of disorders that can be dichotomized at the first level according
males and in female carriers. In the US, about one in 12 males to what is missing to cause the perceptual loss, and at a second
is affected by red-green color vision deficiency and one in seven level according to the degree of color vision that remains
females is a carrier. (Box 123.4). The most common cause of color vision deficiency
When color defective arrays initially began to accumulate in is the deletion of all OPN1LW (protan defects) or all OPN1MW
the population, female carriers would have had one normal (deutan defects) genes. The degree to which color vision is
array with one OPN1LW and one OPN1MW gene and another impaired is determined by the spectral properties of the
array that either had one or three opsin genes (Fig. 123.8a,b). In pigments encoded by the genes that remain.
either case, the unequal number of opsin genes on the two
X-chromosomes produces instability because there is no perfect
alignment of the two arrays during meiotic cell division. Recom- DICHROMACY
bination between two arrays with different numbers of opsin The most severe of the common inherited red-green color vision
genes will give rise to variability in the number of opsin genes defects are the dichromacies, protanopia and deuteranopia in
per X-chromosome and an increase in the prevalence of chimeric which color vision is mediated by just two pigments in two types
genes resulting from intermixing of OPN1LW and OPN1MW of cone. Protanopia and deuteranopia (Box 123.4) each occur at
genes (Fig. 123.8c). As will be discussed below, the diversity a rate of ~1% in Caucasian males, and although dichromacy is
introduced by intermixing L and M opsin gene sequences much rarer in females, about one in 4000 females is affected.
underlies the variety of phenotypes associated with anomalous In most cases, the direct cause of dichromacy is the deletion
trichromacies. of the genes that encode one class of cone photopigment from
Only among humans is there widespread variability in the the X-chromosome through unequal homologous recom-
number of visual pigment genes on the X-chromosome with bination31,44–47 as illustrated in Figure 123.8a,b. For example, in
a high frequency of arrays containing more than two opsin a recent study 53 of 55 protanopes lacked genes for L opsin, and
genes.31,40 Since normal trichromatic color vision requires expres- 51 of 73 deuteranopes lacked genes for M opsin.47
sion of only one L and one M opsin gene, this raises the ques-
tion of whether the extra genes beyond the necessary two are
expressed. Experiments have shown both by inference41 and by PROTANOPIA
direct analysis of opsin gene expression in human retinas from One common cause of protanopia is the deletion of all but one
color deficient donors42 that usually only the two opsin genes at opsin gene on the X-chromosome with the one remaining gene
the 5’ end of the X-chromosome opsin gene array are expressed, encoding an M-class pigment (Fig. 123.8a).31,48–49 Another com-
although exceptions have been observed.43 Thus, the order of mon gene arrangement among protanopic men is an array that
the genes in the array on the X-chromosome and the spectral lacks all OPN1LW genes, and that has a chimeric OPN1LW/
sensitivity of the photopigments encoded by the expressed genes OPN1MW gene in the first position and an OPN1MW gene in
play a central role in determining color vision phenotype. the second position (Fig. 123.8c).46,47 For protanopes with this
1600
Visual Acuity, Adaptation, and Color Vision

arrangement, the chimeric gene and the OPN1MW gene do not small spectral separation on which the person must rely to make
encode photopigments that differ in spectral properties, color discriminations, there appear to be a variety of other fac-
accounting for the protanopic phenotype.47 tors including physiological factors, personality factors, differ-
Occasionally, protanopes who have an apparently intact ences between naïve and experienced observers, and differences
OPN1LW gene have been identified31,46,47 and presumably the in the relative ratios of the underlying cone photoreceptors to
gene is either not expressed or does not encode a functional name a few, that contribute to variability in phenotype.
photopigment. In cases where this hypothesis has been tested In summary, the most severe red-green color vision defects,
experimentally, the OPN1LW gene has either been found to be the dichromacies, are commonly explained by the straight-
displaced to the 3’ end of the array where it is not expressed,46,47 forward deletion of cone opsin genes. Another relatively com-
or it has been found to carry a particular deleterious com- mon cause is a point mutation that disrupts the function of the
bination of amino acids at polymorphic positions encoded by encoded opsin. Recent evidence indicates that there is a funda-
exon 3. The same combination was observed in OPN1MW mental difference in the effects on the cone mosaic that results
genes in deuteranopes where its effect on the cone mosaic and from these two mechanisms for color vision deficiency, specifi-
its contribution to dichromacy has been examined in more cally with regard to what happens to the subpopulation of cones
detail, as will be described below under deuteranopia. that do not contribute to color vision. In the case of the single-
gene dichromat, it appears that all of the cones that would have
become L or M cones express the available X-chromosome
DEUTERANOPIA opsin gene and so no cone photoreceptors are lost. In contrast,

CHAPTER 123
The majority (about two-thirds in some studies) of deuteranopes evidence indicates that in dichromats with two or more opsin
have undergone a deletion of all X-chromosome opsin genes genes in which the first or second gene encodes an opsin with
except for one remaining OPN1LW gene, accounting for the an inactivating amino acid substitution(s), a subpopulation of
phenotype.31,44,50,51 Nearly one-third of deuteranopes have an photoreceptors express the mutant gene, which ultimately results
OPN1MW gene; however quite often they have a chimeric in the death of the photoreceptor, giving rise to a lower than
OPN1LW/OPN1MW gene inserted between the OPN1LW and normal cone density and leaving gaps in the cone mosaic.60 What
OPN1MW genes, displacing the OPN1MW gene to a non- occupies the gaps left by the absent cones remains unknown.
expressed position.31,52 Another relatively common cause of
deuteranopia is the presence of an inactivating mutation in the
OPN1MW.47,53 By far the most common inactivating mutation ANOMALOUS TRICHROMACIES
found in OPN1MW genes is a nucleotide change that results in The milder forms of red-green color vision deficiencies are the
the substitution of arginine for a highly conserved cysteine at anomalous trichromacies. As the term for their condition
position 203 (C203R) of the cone opsin molecule, preventing the implies, affected individuals have trichromatic color vision, but
opsin from folding properly.54,55 Other inactivating amino acid it is not based on L, M, and S pigments like normal color vision.
substitutions have also been found but most are quite rare.47 Classical descriptions of anomalous trichromacy postulated the
Perhaps the most interesting inactivating mutation found in existence of ‘anomalous pigments’ such that in addition to S
OPN1MW, which as was alluded to above has also been found cones, protanomalous individuals are said to have normal M
in OPN1LW genes, is a combination of amino acids at normally and anomalous L pigments while deuteranomalous individuals
polymorphic positions. OPN1LW and OPN1MW have been are said to have normal L and anomalous M pigments. Results
intermixed by recombination so that in the present day of molecular genetic analyses have provided insight into what
population of humans with normal color vision, there are 11 the anomalous pigments are, and as a consequence it has become
dimorphic positions among L and M pigments. As a result, clear that the classical concept of the ‘anomalous pigment’ is
there is tremendous variation in the amino acid sequences of unbefitting.52 For example, ‘anomalous L’ pigments are often
the L and M cone photopigments found in humans with normal indistinguishable from normal M pigments in spectral sensi-
color vision.56–59 A specific combination of amino acids at the tivity and in amino acid sequence, and there is similar overlap
dimorphic positions has been observed to always be associated between ‘anomalous M’ pigments and normal L pigments.51,52
with a color vision deficiency in which there is a perfect The ‘anomalous pigments’ have been generated by recombi-
correlation between the opsin with the deleterious combination nation between the ancestral OPN1LW and OPN1MW genes;
and the absence of function of the corresponding cone. High however, over evolutionary time, multiple rounds of recombina-
resolution adaptive optics imaging of the retina of a deuter- tion have intermixed the ancestral pigment genes so thoroughly
anope, whose OPN1MW gene specified an M pigment with the that among modern human males with normal color vision,
deleterious combination, was shown to have gaps in his cone there is a family of photopigments specified by the OPN1LW
mosaic, presumably where M cones had once been.60 In addi- genes that differ in amino acid sequence and in the wavelength
tion, the deuteranope was shown to have a reduction in cone of peak sensitivity (Fig. 123.9). Similarly, there is a family of
density by about one-third. When the gaps in his mosaic were pigments encoded by the OPN1MW genes found in color
modeled as M cones and cone density recalculated taking into normal individuals (Fig. 123.9). The variant forms of the L and
account the modeled cones, the density estimate was normal. M photopigments underlying normal color vision overlap with
Taken together, these observations support the hypothesis that the forms that correspond to what were classically termed the
in some forms of color vision deficiency, the cause is a loss of ‘anomalous pigment’. Referring to the photopigments underlying
photoreceptor cells that is due to a malfunction in the produc- anomalous trichromacy according to their spectral sensitivities
tion or function of the photopigment. promotes a clearer understanding of the cause of the differences
Males who have opsin gene arrays in which the first two between normal versus anomalous trichromacy.
genes encode photopigments of the same functional class but The genes can be categorized as encoding an L-class or an M-
with a difference in peak sensitivity of less than 2.5 nm, occa- class pigment by the sequence of exon 5. Whether the encoded
sionally perform as dichromats on standard color vision tests, pigment will have peak sensitivity near 560 nm or near 530 nm
including the anomaloscope color matching tests.47 Thus, their is determined by the amino acids at two of the polymorphic
performance is worse than would be predicted strictly by the amino acid positions encoded by exon 5 (Fig. 123.10).61–63 Amino
complement of opsin genes they have. When there is a very acids encoded by five other polymorphic positions encoded by
1601
RETINA AND VITREOUS

red-to-green region of the visible spectrum that is enjoyed by


people with normal color vision. Individuals with deuteranomaly
lack M cones but they have two distinct classes of L-cone that
are different enough in spectrum to provide the basis for limited
color vision in the red-green region of the spectrum.62–64 Like-
wise, individuals with protanomaly lack L cones but they have
two different classes of M-cone. An anomalous trichromat
with a large spectral difference between the L or M cone sub-
types has the basis for much better color vision than a person
with two cone subtypes that are nearly identical.44,51,64–65 Spectral
separations of 5 nm or larger give rise to only a very mild color
vision deficit,64 and affected individuals often perform as nearly
normal in standard color vision tests. When the underlying
FIGURE 123.9. During human evolution, recombination has
intermixed the OPN1LW and OPN1MW genes so thoroughly that there pigments are separated in peak sensitivity by between 2.5 and
is not just one L- and one M-pigment encoded. Instead, the OPN1LW 5 nm, color vision is more than mildly impaired, but is none-
and OPN1MW genes found in males with normal color vision encode theless quite excellent compared to smaller separations. When
a family of L-cone photopigments (L-class) and a family of M-cone color vision in the red-green region of the spectrum is mediated
photopigments (M-class). The OPN1SW gene has not been observed by cones that differ in peak sensitivity by fewer than 2.5 nm,
SECTION 10

to vary, and hence among humans there is only one know functional color discrimination is quite impaired, and some affected indi-
S-cone photopigment. viduals’ performance on standard color vision tests is indistin-
guishable from the performance of a dichromat.64
C-terminus
PROTANOMALY
Protanomalous individuals usually have a rearranged visual
pigment gene locus in which the first gene is an
OPN1LW/OPN1MW chimera with exon 5 derived from the
233
parental OPN1MW gene and thus it encodes a pigment of the
277 M class. The chimeric gene is followed by an OPN1MW gene
230 (Fig. 123.8c). Amino acid sequence differences between the
chromophore M-class pigments encoded by the two genes produce a spectral
65 180
285
attachment site difference between them, enough to support a small degree of
312
309 trichromatic color vision (Fig. 123.10).51,52,64
116

dimorphic sites:
DEUTERANOMALY
spectral tuning sites:
Long (Y277, T285) vs middle Deuteranomalous individuals usually have a rearranged visual
N-terminus
(F277, A285)-wave determining pigment gene locus so that the first gene is a normal OPN1LW
sites
gene, which is followed by a chimeric gene that is an
FIGURE 123.10. Spectral tuning of the L and M cone photopigments OPN1MW/OPN1LW gene that was produced by unequal homo-
is achieved through amino acid substitutions at a limited number of
logous recombination and that encodes an L-class photopig-
positions. The balls represent the amino acids that comprise the L and
M opsins. The photopigments are seven transmembrane proteins. The
ment (Fig. 123.8a). It is not uncommon for the chimeric gene to
red balls indicate amino acids positions 277 and 285. Substitutions at be followed by one or more additional chimeric genes or normal
these positions distinguish the L-class from the M-class pigments. OPN1MW genes (Fig. 123.8a), however, because genes down-
The yellow balls indicate amino acid positions at which substitutions stream of the first two at the 5„ end of the array are usually not
produce spectral shifts, and that are responsible for variability in the expressed, the additional genes do not bear on the color vision
absorption spectrum among the L-class, among the M-class, and phenotype.
between the L- and M-classes. Substitutions at the yellow positions
have a relatively small effect on the absorption spectrum in
comparison to the red positions. The blue balls indicate amino acid
TRITAN COLOR VISION DEFICIENCY
positions that are variable among L- and M-class pigments but that
Color vision defects caused by abnormalities of the S cones are
do not influence the absorption spectrum.
denoted with the prefix tritan, and exhibit autosomal dominant
inheritance. In addition, tritan defects show incomplete
exon 2, 3, and 4 produce spectral variants of the L-class. Only penetrance, meaning that there is variability in the degree to
the polymorphic amino acid positions specified by exon 3 and 4 which color vision is impaired among individuals with the same
produce shifts in spectral peak of M pigments, and thus, there underlying gene defect, even within a family. That is, even
are fewer variant forms of M- than of L-class pigments62 among members of the same family, some individuals might
(Fig. 123.9). In addition, the spectral shifts produced by the exhibit a complete loss of S-cone function, whereas other mem-
polymorphisms in the M-class pigments are relatively small bers may exhibit a milder, incomplete loss.66–69 Thus, tritan
compared to shifts made by the same amino acid substitutions defects do not parallel red-green color vision defects and cannot
at the corresponding positions of the L pigments. From the be dichotomized into analogous dichromatic and anomalous
deduced amino acid sequences of the pigments encoded by the trichromatic forms.
genes in the first two positions of the X-chromosome opsin gene Tritan deficiencies have been associated with mutations of
array, the spectral separation of the pigments underlying color the S-opsin gene resulting in four different amino acid
vision can be predicted. substitutions.70–72 In one study,70 it was concluded that unlike
It is the large difference in spectral absorption between L and mutations in the rod pigment rhodopsin that cause autosomal
1602 M cones that underlies the excellent color discrimination in the dominant retinitis pigmentosa (adRP), amino acid substitutions
Visual Acuity, Adaptation, and Color Vision

in the S opsin do not cause retinal degeneration. Given that lying genetic cause of the disorder among the Pingelapses is an
only ~5% of the cone photoreceptors in humans are S amino acid substitution in the beta subunit of the cyclic-GMP
cones,73–74 this is not surprising. However, the absence of retinal gated ion channel. Phototransduction in all three cone types
degeneration does not imply that the S cones do not degenerate. relies on the function of the same cyclic-GMP gated ion channel,
An explanation for the low penetrance of tritan defects has yet which has two subunits, the alpha subunit encoded by the
to be found, but one interesting possibility is that the S cones CNGA3 gene on chromosome 2, and the beta subunit encoded
degenerate over time, analogous to the degeneration of rods in by the CNGB3 gene on chromosome 8. Mutations in the gene
adRP. If so, the tritan phenotype would be a function of age, encoding the alpha subunit have also been found in families
reflecting a progressive loss of S cones over time. The low pene- with rod monochromacy, and in patients with incomplete achro-
trance aspect of the disorder may simply reflect that younger matopsia.78 Patients with incomplete forms of achromatopsia
observers have not yet lost enough S cones to manifest symp- appear to have residual cone function whereas patients with the
toms. The prevalence of inherited tritan defects have been complete forms do not, implying that not all of the mutations
reported to be quite low, but they may be grossly underestimated identified completely abolish channel function.
for a variety of reasons including the fact that standard
color vision tests do not test for tritan defects, they are COLOR APPEARANCE
extremely difficult to test for, and the phenotype may be age-
dependent. The true incidence of inherited tritan defects and The human eye is popularly described as being capable of dis-
the ultimate fate of the S cones in affected individuals must criminating as many as 10 million ‘colors’. This is offered as a

CHAPTER 123
await further experimentation. A particularly exciting prospect reason that computer displays are made to be capable of
is the application of cutting edge imaging technologies using displaying 256 intensities for each of the red, green, and blue
adaptive optics to the study of the retinal architecture in tritan channels which makes the total number of possible ‘colors’ for
subjects. each pixel equal to 16 777 216 (often approximated as 16 million).
In this context, differences in ‘color’ include differences in
ACHROMATOPSIA brightness, hue, and saturation. It has long been understood
that the ‘millions’ of colors humans can discriminate represent
Also extremely rare are the monochromatic color vision defects subtle gradations of a much smaller set of basic sensations. For
known as achromatopsias. These disorders are associated with example, most people agree that all color experience can be
reduced or absent cone function, denoted as incomplete and described using eleven basic color terms which in English are
complete achromotopsia, respectively. Blue cone monochro- white, black, red, green, yellow, blue, brown, gray, orange,
macy is a form of incomplete achromatopsia in which affected purple, and pink. Among these ‘basic’ colors, theorists agree that
individuals base their vision on S cones and rods, and thus have some are more fundamental than others. For example, pink
diminished capacity for all aspects of vision mediated by cones might be described as a very pale red, brown – a very dark
including color vision and acuity. Rod monochromacy is a form orange, and purple – a reddish-blue. Seven of the basic colors-
of complete achromatopsia in which vision is mediated only by red, green, blue, yellow, black, white, and gray-seem to be truly
rods. Affected individuals are completely colorblind and have fundamental in that each of these sensations seem to be unique
very poor acuity. Achromatopsia has been reported to affect and not describable as a combination of the others.
fewer than one in 30 000 individuals.29 Thus, color experience can be reasonably explained as the
combination of six unique sensations. Accordingly, navy blue is
the simultaneous sensation of blue and black. Pale violet is the
BLUE CONE MONOCHROMACY combined sensation of white, red, and blue. The seventh ‘color’,
Genetically, blue cone monochromacy is a heterogeneous dis- gray, in this scheme is the absence of all color sensation. Unique
order, but in all cases the underlying cause is loss of function of hues were first described by Hering.79 He also noted that red
both L and M cones. One major cause of blue cone mono- and green are opposite hues because they cannot be elicited
chromacy is the deletion of a critical DNA element known as simultaneously by a single color stimulus. There is no reddish-
an enhancer or LCR responsible for facilitating the expression green color nor is there a bluish-yellow. Blue and yellow form a
of the L and M opsin genes. In the absence of the LCR, none of second opponent pair. The same is true of black and white if
the X-chromosome opsin genes are expressed normally, and one accepts that gray is not the simultaneous sensation of black
thus functional L and M cones are not produced.35–37 The and white but rather the absence of either. Hering’s concept of
second major cause of blue cone monochromacy is the deletion opponency consequently organizes the six fundamental sensa-
of all except one of the X-chromosome opsin genes and the tions into the activity of three pairs of opponent sensations,
presence of an inactivating mutation in the remaining gene.35,37 black and white, red and green, and blue and yellow. In systems
The most common mutation is the C203R mutation that has designed to represent all the possible hues on a continuous
been found in conjunction with red-green color vision defects. surface such as Figure 123.11, the four fundamental hues
Blue cone monochromats who in psychophysical tests appear to standout as unique while all other colors are seen as blends of
have more than one class of functional cone have been the unique hues.
reported.75–76 A complete understanding of this disorder must The facts of our vision are that three types of cone photo-
await further experimentation. receptor are responsible for three pairs of sensations. The
question is: how are the signals from the three types of receptors
combined in the nervous system to yield the three opponent
COMPLETE ACHROMATOPSIA AND FORMS pathways? A great deal is known about the anatomy and physi-
OF INCOMPLETE ACHROMATOPSIA OTHER ology of the visual system that can be brought to bear on that
THAN BLUE CONE MONOCHROMACY question, but many aspects of the neural circuits for coding
Although achromatopsias are extremely rare, specific human color remain puzzling. In the task of understanding the neural
populations have been identified that exhibit an extraordinarily operations responsible for transforming the cone signals into
high incidence of the disorder. One example is autosomal reces- perception, a simplification is that of considering reduced color
sive incomplete achromatopsia which has a prevalence of 5% vision systems with fewer cone types and fewer fundamental
among the Pingelapese islanders in Micronesia.77 The under- sensations. 1603
SECTION 10 RETINA AND VITREOUS

FIGURE 123.11. Fan of colors in three-dimensional pigment space.


The fan represents the signal that various colored lights evoke in the
S, M, and L cones.
FIGURE 123.12. The wiring diagram of midget ganglion cells in the
human fovea. In the central retina, each midget bipolar cell receives
THE CIRCUITRY FOR CODING BLACK- input from a single middle- (M) or long-wavelength sensitive (L) cone,
WHITE, RED-GREEN, AND BLUE-YELLOW and contacts a single midget ganglion cell. This one-to-one
organization allows the signal from each M or L cone to be
Information from the retina is carried to higher centers via transmitted to higher brain regions. However, at the cone terminal
the axons of ganglion cells. At the ganglion cell stage, the prior to the transmission to the bipolar cell, a cone’s response is
responses of receptors has already been combined by post- compared in an opponent fashion to the responses of the neighboring
receptoral circuitry to produce neurons with specialized response cones. This comparison is achieved via horizontal cells that provide
lateral reciprocally inhibitory interconnections between all cones.
properties. As introduced in Chapter 111, this processing
Every cone is served by two midget ganglion cells, one ON-center
begins at the output terminal of the cone itself, the cone pedicle. and one OFF-center. Neither will respond to stimuli, such as diffuse
Every cone pedicle receives a lateral inhibitory input from sur- uniform white light, that produce activity in a cone that is equal to the
rounding cones via horizontal cells (Fig. 123.12). Thus, for a average activity of its neighbors.
cone near the fovea, the response communicated to post-
synaptic bipolar cells is the result of a cone’s own electrical
response to light and the opposing responses of its neighboring
cones that arrive via horizontal cell input. Thus, at its synaptic
terminal (pedicle), every cone compares the number of quanta corresponding pair of ON- and OFF-midget bipolar cells that
it absorbs as opposed to the average number of quanta absorbed have opposite responses to the neurotransmitter, glutamate
by its neighbors. Assuming that the potential change at the (Chapter 112), released by cones. While some types of ganglion
pedicle produced by a cone absorbing light is evenly balanced cells are specialized to carry information collected from many
with the opposing input from the average of its neighbors, then, cones, each midget ganglion cell of the central retina is special-
if the number of quanta absorbed by a cone is greater than the ized to transmit information from a single cone.
average absorbed by its neighbors, it hyperpolarizes. If a cone By virtue of its horizontal cell interconnections, a cone com-
absorbs fewer quanta than its average neighbor it depolarizes. pares the number of quanta it absorbs opposed to the average
Most importantly, when the number of quanta absorbed by a number of quanta absorbed by its neighbors. The ON-center
cone is somewhere near equal to its average neighbor, no signal midget ganglion cell is specialized to signal with an increased
is transmitted, i.e., the two opposing inputs null. Thus, a cone rate of action potentials when its cone absorbs a greater number
does not signal information about its photon catch, rather it of photons than the average absorbed by its neighbors as would
signals information about the number of photons it has absorbed happen when a relatively lighter region of an image falls on a
relative to the average number it’s neighbors have absorbed. In cone while its neighboring cones ‘see’ adjacent darker parts of
the central retina a cone’s neighbors would include an average the image. The OFF-center ganglion cell is specialized to signal
of six cones that immediately surround it and cones more the reverse, i.e., when a cone absorbs fewer quanta than its
distant tiers away; however, the strength of the signal falls off average neighbor as when a relatively darker region of the scene
exponentially with distance making the nearest neighbors the falls on the cone. When the number of quanta absorbed by a cone
most important. is equal to its average neighbor, no signal is transmitted, i.e., the
The central retina near the fovea, the region that serves two opposing inputs null and both ON- and OFF-center
highest visual acuity, more than 90% of ganglion cells are of the ganglion cells fire at their spontaneous rates.
‘midget’ variety, so called because of their small dendritic arbor, The midget ON and OFF midget gangion cells have all the
which in the central 7–10° connects to a single cone via a qualities to serve as the biological substrate for the opponent
‘midget’ bipolar cell. Each cone provides input to many ganglion percepts of black and white as proposed by Hering. This is
cells each presumably specialized to carry specific information particularly evident for individuals with a red-green color vision
about the visual stimulus. These include one of each of two defect who have a reduced number of cone types from three
midget ganglion cell subtypes for each cone (Fig. 123.12), one to two and they have a reduced number of fundamental color
1604 ON-center and one OFF-center which receive input through a sensations. As explained above, individuals with only two
Visual Acuity, Adaptation, and Color Vision

FIGURE 123.13. (a) A digital image of fruit that


has been digitally processed in subsequent
panels to represent the activity of different
subsystems for coding color in the human
visual system. (b) The image processed to
appear as it would to a dichromat having color
vision based on just S and L cones. The
dichromat has only four color sensations, blue,
yellow, black, and white. Because blue and
yellow are processed in an opponent fashion,
for a dichromat, there are no intermediate hues
d that represent the simultaneous sensation of
a both blue and yellow. (c) Digitally resampling
the image at a much lower spatial resolution
removes the fine details from the image and
significantly degrades its quality. (d) The black
and white components are separated from
(e) the hue components of the image. Here the
hue components have been resampled with the

CHAPTER 123
same reduced resolution as in panel (c). (f) The
image is reconstituted combining the high
resolution black and white components of (d)
with the low resolution hue components of (e).
The spatial resolution of the hue components
(b) and (f) are very different but on casual
inspection they appear nearly identical. If you
b e
look closely you can see that the yellow color is
blurred outside the black lines. This is largely
ignored by the visual system that uses the
black and white edges to define detailed
boundaries in the image. The hue information is
used to define qualities of the objects other
than spatial detail.

c f

spectrally different types of cones are refered to as dichromats. cones are many times fewer in number (~5% of the total in
The prefix ‘di-’ in dichromacy does not refer to two types of humans). Assuming a balanced center versus surrounds, this
cone. Dichromacy means, literally, two hues and derives from system signals the presence of light/dark boundaries in a visual
the fact that dichromats can match any color using mixtures of scene. If a dark area in an image falls on a cone, but adjacent
just two ‘primary’ hues. However, the term dichromat is also lighter areas illuminate neighboring cones, the OFF-center
appropriate because dichromats see only two hues. To them, midget ganglion will signal with increased firing. Conversely, a
objects are black, white, shades of gray, or one of two hues. The light area bounded by darker regions will be signaled by increased
image in Figure 123.13 has been digitally altered to simulate the firing of the ON-center ganglion cell. These are exactly the
appearance for a dichromat. For the dichromat there is only stimulus conditions we associate with the percepts of black
black and white and blue and yellow. In contrast, people with and white.
normal color vision see more than 100 different hues in In addition to serving the percepts of black and white, this
addition to black, white, and gray. Dichromats confuse red with system presumably evolved to serve high acuity spatial vision,
green, and they confuse, with red and green, all colors in the signaling the presence of dark/light edges with great detail and
spectrum that fall between them, including yellow, orange, and precision. Representing more than 90% of the ganglion cells that
brown. They see blue and violet as the same color, and blue- serve the central retina, the midget ganglion cells are the only
green is indistinguishable from white or gray. Magenta and its output neurons that have sufficient numbers to transmit infor-
pastel counterpart pink also appear white or gray. mation about fine detail in the image. Accordingly, the midget
During evolution, the midget ganglion cell system is believed ganglion cells appear to simultaneously serve two purposes in
to have arisen in an ancestor to modern primates prior to the vision for a dichromat. The ON- and OFF-center midget ganglion
emergence of trichromatic color vision. Like most other mam- cells form the basis for the percepts of black and white, respec-
mals, that ancestor presumably had two cone types, S cones and tively. In turn, the percepts of black and white in the
a second type of cone sensitive in the middle-to-long wave- dichromatic image are responsible for coding information about
lengths (Fig. 123.7b). The midget ganglion cells do not receive fine detail in the image. This is evident when the black and
direct center input from S cones. Thus, in a dichromatic ancestor white of an image (Fig. 123.13c) is separated digitally from its
to humans, the midget ganglion cell system’s major function color, all the detail of the image is preserved.
was to compare absorptions of middle-to-long wavelength cones We use information about hue to find out about the internal
with their neighbors. The neighbors of an L/M cone would have qualities of objects. For example, a trichromat can tell that a
been predominately of the identical spectral type because S banana is ripe by its yellow color. However, to determine its 1605
RETINA AND VITREOUS

ripeness we do not need information about the fine detail of the vision and they could be important for providing the required
spatial distribution of the yellow color. This is true of all the two opponent parts of the system.
information we extract from the hue of objects. We know that The story would be relatively simple if humans were all
it is evening because of the red of the sunset or that a colleague dichromats. If this were true, details of the specialized functions
is embarrassed because of the redness in his face. However, we of each of three major ganglion cell populations carrying infor-
do not need to know details of the spatial distribution of the mation to the brain would be easily understood. The midget
colored areas. Even when we use hue to locate objects in visual ganglion cells provide the biological substrate for percepts of
search, the color can ‘catch our eye’ in the absence of the spatial black and white and are responsible for carrying information
details of the object. The S cones are used in color vision but about high acuity spatial detail. The second major ganglion cell
they do not participate in providing high acuity. They can parti- population, the parasol cells, are responsible for the perception
cipate in providing hue information at a much lower sampling of motion. These make up only ~5% of ganglion cells in the
density than the L and M cones that provide information about fovea but are a much larger proportion of ganglion cells in the
spatial detail. The unimportance of extracting color vision with peripheral retina where our motion perception is most acute.
high spatial resolution is illustrated in Fig. 123.13. In Fig 123.13e, Although details are left to be worked out, the third major
the hue components of the image were separated and resampled ganglion cell population, the small bistratified ganglion cells
at a much lower resolution. These low resolution hue compo- must participate in blue-yellow color vision. However, the
nents provide almost no information that allows us to recognize introduction of red-green color vision complicates the story
the objects in the scene. However, when the low quality hue because adding a randomly distributed third cone type makes
SECTION 10

components are recombined with (d), the black and white almost every midget ganglion cell respond to either diffuse red
components of the image, (Fig. 123.13f) the result is almost or diffuse green light in addition to dark and light edges. A
imperceptibly different than the original image (b). guiding principle in our understanding the biological basis of
It is not completely clear which ganglion cell subtypes are sensation for more than 150 years is Johannes Müller’s (1833)
specialized for carrying information corresponding to Herring’s ‘Law of Specific Nerve Energies’ which states “Qualitatively
blue-yellow opponent channel. Small bistratified ganglion cells different sensations must derive from different organs.” Accord-
make up less than 10% of the total output of the retina and they ingly, even though the midget ganglion cells in trichomats
have opponent blue-yellow responses. Thus, they are candidates respond to both diffuse red and green lights and black and white
to have some role in blue-yellow color vision. However, they all edges, ultimately the two percepts must be separated from each
respond to blue and inhibit to yellow, leaving us without an other at a higher level of visual processing, into two perceptually
explanation of the physiological basis for the yellow half of the opponent processes of black and white and red and green.
blue-yellow opponent system. It is possible that a subpopulation How this is accomplished is one of the fascinating remaining
of midget ganglion cells is also involved in blue-yellow color mysteries of our visual system.

REFERENCES
1. Green DG, Powers MK, Banks MS: Depth 12. Hyvarinen L, Rovamo J, Laurinen P, et al: 23. Yarbus AL: The perception of an image
of focus, eye size and visual acuity. Vision Contrast sensitivity in monocular glaucoma. fixed with respect to the retina. Biophysics
Res 1980; 20:827. Acta Ophthalmol 1983; 61:742. 1957; 2:683.
2. VanMeeteren A: Calculations on the optical 13. Hyvarinen L, Laurinen P, Rovamo J: 24. Riggs LA, Tulunay SU: Visual effects of
modulation transfer function of the human Contrast sensitivity in evaluation of visual varying the extent of compensation for
eye for white light. Opt Acta 1974; 15:47. impairment due to macular degeneration eye movements. J Opt Soc Am 1959;
3. Campbell FW, Gregory AH: Effect of size of and optic nerve lesions. Acta Ophthalmol 49:741.
pupil on visual acuity. Nature 1960; 1983; 61:161. 25. Barlow HB: Slippage of contact lenses and
187:1121. 14. Hyvarinen L, Laurinen P, Rovamo J: other artefacts in relation to fading and
4. Campbell FW, Green DG: Optical and Contrast sensitivity in evaluation of visual regeneration of supposedly stable retinal
retinal factors affecting visual resolution. impairment due to diabetes. Acta images. Q J Exp Psychol 1963; 15:36.
J Physiol 1965; 181:576. Ophthalmol 1983; 61:94. 26. Tulunay-Keesey U, Jones RM: The effect of
5. Green DG: Visual acuity: The influence of 15. Koskela PU, Hyvarinen L: Contrast micromovements of the eye and exposure
refraction and diffraction and the use of sensitivity in amblyopia. IV. Assessment of duration on contrast sensitivity. Vision Res
interference fringes. Int Ophthalmol Clin vision using vertical and horizontal gratings 1976; 16:481.
1978; 18:21. and optotypes at different contrast levels. 27. Aquilar M, Stiles WS: Saturation of the rod
6. Green DG: Regional variations in the visual Acta Ophthalmol 1986; 64:570. mechanisms of the retina at high levels of
acuity for interference fringes on the retina. 16. Gaskill JD: Linear systems, Fourier stimulation. Opt Acta 1954; 1:59.
J Physiol 1970; 207:351. transforms, optics. New York: John Wiley; 28. Rushton WAH: The sensitivity of rods under
7. Le Grand Y: Form and space vision. 1978. illumination. J Physiol 1965; 178:141.
Bloomington: Indiana University Press; 17. Bodis-Wollner I: Visual acuity and contrast 29. Sharpe LT, Stockman A, Jägle H,
1967. sensitivity in patients with cerebral lesions. Nathans J: Opsin genes, cone
8. Thibos LN, Cheney FE, Walsh DJ: Retinal Science 1972; 178:679. photopigments, color vision, and color
limits to the detection and resolution of 18. Brown B: Reading performance in low blindness. In: Gegenfurtner KR, Sharpe LT,
gratings. J Opt Soc Am A 1987; 4:1524. vision patients: relation to contrast and eds. Color vision: from genes to
9. Lovegrove WJ, Bowling A, Badcock D, contrast sensitivity. Am J Optom Physiol perception. New York: Cambridge
Blackwood M: Specific reading disability: Opt 1981; 58:218. University Press; 1999:3.
differences in contrast sensitivity as a 19. Rubin GS, Legge GE: Psychophysics of 30. Dean M, Carrington M, O’Brien SJ:
function of spatial frequency. Science 1980; reading. VI. The role of contrast in low Balanced polymorphism selected by
210:439. vision. Vision Res 1989; 29:79. genetic versus infectious human disease.
10. Hyvarinen L, Rovamo J, Laurinen P, 20. Pirenne MH, Denton EJ: Accuracy and Ann Rev Genom Human Genet 2002;
Peltomaa A: Contrast sensitivity function in sensitivity of the human eye. Nature 1952; 3:263.
evaluation of visual impairment due to 170:1039. 31. Nathans J, Piantanida TP, Eddy RL, et al:
retinitis pigmentosa. Acta Ophthalmol 21. Shlaer S, Smith E, Chase A: Visual acuity Molecular genetics of inherited variation in
1981; 59:763. and illumination in different spectral human color vision. Science 1986; 232:203.
11. Hyvarinen L: Contrast sensitivity in visually regions. J Gen Physiol 1942; 25:553. 32. Jacobs GH: The distribution and nature of
impaired children. Acta Ophthalmol 1983; 22. Mendelbaum J, Sloan LL: Peripheral visual colour vision among the mammals. Biol
157(Suppl):58. acuity. Am J Ophthalmol 1947; 30:581. Rev 1993; 68:413.
1606
Visual Acuity, Adaptation, and Color Vision

33. Jacobs GH: A perspective on color vision 49. Neitz J, Neitz M, He JC, Shevell SK: 63. Merbs SL, Nathans J: Absorption spectra
in platyrrhine monkeys. Vision Res 1998; Trichromatic color vision with only two of the hybrid pigments responsible for
38:3307. spectrally distinct photopigments. Nat anomalous color vision. Science 1992;
34. Jacobs GH, Neitz J: Color vision in Neurosci 1999; 2:884. 258:464.
monkeys: sex related differences suggest 50. Deeb SS, Lindsey DT, Hibiya Y, et al: 64. Neitz J, Neitz M, Kainz PM: Visual pigment
the mode of inheritance. Vision Res 1985; Genotype-phenotype relationships in gene structure and the severity of human
25:141. human red/green color-vision defects: color vision defects. Science 1996; 274:801.
35. Nathans J, Davenport CM, Maumenee IH, molecular and psychophysical studies. Am 65. Regan BC, Reffin JP, Mollon JD:
et al: Molecular genetics of blue cone J Hum Genet 1992; 51:687. Luminance noise and the rapid
monochromacy. Science 1989; 245:831. 51. Carroll J, Neitz M, Neitz J: Testing determination of discrimination ellipses in
36. Wang Y, Macke JP, Merbs SL, et al: A locus hypotheses about visual pigments colour deficiency. Vision Res 1994; 34:1279.
control region adjacent to the human red underlying deutan color vision. Color Res 66. Wright WD: The characteristics of
and green visual pigment genes. Neuron Appl 2001; 26:S106. tritanopia. J Opt Soc Am 1952; 42:509.
1992; 9:429. 52. Neitz M, Neitz J: Molecular genetics of 67. Kalmus H: The familial distribution of
37. Nathans J, Maumenee IA, Zrenner E, et al: color vision and color vision defects. Arch congenital tritanopia. Ann Hum Genet
Genetic heterogeneity among blue-cone Ophthalmol 2000; 118:691. 1955; 20:39.
monochromats. Am J Hum Genet 1993; 53. Winderickx J, Sanocki E, Lindsey DT, et al: 68. Pokorny J, Smith VC, Went LN: Color
53:987. Defective colour vision associated with a matching in autosomal dominant tritan
38. Smallwood PM, Wang Y, Nathans J: Role missense mutation in the human green defect. J Opt Soc Am 1981; 71:1374.
of a locus control region in the mutually visual pigment gene. Nat Genet 1992; 1:251. 69. Went LN, Pronk N: The genetics of tritan

CHAPTER 123
exclusive expression of human red and 54. Karnik SS, Khorana HG: Assembly disturbances. Hum Genet 1985; 69:255.
green cone pigment genes. PNAS 2002; functional rhodopsin requires a disulfide 70. Weitz CJ, Miyake Y, Shinzato K, et al:
99:1008. bond between cysteine residues 110 and Human tritanopia associated with two
39. Hagstrom SA, Neitz M, Neitz J: Cone 187. J Biol Chem 1990; 265:17520. amino acid substitutions in the blue
pigment gene expression in individual 55. Kazmi MA, Sakmar TP, Ostrer H: Mutation sensitive opsin. Am J Hum Genet 1992;
photoreceptors and the chromatic of a conserved cysteine in the X-linked 50:498.
topography of the retina. J Opt Soc Am A cone opsins causes color vision 71. Weitz CJ, Went LN, Nathans J: Human
Opt Image Sci Vis 2000; 17:527. deficiencies by disrupting protein folding tritanopia associated with a third amino
40. Onishi A, Koike S, Ida M, et al: and stablilty. Invest Ophthalmol Vis Sci acid substitution in the blue sensitive visual
Dichromatism in macaque monkeys. Nature 1997; 38:1074. pigment. Am J Hum Genet 1992; 51:444.
1999; 402:139. 56. Nathans J, Thomas D, Hogness DS: 72. Gunther KL, Neitz J, Neitz M: A novel
41. Hayashi T, Motulsky AG, Deeb SS: Position Molecular genetics of human color vision: mutation in the short-wavelength sensitive
of a ‘green-red’ hybrid gene in the visual the genes encoding blue, green, and red cone pigment gene associtated with a tritan
pigment array determines colour-vision pigments. Science 1986; 232:193. color vision defect. Vis Neurosci 2006;
phenotype. Nat Genet 1999; 22:90. 57. Winderickx J, Battisti L, Hibibya Y, et al: 23:403.
42. Bollinger K, Sjoberg S, Neitz M, Neitz J: Haplotype diversity in the human red and 73. Curcio CA, Allen KA, Sloan KR, et al:
Topographical cone photopigment gene green opsin genes: evidence for frequent Distribution and morphology of human
expression in deutan-type red-green color sequence exchange in exon 3. Hum Mol cone photoreceptors stained with anti-blue
vision defects. Vision Res 2004; 34:135. Genet 1993; 2:1413. opsin. J Comp Neurol 1991; 312:610.
43. Sjoberg SA, Neitz M, Balding SD, Neitz J: 58. Neitz M, Neitz J, Grishok A: Polymorphism 74. Hofer H, Carroll J, Neitz J, et al:
L-cone pigment genes expressed in normal in the number of genes encoding long- Organization of the human trichromatic
colour vision. Vision Res 1998; 38:3213. wavelength sensitive cone pigments among cone mosaic. J Neurosci 2006; 26:722.
44. Shevell SK, He JC, Kainz PM, et al: males with normal color vision. Vision Res 75. Smith VC, Pokorny J, Delleman JW, et al:
Relating color discrimination to 1995; 35:2395. X-linked incomplete achromatopsia with
photopigment genes in deutan observers. 59. Carroll J, McMahon C, Neitz M, Neitz J: more than one class of functional cones.
Vision Res 1998; 38:3371. Flicker-photometric electroretinogram Invest Ophthalmol Vis Sci 1983; 24:451.
45. Neitz M, Neitz J: A new mass screening estimates of L : M cone photoreceptor ratio 76. Crognale MA, Fry M, Highsmith J, et al:
test for color-vision deficiencies in children. in men with photopigment spectra derived Characterization of a novel form of X-linked
Color Res Appl 2001; 26:S239. from genetics. J Opt Soc Am A Opt Image incomplete achromatopsia. Vis Neurosci
46. Jagla WM, Jägle H, Hayashi T, et al: Sci Vis 2000; 17:499. 2004; 21:197.
The molecular basis of dichromatic color 60. Carroll J, Neitz M, Hofer H, et al: Functional 77. Sundin OH, Yang JM, Li Y, et al: Genetic
vision in males with multiple red and green photoreceptor loss revealed with adaptive basis of total colourblindness among the
visual pigment genes. Hum Mol Genet optics: an alternate cause of color Pingelapese islanders. Nat Genet 2000;
2002; 11:23. blindness. Proc Natl Acad Sci USA 2004; 25:289.
47. Neitz M, Carroll J, Renner A, et al: Variety 101:8461. 78. Wissinger B, Gamer D, Jägle H, et al:
of genotypes in males diagnosed as 61. Neitz M, Neitz J, Jacobs GH: Spectral CNGA3 mutations in hereditary cone
dichromatic on a conventional clinical tuning of pigments underlying red-green photoreceptor disorders. Am J Hum Genet
anomaloscope. Vis Neurosci 2004; 21:205. color vision. Science 1991; 252:971. 2001; 69:722.
48. Kainz PM, Neitz M, Neitz J: Molecular 62. Asenjo AB, Rim J, Oprian DD: Molecular 79. Hering E: Outlines of a theory of the light
genetic detection of female carriers of determinants of human red/green color sense. (Hurvich LM, Jameson D, transl)
protan defects. Vision Res 1998; 38:3365. discrimination. Neuron 1994; 12:1131. Cambridge: Harvard University Press; 1964.

1607
CHAPTER

124 Objective Assessment of Retinal Function


Michael A. Sandberg

INTRODUCTION with dominant retinitis pigmentosa based on generalized rod


loss versus regionalized rod and cone loss6 and in monitoring
This chapter describes the methodology application, and the course of diseases affecting the uvea and retinal pigment
normal variation of some objective measures of retinal function epithelium (RPE).7
that are used to establish diagnoses in known or suspected Two types of reflectometer are in use at present, normally in
hereditary retinal diseases. Objective assessment of retinal a clinical research setting. The first, and original, type involves
function is important for at least three reasons. First, it provides collecting and comparing the amount of light reflected by 1–2°
evidence about the site of localization of visual loss (e.g., of the fundus, initially from the dark-adapted eye and then again
whether the site of visual loss is within the eye or visual after exposure to a light that bleaches most of the available visual
pathways or whether the abnormality is restricted to the macula pigment.8–10 The light from a brief test flash, which in itself
or involves the entire retina). Second, in most cases it permits bleaches little of the visual pigment, is reflected from the fundus
a quantitative assessment of the degree of malfunction that can and focused on the head of a photomultiplier tube. The differ-
be followed up over time for the purpose of projecting long-term ence in the reflected light before and after the bleaching episode
prognosis or evaluating a prospective treatment. Third, outcomes provides a measure of the amount of visual pigment. The second,
from these measures may be shown to the patient as variations and more recent, type of reflectometer involves imaging the fundus
in fundus reflectance or as waveforms on photographs or paper over a visual angle of at least 10° and capturing the reflected
so that the patient can appreciate the type and magnitude of his light either photographically or on videotape. Density differences
or her visual malfunction and thereby actively participate with are then quantified by comparing unbleached with bleached
the ophthalmologist in the initial and follow-up examinations. areas captured in a single image11,12 or between successive
The techniques addressed include fundus reflectometry, fundus images.13,14
autofluorescence, optical coherence tomography (OCT), early Both systems may be properly used only in patients with clear
receptor potential (ERP) recording, electrooculography, full-field media and stable fixation. The pupil is maximally dilated and
and focal flash electroretinography, pattern-reversal electroretino- the eye undergoes dark adaptation for at least 30 min, during
graphy, and pattern-reversal visual evoked response (VER) record- which time an impression of the patient’s bite is made with
ing. The first three methods, though not measures of function dental wax. With the patient’s head stabilized by the wax impres-
per se, are always presented in the context of measures of function sion and the fellow eye fixating on a red lamp or light-emitting
in interpreting hereditary retinal disease. (OCT is considered in diode, the examiner aligns the eye to be tested in dim red or
greater detail elsewhere in this publication.) Although VER record- infrared light that causes negligible bleaching. For assessing
ing does not reflect retinal function directly, it is a necessary test rhodopsin density, the mid-peripheral fundus, in which cone
when measures of retinal function fail to disclose the site of photoreceptors are scarce, is generally chosen. For assessing cone
abnormality. It is therefore discussed briefly at the end of the pigment density, the fovea, in which rods are fewest, is chosen.
chapter. Guidelines are presented for obtaining reliable and repro- A test flash of narrow-band wavelength near the peak of the absorp-
ducible results and, in some cases, for interpreting variations tion spectrum (i.e., ~500 nm for rods and ~560 nm for cones)
within a single session and between visits in a given patient as is then presented to quantify reflectance of the dark-adapted eye.
well as differences that may exist among normal subjects. After reaffirming alignment in red light, a bright white light is
usually presented for many seconds to bleach at least 95% of the
FUNDUS REFLECTOMETRY visual pigment. The test flash is presented again to quantify reflect-
ance without significant absorption by visual pigment. The
Fundus reflectometry (also known as retinal densitometry) is a logarithmic difference between the two measurements repres-
method for estimating the mass optical density, absorption spectra, ents the pigment density. This is a ‘double-density’ difference in
and regeneration kinetics of the photolabile pigments within that the test beam has passed through the photoreceptor layer
photoreceptors. Although still primarily a research tool, it may twice, the second time by reflection from the pigment epithelium
be used clinically to help identify stationary forms of nyctalopia and choroid. Additional test flashes may be presented either at
that have normal rhodopsin densities but a defect in transmission other wavelengths in rapid succession to both dark-adapted and
between rod photoreceptors and more proximal retinal cells1; bleached eyes to determine the absorption spectrum of the visual
diseases of dark adaptation involving slowed pigment regenera- pigment or at known intervals after the bleaching episode to
tion, such as fundus albipunctatus (Fig. 124.1)2; diseases of dark assess the time course of pigment regeneration. Figure 124.2
adaptation with normal pigment regeneration, such as Oguchi’s illustrates results from an imaging densitometer. In this case, a
disease3; and photoreceptor mosaicism in carriers of X-linked mid-peripheral region of retina was initially bleached for 10 s
retinitis pigmentosa.4,5 It is also useful in subtyping patients over only the right half of the image, the left half remaining 1609
RETINA AND VITREOUS

FIGURE 124.1. Visual pigment regeneration for two brothers (LD and
ED) with fundus albipunctatus. Recovery times to 50% of maximum
were ~1 h for rhodopsin in the peripheral retina and 20 min for cone
SECTION 10

pigments in the fovea, increases of approximately 20-fold and 16-fold,


respectively, compared with normal times with this test system.
Modified from Carr RE, Ripps H, Siegel IM: Visual pigment kinetics and
adaptation in fundus albipunctatus. Doc Ophthalmol Proc Ser 1974; 4:193. FIGURE 124.2. Midperipheral regions of a normal human fundus
Reprinted from Kluwer Academic Publishers. photographed on videotape with 502-nm light immediately after a
greater than 95% rhodopsin bleach over the right half performed with
an imaging reflectometer. Squares at the bottom were copied from
adapted to the dark. The figure shows the image photographed areas designated with letters to facilitate comparison of double-
by a 500-nm test flash and captured on videotape. The bleached density gray values, which differ by ~0.2 log unit.
half appears brighter than the unbleached half, indicating less
remaining visual pigment in the former.
Normal values for rhodopsin double density range from 0.08 macular pigment and by the increased melanin in this region,
to 0.15 log-unit in different peripheral regions for young adult and increased markedly with increasing age for excitation
subjects15 and from 0.06 to 0.14 log-unit between regions of a >470 nm (Fig. 124.3, left).
single subject.16 For cone pigment, the range between subjects is Later in the same year, Delori et al showed that patients with
0.14–0.30 log-unit within the central 2°, which falls to 0.05–0.11 autosomal recessive juvenile macular degeneration (Stargardt
log-unit at an average eccentricity of 2.5° degrees.14 Foveal cone disease/fundus flavimaculatus) and a dark choroid on fluorescein
pigment density has been reported to decline linearly with age.17 angiography had increased autofluorescence of the RPE after
taking into account age (Fig. 124.3, right), indicating abnormally
FUNDUS AUTOFLUORESCENCE high accumulation of lipofuscin in these patients.19 Since then,
the adaptation of a confocal scanning laser ophthalmoscope in
Delori and associates in 1995 used noninvasive fundus spectro- London has led to a series of papers describing the imaging of
photometry to demonstrate that lipofuscin in the human RPE fluorescence of the RPE in patients with different retinal diseases.
exhibits a red fluorescence when stimulated by a shorter-wave- These authors reported increased autofluorescence in patients with
length light.18 Emission peaked between 620 and 640 nm, and macular dystrophy in general (i.e., even in the absence of a dark
excitation peaked at 510 nm. The shape of the emission spectrum choroid on fluorescein angiography),20 although some patients
changed for excitation wavelengths ≤ 470 nm, indicating a second with Stargardt disease had reportedly normal or reduced auto-
fluorophore. The authors also showed that fluorescence was fluorescence.21 Measurements with this instrument have also
minimal at the fovea, apparently due to excitation absorbance by been performed in retinitis pigmentosa, where rings of increased

FIGURE 124.3. (Left) Variation of RPE fluorescence with retinal location and age. Fluorescence at 620 nm was excited by a wavelength of
510 nm. Spatial resolution = 2°. (Right) Fluorescence of the RPE at 620 nm to an excitation of 510 nm for one or both eyes of patients with
Stargardt disease/fundus flavimaculatus (triangles) and for healthy controls (circles, n = 45). Error bars designate ±1 standard deviation. The solid
and dashed lines represent the best-fitting regression line and its 99.99% confidence limits.
(Left) From Delori FC, Dorey CK, Staurenghi G, et al: In vivo fluorescence of the ocular fundus exhibits retinal pigment epithelium lipofuscin characteristics. Invest
Ophthalmol Vis Sci 1995; 36:718. (Right) From Delori FC, Staurenghi G, Arend O, et al: In vivo measurement of lipofuscin in Stargardt’s disease – Fundus
1610 Flavimaculatus. Invest Ophthalmol Vis Sci Mar 1995; 36:2327.
Objective Assessment of Retinal Function

FIGURE 124.4. Tomograms recorded with a


OD OS
Zeiss Stratus High-Resolution Optical
Normal Coherence Tomographer from a 39-year-old
female normal control with Snellen visual
ONL
acuities of 20/20 OD and 20/20 OS (upper
rd
3 HRB
RPE
images), a 34-year-old male with retinitis
pigmentosa (RP) and visual acuities of 20/20
OD and 20/20 OS (middle images), and a 33-
year-old male with RP and visual acuities of
20/200 OD and 20/80 OS (lower images). Each
RP tomogram subtended 6 mm centered on the
fovea. The horizontal arrows (upper right)
designate the low-reflective outer nuclear layer
(ONL), the third high-reflectance band possibly
designating the photoreceptor inner
segment/outer segment junction (third HRB),
and the high-reflective RPE/choriocapillaris
(RPE).
RP Sandberg MA, Brockhurst RJ, Gaudio AR, Berson EL:

CHAPTER 124
The association between visual acuity and central
retinal thickness in retinitis pigmentosa. Invest
Ophthalmol Vis Sci 2005; 46:3349.

autofluorescence in the macula and areas of decreased fluores- these investigators showed that both retinal thinning (due to
cence in the periphery have been observed.22,23 Conversely, some cell loss) and retinal thickening (due to presumed edema) were
patients with Leber congenital amaurosis, an early-onset severe associated with lower visual acuity (Fig. 124.5).24 This study
form of retinitis pigmentosa, have been found to have minimally, also found that an increase or decrease in retinal thickness of
if at all, disturbed autofluorescence, which the authors interpreted more than 17 mm at fixation at follow-up can be considered a sig-
as indicating morphologically preserved photoreceptors and nificant (p < 0.01) change in patients with retinitis pigmentosa.
RPE in those regions.23 However, since the confocal scanning OCT has also been used to detect cystoid macular edema
laser ophthalmoscope excites autofluorescence with a lower wave- (CME) in patients with retinitis pigmentosa. One of these studies
length (488 nm) and measures autofluorescence over a broader found that the area of the cystoid spaces was positively correlated
spectrum (with a 50% cut-on at 510 nm) than the results of Delori with the grade of the fluorescein angiogram and the visual acuity,25
and colleagues would recommend, it is possible that differences and two studies have reported that some eyes with cystoid lesions
in lenticular absorption, in macular pigment density, in uveal showed no angiographic evidence of dye leakage or pooling.25,26
pigment density, and in the relative concentrations of multiple
retinal and RPE fluorophores may, in part, confound interpre-
tation when this instrument is applied to different types and
stages of hereditary retinal degeneration.

OPTICAL COHERENCE TOMOGRAPHY


OCT is a new rapid noninvasive method for obtaining cross-
sectional images of the retina based on differential near-infrared
light reflection at optical interfaces. Although its most obvious
application has been to help monitor the surgical treatment of
macular holes, epiretinal membranes, and retinal tears, OCT can
also be used to help assess hereditary retinal disease. Figure 124.4
illustrates representative tomograms from a normal control sub-
ject and two patients with retinitis pigmentosa. The tomograms
were obtained with a commercial third-generation instrument
(OCT3). The upper tomograms from the normal control show
the low-reflectance outer nuclear layer peaking in thickness in
the center beneath the foveal depression and visible from edge
to edge. A third high-reflectance band can be distinguished just
above a thicker high-reflectance RPE/choriocapillaris complex; FIGURE 124.5. Regression of ETDRS visual acuity on OCT retinal
the third high-reflectance band is slightly convex in the center, thickness at fixation based on data from 288 eyes of 162 patients with
possibly reflecting longer cone outer segments in this region. retinitis pigmentosa without macular cysts. The straight line with large
The middle tomograms from a patient with retinitis pigmentosa dashes is the best-fitting linear function. The curve with small dashes
and normal visual acuities have a normal appearance in the is the best-fitting log function. The solid curve is the best-fitting
second-order polynomial and best describes the data. The different
center (with an intact third high-reflectance band) but show loss
symbols designate the definition of the third high-reflectance band
of the outer nuclear layer more peripherally. The lower tomo- (third HRB) which is thought to represent the inner segment/outer
grams from a patient with retinitis pigmentosa and poor visual segment junction.
acuities show widespread loss of the outer nuclear layer with From Sandberg MA, Brockhurst RJ, Gaudio AR, Berson EL: The association
apparent thinning of the inner retina. By examining patients between visual acuity and central retinal thickness in retinitis pigmentosa.
with retinitis pigmentosa and no cystic changes in the macula, Invest Ophthalmol Vis Sci 2005; 46:3349. 1611
RETINA AND VITREOUS

FIGURE 124.6. OCT images from the right eye


of a 44-year-old male with retinitis pigmentosa
before (a) and 3 weeks after (b) beginning
treatment with acetazolamide, at which time
the macular cysts had decreased in size.
From Apushkin MA, Fishman GA, Janowicz MJ:
Monitoring cystoid macular edema by optical
coherence tomography in patients with retinitis
pigmentosa. Ophthalmology 2004; 111:1899.
a b

Figure 124.6 illustrates tomograms from a patient with retinitis


pigmentosa, before and after treatment with acetazolamide for
CME; the patient showed no evidence of CME on fundus
examination or fluorescein angiography prior to treatment.26
Several studies have also used OCT to visualize cystic lesions in
patients with hereditary retinoschisis.27–30
SECTION 10

EARLY RECEPTOR POTENTIAL


RECORDING
The ERP is a very short latency, biphasic response from the eye
elicited by a very bright flash of light.31 It consists of a cornea-
FIGURE 124.7. Normal human ERP followed by an a-wave of ERG
positive component (R1) followed by a cornea-negative compo-
(left tracing) and normal ERP with high sweep speed and amplification
nent (R2), which is then followed by the cornea-negative a-wave (right tracing). Both cornea-positive peak (R1) and later cornea-
of the electroretinogram (ERG) (Fig. 124.7). It derives almost negative peak (R2) of ERP are designated. Stimulus onset is at the
entirely from photoreceptors32 – R1 reflects the conversion of beginning of each trace. Calibration symbol = 2 ms horizontally and
lumirhodopsin to metarhodopsin I,33 whereas R2 reflects the 100 µV vertically for the left tracing; 0.5 ms horizontally and 50 µV
conversion of metarhodopsin I to metarhodopsin II.34 As the ERP vertically for the right tracing.
amplitude is proportional to the amount of pigment bleached by From Berson EL, Goldstein EB: The early receptor potential in dominantly
the flash35 and depends on the orientation of visual pigment mole- inherited retinitis pigmentosa. Arch Ophthalmol 1970; 83:412. Copyright 1970,
cules within the outer segment,36 it may be used as a measure American Medical Association.
of mass outer segment optical density over most of the retina.
In humans, 60–70% of the R2 response is generated by cones
and the remainder by rods,37,38 so that it may not be possible to
specify whether one or both receptor systems are involved if the
amplitude reduction is less than 40%. The ERP has been used
to demonstrate photoreceptor impairment in a variety of retinal
diseases, including retinitis pigmentosa.31,39,40
The ERP must be elicited with a light flash ~10 million
times brighter than that required to elicit the b-wave of the ERG
(Fig. 124.8).41 This can be achieved, for example, with a flash-
gun focused in the plane of the pupil in Maxwellian view. Care
must be taken to avoid a photovoltaic artifact superimposed on
R1. Investigators have developed custom monopolar contact
lenses with either nonmetallic electrodes or metallic electrodes
shielded from incident light42–44 to record an ERP uncontami-
nated by a photovoltaic artifact. Since the light flash must bleach
a considerable amount of visual pigment to generate a detect-
able response, time for pigment regeneration must be allowed
before presenting successive flashes to illustrate reproducibility.
Intraindividual and interindividual variations in ERP amplitude
may be as much as 3:1.45
FIGURE 124.8. Amplitudes of the b-wave and a-wave of the ERG
ELECTROOCULOGRAPHY and the R2 component of the ERP for the dark-adapted albino rat as
a function of the log of the flash energy. Responses were obtained
The light-rise of the electrooculogram (EOG) is a measure of the with 0.7-ms full-field white flashes. Log flash energy of zero
integrity of the RPE and overlying photoreceptors. It arises from corresponds to one quantum absorbed/rod. Amplitudes were
a depolarization of the basal (i.e., choroidal side) membrane of measured from baseline.
the RPE.46 The EOG is a measure of the slowly changing voltage From Cone RA: The early receptor potential of the vertebrate eye. Cold Spring
Harbor Symp Quant Biol 1965; 30:483.
difference between the front (positive) and the rear (negative)
surfaces of the globe recorded over time under different condi-
tions of illumination (Fig. 124.9). The light-rise represents the the EOG light-rise is reduced or absent.48–51 Asymptomatic carriers
largest difference measured in illumination divided by the of Best’s disease may also have abnormal EOGs.50 The EOG
smallest difference measured in darkness.47 In clinical practice, also differentiates Best’s disease from pseudovitelliform macular
its important use is to help diagnose Best’s vitelliform macular degeneration, in which there is generally a normal ratio.51 In
1612 degeneration, a dominantly inherited condition. In this disease, other retinal diseases, EOG testing does not add to whatever
Objective Assessment of Retinal Function

FIGURE 124.9. EOG recorded from a normal subject. Eye


movements were made twice each minute by alternately viewing
fixation points separated by 30° in a Ganzfeld dome, differentially
amplified at a gain of 200 (–3 dB at 0.1 and 100 Hz), digitized, and
peak-to-peak amplitudes for each saccade quantified by computer.
Vertical dotted lines are separated by 2.5-min intervals; horizontal

CHAPTER 124
dotted lines are separated by 200 µV. Arrow designates onset of
background illumination.

diagnosis may have been made with the ERG alone.52 Fast
oscillations of the EOG have also been recorded53 and have been
found to be dissociated from the light-rise of the EOG in some
maculopathies. For example, in one report the fast oscillations
were found to be normal or nearly normal, while the light-rise
was abnormal, in Best’s disease (Fig. 124.10).54
The EOG may be measured in cooperative patients with
stable fixation who have a visual-field diameter of at least 60°.
After exposure to ambient room illumination for 30 min or
longer, during which time the pupils are dilated, cup electrodes
filled with electrode cream are attached with tape just lateral to
the inner and outer canthus of each eye (i.e., two electrodes per
eye) and to the forehead as ground. On verbal instruction from
the examiner, the subject alternately fixates red light-emitting
diodes in a Ganzfeld dome placed at 0° and 30° with respect to FIGURE 124.10. EOG from a normal subject (A) and from three
forward gaze, first in the dark for ~12 min and then in the patients with Best disease (B–D). Event marker line below indicates
presence of a full-field 10 ftL white background for ~12 min to ‘on’ or ‘off’ of the 20 ftL background illumination. Fast oscillation
monitor the light-rise. A Ganzfeld dome, rather than an X-ray peaks, indicated on normal EOG by closed arrows, are normal for the
viewing box (or equivalent), should be used so that the entire patients. Peaks of the light-rise are indicated by open arrows, and are
prolonged and/or subnormal for the patients.
retina will be illuminated as evenly as possible. Otherwise the
From Weleber RG: Fast and slow oscillations of the electrooculogram in Best’s
EOG will reflect primarily only posterior pole function.55 Inter- macular dystrophy and retinitis pigmentosa. Arch Ophthalmol 1989; 107:530.
vals of ~12 min of darkness and 12 min of illumination are said
to reduce the normal variation.56 The maximal voltage in the
light is compared with the minimal voltage in the dark to derive showed a sinusoidal temporal pattern in which the ratio was
the light-rise to dark-trough ratio, which is normally greater than highest in the early morning and late afternoon and lowest
1.8 in patients younger than 50 years. Care should be taken around midday.
that the onset of illumination is not too abrupt or else the tested
eye might begin to tear, which could alter electrode resistance. Fast FULL-FIELD ELECTRORETINOGRAPHY
oscillations may be elicited with light-dark periods of 2.5 min
and can be immediately followed by monitoring the dark phase The normal human ERG elicited by a moderate-intensity white
of the conventional EOG.54 Responses from each eye should be flash from the dark-adapted eye consists of a cornea-negative
differentially amplified at a gain of ~200 (0.1-100 Hz) and deflection, called the a-wave, followed by a cornea-positive
displayed on an x–y plotter or digitized and displayed by com- deflection, called the b-wave (Fig. 124.11). The a-wave is known
puter. Repeat recordings on a given patient should be done at to reflect photoreceptor function,65,66 and the b-wave is
approximately the same time of day because of the presence of generated by Müller’s cells reflecting activity in the inner
an underlying circadian rhythm (see further on). nuclear layer of the retina.67,68 In actuality, the recorded b-wave
Intraindividual variability in the light-rise to dark-trough ratio is a summation of photoreceptor and more proximal retinal
of the EOG generally does not exceed 60%.57–59 The normal range function, since the photoreceptor component continues beyond
for the light-rise to dark-trough ratio has been placed at 1.9–2.8,60 the onset of the b-wave.66 Several wavelets, known as oscillatory
although values of 1.5–3.4 have been reported.61 It should be potentials (Fig. 124.11), may normally be seen superimposed on
noted that the ratio increases with luminance.62 The ratio also the ascending portion of the b-wave. These wavelets reflect bipo-
appears to decline with age, at least among women.60,61,63 Signifi- lar cell responses generated by feedback from amacrine cells.69,70
cant differences in the ratio between sexes have been reported, It is also possible to record a positive response – the d-wave – to
with larger values for females.59,61 Between 20% and 50% of the light offset, most typically for square-wave light modulation
variation in the EOG light-rise may be due to circadian rhyth- but, recently, also to sawtooth-modulated stimuli.71,72 If the
micity.64 Recordings done at 2-h intervals for six normal subjects conventional brief strobe- or photo-flash is used to elicit the 1613
SECTION 10 RETINA AND VITREOUS

FIGURE 124.12. Dark-adapted ERGs recorded from a normal


subject, a patient with juvenile X-linked retinoschisis, and a patient
with congenital nyctalopia with myopia in response to a full-field white
flash. Calibration – 100 mV vertically and 50 ms horizontally.

compared to their positive d-wave to light offset (Fig. 124.13).71


The authors concluded that the patients had a greater
impairment of their depolarizing bipolar system than their
FIGURE 124.11. Dark-adapted ERGs recorded from a normal subject hyperpolarizing bipolar system. Selective loss of oscillatory
in response to full-field white flashes of varying integrated luminance. potentials, in contrast, is usually interpreted as specifically reflect-
Traces begin at flash onset. Cornea-negative a-wave and cornea- ing inner retinal ischemia, as occurs in diabetic retinopathy75
positive b-wave are designated by letters; oscillatory potentials are and central retinal vein occlusion (Fig. 124.14).76
designated by asterisks. Arrow points to inflection in the a-wave, Careful inspection of the a-wave reveals an inflection (see
representing a combination of cone and rod components.
Fig. 124.11, arrow) that reflects a summation of cone and rod
From Sandberg MA, Lee H, Gaudio AR, et al: Relationship of oscillatory potential
amplitudes to a-wave slope over a range of flash luminances in normal subjects.
components of differing time course. Although not apparent, the
Invest Ophthalmol Vis Sci 32:1508, 1991. © Association for Research in Vision same is true of the b-wave. The fact that the normal dark-adapted
and Ophthalmology. ERG is a summation of rod and cone components may be demon-
strated in two ways. First, placing in turn a blue filter or a red
filter scotopically matched to the blue filter (i.e., matched in
brightness for the rods) in front of the eye and then flashing a
ERG, it is thought that the response lacks the positive d-wave white light results in different waveforms (Fig. 124.15). The
that otherwise occurs at light-offset.73 waveform with the blue filter in front of the eye consists of a
Separation of the ERG into a-wave, b-wave, and oscillatory late-onset, bell-shaped b-wave, whereas that with the red filter
potentials has important application for objectively diagnosing, in front of the eye consists of an early-onset a-wave and b-wave
classifying, and staging retinal diseases. For example, in (with oscillations) followed by the same late-onset b-wave. The
response to a moderate-intensity white light presented to the early components observed for red, but not blue, light represent
dark-adapted eye, loss of the a-wave with a slowing of the b-wave cone activity, whereas the late b-wave represents rod activity. When
in patients with clear media may signify a photoreceptor degen- the cone and rod components are well separated in time, they
eration in which photoreceptors have lost optical density. This appear to summate linearly.77–79 However, when the two b-waves
follows from the fact that in normal eyes reducing stimulus occur at the same time with near-maximal amplitudes, as for
intensity affects a-wave amplitude before b-wave amplitude (see the conventional white flash, the summation appears to be non-
Fig. 124.11). Conversely, loss of the b-wave with preservation of linear, as may be seen by using a method of digital subtraction.80
the a-wave may signify retinoschisis74 or congenital nyctalopia Linear subtraction of a cone-isolated response to red light from
with myopia1 (Fig. 124.12). In both of these conditions, synaptic a mixed cone and rod response to a bright blue light photo-
transmission from photoreceptors to more proximal elements is pically matched to the red light yields a rod-isolated waveform
disturbed. When ‘on’ versus ‘off ’ responses to sawtooth stimuli in which the b-wave is ‘scalloped’ (Fig. 124.16). This implies
were compared in patients with retinoschisis, the investigators that the b-wave to the bright blue light represents a sublinear
found that the patients with retinoschisis had a normal a-wave summation of rod and cone components. Conversely, even for
but a b-wave to light onset that was relatively reduced when these bright lights, the two a-waves appear to summate linearly.
1614
Objective Assessment of Retinal Function

FIGURE 124.13. ERG waveforms of patients


with X-linked retinoschisis (XLRS) (panels 1 and
3) and control subjects (panels 2 and 4) in
response to 8-Hz rapid-on (panels 1 and 2) and
rapid-off (panels 3 and 4) sawtooth flicker.
Dashed lines: time of stimulus onset; stimulus
waveform is illustrated on the x-axis. The
waveforms are arranged in order of increasing
b-wave amplitude. Numbers next to the
waveforms in the left panel refer to patient
designations.
From Alexander KR, Fishman GA, Barnes CS,
Grover S: ON-response deficit in the electroretinogram
of the cone system in X-linked retinoschisis. Invest
Ophthalmol Vis Sci 2001; 42:453.

CHAPTER 124
Isolation of rod and cone contributions to the ERG, like com-
parative analysis of the a-wave and b-wave components, is also
important for classifying retinal diseases. For example, selective
loss of cone function may signify congenital rod monochroma-
tism or advanced cone degeneration, whereas loss of a rod con-
tribution may signify an early stage of dominant retinitis
pigmentosa. In the cone-isolated response to flicker, the high
rate of presentation and sinusoidal nature of the waveform in
cases of advanced retinitis pigmentosa make it possible to
resolve amplitudes as small as 0.05 mV with signal averaging
and electronic filtering (see further on), which can be used to
follow up the course of this condition.83
The full-field ERG may also be used to quantify the amount
of remaining function for each of the three cone mechanisms.
The middle-wavelength or green-cone system and the long-
wavelength or red-cone system may be evaluated by comparing
cone ERGs elicited by a short-wavelength flash or a photopically
matched long-wavelength flash superimposed on a photopic
background (see Fig. 124.15)78 or flickering at 30 Hz.84 If the
responses are equivalent in amplitude, the patient is considered
to have comparable numbers of cones of each type, as has been
observed in carriers of blue-cone monochromatism.85 However,
if the two responses are unequal in amplitude or differ by a factor
of two or greater, then one of the two cone systems is considered
to be reduced in number relative to the other or absent,
respectively.84 Use of a short-wavelength flash superimposed on
a bright white background has been shown to isolate in time the
short-wavelength or blue-cone system from the other systems
(Fig. 124.18).86
Interest has developed in clinical recording of b-wave ampli-
tude versus flash intensity functions under conditions of dark
FIGURE 124.14. Dark-adapted ERGs recorded from a normal adaptation in patients with retinal disease. The intent of this
subject, a patient with central retinal vein occlusion (CRVO), and a approach is to distinguish changes in maximal amplitude (Vmax),
patient with diabetic retinopathy in response to a full-field bright white which are thought to reflect cell number and response gain,
flash. from changes in sensitivity (k), which are thought to reflect outer
segment optical density and media clarity. Some studies have
used white flashes to elicit these functions,87–89 which complicates
The presence of a steady white background that desensitizes rods interpretation because of the variable summation of cone and
and eliminates their contribution to the ERG reveals a ‘photopic’ rod contributions in diseases that may affect these two systems
a-wave and b-wave from the cone system (Fig. 124.17).81 In unequally. One study used digital subtraction to isolate rod func-
addition to background illumination, a flash rate of 30 Hz may be tion in patients with retinitis pigmentosa or cone–rod degen-
used to isolate cone function to white light (see Fig. 124.15).78 eration and showed that reductions in sensitivity, irrespective of
This is true because the rod system is normally incapable of changes in maximal amplitude, may be used to infer losses of
responding to these rates of flicker. Lights of different wave- rod photoreceptor optical density.90 Another study showed that
length and background adaptation have been used to demonstrate patients with an apparently rare form of retinal degeneration could
both a rod and a cone contribution to oscillatory potentials.82 have increased maximal rod b-wave amplitude with reduced
1615
RETINA AND VITREOUS

FIGURE 124.15. Full-field ERGs to scotopically matched red (column 1) and blue (column 2) flashes, to photopically matched orange (column 3)
and blue-green (column 4) flashes in the presence of 5-10 ftL of background light and to 30-Hz white flashes (column 5) are shown successively
from top to bottom for a patient with Nougaret-type night blindness, a normal subject, and a patient with advanced cone degeneration. Two or
three responses to the same stimulus are superimposed. Calibration – 60 ms horizontally and 50 µV vertically for columns 1 and 2; 30 ms
horizontally and 50 µV vertically for columns 3 and 4; 60 ms horizontally and 100 µV vertically for column 5. Corneal positivity is an upward
SECTION 10

deflection. Stimulus onset, vertical hatched line for columns 1–4; shock artifacts for column 5.
From Berson EL, Gouras P, Hoff M: Temporal aspects of the electroretinogram. Arch Ophthalmol 1969; 81:207-214. Copyright 1969, American Medical Association.

FIGURE 124.16. Computer-averaged dark-adapted full-field ERGs


from a normal subject to photopically matched blue (top) and red FIGURE 124.17. Analysis of ERG in a dark-adapted human eye as
(middle) flashes and the result of subtracting the second response the resultant of photopic (dashed line) and scotopic (dotted line)
from the first to derive a rod ERG in isolation (bottom). The rod components. The a-wave is composed of photopic (ap) and scotopic
component elicited by the red flash had already been eliminated by (as) components, and the b-wave is similarly composed of photopic
subtracting the response to a scotopically matched blue flash. In the (bp) and scotopic (bs) components. The solid line shows the algebraic
bottom waveform, the lower solid line represents the result of sum of the lower waveforms.
subtraction and the hatched area illustrates the suggested rod b-wave From Armington JC, Johnson EP, Riggs LA: The scotopic a-wave in the
correction for nonlinear summation of the cone and rod components electrical response of the human retina. J Physiol (Lond) 1952; 118:289.
to blue light.
From Sandberg MA, Miller S, Berson EL: Rod electroretinograms in an elevated
cyclic guanosine monophosphate-type human retinal degeneration. Comparison
with retinitis pigmentosa. Invest Ophthalmol Vis Sci 1990; 31:2283.
© Association for Research in Vision and Ophthalmology.

1616
Objective Assessment of Retinal Function

FIGURE 124.18. Full-field ERGs elicited by red


and blue flashes at different levels of
background illumination indicated by the
photopic troland values on the left above each
trace. L, M, and S signify the responses of,
respectively, the long-wavelength-, the middle-
wavelength-, and the short-wavelength-
sensitive cone systems. Calibration – vertically
4 µV for the upper four and 40 µV for the lower
four traces and 9 ms horizontally.
From Gouras P, MacKay CJ: Electroretinographic
responses of the short-wavelength-sensitive cones.
Invest Ophthalmol Vis Sci 1990; 31:1203.
© Association for Research in Vision and
Ophthalmology.

CHAPTER 124
sensitivity, as well as implicit times that were more delayed for
dim flashes than for bright flashes (Fig. 124.19), a combination
that could better be explained by an elevation of retinal cyclic
guanosine monophosphate than in terms of cell and optical
density loss.80
A corresponding approach has been used by a few laboratories
to evaluate the a-wave and, by inference, the photoreceptor
response. a-Waves were elicited by a series of flash intensities
that included intensities far brighter than those needed to saturate
the b-wave. A computational model was then applied to the
leading edge of the a-wave to estimate parameters of the photo-
transduction cascade. Representative cone a-waves obtained in the
presence of a rod-saturating background and rod a-waves derived
from subtracting the cone a-waves from dark-adapted responses
of a normal observer and the fitted functions are illustrated in
Fig. 124.20.91 With such fitted functions it is possible to
estimate the maximal amplitude, sensitivity, and delay of the
respective photoresponse. Normative values are presented in
the afore-mentioned paper.91 When this methodology was applied
to patients with retinitis pigmentosa, the authors found that
cone and rod a-waves were quantifiable in 30–33% of cases and,
in those cases, the maximum amplitude and sensitivity parameters
were below normal in all subgroups.92 In this cross-sectional
study there was no evidence that phototransduction efficiency
decreased with increasing age.
Full-field ERGs, like EOGs, are conventionally elicited with a
Ganzfeld dome (Fig. 124.21) that provides a nearly homoge-
neous distribution of light over the central 120° of the retina.93 FIGURE 124.19. Rod b-wave amplitude and implicit time (i.e., time to
Although retinal illuminance falls as a consequence of decreas- peak) versus retinal illuminance functions for 17 normal subjects
ing apparent pupillary area for retinal eccentricities greater than (mean ± SD) and three patients with an elevated cyclic guanosine
60°, this is compensated in large part by the curvature of the monophosphate-type retinal degeneration. Rod ERGs were elicited
retina and by reduced light absorption in the ocular media with with full-field blue flashes at low retinal illuminances and by a method
eccentricity.94 With virtually uniform retinal illumination, the of digital subtraction involving photopically matched blue and red
faster cone and slower rod components of the ERG across the flashes at high retinal illuminances.
retina respond with a minimal variation in latency and there- From Sandberg MA, Miller S, Berson EL: Rod electroretinograms in an elevated
cyclic guanosine monophosphate-type human retinal degeneration. Comparison
fore may be separated in time and quantified. In addition, this
with retinitis pigmentosa. Invest Ophthalmol Vis Sci 1990; 31:2283.
retinal light distribution is altered little by small changes in eye
© Association for Research in Vision and Ophthalmology.
position, which fosters reproducibility between successive
responses.93,95
The eye(s) to be tested should be initially dilated and adapted
to the dark for at least 45 min. Dilatation maximizes amplitudes best done with a bipolar contact lens electrode, with the positive
and generally minimizes implicit times.96,97 Complete dark adap- electrode being a ring around the contact lens and the negative
tation, which may require 45 min or longer depending on the level or reference electrode a conductive coating on a lid speculum
of prior exposure, also maximizes amplitudes (Fig. 124.22)98 (Fig. 124.23). The bipolar configuration, in which the lid versus
while also tending to maximize implicit times. Recordings are ground response is subtracted from the cornea versus ground 1617
RETINA AND VITREOUS

FIGURE 124.20. Representative a-waves from


a b a 65-year-old control subject. (a) Responses in
dark to intensities ranging from 3.2 to 4.4 log
scotopic troland-seconds (log sc td-s).
(b) Same 4 intensities presented against a
3.2-log td background. (c) Rod-isolated
responses and fitted functions (dashed lines).
(d) Cone responses and model fits (dashed
lines.
From Birch DG, Hood DC, Locke KG, et al:
c d Quantitative measures of phototransduction in cone
and rod photoreceptors. Normal aging, progression
with disease, and test-retest variability. Arch
Ophthalmol 2002; 120:1045.
SECTION 10

FIGURE 124.22. Computer-averaged full-field rod ERG percent


amplitudes to a 0.5-Hz 10-µs blue flash (max 440 nm) of 16 ftL
recorded over time in the dark from a normal observer after a 10-min
full-field white light bleach of 10 or 500 ftL presented to the dilated
pupil. Amplitudes for 60 min of dark adaptation were arbitrarily
designated 100%. All the data appear to reflect rod function, except
for the low-amplitude 5-min value after the stronger bleach, which
probably represents residual cone function. Each set of data
represents a single run.
From Sandberg MA: Technical issues in electroretinography. In: Heckenlively J,
Arden G, eds. Principles and practice of clinical electrophysiology of vision.
St Louis: Mosby-Year Book; 1991.

factually reduce ERG amplitudes and increase implicit times.


FIGURE 124.21. Ganzfeld stimulator. Flashlamp enclosed in case and Some facilities use alternative electrodes for recording ERGs. In
attached to the top of the diffusing sphere illuminates the inner white this country, these include the disposable Jet electrode, the Arden
surface of this dome (40 cm in diameter), providing a full-field stimulus. gold foil electrode, and the Dawson–Trick–Litzkow (DTL) fiber
Lights are recessed in the top of the dome so that the patient can be electrode. All three are monopolar electrodes that will give
tested in the presence of steady full-field background light. larger voltages than the bipolar Burian–Allen electrode,99 but
From Rabin AR, Berson EL: A full-field system for clinical electroretinography.
also be more subject to artifact contamination.
Arch Ophthalmol 1974; 92:59. Copyright 1974, American Medical Association.
With the patient fixating on a red light-emitting diode, 0.5-Hz
dim, short-wavelength flashes may be given first to isolate the
rod ERG. Next, 0.5-Hz scotopically matched long-wavelength
flashes can be given to obtain a mixed response consisting of a
response, localizes the response to the eye and provides the best faster cone component and a slower rod component of amplitude
elimination of surrounding 60-Hz noise and any photovoltaic comparable with that elicited by the short-wavelength flash.
artifact that may be generated by the flash lamp. The lid spe- After that, 0.5-Hz dim white flashes may be given to elicit an
culum also prevents the upper and lower eyelids from partially a-wave and maximal b-wave consisting of a summation of both
covering the cornea and thereby obstructing the passage of light cone and rod components. Finally, 30-Hz dim white flashes or
into the eye. A reduction in retinal illuminance could arti- 0.5-Hz dim white flashes superimposed on a background to
1618
Objective Assessment of Retinal Function

single flashes because of reduced stimulus retinal illuminance.


An electronic photoflash, with ~1000 times the energy of the
conventional full-field flash when illuminating a Ganzfeld dome,
can be used to elicit larger responses from such eyes.103 In order
to separate the optical density effect of the media obstruction from
any change that may be due to a retinal abnormality, responses
to a series of stimulus intensities should be compared with
those obtained with conventional full-field flashes in normal
eyes (e.g., see Fig 124.11). If a-wave and b-wave amplitudes and
implicit times to the brighter flashes in eyes with opaque media
can be matched to those obtained to the dimmer flashes in normal
eyes with clear media, large areas of the retina can be considered
to be functioning normally.104 In eyes with large pupils and
relatively clear media, these bright flashes may be used to elicit
a maximal a-wave with oscillatory potentials superimposed on
the ascending limb of the b-wave.103 Bright flash stimulation
should be presented to the dark-adapted eye (i.e., before flicker
or background illumination), with each flash separated from the

CHAPTER 124
next by an interval of ~1 min to minimize light adaptation
from the preceding flash.
Full-field ERG responses may be photographed in real time
from an oscilloscope or digitized and stored for subsequent analy-
sis and hard copy. Digitization may be used to eliminate baseline
variation and to isolate oscillatory potentials, whose frequency
FIGURE 124.23. Double-electrode (Burian–Allen) contact lenses used content (50–180 Hz) exceeds that of the a-wave (<50 Hz) and
to obtain ERG responses. b-wave (<25 Hz).103,105–107 Oscillatory potentials may then be
From Rabin AR, Berson EL: A full-field system for clinical electroretinography. quantified either in conventional time domain or, by Fourier
Arch Ophthalmol 1974; 92:59. Copyright 1974, American Medical Association.
analysis, in frequency domain (Fig. 124.24). Consecutive digi-
tized responses may also be summed and averaged and, for 30-Hz
flicker, smoothed with narrow band-pass filtering to resolve very
small amplitudes (Fig. 124.25).83,108 However, for this purpose,
isolate a cone response may be given.95 For cases of generalized recordings should be performed with a bipolar contact lens elec-
cone degeneration or when abnormal color vision is found, the trode to minimize contamination of the waveform with electrical
spectral lights described earlier may be presented to assess the or photovoltaic artifacts. Figure 124.26 shows responses to 30-Hz
different cone systems across the retina. When isolating the cone xenon flashes recorded without and with narrow band-pass
response with flicker or background illumination, the retina may filtering from different electrodes placed in saline solution. The
need to be illuminated for several minutes before a maximal monopolar gold foil electrode generates a large artifact with con-
response is obtained.100–102 stant phase for both types of filtering. The monopolar Jet elec-
Patients with small dilated pupils or those whose pupils can- trode generates smaller responses reflecting an electrical artifact
not be dilated or, in some cases, those who have media opacities alone. In contrast, the two bipolar electrodes (the Burian–Allen
that obscure visualization of the fundus may have ERGs that and GoldLens) show minimal noise.109 With a well-conditioned
are smaller in amplitude than expected or nondetectable with Burian–Allen contact lens electrode, test–retest reproducibility for

FIGURE 124.24. Quantification of mean oscillatory potential amplitude in the time domain between the a-wave and the b-wave peaks after
62-Hz high-pass filtering for a normal subject in response to a bright full-field flash white flash. Vertical lines represent some of the amplitudes
whose absolute values with respect to baseline were summed before calculation of the mean (left). Quantification of mid- and high-frequency
amplitudes for oscillatory potentials by the magnitude fast Fourier transform for a normal subject in response to the same flash (right).
From Sandberg MA, Lee H, Gaudio AR, et al: Relationship of oscillatory potential amplitudes to a-wave slope over a range of flash luminances in normal subjects.
Invest Ophthalmol Vis Sci 1991; 32:1508. © Association for Research in Vision and Ophthalmology.
1619
RETINA AND VITREOUS

b-wave implicit time increases.110,114 The same trends have


been observed in the b-wave component isolated from the short-
wavelength-sensitive cones in response to a blue flash on a bright
background.115 These decreases in ERG amplitudes and increases
in ERG implicit times with increasing age probably reflect
decreasing retinal illuminance with increasing age caused by
reduced light transmissivity by the lens116 and, possibly, to a
smaller dilated pupillary diameter with increasing age. Similarly,
increased uveal pigmentation lowers amplitude, and decreased
uveal pigmentation raises it.117,118 Increased uveal pigmentation
related to race appears to reduce the maximal amplitude by an
average of 17%, but does not affect the sensitivity or the implicit
time of the b-wave as measured in response to blue flashes of
varying retinal illuminance.119 The ERG b-wave is also larger in
hyperopic eyes than in myopic eyes (Fig. 124.28)112,120,121 and is
slightly larger in women than in men.110,112,120,122,123
Intrasubject variation in amplitude in normal subjects across
days can be as much as ±25% for large responses. Normal sub-
SECTION 10

jects, unentrained to a cyclical pattern of illumination, have shown


no significant variations in rod ERG amplitude over daylight
hours.124 However, in normal subjects entrained to cyclical illu-
mination for at least 3 days, rod ERG amplitudes follow a regular
pattern, being on average 10–15% smaller 1.5 h after light onset
than at other times of day.124 The physiologic basis for this ERG
diurnal rhythm appears to be an alteration in photoreceptor
function associated with rod outer segment disk shedding, since
recordings in normal light-entrained rats have demonstrated an
ERG variation similar to that seen in humans and that is sig-
nificantly correlated with the number of phagosomes appearing
in the RPE.125

FOCAL CONE ELECTRORETINOGRAPHY


Since only some 7% of cone photoreceptors are in the central
macula, patients with macular degeneration typically retain nor-
mal full-field ERGs and must be assessed with a focal stimulus
in order to reveal and quantify malfunction.52 Foveal cone ERGs
have been used by various centers since the late 1960s to detect
macular malfunction.126–133 Abnormal foveal cone responses have
been found in patients with Stargardt’s disease,129,134 in which
amplitude varied directly with visual acuity, and in those with
FIGURE 124.25. ERGs elicited with full-field white 30-Hz flashes from age-related macular degeneration (Fig. 124.29).130,135 Patients
a normal subject and four patients with different genetic types of RP with age-related macular degeneration may also show delays in
at ages spanning intervals of 11–15 years. Responses were recorded foveal cone ERG implicit time that are associated with abnormal
without (left column) or with (right column) computer averaging and choroidal perfusion.136 In patients with macular holes, foveal
band-pass filtering. Stimulus onset, vertical markers. Calibration (left amplitude varied inversely with hole diameter, and subnormal
column, lower right) – 100 µV vertically for the normal subject and the responses appeared to predict impending holes in the fellow eye.137
top three patients; 40 µV vertically for the bottom patient; 50 ms
Foveal ERGs may be abnormal in symptomatic patients even
horizontally for all traces. Calibration (right column, lower right) – 2 µV
vertically for the dominant, X-linked, and isolate patients; 0.3 µV for when no diagnostic abnormalities can be seen by ophthalmoscopy
the recessive patient; 20 ms horizontally for all traces. The b-wave and fluorescein angiography,138,139 and thus they provide an aid
implicit times are designated with arrows. in early detection of macular malfunction (i.e., the so-called ‘occult
From Andreasson SOL, Sandberg MA, Berson EL: Narrow-band filtering for maculopathy’). Conversely, foveal cone ERGs are normal in
monitoring low-amplitude cone electroretinograms in retinitis pigmentosa. patients with strabismic amblyopia140 or optic neuropathy,140–142
Am J Ophthalmol 1988; 105:500, copyright 1988, from Elsevier Science. indicating that the test provides a differential diagnosis for dis-
eases of the macula versus diseases of the optic nerve or visual
cortex (Fig. 124.30). Focal cone ERGs should, in general, be
sub-mV full-field cone ERGs to 30-Hz flashes revealed highly used in conjunction with full-field recording in order to rule out
correlated implicit times and amplitudes.109 Signal averaging may generalized cone degeneration, which may not have visible signs
also be used to reduce large voltages due to eye movements, as, of peripheral retinal disease.
for example, in cases of nystagmus. Focal cone ERGs may be elicited with a commercially avail-
The variation in full-field ERG amplitude for a given stimulus able hand-held stimulator ophthalmoscope, the Maculoscope.
condition across normal subjects is as much as 2.5:1.110,111 Age, With this instrument, the examiner can visualize the fundus
sex, refraction, ocular pigmentation, and time of day may all through a short-stalk Burian–Allen bipolar contact lens electrode
contribute to this variation: with increasing age among adults, over the dilated pupil. A 42-Hz, white, 4° diameter stimulus is
a-wave amplitude declines,110 b-wave amplitude declines,110,112–114 placed on the fovea and centered within a steady white 12° sur-
oscillatory potential amplitude declines (Fig. 124.27), and round. This stimulus/surround configuration allows the
1620
Objective Assessment of Retinal Function

FIGURE 124.26. Responses to full-field 30-Hz


xenon flashes of 1.3 log td-s without (left) and
with (right) narrow-band filtering from four
different electrodes placed in saline solution.
From Birch DG, Sandberg MA: Sub-microvolt full-field
cone electroretinograms: artifacts and reproducibility.
Doc Ophthalmol 1997; 92:269. Reprinted from Kluwer
Academic Publishers, Spuiboulevard 50, P.O. Box 17,
3300 AA Dordrecht, The Netherlands.

CHAPTER 124
examiner to visually track an area to be tested even in patients 16 min from up to 241 locations within the central 50°, called
with eccentric or variable fixation. The high rate of flicker not the ‘multifocal ERG’.147 Although this approach assumes phase
only isolates cone function but also renders the response sinu- (implicit time) constancy between adjacent 5° locations and relies
soidal so that amplitudes less than 1 mV, which occur normally, on the voluntary fixation of the patient, the output is a detailed
can be resolved with the help of electronic filtering, computer three-dimensional plot of response amplitude by location.
averaging, and Fourier analysis.129 White flashes, rather than red, These a-wave and b-wave responses appear to be the same as
are used to avoid eliciting subnormal amplitudes from patients those obtained by conventional recording and, thereby, appear
with congenital protanopia. Focal cone ERGs may be performed to be of value in identifying which cells are contributing to the
in an unshielded room in dim ambient room illumination. Since retinal malfunction.148 This approach ought to be most useful for
foveal cone ERG amplitudes are very small and tend to increase characterizing maculopathies149–151 by revealing a relative foveal
during continued light adaptation,143 consecutive response amplitude depression (e.g., Fig. 124.31),150 but the diagnosis of
averages should be recorded over a period of a few minutes to maculopathy rests on the assumption that the patients
prove reproducibility. maintained stable central fixation throughout testing.
Foveal cone ERG amplitude has been reported to be inversely The multifocal ERG may be most appropriate for generating an
correlated with age for 100 normal eyes of subjects between the objective map of the central visual field of patients with gener-
ages of 5 and 75 years.130 A fourfold range for normal foveal cone alized retinal disease who retain central fixation. Several studies,
ERG amplitude has been reported.134 Although the reason for for example, have performed multifocal ERG testing on patients
this large normal range is unclear, it can be effectively reduced with early stages of retinitis pigmentosa. Based on a sample of
by 30% on average by adopting a foveal to parafoveal amplitude 111 patients with retinitis pigmentosa, 24% who were anti-
ratio.144 This approach should be used with caution, as some cipated to have sufficiently large signals for testing were found
regional variation in the parafoveal cone ERG has been to have a detectable multifocal ERG, consisting of a central peak
reported.145 The foveal cone ERG is also smaller in amplitude of variable diameter, with implicit times that tended to increase
in eyes with brown irides than in eyes with lighter-pigmented towards the periphery.152 A study on a small group of patients
irides, presumably because of the corresponding differences in with retinitis pigmentosa tried to correlate multifocal cone ERGs
uveal pigmentation.146 Normal intervisit variability in foveal with multifocal rod ERGs and multifocal ERG values with visual
cone ERG amplitude has not been reported. field sensitivities by location. The authors reported a significant
Another commercial instrument (the VERIS System) exists correlation between multifocal cone ERG amplitude and cone-
for recording focal cone ERGs simultaneously over an interval of mediated visual field sensitivity, but there were many instances
1621
RETINA AND VITREOUS

FIGURE 124.28. Linear regression of b-wave amplitude on refraction


SECTION 10

based on average of regression equations for men and women from


measurements on 86 normal subjects. Maximal b-waves were elicited
from the semi-dark-adapted eye with white light flashes derived from
a lamp at different distances from the subject and presented through
the dilated pupil.
From Sandberg MA: Technical issues in electroretinography. In: Heckenlively J,
Arden G, eds. Principles and practice of clinical electrophysiology of vision.
St Louis: Mosby-Year Book; 1991. Data from Pallin E: The influence of the axial
size of the eye on the size of the recorded b-potential in the clinical single-flash
electroretinogram. Acta Ophthalmol 1969; 101(Suppl):1.

these results suggest that multifocal ERG recording may not be


a useful additional test to detect carriers of X-linked retinitis
pigmentosa.
Multifocal ERG recording has also been used to reveal the dis-
tribution of retinal malfunction in patients with X-linked juvenile
retinoschisis.157,158 These patients present with a foveal schisis
in all cases and a peripheral schisis in 50% of cases. Multifocal
ERG recording has demonstrated a greater amplitude loss cen-
trally than peripherally, and amplitudes have been found to be
normal even in areas with visible peripheral schisis. The multi-
FIGURE 124.27. A-wave, b-wave, and oscillatory potential amplitude focal ERG waveform also generally does not show the negativity
versus age in normal subjects. Responses were elicited from dark- characteristic of the full-field ERG, perhaps because of constraints
adapted eyes with full-field dim white flashes for the upper and middle in the software algorithm that derives the multifocal ERG wave-
graphs or bright white flashes for the lower graph. Oscillatory potential form. Interestingly, multifocal ERG implicit times have been
amplitudes are calculated as described for Figure 124.24. found to be delayed at all, or nearly, all tested locations, indi-
cating involvement of areas without visible schisis and areas
with normal amplitudes.

of reduced amplitude with normal sensitivity. The correlations FOCAL ROD ELECTRORETINOGRAPHY
between rod ERG amplitude and rod-mediated visual field sen-
sitivity and between cone ERG amplitude and rod ERG ampli- Since rod photoreceptors are especially sensitive to reflected light,
tude were very low, perhaps due to methodological problems in it has been impossible, until recently, to record rod ERGs to bright
isolating multifocal rod ERGs.153 flashes of light from localized areas. In addition to allowing one
Although full-field ERG recording has been shown to detect to characterize transduction among homogeneous populations
retinal malfunction in 90–96% of obligate carriers of X-linked of rod photoreceptors,159 these techniques allow the assessment
retinitis pigmentosa,154,155 complementary tests of localized of retinal function in areas outside of the macula that are not
function could potentially enhance this sensitivity by revealing an easily assessed by focal cone ERGs because cone photoreceptors
underlying mosaicism as predicted by random X-chromosome are normally scarce. For example, Figure 124.32 shows dark-
inactivation. Multifocal ERG recording in five carriers from adapted focal rod ERGs from a patient with the multiple
three families revealed local amplitude losses and implicit time evanescent white dot syndrome and an enlarged blind spot in her
delays in two patients, local amplitude losses alone in one patient, left eye.160 In comparison with the large, symmetric focal responses
local implicit time delays alone in one patient, and no abnor- recorded from the nasal and temporal retina of a normal subject,
mality in one patient. In the two patients with both types of the response from the nasal retina (within the scotoma) was non-
defect there was poor correspondence between the locations show- detectable whereas the response from the temporal retina was
ing amplitude losses and the locations showing implicit time normal in the patient. These recordings prove that her visual
delays.156 Although based on only a small number of patients, loss was of retinal origin.
1622
Objective Assessment of Retinal Function

FIGURE 124.29. Representative foveal cone


ERGs from a normal subject and four patients
with AMD. Responses were recorded with a
stimulator ophthalmoscope. Each 100-ms trace
contains 4 responses to the 42-Hz stimulus.
Two consecutive traces (each the average of
200 sweeps) are shown for each patient.
Arrows indicate b-wave implicit time (time
between light flash and response).
From Fish GE, Birch DG, Fuller DG, et al: A
comparison of visual function tests in eyes with
maculopathy. Ophthalmology 1986; 93:1177–1182.

CHAPTER 124
One approach involving three steps allows one to record the retinal illuminance (d). The focal response appeared to be gener-
focal rod a-wave.159 In step 1, ERGs are recorded from the dark- ated entirely by rods because the ERG to a 30°-diameter red flash
adapted eye in response to a 40°-diameter stimulus. In step 2, that would elicit the same cone response as the blue flash was
ERGs are recorded in response to the same stimulus 1 s after a nondetectable for these recording conditions (e). When using a
white conditioning flash of the same diameter. The conditioning 10°-diameter stimulus, the stray light component may not over-
flash is used to desensitize rods directly illuminated by the flash lap the focal component that can then be quantified without
without significantly affecting the a-wave generated by rods and resorting to a subtractive technique.
cones illuminated by reflected light. Subtracting the a-wave
obtained in step 2 from that obtained in step 1 derives the a-wave PATTERN ELECTRORETINOGRAPHY
generated by rods directly illuminated by the flash. The validity of
this method rests on the assumption that the rods and cones out- Pattern electroretinography uses a spatiotemporal exchange of
side the stimulus are not light-adapted by the conditioning flash. stripes or checks in which mean luminance is held fixed to elicit
A second technique allows recording both a-waves and b-waves a focal ERG.161 The theory is that local, time-varying changes in
from the dark-adapted eye in response to stimuli as small as 10°.160 luminance responses generated by photoreceptors cancel at the
This technique is illustrated in Figure 124.33. The response to cornea (or at least make a minimal contribution to the response),
a 30°-diameter focal blue flash (a) consists of a faster, smaller whereas cells with center-surround receptive fields (i.e., retinal
‘focal’ component and a slower, larger ‘stray light’ component. ganglion cells) are strongly stimulated. One defense of this idea
The response to some dimmer full-field blue flash (b) consists is that the pattern electroretinogram (PERG) shows spatial
of a negative deflection (a-wave) followed by a positive deflection tuning under some situations. That is, amplitude is larger for
(b-wave) that closely matches the stray light b-wave of the first stimulus elements of medium size and smaller for stimulus
response. Subtraction of the second from the first response (a–b) elements of smaller or larger sizes (Fig. 124.34).162–170 In other
results in isolation of a local response (c) with an a-wave and instances, however, a low-pass relationship occurs in which
b-wave that are smaller than, but otherwise similar to, those of amplitude increases monotonically with the size of the
the full-field, rod-isolated response to a blue flash of the same stimulus element.168 In these cases, primarily resulting from
1623
RETINA AND VITREOUS

FIGURE 124.30. Foveal and parafoveal cone


ERGs recorded with a stimulator
ophthalmoscope from a normal subject and
four patients with visual acuity 6/60. Two or
more consecutive computer summations
(n = 128) are shown. Vertical lines denote
stimulus onset; arrows denote b-wave implicit
time to corresponding response peak.
Calibration symbol in lower right corner
denotes 20 ms horizontally and 0.25 µV
vertically.
From Jacobson SG, Sandberg MA, Effron MH, Berson
EL: Foveal cone electroretinograms in strabismic
amblyopia. Comparison with juvenile macular
degeneration, macular scars and optic atrophy. Trans
Ophthalmol Soc UK 1979; 99:353.
SECTION 10

FIGURE 124.31. Multifocal ERGs and the


corresponding three-dimensional plot for a
normal control subject (left) and a patient with
occult macular degeneration (right).
From Piao CH, Kondo M, Tanikawa A, et al: Multifocal
electroretinogram in occult macular dystrophy. Invest
Ophthalmol Vis Sci 2000; 41:513.

high-luminance, high-contrast targets, a significant luminance supports the idea that it derives from ganglion-cell activity. It
component is thought to contaminate the responses to large has been reported to be both more sensitive179 and less
stimulus elements.162,167 Luminance components presumably sensitive180 than full-field oscillatory potentials in detecting inner
arise from nonlinear summation of opposite sign responses to retinal malfunction in patients with early stages of diabetic reti-
on and off stimulation.169,170 nopathy. It remains controversial whether the PERG tends to be
Unlike the focal luminance-evoked ERG (usually referred to abnormal in ocular hypertension, as some studies have found
as the focal ERG or FERG), the PERG has been found to be many abnormal responses,181,182 whereas others have found mostly
predominantly abnormal in patients with glaucoma,171–174 optic normal responses.183 It also remains controversial whether the
neuritis,175,176 or optic atrophy (Fig. 124.35),177,178 which further PERG is abnormal in amblyopia, as some studies have found
1624
Objective Assessment of Retinal Function

CHAPTER 124
b

FIGURE 124.32. (a) Kinetic visual field for the left eye (visual acuity =
20/25) of a patient with multiple evanescent white dot syndrome
(MEWDS) and an enlarged blind spot. The visual field was to a V4e
white test light presented with a Goldmann perimeter on a
background luminance of 31.5 asb. The blind spot normally subtends
~5° between 10° and 20° eccentricity in the temporal field. (b) Focal
rod ERGs to a 30°-diameter blue flash of 2.1 log scot. td.-s. along the
horizontal meridian at an eccentricity of 25° in the temporal or nasal
field of the eye of a normal volunteer and the affected eye of the
patient. Responses were computer averaged (n = 4). Focal rod ERGs
for another normal volunteer (not illustrated) had b-wave amplitudes
that were 44 µV temporally and 43 µV nasally.
(a and b) From Sandberg MA, Pawlyk BS, Berson EL: Isolation of focal rod
electroretinograms from the dark-adapted human eye. Invest Ophthalmol Vis Sci
1996; 37:930. Reprinted from Lippincott-Raven Publishers, 227 East Washington
Square, Philadelphia.

mainly abnormal responses,184–186 whereas others have found


normal responses.165,187 FIGURE 124.33. ERGs from a normal volunteer. (a) ERG to a 30°-
Based on the preceding normative and clinical studies, studies diameter blue flash of 2.1 log scot. td.-s. at a temporal retinal
in mammals involving section of the optic nerve,188,189 and cur- eccentricity of 30°. (b) Full-field ERG to a blue flash of 0.4 log scot.
rent source density analysis,190 it is now fairly certain that the td.-s. (c) Subtraction of (b) from (a). (d) ERG to a full-field blue flash of
PERG derives, at least in part, from the ganglion cells. However, the 2.1 log scot. td.-s. after subtraction of the response to a photopically
matched full-field red flash. (e) ERG to a 30°-diameter red flash at a
PERG should not be used alone to diagnose the site of retinal mal-
temporal retinal eccentricity of 30°s and photopically matched to the
function, since it necessarily depends on photoreceptor function blue flash used to elicit (a). Responses were computer averaged
and has been shown to be abnormal in macular disease involving (n = 4). A second normal volunteer gave similar ERGs (not illustrated).
the photoreceptors.191 If the PERG is abnormal, it would also be From Sandberg MA, Pawlyk BS, Berson EL: Isolation of focal rod
advisable to obtain a focal flash ERG to evaluate photoreceptor electroretinograms from the dark-adapted human eye. Invest Ophthalmol Vis Sci
function before assuming that the inner retina is abnormal. 1996; 37:930. Reprinted from Lippincott-Raven Publishers, 227 East Washington
PERGs are usually elicited by phase-reversing square-wave Square, Philadelphia.
stripes or checks presented on a television monitor or modulated
by a moving mirror within an optical system. The temporal modu- positive deflection (see Fig. 124.35), much like the luminance
lation may be at a low frequency (i.e., <8 Hz) to elicit transient ERG. The temporal modulation is usually square wave but may
responses (see Fig. 124.35), or at a high frequency (i.e., ≥8 Hz) to be sinusoidal. Contrast (i.e., the luminance difference between
elicit a steady-state, sinusoidal response. The transient response light and dark elements relative to the mean luminance) may be
consists of a cornea-negative deflection followed by a cornea- 100% or less. Field size is typically 20° or less, but the pattern
1625
RETINA AND VITREOUS

FIGURE 124.34. Pattern ERG amplitude and


phase versus spatial frequency for a normal
subject. Responses were elicited with a
1.6 µ 1.7°, 100% contrast sinusoidal grating
reversing sinusoidally at 8 Hz. Responses were
recorded with a gold-foil electrode.
From Hess RF, Baker CL Jr: Human pattern-evoked
electroretinogram. J Neurophysiol 1984; 51:939.
SECTION 10

must contain an even number of spatial elements in order to between males and females,195 but a small decline in ampli-
minimize any luminance component, particularly if low spatial tude195,202 and an increase in latency195 with increasing age have
frequencies are used. been reported. In a comparison of responses for a group of normal
The patient’s pupil may or may not be dilated. In either case, subjects of mean age 21 years with a group of normal subjects
care must be taken that the patient is properly refracted and can of mean age 72 years, the latter had an average 40% reduction
focus on the pattern, which is usually placed between 0.5 and 1 m in amplitude.203
distant. A pattern that is not focused will have reduced retinal
contrast and result in a reduction in amplitude.162,165 Two diopters VISUAL EVOKED RESPONSE TESTING
of blur may reduce the amplitude by 50%.165 The probability that
patterns will be out of focus for patients with reduced acuity must Pattern-reversal VERs are now preferred over flash VERs for the
be considered when interpreting responses, an issue that has evaluation of the visual pathways, owing in large part to their
been raised with respect to evaluating strabismic amblyopia.165 smaller range of variation in normal subjects and, undoubtedly
Signals may be monitored with a silver cup skin electrode on related to this, their enhanced sensitivity in detecting axonal con-
the lower lid,192 a corneal gold foil or DTL electrode placed in duction defects. The pattern VER is a multiphasic response that,
the lower conjunctival sac,192–194 or a Burian–Allen bipolar contact when detectable, contains an inion-positive component with a
lens electrode.195 With the gold foil electrode, but apparently not latency of ~100 ms, called P100 (see Fig. 124.35). The primary
with the DTL electrode,196 pattern stimulation of one eye can use of the pattern-reversal VER is to identify visual loss second-
by volume conduction lead to an artifactual response from the ary to diseases of the optic nerve and anterior visual pathways
fellow eye.197–199 With the DTL electrode, the ratio of signal to versus those of psychogenic origin. For example, demyelination
noise was said to be best if the reference electrode was placed by and compressive lesions of the optic nerve produce reliable
the outer canthus of the stimulated eye.196 If a contact lens is used, slowing of the pattern response (Fig. 124.36), whereas flash
the eye must be refracted with it in place. PERG signals are typi- responses in these cases often have normal latencies.204 As many
cally less than 5 mV in amplitude and must be averaged, prefer- as 96% of patients with multiple sclerosis might be expected to
ably with an artifact reject buffer. The skin electrode is reported have delayed pattern VERs.205 Although P100 remains delayed,
to be little affected by blinking but yields smaller amplitudes its amplitude varies monotonically with visual acuity during
than does a corneal electrode.192 The contact lens electrode alone recovery from optic neuritis.206 Determining contrast thresholds
prevents reduction of stimulus luminance resulting from lid for a range of spatial frequencies permits the objective assess-
closure. ment of visual acuity (at 100% contrast),207 which can be used
Although the stimulus fields for eliciting PERGs are usually to detect malingering.
large relative to those used for eliciting FERGs, fixation near the The stimulus for the pattern VER may be essentially the same
center of the pattern is still important because the major contri- as for the PERG, allowing for both measures to be recorded simul-
bution to the response for a mid-frequency check size comes taneously, as is becoming increasingly the custom.163,184,195,200,202
from the fovea. It has been shown that PERG amplitude can fall It is essential that the checkerboard pattern be in proper focus,
by 50% for an eccentricity of 4°165 and by 63% for an eccentricity since lack of focus leads to decreases in VER amplitude208,209
of 12°.200 Eccentric fixation may also help to generate a lumi- and increases in VER latency.210 Recordings are generally made
nance component, which may be conveniently checked by quan- from the midline, using either a bipolar configuration with the
tifying the fundamental component of the response by Fourier positive electrode above the inion and the negative electrode on
analysis.166 the vertex or monopolar configurations with positive electrodes
Interocular differences in PERG amplitude among normal sub- above the inion and over the parietal lobe. Earlobes may serve
jects have been estimated to be as much as 100%.165 Test–retest as reference and ground. The electrodes may be conventional
correlations were 0.58 comparing two measurements made on the electroencephalographic cup electrodes and should be filled with
same day but only 0.01 comparing measurements made 1 week electrode cream and applied to the scalp after reducing scalp
apart.201 The PERG does not appear to be significantly different resistance with an abrasive. The intent is to obtain a resistance
1626
Objective Assessment of Retinal Function

Unlike retinal responses, scalp responses to pattern reversal


show marked interindividual variation in waveform among nor-
mal subjects.212 Nevertheless, remarkable agreement exists for
the normal latency for the P100 component across studies. Upper
limits for latency differences between left and right eyes of normal
subjects have been estimated at 8–10 ms.213 Repeat testing of
the same subjects on separate days showed a substantial variation
in latency with values that still mostly fell within normal limits
obtained on the same patients at a single session.213 P100 latency
increases with age in normal subjects, at a rate of ~2 ms/decade
depending on spatial frequency (Fig. 124.38).214 In addition, a
50-year increase among adults was associated with a 50%
reduction in VER amplitude for temporal modulation below 6 Hz.
For faster modulation, age had no effect on amplitude.203 VER
acuity (derived by extrapolating VER amplitude as a function of
increasing spatial frequency to zero amplitude) increased 300%
in infants for increasing age up to the adult level reached at age
30 weeks.215 P100 latency is also greater for small pupils than

CHAPTER 124
for large pupils.216 Some (but not all) of the increase with age
may be due to age-related miosis.

CHOOSING A TEST
When a patient presents with reduced visual acuity, blur, or meta-
morphopsia or with difficulty seeing at night, increased sensi-
tivity to bright lights, or an impairment in discriminating colors
(or any combination of these symptoms), the ophthalmologist
must decide whether objective tests of visual function are required
in addition to a routine examination. It has been our preference
to first rule out distal retinal malfunction by performing full-field
and focal flash electroretinography. Tests based on differential
fundus reflectance – especially OCT – can be used to corroborate
this impression. If results from these tests are abnormal, no addi-
tional measurements of function are necessary, and the patient
can be advised that the problem is within the eye in the outer
retina. If results from flash electroretinography are normal, it
would be appropriate to consider the PERG to rule out ganglion
FIGURE 124.35. Pattern-reversal ERGs (PERRs) (a) and VERs (b)
cell and optic nerve dysfunction, and then the pattern VER to
from the right and left eyes of a patient with a traumatic unilateral rule out visual pathway and cortical dysfunction.
(right) optic nerve section. Responses were elicited by a matrix of
40 min of arc dots, as illustrated at upper right. Arrows designate Key Features
major negative and positive components, respectively. The pattern • Measures optical density and regeneration kinetics of
ERGs were recorded simultaneously from both eyes with DTL
rhodopsin or cone visual pigment
electrodes. The pattern VERs were recorded sequentially with the
positive electrode 2 cm above the inion referenced to an earlobe. • Measures lipofuscin concentration
(a and b) From Dawson WW, Maida TM, Rubin ML: Human pattern-evoked • In vivo microscopy of the retina
retinal responses are altered by optic atrophy. Invest Ophthalmol Vis Sci 1982; • Noninvasive detection and quantification of cystoid macular
22:796. © Association for Research in Vision and Ophthalmology. edema
• Amplitude proportional to amount of bleached visual pigment
• Requires very bright light flashes
• Measures RPE integrity
• Used to distinguish Best disease from pseudovitelliform
macular degeneration
• Measures mass retinal response from the entire retina to
less than 5000 W. The electrodes may be held in place with flashes of light
colloidin. Responses must be averaged by computer because • Components derive from different cell types
amplitudes are generally less than 10 mV. • Insensitive to maculopathies
The use of on–off modulated gratings (in which a grating alter- • Allows detection of maculopathies
nates with a blank screen of the same mean luminance) has • Should employ visualization of the retina during testing to
been recommended as superior to pattern-reversal stimulation insure appropriate placement and stabilization of stimulus
for eliciting the steady-state VER.211 The on–off grating stimu- • Allows detection of regional losses of retinal function due to
lus has been reported to evoke a bell-shaped spatial-frequency rod system disease
tuned response more consistently across normal subjects than • Can be used to detect specific ganglion cell dysfunction when
the pattern-reversal stimulus. In addition, the spatial-tuning distal foveal retinal function is normal
characteristics obtained by psychophysical testing and by the • Can be used to detect specific cortical malfunction when
VER are more similar for the on–off grating stimulus than for ocular function is normal
the pattern-reversal stimulus (Fig. 124.37).
1627
RETINA AND VITREOUS

FIGURE 124.36. (Left) Pattern-reversal VERs


recorded from a midline occipital electrode
from the left and right eyes of a normal subject
(a) and two patients who were recovering from
acute attacks of optic neuritis in the right eye
with onset 4 weeks (b) and 3 weeks
(c) previously. Pattern reversal is at 20 ms;
positivity downward. (Right) Distribution of the
peak latencies of the major positive component
of the pattern-reversal VER from the affected
eye of 18 patients with optic neuritis (upper
histogram), from the unaffected eye of 19
patients with optic neuritis (middle histogram),
and from the right eye of 17 normal subjects
(lower histogram).
From Halliday AM, McDonald WI, Mushin J: Delayed
visual evoked response in optic neuritis. Lancet 1972;
1:982.
SECTION 10

FIGURE 124.37. Contrast-sensitivity function


(CSF) as obtained through the visual-evoked
potential (VEP) regression technique (filled
symbols) and psychophysically (open symbols).
(Left) Pattern reversal (Rev.). (Right) On–off
modulation.
From Strasburger H, Remky A, Murray IJ, et al:
Objective measurement of contrast sensitivity and
visual acuity with the steady-state visual evoked
potential. Germ J Ophthalmol 1996; 5:42. Reprinted
from Springer-Verlag.

FIGURE 124.38. P100 (P1) latency for 48 min


(closed circles) and 12 min (open circles)
checks as a function of age. Each data point is
the mean latency of the right and left eyes of
each subject. For 48-min checks, y = 0.14x +
101.8; for 12-min checks, y = 0.26x + 105.6.
For ease of comparison, the regression line for
48-min checks has also been plotted as a
dashed line on the 12-min graph.
From Sokol S, Moskowitz A, Towle VL: Age-related
changes in the latency of the visual evoked potential:
influence of check size. Electroencephalogr Clin
Neurophysiol 1981; 51:559.

1628
Objective Assessment of Retinal Function

REFERENCES
1. Carr RE, Ripps H, Siegel IM, et al: dystrophy-fundus flavimaculatus. Am J 40. Tamai A: Studies on the early receptor
Rhodopsin and the electrical activity of the Ophthalmol 2004; 138:55. potential in the human eye. III. ERP in
retina in congenital night blindness. Invest 22. Robson AG, Egan CA, Luong VA, et al: primary retinitis pigmentosa. Yonago Acta
Ophthalmol Vis Sci 1966; 5:497. Comparison of fundus autofluorescence Med 1974; 18:18.
2. Carr RE, Ripps H, Siegel IM: Visual with photopic and scotopic fine-matrix 41. Cone RA: The early receptor potential of
pigment kinetics and adaptation in fundus mapping in patients with retinitis the vertebrate eye. Cold Spring Harbor
albipunctatus. Doc Ophthalmol Proc Ser pigmentosa and normal visual acuity. Invest Symp Quant Biol 1965; 30:483.
1974; 4:193. Ophthalmol Vis Sci 2004; 45:4119. 42. Berson EL, Goldstein EB: The early
3. Carr RE, Ripps H: Rhodopsin kinetics and 23. Sholl HPN, Chong NHV, Robson AG, et al: receptor potential in dominantly inherited
rod adaptation in Oguchi’s disease. Invest Fundus autofluorescence in patients with retinitis pigmentosa. Arch Ophthalmol
Ophthalmol Vis Sci 1967; 6:426. Leber congenital amaurosis. Invest 1970; 83:412.
4. Bird AC, Hyman V: Detection of Ophthalmol Vis Sci 2004; 45:2747. 43. Barber C, Cotterill DJ, Larke JR: A new
heterozygotes in families with X-linked 24. Sandberg MA, Brockhurst RJ, Gaudio AR, contact lens electrode. Doc Ophthalmol
pigmentary retinopathy by measurements Berson EL: The association between visual Proc Ser 1977; 13:385.
of retinal rhodopsin concentration. Trans acuity and central retinal thickness in 44. Sieving PA, Fishman GA, Maggiano JM:
Ophthalmol Soc UK 1972; 93:221. retinitis pigmentosa. Invest Ophthalmol Vis Corneal wick electrode for recording
5. Bird AC: X-linked retinitis pigmentosa. Sci 2005; 46:3349. bright flash electroretinograms and early
Br J Ophthalmol 1975; 59:175. 25. Hirakawa H, Iijima H, Gohdo T, Tsukahara S: receptor potentials. Arch Ophthalmol 1978;

CHAPTER 124
6. Kemp CM, Jacobson SG, Faulkner DJ: Optical coherence tomography of cystoid 96:899.
Two types of visual dysfunction in macular edema associated with retinitis 45. Muller W, Topke H: The early receptor
autosomal dominant retinitis pigmentosa. pigmentosa. Am J Ophthalmol 1999; potential (ERP). Doc Ophthalmol 1987;
Invest Ophthalmol Vis Sci 1988; 29:1235. 128:185. 66:35.
7. Alpern M, Krantz D: Visual pigment kinetics 26. Apushkin MA, Fishman GA, Janowicz MJ: 46. Steinberg RH, Linsenmeier RA, Griff ER:
in abnormalities of the uvea-retinal Monitoring cystoid macular edema by Three light-evoked responses of the retinal
epithelium in man. Invest Ophthalmol Vis optical coherence tomography in patients pigment epithelium. Vision Res 1983;
Sci 1981; 20:183. with retinitis pigmentosa. Ophthalmology 23:1315.
8. Campbell FW, Rushton WAH: Measurement 2004; 111:1899. 47. Arden GB, Barrada A, Kelsey JH: New
of the scotopic pigment in the living human 27. Azzolini C, Pierro L, Codenotti M, Brancato clinical test of retinal function based upon
eye. J Physiol 1955; 130:131. R: OCT images and surgery of juvenile the standing potential of the eye. Br J
9. Weale RA: Photo-sensitive reactions in Macular retinoschisis. Eur J Ophthalmol Ophthalmol 1962; 46:449.
foveae of normal and cone monochromatic 1997; 7:196. 48. Krill AE, Morse PA, Potts AM, et al:
observers. Optica Acta 1959; 6:158. 28. Eriksson U, Larsson E, Holmstrom G: Hereditary vitelliruptive macular
10. Alpern M: Rhodopsin kinetics in the human Optical coherence tomography in the degeneration. Am J Ophthalmol 1966;
eye. J Physiol 1971; 217:447. diagnosis of juvenile X-linked retinoschisis. 61:1405.
11. Highman VN, Weale RA: Rhodopsin density Acta Ophthalmol Scand 2004; 82:218. 49. Francois J, De Rouck A, Fernandez-Sasso
and visual threshold in retinitis pigmentosa. 29. Apushkin MA, Fishman GA, Janowicz MJ: D: Electro-oculography in vitelliform
Am J Ophthalmol 1973; 75:822. Correlation of optical coherence degeneration of the macula. Arch
12. Sheorey UB: Clinical assessment of tomography findings with visual acuity and Ophthalmol 1967; 77:726.
rhodopsin in the eye. Br J Ophthalmol macular lesions in patients with X-linked 50. Deutman AF: Electro-oculography in
1976; 60:135. retinoschisis. Ophthalmology 2005; families with vitelliform dystrophy of the
13. Kemp CM, Faulkner DJ: Rhodopsin 112:495. fovea. Detection of the carrier state. Arch
measurement in human disease: fundus 30. Hayashi T, Kitahara K: Optical coherence Ophthalmol 1969; 81:305.
reflectometry using television. Dev tomography in enhanced S-cone 51. Sabates R, Pruett RC, Hirose T:
Ophthalmol 1981; 2:130. syndrome: large macular retinoschisis The electrooculogram in ‘vitelliform’
14. Kilbride PE, Fishman M, Fishman GA, et al: with disorganized retinal lamination. Eur J macular lesions. Doc Ophthalmol Proc Ser
Foveal cone pigment density difference and Ophthalmol 2005; 15:643. 1983; 37:93.
reflectance in retinitis pigmentosa. Arch 31. Berson EL, Goldstein EB: The early 52. Berson EL: Electrical phenomena in the
Ophthalmol 1986; 104:220. receptor potential in dominantly inherited retina. In: Hart WM, ed. Adler’s physiology
15. Ripps H, Brin KP, Weale RA: Rhodopsin retinitis pigmentosa. Arch Ophthalmol of the eye. Clinical application. 9th edn.
and visual threshold in retinitis pigmentosa. 1970; 83:412. St Louis: CV Mosby; 1992.
Invest Ophthalmol Vis Sci 1978; 17:735. 32. Brown KT, Crawford JM: Melanin and the 53. Kolder H, Brecher GA: Fast oscillations of
16. Faulkner DJ, Kemp CM: Human rhodopsin rapid light-evoked responses from pigment the corneoretinal potential in man. Arch
measurement using a T.V.-based imaging epithelial cells of the frog eye. Vision Res Ophthalmol 1966; 75:232.
fundus reflectometer. Vision Res 1984; 1967; 7:165. 54. Weleber RG: Fast and slow oscillations of
24:221. 33. Pak WL: Some properties of the early the electro-oculogram in Best’s macular
17. Kilbride PE, Hutman LP, Fishman M, et al: electrical response in the vertebrate retina. dystrophy and retinitis pigmentosa. Arch
Foveal cone pigment density difference in Cold Spring Harbor Symp Quant Biol 1965; Ophthalmol 1989; 107:530.
the aging human eye. Vision Res 1986; 30:493. 55. Fishman GA, Young RSL, Schall SP, et al:
26:321. 34. Cone RA: Early receptor potential: Electro-oculogram testing in fundus
18. Delori FC, Dorey CK, Staurenghi G, et al: photoreversible charge displacement in flavimaculatus. Arch Ophthalmol 1979;
In vivo fluorescence of the ocular fundus rhodopsin. Science 1967; 155:1128. 97:1896.
exhibits retinal pigment epithelium 35. Cone RA: Early receptor potential of 56. Kolder HE, Hochgesand P: Empirical model
lipofuscin characteristics. Invest the vertebrate retina. Nature 1964; of electro-oculogram. Doc Ophthalmol
Ophthalmol Vis Sci Mar 1995; 36:718. 204:736. 1973; 34:229.
19. Delori FC, Staurenghi G, Arend O, et al: 36. Cone RA, Brown PK: Dependence of the 57. Kelsey JH: Variations in the normal electro-
In vivo measurement of lipofuscin in early receptor potential on the orientation oculogram. Br J Ophthalmol 1967; 51:44.
Stargardt’s disease – Fundus of rhodopsin. Science 1967; 156:536. 58. Lith GHM, Balik J: Variability of the electro-
Flavimaculatus. Invest Ophthalmol Vis Sci 37. Goldstein EB, Berson EL: Cone dominance oculogram (EOG). Acta Ophthalmol 1970;
Mar 36:2327. of the human early receptor potential. 48:1091.
20. Von Ruckmann A, Fitzke FW, Bird AC: Nature 1969; 222:1272. 59. Jones RM, Stevens TS, Gould S: Normal
In vivo fundus autofluorescence in macular 38. Carr RE, Siegel IM: Action spectrum of the EOG values of young subjects. Doc
dystrophies. Arch Ophthalmol 1997; early receptor potential. Nature 1970; Ophthalmol Proc Ser 1977; 13:93.
115:609. 225:88. 60. Arden GB, Barrada A: Analysis of the
21. Lois N, Halfyard AS, Bird AC, et al: Fundus 39. Galloway NR: Early receptor potential in the electro-oculogram of a series of normal
autofluorescence in Stargardt macular human eye. Br J Ophthalmol 1967; 51:261. subjects. Br J Ophthalmol 1962; 46:468.
1629
RETINA AND VITREOUS

61. Adams A: The normal electro-oculogram guanosine monophosphate-type human 100. Miyake Y, Horiguchi M, Ota I, et al:
(EOG). Acta Ophthalmol 1973; 51:551. retinal degeneration. Comparison with Characteristic ERG flicker anomaly in
62. Jackson SA: The optimum illuminance level retinitis pigmentosa. Invest Ophthalmol Vis incomplete congenital stationary night
for clinical electro-oculography. Acta Sci 1990; 31:2283. blindness. Invest Ophthalmol Vis Sci 1987;
Ophthalmol 1979; 57:665. 81. Armington JC, Johnson EP, Riggs LA: The 28:1816.
63. Krogh E: Normal values in clinical scotopic a-wave in the electrical response 101. Gouras P, MacKay CJ: Growth in amplitude
electrooculography. IV. Analysis of two of the human retina. J Physiol (Lond) 1952; of the human cone electroretinogram with
dimensionless EOG parameters and their 118:289. light adaptation. Invest Ophthalmol Vis Sci
relation to other variables. Acta Ophthalmol 82. King-Smith PE, Loffing DA, Jones R: Rod 1989; 30:625.
1977; 55:739. and cone ERGs and their oscillatory 102. Miller S, Sandberg MA: Cone
64. Anderson ML, Purple RL: Circadian potentials. Invest Ophthalmol Vis Sci 1986; electroretinographic change during light
rhythms and variability of the clinical 27:270. adaptation in retinitis pigmentosa. Invest
electro-oculogram. Invest Ophthalmol Vis 83. Berson EL, Sandberg MA, Rosner B, et al: Ophthalmol Vis Sci 1991; 25:2536.
Sci 1980; 19:278. Natural course of retinitis pigmentosa over 103. Sandberg MA, Lee H, Gaudio AR, et al:
65. Brown KT, Wiesel TN: Localization of a three-year interval. Am J Ophthalmol Relationship of oscillatory potential
origins of electroretinogram components by 1985; 99:240. amplitudes to a-wave slope over a
intraretinal recording in the intact cat eye. 84. Reichel E, Bruce AM, Sandberg MA, et al: range of flash luminances in normal
J Physiol (Lond) 1961; 158:257. An electroretinographic and molecular subjects. Invest Ophthalmol Vis Sci 1991;
66. Brown KT, Watanabe K: Isolation and genetic study of X-linked cone degeneration. 32:1508.
identification of a receptor potential from Am J Ophthalmol 1989; 108:540. 104. Fuller DG, Knighton RW, Machemer R:
the pure cone fovea of the monkey retina. 85. Berson EL, Sandberg MA, Maguire A, et al: Bright-flash electroretinography for the
SECTION 10

Nature 1962; 193:958. Electroretinograms in carriers of blue cone evaluation of eyes with opaque vitreous.
67. Miller RF, Dowling JE: Intracellular monochromatism. Am J Ophthalmol 1986; Am J Ophthalmol 1975; 80:214.
responses of the Müller (glial) cells of 102:254. 105. Algvere P, Westbeck S: Human ERG in
mudpuppy retina: their relation to b-wave 86. Gouras P, MacKay CJ: Electroretinographic response to double flashes of light during
of the electroretinogram. J Neurophysiol responses of the short-wavelength- the course of dark adaptation: a Fourier
1970; 33:323. sensitive cones. Invest Ophthalmol Vis Sci analysis of the oscillatory potentials. Vision
68. Newman EA: Current source density 1990; 31:1203. Res 1972; 12:195.
analysis of the b-wave of the frog retina. 87. Arden GB, Carter RM, Hogg CR, et al: 106. Gur M, Zeevi Y: Frequency-domain analysis
Am J Neurophysiol 1980; 43:1335. A modified ERG technique and the results of the human electroretinogram. J Opt Soc
69. Ogden TE: The oscillatory waves of the obtained in X-linked retinitis pigmentosa. Am 1980; 70:53.
primate electroretinogram. Vision Res 1973; Br J Ophthalmol 1982; 67:419. 107. Van der Torren K, Groenweg G, Van Lith G:
13:1059. 88. Moloney JB, Mooney DJ, O’Connor MA: Measuring oscillatory potentials: Fourier
70. Heynen H, Wachtmeister L, van Norren D: Retinal function in Stargardt’s disease and analysis. Doc Ophthalmol 1988; 69:153.
Origin of the oscillatory potentials in the fundus flavimaculatus. Am J Ophthalmol 108. Andreasson SOL, Sandberg MA, Berson
primate retina. Vision Res 1985; 25:1365. 1983; 96:57. EL: Narrow-band filtering for monitoring
71. Alexander KR, Fishman GA, Barnes CS, 89. Massof RW, Wu L, Finkelstein D, et al: low-amplitude cone electroretinograms in
Grover S: ON-response deficit in the Properties of electroretinographic intensity retinitis pigmentosa. Am J Ophthalmol
electroretinogram of the cone system in response functions in retinitis pigmentosa. 1988; 105:500.
X-linked retinoschisis. Invest Ophthalmol Doc Ophthalmol 1984; 57:279. 109. Birch DG, Sandberg MA: Sub-microvolt
Vis Sci 2001; 42:453. 90. Birch DG, Fish GE: Rod ERGs in retinitis full-field cone electroretinograms: artifacts
72. Dryja TP, McGee TL, Berson EL, et al: pigmentosa and cone-rod degeneration. and reproducibility. Doc Ophthalmol 1997;
Night blindness and abnormal cone Invest Ophthalmol Vis Sci 1987; 28:140. 92:269.
electroretinogram ON responses in patients 91. Birch DG, Hood DC, Locke KG, et al: 110. Martin DA, Heckenlively JR: The normal
with mutations in the GRM6 gene encoding Quantitative measures of electroretinogram. Doc Ophthalmol Proc
mGluR6. PNAS 2005; 102:4884. phototransduction in cone and rod Ser 1982; 31:135.
73. Seiple W, Holopigian K: The ‘off’ response photoreceptors. Normal aging, progression 111. Berson EL: Electroretinographic findings in
of the human electroretinogram does not with disease, and test-retest variability. retinitis pigmentosa. Jpn J Ophthalmol
contribute to the brief flash ‘b-wave’. Visual Arch Ophthalmol 2002; 120:1045. 1987; 31:327.
Neurosci 1994; 11:667. 92. Tzekov RT, Locke KG, Hood DC, Birch DG: 112. Peterson H: The normal b-potential in the
74. Hirose T, Wolf E, Hara A: Cone and rod ERG phototransduction single-flash clinical electroretinogram:
Electrophysiological and psychophysical parameters in retinitis pigmentosa. Invest a computer technique study of the
studies in congenital retinoschisis of Ophthalmol Vis Sci 2003; 44:3993. influence of sex and age. Acta Ophthalmol
X-linked recessive inheritance. Doc 93. Berson EL, Gouras PL, Gunkel R: Rod 1968; 99(Suppl):5.
Ophthalmol Proc Ser 1977; 13:173. responses in retinitis pigmentosa, 113. Weleber RG: The effect of age on human
75. Yonemura D, Aoki T, Tsuzuki K: dominantly inherited. Arch Ophthalmol cone and rod Ganzfeld electroretinograms.
Electroretinogram in diabetic retinopathy. 1968; 80:58. Invest Ophthalmol Vis Sci 1981; 20:392.
Arch Ophthalmol 1962; 68:19. 94. Le Grand Y: Form and Space Vision. 114. Wright CE, Williams DE, Drasdo N, et al:
76. Gaudio AR, Lee H, Kini M, et al: Oscillatory Bloomington: Indiana University Press; 1967. The influence of age on the
potentials in central retinal vein occlusion. 95. Rabin AR, Berson EL: A full-field system for electroretinogram and visual evoked
Invest Ophthalmol Vis Sci 1989; clinical electroretinography. Arch potential. Doc Ophthalmol 1985; 59:365.
30(Suppl):477. Ophthalmol 1974; 92:59. 115. Gouras P, MacKay CJ, Yamamoto S: The
77. Gouras P: Rod and cone independence in 96. Karpe G, Wulfing B: Importance of pupil human S-cone electroretinogram and its
the electroretinogram of the dark-adapted size in clinical ERG. Acta Ophthalmol 1961; variation among subjects with and without
monkey’s perifovea. J Physiol 1966; 70(Suppl):53. L- and M-cone function. Invest Ophthalmol
187:455. 97. Brunette JR, Olivier P, Lafond G: Decreased Vis Sci 1993; 34:2437.
78. Berson EL, Gouras P, Hoff M: Temporal ocular light penetration and 116. Said FS, Weale RA: The variation with age
aspects of the electroretinogram. Arch electroretinographic response time. Can J of the spectral transmissivity of the living
Ophthalmol 1969; 81:207. Ophthalmol 1980; 15:24. human crystalline lens. Gerontologia 1959;
79. Chatrian GE, Nelson PL, Lettich E, et al: 98. Sandberg MA: Technical issues in 3:213.
Computer-assisted quantitative electroretinography. In: Heckenlively J, 117. Dodt E, Copenhaver RM, Gunkel RD:
electroretinography. II. Separation of rod Arden G, eds. Principles and practice of Electroretinographic measurement of the
and cone components of the clinical electrophysiology of vision. spectral sensitivity in albinos, caucasians,
electroretinogram in congenital St Louis: Mosby-Year Book; 1991. and negroes. Arch Ophthalmol 1959;
achromatopsia and congenital nyctalopia. 99. Hennessy MP, Vaegan: Amplitude scaling 62:795.
Am J EEG Technol 1980; 20:79. of Burian–Allen, gold foil and Dawson, Trick 118. Krill AE, Lee GB: The electroretinogram in
80. Sandberg MA, Miller S, Berson EL: Rod and Litzkow electrodes. Doc Ophthalmol albinos and carriers of the ocular albino
1630 electroretinograms in an elevated cyclic 1995; 89:235. trait. Arch Ophthalmol 1963; 69:32.
Objective Assessment of Retinal Function

119. Wali N, Leguire LE: Fundus pigmentation 138. Miyake Y, Ichikawa K, Shiose Y, et al: 156. Vajaranant TS, Seiple W, Szlyk JP,
and the dark-adapted electroretinogram. Hereditary macular dystrophy without Fishman GA: Detection using the multifocal
Doc Ophthalmol 1992; 80:1. visible fundus abnormality. Am J electroretinogram of mosaic retinal
120. Pallin E: The influence of the axial size of the Ophthalmol 1989; 108:292. dysfunction in carriers of X-linked retinitis
eye on the size of the recorded b-potential in 139. Matthews GP, Sandberg MA, Berson EL: pigmentosa. Ophthalmology 2002;
the clinical single-flash electroretinogram. Foveal cone electroretinograms in patients 109:560.
Acta Ophthalmol 1969; 101(Suppl):1. with central visual loss of unexplained 157. Piao C-H, Kondo M, Nakamura M, et al:
121. Perlman I, Meyer E, Haim T, et al: Retinal etiology. Arch Ophthalmol 1992; 110:1568. Multifocal electroretinograms in X-linked
function in high refractive error assessed 140. Jacobson SG, Sandberg MA, Effron MH, retinoschisis. Invest Ophthalmol Vis Sci
electroretinographically. Br J Ophthalmol et al: Foveal cone electroretinograms in 2003; 44:4920.
1984; 68:79. strabismic amblyopia. Comparison with 158. Huang S, Wu D, Jiang F, et al: The
122. Karpe G, Rickenbach K, Thomasson S: juvenile macular degeneration, macular multifocal electroretinogram in X-linked
The clinical electroretinogram. I. The scars and optic atrophy. Trans Ophthalmol juvenile retinoschisis. Doc Ophthalmol
normal electroretinogram above fifty years Soc UK 1979; 99:353. 2003; 106:251.
of age. Acta Ophthalmol 1950; 28:301. 141. Biersdorf WR: The foveal electroretinogram 159. Nusinowitz S, Hood DC, Birch DG: Rod
123. Zeidler I: The clinical electroretinogram. IX. is normal in optic atrophy. Doc Ophthalmol transduction parameters from the a wave
The normal electroretinogram. Value of the Proc Ser 1984; 40:127. of local receptor populations. J Opt Soc
b-potential in different age groups and its 142. Sandberg MA, Baruzzi CM, Berson EL: Am 1995; 12:2259.
difference in men and women. Acta Foveal cone ERGs in optic neuropathy. 160. Sandberg MA, Pawlyk BS, Berson EL:
Ophthalmol 1959; 37:294. Invest Ophthalmol Vis Sci 1986; Isolation of focal rod electroretinograms

CHAPTER 124
124. Birch DG, Berson EL, Sandberg MA: 27(Suppl):104. from the dark-adapted human eye. Invest
Diurnal rhythm in the human rod ERG. 143. Weiner A, Sandberg MA: Normal change in Ophthalmol Vis Sci 1996; 37:930.
Invest Ophthalmol Vis Sci 1984; 25:236. the foveal cone ERG with increasing 161. Riggs LA, Johnson EP, Schick AML:
125. Sandberg MA, Pawlyk BS, Berson EL: duration of light exposure. Invest Electrical responses of the human eye to
Electroretinogram (ERG) sensitivity and Ophthalmol Vis Sci 1991; 32:2842. moving stimulus pattern. Science 1964;
phagosome frequency in the normal 144. Weiner A, Sandberg MA, Berson EL: 144:567.
pigmented rat. Exp Eye Res 1986; 43:781. Abnormal foveal to parafoveal cone ERG 162. Odom JV, Maida TM, Dawson WW: Pattern
126. Arden GB, Bankes JLK: Foveal amplitude ratios in patients with reduced evoked retinal response (PERR) in human:
electroretinogram as a clinical test. visual acuity and suspected foveal disease. effects of spatial frequency, temporal
Br J Ophthalmol 1966; 50:740. Invest Ophthalmol Vis Sci 1990; frequency, luminance and defocus. Curr
127. Biersdorf WR, Diller DA: Local 31(Suppl):426. Eye Res 1982–83; 2:99.
electroretinogram in macular degeneration. 145. Miyake Y, Shiroyama N, Horiguchi M, et al: 163. Sokol S, Jones K, Nadler D: Comparison of
Am J Ophthalmol 1969; 68:296. Asymmetry of focal ERG in human macular the spatial response properties of the
128. Hirose T, Miyake Y, Hara A: Simultaneous region. Invest Ophthalmol Vis Sci 1989; human retina and cortex as measured by
recording of electroretinogram and visually 30:1743. simultaneously recorded pattern ERGs and
evoked response: focal stimulation under 146. Sandberg MA, Miller S, Gaudio AR: Foveal VERs. Vision Res 1983; 23:723.
direct observation. Arch Ophthalmol 1977; cone ERGs in fellow eyes of patients with 164. Hess RF, Baker CL Jr: Human pattern-
95:1205. unilateral neovascular age-related macular evoked electroretinogram. J Neurophysiol
129. Sandberg MA, Jacobson SG, Berson EL: degeneration. Invest Ophthalmol Vis Sci 1984; 51:939.
Foveal cone electroretinograms in retinitis 1993; 34:3477. 165. Hess RF, Baker CL Jr, Verhoeve JN, et al:
pigmentosa and juvenile macular 147. Bearse MA, Sutter EE: Imaging localized The pattern evoked electroretinogram:
degeneration. Am J Ophthalmol 1979; retinal dysfunction with the multifocal its variability in normals and its relationship
88:702. electroretinogram. J Opt Soc Am 1996; to amblyopia. Invest Ophthalmol Vis Sci
130. Fish GE, Birch DG, Fuller DG, et al: 13:634. 1985; 26:1610.
A comparison of visual function tests in 148. Hood DC, Seiple W, Holopigian K, 166. Plant GT, Hess RF, Thomas SJ: The pattern
eyes with maculopathy. Ophthalmology Greenstein V: A comparison of the evoked electroretinogram in optic neuritis.
1986; 93:1177. components of the multifocal and full-field Brain 1986; 109:469.
131. Seiple WH, Siegel IM, Carr RE, et al: ERGs. Vis Neurosci 1997; 14:1. 167. Drasdo N, Thompson DA, Thompson CM,
Evaluating macular function using the focal 149. Kretschmann U, Seeliger MW, Ruether K, et al: Complementary components and local
electroretinogram. Invest Ophthalmol Vis et al: Multifocal electroretinography in variations of the pattern electroretinogram.
Sci 1986; 27: 1123. patients with Stargardt’s macular Invest Ophthalmol Vis Sci 1987; 28:158.
132. Miyake Y, Shiroyama N, Ota I, et al: Local dystrophy. Br J Ophthalmol 1998; 82:267. 168. Trick GL, Wintermeyer DH: Spatial and
macular electroretinographic responses in 150. Piao CH, Kondo M, Tanikawa A, et al: temporal frequency tuning of pattern-
idiopathic central serous chorioretinopathy. Multifocal electroretinogram in occult reversal retinal potentials. Invest
Am J Ophthalmol 1988; 106:546. macular dystrophy. Invest Ophthalmol Vis Ophthalmol Vis Sci 1982; 23:774.
133. Brodie SE, Naidu EM: Combined amplitude Sci 2000; 41:513. 169. Spekreijse H, Estevez O, Van der Tweel LH:
and phase criteria for evaluation of macular 151. Wildberger H, Niemeyer G, Junghardt A: Luminance responses to pattern reversal.
electroretinograms. Invest Ophthalmol Vis Multifocal electroretinogram (mfERG) in a Doc Ophthalmol Proc Ser 1973; 2:205.
Sci 1990; 31(Suppl):425. family with occult macular dystrophy 170. Baker CL, Hess RF: Linear and nonlinear
134. Sandberg MA, Hanson AH, Berson EL: (OMD). Klin Monatsbl Augenheilkd 2003; components of human electroretinogram.
Foveal and parafoveal cone 220:111. J Neurophysiol 1973; 51:9521984.
electroretinograms in juvenile macular 152. Seeliger M, Kretschmann U, Apfelstedt- 171. Arden GB, Vaegan, Hogg CR: Clinical and
degeneration. Ophthalmic Paediatr Genet Sylla E, et al: Multifocal electroretinography experimental evidence that the pattern
1983; 3:83. in retinitis pigmentosa. Am J Ophthalmol electroretinogram (PERG) is generated in
135. Birch DG, Fish GE: Focal cone 1998; 125:214. more proximal retinal layers than the focal
electroretinograms: aging and macular 153. Holopigian K, Seiple W, Greenstein VC, et electroretinogram (FERG). Ann N Y Acad
disease. Doc Ophthalmol 1988; 69:211. al: Local cone and rod system function in Sci 1982; 388:580.
136. Remulla JFC, Gaudio AR, Miller S, patients with retinitis pigmentosa. Invest 172. May JG, Ralston JV, Reed JL, et al: Loss in
Sandberg MA: Foveal electroretinograms Ophthalmol Vis Sci 2001; 42:779. pattern-elicited electroretinograms in optic
and choroidal perfusion characteristics in 154. Berson EL, Rosen JB, Simonoff EA: nerve dysfunction. Am J Ophthalmol 1982;
fellow eyes of patients with unilateral Electroretinographic testing as an aid in 93:418.
neovascular age-related macular detection of carriers of X-chromosome- 173. Wanger P, Persson HE: Pattern-reversal
degeneration. Br J Ophthalmol 1993; linked retinitis pigmentosa. Am J electroretinograms in unilateral glaucoma.
79:558. Ophthalmol 1979; 87:460. Invest Ophthalmol Vis Sci 1983; 24:749.
137. Birch DG, Jost BF, Fish GE: The focal 155. Fishman GA, Weinberg AB, McMahon TT: 174. Boback P, Bodis-Wollner I, Harnois C, et al:
electroretinogram in fellow eyes of patients X-linked recessive retinitis pigmentosa: Pattern electroretinograms and visual
with idiopathic macular holes. Arch clinical characteristics of carriers. Arch evoked potentials in glaucoma and multiple
Ophthalmol 1988; 106:1558. Ophthalmol 1986; 104:1329. sclerosis. Am J Ophthalmol 1983; 96:72. 1631
RETINA AND VITREOUS

175. Kirkham TH, Coupland SG: The pattern 189. Maffei L, Fiorentini A, Bisti S, et al: Pattern 204. Halliday AM, McDonald WI, Mushin J:
electroretinogram in optic nerve ERG in the monkey after section of the Delayed visual evoked response in optic
demyelination. Can J Neurol Sci 1983; optic nerve. Exp Brain Res 1985; 59:423. neuritis. Lancet 1972; 1:982.
10:256. 190. Sieving PA, Steinberg RH: Proximal retinal 205. Halliday AM, McDonald WI, Mushin J:
176. Mashima Y, Oguchi Y: Clinical study of the contribution to the intraretinal 8-Hz pattern Visual evoked responses in the diagnosis
pattern electroretinogram in patients with ERG of cat. J Neurophysiol 1987; 57:104. of multiple sclerosis. BMJ 1973; 4:661.
optic nerve damage. Doc Ophthalmol 1985; 191. Vaegan, Billson FA: Macular 206. Halliday AM, McDonald WI, Mushin J:
61:91. electroretinograms and contrast sensitivity Delayed pattern evoked responses in optic
177. Fiorentini A, Maffei L, Pirchio M, et al: The as sensitive detectors of early neuritis in relation to visual acuity. Trans
ERG in response to alternating gratings in maculopathy. Doc Ophthalmol 1986; Ophthalmol Soc UK 1973; 93:315.
patients with diseases of the peripheral 63:399. 207. Howe JW, Mitchell KW, Robson C: Some
visual pathway. Invest Ophthalmol Vis Sci 192. Kakisu Y, Mizota A, Adachi E: Clinical clinical experiences using contrast evoked
1981; 21:490. application of the pattern electroretinogram potential techniques in organic and non-
178. Dawson WW, Maida TM, Rubin ML: Human with lid skin electrode. Doc Ophthalmol organic visual dysfunction. Doc Ophthalmol
pattern-evoked retinal responses are 1986; 63:187. Proc Ser 1982; 31:353.
altered by optic atrophy. Invest Ophthalmol 193. Arden GB, Carter RM, Hogg C, et al: 208. Harter MR, White CT: Evoked cortical
Vis Sci 1982; 22:796. A gold foil electrode: extending the responses to checkerboard patterns: effect
179. Arden GB, Hamilton AMP, Wilson-Holt J, horizons for clinical electroretinography. of check size as a function of visual acuity.
et al: Pattern electroretinograms become Invest Ophthalmol Vis Sci 1979; 18:421. Electroencephalogr Clin Neurophysiol
abnormal when background diabetic 194. Dawson WW, Trick GL, Litzkow CA: 1970; 28:48.
retinopathy deteriorates to a preproliferative Improved electrode for electroretinography. 209. Millodot M, Riggs LA: Refraction
SECTION 10

stage: possible use as a screening test. Invest Ophthalmol Vis Sci 1979; 18:988. determined electrophysiologically:
Br J Ophthalmol 1986; 70:330. 195. Celesia GG, Kaufman D, Cone S: Effects of responses to alternation of visual contours.
180. Coupland SG: A comparison of oscillatory age and sex on pattern electroretinograms Arch Ophthalmol 1970; 84:272.
potential and pattern electroretinogram and visual evoked potentials. 210. Sokol S, Moskowitz A: Effect of retinal blur
measures in diabetic retinopathy. Doc Electroencephalogr Clin Neurophysiol on the peak latency of the pattern evoked
Ophthalmol 1987; 66:207. 1987; 68:161. potential. Vision Res 1981; 21:1279.
181. Weinstein GW, Arden GB, Hitchings RA, 196. Odom JV, Maida TM, Dawson WW, et al: 211. Strasburger H, Remky A, Murray IJ, et al:
et al: The pattern electroretinogram (PERG) Pattern electroretinogram: effects of Objective measurement of contrast
in ocular hypertension and glaucoma. Arch reference electrode position. Doc sensitivity and visual acuity with the
Ophthalmol 1988; 106:923. Ophthalmol 1987; 65:297. steady-state visual evoked potential. Germ
182. Trick GL, Bickler-Bluth M, Cooper DG, 197. Seiple WH, Siegel IM: Recording the J Ophthalmol 1996; 5:42.
et al: Pattern reversal electroretinogram pattern electroretinogram: a cautionary 212. Airas KA: Interindividual variation and
(PERG) abnormalities in ocular note. Invest Ophthalmol Vis Sci 1983; additivity of the visual evoked potentials to
hypertension: correlation with glaucoma 24:796. the local checkerboard stimulation of the
risk factors. Curr Eye Res 1988; 7:201. 198. Peachey NS, Sokol S, Moskowitz A: central and paracentral retina. Acta
183. Wanger P, Persson HE: Pattern-reversal Recording the contralateral PERG: effect of Ophthalmol 1986; 64:557.
electroretinograms from normotensive, different electrodes. Invest Ophthalmol Vis 213. Meienberg O, Kutak L, Smolenski C, et al:
hypertensive and glaucomatous eyes. Sci 1983; 24:1514. Pattern reversal evoked cortical responses
Ophthalmologica 1987; 195:205. 199. Arden GB, Carter RM, Hogg C: Uniocular in normals. A study of different methods of
184. Sokol S, Nadler D: Simultaneous recording of pattern ERG. Vision Res 1986; stimulation and potential reproducibility.
electroretinograms and visual evoked 26:281. J Neurol 1979; 222:81.
potentials from adult amblyopes in 200. Rover J, Bach M: Pattern electroretinogram 214. Sokol S, Moskowitz A, Towle VL:
response to pattern stimuli. Invest plus visual evoked potential: a decisive test Age-related changes in the latency of the
Ophthalmol Vis Sci 1979; 18:848. in patients suspected of malingering. Doc visual evoked potential: influence of check
185. Arden GB, Vaegan, Hogg CG, et al: Pattern Ophthalmol 1987; 66:245. size. Electroencephalogr Clin Neurophysiol
ERGs are abnormal in many amblyopes. 201. Jacobi PC, Walter P, Brunner R, Krieglstein 1981; 51:559.
Trans Ophthalmol Soc UK 1980; GK: Reproducibility and intraindividual 215. Allen D, Tyler CW, Norcia AM: Development
100:435–460. variability of the pattern electroretinogram. of grating acuity and contrast sensitivity in
186. Arden GB, Wooding SL: Pattern ERGs in Germ J Ophthalmol 1994; 3:216. the central and peripheral visual field of the
amblyopia. Invest Ophthalmol Vis Sci 1985; 202. Trick GL, Trick LR, Haywood KM: Altered human infant. Vision Res 1996; 36:1945.
26:88. pattern evoked retinal and cortical 216. Hawkes CH, Stow B: Pupil size and the
187. Gottlob I, Welge-Lussen L: Normal pattern potentials associated with human pattern evoked visual response. J Neurol
electroretinograms in amblyopia. Invest senescence. Curr Eye Res 1986; 10:717. Neurosurg Psychiatry 1981; 44:90.
Ophthalmol Vis Sci 1987; 28:187. 203. Porciatti V, Burr DC, Morrone MC, Fiorentini
188. Maffei L, Fiorentini A: Electroretinographic A: The effects of ageing on the pattern
responses to alternating gratings before electroretinogram and visual evoked
and after section of the optic nerve. potential in humans. Vision Res 1992;
Science 1981; 211:953. 32:1199.

1632
CHAPTER

125 Müller Cells and the Retinal Pigment Epithelium


Vamsi K. Gullapalli, Ilene K. Sugino, and Marco A. Zarbin

MÜLLER CELLS diencephalon leads to the formation of the optic vesicle that
abuts against the surface ectoderm. The cornea and lens develop
from the surface ectoderm. The optic vesicle then invaginates
INTRODUCTION on day-27 to form a bilayered optic cup. The inner layer com-
Müller cells, first described by Heinrich Müller in 1851,1 often prises progenitor cells that give rise to the cells of the neural
are thought of as providing a structural framework for the retina; the outer layer develops into retinal pigment epithelium
retina. They are large cells spanning the entire width of the (RPE). As in other parts of nervous system, the neural retina
retina, from the internal limiting membrane (ILM) to processes initially consists of a neural epithelium and inner marginal
extending into the subretinal space. Müller cells belong to the zone. The neural epithelium develops into an inner and outer
class of glial cells called astrocytes, which are characterized by a neuroblastic layer and a transient layer of Chievitz. By the
large nucleus, prominent cellular processes, and participation in fourth month, all the layers of retina are discernible.3
neural homeostasis. However, Müller cells also exhibit func- Cell division generating the cells of the neural retina occurs
tions of another type of glia, oligodendrocytes, which supply in two phases. In phase one, ganglion, cone, and horizontal cells
energy to neurons. Müller cells are truly retinal cells, as they develop. In phase two, rod, bipolar, and Müller cells develop.
originate in the retina, are found exclusively in the retina, and Amacrine cells develop during both phases.4 Animal studies
are the major glial cells of the retina. suggest that a single progenitor cell gives rise to a column of
cells containing one Müller cell ensheathing a defined number
of rod, bipolar, and amacrine cells.5
OVERVIEW OF THE MÜLLER CELL AND ITS Immunohistochemical studies of fetal human tissue show
FUNCTIONS that the progenitor cell itself may differentiate into a Müller
Based on the intricate and extensive processes emanating from cell.6 Müller cells are thought to be a key component in this
a single cell body, it is easy to imagine the Müller cell having the columnar arrangement of cells, providing nutrients, guiding
function of support, as Müller cells fill all extracellular spaces in associated neurons in migration to their final positions, and
the retina. Besides providing support for retinal neurons, Müller acting as a substrate for neurite outgrowth.5,7 In cell culture
cells define outer and inner boundaries of the retina. Their experiments utilizing stratospheroids of retinal cells of two dif-
endfeet terminate at the ILM, and their junctional complexes ferent species (chick and quail), Willbold and coworkers demon-
form the outer limiting membrane (OLM). The elaborate struc- strated that spheroids composed of species-specific radial
ture of the Müller cell involves contact with all parts of the sectors or columns form, surrounded by a Müller cell forming a
retina, which enables Müller cells to provide functions other barrier to cross species cell lateral migration.8 Selective damage
than support to the various cells they contact. During retinal to Müller cells showed their continued presence was necessary
development, Müller cells show extreme plasticity; they localize to maintain the columnar arrangement.8 Cell recognition markers
to areas of the retina undergoing development and differen- such as L1/NgCAM, 5A11, and F11 are present in Müller cells,
tiation and show localized development of processes in parallel which supports their role in migration and formation of radial
with development of the adjacent neuroblast. In the mature columns. In addition to these migration cues, Müller cells also
retina, Müller cells provide support for adjacent cells, main- express barrier proteins providing negative cues that may con-
taining control of the extracellular environment and providing stitute a repulsive barrier (e.g., EAP-300; embryonic avian poly-
nutrients to maintain the high metabolic rate of the retina. peptide and clustrin).4,8 Of note, these experiments showed that
Additionally, Müller cells are very important in the visual cycle while cross species cell migration between radial columns is
of cones, providing the substrate for regeneration of cone opsin. inhibited, lateral processes from Müller cells and their associ-
Regional specializations to support these functions are present ated neurons can cross the species-specific border, suggesting
within each Müller cell. their role in neurite outgrowth. Studies of neonatal rat retinal
cells show that Müller cells can support long neurite extensions
in rods while other retinal neurons are less supportive.7
EMBRYOLOGY AND DEVELOPMENT OF Morphological studies of the human fetus have demonstrated
MÜLLER CELLS the anatomic distribution of Müller cells during develop-
The earliest morphologically distinct step in the development of ment.9,10 Müller cells are present at gestation week-7 (20 mm)
the eye is the bilateral evagination of the diencephalon region and are present in both neuroblastic layers although they are
of the neural tube on day-25 postconception.2 However, it is more common in the inner half of the inner neuroblastic layer.
likely that the cells in this region are already committed to Dense processes extend from the Müller cells to the surface of
the eye long before this event. Continued evagination of the the nerve fiber layer (NFL), and radial processes are found 1633
RETINA AND VITREOUS

between ganglion cell axons. At gestation week-9 (40–43 mm), INTERNAL


Müller cells are the major component of the inner neuroblastic LIMITING MEM.
layer, forming columns and extending across the full thickness
of the retina. Dense inner processes are present near ganglion c NERVE
FIBER L.
cells and are thought to guide the ganglion cells into the inner
retina. At 66 mm, Müller cell nuclei and outer processes are
observed between differentiating cones. By gestation week-13
GANGLION
(96 mm), Müller cells are present in all layers of the retina. At b CELL L.
159–200 mm, when the rods and cones are present, Müller cell
nuclei are still present in the outer nuclear layer (ONL), are
numerous in the inner nuclear layer (INL), and are still present
in the ganglion cell layer and NFL. At week-20 (180 mm), a INTERNAL
c
PLEXIFORM L.
Müller cell processes extend microvilli into the subretinal space.
Observations from these studies show that Müller cell pro-
cesses differentiate sequentially from the inner (vitread) to the
outer (sclerad) retina, differentiating in synchrony with the cell
on which the process abuts. Different parts of the same cell can
differentiate at different times while other processes can differ-
SECTION 10

INTERNAL
entiate at the same time but in morphologically different ways. b
NUCLEAR L.
It is not clear whether the differentiation of different portions of
the Müller cell is in response to the demands of adjacent cells
or if Müller cell process differentiation leads to an induction of
neuronal differentiation.10 The apparent shifts in location of
Müller cell nuclei are believed to be due to migration of the
c
nucleus along the cell length, cell loss, and, possibly, cell
division.9 EXTERNAL
PLEXIFORM L.
a
ANATOMY
Generally, Müller cells are recognized histologically and ultra-
structurally by their relatively dense cytoplasm, which contains a – RADIAL PROCESSES
numerous glycogen particles, abundant filaments measuring b – HONEYCOMB
100Å in diameter, and well-developed smooth endoplasmic MESHWORK
EXTERNAL
reticulum consisting of vesicles of varying sizes and shapes.11 In b c – HORIZONTAL FIBERS NUCLEAR L.
the adult eye, Müller cells span the entire width of the retina d – FIBER BASKETS
with their cell bodies located in the INL and nuclei located in
the middle of this layer. Radial processes (Fig. 125.1a) extend EXTERNAL
vitread to terminate near the ILM and sclerad to contribute to LIMITING MEM.
d
the OLM, and lateral processes extend out at all levels of the
neural retina. In the nuclear layers, lamellae form a network
surrounding cell bodies (Fig. 125.1b, ‘honeycomb meshwork’).
In the plexiform layers, processes are interwoven between FIGURE 125.1. Schematic drawing illustrating the complex structure
synaptic processes of neurons, ensheath dendrites, and of Müller cells. Müller cells (shown with dark cytoplasm and dark
surround axons in the NFL (Fig. 125.1c, ‘horizontal fibers’). nuclei) extend radial processes (a) from the cell body to form the
Müller cells cover most, but not all, of the neuronal surfaces. internal limiting membrane of the retina surface and the external
Small branches extend out to contact and surround blood limiting membrane facing the retinal pigment epithelium (RPE). Lateral
processes (b, honey comb meshwork) extend from the radial
vessels along with astrocytes, forming alternate layers of astro-
processes to ensheath cell bodies of the ganglion cells, cells of the
cyte and Müller cell processes, completely isolating vessels from internal (inner) nuclear layer, and cell bodies of the photoreceptors in
the rest of the neural retina.12 Radial trunk extensions thin in the external (outer) nuclear layer. Horizontal fibers (c) are found in the
the inner plexiform layer (IPL), pass through the ganglion cell plexiform layers and the nerve fiber layer where they ensheath
layer, and intertwine with astrocytes and microglia forming an neurons and their processes.
endfoot expansion terminating at the ILM. Viewed en face, the From Hogan MJ, Alvarado JA, Weddall JE: Histology of the human eye.
endfeet form a mosaic pattern. Particle arrays, referred to as Philadelphia: W.B. Saunders; 1971, Figure 9–57.
intramembranous orthogonal arrays of particles (OAPs), are
prominent in the endfeet near the vitreous surface and in areas
where processes contact blood vessels. The function of OAPs is In the fovea centralis, Müller cell processes occupy most of
not certain, although based on their location, they have been the inner third of the retinal thickness. The processes form an
proposed to function as sites of K+ conductance.13 inverted cone-shaped zone whose apex faces the ONL (Fig. 125.2).
At their outermost ends, Müller cells exhibit junctions (zonula In the 50 mm diameter apex of the Müller cell cone, Müller cell
adherens) between other Müller cells and photoreceptors to processes are present with atypical watery cytoplasm separating
form the OLM, which is thought to have a supportive function the foveal cone cells.15 The Müller cell cone is hypothesized to
in maintaining the alignment and orientation of the photo- be a reservoir for retinal xanthophylls in addition to providing
receptors.14 Extending past the OLM into the subretinal space structural support for the receptor cells of the fovea.16 The
are long, thin apical Müller cell processes filling the spaces Müller cell cone is thought to be involved in idiopathic age-
between photoreceptor inner segments (Figure 125.1d, ‘fiber related macular holes.16 The role of Müller cells in creating
baskets’).11 structural support in the retina is illustrated by the condition,
1634
Müller Cells and the Retinal Pigment Epithelium

b
Line M
Line N
b
a Line P
a

a
FIGURE 125.3. B-scans of 3-D OCT from a patient with juvenile
X-linked retinoschisis. Two wide hyporeflective spaces split the retina.
Anteroposterior or oblique linear columns form a bridge across a
superficial wide hyporeflective space. In the same layer, there is a
large cystoid space in the fovea (line N). This layer is probably the
outer plexiform layer. Deeper cleavage is seen in the parafoveal area
but not in the fovea (line P). This layer is probably the outer nuclear
layer. Small cystoid spaces (arrowhead) are seen in the superficial
FIGURE 125.2. Details of the fovea centralis. The nuclei of the parafoveal retina that split the retina (line M). This layer is probably the

CHAPTER 125
photoreceptors are displaced internally (a) and the curving internal nerve fiber layer or the ganglion cell layer.
receptor fibers are seen at (b). The lighter cytoplasm of the Müller Reproduced with permission from Minami Y, Ishiko S, Takai Y, et al: Retinal
cells (arrow) is seen in the center of the fovea. changes in juvenile X linked retinoschisis using three dimensional optical
Legend and figure from Hogan MJ, Alvarado JA, Weddell JE: Histology of the coherence tomography. Br J Ophthalmol 2005; 89:1663–1664 (BMJ Publishing
human eye. Philadelphia: W.B. Saunders; 1971, Figure 9–80. Group Ltd).

juvenile X-linked retinoschisis (JXRS). JXRS is caused by [K+]0 to low concentration is thought to be achieved by
mutations in RS1, which encodes the protein, retinoschisin. nonuniform distribution of potassium channels.22 Several types
Retinoschisin is secreted by photoreceptor and bipolar cells.17–19 of potassium channels have been identified in Müller cells, and
JXRS is a congenital disease characterized by foveal schisis in it appears that the type of channel may vary considerably
100% of patients and peripheral retinoschisis in ~50% of among different species.14 An important K+ channel thought to
patients. The electroretinogram demonstrates reduced b-wave contribute significantly to the Müller cell K+ buffering system is
amplitude and relative a-wave preservation. Visual acuity usually the Kir channel, found in the plasma membrane. Studies per-
is in the range of 20/40–20/60. Three dimensional optical formed in mice identified two Kir channels with very different
coherence tomography demonstrates a large cystic space in the subcellular concentrations.23 Kir2.1, a strongly inward rectify-
fovea (probably in the outer plexiform layer (OPL)) as well as ing channel, was identified in regions of high K+ flow into
cleavage planes in the NFL, GCL, and OPL (Fig. 125.3).20 Müller cells (i.e., membrane domains localized near retinal
Bridging columns spanning these cystic spaces probably neurons, including the synaptic layers). These channels are
represent Müller cells and demonstrate the mechanical effect of such that K+ enters more readily than it can exit; thus, these
these cells on retinal architecture. channels are described as inwardly rectifying potassium
channels. A weakly rectifying channel (i.e., movement of K+
inward or outward depends upon the K+ concentration), Kir4.1,
FUNCTIONS was detected in Müller cells throughout the retinal layers and
The complex structure of Müller cells, spanning the width of was highly concentrated in the membrane domains of the endfeet
the retina with processes extending laterally, places them in a facing the vitreous humor and in membranes of processes
position to perform many different functions in the retina. ensheathing blood vessels. Thus, the vitreous humor and blood
Across the width of the retina, Müller cells are thought to vessels may act as potassium sinks. A third Kir channel, Kir5.1,
control and regulate molecules and ions. In localized areas, was found in rats to be present diffusely in Müller cell bodies
Müller cells can control the extracellular microenvironment and appeared to form a heteromeric channel with Kir4.1 in the
of neuronal cell bodies and synapses. Müller cells play an distal processes (fiber baskets, Fig. 125.1) in the subretinal
important role in retinal signaling processes by removing space and in processes of Müller cells located in the IPL, INL,
neurotransmitters rapidly from the extracellular space. Also, OPL, and ONL but not in the endfeet and processes surround-
due to intimate association with the retinal vasculature, Müller ing blood vessels where homomeric Kir4.1 channels are found.
cells are implicated in movement of substances into and out of The properties of the two channels are different (e.g., pH
the retinal vessels. Specialized regions of Müller cells, located sensitivity of the Kir4.1/Kir5.1 channel), and therefore, they are
adjacent to the regulated region of the neuron or endothelial thought to play different functional roles in K+ buffering.24
cell, contain the specific molecules needed to carry out these Studies in rodents have shown co-localization of Kir4.1 chan-
support functions. The following sections describe some of the nels with the water channel, aquaporin-4, in membrane domains
functions attributed to Müller cells in the adult eye. facing the vitreous humor and blood vessels, indicating that
potassium and water movement are closely linked in Müller
Regulation of the Extracellular Space cells. The localized distribution of aquaporin in Müller cell mem-
Potassium branes indicates that Müller cells mediate osmotic gradients
Light-evoked neuronal activity causes an increase in extra- associated with K+ movement and function to direct water flux
cellular K+ concentration ([K+]0) in the synaptic layers while to specific extracellular compartments. The tight co-localiza-
[K+]0 decreases adjacent to the photoreceptor nuclei. One very tion of the two channels may support the movement of K+
important function of Müller cells is the regulation of [K+]0 while preventing osmotic imbalances in the retina.25
since fluctuations alter neuronal excitability. In a process Another type of potassium channel, the tandem pore chan-
termed ‘K+ siphoning’,21 movement of K+ from areas of high nels TASK-1 and TASK-2, has been localized in the cell body
1635
RETINA AND VITREOUS

and fiber baskets of mammalian Müller cells (e.g., rat, mouse,


and guinea pig). These channels, along with Kir 4.1 channels, Cone Müller cell
are thought to maintain the negative resting potential of Müller
cells in nonpathological conditions and could play a minor role Light E
in K+ siphoning. Skatchkov et al. suggest these channels are X 11-cis-retinal
T
important in maintaining membrane hyperpolarization in areas R RGR
where Kir4.1 channels are sparse and strongly rectifying A all-trans-retinal
channels, such as Kir2.1, are located (e.g., photoreceptor cell 11-cis-retinal C
11-cis-retinol E
bodies). TASK channels may become important in potassium L RDH10
regulation when Kir4.1 channels are downregulated (see section all-trans-retinal L
on Müller cell gliosis).26 U
11-cis-retinol
L
all-trans-retinol A
CO2 and pH regulation (vitamin A) R all-trans-retinol
The high metabolic rate of the retina leads to the generation S isomerase
of large amounts of CO2. Müller and RPE cells contain the P
sodium–bicarbonate co-transporter, pNBC1,27 and large stores A
C
of carbonic anhydrase, an enzyme that converts CO2 to bicar- E
bonate in a reaction helping to maintain the extracellular pH.28
SECTION 10

One form of carbonic anhydrase, CAXIV, is located in the FIGURE 125.4. Cone opsin recycling pathway in Müller cells.
membrane domains of Müller cells facing the photoreceptors Generation of 11-cis-retinol from all-trans-retinol (released into the
and is also enriched in the Müller cell endfeet. The presence of extracellular space) (black text).35,36 Proposed generation of 11-cis-
CAXIV in the Müller cell endfeet has led to the notion that the retinal from all-trans-retinol involving the enzymes, retinal G protein-
endfeet are a clearance route for CO2 to the vitreous cavity and coupled receptor (RGR) and all-trans retinol dehydrogenase (RDH10)
retinal blood vessels.28 (blue text).37

Neurotransmitter uptake and conversion


Müller cells are important in regulating the extracellular levels uptake is thought to be stimulated by synaptically released
of glutamate, the major excitatory transmitter of neurons of the glutamate.29 Similarly, glycogen breakdown and release of
retina, and g-aminobutyric acid (GABA), the major inhibitory metabolic substrates is thought to be regulated by neuroactive
transmitter. Potent uptake systems and degradative pathways substances released by neurons.14
exist for both transmitters in Müller cells. Glutamate is also
taken up by neurons where it is recycled, but in Müller cells it Visual Pigment Recycling
is thought to be inactivated by conversion to glutamine in a In studies of cone-dominant eyes of chicken and ground
reversible reaction catalyzed by glutamine synthetase, which in squirrel, Müller cells were found to contain an alternate visual
the retina is present only in Müller cells. The conversion of cycle for cone opsin regeneration that is distinct from the visual
glutamate to glutamine involves ammonia fixation, ammonia cycle in RPE cells (Fig. 125.4, cycle indicated with black text
production being associated with increased synaptic activity. and black arrows). All-trans-retinol, which is released into the
Thus, another important function of Müller cells may be extra- extracellular space by rods and cones following light activation
cellular ammonia detoxification.29 Both glutamine synthetase of an opsin pigment molecule, can be taken up by Müller cells,
and the Müller cell-specific glutamine transporter, GLAST (L- directly converted to 11-cis-retinol by a unique all-trans-retinol
glutamate–L-aspartate transporter), are co-localized in the fine isomerase, and bound by cellular retinaldehyde binding protein
horizontal processes of Müller cells that ensheath rod photo- (CRALBP). The 11-cis-retinol is released into the extracellular
receptor synaptic terminals.30 space where it can be taken up by cones for regeneration of
Müller cells contain GABA transporters (GAT-1 and GAT-3) visual pigment utilizing the cone-specific 11-cis-dehydrogenase.
and the GABA-degrading enzymes, GABA-transaminase Both 11-cis-retinol and all-trans-retinol are bound to interpho-
(GABA-t) and succinic semialdehyde dehydrogenase (SSADH), toreceptor retinoid binding protein (IRBP) during translocation
located in mitochondria, indicating their role in the regulation between the cones and Müller cells.35 The energy source for this
of GABA concentration in the extracellular space. In mam- isomerization may be a palmitoyl CoA-dependent esterification
malian Müller cells, the predominant transporter is GAT-3, of 11-cis-retinol and all-trans-retinol.35,36
which is found in the plasma membrane throughout the cell.31 In RPE cells, RDH10 (an all-trans-retinol dehydrogenase)
Once transported into the cell, GABA and a-ketoglutarate are oxidizes all-trans-retinol to all-trans-retinal in the photic visual
catalyzed by GABA-t to glutamate and succinic semialdehyde. cycle (see section on Role in Visual Cycle in The Retinal
Succinic semialdehyde is then degraded by SSADH to succinate, Pigment Epithelium). When exposed to light, the RPE retinal G
which can then enter the tricarboxylic acid cycle.14 protein-coupled receptor (RGR) isomerizes all-trans-retinal to
11-cis-retinal. Both RDH10 and RGR have been found in
Glycogen Metabolism Müller cells (although in lower levels than found in RPE),
Müller cells are thought to play a central role in maintaining the indicating the possibility that Müller cells can generate 11-cis-
nutritional needs of the retina (see Winkler et. al and references retinal from all-trans-retinol utilizing mechanisms similar to
therein32). Glucose is thought to be transported from the blood those found in RPE cells (Fig. 125.4, proposed cycle indicated by
by Müller cells via facilitated diffusion mediated by the glucose blue text and blue arrows).37
transporter, GLUT 1, converted into glycogen, and stored.33
GLUT 1 has been demonstrated in the plasma membrane of Neuronal Activation of Müller Cells
human Müller cells.33 (GLUT 2 has been localized to the apical As noted above, Müller cells affect the function of retinal
processes of rat Müller cells (fiber baskets of Fig. 125.1), neurons by regulating the extracellular environment. However,
indicating one possible function for these structures.34) Müller neurons seem to be able to affect Müller cells directly by
cells contain the metabolic pathways for glycogen deposition neuronal signaling. Light stimulation of the retina, for example,
1636 and are the sole site of glycogen storage in the retina. Glucose results in a transient increase in Ca2+ in the Müller cell
Müller Cells and the Retinal Pigment Epithelium

processes located in the IPL, which spreads to the cell endfeet at glaucoma, mechanical injury, choroidal melanoma, and retinal
the vitreal surface. The resultant release of ATP by Müller cells tissue obtained from partial retinectomies to relieve traction.50,51
is thought to enhance or depress neuronal activity in a neuronal Loss or diminished activity of ion channels can lead to a change
excitability feedback mechanism.38 in the Müller cell membrane potential and affect the ability of
Müller cells to take up glutamate. Since glutamate is highly toxic
Müller Cell Contribution to the ERG to the photoreceptors, extracellular regulation of glutamate is
Early studies by Witkovsky39 on isolated retina showed that critical for vision preservation.50,52,53
Müller cells contribute to the negative slow PIII of the c-wave of Although there are no diseases of the retina in which Müller
the ERG. The Müller cell response was attributed to the photo- cells are known to be the primary site affected, Müller cells
receptor-dependent light-evoked decrease in subretinal extra- appear to be involved in many retinal disorders.14 The role of
cellular [K+]0.40 (RPE contributes to the positive component Müller cells in specific diseases is addressed elsewhere
of the c-wave; please see section, Ion Transport in The Retinal (Chapters 182 and 183). Müller cell responses in general are
Pigment Epithelium). Further evidence indicating that K+ discussed in the following sections.
movement by Müller cells contributes to the ERG is the loss of
the PIII wave in Kir4.1 knockout animals.41 Some studies Müller Cell Gliosis
indicate that K+ buffering by Müller cells may also contribute to Pathologies involving neuronal degeneration in the retina
the b-wave although conflicting studies attribute the b-wave to appear to be associated with some level of Müller cell activation
depolarizing bipolar cells.14 The lack of change in the b-wave (gliosis).44 Müller cell response or activation can be detrimental

CHAPTER 125
response in the Kir4.1 knockout mice supports the neuronal or beneficial, depending on the magnitude of the response. For
origin of the b-wave.41 example, Müller cell activation can be beneficial if modest, as
activation could be helpful due to increased growth factor
Phagocytosis secretion by the cells (see section on Neuroprotection). If gliosis
Müller cells can phagocytose a variety of substances (e.g., latex results in proliferation and/or is long-lasting, gliosis can lead to
beads, carbon, copper particles, blood cells) and are thought to neuronal degeneration (e.g., subretinal fibrosis following retinal
assist the RPE cells in phagocytosis of outer segments.14 In devel- detachment and epiretinal membrane formation leading to
opment and in retinal disease, Müller cells can phagocytose retinal detachment).54 Müller cell activation can be detected by
debris from dying neurons.42 Also, the phagocytotic capacity of upregulation of the intermediate filaments, vimentin and glial
Müller cells may aid in maintaining the clarity of the vitreous fibrillary acidic protein (GFAP). Since GFAP expression is not
humor.42 detected in healthy retinas, GFAP detection can be used as an
early indicator of reactive Müller cells.14 Another early indicator
of Müller cell activation is activation of ERKs (extracellular
PATHOLOGY signal-regulated kinases) and release of neuroprotective growth
Müller cells play a prominent role in injury and diseases of the factors (see section on Growth Factor, Cytokine, and Neuro-
retina. They exhibit the ability to respond to subtle insults by trophic Factor Secretion).55 Gliosis can lead to changes in cell
modulation of normal functions (e.g., moderating the rise in shape including changes to or loss of processes.56 In the
extracellular K+ and glutamate following plasma leakage into detached retina, gliotic responses also include downregulation
the retina43) as well as the ability to undergo marked functional of glutamine synthetase, cellular retinaldehyde-binding protein,
and morphological change in response to more severe insults. K+ channel expression, and carbonic anhydrase.57 Studies per-
Müller cells can undergo cell division and migration.44 Müller formed in rats (ischemia model), pigs, and rabbits (detachment
cells can respond to a number of growth factors, cytokines, and models) show that one of the consequences of Müller cell
neurotransmitters derived from photoreceptors and other activation can be the loss or relocation of Kir4.1 channels.
neurons and RPE cells. In pathological conditions, inflam- Impairment of potassium buffering capabilities through loss of
matory cells, platelets, and plasma can activate Müller cells.45 Kir channels could lead to the inability of the cells to maintain
Some Müller cell responses are related to tissue repair (e.g., their hyperpolarized state and thus lead to breakdown of normal
resurfacing of Bruch’s membrane following RPE and photo- homeostasis (e.g., K+ buffering and water regulation) resulting
receptor loss to maintain the blood–retina barrier in atrophic in edema and retinal degeneration.57–59 In the rabbit and pig
macular lesions14) and wound healing (e.g., Müller cell wound detachment models, and as sometimes occurs in patients
closure of the OLM following macular hole repair by vitrectomy following retinal reattachment, loss of photoreceptors (or visual
with cortical vitreous and epicortical vitreous membrane function) can be seen in attached areas adjacent to the
peeling46) while other responses are related to neuroprotection reattached retina. This loss is attributed to activation of Müller
(see section on Growth Factor, Cytokine, and Neurotrophic cells adjacent to the area of detachment.57,59
Factor Secretion) and detoxification (e.g., Müller cell response to As previously mentioned, Müller cells can respond to a
elevated ammonia levels in hepatic retinopathy47,48). number of stimuli, in particular mitogenic growth factors.45
Additionally, since Müller cells play a prominent role in Blood-derived factors (e.g., thrombin) released from the break-
retinal homeostasis, any dysfunction involving Müller cells can down of the blood–brain barrier may trigger the proliferative
have a profound effect on retinal function. For example, long- response through downregulation of Kir channels.45,60 In
term replacement of the vitreous with a fluid, such as perfluoro- addition to Kir channels, which mediate potassium levels
carbon or silicon oil, incapable of acting as a K+ sink could during light-activated K+ elevation in the extracellular space (K+
interfere with Kir function and might lead to outer retina siphoning), Müller cells express other ion channels including
degeneration especially when little vitreous fluid remains.49 Ca2+-activated K+ channels.60 Studies comparing Müller cells
Similarly, damage to the Müller cell endfeet, such as occurs in from diseased and healthy retinae show impaired K+
retinoschisis, would impair spatial K+ buffering capabilities of conductance due to loss of Kir channels in cells from diseased
damaged cells.42 In a study comparing healthy versus diseased retinae although Ca2+-dependent K+ channels are activated.50,52
eyes, Müller cell ion channels appeared to be affected in the The Ca2+-dependent K+ channels are activated when the mem-
diseased eye. For example, changes in Müller cell membrane brane potential of the cell is markedly reduced (as in patho-
properties (e.g., down-regulation of Kir current density) have logical conditions when the Kir channels are lost) and are not
been found in diseased retinae from human eyes with secondary thought to play an important role in potassium siphoning in the 1637
RETINA AND VITREOUS

normal retina.61 Müller cell proliferation can be triggered by similar in size to that of macrophage iNOS and thus may
mechanisms activating these Ca2+-dependent K+ channels.60 be important as a reactive mediator in inflammation and
The mechanism of Ca2+-dependent K+ channels activation is infection.79 However, the exact role of iNOS in pathology and
thought to be through activation of P2 receptors. The P2Y regulation of NO by Müller cells is not understood fully.
receptor, minimally active in healthy retinae, is activated by
ATP. Following P2Y receptor activation, the cells become Müller Cell Immune Modulation
unresponsive to ATP and cease to proliferate. The receptors can Experimental evidence from in vitro cell culture studies
be re-sensitized to ATP following exposure to growth factors indicates that Müller cells may provide an immune modulation
(e.g., platelet-derived growth factor (PDGF), epidermal growth function in the retina. Müller cells, along with RPE and vascular
factor (EGF) and nerve growth factor (NGF).57,62 endothelial cells, have been shown to express MHC class II
Müller cell proliferation also can be initiated by mitogenic (MHCII) antigens.14 Müller cells from rat retina can be induced
growth factors. Basic fibroblast growth factor (bFGF), which to proliferate following addition of conditioned media from
has been shown to be released intraretinally following injury activated spleen cells or activated lymphocytes and can express
(mechanical or light-induced) and retinal detachment, is a MHCII antigens.80 Müller cells in the human eye of a patient
potent growth factor in Müller cell activation and proliferation. with hypopigmented subretinal fibrosis and uveitis syndrome
Other growth factors shown to evoke Müller cell proliferation were shown to express MHCII antigens by immunohisto-
include PDGF, heparin-binding epidermal growth factor (HB- chemical staining.81 However, when Müller cells expressing
EGF), and transforming growth factors (TGF).63 MHCII antigens were cultured with activated T cells,
SECTION 10

proliferation of the latter was inhibited. The prevention of T cell


Neuroprotection proliferation required physical contact between Müller cells and
Elevation of the extracellular glutamate concentration is highly T cells, indicating that the inhibition was through a membrane
toxic to retinal neurons.56 Extracellular glutamate concentra- bound protein present in the cells.82 When this membrane
tions are highly regulated through Müller cell uptake involving bound protein is inhibited (e.g., using glucocorticoids), Müller
the GLAST transporter. Excess glutamate can be generated in a cells can function as an antigen presenting cell.83 Thus, Müller
variety of retinal conditions including ischemia, hypoglycemia, cells have the potential to inhibit or stimulate T cell prolifera-
glaucoma, diabetes, and trauma.14,56 Accordingly, some studies tion. Since lymphocytes migrating out of retinal capillaries con-
have correlated increases in GLAST activity with elevated tact Müller cells, the interaction between the two cells could
glutamate levels.56 However, in diabetic retinopathy, GLAST either suppress retinal inflammation or enhance it.14
levels appear to decrease (possibly due to oxidative damage since
the effects can be reversed by exposure to a disulfide-reducing Growth Factor, Cytokine, and Neurotrophic Factor
agent) leading to elevated glutamate levels and resultant glutamate Secretion
excitotoxicity.64,65 Müller cells and RPE can up-regulate the expression of growth
Studies in rodents have shown that intraocular injection of factors, cytokines, and neurotrophic factors in response to
neurotrophic factors can promote photoreceptor survival in retinal injury or other stimuli (Table 125.1).84 In addition to
retinal degenerations caused by genetic or environmental acting on photoreceptors, Müller cells also may be involved in
factors.66–68 Neuroprotection of photoreceptors following injec- ganglion cell and bipolar cell neuroprotection.56 Secreted growth
tion of growth factors (e.g., brain-derived neurotrophic factor factors such as bFGF, VEGF (vascular endothelial growth
(BDNF), ciliary neurotrophic factor (CNTF), neurotrophin-3 factor), and PEDF (pigment epithelium-derived factor) appear
(NT-3), or bFGF) is thought to be mediated by activation of to provide constitutive trophic support to the photoreceptors
Müller cells, since photoreceptors are not activated by exposure and vascular endothelia.85–87 Changes in the endogenous
to these growth factors as indicated by lack of intracellular sig- concentration of these growth factors can affect surrounding
naling pathway activation.69,70 Sustained growth factor delivery cells. For example, PEDF downregulation results in increased
by viral vectors to activate Müller cells offers a possible treat- Müller cell secretion of VEGF and TNF-a (tumor necrosis
ment to prevent retinal degeneration.71,72 The mechanism by factor), which may contribute to inflammation and vascular
which activated Müller cells affect photoreceptor survival is not changes associated with diabetic retinopathy.86,87 Müller cells
known. contribute to the endogenous levels of VEGF, PEDF, TGF-b2,
TSP-1 (thrombospondin), BDNF, and bFGF.88–90 Changes in
Nitric Oxide Synthetase bFGF secretion by Müller cells can result in either protection
Nitric oxide synthetase (NOS) catalyzes the formation of highly (increased bFGF secretion) or increased apoptosis (decreased
reactive nitric oxide (NO) from L-arginine. Nitric oxide has bFGF secretion) in adjacent photoreceptors.91 Microglia are
been implicated in the regulation of retinal blood flow, in visual an endogenous source of growth factors leading to Müller cell
transduction, and is toxic to microorganisms.73 As such, NO stimulation resulting in increased (e.g., NT-3) or decreased (e.g.,
may also contribute to retinal pathology and has been impli- NGF) bFGF secretion.92
cated in reperfusion injury following ischemia, diabetic vascular Both in vivo and in vitro studies show that Müller cells respond
damage, HIV retinitis, and endotoxin-induced uveitis.73,74 to many different stimuli and can secrete different growth factors,
Histochemical staining for NOS has shown its presence depending on the stimulus or culturing conditions (Table 125.1).
throughout the retina;73 mRNA for inducible and constitutive In addition to providing neuroprotective growth factors and
forms of nitric oxide synthetase (iNOS and cNOS, respectively) factors capable of modulating blood flow and vascular
have been found in human retinal tissue.75 cNOS is calcium- permeability, Müller cells can produce several pro-inflammatory
dependent; iNOS is calcium-independent. iNOS is expressed in cytokines in response to viral infections.93,94
Müller cells following stimulation by interferon (IFN)-g and Two types of receptors, Trk (high-affinity neurotrophin
lipopolysaccharide76 and, following upregulation, can produce tyrosine kinase receptor) and p75 (low-affinity neurotrophin
sustained levels of NO. cNOS (found in mammalian Müller receptor), have been identified in pro-survival (Trk) and anti-
cells of mice77 and the tree shrew,78) is relatively short-lived and survival (p75) signaling by neurotrophins.104 In a light damaged
produces picomolar quantities of NO.73 (iNOS has also been mouse model, enhanced TrK and p75 labeling was localized to
detected in human RPE following stimulation by interleukin-1 different parts of Müller cells.92 These studies showed photo-
1638 and IFN.73) mRNA for iNOS found in Müller cells of mice is receptor survival following light damage with p75 blockade
Müller Cells and the Retinal Pigment Epithelium

TABLE 125.1. Müller Cell Growth Factor, Cytokine, and Neurotrophin Secretion Following Stimulation

Growth Factor Secreted Stimulus Model Associated Retina Pathology or Function References
95
BDNF, NGF, NT-3, NT-4, Glutamate Cultured rat MC Neuroprotection during glutamate toxicity
GDNF
96
VEGF TGF-b, hypoxia Cultured rat MC Hypoxia-induced angiogenesis
97
TNF-a, NO IFN-g and LPS RCS cultured PR degeneration in retinal dystrophies
rat MC
93,94
TNF-a, IL-6, IFN-a, IFN-b HSV-1, IFN-g Mouse and Immune and inflammatory responses
cultured mouse MC
44
bFGF, CNTF, GDNF Light, mechanical Light damaged rat Neuroprotection
98,99
injury
88
VEGF, TSP-1 Hypoxia Cultured human TSP-1 angiogenesis inhibition; VEGF
MIO-M1 cells and angiogenesis induction
guinea-pig MC

CHAPTER 125
100
VEGF HB-EGF MIO-M1 PVR
87
VEGF, TNF-a Reduced PEDF level Cultured rat MC Diabetic retinopathy
101
VEGF Advanced glycation Cultured human Diabetic retinopathy
endproducts (AGE) MC
102
bFGF bFGF Cultured rat MC Injury response to secreted bFGF
(neuroprotection)
92
bFGF NT-3 Cultured rat MC Neuroprotection
103
CNTF Optic nerve damage rat Neuroprotection
BDNF, brain-derived neurotrophic factor; NGF, nerve growth factor; NT, neurotrophin; GDNF, glial cell line-derived neurotrophic factor; MC, Müller cells; VEGF, vascular
endothelial growth factor; TGF, transforming growth factor; TNF, tumor necrosis factor; NO, nitric oxide; IFN, interferon; LPS, lipopolysaccharide; RCS, Royal College of
Surgeons; PR, photoreceptor; IL, interleukin; HSV-1, herpes simplex virus type 1; bFGF, basic fibroblast growth factor; CNTF, ciliary neurotrophic factor; TSP,
thrombospondin; MIO-M1, spontaneously immortalized human Müller cell line; HB-EGF, heparin-binding epidermal growth factor-like growth factor; PVR, proliferative
vitreoretinopathy; PEDF, pigment epithelium-derived factor.

or p75 absence in p75 knockout mice p75 activation was of the vision-mediating visual pigments of the photoreceptors,
hypothesized to suppress growth factor release. Based on the transport from the systemic circulation of molecules necessary
results of experimental studies in different model systems, it for the structure and function of photoreceptors and
may be that p75 signaling is not the main pathway mediating maintenance of a dry, pH-balanced, and immune-privileged
cell death by Müller cells.105–107 Additional stress pathways or subretinal space. Unlike Müller cells, RPE are easy to study,
impairment of survival signals such as those initiated by TrK isolate, and culture. This fact has resulted in a vast and rich
receptors may be required for p75 mediated activation of cell database of knowledge on RPE structure and function.108,109
death.104 This chapter provides a broad introduction to the RPE.

FUTURE DEVELOPMENTS EMBRYOLOGY AND DEVELOPMENT OF RPE


Müller cells have complex involvement in a variety of functions RPE progenitors originate from the dorsal portion of the optic
in the developing, normal, and pathological retina. Much work vesicle (Fig. 125.5). Cells in the early stages of optic vesicle
remains to be done to characterize and clarify the mechanisms development are similar morphologically and molecularly and
involved in retinal homeostasis, particularly in the human eye. express numerous transcription factors (e.g., Otx2, Pax6, Rx1,
Additionally, the mechanisms underlying the morphological, Six3)110 that initiate eye development (Fig. 125.5). At this stage,
cellular, and molecular changes Müller cells undergo as part of all the cells of the optic vesicle can give rise to the neural retina,
the retina’s pathophysiological responses are not understood optic stalk, or RPE.111–113 Signals from the surrounding tissues
fully. Future studies will lead to insights to these unanswered determine the fate of different regions of the optic vesicle, e.g.,
questions, which should foster the development of therapies whether the cells develop into RPE or neural retina. The surface
specifically targeting Müller cell reactivity or dysfunction in ectoderm generates signals that promote neural retinal develop-
retinal pathology. ment and suppress RPE development.114 These signals are
molecules that belong to the FGF family. Thus, the outer rim
THE RETINAL PIGMENT EPITHELIUM of optic vesicle adjacent to the surface ectoderm (that later
invaginates to form the inner layer of the optic cup) eventually
develops into the neural retina rather than into RPE.115,116
OVERVIEW OF THE RETINAL PIGMENT Conversely, the mesenchyme surrounding the posterior aspect
EPITHELIAL CELL AND ITS FUNCTIONS of the optic vesicle (the prospective RPE) generates signals that
Retinal pigment epithelium (RPE) is a simple monolayer of activate the RPE development pathway and suppress neural
postmitotic, melanin-laden cells lying between the photorecep- retinal pathway.117 Members of TGF-b superfamily have been
tors and the choroid but serving functions that are critical to the shown to play a role.117,118 These signals act on the target cells
eye. These include the formation of a blood–retinal barrier, and activate or suppress transcription factors like Mitf
phagocytosis of the photoreceptor outer segments (OS), recycling (microphthalmia-associated transcription factor),119,120 Otx 1639
RETINA AND VITREOUS

A 1 2 3
presumptive neural retina
extraocular RPE
RPE
mesenchyme
lens

lens placode
lens pit

presumptive
neural retina

optic stalk optic nerve


optic vesicle

B 1 2 3

extraocular
mesenchyme

Otx2
SECTION 10

TGF-b like Otx Wnt Pax6 Wnt


Otx1 Otx
Pax6 Mitf Hh
Mitf
Six3
Rx1
prospective RPE BMP
melanogenic
genes
Otx2 Pax6 Rx1
Otx1
Pax6 Six3 Chx10 differentiated RPE
Six3
Rx1 FGF
prospective neural retina

lens placode
naive optic vesicle patterned optic vesicle RPE of the optic cup

FIGURE 125.5. Diagrammatic representation of the development of the RPE. A. The optic vesicle develops as an outpouching of the diencephalon
(A-1). The vesicle invaginates to form an optic cup (A2, and A3). Signals from the surface ectoderm (e.g., FGF and others) (A-1) promote neural
retinal development (A3) from the ventral part of the optic vesicle while the dorsal part of the optic vesicle develops into the RPE following activation
by signals (e.g., TGF-b) from the adjacent mesenchyme. B. Until the activation of signals specifying the fate, the cells of the optic vesicle are
indistinguishable (multiple colors). TGF-b from extraocular mesenchyme favors development of RPE while FGF from the lens placode suppresses
RPE development and favors neural retinal development (B1). Different transcription factors activated by the extracellular signals become
confined to prospective RPE and prospective neural retina (B2). The interaction of the activated transcription factors with other regulatory
molecules consolidates the identity of the cells and promotes differentiation. Otx: orthodenticle-related transcription factor; Pax6: paired-box
transcription factor; Hh: hedgehog; BMP: bone morphogenetic protein; Wnt: wingless and integration site gene; Six: sine oculis homeobox;
Chx10: C.elegans ceh-10 homeo domain containing homolog; Mitf: microphthalmia-associated transcription factor; Rx: retina homeobox.
See text for details. Figure based on Martinez-Morales JR, Rodrigo I, Bovolenta P: Eye development: a view from the retina pigmented epithelium. Bioessays 2004;
26:766–777. Reprinted with permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc. copyright 2004.

(orthodenticle-related transcription factors),121,122 and Pax6 important for development of the adjacent neural retina,
(paired-box transcription factors).123,124 Mitf, for example, binds choroid, and sclera.129,130
and transactivates the genes involved in terminal differentiation The presence of RPE is necessary for the formation of photo-
of RPE, including tyrosinase.119 Further differentiation of receptor discs131 and photoreceptor differentiation.132 Loss of
RPE may be influenced by a different set of signaling factors, RPE leads to improper retinal development.129,131 Neural retinal
including bone morphogenetic protein (BMP)125 and hedgehog development (generation of cells and differentiation) occurs along
(Hh)126,127 families and cell-cycle regulators. The factors men- a gradient starting in the center and extending to periphery.133,134
tioned above are not unique to RPE. How these factors promote RPE maturation precedes neural retinal maturation135 but retinal
formation of RPE in the optic vesicle and different structures in development does not depend solely on RPE (e.g., hedgehog
different parts of the forebrain is not clearly understood. signaling from ganglion cells is important for the development
By day-30, the invagination is complete, and the two layers of of laminar organization of neural retina127). Photoreceptors also
the optic vesicle are apposed. RPE initially is pseudostratified influence development of RPE. Neural cell adhesion molecule
but matures into a monolayer.3 On day-35, melanin granules (NCAM) localization to apical regions of RPE, for example,
appear in RPE cells. This is the earliest site of pigmentation requires the presence of photoreceptors; absence of photo-
in the body. By 6-weeks gestation, RPE basement membrane receptors leads to basolateral redistribution.136 One possible
becomes evident, and by 10-weeks, the apical processes develop. mechanism by which RPE may be influencing development of
Further differentiation results in apicobasal polarity and for- neural retina is by release of a soluble factor.129,137 This factor is
mation of tight junctions to establish the outer blood–retinal believed to be ATP, and it increases cell proliferation in the
barrier. Development of tight junctions occurs in three stages. neural retina.138 Melanin-related agents in RPE appear to be
In the first stage, specific proteins, g-tubulin in the apical responsible for some aspects of retinal development. Human
portions of the cells, and a1b3 integrin in basal portions are albino eyes can exhibit foveal hypoplasia and have abnormal
expressed, but tight junctions are rudimentary. In the second chiasmal projections.139,140 Lack of pigment results in temporal
stage, Na+–K+–ATPase is apically polarized. In the final stage, delay in neural retinal differentiation as well as abnormalities in
mature tight junctions composed of ZO-1, occludin, and ganglion cell axon decussation (i.e., greater numbers of crossed
claudin are formed.128 fibers compared to pigmented eyes). These retinae are under-
In accordance with a general theme of development, inter- developed with abnormally thin INLs and ONLs. There is a rod
action of the RPE with the surrounding structures is important cell deficit (30%), but the cone cell numbers and cone mosaic
1640 for RPE development and maturation. Conversely, RPE is are unaffected.140
Müller Cells and the Retinal Pigment Epithelium

RPE cells also may play a role in melanocyte differentiation matin. Many RPE are bi- or multinucleated (one out of 30 RPE
and vascular development in the choroid.130 RPE secretes have multiple nuclei), more commonly in peripheral RPE
growth factors that influence endothelial cell and photoreceptor cells.149,161 Mitosis has not been seen in adult RPE cells in situ.
differentiation.141–144 Through its transport capabilities, RPE If there is a defect resulting from cell loss due to any reason, the
may relay growth regulating signals from the neural retina that neighboring cells spread to fill in the defect.162 However, RPE
influence choroidal and scleral growth, thereby potentially are capable of proliferating if cells are isolated and grown in
modulating the development of the latter structures.145 RPE is culture.
capable of secreting all the components of Bruch’s membrane.146 RPE cell cytoplasm contains mitochondria (present mostly
Bruch’s membrane layers develop sequentially from inside-out, between the nucleus and the basal surface) and extensive
i.e., the RPE basement membrane forms first, followed by pro- smooth endoplasmic reticulum (characteristic of only one other
gressive development of the outer laminae. The choriocapillaris cell in the eye, the Müller cell). In the periphery, the amount of
basement membrane is the last layer to form.147,148 smooth endoplasmic reticulum is lower, but cells have more
rough endoplasmic reticulum (present in the apical portion
of the cell). RPE cells contain lysosomal granules, melanin
ANATOMY AND CELL BIOLOGY granules, and lipofuscin. Pigment granules measure 2–3 mm in
RPE forms a monolayer of cells sclerad to the neural retina, length and 1 mm in diameter. They are present mostly in the apical
extending from the edge of the optic disc, posteriorly, to the ora portion of the cell. The granules are either ovoid or needle
serrata, anteriorly. When viewed en face, the cells appear shaped, the former located in perinuclear cytoplasm and the

CHAPTER 125
hexagonal. In cross section, cells are cuboidal and contain a latter near the apical region. Melanin granules may fuse with
basally located nucleus and apically located pigment granules. lysosomal granules or lipofuscin to form complex melano-
The RPE size varies with the location in the eye; submacular lysosomal or melanolipofuscin granules. The content of ‘pure’
RPE cells have a diameter of 14 mm and are 10–14 mm tall. RPE melanin declines with age, whereas the number of complex
in the peripheral regions, especially near the ora serrata, are melanin granules increases. Submacular RPE cells contain more
flatter and can have a diameter of up to 60 mm.149,150 The RPE complex granules than RPE elsewhere, particularly in young
apical surface is characterized by numerous microvilli, which eyes. With age, lipofuscin granules increase in RPE cells, the
are of two different lengths; fine, filopodia-like microvilli of largest increase occurring between the first and second decade
5–7 mm in length interdigitate with the photoreceptor outer seg- of life. Due to the increase in granules, cytoplasmic volume not
ments; shorter, broader, lamellopodia-like microvilli of ~3 mm occupied by pigments (‘free space’) decreases with age.163
length enclose the tips of the OS (Fig. 125.6). The basal RPE The RPE is polarized with a distinct distribution of molecules
surface exhibits complex infoldings that may extend 1 mm or and ion channels on the apical and basal surfaces. For example,
more into the cytoplasm. The RPE rests on Bruch’s membrane, the apical membrane contains avb5 integrin and Na+–K+–ATPase
which is a pentalaminar structure that includes the RPE base- while the basal membrane contains a6b1 integrin. The polarity
ment membrane (on which RPE rests directly), the inner is achieved by a specific distribution of molecules to either
collagenous layer, the elastic layer, the outer collagenous layer, surface and is maintained by the presence of tight junctions
and the choriocapillaris basement membrane. that prevents intra-membrane diffusion of molecules between
There are ~4–6 million RPE cells in each human eye. The the apical and basal domains of the cell membrane.164 Organ-
cell density decreases from center (4000 cells/mm) to mid- ization of the cytoskeletal elements also helps to maintain the
periphery (3000 cells/mm) to far periphery (1600 cells/mm).151 polarity.165
This distribution is likely due to the growth of the eye being Laterally, RPE connect with each other via tight junctions
confined to the ora–equatorial region from infancy to adoles- (zonula occludens [ZO]), adherens junctions (zonula adherens),
cence and the lack of RPE cell proliferation after development. and gap junctions.365 Tight junctions are responsible for the
RPE cells increase in size in the peripheral region near the ora barrier function of RPE monolayer. In freeze-fracture images,
serrata from infancy to adolescence. In the periphery, RPE cell tight junctions appear as ‘sealing strands’ encircling the cell
density is highest in the nasal quadrant; all other quadrants near the apex. By transmission electron microscopy, the tight
have a lower packing density. Cell density decreases by ~0.3% junctions appear as close apposition of the plasma membrane of
per year with increasing age, most marked under the fovea and adjacent cells near their apices. These strands are extracellular
mid-peripheral retina.151 This rate of loss is more in blacks than domains of tight junction proteins claudin and occludin.164 The
in whites.152 Cell number or cell density is not affected by cytoplasmic side of the cell membrane contains ZO proteins,
gender, but it correlates with rod cell density. The rod:RPE ratio ZO-1 and ZO-2, that connect to the actin filaments present
is 0 in the fovea and increases to ~19 rods for every RPE in the in the cytoplasm as well as activate signal transduction
periphery.151 In the periphery, ratio of RPE to rods is highest in pathways.164
the nasal followed by the superior quadrant. The other two The zonula adherens is formed by interaction of cadherin
quadrants have similar densities. The cone:RPE ratio is highest molecules. These junctions appear as a 200Å separation between
under the fovea (~40 cones/RPE), declines toward mid- the plasma membranes of adjacent cells. The cytoplasmic domain
periphery (~1), and increases toward the far periphery because of cadherins interacts with vinculin, catenins, a-actinins, and
of high cone density in that region.151,153 The ratio of RPE cells actin filaments. Actin filaments are arranged circumferentially
to photoreceptors does not change with age because the cone and maintain the hexagonal shape of RPE cells. Gap junctions
and rod numbers also decline with age.152,154–158 Hexagonal are present toward the base of the cell and are formed by
shape is lost under the foveal area with age.159 connexins.166 These junctions allow intercellular communica-
In addition to the topographic variation, there is cell-to-cell tion and transport of ions and metabolites.
heterogeneity in the RPE monolayer. This heterogeneity has
been termed cellular mosaicism-patches of cells of varying
phenotype forming the monolayer, cells within a patch being FUNCTIONS
identical but differing from cells in an adjacent patch.160 Role in Retinal Adhesiveness
The RPE nucleus is 5–12 mm in diameter and is somewhat The development of the retina by invagination of the optic cup
oval with the long axis parallel to the basal surface. The nucleus results in a potential space between the neural retina and the
contains one or two nucleoli and diffusely distributed chro- RPE. Yet in a fully developed eye the neural retina is attached 1641
RETINA AND VITREOUS

to the RPE. A force of ~100 dynes is needed to detach one process mediated by dopamine (which provides the light signal)
centimeter of retina in live rabbits; ~180 dynes in cats, and and melatonin (which provides the dark signal). It is not clear
~140 dynes in monkeys.167 Numerous factors are responsible whether the initiation of disc shedding occurs in the photo-
for keeping the retina attached. The most important contribu- receptors (e.g., shedding can sometimes occur in the absence of
tions to retinal adhesiveness come from interphotoreceptor RPE in vitro 184), whether RPE cells play a role (i.e., do RPE
matrix (IPM), which passively glues the neural retina to RPE; processes ‘bite’ off the tips?), or whether both processes occur
the metabolic activity of the RPE; and fluid transport across the during the initiation. Regardless, the interdigitation of OS with
RPE.168,169 RPE microvilli is important for phagocytosis to occur. When
Induction of retinal detachment by experimentally induced there is no interdigitation, as occurs in retinal detachment 185 or
perturbations demonstrates the role of RPE metabolic activity in mutant vitiligo mice that have RPE with short apical villi
in retinal adhesiveness. Removal of Ca2+ and Mg2+, for that do not ensheath the OS,186 phagocytosis does not occur
example, leads to reversible decrease in retinal adhesiveness; even though the RPE is capable of phagocytosis. The trigger
reducing the pH also leads to a decrease in adhesiveness.170,171 may be the length of OS,184 or some other signal that has not
A decrease in oxidative metabolism resulting from ischemia yet been identified. Light-induced oxidized tips of OS have been
leads to reduced retinal adhesiveness.168 proposed as a signal for phagocytosis by Sun and co-workers.187
Due to the resistance to free fluid movement across RPE Recognition and binding of OS to the RPE apical surface
resulting from the tight junctions, there is a hydrostatic occurs by receptor-ligand interactions. The receptors on RPE
pressure differential across the retina. This pressure differential that have been implicated include mannose receptor,188 CD36,
SECTION 10

forces fluid movement from the retina into the choroid. This MerTK,189 integrin avb5,190,191 and a gp55 glycoprotein.
differential, which could be as little as 0.001 mmHg, may be Blocking the mannose receptor inhibits RPE phagocytosis of
sufficient to keep the retina attached.172 If the direction of fluid ROS.188,192 The identity of the natural ligand for this receptor is
movement is reversed by injection of hyperosmolar solution not known. CD36 is a macrophage receptor that is involved in
into the vitreous cavity, retinal detachment occurs.173 Similarly, phagocytosis and binds to Toll-like receptor, TLR4.193 CD36
if choroidal osmolarity is increased, there is an increase in may also bind to light-induced oxidative products of photo-
retinal adhesiveness.174 Vitreous gel plays some role in retinal receptor lipids, and the latter may be a physiological signal to
attachment by physically keeping the retina apposed to RPE. Of initiate phagocytosis.187 CD36 binding is a necessary and
uncertain importance to retinal-RPE adhesion is the physical sufficient signal for activation of phagocytosis.190
ensheathment of OS by RPE microvilli as well as the frictional MerTK is tyrosine kinase receptor, i.e., the cytoplasmic
and electrostatic resistance resulting from the ensheathment. domain of the receptor is a tyrosine kinase that becomes
Recently, integrin avb5 has been shown to play a role in retinal- activated when an appropriate ligand binds to the receptor on
RPE adhesion, and, interestingly, the strength of adhesion the cell surface. Cells lacking MerTK are capable of binding to
mediated by avb5 was maximal at 3.5 hr after light onset when OS but are not able to ingest them.194 The cognate receptor for
the phagocytosis is complete.175 MerTK is believed to be a vitamin K-dependent growth arrest-
specific protein, Gas6.195,196 The role of Gas6 in phagocytosis
Phagocytosis has been demonstrated in vitro,197 but its role in vivo is not
While RPE cells are capable of nonspecific phagocytosis, a known. Integrin avb5 present on the apical membrane of RPE
property shared with other phagocytic cells like macrophages, binds to vitronectin. However, vitronectin may not be involved
RPE cells show a strong preference for photoreceptor OS.176 in OS phagocytosis in situ, and there may be other proteins
Nonspecific phagocytosis and phagocytosis in vitro is not involved;191 avb5 interacts with MerTK to mediate phago-
rhythmic while phagocytosis of OS is. By the time a human cytosis. It activates MerTK via focal adhesion kinase.198
RPE cell is 80 years old, it has phagocytosed ~108 OS.177 OS binding to RPE surface receptors activates second
Dysfunctional phagocytosis can lead to accumulation of debris messenger systems that lead to reorganization of cytoskeletal
in the subretinal space and cause loss of vision as is evident in elements to cause ingestion. MerTK activation causes an
Royal College of Surgeons (RCS) rats. These rats have a genetic increase in inositol triphosphate (IP3) which, in turn, leads to an
mutation in the MerTK receptor (vide infra) that mediates increase in intracellular free calcium concentration.199 Together,
binding to OS (vide infra), and, therefore, RCS RPE cells have these two initiate phagocytosis by influencing the reorganiza-
defective OS phagocytosis. Rod OS are maintained at a fixed tion of cytoskeletal filaments. Increased calcium also results in
length,178 and there is a need to create new discs/opsin in photo- activation of protein kinase C that shuts off phagocytosis.200
receptors to replace damaged OS due to light exposure.179,180 Cyclic AMP (cAMP) modulates the rate of phagocytosis:
The constant length is achieved by RPE phagocytosis of OS.179 increased cAMP results in decreased phagocytosis.201,202
Phagocytosis also allows for recycling of retinoids and fatty Following phagocytosis, the phagosomes are guided to the
acids present in photoreceptor OS (discussed earlier). basal region by microtubules203 and fuse with lysosomes in the
The nature of structural interface between RPE and photo- basal region of the cell. Initially, small lysosomes fuse with
receptor OS is different in rods and cones. Rod OS (ROS) con- phagosomes. Subsequently, large lysosomes fuse with the
tact the apical surface of RPE while cone OS (COS) do not reach phagosomes via pore-like structures through which lysosomal
the surface. In the case of COS, RPE cells extend one or more contents may enter the phagosome.204 Cathepsin D is the major
processes of 10–20 mm length to reach the tip. Each process lysosomal enzyme involved in degradation of OS with possible
expands near the COS tip to ensheath it. These processes involvement of cathepsin S.205–207 The degraded products are
surround the COS to form a supracone space. COS discs are either reused or eliminated. The rapid and transient increase in
shed into this space and guided to RPE.181,182 Since the COS is gene and protein expression that follows OS phagocytosis does
shorter than ROS and has a narrow tip, the resulting phago- not occur following nonspecific phagocytosis of latex beads in
somes are smaller, and there are fewer phagosomes in the RPE vitro. These genes include c-fos, zif-268, tis-1, and peroxisome
that contact cones vs. rods. proliferator-activated receptor g that is a regulator of lipid
Phagocytosis can be divided into the following stages: metabolism.208,209
shedding of OS, binding of OS to RPE, internalization, migra- Molecules that have been implicated in regulation of phago-
tion to the basal cytoplasm, and digestion.183 Photoreceptors cytosis include b-adrenergic agonists, adenosine A2 receptors,210
1642 shed their OS cyclically, based on the circadian rhythm, in a serotonin, bFGF, acetylcholine, glutamate dopamine, and
Müller Cells and the Retinal Pigment Epithelium

melatonin.183 The latter two are responsible for the circadian eliminated by light exposure, lipofuscin accumulation is not
regulation of shedding and phagocytosis.211 Melatonin is a significant.236 Thus, phagocytosed photoreceptors probably are
hormone secreted rhythmically by the pineal gland and also by a major source of lipofuscin in RPE. RPE cells phagocytose
photoreceptors at night. It activates light-induced OS shedding 6000–8000 discs every day, and the digestion is not
in dark. During daytime, dopamine synthesis results in complete.183 The residual structures are fluorescent and can be
inhibition of melatonin and inhibition of OS shedding. RPE seen as early as 16 months after birth.237 Autophagy also con-
cells express receptors for both dopamine and melatonin so that tributes to the debris.238–240 Since many RPE cells persist through
shedding and phagocytosis are synchronized. Recently, the the life span, this debris can occupy up to 70% of the cyto-
role of avb5 integrin in synchronized phagocytosis has been plasmic volume, which leads to decreased RPE phagocytic
demonstrated.212 activity.241 Lipofuscin deposition also depends on dietary
vitamin A.242
Secretion The natural autofluorescence of the fundus (between 500 and
RPE cells secrete numerous growth factors.213 These include 750 nm with maximum emission at ~590–630 nm) is attri-
PDGF, PEDF, FGF, TGF, CNTF, VEGF, insulin-like growth buted to RPE lipofuscin. The autofluorescence increases with
factor (IGF), lens-epithelium derived growth factor, cytokines age and declines after age 70.243 The latter could be either due
(interleukins), and tissue inhibitor of metalloproteinases (TIMP). to loss of RPE or due to decreased fluorescence of A2E as a
Some of these are secreted by cultured RPE while others like result of photo-oxidation.244
VEGF, TGF-b, IGF, and FGF are secreted in vivo as well. A prominent component of lipofuscin is A2E (C42H58NO,

CHAPTER 125
PEDF is a 50 kD protein present in RPE and secreted into molecular weight 592): vitamin A aldehyde (generated upon
the adjacent IPM.214 PEDF is neurotrophic, prevents cell photoisomerization of 11-cis-retinal) + ethanolamine in 2:1 ratio.
proliferation, and promotes differentiation in retina and other A2E is often named as N-retinylidene-N-retinylethanolamine,
tissues.215 PEDF has neuroprotective effects against ischemic but the name does not reflect the pyridinium ring present in the
damage,216 glutamate excitotoxicity, oxidative stress, and retinal structure.245 Sparrow hypothesizes that since A2E structure is
degeneration. It has antiangiogenic effects, stabilizing the unprecedented, it may not be recognized by RPE lysosomal
choriocapillaris endothelium.217,218 VEGF219 is secreted in low enzymes.245 A2E formation starts with condensation of
concentrations in healthy eyes and prevents apoptosis of phosphatidylethanolamine (from the photoreceptor membrane)
choriocapillaris endothelium. PEDF is secreted apically from and light-generated 11-cis-retinal to form N-retinylidene-
RPE cells and VEGF basally.220,221 PEDF also influences glial cell phosphatidylethanolamine (NRPE) (NRPE is the substrate for
maturation. RPE removal results in failure of Müller cells to ABCA4; vide infra), followed by a series of intermediate
form adherens junctions with photoreceptors.222 products, including A2PE, the immediate precursor of
TGF-b was initially demonstrated in eyes with proliferative A2E.246,247 A2E is formed by cleavage of A2PE in RPE.247 The
vitreoretinopathy (PVR). Vitreous samples from these eyes con- cyclical phagocytosis of OS prevents accumulation of A2PE in
tained three times more TGF-b than eyes with uncomplicated the photoreceptor OS. A2PE is the fluorescent pigment in
retinal detachment. RPE cells can synthesize and secrete TGF-b photoreceptor OS. Thus, accumulation of A2PE due to defects
normally. RPE cells display increased production of predomi- in phagocytosis contributes to the fluorescence in photo-
nantly TGF-b2 in response to photocoagulation,223 decreased receptors (e.g., RCS rats, Stargardt disease). A2E undergoes
oxygen tension,224 PVR,225 age-related macular degeneration,226 further photoisomerization to generate iso-A2E and other cis-
myopia, and sickle cell retinopathy.227 In PVR, RPE cells are a isomers that are not as abundant as A2E.248
major component of the proliferating cells. In addition, the One hypothesis for age-related accumulation of lipofuscin
TGF-b produced by RPE increases the fibrotic response.228 was age-related decline in activity of lysosomal enzymes like
PDGF is chemotactic and mitogenic to RPE and glial cells.229 cathepsins.249 However, lipofuscin accumulation itself appears
PDGF receptors are also expressed by RPE.230 to alter lysosomal degradation.231 What makes lipofuscin
Growth factors like FGF and CNTF play a neurotrophic role resistant to degradation? It is believed that oxidative damage to
in maintenance of photoreceptors.68 cellular components results in cross-linking of proteins and
other biomolecules,250 rendering them indigestible. Thus, cells
Lipofuscin Accumulation and other Aging exposed to oxidative stress tend to accumulate lipofuscin.
Changes Accumulation of lipofuscin in the cell affects cell function and
Lipofuscin (‘age pigment’) is an intralysosomal complex of health. Lipofuscin-containing cells are more susceptible to
protein, lipid, carbohydrate, metals, and other compounds that oxidative damage.251 Lipofuscin is a photoinducible generator of
accumulates in postmitotic, metabolically active cells.231 The oxygen radicals, e.g., superoxide anion, singlet oxygen, and
amount accumulated in these cells has a linear correlation with hydrogen peroxide.252 These free radical species, in turn, react
age, hence the label, age pigment, and has a negative correlation with biomolecules to cause lipid peroxidation, protein
with longevity. Accumulation in RPE seems to be inversely oxidation, loss of lysosomal integrity, and RPE cell death.253,254
related to melanin content.232 Lipofuscin content of RPE cells A2E is one of the photosensitizers in lipofuscin, and there may
depends on the location in the eye. Cells in the posterior pole be other as yet unidentified molecules in lipofuscin that are also
have a higher amount of lipofuscin with subfoveal RPE showing photosensitizers. Even though lipofuscin is compartmentalized,
a lower accumulation relative to cells in the subparafoveal there is increased production of reactive oxygen species within
region.233 Lipofuscin is nondegradable and cannot be eliminated the lipofuscin-containing lysosomes due to the presence of iron
from the cell via exocytosis.231 Formation and accumulation of that reacts with H2O2.255 Lipofuscin accumulation results in
lipofuscin may occur by autophagy of cytoplasmic organelles decreased phagocytosis by RPE, including that of photoreceptor
and contents and subsequent fusion with lysosomes or by OS.241 Proteins in lipofuscin granules are modified by
phagocytosis of material from a different cell (heterophagy). peroxidation; the presence of mitochondrial proteins indicate
Phagocytosed photoreceptor OS are a major source of autophagy and a possible role in lipofuscin formation.256
lipofuscin; if RPE that are unable to phagocytose photoreceptor Nonenzymatic oxidative modification of amino acids may
OS or have mutations in RPE 65 (and therefore lack 11-cis- result in inhibition of proteolysis, and incomplete proteolysis of
retinal and vitamin A aldehyde), they do not accumulate proteins may promote lipofuscin formation.257 The presence
lipofuscin as much as native RPE.234,235 If photoreceptors are of A2E in RPE results in delayed degradation of phospholipids 1643
RETINA AND VITREOUS

Retina B-wave

600mV
(apical)
2K 2Cl K Na nHCONa Na, K ERG
1
A-wave
100 msec
Vac = -50mV C-wave
B-wave

1 mV
C-wave

3Na
K 3Na
K

10 seconds
TEP Light peak
5mV

4 mV
Light C-wave
response “2nd C”

HCO2
K Cl HCO2
Cl
K

“Fast oscillation”
10 minutes
Light peak

5 mV
Vta = -45mV Slow C
Oscillations
oscillation
SECTION 10

Cl, HCO2 “Fast oscillation”


Cl
Choroid 2 hours
(basal)
FIGURE 125.7. Electrical changes recorded from a corneal electrode
FIGURE 125.6. Ion channels in RPE cells.
in a dark-adapted eye after exposure to a bright light. The response
Redrawn from Marmor MF, Wolfensberger TJ: The retinal pigment epithelium.
has been depicted in four different time scales from top to bottom;
New York: Oxford University Press; 1998.
a- and b-waves occur relatively rapidly and represent neural retinal
responses. The light-induced drop in subretinal K+ concentration
causes hyperpolarization of the RPE apical membrane and the distal
ends of Müller cells resulting in the c-wave. The hyperpolarization of
as well.258 Melanosomes are photoprotective, but with age the RPE basal membrane is recorded as a fast oscillation. The
melanosomes become photoreactive. (This change could be due depolarization of the RPE basolateral membrane by increased
to association of A2E with melanosomes although contamina- conductance of Cl– results in light peak generation.
tion of the melanosome preparation with A2E in the experi- Redrawn from Marmor MF, Wolfensberger TJ: The retinal pigment epithelium.
ments cannot be ruled out.) Aggregates of A2E have a New York: Oxford University Press; 1998.
detergent-like property of compromising membrane integrity
leading to membrane blebbing.259
low, the direction is reversed so that HCO3– moves through RPE
Ion Transport from choroid into the subretinal space. There is a net secretion
RPE cells express ion channels on their apical and basolateral (i.e., movement of ion across the RPE into the subretinal space)
surfaces that mediate transport of Na+, K+, HCO3–, Cl–, etc. of Ca+, with movement out of the apical membrane via a
One of the consequences of RPE cell fluid transport is Na+/Ca2+ exchanger and Ca2+–ATPase. K+ enters RPE on the
dehydration of the subretinal space and maintenance of neural apical side via a Na+K+–ATPase and a Na+K+–2Cl co-
retina–RPE apposition (vide infra). Fluid movement is linked to transporter and leaves via apical or basal K+ channels. There is
the ion movement. In RPE, H+/lactate, and Cl⫺ movement are a net transepithelial transport of K+ from the subretinal space
the main ions that are linked to fluid movement.260,261 The ion to choroid. Movement of Cl– and K+ drives the movement
differences between the cytoplasm and the surrounding tissues of water.
result in a potential difference across the RPE cell monolayer. The ion channels maintain the ion homeostasis in the sub-
This transepithelial potential (TEP) is ~5–15 mV.262 retinal space, especially the changes that occur in ion concen-
Ion channels present in RPE cells are summarized in Figure trations as a result of light stimulation of photoreceptors. In the
125.6. Sodium is secreted actively into the subretinal space by dark, the apical RPE membrane is more hyperpolarized than the
a Na+/K+ ATPase pump present in the apical surface.263,264 This basal membrane. Voltage across the apical membrane in a
pump creates a sodium gradient from the subretinal space to resting RPE cell is ~ –50mV and across the basal membrane is
the RPE cytoplasm that facilitates as well as provides energy –45mV. Thus, a transepithelial potential, TEP (Vba–Vap= 5mV),
for the transepithelial transport. HCO3–, K+, and Cl– are co- exists across the RPE.262 Membrane potential across the apical
transported with Na+ (vide infra). The mechanism of Na+ entry membrane is primarily due to K+ channels with additional
from the basal plasma membrane is not known. Cl– is absorbed contributions from the Na+/K+ pump and a Na+/HCO3–
actively by apical Na+K+–2Cl– co-transport present in the apical cotransporter (Fig. 125.6). K+ and Cl– channels in the basal
membrane,265,266 Ca+-dependent Cl– channels,267,268 ClC-2 membrane contribute to the membrane potential. The
channels (a family of chloride conducting, voltage-gated difference in membrane potential between the apical and basal
channels),269 and cystic fibrosis transmembrane conductance membrane is due to the large Cl– conductance. This difference
regulator (CFTR)270 present in the basolateral membrane. manifests as the standing potential in a dark-adapted eye when
Na+/HCO3– enters the cell via an apical Na+/HCO3– an electrode is placed on the cornea and amplified, with the
cotransporter271,272 and leaves the RPE through a basal cornea positive.
Cl–/HCO3– exchanger and possibly a Na+/HCO3– co-transporter. Exposure to light induces a series of changes that are recorded
The transport of HCO3– regulates the pH of subretinal space in the electroretinogram (ERG) and electrooculogram (EOG).
and the intracellular pH of RPE cells. If the pH is high in the The ERG response begins with a rapid dip in voltage (a-wave)
subretinal space or in RPE, HCO3– is moved out of RPE into the followed by a rapid upward deflection (b-wave) that does not
choroid via a NaHCO3 co-transporter in the apical membranes quite return to the baseline (Fig. 125.7); a- and b-waves reflect
1644 and Cl–/HCO3– exchanger in the basal membranes. When pH is electrical activity in the neural retina.273 These waves are
Müller Cells and the Retinal Pigment Epithelium

followed by a slow rising c-wave for which the RPE is Fluid Transport
responsible.274 In the dark, Na+ flows into the photoreceptors About 10–20% of aqueous fluid is absorbed posteriorly with the
via the cGMP-gated Na+ channels in the OS and is pumped out rate limiting structure being the neural retina. The RPE also
by the Na+/K+ pump in the inner segment. There is a passive limits passive fluid movement across Bruch’s membrane, but it
efflux of K+ out of the inner segment. In response to light, has an active pumping mechanism that has a large capacity and
cGMP-gated channels close, and the OS is hyperpolarized. helps to keep the subretinal space dehydrated. The fluid
Additionally, the passive efflux of K+ is also shut down. permeability across dog retina is 0.03 m1 min–1 mmHg–1 sqcm–1
Decreased concentration of K+ in the subretinal space and the and across dog RPE is 0.01 ml min–1 mmHg–1 sqcm–1.
large conductance of K+ at the RPE apical membrane results in Permeability across monkey RPE is 0.005 m1min–1 mmHg–1
hyperpolarization of the latter.262 Additionally, the distal ends of sqcm–1.288,289 The transretinal pressure of 0.5 µ 10–3 mmHg172
Müller cells hyperpolarize as well, resulting in a slow negative that exists across the retina is sufficient to keep retina attached
transretinal potential termed slow PIII.275 Since the RPE but has no effect on the fluid movement across it. In the
contribution is larger, a positive deflection is recorded on the monkey eye, a pressure drop of ~4 mmHg exists between the
surface of the cornea. In summary, both RPE and Müller cells vitreous and choroid, which implies a possible fluid flow across
contribute to c-wave. The former results in a positive deflection, the RPE of 0.3 m1/min.290
and the latter results in a negative deflection. Since RPE The forces driving the fluid movement across the retina in
contribution is greater than Müller cell’s, the net change is a the eye include intraocular pressure, osmotic/oncotic pressure,
positive c-wave in the EOG. The c-wave is followed by fast and the active solute-linked fluid transport by the RPE mono-

CHAPTER 125
oscillation, which is a negative slow potential fall toward or layer. A change in intraocular pressure from 0 to 38 mmHg
below the dark-adapted baseline. This change is produced by leads to a 39% increase in rate of fluid absorption from sub-
the hyperpolarization of the RPE basal membrane as a retinal space in rabbits.291 Because of the higher protein content
consequence of the above-mentioned fall in subretinal K+.276 in the choroid and because osmotic pressure in vitreous and
The decrease in K+ slows the activity of Na+/K+/2Cl– activity subretinal space is maintained at somewhat similar levels, there
causing a decrease in Cl– influx across the apical membrane of is a net osmotic pressure that drives fluid outwards only.292,293
RPE. The resultant decrease in intracellular Cl– concentration In a normal eye, active transport by RPE cells is the major force
produces a change in Cl– equilibrium across the basolateral Cl– eliminating water from the subretinal space; intraocular
channels leading to a hyperpolarization of basal membrane. pressure and osmotic pressure contribution to fluid transport is
The latter is termed fast because it is fast relative to the next minor but effective when RPE cells are damaged. Müller cells
response, the light peak. The light peak is the slowest and may play a role in water transport in the inner retina.25 RPE is
largest component of the EOG and refers to the slow increase in estimated to transport fluid at a rate of 0.1 m1 h–1 sqmm–1.294
potential that follows the fast oscillation. It is generated by an The ion channels responsible for solute-linked fluid movement
increased conductance of Cl– in the basolateral membrane, are the apical Na+,K+,2Cl– channel, basal Cl– channels, and the
depolarizing it, and increasing the transepithelial potential.262 H+/lactate co-transporter.260,261 Recently aquaporin that forms
“water channels” in secretory and absorptive epithelia,295 have
Transport of other molecules been shown to play a significant role in water transport across
The RPE also eliminates waste produced by photoreceptors. RPE cells.296
Lactic acid is an excellent example. Photoreceptor OS produce
lactic acid, which is removed by RPE via a lactate–H+ co- Melanin
transporter, MCT1, in the apical membrane, and MCT3, in the Melanin granules are ovoid in shape and measure ~2–3 mm in
basal membrane.277 length, 1 mm in diameter, and are distributed near the apical
Glucose is transported from blood to photoreceptors by GLUT region of the cell. Melanin granules contain polymerized
1 and GLUT3. The latter transports glucose routinely, and the melanin in a protein matrix. The melanin can be pheomelanin
former functions when there is an increased metabolic or eumelanin, imparting yellowish/reddish or brown/black color,
demand.278,279 Vitamin A is taken up on the basal surface by respectively. The human eye contains eumelanin.297 While
receptor binding to vitamin A bound to retinol-binding protein/ uveal melanin-containing cells are derived from melanoblasts
transthyretin complex and enters the visual cycle.280,281 RPE originating from neural crest, RPE, ciliary epithelium, and iris
also transports docosahexaenoic acid synthesized in the liver pigment epithelium develop from the neurectoderm.297,298
and circulating in the blood.282 Docosahexaenoic acid, which is Melanin is produced during development. However, it is
enriched in photoreceptor membranes, is not synthesized in possible that melanin is synthesized in adult cells as well.299,300
neuronal tissue.283 Tyrosinase synthesis can be induced in adult cultured RPE by
Mutations in the bestrophin molecule (product of VMD2 feeding them photoreceptor OS.301
gene), which functions as Ca+-sensitive Cl– channel, are Melanin synthesis begins with the production of pre-
associated with three diseases: Best vitelliform macular melanosomes and synthesis of tyrosinase, the two processes
dystrophy,284 adult-onset vitelliform dystrophy, and autosomal being independent of one another (Fig. 125.8).302,303 Preme-
dominant vitreoretinalchoroidopathy.285 In the former two con- lanosomes are vesicular bodies that contain the structural
ditions, appearance of an egg yolk-like yellow lesion in the fun- proteins of melanin but no tyrosinase enzyme. They are
dus is characteristic and results from accumulation of deposits, assembled in smooth endoplasmic reticulum and released into
including lipofuscin, in the subretinal space and sub-RPE region the cytoplasm. Tyrosinase is synthesized in rough endoplasmic
as well as RPE hypertrophy. Vitreoretinalchoroidopathy is cha- reticulum and transported through the Golgi apparatus during
racterized by a circumferential zone of hyperpigmentation which the enzyme undergoes posttranslational modification,
anterior to the equator along with punctate white pre-retinal predominantly glycosylation. The enzyme is then assembled
opacities, fibrillar condensation of vitreous, and breakdown of into coated vesicles that bud off and fuse with the
the blood–retinal barrier.286 premelanosomes to form melanosomes.
Patients with cystic fibrosis have defective Cl– channels in RPE Tyrosinase catalyzes the first two steps of melanin
cells as well.270,287 However, retinal degeneration is not seen, synthesis: hydroxylation of the amino acid, L-tyrosine, to
probably because of the presence of other channels that 3,4–dihydroxyphenylalanine (DOPA) and oxidation of DOPA
compensate. to dopaquinone. Spontaneous endocyclic ring formation 1645
RETINA AND VITREOUS

RPE cells exhibit regional variation in melanin content. Cells


L-tyrosine
in the periphery have higher melanin content, and the amount
decreases posteriorly.304,305 With age, melanin content decreases
Tyrosinase
in the periphery while there is no change under the macula.
DOPA There is no difference in RPE melanin content in whites and
blacks.305
Tyrosinase Melanin absorbs light passing through the retina, preventing
reflection of light within the fundus that might affect the clarity
Dopaquinone of the image formed by the photoreceptors and protecting the
Tyrosinase photoreceptors from excess light.306 Absorption of excess light
raises the possibility of an RPE photoprotective effect. However,
there are conflicting data. In vitro studies show that intra-
Leucodopachrome cellular melanin clearly protects the cell from light-induced
DNA damage.307 Some in vivo studies show that RPE
pigmentation protects the overlying photoreceptors from light-
induced damage in rodents.308,309 In another study using
Dopachrome chimeric mice expressing pigmented and nonpigmented RPE
cells, light damage to photoreceptors was more severe in the
SECTION 10

central retina than in the periphery, regardless of the pigmen-


tation of the underlying RPE.310
Even though synthetic melanin has been shown to have
strong free-radical scavenging properties, natural melanin is not
DHICA DHI very effective.306,311 Thus, it is unlikely that it plays as signifi-
cant a role in RPE cells as other antioxidants, e.g., superoxide
dismutase.306 Melanin is also capable of generating free-radicals
and was thought to be responsible for damage caused by light of
Melanin (polymers of DHICA and DHI) shorter wavelength.312,313 However, this blue light damage is
now attributed to the presence of liposfuscin.314
FIGURE 125.8. Melanin synthesis begins with hydroxylation of Melanin traps radiation and dissipates energy, which can
L-tyrosine to 3,4-dihydroxyphenylalanine (DOPA) followed by oxidation result in tissue damage if there is acute exposure to high
of DOPA to dopaquinone. Both reactions are catalyzed by tyrosinase. intensity radiation as occurs in laser photocoagulation.315
Spontaneous endocyclic ring formation results in formation of Hansen and Fine have demonstrated in melanin models that a
leucodopachrome from dopaquinone. Leucodopachrome undergoes temperature increase above 10°C will cause a zone of
oxidation to form dopachrome and DOPA. This oxidation is coupled denaturation in the vicinity of the granule that would spread
with re-oxidation of DOPA to dopaquinone by tyrosinase. Dopachrome
with the conduction of heat from the granule, or, if the energy
is converted to 5,6 dihydroxyindole-2-carboxylic acid (DHICA). A small
portion of dopachrome undergoes spontaneous conversion to 5,6 is great enough, generation of steam at the granule cytoplasm
dihydroxyindole (DHI). Oxidative polymerization of DHICA and DHI interface. The heat and the steam lead to coagulative damage as
results in melanin formation. well as disruption of the structures.316
Boulton M: Melanin and the RPE. In: Marmor MF, Wolfensberger TJ, eds. The Melanin can bind to numerous chemicals, thus becoming an
retinal pigment epithelium. New York: Oxford University Press; 1998:68–85. effective sink or storage site responsible for toxicity.317,318
Chlorpromazine and chloroquine can bind irreversibly to melanin
and can lead to toxicity.319 Since melanins are polyanions, ionic
interaction is one mechanism by which they bind to chemicals.
results in formation of leucodopachrome from dopaquinone. The role of melanin in development is discussed in the
Leucodopachrome undergoes oxidation to form dopachrome embryology section. With increasing age, complex melanin-
and DOPA; this oxidation is coupled with re-oxidation of DOPA containing granules begin to appear. In addition to melanin,
to dopaquinone by the tyrosinase enzyme. Dopachrome is these contain a cortex of lipofuscin (melanolipofuscin) or a
converted to 5,6 dihydroxyindole-2–carboxylic acid (DHICA). A cortex of enzyme-reactive material (melanolysosomes).163 The
small portion of dopachrome undergoes spontaneous conver- amount of complex granules increases with age while pure
sion to 5,6 dihydroxyindole (DHI). Oxidative polymerization of melanin declines. The highest density of complex granules is in
DHICA and DHI results in eumelanin polymer. Pheomelanin is the submacular region and declines toward periphery and fovea.
formed by addition of cysteine to dopaquinone.297 In human eyes over 90 years old, there are no pure melanin
The different stages of melanin granule maturation can be granules.163 Pigment-free cytoplasm in the RPE also declines
seen by electron microscopy.149 Premelanosomes are with age.163 The fluorescence of the melanosomes shows a shift
membrane-bound, ovoid, nonpigmented organelles containing toward red with increasing age.300
a protein matrix. Stage I granules have protein filaments pole to
pole, in an ordered arrangement; melanin synthesis becomes Role in Visual Cycle
evident at this stage. Increasing melanin synthesis become Transduction of a light signal to vision begins with absorption
evident in stages II and III, and by stage IV, a fully melanized of light by rhodopsin (or cone opsin). Rhodopsin (or cone opsin)
melanosome, i.e., melanin granule, is formed. is formed by binding opsin, a G-protein coupled protein that
RPE cells are the first to synthesize melanin in the eye.149 By is present within the membranes of the photoreceptor discs,
7-weeks gestation, immature melanosomes are visible, and in with the visual chromophore, 11-cis-retinal, in the intradiscal
the subsequent few weeks (~7 or more weeks), continued space (Fig. 125.9). Absorption of light results in irreversible
melanin synthesis is evident by the appearance of melanosomes isomerization of 11-cis-retinal to all-trans-retinal, which leads
of various stages. Production of new melanosomes then stops, to a conformational change in the rhodopsin molecule, thus
but the polymerization of melanin within the granules con- activating it.320,321 Activated rhodopsin initiates the photo-
1646 tinues for up to two years. transduction cascade. Rhodopsin is phosphorylated and
Müller Cells and the Retinal Pigment Epithelium

FIGURE 125.9. Schematic of the rod visual


The Rod Visual Cycle cycle. See text for details. ABCR: ATP-binding
cassette, retina; CRBP: cellular retinol-binding
CRALBP CRALBP CRBP
protein; CRALBP: cellular retinaldehyde-binding
11-Ral 11-Rol t-Rol
protein; IMH: isomerohydrolase; IRBP:
11-RDH LRAT
interphotoreceptor retinoid-binding protein;
11-Ral 11-Rol t-RE t-Rol LRAT: lecithin:retinol acyltransferase; RDH: all-
IMH
trans-retinol dehydrogenase; 11-RDH: 11-cis-
retinol dehydrogenase; Rho: rhodopsin; Rho*:
Retinal Pigment Epithelium
activated rhodopsin; 11-Ral: 11-cis-retinal;
11-Rol: 11-cis-retinol; at-Ral: all-trans-retinal;
11-Ral IRBP ? Subretinal Space ? IRBP t-Rol at-Rol: all-trans-retinol; at-RE: all-trans-retinyl
ester. Reproduced with permission from Saari,
J C: Biochemistry of visual pigment
Rod Outer Segment regeneration: the Friedenwald lecture. Invest
hv Ophthalmol Vis Sci, 2000; 41(2):337–48
t-Ral NADPH NADP (Association for Research in Vision and
Rho Rho* t-Rol Ophthalmology).
11-Ral t-Ral ABCR RDH

CHAPTER 125
t-Ral

Opsin

inactivated by rhodopsin kinase and subsequent binding to production of 11-cis-retinal in the dark. The switch between the
arrestin. Inactivated rhodopsin releases all-trans-retinal and membrane and soluble forms of RPE-65 is controlled by LRAT
binds to 11-cis-retinal so that it can become activated again by and by the concentration of 11-cis-retinol in RPE.328 11-cis-
exposure to light. All-trans-retinal generated in the photo- retinol is either esterified by LRAT and stored in the cell or
receptor disk space cannot be converted back to 11-cis-retinal in oxidized to 11-cis-retinal, a reaction catalyzed by 11-cis-retinol
the photoreceptor. This conversion occurs in RPE cells and dehydrogenase or RDH5.329 Cellular retinaldehyde-binding
possibly in Müller cells. Transport of all-trans-retinal to RPE, protein (CRALBP) accelerates the latter enzymatic reaction:330
however, is not a matter of simple diffusion. All-trans-retinal in 11-cis-retinal is transported back to photoreceptors by
the disk space is transported to the extra-discal space by an IRBP.324,325
ATP-binding cassette protein (ABCR), known as ABCA4.322,323 An alternate pathway for regeneration of 11-cis-retinal has
This is a flippase for NRPE, flipping the molecule across the been described.331 This pathway is the reverse of the light-
disk membrane. All-trans-retinal is converted to NRPE by induced reaction occurring in the photoreceptors, i.e., all-trans-
reacting with phosphatidylethanolamine. Once in the retinal is converted to 11-cis-retinal by RPE retinal G protein
photoreceptor extra-discal cytoplasm, all-trans-retinal is coupled receptor (RGR) using light energy. The source of all-
released. It is then reduced to all-trans-retinol (vitamin A) by a trans-retinal is the RPE pool of all-trans-retinol acquired from
membrane-bound retinol dehdyrogenase (RDH) in the the photoreceptors. The conversion is mediated by a RDH that
photoreceptor extra-discal cytoplasm. All-trans-retinol is then is presumed to be different from RDH5. This alternate pathway
transported to RPE cells from the rods or to Müller cells from maintains a constant level of 11-cis-retinal despite changes in
cones by interphotoreceptor-binding protein (IRBP) present in ambient light, i.e., maintains the levels of 11-cis-retinal after
the interphotoreceptor space.324,325 onset of light. The main pathway is important for the acute
All-trans-retinol transport within the RPE cell is not well changes in levels that occur during the process of vision.
characterized. Water-insoluble all-trans-retinol in the RPE is Therefore, loss of RDH5 results in delayed recovery from light
bound to cellular retinol-binding protein (CRBP), thus trapping exposure.
it within the RPE cell as well as protecting it from oxidation or Why does a two-cell system involving Müller and RPE cells
isomerization. CRBP-bound all-trans-retinol is carried to the exist for 11-cis-retinal renewal? It is speculated that such a
endoplasmic reticulum where it undergoes esterification to system separates molecules in different compartments so that
long-chain fatty acids derived from membrane phospholipids. thermodynamically unfavorable reactions can proceed by mass
This reaction, catalyzed by lecithin:retinol acyl transferase action.332 An alternative explanation could be that there may be
(LRAT), leads to formation of all-trans-retinyl esters. Hydrolysis no room for all the molecules required due to the high packing
of retinyl esters coupled with isomerization results in formation density of visual pigments in the OS.332
of 11-cis-retinol.326 The coupling of hydrolysis is important Mutations in the molecules involved in visual cycle result
because isomerization requires energy and is provided by the in retinal degenerations. Total loss of ABCA4 function causes
energy generated during hydrolysis. An additional protein retinitis pigmentosa.333 Partial loss results in Stargardt disease
involved in the isomerization step is RPE-65, which acts as a or fundus flavimaculatus.323 Loss of ABCA4 may also play a role
chaperone for all-trans retinyl esters.327 RPE-65 exists in two in age-related macular degeneration.334–336 Loss of CRALBP causes
forms: a membrane bound form that is triply palmitoylated and retinitis punctata albescens and autosomal recessive retinitis
a soluble form that is not. The former has high affinity to pigmentosa due to decreased efficiency of regeneration of visual
all-trans retinyl esters and donates a palmitoyl group for the pigment.337,338 Loss of RDH5 causes fundus albipunctatus,339
reaction mediated by LRAT. This ensures 11-cis-retinal production and loss of RGR leads to retinitis pigmentosa.340,341 Mutations
during light. The soluble form of RPE-65 reduces the in RPE-65 can cause Leber congenital amaurosis and autosomal 1647
RETINA AND VITREOUS

recessive childhood-onset severe retinal dystrophy.342,343 Finally, surface of fetal RPE cells.358,359 They do not seem to express
loss of function mutations in LRAT leads to early onset retinal MHC class II antigens normally but do so if exposed to IFN-
dystrophy.344 g.360 Culturing RPE cells also can induce MHC antigen
An opsin that is localized exclusively to RPE has been expression.360 RPE cells can process antigen prior to antigen
identified. It is present in the microvilli of the RPE. The exact presentation, thereby potentially acting as antigen-presenting
role has not been elucidated.345 cells.361 Higher levels of IFN-g-induced MHC class II antigen
expression on RPE cells leads to increased activation of antigen-
Immune Functions specific T-cells as manifested by the production of pro-
The subretinal space is an immune-privileged site and can inflammatory TNF-a.362 This activation does not include T-cell
accept allografts for prolonged periods compared to a proliferation or production of IL-2, however, as is the norm for
nonimmune-privileged site (e.g., the subconjunctival space).346 T-cell activation.
Immune-privileged tissue can survive for prolonged periods in
nonimmune-privileged sites compared to nonimmune-
privileged tissue. RPE is an immune-privileged tissue. Neonatal FUTURE DEVELOPMENTS
RPE sheets, when placed in heterotopic sites (i.e., anatomic Due to the critical functions performed by RPE cells, perturba-
locations in which the transplanted tissue is not found tions in cell function can have serious consequences that affect
normally) do not undergo rejection when compared to vision. Their central role in visual physiology is demonstrated
nonprivileged tissue.347 Constitutive expression of Fas ligand on by the numerous visually debilitating conditions that are caused
SECTION 10

the RPE may contribute to its status as immune-privileged by mutations in RPE. RPE cells already are the target of thera-
tissue.347,348 RPE contributes to the immune-privilege of peutic interventions for conditions such as Leber congenital
subretinal space by maintaining a blood–retinal barrier; RPE amaurosis.363 The ease of RPE isolation, the straightforward
cells create a local immune suppressive microenvironment surgical access to the subretinal space, and the facile inter-
through the production of immuno-modulatory factors such as digitation of transplanted RPE cells with host photoreceptors
TGF-b,349–352 through suppression of T-cell activation,353,354 and renders the RPE a logical choice for initial attempts at cell-based
through the induction of activated-T cell apoptosis.355–357 therapy in the retina.364 Newer experimental techniques like
RPE cells express transplantation antigens. Major and minor gene chip array and protein chips will continue to add to our
histocompatibility (MHC) class I antigens are present on the knowledge of structure and function of RPE.

REFERENCES
1. Müller H: Zur histologie der netzhaut. 13. Wolburg H, Berg K: Distribution of 23. Kofuji P, Biedermann B, Siddharthan V,
Z Wiss Zool 1851; 3:234–237. orthogonal arrays of particles in the Muller et al: Kir potassium channel subunit
2. Chow RL, Lang RA: Early eye development cell membrane of the mouse retina. Glia expression in retinal glial cells: implications
in vertebrates. Annu Rev Cell Dev Biol 1988; 1:246–252. for spatial potassium buffering. Glia 2002;
2001; 17:255–296. 14. Sarthy V, Ripps H: The Retinal Müller cell. 39:292–303.
3. Hollenberg MJ, Spira AW: Early New York: Plenum Publishers; 2001. 24. Ishii M, Fujita A, Iwai K, et al: Differential
development of the human retina. Can J 15. Yamada E: Some structural features of the expression and distribution of Kir5.1 and
Ophthalmol 1972; 7:472–491. fovea centralis in the human retina. Arch Kir4.1 inwardly rectifying K+ channels in
4. Sharma RK, Johnson DA: Molecular signals Ophthalmol 1969; 82:151–159. retina. Am J Physiol Cell Physiol 2003;
for development of neuronal circuitry in the 16. Gass JD: Muller cell cone, an overlooked 285:C260–C267.
retina. Neurochem Res 2000; 25:1257–1263. part of the anatomy of the fovea centralis: 25. Nagelhus EA, Horio Y, Inanobe A, et al:
5. Reichenbach A, Stolzenburg JU, hypotheses concerning its role in the Immunogold evidence suggests that
Eberhardt W, et al: What do retinal muller pathogenesis of macular hole and coupling of K+ siphoning and water
(glial) cells do for their neuronal ‘small foveomacualr retinoschisis. Arch transport in rat retinal Muller cells is
siblings’? J Chem Neuroanat 1993; Ophthalmol 1999; 117:821–823. mediated by a coenrichment of Kir4.1 and
6:201–213. 17. Grayson C, Reid SN, Ellis JA, et al: AQP4 in specific membrane domains. Glia
6. Walcott JC, Provis JM: Muller cells express Retinoschisin, the X-linked retinoschisis 1999; 26:47–54.
the neuronal progenitor cell marker nestin protein, is a secreted photoreceptor protein, 26. Skatchkov SN, Eaton MJ, Shuba YM, et al:
in both differentiated and undifferentiated and is expressed and released by Weri-Rb1 Tandem-pore domain potassium channels
human foetal retina. Clin Experiment cells. Hum Mol Genet 2000; 9:1873–1879. are functionally expressed in retinal (Muller)
Ophthalmol 2003; 31:246–249. 18. Molday LL, Hicks D, Sauer CG, et al: glial cells. Glia 2006; 53:266–276.
7. Kljavin IJ, Reh TA: Muller cells are a Expression of X-linked retinoschisis protein 27. Bok D, Schibler MJ, Pushkin A, et al:
preferred substrate for in vitro neurite RS1 in photoreceptor and bipolar cells. Immunolocalization of electrogenic sodium-
extension by rod photoreceptor cells. Invest Ophthalmol Vis Sci 2001; bicarbonate cotransporters pNBC1 and
J Neurosci 1991; 11:2985–2994. 42:816–825. kNBC1 in the rat eye. Am J Physiol Renal
8. Willbold E, Layer PG: Muller glia cells and 19. Sauer CG, Gehrig A, Warneke-Wittstock R, Physiol 2001; 281:F920–F935.
their possible roles during retina et al: Positional cloning of the gene 28. Nagelhus EA, Mathiisen TM, Bateman AC,
differentiation in vivo and in vitro. Histol associated with X-linked juvenile et al: Carbonic anhydrase XIV is enriched in
Histopathol 1998; 13:531–552. retinoschisis. Nat Genet 1997; specific membrane domains of retinal
9. Rhodes RH: A light microscopic study of 17:164–170. pigment epithelium, Muller cells, and
the developing human neural retina. Am J 20. Minami Y, Ishiko S, Takai Y, et al: Retinal astrocytes. Proc Natl Acad Sci U S A 2005;
Anat 1979; 154:195–209. changes in juvenile X linked retinoschisis 102:8030–8035.
10. Rhodes RH: Ultrastructure of Muller cells in using three dimensional optical coherence 29. Tsacopoulos M: Metabolic signaling
the developing human retina. Graefes Arch tomography. Br J Ophthalmol 2005; between neurons and glial cells: a short
Clin Exp Ophthalmol 1984; 221:171–178. 89:1663–1664. review. J Physiol Paris 2002; 96:283–288.
11. Hogan MJ, Alvarado JA, Weddell JE: 21. Newman EA, Frambach DA, Odette LL: 30. Derouiche A: Possible role of the Muller cell
Histology of the human eye. Philadelphia: Control of extracellular potassium levels by in uptake and metabolism of glutamate in
W.B. Saunders; 1971. retinal glial cell K+ siphoning. Science the mammalian outer retina. Vision Res
12. Ramirez JM, Trivino A, Ramirez AI, et al: 1984; 225:1174–1175. 1996; 36:3875–3878.
Structural specializations of human 22. Kofuji P, Connors NC: Molecular substrates 31. Biedermann B, Bringmann A, Reichenbach
retinal glial cells. Vision Res 1996; of potassium spatial buffering in glial cells. A: High-affinity GABA uptake in retinal
1648 36:2029–2036. Mol Neurobiol 2003; 28:195–208. glial (Muller) cells of the guinea pig:
Müller Cells and the Retinal Pigment Epithelium

electrophysiological characterization, 49. Winter M, Eberhardt W, Scholz C, 65. Puro DG: Diabetes-induced dysfunction of
immunohistochemical localization, and Reichenbach A: Failure of potassium retinal Muller cells. Trans Am Ophthalmol
modeling of efficiency. Glia 2002; siphoning by Muller cells: a new hypothesis Soc 2002; 100:339–352.
39:217–228. of perfluorocarbon liquid-induced 66. Faktorovich EG, Steinberg RH, Yasumura D,
32. Winkler BS, Starnes CA, Sauer MW, et al: retinopathy. Invest Ophthalmol Vis Sci et al: Basic fibroblast growth factor and
Cultured retinal neuronal cells and Muller 2000; 41:256–261. local injury protect photoreceptors from
cells both show net production of lactate. 50. Reichenbach A, Faude F, Enzmann V, et al: light damage in the rat. J Neurosci 1992;
Neurochem Int 2004; 45:311–320. The Muller (glial) cell in normal and 12:3554–3567.
33. Mantych GJ, Hageman GS, Devaskar SU: diseased retina: a case for single-cell 67. LaVail MM, Unoki K, Yasumura D, et al:
Characterization of glucose transporter electrophysiology. Ophthalmic Res 1997; Multiple growth factors, cytokines, and
isoforms in the adult and developing 29:326–340. neurotrophins rescue photoreceptors from
human eye. Endocrinology 1993; 51. Francke M, Pannicke T, Biedermann B, the damaging effects of constant light.
133:600–607. et al: Loss of inwardly rectifying potassium Proc Natl Acad Sci U S A 1992;
34. Watanabe T, Mio Y, Hoshino FB, et al: currents by human retinal glial cells in 89:11249–11253.
GLUT2 expression in the rat retina: diseases of the eye. Glia 1997; 20:210–218. 68. LaVail MM, Yasumura D, Matthes MT, et al:
localization at the apical ends of Muller 52. Reichelt W, Pannicke T, Biedermann B, Protection of mouse photoreceptors by
cells. Brain Res 1994; 655:128–134. et al: Comparison between functional survival factors in retinal degenerations.
35. Mata NL, Radu RA, Clemmons RC, Travis characteristics of healthy and pathological Invest Ophthalmol Vis Sci 1998; 39:592–602.
GH: Isomerization and oxidation of vitamin human retinal Muller glial cells. Surv 69. Wahlin KJ, Campochiaro PA, Zack DJ,
a in cone-dominant retinas: a novel Ophthalmol 1997; 42 (Suppl 1):S105–S117. Adler R: Neurotrophic factors cause

CHAPTER 125
pathway for visual-pigment regeneration in 53. Francke M, Pannicke T, Biedermann B, activation of intracellular signaling
daylight. Neuron 2002; 36:69–80. et al: Sodium current amplitude increases pathways in Muller cells and other cells of
36. Mata NL, Ruiz A, Radu RA, et al: Chicken dramatically in human retinal glial cells the inner retina, but not photoreceptors.
retinas contain a retinoid isomerase activity during diseases of the eye. Eur J Neurosci Invest Ophthalmol Vis Sci 2000;
that catalyzes the direct conversion of all- 1996; 8:2662–2670. 41:927–936.
trans-retinol to 11-cis-retinol. Biochemistry 54. Fisher SK, Lewis GP, Linberg KA, Verardo 70. Wahlin KJ, Adler R, Zack DJ, Campochiaro
2005; 44:11715–11721. MR: Cellular remodeling in mammalian PA: Neurotrophic signaling in normal and
37. Wu BX, Moiseyev G, Chen Y, et al: retina: results from studies of experimental degenerating rodent retinas. Exp Eye Res
Identification of RDH10, an All-trans Retinol retinal detachment. Prog Retin Eye Res 2001; 73:693–701.
Dehydrogenase, in Retinal Muller Cells. 2005; 24:395–431. 71. Di Polo A, Aigner LJ, Dunn RJ, et al:
Invest Ophthalmol Vis Sci 2004; 55. Bringmann A, Pannicke T, Grosche J, et al: Prolonged delivery of brain-derived
45:3857–3862. Muller cells in the healthy and diseased neurotrophic factor by adenovirus-infected
38. Newman EA: Glial modulation of synaptic retina. Prog Retin Eye Res 2006; Muller cells temporarily rescues injured
transmission in the retina. Glia 2004; 25:397–424. retinal ganglion cells. Proc Natl Acad Sci
47:268–274. 56. Garcia M, Vecino E: Role of Muller glia in U S A 1998; 95:3978–3983.
39. Witkovsky P, Dudek FE, Ripps H: Slow PIII neuroprotection and regeneration in the 72. Gauthier R, Joly S, Pernet V, et al: Brain-
component of the carp electroretinogram. retina. Histol Histopathol 2003; derived neurotrophic factor gene delivery
J Gen Physiol 1975; 65:119–134. 18:1205–1218. to muller glia preserves structure and
40. Frishman LJ, Steinberg RH: Light-evoked 57. Francke M, Faude F, Pannicke T, et al: Glial function of light-damaged photoreceptors.
increases in [K+]o in proximal portion of the cell-mediated spread of retinal Invest Ophthalmol Vis Sci 2005;
dark-adapted cat retina. J Neurophysiol degeneration during detachment: a 46:3383–3392.
1989; 61:1233–1243. hypothesis based upon studies in rabbits. 73. Goldstein IM, Ostwald P, Roth S: Nitric
41. Kofuji P, Ceelen P, Zahs KR, et al: Genetic Vision Res 2005; 45:2256–2267. oxide: a review of its role in retinal function
inactivation of an inwardly rectifying 58. Iandiev I, Tenckhoff S, Pannicke T, et al: and disease. Vision Res 1996;
potassium channel (Kir4.1 subunit) in mice: Differential regulation of Kir4.1 and Kir2.1 36:2979–2994.
phenotypic impact in retina. J Neurosci expression in the ischemic rat retina. 74. Roth S: Role of nitric oxide in retinal cell
2000; 20:5733–5740. Neurosci Lett 2006; 396:97–101. death. Clin Neurosci 1997; 4:216–223.
42. Reichenbach A, Robinson SR: The 59. Iandiev I, Uckermann O, Pannicke T, et al: 75. Park CS, Pardhasaradhi K, Gianotti C, et al:
involvement of Muller cells in the outer Glial cell reactivity in a porcine model of Human retina expresses both constitutive
retina. In: Djamgoz MBA, Archer SN, retinal detachment. Invest Ophthalmol Vis and inducible isoforms of nitric oxide
Vallerga S, eds. Neurobiology and clinical Sci 2006; 47:2161–2171. synthase mRNA. Biochem Biophys Res
aspects of the outer retina. London, New 60. Bringmann A, Francke M, Pannicke T, et al: Commun 1994; 205:85–91.
York: Chapman & Hall; 1995:395–416. Role of glial K(+) channels in ontogeny and 76. Goureau O, Regnier-Ricard F, Courtois Y:
43. Kusaka S, Kapousta-Bruneau NV, Puro DG: gliosis: a hypothesis based upon studies Requirement for nitric oxide in retinal
Plasma-induced changes in the physiology on Muller cells. Glia 2000; 29:35–44. neuronal cell death induced by activated
of mammalian retinal glial cells: role of 61. Puro DG: Müller cells; dynamic Muller glial cells. J Neurochem 1999;
glutamate. Glia 1999; 25:205–215. components of the retina. In: Toyoda J-I, 72:2506–2515.
44. Bringmann A, Reichenbach A: Role of Murakami M, Kaneko A, Saito T, eds. The 77. Darius S, Wolf G, Huang PL, Fishman MC:
Muller cells in retinal degenerations. Front retinal basis of vision. Amsterdam, New Localization of NADPH-diaphorase/nitric
Biosci 2001; 6:E72–E92. York: Elsevier; 1999:233–248. oxide synthase in the rat retina: an electron
45. Puro DG: Growth factors and Müller cells. 62. Weick M, Wiedemann P, Reichenbach A, microscopic study. Brain Res 1995;
Prog Retin Eye Res 1995; 15:89–101. Bringmann A: Resensitization of P2Y 690:231–235.
46. Madreperla SA, Geiger GL, Funata M, receptors by growth factor-mediated 78. Cao QL, Murphy HA, Petry HM:
et al:Clinicopathologic correlation of a activation of the phosphatidylinositol-3 Localization of nitric oxide synthase in the
macular hole treated by cortical vitreous kinase in retinal glial cells. Invest tree shrew retina. Vis Neurosci 1999;
peeling and gas tamponade. Ophthalmol Vis Sci 2005; 46:1525–1532. 16:399–409.
Ophthalmology 1994; 101:682–686. 63. Hollborn M, Jahn K, Limb GA, et al: 79. Goureau O, Hicks D, Courtois Y, De Kozak
47. Bringmann A, Kuhrt H, Germer A, et al: Characterization of the basic fibroblast Y: Induction and regulation of nitric oxide
Muller (glial) cell development in vivo and in growth factor-evoked proliferation of the synthase in retinal Muller glial cells.
retinal explant cultures: morphology and human Muller cell line, MIO-M1. Graefes J Neurochem 1994; 63:310–317.
electrophysiology, and the effects of Arch Clin Exp Ophthalmol 2004; 80. Roberge FG, Caspi RR, Chan CC, et al:
elevated ammonia. J Hirnforsch 1998; 242:414–422. Long-term culture of Muller cells from adult
39:193–206. 64. Lieth E, Barber AJ, Xu B, et al: Glial rats in the presence of activated
48. Reichenbach A, Fuchs U, Kasper M, et al: reactivity and impaired glutamate lymphocytes/monocytes products. Curr
Hepatic retinopathy: morphological features metabolism in short-term experimental Eye Res 1985; 4:975–982.
of retinal glial (Muller) cells accompanying diabetic retinopathy. Penn State Retina 81. Kim MK, Chan CC, Belfort R Jr, et al:
hepatic failure. Acta Neuropathol (Berl) Research Group. Diabetes 1998; Histopathologic and immunohistopathologic
1995; 90:273–281. 47:815–820. features of subretinal fibrosis and uveitis 1649
RETINA AND VITREOUS

syndrome. Am J Ophthalmol 1987; degeneration. Ocul Immunol Inflamm 1997; retina but not pigmented epithelium
104:15–23. 5:85–94. differentiation in chick embryos.
82. Caspi RR, Roberge FG: Glial cells as 98. Wen R, Song Y, Cheng T, et al: Injury- Development 1997; 124:805–816.
suppressor cells: characterization of the induced upregulation of bFGF and CNTF 116. Tcheng M, Fuhrmann G, Hartmann MP,
inhibitory function. J Autoimmun 1989; mRNAS in the rat retina. J Neurosci 1995; et al: Spatial and temporal expression
2:709–722. 15:7377–7385. patterns of FGF receptor genes type 1 and
83. Roberge FG, Caspi RR, Nussenblatt RB: 99. Harada C, Harada T, Quah HM, et al: type 2 in the developing chick retina. Exp
Glial retinal Muller cells produce IL-1 Potential role of glial cell line-derived Eye Res 1994; 58:351–358.
activity and have a dual effect on neurotrophic factor receptors in Muller glial 117. Fuhrmann S, Levine EM, Reh TA:
autoimmune T helper lymphocytes. Antigen cells during light-induced retinal Extraocular mesenchyme patterns the optic
presentation manifested after removal of degeneration. Neuroscience 2003; vesicle during early eye development in the
suppressive activity. J Immunol 1988; 122:229–235. embryonic chick. Development 2000;
140:2193–2196. 100. Hollborn M, Tenckhoff S, Jahn K, et al: 127:4599–4609.
84. Chaum E: Retinal neuroprotection by Changes in retinal gene expression in 118. Dohrmann CE, Hemmati-Brivanlou A,
growth factors: a mechanistic perspective. proliferative vitreoretinopathy: glial cell Thomsen GH, et al: Expression of activin
J Cell Biochem 2003; 88:57–75. expression of HB-EGF. Mol Vis 2005; mRNA during early development in
85. Stone J, Maslim J, Valter-Kocsi K, et al: 11:397–413. Xenopus laevis. Dev Biol 1993;
Mechanisms of photoreceptor death and 101. Hirata C, Nakano K, Nakamura N, et al: 157:474–483.
survival in mammalian retina. Prog Retin Advanced glycation end products induce 119. Goding CR: Mitf from neural crest to
Eye Res 1999; 18:689–735. expression of vascular endothelial growth melanoma: signal transduction and
86. Boehm BO, Lang G, Volpert O, et al: Low factor by retinal Muller cells. Biochem transcription in the melanocyte lineage.
SECTION 10

content of the natural ocular anti- Biophys Res Commun 1997; Genes Dev 2000; 14:1712–1728.
angiogenic agent pigment epithelium- 236:712–715. 120. Takeda K, Yasumoto K, Kawaguchi N, et al:
derived factor (PEDF) in aqueous humor 102. Cao W, Wen R, Li F, et al: Induction of basic Mitf-D, a newly identified isoform,
predicts progression of diabetic fibroblast growth factor mRNA by basic expressed in the retinal pigment epithelium
retinopathy. Diabetologia 2003; fibroblast growth factor in Muller cells. Invest and monocyte-lineage cells affected by
46:394–400. Ophthalmol Vis Sci 1997; 38:1358–1366. Mitf mutations. Biochim Biophys Acta
87. Zhang SX, Wang JJ, Gao G, et al: Pigment 103. Chun MH, Ju WK, Kim KY, et al: 2002; 1574:15–23.
epithelium-derived factor (PEDF) is an Upregulation of ciliary neurotrophic factor 121. Bovolenta P, Mallamaci A, Briata P, et al:
endogenous antiinflammatory factor. Faseb in reactive Muller cells in the rat retina Implication of OTX2 in pigment epithelium
J 2006; 20:323–325. following optic nerve transection. Brain Res determination and neural retina
88. Eichler W, Yafai Y, Keller T, et al: PEDF 2000; 868:358–362. differentiation. J Neurosci 1997;
derived from glial Muller cells: a possible 104. Casaccia-Bonnefil P, Gu C, Chao MV: 17:4243–4252.
regulator of retinal angiogenesis. Exp Cell Neurotrophins in cell survival/death 122. Martinez-Morales JR, Signore M,
Res 2004; 299:68–78. decisions. Adv Exp Med Biol 1999; Acampora D, et al: Otx genes are required
89. Ohsato M, Hayashi H, Oshima K, et al: In 468:275–282. for tissue specification in the developing
situ localization of basic fibroblast growth 105. Vecino E, Caminos E, Ugarte M, et al: eye. Development 2001; 128:2019–2030.
factor protein and mRNA in the retina. Immunohistochemical distribution of 123. Grindley JC, Davidson DR, Hill RE: The role
Ophthalmic Res 1997; 29:24–30. neurotrophins and their receptors in the rat of Pax-6 in eye and nasal development.
90. Seki M, Nawa H, Fukuchi T, et al: BDNF is retina and the effects of ischemia and Development 1995; 121:1433–1442.
upregulated by postnatal development and reperfusion. Gen Pharmacol 1998; 124. Baumer N, Marquardt T, Stoykova A, et al:
visual experience: quantitative and 30:305–314. Retinal pigmented epithelium determination
immunohistochemical analyses of BDNF in 106. Nakamura K, Harada C, Okumura A, et al: requires the redundant activities of Pax2 and
the rat retina. Invest Ophthalmol Vis Sci Effect of p75NTR on the regulation of Pax6. Development 2003; 130:2903–2915.
2003; 44:3211–3218. photoreceptor apoptosis in the rd mouse. 125. Belecky-Adams T, Adler R: Developmental
91. Harada T, Harada C, Kohsaka S, et al: Mol Vis 2005; 11:1229–1135. expression patterns of bone
Microglia-Muller glia cell interactions 107. Rohrer B, Matthes MT, LaVail MM, morphogenetic proteins, receptors, and
control neurotrophic factor production Reichardt LF: Lack of p75 receptor does binding proteins in the chick retina. J Comp
during light-induced retinal degeneration. not protect photoreceptors from light- Neurol 2001; 430:562–572.
J Neurosci 2002; 22:9228–9236. induced cell death. Exp Eye Res 2003; 126. Marti E, Bovolenta P: Sonic hedgehog in
92. Harada T, Harada C, Nakayama N, et al: 76:125–129. CNS development: one signal, multiple
Modification of glial-neuronal cell 108. Marmor MF, Wolfensberger TJ: The retinal outputs. Trends Neurosci 2002; 25:89–96.
interactions prevents photoreceptor pigment epithelium. New York: Oxford 127. Wang YP, Dakubo G, Howley P, et al:
apoptosis during light-induced retinal University Press; 1998. Development of normal retinal organization
degeneration. Neuron 2000; 26:533–541. 109. Strauss O: The retinal pigment epithelium depends on Sonic hedgehog signaling from
93. Drescher KM, Whittum-Hudson JA: in visual function. Physiol Rev 2005; ganglion cells. Nat Neurosci 2002;
Evidence for induction of interferon-alpha 85:845–881. 5:831–832.
and interferon-beta in retinal glial cells of 110. Martinez-Morales JR, Rodrigo I, Bovolenta 128. Rahner C, Fukuhara M, Peng S, et al: The
Muller. Virology 1997; 234:309–316. P: Eye development: a view from the retina apical and basal environments of the retinal
94. Drescher KM, Whittum-Hudson JA: Herpes pigmented epithelium. Bioessays 2004; pigment epithelium regulate the maturation
simplex virus type 1 alters transcript levels 26:766–777. of tight junctions during development.
of tumor necrosis factor-alpha and 111. Zuber ME, Gestri G, Viczian AS, et al: J Cell Sci 2004; 117:3307–3318.
interleukin-6 in retinal glial cells. Invest Specification of the vertebrate eye by a 129. Layer PG, Rothermel A, Willbold E:
Ophthalmol Vis Sci 1996; 37:2302–2312. network of eye field transcription factors. Inductive effects of the retinal pigmented
95. Taylor S, Srinivasan B, Wordinger RJ, Development 2003; 130:5155–5167. epithelium (RPE) on histogenesis of the
Roque RS: Glutamate stimulates 112. Wetts R, Fraser SE: Microinjection of avian retina as revealed by retinospheroid
neurotrophin expression in cultured Muller fluorescent tracers to study neural cell technology. Semin Cell Dev Biol 1998;
cells. Brain Res Mol Brain Res 2003; lineages. Development 1991; 9:257–262.
111:189–197. (Suppl 2):1–8. 130. Zhao S, Overbeek PA: Regulation of
96. Behzadian MA, Wang XL, Al-Shabrawey M, 113. Turner DL, Snyder EY, Cepko CL: Lineage- choroid development by the retinal pigment
Caldwell RB: Effects of hypoxia on glial cell independent determination of cell type in epithelium. Mol Vis 2001; 7:277–282.
expression of angiogenesis-regulating the embryonic mouse retina. Neuron 1990; 131. Hollyfield JG, Witkovsky P: Pigmented
factors VEGF and TGF-beta. Glia 1998; 4:833–845. retinal epithelium involvement in
24:216–225. 114. Wetts R, Fraser SE: Multipotent precursors photoreceptor development and function.
97. de Kozak Y, Cotinet A, Goureau O, et al: can give rise to all major cell types of the J Exp Zool 1974; 189:357–378.
Tumor necrosis factor and nitric oxide frog retina. Science 1988; 239:1142–1145. 132. Gaur VP, Liu Y, Turner JE: RPE conditioned
production by resident retinal glial cells 115. Pittack C, Grunwald GB, Reh TA: Fibroblast medium stimulates photoreceptor cell
1650 from rats presenting hereditary retinal growth factors are necessary for neural survival, neurite outgrowth and
Müller Cells and the Retinal Pigment Epithelium

differentiation in vitro. Exp Eye Res 1992; 150. Ts’o MO, Friedman E: The retinal pigment 169. Yao XY, Hageman GS, Marmor MF: Retinal
54:645–659. epithelium. I. Comparative histology. Arch adhesiveness is weakened by enzymatic
133. Rapaport DH, Rakic P, LaVail MM: Ophthalmol 1967; 78:641–649. modification of the interphotoreceptor
Spatiotemporal gradients of cell genesis in 151. Panda-Jonas S, Jonas JB, Jakobczyk- matrix in vivo. Invest Ophthalmol Vis Sci
the primate retina. Perspect Dev Neurobiol Zmija M: Retinal pigment epithelial cell 1990; 31:2051–2058.
1996; 3:147–159. count, distribution, and correlations in 170. Marmor MF, Yao XY, Hageman GS: Retinal
134. Provis JM, van Driel D, Billson FA, Russell normal human eyes. Am J Ophthalmol adhesiveness in surgically enucleated
P: Development of the human retina: 1996; 121:181–189. human eyes. Retina 1994; 14:181–186.
patterns of cell distribution and 152. Dorey CK, Wu G, Ebenstein D, et al: Cell 171. Yao XY, Hageman GS, Marmor MF: Retinal
redistribution in the ganglion cell layer. loss in the aging retina. Relationship to adhesiveness in the monkey. Invest
J Comp Neurol 1985; 233:429–451. lipofuscin accumulation and macular Ophthalmol Vis Sci 1994; 35:744–748.
135. Fleming PA, Braekevelt CR, Harman AM, degeneration. Invest Ophthalmol Vis Sci 172. Fatt I, Shantinath K: Flow conductivity of
Beazley LD: Retinal pigment epithelium 1989; 30:1691–1699. retina and its role in retinal adhesion. Exp
and photoreceptor maturation in a wallaby, 153. Krebs W, Krebs IP: Quantitative Eye Res 1971; 12:218–226.
the quokka. J Comp Neurol 1996; morphology of the central fovea in the 173. Marmor MF: Retinal detachment from
370:47–60. primate retina. Am J Anat 1989; hyperosmotic intravitreal injection. Invest
136. Gundersen D, Powell SK, Rodriguez- 184:225–236. Ophthalmol Vis Sci 1979; 18:1237–1244.
Boulan E: Apical polarization of N-CAM in 154. Curcio CA, Millican CL, Allen KA, Kalina 174. Kita M, Marmor MF: Systemic mannitol
retinal pigment epithelium is dependent on RE: Aging of the human photoreceptor increases the retinal adhesive force in vivo.
contact with the neural retina. J Cell Biol mosaic: evidence for selective vulnerability Arch Ophthalmol 1991; 109:1449–1450.

CHAPTER 125
1993; 121:335–343. of rods in central retina. Invest Ophthalmol 175. Nandrot EF, Anand M, Sircar M, Finnemann
137. Raymond SM, Jackson IJ: The retinal Vis Sci 1993; 34:3278–3296. SC: Novel role for alphavbeta5–integrin in
pigmented epithelium is required for 155. Feeney-Burns L, Burns RP, Gao CL: Age- retinal adhesion and its diurnal peak. Am J
development and maintenance of the related macular changes in humans over Physiol Cell Physiol 2006;
mouse neural retina. Curr Biol 1995; 90 years old. Am J Ophthalmol 1990; 290:C1256–C1262.
5:1286–1295. 109:265–278. 176. Mayerson PL, Hall MO: Rat retinal pigment
138. Pearson RA, Dale N, Llaudet E, Mobbs P: 156. Gao H, Hollyfield JG: Aging of the human epithelial cells show specificity of
ATP released via gap junction hemichannels retina. Differential loss of neurons and phagocytosis in vitro. J Cell Biol 1986;
from the pigment epithelium regulates retinal pigment epithelial cells. Invest 103:299–308.
neural retinal progenitor proliferation. Ophthalmol Vis Sci 1992; 33:1–17. 177. Young RW: The Bowman Lecture, 1982.
Neuron 2005; 46:731–744. 157. Panda-Jonas S, Jonas JB, Jakobczyk- Biological renewal. Applications to the eye.
139. Land PW, Lund RD: Development of the Zmija M: Retinal photoreceptor density Trans Ophthalmol Soc U K 1982; 102
rat’s uncrossed retinotectal pathway and its decreases with age. Ophthalmology 1995; (Pt 1):42–75.
relation to plasticity studies. Science 1979; 102:1853–1859. 178. LaVail MM: Kinetics of rod outer segment
205:698–700. 158. Panda-Jonas S, Jonas JB, Jakobczyk M, renewal in the developing mouse retina.
140. Jeffery G: The albino retina: an abnormality Schneider U: Retinal photoreceptor count, J Cell Biol 1973; 58:650–661.
that provides insight into normal retinal retinal surface area, and optic disc size in 179. Young RW, Bok D: Participation of the
development. Trends Neurosci 1997; normal human eyes. Ophthalmology 1994; retinal pigment epithelium in the rod outer
20:165–169. 101:519–523. segment renewal process. J Cell Biol 1969;
141. Behling KC, Surace EM, Bennett J: 159. Watzke RC, Soldevilla JD, Trune DR: 42:392–403.
Pigment epithelium-derived factor Morphometric analysis of human retinal 180. Bok D: The retinal pigment epithelium: a
expression in the developing mouse eye. pigment epithelium: correlation with age and versatile partner in vision. J Cell Sci Suppl
Mol Vis 2002; 8:449–454. location. Curr Eye Res 1993; 12:133–142. 1993; 17:189–195.
142. Jablonski MM, Tombran-Tink J, Mrazek 160. Burke JM, Skumatz CM, Irving PE, McKay 181. Steinberg RH, Wood I: Pigment epithelial
DA, Iannaccone A: Pigment epithelium- BS: Phenotypic heterogeneity of retinal cell ensheathment of cone outer segments
derived factor supports normal pigment epithelial cells in vitro and in situ. in the retina of the domestic cat. Proc R
development of photoreceptor neurons and Exp Eye Res 1996; 62:63–73. Soc Lond B Biol Sci 1974; 187:461–478.
opsin expression after retinal pigment 161. Ts’o MO, Friedman E: The retinal pigment 182. Steinberg RH, Wood I, Hogan MJ: Pigment
epithelium removal. J Neurosci 2000; epithelium. III. Growth and development. epithelial ensheathment and phagocytosis
20:7149–7157. Arch Ophthalmol 1968; 80:214–216. of extrafoveal cones in human retina. Philos
143. King GL, Suzuma K: Pigment-epithelium- 162. Nagai H, Kalnins VI: Normally occurring Trans R Soc Lond B Biol Sci 1977;
derived factor—a key coordinator of retinal loss of single cells and repair of resulting 277:459–474.
neuronal and vascular functions. N Engl J defects in retinal pigment epithelium in situ. 183. Nguyen-Legros J, Hicks D: Renewal of
Med 2000; 342:349–351. Exp Eye Res 1996; 62:55–61. photoreceptor outer segments and their
144. Burns MS, Hartz MJ: The retinal pigment 163. Feeney-Burns L, Hilderbrand ES, Eldridge phagocytosis by the retinal pigment
epithelium induces fenestration of S: Aging human RPE: morphometric epithelium. Int Rev Cytol 2000; 196:245–313.
endothelial cells in vivo. Curr Eye Res analysis of macular, equatorial, and 184. Young RW: An hypothesis to account for a
1992; 11:863–873. peripheral cells. Invest Ophthalmol Vis Sci basic distinction between rods and cones.
145. Rymer J, Wildsoet CF: The role of the 1984; 25:195–200. Vision Res 1971; 11:1–5.
retinal pigment epithelium in eye growth 164. Aijaz S, Balda MS, Matter K: Tight 185. Williams DS, Fisher SK: Prevention of rod
regulation and myopia: a review. Vis junctions: molecular architecture and disk shedding by detachment from the
Neurosci 2005; 22:251–261. function. Int Rev Cytol 2006; 248:261–298. retinal pigment epithelium. Invest
146. Campochiaro PA, Jerdon JA, Glaser BM: 165. Zhao S, Rizzolo LJ, Barnstable CJ: Ophthalmol Vis Sci 1987; 28:184–187.
The extracellular matrix of human retinal Differentiation and transdifferentiation of 186. Sidman RL, Kosaras B, Tang M: Pigment
pigment epithelial cells in vivo and its the retinal pigment epithelium. Int Rev epithelial and retinal phenotypes in the
synthesis in vitro. Invest Ophthalmol Vis Sci Cytol 1997; 171:225–266. vitiligo mivit, mutant mouse. Invest
1986; 27:1615–1621. 166. Nicholson BJ: Gap junctions – from Ophthalmol Vis Sci 1996; 37:1097–1115.
147. Takei Y, Ozanics V: Origin and development cell to molecule. J Cell Sci 2003; 187. Sun M, Finnemann SC, Febbraio M, et al:
of Bruch’s membrane in monkey fetuses: 116:4479–4481. Light-induced oxidation of photoreceptor
an electron microscopic study. Invest 167. Kita M, Marmor MF: Retinal adhesive force outer segment phospholipids generates
Ophthalmol 1975; 14:903–916. in living rabbit, cat, and monkey eyes. ligands for CD36–mediated phagocytosis
148. Olson MD: Development of Bruch’s Normative data and enhancement by by retinal pigment epithelium: a potential
membrane in the chick: an electron mannitol and acetazolamide. Invest mechanism for modulating outer segment
microscopic study. Invest Ophthalmol Vis Ophthalmol Vis Sci 1992; 33:1879–1882. phagocytosis under oxidant stress
Sci 1979; 18:329–338. 168. Marmor MF, Yao XY: The metabolic conditions. J Biol Chem 2006;
149. Hogan MJ, Alvarado JA, Weddell JE: Retina dependency of retinal adhesion in rabbit 281:4222–4230.
Histology of the Human Eye. Philadelphia: and primate. Arch Ophthalmol 1995; 188. Boyle D, Tien LF, Cooper NG, et al: A
W B Saunders; 1971:393–522. 113:232–238. mannose receptor is involved in retinal 1651
RETINA AND VITREOUS

phagocytosis. Invest Ophthalmol Vis Sci interaction. J Histochem Cytochem 1993; interphotoreceptor matrix: apical secretion
1991; 32:1464–1470. 41:253–263. and distribution. Exp Eye Res 2004;
189. Feng W, Yasumura D, Matthes MT, et al: 205. Rakoczy PE, Lai CM, Baines M, et al: 78:223–234.
Mertk triggers uptake of photoreceptor Modulation of cathepsin D activity in retinal 221. Blaauwgeers HG, Holtkamp GM, Rutten H,
outer segments during phagocytosis by pigment epithelial cells. Biochem J 1997; et al: Polarized vascular endothelial growth
cultured retinal pigment epithelial cells. 324 (Pt 3):935–940. factor secretion by human retinal pigment
J Biol Chem 2002; 277:17016–17022. 206. Zimmerman WF, Godchaux W III, Belkin M: epithelium and localization of vascular
190. Finnemann SC, Silverstein RL: Differential The relative proportions of lysosomal endothelial growth factor receptors on the
roles of CD36 and alphavbeta5 integrin in enzyme activities in bovine retinal pigment inner choriocapillaris. Evidence for a
photoreceptor phagocytosis by the retinal epithelium. Exp Eye Res 1983; 36:151–158. trophic paracrine relation. Am J Pathol
pigment epithelium. J Exp Med 2001; 207. Rakoczy PE, Mann K, Cavaney DM, et al: 1999; 155:421–428.
194:1289–1298. Detection and possible functions of a 222. Jablonski MM, Tombran-Tink J, Mrazek
191. Finnemann SC, Bonilha VL, Marmorstein cysteine protease involved in digestion of DA, Iannaccone A: Pigment epithelium-
AD, Rodriguez-Boulan E: Phagocytosis of rod outer segments by retinal pigment derived factor supports normal Muller cell
rod outer segments by retinal pigment epithelial cells. Invest Ophthalmol Vis Sci development and glutamine synthetase
epithelial cells requires alpha(v)beta5 1994; 35:4100–4108. expression after removal of the retinal
integrin for binding but not for 208. Ershov AV, Lukiw WJ, Bazan NG: Selective pigment epithelium. Glia 2001; 35:14–25.
internalization. Proc Natl Acad Sci U S A transcription factor induction in retinal 223. Yoshimura N, Matsumoto M, Shimizu H,
1997; 94:12932–12937. pigment epithelial cells during et al: Photocoagulated human retinal
192. Gregory CY, Hall MO: The phagocytosis of photoreceptor phagocytosis. J Biol Chem pigment epithelial cells produce an inhibitor
ROS by RPE cells is inhibited by an 1996; 271:28458–28462. of vascular endothelial cell proliferation.
SECTION 10

antiserum to rat RPE cell plasma 209. Ershov AV, Bazan NG: Photoreceptor Invest Ophthalmol Vis Sci 1995;
membranes. Exp Eye Res 1992; phagocytosis selectively activates 36:1686–1691.
54:843–851. PPARgamma expression in retinal pigment 224. Khaliq A, Patel B, Jarvis-Evans J, et al:
193. Kindzelskii AL, Elner VM, Elner SG, et al: epithelial cells. J Neurosci Res 2000; Oxygen modulates production of bFGF and
Toll-like receptor 4 (TLR4) of retinal pigment 60:328–337. TGF-beta by retinal cells in vitro. Exp Eye
epithelial cells participates in 210. Gregory CY, Abrams TA, Hall MO: Stimulation Res 1995; 60:415–423.
transmembrane signaling in response to of A2 adenosine receptors inhibits the 225. Baudouin C, Fredj-Reygrobellet D, Brignole
photoreceptor outer segments. J Gen ingestion of photoreceptor outer segments F, et al: Growth factors in vitreous and
Physiol 2004; 124:139–149. by retinal pigment epithelium. Invest subretinal fluid cells from patients with
194. Chaitin MH, Hall MO: Defective ingestion of Ophthalmol Vis Sci 1994; 35:819–825. proliferative vitreoretinopathy. Ophthalmic
rod outer segments by cultured dystrophic 211. Cahill GM, Besharse JC: Circadian Res 1993; 25:52–59.
rat pigment epithelial cells. Invest rhythmicity in vertebrate retinas: regulation 226. Amin R, Puklin JE, Frank RN: Growth factor
Ophthalmol Vis Sci 1983; 24:812–820. by a photoreceptor oscillator. Prog Retin localization in choroidal neovascular
195. Chen J, Carey K, Godowski PJ: Eye Res 1995; 14:267–291. membranes of age-related macular
Identification of Gas6 as a ligand for Mer, a 212. Nandrot EF, Kim Y, Brodie SE, et al: Loss of degeneration. Invest Ophthalmol Vis Sci
neural cell adhesion molecule related synchronized retinal phagocytosis and age- 1994; 35:3178–3188.
receptor tyrosine kinase implicated in related blindness in mice lacking 227. Lutty GA, Merges C, Crone S, McLeod DS:
cellular transformation. Oncogene 1997; alphavbeta5 integrin. J Exp Med 2004; Immunohistochemical insights into sickle
14:2033–2039. 200:1539–1545. cell retinopathy. Curr Eye Res 1994;
196. Hall MO, Prieto AL, Obin MS, et al: Outer 213. Tanihara H, Inatani M, Honda Y: Growth 13:125–138.
segment phagocytosis by cultured retinal factors and their receptors in the retina and 228. Raymond MC, Thompson JT: RPE-mediated
pigment epithelial cells requires Gas6. Exp pigment epithelium. Prog Retin Eye Res collagen gel contraction. Inhibition by
Eye Res 2001; 73:509–520. 1997; 16:271–301. colchicine and stimulation by TGF-beta.
197. Hall MO, Agnew BJ, Abrams TA, Burgess 214. Tombran-Tink J, Shivaram SM, Chader GJ, Invest Ophthalmol Vis Sci 1990;
BL: The phagocytosis of os is mediated by et al: Expression, secretion, and age- 31:1079–1086.
the PI3-kinase linked tyrosine kinase related downregulation of pigment 229. Campochiaro PA, Glaser BM: Platelet-
receptor, mer, and is stimulated by GAS6. epithelium-derived factor, a serpin with derived growth factor is chemotactic for
Adv Exp Med Biol 2003; 533:331–336. neurotrophic activity. J Neurosci 1995; human retinal pigment epithelial cells. Arch
198. Finnemann SC: Focal adhesion kinase 15:4992–5003. Ophthalmol 1985; 103:576–579.
signaling promotes phagocytosis of 215. Steele FR, Chader GJ, Johnson LV, 230. Nagineni CN, Kutty V, Detrick B, Hooks JJ:
integrin-bound photoreceptors. Embo J Tombran-Tink J: Pigment epithelium- Expression of PDGF and their receptors in
2003; 22:4143–4154. derived factor: neurotrophic activity and human retinal pigment epithelial cells and
199. Heth CA, Marescalchi PA: Inositol identification as a member of the serine fibroblasts: regulation by TGF-beta. J Cell
triphosphate generation in cultured rat protease inhibitor gene family. Proc Natl Physiol 2005; 203:35–43.
retinal pigment epithelium. Invest Acad Sci U S A 1993; 90:1526–1530. 231. Brunk UT, Terman A: Lipofuscin:
Ophthalmol Vis Sci 1994; 35:409–416. 216. Ogata N, Wang L, Jo N, et al: Pigment mechanisms of age-related accumulation
200. Hall MO, Abrams TA, Mittag TW: ROS epithelium derived factor as a neuroprotective and influence on cell function. Free Radic
ingestion by RPE cells is turned off by agent against ischemic retinal injury. Curr Eye Biol Med 2002; 33:611–619.
increased protein kinase C activity and by Res 2001; 22:245–252. 232. Sundelin SP, Nilsson SE, Brunk UT:
increased calcium. Exp Eye Res 1991; 217. Dawson DW, Volpert OV, Gillis P, et al: Lipofuscin-formation in cultured retinal
52:591–598. Pigment epithelium-derived factor: a potent pigment epithelial cells is related to their
201. Hall MO, Abrams TA, Mittag TW: The inhibitor of angiogenesis. Science 1999; melanin content. Free Radic Biol Med
phagocytosis of rod outer segments is 285:245–248. 2001; 30:74–81.
inhibited by drugs linked to cyclic adenosine 218. Barnstable CJ, Tombran-Tink J: 233. Wing GL, Blanchard GC, Weiter JJ: The
monophosphate production. Invest Neuroprotective and antiangiogenic actions topography and age relationship of
Ophthalmol Vis Sci 1993; 34:2392–2401. of PEDF in the eye: molecular targets and lipofuscin concentration in the retinal
202. Edwards RB, Flaherty PM: Association of therapeutic potential. Prog Retin Eye Res pigment epithelium. Invest Ophthalmol Vis
changes in intracellular cyclic AMP with 2004; 23:561–577. Sci 1978; 17:601–607.
changes in phagocytosis in cultured rat 219. Adamis AP, Shima DT, Yeo KT, et al: 234. Katz ML, Drea CM, Eldred GE, et al: Influence
pigment epithelium. Curr Eye Res 1986; Synthesis and secretion of vascular of early photoreceptor degeneration on
5:19–26. permeability factor/vascular endothelial lipofuscin in the retinal pigment epithelium.
203. Burnside MB: Possible roles of microtubules growth factor by human retinal pigment Exp Eye Res 1986; 43:561–573.
and actin filaments in retinal pigmented epithelial cells. Biochem Biophys Res 235. Lorenz B, Wabbels B, Wegscheider E,
epithelium. Exp Eye Res 1976; 23:257–275. Commun 1993; 193:631–638. et al: Lack of fundus autofluorescence to
204. Bosch E, Horwitz J, Bok D: Phagocytosis 220. Becerra SP, Fariss RN, Wu YQ, et al: 488 nanometers from childhood on in
of outer segments by retinal pigment Pigment epithelium-derived factor in the patients with early-onset severe retinal
1652 epithelium: phagosome-lysosome monkey retinal pigment epithelium and dystrophy associated with mutations in
Müller Cells and the Retinal Pigment Epithelium

RPE65. Ophthalmology 2004; 252. Boulton M, Dontsov A, Jarvis-Evans J, 270. Blaug S, Quinn R, Quong J, et al: Retinal
111:1585–1594. et al: Lipofuscin is a photoinducible free pigment epithelial function: a role for
236. Katz ML, Eldred GE: Retinal light damage radical generator. J Photochem Photobiol B CFTR? Doc Ophthalmol 2003; 106:43–50.
reduces autofluorescent pigment 1993; 19:201–204. 271. Hughes BA, Adorante JS, Miller SS, Lin H:
deposition in the retinal pigment epithelium. 253. Davies S, Elliott MH, Floor E, et al: Apical electrogenic NaHCO3 cotransport.
Invest Ophthalmol Vis Sci 1989; 30:37–43. Photocytotoxicity of lipofuscin in human A mechanism for HCO3 absorption across
237. Feeney L: Lipofuscin and melanin of human retinal pigment epithelial cells. Free Radic the retinal pigment epithelium. J Gen
retinal pigment epithelium. Fluorescence, Biol Med 2001; 31:256–265. Physiol 1989; 94:125–150.
enzyme cytochemical, and ultrastructural 254. Schutt F, Davies S, Kopitz J, et al: 272. Lin H, Kenyon E, Miller SS: Na-dependent
studies. Invest Ophthalmol Vis Sci 1978; Photodamage to human RPE cells by pHi regulatory mechanisms in native
17:583–600. A2–E, a retinoid component of lipofuscin. human retinal pigment epithelium. Invest
238. Burke JM, Skumatz CM: Autofluorescent Invest Ophthalmol Vis Sci 2000; Ophthalmol Vis Sci 1992; 33:3528–3538.
inclusions in long-term postconfluent 41:2303–2308. 273. Celesia GG: Anatomy and physiology of
cultures of retinal pigment epithelium. Invest 255. Terman A, Brunk UT: Ceroid/lipofuscin visual evoked potentials and
Ophthalmol Vis Sci 1998; 39:1478–1486. formation in cultured human fibroblasts: the electroretinograms. Neurol Clin 1988;
239. Katz ML, Gao CL, Rice LM: Formation of role of oxidative stress and lysosomal 6:657–679.
lipofuscin-like fluorophores by reaction of proteolysis. Mech Ageing Dev 1998; 274. Steinberg RH, Schmidt R, Brown KT:
retinal with photoreceptor outer segments 104:277–291. Intracellular responses to light from cat
and liposomes. Mech Ageing Dev 1996; 256. Schutt F, Bergmann M, Holz FG, Kopitz J: pigment epithelium: origin of the
92:159–174. Proteins modified by malondialdehyde, electroretinogram c-wave. Nature 1970;

CHAPTER 125
240. Wassell J, Ellis S, Burke J, Boulton M: 4–hydroxynonenal, or advanced glycation 227:728–730.
Fluorescence properties of autofluorescent end products in lipofuscin of human retinal 275. Karowski CJ, Proenza LM: Relationship
granules generated by cultured human RPE pigment epithelium. Invest Ophthalmol Vis between Muller cell responses, a local
cells. Invest Ophthalmol Vis Sci 1998; Sci 2003; 44:3663–3668. transretinal potential, and potassium flux.
39:1487–1492. 257. Katz ML: Incomplete proteolysis may J Neurophysiol 1977; 40:244–259.
241. Sundelin S, Wihlmark U, Nilsson SE, Brunk contribute to lipofuscin accumulation in the 276. Linsenmeier RA, Steinberg RH: Delayed
UT: Lipofuscin accumulation in cultured retinal pigment epithelium. Adv Exp Med Biol basal hyperpolarization of cat retinal
retinal pigment epithelial cells reduces their 1989; 266:109–116; discussion 116–118. pigment epithelium and its relation to the
phagocytic capacity. Curr Eye Res 1998; 258. Finnemann SC, Leung LW, Rodriguez- fast oscillation of the DC electroretinogram.
17:851–857. Boulan E: The lipofuscin component A2E J Gen Physiol 1984; 83:213–232.
242. Katz ML, Drea CM, Robison WG Jr: selectively inhibits phagolysosomal 277. Philp NJ, Yoon H, Grollman EF:
Relationship between dietary retinol and degradation of photoreceptor phospholipid Monocarboxylate transporter MCT1 is
lipofuscin in the retinal pigment epithelium. by the retinal pigment epithelium. Proc Natl located in the apical membrane and MCT3
Mech Ageing Dev 1986; 35:291–305. Acad Sci U S A 2002; 99:3842–3847. in the basal membrane of rat RPE. Am J
243. Delori FC, Goger DG, Dorey CK: Age- 259. Sparrow JR, Fishkin N, Zhou J, et al: A2E, Physiol 1998; 274:R1824–R1828.
related accumulation and spatial a byproduct of the visual cycle. Vision Res 278. Senanayake P, Calabro A, Hu JG, et al:
distribution of lipofuscin in RPE of normal 2003; 43:2983–2990. Glucose utilization by the retinal pigment
subjects. Invest Ophthalmol Vis Sci 2001; 260. Hamann S: Molecular mechanisms of water epithelium: evidence for rapid uptake and
42:1855–1866. transport in the eye. Int Rev Cytol 2002; storage in glycogen, followed by glycogen
244. Sparrow JR, Zhou J, Ben-Shabat S, et al: 215:395–431. utilization. Exp Eye Res 2006; 83:235–246.
Involvement of oxidative mechanisms in 261. Hughes BA, Gallemore RP, Miller SS: 279. Ban Y, Rizzolo LJ: Regulation of glucose
blue-light-induced damage to A2E-laden Transport mechanisms in the retinal transporters during development of the
RPE. Invest Ophthalmol Vis Sci 2002; pigment epithelium. In: Marmor MF, retinal pigment epithelium. Brain Res Dev
43:1222–1227. Wolfensberger TJ, eds. The retinal pigment Brain Res 2000; 121:89–95.
245. Sparrow JR, Boulton M: RPE lipofuscin and epithelium. New York: Oxford University 280. Blomhoff R, Green MH, Berg T, Norum KR:
its role in retinal pathobiology. Exp Eye Res Press; 1998. Transport and storage of vitamin A.
2005; 80:595–606. 262. Steinberg RH, Linsenmeier RA, Griff ER: Science 1990; 250:399–404.
246. Liu J, Itagaki Y, Ben-Shabat S, et al: The Retinal pigment epithelial cell contributions to 281. Bok D, Heller J: Transport of retinol from
biosynthesis of A2E, a fluorophore of aging the electroretinogram and electrooculogram. the blood to the retina: an autoradiographic
retina, involves the formation of the Prog Retin Eye Res 1985; 4:33–66. study of the pigment epithelial cell surface
precursor, A2–PE, in the photoreceptor 263. Jaffe GJ, Burke JM, Geroski DH: Ouabain- receptor for plasma retinol-binding protein.
outer segment membrane. J Biol Chem sensitive Na+-K+ ATPase pumps in Exp Eye Res 1976; 22:395–402.
2000; 275:29354–29360. cultured human retinal pigment epithelium. 282. Bazan NG, Gordon WC, Rodriguez de
247. Ben-Shabat S, Parish CA, Vollmer HR, Exp Eye Res 1989; 48:61–68. Turco EB: Docosahexaenoic acid uptake
et al: Biosynthetic studies of A2E, a major 264. Miller SS, Steinberg RH, Oakley B II:The and metabolism in photoreceptors: retinal
fluorophore of retinal pigment epithelial electrogenic sodium pump of the frog conservation by an efficient retinal pigment
lipofuscin. J Biol Chem 2002; retinal pigment epithelium. J Membr Biol epithelial cell-mediated recycling process.
277:7183–7190. 1978; 44:259–279. Adv Exp Med Biol 1992; 318:295–306.
248. Parish CA, Hashimoto M, Nakanishi K, 265. Hu JG, Gallemore RP, Bok D, Frambach 283. Jeffrey BG, Weisinger HS, Neuringer M,
et al: Isolation and one-step preparation of DA: Chloride transport in cultured fetal Mitchell DC: The role of docosahexaenoic
A2E and iso-A2E, fluorophores from human human retinal pigment epithelium. Exp Eye acid in retinal function. Lipids 2001;
retinal pigment epithelium. Proc Natl Acad Res 1996; 62:443–448. 36:859–871.
Sci U S A 1998; 95:14609–14613. 266. Kennedy BG: Na(+)-K(4)-Cl- cotransport in 284. Petrukhin K, Koisti MJ, Bakall B, et al:
249. Ivy GO, Kanai S, Ohta M, et al: Lipofuscin- cultured cells derived from human retinal Identification of the gene responsible for
like substances accumulate rapidly in brain, pigment epithelium. Am J Physiol 1990; Best macular dystrophy. Nat Genet 1998;
retina and internal organs with cysteine 259:C29–C34. 19:241–247.
protease inhibition. Adv Exp Med Biol 267. Sun H, Tsunenari T, Yau KW, Nathans J: The 285. Yardley J, Leroy BP, Hart-Holden N, et al:
1989; 266:31–45; discussion 45–47. vitelliform macular dystrophy protein defines Mutations of VMD2 splicing regulators
250. Kikugawa K, Kato T, Beppu M, Hayasaka a new family of chloride channels. Proc cause nanophthalmos and autosomal
A: Fluorescent and cross-linked proteins Natl Acad Sci U S A 2002; 99:4008–4013. dominant vitreoretinochoroidopathy
formed by free radical and aldehyde 268. Ueda Y, Steinberg RH: Chloride currents in (ADVIRC). Invest Ophthalmol Vis Sci 2004;
species generated during lipid oxidation. freshly isolated rat retinal pigment epithelial 45:3683–3689.
Adv Exp Med Biol 1989; 266:345–356; cells. Exp Eye Res 1994; 58:331–342. 286. Kaufman SJ, Goldberg MF, Orth DH,
discussion 357. 269. Weng TX, Godley BF, Jin GF, et al: Oxidant et al: Autosomal dominant
251. Terman A, Abrahamsson N, Brunk UT: and antioxidant modulation of chloride vitreoretinochoroidopathy. Arch Ophthalmol
Ceroid/lipofuscin-loaded human fibroblasts channels expressed in human retinal 1982; 100:272–278.
show increased susceptibility to oxidative pigment epithelium. Am J Physiol Cell 287. Wu J, Marmorstein AD, Peachey NS:
stress. Exp Gerontol 1999; 34:755–770. Physiol 2002; 283:C839–C849. Functional abnormalities in the retinal 1653
RETINA AND VITREOUS

pigment epithelium of CFTR mutant mice. melanin and choroidal melanin in human 325. Gonzalez-Fernandez F: Interphotoreceptor
Exp Eye Res 2006; 83:424–428. eyes. Invest Ophthalmol Vis Sci 1986; retinoid-binding protein—an old gene for
288. Tsuboi S, Pederson JE: Volume flow across 27:145–152. new eyes. Vision Res 2003; 43:3021–3036.
the isolated retinal pigment epithelium of 306. Sarna T: Properties and function of the 326. Arshavsky V: Like night and day: rods and
cynomolgus monkey eyes. Invest ocular melanin—a photobiophysical view. cones have different pigment regeneration
Ophthalmol Vis Sci 1988; 29:1652–1655. J Photochem Photobiol B 1992; 12:215–258. pathways. Neuron 2002; 36:1–3.
289. Tsuboi S: Measurement of the volume flow 307. Kobayashi N, Muramatsu T, Yamashina Y, 327. Mata NL, Moghrabi WN, Lee JS, et al:
and hydraulic conductivity across the et al: Melanin reduces ultraviolet-induced Rpe65 is a retinyl ester binding protein that
isolated dog retinal pigment epithelium. DNA damage formation and killing rate in presents insoluble substrate to the
Invest Ophthalmol Vis Sci 1987; cultured human melanoma cells. J Invest isomerase in retinal pigment epithelial cells.
28:1776–1782. Dermatol 1993; 101:685–689. J Biol Chem 2004; 279:635–643.
290. Emi K, Pederson JE, Toris CB: Hydrostatic 308. Rapp LM, Williams TP: The role of ocular 328. Xue L, Gollapalli DR, Maiti P, et al: A
pressure of the suprachoroidal space. Invest pigmentation in protecting against retinal palmitoylation switch mechanism in the
Ophthalmol Vis Sci 1989; 30:233–238. light damage. Vision Res 1980; regulation of the visual cycle. Cell 2004;
291. Negi A, Kawano S, Marmor MF: Effects of 20:1127–1131. 117:761–771.
intraocular pressure and other factors on 309. Sanyal S, Zeilmaker GH: Retinal damage 329. Jang GF, Van Hooser JP, Kuksa V, et al:
subretinal fluid resorption. Invest by constant light in chimaeric mice: Characterization of a dehydrogenase
Ophthalmol Vis Sci 1987; 28:2099–2102. implications for the protective role of activity responsible for oxidation of 11-cis-
292. Takeuchi A, Kricorian G, Yao XY, et al: The melanin. Exp Eye Res 1988; 46:731–743. retinol in the retinal pigment epithelium of
rate and source of albumin entry into 310. LaVail MM, Gorrin GM: Protection from mice with a disrupted RDH5 gene. A model
saline-filled experimental retinal light damage by ocular pigmentation: for the human hereditary disease fundus
SECTION 10

detachments. Invest Ophthalmol Vis Sci analysis using experimental chimeras and albipunctatus. J Biol Chem 2001;
1994; 35:3792–3798. translocation mice. Exp Eye Res 1987; 276:32456–32465.
293. Takeuchi A, Kricorian G, Marmor MF: 44:877–889. 330. Saari JC, Nawrot M, Kennedy BN, et al:
Albumin movement out of the subretinal 311. Sarna T, Menon IA, Sealy RC: Visual cycle impairment in cellular
space after experimental retinal Photosensitization of melanins: a retinaldehyde binding protein (CRALBP)
detachment. Invest Ophthalmol Vis Sci comparative study. Photochem Photobiol knockout mice results in delayed dark
1995; 36:1298–1305. 1985; 42:529–532. adaptation. Neuron 2001; 29:739–748.
294. Chihara E, Nao-i N: Resorption of 312. Felix CC, Hyde JS, Sarna T, Sealy RC: 331. Chen P, Hao W, Rife L, et al: A photic visual
subretinal fluid by transepithelial flow of the Melanin photoreactions in aerated media: cycle of rhodopsin regeneration is dependent
retinal pigment epithelium. Graefes Arch electron spin resonance evidence for on Rgr. Nat Genet 2001; 28:256–260.
Clin Exp Ophthalmol 1985; 223:202–204. production of superoxide and hydrogen 332. McBee JK, Palczewski K, Baehr W,
295. Nielsen S, Smith BL, Christensen EI, Agre peroxide. Biochem Biophys Res Commun Pepperberg DR: Confronting complexity:
P: Distribution of the aquaporin CHIP in 1978; 84:335–341. the interlink of phototransduction and
secretory and resorptive epithelia and 313. Ham WT Jr, Ruffolo JJ Jr, Mueller HA, retinoid metabolism in the vertebrate retina.
capillary endothelia. Proc Natl Acad Sci U Guerry D III: The nature of retinal radiation Prog Retin Eye Res 2001; 20:469–529.
S A 1993; 90:7275–7279. damage: dependence on wavelength, 333. Martinez-Mir A, Paloma E, Allikmets R,
296. Stamer WD, Bok D, Hu J, et al: Aquaporin- power level and exposure time. Vision Res et al: Retinitis pigmentosa caused by a
1 channels in human retinal pigment 1980; 20:1105–1111. homozygous mutation in the Stargardt
epithelium: role in transepithelial water 314. Rozanowska M, Jarvis-Evans J, Korytowski disease gene ABCR. Nat Genet 1998;
movement. Invest Ophthalmol Vis Sci 2003; W, et al: Blue light-induced reactivity of 18:11–12.
44:2803–2808. retinal age pigment. In vitro generation of 334. Allikmets R, Shroyer NF, Singh N, et al:
297. Boulton M: Melanin and the RPE. In: oxygen-reactive species. J Biol Chem Mutation of the Stargardt disease gene
Marmor MF, Wolfensberger TJ, eds. The 1995; 270:18825–18830. (ABCR) in age-related macular degeneration.
retinal pigment epithelium. New York: 315. Marshall J: Lasers in ophthalmology: the Science 1997; 277:1805–1807.
Oxford University Press; 1998:68–85. basic principles. Eye 1988; 2:S98–S112. 335. Stone EM, Webster AR, Vandenburgh K,
298. Boissy RE: The melanocyte. Its structure, 316. Hansen WP, Fine S: Melanin granule et al: Allelic variation in ABCR associated
function, and subpopulations in skin, eyes, models for pulsed laser induced retinal with Stargardt disease but not age-related
and hair. Dermatol Clin 1988; 6:161–173. injury. Appl Opt 1968; 7:155–158. macular degeneration. Nat Genet 1998;
299. Schraermeyer U: Does melanin turnover 317. Koneru PB, Lien EJ, Koda RT: Oculotoxicities 20:328–329.
occur in the eyes of adult vertebrates? of systemically administered drugs. J Ocul 336. Kuroiwa S, Kojima H, Kikuchi T, Yoshimura
Pigment Cell Res 1993; 6:193–204. Pharmacol 1986; 2:385–404. N: ATP binding cassette transporter retina
300. Boulton M, Docchio F, Dayhaw-Barker P, 318. Drager UC: Calcium binding in pigmented genotypes and age related macular
et al: Age-related changes in the and albino eyes. Proc Natl Acad Sci U S A degeneration: an analysis on exudative
morphology, absorption and fluorescence 1985; 82:6716–6720. non-familial Japanese patients. Br J
of melanosomes and lipofuscin granules of 319. Larsson BS: Interaction between chemicals Ophthalmol 1999; 83:613–615.
the retinal pigment epithelium. Vision Res and melanin. Pigment Cell Res 1993; 337. Maw MA, Kennedy B, Knight A, et al:
1990; 30:1291–1303. 6:127–133. Mutation of the gene encoding cellular
301. Schraermeyer U, Kopitz J, Peters S, et al: 320. Arshavsky VY, Lamb TD, Pugh EN, Jr: G retinaldehyde-binding protein in autosomal
Tyrosinase biosynthesis in adult proteins and phototransduction. Annu Rev recessive retinitis pigmentosa. Nat Genet
mammalian retinal pigment epithelial cells. Physiol 2002; 64:153–187. 1997; 17:198–200.
Exp Eye Res 2006; 83:315–321. 321. Burns ME, Baylor DA: Activation, 338. Morimura H, Berson EL, Dryja TP:
302. Seiji M, Fitzpatrick TB, Birbeck MS: The deactivation, and adaptation in vertebrate Recessive mutations in the RLBP1 gene
melanosome: a distinctive subcellular photoreceptor cells. Annu Rev Neurosci encoding cellular retinaldehyde-binding
particle of mammalian melanocytes and the 2001; 24:779–805. protein in a form of retinitis punctata
site of melanogenesis. J Invest Dermatol 322. Sun H, Nathans J: Stargardt’s ABCR is albescens. Invest Ophthalmol Vis Sci 1999;
1961; 36:243–252. localized to the disc membrane of retinal 40:1000–1004.
303. Seiji M, Shimao K, Birbeck MS, Fitzpatrick rod outer segments. Nat Genet 1997; 339. Cideciyan AV, Haeseleer F, Fariss RN, et al:
TB: Subcellular localization of melanin 17:15–16. Rod and cone visual cycle consequences
biosynthesis. Ann N Y Acad Sci 1963; 323. Molday LL, Rabin AR, Molday RS: ABCR of a null mutation in the 11-cis-retinol
100:497–533. expression in foveal cone photoreceptors dehydrogenase gene in man. Vis Neurosci
304. Schmidt SY, Peisch RD: Melanin and its role in Stargardt macular dystrophy. 2000; 17:667–678.
concentration in normal human retinal Nat Genet 2000; 25:257–258. 340. Bernal S, Calaf M, Garcia-Hoyos M, et al:
pigment epithelium. Regional variation and 324. Pepperberg DR, Okajima TL, Wiggert B, Study of the involvement of the RGR,
age-related reduction. Invest Ophthalmol et al: Interphotoreceptor retinoid-binding CRPB1, and CRB1 genes in the
Vis Sci 1986; 27:1063–1067. protein (IRBP). Molecular biology and pathogenesis of autosomal recessive
305. Weiter JJ, Delori FC, Wing GL, Fitch KA: physiological role in the visual cycle of retinitis pigmentosa. J Med Genet 2003;
1654 Retinal pigment epithelial lipofuscin and rhodopsin. Mol Neurobiol 1993; 7:61–85. 40:e89.
Müller Cells and the Retinal Pigment Epithelium

341. Morimura H, Saindelle-Ribeaudeau F, blood-retina barrier by autocrine PGE2 and 358. Rezai KA, Semnani RT, Patel SC, et al: The
Berson EL, Dryja TP: Mutations in RGR, membrane-bound mechanisms. Cell immunogenic potential of human fetal
encoding a light-sensitive opsin Immunol 1993; 149:315–330. retinal pigment epithelium and its relation
homologue, in patients with retinitis 351. Tanihara H, Yoshida M, Matsumoto M, to transplantation. Invest Ophthalmol Vis
pigmentosa. Nat Genet 1999; 23:393–394. Yoshimura N: Identification of transforming Sci 1997; 38:2662–2671.
342. Marlhens F, Bareil C, Griffoin JM, et al: growth factor-beta expressed in cultured 359. Dutt K, Waldrep JC, Kaplan HJ, et al: In
Mutations in RPE65 cause Leber’s human retinal pigment epithelial cells. vitro phenotypic and functional
congenital amaurosis. Nat Genet 1997; Invest Ophthalmol Vis Sci 1993; characterization of human pigment
17:139–141. 34:413–419. epithelial cell lines. Curr Eye Res 1989;
343. Gu SM, Thompson DA, Srikumari CR, et al: 352. Holtkamp GM, de Vos AF, Kijlstra A, Peek 8:435–440.
Mutations in RPE65 cause autosomal R: Expression of multiple forms of IL-1 360. Liversidge JM, Sewell HF, Forrester JV:
recessive childhood-onset severe retinal receptor antagonist (IL-1ra) by human Human retinal pigment epithelial cells
dystrophy. Nat Genet 1997; 17:194–197. retinal pigment epithelial cells: identification differentially express MHC class II (HLA,
344. Thompson DA, Li Y, McHenry CL, et al: of a new IL-1ra exon. Eur J Immunol 1999; DP, DR and DQ) antigens in response to in
Mutations in the gene encoding lecithin 29:215–224. vitro stimulation with lymphokine or purified
retinol acyltransferase are associated with 353. Ishida K, Panjwani N, Cao Z, Streilein JW: IFN-gamma. Clin Exp Immunol 1988;
early-onset severe retinal dystrophy. Nat Participation of pigment epithelium in 73:489–494.
Genet 2001; 28:123–124. ocular immune privilege. III. Epithelia 361. Percopo CM, Hooks JJ, Shinohara T, et al:
345. Sun H, Gilbert DJ, Copeland NG, et al: cultured from iris, ciliary body, and retina Cytokine-mediated activation of a neuronal
Peropsin, a novel visual pigment-like suppress T-cell activation by partially non- retinal resident cell provokes antigen

CHAPTER 125
protein located in the apical microvilli overlapping mechanisms. Ocul Immunol presentation. J Immunol 1990;
of the retinal pigment epithelium. Inflamm 2003; 11:91–105. 145:4101–4107.
Proc Natl Acad Sci U S A 1997; 354. Kaestel CG, Jorgensen A, Nielsen M, et al: 362. Sun D, Enzmann V, Lei S, et al: Retinal
94:9893–9898. Human retinal pigment epithelial cells pigment epithelial cells activate uveitogenic
346. Streilein JW: Regional immunity and ocular inhibit proliferation and IL2R expression of T cells when they express high levels of
immune privilege. Chem Immunol 1999; activated T cells. Exp Eye Res 2002; MHC class II molecules, but inhibit T cell
73:11–38. 74:627–637. activation when they express restricted
347. Wenkel H, Streilein JW: Evidence that 355. Jorgensen A, Wiencke AK, la Cour M, et al: levels. J Neuroimmunol 2003; 144:1–8.
retinal pigment epithelium functions as an Human retinal pigment epithelial cell- 363. Acland GM, Aguirre GD, Bennett J, et al:
immune- privileged tissue. Invest induced apoptosis in activated T cells. Long-term restoration of rod and cone
Ophthalmol Vis Sci 2000; 41:3467–3473. Invest Ophthalmol Vis Sci 1998; vision by single dose rAAV-mediated gene
348. Zamiri P, Zhang Q, Streilein JW: 39:1590–1599. transfer to the retina in a canine model of
Vulnerability of allogeneic retinal pigment 356. Farrokh-Siar L, Rezai KA, Patel SC, Ernest childhood blindness. Mol Ther 2005;
epithelium to immune T-cell-mediated JT: Cryoprecipitate: an autologous 12:1072–1082.
damage in vivo and in vitro. Invest substrate for human fetal retinal pigment 364. Gullapalli VK, Khodair MA, Wang H, et al:
Ophthalmol Vis Sci 2004; 45:177–184. epithelium. Curr Eye Res 1999; 19:89–94. Retinal pigment epithelium and
349. Campochiaro PA: Cytokine production by 357. Rezai KA SR, Farrokh-Siar L, Hamann KJ, photoreceptor transplantation frontiers. In:
retinal pigmented epithelial cells. Int Rev et al: Human fetal retinal pigment epithelial Ryan SJ, ed. Retina. Philadelphia: Elsevier-
Cytol 1993; 146:75–82. cells induce apoptosis in allogenic T-cells in Mosby; 2006:2597–2613.
350. Liversidge J, McKay D, Mullen G, Forrester a fas ligand and PGE2 independent 365. Rizzolo LJ. Development and role of tight
JV: Retinal pigment epithelial cells pathway. Current Eye Research 1999; junctions in the retinal pigment epithelium.
modulate lymphocyte function at the 18:430–439. Int Rev Cytl 2007; 258:195–234.

1655
CHAPTER

126 Retinal and Choroidal Circulations


Constantin J. Pournaras and Guy Donati

The knowledge of the mechanisms underlying the pathophy-


siology of the retinal microcirculation is of fundamental clinical
VASCULAR SUPPLY OF THE RETINA
importance, since ischemic microangiopathies of the inner The retinal circulation is end-arterial without any anastomoses;
retina1–3 are the most common cause of blindness in developed the central retinal artery appears at the optic disk where it
countries.4 Impairment of the choroidal and retinal circulations divides into two major branches. These in turn divides in
results in blood flow modifications, which in turn affects the arterioles extending outward from the optic disk each supplying
delivery of oxygen and metabolic substrates, necessary for the one quadrant of the retina. Retinal arteries and veins divide by
maintenance of the energy generating processes to the retina dichotomous and site-arm branching, the terminal arterioles
tissue. The neuronal cells reach certain energy state through the come off at almost right angles from the main stream. In ~25%
formation of adenosine triphosphate (ATP), produced by means of humans a cilioretinal artery hooks around the temporal
of two fundamental metabolic pathways, namely glycolysis and margin of the optic disk and provides a portion of the macula
oxidative phosphorylation coupled to the citric acid cycle. In with the arteriolar supply.22
order to maintain an adequate ATP level in the cells, both its The larger retinal vessels lie in the inner-most portion of the
production and utilization rates have to be even. This requires retina close to the inner limiting membrane; retinal glial cells,
not only control of the activity of the glycolytic and citric acid mainly astrocytes, extend over large areas in close spatial rela-
cycle enzymes, but also adequate oxygen and glucose delivery. tionship with retinal vessels wall.23 Astrocytes constrains the
The mammalian retina possesses a high rate of glycolysis retinal vessels to the retina and maintains their integrity.24 At
and lactate production5–8 but also, like the brain an elevated rate the arteriovenous crossing sites, the deeper vessels may indent
of oxygen consumption.9–12 The above are needed for the active the retina down to the outer plexiform or outer nuclear layer.21
neuronal transport processes, which maintain the ionic The venous system has a certain similarity to the arteriolar
gradients necessary for electrical activity and visual trans- arrangement; the retinal venous blood is drained by the central
duction.13 In addition to the impairment of the energy- retinal vein that leaves the eye through the optic nerve and
dependent transport processes, the release of excitatory amino drains into the cavernous sinus. Both the terminal vessels,
acids,14,15 the rise of intracellular calcium, the disruption of namely precapillary arterioles, and the postcapillary venules are
calcium-modulated processes,16 the reperfusion-reoxygenation, linked by the interposed capillary bed. Even though there are no
oxygen-derived free readicals release,17,18 nitric oxide (NO) arteriovenous shunts in the normal retina, at the periphery of
mediated neurotoxicity19 and contribution to delayed cell the retina, the terminal arterioles and veins are linked by large
death,20 have been demonstrated as interrelated pathophy- capillary communications. Similar anastomotic capillaries
siologic pathways involved in the ischemic neuronal damage. connect the perifoveal terminal arterioles with the venules,
Developements in fluorescein and indocyanine green leaving a capillary free zone of 400–500 mm in diameter.
angiography, as techniques for non invasive measurements of The retinal arterioles give rise to a plexus of capillaries each
the retinal and choroidal blood flow, and new experimental measuring ~5 mm in diameter. These capillaries lie in an
findings on the regulation of the retinal and choroidal vascular interconnecting network, which is arranged in a basic two-
tone, improved our understanding of retinal and choroidal layered pattern. The first one resides in the nerve fiber and
circulation in health as in diseased eyes. ganglion cell layer, and the second deeper one, lies in the inner
nuclear layer. In the peripapillary area, an additional capillary
ANATOMY OF THE RETINAL AND network, lies in the superficial portion of the nerve fiber layer,
CHOROIDAL CIRCULATIONS which constitutes the radial peripapillary capillaries. It is
distributed around the optic disk and along the temporal
Metabolic substrate and oxygen delivery of the retina in higher superior and inferior retinal vessels.25 Toward the periphery, the
mammals including humans and other primates is supported deep net disappears leaving in this way a single layer of wide-
by the two separate vascular systems, the retinal and choroidal mesh capillaries. At the extreme retinal periphery an area of
circulations. In some lower mammals, as rabbits and guinea ~1.5 mm width is avascular. A capillary free zone also
pigs, the retina is almost completely dependant upon the surrounds the arterioles; it is probably due to local high oxygen
choroid, since retinal vessels can be found only in a small area tension during vascular remodelling occuring during
of the retina or can be totally lacking.21 Both retinal and maturation of the retinal vascular system.26 The outer retinal
choroidal vessels derived from the ophthalmic artery, branch of layers, including the photoreceptors, are avascular and receive,
the internal carotid, have distinctive morphological and as the peripheral avascular retinal area does, metabolic energy
functional behavior. support from the choroid (Fig. 126.1).
1657
RETINA AND VITREOUS

the equator. The retinal arteries differ from those of the same size
in other organs, in that of the unusually developed smooth muscle
layer and that they lack an internal elastic lamina. Near the optic
disk the arterial wall has five to seven layers of smooth muscle
cells, which diminish to two or three layers at the equator and one
to two at the periphery. The muscle cells are oriented both
circularly and longitudinally, each being surrounded by a
basement membrane that contains an increasing amount of
collagen toward the adventitia.27 Retinal arteriolar precapillary
annuli were observed in a number of animals at the side-arm
branches of retinal arteries, but not in humans (see Fig. 126.2).
There is no evidence that annuli contain muscular or elastic
tissue components, instead it is more likely that they are
composed of basement membrane or immature collagen.28
The capillary wall is composed of three distinct elements:
endothelial cells, intramural pericytes, and basement mem-
brane. The endothelial cells are oriented along the axis of the
capillaries; they present at their thickest areas a nucleus bulging
SECTION 3

into the lumen, and express cytoplasmic extensions which


encircle the lumen. Tight junctional complexes with con-
tinuous fusion of the outer leaflets of the cell membranes are
found along the opposing surfaces of adjacent endothelial
cells.29 Frequently the endothelial cell processes overlap, and a
lip like protrusion often marks the junction of two cells. The
continuous layer of endothelial cells is surrounded by a thick
basement membrane within which there is a discontinuous
layer of intramural pericytes in almost a one to one ratio with
FIGURE 126.1. Distribution of the retinal capillaries in a monkey the endothelial cells. Clinical and experimental observations
retina digest preparation. Note the broad capillary-free zone present suggests that pericytes contribute to the regulation of micro-
around the artery, and the absence of sphincters (insert). vascular growth and function.30
FINE STRUCTURE OF THE RETINAL
VASCULATURE VASCULAR SUPPLY OF THE CHOROID
All the branches of the main retinal arteries have the structure The choroid, a highly vascularized and pigmented layer, con-
of small arteries. According to electron microscopic studies, stitutes the posterior portion of the uvea. It is ~0.2 mm thick
these arteries are proved to have an arteriolar structure beyond posteriorly, tapering to 0.1–0.15 mm thick in the periphery. The

FIGURE 126.2. (a) Schematic representation of


capillary distribution within the inner layers of
the retina. The photoreceptor layer is avascular,
receiving oxygen and metabolic substrate
support from choroidal capillaries. ILM, internal
limiting membrane; GL, ganglion cell layer;
IPL, inner plexiform layer; OPL, outer plexiform
layer; ONL, outer nuclear layer; RPE, retinal
pigmentary epithelium. (b) Semithin section of a
human retina.

a b
1658
Retinal and Choroidal Circulations

FIGURE 126.4.
Artwork of the
equatorial choroid of a
60-year-old man.
Scleral view showing
microarchitecture of
the equatorial
choroids, scleral view.
Lobules are marked by
broken lines. A, artery;
v, vein; CH,
choriocapillaris.

CHAPTER 126
FIGURE 126.3. After scleral penetration the short posterior ciliary
arteries expand toward the periphery with a chevron configuration.
Indocyanine green angiograms showing choroidal arterial filling with a
chevron pattern.
with the vascular pattern of the arterial phase of ICG
angiography. This pattern differs from short posterior ciliary
choroid is composed of the choriocapillaris layer, the medium arteries not directed to the macular area, in that it expands in a
vessels layer (Sattler’s layer) and the outer layer of large vessels typical chevron configuration (Fig. 126.3).39
(Haller’s layer). In primates, the medium layer contains large The long posterior ciliary arteries follow long, oblique
arteries measuring 40–90 mm, large veins measuring intrascleral courses and travel in the virtual suprachoroidal
20–100 mm, nerves, and lymphatics.31,32 space, giving off recurrent branches to the macula and to the
The choriocapillaris and the medium-sized choroidal vessels anterior choroid at the ora serrata. Recurrent branches supply
are sandwiched between the apical retinal pigment epithelium choroidal areas at 3 and 9 o’clock meridians.40
(RPE) of neuroepithelial origin and the outer choroidal pigment The anterior ciliary arteries perforate the sclera at the
of neural crest origin. Two collagenous structures, one on the insertion of the tendons of the muscles and pass through the
inside (Bruch’s membrane) and one on the outside (subcapillary suprachoroidal space to enter the ciliary body. These primarily
fibrous tissue), are connected by the intercapillary pillars.31 supply the ciliary body and iris. Before joining the major arterial
Most medium-sized vessels are external to the subcapillary circle of the iris, anterior ciliary arteries are divided into 7–12
fibrous tissue.33 External to it, an anatomical cleavage plane recurrent branches that supply the anterior choroid.40,41
referred to as intervascular space, may be found between the
inner medium sized and outer large-vessel choroidal layers.31 Choriocapillaris
The functional unit of the choroidal circulation is the chorio-
Choroidal Arteries capillary lobule. The lobules measure 0.6–1.0 mm and are
The vascular supply to the choroid derives from the ophthalmic supplied by precapillary arterioles and drained by postcapillary
artery via branches of the anterior and posterior ciliary arteries. veinules.42,43 Lobules subdivide the choroid into several functional
The ophtalmic artery gives off 2–3 main ciliary arteries that is islands. Each lobule consists of a capillary meshwork with radial
the nasal, the temporal, and occasionally the paraoptic one, and circumferential arrangement, that contains an arteriole in
supplying the corresponding hemisphere of the choroid.34,35 the middle and a venule at its periphery (Fig. 126.4).44,45 Lobules
These in turn, branch into 10–20 short posterior ciliary in the equatorial part of the choriocapillaris are larger (200 mm)
arteries, that enters into the globe at the posterior pole and than those located both at the posterior pole (100 mm), and in
assume a peripapillary and perimacular pattern, the papillo- the submacular area (30–50 mm).38 The structure of the
macular oval,36 before branching peripherally in a wheel-shaped choriocapillary resembles mostly a dense network of freely con-
arrangement, and two long posterior ciliary arteries.32,33 nected capillaries in the peripapillary and submacular area. This
Secondary and tertiary branches of the short posterior ciliary pattern changes to a lobule-like arrangement in the posterior
arteries are subsequently divided into the major choroidal pole and to a palm-like organization, more peripherically.44
arteries.34,35 The short posterior ciliary arteries also contribute
paraoptic branches to the Zinn–Haller circle as well as pial Choroidal Veins
branches. They provide segmental supply to the lamina Blood discharges the lobules of the choriocapillaris by collecting
cribrosa, retro and prelaminar optic nerve head (ONH).35,37 venules that join the afferent veins. At the posterior pole, the
Some branches of these short posterior ciliary arteries are venule is located at the periphery of the lobule, stays on the
selectively directed to the macular region vessels, i.e., the very same plane of the lobule, and possibly also drains adjacents
short posterior ciliary arteries.38 Histological examination lobules.40 Small venular channels commonly connect the
showed that they have a spiral shaped configuration, consistent posterior aspect of the choriocapillaris with collecting venules. 1659
RETINA AND VITREOUS

Fine Structure of the Choriocapillaris


The capillaries of the choriocapillaris usually have a diameter
of 18–50 mm. They are flat and often have a narrow segment.48
The endothelial cells of the choriocapillaris are connected by
a discontinuous row of ‘zonulae occludens’. The part of the
choriocapillaris that faces the RPE has large fenestrations
covered by a thin membrane with a central thickening.49,50
The fenestrations are larger and more numerous in the
submacular area.51 Experimental destruction of the RPE
causes focal atrophy of the choriocapillaris, suggesting that
the localization of the fenestrae toward the RPE could be
the result of a modulating action of the RPE on the
choriocapillaris.52 These fenestrations have high permeability,
and as the analysis of suprachoroidal fluids suggests, such
permeability of the choriocapillaris is similar to that of an
isoporous membrane with a pore diameter of 144 Å.53 These
high permeability probably accounts for the maintenance of
an adequate concentration of glucose and other nutrients at
SECTION 3

the RPE level.49

Innervation of Retinal and Choroidal Vessels


Histologic studies and stimulation experiments have revealed a
rich supply of autonomic vasoactive nerves to the choroidal
vessels but not to the retina. Sympathetic nerves derived from
the superior cervical sympathetic ganglion, innervate the
FIGURE 126.5. Intermediate-size veins. Indocyanine green angiogram choroidal vascular bed, as well as the central retinal artery up to
showing a superior vortex vein. the lamina cribrosa.54,55 The choroidal vessels contain alpha-
adrenergic vasoconstrictor receptors but no beta-adrenergic
vasodilator receptors. Thus, alpha adrenergic agonists constrict
Intervenular channels between collecting venules and larger the long posterior ciliary arteries in vitro and reduce blood flow
veins, direct arteriovenous anastomosis, and interdigitation through the choroid, while beta-adrenergic agonist isopro-
between the choriocapillaris and venules were also reported in terenol has no effect.56
histological studies.40,44 Postcapillary venules are closely From this point on, although a- and b-adrenergic receptors57
arranged in the macular region. The meshwork of the venous and receptors for angiotensin58 are present, the retinal vessels
plexus becomes less dense with increasing distance from the are devoid of sympathetic fibers.55 The parasympathetic system
macula, the vessels become straighter, losing the tortuous provides vasodilating fibers to the choroid through the facial
aspect, which is characteristic of the macular region. Vessels of nerve,59 the neurotransmitter may be the vasoactive intestinal
larger lumen form the subcapillary plexus and eventually flow peptide (VIP).60–62 A neuronal NO release has been identified in
into the vortex veins.40 autonomic nerves in the choroid,63 confirming the role of
Four to six vortex veins receive the blood collected in the perivascular nerve fibers around choroidal vessels staining
ampullae of the vortex veins by the afferent ones. The vortex for NOS.64
veins are located 2.5–3 mm posterior to the equator, closer to
the vertical meridian than to the horizontal one and drain
into the superior and inferior orbital veins (Fig. 126.5).46 Some STRUCTURE OF THE BLOOD–OCULAR
drainage also occurs through the anterior ciliary veins of the BARRIERS
ciliary body. Venous drainage sems to be segmentally organized Inner Blood–Retinal Barrier
into quadrants, with watersheds oriented horizontally through The retina is a part of the brain and, as a result, retinal
the disk and fovea and vertically through the papillomacular capillaries have a similar structure to cerebral ones. As in the
area (Fig. 126.6).43,47 brain, adjacent endothelial cells are connected by a continuous

FIGURE 126.6. Indocyanine green angiogram


of early (a) and late (b) choroidal arteriovenous
phase. Note the subfoveal entry of short
posterior macular arteries, the vertical arterial
watershed area running through the papilla, and
the venous watersheds oriented horizontally
though the disk and fovea and vertically
through the papillomacular region.

a b
1660
Retinal and Choroidal Circulations

products across the blood–retinal barrier in and out of the


TABLE 126.1. Comparison between Retinal and Choroidal
retina. Physiologically Bruch’s membrane doesn’t constitute a
Circulation Blood Flow and Oxygen Extraction [82,88,95,98,99].
barrier to the movement of these molecules except in aging.
Retinal Low level of flow High O2 extraction Lipoprotein accumulation occurs in the Bruch’s membrane
circulation (15 – 34 mg/min) (40%) from rod outer segment degradation in aging that may obstruct
Choroidal High level of flow Low O2 extraction the movement of water and perhaps of waste products, from the
circulation (677 – 735 mg/min) (4%) RPE to the choroids.81

PHYSIOLOGY OF THE RETINAL AND


CHOROIDAL CIRCULATION

network of membranous ridges (tight junctions).65 Junctions TECHNIQUES OF BLOOD FLOW


between endothelial cells and pericytes also occur through MEASUREMENTS (ANIMALS/HUMANS)
fenestrations in basement membranes.66 These types of capil- In the last decades, non invasive clinical methods as digital
laries, called nonfenestrated ones, only have two kinds of small fluorescein and ICG angiography Laser Doppler Velocimetry
pores, with a diameter of 9–24 to 70 nm.65,67 The permeability (LDV) coupled to vessels diameter measurements,82 blue field
of such nonfenestrated capillaries has been estimated to be less entoptic phenomenon83 gave much information on the retinal

CHAPTER 126
than 1% of those in skeletal muscle and less than 0.1% of those blood flow modifications and regulation in normal and diseased
in the mesentery.68 In the retinal vessels, endothelial cells and eyes. In addition techniques has also been used in animal
astrocyte interactions,23,24 are of fundamental importance for models in order to obtain quantitative information on retinal
the efficient function of the inner blood–retinal barrier, as and choroidal blood flow including calorimetry84 direct
contact with glial neighboring cells is a prerequisite for the measurements from choroidal veins,85 radioactive krypton
development of tight junctions.69 desaturation,86 labeled microspheres,87,88 hydrogen clearance,89
Thus, the permeability of the retinal capillaries is similar to laser doppler flowmetry (LDF).90–92 Among all these techniques,
that of cerebral vessels with no or at the most minimal leakage injection of radioactive labeled microsphere has been widely
of fluorescein,70 or small ions like sodium.71 As in all vascular used to calculate tissue blood flow in animals.
beds, retinal capillaries are highly permeable to lipid-soluble As such techniques are not useful for humans, most of our
substances such as gases (O2, CO2). Water also diffuses rapidly knowledge about physiology of the retinal and choroidal
through the endothelial cells. However, the inner blood–retinal circulations come from animal studies, confirmed by recent
barrier is largely impermeable not only to large molecules as findings mainly using LDV and flowmetry. Computer-assisted
albumin and others proteins72 but also to small water-soluble analysis of fluorescein93 and ICG94 angiograms have been made
substances, like glucose and amino-acids. This means that to provide qualitative information about choroidal blood flow,
metabolic substrates have to be transported through the inner but they do not measure flow rates.
blood–retinal barrier by means of carrier-mediated transports
systems.
Active transport takes place either through specific channels RETINAL AND CHOROIDAL BLOOD FLOW
constituted of trans membranous proteins, i.e., intracellular A 35–80 mL/min retinal blood flow has been calculated using the
and extracellular lactate to pyruvate ratios are in near equilib- LDV and monochromatic fundus photography in humans.82–95
rium, which is established by monocarboxylate transporters,73,74 Marked regional differences in blood flow through the
or by pinocytosis through the cytoplasm of the endothelial choroid was found, as choroidal blood flow near the fovea and
cells.75 The above has been demonstrated for glucose,76 and around the ONH is much higher than the blood flow in the
amino-acids,77 the main nutrients of neuronal cells. periphery of the eye.88 Choroidal blood flow is extremely high,
~10 times higher than the flow of the gray matter of the brain
Outer Blood Retinal Barrier and four times the flow of kidney (Table 126.1).96
The retinal pigment epithelial (RPE) cells form the outer blood Even though a corresponding difference in metabolic require-
retinal barrier. RPE cells measure 16 mm in height and ments does not exist, the large rate of blood flow in the capillary
10–60 mm in diameter. The apical zonulae occludens bed of the choroid is probably owing to the large caliber of the
constitute the location of the blood–retinal barrier. The basal vascular lumen and the consequent low resistance to flow.
lamina of the RPE form the most inner layer of the Bruch’s Eighty-five percent of the total blood supply to the eye is
membrane. Inner collagen, elastic fibers, outer collagen, and distributed to the choroid and only 4% to the retina. From the
basement membrane of the choriocapillaris constitute remaining 11%, 10% is distributed to the ciliary body and 1% to
the remaining layers.48 Active transport takes place also at the the iris.87,88,96 Consequently, the oxygen extraction from the
outer blood–retinal barrier as it has been demonstrated for uveal blood is very low, the arteriovenous difference for the
glucose and amino-acids.78 choroidal blood is ~3%.97 On the other hand, in humans98 and
Several ocular diseases and surgical trauma alter the permea- pigs,99 the oxygen content of the retinal venous blood is ~38%
bility of the blood–ocular barriers. Disruption of the blood–retinal lower than the one in arterial blood. Despite the low oxygen
barrier has been demonstrated, using ocular fluorophotometry extraction from the choroidal blood, in pigs, ~60% of oxygen
in diabetes and in systemic hypertension.79,80 Proteins and and 75% of glucose are delivered by the choroidal circulation.99
other substances of high molecular weight can enter the The reason for the high volume of choroidal blood flow is not
interstitial space, due to the fenestrated nature of the choroidal completely understood. A thermoregulating action by removal
capillaries. Retinal binding protein, vitamin A, and many other of heat generated during visual transduction has been advo-
micronutrients and ions become available to the RPE for cated,100 as exposition of the eye to a moderate-intensity light
transport to the outer layers of the retina. The movement of source causes a reflexive increase in choroidal blood flow in
metabolites takes place across both the RPE and Bruch’s monkeys and humans.101 Fluid removal from the outer retina
membrane. Both active and passive transport mechanisms by the high choroidal oncotic pressure, or subserving the
facilitate the movement of selected nutrients and of waste metabolic needs of the retina has also been suggested.11 1661
RETINA AND VITREOUS

RETINAL AND CHOROIDAL OXYGEN


DISTRIBUTION
In the retina direct measurements of tissue oxygen partial
pressure (PO2) were performed in animals using oxygen
sensitive microelectrodes.102,103 The PO2 was found to be
heterogenously distributed close to the vitreoretinal interface.
Juxtaarteriolar preretinal or transretinal PO2 profiles indicates
that O2 diffusion from the retinal arterioles affects the juxtaar-
teriolar preretinal and inner retinal layers PO2. In contrast, the
preretinal and inner retinal (30% depth) PO2 far beyond the
retinal vessels remains constant in all retinal areas.104,105
Transretinal intervascular PO2 profiles, indicated that the
intraretinal PO2 gradually decreases from both the retinal
surface and the choroid toward the mid-retina with the
minimum value recorded at 50% of retinal depth.105 Close to
the pigment epithelium the PO2 is significantly higher than that
at the inner limiting membrane level. This is probably due to
SECTION 3

much higher O2 delivery by the choroidal circulation than by


the retinal one and the very low arteriovenous oxygen
a
difference.97 These PO2 profiles indicate that there is O2
diffusion from the inner retina and the choroid toward the
middle of the retina (Fig. 126.7). The above comes in
accordance with previous theoretical calculations,106 and
measurement obtained in other species.12,107,108

REGULATION OF THE RETINAL AND


CHOROIDAL BLOOD FLOW
The general concept of the regulation of blood flow in a vascular
bed is that systemic factors (circulating hormones and the
autonomic nervous system) regulate the distribution of the
b
cardiac output over the different vascular beds as a function of
the hemodynamic situation of the whole body and that local FIGURE 126.7. (a) Intervascular transretinal PO2 profiles recorded in
factors (such as PO2, PCO2, pH, metabolic products) try to miniature pigs during normoxia (each point is the mean + SE, N = 13).
adapt flow to local needs. Minimal PO2 value is reached at 50% retinal depth. ILM, inner limiting
The blood flow through the eye is directly related to the mean membrane; RPE, retinal pigment epithelium; (b) The drawing indicates
perfusion pressure (PPm), and inversely related to the vascular the pathway of the microelectrode through the retina.
resistance (Rm) within the ocular vascular bed.
Rm further depends on blood viscosity (n), the length (L)
and diameter (2r) of the blood vessels (cf. Law of Poiseuille:
R = 8 nL/r4). Thus, the blood flow F = DP pr4/8 nL, is related Viscosity
to the PPm and the diameter of the resistance vessel, and inver- Blood viscosity varies as a function of the shear rate, being high
sely related to the vascular blood viscocity and length of the at low shear rate. Blood viscosity diminishes and becomes
vascular bed. almost constant at a high shear rate.113 An increase in plasma
viscosity (e.g., hyperglobulinemias, high Ht, leukemia, sickle
The PPm cell anemia), substantially influences the flow through the
Is determined as the local arterial blood pressure minus the retinal circulation. Slowdown of the retinal perfusion may lead
venous pressure. As the venous pressure almost equals to stasis in the retinal veins and ultimately to venous occlusion.
intraocular pressure, PPm is defined as the difference between Retinal perfusion could be normalized by correction of the
the mean ophthalmic artery blood pressure (MOAP) minus the hyperviscosity.114
intraocular pressure (IOP).The MOAP, was assumed to be 2/3 of
the mean arterial blood pressure (MABP), and was calculated as Length of the Vascular Bed
MABP = 2/3 [BPdiast + 1/3 (BPsyst - BPdiast)]. BPdiast and BPsyst are Changes in the total length of the vascular bed can be modified
the brachial artery blood pressures during diastole and systole, by precapillary sphincters lying at the bifurcations of the arte-
respectively. rioles, which causes the opening and closing of the capillary
The retinal blood flow is autoregulated (i.e., maintained à bed. Although such a sphincter is seen in many tissues, it is not
constant values) to a mean systemic blood pressure to 41% up found in choroidal or retinal arterioles28; in contrast to the
to baseline values.109 Similarly, autoregulation occurs during an capillaries in the pulmonary circulation, all of which can be
increase of IOP up to 27–30 mmHg, which results in a mean mobilized or not, depending on the circulatory needs.
retinal perfusion pressure decrease of less than 50%.110 As a
result, the inner retinal-tissue partial pressure of O2 (PO2) is The Diameter of Vessels
maintained at constant values during moderate reductions of Is determined by the contractile state of the arteriolar smooth
the perfusion pressure.111,112 The Rm vascular resistance is muscle and the capillaries pericytes. Numerous factors
related to (1) the blood viscosity, (2) length, and (3) the diameter influence the vascular resistance such as systemic factors
of the blood vessel. Any change of each of those parameters including autonomic nerves, circulating substances and
1662 influence retinal blood flow. systemic PaO2 and PCO2, local metabolic factors including
Retinal and Choroidal Circulations

substances released by the vascular endotheliun and/or the Hypercapnia induces an increase in retinal blood flow through
neuroglial cells surrounding the vessels which get involved in a mechanism involving either neuronal NO synthase (NOS-I)130
the moment to moment regulation of the distribution of the or PGE2-mediated endothelial NO synthase (NOS-III) release.131
cardiac output depending on the metabolic needs of the tissue. Metabolic acidosis induced by intravenous acetazolamide
injection produces an increase of preretinal and optic disk
Systemic factors PO2,132 suggesting that acetazolamide increases retinal blood
Innervation The eye has a rich autonomic innervation only to flow, as observed in the case of cerebral blood flow,133 probably
the uvea and the extraocular parts of the retinal blood vessels. by increasing the PaCO2.134 This PaCO2 increase is due to
Sympathetic nerves reach the eye from the sympathetic cervical significant bicarbonate loss in the renal tubules, resulting in
superior ganglion, while parasympathetic nerves reach the hyperchloremic metabolic acidosis. The CO2 produced by the
eye through the oculomotor nerve, the facial nerve and through cells cannot be eliminated by carbonic anhydrase and so
the ophthalmic and maxillary division of the trigeminal nerve. increases the PaCO2 by diffusing through the basement
The choroidal vessels contain alpha-adrenergic vasocon membrane.135 The PaCO2 increase and pH decrease induced by
strictor receptors A considerable number of perivascular nerve acetazolamide are not affected either by hyperventilation or by
fibers around choroidal vessels stain for NOS forming a dense bicarbonate intravenous infusion.128
network of choroidal ganglion cells; 64 such distibution is
found only in humans and higher monkeys which have a fovea Circulating substances The influence of circulating molecules
centralis. Thus the choroidal circulation appears to be under the and hormones on the retinal and choroidal circulation is rather

CHAPTER 126
influence of basal release of NO released either by the vascular unclear.
endothelium or the nitrergic choroidal nerve terminals. High levels of angiotensin-converting enzyme136 and recep-
tors for angiotensin II58 are found in retinal and choroidal blood
Arterial oxygen partial pressure (PaO2) Hyperoxia (100% vessels, suggesting that angiotensin II might play a role in the
oxygen breathing) induces a marked vasoconstriction of the inner regulation of ocular circulation. Studies of the effect of
retinal arterioles in both normal humans,115,116 and angiotensin II on the ocular circulation, however, have yielded
anesthetized animals.104,117 By the autoregulatory vasocon- controversial results. In contrast to previous studies, more
striction in the inner retina, PO2 is maintained at constant recent ones indicated no evidence that angiotensin II contract
values during systemic hyperoxia.104,105 The responses of the retinal arteries,137 or choroidal and optic nerve blood flow.138,139
retinal circulation to changes in arterial O2 are similar to those Similarly controversial results have been reported after
of the cerebral circulation,118 however, the retinal blood flow administration of adrenergic drugs, a decrease, an increase or no
decrease in response to hyperoxia is quantitatively more effect on the retinal circulation has been described.139
important.104,116 Whatever the effects of these molecules on the ocular
In humans and anesthetized animals, hypoxia (decrease in circulation, none seems to have a direct contribution to the
PO2 in the arterial blood) induces vasodilation of retinal adaptations of the choroidal and retinal circulation to the
arterioles117,119 which can be clearly measured only under an varying metabolic needs of the retina.
arterial PO2 (PaO2) of 65 mmHg. A similar observation was
observed in cerebral blood-flow studies.118,120 Local factors
Transretinal PO2, profiles made during variations in PaO2, by These factors may be either ionic or molecular or related to gas
steps of 10 mmHg between 120 and 30 mmHg have shown that modifications under physiological stimuli or pathological
the PO2 values measured in the inner-retina up to half of its conditions. Interactions between substances released either by
thickness remained rather stable during different steps of the vascular endothelial cells or by neuroglial or neuronal tissue
hypoxia. In contrast, the PO2, measured near the choroid and in surrounding the retinal arterioles i.e., relaxing factors as NO,
the outer-retina decreased in a linear manner according to the prostacyclin (PGI2), lactate or contracting factors such as
variations of systemic PaO2.121–123 A decrease in PO2 near the endothelin-1 (ET-1), Angiotensin II, cyclooxygenase (COX)
photoreceptor–pigment epithelium complex during systemic products such as thromboxane-A2 (TXA2) and prostaglandin H2
hypoxia is expected to lower the ATP supply to the rods and, (PGH2), should affect the arteriolar tone, thus regulating the
thus, inhibit the Na+/K+ pump, and inducing a rapid alteration retinal vasomotor responses.140–142
of light-induced response of the RPE.124 Photoreceptors are The endothelium located between the circulating blood and
apparently more vulnerable to steps of hypoxia, indicating that the vascular smooth muscle cells, regulates permeability, affect
high oxygen delivery by the choroidal circulation, is necessary coagulation, platelet function, and fibrinolysis but it also exerts
to maintain mitochondrial respiration in photoreceptors and, metabolic functions by activating and inactivating hormones.
as a result, their normal function. The choroidal circulation is In addition, vasoactive substances that either inhibit (i.e.,
not influenced by hyperoxia possibly because of increased NO endothelium-derived relaxing factors: EDRF) or activate (i.e.,
synthesis.125 endothelium-derived contracting factors: EDCF) the underlying
smooth muscle cells can also be released by endothelial cells.
Arterial PaCO2 Changes in the partial pressure of arteriolar
CO2 (PaCO2) affect retinal blood flow. The sensitivity of the Role of NO Since the initial observation indicating that
retina to variations of PaCO2 is such that a PaCO2 rise of acetylcholine-induced vasodilation is dependent on the
1 mmHg induces a 3% rise in blood flow.126 There is a tight presence or absence of the vascular endothelium,143 from
parallelism between changes of PaO2 and changes of interstitial numerous experiments it became obvious that the endothelium
pH in the inner retina; variations of systemic PaCO2 (DPaCO2 produces a vasodilator, which was initially defined as EDRF. NO
= 3 7.6 ± 6 mmHg) induce a decrease in pH in the retina of released by endothelial cells accounts for the biological
0.158 + 0.025 units. Acidification of the blood by infusing HCl properties of the EDRF.143–147
or by injecting lactate127 affects neither interstitial pH nor NO is a nonpolar gas soluble in tissues, freely diffusible
retinal blood flow, suggesting that interstitial acidosis and not across membranes, synthetized by the enzyme nitric oxide
systemic acidosis might be a step in the induction of vasomotor synthase (NOS) from the oxidation of L-arginine and formation
response in hypercapnia,128 a finding which confirms those of L-citrulline.144,148–150 Previously the NO synthases were
reported in the cerebral cortex.129 classified as inducible and constitutive, but recent experiments 1663
RETINA AND VITREOUS

FIGURE 126.8. Putative NO metabolic


pathway in the retina. NO is constitutively
released by both neuronal cells and
endothelium in the retina and acts on the
vascular smooth muscle.
SECTION 3

have demonstrated that all NOS isoforms are regulated


dynamically. Three isoforms of NOS are classified as neuronal
NOS (NOS1), endothelial NOS (NOS3), and NOS (NOS2).
The isoform NOS-I has been found in neurons of the central
and peripheral nervous system,151 and in the retina of
mammals, in ganglion, amacrine, horizontal, Müller glial cells,
and photoreceptors,147,150–154 and human retina.155 The NOS-III
is mainly expressed by the vascular endothelium cells including
the retinal,156,157 and choroidal157 vessels endothelial cells and
the endothelial cells and pericytes of the retinal capillaries (Fig.
126.8).158
NO is produced when NOS-I and NOS-III are activated via
the calcium/calmodulin complex. Preretinal NO gradient from
the vitreal surface of the retina toward the vitreous indicates a a
continuous release of NO by the retinal tissue (Fig. 126.9), and,
since the NO gradient is also recorded far from visible arterioles,
this confirms that cells in the retina, other than endothelial
cells, may release NO.159 Juxta-arteriolar vitreal microinjections
of nitro-L-arginine (L-NA), a non specific inhibitor of NOS,
inducing segmental vasoconstriction159 suggest that a
continuous production of NO is necessary in order to maintain
a dilating arteriolar tone in the inner retina.
Experimental evidence suggests that in the ocular circulation
NO also plays a role, since relaxation of isolated ophthalmic
and ciliary artery rings induced by acetylcholine is markedly
reduced by the NO synthase inhibitor NG monomethyl-L-
arginine (L-NMMA) nitro-L-arginine.160,161 NO also induces
ralaxation of the contracting tone of the retinal bovine pericytes
via a cGMP dependnt mechanism.162 In addition, endothelium-
derived NO release under basal conditions or stimulated by
bradykinin regulates the ophthalmic circulation of the perfused
porcine eye.163
Finally, red blood cells (RBCs) provide a novel NO mediated b
vasodilator activity in which hemoglobin acts as an O2 sensor
FIGURE 126.9. Intervascular preretinal (NO) profile: (a) Continuous
and O2-responsive NO signal transducer, thereby regulating
NO recording obtained as the NO microprobe was advanced toward
both peripheral and pulmonary vascular tone. In blood, NO the inner retinal surface (rising phase) and then retracted 200 µm into
binds to cysteine-b-93 forming S-nitrosohemoglobin. Deoxy- the vitreous (falling phase). Current was maximal at the retinal surface,
genation is accompanied by allosteric transition in S- that portion of recording between the inner retinal surface (0 µm) and
nitrosohemoglobin that releases the NO group. The NO 200 µm out in the vitreous. (b) Values are means + SEM of nine
1664 released from the Hb may be transferred directly to the vascular recordings of the type shown in (a).159
Retinal and Choroidal Circulations

endothelium. In that way, S-nitrosohemoglobin contracts the the retina is due to the oxidation of glucose to CO2.6 Systemic
blood vessels and decreases perfusion in tissues with high O2 hypoxia induces an increase in the retinal lactate release,7
affinity and relaxes the vessels to improve blood flow in which potentially affects the arteriolar tone as preretinal
structures with low O2 affinity.164,165 microiniections (30–100 nL) of L-lactate (0.5 mol, pH 7.4) close
to the retinal arterioles locally dilate the arteriolar wall.127
Role of prostaglandins In the cerebral circulation, the effects Since retinal metabolism produces significant amounts of L-
of different subclasses of prostaglandins (PGs) have been largely lactic acid which crosses cell membranes, it is not yet clear
studied under physiological and pathological conditions.166–168 whether lactic acid microinjected into the preretinal vitreous
Studies on isolated cerebral vessels,169 isolated pericytes of exerts its effect on the endothelial cells or on the smooth
retinal capillaries170 and incubated rabbit retinas171 have shown muscle cells by interfering with the metabolism of cells
that some subclasses of PGs (PGE2, PGF2, PGI2) may be surrounding the arterioles. Indeed, lactate could be transported
synthesized from their precursor, the arachidonic acid. PGI2 was from the vitreous side of the vascular wall by means of a specific
shown to exert a vasodilating action on isolated bovine retinal transporter,193 which affects endothelial cell metabolism, and
arterioles,172 and acts as a vasodilator in rabbit eyes.173 thus induces endothelial release of vasoactive substances.
Microinjections of PGE2 induced a segmental vasodilation119 of Intravenous administration of sodium lactate increases
the retinal arterioles. retinal bood flow.194 Since intracellular and extracellular lactate
Perfusion of the eye either through the sublingual artery or to pyruvate ratios are in near equilibrium, which is established
microinjections close to the retinal arterioles by inhibitors of by monocarboxylate transporters,73,74 intravenously injected

CHAPTER 126
PG synthase (indomethacin) produced a transitory reversible lactate could be transported affecting the endothelial cell
vasoconstricton of the retinal arterioles in normoxia/ metabolism, inducing endothelial release of vasoactive
normocapnia as well as inhibition of the retinal vasodilation substances (i.e., NO) or could exert its effect on the smooth
normally induced by hypercapnia,119 in contrast did not affect muscle cells by interfering with the metabolism of cells
the hypoxia nor lactate-induced vasodilation.127 Thus, surrounding the arterioles. Indeed, the increase of the lactate-
vasodilater PG release, as in the brain,174 should be a possible to-pyruvate ratio leads to reduction of the NADH-to-NAD+
mechanism maintaining either the arteriolar tone in normo- ratio which in turn increases the intracellular NADH
capnia or of hypercapnia-induced arteriolar vasodilation, but concentration,195 inducing an increase of retinal blood flow in
the hypoxia and lactate induce vasodilation through a PG stimulated retina.196,197
independent pathway. PGE2 and PGF2 are the predominant PGs
produced by the retina and choroid and potentially plays a role
during physiological adaptations, such as hypercapnia and AUTOREGULATION OF THE RETINAL BLOOD
autoregulation.175,176 FLOW
The autoregulation of the blood flow in a tissue vascular bed is
Endothelins Are a family of three 21-amino-acid peptides defined as the ability of the tissue to maintain its blood flow
secreted by a variety of cells in the eye.177 Three members of the relatively constant despite moderate variations of perfusion
ET family have been identified so far: ET-1, ET-2, and ET-3,178 pressure.198,199 This can be achieved to the some extent, by
and two subtypes of receptors with different sensibilities to the changing the vascular resistance. This occurs through changes
three isoforms. of the contractile state of the arterioles and possibly of the
ETA receptor, which is expressed on vascular smooth muscle capillary pericytes. Changes in vessel tone have been observed
cells and pericytes,179 is characterized by a very high affinity to both as a physiologic adaptation and in pathologic conditions.
ET-1. ET-1 is the most potent vasoconstricting substance As retinal vessels are not sympathetically innervated, the
known and has been shown to induce vasoconstriction after mechanism which underlies the autoregulation of retinal blood
intravitreal injection in rabbit,180 cat,181 and rat retinal flow is the balanced result of a myogenic component and
arterioles.182 ET-1 affects smooth muscle cells and pericytes,141 metabolic component i.e., interactions between factors release
and contributes to induced retinal vasoconstriction in human by the retinal metabolism and the vascular endothelium.
retinal arterioles in normoxia and hyperoxia.183,184
Two types of ETB receptors have been identified. The ETB1 Myogenic Mechanism
receptor, which is expressed in endothelial cells, has equal By means of a myogenic mechanism, with the increase or
affinity for each ET isoform and mediates vasorelaxation decrease of the PPm, the arterioles constrict or dilate respec-
through release of NO in the pulmonary circulation of lambs tively, in order to regulate the blood flow to constant values (i.e.,
under physiological conditions185 or under hypoxia.186 The autoregulation). However, regulation becomes ineffective when
activation of ETB1 receptors elicits also the release of NO in the the perfusion pressure rises or falls beyond certain limits.
rat187 and rabbit kidney.188 The variation in the vascular transmural pressure difference
ETB receptors have been identified in cultured bovine retinal serves as the stimulus for a myogenic mechanism. This
pericytes179 and endothelial cells,186,189 suggesting a similar happens by changing the vascular resistance during moderate
vasodilating effect through NO release in the retina. ETB modifications in perfusion pressure, a mechanism which
blockage induces a sustained hypertension in conscious resembles that of kidney and brain. Pacemakers cells in the
nonhuman primates, which is mediated by ETA receptors. arteriolar wall may modify the arteriolar tone and consequently,
These data suggest that ETB1 receptors might influence arterial adapt the vascular resistance.96
blood pressure homeostasis by reducing plasma ET-1 levels and Endothelium dependent regulation of ophthalmic arteries
minimizing ETA activation.190 The ETB2 receptor has a high tone during quick strech200–202 and a contracting endothelium-
affinity for ET-3 and mediates direct vasoconstriction.191 derived factor203 released during increase in transmural pressure
seems to be other components of the myogenic autoregulation.
Role of lactate The mammalian retina has been found to Ion channels in endothelial cells may act as mechano-
possess an unusually high rate of glycolysis: ~70% of the transducers, accounting for the apparent capacity of the
total glucose utilized by the retina is converted to lactate.7 endothelium to sense mechanical forces and respond to
Lactate released by isolated Müller cells is metabolized by them.204 Indeed, L-type voltage-gated calcium channels are
photoreceptors.192 In addition, 70% of oxygen consumption in involved in transforming the stretch of the vascular smooth 1665
RETINA AND VITREOUS

muscle cells induced by an increase of the intraluminal pressure mechanism during the flight or fight response, as well as during
into a contraction.205 However, depolarization-independent moderate hypertension.209,227 Loss of sympathetic innervation
mechanisms on the vascular smooth muscle seem to be may cause an almost linear choroidal perfusion pressure-flow
involved, as after depolarization of the vascular smooth muscle relationships227 leading to accumulation of fluid in the retina
by K+ (120 mM) a pressure-induced contraction is still and retinal edema. This is important in diseases such as
observed.206 diabetes and hypertension, in which autonomic control is
altered.
The Metabolic Control Decreases in mean arterial pressure with the IOP are held
The metabolic control of the retina is defined as the effect of constant, even if inducing a decrease in the perfusion pressure,
factors released by tissue metabolism, which strives to optimize do not affect choroidal blood flow, which is maintained by a
the blood flow according to the metabolic needs of the tissue. myogenic mechanism at constant values.228
The retinal circulation adapts very well to changes in retinal However, a lack of choroidal autoregulation and linear
metabolism. During flicker stimulation the increased choroidal perfusion pressure–flow relationships have been
metabolism of retinal tissue207,208 is coupled to an increase in obtained in studies where the perfusion pressure gradient was
retinal and ONH perfusion.209–211 decreased by raising the venous pressure with ramp or step
In cerebral212 and ocular circulation, NO is in part increases in intraocular pressure.88,143,229 Due to lack of choroidal
responsible for the functional vasodilation. A role for NO was autoregulation, during increments of intraocular pressure, the
also established in neurovascular coupling in the eye as diffuse PO2 in the choroid and the outer retina decreases.112
SECTION 3

flicker light stimulation induces an increase of the retinal213 and Electrical stimulation of the parasympathetic vasodilator
ONH blood flow, associated with a transient increase of NO.214 fibers, provided to the choroid through the facial nerve, results
The retinal blood flow flicker response is blunted by NOS in marked vasodilation in the uvea and increased blood
inhibition.215 In healthy young subjects, the contribution of flow.230,231 In rabbits NO at low frequences61 and VIP released
basal NO release on retinal vascular tone, was investigated at high frequences,62 may be the transmmitters for the effect of
using Zeiss retinal vessel analyzer. L-NMMA significantly the stimulation of the facial nerve. The physiological
reduced arterial diameter (3 mg/kg: –2%) and venous diameter importance of the parasympathetic system in the regulation of
(3 mg/kg: –5%). In addition, the flicker induced vasodilation of the choroidal blood flow, though, is yet to be defined.
the human retinal vasculature was significantly attenuated.216
In dogs, inhibition of NO release by intravenous perfusion of Role of NO
L-NMMA induces a 40% decrease in the choroidal blood flow A neuronal NO release has been identified in autonomic nerves
without any significant modifications of the retinal blood in the choroid,63 confirming the role of perivascular nerve fibers
flow.217 Perfusion of the eye either through the sublingual artery around choroidal vessels staining for NOS.64 Studies in
by inhibitors of PG synthase (indomethacin) produced a tran- rabbits232 showed evidences for a continuous neuronal NO-
sient reversible vasoconstricton of the retinal arterioles in induced vasodilator tone in the choroid.
normoxia/normocapnia127 suggesting that in the retina, Several animal experiments indicate that NO plays an
vasodilating substances release non affected by PGs inhibitors important role in the maintenance of basal vascular tone in the
i.e., NO release, adapt the vascular tone, leading to regulation choroid. Deussen and co-workers first established NO as a
of retinal blood flow. major determinant of uveal blood flow using intravenous
Interaction between the two mediators has been provided infusion L-NMMA in dogs and the radioactive microsphere
previously in other preparations218–222 could be a possible technique. Despite an increase in mean arterial pressure of
mechanism affecting either the arteriolar tone in normocapnia 20mmHg the authors observed a significant decrease in
or the hypercapnia-induced retinal arteriolar vasodilation.223 In choroidal blood flow (–40%), ciliary body blood flow (–40%) and
addition, endothelium-derived NO has been reported to be a iris blood flow (–48%) and a slight reduction in retinal blood
vasodilating mediator in response to hypoxia in retinal vessels flow (–12%).217 These experiments were confirmed in cats by
in cats.224 using LDF, where L-NA reduced choroidal blood flow in a dose-
dependent fashion despite an increase in systemic blood
pressure.233 The observation that NO is a major determinant of
AUTOREGULATION OF THE CHOROIDAL choroidal blood flow was confirmed in a variety of studies using
BLOOD FLOW the radioactive microsphere technique and LDF in various
Autonomic Regulation of the Choroidal Blood species.
Flow In humans, L-NMMA induced a fundus pulsation amplitude
The choroidal vasculature is richly innervated by vasoactive (FPA) reduction, indicating that the choroid is particularly
nerves. Stimulation of the cervical sympathetic chain in sensitive to changes in NO release. L-NMMA induced a slight
monkeys had no effect on blood flow through either the retina increase in systemic blood pressure and reduced NO content in
or ONH,225 in contrast to the marked reduction of the choroidal the exhaled air by ~50%234 indicating that L-NMMA is capable
blood flow in monkeys and cats.225,226 The choroidal vessels of reducing endogenous NO production. Similar findings in
contain alpha-adrenergic vasoconstrictor receptors but no beta- humans where the choroidal perfusion was assessed with LDF,
adrenergic vasodilator receptors. Thus, alpha-adrenergic indicated a dose-dependent reduction of the choroidal blood
agonists constrict the long posterior ciliary arteries in vitro and flow parameter after administration of L-NMMA.235 In
reduce blood flow through the choroid, while beta-adrenergic addition, endothelium-derived NO has been reported to be a
agonist iso-proterenol has no effect.56 vasodilating mediator in response to marked hypoxia in the
Choroidal blood flow changes very little during sudden forearm resistant vessels in healthy humans,236 in interaction
increments in blood pressure, as an increased sympathetic with vasodilating PGs.237,238 These results clearly indicate that
activity and consequent vasoconstriction of the choroidal NO is physiologically released in human choroidal vessels.
vessels maintain choroidal blood flow constant.92,227 The However the reduction of endogenous NO production by the l-
choroidal blood flow seems to be regulated by the autonomic NMMA does not mean that NO production from NOS-1 and/or
nervous system by alpha adrenergic receptors located at the NOS-3 was inhibited by the same extent in the choroid. An
1666 choroidal vessels. This control represents a protective interaction between NO and ET1 in the choroid has been
Retinal and Choroidal Circulations

speculated, as ET-1 causes vasoconstriction of choroidal blood sodium leads to the activation of the Na+/K+-ATPase which
vessels when injected intravenously in cats only when the triggers aerobic glycolysis and lactate production. Once lactate
release of NO was inhibited.239 In fact the effect of ET1 on a is released, it can be transformed by neurons into pyruvate and
vascular bed apart from causing vasoconstriction, it can also enter the TCA cycle to serve as an energy fuel. One molecule of
release both the vasodilators PGI2 and NO.240 lactate entering the TCA cycle can yield in normoxic conditions
17 ATPs.260
PHYSIOPATHOLOGY OF THE RETINAL AND Since glutamate uptake is electrogenic and strongly
CHOROIDAL CIRCULATION dependent on trans-membrane Na+ influx, ischemia, by
causing a decrease in ATP concentration in the cell and, in turn,
inhibition of the Na+ pump, or high extracellular K+ conditions,
RETINAL CIRCULATION impairs the uptake of glutamate in Müller cells or astrocytes.261
Acute retinal ischemia following experimental occlusion of the Intravitreal measurement demonstrates a significant glutamate
retinal arterioles, induces drop of the inner retinal PO2 leading increase in the rabbit vitreous following ischemia–reperfusion
to anoxic damage of the inner retinal cells.123,241,242 Retinal induced by large increases of the intraocular pressure.262,263
areas affected by acute branch vein occlusion (BRVO) reveal also However, measurements done before and following
a inner retinal tissue hypoxia,132,243,244 as oxygen diffusing from experimental BRVO failed to demonstrate a glutamate increase
the choroid does not reach the inner retina. Accordingly, a following the occlusion (personal findings). We cannot exclude
hypoxic damage of the neuronal cells of the inner retina. has that artifacts induced by various amino-acid modifications in

CHAPTER 126
been confirmed by histological data.245,246 Tissue hypoxia is the vitreous after the occlusion (like the observed preretinal
probably secondary to a decrease of blood flow in the affected vitreous exudation from the occluded vein) were not responsible
retinal areas, secondary to myogenic vasoconstriction of the for altered measurements. Yet, measurement of glutamate
retinal arterioles or a decrease of NO release.247 release in the retina and the cerebral cortex by a microdialysis
During systemic hyperoxia, O2 diffusing from the choroid electrode perfused with L-glutamate oxidase, as well as the
supplies also the inner ischemic retina affected by an acute recording of the current evoked between two voltage-clamped
BRVO.123 A decrease of PO2 of the damaged inner retinal electrodes, indicated differences between retina and cerebral
neuronal layers, enables oxygen to diffuse from the choroid and cortex glutamate release following ischemia/reperfusion. More
increases the inner retinal PO2. In contrast, 100% O2 breathing specifically, ischemia was reported to induce in the retina a
fails to produce an increase in PO2 in the center of fresh anoxic decrease of preretinal glutamate release, in contrast to the
postarteriolar embolism foci.248 Thus hyperoxia could be a increase induced in the cerebral cortex. Glutamate release
useful way to prevent hypoxic inner retinal layers damage, in changes were reversible following reperfusion.264
eyes with vein occlusion. Thus, in vivo data are lacking with regard to the extracellular
Carbogen breathing significantly increased preretinal PO2 in glutamate concentration and glutamate-mediated neurotoxicity
normal and in ischemic postexperimental BRVO areas of mini- in acute ischemia/hypoxia induced by BRVO. That knowledge
pigs. The concomitant use of acetazolamide injection and would be crucial for the application of neuroprotective
carbogen breathing or hyperoxia could restore an appropriate treatments.
oxygenation of BRVO areas. Acetazolamide induced an increase
of the preretinal PO2 to a greater extent when it was associated Preretinal L-lactate measurements
with carbogen breathing than when it was combined with Preretinal lactate release was measured, lactate-sensitive
hyperoxia.132,249 microelectrodes, following experimental BRVO. Following a
However, this finding conflicts with previous data obtained in steady state measurements, an experimental BRVO 30 min
humans.249,250 Indeed, the ischemic retina, even if adequately after the occlusion induced a significant decrease in preretinal
supplied with oxygen, still requires a supply of other lactate (83.4 ± 10.0%, n = 10, p <0.01) compared with the
metabolites, such as glucose, to maintain a normal function. values measured before BRVO. One hour after BRVO, a 76.4 ±
12.9% decrease in preretinal lactate (n = 10, p < 0.01) was
Ischemia-Induced Retinal Metabolic Changes measured. Two hours later, this decrease was 58 ± 20.0% (n =
Glutamate uptake 9, p <0.01). Four hours after BRVO, the preretinal lactate was
Electrophysiological and pharmacological evidence support the not further decreased (58 ± 11.3%, n = 5, p < 0.01).265 Those
notion that glutamate is the main excitatory neurotransmitter results suggest that acute BRVO increases the venous blood
in various regions of the brain and the vertebrate retina.251–255 pressure upstream to the occlusion and strongly decreases the
In the retina, it can be electrogenically taken up by neuroglial capillary blood flow and thus the flux of glucose from the
Müller cells and astrocytes.256–258 In astrocytes, glutamate is plasma to Müller cells. Impaired glycolysis could lead to a
predominantly converted to glutamine through an ATP- decrease in preretinal lactate as measured in our experiments.
requiring reaction catalyzed by the astrocytes specific glutamine Perturbation of glycolysis, worsened by hypoxia, could provoke
synthase. Glutamine released by astrocytes is taken up by retinal metabolism modifications leading to cellular dys-
neurons to replenish the neurotransmitter pool of glutamate.259 function and death.
This uptake has physiological significance since there is no
extracellular enzyme to degrade glutamate. Therefore, some The role of NO
cells have to take up glutamate in order to facilitate its clearance The role of NO in the pathophysiology of focal ischemia in the
by diffusion from the synaptic clefts and to maintain synaptic central nervous system is complex, since both modifications in
function. constitutive and induced NO release are implicated. Previous
Glutamate transport into astrocytes triggers aerobic studies showed that NO could protect brain tissue during focal
glycolysis in the glial cells, utilization of glucose and lactate ischemia266 by acting on blood vessels and platelets,149 whereas
production.258,259 Glutamate-stimulated increase in glucose it could increase ischemic damage by mediating neurotoxicity19
uptake and phosphorylation in the astrocytes is abolished in the and by contributing to delayed cell death.20
absence of sodium in the extracellular medium, which is Neurotoxicity has been reported to be associated with NO by
consistent with the necessity of an electrochemical gradient for excessive stimulation of the NMDA receptors.267 Postsynaptic
the ion to drive glutamate uptake. The intracellular increase of NMDA-receptor activation could lead to an intracellular 1667
RETINA AND VITREOUS

FIGURE 126.10. (a) Semithin section through


the vitreoretinal interface over an ischemic
retinal area (R) shows a new vessel lying on the
modified vitreoretinal interface. L, vessel lumen;
ILM, internal limiting membrane. (b) Electron
microscopy shows an interendothelial junction
of a new vessel with nonfenestrated
endothelium that had grown intravitreally.
E, endothelial cell. (c) Electon microscopy of a
fenestrated (arrow) endothelial cell of a new
b vessel lying intravitreally in experimental
vasoproliferative microangiopathy in miniature
pigs. V, vitreous.
SECTION 3

a c

increase in NO production, which in turn would cause PDR278 and animals following experimental branch vein
nitrosylation of thiols (inhibition of GAPDH), DNA nitration, occlusion.279 Preretinal tissue hypoxia was measured at the
oxidation of intracellular protein sulfhydrils, and inhibition of ischemic areas as probably the oxygen which diffuses from both
mitochondrial respiration by binding to iron-sulfur com- the large retinal vessels and the choroid, does not reach the
plexes.19 NO can also act as a second messenger on neighboring ischemic inner retinal territories.280–282 Eyes with ischemic
cells, leading to an excessive increase in cGMP, which is an microangiopathy complicated by neovascularization have poor
intracellular effector regulating many vital cell functions, such clinical prognosis as a result of the abnormal structure and
as the transduction of light signals in retinal photoreceptor function of the new vessels.283–285 The clinical appearance and
cells.268 the fine structure of new vessels, in experimental ischemic
NO can also elicit an indirect neurotoxicity via formation of microangiopathy,281 are similar to those observed in human
peroxynitrite (ONOO–), which decomposes to another reactive eyes with vasoproliferative microangiopathy. The new vessels
oxygen species related to the hydroxy free radical (OH-) and to are composed by continuous-type, endothelial cells, surrounded
the radical nitrogen dioxide (NO2), which is a potent activator by a basal lamina and pericytes. The intercellular junctions of
of lipid peroxidation. This form of toxicity is mainly related to the endothelial cells were scarce, fusion plates are observed
late inflammatory reaction and/or reperfusion of an injured occasionally and exceptionally the endothelial cell fenestrations
area.19,269,270 (Fig. 126.10).
Because of the complex role played by NO and its potential Neovascularization, that occurs in ischemic/hypoxic retinal
implication in both neurotoxicity and free radicals toxicity, areas support the hypothesis that it is tissue hypoxia that
experiments aiming at suppressing or increase NO release have triggers neovascularization. Tissue hypoxia indirectly affects the
been conducted in the past years. It has been shown that new vessels growth by the release of growth modulators by the
selective inhibition of inducible NO synthase by amino- endothelial cells and modyfing the response of the endothelial
guanidine diminishes the cerebral ischemic damage in a rat cells to growth modulators (probably via receptor upregulation).
model,271 whereas supplementation of NO by the NO-donor An increase in hypoxia-inducible factor-1 (HIF-1) expression
nitroprusside can improve blood flow and reduce brain damage has been shown to be correlated with vascular endothelial
after focal ischemia.272 Constitutive NO release is probably growth factor (VEGF) expression both during normal and
impaired in the first hours after the onset of ischemia in the pathologic retinal vascularization in mouse which confirms the
mammalian brain; increasing local NO availability during the essential role of hypoxia in the regulation of VEGF expression
first hours following the onset of ischemia could improve blood in the retina.286
flow and be neuroprotective.273 In addition, in retinal or nonocular microvascular endothelial
In the aorta and cerebral vessels from hypercholesterolemic cells cultures, the release of factors as insulin growth factor-I
rabbits with impaired endothelium-dependent relaxation, L- (IGF-I),287 platelet-derived growth factor (PDGF),288 VEGF,289
arginine restores the vasodilator response to achety- tissue plasminogen activator (t-PA) and its inhibitor PAI-I
lcholine.274,275 In pulmonary arteries and cultured endothelial (factors which modulate extracellular matrix turnover),290,291
cells, depletion of L-arginine causes a loss of endothelium- increases under steps of hypoxia. Moreover basic fibroblast
dependent relaxation that is reversible with addition of growth factor (bFGF), acts as a potent stimulator of endothelial
extracellular L-arginine.276 In humans, the effect of NOS cells in culture to form capillary-like tubules,292 and induces
inhibition by bolus infusion of 3 mg/kg of L-NMMA over 5 min cellular growth if added to hypoxic cultures of aortic capillary
on the renal vasculature can be reversed by simultaneous endothelial cells.293
infusion of L-arginine (17 mg kg–1 min–1 over 30 min).277 The production of the VEGF, known to induce angiogenesis
In the long term evolution of ischemic microangiopathies, as in ischemic regions of tumors,289,294 was detected in retinas
retinal vein occlusion or diabetic microangiopathy, the with ischemic microangiopathy295–297 and in the vitreous of
hemodynamic modifications on the retinal vascular bed leads to human eyes with proliferative diabetic retinopathy.298–300
the formation of ischemic areas, where the blood flow Hypoxia is considered a functional stimulus for VGEF expres-
1668 decreases, as it was measured by LDV in diabetic patients with sion in the ischemic regions of tumors and retina.289,301,302
Retinal and Choroidal Circulations

In ischemic retinas of adult primates, VEGF gene expression circumscribed pigmented lesions the so called Elschnig’s spots.
was reduced by systemic hyperoxia which reverses the retinal A number of systemic diseases as systemic hypertension,
tissue hypoxia.282 toxemia of pregnancy, disseminated intravascular coagulopathy,
acute posterior multifocal placoid pigment epitheliopathy, giant
cell arteritis, Goodpasture’s syndrome, and sickle cell disease
CHOROIDAL CIRCULATION induces this pattern of choroidal lesions.309
Choroidal Pressure Gradients Occlusion of larger choroidal arteries, such as short posterior
Even though there is marked permeability of the chorio- ciliary ones, causes triangular areas of chorioretinal damages
capillaris (as discussed earlier), very little fluid flows into or out much smaller than expected.307 Moreover, ischemic lesions of
from the extravascular space. Colloid osmotic pressure gradient the choroid tend to resolve rapidly over several days, much
across the choriocapillary is 8–9 mmHg toward the lumen of faster than expected by recanalization of a single terminal
the vessels resulting in a tendency to absorb fluid into the arteriole.310,311 Similar observations have been reported first by
choriocapillaris.303 The hydrostatic pressure in the chorio- Amalric after large sectorial choroidal ischemia caused by
capillaris is 7–8 mmHg above the intraocular pressure.304 Thus temporal arteritis or carotid obstruction.312 Thus, it seems that
the hydrostatic pressure gradient across the walls of the chorio- if the RPE and the outer retina can survive even under marked
capillaris is about the same as the colloid osmotic pressure, reductions in choroidal blood flow.
resulting in little or no net flow between the choroidal vascular The anatomically observed anastomotic channels which
bed and the extravascular space. Any change in the hydrostatic connects the lobules together and do not seem to contribute to

CHAPTER 126
or colloid-osmotic pressures of the choroid would disturb the the pattern of blood flow in normal states, may play a role in the
Starling equilibrium causing a net filtration or absorption of regulation of flow, when the blood flow or perfusion pressure is
choroidal extravascular fluid. A marked increase in the hydro- markedly reduced in adjacent lobules as this occurs during
static pressure causing spontaneous choroidal detachment has ischemia.309,313 A partial late filling from episcleral and pial
been reported in patients with arteriovenous fistulas of the arteries, as retrograde filling from the vortex veins have also
cavernous sinus.305 A sudden reduction in IOP brings about the been advocated.314,315 Complete abolition of choroidal blood
same effect, as transient choroidal detachments are common supply would destroy both the RPE and the photo-
after intraocular surgery.306 receptors.316,317 Occlusion of the vortex veins without arterial
occlusion causes venous stasis, with a generalized breakdown of
Choroidal Blood Flow and Aging the blood aqueous barrier. Occlusion of two or more vortex
It was proposed that reduced blood flow through sclerotic veins results in hyphema, forward movement of the iris-lens
choroidal arteries would reduce perfusion mainly in the peri- diaphragm, and increased intraocular pressure.47
phery (watershed area) of the area supplied by one short
posterior ciliary artery. The submacular area is a meeting place SUMMARY
for many such watershed zones, which could explain the
tendency of the macula to undergo degenerative changes. There The vascular tone of the resistant vessels (arterioles, capillaries)
is evidence that when light is focused on the macula, it causes in the eye is modulated by the interaction of multiple
a much higher rise in temperature, whenever choroidal blood mechanisms affecting the arteriolar smooth muscle and the
flow is reduced.101 Dilated choroidal arteries in the submacular vascular pericytes. The retinal blood flow is autoregulated by
area were observed on choroidal angiography in about half the the interaction of myogenic and metabolic mechanisms
patients suffering of age related macular degeneration.81 through the release of vasoactive substances by the retinal
Although, degeneration of the RPE and Bruch’s membrane tissue surrounding the arteriolar wall and/or the vascular
alterations seems to be crucial alterations in age related macular endothelium. Mechanical stretch and increases in arteriolar
degeneration, they are unlike to be due to reduced choroidal transmural pressure evoke the release of contracting factors by
blood flow. Indeed, considering the enormous flow rate through the endothelial cells; NO or lactate, released during neuronal
the choroid, the nutrition of these tissues should not be at risk. activity and energy-generating reactions of the retina, strive to
optimize blood flow according to the metabolic needs of retinal
Choroidal Vascular Occlusions tissue. A close interaction between PG and NO metabolic
The existence of anatomic connections between the lobules in pathways indicates that, when one system is inhibited, the
the choriocapillaris (as discussed earlier) suggests that ischemic other could rapidly compensate for the deficient system, and
lesions caused by vascular occlusions are rare in the choroid. maintain constant blood flow. The interaction of those
However, induced choroidal ischemia tends to have a segmental metabolic pathways is probably implicated in the control of the
geographic pattern not consistent with alternative flow through vasomotion and autoregulation in the inner retina. Therefore, it
rich anastomotic channels. Choroidal blood flow is functionally appears that the impairment of structure and function of the
segmented, and the precapillary arterioles act as end- retinal neuronal tissue and endothelium is the mechanism of
arterioles.35,307,308 The occlusion of a single end arteriole in the retinal blood abnormal regulation observed during the evolution
choriocapillaries lobule leads to the development of small of ischemic microangiopathies.

REFERENCES
1. Group DRSR: Preliminary report of effects in branch vein occlusion. Arch Ophthalmol 5. Cohen LH, Noell WK: Glucose catabolism
of photocoagulation therapy. Am J 1986; 104:34–41. of rabbit retina before and after
Ophthalmol 1976; 81:383–396. 4. Leibowitz HMK, Maunder DE, Milton LR, development of visual function.
2. Group DRSR: Photocoagulation treatment et al: The Framingham Eye Study J Neurochem 1960; 5:253–276.
of proliferative diabetic retinopathy: clinical monograph: an ophthalmological and 6. Winkler BS: Glycolytic and oxidative
application of diabetic retinopathy study epidemiological study of cataract, metabolism in relation to retinal function.
(DRS) findings. DRS report number 8. glaucoma, diabetic retinopathy, macular J Gen Physiol 1981; 77:667–692.
Ophtalmology 1981; 88:583–600. degeneration, and visual acuity in a 7. Winkler BS: The intermediary metabolism of
3. Group BVOS: Argon laser scatter general population of 2631 adults, the retina: biochemical and functional
photocoagulation for prevention of 1973–1975. Surv Ophthalmol 1980; aspects. Am Acad Ophtalmol 1983; 77: 1669
neovascularization and vitreous hemorrhage 24(Suppl):335–610. 227–242.
RETINA AND VITREOUS

8. Graymore CN, ed: Biochemistry of the and their role in the permeability of the 50. Spitznas M, Reale E: Fracture faces of
retina. In: Biochemistry of the eye. London: blood-retinal barrier. Exp Eye Res 1966; fenestrations and junctions of endothelial
Academic Press; 1970:645–735. 5:229–234. cells in human choroidal vessels. Invest
9. Kimble EA, Svoboda RA, Ostroy SE: 30. Hirschi KK, D’Amore PA: Pericytes in the Ophthalmol 1975; 14:98–107.
Oxygen consumption and ATP changes of microvasculature. Cardiovasc Res 1996; 51. Sugita A, Hamasaki M, Higashi R: Regional
the vertebrate photoreceptor. Exp Eye Res 32:687–698. difference in fenestration of choroidal
1980; 31:271–288. 31. Krebs W, Krebs IP: Quantitative capillaries in Japanese monkey eye. Jpn J
10. Weiter JJ, Zuckerman R: The influence of morphology of the central fovea in the Ophthalmol 1982; 26:47–52.
the photoreceptor-RPE complex on the primate retina. Am J Anat 1989; 52. Korte GE, Reppucci V, Henkind P: RPE
inner retina. An explanation for the 184:225–236. destruction causes choriocapillary atrophy.
beneficial effects of photocoagulation. 32. Sattler H: Uber den feineren Bau der Invest Ophthalmol Vis Sci 1984;
Ophthalmology 1980; 87:1133–1139. choroidea des Mensche. Beitragen zur 25:1135–1145.
11. Tsacopoulos M: Physiopathology of the pathologischen un vergleichenden 53. Chylack LT Jr, Bellows AR: Molecular
uveal circulation. J Fr Ophtalmol 1979; Anatomie des Aderhaut. Von Graefe’s Arch sieving in suprachoroidal fluid formation in
2:134–142. 1976; 428–440. man. Invest Ophthalmol Vis Sci 1978;
12. Linsenmeier RA: Effects of light and 33. Nuel IP: De la vascularisation de la 17:420–427.
darkness on oxygen distribution and choroide et de la nutrition de la rétine 54. Ehinger B: Adrenergic nerves to the eye
consumption in the cat retina. J Gen principalement au niveau de la fovea and to related structures in man and
Physiol 1986; 88:521–542. centralis. Arch Ophthalmol 1992; 70–87. cynomolgus monkey. Invest Ophthalmol Vis
13. Penn RD, Hagins WA: Kinetics of the 34. Weiter JJE, JT: Anatomy of the choroidal Sci 1966; 5:42–52.
photocurrent of retinal rods. Biophys J vasculature. Am J Ophthalmol 1974; 55. Laties AM: Central retinal artery
SECTION 3

1972; 12:1073–1094. 78:583–590. innervation. Absence of adrenergic


14. Rothman SM, Olney JW: Glutamate and 35. Hayreh SS: Segmental nature of the innervation to the intraocular branches.
the pathophysiology of hypoxic–ischemic choroidal vasculature. Br J Ophthalmol Arch Ophthalmol 1967; 77:405–409.
brain damage. Ann Neurol 1986; 1975; 59:631–648. 56. Dalske HF: Pharmacological reactivity of
19:105–111. 36. Shubert HD: Topographic anatomy of the isolated ciliary arteries. Invest Ophthalmol
15. Choi DW: Cerebral hypoxia: some new central retina and segmental choroidal 1974; 13:389–392.
approaches and unanswered questions. circulation. In: Yannuzzi LA, Slatker JS, 57. Denis P. Elena PP: Retinal vascular beta-
J Neurosci 1990; 10:2493–2501. eds. Indocyanine green angiography. St adrenergic receptors in man.
16. Stys PKR, Waxman BR, Davis SG, et al: Role Louis: Mosby; 1997:18–23. Ophtalmologie 1989; 3:62–64.
of extracellular calcium in anoxic injury of 37. Hayreh SS: Physiological anatomy of the 58. Ferrari-Dileo GD, Anderson EB, DR:
mammalian central white matter. Proc Natl choroidal vascular bed. Int Ophthalmol Angiotensin binding sites in bovine and
Acad Sci USA 1990; 87:4212–4216. 1983; 6:85–93. human retinal blood vessels. Invest
17. McCord JM: Oxygen-derived free radicals 38. Carella E, Carella G: Microangioarchitecture Ophthalmol Vis Sci 1987; 28:1747–1751.
in postischemic tissue injury. N Engl J Med of the choroidal circulation using latex 59. Ruskell GL: Facial parasympathetic
1985; 312:159–163. casts. In: Yannuzzi LA, Slatker JS, eds. innervation of the choroidal blood vessels
18. Traystman RJ, Kirsch JR, Koehler RC: Indocyanine green angiography. St Louis: in monkeys. Exp Eye Res 1971;
Oxygen radical mechanisms of brain injury Mosby; 1997:24–28. 12:166–172.
following ischemia and reperfusion. J Appl 39. Hayreh SS: Submacular choroidal vascular 60. Stone RAT, Tervo T, Tarkkanen K, A:
Physiol 1991; 71:1185–1195. pattern. Experimental fluorescein fundus Vasoactive intestinal polypeptide-like
19. Dalkara TM: The complex role of nitric angiographic studies. Albrecht Von Graefes immunoreactive nerves to the human eye.
oxide in the pathophysiology of focal Arch Klin Exp Ophthalmol 1974; Acta Ophthalmol (Copenh) 1986;
cerebral ischemia. Brain Pathol 1994; 192:181–196. 64:12–18.
4:49–57. 40. Hayreh SS: The long posterior ciliary 61. Nilsson: Nitric oxide as a mediator of
20. Dawson VL, DT, London ED, Bredt DS, arteries. An experimental study. Albrecht parasympathetic vasodilation in ocular and
Snyder SH: Nitric oxide mediates Von Graefes Arch Klin Exp Ophthalmol extraocular tissues in the rabbit. Invest
glutamate neurotoxicity in primary cortical 1974; 192:197–213. Ophthalmol Vis Sci 1996; 37:2110–2119.
culture. Proc Natl Acad Sci USA 1991; 41. Heimann K: Development of blood vessels 62. Nilsson: Vasoactive intestinal paptide (VIP):
88:6368–6371. in the macular–choroidal zone. Klin effects on the eye and on the regional
21. Wise GN, Dollery CT, Henkind P: The retinal Monatsbl Augenheilkd 1970; 157:636–642. blood flow. Acta Physiol Scand 1984;
circulation. New York: Harper and Row; 42. Torczynski E, Tso MO: The architecture of 121:385–392.
1975:20–82. the choriocapillaris at the posterior pole. 63. Bredt DS, Hwang PM, Snyder SH:
22. Hayreh SS: The cilio-retinal arteries. Br J Am J Ophthalmol 1976; 81:428–440. Localization of nitric oxide synthase
Ophthalmol 1963; 47:71–89. 43. Fryczkowski AW, Sherman MD, Walker J: indicating a neural role for nitric oxide.
23. Rungger-Brandle EM, Niemeyer JM, Observations on the lobular organization of Nature 1990; 347:768–770.
Eppenberger G, HM: Confocal microscopy the human choriocapillaris. Int Ophthalmol 64. Lötjen-Drecoll E: Funktionelle Morphologie
and computer-assisted image 1991; 15:109–120. der Aderhaut mit šberraschungen. 1998.
reconstruction of astrocytes in the 44. Hayreh SS: The choriocapillaris. Albrecht 65. Büngard M: The three-dimensional
mammalian retina. Eur J Neurosci 1993; Von Graefes Arch Klin Exp Ophthalmol organisation of tight jonctions in a capillary
5:1093–1106. 1974; 192:165–179. endothelium revealed by serial-section
24. Zhang Y, Stone J: Role of astrocytes in the 45. Fryczkowski AW: Topographic anatomy of microscopy. J Ultrastruct Res 1984;
control of developing retinal vessels. Invest the central retina and segmental choroidal 88:1–17.
Ophthalmol Vis Sci 1997; 38:1653–1666. circulation. In: Yannuzzi LA, Slatker JS, 66. Carlson EC: Fenestrated subendothelial
25. Henkind P, De Oliveira LF: New eds. Indocyanine green angiography. basement membranes in human retinal
observations on the radial peripapillary St Louis: Mosby; 1997:29–34. capillaries. Invest Ophthalmol Vis Sci 1989;
capillaries. Invest Ophthalmol Vis Sci 1967; 46. TorczynskiE: Choroid and suprachoroid. 30:1923–1932.
6:103–108. Biomedical foundations of ophthalmology. 67. Grotte G: Passage of dextran molecules
26. Henkind P, De Oliveira LF: Development of In: Duane TD, ed. Philadelphia: JB across the blood-lymph barrier. Acta Chir
the retina vessels in the rat. Invest Lippincott; 1987. Scand 1956; 211:1–84.
Ophthalmol Vis Sci 1967; 6:520–523. 47. Hayreh SS, Baines JA: Occlusion of the 68. Crone C: The Malpighi lecture. From
27. Hogan MJ, Feeney L: The ultrastructure of vortex veins. An experimental study. Br J ‘Porositates carnis’ to cellular
the retinal blood vessels. I. The large Ophthalmol 1973; 57:217–238. microcirculation. Int J Microcirc Clin Exp
vessels. J Ultrastruct Res 1963; 39:10–28. 48. Hogan MJ, Alvarado JA, Weddel JE: 1987; 6:101–122.
28. Henkind P, De Oliveira LF: Retinal arteriolar Histology of the human eye. Philadelphia: 69. Neuhaus J, Risau W, Wolburg H: Induction
annuli. Invest Ophthalmol 1968; 7:584–591. Saunders Publishers; 1971:369. of blood-brain barrier characteristics in
29. Shakib M, Cunha-Vaz JG: Studies on the 49. Bill A, Tornquist P, Alm A: Permeability of bovine brain endothelial cells by rat
permeability of the blood-retinal barrier. IV. the intraocular blood vessels. Trans astroglial cells in transfilter coculture. Ann
1670 Junctional complexes of the retinal vessels Ophthalmol Soc UK 1980; 100:332–336. N Y Acad Sci 1991; 633:578–580.
Retinal and Choroidal Circulations

70. Grayson MCL, AM: Ocular localization of study with radioactively labelled 106. Dollery CT, Bulpitt CJ, Kohner EM: Oxygen
sodium fluorescein. Effects of microspheres including flow determinations supply to the retina from the retinal and
administration in rabbit and monkey. Arch in brain and some other tissues. Exp Eye choroidal circulations at normal and
Ophthalmol 1971; 85:600–603. Res 1973; 15:15–29. increased arterial oxygen tensions. Invest
71. Tornquist P, Alm A, Bill A: Studies on ocular 89. Yu DYA, Cringle VA, Brown SJ, MJ: Ophthalmol 1969; 8:588–594.
blood flow and retinal capillary permeability Choroidal blood flow measured in the dog 107. Alder VAC, Constable SJ, IJ: The retinal
to sodium in pigs. Acta Physiol Scand eye in vivo and in vitro by local hydrogen oxygen profile in cats. Invest Ophthalmol
1979; 106:343–350. clearance polarography: validation of a Vis Sci 1983; 24:30–36.
72. Pappenheimer JR: Passage of molecules technique and response to raised 108. Ahmed JB, Dunn RD, Linsenmeier R Jr, RA:
through capillary walls. Physiol Rev 1953; intraocular pressure. Exp Eye Res 1988; Oxygen distribution in the macaque retina.
33:387–423. 46:289–303. Invest Ophthalmol Vis Sci 1993; 34:516–521.
73. Gerhart DZ, Leino RL: Distribution of 90. Gherezghiher T, Okubo H, Koss MC: 109. Robinson FR, Grunwald CE, Petrig JE, et
monocarboxylate transporters MCT1 and Choroidal and ciliary body blood flow al: Retinal blood flow autoregulation in
MCT2 in rat retina. 1999; 92:367–375. analysis: application of laser Doppler response to an acute increase in blood
74. Poole RC: Transport of lactate and other flowmetry in experimental animals. Exp Eye pressure. Invest Ophthalmol Vis Sci 1986;
monocarboxylates across mammalian Res 1991; 53:151–156. 27:722–726.
plasma membranes. Am J Physiol 1993; 91. Riva CEC, Mann SD, Barnes RM, GE: Local 110. Riva CE, Sinclair SH, Grunwald JE:
264:761–782. choroidal blood flow in the cat by laser Autoregulation of retinal circulation in
75. Palade GE, Simionescu M, Simionescu N: Doppler flowmetry. Invest Ophthalmol Vis response to decrease of perfusion
Structural aspects of the permeability of Sci 1994; 35:608–618. pressure. Invest Ophthalmol Vis Sci 1981;

CHAPTER 126
the microvascular endothelium. Acta 92. Riva CET, Hero P, Movaffaghy M, et al: 21(1 Pt 1):34–38.
Physiol Scand Suppl 1979; 463:11–32. Choroidal blood flow during isometric 111. Alm A, Bill A: The oxygen supply to the
76. Alm A, Tornquist P: The uptake index exercises. Invest Ophthalmol Vis Sci 1997; retina. I. Effects of changes in intraocular
method applied to studies on the blood- 38:2338–2343. and arterial blood pressures, and arterial P
retinal barrier. I. A methodological study. 93. Klein GJB, Flower RH, RW: An image O2 and P CO2 on the oxygen tension in the
Acta Physiol Scand 1981; 113:73–79. processing approach to characterizing vitreous body of the cat. Acta Physiol
77. Tornquist P, Alm A: Carrier-mediated choroidal blood flow. Invest Ophthalmol Vis Scand 1972; 84:261–274.
transport of amino acids through the Sci 1990; 31:629–637. 112. Yancey CM, Linsenmeier RA: Oxygen
blood-retinal and the blood-brain barriers. 94. Flower RW: Extraction of choriocapillaris distribution and consumption in the cat
Graefes Arch Clin Exp Ophthalmol 1986; hemodynamic data from ICG fluorescence retina at increased intraocular pressure.
224:21–25. angiograms. Invest Ophthalmol Vis Sci Invest Ophthalmol Vis Sci 1989;
78. Miller S, Steinberg RH: Transport of taurine, 1993; 34:2720–2729. 30:600–611.
L-methionine and 3-o-methyl-D-glucose 95. Feke GTT, Deupree H, Goger DM, et al: 113. Stoltz JF, DM: New trends in clinical
across frog retinal pigment epithelium. Exp Blood flow in the normal human retina. hemorheology: an introduction to the
Eye Res 1976; 23:177–189. Invest Ophthalmol Vis Sci 1989; concept of the hemorheological profile.
79. Krogsaa BL-A, Mehlsen H, Sestoft JL: 30:58–65. Schweiz Med Wochenschr 1991;
Blood-retinal barrier permeability versus 96. Alm A, Bill A: Ocular circulation. In: Moses 43(Suppl):41–49.
diabetes duration and retinal morphology in RA, ed. Adler’s physiology of the eye. 114. Knabben H, WS, Remky A, et al: Retinal
insulin dependent diabetic patients. Acta St Louis: Mosby; 1987. hemodynamics in patients with
Ophthalmol (Copenh) 1987; 65:686–692. 97. Alm A, Bill A: Blood flow and oxygen hyperviscosity syndrome Klin Monatsbl
80. Krogsaa BL-A, Parving H, Bjaeldager HH, extraction in the cat uvea at normal and Augenheilkd 1995; 206:152–156.
P: The blood-retinal barrier permeability in high intraocular pressures. Acta Physiol 115. Hickam JB, Frayser R: Studies of the retinal
essential hypertension. Acta Ophthalmol Scand 1970; 80:19–28. circulation in man: observation on vessel
(Copenh) 1983; 61:541–544. 98. Hickam JB, Frayser R, Ross JC: A study of diameter, arteriovenous oxygen difference
81. Foulds WS: The choroidal circulation and retinal venous blood oxygen saturation in and mean circulation time. Circulation
retinal metabolism – an overview. Eye human subjects by photographic means. 1966; 33:302–316.
1990; 4(Pt 2):243–248. Circulation 1963; 27:375–385. 116. Riva CE, Grunwald JE, Sinclair SH: Laser
82. Riva CEG, Sinclair JE, Petrig SH, BL: Blood 99. Tornquist P, Alm A: Retinal and choroidal Doppler Velocimetry study of the effect of
velocity and volumetric flow rate in human contribution to retinal metabolism in vivo. pure oxygen breathing on retinal blood
retinal vessels. Invest Ophthalmol Vis Sci A study in pigs. Acta Physiol Scand 1979; flow. Invest Ophthalmol Vis Sci 1983;
1985; 26:1124–1132. 106:351–357. 24:47–51.
83. Riva CE, Petrig B: Blue field entoptic 100. Parver LM: Temperature modulating action 117. Eperon G, Johnson M, David NJ: The effect
phenomenon and blood velocity in the of choroidal blood flow. Eye 1991; 5(Pt of arterial PO2 on relative retinal blood flow
retinal capillaries. J Opt Soc Am 1980; 2):181–185. in monkeys. Invest Ophthalmol 1975;
70:1234–1238. 101. Parver LM, Auker C, Carpenter DO: 14:342–352.
84. Bill A: Calorimetric procedures for the study Choroidal blood flow as a heat dissipating 118. Kety SS, Schmidt CE: Effect of altered
of the blood flow through the ciliary region mechanism in the macula. Am J arterial tension of carbon dioxide and
and the choroid in rabbits. Acta Ophthalmol 1980; 89:641–646. oxygen on cerebral blood flow on normal
Ophthalmol (Copenh) 1962; 40:131–148. 102. Tsacopoulos M, Lehmenkuhler A: A young man. J Clin Invest 1948;
85. Bill A: Intraocular pressure and blood flow double-barrelled Pt-microelectrode for 27:484–492.
through the uvea. Arch Ophthalmol 1962; simultaneous measurement of PO2 and 119. Pournaras C, Tsacopoulos M, Chapuis P:
67:336–348. bioelectrical activity in excitable tissues. Studies on the role of prostaglandins in the
86. Friedman E, KH, Smith TR: Retinal and Experientia 1977; 33:1337–1338. regulation of retinal blood flow. Exp Eye
choroidal blood flow determined with 103. Tsacopoulos M, Poitry S, Borsellino A: Res 1978; 26:687–697.
krypton-85 in anesthetized animals. Invest Diffusion and consumption of oxygen in the 120. Kogure KS, Reinmuth P, Fujishima OM,
Ophtalmol Vis Sci 1964; 3:539. superfused retina of the drone (Apis et al: Mechanisms of cerebral
87. Alm A, Bill A: The oxygen supply to the mellifera) in darkness. J Gen Physiol 1981; vasodilatation in hypoxia. J Appl Physiol
retina. II. Effects of high intraocular 77:601–628. 1970; 29:223–229.
pressure and of increased arterial carbon 104. Riva CE, Pournaras CJ, Tsacopoulos M: 121. Linsenmeier RA, Braun RD: Oxygen
dioxide tension on uveal and retinal blood Regulation of local oxygen tension and distribution and consumption in the cat
flow in cats. A study with radioactively blood flow in the inner retina during retina during normoxia and hypoxemia.
labelled microspheres including flow hyperoxia. J Appl Physiol 1986; J Gen Physiol 1992; 99:177–197.
determinations in brain and some other 61:592–598. 122. Moret PP, Munoz CJ, Brazitikos JL, et al:
tissues. Acta Physiol Scand 1972; 105. Pournaras CJR, Tsacopoulos CE, Profile of pO2. I. Profile of transretinal pO2
84:306–319. Strommer M, K: Diffusion of O2 in the retina in hypoxia. Klin Monatsbl Augenheilkd
88. Alm A, Bill A: Ocular and optic nerve blood of anesthetized miniature pigs in normoxia 1992; 200:498–499.
flow at normal and increased intraocular and hyperoxia. Exp Eye Res 1989; 123. Pournaras CJ, Ilic J, Gilodi N: The
pressures in monkeys (Macaca irus): a 49:347–360. physiopathology of retinal circulation: 1671
RETINA AND VITREOUS

consequences of acute retinal vascular the ocular circulation: a new concept in 160. Haefliger IOF, Luscher J, TF: Nitric oxide
occlusion. Klin Monatsbl Augenheilkd 1985; ophthalmology? Surv Ophthalmol 1994; and endothelin-1 are important regulators
186:471–476. 39:123–132. of human ophthalmic artery. Invest
124. Linsenmeier RA, Steinberg RH: Effects of 142. Brown SM, Jampol LM: New concepts of Ophthalmol Vis Sci 1992; 33:2340–2343.
hypoxia on potassium homeostasis and regulation of retinal vessel tone. Arch 161. Haefliger IOF, Luscher J: Heterogeneity of
pigment epithelial cells in the cat retina. Ophthalmol 1996; 114:199–204. endothelium-dependent regulation in
J Gen Physiol 1984; 84:945–970. 143. Furchgott RF, Zawadzki JV: The obligatory ophthalmic and ciliary arteries. Invest
125. Hardy PP, Lahaie KG, Isabelle V, et al: role of endothelial cells in the relaxation of Ophthalmol Vis Sci 1993; 34:1722–1730.
Increased nitric oxide synthesis and action arterial smooth muscle by acetylcholine. 162. Haefliger IO, Zschauer A: Relaxation of
preclude choroidal vasoconstriction to Nature 1980; 288:373–376. retinal pericyte contractile tone through the
hyperoxia in newborn pigs. Circ Res 1996; 144. Palmer RM, Ferrige AG, Moncada S: Nitric nitric oxide-cyclic guanosine
79:504–511. oxide release accounts for the biological monophosphate pathway 1994;
126. Tsacopoulos M, David NJ: The effect of activity of endothelium-derived relaxing 35:991–997.
arterial PCO2 on relative retinal blood flow factor. Nature 1987; 327:524–526. 163. Meyer PF, Luscher J: Endothelium–
in monkeys. Invest Ophthalmol 1973; 145. Ignarro LJB, Wood GM, Byrns KS, et al: dependent regulation of the ophthalmic
12:335–347. Endothelium-derived relaxing factor microcirculation in the perfused porcine
127. Brazitikos PDP, Munoz CJ, Tsacopoulos JL, produced and released from artery and eye: role of nitric oxide and endothelins.
M: Microinjection of L-lactate in the vein is nitric oxide. Proc Natl Acad Sci USA Invest Ophthalmol Vis Sci 1993;
preretinal vitreous induces segmental 1987; 84:9265–9269. 34:3614–3621.
vasodilation in the inner retina of miniature 146. Feelisch MtP, Zamora M, Deussen R, et al: 164. Stamler JS, JL, Eu JP, et al: Blood flow
pigs. Invest Ophthalmol Vis Sci 1993; Understanding the controversy over the regulation by S-Nitrosohemoglobin in the
SECTION 3

34:1744–1752. identity of EDRF. Nature 1994; 368:62–65. Physiological Oxygen Gradient. Science
128. Tsacopoulos M, Levy S: Intraretinal acid- 147. Koch KWL, Haberecht HG, Redburn M, 1997; 276:2034–2037.
base studies using pH glass et al: Functional coupling of a 165. Singel DJ, SJ: Chemical physiology of
microelectrodes: effect of respiratory and Ca2+/calmodulin-dependent nitric oxide blood flow regulation by red blood cells:
metabolic acidosis and alkalosis on inner- synthase and a soluble guanylyl cyclase in the role of nitric oxide and S-
retinal pH. Exp Eye Res 1976; 23:495–504. vertebrate photoreceptor cells. EMBO J nitrosohemoglobin. Annu Rev Physiol 2005;
129. Harper AM: Physiology of cerebral blood 1994; 13:3312–3320. 67:99–145.
flow. Br J Anaesth 1965; 37:225–325. 148. Ignarro LJ: Biosynthesis and metabolism of 166. Pickard JDM, MacKenzie LA, Harper ET:
130. Sato E, STNT, Mori F, et al: Role of nitric endothelium–derived nitric oxide. Ann Rev Response of the cerebral circulation in
oxide in regulation of retinal blood flow Pharmacol Toxicol 1990; 30:535–560. baboons to changing perfusion pressure
during hypercapnia in cats. Invest 149. Moncada S, Palmer RM, Higgs EA: Nitric after indomethacin. Circ Res 1977;
Ophthalmol Vis Sci 2003; 44:4947–4953. oxide: physiology, pathophysiology, and 40:198–203.
131. Checchin D, HX, Hardy P, et al: PGE(2)- pharmacology. Pharmacol Rev 1991; 167. Ellis EF, Wei EP, Kontos HA: Vasodilation of
mediated eNOS induction in prolonged 43:109–142. cat cerebral arterioles by prostaglandins
hypercapnia. Invest Ophthalmol Vis Sci 150. Venturini CM, Knowles RG, Palmer RM, D2, E2, G2, and I2. Am J Physiol 1979;
2002; 43:1558–1566. Moncada S: Synthesis of nitric oxide in the 237:H381–H385.
132. Pournaras JAP, Munoz IK, Pournaras JL, bovine retina. Biochem Biophys Res 168. Kontos HAW, Ellis EP, Dietrich EF, et al:
CJ: Experimental retinal vein occlusion: Commun 1991; 180:920–925. Prostaglandins in physiological and in
effect of acetazolamide and carbogen 151. Dawson TMB, Fotuhi DS, Hwang M, et al: certain pathological responses of the
(95% O2/5% CO2) on preretinal PO2. Invest Nitric oxide synthase and neuronal NADPH cerebral circulation. Fed Proc 1981;
Ophthalmol Vis Sci 2004; 45:3669–3677. diaphorase are identical in brain and 40:2326–2330.
133. Vorstrup S, HI, Paulson OB: Effect of peripheral tissues. Proc Natl Acad Sci USA 169. Hagen AA, White RP, Robertson JT:
acetazolamide on cerebral blood flow and 1991; 88:7797–7801. Synthesis of prostaglandins and
cerebral metabolic rate for oxygen. J Clin 152. Goureau OH, Courtois D, De Kozak Y, Y: thromboxane B2 by cerebral arteries.
Invest 1984; 74:1634–1639. Induction and regulation of nitric oxide Stroke 1979; 10:306–309.
134. Taki K, KH, Endo S, et al: Cascade of synthase in retinal Muller glial cells. 170. Hudes GRL, Rockey WY, White JH:
acetazolamide-induced vasodilatation. Res J Neurochem 1994; 63:310–317. Prostacyclin is the major prostaglandin
Commun Mol Pathol Pharmacol 1999; 153. Yamamoto RB, Snyder DS, Stone SH, RA: synthesized by bovine retinal capillary
103:240–248. The localization of nitric oxide synthase in pericytes in culture. Invest Ophthalmol Vis
135. Harlan JM: Diuretics agents. In: Katzung the rat eye and related cranial ganglia. Sci 1988; 29:1511–1516.
BG, ed. Basic and clinical pharmacology. Neuroscience 1993; 54:189–200. 171. Preud’homme Y, Demolle D, Boeynaems
7th edn. 1998:246–249. 154. Osborne NN, Barnett NL, Herrera AJ: JM: Metabolism of arachidonic acid in
136. Ferrari-Dileo G, RJ, Rockwood EJ, et al: NADPH diaphorase localization and nitric rabbit iris and retina. Invest Ophthalmol Vis
Angiotensin-converting enzyme in bovine, oxide synthetase activity in the retina and Sci 1985; 26:1336–1342.
feline, and human ocular tissues. Invest anterior uvea of the rabbit eye. Brain Res 172. Nielsen PJ, Nyborg NC: Contractile and
Ophthalmol Vis Sci 1988; 29:876–881. 1993; 610:194–198. relaxing effects of arachidonic acid
137. Nyborg NC, NP, Prieto D, Benedito S: 155. Park CSP, Gianotti K, Villegas C, et al: derivatives on isolated bovine retinal
Angiotensin II does not contract bovine Human retina expresses both constitutive resistance arteries. Exp Eye Res 1990;
retinal resistance arteries in vitro. Exp Eye and inducible isoforms of nitric oxide 50:305–311.
Res 1990; 50:469–474. synthase mRNA. Biochem Biophys Res 173. Starr MS: Effects of prostaglandings on
138. Krejcy K, WM, Kreuzer C, et al: Commun 1994; 205:85–91. blood flow in the rabbit eye. Exp Eye Res
Characterization of angiotensin-II effects on 156. Knowles RGM, S: Nitric oxide synthases 1990; 50:305–311.
cerebral and ocular circulation by in mammals. Biochem J 1994; 174. Hsu P, Albuquerque ML, Leffler CW:
noninvasive methods. Br J Clin Pharmacol 298:249–258. Mechanisms of hypercapnia-stimulated PG
1997; 43:501–508. 157. Meyer PC, Schlotzer-Schrehardt C, production in piglet cerebral microvascular
139. Alm A: Effects of norepinephrine, Flammer U, et al: Localization of nitric endothelial cells. Am J Physiol 1995; 268(2
angiotensin, dihydroergotamine, oxide synthase isoforms in porcine ocular Pt 2):H591–H603.
papaverine, isoproterenol, histamine, tissues. Curr Eye Res 1999; 18:375–380. 175. Chemtob SB, Rex K, Chatterjee J, et al:
nicotinic acid, and xanthinol nicotinate on 158. Chakravarthy US, McNally AW, Bailie J, Ibuprofen enhances retinal and choroidal
retinal oxygen tension in cats. Acta et al: Nitric oxide synthase activity and blood flow autoregulation in newborn
Ophthalmol 1972; 50:707–719. expression in retinal capillary endothelial piglets. Invest Ophthalmol Vis Sci 1991;
140. Haefliger IOB, Luscher JL, TF: cells and pericytes. Curr Eye Res 1995; 32:1799–807.
Endothelium-dependent vasoactive 14:285–294. 176. Stiris TS, Hehre C, Goldberg D, et al: Effect
modulation in the ophthalmic circulation. 159. Donati GP, Munoz CJ, Poitry JL, et al: Nitric of cyclooxygenase inhibition on retinal and
Prog Retin Eye Res 2001; 20:209–225. oxide controls arteriolar tone in the retina of choroidal blood flow during hypercarbia in
141. Haefliger IOM, Flammer P, Luscher J, TF: the miniature pig. Invest Ophthalmol Vis Sci newborn piglets. Pediatr Res 1992;
1672 The vascular endothelium as a regulator of 1995; 36:2228–2237. 31:127–130.
Retinal and Choroidal Circulations

177. Levin ER: Endothelins. N Engl J Med 1995; 195. Williamson DH, Krebs HA: The redox strate 215. Kondo M, Wang L, Bill A: The role of nitric
333:356–363. of free nicotinamide-adenine dinucleotide oxide in hyperaemic response to flicker in
178. Inoue AY, Kimura M, Kasuya S, et al: The in the cytoplasm and mitochondria in the the retina and optic nerve in cats. Acta
human endothelin family: three structurally rat liver. Biochem J 1967; 103:514–527. Ophthalmologica Scandinavica 1997;
and pharmacologically distinct isopeptides 196. Ido Y, Chong K, Williamson JR: NADH 75:232–235.
predicted by three separate genes. Proc augments blood flow in physiologically 216. Dorner GT, et al: Nitric oxide regulates
Natl Acad Sci USA 1989; 86:2863–2867. activated retina and visual cortex. Proc Natl retinal vascular tone in man. Am J Physiol
179. McDonald DMB, Archer JR, Chakravarthy Acad Sci USA 117:653–658. Heart Circ Physiol 2003; 10(1152 L1 –
DB: Characterization of endothelin A (ETA) 197. Garhofer G, Huemer KH, et al: Flicker light- Available at:
and endothelin B (ETB) receptors in induced vasodilatation in the human retina: \\Siro\public\Administration\Reference
cultured bovine retinal pericytes. Invest effect of lactate and changes in mean Manager\PDF Files\3953.pdf).
Ophthalmol Vis Sci 1995; 36:1088–1094. arterial pressure. Invest Ophthalmol Vis Sci 217. Deussen A, Sonntag M, Vogel R:
180. Takei KS, Nonoyama T, Miyauchi T, et al: 2003; 44:5309–5314. L-arginine-derived nitric oxide: a major
A new model of transient complete 198. Granger HJ, Shepperd AP: Intrinsic determinant of uveal blood flow. Exp Eye
obstruction of retinal vessels induced by microvascular control of tissu oxygen Res 1993; 57:129–134.
endothelin-1 injection into the posterior delivery. Microvasc Res 1973; 5:49. 218. Moncada S: Pharmacology and
vitreous body in rabbits. Graefes Arch Clin 199. Johnson PC: The myogenic response. In: endogenous roles of prostaglandins
Exp Ophthalmol 1993; 231:476–481. Bohr DF, Somlyo AP, Sparks HV, eds: endoperoxides, thromboxane A2 and
181. Granstam E, Wang, Bill A: Ocular effects of Handbook of physiology. The Prostacyclin 1979; 30:293–331.
endothelin-1 in the cat 1992; 11:325–332. cardiovascular system. Baltimore: Waverly 219. Radomski MW, Moncada S: The

CHAPTER 126
182. Bursell SEC, Oren AC, King B, GL: The in Press; 1980; 2:772–726. antiaggregation properties of vascular
vivo effect of endothelins on retinal 200. Yao KT, Flammer M, Luscher J: Endothelium- endothelium: interactions between
circulation in nondiabetic and diabetic rats. dependent regulation of vascular tone of the prostacyclin and nitric oxide. Br J
Invest Ophthalmol Vis Sci 1995; porcine ophthalmic artery. Invest Ophthalmol Pharmacol 1987; 93:639–646.
36:596–607. Vis Sci 1991; 32:1791–1798. 220. de Nucci GG, Warner RJ, Vane TD:
183. Dallinger SD, Wenzel GT, Graselli R, et al: 201. Harder DR: Pressure-induced myogenic Receptor-mediated release of endothelium-
Endothelin-1 contributes to hyperoxia- activation of cat cerebral arteries is derived relaxing factor and prostacyclin
induced vasoconstriction in the human dependent on intact endothelium. Circ Res from bovine aortic endothelial cells is
retina. Invest Ophthalmol Vis Sci 2000; 1987; 60:102–107. coupled. Proc Natl Acad Sci USA 1988;
41:864–869. 202. Harder DR: Pressure-dependent membrane 85:2334–2338.
184. Polak KL, Frank A, Jandrasits B, et al: depolarization in cat middle cerebral artery. 221. Doni MGW, Palmer BJ, Moncada RM:
Regulation of human retinal blood flow by Circ Res 1984; 55:197–202. Actions of nitric oxide on the release of
endothelin-1. Exp Eye Res 2003; 203. Harder DR, Kauser K, et al: Pressure prostacyclin from bovine endothelial cells in
76:633–640. releases a transferable endothelial culture. Eur J Pharmacol 1988; 151:19–25.
185. Wong J, VP, Fineman JR, et al: contractile factor in cat cerebral arteries. 222. Shimokawa HF, Lorenz NA, Vanhoutte RR:
Endothelin-1 produces pulmonary Circ Res 1989; 65:193–198. Prostacyclin releases endothelium-derived
vasodilation in the intact newborn lamb. 204. Lansman JB, Hallam TJ, Rink TJ: Single relaxing factor and potentiates its action in
Am J Physiol 1993; 265:H1318–H1325. stretch-activated ion channels in vascular coronary arteries of the pig. Br J
186. Eddahibi S, Monirul M, et al: Dilator effect endothelial cells as mechanotransducers? Pharmacol 1988; 95:1197–1203.
of endothelins in pulmonary circulation: Nature 1987; 325:811–813. 223. Petropoulos IK, Munoz JL, Pournaras CJ:
damages associated with chronic hypoxia. 205. Jeppesen P, Aalkjoer C, Bek T: Bradykinin Effect of carbogen breathing and
Am J Physiol 1993; 265:L571–L580. relaxation in small porcine retinal arterioles. acetazolamide on optic disc PO2. Invest
187. Filep JG, Rousseau A, et al: Vascular Invest Ophthalmol Vis Sci 2002; Ophthalmol Vis Sci 2005; 46:4139–4146.
response to endothelin-1 following inhibiton 43:1891–1895. 224. Nagaoka T, Mori F, et al: The effect of nitric
of nitric oxide synthesis in the conscious 206. Delaey C, Van de Voorde J: Pressure- oxide on retinal blood flow during hypoxia
rat 1993; 110:1213–1221. induced myogenic responses in isolated in cats. Invest Ophthalmol Vis Sci 2002;
188. D’Orleans-Juste P, Telemaque S, et al: bovine retinal arteries. Invest Ophthalmol 43:3037–3044.
Block of endothelin-1-induced release of Vis Sci 2000; 41:1871–1875. 225. Alm A: The effect of sympathetic
thromboxane A2 from the guinea pig lung 207. Bill A, Aspects of oxygen and glucose stimulation on blood flow through the uvea,
and nitric oxide from the rabbit kidney by a consumption in the retina: effects of high retina and optic nerve in monkeys (Macaca
selective ETB receptor antagonist, BQ-788. intraocular pressure and light. Graefes Arch irus). Exp Eye Res 1977; 25:19–24.
Br J Pharmacol 1994; 113:1257–1262. Clin Exp Ophthalmol 1990; 228:124–127. 226. Alm A, Bill A: The effect of stimulation of
189. Hirata Y, Eguchi S, et al: Endothelin 208. Wang L, Bill A: Glucose metabolism in cat the cervical sympathetic chain on retinal
receptor subtype B mediates synthesis of outer retina. Effects of light and hyperoxia. oxygen tension and on uveal, retinal and
NO by cultured bovine endothelial cells. Invest Ophthalmol Vis Sci 1997; 38:48–55. cerebral blood flow in cats. Acta Physiol
J Clin Invest 1993; 91:1367–1373. 209. Bill A, Control of retinal and choroidal blood Scand 1973; 88:84–94.
190. Reihart GA, Burke SE, et al: Hypertension flow. Eye 1990; 4:319–325. 227. Bill A, Linder M, Linder J: The protective
induced by blockade of ET(B) receptors in 210. Kondo M, Wang L, Bill A: The role of nitric role of ocular sympathetic vasomotor
conscious nonhuman primates: role of oxide in hyperaemic response to flicker in nerves in acute arterial hypertension. Bibl
ET(A) receptors. Am J Physiol Heart Circ the retina and optic nerve in cats. Acta Anat 1977; 16(Pt 2):30–35.
Physiol 2002; 283:H1555–H1561. Ophthalmol Scand 1997; 75:232–235. 228. Kiel JWS: Autoregulation of choroidal blood
191. Sokolvsky M, Galron R: A novel subtype of 211. Riva CE, Shonat RD, Petrig BL: Flicker flow in the rabbit. Invest Ophthalmol Vis Sci
endothelin receptors. J Biol Chem 1992; evoked increase in optic nerve head blood 1992; 33:2399–2410.
267:20551–20554. flow in anesthetized cats. Neurosci Lett 229. Riva CET, Hero P, Petrig M: Effect of acute
192. Poitry-Yamate CL, Poitry S, Tsacopoulos 1991; 128:291–296. decreases of perfusion pressure on
M: Lactate released by Muller glial cells is 212. Iadecola C: Regulation of the cerebral choroidal blood flow in humans. Invest.
metabolized by photoreceptors from microcirculation during neural activity: is Ophthalmol Vis Sci 1997; 38:1752–1760.
mammalian retina. J Neurosci 1995; nitric oxide the missing link? 1993; 230. Nilsson Bill A: Characteristicss of uvea
15(7 Pt 2):5179–5191. 16:206–214. vasodilation produced by facial nerve
193. Oldendorf WH: Carrier-mediated 213. Scheiner AJR, Kazahaya CE, Petrig K: stimulation in monkeys, cats, and rabbits.
blood–brain barrier transport of short-chain Effect of flicker on macular blood flow Exp Eye Res 1985; 40:841–852.
monocarboxylic organic acids. Am J assessed by the blue field simulation 231. Nilsson SFE: Studies on ocular blood flow
Physiol 1973; 224:1450–1453. technique. Invest Ophthalmol Vis Sci 1994; and acqueous humour dynamics. Acta
194. Garhofer G, Resch H, et al: Effect of 35:3436–3441. Universitatis Upsaliensis 1986; 43:1–38.
intravenous administration of sodium- 214. Buerk DG, Riva CE, Cranstoun SD: Nitric 232. Seligsohn EE, Bill A: Effects of NG-nitro-L-
lactate on retinal blood flow in healthy oxide has a vasodilatory role in cat optic arginine methyl ester on the cardiovascular
subjects. Invest Ophthalmol Vis Sci 2003; nerve head during flicker stimuli. Microvasc system of the anaesthetized rabbit and on
44:3972. Res 1996; 52:13–26. the cardiovascular response to thyrotropin- 1673
RETINA AND VITREOUS

releasing hormone. Br J Pharmacol 1993; 250. Anderson B Jr: Ocular effects of changes in 270. Beckman JS: The double-edged role of
109:1219–1225. oxygen and carbon dioxide tension. Trans nitric oxide in brain function and
233. Mann RM, et al: Nitric oxide and choroidal Am Ophthalmol Soc 1968; 66:423–474. superoxide-mediated injury. J Dev Physiol
blood flow regulation. Invest Ophthalmol 251. Watkins JC, EH: Excitatory amino acid 1991; 15:53–59.
Vis Sci 1995; 36:925–930. transmitters. Annu Rev Pharmacol Toxicol 271. Iadecola C, Zhang F, Xu X: Inhibition of
234. Schmetterer L, et al: The effect of systemic 1981; 21:165–204. inducible nitric oxide synthase ameliorates
nitric oxide-synthase inhibition on ocular 252. Rothman SM: Glutamate and the patho- cerebral ischemic damage. Am J Physiol
fundus pulsations in man. Exp Eye Res physiology of hypoxic-ischemic brain 1995; 268:R286–R292.
1997; 64:305–312. damage 1986; 19:105–111. 272. Zhang F, IC: Nitroprusside improves blood
235. Luksch A, et al: Effects of systemic NO 253. Rothman SM: Excitotoxicity and the NMDA flow and reduces brain damage after focal
synthase inhibition on choroidal and optic receptor. Trends Neurosci 1987; ischemia. Neuroreport 1993; 4:559–562.
nerve head blood flow in healthy subjects. 10:299–302. 273. Carreau A, Duval D, Poignet H, et al:
Invest Ophthalmol Vis Sci 2000; 254. Rothman SM: Synaptic release of Neuroprotective efficacy of N omega-nitro-
41:3080–3084. excitatory amino acids neurotransmitter L-arginine after focal cerebral ischemia in
236. Blitzer ML, LS, Creager MA: Endothelium- mediates anoxic neuronal death. the mouse and inhibition of cortical nitric
derived nitric oxide mediates hypoxic J Neurosci 1984; 4:884–891. oxide synthase. Eur J Pharmacol 1994;
vasodilation of resistance vessels in 255. Andine P, Ellren K et al: The excitatory 256:241–249.
human. Am J Physiol 1996; amino acid antagonist kynurenic acid 274. Cooke JP, Girerd XJ, et al: Arginine restores
271:H1182–H1185. administered after hypoxic-ischemia in cholinegic relaxation of
237. Hardy PN, Abran AM, St-Louis D, et al: neonatal rats offers neuroprotection. hypercholesterolemic rabbit thoracic aorta.
Nitric oxide in retinal and choroidal blood Neurosci Lett 1988; 90:208–212. Circulation 1991; 83:1057–1062.
SECTION 3

flow autoregulation in newborn pigs: 256. Barbour B, Attwell D: Electrogenic uptake 275. Rossitch E, Black McLP, Cooke JP: L-
interactions with prostaglandins. Pediatr of glutamate and aspartate into glial cells arginine normalizes endothelial function in
Res 1996; 39:487–493. isolated from the salamander (Ambystoma) cerebral vessels from hypecholesterolemic
238. Van-Bel F, Roman C, Rudolph AM: retina. J Physiol 1991; 436:169–193. rabbits. J Clin Invest 1991; 87:1295–1299.
Perinatal regulation of the cerebral 257. Brew H: Electrogenic glutamate uptake is a 276. Carville C, Eddahibi S, et al: Loss of
circulation: role of nitric oxide and major current carrier in the membrane of endothelium-dependent relaxation in
prostaglandins. Pediatr Res 1997; axolotl retinal glial cells. Nature 1987; proximal pulmonary arteries from rats
42:299–304. 327:707–709. exposed to chronic hypoxia: effects of in
239. Granstam E, Wang L, Bill A: Vascular 258. Tsacopoulos M: Metabolic coupling vivo and in vitro supplementation with-
effects of endothelin-1 in the cat; between glia and neurons. J Neurosci Arginine. J Cardiov Pharmac 1993;
modification by indomethacin and L-NAME. 1996; 16:877–885. 22:889–896.
Acta Physiol Scand 1993; 148:165–176. 259. Pellerin L: Glutamate uptake into astrocytes 277. Wolzt M, Schmetter L, Ferver W, et al:
240. MacCumber MW, Jampel HD, Snyder SH: stimulates aerobic glycolysis: a mechanism Effect of nitric oxide synthase inhibition on
Ocular effects of the endothelins. Abundant coupling neuronal activity to glucose renal hemodynamics in humans: reversal
peptides in the eye. Arch Ophthalmol 1991; utilization. Proc Natl Acad Sci USA 1994; by L-arginine. In: The American
109:705–709. 91:10625–10629. Physiological Society 1997:F178–F182.
241. Alder VAB-N, Cringle J, PO2 profiles and 260. Magistretti PJ: Astrocytes couple synaptic 278. Grunwald JEB, Grunwald AJ, Riva SE:
oxygen consumption in cat retina with an activity to glucose utilization in the brain. Retinal hemodynamics in proliferative
occluded retinal circulation. Invest News Physiol Sci 1999; 14:177–182. diabetic retinopathy. a laser Doppler
Ophthalmol Vis Sci 1990; 31:1029–1034. 261. Napper GA, Kalloniatis M: Reduced velocimetry study. Invest Ophthalmol Vis
242. Bardy M, Tsacopoulos M: Metabolic glutamate uptake by retinal glial cells under Sci 1993; 34:66–71.
changes in the retina after experimental ischemic/hypoxic conditions. Vis Neurosci 279. Rosen DAM, Kohner J, Hamilton EM, et al:
microembolism in the miniature pig 1999; 16:149–158. Experimental retinal branch vein occlusion
(author’s transl). Klin Monatsbl Augenheilkd 262. Adachi K, Masai H, et al: Mechanism of the in rhesus monkeys. II. Retinal blood flow
1978; 172:451–460. pathogenesis of glutamate neurotoxicity in studies. Br J Ophthalmol 1979; 63:388–392.
243. Pournaras CJR, Munoz A, Abdesselem JL, retinal ischemia. Graefes Arch Clin Exp 280. Pournaras CJT, Strommer M, Gilodi K,
Experimental venous branch occlusion: Ophthalmol 1998; 236:766–774. et al: Experimental retinal branch vein
change in the preretinal oxygen pressure 263. Kageyama T, Tamail M: Glutamate elevation occlusion in miniature pigs induces local
pO2 by dexamethasone. Klin Monatsbl in rabbit vitreous during transient ischemia- tissue hypoxia and vasoproliferative
Augenheilkd 1990; 196:475–480. reperfusion. Jpn J Ophthalmol 2000; microangiopathy. Ophthalmology 1990;
244. Pournaras CJT, Bovet M, Roth J: Diffusion 44:110–114. 97:1321–1328.
of O2 in the normal and the ischemic retina 264. Iijima T, Iwao Y, Sankawa H: Differences in 281. Pournaras CJ: Retinal oxygen distribution.
of miniature pigs. Ophtalmologie 1990; glutamate release between retina and Its role in the physiopathology of
4:17–19. cerebral cortex following ischemia. vasoproliferative microangiopathies. Retina
245. Kohner EMD, Shakib CT, Henkind M, et al: Neurochem Int 2000; 36:221–224. 1995; 15:332–347.
Experimental retinal branch vein occlusion. 265. Pournaras CJ, Poitry S, Donati G: Preretinal 282. Pournaras CJM, Gragoudas JW, Husain
Am J Ophthalmol 1970; 69:778–825. lactate measurements after experimental ES, et al: Systemic hyperoxia decreases
246. Hockley DJ, Tripathi RC, Ashton N: branch retinal vein occlusion. Invest vascular endothelial growth factor gene
Experimental retinal branch vein occlusion Ophthalmol Vis Sci 2001; 42:239. expression in ischemic primate retina. Arch
in rhesus monkeys. III. Histopathological 266. Morikawa E, Huang Z, Moskowitz MA: Ophthalmol 1997; 115:1553–1558.
and electron microscopical studies. Br J L–arginine decreases infarct size caused by 283. Wallow IH, Geldner PS: Endothelial
Ophthalmol 1979; 63:393–411. middle cerebral arterial occlusion in SHR. fenestrae in proliferative diabetic
247. Donati G, Pournaras, Pizzolato CJ, et al: Am J Physiol 1992; 263(5 Pt retinopathy. Invest Ophthalmol Vis Sci
Decreased nitric oxide production accounts 2):H1632–H1635. 1980; 19:1176–1183.
for secondary arteriolar constriction after 267. Garthwaite J, Palmer RMJ, Moncada S: 284. Hamilton CWC, Klintworth D, Machemer
retinal branch vein occlusion. Invest NMDA receptor activation induces nitric GK: A transmission and scanning electron
Ophthalmol Vis Sci 1997; 38:1450–1457. odixe synthesis from arginine in rat brain microscopic study of surgically excised
248. Tsacopoulos M: Role of metabolic factors slices. Eur J Pharmacol 1989; preretinal membrane proliferations in
in the regulation of retinal blood minute 172:413–416. diabetes mellitus. Am J Ophthalmol 1982;
volume. Adv Ophthalmol 1979; 268. Garthwaite J: Nitric oxide and cell-cell 94:473–488.
39:233–273. signaling in the nervous system 1991; 285. Miller HM, Zonis B: Diabetic
249. Bietti G: Effects of experimentally 14:60–88. neovascularization: permeability and
decreased or increased oxygen supply in 269. Beckman JS, et al: Apparent hydroxyl ultrastructure. Invest Ophthalmol Vis Sci
some ophthalmic diseases; practical value radical production by peroxynitrite: 1984; 25:1338–1342.
for diagnosis, prognosis, and treatment. implications for endothelial injury from nitric 286. Ozaki HY, Della AY, Ozaki N, et al: Hypoxia
AMA Arch Ophthalmol 1953; 49: oxide and superoxide. Proc Natl Acad Sci inducible factor-1alpha is increased in
1674 491–513. USA 1990; 87:1620–1624. ischemic retina: temporal and spatial
Retinal and Choroidal Circulations

correlation with VEGF expression. Invest 296. Pierce EA, Avery RL, Smith LEH: Vascular with arteriovenous fistula in the cavernous
Ophthalmol Vis Sci 1999; 40:182–189. endothelial growth factor/vascular sinus area. Arch Ophthalmol 1978;
287. Boulton MP, Khaliq B, Moriarty A, et al: permeability factor expression in a mouse 96:1370–1373.
Modulators and milieu in preretinal model of retinal neovascularisation. Proc 306. Brubaker RF, Pederson JE: Ciliochoroidal
neovascularisation. Eye 1992; 6(Pt Natl Acad Sci 1995; 92:905–909. detachment. Surv Ophthalmol 1983;
6):560–565. 297. Shima DTG, Miller A, Tolentino JW, et al: 27:281–289.
288. Bounelis PM, King W, et al: Hypoxia Cloning and mRNA expression of vascular 307. Shimizu K: Segmental nature of the
stimulates platelet derived growth factor endothelial growth factor in ischemic angioarchitecture of the choroid.
gene expression by pulmonary artery retinas of Macaca fascicularis. Invest Proceedings of the XXIII international
endothelial cells. Clin Res 1988; 36:503A. Ophthalmol Vis Sci 1996; 37:1334–1340. Congress in Ophthalmology. Kyoto; 1978.
289. Shweiki DI, Soffer A, Keshet D: Vascular 298. Adamis AP, Bernal M-T: Increased vascular 308. Hayreh SS: In vivo choroidal circulation and
endothelial growth factor induced by endothelial growth factor levels in the its watershed zones. Eye 1990; 4(Pt
hypoxia may mediate hypoxia-initiated vitreous of eyes with proliferative diabetic 2):273–289.
angiogenesis. Nature 1992; 359:843–845. retinopathy. Am J Ophthalmol 1994; 309. Gass JMD: Stereoscopic atlas of macular
290. Wojta JJ, Jones RL, Binder RL, Hoover BR: 118:445–450. disease. St Louis: Mosby; 1997:187–197.
Reduction in pO2 decreases the fibrinolytic 299. Aiello LPA, Arrigg RL, Keyt PG, et al: 310. Collier RH: Experimental embolic ischemia
potential of cultured bovine endothelial Vascular endothelial growth factor in ocular of the choroid. Arch Ophthalmol 1967;
cells derived from pulmonary arteries and fluid of patients with diabetic retinopathy 77:683–692.
lung microvasculature. Blood 1988; and other retinal disorders. N Engl J Med 311. Buettner HM, Charles R, Anderson S:
71:1703–1706. 1994; 331:1480–1487. Experimental deprivation of choroidal blood

CHAPTER 126
291. Shatos MAD, Stump JM, Thompson DC, 300. Malecaze FC, Simorre-Pinatel S, Mathis V, flow. Retinal morphology, early receptor
et al: Oxygen radicals generated during et al: Detection of vascular endothelial potential, and electroretinography. Am J
anoxia followed by reoxygenation reduce growth factor messenger RNA and vascular Ophthalmol 1973; 75:943–952.
the synthesis of tissue-type plasminogen endothelial growth factor-like activity in 312. Almaric P: Circulation choroidienne. In:
activator and plasminogen activator proliferative diabetic retinopathy. Arch Compte rendu du symposium international
inhibitor-1 in human endothelial cell culture. Ophthalmol 1994; 112:1476–1482. d’angiographie fluorescéinique, Albi 1969.
J Biol Chem 1990; 265:20443–20448. 301. Pe’er j AD, Itin A, Hemo J, et al: Hypoxia- Karger: Basel; 1971:193–203.
292. Montesano RV, Baird JD, Guillemin A, et al: induced expression of vascular endothelial 313. Bischoff PM, Flower RW: High blood
Basic fibroblast growth factor induces growth factor by retinal cells is a common pressure in choroidal arteries as a possible
angiogenesis in vitro. Proc Natl Acad Sci factor in neovascularizing ocular diseases. pathogenetic mechanism in senile macular
USA 1986; 83:7297–7301. Lab Invest 1995; 72:638–645. degeneration. Am J Ophthalmol 1983;
293. Shreeniwas RO, Cozzolino S, Torcia F, et al: 302. Shima DTA, Ferrara AP, Yeo N, et al: 96:398–399.
Macrovascular and microvascular Hypoxic induction of endothelial cell 314. Condon PI, Serjeant GR, Ikeda H: Unusual
endothelium during long-term hypoxia: growth factors in retinal cells: identification chorioretinal degeneration in sickle cell
alterations in cell growth, monolayer and characterization of vascular endothelial disease. Possible sequelae of posterior
permeability, and cell surface coagulant growth factor (VEGF) as the mitogen. Mol ciliary vessel occlusion. Br J Ophthalmol
properties. J Cell Physiol 1991; 146:8–17. Med 1995; 1:182–193. 1973; 57:81–88.
294. Plate KHB, Weich G, Risau HA: Vascular 303. Bill A: Capillary permeability to and 315. Jampol LML, Albert M, Craft DM: Ocular
endothelial growth factor is a potential extravascular dynamics of myoglobin, clinical findings and basement membrane
tumour angiogenesis factor in human albumin and gammaglobulin in the uvea. changes in Goodpasture’s syndrome. Am J
gliomas in vivo. Nature 1992; 359:845–848. Acta Physiol Scand 1968; 73:204–219. Ophthalmol 1975; 79:452–463.
295. Miller JWA, Shima AP, D’Amore DT, et al: 304. Mäepa O: Pressure in the anterior ciliary 316. Spolaore RG, Coscas A, de Margerie G:
Vascular endothelial growth factor/vascular arteries, choroidal veins and Acute sectorial choroidal ischemia. Am J
permeability factor is temporally and choriocapillaris. Exp Eye Res 1992; Ophthalmol 1984; 98:707–716.
spatially correlated with ocular 54:731–735. 317. Gaudric A, Coscas G, Bird AC: Choroidal
angiogenesis in a primate model. Am J 305. Klein RM, Smith SM, Myers JL, et al: ischemia. Am J Ophthalmol 1982;
Pathol 1994; 145:574–584. Abnormal chloroidal circulation: association 94:489–498.

1675
CHAPTER
Examination of the Retina: Ophthalmoscopy and
127 Fundus Biomicroscopy
Thomas R. Friberg

INTRODUCTION
DIRECT OPHTHALMOSCOPE
The essentials of the clinical examination techniques used to To examine the ocular fundus, specialized instruments are neces-
evaluate the ocular fundus have changed little over the past sary. The simplest is the direct ophthalmoscope, which is in
decade. While additional ancillary testing and methods have been essence a miniature flashlight held very close to the patient’s eye
developed, a thorough funduscopic examination remains a main- and shined through the pupil (Fig. 127.1). The fundus is viewed
stay of clinical practice. Excellent and efficient retinal examin- monocularly through a small peephole located just above the
ation skills are critical to virtually all ophthalmologists, and this illumination source of the instrument, producing an upright
chapter explains the principles of this evaluation. virtual image that magnifies the area of interest ~15 times.3
A thorough examination of the ocular fundus requires excel- This ophthalmoscope also has a dial containing neutralizing
lent powers of observation and clinical experience with ophthal- lenses, which is rotated to achieve the clearest retinal image.
moscopy and biomicroscopy. Some individuals have almost a Although magnification and resolution are quite good with the
natural ability to use these specialized instruments to discover direct ophthalmoscope, difficulties inherent with its use include
and document subtle retinal abnormalities. Most have consid- lack of stereopsis, inadequate illumination in the presence of
erably more difficulty, however. For instance, a novice might media opacities, the necessity of placing the examiner’s face in
struggle and then finally see scattered retinal hemorrhages in the close proximity to the patient’s face, a retinal image covering only
fundus of a patient’s eye but then fail to note the location of the ~8° of the fundus,3 and severe degradation of the image when
hemorrhages with respect to retinal landmarks. With practice, pro- significant lens opacities are present.4,5 It may be impossible to
per use of the instrumentation can be learned by almost anyone. adequately examine eyes with a high degree of astigmatism or
Unfortunately, some clinicians never become good observers and spherical ametropia using this instrument. Furthermore, the direct
remain unaware of the important distinction between seeing an ophthalmoscope does not allow an undistorted view of the periph-
abnormality and observing it.1 Observation is a high-level activity, eral retina, limiting its use to visualizing the posterior retina only.
requiring mental processing and categorization of what is seen. Largely because of its portability and simplicity, the direct
One method of improving observational skills is to use an ophthalmoscope is a useful screening device for the general
examination routine whereby different regions of the fundus are practitioner who may wish to rule out papilledema, macular
evaluated in a specific sequence. For instance, an ophthalmo- degeneration, or hypertensive or diabetic retinopathy. Many
logist might begin by examining the optic nerve; move on to the ophthalmologists have become facile with binocular slit-lamp
temporal vascular arcades, macula, and nasal retina; and finish biomicroscopes and specialized contact and noncontact fundus
by evaluating the equatorial and peripheral retina. By using an examination lenses, however, and observation of the ocular
organized system of fundus evaluation, diagnostic oversights fundus with the direct ophthalmoscope is often redundant and
will be minimized. unnecessary in the office setting.

PUPILLARY DILATATION BINOCULAR INDIRECT OPHTHALMOSCOPE


Examination of the fundus and especially the retinal periphery The binocular indirect ophthalmoscope, in conjunction with a
is greatly facilitated by a well-dilated pupil. Suggested mydriatic high-quality aspherical hand-held lens, has become the indis-
agents include 1% tropicamide (Mydriacyl) and 2.5% phenyl- pensable standard for the proper examination of the fundus,
ephrine hydrochloride drops (Mydfrin), one drop of each in both especially when evaluating areas located outside the posterior
eyes. Instillation of a topical anesthetic, such as 0.5% proparacaine pole (Fig. 127.2). Introduced by Schepens,6 binocular indirect
hydrochloride, before instilling the dilating drops promotes ophthalmoscopy offers a typical field of view of 25º or more and
more rapid mydriasis, as the anesthetic prevents reflex tearing excellent resolution of fundus details.5 Stereopsis is enhanced,
and subsequent dilution of the mydriatic agent. Most patients’ allowing the examiner to detect nuances of elevation and exca-
pupils dilate adequately after 20–30 min using this regimen. vation of the retinal contour. The device is portable, permits
However, darkly pigmented irises dilate more slowly and remain evaluation of the retina with the patient in either the sitting or
dilated longer than do lightly colored irises, probably because the supine position, and quickly gives a view of relatively large
the mydriatic agents are bound to the iris melanin and are retinal areas. Furthermore, binocular indirect ophthalmoscopy
released gradually.2 After the examination, mydriasis can be has been incorporated into laser delivery systems, providing an
reversed more quickly by medications designed for this purpose important alternative to slit-lamp photocoagulation systems.7
such as 0.5% dapiprazole hydrochloride (Rev-Eyes, Storz Although an examination of the posterior fundus using the
Ophthalmics). direct ophthalmoscope can be performed after a minimum of 1677
SECTION 10 RETINA AND VITREOUS

FIGURE 127.1. The direct ophthalmoscope is used like a flashlight to


illuminate the patient’s eye while the examiner looks through a small a
peephole. The left eye of the examiner is used to study the fundus of
the patient’s left eye. Conversely, the right eye is used to examine the
patient’s right eye.

instruction, effective use of the indirect ophthalmoscope requires


several hours of practice. The image formed by the indirect
ophthalmoscope is physically located above the plane of the
lens and, as with all real images, is inverted (Fig. 127.3). This
inversion creates considerable problems for the novice, especially
when he or she tries to draw the observed abnormalities. In
addition, the alignment of the indirect ophthalmoscope’s illu-
mination beam, the hand-held lens, the patient’s pupil, and the
ophthalmoscope oculars is crucial to obtaining a sharp image.
Hence, before examining patients, it is beneficial to examine a
model eye (Mira, Waltham, MA) manufactured for the purpose
of practicing indirect ophthalmoscopy (Fig. 127.4). Another aid
to learning this important skill is a specially designed interac-
tive computer program contained on a compact disk (CD-ROM)
(‘Indirect Ophthalmoscopy and Fundus Drawing’, © 1996 by
The New York Eye and Ear Infirmary).

EXAMINATION TECHNIQUE WITH THE


INDIRECT OPHTHALMOSCOPE
To use the indirect ophthalmoscope, the examiner first adjusts
the headbands so that the scope fits comfortably. A lightweight
instrument is preferred. The illumination beam is turned on, and
the interpupillary distance of the eyepieces is adjusted so that
both eyes can see the examiner’s outstretched hand simulta-
neously. Next, the knob on the headset that controls the location
of the illumination beam is rotated until the ‘footprint’ of the b
light illuminates the superior portion of the outstretched hand.
The head and body of the examiner should be positioned so that FIGURE 127.2. (a) An example of a binocular indirect
her or his viewing axis is in a line with the center of the patient’s ophthalmoscope. The coronal and sagittal headbands and the
dilated pupil. When this is done, a bright red reflex should appear interpupillary distance are adjustable. A transformer provides power
through the oculars of the indirect ophthalmoscope. for illumination. (b) With the indirect ophthalmoscope adjusted and in
Once a red reflex is visualized, the indirect hand-held lens is place on the head, the examiner holds a lens over the patient’s eye to
interposed along the imaginary line drawn between the examiner’s form a retinal image.
pupil and the patient’s pupil. The indirect lens should be held
between the thumb and the forefinger of either hand and
1678
Examination of the Retina: Ophthalmoscopy and Fundus Biomicroscopy

CHAPTER 127
FIGURE 127.4. Pliable model eyes with realistic fundus details are
valuable when learning binocular indirect ophthalmoscopy. They can
also be indented to simulate scleral depression.

FIGURE 127.5. The most convex side of the hand-held lens is placed
so that it faces the examiner. By tilting the lens very slightly, the bright
light reflexes produced by the lens surfaces (left) do not enter the
FIGURE 127.3. With indirect ophthalmoscopy, a real, inverted image examiner’s eye.
of the patient’s fundus is formed in a plane located just above the
hand-held lens.

must remain in alignment. A useful aid is to envision a solid


positioned a few centimeters from the patient’s eye. The lens sur- rod representing the viewing axis of the indirect scope to which
face is oriented almost perpendicular to the illumination beam. the center of the hand-held lens is fixed. This imaginary rod
To minimize problems with light reflections, the lens should be pivots at the center of the patient’s pupil when the entire extent
tilted very slightly, with the most convex side of the indirect of the retina is examined (Fig. 127.6). Indirect ophthalmoscopy
lens facing the examiner (Fig. 127.5). is facilitated by having the patient in the supine position with
To steady the lens, the fourth and fifth digits of the hand hold- the examiner standing, as this allows easy access to all retinal
ing the lens should rest on the patient’s face during the exam- regions.
ination. The distance between the patient’s eye and the lens
should otherwise be as great as possible. To optimize the view, Key Feature
the lens is slowly moved toward or away from the patient’s eye. • Proper alignment of the ophthalmoscope and the hand-held
When the lens is properly positioned, the image of the patient’s lens facilitates the examination. When the fundus image is
fundus fills the lens and is in sharp focus. To examine different difficult to visualize, the novice should check this alignment as
areas of the fundus, the viewing axis of the indirect ophthalmo- an initial step.
scope, the center of the hand-held lens, and the patient’s pupil
1679
SECTION 10 RETINA AND VITREOUS

FIGURE 127.7. The field of view of the indirect ophthalmoscope


FIGURE 127.6. To view different regions of the fundus, the examiner using a 20-D lens (outer circle) is much larger than that provided by
moves his or her head and the hand-held lens as if there were an the direct ophthalmoscope (inner circle). However, the size of a given
imaginary rod connecting the center of the patient’s pupil, the center retinal lesion obtained using the direct ophthalmoscope appears
of the lens, and a point midway between the eyepieces of the indirect commensurately bigger.
ophthalmoscope. The examiner then pivots around the central pupil to
view the complete fundus.
SMALL-PUPIL INDIRECT
Many patients, especially those with light complexions, are OPHTHALMOSCOPES
photophobic when examined with bright light. Hence, the
illumination level of the ophthalmoscope must be low enough Many indirect ophthalmoscopes incorporate a small pupil feature
to allow examination of the fundus without causing patient dis- which facilitates the examination of eyes when the pupils dilate
comfort and resultant squeezing of the eyelids. Conversely, the poorly or eyes that harbor focal media opacities. Typically, a
light must be sufficiently bright to allow observation of retinal small lever located under the eye pieces is moved in position to
details. For patient comfort, it is best to start with a dimmer engage this feature. The lever physically places the illumination
light and increase its intensity as necessary. and visualization pathways closer together by moving a
The patient’s eyelids should be held open with the examiner’s triangular prism. Such a feature is extremely useful and makes
fingers or a lid speculum during indirect ophthalmoscopy. Either the instrument more versatile.
the hand holding the lens or the free hand can be used for this
purpose, depending on whether scleral depression is being per- SCLERAL DEPRESSION
formed. Although use of a lid speculum potentially frees up one
of the examiner’s hands, an irrigating balanced salt solution To view the total extent of the peripheral fundus of the emmet-
(BSS) must be periodically instilled to prevent corneal drying ropic eye, the wall of the eye must be depressed inward toward
when a speculum is used. the visual axis. This examination technique, called scleral
depression, should not be attempted until one is first accom-
CHOICE OF HAND-HELD LENS plished at obtaining a sharp image of the posterior pole. If one
cannot reliably image the posterior segment, attempting to view
Numerous lenses are available for binocular indirect ophthal- the retinal periphery with scleral depression is usually futile as
moscopy. When selecting a lens, recall that angular magnifica- well as uncomfortable for the patient.
tion of fundus details is inversely proportional to the power of Several types of scleral depressors have been designed, and
the hand-held lens. For example, a 20-D lens will magnify the each has its advocates. The working end of most depressors con-
fundus details of an emmetropic eye ~2.3 times, whereas a 30-D sists of a metal shaft with an enlargement or crosspiece at the
lens magnifies it ~1.5 times.8 A high-quality 20-D aspherical tip. Some depressors have a more elaborate construction with
lens is widely used because it offers a good compromise between moving parts. Alternatively, one can simply use a cotton-tipped
field of view and magnification. However, when viewing eyes applicator pressed against the lids to accomplish the same goal,
with small pupils or eyes with hazy media, a 30- or 28-D lens although the tip is too bulky for many patients.
is often a better choice for the beginner. The field of view of the Pressure is exerted on the globe by pressing gently on the
lens is generally directly proportional to the lens power, with a patient’s partially opened eyelid until the peripheral retina is
20-D aspherical lens providing about a 35º field.3 Field of view brought into view (Fig. 127.8). If excessive pressure is used, the
is also a function of the diameter of the lens, with a larger field patient will become uncomfortable, squeeze the eyelids shut,
offered by a larger diameter lens. A comparison of field of view and prevent the examination from proceeding. With practice,
obtained when using a direct ophthalmoscope versus an indirect patient discomfort should be minimal. Typically, scleral depres-
1680 ophthalmoscope is shown in Figure 127.7. sion of the entire 360º of the retinal periphery is easier to
Examination of the Retina: Ophthalmoscopy and Fundus Biomicroscopy

accomplish if the patient is in the supine position. The shaft of movement of the depressor in the vicinity of a suspicious area
the depressor, the area of interest in the retinal periphery, and during observation may open up a previously unseen tear or
the central cornea should all be kept in the same plane during demonstrate areas of vitreoretinal traction. Hence, to detect and
scleral depression.9 diagnose subtle retinal abnormalities, dynamic scleral depression
Scleral depression is not a static process whereby the peri- is required.
pheral retina is merely brought into view and observed. Gentle Topical anesthesia, such as a drop of 0.5% proparacaine hydro-
chloride, may facilitate the examination with scleral depression.
To limit patient discomfort, the complete examination should
be performed without placing the depressor directly onto the
conjunctival surface (Fig. 127.9). Occasionally, evaluation of
some peripheral areas, particularly those at the 3 and 9 o’clock
meridians, necessitates placement of the depressor directly on
the globe.

DOCUMENTING THE RETINAL FINDINGS


Detailed sketches of the fundus are made on durable drawing
paper to document the retinal pathologic condition (Fig. 127.10).

CHAPTER 127
The location of a given lesion is drawn in reference to the major
retinal veins, the meridional location of the lesion within the
eye, and its relative peripheral location. The arteries are not
typically drawn. By convention, the 12 o’clock meridian is placed
at the top of the retinal drawing because it represents the upper-
most part of the clock face. Keep in mind that mapping the
retinal surface, which is spherical, on a flat piece of paper neces-
sarily produces inaccuracies of scale in the finished drawing. For
instance, the peripheral retina is disproportionately represented,
just as the size of equatorial regions of the world is exaggerated
in flat, polar maps generated by gnomonic projection.10 Hence,
a lesion of a given area will appear larger on the retinal drawing

FIGURE 127.8. To perform scleral depression, light pressure is


exerted on the globe through the patient’s eyelids with the tip of the
scleral depressor. The shaft of the depressor, the area of interest in
the fundus, and the central cornea should all be in the same plane.

FIGURE 127.10. Drawing of a retinal detachment on conventional


retinal drawing paper. The center of the paper represents the fovea,
the inner circle (E) represents the equator, and the outer circle (O)
represents the ora serrata. The region defined by the two outermost
circles represents the pars plana. On this paper, labeled O.S. for the
left eye, the optic nerve is predrawn and located to the left of the
fovea. Drawing paper labeled O.D. has the optic nerve drawn to the
right of the fovea. Findings located on the drawing correspond to their
clock-hour location within the patient’s eye, with 12 o’clock
FIGURE 127.9. The scleral depressor is carefully repositioned along representing the superiormost portion of the globe. In this figure, a
the upper and lower eyelids to completely examine all 360º of the retinal detachment has been drawn extending from the 9:30 to the
retinal periphery. 12:30 positions. The retinal tear is drawn at the 11:45 position. 1681
RETINA AND VITREOUS

FIGURE 127.11. (Left) Some fundus details located along the meridian at the 1:30 position on the clock are outlined by three circular areas,
representing three areas to be viewed by indirect ophthalmoscopy. (Center) The individual image fields of the fundus, as seen through the
examiner’s lens. Each field has been inverted by the optical system. (Right) The salient details of each image field are drawn onto the fundus
drawing paper. The paper is oriented with the 12 o’clock meridian directed toward the patient’s feet. This allows the examiner to draw what she
or he sees within each field as if it were being ‘pasted’ directly onto the drawing paper. If the drawing paper were not inverted, the examiner
would have to mentally invert the images before drawing them.
SECTION 10

paper if it is located in the peripheral fundus than it would if it


were located in the macula.
Because the retinal image is inverted during indirect ophthal-
moscopy, beginners often make the mistake of assuming that
the pathologic condition seen at the 6 o’clock position in the
hand-held lens must be located at the 12 o’clock position in the
eye. In fact, since only part of the fundus is imaged at a time,
only the small fundus region that is being viewed is inverted
(Fig. 127.11).5 To avoid confusion, one can move the hand-held
lens out of the way occasionally to observe which quadrant of
the patient’s eye the indirect ophthalmoscope is being directed
toward.

Key Feature
• The retinal image is inverted using the indirect
ophthalmoscope examination technique. When learning
ophthalmoscopy, it is often helpful to look at the eye without
the lens in position to confirm what region of the fundus is
being viewed.

When drawing the fundus, it is helpful to place the drawing


paper on a clipboard and have the patient hold or support the
clipboard on his or her chest. By orienting the paper so that its
12 o’clock meridian is directed inferiorly toward the patient’s
feet, the image fields can be visually translated directly onto the
paper without having to mentally invert them. Holding a pencil
in one hand while holding the lens and the patient’s eyelids
open with the other hand greatly speeds up the process of
generating an accurate retinal drawing (Fig. 127.12). FIGURE 127.12. The patient’s fundus findings can be documented
As retinal features are color-coded by convention (Table 127.1), rapidly if the examiner holds the lens in one hand while drawing on
color pencils are characteristically used for the retinal drawing. the paper with a pencil held in the other hand. Again, the paper is
purposely inverted.

LIMITATIONS OF THE INDIRECT


OPHTHALMOSCOPE
TABLE 127.1 Color Coding for Retinal Drawing
Although the indirect ophthalmoscope is an invaluable instru-
ment, it has limitations. A major drawback is that its magnifi- Color Anatomic Feature
cation is insufficient to allow detection of very small retinal
Red Retinal arteries, retinal hemorrhage,
abnormalities, particularly subtle macular lesions. For example, attached retina, retinal neovascularization
retinal microaneurysms, tiny areas of subretinal neovasculari-
zation, foveal cysts, and small round retinal breaks may be Blue Retinal veins, detached retina, retinal
edema
difficult to resolve with this instrument. For this reason, other
ancillary devices are necessary to thoroughly evaluate the fundus. Green Media opacities, vitreous hemorrhage
Yellow Retinal and choroidal exudates
BIOMICROSCOPY OF THE RETINA Brown Pigmented lesions, choroidal detachments
The slit lamp is an essential instrument for fundus examination, Red lined with blue Retinal breaks
1682 particularly when a high degree of magnification is desired. It
Examination of the Retina: Ophthalmoscopy and Fundus Biomicroscopy

consists of a movable binocular biomicroscope mounted on a If the slit beam of the biomicroscope is placed across an ele-
table and an intense illumination beam or slit beam of adjustable vated retinal lesion, the behavior of the scattered light also pro-
width that can be rotated 360º in the vertical plane. Focusing is vides important diagnostic clues. Often the borders of a small
accomplished by moving a joystick located on the microscope serous retinal or pigment epithelial detachment can be made to
platform. The slit lamp is used in conjunction with a diagnostic glow by positioning the slit beam across the elevation, making
contact lens or hand-held lens to provide a high-quality magni- the lesion easier to demarcate. This technique is very useful in
fied stereoscopic image of the fundus. Slit-lamp biomicroscopy evaluating the posterior pole for the presence of a macular hole
of the fundus is particularly useful in determining whether the or for detecting cystic spaces within the fovea.
location of a hemorrhage is preretinal, intraretinal, or sub- Slit-lamp examination methods can be separated into two
retinal; in detecting cystoid macular edema; and in diagnosing major categories: (1) contact methods requiring the placement of
clinically significant macular thickening. a specialized contact lens onto the corneal surface to neutralize
The slit lamp also provides optical sections of the vitreous body its power and (2) noncontact methods, which use the refractive
to allow detection of posterior vitreous detachments, abnormal power of the cornea and a special lens to form a fundus image.
vitreous surfaces, and vitreous floaters. Furthermore, the surface Noncontact fundus examinations are generally performed more
contours of a chorioretinal lesion are more apparent if a narrow rapidly, particularly if only the posterior pole requires evaluation.
beam of light is projected onto the lesion’s surface. If an However, the patient may squeeze the eyelids shut during non-
elevation or depression of a chorioretinal lesion is present, the contact fundus evaluations. With diagnostic contact lenses, the
slit beam on the retina will appear curved rather than straight eyelids cannot be closed, but corneal irritation may result if the

CHAPTER 127
(Fig. 127.13a). In some cases, when there is very little examination is not performed gently. Furthermore, excessive
subretinal fluid, illumination of the retinal vessels with the slit pressure exerted on the eye during a contact lens evaluation may
beam produces shadows of the vessels at the level of the retinal induce vasovagal reactions such as fainting and nausea. These
pigment epithelium (Fig. 127.13b). Such shadows are further side effects rarely occur if the examiner is experienced.
proof of retinal elevation.
NONCONTACT METHODS OF
BIOMICROSCOPIC RETINAL EXAMINATION

PLANOCONCAVE LENSES
A planoconcave lens of high negative optical power, such as a
Hruby lens, is incorporated into some slit-lamp biomicroscopes.
To use this lens, a broad vertical slit beam is first rotated to
illuminate the fundus from virtually a straight-on direction until
the red reflex is clearly seen through the oculars. The lens is
centered over the patient’s cornea, positioned a few centimeters
from the patient’s eye, and focused via a lever until the fundus
comes into view (Fig. 127.14). Some systems focus the lens
more or less automatically. The image formed is upright and
vertical, but the image quality is not uniformly good, especially
near the edges of the field of view. This lens is used almost
exclusively to view the posterior pole, as distortion seriously
degrades the image if the fundus is viewed along any axis other
than the approximate optical axis of the patient’s eye.11

ASPHERICAL LENSES (60 D, 78 D, 90 D) AND


SLIT-LAMP INDIRECT OPHTHALMOSCOPY
A real image of the fundus is formed at the slit lamp several
centimeters in front of the patient’s eye when a high-powered
plus lens is positioned in front of the cornea. This aerial image
of the fundus is magnified by the slit-lamp optics. The resultant
image is real, inverted, and of high quality if a superior high-
power aspherical lens made for this purpose is used. Typically,
indirect biomicroscopic lenses are positioned farther from the
patient’s eye than is the Hruby-type lens. With a lens of lower
power or longer focal length, the image is produced at a location
farther in front of the patient’s eye than with a higher-power
lens. Because some slit-lamp focusing tracks are limited in
travel away from the patient, it may be impossible to obtain a
clear fundus image with certain low-power indirect lenses.
Indirect ophthalmoscopy at the slit lamp is performed by
placing the lens between the thumb and the forefinger, with the
FIGURE 127.13. (a) A slit beam has been directed over a
elbow supported by the slit-lamp table (Fig. 127.15). Because
questionably elevated choroidal lesion. Bending of the beam is
observed, indicating that the lesion is indeed elevated. (b) To detect a
these lenses are of relatively high power, any movement of the
subtle elevation of the sensory retina, the area of suspicion is examiner’s hand induces sizable prism shift movements of the
illuminated by the slit beam. Shadows of the retinal vessels are seen image. Some examiners prefer, therefore, to have the lens
at the pigment epithelial level, indicating the presence of subretinal mounted on a small jointed holder affixed to the slit lamp
fluid and a shallow serous detachment of the sensory retina. (Volk Optical, Mentor, OH). As with indirect ophthalmoscopy, 1683
RETINA AND VITREOUS

a
SECTION 10

FIGURE 127.14. A high-power minus lens placed in front of the


cornea forms a virtual image of the fundus. Moving a lever on the lens
holder focuses the image, which is observed through the slit-lamp
binoculars.

higher-power lenses provide wider fields of view at the expense


of magnification. b
Although noncontact lenses are suitable for viewing the
posterior pole, they do not by themselves provide an adequate view FIGURE 127.15. (a) Binocular indirect ophthalmoscopy at the slit
of the retinal periphery. Scleral depression can be attempted lamp is accomplished by holding a high-power plus lens in front of the
while viewing the fundus through these lenses, but this tech- cornea and focusing the slit lamp on the aerial image that forms
nique requires considerable practice and is technically difficult. anterior to the lens. (b) Lenses of various powers provide the
examiner with an array of magnification and field of view options.
CONTACT LENS METHODS OF
BIOMICROSCOPIC RETINAL EXAMINATION
its surface and the axis of the lens. Through the central posterior
Before placing the diagnostic contact lens on the patient’s cornea, pole portion, the field of view is ~30º for an emmetropic eye.
a drop of a mild anesthetic agent such as 0.5% proparacaine The smallest mirror is used to view the anterior chamber angle
hydrochloride is instilled. A viscous, clear liquid such as methyl- and occasionally the far periphery of the fundus. The middle-sized
cellulose or hydroxypropyl methylcellulose (Goniosol) is placed in mirror is configured to allow viewing of the retinal periphery
the concave portion of the lens to optically couple the lens to the anterior to the equator, and the largest mirror is chosen when
cornea. With the patient looking up, the lids are held open and the area of interest is within the equatorial and posterior
the lens is placed gently on the patient’s cornea (Fig. 127.16). equatorial regions of the fundus (Fig. 127.18).12
Small air bubbles are eliminated by exerting gentle pressure on The beam of the slit lamp should be projected along the radial
the lens. When fundus photography is scheduled directly after axis of the mirror being used. This is achieved by rotating a
removal of the diagnostic contact lens, any residual interface solu- collar or knob on the slit lamp, which, in turn, rotates the slit
tions remaining on the cornea can degrade the retinal image. beam. With the patient gazing straight ahead, the best image is
Hence, many examiners prefer to use low-viscosity BSSs rather obtained when the front surface of the lens is kept perpendicular
than thicker, more tenacious interface liquids. Unfortunately, to the viewing axis of the slit lamp. By rotating the examination
BSSs are of such low viscosity that they spill out of the fundus mirror and readjusting the slit beam, all meridians of the retina
contact lens unless the lens is placed on the cornea in a smooth, can be evaluated.
rapid motion while the patient gazes ahead. Redirection of the patient’s gaze facilitates viewing of more
anteriorly or posteriorly located retinal regions positioned
within a given mirror. If the patient moves her or his eye toward
GOLDMANN THREE-MIRROR LENS the mirror, a more posteriorly located portion of the fundus
The three-mirror lens has a clear central portion for viewing the comes into view. If the eye is moved away from the mirror, more
posterior pole, surrounded by three radially arranged mirrors peripheral areas of the fundus are seen (Fig. 127.19). This change
1684 (Fig. 127.17, left). Each mirror has a different inclination between in image field with a change in the patient’s gaze is especially
Examination of the Retina: Ophthalmoscopy and Fundus Biomicroscopy

FIGURE 127.18. (Left) The Goldmann three-mirror lens has a clear


central zone (D) for examination of the posterior pole. The adjacent
mirrors (A, B, C) are each inclined at different angles to the optical
axis of the lens, providing visual access to different regions of the
fundus, as depicted at right.

CHAPTER 127
a

b
FIGURE 127.19. The patient’s direction of gaze will influence the field
FIGURE 127.16. (a) A diagnostic fundus contact lens containing an
being observed through each mirror of the Goldmann lens. If the
optical coupling fluid is placed on the anesthetized cornea by holding
patient looks toward the particular mirror being used for viewing, a
the lids open while the patient looks up. (b) The patient then gazes
more posterior region of the fundus will be seen.
straight ahead to center the lens, which is held in place with very light
pressure.
important to note when photocoagulating a patient’s eye
through a mirror. The fovea may inadvertently appear within the
treatment mirror of the lens on extreme gaze. Failure to recog-
nize the fovea in this situation can lead to its destruction by
photocoagulation.

THREE-MIRROR LENS EXAMINATION


COUPLED WITH SCLERAL DEPRESSION
A highly magnified view of the ora serrata and the most peri-
pheral retina can be achieved by performing scleral depression
during the three-mirror lens examination. This magnified exam-
ination of the retinal periphery is often easier to accomplish if
the Goldmann lens is placed in a specialized conical holder, which,
in turn, has a small depressor located along its circumference
(Fig. 127.20). The middle-sized or smallest mirror should be
inserted into the position opposite the depressor location. The
lens holder is then held gently but firmly against the patient’s
eye, producing the desired scleral indentation. One type of lens
FIGURE 127.17. Specialized fundus contact examination lenses. holder incorporates an adjustable depressor that can be moved
(Left) Goldmann three-mirror contact lens. (Center) Mainster more posteriorly along the sclera in any meridian.13 Unfortu-
biomicroscopic lens. (Right) Rodenstock panfunduscopic lens. Each nately, many patients find these scleral depression devices very
lens has specific attributes and shortcomings. uncomfortable, especially in inexperienced hands. 1685
RETINA AND VITREOUS

purposes. The image plane is located quite anterior to the lens


surface, however, making it difficult to view the fundi of patients
with hyperopia or exophthalmos when using slit lamps with
limited forward travel.

SPECIALIZED RETINAL EXAMINATION


TECHNIQUES
If a fiber optic probe or other suitable light source is placed against
the sclera in a darkened room, the retina can be visualized using
indirect ophthalmoscopy with the illuminating scope light
turned off. This examination technique (transillumination
ophthalmoscopy) requires that light be transmitted through the
FIGURE 127.20. Placement of the Goldmann lens into a conical sclera and choroid. If an eye contains a solid pigmented mass,
holder containing a scleral depressor at its periphery allows the transillumination is useful in that the tumor will appear dark,
examiner to perform scleral depression while using the slit-lamp whereas other choroidal lesions, such as a choroidal detachment,
biomicroscope. Two of these devices are illustrated. will be much more brightly illuminated.
Alternately, a transillumination light can be placed on the
SECTION 10

cornea so that it shines through the pupil. The examiner then


observes the surface of the sclera, which should glow a uni-
GOLDMANN POSTERIOR FUNDUS CONTACT formly orange color. A pigmented tumor or foreign body within
LENS the choroid will prevent the transillumination light from illumi-
If the examiner is primarily interested in studying the posterior nating the sclera beneath it, and a dark shadow will be present
retina, a small lightweight lens incorporating a peripheral flange within the affected scleral quadrant. In eyes that are lightly
at the point at which it touches the eyelids is often preferred over pigmented, a bright transilluminator may be applied directly to
a larger, bulkier lens. The flange and smaller contact diameter the sclera to achieve a similar effect (Fig. 127.21).
make this lens more difficult to dislodge if the patient squeezes
the eyelids.

PANFUNDUSCOPIC LENS
The panfunduscopic lens (Rodenstock, Munich, Germany) (see
Fig. 127.17, right) consists of a meniscus lens coupled with a
spherical lens located within the same lens holder housing.
When the meniscus lens is placed on the cornea, the resultant
image of the fundus is inverted, real, minified, and located well
forward of the anterior lens surface. Magnification provided by
the slit lamp counteracts the minification produced by the lens
configuration. A wide-angle view of the fundus in an emmetropic
eye, extending from the fovea to the equatorial region, is produced
by such a panfunduscopic lens.14 This device also facilitates the
examination of the fundus through a poorly dilated pupil. How-
ever, panfunduscopic lenses are used more often during laser
photocoagulation than for diagnostic purposes, primarily
because the image of the retina is small and the view through
the peripheral portions of this lens is not excellent.

MAINSTER LENS
The Mainster lens (Ocular Instruments, Bellevue, WA) (see
FIGURE 127.21. Transillumination. The globe is illuminated, through
Fig. 127.17, center) provides a somewhat reduced field of view
the sclera in this illustration, via a fiberoptic light source placed on the
(45º) compared with the panfunduscopic lens but also produces globe externally or on the cornea. In a darkened room, the sclera is
less minification.15 Resolution with this lens is sufficient to detect observed, glowing yellow-orange. A dark choroidal lesion will block
retinal thickening from macular edema. The image size is com- the transmission of light through the eye wall and will appear as a
parable with that provided by the Goldmann lens, making the dark shadow surrounded by the red glow of the remainder of the
Mainster lens versatile for diagnostic as well as therapeutic sclera.

REFERENCES
1. Doyle AC: A scandal in Bohemia. In: The 3. Rubin ML: Magnification; practical 5. Rosenthal ML, Fradin S: The technique
complete works of sherlock holmes. Garden instruments: the indirect ophthalmoscope. of binocular indirect ophthalmoscopy.
City, NY: Doubleday; 1892–1927:162. In: Optics for clinicians. 2nd edn. Highlights Ophthalmol 1966;
2. Thompson HS: The pupil. In: Moses RA, Gainesville, FL: Triad; 1974:235, 298. 9:179–257.
Hart WM, eds. Adler’s physiology of the 4. Schepens CL: Methods of examination. 6. Schepens CL: Progress in detachment
eye, clinical application. St. Louis: CV In: Retinal detachment and allied diseases. surgery. Trans Am Acad Ophthalmol
Mosby; 1987:321. Philadelphia: WB Saunders; 1983:99–133. Otolaryngol 1951; 55:607–615.

1686
Examination of the Retina: Ophthalmoscopy and Fundus Biomicroscopy

7. Friberg TR: Clinical experience with a 11. Tolentino FI, Schepens CL, Freeman HM: ora serrata and pars plana. Am J
binocular indirect ophthalmoscope laser Instrumentation. In: Vitreoretinal Ophthalmol 1967; 64:467–468.
delivery system. Retina 1987; 7:28–31. disorders. Diagnosis and management. 14. Michels RG, Wilkinson CP, Rice TA: Pre-
8. Rubin ML: The optics of indirect Philadelphia: WB Saunders; operative evaluation. In: Michels RG, ed.
ophthalmoscopy. Surv Ophthalmol 1964; 1976:45–70. Retinal detachment. St. Louis: CV Mosby;
9:449–464. 12. Benson WE: Fundus examination and 1990:325–378.
9. Chignell AH: Techniques of examination. pre-operative management. In: Retinal 15. Mainster MA, Crossman JL, Erickson PJ,
In: Retinal detachment surgery. 2nd edn. detachment: diagnosis and management. Heacock GL: Retinal laser lenses:
Berlin: Springer-Verlag; 1988:14–25. 2nd edn. Philadelphia: JB Lippincott; magnification, spot size, and field of view.
10. Kemp P, ed: The oxford companion to 1988:85–111. Br J Ophthalmol 1990; 74:177–179.
ships and the sea. London: Oxford 13. Eisner G: Attachment for Goldmann three-
University Press; 1976:346. mirror contact glass. For examination of the

CHAPTER 127

1687
CHAPTER

128 Principles of Fluorescein Angiography


Timothy J. Bennett, David A. Quillen, and James D. Strong

Since its introduction in the early 1960s, fluorescein angiography Fluorescein sodium absorbs blue light, with peak excitation
has become an essential tool in the understanding, diagnosis, occurring at wavelengths between 465 and 490 nm. The
and treatment of retinal disorders. While direct examination of resulting fluorescence occurs at the yellow–green wavelengths of
the fundus with slit-lamp biomicroscopy and indirect ophthal- 520–530 nm (Fig. 128.1). Dye concentration and pH can affect
moscopy is indispensable in the diagnosis of retinal disease, the intensity of fluorescence. Maximum fluorescence occurs at
fluorescein angiography provides additional information a pH of 7.4, but the pH of fluorescein sodium for angiographic
concerning the anatomy, physiology, and pathology of the retina use is adjusted to a range of 8–9.8 for stability. In powdered
and choroid. This diagnostic procedure utilizes a specialized or concentrated solution form, fluorescein sodium appears
fundus camera or scanning laser ophthalmoscope (SLO) to orange–red in color. Fluorescence is detectable in concentrations
capture rapid-sequence photographs of the retina following an between 0.1% and 0.0000001%. In broad-spectrum illumi-
intravenous injection of fluorescein sodium. Photographic or nation, diluted fluorescein sodium appears bright yellow–green
video images taken as the dye courses through the eye can demon- in color. When illuminated with blue light, the yellow–green
strate abnormalities within the neurosensory retina, pigment color intensifies dramatically.
epithelium, sclera, choroid, and optic nerve, providing clinically The fluorescent properties of this dye have made it useful
useful information for nearly the entire spectrum of posterior in a variety of industrial, scientific, military, and medical appli-
segment disorders. cations. Fluorescein sodium was the first fluorescent dye used
for water tracing purposes.4 It has been used as a visible marker
CHARACTERISTICS OF FLUORESCEIN for search-and-rescue operations, to track and measure flow

FLUORESCENCE
Fluorescein angiography is an application of the physical phe-
nomenon of fluorescence.1 Fluorescence is a type of photolumi-
nescence that occurs when susceptible molecules known as
fluorophores absorb electromagnetic energy, temporarily
exciting them to a higher energy state. As the molecules return
to their original energy level, they emit light of a different,
usually longer wavelength. Unlike phosphorescence, which con-
tinues to occur after the excitation source is removed, fluores-
cence requires continuous excitation. Once the excitation
source is removed, emission of fluorescence stops almost
immediately (10–8 s).
Fluorescence occurs naturally in certain compounds and may
occasionally be observed in the human eye. Optic nerve drusen,
astrocytic hamartomas, lipofuscin pigments in the retina, and
the aging human lens are all believed to exhibit natural fluores-
cence that can be documented with various photographic
techniques.

FLUORESCEIN SODIUM
Although commonly referred to as fluorescein, the dye used
for fluorescein angiography is actually fluorescein sodium
(C20H10Na2O5).2 It is the water-soluble salt of fluorescein, also
known as resorcinolphthalein sodium, or uranine. A member of
the xanthene group of dyes, it is a highly fluorescent chemical
compound synthesized from the petroleum derivatives resor- FIGURE 128.1. Excitation and emission. Representative excitation
cinol and phthalic anhydride.3 The dye was first synthesized in and emission curves of fluorescein sodium. Peak excitation occurs at
1871 by Adolf von Baeyer, who later received the Nobel Prize in wavelengths between 465 and 490 nm (blue–green). Peak
Chemistry (1905) for his work in organic dyes. fluorescence occurs at wavelengths of 520–530 nm (green–yellow). 1689
RETINA AND VITREOUS

dynamics of water sources, map subterranean water courses, In 1959, two medical students, Harold Novotny and David
track hazardous spill dispersion patterns, identify point sources Alvis worked on a research project to develop a photographic
of pollution, and to detect leaks or obstructions in plumbing technique to estimate blood oxygen concentrations in the retinal
and sewage systems. In fact, its common use in industrial vascular bed as a visible segment of the cerebral circulation.
plumbing led a plumbers union to start the tradition of using They mixed fluorescein with a blood sample and measured it
fluorescein to stain the Chicago River green for the annual with a spectrofluorometer to determine the excitation and
St Patrick’s Day celebration. emission wavelengths of fluorescein. They found the peak
Many of the medical and ophthalmic applications of excitation of fluorescein to be 490 nm and peak emission,
fluorescein are analogous to its uses in plumbing or industrial 520 nm. Then they equipped a fundus camera with broadband
flow dynamics. For example, it has been used for intraoperative absorption filters, a Kodak Wratten 47B for excitation and a
assessment of blood flow in surgical resections and grafts with Kodak Wratten 58 green for the barrier. They confirmed the
a Wood’s Light to excite fluorescence.5–9 It has also been employed technique on rabbits and then flipped a coin to see who would
as an intraoperative predictor of intestinal viability,10,11 as an be the first human ‘patient’. Alvis lost, so he was the subject of
indicator of local perfusion in gangrene or severe burns,12,13 and the first successful fluorescein angiogram in a human. After
to monitor perfusate leak in systemic tissues during isolated their initial success, they refined the technique on numerous
limb perfusion with high dose chemotherapeutic agents.14 In patients with diabetes and hypertension. The landmark paper
ophthalmology, topical application of fluorescein sodium is describing their technique was published in the journal
routinely used for applanation tonometry, and as a vital stain in Circulation in 1961, after being rejected by the ophthalmic
SECTION 10

the documentation of ocular surface disorders such as corneal literature as unoriginal work.20 Following their report, several
ulcers, abrasions, or other epithelial defects.15 It is sometimes investigators studied various clinical applications of angiography,
used to determine tear film breakdown time, check the fit of quickly establishing the intrinsic value of the technique.21
contact lenses, verify the patency of lacrimal passageways, and Further innovations were made gradually, including improved
to detect leakage of aqueous humor from corneal or con- filter combinations, fast recycling flash units, digital imaging
junctival wounds using the Seidel Test. techniques, and SLOs, but the basic principles described by
Novotny and Alvis remain unchanged (Fig. 128.2).
HISTORY
In 1881, just a decade after the discovery of fluorescein, Ehrlich INDICATIONS AND USES
observed anterior chamber fluorescence following an injection The most common uses of fluorescein angiography are in retinal
of fluorescein. In 1910, Burke described fluorescent staining of or choroidal vascular diseases such as diabetic retinopathy, age-
retinal and choroidal lesions in white light, after ingestion of a related macular degeneration, hypertensive retinopathy, and
mixture of fluorescein in coffee.16 In the late 1950s, Flocks and vascular occlusions (Table 128.1). For the most part, these are
co-workers investigated methods to determine retinal circulation clinical diagnoses. The angiogram is used to determine the
times with various dyes, including trypan blue and fluorescein.17,18 extent of damage, to develop a treatment plan or to monitor the
Using a 16 mm movie camera attached to a fundus camera with results of treatment. In diabetic retinopathy the angiogram is
a carbon arc light, they succeeded in obtaining fluorescein angio- useful in identifying the extent of ischemia, the location of
grams in cats, but were unsuccessful in attempts on humans microaneurysms, the presence of neovascularization and the
due to insufficient light. MacLean and Maumenee performed extent of macular edema. In age-related macular degeneration,
fluorescein angioscopy with a blue exciter light to diagnose a angiography is useful in identifying the presence, location and
choroidal hemangioma following intravenous injection in 1960.19 characteristic features of choroidal neovascularization for

a b

FIGURE 128.2. Fluorescein angiography, then and now. (a) Photographs from the first fluorescein angiogram by Novotny and Alvis demonstrate
significant crossover of their simple filter combination, resulting in incomplete blockage of the exciting wavelengths. Results were also limited by
the slow recycle time of the flash unit, more than 12 s between exposures. (b) Modern high-resolution digital fluorescein angiogram
demonstrates improvement in technical quality, but the basic technique developed by Novotny and Alvis is essentially the same.
1690 (a) Courtesy of Indiana University Medical Center.
Principles of Fluorescein Angiography

fluorescein include discoloration of the urine for 24–36 h and a


TABLE 128.1. Common Diagnostic Uses and Indications for
slight yellow skin discoloration that fades within a few hours.
Fluorescein Angiography
Nursing mothers should be cautioned that fluorescein is also
Diabetic retinopathy excreted in human milk.26
Age related macular degeneration
Subretinal neovascular membrane from other causes (myopic COMPLICATIONS AND ADVERSE REACTIONS
degeneration, histoplasmosis, etc.)
Central retinal vein occlusion Key Features
Branch retinal vein occlusion Fluorescein angiography is well-tolerated by the vast majority of
individuals. The most common adverse reactions are transient
Central serous chorioretinopathy nausea and occasional vomiting.
Cystoid macular edema
Hypertensive retinopathy Fluorescein sodium is well tolerated by most patients, but
Central retinal artery occlusion angiography is an invasive procedure with an associated risk of
complication or adverse reaction (Table 128.2). Use of fluorescein
Branch retinal artery occlusion
sodium may be contraindicated in patients with a history of

CHAPTER 128
Retinal arterial macroaneurysms allergic hypersensitivity to fluorescein. Although generally con-
Pattern dystrophies of the retinal pigment epithelium sidered safe for patients receiving dialysis, one manufacturer
of fluorescein suggests using half the normal dose in dialyzed
Choroidal tumors patients.27 There are no known risks or adverse reactions asso-
Chorioretinal inflammatory conditions ciated with pregnancy but most practitioners avoid performing
Hereditary retinal dystrophies
fluorescein angiography in pregnant women, especially in their
first trimester.28–30
Historically, adverse reactions occur in 5–10% of patients and
range from mild to severe.31–37 Anecdotal evidence suggests a
possible treatment with laser photocoagulation, photodynamic lower incidence of reaction in recent years and the first large
therapy, or antiangiogenic medications. study conducted in over a decade seems to confirm that,
Fluorescein angiography can be very useful in certain degen- reporting a frequency of adverse reaction of just over 1%.38
erative and inflammatory conditions. Some of these conditions Continued improvements in manufacturing processes and
exhibit characteristic fluorescence patterns, which support the implementation of tighter pharmacopeial standards are credited
diagnosis. For example, Stargardts Disease exhibits a ‘silent with this reduction and may lead to lower rates of reaction in
choroid’ and a central bulls-eye fluorescence pattern in the macula the future.39
while acute posterior multifocal placoid pigment epitheliopathy Transient nausea and occasional vomiting are the most com-
(APMPPE) demonstrates a characteristic ‘block early, stain late’ mon reactions and require no treatment. These mild reactions
pattern. typically occur 30–60 s after injection and last for ~1–2 min.
Angiography has long played a role in advancing the under- Fortunately, they seldom compromise the diagnostic quality of
standing of retinal vascular disorders and potential treatment the angiogram. The incidence of nausea and vomiting seems to
modalities. A number of multicenter clinical trials utilize be related to the volume of dye and rate of injection. A relatively
fluorescein angiography in investigating new treatment options slow rate of injection often reduces or eliminates this type of
in diabetic retinopathy, age-related macular degeneration, and reaction but can adversely affect image quality and alter arm-
retinal vein occlusions.22 to-retina circulation times. Premedication with promethazine

DOSAGE AND ADMINISTRATION


Fluorescein angiography is performed by injecting fluorescein
sodium dye as a bolus into a peripheral vein. The normal adult TABLE 128.2. Complications and Adverse Reactions
dosage is 500 mg, and is typically packaged in doses of 5 mL of Extravasation of dye
10% or 2 mL of 25%. For pediatric patients, the dose is adjusted
to 35 mg/10 pounds of body weight.23 Upon entering the Transient nausea
circulation, ~80% of the dye molecules bind to plasma proteins, Vomiting
which significantly reduces fluorescence because the free electrons
Pruritis
that form this chemical bond are subsequently unavailable for
excitation.1 The remaining unbound or free fluorescein Urticaria
molecules fluoresce when excited with light of the appropriate Bronchospasm
wavelength. With a molecular weight of 376, fluorescein diffuses
freely out of all capillaries except those of the central nervous Laryngeal edema
system, including the retina. Anaphylaxis
The dye is metabolized by the kidneys and is eliminated
Hypotension
through the urine within 24–36 h of administration. During
this period of metabolism and elimination, fluorescein has the Syncope
potential to interfere with clinical laboratory tests that use fluo- Seizures
rescence as a diagnostic marker.24,25 To avoid any false readings,
it may be prudent to schedule clinical lab tests either before the Myocardial infarction
angiogram, or postpone testing for a day or two to allow Cardiac arrest
sufficient elimination of the dye. Side effects of intravenous 1691
RETINA AND VITREOUS

hydrochloride or prochlorperazine may prevent or lessen the


severity of nausea and vomiting in patients with a history of
previous reactions to fluorescein, but is rarely needed and one
study noted a higher frequency of these reactions in patients
that had been premedicated.35 Some patients report a strong
taste sensation or hypersalivation following injection of fluorescein.
Moderate reactions occur less frequently, affecting less than
2% of patients that undergo angiography. Allergic reactions such
as pruritus or urticaria can be treated with antihistamines, but
any patient who experiences these symptoms should be observed
carefully for the possible development of anaphylaxis. The
advisability of performing angiograms in patients with a history
of allergic reaction to fluorescein should be considered carefully,
as allergic sensitization to the dye can increase with each FIGURE 128.3. Accidental arterial injection. Unusual fluorescein
subsequent use. Patients with previous history of mild allergic staining pattern distal to the injection site in a patient with an AV
reaction to fluorescein can be pretreated with an antihistamine, malformation in the forearm. Photograph was taken with a blue filter
such as diphenhydramine, 30–40 min prior to any subsequent over the light source to excite fluorescence to illustrate the distribution
angiograms to limit allergic response, although this may not of dye.
SECTION 10

prevent serious reactions.40 Vasovagal attacks happen in some


patients,41 most likely due to anxiety about the procedure or EQUIPMENT AND TECHNIQUE
their ocular condition. Usually the angiogram needs to be aborted
or postponed, but some patients are able to tolerate the angio- The instrumentation used for fluorescein angiography must
gram during the initial stages of a syncopal episode. However, be capable of delivering the proper excitation wavelengths and
the drop in blood pressure and heart rate can dramatically alter capturing rapid sequence images of the retina as the dye courses
the angiographic results.42 through the vasculature. The modern mydriatic fundus camera
More severe reactions are rare, but include laryngeal edema, is the tool most commonly used for this purpose, but SLOs and
bronchospasm, anaphylaxis, tonic–clonic seizure, myocardial some specialized wide-field fundus cameras can also be used.
infarction and cardiac arrest.43–47 The overall risk of death from Fluorescein angiography can be performed using 35 mm black-
fluorescein angiography has been reported as one in 222 000.35 and-white panchromatic films or with digital cameras. A
Although life-threatening reactions during angiography are rare, number of major ophthalmic instrument manufacturers produce
angiographic facilities and personnel should be properly equipped fluorescein-ready fundus cameras in both film and digital
and prepared to manage serious reactions to the procedure. A configurations. Third-party vendors offer digital conversion
resuscitative crash cart and appropriate agents to treat severe solutions for a variety of film-only cameras.
reactions should be readily available including epinephrine Although fluorescein angiography could be done in color,
for intravenous or intramuscular use, soluble corticosteroids, black-and-white imaging offers increased light sensitivity and
aminophylline for intravenous use, oxygen, and airway instru- ease of contrast enhancement to compensate for the low levels
mentation. It is generally recommended that a physician be of fluorescence in the bloodstream. Film-based angiography
present or available during angiographic procedures. requires either the use of a processing service, or access to a
Extravasation of fluorescein dye during the injection can be darkroom for processing films on-site. Films with an ISO film
a serious complication of angiography. With a pH of 8–9.8, speed rating of 400 or higher are used and push processed in
fluorescein infiltration can be quite painful. If fluorescein dye high contrast developers to increase both light sensitivity and
extravasates, cold compresses should be placed on the affected contrast. Films developed in this way exhibit an increase in the
area for 5–10 min, and the patient should be reassessed until silver halide grain structure and a reduction in apparent
edema, pain, and redness resolve. Serious complications are resolution. These films however are often more forgiving of
more likely to occur when large amounts of dye extravasate. exposure inconsistencies than are digital cameras
Sloughing of the skin, localized necrosis, subcutaneous granuloma,
and toxic neuritis have been reported following extravasation of
fluorescein.48–50 To avoid these problems, continual observation FUNDUS CAMERA
of the injection site during the course of the injection and Successful retinal imaging relies on the interaction between the
monitoring the patient for pain is recommended. Accidental optics of the fundus camera with those of the eye itself. Fundus
arterial injections are rare, but can be quite painful. The dye cameras utilize an aspheric design, that when combined with
remains concentrated and stains the effected extremity with the optics of the subject eye, matches the plane of focus to the
little or no dye reaching the retinal vasculature (Fig. 128.3). With curvature of the fundus. The focus control of the fundus camera
proper technique, these complications of injection can usually is used to compensate for refractive errors in the subject eye.
be avoided. Conditions such as myopia or astigmatism are routinely
In cases when venous access is severely compromised or the encountered and many fundus cameras have additional controls
patient is known to be highly allergic to the dye, fluorescein can to compensate for these optical irregularities in patients’ eyes.
be administered orally.51,52 Although oral fluorescein adminis- The fundus-illuminating beam is delivered axially, through the
tration is typically well tolerated, severe adverse reactions can image forming optics and filters of the fundus camera. Pharma-
occur.53,54 Due to the slow absorption rate, an early transit cologic dilation is necessary to obtain adequate illumination. A
sequence is not possible. The resulting images are less than xenon-filled flash tube delivers a brief burst of intense light to
ideal, but have traditionally provided limited diagnostic expose the photographs.
information in conditions where late phase photographs are Fundus cameras are often categorized by their optical angle of
helpful, such as cystoid macular edema. The use of optical view (Fig. 128.4). An angle of 30–40° is considered the normal
coherence tomography (OCT) to detect the presence of edema angle of view for documenting macular detail and creates a film
or fluid within the retinal tissues has essentially eliminated this image ~2.5 times life size. Fixed-angle cameras usually offer the
1692 use of oral fluorescein. sharpest optics, but variable-angle cameras provide wide-angle
Principles of Fluorescein Angiography

a b c

FIGURE 128.4. Fundus camera angle of view. Images of a patient with age-related macular degeneration taken with a variable angle fundus
camera at (a) 50°, (b) 35°, (c) 20° magnification settings.

CHAPTER 128
capabilities between 45° and 60°. Wide-angle cameras need to band of wavelengths generated by fluorescein will expose only
illuminate a large area of retina, requiring a more-widely dilated the green channel pixels in RGB color sensors, reducing resolution
pupil to accommodate a larger ring of light. through interpolation. However, both monochrome and color
Fundus cameras equipped for fluorescein angiography have a CCDs with pixel counts of three Megapixels or above surpass
timer that records the angiographic sequencing on each frame of the spatial resolution of the 400 speed films commonly employed
the study, a matched pair of exciter and barrier filters and a fast for film-based angiography. Resolution comes at a cost in terms
recycling electronic flash tube that allows a capture rate of up to of light efficiency. High-resolution digital sensors require more
one frame per second. Narrow-band pass-interference filters are light for proper exposure and flash settings often need to be
utilized to allow maximum transmission of peak wavelengths, increased. New high-transmission filter sets have been developed
while minimizing any crossover of transmission curves. The to improve light efficiency and performance with high-resolution
exciter filter transmits blue–green light at 465–490 nm, the peak digital sensors.
excitation range of fluorescein. The barrier filter transmits a Digital imaging offers several distinct advantages over tradi-
narrow band of yellow at fluorescein’s peak emission range tional film-based angiography. Computer technology offers a
of 520–530 nm. The barrier filter effectively blocks all visible variety of powerful tools that can be used to enhance diagnostic
wavelengths but the specific color of fluorescein. information. Software tools provide adjustments for brightness,
Images are captured either with high-speed black-and-white contrast and sharpness. Digital analysis enables measurement
35 mm panchromatic film or electronically, with a charge- of pathologic structures, digital overlays can be used to identify
coupled device (CCD) and computerized system for digital potential changes in lesion size in serial photographs, and
imaging. Increasingly, the use of digital imaging is replacing the multiple fields can be linked together to form composite wide-
majority of film-based systems. field images. Images can be stored on magnetic or optical media
like CD or DVD-ROMs and transmitted electronically across
computer networks for remote viewing or storage on servers.
DIGITAL IMAGING Commercially available digital angiography systems typically
conform to the Digital Imaging and Communications in Medicine
Key Features (DICOM) Standards. This is a set of universal standards for
Digital angiography has become the imaging modality of choice
transferring diagnostic images and associated information
based on the ability to capture, process, enhance, store, and
distribute images electronically.
between devices manufactured by different vendors.56–59 Con-
formance to these standards facilitates connectivity to picture
archiving and communication systems (PACS) used in radio-
The ophthalmic community was quick to adopt digital imaging logy, and integration with electronic medical records and other
technology for fluorescein angiography. Commercial digital hospital information systems.
systems designed specifically for fluorescein angiography and In addition to well-known advantages in capturing, processing,
retrofitted to existing film-capable fundus cameras began to enhancing, storing, and distributing images electronically, having
appear on the market as early as 1983.55 This configuration instant access to digital images can increase clinical efficiency
continues to be the most common type of instrumentation used and enhance patient education opportunities through the ability
for angiography, offering choice and flexibility between film and to review images on large screen computer monitors with the
digital camera backs. In recent years, new instruments have patient. Another significant advantage to digital imaging is that
been developed that rely entirely on digital capture technology it can shorten the learning curve for novice angiographers trying
for angiography. Digital-only devices include SLOs and special- to master the complex techniques of angiography. Having instant
ized wide-field retinal imaging devices that can be configured for feedback allows the operator to adjust exposure settings and
fluorescein angiography. Digital imaging hardware and software camera alignment to correct any flaws in technique.
continues to evolve and improve in quality, and we are likely to Despite these advantages, the high initial cost of digital
see more digital-only devices for angiography in the future. systems has prevented them from being employed universally,
Spatial resolution in current retinal imaging systems varies although they are now more common than film-based systems.
from 800 µ 600 to over 3000 µ 2000 pixels. Monochrome digital A pair of surveys looking at tasks performed by retinal angio-
backs are considered better for angiography than their color graphers indicated that over 70% of respondents (71% in 2002,
counterparts since they are usually more light sensitive, and all 72% in 2004) are utilizing digital technology for some or all of
pixels are available for exposure to fluorescence. The narrow their fluorescein angiograms.60,61 1693
RETINA AND VITREOUS

photographs as well as black-and-white monochromatic green


SCANNING LASER OPHTHALMOSCOPY filter images are routinely taken as baseline views before admin-
Fluorescein angiography can also be recorded using a confocal istering the dye. The early transit phase is the most critical part
SLO in place of the conventional fundus camera. This complex of the angiogram and usually lasts less than a minute. Before
instrument uses a laser beam of the appropriate excitation wave- injecting the dye, the illuminating beam of the fundus camera
length to scan across the fundus in a raster pattern to illuminate is centered within the dilated pupil. The angiographer then
successive elements of the retina, point-by-point.62,63 The laser can prefocuses the camera on the appropriate area of interest. The
deliver a very narrow wavelength band for more efficient excitation dye is administered as a bolus injection, typically through a
of fluorescence than the flash illumination generated by a fundus small gauge needle into an antecubital vein. The timer is started
camera flash tube. A confocal aperture is positioned in front of the and photography commences. The arm-to-retina circulation time
image detector at a focal plane conjugate to the retina, effectively varies, but normally takes 10–12 s. Experienced angiographers
blocking non image-forming light. The confocal optical system and anticipate the initial appearance of the dye and begin the photo-
laser illumination combine to produce high contrast, finely detailed graphic sequence before the dye is visible. Images are routinely
images. The laser scan rate is synchronized at a frame rate captured at a rate of one frame per second until maximum
compatible with digital video display, providing a continuous fluorescence occurs. During this dynamic early phase only one
high-speed representation of the flow dynamics of the retina eye can be captured. After completion of the early phase, photo-
and choroid. This can be especially useful when documentation graphs of the fellow eye or other areas of interest in the primary
of the very early filling stages is necessary, such as in identifi- retina can be taken.
SECTION 10

cation of choroidal neovascular (CNV) feeder vessels. Over the next few minutes, the appearance of the dye stabilizes
The SLO lessens the need for pupillary dilation and patients and begins to slowly fade. The angiographer can capture appro-
easily tolerate the low light level of the laser. SLO technology priate views as necessary without the urgency needed during the
can also be used for indocyanine green (ICG) angiography of the early phase. Late phase photographs are taken as the dye dissipates,
choroidal vasculature, as well as simultaneous fluorescein and anywhere from 7 to 15 min after injection.
ICG angiography, and fundus autofluorescence (FAF) of retinal Many facilities develop disease specific protocols for both
pigment abnormalities. The major drawback of scanning laser sequencing and field of view. For example, peripheral shots of
technology is the high cost of the equipment. the retina are routinely taken after the early transit in diabetic
retinopathy, whereas the macula is the major area of interest in
age-related macular degeneration and peripheral views are not
STEREO IMAGING usually necessary. The angiographer often adjusts the specific
Stereo imaging techniques can be used during angiography to protocol based on visible changes that may occur as the
enhance diagnostic information. Stereo images provide a visual angiogram progresses.
sense of depth that is particularly useful in identifying the histo-
pathologic location of angiographic findings within the retina.
Use of this technique in retinal photography dates back as far as QUALITY ISSUES AND THE ROLE OF THE
1909, but it wasn’t until the 1960s that stereo photography ANGIOGRAPHER
became widely employed after Lee Allen described a practical Some ophthalmologists perform their own angiography, but this
technique for sequential stereo fundus photography.64 Stereo is an exception rather than the rule. Most facilities employ a
imaging is a standard protocol for many clinical trials inves- photographer or technician dedicated to performing ophthalmic
tigating treatment of retinal diseases.22 photography procedures.
Stereo separation is achieved by laterally shifting the fundus Quality angiographic results rely on a number of factors. The
camera a few millimeters between sequential photographs. The skill of the angiographer and the optical and mechanical quality
lateral shift causes the illuminating beam of the fundus camera of the instrumentation can have a direct effect on results, but
to fall on opposite slopes of the cornea. The resulting cornea- there are a number of common factors that can adversely affect
induced parallax creates a hyperstereoscopic effect that is evident angiographic quality. Media opacities can cause illumination
when the sequential pair of photographs is viewed together. artifacts and blurring of the images. Inadequate pupillary dilation
There are a variety of optical stereo viewers available for viewing reduces light reaching the retina, causing uneven illumination.
side-by-side 35 mm film images on a light box, or digital images Excess topical fluorescein staining of the cornea from the initial
on a computer monitor. All stereo viewing devices are designed patient workup can compete with and degrade retinal fluorescence.
to deliver the separate stereo images simultaneously but inde- Inadequate patient cooperation such as poor fixation or inability
pendently to each eye allowing the brain to fuse the pair. to hold steady during the procedure often results in loss of
The common use of digital projection has resulted in a resur- field definition during the important early transit phase.
gence in stereo projection using the color anaglyph technique.65 Extravasation of the dye not only causes discomfort to the
This technique works very well with grayscale angiographic patient, but the resulting incomplete dose reduces the amount
images. A stereo pair can be digitally color encoded as a single of dye in the retinal vessels. Some of these causes are beyond
image using image-editing software. Instead of employing optics the direct control of the angiographer, but every attempt should
or polarization to display a stereo effect, the anaglyph method be made to minimize their detrimental effects in order for each
uses complementary colors to encode and deliver stereo infor- angiogram to be of adequate and consistent diagnostic quality
mation. Digital imaging software facilitates accurate and (Table 128.3).
effective registration of anaglyph stereo pairs. Anaglyph stereo Since fluorescein angiography is a dynamic process, success-
pairs can be viewed with inexpensive eyewear either on a ful results depend on complete preparation before the dye is
computer monitor or projected with an LCD projector and are injected. Many angiographers follow a specific protocol or check-
useful in educational settings. list to ensure that everything is ready. Good communication
between the ophthalmologist and angiographer is essential to
ensure that maximum diagnostic information is obtained. The
SEQUENCING photographic timing sequence and the angiographer’s ability to
Proper sequencing of the angiographic series is essential in adapt to changing conditions are also important elements in
1694 obtaining maximum diagnostic information. Color fundus producing quality angiographic results. Experience is invaluable,
Principles of Fluorescein Angiography

TABLE 128.3. Factors Effecting Image Quality

The skill of the angiographer


Optical/mechanical quality of the instrumentation
Presence of media opacities
Absorption of blue excitation light by cataracts
Residual topical fluorescein staining of the cornea
Inadequate pupillary dilation
Poor fixation
Inadequate patient cooperation
Extravasation of the dye

especially in managing the patient if complications occur during

CHAPTER 128
the critical early phase of the study.
FIGURE 128.5. Blood–retinal barrier. The inner and outer
Unfortunately, there is very little education available in
blood–retinal barriers are demonstrated in this photomicrograph. The
fluorescein angiography. In the absence of formal education, fluorescein is retained within the retinal vessels by the endothelial cell
certification plays an important role in developing competent tight junctions and the choriocapillaris by the RPE.
practitioners in angiography. The Ophthalmic Photographers Reprinted from Eagle RC Jr: Mechanisms of maculopathy. Ophthalmology 1984;
Society offers a voluntary certification program in fluorescein 91:613-625.
angiography that has established standards of competence in
angiography. The Certified Retinal Angiographer (CRA)
program was established in 1978. The program is accredited by
the National Commission for Certifying Agencies and has
certified over 800 individuals to date. Although certification is
not mandatory, the CRA credential offers some assurance of
competence and safety to both patient and physician.
The responsibility for injecting the dye sometimes falls to the
angiographer or a technician. In some practice settings this
makes sense. There are however, some legal issues associated
with unlicensed personnel performing fluorescein injections.66
It is generally recommended that angiographic facilities check
their current state or local laws regarding the credentialing
requirements of personnel performing intravenous injections.67

INTERPRETATION
Fluorescein angiography records the dynamic interaction of
fluorescein with both normal and abnormal anatomic structures
of the ocular fundus. A thorough understanding of the circu-
lation phases and appearance of the dye in a normal eye is
essential for interpretation of abnormalities.
FIGURE 128.6. Patchy choroidal filling. In the posterior fundus, the
THE NORMAL ANGIOGRAM choriocapillaris is arranged in a mosaic of lobules that contributes to
the patchy choroidal fluorescence often seen in the early phases of
In a normal eye, the retinal blood vessels and the retinal the angiogram.
pigment epithelium (RPE) both act as barriers to fluorescein
leakage within the retina (Fig. 128.5). The tight junctions of the
endothelial cells in normal retinal capillaries make them imper- fovea (foveal avascular zone) contribute to the relative hypo-
meable to fluorescein leakage. The tight cellular junctions of fluorescence of the center of the macula (Fig. 128.7).
the healthy RPE provide an outer blood–retinal barrier
preventing the normal choroidal leakage from penetrating the Early Phase
retinal tissues. The early phase of the angiogram can be divided into distinct
Additional anatomical features contribute to the interpretation circulation phases that are useful for interpreting the results
of the fluorescein angiogram. The choriocapillaris is the capillary- (Fig. 128.8a–d).
rich layer of the choroid characterized by fenestrated capillary
walls that leak fluorescein dye freely into the extravascular Choroidal flush
space within the choroid. In the posterior fundus, the chorio- In a normal patient, the dye appears first in the choroid ~10 s
capillaris is arranged in a mosaic of lobules that accounts for the following injection. The major choroidal vessels are imperme-
patchy choroidal fluorescence often seen in the early phases of able to fluorescein, but the choriocapillaris leaks fluorescein dye
the angiogram (Fig. 128.6). The taller, more pigmented retinal freely into the extravascular space. There is usually little detail
pigment epithelial cells along with the presence of xanthophyll in the choroidal flush as the RPE acts as an irregular filter that
pigment and absence of retinal capillaries in the center of the partially obscures the view of the choroid. If a cilioretinal artery 1695
RETINA AND VITREOUS

is present, this fills along with the choroidal flush as both are
supplied by the short posterior ciliary arteries.

Arterial phase
The retinal arterioles typically fill one to 2 s after the choroid;
therefore, the normal ‘arm-to-retina’ circulation time is ~12 s.
A delay in the arm-to-retina time may reflect a problem with
the fluorescein dye injection or circulatory problems with the
patient including heart and peripheral vascular disease.

Arteriovenous phase
Complete filling of the retinal capillary bed follows the arterial
phase and the retinal veins begin to fill. In the early
arteriovenous phase, thin columns of fluorescein are visualized
along the walls of the larger veins (laminar flow). These
columns become wider as the entire lumen fills with dye.

Venous phase
SECTION 10

Complete filling of the veins occurs over the next 10 s with


FIGURE 128.7. Normal macular anatomy. The central 500 mm of the
fovea is devoid of blood vessels (foveal avascular zone or FAZ). This
maximum vessel fluorescence occurring ~30 s after injection.
finding, along with the presence of xanthophyll pigment and taller, The perifoveal capillary network is best visualized in the peak
more pigmented retinal pigment epithelial cells, contributes to the venous phase of the angiogram.
relative hypofluorescence of the macula.

a b c

d e f

FIGURE 128.8. Normal fluorescein angiogram. (a) Choroidal flush. Fluorescein dye appears first in the choroid, 1–2 s before the dye reaches the
retinal arterial circulation. When present, cilioretinal arteries fill along with the choroidal flush since both are supplied by the short posterior ciliary
arteries. (b) Arterial phase. The arterial phase occurs when the fluorescein dye enters the retinal arteries. The normal ‘arm-to-retina’ circulation
time is ~12 s. (c) Arteriovenous phase. The arteriovenous phase of the angiogram comprises the time when the retinal arteries, capillaries, and
veins contain fluorescein. In the early arteriovenous phase, thin columns of fluorescein are visualized along the walls of the larger veins (laminar
flow). (d) Venous phase. As the fluorescein dye begins to exit from the retinal arteries and capillaries, the concentration of fluorescein within the
veins increases, resulting in a decrease in fluorescence of the arteries and an increase of fluorescence of the veins. (e) Mid phase. The
recirculation phase occurs ~2–4 min after injection. The veins and arteries remain roughly equal in brightness. The intensity of fluorescence
diminishes slowly during this phase as fluorescein is removed from the bloodstream by the kidneys. (f) Late phase. The late phase of the
angiogram demonstrates the gradual elimination of dye from the retinal and choroidal vasculature. Staining of the optic disk is a normal finding.
1696 Any other areas of late hyperfluorescence suggest the presence of an abnormality, usually the result of fluorescein leakage.
Principles of Fluorescein Angiography

Mid Phase Pseudofluorescence is the reduction or absence of normal


Also known as the recirculation phase, this occurs ~2–4 min fluorescence. Hypofluorescence is caused by either blockage of
after injection. The veins and arteries remain roughly equal in the normal fluorescence pattern or abnormalities in choroidal
brightness. The intensity of fluorescence diminishes slowly or retinal vascular perfusion.
during this phase as much of the fluorescein is removed from
the bloodstream on the first pass through the kidneys (see Fig. Blocked fluorescence
128.8e). Blocked fluorescence is most commonly caused by blood but
can result from the deposition of abnormal materials such as
Late Phase lipid exudate, lipofuscin, xanthophyll pigment, or melanin
The late or elimination phase demonstrates the gradual pigment. Fluorescein angiography is very helpful in determining
elimination of dye from the retinal and choroidal vasculature the anatomic location of the blocking material, which in turn,
(see Fig. 128.8f). Photographs are typically captured 7–15 min is important in identifying the etiology of the abnormality. For
after injection. Late staining of the optic disk is a normal example, preretinal hemorrhage from proliferative diabetic
finding. Any other areas of late hyperfluorescence suggest the retinopathy blocks visibility of both the retinal and choroidal
presence of an abnormality. vasculature while subretinal blood from exudative age-related
macular degeneration obscures only the choroidal circulation
(Fig. 128.9).
THE ABNORMAL ANGIOGRAM

CHAPTER 128
In evaluating diseases of the macula, fluorescein angiography is Abnormal vascular perfusion
helpful in detecting abnormalities in blood flow, vascular Abnormal vascular perfusion results in hypofluorescence of the
permeability, the retinal and choroidal vascular patterns, the retinal and/or choroidal circulation depending on the location of
RPE, and a variety of other changes.68 Interpretation of the the abnormality. Common causes of retinal hypoperfusion include
abnormal angiogram relies on the identification of areas that retinal arterial and venous occlusions and ischemic disease due
exhibit hypofluorescence or hyperfluorescence. These are to diabetes and other causes. Choroidal hypoperfusion may be
descriptive terms that refer to the time specific, relative produced by ophthalmic artery occlusion, giant cell arteritis,
brightness of fluorescence in comparison with a normal study and hypertensive choroidopathy. It is important to understand
(Table 128.4). the relationship between hypofluorescence due to filling defects
and the specific phase of the angiogram. For example, in many
Hypofluorescence vascular occlusions the hypofluorescence may be a temporary
finding until delayed filling of the affected vessel occurs in the
Key Features later phases of the study (Fig. 128.10).
Hypofluorescence is usually caused by the blockage of normal
fluorescence or abnormal vascular perfusion. Hyperfluorescence

Key Features
TABLE 128.4. Abnormal Angiographic Findings Hyperfluorescence usually results from atrophy of the pigment
epithelium (transmission or ‘window’ defect) or leakage of
Hypofluorescence fluorescein dye.
Filling defect
Blocking defect
Hyperfluorescence is an increase in fluorescence resulting from
Hyperfluoresence the increased transmission of normal fluorescence or an ab-
Autofluorescence normal presence of fluorescein at a given time in the angiogram.
Autofluorescence and pseudofluorescence are terms to
Pseudofluorescence
describe the appearance of apparent hyperfluorescence in the
Transmission or ‘window’ defect absence of fluorescein.
Leakage
Autofluorescence
Pooling Autofluorescence refers to recordable hyperfluorescence that is
Staining believed to occur naturally in certain pathologic entities such as
optic nerve drusen and astrocytic hamartomas. Some, but not

FIGURE 128.9. Hypofluorescence: blockage.


(a) This fluorescein angiogram of an eye with a
central retinal vein occlusion demonstrates
widespread hypofluorescence resulting from
intraretinal hemorrhages. The majority of
hemorrhages are located in the nerve fiber
layer, which accounts for the ‘flame-shaped’
pattern of hypofluorescence along the major
vascular arcades. (b) Hypofluorescence related
to subretinal hemorrhage is demonstrated in
this patient with a ruptured retinal arterial
macroaneurysm. The ability to visualize the
retinal circulation overlying the blocking defect
a b confirms the subretinal location of the
hemorrhage. 1697
RETINA AND VITREOUS

FIGURE 128.10. Hypofluorescence:


nonperfusion. (a) In this patient with advanced
diabetic retinopathy, there is marked
hypofluorescence throughout the retina as a
result of widespread ischemia. Note that there
are scattered patches of more prominent
hypofluorescence from intraretinal hemorrhages.
(b) This patient with diabetic macular edema
has significant macular ischemia with capillary
dropout and enlargement and irregularity of the
foveal avascular zone. Contrast this with the
normal perifoveal capillary network shown in
Figure 128.7.
a b

FIGURE 128.11. Hyperfluorescence:


autofluorescence. (a) Red-free photograph of a
patient with disk drusen. (b) A photograph
SECTION 10

taken with fluorescein exciter and barrier filters


demonstrates autofluorescence of the disk
drusen in the absence of fluorescein.

a b

all disk drusen appear to fluoresce under blue light (Fig.


128.11). Some controversy exists over whether this is actual
fluorescence or reflectance.69 These structures are highly
reflective in the same spectral range of fluorescence and could
actually be exhibiting pseudofluorescence.

Pseudofluorescence
Pseudofluorescence occurs as a result of crossover in the
spectral transmission curves of the exciter and barrier filters. If
too much crossover is present, reflectance from bright fundus
structures will not be fully blocked by the barrier filter.
Crossover can be the result of mismatched or aging filters.
Modern interference filters rarely exhibit significant crossover
unless they have deteriorated. Control photographs are
routinely taken before injection of fluorescein to detect the
possible presence of pseudofluorescence.

Transmission defect
Depending on the density of retinal pigmentation, background
fluorescence from the choroid can be visible as hyper-
fluorescence in the angiogram. A ‘window defect’ is an area of FIGURE 128.12. Hyperfluorescence: transmission defect. A
hyperfluorescence that occurs when there is an absence or transmission or ‘window’ defect is seen in this patient with age-related
reduction of pigmentation due to damage of the RPE. The loss macular degeneration with geographic atrophy. Atrophy of the RPE
of pigment allows visualization of the fluorescence created by allows visualization of the underlying choroidal fluorescence. The
the underlying choriocapillaris (Fig. 128.12). Window defects hyperfluorescence usually appears early in the angiogram and is not
remain uniform in size throughout the angiogram. Their associated with leakage in the late phase.
brightness rises and falls with the choroidal fluorescence. It is
important to differentiate hyperfluorescence due to trans-
mission defects from leakage. or the breakdown of the tight junctions between retinal pigment
epithelial cells (the inner and outer blood–retinal barriers,
Leakage respectively). Examples include macular edema from diabetic
Leakage refers to hyperfluorescence in the angiogram due to retinopathy (Fig. 128.13), cystoid macular edema, and central
extravasation of fluorescein dye. Leakage can result from serous chorioretinopathy. In addition to abnormalities of the
1698 disruption of the retinal vascular endothelial cell tight junctions retinal vascular system or pigment epithelium, leakage is
Principles of Fluorescein Angiography

FIGURE 128.13. Hyperfluorescence: leakage.


The phenomenon of hyperfluorescence related
to fluorescein leakage is demonstrated in this
patient with diabetic retinopathy. (a) The
arteriovenous phase image reveals multiple
hyperfluorescent spots corresponding to
microaneurysms. (b) In the late phase
angiogram, there is diffuse hyperfluorescence
related to leakage from the microaneurysms
and irregular capillaries.

a b

FIGURE 128.14. Hyperfluorescence: leakage.


This pair of images demonstrates fluorescein

CHAPTER 128
leakage from exudative age-related macular
degeneration. (a) During the arteriovenous
phase, the abnormal choroidal blood vessels
become visible (they are surrounded by a zone
of hypofluorescence caused by the blocking
effect of hemorrhage). (b) In the late phase
angiogram, there is significant leakage of
fluorescein throughout the macula.

a b

observed in a variety of conditions associated with the


development of new blood vessels. For example, fluorescein
leakage is seen in eyes with choroidal neovascularization related
to age-related macular degeneration. In these patients,
fluorescein angiography is needed to identify the location and
features of the choroidal neovascular membrane which, in turn,
influences the course of treatment (Fig. 128.14). In eyes with
proliferative diabetic retinopathy, optic disk or retinal
neovascularization is characterized by intense fluorescein
leakage (Fig. 128.15). Leakage can lead to late staining or
pooling of dye.

Staining
Staining refers to late hyperfluorescence resulting from the
accumulation of fluorescein dye into certain tissues. Drusen and
chorioretinal scars commonly exhibit staining. Normal staining
can occur in the optic nerve and sclera as a result of normal
choroidal leakage. Scleral staining is usually only visible when
there is a reduction or absence of the pigment epithelium (window
defect) and the sclera can be seen clinically (Fig. 128.16).
FIGURE 128.15. Hyperfluorescence: leakage. Retinal
Pooling neovascularization is characterized by profuse leakage on fluorescein
Pooling is the accumulation of dye within a distinct anatomic angiography. The areas of retinal neovascularization usually are
space. Pooling can occur in serous detachments of the sensory located at the junction of the perfused and nonperfused retina.
retina or the RPE due to a breakdown of the blood–retinal
barrier. Central serous chorioretinopathy is a condition that
often demonstrates the pooling of fluorescein (Fig. 128.17). insights and value to the ‘Gold Standard’ of retinal imaging.
Color stereo fundus photography is a natural complement to
COMPLEMENTARY IMAGING TECHNIQUES fluorescein angiography and has traditionally been included in
standard angiography protocols along with monochromatic
Fluorescein angiography continues to be a vital tool in the detec- green light ‘red free’ photographs. Monochromatic fundus photo-
tion of retinal vascular disorders. A number of additional graphy with other colors of light has been used for decades, but
diagnostic imaging procedures serve as an adjunct to angiography. is probably underutilized given its inherent value. More
Some have a long history of complementary use with fluorescein recently, ICG angiography, OCT, and FAF have expanded the
angiography, while newer techniques can add new diagnostic available arsenal of diagnostic imaging procedures. 1699
RETINA AND VITREOUS

age-related macular degeneration.73 The goal in age-related


ICG ANGIOGRAPHY macular degeneration is to identify localized areas of choroidal
Digital technology facilitates the use of another fluorescent dye, neovascularization or choroidal feeder vessels that can be
ICG, for retinal and choroidal angiography. With peak absorption treated with a laser to prevent damage to the retina.74–76
(805 nm) and emission (835 nm) in the near-infrared range,
ICG provides greater transmission through the RPE and
hemorrhage than the visible wavelengths used in fluorescein MONOCHROMATIC FUNDUS PHOTOGRAPHY
angiography. ICG also binds more completely with blood Monochromatic fundus photography is the practice of imaging
albumins, so it normally remains within the fenestrated walls the ocular fundus with the use of colored or monochromatic
of the choriocapillaris, unlike fluorescein, which leaks freely illumination. In 1925, Vogt described the use of green light to
from these vessels. enhance the visual contrast of anatomical details of the fundus.77
ICG choroidal angiography was first performed in humans The technique is still commonly used today in combination
in 1972, but results were limited by insufficient sensitivity of with fundus photography. Monochromatic fundus photography
available infrared films.70 In the early 1990s, two groups is based on increased scattering of light at shorter wavelengths
reported improved results with commercially available digital and the use of contrast filters to alter subject tones in black
angiography systems.71,72 The infrared sensitivity of available and white photographs. By limiting the spectral range of the
CCD cameras was combined with new lens coatings applied to illuminating source, the visibility of various fundus structures
fundus camera optics to improve infrared transmission for ICG can be enhanced (Fig. 128.18).78–81 This technique is most
SECTION 10

angiography. In the early to mid 1990s, enthusiasm for this effective when combined with high-resolution black-and-white
procedure exceeded its practical applications. Use of this film or digital sensors.
technique waned by the late 1990s as clinical research had Blue light increases visibility of the anterior retinal layers,
defined a vital, but limited role for ICG angiography as an which normally are almost transparent in white light. Blue light
adjunct to fluorescein angiography. is absorbed by retinal pigmentation and blood vessels, providing
Today, ICG angiography is used in combination with a dark background against which the specular reflections and
fluorescein angiography in a limited number of diagnoses where scattering in the anterior layers of the fundus is enhanced.
choroidal circulation is effected, particularly in patients with Scattering in the ocular media can limit the effectiveness of
these wavelengths. The retinal nerve fiber layer, the internal
limiting membrane, retinal folds, cysts and epiretinal membranes
are examples of semitransparent scattering structures that are
enhanced with short wavelength illumination. Because of excessive
scattering at very short wavelengths, fluorescein exciter filters at
blue–green wavelengths of 490 nm are often used.
Green light is also absorbed by blood, but is reflected more
than blue light by retinal pigmentation. There is less scatter
than with shorter wavelengths, so media opacities are not quite
as troublesome. Green light provides excellent contrast and the
best overall view of the fundus. It enhances the visibility of the
retinal vasculature, and common findings such as hemorrhages,
drusen and exudates. For this reason, green filter ‘red-free’
photos are routinely taken as baseline images before fluorescein
angiography.
Retinal pigmentation appears more transparent in red light,
revealing the choroidal pattern. Overall fundus contrast is
greatly reduced with red illumination. Retinal vessels appear
lighter and become less obvious at longer wavelengths. The
optic nerve appears lighter and almost featureless. Red light is
useful for imaging pigmentary disturbances, choroidal ruptures,
choroidal nevi, and choroidal melanomas.
FIGURE 128.16. Hyperfluorescence: staining. This late phase
Traditionally, monochromatic photography has required use
angiogram demonstrates staining of drusen. Hyperfluorescence is of high-resolution black-and-white films and customized film
observed as fluorescein accumulates within the drusen material. processing techniques to maximize diagnostic information.

FIGURE 128.17. Hyperfluorescence: pooling.


This pair of angiographic images demonstrates
the phenomenon of pooling. (a) Central serous
chorioretinopathy is characterized by a
hyperfluorescent spot that increases in size and
intensity throughout the study. (b) Ten minutes
after injection, this patient has the characteristic
‘smokestack’ appearance as fluorescein
accumulates or ‘pools’ within the localized
neurosensory retinal detachment.

a b
1700
Principles of Fluorescein Angiography

FIGURE 128.18. Monochromatic Imaging.


(a) Fluorescein angiogram of a choroidal lesion.
(b) Mononchromatic red light (peak
transmission at 615 nm) imaging is particularly
useful for documenting lesions deep within the
retina and choroid. Longer wavelengths partially
penetrate the retina pigment epithelium and
make it appear more transparent, revealing the
borders of the nevus.

a b

FIGURE 128.19. Optical coherence


tomography. OCT imaging can be used as an

CHAPTER 128
adjunct to, or a replacement for, fluorescein
angiography. OCT imaging has become the
diagnostic procedure of choice to detect
cystoid macular edema, a condition that was
traditionally confirmed with fluorescein
angiography.

a b

These film and developer combinations could be difficult to Fluorescein angiography was originally developed to look
control, which may explain why monochromatic photography specifically at retinal blood flow. An unexpected benefit was the
never gained universal acceptance. Digital imaging facilitates ability to detect structural changes in certain retinal conditions
immediate control of exposure settings and easy contrast by observation of fluorescein staining patterns in abnormal
enhancement, making the monochromatic technique more retinal tissues or anatomic spaces caused by structural change.
practical for widespread use. OCT has demonstrated great application in detecting many of
these structural abnormalities and fluorescein angiography is
no longer needed for some of these diagnoses (Fig. 128.19). The
OPTICAL COHERENCE TOMOGRAPHY true strength of angiography lies in its original purpose, to look
OCT is useful in the diagnosis of several retinal disorders that specifically at flow dynamics in the retinal and choroidal vascu-
have traditionally been imaged with fundus photography or lature. Used in tandem, OCT and fluorescein angiography provide
fluorescein angiography. OCT imaging provides direct cross- a wealth of diagnostic information to aid in the management of
sectional images of the macula, retinal nerve fiber layer and retinal disease (Fig. 128.20).
optic nerve for objective measurement and clinical evaluation in
the detection of retinal diseases and glaucoma. It is particularly
useful in the detection of changes in normal retinal architecture FUNDUS AUTOFLUORESCENCE
such as macular holes, epiretinal membranes, vitreomacular FAF is a recently developed noninvasive imaging technique for
traction, cystoid macular edema, subretinal fluid, and retinal documenting the presence of lipofuscin in the RPE. Lipofuscin
pigment epithelial detachments.82,83 Its greatest value however, is a fluorescent pigment that accumulates in the RPE as a result
may lie in the ability to quantify and monitor change in retinal of incomplete degradation of photoreceptor outer segments as
thickness due to macular edema from diabetic retinopathy or the eye ages. When excited with short wavelength illumination,
other causes. lipofuscin granules autofluoresce, exhibiting a broad emission
Although the technology has been available since 1991, OCT spectrum from 500 to 750 nm with peak emission at ~630 nm.84
was mostly used in academic settings or for research appli- The original technique used a confocal SLO with the excita-
cations until 2002, when the evolution of the instrumentation, tion wavelength set at 488 nm and a wide band-pass filter with
in the form of increased resolution and ease of operation, short wavelength cutoff at 521 nm.85 Several frames are captured
matured to the point where OCT imaging became both practical with the SLO, then aligned and averaged to reduce noise. Because
and affordable. Since its introduction, OCT has rapidly gained several frames are required, image quality may be affected by
widespread acceptance and for certain retinal conditions, is now eye movement during capture. More recently, digital fundus-
the diagnostic procedure of choice. Compared to angiography, camera based systems have been developed which use high-
OCT is less expensive, noninvasive, faster, well tolerated and sensitivity monochrome sensors with an excitation filter at
easy to perform. The procedure can be conducted in ~10 min, 580 nm and a barrier filter at 695 nm to avoid confounding
usually without dilation or discomfort to the patient. Enthusiasm autofluorescence from the crystalline lens.86 Both systems
for OCT continues to grow as new instruments are being devel- require significant amounts of light and increased gain settings
oped by a number of manufacturers. Advances in technology to achieve adequate exposure, and are subject to image noise.
that use spectral or Fourier domain OCT techniques, promise Despite the disparity in excitation wavelength and barrier filters
improvement in resolution and acquisition speed, as well as between the SLO and fundus camera systems, these two tech-
3-D viewing of retinal structures. niques obtain results that are quite similar in appearance. 1701
RETINA AND VITREOUS

FIGURE 128.20. Optical coherence


tomography. The combination of fluorescein
angiography and OCT can provide the clinician
with greater diagnostic information as
demonstrated in this case of idiopathic central
serous chorioretinopathy. Angiography identifies
the area of active leakage, while OCT illustrates
a compartmentalized serous detachment.

a b

FIGURE 128.21. Fundus autofluorescence.


(a) Fluorescein angiography image of a patient
with geographic atrophy in age-related macular
degeneration. (b) Fundus autofluorescent
SECTION 10

imaging (excitation at 580 nm and barrier at


695 nm) noninvasively identifies RPE atrophy as
areas of hypofluorescence due to an absence
or reduction of lipofuscin accumulation.

a b

Autofluorescence imaging has the potential to provide useful may precede development or progression of geographic atrophy
information in conditions where the health of the RPE plays a in AMD.91
key role. Hyperfluorescence is a sign of increased lipofuscin
accumulation, which may indicate degenerative changes or SUMMARY
oxidative injury. Areas of hypofluorescence indicate missing or
dead RPE cells. Geographic atrophy that appears as a window Fluorescein angiography has revolutionized the understanding,
defect in fluorescein angiography will appear dark in autofluo- diagnosis and treatment of retinal vascular disorders. For over
rescent imaging (Fig. 128.21). A number of investigators have 40 years, ophthalmologists have used fluorescein angiography
been exploring potential applications of this imaging technique as a guide for laser treatment, benefiting many thousands of
in a variety of retinal diseases including: retinitis pigmentosa,87 patients. This important diagnostic tool continues to evolve
central serous chorioretinopathy,88 macular dystrophies,89 pseu- with new advances in digital imaging technique. As new treat-
doxanthoma elasticum,90 and age-related macular degeneration.86 ment modalities are developed, fluorescein angiography will
The role of lipofuscin in the pathogenesis of macular degen- continue to play an important role in the management of
eration is currently unknown, but increased autofluorescence retinal conditions.

REFERENCES
1. Wolfe DR: Fluorescein angiography basic viability of arterialized flaps. Plast Reconstr of fluorescein as an indicator of local blood
science and engineering. Ophthalmology Surg 1977; 60:710–719. flow: fluorescein test in experimental
1986; 93:1617–1620. 7. Singer R, Lewis CM, Franklin JD, et al: frostbite. J Clin Invest 1947; 26:259–268.
2. Morris PF: Fluorescein sodium and Fluorescein test for prediction of flap 13. Gatti JE, LaRossa D, Silverman DG, et al:
indocyanine green: uses and side effects. viability during breast reconstructions. Plast Evaluation of the burn wound with
In: Saine PJ, Tyler ME, eds. Ophthalmic Reconstr Surg 1978; 61:371–375. perfusion fluorometry. J Trauma 1983;
photography: retinal photography, 8. Smith JA Jr, Middleton RG: The use of 23:202–206.
angiography and electronic imaging. 2nd fluorescein in radical inguinal 14. Minor DR, Allen RE, Alberts D, et al: A
edn. Boston: Butterworth-Heinemann; lymphadenectomy. J Urol 1979; clinical and pharmacokinetic study of
2002:137–165. 122:754–756. isolated limb perfusion with heat and
3. Jacobs J: Fluorescein sodium – what is it? 9. Sloan GM, Sasaki GH: Noninvasive melphalan for melanoma. Cancer 1985;
J Ophthal Photography 1992; 14:62. monitoring of tissue viability. Clin Plast 55:2638–2644.
4. Dole RB: Use of fluorescein in the study of Surg 1985; 12:185–195. 15. Martonyi CL, Bahn CF, Meyer RF: Clinical
underground waters. USGS Water Supply 10. Stolar CJ, Randolph JG: Evaluation of slit lamp biomicroscopy and photo slit lamp
Paper 1906; 160:73–85. ischemic bowel viability with a fluorescent biomicrography. Ann Arbor, MI: Time One
5. Myers B: Prediction of skin sloughs at the technique. J Pediatr Surg 1978; 13:221–225. Ink; 1985:64–67.
time of operation with the use of 11. Marfuggi R, Greenspan M: Intraoperative 16. Saine PJ: Landmarks in the development of
fluorescein dye. Surgery 1962; prediction of intestinal viability. Surg Gynae fluorescein angiography. J Ophthal
51:158–162. Obstet 1981; 152:33–35. Photography 1993; 15:17–23.
6. McCaw JB, Myers B, Shanklin KD: The 12. Crimson J, Fuhrman F: Studies on 17. Chao P, Flocks M: The retinal circulation
1702 value of fluorescein in predicting the gangrene following cold injury. IV. The use time. Am J Ophthalmol 1958; 46:8–10.
Principles of Fluorescein Angiography

18. Flocks M, Miller J, Chao P: Retinal reactions revisited: the Lions Eye experience. 61. Benetz BA, Bennett TJ, Tomer TL: What
circulation time with the aid of fundus Clin Exp Ophthalmol 2006; 34:33–38. does today’s ophthalmic photographer do?
cinematography. Am J Ophthalmol 1959; 39. Eutick M: Sodium fluorescein – colourful J Ophthal Photography 2005; 27:85–87.
48:3–6. past, bright future. J Ophthal Photography 62. Woon WH, Fitzke FW, Chester GH, et al:
19. MacLean AL, Maumenee AE: Hemangioma 2006; 28:66–70. The scanning laser ophthalmoscope: basic
of the choroid. Am J Ophthalmol 1960; 40. Ellis PP, Schoenberger M, Rendi MA: principles and applications. J Ophthal
50:2–11. Antihistamines as prophylaxis against side Photography 1990; 12:17–23.
20. Novotny HR, Alvis DL: A method of reactions to intravenous fluorescein. Trans 63. Clark TM: Scanning laser
photographing fluorescence in circulating Am Ophthalmol Soc 1980; 78:190–205. ophthalmoscopes. In: Saine PJ, Tyler ME,
blood of the human retina. Circulation 41. Buchanan RT, Levine NS: Blood pressure eds. Ophthalmic photography: retinal
1961; 24:82–86. drop as a result of fluorescein injection. photography, angiography and electronic
21. David NJ, Justice J: The early days of Plast Reconstr Surg 1982; 70:363–368. imaging. 2nd edn. Boston: Butterworth-
fluorescein angiography. J Ophthal 42. Merin LM, Lam BL: Case report: fluorescein Heinemann; 2002:306–321.
Photography 1994; 16:83–86. angiogram during vasovagal syncope. 64. Allen L: Ocular fundus photography:
22. Quillen DA, Gardner TW, Blankenship GW: J Ophthal Photography 1994; 16:94–95. suggestions for achieving consistently
The Diabetic Retinopathy Study. In: Kertes 43. Gombos GM, Lieberman RM: Seizures good pictures and instructions for
PJ, Conway MD, eds. Clinical trials in associated with fluorescein angiography. stereoscopic photography. Am J
ophthalmology: a summary and practice Ann Ophthalmol 1989; 21:89–90. Ophthalmol 1964; 57:13–28.
guide. Baltimore: Williams & Wilkins; 44. Kelly SP, Mcdermott NJ, Saunders DC, 65. Bennett TJ: Stereo anaglyph preparation
1998:1–13. et al: Convulsion following fluorescein for PowerPoint. J Ophthal Photography

CHAPTER 128
23. Physicians’ desk reference for ophthalmic angiography. Br J Ophthalmol 1989; 2005; 27:38–45.
medicines. Montvale: Thompson PDR; 73:655–656. 66. Ellis JH, Weber P: Legal issues arise when
2006:207. 45. Hess JB, Pacirariu RI: Acute pulmonary unlicensed personnel administer IV
24. Palestine AG: Does intravenous fluorescein edema following intravenous fluorescein fluorescein. Argus 1995; Nov–Dec:28–31.
interfere with clinical laboratory testing? angiography. Am J Ophthalmol 1976; 67. Ophthalmic photographers’ society
J Ophthal Photography 1991; 13:27–28. 82:567–570. standards of practice. J Ophthal
25. Bloom JN, Herman DC, Elin RJ, et al: 46. Deglin SM, Deglin EA, Chung EK: Acute Photography 1999; 21:26.
Intravenous fluorescein interference with myocardial infarction following fluorescein 68. Gass JDM: Stereoscopic atlas of macular
clinical laboratory tests. Am J Ophthalmol angiography. Heart Lung 1977; 6:505–509. diseases: diagnosis and treatment. 4th edn.
1989; 108:375–379. 47. Acasco FJ, Tiestos MT, Navales J, et al: St Louis: Mosby; 1997.
26. Mattern J, Mayer PR: Excretion of Fatal acute myocardial infarction after 69. Barry C, Singh J, Constable IJ: Are optic
fluorescein into breast milk [letter]. Am J intravenous fluorescein angiography. Retina disc drusen exhibiting Autofluorescence,
Ophthalmol 1990; 109:598. 1993; 13:238–239. pseudofluorescence or reflectance?
27. AK-FLUOR package insert, rev. 08/05. 48. Schatz H: Sloughing of skin following J Ophthal Photography 2000; 22:32–35.
Akorn, Inc Buffalo Grove, IL. Available: fluorescein extravasation. Ann Ophthalmol 70. Flower RW, Hochheimer BF: Clinical
http://www.akorn.com/documents/catalog/ 1978; 10:625. infrared absorption angiography of the
package_inserts/17478–254–10.pdf. 49. Elman MJ, Fine SL, Sorenson J, et al: Skin choroids. Am J Ophthalmol 1972;
28. Halperin LS, Olk RJ, Soubrane G, et al: necrosis following fluorescein 73:458–459.
Safety of fluorescein angiography during extravasation. A survey of the Macula 71. Guyer DR, Puliafito CP, Mones JM, et al:
pregnancy. Am J Ophthalmol 1990; Society. Retina 1987; 7:89–93. Digital indocyanine-green angiography in
109:563–566. 50. Lipson BK, Yannuzzi LA: Complications of chorioretinal disorders. Ophthalmology
29. Greenberg F, Lewis RA: Safety of fluorescein fluorescein injections. Int Ophthalmol Clin 1992; 99:287–290.
angiography during pregnancy [letter]. Am J 1989; 29:200–205. 72. Yannuzzi LA, Slakter JS, Sorenson JA, et al:
Ophthalmol 1991; 110:323–325. 51. Kelley JS, Kincaid M: Retinal fluorography Digital indocyanine green videoangiography
30. Berkow JW, Flower RW, Orth DH, Kelley using oral fluorescein. Arch Ophthalmol and choroidal neovascularization. Retina
JS: Fluorescein and indocyanine green 1979; 97:2331–2332. 1992; 12:191–223.
angiography. 2nd edn. San Francisco: 52. Hara T, Inami M, Hara T: Efficacy and 73. Stanga PE, Lim JI, Hamilton P: Indocyanine
American Academy of Ophthalmology; safety of fluorescein angiography with green angiography in chorioretinal
1997:7. orally administered sodium fluorescein. Am diseases: indications and interpretation: an
31. Chazan BI, Balodimos MC, Koncz L: J Ophthalmol 1998; 126:560–564. evidence-based update. Ophthalmology
Untoward effects of fluorescein retinal 53. Kinsella FP, Mooney DJ: Anaphylaxis 2003; 110:15–21.
angiography in diabetic patients. Ann following oral fluorescein angiography. Am 74. Shiraga F, Ojima Y, Matsuo T, et al: Feeder
Ophthalmol 1971; 3:42. J Ophthalmol 1988; 106:745–746. vessel photocoagulation of subfoveal
32. Pacirariu RI: Low incidence of side effects 54. Gomez-Ulla F, Gutierrez C, Seonae I: choroidal neovascularization secondary to
following intravenous fluorescein Severe anaphylactic reaction to orally age-related macular degeneration.
angiography. Ann Ophthalmol 1982; administered fluorescein. Am J Ophthalmol Ophthalmology 1998; 105:662–691.
14:32–36. 1991; 112:94. 75. Flower RW: Experimental studies of
33. Butner RW, McPherson AR: Adverse 55. Saine PJ, Tyler ME, eds: Ophthalmic indocyanine green dye-enhanced
reactions in intravenous fluorescein photography: retinal photography, photocoagulation of choroidal
angiography. Ann Ophthalmol 1983; angiography and electronic imaging. 2nd neovascularization feeder vessels. Am J
15:1084–1086. edn. Boston: Butterworth-Heinemann; Ophthalmol 2000; 129:501–512.
34. Marcus DF, Bovino JA, Williams D: Adverse 2002:247–248. 76. Flower RW: Optimizing treatment of
reactions during intravenous fluorescein 56. Oosterwijk H: DICOM versus HL7 for choroidal neovascularization feeder vessels
angiography. Arch Ophthalmol 1984; modality interfacing. J Digit Imaging 1998; associated with age-related macular
102:825. 11(3 Suppl 1):39–41. degeneration. Am J Ophthalmol 2002;
35. Yannuzzi LA, Rohrer KT, Tindel LJ, et al: 57. Kabachinski J: DICOM: key concepts-part 134:228–239.
Fluorescein angiography complication I. Biomed Instrumen Technol 2005; 77. Vogt A: Die Ophthalmoskopie im rotfreiem
survey. Ophthalmology 1986; 93:611–617. 39:214–216. licht. GraefSaemisch Handbuch der
36. Karhunen U, Raitta C, Kala R: Adverse 58. Kabachinski J: DICOM: key concepts-part gesamten Augenheilkunde. Berlin: Springer
reactions to fluorescein angiography. Acta II. Biomed Instrumen Technol 2005; Verlag; 1925.
Ophthalmol 1986; 64:282–286. 39:292–294. 78. Delori FC, Gragoudas ES: Examination of
37. Kwiterovitch KA, Maguire MG, Murphy RP, 59. Bidgood WD Jr, Horii SC: Introduction to the ocular fundus with monochromatic
et al: Frequency of adverse systemic the ACR-NEMA DICOM standard. light. Ann Ophthalmol 1976; 8:703–709.
reactions occurring after fluorescein Radiographics 1992; 12:345–355. 79. Behrendt T, Slipakoff E: Spectral
angiography – results of a prospective 60. Bennett TJ, MacGregor G, Scheuneman reflectance photography. Int Ophthalmol
study. Ophthalmology 1991; 98:1139–1142. JD: Digital Imaging and the certified retinal Clin 1976; 16:95–100.
38. Kwan AS, Barry CJ, McAllister IL, et al: Angiographer Program. J Ophthal 80. Delori FC, Gragoudas ES, Francisco R,
Fluorescein angiography and adverse drug Photography 2004; 26:28–36. et al: Monochromatic ophthalmoscopy and 1703
RETINA AND VITREOUS

fundus photography. The normal fundus. exhibits retinal pigment epithelium chorioretinopathy. Ophthalmology 2005;
Arch Ophthalmol 1977; 95:861–868. lipofuscin characteristics. Invest 112:825–833.
81. Ducrey NM, Delori FC, Gragoudas ES: Ophthalmol Vis Sci 1995; 36:718–729. 89. Von Ruckmann A, Fitzke FW, Bird AC: In
Monochromatic ophthalmoscopy and 85. von Ruckman A, Fitzke FW, Bird AC: vivo fundus autofluorescence in macular
fundus photography. II. The pathological Distribution of fundus autofluorescence dystrophies. Arch Ophthalmol 1997;
fundus. Arch Ophthalmol 1979; with a scanning laser ophthalmoscope. 115:609–615.
97:288–293. Br J Ophthalmol 1995; 79:407–412. 90. Sawa M, Ober MD, Freund KB, et al:
82. Puliafito CA, Hee MR, Lin CP, et al: Imaging 86. Spaide RF: Fundus autofluorescence and Fundus autofluorescence in patients with
of macular diseases with optical coherence age-related macular degeneration. pseudoxanthoma elasticum.
tomography. Ophthalmology 1995; Ophthalmology 2003; 110:392–399. Ophthalmology 2006; 113:814–820.
102:217–229. 87. Robson AG, Egan C, Holder GE, et al: 91. Hopkins J, Walsh A, Chakravarthy U:
83. Costa RA, Skaf M, Melo LA Jr, et al: Retinal Comparing rod and cone function with Fundus autofluorescence in age-related
assessment using optical coherence fundus autofluorescence images in retinitis macular degeneration: an epiphenomenon?
tomography. Prog Retin Eye Res 2006; pigmentosa. Adv Exp Med Biol 2003; Invest Ophthalmol Vis Sci 2006;
25:325–353. 533:41–47. 47:2269–2271.
84. Delori FC, Dorey CK, Staurenghi G, et al: 88. Spaide RF, Klancnik JM Jr: Fundus
In vivo fluorescence of the ocular fundus autofluorescence and central serous
SECTION 10

1704
CHAPTER

129 Indocyanine Green Videoangiography


Jordi Monés, David R. Guyer, Sara Krupsky, Ephraim Friedman,
Evangelos S. Gragoudas, and Antonio P. Ciardella

Fluorescein angioscopy1 and angiography2 were important tech- this technique, direct fluorescence of the vessels occurred, the
nological advances in the study of retinal disorders. Although resolution improved, and the arterial and capillary dye phases
choroidal circulation patterns have been described with fluor- were evident. In 1974, Flower developed a multispectral fundus
escein angiography, the limitations of this technique for studying camera.12 Simultaneous ICG fluorescence and absorption angio-
the choroid are well known.3–6 The excitation and fluorescence graphy with fluorescein angiography was performed. This
of blue-green wavelengths (peak absorption 465 nm peak fluor- combination allowed comparison of the various types of angio-
escence 525 nm) are absorbed and scattered by the pigment layers grams. Clinical reports of ICG angiographic imaging of choroidal
of the fundus, including the macular xanthophyll. Thus, the neovascularization (CNV), choroidal tumors, choroideremia,
choroidal layers cannot be well visualized.7–10 In addition, sodium choroidal hemangiomas, and the presumed ocular histoplas-
fluorescein, which is 60–80% bound to plasma albumin,11 rapidly mosis syndrome were published.24–31 Further technological
leaks from the fenestrated choriocapillaris4 and produces a dif- improvements followed.32–34 Visualization of the choriocapillaris
fuse background fluorescence, which further obscures the details filling pattern with ICG and study of the temporal differences
of the choroidal vessels.12 Finally, the intricate branching of the of ICG and fluorescein choroidal filling were performed in
choroidal vascular system13 is difficult to study with fluorescein monkeys.33 Hyvärinen and Flower presented a case of CNV in
angiography.12 which the feeding choroidal artery was identified and
Because the choroid is the major blood supply of the eye14 photocoagulated.35
and the outer retinal layers, a better method of studying this In 1985, Bischoff and Flower reported their 10 years of expe-
important tissue was needed. Near-infrared absorption angio- rience with ICG choroidal angiography.36 This series included
graphy, using indocyanine green (ICG) as an absorbing dye, was 180 angiograms of normal volunteers and 500 angiograms of
first studied in the canine brain by Kogure and Choromokos in patients with various fundus diseases. Hayashi and colleagues in
1969.15 This study led to the development of ICG choroidal 1986 performed ICG angiography in patients with central serous
angiography. chorioretinopathy using an infrared-sensitive video camera.37
Videoangiography has also been used by other investigators to
HISTORY OF ICG ANGIOGRAPHY study CNV38–40 and choroidal blood flow.41,42 A digital computer
system has also been used to study choroidal blood flow.43,44
In 1969, a qualitative method of studying choroidal blood flow Preliminary results with ICG videoangiography with the scanning
using reflective densitometry with ICG was reported.16 After laser ophthalmoscope were reported by Scheider and Schroedel
using infrared absorption ICG angiography to study the circula- in 1989,45 and Scheider and co-workers described their first
tion of the dog cerebral surface,15 Kogure and associates first experience with this technique and CNV.46
demonstrated choroidal absorption angiography using intra- In 1992, Guyer and co-workers47 and Yannuzzi and asso-
arterial ICG injection and false-color infrared film in monkeys.17 ciates48 introduced the use of a 1024-line digital imaging system
These investigators demonstrated filling of the smaller choroidal to produce high-resolution enhanced ICG images. These
veins and occasional laminar filling of the larger choroidal veins. systems have improved the resolution of ICG videoangiography
A choroidal arterial phase was not noted. David was the first to such that this technique is now of practical clinical value.
perform ICG absorption choroidal angiography in human patients
in 1969.18 These patients underwent intraarterial ICG injections DIGITAL IMAGING SYSTEMS
during carotid angiography. He described diffuse choriocapillaris
filling and choroidal veins draining toward the vortex veins. In The coupling of a digital imaging system with an ICG camera
1971, Hochheimer performed choroidal absorption angiography allows production of enhanced high-resolution (1024-line) images,
in cats using intravenous ICG injections and black-and-white which are necessary for ICG angiography. These systems pro-
infrared film.19 This study solved two major problems. The first duce instantaneous images that decrease patient waiting time
was the use of intraarterial injections and the second was the and expedite possible laser photocoagulation treatment. Digital
inconsistency of the false-color infrared film. imaging systems allow image archiving, hard-copy generation,
Intravenous ICG absorption angiography was first successfully and direct qualitative comparison between fluorescein and ICG
performed in humans by Flower and Hochheimer in 1972.20,21 angiography findings. In addition, these systems are useful for
With this method, the fundus is illuminated with infrared light planning preoperative laser treatment strategies and for moni-
and the reflected light exposes the photographic film. If larger toring the adequacy of treatment postoperatively.
vessels are filled with enough dye to absorb the incident light, the Digital imaging systems contain film and video cameras with
film will not be exposed.21,22 In 1973, Flower and Hochheimer special antireflective coatings and appropriate excitatory and bar-
described a method of ICG fluorescence angiography.23 With rier filters. A video camera is mounted in the camera viewfinder 1705
RETINA AND VITREOUS

and is connected to a video monitor. The photographer selects


the image and activates a trigger that sends the image to the video
adapter. The digitally charged coupling device camera (mega-
plus camera) then captures the images and transmits these
digitized (‘1024 line µ 1024 line’ resolution) images to a video
board within a computer processing unit. Flash synchronization
allows high-resolution image capture. These images are captured
at one frame per second, stored in buffer memory, and displayed
on a high-contrast, high-resolution video monitor. Optical discs
are used to store images after editing. Hard-copy photographs
can be obtained through a printer or slides can be produced
directly from a slide-making device. Finally, via telecommuni-
cations, satellite stations can be placed in laser treatment areas
and in other offices.

TECHNIQUES IN ICG ANGIOGRAPHY


SECTION 10

Advances in ICG angiography were real-time angiography,49


wide-angle angiography,49 and digital subtraction ICG
angiography.50
Real-time ICG angiography uses a modified fundus camera
with a diode laser illumination system that has an output at
805 nm that can produce images at 30 frames per second, and
allows continuous recording. The images can be acquired either
FIGURE 129.1. Wide-angle ICG angiography picture of a patient with
as a videotape or as a single image at a frequency of 30 images
a choroidal nevus. Note the typical butterfly distribution of the
per second. choroidal circulation.
Wide-angle image of the fundus can be obtained by perform- From Yannuzzi LA, Flower RW, Slakter JS: Indocyanine green angiography.
ing ICG angiography with the aid of wide-angle contact lenses. St. Louis: Mosby-Year Book; 1997.
The contact lenses used are the Volk SuperQuad 160, the Volk
Quadraspheric, or the Volk Transequator (Volk, Mentor, OH).
Because the image formed by these lenses lies ~1 cm in front of
the lens, the fundus camera is set on A or +, so that the camera
is focused on the image plane of the contact lens.
This technique allows instantaneous imaging of a large area of
the fundus. The combined use of the contact lens and of the laser
illumination system allows real-time imaging of a wide field of
the choroidal circulation up to 160º of field of view (Figs 129.1
and 129.2).
Digital subtraction ICG angiography uses digital subtraction
of sequentially acquired ICG angiographic frames to image the
progression of the dye front in the choroidal circulation. A
method of pseudocolor imaging of the choroid allows differen-
tiation and identification of choroidal arteries and veins. Digital
subtraction ICG angiography allows imaging of occult CNV
with greater detail and in a shorter period of time than with
conventional ICG angiography.

PHARMACOLOGY OF ICG
ICG was first used in 1957 to measure cardiac output.51 It is a
water-soluble tricarbocyanine dye, which is an anhydro-
3,3,3„,3„-tetramethyl-1,1„-di-(4-sulfobutyl)-4,5,4„,5„-
dibenzoindotricarbocyanine hydroxide sodium salt.
Its molecular weight is 775 Da, and its empirical formula is FIGURE 129.2. A wide-angle ICG angiography picture of a patient
C43H47N2O6S2Na.52 After intravenous injection, ICG is rapidly with a subretinal hemorrhage secondary to idiopathic polypoidal
and almost completely bound to plasma proteins. It has been choroidal vasculopathy. The image shows 160º of the fundus.
thought that ICG is bound to albumin in the blood.53 However, Courtesy of Richard F. Spaide, MD, New York.
80% of ICG in human serum is actually bound to globulins,
such as a-lipoproteins.54
The spectral absorption of ICG in aqueous solution with TOXICITY OF ICG
albumin is between 790 and 805 nm.54–56 ICG is eliminated
from the blood almost exclusively by the liver and is excreted ICG is not very toxic in animals.61–65 Only a few side effects
into the bile. Reabsorption of ICG from the intestine does not have been reported with clinical use.53,61,66,67 In 1978, 240 000
occur.55–58 ICG is not detected in the cerebrospinal fluid,58,59 nor cases of ICG use were reviewed.68 In the early years, when the
1706 does it cross the placental barrier.60 presence of 5% iodide in Cardio-Green was not appreciated,
Indocyanine Green Videoangiography

there were some side effects in patients with iodide allergies. In fundus cannot be observed. This transit time is ~10 s in young
addition, one patient had urticaria and three patients had anaphy- patients and ~12–18 s in older patients.11
lactic reactions. One of these patients died. Among 43 patients
who underwent chronic hemodialysis, three patients with nausea CHARACTERISTICS OF INFRARED
and one patient with a reversible anaphylactoid reaction to ICG WAVELENGTHS
were reported.69
No side effects have been reported in the ophthalmic litera- Near-infrared light is used to perform choroidal angiography
ture. There were no complications after intravenous doses of because it can penetrate pigmented layers7–9 better than the shorter
150–200 mg of ICG in one study.70 During 700 procedures in wavelengths of visible light. The percentage of absorption in
another study, no side effects were reported.36 Thus, ICG is human retinal pigment epithelium and choroid for equal inten-
relatively nontoxic and appears to be safer than fluorescein dye. sities is between 59% and 75% for 500 nm (blue-green visible
Between 5% and 20% of patients receiving fluorescein dye suffer light) and between 21% and 38% for 800 nm (near-infrared light).10
from nausea, headache, or dizziness, and 5–10 in 1000 patients Near-infrared light causes ICG to fluoresce. Patients note that
have allergic reactions.11 infrared light appears as barely visible red light. Therefore, photo-
In a study that reported on ICG angiography performed on phobic patients may tolerate this procedure better than
1226 consecutive patients, there were three (0.15%) mild adverse fluorescein angiography.20
reactions, four (0.2%) moderate reactions, and one (0.05%) Since longer wavelengths are less scattered than shorter wave-
severe adverse reaction. There were no deaths.71 lengths, near-infrared angiography may be performed in patients

CHAPTER 129
Nevertheless, ICG may cause a severe anaphylactic reaction. with diffuse opacities in the ocular media, such as cataract or
ICG angiography should not be performed in patients who are vitreous hemorrhage.24 Infrared light is less harmful than
allergic to iodide, in those who have a history of severe allergies, shorter-wavelength light to the retina; thus, a continuous light
or in those who are uremic.36 In addition, we would not rec- source may be used for high-speed angiography.36 Thermal retinal
ommend its use in patients with liver disease, as the dye is damage, however, can be produced with the infrared light. With
eliminated exclusively by this organ. energies greater than 1 W/cm2, the retinal temperature may rise by
10°C, and acute retinal damage can occur. The safe time expo-
SPECIAL PROPERTIES OF ICG FOR sure must be calculated.80 The risk of near-infrared-induced
CHOROIDAL ANGIOGRAPHY lens damage is minimal with choroidal angiography.36

ICG absorbs light in the near-infrared region of the spectrum MORPHOLOGIC FEATURES OF THE
(maximal absorption is approximately at 790 nm)17 and also CHOROIDAL VASCULATURE
fluoresces in the near-infrared region (maximal emission is
approximately at 835 nm).72–75 Because of its activity at these
longer wavelengths, ICG fluoresces through pigment and hemor- NORMAL MORPHOLOGIC CHOROIDAL
rhage when it is excited by near-infrared light. PATTERNS
Approximately 98% of ICG is bound to plasma proteins,53 Most of the short posterior ciliary arteries enter the eye near the
and, therefore, the dye probably does not leak from the chorio- macula. These choroidal arteries travel radially to the equator.
capillaris.72 This property allows better visualization of the cho- After they have perforated the sclera and have entered the choroid,
roidal vessels because ICG remains in the choroidal vasculature they divide into smaller branches. Interarterial anastomoses are
longer than fluorescein does. However, some authors believe that common in the choroid,81 but they cannot be distinguished with
ICG can pass through the fenestrations of the choriocapillaris.37,40 ICG angiography. The choroidal arteries do not fill at the same
Bill stated that the choroidal blood vessels are permeable to sub- time. The earliest detectable filling of the arteries is usually nasal
stances with molecular weights of 17–156 kDa, such as myo- to the fovea. This region is the area of highest blood perfusion
globin, albumin, and gamma globulin.76 Thus, protein-bound pressure in the eye.21 The individual vessels of the chorio-
ICG may pass through the fenestrations of the choriocapillaris to capillaris cannot be distinguished. The choriocapillaris filling
some extent.36 pattern produces a faint and diffuse homogeneous fluorescence
The liver rapidly removes ICG from the blood after intra- that prevents a clear visualization of the deeper choroidal layers
venous injection.53,57,77 Therefore, there is no significant ICG (Fig. 129.3).79 Choroidal veins run parallel to the periphery and
staining of normal ocular tissues.17,78 eventually form the vortex veins (Fig. 129.4). Venous anastomoses
Another advantage of ICG is that its peak absorption coincides occur between large vessels.81 Veins are larger and more fluor-
with the emission spectrum of the diode laser. This property escent than arteries, but these changes cannot be used to differ-
may allow selective ablation of chorioretinal lesions using ICG entiate them. A horizontal watershed area between the superior
dye-enhanced diode laser photocoagulation when a target tissue and the inferior vessels is sometimes present (Fig. 129.5).36
containing ICG is exposed to the diode laser beam.49,50,75 Choroidal arteries fill between 0.5 and 1 s earlier than does the
central retinal artery. Different phases of the ICG angiogram
TECHNIQUE OF ICG INJECTION have been described.36,82 The mean time values reported follow:
Choroidal artery to choroidal vein 1.8 s
A dye concentration of 0.03 mg/mL is required for maximal Central retinal artery to retinal vein (laminar) 2.0 s
fluorescence of ICG in the choroidal vessels. The dye is diluted Central retinal artery to retinal vein (full) 6.2 s
600 times before it enters the choroidal circulation. Thus, ~20 mg Choroidal artery to vortex vein (initial) 2.0 s
of ICG in 1 mL of aqueous solvent has to be rapidly injected Choroidal artery to vortex vein (maximal) 5.0 s
intracubitally. We currently inject ~50 mg of ICG for diagnostic
studies. Rapid injection is essential in order to delineate various
choroidal filling phases because the majority of the dye bolus ICG ANGIOGRAPHY OF CHORIORETINAL
must be in the choroidal vessels before reaching the retinal DISORDERS
vasculature.79 This injection should be immediately followed by Age-Related Macular Degeneration
a flush of 5 mL of normal saline solution. The timing of photo- Macular disorders may occur secondary to specific choroidal
graphy should be determined by arm to retina time,36 since the vascular properties of this specialized area.83 Only short arteries 1707
RETINA AND VITREOUS

choroidal vessels. They suggested that chronic choroidal


insufficiency may cause CNV.88
Bischoff and Flower36 reviewed 100 ICG angiograms of age-
related macular degeneration (AMD) and described four abnormal
findings:
1. Delayed or irregular choroidal filling, or both. In some
patients, the interval between choroidal arterial and
choroidal venous filling was 3–4 s. However, as these
authors suggest, the significance of this finding is
uncertain because the investigators did not study an age-
matched control group.
2. Generalized arterial changes. These authors describe
‘wandering arteries’ in some patients with AMD.
3. Localized arterial changes. Marked dilatation of macular
choroidal arteries was observed in some patients. These
arteries filled earlier than other vessels and often formed a
loop close to the entrance site. These loops were
anatomically related to overlying fundus lesions. The
SECTION 10

authors felt that a higher blood pressure in the


choriocapillaris overlying vascular abnormalities may cause
dilatation of the choriocapillaris and subsequent macular
disease (Fig. 129.6).84
4. CNV. This finding was observed by ICG angiography in
FIGURE 129.3. The choroidal venous phase of ICG angiography of a only a small number of cases in Bischoff and Flower’s
normal fundus. A diffuse homogeneous fluorescence is noted in the
series (Fig. 129.7).
macular area. Details of the deeper vessels in this area are not seen.
Around the macular area, the choroidal veins are homogeneous in
caliber and distributed uniformly to converge to form the vortex veins. Hayashi and co-workers reported that ICG angiography was par-
ticularly useful to detect occult CNV.38 The same investigators
showed that ICG leaks from CNV into the subretinal space.
and arterioles are present between the ciliary arteries and the However, the leakage is slower and of lesser magnitude than
choriocapillaris in the macula. This cluster of arterial branches fluorescein leakage of CNV. This slow ICG leakage is some-
is greater in the macula than in any other region. These findings times useful in defining the borders of CNV and has delineated
may be responsible for the high pressure and rapid blood flow of the neovascularization not well identified with fluorescein angio-
macula.13,21,83 These pressure changes may cause choriocapil- graphy.40 Destro and Puliafito39 also found this technique very
laris disease83 and CNV.84 The loss of contractility of the arterial useful to study occult CNV, particularly those with overlying
wall that occurs with age may cause dilatation of the vessel.85,86 hemorrhage or those that had recurred on the edge of previously
Watershed areas of both arterial and venous circulations may treated areas. CNV adjacent to chorioretinal scars showed greater
be present in the macular area.87,88,89 Hayashi and de Laey contrast between the new vessels and the adjacent scar with
described a relationship between these choroidal ‘watershed ICG than with fluorescein angiography. Chorioretinal scars
zones’ and macular lesions and showed evidence of abnormal were hypofluorescent because of less ICG extravasation and the

a b

FIGURE 129.4. (a and b) The choroidal veins converge to form the vortex veins, with a large variability of patterns.
1708
Indocyanine Green Videoangiography

patients in their series because of information obtained through


digital ICG videoangiography. These authors found that digital
ICG videoangiography is especially useful for patients with
poorly defined CNV, pigment epithelial detachments (PEDs),
and recurrent CNV. In many cases, the late ICG images reveal
a hyperfluorescent area corresponding to an area of subretinal
neovascularization that cannot be detected by fluorescein angio-
graphy. The finding of a hyperfluorescent spot on the late ICG
angiogram can separate the neovascularized portion from the
serous portion of a PED.
Yannuzzi and co-workers used ICG angiography to study 235
consecutive AMD patients with occult CNV and associated vas-
cularized PED. These eyes were divided into two groups, depend-
ing on the size and delineation of the CNV. Of the 235 eyes, 89
(38%) had a solitary area of neovascularization that was well
delineated, no more than one disk diameter in size, and defined
as focal CNV. The other 146 eyes (62%) had a larger area of
neovascularization, with variable delineation defined as a

CHAPTER 129
plaque CNV.90
In a further report, 657 consecutive eyes with occult CNV by
fluorescein angiography were studied with ICG angiography. Of
413 eyes with occult CNV without PEDs, focal areas of neovas-
cularization were noted in 89 (22%). Overall, 142 eyes (34.3%)
had lesions that were potentially treatable by laser photoco-
agulation based on additional information provided by ICG
FIGURE 129.5. A horizontal watershed area between the superior angiography. Of the 235 eyes with occult CNV and vascularized
and the inferior vessels found in a minority of patients.
PEDs, 98 (42%) were eligible for laser therapy based on ICG
angiography findings. The authors calculated that ICG angio-
graphy enhances the treatment eligibility by approximately
one-third.91
In an expanded series, the same authors reported their results
on ICG angiography study of 1000 consecutive eyes with occult
CNV by fluorescein angiography.92 They recognized three morpho-
logic types of CNV, which included focal spots (Figs 129.8 and
129.9), plaques (Fig. 129.10) (well-defined and poorly defined),
and combination lesions (Fig. 129.11) (in which both focal spots
and plaques are noted). Combination lesions were further sub-
divided into marginal spots (focal spots at the edge of a plaque of
neovascularization), overlying spots (hot spots overlying plaques
of neovascularization), or remote spots (a focal spot remote from
a plaque of neovascularization).
The relative frequency of these lesions was as follows: focal
spots 29%; plaques 61%, consisting 27% of well-defined plaques
and 34% of poorly defined plaques; and combination lesions
8%, consisting of 3% of marginal spots, 4% of overlying spots,
and 1% of remote spots (Table 129.1).92 A follow-up study from
the same authors of patients with newly diagnosed unilateral
occult CNV secondary to AMD showed that the patients tended
to develop the same morphologic type of CNV in the fellow eye.93
Finally, Chang and associates94 reported on the clinicopatho-
logic correlation of AMD with CNV detected by ICG angio-
FIGURE 129.6. The choroidal mid-transit phase of an ICG angiogram graphy. Histopathologic examination of the lesion revealed a
in a patient with age-related macular degeneration. Dilated tortuous thick subretinal pigment epithelium CNV corresponding to the
arterial loops can be observed on the temporal side. plaquelike lesion seen with ICG angiography.
The studies just discussed demonstrate that ICG videoangio-
graphy is an important adjunctive study to fluorescein angiography
relative lack of choroidal vessels in those areas. ICG remained in the detection of CNV. Fluorescein angiography appears to be
selectively in and around the CNV.39 In addition, Scheider and more sensitive than ICG videoangiography in imaging fine capil-
colleagues described enhanced imaging of CNV by using the laries that connect larger vessels and capillaries at the prolifer-
scanning laser ophthalmoscope with ICG angiography.46 ating edge of well-defined CNV. Although fluorescein angiography
High-resolution ICG images can now be produced by com- may image well-defined CNV better than ICG videoangio-
bining digital imaging systems and an ICG camera.47,48 This graphy in some cases, ICG videoangiography can convert occult
technological advance now allows the theoretical advantage of CNV by fluorescein angiography into well-defined classic CNV
ICG over fluorescein dye to finally be realized. eligible for ICG-guided laser treatment in ~30% of cases.95,96
Yannuzzi and associates48 showed that occult CNV could be Thus, the best imaging strategy to detect CNV is to perform
converted into classic, well-defined CNV in 39% of the 129 fluorescein angiography and ICG videoangiography.
1709
RETINA AND VITREOUS

FIGURE 129.7. (a and b) Red-free


photography and the late phase of the
fluorescein angiogram of a poorly defined
choroidal neovascular membrane. (c–e) In the
ICG angiograms, the limits of the neovascular
membrane can be better visualized.
(f) Increased magnification shows some FVs.
(g and h) With computer analysis, the
membrane observed in the ICG angiogram can
be traced and superimposed on the fluorescein
angiogram. In some instances, ICG
angiography improves the definition of a
choroidal neovascular membrane.

a b
SECTION 10

c d

e f

g h

1710
Indocyanine Green Videoangiography

a b c

FIGURE 129.8. (a) Clinical photograph of a patient with exudative macular detachment. (b) Fluorescein angiography shows occult CNV and a
serous PED. (c) ICG angiography study reveals a focal area, ‘hot spot’, of neovascularization at the superonasal edge of the serous PED.
(a–c) From Yannuzzi LA, Flower RW, Slakter JS: Indocyanine green angiography. St. Louis: Mosby-Year Book; 1997.

CHAPTER 129
a b c

FIGURE 129.9. (a) Clinical photograph of a patient with exudative macular detachment and subretinal hemorrhage. Note the presence of a
blood level in the PED. (b) Fluorescein angiography shows blockage of fluorescence by the subretinal hemorrhage. By fluorescein angiography,
there is probably occult CNV in the temporal macula. (c) ICG angiography of the same patient reveals a hot spot superior to the optic disk
corresponding to a polypoidal choroidal vasculopathy. No active CNV is visible in the macular area.
(a–c) From Yannuzzi LA, Flower RW, Slakter JS: Indocyanine green angiography. St. Louis: Mosby-Year Book; 1997.

FIGURE 129.10. (a) Clinical photograph of a


patient who underwent multiple laser
treatments for recurrent CNV. (b) Fluorescein
angiography shows a large area of staining in
the macula that may be interpreted as occult
CNV. (c) Mid-phase ICG study reveals a plaque
of CNV corresponding in morphology to the
area of staining on fluorescein angiography.
(d) Late-phase ICG study better shows the
plaque lesion.
(a–d) From Yannuzzi LA, Flower RW, Slakter JS:
Indocyanine green angiography. St. Louis: Mosby-Year
a b Book; 1997.

c d
1711
RETINA AND VITREOUS

a b c

FIGURE 129.11. (a) Clinical photograph of a large serous PED surrounded by an exudative detachment of the neurosensory retina. (b) Mid-
phase ICG study demonstrates a focal spot at the margin of the PED. (c) Late-phase ICG study reveals the presence of a plaque of inactive CNV
that is not evident in the earlier phase of the study. This is an example of combination lesion: a hot spot at the margin of a plaque. As in this
case, many hot spots evidenced by ICG angiography may correspond to the variant neovascularization secondary to polypoidal choroidal
vasculopathy.
SECTION 10

(a–c) From Yannuzzi LA, Flower RW, Slakter JS: Indocyanine green angiography. St. Louis: Mosby-Year Book; 1997.

(37.5%) of 16 eyes.99 Importantly, these studies set the founda-


TABLE 129.1 Relative Frequency of Various Lesions Using
Indocyanine Green Angiography in Eyes With Occult Choroidal
tion for future prospective studies of ICG-guided laser treat-
Neovascularization ment. In addition, they proved that the presence of a PED is a
poor prognostic factor in the treatment of exudative AMD.
Lesion No. of Eyes (%) A recent study prospectively evaluated 185 consecutive eyes
Focal spots 283 (29) with exudative AMD and a well-delineated area (hot spot or focal
area) of hyperfluorescence by ICG angiography. All the patients
Plaques 597 (61) were divided into two groups (with PED and without PED). Of
Well-defined plaques 265 (27) the 185 eyes, 99 eyes without PED achieved a 71% rate of oblitera-
tion at 6 months and a 48% rate of obliteration at 12 months.
Poorly defined plaques 332 (34)
Eyes with PED did significantly worse, with an obliteration rate
Combination lesions 84 (8) of the CNV of 23% at 12 months. The overall success rate was
Marginal spots 35 (3) 36% at 12 months.96
A possible explanation for the high recurrence rate of occult
Overlying spots 37 (4) CNV after laser photocoagulation, particularly when a vascu-
Remote spots 12 (1) larized PED is present, may be found in the peculiar anatomy of
the CNV in such cases. It has been observed that there is a
Other lesions 20 (2)
variant of CNV where the neovascularization is fed both by a
Multiple spots 7 (1) choroidal and by a retinal component to create a retinochoroidal
Null 13 (1) anastomosis (RCA) (Figs 129.12 and 129.13) (JS Slakter, LA
Yannuzzi, U Schneider, et al, personal communication, 1998).100
From Guyer DR, Yannuzzi LA, Slakter JS, et al: Classification of choroidal
neovascularization by indocyanine green angiography. Ophthalmology 1996;
In a report by Kuhn and colleagues, RCAs were identified as
103:2054, 1996. occurring in 93% of patients with CNV associated with a serous
PED. They reported a poor success rate from laser treatment
as well.100
Slakter and co-workers followed prospectively 150 patients
with newly diagnosed exudative AMD (JS Slakter, LA Yannuzzi,
ICG-Guided Laser Treatment of CNV in AMD U Schneider, et al, personal communication, 1998). All had
Slakter and associates performed ICG-guided laser photocoagu- clinical and fluorescein angiographic evidence of occult CNV,
lation in 79 eyes with occult CNV.97 The occult CNV was suc- and each demonstrated focal areas of hyperfluorescence on ICG
cessfully eliminated with stabilized or improved visual acuity in angiography, believed to be representative of CNV. Thirty-one
29 (66%) of 44 eyes with occult CNV associated with neurosen- (21%) of the 150 eyes were found to have an RCA. In 82 eyes,
sory retinal elevations, and in 15 (43%) of 35 eyes with occult the occult CNV was associated with a serous PED. Twenty-two
CNV associated with PED. This study demonstrated that in (27%) of these patients were noted to have RCA. In the remain-
some cases, ICG videoangiography imaging can successfully ing 68 cases (occult CNV without serous PED), nine eyes (13%)
guide laser photocoagulation of occult CNV. were found to have an RCA. Associated clinical features of
In another pilot study of ICG-guided laser treatment of occult RCAs were identified in preretinal or intraretinal hemorrhages
CNV, Regillo and colleagues had similar results.98 at the site of the lesion, dilated tortuous retinal vessels, sudden
Guyer and co-workers reported on a pilot study of ICG- termination of a retinal vessel, and cystoid macular edema.
guided laser photocoagulation of 23 eyes that had untreatable The same authors found that the success rate of laser photo-
occult CNV secondary to AMD with focal spots at the edge of a coagulation of RCAs without serous PED was 66%, whereas with
plaque of neovascularization on the ICG study.99 ICG-guided serous PED it dropped to 14% (JS Slakter, LA Yannuzzi, U
laser photocoagulation was applied solely to the focal spot at the Schneider, et al, personal communication, 1998). Thus, the pres-
edge of the plaque. At 24 months of follow-up, anatomic success ence of an RCA may well provide a key to understanding the
1712 with resolution of the exudative findings was obtained in six poor outcome for laser treatment in this subgroup of patients.
Indocyanine Green Videoangiography

FIGURE 129.12. (a) Clinical photograph of a


patient with a serous PED and subretinal
hemorrhage. (b) High-magnification early-phase
ICG study reveals retina arterial and venous
branches connecting to the intraretinal
neovascularization of a retinal angiomatous
proliferation.
(a and b) From Yannuzzi LA, Flower RW, Slakter JS:
Indocyanine green angiography. St. Louis: Mosby-Year
Book; 1997:166.

a b

CHAPTER 129
FIGURE 129.13. (a) Clinical photograph
demonstrates a serous PED and two focal
subretinal hemorrhages at the site of two RAPs.
(b) High-magnification red-free photograph
better shows two small retinal vessels that
appear to extend into each of intraretinal new
vessels. (c) Mid-phase ICG study reveals two
focal hot spots corresponding to the RAPs.
(d) Clinical photograph after laser treatment.
Note the persistence of an anastomotic red
blood vessel at the center of the white laser
burn.
a b (a–d) From Yannuzzi LA, Flower RW, Slakter JS:
Indocyanine green angiography. St. Louis: Mosby-Year
Book; 1997:166.

c d

Sorenson and associates reported on ICG-guided laser treat- were identified. Hot spots were noted to be of three distinct
ment of recurrent occult CNV secondary to AMD.101 Of 66 eyes patterns: polypoidal choroidal neovascularization (polypoidal
that entered in the study, only 29 (44%) were eligible for laser CNV) in 21 of 34 eyes, or 62%; retinal angiomatous
treatment, and of these 29 eyes, 18 (62%) had anatomic success proliferation (RAP) in 11 of 34 eyes, or 30%; and focal occult
with an average follow-up of 6 months. Interestingly, 56% of the CNV in two of 34 eyes, or 8%. The authors concluded that focal
patients remained untreatable by ICG angiography guidance, area of intense hyperfluorescence or so-called hot spot seen
and even with treatment, 11 of 29 patients had incomplete on ICG angiography in exudative ARMD was due to one of
resolution or worsening of the exudative manifestations. three possible forms of neovascularization: most frequently
The spectrum of neovascular AMD expanded over the past polypoidal CNV, less commonly RAP, and infrequently
decade, and entities such as retinal angiomatous proliferation nonspecific, focal occult CNV. 108
(RAP) and polypoidal choroidal vasculopathy (PCV) emerged.102–107
After retina specialists became aware of these variants of neo- Central Serous Chorioretinopathy
vascular AMD, many of the previously described ‘hot spots’ ICG angiography of patients with central serous chorioretino-
appeared to be RAP or polypoidal lesions. That might explain pathy may show, in the early phase of the study, diffuse or
the variability in the response of ICG guided treated CNV, espe- multifocal areas of choroidal hyperpermeability not associated
cially regarding the ‘hot spots’. Fernandes et al described the with abnormalities detectable by fluorescein angiography or
nature of focal areas of hyperfluorescence or hot spots imaged clinical examination (Fig. 129.14); in the late phase of the ICG
with ICG from a total of 190 patients (220 eyes) with exudative study, there is dispersion of the fluorescence and a distinctive
ARMD. Thirty patients and 34 eyes (16%) with hot spots silhouetting of the larger choroidal vessels.47,127–129 1713
RETINA AND VITREOUS

a b c

FIGURE 129.14. (a) Fluorescein angiography of a patient with central serous chorioretinopathy demonstrates multifocal pinpoint areas of
leakage at the posterior pole. (b) Wide-angle ICG study reveals diffuse choroid hyperpermeability and scattered areas of hyperfluorescence.
SECTION 10

(c) Late-phase ICG study shows multifocal areas of leakage throughout the fundus.

Diabetic Retinopathy the borders of the pigmented tumors may be better delineated
The choroidal angiography findings of 60 patients with diabetic by ICG angiography than by fluorescein angiography, and there-
retinopathy were reported by Bischoff and Flower.36 Most of the fore ICG angiography may serve as a more accurate tool for
patients with proliferative diabetic retinopathy showed irregular assessment of tumor growth.24,25,36 Bischoff and Flower reported
and delayed choroidal filling. Approximately 50% of patients that in some lightly pigmented tumors, ICG angiography was
with background diabetic retinopathy showed such changes. able to resolve some of the larger vessels of the tumor with
staining of their walls and leakage into the mass.34
Choroidal Tumors In contrast to pigmented choroidal melanomas, choroidal
Choroidal ICG absorption angiography and ICG fluorescence hemangiomas show progressive hyperfluorescence because they
angiography have been used to study choroidal are composed of vascular channels29,112–114; thus, ICG angio-
tumors.24,25,27,29,36,70,112–115 This technique can visualize the vas- graphy may be useful in distinguishing choroidal hemangiomas
cularization and filling patterns of nonpigmented and lightly from some choroidal melanomas. Choroidal metastasis originat-
pigmented tumors. The near-infrared light is absorbed by mela- ing from different primary tumors will show different angiographic
nin of heavily pigmented tumors, such as choroidal melanomas, characteristics, depending on the vascularity and pigmentation
and thus blocks ICG fluorescence (Fig. 129.15). Nevertheless, of the mass: Metastasis of breast carcinoma blocks choroidal
fluorescence,115 whereas metastasis of thyroid carcinoma70 and
metastatic bronchial carcinoid tumors115 are hyperfluorescent on
ICG angiography. However, intense late hyperfluorescence may
be more characteristically observed with choroidal hemangio-
mas than with choroidal metastasis.

Choroiditis
In patients with birdshot choroiditis, ICG angiography was found
to be superior to fluorescein angiography in defining the typical
patches. Multiple hypofluorescent lesions radiating to the peri-
phery are observed between the choroidal veins.115 These lesions
are consistent with choriocapillary dropout and sometimes are
more evident on later stages of the angiogram (Fig. 129.16). In
patients with serpiginous choroiditis, actively inflamed areas
within the lesion block choroidal fluorescence (Fig. 129.17).114
With resolution, the fluorescence of choroidal vessels can be
seen in the previously hypofluorescent areas. Late hyperfluor-
escence can be seen at sites at which CNV has evolved. Overt
leakage from choroidal vessels was observed on ICG angiography
in a patient with acute choroiditis.115 Complete clinical angio-
graphic resolution was noted after treatment.
Slakter and associates reported on the ICG angiography find-
ings in a series of 14 patients with multifocal choroiditis. Fourteen
(50%) of the 28 eyes were found to have large hypofluorescent
spots in the posterior pole on ICG angiography, which, in most
cases, did not correspond to clinically or fluorescein angiograph-
FIGURE 129.15. Late-phase ICG angiogram of a patient with a ically detectable lesions. In seven eyes exhibiting enlarged blind
juxtapapillary choroidal melanoma. No intratumoral vessels can be spots on visual-field testing, ICG angiography showed confluent
noted because the heavy pigmentation of the tumor absorbs the hypofluorescence surrounding the optic nerve (Fig. 129.18).
1714 near-infrared wavelengths. The ICG angiogram was found useful in evaluating the natural
Indocyanine Green Videoangiography

showed a reduction in the size and number of the hypofluor-


escent spots in three patients, with complete resolution of these
angiographic lesions noted in the fourth patient.116
Acute multifocal posterior placoid pigment epitheliopathy
(AMPPPE) lesions are hypofluorescent by ICG angiography in
both the early and the late phases of the study (Fig. 129.19).
The ICG choroidal hypofluorescence in this condition may be
due to a partial choroidal vascular occlusion secondary to
occlusive vasculitis.117,118
Multiple evanescent white dot syndrome presents with a
characteristic ICG angiography picture in the acute phase of the
disease (Fig. 129.20).119 Unlike the subtle white dots seen
clinically or the indistinct punctate hyperfluorescence seen with
fluorescein angiography, ICG angiography shows a pattern of
hypofluorescent spots throughout the posterior pole and periph-
eral retina. These hypofluorescent spots appear ~10 min after
the dye injection in the mid-ICG phase and persist throughout
the remainder of the study. These spots appear larger than the

CHAPTER 129
white dots seen clinically, varying in diameter from less than 50
to ~500 mm. Many more lesions can easily be identified on ICG
angiography than on fundus examination or fluorescein
angiography.
In a few cases, there is also a ring of hypofluorescence sur-
FIGURE 129.16. Multiple hypofluorescent lesions straddle the rounding the optic nerve. In these patients, a blind spot enlarge-
choroidal veins in birdshot choroiditis. ment on visual-field examination is always present. The
resolution of the hypofluorescent ring around the optic nerve is
accompanied by a normalization of the visual field.
course in two patients with multifocal choroiditis as well as a During the convalescent phase, there is resolution of the hypo-
response to oral prednisone therapy in four others. The ICG angio- fluorescent spots seen on ICG angiography with return of visual
graphy performed in these patients showed changes correlating function and normalization of the clinical examination.
with the clinical course. After administration of the oral pred- Digital ICG videoangiography is thus valuable in the diagnosis
nisone, the patients were noted to have decreased symptoms and monitoring of patients with choroiditis. Further investigation
and less vitreitis on clinical examination. The ICG angiography is warranted in this area.

FIGURE 129.17. (a and b) Late-phase


fluorescein angiograms of an eye with a
recurrence of serpiginous choroidopathy.
(c and d) Late-phase ICG angiograms show
marked hypofluorescence in the areas of active
inflammation.

a b

c d
1715
RETINA AND VITREOUS

a b c

FIGURE 129.18. (a) Fluorescein angiography of a patient with multifocal choroiditis. Note the presence of an atrophic ring around the optic disk.
(b) Late-phase ICG angiography reveals the presence of multifocal hyperfluorescent lesions that do not have a clinical or fluorescein angiography
correspondence. (c) Wide-angle ICG shows scattered hypofluorescent spots around the optic disk and around the superotemporal vortex vein.
SECTION 10

Polypoidal Choroidal Vasculopathy These polypoidal structures correspond to the reddish orange
ICG angiography has been used to detect and characterize the choroidal excrescence seen on clinical examination. In the later
abnormality of polypoidal choroidal vasculopathy with enhanced phase of the angiogram, there is a uniform disappearance of the
sensitivity and specificity.120–122 In the initial phases of the ICG dye (‘washout’) from the bulging polypoidal lesions. The late
study, a distinct network of vessels within the choroid becomes ICG staining characteristic of occult CNV is not seen in the
visible. polypoidal choroidal vasculopathy vascular abnormality.
Early in the course of the ICG study, the larger vessels of the
polypoidal choroidal vasculopathy network start to fill before the Retinal Angiomatous Proliferation
retinal vessels, but the area within and surrounding the network ICG angiography is useful in determining the characteristics of
is relatively hypofluorescent compared with the uninvolved the variant of exudative AMD, retinal angiomatous proliferation,
choroid. Shortly after the network can be identified on the ICG better than fluorescein angiography.102–107 At stage I, the neo-
angiogram, small hyperfluorescent ‘polyps’ become visible vascularization appears as a focal area of usually very intense
within the choroid. hyperfluorescence (hot spot). There is also some late extension

FIGURE 129.19. (a) Clinical photograph of a


patient with acute multifocal posterior placoid
pigment epitheliopathy demonstrates the pale,
flat, and the placoid lesions involving the
pigment epithelium. (b) Late-phase fluorescein
angiography demonstrates the optic nerve
staining and mottled hyperfluorescence at the
periphery of the lesion. (c) Early-phase ICG
study shows uniform hypofluorescence of the
lesion. (d) Late-phase ICG study demonstrates
a well-demarcated hypofluorescent lesion.
(a–d) From Yannuzzi LA, Flower RW, Slakter JS:
Indocyanine green angiography. St. Louis: Mosby-Year
a b Book; 1997:242.

c d
1716
Indocyanine Green Videoangiography

a b c

CHAPTER 129
d e f

g h i

FIGURE 129.20. (a) Color photograph of a patient with acute onset of multiple evanescent white
dot syndrome. Only a few yellowish spots are visible temporally. (b) The spots are better seen
with a red-free light. (c) Mid-phase fluorescein angiography demonstrates a few hyperfluorescent
spots and staining of the disk margin. (d) Late-phase fluorescein angiography shows marked
staining of the optic nerve. (e) Late-phase ICG study shows a hypofluorescent ring around the
optic nerve. (f and g) There is also evidence of multiple hypofluorescent spots at the posterior
pole and in the mid-periphery. (h) Goldmann visual field demonstrates an enlarged blind spot.
(i) Late-phase ICG study 4 months later demonstrates resolution of the hypofluorescent ring
around the optic nerve. (j) Goldmann visual field of the same day shows resolution of the
enlarged blind spot.
(a–j) From Yannuzzi LA, Slakter JS, Flower R: Indocyanine green angiography. St. Louis: Mosby-Year Book; 1997.

1717
RETINA AND VITREOUS

of leakage within the retina from the intraretinal neovasculari- Shiraga et al110 and Staurenghi et al111 reported improved or
zation (IRN) which is quite characteristic for RAP. In some stabilized visual acuity between 70% and 75% of the cases
cases, a retinal–retinal anastomosis can be visualized. In stage II, following FV photocoagulation. In many cases, the resolution of
the ICG angiogram, besides the hot spot of the IRN, reveals a CNV and associated edema resolved dramatically and within
serous PED that remains hypofluorescent. In stage III, ICG angio- hours of the FVT.
graphy differentiates the serous component of the PED that FVT is a two-step process, including identification and location
remains dark, from the vascularized component which becomes of FVs with high-speed ICG angiography, and FV photocoagu-
hyperfluorescent. At this stage, ICG angiography may sometimes lation delivery of laser energy through a slit-lamp delivery sys-
reveal RCA. tem. It is necessary, therefore, to transfer information regarding
FV location from the ICG image of the angiograms to the slit-
OTHER CONDITIONS lamp view of the fundus of the eye to be treated. Since the
landmarks between the angiograms and the fundus are different,
Choroidal involvement in the presumed ocular histoplasmosis their reinterpretation is needed, with potential minor deviations
syndrome has been described with ICG angiography.31,36 and thus lack of precision. This may result in a larger area being
Bischoff and Flower reported no significant abnormality in ICG treated than really necessary, which limits how close the treat-
angiograms of patients with Best’s disease.36 In these eyes, it is ment can be applied to the fovea.
our experience that a hypofluorescent lesion in the macula can The main advantages of FVT are: (1) Even when the CNV is
be found, which most likely corresponds to a blocking effect of large only a small fundus area is photocoagulated; (2) the photo-
SECTION 10

the abnormal material, although choroidal nonperfusion at that coagulation is away from the CNV, thus sparing the fovea and
area cannot be ruled out. the presumably damaged RPE associated with the CNV; (3)
Reduced filling of the choriocapillaris in cases of myopia has there are no adverse events due to the laser burn even after
been noted with ICG angiography.36 ICG angiography is also multiple laser treatments; (4) there are no untoward side effects
particularly useful in patients with mild vitreous hemorrhage, to the treatment, even when administered multiple times; and
which prevents evaluation by fluorescein angiography.36 (5) the worst adverse event reported to date is failure of treat-
ment, in which case other treatments can be applied. FVT does
CHOROIDAL BLOOD FLOW STUDIES not cause irreversible damage or adverse events that might
impede other therapies.126 The main disadvantages are: (1) the
ICG angiography has also been used to investigate ocular hemo- difficulty in precisely aiming the laser beam to the FV since the
dynamics. This technique is enhanced by the low affinity of ICG location needs be transferred from the angiogram to the fundus
to ocular tissue structures as shown with 123I ICG measure- view; (2) reopening of the successfully treated FVs; (3) damage
ments.76 Flower, Bischoff, Prünte and their associates41–44 have to the overlying RPE producing a paracentral scotoma that can
used choroidal filling times to assess the velocity of the choroidal be seen bt the patient; (4) changes in fundus pigmentation
circulation with some success. These techniques have yet to may cause a large variability in the efficiency of the photocoa-
demonstrate clinical usefulness. In 1979, Ernest and Goldstick gulation; and (5) limitations in getting the treatment close to
estimated choroidal blood flow in monkeys by determining the the fovea.126
rate of clearance of ICG from the choroidal circulation.117 Several Characterizing and defining FVs is not an easy task; the same
authors have attempted to quantify morphologic and dynamic can be stated about understanding the angiographic–anatomic
parameters in the choroidal circulation. Quantification of effec- correlation, epecially when often the anatomic and functional
tive nutrient choroidal blood flow is difficult, however. Fluor- relationships are also difficult to interpret. To solve this problem
escence is a superficial phenomenon. It is not proportional to Flower proposed a theoretical anthropomorphic model of the CNV
the quantity of dye in the blood vessel,79,124 and it depends on and the FVs.130 The model implies that FVs are not the small
multiple variables such as vessel size, the cardiovascular system, abnormal capillary-like vessels that arise from the choriocapil-
and the injection technique.125 laris and feed directly the CNV, but vessels of the inner layer of
medium-sized choroidal arterioles and venules that supply the
FEEDER VESSEL TREATMENT OF region of the choriocapillaris that feeds the CNV. Thus, it is not
CHOROIDAL NEOVASCULARIZATION necessary that there is a direct anatomic connection between the
FVs identified in the ICG angiogram and the CNV. The close
Photocoagulation of the feeder vessels (FVs) has been investigated proximity of these FVs in the inner layer of the medium-sized
for the treatment of the CNV, especially when the neovascular choroidal arterioles and the associated CNV vessel penetrating
membranes are under the fovea in which direct treatment with Bruch’s membrane may result in the FV and CNV appearing to
laser would cause a large and irreversible scotoma. Before the be contiguous in an ICG angiogram image. In the model system,
availability of antiangiogenic therapy, many cases of subfoveal the choriocapillaris acts like a relief valve inserted in the afferent
CNV had a poor visual prognosis, both with laser photocoa- vessel pathway, proximal to the CNV membrane.
gulation and with photodynamic therapy, due to the size or In some classic lesions, the capillary-like segments connect-
angiographic subtype of the lesion. ing the CNV to the choriocapillaris (i.e., the penetrating vessels)
Feeder vessel treatment (FVT) is an attractive combination actually can be long enough to be detectable as FVs in angiogram
since it is not drug-based and acts directly on the source of CNV images and, therefore, directly treatable by photocoagulation,
blood flow, rather than the membrane itself.126 The rationale of instead of photocoagulating the associated Sattler’s layer vessel
treatment of the FVs of the choroidal neovascular membranes as in the case of occult CNV lesions (Fig. 129.21).
arose from the initial concept of laser photocoagulation of the Since the submacular choriocapillaris is a true vascular plexus,
FVs in diabetic retinopathy.109 based on available histologic and hemodynamic data, a more
Early attempts of FVT failed due to lack of optimal technology sophisticated model was developed to simulate changes in blood
in the imaging of the fundus as well as in delivery of laser with flow through larger choriocapillaris areas after FV photocoagu-
precision.126 New equipment in the late 1990s demonstrated lation.131 This model can explain why partial closure of the FV
the feasibility of FVT of subfoveal and juxtafoveal CNV with is enough to reduce the blood flow through the CNV achieving
efficacy.110,111,130 its functional closure. If reduced choriocapillaris blood flow is a
1718
Indocyanine Green Videoangiography

a b

CHAPTER 129
c d

FIGURE 129.21. Treatment of a ‘mixed’ CNV lesion. (a) Pretreatment angiogram. (b) The penetrating vessel (yellow arrow) leading to the
‘classic’ component was photocoagulated, resulting in closure of only the ‘classic’ component, as demonstrated on subsequent ICGA. (c) About
2 weeks later, the photocoagulated penetrating vessel reopened. Rather than re-treating the penetrating vessel, the FV (green arrow) that
supplied blood to both the ‘classic’ and ‘occult’ components was photocoagulated. (d) Subsequent ICGA showed that both lesion components
had been closed, demonstrating that closure of the Sattler’s layer FV supplying blood to the area of choriocapillaris (CC) from which the lesion’s
penetrating vessels originated resulted in stopping blood flow through both CNV components. The mixed lesion is shown schematically in the
cartoon to the left of the angiogram images showing the penetrating vessels in yellow and the FVs in green.
(a–d) From Flower RW, Staurenghi G: Trataminento de los vasos nutrícios de la neovascularización coroidea. In: Monés J, Gómez Ulla, eds. Degeneración Macular
Asociada a la Edad. Barcelona: Prous Science; 2005.

hemodynamic mechanism of successful CNV treatment, the end- be more accurately placed and it is possible to get closer to the
point of any modality of laser treatment is not total obliteration fovea. On the other hand, the thermal energy is more selectively
of the CNV, which may result frequently in recurrence, but confined in the vessel, making its closure more efficient and
reduction of the CNV blood flow.131,132 thus avoiding unnecessary thermal energy absorption by the
Dye-enhanced photocoagulation of FVs using ICG had been surrounding tissues (Fig. 129.22). Immediately following FV
demonstrated in animal studies,130 and in human eyes.133 In prev- closure, the presence of incarcerated ICG dye in the vessel
ious studies, dye-enhanced photocoagulation with ICG con- adjacent to the burn proves that the blood flow has stooped in
sisted of intravenous injection of large amounts of ICG dye in the treated vessel. (Fig. 129.23)
order to accumulate the dye in the CNV and thus theoretically Compared to some of the current treatments for CNV, FVT
enhancing the uptake of laser energy during confluent has some advantages for the patient, if ICG is not contra-
photocoagulation.134,135 indicated: it is minimally invasive, safe, inexpensive, has no
Flower has described a technique by which FV can be identified cumulative effect, and its failure does not preclude use of other
and treated at the same time with ICG dye-enhanced photo- therapies since it does not leave an irreversible damage. These
coagulation with an adapted fundus camera that can deliver a attributes make FVT another attractive first-line option to treat
diode laser beam.136,137 With this modality, the laser burns can exudative AMD.126
1719
RETINA AND VITREOUS

FIGURE 129.22. The effects of FVT by


conventional photocoagulation compared to
ICG-dye enhanced photocoagulation (ICG-
DEP). The cartoons suggest the differences in
location for the epicenter of light to heat
transduction and the volume of fundus tissue
involved in the two methods of
photocoagulation. With ICG-DEP the tissue
volume in which transduction takes place is
shifted away from the RPE to the target vessel,
and it becomes more focused, thereby
reducing the volume of surrounding tissue
damaged. The left-hand image is from a late
phase ICGA of an eye in which FVT was by
conventional photocoagulation; the thermal
damage was great enough that the nerve fiber
layers overlying the laser spot were sufficiently
damaged to produce a scotoma, and the
associated dark arcuate area presumably was
due to atrophic nerve fibers. The right-hand
SECTION 10

image is from a posttreatment ICGA that


demonstrates an additional advantage to ICG-
DEP, namely that immediately following
successful FV closure, incarcerated ICG dye
often can be detected at the laser burn site,
which provides immediate proof that blood flow
in the target vessel had stopped.
From Flower RW, Staurenghi G: Trataminento de los
vasos nutrícios de la neovascularización coroidea. In:
Monés J, Gómez Ulla, eds. Degeneración Macular
Asociada a la Edad. Barcelona: Prous Science; 2005.

FIGURE 129.23. Results of ICG-DEP FVT of a


classic CNV. The circles indicate the location of
the CNV area in each ICGA image, and the
arrow in the pretreatment image identifies the
FV. In this case, immediately posttreatment,
ICG-stained blood was incarcerated in the CNV
as well as in the short FV segment distal to the
photocoagulation site. Shortly thereafter, a
posttreatment angiogram demonstrated no
filling of the CNV’s FV, although fluorescence
from the dye-stained blood incarcerated in the
CNV persisted – less intensely, however, than in
the immediate posttreatment angiogram.
Subsequent angiograms demonstrated that
blood flow to the CNV had been successfully
stopped by the treatment.
From Flower RW, Staurenghi G: Trataminento de los
vasos nutrícios de la neovascularización coroidea. In:
Monés J, Gómez Ulla, eds. Degeneración Macular
Asociada a la Edad. Barcelona: Prous Science; 2005.

1720
Indocyanine Green Videoangiography

REFERENCES
1. MacLean AL, Maumenee AE: Hemangioma 23. Flower RW, Hochheimer BF: A clinical 40. Hayashi K, Hasegawa Y, Tazawa Y, et al:
of the choroid. Trans Am Ophthalmol Soc technique and apparatus for simultaneous Clinical application of indocyanine
1959; 57:171. angiography of the separate retinal and angiography to choroidal
2. Novotny HR, Alvis DL: A method of choroidal circulations. Invest Ophthalmol neovascularization. Jpn J Ophthalmol
photographing fluorescence in circulating 1973; 12:248. 1989; 33:57.
blood of the human eye. Am J Ophthalmol 24. Patz A, Flower RW, Klein ML, et al: Clinical 41. Prünte P, Niesel P: Quantification of
1960; 50:176. application of indocyanine green choroidal blood-flow parameters using
3. Maumenee AE: Fluorescein angiography in angiography. Doc Ophthalmol Proc Ser indocyanine green video-fluorescence
the diagnosis and treatment of lesions of 1976; 9:245. angiography and statistical picture analysis.
the ocular fundus. (The 1968 Doyne 25. Orth DH, Patz A, Flower RW: Potential Ophthalmologica 1988; 226:55.
Lecture.) Trans Ophthalmol Soc UK 1968; clinical applications of indocyanine green 42. Bischoff P: Quantitative Untersuchung der
88:529. choroidal angiography. Preliminary report. normalen Aderhautzirkulation. Fortschr
4. Hyvärinen L, Maumenee AE, George T, Eye Ear Nose Throat Monthly 1976; 55:15. Ophthalmol 1989; 86:107.
et al: Fluorescein angiography of the 26. Craandijk A, Van Beek CA: Indocyanine 43. Flower RW, Klein GJ: Pulsatile flow in the
choriocapillaris. Am J Ophthalmol 1969; green fluorescence angiography of the choroidal circulation: a preliminary
67:653. choroid. Br J Ophthalmol 1976; 60:377. investigation. Eye 1990; 4:310.
5. Archer D, Krill AF, Newell FW: Fluorescein 27. Chopdar A, Turk AM, Hill DW: Fluorescent 44. Klein GJ, Baumgartner R, Flower RW:
studies of normal choroidal circulation. Am infra-red angiography of the fundus oculi An image processing approach to

CHAPTER 129
J Ophthalmol 1970; 69:543. using indocyanine green dye. Trans characterizing choroidal blood flow.
6. Archer DB, Krill AE, Ernest JT: Choroidal Ophthalmol Soc UK 1978; 98:142. Invest Ophthalmol Vis Sci 1990; 31:629.
vascular aspects of degenerations of the 28. Forsius H, Hyvärinen L, Nieminen H, et al: 45. Scheider A, Schroedel C: High resolution
retinal pigment epithelium. Trans Fluorescein and indocyanine green indocyanine green angiography with
Ophthalmol Soc UK 1972; 92:187. fluorescence angiography in study of scanning laser ophthalmoscope.
7. Wald G: The photochemistry of vision. Doc affected males and in female carriers with Am J Ophthalmol 1989; 108:458.
Ophthalmol 1949; 3:94. choroideremia. A preliminary report. Acta 46. Scheider A, Kaboth A, Neuhauser L:
8. Geeraets WJ, Williams RC, Chang G, et al: Ophthalmol 1977; 55:459. Detection of subretinal neovascular
The loss of light energy in retina and 29. Bonnet M, Habozit F, Magnard G: Valeur de membranes with indocyanine green and
choroid. Arch Ophthalmol 1960; 64:606. l’angiographie en infra-rouge au vert infrared scanning laser ophthalmoscope.
9. Behrendt T, Wilson LA: Spectral reflectance d’indocyanine dans le diagnostic clinique Am J Ophthalmol 1992; 113:45.
photography of the retina. Am J des angiomes de la choroide. Bull Soc 47. Guyer DR, Puliafito CA, Monés JM, et al:
Ophthalmol 1965; 59:1079. Ophthalmol Fr 1976; 76:713. Indocyanine-green digital angiography and
10. Geeraets WJ, Berry ER: Ocular spectral 30. Bacin F, Buffet JM: Un diagnostic choroidal neovascularization.
characteristics as related to hazards from difficile: L’angiome choroidien isole. Ophthalmology 1992; 92:287.
lasers and other light sources. Am J J Fr Ophthalmol 1978; 1:197. 48. Yannuzzi LA, Slakter JS, Sorenson J, et al:
Ophthalmol 1968; 66:15. 31. Saari M: Disciform detachment of the Digital indocyanine green videoangiography
11. Bloome MA: Fluorescein angiography: macula. II. Fluorescein and indocyanine and choroidal neovascularization. Retina
risks. Vision Res 1980; 20:1083. green fluorescence angiographic findings in 1992; 12:191.
12. Flower RW: Choroidal angiography using juvenile haemorrhagic macular 49. Spaide RF, Orlock DA, Herrmann-
indocyanine green: a review and progress choroidopathy. Acta Opthalmol 1977; Delamazure B, et al: Wide-angle
report. Opthalmol Dig 1974; 36:18. 55:530. indocyanine green angiography. Retina
13. Ring HG, Fujino T: Observations on the 32. Flower RW: Simple adaptors for fast 1998; 18:44.
anatomy and pathology of the choroidal conversion of a fundus camera for rapid- 50. Spaide RF, Orlock DA, Yannuzzi LA, et al:
vasculature. Arch Ophthalmol 1967; sequence ICG fluorescence choroidal Digital subtraction indocyanine green
78:431. angiography. J Biol Photogr Assoc 1977; angiography of occult choroidal
14. Friedman E, Kopald HH, Smith TR, et al: 45:43. neovascularization. Ophthalmology 1998;
Retinal and choroidal blood flow 33. Flower RW: Choroidal fluorescent dye filling 105:680.
determined with krypton-85 anesthetized patterns. A comparison of high-speed 51. Fox IJ, Wodd EH: Applications of dilution
animals. Invest Ophthalmol 1964; 3:539. indocyanine green and fluorescein curves recorded from the right side of the
15. Kogure K, Choromokos E: Infrared angiograms. Int Ophthalmol 1980; 2:143. heart or venous circulation with the aid of a
absorption angiography. J Appl Physiol 34. Hayashi K, Nakase Y, Nishiyama A, et al: new indicator dye. Proc Mayo Clin 1957;
1969; 26:154. Indocyanine green fluorescence 32:541.
16. Collela ME, Pilkerton AR: Determination of angiography, report 2: studies of new 52. Paumgartner G: The handling of
choroidal blood flow in man. Invest interference filters. Acta Soc Ophthalmol indocyanine green by the liver. Schweiz
Ophthalmol 1969; 8:460. Jpn 1982; 86:1532. Med Wochenschr 1975; 105(17 Suppl):1.
17. Kogure K, David NJ, Yamanouchi U, et al: 35. Hyvärinen L, Flower RW: Indocyanine green 53. Cherrick GR, Stein SW, Leevy CM, et al:
Infrared absorption angiography of the fluorescence angiography. Acta Ophthalmol Indocyanine green: observations on its
fundus circulation. Arch Ophthalmol 1970; 1980; 58:528. physical properties, plasma decay, and
83:209. 36. Bischoff PM, Flower RW: Ten years’ hepatic extraction. J Clin Invest 1960;
18. David NJ: Infrared absorption fundus experience with choroidal angiography 39:592.
angiography. In: Proceedings international using indocyanine green dye: a new routine 54. Baker KJ: Binding of sulfobromophthalein
symposium fluorescein angiography, Albi examination or an epilogue? Doc (BSP) sodium and indocyanine green (ICG)
1969. Basel: Karger; 1971:189. Ophthalmol 1985; 60:235. by plasma a1-lipoproteins. Proc Soc Exp
19. Hochheimer BF: Angiography of the retina 37. Hayashi K, Hasegawa Y, Tokoro T: Biol Med 1966; 122:957.
with indocyanine green. Arch Ophthalmol Indocyanine green angiography of central 55. Leevy CM, Bender J, Silverberg M, et al:
1971; 86:564. serous chorioretinopathy. Int Ophthalmol Physiology of dye extraction by the liver:
20. Flower RW, Hochheimer BF: Clinical 1986; 9:37. comparative studies of
infrared absorption angiography of the 38. Hayashi K, Hasegawa Y, Tokoro T, et al: sulfobromophthalein and indocyanine
choroid. Am J Ophthalmol 1972; 73:458. Value of indocyanine green angiography in green. Ann N Y Acad Sci 1963; 111:161.
21. Flower RW: Infrared absorption the diagnosis of occult choroidal 56. Goresky CA: Initial distribution and rate of
angiography of the choroid and some neovascular membrane. Jpn J Clin uptake of sulfobromophthalein in the liver.
observations on the effects of high Ophthalmol 1988; 42:827. Am J Physiol 1964; 207:13.
intraocular pressures. Am J Ophthalmol 39. Destro M, Puliafito CA: Indocyanine green 57. Wheeler HO, Cranston WI, Mettzer JI:
1972; 74:600. videoangiography of choroidal Hepatic uptake and biliary excretion of
22. Brown N, Strong R: Infrared fundus neovascularization. Ophthalmology 1989; indocyanine green in the dog. Proc Soc
angiography. Br J Ophthalmol 1973; 57:797. 96:846. Exp Biol Med 1958; 99:11.
1721
RETINA AND VITREOUS

58. Ketterer SG, Wiegand BD: Hepatic indocyanine green and its use in estimation 96. Schwartz SG, Guyer DR, Yannuzzi LA, et al:
clearance of indocyanine green. Clin Res of hepatic blood flow in dogs. Am J Physiol Indocyanine green videoangiography
1959; 7:289. 1960; 199:481. guided laser photocoagulation of primary
59. Ketterer SG, Wiegand BD: The excretion of 78. Ansari A, Lambrecht RM, Packer S, et al: occult choroidal neovascularization in
indocyanine green and its use in the Note on the distribution of iodine age-related macular degeneration. Invest
estimation of hepatic blood flow. Clin Res 123-labeled indocyanine green in the eye. Ophthalmol Vis Sci 1995; 36:186.
1959; 7:71. XVIII. Invest Ophthalmol 1975; 14:780. 97. Slakter JS, Yannuzzi LA, Sorenson JA,
60. Probst P, Paumgartner G, Caucig H, et al: 79. Flower RW: Injection technique for et al: A pilot study of indocyanine green
Studies on clearance and placental transfer indocyanine green and sodium fluorescein videoangiography-guided laser
of indocyanine green during labor. Clin dye angiography of the eye. Invest photocoagulation of occult choroidal
Chim Acta 1970; 29:157. Ophthalmol 1973; 12:881. neovascularization in age-related macular
61. Fox IJ, Wodd EH: Indocyanine green: 80. American National Standards Institute degeneration. Arch Ophthalmol 1994;
physical and physiologic properties. Mayo (ANSI): Safe use of lasers, Standard 112:465.
Clin Proc 1960; 35:732. Z-136.1. New York: ANSI; 1976. 98. Regillo SD, Benson WE, Maguire JI, et al:
62. Warner GS: Laboratory animal toxicity 81. Shimizu K, Ujiie K, eds: The structure of Indocyanine green angiography and occult
studies on cardiogreen. Documentation of the ocular vessels. Tokyo, New York: choroidal neovascularization.
Hynson. Baltimore: Wescott & Dunning; 1971. Ikagu-Shoin; 1978:51. Ophthalmology 1994; 101:280.
63. Tokoro T, Hayashi K, Muto M, et al: Studies 82. Bischoff PM, Speiser P: Angiographic 99. Guyer DR, Yannuzzi LA, Ladas ID, et al:
on choroidal circulation. Report I: study of the vortex vein circulation. Graefes Indocyanine green-guided laser
fundamental studies on the infrared Arch Clin Exp Ophthalmol 1986; 224:122. photocoagulation of focal spots at the
absorption angiography. Jpn J Ophthalmol 83. Potts AM: An hypothesis on macular edge of plaques of choroidal
SECTION 10

1976; 30:173. disease. Trans Am Acad Ophthalmol neovascularization. Arch Ophthalmol 1996;
64. Lutty GA: The acute intravenous toxicity of Otolaryngol 1966; 70:1058. 114:693.
biological stains, dyes, and other 84. Bischoff PM, Flower RW: High blood 100. Kuhn D, Meunier I, Soubrane G, Coca G:
fluorescent substances. Toxicol Appl pressure in choroidal arteries as a possible Imaging of chorioretinal anastomoses in
Pharmacol 1978; 44:225. pathogenetic mechanism in senile macular vascularized retinal pigment epithelium
65. Lutty GA: An intraperitoneal survey of degeneration. [Letter] Am J Ophthalmol detachments. Arch Ophthalmol 1995;
biological stains, dyes, and other 1983; 96:398. 113:1392.
fluorescent substances. Bull Nippon 85. Green WR, Key SN: Senile macular 101. Sorenson JA, Yannuzzi LA, Slakter JS,
Kanhoh-Shihiso Kenkyusho 1979; 50:25. degeneration. A histopathological study. et al: A pilot study of digital indocyanine
66. Leevy CM, Smith F, Kierman T: Liver Trans Am Ophthalmol Soc 1977; 75:180. green videoangiography for recurrent
function test. In: Bochus HL, ed. 86. Garner A: Vascular disorders. In: Garner A, occult choroidal neovascularization in
Gastroenterology. 3rd edn. Philadelphia: Klintworth GK, eds. Pathobiology of ocular age-related macular degeneration. Arch
WB Saunders; 1976:68. disease, part B. New York: Dekker; Ophthalmol 1994; 112:473.
67. Shabetai R, Adolph RJ: Principles of 1982:1479. 102. Hartnett ME, Weiter JJ, Gardts A, Jalkh AE:
cardiac catheterization. In: Fowler NO, ed. 87. Hayreh SS: Recent advances in fluorescein Classification of retinal pigment epithelial
Cardiac diagnosis and treatment. 3rd edn. fundus angiography. Br J Ophthalmol 1974; detachments associated with drusen.
Hagerstown, MD: Harper & Row; 1980:117. 58:391. Graefes Arch Clin Exp Ophthalmol 1992;
68. Carski TR, Staller BJ, Hepner G: Adverse 88. Hayashi K, de Laey JJ: Indocyanine green 230:11–19.
reactions after administration of angiography of choroidal neovascular 103. Yannuzzi LA, Negrao S, Iida T, et al: Retinal
indocyanine green. JAMA 1978; 240:635. membranes. Ophthalmologica 1985; angiomatous proliferation in age-related
69. Iseki K, Onoyama K, Fujimi S: Shock 190:30. macular degeneration. Retina 2001;
caused by indocyanine green dye in 89. François P, Turut P, Delannoy C: 21:416–434.
chronic hemodialysis patients. [Letter] Clin L’angiofluorographie choroidienne a 104. Kuhn D, Meunier I, Soubrane G, Coscas G:
Nephrol 1980; 14:210. l’indocyanine. Bull Soc Ophthalmol Fr Imaging of chorioretinal anastomoses in
70. Bacin F, Buffet JM, Mutel N: Angiographie 1977; 77:971. vascularized retinal pigment epithelium
par absorption, en infrarouge, au vert 90. Yannuzzi LA, Hope-Ross M, Slakter JS, detachments. Arch Ophthalmol 1995;
d’indocyanine. Aspects chez le sujet et al: Analysis of vascularized pigment 113:1392–1398.
normal et dans les tumeurs choroidiennes. epithelium detachments using indocyanine 105. Hartnett ME, Weiter JJ, Staurenghi G,
Bull Soc Ophthalmol Fr 1981; 81:315. green videoangiography. Retina 1994; Elsner AE: Deep retinal vascular anomalous
71. Hope-Ross M, Yannuzzi LA, Gragoudas 14:99. complexes in advanced age-related
ES, et al: Adverse reactions due to 91. Guyer DR, Yannuzzi LA, Slakter JS, et al: macular degeneration. Ophthalmology
indocyanine green. Ophthalmology 1994; Digital indocyanine-green 1996; 103:2042–2053.
101:529. videoangiography of occult choroidal 106. Slakter JS, Yannuzzi LA, Schneider U, et al:
72. Flower RW: High-speed human choroidal neovascularization. Ophthalmology 1994; Retinal choroidal anastomoses and occult
angiography using indocyanine green dye 101:1727. choroidal neovascularization in age-related
and a continuous light source. Doc 92. Guyer DR, Yannuzzi LA, Slakter JS, et al: macular degeneration. Ophthalmology
Ophthalmol Proc Ser 1976; 9:59. Classification of choroidal 2000; 107:742–754.
73. Flower RW, Hochheimer BF: Indocyanine neovascularization by digital indocyanine 107. Lafaut BA, Aisenbrey S, Broecke CV,
green dye fluorescence and infrared green videoangiography. Ophthalmology Bartz-Schmidt KU: Clinicopathological
absorption choroidal angiography 1996; 103:2054. correlation of deep retinal vascular
performed simultaneously with fluorescein 93. Chang B, Yannuzzi LA, Ladas ID, et al: anomalous complex in age-related macular
angiography. Johns Hopkins Med J 1976; Choroidal neovascularization in second degeneration. Br J Ophthalmol 2000;
138:33. eyes of patients with unilateral exudative 84:1269–1274.
74. Benson RC, Kues HA: Fluorescence age-related macular degeneration. 108. Fernandes LH, Freund KB, Yannuzzi LA,
properties of indocyanine green as related Ophthalmology 1995; 102:1380. et al: The nature of focal areas of
to angiography. Phys Med Biol 1978; 94. Chang TS, Freund KB, de la Cruz Z, et al: hyperfluorescence or hot spots imaged
23:159. Clinicopathologic correlation of choroidal with indocyanine green angiography. Retina
75. Puliafito CA, Destro M, To K, et al: Dye neovascularization demonstrated by 2002; 22:557–568
enhanced photocoagulation of choroidal indocyanine green angiography in a patient 109. Behrendt T: Therapeutic vascular
neovascularization. ARVO abstracts. Invest with retention of good vision for almost four occlusions in diabetic retinopathy. Arch
Ophthalmol Vis Sci 1988; 29(Suppl):414. years. Retina 1994; 14:114. Ophthal. 1972; 87:629–633.
76. Bill A: Capillary permeability to and 95. Guyer DR, Yannuzzi LA, Ladas ID, et al: 110. Shariga F, Ojima Y, Matsuo T, et al:
extravascular dynamics of myoglobin, Indocyanine green guided laser Feeder vessel photocoagulation of
albumin and gammaglobulin in the uvea. photocoagulation of focal spots at the subfoveal choroidal neovascularization
Acta Physiol Scand 1968; 73:204. edge of plaques of choroidal secondary to age-related macular
77. Ketterer SG, Wiegard BD, Rapaport E: neovascularization: a pilot study. Arch degeneration. Ophthalmology 1998;
1722 Hepatic uptake and biliary excretion of Ophthalmol 1996; 114:693. 105:662–669.
Indocyanine Green Videoangiography

111. Staurenghi G, Orzalesi N, La Capria A, 121. Spaide RF, Hall L, Haas A, et al: choriocapillaris blood flow in treatment of
Aschero M: Laser treatment of feeder Indocyanine green videoangiography of sub-foveal choroidal neovascularization
vessels in subfoveal choroidal neovascular older patients with central serous associated with age-related Macular
membranes: a revisitation using dynamic chorioretinopathy. Retina 1996; 16:203. degeneration. Am J Ophthalmol 2001;
indocyanine green angiography. 122. Yannuzzi LA, Ciardella AP, Spaide RF, et al: 132:85–93.
Ophthalmology 1998; 105:2297–2305. The expanding clinical spectrum of 132. Staugenghi G, Flower RW: Clinical
112. Bonnet M, Francoz-Taillanter N: idiopathic polypoidal choroidal observations supporting a theoretical
Hemangiomes caverneux de la choroide vasculopathy. Arch Ophthalmol 1997; model of choriocapillaris blood flow in
(revue clinique de 10 cas). Bull Soc 115:478. treatment of choroidal neovascularation
Ophthalmol Fr 1981; 81:455. 123. Ernest JT, Goldstick TK: Choroidal blood associated with age-related macular
113. Quentel G, Coscas G: Angiographie en flow measurement in the monkey by degeneration. Am J Ophthalmol 2002;
fluorescence infrarouge au vert clearance of indocyanine green dye. Exp 133:801–808.
d’indocyanine. Bull Soc Ophthalmol Fr Eye Res 1979; 29:7. 133. Flower RW: Optimizing treatment of
1984; 84:559. 124. Miszalok V: Video-Angiographie: Methode choroidal neovascularization feeder vessels
114. Bischoff P: Badeutung der und Nomenklatur. Klin Monatsbl associated with age-related macular
Infrarotangiographie fur die Augenheilkd 1987; 190:217. degeneration. Am J Ophthalmol 2002;
Differentialdiagnostik der Aderhauttumoren. 125. Flower RW, Hochheimer BF: Quantification 134:228–239.
Klin Monatsbl Augenheilkd 1985; 186:187. of indicator dye concentration in ocular 134. Reichel E, Puliafito CA, Duker JS, Guyer DR:
115. Krupsky S, Friedman E, Foster CS, et al: blood. Exp Eye Res 1977; 25:103. Indocyanine green dye-enhanced diode
Indocyanine green angiography in choroidal 126. Flower RW, Staurenghi G: Trataminento de laser photocoagulation of poorly defined

CHAPTER 129
diseases. Invest Ophthalmol Vis Sci 1992; los vasos nutrícios de la neovascularización subfoveal choroidal neovascularization.
33:723. coroidea. In: Monés J, Gómez Ulla, eds. Ophthalmic Surg 1994; 25:195–201.
116. Slakter JS, Giovannini A, Yannuzzi LA, et al: Degeneración Macular Asociada a la Edad. 135. Obana A, Gohto Y, Nishiguchi K, et al:
Indocyanine green angiography of Barcelona: Prous Science; 2005. A retrospective pilot study of indocyanine
multifocal choroiditis. Ophthalmology 1997; 127. Guyer DR, Yannuzzi LA, Slakter JS, et al: green enhanced diode laser
104:1813. Digital indocyanine green videoangiography photocoagulation for subfoveal choroidal
117. Howe LJ, Woon H, Graham EM, et al: of central serous chorioretinopathy. Arch neovascularization associated with
Choroidal hypoperfusion in acute multifocal Ophthalmol 1994; 112:1057. age-related macular degeneration.
posterior placoid pigment epitheliopathy. 128. Spaide RF, Hall L, Haas A, et al: Jpn J Ophthalmol 2000; 44:668–676.
An indocyanine green angiography study. Indocyanine green videoangiography of 136. Flower RW: Extraction of choriocapillaris
Ophthalmology 1995; 102:790. older patients with central serous hemodynamic data from ICG fluorescence
118. Park D, Schatz H, McDonald HR, et al: chorioretinopathy. Retina 1996; 16:203. angiograms. Invest Ophthalmol Vis Sci
Indocyanine green angiography of acute 129. Spaide RF, Campeas L, Hass A, et al: 1993; 34:2720–2729
multifocal posterior placoid pigment Central serous chorioretinopathy in younger 137. Flower RW: Variability in choriocapillaris
epitheliopathy. Ophthalmology 1995; and older adults. Ophthalmology 1996; blood flow distribution. Invest Ophthalmol
102:1877. 103:2070. Vis Sci 1995; 36:1247–1258.
119. Ie D, Glaser BM, Murphy RP, et al: 130. Flower RW: Experimental studies of
Indocyanine green angiography in multiple indocyanine green dye enhanced
evanescent white dot syndrome. Am J photocoagulation of choroidal
Ophthalmol 1994; 117:7. neovascularization feeder vessels. Am J
120. Yannuzzi LA, Sorenson JS, Spaide RF, et Ophthalmol 2000; 129:501–512.
al: Idiopathic polypoidal choroidal 131. Flower RW, von Kerczek C, Zhu L, et al:
vasculopathy. Retina 1990; 10:1. A theoretical investigation of the role of

1723
CHAPTER

130 Optical Coherence Tomography


Michael M. Altaweel and David L. Johnson

INTRODUCTION OCT is also useful in other medical specialties, including


otolaryngology, cardiology, gastroenterology, and vascular
Optical coherence tomography (OCT) allows noninvasive real- surgery.3-7 Conventional imaging modalities such as magnetic
time cross-sectional imaging of living tissues by measuring resonance imaging (MRI), computerized tomography (CT), or
their optical properties (see Fig. 130.1). The use of OCT in ultrasound have maximum resolutions that are 1/10th to
posterior segment imaging was first reported by Huang et al in 1/100th that of commercially available OCT systems. OCT
1991.1 Since, it became readily available for clinical use in the affords unprecedented in vivo resolution of biological tissues.
mid-1990s, OCT has revolutionized the way ophthalmologists The purpose of this chapter is to introduce the technology of
diagnose and manage certain eye conditions. This is in large OCT imaging, more specifically retinal imaging. It will also
part due to its technical ease of use, noninvasive nature, and the provide a framework for interpretation of normal images and
incredible amount of information it provides, particularly those of certain pathologic conditions. The examples will
regarding the condition of the retina and its relationship to highlight how OCT has improved our diagnostic ability, added
surrounding structures. It has also gained greater use as an to our understanding the pathogenesis of retinal disorders, and
imaging modality for glaucoma and for anterior segment influenced their management. The role of OCT in clinical trials
evaluation and management.2 will be discussed. OCT imaging of multiple retinal disorders
will also be found in other clinical chapters. OCT imaging for
the anterior segment and glaucoma will be discussed in other
portions of this book.

Low-Coherence TECHNOLOGY
Light Source

Reference The globe lends itself well to be studied using OCT because of
Fiber-Optic Mirror its optical accessibility with relatively transparent tissues.11
Interferometer Posterior segment OCT utilizes light in the 800–820 nm
Detector Transverse wavelength generated from a superluminescent diode. Anterior
Scanning
segment OCT utilizes a longer wavelength of 1300 nm in order
Electronics Computer to permit deeper penetration of denser biologic tissues such as
Condensing the sclera. OCT has limited clinical usefulness in tissue depths
Lens
greater than 2–3 mm. Because of the extremely high detection
Beamsplitter
sensitivity of the photodetector, <1 mW of power is required to
Slit-lamp obtain adequate images. This is well within safe acceptable
Viewing
Objective limits.
‘Tomography’ refers to the fact that OCT forms a two-
FIGURE 130.1. OCT performs cross-sectional imaging of structures dimensional cross-sectional image of the retina based on a
within the eye, allowing a noninvasive ‘optical biopsy’ with resolutions series of axial or longitudinal scans and measurements taken by
nearing that of histopathology. OCT is similar in principle to ultrasound
the machine. With each scan pass of the retina, anywhere from
but uses light rather than sound to generate images. For posterior
segment imaging, light is generated from a superluminescent diode. 128 to 512 axial range samples are captured. Each range sample
This light is split into a reference beam and a measurement beam by a is analogous in many ways to conventional A-scans. Each of
partially reflecting mirror. A portion of the measurement beam light is these scans is composed of 1024 data points which are 1–2 mm
backscattered or reflected back from different microstructural features in depth. In order to construct a tomographic image, these data
in the posterior segment. The reference beam is reflected back from points are integrated and transformed into an image displayed
the mirror which is at a known distance. The two light waves can in a meaningful way on the computer screen.12
combine and interfere, producing an output which is processed and Commercially available instrument such as the Stratus
quantified by the photodetector within the machine.8–10 The technique OCT-3 (Carl Zeiss Meditec Inc, Dublin, CA) have axial reso-
of low-coherence interferometry is utilized to allow measurement.8,9
lutions of 8 mm and transverse resolution of 20 mm. Machines
The low-coherence light utilized by the OCT only causes interference
when the reference beam echo delay equals the echo delay in the
used in research settings are capable of axial resolutions of
measurement beam. The interferometer therefore correlates the 2–3 mm13–15 These ultrahigh-resolution OCT machines provide
measurement light with the reference light that has a known delay to an unprecedented level of microstructural detail.16
allow measurement of the echo delay and amplitude of the The OCT unit consists of an acquisition module connected
backscattered or backreflected light. to a computer. Test results are displayed on a computer screen. 1725
RETINA AND VITREOUS

Currently available software packages have ~20 acquisition high-resolution cross-hair scan is often used to provide greater
modules or scanning modes. Modes vary according to scan detail for assessment of morphological changes in the retina
patterns, resolution of microstructure, acquisition times, and and adjacent structures.
target tissue of interest. Automated analysis protocols facilitate
interpretation of data. Quantitative data regarding retinal Key Features of OCT
thickness and qualitative information regarding morphology are • Non-invasive
obtained from the evaluation of an OCT scan. Longitudinal • Cross-sectional image of retinal architecture
follow-up is possible as data from prior scans is stored on the • Retinal thickness can be assessed and followed longitudinally
computer hard drive or an external storage device. • Provides new information on pathophysiology of retinal
disorders and response to therapy
OBTAINING IMAGES • Excellent for patient education

One of the chief advantages of OCT versus other retinal


imaging modalities is the noninvasive nature of the test. The INTERPRETATION OF NORMAL RETINAL
patient is placed into the acquisition module, which is similar
in design to a slit lamp. Mydriasis is helpful but not always
ANATOMY
necessary. The operator views in real time an infrared video The ability to image normal morphology and structural abnor-
image of the fundus displayed on the computer monitor to malities with OCT is based upon the fact that biologic tissues
SECTION 10

ensure proper alignment. The patient views a fixation light. As of differing densities (i.e., normal retina, RPE, choroid, vitreous,
the scanning light used is near infrared, the patient rarely hemorrhage, edema, drusen, etc) have different backscattering
perceives it during the scan and therefore experiences minimal properties.12 The OCT scan of a normal retina demonstrates
or no discomfort. characteristic features, including a distinct ability to distinguish
A sequence of axial measurements at various transverse between the retina and the vitreous, the retina and the RPE, and
positions is obtained in order to form a tomographic or two- between the RPE and the choroid. The foveal depression is well
dimensional representation of the retina. The faster the image demonstrated. Details of retinal layers have been progressively
acquisition time, the lower the pixel density or transverse better visualized with more recent versions of the OCT.
resolution will be. Conversely, the opposite is true. Commer- Displayed in Figure 130.2a is a cross-sectional image of the
cially available OCT instruments obtain scans at ~400 axial posterior pole in a normal eye obtained on the Stratus OCT-III
scans per second. In order to obtain higher-resolution scans machine. By convention the vitreous cavity is superior and the
with 512 transverse pixels, 1.28 s are required (T = 512/400) external layers of the retina and choroid are inferior. Figure
per scan line. Lower resolution scan lines with a transverse pixel 130.2b is an ultrahigh-resolution scan of a normal retina, which
density of 128 require only 0.32 s to acquire. These faster scans demonstrates much more distinction between structural
sacrifice transverse resolution while decreasing the amount of elements within the retina.
motion artifact and required patient cooperation.12 As demonstrated above, the OCT image closely resembles
Typical macular scans are 6 mm in length but can be adjusted the histological anatomy of the retina. Delineation of cellular
by the operator. Depending upon the acquisition protocol, details with commercially available OCT technology is not yet
anywhere from 2 to 24 scan lines are performed. Scan patterns possible. However, it is possible to distinguish between cellular
can also be adjusted. The most commonly employed scanning and noncellular elements of the retina.12 Ultrahigh-resolution
patterns for macular pathology are the ‘fast macular’ scan and imaging technology delivers an even greater level of resolu-
high-resolution radial scan. Fast macular scans obtain six cross- tion.13,15 The interface between the posterior hyaloid and the
sectional scans 30° apart, with each passing through the center internal limiting membrane (ILM) can usually only be
of the macula. The fast macular scan has a transverse reso- visualized if the hyaloid is elevated off the surface of the retina
lution of 128 pixels and is obtained in 2.5 s. The radial line scan (Fig. 130.7).10 The fovea is displayed with its normal thinning,
has a transverse resolution of 512 pixels, thus providing a much which is due to the absence of the inner retinal layers and the
higher level of detail, but requires much more time to obtain presence of only a thickened photoreceptor layer and the outer
and repeated efforts at fixation. Macular thickness and gross nuclear layer.
abnormalities can be sufficiently interpreted using the fast A false color coding system ascribed by the image processing
macular protocol12 and the results of testing are more reliable algorithm is used to distinguish between the different micro-
in situations where fixation is difficult for the patient.17 The structural layers of the retina. The reflectivity of different

Nerve Fiber Layer FIGURE 130.2. (a) Normal retinal anatomy.


Ganglion Cell Layer Notice how the NFL increases in thickness
Inner Plexiform Layer nasally toward the optic nerve (right side of
Inner Nuclear Layer image). Cellular layers, such as the ganglion cell
Outer Plexicom Layer
Outer Nuclear Layer layers and the inner and outer nuclear layers
Inner/Outer Photoreceptor Junction have less intense backscattering due to the
Retinal Pigment density and orientation of their respective
a Epithelium/Choriocapillaris complex elements. Layers with more intense reflectivity
include the RPE/Choriocapillaris, the junction
between the inner and outer segments of the
photoreceptor layer, the inner and outer
plexiform layers, as well as the NFL. Axial
resolution is 8 mm. (b) Ultrahigh-resolution OCT
of a normal retina. Axial resolution is 2 mm.
Notice the greater ability to identify retinal
structures/layers.
Photo courtesy of Dr JS Duker.
b
1726
Optical Coherence Tomography

a b

CHAPTER 130
FIGURE 130.3. (a) The retinal thickness map is derived from six radial scans oriented 30° apart, all passing through the center of the macula.
(b) Retinal thickness map: the thickness for each of the nine macular subfields is displayed in both numerical as well as color coded formats.
Central point thickness in this patient is 181 mm, and the standard deviation is 5 mm. The displayed scan is oriented from inferior to superior.

retinal layers is based upon the arrangement of their structures NFL. The posterior surface of the retina is typically defined by
as well as their biological densities and degree of pigmentation. the high reflectivity of the junction between retinal tissue and
Those structures with high biological reflectivity are repre- the RPE/choriocapillaris complex.12 Normal retinal thickness is
sented by red, medium reflectivity with yellow/green, and those 182 mm for the OCT-III.18 Determinations of retinal thickness
with low reflectivity are blue. Absence of a reflectivity signal is are performed automatically and can be the source of erroneous
represented by black. The nerve fiber layer (NFL) and plexiform measurements.
layers are highly reflective due to the horizontally oriented
axonal structures. The NFL increases in thickness and DISTINGUISHING ARTIFACTS
reflectivity close to the optic nerve. The nuclear layers are less
reflective and appear blue to black on cross-sectional images. In order to interpret OCT and determine whether the quanti-
The RPE and choriocapillaris form the outermost highly tative information is accurate, one must assess the quality of an
reflective structure visible due to their high melanin and vas- OCT scan and recognize OCT artifacts. Fortunately, obtaining
cular content respectively. These two layers cannot usually be adequate OCT images is a quick process. Most artifacts are
distinguished on conventional OCT imaging because of their derived from difficulty with patient cooperation or abnor-
close proximity to one another. External to the choriocapillaris malities within the ocular media rather than operator error.19
are the larger sized choroidal vessels. These vessels, as well as Mydriasis is not always necessary to obtain OCT but may
deeper structures, cannot usually be seen on OCT due to the provide a higher signal-to-noise ratio and consequently better
shadowing from the overlying RPE and choriocapillaris.10 microstructural details. Dilation also decreases the amount of
Scanned images have a legend that indicates the direc- ‘vignetting’ which is described below. Obtaining quality OCT
tionality of the currently displayed scan, as demonstrated in images depends upon the light being able to reach the target
Figure 130.3b. The scan direction is proceeded from inferior to tissue. If there is significant media opacification such as corneal
superior in the scan displayed. By convention, the right side of opacification or severe cataract there will be less light reaching
the image corresponds to the arrowhead. OCT quantitative the posterior segment and less light returning to the infero-
results are generally represented in a retinal map. The macula meter for detection. This ‘signal-to-noise ratio’ is displayed on
is artificially divided into nine regions and the average retinal the screen and labeled as ‘signal strength’. A signal strength of
thickness is calculated for each region. The inner circle of the 10 represents the highest quality imaging whereas a signal
map has a diameter of 1.0 mm and correlates roughly with the strength of zero represents the lowest quality. If there is little
fovea. The middle circle has a diameter of 3.0 mm and the outer light reaching the tissue, the images appear dull or ‘washed out’
circle a diameter of 6.0 mm, which is the length of the axial and it is difficult to detect subtle abnormalities and differences
scans. A color coded map along with a legend is displayed in in tissue reflectivity. Figure 130.4 shows an example of low
order to facilitate rapid interpretation of numerical values. signal strength. Notice how the ordinarily highly reflective
Greater retinal thickness is represented by the ‘hotter’ colors structures appear blue or even black. Moderate media opacities
such as red and white. Average retinal thickness is represented such as early cataract, asteroid hyalosis, and mild vitreous
by green, and thin or atrophic areas of retina are represented by hemorrhage do not preclude adequate imaging.20,21 OCT can be
the ‘cooler’ colors such as blue or black. Central retinal or foveal performed through silicone oil, but not through intraocular gas.
thickness and total volume of the macula are displayed in Another artifact that must be recognized is that of ‘vignetting’.
numerical format. The standard deviation of the center point is Vignetting occurs as the light beam is obscured by part of the
determined by comparing the six points which cross through iris, causing less light to reach the retina in that area.12 The find-
the center in the scanning protocol and is recorded with the ings are quite characteristic and easily recognized (Fig. 130.5).
center point. Poor patient cooperation can lead to abnormalities on the
Image processing algorithms allow automated measurement OCT image. Image processing software corrects for a certain
of retinal thickness. This is accomplished by first determining amount of longitudinal movement by the patient but there is
the anterior and posterior surfaces of the retina. The anterior little to no software correction for transverse movement or poor
surface of the retina is defined by the transition from the low patient fixation. This results in reduced image quality and
reflectivity of the vitreous cavity to the high reflectivity of the transverse resolution. This is particularly a problem in patients 1727
RETINA AND VITREOUS

interface and subsequent measurement data.22 This tool is


being developed for future versions of the OCT.

ENSURING HIGH-QUALITY OCT DATA


1. All six underlying scans present for retinal thickness map
2. ILM and RPE borders drawn correctly
3. Standard deviation of the center point <10% (of center-
point value)
4. Adequate signal strength (5 or more)
5. Macula centered in the scan

CLINICAL CONDITIONS
OCT has improved our understanding of the pathophysiology
of retinal disorders, our ability to distinguish between differen-
tial diagnoses with a similar clinical appearance, our ability to
FIGURE 130.4. Example of low ‘signal-to-noise ratio’. Notice how the follow conditions in a noninvasive manner, our ability to make
SECTION 10

entire image appears ‘washed out’. It is difficult to discern details of treatment decisions, and our ability to educate our patients.
the retinal microstructure. The signal strength is relatively low at three. The following examples demonstrate the utility of OCT and
illustrate commonly identified abnormalities.

VITREORETINAL INTERFACE ABNORMALITIES


Posterior Vitreous Detachment
Posterior vitreous detachment (PVD) is a common pheno-
menon, observed in 63% of eyes over the age of 70. It results
from syneresis of the vitreous gel. When the liquefied vitreous
obtains access to the retro-hyaloid space through a defect in the
posterior hyaloid face, PVD can occur.23–25
FIGURE 130.5. OCT image of a normal macula. The right side of the PVD often shows up on OCT as a thin, faint hyper reflective
image displays a common artifact known as vignetting. This occurs line lying above the surface of the retina. In order for the
when the iris blocks a portion of the light. This may occur with poor posterior hyaloid to be visualized on OCT the hyaloid must be
pupil dilation. within 100–200 mm of the surface of the retina.12,26 Figure
130.7, shows an example of a PVD.

Vitreomacular Traction Syndrome


Vitreomacular traction syndrome (VMT) results from an
abnormally firm adherence of the posterior hyaloid to the
macular region with anteroposterior and tangential traction
causing distortion of the macular architecture and retinal
thickening.27,28 Vision is frequently decreased secondary to the
retinal edema as well as the abnormal macular contour. The
condition may be difficult to appreciate on clinical exam, but is
clearly demonstrated by OCT imaging.29–36
Figure 130.8 demonstrates a case of VMT traction. This is a
series of OCT images taken from the same patient over the course
of 13 months. Notice the thin band representing the posterior
hyaloid which is inserting onto the surface of the retina. The
retina displays cystic changes and there can be subretinal and
FIGURE 130.6. Misdrawn borders results in erroneous measurement
in a portion of the retinal thickness map.

with poor visual acuity. Retinal thickness data is often less


reliable in such situations. This is best demonstrated as a large
standard deviation in the center-point thickness measurement.
The problem is partially alleviated by utilizing the fast map
protocol rather than the high-resolution scanning protocol for
such patients.17
The automated algorithm, which determines and draws the
anterior and posterior borders of the retina to allow retinal
thickness measurements, can be erroneous. This is noted as
misdrawn borders on the retinal thickness map (Fig. 130.6). If
this occurs, the data generated by the OCT would not be FIGURE 130.7. PVD. Notice the thin hyperreflective band just above
correct. For a prior version of the OCT (OCT II), a graphical the surface of the retina representing the posterior vitreous cortex, or
1728 user interface was developed which allowed correction of this hyaloid.
Optical Coherence Tomography

a a

CHAPTER 130
b b

FIGURE 130.9. (a) demonstrates an epiretinal membrane causing


distortion of macular anatomy with loss of the foveal depression and
thickening of the retina. The epiretinal membrane is easily identified
as the highly reflective structure lying on the surface of the retina.
(b) Four months after membrane peeling, the patient’s macular
anatomy has returned to near normal with return of a foveal
depression. One can appreciate the extent of the epiretinal membrane
peeling by noticing the residual membrane on the right side of the
image.

FIGURE 130.8. (a–c) This series of figures demonstrates a case of


VMT that progressed over the course of 13 months. The patient
complained of increasingly distorted and decreased vision.

sub-RPE fluid as well. There is obvious distortion of normal


foveal anatomy in the later images.
OCT is very useful in understanding and planning the
management for VMT and provides an excellent tool for patient
education. These patients can clearly understand the rationale for
proceeding with a vitrectomy and release of adhesions as their
condition deteriorates. The OCT is equally useful in assessing FIGURE 130.10. Macular pseudohole associated with an epiretinal
management outcomes. membrane. Notice the highly reflective membrane lying just above the
surface of the retina. One can appreciate that this membrane has
Epiretinal Membrane caused an irregular retinal surface as well as intraretinal edema. There
is a layer of photoreceptors at the base of the pseudohole.
Epiretinal membranes are caused by glial proliferation and con-
tracture on the surface of the retina. This can lead to abnormal
macular and vascular architecture. Fluorescein angiography brane causing decreased vision with images of the same patient
frequently will demonstrate vascular leakage.27,37–41 after epiretinal membrane peeling.
On OCT, epiretinal membranes are manifested by an abnor- Epiretinal membranes can lead to the appearance of a
mal highly reflective structure lying on the surface of the retina. ‘pseudohole’. OCT can be useful in distinguishing a ‘pseu-
The membrane is more easily distinguished if there is some dohole’ from a full thickness macular hole, which can be a
separation between the membrane and the surface of the retina. difficult clinical distinction. Figure 130.10 demonstrates a severe
If there is no separation one may be able to use indirect clues to ERM with pseudohole resulting in the appearance of a
appreciate the presence of the membrane such as surface con- steepened foveal contour, intraretinal edema, and separation of
tracture and macular edema. the outer nuclear layer and the outer plexiform layer. In contrast
OCT is helpful in confirming the diagnosis as well as quanti- to a macular hole, the photoreceptor layer remains intact.42–45
fying the amount of macular distortion present. It is often quite In providing patient education regarding their ERM, the OCT
useful in following the patients in the pre- and postoperative is invaluable, particularly since the abnormalities may be subtle
stages. Figure 130.9 demonstrates a case of epiretinal mem- on fundus photographs. 1729
RETINA AND VITREOUS

a
SECTION 10

FIGURE 130.11. (a) Demonstrates an impending macular hole with a


foveolar detachment. Notice the thin posterior hyaloid band above the
surface of the retina inserting onto the fovea. (b) Three months later
the posterior hyaloid completely detached and the stage I hole
spontaneously resolved. Notice once again the separated posterior
hyaloid lying above the surface of the retina.

Idiopathic Macular Holes


Macular holes are full thickness defects in the neurosensory
retina caused by abnormal tangential or oblique vitreous trac-
tion on the foveal region. OCT assists in the diagnosis of b
macular hole by clearly demonstrating the retinal defect and has
enhanced our understanding of the pathophysiology of hole FIGURE 130.12. (a and b) Stage IIA macular hole with a persistent
attachment of the posterior vitreous leading to traction. Stage IIB hole
formation.45–47 Such images are of great benefit in patient
with posterior vitreous separation may be more appropriate to treat
education regarding their disorder and proposed surgical with additional intervention such as ILM peel.
correction. Demonstration of the pathophysiology of various
stages of macular holes also assists the surgeon in making
treatment decisions, including whether surgery is required or OCT can also be useful in determining the prognosis for
not.48,49 Early stage holes may appear as a yellow dot or circle on patients with a macular hole. Figure 130.13a represents a full
biomicroscopic examination, with a long differential diagnoses. thickness stage IV macular hole. The rounded edges of the hole,
OCT can demonstrate the early stages of a hole and the cystic changes in the adjacent retina, and large width of the hole
tractional elements responsible for hole formation. Figure indicates a severe macular hole and may suggest features of
130.11 demonstrates a stage I impending macular hole. There chronicity. There is a higher percentage of nonclosure or lack of
is mild loss of visual acuity and mild metamorphopsia. A visual improvement despite hole closure with stage IV holes
foveolar detachment is present. 50% of stage I holes will than with smaller macular holes.61–64 The OCT is an excellent
spontaneously resolve.50,51 Therefore the OCT helped in tool in providing patient education regarding macular hole and
making the correct diagnosis and in making the decision to in demonstrating the anatomical success of surgical repair
observe the condition rather than operate. (Fig. 130.13b).
Surgical developments in the repair of macular hole have
included the increasing addition of ILM peeling in an effort to
improve hole closure rates.52–56 Controversy exists concerning VASCULAR ABNORMALITIES
whether final visual outcome is improved, and whether ILM Diabetic Macular Edema
peeling is necessary.57–59 For stage II macular holes, OCT may Macular edema is the leading cause of vision loss in diabetics.
be instrumental in directing the choice of surgical procedure. As It affects ~14–25% of patients who have had diabetes for 10
illustrated in Figure 130.12, stage II holes (Gass biomicroscopic years or more.65–69 OCT can be used to help detect and quantify
staging) may be distinguished as stage IIA if the posterior the amount of diabetic macular edema present, the proximity
hyaloid is still attached and causing traction on the edge of the and involvement of the foveal region by the edema, as well as
macular hole or stage IIB if such traction has already been the response to therapy.70,71 Subclinical macular edema may
released.60 It is postulated that a stage IIA hole may be less only be appreciated with OCT imaging. A National Eye Institute
likely to require an ILM peel to improve closure rates than a sponsored trial is assessing the utility of early intervention for
stage IIB hole. This would avoid the additional risk imposed by such patients (Subclinical Diabetic Macular Edema Study). A
1730 performing a membrane peel during surgery. number of clinical trials have recently incorporated retinal
Optical Coherence Tomography

aii

CHAPTER 130
ai b

FIGURE 130.13. (a) Represents a stage IV full thickness macular hole with a PVD (not visible). Notice the signs of severity such as the rounded
contour at the edge of the hole as well as the cystic changes within the retina adjacent to the hole. (b) Successful closure of this macular hole
after pars plana vitrectomy with ILM peeling and intraocular gas tamponade. There is a return to a relatively normal foveal anatomy.

a b

FIGURE 130.14. OCT and corresponding color fundus image of a patient with clinically significant diabetic macular edema. The transverse scan
demonstrates intraretinal edema collecting on the temporal side of the fovea. The topographical image shows the location and extent of the
retinal edema. Notice on the cross-sectional image the hard exudate immediately temporal to the fovea causing a mild shadowing effect.

thickness as measured by OCT as a secondary endpoint. Com- Figure 130.15 illustrates the utility of OCT in surgical
parisons with standardized visual acuity have found variable planning, with points of adhesion and traction evident.75 Pars-
correlation, however morphological responses to therapy are plana vitrectomy with membrane peeling resulted in an
well visualized with OCT.72–74 Retinal thickness parameters are improved macular anatomy.
utilized in making re-treatment decisions. In clinical practice,
the use of OCT to follow patients with diabetic macular edema Cystoid Macular Edema
has increased greatly, particularly for those managed with Visually significant cystoid macular edema (CME) is one of the
intravitreal steroid or anti-VEGF injections. most common reasons for suboptimal visual recovery after
Figure 130.14, shows the transverse image and macular uncomplicated cataract extraction, occurring in ~1–2% of
thickness assessment in a patient with clinically significant patients. Occurrence rates may be higher in complicated cases,
diabetic macular edema. aphakia, diabetics, and in patients receiving topical glaucoma
therapy with prostaglandin analogs.76,77 Onset is usually
Proliferative Diabetic Retinopathy 6–8 weeks after surgery.
Diabetes can lead to the development of extraretinal fibro- OCT is a noninvasive alternative to fluorescein angiography
vascular proliferation extending beyond the ILM into the in making the diagnosis and following CME. The amount of
vitreous. Partial PVD in these cases can lead to the development intraretinal edema can be quantified. The edema is often con-
of vitreous hemorrhage, macular heterotopia, and tractional tained within cystoid spaces in the outer plexiform layer. OCT
retinal detachment. also helps to determine if there is a component of vitreous 1731
RETINA AND VITREOUS

aii

ai b
SECTION 10

FIGURE 130.15. (a) Diabetic tractional detachment of the macula. There is a thick fibrovascular membrane lying on the surface of the macula
causing a tractional detachment with accumulation of a large amount of intraretinal edema. (b) Following pars plana vitrectomy with membrane
peeling there is a return to a more normal macular architecture. There is still a large amount of cystic intraretinal edema present.

ai aii

aiii b

FIGURE 130.16. (a) Patient with CME after uncomplicated extracapsular cataract extraction. Cystoid spaces are evident. There is no VMT.
1732 (b) After 2 months of treatment with topical nonsteroidal antiinflammatory drops, much of the edema has resolved.
Optical Coherence Tomography

CHAPTER 130
b

ai

FIGURE 130.17. Inferior branch vein occlusion. Notice the cystic


aii intraretinal edema on the cross-sectional image. The macular thickness
map confirms that most of the edema is inferior to the fovea.

traction contributing to the macular edema. Figure 130.16 OCT is useful for evaluating the severity of macular edema
reveals a case of CME after uncomplicated cataract extraction. associated with retinal vascular occlusion. It does not replace
OCT is particularly helpful in the diagnosis of CME associated fluorescein angiography in the initial diagnostic evaluation
with the therapeutic use of Niacin, as the fluorescein angiogram but compliments it and quantifies the degree of edema (Fig.
generally does not demonstrate leakage in these cases.78–80 130.17).81,82 It is very useful in following the natural history or
response to therapy in a noninvasive manner. It has been used
Branch Retinal Vein Occlusion to assist in management decisions and is a secondary endpoint
Branch retinal vein occlusions (BRVO) occur most commonly at in current therapeutic trials, including the standard care versus
arteriovenous crossings in the setting of systemic hypertension. intravitreal triamcinolone for retinal vein occlusion (SCORE)
The retinal artery can compress the vein within their common study. The standard of care for BRVO with secondary center
adventitial sheath, resulting in turbulent flow and thrombosis. involving macular edema has been observation for a period of 3
Systemic abnormalities such as vasculitis or coagulation months, followed by a treatment with grid laser in the area of
disorders may also lead to BRVO. The clinical picture is one of ischemia and leakage if the visual acuity was less than or equal
sudden painless vision loss due to ischemia, hemorrhage, or to 20/40.83 OCT can be helpful in determining if the macular
associated macular edema. edema is worsening before this point and whether intervention 1733
RETINA AND VITREOUS

ai
SECTION 10

FIGURE 130.18. (a) CRVO. Notice the large amount of intraretinal


edema manifest on the cross-sectional image and the macular thickness
map. The apparent concavity of the RPE/choriocapillaris band is an
artifact due to the automated image processing software. (b) Four weeks
aii after receiving 4 mg of intravitreal Triamcinolone, the patient’s macular
thickness decreased markedly.

may be required earlier. Conversely, it can also determine if the dilated and tortuous veins, optic disk edema, intraretinal
macular edema has been improving over the 3 months and hemorrhages, and retinal edema. Vision loss in CRVO may due
further observation might be reasonable. Intravitreal steroids and to ischemic insult or macular edema or both. OCT has been
anti-VEGF intravitreal agents have been utilized as alternative useful in evaluating the degree of retinal thickening, which is
treatments with the response to therapy generally evaluated by often very severe (center-point thickness greater than 500 mm is
a combination of visual acuity and retinal thickness on OCT. As common) (Fig. 130.18a). The Central Vein Occlusion Study
these agents have a temporary effect, repeat OCTs assist the found that grid laser reduced macular edema but did not result
clinician in determining the optimal timing for re-treatment. in improved visual acuity.85,86 Therefore the standard of care for
macular edema associated with this disorder has been under
Central Retinal Vein Occlusion observation. Among the many treatments more recently
Central retinal vein occlusion (CRVO) is due to occlusion of the attempted for CRVO, the most frequently utilized is the
central retinal vein at or posterior to the level of the lamina injection of intravitreal steroid or anti-VEGF agents. OCT
cribrosa. An atherosclerotic central retinal artery may impinge demonstrates well the effect of such medications on the
upon the vein, causing turbulence, endothelial damage, and macular edema (Fig. 130.20b) and can assist the clinician in
1734 thrombus formation.84 Clinical examination often reveals making treatment decisions.87,88 If the edema resolves but the
Optical Coherence Tomography

FIGURE 130.19. CSCR with accumulation of subretinal fluid. The FIGURE 130.21. Geographic atrophy associated with ARMD. There is
shadow effects lateral to the neurosensory detachment are a result of increased reflectivity of deeper structures centrally due to RPE
larger caliber native intraretinal blood vessels. atrophy beneath the fovea. Irregularities of the RPE/choriocapillaris
complex on the right side of the scan are representative of drusen.

CHAPTER 130
a

FIGURE 130.20. CSCR with a pigment epithelial detachment. The


pigment epithelial detachment was much more clearly visualized with
OCT than with fluorescein angiography.

visual acuity remains poor then the degree of ischemia probably


limits the visual potential and makes further injections less
valuable. Conversely, if the edema resolves well and visual
acuity improves, then repeating injections for future
recurrences of edema makes sense. In ongoing clinical trials,
retinal thickness measured by OCT is a secondary endpoint.

Central Serous Chorioretinopathy


Central serous chorioretinopathy (CSCR) results from localized
dysfunction of the retinal pigment epithelial cells, resulting in
accumulation of fluid in the subretinal space (Fig. 130.19).89,90,27
OCT is helpful in diagnosing the condition as well as following
the patients longitudinally. OCT may also demonstrate asso-
ciated lesions which may be more difficult to discern on
examination or with fluorescein angiography, such as pigment
epithelial detachment (Fig. 130.20). The OCT is very useful in
educating the patient regarding their condition, and in
b
demonstrating their progress and/or resolution over time.
FIGURE 130.22. OCT and corresponding fluorescein angiography of
Age-Related Macular Degeneration an occult choroidal neovascular membrane with a large amount of
Age-related macular degeneration (ARMD) is the leading cause intraretinal fluid present. There is disruption and elevation of the RPE.
of irreversible vision loss in the Western world. It affects ~10
million people in the United States with 200 000 new cases of
advanced ARMD diagnosed annually. The disease is divided Exudative ARMD
into two basic categories: nonexudative and exudative. Exudative ARMD lesions are classified according to fluorescein
angiography findings as either predominantly classic,
Nonexudative ARMD minimally classic, or occult with no classic features. OCT
Nonexudative ARMD can lead to changes in the macula such findings do not necessarily correlate with the fluorescein
as RPE hyper- and hypopigmentation as well as basal laminar classification. OCT demonstrates abnormalities in the retinal
and basal linear deposits. RPE hyperpigmentation can cause architecture in most cases of exudative AMD, with common
increased reflectivity of the external band on OCT while RPE findings of subretinal fluid, retinal thickening, pigment
atrophy results in hyperreflectivity of structures deep to the epithelial detachment, (Fig. 130.24) and choroidal neovasculari-
RPE/choriocapillaris (Fig. 130.21).91 Drusen will often produce zation (CNV).92 Occult CNV generally causes thickening of the
a stippling effect of the external band consistent with deposits RPE layer. The typical appearance with inability to distinguish
along Bruch’s membrane.92 the CNV from the RPE layer is termed type I (Fig. 130.22). This 1735
RETINA AND VITREOUS

FIGURE 130.23. OCT and corresponding fluorescein angiogram of a


classic CNV. Notice the discontinuity in the RPE/choriocapillaris band
with a thick hyperreflective neovascular membrane in the subretinal
space. There is accumulation of subretinal fluid as well as intraretinal
fluid.
SECTION 10

is by far more common. Classic CNV may have a similar


appearance, or in some cases may break through Bruch’s
membrane and grow more clearly in the subretinal space (Fig.
130.23). This is termed a type II CNV. From a therapeutic
perspective, this may influence the response to surgical therapy.
A type II CNV complex would be more likely to respond to
surgical removal without RPE damage, whereas removal of a
type I membrane would be much more likely to damage the
normal structure of the RPE and subsequently result in poor
visual acuity. This was demonstrated in the Submacular
Surgery Trial.93,94

OCT in the management of age related macular FIGURE 130.24. Serous Pigment Epithelial Detachment associated
degeneration with ARMD. Notice the excrescences on the surface of the RPE. There
is a small amount of subretinal fluid as well.
Traditional therapy for exudative macular degeneration has
consisted of either thermal laser photocoagulation or photo-
dynamic therapy (PDT). Intravitreal injections with anti-
vascular endothelial growth factor (anti-VEGF) compounds Data from a recent study using OCT to guide decisions
have demonstrated higher rates of vision retention and about re-treatment suggests that this is an efficient and effective
improvement and have increased greatly in use.95–98 way to follow patients with exudative ARMD. In this study,
OCT has been invaluable in helping retina specialists decide patients received on average five to six intravitreal injections of
when to re-treat their patients with exudative ARMD. an anti-VEGF compound over 1 year based on recurrence of
Fluorescein angiography is no longer needed at every follow up retinal thickening rather than the 12 consecutive monthly
visit to make a determination of whether to re-treat or not. injections required over a period of 1 year in the phase III
Typically fluorescein angiography is obtained along with OCT trials.99,100 The visual outcomes were similar with both dosing
at the initial visit in order to confirm the diagnosis and to regimens.
characterize the choroidal neovascular membrane. Once treat- Despite the utility of this imaging method in guiding
ment is initiated, particularly with an intravitreal injection of management, retinal thickness measured on OCT does not
an anti-VEGF agent, the patients can often be followed-up with always correlate with visual acuity. Resolution of retinal
serial OCT imaging and re-treated if intraretinal or subretinal thickening may occur with the formation of a disciform scar or
fluid persists or recurs. with the development of atrophy resulting in poor visual acuity.
Resolution of retinal thickening could also occur with a
favorable response to treatment resulting in stable or improved
Key Features: Utility of OCT in Retinal Disorders visual acuity. Therefore interpretation of the OCT data must be
• Supplements other imaging modalities in making diagnosis
correlated with the visual acuity and the findings on biomicros-
• Can discern and quantify more subtle changes in response to
copic examination.
therapy
• Non-invasive test-very helpful considering that some treatment
OCT in clinical trials
regimens require frequent intervention
The use of OCT measurements as secondary endpoints in
• Can assist in decision making process regarding retreatment
clinical trials has increased greatly in the last few years. The
1736 advantages of OCT include the noninvasive nature of the test,
Optical Coherence Tomography

the ease of use for photographers and subjects, the objective Grading morphology In addition to evaluating OCT quantita-
measurements of retinal thickness, and the additional infor- tive results, reading centers also assess morphological changes
mation regarding morphology of the retina and adjacent such as vitreoretinal interface abnormalities, intraretinal cysts,
structures. Quantitative results are usually reported by CNV, subretinal fluid, and PED. These features are graded for
comparing the mean change in retinal thickness or abnormal presence or absence and may be quantified (extent of cysts,
retinal thickening (total retinal thickness minus normal retinal thickness of subretinal fluid, thickness of CNV complex).
thickness) between the treatment groups and sham group. Correlations are performed to evaluate whether the presence of
Results may also be reported as the percentage of a group which such features at baseline are predictive of outcome, and whether
reach a prespecified goal, such as reduction of retinal thickness the features change over time, in response to therapy.
by 100 mm or reduction of abnormal thickening by 50%.
Longitudinal follow-up As a secondary endpoint OCT has
Eligibility determination OCT determined retinal thickness been very useful in assessing response to therapy for macular
criteria have been utilized in recent trials of ARMD, retinal vein disease. Retinal thickness measurements on OCT can be used
occlusion, and diabetic macular edema to ensure that subjects to make retreatment decisions. For example, in ongoing trials of
who are enrolled have sufficient center involving retinal intravitreal triamcinolone for the management of macular
thickening to evaluate if the therapy under investigation is edema associated with retinal vein occlusion and diabetic
efficacious. A common requirement is the presence of a center- retinopathy, comparisons of baseline and current OCT findings
point retinal thickness of at least 250 mm. In addition, OCT is are used to help determine whether treatment has been

CHAPTER 130
used to exclude patients who demonstrate vitreoretinal successful or not, and secondarily whether to continue
abnormalities which would preclude resolution of retinal treatment or not.
thickening with medical therapy.101
FUTURE TRENDS
OCT quality Several clinical trials have found that the
reliability and reproducibility of the measurements obtained by Several aspects of OCT are being improved upon at present.
OCT scanning are much higher if the OCT meets certain Ultrahigh-resolution OCT improves resolution from 8 to 2 mm,
quality criteria. All six underlying scans must be present for the allowing greater visualization of structural detail which may
retinal thickness map, the ILM and RPE borders must be drawn enhance further our understanding of the pathophysiology of
correctly, the standard deviation of the center point must be macular disorders.13–15
<10% (of center-point value), signal strength should be five or Spectral domain OCT also allows improved resolution,104
greater, and the macula must be centered in the scan. In a study with decreased acquisition time. The combination of OCT with
comparing OCT II and OCT III (current generation) simultaneous registration of retinal images with scanning laser
measurements for diabetic macular edema, OCT III ophthalmoscopy will allow improved reproducibility of measure-
measurements were found to be reliable in 91.5% of cases ments and ensure centration of measurements. 3 dimensional
compared with only 59.6% for the OCT II measurements. The representation of retinal morphology has become possible.105
advantage of the OCT III was the ability to obtain the Software developments including a graphical user interface to
measurements over 2.5 s compared with the requirement to allow correction of misdrawn borders will enhance our ability to
obtain six separate scans for the OCT II.102 This finding was obtain accurate measurements.
corroborated by a comparison of fast macular scans versus six
high-resolution scans to obtain retinal thickness data for an SUMMARY
exudative macular degeneration study. With 383 visits, 93.7%
(359/383) of fast macular thickness maps had a center-point OCT is a noninvasive test that provides clinicians and
standard deviation of less than or equal to 10% (good quality) researchers objective information on the presence and severity
versus 59.0% (226/383) of high-resolution macular thickness of retinal, RPE, and vitreoretinal interface abnormalities. Our
maps. Subjects with this disorder often have difficulty fixating understanding of the pathophysiology of macular diseases has
and were much more likely to have reliable data with the fast been enhanced by OCT, leading to modifications in clinical
macular scans.17 Scans of high quality are highly reproducible management based on OCT findings. OCT has improved our
with a correlation coefficient of 0.981 for repeated OCT III diagnostic ability, especially for subtle macular disease. There
scans. Ensuring OCT quality in a clinical trial setting is critical has ensued a great increase in the use of this imaging modality
to our ability to trust the data. for the baseline evaluation and follow-up of retinal disorders.
The assessment of retinal thickness and morphology has
Correlation with visual acuity Hee et al demonstrated a become particularly useful in making re-treatment decisions,
correlation of visual acuity with retinal thickness in patients and has led to a decreased necessity for invasive imaging tests
with diabetic macular edema (R2 = 0.79).73 Subsequent clinical in follow-up. Therapeutic clinical trials have increasingly
trials have found variable results and in some cases the incorporated OCT measurements as secondary endpoints. OCT
correlation is poor (R2 = 0.07).103 OCT measurements of retinal images are very useful in enhancing patient education regarding
thickness therefore continue to be a secondary rather than their disorder and management. Further refinements in
primary endpoint in clinical trials. They can be useful as a technology will further improve our ability to incorporate OCT
marker of the biological activity of a treatment. into clinical and research applications.

REFERENCES
1. Huang D, Swanson EA, Lin CP, et al: 3. Kwon RS, Brugge WR: New advances in 5. Pitris C, Jesser C, Boppart SA, et al:
Optical coherence tomography. Science pancreatic imaging. Curr Opin Feasibility of optical coherence tomography
1991; 254:1178–1181. Gastroenterol 2005; 21:561–567. for high-resolution imaging of human
2. Schuman JS, PC, Fujimoto JG: Optical 4. Fujimoto JG, Pitris C, Boppart SA, et al: gastrointestinal tract malignancies.
coherence tomography of ocular diseases. Optical coherence tomography: an emerging J Gastroenterol 2000; 35:87–92.
2nd edn. Thoroughfare, New Jersey: Slack; technology for biomedical imaging and 6. Boppart SA, Brezinski ME, Pitris C, et al:
2004. optical biopsy. Neoplasia 2000; 2:9–25. Optical coherence tomography for 1737
RETINA AND VITREOUS

neurosurgical imaging of human intracortical 25. Foos RY, Kreiger AE, Forsythe AB, et al: 43. Mori K, Gehlbach PL, Sano A, et al:
melanoma. Neurosurgery 1998; 43:834–841. Posterior vitreous detachment in diabetic Comparison of epiretinal membranes of
7. Brezinski ME, Tearney GJ, Bouma BE, et al: subjects. Ophthalmology 1980; differing pathogenesis using optical
Imaging of coronary artery microstructure 87:122–128. coherence tomography. Retina 2004;
(in vitro) with optical coherence 26. Witkin AJ, Ko TH, Fujimoto JG, et al: 24:57–62.
tomography. Am J Cardiol 1996; 77:92–93. Redefining lamellar holes and the 44. Suzuki T, Terasaki H, Niwa T, et al: Optical
8. Born M, Wolf E, Bhatia AB: Principles of vitreomacular interface: an ultrahigh- coherence tomography and focal macular
optics: electromagnetic theory of resolution optical coherence tomography electroretinogram in eyes with epiretinal
propagation, interference and diffraction of study. Ophthalmology 2006; 113:388–397. membrane and macular pseudohole. Am J
light. 7th edn. Cambridge, England: 27. Gass J: Stereoscopic atlas of macular Ophthalmol 2003; 136:62–67.
Cambridge University Press; 1999. diseases: diagnosis and treatment. 4th edn. 45. Haouchine B, Massin P, Tadayoni R, et al:
9. Youngquist R, Carr S, Davies D: Optical St Louis: Mosby; 1997. Diagnosis of macular pseudoholes and
coherence domain reflectometry: a new 28. Smiddy WE, Green WR, Michels RG, et al: lamellar macular holes by optical
optical evaluation technique. Opt Lett Ultrastructural studies of vitreomacular coherence tomography. Am J Ophthalmol
1987; 12:158. traction syndrome. Am J Ophthalmol 1989; 2004; 138:732–739.
10. Hee MR, Izatt JA, Swanson EA, et al: 107:177–185. 46. Gaudric A, Haouchine B, Massin P, et al:
Optical coherence tomography of the 29. Smiddy WE, Michels RG, Glaser BM, et al: Macular hole formation: new data provided
human retina. Arch Ophthalmol 1995; Vitrectomy for macular traction caused by by optical coherence tomography. Arch
113:325–332. incomplete vitreous separation. Arch Ophthalmol 1999; 117:744–751.
11. Swanson EA, Izatt JA, Hee MR, et al: In Ophthalmol 1988; 106:624–628. 47. Hee MR, Puliafito CA, Wong C, et al:
vivo retinal imaging by optical coherence 30. Margherio RR, Trese MT, Margherio AR, Optical coherence tomography of macular
SECTION 10

tomography. Opt Lett 1993; 18:1864–1866. et al: Surgical management of holes. Ophthalmology 1995; 102:748–756.
12. Schuman JS, Puliafito CA, Fujimoto JG: vitreomacular traction syndromes. 48. Tanner V, Chauhan DS, Jackson TL, et al:
Principles of optical coherence Ophthalmology 1989; 96:1437–1445. Optical coherence tomography of the
tomography. In: Schuman JS, Fujimoto JG, 31. McDonald HR, Johnson RN, Schatz H: vitreoretinal interface in macular hole
eds. Optical coherence tomography of Surgical results in the vitreomacular formation. Br J Ophthalmol 2001;
ocular diseases. 2nd edn. Thoroghfare, traction syndrome. Ophthalmology 1994; 85:1092–1097.
New Jersey: Slack; 2004:3–20. 101:1397–1402; discussion 1403. 49. Haouchine B, Massin P, Gaudric A: Foveal
13. Drexler W, Morgner U, Ghanta RK, et al: 32. Hikichi T, Yoshida A, Trempe CL: Course of pseudocyst as the first step in macular hole
Ultrahigh-resolution ophthalmic optical vitreomacular traction syndrome. Am J formation: a prospective study by optical
coherence tomography. Nat Med 2001; Ophthalmol 1995; 119:55–61. coherence tomography. Ophthalmology
7:502–507. 33. Carpineto P, Ciancaglini M, Aharrh-Gnama A, 2001; 108:15–22.
14. Drexler W, Sattmann H, Hermann B, et al: et al: Optical coherence tomography 50. Smiddy WE, Michels RG, de Bustros S,
Enhanced visualization of macular imaging of surgical resolution of bilateral et al: Histopathology of tissue removed
pathology with the use of ultrahigh- vitreomacular traction syndrome related to during vitrectomy for impending idiopathic
resolution optical coherence tomography. incomplete posterior vitreoschisis: a case macular holes. Am J Ophthalmol 1989;
Arch Ophthalmol 2003; 121:695–706. report. Eur J Ophthalmol 2004; 108:360–364.
15. Drexler W: Ultrahigh-resolution optical 14:438–441. 51. Smiddy WE, Michels RG, Glaser BM, et al:
coherence tomography. J Biomed Opt 34. Carpineto P, Mastropasqua L: Incomplete Vitrectomy for impending idiopathic
2004; 9:47–74. posterior vitreoschisis and vitreomacular macular holes. Am J Ophthalmol 1988;
16. Gloesmann M, Hermann B, Schubert C, traction. Am J Ophthalmol 2005; 105:371–376.
et al: Histologic correlation of pig retina 140:765–766; author reply 766. 52. Ang A, Snead DR, James S, et al: A rationale
radial stratification with ultrahigh-resolution 35. Thomas D, Bunce C, Moorman C, et al: for membrane peeling in the repair of stage
optical coherence tomography. Invest Frequency and associations of a taut 4 macular holes. Eye 2006; 20:208–214.
Ophthalmol Vis Sci 2003; 44:1696–1703. thickened posterior hyaloid, partial 53. Posselt D, Rahman R, Smith M, et al:
17. Altaweel M, Timm R, Webster M, et al: vitreomacular separation, and subretinal Visual outcomes following ICG assisted
Assessing AMD with optical coherence fluid in patients with diabetic macular ILM peel for Macular Hole. Eye 2005;
tomgraphy (OCT): fast macular thickness edema. Retina 2005; 25:883–888. 19:279–283.
maps versus high resolution macular 36. Catier A, Tadayoni R, Paques M, et al: 54. Smiddy WE, Feuer W, Cordahi G: Internal
thickness maps. IOVS 2006; 47:abstract Characterization of macular edema from limiting membrane peeling in macular hole
5716. various etiologies by optical coherence surgery. Ophthalmology 2001;
18. Chan A, Duker JS: A standardized method tomography. Am J Ophthalmol 2005; 108:1471–1476; discussion 1477–1478.
for reporting changes in macular thickening 140:200–206. 55. Da Mata AP, Burk SE, Foster RE, et al:
using optical coherence tomography. Arch 37. Smiddy WE, Maguire AM, Green WR, et al: Long-term follow-up of indocyanine green-
Ophthalmol 2005; 123:939–943. Idiopathic epiretinal membranes. assisted peeling of the retinal internal
19. Goulding A, Altaweel M: Reproducibility Ultrastructural characteristics and limiting membrane during vitrectomy
and quality assessment of stratus optical clinicopathologic correlation. Ophthalmology surgery for idiopathic macular hole repair.
coherence tomography (OCT3): fast 1989; 96:811–820; discussion 821. Ophthalmology 2004; 111:2246–2253.
macular thickness scans. IOVS 2006; 38. Smiddy WE, Maguire AM, Green WR, et al: 56. Da Mata AP, Burk SE, Riemann CD, et al:
46:abstract 1545. Idiopathic epiretinal membranes: Indocyanine green-assisted peeling of the
20. Hwang JC, Barile GR, Schiff WM, et al: ultrastructural characteristics and retinal internal limiting membrane during
Optical coherence tomography in asteroid clinicopathologic correlation. Retina 2005; vitrectomy surgery for macular hole repair.
hyalosis. Retina 2006; 26:661–665. 25(5 Suppl):811–820; discussion 821. Ophthalmology 2001; 108:1187–1192.
21. Browning DJ, Fraser CM: Optical 39. Appiah AP, Hirose T, Kado M: A review of 57. Haritoglou C, Reiniger IW, Schaumberger M,
coherence tomography to detect macular 324 cases of idiopathic premacular gliosis. et al: Five-year follow-up of macular hole
edema in the presence of asteroid hyalosis. Am J Ophthalmol 1988; 106:533–535. surgery with peeling of the internal limiting
Am J Ophthalmol 2004; 137:959–961. 40. Smiddy WE, Michels RG, Gilbert HD, et al: membrane: update of a prospective study.
22. Koozekanani D, Boyer K, Roberts C: Clinicopathologic study of idiopathic macular Retina 2006; 26:618–622.
Retinal thickness measurements from pucker in children and young adults. Retina 58. Kumagai K, Furukawa M, Ogino N, et al:
optical coherence tomography using a 1992; 12:232–236. Long-term outcomes of internal limiting
Markov boundary model. IEEE Trans Med 41. Smiddy WE, Michels RG, Green WR: membrane peeling with and without
Imaging 2001; 20:900–916. Morphology, pathology, and surgery of indocyanine green in macular hole surgery.
23. Hayreh SS, Jonas JB: Posterior vitreous idiopathic vitreoretinal macular disorders. A Retina 2006; 26:613–617.
detachment: clinical correlations. review. Retina 1990; 10:288–296. 59. Kimura T, Takahashi M, Takagi H, et al: Is
Ophthalmologica 2004; 218:333–343. 42. Wilkins JR, Puliafito CA, Hee MR, et al: removal of internal limiting membrane
24. Foos RY: Vitreoretinal juncture; Characterization of epiretinal membranes always necessary during stage 3 idiopathic
topographical variations. Invest Ophthalmol using optical coherence tomography. macular hole surgery? Retina 2005;
1738 1972; 11:801–808. Ophthalmology 1996; 103:2142–2151. 25:54–58.
Optical Coherence Tomography

60. Altaweel M, Ip M: Macular hole: improved diabetic retinopathy. Am J Ophthalmol 93. Stone TW, Sternberg P Jr: Submacular
understanding of pathogenesis, staging, 2001; 132:458–461. surgery trials update. Ophthalmol Clin
and management based on optical 76. Jaffe GJ: Cystoid macular edema. focal North Am 2002; 15:479–488.
coherence tomography. Semin Ophthalmol points: clinical modules for 94. Bressler NM, Bressler SB, Hawkins BS,
2003; 18:58–66. ophthalmologists; 1994. et al: Submacular surgery trials randomized
61. Williams GA: Macular holes: the latest in 77. Kraff MC, Sanders DR, Jampol LM, et al: pilot trial of laser photocoagulation versus
current management. Retina 2006; 26(6 Factors affecting pseudophakic cystoid surgery for recurrent choroidal
Suppl):S9–S12. mascular edema: five randomized trials. neovascularization secondary to age-
62. Tadayoni R, Massin P, Haouchine B, et al: J Am Intraocul Implant Soc 1985; related macular degeneration. I.
Spontaneous resolution of small stage 3 11:380–385. Ophthalmic outcomes submacular surgery
and 4 full-thickness macular holes viewed 78. Dajani HM, Lauer AK: Optical coherence trials pilot study report number 1. Am J
by optical coherence tomography. Retina tomography findings in niacin maculopathy. Ophthalmol 2000; 130:387–407.
2001; 21:186–189. Can J Ophthalmol 2006; 41:197–200. 95. Rosenfeld PJ, Intravitreal avastin: the low
63. Larsson J, Holm K, Lovestam M-Adrian: 79. Bressler NM: Cystoid macular edema from cost alternative to lucentis? Am J
The presence of an operculum verified by niacin typically is not accompanied by Ophthalmol 2006; 142:141–143.
optical coherence tomography and other fluorescein leakage on angiography. Am J 96. Gragoudas ES, Adamis AP, Cunningham ET,
prognostic factors in macular hole surgery. Ophthalmol 2005; 139:951; author reply 951. et al: Pegaptanib for neovascular age-
Acta Ophthalmol Scand 2006; 84:301–304. 80. Callanan D, Blodi BA, Martin DF: Macular related macular degeneration. N Engl J
64. Jaycock PD, Bunce C, Xing W, et al: edema associated with nicotinic acid Med 2004; 351:2805–2816.
Outcomes of macular hole surgery: (niacin). JAMA 1998; 279:1702. 97. Rosenfeld PJ, Brown DM, Heier JS, Boyer

CHAPTER 130
implications for surgical management and 81. Spaide RF, Lee JK, Klancnik JK Jr, et al: DS: MARINA Study Group. Ranibizumab
clinical governance. Eye 2005; 19:879–884. Optical coherence tomography of branch for neovascular age-related macular
65. Wong TY, Klein R, Islam FM, et al: Diabetic retinal vein occlusion. Retina 2003; degeneration. N Engl J Med 2006 Oct 5;
retinopathy in a multi-ethnic cohort in the 23:343–347. 355(1):1419–1431.
United States. Am J Ophthalmol 2006; 82. Lerche RC, Schaudig U, Scholz F, et al: 98. Brown DM, Kaiser PK, Michels M,
141:446–455. Structural changes of the retina in retinal Soubrane G, Heier JS: ANCHOR Study
66. Knudtson MD, Klein BE, Klein R, et al: Age- vein occlusion – imaging and quantification Group. Ranibizumab versus verteporfin for
related eye disease, quality of life, and with optical coherence tomography. neovascular age-related macular
functional activity. Arch Ophthalmol 2005; Ophthalmic Surg Lasers 2001; 32:272–280. degeneration. N Eng J Med 2006 Oct 5;
123:807–814. 83. BVOS: Argon laser scatter 355(14):1432–1444.
67. Kempen JH, O’Colmain BJ, Leske MC, photocoagulation for prevention of 99. Rosenfield PJ, Moshfeghi AA, Puliafito CA:
et al: The prevalence of diabetic neovascularization and vitreous Optical coherence tomography findings
retinopathy among adults in the United hemorrhage in branch vein occlusion. A after an intravitreal injection of bevacizumab
States. Arch Ophthalmol 2004; randomized clinical trial. Branch Vein (avastin) for neovascular age related
122:552–563. Occlusion Study Group. Arch Ophthalmol macular degeneration. Ophthalmic Surg
68. Klein R, Klein BE, Moss SE, et al: The 1986; 104:34–41. Lasers Imaging 2005; 26:331–335.
Wisconsin epidemiologic study of diabetic 84. Green WR, Chan CC, Hutchins GM, et al: 100. Fung AE, Lalwani GA, Rosenfeld PJ,
retinopathy: XVII. The 14-year incidence Central retinal vein occlusion: a prospective Dubovy SR: An optical coherence
and progression of diabetic retinopathy and histopathologic study of 29 eyes in 28 tomography-guided, variable dosing
associated risk factors in type 1 diabetes. cases. Retina 1981; 1:27–55. regimen with intravitreal ranibizumab
Ophthalmology 1998; 105:1801–1815. 85. CVOS: Baseline and early natural history (Lucentis) for neovascular age-related
69. Klein R, Klein BE, Moss SE, et al: The report. The Central Vein Occlusion Study. macular degeneration. Am J Ophthalmol
Wisconsin epidemiologic study of diabetic Arch Ophthalmol 1993; 111:1087–1095. 2007 Apr; 143(4):566–583.
retinopathy. XV. The long-term incidence of 86. Hayreh SS: The CVOS Group M and N 101. Altaweel M, Hubbard L, Gagnon R: The
macular edema. Ophthalmology 1995; reports. Ophthalmology 1996; utility of optical coherence tomography in
102:7–16. 103:350–352; author reply 353–354. eligibility determination for clinical trials of
70. Kim BY, Smith SD, Kaiser PK: Optical 87. Ip M, Kahana A, Altaweel M: Treatment of macular disease therapy. Invest Ophthalmol
coherence tomographic patterns of central retinal vein occlusion with Vis Sci 2005; 46:abstract 1577.
diabetic macular edema. Am J Ophthalmol triamcinolone acetonide: an optical 102. Salm D, Altaweel M, Chew E, et al:
2006; 142:405–412. coherence tomography study. Semin Reproducibility of macular thickness
71. Goebel W, Franke R: Retinal thickness in Ophthalmol 2003; 18:67–73. measurements within stratus OCT3 and
diabetic retinopathy: comparison of optical 88. Ip MS, Gottlieb JL, Kahana A, et al: between OCT3 and OCT2. IOVS 2005;
coherence tomography, the retinal Intravitreal triamcinolone for the treatment 46:abstract 1591.
thickness analyzer, and fundus of macular edema associated with central 103. Diabetic Retinopathy Clinical Research
photography. Retina 2006; 26:49–57. retinal vein occlusion. Arch Ophthalmol Network, Browning DJ, Glassman AR,
72. Cunningham ET Jr, Adamis AP, Altaweel M, 2004; 122:1131–1136. Aiello LP, Beck RW. Relationship between
et al: A phase II randomized double- 89. Hussain D, Gass JD: Idiopathic central optical coherence tomography-measured
masked trial of pegaptanib, an anti- serous chorioretinopathy. Indian J central retinal thickness and visual acuity in
vascular endothelial growth factor aptamer, Ophthalmol 1998; 46:131–137. diabetic macular edema. Ophthalmology
for diabetic macular edema. 90. Quillen DA, Gass DM, Brod RD, et al: 2007 Mar; 114(3):525–536.
Ophthalmology 2005; 112:1747–1757. Central serous chorioretinopathy in women. 104. Chen TC, Cense B, Pierce MC, et al:
73. Hee MR, Puliafito CA, Duker JS, et al: Ophthalmology 1996; 103:72–79. Spectral domain optical coherence
Topography of diabetic macular edema 91. Ahlers C, Michels S, Elsner H, et al: tomography: ultra-high speed, ultra-high
with optical coherence tomography. Topographic angiography and optical resolution ophthalmic imaging. Arch
Ophthalmology 1998; 105:360–370. coherence tomography: a correlation of Ophthalmol 2005; 123:1715–1720.
74. Hee MR, Puliafito CA, Wong C, et al: imaging characteristics. Eur J Ophthalmol 105. Hangai M, Ojima Y, Gotoh N, Inoue R:
Quantitative assessment of macular edema 2005; 15:774–781. Three-dimensional imaging of macular
with optical coherence tomography. Arch 92. Hee MR, Baumal CR, Puliafito CA, et al: holes with high-speed optical coherence
Ophthalmol 1995; 113:1019–1029. Optical coherence tomography of age- tomography. Ophthalmology 2007 Apr;
75. Imai M, Iijima H, Hanada N: Optical related macular degeneration and choroidal 114(4):763–773.
coherence tomography of tractional neovascularization. Ophthalmology 1996;
macular elevations in eyes with proliferative 103:1260–1270.

1739
CHAPTER

131 Retinal Arterial Occlusions


Craig M. Greven and Wesley H. Adams

INTRODUCTION
Overview
Retinal artery occlusion leads to profound functional deficits in Occlusion or obstruction of the ophthalmic, central retinal,
the distribution of the visual field supplied by the affected vessel. branch retinal, or a cilioretinal artery leads to acute loss of vision
These events, although relatively rare, not only have effects on in the distribution of the affected retina. von Graefe initially
the visual system of the patient, but may be the initial indication described CRAO secondary to emboli related to bacterial endo-
of a systemic life-threatening disease.
carditis in the nineteenth century.1 Since then our understanding
The average age at presentation of patients with central retinal
artery occlusion (CRAO) is the early to mid-60s. However, arterial
of retinal arterial occlusion has increased dramatically. Although
occlusive disease can occur at any age. Males are affected relatively infrequent, these events can profoundly impact not
slightly more commonly than females, and there is no racial only the visual function of the individual, but also can be the
predilection. initial indication of life-threatening systemic disease. An under-
Numerous mechanisms of retinal arterial occlusion have been standing of the basic anatomy and pathologic mechanisms of
documented, but the most common are embolic, thrombotic, occlusion is essential in determining the appropriate manage-
vasculitic, and vasospastic. The most common sources of ment and evaluation of patients presenting with acute retinal
embolic occlusion are atherosclerotic disease of the carotid arterial occlusions.
artery and cardiac disease. Thrombosis is also a major cause of
retinal arterial occlusions. Hereditary and acquired conditions, as
well as local factors, can promote thrombosis. The number of VASCULAR ANATOMY
hereditary conditions associated with retinal arterial occlusive
disease continues to grow as our understanding of the The ophthalmic artery is the first intracranial branch of the
coagulation system increases. Vasculitis is another mechanism internal carotid artery. After entering the orbit, the ophthalmic
associated with arterial occlusive disease of the retina. Systemic artery gives off the central retinal artery, which runs adjacent to
vasculitic conditions like giant cell arteritis, as well as local areas the inferior margin of the optic nerve. The central retinal artery
of vasculitis of the retina, can promote obstruction. penetrates 13 mm posterior to the globe to course through the
Retinal arterial occlusive disease can be classified as substance of the nerve to supply the inner retina. Approximately
ophthalmic artery, central retinal artery, branch artery, and 20 short posterior ciliary arteries arise from the ophthalmic artery
cilioretinal artery. Ophthalmic and central have the worst visual
and penetrate the sclera in a ring around the optic nerve to supply
prognosis, whereas branch and cilioretinal artery occlusions
typically have better visual prognosis. The hallmark of arterial
the choroid and outer retina. Cilioretinal arteries arise from
occlusive disease of the retina on clinical examination is ischemic these short posterior ciliary arteries and are present in 30% of
retinal whitening, and on fluorescein angiography is delayed eyes and 50% of individuals.2,3 In 15% of people a cilioretinal
perfusion of the affected vessel. artery supplies some portion of the macular circulation.3
Management of retinal arterial occlusive disease includes The surface of the optic disk is supplied by branches of the
attempting to reestablish retinal arterial blood flow, determining central and branch retinal arteries. The laminar and prelaminar
the associated systemic disease processes and reason for optic nerve is supplied by branches of the posterior ciliary
occlusion, and management of long-term ocular complications. arteries. The deeper optic nerve in supplied by intraneuronal
The single most important factor in determining visual outcome branches from the central retinal artery and recurrent pial
in patients with retinal arterial occlusive disease is duration of
branches of the central retinal and ophthalmic artery.
retinal ischemia. Histopathologic studies have shown that total
occlusion of the central retinal artery leads to irreversible visual Within the retina, larger branches of the central artery course
loss in less than 2 h. Aggressive management is indicated in through the nerve fiber and ganglion cell layers. As the arterial
patients who present within 24 h of symptoms and the individual circulation divides and lumen size narrows smaller arterioles
management of each patient must be determined on a case-by- and capillaries extend through these layers into the inner
case basis. nuclear layer.4 Central retinal artery circulation does not extend
Systemic workup should include carotid ultrasound to look for past the boundary of the inner nuclear and inner plexiform
atherosclerotic disease, echocardiography, and hematologic layers. The choroidal circulation and choriocapillaris supply the
workup to rule out a potential thrombotic or vasculitic condition. outer retina.
In age-appropriate patients, a sedimentation (sed) rate and C-
While retinal circulation is generally end arterial in nature,
reactive protein must be considered when the diagnosis of
temporal arteritis is entertained.
two anastomoses exist in the region of the optic disk between
branches of the central retinal artery and short posterior ciliary
arteries.5 The first occurs in the capillary beds of the optic nerve
head. Here superficial capillaries derived from the central
1741
RETINA AND VITREOUS

retinal artery lie adjacent to those capillaries that penetrate the dendritic lysis by 3.5 h and extensive cellular loss by 16 h.7
deeper regions of the prelaminar and lamina cribrosa, that Hayreh and associates found that rhesus monkey retinas could
originate from posterior ciliary circulation. The second anasto- tolerate 97–98 min of total occlusion, and that only half of eyes
mosis occurs within the dural sheath of the optic nerve. There occluded for 102 min recovered vision. However, all eyes
intraneuronal branches of the central retinal artery communi- occluded 105 min or more suffered irrecoverable ischemia.6,12,13
cate with recurrent pial branches.
DEMOGRAPHICS/INCIDENCE/
HISTOPATHOLOGY OF ARTERIAL PREVALENCE
OCCLUSIONS
Retinal arterial occlusions may occur at any age, but the average
The retina has a high oxidative capacity and a high level of age of patients with CRAO in two large series is 62 and 67.8,14
glycolytic activity. The decrease or absence of perfusion caused Most series demonstrate a slight male predominance.8,14,15
by central retinal artery obstruction leads to decreased delivery There does not appear to be any racial predilection, however.
of glucose and more importantly oxygen6 to the tissues of the Additionally, the right and left eyes appear to be involved equally.
retina. Ischemia leads to edema as neuron cellular membranes Common systemic risks factors for retinal arterial occlusive
burst4,7 producing the associated funduscopic whitening of the disease include hypertension, diabetes, lipid disorders, and car-
retina caused by opacification of the ganglion cell layer. The diac and systemic atherosclerotic disease.
opacification is most prominent in the macular region where It is difficult to determine the precise incidence and preva-
SECTION 10

the multiple layers of ganglion cells reside. Retinal opacification lence of arterial occlusive events in the retina. However, Brown
generally decreases toward the periphery as the ganglion cell and co-authors suggested that 1 in 10 000 patient visits to a
population diminishes, and is also absent from the avascular tertiary-care eye hospital was related to CRAO.16 Additionally,
zone of the foveola where ganglion cells are absent.8 The classic he estimated that less than 1 in 50 000 outpatient visits to an
‘cherry red spot’ is a result of the visible red reflex of the ophthalmologist will be a patient less than age 30 with retinal
perfused choroid at this location. arterial occlusive disease.16
Optical coherence tomography (OCT) of acute CRAO corre-
lates with these histopathologic features and/or characteristics. MECHANISMS OF OCCLUSION
Findings on OCT demonstrate an increase in inner layer
reflectivity and muted outer layer reflectivity around the foveola The principal mechanisms operative in occlusion of the retinal
with normal reflectivity of the retinal pigment epithelium arterial system are embolic, thrombotic, vaso spastic, extra-
beneath the foveola (Fig. 131.1).9 Eventually, cellular debris in vascular compression, and vasculitic. Other mechanisms such
the ischemic inner layers of the retina undergoes phagocytosis as radiation and elevated intraocular pressure occur much less
and gliosis leading to atrophy. commonly. It is important to note that more than one mechan-
Animal studies have demonstrated swelling of the organelles ism may occur at the same time in a patient. An embolism to
that precede axonal and dendritic rupture with decreased the retinal arterial circulation can originate as a venous throm-
neuronal cell count. Mitochondria, the first organelle affected in bosis reaching the arterial circulation via a right to left cardiac
the ganglion cells,6 began to swell as early as 15 min after shunt. CRAO secondary to giant cell arteritis is primarily a
CRAO in squirrel monkeys.7 Microtubules and cisternae of the vasculitic phenomenon, with thrombosis being the ultimate
endoplasmic reticulum are also acutely susceptible to ischemia.6 occlusive event.
In Kroll’s experimental occlusion of the central retinal artery in In general, age is an important factor in determining the mech-
squirrel monkeys, swelling of the endoplasmic reticulum had anism of occlusion. Patients aged 50 and greater are likely to
occurred by 3.5 h.7 Müller cells were relatively more resistant to have occlusions associated with embolic disease while younger
ischemia than the ganglion cells in both species, presumably patients are more likely to have occlusions secondary to
due to their proximity to retinal capillaries and the chorio- thrombosis.
capillaris, and a greater ability to metabolize under anaerobic
conditions. These changes are similar to those seen in
humans.7,10,11 EMBOLISM/ENDOGENOUS
Retinal cellular survival and thus final visual outcome in Since retinal arterial occlusive disease is primarily a disease of
humans is related to duration of ischemia and the presence of the elderly, embolic occlusion represents the most common
residual circulation. However, no controlled data are available mechanism. Atherosclerosis of the carotid system is the most
in humans. In squirrel monkeys, Kroll observed axonal and common source of retinal emboli with 80% being associated
with carotid artery disease.17,18 A common misconception
regarding carotid disease leading to emboli is that the degree of
obstruction of the carotid artery is of paramount importance.
Although degree of obstruction is important, the presence of an
ulcerative plaque, even with minimal stenosis, can be more
likely to lead to emboli than an extensively occluded carotid
artery. Carotid artery disease can be evaluated by carotid ultra-
sonography, arteriography, and CT angiography.
The heart and major vessels are another important source of
emboli that must be considered when confronted with a patient
with embolic retinal arterial occlusions. Valvular abnormal-
ities,19–21 tumors, like atrial myxomas,22,23 and thrombus forma-
tion in the left atrium secondary to conditions like atrial
FIGURE 131.1. OCT of CRAO demonstrating increased thickness in fibrillation, can lead to emboli formation and vascular occlu-
the inner retinal layers and muted outer layer/retinal pigment sion. Additionally, right to left shunts, like patent foramen
epithelium reflectivity surrounding the foveola. Note that there is ovale, can allow emboli from venous thrombotic disease to
1742 normal reflectivity of the retinal pigment epithelium at the foveola. reach the arterial circulation.24 Important in evaluating embolic
Retinal Arterial Occlusions

disease originating from the heart is echocardiography. Trans-


thoracic echocardiography is the preferred initial screening
technique. In cases where there is a high suspicion of disease
that is not detected by transthoracic echocardiography, trans-
esophageal echocardiography can be helpful in identifying a
source of emboli.25,26 Other rare sources of endogenous emboli
include fat emboli from long bone fractures,27 amniotic fluid
emboli at the time of delivery,28,29 and leukoembolization
secondary to conditions like pancreatitis.

Emboli/Exogenous Source
Exogenous emboli can reach the retinal circulation leading to
arterial occlusive events. Examples include talc emboli associ-
ated with intravenous drug abuse30 and emboli from injections
of steroids in the nasal or periorbital area,31,32 or related to blood
products like platelet emboli during transfusion. Fragments of
catheter tips and artifical heart valves may also cause arterial
occlusion.

CHAPTER 131
FIGURE 131.3. CRAO with cilioretinal sparring. Note elongated
Retinal Emboli/Types platelet fibrin emboli in retinal arterial tree.
Although embolic obstruction is presumably the most common
cause of retinal arterial occlusions, visible emboli detected
ophthalmoscopically are only evident in 20–30% of patients
with retinal arterial occlusive disease.14 Clinically the most
common are cholesterol emboli, platelet fibrin emboli, and
calcific emboli (Figs 131.2–131.4).
Cholesterol emboli are small, glistening yellow in color, and
typically do not occlude the vessel distal to the emboli. These
most commonly arise from atherosclerotic plaques of the
carotid artery. Platelet fibrin emboli may appear gray-white, are
larger and longer than cholesterol emboli, and may be seen to
pass through the retinal vessels.
Calcific emboli are larger, gray-white in color, and associated
with more severe local obstructions. Arruga felt that calcific
emboli were more likely to arise from cardiac valvular disease.15
A comprehensive list of emboli types is provided in Table 131.1.

THROMBOSIS
Thrombosis is another mechanism causing arterial occlusive
disease of the retina. Virchow’s classic triad concerning the
pathogenesis of thrombosis includes vessel wall abnormalities,
stasis of blood flow, and changes in the blood components.
While any one of these factors alone may lead to thrombosis, FIGURE 131.4. Inferior hemiretinal artery occlusion in right eye. Note
calcific emboli at the inferior margin of the disk causing occlusion.
combinations of these factors significantly increase the likelihood

TABLE 131.1 Emboli

I. Endogenous
A. Carotid atheroma
B. Cardiac valvular disease
C. Cardiac tumor
D. Fat emboli
E. Leukoemboli
F. Amniotic fluid emboli
G. Septic emboli
II. Exogenous
A. Injected steroids
B. Catheter tips
C. Talc
FIGURE 131.2. Cholesterol emboli at bifurcation of the inferior nasal
retinal artery. Note glistening characteristics. No functional deficit was D. Artificial heart valves
present. 1743
RETINA AND VITREOUS

of these episodes. While venous thrombosis occurs most com-


monly, arterial thrombotic disease of the retina also does occur.
Structural abnormalities in a vessel like prepapillary arterial
loop can contribute to retinal arterial occlusion by thrombosis.
This structural abnormality, which leads to turbulence and
stasis of flow, has been reported to cause CRAOs.33
Tremendous advances have occurred during the last two
decades in understanding abnormalities of blood components
and their relationship to intravascular thrombosis. Both here-
ditary and acquired abnormalities have been identified. Throm-
bophilia is the term used to describe conditions that are
genetically determined and increase the likelihood of vascular
thrombosis (Table 131.2). These abnormalities typically lead to
venous thrombosis, but when they interact with other local
or systemic hypercoagulable factors like trauma, malignancy,
pregnancy, oral contraceptive use, autoimmune disease, and
cigarette smoking, they may lead to arterial thrombosis. These
hereditary thrombophilic states should be considered in patients
SECTION 10

with retinal arterial occlusions, particularly with a positive family


FIGURE 131.5. Twenty-four-year-old with diabetes, pregnancy, and
history of thrombotic events occurring at a young age. While protein S deficiency. Note branch macular artery occlusion in left eye.
entire textbooks have been devoted to discussing these throm-
bophilic conditions, a brief explanation of some of the more
common of these conditions that have been associated with
retinal arterial occlusive events will be useful.

Antithrombin Deficiency
Human antithrombin (AT) is a plasma protein that is a protease
inhibitor and prevents the formation of thrombi.34 Previously
termed AT III deficiency, AT deficiency was the first hereditary
deficiency of an anticoagulant protein described. Inherited as an
autosomal dominant trait, AT deficiency is associated with a
thrombotic tendency typically manifested as venous thrombo-
embolism. However, retinal arterial occlusions secondary to
thrombosis have been reported.35

Protein C and S Deficiency


Protein C and protein S are vitamin K dependent plasma glyco-
proteins synthesized in the liver. Deficiency in these proteins
occurs primarily as a hereditary condition inherited as an auto-
somal dominant trait, but can be an acquired defect associated
with advanced severe liver disease. Both protein C and protein FIGURE 131.6. Fluorescein angiogram of BRAO secondary to protein
S serve as cofactors for the anticoagulant function of activated S deficiency in left eye.
protein C, a protein that inactivates factors 5A and 8A, and

TABLE 131.2 Thrombophilias Associated with Retinal Arterial downregulates thrombosis.34 Deficiencies of either protein C or
Occlusions protein S leads to a thrombotic tendency, and arterial occlusive
disease of the retina has been described with each (Figs 131.5
I. Factor V Leiden
and 131.6).36,37
II. Antithrombin deficiency
Activated Protein C Resistance and Factor V
III. Protein C deficiency
Leiden
IV. Protein S deficiency As mentioned previously, activated protein C inactivates factor
V. Hyperhomocystinemia V-A and factor VIII-A and downregulates thrombosis. Activated
protein C resistance refers to an abnormally reduced anti-
VI. Anti-phospholipid antibodies
coagulant response of a subject’s plasma to activated protein C
VII. Lupus anticoagulant in in vitro testing. Although any genetic abnormality of the
VIII. Anticardiolipin protein C pathway can cause activated protein C resistance, more
than 90% of hereditary activated protein C resistance subjects
IX. Homozygous C677T polymorphism in methylene have the same genetic abnormality termed factor V Leiden.
tetrahydrofolate reductase
Factor V Leiden is an abnormal factor V associated with a point
X. Prothrombin G20210A mutation and a single amino acid change. Factor V Leiden is
XI. Dysfibrogenemia resistant to the proteolytic action of activated protein C, leading
to increased thrombin formation.34 The subsequent hyper-
XII. Elevated factor VIII coagulable state is one of the most common hereditary causes
XIII. Elevated factor IX of thrombophilia and has been associated with arterial occlusive
1744 disease of the retina.38,39
Retinal Arterial Occlusions

Hyperhomocystinemia
Homocysteine is an intermediate in the metabolism of the
amino acids methionine and cysteine. Hyperhomocystinemia
refers to elevated plasma levels of homocysteine and can be
caused by numerous factors. The most common genetic cause
of hyperhomocystinemia is a mutation in the enzyme methylene
tetrahydrofolate reductase (MTHFR) leading to elevated levels
of plasma homocysteine. Present in 10–20% of Caucasians,
10% of Asians, and rare in blacks, either alone or in combi-
nation with low folate, B6 or B12 levels or other conditions like
renal failure, hypothyroidism, smoking, excess caffeine con-
sumption, and inflammatory bowel disease, this defect can be
associated with increased levels of plasma homocysteine. The
exact mechanism of thrombosis associated with hyperhomo-
cystinemia is poorly understood40 but endothelial toxicity and
smooth muscle proliferation have been proposed. Arterial
thrombotic disease of the retina has been reported.41,42

CHAPTER 131
Antiphospholipid Syndrome
The antiphospholipid syndrome (previously known as anti-
phospholipid antibody syndrome, anticardiolipin antibodies FIGURE 131.7. BRAO associated with vasculitis secondary to retinitis
syndrome, and lupus anticoagulant) is an acquired disorder due to Bartonella henselae.
associated with thrombotic complications. The condition is
considered secondary if associated with other diseases like
systemic lupus erythematosus, other autoimmune disorders,
viral infections, malignancies, or those associated with medi-
VASOSPASM
cations. It is considered primary when it occurs in the absence Vasospasm as a primary cause of retinal arterial occlusive
of any of these conditions. The syndrome is characterized by disease57–62 is felt to be a rare occurrence. Gass has suggested
the presence of antibodies to phospholipid protein complexes that some degree of reflex spasm may play a role in retinal
(anticardiolipin, antiphosphatidyl serine or anti-b 2 glyco- arterial occlusions from many causes including migraine, collagen
protein) or by the detection of lupus anticoagulants.43 The vascular disease, and sickle cell hemoglobinopathies.63,64
precise mechanism by which these antiphospholipid antibodies
promote thrombosis is poorly understood, but may be related to
the inhibition of prostacyclin, a platelet inhibitory prosta- OTHER MECHANISMS
glandin. It has also been speculated that it might impair protein Local conditions in the eye and orbit may lead to situations pre-
C and AT III anticoagulant mechanisms. While venous occlusive disposing to retinal arterial occlusive events. Examples of con-
disease is most common, arterial occlusive disease of the ditions associated with retinal arterial occlusion from intrinsic
retina also has been associated with the antiphospholipid structural anomalies include peripapillary arterial33 loop, optic
syndrome.44–50 nerve head drusen,65 and intraocular foreign body (Figs 131.8
and 131.9).
Elevated intraocular pressure secondary to angle-closure
VASCULITIS AND GIANT CELL ARTERITIS glaucoma or compression of the globe can lead to CRAO.
The classic vasculitis-related CRAO occurs in giant cell arteritis. External compression of the ophthalmic artery or central retinal
As mentioned previously, giant cell arteritis is an example of an
arterial occlusion occurring from a combined mechanism, both
vasculitis and thrombosis.51 Often associated with ocular pain
because of the profound ischemia, patients of the appropriate
age (greater than 55 years) who present with CRAO should be
questioned for associated symptoms like headache, scalp tender-
ness, jaw claudication, malaise, anorexia, fever, and weight loss,
recognizing that 20% of patients with giant cell arteritis are with-
out systemic symptoms. Patients with acute CRAO secondary
to giant cell arteritis may have prodromal transient blurring of
vision for weeks to months prior to the occlusive event.
The suspected diagnosis is solidified by an elevated erythrocyte
sedimentation rate and C-reactive protein and confirmed by
temporal artery biopsy. Making an accurate diagnosis and
initiation of high-dose steroid therapy is imperative because
second eye involvement may occur soon following presentation.
Systemic vasculitis associated with collagen vascular diseases
can cause retinal arterial occlusions. Classic examples are rheu-
matoid vasculitis and systemic lupus erythematosus. Other
systemic vasculities like Behçet’s disease52 may lead to branch
retinal arterial occlusions. Localized vasculitis in the retina
can occur in other conditions like toxoplasmosis retinitis
and Bartonella henselae leading to retinal arterial occlusions FIGURE 131.8. BRAO in right eye secondary to prepapillary arterial
(Fig. 131.7).53–56 loop. 1745
SECTION 10 RETINA AND VITREOUS

FIGURE 131.9. Inferior BRAO secondary to intraocular foreign body


imbedded in the retina with distal occlusion of the inferior temporal
artery. There is hemorrhage overlying the intraocular foreign body.
FIGURE 131.10. Ophthalmic artery occlusion in left eye secondary to
atrial myxoma emboli. Note pallid swelling of the optic nerve. No
evidence of cherry red spot. Note segmentation of the blood column
artery caused by orbital cellulitis, orbital hemorrhage abscess in the retinal vessels.
formation, cavernous sinus thrombosis, and neoplastic disease
may lead to CRAO.

CLINICAL FINDINGS

AMAUROSIS FUGAX
Amaurosis fugax refers to transient monocular loss of vision,
either total or altitudinal, that may serve as a prodromal
symptom for retinal arterial occlusion. Visual loss in amaurosis
fugax typically lasts 7–30 min with total resolution to normal.
Patients with classic symptoms of amaurosis fugax should
undergo complete ophthalmic examination, specifically looking
for the presence of retinal emboli. In these patients, a systemic
workup should be performed for the presence of embolic
disease. Specifically, an evaluation of the carotid artery system
as well as cardiac evaluation with echocardiography should be
performed. Consultation with the patient’s internist or
neurologist is indicated to determine whether anticoagulants
should be initiated.

OPHTHALMIC ARTERY OCCLUSIONS


Ophthalmic artery occlusion causes profound painless unilateral FIGURE 131.11. Chronic ophthalmic artery occlusion in right eye.
loss of vision. Visual function in these cases is extremely poor Note pale disk and ischemic retinal arteries and veins.
as both the retinal and choroidal circulation is interrupted.
Visual acuity ranges from counting fingers to no light per-
ception. An afferent pupillary defect is present. The classic both the B wave and A wave are absent, while in CRAO with
fundus picture is that of an acutely infarcted retina with intact choroidal perfusion, the A wave is present.
extensive retinal whitening (Fig. 131.10). Often there is pallid The chronic picture of ophthalmic artery occlusion is a pale
swelling of the optic disk. Because the choroidal circulation is disk with attenuation of the retinal arterials and venules
compromised, a cherry red spot is not visualized. There is often (Fig. 131.11). Often, retinal pigment epithelial abnormalities
boxcarring or segmentation of the blood column of the retinal will occur related to the profound ischemia of the retinal
vessels. Fluorescein angiography shows no or markedly delayed pigment epithelium and choriocapillaris.
background choroidal flush and minimal or delayed filling of
the retinal vasculature.
Visual recovery is rare in cases of ophthalmic artery occlusion. CENTRAL RETINAL ARTERY OCCLUSIONS
Electroretinographic testing can help differentiate ophthalmic CRAOs also present with painless unilateral profound loss of
artery occlusions from CRAO. In ophthalmic artery occlusions, vision. Visual acuity of less than 20/400 occurs in more than
1746
Retinal Arterial Occlusions

CHAPTER 131
a

FIGURE 131.12. CRAO in left eye. Note cherry red spot with
edematous ischemic retina. Note segmentation of blood column.

90% of eyes at the time of presentation. An afferent pupillary


defect is present. Visual fields are depressed in the area of the
ischemic retina. The retina shows ischemic whitening, and
the retinal arterials and veins manifest boxcarring. A cherry
red spot is present due to the intact choroidal circulation
(Fig. 131.12). The diagnosis may not be obvious in patients
presenting within 1–2 h of occlusion, prior to the development
of an edematous retina. In these cases, fluorescein angiography
is useful and will show an intact choroidal flush with absent,
incomplete, or delayed filling and a leading edge of dye in the
retinal circulation (Fig. 131.13a,b).
The median final visual acuity following resolution of edema
in CRAOs is counting fingers. However, in select cases with
intact cilioretinal arteries supplying the fovea, Snellen visual
acuity may improve to 20/20 (Figs 131.14 and 131.15). Visual
field defects persist in the area of the infarcted retina. Chronic b
fundus changes in CRAO include optic atrophy, attenuated
retinal arterials and venules, and an atrophic-appearing retina. FIGURE 131.13. Fluorescein angiogram of CRAO left eye at (a) 33 s
and (b) 47 s showing a leading edge of dye in the arterial system.
BRANCH RETINAL ARTERY OCCLUSIONS
Branch retinal artery occlusion (BRAO) presents as an acute
painless loss of visual field in the distribution of the occluded
artery. Visual acuity may range from 20/20 to 20/400 depending
CILIORETINAL ARTERY OCCLUSIONS
on the degree of foveal involvement. An afferent pupillary defect Approximately 35% of eyes and 50% of people have cilioretinal
may be present, depending on the degree of retina involvement. arteries. As mentioned previously, cilioretinal arteries are derived
Retinal whitening, along the distribution of the artery is usually from the short ciliary arteries. Typical symptoms of patients
present, but may not occur in nasal branch artery occlusions with cilioretinal artery occlusions are pericentral scotomas,
due to the single layer of ganglion cells present there. Emboli are often subtle, in the distribution of the artery.67 As the area
identified in ~30% of cases. Fluorescein angiogram depicts supplied by the occlusion is usually small, afferent pupillary
delayed transit of the dye through the affected vessel, and often defects are often not present. Although any of the previously
retrograde filling of the vessel can be observed later in the described mechanisms of occlusion may be operational, cilio-
angiogram (Figs 131.16 and 131.17). Combined branch retinal retinal artery occlusions may occur in conjunction with central
artery and vein occlusions are relatively rare (Fig. 131.18). retinal venous occlusive disease and anterior ischemic optic
Patients with BRAOs typically retain excellent visual acuity neuropathy (Figs 131.19 and 131.20).68
following the acute event, as long as a portion of the foveal Cilioretinal artery occlusions occurring in isolation usually
vasculature maintains normal perfusion. Visual acuity improves have a prognosis as good as or better than BRAOs, with 90%
to 20/40 or better in 80% of these eyes, but visual field defects improving to 20/40 or better visual acuity. When cilioretinal
persist.66 The fundus appearance may return to near normal artery occlusions occur concomitant with CRVO, the prognosis
weeks to months following the event. varies depending on resolution of complications associated with
1747
SECTION 10 RETINA AND VITREOUS

FIGURE 131.16. Inferior BRAO with calcific emboli at inferior margin


FIGURE 131.14. Fundus photograph of right eye with CRAO with of the optic disk beneath overlying vein.
cilioretinal sparing. Visual acuity improved to 20/20.

FIGURE 131.17. Fluorescein angiogram of inferior BRAO showing


delayed perfusion of inferior retina with retrograde filling of inferior
retinal veins.

FIGURE 131.15. Fluorescein angiogram of right eye showing


perfused cilioretinal artery, but CRAO and delayed filling of the retinal
arterials. patient management. With regards to visual function, attempts
to restore visual function are most critical in central and
ophthalmic artery occlusions and many of the methods we will
describe are often not utilized in branch or cilioretinal artery
the central retinal vein occlusion like macular edema, ischemia, occlusions. Patients with BRAO or cilioretinal artery occlusion
and hemorrhage.69 typically have an excellent prognosis for regaining visual acuity.
However, the management of each case much be individualized.
MANAGEMENT As previously mentioned, the duration of retinal ischemia is
the most important factor in determining prognosis. Since it is
Management of patients with retinal arterial occlusions can be often difficult to pinpoint the actual initial occlusive event,
considered in three aspects: (1) management of the acute occlusive aggressive management is appropriate, particularly in patients
event in an attempt to restore visual function; (2) workup of the with a duration of occlusion less than 24 h. While there are
patient looking for potential systemic conditions requiring many reports of successful outcomes with various treatments in
treatment; and (3) management of the remote complications select cases, unfortunately, there is no proven beneficial
and sequelae of the arterial occlusive event. Although the first treatment for improving visual outcomes in patients with these
two are the most important aspects, all are critical for optimal events.70 Therapeutic goals are to restore blood flow as quickly
1748
Retinal Arterial Occlusions

CHAPTER 131
FIGURE 131.20. Cilioretinal artery occlusion in right eye, associated
with anterior ischemic optic neuropathy.
FIGURE 131.18. Combined branch retinal artery and vein occlusion in
right eye.

an embolus have been attempted to improve retinal arterial


circulation. Studies have shown no significant difference in final
versus initial visual outcomes in use of topical beta blockers,
oral acetazolamide, or paracentesis.71,72
Medical vasodilatation utilizing sublingual nitroglycerin,
calcium channel blockers and pentoxifylline have also been
used to improve the retinal arterial circulation. While results of
one study found that the pentoxifylline group had less neovas-
cularization compared to standard treatment, there was no defi-
nitive improvement in visual outcomes.73 To date there are only
anecdotes of effectively treating CRAO with nitroglycerin.74,75 It
has been hypothesized that nifedipine or other calcium channel
blockers may reduce the effects of calcium overload in ischemic
retina;76 however, to date no studies showing an improvement
in final visual outcome have been documented. Carbogen, 95%
oxygen and 5% carbon dioxide, is an inhalation treatment that
may cause dilation of retinal arterioles,77 and has been used to
treat CRAO. This modality of treatment has not shown benefit
to final visual outcome and is generally no longer used.72
There are case reports of thrombolytic therapy improving
visual recovery even when initiated more than 2 h after occlu-
sion. However, less than 16% of CRAO cases are due to platelet-
fibrin obstruction.15 Given the lack of clinical evidence of
improved visual outcome, potential for systemic complications,
and inherent difficulty establishing nonembolic etiology, the
decision to attempt fibrinolytic therapy using drugs like TPA
FIGURE 131.19. Inferior cilioretinal artery occlusion with associated should be made carefully.8,78 More data may justify more
central retinal vein occlusion. widespread future use of local or systemic modalities of
treatment.71,79–82
Surgical treatments have been tried with embolic arterial
as possible to limit the damage to the cellular constituents of occlusion with occasional/rare efficacy. Laser photo disruption
the retina. of an embolus has been reported in select patients to cause
Digital ocular massage is a simple, common, noninvasive passage of the embolus through the arterial tree with improve-
initial technique utilized to restore retinal circulation. In this ment in outcomes.83 Vitrectomy with cannulation of the central
technique the fingers are used to compress the globe, raising the retinal artery has also been proposed for management of
intraocular pressure with subsequent release of the pressure embolic retinal arterial occlusive disease.84 No randomized trial
hoping that the arterial pulse will displace any occlusive emboli data have confirmed the efficacy of any of these treatments.
into the distal retinal arterioles and restore flow. No statistical Although mentioned previously, in patients with CRAO, if
benefit of ocular massage has been noted, but as it is a simple giant cell arteritis is suspected by clinical and laboratory para-
treatment with low morbidity, it should be tried in all patients meters, high-dose steroid therapy should be initiated while
with CRAO. awaiting temporal artery biopsy.
Reduction of intraocular pressure through ocular antihyper- The second aspect of management of retinal arterial occlusive
tensives and paracentesis which theoretically may also dislodge disease is systemic workup. This has been covered thoroughly
1749
RETINA AND VITREOUS

is estimated at up to 40%.95 Visual prognosis in the ocular


TABLE 131.3 Initial Workup
ischemic syndrome is poor with the majority of cases pro-
I. Carotid ultrasound gressively losing vision. Panretinal laser photocoagulation may
be efficacious in preventing the development of neovascular
II. Echocardiography – transthoracic, transesophageal
glaucoma. Focal laser photocoagulation in ocular ischemic
III. ESR, C-reactive protein syndrome patients with macular edema is another treatment
IV. CBC, platelet count modality where efficacy has not been proven through random-
ized controlled clinical trials, but may be reasonable. Prompt
V. PT, PTT referral to an internist, neurologist, or vascular surgeon is
VI. Protein C, protein S, activated protein C, factor V Leiden, necessary to address modifiable risk factors and to consider
fasting plasma homocysteine level, anti-phospholipid carotid endarterectomy in the appropriate patient. The efficacy
antibodies of endarterectomy in stroke prevention in symptomatic patients
with 70–99% stenosis has been demonstrated in the North
American symptomatic carotid endarterectomy trial.96
within this chapter, but carotid artery studies including duplex
Doppler ultrasound, transthoracic or transesophageal echo-
cardiography,85 and hematologic testing to rule out various
vasculitic and thrombosis type syndromes must be performed
SECTION 10

(see Table 131.3).86 In general the workup should be tailored to


the patient’s age16,87,88 and co-morbidities. In up to 30% of
patients, no systemic associated condition leading to arterial
occlusion is documented.
The most devastating long-term ocular complication of retinal
arterial occlusive disease is neovascular glaucoma. Neovascular
glaucoma has been reported to occur in 2–16% of these eyes.89,90
The time from occlusion to development of neovascular
glaucoma ranges from one week to years with most events
occurring during the first 2 months. While the eye typically has
limited vision secondary to the initial occlusion, the preference
of a limited vision, comfortable, cosmetically acceptable eye
versus a blind painful eye is obvious. Treatment is as with any
other form of neovascular glaucoma with laser or cryotherapy
to the retina and ciliary body and topical antihypertensive
medications.

OCULAR ISCHEMIC SYNDROME


While the ocular ischemic syndrome is not a ‘retinal arterial
occlusion’ it does represent chronic arterial insufficiency to the FIGURE 131.21. Red free photograph of right eye showing fairly
retina, choroid, and entire globe. The ocular ischemic syndrome normal posterior pole. Neovascularization of the disk is present.
can be defined as a syndrome of hypoperfusion to the globe
secondary to carotid artery or more rarely ophthalmic artery
occlusive disease. Patients with the ocular ischemic syndrome
may be asymptomatic initially or may note gradual blurring,
often of a transient nature. Patients are typically 50 years old or
older and often have significant atherosclerotic disease with
other cardiovascular or cerebral vascular symptoms.91
Anterior segment findings include chronic flare with minimal
cellular response and neovascularization of the iris or angle
structures. Posterior segment findings include retinal hemor-
rhages in all four quadrants, typically present in the midzone.
Arterials are narrowed and veins are typically of normal caliber.
This is a differentiating feature from central retinal vein occlu-
sion where veins are dilated. Microaneurysms and macular
edema may be present and in advanced cases neovascular-
ization of the disk or retina may be present.
On fluorescein angiography there is often delayed asym-
metric filling of the choroid, delayed retinal arterial filling,92 and
prolonged venous transit time.93 Additionally, the retinal
vessels often stain late in the study indicating chronic retinal
ischemia.94 Electroretinography typically shows decreased
amplitude of the A and B wave secondary to global posterior
ischemia (Figs 131.21–131.24).
It is critical to make the diagnosis of ocular ischemic syn-
drome because these patients often develop cerebral vascular FIGURE 131.22. Fluorescein angiogram shows delayed filling of
accidents. The 5-year mortality in ocular ischemic syndrome retinal arterials with asymmetric filling of the choroid.
1750
Retinal Arterial Occlusions

CHAPTER 131
FIGURE 131.23. Progressive arterial filling with delayed venous filling FIGURE 131.24. Leakage from neovascularization of the disk with
and prolonged venous transit time and continued asymmetric filling of late staining of the retinal vessels.
the choroid.

REFERENCES
1. von Graefe A: Uber embolie der arteria 14. Brown GC, Magargal LE: Central retinal echocardiography in the evaluation of
centralis retinae als ursache plötzlicher artery obstruction and visual acuity. patients with retinal artery occlusion.
erblingdung. Arch für Ophthalmology 1859; Ophthalmology 1982; 89:14–19. Ophthalmology 2001; 108:1461–1464.
5:136–157. 15. Arruga J, Sanders MD: Ophthalmologic 26. Wisotsky BJ, Engel HM: Transesophageal
2. Awan KJ: Arterial vascular anomalies of the findings in 70 patients with evidence of echocardiography in the diagnosis of
retina. Arch Ophthalmol 1977; 95:1197–1202. retinal embolism. Ophthalmology 1982; branch retinal artery obstruction. Am J
3. Brown GC, Shields JA: Cilioretinal arteries 89:1336–1347. Ophthalmol 1993; 115:653–656.
and retinal arterial occlusion. Arch 16. Brown GC, Magargal LE, Shields JA, et al: 27. Chuang EL, Miller FS 3rd, Kalina RE. Retinal
Ophthalmol 1979; 97:84–92. Retinal arterial obstruction in children and lesions following long bone fractures.
4. Wise GN, Dollery CT, Henkind P, et al: The young adults. Ophthalmology 1981; Ophthalmology 1985; 92:370–374.
retinal circulation. New York, NY: Harper & 88:18–25. 28. Chang M, Herbert WN: Retinal arteriolar
Row; 1971. 17. Kollarits CR, Lubow M, Hissong SL: Retinal occlusions following amniotic fluid
5. Singh SS, Dass R: The central artery of the strokes. I. Incidence of carotid atheromata. embolism. Ophthalmology 1984;
retina – II. Distribution and anastomoses. JAMA 1972; 222:1273–1275. 91:1634–1637.
Br J Ophthalmol 1960; 44:280–299. 18. Shah HG, Brown GC, Goldberg RE: Digital 29. Kim IT, Choi JB: Occlusions of branch
6. Hayreh SS, Weingeist TA: Experimental subtraction carotid angiography and retinal retinal arterioles following amniotic fluid
occlusion of the central retinal artery of the arterial obstruction. Ophthalmology 1985; embolism. Ophthalmologica 2000;
retina. IV. Retinal tolerance time to acute 92:68–72. 214:305–308.
ischemia. Br J Ophthalmol 1980; 19. Penner R, Font RL: Retinal embolism from 30. AtLee WE Jr: Talc and cornstarch emboli
64:896–912. calcified vegetations of aortic valve. in eyes of drug abusers. JAMA 1972;
7. Kroll AJ: Experimental central retinal artery Spontaneous complication of rheumatic 219:49–51.
occlusions. Arch Ophthalmol 1968; 79. heart disease. Arch Ophthalmol 1969; 31. Whiteman DW, Rosen DA, Pinkerton RM:
8. Hayreh SS, Zimmerman B: Central retinal 81:565–568. Retinal and choroidal microvascular
artery occlusion: visual outcome. Am J 20. Rush JA, Kearns TP, Danielson GK: Cloth- embolism after intranasal corticosteroid
Ophthalmol 2005; 140:376–391. particle retinal emboli from artificial cardiac injection. Am J Ophthalmol 1980;
9. Ozdemir H, Karacorlu S, Karacorlu M, et al: valves. Am J Ophthalmol 1980; 89:851–853.
Optical coherence tomography findings in 89:845–850. 32. Wilkinson WS, Morgan CM, Baruh E,
central retinal artery occlusion. Retina 21. Jampol LM, Wong AS, Albert DM: Atrial Gitter KA: Retinal and choroidal vascular
2006; 26:110–112. myxoma and central retinal artery occlusion secondary to corticosteroid
10. Dahrling BE: The histopathology of early occlusion. Am J Ophthalmol 1973; embolisation. Br J Ophthalmol 1989;
central retinal artery occlusion. Arch 75:242–249. 73:32–34.
Ophthalmol 1965; 73:506–510. 22. Zamora RL, Adelberg DA, Berger AS, et al: 33. Brown GC, Magargal L, Augsburger JJ,
11. Ashton N, et al: Focal retinal ischemia, Branch retinal artery occlusion caused by a Shields JA: Preretinal arterial loops and
ophthalmoscopic, circulatory and mitral valve papillary fibroelastoma. Am J retinal arterial occlusion. Am J Ophthalmol
ultrastructural changes. Br J Ophthalmol Ophthalmol 1995; 119:325–329. 1979; 87:646–651.
1966; 50:285–384. 23. Zamora RL, Adelberg DA, Berger AS, et al: 34. Seligsohn U, Griffin JH: Hereditary
12. Hayreh SS, Jonas JB: Optic disc and Branch retinal artery occlusion caused by a Thrombophilia.. In: Lichtman MA, et al, eds.
retinal nerve fiber layer damage after mitral valve papillary fibroelastoma – Williams Hematology. 7th edn. New York:
transient central retinal artery occlusion: an correction. Am J Ophthalmol 1995; 120:126. McGraw-Hill; 2005:1981–1996.
experimental study in rhesus monkeys. Am 24. Clifford L, Sievers R, Salmon A, Newsom 35. Bertram B, Remky A, Arend O, et al:
J Ophthalmol 2000; 129:786–795. RS: Central retinal artery occlusion: Protein C, protein S, and antithrombin III in
13. Hayreh SS, Karacorlu S, Karacorlu M, et al: association with patent foramen ovale. acute ocular occlusive diseases. Ger J
Central retinal artery occlusion and retinal Eye 2005. Ophthalmol 1995; 4:332–335.
tolerance time. Ophthalmology 1980; 25. Kramer M, Goldenberg-Cohen N, Shapira 36. Greven CM, Weaver RG, Owen J, Slusher
87:75–78. Y, et al: Role of transesophageal MM: Protein S deficiency and bilateral
1751
RETINA AND VITREOUS

branch retinal artery occlusion. with ocular Behcet’s disease. Eur J Lysis in the Eye, et al: Evaluation of
Ophthalmology 1991; 98:33–34. Ophthalmol 2002; 12:328–330. minimally invasive therapies and rational for
37. Greiner K, Hafner G, Dick B, et al: Retinal 53. Williamson TH, Meyer PA: Branch retinal a prospective randomized trial to
vascular occlusion and deficiencies in the artery occlusion in toxoplasma evaluateselective intra-arterial lysis for
protein C pathway. Am J Ophthalmol 1999; retinochoroiditis. Br J Ophthalmol 1991; clinically complete central retinal artery
128:69–74. 75:253. occlusion. Arch Ophthalmol 2003;
38. Tayyanipour R, Pulido JS, Postel EA, et al: 54. Morgan CM, Gragoudas ES: Branch retinal 121:1377–1381.
Arterial vascular occlusion associated with artery occlusion associated with recurrent 72. Atebara NH, Brown GC, Cater J, et al:
factor V Leiden gene mutation. Retina toxoplasmic retinochoroiditis. Arch Efficacy of anterior chamber paracentesis
1998; 18:376–377. Ophthalmol 1987; 105:130–131. and carbogen in treating acute nonarteritic
39. Dhar-Munshi S, Ayliffe WH, Jayne D: 55. Gray AV, Michels KS, Lauer AK, Samples central retinal artery occlusion.
Branch retinal arteriolar occlusion JR: Bartonella henselae infection Ophthalmology 1995; 102:2029–2035.
associated with familial factor V Leiden associated with neuroretinitis, central 73. Iwafune Y, Yoshimoto H: Clinical use of
polymorphism and positive rheumatoid retinal artery and vein occlusion, pentoxifylline in haemorrhagic disorders of
factor. Arch Ophthalmol 1999; neovascular glaucoma, and severe the retina. Pharmatherapeutica 1980;
117:971–973. vision loss. Am J Ophthalmol 2004; 2:429–438.
40. Weger M, Stanger O, Deutschmann H, 137:187–189. 74. Kuritzky S: Nitroglycerin to treat acute loss
et al: The role of hyperhomocysteinemia 56. Ormerod LD, Skolnick KA, Menosky MM, of vision. N Engl J Med 1990; 323:1428.
and methylenetetrahydrofolate reductase et al: Retinal and choroidal manifestations 75. Charness ME, Liu GT: Central retinal artery
(MTHFR) C677T mutation in patients with of cat-scratch disease. Ophthalmology occlusion in giant cell arteritis: treatment
retinal artery occlusion. Am J Ophthalmol 1998; 105:1024–1031. with nitroglycerin. Neurology 1991;
SECTION 10

2002; 134:57–61. 57. Katz B: Migrainous central retinal artery 41:1698–1699.


41. Pianka P, Almog Y, Man O, et al: occlusion. J Clin Neuroophthalmol 1986; 76. Crosson CE, Willis JA, Potter D, et al:
Hyperhomocystinemia in patients with 6:69–75. Effect of the calcium antagonist, nifedipine,
nonarteritic anterior ischemic optic 58. Beversdorf D, Stommel E, Allen C, et al: on ischemic retinal dysfunction. J Ocul
neuropathy, central retinal artery occlusion, Recurrent branch retinal infarcts in Pharmacol 1990; 6:293–299.
and central retinal vein occlusion. association with migraine. Headache 1997; 77. Frayser R, Hickam JB, et al: Retinal
Ophthalmology 2000; 107:1588–1592. 37:396–399. vascular response to breathing increased
42. Wilson RS, Ruiz RS: Bilateral central 59. Clarke WN, Vomiero G, Leonard BC: carbon dioxide and oxygen concentrations.
retinal artery occlusion in homocystinuria. Bilateral simultaneous retinal arteriolar Invest Ophthalmol 1964; 3:427–431.
A case report. Arch Ophthalmol 1969; obstruction in a child with hemoglobin SS 78. Hayreh SS: Retinal artery occlusion with
82:267–268. sickle cell disease. J Aapos 2001; LIF using rTPA. Ophthalmology 1999;
43. Rand JH, Senzel L: The Antiphospholipid 5:126–128. 106:1236–1238.
Syndrome.. In: Lichtman MA, et al, eds. 60. Al-Abdulla NA, Haddock TA, Kerrison JB, 79. Richard G, Lerche RC, Krospe V, et al:
Williams Hematology. 7th edn. New York: Goldberg MF: Sickle cell disease Treatment of retinal arterial occlusion with
McGraw-Hill; 2005:2009–2027. presenting with extensive peri-macular local fibrinolysis using recombinant tissue
44. Dori D, Beiran I, Gelfand Y, et al: Multiple arteriolar occlusions in a nine-year-old boy. plasminogen activator. Ophthalmology
retinal arteriolar occlusions associated with Am J Ophthalmol 2001; 131:275–276. 1999; 106:768–773.
coexisting primary antiphospholipid 61. Fine LC, Petrovic VV, Irvine AR, Bhisitkul 80. Richard G: Retinal artery occlusion with LIF
syndrome and factor V Leiden mutation. RB: Correction-spontaneous central retinal using rTPA. Ophthalmology 1999;
Am J Ophthalmol 2000; 129:106–108. artery occlusion in hemoglobin SC disease. 106:1238–1239.
45. Gold D, Feiner L, Henkind P: Retinal arterial Am J Ophthalmol 2000; 130:906–907. 81. Kattah JC, Wang DZ, Reddy C, et al:
occlusive disease in systemic lupus 62. Kachmaryk MM, Trimble SN, Gieser RG: Intravenous recombinant tissue-type
erythematosus. Arch Ophthalmol 1977; Cilioretinal artery occlusion in sickle cell plasminogen activator thrombolysis in
95:1580–1585. trait and rheumatoid arthritis. Retina 1995; treatment of central retinal artery occlusion.
46. Levine SR, Deegan MJ, Futrell N, Welch 15:501–504. Arch Ophthalmol 2002; 120:1234–1236.
KM: Cerebrovascular and neurologic 63. Gass JD, Tiedeman J, Thomas MA: 82. Beatty S, AuEong AK: Local intra-arterial
disease associated with antiphospholipid Idiopathic recurrent branch retinal arterial fibrinolysis for acute occlusion of the
antibodies: 48 cases. Neurology 1990; occlusion. Ophthalmology 1986; central retinal artery: a meta-analysis of
40:1181–1189. 93:1148–1157. published data. Br J Ophthalmol 2000;
47. Snyers B, Lambert M, Hardy JP: Retinal 64. Johnson MW, Thomley ML, Huang SS, 84:914–916.
and choroidal vaso-occlusive disease in Gass JD: Idiopathic recurrent branch retinal 83. Ciulla TA, D’Amico DJ, Miller JW:
systemic lupus erythematosus associated arterial occlusion. Natural history and Laser photodisruption of visible retinal
with antiphospholipid antibodies. Retina laboratory evaluation. Ophthalmology 1994; artery emboli. Br J Ophthalmol 1995;
1990; 10:255–260. 101:480–489. 79:964–965.
48. Singh R, Conway MD, Wilson WA: 65. Newman NJ, Lessell S, Brandt EM: 84. Peyman GA: Removal of emboli from the
Antiphospholipid syndrome in a patient Bilateral central retinal artery occlusions, branches of the central retinal artery. Arch
with sarcoidosis: a case report. Lupus disk drusen, and migraine. Am J Ophthalmol 2001; 119:1224–1225.
2002; 11:756–758. Ophthalmol 1989; 107:236–240. 85. Sharma S, Sharma SM, Cruess AF, Brown
49. Cobo-Soriano R, Sanchez-Ramon S, 66. Ros MA, Magargal LE, Uram M: Branch GC: Transthoracic echocardiography in
Aparicio MJ, et al: Antiphospholipid retinal-artery obstruction: a review of 201 young patients with acute retinal arterial
antibodies and retinal thrombosis in eyes. Ann Ophthalmol 1989; 21:103–107. obstruction. RECO Study Group. Retinal
patients without risk factors: a prospective 67. Leavitt JM: Occlusion of the cilioretinal Emboli of Cardiac Origin Group. Can J
case-control study. Am J Ophthalmol 1999; artery. Arch Ophthalmol 1948; 40:152–156. Ophthalmol 1997; 32:38–41.
128:725–732. 68. Brown GC, Moffat K, Cruess A, et al: 86. Sharma S: The systemic evaluation of
50. Glacet-Bernard A, Bayani N, Chretien P, Cilioretinal artery obstruction. Retina 1983; acute retinal artery occlusion. Curr Opin
et al: Antiphospholipid antibodies in retinal 3:182–187. Ophthalmol 1998; 9:1–5.
vascular occlusions. A prospective study of 69. Keyser BJ, Duker JS, Brown GC, et al: 87. Greven CM: Retinal arterial occlusions in the
75 patients. Arch Ophthalmol 1994; Combined central retinal vein occlusion young. Curr Opin Ophthalmol 1997; 8:3–7.
112:790–795. and cilioretinal artery occlusion associated 88. Greven CM, Slusher MM, Weaver RG:
51. Fineman MS, Savino PJ, Federman JL, with prolonged retinal arterial filling. Am J Retinal arterial occlusions in young adults.
Eagle RC, Jr: Branch retinal artery Ophthalmol 1994; 117:308–313. Am J Ophthalmol 1995; 120:776–783.
occlusion as the initial sign of giant cell 70. Rumelt S, Brown GC: Update on treatment 89. Duker JS, Sivalingam A, Brown GC, Reber R:
arteritis. Am J Ophthalmol 1996; of retinal arterial occlusions. Curr Opin A prospective study of acute central retinal
122:428–430. Ophthalmol 2003; 14:139–141. artery obstruction. The incidence of
52. Ozdal PC, Ortac S, Taskintuna I, et al: 71. Mueller AJ, Neubauer AS, Schaller U, secondary ocular neovascularization. Arch
Central retinal artery occlusion associated Kampik A: European Assessment Group for Ophthalmol 1991; 109:339–342.
1752
Retinal Arterial Occlusions

90. Hayreh SS, Podhajsky P: Ocular 93. Brown GC: The ocular ischemic syndrome. 96. Beneficial effect of carotid endarterectomy
neovascularization with retinal vascular In: Ryan SJ. Retina. St Louis, MO: Mosby; in symptomatic patients with high-grade
occlusion. II. Occurrence in central and 1989:547–559. carotid stenosis. North American
branch retinal artery occlusion. Arch 94. Brown GC, Magargal LE: The ocular symptomatic carotid endarterectomy trial
Ophthalmol 1982; 100:1585–1596. ischemic syndrome. Int Ophthalmol 1988; collaborators. N Engl J Med 1991;
91. Mizener JB, Podhajsky P, Hayreh SS: 11:239–251. 325:445–453.
Ocular ischemic syndrome. Ophthalmology 95. Sivalingam A, Brown GC, Magargal LE,
1997; 104:859–864. Menduke H: The ocular ischemic
92. Wessing A: Fluorescein angiography of the syndrome. II. Mortality and systemic
retina; textbook and atlas. St Louis, MO: morbidity. Int Ophthalmol 1989;
Mosby; 1969:xii, 222. 13:187–191.

CHAPTER 131

1753
CHAPTER

132 Retinal Venous Occlusive Disease


Jonathan Gunther, Ingrid U. Scott, and Michael Ip

INTRODUCTION 11.9% in the fellow eye. Of all the RVO in the BMES, 69.5%
were BRVO, 25% CRVO, 5.1% HRVO, and 5.1% were bilateral.
Retinal vein occlusion (RVO) is the second most common
retinal vascular disease following diabetic retinopathy.1 There BRANCH RETINAL VEIN OCCLUSION
are three distinct types of RVO: branch retinal vein occlusion
(BRVO), central retinal vein occlusion (CRVO), and an
anatomical variant of CRVO, namely, hemiretinal vein occlu- Synonym
sion (HRVO) (Table 132.1). Retinal venous occlusive disease • Branch vein occlusion
typically occurs at an arteriovenous crossing in BRVO or at the
lamina cribrosa in CRVO and HRVO. Retinal vein occlusions
have a characteristic, although somewhat variable, appearance Key Features
with intraretinal hemorrhage, cotton-wool spots, tortuous and • Segmental retinal involvement in the distribution of the
dilated retinal veins, retinal edema and, occasionally, optic disk involved occluded vein
swelling. These findings are present segmentally in BRVO, in • Dilated, tortuous veins
either the superior or inferior two quadrants in HRVO, and in • Superficial and deep retinal hemorrhages
all quadrants of the fundus in CRVO. Vision loss can vary from • Cotton-wool spots
minimal or no vision loss to complete blindness. Causes of • Optic nerve swelling/hemorrhages
vision loss associated with RVO include macular edema, • Macular/retinal edema
macular nonperfusion, epiretinal membrane, dense intraretinal • Optociliary shunt vessels on disk
hemorrhages, vitreous hemorrhage, neovascular glaucoma, or • Neovascularization of the retina (less common: disk, iris)
tractional retinal detachment (TRD).2–6 Timely intervention
can reduce the incidence and severity of these complications.
Many systemic diseases are risk factors for RVO development. INTRODUCTION
Recognition and treatment of an underlying systemic disease
can benefit the patient visually and improve overall health. The fundoscopic findings of BRVO are similar to those of
The overall incidence of RVO in population-based studies CRVO; however, the extent of retinal involvement is typically
varies depending on age and the study. In a large population- much less. The involved vein takes on a dilated and tortuous
based study in Israel, the 4 year incidence of retinal vein occlu- appearance, deep and superficial retinal hemorrhages form,
sion was 2.14/1000 among persons aged 40 years and older, and cotton-wool spots can be observed, and retinal edema may
5.36/1000 among individuals older than 64 years.7 In the Blue develop. A BRVO typically occurs at the crossing of a retinal
Mountains Eye Study8 (BMES) in Australia, the prevalence of artery and vein. The involved area is usually wedge-shaped with
retinal vein occlusion was 0.7% among persons aged 49–60 the apex near the site of the occlusion, extending outward to the
years, 1.2% for persons aged 60–69 years, 2.1% for those 70–79 periphery (Fig. 132.1). The area drained by the involved vein
years, and 4.6% for persons 80 years and older. The mean age defines the extent of retinal involvement. The greater the area
of onset of vein occlusion was 63 years. Hayreh9 reported that of retinal involvement, the greater the impact it is likely to have
after any RVO in one eye, the incidence of developing a second on vision. The location of the occlusion is an important factor
vein occlusion in the next 4 years was 2.5% in the same eye and in the visual prognosis. In general, the closer the occlusion
occurs to the optic nerve, the more extensive the retinal damage
and visual impact.10–12 The clinical course and visual outcome
of BRVO is highly variable.
TABLE 132.1. Key Characteristics of Vein Occlusions

RVO type Occlusion location Area involved Neovascular EPIDEMIOLOGY


location
BRVO is more than twice as common as CRVO and more than
BRVO Retinal A–V crossing Segmental Posterior pole 10 times as common as HRVO.7,8,13,14 Numerous studies have
CRVO Lamina cribrosa 4-Quadrant Anterior assessed the incidence of venous occlusive diseases over the
segment years. The Beaver Dam Eye Study13 was a population-based
HRVO Lamina cribrosa 2-Quadrant Post. Pole>ant. study of 4926 residents of Beaver Dam, WI aged 43–84 years.
(vertical) segment Initial RVO evaluation was performed between 1988 and 1990
with follow-up examinations 5 years later (1993–1995). Diag- 1755
RETINA AND VITREOUS

Another theory is that venous compression is not the cause


of BRVO. Seitz demonstrated histologically that the retinal vein
is not compressed by the artery at intersections at which
nicking is observed.28 Jefferies found that the vein took a much
more pronounced deviation around an artery when the artery
overlies the vein, compared to when the vein overlies the artery.
He also found focal stratification of the venous endothelial base-
ment membrane opposite the point of contact with the artery.
Localized venous luminal narrowing was not typical. Jefferies
proposed that hemodynamic variations due to venous con-
touring are responsible for the much higher risk in artery over
vein intersections.29
It has been suggested that BRVO is the result of arterial
insufficiency.24,25 However, in a histopathologic study of nine
cases of BRVO, Franghieh demonstrated that all cases had fresh
or recanalized venous thrombus at the site of occlusion. He con-
cluded that branch vein thrombosis was likely the primary event
and that the other vascular findings occurred secondarily.30 Other
SECTION 10

studies have demonstrated that the manifestations of arterial


FIGURE 132.1. Large superotemporal branch retinal vein occlusion insufficiency can be reproduced in animals by experimental
(BRVO) occurring at an arteriovenous intersection near the superior occlusion of the retinal veins.31–36
margin of the disc. The average age of patients with BRVO is between 60 and
70 years.7,8,13,14 The incidence increases with age.8 Certain
systemic diseases have also been shown to be risk factors for
nosis of RVO was made by grading 30° stereo fundus photo- BRVO. The Eye Disease Case-Control Study Group reported
graphs. The prevalence and 5 year incidence of BRVO were each ‘cardiovascular risk profile’ as a risk factor for BRVO.37,38 A
0.6%. Prevalence increased with age and there was no gender significantly increased risk was found for systemic hyperten-
predilection. sion, history of cardiovascular disease, smoking, increased body
The Blue Mountain Eye Study8 in Australia was a prospective mass index and higher serum levels of alpha2-globulin. One-
study including 3654 participants aged 49 years or older. half to two-thirds of patients in large series of BRVO have been
Follow-up consisted of detailed eye examinations including reported to have systemic hypertension.10–12,20,39,40 A decreased
stereoscopic fundus photography. Diagnosis of RVO was based risk of BRVO was associated with alcohol consumption and
on clinical findings and fundus photographic grading. The higher levels of high density lipoprotein (HDL) cholesterol.
overall incidence of BRVO was 1.1% with a strong age-related Diabetes mellitus and open angle glaucoma occurred more
predilection and no gender predilection. frequently among BRVO patients than among controls, but the
differences were not statistically significant.41
PATHOPHYSIOLOGY
BRVO is defined as a focal occlusion of a retinal vein.15–17 With CLINICAL FEATURES
rare exceptions, such as in association with sarcoidosis and Patients with BRVO may complain of painless, decreased vision,
other ocular inflammatory disorders, BRVO develops at arterio- complete loss of vision, or a blind spot in their visual field.
venous crossing sites where the artery passes anteriorly Others may be asymptomatic. Vision loss in BRVO may be due
(superficially) to the vein.16–19 In normal eyes, retinal arteries to macular edema, macular nonperfusion, dense intraretinal
cross over retinal veins in 70–75% of the intersections.15,17 The hemorrhages, vitreous hemorrhage, neovascular glaucoma,
most common location of a BRVO is the superotemporal epiretinal membrane, or tractional retinal detachment.2,42
quadrant of the retina. This may be due to a higher risk of A complete ocular examination should be performed,
occlusion in this area, or may be due to increased diagnosis due including evaluation for iris or angle neovascularization and a
to increased symptoms of a temporal versus a nasal occlusion. dilated fundus examination to evaluate for macular edema,
Most BRVOs involve the area inside the temporal vascular neovascularization of disk and/or retina, presence of intraretinal
arcades (macular BRVO), whereas peripheral BRVOs are hemorrhages, cotton wool spots, vitreous hemorrhage,
observed less commonly, likely because they tend to be less narrowing and sheathing of the adjacent artery. Close attention
symptomatic. Of temporal BRVOs, ~62% occur in the should be paid to the distribution of the involved area of the
superotemporal quadrant and 38% in the inferotemporal fundus.
quadrant.10,11,15,17,20–23 Visual acuity is generally less affected in BRVO when com-
The precise mechanism causing the venous occlusion is pared to CRVO. However, there can be great variation in visual
poorly understood. Multiple theories, often contradictory, exist. outcome ranging from no visual impairment to severe vision
Histological examination has shown that BRVO is associated loss. Typically, patients presenting with visual complaints have
with arteriosclerotic changes in the retinal arterioles.17 The associated retinal hemorrhage, edema, and/or nonperfusion.
resultant thickening of the artery could cause compression of Symptoms may worsen or improve with time. At later stages,
the adjacent vein, a process that may be aggravated because, at visual complications may include macular edema, vitreous
arteriovenous crossing sites, the two vessels are confined within hemorrhage from neovascularization, epiretinal membrane, or
a common adventitial sheath.24 With increased compression, retinal detachment. Generally, the visual prognosis of BRVO is
venous blood flow velocity at the crossing site may gradually quite good. Approximately 50–60% of untreated patients will
increase until local shear stress causes endothelial cell loss, have a final visual acuity of 20/40 or better. However, severe
thrombus formation, and vein occlusion.24–26 Theoretically, vision loss is not uncommon, with 20–25% of patients having
shear stress is maximal when the velocity is high enough to a visual acuity of 20/200 or worse. A final visual acuity of hand
1756 induce transition to a turbulent flow pattern.27 motion or worse is uncommon.2,10,11,20,42
Retinal Venous Occlusive Disease

The classic fundus findings of BRVO are similar to CRVO, associated with BRVO.10,46 In patients with perfused macular
although the area of distribution is distinct. BRVO fundus find- edema, approximately one-third will regain some vision spon-
ings may include optic disk swelling, retinal swelling, including taneously. However, this becomes less likely the longer the
the macula, markedly dilated and tortuous retinal veins, super- edema persists. Spontaneous visual improvement is less com-
ficial and deep retinal hemorrhages radiating outward from the mon in nonperfused macular edema. Interleukin-6 (IL-6) and
optic disk, and cotton-wool spots (Fig. 132.2). However, a small vascular endothelial growth factor (VEGF) have been implicated
or peripheral BRVO may produce findings that are much more in the development of macular edema following nonperfused
subtle and difficult to detect on examination. With time, the BRVO.47
fundus findings may become less distinct and appear similar to Other microvascular changes result in dilated capillaries,
what one would expect to see in diabetic retinopathy, hyper- causing formation of microaneurysms and dot-blot hemor-
tensive retinopathy, or other systemic diseases causing retinal rhages.48,49 Retinal vessels may become fibrosed leading to
microaneurysms and hemorrhages. Clues suggestive of an old sheathing. Collateral vessels may develop between an area of
BRVO include segmental microvascular abnormalities and nonperfused and an area of perfused retina. Collateral vessels
intraretinal collateral vessels draining across the median raphe. appear as small, tortuous venous channels that cross the hori-
In nonperfused cases, sclerosis and sheathing of the retinal zontal raphe and drain into an uninvolved venous circulation
veins and arteries in the distribution of the occlusion may be (Fig. 132.4). Collateral vessels are distinct from neovascular
observed.43,44 vessels in that collaterals are flat while neovascular vessels are
Intravenous FA can assist in evaluating BRVO. Angiographic elevated; the former do not leak on FA while the latter leak.

CHAPTER 132
findings reflect changes in vessel permeability, caliber, and Retinal hemorrhage is one of the most striking features of
patency and assist in identifying areas of macular edema, BRVO. Intraretinal hemorrhages located within the macula can
neovascularization, and nonperfusion. Vitreous or retinal
hemorrhage may impair the utility of FA. Venous filling delay is
often noted in the area drained by the occluded vein. The
venous tributaries often appear narrowed. Occasionally, early
hyperfluorescence just proximal to the site of occlusion is
observed.10,45 Retinal edema often appears as diffuse hyper-
fluorescence on angiography. The edema is commonly found
within the area of distribution of the occluded vein. Macular
edema often takes on a petalloid pattern typical of cystoid
macular edema. When evaluating for macular edema, it is
important to note whether capillary perfusion is present. Retinal
nonperfusion appears on fluorescein angiography (FA), as areas
of capillary hypofluorescence. Neovascularization of the retina
or disk appear as early hyperfluorescent, thin, tortuous vessels
that leak in the later stages of testing.

COMPLICATIONS
Microvascular changes result in numerous findings. Leaking
vessels lead to retinal edema and deposition of hard exudates.
Retinal edema becomes much more visually significant when it FIGURE 132.3. A small inferotemporal BRVO. Although the area of
involves the fovea (Fig. 132.3). Macular edema is the most affected retina is small, the hemorrhage and edema extend into the
common complication, and leading cause of vision loss, fovea and are associated with reduced vision.

FIGURE 132.4. Appearance of venous collaterals after a BRVO. The


FIGURE 132.2. BRVO with hemorrhage, cotton-wool spots, and channels cross the median raphe temporal to the fovea and drain
fibrous sheathing of the retinal veins. from the affected to the nonaffected quadrant. 1757
SECTION 10 RETINA AND VITREOUS

FIGURE 132.5. Fluorescein angiogram of a nonperfused BRVO


demonstrates widespread capillary nonperfusion and staining of the FIGURE 132.6. A tuft of retinal neovascularization (arrow) along the
retinal veins. The two areas of intense hyperfluorescence (arrows) are vascular arcade in an eye with long-standing inferotemporal BRVO.
due to retinal neovascularization.

cause severe vision loss. Vision often will improve as the response to the release of angiogenic factors (such as VEGF and
hemorrhage resolves. Retinal hemorrhages typically resolve over IL-6) from nonperfused retina. Retinal neovascularization
weeks to months, but can persist for years. Hemorrhage blocks typically forms in the first 6–12 months following a BRVO;
fluorescence on angiography and can impair the usefulness of however, it may develop years later also.51
such evaluation. Vitreous hemorrhage, if it occurs, is typically Tractional retinal detachment (TRD) can form following
observed later in the course of BRVO. Vitreous hemorrhage may BRVO if fibrovascular proliferation develops. Rhegmatogenous
develop after rupture of thin, friable neovascular vessels that retinal detachments are a rare complication of BRVO. They
grow in response to retinal nonperfusion. The majority of eyes typically form following posterior retinal breaks caused by
with retinal or disk neovascularization will develop vitreous fibrovascular proliferation and traction.52–55 Nonperfused
hemorrhage if left untreated.10,11,39,42 retina can lead to degeneration and retinal hole formation
Nonperfusion can lead to both early and late complications of following BRVO.55–58 Exudative retinal detachments can occur
BRVO. Early in the course of BRVO, nonperfusion can result in in the area of occlusion and are usually associated with
permanent vision loss. The closer the nonperfusion is to the nonperfusion.54,59,60
macula, the more significant the visual loss will be. Large areas Other visually significant complications of BRVO include
of persistent retinal nonperfusion can lead to neovasculariza- epiretinal membrane formation, retinal pigment epithelial
tion of the retina or disk (Fig. 132.5). It is important to recog- irregularity, and subretinal scarring.54,59,60
nize that perfused BRVO may progress to the nonperfused type
during the ensuing weeks, months and even years after the
initial insult.39,42,44,50 DIFFERENTIAL DIAGNOSIS
The most common site of neovascularization following Diabetic retinopathy can present with findings similar to BRVO
BRVO is the retina (Fig. 132.6). Optic disk neovascularization is with dot-blot hemorrhages, microaneurysms, and neovascu-
much less common and iris neovascularization is rare in BRVO. larization. In diabetic retinopathy, the retinal findings are
When disk neovascularization does occur, retinal neovascu- generally bilateral and involve all four quadrants. In BRVO, the
larization typically is present as well.39 Numerous studies have retinal hemorrhages are almost always unilateral, except in
reported a 20–30% incidence of retinal neovascularization the infrequent case of a bilateral BRVO. Also, the retinal
following BRVO.10,20,42,51 The Branch Vein Occlusion Study hemorrhages involve only the sector of retina that is drained by
(BVOS)42 was a multicentric, randomized, controlled clinical the occluded vein.
trial designed to answer questions regarding the management Hypertensive retinopathy is another disease that must be
of complications of branch vein occlusion. These questions considered in the differential diagnosis of BRVO. Distinguishing
included: ‘Can peripheral scatter argon laser photocoagulation characteristics of hypertensive retinopathy include bilateral
prevent the development of neovascularization?’, and ‘Can narrowed retinal arterioles and hemorrhages that involve the
peripheral scatter argon laser photocoagulation prevent vitreous entire fundus.
hemorrhage?’ In the BVOS, retinal neovascularization developed Radiation retinopathy may be unilateral or bilateral. A
in 22% of all eyes with BRVO. The incidence of retinal neo- history of irradiation is essential for the diagnosis. Similar to
vascularization increased to 36% in eyes with five disk- BRVO, disk swelling, disk hemorrhages, retinal neovascu-
diameters or more of retinal nonperfusion. Shilling and Kohner larization, and cotton-wool spots may be present.
reported a 62% incidence of neovascularization among eyes
with greater than four disk-diameters of nonperfusion and 0%
among eyes with less nonperfusion.39 Retinal neovascu- TREATMENT
larization typically develops at the border between perfused and The treatment of BRVO is continually evolving. Available data
nonperfused retina; it rarely develops outside the area of concerning many of the reported treatment modalities are based
1758 nonperfusion. Neovascular vessels are believed to form in on case series without controls or randomization. As our
Retinal Venous Occlusive Disease

understanding of the disease improves, the standard of care is present, higher laser powers may be necessary for adequate
likely to continue to evolve. uptake.
Typical follow-up is approximately 8–12 weeks after treat-
Medical Treatment ment. If macular edema persists and the acuity and other
Evaluation should include a thorough medical history, clinical findings have not improved, repeat FA is suggested. If
including past ocular history, review of current medications, residual fluorescein leakage is observed, another session of grid
and a thorough review of systems. The most common reported laser photocoagulation may be indicated.63
risk factors associated with BRVO are systemic hypertension,
diabetes, hyperlipidemia, glaucoma, smoking, and age-related Scatter Laser Photocoagulation
atherosclerosis.10–12,20,39–41 Identifying and treating individual Retinal and disk neovascularization can lead to visually com-
risk factors are important in the early treatment and prevention promising vitreous hemorrhage. The BVOS showed that scatter
of future RVO. Systemic evaluation of commonly associated risk sectoral laser photocoagulation reduces the risk of developing
factors is an important part of the examination process. retinal neovascularization from 22% to 12%. However, the
Extensive work-up following BRVO is typically not necessary in study concluded that treatment should be reserved until after
older individuals as the common risk factors are often elicited the development of retinal neovascularization. In eyes with
on basic screening. However, in patients under age 50 years or retinal neovascularization, subsequent laser photocoagulation
with bilateral vein occlusions, a more thorough work-up may be reduces the risk of vitreous hemorrhage from 60% to 30%.2 If
warranted. The primary care physician should be notified when vitreous hemorrhage is already present, staged sessions of

CHAPTER 132
a patient is diagnosed with a BRVO. Initial evaluation may scatter laser photocoagulation may be necessary, or peripheral
include testing for hypertension, diabetes, and a hyper- cryoablation may be performed. Scatter photocoagulation is not
coagulable state. Initial testing may include a fasting blood without its own set of complications. Most notably, some
glucose level, complete blood count with differential and plate- patients report peripheral scotoma following scatter photo-
lets, coagulation studies, and erythrocyte sedimentation rate. coagulation,64 and this has been confirmed with visual field
Systemic anticoagulation has not been shown to be of ocular testing showing peripheral visual field loss.65
benefit in patients with BRVO and may potentially worsen con- Commonly, scatter laser photocoagulation is applied with the
current or future intraretinal hemorrhage.61 Further argon green laser to achieve ‘medium’ white burns (200–500 mm
cardiovascular disease evaluation may be needed; however, the in diameter) spaced one burn width apart and covering the
primary care physician often is helpful in coordinating this. entire area of capillary nonperfusion, as defined by fluorescein
Follow-up generally consists of ophthalmologic evaluation every angiogram. Treatment should extend no closer than two disk-
1–3 months for the first year, followed by every 3–12 months diameters from the center of the fovea.
thereafter.
Conventional Pars Plana Vitrectomy for
Grid Pattern Laser Photocoagulation Complications of BRVO
In 1984, the BVOS2 reported the results of a randomized trial of Various studies have shown favorable surgical result in subjects
no treatment versus grid laser photocoagulation for treatment who undergo vitrectomy for complications of BRVO.57,59,60,66–70
of macular edema associated with BRVO in patients with a Surgical indications include vitreous hemorrhage, TRD involv-
visual acuity of 20/40 to 20/200. Patients with distinct areas of ing the macula, epiretinal membrane, removal of loculated
macular capillary nonperfusion were excluded. At 3 years, 65% preretinal subhyaloid hemorrhage, removal of subfoveal laser-
of treated eyes gained two or more lines of visual acuity induced choroidal neovascularization, and complications of
compared to 37% of untreated eyes. The mean visual acuity ghost cell glaucoma.
improvement was 1.3 ETDRS lines in the treated group versus Several preoperative clinical features are associated with
0.2 lines in the untreated group. Overall, mean visual acuity in better postoperative visual acuity outcome, including better pre-
the treatment group was 20/40 to 20/50, compared to 20/70 operative visual acuity, absence of an afferent pupillary defect,
in the untreated group. and absence of macular edema. The presence of disk neovascu-
Although it is not understood how laser treatment to the larization, retinal tear, or retinal detachment are associated with
retinal pigment epithelium reduces retinal edema, experimental a less favorable outcome.67
studies in the normal primate have shown a decrease in
capillary diameter when this form of therapy is administered.62 Pars Plana Vitrectomy Alone for Macular Edema
Grid treatment may produce retinal thinning that permits the Pars plana vitrectomy (PPV) with the creation of a posterior
choroidal vasculature to provide oxygen to the inner retina; this vitreous detachment (PVD) has been reported to be effective in
may lead to autoregulatory constriction of the retinal vascu- reducing macular edema and improving visual acuity, although
lature, which may result in decreased macular edema. the mechanisms that contribute to this have not been
Grid pattern photocoagulation is typically delayed for at least elucidated.71,72 It may not be intuitive that vitrectomy with
3 months after the development of a BRVO to allow for posterior hyaloid removal alone would improve macular edema,
spontaneous resolution of edema and intraretinal blood. Treat- because macular traction is uncommon in eyes with BRVO.73
ment may be considered in patients with a visual acuity of However, it has been suggested that removal of the posterior
20/40 or worse in the absence of capillary nonperfusion. Grid hyaloid by vitrectomy may improve oxygenation of the retina
laser is applied to the area of macular edema. Suggested treat- resulting in vasoconstriction and thus decreased vascular
ment parameters include argon green laser with a spot size of leakage and macular edema.74 Another theory postulates that
50–100 mm, 0.1 second duration and a power setting sufficient vitrectomy may facilitate diffusion of harmful cytokines that
to produce a light to medium white burn. The laser should be promote increased vascular permeability, such as VEGF, away
focused on the edematous retina within the arcades and the from the retina.
areas of perifoveal capillary leakage as identified by FA. Laser
photocoagulation should not be placed over substantial intra- Arteriovenous Adventitial Sheathotomy
retinal hemorrhage, as the nerve fiber layer may be damaged, Most BRVOs are believed to occur at an arteriovenous crossing
producing preretinal fibrosis. The fovea should be avoided in site, where the arteriole and venule share a common adventitial
the treatment process. When significant intraretinal edema is sheath. Arteriovenous adventitial sheathotomy (AAS) is an 1759
RETINA AND VITREOUS

attempt to decompress the involved venule by separating the The Standard Care versus COrticosteroid for REtinal Vein
overlying retinal artery from the underlying branch vein by Occlusion (SCORE) study is a multicenter, randomized, phase
sectioning the shared adventitial sheath. Patients who have III National Eye Institute-sponsored study that is investigating
macular edema recalcitrant to grid laser photocoagulation may the efficacy and safety of standard care versus intravitreal
be candidates for AAS.75 injection(s) of triamcinolone for macular edema secondary to
Numerous nonrandomized studies have demonstrated BRVO and CRVO.111 Individuals with BRVO or CRVO with
improved visual acuity, reduction in intraretinal hemorrhage, associated macular edema of up to 24 months duration and
return of retinal perfusion and reduction of macular edema after best-corrected visual acuity between 19 and 73 ETDRS letters
AAS surgery.76–96 In one such series, Mason et al79 reported a (corresponding to approximately 20/40 to 20/400 Snellen visual
prospective, nonrandomized interventional trial of 40 eyes with acuity) are eligible for participation in the SCORE study.
decreased visual acuity secondary to BRVO. Twenty of the eyes Another prospective randomized study (Posurdex Trial) is
underwent vitrectomy and surgical decompression by means of underway to evaluate treatment with extended delivery of a
arteriovenous sheathotomy with a microvitreoretinal blade and bioerodable dexamethasone PLGA (polylactic acid polyglycolic
were compared with 20 control eyes (10 observation and 10 grid acid) copolymer complex in patients with RVO.
laser photocoagulation at a mean of 9 months after diagnosis).
The mean preoperative visual acuity was 20/250 in the surgical Isovolemic Hemodilution
group and 20/180 in the control group (statistically similar BRVO has been reported to be associated with hyperviscosity,
in the observation group and laser-treated group). The mean due to higher hematocrit and plasma viscosity.112 Higher blood
SECTION 10

14 month visual acuity was 20/63 in the surgical group and viscosity is less important when blood flow is rapid but in
20/125 in the control group (P = 0.02). Seventy-five percent conditions of low flow, as is likely in a vein predisposed to
of the surgical group halved their visual angle compared with occlusion, the effect of viscosity becomes increasingly signi-
40% of the control group (40% observation versus. 40% laser; ficant as a result of increased red cell aggregation. Viscosity is
P = 0.025). Average lines of visual acuity gained were 4.55 in dependent primarily upon the hematocrit (the greater the
the surgical group and 1.55 in the control group (P = 0.0226; number of red cells, the larger the aggregates) and plasma
1.1 lines in observation group and laser-treated group). Reported fibrinogen (required for aggregation to occur). Studies have demon-
complications include retinal gliosis at the incision site, nerve strated that hypervolemic113 or isovolemic hemodilution,114 com-
fiber layer defects, vitreous hemorrhage, retinal tears, retinal menced within 3 months of the onset of symptoms of a BRVO,
detachment and acceleration of nuclear sclerosis.95 It is accelerates the rate of visual recovery and also has a positive
important to recognize that AAS has not been investigated in a effect on the final visual acuity at 1 year. However, reported
large-scale, prospective, randomized controlled clinical trial. complications of hemodilution include deep-vein thrombosis
and hypotension.115
Intraocular Corticosteroids
Corticosteroids have been proposed for the management of Antivascular Endothelial Growth Factor
macular edema due to various retinal vascular disorders. Intra- (Anti-VEGF)
vitreal triamcinolone acetonide has been investigated for per- Experimental studies have shown that functional and structural
sistent macular edema in RVO, diabetic retinopathy, chronic changes occur in the retinal capillaries induced by the hypoxic
uveitis, proliferative vitreoretinopathy, and postsurgical cystoid environment after RVO. This can lead to increased capillary
macular edema.97–100 The mechanism of action of corti- permeability and accompanying retinal edema. This appears to
costeroids in the treatment of macular edema in RVO is not be mediated, at least in part, by VEGF, a 45 kDa glycoprotein.35
entirely clear. Experimental studies have shown that functional Intravitreal VEGF levels have shown to be increased in patients
and structural changes occur in the retinal capillaries induced with macular edema with BRVO and are correlated significantly
by the hypoxic environment after venous occlusion, leading to with the area of nonperfusion and the severity of macular
increased capillary permeability and accompanying retinal edema.116 The role of anti-VEGF agents in the treatment in
edema possibly mediated in part by VEGF, a 45kDa BRVO has yet to be determined.
glycoprotein.35 Triamcinolone has been shown experimentally
to reduce the breakdown of the blood–retinal barrier by inhibiting CENTRAL RETINAL VEIN OCCLUSION
such factors as prostaglandins, interleukins, VEGF and protein
kinase C.101,102
Small case studies have suggested that there may be a benefit Synonym
from intravitreal injection of steroids for macular edema and • Central vein occlusion
vision loss associated with BRVO.103–109 Some patients with
BRVO may have a favorable anatomical response to this treat-
ment, as demonstrated by optical coherence tomographic (OCT) Key Features
images, measurements demonstrating reduction in macular • 4 quadrant retinal involvement
thickness, and resolution of the large cystic spaces in the outer • Dilated, tortuous veins
plexiform layer within several weeks of injection. However, a • Superficial and deep retinal hemorrhages
favorable visual acuity response may be more likely in patients • Cotton-wool spots
with perfused rather than nonperfused macular edema. Re- • Optic nerve swelling/hemorrhages
treatment may also be performed in some patients due to • Macular/retinal swelling
recurrent macular edema. A human pharmacokinetics study of • Optociliary shunt vessels on disc
nonvitrectomized eyes, found a single 4 mg intravitreal injec- • Neovascularization of the iris (less commonly of the optic disc
tion of triamcinolone to have a mean half-life of 18.6 days and retina)
with measurable concentrations expected to last approximately
3 months.110 Reported side effects include cataract, increased
intraocular pressure and injection-related complications
INTRODUCTION
including noninfectious and infectious endophthalmitis, retinal CRVO typically presents in a fairly acute and dramatic fashion,
1760 detachment, vitreous hemorrhage, and lens injury. often permitting early diagnosis. Classically, a CRVO presents
Retinal Venous Occlusive Disease

with marked dilation and tortuosity of the retinal veins, exten- predicting the clinical course and choosing treatment options
sive retinal edema, pronounced superficial and deep retinal hemor- can be much more difficult and challenging. The pathogenesis
rhages radiating outward from the optic disk in all quadrants, and treatment of CRVO continues to be highly debated.
cotton-wool spots, and optic disk swelling (Fig. 132.7). In its Research continues to direct the understanding and treatment
acute phase, the diagnosis is often made with little doubt. In options of the disease.
less severe instances or late in the course of the disease, the
diagnosis may be less obvious. With time, the dramatic retinal
findings may resolve and the diagnosis may become more EPIDEMIOLOGY
difficult. CRVO is a relatively common cause of severe vision loss. The
In its mildest form, CRVO may consist of only vascular majority of patients who develop CRVO are over the age of 50
dilatation and tortuosity, disk hyperemia, and a few retinal years and 50–70% have associated hypertension, cardiovascular
hemorrhages (Fig. 132.8). In this form, the condition is likely to disease, or diabetes mellitus.117 Numerous studies have
resolve without sequelae. At the other extreme, there may be assessed the incidence of CRVO with varying results. In the
nearly confluent retinal hemorrhages, cotton-wool spots, Beaver Dam Eye Study,13 the incidence of CRVO was 0.2%
massive retinal and macular edema, and extensive capillary during the 5-year follow-up study of 4926 residents of Beaver
nonperfusion. In the latter cases, one expects to observe severe Dam, WI aged 43–84 years. There was no gender predilection.
visual impairment and a tendency for the development of In the BMES8 the overall incidence of CRVO was 0.4% with an
macular edema and anterior segment neovascularization, increasing incidence with age. The BMES also showed no

CHAPTER 132
including neovascular glaucoma. gender predilection.
While early diagnosis of a dramatic presentation of a CRVO
may not require much effort on the part of the clinician,
PATHOGENESIS
The pathogenesis of CRVO is not completely understood,
despite being first described by Leibreich in 1855.118 Numerous
theories have been presented over the years, without consensus
on the pathogenesis of the disease. Hayreh has suggested that
perfused CRVO occurs after occlusion of retinal venous flow,
while nonperfused CRVO develops after occlusion of venous
and arterial flow. His studies were based on animal models with
occlusion of the retinal vessels at their entry into the optic
nerve.83–85 However, Fujino’s pathologic studies found that
occlusion of the retinal vein alone could produce clinical
findings similar to nonperfused CRVO.86
Green et al reported a histopathologic study which supports
the theory of thrombus formation as the inciting event causing
CRVO.87 In this study, a fresh or a re-canalized thrombus was
observed in 29 eyes from 29 patients with CRVO. The study
considered the temporal aspects (6 h to 10 years from onset) of
the cases, and noted the different morphologic features of the
occlusion. These observations explain most of the variability of
the changes observed in previous reports and are likely to be
associated with the evolution of the thrombus. Endothelial cell
FIGURE 132.7. Typical appearance of a fresh central retinal vein
proliferation was a conspicuous feature in 14 (48.3%) of the
occlusion (CRVO), with disk edema, venous dilatation and tortuosity,
cotton-wool spots, and retinal hemorrhages in all quadrants.
eyes, chronic inflammation in the area of the thrombus and/or
vein wall or perivenular area was observed in 14 (48.3%),
arterial occlusive disease in seven (24.6%), and cystoid macular
edema in 26 (89.7%) of the eyes. Most of the eyes included in
the study had chronic, nonperfused CRVO and had been
enucleated for neovascular glaucoma. The few recent-onset
occlusions occurred in patients in the terminal stages of severe
systemic disease. Eyes with recent-onset and perfused CRVOs
were underrepresented in this series.
The reason that thrombus formation tends to occur in the
region of the lamina cribrosa is unknown. The close anatomic
association of the central retinal artery and the central retinal
vein in this region, as well as the narrowing of the central
retinal vessels as they pass through the lamina cribrosa, may
contribute to turbulent flow and thrombus formation.119
Numerous risk factors for CRVO have been identified,
including systemic, ocular, and localized abnormalities. A con-
current systemic disease is present in at least half of the patients.
The Eye Disease Case-Control Study found an increased risk of
either type of CRVO in persons with systemic hypertension and
diabetes mellitus.120 Other studies have shown that approxi-
mately 60% of patients diagnosed with CRVO have a history of
FIGURE 132.8. CRVO demonstrates disk hyperemia and venous hypertension. Vasculitis associated with diseases such as
dilatation and tortuosity, with mild retinal hemorrhages. sarcoid, syphilis, systemic lupus erythematosus is also a risk 1761
RETINA AND VITREOUS

factor. Retrobulbar external compression from thyroid disease patients younger than 50 years with a CRVO found that 26% of
or orbital tumor also increases the risk of CRVO.25,121–126 the subjects had activated protein C resistance, compared with
Systemic disease appears to play a significant role in CRVO 2–7% in the general population.151 Some studies have suggested
development. Atherosclerosis has been reported to be a major that factor V Leiden is a risk factor for the development of
risk factor for CRVO development. One theory is that athero- CRVO in patients younger than 60 years of age,152,146 however,
sclerosis of the adjacent central retinal artery compresses the other studies have not supported this notion.140,145
central retinal vein in the region of the lamina cribrosa, second- Lupus anticoagulant factor is a circulating immunoglobulin
arily inducing thrombosis in the lumen of the vein. Younger that may cause mild prolongation of coagulation studies,
patients with CRVO have been reported to have an increased especially partial thromboplastin time, but paradoxically is
risk of cardiovascular death and collagen vascular disease.121,127,128 associated with thrombosis. This factor can be seen in some
However, much of these data have come from case series patients with systemic lupus, but can be present in the absence
without control groups. Elman compared the prevalence of of lupus. Patients with this factor frequently test positive for
hypertension, diabetes, cardiovascular and cerebrovascular antiphospholipid antibodies, including anticardiolipin, and may
disease, and mortality among CRVO patients and a control test falsely positive on Venereal Disease Research Laboratory
group from the Wilmer Ophthalmological Institute and another (VDRL) testing. Systemic manifestations of the disease include
control group from a national survey.129 He found a higher retinal vein, retinal artery, and choroidal occlusions as well as
prevalence of hypertension in the CRVO patients when com- spontaneous abortions and systemic occlusions.149,153–158
pared to both control groups, but diabetes was more prevalent Hyperviscous states such as elevated hematocrit, increased
SECTION 10

only when compared with the national survey control group. erythrocyte aggregation, decreased erythrocyte deformability,
Rates of cardiovascular disease, cerebrovascular disease, and elevated fibrinogen, dehydration, polycythemia, lymphoma,
mortality did not differ significantly among the groups. When leukemia, sickle cell disease, multiple myeloma, cryo-
comparing with a general ophthalmology patient population, globulinemia, and Waldenstrom macroglobulinemia have been
Rath found a higher association of CRVO with male gender, associated with CRVO.159–161 Common medications that can
systemic hypertension, and open-angle glaucoma.126 Elevated contribute to hematologic abnormalities that may increase the
serum lipid levels have also been reported to be associated with risk of CRVO include diuretics and oral contraceptives.
increased risk.125,130 The Eye Disease Case-Control Study Abnormal platelet function can also contribute to CRVO
Group compared patients with CRVO to age-matched controls. formation. Some have speculated that increased blood viscosity
There was a higher cardiovasacular risk profile, which included may increase the risk of nonperfused CRVO.112,125,132,162–164
hypertension or diabetes, less physical activity, and decreased Certain ocular findings have been shown to be risk factors for
alcohol consumption, among patients with CRVO. The CRVO; however, the mechanism affecting the risk of CRVO is
association was more significant in patients with nonperfused poorly understood. An increased risk has been noted in eyes
rather than perfused CRVO. In women, the risk increased with with open-angle glaucoma. In uncontrolled studies, 40% of
higher erythrocyte sedimentation rates and decreased with use patients with CRVO had preexisting open-angle glaucoma, or
of postmenopausal estrogen.120 developed glaucoma during follow-up.165,166 The Eye Disease
Certain hematologic abnormalities are risk factors for CRVO, Case-Control Study found that patients with a CRVO were five
especially in young adults. Thrombophilic factors have been times more likely than age-matched controls to have either
shown to be associated with an increased risk of RVO. Hyper- glaucoma or an intraocular pressure greater than 20 mmHg.120
homocysteinemia appears to be one of the most common and Other ocular findings associated with increased risk of CRVO
significant thrombophilic risk factors for development of CRVO. include increased cup-to-disk ratio (independent of glaucoma
Less evidence exists for Factor V Leiden mutation, protein C and increased introcular pressure),167 ischemic optic neuropathy,
and S deficiency, antithrombin deficiency, antiphospholipid tilted optic nerve head, optic nerve head drusen, optic disk
antibodies, activated protein C resistance, prothrombin gene traction syndrome, pseudotumor cerebri, external compression
mutation, anticardiolipin antibodies, abnormal fibrinogen of the optic nerve and globe from thyroid-related ophthalmo-
levels, and lupus anticoagulant.120,131–148 pathy, and mass lesions or head trauma with orbital fracture.117
Homocysteine is a naturally occurring molecule in the body Systemic workup is a vital part of the examination process if
and is required in several reactions that occur within the cells. no identifiable risk factor is known. Many of the risk factors
If the pathways to either cysteine or methionine are blocked, for CRVO are treatable systemic diseases that, if left untreated,
then homocysteine levels may rise. A patient who is hetero- can result in high morbidity and mortality. Common diseases
zygous for this mutation has no evidence of hyperhomo- associated with CRVO include hypertension, diabetes mellitus,
cysteinemia or increased risk of thrombotic disorders. Patients and atherosclerosis.25,120–126,129 However, other diseases should
who are homozygous for the defect can develop hyperhomo- be considered, especially in persons younger than 56 years of
cysteinemia. Additionally, deficiencies in vitamin B6 and folate age and in those with bilateral CRVO. In general, further work-
can lead to increased levels of homocysteine. Other causes up is not indicated in persons older than 55 years of age with
include certain medications and kidney disease. The prevalence known systemic vascular risk factors for CRVO.168,169
of hyperhomocysteinemia in the general population is not
known. Hyperhomocysteinemia can be treated with vitamin
supplementation; folate and vitamin B12 have shown to CLINICAL FEATURES
decrease serum homocysteine levels. However, there is no con- Historically, confusion has existed regarding the classification of
vincing data that decreasing homocysteine levels with vitamin CRVO subtypes. In 1904, Coats was the first to describe two
supplementation will reduce the risk of CRVO.143,144,148 types of CRVO. He recognized that one group of patients often
Protein C is a naturally occurring anticoagulant. Protein C had marked vision loss and a poor prognosis, while another
deficiency is a rare cause of RVO; however, activated protein C group had much less visual disturbance and had a better
resistance is the most common identifiable cause of systemic prognosis.170 With the advent of FA, these two types of CRVO
venous thrombosis.149,150 The inheritable condition is trans- were then classified based on retinal capillary perfusion
ferred in an autosomal dominant pattern. While systemic levels characteristics. Multiple terms have been used in an attempt to
of protein C are normal, the usual anticoagulation response is differentiate the two clinical pictures. Terms used to refer to the
1762 abnormal leading to increased thrombosis. One study of 31 more severe type include ‘hemorrhagic retinopathy’, ‘complete’,
Retinal Venous Occlusive Disease

‘nonperfused’, or ‘ischemic CRVO’. Conversely, the more only certain areas appear nonperfused on evaluation. Retinal
benign type has been referred to as ‘venous stasis retinopathy’, hemorrhage may prevent correct quantification of retinal
‘partial’, ‘perfused’, or ‘nonischemic’.8 Our discussion will use nonperfusion due to blockage of fluorescence in the area of
the terms perfused and nonperfused. It is important to under- hemorrhages. Although no standard criteria have been
stand the distinguishing features of the two groups in order to established, the CVOS defined ‘ischemia’ (synonymous with
guide patient counseling, understand prognosis, and guide nonperfusion in our discussion) as 10 or more disk areas of
treatment (Table 132.2). nonperfusion on FA.
A common presenting symptom in CRVO is abrupt decrease Perfused CRVO typically has mild fundus changes, no
in vision. However, some patients will describe transient vision afferent pupillary defect, visual acuity better than 20/400, less
loss lasting a few seconds to minutes over the preceding days to extensive capillary nonperfusion on FA, and nearly normal
weeks. Some patients will report symptoms of photophobia and ERG testing. Ocular neovascularization rarely develops in
red eye; however, these symptoms, if present, typically last for a perfused CRVO eyes and visual prognosis is generally more
few days to weeks only. Rarely, presenting symptoms include favorable.
pain, blurry vision from corneal haze, and increased intraocular A complete ocular examination, including intraocular
pressure. These patients may have a subacute or chronic CRVO pressure measurement, slit-lamp biomicroscopy, gonioscopy to
that has not been detected and has resulted in the more severe rule out neovascularization of the iris or angle, and a dilated
complication of neovascular glaucoma. fundus examination is recommended at the initial presenta-
Initial evaluation should include distinguishing between tion and at monthly follow-up examinations during the first

CHAPTER 132
perfused and nonperfused CRVO. Typically, two-thirds of all 9 months. Neovascularization of the iris or angle is an ominous
CRVOs are perfused, while the remaining one-third are sign of potential development of neovascular glaucoma.
nonperfused.51,121,171 Clinical examination combined with FA
are most commonly used to differentiate between the two broad Visual Acuity
categories. Electrophysiologic testing and visual field testing There is wide variability in visual acuity following a CRVO;
may also be helpful in the evaluation process when FA cannot however, the presenting visual acuity is highly predictive of final
demonstrate the extent of nonperfusion (e.g., in cases with visual outcome. Visual acuity can range from 20/20 to hand
extensive intraretinal hemorrhage). Serial monitoring of the motion, or even no light perception in those with end stage
CRVO subtype is important during follow-up examinations, as neovascular glaucoma. The CVOS4–6 reported that in 65% of
up to one-third of perfused CRVOs progress to nonperfused over patients with presenting visual acuity better than 20/40, the
the course of the disease.4,51,121,123,172 The Central Vein vision remained stable. Patients with poor visual acuity at
Occlusion Study (CVOS)4–6 was a multicentric, international presentation (<20/200) had an 80% chance of having a visual
clinical trial sponsored by the National Eye Institute evaluating acuity worse than 20/200 at the final visit.
725 patients and 728 eyes with CRVO followed at least 3 years. Visual acuity can be helpful in distinguishing perfused versus
Eyes were entered into four predefined study groups: perfused, nonperfused CRVO. Patients with perfused CRVO tend to have
nonperfused, indeterminate, and macular edema. The CVOS better visual acuity; however, vision may still be quite poor due
reported that in the first 4 months of follow-up, 81 (15%) of the to macular edema or other complications.51,121,122 While poor
547 eyes with perfusion converted to nonperfusion. During the visual acuity can be seen in either perfused or nonperfused
next 32 months of follow-up, an additional 19% of eyes were CRVO, relatively good initial visual acuity is suggestive of
found to have converted to nonperfusion for a total 34% after perfused CRVO. Hayreh reported a visual acuity better than
3 years. Risk factors for progression include worse presenting 20/200 (6/60) in 58% of the eyes with perfused CRVO, as
visual acuity, severe macular edema, and progressive intra- compared to only 1.7% of eyes with nonperfused CRVO. A
retinal hemorrhage.172 visual acuity better than 20/400 (6/120) was measured in 81%
Nonperfused CRVO is often more dramatic in its presen- of eyes with perfused CRVO, compared to about 7% of the eyes
tation and is associated with a worse visual outcome than with nonperfused CRVO. A visual acuity of 20/400 or worse was
perfused CRVO.119 The nonperfused type typically presents measured in only 19% of the eyes with perfused CRVO,
with multiple cotton-wool spots, extensive retinal hemorrhage, compared to 93% of the eyes with nonperfused CRVO.119
a relative afferent pupillary defect, visual acuity worse than
20/400, widespread capillary nonperfusion on FA, and Afferent Pupillary Defect
abnormal electroretinography (ERG) testing. One of the great Evaluation for a relative afferent pupillary defect can be helpful
difficulties in evaluating CRVO is understanding the risks in distinguishing perfused and nonperfused CRVO. Eyes with
associated with varying degrees of retinal nonperfusion. Retinal nonperfused CRVO are much more likely to have a relative
nonperfusion rarely occurs throughout the entire fundus. Often, afferent pupillary defect. Ninety percent of eyes with perfused
CRVO were found to have a relative afferent pupillary defect of
0.3 log units or less. However, 91% of eyes with nonperfused
CRVO had a relative afferent pupillary defect of 1.2 log units
TABLE 132.2. Common Clinical Findings in CRVO or more.173
Test Perfused Nonperfused
Intraocular Pressure
Fluorescein angiogram Uniform capillary Patchy capillary Relative intraocular pressure difference is less helpful in the
fluorescence hypoflouresence evaluation process. However, immediately after CRVO, the
Afferent papillary defect Minimal/absent Present intraocular pressure is typically slightly lower in the affected eye
as compared to the fellow eye. This relative difference in intra-
Visual acuity >20/400 <20/400
ocular pressure diminishes with time, and symmetry returns
Electroretinogram Normal/ Delayed over the ensuing weeks to months.174
supernormal
Risk of Low risk High risk Visual Field Testing
neovascularization Visual field testing is widely variable in CRVO, but may be bene-
ficial for better understanding the patient’s visual perception. 1763
RETINA AND VITREOUS

Abnormalities are more common and more severe in eyes with walls of the retinal veins has been demonstrated to be an
nonperfused rather than perfused CRVO.175 indicator of nonperfusion. Retinal capillary nonperfusion is an
important feature in identifying those eyes at greatest risk of
Fluorescein Angiography (FA) developing neovascularization.171,176,177 There are no definite
FA can be very helpful in evaluating the extent and mechanism guidelines for assessing the risk of neovascularization; however,
of retinal dysfunction based on perfusion characteristics. The the CVOS used 10 disk diameters of nonperfusion as its cut-off
characteristic findings in CRVO are due to changes in vascular for predicting risk.4 The location of the nonperfusion is also
caliber, vascular permeability, and capillary nonperfusion. Wide- important in terms of predicting long-term visual potential.
angle FA using a 60° fundus camera is helpful in evaluating Nonperfusion near the fovea is associated with worse visual
peripheral retinal perfusion. The overall arterial perfusion time prognosis. Overlying retinal hemorrhage may impede the ability
after injection is normal to slightly delayed in CRVO; however, to evaluate the underlying capillary perfusion status.
the delay in arteriovenous transit time is typically significantly FA is also helpful in evaluating causes of central vision loss.
delayed (Fig. 132.9).43 A delay beyond 20 seconds is associated Central vision loss is common following CRVO and can be the
with a greater risk of iris neovascularization.123 Nonperfused result of macular edema, parafoveal capillary nonperfusion,
areas may be visualized as patchy areas of hypofluorescence intraretinal hemorrhage, or retinal pigment epithelial damage
with a ground-glass appearance (Fig. 132.10). Staining of the due to deep retinal or subretinal hemorrhage. In macular edema,
FA shows parafoveal and paramacular capillary leakage that
appears as hyperfluorescence that increases in size and intensity
SECTION 10

with time, often in a petalloid type pattern. The hyper-


fluorescence persists longer than that of normal retinal tissue.
Hemorrhage causes persistent blocking of the fluorescence
throughout the angiographic evaluation.

Electrophysiologic Testing
Electrophysiologic testing with electroretinogram (ERG) may be
helpful in distinguishing between perfused and nonperfused
CRVO.178 Matsui et al reported that the ERG b/a-wave ampli-
tude ratios, photopic and scotopic b-wave amplitudes, and
flicker amplitudes were significantly smaller in eyes with exten-
sive capillary nonperfusion, than in eyes without. When the
photopic or scotopic b-wave amplitudes were normal or super-
normal, extensive capillary nonperfusion on FA was absent in
all eyes. When the b/a-wave ratios were greater than or equal
1.0, or when the b-wave amplitudes with white flash or flicker
amplitudes were normal or supernormal, extensive capillary
nonperfusion was present in less than 32% of eyes. When the
b/a ratio was less than one, there was a higher probability of
retinal nonperfusion and a higher risk of developing iris
neovascularization.179
FIGURE 132.9. Delayed filling of the retinal venous system in a
CRVO. This frame, taken 14 s after the appearance of fluorescein in
the retinal arteries, shows minimal filling of the retinal venous system. Optical Coherence Tomography (OCT)
OCT is a helpful tool in evaluating the macula following CRVO.
The OCT can aid the clinician in evaluating macular edema,
epiretinal membrane formation, and subretinal fluid accumula-
tion following CRVO.180,181

COMPLICATIONS
The clinical course after a CRVO is highly variable. The retinal
hemorrhages and microaneurysms may resolve over weeks or
may persist for years. The Blue Mountains Eye Study reported
a 10% incidence of residual retinopathy lesions (microaneurysms,
hemorrhages, hard or soft exudates) at the 5 year follow-up in
nondiabetic patients.182 Macular edema may resolve quickly,
persist chronically, vary intermittently, or develop late in the
course of the disease. Neovascularization typically occurs on the
iris in CRVO; however, neovascularization may develop on
the disk and, or retina. Fundus neovascularization very rarely
develops in eyes with perfused CRVO.8,121,123,176,183,184 If neo-
vascularization develops, it usually is present within the first 7
months of the CRVO. The venous dilatation and tortuosity
typically resolve with time, and marked fibrous sheathing of the
retinal veins and arteries may develop. The disk swelling slowly
resolves and may develop disk pallor in nonperfused cases.
Collateral vessels at the optic disk may develop.
FIGURE 132.10. Fluorescein angiogram of a profoundly nonperfused Macular dysfunction is common following CRVO. The visual
1764 CRVO. Note the nearly complete absence of retinal capillaries. disturbances may be transient or chronic. A common cause of
Retinal Venous Occlusive Disease

macular dysfunction is retinal edema. Retinal edema may ischemia with iris and retinal neovascularization in CRVO.
develop from abnormal vascular permeability following CRVO Upregulation of VEGF production appears to occur most
in any part of the retina. It often presents as a petalloid pattern consistently in the inner nuclear layer in hypoxic retina.188–190
of cystoid macular edema.180 Severe macular edema is a risk The clinician must take care not to confuse disk neovascu-
factor for progression of perfused CRVO to nonperfused.172 The larization with optociliary shunt vessels of the disc. Optociliary
exact mechanism causing the leakage is unknown, but may shunt vessels develop commonly following CRVO (Fig. 132.12).
be due to vascular congestion, capillary damage, or localized The vessels form as collateral channels between the retinal and
inflammatory reactions. ciliary circulations. The shunt vessels are typically larger in
Serous retinal detachment has also been described following caliber and do not leak on FA as compared to typical disk
CRVO. With the advent of better cross-sectional retinal imaging neovascularization. Opinions differ as to whether optociliary
using OCT, serous retinal detachments are reported to occur vessel formation is associated with an improved visual
more frequently than recognized previously.181 prognosis;121,127,191,192 there may be a decreased risk of
Microaneurysms are a common finding following CRVO (Fig. developing anterior segment neovascularization in eyes with
132.11). Less commonly, macroaneurysms may develop.48 Hard retinochoroidal collateral vein formation.167
exudates are a less common finding following CRVO. Large
amounts of hard exudates are associated with nonperfusion,
poor visual acuity, and elevated serum triglyceride levels.185,186 DIFFERENTIAL DIAGNOSIS
The hard exudates typically resolve with time. Retinal hemor- Diabetic retinopathy can present with findings similar to CRVO

CHAPTER 132
rhage can vary in its presentation; however, at least mild retinal with dot-blot hemorrhages and microaneurysms. However, in
hemorrhage is virtually always present following CRVO. The diabetic retinopathy, the retinal findings are generally bilateral,
location of the hemorrhage is typically intraretinal and superficial. compared with CRVO, in which the insult is typically uni-
Occasionally, vitreous hemorrhage may develop following an lateral. Bilateral CRVO is an uncommon presentation.
acute CRVO; vitreous hemorrhage is usually a result of neovas- Ocular ischemic syndrome due to carotid occlusive disease
cularization of the retina or disk at a later stage of the disease. must also be distinguished from CRVO. In the ocular ischemic
Iris and angle neovascularization are dreaded complications syndrome the veins are similarly dilated and irregular, but are
of CRVO because of the risk of developing neovascular not as tortuous as those seen in CRVO. Neovascularization of
glaucoma. Neovascular glaucoma often leads to intractable the disk may be present in either disease without disk swelling
elevated intraocular pressure, blindness, and extreme eye pain. or hemorrhages. Retinal hemorrhages are typically seen in the
Evisceration or enucleation, is all too often the final treatment mid-periphery in ocular ischemic syndrome. Patients with
option once neovascular glaucoma becomes medically uncon- ocular ischemic syndrome may have a history of amaurosis
trollable. Of those that develop neovascular glaucoma, 66–71% fugax, transient ischemic attacks, or orbital pain. Intraocular
do so within the first 6 months of CRVO development. The pressure may be low.
incidence of rubeosis among all CRVOs is approximately 20%. Papilledema may present similarly to CRVO. Disk swelling is
Among non-perfused eyes, the incidence is 45–80%. In perfused generally bilateral due to increased intracranial pressure.
eyes, the rate of iris neovascularization is one to ten percent. Similar disk hemorrhages may be observed; however, the
Of eyes that develop iris neovascularization, approximately hemorrhages are generally localized to the optic disk with
two-thirds will develop neovascular glaucoma without papilledema and typically do not involve the peripheral retina.
treatment.51,121,123,176,183 Early treatment of neovascularization Radiation retinopathy may be unilateral or bilateral. A his-
of the iris with PRP results in resolution of the iris tory of irradiation is essential for the diagnosis. Similar to
neovascularization in approximately two-thirds of the cases.187 CRVO, disk swelling, disk hemorrhages, retinal neovascu-
VEGF appears to play an important role in the development of larization, and cotton-wool spots may be present.
neovascularization and macular edema. Hypoxia-induced Hypertensive retinopathy also may be considered in the
upregulation of VEGF may be an important factor in retinal differential diagnosis of CRVO. Distinguishing characteristics of

FIGURE 132.11. Large capillary aneurysms that occurred after a FIGURE 132.12. Typical, tortuous appearance of disk collaterals after
CRVO. Peripheral to the aneurysms is a large area of nonperfusion. a CRVO. 1765
RETINA AND VITREOUS

hypertensive retinopathy include narrowed retinal arterioles perfused macular edema and 20/50 acuity or worse. Laser treat-
and bilateral involvement of the retinal hemorrhages. ment involved a grid pattern in the area of leaking capillaries
within two disk diameters of the foveal center but not within
TREATMENT the foveal avascular zone. There was no significant difference in
mean visual acuity between treated (20/200) and untreated
Medical Treatment (20/160) eyes at 36 months. There was angiographic resolution
Identification and treatment of systemic vascular risk factors in of the macular edema by 1 year in 31% of treated eyes (com-
conjunction with the internist is important in individuals with pared with 0% of untreated eyes), but there was a lack of visual
CRVO. In general, a systemic workup is not indicated in per- recovery likely secondary to widespread damage to the
sons older than 55 years of age with known systemic vascular perifoveal capillary network. Therefore, the CVOS did not
risk factors for CRVO.168,169 If no known systemic vascular risk recommend grid laser photocoagulation for CRVO-associated
factor is present, initial investigation may include checking macular edema. However, in the younger patients there was a
blood pressure, intraocular pressure, complete blood count, trend toward improved visual acuity in the treatment group.
glucose levels, and a lipid panel on all patients with CRVO. This possibly was not statistically significant given the small
More extensive evaluation may be considered if the initial sample size.
screen does not reveal the presence of a systemic risk factor. The
role of systemic anticoagulation in CRVO is unclear, as there is Chorioretinal Venous Anastomosis
no evidence that agents such as aspirin, heparin or warfarin Laser-induced chorioretinal venous anastomosis was initially
SECTION 10

prevent or alter the natural history of CRVO. Patients taking described by McAllister and Constable.197 The goal is to bypass
warfarin have been reported to develop CRVO despite main- the occluded vein in perfused CRVO. Overall, the treatment
taining therapeutic levels of anticoagulation.193 Limited data consists of producing an intense focal laser burn directed at or
suggests that systemic hemodilution and/or pentoxifylline near a tributary vein in an attempt to disrupt the wall of the
(lowers blood viscosity) may be beneficial in improving visual vein and rupture underlying Bruch’s membrane.198,199 Various
acuity and macular edema, and reducing the risk of progression laser wavelengths have been employed, including argon green,
to ischemic CRVO.194,195 argon blue–green, dye yellow, and Nd-YAG. Successful anasto-
mosis formation is achieved in 33–100% of cases with variable
Laser Photocoagulation visual function recovery.197,198,200–202 However, the treatment is
One of the most feared complications of CRVO is neovascular associated with complications in up to 67% of cases; these
glaucoma. Early signs of developing neovascular glaucoma complications include posterior vitreous detachment, choroidal
include neovascularization of the iris and angle. The CVOS4–6 or vitreous hemorrhages, preretinal fibrosis, choroidal neo-
found that the most important predictive factor of iris neo- vascularization, segmental retinal ischemia, choriovitreal
vascularization in CRVO is poor visual acuity. Other risk factors neovascularization, and retinal detachment.197,198,200–206
identified included increasing amounts of retinal capillary Some surgeons have attempted to create a successful chorio-
nonperfusion and intraretinal blood. The study found that, retinal venous anastomosis by means of surgery, especially in
prophylactic PRP decreased the risk of iris neovascularization nonperfused CRVO.207,208 This technique may offer improved
compared to untreated eyes, but the difference was not statis- visual status in eyes with perfused CRVO and BRVO, but the
tically significant. Laser treatment after the development of iris precise treatment variables and method to reliably create an
or angle neovascularization was followed by prompt regression anastomosis while minimizing complications have yet to be
(within 1 month) in 18 (56%) of 32 previously untreated eyes determined.
and in four (22%) of 18 eyes that had undergone prophylactic
PRP. The authors recommended PRP be delivered promptly Conventional Pars Plana Vitrectomy for
after the development of iris or angle neovascularization in eyes Complications of CRVO
with nonperfused CRVO. Hayreh et al conducted a prospective, Pars plana vitrectomy techniques are often employed to address
study of argon laser PRP in nonperfused CRVO over a 10 year complications of CRVO, especially nonclearing vitreous
period in 123 eyes. On comparing the laser-treated eyes versus hemorrhage.209 At the time of vitrectomy, evacuation of the
the eyes not treated by laser, there was no statistically sig- hemorrhage can be combined with removal of epiretinal
nificant difference between the two groups in the incidence of membranes and endolaser photocoagulation if warranted.
development of angle neovascularization (NV), neovascular
glaucoma (NVG), retinal and/or optic disk NV, or vitreous Pars Plana Vitrectomy for Macular Edema
hemorrhage, or in visual acuity. The study, however, did show a Some authors believe that the absence of posterior vitreous
statistically significant (P = 0.04) difference in the incidence of detachment can contribute to the occurrence or the persistence
iris NV between the two groups, with iris NV less prevalent in of macular edema in CRVO and that relieving the vitreous
the laser group than in the nonlaser group, but only when the traction over the macula by means of vitrectomy-induced
PRP was performed within 90 days after the onset of CRVO.196 posterior vitreous detachment may assist in improving the
The delivery of PRP before the development anterior segment macular edema.71,209,210 Sekiryu et al211 reported on five patients
neovascularization may be considered in eyes with nonperfused with macular edema caused by CRVO that were examined
CRVO when monthly ophthalmologic examination is not possible. using OCT. The retinal thickness through fixation was reduced
However, 20% of these patients will develop iris-neovascular- in all five eyes. Preoperatively, the retina thickness at the
ization despite laser photocoagulation treatment. It should be foveola was more than 500 mm in three eyes and more than
kept in mind that some patients who undergo PRP will have 1000 mm in two eyes. The retina thickness was reduced to
permanent peripheral visual field defects following treatment.196 311+/– 80 mm within 2 weeks on average. Visual acuity was
Patients with CRVO who present with neovascular glaucoma improved by two or more lines in three of five eyes.
should undergo PRP, or, if the media is hazy, cryoablation.
Topical intraocular pressure lowering drugs should be given and Radial Optic Neurotomy
a surgical glaucoma procedure may be indicated. Opremcak et al212 hypothesized that the pathogenesis of CRVO
The CVOS also studied the effectiveness of grid pattern argon may be similar to ‘compartment syndromes’ elsewhere in the
1766 laser photocoagulation in improving visual acuity in eyes with body, where pressure within a confined space results in tissue
Retinal Venous Occlusive Disease

ischemia. According to this hypothesis, an anatomic ‘bottle- Lam et al236 reported subretinal peripapillary tPA injection
neck’ exists at the lamina cribrosa where the central retinal with vitreopapillary and epipapillary membrane dissection and
artery, central retinal vein and optic nerve lie within a 1.5 mm- peripheral photocoagulation in a patient with unresolved CRVO
diameter area with dense connective tissue encircling these at 4 months with a modest improvement of visual acuity from
structures, restricting the central retinal vein luminal finger counting to 20/400. Endovascular delivery of r-tPA
diameter.213 Optic nerve sheath decompression was initially involves cannulation of retinal vessels, either through a
attempted using an external approach by Vasco–Posada214 and neuroradiologic237 or a vitreoretinal approach238–240 and allows
Arcienigas.215 More recently, Opremcak proposed radial optic for delivery of minute quantities of r-tPA directly to the
neurotomy to achieve the decompression of the scleral outlet occluded vessels. Retinal endovascular surgery (REVS) involves
via an internal, vitreoretinal approach.212 Such a surgical vitrectomy followed by insertion of a microcannula into
approach was performed in 11 cases, achieving visual acuity branches of the retinal vasculature with injection of pharma-
improvement in eight cases (73%). Seven of the 11 eyes (64%) cologic agents such as tPA. Weiss and Bynoe have reported their
achieved a final visual acuity of 20/200 or better, and five (45%) technique of pars plana vitrectomy followed by cannulation of a
achieved a final acuity of 20/70 or better. No significant com- branch retinal vein with injection r-tPA toward the optic nerve
plications were observed postoperatively. Other authors have head.239 Of 28 eyes with CRVO of greater than 1 month dura-
noted varying degrees of clinical improvement following surgical tion and worse than 20/400 preoperative acuity, 50% recovered
radial optic neurotomy including a decline in macular edema more than three lines of acuity with a mean follow-up of
and decreased or resolved intraretinal hemorrhages.216–220 12 months. There was a trend toward increased perfusion on

CHAPTER 132
However, others have reported no statistically significant FA attributed in part to the resolution of intraretinal
improvement of vision following the procedure.221,222 Reported hemorrhages after the procedure. Complications included
complications associated with this procedure include optic vitreous hemorrhage in seven eyes and retinal detachment in
nerve damage, vitreous hemorrhage, subretinal hemorrhage, one eye.
peripapillary retinal detachment, choroidal neovascularization,
and temporal visual field defect.223–230 Intraocular Corticosteroids
High-dose oral corticosteroids have been reported to be
Tissue Plasminogen Activator associated with a transient reduction in macular edema with
Thrombolytic agents have been proposed as a treatment improved vision over the short term in a small number of
directed against a suspected thrombus in the central retinal patients with CRVO, but were associated with systemic side
vein. Recombinant tissue plasminogen activator (r-tPA) is a effects and recurrence of macular edema.242 Small case studies
synthetic fibrinolytic agent that converts plasminogen to have suggested that there may be a transient benefit from
plasmin and destabilizes intravascular thrombi. Reduction in intravitreal injection of steroids for macular edema and vision
clot size may facilitate dislodging of the entire thrombus or re- loss associated with CRVO.243–251 The outcome appears to be
canalization of the occluded retinal vein. As therapy for CRVO, dependent on whether the CRVO is perfused or nonperfused.
r-tPA has been administered by several routes: systemic, One such study by Ip et al248 reviewed the medical records of 13
intravitreal, subretinal and via endovascular cannulation of consecutive patients (13 eyes) with macular edema associated
retinal vessels. with CRVO who were treated with an injection of intravitreal
Systemic administration of low-dose (50 mg) front-loaded triamcinolone acetonide (4 mg) at the University of Wisconsin
r-tPA has been attempted in two pilot studies with visual acuity and the Bascom Palmer Eye Institute. Each intravitreal injection
improvement in 30–73% of patients.229,231 However, this treat- was delivered through the pars plana using a 27- or 30-gauge
ment has been associated with serious complications, including needle. The median age of the 13 patients was 67 years
patient mortality, which have curbed the use of systemic (interquartile range, 57–77 years), and the median duration of
administration of r-tPA for CRVO. symptoms before injection was 8 months (interquartile range,
Intravitreal delivery of r-tPA has potential advantages includ- 4–9 months). Mean baseline visual acuity was 20/500 in the
ing directed delivery to the retinal vessels as well as decreased affected eye. Mean visual acuity at the 6 month follow-up
risk of ocular and systemic complications. Of 47 persons in examination was 20/180 in the affected eye. All 13 patients
three studies of intravitreal r-tPA for both ischemic and completed the 6 month examination. Eyes with perfused CRVO
nonischemic CRVO of less than 21 days duration, 28–44% (n = 5) demonstrated a significant improvement in visual
experienced three lines of visual acuity improvement with acuity, whereas eyes with nonperfused CRVO (n = 8)
6 months follow-up.232–234 However, the significance of the demonstrated a nonsignificant visual acuity improvement. No
visual outcome is severely limited, as none of the studies patient had a decrease in visual acuity. Mean baseline foveal
included a control group. Administration of r-tPA did not sig- thickness as measured by OCT was 590 mm (retinal thickening
nificantly alter final perfusion status. Ghazi et al reported = 416 mm). Mean foveal thickness as measured by OCT at the
intravitreal r-tPA in 12 eyes with acute nonperfused CRVO of 1month follow-up examination in 12 patients was 212 mm
less than 3 days duration.235 In all eyes, perfusion status (retinal thickening = 38 mm). Between the 3- and 6-month
remained unchanged at last follow-up. Nine patients (75%) had follow-up examinations, four patients developed a recurrence of
best-corrected visual acuity of 20/200 or worse at presentation macular edema. Three of the four patients were retreated with
compared with four patients (33%) at the last follow-up after a second injection of triamcinolone. Two of these three patients
treatment. Five (55%) of these nine patients had final visual experienced an improvement in visual acuity following retreat-
acuity that improved to 20/50 or better. The remaining four ment. At the 6 month follow-up examination, mean foveal
patients did not have improvement or their vision continued to thickness as measured by OCT for 13 patients was 281 mm
worsen. Overall, eight (67%) of 12 patients had a final visual (retinal thickening = 107 mm).
acuity of 20/50 or better. No side effects related to tPA injection The mechanism of action of corticosteroids for macular
were observed. This report suggests that prompt use of intra- edema in CRVO is not clear and has been discussed above with
vitreal r-tPA, especially in perfused CRVO, may provide signi- BRVO. Most patients with CRVO, both nonperfused and
ficant visual improvement, although study limitations, such perfused, may have a favorable anatomical response to this
as the lack of a control group, preclude definitive conclusions treatment as demonstrated by OCT images and measurements
to be drawn. demonstrating reduction in macular thickness and resolution of 1767
RETINA AND VITREOUS

the large cystic spaces in the outer plexiform layer within


several weeks of injection. However, a favorable visual acuity INTRODUCTION
response appears more likely in patients with perfused CRVO. Epidemiology
Re-treatment may also be necessary in some patients due to the HRVO is the least common type of venous occlusive disease. As
recurrence of macular edema. There may be intravitreal steroid- discussed previously, HRVO makes up only a fraction of the
related adverse events (including cataract and increased intra- incidence of vein occlusions. In the BMES, the overall incidence
ocular pressure) and injection-related adverse events (including of RVO was 1.6% and only 5.1% of those were HRVO.8
noninfectious and infectious endophthalmitis, retinal detach-
ment, vitreous hemorrhage, and lens injury) that could negate Pathogenesis
the benefit of reduction in macular edema. The SCORE study In approximately 20.3% of humans, a two-trunked central retinal
and the Posurdex trial will further clarify the role of cortico- vein persists proximal to the lamina cribosa.255 One of the two
steroids in CRVO. trunks may become occluded near the lamina cribrosa, as in
CRVO, to produce a HRVO.256,257 The trunks typically drain the
Anti-VEGF Agents superior and inferior halves of the retina, respectively. The risk
Experimental studies have shown that functional and structural factors of HRVO appear to be similar to those for CRVO.258
changes occur in the retinal capillaries induced by the hypoxic
environment after venous occlusion. This leads to increased Clinical Features
capillary permeability and accompanying retinal edema. VEGF, HRVO presents clinically with involvement of either the
SECTION 10

a 45kDa glycoprotein redundant,35 appears to play a role in this superior or inferior retinal hemisphere, (Fig. 132.13) although it
process. Numerous anti-VEGF agents are available. Pegaptanib may involve one-third to two-thirds of the retina.
(Macugen) and ranibizumab (Lucentis) have been approved by Visual prognosis appears to be better in HRVO than in CRVO.
the Food and Drug Administration (FDA) for the treatment Macular edema is common. Prognosis appears to correlate with
of neovascular age-related macular degeneration. The use of the extent of retinal perfusion. The extent of nonperfusion has
pegaptanib in a small, randomized series of patients with CRVO been reported to correlate positively with a worse prognosis.259
is currently in progress. Another anti-VEGF medication is In one study including 97 eyes, 32% of HRVO were nonper-
bevacizumab (Avastin), which is approved by the FDA for the fused, while 68% were perfused. Like CRVO, neovascularization
treatment of metastatic colon cancer, but has been used in off- is uncommon in perfused HRVO. The distribution of neovascu-
label settings for the treatment of numerous ocular conditions. larization is distinct from CRVO. The study showed that 13%
A single case report of intravitreal injection of bevacizumab in of nonperfused HRVO developed iris neovascularization, 29%
CRVO reported resolution of macular edema and improvement developed disk neovascularization, and 42% developed retinal
in visual acuity from 20/200 to 20/50 maintained at 4 weeks.252 neovascularization.51 Collateral vessels may form at the disk as
A retrospective study of 16 eyes in 15 patients with macular in CRVO as well as across the median raphe, as in BRVO.257
edema secondary to CRVO supported the previous case report.
The series included nine patients who had received previous Differential Diagnosis
intravitreal triamcinolone injections but had either not im- The differential diagnosis of HRVO is similar to that of CRVO
proved or developed intraocular pressure (IOP) elevation. The and BRVO, including diabetes mellitus, hypertensive retino-
patients received a mean of 2.8 injections of 1.25 mg in pathy, ocular ischemic syndrome, and radiation retinopathy.
0.05 mL of bevacizumab per eye. The mean baseline acuity was
20/600 that improved to a mean of 20/200 at 1 month, and Treatment
20/138 at 3 months. Halving of the visual angle was observed Although the utility of photocoagulation for prevention and
in 14 of the 16 eyes.253 There is a published report of intra- treatment of complications in HRVO has not been established,
vitreal bevacizumab injection for treatment of neovascular prophylactic application of scatter photocoagulation in the
glaucoma after failed intraocular pressure control with affected retinal hemisphere has been suggested in view of the
transscleral cyclophotocoagulation and PRP; the intraocular relatively higher rate of neovascular complications after
pressure improved within 2 days and the patient experienced
marked improvement in comfort.254 The long term risks and
effects of intravitreal anti-VEGF therapy remain unclear.
Until the results of long-term, randomized studies are avail-
able, the optimal management of CRVO will remain unclear.

HEMIRETINAL VEIN OCCLUSION

Synonym
• Hemivein occlusion

Key Features
• Hemi-retinal involvement in the distribution of the involved
occluded vein
• Dilated, tortuous veins
• Superficial and deep retinal hemorrhages
• Cotton-wool spots
• Optic nerve swelling/hemorrhages
• Macular/retinal swelling
• Optociliary shunt vessels on disk FIGURE 132.13. Typical appearance of an inferior hemicentral retinal
• Neovascularization of the optic disk, retina, or iris vein occlusion. Findings are similar to those of CRVO but affect only
1768 half of the retina.
Retinal Venous Occlusive Disease

nonperfused HRVO. Many of the treatment modalities for HRVO. Treatment decisions for HRVO will continue to be based
BRVO and CRVO may be beneficial in HRVO; however, formal on the discretion of the clinician until more study results
studies have not been performed due to the low incidence of become available.

REFERENCES
1. Orth DH, Patz A: Retinal branch vein variation in arteriovenous crossings. Am J 36. Rosen DA, Marshall J, Kohner EM, et al:
occlusion. Surv Ophthalmol 1978; 22:357. Ophthalmol 1992; 113:664. Experimental retinal branch vein occlusion
2. Branch Vein Occlusion Study Group: Argon 19. Zhao J, Sastry SM, Sperduto RD, et al: in rhesus monkeys. Ii. Retinal blood flow
laser photocoagulation for macular edema Arteriovenous crossing patterns in branch studies. Br J Ophthalmol 1979; 63:388.
in branch vein occlusion. Am J Ophthalmol retinal vein occlusion. The eye disease 37. The Eye Disease Case-control Study
1984; 98:271. case-control study group. Ophthalmology Group: Risk factors for branch retinal vein
3. Central Vein Occlusion Study: Baseline and 1993; 100:423. occlusion. Am J Ophthalmol 1993;
early natural history report. Arch 20. Michels RG, Gass JD: The natural course 116:286.
Ophthalmol 1993; 111:1087. of retinal branch vein obstruction. Trans Am 38. Martin SC, Butcher A, Martin N, et al:
4. Central Vein Occlusion Study Group: Acad Ophthalmol Otolaryngol 1974; Cardiovascular risk assessment in patients
Natural history and clinical management of 78:OP166. with retinal vein occlusion. Br J Ophthalmol
central retinal vein occlusion. Arch 21. Sedney SC: Photocoagulation in retinal 2002; 86:774.
Ophthalmol 1997; 115:486. vein occlusion. Doc Ophthalmol 1976; 40:1. 39. Shilling JS, Kohner EM: New vessel
5. Central Vein Occlusion Study Group: 22. Weinberg DV, Egan KM, Seddon JM: formation in retinal branch vein occlusion.

CHAPTER 132
Evaluation of grid pattern photocoagulation Asymmetric distribution of arteriovenous Br J Ophthalmol 1976; 60:810.
for macular edema in central vein occlusion. crossings in the normal retina. 40. Appiah AP, Trempe CL: Risk factors
The central vein occlusion study group m Ophthalmology 1993; 100:31. associated with branch vs. Central retinal
report. Ophthalmology 1995; 102:1425. 23. Christoffersen NL, Larsen M: vein occlusion. Ann Ophthalmol 1989;
6. Central Vein Occlusion Study Group: A Pathophysiology and hemodynamics of 21:153.
randomized clinical trial of early panretinal branch retinal vein occlusion. 41. Johnston RL, Brucker AJ, Steinmann W,
photocoagulation for ischemic central vein Ophthalmology 1999; 106:2054. et al: Risk factors of branch retinal vein
occlusion. The central vein occlusion study 24. Rabinowicz IM, Litman S, Michaelson IC: occlusion. Arch Ophthalmol 1985;
group n report. Ophthalmology 1995; Branch venous thrombosis—a pathological 103:1831.
102:1434. report. Trans Ophthalmol Soc UK 1969; 42. Branch Vein Occlusion Study Group: Argon
7. David R, Zangwill L, Badarna M, et al: 88:191. laser scatter photocoagulation for
Epidemiology of retinal vein occlusion and 25. Paton A, Rubinstein K, Smith VH: Arterial prevention of neovascularization and
its association with glaucoma and insufficiency in retinal venous occlusion vitreous hemorrhage in branch vein
increased intraocular pressure. (a short symposium). Trans Ophthalmol Soc occlusion. A randomized clinical trial. Arch
Ophthalmologica 1988; 197:69. UK 1964; 84:559. Ophthalmol 1986; 104:34.
8. Mitchell P, Smith W, Chang A: Prevalence 26. Bowers DK, Finkelstein D, Wolff SM, et al: 43. Gass JD: A fluorescein angiographic study
and associations of retinal vein occlusion in Branch retinal vein occlusion. A of macular dysfunction secondary to retinal
australia. The blue mountains eye study. clinicopathologic case report. Retina 1987; vascular disease. Ii. Retinal vein obstruction.
Arch Ophthalmol 1996; 114:1243. 7:252. Arch Ophthalmol 1968; 80:550.
9. Hayreh S: Ocular neovascularization in 27. Kumar B, Yu DY, Morgan WH, et al: The 44. Shilling JS: Vascular changes after retinal
central retinal vein occlusion. In: distribution of angioarchitectural changes branch vein occlusion. Trans Ophthalmol
Symposium on Central Vein Occlusion, within the vicinity of the arteriovenous Soc UK 1976; 96:193.
10th Annual Macula Society Meeting. crossing in branch retinal vein occlusion. 45. Clemett RS: Retinal branch vein occlusion.
France: Cannes. Ophthalmology 1998; 105:424. Changes at the site of obstruction. Br J
10. Gutman FA, Zegarra H: The natural course 28. Seitz R: The retinal vessels. In: The Ophthalmol 1974; 58:548.
of temporal retinal branch vein occlusion. crossing phenomenon. St Louis: Mosby; 46. Gutman FA: Macular edema in branch
Trans Am Acad Ophthalmol Otolaryngol 1964:20 retinal vein occlusion: prognosis and
1974; 78:OP178. 29. Jefferies P, Clemett R, Day T: An management. Trans Sect Ophthalmol Am
11. Blankenship GW, Okun E: Retinal tributary anatomical study of retinal arteriovenous Acad Ophthalmol Otolaryngol 1977;
vein occlusion. History and management crossings and their role in the pathogenesis 83:488.
by photocoagulation. Arch Ophthalmol of retinal branch vein occlusions. Aust N Z 47. Noma H, Funatsu H, Yamasaki M, et al:
1973; 89:363. J Ophthalmol 1993; 21:213. Pathogenesis of macular edema with
12. Joffe L, Goldberg RE, Magargal LE, et al: 30. Frangieh GT, Green WR, Barraquer- branch retinal vein occlusion and
Macular branch vein occlusion. Somers E, et al: Histopathologic study of intraocular levels of vascular endothelial
Ophthalmology 1980; 87:91. nine branch retinal vein occlusions. Arch growth factor and interleukin-6. Am J
13. Klein RKB, Moss SE, Meuer SM: The Ophthalmol 1982; 100:1132. Ophthalmol 2005; 140:256.
epidemiology of retinal vein occlusion: the 31. Kohner EM, Dollery CT, Shakib M, et al: 48. Schulman J, Jampol LM, Goldberg MF:
beaver dam eye study. Trans Am Experimental retinal branch vein occlusion. Large capillary aneurysms secondary to
Ophthalmol Soc 2000; 98:133. Am J Ophthalmol 1970; 69:778. retinal venous obstruction. Br J Ophthalmol
14. Hayreh SS, Zimmerman MB, Podhajsky P: 32. Hamilton AM, Marshall J, Kohner EM, et al: 1981; 65:36.
Incidence of various types of retinal vein Retinal new vessel formation following 49. Sanborn GE, Magargal LE: Venous
occlusion and their recurrence and experimental vein occlusion. Exp Eye Res macroaneurysm associated with branch
demographic characteristics. Am J 1975; 20:493. retinal vein obstruction. Ann Ophthalmol
Ophthalmol 1994; 117:429. 33. Hamilton AM, Kohner EM, Rosen D, et al: 1984; 16:464.
15. Jensen V: Clinical studies of tributary Experimental retinal branch vein occlusion 50. Brown GC, Kimmel AS, Magargal LE, et al:
thrombosis in central retinal vein. Acta in rhesus monkeys. I. Clinical appearances. Progressive capillary nonperfusion in
Ophthalmol Supp 1936; 10:1. Br J Ophthalmol 1979; 63:377. temporal branch retinal vein obstruction.
16. Duker JS, Brown GC: Anterior location of 34. Hockley DJ, Tripathi RC, Ashton N: Ann Ophthalmol 1989; 21:290.
the crossing artery in branch retinal vein Experimental retinal branch vein occlusion 51. Hayreh SS, Rojas P, Podhajsky P, et al:
obstruction. Arch Ophthalmol 1989; in the monkey. Histopathological and Ocular neovascularization with retinal
107:998. ultrastructural studies. Trans Ophthalmol vascular occlusion-iii. Incidence of ocular
17. Weinberg D, Dodwell DG, Fern SA: Soc UK 1976; 96:202. neovascularization with retinal vein
Anatomy of arteriovenous crossings in 35. Hockley DJ, Tripathi RC, Ashton N: occlusion. Ophthalmology 1983; 90:488.
branch retinal vein occlusion. Am J Experimental retinal branch vein occlusion 52. Joondeph HC, Joondeph BC: Posterior
Ophthalmol 1990; 109:298. in rhesus monkeys. Iii. Histopathological tractional retinal breaks complicating
18. Feist RM, Ticho BH, Shapiro MJ, et al: and electron microscopical studies. branch retinal vein occlusion. Retina
Branch retinal vein occlusion and quadratic Br J Ophthalmol 1979; 63:393. 1988; 8:136. 1769
RETINA AND VITREOUS

53. Joondeph HC, Goldberg MF: combined with gas/air tamponade for 88. Fujii GY, de Juan E Jr, Humayun MS:
Rhegmatogenous retinal detachment after treating macular edema associated with Improvements after sheathotomy for
tributary retinal vein occlusion. Am J branch retinal vein occlusion: Retinal branch retinal vein occlusion documented
Ophthalmol 1975; 80:253. tomography and visual outcome. Graefes by optical coherence tomography and
54. Gutman FA, Zegarra H: Retinal detachment Arch Clin Exp Ophthalmol 2001; 239:729. scanning laser ophthalmoscope.
secondary to retinal branch vein occlusions. 73. Thompson JT: What is the role of Ophthalmic Surg Lasers Imaging
Trans Sect Ophthalmol Am Acad vitrectomy for macular edema from branch 2003; 34:49.
Ophthalmol Otolaryngol 1976; 81:491. retinal vein occlusion? Am J Ophthalmol 89. Cahill MT, Fekrat S: Arteriovenous
55. Regenbogen L, Godel V, Feiler-Ofry V, et al: 2004; 138:1037. sheathotomy for branch retinal vein
Retinal breaks secondary to vascular 74. Stefansson E, Novack RL, Hatchell DL: occlusion. Ophthalmol Clin North Am 2002;
accidents. Am J Ophthalmol 1977; 84:187. Vitrectomy prevents retinal hypoxia in 15:417.
56. Ramos-Umpierre A, Berrocal JA: Retinal branch retinal vein occlusion. Invest 90. Mandelcorn MS, Nrusimhadevara RK:
detachment following branch vein occlusion: Ophthalmol Vis Sci 1990; 31:284. Internal limiting membrane peeling for
Case report. Ann Ophthalmol 1977; 9:339. 75. Osterloh MD, Charles S: Surgical decom- decompression of macular edema in retinal
57. Chess J, Eichen AL: Rhegmatogenous pression of branch retinal vein occlusions. vein occlusion: a report of 14 cases. Retina
retinal detachment associated with branch Arch Ophthalmol 1988; 106:1469. 2004; 24:348.
vein occlusion. Ann Ophthalmol 1989; 76. Opremcak EM, Bruce RA: Surgical 91. Han DP, Bennett SR, Williams DF, et al:
21:309. decompression of branch retinal vein Arteriovenous crossing dissection without
58. Zauberman H: Retinopathy of retinal occlusion via arteriovenous crossing separation of the retina vessels for
detachment after major vascular sheathotomy: a prospective review of 15 treatment of branch retinal vein occlusion.
occlusions. Br J Ophthalmol 1968; 52:117. cases. Retina 1999; 19:1. Retina 2003; 23:145.
SECTION 10

59. Schatz H, Yannuzzi L, Stransky TJ: Retinal 77. Shah GK, Sharma S, Fineman MS, et al: 92. Liu DT, Lai WW, Chan WM, et al:
detachment secondary to branch vein Arteriovenous adventitial sheathotomy for Sheathotomy for vein occlusion.
occlusion. Part I. Ann Ophthalmol 1976; the treatment of macular edema associated Ophthalmology 112:1321; author reply
8:1437. with branch retinal vein occlusion. Am J 1321, 2005.
60. Schatz H, Yannuzzi L, Stransky TJ: Retinal Ophthalmol 2000; 129:104. 93. Le Rouic JF, Bejjani RA, Rumen F, et al:
detachment secondary to branch vein 78. Mester U, Dillinger P: Vitrectomy with Adventitial sheathotomy for decompression
occlusion: Part ii. Ann Ophthalmol 1976; arteriovenous decompression and internal of recent onset branch retinal vein
8:1461. limiting membrane dissection in branch occlusion. Graefes Arch Clin Exp
61. Fekrat S, Finkelstein D: In: Regillo B, retinal vein occlusion. Retina 2002; 22:740. Ophthalmol 2001; 239:747.
Brown GC, Flynn HW, eds: Vitreoretinal 79. Mason J 3rd, Feist R, White M Jr, et al: 94. Kroll P, Meyer CH, Mester U, et al:
disease: the essentials. New York: Thieme Sheathotomy to decompress branch retinal Sheathotomy to decompress brvo.
Medical Publishers; 1999:117. vein occlusion: A matched control study. Ophthalmology 2005; 112:528.
62. Wilson DJ, Finkelstein D, Quigley HA, et al: Ophthalmology 2004; 111:540. 95. Shah GK: Adventitial sheathotomy for
Macular grid photocoagulation. An 80. Horio N, Horiguchi M: Effect of treatment of macular edema associated
experimental study on the primate retina. arteriovenous sheathotomy on retinal blood with branch retinal vein occlusion. Curr
Arch Ophthalmol 1988; 106:100. flow and macular edema in patients with Opin Ophthalmol 2000; 11:171.
63. Esrick E, Subramanian ML, Heier JS, et al: branch retinal vein occlusion. Am J 96. Scott IU: Vitreoretinal surgery for
Multiple laser treatments for macular Ophthalmol 2005; 139:739. complications of branch retinal vein
edema attributable to branch retinal vein 81. Yamaji H, Shiraga F, Tsuchida Y, et al: occlusion. Curr Opin Ophthalmol 2002;
occlusion. Am J Ophthalmol 2005; 139:653. Evaluation of arteriovenous crossing 13:161.
64. Early Treatment Diabetic Retinopathy Study sheathotomy for branch retinal vein 97. Jonas JB, Kreissig I, Sofker A, et al:
Research Group: Early photocoagulation occlusion by fluorescein videoangiography Intravitreal injection of triamcinolone for
for diabetic retinopathy. Ophthalmology and image analysis. Am J Ophthalmol diffuse diabetic macular edema. Arch
1991; 98:766. 2004; 137:834. Ophthalmol 2003; 121:57.
65. Hayreh SS, Rubenstein L, Podhajsky P: 82. Charbonnel J, Glacet-Bernard A, 98. Martidis A, Duker JS, Greenberg PB, et al:
Argon laser scatter photocoagulation in Korobelnik JF, et al: Management of branch Intravitreal triamcinolone for refractory
treatment of branch retinal vein occlusion. retinal vein occlusion with vitrectomy and diabetic macular edema. Ophthalmology
A prospective clinical trial. Ophthalmologica arteriovenous adventitial sheathotomy, the 2002; 109:920.
1993; 206:1. possible role of surgical posterior vitreous 99. Young S, Larkin G, Branley M, et al: Safety
66. Amirikia A, Scott IU, Murray TG, et al: detachment. Graefes Arch Clin Exp and efficacy of intravitreal triamcinolone for
Outcomes of vitreoretinal surgery for Ophthalmol 2004; 242:223. cystoid macular oedema in uveitis. Clin
complications of branch retinal vein 83. Kube T, Feltgen N, Pache M, et al: Experiment Ophthalmol 2001; 29:2.
occlusion. Ophthalmology 2001; 108:372. Angiographic findings in arteriovenous 100. Benhamou N, Massin P, Haouchine B, et al:
67. Ikuno Y, Tano Y, Lewis JM, et al: Retinal dissection (sheathotomy) for Intravitreal triamcinolone for refractory
detachment after branch retinal vein decompression of branch retinal vein pseudophakic macular edema. Am J
occlusion: Influence of the type of break on occlusion. Graefes Arch Clin Exp Ophthalmol 2003; 135:246.
the outcome of vitreous surgery. Ophthalmol 2005; 243:334. 101. Wilson CA, Berkowitz BA, Sato Y, et al:
Ophthalmology 1997; 104:27. 84. Lu L, Li Y, Yi C, et al: Preliminary clinical Treatment with intravitreal steroid reduces
68. Ikuno Y, Ikeda T, Sato Y, et al: Tractional observation of arteriovenous sheathotomy blood-retinal barrier breakdown due to
retinal detachment after branch retinal vein for treatment of branch retinal vein retinal photocoagulation. Arch Ophthalmol
occlusion. Influence of disc neovascu- occlusion. Yan Ke Xue Bao 2003; 19:33. 1992; 110:1155.
larization on the outcome of vitreous 85. Fujimoto R, Ogino N, Kumagai K, et al: 102. Fischer S, Renz D, Schaper W, et al: In
surgery. Ophthalmology 1998; 105:417. [the efficacy of arteriovenous adventitial vitro effects of dexamethasone on hypoxia-
69. Murakami K, Ho PC, Trempe CL, et al: sheathotomy for macular edema in branch induced hyperpermeability and expression
Tractional detachment of the macula retinal vein occlusion]. Nippon Ganka of vascular endothelial growth factor. Eur J
following branch retinal vein occlusion. Gakkai Zasshi 2004; 108:144. Pharmacol 2001; 411:231.
Ann Ophthalmol 1983; 15:760. 86. Cahill MT, Kaiser PK, Sears JE, et al: The 103. Lee H, Shah GK: Intravitreal triamcinolone
70. Smiddy WE, Isernhagen RD, Michels RG, effect of arteriovenous sheathotomy on as primary treatment of cystoid macular
et al: Vitrectomy for nondiabetic vitreous cystoid macular oedema secondary to edema secondary to branch retinal vein
hemorrhage. Retinal and choroidal vascular branch retinal vein occlusion. Br J occlusion. Retina 2005; 25:551.
disorders. Retina 1988; 8:88. Ophthalmol 2003; 87:1329. 104. Hayashi K, Hayashi H: Intravitreal versus
71. Tachi N, Hashimoto Y, Ogino N: Vitrectomy 87. Chalam KV, Shah GY, Shah VA: Vitrectomy retrobulbar injections of triamcinolone for
for macular edema combined with retinal with or without arteriovenous adventitial macular edema associated with branch
vein occlusion. Doc Ophthalmol 1999; sheathotomy for macular edema associated retinal vein occlusion. Am J Ophthalmol
97:465. with branch retinal vein occlusion. Am J 2005; 139:972.
72. Saika S, Tanaka T, Miyamoto T, et al: Ophthalmol 139:1146; author reply 1146, 105. Yepremyan M, Wertz FD, Tivnan T, et al:
1770 Surgical posterior vitreous detachment 2005. Early treatment of cystoid macular edema
Retinal Venous Occlusive Disease

secondary to branch retinal vein occlusion 125. McGrath MA, Wechsler F, Hunyor AB, et al: 143. Weger M, Stanger O, Deutschmann H,
with intravitreal triamcinolone acetonide. Systemic factors contributory to retinal vein et al: Hyperhomocyst(e)inemia and mthfr
Ophthalmic Surg Lasers Imaging 2005; occlusion. Arch Intern Med 1978; 138:216. c677t genotypes in patients with central
36:30. 126. Rath EZ, Frank RN, Shin DH, et al: Risk retinal vein occlusion. Graefes Arch Clin
106. Ozkiris A, Evereklioglu C, Erkilic K, et al: factors for retinal vein occlusions. A case- Exp Ophthalmol 2002; 240:286.
The efficacy of intravitreal triamcinolone control study. Ophthalmology 1992; 99:509. 144. Brown BA, Marx JL, Ward TP, et al:
acetonide on macular edema in branch 127. Priluck IA, Robertson DM, Hollenhorst RW: Homocysteine: A risk factor for retinal
retinal vein occlusion. Eur J Ophthalmol Long-term follow-up of occlusion of the venous occlusive disease. Ophthalmology
2005; 15:96. central retinal vein in young adults. Am J 2002; 109:287.
107. Jonas JB, Akkoyun I, Kamppeter B, et al: Ophthalmol 1980; 90:190. 145. Aras S, Yilmaz G, Alpas I, et al: Retinal vein
Branch retinal vein occlusion treated by 128. Tsaloumas MD, Kirwan J, Vinall H, et al: occlusion and factor v leiden and
intravitreal triamcinolone acetonide. Eye Nine year follow-up study of morbidity and prothrombin 20210 g:A mutations. Eur J
2005; 19:65. mortality in retinal vein occlusion. Eye Ophthalmol 2001; 11:351.
108. Tewari HK, Sony P, Chawla R, et al: 2000; 14:821. 146. Marcucci R, Bertini L, Giusti B, et al:
Prospective evaluation of intravitreal 129. Elman MJ, Bhatt AK, Quinlan PM, et al: The Thrombophilic risk factors in patients with
triamcinolone acetonide injection in risk for systemic vascular diseases and central retinal vein occlusion. Thromb
macular edema associated with retinal mortality in patients with central retinal vein Haemost 2001; 86:772.
vascular disorders. Eur J Ophthalmol 2005; occlusion. Ophthalmology 1990; 97:1543. 147. Johnson TM, El-Defrawy S, Hodge WG,
15:619. 130. Dodson PM, Galton DJ, Hamilton AM, et al: et al: Prevalence of factor v leiden and
109. Chen SD, Sundaram V, Lochhead J, et al: Retinal vein occlusion and the prevalence activated protein c resistance in central

CHAPTER 132
Intravitreal triamcinolone for the treatment of lipoprotein abnormalities. Br J retinal vein occlusion. Retina 2001; 21:161.
of ischemic macular edema associated Ophthalmol 1982; 66:161. 148. Weger M, Stanger O, Haas A:
with branch retinal vein occlusion. Am J 131. Green WR, Chan CC, Hutchins GM, et al: Hyperhomocysteinemia: a risk factor for
Ophthalmol 2006; 141:876. Central retinal vein occlusion: a prospective central retinal vein occlusion. Am J
110. Beer PM, Bakri SJ, Singh RJ, et al: histopathologic study of 29 eyes in 28 Ophthalmol 2001; 131:290.
Intraocular concentration and cases. Trans Am Ophthalmol Soc 1981; 149. Vine AK, Samama MM: The role of
pharmacokinetics of triamcinolone 79:371. abnormalities in the anticoagulant and
acetonide after a single intravitreal 132. Williamson TH, Rumley A, Lowe GD: Blood fibrinolytic systems in retinal vascular
injection. Ophthalmology 2003; 110:681. viscosity, coagulation, and activated occlusions. Surv Ophthalmol 1993; 37:283.
111. Scott IU IM: It’s time for a clinical trial to protein c resistance in central retinal vein 150. Svensson PJ, Dahlback B: Resistance to
investigate intravitreal triamcinolone for occlusion: A population controlled study. activated protein c as a basis for venous
macular edema due to retinal vein Br J Ophthalmol 1996; 80:203. thrombosis. N Engl J Med 1994; 330:517.
occlusion: The score study. Arch 133. Hayreh SS: Pathogenesis of occlusion of 151. Larsson J, Olafsdottir E, Bauer B: Activated
Ophthalmol 2005; 123:581. the central retinal vessels. Am J protein c resistance in young adults with
112. Ring CP, Pearson TC, Sanders MD, et al: Ophthalmol 1971; 72:998. central retinal vein occlusion. Br J
Viscosity and retinal vein thrombosis. 134. Hayreh SS: Prevalent misconceptions Ophthalmol 1996; 80:200.
Br J Ophthalmol 1976; 60:397. about acute retinal vascular occlusive 152. Arsene S, Delahousse B, Regina S, et al:
113. Remky A, Wolf S, Hamid M, et al: [effect of disorders. Prog Retin Eye Res 2005; Increased prevalence of factor v leiden in
hemodilution on retinal hemodynamics in 24:493. patients with retinal vein occlusion and
retinal branch vein occlusion]. 135. Gottlieb JL, Blice JP, Mestichelli B, et al: under 60 years of age. Thromb Haemost
Ophthalmologe 1994; 91:288. Activated protein c resistance, factor v 2005; 94:101.
114. Chen HC, Wiek J, Gupta A, et al: Effect of leiden, and central retinal vein occlusion in 153. Levine SR, Crofts JW, Lesser GR, et al:
isovolaemic haemodilution on visual young adults. Arch Ophthalmol 1998; Visual symptoms associated with the
outcome in branch retinal vein occlusion. 116:577. presence of a lupus anticoagulant.
Br J Ophthalmol 1998; 82:162. 136. Cahill MT, Stinnett SS, Fekrat S: Meta- Ophthalmology 1988; 95:686.
115. Janvrin SB, Davies G, Greenhalgh RM: analysis of plasma homocysteine, serum 154. Pulido JS, Ward LM, Fishman GA, et al:
Postoperative deep vein thrombosis folate, serum vitamin b(12), and Antiphospholipid antibodies associated
caused by intravenous fluids during thermolabile mthfr genotype as risk factors with retinal vascular disease. Retina 1987;
surgery. Br J Surg 1980; 67:690. for retinal vascular occlusive disease. Am J 7:215.
116. Noma H, Minamoto A, Funatsu H, et al: Ophthalmol 2003; 136:1136. 155. Kleiner RC, Najarian LV, Schatten S, et al:
Intravitreal levels of vascular endothelial 137. Janssen MC, den Heijer M, Cruysberg JR, Vaso-occlusive retinopathy associated with
growth factor and interleukin-6 are et al: Retinal vein occlusion: a form of antiphospholipid antibodies (lupus
correlated with macular edema in branch venous thrombosis or a complication of anticoagulant retinopathy). Ophthalmology
retinal vein occlusion. Graefes Arch Clin atherosclerosis? A meta-analysis of 1989; 96:896.
Exp Ophthalmol 2006; 244:309. thrombophilic factors. Thromb Haemost 156. Asherson RA, Merry P, Acheson JF, et al:
117. Gutman FA: Evaluation of a patient with 2005; 93:1021. Antiphospholipid antibodies: a risk factor
central retinal vein occlusion. 138. Chua B, Kifley A, Wong TY, et al: for occlusive ocular vascular disease in
Ophthalmology 1983; 90:481. Homocysteine and retinal vein occlusion: a systemic lupus erythematosus and the
118. Liebreich R: Apoplexia retinae. Albrecht population-based study. Am J Ophthalmol ‘primary’ antiphospholipid syndrome. Ann
von Graefes Arch Ophthalmol 1855; 1:346. 2005; 139:181. Rheum Dis 1989; 48:358.
119. Green WR: Retina. In: Spencer WH, ed: 139. Yildirim C, Yaylali V, Tatlipinar S, et al: 157. Snyers B, Lambert M, Hardy JP: Retinal
Ophthalmic pathology, an atlas and Hyperhomocysteinemia: a risk factor for and choroidal vaso-occlusive disease in
textbook. WB Saunders; 1985:589. retinal vein occlusion. Ophthalmologica systemic lupus erythematosus associated
120. Eye Disease Case-Control Study Group: 2004; 218:102. with antiphospholipid antibodies. Retina
Risk factors for central retinal vein 140. Cruciani F, Moramarco A, Curto T, et al: 1990; 10:255.
occlusion. Arch Ophthalmol 1996; 114:545. Mthfr c677t mutation, factor ii g20210a 158. Glacet-Bernard A, Bayani N, Chretien P,
121. Quinlan PM, Elman MJ, Bhatt AK, et al: The mutation and factor v leiden as risks factor et al: Antiphospholipid antibodies in retinal
natural course of central retinal vein for youth retinal vein occlusion. Clin Ter vascular occlusions. A prospective study of
occlusion. Am J Ophthalmol 1990; 110:118. 2003; 154:299. 75 patients. Arch Ophthalmol 1994;
122. Zegarra H, Gutman FA, Conforto J: The 141. Hvarfner C, Hillarp A, Larsson J: Influence 112:790.
natural course of central retinal vein of factor v leiden on the development of 159. Spalter HF: Abnormal serum proteins and
occlusion. Ophthalmology 1979; 86:1931. neovascularisation secondary to central retinal vein thrombosis. Arch Ophthalmol
123. Sinclair SH, Gragoudas ES: Prognosis for retinal vein occlusion. Br J Ophthalmol 1959; 62:868.
rubeosis iridis following central retinal vein 2003; 87:305. 160. Luxenberg MN, Mausolf FA: Retinal
occlusion. Br J Ophthalmol 1979; 63:735. 142. Abu El-Asrar AM, Abdel Gader AG, circulation in the hyperviscosity syndrome.
124. Zegarra H, Gutman FA, Zakov N, et al: Al-Amro SA, et al: Hyperhomocysteinemia Am J Ophthalmol 1970; 70:588.
Partial occlusion of the central retinal vein. and retinal vascular occlusive disease. Eur 161. Francis PJ, Stanford MR, Graham EM:
Am J Ophthalmol 1983; 96:330. J Ophthalmol 2002; 12:495. Dehydration is a risk factor for central 1771
RETINA AND VITREOUS

retinal vein occlusion in young patients. 180. Lerche RC, Schaudig U, Scholz F, et al: vein occlusion. Arch Ophthalmol 1995;
Acta Ophthalmol Scand 2003; 81:415. Structural changes of the retina in retinal 113:456.
162. Trope GE, Lowe GD, McArdle BM, et al: vein occlusion-imaging and quantification 198. Fekrat S, Goldberg MF, Finkelstein D:
Abnormal blood viscosity and haemostasis with optical coherence tomography. Laser-induced chorioretinal venous
in long-standing retinal vein occlusion. Ophthalmic Surg Lasers 2001; 32:272. anastomosis for nonischemic central or
Br J Ophthalmol 1983; 67:137. 181. Ozdemir H, Karacorlu M, Karacorlu S: branch retinal vein occlusion. Arch
163. Arend O, Remky A, Jung F, et al: Role of Serous macular detachment in central Ophthalmol 1998; 116:43.
rheologic factors in patients with acute retinal vein occlusion. Retina 2005; 25:561. 199. Leonard BC, Coupland SG, Kertes PJ,
central retinal vein occlusion. 182. Cugati S, Cikamatana L, Wang JJ, et al: et al: Long-term follow-up of a modified
Ophthalmology 1996; 103:80. Five-year incidence and progression of technique for laser-induced chorioretinal
164. Glacet-Bernard A, Chabanel A, Lelong F, vascular retinopathy in persons without venous anastomosis in nonischemic central
et al: Elevated erythrocyte aggregation in diabetes: The blue mountains eye study. retinal vein occlusion. Ophthalmology 2003;
patients with central retinal vein occlusion Eye 2005. 110:948.
and without conventional risk factors. 183. Magargal LE, Donoso LA, Sanborn GE: 200. Browning DJ: Fundus photographic,
Ophthalmology 1994; 101:1483. Retinal ischemia and risk of fluorescein angiographic, and indocyanine
165. Dryden RM: Central retinal vein occlusions neovascularization following central retinal green angiographic signs in successful
and chronic simple glaucoma. Arch vein obstruction. Ophthalmology 1982; laser chorioretinal venous anastomosis for
Ophthalmol 1965; 73:659. 89:1241. central retinal vein occlusion.
166. Bertelsen T: The relationship between 184. Ferris FL 3rd SR: Standardized illumination Ophthalmology 1999; 106:2261.
thrombosis in the retinal veins and primary for visual acuity testing in clinical research. 201. Browning DJ, Antoszyk AN: Laser
glaucoma. Acta Ophthalmol (Copenh) Am J Ophthalmol 1982; 94:97. chorioretinal venous anastomosis for
SECTION 10

1961; 39:603. 185. Weinberg D, Jampol LM, Schatz H, et al: nonischemic central retinal vein occlusion.
167. Fuller JJ, Mason JO 3rd, White MF Jr, et al: Exudative retinal detachment following Ophthalmology 1998; 105:670.
Retinochoroidal collateral veins protect central and hemicentral retinal vein 202. McAllister IL, Douglas JP, Constable IJ,
against anterior segment neovascularization occlusions. Arch Ophthalmol 1990; 108:271. et al: Laser-induced chorioretinal venous
after central retinal vein occlusion. Arch 186. Brown GC: Central retinal vein obstruction anastomosis for nonischemic central retinal
Ophthalmol 2003; 121:332. with lipid exudate. Arch Ophthalmol 1989; vein occlusion: Evaluation of the
168. Lahey JM, Kearney JJ, Tunc M: 107:1001. complications and their risk factors. Am J
Hypercoagulable states and central retinal 187. Duker JS, Brown GC: The efficacy of Ophthalmol 1998; 126:219.
vein occlusion. Curr Opin Pulm Med 2003; panretinal photocoagulation for 203. Luttrull JK: Epiretinal membrane and
9:385. neovascularization of the iris after central traction retinal detachment complicating
169. Lahey JM, Tunc M, Kearney J, et al: retinal artery obstruction. Ophthalmology laser-induced chorioretinal venous
Laboratory evaluation of hypercoagulable 1989; 96:92. anastomosis. Am J Ophthalmol 1997;
states in patients with central retinal vein 188. Boyd SR, Zachary I, Chakravarthy U, et al: 123:698.
occlusion who are less than 56 years of Correlation of increased vascular endothelial 204. Eccarius SG, Moran MJ, Slingsby JG:
age. Ophthalmology 2002; 109:126. growth factor with neovascularization and Choroidal neovascular membrane after
170. Coats G: Thrombosis of the central vein of permeability in ischemic central vein laser-induced chorioretinal anastomosis.
the retina. R Long Ophthalmic Hosp Rep occlusion. Arch Ophthalmol 2002; Am J Ophthalmol 1996; 122:590.
1904; 16:62. 120:1644. 205. Browning DJ, Rotberg MH: Vitreous
171. Laatikainen L, Kohner EM, Khoury D, et al: 189. Pe’er J, Folberg R, Itin A, et al: Vascular hemorrhage complicating laser-induced
Panretinal photocoagulation in central endothelial growth factor upregulation in chorioretinal anastomosis for central retinal
retinal vein occlusion: a randomised human central retinal vein occlusion. vein occlusion. Am J Ophthalmol 1996;
controlled clinical study. Br J Ophthalmol Ophthalmology 1998; 105:412. 122:588.
1977; 61:741. 190. Chen KH, Wu CC, Roy S, et al: Increased 206. Bavbek T, Yenice O, Toygar O: Problems
172. Minturn J, Brown GC: Progression of interleukin-6 in aqueous humor of with attempted chorioretinal venous
nonischemic central retinal vein obstruction neovascular glaucoma. Invest Ophthalmol anastomosis by laser for nonischemic crvo
to the ischemic variant. Ophthalmology Vis Sci 1999; 40:2627. and brvo. Ophthalmologica 2005; 219:267.
1986; 93:1158. 191. Blinder KJ, Khan JA, Giangiacomo J, et al: 207. Fekrat S, de Juan E Jr: Chorioretinal
173. Servais GE, Thompson HS, Hayreh SS: Optociliary veins and visual prognosis after venous anastomosis for central retinal vein
Relative afferent pupillary defect in central central retinal vein occlusion. Ann occlusion: Transvitreal venipuncture.
retinal vein occlusion. Ophthalmology 1986; Ophthalmol 1989; 21:192. Ophthalmic Surg Lasers 1999; 30:52.
93:301. 192. Im CY, Lee SY, Kwon OW: Collateral 208. Peyman GA, Kishore K, Conway MD:
174. Hayreh SS, Zimmerman MB, Beri M, et al: vessels in branch retinal vein occlusion. Surgical chorioretinal venous anastomosis
Intraocular pressure abnormalities associated Korean J Ophthalmol 2002; 16:82. for ischemic central retinal vein occlusion.
with central and hemicentral retinal vein 193. Mruthyunjaya P, Wirostko WJ, Ophthalmic Surg Lasers 1999; 30:605.
occlusion. Ophthalmology 2004; 111:133. Chandrashekhar R, et al: Central retinal 209. Kado M, Jalkh AE, Yoshida A, et al:
175. Hayreh SS: Classification of central retinal vein occlusion in patients treated with long- Vitreous changes and macular edema in
vein occlusion. Ophthalmology 1983; term warfarin sodium (coumadin) for central retinal vein occlusion. Ophthalmic
90:458. anticoagulation. Retina 2006; 26:285. Surg 1990; 21:544.
176. Magargal LE, Brown GC, Augsburger JJ, 194. Hansen LL, Wiek J, Schade M, et al: Effect 210. Hikichi T, Konno S, Trempe CL: Role of the
et al: Neovascular glaucoma following and compatibility of isovolaemic vitreous in central retinal vein occlusion.
central retinal vein obstruction. haemodilution in the treatment of Retina 1995; 15:29.
Ophthalmology 1981; 88:1095. ischaemic and non-ischaemic central 211. Sekiryu T, Yamauchi T, Enaida H, et al:
177. May DR, Klein ML, Peyman GA, et al: retinal vein occlusion. Ophthalmologica Retina tomography after vitrectomy for
Xenon arc panretinal photocoagulation for 1989; 199:90. macular edema of central retinal vein
central retinal vein occlusion: A randomised 195. Wolf S, Arend O, Bertram B, et al: occlusion. Ophthalmic Surg Lasers 2000;
prospective study. Br J Ophthalmol 1979; Hemodilution therapy in central retinal vein 31:198.
63:725. occlusion. One-year results of a 212. Opremcak EM, Rehmar AJ, Ridenour CD,
178. Matsui Y, Katsumi O, Mehta MC, et al: prospective randomized study. Graefes et al: Radial optic neurotomy for central
Correlation of electroretinographic and Arch Clin Exp Ophthalmol 1994; 232:33. retinal vein occlusion: 117 consecutive
fluorescein angiographic findings in 196. Hayreh SS, Klugman MR, Podhajsky P, et al: cases. Retina 2006; 26:297.
unilateral central retinal vein obstruction. Argon laser panretinal photocoagulation in 213. Lit ES, Tsilimbaris M, Gotzaridis E, et al:
Graefes Arch Clin Exp Ophthalmol 1994; ischemic central retinal vein occlusion. A Lamina puncture: Pars plana optic disc
232:449. 10-year prospective study. Graefes Arch surgery for central retinal vein occlusion.
179. Morrell AJ, Thompson DA, Gibson JM, Clin Exp Ophthalmol 1990; 228:281. Arch Ophthalmol 2002; 120:495.
et al: Electroretinography as a prognostic 197. McAllister IL, Constable IJ: Laser-induced 214. Vasco-Posada J: Modification of the
indicator or neovascularisation in crvo. Eye chorioretinal venous anastomosis for circulation in the posterior pole of the eye.
1772 1991; 5(Pt 3):362. treatment of nonischemic central retinal Ann Ophthalmol 1972; 4:48.
Retinal Venous Occlusive Disease

215. Arciniegas A: Treatment of the occlusion of neurotomy for central retinal vein occlusion. 245. Jonas JB, Kreissig I, Degenring RF:
the central retinal vein by section of the Jpn J Ophthalmol 2006; 50:158. Intravitreal triamcinolone acetonide as
posterior ring. Ann Ophthalmol 1984; 231. Hattenbach LO, Wellermann G, Steinkamp treatment of macular edema in central
16:1081. GW, et al: Visual outcome after treatment retinal vein occlusion. Graefes Arch Clin
216. Nagpal M, Nagpal K, Bhatt C, et al: Role of with low-dose recombinant tissue Exp Ophthalmol 2002; 240:782.
early radial optic neurotomy in central plasminogen activator or hemodilution in 246. Ip M, Kahana A, Altaweel M: Treatment of
retinal vein occlusion. Indian J Ophthalmol ischemic central retinal vein occlusion. central retinal vein occlusion with
2005; 53:115. Ophthalmologica 1999; 213:360. triamcinolone acetonide: An optical
217. Martinez-Jardon CS, Meza-de Regil A, 232. Lahey JM, Fong DS, Kearney J: Intravitreal coherence tomography study. Semin
Dalma-Weiszhausz J, et al: Radial optic tissue plasminogen activator for acute Ophthalmol 2003; 18:67.
neurotomy for ischaemic central vein central retinal vein occlusion. Ophthalmic 247. Degenring RF, Kamppeter B, Kreissig I,
occlusion. Br J Ophthalmol 2005; 89:558. Surg Lasers 1999; 30:427. et al: Morphological and functional
218. Garciia-Arumii J, Boixadera A, Martinez- 233. Glacet-Bernard A, Kuhn D, Vine AK, et al: changes after intravitreal triamcinolone
Castillo V, et al: Chorioretinal anastomosis Treatment of recent onset central retinal acetonide for retinal vein occlusion. Acta
after radial optic neurotomy for central vein occlusion with intravitreal tissue Ophthalmol Scand 2003; 81:399.
retinal vein occlusion. Arch Ophthalmol plasminogen activator: A pilot study. Br J 248. Ip MS, Gottlieb JL, Kahana A, et al:
2003; 121:1385. Ophthalmol 2000; 84:609. Intravitreal triamcinolone for the treatment
219. Weizer JS, Stinnett SS, Fekrat S: Radial 234. Elman MJ, Raden RZ, Carrigan A: of macular edema associated with central
optic neurotomy as treatment for central Intravitreal injection of tissue plasminogen retinal vein occlusion. Arch Ophthalmol
retinal vein occlusion. Am J Ophthalmol activator for central retinal vein occlusion. 2004; 122:1131.

CHAPTER 132
2003; 136:814. Trans Am Ophthalmol Soc 2001; 99:219. 249. Bashshur ZF, Ma’luf RN, Allam S, et al:
220. Patelli F, Radice P, Zumbo G, et al: Optical 235. Ghazi NG, Noureddine B, Haddad RS, et Intravitreal triamcinolone for the
coherence tomography evaluation of al: Intravitreal tissue plasminogen activator management of macular edema due to
macular edema after radial optic neurotomy in the management of central retinal vein nonischemic central retinal vein occlusion.
in patients affected by central retinal vein occlusion. Retina 2003; 23:780. Arch Ophthalmol 2004; 122:1137.
occlusion. Semin Ophthalmol 2004; 19:21. 236. Lam HD, Blumenkranz MS: Treatment of 250. Krepler K, Ergun E, Sacu S, et al:
221. Kim TW, Lee SJ, Kim SD. Comparative central retinal vein occlusion by vitrectomy Intravitreal triamcinolone acetonide in
evaluation of radial optic neurotomy and with lysis of vitreopapillary and epipapillary patients with macular oedema due to
panretinal photocoagulation in the adhesions, subretinal peripapillary tissue central retinal vein occlusion. Acta
management of central retinal vein plasminogen activator injection, and Ophthalmol Scand 2005; 83:71.
occlusion. Korean J Ophthalmol. 2005; photocoagulation. Am J Ophthalmol 2002; 251. Williamson TH ODA: Intravitreal triamcinolone
19:269. 134:609. acetonide for cystoid macular edema in
222. Martinez-Jardon CS, Meza-de Regil A, 237. Paques M, Vallee JN, Herbreteau D, et al: nonischemic central retinal vein occlusion.
Dalma-Weiszhausz J, et al. Radial optic Superselective ophthalmic artery fibrinolytic Am J Ophthalmol. 2005; 139:860.
neurotomy for ischaemic central vein therapy for the treatment of central retinal 252. Rosenfeld PJ, Fung AE, Puliafito CA:
occlusion. Br J Ophthalmol. 2005; 89:558. vein occlusion. Br J Ophthalmol 2000; Optical coherence tomography findings
223. Hayreh SS: Radial optic neurotomy for 84:1387. after an intravitreal injection of bevacizumab
central retinal vein occlusion. Retina 2002; 238. Weiss JN: Treatment of central retinal vein (avastin) for macular edema from central
22:374. occlusion by injection of tissue retinal vein occlusion. Ophthalmic Surg
224. Williamson TH, Poon W, Whitefield L, et al: plasminogen activator into a retinal vein. Lasers Imaging 2005; 36:336.
A pilot study of pars plana vitrectomy, Am J Ophthalmol 1998; 126:142. 253. Iturralde D, Spaide RF, Meyerle CB, et al:
intraocular gas, and radial neurotomy in 239. Weiss JN, Bynoe LA: Injection of tissue Intravitreal bevacizumab (avastin) treatment
ischaemic central retinal vein occlusion. plasminogen activator into a branch retinal of macular edema in central retinal vein
Br J Ophthalmol 2003; 87:1126. vein in eyes with central retinal vein occlusion: A short-term study. Retina 2006;
225. Schneider U, Inhoffen W, Grisanti S, et al: occlusion. Ophthalmology 2001; 108:2249. 26:279.
Characteristics of visual field defects by 240. Bynoe LA, Weiss JN: Retinal endovascular 254. Kahook MY, Schuman JS, Noecker RJ:
scanning laser ophthalmoscope surgery and intravitreal triamcinolone Intravitreal bevacizumab in a patient with
microperimetry after radial optic neurotomy acetonide for central vein occlusion in neovascular glaucoma. Ophthalmic Surg
for central retinal vein occlusion. Retina young adults. Am J Ophthalmol 2003; Lasers Imaging 2006; 37:144.
2005; 25:704. 135:382. 255. Chopdar A: Dual trunk central retinal vein
226. Takaya K, Suzuki Y, Nakazawa M: Massive 241. Bynoe LA, Hutchins RK, Lazarus HS, et al: incidence in clinical practice. Arch
hemorrhagic retinal detachment during Retinal endovascular surgery for central Ophthalmol 1984; 102:85.
radial optic neurotomy. Graefes Arch Clin retinal vein occlusion: initial experience of 256. Chopdar A: Hemi-central retinal vein
Exp Ophthalmol 2006; 244:265. four surgeons. Retina 2005; 25:625. occlusion. Pathogenesis, clinical features,
227. Samuel MA, Desai UR, Gandolfo CB: 242. Shaikh S, Blumenkranz MS: Transient natural history and incidence of dual trunk
Peripapillary retinal detachment after radial improvement in visual acuity and macular central retinal vein. Trans Ophthalmol Soc
optic neurotomy for central retinal vein edema in central retinal vein occlusion UK 1982; 102 (Pt 2):241.
occlusion. Retina 2003; 23:580. accompanied by inflammatory features 257. Hayreh SS, Hayreh MS: Hemi-central
228. Bakri SJ, Beer PM: Choroidal after pulse steroid and anti-inflammatory retinal vein occulsion. Pathogenesis,
neovascularization after radial optic therapy. Retina 2001; 21:176. clinical features, and natural history. Arch
neurotomy for central retinal vein occlusion. 243. Greenberg PB, Martidis A, Rogers AH, et al: Ophthalmol 1980; 98:1600.
Retina 2004; 24:610. Intravitreal triamcinolone acetonide for 258. Appiah AP, Trempe CL: Differences in
229. Hattenbach LO, Steinkamp G, Scharrer I, macular oedema due to central retinal vein contributory factors among hemicentral,
et al: Fibrinolytic therapy with low-dose occlusion. Br J Ophthalmol 2002; 86:247. central, and branch retinal vein occlusions.
recombinant tissue plasminogen activator 244. Ip MS, Kumar KS: Intravitreous Ophthalmology 1989; 96:364.
in retinal vein occlusion. Ophthalmologica triamcinolone acetonide as treatment for 259. Zhang H, Xia Y: [analysis of visual
1998; 212:394. macular edema from central retinal vein prognosis and correlative factors in retinal
230. Tsujikawa A, Hangai M, Kikuchi M, et al: occlusion. Arch Ophthalmol 2002; vein occlusion]. Zhonghua Yan Ke Za Zhi
Visual field defect after radial optic 120:1217. 2002; 38:98.

1773
CHAPTER
Diagnosis, Management, and Treatment of
133 Nonproliferative Diabetic Retinopathy
Lloyd P. Aiello, Jerry Cavallerano, Manvi Prakash, and Lloyd M. Aiello

SIGNIFICANCE OF APPROACHING Nevertheless, DR remains a leading cause of new-onset


HIGH-RISK PROLIFERATIVE DIABETIC blindness in the United States for citizens between the ages of
RETINOPATHY AND CLINICALLY 20 and 74 years. Today this blindness usually results from non-
SIGNIFICANT DIABETIC MACULAR EDEMA resolving vitreous hemorrhage, traction retinal detachment, or
DME. However, the 5-year risk of severe visual loss (SVL –
Studies first demonstrated evidence of the benefit of scatter visual acuity of 5/200 or worse on two consecutive visits 4
(panretinal) laser photocoagulation (PRP) surgery to treat months apart) can be reduced to less than 5% if a person with
diabetic retinopathy (DR) in 1967.1 (see Table 133.1 for a list of DR approaching or just reaching high-risk proliferative retino-
common abbreviations used in this chapter). Since these pathy (defined below) receives timely PRP. Furthermore, people
promising beginnings, dramatic strides in treating proliferative with clinically significant diabetic macular edema (CSME) can
diabetic retinopathy (PDR) and diabetic macular edema (DME) have the risk of moderate visual loss (MVL - doubling of the
have been made through the effective use of PRP and other visual angle i.e., 20/20 to 20/40) reduced to ~12% or less if they
surgical techniques. These benefits have been strongly sup- undergo appropriate focal laser surgery. Along with the
ported by the findings of three major nationwide randomized mainstays of current therapy which include laser surgery and
controlled clinical trials: the Diabetic Retinopathy Study intensive glycemic, blood pressure and lipid control, newer and
(DRS),2–15 the Early Treatment Diabetic Retinopathy Study evolving strategies for eye care include oral protein kinase C
(ETDRS),16–34 and the Diabetic Retinopathy Vitrectomy inhibitors and intravitreal injections of corticosteroids, and
Study (DRVS).35–39 With appropriate diagnosis, timely antiangiogenic agents. These novel therapies hold the promise
intervention and careful follow-up, the risk of severe visual of providing additional efficacious treatment modalities, with
loss from PDR can be virtually eliminated. fewer side effects than present-day interventions.
Since diabetic retinopathy is often asymptomatic in its most
treatable stages, its early detection through regularly scheduled
evaluations of DR and DME severity is critical. This chapter
TABLE 133.1. Abbreviations and Definitions reviews the prognostic implications of the lesions of diabetic
retinopathy and the risks of progression, with particular
PDR Proliferative diabetic retinopathy emphasis on identifying patients at risk of visual loss and in
NPDR Nonproliferative diabetic retinopathy need of laser surgery. The laser treatment techniques are only
generally described in this chapter but are discussed in the
H/Ma Hemorrhages or microaneurysms, or both
chapter on Proliferative DR and are carefully detailed in ETDRS
HE Hard exudates Report no 3 & 4.18,19
SE (CWS) Soft exudates (cotton-wool spots)
EPIDEMIOLOGY OF DIABETIC
VB Venous beading
RETINOPATHY
IRMA Intraretinal microvascular abnormality
The Center for Disease Control and Prevention estimates that
NVD New vessels on or within 1 disk diameter of disc
20.8 million people or 7.0% of the total U.S. population have
margin
diabetes, of which 6.2 million people are undiagnosed.40,41 The
NVE New vessels elsewhere in the retina outside of disc vast majority (>90%) of diabetic patients have type 2 diabetes,
and 1 disc diameter from disk margin which is usually diagnosed after 40 years of age, although the
FPD Fibrous proliferations on or within 1 disc diameter of prevalence of type 2 diabetes is increasing in children and
disc margin adolescent populations.42 People with type 1 diabetes have a
FPE Fibrous proliferations elsewhere, not FPD lack of insulin production, generally requiring insulin therapy,
while people with type 2 diabetes initially demonstrate insulin
SVL Severe visual loss: Visual acuity ≤5/200 at two resistance, although they may also eventually require insulin
consecutive completed 4-mo follow-up visits
treatment as the disease progresses or to maximize diabetes
MVL Moderate visual loss: A doubling of the visual angle control. Although, the likelihood of developing some level of
(e.g., 20/40 to 20/80 at two consecutive completed diabetic retinopathy during a lifetime is somewhat higher for
4-mo follow-up visits) persons with type 1 diabetes, people with type 2 diabetes
CSME Clinically significant macular edema account for the majority of clinical cases of diabetic eye disease
because of their overall larger numbers. 1775
RETINA AND VITREOUS

TABLE 133.2. Medical Complications

Condition Comment

Risk Indicators of Diabetic Retinopathy


Joint contractures Association of retinopathy and contractures has been established. Eye examination is indicated.
Care of joint contractures is important.
Neuropathy Peripheral neuropathy may result in difficulty handling contact lenses. Neuropathy in lower extremities
may alter mobility in such a way that restoration and maintenance of as much vision as possible is
important. Cardiovascular autonomic neuropathy is an independent risk factor for proliferative
diabetic retinopathy.
Conditions That May Affect the Course of Diabetic Retinopathy
Hypertension Appropriate medical treatment is indicated for prevention of cardiovascular disease, stroke, and death.
Hypertension itself may result in hypertensive retinopathy superimposed on diabetic retinopathy.
Elevated triglycerides and lipids Appropriate management to normalize is important. Proper diet and reduced levels may result in less
retinal vessel leakage.
Proteinuria; elevated creatinine Aggressive management of renal disease is indicated to avoid renal retinopathy, which may increase
SECTION 10

risk of progression of diabetic retinopathy.


Cardiovascular disease Increased risk of cardiac disease, particularly coronary vascular disease, is often associated with an
increase in the attenuation and arteriosclerotic closure of the arterial system of the retina.
A decreased risk of hemorrhage into the vitreous may result, but there also may be a decrease in
retinal function with associated decrease in vision. Management of cardiovascular disease may help
relieve some of the ischemic process in the retina. Aggressive cardiovascular management is
important.
Clinical trials There are no clinical trials that have specifically shown that control of systemic conditions that may
affect the eyes (the four previous entries) prevents the progression of diabetic retinopathy. However,
clinical experience suggests an association with the systemic benefits of appropriate treatment of
these problems.

DR is a highly specific vascular complication common to of DR. (Table 133.2). These factors include pregnancy49–51 and
both type 1 and type 2 diabetes mellitus. Duration of diabetes the metabolic syndromes like chronic hyperglycemia,52–55
is a significant risk factor for the development of retinopathy. hypertension,56 renal disease,54 abdominal obesity, hypercholes-
After 20 years of diabetes, nearly all patients with type 1 terolemia, and dyslipidemia.34,57,58 Cardiovascular and
diabetes, and more than 60% of patients with type 2 diabetes, peripheral autonomic neuropathies are associated with the
have some degree of retinopathy.43,44 A pooled data analysis presence of PDR.59 Patients with these conditions require
of eight population-based surveys estimates that among ~10.2 careful medical evaluation and treatment, diligent follow-up for
million US adults known to have diabetes who are age 40 years progression of diabetic retinopathy, and consideration of early
or older, the prevalence rates for DR and sight-threatening laser photocoagulation surgery.
retinopathy are 40.3% and 8.2%, respectively.45 Overall, DR Research studies are investigating how lifestyle changes can
affects ~4.1 million US adults age ≥ 40 years, and one out of delay or even prevent the onset of type 2 diabetes in individuals
every 12 persons in this age group has advanced, vision- at high-risk such as >20 million Americans with impaired
threatening retinopathy.46 glucose tolerance. The Diabetes Prevention Program, a study
At the present time, there are no known cures for DR or looking at preventing type 2 diabetes in people at high risk,
DME, and no known means to completely prevent these con- found that the development of diabetes was reduced 58% over
ditions from occurring. Laser photocoagulation surgery reduces 3 years by lifestyle interventions including diet and physical
the risk of moderate vision loss from DME and severe vision activity.60 The study also found that in some populations the
loss from PDR, but in general does not restore vision once loss drug metformin reduced the risk of developing diabetes by 31%
has occurred. Laser treatment is generally most effective when over 3 years. The group with greatest response consisted of
initiated at the time a person approaches or just reaches younger (20–40 years old) people 50–80 pounds overweight.
‘high-risk PDR’ and before the visual acuity is lost from DME.24 Another medication, acarbose, was found to reduce the risk of
The 5-year risk of SVL from untreated high-risk PDR may be as developing diabetes by 25% over 3 years in the STOP-NIDDM
high as 60%. DME will develop in up to 10% of all diabetic Trial.61
patients. In 4% of cases, DME affects the central fovea, and, up
to 30% of patients with CSME will develop MVL.47 Since PDR FUNDAMENTAL CLINICAL TRIALS
and CSME may cause no ocular or visual symptoms when the
retinal lesions are most amenable to treatment, the overriding Large randomized clinical trials have largely determined the
concern is to identify eyes at risk of visual loss and ensure that strategies for appropriate clinical eye care and management of
patients receive timely evaluation and initiation of laser surgery patients with diabetic retinopathy.
and other interventions when indicated. Even minor clinical The DRS (Table 133.3) conclusively demonstrated that PRP
errors in diagnosing the severity of retinopathy (Table 133.9) significantly reduces the risk of SVL from PDR, particularly
can result in delay in appropriate care plans and significantly when high-risk PDR is present.
increase the person’s risk of visual loss.48 The ETDRS provided valuable information concerning the
In addition, identification and optimum control of coexisting timing of PRP for advancing diabetic retinopathy and
health and medical problems are critically important as they conclusively demonstrated that focal photocoagulation for
1776 present a significant risk for the development and progression CSME reduces the risk of MVL by 50% or more (Table 133.4).
Diagnosis, Management, and Treatment of Nonproliferative Diabetic Retinopathy

TABLE 133.3. Diabetic Retinopathy Study TABLE 133.4. Early Treatment Diabetic Retinopathy Study

Major Eligibility Criteria Major Eligibility Criteria


1. Visual acuity: ≥20/100 in each eye. 1. Visual acuity: ≥20/40 (≥20/400 if reduction caused by macular
edema).
2. PDR in at least one eye or severe NPDR in both.
2. Mild to very severe NPDR and/or non-high-risk PDR, with or
3. Both eyes suitable for photocoagulation. without macular edema.
Major Design Features 3. Both eyes suitable for photocoagulation.
1. One eye of each patient was assigned randomly to Major Design Features
photocoagulation (scatter [panretinal], local [direct confluent
treatment of surface new vessels], and focal [for macular edema] 1. One eye of each patient assigned randomly to early
as appropriate). The other eye was assigned to follow up without photocoagulation and the other to deferral (careful follow-up and
photocoagulation. photocoagulation if high-risk PDR develops).
2. The eye assigned to treatment was then randomly assigned to 2. Patients assigned randomly to aspirin or placebo.
argon laser or xenon arc photocoagulation.
3. Eyes with diabetic macular edema were assigned to immediate
Major Conclusions or deferred focal photocoagulation.

CHAPTER 133
1. Photocoagulation reduced risk of severe visual loss by 50% or Major Conclusions
more. (SVL = VA* <5/200 at two consecutively completed
4-month follow-up visits.) 1. Focal photocoagulation (direct laser for focal leaks and grid laser
for diffuse leaks) reduced the risk of moderate visual loss
2. Modest risks of decrease in visual acuity (usually only 1 line) and (doubling of the visual angle) by 50% or more and increased the
visual field (risks greater with xenon than argon chance of a small improvement in visual acuity.
photocoagulation).
2. Both early scatter with or without focal photocoagulation and
3. Treatment benefit outweighs risks for eyes with high-risk PDR deferral were followed by low rates of severe visual loss (5-yr
(50% 5-yr rate of SVL in such eyes without treatment was rates in deferral subgroups were 2–10%; in early
reduced to 20% by treatment). photocoagulation groups these rates were 2–6%).
Prepared by M. Davis, M.D. and the ETDRS Research Group for the American 3. Focal photocoagulation should be considered for eyes with CSME.
Academy of Ophthalmology Diabetes 2000 Program
*VA = visual activity 4. Scatter photocoagulation is not indicated for mild to moderate
NPDR but should be considered as retinopathy approaches the
high-risk stage and usually should not be delayed when the
high-risk stage is present.
Furthermore, the ETDRS demonstrated that both early PRP Prepared by M. Davis, M.D. and the ETDRS Research Group for the American
(before the onset of high-risk PDR) and deferral of treatment Academy of Ophthalmology Diabetes 2000 Program.
‘until and as soon as high-risk PDR developed’ are effective in
reducing the risk of SVL. PRP, therefore, should be considered
as an eye approaches the high-risk PDR and “usually should with type 1 diabetes of moderate duration.63 The group taking
not be delayed if the eye has reached the high-risk proliferative sorbinil, however, did show a slightly slower progression rate
stage.”24 in microaneurysm count. Findings also showed no beneficial
Subsequent analysis of the ETDRS data shows that early PRP effect of sorbinil on DR.64 There were complications from the
is particularly beneficial for patients with type 2 diabetes.62 In use of this drug in the study population. Nearly 7% of the initial
type 2 diabetes patients, early PRP substantially reduced sub- 202 participants had adverse reactions, including toxic
sequent visual loss and the need for vitrectomy. In contrast, in epidermal necrolysis, erythema multiforme, and the Stevens–
type 1 diabetic patients there was no major benefit compared to Johnson syndrome. It is unlikely that sorbinil will achieve
the waiting for development of high risk characteristics. prophylactic usage because of the hypersensitivity reactions and
Consequently, PRP should be considered in patients with severe outcome similarities between the group treated with sorbinil
or very severe nonproliferative diabetic retinopathy (NPDR) and the control group in the study.
levels, or with PDR less than high risk, especially if they have The Diabetes Control and Complications Trial (DCCT)
type 2 diabetes. (Table 133.5) was designed to test whether intensive insulin
The DRVS provided guidelines for the most opportune time therapy resulting in improved glycemic control could reduce
to consider vitrectomy surgery for patients with type 1 and the risk of onset or progression of retinopathy for persons with
type 2 diabetes mellitus with vitreous hemorrhage35,36,39 or type 1 diabetes.(Table 133.6)65–68 The DCCT found that regard-
severe PDR in eyes with useful vision.37,38 Early vitrectomy for less of the baseline level of glycemic control, a 10% reduction
eyes with recent severe vitreous hemorrhage and visual acuity of glycosylated hemoglobin level significantly reduced the risk of
less than 5/200 was beneficial, especially for patients with type onset of DR, retarded the progression of DR, and reduced
1 diabetes mellitus. Furthermore, the chances of achieving the need for laser treatment. Intensive therapy reduced the risk
visual acuity of 10/20 or better increased when early vitrectomy of onset of retinopathy by 76%, and resulted in a 63% reduction
was performed in eyes with severe new vessels, again, especially in the risk of progression of retinopathy. In the intensive insulin
for patients with type 1 diabetes mellitus. At the present time, therapy group, the risk of developing severe NPDR or PDR was
the findings of the DRVS should be viewed with the under- reduced by 47%, the risk of developing CSME was reduced by
standing that substantial subsequent developments in vitrectomy 23%, and the risk of requiring laser surgery was reduced by 56%.
instrumentation, techniques, and the application of endo-laser The Epidemiology of Diabetes Intervention and Compli-
have come into common use after the conclusion of this study. cations (EDIC) Study followed the DCCT patient cohort after
The multicenter Sorbinil Retinopathy Trial tested whether a all patients were converted to intensive insulin therapy. Of
daily dose of sorbinil, an aldose reductase inhibitor (ARI), major clinical importance was the finding that despite the fact
reduces the complications of DR. Over a 3 year period, the drug that glycemic control as measured by HbA1c became virtually
had no clinically important effect on the course of DR in adults equivalent between groups, the benefits of early intensive 1777
RETINA AND VITREOUS

TABLE 133.5. Diabetes Control and Complications Trial TABLE 133.6. PDR AT 1-YR Visit by Severity of Individual
Lesion
Major Eligibility Criteria
Lesion Grade PDR in 1 Year (%)
Type 1 Insulin Dependent Diabetes Mellitus Age 13–39 years
HMA Present in 2–5 fields 9
Absence of hypertension, hypercholesterolemia, and severe
diabetic or medical complications Very severe 57
Primary-Prevention Cohort IRMA None 9
IDDM for one to five years Moderate in 2–5 fields 57
No diabetic retinopathy of seven-field stereoscopic fundus VB Lacking 15
photography
Present in 2–5 fields 59
Urinary albumin secretion < 40 mg/24 hrs
Secondary-Intervention Cohort
IDDM for 1 to 15 years
Very-mild-to-moderate nonproliferative diabetic retinopathy
SECTION 10

Urinary albumin secretion < 200 mg/24 h


Major Design Features
Patients randomized to Conventional Treatment or Intensive
Treatment Group
Conventional Treatment Group
insulin injections once or twice a day
daily blood glucose tests
daily self-monitoring of urine or blood glucose
clinical visits every three months
education about diet and exercise
Intensive Treatment Group
insulin pump or three or more insulin injections a day
insulin dosage adjusted according to self-monitoring of blood
glucose, diet, and exercise FIGURE 133.1. Standard photograph No. 2A of the Modified Airlee
House Classification of Diabetic Retinopathy demonstrating a
blood sugar tests four or more times/day moderate degree of hemorrhage or microaneurysms, or both.
diet and exercise plan Courtesy of the Early Treatment Diabetic Retinopathy Study [ETDRS].

initial hospitalization to implement treatment


weekly to monthly clinical visits with frequent telephone
contact
DIAGNOSIS, CLASSIFICATION, AND
MANAGEMENT OF DIABETIC
Major Conclusions
RETINOPATHY
Reduced clinically meaningful retinopathy by 27–76%
Reduced clinically meaningful nephropathy by 34–57% RETINAL LESIONS
Various retinal lesions identify the risk of progression of
retinopathy and visual loss. (Table 133.6)
therapy persisted with continuing benefit of reduced rates of Subtle venous caliber changes and retinal ‘microaneurysms’
retinopathy progression observed in patients who formerly had are generally the first clinically evident signs of DR.
intensive insulin therapy. Similar risk reductions of other Microaneurysms are saccular outpouchings of retinal capillaries
microvascular complications such as renal disease and possibly due to endothelial cell proliferation.(Fig. 133.1)
neuropathy were also observed. Thus, early optimal control of Ruptured microaneurysms, decompensated capillaries, and
blood glucose is critically important for long-term ocular and intraretinal microvascular abnormalities result in ‘intra-retinal
systemic outcome.69,70 hemorrhages.’ The clinical appearance of these hemorrhages
The United Kingdom Prospective Diabetes Study reflects the retinal architecture at the level at which the
(UKPDS)71,72 and studies in Japan73 found similar results in hemorrhage occurs. Hemorrhages in the nerve fiber layer
patients with type 2 diabetes and also identified elevated blood assume a more flame-shaped appearance, coinciding with the
pressure as an independent risk factor for the onset and pro- structure of the nerve fiber layer that runs parallel to the retinal
gression of DR. The UKPDS enrolled 3867 patients with newly surface. Hemorrhages deeper in the retina, at which point the
diagnosed type 2 DM. Intensive blood glucose control with arrangement of nerve fibers is more or less perpendicular to the
either sulphonylureas or insulin resulted in a 17% risk reduc- surface of the retina, assume a dot or blot shape.
tion for progression of DR, a 29% risk reduction in the need for ‘Intra-retinal microvascular abnormalities’ (IRMAs)
laser photocoagulation surgery, a 23% risk reduction for the (Fig. 133.2) represent preexisting vessels with endothelial cell
development of vitreous hemorrhage, and a 16% risk reduction proliferation that become ‘shunts’ through the areas of
1778 in legal blindness. nonperfusion. IRMAs may be seen adjacent to cotton-wool
Diagnosis, Management, and Treatment of Nonproliferative Diabetic Retinopathy

CHAPTER 133
FIGURE 133.2. Standard photograph No. 8A of the Modified Airlie FIGURE 133.4. Standard photograph No. 10A of the Modified Airlie
House Classification of Diabetic Retinopathy demonstrating House Classification of Diabetic Retinopathy demonstrating
intraretinal microvascular abnormalities (IRMAs) (arrows). neovascularization of the optic disc (NVD) (arrow), covering
Courtesy of the ETDRS. approximately one quarter to one third of the disc area.
Courtesy of the ETDRS.

FIGURE 133.3. Standard photograph No. 6B of the Modified Airlie


House Classification of Diabetic Retinopathy demonstrating venous FIGURE 133.5. Standard photograph No. 7 of the Modified Airlie
beading. House Classification of Diabetic Retinopathy demonstrating
Courtesy of the ETDRS. neovascularization elsewhere (NVE) in the retina greater than one half
disk diameter with a fresh hemorrhage present.
Courtesy of the ETDRS.

spots. Multiple IRMAs mark a severe stage of nonproliferative


retinopathy with a high risk of development of frank neovas-
cularization. Retinal neovascularization commonly occurs in Patients with high-risk PDR require immediate PRP (see
areas of previously existing IRMA. Chapter 135). High-risk PDR is characterized by one or more of
‘Venous caliber abnormalities’ (Fig. 133.3) are indicators of the following lesions:
retinal hypoxia. These abnormalities can be venous dilatation, • NVD that is ~one-quarter to one-third disc area or more
venous beading, or venous loop formation. There are often large in size (i.e., ≥ 1/4 – 1/3 disk area) (Fig. 133.4),
areas of nonperfusion adjacent to these veins. PRP may cause • NVD < one-quarter disc area in size if fresh vitreous or
these abnormal veins to become less dilated and more regular. preretinal hemorrhage is present, and
‘Proliferative retinopathy’ (Fig. 133.4) is marked by devel- • NVE ≥ 1/2 disc area in size if fresh vitreous or preretinal
opment of retinal neovascularization. The rate of growth of hemorrhage is present (Fig. 133.5).
these new vessels is variable. They most commonly appear at or
near the optic disc (neovascularization of the disk NVD) or Thus, the evaluation of the diabetic retina for PDR requires
elsewhere in the retina (neovascularization elsewhere NVE) and attention to the presence, location and extent of new vessels
proliferate on the posterior vitreous face. They may also arise in and the presence or absence of preretinal or vitreous
the perifoveal area. hemorrhages.4 1779
RETINA AND VITREOUS

NONPROLIFERATIVE DIABETIC RETINOPATHY TABLE 133.7. Levels of Retinopathy


(NPDR) AND EARLY PDR Nonproliferative Diabetic Retinopathy (NPDR)
As described earlier, it is critically important to consider PRP
A. Mild NPDR
as retinopathy approaches or reaches the high-risk stage of
PDR. An eye is considered to be approaching the high-risk stage At least one microaneurysm
when there are retinal signs of severe or very severe NPDR or Definition not met for B, C, D, E, F
the presence of PDR without high risk characteristics. The
baseline severity of retinopathy is closely correlated with the B. Moderate NPDR
risk of NPDR progression to early PDR and to high-risk PDR H/Ma ≥ standard photograph No. 2A
(Tables 133.6 to 133.8), making accurate assessment of NPDR
Soft exudates, VB, and IRMA definitely present
severity not only important for identifying timing of laser
surgery, but also for determining future follow-up schedules to Definition not met for C, D, E, F
minimize risk between ophthalmic examinations. C. Severe NPDR

Nonproliferative Diabetic Retinopathy Severity H/Ma ≥ standard photograph No. 2A (Fig. 133.1) in all
4 quadrants
DR is broadly classified as NPDR and PDR. DME can occur
with either NPDR or PDR and is discussed separately. Accurate VB in 2 or more quadrants (Fig. 133.3)
SECTION 10

diagnosis of a patient’s ‘DR severity level’ is critical because IRMA > standard photograph No. 8A in at least 1 quadrant
there is a varying risk of progression to PDR and high-risk PDR (Fig. 133.2)
depending on the specific NPDR severity level.(Fig. 133.6);
(Table 133.8) D. Very Severe NPDR
‘Mild NPDR’ is marked by at least one retinal micro- Any two or more of C.
aneurysm, but hemorrhages and microaneurysms are less than Definition not met for E, F
those in ETDRS standard photograph No. 2A (Fig. 133.1 and
Table 133.7). No other retinal lesion or abnormality associated Proliferative Diabetic Retinopathy (PDR)
with diabetes is present. Those with mild NPDR have a 5% risk (Composed of:(1) NVD or NVE, (2) preretinal or vitreous
of progression to PDR within 1 year and a 15% risk of hemorrhage, (3) fibrous tissue proliferation)
progression to high-risk PDR within 5 years (Table 133.8).
E. Early PDR
‘Moderate NPDR’ (Table 133.7) is characterized by
hemorrhages or microaneurysms, or both (H/Ma), greater than New vessels
or equal to those pictured in ETDRS standard photograph No. Definition not met for F
2A. Soft exudates, venous beading, and IRMAs are definitely
present in a mild degree. The risk of progression to PDR within F. High-risk PDR
1 year is 12–27%, and the risk of progression to high-risk PDR NVD ( 1/3 – 1/2 disc area (Fig. 133.4) or
within 5 years is 33 percent (Table 133.8).
NVD and vitreous or preretinal or vitreous hemorrhage
Patients with mild or moderate NPDR generally are not (Fig. 133.5) or
candidates for PRP and can be followed safely at 6–12 month
intervals as determined by the examiner. The presence of NVE ≥ 1/2 disc area and vitreous or preretinal hemorrhage
macular edema, even with mild or moderate degrees of NPDR, Clinically Significant Macular Edema (CSME)
requires follow-up in a shorter period, and if CSME is present,
1. Thickening of the retina at or within 500 mm from the center of
focal laser treatment should be considered (Table 133.8). the macula or
Coincident medical problems or pregnancy will generally
necessitate a more frequent period of reevaluation. 2. Hard exudates with thickening of the adjacent retina located at
or within 500 mm from the center of the macula or
‘Severe NPDR’, based on the severity of H/Ma, IRMAs, and
venous beading, is characterized by any one of the following 3. A zone of retinal thickening, 1 disc area or larger in size located
lesions (see Table 133.7): at or within 1 disc diameter from the center of the macula.
• H/Ma ≥ standard photograph No. 2A (Fig. 133.1) in four
quadrants, or
• Venous beading (Fig. 133.3) in two or more quadrants, or very severe NPDR may be candidates for PRP, and, macular
• IRMAs ≥ standard photograph No. 8A (Fig. 133.2) in at edema if present, may require treatment. Very close follow-up
least one quadrant. evaluation at 2–3 month intervals is important (Table 133.8).
For type 2 diabetes, early PRP should be considered for patients
These criteria are often referred to as the ‘4–2–1- rule’ for with severe or very severe NPDR.62
determination of severe NPDR. Any two or more of the above
findings reflects very severe NPDR. Eyes with severe NPDR Early Proliferative Diabetic Retinopathy
have a 52% risk of developing PDR within 1 year and a Diabetic retinopathy marked by NVD or NVE on the retina
60% risk of developing high-risk PDR within 5 years. These or by fibrous tissue proliferation is designated PDR. Early PDR
patients require follow-up evaluation at 2–4 months intervals. does not meet the definition of high-risk PDR (Table 133.7).
Treatment of CSME is strongly indicated because of the risk of Eyes with early PDR (< high-risk PDR) have a 75% risk of
the development of PDR and high-risk PDR. In addition, some developing high-risk PDR within a 5-year period. These eyes
eyes with macular edema, even if not clinically significant, may may prompt PRP; and macular edema, sometimes even if not
require focal treatment in preparation for impending PRP clinically significant, may benefit from focal treatment before
(Table 133.8). scatter treatment is initiated (Table 133.8).
Eyes with very severe NPDR (Table 133.7) have two or more Patients with severe and very severe NPDR and early PDR
lesions of severe NPDR but no frank neovascularization. There (<high-risk PDR) should be considered for early PRP. This is
1780 is a 75% risk of developing PDR within 1 year. Patients with particularly true if there are new vessels in the presence of
Diagnosis, Management, and Treatment of Nonproliferative Diabetic Retinopathy

TABLE 133.8. General Management Recommendations

Natural Course Evaluation Treatment Strategies


Rate of Progression to:
Level of Retinopathy PDR 1 Yr HRC* 5 Yr Color Fundus FA† PRP‡ Focal Follow-Up
Photograph (mos)

Mild NPDR 5% 15%


No macular edema No No No No 12
Macular edema Yes Occasionally No No 4–6
CSME Yes Yes No Yes 2–4
Moderate NPDR 12–27% 33%
No macular edema Yes No No No 6–8
Macular edema (not CSME) Yes Occasionally No No 4–6

CHAPTER 133
CSME Yes Yes No Yes 2–4
Severe NPDR 52% 60%
No macular edema Yes No Rarely No 3–4
Macular edema (not CSME) Yes Occasionally Occasionally Occasionally 2–3
after focal
CSME Yes Yes Occasionally Yes 2–3
after focal
Very severe NPDR 75% 75%
No macular edema Yes No Occasionally No 2–3
Macular edema (not CSME) Yes Occasionally Occasionally Occasionally 2–3
after focal
CSME Yes Yes Occasionally Yes 2–3
after focal
Nonhighrisk PDR 75%
No macular edema Yes No Occasionally No 2–3
Macular edema (not CSME) Yes Occasionally Occasionally Occasionally 2–3
after focal
CSME Yes Yes Occasionally Yes 2–3
after focal
Highrisk PDR
No macular edema Yes No Yes No 2–3
Macular edema Yes Yes Yes Usually 1–2
CSME Yes Yes Yes Yes 1–2
* HRC = highrisk characteristic proliferative retinopathy
† FA = fluorescein angiography.
‡ PRP = pan-retinal photocoagulation.

severe or very severe NPDR, elevated new vessels, or NVD. In diabetic retinopathy: no apparent retinopathy (no abnormalities),
the presence of macular edema, patients with severe NPDR or mild NPDR (microaneurysms only), moderate NPDR (more
worse should be considered for prompt focal treatment of than microaneurysms only but less than severe NPDR), severe
macular edema, possibly whether the macular edema is NPDR (any of the following: >20 intraretinal hemorrhages in
clinically significant or not, in preparation for the impending each of four quadrants, definite VB in two or more quadrants,
need of PRP which can exacerbate any DME present at the time prominent IRMA in one or more quadrant and no PDR), and
of treatment (Table 133.8). PDR (one or more of retinal neovascularization, vitreous
hemorrhage, or preretinal hemorrhage). Table 133.9 compares
INTERNATIONAL CLASSIFICATION OF levels of DR in the International Classification of DR to ETDRS
DIABETIC RETINOPATHY levels of DR.
The International Classification identified two broad levels of
In an effort to simplify classification and standardize com- DME: macular edema apparently absent (no apparent retinal
munication, the American Academy of Ophthalmology thickening or hard exudates (HE) in posterior pole) and macular
initiated a project to establish a consensus International edema apparently present (some apparent retinal thickening
Classification of DR and DME.74,75 This International or HE in posterior pole); if present, macular edema was
Classification of DR and DME described five clinical levels of subclassified as ‘mild DME’ (some retinal thickening or HE in 1781
RETINA AND VITREOUS

ROLE OF FLUORESCEIN ANGIOGRAPHY IN


THE MANAGEMENT OF DIABETIC
RETINOPATHY
Fluorescein angiography of the macula in the presence of
CSME is fundamental for the detection of treatable lesions, as
described below. However, its use to identify lesions such as
NVE, NVD, or retinal thickening is generally not necessary as
these lesions are usually detectable by clinical exam.
Angiographic risk factors for progression of NPDR to PDR have
been identified.26,28 Analysis of data from the untreated
(deferred) eyes in the ETDRS indicates that the following
lesions are independently related to outcome: (1) fluorescein
leakage, (2) capillary loss on fluorescein angiography,
(3) capillary dilatation on fluorescein angiography, and (4) the
following color fundus photographic risk factors: IRMAs,
FIGURE 133.6. Life table cumulative event rates of high-risk venous beading, and H/Ma. Hard and soft exudates have an
proliferative retinopathy by level of retinopathy severity scale at inverse relationship to progression. It is widely accepted that
SECTION 10

baseline in eyes assigned to deferral of photocoagulation in the capillary loss as documented on fluorescein angiography is a
ETDRS. Level < 35, mild nonproliferative diabetic retinopathy (NPDR); risk factor for progression of NPDR to PDR.26,28,76–78 However,
level 43, moderate NPDR; level 47, moderate to severe NPDR; level capillary dilatation on fluorescein angiography, fluorescein leak-
53A-D, severe NPDR; level 53E, very severe NPDR; level 61, early age, and capillary loss on fluorescein angiography, and the
proliferative diabetic retinopathy (PDR); level 65, PDR < high-risk ETDRS color fundus photographic retinopathy severity levels
PDR.27
are all closely correlated. Although the fluorescein angiography
Courtesy of the ETDRS.
abnormalities provide additional prognostic information, the
color fundus photographic grading of retinopathy severity levels
posterior pole but distant from center of the macula), ‘moderate give the same prognostic results.27,28 The increased power to
DME’ (retinal thickening or HE approaching the center of predict progression from NPDR to PDR by fluorescein
the macula but not involving the center), or ‘severe DME’ angiography is generally considered “not of significant clinical
(retinal thickening or HE involving the center of the macula). importance to warrant routine fluorescein angiography.”28
Table 133.10 compares levels of DME in the International Periodic follow-up retinal examinations, however, are
Classification to ETDRS levels of DME. necessary. The appropriate interval can be determined by skillful
This International Classification of DR and DME reduces grading of seven standard field stereo color fundus photographs
the number of levels of DR, simplifies descriptions of the or by retinal examination by an ophthalmologist experienced in
categories, and describes the levels without relying on reference the management of diabetic eye disease. Since the severity of
to the standard photographs of the Airlie House Classification diabetic retinopathy is predictive of progression and establishes
of DR. Thus, the International Classification simplifies the frequency of follow-up, and since initiation of PRP should be
clinical levels of diabetic retinopathy, easing the communication considered as diabetic retinopathy approaches the high-risk
between clinicians. However, since the International Classifica- stage, routine life-long “periodic follow-up of all patients with
tion is less detailed than the ETDRS retinopathy severity scale, diabetic retinopathy continues to be of fundamental clinical
the International Classification of DR and DME is not a importance.”28
replacement for the ETDRS scale in large clinical trials or Other imaging modalities also add vital information to the
studies where precise retinopathy classification is required. evaluation of DR. The introduction of optical coherence

TABLE 133.9. Comparison of the International Clinical DR Scale and the Early
Treatment Diabetic Retinopathy Study (ETDRS) Scale of Diabetic Retinopathy

International Classification Level of DR ETDRS Level of DR

No apparent retinopathy Level 10: DR absent


Mild NPDR Level 20; very mild NPDR
Moderate NPDR Levels 35, 43, 47; moderate NPDR
Severe NPDR Levels 53A-E; severe to very severe NPDR
PDR Levels 61,65,71,75,81,85; PDR, high-risk
PDR, very severe or advanced PDR
DR =diabetic retinopathy; NPDR = Nonproliferative diabetic retinopathy; PDR = Proliferative diabetic
retinopathy
Derived from: (Wilkinson CP, Ferris FL III, Klein RE, et al: Proposed international clinical diabetic retinopathy
and diabetic macular edema disease severity scales. Ophthalmology 2003; 110:1677–1682. Chew EY: A
simplified diabetic retinopathy scale. Ophthalmology 2003; 110:1675–1676. Fundus Photographic Risk
Factors for Progression of Diabetic Retinopathy. ETDRS Report Number 12. Early Treatment Diabetic
Retinopathy Study Research Group. Ophthalmology 1991; 98(5 Suppl):823–833. Grading Diabetic
Retinopathy From Stereoscopic Color Fundus Photographs—an Extension of the Modified Airlie House
Classification. ETDRS Report Number 10. Early Treatment Diabetic Retinopathy Study Research Group.
Ophthalmology 1991; 98(5 Suppl):786–806.)
1782
Diagnosis, Management, and Treatment of Nonproliferative Diabetic Retinopathy

TABLE 133.10. Comparison of International Clinical DME Scale and ETDRS Scale

Disease Severity Level Findings DME vs. ETDRS scale

DME apparently absent No apparent retinal thickening or hard No DME


exudates (HE) in posterior pole
DME apparently present Some apparent retinal thickening or HE Mild DME: some retinal thickening or HE in posterior pole but
in posterior pole distant from center of the macula (ETDRS: DME but not CSME)
Moderate DME: retinal thickening or HE approaching the center
but not involving the center (ETDRS: CSME)
Severe DME: retinal thickening or HE involving the center of the
macula (ETDRS: CSME, center involved)
DME=diabetic macular edema; HE=hard exudates; CSME=clinically significant macular edema.

tomography (OCT) has permitted evaluation of macular thick-


ening and vitreo-retinal interface abnormalities in a quan-

CHAPTER 133
titative, highly sensitive and reproducible manner.79 OCT is
valuable in confirming or elucidating subtle macular pathology
such as persistent edema, preretinal fibrosis, and vitreo-macular
traction that might otherwise be difficult or impossible to
appreciate on clinical exam. However, OCT does not provide
substantive information on the extent or location of the retinal
lesions characterizing NPDR as does photography, nor does it
identify the source or activity of retinal leakage as can
angiography. Although visual acuity is generally correlated with
central macular thickness, the variability is large, making
current models of OCT inadequate for use as a surrogate for
visual acuity in clinical trials.80 New spectral domain models
of OCT have been approved by the FDA and promise entirely
new methods and sensitivity of evaluating retinal pathology
that will require study to fully elucidate their role in the
clinical management and research investigation of diabetic
retinopathy. FIGURE 133.7. Schematic representation of clinically significant
macular edema (CSME), with thickening of the macula less than
500 mm from the center of the macula.
DIABETIC MACULAR EDEMA Courtesy of Robert Murphy, M.D.
Diabetes can affect the macula and macular function through a
variety of mechanisms including:
1. Macular edema; i.e., a collection of intra-retinal fluid in 3. An area or areas of retinal thickening at least one disk area
the macula, with or without lipid exudates and with or in size, at least part of which is within 1 disk diameter of
without cystoid changes; the center of the macula (Figs 133.9 and 133.10).
2. Nonperfusion of perifoveal capillaries, with or without
intra-retinal fluid; There are particular retinal lesions identified on fluorescein
3. Traction in the macula by fibrous or glial tissue causing angiography that are amenable to treatment. The ‘treatable
dragging of the retinal tissue, surface wrinkling, or lesions’ associated with macular edema include:
detachment of the macula; 1. Focal leaks >500 mm from the center of the macula
4. Intra-retinal or preretinal hemorrhage in the macula; thought to be causing retinal thickening or hard exudates
5. Lamellar or full-thickness retinal hole formation; (Fig. 133.11),
6. Any combination of the preceding and other 2. Focal leaks 300 to 500 mm from the center of the macula
mechanisms. thought to be causing retinal thickening or hard exudates if
the treating ophthalmologist does not believe that
Diabetic macular edema may be present at any level of treatment is likely to destroy the remaining perifoveal
retinopathy (Table 133.8). For clinical purposes, ‘macular capillary network (Fig. 133.11),
edema’ is defined as retinal thickening within two disk diam- 3. Areas of diffuse leakage that have not been treated
eters of the center of the macula (not fluorescein leakage previously (Fig. 133.12), or
without thickening). Retinal thickening or hard exudates with 4. Avascular zones, other than the normal foveal avascular
adjacent retinal thickening that threatens or involves the center zone, not previously treated (Fig. 133.12b).
of the macula is considered to be clinically significant.
CSME as defined by the ETDRS includes any one of these Focal laser surgery for CSME consists of either direct laser treat-
lesions (see Table 133.7): ment, grid laser treatment, or a combination of direct laser to
1. Retinal thickening at or within 500 mm of the center of the focal leaks and grid laser treatment to diffuse leakage or thick-
macula (Fig. 133.7); or ened avascular macula. These treatment methods are described
2. Hard exudates at or within 500 mm s of the center of in detail elsewhere.17,19 Table 133.8 summarizes the manage-
the macula, if there is thickening of the adjacent retina ment recommendations for CSME at the various retinopathy
(Fig. 133.8); or levels. 1783
RETINA AND VITREOUS

a b
SECTION 10

FIGURE 133.8. (a) Schematic representation of CSME with hard exudates at or within 500 mm of the center of the macula, with thickening of the
retina adjacent to the exudates. (b) Clinical appearance of hard exudates less than 500 mm from the center of the macula. There is thickening of
the adjacent retina, which is not appreciated without stereoscopic observation.
(a) Courtesy of Robert Murphy, M.D. (b) Courtesy of the ETDRS.

FIGURE 133.9. Schematic representation of area of thickening, 1 disk


diameter in size, part of which is within 1 disk diameter of the center FIGURE 133.10. Clinical photograph showing macular edema greater
of the macula. than 500 mm from the center of the macula (the edema is not
Courtesy of Robert Murphy, M.D. appreciated without stereoscopic evaluation).
Courtesy of the ETDRS.

LASER PHOTOCOAGULATION FOR DIABETIC


is unlikely to increase the risk of SVL provided that the
MACULAR EDEMA AND NONPROLIFERATIVE retinopathy is not progressing rapidly and careful follow-up can
DIABETIC RETINOPATHY be maintained. In contrast, in eyes with CSME and high-risk
The 5-year risk of MVL from macular edema in the ETDRS PDR, delaying PRP while focal treatment is completed usually
without focal laser treatment was 30%. Focal laser surgery for is not advisable. In these cases, the DME treatment will
CSME reduced this risk to 15%,16 amounting to a 50% generally be completed and the PRP begun during the same
reduction in the risk of moderate visual loss. Focal treatment initial laser treatment session.
also increased the chance of improvement in visual acuity of Focal treatment was not attended by adverse effects on
one line or more, but in general, vision remains approximately central visual field or color vision when compared with eyes
constant. Conversely, PRP was not effective in treating diabetic assigned to deferral of focal treatment.17Any harmful effects
macular edema and in some cases may have had a deleterious of early photocoagulation reflected by constriction of the
effect on the progression of macular edema. peripheral visual fields were primarily attributable to PRP. Since
Eyes with CSME and retinopathy that is approaching high- the principal benefit of laser for CSME is to prevent a further
risk PDR are best treated first with focal photocoagulation for decrease in visual acuity, laser surgery should be considered in
the macular edema 6–8 weeks before initiating PRP. Eyes with all eyes with CSME, especially if the center of the macula is
mild or moderate NPDR and CSME respond best to prompt threatened or involved, even if normal visual acuity is present.
focal photocoagulation, with scatter treatment delayed unless The DRS had demonstrated in 1976 that PRP was effective
severe or very severe NPDR or high-risk PDR occurs. Delaying in reducing the risk of severe visual loss from high-risk PDR.
1784 PRP while focal treatment is being completed in these patients Since, the DRS did not provide a clear choice between prompt
Diagnosis, Management, and Treatment of Nonproliferative Diabetic Retinopathy

a b c

CHAPTER 133
d e

FIGURE 133.11. (a) There is a small area of retinal thickening just above the center of the macula (poorly appreciated without stereopsis), which
is detectable monocularly because of blurring of the choroidal pattern. Several microaneurysms are visible within the thickened area. There are
hard exudates around the edges of the edematous patch, some of which extends almost to the center of the macula. Thickening extends to
within 500 mm of the center of the macula (clinically significant macular edema). Visual acuity is 20/15. (b) In the 17- to 18-sec phase of the
fluorescein angiogram, microaneurysms and slightly dilated capillaries are visible in the area of thickening. (c) The 7-min phase of the angiogram
shows leakage into the retina from the two groups of microaneurysms noted in (b). (d) Treatment has been applied to most of the
microaneurysms. (e) Four months later, the appearance of the retina is satisfactory: The center of the macula is flat and most of the thickening
noted before treatment has disappeared. Visual acuity remains at 20/15.
(a–e) Courtesy of the ETDRS.

treatment and deferral of treatment unless there was progres- generally uses a 50 mm spot size (50–100 mm in the ETDRS) to
sion to high-risk PDR, one question of concern for the photocoagulate discrete lesions (such as microaneurysms that
subsequent ETDRS was whether earlier PRP, before the fill or leak during fluorescein angiography) or to place a
development of high-risk PDR, justified the side effects and grid over diffusely thickened areas and nonperfusion. Treatment
attendant risks of laser surgery. (Table 133.11) is placed from 500–3000 mm from the center of the macula
In the ETDRS, early treatment, compared with deferral of as discussed earlier. Response to treatment is generally asses-
photocoagulation until high-risk PDR developed,22 was sed after 3–4 months and retreatment or a new therapeutic
associated with a small reduction in the incidence of SVL, but approach considered at that time for persistent or worsening
5 year rates of SVL were low for both the early treatment group CSME. Complications and side effects of laser photocoa-
and the group assigned to deferral of treatment (2.6% and gulation are summarized in Table 133.11.
3.7%, respectively). However, for patients with type 2 diabetes, In summary, focal/grid laser photocoagulation reduces
earlier laser photocoagulation, even for eyes with severe or very moderate visual loss from CSME by 50%. PRP significantly
severe NPDR, does reduce the risk of severe vision loss.62 reduces the risk of SVL from PDR but can also exacerbate DME.
Provided careful follow-up can be maintained, PRP is not Thus, treatment of DME in patients approaching high risk
recommended for eyes with mild or moderate NPDR since the retinopathy should be considered before PRP is urgently
benefits are small and the risks equivalent as compared with required (i.e., with the development of high risk PDR). Early
treating more severe retinopathy.24 When retinopathy is more PRP reduces the risk of severe visual loss, but the rates of SVL
severe (i.e., severe or very severe NPDR and early PDR), PRP are low. Nevertheless, there is a clear benefit for early treatment
should be considered especially in patients with type 2 diabetes in patients with type 2 diabetes. Consequently, it is recom-
24
as the benefits of early photocoagulation are greater. When mended that PRP not be used for mild to moderate NPDR. For
both eyes are approaching the high-risk stage, initiating PRP severe NPDR and early PDR, PRP is most appropriate when
early in at least one eye seems particularly appropriate as close follow-up is unlikely, the disease process is progressing
optimal timing of photocoagulation may be difficult if both eyes rapidly, or for patients with type 2 diabetes.
need photocoagulation simultaneously. Also, prompt PRP
should be considered for eyes with neovascularization in the
anterior chamber angle, whether or not high-risk proliferative NOVEL AND EVOLVING THERAPIES FOR
retinopathy is present. DIABETIC RETINOPATHY AND DIABETIC
Table 133.8 presents the treatment program for DR, and the MACULAR EDEMA
evaluation and treatment of PDR is discussed extensively in The rapid advancement of scientific research over the past
the PDR chapter. Focal photocoagulation for macular edema decade has tremendously expanded our understanding of the 1785
RETINA AND VITREOUS

a b c
SECTION 10

d e f

FIGURE 133.12. (a) Clinical photograph showing retinal thickening temporal to the center of the
macula extending just to the center. Visual acuity is 20/40+3. (b) Early phase of the angiogram
shows capillary loss adjacent to the foveal avascular zone, capillary dilatation, and scattered
microaneurysms. (c) The 7-min phase of the angiogram shows extensive small cystoid spaces
temporal to the center of the macula and above and below it. The center appears uninvolved.
(d) The microaneurysms have been treated focally. In addition, laser burns have been applied in
a grid pattern to the areas of diffuse leakage. (e) The temporal extent of the grid laser treatment.
(f) Four months later, hemorrhages and hard exudates have decreased, and the retinal thickening
can no longer be detected. Visual acuity is 20/25. (g) The 7-min phase of the angiogram showing
disappearance of most of the cystoid spaces visible in (c).
(a–g) Courtesy of the ETDRS.
g

molecular and cellular mechanisms underlying the develop- was also assessed.81 The study showed occasional diurnal
ment of NPDR, PDR and DME. With this added knowledge variation in retinal thickening in patients with CSME but not
has come new insight into targets for therapeutic intervention. to an extent that would generally affect clinical trial research.
Currently new agents that may reduce complications of diabetes However, it was found that although retinal thickness and
in the eye are in various stages of evaluation. Among those with visual acuity are modestly correlated, there is substantial
significant clinical trial and use are intra-vitreal steroids, intra- variability. Substantial proportions of patients may have
vitreal antiangiogenic agents and oral PKC-b inhibitors as paradoxical responses (i.e., improved vision despite retinal
agents of particular interest. These agents hold promise not thickening, or declining vision with resolution of excess retinal
only for additional, possibly more efficacious therapies, but also thickening).
for fewer associated side effects since they do not share the The second study compared standard laser (modified ETDRS
inherently retinal destructive nature of laser treatment. protocol) with a mild macular grid (MMG) treatment for
The need to evaluate numerous new therapies in a treating CSME.82 MMG entailed light burns spread across the
scientifically rigorous and timely manner has prompted macula in areas of both thickened and unthickened retina.
development of clinical trial networks. The National Eye Although both MMG and modified ETDRS treatment were
Institute funded Diabetic Retinopathy Clinical Research found effective in reducing retinal thickening and improving
Network (DRCR.net), initiated in 2002, is a collaborative effort vision in patients with CSME, the magnitude of effect of MMF
with the goal of facilitating multicentric clinical research to was not as robust as modified ETDRS and side effects were
study the effectiveness of new treatments of diabetic similar. Thus, the MMG approach is not being evaluated
retinopathy and diabetic macular edema. The DRCR.net further by large scale studies.
currently includes over 160 participating sites in more than 40 There are three DRCR.net studies currently in the follow-up
states, representing in excess of 500 physicians and 1000 other phase, all involving the treatment of CSME. The first study is
personnel. comparing peribulbar triamcinolone acetonide (Kenalog) with
To date, two DRCR.net studies have been reported. The first focal laser (modified ETDRS) photocoagulation. Patients were
study was an observational study evaluating the relationship randomized to receive either laser, steroid (by anterior subtenon
between OCT measured retinal thickness and visual acuity.80 or posterior subtenon injection), or both. Completion of the
1786 Diurnal variation in retinal thickening as measured by OCT initial primary endpoint follow-up phase presented at a national
Diagnosis, Management, and Treatment of Nonproliferative Diabetic Retinopathy

endophthalmitis. (see also PDR chapter for further details on


TABLE 133.11. Complications and Side Effects of Scatter
intra-vitreal steroid therapies).
(Panretinal) Laser Photocoagulation
Another set of DRCR.net studies is evaluating the anti-
Side Effects angiogenic agents Bevacizumab (Avastin, Genentech, Inc.)84,85
Field constriction and nyctalopia: Depends upon extent and
and Ranibizumab (Lucentis, Genentech, Inc.)86 for the treat-
intensity of scatter laser burns ments of CSME. These agents block the potent angiogenic and
permeability factor called vascular endothelial growth factor
Serous or choroidal detachment (VEGF). VEGF is known to be involved in mediating PDR
Pain and DME and is described in detail in the chapter on PDR.
Bevacizumab and Ranibizumab are humanized antibodies or
Peripheral retina-more painful
antibody fragments, respectively, that bind all isoforms of VEGF
Use retrobulbar anesthesia as needed with high affinity. Ranibizumab is FDA approved for the treat-
Reassurance ment of neovascular age-related macular degeneration (AMD)
while bevacizumab is FDA approved for the systemic treatment
Hypoglycemia of certain cancers.
Anxiety, pain, shock, seizure Pegaptanib (Macugen, Eyetech Pharmaceuticals) is an
Complications
aptamer with high affinity for only the VEGF120 isoform and is
FDA approved for the treatment of neovascular AMD. Recently,

CHAPTER 133
Foveal burn a pharmaceutically sponsored Phase II trial of pegaptanib intra-
Identify landmarks prior to treatment vitreally injected every 6 weeks was completed involving 172
patients. Median visual acuity, visual acuity improvement of
Macular edema 10 or more letters, median retinal thickness and need for
May result in permanent decrease in visual acuity photocoagulation were all better after 36 weeks of treatment
Foveal traction
with 0.3 mg pegaptanib as compared with sham injections.87
Phase III trials with pegaptanib for diabetic macular edema are
Do not treat over blood currently underway.
Avoid puncture of Bruch’s membrane The DRCR.net is currently recruiting or will soon initiate
several additional clinical trials. These trials include evaluating
Acute angle-closure glaucoma
if subgroups of DME will benefit from vitrectomy, determining
Possible but rare the incidence of macular edema (as measured by OCT) follow-
Anterior segment
ing various modalities of PRP, identifying risks of subsequent
DME in patients with subclinical DME, evaluating the effect of
Posterior synechiae adjunctive therapies (steroids or VEGF inhibitors) when used
Cornea and lens burns with PRP for the prevention and/or treatment of macular
edema, and the effect of adjunctive therapies at the time of cat-
Internal ophthalmoplegia
aract surgery to evaluate their ability to prevent and/or improve
Less frequent with multiple sessions and light to moderately diabetic macular edema.
intense burns At this time, the available clinical evidence suggests that
Retrobulbar hemorrhage following retrobulbar injection anti-VEGF and possibly other antiangiogenic agents will have
a substantial effect on ameliorating neovascular disease.
Continue with treatment Although it is likely that these will also have benefit for the
Watch central retinal artery treatment of CSME, initial data suggests that the effect may be
partial and require repetitive administration to maintain the
Retrobulbar anesthesia required infrequently
effects. Results of ongoing clinical trials will be necessary before
Loss of follow-up may result from enough is known about the efficacy and side effects of these new
Pain agents to assess their impact on the clinical care of diabetic eye
disease. The role of these agents for preventing progression of
Retrobulbar hemorrhage NPDR is theoretical at present and awaits trials specifically
Intercurrent illness designed to address this issue.
Lack of caring explanation
PKC-b activation is induced by hyperglycemia through
several mechanisms and in part mediates the development of
Lack of persistent rescheduling vascular dysfunction, vascular permeability, VEGF expression
and VEGF signal transduction. Thus, inhibition of PKC-b might
be expected to ameliorate diabetes-induced vascular compli-
scientific meeting demonstrated that the peribulbar application cations by several mechanisms. The synthesis of a PKC-b
of Kenalog did not show any additional benefit to laser isoform-selective inhibitor (ruboxistaurin, Eli Lilly) has pro-
treatment alone.83 vided diverse data to substantiate this hypothesis.88 Orally
Another study is comparing 1 or 4 mg intra-vitreal injection ingested ruboxistaurin ameliorates diabetes-induced abnor-
of a preservative free triamcinolone acetonide with laser for malities of retinal blood flow, glomerular filtration rate and
treatment of CSME. This study is currently ongoing. Early albumin excretion rate in animals.89 These data supported the
studies of intravitreal administration of corticosteroids have evaluation of orally administered ruboxistaurin in clinical trials
shown a transient reduction in excess retinal thickening in as a potential noninvasive and nondestructive inhibitor of the
patients with CSME. Vision can improve, although this progression of diabetic retinopathy and diabetic macular
improvement does not always occur, and the effect generally edema.90
wears off necessitating repetitive injections. Intra-vitreal Data from two 3-year Phase III trials of ruboxistaurin in
injection of steroid is associated with development of cataract patients with moderate to severe NPDR have been reported.91,92
and the potentially severe side effects of glaucoma and Results were very similar in both trials. In the larger PKC-DRS2 1787
RETINA AND VITREOUS

TABLE 133.12. Eye Examination Schedule

Type of Diabetes Mellitus Recommendation Time of Routine Minimal


First Examination Follow-up*

Type 1 Diabetes Mellitus 5 yr after onset or during puberty Yearly


Type 2 Diabetes Mellitus At time of diagnosis Yearly
During pregnancy Prior to pregnancy for counseling 1–2 months post partum
Early in first trimester
Each trimester or more frequently
as indicated
*Abnormal findings dictate more frequent follow-up examinations.

study, 685 patients with moderate to severe NPDR and no PRP nation of retinopathy severity, optimization of systemic factors,
SECTION 10

in at least one eye were evaluated after receiving orally routine careful follow-up and timely laser photocoagulation,
administered ruboxistaurin (32 mg/day) over a period of 36 vitrectomy surgery and/or novel therapies. Proper care results
months.92 Moderate visual loss sustained for at least 6 months in substantial reduction of personal suffering and substantial
(SMVL) occurred in 5.5% of ruboxistaurin-treated patients and cost savings.107
9.1% of placebo-treated patients (40% risk reduction, p=0.03). For patients who do not have access to diabetes eye care,
In individual eyes, ruboxistaurin reduced SMVL by 45% novel methods of delivery of such care have been evolving.
(p=0.01). Mean visual acuity was better in the ruboxistaurin- Ocular telemedicine is one approach that has the potential to
treated patients from 12 months onward. In ruboxistaurin- extend high quality diabetes eye care to patients who face socio-
treated patients, visual improvement of 15 or more letters was economic, geographic, or other challenges to care. While some
twice as frequent (4.9% vs 2.4%). Ruboxistaurin treatment also programs have demonstrated a high degree of reliability in
reduced progression of macular edema to within 100 mm from identifying level of diabetic retinopathy compared to clinical
the center of the macula and the need for initial laser treatment examination and standard retinal photography, it is important
for macular edema was 26% less frequent in eyes of ruboxis- for both patients and health care providers to recognize
taurin-treated patients. Ruboxistaurin therapy was very well strengths and limitations of ocular telemedicine programs. To
tolerated and has had an excellent safety profile to date.93 No assist in guiding expectations, the American Telemedicine
effect was observed on progression of NPDR severity. The com- Association has prepared ‘Practice Recommendations for
pound has received an ‘Approvable’ rating by the FDA under the Ocular Telemedicine for Diabetic Retinopathy’.
trade name Arxxant (Eli Lilly). Requirements for additional Strict guidelines have been established for the ocular care of
studies for full regulatory approval are currently under discussion. people with diabetes (Tables 133.8 and 133.12). All diabetic
patients should be informed of the possibility of the devel-
opment of retinopathy with or without symptoms and the
PRECLINICAL STUDIES associated threat of visual loss. The natural course and treat-
A wide array of other interventions are in various stages of ment of DR should be discussed and the importance of routine
preclinical evaluation. Such studies hold promise for eventually examination stressed. The patient must understand that the
even more effective, more specific and safer therapies than are risks of diabetes complications in the eye and elsewhere in the
currently available. However, several years of further inves- body may be reduced by diligent personalized health care and
tigation will be required before any potential clinical impact is routine follow-up examinations, and that early efforts despite
known. Ongoing research is addressing mechanisms contri- lack of symptoms can yield long term benefits that are lost if
buting to altered retinal blood flow and retinal vascular care is initiated late. Thus, in many ways, the care of the
complications in diabetes.94,95 Retinal blood flow is decreased in patient with no or early NPDR is of paramount importance.
patients who have had diabetes less than 5 years and may be Patients should be informed of the strong relationship between
important in the onset of clinical retinopthy.94 A direct relation- diabetes control and the subsequent development of ocular and
ship between decreased retinal blood flow and the activation of other medical complications. The association of hypertension,
protein kinase C in the rat model has been shown.95 Oxidant dyslipidemia, abdominal obesity, eating disorders, renal disease,
stress may also be involved in the early progression of DR pregnancy, joint contractures, cardiovascular disease, and
suggesting that antioxidants may have therapeutic usefulness. peripheral neuropathy with onset and progression of DR should
Modulators of NPDR may also include vasoactive agents such be noted and care taken to assure that the patient has
as angiotensin II, histamine, and oxygen. A wide array of other appropriate medical follow-up for these conditions. Patients
targets are evolving from the studies of angiogenic agents and with permanent visual impairment, including legal or total
their mechanism of action over the past decade based on work blindness, should be informed of the availability of visual,
initiated by Michelson nearly six decades ago.96–106 vocational, and psychosocial rehabilitation programs. Diabetic
women contemplating pregnancy should have a complete eye
PATIENT CARE AND GUIDELINES examination prior to conception if possible. Since pregnancy
may dramatically exacerbate existing retinopathy and may be
Although this is an exciting period with new and forthcoming associated with hypertension, diabetic women should have their
clinical trial data on a variety of novel therapeutic approaches, eyes examined early in the first trimester of pregnancy and
until we have treatment modalities to prevent or cure diabetic generally at least each trimester thereafter and again 1–2
retinopathy, the emphasis for our patients with diabetes months post partum. Severe retinopathy may impact optimal
1788 remains focused upon early identification, accurate determi- delivery methods.
Diagnosis, Management, and Treatment of Nonproliferative Diabetic Retinopathy

CONCLUSIONS team share the responsibility of assuring such care is offered to


the patient.
In its earliest stages, DR usually causes no symptoms. Visual Faced with the current inability to prevent or cure diabetic
acuity may be excellent, and the patient may be completely retinopathy, the eye care for patients with diabetes must pri-
unaware of even advanced retinopathy. Preservation of vision can marily focus on patient access, early detection, accurate
be maximized by early initiation of a careful eye care program retinopathy assessment, careful medical and ophthalmic follow-
which includes patient education, close follow-up, and efficient up, timely laser photocoagulation and appropriate use of novel
communication between the entire team of health care providers. therapies. With this approach, and the continuum of connected
Accurate assessment of DR severity is essential on an ongoing diabetes eye and medical care evolving from advances in
basis to determine the likelihood of retinopathy progression, information technology and telemedicine, our twenty-first
risk of vision loss and timing of follow-up and therapy. Optimal century mission of preserving vision in patients with diabetes
control of systemic factors such as blood glucose, blood will become increasingly successful.
pressure, and lipids is also critical. All members of the health

REFERENCES
1. Aiello LM, Beetham WP, Balodimos MC, et 13. Diabetic Retinopathy Study Report Number Airlie House Classification. Ophthalmology

CHAPTER 133
al: Ruby laser photocoagulation in 12: Macular edema in Diabetic Retinopathy 1991; 98:786–806.
treatment of diabetic proliferating Study patients. Ophthalmology 1987; 26. Early Treatment Diabetic Retinopathy Study
retinopathy: Preliminary report. In: Goldberg 94:754–760. Report Number 11: Classification of
MF, Fine S, eds. Symposium on the 14. Diabetic Retinopathy Study Report Number diabetic retinopathy from fluorescein
Treatment of Diabetic Retinopathy. Public 13: Factors associated with visual outcome angiograms. Ophthalmology 1991;
Health Service Publication No. 1890. after photocoagulation for diabetic 98:807–822.
Washington, DC: US Government Printing retinopathy. Invest Ophthalmol Vis Sci 1989; 27. Early Treatment Diabetic Retinopathy Study
Office; 1969:437–463. 30:23–28. Report Number 12: Fundus photographic
2. Diabetic Retinopathy Study Report Number 15. Diabetic Retinopathy Study Report Number risk factors for progression of diabetic
1: Preliminary report on effects of 14: Indications for photocoagulation retinopathy. Ophthalmology 1991; 98:823.
photocoagulation therapy. Am J Ophthalmol treatment of diabetic retinopathy. Int 28. Early Treatment Diabetic Retinopathy Study
1976; 81:1–14. Ophthalmol Clin 1987; 27:239–253. Report Number 13: Fluorescein
3. Diabetic Retinopathy Study Report Number 16. Early Treatment Diabetic Retinopathy Study angiographic risk factors for progression of
2: Photocoagulation of proliferative diabetic Report Number 1: Photocoagulation for diabetic retinopathy. Ophthalmology 1991;
retinopathy. Ophthalmology 1978; 85:82. diabetic macular edema. Arch Ophthalmol 98:834–840.
4. Diabetic Retinopathy Study Report Number 1985; 103:1796–1806. 29. Early Treatment Diabetic Retinopathy Study
3: Four risk factors for severe visual loss in 17. Early Treatment Diabetic Retinopathy Study Report Number 14: Aspirin effects on
diabetic retinopathy. Arch Ophthalmol 1979; Report Number 2: Treatment techniques mortality and morbidity in patients with
97:658. and clinical guidelines for photocoagulation diabetes mellitus. JAMA 1992;
5. Diabetic Retinopathy Study Report Number of diabetic macular edema. Ophthalmology 268:1292–1300.
4: A short report of long-range results. 1987; 94:761–774. 30. Chew EY, Williams GA, Burton TC, et al:
Proceedings of the 10th Congress of 18. Early Treatment Diabetic Retinopathy Study Early Treatment Diabetic Study Report 16.
International Diabetes Federation. New Report Number 3: Techniques for scatter Aspirin effects on the development of
York: Excerpta Medica; 1980. and local photocoagulation treatment of cataracts in patients with diabetes mellitus.
6. Diabetic Retinopathy Study Report Number diabetic retinopathy. Int Ophthalmol Clin Arch Ophthalmol 1992; 110:339–342.
5: Photocoagulation treatment of 1987; 27:254–264. 31. Flynn HW, Chew EY, Simons BD, et al: Early
proliferative diabetic retinopathy. 19. Early Treatment Diabetic Retinopathy Study Treatment Diabetic Retinopathy Study
Relationship of adverse treatment effects to Report Number 4: Photocoagulation for Report Number 17. Pars plana vitrectomy in
retinopathy severity. Dev Ophthalmol 1981; diabetic macular edema. Int Ophthalmol the Early Treatment Diabetic Retinopathy
2:1–15. Clin 1987; 27:265–272. Study. Ophthalmology 1992; 99:1351–1357.
7. Diabetic Retinopathy Study Report Number 20. Early Treatment Diabetic Retinopathy Study 32. Early Treatment Diabetic Retinopathy Study
6: Design, methods, and baseline results. Case Reports Numbers 3 and 4: Case Report Number 19: Focal photocoagulation
Invest Ophthalmol Vis Sci 1981; reports to accompany Early Treatment treatment of diabetic macular edema:
21:149–209. Diabetic Retinopathy Study Reports Relationship of treatment effect to
8. Diabetic Retinopathy Study Report Number Numbers 3 and 4. Int Ophthalmol Clin fluorescein angiographic and other retinal
7: A modification of the Airlie House 1987; 27:273–333. characteristics at baseline. Arch Ophthalmol
Classification of Diabetic Retinopathy. 21. Early Treatment Diabetic Retinopathy Study 1995; 113:1144–1155.
Invest Ophthalmol Vis Sci 1981; Report Number 5: Detection of diabetic 33. Chew EY, Klein ML, Murphy RP, et al: Early
21:210–226. macular edema. Ophthalmoscopy versus Treatment Diabetic Retinopathy Study
9. Diabetic Retinopathy Study Report Number photography. Ophthalmology 1989; Report Number 20. Effects of aspirin on
8: Photocoagulation treatment of 96:746–751. vitreous/preretinal hemorrhage in patients
proliferative diabetic retinopathy. Clinical 22. Early Treatment Diabetic Retinopathy Study with diabetes mellitus. Arch Ophthalmol
application of Diabetic Retinopathy Study Report Number 7: Early Treatment Diabetic 1995; 113:52–55.
(DRS) Findings. Ophthalmology 1981; Retinopathy Study design and baseline 34. Chew EY, Klein ML, Ferris FL III, et al: Early
88:583–600. patient characteristics. Ophthalmology Treatment Diabetic Retinopathy Study
10. Diabetic Retinopathy Study Report Number 1991; 98:741–756. Report Number 22. Association of elevated
9: Assessing possible late treatment effects 23. Early Treatment Diabetic Retinopathy Study serum lipid levels with retinal hard exudates
in stopping clinical trials early: a case study. Report Number 8: Effects of aspirin in diabetic retinopathy. Arch Ophthalmol
Controlled Clin Trials 1984; 5:373–381. treatment on diabetic retinopathy. 1996; 114:1079–1084.
11. Diabetic Retinopathy Study Report Number Ophthalmology 1991; 98:757–765. 35. Diabetic Retinopathy Vitrectomy Study
10: Factors influencing the development of 24. Early Treatment Diabetic Retinopathy Study Report Number 1: Two-year course of visual
visual loss in advanced diabetic retinopathy. Report Number 9: Early photocoagulation acuity in severe proliferative diabetic
Invest Ophthalmol Vis Sci 1985; for diabetic retinopathy. Ophthalmology retinopathy with conventional management.
26:983–991. 1991; 98:766–785. Ophthalmology 1985; 92:492–502.
12. Diabetic Retinopathy Study Report Number 25. Early Treatment Diabetic Retinopathy Study 36. Diabetic Retinopathy Vitrectomy Study
11: Intraocular pressure following panretinal Report Number 10: Grading diabetic Report Number 2: Early vitrectomy for
photocoagulation for diabetic retinopathy. retinopathy from stereoscopic color fundus severe vitreous hemorrhage in diabetic
Arch Ophthalmol 1987; 105:807–809. photographs-An extension of the modified retinopathy. Two-year results of a 1789
RETINA AND VITREOUS

randomized trial. Arch Ophthalmol 1985; 54. Chase HP, Jackson WE, Hoops SL, et al: of intensive therapy. N Engl J Med 2000;
103:1644–1652. Glucose control in the renal and retinal 342:381–389.
37. Diabetic Retinopathy Vitrectomy Study complications of insulin-dependent 70. The Writing Team for the Diabetes Control
Report Number 3: Early vitrectomy for diabetes. JAMA 1989; 261:1155–1160. and Complications Trial/ Epidemiology of
severe proliferative diabetic retinopathy in 55. Brinchmann-Hansen O, Dahl-Jorgensen K, Diabetes Interventions and Complications
eyes with useful vision. Results of a Hanssen KF, et al: Effects of intensified Research Group: Effect of intensive therapy
randomized trial. Ophthalmology 1988; insulin treatment on various lesions of on the microvascular complications of type
95:1307–1320. diabetic retinopathy. Am J Ophthalmol 1 diabetes mellitus. JAMA 2002;
38. Diabetic Retinopathy Vitrectomy Study 1985; 100:644–653. 287:2563–2569.
Report Number 4: Early vitrectomy for 56. Krolewski AS, Canessa M, Warram JH, 71. Kohner EM, Aldington SJ, Stratton IM, et
severe proliferative diabetic retinopathy in et al: Predisposition to hypertension and al: Diabetic retinopathy at diagnosis of non-
eyes with useful vision. Clinical application susceptibility to renal disease in insulin- insulin-dependent diabetes mellitus and
of results of a randomized trial. dependent diabetes mellitus. N Engl J Med associated risk factors: United Kingdom
Ophthalmology 1988; 95:1331–1334. 1988; 318:140–145. Prospective Diabetes Study, 30. Arch
39. Diabetic Retinopathy Vitrectomy Study 57. Aiello LP, Cahill MT, Wong JS: Perspective: Ophthalmol 1998; 116:297–303.
Report Number 5: Early vitrectomy for Systemic considerations in the 72. UK Prospective Diabetes Study Group:
severe vitreous hemorrhage in diabetic management of diabetic retinopathy. Am J Effect of intensive blood-glucose control
retinopathy. Four-year results of a Ophthalmol 2001; 132:760–776. with sulphonylureas or insulin compared
randomized trial. Arch Ophthalmol 1990; 58. Stern MP, Patterson JK, Haffner SM, et al: with conventional treatment and risk of
108:958–964. Lack of awareness and treatment of complications in patients with type 2
40. Cowie CC, Rust KF, Byrd-Holt D, et al: hyperlipidemia in type II diabetes in a diabetes: UKPDS 33. Lancet 1998;
SECTION 10

Prevalence of diabetes and impaired community survey. JAMA 1989; 352:837–853.


fasting glucose in adults – United States, 262:360–364. 73. Ohkubo Y, Kishikawa H, Araki E, et al:
1999–2000. MMWR 2003; 52:833–837. 59. Fong DS, Warram JH, Aiello LM, et al: Intensive insulin therapy prevents the
41. National Diabetes Surveillance System. Cardiovascular autonomic neuropathy and progression of diabetic microvascular
Centers for Disease Control and proliferative diabetic retinopathy. Am J complications in Japanese patients with
Prevention. Available: Ophthalmol 1995; 120:317–321. non-insulin-dependent diabetes mellitus: a
http://www.cdc.gov/diabetes/statistics/inde 60. Diabetes Prevention Program Research randomized prospective 6-year study.
x/htm. Accessed 12 April 2006. Group: Diet and exercise dramatically delay Diabetes Res Clin Pract 1995; 28:103–117.
42. National Diabetes Fact Sheet: CDC’s type 2 diabetes. Reduction in the incidence 74. Wilkinson CP, Ferris FL III, Klein RE, et al:
Diabetes Program-Publications & Products; of type 2 diabetes with lifestyle intervention Proposed international clinical diabetic
2003. or metformin. N Engl J Med 2002; retinopathy and diabetic macular edema
43. Klein R, Klein BEK, Moss SE, et al: The 346:393–403. disease severity scales. Ophthalmology
Wisconsin Epidemiologic Study of Diabetic 61. Chiasson JL, Josse RG, Gomis R, et al: 2003; 110:1677–1682.
Retinopathy. II. Prevalence and risk of Acarbose for prevention of type 2 diabetes 75. Chew EY: A simplified diabetic retinopathy
diabetic retinopathy when age at diagnosis mellitus: the STOP-NIDDM randomised scale. Ophthalmology 2003; 110:1675–1676.
is less than 30 years. Arch Ophthalmol trial. Lancet 2002; 359:2072–2077. 76. Bresnick GH: Background diabetic
1984; 102:520–526. 62. Ferris FL III: Early photocoagulation in retinopathy. In: Ryan SJ, ed. Retina.
44. Klein R, Klein BEK, Moss SE, et al: The patients with either Type I or Type II diabetes. Medical Retina. St Louis: Mosby;
Wisconsin Epidemiologic Study of Diabetic Tr Am Ophth Soc 1996; 94:505–537. 1989:327–366.
Retinopathy. III. Prevalence and risk of 63. The Sorbinil Retinopathy Trial Research 77. Shimizu K, Kobayashi Y, Muraoka K:
diabetic retinopathy when age at diagnosis Group: A randomized trial of sorbinil, an Midperipheral fundus involvement in
is 30 or more years. Arch Ophthalmol 1984; aldose reductase inhibitor in diabetic diabetic retinopathy. Ophthalmology 1981;
102:527–532. retinopathy. Arch Ophthalmol 1990; 88:601–612.
45. Vision Problems in the US Prevent 108:1234–1244. 78. Takashi N, Muroka K, Shimizu K:
Blindness America, National Eye Institute; 64. The Sorbinil Retinopathy Trial Research Distribution of capillary nonperfusion in
2002. Group. The Sorbinil Retinopathy Trial: early-stage diabetic retinopathy.
46. The Eye Diseases Prevalence Research Neurology results. Neurology 1993; Ophthalmology 1984; 91:1431–1439.
Group: The Prevalence of Diabetic 43:1141–1149. 79. Schuman JS, Puliafito CA, Fujimoto JG:
Retinopathy Among Adults in the United 65. The Diabetes Control and Complications Optical coherence tomography of ocular
States. Arch Ophthalmol 2004; Trial Research Group: The effect of disease second edition. Slack Copyright;
122:552–563. intensive treatment of diabetes on the 2004.
47. Ali FA:. A Review of Diabetic Macular development and progression of long term 80. The Diabetic Retinopathy Clinical Research
Edema. DJO 2002. complications in insulin-dependent Network: The Relationship between OCT-
48. Aiello LM: Perspective on diabetic diabetes mellitus. N Engl J Med 1993; measured Central Retinal Thickness and
retinopathy. Am J Ophthalmol 2003; 329:977–986. Visual Acuity in Diabetic Macular Edema.
136:122–135. 66. The Diabetes Control and Complications Ophthalmology 2007; 114:525–536.
49. Moloney JBM, Drury MI: The effect of Trial Research Group: The relationship of 81. The Diabetic Retinopathy Clinical Research
pregnancy on the natural course of diabetic glycemic exposure (HbA1c) to the risk of Network: Diurnal Variation in Retinal
retinopathy. Am J Ophthalmol 1982; development and progression of Thickening Measurement by OCT in
93:745–756. retinopathy in the Diabetes Control and Center-involved Diabetic Macular Edema.
50. Serup L: Influence of pregnancy on diabetic Complications Trial. Diabetes 1995; Arch Ophthalmol 2006; 124:1701–1707.
retinopathy. Acta Endocrinol 1986; 44:968–983. 82. Diabetic Retinopathy Clinical Research
277:122–124. 67. The Diabetes Control and Complications Network: Comparison of Modified-ETDRS
51. Phelps RL, Sakol P, Metzger BE, et al: Trial Research Group: Progression of and Mild Macular Grid Laser
Changes in diabetic retinopathy during retinopathy with intensive versus Photocoagulation Strategies for Diabetic
pregnancy: Correlations with regulation of conventional treatment in the Diabetes Macular Edema. Arch Ophthalmol 2007;
hyperglycemia. Arch Ophthalmol 1986; Control and Complications Trial. 125:469–480.
104:1806–1810. Ophthalmology 1995; 102:647–661. 83. Diabetic Retinopathy Clinical Research
52. The Kroc Collaborative Study Group: Blood 68. The Diabetes Control and Complications Network: Retinal Subspecialty Day
glucose control and the evolution of Trial Research Group: Hypoglycemia in the Symposium, American Academy of
diabetic retinopathy and albuminuria. N Diabetes Control and Complications Trial. Ophthalmology Annual Meeting. Las Vegas;
Engl J Med 1984; 311:365–372. Diabetes 1997; 46:271–286. 2006.
53. Grunwald JE, Riva CE, Martin DB, et al: 69. Diabetes Control and Complications 84. Spaide RF, Fisher YL: Intravitreal
Effect of insulin-induced decrease in blood Trial/Epidemiology of Diabetes Intervention bevacizumab (Avastin) treatment of
glucose on the human diabetic retinal and Complications Research Group: proliferative diabetic retinopathy
circulation. Ophthalmology 1987; Retinopathy and nephropathy in patients complicated by vitreous hemorrhage.
1790 94:1614–1620. with type 1 diabetes four years after a trial Retina 2006; 26:275–278.
Diagnosis, Management, and Treatment of Nonproliferative Diabetic Retinopathy

85. Avery RL, Joel Pearlman J, Pieramici DJ, et 95. Shiba T, Bursell S-E, Clermont A, et al: 106. Ishii H, Jiroousek MR, Koya D, et al:
al: Intravitreal bevacizumab (Avastin) in the Protein kinase C (PKC) activation is a Amerlioration of vascular dysfunctions in
treatment of proliferative diabetic causal factor for the alteration of retinal diabetic rats by an oral PKC beta inhibitor.
retinopathy. Ophthalmology 2006; blood flow in diabetes of short duration. Science 1996; 279:728–731
113:1695–1715. Invest Ophthalmol Vis Sci 1991; 32:785. 107. Javitt JC, Aiello LP, Bassi LJ, et al:
86. Chun DW, Heier JS, Topping TM, et al: A 96. Michaelson IC. The mode of development Detecting and treating retinopathy in
pilot study of multiple intravitreal injections of the vascular system of the retina, with patients with type I diabetes mellitus.
of ranibizumab in patients with center- some observations on its significance for Savings associated with improved
involving clinically significant diabetic certain retinal disease. Trans Ophthalmol implementation of current guidelines.
macular edema. Ophthalmology 2006; Soc UK 1948; 68:137–180. Ophthalmology 1991; 98:1565–1574.
113:1706–1712. 97. Ashton N: Retinal vascularization in health 108. Aiello LM, Cavallerano J, Cavallerano A,
87. Macugen Diabetic Retinopathy Study and disease. Am J Ophthalmol 1957; Bursell S-E. The Joslin Vision Network
Group: A phase II randomized double- 44:7–24. (JVN) Innovative Telemedicine Care for
masked trial of pegaptanib, an anti- 98. Aiello LP, Avery RL, Arrigg PG, et al: Diabetes. Ophthalmol Clin North Am 2000;
vascular endothelial growth factor aptamer, Vascular endothelial growth factor in ocular 13: 213–224.
for diabetic macular edema. fluids of patients with diabetic retinopathy 109. Wilson C, Horton M, Cavallerano J, Aiello
Ophthalmology 2005; 112:1747–1757. and other ocular disorders. N Engl J Med LM. Addition of primary-care based retinal
88. Jirousek MR, Gillig JR, Gonzalez CM, et al: 1994; 331:1480–1487. imaging technology to an existing eye care
(S)-13-[(dimethylamino)methyl]- 99. Amin RH, Frank RN, Kennedy A, et al: professional referral program increased the
10,11,14,15-tetrahydro-4,21; 9:16- Vascular endothelial growth factor is rate of surveillance and treatment of

CHAPTER 133
dimetheno-1H, 13H- present in glial cells of the retina and optic diabetic retinopathy. Diabetes Care 2005;
dibenzo[e,k]pyrrolo[3,4- nerve of human subjects with 28:318–322.
h][1,4,13]oxadiazacyclohexadecene- nonproliferative diabetic retinopathy. Invest 110. Bursell S-E, Cavallerano JD, Cavallerano
1,3(2H)-d ione (LY333531) and related Ophthalmol Vis Sci 1997; 38:36–47. AA, et al: Stereo nonmydriatic digital-video
analogues: isozyme selective inhibitors of 100. Pierce EA, Avery R, Folet ED, et al: color retinal imaging compared with Early
protein kinase C beta. J Med Chem 1996; Vascular endothelial growth factor/vascular Treatment Diabetic Retinopathy Study
39:2664–2671. permeability factor expression a mouse seven standard field 35-mm stereo color
89. Ishii H, Jirousek MR, Koya D, et al: model of retinal neovascularization. Proc photos for determining level of diabetic
Amelioration of vascular dysfunctions in Natl Aca Sci USA 1995; 92:905–909. retinopathy. Ophthalmology 2001;
diabetic rats by an oral PKC beta inhibitor 101. Adamis AP, Miller JW, Bernal MT: Increased 108:572–585.
[see comments]. Science 1996; vascular endothelial growth factor levels in 111. Cavallerano AA, Cavallerano JD, Katalinic
272:728–731. the vitreous of eyes with proliferative P, et al: Use of Joslin Vision Network
90. Aiello LP. The potential role of PKC beta in diabetic retinopathy. Am J Ophthalmol digital-video nonmydriatic retinal imaging to
diabetic retinopathy and macular edema. 1994; 118:445–450. assess diabetic retinopathy in a clinical
Surv Ophthalmol 2002; 47:S263–S269. 102. Tilton RG, Kawamura T, Chang KC, et al: program. Retina 2003; 23:215–223.
91. The PKC-DRS Study Group: The effect of Vascular dysfunction induced by elevated 112. Cavallerano JD, Aiello LP, Cavallerano AA,
ruboxistaurin on visual loss in patients with glucose levels in rats mediated by Vascular et al: Nonmydriatic digital retinal imaging
moderately severe to very severe endothelial growth factor. J Clin Invest alternative for annual retinal examination in
nonproliferative diabetic retinopathy: initial 1997; 99:2192–2202. persons with previously documented no or
results of the Protein Kinase C beta 103. Pu X, Aiello LP, Ishii H, et al: mild diabetic retinopathy. Am J Ophthalmol
Inhibitor Diabetic Retinopathy Study (PKC- Characterization of vascular endothelial 2005; 140:667–673.
DRS) multicenter randomized clinical trial. growth factor’s effect on the activation of 113. Pey S-P, Aiello LM, Cavallerano JD, et al:
Diabetes 2005; 54:2188–2197. protein kinase C, its isoforms and Comparison of nonmydriatic digital retinal
92. The PKC-DRS2 Research Group: Effect of endothelial growth. J Clin Invest 1996; imaging vs. dilated ophthalmic examination
ruboxistaurin on visual loss in patients with 98:2018–2026. for nondiabetic eye disease in persons with
diabetic retinopathy. Ophthalmology 2006; 104. Aiello LP, Bursell SE, Clermont A, et al: diabetes. Ophthalmology 2006;
113:2221–2230. Vascular endothelial growth factor-induced 113:833–840.
93. McGill JB, King GL, Berg PH, et al: Clinical retinal permeability is mediated by protein 114. Cavallerano J, Lawrence MG, Zimmer-
safety of the selective PKC-beta inhibitor, kinase C in vivo and suppressed by an Galler I, et al: Telehealth practice
ruboxistaurin. Expert Opin Drug Saf 2006; orally effective beta isoform-selective recommendations for diabetic retinopathy.
5:835–845. inhibitor. Diabetes 1997; 46:1473–1480. Telemedicine J e-Health 2004; 10:469–482.
94. de la Rubia Sanchez G, Oliver-Pozo J, 105. Robinson GS, Pierce EA, Rook SL, et al:
Shiba T, King GL: Modulation of Oligodeoxynucleotides inhibit retinal
endothelin-1 (ET-1) receptor on retinal neovascularization in a murine model of
pericytes by elevated glucose levels. Invest proliferative retinopathy. Proc Natl Acad Sci
Ophthalmol Vis Sci 1991; 32:3110. 1996; 93:4851–4856.

1791
CHAPTER

134 Diabetic Macular Edema


Ronald P. Danis

Overview Key Features: ETDRS Criteria for Clinically Significant


Macular Edema
Diabetic macular edema (DME) remains the most common cause • Retinal thickening at the center of the macula.
of vision loss among diabetic patients. New understanding of the • Retinal thickening and/or adjacent hard exudates at or within
underlying pathophysiology has heightened interest in the 500 microns of the center of the macula.
potential benefits of specific pharmacologic therapy, such as • An area of retinal thickening greater than or equal to one disc
treatment with intraocular steroids, anti-vascular endothelial area, any part of which is within 1 disc diameter of the center
growth factor (VEGF), and protein kinase C-beta (PKCb) of the macula.
inhibition. At this time, laser photocoagulation according to the • A disk diameter is a standardized unit of measurement
guidelines of the Early Treatment of Diabetic Retinopathy Study derived from the measurement of the average optic nerve
(ETDRS) continues to be the primary standard of care treatment head on slide film images and, in the ETDRS, presumed to
in most communities, while the evidence base for alternative or correspond to 1.5 mm on the retina.
supplementary treatments accrues. Recent advances in imaging • Most digital camera systems today use a revised
technology, most notably the wide commercial availability and assumption that a disc diameter measures 1.8 mm for disc
acceptance of optical coherence tomography (OCT), have diameter and a disk area 2.54 mm2.
yielded new tools for the clinician and researcher in monitoring • The disc diameter or disc area as a measurement on the
photographic image is the same, the underlying
macular edema progression and response to treatment.
presumptions regarding the image magnification scale
have changed.

DEFINITION
Diabetic macular edema is manifested as retinal thickening at less severe stages.3 In recognition that eyes with severe
primarily due to exudation from incompetent macular retinal edema at the center of the macula may not respond well to laser
capillaries. While retinal thickening may occur peripheral to the treatment, and with the availability of new pharmacologic
macula, such edema is not vision threatening. In order to define agents administered by intravitreal injection, many clinicians
a threshold severity level of edema at which retreatment was are using drugs off-label to treat DME, without the evidence
specified for the protocol, the ETDRS coined the term ‘clinically from large randomized trials.
significant macular edema’.1,2 This term has been incorporated
into most clinicians’ understanding as a level of disease at PREVALENCE AND INCIDENCE
which it is appropriate to initiate laser treatment. The phrase
clinically significant macular edema is often applied as a clinical In the first detailed population-based assessment of diabetic
description of ocular status (e.g., the eye has ‘non-clinically retinopathy (DR), the Wisconsin Epidemiologic Study of
significant DME’). While a useful and time-honored term, a Diabetic Retinopathy (WESDR), documented a prevalence of
number of other descriptors can be applied to the clinical macular edema (defined as retinal thickening within one
appearance of DME which may be more appropriate as new disk-diameter of the macular center in stereoscopic retinal
treatments become available. For instance, an eye with photographs) of 11.1% overall among patients with diabetes in
clinically significant macular edema by the ETDRS definition Southern Wisconsin in the early 1980s.4 The prevalence was
could have center involvement with cystoid changes, or, it could slightly higher in early onset compared to older-onset diabetes
have focal edema with a normal foveal contour and thickness. and was strongly associated with duration of diabetes and
These very different presentations may merit different glycemic control. Proteinuria and vascular hypertension were
treatments. additional factors associated with increased prevalence.
The standard of care for varying presentations of DME is in Subsequent population-based studies have yielded prevalence
evolution at this time. For several decades, the guidelines of the rates between 2%,5 and over 10%6–9 and confirmed the afore-
ETDRS were generally applied in the medical community to mentioned associations. In some diabetic populations, the
manage patients with a level of DME meeting threshold criteria prevalence of retinopathy, including DME, appears to be declining
for clinically significant macular edema, particularly when the due to improved glycemic control of diabetes in the community.6,10
center of the macula was involved or immanently threatened by However, the absolute prevalence of DME might be increasing
retinal thickening or hard exudates very close to it. However, due to the overall increased prevalence of diabetes in
recent clinical trials have been investigating treatment of DME industrialized nations.11 The incidence of new cases of DME 1793
RETINA AND VITREOUS

in a diabetic population has been characterized in a population- Hypertension significantly exacerbates vision loss from
based longitudinal assessment within the WESDR. The WESDR macular edema.22,31 The UKPDS has demonstrated that strict
determined a 10 year rate of developing DME of 20.1% in control of systemic hypertension has an ameliorative effect on
younger onset diabetic patients, 25.4% in older-onset diabetic retinopathy progression and vision loss from macular edema in
patients taking insulin, and 13.9% in the older-onset group type 2 diabetes.32,33 The WESDR study also documented an
not taking insulin.12 It is expected that the incidence of DME adverse association of hypertension with vision loss from diabetic
will decrease as excellent metabolic control is increasingly retinopathy.31 It has been suggested that not only is hyper-
embraced as a therapeutic goal by patients and health care tension a risk factor for the development of macular edema, but
workers. its treatment may have important benefits in patients with
uncontrolled hypertension.34
CLINICAL ASSOCIATIONS AND RISK Proteinuria and retinopathy are strongly correlated because of
FACTORS their common risk factors of glycemic control, duration of
diabetes, and hypertension.17,31,35–37 Less clear is the relationship
Macular edema is strongly positively associated with diabetic between renal failure and macular edema. There appear to be
retinopathy severity.12 This is particularly problematic for the patients who have increased macular edema as a collateral
clinician who must frequently manage coexistent DME and adverse event due to fluid retention, and who improve after
proliferative diabetic retinopathy (PDR), as will be discussed. institution of diuretics or hemodialysis,38 but this is difficult
Undoubtedly, the molecular processes leading to the loss of to demonstrate among populations of patients undergoing
SECTION 10

retinal capillary integrity and subsequent edema are closely dialysis.34,39,40 Retinal HE and macular edema were significantly
related to, but not identical to those causing capillary closure and independently associated with proteinuria in a cohort of
and ultimate neovascularization. This is evident from the African American patients.41 It is accepted that fluid retention
observation that most patients with macular edema do not have from cardiac failure, renal failure, or other causes can exacerbate
PDR, and many patients with proliferative retinopathy do not DME and may be important concerns in managing it.1
have DME.13 This partial disconnect between related com- Dyslipidemia has been implicated as an independent risk
plications in the same tissue led the ETDRS to exclude retinal factor for vision loss and DME.42–44 This may be in part due to
thickening from the overall diabetic retinopathy (DR) severity increased quantities of HE in the eyes of hyperlipidemic diabetic
scale.14,15 The severity scale is based on rate of progression to patients, which has been a consistent observation between
high-risk PDR, and retinal thickening per se was not identified studies.41,45–48 However, such observations also suggest that
as conferring additional risk beyond that represented by the dyslipoproteinemia or excessive dietary exposure to fats may
other abnormalities characteristic of diabetic retinopathy. contribute to macular edema, perhaps through lipid peroxidation
Glycemic control is a conclusively identified risk factor for or other vasculopathic influences.49–51
retinopathy progression as well as for DME. The Diabetes Development or worsening of DME during pregnancy is
Complications and Control Trial (DCCT) evaluated intensive occasionally observed and is most often associated with poor
glycemic control versus good control in type 1 diabetic patients glycemic control,32,52 preexisting retinopathy, and pre-
with no or minimal DR over 4–6 years, and found a marked eclampsia.53–55 The DME associated with pregnancy may at
reduction in the progression of retinopathy, nephropathy, and least partially respond to laser treatment, but may also resolve
neuropathy.16 In the follow-on study of this cohort, the postpartum.56 Clinicians faced with a pregnant patient with
Epidemiology of Diabetes Interventions and Control (EDIC) DME may, therefore, elect to observe the patient without treat-
study demonstrated that subjects in the former intensive treat- ment for several months during her pregnancy, and following
ment group had a lower rate of progression to DME requiring her delivery before making the determination that laser
laser treatment than those in the good control group.17,18 treatment is required. With excellent prenatal care and careful
Similar results have been demonstrated in type 2 diabetic metabolic control, those women who experience some retino-
patients in the United Kingdom Prospective Diabetes Study pathy worsening during pregnancy generally have only transient
(UKPDS).19 Clear relationships between serum glycohemo- changes.32,53,57 Some of the worsening that may occur during
globin levels and DR complications have been established.20–22 pregnancy may be due to rapid institution of strict glycemic
Older-onset patients requiring insulin have a higher incidence control, which can cause transient worsening as well.58–60
of macular edema (and other diabetic complications) than non- Cataract surgery, other types of intraocular surgery, and ocular
insulin dependent diabetic patients.23 This is likely an effect inflammatory disease may produce inflammatory and angio-
related to poorer glycemic control in patients ultimately genic mediators which can produce macular edema in eyes with
requiring insulin therapy. or without diabetic retinopathy.61 However, eyes with pre-existing
Duration of diabetes is strongly correlated with prevalence DME are at high risk for exacerbation. Patients with cataract
and incidence of macular edema, retinopathy progression, and and DME, who undergo surgery, will occasionally experience vision
other diabetic complications.12,24 The diagnosis of diabetes in worsening due to increased DME rather than the expected
type 2 subjects occasionally occurs some time after subclinical visual improvement from cataract removal.62,63 Therefore,
diabetes has been manifest, which yields a small proportion of DME should be treated prior to surgery to minimize this risk.64
patients who may present with macular edema at the time Laser treatment for PDR, panretinal photocoagulation (PRP), or
of diagnosis, or even have decreased vision from macular edema scatter laser is documented to cause vision loss due to worsening
as the presenting sign.25–27 In contrast, persons with type 1 macular edema.65 Eyes with preexisting macular edema appear to
diabetes are very unlikely to experience advanced retinopathy be more at risk. The ETDRS Research group suggested that in eyes
and macular edema before 5 years of duration. Age-corrected with both DME and PDR, the risk of vision loss from PRP was
prevalence rates suggest that there is no definite age interaction minimized by first treating the DME with macular focal/grid
with duration of disease8,11 but because of the increasing inci- photocoagulation, and later treating the PDR (if such a delay is
dence of diabetes with age, retinopathy rates are correspondingly acceptable).1 The underlying mechanism may be the production of
higher with advancing age. Consequently, early detection of an inflammatory response to the laser treatment, which worsens
type 2 diabetes and institution of excellent glycemic control is vascular leakage and exacerbates DME.66 This has led some
paramount in decreasing the risk of vision loss from macular investigators to consider adjunctive steroid treatment with PRP to
1794 edema in the older population.28–30 minimize the risk of early vision loss.67,68
Diabetic Macular Edema

Extracellular fluid in the retina may cause diffuse thickening


Key Features: Clinical Associations with Diabetic
of the tissue. In early stages, Mueller cell and other glial cell
Macular Edema Severity
edema can be observed, which appear to be compensatory.91,92
• Diabetic retinopathy severity level
Severe edema generally results in development of loculated
• Duration of Diabetes
fluid, called cystoid spaces, in the middle retinal layers (Fig.
• Glycemic control
134.1). Cystoid spaces initially develop in the outer plexiform
• Hypertension
layer of the retina, hypothesized to be the loosest and most
• Dyslipidemia
elastic cellular stratum of the retina and therefore the most
• Nephropathy
accommodating layer for this expansion.93 In very severe or
• Fluid retention
chronic edema, the cystoid spaces may occupy nearly the entire
• Pregnancy
retinal thickness with gross distortion of the retina. In very
• Intraocular surgery
chronic cases, there is eventual disorganization of the retinal
• Uveitis
tissue, which may be partly related to co-existent capillary
• Panretinal Photocoagulation
ischemia and reactive glial responses. At this point, the retina
may resume normal thickness, or even become abnormally
thin, as the macula assumes a more atrophic appearance.
PATHOLOGY AND PATHOPHYSIOLOGY Cystoid spaces may rapidly disappear with treatment, yielding a
normal-appearing retina in cases where chronic cellular changes

CHAPTER 134
OF DME
have not yet occurred.
Capillary damage is manifest in the earliest stages of DR by loss In the foveal region, the outer plexiform layer has a radial
of the retinal pericytes and capillary basement membrane orientation due to the centrifugal displacement of the bipolar
thickening.69 With increasing duration and severity of hyper- cell layer, the nerve-fiber layer of Henle. This layer may host
glycemia, eventual capillary closure and microaneurysm cystoid spaces of very large proportions. Most cases of clinically
formation occur.70 At what point, the blood–retinal permeability apparent cystoid change are in this region, due to the large size
barrier is permanently compromised in this cascade of events is of the cysts. The clinical cyst pattern often appears as one or
not clear, but hyperglycemia alone of relatively short duration more central large cysts surrounded by a region of somewhat
can cause retinal vascular leakage in the absence of detectable smaller cysts, and finally by a region of smaller cysts that may
retinopathy or structural damage by light microscopy in animal not be clinically visible (Fig. 134.2). Fluorescein angiography
models.71 The intimate ultrastructural connections between the has been historically helpful in defining cystoid edema, since
capillary cells and the neighboring neuronal and glial tissue the fluorescein dye from the leaking capillaries tends to pool in
suggest important functional relationships.72,73 This raises the the cystoid spaces in the late phases of the study, highlighting
question as to whether the observed vascular changes are primary these structures (Figs 134.2d and 134.3d).94 OCT has allowed
or secondary to neuroglial dysfunction. The ultrastructural visualization of high resolution cross sectional retinal morphology
basis for the blood–retinal barrier is the tight junctions between and demonstrated that cystoid spaces are more common than
the retinal capillary endothelial cells and the relative paucity of has been appreciated by clinical examination or fluorescein
transendothelial transport vesicles compared to endothelial cells angiography (Figs 134.2d and 134.3d).95 An additional feature
in other tissues.74 With hyperglycemia, there is a disorganiza- visible by OCT and not apparent clinically or on angiography is
tion of the interendothelial junctions and an increase in a common appearance of subretinal fluid in cases of severe
transcellular endocytosis, with resultant fluid flow through the DME (Fig. 134.2e).96 The source of the fluid has been presumed
capillary walls into the retinal tissue.74,75 Fluid homeostasis of to be due to excessive retinal capillary leakage, but there is some
the retina is poorly understood. However, increased permeability evidence that the retinal pigment epithelium may also have
of the retinal capillaries at some point overwhelms the fluid dysfunctional fluid homeostatic function in diabetes and
reuptake mechanisms and retinal edema occurs. The extent of contribute to this appearance.93,97
retinal vascular leakage into the vitreous cavity can also be
detected in vivo.76
The pathophysiologic mechanisms underlying the breakdown
of the blood–retinal barrier are multifactorial and are continuing
to be understood in the context of the myriad biochemical
pathways affected by hyperglycemia. One of the dominant
emerging themes surrounds the role of the soluble growth factor
VEGF in the diabetic retina. While better known as an
angiogenic stimulus upregulated by hypoxia,77,78 VEGF is a
potent permeability agent which is found in animal diabetic
models and in human diabetic retinas associated with increased
vasopermeability.79–81 Therefore, VEGF inhibition has emerged
as a target molecule for treatment of DME. Another target
molecule for pharmacotherapy is the intracellular signaling
molecule PKCb (protein kinase Cb). Selectively upregulated in
hyperglycemia, this protein appears to mediate in part a variety
of diabetes-induced pathophysiologic events,82 including VEGF
production,83 leukostasis and vascular permeability,83,84 abnormal
blood flow,82 and angiogenesis.85 The generation of reactive
lipoxidative products and glycation end products86 in diabetes
can cause direct cellular damage and also induce inflammation.87
Steroids can inhibit inflammatory responses, stabilize inter- FIGURE 134.1. Photomicrograph of cystoid spaces and subretinal
endothelial junctions, and inhibit VEGF, thereby accounting for fluid in the retina of a diabetic patient with severe DME. H&E, original
interest in this class of compounds for treating DME.88–90 magnification µ100. Image furnished by Daniel Albert MD. 1795
RETINA AND VITREOUS

a b c
SECTION 10

d e

FIGURE 134.2. Right eye of a patient with untreated DME. Visual acuity measured 66 letters (~20/50). (a) Red-free (green channel) image from a
digital fluorescein angiogram of a patient with diffuse DME with cystoid changes. (b) Early phase of the fluorescein angiogram with numerous
microaneurysms and patchy loss of capillaries. (c) Mid-phase of the angiogram demonstrating early diffuse fluorescein dye leakage. (d) Late
phase of the angiogram with fluorescein dye pooling in cystoid spaces and diffuse retinal fluorescein staining. (e) OCT scan demonstrating retinal
thickening and a large central cyst and smaller cystoid spaces in the outer plexiform layer of the retina and retinal elevation from subretinal fluid.
Printed with permission of the Diabetic Retinopathy Clinical Research (DRCR) Network.

One of the most characteristic clinical appearances of DME Chronic severe DME, in addition to retinal atrophy, can lead
is the presence of hard exudates (HE), which are lipoproteinaceous to subretinal fibrosis, particularly if chronic hard exudates and
deposits from the transudation of fluid from retinal capillaries subretinal fluid are present.101 In some cases, subretinal non-
(Fig. 134.4). Abnormal fluid homeostasis in the retina results vascular membranes appear to be stimulated by laser treatment
in a high rate of flux of plasma components in and out of the of macular edema. Often, such membranes are subclinical but
retinal capillaries. Slower absorption of transudated larger may be noted on histopathology in autopsy eyes or clinically by
plasma molecules from the retinal tissues results in a net OCT. If subretinal fibrosis occurs under the fovea, eventual
accumulation, which may eventually form a macroscopically photoreceptor loss and poor vision result. Macular ischemia
visible accretion, the hard exudate. As with cystoid spaces, hard may coexist with DME, which may also yield visual acuity
exudates develop in the outer plexiform layer (Fig. 134.5).98 Not worse than expected based on the appearance of the macula.
unexpectedly, several reports correlate increased quantities of Other confounding abnormalities, that can coexist and confuse
hard exudates in patients with DME who also suffer from the relationship between DME and vision and treatment
dyslipidemia.48 Hard exudates may accumulate in the foveal include, epiretinal membranes and vitreous traction.95,102,103
center, leading to loss of central visual function.99 A large
amount of hard exudates at the center may tend to cause fibrotic CLINICAL PRESENTATION OF DIABETIC
organization which portends poor visual prognosis. MACULAR EDEMA
A paradoxical increase in the presence of HE may occur
temporarily in recently treated eyes with DME. This may be Patients with DME present with a range of visual symptoms
due to increased fluid reuptake in the retinal capillaries, leaving depending on the degree to which the fovea is involved and the
behind a greater proportion of lipoproteins that previously were chronicity of the edema. If the macula center is not involved,
dispersed throughout the edema in the retinal tissue. This effect patients are rarely symptomatic; only a few very observant
has clinical relevance for those eyes that have hard exudates individuals may notice relative paracentral scotomas corresponding
close to, but not yet in, the macular center. Treating the macular to focal edema and hard exudates. Some patients with central
edema could, in such a case, cause increased hard exudate macular involvement have excellent acuity and no visual
accumulation into the center leading to loss of visual acuity.100 complaints, presumably because of only recent involvement of
HE may gradually reabsorb over a period of months if macular the center. Over time, patients experience a gradual progressive
edema is reduced. Eyes with aggregation of hard exudates in the vision loss over weeks to months. Patients may complain of
center of the macula may have some visual recovery at that loss of color vision,104 poor night vision, and washing-out of
1796 point. vision in bright sunlight with poor dark–light adaptation.
Diabetic Macular Edema

a b c

CHAPTER 134
d e

FIGURE 134.3. Same eye from the patient shown in Figure 134.2, one year later, after one modified ETDRS laser treatment was performed at
baseline. Visual acuity improved to 73 letters (20/40) at the 12 month visit. (a) Red-free image demonstrating no visible cystoid spaces.
(b) Early phase of the angiogram with early leakage from neovascularization from the optic nerve head and the inferior temporal arcade vessels.
(c) Mid-phase of the angiogram demonstrating persistent diffuse retinal capillary leakage and retinal staining. (d) Late phase of the angiogram,
showing diffuse leakage, but less than at baseline, with reduction in pooling of dye in cystoid spaces. (e) OCT scan showing resolution of
subretinal fluid, resolution of cystoid spaces, and marked reduction in retinal thickening after laser treatment.
Printed with permission of the DRCR Network.

FIGURE 134.5.
Photomicrograph of
hard exudates in the
outer plexiform layer of
the retina in the retina
of a diabetic patient.
H&E, original
magnification µ100.
Image furnished by
Daniel Albert, MD.
Printed with
permission of the
DRCR Network.

FIGURE 134.4. Color photograph of the left eye of a diabetic patient


with focal macular edema, center-involved, with circinate hard
exudates roughly circumscribing the area of retinal thickening.
Printed with permission of the DRCR Network.

1797
RETINA AND VITREOUS

Metamorphopsia is not uncommon. Frequently, patients with progress. The standardized seven-fields photography protocol,
center-involved DME note fluctuation of vision from day-to-day allow thorough documentation of mid-peripheral and posterior
or even over the course of a day.34 In some cases, the patient retinal lesions and has been a mainstay in clinical trials where
may relate such changes to fluid retention, hyper or hypoglycemia, both DR severity and macular edema are to be analyzed. As
or ambient lighting. Some patients may experience a diurnal many ophthalmology offices transition from, film imaging to
variation in macular edema,105–109 though this appears to be of digital camera systems for a variety of reasons, issues with
low frequency and magnitude and of uncertain clinical relevance. color contrast, display, and stereo viewing in the digital
On fundus examination with slit lamp biomicroscopy or environment bring some challenges that can decrease the
contact lens, retinal thickening may present in some commonly utility of color imaging. However, with adequate quality and
identified clinical patterns. Focal edema often occurs associated display, high resolution color images are comparable to film.110
with a cluster of microaneurysms, sometimes surrounded by an Standardized protocol color imaging (film and digital) is employed
incomplete ring of hard exudates (this pattern is termed extensively for clinical research and is comparable to biomicro-
circinate hard exudates) (Fig. 134.4). Multiple foci of edema scopic clinical examination for the detection and classification
may be present, varying in size, and sometimes merging of retinal thickening.111 However, many clinicians rely
together over time. The macula center may or may not be increasingly on OCT to monitor the status of the macula,
involved. Diffuse DME may be very difficult to identify particularly if the center is involved.
clinically if the retina is of uniform thickness, due to the lack of Fluorescein angiography yields useful information regarding
reference landmarks. Clues include the height of the retinal macular perfusion and the extent of capillary leakage. At this
SECTION 10

blood vessels over the pigment epithelium, loss of the foveal time, angiography is the only common clinical method that can
depression or even cystoid spaces. If the pupil is small or there document retinal capillary ischemia, although good photographic
is lens opacity interfering with the examination, such features quality is required. Areas of fluorescein leakage which may not
are easily missed. Other features sometimes seen with macular correlate with retinal thickening are sometimes noted (Fig. 134.6);
edema include variable loss of retinal transparency, a large burden in contrast, sometimes areas of retinal thickening may not
of microaneurysms and intraretinal hemorrhages, and dispersed demonstrate notable fluorescein leakage. Partly for this reason,
flecks of hard exudate. the ETDRS did not include fluorescein angiographic
Stereoscopic fundus photographs have been obtained historically characteristics in the definition of clinically significant macular
to document the extent of macular edema and monitor edema or criteria for retreatment,2 although angiography was

a b c

d e

FIGURE 134.6. Left eye of a diabetic patient prior to laser treatment. Baseline visual acuity measured 42 letters (20/160). (a) Color fundus
photograph with central edema, hard exudates, and scattered intraretinal hemorrhages and microaneurysms. (b) Early phase angiogram,
scanned from a negative film strip, with early leakage from microaneurysms. (c) Mid-phase of the angiogram showing patching fluorescein
leakage. (d) Late cystoid staining. (e) OCT scan demonstrates cystoid edema. The hyper-reflective dot at the center (arrow) represents hard
1798 exudate in the outer plexiform layer.
Diabetic Macular Edema

performed within the study.100,112 However, angiographic Successful reduction of DME early in the course of the
features were used to guide treatment. The treatable lesions disease is likely to stabilize or improve visual function. The
associated with macular edema include: focal leaks greater than ETDRS demonstrated that laser-treated eyes not only had a
500 mm from the center of the macula believed to be causing significant reduction in the rate of vision loss, but those eyes
retinal thickening or HE; focal leaks 300–500 mm from the were also twice as likely to experience visual acuity improvement.
center of the macula believed to be causing retinal thickening Unfortunately, eyes with long-standing DME that have
or HE if the treating ophthalmologist does not believe that reduction in retinal thickening, may not experience any visual
treatment is likely to destroy the remaining perifoveal capillary improvement with treatment.124
network; areas of diffuse leakage that have not been treated Persons with no or minimal diabetic retinopathy may have
previously; areas of capillary nonperfusion not previously subtle abnormalities of contrast sensitivity and color perception.
treated. In many eyes with significant disease, the location of DME decreases contrast sensitivity roughly in proportion to
the fovea is not distinct, and angiography assists with topographical visual acuity. Threshold retinal sensitivity also decreases, and
localization of the center and identification of microaneurysms color vision is progressively impaired.125,104 Light–dark adap-
for laser treatment. tation is also prolonged.126 These measures confirm the
OCT technology was commercialized for retinal imaging in subjective complaints of many subjects with DME regarding
1995 and quickly penetrated ophthalmology offices because of loss of night vision, dimness of vision, and impairment of color
the advantageous and unique display of retinal topography and perception. On multifocal electroretinogram testing, reduced
cross sectional anatomy.113 Third-generation OCT machines amplitudes and latencies can be found, again roughly parallel to

CHAPTER 134
display mean retinal thickness measurements in nine subfields the extent of acuity loss.127,128 The oscillatory potentials measured
that approximate (but are slightly smaller than) the macular from standard bright flash electroretinogram protocols may also
grid used for measurement and grading in the ETDRS. The be more sensitive than visual acuity as a measure of early
center point thickness and total macular volume within the grid impairment from diabetic retinal vasculopathy.126,34 Carefully
are provided along with a false-color topographical map. The microperimetric analyses of diabetic maculae correlated with
reproducibility of good-quality scans is very high and allows fluorescein angiograms demonstrate relative scotomas and
detection of moderate change in center retinal thickness over other abnormalities.129,130
time.114–116 However, quality issues in scan acquisition, media
opacity, unstable fixation, and indistinct tissue planes causing
measurement algorithm errors are relatively common and
LASER PHOTOCOAGULATION FOR DIABETIC
require interpretation.117 Nevertheless, the ability to measure MACULAR EDEMA
retinal thickness has not been available with other modalities The ETDRS conclusively demonstrated that focal/grid laser
and has been an important advance. Detection of subtle photocoagulation was safe and effective in reducing vision loss
thickening at the center is more sensitive by OCT scans than due to DME (Figs 134.2, 134.3, 134.6, and 134.7). In eyes with
clinical examination.118,119 Cystoid spaces, subretinal fluid, and center-involved DME, the rate of moderate visual loss (loss of
vitreoretinal abnormalities not visible on clinical examination 15 or more letters on the ETDRS chart) was reduced from 33%
or photography are often imaged on OCT scans.96 Less clear among eyes in the deferral of treatment arm to 13% among
is how robust current OCT technology is for measurement those eyes in the immediate laser treatment group,123 a relative
and tracking of noncenter involved macular edema. DME reduction in the rate by ~60%. At 3 years, in eyes with center-
features on OCT correlate well with classification by fundus involved DME and acuity 20/40 or worse, improvement in
photographs114,120,121 and fluorescein angiography.122 acuity by six or more letters was observed in ~40% of eyes
assigned to immediate treatment, versus ~20% in eyes assigned
to deferral.2 There was a similar two-fold increase in the rate
RELATIONSHIP BETWEEN MACULAR EDEMA
of gain of 15 or more letters, ~15% versus ~7% (ETDRS,
AND VISUAL ACUITY AND OTHER MEASURES unpublished data). The 3 year results were sufficiently
OF VISUAL FUNCTION compelling that the data safety and monitoring committee of
Untreated patients with center-involved DME experience a the ETDRS halted the study before the scheduled end of the
gradual loss of visual acuity. In the ETDRS, the 3-year risk of trial so that subjects in the deferred laser treatment group
moderate vision loss (defined as loss of three lines of acuity on could be offered treatment. While laser treatment increased the
the standardized acuity chart) was ~33% at 3 years in the group probability that a patient might have visual improvement, the
with CSME.2 However, the rate of moderate vision loss for eyes number of patients actually manifesting substantial improve-
with clinically significant edema without center involvement ment was disappointingly small. In addition, the mean acuity of
was close to 20%, and untreated eyes with DME that was less treated patients in the ETDRS had a progressive decline in
than clinically significant had a rate of ~15%.123 Unless vision over time despite treatment. The advent of laser photo-
ischemia, traction, or other confounding macular pathology is coagulation for DME was a great contribution for management
present, DME does not cause visual acuity loss until the center of an otherwise untreatable condition, and laser treatment has
of the macula is involved. Even then, vision loss tends to be saved useful vision in countless individuals. However, the
gradual, and it is not unusual to observe patients with obvious burden of vision loss among patients with DME remains high,
involvement of the foveal region by DME, yet have excellent thus spurring the investigation of pharmacologic treatments
acuity and no symptoms.123 Conversely, as noted previously, that might be adjunctive to laser treatment, salvage therapy for
patients with chronic edema may eventually undergo retinal those patients who continue to lose vision after laser, or
disorganization and atrophy; such maculae may have poor potential new primary therapies.
acuity yet retinal thickness within the normal range. Therefore, The technique of ETDRS focal/grid photocoagulation has
a cross sectional assessment of a population with DME may been used as standard therapy in the community since the
display only a fair relationship between thickness and acuity, study results were disseminated, with little modification over
because it includes patients with a range of disease duration. the years.123 The original study employed argon blue–green
OCT assessment of retinal thickness is therefore a limited laser with laser spot sizes up to 100 mm in diameter. Newer
predictor of vision in a cross-sectional sample of persons with lasers routinely allow spot sizes of 50 mm in diameter, which is
DME. preferable to avoid complications from laser scar expansion. 1799
RETINA AND VITREOUS

a b c
SECTION 10

d e

FIGURE 134.7. Same eye shown in Figure 134.6 at one year post baseline, after a total of 3 modified ETDRS focal/grid photocoagulation
treatments at baseline, 3.5 months and 8 months. Visual acuity improved to 65 letters (20/50) at 12 months. (a) Color photograph demonstrating
reduction in hard exudates and visible microaneurysms. (b) Early phase fluorescein angiogram showing hyperfluorescent pigment epithelial
defects from laser burns (arrow). (c) Mid-phase angiogram showing reduced leakage. (d) Late phase angiogram with only mild fluorescein
leakage and no cystoid edema. (e) OCT scan from the 12 month visit demonstrating no significant retinal thickening, and resolution of
cystoid spaces.
Printed with permission of the DRCR Network.

a b c

FIGURE 134.8. Same eye shown in Figure 134.6, immediately after the first laser application (a), and immediately following the second laser
application (b), at 3.5 months after the first. Pale gray laser spots can be observed in both photographs (arrows). (c) OCT scan of the same eye
at 8 months, demonstrating reduction in retinal edema, but persistent cystoid changes and hard exudate.
Printed with permission of the DRCR Network.

The blue argon wavelengths have been filtered out from modern fovea and grid laser treatment to areas of diffuse retinal thickening.
argon lasers, since, the luteal pigments in the inner retinal In addition, areas of ischemia identified on fluorescein angio-
layers absorb blue light and yield unnecessary thermal damage graphy could be treated with grid laser. Repeat laser treatment
to the retinal nerve fiber layer. In addition, solid state lasers are may be applied if clinically significant DME persists. In the
available to provide treatment with green wavelengths. The ETDRS, leaking microaneurysms up to 300 mm from the fovea
intensity of application tends to be slightly less among retinal could be treated if additional treatment was needed at sub-
surgeons today, again out of concern for laser scar expansion sequent visits. Laser burns this close to the fovea increase the
(Fig. 134.8). The ETDRS protocol recommends focal treatment risk of scotomas, accidental fovea burns, and late damage to
1800 to leaking microaneurysms that are at least 500 mm from the the fovea from laser scar enlargement and are rarely used today.
Diabetic Macular Edema

reading center interpretation of stereoscopic color photographs


Tips: Modified ETDRS Focal/Grid Laser
for statistical analysis. Patients with edema detectable only on
Photocoagulation of DME
OCT fall into a category that is unstudied in regard to risk of
• Do not treat lesions closer than 500 mm from the fovea on the
vision loss and requirement for treatment. The DRCR.net is
first treatment
currently enrolling subjects with subclinical DME, detected on
• Avoid intense laser burns/scars – endpoint of light gray burns
OCT but not on clinical examination, into a protocol for a
• Avoid excessive density – space burns at least one burn width
longitudinal cohort study (www.DRCRnet.org).
apart
• Do not treat intraretinal hemorrhage or over preretinal
hemorrhage Complications: Potential Complications of Laser
• Consider treat large (>40 mm) microaneurysms that appear to Photocoagulation of DME
be principal causes of leakage focally to the ETDRS endpoint • Fovea burns
of color change (either whitening or darkening) • Subretinal hemorrhage
• Apply grid treatment over areas of diffuse edema • Vitreous hemorrhage
• Optionally grid areas of nonperfusion seen on angiography • Laser scar expansion over time
• Paracentral scotomas
• Subretinal fibrosis
Adverse effects of macular laser treatment according to the • Choroidal neovascularization
ETDRS protocol are remarkable few. The most feared

CHAPTER 134
• Exacerbation of ischemia
complication is accidental foveal photocoagulation, which may • Migration of hard exudates into the fovea
occur from inopportune ocular movement or from surgeon
disorientation and error.131 This may produce immediate and
permanent visual acuity loss and a central scotoma. Closure of
microaneurysms in a very ischemic macula may contribute to VITRECTOMY SURGERY
worsening of ischemia.123 Very severe laser burns, particularly if A subset of patients with DME has coexistent epiretinal
they are small and well focused, may rupture Bruch’s membrane membranes and/or partial posterior vitreous detachment with
and cause subretinal and vitreous bleeding. Late complications retinal traction. These patients may benefit from pars plana
from severe laser scars include choroidal neovascularization. vitrectomy to address the mechanical issues contributing to
Subretinal fibrosis may occur associated with laser scars, the retinal edema.139,102,140 Even without obvious retinal
perhaps more commonly in patients with very severe DME.132 traction, some clinicians believe that many cases of DME
Very severe laser burns, particularly if applied over hemorrhage, respond to removal of the vitreous, with or without removal of
may also cause epiretinal proliferation with retinal traction. the internal limiting membrane.141 The rationale for this
Finally, a common observation is that laser scars tend to enlarge approach is that there may be some subclinical traction from a
over time due to a phenomenon termed ‘RPE creep’. The retinal taut posterior vitreous face, or that the intact vitreous serves as
pigment epithelium (RPE) at the margins of macular laser scars a reservoir for inflammatory macromolecules which promote
tends to atrophy over time, causing progressively enlarging edema.81,142,143,144 No large randomized trials have evaluated
regions of RPE that may become confluent or may erode under this treatment to date.
the fovea, causing photoreceptor dysfunction and vision loss.1,100
Because focal/grid macular laser treatment is not entirely
without risk, the ETDRS noted that for eyes with clinically TRIAMCINOLONE ACETONIDE
significant macular edema without center involvement, The rationale for use of steroids to treat DME is compelling.88,145
observation for a period is acceptable management.100 In As noted previously, the molecular biology of diabetic vascular
particular, if a patient has fluid retention, management of the change includes leukostasis and other mechanisms of
systemic issues may occasionally benefit the patients DME and inflammation, endothelial decompensation, and increased
forestall the need for laser treatment.34 levels of pro-inflammatory cytokines. Steroids may be useful to
Alternatives to ETDRS-style focal/grid macular photo- treat DME because of antiinflammatory effects which include
coagulation have been evaluated in numerous studies. The decreased inflammatory cell activation and adhesion, and growth
Diabetic Retinopathy Clinical Research Network (DRCR.net) factor signaling. Steroids also have a direct effect on the maturation
has evaluated a diffuse grid style of very mild burns compared of interendothelial cell junctions and improved barrier properties.
to standard of care focal/grid treatment, and found no advantage Intravitreous injection of triamcinolone acetonide often has
to the former (DRCR Network Research Group, manuscript a marked beneficial effect on retinal thickening in DME and
submitted). Subthreshold burns with diode laser133 or green thus has sparked considerable interest and clinical use.
laser134,135 have been employed to reduce the risk of perifoveal Numerous small studies have demonstrated robust anatomic
scotoma and laser scarring. results, but often the visual results do not correlate well with
Complicating any discussion of laser treatments alternative reduction in retinal thickening,124,146,147 probably because of the
to the ETDRS method is the fact that the underlying cause for aforementioned complicating factors for lack of vision recovery
the treatment effect of laser is not well understood. Some with chronic DME. The duration of drug in the vitreous cavity
evidence for increased oxygen tension after laser suggests an approximates the duration of clinical effect (2–3 months for a
effect in downregulating VEGF production.136 Pigment epithelium 4.0 mg injection of triamcinolone).147–151 Repeated injections
derived growth factor (PEDF) is a candidate inhibitor of vascular may be needed to maintain clinical benefit. The complications
permeability that has VEGF antagonist and antiinflammatory of triamcinolone acetonide injection are well documented and
properties,137 and is upregulated in RPE cells by laser treatment.138 include endophthalmitis, elevated intraocular pressure
OCT is more sensitive than clinical examination for requiring topical therapy or even surgical intervention to
identifying subtle thickening of the foveal region.118,119 This prevent vision loss,152–155 and cataract.156,157
raises the question of what threshold of retinal thickening is A large randomized clinical trial of serial intravitreal
of sufficient risk to merit laser or any other treatment. The injections of triamcinolone has completed enrollment within
findings of the ETDRS are based upon clinician-determined the DRCR.net, with results not yet available at this time. While
identification of retinal thickening to guide treatment and short-term effects of intravitreal triamcinolone are encouraging, 1801
RETINA AND VITREOUS

a longer-term trial is essential to determining whether the specific PKCb inhibitor, ruboxistaurin (Arxxant: Eli Lilly),
known risks of intravitreal injection of steroids are outweighed decreased abnormal vascular permeability and blood flow,82,83
by the ultimate clinical benefits. and also inhibited angiogenesis in a nondiabetic retinal ischemia
Because the risks of intravitreal injection are relatively high, model.85
there is high-clinical interest in evaluating the potential benefits Phase III clinical trials of oral administration of ruboxistaurin
of peribulbar injection of high-dose triamcinolone acetonide. have demonstrated efficacy in the prevention of sustained
The risks of cataract and intraocular pressure elevation are moderate vision loss in diabetic patients with nonproliferative
somewhat less from peribulbar injection, but with repeated DR.3 In a trial designed to test the effects of the study drug on
injections there may be ptosis, orbital fat atrophy and progression of diabetic retinopathy, it was found that
enophthalmos, and diplopia from extraocular muscle damage. ruboxistaurin reduced the rate of sustained moderate vision loss
A 40 mg dose of triamcinolone in the orbit may produce (loss of 15 or more letters for at least two consecutive study
detectable serum levels and affect glycemic control in diabetic visits) in eyes with definite diabetic macular edema at baseline
patients. Small studies have suggested benefit in the treatment (10% 32 mg/day study drug versus 25% placebo, P = 0.017). A
of DME, although there is considerable variability in intraocular separate study evaluated the ruboxistaurin specifically in regard
penetration from peribulbar injection, and clearly intraocular to the reduction of progression of retinal thickening from more
drug levels are lower than with direct intraocular admini- than 300 m to within 100 m of the center as measured in
stration.67,158 At this time, both peribulbar and intravitreous stereoscopic fundus photographs. The 32 mg/day dose reduced
triamcinolone steroids are widely employed clinically in the progression of DME compared to placebo (hazard ratio = 0.66
SECTION 10

management of DME, but without a sufficient evidence base to (95% CI 0.47–0.93P = 0.016) (PKC-DMES Study Group, manu-
merit recommendation as standard therapy. script submitted). This product is currently under regulatory
review for the treatment of early diabetic macular edema.
ANTI-VEGF THERAPY
Complications of local drug injection
Given the previous discussion of the importance of VEGF in • Intraocular steroids
vascular permeability and its upregulation in diabetic retino- • Endophthalmitis (rare) or sterile pseudoendophthalmitis
pathy, the rationale for use of anti-VEGF drugs is clear. Current (rare)
specific anti-VEGF therapy is given intravitreal at frequent • Intraocular pressure elevation (common, about 25%)
intervals, which may temporarily blunt the effects of VEGF and • Cataract (moderate frequency)
lessen macular edema. Unknown is whether treatment will • Needle damage (retinal tears, lens damage, vitreous
alter the underlying disease process (although it may cause at hemorrhage)(rare)
least temporary regression of neovascularization).159,160 There- • Injection site pain and bleeding (mild)
fore, as the drug effect wears off, the underlying production of • Periocular Steroids
VEGF appears to continue unabated and it is likely that long • Accidental globe penetration (rare)
term intravitreal therapy is required for management.161 • Orbital fat atrophy – enophthalmos (rare)
Results from a phase II clinical trial of intravitreal injection • Diplopia (rare)
of pegaptanib (Macugen: Eyetech OSI) over 36 weeks • Ptosis (rare)
demonstrated beneficial effects,162 and a phase III trial is in • Foreign body encapsulation (rare)
progress at this time. Median VA was better at week 36 with • Cataract (rare)
0.3 mg/0.05 ml pegaptanib (20/50), as compared with sham • Intraocular pressure elevation (common, not as common
injection (20/63) (P = 0.04), and a larger proportion of patients as intraocular)
receiving pegaptanib had > or =15 letters of visual gain (18% • Injection site pain and bleeding (mild)
versus. 7%, P = 0.12). Mean central retinal thickness decreased • Anti-VEGF Product Intravitreal Injection
by 68 mm with 0.3 mg, versus an increase of 4 mm with sham • Endophthalmitis (rare)
(P = 0.02). Pegaptanib is an engineered RNA fragment bearing • Injection site pain and bleeding (mild)
specific binding sites for the VEGF 165 isoform. It is currently • Needle damage to globe (tears, cataract, vitreous
administered as an intravitreal injection at 6 week intervals. hemorrhage) (rare)
Early phase clinical trials of ranabizumab (Lucentis: Genentech)
are in progress. Off label use of bevacizumab (Avastin: Genentech)
has been presented in small cohorts of patients with macular SUMMARY AND FUTURE DIRECTIONS
edema from various causes,163,164 but no randomized trials of
DME have been reported. Ranabizumab and bevacizumab are The landscape of DME management is rapidly changing with
fusion proteins with a human antibody backbone and bind all the advent of research advances leading to better understanding
VEGF subtypes. Both of these drugs are given as intravitreal of pathophysiologic mechanisms and discovery of potential
injections, but the optimal delivery schedule and dosing level therapeutic compounds. There are several challenges that remain
have not been elucidated. in bringing forth new treatments with an adequate evidence
base to guide clinicians in timely patient care.
1. Diabetic retinopathy is a complicated disease. It is clear
INHIBITION OF PROTEIN KINASE C that a number of altered cellular mechanisms work in
PKC is a family of intracellular signaling peptides that act as concert to bring about clinical complications. It is unclear
intermediaries for growth factor and integrin signaling between how impacting only one or a few of these pathways will
ligand binding and nuclear transcription. Broad inhibition of result in effective treatment, and it may be that a
PKC is highly toxic, however specific isoforms are upregulated combination approach will be necessary for management of
in response to particular stimuli. The PKCb isoform is specifically some cases.
upregulated in hyperglycemia in multiple tissues, including 2. Vascular leakage from hyperglycemia can be produced in
vascular endothelial cells, and therefore is hypothesized to animal models, but there is no robust animal model of
mediate some of the myriad biochemical disturbances induced DME, which limits the preclinical testing of potential new
1802 by chronic hyperglycemia.165 In animal models of diabetes, a treatments before clinical development.
Diabetic Macular Edema

3. Diabetic retinopathy is a chronic disease: onset is gradual Despite these caveats, research continues intensively
over years, and treatment may be required indefinitely. because the public health burden of DME and other
Consequently, clinical trials are large and lengthy, creating diabetic compli-cations is so great. This has led to the
huge financial burdens in bringing potential new current era of an array of treatments of uncertain benefit
treatments to market, slowing their introduction and and risk, and lack of clinician certainty as to how and
adding to the costs, thereby creating barriers for patient care. when to apply what treatments and in what combination,
4. Visual acuity is an imprecise and subjective measure of visual if any. It will take years to sort out these issues, and in the
function, although it is highly relevant and clearly the best meantime, new treatments will be introduced which may
test available. It correlates only modestly with advanced further complicate the decision making process for patient
DME, not at all with early (not center involved) DME. and physician. Nevertheless, this is clearly a superior
5. The OCT has become the standard method for assessing situation over the converse, having no promising
the status of the central macula in DME, but current treatments to offer. While clinical trials continue to work
technology is of uncertain sensitivity for detection or toward testing new approaches, the results of the ETDRS
monitoring of edema that is not center-involved (for both remains a solid foundation for clinicians to refer to when
patient care and in clinical trials). Retinal thickness faced with a difficult treatment decision.
measurement (on exam, in photos, or by OCT) correlates
only moderately with visual acuity, but remains the most The results of the DCCT/EDIC and UKPDS trials demonstrate
important variable for disease presence and progression. that DME and other complications can be prevented or delayed

CHAPTER 134
through excellent metabolic management, and this should
There is, therefore, room for improvement in functional continue to be a major focus of conversation between physician
and anatomic assessments of DME. Validation of other, and patient at any phase of the disease. Early disease detection
early functional assessments with disease outcome may and treatment are the next greatest priority, and all health care
shorten the drug development timeline and result in providers need to work in concert to educate, screen and monitor
increased efficiency of clinical research. patients with diabetes and those at high risk for diabetes.

REFERENCES
1. Early Treatment Diabetic Retinopathy Study 10. Lecaire T, Palta M, Zhang H et al. Lower- type 1 diabetes mellitus. JAMA. 2002;
Research Group: Treatment techniques and than-Expected Prevalence and Severity of 287:2563–2569.
clinical guidelines for photocoagulation of Retinopathy in an Incident Cohort followed 19. UK Prospective Diabetes Study (UKPDS)
diabetic macular edema. Early treatment during the First 4–14 Years of Type 1 Group. Effect of intensive blood-glucose
diabetic retinopathy study report number 2. Diabetes. Am. J. Epidemiol. 2006. control with metformin on complications in
Ophthalmology 1987; 94:761–774. 11. Cugati S, Kifley A, Mitchell P, Wang JJ. overweight patients with type 2 diabetes
2. Early Treatment Diabetic Retinopathy Study Temporal trends in the age-specific (UKPDS 34). Lancet. 1998; 352:854–865.
Research Group. Photocoagulation for prevalence of diabetes and diabetic 20. Klein R, Klein BE, Moss SE et al.
diabetic macular edema. Early Treatment retinopathy in older persons: Population- Glycosylated hemoglobin predicts the
Diabetic Retinopathy Study report number based survey findings. Diabetes Res. Clin. incidence and progression of diabetic
1. Arch. Ophthalmol. 1985; Pract. 2006. retinopathy. JAMA. 1988; 260:2864–2871.
103:1796–1806. 12. Klein R, Klein BE, Moss SE, Cruickshanks 21. The absence of a glycemic threshold for
3. PKC-DRS Study Group. The effect of KJ. The Wisconsin Epidemiologic Study of the development of long-term
ruboxistaurin on visual loss in patients with Diabetic Retinopathy. XV. The long-term complications: the perspective of the
moderately severe to very severe incidence of macular edema. Diabetes Control and Complications Trial.
nonproliferative diabetic retinopathy: initial Ophthalmology. 1995; 102:7–16. Diabetes. 1996; 45:1289–1298.
results of the Protein Kinase C beta 13. Early Treatment Diabetic Retinopathy Study 22. Zander E, Herfurth S, Bohl B et al.
Inhibitor Diabetic Retinopathy Study Research Group. Case reports to Maculopathy in patients with diabetes
(PKC-DRS) multicenter randomized clinical accompany Early Treatment Diabetic mellitus type 1 and type 2: associations
trial. Diabetes. 2005; 54:2188–2197. Retinopathy Study Reports 3 and 4. Int. with risk factors. Br. J. Ophthalmol. 2000;
4. Klein R, Klein BE, Moss SE et al. The Ophthalmol. Clin. 1987; 27:273–333. 84:871–876.
Wisconsin epidemiologic study of diabetic 14. Early Treatment Diabetic Retinopathy Study 23. Klein R, Klein BE. Relation of glycemic
retinopathy. IV. Diabetic macular edema. Research Group. Grading diabetic control to diabetic complications and
Ophthalmology. 1984;91:1464–1474 retinopathy from stereoscopic color fundus health outcomes. Diabetes Care. 1998;
5. Klein R, Klein BE, Moss SE, Linton KL. The photographs—an extension of the modified 21:C39–43.
Beaver Dam Eye Study. Retinopathy in Airlie House classification. ETDRS report 24. Moss SE, Klein R, Klein BE. The 14–year
adults with newly discovered and number 10. Ophthalmology. 1991; incidence of visual loss in a diabetic
previously diagnosed diabetes mellitus. 98:786–806. population. Ophthalmology. 1998;
Ophthalmology. 1992; 99:58–62. 15. Group. ETDRSR. Fundus photographic risk 105:998–1003.
6. Wong TY, Klein R, Islam FM et al. Diabetic factors for progression of diabetic 25. Balme M, McAllister I, Davis WA, Davis TM.
retinopathy in a multi-ethnic cohort in the retinopathy. ETDRS report number 12. Retinopathy in latent autoimmune diabetes
United States. Am. J. Ophthalmol. 2006; Ophthalmology. 1991; 98:823–833. of adults: the Fremantle Diabetes Study.
141:446–455. 16. The effect of intensive treatment of Diabet. Med. 2002; 19:602–605.
7. Roy MS, Klein R, O’Colmain BJ et al. The diabetes on the development and 26. Wong TY, Mohamed Q, Klein R, Couper DJ.
prevalence of diabetic retinopathy among progression of long-term complications in Do retinopathy signs in non-diabetic
adult type 1 diabetic persons in the United insulin-dependent diabetes mellitus. N. individuals predict the subsequent risk of
States. Arch. Ophthalmol. 2004; Engl. J. Med. 1993; 329:977–986. diabetes? Br. J. Ophthalmol. 2006;
122:546–551. 17. Retinopathy and nephropathy in patients 90:301–303.
8. Kempen JH, O’Colmain BJ, Leske MC et with type 1 diabetes four years after a trial 27. Kohner EM, Aldington SJ, Stratton IM et al.
al. The prevalence of diabetic retinopathy of intensive therapy. . N. Engl. J. Med. United Kingdom Prospective Diabetes
among adults in the United States. Arch. 2000; 342:381–389. Study, 30: diabetic retinopathy at diagnosis
Ophthalmol. 2004; 122:552–563 18. Writing Team for the Diabetes Control and of non-insulin-dependent diabetes mellitus
9. Varma R, Torres M, Pena F et al. Complications Trial/Epidemiology of and associated risk factors. Arch.
Prevalence of diabetic retinopathy in adult Diabetes Interventions and Complications Ophthalmol. 1998; 116:297–303.
Latinos: the Los Angeles Latino eye study. Research Group. Effect of intensive therapy 28. Newcomb PA, Klein R, Massoth KM.
Ophthalmology. 2004; 111:1298–1306. on the microvascular complications of Education to increase ophthalmologic care 1803
RETINA AND VITREOUS

in older onset diabetes patients: indications retinopathy. Diabetes Care. 1996; 59. Hellstedt T, Kaaja R, Teramo K, Immonen I.
from the Wisconsin Epidemiologic Study of 19:1291–1293. The effect of pregnancy on mild diabetic
Diabetic Retinopathy. J. Diabetes 45. Klein R, Klein BE, Moss SE, Surawicz TS. retinopathy. Graefes Arch. Clin. Exp.
Complications. 1992; 6:211–217. The Wisconsin Epidemiologic Study of Ophthalmol. 1997; 235:437–441.
29. Klein R. Prevention of visual loss from Diabetic Retinopathy. XIII. Relationship of 60. Davis MD. Worsening of diabetic
diabetic retinopathy. Surv. Ophthalmol. serum cholesterol to retinopathy and hard retinopathy after improvement of glycemic
2002; 47 Suppl 2:S246–252. exudate. Ophthalmology. 1991; control. Arch. Ophthalmol. 1998;
30. Harris MI, Klein R, Welborn TA, Knuiman 98:1261–1265. 116:931–932.
MW. Onset of NIDDM occurs at least 4–7 46. Klein R, Sharrett AR, Klein BE et al. The 61. Patel JI, Hykin PG, Cree IA. Diabetic
yr before clinical diagnosis. Diabetes Care. association of atherosclerosis, vascular risk cataract removal: postoperative
1992; 15:815–819. factors, and retinopathy in adults with progression of maculopathy—growth factor
31. Klein R, Klein BE, Moss SE, Cruickshanks diabetes : the atherosclerosis risk in and clinical analysis. Br. J. Ophthalmol.
KJ. The Wisconsin Epidemiologic Study of communities study. Ophthalmology. 2002; 2006; 90:697–701.
Diabetic Retinopathy: XVII. The 14–year 109:1225–1234. 62. Krepler K, Biowski R, Schrey S et al.
incidence and progression of diabetic 47. Davis MD, Fisher MR, Gangnon RE et al. Cataract surgery in patients with diabetic
retinopathy and associated risk factors in Risk factors for high-risk proliferative retinopathy: visual outcome, progression of
type 1 diabetes. Ophthalmology. 1988; diabetic retinopathy and severe visual loss: diabetic retinopathy, and incidence of
105:1801–1815. Early Treatment Diabetic Retinopathy Study diabetic macular oedema. Graefes Arch.
32. Effect of pregnancy on microvascular Report #18. Invest. Ophthalmol. Vis. Sci. Clin. Exp. Ophthalmol. 2002; 240:735–738.
complications in the diabetes control and 1998; 39:233–252. 63. Squirrell D, Bhola R, Bush J et al. A
complications trial. The Diabetes Control 48. Chew EY, Klein ML, Ferris FLr et al. prospective, case controlled study of the
SECTION 10

and Complications Trial Research Group. Association of elevated serum lipid levels natural history of diabetic retinopathy and
Diabetes Care. 2000; 23:1084–1091. with retinal hard exudate in diabetic maculopathy after uncomplicated
33. Matthews DR, Stratton IM, Aldington SJ et retinopathy. Early Treatment Diabetic phacoemulsification cataract surgery in
al. Risks of progression of retinopathy and Retinopathy Study (ETDRS) Report 22. patients with type 2 diabetes.
vision loss related to tight blood pressure Arch. Ophthalmol. 1996; 114:1079–1084. Br. J. Ophthalmol. 2002; 86:565–571.
control in type 2 diabetes mellitus: UKPDS 49. Jenkins AJ, Lyons TJ, Zheng D et al. 64. Menchini U, Cappelli S, Virgili G. Cataract
69. Arch. Ophthalmol. 2004; 122:1631–1640. Serum lipoproteins in the diabetes control surgery and diabetic retinopathy. Semin.
34. Bresnick GH. Diabetic macular edema. A and complications trial/epidemiology of Ophthalmol. 2003; 18:103–108.
review. Ophthalmology. 1986; 93:989–997. diabetes intervention and complications 65. Ferris FLr, Podgor MJ, Davis MD. Macular
35. Sjolie AK, Porta M, Parving HH et al. The cohort: associations with gender and edema in Diabetic Retinopathy Study
DIabetic REtinopathy Candesartan Trials glycemia. Diabetes Care. 2003; patients. Diabetic Retinopathy Study
(DIRECT) Programme: baseline 26:810–818. Report Number 12. Ophthalmology. 1987;
characteristics. J Renin Angiotensin 50. Klein RL, McHenry MB, Lok KH et al. 94:754–760.
Aldosterone Syst. 2005; 6:25–32. Apolipoprotein C-III protein concentrations 66. Shimura M, Yasuda K, Nakazawa T et al.
36. Klein R, Zinman B, Gardiner R et al. The and gene polymorphisms in Type 1 Quantifying alterations of macular thickness
relationship of diabetic retinopathy to diabetes: associations with microvascular before and after panretinal
preclinical diabetic glomerulopathy lesions disease complications in the DCCT/EDIC photocoagulation in patients with severe
in type 1 diabetic patients: the cohort. J. Diabetes Complications. 2005; diabetic retinopathy and good vision.
Renin-Angiotensin System Study. Diabetes. 19:18–25. Ophthalmology. 2003; 110:2386–2394.
2005; 54:527–533. 51. Lyons TJ, Jenkins AJ, Zheng D et al. 67. Wilson CA, Berkowitz BA, Sato Y et al.
37. Agardh CD, Agardh E, Torffvit O. The Diabetic retinopathy and serum lipoprotein Treatment with intravitreal steroid reduces
association between retinopathy, subclasses in the DCCT/EDIC cohort. blood-retinal barrier breakdown due to
nephropathy, cardiovascular disease and Invest. Ophthalmol. Vis. Sci. 2004; retinal photocoagulation. Arch. Ophthalmol.
long-term metabolic control in type 1 45:910–918. 1992; 110:1155–1159.
diabetes mellitus: a 5 year follow-up study 52. Klein BE, Moss SE, Klein R. Effect of 68. Zein WM, Noureddin BN, Jurdi FA et al.
of 442 adult patients in routine care. pregnancy on progression of diabetic Panretinal photocoagulation and intravitreal
Diabetes Res. Clin. Pract. 1997; retinopathy. Diabetes Care. 1990; triamcinolone acetonide for the
35:113–121. 13:34–40. management of proliferative diabetic
38. Bresnick GH. Diabetic maculopathy. A 53. Temple RC, Aldridge VA, Sampson MJ et retinopathy with macular edema. Retina.
critical review highlighting diffuse macular al. Impact of pregnancy on the progression 2006; 26:137–142.
edema. Ophthalmology. 1983; of diabetic retinopathy in Type 1 diabetes. 69. Cogan DG, Kuwabara T. The mural cell in
90:1301–1317. Diabet. Med. 2001; 18:573–577. perspective. Arch. Ophthalmol. 1967;
39. Tokuyama T, Ikeda T, Sato K. Effects of 54. Lovestam-Adrian M, Agardh CD, Aberg A, 78:133–139.
haemodialysis on diabetic macular leakage. Agardh E. Pre-eclampsia is a potent risk 70. Kuwabara T, Cogan DG. Retinal vascular
Br. J. Ophthalmol. 2000; 84:1397–1400. factor for deterioration of retinopathy patterns. VII. Acellular change. Invest.
40. Watanabe Y, Yuzawa Y, Mizumoto D et al. during pregnancy in Type 1 diabetic Ophthalmol. 1965; 4:1049–1064.
Long-term follow-up study of 268 diabetic patients. Diabet. Med. 1997; 71. King GL, Shiba T, Oliver J et al. Cellular
patients undergoing haemodialysis, with 14:1059–1065. and molecular abnormalities in the vascular
special attention to visual acuity and 55. Axer-Siegel R, Hod M, Fink-Cohen S et al. endothelium of diabetes mellitus. Annu.
heterogeneity. Nephrol. Dial. Transplant. Diabetic retinopathy during pregnancy. Rev. Med. 1994; 45:179–188.
1993; 8:725–734. Ophthalmology. 1996; 103:1815–1819. 72. Gardner TW, Antonetti DA, Barber AJ et al.
41. Roy MS, Klein R. Macular edema and 56. Sinclair SH, Nesler C, Foxman B et al. Diabetic retinopathy: more than meets the
retinal hard exudates in African Americans Macular edema and pregnancy in eye. Surv. Ophthalmol. 2002; 47 Suppl
with type 1 diabetes: the New Jersey 725. insulin-dependent diabetes. Am. J. 2:S253–262.
Arch. Ophthalmol. 2001; 119:251–259. Ophthalmol. 1984; 97:154–167. 73. Feit-Leichman RA, Kinouchi R, Takeda M et
42. Miljanovic B, Glynn RJ, Nathan DM et al. A 57. Chew EY, Mills JL, Metzger BE et al. al. Vascular damage in a mouse model of
prospective study of serum lipids and risk Metabolic control and progression of diabetic retinopathy: relation to neuronal
of diabetic macular edema in type 1 retinopathy. The Diabetes in Early and glial changes. Invest. Ophthalmol. Vis.
diabetes. Diabetes. 2004; 53:2883–2892. Pregnancy Study. National Institute of Child Sci. 2005; 46:4281–4287.
43. Effects of intensive diabetes therapy on Health and Human Development Diabetes 74. Harhaj NS, Antonetti DA. Regulation of
neuropsychological function in adults in the in Early Pregnancy Study. Diabetes Care. tight junctions and loss of barrier function
Diabetes Control and Complications Trial. 1995; 18:631–637. in pathophysiology. Int. J. Biochem. Cell
The DCCT Research Group. Ann. Intern. 58. Phelps RL, Sakol P, Metzger BE et al. Biol. 2004; 36:1206–1237.
Med. 1996; 124:379–388. Changes in diabetic retinopathy during 75. Vinores SA, Derevjanik NL, Ozaki H et al.
44. Ferris FL, 3rd, Chew EY, Hoogwerf BJ, pregnancy. Correlations with regulation of Cellular mechanisms of blood-retinal barrier
Early Treatment Diabetic Retinopathy Study hyperglycemia. Arch. Ophthalmol. 1986; dysfunction in macular edema. Doc.
1804 Research Group. Serum lipids and diabetic 104:1806–1810. Ophthalmol. 1999; 97:217–228.
Diabetic Macular Edema

76. Bursell SE, Delori FC, Yoshida A et al. edema. Surv. Ophthalmol. 1984; 28 macular edema. Arch. Ophthalmol. 2006;
Vitreous fluorophotometric evaluation of Suppl:505–511. 124:1701–1707.
diabetics. Invest. Ophthalmol. Vis. Sci. 92. Kincaid MC, Green WR, Fine SL et al. An 110. Fransen SR, Leonard-Martin TC, Feuer WJ,
1984; 25:703–710. ocular clinicopathologic correlative study of Hildebrand PL. Clinical evaluation of
77. Ferrara N. Role of vascular endothelial six patients from the Diabetic Retinopathy patients with diabetic retinopathy: accuracy
growth factor in regulation of physiological Study. Retina. 1983; 3:218–238. of the Inoveon diabetic retinopathy-3DT
angiogenesis. Am J Physiol Cell Physiol. 93. Antcliff RJ, Marshall J. The pathogenesis of system. Ophthalmology. 2002;
2001; 280:C1358–1366. edema in diabetic maculopathy. Semin. 109:595–601.
78. Adamis AP, Shima DT, Tolentino MJ et al. Ophthalmol. 1999; 14:223–232. 111. Kinyoun J, Barton F, Fisher M et al.
Inhibition of vascular endothelial growth 94. Fluorescein angiographic risk factors for Detection of diabetic macular edema.
factor prevents retinal ischemia-associated progression of diabetic retinopathy. ETDRS Ophthalmoscopy versus photography—
iris neovascularization in a nonhuman report number 13. Ophthalmology. 1991; Early Treatment Diabetic Retinopathy Study
primate. Arch. Ophthalmol. 1996; 98:834–840. Report Number 5. Ophthalmology. 1989;
114:66–71. 95. Catier A, Tadayoni R, Paques M et al. 96:746–750, discussion 750–741.
79. Suarez S, Ballmer-Hofer K. VEGF Characterization of macular edema from 112. Classification of diabetic retinopathy from
transiently disrupts gap junctional various etiologies by optical coherence fluorescein angiograms. ETDRS report
communication in endothelial cells. J. Cell tomography. Am. J. Ophthalmol. 2005; number 11. Ophthalmology. 1991;
Sci. 2001; 114:1229–1235. 140:200–206. 98:807–822.
80. Antonetti DA, Barber AJ, Hollinger LA et al. 96. Panozzo G, Parolini B, Gusson E et al. 113. Hee MR, Puliafito CA, Duker JS et al.
Vascular endothelial growth factor induces Diabetic macular edema: an OCT-based Topography of diabetic macular edema

CHAPTER 134
rapid phosphorylation of tight junction classification. Semin. Ophthalmol. 2004; with optical coherence tomography.
proteins occludin and zonula occluden 1. 19:13–20. Ophthalmology. 1998; 105:360–370.
A potential mechanism for vascular 97. Ferris FL, 3rd, Patz A. Macular edema. A 114. Diabetic Retinopathy Clinical Research
permeability in diabetic retinopathy and complication of diabetic retinopathy. Surv. Network. Reproducibility of macular
tumors. J. Biol. Chem. 1999; Ophthalmol. 1984; 28 Suppl:452–461. thickness and volume using Zeiss optical
274:23463–23467. 98. Yanko L, Ungar H, Michaelson IC. The coherence tomography in patients with
81. Patel JI, Tombran-Tink J, Hykin PG et al. exudative lesions in diabetic retinopathy diabetic macular edema. Submitted.
Vitreous and aqueous concentrations of with special regard to the hard exudate. 115. Browning DJ. Interobserver variability in
proangiogenic, antiangiogenic factors and Acta Ophthalmol. (Copenh). 1974; optical coherence tomography for macular
other cytokines in diabetic retinopathy 52:150–160. edema. Am. J. Ophthalmol. 2004;
patients with macular edema: Implications 99. Patz A, Fine SL. Diabetic macular edema. 137:1116–1117.
for structural differences in macular Int. Ophthalmol. Clin. 1976; 16:105–113. 116. Polito A, Del Borrello M, Isola M et al.
profiles. Exp. Eye Res. 2006; 82:798–806. 100. Early Treatment Diabetic Retinopathy Study Repeatability and reproducibility of fast
82. Ishii H, Jirousek MR, Koya D et al. Research Group. Focal photocoagulation macular thickness mapping with stratus
Amelioration of vascular dysfunctions in treatment of diabetic macular edema. optical coherence tomography. Arch.
diabetic rats by an oral PKC beta inhibitor. Relationship of treatment effect to Ophthalmol. 2005; 123:1330–1337.
Science. 1996; 272:728–731. fluorescein angiographic and other retinal 117. Sadda SR, Wu Z, Walsh AC et al. Errors in
83. Aiello LP, Bursell SE, Clermont A et al. characteristics at baseline: ETDRS report retinal thickness measurements obtained
Vascular endothelial growth factor-induced no. 19. Arch. Ophthalmol. 1995; by optical coherence tomography.
retinal permeability is mediated by protein 113:1144–1155. Ophthalmology. 2006; 113:285–293.
kinase C in vivo and suppressed by an 101. Chester EM, Banker BQ. The role of lipid 118. Brown JC, Solomon SD, Bressler SB et al.
orally effective beta-isoform-selective thrombi in the pathogenesis of diabetic Detection of diabetic foveal edema: contact
inhibitor. Diabetes. 1997; 46:1473–1480. retinopathy. Arch. Intern. Med. 1967; lens biomicroscopy compared with optical
84. Miyamoto K, Khosrof S, Bursell SE et al. 120:397–407. coherence tomography. Arch. Ophthalmol.
Prevention of leukostasis and vascular 102. Johnson MW. Tractional cystoid macular 2004; 122:330–335.
leakage in streptozotocin-induced diabetic edema: a subtle variant of the 119. Browning DJ, McOwen MD, Bowen RM,
retinopathy via intercellular adhesion vitreomacular traction syndrome. Am. Jr., O’Marah TL. Comparison of the clinical
molecule-1 inhibition. Proc. Natl. Acad. Sci. J. Ophthalmol. 2005; 140:184–192. diagnosis of diabetic macular edema with
U. S. A. 1999; 96:10836–10841. 103. Gaucher D, Tadayoni R, Erginay A et al. diagnosis by optical coherence
85. Danis RP, Bingaman DP, Jirousek M, Yang Optical coherence tomography assessment tomography. Ophthalmology. 2004;
Y. Inhibition of intraocular of the vitreoretinal relationship in diabetic 111:712–715.
neovascularization caused by retinal macular edema. Am. J. Ophthalmol. 2005; 120. Strom C, Sander B, Larsen N et al. Diabetic
ischemia in pigs by PKCbeta inhibition with 139:807–813. macular edema assessed with optical
LY333531. Invest. Ophthalmol. Vis. Sci. 104. Fong DS, Barton FB, Bresnick GH. coherence tomography and stereo fundus
1998; 39:171–179. Impaired color vision associated with photography. Invest. Ophthalmol. Vis. Sci.
86. Pachydaki SI, Tari SR, Lee SE et al. diabetic retinopathy: Early Treatment 2002; 43:241–245.
Upregulation of RAGE and its ligands in Diabetic Retinopathy Study Report No. 15. 121. DRCR.net. Comparison of the modified
proliferative retinal disease. Exp. Eye Res. Am. J. Ophthalmol. 1999; 128:612–617. Early Treatment Diabetic Retinopathy and
2006; 82:807–815. 105. Frank RN, Schulz L, Abe K, Iezzi R. mild macuilar grid laser strategies for
87. Joussen AM, Poulaki V, Le ML et al. A Temporal variation in diabetic macular diabetic macular edema. Arch Ophthalmol.
central role for inflammation in the edema measured by optical coherence 2007; 114:525–536.
pathogenesis of diabetic retinopathy. tomography. Ophthalmology. 2004; 122. Kang SW, Park CY, Ham DI. The correlation
FASEB J. 2004; 18:1450–1452. 111:211–217. between fluorescein angiographic and
88. Ip MS. Intravitreal injection of 106. Sternberg P, Jr., Fitzke F, Finkelstein D. optical coherence tomographic features in
triamcinolone: an emerging treatment for Cyclic macular edema. Am. J. Ophthalmol. clinically significant diabetic macular
diabetic macular edema. Diabetes Care. 1982; 94:664–669. edema. Am. J. Ophthalmol. 2004;
2004; 27:1794–1797. 107. Larsen M, Wang M, Sander B. Overnight 137:313–322.
89. Martidis A, Duker JS, Greenberg PB et al. thickness variation in diabetic macular 123. Diabetes Control and Complications Trial
Intravitreal triamcinolone for refractory edema. Invest. Ophthalmol. Vis. Sci. 2005; (DCCT): results of feasibility study. The
diabetic macular edema. Ophthalmology. 46:2313–2316. DCCT Research Group. Diabetes Care.
2002; 109:920–927. 108. Polito A, Del Borrello M, Polini G et al. 1987; 10:1–19.
90. Felinski EA, Antonetti DA. Glucocorticoid Diurnal variation in clinically significant 124. Massin P, Audren F, Haouchine B et al.
regulation of endothelial cell tight junction diabetic macular edema measured by the Intravitreal triamcinolone acetonide for
gene expression: novel treatments for Stratus OCT. Retina. 2006; 26:14–20. diabetic diffuse macular edema: preliminary
diabetic retinopathy. Curr. Eye Res. 2005; 109. Danis RP, Glassman AR, Aiello LP et al. results of a prospective controlled trial.
30:949–957. Diurnal variation in retinal thickening Ophthalmology. 2004; 111:218–224.
91. Yanoff M, Fine BS, Brucker AJ, Eagle RC, measurement by optical coherence 125. Abraham FA, Haimovitz J, Berezin M. The
Jr. Pathology of human cystoid macular tomography in center-involved diabetic photopic and scotopic visual thresholds in 1805
RETINA AND VITREOUS

diabetics without diabetic retinopathy. 139. Lewis H, Abrams GW, Blumenkranz MS, 153. Jonas JB, Degenring RF, Kreissig I et al.
Metab. Pediatr. Syst. Ophthalmol. 1988; Campo RV. Vitrectomy for diabetic macular Intraocular pressure elevation after
11:76–77. traction and edema associated with intravitreal triamcinolone acetonide
126. Brinchmann-Hansen O, Dahl-Jorgensen K, posterior hyaloidal traction. Ophthalmology. injection. Ophthalmology. 2005;
Hanssen KF, Sandvik L. Oscillatory 1992; 99:753–759. 112:593–598.
potentials, macular recovery time, and 140. Yanyali A, Nohutcu AF, Horozoglu F, Celik 154. Bakri SJ, Beer PM. The effect of intravitreal
diabetic retinopathy through 3 years of E. Modified grid laser photocoagulation triamcinolone acetonide on intraocular
intensified insulin treatment. versus pars plana vitrectomy with internal pressure. Ophthalmic Surg Lasers Imaging.
Ophthalmology. 1988; 95:1358–1366. limiting membrane removal in diabetic 2003; 34:386–390.
127. Han Y, Adams AJ, Bearse MA, Jr., Schneck macular edema. Am. J. Ophthalmol. 2005; 155. Wingate RJ, Beaumont PE. Intravitreal
ME. Multifocal electroretinogram and short- 139:795–801. triamcinolone and elevated intraocular
wavelength automated perimetry measures 141. Stefansson E, Landers MB, 3rd. How does pressure. Aust. N. Z. J. Ophthalmol. 1999;
in diabetic eyes with little or no retinopathy. vitrectomy affect diabetic macular edema? 27:431–432.
Arch. Ophthalmol. 2004; 122:1809–1815. Am. J. Ophthalmol. 2006; 141:984; author 156. Challa JK, Gillies MC, Penfold PL et al.
128. Bearse MA, Jr., Han Y, Schneck ME, reply 984–985. Exudative macular degeneration and
Adams AJ. Retinal function in normal and 142. La Heij EC, Hendrikse F, Kessels AG, intravitreal triamcinolone: 18 month follow
diabetic eyes mapped with the slow flash Derhaag PJ. Vitrectomy results in diabetic up. Aust. N. Z. J. Ophthalmol. 1998;
multifocal electroretinogram. Invest. macular oedema without evident 26:277–281.
Ophthalmol. Vis. Sci. 2004; 45:296–304. vitreomacular traction. Graefes Arch. Clin. 157. Thompson JT. Cataract formation and
129. Bengtsson B, Heijl A, Agardh E. Visual Exp. Ophthalmol. 2001; 239:264–270. other complications of intravitreal
fields correlate better than visual acuity to 143. Stolba U, Binder S, Gruber D et al. triamcinolone for macular edema. Am.
SECTION 10

severity of diabetic retinopathy. Vitrectomy for persistent diffuse diabetic J. Ophthalmol. 2006; 141:629–637.
Diabetologia. 2005; 48:2494–2500. macular edema. Am. J. Ophthalmol. 2005; 158. Cardillo JA, Melo LA, Jr., Costa RA et al.
130. Agardh E, Stjernquist H, Heijl A, Bengtsson 140:295–301. Comparison of intravitreal versus posterior
B. Visual acuity and perimetry as measures 144. Ouchi M, West K, Crabb JW et al. sub-Tenon’s capsule injection of
of visual function in diabetic macular Proteomic analysis of vitreous from triamcinolone acetonide for diffuse diabetic
oedema. Diabetologia. 2006; 49:200–206. diabetic macular edema. Exp. Eye Res. macular edema. Ophthalmology. 2005;
131. Ishiko S, Ogasawara H, Yoshida A, Hanada 2005; 81:176–182. 112:1557–1563.
K. The use of scanning laser 145. Flynn HWJ, Scott IU. Intravitreal 159. Avery RL. Regression of retinal and iris
ophthalmoscope microperimetry to detect triamcinolone acetonide for macular edema neovascularization after intravitreal
visual impairment caused by macular associated with diabetic retinopathy and bevacizumab (Avastin) treatment. Retina.
photocoagulation. Ophthalmic Surg. venous occlusive disease: it’s time for 2006; 26:352–354.
Lasers. 1998; 29:95–98. clinical trials. Arch. Ophthalmol. 2005; 160. Spaide RF, Fisher YL. Intravitreal
132. Fong DS, Segal PP, Myers F et al. 123:258–259. bevacizumab (Avastin) treatment of
Subretinal fibrosis in diabetic macular 146. Tewari HK, Sony P, Chawla R et al. proliferative diabetic retinopathy
edema. ETDRS report 23. Early Treatment Prospective evaluation of intravitreal complicated by vitreous hemorrhage.
Diabetic Retinopathy Study Research triamcinolone acetonide injection in Retina. 2006; 26:275–278.
Group. Arch. Ophthalmol. 1997; macular edema associated with retinal 161. Rosenfeld PJ, Fung AE, Puliafito CA.
115:873–877. vascular disorders. Eur. J. Ophthalmol. Optical coherence tomography findings
133. Akduman L, Olk RJ. Subthreshold 2005; 15:619–626. after an intravitreal injection of
(invisible) modified grid diode laser 147. Patelli F, Fasolino G, Radice P et al. Time bevacizumab (avastin) for macular edema
photocoagulation in diffuse diabetic course of changes in retinal thickness and from central retinal vein occlusion.
macular edema (DDME). Ophthalmic Surg. visual acuity after intravitreal triamcinolone Ophthalmic Surg Lasers Imaging. 2005;
Lasers. 1999; 30:706–714. acetonide for diffuse diabetic macular 36:336–339.
134. Bandello F, Polito A, Del Borrello M et al. edema with and without previous macular 162. Cunningham ET, Jr., Adamis AP, Altaweel M
“Light” versus “classic” laser treatment for laser treatment. Retina. 2005; 25:840–845. et al. A phase II randomized
clinically significant diabetic macular 148. Schindler RH, Chandler D, Thresher R, double-masked trial of pegaptanib, an
oedema. Br. J. Ophthalmol. 2005; Machemer R. The clearance of intravitreal anti-vascular endothelial growth factor
89:864–870. triamcinolone acetonide. Am. aptamer, for diabetic macular edema.
135. Sinclair SH, Alaniz R, Presti P. Laser J. Ophthalmol. 1982; 93:415–417. Ophthalmology. 2005; 112:1747–1757.
treatment of diabetic macular edema: 149. Tano Y, Chandler D, Machemer R. 163. Iturralde D, Spaide RF, Meyerle CB et al.
comparison of ETDRS-level treatment with Treatment of intraocular proliferation with Intravitreal bevacizumab (Avastin) treatment
threshold-level treatment by using high- intravitreal injection of triamcinolone of macular edema in central retinal vein
contrast discriminant central visual field acetonide. Am. J. Ophthalmol. 1980; occlusion: a short-term study. Retina. 2006;
testing. Semin. Ophthalmol. 1999; 90:810–816. 26:279–284.
14:214–222. 150. Scholes GN, O’Brien WJ, Abrams GW, 164. Mason JO, 3rd, Albert MA, Jr., Vail R.
136. Pournaras CJ. Retinal oxygen distribution. Kubicek MF. Clearance of triamcinolone Intravitreal bevacizumab (Avastin) for
Its role in the physiopathology of from vitreous. Arch. Ophthalmol. 1985; refractory pseudophakic cystoid macular
vasoproliferative microangiopathies. Retina. 103:1567–1569. edema. Retina. 2006; 26:356–357.
1995; 15:332–347. 151. Bakri SJ, Beer PM. Intravitreal 165. Aiello LP. The potential role of PKC beta in
137. Zhang SX, Wang JJ, Gao G et al. Pigment triamcinolone injection for diabetic macular diabetic retinopathy and macular edema.
epithelium-derived factor (PEDF) is an edema: a clinical and fluorescein Surv. Ophthalmol. 2002; 47 Suppl
endogenous antiinflammatory factor. angiographic case series. Can. J. 2:S263–269.
FASEB J. 2006; 20:323–325. Ophthalmol. 2004; 39:755–760.
138. Ogata N, Tombran-Tink J, Jo N et al. 152. Smithen LM, Ober MD, Maranan L, Spaide
Upregulation of pigment epithelium-derived RF. Intravitreal triamcinolone acetonide and
factor after laser photocoagulation. Am. J. intraocular pressure. Am. J. Ophthalmol.
Ophthalmol. 2001; 132:427–429. 2004; 138:740–743.

1806
CHAPTER

135 Proliferative Diabetic Retinopathy


Jennifer K. Sun, Joan W. Miller, and Lloyd P. Aiello

INTRODUCTION AND DEFINITION PATHOGENESIS


With the introduction of insulin in the 1930s, diabetes mellitus Early functional changes have been observed in diabetes,
rapidly ceased to be a fatal disease of young people, with coma including alterations in retinal blood flow4b and a breakdown in
as a cause of death falling from 60–4%.1 The medical care the blood–retinal barrier, which is demonstrated by increased
of diabetic patients instead shifted to the management of vas- leakage from retinal blood vessels measured by vitreous fluoro-
cular complications, including cardiac disease, renal failure, and photometry.5–7 Recent evidence suggests that angiogenic factors
diabetic retinopathy. In the 1970s, it was well demonstrated may play a role in these early changes as well.8,9 Morphologic
that laser photocoagulation could substantially reduce visual changes in nonproliferative diabetic eye disease include micro-
loss and blindness from diabetic retinopathy.2,3 Advances in aneurysms, blot and dot hemorrhages, cotton-wool spots, venous
vitreoretinal surgery have made a further impact on severe caliber abnormalities, intraretinal microvascular abnormalities
proliferative diabetic retinopathy (PDR). Even so, diabetic eye (IRMAs), hard exudates, and macular edema. Microaneurysms
disease remains a severe medical and social problem afflicting appear in clusters and surround areas of capillary closure.
individuals during their productive years. The prevalence of IRMAs are also found adjacent to areas of capillary closure.
diabetes and its associated micro and macrovascular compli- Controversy has existed as to whether IRMAs represented
cations is steadily increasing. Currently, at least 177 million dilated and abnormal retinal capillaries or early intraretinal
people suffer from diabetes worldwide. It is estimated that neovascularization.6 However, recent evidence has demon-
by the year 2030, this number will increase to approximately strated the continuity of intraretinal microvascular changes and
370 million individuals.3a Diabetic retinopathy is responsible for preretinal neovascularization.10 In a retrospective study of serial
at least 12% of the new cases of blindness each year in the United photographs of NVE, Chang and associates found that NVE
States.4 In the United States alone approximately 700 000 persons originated at the same location as an earlier IRMA in 40 of
have PDR. The health care costs of retinopathy-associated dis- 57 locations (70.2%), and the average time span between the
orders exceeds $620 million in the US each year.4a appearance of IRMA and that of NVE was 3 years.11
PDR is characterized by new vessels arising from the surface As more advanced retinopathy develops, retinal neovascu-
of the retina. New vessels may grow from the vessels on the larization occurs. NVE develops from retinal vessels and tends
surface of the optic disk (neovascularization at the disk (NVD)). to appear adjacent to regions of nonperfusion. NVD originates
They may also originate from the retinal vascular arcades else- from vessels on the disk and appears coincident with significant
where (neovascularization elsewhere in the retina (NVE)) and areas of nonperfusion in the midperiphery. The exact patho-
grow along the partially detached posterior hyaloid, also termed genesis of retinal neovascularization remains to be elucidated.
the ‘posterior vitreous face’ (Fig. 135.1). Fibroblasts and glial Retinal neovascularization is not a response limited to diabetes
elements accompany the new vessels, and proliferation of this but is probably a programmed response to retinal ischemia. In
fibroglial tissue can become the predominant feature of the 1948, Michaelson hypothesized that a diffusible factor was
retinopathy. PDR also includes rubeosis iridis or new vessels on released from ischemic retina that stimulated the development
the surface of the iris or in the anterior chamber angle. of embryonic vasculature.12 Ashton proposed that oxygen

FIGURE 135.1. Proliferative diabetic


retinopathy (PDR). (a) Neovascularization at the
disk (NVD) a fan-shaped configuration of new
vessels branching out from the optic nerve
head. Additional findings include extensive
vascular caliber changes, intraretinal
hemorrhages and microaneurysms and cotton
wool spots. (b), Neovascularization elsewhere
in the retina (NVE) new vessels along the
superotemporal arcade with associated
preretinal hemorrhage.
a b

1807
RETINA AND VITREOUS

FIGURE 135.2. Severe retinal ischemia with


rubeosis. (a) The right eye of a 27-year old
diabetic man who presented with rubeosis and
neovascular glaucoma in each eye. There is
NVD, and visual acuity is 20/200. (b) View of
the inferotemporal arcade with numerous
intraretinal hemorrhages and subtle intraretinal
whitening. (c) Fluorescein angiogram
documents severe capillary nonperfusion of the
entire temporal macula and beyond. There is
also hyperfluorescence indicating NVD.
a b (d) Inferior view shows nearly complete
absence of capillary perfusion outside the
peripapillary area.
SECTION 10

c d

deprivation stimulated the production of an angiogenic factor However, FGF has not been shown to have a causal role. The
in both development and retinopathy of prematurity.13 Wise FGFs have a high affinity for heparin and heparin sulfate, and a
expanded the idea of an angiogenic factor to explain a number significant proportion remains associated with the cell matrix.31
of adult retinopathies, including diabetic retinopathy, and This has led to the concept of matrix-associated FGF as a stored
the clinical correlation of retinal ischemia and retinal growth factor. Intracellular FGF may be secreted via alternate
neovascularization has been well established.2,14–16 These pathways, possibly in response to sublethal injury.32
angiogenic factors are released from ischemic retina and The IGFs (IGF-I and IGF-II) are growth-promoting peptides
stimulate new vessel growth from susceptible vessels both with multiple biologic effects.33 IGF-I was initially identified as
locally and at distant locations in the eye (Fig. 135.2). a circulating factor that appeared to mediate the effects of
Angiogenic factors have been isolated from ocular and other growth hormone.34 The role of this growth factor and its
tissues and studied in clinical specimens and experimental associated factors in ocular neovascularization was first
models of neovascularization. Whereas the number of known suggested by the clinical observation of regression of PDR after
angiogenic factors continues to increase, growth factors and infarction of the pituitary during pregnancy.35 IGF-I stimulates
signaling molecules that appear to have particular relevance migration and proliferation of endothelial cells36 and has been
to diabetic retinopathy include basic fibroblast growth factor demonstrated to be angiogenic in vivo at high doses.37 Grant
(bFGF), insulin-like growth factor (IGF), vascular endothelial and colleagues demonstrated increased levels of IGF-I in the
growth factor (VEGF), and the beta isoform of protein vitreous of patients with PDR compared with controls.38
kinase c (PKC). VEGF was first identified in tumor and developmental sys-
bFGF is the best characterized of the fibroblast growth factors tems as a vasopermeability factor and angiogenesis factor, which
(FGFs) and the one-most strongly implicated in the angiogenic was upregulated by hypoxia, a component of ischemia.39 VEGF
process. bFGF stimulates endothelial cell proliferation and and mRNA and protein were shown to be elevated in several
migration, induces protease production by endothelial cells, and experimental systems, including a model of retinal ischemia
stimulates endothelial cells to migrate into 3-dimensional and iris neovascularization in the monkey and retinopathy of
collagen matrices to form capillary-like tubes.17–19 bFGF stimu- prematurity in the mouse and the rat.40–43 Various strategies to
lates angiogenesis in vivo in the chick chorioallantoic mem- inhibit VEGF either completely prevented or greatly reduced
brane and corneal micropocket, in concentrations as low as neovascularization in these models.44,45 VEGF protein levels
100 ng.20–22 bFGF has been isolated from corpus luteum, adrenal were found to be increased in the ocular fluids of patients with
gland, kidney, and retina, and it has been implicated in tumor- PDR or ischemic retinal vein occlusion and decreased after
mediated angiogenesis.17,23 successful laser retinal ablation.46,47 Surgical specimens from
When considering the FGFs as ocular angiogenic factors, one eyes with PDR consistently demonstrated VEGF expression, in
perplexing attribute of both acidic fibroblast growth factor contrast to other growth factors.48 Finally, VEGF has been
(aFGF) and bFGF is their lack of a consensus signal peptide for demonstrated to be sufficient to produce neovascularization, by
secretion.24 As a result, demonstration of aFGF25,26 and bFGF27 injecting VEGF into normal animal eyes49 or by means of a
in the retina does not necessarily mean that the factors are transgenic mouse model.50
exerting any action at that moment. bFGF related messenger Recent work has further suggested that the serine/threonine
RNA (mRNA) and protein levels have been reported to be kinase PKC also appears to play a role in the development of
elevated in a number of experimental systems and clinical diabetes-associated neovascularization, primarily through its
settings, including the retinas of newborn mice exposed to influence on the activity of growth factors such as VEGF.50a In
hyperoxia,28 the retinal pigment epithelium (RPE) after krypton human vascular smooth muscle cells, hyperglycemia-induced
1808 laser injury,29 and ocular fluids from individuals with PDR.30 expression of VEGF is prevented by administration of PKC
Proliferative Diabetic Retinopathy

inhibitors.50b Inhibition of PKCb by the selective inhibitor to PDR.54 In those not using insulin, these numbers were all
ruboxistaurin blocks VEGF’s proliferative, angiogenic, and pro- lower, with only a 34% incidence of any retinopathy. The
permeability effects.50c,50d Using a model of ischemia-induced incidence of legal blindness was 3% over 4 years in the older-
proliferative retinopathy, there is a significant increase in onset group on insulin versus 1.5% in the younger-onset group.
VEGF-mediated retinal neovascularization in transgenic mice, Various investigators have attempted to determine the
overexpressing the PKCb2 isoform, and a corresponding characteristics associated with the severity or progression of
decrease in angiogenic activity in PKCb isoform null mice.50e retinopathy. As already evidenced, the duration of diabetes is
Besides acting as a reservoir for growth factors, the vitreous important in all patients. The WESDR also found that elevated
may exert a mechanical effect in the development of pro- glycosylated hemoglobin levels were associated with more
liferative tissue. Retrospective studies have shown that, whereas severe retinopathy in all groups. Proteinuria was associated with
a complete posterior vitreous detachment (PVD) occurs in severe retinopathy in younger-onset subjects who had diabetes
diabetes as a function of aging, partial PVD is seen in diabetics for 10 years or more, as well as in the older-onset group.
with PDR far more commonly than in individuals without Elevated diastolic blood pressure and male sex were associated
diabetes or in diabetics with background retinopathy.51 The with more severe retinopathy in the younger-onset group and
posterior vitreous face remains attached to the retinal vessels at with elevated systolic blood pressure in the older-onset
the sites of proliferation and provides a scaffold along which the group.53,54
new vessels and fibrous tissue can grow. The resulting tractional A prospective study was carried out at the Joslin Clinic in
forces can lead to the development of traction retinal detach- 153 patients with long-standing diabetes, 90% of whom were in

CHAPTER 135
ment. Precocious PVD, before the development of proliferative the early-onset category.56 The risk of progression to preprolif-
retinopathy, appears to protect patients from traction retinal erative or proliferative retinopathy increased in the presence of
detachments.52 background retinopathy, even if it was slight. The risk of pro-
gression increased exponentially with increasing hemoglobin
EPIDEMIOLOGY A1c. Diastolic blood pressure less than 70 mmHg, and increas-
ing age were protective factors. Since the study involved patients
Epidemiologic studies are important in elucidating the sys- with long-standing diabetes (duration 16–60 years), the results
temic, ocular, and genetic factors associated with the develop- may not apply to patients in the early course of the disease.
ment and progression of diabetic retinopathy. Understanding Genetic factors have been investigated, particularly the histo-
these factors gives insight into the pathophysiology of the disease, compatibility antigens (human leukocyte antigen (HLA)) with
as well as indicating where treatment should be directed, in varying findings.57 Baker and Rand and associates reported that
individual patients, and in the healthcare system as a whole. patients with HLA-DR phenotypes 4/0, 3/0, and X/X (neither
The Wisconsin Epidemiologic Study of Diabetic Retinopathy 3 nor 4) had three to four times the risk of proliferative
(WESDR) is a large population-based study of 2366 subjects retinopathy of that found in patients with the HLA-DR pheno-
divided into two groups: the younger-onset group, or those types usually associated with diabetes (HLA-DR 3/4, 3/X, 4/X).
diagnosed with diabetes before age 30 years, and the older-onset In patients with myopia (2 D or greater), this risk was not
group, or those diagnosed with diabetes after age 30 years.53,54 evident.57,58
The older-onset group was subdivided into those taking insulin Several studies have reported significant mortality associated
and those who did not. In the younger-onset group, the with PDR. In a cohort study at the Joslin Clinic, Rand and col-
prevalence of any retinopathy was closely related to duration of leagues demonstrated that the risk of death was not attributable
diabetes, ranging from 2% in subjects with diabetes for less than to PDR itself.59 The increased risk of death was found only in
2 years to 98% in subjects with diabetes for 15 years or more.53 PDR patients with diabetic nephropathy as evidenced by per-
The severity of retinopathy in this group also increased with sistent proteinuria.
duration of diabetes: the prevalence of PDR ranged from 0% in
subjects with diabetes for less than 5 years to 25% in subjects OCULAR AND SYSTEMIC RISK FACTORS
with diabetes for 15 years and 56% in subjects with diabetes for FOR PROGRESSION TO PDR
20 years or more. In the younger-onset group, macular edema is
rare before diabetes has been present for 10 years, but the
incidence reaches 21% after 20 or more years of disease. In a GLUCOSE CONTROL
population examined at the Joslin Clinic, macular edema was With the introduction of insulin arose the question, is there a
more common in the older-onset group; and in patients under level of glycemic control that will prevent the secondary
the age of 50 years, it was associated with preproliferative or complications of diabetes? If there is such a level, at what point
proliferative retinopathy.55 in the course of the disease must the intervention be made?
In contrast to the younger-onset group, the older-onset group Most clinical studies have shown an association between
was more likely to have retinopathy present at the time diabetes poor control of blood glucose and increased severity of
was diagnosed. The prevalence of any retinopathy in this group retinopathy.53,54,56,58,60 In the WESDR, an elevated glycosylated
in the WESDR was 30% in insulin-taking subjects and 23% hemoglobin was associated with more severe retinopathy in the
in those not taking insulin who had diabetes for less than younger-onset and older-onset diabetics.53,54 In a series at the
2 years.54 This increased to 85% in the group taking insulin and Joslin Clinic, the risk of PDR in long-standing diabetes was
58% in the group not taking insulin after 15 years or more of related to elevated glycosylated hemoglobin levels.56,58 Several
diabetes. After 15 years or more of diabetes, the prevalence prospective studies have begun placing diabetic patients on
of PDR was 20% in those taking insulin and 4% in those not continuous subcutaneous insulin infusion (CSII) systems or
taking insulin. multidose (three to four doses per day) insulin treatment
Incidence and progression of retinopathy were also studied in regimens with home glucose monitoring.61–63
the WESDR. The younger-onset group had the highest 4 year The Kroc Collaborative Study involved 70 patients with
incidence of any retinopathy (59%), as well as progression diabetes and nonproliferative retinopathy who were randomized
(41%), and progression to PDR (10.5%).53 Older-onset patients to CSII or conventional injection treatment.61 Over 8 months,
taking insulin had a 47% 4 year incidence of any retinopathy, glycosylated hemoglobin and blood glucose levels reached near-
34% incidence of progression, and 7% incidence of progression normal levels in the treatment group. The level of retinopathy 1809
RETINA AND VITREOUS

progressed in both groups but was worse in the treatment group,


with increased cotton-wool spots and IRMA. After 2 years of
OCULAR SURGERY
follow-up, however, little difference was found in the rates of Cataracts occur commonly in diabetic patients and may require
progression between the two groups. The Oslo group, studying surgical intervention for both visual rehabilitation and visual-
45 patients randomized to CSII, multidose insulin, or con- ization of the fundus. Cataract surgery in diabetics may be
ventional insulin therapy, also reported an initial worsening of more complicated than in the general population, however, with
retinopathy, with microaneurysms and IRMA.62 One patient poorer postoperative results, including an increased incidence
experienced severe preproliferative retinopathy in both eyes of cystoid macular edema, an increased risk of progression of
after 3 months of CSII and was followed up without laser inter- the retinopathy, and an increased risk of anterior segment
vention but continued on CSII with regression of the neovascularization. The outcome of cataract surgery in
retinopathy after several months.64 After 2 years of follow-up, individuals with diabetes has been studied by a number of
patients receiving CSII or multiple injections had a lower rate investigators.
of progression of retinopathy, as measured by number of Visual outcome in cataract surgery appears to be affected by
microaneurysms and hemorrhages, than did the conventionally the presence or absence of retinopathy preoperatively. In a series
treated group. Both studies, as well as other case reports,65 of 28 eyes of patients with diabetes, but without retinopathy
suggest that rapid achievement of good glycemic control in undergoing extracapsular surgery with lens implantation, the
diabetics with previously poor metabolic control may be detri- corrected visual acuity achieved 12 months postoperatively was
mental, particularly in patients with preproliferative or prolifer- 20/40 or better in 88% of the patients.69 These results compare
SECTION 10

ative retinopathy. favorably with those in nondiabetic individuals. Conversely,


These trials are limited because of their small study popula- patients with retinopathy preoperatively had a poorer visual
tion, short duration, and patient makeup, with varying degrees outcome, with one-third of the 18 eyes achieving a post-
of retinopathy. The Diabetes Control and Complications Trial operative visual acuity less than 20/200. Eight out of the 18 eyes
(DCCT) was a large, randomized, controlled trial, involving developed cystoid macular edema. Severe exudative maculopathy
1441 insulin-dependent subjects, designed to study both primary has been described after cataract extraction in patients with
prevention and secondary intervention.63,66,67 Patients were ran- preoperative background diabetic retinopathy.70 Postoperative
domized to standard therapy or intensive therapy of multidose visual acuity may be worse, despite medical treatment and
insulin injections or CSII. The primary prevention study involved macular laser photocoagulation.
726 subjects who had diabetes for 1–5 years and lacked retino- In another retrospective series, Benson and associates studied
pathy or evidence of renal disease. The secondary intervention the visual acuity results in diabetic patients undergoing
study involved 715 subjects who had diabetes for 1–15 years, extracapsular cataract extraction with placement of a posterior
with minimal background retinopathy and minimal diabetic chamber lens.71 They found that in this referral population, the
nephropathy. Retinopathy was the principal outcome in the final visual acuity was 20/40 or better in 48% of eyes, and that
DCCT. The primary prevention cohort was followed up to deter- 28% had 20/200 or worse visual acuity. Sixty-five percent had
mine whether intensive therapy could prevent the development an improvement in visual acuity of two or more Snellen lines.
of retinopathy, and the secondary intervention cohort was Interestingly, they found that age was a predictor of final visual
followed up to determine whether intensive therapy could delay acuity, patients aged 63 years or younger were more likely to
the progression of retinopathy. Patients in the primary cohort achieve improved visual acuity. The poorer results in the older
were followed up for an average of 6.0 years, and those in the patients were due to complications of macular edema.
secondary intervention cohort were followed-up for 7.0 years. In Neovascularization of the iris developed in 6% of patients, and
the primary prevention cohort, both treatment groups showed a the risk of iris neovascularization in their study population was
steady rise in the cumulative incidence of any retinopathy not increased by posterior capsulotomy.
(defined as ≥1 microaneurysm in two consecutive 6 month sets Studies of the risk of progression of diabetic retinopathy,
of photographs), reaching 90% in the conventional group and apart from macular edema, have reached differing conclusions.
70% in the intensive treatment group. Intensive treatment Sebestyen studied 74 patients with, either no background
slowed the development of retinopathy over the 9 years of diabetic retinopathy, or mild background diabetic retinopathy,
follow-up, with an average risk reduction of 27%, but did not who were undergoing cataract extraction with lens implanta-
prevent its onset.68 Using a more conservative outcome of at tion. He found a similar risk of progression between the operated
least three microaneurysms at two consecutive visits, the size of and the unoperated eyes.72 In another study comparing extra-
the risk reduction with intensive retreatment increased to 63%. capsular and intracapsular techniques, Alpar found the lowest
Intensive therapy was more effective when initiated early in the incidence of progression of retinopathy in the group undergoing
course of insulin-dependent diabetes. extracapsular extraction.73 However, he did not differentiate on
In the secondary intervention cohort, the 9 year cumulative the basis of preoperative retinopathy, which appears from other
incidence of three or more steps of progression, was 56% in studies to be an important factor.
patients who received intensive treatment versus 78% in patients Aiello and co-workers retrospectively studied 154 diabetic
treated conventionally. The incidence of severe nonproliferative patients, who had undergone routine intracapsular cataract
retinopathy was 9.2% versus 26%, and the incidence of neo- extraction in one eye only.74 The second eye served as a control,
vascularization was 8% versus 24%. For macular edema, the and neither eye received laser photocoagulation before surgery
cumulative incidence was 27% versus 44%. Although all levels or during the first year after surgery. Although most of the eyes
of retinopathy included in the study benefited from intensive had either no diabetic retinopathy or background retinopathy,
therapy, the effects were less marked in eyes with more advanced eyes were included with both quiescent PDR and active PDR. In
retinopathy. Since the DCCT did not include patients with all patients, regardless of the degree of preoperative retinopathy,
truly advanced retinopathy, it does not address the question of there was a statistically significant increased risk of rubeosis
whether these eyes would benefit from intensive therapy. All iridis and neovascular glaucoma (8% vs 0%). In patients with
of the observed risk reductions reflected an initial period of 2 active PDR, preoperatively the risk was even higher (40% vs
years or more in which there was no benefit from intensive 0%). There was also an increased risk of vitreous hemorrhage
treatment, averaged against a period beyond 3 years in which after surgery, but this was significant only for the group with
1810 intensive therapy showed increasing reductions in risk. no background retinopathy or mild background retinopathy
Proliferative Diabetic Retinopathy

preoperatively, and not for those with proliferative retinopathy, Klein and co-workers reported on a prospective case-control
probably because of the small number of patients in this group. study comparing pregnant and nonpregnant insulin-taking
Hykin and colleagues retrospectively compared visual acuity diabetic women.80 Risk factors that were evaluated for the
results in 90 patients with PDR or background retinopathy under- progression of retinopathy included glycosylated hemoglobin,
going extracapsular cataract extraction with lens implantation.75 duration of diabetes, current age, diastolic blood pressure, num-
They found that final visual acuity was better in eyes with ber of past pregnancies, and current pregnancy. Women with
background retinopathy (67% ≥ 20/40) compared with eyes with evidence of proliferative retinopathy or evidence of previous
PDR (21% ≥ 20/40). Eyes with quiescent PDR and lacking panretinal photocoagulation were not considered to be at risk
maculopathy did somewhat better, with 52% achieving 20/40 or for progression, and their data were not included in evaluating
better. Deterioration of retinopathy occurred postoperatively, in risk factors for progression. After correcting for glycosylated
50% of eyes with active PDR preoperatively, and immediate hemoglobin level, current pregnancy was found significantly
postoperative fibrinous anterior uveitis preventing early associated with progression, with an adjusted odds ratio of 2:3.
panretinal photocoagulation occurred in over half the patients Diastolic blood pressure had a smaller effect on progression.
with active proliferative retinopathy. Eyes with active prolifer- Pregnant women were significantly more likely to have a
ative retinopathy and cataract are better treated with combined decrease in visual acuity, although this was only a mean dif-
cataract removal and pars plana vitrectomy with intraoperative ference of less than 1 Snellen letter poorer. Klein and co-workers
endolaser. noted that the metabolic control of the pregnant group was
Given the increased risks associated with cataract surgery in markedly better than that of the nonpregnant group.80 This is

CHAPTER 135
patients with diabetic retinopathy, special consideration should probably because of the current obstetric practice of attempting
be made, both preoperatively and postoperatively. The preoper- tighter metabolic control during pregnancy. It has been pos-
ative assessment of the visual impairment caused by cataract tulated that progression of retinopathy during pregnancy may
may be more difficult in the diabetic patient, and use of blue- be at least partly related to the rapid tightening of metabolic
field entoptoscope and potential acuity meter may be helpful.76 control. Laatikainen and associates studied 40 pregnant
Even in cases where the retina appears unremarkable, patients patients randomized at the end of their first trimester to
should be cautioned that there may be progression of diabetic conventional insulin therapy and CSII.81 They found that the
changes postoperatively, with resultant poorer visual acuity. risk of progression of retinopathy was the same. However, two
When a cataract is developing in a diabetic patient, effort should of the patients in the CSII group progressed from background to
be made to perform indicated laser photocoagulation, particu- proliferative retinopathy. Both patients had a rapid decrease in
larly panretinal photocoagulation for active PDR, before the their glycosylated hemoglobin level as they entered CSII treat-
density of the cataract precludes treatment. ment. None of the patients in the conventional therapy group
Surgery should be directed toward maintaining an adequate developed proliferative changes.
aperture for viewing (and possibly treating) the fundus; and Diabetic retinopathy may also be a predictor of pregnancy
phacoemulsification or extracapsular technique with ‘in the bag’ outcome. Diabetic patients taking insulin are known to be at
placement of an intraocular lens is preferred. Because of the increased risk of an adverse pregnancy outcome, including abor-
increased rate of macular edema postoperatively in diabetic tion, perinatal death, and severe congenital anomalies. Klein
patients, it may be worth considering topical or oral non- and colleagues evaluated various risk factors as predictors for
steroidal antiinflammatory agents as well as topical or depot adverse outcome and found that the severity of retinopathy was
steroids. Wounds should be sutured well to allow laser photo- the only variable to significantly predict an adverse outcome.82
coagulation with a contact lens within 2–3 weeks of surgery Pregnancy in diabetic patients is a crucial time for coordi-
if this proves necessary. If yttrium–aluminum–garnet (YAG) nated care by ophthalmologists, obstetricians, and internists.
capsulotomy is indicated postoperatively, again the aperture Guidelines for patient care should be understood by all
should allow visualization of the retina to the equator. Close members of the team. Pregnant diabetic patients should be seen
follow-up postoperatively is essential, with examination of the during the first month of the pregnancy. If no retinopathy is
iris for neovascularization, and a dilated examination of the found, follow-up in each trimester is sufficient unless the
fundus for macular edema or progression of retinopathy, with patient becomes symptomatic. An exception is made if the
consideration of focal or scatter photocoagulation, as indicated. patient is being brought under tighter metabolic control, which
could increase the risk of progression of the retinopathy.
Improvement in metabolic control should be gradual. Patients
PREGNANCY with preproliferative or proliferative disease should be followed
The role of pregnancy in the progression of diabetic retinopathy up every 1–2 months. Laser photocoagulation should be con-
remains somewhat controversial. In his 1950 report, Beetham sidered earlier in pregnant diabetics than in nonpregnant
found that disease in pregnant diabetic patients without retino- diabetics, when there is evidence of early proliferative or active
pathy did not progress during the pregnancy.77 However, preproliferative retinopathy. These findings will be revealed by
patients with evidence of proliferative retinopathy at the onset careful ophthalmoscopic and biomicroscopic examination, and
of pregnancy did so poorly, that he recommended them not to fluorescein angiography can usually be avoided in the assessment
undertake pregnancy. In a study by Rodman and associates in of the pregnant diabetic patient. Although there are no firm data
1979, 8% of 201 pregnant diabetic women, with no background on the risk of vitreous hemorrhage in patients with proliferative
retinopathy, or mild background retinopathy at the onset of retinopathy, PDR is not an indication for cesarean section.
pregnancy, had progression of their retinopathy during
pregnancy.78 In 127 women with proliferative disease, 25% had
retinopathy progress during pregnancy. Laatikainen and col- HYPERTENSION
leagues prospectively studied 73 consecutive pregnant patients The relationship between hypertension and the development of
and noted no significant progression of retinopathy in patients diabetic retinopathy is not clear. In the WESDR, the presence
who lacked or had minimal retinopathy at the beginning of and severity of retinopathy were associated, with elevated
pregnancy.79 However, 13 of the 20 patients (65%) with frank diastolic blood pressure in younger-onset diabetics after 10 years
retinopathy in the first trimester were observed to progress or more of diabetes, and with elevated systolic blood pressure
during pregnancy. in the older-onset diabetics.53,54 In a case-control study at the 1811
RETINA AND VITREOUS

Joslin Clinic, Rand and co-workers found that hypertension was group eyes versus two of the control eyes had an increase in
associated with proliferative retinopathy in patients who had preretinal gliosis. One eye in each group had worsening of
had diabetes for 15 years or more.58 This association remained proliferative retinopathy. Comparison of the variables of visual
significant even if cases with renal disease were excluded. In a acuity, macular edema, capillary closure, NVD, preretinal
prospective study at the Joslin Clinic, Janka and associates neovascularization, preretinal gliosis, and severity of retino-
found different rates of progression of severe retinopathy in pathy showed no statistically significant difference between
patients with diastolic blood pressures less than or greater than the two groups.
70 mmHg.56,83 Chase and colleagues also found that elevated Ramsay and co-workers reported on the ophthalmic out-
diastolic blood pressure, even just to ‘high-normal’ values, comes of pancreas transplant recipients, again comparing suc-
carried an increased risk of retinopathy in young diabetics.84 cessful and unsuccessful transplantations.88 Twenty-two
Since hypertension is a known risk factor for stroke and myo- patients were in the successful transplantation group and 16
cardial infarction, it is important that elevated blood pressure in the control group, with follow-up averaging 24 months.
be treated in the diabetic patient, regardless of its effect on Although the investigators noted no substantial difference
retinopathy. in the progression of retinopathy in the two groups after 2
years, they observed a trend toward less progression of
retinopathy in the treated group after 3 years. In order to
RENAL DISEASE adequately study the ophthalmic outcomes of pancreas
Approximately one-third of juvenile-onset insulin-dependent transplant recipients, it will probably be necessary to pool the
SECTION 10

diabetics experience diabetic nephropathy, with the highest risk patients in transplantation centers across the United States
in the second decade of diabetes.85 Several risk factors may be under a common protocol.
involved in the development of diabetic nephropathy, including Pancreas transplantation still carries significant morbidity
genetic predisposition, hypertension, and poor glycemic control. and mortality and requires chronic immunosuppression. As a
Severe retinopathy is more likely to be found in patients with result, the procedure has been limited to patients with end-
renal insufficiency. In the WESDR, proteinuria was strongly stage nephropathy, which is a group with a high incidence of
associated with severe retinopathy in the younger-onset dia- advanced retinopathy, many of whom have already received
betics with 10 years or more of diabetes.53 Older-onset diabetics panretinal photocoagulation. It may be difficult to show an
with proteinuria were also more likely to have proliferative effect of pancreas transplantation and normoglycemia on this
retinopathy.54 Renal retinopathy will overlie diabetic retino- advanced and already treated eye disease.
pathy in uremic patients and consists of a hypertensive com-
ponent and a uremic component.12 The hypertensive changes OCULAR CONSEQUENCES OF
include nerve fiber layer hemorrhages, cotton-wool spots, and a NEOVASCULAR PROLIFERATION
narrowing and irregular caliber of the retinal arterioles. The
uremic changes include disk edema and diffuse retinal edema,
which may lead to massive macular edema. Treatment of the NVD AND NVE
renal failure, with diuretics, dialysis, or renal transplantation, Neovascularization occurs most frequently in the posterior pole
may result in decreased retinal and macular edema. Laser or within 45o of the optic disk.89 It is very commonly observed
photocoagulation is not very effective in this group of patients; on the disk itself. New vessels on the disk can be easily over-
it is important to consider laser photocoagulation treatment for looked in their early stages, beginning as fine loops or networks
preproliferative retinopathy or early proliferative changes in the of vessels. The vessels gradually increase in caliber from one-
diabetic patient experiencing renal failure. eighth to one-quarter the diameter of a retinal vein at the optic
The treatment of renal failure may result in a different set of disk. The vessels often form a cartwheel configuration with vessels
ocular problems. Patients on hemodialysis are at increased risk radiating out from the center to a circumferential peripheral
of developing elevated intraocular pressure, particularly in eyes vessel (Fig. 135.1a). Occasionally, new vessels will appear similar
that have undergone vitrectomy. Renal transplant patients are to normal retinal vessels, with a large caliber and extending
at an increased risk of acquiring cataract from chronic cortico- across the retina without forming networks. However, these
steroid treatment or cytomegalovirus retinitis from chronic vessels can be distinguished from normal retinal vasculature by
immunosuppression.86 the fact that they cross both arterioles and venules. NVD is best
identified using a magnified stereoscopic view, either by contact
or precorneal lens or with stereoscopic photography.
PANCREAS TRANSPLANTATION NVE is best detected by a thorough fundus examination, with
Pancreas transplantation, alone or in combination with renal binocular indirect ophthalmoscopy combined with biomicro-
transplantation, is a relatively new and risky procedure per- scopy using a lens. Fundus photographs are also helpful in
formed on diabetic patients with advanced diabetic nephropathy detecting early NVE. Neovascularization typically occurs
in conjunction with renal transplantation. Several investigators adjacent to areas of capillary closure, marked by cotton-wool
have begun to examine the effect of transplantation on diabetic spots and hemorrhagic microaneurysms (Fig. 135.1b). IRMA
retinopathy, although all the studies to date have been limited may be difficult to differentiate from early NVE. IRMA is used
by low numbers and short follow-up. Petersen and Vine reported to describe irregular, segmental dilatation of intraretinal
on eight patients who underwent combined pancreas and renal capillaries, representing early neovascular changes or shunt
transplantation from cadaver donors.87 Four patients success- vessels. On fluorescein angiography, IRMA typically does not
fully retained functioning pancreas transplants for at least leak as profusely as do new vessels (Fig. 135.3).
12 months, whereas four patients with failed pancreas transplants
but functioning renal transplants served as the controls. Suc-
cessful pancreas transplantation led to a euglycemic state, with VITREOUS AND PRERETINAL HEMORRHAGE
normal fasting blood glucose, and glycosylated hemoglobin levels. While the posterior hyaloid remains attached, neovascular
However, the progression of retinopathy appeared unaffected. proliferations appear to be on or slightly anterior to the retina,
Three of the study group eyes had an increase in capillary and are usually asymptomatic. Small hemorrhages may occur
1812 closure versus no increase in the control eyes. Four of the study near the growing tips of the vessels, but these usually remain
Proliferative Diabetic Retinopathy

a b c

FIGURE 135.3. Fluorescein angiography of neovascularization. (a) NVE along the vascular arcade. (b) Early fluorescein transit defines NVE.
(c) Late transit shows continued leakage from NVE.

CHAPTER 135
subhyaloid. As the posterior hyaloid detaches, hemorrhages
become less confined and visual acuity may diminish in propor-
tion to the extent and diffusion of the hemorrhage (Fig. 135.4).
Vitreous detachment usually begins in the posterior pole on
either side of the vascular arcades, over the fovea, or temporal
to the macula. Progression of detachment of the vitreous is
halted whenever a tuft of neovascularization or a particularly
strong attachment to a retinal vessel is encountered. The tuft
may be pulled forward as the vitreous contracts, with or without
detachment of the underlying retina. The vitreous detachment
will continue to the periphery, at which point the vitreous is
permanently attached to the vitreous base. The vitreous usually
remains attached at the disk if fibrovascular proliferations are
present.
This process leads to a variety of complex patterns of partial
vitreous detachment. In general, however, the pathophysiology
of partial vitreous detachment favors attachment of the vitreous
to neovascularization, major retinal vessels, the disk, and
obligatorily, the peripheral vitreous base. The outer contour of
the partially detached vitreous has been termed the ‘vitreous
cone’, with the widest aspect of the cone anteriorly and a
variable stem of the cone oriented at the posterior pole. The FIGURE 135.4. A complication of neovascularization. Vitreous
posterior vitreous face and its attachments in a given eye are hemorrhage from NVD.
critical to the understanding of the surgical approaches in PDR
(Fig. 135.5). The posterior vitreous face serves as a surface that seen adjacent to the vessels. This tissue increases as the vessels
may loculate hemorrhage. In addition, it may also have perfo- develop, and it may subsequently contract. Distortion and dis-
rations, which at times may cause it to be confused with an ruption of the normal retinal tissue may result in conjunction
epiretinal membrane or even a macular hole. with the process of vitreous detachment (Fig. 135.6).
Vitreous hemorrhage may occur as a result of vitreous trac- Contraction of the posterior vitreous face and fibrovascular
tion on new vessels. Contraction of the vitreous or fibrovascular proliferation may lead to tractional retinal detachment. Since
proliferation can lead to avulsion of a retinal vessel, usually fibrous proliferation usually progresses along the temporal
a vein, and vitreous hemorrhage. Hemorrhage may also be arcades, the retina along the temporal arcades is usually the
associated with Valsalva’s maneuvers related to coughing or first to detach. Progression of the detachment can then extend
vomiting or occasionally is associated with insulin reactions.90 centrally and peripherally. The rate of progression of an extra-
Most of the time, it occurs during sleep and is unrelated to macular tractional detachment to involve the macula may be as
any obvious factor. Results of the Early Treatment Diabetic low as 15% in a single year.92 In contrast to rhegmatogenous
Retinopathy Study (ETDRS) indicate that aspirin use does not retinal detachments, tractional detachments are typically con-
increase the risk of vitreous hemorrhage.91 cave, are localized, and do not extend to the ora serrata. Trac-
Blood in the fluid vitreous behind the detached posterior tional retinal detachments in PDR have many configurations,
vitreous face remains red until it is absorbed, which usually depending on the location, number, and severity of attachments
occurs over weeks to months. Hemorrhage into the formed of the posterior vitreous face to neovascularization and its
vitreous tends to turn white over time and may require months associated fibrovascular tissue. The activity of the neovascu-
to clear. larization also plays a role. Configurations of tractional detach-
ments may range from an isolated tractional detachment
outside the arcades associated with an area of NVE that has
FIBROUS PROLIFERATION AND RETINAL regressed, to a tractional detachment of the entire posterior
TRACTION retina with extensive contact of the retina, neovascular tissue,
Early in the course of neovascularization, the new vessels appear and the posterior vitreous face. The various configurations have
bare, but as they develop, delicate white fibrous tissue can be implications for the indications for surgical intervention and for 1813
RETINA AND VITREOUS

FIGURE 135.5. Posterior hyaloid (posterior


vitreous face) configurations in PDR. (a) The
posterior hyaloid is attached across the macula,
at the disc, and just beyond each arcade.
Outside of these areas, the vitreous is
detached. In this eye, the posterior hyaloid is
fibrotic and slightly separated from the surface
of the macula, rendering it visible. The
underlying retina is totally attached and the
visual acuity is 20/30. (b) Posterior hyaloid with
a large hole centrally, simulating an epiretinal
membrane with hole. The residual attachment
a b
points of the posterior hyaloid at the disk in
superior, inferior, and inferotemporal locations
may be seen. The retina is attached, and visual
acuity is 20/30. (c) Ultrasound demonstrates
traction retinal detachment and vitreous
attachments (vitreous cone). (d) Intraoperative
view of vitreous surgery with creation of an
opening in the detached peripheral vitreous
SECTION 10

with the vitrectomy instrument. The


membranous character of this tissue is shown.

c d

the prognosis for successful reattachment. Tractional retinal


detachments may also develop retinal breaks, either atrophic or
tractional. The resulting combined tractional–rhegmatogenous
detachment typically progresses, although in some cases the
detachment may remain stable despite the break, particularly in
an eye with extensive panretinal photocoagulation (Fig. 135.7).
Detachments involving the fovea have obvious impact on
vision. However, in other cases, the fibrovascular tissue may
overgrow and obscure the fovea, reducing vision without actual
foveal detachment. Contraction of fibrovascular tissue can also
lead to distortion or horizontal displacement of the macula
(tangential traction). Visual acuity may be reduced in this
condition owing to striae in the macula, and surgery may result
in visual improvement.93

RUBEOSIS
Iris neovascularization or rubeosis is a complication of pro-
liferative retinopathy. Rubeosis is characterized by the networks FIGURE 135.6. Very severe neovascularization with vascular
of branching vessels over the surface of the iris, with vessels overgrowth obscuring the retina and traction retinal detachment.
crossing the scleral spur on gonioscopy. Usually, the new vessels
are apparent on clinical examination, but fluorescein angio-
graphy is occasionally helpful in demonstrating leakage from
the new vessels. Rubeosis is observed in the setting of
proliferative changes and retinal ischemia and also in long- detachment is completed, except in the areas where fibro-
standing retinal detachment related to diabetic retinopathy. It is vascular proliferations prevent complete separation, and
most dramatically evident in eyes with retinal detachment after fibrovascular proliferation ceases. In an untreated eye, there is
unsuccessful vitrectomy. In this setting, rubeosis will develop in usually some element of tractional detachment by the time this
almost all cases and will progress rapidly if the underlying phase is reached, and it typically involves the macula. The optic
detachment is not repaired. nerve develops some pallor, and the retinal arterioles become
attenuated. Retinal hemorrhages and microaneurysms are rare,
and pigmentary changes in the RPE may be seen. Fibrous tissue
INVOLUTIONAL PROLIFERATIVE DIABETIC may be thinner and translucent, with the appearance of vitreous
RETINOPATHY veils in contact with diaphanous retina. Visual loss at the
All diabetic retinopathy eventually reaches an involutional involutional phase may relate to macular detachment, macular
stage. The visual outcome is variable and depends on the ischemia, long-standing macular edema, or optic nerve disease
1814 degree of structural alteration in the posterior pole. Vitreous (Fig. 135.8).
Proliferative Diabetic Retinopathy

a b c

FIGURE 135.7. Posterior break in tractional retinal detachment. (a) Tractional detachment along the superior arcade with a retinal break just
inferior to the neovascular stalk. (b) Confluent laser applied to surround the detachment. (c) Four months later, the detachment is unchanged
and visual acuity remains 20/20. The detachment has remained stable over the subsequent 2.5 years of observation.

CHAPTER 135
a b c

d e f

g h

FIGURE 135.8. Macular ischemia in a 21-year-old man with PDR. (a) Intraretinal hemorrhages and cotton-wool spots. Visual acuity is 20/30.
(b) Fluorescein angiogram demonstrates macular capillary nonperfusion and early disk neovascularization. (c) Late phase of the angiogram
shows continued ischemia with diffuse leakage in the macula. (d) Six months later, the cotton-wool spots have increased and visual acuity is
20/50. (e) Fluorescein angiogram now displays extensive capillary nonperfusion, most prominent in the temporal macula. NVD has increased,
and visual acuity is 20/50. (f) Eight years later, after extensive panretinal photocoagulation and also a vitrectomy for vitreous hemorrhage,
regressed proliferative changes are noted. Sclerotic vessels are seen temporally, and visual acuity is 20/80. (g) Fluorescein angiogram shows
delayed filling of temporal vessels and a greatly increased foveal avascular zone. (h) Late phase of the angiogram shows continued ischemia,
vascular wall leakage in the temporal vessels, and macular edema due to intraretinal vascular abnormalities. 1815
RETINA AND VITREOUS

TREATMENT OF PROLIFERATIVE DIABETIC truction. The best results were obtained by Meyer-Schwickerath
RETINOPATHY and Schott in patients with nonproliferative and early PDR.99
Several months to years after treatment with the xenon arc, flat
neovascular patches remained scarred and atrophic, there was a
PITUITARY ABLATION decrease in exudates, retinal edema, and venous dilatation. New
Some of the earliest attempts at treating PDR involved pituitary retinal vessels disappeared even if not treated directly. Atrophic
ablation. However, this treatment modality has subsequently changes progressed over 4–8 years. Of 73 patients with non-
been supplanted by other procedures that are more effective and proliferative diabetic retinopathy treated with the xenon arc and
that have fewer complications, including laser photocoagulation followed up for 1–9 years, only one patient developed PDR. How-
and vitrectomy. Thus, the technique of pituitary ablation is of ever, diffuse treatments of the retina resulted in large visual-
primarily historical interest today. field defects, hemorrhage, and fibrous proliferation and traction.
Houssay first noted that hypophysectomy reduced the severity Aiello and co-workers, postulated that in order to reproduce
of diabetes in pancreatectomized dogs in 1930.94 Interest was the involutional fundus picture, one needed to be able to
further generated by a report by Poulsen in 1953 of remission of produce multiple small ‘harmless’ chorioretinal scars in the
diabetic retinopathy in a woman with Sheehan’s syndrome, or posterior pole.100 Clearly, this was not possible with the xenon
postpartum anterior pituitary insufficiency.35 Luft and co- arc. However, with the development of the ruby laser, this
workers introduced the technique in humans in the 1950s.95 treatment approach became possible. The ruby laser emits
Various techniques of pituitary ablation were tried, including monochromatic energy at 694.3 nm, a wavelength transmitted
SECTION 10

surgical hypophysectomy, transsphenoidal hypophysectomy, through ocular media and absorbed well by the RPE and
yttrium-90 implantation, x-irradiation, proton beam irradia- choroid. It transmits fairly well through blood, permitting treat-
tion, and stereotactic hypophysectomy using radiofrequency ment through mild vitreous hemorrhage but also making the
coagulation. laser less effective in treating vessels directly. The laser emits
Both short- and long-term follow-up of patients undergoing energy for less than 1 ms. In experimental studies by Campbell
pituitary ablation demonstrated a stabilization of visual acuity and associates, the thermal changes were smaller and more
in survivors, an improvement in both disk and peripheral neo- narrowly confined than those found with the xenon arc. In
vascularization, and a reduction in exudates, microaneurysms, 1965, Campbell and associates reported the results of treating
and hemorrhages.12,35,96 However, the complications of treat- 220 patients for a variety of conditions, including retinal tears,
ment were frequent and significant. As reported by Sharp and retinoschisis, retinal detachment, angiomas, and chorioretinitis.101
colleagues, they included debilitating postural hypotension and Beetham and colleagues began using the ruby laser to treat
asymptomatic hypoglycemia, with several related deaths. patients with diabetic retinopathy in 1967. By 1969, they had
Osteoporosis and avascular necrosis of the hip were reported treated 329 patients with various degrees of proliferating retino-
and were related to steroid replacement. Sterility led to episodes pathy, treating one eye and using the second eye as a control, in
of severe depression in several patients and suicide in at least one patients in whom the degree of retinopathy was approximately
case. In the group from London treated with yttrium-90, the the same.97 Most of the patients treated had flat neovascu-
5 year mortality rate was 18% and the 10 year rate was 51%.96 larization of the retina, or early NVD plus or minus NVE.
Patients were medically evaluated and followed up by complete
ocular examinations, including refraction, field examination,
BEETHAM’S OBSERVATIONS LEADING TO fundus photography, and fluorescein angiography. The ruby
RATIONALE FOR LASER THERAPY laser was used at a 2.5o aperture to produce 750 individual
Early investigators observed that certain ocular conditions seemed chorioretinal lesions of grade I or II intensity. Applications were
to prevent severe diabetic retinopathy. Eyes with chorioretinal placed in all areas of the posterior pole, avoiding the disk,
scarring, optic atrophy, retinitis pigmentosa, and high myopia macula, and papillomacular bundle.
were protected from severe proliferative retinopathy.97 Beetham, With 1–2 years of follow-up, there was regression of neovas-
studying patients at the Joslin Clinic, described spontaneous cularization in the treated eyes that was statistically significant.97
resolution of PDR in approximately 10% of patients.98 The Eighty percent of the treated eyes showed some regression, and
fundus picture in these patients consisted of lacy, reticulated 54% had complete disappearance of neovascularization, with
proliferative tissue; attenuated arterioles, and obliterated vessels the control eyes remaining unchanged or worsening. Aiello and
appearing as white lines. The fundus picture resembled that of co-workers also noted a decrease in the diffuse angiopathy of the
the ocular conditions described earlier, as well as the fundus posterior pole, even in the untreated area of the papillomacular
picture that developed after successful hypophysectomy for PDR. bundle.100 Results in patients with more severe proliferative
Over a period of time, it was recognized that this appearance retinopathy were more disappointing, although with fewer
and the subsequent possibility of inducing regression of PDR patients falling into these groups so that statistical comparisons
with preserved visual acuity could be achieved with laser could not be made. Of seven patients with elevated disk neo-
photocoagulation. vascularization who were treated with ruby laser, two patients
showed improvement, three patients suffered severe vitreous
hemorrhage with one developing severe fibrous proliferation
EARLY LASER TRIALS (DIRECT ABLATION) after the hemorrhage, and two patients remained stable. None
Meyer-Schwickerath and Schott first used light photo- of the treated patients in any group developed sector visual-field
coagulation to treat diabetic retinopathy in 1955.99 The xenon defects, and there was no related vitreous traction, iritis, cata-
arc they used produced white light emitting multiple wave- racts, or elevated intraocular pressure.
lengths. Lesions of moderate therapeutic intensity were full- Aiello and Beetham and their associates noted the following
thickness retinal burns histologically, with destruction of inner results after treatment: neovascular nets disappeared or became
retina, explaining the frequent occurrence of visual-field defects. nonfilling, diffuse angiopathy improved, dot-blot hemorrhages
With the xenon arc, one could obliterate vessels directly, treat- decreased, retinal edema decreased, the disk became paler, there
ing flat neovascular patches, dilated retinal capillaries, micro- was no change in fibrous proliferation, and the retinal veins
aneurysms, and edematous retina. Treating elevated vessels were noted to be less ‘leaky’ by fluorescein.97,100 The patho-
1816 required increased power and resulted in extensive retinal des- physiologic mechanism by which the laser treatment worked
Proliferative Diabetic Retinopathy

was unclear, possibly involving the reduction in metabolic glaucoma. Special consideration should also be given to the
demand by destroying functioning retinal tissue, or altering the history, compliance, medical complications precluding follow-
hemodynamic statues of the retinal-choroid layer. Several up, and other clinical aspects seen in the patient. For instance,
decades later, the issue is still not completely resolved, although it may be argued that a juvenile diabetic with visual loss caused
alterations in the levels of various modulators of angiogenesis, by severe proliferative retinopathy in one eye should be treated
such as VEGF, likely play a role.46,47 Aiello and Beetham and in the second eye when any degree of proliferative retinopathy,
their associates believed that a ‘green’ laser might be useful to preproliferative retinopathy, or increasing retinal ischemia is
directly treat elevated vessels prone to hemorrhage because documented. Similarly a pregnant woman with proliferative
there would be greater absorption by the blood column. They changes may be treated sooner because of the possibility of
also believed that their results warranted a large, long-term, rapid progression of the retinopathy, particularly if tight
controlled study. metabolic control is instituted.

Technique
DIABETIC RETINOPATHY STUDY AND Laser photocoagulation has effectively replaced the xenon arc
MODERN PANRETINAL PHOTOCOAGULATION photocoagulator as the instrument of choice. Multiple wave-
(PRP) lengths and instruments are available and effective. Argon green
Indications (514 nm) is most commonly employed, and the energy is well
The early studies of Beetham and Aiello and associates absorbed by blood-filled vessels and by pigment in the RPE.

CHAPTER 135
suggested a beneficial effect of laser photocoagulation in con- Argon blue-green (488 nm) is rarely used, although it was the
trolling PDR. DRS was a multicentric randomized prospective standard of therapy for the DRS. It is more widely scattered by
study designed to determine whether laser treatment in media and may lead to long-term retinal toxicity in the treating
diabetics prevented severe visual loss. It proved very quickly physician. Dye yellow (577 nm) is well absorbed by blood and
that severe visual loss (visual acuity < 5/200) occurred ~60% less may permit direct treatment of new vessels in certain instances,
frequently in eyes treated with photocoagulation than in eyes such as NVE persisting after panretinal photocoagulation.
assigned to no treatment.2,3 The study was also able to identify Krypton red (647 nm) offers better penetration through nuclear
certain high-risk characteristics in subgroups of patients that sclerotic cataracts and, to a lesser extent, vitreous hemorrhage
led to poor outcome without treatment and in which treatment than does the argon green wavelength. Improved penetration to
clearly was of benefit.102 After 4 years, the study was changed to the choroid may make this wavelength more painful to the
allow treatment of previously untreated eyes that developed patient. More recently, solid-state diode lasers emitting wave-
these high-risk characteristics. lengths between 780 and 850 nm have become available for
The high risk characteristics demonstrated by the DRS are: medical use. They offer the advantage of small size and porta-
• moderate to severe NVD (greater than standard bility, as well as low power requirements. The longer wave-
photograph 10A), lengths can penetrate media opacity, but they do require more
• any NVD with vitreous or preretinal hemorrhage, and power to produce equivalent retinal lesions, and they may be
moderate to severe NVE with vitreous or preretinal associated with more patient discomfort.
hemorrhage. Laser photocoagulation can be delivered through a slit-lamp
system, an indirect ophthalmoscope, or an endolaser probe.
Other indications, although not clearly demonstrated by the Transpupillary slit-lamp delivery is the most common delivery
study, are also widely employed, and these are summarized in system for the treatment of adults. The indirect ophthalmo-
Table 135.1. These include widespread capillary dropout, moderate scope laser is available with argon, krypton, or diode lasers and
to severe NVE alone, and rubeosis with or without neovascular permits panretinal photocoagulation in patients under general
anesthesia or in a recumbent position. The endolaser, argon,
krypton, or diode is restricted to be used at vitrectomy.
TABLE 135.1 Indications for Panretinal Photocoagulation for
Topical anesthesia is usually adequate, although retrobulbar
Proliferative Diabetic Retinopathy or peribulbar anesthesia may be needed for re-treatments, or for
treatment with longer wavelengths, or for indirect ophthalmo-
DRS High-Risk Characteristics scope delivery. Oral diazepam supplementation can also be
NVD of moderate to severe degree (greater than standard useful in a patient who is very nervous. The lenses used for slit-
photo 10A)
NVD of any degree if associated with preretinal or vitreous lamp delivery include the Rodenstock panfunduscopic lens, the
hemorrhage Volk quadraspheric lens, and the Goldmann three-mirror lens.
NVE of moderate to severe degree if associated with preretinal or The Rodenstock and Volk lenses allow one to view a large area
vitreous hemorrhage of the fundus during treatment and are popular for the perfor-
Additional Indications Widely Employed mance of panretinal photocoagulation. With these lenses, the
Rubeosis with or without neovascular glaucoma image is inverted and far peripheral burns are more difficult to
Moderate to severe NVE alone, particularly in juvenile diabetic place than with the Goldmann lens. The Rodenstock and Volk
patients lenses magnify the spot size, and relatively more power is
Widespread retinal ischemia and capillary drop-out on fluorescein required for these lenses. The Goldmann lens allows placement
angiography of far peripheral burns, but it provides a view of only a small
Special Situations for Consideration of Panretinal area of the retina.
Photocoagulation Spot size depends on the lens selected, usually 500 mm for the
PDR developing in pregnancy, particularly with the institution of Goldmann lens and 200 mm for the Rodenstock lens, to achieve
tight metabolic control
~500 mm burn. Occasionally, larger lesions are employed for
Preproliferative retinopathy in the second eye of a juvenile diabetic
patient with severe PDR in the other eye heavy therapy (e.g., when treating rubeosis). The duration is
typically 0.1–0.2 s. Longer durations can be used in patients
Abbreviations: DRS, Diabetic Retinopathy Study; NVD, neovascularization at the
disc; NVE, neovascularization elsewhere in the retina; PDR, proliferative diabetic
with media opacity, as well as for treatment with longer wave-
retinopathy. lengths. The power should be titrated to produce a gray-white
burn. Treatment should begin at 100 mW with the Goldmann 1817
RETINA AND VITREOUS

lens or 150 mW with the Rodenstock or Volk lens, although a Blankenship have shown that regression of neovascularization
heavily pigmented fundus in an aphakic or pseudophakic eye 3 weeks after treatment is a good indicator of longer-term visual
suggests an even lower initial setting. The number of spots is results.109 Vine suggested that eyes be assessed 6–8 weeks
less critical than the response to therapy and follow-up. The after laser treatment and that treatment be augmented if high-
DRS protocol specified 800–1600 spots 500 mm in size. The risk characteristics persist.110 He described a group of
burns should be placed 1–1.5 burn widths apart, with focal con- ‘nonresponders’ who continued to have high-risk characteristics
fluent bombardment of the NVE. If possible, the treatment despite augmented PRP averaging 3000 Goldman burns.
inferiorly should be heavier than the treatment superiorly to Approximately 50% of these ‘nonresponders’ did respond to
preserve downgaze field. In treating the temporal raphe, a additional low-intensity, but extensive, photocoagulation,
barrier line should be placed 2.5 disk diameters temporal to the reaching an average of 7550 Goldman burns, with preservation
center of the macula, with treatment extending distal to the of visual acuity.
barrier. Panretinal treatments are usually divided over two to Although the DRS used blue-green argon wavelengths or
three sessions, but they may be given in a single session if xenon arc for photocoagulation, other wavelengths are
required. One study found no significant long-term differences employed by treating ophthalmologists. Argon green has
in single-session versus multiple-session treatment, but fewer replaced argon blue-green to avoid long-term retinal toxicity in
transient choroidal and exudative retinal detachments were the treating physician. Dye yellow, krypton red, and diodes
observed in the multiple-session group.103 emitting in the near-infrared have all been utilized for PRP.
Studies have compared the effectiveness of certain wavelengths
SECTION 10

Results but not all. Krypton red photocoagulation was demonstrated to


The DRS clearly demonstrated that photocoagulation in be as effective as argon blue-green for the treatment of PDR,
selected diabetics reduced the risk of severe visual loss. Eligible comparing visual outcome, regression of vessels, and incidence
patients for the DRS had proliferative retinopathy in at least of complications.111
one eye or severe nonproliferative retinopathy in two eyes, and There is some controversy regarding photocoagulation in the
visual acuity of 20/100 or better in each eye. One eye was ran- presence of tractional retinal detachment. If the tractional
domly assigned to treatment, and treatment was randomized retinal detachment involves the fovea, vitrectomy is indicated.
between argon laser and xenon arc photocoagulation. The rate However, concerns are often expressed that photocoagulation in
of severe visual loss (visual acuity < 5/200) was reduced by treat- patients with extrafoveal tractional detachments will lead to
ment from 16% in nontreated eyes over 2 years to 6% in treated worsening of the detachment and involvement of the fovea. The
eyes, a reduction of 57%.2,3,104 The DRS identified certain sub- DRS found that harmful treatment effects, including decreased
groups, based on severity of the retinopathy, for which the treat- visual acuity, were associated with preexisting fibrous prolifera-
ment effect outweighed any harmful effect. Severe visual loss in tions and localized retinal detachments, particularly in the
patients with these retinopathy gradings (called ‘high-risk cha- xenon-treated group.105 However, the report also confirmed that
racteristics’ and discussed earlier) fell from 26% in nontreated those patients with severe proliferative retinopathy still bene-
eyes to 11% in treated eyes.3,102,104 Harmful effects of treatment fited from argon laser treatment. Some authors have suggested
were also identified and were somewhat greater in the xenon- ‘prophylactic vitrectomy’ for traction retinal detachments
treated group of the DRS. Estimates of persistent visual acuity ‘threatening’ the macula.112 This approach may unnecessarily
loss attributable to treatment in the xenon-treated eyes were subject some patients to the risks of vitrectomy surgery. One
19% with loss of one line of visual acuity and 11% with loss of study investigating argon laser photocoagulation in patients
two lines.104,105 In the argon-treated group, these numbers were with severe proliferative retinopathy and posterior extrafoveal
11% and 3%, respectively. Twenty-five percent of the xenon- traction detachments found that detachments rarely progress
treated eyes demonstrated a modest loss of visual field, and an after treatment to involve the fovea.113 In this study, the area of
additional 25% had more severe field loss. Five percent of the detachment was avoided with otherwise standard photocoagu-
argon group showed some constriction in visual field. It was felt lation techniques and without any effort to wall off the
that some adverse treatment effects were related to focal treat- detachment.113 PRP with careful follow-up should be the first
ment of NVD and elevated NVE, and this aspect of treatment line of therapy in patients with proliferative retinopathy and
was abandoned.104 Other studies comparing the use of xenon traction retinal detachments not involving the fovea.
arc and laser photocoagulation failed to demonstrate any signi- The DRS found that eyes with high-risk characteristics have
ficant difference in the two modalities, either in effectiveness or a 2 year risk of severe visual loss of 25%. Scatter photocoagula-
in the rate of visual loss related to treatment.106,107 However, they tion reduces the risk of severe visual loss by 50% or more. Eyes
did confirm the greater risk of field loss occurring after xenon with severe nonproliferative retinopathy or proliferative retino-
arc treatment, which probably correlates with the inner retinal pathy without high-risk characteristics have a 2 year risk of
damage with a xenon arc burn evident on histopathology.108 severe visual loss of 3–7%. Thus, the risk of visual-acuity loss
Regression of neovascularization occurs in 30–55% of eyes relating to treatment assumes greater relative importance.104
after laser photocoagulation using various treatment approaches The DRS was unable to determine whether deferral of treat-
and may correlate with visual prognosis (Fig. 135.9). The DRS ment with observation in this group was better than early
found complete regression of NVD in 29.8% and partial regres- treatment. The Early Treatment of Diabetic Retinopathy Study
sion in 24.5% of eyes 12 months after treatment.2 Blankenship was designed in part to determine the optimal timing of photo-
compared central and peripheral photocoagulation and found a coagulation. Early treatment for nonproliferative retinopathy
trend toward decreased visual loss (related to treatment) in the was compared with deferral of photocoagulation until high-risk
peripheral distribution group and a slightly smaller loss of characteristics developed. There was a small reduction in the
visual field. Regression of NVD was similar in both groups, with rate of severe visual loss with early treatment, but the rates of
47% complete regression in the peripheral distribution group severe visual loss were low in both groups (2.6% for the early
versus 40% in the central distribution group. In both groups, treatment group, and 3.7% for the deferral group).114 Therefore,
33% of patients had partial regression of NVD after photo- the study group initially recommended that as long as careful
coagulation.108 Regression of NVD should be assessed several follow-up can be provided for the patient, PRP is not recom-
weeks after photocoagulation as a prognostic indicator and to mended for eyes with mild or moderate nonproliferative retino-
1818 determine whether additional treatment is necessary. Doft and pathy. It is important to note that the data was subsequently
Proliferative Diabetic Retinopathy

a b c

CHAPTER 135
d e f

g h i

j k

FIGURE 135.9. Progression of PDR despite panretinal photocoagulation. (a) A 29-year-old man with preproliferative changes in the right eye.
Vascular dilatation, cotton-wool spots, and intraretinal hemorrhages are seen. Visual acuity is 20/25. (b) Superior retina with similar
preproliferative changes. (c) Fluorescein angiogram shows numerous microaneurysms with leakage and an enlarged foveal avascular zone.
(d) Superior retina with capillary nonperfusion and leakage into the vascular walls. (e) Patient was lost to follow-up for 1.5 years and presented
with severe NVD. Visual acuity is 20/25. (f) Fluorescein angiogram shows marked progression of ischemic changes in association with prominent
leakage from NVD. (g) Angiogram of the superior retina shows greatly increased ischemia compared with the previous angiogram. (h) Angiogram
of the nasal retina documents almost total closure of the capillary bed in association with areas of neovascularization. (i) Six months later, NVD
progressed despite extensive and repeated panretinal photocoagulation. (j) After an additional 10 months, a traction retinal detachment involving
the fovea developed. The retina was obscured by neovascular tissue, and visual acuity is counting fingers at 1 ft. (k), One year after vitrectomy
and endolaser treatment, the retina is attached but displays vascular and optic atrophy, and visual acuity is 20/200.

1819
RETINA AND VITREOUS

re-analyzed taking into account the type of diabetes, type 1 or pressure can occur, particularly after heavy treatment, because
type 2. In this analysis, there was an advantage in treating of choroidal swelling and angle shallowing, but it usually
severe nonproliferative or early PDR with PRP in patients with resolves after 48 h.116 It can usually be avoided with divided
type 2 diabetes.115 The physician must also incorporate clinical sessions or lighter treatment. The cornea in diabetic patients is
judgment, including assessment of the fellow eye, progression very sensitive to contact lens trauma, and corneal abrasion
of lens opacities, and other conditions. during treatment may result in a persistent epithelial defect.
The cornea should be inspected after treatment and any abrasions
Complications treated appropriately. Mydriasis is the result of laser damage to
Laser photocoagulation is clearly an effective treatment but can nerves in the uveal tract and is permanent. Paralysis of accom-
result in complications (Table 135.2). These complications may modation can occur but is usually transient.117
occur during treatment in the immediate postoperative period Macular edema and visual acuity loss of one to three lines can
or present as long-term problems (Fig. 135.10). Pain during occur and are more common in patients with perifoveal capillary
treatment is usually transient but may require retrobulbar nonperfusion. Maximum macular edema after panretinal
anesthesia for completion of the session. Increased intraocular photocoagulation can occur anywhere from 4 to 7 weeks after
the initial laser treatment. However, visual recovery is usually
good.117a Visual-field loss secondary to photocoagulation was
TABLE 135.2 Complications of Panretinal Photocoagulation documented in the DRS and is related to the extent of
treatment;104 loss of dark adaptation can also occur after PRP.118
SECTION 10

Foveal burn Choroidal detachment and exudative retinal detachment are


Optic disc damage usually the result of very heavy PRP and generally resolve
spontaneously.119 Choroidal hemorrhage can occur with a very
Macular edema
heavy burn, particularly with longer wavelengths, but is usually
Choroidal hemorrhage limited and resolves spontaneously. Subretinal neovasculariza-
Choroidal neovascularization
tion has been reported and should be treated if it is macular.120
Foveal burns are the result of the surgeon’s disorientation or
Choroidal detachment unfortunate ocular movement and result in permanent loss of
Exudative retinal detachment central acuity, but there may be some improvement after the
initial loss. Vision loss has also been reported in association
Vitreous hemorrhage
with peripapillary treatment.121 Vitreous hemorrhage can result
Pain during treatment from rupture of neovascular vessels during treatment (rarely) or
Increased intraocular pressure from shrinkage and regression of neovascular tissue after treat-
ment (commonly). Hemorrhage that occurs after panretinal
Corneal abrasion photocoagulation usually resolves with time but may occasion-
Mydriasis and paresis of accommodation ally require vitrectomy. Direct treatment of elevated neovascu-
larization with green (514 nm) or yellow (577 nm) wavelengths
Loss of visual field
has a limited role and is restricted to eyes with recurrent
Loss of dark adaptation vitreous hemorrhage and persistent neovascularization despite
Lens opacities complete PRP.
Lens opacities can occur with high energy and misfocusing,
Increase in traction detachments particularly with the panfundus-style lenses, and are usually
permanent but nonprogressive.122,123 Vascular occlusion can

FIGURE 135.10. Panretinal photocoagulation


and complications. (a) Wide-angle photograph
of a typical panretinal photocoagulation pattern.
(b) Choroidal and exudative retinal detachment
after extensive photocoagulation for rubeosis.
(c) Macular edema 2 weeks after panretinal
photocoagulation. Visual acuity is at the
counting fingers at 2 ft level, and was 20/30
before laser. (d) Fluorescein angiogram
documents cystoid edema. Edema resolved in
1 month with visual recovery.
a b

c d
1820
Proliferative Diabetic Retinopathy

result from placement of burns over a vessel. Frequently, the standardized photocoagulation management in the deferred
vessel will again become patent. Although heavy treatment with vitrectomy group and without the use of endolaser photocoagu-
the xenon arc photocoagulator in eyes with active vitreoretinal lation in diabetic vitrectomy in either group, and the validity
traction can lead to progression of an extrafoveal tractional of its conclusions may be questioned. Nevertheless, early
retinal detachment into the fovea in certain cases, data indicate vitrectomy offers the chance for prompt recovery of vision,
that argon laser therapy may be safely performed in these which is of greatest importance to patients who do not have
eyes.113 Care should be taken to avoid heavy treatment near useful vision in the fellow eye.126
areas of vitreoretinal traction. Progressive fibrovascular proliferation in diabetes can lead to
tractional retinal detachment. Posterior tractional detachments
THE ROLE OF VITRECTOMY FOR not involving the fovea may remain stable and should be
PROLIFERATIVE DIABETIC RETINOPATHY observed.92,113,126 However, once the fovea is involved, vitrec-
tomy is indicated. Surgery should be performed promptly because
Indications degenerative changes in the detached retina will prevent visual
Laser photocoagulation allows effective treatment of moderate recovery after reattachment (Fig. 135.11). The combination of
to severe PDR. However, although treatment substantially vitreous traction and membrane contraction can lead to retinal
reduces the risk of severe visual loss, PDR in a certain number holes and combined traction-rhegmatogenous retinal detach-
of eyes progresses to tractional retinal detachment and vision ments, which need to be approached via pars plana vitrectomy.
loss. Standard laser cannot be performed in eyes with vitreous Some patients will present with cataract and proliferative

CHAPTER 135
hemorrhage precluding visualization of the retina. Vitrectomy disease. If adequate PRP cannot be performed because of media
was first introduced by Machemer in the 1970s as a method opacity, there are several management options. Cataract surgery
of removing nonclearing vitreous hemorrhage.124 As techniques can be performed using extracapsular or phacoemulsification
and instrumentation have improved, its indications have techniques, with PRP performed within a few days of surgery. It
broadened (Table 135.3).125,126 should be kept in mind that proliferation may progress rapidly
Removing dense, nonclearing vitreous hemorrhage remains after cataract extraction. Alternatively, pars plana vitrectomy and
an important indication for vitrectomy in diabetics. The Diabetic the endolaser can be performed in conjunction with cataract
Vitrectomy Study Group studied the timing of vitrectomy and removal, either pars plana lensectomy or phacoemulsification
its effect on visual outcome and complications.127,128 They via a limbal approach, with intraocular lens placement.92,126,129,130
included patients with severe vitreous hemorrhage (visual Lensectomy can be performed with preservation of the anterior
acuity < 5 /200) and compared early vitrectomy (after 1 month) capsule and sulcus fixation of a posterior chamber intraocular
with deferred vitrectomy (after 1 year). In patients with type 1 lens after completion of the vitrectomy. Alternatively,
diabetes, early vitrectomy offered a greater chance of visual phacoemulsification may be performed, with placement of the
recovery. In type 2 diabetics and mixed type, the visual results intraocular lens in the capsular bag either preceding pars plana
were essentially the same whether vitrectomy was performed vitrectomy or after its completion. The advantage to placement
early or after 1 year. However, the study was performed without of the intraocular lens after the vitrectomy is improved visual-
ization of the retina during vitrectomy. A combined surgical
approach may be most useful in cases when the cataract is
visually significant and would prevent visualization of the fun-
TABLE 135.3 Indications for Vitrectomy in Proliferative Diabetic dus during vitrectomy and when the anticipated retinal surgery
Retinopathy
is not unduly complicated.
Vitreous hemorrhage Iris neovascularization, with or without angle involvement
and glaucoma, is conventionally treated with panretinal photo-
Nonclearing, based on duration and visual need
coagulation. In cases of rubeosis, vitrectomy with endolaser use,
To permit photocoagulation of active retinopathy with or without lensectomy, is indicated in patients with media
Traction retinal detachment involving the fovea opacities such as cataract or vitreous hemorrhage or when
retinal reattachment is also necessary.126,131
Visual loss owing to macular striae or distortion (tangential traction) Epiretinal membranes develop in diabetes after premacular
Combined traction-rhegmatogenous retinal detachment hemorrhages or after extensive photocoagulation. Vitrectomy is
indicated when the patient’s vision is compromised enough to
Rubeosis with media opacity precluding panretinal photocoagulation
warrant surgery.
Visual loss owing to epiretinal membrane or opacified posterior
vitreous face Technique
Progressive neovascularization unresponsive to retinal ablation The goals of vitrectomy in diabetic retinopathy are simple in
concept. One major objective is to remove media opacities,

FIGURE 135.11. Tractional retinal detachment


involving the fovea. (a) Preoperative
photograph illustrates extensive
neovascularization emanating from the disk
with tractional detachment. Visual acuity is
20/400. (b) Postoperative photograph after
vitrectomy shows retinal reattachment and
removal of neovascular tissue. Visual acuity is
20/40.

a b
1821
RETINA AND VITREOUS

either vitreous hemorrhage or cataract. The second objective is frequency of retinal breaks and a possibility of increased intra-
to relieve vitreous traction causing detachment, distortion, or ocular hemorrhage from transection across the neovascular
displacement of the fovea, either transvitreal traction from stalks. In actual practice, a combination of surgical approaches
anterior vitreous to posterior attachments or tangential traction is frequently required in a given eye.
from proliferative tissue adherent to the retinal surface. As Diathermy is used as needed to obtain hemostasis, but most
dissection is performed, it is important to obtain hemostasis to hemorrhages can be controlled by elevation of the infusion fluid
preserve visualization and allow for effective endophoto- height.126 Airfluid exchange with internal drainage of subretinal
coagulation. The final goal is to perform retinal ablation, usually fluid is used in cases of retinal detachment with a posterior
by laser, to prevent subsequent neovascular proliferation.126 retinal break. Panretinal endophotocoagulation is performed
Two major differences in the technical approach to diabetic when there is active neovascularization or preoperative iris
vitrectomy are (1) the segmentation technique, and (2) the neovascularization or if panretinal photocoagulation has not
delamination and en-bloc resection techniques. In all approaches, been performed before surgery.92,131 Endophotocoagulation is
the anterior vitreous is removed to permit visualization and to also used to treat iatrogenic retinal breaks. Trans-scleral
create a fluid space posterior to the lens. In segmentation, an cryopexy remains a useful tool when extensive retinal ablation
opening is made in the mid-peripheral detached vitreous where is required or the fundus view is lost. Intraocular tamponade
it is typically separated from the underlying retina.124,126 This with air, long-acting gas, or silicone oil is used in cases of retinal
opening is extended 360o at the equator and eliminates antero- detachment.
posterior traction. This ‘truncation of the vitreous cone’ sepa-
SECTION 10

rates the posterior vitreous face with its attachments to the Results
retina from the vitreous base anteriorly. Segmentation then The results of vitrectomy for PDR have improved steadily since
proceeds with dissection of the posterior vitreous face with the mid-1970s owing to increasing vitreoretinal surgical sophis-
vertical scissors cuts around each neovascular attachment of tication in general and the introduction of the endolaser and the
the posterior vitreous face to the underlying retina.92,126 The continuing development of intraocular tamponade in particular.
end result is the segmentation of the posterior vitreous face into Surgical results in patients with vitreous hemorrhage alone
small islands of residual neovascular attachments, and this suggest that 71–78% of patients will have improved vision
segmentation relieves contraction between these points and 6 months after vitrectomy133–135 and that 76% of patients will
permits reattachment of the retina. These islands can then be have visual acuity of 5/200 or better. For patients with traction
trimmed with the vitrectomy instrument. retinal detachment involving the fovea, 57–75% of patients will
In delamination and en-bloc resection techniques, the have improved visual acuity,136–141 with 20/200 or greater acuity
anteroposterior traction is not relieved initially but is used to at 6 months in 40–54% of patients (Fig. 135.12).138,140,141
facilitate dissection of the posterior vitreous face in a horizontal Anatomic reattachment of the macula can now be obtained
fashion. In the delamination technique, an opening is created in intraoperatively in the great majority of cases,137,138 but recur-
the posterior vitreous face over the macula, gaining access to rent macular detachment (resulting from recurrent epiretinal
the surgical plane between the posterior vitreous face and the tissue or rhegmatogenous detachment) occurs in 5–23%
retina. Horizontal scissors transection across the neovascular of cases.137,140,141 For patients with combined tractional–
attachments above the disk and major vascular arcades is rhegmatogenous retinal detachment who require vitrectomy for
performed radiating outward from the posterior pole, ultimately management, long-term macular re-attachment is obtained in
freeing the retina from the posterior vitreous face entirely.92 In 64% of cases, and improved visual acuity is observed in 53% of
the conceptually similar en bloc technique, access to this same cases, with 55% of these patients having postoperative visual
surgical plane is accomplished by creating an opening in the acuity greater than 5/200.142 These results are an improvement
peripheral detached vitreous and then proceeding with hori- over earlier series in which scleral buckling alone was available
zontal scissors transection across the posterior pole.132 In both for repair, in which the retina was reattached in 46% of cases
techniques, the dissection is concluded by using the vitrectomy and 43% had improved visual acuity.143 For patients with
instrument to remove the now freed posterior vitreous face as it distortion of the macula from contraction of proliferative tissue
hangs supported by the remaining attachments to the anterior (tangential traction syndrome), visual acuity was improved in
vitreous base. four of four of patients in one report (Fig. 135.13).93 In a large
It should be emphasized that each of the techniques men- series, preoperative factors associated with a negative prognostic
tioned previously has adherents and detractors. The segmenta- value for visual recovery include iris neovascularization, cata-
tion technique is criticized for the incomplete removal of ract, visual acuity less than 5/200, and retinal detachment.144
proliferative tissue from the retina with possible recurrent Long-term studies indicate the stability of visual and anatomic
hemorrhage or membrane proliferation, and the horizontal dis- results of vitreoretinal surgery for PDR.145 However, postoper-
section techniques are criticized because there is a greater ative complications such as rubeosis (with or without neovascular

FIGURE 135.12. Tractional retinal detachment


involving the fovea. (a) Preoperative photograph
shows severe neovascularization with
detachment of the fovea. Visual acuity is
20/400. (b) Postoperative appearance after
vitrectomy shows retinal reattachment with
continued inferior distortion of the macula.
Visual acuity is 20/200.

a b
1822
Proliferative Diabetic Retinopathy

a b c

FIGURE 135.13. Tangential traction on the fovea with decreased vision. (a) Preoperative appearance shows macular striae to regressed
neovascular tissue to the disc. Visual acuity is 20/80. (b) Fluorescein angiogram documents extensive cystoid edema, primarily affecting the area
of traction. (c) Postoperative photograph 2 months after vitrectomy and removal of posterior vitreous face. Traction is relieved and visual acuity
is 20/70.

CHAPTER 135
a b

FIGURE 135.14. Complications of vitrectomy. (a) Rubeosis, hyphema, neovascular glaucoma, and recurrent detachment after failed vitrectomy
for traction retinal detachment involving the fovea; visual acuity is at the no-light perception level. (b) Anterior hyaloid fibrovascular proliferation in
a 22-year-old man after phakic diabetic vitrectomy. Visual acuity is at the seeing-hand motions level, and the eye was lost despite repeat surgery.

glaucoma), retinal detachment, recurrent vitreous hemorrhage, nized retinal holes or traction and requires reoperation.92,126
and anterior hyaloid fibrovascular proliferation are the chief Endophthalmitis is rare and should be diagnosed and treated
causes of irreparable visual loss in these patients.133–142,146,147 emergently. Glial recurrence occurs rarely, the glial tissue
proliferates directly on the retinal surface and is managed
Complications surgically with a segmentation–delamination approach.92
Postoperative complications after vitrectomy can be early Iris and angle neovascularization may develop after vitrectomy;
(within the first week) or late (weeks to months later). Early it is usually related to rhegmatogenous retinal detachment or
complications are frequent but can usually be treated extensive retinal ischemia (Fig. 135.14a).92,126 Surgery for the
successfully. The cornea of the diabetic individual is susceptible retinal detachment with extensive panretinal photocoagulation
to injury at surgery, and epithelial defects may be difficult to is indicated. Anterior hyaloidal fibrovascular proliferation is a
heal, requiring pressure patching and, occasionally, bandage serious complication of diabetic vitrectomy and is most
contact lens for resolution.126 Cataract formation may occur commonly observed in juvenile diabetics with severe retinal
from direct lens trauma at the time of vitrectomy or from ischemia undergoing vitrectomy without lens removal.146,147 In
exposure to intraocular gas postoperatively.92,126 Lens removal this situation, extensive neovascularization develops posterior
should be performed, if visually significant, after the posterior to the lens and along the anterior hyaloid (Fig. 135.14b). Unless
segment is stable, or sooner if significant complications arise surgical intervention, including lensectomy and extensive
from lens-induced glaucoma or inflammation. peripheral retinal ablation, is performed promptly, these eyes
Postoperative hemorrhage usually resolves spontaneously are invariably lost.
over several weeks. If the fundus is obscured, ultrasound should
be performed periodically to exclude retinal detachment.92,126 If INTRAVITREAL INJECTION OF
the blood does not clear within several weeks or if there is a ANTIANGIOGENIC AGENTS
concurrent retinal detachment, reoperation is indicated. Ghost
cell glaucoma is an infrequent complication but usually requires Much recent attention has focused on the role of intravitreal
a washout procedure to control the intraocular pressure.92 injections of antiangiogenic agents in the treatment of a number
Retinal detachment after vitrectomy may result from unrecog- of retinal conditions, including age-related macular degenera- 1823
RETINA AND VITREOUS

tion, diabetic macular edema, and diabetic PDR. There are a its effects are less well-established regarding proliferative
number of potential complications of intraocular injections, disease.151 Some surgeons have advocated the intraoperative use
including endophthalmitis, retinal tears/detachments, vitreous of triamcinolone acetonide in order to improve staining and
hemorrhage, cataract, hypotony, and glaucoma. In spite of these visualization of the vitreous to facilitate the identification and
risks, however, a review of the literature indicates that the risk removal of the posterior hyaloid face.
of a serious adverse event from an intravitreal injection is relatively Anti-VEGF agents that have been used in the treatment of
low. Jager et al analyzed data from 14 866 intravitreal injections PDR include the nonselective VEGF inhibitors ranibizumab
in 4382 eyes and found a prevalence of 0.2% per injection and and bevacizumab as well as pegaptanib sodium which is a
0.5% per eye of endophthalmitis after excluding cases reported targeted aptamer against the VEGF isoform 165. Although there
specifically as pseudoendophthalmitis.148 Guidelines for proper is extensive data on these agents in the treatment of choroidal
technique in intravitreous injections are still evolving,149 neovascularization from age-related macular degeneration, the
although certain procedures, including the routine use of a wire literature on their use in treatment of PDR is limited to case
lid speculum, sterile preparation with povidone iodine, and the reports and series. Nonetheless, the anecdotal evidence that
avoidance of a paracentesis have gained widespread acceptance exists suggests that anti-VEGF therapy is effective in at least
as means toward reducing the risk of endophthalmitis. some patients in regressing iris and retinal neovascularization
Intravitreal steroids have.been used as adjuncts to both PRP and reducing vitreous hemorrhage.152,153 The Macugen Diabetic
and vitrectomy for PDR. Although there are no large-scale, Retinopathy Study Group reported that in patients with PDR
randomized, placebo-controlled clinical trials in the literature to who were enrolled in a study of pegaptanib sodium for diabetic
SECTION 10

address the efficacy of intravitreal steroids for retinal neovascu- macular edema, 62% (eight of 13) of patients treated with
larization, the available case reports suggest that intraocular pegaptanib demonstrated regression of neovascularization on
steroids are well tolerated at least in the short term.150 Well- fundus photographs and/or decreased fluorescein leakage from
documented complications of long-term steroid administration their neovascularization. None of the fellow, nonstudy eyes or
include cataract and glaucoma. Although intraocular steroid eyes randomized to sham injection showed a similar regression
often has a beneficial, if temporary effect on macular edema, of neovascularization.154

REFERENCES
1. Waite J, Beetham W: The visual mechanism Diabetes Mellitus. 12th edn. Philadelphia: 19. Montesano R, Vassali JD, Baird A, et al:
in diabetes mellitus: comparative study of Lea & Febiger; 1985. Basic fibroblast growth factor induces
2002 diabetics and 457 non-diabetics for 8. Tolentino MJ, Miller JW, Gragoudas ES, angiogenesis in vitro. Proc Natl Acad Sci
control. N Engl J Med 1935; 212:367–429. et al: Intravitreous injections of vascular USA 1986; 83:7297–7301.
2. The Diabetic Retinopthay Study Research endothelial growth factor produce retinal 20. Lobb RR, Alderman EM, Fett JW: Induction
Group: Photocoagulation treatment of ischemia and microangiopathy in an adult of angiogenesis by bovine brain derived
proliferative diabetic retinopathy: the primate. Ophthalmology 1996; class I heparin-binding growth factor.
second report of Diabetic Retinopathy 103:1820–1828. Biochemistry 1985; 24:4969–4973.
Study findings. Ophthalmology 1978; 85:82. 9. Amin R, Frank R, Kennedy A, et al: Vascular 21. Shing Y, Folkman J, Haudenschild C, et al:
3. The Diabetic Retinopathy Study Group: endothelial growth factor is present in glial Angiogenesis is stimulated by a tumor-
Preliminary report on effects of cells of the retina and optic nerve of human derived endothelial cell growth factor. J Cell
photocoagulation therapy. Am J Ophthalmol subjects with nonproliferative diabetic Biochem 1985; 29:275–287.
1976; 81:383–396. retinopathy. Invest Ophthalmol Vis Sci 1997; 22. Esch F, Ueno N, Baird A, et al: Primary
3a. World Health Organization. Diabetes fact 38:36–47. structure of bovine brain acidic fibroblast
sheet. Available: http://www.who.int/ 10. Pascal DI, Bindley CO, Wallow IHL: growth factor. Biochem Biophys Res
hpr/NPH/docs/gs_diabetes.pdf#search=%2 Clinicopathologic correlation of intraretinal Commun 1986; 133:554–562.
2370%20million%20diabetes%20world%2 microvascular abnormalities. Retina 1997; 23. Gospodarowicz D: Fibroblast growth
0health%20organization%22 30 Sept 17:321–329. factors. In: Pimental E, Perucho M, eds.
2006. 11. Chang S, Leonard-Martin T, Feman S: Critical Reviews in Oncogenesis. Boca
4. Klein R, Klein B: Vision disorders in Relationship between IRMA and diabetic Raton, FL: CRC; 1989:1–26.
diabetes. In: Harris M, Hamman R, eds. neovascularization. Invest Ophthalmol Vis 24. Abraham JA, Mergia A, Whang JL, et al:
Diabetes Data Compiled 1984, p XIII. Sci 1995; 36:S483. Nucleotide sequence of a bovine clone
Washington, DC: US Government Printing 13. Ashton N: Retinal vascularization in health encoding the angiogenic protein, basic
Office; 1985. and disease. Am J Ophthalmol 1957; fibroblast growth factor. Science 1986;
4a. Aiello LP: Angiogenic pathways in diabetic 44:7–24. 233:545–548.
retinopathy. N Engl J Med 2005; 14. Kohner E, Schilling J, Hamileton A: The role 25. D’Amore PA, Klagsbrun M: Endothelial
353:839–841. of avascular retina in new vessel formation. mitogens derived from retina and
4b. Bursell SE, Clermont AC, Kinsley BT, et al: Metab Ophthalmol 1976; 1:15. hypothalamus: Biological and biochemical
Retinal blood flow changes in patients with 15. Wise G: Retinal neovascularization. Trans similarities. J Cell Biol 1984; 99:1545–1549.
insulin-dependent diabetes mellitus and no Am Ophthalmol Soc 1956; 54:729–826. 26. Caruelle D, Groux MB, Gaudric A, et al:
diabetic retinopathy. Invest Ophthalmol Vis 16. Branch Vein Occlusion Study Group: Argon Immunological study of acidic fibroblast
Sci 1996; 37:886–897. laser scatter photocoagulation for prevention growth factor (aFGF) distribution in the eye.
5. Michaelson IC: The mode of development of neovascularization and vitreous J Cell Biochem 1989; 39:117–128.
of the vascular system of the retina, with hemorrhage in branch vein occlusion. Arch 27. Gao H, Hollyfield JG: Basic fibroblast
some observations on its significance for Ophthalmol 1986; 104:34–41. growth factor (bFGF) immunolocalization in
certain retinal disease. Trans Ophthalmol 17. Baird A, Böhlen P: Fibroblast growth the rodent outer retina demonstrated with
Soc UK 1948; 68:137–180. factors. In: Roberts AB, Sporn MB, eds. an anti-rodent bFGF antibody. Brain Res
5. Plehwe W, Sleightholm M, Kohner E, et al: Handbook of Experimental Pharmacology: 1992; 585:355–360.
Does vitreous fluorophotometry reflect Peptide Growth Factors and Their 28. Nyberg F, Hahnenberger R, Jakobson AM,
severity of early diabetic retinopathy? Br J Receptors. Berlin: Springer-Verlag; et al: Enhancement of FGF-like
Ophthalmol 1989; 73:225–260. 1990:369–418. polypeptides in the retinae of newborn mice
6. Frank R: Diabetic retinopathy: current 18. Mignatti P, Tsuboi R, Robbins E, et al: In exposed to hyperoxia. FEBS Lett 1990;
concepts of evaluation and treatment. Clin vitro angiogenesis on the human amniotic 267:75–77.
Endocrinol Metab 1986; 15:933–969. membrane: requirement for basic fibroblast 29. Zhang NL, Samadani EE, Frank RN:
7. Aiello L, Li R, Sebestyen J, et al: The eyes growth factor-induced proteinases. J Cell Mitogenesis and retinal pigment epithelial
1824 and diabetes. In: Marble A, ed. Joslin’s Biol 1989; 108:671–682. cell antigen expression in the rat after
Proliferative Diabetic Retinopathy

krypton laser photocoagulation. Invest endothelial growth factor in ocular fluid of 56. Janka H, Warram J, Rand L, et al: Risk
Ophthalmol Vis Sci 1993; 34:2412–2424. patients with diabetic retinopathy and other factors for progression of background
30. Sivalingam A, Kenney J, Brown GC, et al: retinal disorders. N Engl J Med 1994; retinopathy in long-standing IDDM.
Basic fibroblast growth factor levels in the 331:1480–1487. Diabetes 1989; 38:460–464.
vitreous of patients with proliferative 47. Adamis AP, Miller JW, Bernal MT, et al: 57. Baker R, Rand L, Krolewski A, et al:
diabetic retinopathy. Arch Ophthalmol Elevated vascular permeability Influence of HLA-DR phenotype and
1990; 108:869–872. factor/vascular endothelial growth factor myopia on the risk of nonproliferative and
31. Vlodavsky I, Folkman J, Sullivan R, et al: levels in the vitreous of eyes with proliferative diabetic retinopathy. Am J
Endothelial cell-derived basic fibroblast proliferative diabetic retinopathy. Am J Ophthalmol 1986; 102:693–700.
growth factor: Synthesis and deposition Ophthalmol 1994; 118:445–450. 58. Rand L, Krolewski A, Aiello L, et al: Multiple
into subendothelial extracellular matrix. 48. Malecaze F, Clamens S, Simorre-Pinatel V, factors in the prediction of risk of
Proc Natl Acad Sci USA 1987; et al: Detection of vascular endothelial proliferative diabetic retinopathy. N Engl J
84:2292–2296. growth factor messenger RNA and vascular Med 1985; 313:1433–1438.
32. McNeil PL, Muthukrishnan L, Warder E, endothelial growth factor-like activity in 59. Rand L, Krolewski A, Warram J: Late
et al: Growth factors are released by proliferative diabetic retinopathy. Arch complications: The critical period. In:
mechanically wounded endothelial cells. Ophthalmol 1994; 112:1476–1482. Friedman E, L’Esperance A, eds. Diabetic
J Cell Biol 1989; 109:811–822. 49. Tolentino M, Miller J, Gragoudas E, et al: Renal-Retinal Syndrome. New York: Grune
33. LeRoith D, Roberts CT Jr: Insulin-like Vascular endothelial growth factor is & Stratton; 1986:3.
growth factors. Ann N Y Acad Sci 1993; sufficient to produce iris neovascularization 60. Orchard T, Dorman J, Maser R, et al:
692:1–9. and neovascular glaucoma in a nonhuman Factors associated with avoidance of

CHAPTER 135
34. Salmon WD Jr, Daughaday WH: A primate. Arch Ophthalmol 1996; severe complications after 25 yr of IDDM.
hormonally controlled serum factor which 114:964–970. Pittsburgh Epidemiology of Diabetes
stimulates sulfate incorporation by cartilage 50. Okamoto N, Tobe T, Hackett S, et al: Complications Study I. Diabetes Care
in vitro. J Lab Clin Med 1957; 49:825–836. Transgenic mice with increased expression 1990; 13:741–747.
35. Poulsen JE: The Houssay phenomenon in of vascular endothelial growth factor in the 61. Kroc Collaborative Study Group: Blood
man. Recovery from retinopathy in a case retina: a new model of intraretinal and glucose control and the evolution of
of diabetes with Simmonds’ disease. subretinal neovascularization. Am J Pathol diabetic retinopathy and albuminuria:
Diabetes 1953; 2:7–12. 1997; 151:281–291. A preliminary multicenter trial. N Engl J
36. Grant M, Jerdan J, Merimee TJ: Insulin-like 50a. Meier M, King GL: Protein kinase C Med 1984; 311:365–372.
growth factor-I modulates endothelial cell activation and its pharmacological 62. Dahl-Jorgensen K, Brinchmann-Hanses O,
chemotaxis. J Clin Endocrinol Metab 1987; inhibition in vascular disease. Vascular Med Hanssen K, et al: Effect of near
65:370–371. 2000; 5:173–185. normoglycemia for two years on
37. Grant MB, Mames RN, Fitgerald C, et al: 50b. Williams B: Factors regulating the progression of early diabetic retinopathy,
Insulin-like growth factor I as an angiogenic expression of vascular nephropathy, and neuropathy: The Oslo
agent. In vivo and in vitro studies. Ann N Y permeability/vascular endothelial growth Study. BMJ 1986; 293:1195–1199.
Acad Sci 1993; 692:230–242. factor by human vascular tissues. 63. The DCCT Research Group: Diabetes
38. Grant M, Russell B, Fitgerald C, et al: Diabetologia 40: S118–S120, 1997 Control and Complications Trial (DCCT):
Insulin-like growth factors in vitreous. 50c. Ishii H, Jirousek MR, Koya D et al: Results of feasibility study. Diabetes Care
Studies in control and diabetic subjects Amerlioration of vascular dysfunctions in 1987; 10:1–19.
with neovascularization. Diabetes 1986; diabetic rats by an oral PKC inhibitor. 64. Rosenlund E, Hasskens K, Brinchmann-
35:416–420. Science 1996; 272:728–731 Hanses O, et al: Transient proliferative
39. Miller J, Adamis A, Aiello L: Vascular 50d. Aiello LP, Bursell SE, Clermont A, et al: diabetic retinopathy during intensified
endothelial growth factor in ocular Vascular endothelial growth factor-induced insulin treatment. Am J Ophthalmol 1988;
neovascularization and proliferative diabetic retinal permeability is mediated by protein 105:618–625.
retinopathy. Diabetes Metab Rev 1997; kinase C in vivo and suppressed by an 65. Dandona P, Bolger J, Boag F, et al: Rapid
13:37–50. orally effective beta-isoform-selective development and progression of
40. Miller JW, Adamis AP, Shima D, et al: inhibitor. Diabetes 1997; 46:1473–1480 proliferative retinopathy after strict diabetic
Vascular endothelial growth factor/vascular 50e. Suzuma K, Takahara N, Suzuma I, et al: control. BMJ 1985; 290:895–896.
permeability factor is temporally and Characterization of protein kinase C 66. Zinman B: The physiologic replacement of
spatially correlated with ocular isoform’s action on retinoblastoma protein insulin, an elusive goal. N Engl J Med 1989;
angiogenesis in a primate model. Am J phosphorylation, vascular endothelial 321:363–370.
Pathol 1994; 145:574–584. growth factor-induced endothelial cell 67. The Diabetes Control and Complications
41. Pierce EA, Avery R, Foley E, et al: Vascular proliferation, and retinal neovascularization. Trial Research Group: The effect of
endothelial growth factor/vascular Proc Natl Acad Sci USA 2002; 99:721–726. intensive treatment of diabetes on the
permeability factor expression in a mouse 51. Tagawa H, McMeel J, Furukawa H, et al: development and progression of long-term
model of retinal neovascularization. Proc Role of the vitreous in diabetic retinopathy. complications in insulin-dependent diabetes
Natl Acad Sci USA 1995; 92:905–909. I. Vitreous changes in diabetic retinopathy mellitus. N Engl J Med 1993; 329:977–986.
42. Dorey C, Aouididi S, Reynaud X, et al: and in physiologic aging. Ophthalmology 68. The Diabetes Control and Complications
Correlation of vascular permeability 1986; 93:596–601. Trial Research Group: Progression of
factor/vascular endothelial growth factor 52. Foos R, Kreiger A, Forsythe A, et al: retinopathy with intensive versus
with extraretinal neovascularization in the Posterior vitreous detachment in diabetic conventional treatment in the Diabetes
rat. Invest Ophthalmol Vis Sci 1996; subjects. Ophthalmology 1980; Control and Complications Trial.
114:1210–1217. 87:122–128. Ophthalmology 1995; 102:647–661.
43. Stone J, Chan-Ling T, Pe’er J, et al: Roles 53. Klein R, Klein B, Moss S, et al: Diabetic 69. Cheng H, Franklin S: Treatment of cataract
of vascular endothelial growth factor and retinopathy. II. Prevalence and risk of in diabetics with and without retinopathy.
astrocyte degeneration in the genesis of diabetic retinopathy when age at diagnosis Eye 1988; 2:607–614.
retinopathy of prematurity. Invest is less than 30 years. Arch Ophthalmol 70. Jaffe G, Burton T: Progression of
Ophthalmol Vis Sci 1996; 37:290–299. 1984; 105:520–526. nonproliferative diabetic retinopathy
44. Adamis A, Shima D, Tolentino M, et al: 54. Klein R, Klein B, Moss S, et al: The following cataract extraction. Arch
Inhibition of VEGF prevents ocular Wisconsin Epidemiologic Study of Diabetic Ophthalmol 1988; 106:745–749.
neovascularization in a non-human primate. Retinopathy. III. Prevalence and risk of 71. Benson W, Brown G, Tasman W, et al:
Arch Ophthalmol 1996; 114:66–71. diabetic retinopathy when age at diagnosis Extracapsular cataract extraction with
45. Aiello L, Pierce E, Foley E, et al: Inhibition is 30 or more years. Arch Ophthalmol 1984; placement of a posterior chamber lens in
of vascular endothelial growth factor 102:527–532. patients with diabetic retinopathy.
suppresses retinal neovascularization in 55. Aiello L, Rand L, Briones J, et al: Diabetic Ophthalmology 1993; 100:730–738.
vivo. Proc Natl Acad Sci USA 1995; retinopathy in Joslin Clinic patients with 72. Sebestyen J: Introcular lenses and diabetes
92:10457–10461. adult-onset diabetes. Ophthalmology 1981; mellitus. Am J Ophthalmol 1986;
46. Aiello L, Avery R, Arrigg P, et al: Vascular 88:619–623. 101:425–428. 1825
RETINA AND VITREOUS

73. Alpar J: Diabetes: Cataract extraction and 92. Charles S: Vitreous microsurgery. factor regression after laser panretinal
intraocular lenses. J Cataract Refract Surg Baltimore: Williams & Wilkins; photocoagulation for proliferative diabetic
1987; 13:43–46. 1981:107–120. retinopathy. Ophthalmology 1984;
74. Aiello L, Wand M, Liang G: Neovascular 93. Packer A: Vitrectomy for progressive 91:1453–1457.
glaucoma and vitreous hemorrhage macular traction associated with 110. Vine A: The efficacy of additional argon
following cataract surgery in patients with proliferative diabetic retinopathy. Arch laser photocoagulation for persistent,
diabetes mellitus. Ophthalmology 1983; Ophthalmol 1987; 105:1679–1682. severe proliferative diabetic retinopathy.
90:814–820. 94. Poulsen J: Diabetes and anterior pituitary Ophthalmology 1985; 92:1532–1537.
75. Hykin P, Gregson R, Stevens J, et al: insufficiency. Final course and postmortem 111. Blankenship G: Red-krypton and blue-
Extracapsular cataract extraction in study of a diabetic patient with Sheehan’s green argon panretinal laser
proliferative diabetic retinopathy. syndrome. Diabetes 1966; 15:73–77. photocoagulation for proliferative diabetic
Ophthalmology 1993; 100:394–399. 95. Luft R, Olivecrona H, Ikkos D, et al: retinopathy: A laboratory and clinical
76. Lischwe T, Ide C: Predicting visual acuity Hypophysectomy in man: further comparison. Trans Ophthalmol Soc UK
after cataract surgery using the blue field experiences in severe diabetes mellitus. 1986; 134:967–1003.
entoptoscope and projected slides. BMJ 1955; 2:752-756. 112. Shea M: Early vitrectomy in proliferative
Ophthalmology 1988; 95:256–260. 96. Sharp P, Fallon T, Brazier O, et al: Long-term diabetic retinopathy. Arch Ophthalmol
77. Beetham W: Diabetic retinopathy in follow-up of patients who underwent yttrium- 1983; 101:1204–1205.
pregnancy. Trans Ophthalmol Soc UK 90 pituitary implantation for treatment of 113. D’Amico D: Diabetic traction retinal
1950; 48:205–216. proliferative diabetic retinopathy. Diabetologia detachments threatening the fovea and
78. Rodman H, Singerman LJ, Aiello L, et al: 1987; 30:199–207. panretinal argon laser photocoagulation.
Diabetic retinopathy and its relationship to 97. Beetham W, Aiello L, Balodimos M, et al: Semin Ophthalmol 1991; 6:11–18.
SECTION 10

pregnancy. In: Merkaty T, Adams P, eds. Ruby laser photocoagulation of early 114. Early Treatment Diabetic Retinopathy Study
The Diabetic Pregnancy: A Perinatal diabetic neovascular retinopathy. Arch Group: Early photocoagulation for diabetic
Perspective. New York: Grune & Stratton; Ophthalmol 1970; 83:261–272. retinopathy. Ophthalmology 1991;
1979. 98. Beetham W: Visual prognosis of 98:766–785.
79. Laatikainen L, Larinkari J, Teramo K, et al: proliferating diabetic retinopathy. Br J 115. Ferris F: Effect of Scatter
Occurrence and prognostic significance of Ophthalmol 1963; 47:611–619. Photocoagulation: Type I vs. Type II
retinopathy in diabetic pregnancy. Metab 98. Meyer-Schwickerath G, Schott K: Diabetic Diabetes. In: Vitreoretinal Frontiers. 21st
Pediatr Ophthalmol 1980; 4:191–195. retinopathy and photocoagulation. Am J Century. San Francisco: American
80. Klein B, Moss S, Klein R: Effect of Ophthalmol 1968; 66:597–603. Academy of Ophthalmology; 1996:93.
pregnancy on progression of diabetic 100. Aiello L, Beetham W, Balodimos M, et al: 116. Blondeau P, Pavan P, Phelps C: Acute
retinopathy. Diabetes Care 1990; 13:34–40. Ruby laser photocoagulation in treatment pressure elevation following panretinal
81. Laatikainen L, Teramo K, Hieta- of diabetic proliferating retinopathy: photocoagulation. Arch Ophthalmol 1981;
Heikeraninen, et al: A controlled study of Preliminary report. In: Goldberg M, Fine S, 99:1239–1241.
the influence of continuous subcutaneous eds. Symposium on the Treatment of 117. Lobes L, Bourgon P: Pupillary
insulin infusion treatment on diabetic Diabetic Retinopathy. Washington, DC: abnormalities induced by argon laser
retinopathy during pregnancy. Acta Med Public Health Service; 1969:437. photocoagulation. Ophthalmology 1985;
Scand 1987; 221:367–376. 101. Campbell C, Koester C, Curtice V, et al: 92:234–236.
82. Klein B, Klein R, Meur S, et al: Does the Clinical studies in laser photocoagulation. 117a. Shimura M, Yasuda K, Nakazawa T, et al:
severity of diabetic retinopathy predict Arch Ophthalmol 1965; 74:57–65. Quantifying alterations of macular thickness
pregnancy outcome? J Diabetes 102. The Diabetic Retinopathy Study Research before and after panretinal
Complications 1988; 2:179–184. Group: Four risk factors for severe visual photocoagulation in patients with severe
83. Janka H, Ziegler A, Valsania P, et al: Impact loss in diabetic retinopathy. The third report diabetic retinopathy and good vision.
of blood pressure on diabetic retinopathy. from the DRS. Arch Ophthalmol 1979; Ophthalmology 2003; 110:2386–2394.
Diabetes Metab 1989; 15:333–337. 97:654–655. 118. Pender P, Benson W, Compton H, et al:
84. Chase H, Garg S, Jackson W, et al: Blood 103. Doft B, Blankenship G: Single versus The effects of panretinal photocoagulation
pressure and retinopathy in type I diabetes. multiple treatment sessions of argon laser on dark adaptation in diabetics with
Ophthalmology 1990; 97:155–159. panretinal photocoagulation for proliferative proliferative retinopathy. Ophthalmology
85. Krolewski A, Canessa M, Warram J, et al: diabetic retinopathy. Ophthalmology 1982; 1981; 88:635–638.
Predisposition to hypertension and 89:772–779. 119. Kleiner R, Elman M, Murphy R, et al:
susceptibility to renal disease in insulin- 104. The Diabetic Retinopathy Study Research Transient severe visual loss after panretinal
dependent diabetes mellitus. N Engl J Med Group: Photocoagulation treatment of photocoagulation. Am J Ophthalmol 1988;
1988; 318:140–145. proliferative diabetic retinopathy. Clinical 106:298–306.
86. Klein R: Recent developments in the application of DRS findings. 120. Wallow I, Johns K, Barry P, et al:
understanding and management of diabetic Ophthalmology 1981; 88:583–600. Chorioretinal and choriovitreal
retinopathy. Med Clin North Am 1988; 105. The Diabetic Retinopathy Study Research neovascularization after photocoagulation
72:1415–1437. Group: Photocoagulation treatment of for proliferative diabetic retinopathy.
87. Petersen M, Vine A: The University of proliferative diabetic retinopathy: Ophthalmology 1985; 92:523–532.
Michigan Pancreas Transplant Evaluation Relationship of adverse treatment effects to 121. Swartz M, Apple D, Creel D: Sudden
Committee: Progression of diabetic retinopathy severity. Dev Ophthalmol 1981; severe visual loss associated with
retinopathy after pancreas transplantation. 2:248–261. peripapillary burns during panretinal argon
Ophthalmology 1990; 97:496–500. 106. Okun E, Johnston G, Boniuk I, et al: Xenon photocoagulation. Br J Ophthalmol 1983;
88. Ramsay R, Goetz F, Sutherland D, et al: arc photocoagulation of proliferative 67:517–519.
Progression of diabetic retinopathy after diabetic retinopathy. a review of 2688 122. McCanna R, Chandra S, Stevens T, et al:
pancreas transplantation for insulin- consecutive eyes in the format of the Argon laser-induced cataract as a
dependent diabetes mellitus. N Engl J Med Diabetic Retinopathy Study. Ophthalmology complication of retinal photocoagulation.
1988; 318:208–214. 1984; 91:1458–1463. Arch Ophthalmol 1982; 100:1071–1073.
89. The Diabetic Retinopathy Study Research 107. Plumb A, Swan A, Chignell A, et al: 123. Lakhanpal V, Schocket S, Richards R, et al:
Group: Design, methods, and baseline A comparative trial of xenon arc and argon Photocoagulation-induced lens opacity.
results. Invest Ophthalmol Vis Sci 1981; laser photocoagulation in the treatment of Arch Ophthalmol 1982; 100:1068–1070.
21:149–226. proliferative diabetic retinopathy. Br J 124. Machemer R: Vitrectomy: A Pars Plana
90. Davis M: Proliferative diabetic retinopathy. Ophthalmol 1982; 66:213–218. Approach. New York: Grune & Stratton;
In: Ryan S, ed. Retina. St Louis: Mosby; 108. Blankenship G: A clinical comparison of 1975.
1989:367–402. central and peripheral argon laser 125. Aaberg T, Abrams G: Changing indications
91. Early Treatment of Diabetic Retinopathy panretinal photocoagulation for proliferative and techniques for vitrectomy in
Study Group: Effects of aspirin treatment diabetic retinopathy. Ophthalmology 1988; management of complications of diabetic
on diabetic retinopathy. Ophthalmology 95:170–177. retinopathy. Ophthalmology 1987;
1826 1991; 98:107–120. 109. Doft B, Blankenship G: Retinopathy risk 94:775–779.
Proliferative Diabetic Retinopathy

126. Michels R: Proliferative diabetic vitreous hemorrhage. Br J Ophthalmol 146. Lewis H, Abrams G, Williams G: Anterior
retinopathy: pathophysiology of extraretinal 1976; 60:752–755. hyaoidal fibrovascular proliferation after
complications and principles of vitreous 136. Barrie T, Feretis E, Leaver P, et al: Closed diabetic vitrectomy. Am J Ophthalmol
surgery. Retina 1981; 1:1–17. microsurgery for diabetic traction macular 1987; 104:607–613.
127. The Diabetic Retinopathy Vitrectomy Study detachment. Br J Ophthalmol 1982; 147. Lewis H, Abrams G, Foos R:
Research Group: Early vitrectomy for 66:754–758. Clinicopathologic findings in anterior
severe vitreous hemorrhage in diabetic 137. Williams D, Williams G, Hartz A, et al: hyaloidal fibrovascular proliferation after
retinopathy. Arch Ophthalmol 1985; Results of vitrectomy for diabetic traction diabetic vitrectomy. Am J Ophthalmol
103:1644–1652. retinal detachments using the en bloc 1987; 104:614–618.
128. The Diabetic Retinopathy Vitrectomy Study excision technique. Ophthalmology 1989; 148. Jager RD, Aiello LP, Patel SC, et al: Risks
Group: Two-year course of visual acuity in 96:752–758. of intravitreous injection: a comprehensive
severe proliferative diabetic retinopathy 138. Aaberg T: Clinical results in vitrectomy for review. Retina 2004; 24:676–698.
with conventional management. diabetic traction retinal detachment. Am J 149. Aiello LP, Brucker AJ, Chang S, et al:
Ophthalmology 1985; 92:492–502. Ophthalmol 1979; 88:246–253. Evolving guidelines for intravitreous
129. Koenig S, Mieler W, Han D, et al: 139. Hutton W, Bernstein I, Fuller D: Diabetic injections. Retina 2004; 24:S3–S19.
Combined phacoemulsification, pars plana traction retinal detachment: Factors 150. Bandello F, Polito A, Pognuz DR, et al:
vitrectomy, and posterior chamber influencing final visual acuity. Triamcinolone as adjunctive treatment to
intraocular lens insertion. Arch Ophthalmol Ophthalmology 1980; 87:1071–1077. laser panretinal photocoagulation for
1992; 110:1101–1104. 140. Tolentino F, Freeman H, Tolentino F: Closed proliferative diabetic retinopathy. Arch
130. Senn P, Schipper I, Perren B: Combined vitrectomy in the management of diabetic Ophthalmol 2006; 124:643–650.

CHAPTER 135
pars plana vitrectomy, phacoemulsification, traction retinal detachment. Ophthalmology 151. Jonas JB, Sofker A, Degenring R:
and intraocular lens implantation in the 1980; 87:1078–1089. Intravitreal triamcinolone acetonide as an
capsular bag: a comparison to vitrectomy 141. Rice T, Michels R, Rice E: Vitrectomy for additional tool in pars plana vitrectomy for
and subsequent cataract surgery as a two- diabetic traction retinal detachment proliferative diabetic retinopathy. Eur J
step procedure. Ophthalmic Surg Lasers involving the macula. Am J Ophthalmol Ophthalmol 2003; 13:468–473.
1995; 26:420–428. 1983; 95:22–33. 152. Oshima Y, Sakaguchi H, Gomi F, et al:
131. McCuen BD, Rinkoff J: Silicone oil for 142. Rice T, Michels R, Rice E: Vitrectomy for Regression of iris neovascularization after
progressive ocular neovascularization after diabetic rhegmatogenous retinal detachment. intravitreal injection of bevacizumab in
failed diabetic vitrectomy. Arch Ophthalmol Am J Ophthalmol 1983; 95:34–44. patients with proliferative diabetic
1989; 107:677–682. 143. Miller S, Shafrin G, Bresnick G, et al: retinopathy. Am J Ophthalmol 2006;
132. Abrams G, Williams G: “En bloc” excision Scleral buckling for diabetic retinal 142:155–158.
of diabetic membranes. Am J Ophthalmol detachments secondary to proliferative 153. Spaide RF, Fisher YL: Intravitreal
1987; 103:302–308. diabetic retinopathy. Am J Ophthalmol bevacizumab (Avastin) treatment of
133. Mandelcorn M, Blankenship G, Machemer 1980; 89:103–112. proliferative diabetic retinopathy
R: Pars plana vitrectomy for the 144. Thompson J, Auer C, de Bustros S, et al: complicated by vitreous hemorrhage.
management of severe diabetic retinopathy. Prognostic indicators of success and failure Retina 2006; 26:275–278.
Am J Ophthalmol 1976; 81:561–570. in vitrectomy for diabetic retinopathy. 154. Adamis AP, Altaweel M, Bressler NM, et al;
134. Michels R, Rice T, Tice E: Vitrectomy for Ophthalmology 1986; 93:290–295. Macugen Diabetic Retinopathy Study
diabetic vitreous hemorrhage. Am J 145. Blankenship G, Machemer R: Long-term Group: Changes in retinal
Ophthalmol 1983; 95:12–21. diabetic vitrectomy results: Report of neovascularization after pegaptanib
135. Peyman G, Raichand M, Huamonte F, et al: 10-year follow-up. Ophthalmology 1985; (Macugen) therapy in diabetic individuals.
Vitrectomy in 125 eyes with diabetic 92:503–506. Ophthalmology 2006; 113:23–28.

1827
CHAPTER

136 Advanced Retinopathy of Prematurity


Peter Hovland, Shizuo Mukai, and Tatsuo Hirose

INTRODUCTION ments posterior to the ridge can proceed to total retinal detach-
ment within a day, and total detachments can become closed
Retinopathy of prematurity (ROP) – the proliferation of funnels within a week in very active vasoproliferative ROP.
abnormal retinal blood vessels that occurs in premature infants Peripheral retinal cryoablation during the acute phase of ROP
and its pathologic sequelae – has been and continues to be an appears to reduce by half the incidence of childhood RD.3
important cause of childhood blindness. Since 1942 when ROP Despite prophylactic ablative therapy RD can occur. Hartnett
was first recognized by Terry,1,2 advances have been made both and McColm report4 that factors associated with progression to
in understanding the etiology and characterizing the stage 4 RD following laser ablative therapy included the absence
pathological progression of ROP, as well as establishing inter- of clear vitreous, ridge elevation of at least six clock hours,
ventions timed to prevent or even reverse visual loss. Progress and plus disease encompassing two or more quadrants.
in the field has been based on key observations, well designed In contrast, exudative retinal detachments occur less
studies, and improvements in surgical techniques and commonly in acute ROP. These occur as a result of plasma
technology. leakage from abnormal neovascular tufts, with subsequent
The pathophysiology, diagnosis and management of acute subretinal fluid accumulation. Chronic vitreoretinal traction to
ROP are covered extensively in Chapter 318. In this chapter, the the retinal vessels may play some role in causing exudation.
concentration will be on advanced stages of ROP, including Exudative RD may occur more frequently in the adult patient
retinal detachment and preretinal fibrovascular proliferation, with ROP. The fundus usually shows smooth retinal
and their management. detachment with yellow subretinal exudates. Abnormal vessels
such as small angiomas with retinal hemorrhage can be seen.
RETINAL DETACHMENT IN ROP Focal and diffuse pigmentary changes indicate the chronic
nature of the detachment. Chronic exudation may result in
anterior segment pathology (ischemia, cataract, neovasculari-
Key Features: Forms of Retinal Detachment zation) and posterior segment dysfunction by exudative RD or
Tractional RD – Most common form observed in acute ROP macular edema. Shallow exudative detachments of avascular
Exudative RD – More common in chronic forms retina anterior to the ridge do not normally require surgical
Rhegmatogenous RD – rarely noted in acute ROP. May be intervention.5 The abnormal neovascular tufts may be treated
iatrogenic
with cryotherapy to reduce fluid leakage (Fig. 136.1). Newer
therapies such as intravitreal antivascular endothelial growth
Patients with ROP have an increased lifetime risk for various factor (anti-VEGF) compounds, which decrease vessel perme-
forms of retinal detachment. In acute ROP, the tractional RD is ability may be of some use in treating this condition in
most commonly observed. Tractional RDs originate at the ridge, the adult.
at which point myofibroblasts pull centripetally and anteriorly Rhegmatogenous detachments are seen only rarely in acute
toward the lens in a purse-string configuration. Once retinal ROP and are usually iatrogenic at this age. In contrast, they are
detachment occurs, it can progress rapidly. Peripheral detach- the most common retinal detachment seen in older children

FIGURE 136.1. (a) Fundus photograph of


stage 3 moderate retinopathy of prematurity.
A large ridge with extraretinal fibrovascular
proliferation is visible. Peripheral to this ridge is
a large area of avascular retina. (b) Fundus
photograph of the same eye immediately after
ablation of the avascular retina anterior to the
ridge by diode laser coagulation. In this case,
laser coagulation was applied in three to four
contiguous rows anterior to the ridge without
treating all the way up to the ora serrata.
Treatment was as effective as cryotherapy.

a b
1829
RETINA AND VITREOUS

and adults with cicatricial ROP. During early adolescence, there remains disagreement as to whether to cut or remove the
patients may be at an increased risk of developing RD.6 Many band at some time after scleral buckling surgery. Machemer and
of these cases are long-standing and are not noticed by the deJuan17 do not routinely remove the band unless there is cli-
patient until subretinal fluid expands into a large enough area nically apparent retardation of ocular growth. Greven and
to produce visual symptoms. The detached retina is usually Tasman15 also leave the band in place in most cases. McPherson
thin, with multiple small equatorial breaks hidden by the and coworkers18 and Orellana5 state that the band ideally
vitreous membrane. These late-occurring rhegmatogenous should be transected 3–6 months postoperatively to permit
detachments may be repaired with scleral buckle (SB) or normal ocular growth.
vitrectomy techniques,7,8 though successful repair of late-
occurring RD is more likely to require several procedures.9
Because of the increased risk of developing RD, regular eye PROPHYLACTIC SB
examinations are recommended in high-risk or preverbal The uniform loss of macular vision in infants who have
children with a history of ROP.8 undergone therapeutic scleral buckling for stage 4b or stage 5
ROP has been noted by Mintz-Hittner and Kretzer.21 They
SCLERAL BUCKLING proposed two reasons for the poor outcome in these infants.
First, rapid ocular growth occurs after development of ROP in
Scleral buckling can be used to treat milder forms of traction these infants, whereas the area of vascularized retina does not
retinal detachment in acute ROP.10–13 Retinal detachment in increase. The distance between the temporal optic disk and the
SECTION 10

posterior zone ROP in which the ridge is located very posterior center of the macula remains 4 mm from preterm to adulthood.
to the equator showing a large attached avascular retinal Thus, traction is exaggerated with ocular growth. Second, the
anteriorly can not be treated with a buckle. Beyrau and Danis retinal interaction with retinal pigment epithelium is critical to
reported 14 that primary scleral buckling in 22 eyes of 14 infants normal retinal development. An interruption of the normal
with stage 4a ROP resulted in successful maintenance of configuration for even a few days may result in irreversible
attachment of the macula in 70% initially, and useful vision in dysmorphic changes. These considerations led to the proposal
60%. Greven and Tasman15 reported successful reattachment that these extremely low-birth-weight infants be treated with a
with scleral buckling in 13 of 22 eyes with stage 4b and stage 5 prophylactic SB at the time of cryotherapy for threshold ROP.
(open–open configuration) ROP. More than 50% of patients Macular detachment or ectopia did not develop in 80% of eyes
had satisfactory anatomic results in most other studies.5,16–18 in these infants with zone 1 disease who were treated with a
Prost reports19 that a modified SB encircling procedure was prophylactic SB. This approach to managing ROP has not
used in 121 eyes of 68 infants with stage 5 ROP, resulting in a become widely adopted, however. When the ridge is located
52% total (and 24% partial) reattachment rate. However, only posterior to the equator, as in zone I ROP it is better to approach
20% of the operated eyes were judged by the author to have by vitrectomy.
useful vision. Scleral buckling is also used in the treatment of
rhegmatogenous detachment in ROP eyes.20 VITRECTOMY
The timing or even indication of surgery in the management
of stage 4a detachment is controversial because many cases of Vitrectomy may be considered when scleral buckling fails, a
partial nonrhegmatogenous detachments often reattach spon- high retinal detachment is present, media opacification by
taneously particularly when there is neither active vasopro- vitreous strands or hemorrhage occurs, or a fibrous membrane
liferation nor plus disease. However, since the duration of is observed behind the lens. Vitrectomy is considered as a
retinal detachment influences prognosis, as the retina becomes primary procedure for retinal detachment in posterior zone
atrophic after relatively brief periods of detachment,18 scleral ROP. With advances in surgical approaches to ROP, the
buckling should be considered early in progressive traction retina can be reattached in an apparently unsalvageable eye
detachments and in large exudative detachments. There is less (Fig. 136.2). Optimal results are achieved by meticulous
agreement about surgery in traction detachments that do not removal of proliferative membranes; this may be assisted by
involve the posterior pole. Until a randomized, controlled trial intraoperative injection of triamcinolone into the eye to help in
of surgery for stage 4 ROP is performed, the decision to under- the visualization of the vitreous and membranes. Sometimes
take surgery in these controversial cases must be determined on scleral buckling is combined with vitrectomy surgery.18,22–36
an individual basis. There are two distinct approaches to vitrectomy in advanced
Cryotherapy or laser therapy is often performed at the time ROP: the closed technique which is more commonly used for
of retinal detachment surgery, especially in traction or exudative stage 4a, 4b, and stage 5 when the retina is well visible; and the
cases with continued active neovascularization. Cryotherapy ‘open sky’ method which is typically reserved for stage 5 ROP
can be applied to the active neovascular ridge if the detachment when the retina is pulled well behind the lens, including cases
is shallow. The scleral depression made by a cryoprobe can with leukocoria.
reach the ridge. The laser can be applied on the ridge on the
buckle after external drainage of subretinal fluid and after the
retina is attached. Cryotherapy or laser is unnecessary if there CLOSED VITRECTOMY WITH LENS REMOVAL
is no active neovascularization. When the traction is effectively Charles,22 Machemer and deJuan,17,23 Trese,28 and Fuchino and
released additional treatment to create chorioretinal adhesive associates34 pioneered closed vitrectomy (CV) techniques for
force is not required (athermal buckling). A silicone band is management of ROP. The small size of the eye and extensive
used to encircle the globe along the site of the ridge. External proliferation necessitated several modifications of the standard
drainage of subretinal fluid is usually necessary to indent the closed-vitrectomy approach used in adult eyes. When the retina
sclera effectively without raising the intraocular pressure due to is pulled so far anteriorly that it abuts the lens, the instruments
the SB. Greven and Tasman15 recommended scleral dissection must be inserted through the iris root or anterior ciliary body
in some patients and fluid drainage in nearly all patients. to avoid entering the subretinal space. Typically, a two-port
The silicone band in such small eyes may impair ocular system is used with an irrigating light pipe in one port.
growth. How the tightly applied band affects the intraocular Alternatively, infusion can be placed in the anterior chamber
1830 circulation and future development of vision is not known. Yet after removal of the lens (three-port). Machemer and deJuan17
Advanced Retinopathy of Prematurity

FIGURE 136.2. (a) Photograph of the eye of a


16-month-old boy with stage 5 retinopathy of
prematurity. The retina is totally detached and
pulled forward toward the lens. The vessels of
the detached retina are seen through the thin
retrolental fibrous membrane. (b) A B-scan
image of a stage 5 ROP detachment.
(c) Interpretive drawing of the B-scan.
(d) Fundus photograph of a former stage
5 ROP eye after successful open-sky
reattachment.

a b

CHAPTER 136
c d

recommended the corneal limbal approach to avoid tearing of successful LSV treatment of Stage 4a. They report that of 23
the anteriorly pulled retina by the instrument as it is inserted eyes tested in an average follow-up of 3.5 years, 83% had normal
through the pars plicata. This has the advantage of keeping the appearance of the macula, and most patients had an average
anterior chamber deep and the retina back, especially when visual acuity of 20/80. Moshfeghi and coworkers report38 that a
instruments are exchanged through the surgical wound. The single LSV for treatment of stage 4a resulted in complete retinal
disadvantage of the anterior chamber infusion is that the reattachment in 30 of 32 eyes (94%). In a nonrandomized
infusion fluid can push the lens posteriorly and pressure the retrospective comparison of SB to LSV for treatment of stage 4a,
detached retina behind it causing a retinal dialysis. Sector iridec- Harnett39 found that LSV was a more successful initial proce-
tomy or iris spreading hooks may be required for visualization. dure (72%), than SB (31%). Hubbard and coworkers reported40
After lensectomy, the epiretinal–retrolental membrane is that a two-port LSV performed in 37 eyes of 24 patients with a
dissected by delamination or segmentation with scissors or micro- stage 4 ROP resulted in complete retinal reattachment in 84%
vitreoretinal (MVR) blade and removed with the vitrectomy of 4a and 92% of 4b at a median follow-up of 13 months.
instrument. Additional ports may be made to allow for more Capone and Trese performed a two-port, LSV in 40 eyes of 31
complete dissection and removal of the membranes. Hyaluronic patients with a reported 90% reattachment at a follow-up mean
acid is used to aid in the dissection by Trese28 and others. of 12 months.41 Holz and coworkers report42 that a three-port
LSV technique allowed for a 85% successful reattachment in
108 eyes of 102 consecutive patients with stage 4a and 4b. They
LENS-SPARING VITRECTOMY also report43 94% sustained lens clarity on a mean follow-up of
In recent years the lens-sparing vitrectomy (LSV)37–43 technique 32 months. Despite these successes, many of the stage 4a cases
has been advanced in the treatment of retinal detachment, may not advance and may not require surgery.
particularly in posterior zone type ROP in which the ridge is
located posterior to the equator. It usually is performed in eyes
that have been treated previously by laser photocoagulation or OPEN-SKY VITRECTOMY
cryotherapy. The technique usually can not be applied effec- The open-sky vitrectomy (OSV) technique was developed by
tively in cases where the ridge is very anterior and the mem- Schepens in 1981.25 It addresses the difficult surgical scenario
brane to be cut is located very peripherally. The patient benefits of stage 5 ROP where the retina is drawn anteriorly by the
by remaining phakic. This technique is more challenging if retrolental membrane, which makes CV difficult. It also allows
the membrane to be cut and cleaned is anterior to the equator for an approach to cases with corneal opacification. To initiate
due to the relatively large size of the crystalline lens at this age. the OSV, A Flieringa ring is sewn in place to preserve the shape
In questionable cases, one may plan the LSV to start. The of the globe during surgery. Then the cornea is removed with a
sclerotomy is through the pars plicata or pars plana and not 7- to 8-mm trephine and stored in culture medium during
through ablated retina. Occasional touch of the lens with an surgery.44 The eye is irrigated with chilled balanced salt solution
intraocular instrument does not lead to cataract postoperatively. during the procedure to minimize fibrin formation.
However, if the membrane could not be removed effectively Iridotomies made at the 12 and the 6 o’clock positions in the
without lensectomy, the lens can be sacrificed at that point. past have been replaced by iris retractors (e.g., ‘perfect pupil’
Despite technical difficulties, several groups have reported designed by John Milverton). Intracapsular extraction of the
successful case series. Trese and coworkers reported37a series of crystalline lens is performed using a cryoprobe. This allows 1831
RETINA AND VITREOUS

direct visualization of the transparent anterior hyaloid and the vitrectomy is the difficulty in exposing the posterior pole in eyes
fibrous, white, retrolental membrane. Incision on the retro- with a narrow funnel. The use of additional hyaluronic acid can
lental fibrous (RLF) membrane starts far in the periphery be helpful in these situations.
usually in front of the ciliary body until the surface of the retina
is reached. The RLF membrane is circumcised. Then the dissec-
tion is continued centrally to the opening of the funnel of the TIMING OF VITRECTOMY
detachment. In order to make space inside of the funnel larger The optimal timing of vitrectomy remains to be determined. In
and make the location of the adhesion of the membrane to the patients with stage 5 ROP, a session of cryotherapy or laser
retina visible hyaluronic acid is injected into the mouth of the therapy before surgery may promote quiescence of the
funnel. In order to view the inside of the funnel better, flat round vasoproliferative process, allowing earlier repair of the detach-
coverglass, 5 mm in diameter, is used to touch the surface of the ment, but this is difficult to perform with the presence of partial
hyaluronic acid filling the funnel. The dissection continues retinal detachment, and impossible when the detachment is
toward the disk. The fibrous mass filling the funnel of detached total or nearly total.48 Chong and colleagues49 recommended
retina is cut near the optic nerve head and removed en bloc. operating as soon as plus disease has regressed. At this stage,
The iridotomies, if made, are closed with 10-0 polypropylene although the traction retinal detachment is typically still an
sutures, and the corneal button is replaced with 10-0 nylon open funnel, adhesion of the membrane to the retina is very
sutures. Hyaluronic acid is injected to deepen the anterior strong, making complete removal of the fibrous tissues from the
chamber, as well as into the open mouth of the funnel. When retina extremely difficult. There is also an increased chance of
SECTION 10

the intraocular pressure goes up and the retina is found to be reproliferation because the membrane is still continuing to
still highly elevated, the subretinal fluid is drained externally form and there is a higher chance of hemorrhage.
and hyaluronic acid is further injected into the vitreous cavity. Thus, the potential functional advantage of operating early
At the end of the surgery the detached retina should be placed must be weighed against the increased difficulty of extracting
away from the back of the iris. No effort is made to directly adherent membranes and the increased postoperative
reattach the retina by intravitreous injection. hemorrhage and fibrin production (with development of
Scleral buckling was performed by Hirose and colleagues45 secondary membranes) if vitrectomy is performed too soon.
4–8 weeks later if the retina showed no sign of reattachment. Use of the recently available anti-VEGF agents may serve as a
They reported a 39% anatomic success rate using this tech- useful adjuvant in reducing neovascularization and vascular
nique. Eyes with the closed–closed configuration had a 29% permeablility prior to vitrectomy. Perhaps an approach con-
reattachment rate; all other eyes had reattachment rates of sisting of intraocular anti-VEGF injection followed few to
greater than 60%. McPherson and coworkers30 had a 22% several days later by vitrectomy and membrane removal may
anatomic success rate with open funnels and an 11% success work. One must always weigh the risk of systemic absorption of
rate with closed funnels using the open-sky technique, whereas any anti-VEGF drug used in the eye.
Tasman and associates 31 had a 35% anatomic success rate. The timing of vitrectomy in relation to the progression of
The preoperative presence of subretinal hemorrhage in stage stage 4a and 4b disease is likely very important as well.
5 confers a poor prognosis. Ultrasonographic evaluation of stage Hartnett50 identified preoperative factors which correlated with
5 eyes should be performed prior to OSV. Steidl and Hirose, in outcome of vitrectomy and SB procedures performed on stage
a retrospective review46 of 426 eyes of 263 patients who under- 4 and 5 eyes which had received previous laser ablation. In eyes
went OSV for stage 5 ROP, identified the presence of subretinal which failed first procedures, vitreous haze or organization and
organization (bands, plaques, or diffuse hemorrhage) to be plus disease were found to be significant in stage 4 eyes, while
associated with incomplete retinal reattachment or retinal 6 clock hours of ridge elevation and plus disease were found to
attachment without any useful vision. be significant in stage 5 eyes. Hartnett recommends additional
laser be performed before surgery in eyes manifesting neo-
vascularization or plus disease. The optimal timing of LSV with
COMPARISON OF OSV TO CV respect to laser ablation is unknown. An Argentine group51
The relative rates of success of CV versus OSV depend in part reports in a small series of eight patients, that those patients
on the selection of cases. The advantage of the closed technique who had received laser treatment prior to LSV had more organi-
is the avoidance of removal and replacement of the corneal zation of the vitreous which made surgery more difficult, with
button. The disadvantages include the need for a pars plicata or the previously untreated patients faring better.
iris root entry site because of the extreme anterior displacement The optimal time for surgery may be after 6 months of age
of the retina and the difficulty maneuvering vitrectomy but before the patient is 1 year old. One may operate earlier if
instruments within the small closed space of the premature the eye has been treated with cryotherapy or photocoagulation
infant eye. The use of multiple ports may allow for more com- and the fundus shows no sign of active vasoproliferation. Even
plete removal of membranes. Incomplete removal of residual when surgery is delayed for years, it may be worth attempting
peripheral membranes results in only ‘partial’ reattachment. repair in some cases because ambulatory acuity has been
Reported disadvantages of the open-sky technique include obtained postoperatively in patients with long-standing retinal
increased complexity of postoperative management because of detachment up to 3 years of age.45
the need for a corneal graft, prolonged hypotony, and a longer
operating time. In experienced hands, however, operative time
is less than 2 h.45 Choroidal detachments are rare, and corneal COMPLICATIONS OF VITRECTOMY
endothelial cell-density changes little.47 The advantages of the Iatrogenic retinal breaks produced during vitrectomy surgery are
open-sky technique are the excellent visualization of the extremely difficult to close. Some peripheral breaks may be closed
pathologic process and the direct access to the retrolental by scleral buckling, but breaks formed posterior to the ridge or
membrane. This makes more complete removal of peripheral in the posterior pole in infants with ROP are impossible to close
membranes possible and less residual traction on the retina either with scleral buckling or with air–fluid exchange and
when it is reattached at the posterior pole. Some cases that internal drainage of subretinal fluid probably due to the inability
would be inoperable by the closed technique can be repaired to completely release the traction by vitrectomy or retinal
1832 using the open-sky method. The main drawback of open-sky foreshortening, or a combination of both. Other factors that can
Advanced Retinopathy of Prematurity

make ROP surgery more difficult and associated with higher hypotony, and vitreous hemorrhage were similar in the two
risk include the presence of a persistent hyaloid artery,52 groups. Thus, although surgical approaches to ROP are meeting
intraocular bleeding, multiple circular retinal folds, and tilting with improved rates of retinal reattachment in ROP, the visual
of the funnel toward one quadrant.45 outcomes to date indicate that the emphasis must remain on
Although anatomic reattachment can be achieved in eyes prevention of retinal detachment.35,54
with severe disease using either technique, vision has been
disappointingly poor.49,53 Machemer and deJuan17 reported an SUMMARY
anatomic success rate of 64% and a functional success rate of
43% in stage 4 cases. Patients with stage 5 ROP do not fare as Interest in ROP has grown along with the increasing incidence
well. Stage 5 cases with the open–open configuration have the of the disease resulting in many advances for treament of acute
best prognosis; the closed–closed configuration is least likely to ROP. Treatments of advanced stages of acute ROP have also
result in recovery of useful vision.26,32–36 However, the limited been refined, with properly timed cryotherapy or laser photo-
vision, which may detect the movement of objects, for example, coagulation preventing retinal detachment and meticulous
which develops after reattachment of stage 5 ROP, is very useful vitrectomy techniques successfully removing traction forces.
for activities of daily life and as such very enhancing to the Smaller gauge (25 G and 23 G) vitrectomy techniques are being
quality of life in the affected young children. pioneered, yet have not been widely adapted to ROP surgery,
In the cryo-ROP study, the visual outcome of infants who though ultimately they may be found to be advantageous when
progressed to stage 5 retinal detachment was studied retro- operating on the smaller eyes of infants. Adjuvant to surgery

CHAPTER 136
spectively.35,54 Surgery was performed by several different such as the use of recently developed anti-VEGF agents may
surgeons using different techniques. There were no standard- make surgical success higher in addition to being potentially
ized preoperative criteria for undertaking vitrectomy, and useful in treating the acute stages of ROP.
patients were not randomized. The configuration of detach- The improved anatomic success rates of vitreoretinal surgery
ment (open vs closed funnel) was not specified. The anatomic have been confounded by disappointingly poor visual function.
success rate was 28% at 1 year and 21% at 5.5 years in operated Still, surgery should be pursued when indicated, as even
eyes, with a functional success rate of 1–3%. Although data attaining hand-motion visual acuity allows many patients to
were not specified for all eyes that did not undergo vitrectomy, remain ambulatory, and anatomic success may avoid the need
one of the 10 eyes described had spontaneous resolution of the for enucleation. Further modifications of treatment protocols,
retinal detachment. Eyes that underwent vitrectomy had a with an emphasis on preventing retinal detachment, will be
lower incidence of glaucoma and shallow anterior chamber necessary before significantly better acuity can be retained in
compared with unoperated eyes. The rates of corneal opacity, eyes with severe stages of this devastating disease.

REFERENCES
1. Terry TL: Extreme prematurity and 9. Kaiser RS, Trese MT, Williams GA, 18. McPherson AR, Hittner HM, Kretzer FL:
fibroblastic overgrowth of persistent Cox MS Jr: Adult retinopathy of prematurity: Treatment of acute retinopathy of prematurity
vascular sheath behind each crystalline outcomes of rhegmatogenous retinal by scleral buckling. In: McPherson AR,
lens. I: Preliminary report. Am J Ophthalmol detachments and retinal tears. Hittner HM, Kretzer FL, eds. Retinopathy
1942; 25:203–204. Ophthalmology 2001; 108:1647–1653. of prematurity-current concepts and
2. Tasman W, Patz A, McNamara JA, et al: 10. Topilow HM, Ackerman AL, Wang FM: controversies. Toronto: BC Decker;
Retinopathy of prematurity: the life of a The treatment of advanced retinopathy of 1986:179–192.
lifetime disease. Am J Ophthalmol 2006; prematurity with cryotherapy and scleral 19. Prost ME:Results of treatment of retinal
141:167–174. buckling procedure. Ophthalmology 1985; detachment in active stage-5 retinopathy
3. Cryotherapy for Retinopathy of Prematurity 92:379–387. of prematurity. Klin Oczna 2003;
Cooperative Group: Multicenter trial of 11. Cryotherapy for Retinopathy of Prematurity 105:387–391.
cryotherapy for retinopathy of prematurity: Cooperative Group: Multicenter trial of 20. Sneed SR, Pulido JS, Blodi CF, et al:
ophthalmological outcomes at 10 years. cryotherapy for retinopathy of prematurity. Surgical management of late-onset retinal
Arch Ophthalmol 2001; 119:1110–1118. One year outcome-structure and detachments associated with regressed
4. Hartnett ME, McColm JR: Retinal features function. Arch Ophthalmol 1990; retinopathy of prematurity. Ophthalmology
predictive of progressive stage 4 retinopathy 108:1408–1416. 1990; 97:179–183.
of prematurity. Retina 2004; 24:237–241. 12. McPherson AR, Hittner HM: Scleral 21. Mintz-Hittner HA, Kretzer FL: The rationale
5. Orellana J: Scleral buckling in acute buckling in 2- to 11-month-old premature for cryotherapy with a prophylactic scleral
retinopathy of prematurity stages 4 and 5. infants with retinal detachment associated buckle for zone 1 threshold retinopathy of
In: Eichenbaum JW, Mamelok AE, Mittl RN, with acute retrolental fibroplasia. prematurity. Doc Ophthalmol 1990;
Orellana J, eds. Treatment of retinopathy of Ophthalmology 1979; 86:819–835. 74:263–268.
prematurity. Chicago: Year Book Medical; 13. Ricci B, Santo A, Ricci F, et al: Scleral 22. Charles S: Vitrectomy with ciliary body entry
1991:194–213. buckling surgery in stage 4 retinopathy of for retrolental fibroplasia. In McPherson AR,
6. Palmer EA, Hardy RJ, Dobson V, et al: prematurity. Graefes Arch Clin Exp Hittner HM, Kretzer FL, eds. Retinopathy
Cryotherapy for Retinopathy of Prematurity Ophthalmol 1996; 234:S38–S41. of prematurity-current concepts and
Cooperative Group 15-year outcomes 14. Beyrau K, Danis R:Outcomes of primary controversies. Toronto: BC Decker;
following threshold retinopathy of scleral buckling for stage 4 retinopathy of 1986:225–234.
prematurity: final results from the prematurity.Can J Ophthalmol 2003; 23. deJuan E, Machemer R: Retinopathy of
multicenter trial of cryotherapy for 38:267–271. prematurity. Surgical technique. Retina
retinopathy of prematurity. Arch 15. Greven C, Tasman W: Scleral buckling in 1987; 7:63–69.
Ophthalmol. 2005; 123:311–318. stages 4B and 5 retinopathy of prematurity. 24. Jabbour NM, Eller AE, Hirose T, et al:
7. Tufail A, Singh AJ, Haynes RJ, et al: Late Ophthalmology 1990; 97:817–820. Stage 5 retinopathy of prematurity.
onset vitreoretinal complications of 16. Trese MT: Scleral buckling for retinopathy Prognostic value of morphologic findings.
regressed retinopathy of prematurity. Br J of prematurity. Ophthalmology 1994; Ophthalmology 1987; 94:1640–1646.
Ophthalmol 2004; 88:243–246. 101:23–26. 25. Schepens CL: Clinical and research
8. Terasaki H, Hirose T: Late-onset retinal 17. Machemer R, deJuan E: Retinopathy of aspects of subtotal open-sky vitrectomy.
detachment associated with regressed prematurity: approaches to surgical Am J Ophthalmol 1981; 91:143.
retinopathy of prematurity. Jpn J therapy. Aust N Z J Ophthalmol 1990; 26. Zilis JD, deJuan E, Machemer R: Advanced
Ophthalmol 2003; 47:492–497. 18:47–45. retinopathy of prematurity. The anatomic 1833
RETINA AND VITREOUS

and visual results of vitreous surgery. 36. Mintz-Hittner HA, O’Malley RE, Kretzer FL: 45. Hirose T, Schepens CL, Katsumi O,
Ophthalmology 1990; 97:821–826. Long-term form identification vision after Mehta MC: Open-sky vitrectomy for severe
27. Charles S: Vitreoretinal surgery for early, closed, lensectomy-vitrectomy for retinal detachment caused by advanced
retinopathy of prematurity. Birth Defects stage 5 retinopathy of prematurity. retinopathy of prematurity. In: Flynn J, ed.
1988; 24:287–293. Ophthalmology 1997; 104:454–459. Retinopathy of prematurity. New York:
28. Trese MT: Surgical results of stage V 37. Prenner JL, Capone A Jr, Trese MT: Visual Springer-Verlag; 1992:95–114.
retrolental fibroplasia and timing of surgical outcomes after lens-sparing vitrectomy for 46. Steidl SM, Hirose T: Subretinal organization
repair. Ophthalmology 1984; 91:461–466. stage 4A retinopathy of prematurity. in stage 5 retinopathy of prematurity.
29. Blacharski PA, Charles S: Thrombin Ophthalmology 2004; 111:2271–2273. Graefes Arch Clin Exp Ophthalmol 2003;
infusion to control bleeding during 38. Moshfeghi AA, Banach MJ, Salam GA, 241:263-268; Epub 2003 Mar 18.
vitrectomy for stage V retinopathy of Ferrone PJ: Lens-sparing vitrectomy for 47. Sawa M, Hirose T, Kenyon KR: Endothelial
prematurity. Arch Ophthalmol 1987; progressive tractional retinal detachments specular microscopy in children with
105:203–205. associated with stage 4A retinopathy of retrolental fibroplasia undergoing open-sky
30. McPherson AR, Hittner HM, Moura RA, prematurity. Arch Ophthalmol 2004; vitrectomy. Jpn J Ophthalmol 1990;
Kretzer FL: Treatment of retrolental 122:1816–1818. 34:1–14.
fibroplasia with open-sky vitrectomy. In: 39. Hartnett ME, Maguluri S, Thompson HW, 48. Trese MT: Surgical therapy for stage V
McPherson AR, Hittner HM, Kretzer FL, McColm JR: Comparison of retinal retinopathy of prematurity. A two-step
eds. Retinopathy of prematurity-current outcomes after scleral buckle or lens- approach. Graefes Arch Clin Exp
concepts and controversies. Toronto: sparing vitrectomy for stage 4 retinopathy Ophthalmol 1987; 225:266–268.
BC Decker; 1986:225–234. of prematurity. Retina 2004; 24:753–757. 49. Chong LP, Machemer R, deJuan E:
31. Tasman W, Borrone RN, Bolling J: 40. Hubbard GB III, Cherwick DH, Burian G: Vitrectomy for advanced stages of
SECTION 10

Open-sky vitrectomy for total retinal Lens-sparing vitrectomy for stage 4 retinopathy of prematurity. Am J
detachment in retinopathy of prematurity. retinopathy of prematurity. Ophthalmology Ophthalmol 1986; 102:710–716.
Ophthalmology 1987; 94:449–452. 2004; 111:2274–2277. 50. Hartnett ME: Features associated with
32. Hirose T, Katsumi O, Mehta MC, 41. Capone A Jr, Trese MT: Lens-sparing surgical outcome in patients with stages 4
Schepens CL: Vision in stage 5 retinopathy vitreous surgery for tractional stage 4A and 5 retinopathy of prematurity. Retina
of prematurity after retinal reattachment by retinopathy of prematurity retinal 2003; 23:322–329.
open-sky vitrectomy. Arch Ophthalmol detachments. Ophthalmology 2001; 51. Luna JD, Caribaux LJ, Reviglio VE,
1993; 111:345–349. 108:2068–2070. Juarez CP: Lens-sparing surgery for
33. Seaber JH, Machemer R, Eliott D, et al: 42. Lakhanpal RR, Sun RL, Albini TA, Holz ER: retinopathy of prematurity. Ophthalmology
Long-term visual results of children after Anatomic success rate after 3-port lens- 2003; 110:1669.
initially successful vitrectomy for stage V sparing vitrectomy in stage 4A or 4B 52. Eller AW, Jabbour NM, Hirose T,
retinopathy of prematurity. Ophthalmology retinopathy of prematurity. Ophthalmology Schepens CL: Retinopathy of prematurity.
1995; 102:199–204. 2005; 112:1569–1573. The association of a persistent hyaloid
34. Fuchino Y, Hayashi H, Kono T, Ohshima K: 43. Lakhanpal RR, Davis GH, Sun RL, et al: artery. Ophthalmology 1987; 94:444–448.
Long-term follow-up of visual acuity in eyes Lens clarity after 3-port lens-sparing 53. Topilow HW, Ackerman AL, Wang FM,
with stage 5 retinopathy of prematurity vitrectomy in stage 4A and 4B retinal Strome RR: Successful treatment of
after closed vitrectomy. Am J Ophthalmol detachments secondary to retinopathy of advanced retinopathy of prematurity.
1995; 120:308–316. prematurity. Arch Ophthalmol 2006; Ophthalmic Surg 1988; 19:781–785.
35. Quinn GE, Dobson V, Barr CC, et al: Visual 124:20–23. 54. Quinn GE, Dobson V, Barr CC, et al: Visual
acuity of eyes after vitrectomy for retinopathy 44. Hirose T, Lou PL: Retinopathy of acuity in infants after vitrectomy for severe
of prematurity: follow-up at 5 1/2 years. prematurity. Int Ophthalmol Clin 1986; retinopathy of prematurity. Ophthalmology
Ophthalmology 1996; 103:595–600. 26:1–23. 1991; 98:5–13.

1834
CHAPTER

137 Eales’ Disease


Marissa L. Albano and Robert P. Murphy

Key Features HISTORY


• Eales’ disease is an idiopathic obliterative vasculopathy of the
peripheral retina
The disease is named after Henry Eales, an ophthalmologist
• Initially presents as retinal periphlebitis and later as retinal
who in 1880 described a syndrome of recurrent vitreous
ischemia, leading to vascular alterations, neovascularization,
hemorrhages in young men with epistaxis and constipation.1
and vitreous hemorrhage
He termed this new entity primary recurrent retinal
• Affects persons of all ages and is usually bilateral; most
hemorrhage. Using the newly developed direct ophthal-
commonly affects young, healthy males in the Indian
moscope, Eales documented abnormal retinal veins and areas in
subcontinent and Middle East
the peripheral retina that were free of capillaries.2 In spite of
• The etiology is believed to be multifactorial
these astute early observations, the entity he described differs
• Early manifestations include vascular sheathing and focal
considerably from the disease that now bears his name. He did
occlusion of peripheral retinal vessels
not observe any new vessels or inflammation preceding or
• Therapy is directed toward the presenting symptoms of the
accompanying the hemorrhages.
disease:
In 1887, Wadsworth described associated inflammation and
• Oral, peribulbar and intravitreal corticosteroids are used in
neovascularization with this disease.3 Since then, numerous
the active inflammatory stage
authors have grouped periphlebitis retinae and idiopathic
• Retinal neovascularization appears to respond well to laser
recurrent vitreous hemorrhages with or without retinal
photocoagulation
perivasculitis under the term Eales’ disease.
• Vitrectomy for vitreous hemorrhage with or without retinal
Not all authors have believed that Eales’ disease is a specific
detachment
entity. Duke–Elder thought that Eales’ disease represented the
clinical manifestation of many diseases.4 Indeed, refined
diagnostic tests have demonstrated that many of the so-called
idiopathic hemorrhages are the result of diseases with known
causes, such as sarcoidosis, systemic lupus erythematosus, diabetes
Eales’ disease is an idiopathic obliterative vasculopathy of the mellitus, sickle-cell disease, and collagen vascular disease.
peripheral retina, initially presenting as retinal periphlebitis However, after elimination of these causes, a group of patients
and later as retinal ischemia, leading to vascular alterations, remains with idiopathic peripheral nonperfusion and perivas-
neovascularization, and vitreous hemorrhage. It can affect per- culitis of the retina. Many investigators now agree that Eales’
sons of all ages and is usually bilateral; it most commonly disease is a distinct entity comprising characteristic fundoscopic
affects young, healthy males in the Indian subcontinent and and fluorescein angiographic features.5 Although this disease has
Middle East. The etiology is believed to be multifactorial, been called periphlebitis retinae,6 emphasizing the abnorma-
although it currently remains unclear. Recent immunological, lities of retinal venules, evidence suggests that the inflammation
biochemical, and histopathological studies have implicated in this disease affects both arterioles and venules.7
Mycobacterium tuberculosis, retinal autoimmunity, human
leukocyte antigen, and free radical mediated damage in the EPIDEMIOLOGY
pathogenesis of this disease. Early manifestations include vas-
cular sheathing and focal occlusion of peripheral retinal vessels. Eales’ disease is uncommon in North America; however, this
With progression, areas of nonperfusion can develop. disorder is responsible for widespread visual loss in the Indian
Neovascularization can occur at the junction of the perfused subcontinent. It often affects healthy young adults, with the
and nonperfused retina, frequently resulting in recurrent average age of onset at 20–30 years. Patients usually present
vitreous hemorrhages. As the exact etiopathogenesis remains with symptoms of vitreous hemorrhage, such as floaters or
unclear, therapy is directed toward the presenting symptoms of decreased vision. The majority of patients experience bilateral,
the disease. Oral, peribulbar, and intravitreal corticosteroids are although often asymmetric, involvement.
used in the active inflammatory stage, while retinal neovas- Most reports, including Eales’ original description, indicate a
cularization in Eales’ disease appears to respond well to laser male predominance. However, Murphy and co-workers found
photocoagulation. Vitrectomy is utilized for vitreous hemor- an equal prevalence of men and women in their study of 55
rhage with or without retinal detachment. patients in North America.7

1835
RETINA AND VITREOUS

CLINICAL FEATURES areas in the far periphery to massive nonperfusion extending


into the posterior pole (Fig. 137.3). The nonperfusion is
generally confluent and sharply demarcated from the posterior
Clinical Features of Eales’ Disease perfused retina. The temporal retina is most commonly
• Vascular sheathing affected.
• Retinal capillary nonperfusion with or without The microvascular abnormalities may be so severe that the
neovascularization fundus resembles Coats’ disease. Fine white lines representing
• Recurrent vitreous hemorrhages the remains of obliterated large vessels (ghost vessels) are often
• Cystoid macular edema seen in the area of nonperfusion.
Elliot8 and Spitznas and colleagues9 have documented the
INFLAMMATION vascular abnormalities at the junction of the anteroperipheral
nonperfused retina and the posterior perfused retina.
Most patients with Eales’ disease develop ocular inflammation, Intraretinal hemorrhages often first appear in the affected area,
especially early in its course. Vascular sheathing is seen in most followed by an increase in vascular tortuosity with frequent
patients (Fig. 137.1). The degree of sheathing ranges from fine collateral formation around occluded vessels (Fig. 137.3).
white lines on both sides of the blood column to thick exudative Microaneurysms, arteriovenous shunts, and venous beading
sheathing. The thin white lines tend to be continuous, whereas are commonly seen at the junction (Fig. 137.4). Fluorescein
the heavy, exudative sheathing is often segmental. angiography enhances these abnormalities and often demon-
SECTION 10

Areas of vascular sheathing frequently demonstrate hyper- strates staining at the stumps of obliterated vessels.
fluorescence in fluorescein angiography (Fig. 137.2). However, Patients with Eales’ disease can also experience branch vein
there is no direct correlation between the regions of sheathing occlusion. The branch vein occlusion can be either single or
and staining. The intensity of the hyperfluorescence seen on the multiple. Unlike patients with primary branch vein occlusion,
fluorescein angiograms does not correlate with the intensity of in whom the pathologic condition is confined to one quadrant,
inflammation. patients with Eales’ disease have more extensive involvement of
In the century since Eales’ observation of altered retinal the peripheral retina that does not respect the horizontal
veins, many investigators have described Eales’ disease as a midline.
primary disease of altered retinal veins. Elliot and Harris sug-
gested the term periphlebitis retinae for this disorder.6 However,
more recent studies have reported equal involvement of NEOVASCULARIZATION
arteriolar and venular sheathing.7 Because of the evidence of The retinal nonperfusion leads to the eventual development of
arteriolar involvement (Fig. 137.1b), this disease should be new vessels. The neovascularization can form either on the disk
considered a retinal vasculitis or vasculopathy. or elsewhere in the retina. Neovascularization of the iris has
Keratic precipitates, anterior chamber cell and flare, and also been described. These abnormal blood vessels frequently
vitreous cells have been observed in patients with Eales’ bleed and are the major cause of visual loss in this disease.
disease.7 Cystoid macular edema occurs in eyes with extensive Ischemic retina produces a diffusible substance that stimu-
sheathing. Although the exact cause of the macular edema is lates neovascularization, vascular endothelial growth factor
unknown, it may be associated with low-grade inflammation. (VEGF). Perentes and colleagues demonstrated a close relation-
ship between prominent neovascular proliferation and the
intense expression of VEGF via histopathological and
NONPERFUSION immunohistochemical examination of an enucleated eye from
Peripheral retinal nonperfusion is present in all patients with a patient with documented Eales’ disease.10 The neovas-
this disease. The extent of the perfusion ranges from small cularization often occurs along the junction of perfused and

a b

FIGURE 137.1. (a) Thin white lines representing vascular sheathing surround a retinal venule in a patient with Eales’ disease. (a) Extensive
arteriolar sheathing with retinal vascular nonperfusion nasal to disk.
1836 (a) From Gieser SC, Murphy RP: Eales disease. In: Ryan S, ed. Retina. St Louis, MO: CV Mosby; 1989.
Eales’ Disease

CHAPTER 137
FIGURE 137.2. Fluorescein angiogram demonstrates abnormal
staining of a small retinal venule (arrows) in a patient with Eales’
disease. There was venous sheathing in this area.

FIGURE 137.4. Fluorescein angiogram demonstrates severe


nonperfusion of the retinal vasculature involving the macula.

FIGURE 137.3. Fluorescein angiogram of the peripheral retina


demonstrates the junction of normally perfused retinal vessels
adjacent to an area of nonperfused retina. Note the vascular
abnormalities and the small area of neovascularization at the junction.

FIGURE 137.5. Hypovascular fibroproliferation emanates from the


disk of a patient with advanced Eales’ disease.

nonperfused retina, similar to the appearance of sickle-cell


retinopathy. The neovascularization frequently has a prominent VISUAL PROGNOSIS
fibrous component. Occasionally, patients have extensive
retinal and vitreal proliferation of avascular sheets and strands The natural course of this disease is variable. Although the
of fibrous scar tissue (Fig. 137.5). The anteroposterior traction visual acuity in patients with Eales’ disease ranges from normal
resulting from the fibrovascular membrane places these eyes at to no-light perception, most eyes retain good acuity. In spite of
risk of developing traction retinal detachment. Badrinath and extensive anteroperipheral nonperfusion, the macula is usually
colleagues found multifocal fibrous and fibrovascular spared, preserving central vision. Murphy and co-workers
vitreoretinal adhesions in 83% of eyes undergoing vitreous reported that 67% of their patients had final visual acuity in the
surgery for complications of Eales’ disease, in addition to noting better eye that ranged from 20/15 to 20/40.7 Twenty-four
type II collage in the epiretinal membranes causing tangential percent had visual acuity that ranged from 20/50 to 20/200,
traction.11 and 9% had visual acuity worse than 20/200. 1837
SECTION 10 RETINA AND VITREOUS

a b

FIGURE 137.6. (a) Neovascularization of the disk. Note the segmental exudative arteriolar sheathing. (b) Same patient as in (a) 2.5 years later.
The patient has had total regression of the neovascularization after treatment with scatter photocoagulation of all areas of nonperfused retina.
Note that the former areas of arteriolar sheathing have resolved.
(a and b) From Gieser SC, Murphy RP: Eales disease. In: Ryan S, ed. Retina. St Louis, MO: CV Mosby; 1989.

Vitreous hemorrhage is the most frequent cause of visual systemic steroid efficacy. In a prospective study by Bali, pulsed
loss. Usually the hemorrhage settles to the lower portion of the oral methotrexate therapy at a dose of 12.5 mg/week was shown
vitreous and is gradually reabsorbed within several weeks or to be clinically effective within 4 weeks and is associated with
months, with the return of normal central vision. Severe, an acceptable safety profile.18 Recently, intravitreal triam-
permanent visual loss usually results from complications cinolone acetonide was successfully used in three eyes of
associated with neovascularization, such as persistent vitreous two patients with Eales’ disease in the acute inflammatory
hemorrhage, retinal detachment, and neovascular glaucoma. stage who were not responding to oral and peribulbar
Occasionally, loss of vision is caused by cystoid macular edema, corticosteroids.19
macular holes, retinal telangiectasia, or epiretinal membrane. Investigators generally agree that peripheral scatter
In some patients, relentless nonperfusion progresses across the photocoagulation of the ischemic retina is invaluable in the
macula (see Fig. 137.4); visual acuity in these eyes is usually treatment of neovascularization (Fig. 137.6).20,21 This is similar
less than 20/400. to the treatment used for other vasoproliferative diseases of the
A long-term follow-up study of 130 patients with Eales’ retina, such as diabetic and sickle-cell retinopathies. Several
disease showed visual acuity improved in 20%, maintained in investigators have demonstrated favorable results with light-
43%, and worsened in 37%. The complications noted after intensity, full-scatter argon laser photocoagulation to the
5 years were tractional detachment, cataract, rubeosis iridis, nonperfused retina and to the junction of perfusion and non-
neovascular glaucoma, and phthisis bulbi.12 perfusion.21 Others have used a combination of cryotherapy and
laser photocoagulation.22 Because nonperfused retina is more
TREATMENT fragile than perfused retina, caution should be exercised when
treating these patients. There is evidence to show that
Henry Eales offered his patients a mixture of laxative, digitalis, prophylactic pan retinal photocoagulation in areas of ischemic
and belladonna.1 Presumably, these remedies were not retina in asymptomatic fellow eyes of patients is effective in
beneficial. Lowered levels of antioxidant vitamins in patients controlling the progression and future complications of this
with Eales’ disease and consequent accumulation of oxygen and disease.23 Due to the intense expression of VEGF in Eales’
lipid free radicals have prompted the belief that vitamins E, C, disease patients, anti-VEGF therapy with either Avastin or
and A may be of value.13 Bhooma and colleagues demonstrated Lucentis (Genentech Corp, San Francisco, CA) may be of some
an increase in serum thiobarbituric acid nactine substances, an treatment benefit, however this remains to be studied.
indicator of lipid peroxidation products produced by oxygen Vitrectomy can be employed for removing persistent vitreous
and free radicals.13 Some investigators believe there is an asso- hemorrhages and fibrosis, often with good results.24 Kumar and
ciation of Eales’ disease with tuberculoprotein hypersensitivity, colleagues’ study demonstrated the importance of early vit-
therefore prompting the use of antituberculous therapy as rectomy for persistent hemorrhages. Delaying vitrectomy more
well.14–16 The benefit of these therapies is untested. than 6 months is believed to allow the development of cystoid
As the basic pathologic lesion of Eales’ disease is macular edema, macular scar, and macular pucker formation
inflammatory occlusion of retinal vessels, corticosteroids are with poorer visual outcome.25 No treatment is known to
becoming a mainstay of treatment. For the acute inflammatory prevent or reverse the nonperfusion or capillary drop-out.
stage, oral prednisone (1 mg/kg) is started and the dose tapered
for 6–8 weeks. If the patient is intolerant to systemic cor- DIFFERENTIAL DIAGNOSIS AND SYSTEMIC
ticosteroids or if macular edema develops, periocular depot EVALUATION
corticosteroid/triamcinolone acetonide injections have been
found to be beneficial.17 Immunosuppressive agents are As an idiopathic entity, Eales’ disease must first be differen-
1838 reserved for intolerance to systemic steroids or for lack of tiated from retinal vasculopathies with known causes. Systemic
Eales’ Disease

with Eales’ disease.14 This protein has not been seen in healthy
TABLE 137.1. Laboratory Investigations for Eales„ Disease controls or patients with diabetic retinopathy; however, it
CBC has been isolated from the serum of patients with posterior
uveitis, tuberculosis, leprosy, and rheumatoid arthritis.
Erythrocyte sedimentation rate
Immunohistochemistry on epiretinal membranes of Eales’
Reticulocyte count disease patients localized the 88-kDa protein to inflammatory
Postprandial blood sugar
cells and nonvascular endothelium.27 Further investigation is
necessary to decipher the role of this protein.
Mantoux test (tuberculosis) Electrophoretic study of serum proteins in patients with
Sickle cell preparation Eales’ disease has shown a rise in a-globulins and reduced
albumin levels.28 Isoelectric focusing of the serum samples has
Hemoglobin electrophoresis (sickle cell retinopathy)
revealed several unique proteins in these patients.13 It is
Immunoglobulin profile VDRL and Treponema pallidum possible that altered immune reactivity to an extraneous agent
Hemagglutination Test may play a role in the pathogenesis of Eales’ disease. A small
Antinuclear antibody (SLE) study by Saxena and associates found a lymphocyte proliferative
response in 6 of 24 patients against S-antigen, its uveitogenic
Serum angiotensin-converting enzyme (sarcoidosis) fragments or interphotoreceptor retinoid-binding protein,
further suggesting that retinal antigens play a role in the

CHAPTER 137
Lysozyme (sarcoidosis)
X-ray chest (tuberculosis and sarcoidosis)
pathogenesis of Eales’ disease.29
Recent evidence may suggest that HLA haplotypes have a
possible predilection for Eales’ disease. HLA phenotypes DR3,
A1, B8, B5(51), and DR15(2) occurred in the majority of cases
of Eales’ disease, while they are not found in normal controls.
diseases such as diabetes mellitus, sickle-cell hemoglo- HLA DQ2, DR52, and Bw6 were found in higher frequency of
binopathies, sarcoidosis, and systemic lupus erythematosus can Eales’ patients, and are believed to be strongly associated with
manifest retinal inflammation, nonperfusion, and neovas- the disease.30
cularization. Ocular diseases such as branch and central retinal Some patients with Eales’ disease have a concomitant vesti-
vein occlusions, Coat’s disease, parsplanitis, dragged disk buloauditory dysfunction. Renie and associates discovered
syndrome, and macular telangiectasia.17 However, one can rule sensorineural hearing loss in 24% of their 35 patients with this
out these conditions with a careful history and appropriate disease and vestibuloauditory dysfunction in 50% of their
laboratory tests (Table 137.1). patients.26 Wagner and Fehrmann’s case report documented a
Many investigators have emphasized a relationship between man diagnosed with Eales’ disease presenting with concurrent
Eales’ disease and M. tuberculosis.8,9,14–17 Although there is a visual acuity loss and Meniere-like vestibulocochlear
higher than normal incidence of positive reaction to the symptoms, who responded well to high-dose IV corticosteroids.
tuberculin protein, no one has demonstrated any evidence that They hypothesized a common pathogenic cause such as
the ocular changes are related to infection of the eye or retina by autoimmune-mediated microvasculitis of retinal and
tuberculin bacteria. The most widely held belief is that Eales’ vestibulocochlear vessels.31
disease is an immune-mediated hypersensitivity process result- There are multiple case reports of diseases of the central
ing from prior exposure to the tuberculoprotein.15 Renie and nervous system in patients with Eales’ disease, including mul-
associates noted in their group of 32 patients that 48% had tiple sclerosis, cerebellar ataxia, myelopathy, hemiplegia, and
either tuberculosis or a history of exposure to tuberculosis.26 stroke.32 Internuclear ophthalmoplegia33 and an internal carotid
The detection of M. tuberculosis DNA by PCR in a significant artery aneurysm34 have also been reported in patients with
number of vitreous fluid samples and epiretinal membranes of Eales’ disease. Additionally, several investigators have noted an
patients with Eales’ disease compared to normal controls increased prevalence of immunologic disorders.35
further emphasizes the association of tuberculosis with the Eales’ disease is a long-recognized, but poorly understood,
pathogenesis of Eales’ disease.16 disorder. Until we understand more about the cause and
Recently, a novel acute phase reactant 88-kDa protein has pathogenesis of Eales’ disease, we will continue to be able to
been isolated in the vitreous humor and serum of patients treat only the complications of this disorder.

REFERENCES
1. Eales H: Causes of retinal hemorrhage 7. Murphy RP, Gieser SC, Fine SL, Patz A: 12. Atmaca LS, Batioglu F, Atmaca Sonmez P:
associated with epistaxis and constipation. Retinal and vitreous findings in Eales A long-term follow-up of Eales’ disease.
Birm Med Rev 1880; 9:262. disease. Invest Ophthalmol Vis Sci 1986; Ocul Immunol Inflamm 2002; 10:213.
2. Eales H: Primary retinal hemorrhages in 27:121. 13. Bhooma V, Sulochana KN, Biswas J,
young men. Ophthalmol Rev 1882; 1:41. 8. Elliot AJ: Thirty-year observation of patients Ramakrishnan S: Eales’ disease:
3. Wadsworth OF: Recurrent retinal with Eales disease. Am J Ophthalmol 1975; accumulation of reactive oxygen
hemorrhage, followed by the development 80:404. intermediates and lipid peroxides and
of blood vessels in the vitreous. Ophthalmol 9. Spitznas M, Meyer-Schwickerath G, decrease of antioxidants causing
Rev 1887; 6:289–299. Stephan B: The clinical picture of Eales inflammation, neovascularization and retinal
4. Duke-Elder WS: Diseases of the retina. In: disease. Graefes Arch Clin Exp Ophthalmol damage. Curr Eye Res 1997; 16:91.
Duke-Elder WS, ed. System of 1975; 194:73. 14. McCaughan F, Holmes A, Lynn W, Friedland
ophthalmology. St Louis, MO: Mosby; 1967. 10. Perentes Y, Chan CC, Bovey E, et al: JS: Mycobacterium tuberculosis infection
5. Gieser SC, Murphy RP: Eales disease. In: Massive vascular endothelium growth factor complicated by Eales disease with
Ryan S, ed. Retinal disease. St Louis, MO: (VEGF) expression in Eales’ disease. Klin peripheral neuropathy. Clin Infec Dis 2002;
Mosby; 1989. Monatsbl Augenheilkd 2002; 219:311. 35:89.
6. Elliot AJ, Harris GS: The present status of 11. Badrinath SS, Gopal L, Sharma T, et al: 15. Madhavan HN, Therese KL, Doraiswamy K:
the diagnosis and treatment of periphlebitis Vitreoschisis in Eales’ disease: pathogenic Further investigations on the association of
retinae (Eales disease). Can J Ophthalmol role and significance in surgery. Retina Mycobacterium tuberculosis with Eales’
1969; 4:117. 1999; 19:51. disease. Indian J Ophthalmol 2002; 50:35.
1839
RETINA AND VITREOUS

16. Madhavan HN, Therese KL, Gunisha P, 23. Ishaq M, Niazi MK: Usefulness of laser 30. Ishaq M, Karamat S, Niazi MK: HLA typing
et al: Polymerase chain reaction for photocoagulation in managing in patients of Eales disease. J Coll
detection of Mycobacterium tuberculosis asymptomatic eyes of Eales’ disease. Physicians Surg Pak 2005; 15:288.
in epiretinal membrane in Eales’ disease. J Ayub Med Coll Abbottabad 2002; 14:22. 31. Wagner W, Fehrmann A: Association of
Invest Ophthalmol Vis Sci 2000; 41:822. 24. Smiddy WE, Isernhagen RD, Michels RG, retinal vasculitis (Eales’ disease) and
17. Biswas J: Eales disease – an update. Glaser BM: Vitrectomy for nondiabetic Meniere-like vestibulocochlear symptoms.
Survey Ophthalmol 2002; 47:197. vitreous hemorrhage. Retina 1988; 8:88. Eur Arch Otorhinolaryngol 2006; 263:100.
18. Bali T, Saxena S, Kumar D, Nath R: 25. Kumar A, Tiwari HK, Singh RP, et al: 32. Misra UK, Jha S, Kalita J, Sharma K:
Response time and safety profile of pulsed Comparative evaluation of early vs. Stroke: a rare presentation of Eales’
oral methotrexate therapy in idiopathic deferred vitrectomy in Eales’ disease. Acta disease. A case report. Angiology 1996;
retinal periphlebitis. Eur J Ophthalmol Ophthalmol Scand 2000; 78:77. 47:73.
2005; 15:374. 26. Renie WA, Murphy RP, Anderson KC, et al: 33. Atabay C, Erdem E, Kansu T, Eldem B:
19. Agrawal S, Agrawal J, Agrawal T: The evaluation of patients with Eales Eales disease with internuclear
Intravitreal triamcinolone acetonide in disease. Retina 1983; 3:243. ophthalmoplegia. Ann Ophthalmol 1992;
Eales’ disease. Retina 2006; 26:227. 27. Rajesh M, Sulochana KN, Sundaram AL, 24:267.
20. Spitznas M, Meyer-Schwickerath G, et al: Presence of a 88 kDa Eales protein in 34. Rangasetty UC, Tyagi S, Mukhopadhyay S,
Stephan B: Treatment of Eales disease with uveitis, tuberculosis, leprosy and rheumatoid et al: Isolated extra-cranial internal carotid
photocoagulation. Graefes Arch Clin Exp arthritis. Med Sci Monit 2003; 9:95. artery aneurysm in a young adult with
Ophthalmol 1975; 194:73. 28. Rengarajan K, Muthukkaruppan VR, Eale’s disease. J Assoc Phys India 2003;
21. Magargal LE, Walsh AW, Magargal HO, Namperumalsamy P: Biochemical analysis 51:830.
Robb-Doyle E: Treatment of Eales disease of serum proteins from Eales patients. Curr 35. Muthukkaruppan V, Rengarajan K,
SECTION 10

with scatter photocoagulation. Ann Eye Res 1989; 8:1259. Chakkalath HR, Namperumalsamy P:
Ophthalmol 1989; 21:300. 29. Saxena S, Rajasingh J, Biswas S, et al: Immunological status of patients of Eales
22. Das T, Namperumalsamy P: Combined Cellular immune response to retinal disease. Indian J Med Res 1989; 90:351.
photocoagulation and cryotherapy in S-antigen and interphotoreceptor retinoid-
treatment of Eales retinopathy. Indian J binding protein fragments in Eales’ disease
Ophthalmol 1987; 35:108. patients. Pathobiology 1999; 67:39.

1840
CHAPTER

138 Retinal Arterial Macroaneurysms


Diana V. Do, Quan Dong Nguyen, and Julia A. Haller

Aneurysmal alterations of the retinal vasculature are a frequent Hemorrhage can also occur within the retina. Often a typical
finding in clinical ophthalmic practice. These changes most com- ‘hourglass hemorrhage’ occurs, consisting of simultaneous sub-
monly involve the retinal veins or capillaries and are usually retinal and preretinal collections of blood. Serous fluid can collect
seen as a sequel to diabetes mellitus, venous occlusive disease, within the retina, producing diffuse, focal, or cystoid macular
sickle-cell disease, or radiation retinopathy. Less commonly, edema, or it can accumulate in the subretinal space, detaching
larger aneurysms arise directly from the major retinal arteries. the macula and producing a gradual diminution in visual acuity.
Although mentioned sporadically in the earlier literature,1,2 the Lipid exudates can also cause a gradual decrease in vision by
entity currently known as retinal arterial macroaneurysms was migrating into the macula (Fig. 138.2).
formally described in 1973 by Robertson.2 Secondary epiretinal membranes may form as subhyaloid or
subinternal limiting membrane hemorrhage resorbs, and these
DEFINITION AND DESCRIPTION can cause persistent decreased visual acuity even after the macro-
aneurysm itself and bleeding/exudate caused by it have resolved.
Retinal arterial macroaneurysms may be defined as fusiform or If the macroaneurysm is large and occurs at an arteriovenous
saccular dilatations of the retinal arteries, usually arising within crossing, a branch retinal vein occlusion may be produced sec-
the first three orders of bifurcation. Their diameter exceeds ondarily, and symptoms may result from manifestations of the
100 mm (arbitrarily the upper limit of typical microaneurysms) vein occlusion or from the macroaneurysm, or both.
but generally is not greater than ~250 mm. Although most cases
are unilateral, 10% of patients have bilateral disease. Multiple ANGIOGRAPHIC FINDINGS
aneurysms occur in ~20% of affected eyes and usually involve
different arteries in the same eye. Patients may also present The most common fluorescein angiographic appearance of a
with multiple aneurysms along the same artery. The most com- macroaneurysm is uniform filling during the early arterial phase
monly reported site of involvement is along the superotemporal of the angiogram. Partial filling may occur if the aneurysm is spon-
arcade because individuals with aneurysms at this location are taneously involuting or is partially thrombosed (Fig. 138.3). The
more likely to have visual symptoms.2a Rarely, macroaneurysms late phase of the fluorescein angiogram may reveal leakage from
can occur directly on the optic nervehead3–5 or arise from a cilio- the macroaneurysm or staining of the vessel wall. The involved
retinal artery.6,7 They often occur at bifurcation sites and at artery is typically patent but may be narrowed proximal and
arteriovenous crossings and have been seen to develop in vessels distal to the macroaneurysm, although obliteration of the distal
with a documented history of embolic damage.7–9 Approximately portion of the artery has been reported.13 The area surrounding
10% of macroaneurysms are pulsatile on initial presentation,
but the literature is discordant as to whether this is a sign of
impending rupture.10–12

EPIDEMIOLOGY
The typical patient presenting with a macroaneurysm is a woman,
greater than 60 years of age, with an established history of sys-
temic hypertension. The female preponderance is on the order
of 3:1.2,10,11 Approximately 75% of patients have a history of hyper-
tension. Hypertension and arteriosclerotic vascular disease are
the only consistent disease associations in patients harboring
macroaneurysms.

FUNDUS APPEARANCE
The presentation of a patient with a macroaneurysm is variable.
Retinal arterial macroaneurysms can be found on routine exam-
ination in asymptomatic patients. Sudden, severe visual loss
may result from rupture of the aneurysm and resultant hemor-
rhage into the subretinal space, the subinternal limiting mem-
brane space, the retrohyaloid space, or the vitreous (Fig. 138.1). FIGURE 138.1. Large hemorrhage obscures the macroaneurysm. 1841
RETINA AND VITREOUS

FIGURE 138.2. (a and b) Peripherally located


aneurysm with lipid exudate involving the fovea.

a b
SECTION 10

ations have led investigators to at least three hypotheses – focal


damage to the blood vessel wall, chronic hypertensive vascular
damage, and inherent structural defects in blood vessels.
Lewis and colleagues9 observed a patient who developed
macroaneurysm at the site of a documented incomplete embolic
occlusion of a retinal artery. The authors postulated that embolic
injury results in focal damage to the blood vessel wall, causing
weakening of the wall and subsequent aneurysm formation.
Wiznia7 reported a macroaneurysm occurring in a cilioretinal
artery after an embolic occlusion of the artery. Ohga and asso-
ciates14 described a case of a macroaneurysm in an artery that
crossed over a toxoplasmosis scar. Periarteritis from the toxo-
plasmic inflammation may have caused structural damage that
weakened the arterial wall and caused an aneurysm to form.
The role of previous vascular damage was also suggested by the
study of Panton and co-workers.14a They noted a high concordance
rate between the location of macroaneurysms and that of branch
retinal vein occlusions when these occurred in the same eye.
Macroaneurysms occurred 12 times more often in the quadrant
served by the affected vein than elsewhere. In two eyes, the branch
vein occlusion was known to have preceded development of the
FIGURE 138.3. Fluorescein angiogram shows rapid, incomplete filling macroaneurysm.
of the macroaneurysm, which is consistent with partial thrombosis. Retinal arterial macroaneurysms have been noted to be
similar to intracerebral miliary aneurysms seen in elderly
hypertensive patients, especially women.2,11 Since hypertension
the macroaneurysm often shows capillary microaneurysms and is commonly associated with retinal arterial macroaneurysms,
nonperfusion, intraretinal microvascular abnormalities, telang- it is believed that chronic vascular wall damage caused by
iectasis, and fluorescein dye leakage. hypertension and associated arteriosclerotic changes predispose
Occasionally, fluorescein angiography may not demonstrate a the vessels to focal dilatation in the presence of continued
macroaneurysm because of blockage by overlying hemorrhage. increased intraluminal pressure.
A high index of suspicion combined with serial examinations and Lavin and colleagues11 noted that at the point of arteriovenous
angiography as the hemorrhage clears will result in an accurate crossing, there is no adventitia and the two blood vessels share
diagnosis. When hemorrhage prevents view of underlying struc- a common coat. The arterial wall thus has less support at these
tures, indocyanine green angiography may be useful in viewing the locations, and with increased intraluminal pressure in the
macroaneurysm because the absorption and emission spectra of hypertensive patient, ectasia of the arterial wall may result.
indocyanine green are in the near-infrared range, and the dye After development of the macroaneurysm, symptoms are
can often be seen through hemorrhage, allowing views of struc- produced by leakage through the thinned wall of the lesion as
tures that would otherwise be obscured. A retrospective case well as from the surrounding microvascular alterations that are
series using indocyanine green angiography in eyes with dense usually present. Histopathologically, actual aneurysmal sites
preretinal and subretinal hemorrhages demonstrated that show thickening of the arterial walls secondary to a fibrin-
indocyanine green angiography could correctly diagnose retinal laminated clot accompanied by hypertrophy of the muscular
macroaneurysms as the cause of hemorrhage when fluorescein layer. A thrombus will often partially or entirely fill the macro-
angiography was inconclusive.13a aneurysm. Thickened, hyalinized arterial walls are common in
adjacent arterioles. The areas surrounding the macroaneurysm,
PATHOGENESIS AND PATHOLOGIC clinically noted to show microvascular changes such as capillary
APPEARANCE loss, telangiectasias, and products of vascular leakage, typically
show corroborating histologic findings including dilatation of
The exact pathogenetic mechanism of macroaneurysm forma- the capillary bed, hemorrhage, lipid, edema, and photoreceptor
1842 tion is uncertain. However, several observations and associ- degeneration.14b
Retinal Arterial Macroaneurysms

NATURAL HISTORY In the group of patients with chronic decompensation, several


macroaneurysms were treated with photocoagulation because
The natural history of macroaneurysms varies and is dependent edema and lipid exudate had already involved the central macular
on the clinical presentation. In some patients with hemorrhage area and caused decreased visual acuity. In five subjects in whom
and no involvement of the macula, the visual acuity is not affected macular function was not threatened, no treatment was under-
and the hemorrhage can resolve without any adverse effects taken. None of these cases ever progressed to involve the macula,
(Fig. 138.4). although only one spontaneously closed during the study.
Abdel-Khalek and Richardson10 reported the natural history After closure of these chronically decompensating aneurysms,
of 18 cases of macroaneurysm, segregating them by clinical either spontaneously or as a result of photocoagulation, kinking
presentation into acute and chronic decompensation. Acute occurred at the aneurysmal site. Arteriolar constriction also
decompensation was typified by hemorrhage into the retina, occurred, usually proximal but occasionally distal to the macro-
subretinal space, subhyaloid space, or vitreous, and it produced aneurysm site. Arterial sheathing distal to the macroaneurysm
sudden visual loss. Chronic decompensation was characterized was commonly seen, but distal arterial closure was rare, even
by more gradual visual loss from the accumulation of macular after therapy.
edema and lipid exudate. These authors reported a relatively good visual prognosis
After presumed aneurysmal rupture associated with the acute with macroaneurysms presenting with acute hemorrhage, as
decompensation category, several pathways were followed. In have some other investigators.11,12 Of the eight patients in this
some subjects, the arterial perforation closed, leaving an intact category, only two patients had a final visual acuity of 6/18 or

CHAPTER 138
aneurysm that could rebleed or continue to leak fluid and exudate. worse.10 The remaining six patients had visual acuity of 6/12 or
Other aneurysms spontaneously closed, forming Z-shaped kinks better. Of the patients with chronic aneurysmal decompen-
at the former aneurysm site (Fig. 138.5). Subjects with only an sation, one patient had visual acuity of 6/60, two patients had
intraretinal hemorrhage often experienced a yellow-gray, saddle- visual acuity of 6/36, and one patient had visual acuity of 6/18,
shaped plaque centered on the macroaneurysm, with surrounding with only six patients arriving at visual acuity of 6/12 or better.
exudate. The involved arteriole often became heavily sheathed. These authors concluded that acute hemorrhage infrequently

FIGURE 138.4. (a) Subretinal hemorrhage not


involving the fovea. (b) Same eye as in (a)
6 weeks later without treatment.

a b

FIGURE 138.5. Spontaneous closure of the


macroaneurysm seen in Figure 138.1. (a) At
6 days. (b) At 20 days. (c) At 42 days. (d) At
close to 6 months after Figure 138.1.
(e) Fluorescein angiogram shows kinks at the
former aneurysm site.

a b

c d e
1843
RETINA AND VITREOUS

leads to marked decreases in visual acuity, whereas chronic retinal arterial macroaneurysms and are not always easily
macular edema and exudate commonly lead to a poorer visual distinguishable. Spalter20 called retinal arterial macroaneurysms
outcome. another ‘masquerade’ syndrome.
Palestine and associates12 investigated the natural course of Microaneurysms such as those occurring secondary to
macroaneurysms using a similar strategy. They divided their diabetes mellitus, radiation retinopathy, sickle-cell disease, and
patients into three groups. Group A included eyes with branch retinal vein occlusion are usually not confused with
hemorrhage, exudate, edema, or the macroaneurysm within the retinal arterial macroaneurysms. They are smaller and the
vascular arcades, accompanied by decreased visual acuity. nature of the underlying disease is often well established.
Group B included eyes with hemorrhage, exudate, edema, or the Schulman and colleagues21 described large capillary macro-
macroaneurysm within the arcades, but without an effect on aneurysms occurring secondary to retinal venous obstructive
visual acuity. Group C included eyes in which all the pathologic disease. These were similar in size to macroaneurysms but ori-
features were peripheral to the vascular arcades. These ginated from the venous side of the capillary bed. They caused
investigators concluded that eyes in group A could do poorly decreased visual acuity as a result of macular edema and exudate
and that the prognosis was variable. Eyes in group B did better as well as serous elevation of the macula, mechanisms shared
but needed to be observed periodically to be certain they did not in common with retinal arterial macroaneurysms. Parodi and
convert to the group A type of eyes. Group C eyes typically did co-workers22 reported multiple 150–300 mm capillary macro-
well without therapy. These investigators also noted that eyes aneurysms in an eye with central retinal vein occlusion.
presenting with acute vitreous or subinternal limiting A variety of macroaneurysms has been reported after branch
SECTION 10

membrane hemorrhage tended to do well. retinal vein occlusion.23,24 Cousins and associates24 reviewed
Cleary and co-workers13 reviewed the natural history of the photographs and fluorescein angiograms of patients who
macroaneurysms in 20 patients. They observed that the macro- participated in the Collaborative Branch Vein Occlusion Study
aneurysms almost always closed spontaneously after acute through the Bascom Palmer Eye Institute. They noted that four
hemorrhage, whereas the lesions rarely did so when macular types of macroaneurysm could be seen after branch retinal vein
edema was the presenting pathologic condition. The authors occlusion. Typical retinal arterial macroaneurysms, indis-
concluded that the prognosis was good in patients presenting tinguishable from the idiopathic variety, were common and
with hemorrhage and poorer in those presenting with macular occurred within the zone of the branch retinal vein occlusion.
edema. Both the degree of chronicity and the severity of the Venous macroaneurysms were similar in size but slower to fill
macular edema affected the prognosis. angiographically because they occurred along the obstructed
McCabe and associates reviewed 41 patients who presented vein. Capillary aneurysms were also seen, similar to those
with macular hemorrhage secondary to macroaneurysms and reported by Schulman and colleagues.21 Finally, they described a
were managed with observation.15 After an average follow-up of new type of macroaneurysm that they called the collateral-
15.7 months, a visual acuity of 20/40 was achieved in 37%, associated macroaneurysm. These were associated with clearly
between 20/50 and 20/100 in 29%, and 20/200 or worse in 34%. identifiable dilated collateral vessels on fluorescein angiography.
Poorer visual acuity outcomes were associated with macular All four types of these macroaneurysms were associated with
pigmentary changes after resorption of blood. intraretinal lipid or hemorrhagic exudation, often involving the
More recently, Yang and colleagues conducted a retrospective macula. On fluorescein angiography, the macroaneurysms were
review of arterial macroaneurysms followed at their institution typically found to be present in areas of capillary nonperfusion.
over a 17-year period.16 The authors classified the 31 patients Eighty-four percent of these eyes were classified as having severe
in their study as having either predominantly hemorrhagic or nonperfusion according to the criteria of the Branch Vein
exudative macroaneurysms. Eyes with hemorrhagic lesions had Occlusion Study.
a greater improvement in vision after resolution of the blood There has been one case report of a spontaneous venous
compared to eyes in the exudative cohort, and foveal exudate macroaneurysm occurring without an antecedent vein occlusion.
was a statistically significant risk factor for final visual acuity Brourman and colleagues25 described a macroaneurysm of the
equal to or worse than 6/30. proximal superotemporal vein in a patient with pericarditis.
Although arterial closure is not a common occurrence, some They suggested that increased venous pressure may have been
investigators have described a significant number in their case a causative factor.
series. Panton and co-workers14a reported an 8% rate of branch Branch retinal vein occlusions have been reported to
retinal artery occlusion in untreated cases of macroaneurysm masquerade as retinal arterial macroaneurysms; this is known
and a 16% incidence after treatment. There has been at least as the Bonnet sign.26 The classic Bonnet sign consists of
one other case report of spontaneous branch retinal artery intraretinal hemorrhage at an arteriovenous crossing simulating
occlusion.17 a macroaneurysm.
As already noted, a question has been raised in the literature There is one report of a retinal arterial macroaneurysm
regarding the significance of pulsation of the macroaneurysm. presenting as a mass lesion of the optic nerve; only with serial
Some investigators consider this a sign of imminent follow-up was the proper diagnosis ultimately made.3 There has
rupture,10,18,19 although others believe that it has no relation- been one case report of Valsalva’s retinopathy occurring as the
ship to eventual rupture and that there are no reliable signs of result of a ruptured retinal arterial macroaneurysm.27
impending rupture.11 Before retinal arterial macroaneurysms became a familiar
clinical entity, they were commonly confused with retinal
DIFFERENTIAL DIAGNOSIS telangiectasis, Leber’s miliary aneurysms, or Coats’ disease.
These patients have unilateral disease consisting of multiple
In cases uncomplicated by a poor view of the fundus, the diag- telangiectatic vessels in the midperiphery, predominantly on
nosis of retinal arterial macroaneurysm is easily established. the venous side of the circulation and usually present in
Often, however, the true nature of the pathologic condition is childhood, often with massive exudation including exudative
obscured by hemorrhage, whether in the vitreous, on the surface retinal detachment.
of the retina, or within the retina itself. A typical hourglass type von Hippel’s angiomatosis usually shows a distinct genetic
of hemorrhage should alert the clinician to the possibility of a component. When small, the angiomas may be confused with
1844 macroaneurysm. In addition, several entities closely resemble macroaneurysms, but when well developed, they are typically
Retinal Arterial Macroaneurysms

much larger and accompanied by large, tortuous feeder and involve or already involve the central macula should be
draining vessels. Similar-sized retinal angiomas may occur considered for therapy (Fig. 138.7). Treatment has been shown
unassociated with von Hippel’s disease; they do not have pro- to shorten the duration of macroaneurysm patency.11 The
minent feeder and drainage vessels unless the tumors become duration of the hemorrhage and exudate as well as its severity
quite large. will determine the ultimate visual outcome.
A common presenting situation is that of an elderly patient Abdel-Khalek and Richardson10 treated macroaneurysms with
with an acute subretinal hemorrhage in the macula. There is direct photocoagulation using the argon laser and xenon arc.
often a surrounding area of subretinal lipid exudate, and the Two of their patients also received perianeurysmal treatment
appearance can closely mimic neovascular age-related macular preceding the direct treatment. No bleeding or complications
degeneration. The macroaneurysm may be obscured by the occurred from the direct therapy. The macroaneurysms were
retinal hemorrhage. A high index of suspicion and careful closed in all cases. Some investigators recommend treatment to
examination of the fellow eye, coupled with indocyanine green be undertaken early in the course of the disease because the
angiography may lead to the proper diagnosis, confirmed after longer the edema and exudate are present, the less the chance
hemorrhage resolution. of improving the visual acuity and preventing permanent
Since ~10% of retinal arterial macroaneurysms present with macular damage. Closure of the macroaneurysm has been
vitreous hemorrhage, this diagnosis must be suspected when no shown to enhance the rate of resorption of retinal exudate,9
retinal detachment, retinal tear, or avulsed retinal vessel is although the exudate may initially appear to worsen or increase
found after the vitreous hemorrhage clears. The retinal arterial because of more rapid and selective resorption of retinal edema

CHAPTER 138
macroaneurysm may involute before adequate fundus viewing fluid, which may cause further precipitation of lipid exudate.
and leave only subtle telltale signs of its presence as the cause Palestine and associates12 treated several eyes with macro-
of the vitreous hemorrhage. aneurysms that had produced macular edema and exudate.
If large subretinal or subretinal pigment epithelial hemorr- They specifically advised against treating the aneurysm or the
hage occurs, this presentation may be mistaken for a choroidal artery directly because of the possibility of rupture of the
melanoma (Fig. 138.6). aneurysm, although they acknowledged that this complication
had not been reported. These investigators, as did Lewis and
MANAGEMENT colleagues,9 treated the area of the microvascular changes sur-
rounding the aneurysm and found that this predictably produced
Although there have been no prospective, randomized clinical involution of the aneurysm. They also noted an early increase
trials involving laser photocoagulation treatment of retinal in lipid exudation, as less protein-rich fluid was initially resorbed.
arterial macroaneurysms, empirical treatment guidelines have Complete resolution of the exudate took several months.
been established by numerous investigators. Many investigators Lavin and colleagues11 treated hemorrhagic and exudative
apply laser photocoagulation directly to macroaneurysms macroaneurysms when the macula was involved, with a combi-
associated with exudates, macular edema, and visual acuity nation of direct and perianeurysmal techniques using the argon
decrease. Although advances in vitreous surgery and the laser. They achieved closure of the macroaneurysms without
development of adjunctive pharmacologic agents have made the complications and again noted that the visual prognosis
removal of submacular hemorrhage technically feasible, and depended on the degree of intraretinal hemorrhage and exudate
good results with this strategy have been reported, the useful- in the central macula.
ness of surgical intervention has not been established as yet. Joondeph and co-workers35 treated macroaneurysms using
Natural history studies suggest that spontaneous resorption of the yellow dye laser. They used a technique of partially over-
blood usually occurs with relatively good visual outcomes.28–33 lapping burns applied to just the peripheral margin of the
Most authorities agree that a quiet macroaneurysm that is macroaneurysm, sparing the feeding and draining arterioles.
asymptomatic without visible leakage should not be treated but They successfully closed all the macroaneurysms in one treat-
should continue to be monitored until spontaneous fibrosis ment and reported no complications of therapy.
occurs. Ghost macroaneurysms (i.e., those that are already Mainster and Whitacre36 also used the yellow dye laser with
spontaneously fibrosed and show the kinked-vessel Z-sign or a similar technique to successfully close five macroaneurysms.
the saddle-shaped plaque) also should not be treated, as they They also reported no complications. However, Russell and
have run their natural course and will remain quiescent. Folk,37 after having successfully treated 15 macroaneurysms
Similarly, macroaneurysms that have ruptured and bled acutely with the argon laser, used the yellow dye laser on one patient
rarely rebleed,34 so observation is adequate in these cases. and immediately produced a branch retinal artery occlusion
Aneurysms that leak fluid or exudate, or both, and threaten to after applying only four burns to the macroaneurysm. Their

a b c

FIGURE 138.6. Patient with a large subretinal and subpigment epithelial hemorrhage referred to as a choroidal melanoma. (a) At presentation.
(b) Fluorescein angiogram at presentation. (c) Appearance 5 months later. 1845
RETINA AND VITREOUS

FIGURE 138.7 Treatment algorithm for retinal


Arterial macroaneurysm arterial macroaneurysm.

Exudate and macular edema Not threatening fovea Hemorrhage

(Decreased visual acuity) (Asymptomatic) Into fovea

Consider laser photocoagulation Observe Observe

treatment technique was not well described but appeared to and colleagues39 used a Q-switched neodymium:yttrium–
involve a more direct treatment to the macroaneurysm than did aluminum garnet laser to release a retrohyaloid hemorrhage
either of the previous two reports involving the yellow dye laser. resulting from a ruptured macroaneurysm, thus facilitating
SECTION 10

Recent work by Brown and associates38 suggested that direct more rapid resorption of the blood than would have occurred
laser treatment of macroaneurysms may be harmful and, in spontaneously. Raymond40 reported two cases in which
their study, it involved a significant risk factor for poor visual subinternal limiting membrane hemorrhage from macro-
outcome. In a retrospective study, they compared the long-term aneurysms was liberated into the vitreous with the same type of
visual outcome between patients treated with direct laser photo- laser.
coagulation and untreated patients. They treated eight eyes Subfoveal hemorrhage, whether from a macroaneurysm or
with argon green and eight eyes with yellow dye lasers. Even associated with other pathology, has traditionally been believed
when statistically controlling for the effect of subfoveal hemor- to have a uniformly poor prognosis for visual acuity. Advances
rhage and subfoveal lipid, laser photocoagulation remained the in vitrectomy surgery have attempted to address this problem.
strongest predictor for final visual acuity of less than 20/80. Hanscom and Diddie28 first reported drainage of submacular
They noted four cases of branch retinal artery occlusion, at least hemorrhage resulting from ruptured retinal arterial macro-
two of which led to further foveal capillary dropout and subsequent aneurysms in 1987. There have been numerous subsequent
decreased visual acuity. They suggested that indirect treatment reports, some of which described the adjunctive use of tissue
may lower the complication rate and show better results. plasminogen activator to evacuate submacular hemorrhage in
Panton and co-workers14a observed a 16% rate of branch this situation.29–33 None of these reports described more than four
retinal artery occlusion after laser photocoagulation, but noted eyes with subfoveal hemorrhage. Patients showed improvement
that none occurred with exclusively perianeurysmal treatment. from their preoperative visual acuities, but it has yet to be deter-
They also acknowledged that other investigators believed that mined whether this aggressive approach improves on the natural
direct treatment was riskier and more likely to cause arterial history, even for this serious presentation of macroaneurysms.
closure than were indirect techniques. There can be significant and sometimes marked improvement
Chaum and colleagues reported the development of retino- in vision even without treatment, although natural history data
choroidal anastomoses and choroidal neovascularization (CNV) are scant. Berrocal and associates41 studied variations in the
in one patient after applying laser photocoagulation directly to clinical course of subfoveal hemorrhages. Two of their patients
a macroaneurysm.38a Although the CNV was not located directly had hemorrhage due to macroaneurysms. One did poorly, but
beneath the site of the photocoagulation, the authors hypo- the other recovered to 20/30 from an initial vision of 5/200. The
thesize that the laser treatment induced changes in the blood authors recently observed a case of subretinal hemorrhage
flow in the macula which induced retinochoroidal anastomoses extending beneath the fovea to cause counting fingers visual
formation and CNV. acuity with eventual spontaneous recovery to 20/40 (Fig. 138.8).
These aforementioned treatment techniques are routinely used Although vitreous hemorrhage from macroaneurysms typically
for managing the vision-threatening consequences of macro- clears rapidly, pars plana vitrectomy surgery is effective for the
aneurysms. Ancillary techniques have been reported. Tassignon occasional case of dense, nonclearing vitreous hemorrhage.33

a b c

FIGURE 138.8. Patient with subretinal hemorrhage extending into the fovea. (a) Vision is counting fingers. (b) Early resorption of hemorrhage.
1846 (c) Vision is 20/40 10 weeks after (a).
Retinal Arterial Macroaneurysms

SUMMARY involvement with hemorrhage, exudate, edema, or serous detach-


ment or from vitreous or preretinal hemorrhage. Selected patients
Retinal arterial macroaneurysms are a well-defined retinal vascu- may be successfully treated using laser photocoagulation, but the
lar disorder consisting of saccular or fusiform dilatations of the visual prognosis cannot be reliably predicted in individual cases.
major retinal arterioles, usually within the first three orders of
bifurcation. They usually occur in older, hypertensive women. Key Features
Diagnosis can sometimes be difficult, and a high index of • Fusiform or saccular dilation of the retinal artery
suspicion is required. Indocyanine green angiography may be • Arises within the first three orders of artery bifurcation
helpful in cases where hemorrhage obscures the macroaneurysm. • Most commonly found along superotemporal arcade
Their natural history is often benign, with many progressing to • 90% of cases are unilateral
spontaneous fibrosis and involution with retention of good visual • Often associated with hypertension and arteriosclerosis
acuity. The visual acuity is decreased as the result of macular

REFERENCES
1. Rabb MF, Gagliano DA, Teske MP: Retinal 14a. Panton RW, Goldberg MF, Farber MD: 29. Peyman GA, Nelson NC, Alturki W, et al:
arterial macroaneurysms. Surv Ophthalmol Retinal arterial macroaneurysms: risk Tissue plasminogen activating factor
1988; 33:73. factors and natural history. Br J Ophthalmol assisted removal of subretinal hemorrhage.

CHAPTER 138
2. Robertson DM: Macroaneurysms of the 1990; 74:595. Ophthalmic Surg 1991; 22:575.
retinal arteries. Trans Am Acad Ophthalmol 14b. Fichte C, Streeten BW, Friedman AH: 30. Moriarty AP, McAllister IL, Constable IJ:
Otolaryngol 1973; 77:OP55. A histopathologic study of retinal arterial Initial clinical experience with tissue
2a. Moosavi RA, Fong KCS, Chopdar A. aneurysms. Am J Ophthalmol 1978; plasminogen activator (tPA) assisted
Retinal artery macroaneurysms: clinical and 85:509. removal of submacular hemorrhage. Eye
fluorescein angiographic features in 34 15. McCabe C, Flynn HW, McLean WC, et al: 1995; 9:582.
patients. Eye 2006; 20:1011–1020. Nonsurgical management of macular 31. Lim JI, Drews-Botsch C, Sternberg P, et al:
3. Brown GC, Weinstock F: Arterial hemorrhage secondary to retinal artery Submacular hemorrhage removal.
macroaneurysm on the optic disc macroaneurysms. Arch Ophthalmol 2000; Ophthalmology 1995; 102:1393.
presenting as a mass lesion. Ann 118:780–785. 32. Ibanez HE, Williams DF, Thomas MA, et al:
Ophthalmol 1985; 17:519. 16. Yang CS, Tsai DC, Lee FL, Hsu WM: Surgical management of subretinal
4. Kowal L, Steiner H: Arterial macroaneurysm Retinal arterial maacroaneurysms: risk hemorrhage: a series of 47 consecutive
of the optic disc. Aust N Z J Ophthalmol factors of poor visual outcome. cases. Arch Ophthalmol 1995; 113:62.
1991; 19:75. Ophthalmologica 2005; 219:366–372. 33. Brent BD, Gonce M, Diamond JG: Pars
5. Iwasawa A, Majima A, Shirai S, et al: Bilateral 17. Oo A, Saito Y, Okamoto S, et al: Retinal plana vitrectomy for complications of
prepapillary macroaneurysm. Report of a macroaneurysm and branched retinal artery retinal arterial macroaneurysms – a case
case. Jpn J Ophthalmol 1989; 43:619. occlusion. Ther Res 1989; 10:11. series. Ophthalmic Surg 1993; 24:534.
6. Giuffre G, Montalto FP, Amodei G: 18. Psinakis A, Kokolakis PG, Theodossiadis C, 34. Nadel AJ, Gupta KK: Macroaneurysms of
Development of an isolated retinal et al: Pulsatile retinal arterial macroaneurysm: the retinal arteries. Arch Ophthalmol 1976;
macroaneurysm of the cilioretinal artery. Br treatment with argon laser photocoagulation. 94:1092.
J Ophthalmol 1987; 71:445. J Fr Ophtalmol 1989; 12:673. 35. Joondeph BC, Joondeph HC, Blair NP:
7. Wiznia RA: Development of a retinal artery 19. Yokoi N, Tohsaka S, Yamamoto T: A case of Retinal macroaneurysms treated with the
macroaneurysm at the site of a previously pulsating retinal arterial macroaneurysm. yellow dye laser. Retina 1989; 9:187.
detected retinal artery embolus. Am J Folia Ophthalmol Jpn 1990; 41:584. 36. Mainster MA, Whitacre MM: Dye yellow
Ophthalmol 1982; 114:642. 20. Spalter HF: Retinal macroaneurysms: a photocoagulation of retinal arterial
8. El-Asrar AMA, Awad A, Tabbara KF: Retinal new masquerade syndrome. Trans Am macroaneurysms. Am J Ophthalmol 1988;
artery macroaneurysm in a patient with Ophthalmol Soc 1982; 80:113. 105:97.
congenital heart disease. Br J Ophthalmol 21. Schulman J, Jampol LM, Goldberg MF: 37. Russell SR, Folk JR: Branch retinal artery
1993; 77:606. Large capillary aneurysms secondary to occlusion after dye yellow
9. Lewis RA, Norton MH, Wise GN: Acquired retinal venous obstruction. Br J Ophthalmol photocoagulation of an arterial
arterial macroaneurysms of the retina. Br J 1981; 65:36. macroaneurysm. Am J Ophthalmol 1987;
Ophthalmol 1976; 60:21. 22. Parodi MB, Bondel E, Ravalico G: Capillary 104:186.
10. Abdel-Khalek MN, Richardson J: Retinal macroaneurysms in central retinal vein 38. Brown DM, Sobol WM, Folk JC, et al:
macroaneurysm: natural history and occlusion. Ophthalmologica 1995; 209:248. Retinal arteriolar macroaneurysms: long
guidelines for treatment. Br J Ophthalmol 23. Sanborn GE, Magargal LE: Venous term visual outcome. Br J Ophthalmol
1986; 70:2. macroaneurysm associated with branch 1994; 78:534.
11. Lavin MJ, Marsh RJ, Peart S, et al: Retinal retinal vein obstruction. Ann Ophthalmol 38a. Chaum E, Greenwald MA: Retinochoroidal
arterial macroaneurysms: a retrospective 1984; 16:464. anastomoses and a choroidal neovascular
study of 40 patients. Br J Ophthalmol 24. Cousins SW, Flynn HW, Clarkson JG: membrane in a macular exudate following
1987; 71:817. Macroaneurysms associated with retinal treatment for retinal macroaneurysms.
12. Palestine AG, Robertson DM, Goldstein branch vein occlusion. Am J Ophthalmol Retina 2002; 22:363–366.
BG: Macroaneurysms of the retinal arteries. 1990; 109:567. 39. Tassignon MJ, Stempels N, Van Mulders L:
Am J Ophthalmol 1982; 93:164. 25. Brourman ND, Goldberg RE, Augsburger Retrohyaloid premacular hemorrhage
13. Cleary PE, Kohner EM, Hamilton AM, et al: JJ, et al: Isolated venous macroaneurysm. treated by q-switched Nd-YAG laser.
Retinal macroaneurysms. Br J Ophthalmol Ophthalmic Surg 1990; 21:646. Graefes Arch Clin Exp Ophthalmol 1989;
1975; 59:355. 26. Kimmel AS, Magargal LE, Morrison DL, 227:440.
13a. Townsend-Pico WA, Meyers SM, Lewis H: et al: Temporal branch retinal vein 40. Raymond LA: Neodymium:YAG laser
Indocyanine green angiography in the obstruction masquerading as a retinal treatment for hemorrhages under the
diagnosis of retinal arterial arterial macroaneurysm: the Bonnet sign. internal limiting membrane and posterior
macroaneurysms associated with Ann Ophthalmol 1989; 21:251. hyaloid face in the macula. Ophthalmology
submacular and preretinal hemorrhages: a 27. Avins LR, Krummenacher TK: Valsalva 1995; 102:406.
case series. Am J Ophthalmol 2000; maculopathy due to a retinal arterial 41. Berrocal MH, Lewis ML, Flynn HW:
129:33–37. macroaneurysm. Ann Ophthalmol 1983; Variations in the clinical course of
14. Ohga H, Egi K, Katayama T, et al: A case of 15:421. submacular hemorrhage. Am J Ophthalmol
retinal macroaneurysm with congenital 28. Hanscom TA, Diddie KR: Early surgical 1996; 122:486.
ocular toxoplasmosis. Folia Ophthalmol drainage of macular subretinal hemorrhage.
Jpn 1990; 41:898. Arch Ophthalmol 1987; 105:1722.
1847
CHAPTER

139 Coats’ Disease and Retinal Telangiectasia


Grant M. Comer and Mark W. Johnson

PRIMARY OR CONGENITAL RETINAL by 20 years of age, with a median age at diagnosis of 5 years.10
TELANGIECTASIA (COATS’ DISEASE) Roughly 75% of cases occur in males, and 95% of cases are
limited to one eye with asymmetric involvement when bilateral.
Most cases are diagnosed after parents note decreased vision,
INTRODUCTION leukocoria or strabismus (Fig. 139.1).10 Milder cases may be
Coats’ disease (idiopathic retinal telangiectasia, Leber’s miliary detected at the time of routine ophthalmic examination.
aneurysms) is an idiopathic, progressive exudative retinopathy
consisting of incompetent retinal telangiectasia that results in a
variable clinical spectrum ranging from isolated vascular abnor- ETIOLOGY
malities with no exudation to extensive sub- and intraretinal Although numerous authors have described Coats’-like condi-
exudation and detachment. Initially described as separate entities, tions associated with a number of ocular and systemic diseases,
the milder (Leber’s miliary aneurysms)1 and more severe (Coats’ Coats’ disease, by definition, is isolated and idiopathic with no
disease)2,3 forms of this retinal vascular anomaly are now con- known inheritance pattern.10 One report suggested involvement
sidered by most authorities to be variable expressions of the same of the Norrie disease pseudoglioma (NDP) gene (Xp11.3), which
disease and are, therefore, currently grouped under the common produces norrin protein.13 Norrin affects retinal angiogenesis
name of Coats’ disease.4,5 by modification of endothelial cell development. Known
phenotypic manifestations of NDP gene mutations include
Norrie disease, X-linked primary retinal dysplasia, and X-linked
CLASSIFICATION exudative vitreoretinopathy, which all exhibit variable degrees of
Coats initially divided this entity into three varieties: type I congenital vascular abnormalities within the retina.14,15 Black
included cases of abnormal exudation without apparent and colleagues reported an NDP mutation in a mother with
vascular changes; type II included both exudation and abnormal Coats’ disease who gave birth to a son with Norrie disease and
vessels; and type III exhibited exudation surrounding a large suggested that a somatic NDP gene mutation may be
retinal angioma.2,3 With the advent of fluorescein angiography, responsible for Coats’ disease. They also found a mixture of
it became apparent that abnormal retinal vessels were present both normal and mutant NDP alleles within retinal tissue, but
in both types I and II. Type III was subsequently determined not nonretinal tissue, of an enucleated eye from a patient with
synonymous with von Hippel’s angiomatosis retina.6 Other Coats’ disease.13
authors6,7 have attempted to stage Coats’ disease based on
specific clinical findings at presentation.
More recently, reports have attempted to classify Coats’ disease
by associating the degree of retinal involvement with prognosis.
Cahill and associates classified Coats’ disease as severe, focal,
juxtafoveal, or associated with other diseases. While severe
Coats’ disease had a dismal prognosis, the other groups were
generally amenable to treatment with 67% achieving a visual
acuity at or better than presentation.8 Similarly, Shields and
co-authors classified Coats’ disease as: stage 1, telangiectasia only;
stage 2, telangiectasia and exudation (2A, extrafoveal; 2B foveal);
stage 3, exudative retinal detachment (3A, subtotal; 3B total);
stage 4, total detachment with secondary glaucoma; and stage 5,
advanced end-stage disease. Despite attempts at treatment,
visual prognosis decreased as the extent of retinal involvement
increased, especially once the fovea was involved.9

DEMOGRAPHICS
No strong evidence supports a specific racial or ethnic predis-
position or inheritance pattern.10 Coats’ disease has reportedly
occurred in patients ranging from 3 months of age11 to those in FIGURE 139.1. Leukocoria secondary to total exudative retinal
the eighth decade of life;12 however, the majority of cases occur detachment in a child with Coats’ disease. 1849
RETINA AND VITREOUS

asymptomatic in the periphery, vision usually becomes affected


CLINICAL PRESENTATION with the onset of hard exudates beneath the fovea, cystoid
Although the extent and rate of development of clinical findings macular edema, or exudative macular detachment (Fig. 139.4).
are variable, the disease is usually progressive.16 Primary abnor- Massive exudation contributes to numerous secondary abnor-
malities in Coats’ disease are confined to retinal vessels, with a malities throughout the eye. While often normal, the anterior
spectrum of secondary findings depending on the degree and segment can demonstrate corneal edema, megalocornea,
chronicity of vascular incompetence. Abnormalities can occur cholesterolosis in the anterior chamber, neovascularization of
in all levels of the vasculature and include: micro- and macro- the iris, cataract, peripheral anterior synechiae, angle closure,
aneurysms, capillary dilation (telangiectasis) and nonperfusion, and neovascular or secondary open-angle glaucoma.10,18,20,21
and saccular outpouchings of retinal venules.12,17 In mild cases, Secondary posterior segment abnormalities may include: retinal
one or more localized foci of retinal telangiectasia are noted hemorrhages, macrocysts, vasoproliferative tumors, neovascu-
within the retinal capillary bed, typically in the temporal quad- larization, vitreous hemorrhage, lamellar macular holes, and
rants between the equator and ora serrata; however, all preretinal membranes.10,12,18,22,23
quadrants, including the macula, can be affected (Fig. 139.2).18,19 The clinical presentation differs in adult patients in that the
In more advanced Coats’ disease, areas of lipoproteinaceous vascular abnormalities are often present in both the peripheral as
exudation develop adjacent to telangiectatic vessels, which have well as juxtamacular areas. In addition, adults more commonly
permeable endothelial cells, and slowly progress to involve the demonstrate localized lipid deposition, hemorrhage around
entire retina (Fig. 139.3a–c).19 With accumulation of exudate, a macroaneurysms, and slower disease progression.12 Long-term
SECTION 10

yellow or greenish-yellow subretinal deposit may develop in surveillance is important, even after treatment, as multiple
association with localized exudative retinal detachment. This recurrences are seen to occur in up to 33% patients with Coats’
subretinal lipid may coalesce into crystalline bodies and disease.24
multiple subretinal deposits. Intraretinal macular exudation
often occurs in a stellate pattern without connection to the
peripheral telangiectasis.19 With further progression, total
exudative retinal detachment may develop.10 Although often

FIGURE 139.2. Asymptomatic focus of retinal telangiectasia in a FIGURE 139.4. Massive lipid exudation in macula associated with
patient with mild Coats’ disease. inferior exudative retinal detachment.

a b c

FIGURE 139.3. (a) Retinal telangiectasia and exudation in the superotemporal quadrant. (b) Focal vascular incompetence and nonperfusion on
1850 composite fluorescein angiogram. (c) Subfoveal exudation extending from distant nidus of leakage as seen on optical coherence tomography.
Coats’ Disease and Retinal Telangiectasia

ANCILLARY TESTS TABLE 139.1 Differential Diagnosis of Coats’ Disease


Fluorescein angiography and optical coherence tomography Retinoblastoma
provide an objective measure of disease extent. Fluorescein
Familial exudative vitreoretinopathy
angiography demonstrates areas of capillary nonperfusion,
saccular and beadlike ‘light bulb’ dilatations of larger retinal Retinopathy of prematurity
vessels, and general dilatation (telangiectasia) of the capillary Retinal toxocariasis
bed (Fig. 139.5 a,b).19,25 Abnormal retinal vessels typically fill in
the late arterial or early venous phase and slowly leak Persistent hyperplastic primary vitreous
fluorescein, which may pool within the intraretinal spaces of Retinal angiomatosis
associated cystoid edema or within the subretinal space.19,25,26
Incontinentia pigmenti
Children as young as 11 months, under general anesthesia,27
and 4 years, unsedated,28 have tolerated optical coherence Pars planitis
tomography (OCT) to document and quantify subretinal fluid Congenital cataract
and macular edema to a resolution of 10 µm.29
Additional tests help differentiate Coats’ disease from ocular Malignant melanoma
tumors such as retinoblastoma, since a definitive diagnosis Choroidal metastasis
cannot be established from clinical examination alone in 20% of

CHAPTER 139
Choroidal hemangioma
cases.19,30 In A- and B-scan echography, advanced Coats’ disease
often demonstrates a narrow or closed funnel retinal detachment, Eccentric disciform age-related macular degeneration
poor retinal mobility, retinal thickening, low to medium reflective Exophytic retinal capillary hemangioma
subretinal opacities with a slow convection movement, and
absence of a solid mass lesion or calcium.31 Computed tomo- Branch retinal vein occlusion
graphy demonstrates subtle homogeneous subretinal densities Idiopathic acquired juxtafoveal telangiectasia
without contrast enhancement. This is differentiated from retino-
Other causes of acquired juxtafoveal telangiectasia (see Table 139.2)
blastoma which demonstrates foci of calcium and diffuse con-
trast enhancement of the mass.32 Subretinal fluid demonstrates
homogeneous hyperintensity on T2- more than T1-weighted
magnetic resonance images without further enhancement on Eales’ disease,41 retinal capillary hemangioma,19 retinal cavernous
contrast-enhanced sequences.30,33,34 In contrast, solid intraocular hemangioma,19 and various causes of vasculitis (Table 139.1).42
tumors greater than 2.0 mm in thickness, such as retinoblastoma, Retinoblastoma, which is potentially lethal if untreated, is the
demonstrate variable intensity on T1-weighted images and are most significant condition that must be differentiated from Coats’
hypointense on T2-weighted images.30,34 Calcification is disease. Coats’ disease generally presents as a unilateral disorder
accentuated on T2-weighted images.30 Finally, subretinal fluid in older boys with no family history, whereas retinoblastoma
aspiration can establish a cytopathologic diagnosis characterized is more often bilateral with no sex predilection and a positive
by lipid-laden macrophages and cholesterol crystals.35,36 family history.19 The presence of sub- and intraretinal calcifica-
tion strongly suggests retinoblastoma; however, limited reports
have confirmed the presence of subretinal43 and intraretinal
DIFFERENTIAL DIAGNOSIS calcium44 in enucleated eyes found to have Coats’ disease.
Since Coats’ disease spans a large spectrum of clinical present- True Coats’ disease is defined as an idiopathic, progressive exu-
ations and ages, the differential is vast. Coats’ disease often dative retinopathy with associated retinal telangiectasia. However,
presents in children as leukocoria and strabismus, as such, the numerous reports have described predominantly nonocular dis-
differential diagnosis includes retinoblastoma, toxocariasis, per- orders, in which Coats’-like changes can comprise an element of
sistent hyperplastic primary vitreous, advanced retinopathy of the condition, including pericentric inversion on chromosome 3,45
prematurity, congenital cataract, incontinentia pigmenti, endo- chromosome 13 deletion,46 fasciocapsulohumeral muscular
phthalmitis, Norrie disease, and inflammatory uveitis.14,18,19 In dystrophy,47 Turner ’s syndrome,48 Senior–Loken syndrome
both children and adults, other retinal telangiectatic and vascular (familial renal–retinal dystrophy),49 epidermal nevus syn-
diseases may also mimic Coats’ disease, including branch retinal drome,50 VATER association (vertebral defects, imperforate
vein occlusion,37 diabetic retinopathy,38 radiation retinopathy,39 anus, tracheoesophageal fistula, and radial and renal dysplasia),51
familial exudative vitreoretinopathy,18 juxtafoveal telangiectasia,40 congenital plasminogen deficiency type I,52 Hallermann–Streiff

a b

FIGURE 139.5. (a) Typical ‘light bulb’ dilations and (b) capillary nonperfusion with focal leakage in late-phase angiogram. 1851
RETINA AND VITREOUS

FIGURE 139.6. Cholesterol crystals of


subretinal fluid aspirate in a patient with Coats’
disease as visualized with H & E staining (a)
and polarized microscopy (b).

a b

syndrome,53 Coats’ plus syndrome,54 Cornelia de Lange syn- cryopexy. In more advanced cases, excessive exudate may
drome,55 Alport’s syndrome,56 renal transplantation,57 aplastic require drainage through a sclerotomy or retinotomy to allow
anemia,58 multiple glomus tumors,59 and telangiectasia of the for adequate treatment of the telangiectasis. As it is important
nasal mucosa.60 to treat the entire area of abnormal vessels, fluorescein angio-
Similarly, other reports have described ocular conditions graphy may aid in visualizing the full extent of vascular
SECTION 10

associated with separate Coats’-like changes, including: disease.71


idiopathic retinal gliosis,61 branch retinal vein occlusion,37 Shields and associates recommend management based on
retinitis pigmentosa,62,63 morning glory optic disk,64 uveitis,65 their staging scheme. Stage 1 disease, telangiectasia only, receives
ocular toxoplasmosis,66 and retinal dysplasia.67 either periodic observation or laser photocoagulation. Stage 2,
telangiectasia and exudation, is treated with cryopexy or laser
photocoagulation. Stage 3A, subtotal detachment, requires
PATHOLOGY cryopexy or laser photocoagulation; however, 3B, total detach-
Pathologic specimens usually come from globes enucleated ment, receives cryopexy for shallow detachments and either
because of advanced disease causing a blind and painful eye or scleral buckling or pars plana vitrectomy for more extensive
when retinoblastoma could not be excluded in the diagnosis.18 involvement. Stage 4, total retinal detachment and glaucoma,
These specimens allow extensive investigation into the primary usually requires enucleation to relieve severe pain. However,
and secondary abnormalities in Coats’ disease. stage 5, advanced end-stage disease, requires no treatment
The primary mechanism underlying Coats’ disease is because the eye is usually blind but comfortable.9
abnormal permeability of the vascular endothelial cells from Reports have described resolution of exudation using xenon
hyalinization and endothelial cell separation. This vascular arc,72 argon,73 yellow-dye,74 and large spot diode75 lasers; how-
incompetence results in subsequent leakage of blood com- ever, resolution often takes months and the final visual acuity
ponents into the retinal tissue and subretinal space.68 Light is often poor. Areas of thick exudation may prevent adequate
microscopy reveals variability among the retinal vessels absorption of laser energy; in such areas, the triple freeze-thaw
including: dilation, hyaline thickening, intraluminal and peri- method of transscleral retinal cryopexy may be more effective.76
vascular gliosis, intraluminal narrowing, and aneurysms devoid Multiple treatment sessions are often necessary to accomplish
of endothelium. In addition, spindle cells, eosinophils, poly- complete vascular closure, and careful follow-up is required to
morphonuclear leukocytes, and mononuclear cells are found document regression of the abnormal vessels and to detect new
both around and within vessels.69 Electron microscopy demon- areas of telangiectasia, which have been reported to occur as late
strates intramural thickening with a basement membrane-like as 5 years after the initial treatment (Fig. 139.7).77
material, patchy absence of endothelium and pericytes, and
plasmoid and fibrinous infiltration of aneurysmal and telangi-
ectatic vessel walls.69
With breakdown of the blood–retinal barrier, massive out-
pouring of lipid-rich exudate occurs into the retina and
subretinal space. Light microscopy reveals irregular thickening
of the inner retina from cystic cavities with periodic acid-Schiff
(PAS) positive eosinophilic fluid and infiltration of foam and
‘ghost’ cells. In addition, subretinal fibrin, cholesterol crystals,
and cholesterol- and pigment-laden macrophages are observed
(Fig. 139.6).18,69,70 Electron microscopy reveals inner retina
infiltration by foam and ‘ghost’ cells, along with macrophages,
hypertrophic Muller cells, and perivascular proliferation of glial
cells. The outer retina demonstrates patchy degeneration and
atrophy of the photoreceptors. The subretinal space is generally
electron-optically empty.69 The external globe, cornea, trabecular
meshwork, iris, ciliary body, Bruch’s membrane, and choroid
are usually normal.10,69

TREATMENT
The primary goal of treatment is obliteration of the abnormal
leaking retinal vessels to allow subsequent absorption of the
lipid exudates. In mild cases, ablation of these vessels may be FIGURE 139.7. Nearly complete resolution of exudation observed in
1852 accomplished with laser photocoagulation or transscleral Figure 139.3 after laser photocoagulation.
Coats’ Disease and Retinal Telangiectasia

FIGURE 139.8. (a) B-scan ultrasound depicts


total exudative retinal detachment in a patient
with Coats’ disease. (b) This patient’s retina
was successfully reattached via pars plana
vitrectomy with internal drainage of subretinal
fluid and endolaser photocoagulation.

a b

In advanced cases associated with exudative retinal detach-


TABLE 139.2 Differential Diagnosis of Idiopathic Telangiectasia
ment, surgical drainage of subretinal fluid and exudate may be
necessary to allow treatment of abnormal retinal vessels with Diabetic retinopathy
either photocoagulation or cryopexy.72,76,77 Although scleral

CHAPTER 139
Branch retinal vein occlusion/central retinal vein occlusion
buckling with external drainage remains a viable treatment
option (Fig. 139.8),6,16 accumulations of cholesterol deposits Radiation retinopathy
can hinder subretinal fluid outflow at the draining sclerotomy Tapetoretinal dystrophy
site, insulate retinal telangiectasia from the cryoablation, and
Inflammatory retinopathy/Irvine–Gass syndrome
prevent anatomical retinal reattachment.78 Pars plana vitrec-
tomy with78,79 or without80 scleral buckling achieves anatomic Coats’ disease
success and allows controlled subretinal fluid and cholesterol Eales’ disease
drainage through an internal retinotomy, direct ablation of
telangiectasia with diathermy or laser, and the ability to peel Adult-onset pseudovitelliform foveal macular dystrophy
associated preretinal membranes. Although visual function may Best’s disease
be severely limited, particularly in cases of total retinal detach-
Age-related macular degeneration
ment, these surgical maneuvers may stabilize the eye and
prevent the subsequent development of phthisis, painful neo- Choroiditis
vascular glaucoma, and the need for enucleation.16 Sickle-cell retinopathy
With the recent advent of anti-vascular endothelial growth
factor (anti-VEGF) therapy, Smithen and associates12 speculate Polycythemia vera retinopathy
that anti-VEGF medications may limit vascular leakage found Canthaxanthine crystalline retinopathy
in Coats’ disease since VEGF injections into nonhuman
Other crystalline retinopathies
primates caused vascular changes similar to those found in
Coats’ disease.81,82 However, no published reports have evalu- Localized retinal capillary hemangioma
ated intravitreal VEGF levels or the use of anti-VEGF therapy in Hypertensive retinopathy
Coats’ patients.
Ocular ischemic syndrome/carotid artery obstruction
IDIOPATHIC MACULAR TELANGIECTASIA
Retinal telangiectasia localized to the foveal region alone in one the superficial and deep retinal capillary circulations.84 Vascular
or both eyes is termed macular (juxtafoveal) telangiectasia. abnormalities are noted within 2 disk diameters of the center of
Such changes may be congenital, and thus a mild variant of the fovea, primarily along the temporal parafoveal region, and
Coats’ disease, or may be acquired and seen as an isolated often associated with localized capillary leakage and lipid
finding in middle-aged or older patients.40 These idiopathic exudate (Fig. 139.9a,b). Visual loss in this group of patients tends
forms of telangiectasia must be distinguished from secondary to occur with the accumulation of foveolar exudate and associated
telangiectasia caused by other retinal vascular diseases cystoid macular edema.83,84 Despite maintenance of excellent
(Table 139.2). visual acuity for years without treatment and spontaneous reso-
Gass and Blodi classified these patients into three groups lution in occasional patients, Gass and Blodi recommended early
with multiple subgroups based on historical, biomicroscopic, photocoagulation when visual loss is associated with central
and fluorescein angiographic findings.83 Recently, Yannuzzi and accumulation of yellow exudate.83
associates simplified the classification scheme into two types of
idiopathic macular telangiectasia by eliminating rare forms of
the disease.84 TYPE II (PERIFOVEAL TELANGIECTASIA)
Also known as group II, occult and nonexudative idiopathic
juxtafoveal retinal telangiectasia under the Gass and Blodi
TYPE 1 (ANEURYSMAL TELANGIECTASIS) classification, this is the most common form of macular telangi-
Also known as group 1, visible and exudative idiopathic juxtafoveal ectasia and occurs equally in both sexes. Patients typically
retinal telangiectasia under the Gass and Blodi classification, present in the fifth and sixth decades of life with mild blurring
middle-aged male patients typically present with a unilateral of vision in one or both eyes that can progress, primarily from
mild blurring of central vision.83 This form of macular telang- foveolar atrophy or subretinal neovascularization.83 Biomicro-
iectasia is considered a form of Coats’ disease that is limited to scopy reveals symmetric blunting of the foveal reflex with a
the macula.83,84 Biomicroscopy reveals arteriolar, capillary, and mild grayish appearance of the parafoveal retina, minimal serous
venular aneurysms and telangiectasia which are found in both exudation, and no lipid deposition.83,84 In addition, glistening 1853
RETINA AND VITREOUS

FIGURE 139.9. (a) Unilateral type 1


aneurysmal telangiectasis. Note the aneurysms
and telangiectatic capillaries in the parafoveolar
region associated with lipid exudates.
(b) Fluorescein angiogram demonstrating
capillary telangiectasia, microaneurysms, and
leakage.

a b
SECTION 10

a c e

b d f

FIGURE 139.10. (a and b) Right and left color fundus photographs of type II idiopathic macular telangiectasia demonstrating a mild parafoveal
grayish appearance. (c and d) Classic fluoroscein staining temporal to fovea on late-phase angiogram of right and left eyes. (e and f) Horizontal
5mm optical coherence tomography scans through fovea of right and left eyes demonstrating a nearly normal appearance without fluid in areas
of staining on angiogram. There are tiny hyporeflective spaces in the foveola of each eye.

white or yellowish-white crystalline deposits may be noted in


the superficial parafoveal retina in ~40% of patients.85 In some
patients, a small, yellow lesion, 1/3 disk diameter in size,
develops within the foveal avascular zone and appears to repre-
sent an inner retinal cavitation on OCT evaluation.83,84
Initially, telangiectasis is not visible on biomicroscopy, and
fluorescein angiography shows only mild staining within
one disk diameter of the foveola, especially temporally
(Fig. 139.10a–f). With progression, these eyes may develop
dilated right-angled retinal venules and arterioles and stellate
plaques of retinal pigment epithelial hyperplasia, as well as
intra- and subretinal anastomosis and subretinal neovascular-
ization.26,40,84 Optical coherence tomography can demonstrate
hyporeflective intraretinal cavitation without foveolar thickening
(Fig. 139.11) and middle or inner layer hyperreflectivity
corresponding to areas of retinal pigment epithelial migration.86 FIGURE 139.11. Hyporeflective inner layer cavitation without retinal
When subretinal neovascularization occurs, central visual thickening on optical coherence tomography scan.
1854
Coats’ Disease and Retinal Telangiectasia

a b c

FIGURE 139.12. (a) Color photograph of subretinal neovascularization secondary to type II idiopathic macular telangiectasia. (b) Early phase
fluorescein angiogram demonstrating subretinal neovascularization with probable retinal feeding and draining vessels. (c) Horizontal 5 mm optical
coherence tomography image demonstrating a hyperreflective subretinal focus corresponding to the area of neovascularization on clinical exam.

CHAPTER 139
acuity loss may be rapid, and 81% of untreated eyes have a final have demonstrated efficacy; however, the subretinal neovascu-
acuity of 20/200 or worse (Fig. 139.12a–c).87 larization in idiopathic juxtafoveal retinal telangiectasis arises
Grid laser is not indicated for treatment of type II perifoveal from the retina, as opposed to the choroid, and activation of
telangiectasia because it does not improve or stabilize long-term photosensitizing dye in the retinal layers may cause iatrogenic
visual acuity.88 In addition, laser has led to the development of damage.90,93 Laser photocoagulation may help stabilize central
subretinal hemorrhage89 and choroidal neovascularization.88 A visual acuity but induces an iatrogenic scotoma,94 and sub-
variety of therapeutic modalities have been employed to treat macular surgery has demonstrated poor outcomes.95 Anti-VEGF
subretinal neovascularization complicating this disorder. Photo- drugs hold promise for treating neovascularization associated
dynamic therapy90,91 and transpupillary thermotheraphy92,93 with this disease.

REFERENCES
1. Leber T: Verber ein durch Yorkommen 12. Smithen L, Brown G, Brucker A, et al: 22. Machemer R, Williams J: Pathogenesis and
miltipler Miliaraneurisi men Characterisierte Coats’ disease diagnosed in adulthood. therapy of traction detachment in various
Form von Retinal-degeneration. Graefes Ophthalmology 2005; 112:1072–1078. retinal vascular diseases. Am J Ophthalmol
Arch Clin Exp Ophthalmol 1912; 81:1–14. 13. Black G, Perveen R, Bonshek R, et al: 1988; 105:170–181.
2. Coats G: Forms of retinal disease with Coats’ disease of the retina (unilateral 23. Ioannidis A, Liasis A, Sheldrick J, et al:
massive exudation. R Lond Ophthalmic retinal telangiectasis) caused by somatic Lamellar macular hole as a presenting
Hosp Rep 1908; 17:440–525. mutation in the NDP gene: a role for norrin feature in a child with Coats’ disease.
3. Coats G: Ueber Retinitis exudativa (retinitis in retinal angiogenesis. Hum Mol Gen 1999; J Pediatr Ophthalmol Strabismus 2005;
hemorrhagiga externa). Graefes Arch Clin 8:2031–2035. 42:378–379.
Exp Ophthalmol 1912; 81:275–327. 14. Mintz-Hittner H, Ferrell R, Sims K, et al: 24. Shienbaum G, Tasman W: Coats disease.
4. Reese A: Telangiectasis of the retina and Peripheral retinopathy in offspring of A lifetime disease. Retina 2006; 26:422–424.
Coats’ disease. Am J Ophthalmol 1956; carriers of Norrie disease gene mutations. 25. Theodossiadis G: Some clinical,
42:1–8. Ophthalmology 1996; 103:2128–2134. fluorescein-angiographic, and therapeutic
5. Leber T: Retinitis exudativa (Coats), 15. Luhmann U, Lin J, Acar N, et al: Role of aspects of Coats’ disease. J Pediatr
Retinitis und Chorioretinitis Serofibrinosa Norrie Disease Pseudoglioma gene in Ophthalmol Strabismus 1979; 16:257–262.
Degeraus. Graefe Saemisch Augenheikld sprouting angiogenesis during 26. Gass J: Stereoscopic atlas of macular
1915; 7:1267–1319. development of the retinal vasculature. diseases: diagnosis and treatment. 3rd edn.
6. Gomez-Morales A: Coats’ disease. Am J Invest Ophthalmol Vis Sci 2005; St. Louis, MO: CV Mosby; 1987.
Ophthalmol 1965; 60:855–864. 46:3372–3382. 27. Chen S, Patel C: Optical coherence
7. Woods A, Duke J: Coats’ disease. I. 16. Silodor S, Augsburger J, Shields J: Natural tomography in uncooperative children
Review of the literature, diagnostic criteria, history and management of advanced under general anesthesia [letter]. J Pediatr
clinical findings, and plasma lipid studies. Coats’ disease. Ophthalmic Surg 1988; Ophthalmol Strabismus 2005; 42:71.
Br J Ophthalmol 1963; 47:385–412. 19:89–93. 28. Shields C, Mashayekhi A, Luo C, et al:
8. Cahill M, O’Keefe M, Acheson R, et al: 17. Green W, McDonnell P, Yeo J: Pathologic Optical coherence tomography in children:
Classification of the spectrum of Coats’ features of senile macular degeneration. analysis of 44 eyes with intraocular tumors
disease as subtypes of idiopathic retinal Surv Ophthalmol 1985; 92:615–627. and simulating conditions. J Pediatr
telangiectasis with exudation. Acta 18. Chang M, McLean I, Merritt J: Coats’ Ophthalmol Strabismus 2004; 41:338–344.
Ophthalmol Scand 2001; 79:596–602. disease: a study of 62 histologically 29. Puliafito C, Hee M, Lin C, et al: Imaging of
9. Shields J, Shields C, Honavar S, et al: confirmed cases. J Pediatr Ophthalmol macular diseases with optical coherence
Classification and management of Coats Strabismus 1984; 21:163–168. tomography. Ophthalmology 1995;
disease: the 2000 Proctor Lecture. Am J 19. Shields J, Shields C: Review: Coats 102:217–229.
Ophthalmol 2001; 131:572–583. disease. Retina 2002; 22:80–91. 30. Haik B, Saint Louis L, Smith M, et al:
10. Shields J, Shields C, Honavar S, Demirci H: 20. Shields J, Eagle R, Fammartino J, et al: Magnetic resonance imaging in the
Clinical variations and complications of Coats disease as a cause of anterior evaluation of leukocoria. Ophthalmology
Coats disease in 150 cases. The 2000 chamber cholesterolosis. Arch Ophthalmol 1985; 92:1143–1152.
Sanford Gifford Memorial Lecture. Am J 1995; 113:975–977. 31. Atta H, Watson N: Echographic diagnosis
Ophthalmol 2001; 31:561–571. 21. Eibschitz-Tsimhoni M, Johnson M, Johnson of advanced Coats’ disease. Eye 1992;
11. Maruoka K, Yamamoto M, Fujita H, et al: A T, Moroi S: Coats’ syndrome as a cause of 6:80–85.
case of Coats’ disease in a low-birth- secondary open-angle glaucoma. 32. Haik B, Saint Louis L, Smith M, et al:
weight infant. Ophthalmologica 2005; Ophthalmic Surg Lasers Imaging 2003; Computed tomography of the
219:401–403. 34:312–314. nonrhegmatogenous retinal detachment in
1855
RETINA AND VITREOUS

the pediatric patient. Ophthalmology 1985; 53. Newell S, Hall B, Anderson C, Lim E: 73. Haik B: Advanced Coats’ disease. Trans
92:1133–1142. Hallermann Streiff syndrome with Coats Am Ophthalmol Soc 1991; 89:371–476.
33. Kirath H, Eldem B: Management of disease. J Pediatr Ophthalmol Strabismus 74. Nucci P, Bandello F, Serafino M, Wilson M:
moderate to advanced Coats’ disease. 1994; 31:123–125. Selective photocoagulation in Coats’
Ophthalmologica 1998; 212:19–22. 54. Crow Y, McMenamin J, Haenggeli C, et al: disease: ten-year follow-up. Eur J
34. Potter P, Shields C, Shields J, Flanders A: Coats’ Plus: a progressive familial syndrome Ophthalmol 2002; 12:501–505.
The role of magnetic resonance imaging in of bilateral Coats’ disease, characteristic 75. Couvillion S, Margolis R, Mavrofrides E,
children with intraocular tumors and cerebral calcification, leukoencephalopathy, et al: Laser treatment of Coats’ disease.
simulating lesions. Ophthalmology 1996; slow pre- and post-natal linear growth and J Pediatr Ophthalmol Strabismus 2005;
103:1774–1783. defects of bone marrow and integument. 42:367–368.
35. Haik B, Koizumi J, Smith M, Ellsworth R: Neuropediatrics 2004; 35:10–19. 76. Sigelman J: Retinal diseases:
Fresh preparation of subretinal fluid 55. Folk J, Genovese F, Biglan A: Coats pathogenesis, laser therapy, and surgery.
aspirations in Coats’ disease. Am J disease in a patient with Cornelia de Lange Boston, MA: Little Brown & Co; 1984.
Ophthalmol 1985; 100:327–328. syndrome. Am J Ophthalmol 1981; 77. Chisolm I, Foulds W, Christison D:
36. Shields J, Shields C, Ehya H, et al: Fine 91:607–610. Investigation and therapy of Coats’ disease.
needle aspiration biopsy of suspected 56. Kondra L, Cangemi F, Pitta C: Alport’s Trans Ophthalmol Soc UK 1974; 94:335.
intraocular tumors. The 1992 Urwick syndrome and retinal telangiectasia. Ann 78. Yoshizumi M, Kreiger A, Lewis H, et al:
lecture. Ophthalmology 1993; Ophthalmol 1983; 15:550–551. Vitrectomy techniques in late-stage
100:1677–1684. 57. Berger M, Lieberman K, Schoeneman M, Coats’-like exudative retinal detachment.
37. Luckie A, Hamilton A: Adult Coats’ disease et al: Coats disease in a renal transplant Doc Ophthalmol 1995; 90:387–394.
in branch retinal vein occlusion. Aust N Z J recipient. Nephrol Dial Transplant 1987; 79. Schmidt-Erfurth U, Lucke K: Vitreoretinal
SECTION 10

Ophthalmol 1994; 22:203–206. 2:120–123. surgery in advanced Coat’s disease. Ger J


38. Ashton N: Studies of the retinal capillaries 58. Kajtar P, Mehes K: Bilateral Coats Ophthalmol 1995; 4:32–36.
in relation to diabetic and other retinopathy associated with aplastic anemia 80. Kranias G, Krebs T: Advanced Coats’
retinopathies. Am J Ophthalmol 1980; and mild dyskeratotic signs. Am J Med disease successfully managed with vitreo-
90:203. Genet 1994; 49:374–377. retinal surgery. Eye 2002; 16:500–501.
39. Hayreh S: Post-radiation retinopathy: 59. Bhushan M, Kumar S, Griffiths C: Multiple 81. Tolentino M, Miller J, Gragoudas E, et al:
a fluorescence fundus angiographic study. glomus tumours, Coats disease and basic Intravitreous injections of vascular
Br J Ophthalmol 1970; 54:705. fibroblast growth factor. Br J Dermatol endothelial growth factor produce retinal
40. Gass J, Oyakawa R: Idiopathic 1997; 137:454–456. ischemia and microangiopathy in an adult
juxtafoveolar retinal telangiectasis. Arch 60. Gartner J, Draf W: Leber’s miliary primate. Ophthalmology 1996;
Ophthalmol 1982; 100:769. aneurysms associated with telangiectasia 103:1820–1828.
41. Recine W, Murphy R, Anderson K, et al: of the nasal mucosa. Am J Ophthalmol 82. Tolentino M, McLeod D, Taomoto M, et al:
The evaluation of patients with Eales’ 1975; 79:56–58. Pathologic features of vascular endothelial
disease. Retina 1983; 3:243. 61. Green W: Bilateral Coats disease. Massive growth factor-induced retinopathy in the
42. Ffytche T: Retinal vasculitis: a review of the gliosis of the retina. Arch Ophthalmol 1967; nonhuman primate. Am J Ophthalmol 2002;
clinical signs. Trans Ophthalmol Soc UK 77:378–383. 133:373–385.
1977; 97:457. 62. Pruett R: Retinitis pigmentosa: clinical 83. Gass J, Blodi B: Idiopathic juxtafoveolar
43. Senft S, Hidayat A, Cavender J: Atypical observations and correlations. Trans Am retinal telangiectasis. Update of
presentation of Coats disease. Retina Ophthalmol Soc 1983; 81:693–735. classification and follow-up study.
1994; 14:36–38. 63. Khan J, Ide C, Strickland M: Coats’-type Ophthalmology 1993; 100:1536–1546.
44. Miller D, Benz M, Murray T, Dubovy S: retinitis pigmentosa. Surv Ophthalmol 84. Yannuzzi L, Bardal A, Freund K, et al:
Intraretinal calcification and osseous 1988; 32:317–332. Idiopathic macular telangiectasia. Arch
metaplasia in Coats disease. Arch 64. Kremer I, Cohen S, Izhak R, Ben-Sira I: Ophthalmol 2006; 124:450–460.
Ophthalmol 2004; 122:1710–1712. An unusual case of congenital unilateral 85. Moisseiev J, Lewis H, Bartov E, et al:
45. Skuta G, France T, Stevens T, et al: Coats disease associated with morning Superficial retinal refractile deposits in
Apparent Coats’ disease and pericentric glory optic disc anomaly. Br J Ophthalmol juxtafoveal telangiectasis. Am J Ophthalmol
inversion of chromosome 3. Am J 1985; 69:32–37. 1990; 109:604–605.
Ophthalmol 1987; 104:84–86. 65. Lim W-K, Nussenblatt R, Chan C-C: 86. Gupta V, Gupta A, Dogra M, Agarwal A:
46. Genkova P, Toncheva D, Tzoneva M: Immunopathologic features of inflammatory Optical coherence tomography in group 2A
Deletion of 13q 12.1 in a child with Coats’ Coats disease. Arch Ophthalmol 2005; idiopathic juxtafoveolar telangiectasis.
disease. Acta Paediatr Hung 1986; 123:279–281. Ophthalmic Surg Lasers Imaging 2005;
27:141–143. 66. Frezzotti R, Berengo A, Guerra R, 36:482–486.
47. Gurwin E, Fitzsimons R, Sehmi K, Bird A: Cavallini F: Toxoplasmic Coats retinitis. 87. Engelbrecht N, Aaberg TJ, Sung J, Lewis
Retinal telangiectasis in Am J Ophthalmol 1965; 59:1099–1102. M: Neovascular membranes associated
fascioscapulohumeral muscular dystrophy 67. Egerer I, Rodrigues M, Tasman W: Retinal with idiopathic juxtafoveolar telangiectasis.
with deafness. Arch Ophthalmol 1985; dysplasia in Coats disease. Can J Arch Ophthalmol 2002; 120:320–324.
103:1695–1700. Ophthalmol 1975; 10:79–85. 88. Park D, Schatz H, McDonald H, Johnson R:
48. Cameron J, Yanoff M, Frayer W: Coats 68. McGettrick P, Loeffler K: Bilateral Coats’ Grid laser photocoagulation for macular
disease and Turner’s syndrome. Am J disease in an infant (a clinical, edema in bilateral juxtafoveal
Ophthalmol 1974; 78:852–854. angiographic, light and electron telangiectasis. Ophthalmology 1997;
49. Schuman J, Lieberman K, Friedman A, microscopic study). Eye 1987; 1:136–145. 104:1838–1846.
et al: Senior-Loken syndrome (familial 69. Tripathi R, Ashton N: Electron 89. Friedman S, Mames R, Stewart M:
renal-retinal dystrophy) and Coats’ disease. microscopical study of Coats’s disease. Subfoveal hemorrhage after grid laser
Am J Ophthalmol 1985; 100:822–827. Br J Ophthalmol 1971; 55:289–301. photocoagulation for idiopathic
50. Burch J, Leveille A, Morse P: Ichthyosis 70. Kremer I, Nissenhorn I, Ben-Sira I: juxtafoveolar retinal telangiectasis.
hystrix epidermal nevus syndrome and Cytologic and biochemical examination of Ophthalmic Surg 1993; 24:551–553.
Coats’ disease. Am J Ophthalmol 1980; the subretinal fluid in the diagnosis of 90. Potter M, Szabo S, Chan E, Morris A:
89:25–30. Coats’ disease. Acta Ophthalmol (Copenh) Photodynamic therapy for subretinal
51. Hon C, Ko T-C: Coats disease and VATER 1989; 67:342–346. neovascular membranes in Type 2A
association in a 5-year-old boy. Arch 71. Sneed S, Blodi C, Pulido J: Treatment of idiopathic juxtafoveolar retinal
Ophthalmol 2004; 122:1232–1233. Coats’ disease with the binocular indirect telangiectasis. Am J Ophthalmol 2002;
52. Patrassi G, Sartori M, Piermarocchi S, et al: argon laser photocoagulator [letter]. Arch 133:149–151.
Unusual thrombotic-like retinopathy (Coats’ Ophthalmol 1989; 107:789–790. 91. Hershberger V, Hutchins R, Laber P:
disease) associated with congenital 72. Egerer I, Tasman W, Tomer T: Coats’ Photodynamic therapy with verteporfin for
plasminogen deficiency type I. J Int Med disease. Arch Ophthalmol 1974; subretinal neovascularization secondary to
1993; 234:619–623. 97:109–112. bilateral idiopathic acquired juxtafoveolar
1856
Coats’ Disease and Retinal Telangiectasia

retinal telangiectasis. Ophthalmic Surg 93. Nachiappan K, Shanmugam M: Treatment Laser photocoagulation of the macula.
Lasers Imaging 2003; 34:318–20. of CNVM secondary to idiopathic Philadelphia, PA: Lippincott; 1989.
92. Shukla D, Singh J, Kolluru C, et al: juxtafoveal retinal telangiectasis by 95. Berger A, McCuen B, Brown G, Brownlow
Transpupillary thermotherapy for subfoveal transpupillary thermotherapy [letter]. R: Surgical removal of subfoveal
neovascularization secondary to group 2A Am J Ophthalmol 2005; 139:577–578. neovascularization in idiopathic juxtafoveal
idiopathic juxtafoveolar telangiectasis. 94. Brucker A, Robinson F: Primary retinal retinal telangiectasis. Retina 1997;
Am J Ophthalmol 2004; 138:147–149. vascular abnormalities. In: Yannuzzi L, ed. 17:94–98.

CHAPTER 139

1857
CHAPTER

140 Neuroretinitis
Valerie Purvin

Key Features
• Acute, painless, unilateral visual loss of variable severity
• Usually affects healthy young individuals
• Acutely: disk edema often with peripapillary serous
detachment
• Ten days later: macular hard exudates in star pattern
• Due to cat scratch disease in two-thirds of cases
• One-quarter of cases are idiopathic despite thorough
evaluation
• Generally good prognosis regardless of treatment
• Consider antibiotics for suspected bacterial infection
• Steroids may be considered as well

HISTORIC PERSPECTIVE
In 1916 Theodor Leber described a condition characterized by
acute unilateral visual loss with disk edema and macular exudates
arranged in a star pattern.1 Believing the condition to be a pri-
mary retinal disorder, he used the term ‘stellate maculopathy’.
This concept was challenged by Gass in 1977, who noted that disk FIGURE 140.1. Ophthalmoscopic appearance of idiopathic NR in a
36-year-old woman. Fundus photograph of the patient’s left eye taken
edema precedes the formation of exudates in this condition.2
1 month after onset of visual loss shows mild residual disk edema
He further demonstrated by fluorescein angiography that the with pallor and a robust macular star. Serologic testing for specfic
site of the leakage is in fact not the macula but the optic disk inflammatory diseases was unrevealing.
and suggested the term neuroretinitis (NR). In some patients a
specific infectious agent has been implicated; cases in which no
infectious etiology is identified are designated as Leber’s idio-
pathic stellate NR. The series by Dreyer et al3 and by Maitland TABLE 140.1 Etiologies
and Miller4 in 1984 furnished a description of the typical
Bartonella species (mostly B. henselae)
clinical features of this disorder. Since that time a number of
papers have reported additional cases of NR due to a wide range Spirochete: syphilis (2º or 3º), Lyme disease (stage II), leptospirosis
of infections (this is discussed in the following). Viral or postviral: mumps, chicken pox, herpes simples, herpes
zoster, HIV, hepatitis B, Coxsackie B, influenza A, Epstein–Barr,
PATHOPHYSIOLOGY cytomegalovirus
Tuberculosis
Inflammation of disk capillaries causes leakage of protein and
lipid that spreads into the peripapillary subretinal space and Toxoplasmosis
outer plexiform layer. Resorption of the serous component Nematode: Toxocara canis, DUSN
leaves lipid exudates that are subsequently ingested by
Histoplasmosis capsulatum
macrophages. Due to the loose, radial configuration of the outer
plexiform layer, a star pattern is formed5 (Fig. 140.1). This loca- Rocky Mountain spotted fever
tion of fluid accumulation has more recently been confirmed Salmonella
with ocular coherence tomography (OCT).6 It is not entirely
clear whether the disk inflammation in this disorder represents Postvaccination: Rabies
direct infection or a secondary autoimmune response. Noninfectious uveitis: sarcoidosis, inflammatory bowel disease,
periarteritis nodosa
ETIOLOGIES Uncertain mechanism: Parry–Romberg syndrome (progressive
facial hemiatrophy), IRVAN syndrome, tubulointerstitial nephritis
Cases of NR have been reported in association with a variety of and uveitis (TINU syndrome)
infectious agents (see Table 140.1).7–23 The single most 1859
RETINA AND VITREOUS

TABLE 140.2 Differential Diagnosis for Disk Edema with


Macular Star

Hypertensive retinopathy
Papilledema (Øintracranial pressure)
Anterior ischemic optic neuropathy
Diabetic papillopathy
Branch retinal vein occlusion/papillophlebitis
Optic disk tumor: melanocytoma, juxtapapillary angioma

common of these is cat scratch disease (CSD) which accounted


for two-thirds of cases in one large series.24 Despite thorough
evaluation, approximately one quarter of cases remain idio-
pathic.24 Occasional cases are due to noninfectious forms of
uveitis such as sarcoidosis, inflammatory bowel disease, and
SECTION 10

periarteritis nodosa. NR has also been described as a part of


other retinal inflammatory disorders including idiopathic
retinal vasculitis and aneurysms (IRVAN syndrome) and diffuse FIGURE 140.2. Ophthalmoscopic appearance of NR due to CSD. The
unilateral subacute neuroretinitis (DUSN) which is caused by a patient was a 22-year-old man who developed decreased vision in the
nematode.25 Other noninflammatory conditions may also cause left eye 2 weeks after onset of fever, headache, and muscle aches.
disk edema with a macular star figure26 and it is important for Acutely the disk is swollen with nerve fiber layer hemorrhage and
the clinician to be alert to these (see Table 140.2). Notably, peripapillary serous detachment.
increased intracranial pressure (papilledema), hypertensive reti-
nopathy, ischemic optic neuropathy and diabetic papillopathy
may all cause a similar funduscopic appearance. The term NR
should be reserved for those cases in which the underlying
mechanism is inflammatory.

CLINICAL CHARACTERISTICS
NR usually affects children and young adults (average age
22–30 years) but the age range is broad.3,4 Males and females
are affected equally. Approximately 50% of affected individuals
experience an antecedent flu-like illness, usually affecting the
upper respiratory tract.3,4 In patients with cat scratch NR these
prodromal symptoms usually include sore throat, headache,
myalgias, and lymphadenopathy. Visual loss typically takes the
form of painless unilateral blurring of central vision which
progresses over several days. Visual loss is usually painless
although occasional patients experience a mild retrobulbar
ache. While mild disk edema is occasionally seen in the fellow
eye, symptomatic bilateral NR is quite unusual and should
always suggest the alternative possibility of hypertensive
FIGURE 140.3. Same patient as in Figure 140.1 but 4 weeks later.
retinopathy or raised intracranial pressure. There has been much resolution of disk edema with the new
appearance of hard exudates tracking to the macula in a star pattern.
VISUAL FUNCTION Serologic testing confirmed recent B. henselae infection.

Visual acuity is usually between 20/50 and 20/200 with a range appearance depends in large part on when the patient is
from 20/20 to light perception. The most common pattern of examined. Isolated optic disk edema is the earliest finding
visual field loss, found in 24 of 29 eyes in the series of Dreyer (Fig. 140.2). Disk swelling is usually diffuse but can be segmental,
et al, is a cecocentral or central scotoma, consistent with the a finding that may be better appreciated with fluorescein
presence of edema in the papillomacular bundle.3 Other patterns angiography. The degree of disk edema is variable; when severe
including arcuate defects and altitudinal loss are occasionally it may be accompanied by peripapillary nerve fiber layer
seen. A relative afferent pupillary defect (RAPD) is often present hemorrhages. In most cases, disk edema is associated with
although not with the same constancy or magnitude as in an exudative peripapillary serous retinal detachment. It takes
demyelinating optic neuritis. This difference reflects a different 9–12 days for the characteristic macular hard exudates to
mechanism of visual loss in these two conditions: visual loss appear and at this stage the disk edema is usually diminishing
is due in large part to maculopathy in NR versus optic nerve (Fig. 140.3). The star is initially sharply defined and spoke-like;
dysfunction in optic neuritis (ON). over time the exudates develop a more globby appearance and
eventually, after a number of months, disappear completely,
often leaving residual subfoveal RPE defects in the macula. Disk
FUNDUSCOPIC FINDINGS edema resolves over 8–12 weeks in most cases.3 The disk may
Posterior vitreous cells are usually present; anterior chamber eventually return to normal or may show pallor and/or gliotic
1860 cell and flare are occasionally seen as well. The funduscopic changes.
Neuroretinitis

In addition to these characteristic changes involving the disk 35 patients with NR seen in a single university-based practice
and macula, small, discrete yellow-white chorioretinal spots are identified three patients who had already been diagnosed with
sometimes identified.27 These spots are at the level of the deep MS.30 All three of these patients were on b-interferon and the
retina or choriocapillaris and may be seen in both symptomatic authors suggested that perhaps this treatment influenced the
and asymptomatic eyes. These CR lesions are especially permeability of disk vessels thus affecting the funduscopic
characteristic of patients with NR due to CSD but have also appearance. Despite this one report, the management of patients
been found in patients with other causes of NR and in those with NR should be clearly distinguished from those with ON.
with the idiopathic variety. Their occurrence serves as evidence
of a hematogenously spread infectious agent rather than an DIFFERENTIAL DIAGNOSIS
autoimmune process. These spots resolve slowly, becoming
small chorioretinal scars. The approach to diagnosis depends in part on when the patient
is seen in the course of the illness. When seen acutely, i.e.,
before formation of a macular star, the main alternative diag-
ANCILLARY TESTING nosis is ON. Both NR and ON typically present as acute
Fluorescein angiography in patients with acute NR demonstrates monocular visual loss in a previously healthy young person and
abnormal capillary permeability, particularly from the capillaries optic disk edema is seen in both conditions. Several features are
deep within the optic disk. Such leakage may persist even after the helpful in making this distinction. The presence of eye pain in
disk has returned to its normal appearance. While there may be most patients with ON is extremely helpful since this is a very

CHAPTER 140
slight staining of peripapillary vessels, the macular vasculature is uncommon symptom in NR. The absence of a RAPD in the
entirely normal. Disk staining is often segmental and is found in presence of monocular visual loss speaks strongly against ON
the contralateral (asymptomatic) eye in 10–15% of cases. Chorio- as the cause. Similarly, a disproportion between the visual loss
retinal white spots, when present, show late hyperfluorescence. and the RAPD (large drop in visual acuity with only a small
OCT has shown accumulation of fluid in the outer plexiform RAPD) speaks in favor of NR. The presence of peripapillary
layer with or without serous detachment of the sensory retina.6 serous detachment is characteristic of NR but unexpected in
ON. Disk hemorrhages are sometimes present in NR but
PROGNOSIS extremely unusual in ON. Finally, the results of orbital MRI
scanning in ON are abnormal in 90% of cases, whereas in NR
Most patients with NR enjoy excellent recovery of vision such scans are typically normal.
without intervention. In the series of Dreyer et al, for example, When seen after appearance of the macular star the differ-
visual acuity at last follow-up examination (median 14 months) ential diagnosis is different. The first step is to decide if the
was 20/20 or better in 66% of cases, 20/25 to 20/40 in 31% and patient actually has NR or has, instead, another cause of macular
remained at 20/300 in one eye.3 A repeat event either in the hard exudates. The presence of more extensive retinopathy
previously affected or the fellow eye is unusual. should suggest an alternative diagnosis such as CRVO or BRVO
There appears to be a subset of patients, however, with a (retinal hemorrhages predominate) or severe hypertension
somewhat different clinical picture in whom the prognosis is (hemorrhages and exudates usually present). When these
more guarded. Two of the patients in the series of Dreyer et al changes are bilateral, particular attention should be directed to
had disk-related field defects and a large relative afferent defect the possibility of hypertensive retinopathy. In increased intra-
and in both of these cases the visual outcome was poor.3 Of the cranial pressure (papilledema) the findings are also typically
12 patients with NR described by Maitland and Miller, only bilateral but the changes are usually confined to the disk and
three failed to experience excellent recovery and in two of these macula. In the case of papilledema and in anterior ischemic
cases there was evidence of a previous similar event in the fellow optic neuropathy the macular star is usually partial, seen just
eye.4 Purvin and Chioran reported a series of seven such patients on the nasal side of the macula in contrast with its appearance
who experienced 2–7 attacks of NR at intervals of 1–10 years 360° around the macula in most cases of NR. Finally, prolonged
(average interval 2.7 years).28 Following repeated episodes, duration of disk edema and macular exudates should suggest a
visual loss is cumulative and can be quite substantial. This noninflammatory disorder, specifically CRVO, papillophlebitis,
variant of NR may be suspected following an initial episode by or an optic disk tumor.
virtue of a large relative afferent pupillary defect, disk-related
field loss and poor visual recovery. Such patients should be EVALUATION
considered to be at relatively high risk for a future similar event
and should be counseled accordingly. In some cases, prophy-
lactic treatment to prevent a repeat attack may be considered
HISTORY AND PHYSICAL
(this is discussed in the following). A thorough history is important and should include the following
features: travel (particularly to areas in which Lyme disease is
RELATIONSHIP TO MULTIPLE SCLEROSIS prevalent), animal exposure, sexual contact, ingestion of unpas-
teurized or uncooked foods. The patient should be questioned
One particular aspect of prognosis bears specific comment, about systemic symptoms including fever, lymphadenopathy,
namely the possible relationship of NR to multiple sclerosis myalgias, skin rash or other skin lesions, sore throat, and
(MS). It has been well established that patients who experience headache. Complete physical and ocular examinations are also
an attack of ON are at increased risk for the future development essential.
of MS. Insofar as NR can be considered a form of papillitis, one
might assume that patients with this condition would be at
similarly high risk. A large survey of affected patients, however, LABORATORY TESTING
found no such increased risk.29 This difference in prognosis is The list of diseases that have on occasion caused NR is exten-
presumably related to differences in pathophysiology in these two sive. Laboratory testing should be tailored to the individual
conditions. In demyelinating ON the target tissue of the inflam- patient based on information obtained from the history and
matory response is the myelin sheath, whereas in NR the target physical examinations. In most cases, serologic testing should
is the optic disk vasculature. However, a retrospective review of include cat scratch titers (Bartonella species), a fluorescent 1861
RETINA AND VITREOUS

treponemal antibody absorption (FTA-ABS) test, and a PPD. In although the possible benefit of such treatment is unknown. In
endemic areas and in patients with a history of tick exposure cases with particularly severe visual loss (either idiopathic or
and/or characteristic symptoms of Lyme disease, titers for these due to a specific organism) treatment with corticosteroids is
antibodies should also be obtained. Other testing should be often advised. In patients with recurrent idiopathic NR, the
obtained as indicated by specific findings from the history and addition of long-term immunosuppression has been shown to
physical examination. decrease the number of recurrences.32

TREATMENT
Summary: Cat Scratch NR
Treatment generally depends on the results of testing for specific • Most common identifiable cause of NR
infectious agents. The most common causative organism, • Usually due to B. henselae
Bartonella henselae, is sensitive to a variety of antibiotics and • Most often affects children
most authors recommend treatment accordingly. There are no • 80% of CSD infections are self-limited
controlled studies, however, comparing the results of treatment • Mild local infection is followed by tender regional
with the natural history of the disease or comparing the efficacy lymphadenopathy then fever, malaise, and headache
of different antibiotic regimen. A retrospective study found the • Other ocular manifestations include conjunctivitis, anterior
following oral medications to be of benefit: rifampin (effective uveitis, focal retinal vasculitis or choroiditis, branch retinal
in 87% of patients), ciprofloxacin (84%), and trimethaprim-
SECTION 10

artery occlusion, and peripapillary angiomatous lesions


sulfamethoxazole (58%).31 In another smaller series, patients
with NR due to CSD did well with a combination of doxycycline
100 mg and rifampin 300 mg twice daily with minimal side-
effects.9 Golnik and associates reported a similar favorable out-
Tip File
come in four patients with CSD–NR treated with ciprofloxacin
1. Other conditions besides NR can cause disk edema with
500 mg twice daily.11 Treatment is usually initiated while
macular hard exudates. Before initiating an extensive
results of serologic testing are pending and continued for any-
evaluation for specific etiologies, rule out other mechanisms
where from 10 days to 6 weeks if results confirm CSD infection.
2. Be especially cautious in cases of bilateral NR. In most cases
The use of doxycycline and ciprofloxacin should be avoided in
this clinical picture is due to hypertensive retinopathy or
children, however. Some patients are also treated with steroids
occasionally increased intracranial pressure
although there is no evidence that this addition improves the
3. NR is most common in young people. In older individuals
visual outcome. Unlike treatment considerations for demyeli-
consider the alternative possibility of anterior ischemic optic
nating ON, there is no reason to favor intravenous over oral
neuropathy
administration in this setting.
4. In cases with acute unilateral visual loss, suspect NR rather
Patients with idiopathic NR usually enjoy good visual recovery
than ON in cases with little or no RAPD
regardless of treatment. Some are treated with corticosteroids

REFERENCES
1. Leber T: Die pseudonephritischen and sequelae. Ophthalmology 1998; Syphilitic neuroretinitis. Am J Ophthalmol
Netzhauterkrankungen, die Retinitis stellata: 105:459–466. 1983; 95:480–486.
Die Purtschersche Netzhautaffektion nach 10. Fish RH, Hoskins JC, Kline LB: 19. Lesser RL, Kornmehl EW, Pachner AR,
schwerer Schadelverletzung. In: Graefe AC, Toxoplasmosis neuroretinitis. et al: Neuro-ophthalmologic manifestations
Saemische T, eds. Graefe-Saemisch Ophthalmology 1993; 100:1177–1182. of Lyme disease. Ophthalmology 1990;
Handbuch der Augerheilkunde. 2nd edn. 11. Golnik KC, Marotto ME, Fanous MM, et al: 97:699–706.
Leipzig, Germany: Englemann; 1916:1319. Ophthalmic manifestations of Rochalimaea 20. Mansour AM: Cytomegalovirus optic
2. Gass JDM: Diseases of the optic nerve that species. Am J Ophthalmol 1994; neuritis. Curr Opinion Ophthalmol 1997;
may simulate macular disease. Trans Am 118:145–151. 8:55–58.
Acad Ophthalmol Otolaryngol 1977; 12. Ulrich GG, Waecker NJ Jr, Meister SJ, et al: 21. Fusco R, Magli A, Guacci P: Stellate
83:766–769. Cat scratch disease associated with maculopathy due to Salmonella typhi.
3. Dreyer RF, Hopen G, Gass JDM, Smith JL: neuroretinitis in a 6-year-old girl. A case report. Ophthalmologica 1986;
Leber’s idiopathic stellate neuroretinitis. Ophthalmology 1992; 99:246–249. 192:154–158.
Arch Ophthalmol 1984; 102:1140–1145. 13. Weiss AH, Beck RW: Neuroretinitis in 22. Beck RW, Sergott RC, Barr CC,
4. Maitland CG, Miller NR: Neuroretinitis. Arch childhood. J Pediatr Ophthalmol Annesley WH: Optic disc edema in the
Ophthalmol 1984; 102:1146–1150. Strabismus 1989; 26:198–203. presumed ocular histoplasmosis syndrome.
5. Wolter JR, Philips RL, Butler GR: The star- 14. Ormerod LD, Skolnick KA, Menosky MM, Ophthalmology 1984; 91:183–185.
figure of the macular area. Arch Ophthalmol et al: Retinal and choroidal manifestations 23. Saxena R, Sethi HS, Rai HK, Menon V:
1958; 60:49–59. of cat scratch disease. Ophthalmology Bilateral neuro-retinitis following chick
6. Stewart JW, Brazis PW, Barrett KM, et al: 1998; 105:1024–1031. embryo cell anti-rabies vaccination:
Optical coherence tomography in a case of 15. Solley WA, Martin DF, Newman NJ, et al: a case report. BMC Ophthalmol
bilateral neuroretinitis. J Neuroophthalmol Cat scratch disease: posterior segment 2005; 5:20.
2005; 25:131–133. manifestations. Ophthalmology 1999; 24. Suhler EB, Lauer AK, Rosenbaum JT:
7. Bar S, Segal M, Shapira R, Savir H: 106:1546–1553. Prevalence of serologic evidence of cat
Neuroretinitis associated with cat-scratch 16. Johnson BL, Wisotzkey HM: Neuroretinitis scratch disease in patients with
disease. Am J Ophthalmol 1990; associated with herpes simplex neuroretinitis. Ophthalmology 2000;
110:703–705. encephalitis in an adult. Am J Ophthalmol 107:871–876.
8. Chrousos GA, Drack AV, Young M, et al: 1977; 83:481–489. 25. Bird AC, Smith JL, Curtin VT: Nematode
Neuroretinitis in cat scratch disease. J Clin 17. Selbst RG, Selhorst JB, Harbison JW, et al: optic neuritis. Am J Ophthalmol 1970;
Neuroophthalmol 1990; 10:92–94. Parainfectious optic neuritis: report and 69:72–77.
9. Reed JB, Scales DK, Wong MT, et al: review following varicella. Arch Neurol 26. Shoari M, Katz B: Recurrent neuroretinitis
Bartonella henselae neuroretinitis in cat 1983; 40:347–350. in an adolescent with ulcerative colitis.
scratch disease: diagnosis, management 18. Folk JC, Weingeist TA, Corbett JJ, et al: J Neuroophthalmol 2005; 25:286–288.
1862
Neuroretinitis

27. Carroll DM, Franklin RM: Leber’s idiopathic sclerosis? Arch Neurol 1987; review of the literature. Pediatr Infect Dis J
stellate retinopathy. Am J Ophthalmol 1982; 44:1045–1048. 1992; 11:474–478.
93:96–101. 30. Williams KE, Johnson LN: Neuroretinitis in 32. Purvin V, Ranson N, Kawasaki A: Idiopathic
28. Purvin VA, Chioran G: Recurrent patients with multiple sclerosis. recurrent neuroretinitis: effects of long-term
neuroretinitis. Arch Opthalmol 1994; Ophthalmology 2004; 111:335–341. immunosuppression. Arch Ophthalmol
112:365–371. 31. Margileth A: Antibiotic therapy for 2003; 121:65–67.
29. Parmley VC, Schiffman JS, Maitland CG, cat-scratch disease: clinical study of
et al: Does neuroretinitis rule out multiple therapeutic outcome in 268 patients and a

CHAPTER 140

1863
CHAPTER

141 Familial Exudative Vitreoretinopathy


Shizuo Mukai and Christopher M. Andreoli

In 1969, Criswick and Schepens1 reported on six patients with • Falciform retinal fold
abnormalities of the retina and vitreous that were clinically • Rhegmatogenous retinal detachment
similar to retinopathy of prematurity (ROP) but without a history • Massive intraretinal and subretinal exudation
of prematurity or oxygen supplementation. Two of the patients
were related by blood, and the other four patients were members
of another family and were related by blood. Criswick and Stage 1 describes the mild form of FEVR. Fundus examination
Schepens called this entity familial exudative vitreoretinopathy during this stage reveals vitreoretinal changes in the periphery
(FEVR; which is also known as Criswick–Schepens syndrome) between the equator and the ora serrata, including the white-with-
because of the familial and exudative nature of the disease. Both pressure and white-without-pressure signs, peripheral cystoid
FEVR and ROP are characterized by avascularity of the peripheral degeneration, and vitreous bands. There is a peripheral avascular
retina, which is especially evident on fluorescein angiography. zone that is difficult to see clinically. Exudation, neovasculariz-
In both diseases, reactive fibrovascular proliferations develop ation, and fibrovascular proliferation are not present at this
that may lead to cicatricial changes and retinal traction in the initial stage. Fluorescein angiography demonstrates the periph-
temporal periphery, resulting in dragged disks, ectopic maculae, eral avascular zone clearly (Fig. 141.1) that is present in the
retinal detachments, and falciform retinal folds. Visual loss is temporal periphery and can leak fluorescein. There is abnormal
primarily due to retinal detachment.2 arborization of the vessels in the periphery that terminates along
a scalloped or curvilinear border with the avascular zone. In cases
Synonyms with wider zones of peripheral avascularity, a wedge-shaped area
• FEVR of avascularization may be seen in the temporal meridian. Retinal
• Criswick–Schepens syndrome pigment epithelial changes are often associated with this V-shaped
zone of avascularization.4 In many individuals, the peripheral
avascular zone persists into adult life without regression or pro-
CLINICAL CHARACTERISTICS gression to proliferation.5 This is in contrast to ROP in which
the peripheral retina subsequently vascularizes.
FEVR is a vitreoretinal disorder that is clinically similar to ROP, It is not known why the peripheral retina does not vascularize
except that there is usually a positive family history, and there completely in FEVR. Whether this disease is a primary disorder
is no history of prematurity or oxygen supplementation. The
disease is often asymptomatic in the early stages and progresses
very slowly. Based on funduscopic and fluorescein angiographic
observations, Canny and Oliver3 developed a useful staging
scheme. The three stages describe mild, moderate, and severe
forms of FEVR, and each stage can be seen at any age.

Key Features
Mild (Stage 1)
• Peripheral avascular zone
• Abnormal aborization in periphery with:
• Vascular engorgement and telangiectasia
• Capillary microaneurysms
• Arteriovenous shunts
• Straightened vessels
Moderate (Stage 2)
• Neovascularization
• Fibrovascular proliferation
• Subretinal and intraretinal exudation
Severe (Stage 3)
• Cicatricial lesion in the temporal periphery FIGURE 141.1. Fluorescein angiogram demonstrates the peripheral
• Traction retinal detachment avascular zone, vascular engorgement and telangiectasia,
microaneurysms, and shunt vessels in stage-1 disease. 1865
RETINA AND VITREOUS

a b c
SECTION 10

FIGURE 141.2. (a) The temporal periphery in stage-2 FEVR. Fibrovascular ridge and retinal exudation are seen. (b) Early fluorescein angiogram
of the same area shows neovascularization at the vascular–avascular border. (c) Late fluorescein angiogram of the same area shows extensive
leakage of the fluorescein dye.
(a–c) Courtesy of Tatsuo Hirose, MD.

affecting retinal vessel development6,7 rather than a true vitreo-


retinopathy, as originally suggested by Criswick and Schepens,1
is still to be determined. Although congenital nonvascularization
of the retinal periphery is the most common characteristic of
the mildest phenotypes, vitreous changes are also noted in these
patients. It also remains uncertain whether a secondary vaso-
occlusive process (e.g., associated with hematologic disease)
contributes to the pathogenesis of FEVR. Laqua7 performed
extensive hematologic examinations on one of his patients with
FEVR and did not find any evidence of a hematologic disorder.
Platelet dysfunction may play a role in FEVR, but this is still
not fully determined.8–10
Stage 2 FEVR is the moderate form of the disease and
represents the proliferative and exudative stage. In addition to
the stage 1 findings already described, neovascularization, fibro-
vascular proliferation, and subretinal and intraretinal exudation
are seen, especially in the temporal periphery (Figs 141.2 and
141.3). A localized traction retinal detachment may also be
present. The fibrovascular lesions contract and exert traction on
the retina, producing ectopic maculae and dragged disks (Fig.
141.4).3 A fluorescein angiogram shows neovascularization FIGURE 141.3. The temporal periphery in stage 2 disease
developing from the retinal vessels at the vascular–avascular demonstrates intraretinal and subretinal exudation, pigmentary
border and growing anteriorly into the avascular retina. The changes, and straightening of retinal vessels.
temporal region of the retina is again the most common area for
neovascularization. There is abrupt cessation of the retinal
capillary network at the scalloped edge posterior to the fibro- constitute the major cause of visual loss in these patients. The
vascular mass. These terminal capillaries also leak fluorescein. incidence of retinal detachment ranges between 20% and
Fibrovascular masses stain with fluorescein. Large feeder vessels 32%.2,13–15 Rhegmatogenous retinal detachment is common –
may also be seen.11 between 25% and 63% of all retinal detachments in FEVR are
Stage 3 represents the advanced stage of FEVR. A cicatricial of this type. A variety of retinal breaks have been observed,
lesion in the temporal periphery pulls the retina and causes including round holes, horseshoe tears, and giant tears. Rheg-
traction retinal detachments, falciform retinal folds, and matogenous retinal detachments usually occur in the second
rhegmatogenous retinal detachments. Massive intraretinal and and third decades of life. At least in the Japanese population,
subretinal exudation is present and can result in an exudative rhegmatogenous retinal detachments from FEVR appear to
retinal detachment (Fig. 141.5). The exudative component of constitute a significant proportion of all rhegmatogenous retinal
the disease may resemble Coats’ disease and is different from detachments in this age group. Hashimoto and associates14
ROP. The degree of exudation is usually less than in Coats’ showed that in 576 consecutive cases of all rhegmatogenous
disease, and total retinal detachments from exudation are rare. retinal detachments, 5% were in FEVR patients. When these
Anterior segment changes, including cataract, iris atrophy, authors looked at all rhegmatogenous retinal detachments in
rubeosis iridis, and neovascular glaucoma, nonneovascular patients younger than 30 years of age, a surprising 12% were in
chronic angle-closure glaucoma, as well as band keratopathy patients with FEVR.
can also occur.3,12 Retinal breaks in FEVR are thought to be caused by a
1866 Retinal detachments are relatively common in FEVR and combination of vitreoretinal traction and retinal atrophy.
Familial Exudative Vitreoretinopathy

Falciform retinal folds are also relatively common and are


found in 15–39% of the eyes with FEVR.2,19 The folds usually
extend from the optic nerve head to the temporal or infero-
temporal periphery, at which point there are cicatricial
connections to the mass of fibrous tissue at the peripheral
retina and lens equator.20,21 The distance between the peripheral
retina and the lens equator is very short in the neonatal eye, and
this accounts for the high frequency of adhesions anteriorly to
the lens. The folds appear to be caused by severe traction of the
retina resulting from the organization of peripheral neovas-
cularization and formation of a fibrovascular mass. The folds
usually occur in the first decade of life and do not show
progression.
Traction retinal detachments are less common and occur at a
rate of 6–10% in eyes with FEVR.2,14 They usually occur before
the age of 10 years and show very little progression. In the cases
with isolated traction retinal detachment, vitrectomy alone may
be effective.22 Massive Coats’-like exudation can cause total

CHAPTER 141
retinal detachments (Fig. 141.5),23 but in general, significant
exudative retinal detachments appear to be rare.2
FIGURE 141.4. Patient with stage 2 FEVR with a dragged disk.
DIFFERENTIAL DIAGNOSIS

Differential Diagnosis
• Retinopathy of Prematurity
• Coats’ disease
• Sickle cell trait and disease
• Other hemoglobinopathies
• Persistent hyperplastic primary vitreous/persistent fetal
vasculature
• Toxocara canis infection
• Norrie disease
• Incontinentia pigmenti
• Hereditary falciform retinal fold
• Angiomatosis of the retina
• Autosomal dominant neovascularization
• Eales’ disease
• Pars planitis

FEVR is characterized by nonvascularization of the peripheral


retina, familial occurrence (usually autosomal dominant,
X-linked, or autosomal recessive), dragged disk and macula,
retinal exudation, retinal detachment, and retinal folds. The
FIGURE 141.5. Stage 3 FEVR in a patient with retinal hemorrhage clinical features are most similar to those of ROP and include
and severe exudative retinal detachment. peripheral nonvascularization, reactive fibrovascular prolifer-
ations, and cicatricial changes with severe traction on the retina.
Important differentiating features are a family history of the dis-
Vitreous bands and adhesions are seen even in the early stages order and no history of prematurity or oxygen supplementation.
of FEVR. Neovascularization and fibrovascular proliferation The peripheral avascular retina does not vascularize in FEVR,
produce additional traction. The peripheral retina in FEVR as it typically does in ROP, and exudation, which is common in
patients is also avascular and atrophic. Atrophic holes are seen FEVR, is very rare in ROP.7
in the periphery in many of the cases. The combination of In addition to ROP, possible alternative differential diagnoses
vitreoretinal traction and atrophic retina makes these eyes more of patients having peripheral retinal nonvascularization with
susceptible to retinal tears.2,15,16 The combined traction and neovascular proliferation include sickle-cell trait and disease as
rhegmatogenous retinal detachments that result tend to be well as other hemoglobinopathies, Eales’ disease, incontinentia
surgically difficult to repair and often lead to proliferative pigmenti, and autosomal dominant neovascularization as
vitreoretinopathy and require multiple procedures.2,15,17 There described by Gitter and co-workers.24 Diseases causing dragged
is also one reported case of significant postoperative uveitis.18 disks, ectopic maculae, and falciform retinal folds also need
Vitreous hemorrhage in FEVR patients is rare but may be an to be considered, especially hereditary falciform retinal folds,
ominous sign; two cases described by van Nouhuys2 resulted in persistent hyperplastic primary vitreous/persistent fetal
rapid progression to closed-funnel retinal detachments. In vasculature, Toxocara canis infection, Norrie disease, and
summary, the cases of retinal detachment in FEVR have many incontinentia pigmenti.19,25,26 Causes of peripheral retinal
of the difficulties seen in combined traction-rhegmatogenous fibrous or exudative mass lesion include Coats’ disease,
retinal detachment in young adults with predilection for pro- angiomatosis of the retina, Toxocara canis infection, and pars
liferative vitreoretinopathy. planitis.7,16 1867
RETINA AND VITREOUS

GENETICS number of fenestrated blood vessels as well as persistence of


hyaloid vasculature.44,45
A third locus, EVR3, segregating with a large kindred
Genetics demonstrating autosomal dominant FEVR has been identified
• Genetically heterogeneous on chromosome 11 at locus 11p12-13. The gene is currently
• Autosomal dominant unidentified.46
• X-linked A fourth locus, EVR4, contains the LRP5 gene which encodes
• Autosomal recessive a Wnt co-receptor, low-density lipoprotein receptor-related
• Variable clinical expression protein-5 (LRP5). Mutations in this gene cause an autosomal
dominant form of FEVR.47,48 This gene is located on
chromosome 11 at locus 11q13, distinct from EVR1 (FZD-4)
FEVR is a genetically heterogeneous disease with autosomal and EVR3. It has been shown that mutations in this same gene
dominant, autosomal recessive, and X-linked inheritance pos- can cause an autosomal recessive form of FEVR as well.49
sible with variable clinical expression. Multiple loci and genes Frizzled-4 and LRP5 act as a functional receptor pair in the Wnt
have been implicated. Penetrance is variable, depending on the signaling pathway.50
method of diagnosis. When using fluorescein angiography to Toomes has suggested a fourth autosomal dominant locus
determine clinical status, penetrance is reported to be 100% distinct from EVR1, EVR3, and EVR4.51 Autosomal recessive
because all affected individuals have a sector of avascular periph- pedigrees have also been described; however, an underlying
SECTION 10

eral retina.27 When using examination of the retina with an genetic defect has not been determined.52
indirect ophthalmoscope through a dilated pupil to determine Genetic sequence analysis is available on a research basis for
clinical status, penetrance is considered to be ~90%.20 When NDP, FZD-4, and LRP. Because the autosomal dominant,
using reduced vision or other clinical symptoms to determine autosomal recessive, and X-linked forms of FEVR cannot be
clinical status, penetrance as low as 10% has been reported. distinguished clinically, determining the mode of inheritance
The first locus discovered, EVR1, contains the FZD-4 gene for a particular family can be difficult, and for the simplex case,
encoding the Wnt receptor Frizzled-4.28,29 Mutations in FZD-4 may not be possible.
are responsible for autosomal dominant forms of FEVR. This
gene maps to the long arm of human chromosome 11 at locus PATHOLOGIC FEATURES
11q13-23. Multiple families have been discovered to have novel
missense and nonsense mutations in this gene causing a similar Histopathologic studies of six eyes enucleated for end-stage
FEVR phenotype.28,30–34 The FZD-4 family of genes encode for FEVR have been reported.53–56 These eyes were enucleated for
the Frizzled family of seven-pass transmembrane receptors that phthisis, neovascular glaucoma, acute angle closure, and possible
bind to multiple ligands including the Wnt family of proteins. retinoblastoma (in a 6-week-old infant). Many of the histo-
This receptor–ligand pair is implicated in development, cell pathologic findings are those typically associated with chronic
proliferation, and carcinogenesis.29,35 A missense mutation in retinal detachments, neovascular glaucoma, and phthisis
FZD4 has been reported in a single case of advanced ROP.36 bulbi. This discussion concentrates on the findings unique to
The second FEVR locus, EVR-2, has been found to contain the FEVR. No pathology of either stage 1 or stage 2 disease is
same gene responsible for Norrie disease (ND) and has been shown available.
to be mutated in cases of X-linked FEVR.37,38 ND is clinically Retinal detachment and prominent preretinal and vitreal mem-
characterized by bilateral retrolental grayish-yellow fibrovascular brane formation are seen in all cases. Brockhurst and colleagues55
masses. This has traditionally been thought to arise from primary observed a thick, acellular, preretinal membrane consisting of
retinal dysplasia.39 With age, the disease progresses to include amorphous material. This membrane is seen just posterior to
cataract, posterior and anterior synechiae, iris atrophy, corneal the ora serrata and extends posteriorly. Glazer and associates
opacification, band keratopathy, and blindness with phthisis bulbi. showed by electron microscopy that the membranes consist of
Mutations in the Norrie gene (NDP) on the human X chromo- fibrovascular tissue with astrocytes.57 Multiple adhesions to the
some at locus Xp11.4 which encodes ND protein (Norrin), have retina cause large retinal folds. Acellular fibrous membrane with
been shown to cause a spectrum of diseases including ND pigment is also seen in the vitreous. Van Nouhuys’54 case of the
(nonsense mutation) and X-linked FEVR (missense mutations). 6-week-old infant with possible retinoblastoma shows the
There are also reported cases suggesting a role of this gene in preretinal membrane to extend much more anteriorly and
the pathogenesis of persistent hyperplastic primary vitreous/ attach to the lens capsule. Intraretinal and subretinal exudation
persistent fetal vasculature, Coats’ disease, and advanced is also seen, but the degree of exudation and the extent of
ROP.40 These diseases all appear to share abnormalities in telangiectatic vessels are less than those usually seen in Coats’
retinal vasculature. disease.56 Nicholson and Galvis53 observed areas of necrosis and
There is some evidence that a genotype–phenotype relationship acute inflammation surrounding the fibrovascular tissue in the
exists with mutations in NDP. It is suggested that missense temporal periphery of the retina.
mutations affecting the carboxy terminus of the protein result in
a less severe phenotype manisfested as X-linked FEVR.37,41 How- PREVENTION AND TREATMENT
ever, there is also significant phenotypic variation within families
carrying the same NDP mutation.37,42 Allen et al suggests that all The major cause of visual loss in cases of FEVR is retinal
phenotypes associated with a defect in NDP should be classified detachment. When retinal detachments occur in eyes with
as ND with a range of expressivity, and that X-linked FEVR FEVR, they tend to be complicated and difficult to repair, often
should be reserved for any cases which do not involve a defect requiring multiple surgical procedures.2,57,58 Some of the
in NDP.42 detachments are similar to those seen in ROP. Retinal detach-
Xu et al43 established Norrin as a ligand for Frizzled-4, a pre- ments are more difficult and have the worst prognosis in
sumptive Wnt receptor. They propose that Norrin–Frizzled-4 younger children; rhegmatogenous retinal detachment in an
binding serves as a signaling system involved in vascular develop- adolescent FEVR patient has high likelihood of proliferative
ment.43 Both FZD-4 and NDP knockout mice have been shown to vitreoretinopathy and redetachment. The key to retinal
1868 display increased retinal vascular permeability with an increased detachment repair in FEVR patients is to release the peripheral
Familial Exudative Vitreoretinopathy

vitreoretinal traction using meticulous membrane dissection would be difficult to carry out because of the rarity and slow
and scleral buckling.17 progression of the disease. It is currently not clear whether we
As in ROP, the goal is to try to prevent the detachments of can prevent the retinal detachments that occur quite early in life,
the retina seen in stage 3, advanced FEVR. Early examination since stage 3 FEVR can be already present in very young patients.60
(including dilated fundus examination with scleral depression) Presence of the cicatricial process makes the direct detection
of children who have a positive family history is mandatory in of subtle retinal detachments difficult. Vitreous cells and haze,
identifying those with early proliferative changes. The neovascular poor mydriasis, aqueous cells and flare, and posterior synechiae
and fibrovascular process can then be treated by prophylactic are all signs of retinal detachment in a FEVR patient. Ultra-
photocoagulation or cryotherapy. There are very few descriptions sonography is an important test in the diagnosis of retinal
of FEVR in neonates and infants, and early examination will detachments in cases with media opacities. An increase in angle
also give us a better understanding of how FEVR presents in this kappa is evidence of increasing peripheral traction.2 The role of
population. The application of retinoablative procedures to the electrophysiologic testing is uncertain.61
avascular retina alone or together with the neovascular frond
appears to be effective in aborting the proliferative process.2,59
Treatment Options
As in treated cases of ROP, the destruction of the proliferative
• Photocoagulation or cryotherapy
process is thought to diminish the subsequent cicatricial process
• Scleral buckle
that leads to retinal detachment. A randomized study to test the
• Vitrectomy with meticulous membrane dissection
effectiveness of prophylactic cryotherapy or photocoagulation

CHAPTER 141
REFERENCES
1. Criswick VG, Schepens CL: Familial 15. Benson WE: Familial exudative 28. Li Y, Fuhrmann C, Schwinger E, et al: The
exudative vitreoretinopathy. Am J vitreoretinopathy. Trans Am Ophthalmol gene for autosomal dominant familial
Ophthalmol 1969; 68:578–594. Soc 1995; 93:473–521. exudative vitreoretinopathy (Criswick-
2. van Nouhuys CE: Juvenile retinal 16. Dudgeon J: Familial exudative vitreo- Schepens) on the long arm of chromosome
detachment as a complication of familial retinopathy. Trans Ophthalmol Soc UK 11. Am J Ophthalmol 1992; 113:712–713.
exudative vitreoretinopathy. Fortschr 1979; 99:45–49. 29. Robitaille J, MacDonald ML, Kaykas A,
Ophthalmol 1989; 86:221–223. 17. Tano Y, Ikeda T: Treatment of familial et al: Mutant frizzled-4 disrupts retinal
3. Canny CL, Oliver GL: Fluorescein exudative vitreoretinopathy with pars plana angiogenesis in familial exudative
angiographic findings in familial exudative vitrectomy. Ophthalmology 1990; vitreoretinopathy. Nat Genet 2002;
vitreoretinopathy. Arch Ophthalmol 1976; 97(Suppl):152. 32:326–330.
94:1114–1120. 18. Okubo Y, Okubo A, Kubono T, Shimizu H: 30. Yoshida S, Arita R, Yoshida A, et al: Novel
4. Miyakubo H, Hashimoto K, Miyakubo S: Familial exudative vitreoretinopathy, mutation in FZD4 gene in a Japanese
Retinal vascular pattern in familial rhegmatogenous retinal detachment and pedigree with familial exudative
exudative vitreoretinopathy. Ophthalmology postoperative uveitis with massive vitreoretinopathy. Am J Ophthalmol 2004;
1984; 91:1524–1530. subretinal exudation. Nippon Ganka Gakkai 138:670–671.
5. Tasman W, Augsburger JJ, Shields JA, Zasshi 1984; 88:1151–1156. 31. Toomes C, Bottomley HM, Scott S, et al:
et al: Familial exudative vitreoretinopathy. 19. Nishimura M, Yamana T, Sugino M, et al: Spectrum and frequency of FZD4 mutations
Trans Am Ophthalmol Soc 1981; Falciform retinal fold as sign of familial in familial exudative vitreoretinopathy. Invest
79:211–226. exudative vitreoretinopathy. Jpn J Ophthalmol Vis Sci 2004; 45:2083–2090.
6. Gow J, Oliver GL: Familial exudative Ophthalmol 1983; 27:40–53. 32. Price SM, Periam N, Humphries A, et al:
vitreoretinopathy. An expanded view. 20. van Nouhuys CE: Dominant exudative Familial exudative vitreoretinopathy linked
Arch Ophthalmol 1971; 86:150–155. vitreoretinopathy and other vascular to D11S533 in a large Asian family with
7. Laqua H: Familial exudative developmental disorders of the consanguinity. Ophthalmic Genet 1996;
vitreoretinopathy. Albrecht Von Graefes Arch peripheral retina. Doc Ophthalmol 1982; 17:53–57.
Klin Exp Ophthalmol 1980; 213:121–133. 54:1–414. 33. Kondo H, Hayashi H, Oshima K, et al:
8. Chaudhuri PR, Rosenthal AR, Goulstine DB, 21. Campo RV: Similarity of familial exudative Frizzled 4 gene (FZD4) mutations in
et al: Familial exudative vitreoretinopathy vitreoretinopathy and retinopathy of patients with familial exudative
associated with familial thrombocytopathy. prematurity. Arch Ophthalmol 1983; vitreoretinopathy with variable expressivity.
Br J Ophthalmol 1983; 67:755–758. 101:821. Br J Ophthalmol 2003; 87:1291–1295.
9. Friedrich CA, Francis KA, Kim HC: Familial 22. Maguire AM, Trese MT: Visual results of 34. Omoto S, Hayashi T, Kitahara K, et al:
exudative vitreoretinopathy (FEVR) and lens-sparing vitreoretinal surgery in infants. Autosomal dominant familial exudative
platelet dysfunction. Br J Ophthalmol 1989; J Pediatr Ophthalmol Strabismus 1993; vitreoretinopathy in two Japanese families
73:477–478. 30:28–32. with FZD4 mutations (H69Y and C181R).
10. Gole GA, Goodall K, James MJ: Familial 23. Ebert EM, Mukai S: Familial exudative Ophthalmic Genet 2004; 25:81–90.
exudative vitreoretinopathy. Br J vitreoretinopathy. Int Ophthalmol Clin 1993; 35. Huang HC, Klein PS: The Frizzled family:
Ophthalmol 1985; 69:76. 33:237–247. receptors for multiple signal transduction
11. Nijhuis FA, Deutman AF, Aan de Kerk AL: 24. Gitter KA, Rothschild H, Waltman DD, et al: pathways. Genome Biol 2004; 5:234.
Flourescein angiography in mild stages of Dominantly inherited peripheral retinal 36. MacDonald ML, Goldberg YP, Macfarlane
dominant exudative vitreoretinopathy. Mod neovascularization. Arch Ophthalmol 1978; J, et al: Genetic variants of frizzled-4 gene
Probl Ophthalmol 1979; 20:107–114. 96:1601–1605. in familial exudative vitreoretinopathy and
12. Azuara-Blanco A, Pesin SR, Katz LJ, et al: 25. van Nouhuys CE: Congenital retinal fold as advanced retinopathy of prematurity. Clin
Familial exudative vitreoretinopathy a sign of dominant exudative Genet 2005; 67:363–366.
associated with nonneovascular chronic vitreoretinopathy. Albrecht Von Graefes 37. Riveiro-Alvarez R, Trujillo-Tiebas MJ,
angle-closure glaucoma. J Glaucoma 1997; Arch Klin Exp Ophthalmol 1981; 217:55–67. Gimenez-Pardo A, et al: Genotype-
6:47–49. 26. Chang-Godinich A, Paysse EA, Coats DK, phenotype variations in five Spanish
13. Miyakubo H, Inohara N, Hashimoto K: Holz ER: Familial exudative families with Norrie disease or X-linked
Retinal involvement in familial exudative vitreoretinopathy mimicking persistent FEVR. Mol Vis 2005; 11:705–712.
vitreoretinopathy. Ophthalmologica 1982; hyperplastic primary vitreous. Am J 38. Shastry BS, Hejtmancik JF, Trese MT:
185:125–135. Ophthalmol 1999; 127:469–471. Identification of novel missense mutations
14. Hashimoto K, Miyakubo H, Inohara N, Tada 27. Ober RR, Bird AC, Hamilton AM, Sehmi K: in the Norrie disease gene associated with
H: Juvenile retinal detachment and familial Autosomal dominant exudative one X-linked and four sporadic cases of
exudative vitreoretinopathy. Jpn J Clin vitreoretinopathy. Br J Ophthalmol 1980; familial exudative vitreoretinopathy. Hum
Ophthalmol 1983; 37:797–803. 64:112–120. Mutat 1997; 9:396–401. 1869
RETINA AND VITREOUS

39. Warburg M: Norrie’s disease. A congenital 47. Toomes C, Downey LM, Bottomley HM, 54. Mintz-Hittner HA, Ferrell RE, Sims KB,
progressive oculo-acoustico-cerebral et al: Identification of a fourth locus (EVR4) et al: Peripheral retinopathy in offspring of
degeneration. Acta Ophthalmol (Copenh) for familial exudative vitreoretinopathy carriers of Norrie disease gene mutations.
1966; 89(Suppl):1–47. (FEVR). Mol Vis 2004; 10:37–42. Possible transplacental effect of abnormal
40. Shastry BS, Pendergast SD, Hartzer MK, 48. Qin M, Hayashi H, Oshima K, et al: Norrin. Ophthalmology 1996;
et al: Identification of missense mutations Complexity of the genotype-phenotype 103:2128–2134.
in the Norrie disease gene associated with correlation in familial exudative 55. Brockhurst RJ, Albert DM, Zakov ZN:
advanced retinopathy of prematurity. Arch vitreoretinopathy with mutations in the Pathologic findings in familial exudative
Ophthalmol 1997; 115:651–655. LRP5 and/or FZD4 genes. Hum Mutat vitreoretinopathy. Arch Ophthalmol 1981;
41. Meindl A, Lorenz B, Achatz H, et al: 2005; 26:104–112. 99:2143–2146.
Missense mutations in the NDP gene in 49. Jiao X, Ventruto V, Trese MT, et al: 56. Boldrey EE, Egbert P, Gass JD, Friberg T:
patients with a less severe course of Norrie Autosomal recessive familial exudative The histopathology of familial exudative
disease. Hum Mol Genet 1995; 4:489–490. vitreoretinopathy is associated with vitreoretinopathy. A report of two cases.
42. Allen RC, Russell SR, Streb LM, et al: mutations in LRP5. Am J Hum Genet 2004; Arch Ophthalmol 1985; 103:238–241.
Phenotypic heterogeneity associated with a 75:878–884. 57. Glazer LC, Maguire A, Blumenkranz MS,
novel mutation (Gly112Glu) in the Norrie 50. Pinson KI, Brennan J, Monkley S, et al: et al: Improved surgical treatment of
disease protein. Eye 2006; 6:234–241. An LDL-receptor-related protein mediates familial exudative vitreoretinopathy in
43. Xu Q, Wang Y, Dabdoub A, et al: Wnt signalling in mice. Nature 2000; children. Am J Ophthalmol 1995;
Vascular development in the retina and 407:535–538. 120:471–479.
inner ear: control by Norrin and Frizzled-4, 51. Toomes C, Downey LM, Bottomley HM, et al: 58. Uto H, Shigeto M, Tanaka H, et al: A case
a high-affinity ligand-receptor pair. Cell Further evidence of genetic heterogeneity of 11q-syndrome associated with
SECTION 10

2004; 116:883–895. in familial exudative vitreoretinopathy; abnormalities of the retinal vessels.


44. Richter M, Gottanka J, May CA, et al: exclusion of EVR1, EVR3, and EVR4 in a Ophthalmologica 1994; 208:233–236.
Retinal vasculature changes in Norrie large autosomal dominant pedigree. Br J 59. Bergen RL, Glassman R: Familial exudative
disease mice. Invest Ophthalmol Vis Sci Ophthalmol 2005; 89:194–197. vitreoretinopathy. Ann Ophthalmol 1983;
1998; 39:2450–2457. 52. de Crecchio G, Simonelli F, Nunziata G, 15:275–276.
45. Rehm HL, Zhang DS, Brown MC, et al: et al: Autosomal recessive familial 60. Slusher MM, Hutton WE: Familial exudative
Vascular defects and sensorineural exudative vitreoretinopathy: evidence for vitreoretinopathy. Am J Ophthalmol 1979;
deafness in a mouse model of Norrie genetic heterogeneity. Clin Genet 1998; 87:152–156.
disease. J Neurosci 2002; 22:4286–4292. 54:315–320. 61. Ohkubo H, Tanino T: Electrophysiological
46. Downey LM, Keen TJ, Roberts E, et al: 53. Nicholson DH, Galvis V: findings in familial exudative
A new locus for autosomal dominant Criswick–Schepens syndrome (familial vitreoretinopathy. Doc Ophthalmol 1987;
familial exudative vitreoretinopathy maps to exudative vitreoretinopathy). Study of a 65:461–469.
chromosome 11p12-13. Am J Hum Genet Colombian kindred. Arch Ophthalmol 1984;
2001; 68:778–781. 102:1519–1522.

1870
CHAPTER

142 Central Serous Chorioretinopathy


Catherine B. Meyerle and Richard F. Spaide

DEFINITION In terms of gender, there is a male predilection with the


reported male:female ratio ranging from 6:1 in older studies1–10
Central serous chorioretinopathy (CSC) is characterized by an and less than 3:1 in more recent literature.12,13 Race also plays
idiopathic circumscribed serous retinal detachment, usually a role in CSC, with more whites, Hispanics, and Asians
confined to the central macula, caused by leakage of fluid affected. CSC has been reported as uncommon in blacks, but
through the retinal pigment epithelium (RPE) as defined by some authors refute this point.15 The disease may run a more
fluorescein angiography. Eyes with CSC do not have signs of severe course in Hispanics and Asians with extensive bullous
intraocular inflammation, accelerated hypertension, infiltration changes. Refractive error is significant with hyperopic or
or infarction of the choroid or RPE. Serous detachments of the emmetropic patients affected more in Western nations.16 This
RPE are frequently present. Although most cases are acute with association, however, does not hold for those in Asian regions
minimal sequela, some patients have a more chronic version of with a high prevalence of myopia such as in Japan.
the disease with poorer visual prognosis. Corticosteroids have been implicated in CSC by multiple
studies. Tittl and associates, in a retrospective case-control study
HISTORICAL BACKGROUND of 230 patients, found that corticosteroids (used by 9.1% of
patients) are risk factors in addition to psychotropic medications
CSC has a long history of changing names reflective of the and hypertension.12 Haimovici and co-workers13 also reported
previous uncertainty about the etiology of the pathological pro- an association of CSC with corticosteroids (used by 14.4% of
cess. von Graefe first described it as a recurrent central retinitis.1 patients) in a retrospective case-control study of 312 patients.
Horniker2 agreed that the pathology was localized to the retina but Other risk factors, according to Haimovici13, included pregnancy,
thought these patients had an underlying angioneurosis causing antibiotics, alcohol, untreated hypertension, and allergic respi-
angiospasm and exudation. He used the name capillarospastic ratory disease, with the pregnancy association also reported by
central retinitis. Gifford and Marquardt3 shared Horniker’s view Gass.17 Karadimas eventually confirmed that corticosteroids are
and coined the term central angiospastic retinopathy. Bennet a risk factor for CSC in a smaller, but prospective, case-control
also localized the disease to the retina and applied the name study.18 Systemic corticosteroid treatment in organ transplant
central serous retinopathy.4 Through fluorescein angiography, patients has been associated with aggressive CSC.19,20 More recent
Maumenee5 changed the concept of the primary tissue affected studies have shown that fundus changes previously attributed
by describing the leak occurring at the level of the RPE. Gass6 to organ transplantation21 were in fact most likely due to corti-
expanded on this view with his description of the fluorescein costeroid-induced CSC, as patients who never had organ trans-
angiographic findings and named the condition idiopathic plantation but were on corticosteroids had similar findings.22
central serous choroidopathy. Today, since we understand that Inhaled, intranasal, intramuscular, and topical dermatological23–26
hyperpermeability at the choroid causes a leak through the RPE corticosteroids have also been implicated in CSC pathogenesis.
resulting in a neurosensory retinal detachment, CSC is the There have even been isolated case reports of CSC following
preferred term. vitrectomy with intravitreal triamcinolone acetonide for diabetic
macular edema27 and CSC after a periocular steroid injection
DEMOGRAPHICS for HLA-B27-associated iritis.28 Our associates, however, have
performed hundreds of intravitreal triamcinolone acetonide
CSC has certain common demographic features.1–11 Historically injections and have not yet observed a case of CSC. Endogenous
the mean patient age was reported as 20–45 years, but more corticosteroids are also thought to contribute to the pathoge-
recent studies have not substantiated these numbers and actually nesis of disease as many CSC patients have elevated 24-h urine
report an older mean age. In a 1996 study of 130 patients with corticosteroids.29 Repeated injections of intramuscular pred-
CSC, the mean age was 51 years.11 Two recent large retrospec- nisolone in combination with intravenous epinephrine in a
tive case control studies concur with this older mean age. A cynomolgus monkey (Macaca irus) resulted in CSC after the
study of 230 patients found the mean age to be 51 years12 and thirty-second injection.30 Some question the validity of this animal
another study of 312 patients revealed a mean age of 45 years.13 model, however, based on the histopathology. Ultrastructural
If diagnosed in a patient older than 50 years of age, however, findings of RPE degeneration with underlying damaged endo-
one must carefully assess for macular degeneration or choroidal thelial cells within the inner surface of the choriocapillaris31 may
neovascularization (CNV) before making the diagnosis of CSC. be more consistent with infarction seen in malignant hyperten-
CSC is very rare in patients less than 20 years old. Although sion. Yoshioka defends his CSC animal model by pointing out
CSC in a 7-year-old girl has been reported, this patient may that the monkeys in his study did not have any Elsching’s spots
actually have had posterior scleritis.14 or retinal vasculature changes consistent with hypertension.30 1871
RETINA AND VITREOUS

Similar to corticosteroids, both exogenous and endogenous (DRPE), and bullous retinal detachment. Classic or acute CSC
sympathomimetic agents have been implicated in CSC. Exces- is the most common form, consisting of a solitary, localized
sive use of sympathomimetic compounds has been associated neurosensory detachment in the posterior pole. A round to oval,
with CSC.32 Endogenous plasma concentrations of epinephrine well-delineated, shallow serous retinal detachment elevates the
and norepinephrine were found to be higher among CSC macula and blunts the normal foveal reflex (Fig. 142.1). This
patients than in controls in one study.33 Repeated injections of blister of clear fluid has a characteristic halo formed by light
intravenous epinephrine alone in a Japanese monkey model reflexes where the sloping retina reflects light back to the observer.
(Macaca fuscatus) resulted in CSC after 39 injections.30 The size of the detachment can vary considerably, but the average
Personality traits are thought to play a role in CSC. Patients size is 2 disk diameters. Serous retinal pigment epithelial detach-
with CSC commonly are stressed by multiple events in their ments (PEDs) are also commonly seen in association with CSC
life.34 Based on the the Jenkins Activity Survey, Yannuzzi showed (Fig. 142.2). On biomicroscopy, a PED appears as a well-
that these patients are more likely to have type A personality circumscribed, orange-colored elevation with a slightly darker
characteristics when compared with controls.35 Werry and rim that can illuminate with a slit beam aimed from the side. A
Arends,36 using the Minnesota Multiphasic Personality Inven- PED forms when the RPE cells, and their associated basement
tory Test, showed that CSC patients are more likely to show membrane, separate from the underlying Bruch’s membrane.
hypochondria or hysteria and have a conversional neurosis. These detachments are often less than 0.25 disk diameter, but
can be larger. Prominent PEDs are found in two other conditions
OCULAR SYMPTOMS besides CSC: CNV, particularly occult CNV, and polypoidal
SECTION 10

choroidal vasculopathy, a variant of CNV. Therefore, if turbid


Common symptoms of CSC are decreased vision with distor- fluid or subretinal blood is noted, the macular pathology is most
tions including metamorphopsia, micropsia, scotomas, chroma- likely not CSC and is rather secondary to CNV.
topsia, and prolonged after-images. The visual acuity, somewhat Some patients may have the deposition of subretinal fibrin.
improvable with mild hyperopic correction, is usually reduced The subretinal fibrin is a grayish-white sheet overlying promi-
to between 20/30 and 20/60 but can decline to worse than nent leaks. Frequently, there may be a concomitant underlying
20/200 in severe or recurrent disease. Superior field defects can PED with the fibrin accumulating radially around the top of
occur in patients with inferior detachments from gravitating the PED making a ring appearance.
fluid. Younger CSC patients usually have unilateral involve-
ment, while older patients are more likely to have bilateral FLUORESCEIN ANGIOGRAPHIC FINDINGS
involvement. OF ‘CLASSIC’ CSC
OCULAR FINDINGS OF CLASSIC CSC Fluorescein angiography in acute cases of classic CSC demon-
strates one or several hyperfluorescent leaks at the level of the
CSC can present in three different ways: classic acute chorio- RPE. Early in the angiogram, there is a focal dot-like hyper-
retinopathy, chronic diffuse retinal pigment epitheliopathy fluorescence representing the leakage of dye from the choroid

FIGURE 142.1. (a) Red-free photography


shows a trace amount of fluid. (b) OCT better
illustrates the presence of both subretinal fluid
(asterisk) and a small pigment epithelial
detachment (arrow).

a b

a b c

FIGURE 142.2. (a) Red-free photograph illustrates a classic pigment epithelial detachment. (b and c) ICG angiography shows the typical early
hyperfluorescence and late hypofluorescence of the pigment epithelial detachment.
1872
Central Serous Chorioretinopathy

FIGURE 142.3. (a) The typical pinpoint leak is


present on fluorescein angiogram. (b) Magnified
view of the macula shows a pigment epithelial
detachment resulting from the leak.

a b

through the RPE. Later, dye accumulates beneath the sensory INDOCYANINE GREEN ANGIOGRAPHIC

CHAPTER 142
retinal detachment but does not extend outside the borders of FINDINGS OF ‘CLASSIC’ CSC
the detachment. Two patterns of leakage occur. First, the classic
smokestack leak described by Shimizu and Tobari in 197137 Indocyanine green (ICG) angiography demonstrates multifocal
occurs in the minority of cases (10%), but has a dramatic areas of choroidal vascular hyperpermeability (Fig. 142.4).40–42
appearance. The leakage first arises superiorly, resembling a These areas are best seen in the mid-phases of the angiogram,
smokestack, and then plumes out laterally. This pattern is and appear localized within the inner choroid. With time the
thought to be related to convection currents and a protein liver removes the ICG from the circulation, and the dye that has
gradient, with protein concentration increasing in the fluid leaked into the choroid appears to disperse somewhat, parti-
accumulating under the neurosensory detachment.38 Smoke- cularly into the deeper layers of the choroid.42 This produces a
stack leaks are usually associated with larger areas of retinal characteristic appearance of larger but less prominent hyper-
detachment. The second and more common pattern of dye fluorescent patches in the choroid with silhouetting of the larger
leakage, occurring in up to 90% of cases, is manifested as a choroidal vessels in the later phases of the ICG angiographic
small dot leak with a uniform filling pattern within the evaluation. Patients with CSC can actually present with no
detachment (Fig. 142.3). fluorescein leakage during a quiescent phase, but will still have
In both types of leakage patterns, focal leaks are somewhat areas of underlying choroidal vascular hyperpermeability that
more common nasally than temporally, superiorly than serves as a marker of the disease. Younger patients may have
inferiorly.9,39 Most leaking points are in a ring 1 mm wide, PEDs as a forme fruste of CSC in that underlying choroidal
starting 0.5 mm from the center of the fovea, but can occur hyperpermeability may cause elevations of the RPE without
further than 3 mm from the foveal avascular zone in 11.8% of creating breakthrough leaks.
cases. The leakage point occurs in the fovea in less than
10% of cases and within the papillomacular bundle in 20–25% AUTOFLUORESCENCE PATTERNS OF
of cases. If a leakage point is not readily apparent on fluorescein CLASSIC CSC
angiography, the superior extramacular area should be inspected
carefully as gravity might cause a detachment below the leak. In Autofluorescence photography is a noninvasive test that pro-
some cases, a leakage point will not be found because the leak vides functional images of the fundus based on stimulated
has healed. In these cases, the detachment will usually resolve emission of light from lipofuscin and related molecules.
in days to weeks. Finally, other conditions such as exudative Lipofuscin, found within the RPE, is a cellular waste product
age-related macular degeneration (AMD) may mimic CSC containing lipid, protein, and fluorophores such as A2E. Addi-
clinically, and thus a leakage point will not be found. tional sources of autofluorescence are the precursors to A2E

a b c

FIGURE 142.4. (a) Early phase; (b) mid phase; (c) late phase of ICG. ICG angiography demonstrates multiple patchy areas of choroidal
hyperpermeability, best seen in mid phase (b). These hyperpermeability regions may persist in chronic central serous even when there is no
active leakage on fluorescein angiography.
1873
RETINA AND VITREOUS

that accumulate in the outer retina prior to phagocytosis. These patients. This imaging technique will not only scientifically
A2E precursors increase in number if the outer segments are help us better understand the pathogenesis of CSC, but will also
not being phagocytized. clinically guide us in management of individual patients.
Autofluorescence images can be generated by excitation of
the fluorophores with light centered at 580 nm, detection above DIFFUSE RETINAL PIGMENT
695 nm with a barrier filter and recording by a Topcon 50µ EPITHELIOPATHY
fundus camera. The filter combination ensures that autofluor-
escence from the natural crystalline lens is bypassed. Alter- A second principal presentation of CSC shows widespread
natively, a Heidelberg retina angiograph (HRA) can be used with alteration of pigmentation of the decompensating RPE in the
excitation at 488 nm and detection above 500 nm. HRA is based posterior pole that appears to be related to the chronic presence
on confocal imaging. The advantage of the confocal aspect is of subretinal fluid. This variant of CSC has been termed ‘DRPE’
that it allows the HRA system to reject lens autofluorescence. or ‘chronic CSC’.42 Just as CSC has had many names during its
The disadvantage of the confocal imaging is that it collects light history, so has this more chronic variant. DRPE is related to not
only from the RPE. If there is a retinal detachment, as in CSC, only a past history of CSC, but also to the age of the patient
the HRA does not record the light coming from the retina. With at the time of diagnosis.11 Patients with DRPE generally have a
a fundus camera, however, it is possible to see the autofluorescence more pronounced loss of visual acuity, and may have perma-
from the RPE and outer retina simultaneously. The resulting nent loss of visual acuity to the level of legal blindness.
autofluorescence photograph obtained with the fundus camera,
SECTION 10

therefore, is a summation of the autofluorescence from the OCULAR FINDINGS OF DRPE


retina and underlying RPE. Visualization of the autofluorescent
signal depends on the distribution pattern of the fluorophore- Patients with DRPE have widespread disease. Close examin-
containing lipofuscin and A2E precursors. Autofluorescence ation of the retina by slit-lamp biomicroscopy may show thin-
photography is important in CSC because it provides a non- ning of the retina and possible cystic changes within the retina.
invasive technique to study the status of both the RPE and outer There are often RPE alterations that are manifest in three differ-
retina and to assess atrophic changes. ent ways. First, the RPE can show atrophy where there is a loss
Von Ruckman and Bird reported abnormalities of fundus of pigmentation and increased visibility of the underlying larger
autofluorescence in a prospective study of 69 eyes with CSC.43 choroidal vessels. This atrophy is readily seen with autofluor-
In this study, acute CSC was defined by a focal leak on angio- escence photography. Second, the RPE can have areas of focal
graphy with serous retinal detachment or pigment epithelial hyperpigmentation. Finally, some patients may have RPE hyper-
detachment. In the acute CSC eyes, increased autofluorescence plasia to the point where they develop bone spicules. Within the
noted at the leakage site and in the area of retinal detachment detachment, the subretinal fluid is clear, but sometimes has
was attributed to increased metabolic activity of the RPE. In flecks of subretinal lipid. This feature may suggest the presence
longstanding lesions of acute or classic CSC patients, decreased of occult CNV when in fact the diagnosis is CSC. Because CNV
or absent autofluorescence was reported to indicate reduced has a different course and treatment, great care must be made
metabolic activity of the RPE due to photoreceptor cell loss. in establishing the diagnosis of CSC in an older patient with
A retrospective German study of 85 patients evaluated auto- subretinal lipid. Broad areas of detachment in the posterior pole
fluorescence changes in both acute and chronic CSC patients, are present and tracts of fluid may descend inferiorly toward the
with acute defined as less than 6 weeks.44 The findings for the equator. Yannuzzi and colleagues7 described 25 patients with
acute patients were that 72% had decreased autofluorescence at extramacular inferior hemispheric RPE atrophic tracts related to
the leakage site and 77% had decreased autofluorescence in the an antecedent retinal detachment. Five of these patients
area of the neurosensory detachment. The authors suggest that actually had a peripheral retinal detachment.
the hypoautofluorescence in the acute stage is due to blockage
caused by edema. ANGIOGRAPHIC FINDINGS OF DRPE
Our group’s case series evaluating autofluorescence in 58
eyes with CSC expanded on these previous studies by including The various diffuse areas of altered RPE are readily apparent on
optical coherence tomography (OCT) analysis and statistical fluorescein angiography. These areas have a granular hyper-
measurement of autofluorescence values.45 The acute leaks of fluorescence due to relative atrophy of the involved RPE and
classic CSC, if imaged within the first month, showed no apparent associated subtle, indistinct leaks (Fig. 142.5). There may be
abnormal autofluorescence other than a serous detachment of dependent retinal detachments into the inferior fundus with
the macula. Beyond 1 month, however, the area of detachment associated atrophic tracts of the RPE.7 Capillary telangiectasis,
showed increased autofluorescence in a diffuse pattern with capillary nonperfusion,46 and secondary neovascularization
some subretinal granular deposits. OCT analysis demonstrated associated with the chronic detachments may occur. Because of
that the hyperautofluorescent areas had material on the outer the widespread pigmentary alterations and chronically reduced
surface of the retina. The thickness of the outer retinal material visual acuity, these patients are sometimes misdiagnosed as having
was proportional to the amount of hyperautofluorescence. We an inherited retinal or macular dystrophy. ICG angiography of
hypothesize that the material on the outer surface of the retina DRPE shows the same type of widespread choroidal vascular
represents accumulated photoreceptor outer segments secondary hyperpermeability as patients with typical CSC have, except the
to lack of direct apposition and phagocytosis by the RPE. Auto- number and area of hyperpermeability seems to be greater in
fluorescence, therefore, is not limited to RPE cells but can include patients with DRPE.
outer retina as well. Our study also indicated that measurement
of autofluorescence of the central macula is highly correlated with AUTOFLUORESCENCE PATTERNS OF DRPE
visual acuity. While a healthy macula has a relatively uniform
distribution of autofluorescence, an unhealthy macula has more Autofluorescence imaging of DRPE patients varies according to
variance related to cell injury and accumulation of abnormal the degree of cellular injury. Von Ruckman and Bird43 concluded
amount of fluorophores. in their study that decreased or absent autofluorescence within
Autofluorescence photography, therefore, provides a noninvasive longstanding lesions indicates reduced metabolic activity of the
1874 assessment of the status of the RPE and outer retina in CSC RPE due to photoreceptor cell loss. A German study44 reported
Central Serous Chorioretinopathy

FIGURE 142.5. (a and b). As CSC becomes


chronic, the hyperfluorescence on fluorescein
angiography loses its pinpoint character and
becomes more diffuse.

a b

that the irregular and increased autofluorescence changes have one to three leaks seen during fluorescein angiography,
observed among their chronic patients is reflective of RPE some patients in an unusually severe variant have numerous

CHAPTER 142
reactive changes. exuberant leaks, multiple PEDs, and bullous retinal detach-
Our study of 58 eyes45 helps explain the varying patterns of ments that extend into the inferior periphery of the fundus.47
hyper- and hypo-autofluorescence seen in chronic CSC. The These detachments are often associated with subretinal
descending atrophic tracts often seen in DRPE have character- fibrinous exudates and shifting subretinal fluid. Several reports
istic patterns. Tracts of more recent origin and the outer edge of of this condition originated in Japan, where this variant seems
more chronic descending tracts exhibit hyperautofluorescence, more common.41,46,48 Bullous serous retinal detachments also
reflecting the increased lipofuscin within the RPE and A2E have been reported in organ transplant patients.49 Patients with
products within the outer retina. Patients with a history of CSC bullous detachment have the same findings during ICG angio-
that has been quiescent for many years have only hypoauto- graphy as the patients with classic CSC and DRPE, except the
fluorescent areas, indicative of the RPE atrophy. Autofluor- number and size of choroidal hyperpermeability areas are greater.
escence photography, therefore, provides a noninvasive tool to An observational case series of 25 patients showed that this
monitor cellular function in CSC patients and indirectly severe variant can occur in both healthy patients (21/25) and
determine visual prognosis (Figs 142.6 and 142.7). patients treated with corticosteroids for metabolic or auto-
immune disease (4/25).50 Bilaterality is the norm (84%). While
BULLOUS DETACHMENT OF THE RETINA some of the patients initially presented with bullous changes,
SECONDARY TO CSC others converted from classic CSC (36%). The conversion time
ranged from 7 months to 9 years after onset. Recurrence rate
There is an additional, but rare, form of CSC that causes
bullous retinal detachments. Although most patients with CSC

FIGURE 142.7. This autofluorescence photograph taken with a


FIGURE 142.6. This autofluorescence photograph taken with a Topcon 50µ camera shows dark hypoautofluorescent descending
Topcon 50µ camera illustrates the diffuse decompensation of the RPE tracts reflecting the atrophic patterns resulting from inferiorly
seen in chronic CSC. gravitating fluid. 1875
RETINA AND VITREOUS

was as high as 52% in this study, but 80.4% recovered central the inability to visualize the external limiting membrane or the
visual acuity of 20/40 or better. Those patients who experienced boundary between photoreceptor bodies and outer segments.
visual loss greater than 20/40 had macular damage.
DIFFERENTIAL DIAGNOSIS
SUBRETINAL DEPOSITS
Although the clinical and fluorescein angiographic features of
Patients with CSC, of any variety, may have deposition of sub- CSC are often classic, several entities should be considered in
retinal material that occurs in four forms:17,51 fibrin (see section the differential diagnosis: CNV, tumors, inflammatory disorders,
on Ocular Findings of Classic CSC), lipid, macrophages, and vascular pathology and rhegmatogenous retinal detachment can
outer photoreceptor segments. Subretinal lipid is usually found sometimes have similar clinical characteristics to CSC. Careful
in chronic CSC in older patients and appears as discrete, hard- biomicroscopy and fundus imaging, however, allows for the
edged, subretinal accumulations typically at the borders of a correct diagnosis.
neurosensory detachment. This older age group is at risk for The principal condition that needs to be differentiated from
CNV, which is a more common disorder causing subretinal lipid. CSC is CNV, particularly occult CNV. The ocular findings of
Because CNV has a different course and treatment, great care CNV share many similarities with those of CSC. Both groups
must be exercised in establishing the diagnosis of CSC in an of patients may have neurosensory detachments, PEDs, mottled
older patient with subretinal lipid. depigmentation, hyperpigmentation, areas of RPE atrophy, and
Diffuse deposition of macrophages and outer photoreceptor subretinal deposits of fibrin and lipid.11 Patients with CNV,
SECTION 10

segments gives the appearance of outer retinal small dots on however, have thickening at the level of the RPE, notched PEDs
ophthalmoscopy. This can be seen in almost every patient with and subretinal or subpigment epithelial blood. These findings
CSC lasting more than a few months. Pronounced accumulation are not seen in CSC. In addition, eyes with CNV generally have
of material may even produce the appearance of diffuse yellow coexistent ocular findings related to the generation of new blood
flecks in some patients. Because of the subretinal deposits, vessel growth. These factors include punched-out chorioretinal
some of these patients have been initially suspected of having scars in ocular histoplasmosis syndrome, lacquer cracks, and
retinitis, choroidal tumors, or CNV. Based on autofluorescence areas of choroidal atrophy in pathological myopia, breaks in
studies,45 the material on the outer surface of the elevated Bruch’s membrane in cases of choroidal rupture, and drusen
retina may perhaps represent an accumulation of photoreceptor and pigmentary clumping in patients with age-related macular
outer segments secondary to lack of phagocytosis, given that the degeneration. The CNV secondary to proximal causes such as
RPE in these cases is not in direct apposition with the retina. chorioretinal scars or choroidal ruptures generally has ‘classic’
Interestingly, the granular dots may vary in number, but do not findings on fluorescein angiography. These cases demonstrate a
vary in size. This uniformity of size and their autofluorescent lacy vascular pattern of hyperfluorescence with increasing
nature suggest that these dots may be macrophages, engorged with leakage and staining throughout a fluorescein angiographic
phagocytized outer segments. The occasional yellow fleck evaluation. Occasionally a specific feeder vessel can be seen
appearance is explained by the lutein component of outer extending from the chorioretinal scar. The fluorescein angio-
photoreceptor segments.52 graphic findings of exudative age-related macular degeneration
may be more difficult to differentiate from CSC because the
OPTICAL COHERENCE TOMOGRAPHY majority of AMD leakage is occult CNV. ICG angiography of
IN CSC CSC demonstrates multifocal, and usually bilateral, choroidal
vascular hyperpermeability that has specific temporal and
OCT provides anatomical information in all types of CSC. A topographical characteristics. The hyperpermeability in CSC is
Spanish study compared conventional OCT findings with most evident in the mid-phases of the angiographic evaluation.
fluorescein angiographic patterns in 39 eyes with CSC.53 The The later phases of ICG angiography show dispersion of the dye
authors report that 36 eyes had an optically empty vaulted area with negative staining of the larger choroidal vessels. CNV, on
under the neurosensory retina consistent with detachment that the other hand, shows a unilateral, unifocal area of hyperfluor-
related to fluorescein-filled areas. Thirty-five eyes had highly escence that usually shows progressively increasing contrast
characteristic small bulges protruding from the RPE, angiogra- with the surrounding choroid in the later phases of the angio-
phically related to leaking spots. Three eyes showed an almost gram. ICG angiography may provide important information to
semicircular space under the RPE, with thinner overlying retina. help rule out the presence of occult CNV. Also, repeating the
Additionally, our group’s study showed that conventional OCT fluorescein angiogram 2–3 weeks later may be helpful as CNV
is useful for detecting subclinical cystoid macular degeneration may become more apparent with time.
or foveal atrophy.54 OCT, therefore, provides a noninvasive imag- Tumors and infiltrative conditions, such as leukemia, amela-
ing test complementary to fluorescein angiography. notic melanoma, or metastatic disease, can also appear similar to
A study of 38 CSC eyes with OCT ophthalmoscope producing CSC. Clinical examination, however, shows that these infiltra-
en face OCT scans (OCT C-scans) provides information beyond tive lesions generally have a different color than the surrounding
conventional OCT.55 In this study, active CSC was associated normal choroid, demonstrate thickening of the choroid by ultra-
with large neurosensory detachment (23/29), subretinal hyper- sonography, and do not have serous PEDs. Inflammatory con-
reflective deposits (20/29), and pigment epithelial detachment ditions with serous retinal detachments can also masquerade as
(15/29). Multiple small PEDs located both within and outside CSC, but clinical diagnostic clues exist. For example, eyes
the neurosensory detachment were detected in one-third of the affected with posterior scleritis or Harada’s disease show signs
patients with either active or inactive CSC. of intraocular inflammation such as iritis or vitritis, have patches
In a retrospective study of unilateral resolved CSC,56 the of yellowish discoloration in the posterior pole, demonstrate
involved eyes had a decrease in central foveal thickness which staining of the optic nerve head during fluorescein angiography,
had a statistically significant correlation with visual acuity. The and have thickening of the choroid by ultrasonography. These
foveal thickness of the eye with resolved CSC was normalized findings are not seen in CSC. Extraocular symptoms, such as
by dividing its thickness by that of the uninvolved eye. Addi- meningeal signs including headache, neck stiffness, and
tional factors on OCT associated with poorer visual acuity were vomiting, are common in Harada’s disease but not in CSC.
1876
Central Serous Chorioretinopathy

Anatomical changes can also complicate the picture. For exam- with CSC led to new theoretical considerations. During ICG
ple, patients with optic nerve pits may have a serous detachment angiography, the choroidal circulation appears to have multi-
of the macula that may appear similar to CSC. Fortunately, the focal areas of hyperpermeability.40,42,60–63 These areas of hyper-
optic nerve pathology is generally readily visible. Also, the macu- permeability may arise from venous congestion. Excessive
lar elevation due to optic nerve pits differs from CSC because it tissue hydrostatic pressure within the choroid from the vascular
is generally a bilayer detachment caused by retinoschisis in the hyperpermeability may lead to PEDs, disruption of the retinal
macula. There are no leaks from the level of the RPE during fluor- pigment epithelial barrier, and abnormal egress of fluid under
escein angiography in patients with optic nerve pits. Another the retina. In past studies, leaks demonstrable at the level of the
anatomical abnormality that can mimic CSC is rhegmatogenous RPE invariably are contiguous with areas of choroidal vascular
retinal detachments. They may cause elevation of the macula like hyperpermeability.40,42,60–63 On the other hand, most areas of
CSC, but differ in that they have an associated retinal hole or tear hyperpermeability are not associated with actual leaks. These
and do not have leaks visible during fluorescein angiography. areas of hyperpermeability without leaks may affect the size,
Vascular disorders are also in the differential diagnosis. shape, and chronicity of any overlying neurosensory detachment
Malignant hypertension can produce a serous retinal detachment. by inducing changes in the ability of the overlying RPE to pump.
The presence of systemic hypertension, Elschnig’s spots, shifting Theoretical considerations about why the choriocapillaris
fluid, and choroidal or retinal vasculature changes, or both, can would develop increased permeability have been described else-
distinguish it from CSC. Serous retinal detachment can occur where. Increased circulating epinephrine and norepinephine
in toxemia of pregnancy. The systemic findings of hypertension, levels have been found in patients with CSC. Administration of

CHAPTER 142
proteinuria, and edema will separate this condition from CSC sympathomimetic compounds has been associated with CSC in
seen in pregnant women. Disseminated intravascular coagulo- humans. A CSC-like condition exists in monkeys given both
pathy should also be considered in the differential diagnosis of sympathomimetic agents and corticosteroids. It is possible to
serous retinal detachment and can be distinguished by its sys- postulate that sympathomimetic compounds or corticosteroids,
temic findings. either endogenous or exogenous, alter the permeability of the
choriocapillaris directly, or through secondary means such as
PATHOPHYSIOLOGY affecting choroidal vasculature autoregulation. Although the rate
of developing CSC with corticosteroid use is not known, CSC is
Multiple theories of pathophysiology exist and continue to a relatively common disease. Clearly, systemic administration
evolve as we understand the disease better. Fluorescein angio- of cortiosteroids can lead to CSC, but local administration does
graphy contributed to the current pathogenesis concept. With not appear to do so with anywhere near the same frequency.
the advent of this retinal imaging test, ophthalmologists had a Although there has been one isolated case report of CSC
more precise method of diagnosing and evaluating CSC. Fluor- following intravitreal triamcinolone acetonide for diabetic
escein angiography demonstrates one or more sites of leakage in macular edema,28 the author has given hundreds of intravitreal
cases of active CSC. With cessation of these leaks, the detach- steroid injections to patients and has not seen one case of CSC.
ment regresses. This suggested, at least to some observers, that Perhaps systemic administration causes particular physiologic
the leak seen during fluorescein angiography represented fluid alterations not as readily induced by local application.
coming from the choroid into the subretinal space through a
precisely located defect in the RPE. The fluorescein, contained HISTOPATHOLOGY OF CSC
in the choroidal fluid, was brought into the subretinal space with
the bulk fluid flow going from the choroid toward the retina. There exists only limited histopathologic information on CSC.
The balance of the tissue oncotic and hydrostatic pressures Neurosensory detachment with subretinal and subpigment
ordinarily causes fluid flow from the retina towards the choroid. epithelial deposition of fibrin has been reported. A model of
In experimental models, injury or destruction of the RPE was exudative detachment has been produced in monkeys with
seen to speed up the resorption of subretinal fluid.57 These repeated injection of corticosteroids and epinephrine.64–67 In
findings suggested that an isolated defect in the RPE could not their monkey model of CSC, Yoshioka and Katsume31 described
account for the findings seen in CSC, but rather that more a focal area of degenerated RPE with adjacent damaged chorio-
diffuse RPE abnormalities would be required to overcome the capillaris endothelial cells. These endothelial abnormalities
RPE pumping function. To clarify the pathophysiology of CSC, were sealed by platelet-fibrin clots. Some dispute the validity of
several newer theories were postulated based in part on findings Yoshioka’s animal model and suggest the monkeys likely had
from animal models. One theory stated that what appeared to accelerated hypertension rather than just simple CSC.
be leaks at the level of the RPE were in fact not necessarily OCT, thanks to its high resolution, can effectively provide
active leaks, but were areas where dye diffused into the sub- optical biopsies. Although OCT has been commonly used to
retinal space.58 The neurosensory detachment was thought to determine the presence of subretinal fluid, it can provide even
be secondary to widespread areas of RPE dysfunction. This theory, more useful information. Retinal atrophy has been seen in some
however, did not clearly elucidate why the areas of RPE dysfunc- patients. In addition, the ability to visualize finer anatomic
tion occurred or why CSC spontaneously improves, as it fre- details such as the external limiting membrane was much less
quently does. This theory also did not explain why patients in patients with lower levels of visual acuity, suggesting that
with CSC frequently develop PEDs, or why laser treatment to a anatomic alterations occur that are associated with decreased
‘leak’ causes a rapid resolution of the neurosensory detachment. acuity.56 Patients with a history of chronic detachment and poor
Another theory suggested that a focus of RPE cells, losing their visual acuity after reattachment may have cystoid spaces within
normal polarity, pumps fluid from a choroid to retina direction, the retina, a condition that has been termed cystoid macular
causing a neurosensory detachment.59 Yet this theory could neither degeneration.54
explain the presence of PEDs and subretinal fibrin nor how a
few RPE cells pumping in the wrong direction could overcome NATURAL COURSE
the pumping ability of broad areas of surrounding RPE cells.
Integration of the clinical findings of CSC with the ICG angio- Most patients with CSC spontaneously resolve and experience
graphic abnormalities of the choroidal circulation in patients an almost complete restoration of vision. In the series of Klein
1877
RETINA AND VITREOUS

and colleagues,68 resolution was noted in all 34 eyes studied When is it appropriate to laser photocoagulate CSC? Because
prospectively without any treatment. The average resolution of the unfavorable risk–benefit ratio, laser photocoagulation
time in this study was 3 months. All patients had visual acuity generally is reserved for patients with the following criteria:
of 20/40 or better, and 94% of the eyes had visual acuity of symptoms greater than 4 months, leakage sites located greater
20/30 or better at follow-up examination (average 23 months). than 375 mm from fixation, a history of CSC in the fellow eye
However, even if patients do recover Snellen visual acuity, with an unfavorable outcome, and the need or desire for treat-
many still complain of decreased color vision, relative scotomas, ment. If the leak is located well away from the central macula,
micropsia, metamorphopsia, decreased contrast sensitivity, and then there is less reason to hesitate about laser photocoagula-
nyctalopia in the affected eye. Some may even notice a slight tion. A detailed biomicroscopic examination and fluorescein
distortion in their central vision. Although these patients have angiogram is essential to monitor for CNV. If laser is initiated,
resolution of their neurosensory detachment, they regain only it is imperative that the patient understands that treatment
part of their central vision because they have suffered photo- only shortens the duration of the disease and does not affect the
receptor damage, atrophy, irregular RPE pigmentation, or final visual acuity or recurrence rate.
subretinal fibrosis.
Recurrence of CSC is a problem, affecting 40–50% of METHODS OF PHOTOCOAGULATION
patients.10,68 Some of these patients will go on to have recurrent
focal leaks while others will inexorably progress to the more Laser photocoagulation should be performed to the leakage site
visually threatening DRPE. Recurrences can occur many years with low-intensity energy. The laser is set for a spot size of
SECTION 10

later. The recurrent leakage point is within 1 mm of the initial 100–200 mm, power of 100–150 mW, with application time of
leakage point in 80% of patients.9,l0 Secondary CNV may occur, 0.1–0.2 s. With a recent angiogram as guidance, the more
particularly in patients over 50 years of age.11 peripheral leaks are treated first. The amount of laser uptake is
affected by several variables. These include the amount of
TREATMENT subretinal fluid present, the degree of pigmentation of the RPE,
which is variably pigmented in areas of chronic subretinal fluid,
Each treatment technique for CSC has been based to a certain the degree of RPE detachment, and the wavelength of laser
extent on proposed mechanisms of pathophysiology at the time. used. The leakage point is treated as well as a small sur-
The resultant treatment approaches for CSC have been varied rounding region of normal RPE. Great care should be taken to
and have usually been examined as part of uncontrolled studies. obtain only a dull gray coagulation to avoid the possibility of
No medical therapy has been shown to be effective for patients secondary CNV.
with CSC. Tranquilizers, sedatives, and barbiturates have been The patient should be monitored carefully to assess for
advocated to decrease the psychogenic component of this dis- recurrence or laser-induced complications. The subretinal fluid
order, but their efficacy has not been demonstrated. Because of generally takes a few weeks to resorb. The visual symptoms start
the suggestion that CSC may be related to abnormal levels of to abate with diminution of the subretinal fluid, but the time it
circulating epinephrine, the use of b-blockers has been suggested takes for the patient to regain final visual acuity seems proportional
as a treatment. A small study suggested a possible benefit,69 but to the amount of time the retina was detached. The initial
the findings have not been confirmed with either a larger study follow-up examinations are to evaluate the patient for CNV.
or a randomized trial. Epinephrine stimulates a and b receptors; If hemorrhage, increased turbidity of the subretinal fluid, or
blocking only b receptors would allow unopposed a stimulation. thickening at the level of the RPE in or adjacent to the area of
This might produce unwanted vascular constriction. If a patient laser treatment is noted, secondary CNV should be suspected.
is on corticosteroids, the medical treatment includes withdrawal The patient should have a repeat fluorescein angiogram at that
of the drug. Ketoconazole has been shown to reduce endogenous point to help in establishing the diagnosis. Secondary CNV
corticosteroid levels and is a theoretical treatment, but no study generally causes a nodular or crescent-shaped area of hyper-
regarding this treatment is published at this time. Currently, no fluorescence under or adjacent to the area of previous laser photo-
randomized controlled study has shown any drug to be useful in coagulation. If the original site of treatment was sufficiently
the treatment of CSC. extrafoveal, it is possible to discover and treat secondary CNV,
in many cases, before the neovascularization extends under the
PHOTOCOAGULATION THERAPY fovea. The CNV may be treated with either thermal laser, if
sufficient room exists, or with photodynamic therapy (PDT).
Laser photocoagulation is the most commonly studied modality
in the treatment of CSC. The principal goal of this treatment is PHOTODYNAMIC THERAPY
to reduce the leakage through the RPE and cause resolution of
the subretinal fluid with improvement in visual acuity. Laser PDT is a newer treatment modality for CSC. While the thera-
photocoagulation to the site of leakage seen during fluorescein peutic mechanism of PDT is known for the CNV in age-related
angiography shortens the duration of macular detachment in macular degeneration, it is not known for CSC. Studies have
patients with typical CSC, but does not appear to affect the final shown that PDT may be useful for treatment of DRPE, and
visual acuity.70–75 Similarly for DRPE, thermal grid laser to an even acute classical CSC in certain circumstances.
area with small leaks appeared to cause a decrease in the DRPE is a challenge to treat because of the wide distribution
amount of subretinal fluid present,76 but did not cause a long- of multiple indistinct leaks. Several groups have investigated
term change in the visual acuity. The effect of laser treatment the use of PDT with verteporfin for more chronic forms of
on the rate of recurrence is inconclusive as it reduced the rate of CSC.78–80 Generally PDT causes the subretinal fluid to decrease
recurrence in some studies,74,75 but not in others.70,71,73 For the or resolve completely. Recurrences of subretinal fluid do occur,
severe bullous variant, laser photocoagulation did not confer but they are responsive to retreatment with PDT. Pigmentary
any significant advantage in terms of temporal resolution of changes persist, however, so earlier treatment is desired.
serous retinal detachment or final visual acuity outcome.77 Our group conducted a study of ICG angiography-guided
Potential dire side effects of photocoagulation include CNV, PDT of 20 chronic CSC eyes in which we found an inverse
scotoma, and RPE scar expansion. correlation between baseline visual acuity at the time of treatment
1878
Central Serous Chorioretinopathy

initiation and improvement of visual acuity afterwards.79 The RPE leaks treated with PDT.81 No areas of hyperpermeability
location of laser light application, in our study, was based on distant from the RPE leak were included in the treatment. All
regions of choroidal vascular hyperpermeability seen during nine eyes showed resolution of both fluorescein leakage and
ICG angiography that were responsible for the fluid leakage into anatomic macular fluid. Visual acuity improved in seven eyes
the macula. In typical clinical practice, conventional fluorescein and remained the same in two eyes. There were no adverse
angiography may be used in lieu of ICG as it provides sufficient events. Interestingly, many of the study cases treated did not
useful information. Safety precautions include avoidance of have ICG. The leak plus 1 mm was treated in these patients
directly treating the central fovea to help reduce the possibility and the clinical results showed resolution of the leakage and
of inducing foveal atrophy with the PDT. Although we used fluid. Therefore, it probably is not necessary to treat the hyper-
the usual dose of verteporfin, it may be possible to reduce the permeable area as delineated by ICG for acute leaks. Although
dosage, possibly resulting in decreased costs. our case series suggests that PDT with verteporfin can lead to
On occasion PDT has been used for typical acute leaks. resolution of focal RPE leaks in CSC, the study is limited by its
Because of the high cost of PDT, its use has typically been small size, retrospective nature, and lack of a control group to
limited in classic CSC to those patients with focal leaks near help ascertain if the resolution was due to the PDT or the
the center of the fovea where laser photocoagulation may induce natural disease course.
excessive harm. Our group conducted a small case series of focal

CHAPTER 142
REFERENCES
1. von Graefe A: Ueber centrale recidivierende 15. Desai UR, Alhalel AA, Campen TJ, et al: acetonide for diabetic macular oedema.
Retinitis. Graefes Arch Clin Exp Ophthalmol Central serous chorioretinopathy in African Acta Ophthalmol Scand 2005; 83:132–133.
1866; 12:211–215. Americans. J Natl Med Assoc 2003; 28. Baumal C, Martidis A, Truong S: Central
2. Horniker E: Su di una forma retinite 95:553–559. serous chorioretinopathy associated with
centrale di origine vasoneurotica (retinite 16. Spitznas M: Central serous retinopathy. In: periocular corticosteroid injection treatment
central capillaro spastica). Ann Ottal 1927; Ryan SJ, ed. Retina. St Louis, MO: CV for HLA-B27-associated iritis. Arch
55:578–600. Mosby; 1989: 217–227. Ophthalmol 2004; 122:926–928.
3. Gifford SR, Marquardt G: Central 17. Gass JDM: Central serous 29. Haimovici R, Rumelt S, Melby J: Endocrine
angiospastic retinopathy. Arch Ophthalmol chorioretinopathy and white subretinal abnormalities in patients with central
1939; 21:211–228. exudation during pregnancy. Arch. serous chorioretinopathy. Ophthalmology
4. Bennett G: Central serous retinopathy. Ophthalmol 1991; 109:677. 2003; 110:698–703.
Br J Ophthalmol 1955; 39:605–618. 18. Karadimas P, Bouzas EA: Glucocorticoid 30. Yoshioka H, Katsume Y, Akune H:
5. Maumenee AE: Symposium: macular use represents a risk factor for central Experimental central serous
diseases, clinical manifestations. Trans Am serous chorioretinopathy: a prospective, chorioretinopathy in monkey eyes:
Acad Ophthalmol Otolaryngol 1965; case-control study. Graefes Arch Clin Exp fluorescein angiographic findings.
69:605–613. Ophthalmol 2004; 122:784–786. Ophthalmologica 1982; 185:168–178.
6. Gass JDM: Pathogenesis of disciform 19. Polak BC, Baarasma GS, Snyers B: Diffuse 31. Yoshioka H, Katsume Y: Experimental
detachment of the neuroepithelium. II. retinal pigment epitheliopathy complicating central serous chorioretinopathy. III.
Idiopathic central serous choroidopathy. systemic corticosteroid treatment. Br J Ultrastructural findings. Jpn J Ophthalmol
Am J Ophthalmol 1967; 63:587–615. Ophthalmol 1995; 79:922–925. 1982; 26(4):397–409.
7. Yannuzzi L, Shakin J, Fisher Y, et al: 20. Gass JD, Little H: Bilateral bullous 32. Michael JC, Pak J, Pulido J, de Venecia G:
Peripheral retinal detachment and retinal exudative retinal detachment complicating Central serous chorioretinopathy
pigment epithelial atrophic tracts idiopathic central serous chorioretinopathy associated with administration of
secondary to central serous pigment during systemic corticosteroid therapy. sympathomimetic agents. Am J
epitheliopathy. Ophthalmology 1984; Ophthalmology 1995; 102:737–747. Ophthalmol 2003; 136:182–185.
91:1554–1572. 21. Gass JDM, Slamovits TL, Fuller DG, et al: 33. Sun J, Tan J, Wang Z, et al: Effect of
8. Spaide RF: Central serous Posterior chorioretinopathy and retinal catecholamine on central serous
chorioretinopathy and other causes of detachment after organ transplantation. chorioretinopathy. J Huazhong Univ Sci
serous detachment of the retina. In: Spaide Arch Ophthalmol 1992; 110:1717–1722. Technolog Med Sci 2003; 23:313–316.
RF, ed. Diseases of the retina and vitreous. 22. Iida T, Spaide RF, Haas A, et al: Leopard- 34. Gelber GS, Schatz H: Loss of vision due to
Philadelphia, PA: W.B. Saunders Co.; 1999. spot pattern of yellowish subretinal central serous chorioretinopathy following
9. Spitznas M, Huke J: Number, shape, and deposits in central serous psychological stress. Am J Psychiatry
topography of leakage points in acute type chorioretinopathy. Arch Ophthalmol 2002; 1987; 144:46–50.
I central serous retinopathy. Graefes Arch 120:37–42. 35. Yannuzzi L: Type A behavior and central
Clin Exp Ophthalmol 1987; 225:438–440. 23. Haimovici R, Gragoudas E, Duker J, et al: serous chorioretinopathy. Trans Am
10. Gilbert CM, Owens SL, Smith PD, Fine SL: Central serous chorioretinopathy Ophthalmol Soc 1986; 84:799–845.
Long-term follow-up of central serous associated with inhaled or intranasal 36. Werry H, Arends C: Untersuchung zur
chorioretinopathy. Br J Ophthalmol 1984; corticosteroids. Ophthalmol 1997; Objektivierung von
68:815–820. 104:1653–1660. Personlichkeitsmerkmalen bei Patienten mit
11. Spaide RF, Campeas L, Haas A, et al: 24. Fernandez C, Mendoza A, Arevola J: Retinopathia centralis serosa. Klin
Central serous chorioretinopathy in younger Central serous chorioretinopathy Monatsbl Augenheilkd 1978; 172:363–370.
and older adults. Ophthalmology 1996; associated with topical dermal 37. Shimizu K, Tobari I: Central serous
103: 2070–2080. corticosteroids. Retina 2004; 24:471–474. retinopathy dynamics of subretinal fluid.
12. Tittl MK, Spaide RF, Wong D, et al: 25. Karadimas P, Kapetanios A, Bouzas E: Mod Probl Ophthalmol 1971; 9:152–157.
Systemic and ocular findings in central Central serous chorioretinopathy after local 38. Shimizu K, Tobari I: Central serous
serous chorioretinopathy. Am J Ophthalmol application of glucocorticoids for skin retinopathy dynamics of subretinal fluid.
1999; 128:63–68. disorders. Arch Ophthalmol 2004; Mod Probl Ophthalmol 1971; 9:152–157.
13. Haimovici R, Koh S, Gagnon DR: Risk 122:784–786. 39. Vukojevic N, Sikic J, Katusic D, et al: Types
factors for central serous chorioretinopathy: 26. Williamson J, Nuki G: Macular lesions during of central serous retinopathy, analysis of
a case-control study. Ophthalmology 2004; systemic therapy with depot tetracosactrim. shape, topographic distribution and
111:244–249. Br J Ophthalmol 1970; 54:405–409. number of leakage sites. Coll Antropol
14. Fine SL, Owens SL: Central serous 27. Imasawa M, Ohshiro T, Gotoh T, et al: 2001; 25(Suppl):83–87.
retinopathy in a 7-year-old girl. Am J Central serous chorioretinopathy following 40. Prunte C, Flammer J: Choroidal capillary
Ophthalmol 1980; 90:871–873. vitrectomy with intravitreal triamcinolone and venous congestion in central serous
1879
RETINA AND VITREOUS

chorioretinopathy. Am J Ophthalmol 1996; serous chorioretinopathy. Retina 2003; 68. Klein ML, van Buskirk EM, Friedman E,
121:26–34. 23:1–7; quiz 137–138. et al: Experience with nontreatment of
41. Okushiba U, Takeda M: Study of choroidal 55. van Velthoven M, Verbraak F, Garcia P, central serous choroidopathy. Arch
vascular lesions in central serous et al: Evaluation of central serous Ophthalmol 1974; 91:247–250.
chorioretinopathy using indocyanine green retinopathy with en face optical coherence 69. Avci R, Deutman AF: Treatment of central
angiography. Nippon Ganka Gakkai Zasshi tomography. Br J Ophthalmol 2005; serous chorioretinopathy with the beta
1997; 101:74–82. 89:1483–1488. receptor blocker metoprolol. Klin Monatsbl
42. Spaide RF, Hall L, Haas A, et al: 56. Eandi CM, Chung JE, Cardillo-Piccolino F, Augenheilkd 1993; 202:199–205.
Indocyanine green videoangiography of et al: Optical coherence tomography in 70. Ficker L, Vafidis G, While A, Leaver P:
central serous chorioretinopathy in older unilateral resolved central serous Long-term follow-up of a prospective trial
adults. Retina 1996; 16:78–80. chorioretinopathy. Retina 2005; of argon laser photocoagulation in the
43. von Ruckmann A, Fitzke F, Fan J, et al: 25:417–421. treatment of central serous retinopathy.
Abnormalities of fundus autofluorescence 57. Negi A, Marmor MF: The resorption of Br J Ophthalmol 1988; 72:829–834.
in central serous retinopthy. Am J subretinal fluid after diffuse damage to the 71. Gilbert CM, Owens SL, Smith PD, Fine SL:
Ophthalmol 2002; 133:780–786. retinal pigment epithelium. Invest Long-term follow-up of central serous
44. Framme C, Walter A, Gabler B, et al: Ophthalmol Vis Sci 1983; 24:1475–1479. chorioretinopathy. Br J Ophthalmol 1984;
Fundus autofluorescence in acute and 58. Marmor MF: New hypotheses on the 68:815–820.
chronic-recurrent central serous pathogenesis and treatment of serous 72. Leaver P, Williams C: Argon laser
chorioretinopathy. Acta Ophthalmol Scand retinal detachment. Graefes Arch Clin Exp photocoagulation in the treatment of
2005; 83:161–167. Ophthalmol 1988; 226:548–552. central serous retinopathy. Br J Ophthalmol
45. Spaide RF, Klancnik JM: Fundus 59. Spitznas M: Pathogenesis of central serous 1979; 63:674–677.
SECTION 10

autofluorescence and central serous retinopathy: a new working hypothesis. 73. Brancato R, Scialdone A, Pece A, et al:
chorioretinopathy. Ophthalmolology 2005; Graefes Arch Clin Exp Ophthalmol 1986; Eight-year follow-up of central serous
112:825–833. 224:321–324. chorioretinopathy with and without laser
46. Akiyama K, Kawamura M, Ogata T, 60. Hayashi K, Hasegawa Y, Tokoro T: treatment. Graefes Arch Clin Exp
Tanaka E: Retinal vascular loss in idiopathic Indocyanine green angiography of central Ophthalmol 1987; 225:166–168.
central serous chorioretinopathy with serous chorioretinopathy. Int. Ophthalmol. 74. Yap EY, Robertson DM: The long-term
bullous retinal detachment. Ophthalmology 1986; 9:37–341. outcome of central serous
1987; 94:1605–1609. 61. Scheider A, Nasemann JE, Lund OE chorioretinopathy. Arch Ophthalmol 1996;
47. Gass JDM: Bullous retinal detachment: an Fluorescein and indocyanine green 114:689–692.
unusual manifestation of idiopathic central angiographies of central serous 75. Burumcek E, Mudum A, Karacorlu S,
serous choroidopathy. Am J Ophthalmol choroidopathy by scanning laser Arslan MO: Laser photocoagulation for
1973; 75:810. ophthalmoscopy. Am J Ophthalmol 1993; persistent central serous chorioretinopathy:
48. Tsukahara I, Uyama M: Central serous 115:50–56. results of long-term follow-up.
choroidopathy with bullous retinal 62. Piccolino FC, Borgia L: Central serous Ophthalmology 1997; 104:616–622.
detachment. Abrecht Von Graefes Arch Klin chorioretinopathy and indocyanine green 76. Yannuzzi LA, Slakter JS, Kaufman SR,
Exp Ophthalmol 1978; 206:169–178. angiography. Retina 1994; 14:231–242. Gupta K: Laser treatment of diffuse retinal
49. Friberg TR, Eller AW: Serous retinal 63. Guyer DR, Yannuzzi LA, Slakter JS, et al: pigment epitheliopathy. Eur J Ophthalmol
detachment resembling central serous Digital indocyanine green videoangiography 1992; 2:103–114.
chorioretinopathy following organ of central serous chorioretinopathy. Arch 77. Sahu DK, Namperumalsamy P, Hilton G,
transplantation. Graefes Arch Clin Exp Ophthalmol 1994; 112:1057–1062. et al: Bullous variant of idiopathic central
Ophthalmol 1990; 228:305–309. 64. Nagayoski K: Experimental study of serous chorioretinopathy. Br J Ophthalmol
50. Otsuka S, Ohba N, Nakao K: A long-term chorioretinopathy by intravenous injection 2000; 84:485–492.
follow-up study of severe variant of central of adrenaline. Acta Soc Ophthalmol Jpn 78. Battaglia Parodi M, Da Pozzo S, Ravalico G:
serous chorioretinopathy. Retina 2002; 1971; 75:1720–1727. Photodynamic therapy in chronic central
22:25–32. 65. Miki T, Sunada I, Higaki T: Studies on serous chorioretinopathy. Retina 2003;
51. Ie D, Yannuzzi LA, Spaide RF, et al: chorioretinitis induced in rabbits by stress 23:235–237.
Subretinal exudative depostis in central (repeated administration of epinephrine). 79. Yannuzzi LA, Slakter JS, Gross NE, et al:
serous chorioretinopathy. Br J Ophthalmol Acta Soc Ophthalmol Jpn 1972; Indocyanine green angiography-guided
1993; 77:349–353. 75:1037–1045. photodynamic therapy for treatment of
52. Rapp LM, Maple SS, Choi JH: Lutein and 66. Yasuzumi T, Miki T, Sugimoto K: Electron chronic central serous chorioretinopathy:
zeaxanthin concentrations in rod outer microscopic studies of epinephrine a pilot study. Retina 2003; 23:288–298.
segment membranes from perifoveal and choroiditis in rabbits. I. Pigment epithelium 80. Cardillo Piccolino F, Eandi CM, Ventre L,
peripheral human retina. Invest Ophthalmol and Bruch’s membrane in the healed stage. et al: Photodynamic therapy for chronic
Vis Sci 2000; 41(5):1200–1209. Acta Soc Ophthalmol Jpn 1974; central serous chorioretinopathy. Retina
53. Montero J, Ruiz-Moreno J: Optical 78:588–598. 2003; 23:752–763.
coherence tomography characterisation of 67. Yoshioka H, Sugita T, Nagayoski K: 81. Ober M, Angelilli A, Do D, et al:
idiopathic central serous chorioretinopathy. Fluorescein angiography findings in Photodynamic therapy for acute central
Br J Ophthalmol 2005; 89:562–564. experimental retinopathy produced by serous chorioretinopathy. Ophthalmology
54. Iida T, Yannuzzi L, Spaide R, et al: Cystoid intravenous adrenaline injection. Folia 2005; 112(12):2088–2094.
macular degeneration in chronic central Ophthalmol Jpn 1970; 21:648–652.

1880
CHAPTER

143 Genetics of Age-related Macular Degeneration


Margaret M. DeAngelis and Fei Ji

INTRODUCTION hence pharmacological targets in preventing or slowing down


progression of disease. This chapter focuses on genetic
Age-related macular degeneration (AMD) is the most common contributions to the etiology of AMD.
form of legal blindness in the United States as well as other
developed countries. The Eye Disease Prevalence Group
estimates that 1.75 million United States citizens have BACKGROUND STUDIES DEMONSTRATING
advanced AMD in at least one eye.1 Approximately 10% of the A GENETIC COMPONENT: TWIN AND
population aged 43 and older is affected with some form of the FAMILIAL AGGREGATION STUDIES
disease and 30% of the population aged 75 and older is
affected.2 As the population lives longer this number will likely AMD is a challenging disease to study from a genetic
increase. It is estimated that 2.95 million individuals in the US perspective because unlike disorders that exhibit Mendelian
will have advanced AMD by the year 2020.1 inheritance patterns (where one gene is responsible for the
Current diagnostic methods focus on the detection of phenotype observed in a given family), complex diseases like
neovascular AMD, since available treatments are directed AMD have the following characteristics. Since the disease is
against this advanced stage of the disease. Although the newest common or highly prevalent (with 1.75 million United States
treatments offer some chance of visual improvement, they citizens having advanced AMD in at least one eye),1 it is likely
require invasive delivery methods and have limited ability to that there is more than one gene or environmental factor in
prevent or reverse vision loss. Assessments of an individual's addition to the interactions of these factors that influence an
risk of developing advanced AMD are based on ocular findings individual's susceptibility to disease. AMD is also a
in those who already have the early stages.3 Methods are yet to heterogeneous disease and its many subtypes (reviewed in other
be refined that determine which individuals are at highest risk chapters in this book) present the possibility that there are
of vision loss due to AMD prior to the development of any signs different disease mechanisms and hence different genes
of the disease. responsible for each subtype.
Previous studies of genetic and epidemiologic factors have Although the causes of AMD remain elusive, there is
not been in agreement as to predictors of AMD. Cigarette evidence demonstrating that genetics plays an important role.
smoking appears to be the only epidemiologic risk factor Table 143.1 summarizes many of the studies that have
generally accepted as being associated with an increased risk for supported a role for genetics in the etiology of AMD. These
AMD,4 while an allelic variant in the complement factor H gene studies consist of data from both familial aggregation and twin
(CFH) appears to be the strongest genetic risk factor.
Specifically, several independent reports have shown the
functional polymorphism Y402H in CFH, where a tyrosine is
substituted by a histidine, to be associated with increased risk TABLE 143.1. Studies Showing Hereditability
of both early5–9 and late stages of AMD (both neovascular and
geographic atrophy).6,7,9–12 These findings further suggest that Type of Study Conclusion Authors
the histidine allele, or disease allele, contributes to almost half Monozygotic Twins shared identical Melrose et al.89,
of all cases of AMD in the population. Although having these twin case health problems and Meyers et al.90,
factors may increase one's risk of disease, there are still many report had similar features Dosso et al.91
individuals who both smoke and have the CFH-associated of AMD in
disease variant but have no signs of AMD. For this reason, the corresponding eyes
magnitude of risk for AMD probably depends on exposure to Twin study Showed higher Klein et al.92,
other environmental and genetic risk factors in combination concordance in Meyers et al.93
with smoking and CFH variation. monozygotic twins than Gottfredsdottir et al.
94
Discovering precisely which genes and environmental factors in dizygotic, confirmed Grizzard et al.
95
that stage and type Seddon et al.
contribute to the pathophysiology of AMD could provide targets 96
of AMD was similar Hammond et al.
that could be modifiable through therapeutic or behavioral between twins 2002,14
intervention, thereby reducing or preventing the incidence of
this disease. Knowing which common variants in a handful of Familial Prevalence of AMD Seddon et al.13,
aggregation was higher among Klaver et al.52,
genes may predict which individuals in the population are at study first-degree relatives Assink et al.97
greatest risk for converting to the more advanced stages of of AMD patients
AMD. These genes in turn could serve as biomarkers and 1881
RETINA AND VITREOUS

studies. Data from the Rotterdam study showed that first- ELOVL4
degree relatives of affected individuals are at 25% greater risk
of developing disease than individuals in the general population Like ABCA4, the protein product of ELOVL4 is involved in lipid
without any affected family members. It has also been shown transport and metabolism. Unlike ABCA4, ELOVL4 is located
that AMD has a tendency to cluster or aggregate within in a region (6q24–q14) previously identified from genome-wide
families.13 Further, Hammond and colleagues14 demonstrated scans to harbor AMD susceptibility genes.37,38 A five base pair
that monozygotic twins with AMD had a higher degree of deletion in this gene has been found to be associated with
concordance (both members having AMD) than dizygotic Stargardt disease 3, which has phenotypic similarity to the dry
twins, 37% compared to 19%, respectively. or atrophic form of AMD. A recent report has suggested that the
M299V variant, where a methionine is substituted by a valine
Key Features in the protein sequence of ELOVL4, could increase risk of
A genetic component is supported by the following: neovascular AMD,10 although an earlier study did not find
• A higher prevalence of AMD is found in identical twins this association for either the early or more advanced stages
compared to fraternal twins of AMD.39 Similarly, a recently published study of a Finnish
• A higher prevalence of AMD is found in first-degree relatives of population also did not find an association between the
AMD patients compared to unrelated individuals from the M299V variant in ELOVL4 and patients with large drusen,
general population neovascular, or atrophic AMD.40 It appears that more studies
may need to be conducted before the contribution of the
SECTION 10

ELOVL4 gene to the etiology of AMD can be confirmed.


JUVENILE FORMS (MENDELIAN) OF
MACULAR DEGENERATION: CANDIDATE Key Features
GENES FOR AMD Of the six genes responsible for juvenile forms of retinal
degeneration (VMD2, TIMP-3, RDS/peripherin, EFEMP1, ABCA4,
Because of evidence suggesting a genetic component, some ELOVL4), only ABCA4 and ELOVL4 possibly influence
genes known to be associated with juvenile-onset forms of susceptibility to AMD. Both ABCA4 and ELOV4L4 function in lipid
retinal degeneration have been studied for their association with metabolism and transport
AMD. No studies to date have found an association between
mutations in vitelliform macular dystrophy 2 (VMD2) (Best
disease),15–19 tissue inhibitor of metalloproteinase–3 (TIMP–3) GENOME-WIDE SCANS
(Sorsby fundus dystrophy),20,21 retinal degeneration, slow
RDS/peripherin (retinitis pigmentosa),22,23 and epidermal A genetic component is further supported by the multiple
growth factor-containing fibulin like extracellular martrix genome-wide scans which have identified similar chromosomal
protein 1 (EFEMP1) (Malattia Leventinese/Doyne honeycomb locations harboring AMD susceptibility genes. The purpose
retinal dystrophy)24–26 and any type of AMD, either early or late of a genome-wide scan is to search the 22 pairs of autosomes
stages. Table 143.2 shows the names of these genes, their (no sex chromosomes) with the goal of identifying regions
genomic locations, the retinal disease associated with the gene significantly linked to the disease under investigation. Although
and the AMD population studied. Table 143.2 also shows that linkage studies are powerful and can uncover novel disease
two genes, the ATP-binding cassette transporter gene, subfamily genes, the significant or linked region could harbor hundreds
A, member 4 (ABCA4) (autosomal recessive Stargardt disease) of these disease loci or genes making the identification of one
and the elongation of very long chain fatty acids gene (ELOVL4) locus a challenge. Another disadvantage of a linkage study is
(Stargardt disease type 3 or autosomal dominant Stargardt that regions that contain genes contributing modest or subtle
disease) are inconsistent with respect to whether or not effects to a given phenotype may be missed.
variations in these genes are associated with AMD. Linkage refers to the physical proximity of the disease locus
with the marker locus. The degree of linkage is determined by
ABCA4 the logarithm of the odds ratio (lod score). A lod score of
3 translates to 1000 to 1 odds in favor of linkage (P = 0.0001)
ABCA4 is located on the short arm of chromosome 1 (1p22.1–p21) and a lod score of –3 is 1000 to 1 against the genotype being
and functions in lipid metabolism and transport. Mutations in linked to the disease.
ABCA4, also known as ABCR, cause Stargardt disease, a For complex diseases such as AMD generally no assumption
juvenile form of macular degeneration characterized by deposits regarding the mode of inheritance is made a priority – this is
of lipofuscin in and beneath the retinal pigment epithelium. In called model-free linkage analysis. Model-free linkage analysis
1997, Allikmets et al found an association of heterozygous employs statistical tests that calculate the number of alleles
ABCA4 alleles with AMD.27 In particular, G1961E (glycine is shared between related individuals with and without disease.
substituted by glutamic acid) and D2177N (aspartic acid is Markers called microsatellites or single nucleotide poly-
substituted by asparagine), the two most common variants, morphisms (SNPs) can be used for this purpose. A
along with rare variants in ABCA4 were reported to be microsatellite is a tandemly repetitive unit of base pairs in
associated with ~4% of AMD cases (mostly the dry form). DNA. The repetitive units usually consist of 2, 3, or 4 base
Further, in a larger screen of more than 1200 patients with both pairs and it is the number of these repetitive units that varies
dry and neovascular AMD, Allikmets et al in 2000 reported that between individuals. Because of the high variation or
ABCA4 variants could probably explain up to 8% of AMD.28 heterozygosity in the number of these repetitive units for a
However, several groups have tried to replicate these findings given microsatellite between individuals in a population, they
and were unable to conclude that variants in ABCA4 were are very informative for linkage studies as they are highly
associated with the dry or more advanced stages of AMD.29–36 It polymorphic. The disadvantage is that microsatellites are not
could be that if variation in ABCA4 increases susceptibility to evenly distributed throughout the genome. An SNP is a single
AMD, the risk of disease would only be small or modest, or base change, located every 1000–3000 base pairs in DNA
there exists multiple susceptibility genes for AMD, not sequence and therefore they are abundant in the genome.
1882 necessarily expressed in every patient with AMD. Over the last few years the discovery rate of SNPs throughout
Genetics of Age-related Macular Degeneration

TABLE 143.2. Candidate Genes for AMD Derived from Juvenile Forms of Hereditary Retinal Degenerations

Gene Name Symbol Location Function Disease Association Study Population Author

Retinal RDS 6p21.2– Protein binding Retinitis pigmentosa 56 y.o. index Kemp et al.22
Degeneration, p12.3 patient w/AMD
Slow & his 41 y.o. niece,
42 y.o. index
patient w/RP
& her 15 y.o. son
3 unrelated families, Gorin et al.23
12 patients w/AMD
or peripheral retinal
degeneration, 100
related controls
Tissue TIMP3 22q12.3 Metalloendopeptidase Sorsby's fundus 68 sibpairs w/AMD De La Paz et al.20
Inhibitor of inhibitor activity dystrophy (by AREDs), mean
metallopeptidase-3 age 71.5 ±
10.2 years, 79 age,

CHAPTER 143
gender and ethnically
matched controls
143 German AMD Felbor et al.21
patients (age 48–91),
74 w/other macular
dystrophies
ATP-Binding ABCA4 1p22.1 – ATP binding, ATPase Stargardt 167 unrelated Allikmets et al.27*
Cassette, p21 activity, lipid disease patients w/drusen,
sub-family A transport GA, RPE
(ABC1), & metabolism detachment and/or
member 4 CNV, 220 racially
matched controls
182 patients w/AMD Stone et al.29
(60% w/CNV in at
least one eye),
96 unrelated
subjects expected
to develop AMD
212 patients De La Paz et al.30
w/intermediate
or large soft drusen,
RPE detachments,
GA and/or CNV
(159 familial, 53
sporadic), 56 racially
matched controls
52 unrelated French Souied et al.31
patients w/CNV, 90
French unrelated
self-reported
unaffecteds as
controls
1218 unrelated Allikmets et al.28*
patients w/dry
AMD or CNV from
North America and
Western Europe,
1258 controls
100 patients Rivera et al.32
w/GA, 100
patients w/CNV,
153 unaffecteds age
and geography-
matched controls
544 AMD patients Guymer et al.33
(mostly CNV)
from US, Australia
& Switzerland,
689 controls from
the same areas,
62 unaffected
Somalians 1883
Continued
RETINA AND VITREOUS

TABLE 143.2. Candidate Genes for AMD Derived from Juvenile Forms of Hereditary Retinal Degenerations (Cont’d)

Gene Name Symbol Location Function Disease Association Study Population Author

182 patients Webster et al.34


w/AMD
(Int. ARM Epi.
Study Class.
System),
96 controls from
the same clinic
population as
AMD patients
26 AMD patients Bernstein et al.35
≥ grade 2 (AREDS
scale) from
15 families,
33 unaffected
siblings
SECTION 10

165 AMD patients Schmidt et al36


≥ grade 3 (Int. ARM
Study Class. System)
from 70 families,
33 unaffected
siblings, 59 unrelated
controls
Epidemial EFEMP1 2p16 Maintenance of Malattia Leventinese 494 AMD patients, Stone et al.24
Growth Factor – extracellular matrix (ML)/Doyne 56 ML/DHRD
containing integrity Honeycomb patients, 477
fibulin-like Retinal Dystrophy controls
extracellular (DHRD)
matrix protein 1
13 index patients Guymer et al.25
w/onset drusen,
15 family members,
54 familial cases of
AMD, 150 age and
ethnicity-matched
controls, all at least
50 y.o.
54 patients w/early Narendran et al.26
onset drusen, 114
unrelated controls
≥ 60 y.o.
Vitelliform VMD2 11q13 Chloride ion binding Best Disease 259 unrelated Allikmets et al.15
Macular & ion channel patients w/drusen,
Dystrophy 2 activity GA, RPE detatchment
(Best disease, and/or CNV, 30
bestrophin) patients w/other
disorders (1 w/Best)
41 unrelated Kramer et al.16
patients w/Best's
Disease, 200 AMD
patients,
140 unaffected
controls
321 patients Lotery et al.17
w/drusen and/or
atrophy of RPE,
96 unrelated
patients w/Best
disease,
192 controls
at least 40 y.o.
85 Japanese AMD Akimoto et al.18
patients, 105 age-
matched Japanese
controls

Continued

1884
Genetics of Age-related Macular Degeneration

TABLE 143.2. Candidate Genes for AMD Derived from Juvenile Forms of Hereditary Retinal Degenerations (Cont’d)

Gene Name Symbol Location Function Disease Association Study Population Author

259 AMD patients, Seddon et al.19


28 patients
w/maculopathies
other than Best's
Elongation of ELOVL4 6q14 Lipid metabolism & Stargardt Disease, 778 AMD patients Ayyagari et al.39
very long chain transport type III (Int. Class. System),
fatty acids 551 age and race-
matched controls
992 Caucasian Conley et al.10*
patients w/extensive
drusen, pigmentary
changes, GA and/or
CNV, 120 unrelated
controls

CHAPTER 143
335 Finnish patients Seitsonen et al.40
w/more than 5 small
drusen (< 63µm),
at least 10 large
drusen (> 125µm),
GA, or CNV, of which
154 were sporadic
cases & 181 familial,
105 controls
* Denotes studies finding positive associations.
RP – Retinis Pigmentosa; y.o. – years old; AREDS – age-related eye disease study; GA – geographic atrophy; RPE – retinal pigment epithelial;
CNV – choroidal neovascularization; Int. ARM Epi. Study Class. System – International Age-Related Maculopathy Epidemiology Study Classification System;
Int. Class. System – International Classification System; ML – Malattia Leventinese; DHRD – Doyne Honeycomb Retinal Dystrophy.

the genome has accelerated, thus making them useful for as the AMD population studied. A summary of chromosomal
genome-wide scans; however, they are not as polymorphic regions found by more than one genome-wide scan to be
and many more are needed for linkage analysis. Almost all significantly linked to AMD is shown in Table 143.3 and
of the genome-wide scans conducted to date are linkage includes 1q23.3–q31.1,6,41–46 2p25.3–p12,46,47 6q14,37,38
studies using microsatellite markers, except Klein's scan which 9q31,46,4710q26,42–46,48,49 12q23–q24.31,37,4615q13–q15,37,46
is an association study employing SNPs in the identification of 16p12–p12.1,37,38,46 17q25,42,43 and 22q12.1.45,47 A recent
CFH (Fig. 143.1 and Table 143.3). pooled analysis of several of these genome-wide scans that used
Table 143.3 illustrates results from the AMD genome-wide linkage analysis and examined all types of AMD confirmed
scans to date including regions found to be significant, micro- two of the most consistently reported chromosomal regions
satellite markers used to identify the region, microsatellite (1q23.3–q31.1 and 10q26) while identifying others that were
markers/SNPs, chromosomal band, LOD score, P value, as well not initially significant in the separate studies alone.50 This
pooled analysis or meta-analysis also confirmed two regions
that had been reported to be significant by only individual
studies: 3p14.1–p25.350,51 and 4q32.44,50 Many of the AMD
Genome-wide Scans genome-wide scan studies illustrated in Table 143.3 have
6 resulted in the identification of various chromosomal regions
which have not been replicated by other studies. Differences
5 across studies in definition and measurement of AMD type and
grade coupled with simultaneous analysis of different subtypes
Number of Studies

4 within study populations may make it difficult to compare and


interpret findings. For example, sibling pairs with advanced
3 AMD examined from the family age-related maculopathy study
(FARMS) (Wisconsin grading scale) showed significance to
2 5q33.3, 14q32.33, 16p13.13, 19q13.31, 21q21.2,51 while other
investigators who also studied populations characterized by
1
advanced AMD did not report significant linkage to these same
regions.42–44,46 Moreover, another group's finding of a significant
association between neovascular AMD and 16p12.1 was depend-
0
1998 1999 2000 2001 2002 2003 2004 2005 2006 ent on factors such as body mass index (BMI) and hypertension,38
Year of Study
although the 16p12 region has been shown by other studies to
FIGURE 143.1. The number of genome wide scans for AMD be significantly associated with advanced AMD independent of
performed each year since the first, Klein et al, in 1998. Note that these factors.46 In summary, genome-wide scans have helped to
none were completed in 1999 nor have there been any thus far in identify candidate genes for AMD as well as to highlight the
2006. need for further evaluation of candidate regions. 1885
RETINA AND VITREOUS

TABLE 143.3. Summary of genes uncoved by genome wide scans and candidate gene studies for AMD

Author Region Band LOD P value AMD population Grading scale


41
Klein et al. D1S466 – 1q31 3.00† 0.0001 21 individuals from a family Wis
D1S413 w/ ARMD
Weeks et al.42 D1S1660 1q31 2.46 0.0004 630 individuals w/ either GA modified
and/or CNV from 364 families
Majewski et al.44 D1S518 1q31 2.07 0.001§ 70 families w/ 258 individuals None
w/ advanced AMD, stratified
according to age and dry
vs. wet
Seddon D1S1589 1q24 1.33 0.0070 490 affected individuals AREDS
et al.45 ≥ grade 3, 608 relative pairs
Abecasis D1S549 1q41 1.55 0.0040 113 families, 331 individuals Intnl ARM Epid
et al.47 w/ any type of AMD
Iyengar et al. D1S202 1q31 1.02† 0.0149 297 individuals w/ CNV Wis
SECTION 10

or GA from 34 families,
349 sib pairs, corrected
for age
Weeks et al.43 D1S1660 1q31 2.72 0.0610 1274 individuals w/ large modified
drusen, either GA and/or
CNV from 530 families
Klein et al.6 rs380390 1q31 >5.5† 0.00000004 96 individuals w/ GA and/or AREDS
rs1329428 4.77† 0.00000140 CNV, 50 controls, all from
AREDS
Fisher 2005 D1S202 – 1q31.1 – 0.90† 0.0209 Meta-analysis of Abecasis, n/a
D1S425 q32 Iyengar, Majewski, Schick,
Seddon and Weeks
Fisher 2005 D1S2705 – 1q23.3 – 1.16† 0.0104 Meta-analysis of Abecasis, n/a
D1S202 q31.1 Iyengar, Majewski, Schick,
Seddon and Weeks
Summary region 1q23.3 – q31.1
45
Seddon et al. D2S1391 2q33 1.81 0.0020 490 affected individuals AREDS
≥ grade 3, 608 relative pairs
Abecasis et al.47 D2S1780 2p25.3 1.84 0.0020 113 families, 331 individuals Intnl ARM Epid
w/ any type of AMD (69%
w/ GA and/or CNV) - only
CNV patients
Iyengar et al. D2S1356 2p21 1.52† 0.0041 297 individuals w/ CNV or GA Wis
2004 from 34 families, 349 sib pairs,
corrected for age
Fisher 2005 D2S297 – 2p25.1 – 0.76† 0.0308 Meta-analysis of Abecasis, n/a
D2S2312 p23.2 Iyengar, Majewski, Schick,
Seddon and Weeks
Fisher 2005 D2S2312 – 2p23.2 – 0.84† 0.0244 Meta-analysis of Abecasis, n/a
D2S2251 p16.2 Iyengar, Majewski, Schick,
Seddon and Weeks
Fisher 2005 D2S2251 – 2p16.2 – 0.59† 0.0495 Meta-analysis of Abecasis, n/a
D2S139 p12 Iyengar, Majewski, Schick,
Seddon and Weeks
Summary 2p25.3 –
region p12
Majewski D3S1304 3p13 2.19 0.0007 70 families w/ 258 individuals None
et al.44 w/ advanced AMD, samples
limited to individuals w/
predominantly dry AMD
Schick et al.37 ^ D3S1763 3q26.1 1.25† 0.0081 Beaver Dam Eye Study Wis
(105 families w/ 258
individuals having scores
≥ 4.5 - No late AMD),
adjusted for age

Continued
1886
Genetics of Age-related Macular Degeneration

TABLE 143.3. Summary of genes uncoved by genome wide scans and candidate gene studies for AMD (Cont’d)

Author Region Band LOD P value AMD population Grading scale

Summary region 6q14


Weeks et al.48 D9S925 – 9q21–q22 1.87 0.0017 212 individuals w/ either GA modified
D9S301* – and/or CNV from 225 families
D9S1120
Weeks et al.42 D9S1118 9p13 1.79 0.0020 630 individuals w/ either GA modified
and/or CNV from 364 families
Majewski et al.44 D9S934 9q33 2.07 0.0010 70 families w/ 258 individuals None
w/ advanced AMD, stratified
according to age and dry vs. wet
Abecasis et al.47 D9S938 9q31.1 1.78 0.0020 113 families, 331 individuals Intnl ARM Epid
w/ any type of AMD
Iyengar D9S1871 9p24 1.46† 0.0048 297 individuals w/ advanced Wis
et al. 2004 AMD from 34 families,

CHAPTER 143
349 sib pairs, corrected for age
Iyengar D9S938 9q31 1.26† 0.0079 297 individuals w/ advanced Wis
et al. 2004 AMD from 34 families,
349 sib pairs, corrected for age
Summary Region 9q31
48
Weeks et al. D10S1230 10q26 1.42 0.0053 212 individuals w/ either modified
GA and/or CNV from
225 families
Weeks et al.42 D10S1230 10q26 2.00 0.0400 630 individuals w/ either GA modified
and/or CNV from 364 families
Majewski et al.44 D10S1230 10q26 3.06 0.00009 70 families w/ 258 individuals None
w/ advanced AMD, stratified
according to age and dry vs. wet
Seddon et al.45 D10S1222 10q26 1.61 0.0030 490 affected individuals AREDS
≥ grade 3, 608 relative pairs
Iyengar et al. D10S1248 10q26 1.55† 0.0037 297 individuals w/ advanced Wis
2004 AMD from 34 families,
349 sib pairs, corrected for age
Kenealy et al.49 D10S1230 10q26 1.52 133 affected sib pairs modified
(graded 3, 4, or 5)
Weeks et al.43 D10S1230 10q26 1.91 0.0600 1274 individuals w/ either modified
GA and/or CNV from
530 families
Fisher 2005 D10S1690 – 10q23.33 – 1.39† 0.0057 Meta-analysis of Abecasis, n/a
D10S1483 q26.13 Iyengar, Majewski, Schick,
Seddon and Weeks
Fisher 2005 D10S1483 – 10q24.31 – 2.63† 0.0003 Meta-analysis of Abecasis, n/a
qter q26.13 Iyengar, Majewski, Schick,
Seddon and Weeks
Summary region 10q26
Fisher 2005 D12S318 – 12q23.2 – 0.76† 0.0305 Meta-analysis of Abecasis, n/a
D12S1349 q24.31 Iyengar, Majewski, Schick,
Seddon and Weeks
Schick et al.37 D12S1300 12q23– 1.35† 0.0063 Beaver Dam Eye Study Wis
12q24 (105 families w/ 258
individuals having scores
≥ 4.5 - No late AMD)
Schick et al.37 D12S346 12q22– 1.53† 0.0040 Beaver Dam Eye Study Wis
12q23 (105 families w/ 258 individuals
having scores ≥ 4.5 - No
late AMD)
Iyengar et al. D12S2078 12q23 1.88† 0.0016 297 individuals w/ CNV or Wis
2004 GA from 34 families, 349 sib
pairs, corrected for age
Iyengar et al. D12S297 12q13 1.33† 0.0066 297 individuals w/ CNV or GA Wis
2004 from 34 families, 349 sib pairs,
corrected for age
1887
Continued
RETINA AND VITREOUS

TABLE 143.3. Summary of genes uncoved by genome wide scans and candidate gene studies for AMD (Cont’d)

Author Region Band LOD P value AMD population Grading scale

Summary region 12q23


– q24.31
Schmidt et al.38 D14S608, 14q13 3.20 0.0020 62 families: 2+ sampled relatives Wis
D14S599 w/ early or late AMD in 28
families w/ lower-than-average
IOP values
Jun et al.51 D14S1007 14q32.33 1.97† 0.0013 346 sib pairs from FARMS Wis
w/ advanced AMD
Summary: No overlapping regions found
Schick et al.37 ^ D15S659 15q11.1– 0.60† 0.0479 Beaver Dam Eye Study Wis
15q14 (105 families w/ 258 individuals
having scores ≥ 4.5 - No
late AMD)
SECTION 10

Schick et al.37 ^ D15S816 15q25– 0.63† 0.0447 Beaver Dam Eye Study Wis
15q26 (105 families w/ 258 individuals
having scores ≥ 4.5 - No
late AMD)
Iyengar et al. GATA50C03 15q14 3.94† 0.00001 297 individuals w/ advanced Wis
2004 AMD from 34 families,
349 sib pairs, corrected for age
Iyengar et al. D15S1012 15q13–15q15 5.34† 0.00000035 297 individuals w/ advanced Wis
2004 AMD from 34 families,
349 sib pairs, corrected for age
Summary region 15q13 – q15
Schick et al.37 ^ D16S769 16p12.1 1.23† 0.0086 Beaver Dam Eye Study Wis
(105 families w/ 258 individuals
having scores ≥ 4.5 - No
late AMD)
Iyengar et al. D16S769 16p12.1 1.10† 0.0120 297 individuals w/ advanced Wis
2004 AMD from 34 families,
349 sib pairs, corrected for age
Schmidt et al.38 D16S403 16p12 2.90 0.0400 32 families (late AMD) w/ higher Wis
than average BMI, SBP & IOP
values
Fisher 2005 D16S3103 –≠ 16p13 – 0.92† 0.0195 Meta-analysis of Abecasis, n/a
D16S415 q12.2 Iyengar, Majewski, Schick,
Seddon and Weeks
Fisher 2005 D16S415 – 16q12.2 – 0.94† 0.0187 Meta-analysis of Abecasis, n/a
D16S516 q23.1 Iyengar, Majewski, Schick,
Seddon and Weeks
Jun et al.51 * D16S2616 16p13.13 1.91† 0.0015 346 sib pairs from FARMS Wis
w/ advanced AMD
Summary region 16p12 – p12.1
42
Weeks et al. D17S928 17q25 3.53 0.0070 630 individuals w/ either GA modified
and/or CNV from 364 families
D17S928 17q25 1.56 0.0037 1274 individuals w/ either GA modified
and/or CNV from 530 families
Summary region 17q25
Iyengar et al. GATA178F11 18p11 1.60† 0.0033 297 individuals w/ advanced Wis
2004 AMD from 34 families, 349 sib
pairs, corrected for age
Jun et al.*51 D19S245 19q13.31 2.35† 0.0005 225 sib pairs from BDES & 346 Wis
from FARMS w/ advanced AMD
Iyengar et al. D20S451 20q13 1.42† 0.0052 297 individuals w/ advanced Wis
2004 AMD from 34 families,
349 sib pairs, corrected for age
Jun et al.51 * D21S2052 21q21.2 1.31† 0.007 346 sib pairs from FARMS Wis
w/ advanced AMD

1888 Continued
Genetics of Age-related Macular Degeneration

TABLE 143.3. Summary of genes uncoved by genome wide scans and candidate gene studies for AMD (Cont’d)

Author Region Band LOD P value AMD population Grading scale


45
Seddon et al. D22S1045 22q12 – q13 2.00 0.0010 490 affected individuals AREDS
≥ grade 3, 608 relative pairs
Abecasis et al.47 AGAT055Z 22q12.1 2.55 0.0003 113 families, 331 individuals Intnl ARM
w/ any type of AMD Epdiem
(69% w/ GA and/or CNV) – only
CNV patients
Summary region 22q12 – q13
Note: All maps Marshfield unless * = not indicated and ^ = Harvard Partners, Marshfield and Weizmann.
§ indicates P value estimated by LOD score reported, † indicates LOD score estimated by P value reported.
ARMD - Age Related Macular Degeneration, Wis - Wisconsin, GA - Geographic Atrophy, CNV - Choroidal Neovascularization, AMD - Age-related Macular Degeneration,
AREDS - Age Related Eye Disease Study, Intnl ARM Epid - International Age Related Maculopathy Epidemiology, BDES - Beaver Dam Eye Study, FARMS - Family Age
Related Maculopathy Study, IOP - Intaocular Pressure, BMI - Body Mass Index, SBP - Systolic Blood Pressure, n/a - non applicable.

CHAPTER 143
by the Rotterdam group from the Netherlands in 1998 to be
Key Features associated with a 'decreased' risk of AMD.52 A trend toward
Genome-wide scans can be an important tool in finding genes increased risk of development of AMD was observed with the
associated with disease. Genome-wide scans applying either E2 allele but this finding was not statistically significant. This
linkage or association methods are an important approach in study further demonstrated histologically the presence of APOE
finding genes associated with disease. Because AMD is in large soft drusen from eyes of patients with AMD as well as
a multifactorial disease with both genetic and environmental the absence of this protein from small hard drusen. Many other
factors influencing susceptibility, genes with modest or subtle studies have supported the finding that the E4 allele is
effects may go undetected by linkage studies, yet may be associated with a decreased risk of either the early form or more
uncovered by an association study. For this reason, it is important advanced stages of AMD.53–58 Although many of these studies
to have multiple approaches in elucidating the genetic etiology of a have shown a trend toward an association of the E2 allele with
complex or multifactorial phenotype such as AMD. In genome- increased risk of AMD,52,54,56 this finding did not reach
wide scans, there is the problem of multiple testing issue, which statistical significance with the exception of Baird et al who in
can lead to false positive results. The point-wise significance level 2004 showed that the presence of the E2 allele was statistically
for linkage analysis (lod = 3, P = 0.0001) and association analysis significantly associated with an earlier onset of AMD in women
( P = 0.05) need to be set to more stringent values to obtain a with the neovascular form of AMD.58 Conversely, Pang et al,59
reasonable family-wise significance. Schmidt et al,60 Schultz et al,61 and Gotoh et al62 found no
evidence for either a protective effect of APOE E4 or a causal
effect of APOE E2 on AMD risk. Further supporting the findings
CANDIDATE GENES of a lack of association of APOE and AMD is that of the many
genome-wide scans conducted, none has shown the 19q13.2
Based on the evidence for a genetic component, a variety of region to be linked to AMD. Moreover, a study that performed
candidate genes have been evaluated either for their roles in a genome-wide scan on 34 families with advanced stages of AMD
diseases that share phenotypic similarity to AMD and/or because comprising 346 sibling pairs showed linkage to 19q13.31,51 a
they were identified in genome-wide scans that searched for region in close proximity to APOE, and concluded that the
areas of the genome harboring AMD susceptibility genes as signal was not due to the effect of the APOE gene. Specifically,
described earlier in this chapter. The comprehensive discussion this group investigated all six APOE genotypes in conjunction
that follows includes the genes that have been found by at least with the linkage findings from the FARMS cohort. This resulted
one study to be statistically associated with either increased or in diminished significance of the linkage peak on 19.31,
decreased risk of AMD. Table 143.4 summarizes these findings. suggesting that a gene in this region, other than APOE, may be
responsible for risk of AMD. Furthermore, in a study that
examined the effects of APOE genotypes along with smoking,
APOLIPOPROTEIN GENE (APOE) APOE genotype alone was not found to influence risk of
Like ELOVL4, APOE functions in lipid transport and meta- AMD;60 however, any association observed between APOE and
bolism. APOE was the first candidate gene investigated for a AMD was dependent on smoking history. For example, risk of
role in AMD outside of the genes responsible for the juvenile neovascular AMD increased if an individual was a carrier of the
forms of macular degenerations. The role of apolipoprotein gene E2 allele and had a history of smoking compared to individuals
(APOE) in susceptibility to Alzheimer's disease is well estab- who had the E3/E3 genotype and never smoked.60 This finding
lished, i.e., APOE is a component of the amyloid plaques that may suggest that if APOE is involved in AMD pathogenesis, it
are a hallmark of this aging degenerative disease of the central may function as a weak modifier on risk for either early or more
nervous system. Three alleles E2, E3, E4 (E3 is the most advanced stages of AMD.
frequent allele in the general population) in the last exon, exon
4, of APOE influence disease risk for Alzheimer's. The combi-
nation of these three alleles makes up six genotypes: E2/E2, PAROXONASE 1 GENE (PON-1)
E2/E3, E2/E4, E3/E3, E3/E4, and E4/E4. The E4 allele, and in Paroxonase 1 gene (PON-1) like APOE encodes for a protein
particular the E4/E4 genotype, is thought to be associated that regulates both lipid metabolism and transport. The three
with increased risk of an earlier onset of Alzheimer's disease studies to date that have been published are not in agreement
while the E2 allele is thought to be protective for development as to the contribution of PON1 to the etiology of AMD.
of Alzheimer's disease. The E4 allele of APOE was first reported Specifically the M55L (a methionine is substituted by leucine) 1889
RETINA AND VITREOUS

TABLE 143.4. Candidate Genes for AMD

Previously
Identified
Gene name Symbol Location Function Region? Study Population Study

APOlipoprotein E APOE 19q13.2 Lipid 88 AMD patients (Int. Class. Klaver et al.52
metabolism System), 901 controls from
and transport the Netherlands ≥ 55 y.o.
116 unrelated French Souied et al.53
patients w/CNV, 168 age
and sex-matched controls
≥ 60 y.o.
39 Chinese patients w/ Pang et al.59*
disciform scarring or CNV,
133 normal controls
230 patients ≥ grade 3 by Int. Schmidt et al.55*
Class. System - 61
independent familial cases,
SECTION 10

101 sporadic cases &


68 familial cases used as
sporadic, 333 spouses
used as controls
87 Italian patients w/AMD Simonelli et al.54
(Int. ARM Epi. Study),
47 age-matched controls,
1287 individuals from
the general population
Pooled analysis of Schmidt et al.56
4 independent case-control
data sets – 377 patients
w/ any extensive,
intermediate or large soft
drusen, GA or CNV,
198 ethnically-matched
controls of similar age
259 affected in 56 families Schultz et al.61*
w/AMD, 207 unaffected
from these families,
104 unrelated affected,
88 unrelated controls
(AMD = GA, CNV and/or
extensive drusen)
322 Anglo-Celtic patients Baird et al.64
w/drusen > 63µm, CNV or
dry AMD, 123 unrelated
controls matched for
ethnicity
85 unrelated Japanese Gotoh et al.83*
patients w/CNV,
82 unrelated Japanese
controls ≥ 50 y.o.
632 white unrelated patients Zareparsi et al.57
w/GA, CNV, coarse RPE
changes or large drusen,
206 controls
992 Caucasian patients Conley et al.10*
w/extensive drusen,
pigmentary changes,
GA and/or CNV,
120 unrelated controls
377 unrelated patients Schmidt et al.60*
w/extensive intermediate
or soft drusen, GA and/or
CNV (221 ever smokers),
198 unrelated controls
(90 ever smokers)

Continued
1890
Genetics of Age-related Macular Degeneration

TABLE 143.4. Candidate Genes for AMD (Cont’d)

Previously
Identified
Gene name Symbol Location Function Region? Study Population Study

95 patients w/white Asensio-Sanchez


drusen with or without et al. 2006
pigmentation, CNV or
geographic AMD,
65 spouses as controls
160 patients w/GA Bojanowski et al.
and/or CNV, 227 controls – 2006
133 screened unaffecteds,
94 volunteers with a
younger mean age
SuperOxide SOD2 6q25.3 Regulator 102 Japanese patients Kimura et al.67
Dismutase 2, of oxidative w/CNV, 200 controls
mitochondrial stress

CHAPTER 143
94 patients from Northern Esfandiary et al.65*
Ireland w/CNV, 95 controls
ParaoxONase 1 PON1 7q21.3 Lipid 72 unrelated Japanese Ikeda et al.63
metabolism patients w/CNV, 140 age
and transport and sex-matched controls
62 Anglo-Celtic patients Baird et al.64*
w/ late AMD (Int. ARM Epi.
Study), 115 controls
matched for age and
ethnicity
94 patients from Northern Esfandiary et al.65*
Ireland w/CNV, 95 controls
CySTatin C CST3 20p11.21 Protease 167 German patients w/ CNV Zurdel et al.68
inhibitor (Int. ARM Epi. Study),
517 unrelated controls from
Germany, Switzerland,
Italy & USA
Angiotensin I ACE 17q23.3 Regulator of 173 patients w/CNV or Hamdi et al.69
Converting systemic extensive small (< 63 µm) or
Enzyme 1 blood intermediate (>125 µm)
pressure drusen, 189 age-matched
regulation controls
992 Caucasian patients Conley et al.10*
w/extensive drusen,
pigmentary changes,
GA and/or CNV,
120 unrelated controls
Hemicentin-1 HMCN1 1q25.3 – Maintenance of Klein 1998, 100 families w/3 or more Schultz et al.61
q31.1 extracellular Weeks 2001, living AMD patients,
matrix Majewski 2003, 188 sporadic cases of AMD
integrity Seddon 2003 (Wisconsin Grading Scale),
& Iyengar 2004 174 controls matched for
age, sex and ethnicity
368 patients ≥ grade 2 Hayashi et al.77*
(Int. Class. System), equal
matched unaffected
controls
508 patients ≥ Grade 3 McKay et al.78*
(Rotterdam Study class),
25 possibly affected
and 163 controls
992 Caucasian patients Conley et al.10*
w/extensive drusen,
pigmentary changes,
GA and/or CNV,
120 unrelated controls

Continued

1891
RETINA AND VITREOUS

TABLE 143.4. Candidate Genes for AMD (Cont’d)

Previously
Identified
Gene name Symbol Location Function Region? Study Population Study

335 Finnish patients w/large Seitsonen et al.40*


drusen, GA, or CNV,
of which 154 were sporadic
cases & 181 familial,
105 controls
Chemokine CX3CR1 3p21.3 Regulator Majewski 2003 85 patients w/GA or CNV in Tuo et al.70
(C-X3-C motif) of immune & Jun 2005 at least one eye,
Receptor 1 acute phase 105 controls (Clinically
response diagnosed group),
21 patients w/CNV,
19 w/areolar AMD &
171 controls (pathologically
diagnosed group)
SECTION 10

Fibulin-5 FBLN5 14q32.1 Maintenance of Jun 2005 402 patients w/drusen, Stone et al.79
extracellular & Schmidt disruption or atrophy of
matrix 2004 RPE or CNV, 429 controls
integrity (80% of both groups
Caucasian)
805 European patients Lotery et al.80
w/ AMD (Int. Class. System),
279 unrelated controls
≥ 65 y.o.
Toll-Like TLR4 9q32 – Regulator Majewski 667 unrelated AMD patients Zareparsi et al.81
Receptor 4 q33 of innate 2003 (Int. ARM Epi. Study),
immunity 439 unrelated controls,
all Caucasian
Complement C2 6p21.3 Regulator 898 unrelated patients Gold et al. 2005
component 2 of alternative- ≥ 60 y.o. w/AMD (Int. ARM
complement- Epi. Study), 389 unrelated
pathway age and ethnicity matched
controls, all of
European-American descent
Complement CFB 6p21.3 Regulator 898 unrelated patients Gold et al. 2005
Factor B of alternative- ≥ 60 y.o. w/AMD
complement- (Int. ARM Epi. Study),
pathway 389 unrelated age and
ethnicity matched controls,
all of European-American
descent
Complement CFH 1q32 Regulator of Klein 1998, 992 Caucasian patients Conley et al.10
Factor H alternative- Weeks 2001, w/extensive drusen,
complement- Majewski 2003, pigmentary changes,
pathway and Seddon 2003 GA and/or CNV,
innate & Iyengar 2004 120 unrelated controls
immunity
400 patients w/extensive Edwards et al.5
drusen or macular
pigmentary abnormalities,
202 controls w/no more
than 4 small, hard drusen
954 unrelated AMD patients Hageman et al.9
60 or older, 406 age- and
ethnicity-matched controls
Patients w/CNV and/or GA Jakobsdottir
– 1443 affected from et al.71
594 families, 196 sporadic
cases, 296 unrelated
controls
96 white patients with large Klein et al.6
drusen, GA and/or CNV,
50 white controls

Continued
1892
Genetics of Age-related Macular Degeneration

TABLE 143.4. Candidate Genes for AMD (Cont’d)

Previously
Identified
Gene name Symbol Location Function Region? Study Population Study

1166 German AMD patients Rivera et al.72


(Int. Class. System),
945 unrelated controls
2 unrelated CNV groups from Souied et al.11
France, 60 sporadic and
81 familial cases compared
w/ 91 controls
616 white patients w/CNV, Zareparsi et al.81
GA and/or large drusen,
275 white controls at least
68 y.o.
The Rotterdam Study – Despriet et al.

CHAPTER 143
2387 patients, of whom 2006
49 had soft drusen, GA or
CNV, remainder controls
146 unrelated Japanese Gotoh et al.62*
patients w/CNV,
105 controls
163 Chinese patients w/CNV, Lau et al. 2006
232 age-matched controls
Patients w/soft drusen, Magnusson et al.
GA and/or CNV, 1016 from 2006
Iceland and 431 from
the US, 1108 unaffected
Icelandic relatives,
431 controls
96 Japanese patients w/CNV, Okamoto et al.
89 matched controls 2006
between 50-85 y.o.
647 patients ≥ grade 3 Postel et al. 2006
(AREDS scale) and at least
55 y.o., 163 controls grade
1 or 2
111 white males w/drusen, Schaumberg et al.
RPE changes, atrophy, 2006
hypertrophy, RPE
detachment, GA, subretinal
CNV membrane and/or
disciform scars, 401 male
controls
335 Finnish patients w/large Seitsonen et al.40
drusen, GA, or CNV, of
which 154 were sporadic
cases & 181 familial,
105 controls
443 unrelated English Sepp et al.12
patients w/GA or CNV
(mean number of pack
years of cigarette smoking
= 15.4±18.8), 262 spouses
as controls (pack years
=10.7±14.5)
104 unrelated Italian AMD Simonelli et al.
patients (Int. Class. System) 2006
and 131 unrelated controls
Very Low VLDLR 9p24 Lipid Iyengar 992 Caucasian patients Conley et al.10*
Density metabolism 2004 w/extensive drusen,
Lipoprotein and transport pigmentary changes,
Receptor GA and/or CNV,
120 unrelated controls

Continued
1893
RETINA AND VITREOUS

TABLE 143.4 Candidate Genes for AMD (Cont’d)

Previously
Identified
Gene name Symbol Location Function Region? Study Population Study

Two independent data sets – Haines et al.66


314 affected and
97 unaffected from the
family dataset, 399 affected
and 159 unaffected from
the case-control dataset.
Affecteds had extensive
drusen ( ≥ 63–125 µm),
GA or CNV
PLEcKstrin PLEKHA1 10q26.13 Phospholipid Weeks 2001, Patients w/CNV and/or GA – Jakobsdottir
Homology domain binding Majewski 2003, 1443 affected from et al.71
containing, indirectly Seddon 2003, 594 families, 196 sporadic
family A immune Iyengar 2004 cases, 296 unrelated
(phosphoinositide response & Kenealy controls
SECTION 10

binding specific) 2004


member 1
1166 German AMD patients Rivera et al.72*
(Int. Class. System),
945 unrelated controls
810 white unrelated patients Schmidt et al.73*
w/extensive intermediate
or large (≥ 125 µm) drusen,
GA or CNV (39% reported
smokers), 259 unrelated
controls
LOC387715 LOC 10q26.13 Unknown Weeks 2001, Patients w/CNV and/or GA – Jakobsdottir
387715 Majewski 2003, 1443 affected from et al.71
Seddon 2003, 594 families, 196 sporadic
Iyengar 2004 cases, 296 unrelated
& Kenealy 2004 controls
1166 German AMD patients Rivera et al.72
(Int. Class. System),
945 unrelated controls
810 caucasion unrelated Schmidt et al.73
patients w/extensive
intermediate or large
(≥ 125 µm) drusen, GA or
CNV (39% reported smokers),
259 unrelated controls
HtrA serine HTRA1 10q26 Unknown Weeks 2001, 96 patients with neovascular Dewan et al.,
peptidase 1 Majewski 2003, AMD and 130 age-matched 2006
Seddon 2003, controls of Southeast Asian
Iyengar 2004 descent
& Kenealy 2004
442 AMD cases (265 CNV Yang et al., 2006
and 177 with soft confluent
drusen) and 309 normal
controls from a Caucasian
population in Utah
Vascular VEG-F 6p12 Regulator of Two independent data sets – Haines et al.66
Endothelial angiogenesis 314 affected and 97
Growth Factor unaffected from the family
dataset, 399 affected and
159 unaffected from the
case-control dataset.
Affecteds had extensive
drusen ( ≥ 63–≤ 125 µm),
GA or CNV

Continued

1894
Genetics of Age-related Macular Degeneration

TABLE 143.4 Candidate Genes for AMD (Cont’d)

Previously
Identified
Gene name Symbol Location Function Region? Study Population Study

Low density LRP6 12p13 – Lipid Two independent data sets – Haines et al.66
lipoprotein p11 metabolism 314 affected and
Receptor-related and transport 97 unaffected fromthe
Protein 6 family dataset, 399 affected
and 159 unaffected from
the case-control dataset.
Affecteds had extensive
drusen (≥ 63–≤ 125 µm),
GA or CNV
Excision ERCC6 10q11.23 DNA repair 460 advanced AMD cases and Tuo et al.76
Repair Cross- 269 age-matched controls
complementing
rodent repair
deficiency,

CHAPTER 143
Complementation
group 6
*Indicates no association found.
Int. Class System - International Classification System, CNV - Choroidal Neovascularization, GA - Geographic Atrophy,
Int. ARM Epi. Study - International Age Related Maculopathy Epidemiology Study, RPE - Retinal Pigment Epithelial, Int. Class. System - International Classification
System, y.o. - years old, AREDS - Age Related Eye Disease Study.

and Q192R (a glutamine is substituted by arginine) variants MANGANESE SUPEROXIDE DISMUTASE 2


were reported to be associated with neovascular AMD in GENE (SOD-2)
72 individuals from Japan.63 Further the affected individuals
also had significantly higher levels of low-density lipoprotein SOD2, localized to 6q25.3, encodes an enzyme with
in the plasma.63 Two other studies, however, found no antioxidant properties and is also expressed in the retina. The
association between these variants in patients diagnosed with substitution of an alanine for a valine homozygously in exon
late AMD of Anglo-Celtic origin64 or patients with neovascular 2 of this gene was found to be associated with a 10-fold
AMD from Northern Ireland.65 Again, it is possible that there increased risk of neovascular AMD in 102 Japanese patients
exists multiple susceptibility genes for AMD, that are not when compared to 200 ethnically matched controls.67 Like
necessarily expressed in every patient with AMD, possibly due in the case of PON1, the variant in SOD2 could not be found
to ethnic heterogeneity. in patients with neovascular AMD from Northern Ireland at
a statistically significant level when compared to controls.65
Again, it is possible that there exist multiple susceptibility
VERY-LOW-DENSITY LIPOPROTEIN genes for AMD, not necessarily expressed in every patient with
RECEPTOR GENE (VLDLR) AMD, possibly due to ethnic heterogeneity.
Like APOE and PON-1, VLDLR is also involved in lipid meta-
bolism and transport. There is a lack of agreement between the
two studies conducted to date as to the association of VLDR CYSTATIN C GENE (CST3)
and AMD. VLDR is located on the short arm of chromosome 9 CST3, similar to TIMP3 (Sorsby's fundus dystrophy), is a
(9p24), a region found to be significantly associated with late protease inhibitor. CST3 is localized to 20p11.21, a region as
AMD in one genome-wide scan (Tables 143.3 and 143.4).46 yet to be reported as being linked to any type of AMD. The
While one study found a significant association of this gene homozygous variant in the 5' untranslated region of CST3 state
with all types of AMD in Caucasian patients including those was demonstrated to be associated with almost 7% of German
with large, extensive drusen, neovascularization, and geo- patients with neovascular AMD.68 Further study of this gene
graphic atrophy using SNPs,66 another large study on Caucasian in the etiology of AMD is warranted as this is the only
patients with similar AMD diagnosis did not find a significant investigation to date.
association with VLDR.10 More studies may need to be done to
validate the role of VLDR in the pathophysiology of AMD.
ANGIOTENSIN I CONVERTING ENZYME GENE
(ACE)
LOW-DENSITY LIPOPROTEIN RECEPTOR- The role of ACE as a regulator of blood pressure has been
RELATED PROTEIN 6 GENE (LRP6) established. ACE is localized to the long arm of chromosome
Another gene involved in lipid metabolism and transport is 17 (17q23.3). The polymorphism in this gene that was found to
LRP6. Using a combination of linkage and association be associated with 'decreased' susceptibility to AMD is a
analysis on family-based and case-control populations, repetitive element called an Alu.69 This polymorphism was
respectively, Haines et al demonstrated that LRP6 was found to be significantly associated homozygously with a
associated with advanced AMD in Caucasian patients.66 fivefold decreased risk from the early dry or atrophic forms
Although LRP6 is located in a region, 12p11–p13, yet to be of AMD.69 This association was found after studying patients
identified by genome-wide scans as significant, its recent with extensive small (<63 mm) drusen, intermediate (>125 mm)
discovery as a candidate gene in the etiology of AMD may drusen, geographic atrophy as well as neovascular AMD.
warrant further study. However, another study that examined a similar spectrum of 1895
RETINA AND VITREOUS

AMD phenotypes concluded that there was no association of another, teasing out precisely which gene in this region is
ACE with either a decreased or increased risk of AMD.10 associated with AMD may require further investigation.

CHEMOKINE (C-X3-C MOTIF) RECEPTOR 1 VASCULAR ENDOTHELIAL GROWTH FACTOR


GENE (CX3CR1) GENE (VEGF)
CX3CR1 functions as a regulator of the immune response in VEGF is localized to the short arm of chromosome 6 (6p12), a
the acute phase. region as yet to be identified in any genome-wide scan (Table
CXCR1 is localized to the short arm of chromosome 143.2). The role of VEGF as a regulator of angiogenesis is well
3 (3p21.3), a region found to be significantly linked to the established. Further, VEGF is a target for therapeutic
advanced stages of AMD by two independent genome-wide interventions, such as Macugen and Lucentis. Only one
scans (Tables 143.3 and 143.4).44,51 Two polymorphisms study to date has examined the genetic contribution of VEGF
CX3CR1, V249I (a valine is substituted by isoleucine) and to AMD.66
T280M (a threonine is substituted by methionine), were Employing a combination of linkage (family-based) and
found both homozygously and heterozygously to be associated association (sporadic) analysis using SNPs, common variation
with cases of advanced AMD.70 Further, this study also in this gene was significantly found to be associated with
demonstrated decreased level of expression of this gene in the AMD.66 The affected individuals consisted of those with drusen
maculae of a patient with the T280M variation in the only (63–125 mm) geographic atrophy or neovascularization.
SECTION 10

homozygous state, when compared to the maculae from


a control individual. Given that CX3CR1 is in a previously
linked region to AMD, the findings of a positive association
EXCISION REPAIR CROSS-
with risk of AMD and decreased level of expression of this COMPLEMENTATION GROUP 6 GENE (ERCC6)
gene in an AMD retina should warrant further study. ERCC6 is localized to 10q11.23 and functions in the repair of
DNA as well as aging. A SNP in the 5'-untranslated region of
this gene was reported to be significantly associated in the
PLECKSTRIN HOMOLOGY DOMAIN homozygous state with advanced AMD.76 Further, increased
CONTAINING FAMILY A (PHOSPHOINOSITIDE expression of this gene was observed in lymphocytes from
BINDING SPECIFIC) MEMBER 1 GENE individuals that were homozygous for his variation. To date
(PLEKHA 1), HYPOTHETICAL LOC387715 this is the only study showing an association of this gene
GENE (LOC387715), AND HTRAA SERINE with AMD.
PEPTODASE 1 (HTRA1)
PLEKHA1, LOC387715, and HRTA1 are localized to the long HEMICENTIN-1 GENE (FIBULIN 6, FIBL-6)
arm of chromosome 10 (10q26.13), a region consistently found In 1998 Klein (Oregon) described a family linked to the long
by many genome-wide scans to be linked to both early and the arm of chromosome 1 (1q25–1q32).41 Subsequently, this same
more advanced stages of AMD (Tables 143.3 and 143.4).42–46,48,49 region has been found by many studies to be linked or
Meta-analysis or pooled findings from six genome-wide scans associated with both early and advanced stages of AMD (Table
demonstrated that the 10q26 demonstrated the greatest 143.3).6,42–46,50 In 2003, Schultz et al analyzed this same
significance for linkage compared to other significant regions, multigenerational family and found that a mutation in
such as the 1q25-q32 region.50 PLEKHA1 is believed to hemicentin-1 (Fibl-6) was associated with the early stage of
participate in phospholipid binding and more indirectly in the AMD.77 Specifically, this group found that a Gln5345Arg
immune response, whereas LOC387715, a gene that is not well change (a glutamine is replaced by arginine) in exon 104
conserved across species, has no attributable function. segregated with the disease. Based on these results, it was
Although the function of HTRA1 is unknown, it ahs been suggested that hemicentin-1 was the gene responsible for the
reported that the encoded protein is upregulated in aging and phenotype observed in this family.76 Since this time, there
downregulated in certain tumors. Using a combination of have been no other confirmatory reports that the Gln5345Arg
linkage and association employing SNPs specific to the 10q26 variant in hemicentin-1 is associated with any type of AMD
region, Jakobsdottir et al demonstrated that while both (Table 143.3).10,40,78,79 However, Iyengar et al46 reported that
PLEKHA1 and LOC387715 were significantly associated with this gene could be involved in the etiology of AMD because
both neovascular and atrophic AMD, the association was linkage was observed for four SNPs in this gene. Iyengar then
stronger for PLEKHA1. Over 1400 affected individuals further concluded that although they did not find the specific
comprised of both familial and sporadic cases of AMD were Gln5345Arg that Schultz did, that maybe other variants in
examined.71 Rivera et al72 extended these findings by FIBL-6, as yet to be discovered, could be associated with AMD.46
demonstrating that a specific variant within the LOC387715
locus was associated with increased AMD risk after examining
a cohort that consisted of both early and advanced stages. FIBULIN 5 (FBLN5)
Similar findings were replicated in a Caucasian population that FBLN5, located on the long arm of chromosome 14 (14q32.1),
consisted of advanced cases, including large drusen (=125 µm), was chosen as a candidate gene because of its similarity to
geographic atrophy and neovascularization.73 These two studies Fibulin 3, for which a mutation in this gene is associated with
concluded that LOC387715, and not PLEKHA1, was associated Doyne's honeycomb dystrophy, a disease with some overlapping
with AMD. More recently, it was demonstrated that another phenotype to AMD (Table 143.2). More recently, the 14q32
variant or SNP in the HTRA1 promotor, in linkage region has been reported by one genome-wide scan to be linked
disequilibrium (LD) with the LOC 387715 variant, is likely the to advanced AMD.51 Like hemicentin-1, FIBLN5 is an
functional polymorphism responsible for increased AMD extracellular matrix protein that functions in maintaining the
risk.74,75 Given that 10q26 is a region that has been consistently integrity of elastic lamina, similar to what is found in Bruch's
linked with AMD by many genome-wide scans and that membrane. Various mutations in FIBLN5 were reported to be
1896 PLEKHA1, LOC387715, and TRA1 are located adjacent to one associated with AMD in 7 out of 40 AMD patients in
Genetics of Age-related Macular Degeneration

a case-control study.80 Phenotypically, the seven patients all had population. CFH is localized to 1q32, a region found by both
a distinct and unusual pattern of drusen in common. Three linkage6,42–46,50 and by a study of association6 to be associated
out of the seven patients also had neovascularization. None with any subtype of AMD. CFH functions in regulation of the
of these mutations were found in unaffected individuals. alternative complement pathway as well as innate immunity.
Stone et al concluded that fibulin 5 accounts for 1.7% of all Some groups have reported that common variation in CFH,
cases of AMD.80 A recent study has replicated these as well as other than Y402H, is associated with both early and advanced
showed that two additional novel variants in FIBLN5 are forms of AMD.9,74,83 Interestingly, some of these same variants
associated with other subtypes of AMD.80 This study further (or a combination of them), including Y402H, are associated
demonstrated in vitro that there is a reduction in secretion of with increased risk for other diseases such as myocardial
FIBLN5 in COS7 cells from patients expressing these infarction,85 hemolytic uremic syndrome (HUS),86 and
variants.81 membranoproliferative glomerulonephritis (MPGN).87 This by
no means indicates that CFH does not play an important role
in the pathophysiology of AMD but underscores the importance
TOLL-LIKE RECEPTOR 4 GENE (TLR4) of examining common variation in multiple susceptibility genes
TLR4 is localized to the long arm of chromosome 9 (9q32–q33) as well as contributing environmental factors simultaneously to
a region previously linked to patients with the advanced forms get a better estimate of an individual's risk of AMD. Several
of AMD.44 A variant in this gene which functions in innate groups have begun to do this with respect to the contribution of
immunity and lipid transport and metabolism efflux has been CFH along with other reported genetic and epidemiological risk

CHAPTER 143
SECTION 10

previously shown to be associated with a 'decreased' risk of factors for AMD. Some of these studies include, CFH,
arteriosclerosis. This same variant, D299G (asparagine is LOC387715 and smoking,73 CFH and smoking,12 CFH,
substituted by glycine), was found to be associated with a Complement Component 2 Gene (C2), Complement Factor B
2.6-fold 'increase' in risk of AMD.82 Further, this study Gene (CFB),87 and CFH, C2, CFB, and LOC387715.
concluded that an individual's risk is increased even more
when this variant is present in the same individual along with
variants in APOE and/or ATP-binding cassette transporter-1
COMPLEMENT COMPONENT 2 GENE (C2)
gene (ABCA1) gene. Given the suggestive findings of linkage AND COMPLEMENT FACTOR B GENE (CFB)
and association between TLR4 and AMD further studies are C2 and CFB are both part of a major histocompatibility com-
warranted. plex that, like CFH, regulates the alternative complement
pathway. Recently, Gold et al postulated that because
variation in CFH had been significantly associated with AMD,
COMPLEMENT FACTOR H GENE (CFH) genes in the alternative complement pathway may also play a
The last year in the field of AMD genetics has focused on role in the pathophysiology of AMD.88 This study demonstrated
the exciting discovery of the association of CFH with AMD that combinations of SNPs in these two genes decreased
(Fig. 143.2 and Table 143.4). As stated in the beginning of an individual's risk of AMD. Specifically, the protective effect
this chapter, numerous independent reports have shown that on an individual in having variation in these genes was most
the functional polymorphism Y402H in CFH, where a tyrosine significant in those who had two histidine alleles in the CFH
is substituted by a histidine, to be associated with a two- to gene. In other words individuals who were homozygous for
sevenfold increased risk of both early and/or late stages of the Y402H variation in exon 9 of CFH were afforded the
AMD.5–12,83 Only one study has found no association of the most protection from risk of AMD.87 These findings were
CFH gene to AMD.83 This study examined the Y402 variant in recently confirmed in a published report that examined
a Japanese cohort with neovascular AMD.84 This negative simultaneously CFH, C2, CFB, and LOC387715 on risk of
finding may be possibly due to the fact that other AMD advanced AMD.
susceptibility genes may explain the phenotype in this
Key Features
• A role for Inflammation, lipid transport and metabolism as well
Candidate Genes
18
as oxidative stress in the pathophysiology of AMD may be
supported by findings that show that candidate genes
16 associated with these pathways may be involved in the early
14 or late stages of AMD
Number of Studies

• The CFH Y402H variant is the most consistently associated


12
genetic risk factor for AMD
10 • Genome-wide scans and candidate gene association studies
8
are complementary methods

6
In summary, genes could potentially be used as biological
4
markers in the early or presymptomatic diagnosis of the
2 neovascular form of AMD. Genes or biological markers that
are identified can then be used as possible disease targets for
0
1998 1999 2000 2001 2002 2003 2004 2005 2006 the development of preventative therapies for AMD. Further the
Year of Study
findings of candidate gene associations with risk of AMD
point to possible mechanisms or pathways involved in the
FIGURE 143.2. The number of studies to determine the association
etiology of AMD. Studying the hypothesized mechanisms
between the candidate genes and AMD. With the number of
candidate genes increasing, the number of studies performed has involving oxidative stress, inflammation, and lipid metabolism
also increased each year. Because of recent findings of association of from both an epidemiologic (reviewed in detail elsewhere in
AMD with the CFH gene, a large number of studies were performed in this book) and genetic perspective is necessary to further
2005 and 2006. elucidate the role of these pathways in AMD. Given the 1897
RETINA AND VITREOUS

multifactorial and heterogeneous nature of AMD, comple- AMD. Both population stratification between cases and
mentary methods are necessary to detect weak to moderate controls as well as heterogeneity in phenotype among cases, can
associations to identify the contribution epidemiologic and confound findings in analysis of data. Inconsistency in
genetic risk factors make independently and in combination replication of findings among studies could possibly be due to
in order to more accurately determine the overall risk of these factors.

REFERENCES
1. The Eye Diseases Prevalence Research degeneration and other maculopathies. 28. Allikmets R: Further evidence for an
Group: Prevalence of Age-Related Macular Hum Genet 1999; 104:449–453. association of ABCR alleles with age-
Degeneration in the United States. Arch 16. Kramer F, White K, Pauleikhoff D, et al: related macular degeneration. The
Ophthalmol 2004; 122:564–572. Mutations in the VMD2 gene are associated International ABCR Screening Consortium.
2. Klein R, Klein BE, Linton KL: Prevalence of with juvenile-onset vitelliform macular Am J Hum Genet 2000; 67:487–491.
age-related maculopathy. The beaver dam dystrophy (Best disease) and adult 29. Stone EM, Webster AR, Vandenburgh K,
eye study. Ophthalmology 1992; vitelliform macular dystrophy but not age- et al: Allelic variation in ABCR associated
99:933–943. related macular degeneration. Eur J Hum with Stargardt disease but not age-related
3. Ferris FL, Davis MD, Clemons TE, et al: A Genet 2000; 8:286–292. macular degeneration [letter]. Nat Genet
simplified severity scale for age-related 17. Lotery AJ, Munier FL, Fishman GA, et al: 1998; 20:328–329.
SECTION 10

macular degeneration. Arch Ophthalmol Allelic variation in the VMD2 gene in best 30. De La Paz MA, Guy VK, Abou-Donia S,
2005; 123:1570–1574. disease and age-related macular et al: Analysis of the Stargardt disease
4. Thornton J, Edwards R, Mitchell P, et al: degeneration. Invest Ophthalmol Vis Sci gene (ABCR) in age-related macular
Smoking and age-related macular 2000; 41:1291–1296. degeneration. Ophthalmology 1999;
degeneration: a review of association. Eye 18. Akimoto A, Akimoto M, Kuroiwa S, et al: Lack 106:1531–1536.
2005; 19:935–944. of association of mutations of the bestrophin 31. Souied EH, Ducroq D, Gerber S, et al:
5. Edwards AO, Ritter R III, Abel KJ, et al: gene with age-related macular degeneration Age-related macular degeneration in
Complement factor H polymorphism and in non-familial Japanese patients. Graefes grandparents of patients with Stargardt
age-related macular degeneration. Science Arch Clin Exp Ophthalmol 2001; 239:66–68. disease: genetic study. Am J Ophthalmol
2005; 308:421–424. 19. Seddon JM, Afshari MA, Sharma S, et al: 1999; 128:173–178.
6. Klein RJ, Zeiss C, Chew EY, et al: Assessment of mutations in the Best 32. Rivera A, White K, Stohr H, et al:
Complement factor H polymorphism in macular dystrophy (VMD2) gene in patients A comprehensive survey of sequence
age-related macular degeneration. Science with adult-onset foveomacular vitelliform variation in the ABCA4 (ABCR) gene in
2005; 308:385–389. dystrophy, age-related maculopathy, and Stargardt disease and age-related macular
7. Haines JL, Hauser MA, Schmidt S, et al: bull's-eye maculopathy. Ophthalmology degeneration. Am J Hum Genet 2000;
Complement factor H variant increases the 2001; 108:2060–2067. 67:800–813.
risk of age-related macular degeneration. 20. De La Paz MA, Pericak-Vance MA, 33. Guymer RH, Heon E, Lotery AJ, et al:
Science 2005; 308:419–421. Lennon F, et al: Exclusion of TIMP3 as a Variation of codons 1961 and 2177 of
8. Zareparsi S, Branham KEH, Li M, et al: candidate locus in age-related macular the Stargardt disease gene is not
Strong association of the Y402H variant in degeneration. Invest Ophthalmol Vis Sci associated with age-related macular
complement factor H at 1q32 with 1997; 38:1060–1065. degeneration. Arch Ophthalmol 2001;
susceptibility to age-related macular 21. Felbor U, Doepner D, Schneider U, et al: 119:745–751.
degeneration. Am J Hum Genet 2005; Evaluation of the gene encoding the tissue 34. Webster AR, Heon E, Lotery AJ, et al:
77:5–13. inhibitor of metalloproteinases-3 in various An analysis of allelic variation in the
9. Hageman GS, Anderson DH, Johnson LV, maculopathies. Invest Ophthalmol Vis Sci ABCA4 gene. Invest Ophthalmol Vis Sci
et al: A common haplotype in the 1997; 38:1054–1059. 2001; 42:1179–1189.
complement regulatory gene factor H 22. Kemp CM, Jacobson SG, Cideciyan AV, 35. Bernstein PS, Leppert M, Singh N, et al:
(HF1/CFH) predisposes individuals to age- et al: RDS gene mutations causing retinitis Genotype-phenotype analysis of ABCR
related macular degeneration. Proc Natl pigmentosa or macular degeneration lead variants in macular degeneration probands
Acad Sci USA 2005; 102:7227–7232. to the same abnormality in photoreceptor and siblings. Invest Ophthalmol Vis Sci
10. Conley YP, Thalamuthu A, Jakobsdottir J, function. Invest Ophthalmol Vis Sci 1994; 2002; 43:466–473.
et al: Candidate gene analysis suggests a 35:3154–3162. 36. Schmidt S, Postel EA, Agarwal A, et al:
role for fatty acid biosynthesis and 23. Gorin MB, Jackson KE, Ferrell RE, et al: Detailed analysis of allelic variation in the
regulation of the complement system in the A peripherin/retinal degeneration slow ABCA4 gene in age-related maculopathy.
etiology of age-related maculopathy. Hum mutation (Pro-210-Arg) associated with Invest Ophthalmol Vis Sci 2003;
Mol Genet 2005; 14:1991–2002. macular and peripheral retinal degeneration. 44:2868–2875.
11. Souied EH, Leveziel N, Richard F, et al: Ophthalmology 1995; 102:246–255. 37. Schick JH, Iyengar SK, Klein BE, et al:
Y402H complement factor H polymorphism 24. Stone EM, Lotery AJ, Munier FL, et al: A whole-genome screen of a quantitative
associated with exudative age-related A single EFEMP1 mutation associated with trait of age-related maculopathy in sibships
macular degeneration in the French both Malattia Leventinese and Doyne from the Beaver Dam Eye Study. Am J
population. Mol Vis 2005; 11:1135–1140. honeycomb retinal dystrophy. Nat Genet Hum Genet 2003; 72:1412–1424.
12. Sepp T, Khan JC, Thurlby DA, et al: 1999; 22:199–202. 38. Schmidt S, Scott WK, Postel EA, et al:
Complement factor H variant Y402H is a 25. Guymer RH, McNeil R, Cain M, et al: Ordered subset linkage analysis supports a
major risk determinant for geographic Analysis of the Arg345Trp disease- susceptibility locus for age-related macular
atrophy and choroidal neovascularization in associated allele of the EFEMP1 gene in degeneration on chromosome 16p12. BMC
smokers and nonsmokers. Invest individuals with early onset drusen or Genet 2004; 5:18.
Ophthalmol Vis Sci 2006; 47:536–540. familial age-related macular degeneration. 39. Ayyagari R, Zhang K, Hutchinson A, et al:
13. Seddon JM, Ajani UA, Mitchell BD. Familial Clin Exp Ophthalmol 2002; 30:419–423. Evaluation of the ELOVL4 gene in patients
aggregation of age-related maculopathy. 26. Narendran N, Guymer RH, Cain M, with age-related macular degeneration.
Am J Ophthalmol 1997; 123:199–206. Baird PN. Analysis of the EFEMP1 gene in Ophthalmic Genet 2001; 22:233–239.
14. Hammond CJ, Webster AR, Snieder H, individuals and families with early onset 40. Seitsonen S, Lemmela S, Holopainen J,
et al: Genetic influence on early age-related drusen. Eye 2005; 19:11–15. et al: Analysis of variants in the
maculopathy: a twin study. Ophthalmology 27. Allikmets R, Shroyer NF, Singh N, et al: complement factor H, the elongation of
2002; 109:730–736. Mutation of the Stargardt disease gene very long chain fatty acids – like 4 and the
15. Allikmets R, Seddon JM, Bernstein PS, (ABCR) in age-related macular hemicentin 1 genes of age-related macular
et al: Evaluation of the Best disease gene degeneration. Science 1997; degeneration in the Finnish population.
1898 in patients with age-related macular 277:1805–1807. Mol Vis 2006; 12:796–801.
Genetics of Age-related Macular Degeneration

41. Klein ML, Schultz DW, Edwards A, et al: 57. Zareparsi S, Reddick AC, Branham KE, macular degeneration, contributing
Age-related macular degeneration. Clinical et al: Association of apolipoprotein E alleles independently of complement factor H to
features in a large family and linkage to with susceptibility to age-related macular disease risk. Hum Mol Genet 2005;
chromosome 1q. Arch Ophthalmol 1998; degeneration in a large cohort from a single 14:3227–3236.
116:1082–1088. center. Invest Ophthalmol Vis Sci 2004; 73. Schmidt S, Hauser MA, Scott WK, et al:
42. Weeks DE, Conley YP, Tsai HJ, et al: 45:1306–1310. Cigarette smoking strongly modifies the
Age-related maculopathy: an expanded 58. Baird PN, Guida E, Chu DT, et al: The association of LOC387715 and age-related
genome-wide scan with evidence of epsilon2 and epsilon4 alleles of the macular degeneration. Am J Hum Genet
susceptibility loci within the 1q31 and apolipoprotein gene are associated with 2006; 78:852–864.
17q25 regions. Am J Ophthalmol 2001; age-related macular degeneration. Invest 74. Dewan A, Liu M, Hartman S et al: HTRA1
132:682–692. Ophthalmol Vis Sci 2004; 45:1311–1315. promoter polymorphism in wet age-related
43. Weeks DE, Conley YP, Tsai HJ, et al: Age- 59. Pang CP, Baum L, Chan WM, et al: The macular degeneration. Science
related maculopathy: a genomewide scan apolipoprotein E epsilon4 allele is unlikely 2006;10;314:989–992.
with continued evidence of susceptibility to be a major risk factor of age-related 75. Yang Z, Camp NJ, Sun H et al: A variant of
loci within the 1q31, 10q26, and 17q25 macular degeneration in Chinese. the HTRA1 gene increases susceptibility to
regions. Am J Hum Genet 2004; Ophthalmologica 2000; 214:289–291. age-related macular degeneration. Science
75:174–189. 60. Schmidt S, Haines JL, Postel EA, et al: 2006 November 10;314:992–993.
44. Majewski J, Schultz DW, Weleber RG, et al: Joint effects of smoking history and APOE 76. Tuo J, Ning B, Bojanowski CM, et al:
Age-related macular degeneration – a genotypes in age-related macular Synergic effect of polymorphisms in
genome scan in extended families. Am J degeneration. Mol Vis 2005; 11:941–949. ERCC6 5' flanking region and complement

CHAPTER 143
Hum Genet 2003; 73:540–550. 61. Schultz DW, Klein ML, Humpert A, et al: factor H on age-related macular
45. Seddon JM, Santangelo SL, Book K, et al: Lack of an association of apolipoprotein E degeneration predisposition. Proc Natl
A genomewide scan for age-related gene polymorphisms with familial age- Acad Sci USA 2006; 103:9256–9261.
macular degeneration provides evidence related macular degeneration. Arch 77. Schultz DW, Klein ML, Humpert AJ, et al:
for linkage to several chromosomal regions. Ophthalmol 2003; 121:679–683. Analysis of the ARMD1 locus: evidence
Am J Hum Genet 2003; 73:780–790. 62. Gotoh N, Kuroiwa S, Kikuchi T, et al: that a mutation in HEMICENTIN-1 is
46. Iyengar SK, Song D, Klein BEK, et al: Apolipoprotein E polymorphisms in associated with age-related macular
Dissection of genomewide-scan data in Japanese patients with polypoidal degeneration in a large family. Hum Mol
extended families reveals a major locus choroidal vasculopathy and exudative Genet 2003; 12:3315–3323.
and oligogenic susceptibility for age-related age-related macular degeneration. Am J 78. Hayashi M, Merriam JE, Klaver CC, et al:
macular degeneration. Am J Hum Genet Ophthalmol 2004; 138:567–573. Evaluation of the ARMD1 locus on 1q25–31
2005; 74:39. 63. Ikeda T, Obayshi H, Hasegawa G, et al: in patients with age-related maculopathy:
47. Abecasis GR, Yashar BM, Zhao Y, et al: Paraoxonase gene polymorphisms and genetic variation in laminin genes and in
Age-related macular degeneration: a high- plasma oxidized low-density lipoprotein exon 104 of HEMICENTIN-1. Ophthalmic
resolution genome scan for susceptibility level as possible risk factors for exudative Genet 2004; 25:111–119.
loci in a population enriched for late-stage age-related macular degeneration. Am J 79. McKay GJ, Clarke S, Hughes A, et al:
disease. Am J Hum Genet 2004; Ophthalmol 2001; 132:191–195. A novel diagnostic test detects a low
74:482–494. 64. Baird PN, Chu D, Guida E, et al: frequency of the hemicentin Gln5345Arg
48. Weeks DE, Conley YP, Mah TS, et al: A full Association of the M55L and Q192R variant among Northern Irish age related
genome scan for age-related maculopathy. paraoxonase gene polymorphisms with macular degeneration patients. Mol Vis
Hum Mol Genet 2000; 9:1329–1349. age-related macular degeneration. Am J 2004; 10:682–687.
49. Kenealy SJ, Schmidt S, Agarwal A, et al: Ophthalmol 2004;138:665–666. 80. Stone EM, Braun TA, Russell SR, et al:
Linkage analysis for age-related macular 65. Esfandiary H, Chakravarthy U, Patterson C, Missense variations in the fibulin 5 gene
degeneration supports a gene on et al: Association study of detoxification and age-related macular degeneration.
chromosome 10q26. Mol Vis 2004; 10:57–61. genes in age related macular degeneration. N Engl J Med 2004; 351:346–353.
50. Fisher SA, Abecasis GR, Yashar BM, et al: Br J Ophthalmol 2005; 89:470–474. 81. Lotery AJ, Baas D, Ridley C, et al: Reduced
Meta-analysis of genome scans of age- 66. Haines JL, Schnetz-Boutaud N, Schmidt S, secretion of fibulin 5 in age-related macular
related macular degeneration. Hum Mol et al: Functional candidate genes in age- degeneration and cutis laxa. Hum Mutat
Genet 2005; 14:2257–2264. related macular degeneration: significant 2006; 27:568–574.
51. Jun G, Klein BE, Klein R, et al: Genome- association with VEGF, VLDLR, and LRP6. 82. Zareparsi S, Buraczynska M, Branham
wide analyses demonstrate novel loci that Invest Ophthalmol Vis Sci 2006; KEH, et al: Toll-like receptor 4 variant
predispose to drusen formation. Invest 47:329–335. D299G is associated with susceptibility to
Ophthalmol Vis Sci 2005; 46:3081–3088. 67. Kimura K, Isashiki Y, Sonoda S, et al: age-related macular degeneration. Hum
52. Klaver CC, Kliffen M, Van Duijn CM, et al: Genetic association of manganese Mol Genet 2005; 14:1449–1455.
Genetic association of apolipoprotein E superoxide dismutase with exudative 83. Li M, tmaca-Sonmez P, Othman M, et al:
with age-related macular degeneration. age-related macular degeneration. Am J CFH haplotypes without the Y402H coding
Am J Hum Genet 1998; 63:200–206. Ophthalmol 2000; 130:769–773. variant show strong association with
53. Souied EH, Benlian P, Amouyel P, et al: 68. Zurdel J, Finckh U, Menzer G, et al: CST3 susceptibility to age-related macular
The e4 allele of the alipoprotein E gene as genotype associated with exudative age degeneration. Nat Genet 2006;
a potential protective factor for exudative related macular degeneration. Br J 38:1049–1054.
age-related macular degeneration. Am J Ophthalmol 2002; 86:214–219. 84. Gotoh N, Yamada R, Hiratani H, et al: No
Ophthalmol 1998; 125:353–359. 69. Hamdi HK, Reznik J, Castellon R, et al: association between complement factor H
54. Simonelli F, Margaglione M, Testa F, et al: Alu DNA polymorphism in ACE gene is gene polymorphism and exudative age-
Apolipoprotein E polymorphisms in age- protective for age-related macular related macular degeneration in Japanese.
related macular degeneration in an Italian degeneration. Biochem Biophys Res Hum Genet 2006; 120:139–143.
population. Ophthalmic Res 2001; Commun 2002; 295:668–672. 85. Kardys I, Klaver CC, Despriet DD, et al: A
33:325–328. 70. Tuo J, Smith BC, Bojanowski CM, et al: common polymorphism in the complement
55. Schmidt S, Saunders AM, De La Paz MA, The involvement of sequence variation and factor H gene is associated with increased
et al: Association of the apolipoprotein E expression of CX3CR1 in the pathogenesis risk of myocardial infarction: the Rotterdam
gene with age-related macular of age-related macular degeneration. Study. J Am Coll Cardiol 2006;
degeneration: possible effect modification FASEB J 2004; 18:1297–1299. 47:1568–1575.
by family history, age, and gender. Mol Vis 71. Jakobsdottir J, Conley YP, Weeks DE, 86. Fremeaux-Bacchi V, Kemp EJ, Goodship
2000; 6:287–293. et al: Susceptibility genes for age-related JA, et al: The development of atypical
56. Schmidt S, Klaver CC, Saunders AM, et al: maculopathy on chromosome 10q26. haemolytic-uraemic syndrome is influenced
A pooled case-control study of the Am J Hum Genet 2005; 77:407. by susceptibility factors in factor H and
apolipoprotein E (APOE) gene in age- 72. Rivera A, Fisher SA, Fritsche LG, et al: membrane cofactor protein: evidence from
related maculopathy. Ophthalmic Genet Hypothetical LOC387715 is a second two independent cohorts. J Med Genet
2002; 23:209–223. major susceptibility gene for age-related 2005; 42:852–856. 1899
RETINA AND VITREOUS

87. brera-Abeleda MA, Nishimura C, Smith JL, 90. Meyers SM, Zachary AA: Monozygotic degeneration in monozygotic twins and
et al: Variations in the complement twins with age-related macular their spouses in Iceland. Acta Ophthalmol
regulatory genes factor H (CFH) and factor degeneration. Arch Ophthalmol Scand 1999;77:422–425.
H related 5 (CFHR5) are associated with 1988;106:651–553. 95. Grizzard SW, Arnett D, Haag SL: Twin study
membranoproliferative glomerulonephritis 91. Dosso AA, Bovet J: Monozygotic twin of age-related macular degeneration.
type II (dense deposit disease). J Med brothers with age-related macular Ophthalmic Epidemiol 2003;10:315–322.
Genet 2006; 43:582–589. degeneration. Ophthalmologica 96. Seddon JM, Cote J, Page WF, et al: The
88. Gold B, Merriam JE, Zernant J, et al: 1992;205:24–28. US twin study of age-related macular
Variation in factor B (BF) and complement 92. Klein ML, Mauldin WM, Stoumbos VD. degeneration: relative roles of genetic and
component 2 (C2) genes is associated with Heredity and age-related macular environmental influences. Arch Ophthalmol
age-related macular degeneration. Nat degeneration. Observations in monozygotic 2005;123:321–327.
Genet 2006; 38:458–462. twins. Arch Ophthalmol 1994;112:932–937. 97. Assink JM, Klaver CC, Houwing-
89. Melrose MA, Magargal LE, Lucier AC: 93. Meyers SM: A twin study on age-related Duistermaat JJ et al: Heterogeneity of the
Identical twins with subretinal macular degeneration. Trans Am genetic risk in age-related macular disease:
neovascularization complicating senile Ophthalmol Soc 1994;92:775–843. A population-based familial risk study.
macular degeneration. Ophthalmic Surg 94. Gottfredsdottir MS, Sverrisson T, Musch Ophthalmology 2005;112:482–487.
1985;16:648–651. DC, Stefansson E: Age related macular
SECTION 10

1900
CHAPTER
Age-Related Macular Degeneration: Drusen and
144 Geographic Atrophy
Susan B. Bressler, Diana V. Do, and Neil M. Bressler

Age-related macular degeneration (AMD) is the leading cause of appear hyperfluorescent (brighter than the background fluor-
irreversible blindness in individuals age 55 and older.1 AMD escence), hypofluorescent (darker than the background fluor-
may be classified into a neovascular (exudative) form (discussed escence), or isofluorescent (unable to be distinguished from the
in Chapter 146) and a non-neovascular (nonexudative) form. background fluorescence).11 In a case series of 180 consecutive
The non-neovascular form features drusen and abnormalities eyes, small, hard drusen appeared as isofluorescent spots that
of the retinal pigment epithelium (RPE), such as geographic were difficult to discern from the background fluorescence.11
atrophy (GA), nongeographic areas of atrophy, and focal areas Small, hard drusen are not sufficient to diagnose AMD for
of hyperpigmentation within the macula. Clinical features, the following reasons:
clinicopathologic correlation, differential diagnosis, natural 1. The presence of at least one small drusen in the macula is
course, and treatment of the non-neovascular form of AMD nearly ubiquitous on fundus photographs3,6 and
are reviewed in this chapter. (The epidemiology of AMD is postmortem examination12 in individuals older than 40
discussed in Chapter 38.) years.
2. The incidence of small, hard drusen is not age-related.13
CLINICAL FEATURES AND 3. The presence of small drusen is not associated with an
CLINICOPATHOLOGIC CORRELATION increased risk of the development of the neovascular form
of AMD when compared with the risk associated with
large, soft drusen.4,14,15
DRUSEN
Multiple small, yellow-white lesions located within Bruch’s
membrane in the macula, commonly found in patients older
than 50 years, are called drusen. The term drusen is German
and is the plural for geode, a nodular stone with an interior
cavity lined with crystals.2 The relationship of drusen to AMD
may be somewhat confusing as drusen may vary greatly in
appearance and several types of yellow-white lesions may be
seen at the level of the RPE within the macula. These lesions
may be conveniently categorized into (1) small, hard drusen; (2)
large, soft drusen; and (3) cuticular (basal laminar) drusen.
Large, soft drusen, identified within two disc diameters of the
foveal center, are the form of drusen most commonly
considered to be a feature of AMD.

Small, Hard Drusen


Small drusen have been defined in most studies as having a
greatest linear dimension of less than 50 mm3,4 or less than
63 mm in diameter (Fig. 144.1).5–7 The latter definition has been
adopted, internationally, as the standard and was incorporated
into the Age-Related Eye Disease Study (AREDS) classification
of AMD.8,9 The borders of small drusen are almost always
distinct and well defined, contributing to the designation of
hard drusen.3,4
The fluorescein angiographic characteristics of drusen can
vary depending on their size, content, and RPE depigmentation
on their surface. Small drusen may appear as well-defined focal
spots of hyperfluorescence within the first few minutes of the
FIGURE 144.1. Hard drusen usually appear clinically as small yellow
angiogram.10 In the late frames these minute spots frequently punctate lesions at the level of the retinal pigment epithelium (RPE)
fade. Fluorescein angiography may also reveal the presence of with sharp discrete borders. These changes have been shown to
many more drusen than are apparent on slit-lamp bio- correspond to lipoid degeneration of a few discrete RPE cells without
microscopy. Indocyanine green angiography (ICG) of drusen evidence of diffuse thickening of the inner aspect of Bruch's
has demonstrated several patterns of fluorescence. Drusen may membrane throughout the macula. 1901
RETINA AND VITREOUS

4. Clinicopathologic correlation demonstrates that these 3. localized RPE detachments due to focal accumulation of
small lesions represent either a lipidization of a few RPE basal linear deposit in eyes without diffuse basal linear
cells16 or a localized accumulation (nodule) of hyaline deposits.18,19
material in the inner collagenous zone of Bruch’s
membrane,16 which can be otherwise totally normal on Basal linear deposit, also known as diffuse drusen or diffuse
either side of this nodule. The presence of these localized thickening of the inner aspect of Bruch’s membrane, lies
accumulations in an otherwise normal Bruch’s membrane external to the RPE basement membrane or within the inner
makes it unlikely that these small, hard drusen are a collagenous zone of Bruch’s membrane, in contrast to basal
feature of a diffusely dysfunctioning RPE–Bruch’s laminar deposit, which is situated between the plasma
membrane–choriocapillaris complex. membrane and the basement membrane of the RPE. These
deposits also differ in their ultrastructural characteristics. Basal
linear deposits consist of granular and vesicular lipid-rich
However, many small drusen may be a precursor lesion for the material and may contain widely spaced collagen, whereas basal
development of AMD, as the Chesapeake Bay Waterman Study laminar deposits consist mainly of widely spaced collagen.20
and the Beaver Dam Eye Study have each found that eyes with Clinically, one cannot detect the diffuse thickening of the inner
numerous small, hard drusen are at increased risk of developing aspect of Bruch’s membrane associated with basal linear or
soft or large drusen over time.13,17 The AREDS also reported basal laminar deposit; however, when there are areas of RPE
that roughly one-third of eyes with extensive small drusen hypopigmentation overlying this diffusely thickened Bruch’s
SECTION 10

progress to extensive numbers of medium size drusen or membrane or when this diffuse thickening weakens the inner
develop large drusen within 5 years; thus the presence of many aspect of Bruch’s membrane and predisposes it to separation,
small drusen do increase the chances in the long run that an eye or both, one recognizes these areas clinically as soft drusen
may develop more substantial evidence of AMD.15 (Fig. 144.2b).
Clinically, soft drusen are believed to be a feature of non-
Large, Soft Drusen neovascular AMD for the following reasons:
Large drusen have been defined in most studies as having 1. The incidence and prevalence of soft drusen have been
a dimension that is greater than or equal to 63 mm.5–8 The shown to be age related among many different population-
borders of large drusen are generally poorly demarcated, without based studies both nationally and abroad.3,6,13,17,21–25
sharp edges,3,5,8 contributing to the designation of soft drusen. 2. Soft drusen are associated with an increased risk of
Soft drusen are typically indistinct (Fig. 144.2a) meaning the development of RPE abnormalities,13 GA,13,15 and
density of the deposit decreases from center to periphery and choroidal neovascularization (CNV).4,13–15,25,26
the edges are fuzzy. In contast, some large drusen may be 3. Clinicopathologic correlation demonstrates that the
considered soft distinct drusen as they have a solid appearance presence of soft drusen represents diffuse thickening of the
with distinct edges, appreciable thickness, and a more uniform inner aspect of Bruch’s membrane throughout the
pigment distribution. macula.27,18
Large drusen vary in size, shape, and degree of confluence 4. In the neovascular form of AMD (see Chapter 146)
(merging of their borders) with neighboring drusen. Sizes of pathologic features, including CNV or disciform scarring,
‘large’ drusen have more recently been classified into 64 to are usually noted in eyes that also have diffuse thickening
<125 mm (medium), 125 to <250 mm (large), and ≥250 mm of the inner aspect of Bruch’s membrane throughout the
(very large). Histologically, three types of soft drusen have been macular region.27,18–20
identified:
1. localized detachments of RPE and basal linear deposit During fluorescein angiography staining of drusen (Fig. 144.2c
(diffuse drusen) in eyes with diffuse basal linear deposits, and 144.2d) may be noted in areas in which overlying RPE
2. localized detachments of RPE and basal laminar deposit in hypopigmentation allows increased visualization of choroidal
eyes with diffuse basal laminar deposits, and fluorescence or in areas in which fracturing of this diffuse

FIGURE 144.2. (a) Soft drusen are seen


clinically as large yellow lesions with
amorphous, ill-defined borders (arrow) at the
level of the RPE. (b) It is suspected that the soft
drusen noted clinically usually correspond to
areas of diffuse thickening of the inner aspect
of Bruch's membrane at the point at which
focal areas of RPE hypopigmentation have
developed in areas overlying this diffuse
thickening or at the point at which fracturing of
a b the diffusely thickened inner aspect of Bruch’s
membrane has separated from the remaining
outer aspect of Bruch's membrane. (c) and (d)
It is suspected that drusen may stain more
intensely (arrows) in the late phases of the
angiogram when this fracturing is present,
presumably because the fluorescein molecules
collect between the detached inner aspect and
the remainder of Bruch’s membrane.

c d
1902
Age-Related Macular Degeneration: Drusen and Geographic Atrophy

thickening provides a space for fluorescein molecules to pool of discrete absence or attenuation (depigmentation) of the RPE
(Fig. 144.2d). Some investigators have suggested that the overlying choriocapillaris atrophy such that larger-caliber
hydrophobic or hydrophilic properties of drusen, determined by choroidal vessels may be seen (Fig. 144.3a). The neurosensory
their lipid composition, may account for hypo- or hyper- retina overlying GA may appear thinned. GA represents the
fluorescence, respectively.28 However, this latter theory does not most advanced form of nonneovascular AMD. Although it is far
as yet have direct fluoroangiographic–pathologic correlation. less common than other manifestations of nonneovascular
Indocyanine green imaging of soft large drusen (≥125 mm) with AMD, when it involves the foveal center it accounts for 12–21%
indistinct margins may demonstrate hypofluorescent spots with of the cases of legal blindness attributed to AMD.30–32 Both the
a thin hyperfluorescent margin on the outside border of the incidence and the prevalence of GA are age-related.1,6,13,21,22
drusen in the early phase which persists in the late phase.11 The After the age of 70 prevalence of this nonneovascular AMD
thin hyperfluorescent margin may represent localization of feature rises sharply.
indocyanine green to the periphery of the drusen with poor Clinically, four patterns have been suggested to lead to GA in
penetration into the substance of the drusen. eyes that are presumed to have AMD:
More recently, drusen have been imaged by optical coherence 1. Areas of large, soft, confluent drusen may regress and lead
tomography (OCT) and morphologic changes may be present at to GA in multiple areas. These multifocal areas eventually
the level of the external highly reflective band that represents enlarge and coalesce with other similarly derived areas and
the RPE/Bruch’s membrane/choriocapillaris complex.29 In eyes forms a ring around the fovea.
with soft, large drusen, OCT may demonstrate irregularity in 2. Alternatively, the central macula may contain tiny areas

CHAPTER 144
the contour of the external highly reflective band consistent of reticulated hypo- and hyperpigmentation, which may
with accumulation of material within or beneath Bruch’s mem- progress to one large area of GA that spreads fairly
brane. In eyes with drusenoid pigment epithelial detachments, contiguously in a horseshoe pattern around the fovea,
a term generally used to describe a confluent mound of large eventually completely surrounding it leading to a
drusen, OCT shows focal elevation of the external highly reflective bull’s-eye maculopathy. After many years, doughnut-
band with moderately reflective material underneath this band. shaped areas of GA finally spread to include the foveal
center.
3. Spontaneous flattening of a serous pigment epithelial
ABNORMALITIES OF THE RPE detachment can also give rise to GA.
The development of diffuse thickening of the inner aspect of 4. Lastly, resorption of drusenoid pigment epithelial
Bruch’s membrane associated with the clinical recognition of detachments, can evolve to GA.33 Drusenoid pigment
soft drusen may also be accompanied by abnormalities of the epithelial detachments (PEDs) may be defined as elevated
RPE. These abnormalities include GA of the RPE, non-GA of mounds of large drusen or many confluent drusen that
the RPE (also known as RPE depigmentation), focal hyperpig- have coalesced. These lesions have well-defined borders,
mentation, and dystrophic calcification of Bruch’s membrane. are pale yellow to white in color, and have a minimum
diameter of 360 microns. Nearly half of the 139 eyes
Geographic Atrophy of the RPE monitored within AREDS that had drusenoid PEDs at or
GA of the RPE (also called areolar atrophy) consists of one to shortly after study entry developed central GA during
several areas, usually 175 mm or larger in diameter and circular, follow-up of up to 9 years.

FIGURE 144.3. Abnormalities of the RPE


secondary to age-related macular degeneration.
(a) An area of GA, with well-demarcated loss of
pigmentation of the RPE and overlying thinning
of the sensory retina with more apparent
underlying choroidal vasculature. (b) Calcified
drusen that correspond histologically to
dystrophic calcification at the level of the outer
retina. (c) Focal hyperpigmentation or pigment
clumps correspond to clumps of pigment at the
level of the RPE or within the outer aspects of
the sensory retina. (d) Non-GA refers to a
finding of tiny mottled areas of hypo- and
a b hyperpigmentation that may show some
thinning of the overlying sensory retina.

c d
1903
RETINA AND VITREOUS

Clinicopathologic correlation has shown replacement of of development of soft drusen,13 GA,13 and CNV.4,14,35,36 Some
soft drusen with fibrous tissue or dystrophic calcification.34 investigators hypothesized that areas of focal hyperpigmen-
The RPE overlying these areas ultimately disappears, producing tation may be associated with an increased risk of CNV because
small areas of GA. The underlying choriocapillaris may be the areas of hyperpigmentation represent disturbances of the
sclerosed, with thickening of the intercapillary septae.27 The RPE overlying existing occult CNV.37 However, clinico-
areas of GA are usually accompanied by loss of overlying photo- pathologic correlation has shown that the areas of focal
receptors, accounting for the visual loss that is noted using pigment can correlate with areas of subretinal or intraretinal
scanning laser ophthalmoscopy over these regions. Dystrophic pigment migration to the level of the photoreceptor nuclei,
calcification may accompany GA and appears clinically as overlying diffuse thickening of the inner aspect of Bruch’s
glistening, bright yellow specks within drusen that are under- membrane without evidence of CNV.19
going atrophy (Fig. 144.3b), contributing to the term calcified
drusen. Nongeographic Atrophy
Angiography of GA demonstrates early and discrete hyper- Non-GA of the RPE (also known as RPE degeneration or RPE
fluorescence of atrophic areas, presumably resulting from depigmentation) consists of areas of stippled, punctate hypopig-
increased transmission of choroidal fluorescence because of mentation and pigment mottling in which the underlying
hypopigmentation, attenuation, or lack of RPE. The chorio- choroidal vessels are not more readily apparent than in areas
capillaris may fill slowly or may be entirely absent within without non-GA, but in which the overlying sensory retina
these zones, and the size and shape of these areas do not appears to be thinned on stereoscopic examination (Fig. 144.3d).3
SECTION 10

change during the study. In the late frames, persistent staining In comparison to GA, areas of nongeographic atophy are less
of the areas of GA will be noted, owing to increased visibility well defined, less regular in shape, and less severe. Fluorescein
of the fluorescein staining of the choroidal and scleral tissue angiography of these areas reveals diffuse hyperfluorescence
through the atrophic RPE. OCT imaging of GA demonstrates with a pattern of reticular or punctate blockage corresponding
disruption, attenuation, or absence of the RPE/Bruch’s to the pigment clumping. Clinicopathologic correlation has shown
membrane complex with variable thinning of the overlying mottled areas of relative RPE hypopigmentation or atrophy
neurosensory retina due to loss of photoreceptors.29 In addition, overlying diffuse basal linear and basal laminar deposit.19 These
areas of GA have a highly reflective signal from the choroid areas are similar to the pathologic correlate of large, soft
corresponding to enhanced penetration and reflection of the drusen identified clinically, except the areas of non-GA have
signal from the choroid due to attenuation of the RPE/Bruch’s minute foci of hypopigmentation often interspersed with
membrane/choriocapillaris complex (Fig. 144.4). clumps of hyperpigmentation, whereas clinically apparent soft
drusen have broader areas of RPE hypopigmentation, but often
Focal Hyperpigmentation do not have clumps of hyperpigmentation within the drusen.
Focal hyperpigmentation of the RPE consists of areas or Incidence and prevalence rates of this AMD feature are
clumps of increased gray or black pigmentation at the level of also age dependent6,13,22,23 and eyes with large drusen are more
the outer retina or subretinal space (Fig. 144.3c). It may be likely to develop this characteristic than eyes without large
punctate, linear, or reticular. During fluorescein angiography, drusen.13 Similarly, eyes with non-GA are more likely to develop
areas of focal hyperpigmentation block background choroidal other features of AMD including soft drusen,13 GA,13,38 and
fluorescence. CNV.13,38
The incidence and prevalence rates of focal hyperpigmen-
tation increase with age,6,13,17,21–23 and eyes with soft or large
drusen are more likely to develop increased retinal pigment GRADING DRUSEN AND RPE
than eyes with no drusen or small drusen.13 Clinical studies ABNORMALITIES AND FORMING
have shown that eyes with focal clumps of hyperpigmentation CLINICALLY RELEVANT CLASSIFICATION
identified on color fundus photographs have an increased risk OF AMD
The Age-Related Eye Disease Study (AREDS), a prospective
FIGURE 144.4. multicenter randomized clinical trial involving 4757 partici-
(a) Fundus photograph pants designed to evaluate the effects of high-dose vitamin and
of GA that involves the mineral supplements on the development and progression of
center of the fovea and
AMD, has also supplied a wealth of natural history information
(b) corresponding
optical coherence about AMD.9,15,38 Features of nonneovascular AMD that impart
tomography a greater degree of risk of progression of AMD have been clearly
demonstrates a highly identified by performing detailed evaluation of baseline stereo-
reflective signal from scopic color fundus photographs of the macula and monitoring
the choroid patients annually with sequential color fundus photographs to
corresponding to document clinically relevant outcomes of advanced AMD such
enhanced penetration as foveal GA or CNV.
and reflection of the The AREDS system for classifying age-related macular
signal from the choroid
degeneration was based on reasonable quality film based
due to attenuation of
the RPE/Bruch's
stereoscopic color fundus photographs of the disc (field 1) and
a membrane/chorio- macula (field 2) of each eye of study participants.9 A standard
capillaris complex. grid template adapted from the Early Treatment Diabetic
Retinopathy Study composed of three circles concentric with
the center of the macula was used to identify the area of
interest.39 All features of AMD found within the outer circle
were recorded. The outer circle has a radius of 3000–3600 mm
b (two disc diameters using 1500–1800 mm for the standard disc).
1904 An additional standard template which consisted of a set of
Age-Related Macular Degeneration: Drusen and Geographic Atrophy

graduated circles was used to estimate maximum drusen size, • Intermediate AMD. Extensive medium drusen (occupying
maximum area occupied by drusen, and total area involved an area of at least 360 mm diameter circle, which is
by pigment abnormalities. Figure 144.5 illustrates the grading equivalent to 20 drusen) if the boundaries are indistinct or
grid and the graduated measurement circles with their occupying an area of 656 mm diameter circle (equivalent to
corresponding diameters. 65 drusen) if the boundaries are distinct or at least one
The detailed AREDS system for classifying AMD features was large druse (≥125 mm, approximately the width of a retinal
found to have good reproducibility with kappa statistics ranging vein as it crosses the optic nerve) or the presence of GA
from 0.54 to 0.88; however, this system was dependent on that spares the foveal center (nonfoveal GA).
photographs with at least fair quality read by trained and experi- • Advanced AMD. Geographic atrophy involving the center
enced readers.38 A simpler grading method to anticipate risk of of the fovea or CNV or disciform scar.
developing advanced AMD was also developed for use in clinical
practice (see section on Data from Cohorts of Participants with
Bilateral Non-Neovascular AMD and Progression to CNV or DIFFERENTIAL DIAGNOSIS
Foveal GA).40 Nevertheless, the AREDS system for grading non-
neovascular AMD features has led to a simple classification of A variety of maculopathies might be confused with the non-
AMD that can be easily adapted for clinical practice by neovascular features of AMD, and they should be differentiated
estimating the number of drusen of a particular size and the from drusen or abnormalities of the RPE. These other condi-
presence, type, and location of pigment abnormalities. tions often have a different prognosis from the non-neovascular

CHAPTER 144
features of AMD. A variety of factors, including demographic,
morphologic features, and distribution of the fundus and
Key Features angiographic changes, will help to differentiate these conditions
Age-Related Eye Disease Study Simplified Severity from AMD.
Scale
No = 0
Large drusen CUTICULAR (BASAL LAMINAR) DRUSEN
Yes = 1 1
Innumerable small, uniformly sized, discretely round, slightly
Right eye
raised, yellow subretinal lesions (Fig. 144.6a) that are best seen
No = 0 with angiography (Fig. 144.6b) and usually present in middle
Pigment abnormalities age (forties to sixties) are called cuticular (basal laminar)
Yes = 1 1 drusen.41–43 They may be differentiated from more typical soft
drusen in that retroillumination biomicroscopically demon-
No = 0 strates semitranslucency of innumerable similar–sized basal
Large drusen laminar drusen, as opposed to variable–sized, more typical soft
Yes = 1 1
drusen that appear opaquely yellow and are not semitranslu-
Left eye
cent. During fluorescein angiography, cuticular drusen will
No = 0 demonstrate early, bright, uniform hyperfluorescence compared
Pigment abnormalities with the variable, less bright hyperfluorescence of more typical
Yes = 1 1 soft drusen. With ICG angiography, basal laminar drusen may
appear as hyperfluorescent spots early in the study which

Large drusen & pigment Patient severity scale persist in the late phase of the angiogram.11
abnormalities = 4 risk factors The term basal laminar drusen should not be confused with
Severity Score (Risk Factors) – 5 Year Risk for Advanced the term basal laminar deposits, which refers to the
Both Eyes AMD in at Least 1 Eye wide–spaced collagen located between the plasma membrane
0 0.5% and the basement membrane of the RPE, or the term basal
1 3% linear deposits, which refers to the granular, electron–dense,
2 12% lipid–rich material seen ultrastructurally external to the basement
3 25% membrane of the RPE. In fact, the term basal linear deposits
4 50%
corresponds to the diffuse thickening of the inner aspect of
Scoring System Bruch’s membrane, which was described previously as diffuse
Assess whether large drusen and pigment abnormalities are
present in each eye and add the number of risk factors. The
drusen. Clinicopathologic correlation has shown that cuticular
number of risk factors correlates with the patient’s estimated risk drusen consist of an extremely thick inner aspect of Bruch’s
of progression to advanced AMD in five years. membrane with overlying nodular excrescences, all beneath the
RPE. Since no cuticle exists, and to avoid the use of the
confusing terms basal laminar and basal linear deposits, it has
AREDS has provided the following clinical classification which been proposed that the clinical features depicted in Figure
can be used to describe adults at risk for AMD or vision loss 144.6, typical of cuticular or basal laminar drusen be called
from AMD:9 diffuse drusen with overlying nodular excrescences to more
• No AMD. Absence of any drusen or presence of a few small accurately describe what they are.
drusen (<63 mm diameter drusen occupying <125 mm Patients with this unusual form of drusen may experience
diameter circle [equivalent to 5–15 small drusen]) in the pseudovitelliform detachments consisting of yellowish material
absence of any RPE abnormalities or any later stages of at the level of the outer retina. The material appears to obscure
AMD. details of the RPE, suggesting that it is present between the
• Early AMD. Extensive small drusen (occupies at least sensory retina and the RPE. During fluorescein angiography,
125 mm diameter circle), or nonextensive medium size these detachments show early hypofluorescence ( Fig. 144.6b),
drusen (63 to <125 mm diameter drusen) with or without presumably because of the ability of the yellowish material to
pigment abnormalities (increased pigment or block the underlying fluorescence of the choriocapillaris.
depigmentation) and no other later stages of AMD. Progressive staining of the yellowish material becomes apparent 1905
RETINA AND VITREOUS

a b

FIGURE 144.5. (a) Maculopathy grading grid used in the Age-Related Eye Disease Study. The grid is affixed to a fundus photograph of a left
eye to illustrate the three circles concentric with the center of the fovea and four radial lines in the 1:30, 4:30, 7:30, and 10:30 meridians. The
radius of the inner circle corresponds to 1/3 disc diameter (500 mm), the radius of the middle circle to 1 disc diameter (1500 mm), and the radius
of the outer circle to 2 disc diameters (3000 mm). (b) A set of standard measurement circles for estimating the area involved by various
abnormalities. Three groups of open circles are available and designated C for central, I for inner, and O for outer subfields. For each subfield,
SECTION 10

circle 1 corresponds to 1/64 of the subfield area and circle 2 to 1/16 of the area.
Reproduced from the Age-Related Eye Disease Study Research Group: The age-related eye disease study system for classifying age-related macular degeneration
from stereoscopic color fundus photographs: the AREDS report 6. Am J Ophthalmol 2001; 132:668–681.

FIGURE 144.6. Basal laminar or cuticular


drusen. (a) Color photograph shows
innumerable small, uniformly sized, discretely
round, and slightly raised yellow subretinal
lesions and dull yellow material in the central
macula referred to as a pseudovitelliform
detachment. (b) Angiogram helps highlight
basal laminar drusen. In addition,
hypofluorescence, presumably from the
associated pseudovitelliform detachment,
blocks the underlying fluorescence of the
choriocapillaris. (c) Progressive staining of the
pseudovitelliform detachment material
a b becomes apparent in the middle- and late-
transit phases of the angiogram. (d) With time,
the pseudovitelliform detachment can clear
spontaneously. Geographic atrophy will
sometimes develop with clearing.

c d

in the middle- and late-transit phases of the angiogram development of atrophy or rapid clearing associated with
(Fig. 144.6c), likely due to incompetence of the RPE’s zonula marked GA.
occludens, which fails to keep fluorescein from diffusing from These pseudovitelliform detachments should be differentiated
the choriocapillaris to the subsensory retinal space, with from true vitelliform detachments seen in Best’s disease (in
subsequent staining of the yellowish material. Pseudovitelliform which the electrooculogram will be abnormal) and from pattern
detachments appear as a mound of highly reflective material dystrophies of the RPE that may show pseudovitelliform-like
under the neurosensory retina without any adjacent subretinal detachments.
fluid on OCT imaging.29 This highly reflective substance Patients with basal laminar or cuticular drusen have a
appears to be located between the retina and RPE. diseased Bruchs membrane and are at risk of development of
Since the fluorescein hyperfluorescence of a vitelliform CNV. Therefore, careful scrutiny of any bright hyperfluores-
lesion may mimic CNV, one must recognize its appearance cence that appears to leak in these patients is needed to deter-
in order to avoid unnecessary delivery of treatment intended mine whether the features are due to the presence of CNV or to
to manage CNV. The natural course of these detachments can progressive staining of the pseudovitelliform detachment.
1906 be spontaneous clearing (Fig. 144.6d) with extremely slow Sometimes this differentiation is extremely difficult to make.
Age-Related Macular Degeneration: Drusen and Geographic Atrophy

been observed both within families and sporadically. Although,


DOMINANT DRUSEN IN YOUNG INDIVIDUALS they are often located in the center of the macula, they may
Patients in their teens or twenties may present with large present eccentrically (Fig. 144.9), accounting for the variety
(>63 mm), discrete nodular drusen. Typically these drusen are of appearances on presentation. The pseudovitelliform detach-
bilateral and in a very symmetric distribution between the two ments will not show disruption or layering of the yellow
eyes, such as temporal to the fovea. Even the young children of pigment dependently, as is seen in the vitelliform lesions of
these individuals, those under age 10 years, may have discrete Best’s disease.
drusen (Fig. 144.7). In the experience of the authors, these Clinicopathologic correlation of pattern dystrophies of the
patients may go years without CNV or atrophy developing RPE has shown a thick layer of slightly granular, eosinophilic,
and therefore suffer no significant visual loss. When these periodic acid-Schiff (PAS)-positive material lying between a
lesions are in the macula, they could presumably cause focal thinned atrophic RPE and Bruch’s membrane, with central
disturbances of Bruch’s membrane and may increase the risk pigment clumping from large pigment-laden cells and extra-
of the development of CNV but there are no natural history cellular melanin pigment lying between the sensory retina and
studies of this condition to date. Bruch’s membrane.45 A recent (unpublished) clinicopathologic
correlation of an eye with pattern dystrophy and pseudovitel-
liform lesion demonstrated lipofuscin at the outer retinal level
PATTERN DYSTROPHY OF THE RPE (ADULT to account for the yellow deposit. The diffuse disturbance of
VITELLIFORM DYSTROPHY, ADULT-ONSET Bruch’s membrane histologically presumably places these

CHAPTER 144
FOVEAL PIGMENT EPITHELIAL DYSTROPHY, patients at increased risk of development of CNV through
BUTTERFLY-SHAPED PIGMENT DYSTROPHY, breaks in the outer aspect of Bruch’s membrane.
RETICULAR DYSTROPHY OF THE PIGMENT More recently, OCT imaging and correlation with fluore-
scein angiographic features of eyes with pseudovitelliform
EPITHELIUM) lesions has provided additional morphologic information on
Patients with pattern dystrophy of the RPE will show a the location of the yellow deposit. In sixteen eyes with adult-
reticulated pattern of pigmentation, usually fairly symmetric onset foveomacular vitelliform dystrophy, the yellow foveal
between the two eyes and often without the presence of more lesion demonstrated early hypofluorescence on fluorescein
typical soft drusen (Fig. 144.8).44 These dystrophies may have a angiography with staining and absence of leakage in the late
yellow deposit at the level of the outer retina (pseudovitelliform frames.46 Corresponding OCTs in these eyes revealed a hyper-
detachment), often with a central area of greenish hyper- reflective structure located between the photoreceptors and
pigmentation (sometimes best seen with transillumination RPE. The retina overlying the hyperreflective structure was
of the yellowish material), and occasionally surrounded by a raised by the pseudovitelliform lesion with subsequent loss
petaloid pattern of hyperpigmentation, which is more obvious of the normal foveal depression but the RPE layer was not
on fluorescein angiography (Fig. 144.8b). These lesions have elevated. In five eyes in which fluorescein angiography

a b c

FIGURE 144.7. Dominant drusen. (a) A young woman in her twenties demonstrates typical large drusen, some of which have discrete
boundaries. Similar findings are noted in her 9-year-old son (b) and her 6-year-old son (c).

FIGURE 144.8. Pattern dystrophy of the RPE.


(a) Typical pattern dystrophy in which blocked
fluorescence on angiography corresponds to
pigment clumping or greenish discoloration
seen at the level of the RPE, surrounded by
hyperfluorescence corresponding to dull
yellowish material that has sometimes been
termed a pseudovitelliform detachment. (b) The
angiogram highlights not only the blocked
fluorescence but also very prominent staining
with persistent bright hyperfluorescence in the
late phase of the angiogram, but no leakage.
The absence of leakage helps confirm the
absence of choroidal neovascularization.
a b
1907
RETINA AND VITREOUS

Key Features
The Age-Related Eye Disease Study Clinical
Classification of Age-Related Macular Degeneration
(AMD)
• No AMD. Absence of any drusen or presence of a few small
drusen (<63 µm diameter drusen occupying <125 µm diameter
circle (equivalent to 5–15 small drusen)) in the absence of any
RPE abnormalities or any later stages of AMD
• Early AMD. Extensive small drusen (occupies at least 125 µm
diameter circle), or nonextensive medium size drusen
(63 to <125 µm diameter) with or without pigment
abnormalities (increased pigment or depigmentation) and
no other later stages of AMD
• Intermediate AMD. Extensive medium drusen (occupying
an area of at least 360 µm diameter circle (equivalent to
20 drusen) if the boundaries are indistinct or occupying an
area of 656 µm diameter circle (equivalent to 65 drusen) if the
SECTION 10

boundaries are distinct) or at least one large druse (≥125 µm,


approximately the width of a retinal vein as it crosses the optic
nerve) or the presence of GA that spares the foveal center
FIGURE 144.9. Pattern dystrophy of the RPE in which the blocked
(nonfoveal GA)
fluorescence and hyperfluorescence occur in a multifocal distribution
• Advanced AMD. Geographic atrophy involving the center of
outside of the foveal center.
the fovea or CNV or disciform scar
demonstrated persistent central hypofluorescence in the late
frames corresponding to the yellow deposit, OCT revealed no
hyperreflective structure between the photoreceptors and Data from Fellow Eye Studies and Progression
RPE layers. In these eyes, OCT demonstrated focal thickening to CNV
of the hyperreflective RPE layer but no distinguishable Prior to the AREDS much of the detailed information generated
substance between the RPE and retina. Further studies are on the natural course of eyes with non-neovascular AMD had
needed to confirm these observations. been acquired by studying the fellow eyes of patients with
unilateral neovascular maculopathy that have participated in
the Macular Photocoagulation Study (MPS) Trials. In these
BULL’S-EYE MACULOPATHY investigations, a standardized classification was used to grade
A variety of conditions may produce a bull’s-eye maculopathy macular characteristics on the baseline fundus photographs
(usually referring to an area of central pigmentation of the of the eyes with nonneovascular AMD. Fundus photograph
retina surrounded circumferentially by an area of relative readers had no knowledge of the subsequent course of these
hypopigmentation and sometimes surrounded, once again eyes. In the 5 year prospective study of fellow eyes of partici-
circumferentially, by an area of increased pigmentation). This pants in the Extrafoveal AMD Trial of the MPS, eyes with no
condition may progress to GA of the RPE, which typically is large drusen and no focal hyperpigmentation were at a 10% risk
similar to the GA seen in AMD.47 These conditions usually of developing CNV within 5 years.4 Eyes with large drusen or
can be differentiated from GA associated with AMD. The bull’s- focal hyperpigmentation had a 30% risk of developing CNV, and
eye maculopathies result in a central area of GA with no eyes with both large drusen and focal hyperpigmentation had
associated soft drusen, whereas GA in AMD is usually about a 60% risk of acquiring CNV within 5 years.4 Subsequent
multifocal, with preservation of the fovea until the very late analyses on a larger number of fellow eyes, ~ 670, among
stages. In addition, the bull’s-eye maculopathies occur in early the individuals participating in the Juxtafoveal and Subfoveal
or midlife, whereas GA is seen most often in patients in their AMD Trials of the MPS have confirmed the heightened risk
late 70s and 80s, with increasing prevalence into the 90s. The associated with large drusen and have permitted more precise
causes of some of these maculopathies include central areolar quantification of that risk. In these subsequent studies, fellow
choroidal–RPE dystrophy, the Bardet–Biedl syndrome, con- eyes with large drusen had a 46% chance of developing CNV,
centric annular macular dystrophy, chloroquine retinopathy, those with focal pigment had a 38% chance, and those eyes with
cone dystrophy, fenestrated sheen macular dystrophy, the both factors were at greatest risk with incidence rates of 73% at
Hallervorden–Spatz syndrome, and fundus flavimaculatus. 5 years.14 Systemic hypertension and numerous drusen (≥5)
were also found to be independent risk factors; an individual
NATURAL COURSE with hypertension, numerous and large drusen, and an area
of pigment had an 87% risk of progressing to CNV within
5 years.14 The development of severe visual loss in these fellow
DRUSEN AND RPE ABNORMALITIES eyes with nonneovascular AMD at baseline occurred almost
The natural course of an eye with non-neovascular AMD may exclusively in the eyes in which CNV developed during follow-
ultimately lead to foveal GA, CNV, or both forms of advanced up. Among the fellow eyes that did not develop CNV, the
AMD. The probability that an eye with non-neovascular average visual acuity loss over the 5 year follow-up period was
AMD will progress to CNV depends on whether the fellow only 0.4 lines.48
eye in that individual has non-neovascular AMD in the absence In 2001, the AREDS Group reported on the risk of prog-
of CNV or disciform scarring or whether the eye with drusen ression to advanced AMD among individuals who were
or RPE abnormalities, or both, is the fellow eye of a person assigned to the placebo group.15 The risk of progression to
whose contralateral eye has already developed the neovas- advanced AMD (CNV or foveal GA) within 5 years was 1.3%
1908 cular form of AMD. for individuals with early AMD, 18% for individuals with
Age-Related Macular Degeneration: Drusen and Geographic Atrophy

intermediate AMD, and 43% for the remaining eye (please refer to Key Features: Age-Related Eye Disease Study
among individuals with unilateral advanced AMD (fellow Simplified Severity Scale on page 5). For example, an individual
eyes). Among AREDS participants who had neovascular AMD with large drusen in the right eye and focal pigment in the left
in their first eye affected with advanced AMD about 37% eye would generate a person score of 2, whereas the individual
progressed to neovascular AMD in their second eye during with bilateral large drusen and bilateral pigment abnormalities
a mean follow-up period of 6.2 years. Three ocular risk would have a person score of 4. The scale can be adapted for use
factors (largest drusen size, total area occupied by drusen, and in persons with unilateral advanced AMD by scoring the eye
pigmentary abnormalities) were identified as imparting with advanced AMD as 2 and then continuing on to the
increased risk that a fellow eye would develop CNV. Logistic contralateral eye to assess features (1) and (2). The range of
regression analysis adjusted for age, gender, and AREDS person scores is 0–4. The 5 year risk for developing advanced
treatment assignment was used to predict development of AMD is 0.5% for individuals with a score of 0, 3% for a score of
CNV in the second eye. The strongest predictive factor for 1, 12% for a score of 2, 25% for a score of 3, and 50% for a score
fellow eye CNV development was total drusen area, however, of 4. The AREDS simplified scale40 and the nine-step severity
substitution of largest drusen size and presence of focal scale38 both confirm previous observations made in the MPS
hyperpigment had nearly the same predictive value. Fellow fellow eye studies: drusen size and pigmentary abnormalities
eyes with only small drusen with or without focal pigment are independent risk factors for progression to the advanced
were at relatively low risk for CNV with rates of 5% over stage of AMD.
5 years; whereas fellow eyes with very large drusen and In addition to the MPS and AREDS, several large population-

CHAPTER 144
pigment were at the highest risk of approximately 50% for based studies have also described the evolution of non-
incident CNV during follow-up.49 Treatment with high-dose neovascular AMD fundus manifestations. In the Beaver Dam
antioxidants and zinc resulted in an overall 25% risk Study, 7–12% of eyes with intermediate or large drusen
reduction for development of advanced AMD in individuals demonstrated new areas of drusen (progression) and 10–33%
who had intermediate or unilateral advanced AMD. The had areas of drusen disappear (regression) over a 5 year
greatest absolute reduction in risk was identified among the period.13 Eyes with RPE abnormalities also showed significant
fellow eyes of participants with unilateral advanced AMD. change over time with 32–39% developing additional areas of
pigment and 28% demonstrating less involvement during the 5
Data from Cohorts of Participants with Bilateral year period.13 The Chesapeake Bay Waterman follow-up study
Non-Neovascular AMD and Progression to CNV had also found drusen and RPE abnormalities to progress in
or Foveal GA some and spontaneously regress in others.17 Regarding risk
In addition to evaluating the use of micronutrient supplements factors for progression, Beaver Dam eyes with large drusen
for AMD, another goal of the AREDS was to develop an AMD (≥125 mm) were more likely to develop increased drusen
severity scale that could provide baseline risk categories to area, confluence, focal hyperpigmentation, non-GA, GA, or
track progression of AMD in an orderly fashion to appro- neovascular AMD, than were eyes with smaller drusen.13
ximate risks of progression to advanced AMD. Furthermore, Eyes with RPE abnormalities were more likely to develop
development of a severity scale might aid in providing soft drusen, increased drusen area, confluent drusen, GA, or
surrogate outcomes for advanced AMD. A nine-step severity neovascular AMD compared with eyes without pigmentary
scale was created that combines six progessive levels of total abnormalities.13 Ten-year data from the Beaver Dam eye study
drusen area and five progressive levels of pigmentary also confirmed that eyes with soft indistinct drusen or
abnormalities (increased pigment, depigmentation, and non- pigmentary abnormalities at baseline were more likely to
foveal GA).38 Five-year rates of progression to advanced AMD develop advanced AMD at follow-up than eyes without
were as low as less than 1% in step 1 (eyes with minimal these lesions (15.1% vs 0.4% for soft indistinct drusen and 20%
drusen area and absence of pigment abnormalities) of the vs 0.8% for pigmentary abnormalities).51 Therefore, charac-
scale to ~50% in step 9 (eyes with variable drusen area in the teristics of large drusen and pigmentary abnormalities appear
presence of nonfoveal GA) of the scale. The risk of neo- to confer increased risk of developing more advanced AMD
vascular AMD increased from steps 1 through 8 in the scale and vision loss, whether found in an individuals with
and then decreased in step 9. The risk of foveal GA was low bilateral nonneovascular disease or individuals with uni-
from steps 1 through 5 and then increased steadily from steps lateral neovascular maculopathy. Furthermore, these observa-
5 through 9, an area of the scale that is associated with a greater tions have been made in prospective clinical trials and
degree of pigmentary abnormalities. Additional AREDS confirmed in population based studies.
analyses identified presence of calcified drusen as another The Blue Mountains Eye Study also explored the incidence
independent risk factor for development of GA.50 of advanced AMD among a population based sample in
Although this nine-step severity scale may have utility in a Australia.52 In this cohort, the 5 year incidence of developing
research setting this scale does not lend itself to use in clinical neovascular AMD or GA (foveal or nonfoveal) was 1.1%. Several
practice. Therefore, the AREDS Group developed a simplified ocular risk factors were associated with progression to
scale for clinical use to predict risk for progression to advanced advanced AMD; among right eyes that had non-neovascular
AMD. The simple scale was formulated on several observa- AMD at baseline, the following features conveyed risk: large
tions made during the construct of the research severity scale, drusen (≥125 mm, age-adjusted relative risk [RR] 5.7 [95%
notably: (1) there was a strong association between drusen confidence interval (CI) 3.6–9.0]), indistinct soft drusen (RR 9.9
area and largest drusen size; (2) there was a very low frequency [95% CI 6.4–15.4]), location of drusen within 1500 mm of the
of RPE depigmentation and/or GA in the absence of increased foveal center (RR 11.6 [95% CI 6.7–21.1]), area involved by
pigment; and (3) bilateral large drusen is a stronger risk large drusen equal to at least one half of the optic disc
factor than unilateral large drusen. In the simplified scale, (RR 13.5 [95% CI 8.0–22.8]), and presence of hyperpigmen-
clinicians are asked to assess the presence of only two tation (RR 8.0 [95% CI 5.4–11.9]). In addition, eyes classified
fundus characteristics: (1) large drusen and (2) pigmentary as having AREDS category 3 (intermediate AMD) or
abnormalities (hyperpigmentation or hypopigmentation, or 4 (unilateral advanced AMD) at baseline were 3.9 times (95%
nonfoveal GA).40 Each eye of a person is graded separately for (1) CI 2.3–6.7) more likely to develop advanced AMD over the
and (2) and for each factor present the eye receives a score of 1 5 year period compared to eyes with category 1 or 2. 1909
RETINA AND VITREOUS

The Rotterdam Study, another population-based cohort AMD. Earlier studies have tried to learn similar information by
performed in the Netherlands, also evaluated the incidence of identifying eyes with GA and evaluating case files retro-
advanced AMD. The 2 year cumulative incidence of advanced spectively. One area of interest has been quantifying the rate of
AMD (neovascular AMD or GA) was 0.2% among all study enlargement of the atrophic areas.34,55,56 Schatz and McDonald
participants, although this incidence increased to 1.8% among retrospectively reviewed 50 eyes with GA; 40% had multifocal
participants 85 years of age and older.53 Baseline fundus regions of GA to study.55 The average greatest horizontal linear
features that increased the odds that an eye would progress to dimension of the GA was 509 mm at presentation; although the
advanced AMD included more than 10% of the macular area range was 200–5300 mm. This dimension expanded at an average
covered by drusen (odds ratio (OR) 5.7, 95% CI 2.9–11.3), rate of 139 mm/yr (~1/10 of a disc diameter). Patients under age
presence of depigmentation (OR 4.0, 95% CI 2.5–6.4), presence 75 years tended to progress faster than patients 75 years and
of hyperpigmentation (OR 3.4, 95% CI 2.1–5.4), and ≥10% over, but great variability was noted in both the magnitude and
drusen confluence (OR 2.5, 95% CI 1.7–3.8). Although the the direction of progression in the cases reviewed. Sarks and
incidence of advanced AMD is quite low in this population colleagues reported on a series of 208 eyes with GA.34 They
given the limited 2 year follow-up, the results suggest again that noted that GA tended to start outside the foveal center, and as
drusen area and pigmentary abnormalities are important risk the atrophy encroached within 750 mm of the foveal center, it
factors for progression to advanced stages of AMD. approached one disc diameter. Total involvement of the fovea
More recently, the Copenhagen City Eye Study described the occurred only in the later stages, such that the average affected
incidence and progression of many AMD features.54 In this area in eyes with complete GA of the fovea measured more than
SECTION 10

population-based cohort study, 359 persons of the original 946 seven disc areas (DAs). Visual acuity was related to the per-
subjects were able to participate in the 14 year follow-up centage of fovea affected, but it varied widely, such that it was
examination. Older subjects at baseline were more apt to difficult to predict visual acuity within a narrow margin of
develop features of AMD at follow-up. Among subjects 75–80 certainty from the anatomic appearance of the atrophy alone.
years of age at baseline, medium size drusen were 1.8 times as Nevertheless, central fixation tended to be lost when atrophy
likely to develop and large drusen were 3.5 times as likely to occupied 85% or more of the fovea.
develop compared with subjects 60–64 years of age. In addition, Patients with GA often have bilateral and symmetric disease,
the incidence of soft drusen increased significantly with age although there may be differences in onset and progression
from 21.4% among persons aged 60–64 years to 45.2% among rates between the two eyes.34,55,56 One-half of the subjects in
persons aged 75–80 years. Pigmentary abnormalities also Sarks and colleagues’ study had bilateral GA,34 whereas 13% of
increased with age such that persons aged 75–80 years were 1.8 the Beaver Dam population-based study participants with pure
times as likely to have hyperpigmentary abnormalities GA had bilateral involvement at baseline.6 However, individuals
compared to persons aged 60–64 years. The risk of developing with uniocular GA at baseline were at increased risk of devel-
the advanced stage of AMD was strongly associated with the oping GA in their fellow eye by the time of the 5 year follow-up
presence of soft indistinct drusen (OR 12.2, 95% CI 3.2–47.0), examination. Among the 12 patients with uniocular GA at the
increased drusen size (63–124 mm: OR 5.0, 95% CI 1.7–14.4; initial examination, three (25%) developed GA during the 5 year
>125 mm: OR 7.6, 95% CI 1.0–20.2). follow-up. After 10 years of follow-up, individuals with
uniocular GA at baseline were 6.3 times (95% CI 0.9–42.1) as
NATURAL COURSE OF GEOGRAPHIC likely to develop advanced AMD in the uninvolved, fellow eye
ATROPHY: DEVELOPMENT AND as those individuals with early AMD in both eyes at baseline.
Sunness and coworkers prospectively followed 74 patients
PROGRESSION with bilateral GA and documented median total area of
The natural progression of non-neovascular AMD fundus involvement of 4.4 DA at presentation (range 0.1–27.7 DA).56
features to GA has been described in several population-based There was a strong correlation between the two eyes of a patient
studies (as summarized above); however, additional information for the size of the atrophic area. The median rate of spread was
gathered from serial color fundus photographs from AREDS 0.73 DA/yr, taking into account changes in all dimensions of
participants has provided greater detail on predisposing an atrophic area. The median absolute difference in rates of
anatomic features and the sequence of events leading to the progression between eyes of a patient was 0.38 DA/yr.
formation of GA. Among a cohort of 95 eyes from two AREDS Central visual acuity loss may be gradual and progressive in
clinical centers in which GA developed at least 4 years after eyes with GA, with the degree of impairment directly related to
their initial study evaluation (range 4–11 years), the following foveal involvement.57 Since GA involves the foveal center late in
lesions were determined to have been present at the site of the course of the disease, visual acuity is a poor guide to the
future GA formation: drusen 95/95 eyes (100%), large drusen extent of the atrophy or visual impairment a patient may have.
(>125 mm) 91/95 eyes (95.8%), focal hyperpigmentation 91/95 In a study on 83 patients with bilateral GA, with a median
eyes (95.8%), confluent drusen 89/95 eyes (93.7%), very large visual acuity of 20/54 in the better-seeing eye, the majority
drusen (>250 mm) 79/95 eyes (83.2%), hypopigmentation 77/95 reported difficulty reading, a need to use low-vision devices,
eyes (81%), and calcification 20/57 eyes (35%).33 The average difficulty seeing in a dim environment, trouble recognizing faces,
interval from the time of appearance of these specific lesions to and an awareness of a blurry area.58 These symptoms remained
the development of GA was >6.5 years for drusen or confluent frequent even in a subgroup of 39 patients with visual acuity
drusen, 4.0 years for hyperpigmentation, 2.3 years for hypopig- better than 20/50 (median 20/35). Scanning laser ophthal-
mentation, and 1.5 years for calcification. Among this subset moscope testing demonstrated dense scotomas in all patients;
of AREDS participants, the sequence of events leading to GA therefore, GA-induced scotomas may have a significant impact
appeared to be accumulation of very large, confluent drusen, on visual function even when they do not involve the foveal
followed by hyperpigmentation appearing at the site and then center.
regression or fading of drusen with loss of hyperpigmentation Although GA-induced scotomas impair visual function,
leading to hypopigmentation and then formation of GA. patients with GA often develop an eccentric retinal locus for
Several authors have concentrated on eyes that already fixation. A prospective natural history study of patients with
manifest GA and monitored them prospectively to learn more central GA has demonstrated that 77% of eyes had an eccentric
1910 about the natural course of this feature of non-neovascular preferred retinal locus for fixation at baseline.59 After a median
Age-Related Macular Degeneration: Drusen and Geographic Atrophy

follow-up period of 5.3 years, 91% of eyes had an eccentric are often fairly subtle, such that careful scrutiny of these
preferred retinal locus for fixation with 81% of eyes retaining angiograms is needed, particularly with reference to previous
the baseline preferred retinal locus. The most common fixation studies, to determine whether or not CNV has developed.
patterns were fixation with the scotoma to the right and fixation
with the scotoma superior. Eyes that fixated with the scotoma MANAGEMENT
to the left tended to have lower reading rates (reading speed)
than eyes that fixated with right or superior patterns. The risk of visual loss in eyes with drusen or pigment abnor-
The precise rate of visual acuity loss in patients with GA has malities results from the development of advanced AMD either
recently been evaluated prospectively in 74 patients with visual in the form of CNV or foveal GA. Accordingly, management is
acuity of 20/50 or better at presentation.60 Rates of vision loss aimed at preventing CNV or GA. There has been speculation
over a 2 year period are substantial, with 50% doubling their that ultraviolet or visible light exposure may lead to the
visual angle (a loss of three or more lines of acuity) and 25% generation of reactive oxygen species in the outer retina,64,65
quadrupling their visual angle (a loss of six or more lines of which may in turn cause lipid peroxidation of photoreceptor
acuity). Furthermore, these patients presented with reduced outer segment membranes potentially contributing to the devel-
reading speeds, and this visual function significantly deteri- opment of AMD. However, observational epidemiologic studies
orated during the 2 year follow-up period as well. Therefore, all have failed to provide support for the theory that cumulative
eyes with GA, even those with a parafoveal location, are at high sunlight exposure is associated with AMD. No association was
risk of significant visual impairment within a few years of identified between cumulative ultraviolet or visible light

CHAPTER 144
presentation. exposure and the presence of non-neovascular AMD in the
Patients with GA report gradual progression of their vision Chesapeake Bay Waterman Study.66 However, there was a
loss, even when a more precipitous loss of visual acuity is significantly higher exposure to blue light and all wavelengths
documented between visits as an atrophic area enlarges and in the visible spectrum within the 20 year period immediately
involves more of the foveal center. Generally, patients with GA preceding study participation among the eight participants with
will use central fixation if there is a small foveal region without advanced AMD in the cohort when compared with age-matched
atrophy and eccentric fixation when the fovea is totally control subjects.67 The Eye Disorder Case-Control Study also
involved. However, as the atrophy progressively encroaches on failed to demonstrate any positive relationship between lifetime
the foveal center, these patients may go through a transitional sunlight exposure and the presence of neovascular AMD.68 A
period in which they alternate between central and eccentric limited association between light and AMD was noted in the
fixation, and this gradual transfer of the fixation focus may Beaver Dam population, with increased outdoor summertime
explain the lack of perception of a sudden alteration in acuity.61 exposure being associated with increased retinal pigment among
Alternatively, the explanation may lie within the nature of the male subjects and late AMD in male and female subjects.69
residual central vision in eyes with preservation of small foveal Thus, the available data are inconsistent and inconclusive in
islands. As the foveal island progressively decreases in size, the regard to a relationship between light and AMD. Since the
size of the central visual field gets progressively smaller, such Waterman Study has shown that ultraviolet light is associated
that fewer words or facets of the visual image fit into the with lenticular opacities70 and because sunglasses are inex-
functional region, even though relatively good acuity with pensive and associated with few side effects, it seems reasonable
letters may be preserved. At the point at which the patient not to discourage their use.
transfers fixation to an eccentric locus, the amount of useful Multiple epidemiologic studies have reported positive asso-
functional central retina may not provide better vision than the ciations between cigarette smoking71,72 and non-neovascular
eccentric region. AMD, so possible prevention of AMD would seem to be yet
GA appears to act as a barrier for CNV, such that CNV does another reason for physicians to recommend cessation of
not begin within and rarely progresses through atrophic areas. smoking. Although there are inconsistent data from epidemio-
However, CNV may occur contiguous to these regions and logic studies with regard to an association between hyper-
course along the external perimeter of the area. Since patients tension, cardiovascular disease, and increased cholesterol and
with progressive GA rarely complain of abrupt changes in visual saturated fat intake31,68,73–75 and AMD, it would seem
acuity, any such change should prompt an investigation for the reasonable to recommend routine medical examinations with a
development of CNV. Forty-five to 49% of the fellow eyes with generalist physician to monitor blood pressure, cholesterol levels,
non-neovascular maculopathy and zones of GA at study entry and treatable cardiovascular disease in this elderly population.
into the MPS AMD trials of laser photocoagulation progress to AMD has been identified as having a familial component
CNV within 5 years.14,48 Among 45 patients with unilateral GA based on clustering of the disease within multiple family
and a fellow eye with neovascular maculopathy followed members within different generations and greater concordance
prospectively for natural history of GA, 23% developed CNV at rates among monozygotic versus dizygotic twins.76–78 These
the edge of GA within 2 years, whereas CNV did not develop in observations fuel the search to identify specific genetic
any of the 92 patients with bilateral GA during the same time components of AMD. Several investigators have identified a
interval, suggesting that patients with bilateral GA without common variant (Y402H) of the complement factor H gene
evidence of CNV are at low risk for developing CNV.62 (CFH), located on chromosome 1q31, as a major AMD risk
The development of subretinal hemorrhage in eyes with GA gene.79–81 This finding further suggests that inflammation may
does not necessarily imply the presence of CNV. Nasrallah and play an important role in the pathogenesis of AMD as CFH is a
associates reported on eight patients in whom small subretinal component of the immune system. CFH helps to regulate the
hemorrhage spontaneously cleared over a 15 month period body’s inflammatory response by protecting against uncon-
without evidence of CNV at the time of the hemorrhage nor trolled complement activation, and the Y402H variant is located
evidence of CNV or disciform scarring with clearing of the within the binding sites for heparin and C-reactive protein.
hemorrhage.63 These subretinal hemorrhages may reflect rupture In a retrospective case-control study, the CFH variant was
of normal choriocapillaris, as is seen in subretinal hemorrhages associated with an increased risk of developing GA (OR 3.22,
occurring in myopic patients with lacquer cracks in which CNV 95% CI 1.87–5.55) as well as neovascular AMD (OR 2.50, 95%
is not growing through the lacquer crack. However, fluorescein CI 1.74–3.60) compared to controls with no AMD.82 Further
angiographic signs of active CNV in patients with extensive GA investigation is needed to determine how this gene variant may 1911
RETINA AND VITREOUS

increase the risk of AMD. In time, this may lead to new unsaturated fatty acids (eicosapentaenoic acid (EPA) and
screening strategies to identify individuals with greater risk for docosahexaenoic acid (DHA)). Evidence supporting the perfor-
development or progression of AMD. Furthermore, identifica- mance of AREDS 2 involves observational data from many
tion of the role this gene variant may play in pathogenesis may epidemiologic studies as well as case-control studies within
open possibilities for new management strategies to prevent AREDS which identify individuals who consume higher amounts
progression. of food sources rich in these ingredients to have a reduced odds
In a small pilot trial, oral zinc administration was associated or risk of manifesting varying forms of AMD when compared to
with less vision loss in patients with non-neovascular AMD individuals who have lower exposures. AREDS 2 is not expected
when followed for a period of up to 2 years.83 However, a ran- to reach any conclusions for at least 5–8 years from now. Until
domized prospective trial evaluating oral zinc administration AREDS 2 reports its findings, the safety and efficacy of lutein/
versus placebo in decreasing vision loss and development of zeathanine and fish oil in the management of AMD will remain
CNV in 112 patients at risk of neovascular AMD in their unknown.
second eye did not find treatment to be beneficial.84 Exploring Another form of treatment that has been extensively evaluated
the hypothesis that AMD may be caused by cumulative in the management of eyes with nonneovascular AMD to
oxidative insults, cross-sectional and case-control epidemiologic potentially alter the course of the disease is photocoagulation,
investigations have evaluated blood antioxidant levels, dietary either directly or adjacent to soft drusen. Several clinical trials
ingestion of antioxidant-containing food sources, and uses of have now been completed and have not found this treatment to
micronutrient supplements.73,85–88 These studies have suggested be beneficial in the management of patients with soft drusen.
SECTION 10

a small to moderate protective role of antioxidants on varying The Choroidal Neovascularization Prevention Trial (CNVPT),
levels of AMD; however, the results for specific nutrients and halted recruitment when it was recognized that study eyes in
specific AMD types have been inconsistent and therefore the fellow eye group randomly assigned to laser treatment had
inconclusive. These types of studies also have several important a significantly greater rate of CNV formation within the first
limitations. Blood levels may reflect recent behaviors rather 2 years of follow-up when compared with the natural history
than general body stores, blood levels may not reflect tissue eyes.91 Another prospective study, the Prophylactic Treatment
levels, dietary consumption histories may be imprecise, and the of Age-Related Macular Degeneration Research Group (PTAMD
likely effects of uncontrolled confounding factors. A cause-and- Study Group) found similar results in fellow eyes assigned to
effect relationship between antioxidants and AMD cannot grid threatment with the 810 nm diode laser.92 In this trial, the
be concluded from these investigations. Therefore, the AREDS rate of CNV development in laser treated eyes consistently
was designed to prospectively evaluate the use of antioxidants exceeded the rate found among the observation eyes. After
and zinc in individuals at risk of developing advanced AMD. 1 year of follow-up, 15.8% of laser-treated eyes developed CNV
The AREDS Group conducted a prospective multicenter compared to 1.4% of observation eyes (P = 0.05). More recently,
clinical trial to evaluate the effect of daily supplementation with results from the largest investigation of prophylactic laser
high-dose anti-oxidant vitamins and zinc supplements on the treatment, the Complications of Age-Related Macular
development of advanced AMD.15 A total of 3640 subjects were Degeneration Prevention Trial (CAPT) demonstrated that low
randomized to one of four treatment groups in a double masked intensity laser treatment to one eye of individuals with bilateral
fashion: (1) antioxidants (500 mg vitamin C, 400 IU vitamin E, nonneovascular AMD characterized by multiple large drusen
15 mg beta carotene), (2) zinc (80 mg zinc oxide and 2 mg did not demonstrate benefit in vision or anatomic outcomes.93
cupric oxide to prevent potential anemia), (3) combination of The laser protocol specified 60 spots of barely visible burns in a
antioxidants and zinc, or (4) placebo. Results from the AREDS grid pattern within an annulus of 1500 and 2500 mm of the
demonstrate that the combination supplements (anti-oxidants foveal center. Cumulative 5 year incidence rates for develop-
plus zinc) result in a 25% risk reduction for development of ment of CNV (13.3% laser treated vs 13.3% observed) and GA
advanced AMD in individuals at risk of progression and a 21% (7.4% laser treated vs 7.8% observed,) were similar between the
risk reduction in rates of moderate vision loss (doubling of the two groups. In addition, at 5 years the proportion of subjects
visual angle). Individuals at risk of progression are those with with ≥3 line loss of visual acuity from baseline was similar
intermediate AMD or unilateral advanced AMD with a fellow between the laser treated eyes (20.5%) and observed eyes (20.5%).
eye at risk. Individuals with early AMD did not benefit from Therefore, laser photocoagulation is not recommended in eyes
micronutrient supplementation. Therefore, individuals should with bilateral large drusen since it does not alter the natural
consider taking an antioxidant and zinc supplement such as history of AMD in these eyes.
that used in AREDS only if they meet the AREDS classification At the present time, the most important aspect of manage-
of intermediate AMD or unilateral advanced AMD. Potential ment in patients with drusen is education. Patients should be
risks associated with this daily supplementation are small and informed that AMD does not, in the majority of cases, cause
include a mild increase in genitourinary hospitalizations in legal blindness and that even in the worst cases, peripheral
participants taking zinc (7.5% in the zinc group vs 4.9% in the vision is almost always retained. These patients are at increased
placebo group) and a change in skin color (8.3% in the anti- risk of acquiring CNV, especially if one eye has already been
oxidant group vs 6.0% in the placebo group. Although the affected, and data suggest that diagnosis at the earliest onset
AREDS type micronutrient supplementation is generally safe, of the CNV provides the greatest chance of successful
cigarette smokers have an increased risk of developing lung treatment.94,95 Therefore, patients should monitor their central
cancer and increased risk of mortality when taking high doses vision every day in each eye at risk for the development of CNV,
of beta carotene.89,90 Smokers should be counseled regarding and they should contact their ophthalmologist promptly if they
this increased risk and consider avoidance of high dose notice any metamorphopsia or scotoma, which may suggest the
betacarotene exposure. In AREDS supplementation with zinc onset of CNV. Evaluation by an ophthalmologist should include
(in the absence of antioxidants) was found to significantly careful contact lens biomicroscopy for the presence of subretinal
reduce anatomic progression of AMD, and maybe considered as fluid or hemorrhage and, if necessary, an OCT and a fluorescein
an alternate treatment regimen in smokers. angiography to determine whether or not CNV is present.
A second multicenter, prospective clinical trial is now under- Recently, a new perimetry device, the Preferential Hyperacuity
way called AREDS 2. This study will evaluate additional supple- Perimeter (PreView PHP, Carl Zeiss Meditec, Dublin, CA) was
1912 ments consisting of lutein/zeaxanthine and omega-3 poly- developed to detect metamorphopsia in patients with AMD. A
Age-Related Macular Degeneration: Drusen and Geographic Atrophy

pilot study demonstrated that the PHP had a sensitivity of 94% to neovascular AMD. In addition, since extension of GA through
compared to 34% for the Amsler grid in detecting CNV secondary the foveal center can result in severe central visual loss patients
to AMD.96 In addition, a larger clinical trial demonstrated that may benefit from low-vision aids. We advise patients to maintain
the PHP has a sensitivity of 82% and a specificity of 88% in an open dialog with a low-vision specialist to discuss changes in
detecting newly diagnosed CNV in a cohort of individuals com- their needs and new aids that may become available.
prised of recent-onset CNV or intermediate AMD.97 Additional
clinical trials evaluating the PHP as a screening tool in patients CONCLUSIONS AND FUTURE RESEARCH
with high risk macular characteristics are underway. Data from
these studies may provide information on the role of PHP In recent years, standardized descriptions of non-neovascular
testing in the management of patients at risk of CNV. features of AMD have been developed and substantial natural
Unfortunately, some patients develop CNV in the absence of history data on AMD utilizing these standard definitions has
appreciable symptoms,98 and many ophthalmologists recommend been accumulated. Complex and simple scales describing the
that patients who have drusen be followed at 6 month intervals evolution of AMD have been developed. Limited information
in the hope of detecting asymptomatic CNV that might be on the cause or progression of AMD exists. It is hoped that
amenable to treatment.99 current and future studies will lead to better understanding of
the pathogenesis of this disease. The AREDS has confirmed
that antioxidant and zinc supplements can reduce progression
GEOGRAPHIC ATROPHY OF THE RPE of AMD and vision loss in eyes at risk. Additional interven-

CHAPTER 144
Management of eyes with GA requires patient education, self- tional trials may allow us to gain more insight into the disease
monitoring of symptoms and vision, and routine clinical pathogenesis and reduce the burden of vision loss associated
examination by an ophthalmologist to monitor for conversion with this condition.

REFERENCES
1. Eye Diseases Prevalence Research Group: angiography. Clin Exp Ophthalmol 2003; 22. Vingerling JR, Dielemans I, Hofman A, et al:
Prevalence of age-related macular 31:300–304. The prevalence of age-related maculopathy
degeneration in the United States. Arch 12. Coffey AJH, Brownstein S: The prevalence in the Rotterdam Study. Ophthalmology
Ophthalmol 2004; 122:564–572. of macular drusen in postmortem eyes. Am 1995; 102:205–210.
2. Donders FC: Beitrage zur pathogischen J Ophthalmol 1986; 102:164–171. 23. Mitchell P, Smith W, Attebo K, Wang JJ:
Anatomie des Auges. Graefes Arch 13. Klein R, Klein BEK, Jensen SC, Meuer SM: Prevalence of age-related maculopathy in
Ophthalmol 1855; 1:106. The incidence and progression of age- Australia. Ophthalmology 1995;
3. Bressler NM, Bressler SB, West SK, et al: related maculopathy. The Beaver Dam Eye 102:1450–1460.
The grading and prevalence of macular Study. Ophthalmology 1997; 104:7–21. 24. Klein R, Klein BEK, Knudtson MD, et al:
degeneration in Chesapeake Bay 14. Maguire MG, Bressler SB, Bressler NM, Prevalence of age-related macular
watermen. Arch Ophthalmol 1989; et al: Risk factors for choroidal degeneration in 4 racial/ethnic groups in
107:847–852. neovascularization in the second eye of the multi-ethnic study of atherosclerosis.
4. Bressler SB, Maguire MG, Bressler NM, patients with juxtafoveal or subfoveal Ophthalmology 2006; 113:373–380.
Fine SL: The Macular Photocoagulation choroidal neovascularization secondary to 25. Munoz B, Klein R, Rodriguez J, et al:
Study Group: Relationship of drusen and age-related macular degeneration. Arch Prevalence of age-related macular
abnormalities of the retinal pigment Ophthalmol 1997; 115:741–747. degeneration in a population-based
epithelium to the prognosis of neovascular 15. AREDS Research Group: A randomized, sample of Hispanic people in Arizona:
macular degeneration. Arch Ophthalmol placebo-controlled clinical trial of high- proyecto VER. Arch Ophthalmol 2005;
1990; 108:1442–1447. dose supplementation with vitamins C and 123:1575–1580.
5. Macular Photocoagulation Study Group: E, beta carotene, and zinc for age-related 26. Prenner JL, Rosenblatt BJ, Tolentino MJ,
Persistent and recurrent neovascularization macular degeneration and vision loss. Arch et al. Risk factors for choroidal
after krypton laser photocoagulation for Ophthalmol 2001; 119:1417–1436. neovascularization and vision loss in the
neovascular lesions of age-related macular 16. El Baba F, Green WR, Fleischmann J, et al: fellow eye study of CNVPT. Retina 2003;
degeneration. Arch Ophthalmol 1990; Clinicopathologic correlation of lipidization 23:307–314.
108:825–831. and detachment of the retinal pigment 27. Green WR, McDonnell PH, Yeo JH:
6. Klein R, Klein BEK, Linton KLP: Prevalence epithelium. Am J Ophthalmol 1986; Pathologic features of senile macular
of age-related maculopathy. The Beaver 101:576. degeneration. Ophthalmology 1985;
Dam Eye Study. Ophthalmology 1992; 17. Bressler NM, Munoz B, Maguire MG, et al: 92:615–627.
99:933–943. Five-year incidence and disappearance of 28. Pouleikhoff D, Barondes MJ, Minassian D,
7. Klein R, Davis MD, Magli YL, et al: The drusen and retinal pigment epithelium et al: Drusen as risk factors in age-related
Wisconsin age-related maculopathy abnormalities. Waterman Study. Arch macular degeneration. Am J Ophthalmol
grading system. Ophthalmology 1991; Ophthalmol 1995; 113:301–308. 1990; 109:38–43.
98:1128–1134. 18. Green WR, Enger C: Age-related macular 29. Mavrofrides EC, Villate N, Rosenfeld PJ,
8. Bird AC, Bressler NM, Bressler SB, et al: degeneration histopathologic studies: The Puliafito CA: Age-related macular
An international classification and grading 1992 Lorenz E. Zimmerman Lecture. degeneration. In: Schuman JS, Puliafito
system for age-related maculopathy and Ophthalmology 1993; 100:1519–1535. CA, Fujimoto JG, eds. Optical coherence
age-related macular degeneration. Surv 19. Bressler NM, Silva JC, Bressler SB, et al: tomography of ocular diseases. 2nd edn.
Ophthalmol 1995; 39:367–374. Clinicopathologic correlation of drusen and Slack Inc; 2004:243–252.
9. AREDS Research Group: The age-related retinal pigment epithelial abnormalities in 30. Lebowitz HM, Krueger DE, Maunder LR,
eye disease study system for classifying age-related macular degeneration. Retina et al: The Framingham Eye Study
age-related macular degeneration from 1994; 14:130–142. Monograph VI: Macular Degeneration. Surv
stereoscopic color fundus photographs: the 20. Sarks SH: Ageing and degeneration in the Ophthalmol 1980; 24(Suppl):428–457.
age-related eye disease study report no 6. macular region: A clinicopathologic study. 31. Hyman LG, Lilienfeld AM, Ferris FL, Fine
Am J Ophthalmol 2001; 132:668–681. Br J Ophthalmol 1976; 60:324–341. SL: Senile macular degeneration: a case-
10. Gass JDM: Stereoscopic atlas of maular 21. Bressler SB, Munoz B, Phillips D, West SW, control study. Am J Epidemiol 1983;
diseases diagnosis and treatment. 4th edn. for the Salisbury Eye Evaluation Group: 118:213–227.
St Louis: Mosby; 1997:78–79. Prevalence of age-related macular 32. Ferris FL, Fine SL, Hyman L: Age-related
11. Chang AA, Guyer DR, Orlock DR, Yannuzzi degeneration in a population based study: macular degeneration and blindness due to
LA: Age-dependent variations in the drusen SEE Project. Invest Ophthalmol Vis Sci neovascular maculopathy. Arch Ophthalmol
fluorescence on indocyanine green 1998; 39:S493. 1984; 102:1640–1642. 1913
RETINA AND VITREOUS

33. Klein ML, Ferris FL, Gensler G, et al: 52. Wang JJ, Foran S, Smith W, Mitchell P: Risk degeneration. The Beaver Dam Eye Study.
Clinical features and natural history of of age-related macular degeneration in eyes Arch Ophthalmol 1993; 111:514–518.
drusenoid pigment epithelial detachments with macular drusen or hyperpigmentation. 70. Taylor HR, West SK, Rosenthal FS, et al:
in age-related macular degeneration. Arch Ophthalmol 2003; 121:658–663. Effect of ultraviolet radiation on cataract
Poster presentation, ARVO annual meeting; 53. Klaver CC, Assink JJ, van Leeuwen R, formation. N Engl J Med 1988;
2004. et al: Incidence and progression rates of 319:1429–1433.
34. Sarks JP, Sarks SH, Killingsworth MC: age-related maculopathy: the Rotterdam 71. AREDS Study Group: Risk factors for the
Evolution of geographic atrophy of the Study. Invest Ophthalmol Vis Sci 2001; incidence of advanced age-related macular
retinal pigment epithelium. Eye 1988; 42:2237–2241. degeneration in the age-related eye
2:552–577. 54. Buch H, Nielson NV, Vinding T, et al: disease study (AREDS). Ophthalmology
35. Smiddy WE, Fine SL: Prognosis of patients 14-year incidence, progression, and visual 2005; 112:533–539.
with bilateral macular drusen. morbidity of age-related maculopathy: the 72. Tomany SC, Wang JJ, van Leeuwen R et al:
Ophthalmology 1984; 91:271–277. Copenhagen City Eye Study. Risk factors for incident age-related
36. Holz FG, Wolfensberger TJ, Piguet PQ, Ophthalmology 2005; 112:787–798. macular degeneration. Ophthalmology
et al: Bilateral macular drusen in age- 55. Schatz H, McDonald HR: Atrophic macular 2004; 111:1280–1287.
related macular degeneration. Prognosis degeneration: Rate of spread of geographic 73. Goldberg J, Flowerdew G, Smith E, et al:
and risk factors. Ophthalmology 1994; atrophy and visual loss. Ophthalmology Factors associated with AMD: analysis of
101:1522–1528. 1989; 96:1541–1551. the first NHANES. Am J Epidemiol 1988;
37. Jampol LM: Discussion of prognosis of 56. Sunness JS, Bressler NB, Tian Y, et al: The 128:700–710.
patients with bilateral macular drusen. enlargement of geographic atrophy over 74. Klein R, Klein BEK, Franke T: The
Ophthalmology 1984; 91:276–277. time in age-related macular degeneration. relationship of cardiovascular disease and
SECTION 10

38. AREDS Research Group: The age related Invest Ophthalmol Vis Sci 1996; 37:S21. its risk factors to age-related maculopathy.
eye disease study severity scale for age- 57. Maguire P, Vine AK: Geographic atrophy of The Beaver Dam Eye Study.
related macular degeneration. Arch the retinal pigment epithelium. Am J Ophthalmology 1993; 100:406–414.
Ophthalmol 2005; 123:1484–1498. Ophthalmol 1986; 102:621–625. 75. Mares-Perlman JA, Brady WE, Klein R, et al:
39. Early Treatment Diabetic Retinopathy Study 58. Applegate CA, Sunness JS, Haselwood Dietary fat in age-related maculopathy.
Research Group: Classification of diabetic DM: Visual symptoms associated with Arch Ophthalmol 1995; 113:743–748.
retinopathy from fluorescein angiograms. geographic atrophy from age-related 76. Hammond CJ, Webster AR, Snieder H, et
Ophthalmology 1991; 98:807–822. macular degeneration. Invest Ophthalmol al: Genetic influence on early age-related
40. AREDS Research Group. A simplified scale Vis Sci 1996; 37:S112. maculopathy: a twin study. Ophthalmology
for age-related macular degeneration. Arch 59. Sunness JS, Applegate CA: Long-term 2002; 109:730–736.
Ophthalmol 2005; 123:1570–1574. follow-up of fixation patterns in eyes with 77. Seddon JM, Cote J, Page WF, et al: The
41. Gass JDM, Jallow S, Davis B: Adult central scotomas from geographic atrophy US twin study of age-related macular
vitelliform macular detachment occurring in that is associated with age-related macular degeneration: relative roles of genetic and
patients with basal laminar drusen. Am J degeneration. Am J Ophthalmol 2005; environmental influences. Arch Ophthalmol
Ophthalmol 1985; 99:445–459. 140:1085–1093. 2005; 123:321–327.
42. Kenyon KR, Maumenee AE, Ryan SJ, et al: 60. Sunness JS, Ruben GS, Applegate CA, 78. Haddad S, Chen CA, Santangelo SL,
Diffuse drusen and associated et al: Visual function abnormalities and Seddon JM. The genetics of age-related
complications. Am J Ophthalmol 1985; prognosis in eyes with age-related macular degeneration: a review of progress
100:119–128. geographic atrophy of the macula and to date. Surv Ophthalmol 2006;
43. Bressler NM, Bressler SB, Fine SL: Age- good visual acuity. Ophthalmology 1997; 51:316–363.
related macular degeneration. Surv 104:1677–1691. 79. Haines JL, Hauser MA, Schmidt S, et al.
Ophthalmol 1988; 32:375–413. 61. Sunness JS, Bressler NM, Maguire MG: Complement factor H variant increases the
44. Gass JDM: A clinicopathologic study of a Scanning laser ophthalmoscopic analysis risk of age-related macular degeneration.
peculiar foveomacular dystrophy. Trans Am of the pattern of visual loss in age-related Science 2005; 308:419–421.
Ophthalmol Soc 1974; 72:139. geographic atrophy of the macula. Am J 80. Edwards AO, Ritter R III, Abel KJ, et al:
45. Todd K, Schatz H, Crawford JB: Pathologic Ophthalmol 1995; 119:143–151. Complement factor H polymorphism and
findings in pseudovitelliform macular 62. Sunness JS, Bressler NM, Marsh MJ, et al: age-related macular degeneration. Science
degeneration. Invest Ophthalmol Vis Sci The development of choroidal 2005; 308:421–424.
1986; 27:198. neovascularization in eyes with geographic 81. Klein RJ, Zeiss C, Chew EY, et al:
46. Benhamou N, Souied EH, Zolf R, et al: atrophy from age-related macular Complement factor H polymorphism in
Adult-onset foveomacular vitelliform degeneration. Invest Ophthalmol Vis Sci age-related macular degeneration. Science
dystrophy: a study by optical coherence 1997; 38:S967. 2005; 308:385–389.
tomography. Am J Ophthalmol 2003; 63. Nasrallah F, Jalkh AE, Trempe CL, et al: 82. Postel EA, Agarwal A, Caldwell J, et al:
135:362–367. Subretinal hemorrhage in atrophic age- Complement factor H increases risk for
47. Gass JDM: Stereoscopic atlas of macular related macular degeneration. Am J atrophic age-related macular degeneration.
diseases: diagnosis and treatment. 3rd Ophthalmol 1989; 107:38–41. Ophthalmology 2006; 113:1504–1507.
edn. St Louis: Mosby; 1987:264. 64. Young RW: Solar radiation in age-related 83. Newsome DA, Swartz M, Leone NC, et al:
48. Burgess DB, Hawkins BS, Jefferys J, et al: macular degeneration. Surv Ophthalmol Oral zinc in macular degeneration. Arch
Five-year follow-up of fellow eyes with 1988; 32:252–269. Ophthalmol 1988; 106:192–198.
patients with age-related macular 65. Gottsch JD, Bynoe LA, Harlan JB: Light- 84. Stur M, Tittl M, Reitner A, Meisinger V: Oral
degeneration and unilateral extrafoveal induced deposits in Bruch’s membrane in zinc and the second eye in age-related
choroidal neovascularization. Arch protoporphyric mice. Arch Ophthalmol macular degeneration. Invest Ophthalmol
Ophthalmol 1993; 111:1189–1199. 1993; 111:126–129. Vis Sci 1996; 37:1225–1235.
49. Bressler SB, Bressler NM, Clemons T, et al: 66. West SK, Rosenthal FS, Bressler NM, et al: 85. Eye Disease Case-Control Study Group:
Ocular risk factors for developing Exposure to sunlight and other risk factors Antioxidant status in neovascular age-
neovascular AMD in fellow eyes of patients for age-related macular degeneration. Arch related macular degeneration. Arch
with unilateral neovascular AMD. Presented Ophthalmol 1989; 107:875–879. Ophthalmol 1993; 111:104–109.
at ARVO Annual Meeting; 2004. 67. Taylor HR, Munoz B, West SK, et al: Visible 86. Seddon JM, Ajani UA, Sperduto RD, et al:
50. Armstrong J, Hubbard LD, Davis MD, et al: light and risk of age-related macular Dietary carotenoids, vitamins A, C, and E,
Association of calcified drusen with degeneration. Trans Am Ophthalmol Soc and advanced age-related macular
progression of AMD in AREDS participants. 1990; 88:163–178. degeneration. JAMA 1994; 272:1413–1420.
Presented at ARVO Annual Meeting; 2006. 68. Eye Disease Case-Control Study Group: 87. West S, Vitale S, Hallfrisch J, et al: Are
51. Klein R, Klein BE, Tomany Sc, et al: Ten- Risk factors for neovascular age-related antioxidants or supplements protective for
year incidence and progression of age- macular degeneration. Arch Ophthalmol age-related macular degeneration? Arch
related maculopathy; the Beaver Dam 1992; 110:1701–1708. Ophthalmol 1994; 112:222–227.
eye study. Ophthalmology 2003; 69. Cruickshanks KJ, Klein R, Klein BEK: 88. Mares-Perlman JA, Brady WE, Klein R,
1914 109:176–179. Sunlight and age-related macular et al: Serum antioxidants in age-related
Age-Related Macular Degeneration: Drusen and Geographic Atrophy

macular degeneration in a population- 810-nanometer laser to eyes with drusen. related macular degeneration.
based case-control study. Arch Ophthalmol Unilaterally eligible patients. Ophthalmology 2003; 110:966–970.
1995; 113:1518–1523. Ophthalmology 2006; 113:622e1. 97. Preferential Hyperacuity Perimeter
89. Alpha-Tocopherol, Beta Carotene Cancer 93. Complications of Age-Related Macular Research Group: Preferential Hyperacuity
Prevention Study Group: The effect of Degeneration Prevention Trial Research Perimeter (PreView PHP) for detecting
vitamin E and beta carotene on the Group: Laser treatment in patients with choroidal neovascularization study.
incidence of lung cancer and other bilateral large drusen: the complications of Ophthalmology 2005; 112:1758–1765.
changes in male smokers. N Engl J Med age-related macular degeneration 98. Moisseiev J, Bressler NM: Asymptomatic
1994; 330:1029–1035. prevention trial. Ophthalmology 2006; neovascular membranes in the second eye
90. Omenn GS, Goodman GE, Thornquist MD, 113:1974–1986. of patients with visual loss from age-related
et al: Effects of a combination of beta 94. Bird AC: Treatment of senile macular macular degeneration (AMD). Invest
carotene and vitamin A on lung cancer and degeneration by photocoagulation. Br J Ophthalmol Vis Sci 1990; 31:462.
cardiovascular disease. N Engl J Med Ophthalmol 1974; 58:367–376. 99. American Academy of Ophthalmology
1996; 334:1150–1155. 95. Macular Photocoagulation Study Group: Quality of Care Committee Retina Panel:
91. Choroidal Neovascularization Prevention Laser photocoagulation of subfoveal Macular degeneration. American Academy
Trial Research Group: Laser treatment in neovascular lesions in age-related macular of Ophthalmology Preferred Practice
eyes with large drusen: Short-term effects degeneration. Results of a randomized Pattern. San Francisco: American Academy
seen in a pilot randomized clinical trial. clinical trial. Arch Ophthalmol 1991; of Ophthalmology; 1990.
Ophthalmology 1998; 105:11–23. 109:1219–1231.
92. Friberg TR, Musch DC, Lim JI, et al: 96. Loewenstein A, Malach R, Goldstein M,

CHAPTER 144
Prophylactic treatment of age-related et al: Replacing the amsler grid: a new
macular degeneration report number 1: method for monitoring patients with age-

1915
CHAPTER
Foods and Supplements in the Prevention and
145 Treatment of Age-Related Macular Degeneration
Julie A. Mares

INTRODUCTION specific nutrient. For example, people who eat high levels of
the carotenoids lutein and zeaxanthin typically consume high
Food contains numerous bioactive molecules (vitamins, min- levels of fruits and vegetables, particularly dark leafy greens.
erals, fatty acids, and other phytochemicals) that may modulate These foods, and foods often eaten along with them, are also
and attenuate the multifactorial pathogenetic processes that high in vitamins C and E and other carotenoids that might
promote age-related macular degeneration (AMD), including also protect the retina. There is often an attempt to statistically
oxidative stress, inflammation, phototoxicity, and ischemia. adjust for these other aspects of diet in order for investigators
Nutrition may play an even larger role in preventing age-related to draw conclusions about a specific nutrient of interest. This
degenerative changes as endogenous systems to manage these statistical approach is unlikely to be sufficient to fully isolate
pathogenic processes become more challenged with age. the effects of one target nutrient with other related aspects of
Evidence that addresses the practical importance of nutrition on diet. This is because the intake of so many of the potentially
the common degenerative conditions that promote AMD has protective ingredients in food is highly correlated, and study
grown over the past three decades and is the focus of the current samples do not include large numbers of people who have high
chapter. levels of one component and low levels of all other potentially
Throughout the chapter, three general types of scientific protective components to reliably compare risks in these
evidence that inform us about nutrition and AMD will be subgroups. For this reason, evidence of relationships of diets
discussed for each category of food component. One type of high in certain nutrients in the diet with AMD risk cannot be
evidence comes from studies in animals and cell systems in used to reliably predict the influence of supplements that con-
which the short-term effects of nutrient deficiency or high-dose tain the nutrient. The relationships of certain nutrients to
supplementation are tested. These experiments, which push AMD might also reflect the influence of other aspects of a
the limits of physiological systems, elucidate the mechanisms health-conscious lifestyle, such as physical activity and use of
by which particular nutrients can exert physiological or bio- protective sun gear, especially if not known or not measured
chemical influences. These do not, however, provide insights well and adjusted for. Thus, while the observations made from
about the influence of more moderate variations in intake, epidemiological studies illustrate broad diet attributes that
beyond overt deficiency states or pharmacological effects of associate with lower risk for AMD, they do not permit us to
nutrients under isolated conditions. The results of these exper- generalize about the effects of single nutrients, either as distinct
iments do not generally demonstrate the effect of a nutrient in from the foods that contain them, or in nutritional
the routine context where the nutrient is one of many com- supplements.
ponents of foods consumed. Evidence for mechanisms by which The attempt to understand the influence of particular
nutrients may play a role in AMD is insufficient to predict the nutrients when consumed in isolation and at high levels drove
impact of food or supplement sources over decades. several decades of research using clinical trial designs. The
Epidemiological studies in populations provide broader appeal of the results of these trials is that a specific, short-term
insights about relationships between the intake of foods and impact of a nutrient or combination of nutrients can be known
supplements under more relevant conditions in people and with a high level of certainty, and not be confounded by the
over the long term. These observations illustrate how diets other food components or health behaviors that can influence
that are rich in certain foods or nutrients relate to the devel- the results of observational studies. These trials have provided
opment of early and late stages of AMD, usually over decades, insights about the potential for nutritional manipulation to
in people possessing a variety of genetic predispositions, whose impact the progression of AMD. Such trials aim to test the
health is influenced by a variety of medical conditions, such as impact of food ingredients as medicines.
the presence of hypertension or diabetes and lifestyles such Given the limited medical approaches available to treat
as the use of alcohol and cigarettes. The strength of evidence AMD, nutritional supplementation options have been valued.
from observational studies lies in the strength and consistency The specific nutrient doses and combination of ingredients is
of associations across many different samples of people who informed by a limited state of knowledge limiting the ability to
vary in the characteristics that might explain or confound these generalize to the long-term influence of nutrients in foods or
observations, rather from any single study. other supplement compositions.
While relationships with specific nutrients or food compo- Despite limitations of any one type of evidence, an inte-
nents to AMD are often the focus in such studies, the results gration of evidence from these three scientific approaches has
are influenced by numerous other food components that are provided much broader and deeper understanding with which
associated with the kind of diet that provided high levels of that to counsel both patients with AMD and their families, who, due
1917
RETINA AND VITREOUS

to genetic predispositions, might themselves be at higher risk to AMD from intermediate to more advanced stages by 28%.
develop the conditions some day. The current evidence, as well There is also evidence that this treatment reduces the age-
as gaps in our existing knowledge, will be described in the related oxidation of cysteine in the blood, which supports the
remainder of the chapter. We will conclude with a discussion possibility that the benefit is due to an improvement in oxi-
of broad dietary patterns that show promise to lower the rate dative stress.11 A small short-term study of men with atrophic
of AMD. AMD, who received antioxidant supplements along with lutein,
also reported improved visual function in this treatment group
ANTIOXIDANTS and not in randomized, placebo controls.12
Although the level of individual antioxidants in foods is
Oxidative damage occurs to macro molecules (such as proteins, usually much lower than levels provided by current high-dose
lipids, and DNA) when reactive oxygen species (ROS) interact antioxidant supplements, foods provide an even greater array of
with them and compromise their function. The sources of ROS antioxidants than supplements, and the impact of decades of
are many and can be the result of photooxidation of lipids from consuming dietary antioxidants on AMD may be substantial. In
light exposure; they can also be the by-product of metabolic epidemiological studies, when associations with antioxidant
events, such as oxidative metabolism or enzyme reactions that nutrients are considered one at a time, low levels of one or more
use oxygen (such as xanthine oxidase). Alternatively, they may antioxidants in the blood or diet have often, but not always,
be produced directly as a mechanism for biological defense, as been related to higher prevalence or incidence of certain age-
is the case when white blood cells respond immunologically to related changes in the macula. Antioxidant nutrients related to
SECTION 10

pathogens with an oxidant boost. lower occurrence of AMD include vitamin E13–15 and/or one
It is widely known that the retina is particularly susceptible or more carotenoids or zinc.14–20 The potential impact of a
to oxidative stress because of its high rates of oxidative metab- combination of dietary antioxidants was estimated in the
olism and light exposure, and its high concentration of lipids Rotterdam Eye Study.15 After adjusting for potential con-
with double bonds that are vulnerable to attack by free radicals. founders, the 10% of participants who were consuming
Multiple types of oxidative stress occur in cells, and the net antioxidant-rich diets (i.e., diets with above the median intake
damage is a function of the stressors themselves, as well as the of all four antioxidants in AREDS supplements) had a 35%
ability to defend against them and repair the damage done. lower risk of incident AMD than those with more average diets
Food contains numerous antioxidants, such as vitamins C (diets with intakes of one to three antioxidants above the
and E and carotenoids, which are provided by fruits, vegetables, median). Risk in persons with antioxidant-poor diets (with all
nuts, and nut or vegetable oils. These are chemicals that either four antioxidant nutrients at levels below the median) was
directly neutralize ROS, or that are cofactors for enzyme higher, although this was not statistically significant after
systems that neutralize ROS, such as zinc and selenium, which adjustment for potential confounders, and larger samples would
are provided by meat, dairy, and some grains. These antioxi- be needed to better evaluate this trend. Overall, there is a clear
dants work in different cell compartments, and often in comple- trajectory of lower risk for AMD that is associated with eating
mentary ways. Vitamin C is the most abundant dietary diets higher in several antioxidants. Lowering the prevalence of
antioxidant in aqueous compartments of the cell, while vitamin early or intermediate stages of AMD by eating antioxidant-rich
E and vitamin E-related compounds are the most abundant foods has the potential to prevent even more cases of advanced
lipid-phase antioxidants in foods. Other hydrocarbon plant AMD than does beginning supplements once intermediate
compounds such as carotenoids are also active in lipid-soluble AMD becomes manifest.
cell compartments, and may have unique function by virtue
of their ability to align in biomembranes of cells or organelles.
Foods can contain direct oxidants or substances which promote Antioxidants and AMD
the production of ROS (including, potentially, residual herbi- Summary of Evidence
cides and pesticides) but little is known about these effects on Oxidative stress or deficiency of dietary antioxidants in animals
the eye and they will not be discussed here. promotes degenerative changes in the retina.
In people, there is broad evidence that supports the benefit of a
In studies with experimental animals, deficiencies of
combination of antioxidants in slowing early and late AMD:
vitamins E and C, or nutrients such as zinc, which are
• Most previous population studies indicate lower prevalence of
components in enzymes that protect against oxidative stress,
early AMD among persons who have diets that are high,
result in pathologic changes to the retina (as previously
compared with low, in one or more antioxidant nutrients
reviewed1–3). Yet, antioxidants can behave as pro-oxidants under
• Multiple high-dose antioxidants in one large clinical trial
some circumstances. Single or dual high-dose antioxidant
lowered the progression of intermediate to late AMD, but
supplements have not been associated with lower risk of
single antioxidants have not lowered the incidence of AMD
progression of AMD.4,5 This is consistent with evidence that
• The same combination of antioxidants in foods was associated
oxidant defense is carried out by a highly complex system of
with lower 8-year incidence of AMD in the Rotterdam Eye
mechanisms, in which antioxidant effects of several different
Study
individual antioxidant molecules have been found to be
synergistic.6–8 The type of oxidative stress that is most Bottom Line and Unanswered Questions
damaging depends on both the cell type and intracellular Foods rich in antioxidants (fruits, vegetables, and whole grains) are
often associated with lower risk of early AMD.
location. Retinal pigment epithelium cells, when cultured,
• The impact of diet on progression from intermediate to
demonstrate different vulnerabilities to the type of oxidant
advanced AMD has not been adequately studied
stress than do cells from other tissues, and the result of
One specific high-dose antioxidant supplement lowers risk for
oxidants that target the cell surface and cytosol differs from that progression from intermediate to advanced AMD.
which targets specific organelles.9 • The possibility that lower levels of antioxidants may be
In the Age-Related Eye Disease Study (AREDS), a 6.2 year effective and the long-term health influence of high-dose
randomized, placebo controlled clinical trial,10 the antioxidants has not been tested
administering of a high dose of the antioxidants (500 mg of • The optimal combination and dose of nutrients in supplements
vitamin C, 400 IU of vitamin E, 15 mg of beta-carotene, 80 mg is unknown
1918 of zinc along with 2 mg of copper), slowed the progression of
Foods and Supplements in the Prevention and Treatment of Age-Related Macular Degeneration

The number of known and unknown antioxidants in foods pigment epithelial (RPE) cells and increased photoreceptor cell
exceeds that in most supplements, and foods high in death.40 Retinal zeaxanthin has been demonstrated to prevent
antioxidants, such as vegetables, might lower oxidative stress to light-induced photoreceptor death in quail.41
a greater degree than supplements. In a recent randomized, Lower levels of lutein and zeaxanthin have been found in
cross-over trial, eating two or more cups of Brassica vegetables autopsy specimens of donor eyes with AMD.42 Similarly, in two
(such as broccoli) lowered a urinary marker of oxidative stress, cross-sectional studies, people with AMD have lower
whereas moderate levels of supplementation with antioxidants concentrations of these carotenoids in their macula compared
in multivitamins did not.21 However, it is not known whether to those without AMD.43,44 In contrast, in the largest such
the non-nutritive antioxidants in foods reach the retina (besides cross-sectional study, which involved about 1700 women,
lutein and zeaxanthin, which are discussed next) or whether macular pigment was actually higher in those with intermediate
lowering the systemic level of oxidative stress by consuming and advanced AMD.45 However, evidence in subgroups of
such foods influences age-related macular degeneration. women from this study is consistent with the possibility of a
protective effect in the youngest two-thirds of the sample with
LUTEIN AND ZEAXANTHIN stable diets. Prospective studies of the relationships of macular
pigment to the onset of AMD are needed in order to understand
The specific carotenoids lutein, and its structural isomer whether low macular pigment is a consequence of, or a
zeaxanthin, may protect against the development of AMD, as contributor to, AMD.
previously reviewed.22,23 These carotenoids, which comprise

CHAPTER 145
macular pigment24,25 are responsible for the yellow color of the Lutein and Zeaxanthin and AMD
macula lutea and must be provided by the diet, since the body
Summary of Evidence
does not synthesize them. Their structure is similar to pro- In vitro and animal studies indicate that the oxygenated
vitamin A carotenoids, like beta-carotene, but with additional carotenoids lutein and zeaxanthin may lower oxidative stress in the
hydroxyl groups, which makes them more polar and unable to retina directly or by lowering blue light that reaches the
be used in vitamin A synthesis. Lutein and zeaxanthin are photoreceptor outer segments or RPE.
selectively concentrated into the retina and other ocular tissues Lutein and/or zeaxanthin supplementation in animals lowers light-
over the five other carotenoids which predominate in human induced damage to the retina.
blood, and over the ~600 carotenoids present in nature. The long-term intake of diets rich in lutein and zeaxanthin is often,
Evidence for the existence of specific xanthophyll-binding but not consistently, related to lower risk for AMD.
proteins in the vertebrate retina has been reported.26,27 Bottom Line and Unanswered Questions
Lutein and zeaxanthin may be obtained from many fruits and Foods rich in lutein and zeaxanthin are high in antioxidants, which
vegetables, from egg yolks,28,29 or from supplements. are associated with lower risk of AMD.
Particularly rich sources include dark green leafy vegetables, • The possibility that they may enhance vision by augmenting
such as spinach and kale, and green vegetables like broccoli, macular pigment has not been tested
peas, green beans, and brussels sprouts. Americans, on average, The benefit of diets or supplements rich in lutein and zeaxanthin or
high macular pigment levels on incidence and progression of AMD
consume 1–2 mg of these carotenoids daily.30
has not been adequately studied.
Lutein and zeaxanthin are found throughout the retina. The
• Benefits or safety of supplementing with L and Z has not been
highest density occurs in the Henle fiber and inner plexiform
studied in large trials
layers.31 At these locations, they are likely to function as an
optical filter that absorbs short-wavelength visible (blue) light.32
These features not only may block oxidative damage due to Results from observational studies indicate inconsistent
incident light energy, but may also improve visual resolution as associations between the intake and blood levels of lutein and
previously reviewed.33 One mechanism by which macular zeaxanthin and the occurrence of age-related macular
pigment could protect against the development and progression degeneration. In two case-control studies, higher serum
of AMD is by blocking light-induced formation of a toxic levels46,47 and dietary intake of lutein and zeaxanthin48 were
compound, A2E; this is the principal component of lipofuscin associated with a statistically significant reduction in the risk of
that accumulates in the RPE during phagocytosis of the rods neovascular/exudative AMD. Higher levels of dietary lutein and
and cones, and is taken up by the lysosomes. When a critical zeaxanthin were also associated with lower prevalence of retinal
intracellular level of this compound has been reached, cell pigmentary abnormalities among Americans 40–59 years of
damage to DNA occurs, induced by blue light irradiation.34 age, and lower rates of advanced AMD in Americans 60–79
Because macular pigment can absorb 40–90% of incident blue years of age, in the Third National Health and Nutrition
light, it could reduce the amount of A2E formed. This Examination Survey.49 Low levels of plasma zeaxanthin19 in one
possibility has not yet been tested. study, and both lutein and zeaxanthin in another study,16 were
The macular pigment density reduces about twofold between associated with a higher prevalence of early and late AMD.
the central macula, where the zeaxanthin isomer predominates, Other studies have either failed to find an association, or
and the periphery where the lutein isomer predominates.24 The associations did not reach statistical significance.14,50–54 It is too
highest density of xanthophyll carotenoids in the peripheral early to draw conclusions from population studies because the
retina is in the rod outer segment membranes,35 where their inconsistent observations are likely to be biased by recent diet
concentration is consistent with a role in oxidant defense. An change, selective mortality bias or competing risk factors, or
antioxidant role is supported by the presence of oxidation limited to certain stages of AMD (discussed previously55). In the
products of lutein and zeaxanthin in the retina.36 These recent Carotenoids in Age-Related Eye Disease Study
carotenoids may also influence membrane stability by their (CAREDS), high intake of lutein and zeaxanthin was associated
unique alignment in biological membranes.37 with lower odds for intermediate AMD only after excluding
Animal models to study the impact of macular pigment on persons who were likely to have had unstable diets,20 suggesting
AMD or related photoreceptor health are limited to species that that dietary changes contribute to inconsistent results in
accumulate these carotenoids in their ocular tissues: primates, epidemiological studies of AMD. A bias of selective mortality
and some birds such as quail. Primates fed diets deficient in might also explain the inconsistent results in population
these xanthophyll carotenoids38,39 suffer a loss of retinal studies. For example, older participants in observational studies 1919
RETINA AND VITREOUS

may be more likely to have eaten diets rich in fruits and the intake of root and leafy green vegetables because they
vegetables over their adult lifetime than people in their birth contain vitamin K. Vitamin K is necessary for blood clotting by
cohort who are no longer living, making it more difficult to its action on the formation prothrombin, or clotting factor II. A
discern the effects of those diets on disease outcomes. Longer- sudden increase in vitamin K intake can increase blood
term prospective studies, particularly of the youngest persons at prothrombin levels. However, patients can be advised to consult
risk for AMD, will provide more insights in years to come. with the physician who manages the treatment to adjust the
The benefits of lutein and zeaxanthin supplements on warfarin dose to the highest daily green vegetable intake that
preventing or slowing AMD are unknown. One reason is that the patient can consistently eat.
the ability to accumulate carotenoids with supplements appears
to be variable across individuals. Between 20% and 50% of ZINC
subjects in previous investigations have low serum and/or
retinal response to oral supplementation with these Zinc may be particularly important to the retina because
carotenoids.56–58 In general, blood responses to oral carotenoids concentrations of zinc in the retina exceed those elsewhere in
vary in different people, as well.59,60 The influences on the the body, with the exception of the prostate.64 Deficiency of zinc
ability to take up carotenoids by the intestinal tract and the eye in both animals and people impairs retinal functioning, as
are largely unknown. Having high levels of body fat and diabetes previously reviewed.1 The mechanisms by which zinc could
is related to lower macular density.61 These conditions might influence AMD are numerous and fall into three general
reflect levels of oxidative stress or lipoprotein distributions, categories: catalytic, regulatory, and structural.65 Zinc catalyzes
SECTION 10

which could, in turn, influence the ability to accumulate enzymatic reactions, and is a cofactor of over 100 enzymes,
carotenoids. some of which are involved in oxidant defense. Zinc depletion
The benefit of taking lutein and zeaxanthin supplements has in RPE cells has reduced levels of catalase, glutathione
not been studied in prospective studies or clinical trials. There peroxidase, and metallothionein, and has reduced ability to
was, however, one very small and short-term trial in which phagocytize photoreceptor outer segments.66 Zinc performs
visual acuity improved slightly in the fewer than 30 subjects structural roles; its presence facilitates protein folding to
who received exclusively lutein supplements.12 Further produce biologically active molecules that organize into zinc
information about the benefits of supplementation with lutein finger-like structures. Zinc is also necessary for the DNA
and zeaxanthin (and omega-3 fatty acids) on progression of binding capability of over 400 nuclear regulatory units and plays
advanced AMD may come from a large set of multicenter a role in cellular signaling.67 It is involved in many homeostatic
clinical trials (AREDS II) that is currently underway. mechanisms, including immune responses, previously
Despite inconsistent data for these specific carotenoids to reviewed.67,68 Evidence can be found that both zinc deficiency
date, lutein and zeaxanthin may be two of the many and excesses impair immunity.68 Zinc depletion may also
components in fruits and vegetables that may slow earlier trigger apoptosis of RPE cells, or increase the vulnerability of
stages of AMD. It is possible there are combinations of foods RPE cells to photic injury.69 However, zinc supplementation can
that provide the optimal mix of nutrients that protect against also enhance stress-induced effects in RPE cells.70 Thus, there
AMD. Americans who reported high intakes of vitamin A-rich is evidence for numerous mechanisms by which zinc could
fruits and vegetables had lower prevalence of AMD.62 Low fruit influence retinal systems that are involved in the development
and vegetable consumption was associated with higher 5 year of AMD.
incidence of large drusen in the Beaver Dam Eye Study.14 In The longer-term benefit of zinc on risk for AMD is not clear
CAREDS, there was a strong inverse relationship between from observational studies. In the Beaver Dam Eye Study18 high
intermediate AMD and the intake of vegetables in general, and intake of zinc from foods and supplements was related to lower
green vegetables, specifically, in women under 75 years of age prevalence of early AMD, compared to low intake. The
with stable intakes. One small study in Lithuania reported that association for the incidence of early AMD in that population
the consumption of fresh, uncooked vegetables at least twice per pointed in the same direction, but was statistically significant
week (compared to less than twice per week during the only for the incidence of retinal pigment abnormalities, a less
winter/spring months) was inversely associated with AMD, in
women of ages age 65–74 years.63 Inverse associations with
intakes of vegetables and fruits throughout the rest of the year Zinc and AMD
were also observed, but did not reach statistical significance. In Summary of Evidence
a large prospective follow-up study of women in the Nurses’ Zinc availability influences retinal function in animals and humans.
Health Study, and men in the Health Professionals Follow-up Observational studies report inconsistent relationships between
Study, fruit intake was inversely associated with the risk of moderate variation in long-term zinc intake and AMD.
neovascular AMD, while overall fruit and vegetable intake were High-dose zinc supplementation in one large clinical trial lowered
not associated with AMD.50 Better diets and overall health the progression of intermediate to late AMD. (Results of smaller
habits might explain different associations with vegetable and shorter trials were inconsistent.)
intake from those study samples. Bottom Line and Unanswered Questions
In summary, while there is strong biological plausibility that Moderate intake of foods rich in zinc (dairy, meat, shellfish) might
macular carotenoids might protect against AMD, the results of lower risk for AMD, by virtue of the zinc they provide or because
they also contain other protective vitamins that are poorly studied.
existing epidemiological studies are inconsistent, and have not
• Can relationships of zinc intake to early AMD in some
addressed relationships of macular pigment, specifically, to the
population studies be explained by a protective influence of
progression of AMD. A large clinical trial (AREDS II) is
other nutrients in zinc-rich diets?
underway to determine the impact of lutein and zeaxanthin
• Does moderate intake of zinc from food and supplements slow
supplements on the progression of advanced AMD.
advanced AMD?
The possibility that lutein and zeaxanthin are some of the
High-dose zinc supplementation may modestly slow progression
many components of fruits and vegetables that could protect of AMD among persons who already have intermediate AMD.
against the development of AMD seems strong. However, many • What is the long-term benefit and risk of high-dose zinc
older patients who have been prescribed warfarin treatment to supplementation?
1920 prevent blood clotting have been unnecessarily advised to avoid
Foods and Supplements in the Prevention and Treatment of Age-Related Macular Degeneration

common and more intermediate stage of AMD.14 In a longer-


Dietary Fat and AMD
term prospective study in the Netherlands, zinc intake was
related to lower risk for incident AMD.15 In an Australian study Summary of Evidence
with a similar distribution of zinc intake, early AMD was less High-fat diets in mouse models of AMD induce retinal pathology
thought to contribute to AMD in humans.
prevalent among people with high zinc intake as compared to
In population studies:
low zinc intake, but this was not statistically significant.71
• High intakes of total fat are generally associated with risk of
Incident AMD in the same population was unrelated to zinc
early and late AMD in populations studied
intake.51 The power to evaluate associations between the intake
• The type of fats associated with higher risk varies
of zinc and late AMD was low in these three populations.
considerably; there is some evidence that excess intake of
However, zinc intake was unrelated to self-reported incident
either saturated fats or omega-6 polyunsaturated fatty acids
AMD (a combination of early and late AMD) in two other large
could promote maculopathy
study samples.72
• The intake of fish or omega-3 fatty acids is often associated
By contrast, the use of high-dose zinc supplements (80 mg as
with lower risk for AMD
zinc oxide, along with 2 mg of cupric oxide) for 6 years, with or The body of evidence from in vitro and in vivo animal and clinical
without antioxidants, was associated with modestly lower experiments suggest there is evidence that a protective effect of
progression from intermediate to advanced AMD in the omega-3 fatty acids in the body, in general, may involve one or
AREDS.10 One smaller zinc supplementation trial had pre- more of the following:
viously reported a benefit of zinc supplementation on vision

CHAPTER 145
• Lower oxidative stress
loss in patients with AMD,73 while another did not.74 No • Lower inflammatory cytokines
serious safety issues with zinc supplementation were identified • Improved vascular integrity
in the 6 year AREDS study (aside from more frequent • Less atherogenic blood lipid profiles
hospitalization for genitourinary problems in men and more Bottom Line
frequent anemia, unsupported by differences in hematocrit) and Avoidance of excess fats in foods that provide few micronutrients
zinc supplementation was related to lower mortality in this (processed sweet and salty snacks, and high-fat desserts) and
sample.75 However, the longer-term benefits and risks of zinc favoring omega-3 fatty acids over omega-6 in the diet by eating
supplementation at the high levels tested in AREDS are fatty fish, leafy green vegetables, and whole grains, may lower risk
unknown. While an enhanced immune response has been and progression of AMD.
noted in institutionalized elderly who were supplemented for 1 Unanswered Questions
or 2 years with moderate levels of zinc,76 in combination with The benefit and safety of supplementing with fish oils to slow early
one or more other nutrients, controversy about the long-term or advanced AMD is unknown.
safety of high-dose zinc supplementation exists77 and can only
be addressed with longer-term studies.
power to evaluate associations with either early AMD91 or
DIETARY FAT advanced AMD.87–89 In one short-term prospective study,
persons with low, compared to moderate, levels of dietary fat
There are at least three broad mechanisms by which dietary fats were at higher risk for the 5 year incidence of early AMD.91
might enhance or slow age-related macular degeneration. First,
because of the high caloric density of fats, eating high-fat foods
can displace other foods in the diet which have high nutrient SPECIFIC FATS
density that may have otherwise lowered oxidative stress, or There is inconsistency across studies in the type of fat that was
enhanced macular pigment or immunity. Second, eating high- most related to AMD. This suggests that these relationships
fat and low-nutrient dense foods may contribute to high body may reflect, in part, the caloric density of dietary fats that
mass, which is sometimes reported to be a risk factor for replace other micronutrients. In some samples, high intake of
AMD.78–80 Third, fatty acids themselves have numerous saturated fats was more strongly related to risk for AMD or
biological effects as components of biological membranes and similarly to other types of fat,88–90,93 while in other studies total
regulators of biochemical pathways. Dietary fats could promote intake of polyunsaturated fatty acids or monounsaturated fatty
AMD by promoting atherosclerosis, which is related to AMD acids78,92 was more strongly related. The intake of trans fatty
risk in some studies81,82 or parallel atherosclerosis-like acids, provided in diets by margarines and other processed
processes which result in lipid enrichment and mineralization foods, was related to higher risk for AMD in two studies.78,92
of the retina. Certain fatty acids can also have direct Because fat intake often changes, particularly in relation to
physiological effects on the retina by modulating oxidative diagnosis for cardiovascular diseases, which can be related to
stress, or by the inflammatory response which can promote AMD, these relationships are difficult to interpret. Moreover,
AMD pathogenesis (discussed below). there is limited ability in such studies to adjust for the
numerous other protective aspects of diet that accompany a
more moderate, as compared to high, intake of fat.
OVERALL LEVELS OF FAT In contrast, there is more consistency in epidemiological
In laboratory studies of mouse models of atherosclerosis, studies that report lower risk for AMD among people with high
feeding high-fat diets resulted in the accumulation of lipid-like intakes of fish or long-chain omega-3 polyunsaturated fatty
droplets in the retina and degenerative changes in RPE cells and acids (LC omega-3 PUFAs).90–92,94 The protective influence of
Bruch’s membrane.83–86 In epidemiological studies, there is a omega-3 fatty acids could be somewhat dependent on the
consistent relationship of high dietary fat levels to risk for early omega-6 content of the diet. A higher ratio of omega-3 to -6
and late AMD in observational studies. High total levels of fatty acids could increase formation of antiinflammatory
dietary fat have been related to higher prevalence,87–89 eiscosanoids from omega-3 fatty acids, because the omega-6
incidence,78,90,91 or progression92 of both early and advanced fatty acids compete for the desaturase enzyme that creates
AMD in several different study samples, even though not them95 or replace the omega-6 content of membranes. In one
always statistically significant. Some exceptions to this trend past study, LC omega-3 PUFA intake was stronger among
include prevalence or short-term incidence studies with low subjects who had low intake of omega-6 PUFAs.78 1921
RETINA AND VITREOUS

There has been interest in the health benefits of omega-3 cases of bleeding in trials where patients took warfarin or
fatty acids since the 1970s, when it was observed that aspirin daily.
Greenland Inuit who ingested high levels of fatty fish that In summary, at this time, most population studies to date
contain them, had low rates of cardiovascular disease. Omega- suggest lower early and late AMD among people who eat high
3 fatty acids are one of two major classes of polyunsaturated levels of fish or LC omega-3 PUFAs. The evidence for
fatty acids. Alpha linolenic acid, an omega-3 fatty acid, is one of antioxidant and antiinflammatory properties of these fatty acids
two essential fatty acids that must be obtained from the diet is also supportive of this possibility. However, results of
because they cannot be synthesized by the body. In the body, population studies may reflect other benefits of diets that are
this fatty acid which is contained in leafy green vegetables and high in fish. For example, fatty fish also provide vitamin D,
in larger proportion in some vegetable oils (canola, soy, and which also has antiinflammatory properties, and was recently
flaxseed) can be elongated to make the two major LC omega-3 reported to be associated with lower risk for early AMD in one
PUFAs: eicosapentaenoic acid (EPA) and docosopentaenic acid population (discussed below). The current evidence supports
(DPA); however, the efficiency of conversion is not well the possibility that reducing intake of omega-6 PUFAs and
understood. Fatty fish (salmon, tuna, mackerel, and herring) increasing omega-3 PUFAs of the diet may lower risk for AMD
which concentrate EPA and docosahexaenoic acid (DHA) from with little health risk. This can be achieved by eating leafy green
algae, the primary producers of these fatty acids in the vegetables and fatty fish, substituting canola and olive oils for
ecosystem, are concentrated dietary sources of these LC omega- corn and safflower oils, and avoiding processed foods, selecting
3 PUFAs. These fatty acids are also more concentrated in meat fish with low potential to contain heavy metals (for example, by
SECTION 10

of animals fed grass-based diets than grain-based diets.96 choosing wild caught rather than farm-raised fish) can minimize
There is general agreement that the American diet has potential harmful effects.
changed over the last century, so that the ratio of omega-6 fatty The benefit of supplementing with 1 g per day of LC omega-
acids (found in meat, vegetable oils such as corn, safflower, and 3 PUFAS is currently being tested in a large clinical trial
soy, and processed foods made with these oils) to omega-3 fatty (AREDS II).
acids has increased. American diets provide about 10 times the
amount of omega-6 to -3 fatty acids, and there is a general POSSIBLE PROTECTION BY ASPECTS OF
consensus that consuming more omega-3 fatty acids and less DIET LESS WELL INVESTIGATED
omega-6 would be optimal for health.
LC omega-3 PUFAs, such as DHA, may be particularly
important to the health of the retina. The pathogenesis of AMD VITAMIN D
may be influenced by atherosclerosis or involve parallel Newly emerging evidence for the high prevalence of
processes.97 There is a large body of evidence to suggest that complement H-polymorphisms among people with AMD
EPA and DHA lower CVD mortality by several mechanisms: by suggests that inflammation may play an important role in the
an improvement of blood lipids by lowering blood triglycerides, development of AMD.108–110 High levels of C-reactive protein (a
decreasing inflammation, blood pressure, and platelet aggregation systemic marker of inflammation)111,112 and the use of
and by improving vascular reactivity (recently reviewed).98 antiinflammatory medications113 were significantly related to
These may influence AMD pathogenesis directly or indirectly. AMD, independent of other established risk factors, in some
DHA is the most abundant LC omega-3 PUFA in rod outer but not all114,115 previous studies. Proteins associated with
segment membranes99,100 at a concentration that exceeds levels inflammation including C-reactive protein, fibrinogen, and
found elsewhere in the body.101 Its presence in membranes complement components, as well as transcripts of these
affects their biophysical properties and may influence proteins, have been observed to be entrapped within drusen,
membrane-bound enzymes, receptors, and transport. This is suggesting the involvement of immunocompetent cells in
important in visual transduction,102 but may also influence drusen biogenesis.116 Nutrients which attenuate the
the pathogenesis of AMD. LC omega-3 PUFAs may protect inflammatory response or enhance immunity to pathogens
against AMD by direct influence on retinal cell survival.103 might protect against AMD. Vitamin D is an example of one
DHA has also been demonstrated to protect RPE cells from such food component with antiinflammatory properties.117
oxidative stress.103,104 However, high intake of this type of fat One form of the vitamin, vitamin D3 (cholecalciferol), is
might also lower risk for AMD because of its antiinflammatory synthesized in the skin when exposed to sufficient ultraviolet
properties.95 Numerous cell culture studies provide clues for light. The amount of ultraviolet exposure needed to elevate
possible mechanisms by which LC omega-3 PUFAs could blood vitamin D to recommended levels is small, ~5–15 min
enhance the integrity of vascular and basement membranes and per day with face and hands exposed three times per week in the
prevent neovascularization (recently reviewed105). summer or on any available sunny days in the winter. However,
Unfortunately, none of the over 100 clinical trials of LC aging and sunscreen use can reduce synthesis.118,119 Vitamin D2
omega-3 PUFAs in medical research has evaluated its impact on (ergocalciferol) is also provided by some foods: fortified cows
AMD. However, despite lack of proof of effectiveness in treating milk or soy-milk, cheese, eggs, and fatty fish. However, it has
AMD, previous clinical trials provide a large body of evidence to recently been recognized that many older people obtain
support the notion that omega-3 fatty acids are potent inadequate levels of vitamin D, because they neither consume
antiinflammatory agents and reduce the need for medications the foods that contain it nor get adequate sun exposure to
for the chronic inflammatory disease of rheumatoid arthritis.106 manufacture it in sufficient quantities. Thirty percent of elderly
Also, past clinical trials provide a history about the potential Caucasians sampled in Boston were found to be deficient in
safety of long-chain, omega-3 supplements in about 10,000 vitamin D; the intake of this vitamin is particularly low among
subjects of previous studies. In past studies (previously men and women over 51 years of age in the United States.120
reviewed107 in which 0.3–8 g of DHA and EPA per day was Vitamin D intake reduces C-reactive protein, a marker of
supplemented for periods of 1 week to 7 years, no side effects systemic inflammation.121 Low vitamin D status has been
were reported in 52% of the studies. Overall, side effects were associated with the occurrence of common chronic diseases
reported in fewer than 7% of subjects and involved minor that are suspected to be promoted by inflammatory mecha-
gastrointestinal disturbances (such as diarrhea) and several nisms such as cancers, diabetes, and cardiovascular disease.117
1922
Foods and Supplements in the Prevention and Treatment of Age-Related Macular Degeneration

Higher blood levels of vitamin D were recently reported to be combination of high-fruit and -vegetable diets and low-
associated with lower risk for early AMD in the Third National saturated-fat diets were more protective against coronary heart
Health and Nutrition Examination Survey.122 Consistent disease mortality than was either alone.133
findings in other samples are required to better evaluate this as The possibility that certain specific diet patterns slow the
one of many food components that could protect against AMD. development of early or advanced AMD has not yet been
There were too few cases of advanced AMD to reliably evaluate studied. There does exist evidence in mice for the cooperative
relationships with blood vitamin D in this study, but vitamin D influence of dietary nutrients on the retina in mice: the
might protect against neovascular AMD by virtue of its deposition of basal laminar deposits that are stimulated by diets
antiangiogenic properties in endothelial cells.123,124 This high in polyunsaturated fatty acids is minimized by treatment
remains to be investigated. with vitamin E.85 Because many aspects of diet have the
In summary, vitamin D might be another of the food com- potential to protect against ARM, there may be important
ponents that protects the aging retina. Because this vitamin is insights to be gained by considering overall diet patterns and
provided in milk and fatty fish, it might explain, in part, relation- their relationships to the occurrence of AMD. Certainly, the
ships of these foods to AMD in past studies. There is currently body of evidence about specific nutrients and foods that are
insufficient evidence to reliably determine whether vitamin D related to lower occurrence of AMD support the possibility that
status contributes significantly to protection against AMD. Mediterranean diets or DASH diets could also lower risk for
early or advanced AMD.
Glycemic Index

CHAPTER 145
The glycemic index of foods was recently introduced to be CONSIDERATIONS IN USING
another possible aspect of diet that could influence the SUPPLEMENTS TO TREAT AMD
development of AMD.125 While advanced glycation end
products have been found in drusen, it is not yet known There has been a substantial upswing in the marketing of
whether they are a cause or consequence of degenerative vitamin and mineral supplements for aging eyes. This prompts
changes. Degeneration of the retinal vasculature is a well- questions from patients who have AMD about the use of these
known complication of diabetes mellitus, yet, the presence of supplements to slow progression of this condition and about
diabetes has sometimes, but not always, been related to AMD whether their family members who may be at greater risk will
in epidemiological studies. The biological plausibility that benefit from beginning supplementation to prevent or delay
elevations in blood sugar promote AMD, particularly in the getting the condition themselves. It is interesting to note that
absence of diabetes, remains untested. Nevertheless, diets with while regular supplement use has been practiced by more than
a low glycemic index often include few refined grains and sugars half of Americans for decades, the prevalence of early AMD in
and plenty of fruits, vegetables, whole grains, legumes, and milk one primarily Caucasian American community (in Beaver
which have numerous ingredients that could protect against Dam, Wisconsin) was similar to the prevalence among similar,
AMD. Thus, high-glycemic-index diets, like high-fat diets, may primarily Caucasian, communities in Australia and the
be related to higher rates of AMD, in part or in whole, because Netherlands,134 where the frequency of supplement use is about
they are poorer in a wide variety of protective nutrients and one-third that in the US community. In fact, rates of
other diet components. neovascular AMD were lowest in the Dutch community. The
use of multivitamin supplements is consistently unrelated to
AMD in past epidemiological studies (previously reviewed77,126).
HERBAL SUPPLEMENTS The evidence for benefit of supplements in treating AMD is
The use of herbal supplements has increased in the United limited to one study (previously reviewed77,126,135). The AREDS
States. Several herbal supplements such as those containing demonstrated that taking a high-dose combination antioxidant
ginkgo biloba and bilberry have been promoted to benefit the supplement for 6 years resulted in a 28% reduction of risk for
health of the retina. However, there are no scientific studies that progression to advanced AMD among subjects who already had
support their benefit, except one very small (20 persons) study intermediate AMD.10 Given the limited treatment options for
of ginko biloba in patients with AMD, in which improvement AMD, this may have value for individual persons. It may be
in visual acuity was indicated in a preliminary report (recently that supplement use is more or less helpful in people who are
reviewed126). genetically prone to AMD, but this has not been tested. A
considerable public health benefit of taking the AREDS
supplements has been estimated.136 However, it is not yet
LARGER DIETARY PATTERNS possible to weigh the costs, in terms of health risks of long-term
There is evidence that the pathogenesis of AMD, like many use and whether there would be similar or greater overall health
other chronic diseases of aging, is likely to involve a complex benefit relative to an equal amount of money spent on
interaction of cellular and vascular factors, which may be consuming food sources of antioxidants. It is unknown whether
promoted by light damage, oxidative stress, and inflam- a similar benefit would accrue by the consumption of lower
mation.127 Therefore, it comes as no surprise that numerous levels of antioxidants. While greater benefit might be achieved
nutrients and other dietary components may play multiple roles in slowing AMD with even higher doses of antioxidants, this is
which could be additive or complementary. In many recent not recommended because higher doses have the potential to
studies, specific dietary patterns, which include a high density pose greater health risks. Results of a recent meta-analysis
of fruits and vegetables and whole grains, moderate or low fat suggested that intakes of vitamin E higher than the amount in
levels, and low-fat dairy foods, such as the Mediterranean diet AREDS supplements (above 400 IU) was associated with higher
and Dietary Approaches to Stop Hypertension (DASH) diet, risk for mortality in clinical trials.137 A larger impact might be
have been related to reduced occurrence or progression of many possible with intakes over longer periods. Intakes of beta-
chronic diseases of aging,128–132 and results support the idea that carotene above 20 mg have been associated with higher risk of
the small effects of many individual nutrients or compounds cancer in smokers.138,139
could add up to a stronger effect when the overall diet is It is also unclear whether the results can be generalized to
examined. For example, it was recently demonstrated that the other populations who are less-healthy or consume better or
1923
RETINA AND VITREOUS

be appropriately advised by their physicians to ‘eat well’, just as


The Bottom Line: Summary of Recommendations to
they might be cautioned to stop smoking. A full discussion of
Patients with Intermediate or Late AMD
the details of ‘eating well’ is beyond the scope of this chapter,
Supplements but suggestions in the following section summarize
Taking the AREDS tested supplement containing 500 IU vitamin E, recommendations, as relevant to what we currently know about
15 mg beta-carotene, 500 mg vitamin C, and 80 mg zinc with
AMD, and the role of nutrition in chronic diseases of aging.
2 mg copper may reduce progression.
The last five decades of medical research have demonstrated
Caveats the wisdom of refraining from providing patients with
• The optimal dose and combination of high-dose antioxidants is bifurcated lists of ‘good’ or ‘bad’ foods or nutrients. Over that
unknown time, the results of studies (influenced by market forces), have
• The benefits and safety over more than 6 years is unknown been enlisted to extol the benefits of some (margarine, beta-
• Individual health, diet, and genetic characteristics that enhance carotene, vitamin E) and admonished against indulgence in
or reduce the benefits and safety are unknown others (eggs, red meats, whole-fat dairy, high-cholesterol
Foods shellfish) to prevent chronic diseases like cardiovascular disease
The overall body of current research suggests that healthy eating and cancer. Currently, there is a better understanding of the
may slow the progression of AMD. (See recommendations for complexity of roles of foods in the development of chronic
patients who are at risk for AMD because of a family history of diseases and the value of traditional diets, such as the
AMD.) ‘Mediterranean diet’ are being considered by the medical
SECTION 10

community. The process of adaptation has resulted in


worse diets. People with less nutritious diets might benefit traditional diets, specific to the availability of foods in different
more, but may also be at higher risk for side effects of regions of the world that have promoted health and longevity
pharmacological doses of nutrients if diet imbalances already over thousands of years. Evidence is mounting that certain diet
exist. Insufficient numbers of people in the AREDS, or in all patterns, such as the Mediterranean diet, have more potential
trials combined, exist to determine whether the benefit (or risk) to slow the onset, progression, and recurrence of cardiovascular
is greater in subgroups that have other risk factors for AMD or disease and this may be true for eye disease, as well. The
for specific types of AMD. There were too few people who current consensus of medical research, together with specific
developed early stages of AMD to determine whether these knowledge of relationships of foods and nutrients to AMD, as
high-dose antioxidants had value in slowing progression of reviewed in this chapter, suggests the following as
earlier stages. recommendations to patients to slow early and late AMD.
1. Eat an abundance of fruits and vegetables of varying colors.
SUMMARY AND CONSIDERATIONS IN Currently, the US Dietary Guidelines suggest eating three
RECOMMENDING DIETS TO LOWER RISK to five vegetables per day and two to four servings of fruit
to minimize risk for a variety of chronic diseases. This
OR PROGRESSION OF AMD translates into ~3/4 to 1 cup of vegetables and 1/2 cup of
The knowledge base available to suggest foods that may slow fruit three times a day. Choosing a variety of colors
AMD at early or late stages is currently greater than for naturally varies the nutrients supplied by them. Dark
supplements. The results of the AREDS confirmed that green leafy vegetables are rich in lutein and zeaxanthin,
nutrition is important to the health of the aging macula. but are also rich in a wide variety of micronutrients such
Thus, both patients who have AMD and people who might as vitamins E, C, and other carotenoids. Yellow and orange
be at high risk for AMD (perhaps because of a family history of fruits and vegetables are often sources of vitamin A and
AMD) who inquire about what they can do to slow AMD, can sources of the carotenoids beta-carotene, lutein, and
cryptoxanthin. Red fruits and vegetables often contain not
The Bottom Line: Recommendations for Patients Who only vitamin C, but also other carotenoids, such as
Are at Risk for AMD Because of a Family History of AMD lycopene. Blue and purple fruits supply other
phytochemicals such as flavonoids and anthocyanins that
Supplements have antioxidant and antiinflammatory properties.
There is no evidence from population studies or clinical trials that
2. Eat whole grains at most meals, as breads, cereals, or
supplement use of any kind will lower risk for developing AMD.
pasta. These provide many more B vitamins, vitamin E,
Foods and minerals (such as selenium) than refined grains. They
Current scientific evidence supports the possibility that healthy
are also higher in fiber which can cause one to feel satisfied
eating is the most promising means known (besides avoiding
smoking) to lower risk for developing AMD.
sooner. The current US guidelines suggest eating from 6 to
Because of the broad evidence for benefits of numerous food 11 servings of grains per day, depending on the calories
components, a focus on a balanced diet, rather than food needed. Some controversy exists over the optimal amount
supplements, is more likely to lower risk. and frequency of grains needed in diets. However, there is
Diets which show promise to lower risk for AMD include:
no controversy about the fact that whole grains are
• An abundance of plant foods
nutritionally superior to refined grains.
• Fruits and vegetables of a variety of colors
3. Eat a variety of protein sources daily. Daily high-quality
• Whole grains
protein sources are necessary. US Dietary guidelines
• A variety of daily sources of high-protein foods
suggest that one should eat two to three servings of meat
• Nuts
or alternate protein sources (meat, fish, poultry, eggs, nuts
• Fish
or legumes) and three servings of dairy each day. Many of
• Dairy foods or alternate sources of zinc (meat, shellfish)
these have ingredients that may slow AMD, dairy products
and vitamin D (eggs, fish, and sunlight or supplements)
(milk, cheese, yogurt) are one of two main sources of zinc
• A minimum of high-fat and/or highly sugared foods, especially
in the American diet and many are fortified with vitamin
those made of refined grains
D, as well. If dairy foods are not eaten, calcium and many
• A minimum of processed foods
vitamins can be obtained from beans and other plant
1924 foods, instead. If further research confirms a role of
Foods and Supplements in the Prevention and Treatment of Age-Related Macular Degeneration

vitamin D in slowing AMD, as early findings suggest, then increase omega-3 PUFA intake. Avoiding processed foods
people not eating dairy foods can receive vitamin D by will lower trans-fat intake; since processed foods and oils
eating eggs, fatty fish from cold waters, small frequent provide ~80% of trans fats in the diet, compared to 20%,
(three or more times/week) exposure to sunlight, or taking that occur naturally in food from animal sources.
multivitamins.
Zinc can be obtained from meat, shellfish, or poultry. IMPORTANCE OF FOOD TO AMD PATIENTS
Grass-fed meat contains a more favorable ratio of omega-6 BEYOND NUTRIENTS
to omega-3 fatty acids than grain fed meat. Dark fish from
colder waters (tuna, salmon) is also a source of omega-3 Foods nourish patients with AMD by providing numerous
fatty acids and vitamin D. Eating a variety of seafood and bioactive chemicals that directly or indirectly slow the
concentrating on wild-caught fish reduces concern about pathogenesis of AMD. Additionally, the likelihood that foods
getting high levels of mercury or other toxins from some. that they eat (or perhaps supplements they take if eating
Egg yolk is an easily absorbable source of lutein and healthy foods is not possible) can slow the progression of their
zeaxanthin and contains vitamin D and omega-3 fats. condition may provide a sense of hope, optimism, and self-
Nuts, another protein source, are rich in many types of empowerment that cannot come from the limited medical
vitamin E and omega-3 fatty acids. Variety is the key to treatments that are currently available. The overall current
meeting our need for protein sources, as with other types body of scientific evidence provides solid support for this,
of food. whether or not the long-term impact of isolated components is

CHAPTER 145
4. Eat moderate amounts of fat, reducing processed fats and possible or practical to determine conclusively.
omega-6 fats and increasing omega-3 fatty acids. Excess fat Foods can also stimulate the senses. This may be particularly
in the diet lowers the overall nutrient density because nourishing to the patient with AMD who is becoming aware
high-fat foods (such as from high-fat desserts and salty of diminished sense of sight. The role of food in connecting
snacks) are typically low in vitamins and minerals and the person to his or her surroundings is heightened in patients
replace nutrient-dense foods. However, some fat may be with AMD (or any significant vision loss). Since food lies at the
part of a healthy diet for people with or at risk for AMD. center of many of the activities and rituals that bring meaning
Fat is not only important in enhancing the natural flavor and joy to one’s life, the smells and tastes of traditional foods
of foods but also aids digestion of fat-soluble plant can reinforce connections to cherished past experiences.
pigments. For example, salads to which full-fat salad Physicians might consider supplementing their recommen-
dressings or avocados have been added raise blood dations, based on evidence for the pharmacological impact of
carotenoids more than eating fat-free salads that are food components on the pathogenetic process that promote
equally high in carotenoids.140,141 Current AMD, with encouragement toward a focus on enjoying
recommendations to lower omega-6 and increase omega-3 healthful eating.
fats in the diet can be achieved by reducing processed Decades of medical research both inform us of the large
foods, replacing corn oils and margarines with canola, nut, number of vitamins, minerals, and phytochemicals that are
and olive oils, and increasing the intake of cold-water fish, currently known to contribute to retinal health, and foster
which provide LC omega-3 PUFAs. Substituting meat that within us an appreciation of the probability that others, of
has increasingly come from grain-fed animals, with meat which we may not be presently aware, are likely to be elucidated
from traditional grass-fed animals, is another option to in the near future.

REFERENCES
1. Mares-Perlman JA, Klein R: Diet and age- protection against photosensitized oxidation. 13. West S, Vitale S, Hallfrisch J, et al: Are
related macular degeneration. In: Taylor A, Free Radic Biol Med 2003; 35:1319–1329. antioxidants or supplements protective for
ed. Nutritional and environmental 8. Wrona M, Rozanowska M, Sarna T: age-related macular degeneration? Arch
influences on the eye. Boca Raton, FL: Zeaxanthin in combination with ascorbic Ophthalmol 1994; 112:222–227.
CRC Press; 1999:181–214. acid or alpha-tocopherol protects ARPE-19 14. VandenLangenberg GM, Mares-Perlman
2. Handelman GJ, Dratz EA: The role of cells against photosensitized peroxidation JA, Klein R, et al: Associations between
antioxidants in the retina and retinal of lipids. Free Radic Biol Med 2004; antioxidant and zinc intake and the 5-year
pigment epithelium and the nature of 36:1094–1101. incidence of early age-related maculopathy
pro-oxidant damage. Adv Free Radic Biol 9. Lu L, Hackett SF, Mincey A, et al: Effects of in the Beaver Dam Eye Study. Am J
Med 1986; 2:1–89. different types of oxidative stress in RPE Epidemiol 1998; 148:204–214.
3. Beatty S, Koh H, Phil M, et al: The role of cells. J Cell Physiol 2006; 206:119–125. 15. van Leeuwen R, Boekhoorn S, Vingerling
oxidative stress in the pathogenesis of age- 10. A randomized, placebo-controlled, clinical JR, et al: Dietary intake of antioxidants and
related macular degeneration. Surv trial of high-dose supplementation with risk of age-related macular degeneration.
Ophthalmol 2000; 45:115–134. vitamins C and E, beta carotene, and zinc JAMA 2005; 294:3101–3107.
4. Teikari JM, Laatikainen L, Virtamo J, et al: for age-related macular degeneration and 16. Delcourt C, Carriere I, Delage M, et al:
Six-year supplementation with alpha- vision loss: AREDS report no. 8. Arch Plasma lutein and zeaxanthin and other
tocopherol and beta-carotene and age- Ophthalmol 2001; 119:1417–1436. carotenoids as modifiable risk factors for
related maculopathy. Acta Ophthalmol 11. Moriarty-Craige SE, Adkison J, Lynn M, et al: age-related maculopathy and cataract: The
Scand 1998; 76:224–229. Antioxidant supplements prevent oxidation POLA Study. Invest Ophthalmol Vis Sci
5. Taylor HR, Tikellis G, Robman LD, et al: of cysteine/cystine redox in patients with 2006; 47:2329–2335.
Vitamin E supplementation and macular age-related macular degeneration. Am J 17. Cardinault N, Abalain J-H, Sairafi B et al:
degeneration: randomised controlled trial. Ophthalmol 2005; 140:1020–1026. Lycopene but not lutein nor zeaxanthin
BMJ 2002; 325:11. 12. Richer S, Stites W, Statkute L, et al: decreases in serum and lipoproteins in
6. Stahl W, Junghans A, de Boer B, et al: Double-masked, placebo-controlled, age-related macular degeneration patients.
Carotenoid mixtures protect multilamellar randomized trial of lutein and antioxidant Clinica Chimica Acta2005; 357:34–42.
liposomes against oxidative damage: supplementation in the intervention of 18. Mares-Perlman JA, Klein R, Klein BE, et al:
synergistic effects of lycopene and lutein. atrophic age-related macular degeneration: Association of zinc and antioxidant
FEBS Lett 1998; 427:305–308. the Veterans LAST study (Lutein nutrients with age-related maculopathy.
7. Wrona M, Korytowski W, Rozanowska M, Antioxidant Supplementation Trial). Arch Ophthalmol 1996; 114:991–997.
et al: Cooperation of antioxidants in Optometry 2004; 75:216–2130. 19. Gale CR, Hall NF, Phillips DI, et al: Lutein 1925
RETINA AND VITREOUS

and zeaxanthin status and risk of age- 34. Sparrow JR, Vollmer-Snarr HR, Zhou J, and nutrition examination survey. Am J
related macular degeneration. Invest et al: A2E-epoxides Damage DNA in Epidemiol 2001; 153:424–432.
Ophthalmol Vis Sci 2003; 44:2461–2465. Retinal Pigment Epithelial Cells. Vitamin E 50. Cho E, Seddon JM, Rosner B, et al:
20. Moeller SM, Mehta NR, Tinker LF, et al: and other antioxidants inhibit A2E-epoxide Prospective study of intake of fruits,
Associations between intermediate age- formation. J Biol Chem 2003; vegetables, vitamins, and carotenoids and
related macular degeneration and lutein 278:18207–18213. risk of age-related maculopathy. Arch
and zeaxanthin in the Carotenoids in Age- 35. Rapp LM, Maple SS, Choi JH: Lutein and Ophthalmol 2004; 122:883–892.
Related Eye Disease Study (CAREDS), an zeaxanthin concentrations in rod outer 51. Flood V, Smith W, Wang JJ, et al: Dietary
ancillary study of the Women’s Health segment membranes from perifoveal and antioxidant intake and incidence of early
Initiative. Arch Ophthalmol 2006; 124:1–24. peripheral human retina. Invest Ophthalmol age-related maculopathy: the Blue
21. Fowke JH, Morrow JD, Motley S, et al: Vis Sci 2000; 41:1200–1209. Mountains Eye Study. Ophthalmology
Brassica vegetable consumption reduces 36. Khachik F, Bernstein PS, Garland DL: 2002; 109:2272–2278.
urinary F2-isoprostane levels independent Identification of lutein and zeaxanthin 52. Flood V, Rochtchina E, Wang JJ, et al:
of micronutrient intake. Carcinogenesis Oct oxidation products in human and monkey Lutein and zeaxanthin dietary intake and
2006; 27:2096–2102. retinas. Invest Ophthalmol Vis Sci 1997; age related macular degeneration. Br J
22. Mares-Perlman JA, Klein R: Diet and 38:1802–1811. Ophthalmol 2006; 90:927–928.
age-related macular degeneration. In: 37. Sujak A, Gabrielska J, Grudzinski W, et al: 53. Mares-Perlman JA, Klein R, Klein BE, et al:
Taylor A, ed. Nutritional and environmental Lutein and zeaxanthin as protectors of lipid Association of zinc and antioxidant
influences on the eye. Boca Raton, FL: membranes against oxidative damage: the nutrients with age-related maculopathy.
CRC Press; 1999:181–214. structural aspects. Arch Biochem Biophys Arch Ophthalmol 1996; 114:991–997.
23. Moeller SM, Jacques PF, Blumberg JB: 1999; 371:301–307. 54. Mares-Perlman JA, Brady WE, Klein R,
SECTION 10

The potential role of dietary xanthophylls in 38. Malinow MR, Feeney-Burns L, Peterson et al: Serum antioxidants and age-related
cataract and age-related macular LH, et al: Diet-related macular anomalies in macular degeneration in a population-
degeneration. J Am Coll Nutr 2000; 19(5 monkeys. Invest Ophthalmol Vis Sci 1980; based case-control study. Arch Ophthalmol
Suppl):522S–527S. 19:857–863. 1995; 113:1518–1523.
24. Bone RA, Landrum JT, Fernandez L, et al: 39. Leung IY, Sandstrom MM, Zucker CL, et al: 55. Mares J: Carotenoids and eye disease:
Analysis of the macular pigment by HPLC: Nutritional manipulation of primate retinas, epidemiologic evidence. In: Krinsky NI,
retinal distribution and age study. Invest II: effects of age, n-3 fatty acids, lutein, and Mayne S, eds. Carotenoids in health &
Ophthalmol Vis Sci 1988; 29:843–849. zeaxanthin on retinal pigment epithelium. disease. New York, NY: Marcel Dekker;
25. Bernstein PS, Khachik F, Carvalho LS, et al: Invest Ophthalmol Vis Sci 2004; 2003.
Identification and quantitation of 45:3244–3256. 56. Hammond BR, Jr, Johnson EJ, Russell RM,
carotenoids and their metabolites in the 40. Feeney-Burns L, Neuringer M, Gao CL: et al: Dietary modification of human macular
tissues of the human eye. Exp Eye Res Macular pathology in monkeys fed pigment density. Invest Ophthalmol Vis Sci
2001; 72:215–223. semipurified diets. Prog Clin Biol Res 1989; 1997; 38:1795–1801.
26. Yemelyanov AY, Katz NB, Bernstein PS: 314:601–622. 57. Bone RA, Landrum JT, Guerra LH, et al:
Ligand-binding characterization of 41. Thomson LR, Toyoda Y, Langner A, et al: Lutein and zeaxanthin dietary supplements
xanthophyll carotenoids to solubilized Elevated retinal zeaxanthin and prevention raise macular pigment density and serum
membrane proteins derived from human of light-induced photoreceptor cell death in concentrations of these carotenoids in
retina. Exp Eye Res 2001; 72:381–392. quail. Invest Ophthalmol Vis Sci 2002; humans. J Nutr 2003; 133:992–998.
27. Bhosale P, Larson AJ, Frederick JM, et al: 43:3538–3549. 58. Aleman TS, Duncan JL, Bieber ML, et al:
Identification and characterization of a Pi 42. Bone RA, Landrum JT, Mayne ST, et al: Macular pigment and lutein
isoform of glutathione S-transferase Macular pigment in donor eyes with and supplementation in retinitis pigmentosa and
(GSTP1) as a zeaxanthin-binding protein in without AMD: a case-control study. Invest Usher syndrome. Invest Ophthalmol Vis Sci
the macula of the human eye. J Biol Chem Ophthalmol Vis Sci 2001; 42:235–240. 2001; 42:1873–1881.
2004; 279:49447–49454. 43. Beatty S, Murray IJ, Henson DB, et al: 59. Bowen PE, Garg V, Stacewicz-Sapuntzakis
28. Handelman GJ, Nightingale ZD, Macular pigment and risk for age-related M, et al: Variability of serum carotenoids in
Lichtenstein AH, et al: Lutein and macular degeneration in subjects from a response to controlled diets containing six
zeaxanthin concentrations in plasma after Northern European population. Invest servings of fruits and vegetables per day.
dietary supplementation with egg yolk. Am Ophthalmol Vis Sci 2001; 42:439–446. Ann NY Acad Sci 1993; 691:241–243.
J Clin Nutr 1999; 70:247–2451. 44. Bernstein PS, Zhao DY, Wintch SW, et al: 60. Bowen PE, Herbst-Espinosa SM, Hussain
29. U.S. Department of Agriculture, Agricultural Resonance Raman measurement of EA, et al: Esterification does not impair
Research Service: USDA National Nutrient macular carotenoids in normal subjects lutein bioavailability in humans. J Nutr
Database for Standard Reference, Release and in age-related macular degeneration 2002; 132:3668–3673.
16-1. 2004 Jul 29, 2004. Available: patients. Ophthalmology 2002; 61. Mares JA, LaRowe TL, Snodderly DM, et al:
http://www.nal.usda.gov/fnic/foodcomp/ 109:1780–1787. Predictors of macular pigment density in the
Data/SR16-1/wtrank/16-1w338.pdf Aug 5, 45. La Rowe TL, Snodderly DM, et al: Macular Carotenoids in the Age-Related Eye Disease
2004. pigment and age-related maculopathy in Study. Am J Clin Nutr 2006; in press.
30. Mares-Perlman JA, Fisher AI, Klein R, et al: the carotenoids in age-related eye disease 62. Goldberg J, Flowerdew G, Smith E, et al:
Lutein and zeaxanthin in the diet and study, an ancillary study of the women’s Factors associated with age-related
serum and their relation to age-related health initiative. Am J Clin Nutr Manuscript macular degeneration. An analysis of data
maculopathy in the third national health under review, 2006. from the first National Health and Nutrition
and nutrition examination survey. Am J 46. Antioxidant status and neovascular age- Examination Survey. Am J Epidemiol 1988;
Epidemiol 2001; 153:424–432. related macular degeneration. Eye Disease 128:700–710.
31. Snodderly DM, Brown PK, Delori FC, et al: Case-Control Study Group. Arch 63. Vaicaitiene R, Luksiene DK, Paunksnis A,
The macular pigment. I. Absorbance Ophthalmol 1993; 111:104–109. et al: Age-related maculopathy and
spectra, localization, and discrimination 47. Snellen EL, Verbeek AL, Van Den Hoogen consumption of fresh vegetables and fruits
from other yellow pigments in primate GW, et al: Neovascular age-related macular in urban elderly. Medicina (Kaunas) 2003;
retinas. Invest Ophthalmol Vis Sci 1984; degeneration and its relationship to 39:1231–1236.
25:660–673. antioxidant intake. Acta Ophthalmol Scand 64. Karcioglu ZA, Zinc in the eye. Surv
32. Junghans A, Sies H, Stahl W: Macular 2002; 80:368–371. Ophthalmol 1982; 27:114–122.
pigments lutein and zeaxanthin as 48. Seddon J AU, Sperduto R et al: Dietary 65. Trumbo P, Yates A, Schlicker A, et al:
blue light filters studied in liposomes. carotenoids, vitamins A, C, and E, and Scientific evaluation of dietary reference
Arch Biochem Biophys 2001; 391:160–164. advanced age related macular intake. Dietry reference intakes (DRI) for
33. Hammond JBR, Wooten BR, Curran- degeneration. JAMA 1994; 272:1413–1420. vitamin A, vitamin K, arsenic, boron,
Celentano J: Carotenoids in the retina and 49. Mares-Perlman JA, Fisher AI, Klein R, et al: chromium, copper, iodine, iron, maganese,
lens: possible acute and chronic effects on Lutein and zeaxanthin in the diet and molybderum, nickel, silicon, vandadium,
human visual performance. Arch Biochem serum and their relation to age- related and zinc. Washington DC: National
1926 Biophys 2001; 385:41–46. maculopathy in the third national health Academy Press; 2000.
Foods and Supplements in the Prevention and Treatment of Age-Related Macular Degeneration

66. Tate DJ Jr, Miceli MV, Newsome DA: 84. Kliffen M, Lutgens E, Daemen MJ, et al: 101. Arterburn LM, Hall EB, Oken H:
Zinc protects against oxidative damage in The APO(*)E3-Leiden mouse as an animal Distribution, interconversion, and dose
cultured human retinal pigment epithelial model for basal laminar deposit. Br J response of n-3 fatty acids in humans.
cells. Free Radic Biol Med 1999; Ophthalmol 2000; 84:1415–1419. Am J Clin Nutr 2006; 83:S1467–1476.
26:704–713. 85. Espinosa-Heidmann DG, Sall J, Hernandez 102. Litman BJ, Niu SL, Polozova A, et al: The
67. Mocchegiani E, Muzzioli M, Giacconi R: EP, et al: Basal laminar deposit formation in role of docosahexaenoic acid containing
Zinc, metallothioneins, immune responses, APO B100 transgenic mice: complex phospholipids in modulating G protein-
survival and ageing. Biogerontology 2000; interactions between dietary fat, blue light, coupled signaling pathways: visual
1:133–143. and vitamin E. Invest Ophthalmol Vis Sci transduction. J Mol Neurosci 2001;
68. Shankar AH, Prasad AS: Zinc and immune 2004; 45:260–266. 16:237–242; discussion 279–284.
function: the biological basis of altered 86. Dithmar S, Sharara NA, Curcio CA, et al: 103. Rotstein NP, Politi LE, German OL, et al:
resistance to infection. Am J Clin Nutr Murine high-fat diet and laser Protective effect of docosahexaenoic acid
1998; 68(2 Suppl):447S–463S. photochemical model of basal deposits in on oxidative stress-induced apoptosis of
69. Hyun HJ, Sohn JH, Ha DW, et al: Depletion Bruch membrane. Arch Ophthalmol 2001; retina photoreceptors. Invest Ophthalmol
of intracellular zinc and copper with TPEN 119:1643–1649. Vis Sci 2003; 44:2252–2259.
results in apoptosis of cultured human 87. Heuberger RA, Mares-Perlman JA, Klein R, 104. Mukherjee PK, Marcheselli VL, Serhan CN,
retinal pigment epithelial cells. Invest et al: Relationship of dietary fat to age- et al: Neuroprotectin D1: a docosahexaenoic
Ophthalmol Vis Sci 2001; 42:460–465. related maculopathy in the Third National acid-derived docosatriene protects human
70. Wood JPM, Osborne NN: Zinc and energy Health and Nutrition Examination Survey. retinal pigment epithelial cells from
requirements in induction of oxidative Arch Ophthalmol 2001; 119:1833–1838. oxidative stress. Proc Natl Acad Sci USA

CHAPTER 145
stress to retinal pigmented epithelial cells. 88. Mares-Perlman JA, Brady WE, Klein R, 2004; 101:8491–8496.
Neurochem Res 2003; 28:1525–1533. et al: Dietary fat and age-related 105. SanGiovanni JP, Chew EY: The role of
71. Smith W, Mitchell P, Webb K, et al: Dietary maculopathy. Arch Ophthalmol 1995; omega-3 long-chain polyunsaturated fatty
antioxidants and age-related maculopathy: 113:743–748. acids in health and disease of the retina.
the Blue Mountains Eye Study. 89. Smith W, Mitchell P, Leeder SR: Dietary fat Prog Retin Eye Res 2005; 24:87–138.
Ophthalmology 1999; 106:761–767. and fish intake and age-related 106. Calder PC: n-3 polyunsaturated fatty
72. Cho E, Stampter MK, Seddon JM, et al: maculopathy. Arch Ophthalmol 2000; acids, inflammation, and inflammatory
Prospective study of zinc intake and the 118:401–404. diseases. Am J Clin Nutr 2006; 83(6
risk of age-related macular degeneration. 90. Cho E, Hung S, Willett WC, et al: Suppl):1505S–1519S.
Ann Epidemiol 2001; 11:328–336. Prospective study of dietary fat and the risk 107. Wang C, Chung M, Lichtenstein AH, et al:
73. Newsome DA, Swartz M, Leone NC, et al: of age-related macular degeneration. Am J Effects of Omega-3 fatty acids on
Oral zinc in macular degeneration. Arch Clin Nutr 2001; 73:209–218. cardiovascular disease. evidence
Ophthalmol 1988; 106:192–198. 91. Chua B, Flood V, Rochtchina E, et al: report/technology assessment No. 94
74. Stur M, Tittl M, Reitner A, et al: Oral zinc Dietary fatty acids and the 5-year incidence (Prepared by Tufts-New England Medical
and the second eye in age-related macular of age-related maculopathy. Arch Center Evidence-Based Practice Center
degeneration. Invest Ophthalmol Vis Sci Ophthalmol 2006; 124:981–986. under contract no. 290-02-0022.) AHRQ
1996; 37:1225–1235. 92. Seddon JM, Cote J, Rosner B: Progression Publication No. 04-E013–2. Rockville, MD:
75. Clemons TE, Kurinij N, Sperduto RD: of age-related macular degeneration: Agency for Healthcare Research and
Associations of mortality with ocular association with dietary fat, Quality; 2004.
disorders and an intervention of high-dose transunsaturated fat, nuts, and fish intake. 108. Klein RJ, Zeiss C, Chew EY, et al:
antioxidants and zinc in the Age-Related Arch Ophthalmol 2003; 121:1728–1737. Complement factor H polymorphism in
Eye Disease Study: AREDS Report No. 13. Erratum. Arch Ophthalmol 2004; 122:426. age-related macular degeneration. Science
Arch Ophthalmol 2004; 122:716–726. 93. Seddon JM, Cote J, Rosner B: Progression 2005; 308:385–389.
76. Chandra RK: Effect of vitamin and trace- of age-related macular degeneration: 109. Haines JL, Hauser MA, Schmidt S, et al:
element supplementation on immune association with dietary fat, Complement factor H variant increases the
responses and infection in elderly subjects. transunsaturated fat, nuts, and fish intake. risk of age-related macular degeneration.
Lancet 1992; 340:1124–1127. Arch Ophthalmol 2003; 121:1728–1737. Science 2005; 308:419–421.
77. Mares JA, La Rowe TL, Blodi BA: Doctor, 94. Seddon JM, George S, Rosner B: Cigarette 110. Edwards AO, Ritter R, 3rd, Abel KJ, et al:
what vitamins should I take for my eyes? smoking, fish consumption, Omega-3 Complement factor H polymorphism and
Arch Ophthalmol 2004; 122:628–635. fatty acid intake, and associations with age-related macular degeneration. Science
78. Seddon JM, Rosner B, Sperduto RD, et al age-related macular degeneration: The US 2005; 308:421–424.
Dietary fat and risk for advanced age- Twin Study of age-related macular 111. Seddon JM, Gensler G, Milton RC, et al:
related macular degeneration. Arch degeneration. Arch Ophthalmol 2006; Association between C-reactive protein and
Ophthalmol 2001; 119:1191–1199. 124:995–1001. age-related macular degeneration. JAMA
79. Schaumberg DA, Christen WG, Hankinson 95. Larsson SC, Kumlin M, Ingelman-Sundberg 2004; 291:704–710.
SE, et al: Body mass index and the M, et al: Dietary long-chain n-3 fatty acids 112. Seddon JM, George S, Rosner B, et al:
incidence of visually significant age-related for the prevention of cancer: a review of Progression of age-related macular
maculopathy in men. Arch Ophthalmol potential mechanisms. Am J Clin Nutr degeneration: prospective assessment of
2001; 119:1259–1265. 2004; 79:935–945. C-reactive protein, interleukin 6, and other
80. Seddon JM, Cote J, Davis N, et al: 96. Wood JD, Enser M: Factors influencing cardiovascular biomarkers. Arch
Progression of age-related macular fatty acids in meat and the role of Ophthalmol 2005; 123:774–782.
degeneration: association with body mass antioxidants in improving meat quality. 113. Clemons TE, Milton RC, Klein R, et al: Risk
index, waist circumference, and waist-hip Br J Nutr 1997; 78(Suppl 1):S49–S60. factors for the incidence of advanced age-
ratio. Arch Ophthalmol 2003; 121:785–792. 97. Snow KK, Seddon JM: Do age-related related macular degeneration in the Age-
81. Leeuwen VR, Ikram MK, Vingerling JR, macular degeneration and cardiovascular Related Eye Disease Study (AREDS)
et al: Blood pressure, atherosclerosis, and disease share common antecedents? AREDS report no. 19. Ophthalmology 2005;
the incidence of age-related maculopathy: Ophthalmic Epidemiol 1999; 6:125–143. 112:533–539.
the Rotterdam Study. Invest Ophthalmol Vis 98. Breslow JL: n-3 Fatty acids and 114. Klein R, Klein BE, Marino EK, et al: Early
Sci 2003; 44:3771–3777. cardiovascular disease. Am J Clin Nutr age-related maculopathy in the
82. Vingerling JR, Dielemans I, Hofman A, Bots 2006; 83:S1477–1482. cardiovascular health study. Ophthalmology
M: Age-related Macular Degeneration is 99. Bazan NG, Scott BL: Dietary omega-3 2003; 110:25–33.
Assoiciated with Atherosclerosis. Am J fatty acids and accumulation of 115. Klein R, Klein BE, Knudtson MD, et al:
Epidemiol 1995; 142:404–409. docosahexaenoic acid in rod photoreceptor Systemic markers of inflammation,
83. Miceli MV, Newsome DA, Tate DJ, Jr., et al: cells of the retina and at synapses. Ups J endothelial dysfunction, and age-related
Pathologic changes in the retinal pigment Med Sci 1990; 48(Suppl):97–107. maculopathy. Am J Ophthalmol 2005;
epithelium and Bruch’s membrane of fat- 100. Fliesler SJ, Anderson RE: Chemistry and 140(1):35–44.
fed atherogenic mice. Curr Eye Res 2000; metabolism of lipids in the vertebrate 116. Anderson DH MR, Hageman GS et al:
20:8–16. retina. Prog Lipid Res 1983; 22:79–131. A role for local inflammation in the 1927
RETINA AND VITREOUS

formation of drusen in the aging eye. Am J degeneration. Am J Clin Nutr 2006; alone: the Baltimore Longitudinal Study of
Ophthalmol 2002; 134:411–431. 83:880–886. Aging. J Nutr 2005; 135:556–561.
117. Holick MF, Vitamin D: importance in the 126. West AL, GA Oren and SE Moroi Evidence 134. Smith W, Assink J, Klein R, et al: Risk
prevention of cancers, type 1 diabetes, for the Use of Nutritional Supplements and factors for age-related macular
heart disease, and osteoporosis. Am J Clin Herbal Medicines in Common Eye degeneration: Pooled findings from three
Nutr 2004; 79:362–371. Diseases. American Journal of continents. Ophthalmology 2001;
118. MacLaughlin J, Holick MF: Aging Ophthalmology 2006; 141:157–166. 108:697–704.
decreases the capacity of human skin to 127. Hageman GS, Luthert PJ, Victor Chong 135. Evans JR, Antioxidant vitamin and mineral
produce vitamin D3. J Clin Invest 1985; NH, et al: An integrated hypothesis that supplements for slowing the progression of
76:1536–1538. considers drusen as biomarkers of age-related macular degeneration. Cochrane
119. Matsuoka LY, Ide L, Wortsman J, et al: immune-mediated processes at the RPE- Database Syst Rev 2006:CD000254.
Sunscreens suppress cutaneous vitamin Bruch’s membrane interface in aging and 136. Bressler NM, Bressler SB, Congdon NG,
D3 synthesis. J Clin Endocrinol Metab age-related macular degeneration. Prog et al: Potential public health impact of Age-
1987; 64:1165–1168. Retin Eye Res 2001; 20:705–732. Related Eye Disease Study results: AREDS
120. Moore C, Murphy MM, Keast DR, et al: 128. Hoffmann K, Zyriax B-C, Boeing H, et al: report no. 11. Arch Ophthalmol 2003;
Vitamin D intake in the United States. J Am A dietary pattern derived to explain 121:1621–1624.
Diet Assoc 2004; 104:980–983. biomarker variation is strongly associated 137. Miller ER, 3rd, Pastor-Barriuso R, Dalal D,
121. Timms PM, Mannan N, Hitman GA, et al: with the risk of coronary artery disease. Am et al: Meta-analysis: high-dosage Vitamin E
Circulating MMP9, vitamin D and variation J Clin Nutr 2004; 80:633–640. supplementation may increase all-cause
in the TIMP-1 response with VDR 129. Moeller SM, Taylor A, Tucker KL, et al: mortality. Ann Intern Med 2004;
genotype: mechanisms for inflammatory Overall adherence to the dietary guidelines 142(1):37–46.
SECTION 10

damage in chronic disorders? QJM 2002; for americans is associated with reduced 138. The effect of vitamin E and beta carotene
95:787–796. prevalence of early age-related nuclear lens on the incidence of lung cancer and other
122. Parekh N, Chappell R, Millen A, et al: opacities in women. J Nutr 2004; cancers in male smokers. The Alpha-
Association between Vitamin D and age- 134:1812–1819. Tocopherol, Beta Carotene Cancer
related macular degeneration in the Third 130. Appel LJ, Champagne CM, Harsha DW, Prevention Study Group. N Engl J Med
National Health and Nutrition Examination et al: Effects of comprehensive lifestyle 1994; 330:1029–1035.
Survey 1988–94. Arch Ophthalmol, 2007; modification on blood pressure control: 139. Omenn GS, Goodman GE, Thornquist MD,
125:661–669. main results of the PREMIER clinical trial. et al: Effects of a combination of beta
123. Iseki K, Tatsuta M, Uehara H, et al: JAMA 2003; 289:2083–2093. carotene and vitamin A on lung cancer and
Inhibition of angiogenesis as a mechanism 131. Lopes HF, Martin KL, Nashar K, et al: cardiovascular disease. N Engl J Med
for inhibition by 1alpha-hydroxyvitamin D3 DASH diet lowers blood pressure and lipid- 1996; 334:1150–1155.
and 1,25-dihydroxyvitamin D3 of colon induced oxidative stress in obesity. 140. Brown ED, Micozzi MS, Craft NE, et al:
carcinogenesis induced by azoxymethane Hypertension 2003; 41:422–430. Plasma carotenoids in normal men after a
in Wistar rats. Int J Cancer 1999; 132. de Lorgeril M, Salen P, Martin JL, et al: single ingestion of vegetables or purified
81:730–733. Mediterranean diet, traditional risk factors, beta-carotene. Am J Clin Nutr 1989;
124. Bernardi RJ, Johnson CS, Modzelewski RA, and the rate of cardiovascular 49:1258–1265.
et al: Antiproliferative effects of 1alpha, complications after myocardial infarction: 141. Unlu ZN, Bohn T, Clinton S, Schwartz SJ:
25-dihydroxyvitamin D(3) and vitamin D final report of the Lyon Diet Heart Study. Enhanced carotenoid absorption in humans
analogs on tumor-derived endothelial cells. Circulation 1999; 99:779–785. from salad and salsa with addition of
Endocrinology 2002; 143:2508–2514. 133. Tucker KL, Hallfrisch J, Qiao N, et al: The avocado or avocado oil. J Nutr 2005;
125. Chiu CJ, Hubbard LD, Armstrong J, et al: combination of high fruit and vegetable and 135:3.
Dietary glycemic index and carbohydrate in low saturated fat intakes is more protective
relation to early age-related macular against mortality in aging men than is either

1928
CHAPTER
Age-Related Macular Degeneration: Choroidal
146 Neovascularization
Neil M. Bressler, Susan B. Bressler, and Evangelos S. Gragoudas

Although most patients with age-related macular degeneration process of CNV (see end of this section on Pathogenesis).
(AMD) manifest only drusen or abnormalities of the retinal Although inflammatory cells have been shown histologically
pigment epithelium (RPE), the majority of patients who are at to be associated with the presence of the nonneovascular and
risk for severe loss of vision from AMD can lose vision because neovascular stages of AMD, studies have not shown that
of the development of choroidal neovascularization (CNV) and inflammatory cells necessarily are directly linked to the devel-
related manifestations such as serous or hemorrhagic detach- opment of CNV. Therefore, these studies cannot determine
ment of the RPE and fibrovascular disciform scarring. The whether these inflammatory cells represent a response to existing
pathogenesis, clinical features, differential diagnosis, impact on degenerative changes within Bruch’s membrane or whether the
quality of life, and mangement of the neovascular form of AMD inflammatory cells act as essential mediators of degeneration,
are reviewed in this chapter. (The nonneovascular form of with subsequent development of CNV.
AMD, including drusen and abnormalities of the RPE such as Experimentally produced CNV developing around retinal
geographic atrophy, is reviewed in Chapter 144). laser burns in the monkey eye have also shown the presence of
macrophages at the site of developing CNV.16,17 Again, it is
PATHOGENESIS not known whether these macrophages represent a response to
damaged retina from a laser burn or whether they act as
mediators of CNV. Also, this model of CNV differs from CNV
PATHOGENESIS OF DEVELOPMENT OF CNV in AMD in that experimentally produced CNV proliferates
The understanding of the pathogenesis of the development of internal to the RPE (between the sensory retina and the RPE),16
CNV continues to evolve. In part, clinical1–5 and histologic whereas CNV associated with AMD initially proliferates
reports6–9 suggest that the presence of diffuse thickening of the external to the RPE (within the thickened inner aspect of Bruch’s
inner aspect of Bruch’s membrane (associated with large, soft membrane).6–8
drusen clinically) predisposes Bruch’s membrane to develop A relationship of scleral rigidity with CNV in AMD has been
cracks through which ingrowth of new vessels from the suggested in a pilot study in which increased scleral rigidity
choriocapillaris can occur. This hypothesis is supported by the was found to be associated with the presence of AMD.18 The
finding of CNV in other pathologic entities in which breaks in possibility of compromised blood flow in the vortex veins by
Bruch’s membrane occur, such as pathologic myopia10 and progressively increased scleral rigidity was hypothesized to
angioid streaks.11 However, it is unlikely that a break in Bruch’s account for these findings, but further investigation is required.
membrane alone necessarily predisposes to the development of Results from epidemiologic studies19 also demonstrated that
CNV. Histologic studies have shown that in eyes with AMD, the risk of CNV is associated with low blood levels of
breaks in Bruch’s membrane can be identified not only in areas micronutrients with antioxidant potential, cigarette smoking,
of CNV but also in areas in which no new vessels can be and risk factors for cardiovascular disease. Systemic hypertension
identified.6 also has been reported to be a risk factor for developing CNV in
Experimental studies have also suggested that other cellular the fellow eye of a patient who presents with CNV in the first
processes may have a role in the development of the CNV eye.2 Decreased risk was associated with higher levels of
beyond merely a disturbance in Bruch’s membrane. Laboratory carotenoids and use of postmenopausal exogenous estrogens
studies have shown that endothelial cells can elaborate enzymes in women. The associations with estrogens, cigarette smoking,
necessary for the digestion of a basement membrane such as and serum cholesterol are intriguing because sphingomyelins
Bruch’s membrane.12 This finding would support the concept and cholesterol esters similar to those found in arteriosclerotic
that endothelial cells from the choriocapillaris could produce a plaques are found in aging sclera.20 Perhaps these factors in
break in Bruch’s membrane, rather than presuming that CNV some way account for increased scleral rigidity.
grows only through preexisting breaks in Bruch’s membrane.
Other reports have suggested that a granulomatous inflam-
matory response to degenerated Bruch’s membrane may be an PATHOGENESIS OF PROGRESSION OF CNV
important factor in the development of CNV. In histologic While understanding the development of CNV continues to
studies, eyes with AMD had an increased prevalence of evolve, researchers have developed a theoretical construct that
lymphocytes, macrophages, and fibroblasts within Bruch’s correlates the growth pattern of CNV with the pathobiologic
membrane when compared with control eyes without AMD.13–15 dynamic stages of CNV progression.21 Although no two CNV
These findings would suggest that a low-grade chronic growth patterns are identical, two distinct patterns of CNV
inflammatory response may be involved in the development of have been defined. The first pattern involves growth into the
AMD, although inflammation has a role in the involutionary plane between the RPE and Bruch’s membrane (sub-RPE, or 1929
RETINA AND VITREOUS

type 1). The second pattern involves growth between the retina monocyte colonization protein (MCP) and interleukin-8 (IL-8).
and RPE (subretinal, or type 2). This description of CNV growth Monocytes (or macrophages) express tumor necrosis factor a,
patterns represents a spectrum, and it should be noted that which in turn stimulates IL-8, MCP, and VEGF production by
both patterns do not necessarily occur in temporal sequence, the RPE. VEGF signaling produces other responses, including
i.e., subretinal (type 2) may precede sub-RPE (type 1). Also, proliferation and migration of vascular endothelium.23
subretinal and sub-RPE CNV may be present in the same eye, Vasculogenesis may also play a role in CNV. Recent evidence
and there may be (and often are), several foci of sub-RPE CNV.21 supports a role for hematopoietic stem cells (HSC) in the
Patients with sub-RPE CNV may have few visual symptoms, as pathobiology of CNV, and these stem cells express the
is most often seen in some cases imaged on fluorescein functional VEGF-1 receptor.21 After this initiation stage, CNV
angiography with a lesion composition described as occult CNV progresses to an inflammatory active stage. At this point, CNV
or with no classic CNV (see section below describing lesion digests through other tissue planes, and macrophages express
composition on fluorescein angiography of CNV). CNV may be tissue factor, a protein involved in fibrogenesis, leading to a
difficult to detect angiographically due to the subtle patterns of fibrin scaffold on which CNV grows.21 Additionally, growth factors
fluorescence sometimes associated with occult CNV.21 This are expressed by the RPE and vascular endothelium, including
growth pattern is thought to provide nutrients and oxygen to PDGF, angiopoietins,23 aFGF, bFGF, and transforming growth
an ischemic RPE/outer retina that is expressing vascular factor beta.21 During this stage, CNV stabilizes, and
endothelial growth factor (VEGF) and other angiogenic equilibrium appears to be established between the various
cytokines.21 The subretinal growth pattern is characterized by growth and other factors that are released.21
SECTION 10

CNV occurring between the retina and RPE; tufts of CNV At some point, the balance shifts toward antiangiogenic,
break through Bruch’s membrane and extend laterally under antiproteolytic, and antimigratory activity, resulting in the
the RPE. The subretinal growth pattern of CNV typically has inflammatory inactive or involutional stage of CNV. The CNV
one or a few ingrowth sites.21 This pattern is characterized by may become collagenized and form a disciform scar.21 The
a break in Bruch’s membrane, with proliferation of granulation whole process may be orchestrated by the RPE, including the
tissue into the subretinal space, and vascular leakage under the initiation, stabilization, and involution of CNV, much like a
RPE/outer retina, leading to relatively acute visual symptoms.21 ‘traffic cop’.21
Regardless of the pathogenesis of CNV in AMD, clinico-
SUMMARY OF VEGF ROLE FROM pathologic correlative studies6–9,24 and natural history studies3,25–29
ANGIOGENESIS/GROWTH TO INHIBITORY have shown that untreated CNV is often accompanied by the
ingrowth of fibrous scar tissue, eventually resulting in a
(ANTIANGIOGENIC) MODE IN UNTREATED disciform scar. This CNV-scar complex may have a variety of
CNV complex clinical and angiographic appearances. An understanding
Over time, different factors have been implicated in the of these clinical features (described in the next section) is
dynamic process of CNV growth. This dynamic nature is critical in the identification, proper management, and
characterized by progression from an angiogenesis/growth treatment of the choroidal neovascular form of AMD.
promotion mode to an inhibitory (or antiangiogenic) mode, and
is incompletely understood.21 However, it is clear that growth CLINICAL FEATURES
factors are associated with progression of CNV.22 Recent
evidence suggests a central role of VEGF (also called VEGF-A),
in the development of CNV. VEGF-A is highly regulated by
SYMPTOMS OF CNV
hypoxia, an important benchmark condition necessary for CNV should be suspected in any patient (usually >65 years
vascularization in general. Experimental findings suggest that with large, soft drusen) who complains of metamorphopsia,
hypoxia is involved in a feedback mechanism to accommodate central or paracentral scotoma, or any sudden, nonspecific
insufficient tissue oxygenation by promoting vessel formation change in central vision.30,31 Any of these symptoms should
under normal conditions. It is hypothesized that derailment of alert the ophthalmologist to look for signs of CNV, which are
this feedback mechanism may lead to uncontrolled angiogenesis outlined further on. However, not all patients with CNV will be
in neovascular AMD.22 The vascular endothelial growth factor symptomatic32 or will note changes of metamorphopsia on
family includes placenta growth factor (PIGF) VEGF-A, VEGF- home-testing with an Amsler grid.33 Therefore, even
B, VEGF-C, VEGF-D, and the viral VEGF homolog VEGF-E).22 asymptomatic patients older than 65 years with large, soft
In the human, four isoforms of VEGF-A have been identified: drusen on clinical examination should probably be scrutinized
VEGF121, VEGF165, VEGF189, and VEGF206.22 The role of for signs of CNV on periodic examination.
these growth factors in AMD and tumor progression is an area
of active research. However, VEGF has been implicated in the
process of pathologic vessel growth in both experimental animal SIGNS OF CNV
models and in humans.23 VEGF appears to be causal for the In the early stages of the untreated neovascular form of AMD,
blood–retina barrier breakdown that accompanies neovascular- before disciform scarring has developed clinically, biomicroscopic
ization, as well as that which develops independent of it.23 clues to the presence of CNV may include any or all of the
Other important factors implicated in neovascularization following: the presence of subretinal or intraretinal lipids,
include platelet-derived growth factor (PDGF) and the elevation of the RPE, cystic changes in the sensory retina, or
angiopoietins.23 Evidence for the roles of these various growth visualization of the choroidal neovascular lesion itself. The
factors in neovascular AMD suggests a therapeutic use for novel lesion may be seen as a yellow-green discoloration frequently
drugs aimed at these targets. Pharmaceutical companies have surrounded by a pigmented ring and is often visualized best
already developed anti-VEGF treatments for the eye. Taken with transillumination of the RPE with a thin slit beam on
together, research findings suggest an important role for VEGF- biomicroscopy. The presence of subretinal or sub-RPE blood
A as a suitable candidate for targeted antiangiogenic therapy in may be so extensive as to obscure all other signs of CNV; often
AMD patients.22 though, this blood, if present, will be along the periphery of
In the early stages of CNV development, VEGF is produced the CNV (Fig. 146.1). Other causes of subretinal or sub-RPE
1930 by the RPE and retinal photoreceptors. The RPE also produces hemorrhage should be ruled out, including macroaneurysms,
Age-Related Macular Degeneration: Choroidal Neovascularization

precipitate in a circumferential pattern around the CNV and


thereby help to alert the ophthalmologist to the presence of
CNV. Other biomicroscopic signs of CNV, as already
mentioned, include the elevation of the RPE, presumably
caused by the presence of CNV and fibrovascular proliferation
beneath the RPE,24,35 pigment proliferation overlying the
CNV,36 or the actual choroidal neovascular vessels themselves
(occasionally seen when the overlying RPE pigmentation is
markedly attenuated).
If CNV secondary to AMD is suspected from symptoms or
signs, fluorescein angiography is indicated for the following
reasons: (1) to confirm the diagnosis of CNV, (2) to determine
whether treatment is indicated (as discussed further on), (3) if
treatment is indicated, to serve as a guide to treatment
response, and (4) rarely, if laser photocoagulation is indicated
for well-demarcated symptomatic extrafoveal lesions (see more
description below), to guide the placement of photocoagulation.

CHAPTER 146
FLUORESCEIN ANGIOGRAPHIC FEATURES
FIGURE 146.1. Subfoveal choroidal neovascularization (CNV) with OF CNV
contiguous blood (arrow). The boundaries of the entire lesion (the CNV A set of photographs to facilitate the identification of the variety
and blood) are well demarcated; the entire lesion is less than 3.5 disc of appearances of CNV secondary to AMD includes (1) a black-
areas. The lesion would meet the eligibility criteria of the Macular
and-white stereo pair of the macula obtained with green
Photocoagulation Study (MPS).
From Macular Photocoagulation Study Group: Laser photocoagulation of
(monochromatic) filter; (2) rapid-sequence photographs of the
subfoveal neovascular lesions in age-related macular degeneration. Results of a macula taken during the dye transit, including at least one
randomized clinical trial. Arch Ophthalmol 1991; 109:1220-1231. stereo pair; (3) stereo pairs of the macula taken at approximately
30, 40, 60, 90, 120, and 180 s after dye injection; (4) late stereo
pairs of the macula taken at 5 and 10 min after dye injection;
and (5) stereoscopic color fundus photographs of the macula.37
lacquer cracks in pathologic myopia, traumatic choroidal Since CNV growth can be a continuous process,38,39 the size of
rupture, choroidal tumors, or subretinal hemorrhage within the CNV and the lesion composition can change within a short
areas of geographic atrophy when no CNV is seen on time. Therefore, a fluorescein angiogram should ideally be
angiography. (Presumably, the subretinal hemorrhage within obtained within 2 weeks of initiation of treatment, unless laser
areas of geographic atrophy without CNV on angiography may photocoagulation is considered, in which case angiography
be due to CNV obscured by the hemorrhage or to disruption of should be performed ideally on the same day as any
the choriocapillaris in association with the geographic contemplated laser treatment and probably no more than 96 h
atrophy.34) before treatment.
Although CNV secondary to AMD often has been described Two basic angiographic patterns of CNV, recognized in the
in association with subretinal hemorrhage, many cases may macular photocoagulation studies37,40,41 as well as photo-
present with little or no hemorrhage, the predominant sign dynamic therapy trials,42 the submacular surgery trials, 43 trials
being the more subtle finding of subretinal fluid. Often, one evaluating anti-VEGF treatments44–46 and described by
needs a contact lens examination with biomicroscopy and a independent investigators,24,27,30,31,47–49 include classic and
thin slit beam to detect this subretinal fluid or evidence on occult CNV.
optical coherence tomography (OCT). On biomicroscopic
examination, the anterior portion of the beam will bow forward CLASSIC CNV
convexly, and there will be an increased distance between the
surface of the slit beam, which is visualized on the surface of This condition is characterized by an area of well-demarcated
the sensory retina, and the posterior portion of the beam, which hyperfluorescence that can be discerned in the early phase of
is visualized on the surface of the RPE. As subretinal fluid is the angiogram (Fig. 146.2a) with progressive dye leakage
absorbed at the periphery of the CNV, subretinal lipid may pooling in the overlying subsensory retinal space (Fig. 146.2b).

FIGURE 146.2. Classic CNV. (a) Early phase of


a fluorescein angiogram of classic CNV in
which the boundaries of the neovascular lesion
are well demarcated. (b) Late phase of the
angiogram shows pooling of dye in the
subsensory retinal space, obscuring the
boundaries of CNV demarcated in the earlier
phase of the angiogram.
(a and b) From Macular Photocoagulation Study
Group: Subfoveal neovascular lesions in age-related
macular degeneration. Guidelines for evaluation and
treatment in the Macular Photocoagulation Study. Arch
Ophthalmol 1991; 109:1242–1257. Copyright 1991,
American Medical Association.
a b
1931
RETINA AND VITREOUS

FIGURE 146.3. Recurrent classic and occult


CNV (with fibrovascular pigment epithelial
detachment (PED)). (a) Early phase of
fluorescein angiogram shows areas of classic
CNV (small solid arrow), scar from prior laser
treatment (large solid arrow), and irregular
elevation of the retinal pigment epithelium (RPE)
with stippled hyperfluorescence (open arrows)
representing fibrovascular PED inferotemporal
to the scar. (b) One minute after fluorescein
injection, fluorescein leakage is apparent from
the classic CNV, and increased intensity of
stippled hyperfluorescence corresponding to
fibrovascular PED is noted. The boundaries of
a b the fibrovascular PED remain well demarcated.
At each clock hour, the boundary of the lesion
is clearly demarcated and would meet eligibility
criteria in the MPS trials.
(a and b) From Macular Photocoagulation Study
Group: Subfoveal neovascular lesions in age-related
SECTION 10

macular degeneration. Guidelines for evaluation and


treatment in the Macular Photocoagulation Study. Arch
Ophthalmol 1991; 109:1242–1257.

The dye leakage identified by the late phase of the angiogram ANGIOGRAPHIC FEATURES THAT
will obscure the borders of the classic CNV detected in the OBSCURE THE BOUNDARIES OF CNV
early phase of the angiogram. Only occasionally will fluorescein
angiography identify the actual capillary network of the CNV Three features can obscure the boundaries of CNV and are
secondary to AMD. This latter observation is contrary to the important to recognize when attempting to delineate the
widely held view that CNV presents angiographically as a lacy boundaries of the choroidal neovascular lesion. They include (1)
network of vessels. This lacy pattern may be seen commonly in blood contiguous with the CNV that is thick enough to obscure
CNV secondary to other pathologic entities, such as the ocular the normal choroidal fluorescence, (2) elevated areas of blocked
histoplasmosis syndrome, but is unusual in AMD. fluorescence due to hyperplastic pigment or fibrous tissue, and
(3) a serous detachment of the RPE (Fig. 146.5). The first two of
OCCULT CNV these features block the angiographic view of choroidal
fluorescence. Lesions which are predominantly hemorrhage, or
This condition encompasses a variety of fluorescein angiographic predominantly serous PED (serous PED is an area of uniform
appearances that do not conform to the classic description of bright fluorescence in the early phase, with staining of this
CNV. The occult forms may be categorized into fibrovascular area in later phase frames) have not been subject to trials
pigment epithelial detachments (PEDs) and late leakage of evaluating treatments. However, often areas of occult CNV that
undetermined source. In fibrovascular PEDs (Fig. 146.3), areas are irregular elevations of the RPE may be confused with a
of irregular elevation of the RPE are often most easily detectable serous PED if one does not realize that a fibrovascular PED (a
on stereoscopic fluorescein angiography. These areas usually form of occult CNV) has a stippled, nonuniform fluorescence,
are not as discrete or bright as areas of classic CNV in pictures often not visible, and certainly not uniformly bright, in the early
taken during the transit; by 1–2 min after fluorescein injection, phase frames. Such areas of fibrovascular PED were considered
an area of stippled hyperfluorescence becomes apparent occult CNV, not an area of serous PED, and were subjected to
(Fig. 146.3b). By 10 min, there is persistence of fluorescein trials evaluating treatments. The bright, reasonably uniform early
staining or leakage within a sensory retinal detachment hyperfluorescence associated with a serous detachment of the
overlying this area. These fibrovascular PEDs should not be RPE (described further on) may obscure hyperfluorescence from
confused with serous PEDs (discussed further on). classic or occult CNV and interfere with the ability to judge
Late leakage of undetermined source consists of areas of late whether CNV extends under the area of the serous detachment.
choroidal fluorescein leakage, often appearing as speckled Thus, lesions which were predominantly serous PED,
hyperfluorescence, with pooling of dye in the overlying subsensory predominantly hemorrhage, or predominantly scar were excluded
retinal space, in which there is no discernible, discrete, well- from trials evaluating treatments for CNV and it may not be
demarcated area of hyperfluorescence that might be considered appropriate to extrapolate results of trials that excluded these
the source of leakage from earlier photography (Fig. 146.4). lesions in the management of them. On the other hand, one
Numerous clinicopathologic correlations on postmortem should include fibrovascular PEDs as occult CNV and make
examination8,9 or after submacular surgical removal of CNV50,51 treatment decisions as described for occult CNV below based on
have confirmed that all of these angiographic patterns (classic trials evaluating treatments that included occult CNV defined
CNV, fibrovascular PED, and late leakage of an undetermined in this way.
source) correlate with fibrovascular tissue. These studies have
not identified pathologic features to differentiate classic from OTHER CLINICAL AND ANGIOGRAPHIC
occult CNV. When adequate stereoscopic photographs are FEATURES OF CNV SECONDARY TO AMD
obtained, recognizing a pattern of fibrovascular PED rather
than late leakage of an undetermined source might depend on
the thickness9 of the fibrovascular tissue, with thicker tissue STAINING SCAR
corresponding to a pattern of fibrovascular PED on fluorescein Given variable amounts of time, as the lesion develops obvious
1932 angiography. scarring on examination or fundus photographs, the composition
Age-Related Macular Degeneration: Choroidal Neovascularization

FIGURE 146.4. Occult CNV with late leakage of


undetermined source. (a) Early phase of the
angiogram. (b) Middle phase of the angiogram
shows pinpoints of speckled hyperfluorescence
and larger areas of hyperfluorescence with
accumulation of fluorescein leakage in the
overlying subsensory retinal space. The source
of the leakage cannot be discerned from earlier
phases of the angiogram. The lesion does not
meet the MPS eligibility criteria that the
boundaries of neovascularization be well
demarcated. Therefore, treatment is not
considered for this lesion.
(a and b) From Macular Photocoagulation Study
a b Group: Subfoveal neovascular lesions in age-related
macular degeneration. Guidelines for evaluation and
treatment in the Macular Photocoagulation Study. Arch
Ophthalmol 1991; 109:1242–1257.

CHAPTER 146
late leakage, one cannot be sure that this pattern definitively
represents CNV.

FEEDER VESSELS
These vessels may be identified as choroidal vessels apparent
during the transit phase of the angiogram connected
unequivocally to leaking choroidal capillaries (Fig. 146.7).
Although feeder vessels have been described as extending from
a laser-treated area to recurrent CNV across the perimeter of the
laser-treated area,30,37,52 feeder vessels may also be seen in
untreated eyes. In the latter situation, peripheral untreated
areas of CNV may be connected by feeder vessels to more
central areas of CNV that are evolving toward natural scar
formation.37

LOCULATED FLUID
This fluid consists of a well-demarcated area of hyper-
fluorescence that appears to represent pooling of fluorescein in
a compartmentalized space anterior to the choroidal neovascular
leakage.53 Although the loculated fluid may conform to a
pattern of typical cystoid macular edema, it can also pool within
an area deep to the sensory retina in a shape that does not bear
FIGURE 146.5. Early-phase fluorescein angiogram of a serous any resemblance to cystoid macular edema53 (Fig. 146.8).
detachment of the RPE. A uniform elevation of the RPE, with uniform
pooling of fluorescein dye, and a smooth contour to the surface of the
elevated RPE, with well-demarcated borders in the early phase of the TEARS OR RIPS OF THE RPE
angiogram, are noted. The nasal aspect of the lesion shows irregular An acute tear or rip of the RPE may occur spontaneously
elevation of the RPE with mottled and speckled fluorescence (Fig. 146.9) or during laser photocoagulation of a choroidal
indicative of occult CNV.
neovascular lesion.54–59 Visual acuity may fall precipitously,
especially when associated CNV has an opportunity to destroy
foveal photoreceptors. When there is no CNV, RPE tears
through the fovea may be associated with preservation of
of the lesion may include staining of this scar rather than good central visual acuity provided that the torn area, and not
having the scar block fluorescence when a lot of pigment or the scrolled-up RPE, underlies the foveal center.60 Angiography
pigment epithelium is within the scar. When a majority of the demonstrates early, bright, sharply demarcated hyperfluorescence
lesion is composed of scar that stains on fluorescein within the torn region. Blocked fluorescence corresponding to
angiography, such lesion is described as being predominantly heaped-up RPE will be noted at one side of the lesion. The
scar and has not been subjected to trials evaluating treatments. bright hyperfluorescence presumably corresponds to fluorescein
dye within the choriocapillaris that quickly leaks into the
choroidal and scleral tissues and is not blocked by pigment that
FADING CNV is normally otherwise present within the overlying RPE. No
CNV occasionally may be recognized in the early- or middle- leakage of dye is seen if no overlying sensory retinal detachment
transit phase of the angiogram with fading in the late phase, so is present. The lack of a sensory retinal detachment over a tear
that no leakage can be discerned in the area that was presumed of the RPE may be due to the higher osmotic pressure of the
to harbor CNV37 (Fig. 146.6). These areas most likely represent choroid compared with that of the subretinal space, which
CNV histologically, but without evidence of subretinal fluid or allows fluid to be removed from the subretinal space at a rapid 1933
RETINA AND VITREOUS

a b

FIGURE 146.6. Fading fluorescence of CNV. (a) Early phase of the angiogram shows classic CNV (solid arrow) with contiguous areas of slightly
elevated hyperfluorescent RPE (open arrows), presumably representing a fibrovascular PED, and other less well demarcated areas of
hyperfluorescence nasal to fovea. (b) Later phase of the angiogram shows fluorescein leakage from classic CNV (arrow). However, areas of
SECTION 10

elevated hyperfluorescent RPE noted on the early phase of the angiogram begin to fade in the later phase. Faded areas are not considered a
lesion component to be treated in 1991 by MPS treatment protocol because hyperfluorescence does not have enough leakage or staining in the
late phase of the angiogram to be considered occult CNV. Before 1988, most ophthalmologists would have considered treatment of the classic
CNV with late leakage (arrow). By current interpretations, treatment of the area of classic CNV still might be contemplated, but ophthalmologists
would be concerned about the untreated areas of presumed occult CNV that fade in the late phase of the angiogram.
(a and b) From Macular Photocoagulation Study Group: Subfoveal neovascular lesions in age-related macular degeneration. Guidelines for evaluation and treatment in
the Macular Photocoagulation Study. Arch Ophthalmol 1991; 109:1242–1257.

rate when the tight junctions of the RPE are lacking and unable
to prevent free movement of fluid.61

DISCIFORM SCAR
The term disciform scar is used to describe the yellow-white
fibrous tissue that often accompanies CNV and is an extension
of the description of ‘scar’ above. The lesion may also contain
brown or black pigment, depending on the degree of pigment
proliferation from the RPE within the scar. The disciform
scarring may have a variety of appearances depending on its
location within the retina (sub-RPE or subretinal), the degree
of associated CNV with the scarring, the presence of
chorioretinal anastomosis within the scar, or the amount of
RPE atrophy accompanying the scar (Fig. 146.10). The natural
course of most choroidal neovascular lesions secondary to AMD
consists of scarring within the central portion of the lesion
with continued signs of active CNV at the periphery of the
lesion (including subretinal fluid, hemorrhage, or lipid).
Therefore, most disciform scars secondary to AMD could be FIGURE 146.7. Recurrent CNV with feeder vessel (arrow) as well as
termed CNV-scar when they include both a fibrous component larger choroidal vessels seen within the central portion of the scar
noted on biomicroscopy and a neovascular component from previous laser treatment.
represented by subretinal fluid-hemorrhage-lipid on biomicroscopy From Macular Photocoagulation Study Group: Subfoveal neovascular lesions in
and accompanied by leakage from CNV on angiography age-related macular degeneration. Guidelines for evaluation and treatment in the
(Fig. 146.10b). Occasionally, these disciform scars may develop Macular Photocoagulation Study. Arch Ophthalmol 1991; 109:1242–1257.
anterior to the posterior pole. These peripheral disciform scars
may become quite large with irregular shapes or they may be
accompanied by hemorrhage, leading to the suspicion of a hemorrhage. Sonographically, the posterior pole or peripheral
tumor such as a melanoma, until they are evaluated with lesion is relatively flat and broad-based, with a fairly homo-
ultrasound (see further on). geneous pattern and without signs of choroidal excavation. The
vitreous hemorrhage clears in approximately 75% of patients. If
the hemorrhage does not clear, a vitrectomy to restore
VITREOUS HEMORRHAGE peripheral vision should be considered, taking into account how
Occasionally, hemorrhage from CNV or CNV scarring extends restoration of that peripheral vision will improve the patient’s
into the vitreous space.62,63 Any patient with a massive quality of life, given the unlikelihood of restoration of central
vitreous hemorrhage in one eye and features of AMD in the vision. Whether or not submacular surgery should be considered
fellow eye should be suspected of harboring CNV in the eye to remove any subretinal hemorrhage or fibrovascular tissue
with vitreous hemorrhage. Ultrasonography should be performed identified at the time of vitrectomy probably should await
to rule out a rhegmatogenous retinal tear or detachment, results of clinical trials evaluating the benefits and risks of this
1934 choroidal melanoma, and other less common causes of vitreous intervention.
Age-Related Macular Degeneration: Choroidal Neovascularization

FIGURE 146.8. Example of an eye in which the


borders of loculated fluid extend beyond the
borders of CNV. In late-transit frames (a), the
area of loculated fluid (arrow) extends beyond
the area of the borders of the CNV (arrow in b)
as defined in the early-transit phase of the
angiogram.
(a and b) From Bressler NM, Bressler SB, Alexander J,
et al: Loculated fluid: A previously undescribed
fluorescein angiographic finding in choroidal
neovascularization associated with macular
degeneration. Arch Ophthalmol 1991; 109:211–215.

a b

blocked fluorescence is noted within the area of elevated RPE


throughout the angiogram. One of the more difficult
differentiations is between fibrovascular PEDs and serous

CHAPTER 146
detachment of the RPE. Although, they probably have been
lumped together in several series that have examined RPE
detachments,65–70 some of these clinical reports have attempted
to identify certain features that might distinguish between
serous detachments of the RPE and areas of elevation of the
RPE that may harbor occult CNV.67 Using descriptions from
the Macular Photocoagulation Study (MPS), fibrovascular PEDs
(as a subset of occult CNV) have been distinguished from
classic serous detachments of the RPE in that the former have
slow filling with a stippled appearance to the surface of the RPE
by the middle phase of the angiogram and may show pooling of
dye in the overlying subsensory retinal space in the late phase,
whereas the latter show uniform, bright hyperfluorescence in
the early phase with a smooth contour to the RPE by the middle
phase and little, if any, leakage at the borders of the PED by the
late phase. As mentioned earlier, the fluorescent pattern of a
serous PED obscures the ability to determine whether classic or
FIGURE 146.9. RPE tear seen on the early-transit phase of the occult CNV exists within the area of the serous PED.
fluorescein angiogram demonstrates extremely sharp, well- A hemorrhagic detachment of the RPE will block choroidal
demarcated hyperfluorescence. Continued intense staining was seen fluorescence just as blocked fluorescence from hyperplastic
in the late phase of the angiogram with no leakage. An early blocked
pigment or fibrous tissue does. However, in hemorrhagic
fluorescence (arrow) presumably corresponds to the redundant,
folded, torn RPE.
detachments of the RPE, the dark appearance on biomicroscopy
From Bressler NM, Finkelstein D, Sunness JS, et al: Retinal pigment epithelial caused by the mound-like collection of blood beneath the RPE
tears through the fovea with preservation of good visual acuity. Arch Ophthalmol will help to differentiate it from areas of elevated blocked
1990; 108:1694–1697. fluorescence caused by hyperplastic pigment or fibrous tissue as
discussed previously. Occasionally, a hemorrhagic detachment
of the RPE may be mistaken for a choroidal melanoma, but
usually hemorrhagic detachments of the RPE will not
RPE DETACHMENTS IN AMD demonstrate low internal reflectivity, as is seen characteristically
in choroidal melanomas.
Various changes in AMD may result in elevation or detachment The final feature of AMD that will appear as an elevated or
of the RPE as seen on stereoscopic biomicroscopic or detached RPE is a drusenoid RPE detachment or extensive areas
angiographic evaluation. The term RPE detachment secondary of large confluent drusen.64 Drusenoid RPE detachments can be
to AMD in the ophthalmic literature remains confusing distinguished from serous detachments of the RPE in that
because various RPE detachments may have quite different drusenoid RPE detachments will fluoresce faintly during the
prognoses and managements, and yet several series may have transit and do not progress to bright hyperfluorescence in the
included some or all of these RPE detachments in their reports. late phase of the angiogram. In contrast, serous detachments
RPE detachments secondary to AMD that may be readily of the RPE will fluoresce brightly in the early-transit phase and
recognized and probably should be differentiated include (1) remain brightly hyperfluorescent in the late phase. In addition,
fibrovascular PEDs, which are a subset of occult CNV serous detachments usually will have a smoother, sharper
(Fig. 146.3); (2) elevated areas of RPE that block fluorescence boundary compared with drusenoid RPE detachments. Drusenoid
because of hyperplastic pigment or fibrous tissue (Fig. 146.3); RPE detachments can be distinguished from fibrovascular PEDs
(3) serous detachments of the RPE (Fig. 146.5); (4) hemorrhagic in occult CNV by noting that fibrovascular PEDs will show
detachments of the RPE, in which blood from a choroidal areas of stippled hyperfluorescence with persistence of staining
neovascular lesion is noted beneath or exterior to the RPE; and or leakage within a sensory retinal detachment overlying the
(5) drusenoid RPE detachments, in which large areas of area in the late phase of the angiogram. RPE detachments
confluent, soft drusen are noted.64 associated with large, soft, confluent drusen are usually smaller,
Elevated blocked fluorescence may be differentiated from more shallow, and more irregular in outline than are
fibrovascular PEDs and serous detachments of the RPE in that fibrovascular PEDs. In addition, the drusenoid RPE detachments 1935
RETINA AND VITREOUS

FIGURE 146.10. CNV-disciform scarring.


(a) Subretinal and sub-RPE fibrosis, as well as
subretinal fluid and hemorrhage, is seen on the
color photograph. The latter presumably are
indicative of persistent vascular tissue within
the fibrosis. (b) Fluorescein angiography of CNV
scarring demonstrates leakage toward the
periphery of the lesion, presumably from CNV
associated with the scarring.
(a and b) From Macular Photocoagulation Study
Group: Subfoveal neovascular lesions in age-related
macular degeneration. Guidelines for evaluation and
treatment in the Macular Photocoagulation Study. Arch
Ophthalmol 1991; 109:1242–1257.
a b

will often have reticulated pigment clumping overlying the


large, soft, confluent drusen, and a scalloped border. TABLE 146.1. Time Trade-off Values.4 Values Represent the
SECTION 10

Number of Years (of Every 10 Years of Remaining Life


Expectancy) That Participants Would Trade for Perfect Vision.
IMPACT OF NEOVASCULAR AMD ON QUALITY Best Corrected VA Ophthalmologists AMD
OF LIFE (n = 46) Patients (n = 72 )
Health-related quality of life (QOL), also known as patient- 20/30 to 20/50 0.3 1.9
centered QOL, refers to those aspects of life that either foster
or compromise the ability to participate fully in daily life, such 20/60 to 20/100 1.1 4.3
as driving, recognizing faces, reading small print, judging the 20/200 to 20/400 2.3 4.9
depth of stairs or curbs, or watching TV. Because patient-
Counting fingers to 3.3 6.0
centered QOL is subjective,71 AMD patients with similar visual hand motions
acuity (VA) may rate their QOL quite differently, depending
on which activities and skills are valued most.72 To
systematically measure health-related QOL, researchers have
developed standardized questionnaires that can be self-
administered or conducted as an interview. TABLE 146.2. Time Trade-off Utility Values Stratified by Level
of Vision.6

VA (n) Utility Value P value‡ P value§


WHY IS IT IMPORTANT TO EVALUATE QOL IN
AMD PATIENTS? 20/200 to 0.65 0.6 µ 10 —12
0.03
20/400* (33)
QOL instruments are widely used to assess patients with other
chronic diseases that are common in the population at risk LP to counting 0.47 0.007 NA
for AMD, such as arthritis, depression, cancer, hypertension, fingers* (17)
and stroke. QOL measures are often added to clinical trials to NLP† (15) 0.26 NA 0.007
evaluate the cost-effectiveness and therapeutic value of new
LP = light perception; NLP = no light perception.
treatments.71 Researchers and clinicians recognize the * In the better eye.
psychosocial determinants of health, such as the skyrocketing † NLP in one eye; postulated NLP in other eye.
cost of healthcare, and patients’ desire to participate more ‡ P value from Student’s t test when compared with NLP group.
§ P value from Student’s t test when compared with LP to counting fingers
actively in treatment decisions.73 This has led to active group.
soliciting of patient input about QOL. Patient-reported visual
function can reinforce treatment decisions for practitioners,
providing evidence of treatment benefits.
Ophthalmologists may underestimate the impact that AMD vision with CNV was 0.64.75 Preference values were higher in
can have on patients’ health-related QOL. In one study,54 younger patients and those with better VA, and lower in
ophthalmologists were asked to imagine that they had AMD in patients with anxiety or depression.75 These mid-range values
both eyes and were given the hypothetical scenario: regain imply that some CNV patients may consider an expensive or
perfect vision by giving up some remaining lifespan.74 The same potentially risky treatment to improve their QOL.75 In a utility
hypothetical scenario was offered to 72 AMD patients. For study, Brown and colleagues used a time trade-off method,
every 10 years of life remaining, AMD patients with VA of dividing 65 consecutive ambulatory patients into three groups.76
20/30 to counting fingers would give up more years than would In the time trade-off technique, the investigator calculates each
ophthalmologists with the same hypothetical VA (Table 146.1). participant’s life expectancy and then asks how many years of
Another way to measure patient-centered QOL is with remaining life expectancy an individual would be willing to
preference values. In the submacular surgery trials, 996 trade in return for perfect vision77 (Fig. 146.11).
participants with subfoveal CNV were asked to pretend that One group, with no light perception (NLP) in at least one eye,
they were completely blind at their current level of general was asked to imagine that they had NLP in both eyes. One
health. Participants were also asked to pretend that they had group had bilateral vision with light perception to counting
perfect vision and their current level of general health.75 They fingers, and another group had 20/200 to 20/400 vision. Mean
rated these hypothetical situations on a scale of 0 (death) to 100 utility values were lowest in the group with the worst vision.
(perfect health with perfect vision). Among the 792 evaluable (Table 146.2) 6 Patients in the NLP group were willing to trade
1936 participants, the mean preference value assigned to current almost three out of every 4 years of remaining life expectancy in
Age-Related Macular Degeneration: Choroidal Neovascularization

FIGURE 146.11. Time trade-off calculation.


Input Variables

Participantís age 65 years


Participant’s anticipated life expectancy 85 years
How many remaining years of life would 10 years
participant trade in exchange for living without
AMD

Calculation of Time Trade-off

How many more Divide years participant Subtract previous value


years does would trade for perfect from 1.0
participant expect to vision by remaining
live? years of expected life 1.0 – 0.50 = 0.50

85 years 10 years = 0.50


20 years

CHAPTER 146
-65years
20 years
Calculation of Utility Score

Subtract the time trade-off from 1.0 (the utility value for perfect health).
1.0 – 0.50 = 0.50

return for perfect vision in each eye.76 Even patients in the Activities of Daily Vision Scale (ADVS), a 21-item questionnaire
20/200 to 20/400 group would trade one-third of their about vision-specific activities, including driving in darkness
remaining life expectancy for perfect vision.76 or daylight, and nondriving activities involving distance or near
vision, or glare.81 The loss of driving ability has widespread
implications: 1) a decreased ability to care for oneself (eg.,
THE IMPACT OF AMD ON ACTIVITIES OF earning a living, grocery shopping, getting to doctor appointments)
DAILY LIVING (ADLS) and 2) a decreased ability to care for others, such as a
Visual changes affect physical function, even in patients with chronically ill, elderly spouse.82 Data on contrast sensitivity loss
unilateral advanced AMD. The submacular surgery trials for patients with macular disease suggest that patients with
interviewed 789 patients with subfoveal CNV to evaluate QOL. moderate visual loss (20/70 to 20/200) may have difficulty
Investigators used the National Eye Institute Visual Function recognizing faces.83 Patients with visual disabilities in general
Questionnaire (NEI-VFQ). As noted earlier, the NEI-VFQ is a are at risk of becoming socially isolated, due to difficulties with
validated questionnaire that assesses physical aspects of health moving around and performing ADLs. The inability to recognize
(general health, visual health, pain, and near and distance vision a friend or loved one would also increase feelings of social
activities), and how visual disability affects mental health, role isolation and impact functioning.84
difficulties, social functioning and dependency, driving, and
color and peripheral vision. Scores range from 0 (worst
functioning) to 100 (best functioning).78 Patients with bilateral DEPRESSION AND ANXIETY
AMD scored 6–10 points lower on the NEI-VFQ than patients The fear of progression and potential blindness among AMD
with unilateral AMD even after adjusting for VA.79 Preventing patients remains an important factor in patient-centered
and or delaying the progression of AMD is crucial QOL.77,85 A subgroup of patients with newly diagnosed
to reducing the risk of rapidly declining QOL for patients with subfoveal CNV who participated in the Submacular Surgery
vision loss.79 Cahill and colleagues evaluated patient-centered Trials had measurable anxiety and depression at baseline. In
QOL among 70 patients with bilateral severe AMD using the particular, among the participants who had no prior laser
NEI-VFQ. The mean QOL scores for these important quality treatment in the affected eye, 60/447 (13%) were classified as
of vision (general vision, difficulty with distance and near tasks), having either doubtful or definite anxiety, and 57/447 (12%) as
and vision-specific subscales (dependency, role difficulties, mental having doubtful or definite depression, using the Hospital
health, social function limitations) were significantly worse for Anxiety and Depression Scale.79 In one study, patients with
patients with bilateral severe AMD than for patients with AMD severe bilateral AMD scored significantly lower in the social
of varying severity, and persons without eye disease.80 These and mental health domains of the NEI VFQ-25 than patients
data further support the assertion that visual changes affect with AMD of varying severity and persons without ocular
physical function even in patients with relatively mild AMD. disease.80

THE SOCIAL AND PSYCHOLOGICAL IMPACT MANAGEMENT OF CHOROIDAL


OF NEOVASCULAR AMD ON QOL NEOVASCULARIZATION IN AMD
Not surprisingly, many patients with bilateral advanced AMD, When neovascular AMD is suspected, a fluorescein angiogram
usually neovascular, are hesitant and sometimes unable to is obtained to determine the location of the lesion, and its
drive.80 Among a group of patients with AMD of mixed severity, composition. Following the algorithm outlined in Fig. 146.12
there was a direct correlation between higher severity of AMD after one determines the entire extent of the lesion, one can
and poorer scores on the driving activities subscale of the determine if the most posterior extent of the lesion is 1937
SECTION 10 RETINA AND VITREOUS

FIGURE 146.13. Proportion of eyes at each follow-up examination


FIGURE 146.12. Algorithm for management of CNV in AMD. with a decrease in visual acuity of 6 or more lines from baseline in the
Senile Macular Degeneration Study of the MPS. Dashed line indicates
eyes assigned randomly at entry to no treatment; solid line, eyes
extrafoveal (at least 200 mm from the geometric center of the
assigned at entry to laser treatment. All eyes at baseline (time zero)
foveal avascular zone). While not very common in AMD, in had well-demarcated extrafoveal CNV.
such cases, if the lesion has well-demarcated boundaries, and From Macular Photocoagulation Study Group: Argon laser photocoagulation for
the patient is symptomatic, then laser photocoagulation is neovascular maculopathy after five years. Results from randomized clinical trials.
indicated. The immediate goal of laser treatment in all clinical Arch Ophthalmol 1991; 109:1109–1114.
trials evaluating laser use for CNV38,39,86–91 was to
photocoagulate the entire area of CNV. In order to treat the
entire area of CNV, the ophthalmologist has to be able to
identify the boundaries of the choroidal neovascular lesion.
Therefore, at the present time, treatment is indicated only
when the boundaries of the CNV are well demarcated. If the
boundaries are not well-demarcated, or if one is not certain if
the lesion extends under the foveal center, or if the lesion is
so close to the foveal center that the treating ophthalmologist
believes the scotoma from treatment will outweigh any benefits
of treatment, then one could manage the lesion as if its location
were subfoveal as described below.

RISKS AND BENEFITS OF TREATMENT OF


EXTRAFOVEAL CNV (POSTERIOR BOUNDARY
OF CNV BETWEEN 200 AND 2500 µM FROM
GEOMETRIC CENTER OF THE FAZ)
In the MPS, laser treatment was beneficial at decreasing the risk
of severe visual loss in eyes with extrafoveal CNV secondary
to AMD when compared with no treatment.88 The proportion
of eyes with severe visual loss (6 lines or more of vision loss) FIGURE 146.14. Cumulative proportion of laser-treated eyes ever
1 year after presenting with extrafoveal CNV was 41% in the having recurrent CNV documented after initial laser treatment. Dashed
eyes assigned to no treatment and 24% in the eyes assigned to line indicates eyes assigned to laser treatment in the Senile Macular
laser treatment. By 3 years, 63% of the eyes assigned to no Degeneration Study in which patients had extrafoveal CNV secondary
to AMD. For comparison to a cumulative proportion of recurrences
treatment and 45% of the eyes assigned to treatment had severe
after treatment of CNV secondary to other causes, solid line indicates
visual loss. This treatment benefit was maintained by 5 years eyes assigned to laser treatment in the Ocular Histoplasmosis Study,
after treatment, at which time 64% of the eyes assigned to no and dotted line, eyes in the Idiopathic Neovascularization Study.
treatment and 46% of the eyes assigned to treatment had severe From Macular Photocoagulation Study Group: Argon laser photocoagulation for
visual loss92 (Fig. 146.13). The relative risk of losing 6 lines or neovascular maculopathy after five years. Results from randomized clinical trials.
more of visual acuity from baseline among untreated eyes (n = Arch Ophthalmol 1991; 109:1109–1114.
117) compared with laser-treated eyes (n = 119) was 1.5 from
6 months through 5 years after entry into the study (P =0.001).
Furthermore, after 5 years, untreated eyes had lost a mean of of all recurrences occurred between the end of the second year
7.1 lines of visual acuity, whereas laser-treated eyes had lost 5.2 and the fifth year of follow-up (Fig. 146.14). The effect of
lines. recurrence on visual acuity can be seen in Table 146.3; by
Recurrent CNV was observed in 54% of laser-treated eyes by 3 years, only 10% of the treated eyes with no recurrence had
the end of the 5 year follow-up period92 (Fig. 146.14). About severe visual loss compared with 80% of the treated eyes with
75% of all these recurrences occurred by the end of the first recurrence. At the end of the third year of follow-up, the average
year after treatment. An additional 17% of all the recurrences visual acuity of the treated eyes with no recurrence was 20/50
1938 occurred between 1 and 2 years of follow-up. The remaining 7% and that of the treated eyes with recurrence was 20/250.92 This
Age-Related Macular Degeneration: Choroidal Neovascularization

The term persistence was used by the MPS group to indicate


TABLE 146.3. Visual Acuity by History of Recurrence after
the presence of fluorescein leakage on the periphery of the
Laser Treatment of Extrafoveal Choroidal Neovascularization
Secondary to AMD foveal side of the laser-treated area within 6 weeks after
treatment. The investigators believed that fluorescein leakage
Years since Recurrence Number Average Eyes With within this time might represent persistence of neovascularization.
Treatment of Eyes Visual 6-Line The MPS investigators chose to use the term recurrence when
Acuity Loss n (%)
angiography confirmed no leakage for at least 6 weeks after
1 Yes 62 20/40 4 (6) treatment, with leakage subsequently noted sometime later
than 6 weeks after treatment.93 These terms were strictly
No 49 20/125 20 (41)
defined for analysis of the data from these trials. However, one
2 Yes 49 20/40 4 (8) could consider using the term recurrence whenever the
No 54 20/160 31 (57) following conditions apply: (1) leakage is seen at the periphery
of a laser-treated area and (2) one has previously documented
3 Yes 48 20/50 5 (10) unequivocal lack of peripheral leakage after treatment. A
No 46 20/250 37 (80) persistence could be defined as leakage at the periphery of the
laser-treated area without any prior unequivocal documentation
4 Yes 43 20/50 5 (12)
of lack of peripheral leakage on prior fluorescein angiograms.
No 47 20/250 37 (79) When treatments with photodynamic therapy or anti-VEGF

CHAPTER 146
5 Yes 42 20/50 7 (17) drugs are applied, then other terms may be used to describe
angiographic appearances after treatment43 Specifically,
No 50 20/250 39 (78) progression used for leakage from CNV beyond the boundaries
of the lesion identified prior to treatment. Absence of leakage
implies that there is no progression and no leakage within the
area of CNV identified prior to treatment. Minimal leakage
TABLE 146.4. Treatment Protocol for Choroidal means that there is no progression and that less than 50%
Neovascularization from the Macular Photocoagulation Study of the area of leakage from CNV identified prior to treatment
still shows fluorescein leakage from CNV. Finally, moderate
All Lesions
leakage means that there is no progression and that 51-to 99%
Angiogram <96 hr old of the area of leakage from CNV identified prior to treatment
Retrobulbar anesthesia as necessary still shows fluorescein leakage from CNV.
200–500 mm spot; 0.2–0.5 s duration
Cover entire area of neovascular lesion
Intensity sufficient to produce uniform white burn APPLICATION OF LASER
Extrafoveal Lesions (>199 mm from Foveal Center) PHOTOCOAGULATION
Extend treatment additional 100 mm beyond any adjacent blood, Preparation
pigment ring circumferentially surrounding the lesion, or other An angiogram is projected onto a screen or monitor near the
blocked fluorescence surrounding the lesion laser, so that the ophthalmologist can make rapid and repeated
Juxtafoveal Lesions (1–199 mm from Foveal Center) extrapolations from the fluorescein’s retinal vascular landmarks
to the patient’s fundus when identifying the location of the
Extend treatment additional 100 mm beyond the neovascular lesion
CNV with respect to these landmarks. Careful evaluation of
on border away from fovea
Extend treatment additional 100 mm into any blood present on the vascular landmarks in the patient’s fundus in comparison with
foveal side if the hyperfluorescence from the neovascular lesion landmarks concurrently viewed on a projection of a fluorescein
itself is 100 mm or farther from foveal center angiogram during treatment should enable the ophthalmologist
to identify the boundaries of the lesion with confidence and
Subfoveal Lesions (CNV Underlies Foveal Center)
accuracy and avoid inadvertent treatment of retina that is not
Extend treatment additional 100 mm beyond peripheral boundaries involved with the lesion.
of all lesion components except blood Topical anesthesia usually is sufficient if one is careful to
Cover, but not necessarily extend 100 mm beyond areas of blocked
fluorescence into thick blood
ensure that neither ocular motility nor patient discomfort will
compromise the success of treatment by preventing the
Subfoveal Recurrent Lesions (Prior Laser Treatment with ophthalmologist from delivering laser of sufficient intensity
Recurrent Lesion Underlying Foveal Center) and duration of exposure to produce a uniform white treatment
Extend treatment 300 mm into previous treatment scar-recurrent burn. This is especially true when small amounts of
neovascular lesion interface undertreatment may allow persistence of neovascularization93,94
If feeder vessels present, extend treatment 100 mm beyond lateral or small amounts of overtreatment might obliterate foveal
borders of recurrent vessels and 300 mm radially beyond base structures unnecessarily.95
(origin) of feeder vessel
Treatment Parameters
Initial laser burns are placed along the boundary of the CNV
treatment benefit has been confirmed by two independent trials using a 200 µm spot size and 0.2–0.5 s duration. These
comparing treatment and the natural course.86,87 Although, the parameters allow the ophthalmologist to get sufficient heat to
treatment protocol did not permit treatment of recurrences the retina (with the long duration) without risking a sudden
through the foveal center in this trial, the treatment benefit for break in Bruch’s membrane and to minimize the frequency of
these lesions might have been even greater if anti-VEGF bleeding (by spreading the intensity of the burn over a 200-µm
therapies now in use (see below) for subfoveal lesions had been spot rather than a 50 or 100 µm spot). The area to be covered,
applied to the subfoveal recurrences that would have met if one is following the protocol used in the MPS, differs
current criteria for consideration of treatment of subfoveal depending on the location of the lesion and is outlined in Table
lesions. 146.4 . After the boundaries of the lesion have been treated, the 1939
RETINA AND VITREOUS

studies.40,41 Recurrence and persistence rates also were similar


between the two laser wavelengths. The recurrence and
persistence rates were similar to those observed when the
krypton red laser alone was used to treat neovascular lesions in
the MPS trial of juxtafoveal neovascularization. Thus, no visible
wavelength appears to have a significant advantage over other
wavelengths. Small differences in convenience of achieving
the endpoint of a uniform white burn might be seen with red
or yellow wavelengths when penetrating through the increased
nuclear yellow in the older age group afflicted with CNV
secondary to AMD, but any significant differences of clinical
importance have not been shown.

Special Circumstances
When treating CNV that lies under a major retinal vessel, the
laser burns should straddle the retinal vessel to reduce the
possibility of causing hemorrhage or damaging the vessel by
thermal vasculitis. There is no evidence to suggest that this
SECTION 10

technique compromises the effectiveness of treatment.


When treating CNV that is contiguous with the optic nerve,
one must consider that laser treatment directly over the optic
FIGURE 146.15. Treatment intensity standard. The treatment protocol nerve can cause thermal necrosis of disc tissue and nerve fiber
of the MPS specified a uniform, white burn at least as intense as the bundle defects.98 Therefore, one should consider refraining from
treatment standard. treatment within 100–200 µm of the optic nerve. Similarly,
From Macular Photocoagulation Study Group: Persistent and recurrent when treating a parapapillary area of CNV, one may want to
neovascularization after krypton laser photocoagulation for neovascular lesions
consider treatment only when at least 1 1/2 clock hours of
of ocular histoplasmosis. Arch Ophthalmol 1989; 107:344–352.
papillomacular bundle on the temporal side of the disc is
uninvolved with CNV, so that at least 1 1/2 clock hours of
papillomacular bundle can be spared treatment, as was done in
area within the boundaries is treated subsequently with burns several of the MPS trials.88,99 Treatment to nasal or
of the same spot size or larger, using a duration of 0.5–1 s. The parapapillary lesions likely will not lead to severe visual loss
desired endpoint for the intensity of the laser lesion is to create from damage to the nerve fiber layer that serves the central
a uniformly relatively white lesion. The ophthalmologist can macula if the treatment guidelines outlined previously are
achieve this end-point in either of the following ways: (1) by employed.100 In these situations, the MPS group reported that
initially applying relatively white laser burns that meet or severe visual acuity loss was noted after treatment only when
exceed the intensity illustrated by a standard photograph from recurrent CNV extended through the center of the fovea.100
the MPS89 or (2) by applying lighter gray-white laser spots that This finding suggests that severe visual loss only from nerve
overlap again and again until the entire laser lesion is a uniform fiber layer damage after this treatment approach, in the absence
white treatment burn at least as white as the treatment of subfoveal recurrence, must be a rare complication.
intensity standard37 (Fig. 146.15). Certain subgroups in the various trials had different
treatment benefits, which should be considered when
Wavelength Selection determining whether treatment would be beneficial for a
When the MPS was begun in 1978, the argon blue-green laser particular patient.89,101 This subgroup analysis should probably
was the one most commercially available to investigators. This not completely sway someone in recommending or denying
laser is no longer recommended for treatment within the treatment. Rather, the subgroup analysis data should serve as a
macular region because macular xanthophyll pigment directly guideline when trying to decide whether treatment should be
absorbs the blue light of the argon blue-green laser, thereby recommended to a particular individual. For instance, in the
inducing thermal damage to the inner retina.96 In addition, the krypton trial of juxtafoveal lesions,89 patients who were
risk of inducing internal limiting membrane wrinkling, normotensive had a marked treatment benefit. Patients who
although rarely of clinical significance, is probably greatest with had evidence of hypertension, either by elevated systolic or
the argon blue-green laser.97 Subsequent trials in the MPS for diastolic blood pressures or by the use of antihypertensive
juxtafoveal lesions employed the krypton red laser because of its medications, had no treatment benefit. Although similar trends
theoretical advantage of penetrating through xanthophyll and were noted in the argon trial for patients with CNV secondary
passing through thin layers of red hemorrhage, allowing the to the ocular histoplasmosis syndrome,99 similar trends were
uptake of the laser to be concentrated within the RPE and not noted in the argon trial of CNV secondary to AMD102 nor
melanocytes of the inner choroid. When the MPS trials for in the subfoveal trials in AMD.40,41 Therefore, although the
subfoveal lesions were designed, eyes randomized to the data in the juxtafoveal trial failed to detect a treatment benefit
treatment group were further randomized to either the argon for hypertensive patients, lack of corroboration of this finding
green or the krypton red wavelength for treatment. None of the in two other prospective trials on CNV secondary to AMD
findings from the subfoveal trials suggests a reason to favor cautions one from withholding treatment from patients who are
either the argon green or the krypton red wavelength.40,41 If hypertensive.
there were any theoretical advantage to using one wavelength There is only weak evidence that laser photocoagulation of
over another for treatment of CNV, one might have expected to extrafoveal fluorescent spots (‘hot spots’) on ICG angiography
have detected a difference within the MPS subfoveal trials. or feeder vessels on high speed video ICG angiography within a
Although these trials did not have sufficient power to neovascular lesion leads to a better outcome than no treatment.
demonstrate a small or moderate difference between the two The lack of controls in these trials, or comparisons to anti-
1940 wavelengths, a large difference has been ruled out by these VEGF treatments now used for subfoveal lesions, precludes one
Age-Related Macular Degeneration: Choroidal Neovascularization

a b

CHAPTER 146
FIGURE 146.16. Evaluation of CNV treatment. (a) Angiogram is projected onto
an apparatus (such as a microfilm reader or slide viewer) such that the CNV and
key landmarks around the CNV such as subretinal blood, retinal vessels, and
foveal center can be drawn. (b) The foveal avascular zone (FAZ) and the foveal
center (x) are indicated. (c) A posttreatment photograph is then projected, and
the area of heavy treatment and the same landmark vessels can be outlined on
a separate piece of paper. Evaluation of photocoagulation treatment can be
determined by placing the treatment drawing (c) under the pretreatment drawing
(a) on a lightbox. The area of heavy treatment can then be traced onto the
pretreatment drawing to determine whether the treatment has entirely covered
the CNV.
(a and b) From Bressler NM, Bressler SB, Fine SL: Age-related macular degeneration. Surv
Ophthalmol 1988; 32:375–413.) Alternatively, digital imaging software can be used to overlay
the boundaries of treatment from a post-treatment photograph onto the boundaries of a
lesion from a pre-treatment frame from the fluorescein angiogram.
c

from concluding with much confidence that such treatments When a CNV lesion is extrafoveal, it usually is not difficult
should be considered. This conclusion also applies to such for an ophthalmologist who is experienced in treating CNV to
fluorescent spots seen within serous RPE detachments.103 extend laser treatment over the entire extent of the CNV.88,99
On rare occasions, a patient will present with extrafoveal There is probably little hesitancy in extending treatment
CNV contiguous to a serous detachment of the RPE in which slightly beyond the borders of the CNV in an effort to ensure
the serous detachment extends through the foveal center. There adequate coverage when the CNV is far away from the foveal
have been case reports in which only the extrafoveal CNV in center. Slight extension of treatment should have little effect on
these lesions is treated, resulting in prompt flattening of the the visual acuity, since the treatment in these situations will
RPE detachment with improvement of vision in selected not affect central foveal photoreceptors.
cases.104 Nevertheless, with follow-up, many of these eyes have In an attempt to minimize persistent CNV caused by
acquired recurrent CNV with extensive scarring and visual loss. inadequate coverage, one may evaluate the laser treatment by
The more likely situation is that the extrafoveal CNV actually comparing the area of laser treatment from a posttreatment
is associated with fibrovascular PED that extends through the photograph to the area of the lesion to be covered from the
foveal center in which treatment of the extrafoveal CNV alone pretreatment angiogram.30,105,106 The MPS group has described
has not been shown to be of any benefit,91 and anti-VEGF their methods for this evaluation.106 In step 1, the extent and
treatment, when indicated as described below, should be location of the CNV with respect to the vascular landmarks
considered. (Fig. 146.16a) is traced, either on paper of a projected slide or
on available imaging software with digital imaging systems. In
Postoperative management of laser photocoagulation step 2, immediately after treatment, a posttreatment
Data from the MPS group have shown that eyes in which laser photograph (digitally) or Polaroid (film) is taken. The area of
treatment did not cover the CNV completely on the foveal side heavy treatment and the same landmark vessels are outlined
or did not meet the required level of intensity (a uniform on a separate piece of plain white paper (Fig. 146.16b) or with
relatively white burn, as shown in Fig. 146.15) had ~3 times the image software. In step 3, the treatment drawing from step
the risk of having persistent CNV within 6 weeks after 2 is placed on a light box, and the pretreatment drawing from
treatment compared with eyes in which the CNV was covered step 1 is placed over this, superimposing the landmark vessels,
completely by intense, confluent burns.94 Because of this or superimposed by digital image software. The area of heavy
information, it is essential not only to obtain a uniform treatment can then be traced onto the pretreatment drawing
relatively white laser burn during treatment but also to ensure to determine whether the treatment has covered the CNV in its
that the extent of the intense confluent burns completely entirety (Fig. 146.16c). Any areas not adequately treated can be
covers the extent of the CNV. ‘touched up’ while the patient is still in the office. The MPS 1941
RETINA AND VITREOUS

group proved the usefulness of this method using 1-day


posttreatment stereoscopic color photographs; areas of NEI-VFQ Subscale Scores
inadequate treatment were identified, and these eyes did indeed
12
have a higher rate of persistent CNV in the juxtafoveal 9.9 Ranibizumab Sham
trials,93,94 it is likely that inadequate treatment to extrafoveal 10
lesions would result in the same problem.
8 6.9
Subfoveal Lesions 6 5.2
For treatment of subfoveal lesions (Fig. 146.12), one first
determines if the lesion is predominantly CNV (at least 50% 4
of the lesion is CNV, totaling areas of any classic CNV and any 2
occult CNV). If the lesion is predominantly CNV, one then
determines (Fig. 146.12) if the lesion is minimally classic or 0
occult with no classic. If so, then one determines if the patient
-2
has had presumed recent disease progression in this eye.
Presumed recent disease progression includes any of the -4 -2.6
following: blood associated with the lesion, visual acuity loss -4.7
within the past 3 months, or growth of the lesion on fluorescein -6
-5.9
SECTION 10

angiography within the last 3 months. In the absence of -8


presumed recent disease progression for a subfoveal lesion with Near activities Distance activities Dependency+
a minimally classic or occult with no classic lesion
composition, careful follow-up, perhaps at 3 week, and then 6 FIGURE 146.17. MARINA trial. Improvement in vision-specific QOL
week, and 3 month, intervals is indicated to watch for following pharmacologic intervention in AMD.
progression to a predominantly classic lesion (see below) or
development of presumed recent disease progression. Lesions
with these compositions were excluded from the anti-VEGF enjoyable activities such as reading a newspaper or recognizing
trials described below, and may have a different natural course faces in social situations.
from the lesions in these trials, suggesting that it would be If the lesion is predominantly classic (Fig. 146.12), one need
inappropriate to extrapolate the results of these anti-VEGF not have presumed recent disease progression since trials
trials to minimally classic or occult with no classic lesions in evaluating treatments for these lesions did not include a
the absence of presumed recent disease progression. requirement for presumed recent disease progression. The
If presumed recent disease progression is noted, then ANCHOR (Anti-VEGF Antibody for the Treatment of
treatment with ranibizumab is considered based on the Predominantly Classic Choroidal Neovascularization in AMD)
MARINA (Minimally Classic/Occult Trial of the Anti-VEGF trial involved 423 patients with predominantly classic
Antibody Ranibizumab in the Treatment of Neovascular AMD) neovascular AMD. This randomized, double-masked, active
trial. The MARINA trial was a Phase III study of 716 patients treatment-controlled study compared safety and efficacy of
with presumed recent disease progression (blood associated 0.3–mg and 0.5–mg doses of ranibizumab over the course of
with CNV, recent visual acuity loss, or recent growth of CNV on 2 years. Results from this study showed that 94% (132/140)
fluorescein angiography) associated with minimally classic or of ranibizumab-treated patients assigned to 0.3 mg and 96%
occult with no classic CNV in AMD. Participants were (134/139) of ranibizumab-treated patients assigned to 0.5 mg
randomized 2:1 to receive either ITV ranibizumab, or sham avoided 15 or more letter loss compared with 64% (92/143) of
injections. The active treatment group was further randomized patients assigned to verteporfin PDT (P<.0001). No additional
to either a 0.3-mg or 0.5 mg-dose of ranibizumab every 4 weeks ocular or systemic safety problems were identified beyond the
for 2 years. One-year data showed that compared with baseline, information summarized above from the MARINA trial.109,110
95% (452/478) of ranibizumab-treated patients lost fewer than Since the ranibizumab-treated subjects without PDT in
15 letters of visual acuity (VA), compared with 62% (148/238) ANCHOR had similar outcomes to the predominantly classic
of patients in the control group.107 Twenty-five percent (59/238) cases enrolled by investigators in the FOCUS trial (where
of patients treated with 0.3 mg of ranibizumab and 34% subjected were assigned randomly to verteporfin PDT with
(81/240) treated with 0.5 mg of ranibizumab improved vision by sham injection or verteporfin PDT plus ranibizumab), it seems
15 letters or more compared with approximately 5% (11/238) of unlikely that adding PDT to ranibizumab will result in better
patients in the control group. Nearly 40% (188/478) of vision outcomes for these lesions. It remains unknown at this
ranibizumab-treated patients achieved a VA score of 20/40 or time if this combination therapy will lead to the need for fewer
better compared with 11% (26/238) in the control group. Side injections, and no trial has compared PDT plus ranibizumab to
effects that occurred more frequently in the ranibizumab arms ranibizumab alone.
than in the control group at 1 year included mild to moderate To examine effects of ranibizumab on patient-reported
conjunctival hemorrhage, eye pain, and vitreous floaters. vision-specific QOL, investigators administered the NEI VFQ-
Uveitis and endophthalmitis occurred in fewer than 1% of 25 to participants in both the MARINA and ANCHOR trials.
patients in the ranibizumab and control groups. Serious The MARINA trial demonstrated a marked increase in the
nonocular adverse events occurred essentially as frequently proportion of patients with clinically relevant improvement in
among ranibizumab-treated patients as among controls.108 vision-specific QOL following pharmacologic intervention in
These data indicate that ranibizumab not only reduces the risk AMD (Fig. 146.17) (Chang TS, Fine JT, Bressler N.. Self-
of moderate or severe visual acuity loss, but also increases the reported vision-specific quality of life at 1 year in patients with
chance of moderate visual acuity gain. While a majority of neovascular age-related macular degeneration in 2 Phase III
treated patients did not improve by 15 or more letters, patients randomized clinical trials of ranibizumab (Lucentis). Poster
who do have this level of improvement following ranibizumab presented at: Annual Meeting of the Association for Research in
treatment may regain abilities dependent on vision they had Vision and Ophthalmology, Fort Lauderdale, Fl, May 1–52006:
1942 previously lost, and may be able to participate in productive and Poster B667) In the MARINA trial, investigators administered
Age-Related Macular Degeneration: Choroidal Neovascularization

the NEI-VFQ 25–716 subjects with subfoveal CNV due to AMD concept that a drug which is usually administered intravenously
with minimally classic or occult with no classic types at to prevent neovascularization in cancer patients could be
baseline and at months 1, 2, 3, 6, 9, and 12. MARINA patients effective and safe over the short term, administered ITV in an
treated with ranibizumab 0.5 mg were more likely to improve AMD patient who had responded poorly to previous treatments.
10 or more points on the NEI-VFQ in activities related to near The report also indicates that, although it is a larger molecule
and distance vision than were patients who received sham than ranibizumab, bevacizumab apparently penetrated the
injections (P < 0.001), who experienced overall decreases in retina well enough to inhibit VEGF in the vitreous cavity. Since
their ability to perform these activities (Fig. 146.17) (Chang TS, then, additional reports have appeared in the literature
Fine JT, Bressler N.. Self-reported vision-specific quality of life at presenting safety evaluations of the drug when injected into the
1 year in patients with neovascular age-related macular vitreous of rabbits,109 immunofluorescent studies suggesting
degeneration in 2 Phase III randomized clinical trials of the ability of bevacizumab to penetrate the retina,109 and
ranibizumab (Lucentis). Poster presented at: Annual Meeting evidence of potential clinical efficacy.110 A large, well-designed,
of the Association for Research in Vision and Ophthalmology, clinical trial would be needed to establish more definitively the
Fort Lauderdale, Fl, May 1–5, 2006: Poster B667). In addition, strength of applying these results to patients with neovascular
ranibizumab-treated patients were more likely to improve 10 or AMD.
more points in ratings of dependency on others because of In summary, the strength of the evidence to warrant the tse
vision than were sham injection patients. of ranibizumab for specific lesions described above is based on
Similar benefits of ranibizumab for patients with predomi- multiple randomized clinical trials with relevant endpoints with

CHAPTER 146
nantly classic lesions were confirmed in the ANCHOR trial. respect to visual acuity and a patient’s perception of vision-
Investigators administered the NEI-VFQ 25 to 418 AMD patients related quality of life. The strength of evidence to support use of
with predominantly classic type at baseline and at months 1, 2, bevacizumab is weaker, based on case series and anecdotal
3, 6, 9, and 12 as reported at the 2006 Annual Meeting of the reports. It is unknown at this time if intravitreal bevacizumab
American Society of Retina Specialists, although QOL data have is almost as good, better, or worse than ranibizumab for
not yet been published in a peer-reviewed journal. reducing the risk of visual acuity loss and increasing the chance
of visual acuity gain. However, the evidence may be strong
Other treatments for subfoveal neovascular AMD enough to warrant consideration of bevacizumab when
Bevacizumab was approved by the FDA for intravenous ranibizumab is not available to a patients because of regulatory
administration to patients with metastatic colorectal cancer limitations (e.g., not yet approved for use in a country) or
in February 2004. Bevacizumab has a similar molecular financial limitations (since a single-use vial of ranibizumab
structure to ranibizumab. Initially, a report about a series of costs a physician approximately $2000 in 2007 compared with
AMD patients who were treated with systemic bevacizumab, approximately $50 when 1.25 mg of bevacizumab in 0.05 ml is
and one case report of bevacizumab ITV administration, obtained from a compounding pharmacy making multiple doses
showed short-term vision and anatomic outcomes that from a single-use vial of bevacizumab that was packaged for
appeared similar to those seen with ranibizumab and unlike the intravenous use in the treatment of certain cancers).
natural history of this disease. Based on these reports, some
ophthalmologists began to administer bevacizumab off-label to Pegaptanib sodium
treat AMD. During two concurrent, 48 week, randomized, double-blind,
Specifically, an open-label, uncontrolled, clinical study controlled clinical trials, patients received ITV pegaptanib in
evaluated the short-term safety of systemic bevacizumab in one eye at a dosage of 0.3 mg, 1.0 mg, or 3.0 mg. The control
nine AMD patients with subfoveal CNV, with best-corrected VA group was given sham injections every 6 weeks.44 The primary
of 20/40 to 20/400. Patients received an infusion of outcome variable was the proportion of patients who lost fewer
bevacizumab (5 mg/kg) at baseline, followed by one or two than 15 letters of VA at week 54. As compared to 164 (55%) of
additional doses at 2 week intervals. By 12 weeks, the median 296 patients in the control group, 206 (70%) of 294 patients
VA scores in the study eyes increased by 8 letters (P = 0.011). treated with 0.3 mg pegaptanib responded (P < 0.001), as did
The median central retinal thickness decreased by 157 mm (P = 213 (71%) of 300 those treated with 1.0 mg pegaptanib (P <
0.008). No serious ocular or systemic adverse events were 0.001), and 193 (65%) of 296 those treated with 3.0 mg
identified during safety assessments, which were performed at pegaptanib (P = 0.03). The majority of patients in all three
all visits. The only treatment-related adverse event was a pegaptanib groups (95–97%) and in the sham group (95%
treatable, reversible, mild elevation of systolic blood pressure experienced at least one adverse event.111
(+12 mm Hg; P=.035). Although, the improvements in VA, Vitreous hemorrhage occurred in 2% of pegaptanib-treated
OCT measurements, and angiography (marked improvement eyes, endophthalmitis in 1.3%, traumatic cataract in 0.6%, and
in vessel leakage in all eyes) in this small sample suggest that retinal tear or detachment in 0.7%.44,111 Based on this study,
systemic bevacizumab may be efficacious in neovascular AMD, pegaptanib was approved for the treatment of neovascular
a case series of nine patients does not constitute an adequate AMD and could be considered for patients with lesions similar
evidence base to conclude that systemic bevacizumab®therapy to those of patients enrolled in these trials. However, because
is safe and effective for neovascular AMD patients.107 the results were far different from that obtained with
The single case report of ITV bevacizumab involved a 63- ranibizumab, most ophthalmologists would not consider the
year-old woman with neovascular AMD, whose vision had use of pegaptanib over ranibizumab at this time.
deteriorated despite treatment with PDT/triamcinolone acetate A retrospective post hoc analysis was performed in AMD
and pegaptanib was a sentinel event leading to ophthalmologists’ patients with ‘early’ disease defined retrospectively in two ways:
use of intravitreal bevacizumab while FDA approval for Group 1 (n=34) characteristics include lesion size <2 disc
ranibizumab was pending.108 The patient received a single areas, baseline VA letter score ≥ 54, and no prior PDT or laser
1.0 mg ITV dose of bevacizumab. Within 1 week, the subretinal photocoagulation; Group 2 (n = 30) characteristics included
fluid resolved such that the macular contour appeared normal occult with no classic CNV, no lipid, and VA in study eye worse
on OCT, and remained so for at least 4 weeks. VA remained than that in fellow eye.112 It is unknown how many
stable, and neither ocular inflammation nor angiographic combinations of characteristics were examined to form with
leakage was observed. This case report represents proof of these groups. In pegaptanib-treated Groups 1 and 2, 26 (76%) 1943
RETINA AND VITREOUS

and 24 (80%) of 34 and 30 subjects, respectively, lost <15 longer-term efficacy seen with ranibizumab, but not shown for
letters of VA (the primary efficacy measure), compared with 14 pegaptanib or verteporfin PDT).
(50%) and 20 (57%) of 28 and 35 subjects assigned to sham The necessity for ITV administration of ranibizumab as often
injections. In the pegaptanib Groups 1 and 2, 4 (12%; Group 1) as every month and pegaptanib as often as every 6 weeks is a
to 6 (20%; Group 2) of 34 and 30 subjects, respectively, gained distinct disadvantage compared to topical applications used to
at least 15 letters of VA compared with 1 (4%) and none of 28 treat other eye diseases and different from the potential
and 35 subjects, respectively, receiving sham injections. These disadvantages of the intravenous administration of verteporfin
data suggest that pegaptanib may be more effective if treatment PDT, with sunlight restrictions for several days and follow-up as
is started early in the natural history of a patient’s disease. This often as every 3 months. However, clinical trials with
conclusion should be evaluated with caution, however, as based ranibizumab and pegaptanib were able to successfully recruit
on these small numbers, the confidence intervals surrounding participants who continued treatment throughout the 2 years
the estimated percentage values are likely to be rather wide. of the trials, despite the administration of ITV, so it appears
While PDT with verteporfin was shown to be superior to no to be acceptable despite the invasiveness of the procedure.
treatment for predominantly classic subfoveal lesions (TAP
REPORT NO. 1 1999), its inferiority to ranibizumab in the
ANCHOR trial for these lesions suggests that few ophthal- MANAGING PATIENTS AND PHYSICIANS’
mologists would consider verteporfin PDT over ranibizumab in EXPECTATIONS IN THE TREATMENT OF
most circumstances if both drugs are available. Similarly, NEOVASCULAR AMD
SECTION 10

although no head-to-head trial of ranibizumab to verteporfin


PDT has been done for minimally classic or occult with no Ophthalmology patients’ satisfaction in care hinges on an
classic lesions with presumed recent disease progression, the adequate explanation of diagnosis, treatment, and prognosis.
outcomes reported in the MARINA trial for such lesions with Based on interviews of 163 ophthalmology patients interviewed
ranibizumab seem far superior to those reported to verteporfin at either a Veteran’s Administration hospital or private practice,
PDT (VIP REPORT NO. 2). more than 90% of patients rate these explanations as the most
With respect to safety, the risk of endophthalmitis in any important determinant of their satisfaction with the medical
invasive ocular procedure may be minimized with proper visit.116 Although all 19 ophthalmologists interviewed as part of
aseptic technique, the use of a lid speculum, careful this study rated explanations of diagnosis and treatment as very
postinjection monitoring, and educating patients about which important to patient satisfaction, only 40% of them also rated
postinjection symptoms to report to the physician. a discussion of prognosis as highly.Trobe Ophthalmology 1983,
Bevacizumab is not manufactured with the strict particulate P53A, T2, items 1–3 This dichotomy in expectations about
matter limits that are required for ocular drugs, such as revealing prognosis indicates that patients’ expectations may be
ranibizumab, nor does it contain preservatives. Theoretically, at odds with those of their physicians on this issue.
when dividing bevacizumab into smaller doses required for ITV Patients expect that their ophthalmologists will explain AMD
administration, contamination could be introduced if not done treatment options in ways they can understand, yet a sizable
under aseptic technique, and the label currently indicates the group of patients may have problems with health literacy. In
need to use the drug within 6 h of opening a vial. one study of Medicare enrollees in a nationwide managed-care
Although it is possible that the body could develop antibodies system, one third of English-speaking patients, and more than
against VEGF inhibitors, no serum antibodies to ranibizumab one half of Spanish-speaking patients, exhibited either marginal
were detected in the dose-ranging trial.113 One published trial or inadequate health literacy, and thus were unable to
of pegaptanib did not report evaluations of serum antibodies,114 understand consent forms or medicine labels. In addition,
and another reported that there were no serum antibodies or health literacy tended to decline with age, with inadequate
hypersensitivity reactions to pegaptanib.44 health literacy exhibited in only 15.6% of participants 65–69
Another potential concern is the possibility that VEGF years old and increasing to 58.0% of those older than 85. If an
inhibitors may potentiate myocardial infarction and stroke, which ophthalmologist uses the same level of language with all
has occurred at higher rates in treatment than control groups in patients, there is a risk, therefore, that a substantial minority
trials of intravenous bevacizumab among cancer patients.115 of patients will be unable to comply with treatment plans. This,
Thromboembolic events have occurred among intraocular rani- in turn, can lead to patient disappointment and ophthalmologist
bizumab and pegaptanib trial participants, but the frequency frustration, since a treatment would likely be more effective if
was not statistically different between treatment and control the patient were compliant with instructions.
groups.44 No data has been published regarding the risk of such Those patients with AMD who are literate enough to
events following intraocular injection of bevacizumab. understand the implications of the disease process expect that
In summary, based on clinical trials, ranibizumab appears to their preferences and concerns with regard to treatment options
offer the best outcome at this time in the treatment of selected will be considered by their ophthalmologists. They may arrive
cases of neovascular AMD similar to those enrolled in at the ophthalmologist’s office clutching reams of printouts
MARINA, ANCHOR, VISION, the TAP Investigation, or the from the Internet, expecting that their physician will embrace
VIP Trial compared with pegaptanib or PDT with verteporfin. what they have found from web sites that may or may not be
Additional data are needed to determine if bevacizumab results reliable.117 As a result, patients with AMD may develop
in similar efficacy and safety to warrant its use when ranibizumab unrealistic expectations of treatment effectiveness and safety,
is available. When ranibizumab is not available because of based on selective attention to potential positive outcomes that
regulatory limitations or cost, bevacizumab might be considered. are widely publicized on the Internet or in popular publications.
However, the patient must be informed of the risks and benefits Patients may misunderstand and assume that positive
of extrapolating the ranibizumab trial results to bevacizumab outcomes in a proportion of participants treated during a
based on the limited evidence published to date regarding clinical trial will occur to them as individuals. A patient who
potential benefits of bevacizumab and the limited safety data has read about ranibizumab, for example, may assume that if
available compared with the more extensive safety data treated with that drug, he or she will regain vision and be able
available on pegaptanib or verteporfin PDT (assuming the to drive, read, and recognize faces again because ~40% of
1944 short-term efficacy noted with bevacizumab translates to the ranibizumab-treated patients did so.118
Age-Related Macular Degeneration: Choroidal Neovascularization

with them the desire for explanations of diagnosis, treatment,


PATIENT EXPECTATIONS prognosis that the patients can understand and apply. Most
In the face of AMD, a disease with uncertain outcomes, several patients will accept treatment that the physicians suggest (e.g.,
strategies may be useful to manage patient expectations. These watchful waiting, surgery, photodynamic treatment, intravitreal
strategies, first identified among general medical practitioners, injection). Clinicians cannot predict which patients will benefit
include an honest discussion of the natural history of the from a given therapeutic intervention and which will not. Thus,
disease without treatment, the potential outcomes with treatment decisions for AMD patients should be weighed
treatment, and the prognosis with or without treatment carefully to maximize the chances for vision retention or
(watchful waiting).119 improvement. One way to maximize rational treatment choice
The ophthalmologist should first explain that some patients involves the concept of the number needed to treat (NNT).
with treatment will lose vision. For example, when discussing NNT is an estimate of the number of patients who need to be
ranibizumab as a possible treatment with patients, it should be treated with a given regimen (instead of the standard treatment)
explained that 5% of treated patients lost a moderate amount of for one additional patient to benefit from a specific outcome.
vision (15 or more letters, ~3 or more lines on the eye chart). This benefit is often expressed as an outcome to avoid, e.g.,
However, without treatment, patients should understand that three or more lines of vision loss, or a desirable outcome, e.g., 3
35–40% will lose at least a moderate amount of vision. or more line gain. The ideal NNT would be one, meaning that
Likewise, patients should understand that treatment does not every patient treated with a given regimen would benefit.
take away one’s chance of improving, but that not all treated The NNT is the reciprocal of the absolute risk reduction.

CHAPTER 146
patients will improve. Clinical trials suggest that few untreated Assuming the standard treatment is verteporfin PDT, the NNT
patients will improve a moderate amount (three or more lines for ranibizumab to avoid three or more line loss is calculated as
on an eye chart), and that with verteporfin PDT or pegaptanib shown in Figure 146.18, based on efficacy data from the
injections only ~5–10% will gain this amount of vision. With ANCHOR (Anti-VEGF Antibody for the Treatment of
ranibizumab, 25–40% (depending on drug dose and lesion Predominantly Classic CHORoidal Neovascularization in
composition) gained three or more lines of vision by 1 year, with AMD) Trial. If the clinician is considering ranibizumab or
the proportion of study subjects having this improvement pegaptanib for an AMD patient, for example, the treatment
remaining fairly stable through 2 years. This means that a with the lower NNT with avoidance of vision loss as the
majority of patients will not have noticeable improvement, but specified outcome would be the optimal choice for the
it is far more likely that one will experience improvement with patient.122
ranibizumab treatment than without treatment or with other Ophthalmologists want to prescribe treatments that are cost-
treatments currently available. effective for their patients. A costly intervention for AMD may
Another strategy that is essential to managing patients’ be worth the financial outlay if it truly benefits patients, as the
expectations is to be sure the patient understands, and is willing gift of sight may be considered priceless. The next newsletter in
and able to comply with, the treatment plan.Hewson J Gen Int this series will discuss cost-effectiveness of AMD treatments in
Med 1996; P483, T1, item 5 If pegaptanib or ranibizumab is detail.
being considered as a treatment option, for example, one should
describe the need for multiple visits, multiple imaging
procedures, and multiple injections. While it is unclear at this
MANAGEMENT CONSIDERATIONS FOR
time whether monthly injections (MARINA, ANCHOR) are PHYSICIANS
superior to less frequent injections (PIER), patients receiving Between one-quarter and one-half of Americans read at the
ranibizumab need to know that injections could be as often as 8th grade level or below, despite having graduated from high
monthly for at least 2 years, and possibly for a longer time. As school.123 Ideally, therefore, the ophthalmologist should
more knowledge is gained regarding longer term outcomes, this determine the patient’s health literacy level. In a research
information should be shared with patients. The ophthalmologist setting, clinicians might determine the patient’s health literacy
also must make the patient aware that there are potential risks
of treatment due to either the drug itself (none known
definitively at this time with ranibizumab) or to the administration
of the drug by injection (e.g., such as endophthalmitis or retinal
tears.120 Number Needed to Treat
Given the expenses of some drugs in the management of
neovascular AMD, the treating ophthalmologist should be sure
the patient understands the total expected cost of treatment Proportion with vision loss (PDT) .36
over time, and what proportions will be paid by the patient’s
third-party payer, if any, compared with those that the patient
must pay out-of-pocket. A 2 year course of pegaptanib injections Proportion with vision loss (ranibizumab*) .04
every 6 weeks, for example, may cost more than $19 000.121
The price of ranibizumab, which was administered once a
month in the pivotal trials, will be $1950 per injection to the Absolute Risk Reduction (ARR) .32
ophthalmologist, with an additional mark-up to the patient, for
a total of $23 000 per year to the heath care provider just for the
drug. Additional costs for visits, imaging, and mark-up on the NNT = 1/ARR 1/.32=
drug need to be explained as well. 3.125

OPHTHALMOLOGISTS’ EXPECTATIONS *Dosage, ranibizumab .5mg


The factors mentioned above that govern patient expectations
for treatment also influence their physicians. Ophthalmologists FIGURE 146.18. Calculation of NNT based on data from the
want their patients to be satisfied with their care, and share ANCHOR clinical trial. 1945
RETINA AND VITREOUS

level using the Short Test of Functional Literacy in Adults, ranibizumab treatments followed by quarterly injections may
which is available in both English and Spanish, and takes only confer visual acuity, FA, and OCT benefits compared with sham.
12 min to administer. If that is not possible in the private While such a regimen decreases the inconvenience and risk of
setting due to time and staffing constraints, simply asking the more frequent injections, it is unknown how this regimen
patient, “How often do you need to have someone help you to compares with respect to visual acuity outcomes to the monthly
read instructions, pamphlets, or other written material from treatments used in the MARINA and ANCHOR trials.
your doctor or pharmacy?” can be used as a proxy.124 While waiting for data to determine if withholding treatment
The ophthalmologist should consider providing AMD based on parameters that indicate no room for further
patients with recommendations for easy-to-understand improvement or a stable lesion leads to outcomes that are
educational materials, such as those available from the almost as good, better, or worse than monthly injections of
American Academy of Ophthalmology. The Medline Plus Macular ranibizumab or bevacizumab, and while waiting for evidence if
Degeneration Patient Education Page (http://www.nlm.nih.gov/ either retreatment approach with bevacizumab is almost as
medlineplus/maculardegeneration.html) also provides reliable good, better, or worse than either retreatment approach with
material targeted to the lay public. Frequent updates may be ranibizumab, ophthalmologists are approaching follow-up in
needed as new data are presented or published. one of two ways.
One, they may be applying ranibizumab treatments as it was
done in the trials proving its benefits, i.e., monthly injections
MONITORING PATIENTS FOLLOWING for up to 2 years. Two, knowing from the PIER study that less
SECTION 10

INITIATION OF RANIBIZUMAB THERAPY frequent treatments also are beneficial compared with no
FOR NEOVASCULAR AMD treatment (although not knowing if this is almost as good,
better, or worse than monthly injections), some ophthalmologists
Several investigators have published case series about are withholding additional treatments when there no longer is
monitoring retinal structure with OCT following intravitreal room for improvement or the lesion is stabilized. One might
bevacizumab when used to treat CNV. Avery and colleagues consider that there is no longer room for improvement when
treated 79 patients with intravitreal bevacizumab monthly. the visual acuity is as good as it could possibly be, for example,
While they did not specify whether fluorescein angiography was is 20/25, taking into consideration the patient’s media
performed at each visit, patients followed for 4–15 weeks opacities, refraction, and structural damage to the macula from
showed a decline in central retinal thickness as measured by atrophy or scarring or other degeneration. One might consider
OCT of at least 10% in 41 (55%) of 75 patients seen 1 week a lesion stabilized when certain parameters appear absolutely
postinjection.116 Spaide and colleagues used changes in retinal the same as the previous one or two months with respect to
thickness (as measured in the central subfileld on OCT) as a symptoms, visual acuity, biomicroscopic appearance (aided by
primary outcome measure in a retrospective study of 266 documentation with fundus photographs), OCT, and
patients who had been treated with intravitreal bevacizumab.125 fluorescein angiography.
Ophthalmologists for patients in this series justified performing Clearly, this latter approach is more time intensive for the
FA as needed, rather than at every follow-up visit, because they patient and physician, with no strong evidence that outcomes
deemed slit-lamp biomicroscopy and OCT adequate to measure based on this approach are almost the same or better than those
initial response to VEGF-inhibitor therapy.125 However, obtained by monthly injections regardless of these parameters.
measuring a response to therapy does not determine if Also, while symptoms, visual acuity, and biomicroscopic
retreatment decisions based on OCT, or fluorescein angiography, appearance add little cost to a visit, it is unknown if more costly
or both, lead to better outcomes than if none of these fundus photographs aid in determining if a lesion is stable (as
evaluations are performed after initiating anti-VEGF therapy. has been demonstrated with follow-up of diabetic retinopathy.127
The pivotal trials leading to the FDA approval of It is also unknown if making treatment decisions based on the
ranibizumab, ANCHOR (Anti-VEGF Antibody for the Treatment increased cost of periodic OCTs are beneficial or preclude the
of Predominantly Classic CHORoidal Neovascularization in need for the increased cost of fluorescein angiography (with its
AMD) and MARINA (Minimally Classic/Occult Trial of the occasional difficulties with interpretation and its attendant side
Anti-VEGF Antibody Ranibizumab in the Treatment of effects of nausea, vomiting, itching, or rash),128 or making
Neovascular AMD) gave monthly injections of ranibizumab treatment decisions based on fluorescein angiography are
through 2 years, without considering withholding retreatment beneficial or preclude the need for OCT. Since ranibizumab is
based on OCT or fluorescein angiographic findings. Thus, these expensive to the patient and third party payers, if periodic
trials do not permit one to determine if almost as good, worse, fundus photographs, OCT, or fluorescein angiograms were
or better outcomes would be obtained if treatment were shown to identify situations when retreatment could be
withheld when it was judged that there was no room for withheld with outcomes that were almost as good as monthly
continued improvement or when it was judged that the injections, the cost savings on the drug could outweigh the
condition appeared ‘stabilized’. additional monitoring costs of fundus photographs, or OCT, or
Although the protocols for these pivotal trials specified fluorescein angiography or a combination of these. If
follow-up visits and retreatment with ranibizumab once per bevacizumab, which costs far less than ranibizumab for
month, the possibility that the interval between ranibizumab intraocular injection, were shown to provide outcomes that
treatments could be extended to balance efficacy with comfort were almost as good or better than ranibizumab outcomes, then
and safety has not been determined definitively. The PIER trial avoiding the cost of a few injections might not balance the costs
(Phase 3b, Randomized, Double-Masked, Sham Injection- of the photographs, or OCT, or angiograms.
Controlled Study of the Efficacy and Safety of Ranibizumab in Clearly more data is needed to make the ophthalmologist
Subjects With Subfoveal Choroidal Neovascularization With or confident that use of monitoring with one or more procedures,
Without Classic CNV Secondary to AMD) showed that at including fundus photographs, OCT, and fluorescein angiograms,
month 12, patients treated with ranibizumab lost 1.6 letters to determine retreatment leads to almost as good, or better
and 0.2 letters (0.3 mg and 0.5 mg) compared to a loss of 16.3 outcomes than seen with monthly ranibizumab treatments.
letters in the sham group, on average (P < 0.0001)126 The 1 year Until strong evidence is available, some ophthalmologists
1946 results of PIER suggest that an initial induction with 3 monthly appropriately will apply retreatment each month for up to
Age-Related Macular Degeneration: Choroidal Neovascularization

100
0 (n = 35)
1 (n = 105)
90 2 (n = 142) 87
3 (n = 105)
81
4 (n = 45)
Eyes with choroidal neovascularization (%)

80
72
70 68

61
60
53
50 48 49
47
44
40
40 38
36
34
32
30
30 28 28

CHAPTER 146
27
25
23 22 23 23
20 20
20 19
16
15
12 14 13 13
11
10 7 7 7 7 7 7
4 5
3 3 3
0 0 0 0 0 0
0
0 1.5 3 6 12 18 24 30 36 42 48 54 60
Follow-Up (mo)

FIGURE 146.19. Incidence of CNV by number of risk factors present including definite hypertension, 5 or more drusen, greatest linear
dimension of largest druse >63 µm, and focal hyperpigmentation.
From Macular Photocoagulation Study Group: Risk factors for choroidal neovascularization in the second eye of patients with juxtafoveal or subfoveal choroidal
neovascularization secondary to age-related macular degeneration. Arch Ophthalmol 115:741–747, 1997. Copyright 1997, American Medical Association.

2 years, while others will use weaker evidence (inference from The MPS group has shown that if a patient presents with CNV
case series, biologic rationale, and experience) to consider in one eye and no evidence of CNV or scarring in the fellow
withholding treatment when there is no room for further eye, about 40% of these eyes will have CNV in the fellow eye
improvement or when the situation appears stabilized. A trial within 5 years.1,2 The risk of CNV developing in these eyes
announced by the National Eye Institute129 will address some may be further specified depending on the drusen and RPE
of these issues both with ranibizumab and bevacizumab. abnormalities within 1500 mm of the foveal center present in
the fellow eye and certain patient characteristics at baseline.
NATURAL COURSE OF SUBFOVEAL 2 Specific risk factors include: (1) definite hypertension (systolic
SUBRETINAL HEMORRHAGE IN AMD pressure ≥140 mm Hg, diastolic pressure ≥90 mm Hg, or use of
antihypertensive medications), (2) five or more drusen, (3)
Several reports have noted that whereas some eyes with greatest linear dimension of largest druse greater than 63 mm, or
subfoveal subretinal hemorrhage associated with AMD have (4) focal hyperpigmentation. The risk of CNV developing
poor outcomes, the visual acuity of other eyes do not deteriorate within 5 years after presenting with CNV in the first eye ranged
or may improve spontaneously.8,130,131 Such findings underscore from 7% if none of these risk factors was present to 87% if all
the importance of evaluating the role of therapeutic interventions four risk factors were present (Fig. 146.19).2 As one might have
for these cases, such as surgery to remove subretinal suspected, for the fellow eyes that had no evidence of CNV or
hemorrhage,132,133 in randomized clinical trials. In one such scarring at baseline and subsequently developed CNV within 5
trial from the Submacular Surgery Trials, surgical removal of years, the average visual acuity loss was eight lines of vision.141
blood and any associate neovascular lesion was not shown to In contrast, for the two-thirds of fellow eyes that had no
reduce the risk of vision loss in most cases and was associated evidence of CNV or scarring at baseline and did not have CNV
with a high risk of rhegmatogenous retinal detachments. (ref within 5 years, the average visual acuity loss was less than half
sst report no. 13) The control group (no treatment) did note a line of vision (specifically, 0.4 line).141 Therefore, the risk of
that ~20% of predominantly hemorrhagic lesions will have a moderate or severe loss of visual acuity is highly correlated with
breakthough vitreous hemorrhage. Other treatments under the development of CNV during follow-up. Eyes that do not
investigation include the use of intravitreal ranibizumab acquire CNV during follow-up have little or no visual acuity
(although these lesions were excluded from the MARINA and loss. This finding should be considered when evaluating the role
ANCHOR trials), as well as using intravitreal gas to displace of vitamins, minerals, or other treatment modalities in
the hemorrhage. preventing visual acuity loss for eyes with drusen or RPE
abnormalities without the presence of CNV or scarring.
If one couples this information of fellow eye involvement
RISK OF FELLOW EYE INVOLVEMENT WITH with success of treatment in the first eye from the extrafoveal
SUBSEQUENT CNV SCARRING argon MPS trial, one can determine the risk of legal blindness
It is important to have the patient monitor the vision in the (visual acuity of 20/200 or worse in both eyes) when treating the
fellow eye if its macula does not already have CNV scarring. first eye. The risk of legal blindness, again, is highly correlated 1947
RETINA AND VITREOUS

with whether or not the fellow eye had CNV or scarring at into the magnifying system. Closed-circuit television and hand-
baseline. If CNV or scarring was present in the fellow eye at held electronic devices provide electronic magnification at
baseline, it was likely that the fellow eye already had significant greater levels than is possible with optical systems. These also
loss of visual acuity and that there was a high risk of recurrence provide binocular viewing and can be used at a comfortable
developing in the treated eye. Forty-nine percent of patients in reading distance. Unfortunately, not all systems are readily
the argon AMD MPS trial were legally blind within 5 years of portable and all are quite costly. Writing assistance may be
follow-up when the fellow eye initially had CNV or scarring, provided with a weak hand lens or a +5.00 add-in spectacle.
whereas only 12% of patients were legally blind within 5 years Additional invaluable and simple tools to aid in writing include
of follow-up when the fellow eye had no evidence of CNV or signature guides, black felt-tip pens, and bold wide-ruled paper.
scarring initially.134 Similar analyses currently are under way Distance vision may be enhanced with binocular field glasses
for patients who participated in the MPS trials of juxtafoveal or and opera glasses. Lightweight telescopes can be hand-held or
subfoveal lesions from AMD. Of course, if the visual acuity of mounted onto spectacles. Unfortunately, these instruments
the fellow eye is compromised at all, and the first eye has CNV, distort distances and limit peripheral vision; therefore, their
low-vision aids are probably indicated to assist with magnification applicability is limited to tasks of sedentary distance viewing
for near visual tasks, as are telescopic aids for spotting at and distance spotting.
distance. Finally, the success of each of these aids is highly variable,
depending on the visual deficit and the motivation of the
patient. They can attempt to enhance only lighting or
LOW-VISION AIDS
SECTION 10

magnification for the remaining vision. The patient should not


Patients who have central visual impairment in both eyes look on them as a treatment for macular degeneration nor
should be advised about the availability of a variety of low-vision expect to regain central vision. Nevertheless, it is important to
aids. A low-vision evaluation includes not only prescribing give these patients every opportunity to be evaluated for and
appropriate lenses, magnifying aids (optical or electronic), and trained in the use of optical and nonoptical systems that may
other items of assistance but also properly training and serve to improve the quality of life.
encouraging the patient to use them. Patients should also be
told of community resources available to assist them with CONCLUSIONS AND FUTURE RESEARCH
visual impairment such as might be obtained from the directory
of agencies serving the visually handicapped in the United The 1980s and 1990s provided enormous progress in the
States provided by the American Foundation for the Blind. A management of AMD. Specifically, laser treatment has been
realistic appraisal of the prognosis coupled with appropriate shown to be beneficial when compared with no treatment.
counseling and support is necessary to enable the visually However, the fact that laser treatment is more beneficial for
impaired patient to continue functioning as normally as possible preserving vision when compared with observation alone is
despite the central visual impairment. The American Foundation only a start. Successful cases of laser treatment still result in
for the Blind can also inform patients as to which aids, such as central visual acuity loss for the patient, especially when
talking books, large-print books, watches that ‘tell’ time, closed- treatment involves the foveal center as it so often does in AMD.
circuit television readers, and other devices, are available Furthermore, treatment is applicable in only a minority of
through the directory of products for people with vision cases.
problems. Therefore, future research in the management of CNV in
Visual aids for detailed near and distance tasks, such as AMD will have to evaluate other modalities of treatment that
reading, writing, typing, sedentary distance viewing, and do not cause destruction of the foveal center or therapies that
distance spotting for street signs, can be prescribed with a prevent the development of CNV in the first place.135 Such
thorough low-vision examination conducted in a room with investigations will require interdisciplinary approaches to this
glare-free high-intensity lighting and large trial lenses to allow problem that will probably include epidemiologists, clinicians,
for eccentric fixation. There is a wide range of magnification psychophysicists, vitreoretinal surgeons, geneticists, and
levels available in the present armamentarium of low-vision cellular biologists. These investigations may include therapies
aids. Some incorporate their own illumination source. Each to prevent the development of CNV or to minimize the damage
aid has certain advantages and limitations, so a patient must it causes. Medical interventions that affect angiogenesis may
decide with a low-vision aid specialist what combination of hold some promise. Unfortunately, one randomized clinical
aids is best for her or his visual objectives and visual needs. trial evaluating interferon-a showed no benefit and possible
Four types of reading aids are available: reading glasses, hand- harm in the treatment of CNV from AMD.136 Surgical
held lenses, stand magnifiers, and electronic devices. Reading interventions that could remove the neovascular process before
glasses (convex lenses) provide relatively large fields of vision, it destroys a large area of the central retina also may hold some
but the strongest lenses require short working distances. promise, especially because these interventions, as with
Telescopic reading glasses increase the working distance; medical interventions, may be considered for lesions obscured
however, they allow a relatively smaller field of view and shorter by blood or that have poorly demarcated boundaries.137 Other
depth of focus than a simple high plus reader of comparable currently unproven investigational therapies in the 1990s
magnification. Hand-held magnifiers may suffice for quick, include consideration of indocyanine green-guided laser
simple tasks that are conducted at arm’s length, such as photocoagulation,103,138,139 radiation,140 and photodynamic
adjusting a stove dial. A stand magnifier consists of a mounted therapy.141,142 Given the variable natural history of CNV in
lens that will remain in focus if placed on the reading material AMD, acceptance of any of these new therapies likely will
and held so that its focal point corresponds to the focal point of require therapeutic benefits proved through carefully designed
the patient’s near correction. This device offers an alternative randomized clinical trials. As the number of people over the age
to the high plus lens for the weak or tremulous patient; it is of 65 years in the United States and elsewhere doubles by about
also useful when the patient wishes an increase in working 2030, the importance of finding treatments for CNV in AMD
distance or wants to have an illumination source incorporated will grow tremendously.

1948
Age-Related Macular Degeneration: Choroidal Neovascularization

REFERENCES
1. Bressler SB, Maguire MG, Bressler NM, 18. Friedman E, Ivry M, Ebert E, et al: 37. Macular Photocoagulation Study Group:
Fine SL: Macular Photocoagulation Study Increased scleral rigidity and age-related Subfoveal neovascular lesions in age-
Group: Relationship of drusen and macular degeneration. Ophthalmology related macular degeneration: Guidelines
abnormalities of the retinal pigment 1989; 96:104–108. for evaluation and treatment in the Macular
epithelium to the prognosis of neovascular 19. Eye Disease Case-Control Study (EDCCS) Photocoagulation Study. Arch Ophthalmol
macular degeneration. Arch Ophthalmol Group: Risk factors for neovascular age- 1991; 109:1242–1257.
1990; 108:1442–1447. related macular degeneration. Arch 38. Klein ML, Zorizzo PA, Watzke RC: Growth
2. Macular Photocoagulation Study Group: Ophthalmol 1992; 111:1701–1708. features of choroidal neovascular
Risk factors for choroidal 20. Broekhuyse RM: The lipid composition of membranes in age-related macular
neovascularization in the second eye of the aging sclera and cornea. degeneration. Ophthalmology 1989;
patients with juxtafoveal or subfoveal Ophthalmologica 1975; 171:82. 96:1416–1421.
choroidal neovascularization secondary to 21. Grossniklaus HE and Green WR: Choroidal 39. Vander JF, Morgan CM, Schatz H: Growth
age-related macular degeneration. Arch neovascularization. Am J Ophthalmol 2004; rate of subretinal neovascularization in age-
Ophthalmol 1997; 115:741–747. 137:496–503. related macular degeneration.
3. Gass JDM: Drusen and disciform macular 22. Witmer AN, et al: Vascular endothelial growth Ophthalmology 1989; 96:1422–1429.
detachment and degeneration. Arch factors and angiogenesis in eye disease. 40. Macular Photocoagulation Study Group:
Ophthalmol 1973; 90:206–217. Prog Retin Eye Res 2003; 22:1–29. Laser photocoagulation of subfoveal
4. Smiddy WE, Fine SL: Prognosis of patients 23. Adamis AP ,Shima DT: The role of vascular neovascular lesions in age-related macular

CHAPTER 146
with bilateral macular drusen. endothelial growth factor in ocular health degeneration. Results of a randomized
Ophthalmology 1984; 91:271–277. and disease. Retina.2005; 25:111–118. clinical trial. Arch Ophthalmol 1991;
5. Strahlman E, Fine SL, Hillis A: The second 24. Bressler SB, Bressler NM, Alexander J, 109:1219–1231.
eye of patients with senile macular Green WR: Clinicopathologic correlation of 41. Macular Photocoagulation Study Group:
degeneration. Arch Ophthalmol 1983; occult choroidal neovascularization in age- Laser photocoagulation of subfoveal
101:1191–1193. related macular degeneration. Invest recurrent neovascular lesions in age-related
6. Green WR, McDonnell PH, Yeo JH: Ophthalmol Vis Sci 1991; 32:689. macular degeneration. Results of a
Pathologic features of senile macular 25. Bressler SB, Bressler NM, Fine SL, et al: randomized clinical trial. Arch Ophthalmol
degeneration. Ophthalmology 1985; Natural course of choroidal neovascular 1991; 109:1232–1241.
92:615–627. membranes within the foveal avascular 42. Submacular Surgery Trials Research Group:
7. Sarks SH: Ageing and degeneration in the zone in senile macular degeneration. Am J Guidelines for interpreting retinal
macular region: A clinicopathologic study. Ophthalmol 1982; 93:157–163. photographs and coding findings in the
Br J Ophthalmol 1976; 60:324–341. 26. Bird AC: Macular disciform response and submacular surgery trials (SST). SST report
8. Avery RL, Fekrat S, Hawkins BS, Bressler laser treatment. Trans Ophthalmol Soc UK no. 8. Retina 2005; 25:253–268.
NM: Natural history of subfoveal subretinal 1977; 97:490–493. 43. Barbazetto I, Burdan A, Bressler NM, et al:
hemorrhage in age-related macular 27. Bressler NM, Frost LA, Bressler SB, et al: Treatment of Age-Related Macular
degeneration. Retina 1996; 16:183–189. Natural course of poorly defined choroidal Degeneration with Photodynamic Therapy
9. Green RW, Enger C: Age-related macular neovascularization associated with macular Study Group; Verteporfin in Photodynamic
degeneration histopathologic studies. The degeneration. Arch Ophthalmol 1988; Therapy Study Group. Photodynamic
1992 Lorenz E. Zimmerman Lecture. 106:1537–1542. therapy of subfoveal choroidal
Ophthalmology 1993; 100:1519–1535. 28. Chandra SR, Gragoudas ES, Friedman E, neovascularization with verteporfin:
10. Gass JDM: Pathogenesis of disciform et al: Natural history of disciform fluorescein angiographic guidelines for
detachment of the neuroepithelium. degeneration of the macula. Am J evaluation and treatment – TAP and VIP
VI: Disciform detachment secondary to Ophthalmol 1974; 78:579–582. report no. 2. Arch Ophthalmol 2003;
heredodegenerative, neoplastic and 29. Gragoudas ES, Chandra SR, Friedman E, 121:1253–1268.
traumatic lesions of the choroid. Am J et al: Disciform degeneration of the macula. 44. Gragoudas ES, Adamis AP, Cunningham
Ophthalmol 1967; 63:689. II: Pathogenesis. Arch Ophthalmol 1976; ET Jr, et al: VEGF Inhibition Study in Ocular
11. Lim JI, Bressler NM, Marsh MJ, Bressler 94:755–757. Neovascularization Clinical Trial Group.
SB: Laser treatment of choroidal 30. Bressler NM, Bressler SB, Fine SL: Age- Pegaptanib for neovascular age-related
neovascularization in patients with angioid related macular degeneration. Surv macular degeneration. N Engl J Med 2004;
streaks. Am J Ophthalmol 1993; Ophthalmol 1988; 32:375–413. 351:2805–2816.
116:414–423. 31. Boldt C, Bressler NM, Fine SL, Bressler SB: 45. Rosenfeld PJ, Brown DM, Heier JS,
12. Heriot WJ, Henkind P, Bellhorn RW, Burns Age-related macular degeneration. Curr MARINA Study Group, et al: Ranibizumab
MS: Choroidal neovascularization can Opin Ophthalmol 1990; 1:247–257. for neovascular age-related macular
digest Bruch’s membrane: A prior break is 32. Moisseiev J, Bressler NM: Asymptomatic degeneration. N Engl J Med 2006;
not essential. Ophthalmology 1984; neovascular membranes in the second eye 355:1419–1431.
91:1603–1608. of patients with visual loss from age-related 46. Brown DM, Kaiser PK, Michels M,
13. Öffler KU, Lee WR: Basal linear deposit in macular degeneration (AMD). Invest ANCHOR Study Groupet al: Ranibizumab
the human macula. Graefes Arch Clin Exp Ophthalmol Vis Sci 1990; 31:462. versus verteporfin for neovascular age-
Ophthalmol 1986; 224:493–501. 33. Fine AM, Elman MJ, Ebert JE, et al: Earliest related macular degeneration. N Engl J
14. Penfold PL, Killingsworth MC, Sarks SH: symptoms caused by neovascular Med 2006; 355:1432–1444.
Senile macular degeneration: The membranes in the macula. Arch 47. Soubrane G, Coscas G, Francais C, Koenig
involvement of immunocompetent cells. Ophthalmol 1986; 104:513–514. F: Occult subretinal new vessels in age-
Graefes Arch Clin Exp Ophthalmol 1985; 34. Nasrallah F, Jalkh AE, Trempe CL, et al: related macular degeneration.
223:69–76. Subretinal hemorrhage in atrophic age- Ophthalmology 1990; 97:649–657.
15. Penfold PL, Provis JM, Billson FA: Age- related macular degeneration. Am J 48. Jalkh AE, Nasrallah FP, Marinelli I, Van de
related macular degeneration: Ophthalmol 1989; 107:38–41. Velde F: Inactive subretinal
ultrastructural studies of the relationship of 35. Small ML, Green WR, Alpar JJ, et al: Senile neovascularization in age-related macular
leucocytes to angiogenesis. Graefes Arch macular degeneration: Clinicopathologic degeneration. Ophthalmology 1990;
Clin Exp Ophthalmol 1987; 225:70–76. correlation of two cases with 97:1614–1619.
16. Ryan SJ: Development of an experimental neovascularization beneath the retinal 49. Schatz H, McDonald HR, Johnson RN:
model of subretinal neovascularization in pigment epithelium. Arch Ophthalmol 1976; Retinal pigment epithelial folds associated
disciform macular degeneration. Trans Am 94:601–607. with retinal pigment epithelial detachments
Ophthalmol Soc 1979; 77:707–745. 36. Doyle WJ, Davidorf FH, Makley TA, Dieruf in macular degeneration. Ophthalmology
17. Ryan SJ: Subretinal neovascularization: WJ: Histopathology of an active lesion of 1990; 97:658–665.
natural history of an experimental model. ocular histoplasmosis. Ophthalmic Forum 50. Grossniklaus HE, Green RW:
Arch Ophthalmol 1982; 100:1804–1809. 1984; 2:106–111. Histopathologic and ultrastructural findings
1949
RETINA AND VITREOUS

of surgically excised choroidal neovascular complicating age-related macular 85. Krummenauer F, Braun M, Dick HB: Clinical
membranes in the submacular surgery degeneration associated with pigment outcome and subjective quality of life after
trials. Arch Ophthalmol 1998; 116:745–749. epithelial detachments. Retina 1988; photodynamic therapy in patients with age-
51. Lopez PF, Lambert HM, Grossniklaus HE, 8:115–121. related macular degeneration. Eur J
Sternberg P Jr: Well-defined subfoveal 71. Melsop KA, Boothroyd DB, Hlatky MA: Ophthalmol 2005; 15:74–80.
choroidal neovascular membranes in age- Quality of life and time trade-off utility 86. Coscas G, Soubrane G: Photocoagulation
related macular degeneration. measures in patients with coronary artery des neovaisseaux sous-retiniens dans la
Ophthalmology 1993; 100:415–422. disease. Am Heart J 2003; 145:36–41. degenerescence maculaire senile par laser
52. Sorenson JA, Yannuzzi LA, Shakin JL: 72. Slakter JS, Stur M: Quality of life in patients a argon. Resultats de l’etude randomisee
Recurrent subretinal neovascularization. with age-related macular degeneration: de 60 cas. Bull Mem Soc Fr Ophthalmol
Ophthalmology 1985; 92:1059–1075. impact of the condition and benefits of 1982; 94:149–154.
53. Bressler NM, Bressler SB, Alexander J, treatment. Surv Ophthalmol 2005; 87. Moorfields Macular Study Group:
et al: Macular Photocoagulation Study 50:263–273. Treatment of senile disciform macular
Reading Center: loculated fluid: A 73. Sharma S, Oliver-Fernandez A: Age-related degeneration: A single-blind randomized
previously undescribed fluorescein macular degeneration and quality of life: trial by argon laser photocoagulation. Br J
angiographic finding in choroidal how to interpret a research paper in health- Ophthalmol 1982; 66:745–753.
neovascularization associated with macular related quality of life. Curr Opin Ophthalmol 88. Macular Photocoagulation Study Group:
degeneration. Arch Ophthalmol 1991; 2004; 15:227–231. Argon laser photocoagulation for senile
109:211–215. 74. Brown GC, Brown MM, Sharma S: macular degeneration: results of a
54. Cantrill HL, Ramsay RC, Knobloch WH: Difference between ophthalmologists’ and randomized clinical trial. Arch Ophthalmol
Rips in the pigment epithelium. Arch patients’ perceptions of quality of life 1982; 100:912–918.
SECTION 10

Ophthalmol 1983; 101:1074–1079. associated with age-related macular 89. Macular Photocoagulation Study Group:
55. Decker WL, Sanborn GE, Ridley M, et al: degeneration. Can J Ophthalmol 2000; Krypton laser photocoagulation for
Retinal pigment epithelial tears. 35:127–133. neovascular lesions of age-related macular
Ophthalmology 1983; 90:507–512. 75. Bass EB, Marsh MJ, Mangione CM, et al: degeneration. Results of a randomized
56. Gass JDM: Pathogenesis of tears of the Submacular Surgery Trials Research Group. clinical trial. Arch Ophthalmol 1990;
retinal pigment epithelium. Br J Ophthalmol Patients’ perceptions of the value of 108:816–824.
1984; 68:514–519. current vision: assessment of preference 90. Macular Photocoagulation Study Group:
57. Green SN, Yarian D: Acute tear of the values among patients with subfoveal Laser photocoagulation for juxtafoveal
retinal pigment epithelium. Retina 1983; choroidal neovascularization – The choroidal neovascularization. Five-year
3:16–20. submacular surgery trials vision preference results from randomized clinical trials. Arch
58. Hoskin A, Bird AC, Sehow K: Tears of value scale: SST report no. 6. Arch Ophthalmol 1994; 112:500–509.
detached retinal pigment epithelium. Br J Ophthalmol 2004; 122:1856–67. 91. Macular Photocoagulation Study Group:
Ophthalmol 1981; 65:417–422. 76. Brown MM, Brown GC, Sharma S, et al: Occult choroidal neovascularization:
59. Yeo JH, Marcus S, Murphy RP: Retinal Utility values associated with blindness in Influence on visual outcome in patients
pigment epithelial tears: patterns and an adult population. Br J Ophthalmol 2001; with age-related macular degeneration.
prognosis. Ophthalmology 1988; 95:8–13. 85:327–331. Arch Ophthalmol 1996; 114:400–412.
60. Bressler NM, Finkelstein D, Sunness JS, et al: 77. Stein JD, Brown GC, Brown MM, et al: The 92. Macular Photocoagulation Study Group:
Retinal pigment epithelial tears through the quality of life of patients with hypertension. Argon laser photocoagulation for
fovea with preservation of good visual acuity. J Clin Hypertens 2002; 4:181–188. neovascular maculopathy after five years.
Arch Ophthalmol 1990; 108:1694–1697. 78. Mangione CM, Lee PP, Pitts J, et al: Results from randomized clinical trials.
61. Marmor MF: Mechanisms of normal retinal Psychometric properties of the National Arch Ophthalmol 1991; 109:1109–1114.
adhesion. In: Ryan SJ, ed. Retina. St. Eye Institute Visual Function Questionnaire 93. Macular Photocoagulation Study Group:
Louis: CV Mosby; 1989:76. (NEI-VFQ). NEI-VFQ Field Test Persistent and recurrent neovascularization
62. Tani PM, Buettner H, Robertson DM: Investigators. Arch Ophthalmol 1998; after krypton laser photocoagulation for
Massive vitreous hemorrhage and senile 116:1496–1504. neovascular lesions of age-related macular
macular choroidal degeneration. Am J 79. Dong LM, Childs AL, Mangione CM, et al: degeneration. Arch Ophthalmol 1990;
Ophthalmol 1980; 90:525–533. Submacular Surgery Trials Research Group. 108:825–831.
63. Kreiger AE, Sterling JH: Vitreous Health- and vision-related quality of life 94. Macular Photocoagulation Study Group:
hemorrhage in senile macular among patients with choroidal Persistent and recurrent neovascularization
degeneration. Retina 1983; 3:318–321. neovascularization secondary to age- after krypton laser photocoagulation for
64. Caswell AG, Kohen D, Bird AC: Retinal related macular degeneration at enrollment neovascular lesions of ocular
pigment epithelial detachments in the in randomized trials of submacular surgery: histoplasmosis. Arch Ophthalmol 1989;
elderly: Classification and outcome. Br J SST report no. 4. Am J Ophthalmol 2004; 107:344–352.
Ophthalmol 1985; 69:397–403. 138:91–108. 95. Macular Photocoagulation Study Group:
65. Bird AC, Marshall J: Retinal pigment 80. Cahill MT, Banks AD, Stinnett SS, Toth CA: The influence of treatment coverage on the
epithelial detachments in the elderly. Trans Vision-related quality of life in patients with visual acuity of eyes treated with krypton
Ophthalmol Soc UK 1986; 105:674–682. bilateral severe age-related macular laser for juxtafoveal choroidal
66. Elman MJ, Fine SL, Murphy RP, et al: The degeneration. Ophthalmology 2005; neovascularization. Arch Ophthalmol 1995;
natural history of serous retinal pigment 112:152–158. 113:190–194.
epithelium detachments in patients with 81. Mangione CM, Gutierrez PR, Lowe G, et al: 96. Smiddy WE, Fine SL, Quigley HA, et al:
age-related macular degeneration. Influence of age-related maculopathy on Comparison of krypton and argon laser
Ophthalmology 1986; 93:224–230. visual functioning and health-related quality photocoagulation in simulated clinical
67. Gass JDM: Serous retinal pigment epithelial of life. Am J Ophthalmol 1999; 128:45–53. treatment of primate retina. Arch
detachment with a notch. A sign of occult 82. Stevenson MR, Hart PM, Montgomery AM, Ophthalmol 1984; 102:1086–1092.
choroidal neovascularization. Retina 1984; et al: Reduced vision in older adults with age 97. Han DP, Folk JC: Internal limiting membrane
4:205–220. related macular degeneration interferes with wrinkling after argon and krypton laser
68. Poliner LS, Olk RJ, Burgess D, Gordon ME: ability to care for self and impairs role as photocoagulation of choroidal
Natural history of retinal pigment epithelial carer. Br J Ophthalmol 2004; 88:1125–1130. neovascularization. Retina 1986; 6:215–219.
detachments in age-related macular 83. Peli E, Goldstein RB, Young GM, et al: 98. Goldberg MF, Herbst RW: Acute
degeneration. Ophthalmology 1986; Image enhancement for the visually complications of argon laser
93:543–550. impaired. Simulations and experimental photocoagulation. Arch Ophthalmol 1973;
69. Teeters VW, Bird AC: The development of results. Invest Ophthalmol Vis Sci 1991; 89:311–318.
neovascularization in senile disciform 32:2337–2350. 99. Macular Photocoagulation Study Group:
macular degeneration. Am J Ophthalmol 84. Rovner BW, Ganguli M: Depression and Argon laser photocoagulation for ocular
1973; 76:1–18. disability associated with impaired vision: histoplasmosis. Results of a randomized
70. Singerman L: Laser photocoagulation for the MoVies Project. J Am Geriatr Soc 1998; clinical trial. Arch Ophthalmol 1983;
1950 choroidal new vessel membrane 46:617–619. 101:1347–1357.
Age-Related Macular Degeneration: Choroidal Neovascularization

100. Macular Photocoagulation Study Group: 114. Eyetech Study Group: Anti-vascular 129. Press release. NEI Statement: NIH
Laser photocoagulation for neovascular endothelial growth factor therapy for Stimulates the development and testing of
lesions nasal to the fovea associated with subfoveal choroidal neovascularization new therapies for advanced AMD.
ocular histoplasmosis or idiopathic causes. secondary to age-related macular Available:
Arch Ophthalmol 1995; 113:56–61. degeneration: phase II study results. www.nei.nih.gov/news/statements/amd_the
101. Macular Photocoagulation Study Group: Ophthalmology 2003; 110:979–986. rapy.asp Nov 7, 2006.
Visual outcome after laser 115. Kabbinavar F, Hurwitz H, Fehrenbacher L, 130. Bennett SR, Folk JC, Blodi CF, Klugman M:
photocoagulation for subfoveal choroidal et al: Phase II, randomized trial comparing Factors prognostic of visual outcome in
neovascularization secondary to age- bevacizumab plus fluorouracil patients with subretinal hemorrhage. Am J
related macular degeneration: The (FU)/leucovorin (LV) with FU/LV alone in Ophthalmol 1990; 109:33–37.
influence of initial lesion size and initial patients with metastatic colorectal cancer. 131. Berrocal MH, Lewis ML, Flynn HW Jr:
visual acuity. Arch Ophthalmol 1994; J Clin Oncol 2003; 21:60–65. Variations in the clinical course of
112:480–488. 116. Trobe JD, Kraft R, Krischer JP: Doctor: submacular hemorrhage. Am J Ophthalmol
102. Jampol LM: Hypertension and visual patient communication in ophthalmic 1996; 122:486–493.
outcome in the Macular Photocoagulation outpatient visits. Ophthalmology 1983; 132. Vander JF, Federman JL, Greven C, et al.
Study. Arch Ophthalmol 1991; 90:51a-55a. Surgical removal of massive subretinal
109:789–790. 117. Taub SJ: The Internet’s role in hemorrhage associated with age-related
103. Bressler NM, Bressler SB: Indocyanine patient/physician interaction: bringing our macular degeneration. Ophthalmology
green angiography: can it help preserve the understanding in line with online reality. 1991; 98:23–27.
vision of our patients? Arch Ophthalmol Compr Ophthalmol Update. 2006; 7:25–30. 133. Lewis H: Intraoperative fibrinolysis of

CHAPTER 146
1996; 114:747–749. 118. Genentech: Press release. Preliminary submacular hemorrhage with tissue
104. Maguire JI, Benson WE, Brown GC: phase III data from head-to-head study of plasminogen activator and surgical
Treatment of foveal pigment epithelial lucentis shows lucentis improved vision drainage. Am J Ophthalmol 1994;
detachments with contiguous extrafoveal compared to visudyne in patients with wet 118:559–568.
choroidal neovascular membranes. Am J age-related macular degeneration. Jan 14, 134. Macular Photocoagulation Study Group:
Ophthalmol 1990; 109:523–529. 2006. Available: Five-year follow-up of fellow eyes of
105. Bressler NM, Bressler SB: Laser treatment in http://www.gene.com/gene/news/press- individuals with ocular histoplasmosis and
macular degeneration and histoplasmosis. releases/display.do?method=detail&id=930 unilateral extrafoveal or juxtafoveal
Ophthalmol Clin North Am 1989; 4:565–581. 7 Jul 25, 2006. choroidal neovascularization. Arch
106. Chamberlin JA, Bressler NM, Bressler SB, 119. Hewson MG, Kindy PJ, Van Kirk J, et al: Ophthalmol 1996; 114:677–688.
et al: The use of fundus photographs and Strategies for managing uncertainty and 135. Bressler NM, Bressler SB: Preventative
fluorescein angiograms in the identification complexity. J Gen Intern Med 1996; ophthalmology: Age-related macular
and treatment of choroidal 11:481–485. degeneration. Ophthalmology 1995;
neovascularization in the Macular 120. Rosenfeld PJ, Heier JS, Hantsbarger G, 102:1206–1211.
Photocoagulation Study. Ophthalmology Shams N: Tolerability and efficacy of 136. Bressler NM, Maguire MG, Murphy PL,
1989; 96:1526–1534. multiple escalating doses of ranibizumab et al: Macular scatter (‘grid’) laser treatment
107. Schachat AP, Chambers WA, Liesegang TJ, (Lucentis) for neovascular age-related of poorly demarcated subfoveal choroidal
Albert DA: Safe and effective. macular degeneration. Ophthalmology neovascularization in age-related macular
Ophthalmology 2003; 110:2073–4. 2006; 113:632.e1. degeneration: results of a randomized pilot
108. Rosenfeld PJ, Moshfeghi AA, Puliafito CA: 121. Charters L: Intravitreal anti-VEGF drugs are trial. Arch Ophthalmol 1996;
Optical coherence tomography findings step forward in AMD management. 114:1456–1464.
after an intravitreal injection of Ophthalmology Times. Dec 15, 2005. 137. Bressler NM: Submacular surgery: Are
bevacizumab (Avastin) for neovascular age- Available: http://www.agingeye.net/ randomized trials necessary? Arch
related macular degeneration. Ophthalmic mainnews/lucentis.php Jul 21, 2006. Ophthalmol 1995; 113:1557–1560.
Surg Lasers Imaging 2005; 36:331–335. 122. American College of Physicians: Primer on 138. Slakter JS, Yannuzzi LA, Sorenson JA, et
109. Shahar J, Avery RL, Heilweil G, et al: 95% CIs for the number needed to treat. al: A pilot study of indocyanine green
Electrophysiologic and retinal penetration Eff Clin Pract 1999. Available: videoangiography-guided laser
studies following intravitreal injection of www.acponline.org/journals/ecp/mayjun99/ photocoagulation of occult choroidal
bevacizumab (Avastin). Retina 2006; primer.pdf Jul 21, 2006. neovascularization in age-related macular
26:262–269. 123. Ebrahimzadeh H, Davalos R, Lee PP: degeneration. Arch Ophthalmol 1994;
110. Avery RL, Pieramici DJ, Rabena MD, et al: Literacy levels of ophthalmic patient 112:465–472.
Intravitreal bevacizumab (Avastin) for education materials. Surv Ophthalmol 139. Guyer DR, Yannuzzi LA, Ladas I, et al:
neovascular age-related macular 1997; 42:152–156. Indocyanine green-guided laser
degeneration. Ophthalmology 2006; 124. Morris NS, MacLean CD, Chew LD, photocoagulation of focal spots at the edge
113:363–372. Littenberg B: The Single Item Literacy of plaques of choroidal neovascularization.
111. Eyetech Pharmaceuticals Inc: Briefing Screener: evaluation of a brief instrument Arch Ophthalmol 1996; 114:693–697.
Document for the Dermatologic and to identify limited reading ability. BMC Fam 140. Hart PM, Chakravarthy U, Mackenzie G,
Ophthalmic Drugs Advisory Committee. Pract 2006; 24;7:21. Archer DB: Telepathy for subfoveal
Pegaptanib sodium injetion in the treatment 125. Spaide RF, Laud K, Fine HF, et al: choroidal neovascularization of age-related
of neovascular age-related macular Intravitreal bevacizumab treatment of macular degeneration: Results of follow up
degeneration. NDA 21.756, Aug 27, 2004. choroidal neovascularization secondary to in a non-randomised study. Br J
Available: http://www.fda.gov/ohrms/ age-related macular degeneration. Retina Ophthalmol 1996; 80:1046–1050.
dockets/ac/04/briefing/2004–4053B1_01_E 2006; 26:383–390. 141. Gragoudas ES, Schmidt-Erfurth U,
yetech-Briefing-Doc.pdf May 9, 2006. 126. Press release. PIER year 1 clinical trial Sickenberg M, et al: Results and
112. Gonzales CR: VEGF Inhibition Study in results. Available: preliminary dosimetry of photodynamic
Ocular Neovascularization (VISION) Clinical http://www.medicalnewstoday.com/medical therapy for choroidal neovascularization in
Trial Group. Enhanced efficacy associated news.php?newsid=44460 Nov 7, 2006. age-related macular degeneration in a
with early treatment of neovascular age- 127. Edwards MG, Schachat AP, Bressler SB, phase I/II study. Invest Ophthalmol Vis Sci
related macular degeneration with Bressler NM: Outcome of cataract 1997; 38–4:S17.
pegaptanib sodium: an exploratory operations performed to permit diagnosis, 142. Schmidt-Erfurth U, Miller JW, Sickenberg
analysis. Retina 2005; 25:815–827. to determine eligibility for laser therapy, or M, et al: Photodynamic therapy for
113. Rosenfeld PJ, Schwartz SD, Blumenkranz toperform laser therapy of retinal disorders. choroidal neovascularization in a phase I/II
MS, et al: Maximum tolerated dose of a Am J Ophthalmol 1994; 118:440–444. study. Preliminary results of multiple
humanized anti-vascular endothelial growth 128. Kwiterovich KA, Maguire MG, Murphy RP, treatments. Invest Ophthalmol Vis Sci
factor antibody fragment for treating et al: Frequency of adverse systemic 1997; 38–4:S17.
neovascular age-related macular reactions after fluorescein angiography.
degeneration. Ophthalmology 2005; Results of a prospective study.
112:1048–1053. Ophthalmology 1991; 98:1139–1142. 1951
CHAPTER

147 Photodynamic Therapy


Deeba Husain, Evangelos S. Gragoudas, and Joan W. Miller

INTRODUCTION MECHANISM OF ACTION


Photodynamic therapy (PDT) involves the use of photo- PDT requires the administration, usually intravenous, of a
activatable compounds (photosensitizers) which accumulate in, photosensitizer dye that accumulates in neoplastic and
and are retained by, proliferating tissues. When these molecules neovascular tissues. The targeted tissue is then irradiated using
are activated by light at the appropriate wavelength, active light of a wavelength at an absorption peak of the dye. A
forms of oxygen and free radicals are generated, which result in photochemical reaction10 is initiated with the absorption of
photochemical damage to cells in which the photosensitizer is light by the photosensitizer molecule in the ground state (S),
present. Thus PDT can be used to treat diseased areas which is excited to enter a higher-energy triplet state (3S). The
selectively while sparing the normal tissue. PDT has been triplet state molecules are short-lived reactive species that
extensively studied in the treatment of neoplasia and more transfer their energy via two pathways that can cause
recently for neovascularization. At this time in ophthalmology, cytotoxicity leading to physiological responses such as necrosis
it is approved by the US FDA and other agencies for the and/or apoptosis. Energy can be transferred from the triplet
treatment of subfoveal choroidal neovascularization, and has state photosensitizer to molecular oxygen converting it to
been used for management of other ocular neovascularization. singlet oxygen (1O2), while returning to their ground state
(type II reaction).11 The triplet state molecules may also directly
HISTORICAL BACKGROUND transfer energy through a free radical mechanism to form
cytotoxic intermediates (type I reaction) (Fig. 147.1). The type
The earliest report on the action of light-activated chemicals on II mediated pathway accounts for the majority of tissue
biological systems was published in 1900 by Raab, who destruction and is responsible for the oxygen dependence of
described the lethal effect of light on paramecium treated with PDT. The extent of oxygen dependence varies somewhat among
acridine dye.1 von Tappeiner in 1904 demonstrated oxygen photosensitizers; under anoxic conditions PDT effects are
dependence of this photosensitization reaction and coined abolished for Photofrin.12
the term ‘photodynamic action’.2 Meyer-Betz demonstrated PDT offers some degree of selectivity in the destruction of
photosensitizing properties of hematoporphyrin derivative tumor and neovascular tissue, with minimal local and systemic
(HPD) in 1913.3 In 1942, Aueler and Banzer for the first time side effects. This is due to two factors: a preferential localization
used HPD for photodynamic destruction of tumors in animals.4 of the photosensitizer in these tissues and precise laser light
Lipson and colleagues in 1961 reported on the use of HPD for irradiation of the targeted tissue.13 The mechanism of accumu-
fluorescence detection of neoplastic tissue, and subsequent lation of the photosensitizer in the neovascular and neoplastic
treatment in patients with breast cancer.5 tissue is not known, but several theories have been proposed.
In 1978, Dougherty et al reported the first large series of Photosensitizers are taken up by most tissues of the body, but
patients with cutaneous malignancies that exhibited partial or are retained longer in neoplastic tissue, as well as in normal
complete response to photoradiation therapy using HPD and liver, spleen, kidney, and wound healing tissue. Henderson et al
light.6 The active components of HPD were identified to be have suggested that pooling and retention of photosensitizers in
dihematoporphyrin ethers and esters (DHE), and the com- tumors could occur due to a larger interstitial space and poor
mercial preparation of DHE is known as porfimer sodium or lymphatic network.14 The cells with high mitotic activity such
Photofrin (PF).7 PDT with benzoporphyrin derivative (BPD) has as tumor and neovascular endothelial cells were found to have
shown beneficial effect in preliminary trials in the treatment of
many diseases such as basal cell carcinoma, metastatic skin
lesions, and the elimination of leukemia progenitor cells in the
marrow of patients with chronic myelogenous leukemia.8 PDT
has been used for nononcological conditions outside the eye
including psoriasis, atherosclerotic plaque and restenosis, bone
marrow purging for the treatment of leukemias with autologous
bone marrow transplant, inactivation of viruses in blood or
FIGURE 147.1. Schematic representation of photodynamic action.
blood products, and several autoimmune conditions including Photons are absorbed by photosensitizer molecules in ground state (S).
rheumatoid arthritis.9 PDT with BPD (Verteporfin) for choroidal This causes them to enter in a higher-energy excited triplet state (3S).
neovascularization (CNV) secondary to macular degeneration The short-lived reactive triplet state photosensitizer molecule leads to
was approved by US FDA in 2000, and since then it is approved transfer of energy causing the formation of singlet oxygen and free
and being used all over the world for this indication. radicals that cause cytotoxicity. 1953
RETINA AND VITREOUS

TABLE 147.1. Photosensitizers in Ophthalmology

Xanthene Derivative
Rose bengal
Tetrapyrrole Derivatives
Hematoporphyrin derivative
Photofrin (dihematoporphyrin ether)
Benzoporphyrin derivative
Chlorins and Bacteriochlorins
Mono-aspartyl chlorin e6
Bacteriochlorin a
Tin etiopurpurin
ATX-S10
SECTION 10

Phthalocyanines
FIGURE 147.2. Absorption spectra of selected photosensitizers that
Chloroaluminum sulfonated phthalocyanine
have been used in PDT. PF, Photofrin; BPD-MA, benzoporphyrin
Zinc phthalocyanine derivative monoacid; Ce6, Chlorin 6; CASPc, chloroaluminum
sulfonated phthalocyanine.
From Lui H. Anderson RR: Photodynamic therapy in dermatology: Recent
developments. Dermatologic Clinics 1993; 11:1–13.

a high expression of low-density lipoprotein (LDL) receptors


(e.g., the apo B/E receptor),15 which might result in more CLINICALLY APPROVED
efficient uptake of LDL-bound molecules by receptor-mediated PHOTOSENSITIZERS
endocytosis.16 Therefore various methods to enhance the LDL-
mediated mechanism have been investigated, including for- The effective penetration depth of the PDT treatment is dependent
mulation of photosensitizer in liposomes, lipid-based on the wavelength of the light and optical properties such as
emulsions, as well as preincorporation with LDL. Other absorption and scatter of the targeted tissue. Typically, the effective
techniques used are delivery with biodegradable nanospheres, penetration depth is 2–3 mm at 630 nm and increases to 5–6 mm
attaching photosensitizers covalently to polymers, and using at longer wavelengths (700–800 nm).25 These values can be altered
inclusion complexes such as cyclodextrins. Selective local- by changing the biological and physical characteristics of the
ization of photosensitizers in tumors has also been improved by photosensitizer (Table 147.1). In general, photosensitizers with
binding the dye to targeting molecules such as peptides or longer wavelengths and higher molar absorption at these wave-
monoclonal antibodies (Mab) that recognize specific antigens lengths are more efficient photodynamic agents (Fig. 147.2).10
on tumor cells.17 Targeted PDT is a new modality to improve Most clinical experience comes from the porphyrin family
the efficacy of PDT; here photosensitizer is bound to the of photosensitizers, including hematoporphyrin derivative
targeting peptide which in turn binds to VEGF receptors on (HPD) and dihematoporphyrin ether (DHE). DHE is a complex
the neovascularization thus resulting in efficient localization of mixture of oligomeric esters and ethers of HPD, with inherent
the photosensitizer.18 variability (Fig. 147.3 ).26 The commercial preparation of DHE,
The mechanisms of PDT-mediated tissue destruction called Photofrin (Fig. 147.3a), has been used in clinical trials for
include cellular, vascular, and immunological damage. Direct bladder, lung, stomach, and cervix cancer. The main problems
cellular destruction is due to the damage of cellular membranes with Photofrin were prolonged skin photosensitivity and
through lipid peroxidation and protein damage, by singlet relatively low absorption in the wavelength region of
oxygen and free radical intermediates,7 leading to structural and therapeutic interest (600–1100 nm).10 This has provided an
functional damage to cellular membranes. Apoptosis of the cells incentive to develop second-generation photosensitizers.
has also been reported by a biomolecular cascade induced by The clinically tested second-generation photosensitizers
PDT-related damage to mitochondrion, lysosomes, and nuclear include Verteporfin (BPD-MA) (Fig. 147.3b), mono-L-aspartyl
components.19 Vascular damage is an important mechanism of chlorin e6 (NPe6) (Fig. 147.3c), Hypericin, protoporphyrin IX,
tissue destruction in PDT with certain photosensitizers, and Foscan (mTHPC) (Fig. 147.3d), Purlytin (SnET2) (Fig. 147.3e),
probably occurs secondary to endothelial damage and 5-amino-levulinic acid, ALA (Fig. 147.3f), and Lu-Tex. BPD, a
subsequent platelet aggregation, and vessel thrombosis. second-generation photosensitizer, is formally a chlorin, since it
Eiconasoids are released following PDT including has a reduced pyrrole ring as well as a fused six-membered iso-
thromboxane, histamine, and tumor necrosis factor-a and may cyclic ring (Fig. 147.3b).27 BPD has absorption maxima at 354,
contribute to vascular occlusion.20 Finally, immunomodulation 418, 574, 626, and 688 nm; this allows deeper tissue penetra-
may play a role in PDT-induced tissue destruction. Treatment tion and effective treatment. BPD has a rapid rate of clearance,
of various tumors with PDT has demonstrated increased tissue with no significant photosensitivity after 24 h, and is meta-
levels of cytokines,21 enhanced killing activity of specific bolized to inactive forms prior to excretion through the feces.28
cytotoxic T-lymphocytes,22 increased natural killer cell
activity,23 and tumor-associated macrophage accumulation in PDT OF CHOROIDAL
tissue24 with TNF-a release, and macrophage- mediated NEOVASCULARIZATION
cytotoxicity. Immunomodulation following PDT is being inves-
tigated for application in organ transplantation and the PDT is a potentially selective treatment modality for
1954 treatment of autoimmune diseases. neovascularization. It offers a treatment for CNV, where the
Photodynamic Therapy

FIGURE 147.3. Chemical structures of


(a) dihematoporphyrin ether, DHE;
(b) benzoporphyrin derivative monoacid,
BPD-MA; (c) mono-aspartyl chlorin e6, NP e6;
(d) meta-tetra (hydroxyphenyl) chlorin, mTHPC;
(e) tin etiopurpurin, SnET2; (f) 5-amino-levulinic
acid, ALA.

CHAPTER 147
d

b
e

neovascularization can be occluded and the overlying neuro- dye dose was 27 times higher than the dose which has been
sensory retina is spared avoiding the sudden decrease of vision demonstrated to be safe clinically.
seen as a complication of thermal photocoagulation of subfoveal Preliminary work has been carried out with tin etiopurpurins
CNV. Several investigators have studied the efficacy of various (SnET2).33 The treatment parameters include a dye dose of
photosensitizers for PDT of experimental CNV. 1mg/kg with a light of 665 nm, an irradiance of 600 mW/cm2 and
Experimental CNV was created in a monkey model by fluence of 35–70 J/cm2. A spot size of 1200 mm was used and light
inducing argon laser injury to the macula, which leads to devel- exposure lasted 60–120 s. It has been shown to be effective in
opment of CNV in 2–4 weeks (Fig. 147.4a–c).29 Thomas and closure of experimental CNV. Clinical trials were conducted to
Langhofer30 in 1987 demonstrated successful thrombosis of study the role of SnET2 (Purlytin) in the treatment of wet age
experimental CNV without occlusion of the choriocapillaris, in related macular degeneration (AMD). The pooled data at 2 years
the monkey model using DHE. Kliman et al31 studied PDT of showed that 58% of SnET2-treated patients lost 15 letters or less
experimental CNV using chloroaluminum sulfonated compared to 42% of placebo patients. This effect was only seen in
phthalocyanine (CASPc). These preliminary studies suggest lesions smaller than 3 mm and with better visual acuity (46–67
that closure of experimental CNV can be accomplished over a letters).34 The systemic photosensitivity lasted 4 weeks. The
wide range of treatment parameters, but the selectivity of effect, drug failed to reach its efficacy end point in Phase III studies
and the damage to retinal structures and human safety was and at this time is not used in the treatment of AMD.
not investigated. Miller and Miller32 studied the efficacy of Lu-Tex or lutetium texaphyrin is a water-soluble photo-
rose bengal, a xanthene derivative to treat experimental CNV. It sensitizer that has an absorption peak of 732 nm. It has been
was found that PDT with rose bengal led to disruption of CNV shown to localize in choroidal neovascularization in the
but did not destroy the new vessels completely, and the required monkey eye. PDT using 1–2 mg/kg body weight of Lu-Tex, and 1955
RETINA AND VITREOUS

a b c

FIGURE 147.4 (a). Fundus photograph showing elevated yellow-gray areas (arrows) of choroidal neovascularization in the experimental model of
choroidal neovascularization in the monkey eye. (b) Early phase fluorescein angiogram of the eye in (a) showing the typical lacy pattern of
hyperfluorescence in areas of experimental CNV (arrow). (c) Late phase angiogram of the eye in (b) showing increasing hyperfluorescence and
fluorescein leakage by the CNV (arrows).
(a–c) From Husain D, Miller JW, Michaud N, et al: Intravenous infusion of liposomal benzoporphyrin derivative for photodynamic therapy of experimental choroidal
neovascularization. Arch Ophthalmol 1996; 114:978–985.
SECTION 10

a light dose of 50–100 J/cm2 has been shown to cause closure of high density of lipoprotein receptors which may enhance
CNV in the monkey eye.35 Phase I and II clinical trials with preferential uptake of the photosensitizer.44 BPD fundus
Lu-Tex showed that the drug was effective in causing CNV angiography has also demonstrated that liposomal BPD
occlusion, but the drug caused peripheral parasthesias in a accumulates in CNV.45 Dye doses of 0.25–1 mg/kg of liposomal
significant number of patients and therefore the clinical trials BPD were studied and a minimal dose of 0.375 mg/kg caused
were halted. effective closure of CNV. The optimal time for irradiation
Normal choroidal vasculature has been occluded using newer appeared to be 20–50 min after a bolus intravenous injection
photosensitizers and it has been extrapolated that these dyes of dye with a spot size of 1250 mm at an irradiance of
can potentially be used to treat CNV. Mori et al36 have 600 mW/cm2 and a fluence of 150 J/cm2.43
demonstrated effective and selective occlusion of choroidal Since damage to normal retina and choroidal structures was
vessels in Japanese monkey eyes using 2–10 mg/kg of mono-l- difficult to assess in the experimental CNV model, selectivity
aspartyl chlorin e6 (NP e6) and 664 nm light (0.4–7.5 J/cm2). of PDT was assessed in normal retina and choroid using the
Obana et al37 performed PDT on the retina and choroid of same parameters as for CNV.43 Fluorescein angiography demon-
monkeys using ATX-S10, a new chlorin derivative (dose of strated early hypofluorescence in the treated areas followed by
8–12 mg/kg) using a light of 670 nm (3.5 J/cm2), 30–74 min hyperfluorescence starting at the periphery of the lesion in the
after the dye injection. Histopathology of these eyes demon- later frames of the angiogram. A grading system for assessing
strated photothrombosis of choriocapillaris and choroidal damage to retina and choroid was devised. Damage to RPE and
vessels with no obvious damage to the neurosensory retina. choriocapillaris was seen in all irradiated eyes, and the grading
scheme was based on the damage to the neurosensory retina
and medium to large choroidal vessels. Some damage to the
VERTEPORFIN PDT FOR CHOROIDAL photoreceptors was noted throughout all grades, typically mild
NEOVASCULARIZATION vacuolization and disorientation of the inner and outer
Preclinical Studies segments of the photoreceptors. When PDT was performed
Work in our laboratory has used a tetrapyrrol derivative with liposomal BPD doses of 0.25, 0.375, or 0.5 mg/kg, at
benzoporphyrin derivative monoacid (BPD, Verteporfin, Visudyne). 20–50 min after dye injection, the retinal structure was well
This photosensitizer has a long absorption wavelength at preserved and PDT was sufficiently selective.
690 nm,38 therefore allowing deeper tissue penetration for effective Dosimetry experiments were done using a bolus intravenous
treatment, through blood, fluid, and fibrosis that is important in injection of liposomal BPD-MA, but liposomal drugs are typically
the treatment of CNV. When injected intravenously, BPD has a used clinically as an intravenous infusion, and clinical trials in
serum half-life of 30 min. The high-est tissue levels are reached in dermatology using liposomal BPD administered the dye by infu-
3 h, declining rapidly and cleared in the first 24 h, therefore sion with success. Further studies were undertaken and designed to
reducing the risk of the systemic photosensitivity.39 It has been evaluate the efficacy and parameters for PDT of experimental CNV
found to be safe for human use, and was first studied in clinical using an intravenous infusion of liposomal BPD.46 Infusion
trials in dermatology for malignant skin tumors.40 (10 min) was tested and effective closure was achieved when
Initial studies in experimental CNV were done using a lipo- irradiation was performed 20–30 min from the start of infusion
protein-associated preparation of BPD at doses of 1–2 mg/kg using a light of spot size of 1250–3000 mm of 689 nm light at
and light of 692 nm generated from an argon/dye laser that 600 mW/cm2 and 150 J/cm2. Closure of experimental CNV could
was delivered via a slit-lamp delivery system with a spot be seen by fluorescein angiography (Fig. 147.5a–c) and was con-
size of 1250 mm. Effective vascular occlusion was seen firmed by histopathology. Light microscopy showed occluded vessels
when irradiation was performed 1–80 min after the dye injec- in the area of CNV, closure of the underlying choriocapillaris, and
tion, with a fluence of 50–150 J/cm2 and irradiance of the overlying retina was essentially normal. Electron microscopy of
150–600 mW/cm2.41 No thermal damage was seen at the vessels in the CNV showed occlusion of the lumen with red
irradiances as high as 1800 mW/cm2.42 blood cells, white blood cells, and platelets, with damaged
Further refinement of dosimetry was carried out using the endothelial cells showing swelling and vacuolation of the
liposomal preparation of BPD.43 The mechanism of photo- cytoplasm, with breaks in the nuclear membrane (Table 147.2).46
sensitizer uptake by the neovascular tissue is not known, but it Selectivity of PDT using liposomal BPD was studied by
1956 has been observed that neovascular and neoplastic tissue have performing light irradiation of normal monkey retina and
Photodynamic Therapy

a b c

FIGURE 147.5. (a) Fundus photograph 24 hours after PDT with irradiation performed at the end of slow dye infusion to the foveal choroidal
neovascularization (arrow) and at the end of flush infusion to the choroidal neovascularization temporal to the fovea (arrowhead), showing mild
graying of the two treated areas. (b) Early phase fluorescein angiogram 24 h after PDT using liposomal BPD at the end of slow dye infusion
(arrow) and at the end of flush (arrowhead), illustrates hypofluorescence (occlusion of CNV) in both areas with perfusion of the overlying normal
retinal capillaries confirming occlusion of CNV. (c) Late phase angiogram of the eye in Figure 147.4, showing staining at the edge of the

CHAPTER 147
irradiated area. This most likely represents leakage from perfused choriocapillaris through damaged RPE.
(a–c) From Husain D, Miller JW, Michaud N, et al: Intravenous infusion of liposomal benzoporphyrin derivative for photodynamic therapy of experimental choroidal
neovascularization. Arch Ophthalmol 1996; 114:978–985.

with optimal parameters, with histopathology showing a


TABLE 147.2. Standard Clinical Treatment Parameters for
Visudyne PDT
fibrous scar enclosed by proliferating RPE cells with few
open capillaries. In normal eyes treated with CNV, the
1. Dye dose = 6 mg/m2 body surface area choriocapillaris was re-perfused at 4 weeks. The RPE showed
2. Intravenous infusion over 10 min presence of irregular pigmentation with liposomes, phago-
somes, and melanosomes indicating some recovery of function.
3. Treatment at 15 min after start of dye infusion A layer of pigmented macrophages was seen over the RPE.
4. Laser light wavelength of at 689 nm, irradiance of 600 mW/cm2 There was some mild disorganization of the photoreceptors.
and fluence of 100 J/cm2 The rest of the neurosensory retina appeared to be normal.
Therefore the damage to normal retina and retinal pigment
epithelium caused by PDT at 24 h showed histological recovery
choroid with the same parameters. Fundus photography was at 4 weeks.
taken 24 h after PDT and it demonstrated mild graying of the Long-term recurrence or persistence of CNV after PDT
retina at the treated areas (Fig. 147.6a). Fluorescein angiography may necessitate repeat treatments, so further studies were
of these areas showed early hypofluorescence (Fig. 147.6b) with carried out to evaluate the effect of repeat treatments on
late staining starting from the edges of the lesion (Fig. 147.6c). normal monkey retina and choroid.48 Three consecutive
Light and electron microscopy of the lesions showed occlusion treatments were placed in the monkey eye, using different
of the choriocapillaris and damage to the retinal pigment doses of liposomal BPD doses (6, 12, 18 mg/m2) but constant
epithelium in all cases. Most eyes showed preservation of the light dose (689 nm; 600 mW/cm2; 100 J/cm2). Treatments
medium and larger choroidal vessels, some disarray and were separated by an interval of 2 weeks, with histopathologic
vacuolation of the outer segments with mild pyknosis in the examination at 2 and 6 weeks after the third treatment.
outer nuclear layer and normal inner retina. Minimal damage to the retina, choroid, and optic nerve was
Studies were carried out to study the long-term effect present in animals treated at 6 mg/m2. Higher dye doses lead
(4 weeks follow-up) of BPD on the treated CNV.47 Persistent to significant cumulative damage to the normal retina,
closure of CNV was seen in most eyes that had been treated choroid, and optic nerve.

a b c

FIGURE 147.6. (a) Fundus photograph 24 hours after PDT of normal retina and choroid, showing mild graying of the areas irradiated at the end
of dye infusion (small arrow), at the end of flush (long arrow), 10 minutes after end of flush infusion (hollow arrow), and 20 minutes after the end
of flush (arrowhead). (b) Early phase fluorescein angiogram of an eye in (a), taken 24 hours after PDT of the normal retina and choroid, showing
hypofluorescence of the irradiated areas. (c) Late phase angiogram of eye in (b) showing staining of the irradiated area starting from the
periphery of the lesion. This staining is most likely occurring as described in Figure 147.4.
(a–c) From Husain D, Miller JW, Michaud N, et al: Intravenous infusion of liposomal benzoporphyrin derivative for photodynamic therapy of experimental choroidal
neovascularization. Arch Ophthalmol 1996; 114:978–985.
1957
RETINA AND VITREOUS

Clinical Trials
TABLE 147.3. The Landmark Clinical Trials of PDT with
Phase I/II clinical trials were designed to evaluate safety and
Verteporfin for CNV
maximum tolerated doses of verteporfin therapy for the
treatment of patients with classic CNV. Sixty-one patients were 1. The TAP (treatment of age-related macular degeneration with
reported in a dose escalation protocol.49,50 Dye doses were 6 or photodynamic therapy) trial established the efficacy of PDT
12 mg/m2 of liposomal BPD with light of 689 nm at a fluence of for classic subfoveal neovascularization in AMD at 2 years
follow-up
50–150 J/cm2 and an irradiance of 600 mW/cm2. Eligibility
criterion included classic CNV of less than or equal to nine 2. The VIP (Verteporfin in photodynamic therapy) study
macular photocoagulation study (MPS) disc area and refracted concentrated on subfoveal occult-only lesions not included in
vision of 20/40 or worse. In a report on single treatments, the the TAP study. After 2 years, treated eyes were less likely to
experience visual loss. Exploratory analyses of TAP and VIP
results showed that light-activated verteporfin could cause suggest that lesion size is a more significant predictor of the
short-term (1–4 weeks) cessation of fluorescein leakage from treatment benefit than either lesion composition or visual activity
CNV without angiographic damage to retinal blood vessels or
loss of vision.51 At 12 weeks follow-up, leakage occurred in most 3. The VIM (Visudyne in minimally classic) trial altered the standard
PDT light fluence rate in the treatment of subfoveal minimally
cases, defined as extension of the CNV beyond the original classic lesions. This trial again demonstrated a beneficial effect
borders. Systemic safety of verteporfin was very good in this for those receiving treatment with PDT
Phase I/II investigation (Table 147.3). Specifically, the most
frequent adverse event of this open label study was headache 4. Early retreatment (6 weeks) did not improve outcomes in the
SECTION 10

Verteporfin in early re-treatment (VER) trial


(4.7%). Adverse ocular side effects were noted only at the
highest light doses with a significant loss of vision in three 5. The VIO (Visudyne in occult) trial, evaluating PDT in occult-only
patients. Side effects attributed to PDT included retinal lesions as a confirmatory study of the VIP trial, did not achieve
its primary endpoint at 2 years
arteriolar, venular, and capillary occlusion. Side effects which
could be attributed to the disease process itself or to PDT
included vitreous hemorrhage. The results from the Phase I/II
studies suggested that a potentially effective dose of verteporfin Few ocular systemic adverse events judged to be of any
to be considered for Phase III trials would be 6 mg/m2 body clinical relevance were noted in Phase III of the study.
surface area, which would be infused intravenously over 10 min. Compared to placebo patients, verteporfin-treated patients
Irradiation with a diode laser at 689 nm was applied 15 min had more transient vision disturbances (18% vs 12%), injection
after the start of the infusion; the light dose delivered was site adverse events (13% vs 3%), transient photosensitivity
between 50 and 100 J/cm2 at an intensity of 600 mW/cm2 over reactions (3% vs 0%), and infusion-related low back pain (2%
a period of 83 s. vs 0%). However, most of these events were mild to moderate
A Phase III clinical trial was initiated in December 1996, and resolved by themselves. In summary, the TAP52 inves-
termed as the treatment of age-related macular degeneration tigation showed that verteporfin therapy reduced the risk of at
with photodynamic therapy (TAP). This was a randomized, least moderate vision loss compared to placebo for at least
placebo-controlled, double-masked, multicentric study (19 cen- 12 months in patients with predominantly classic CNV who
ters in North America and Europe) designed to determine the presented with subfoveal lesions. In the TAP investigation, the
effect of PDT on vision in patients with subfoveal CNV visual acuity results were complemented by similar outcomes
secondary to AMD. Eligibility criteria included subfoveal new or for contrast sensitivity evaluations.
recurrent CNV secondary to AMD with a classic component, The 2-year follow-up report of the TAP investigation
best corrected visual acuity of 20/40 to 20/200, and lesion size showed53 that the beneficial outcomes with respect to visual
of 5400 mm or less. Patients underwent re-treatment every acuity and contrast sensitivity noted at the month 12 exam-
3 months if angiographic leakage was apparent on follow-up. ination in verteporfin-treated patients were sustained through
The proportion of eyes with at least moderate vision loss (a the month 24 examination. At the month 24 examination for
decrease in the letter score of 15 letters or ~3 lines) was greater the primary outcome, 213 (53%) of 402 verteporfin-treated
in placebo-treated eyes than in verteporfin-treated eyes patients compared to 78 (38%) of 207 placebo-treated patients
throughout the period from examination at month 3 to the lost fewer than 15 letters. In subgroup analyses for predo-
examination at month 12. In subgroup analysis, the visual minantly classic lesions at baseline, 94 (59%) of 159 verte-
acuity benefit of verteporfin therapy was clearly demonstrated porfin-treated patients compared to 26 (31%) of 83 placebo-
in eyes with classic CNV occupying at least 50% of the area treated patients lost fewer than 15 letters at the month 24
of the entire lesion (termed ‘predominantly classic’ lesions). examination. For minimally classic lesions at baseline, no
With respect to predominantly classic lesions, 33% of the statistically significant differences in visual acuity were noted.
verteporfin-treated eyes had at least moderate vision loss at Few additional photosensitivity adverse reactions and injection
12 months compared to 60% of the placebo-treated eyes. site adverse events were associated with verteporfin therapy in
Furthermore, at this time, 12% of verteporfin-treated eyes and the second year of follow-up. Therefore, this study concluded
33% of placebo-treated eyes had severe vision loss. The results that the visual acuity benefits of Verteporfin therapy for AMD
were even greater for predominantly classic lesions and no patients with predominantly classic CNV subfoveal lesions
occult CNV; 23% of verteporfin-treated eyes had at least are safely sustained for 2 years, providing more compelling
moderate vision loss at 12 months compared to 73% of placebo- evidence to use Verteporfin therapy for these cases. In TAP
treated eyes. Ten percent of the verteporfin-treated eyes had Report No. 5, the 36-month vision and safety results of an
severe vision loss compared to 41% of the placebo-treated eyes. open-label extension of the TAP investigation showed that the
These results prompted the TAP Study Group to recommend visual outcomes for patients treated with Verteporfin with
verteporfin therapy for all predominantly classic lesions that predominantly classic lesions at baseline remained stable from
met the eligibility criteria for the studies. Progression of classic month 24 through month 36, and no additional safety
CNV beyond the area of the lesion identified at baseline concerns.54
occurred in only 166 (46%) of 361 verteporfin-treated eyes The role of PDT in the treatment of occult with no classic
compared to 133 (71%) of 187 placebo-treated eyes by the subfoveal choroidal neovascularization was studied in 339
1958 month 12 examination. patients in a multicentric, placebo-controlled, double-masked
Photodynamic Therapy

study (Verteporfin in photodynamic therapy or VIP study) enrolled participants (85%). At the 24 month examination,
carried over 2 years.55 It showed that there was a significant median improvement from baseline in visual acuity of the
reduction in the risk of moderate to severe visual loss. The 22 patients evaluated was six letters; median contrast sensi-
study also suggested that greater benefit was seen in patients tivity improved by 3.5 letters. At the 24 month examination,
with smaller lesion (4 disc area or smaller) and lower levels 10 patients (45%) gained seven or more letters of visual acuity
of visual acuity (20/50 or less). The evidence for efficacy of PDT from baseline, whereas four patients (18%) lost eight or more
in treating the occult-only subtype has weakened with the letters, including two patients (9%) who lost at least 15 letters.
recently completed Visudyne in occult (VIO) trial, in which the There was absence of fluorescein angiographic leakage from
study’s primary visual outcome was not reached at either 1 or classic CNV in 17 of the 20 evaluable lesions (85%), and leak-
2 years. age from occult CNV was absent in all eyes. No serious ocular
A recent clinical trial was designed to compare the treatment adverse events were reported, and no serious systemic event was
effect and safety of PDT with verteporfin using a standard (SF) considered to be associated with treatment. The study59 con-
or reduced (RF) light fluence rate with that of placebo therapy in cluded that the median visual acuity improved and fluorescein
patients with subfoveal minimally classic choroidal neovas- angiographic leakage decreased after verteporfin therapy in this
cularization (CNV) with AMD (Visudyne in minimally classic small, uncontrolled clinical study of patients with subfoveal
or VIM trial). This was a multicentric, double-masked, placebo- CNV resulting from OHS. Verteporfin therapy seemed to be
controlled, randomized clinical trial. Inclusion criteria included relatively safe in these patients.
initial best-corrected visual acuity of at least 20/250 and a

CHAPTER 147
lesion size of no greater than six MPS disc areas. One hundred COMBINATION THERAPY
and seventeen patients were randomly assigned patients (1:1:1)
to verteporfin infusion (6 mg/m2) and light application with an Preliminary study of PDT in combination with intravitreal
RF rate (300 mW/cm2) for 83 s (light dose of 25 J/cm2) or an SF kenalog injection has been reported in CNV secondary to AMD
rate (600 mW/cm2) for 83 s (light dose of 50 J/cm2) or to placebo and was found to be relatively safe and had reasonable results
infusion with RF or SF. Treatment was repeated every 3 months for lesions with some classic component.60 Subsequently, a case
if the treating physician noted fluorescein leakage from CNV on series61 was reported on 26 patients with CNV secondary to
angiography. At month 12, a loss of at least 3 lines of visual AMD treated with PDT immediately followed by intravitreal
acuity occurred in 5 (14%) of 36 eyes assigned to RF and 10 kenalog. Elevated IOP seemed to be the most frequent early side
(28%) of 36 eyes assigned to SF, compared with 18 (47%) of 38 effect of the treatment. Although the number of patients in this
eyes assigned to placebo. At month 24, this loss occurred in 9 pilot study was limited, the improvement of acuity and the
(26%) of 34 eyes assigned to RF and 17 (53%) of 32 assigned to reduced treatment frequency in these patients suggested that
SF, compared with 23 (62%) of 37 eyes assigned to placebo. combination therapy with PDT and intravitreal triamcinolone
Progression to predominantly classic CNV by 24 months was acetonide, particularly when used as first-line therapy, merits
more common in the placebo group 11 (28%) of 39 patients further investigation. The role of the combination of PDT
compared with 2 (5%) of 38 in the RF group and 1 (3%) of 37 in and intravitreal kenalog for nonsubfoveal choroidal neo-
the SF group. No unexpected ocular or systemic adverse events vascularization was studied in another case series of 15 patients
were identified. The study concluded that Verteporfin therapy followed over 12 months.62 This pilot study showed that
safely reduced the risks of losing at least 15 letters (≥3 lines) of the visual acuity response and the low incidence of subfoveal
visual acuity, and, progression to predominantly classic CNV extension suggest that PDT combined with intravitreal
for at least 2 years in individuals with subfoveal minimally triamcinolone for the treatment of nonsubfoveal choroidal
classic lesions due to AMD measuring six MPS disc areas or neovascularization merits further investigation as a first-line
less. Based on the overall evidence available on Verteporfin treatment. Now a clinical trial is underway to study the
therapy for these lesions, the VIM Study Group recommended combined Visudyne therapy with kenalog in CNV secondary
verteporfin therapy for relatively small minimally classic to AMD.
lesions.56 Clinical trials are also ongoing to evaluate the safety and
Preliminary study57 of the treatment of CNV not related to efficacy of combination treatment of PDT and pegaptanib
AMD such as myopia, histoplasmosis, angioid streaks, and sodium (Macugen) for predominantly classic choroidal
idiopathic causes, with Verteporfin therapy, achieved short-term neovascularization secondary to AMD. Preliminary preclinical
cessation of fluorescein leakage from CNV in a small number data indicate that an intravitreal ranibizumab (Lucentis)
of patients without loss of vision. Verteporfin therapy for injection in combination with verteporfin PDT causes a greater
subfoveal CNV caused by pathologic myopia was studied in a reduction in angiographic leakage than PDT and intravitreal
multicentric, double-masked, placebo-controlled, randomized vehicle injection (PDT only) in experimental choroidal
clinical trial in 110 patients through 2 years of follow-up.58 It neovascularization in the monkey eye.63 Clinical trials
showed that PDT maintained a visual benefit compared with (FOCUS) are ongoing to compare Lucentis in combination with
a placebo therapy. Although the primary outcome was not PDT to PDT alone.64
statistically significantly in favor of verteporfin therapy at Targeted PDT is a new modality to improve the efficacy of
2 years as it had been at 1 year of follow-up, the distribution PDT. Preclinical work has been done with Verteporfin bound to
of change in visual acuity at the month 24 examination was the targeting peptide, ATWLPPR, which helps it bind to VEGF-
in favor of the verteporfin-treated group and showed that this receptor 2. The targeted verteporfin resulted in more selective
group was more likely to have improved visual acuity through treatment than the control conjugate or standard verteporfin.
the month 24 examination. The VIP Study Group recom- This study provides the basis for further studies of targeted
mended verteporfin therapy for subfoveal CNV resulting from PDT strategies and subsequent clinical trials.18
pathologic myopia based on both the 1 and 2 year results of this
randomized clinical trial. CONCLUSION
PDT of subfoveal choroidal neovascularization with
verteporfin in the ocular histoplasmosis syndrome was studied PDT causes selective damage to the proliferating tissues with
in an uncontrolled, prospective case series of 26 patients. relative sparing of the surrounding tissues. This led to its vital
A 24 month examination was completed in 22 of the 26 role in the treatment of ocular neovascularization specially 1959
RETINA AND VITREOUS

CNV. PDT of choroidal neovascularization using verteporfin conditions. Further research is ongoing to determine the
(Visudyne) is a proven treatment modality for certain eyes with suitable photosensitizer and the optimal treatment parameters
subfoveal choroidal neovascularization secondary to age-related for the ocular applications of PDT. New photosensitizers that
macular degeneration and other causes such as myopia, may have improved characteristics for PDT of ocular structures
histoplasmosis, angioid streaks, and idiopathic causes. Clinical continue to be developed. Future strategies will address
trials have shown that it is a safe treatment with minimal improved selectivity by complexing photosensitizers to agents
chance of transient systemic side effects. In the experimental that target tumors and neovasculature. Finally, PDT has a
models, PDT has also been efficacious in the treatment of iris, major role in a multifaceted approach such as in combination
corneal neovascularization, ocular tumors, and glaucoma and it with antiangiogenic and immunogenic modalities.
offers an adjunctive therapy to the existing modalities for these

REFERENCES
1. Raab O: Ueber die Wirkung 16. Kessel D: Porphyrin–lipoprotein association 29. Ohkuma H, Ryan SJ: Experimental
Fluorescierenden Stoffe Auf Infusorien. as a factor in porphyrin localization. Cancer subretinal neovascularization in the monkey.
Z Biol 1900; 39:524–546. Lett 1986; 33:183–188. Arch Ophthalmol 1982; 101:1102–1110.
2. Von Tappeiner H, Jodlbauer A: Ueber 17. Jiang FN, Allison B, Lui D, et al: Enhanced 30. Thomas EL, Langhofer M: Closure of
wirking der photodynamischen photodynamic killing of target cells by either experimental subretinal neovascular vessels
(fluorescierenden) Stoffe auf Protozoan und monoclonal antibody or low density with dihematoporphyrin ether augmented
SECTION 10

Enzyme. Dtsch Arch Klin Med 1904; lipoprotein mediated delivery systems. argon green laser photocoagulation.
80:427–487. J Control Release 1992; 19:41–58. Photochem Photobiol 1987; 46:5881–5886.
3. Meyer-Betz F: Untersuchungen uber die 18. Renno RZ, Terada Y, Haddadin MJ, et al: 31. Kliman, Puliafito CA, Stern D, et al:
Biologische (photodynamische) Wirkung Selective photodynamic therapy by Phthalocyanine photodynamic the new
des hamatoporphyrins und anderer targeted verteporfin delivery to experimental strategy for closure of choroidal
Derivative des Blut-und-Gallenfarb-stoffs. choroidal neovascularization mediated by a neovascularization. Laser Surg Med
Dtsch Arch Klin Med 1913; 112:476–503. homing peptide to vascular endothelial 1994; 15:2–10.
4. Aueler H, Banzer G: Untersuchungen uber growth factor receptor-2. Arch Ophthalmol 32. Miller H, Miller B. Photodynamic therapy
die rolle der Porphyrine bei 2004; 122:1002–1011. of subretinal neovascularization in the
geschwulstkranken Menschen und Tieren. 19. Agarwal ML, Larkin HE, Zaidi SIA, et al: monkey eye. Arch Ophthalmol 1993;
Krebsforsch 1942; 53:65–68. Phospholipase activation triggers 111:855–860.
5. Lipson RL, Baldes EJ, Olsen EM: apoptosis in photosensititzed mouse 33. Baumal CR, Puliafito CA, Pieroth L, et al:
Hematoporphyrin derivative for detection lymphoma cells. Cancer Res 1993; Photodynamic therapy (PDT) of
and management of cancer. Proc IX internat 53:5897–5902. experimental choroidal neovascularization
Cancer Congr. 1966:393. 20. Fingar VH, Weiman TJ: Studies on the with tin ethyl etiopurpurin. Invest
6. Dougherty TJ, Kaufman JE, Goldfarb A, mechanism of photodynamic therapy Ophthalmol Vis Sci 1996; 37:S122.
et al: Photoradiation therapy for the induced tumor destruction. In: Proceedings 34. Thomas EL, Rosen R, Murphy R, et al:
treatment of malignant tumors. Cancer Res of the SPIC conference, ‘Photodynamic Purlytin (SnET2)-photodynamic therapy
1978; 38:2628–2635. therapy Mechanisms II’; 1990:168–177. produces closure of subfoveal choroidal
7. Lui H, Anderson RR: Photodynamic therapy 21. Nseyo UO, Whalen RK, Duncan MR, et al: neovascularization in humans. Invest
in dermatology: Recent developments. Urinary cytokines following photodynamic Ophthalmol Vis Sci 1998; 39:S242.
Dermatol Clin 1993; 11:1–13. therapy for bladder cancer: preliminary 35. Graham KB, Arbour JD, Connoly EJ et al:
8. Levy JG: Photosensitizers in photodynamic report. Urology 1990; 36:167–171. Digital angiography using Lutetium
therapy. Sem in Oncology 1994; 22. Steele JK, Lui D, Stammers AT, et al: texaphyrin in a monkey model of choroidal
21(Supp)15:4–10. Suppressor deletion therapy: selective neovascularization. Invest Ophthalmol Vis
9. Richter AM, Chowdhary R, Ratkay L, et al: elimination of T-suppressor cells in vivo Sci 1999; 40:S402.
Nononcologic potentials for the using Hp conjugate to a monoclonal 36. Mori K, Ohta M, Katagiri T, et al:
photodynamic therapy. Proc Soc Photo- antibody permits animal to reject synergic Photodynamic therapy (PDT) with
Optical Instr Eng 1994; 2078:293–304. tumor cells. Cancer Immunol Immunother combination of a new sensitizer: NPe6 and
10. Hasan T, Parish JA: Photodynamic therapy 1988; 26:125–131. a diode laser emitting at 664 nm.
of cancer. In: Holland JF, Frei E, et al, eds. 23. Gomer CJ, Ferrario A, Hayashi N, et al: Investigative Ophthalmol Vis Sci 1996;
Cancer medicine. 4th edn. New York: Molecular, and tissue response following 37/3:S3177.
Williams and Wilkins; 1996:739–751. photodynamic therapy. Lasers Surg Med 37. Obana A, Gohto Y, Miki T, et al: Selective
11. Foote CS: Photosensitized autooxidation 1988; 8:450–463. photodynamic effects of the new
and singlet oxygen: consequences in 24. Evans S, Matthews W, Perry R, et al: Effect photosensitizer ATX-S10(Na) on choroidal
biological systems. In: Proyer WA, ed. Free of Photodynamic therapy on tumor necrosis neovascularization in monkeys. Arch of
radicals in biology. New York: Academic factor production by murine macrophages. ophthalmol 2000 ; 118:650–658.
Press; 1976:85. J Natl Cancer Inst 1990; 82:34–39. 38. Richter A, Waterfield E, Jain A, et al:
12. Henderson BW: Probing the effects of 25. Svaasand LO, Ellingson R: Optical Photosensitizing potency of structural
photodynamic therapy through in vivo in properties of human brain. Photochem analogs of benzoporphyrin derivative
vitro methods. In: Kessel D, ed. Photobiol 1983; 38:283–299. (BPD-MA) in a mouse model. Br J Cancer
Photodynamic therapy of Neoplastic 26. Dougherty TJ, Potter WR, Weishaupt KR: 1991; 63:87–93.
disease. Baco Raton, FL: CRC Press; The structure of active component of 39. Richter AM, Cerruti-Sola S, Sternberg ED,
1990:169–188. hematoporphyrin derivative. In: Doirin DR, et al: Biodistribution of tritiated
13. Fisher AMR, Murphree AL, Gomer CJ: Gomer CJ, eds. Porphyrin localization and benzoporphyrin derivative (3H-BPD-MA),
Clinical and preclinical photodynamic treatment of tumors. New York: Alan R Liss; a new photosensitizer, in normal and
therapy. Lasers Surg Med 1995; 1984: 304–314. tumor-bearing mice. J Photochem
17:2–31. 27. Richter AM, Waterfield E, Jain AK, et al: In Photobiol 1990; 5:231–244.
14. Henderson BW, Dougherty TJ: How does vitro and phototoxic properties of four 40. Lui J, Hruza L, Kollias N, et al:
photodynamic therapy work? Photochem structurally related benzoporphyrin Photodynamic therapy of malignant skin
Photobiol 1992; 55:931–948. derivatives. Photochem Photobiol 1990; tumors with benzoporphyrin derivative-
15. Jori G: Low density lipoproteins–liposome 52:495–500. monoacid ring A (BPD-MA): Preliminary
delivery system for tumor photosensitizers 28. Richter AM, Jain AK, Canaan AJ, et al: investigations. In: Anderson RR, et al,
in vivo. In: Handerson BW, Dougherty TJ, Photosensitizing effeciency of two Chairs/eds. Proceedings of lasers in
eds. Photodynamic therapy, basic regioisomers of the benzoporphyrin otolaryngology, dermatology, and tissue
principles and clinical applications. derivative monoacid ring A. Biochem welding, Jan 16–18, 1993. Los Angeles CA.
1960 New York: Dekker; 1992:173–186. Pharmacol 1992; 43:2349–2358. Bellingham WA: SPEI, C; 1993: 147–151.
Photodynamic Therapy

41. Miller JW, Walsh AW, Kramer M, et al: 50. Miller JW, Schmidt-Erfurth U, Sickenberg photodynamic therapy using verteporfin for
Photodynamic therapy of experimental M, et al: Selected angiographic findings choroidal neovascularization in pathologic
choroidal neovascularization using following photodynamic therapy of myopia, ocular histoplasmosis syndrome,
lipoprotein delivered benzoporphyrin. Arch subfoveal choroidal neovascularization angioid streaks, and idiopathic causes.
Ophthalmol 1995; 113:810–818. using BPD. Invest Ophthal Vis Sci 1996; Arch Ophthalmol 2000; 118:327–336.
42. Moulton RS, Walsh AW, Miller JW, et al: 37:1014. 58. Blinder KJ, Blumenkranz MS, Bressler NM,
Response of retinal and choroidal vessels 51. Miller JW, Schmidt-Erfurth U, Sickenberg et al: Verteporfin therapy of subfoveal
to photodynamic therapy using M, et al: Photodynamic therapy with choroidal neovascularization in pathologic
benzoporphyrin derivative. Invest verteporfin for choroidal neovascularization myopia: 2-year results of a randomized
Ophthalmol 1993; 34:S2294. caused by age-related macular clinical trial – VIP report no. 3.
43. Kramer M, Miller JW, Michaud N, et al: degeneration. Results of a single treatment Ophthalmology 2003; 110:667–673.
Liposomal benzoporphyrin derivative in a Phase 1 and 2 study. Arch Ophthalmol 59. Rosenfeld PJ, Saperstein DA, Bressler NM,
verteporfin photodynamic therapy: 1999; 117:1161–1173. et al: Photodynamic therapy with
selective treatment of choroidal 52. Treatment of Age-related Macular verteporfin in ocular histoplasmosis:
neovascularization in monkeys. Degeneration With Photodynamic Therapy uncontrolled, open-label 2-year study.
Ophthalmology 1996; 103:427–438. (TAP) Study Group: Photodynamic therapy Ophthalmology 2004 Sep; 111:1725–1733.
44. Allison BA, Pritchard PH, Levy JG: of subfoveal choroidal neovascularization in 60. Rechtman E, Danis RP, Pratt LM et al:
Evidence of low-density lipoprotein age-related macular degeneration with Intravitreal triamcinolone with
receptor mediated uptake of BPD. Brit J verteporfin. One-year results of 2 photodynamic therapy for subfoveal
Cancer 1994; 69:833–839. randomized clinical trialsTAP Report 1. choroidal neovascularisation in age-related

CHAPTER 147
45. Kramer M, Kenney AG, Delori F, et al: Arch Ophthalmol 1999; 117:1329–1345. macular degeneration. Br J Ophthalmol
Imaging of experimental choroidal 53. Bressler NM: Photodynamic therapy of 2004; 88:344–347.
neovascularization (CNV) using liposomal subfoveal choroidal neovascularization in 61. Spaide RF, Sorenson J, Maranan L:
Benzoporphyrin Derivative mono-acid age-related macular degeneration with Photodynamic therapy with verteporfin
(BPD-MA) angiography. 1995. Invest verteporfin: two-year results of 2 combined with intravitreal injection of
Ophthal Vis Sci. 36/4:S236. randomized clinical trials-tap report 2. Arch triamcinolone acetonide for choroidal
46. Husain D, Miller JW, Michaud N, et al: Ophthalmol 2001; 119:198–207. neovascularization Ophthalmology 2005;
Intravenous infusion of liposomal 54. Verteporfin Therapy for Subfoveal 112:301–304.
benzoporphyrin derivative verteporfin for Choroidal Neovascularization in Age- 62. Spaide RF, Sorenson J, Maranan L:
photodynamic therapy of experimental Related Macular Degeneration: Four-Year Combined photodynamic therapy and
choroidal neovascularization. Arch Results of an Open-Label Extension of 2 intravitreal triamcinolone for nonsubfoveal
Ophthalmol 1996; 114:978–985. Randomized Clinical Trials: TAP Report No. choroidal neovascularization. Retina 2005;
47. Husain D, Miller JW, Michaud N, et al: 7. Arch Ophthalmol 2005; 123:1283–1285. 25:685–690.
Long term effects of photodynamic therapy 55. Verteporfin therapy of subfoveal choroidal 63. Husain D, Kim I, Gauthier D et al: Safety
(PDT) using liposomal benzoporphyrin neovascularization in age related macular and efficacy of intravitreal injection of
derivative (BPD) on experimental choroidal degeneration: two year results of a ranibizumab in combination with
neovascularization (CNV) and normal retina randomized clinical trial including lesions verteporfin PDT on experimental choroidal
choroid. Invest Ophthal Vis Sci 1996; with no classic choroidal neovascularization in the monkey. Arch
37:1013. neovascularization – Verteporfin in Ophthalmol 2005; 123:509–516.
48. Reinke MH, Canakis C, Husain D, et al: photodynamic therapy report 2. Am J 64. Heier JS, Boyer DS, Ciulla TA, et al:
Photodynamic therapy retreatment o Ophthalmol 2001; 131:541–560. Ranibizumab combined with verteporfin
normal retina and choroid in the primate. 56. Azab M, Boyer DS, Bressler NM, et al: photodynamic therapy in neovascular age-
Invest Ophthal Vis Sci 1997; 38:75. Verteporfin therapy of subfoveal minimally related macular degeneration: year 1
49. Schmidt-Erfurth U, Miller JW, Sickenberg classic choroidal neovascularization in results of the FOCUS Study. Arch
M, et al: Photodynamic therapy of age-related macular degeneration: 2-year Ophthalmol 2006; 24:1532–1542.
subfoveal choroidal neovascularization results of a randomized clinical trial. Arch
using benzoporphyrin derivative: first Ophthalmol 2005; 123:448–457.
results of multicenter trial. Invest Ophthal 57. Sickenberg M, Schmidt-Erfurth U, Miller
Vis Sci 1996; 37:B492. JW, et al: A preliminary study of

1961
CHAPTER
Anti-VEGF and Other Pharmacologic Treatments
148 for Age-Related Macular Degeneration
Ivana K. Kim and Joan W. Miller

OVERVIEW homolog.9 VEGF-C and -D mediate lymphangiogenesis.


Human VEGF-A is alternatively spliced into at least four active
The identification of vascular endothelial growth factor (VEGF) human isoforms, consisting of 121, 165, 189, and 206 amino
as a key mediator of ocular angiogenesis has led to the devel- acids. VEGF121 and VEGF165 are diffusible forms, although
opment of several pharmacologic agents designed for targeted VEGF165 retains heparin-binding ability. The other longer
inhibition of this molecule. Two of these agents, pegabtanib and isoforms remain bound to extracellular matrix. VEGF165 is the
ranibizumab, have been proven efficacious in the treatment of predominant form and possesses greater mitogenic activity.
neovascular age-related macular degeneration (AMD) and are Some recent work in rodent models suggests that this isoform
currently available for clinical use. Additional drugs targeting may be the primary mediator of pathologic neovasculari-
the VEGF pathway as well as other antiangiogenic compounds zation.10 Two tyrosine kinases serve as VEGF-A receptors,
are under evaluation in clinical trials and appear promising. As VEGFR-1 (fms-like tyrosine kinase 1, flt-1) and VEGFR-2
options for pharmacologic therapy multiply, combination (kinase domain region, KDR or fetal liver kinase, flk-1). VEGFR-
therapy using various agents and modalities may provide 2 appears to play the major role in signaling the mitogenic,
improved visual outcomes for patients with choroidal angiogenic, and permeability effects of VEGF.9
neovascularization due to AMD.
Recent progress in the treatment of neovascular AMD has
enabled an evolution from nonspecific, ablative laser therapy VEGF AS A MEDIATOR OF OCULAR
to targeted pharmacologic approaches. These new pharma- NEOVASCULARIZATION
cologic agents represent the culmination of over a decade of It had long been appreciated that neovascularization of the
investigations into the mechanisms of ocular angiogenesis. retina and iris was temporally and spatially correlated with
Early observations of increased vascularity associated with retinal ischemia due to various etiologies. The discovery that
the growth of tumors gave rise to the field of angiogenesis.1 hypoxia could upregulate VEGF expression made VEGF a
Ide and colleagues first proposed the idea of secreted factors plausible candidate for the factor responsible for abnormal
which could promote growth of blood vessels in 1939, based blood vessel growth in the eye.11,12 Evidence from both clinical
on studies of vascular development accompanying the growth and animal studies then accumulated to support the critical role
of transplanted rabbit epithelioma.2 Similarly, in 1948, of VEGF in ocular neovascularization. VEGF expression was
Michaelson suggested that a diffusible ‘Factor X’ from the retina shown to be correlated with iris neovascularization in a primate
stimulated the retinal and iris neovascularization seen in model of ischemic retinal vein occlusion,13 and a similar
diabetic retinopathy.3 However, many years passed before such correlation was demonstrated in a neonatal mouse model of
proposed angiogenic factors were identified. retinal neovascularization.14 Additionally, injection of VEGF in
normal primate eyes produced iris neovascularization, neovas-
THE ROLE OF VEGF IN OCULAR cular glaucoma, and retinal microangiopathy.15,16 Suppression
ANGIOGENESIS of neovascularization was also achieved by VEGF inhibition
through the use of neutralizing antibodies in the primate model
The idea of inhibiting angiogenesis was first proposed as a of iris neovascularization and chimeric proteins acting as
strategy for treating cancer in 1971 by Judah Folkman, who soluble VEGF receptors in the mouse model of retinal
spearheaded the efforts to discover tumor angiogenesis factors.4 neovascularization.17,18
In 1983, Senger and colleagues discovered a protein secreted Human clinical studies confirmed the association of VEGF
from a guinea pig tumor cell line which was a potent inducer of expression with pathologic ocular neovascularization. Measure-
vascular leakage and named it vascular permeability factor ments of vitreous VEGF levels in patients with active prolifer-
(VPF).5 In 1989, Napoleon Ferrara and colleagues at Genentech ative diabetic retinopathy demonstrated significantly higher
identified a molecule in the conditioned media from bovine VEGF concentrations when compared to patients with other
pituitary follicular cells which promoted the proliferation of retinal disorders not characterized by abnormal blood vessel
endothelial cells and called it VEGF.6 The subsequent cloning growth.19 Another study which analyzed both aqueous and
of these two factors demonstrated that they were the same vitreous levels of VEGF in a variety of conditions characterized
protein.7,8 by ocular neovascularization correlated elevated ocular VEGF
This molecule is also known as VEGF-A, as it belongs to concentrations to the presence of active neovascularization.
a family of genes including placental growth factor (PlGF), This study also demonstrated reduction of VEGF levels after
VEGF-B, VEGF-C,VEGF-D, and VEGF-E, a viral-derived panretinal photocoagulation for retinal neovascularization.20
1963
RETINA AND VITREOUS

TABLE 148.1. Agents Approved (*) or in Clinical Trials for Treatment of Neovascular
Macular Degeneration

Agent Target Delivery

Pegaptanib* VEGF Intravitreal


Ranibizumab* VEGF Intravitreal
Bevacizumab VEGF Intravitreal
Bevasiranib (Cand5) VEGF Intravitreal
Sirna-027 VEGFR-1 Intravitreal
VEGF Trap VEGF, PlGF Intravitreal
AdPEDF PEDF (gene transfer) Intravitreal
Anecortave acetate Proteolytic cascade leading to Juxtascleral
extracellular matrix degradation
Squalamine lactate Intracellular signaling Systemic
SECTION 10

VEGF IN CHOROIDAL NEOVASCULARIZATION 0

Mean Change in Visual Acuity


–1
–2
Although hypoxia is not clearly a stimulus for the development –3
–4
of choroidal neovascularization (CNV), VEGF does appear to –5

(no. of letters)
–6 1.0 mg Pegaptanib
–7
function in CNV formation. –8
–9 0.3 mg Pegaptanib

Overexpression of VEGF through gene transfer using viral –10


–11 3.0 mg Pegaptanib
–12
vectors induces choroidal neovascularization in rat and primate –13
–14
models.21,22 Implantation of microspheres containing VEGF –15
–16 Sham injection
–17
into the subretinal space also promotes choroidal neovasculari- 0 6 12 18 24 30 36 42 48 54

zation in primates.23 Additionally, increased VEGF expression Weeks

No. at Risk
has been demonstrated in rodent and primate models of laser- 0.3 mg Pegaptanib 294 286 289 269 273 271 265 271 266 271
1.0 mg Pegaptanib 300 292 291 291 287 285 278 270 267 275
induced CNV.24,25 Conversely, blockade of the VEGF pathway 3.0 mg Pegaptanib 296 286 283 281 283 278 273 267 259 264
Sham Injection 296 291 288 287 281 282 278 275 269 275
using monoclonal antibodies in the primate model of laser-
induced CNV26 and kinase inhibitors in a mouse model27 FIGURE 148.1. Mean change in visual acuity for all dose groups in
prevents CNV formation. This type of experimental data is also the first year of the VISION trials of pegaptanib (Macugen) for
supported by clinical evidence demonstrating the presence of neovascular AMD.
VEGF in choroidal neovascular membranes removed from From Gragoudas ES, Adamis AP, Cunningham ET, Jr, et al: Pegaptanib for
neovascular age-related macular degeneration. N Engl J Med 2004;
patients with neovascular AMD.28,29
351:2805–2816.

ANTI-VEGF AGENTS
Given such weighty evidence for VEGF as a key mediator of of CNV lesions due to AMD30 (Figs 148.1 and 148.2). Patients
ocular neovascularization, it became a prime therapeutic target were randomized to receive 0.3,1.0,or 3.0 mg pegpatanib intra-
in the search for more efficacious treatments for neovascular vitreally or sham injection once every 6 weeks. With pegaptanib
AMD (Table 148.1). (0.3 mg) treatment, 70% of patients avoided moderate vision loss
at 1 year versus 55% of patients in the control group. The
proportion who maintained or gained vision was 33% in the
PEGAPTANIB 0.3 mg pegaptanib group versus 23% of those receiving sham.
However, the number of patients who gained three or more
Key Features lines of vision was small: 6% in the pegaptanib (0.3 mg) group
• RNA aptamer
versus 2% in the control group (clinical example, Fig. 148.3).
• Intravetreal injection
Two-year results of this study suggested that there is a benefit
• Dosing: Every 6 weeks
to continued treatment for a second year31 (Fig. 148.4). Patients
• Efficacy for all lesion types
who received pegaptanib for 2 years were less likely to lose ≥15
letters than those who discontinued treatment after 1 year. Ten
Pegaptanib sodium (Macugen) became the first anti-VEGF agent percent of patients treated with pegaptanib for 2 years gained ≥3
approved by the United States Federal Drug Administration lines of vision. Additionally, the proportion of patients who
(FDA) for ophthalmic use (0.3 mg intravitreal injection) in gained vision was higher in the group that received 2 years of
December 2004. It is a modified 28-base RNA aptamer which treatment than those that received usual care (Fig. 148.5).
binds with high affinity to the 165-amino-acid isoform of The safety profile of pegaptanib appears quite favorable.32
VEGF. The efficacy of pegaptanib in preventing moderate The most common ocular adverse events noted during the trials
vision loss (≥3 lines) from subfoveal CNV was demonstrated by included eye pain, vitreous floaters, punctate keratitis, and
the VEGF Inhibition Study in Ocular Neovascularization increased intraocular pressure. These symptoms were mainly
(VISION), two concurrent, randomized, double-masked, sham- attributed to the injection procedure rather than the drug. The
controlled studies involving 1186 patients, which showed a serious ocular adverse events were also injection-related and
1964 treatment benefit for all subtypes and sizes (up to 12 disc areas) consisted of endophthalmitis (1.3%/patient in year 1), retinal
Anti-VEGF and Other Pharmacologic Treatments for Age-Related Macular Degeneration

FIGURE 148.2. Mean changes in visual acuity


at week 54 of the VISION trials according to
baseline characteristics of (a) angiographic
subtype, (b) visual acuity, and (c) lesion size. *,
p < 0.05 for pegaptanib vs sham injection; †,
p < 0.001; ‡, p < 0.01.
From Gragoudas ES, Adamis AP, Cunningham ET, Jr,
et al: Pegaptanib for neovascular age-related macular
degeneration. N Engl J Med 2004; 351:2805–2816
Copyright © 2004 Massachussets Medical Society.
All rights reserved.

CHAPTER 148
b c

FIGURE 148.3. (a) Color photo and fluorescein angiogram of patient with occult choroidal neovascular membrane. Vision was 20/50. Patient
received pegaptanib therapy. (b) OCT scans of same patient shown in (a). The left scan was taken prior to treatment and reveals a pigment
epithelial detachment and subretinal fluid. The right scan was taken after 4 injections of pegaptanib (6 months of therapy). The subretinal fluid
had resolved and vision had improved to 20/30.

detachment (0.7%/patient in year 1), and traumatic cataract thalmitis in the second year experienced severe vision loss.
(0.6%/patient in year 1). The incidence of these events was Clinical studies of systemically administered bevacizumab
similar for the second year across all cohorts. Only one of the (a monoclonal antibody directed against all VEGF-A isoforms)
cases of endophthalmitis in the first year resulted in severe in oncology patients identified hypertension, hemorrhage, and
vision loss, and none of the patients who developed endoph- thromboembolic events as serious systemic adverse events.33,34 1965
RETINA AND VITREOUS

Patient Treatment Year 1 ˜ Year 2 40


50 0.3 mg ˜ 0.3 mg, N=133
Patient Treatment Year 1 ˜ Year 2
0.3 mg ˜ Discontinue, N=132
0.3 mg ˜ 0.3 mg, N=133
Usual care, N=107
45 0.3 mg ˜ Discontinue, N=132
30
Vision (letters)

Usual care, N=107

40

Percent of Patients
20
35

30
54 60 66 72 78 84 90 96 102 10
Weeks

FIGURE 148.4. Mean visual acuity from week 54 to 102 in the VISION
SECTION 10

trials.
From VISION Clinical Trial Group: Year 2 Efficacy Results of 2 Randomized 0
Controlled Clinical Trials of Pegaptanib for Neovascular Age-Related Macular ≥0 ≥1 ≥2 ≥3
Degeneration. Ophthalmology 2006 Epub July 5, 2006; 113: 1508.el–1508.e25, Lines of Vision Gained
with permission from the American Academy of Ophthamology.)

FIGURE 148.5. Proportion of patients gaining vision after 2 years in


There was no suggestion that intravitreal administration of the VISION trials.
pegaptanib was associated with such systemic side effects From VISION Clinical Trial Group: Year 2 Efficacy Results of 2 Randomized
through year 2 of the VISION trials. Controlled Clinical Trials of Pegaptanib for Neovascular Age-Related Macular
Degeneration. Ophthalmology 2006 Epub Jul 5, 2006; 113: 1508.el–1508.e25,
with permission from the American Academy of Ophthamology.)
RANIBIZUMAB
Key Features A molecule (Fig. 148.6). The benefit of ranibizumab for patients
• Anti-VEGF antibody fragment with minimally classic and occult CNV was demonstrated in
• Intravetreal injection the MARINA (minimally classic/occult trial of the anti-VEGF
• Dosing: Every 4 weeks antibody ranibizumab) study, a randomized, multicenter,
• Efficacy for all lesion types double-masked, sham-controlled trial with an enrollment of
• Superior to photodynamic therapy in randomized trial 716 patients.35 Patients in this study were randomized to
• 30–40% of patients with ≥ 3 lines visual improvement monthly injections of either 0.3 or 0.5 mg ranibizumab or sham
injections. After 1 year, 95% of patients who were treated with
More recently, results of Phase III trials also confirmed the ranibizumab avoided moderate vision loss, compared to 62% of
efficacy of ranibizumab (Lucentis) in neovascular AMD, with patients who were in the control group. Thirty-four percent of
rates of visual improvement not previously achieved. This drug patients treated with ranibizumab 0.5 mg experienced visual
was approved by the US FDA for the treatment of neovascular improvement of three or more lines compared to 5% of patients
AMD in June 2006 at a dose of 0.5 mg given intravitreally. receiving sham injections. The mean change in visual acuity at
Ranibizumab is a recombinant, humanized monoclonal anti- the end of the first year of treatment was a gain of 7.2 letters in
body fragment (48kDa) which binds all isoforms of the VEGF- the 0.5 mg ranibizumab group compared to a loss of 10.4 letters

FIGURE 148.6. Ranibizumab (Lucentis ®) is a


recombinant, humanized, monoclonal antibody
fragment with high affinity for all isoforms of
VEGF-A.
Courtesy of Genentech, Inc.

1966
Anti-VEGF and Other Pharmacologic Treatments for Age-Related Macular Degeneration

FIGURE 148.7. Mean change in visual acuity over 2 years of the


MARINA trial of ranibizumab for minimally classic/occult CNV due to

CHAPTER 148
AMD. Vertical lines are one standard error of the means. *, p < 0.001 FIGURE 148.9. Mean change in visual acuity over 1 year of the
vs sham at each month and p = 0.006 (for 0.3 mg) and 0.003 (for ANCHOR trial of ranibizumab compared to verteporfin PDT for
0.5 mg) at day 7. predominantly classic CNV due to AMD. Vertical bars represent one
From Rosenfeld PJ, Brown DM, Heier JS, Boyer DS, Kaiser PK, Chung CY, standard error of the mean. P < 0.001 for all comparisons vs PDT at
Kim RY; MARWA Study Group. Ranibizunab for neomuscular age-related each month.
macular degeneration. N Engl J Med 2006; 355: 1419–1430 Copyright © 2006 Brown DM, Kaiser PK, Michels M, et al.; ANCHOR Study Group. Ranibizumab
Massachusetts Medical Society. All right reserved. versus verteporfin for neovascular age-related macular degeneration. N Eng J
Med 2006; 355: 1432–1444. Copyright © 2006 Massachusetts Medical Society.
All rights reserved.

group. The mean changes in visual acuity in the higher-dose


ranibizumab group was a gain of 11.3 letters versus a mean loss
of 9.5 letters in the PDT group (Fig. 148.9). Results of a
subgroup analysis of this data suggested a consistent benefit of
ranibizumab compared to PDT regardless of age, visual acuity,
lesion size, or presence of occult CNV at baseline.37
FIGURE 148.8. Percentage of patient achieving 20/40 or better in the The safety profile of ranibizumab appears similar to that of
MARINA trial. Vertical lines are 95% confidence intervals. †, p < 0.001 pegaptanib. The rates of serious ocular adverse events at month
vs sham. 24 of the MARINA study for ranibizumab 0.5 mg were
From Rosenfeld PJ, Brown DM, Heier JS, Boyer DS, Kaiser PK, Chung CY, endophthalmitis 1.3%, uveitis 1.3%, retinal tear 0.4%, and lens
Kim RY; MARWA Study Group. Ranibizunab for neomuscular age-related damage 0.4%.35 Similarly, at month 12 in the ANCHOR study
macular degeneration. N Engl J Med 2006; 355: 1419–1430 Copyright © 2006
for the 0.5 mg ranibizumab group, the rate of endophthalmitis
Massachusetts Medical Society. All right reserved.
was 1.4% and uveitis was seen in 0.7% of patients.36 There did
not appear to be a significant increase in systemic adverse
events such as hypertension or arterial thromboembolic events
in the sham group. Approximately 40% of patients treated with with ranibizumab treatment in either study.
ranibizumab achieved visual acuity of 20/40 or better at the Alternative dosing regiments for ranibizumab remain under
end of the first year versus 11% of the sham-treated group investigation. The PIER (A Phase IIIb, multicenter, randomized,
(Fig. 148.7) The treatment benefit was sustained through 2 double-masked, sham injection-controlled study of the efficacy
years. At month 24, 90% of treated patients (0.5 mg) avoided and safety of ranibizumab in subjects with subfoveal choroidal
moderate vision loss compared to 53% in the control group. neovascularization with or without classic CNV secondary to
Similarly, 33% of treated patients gained three or more lines of AMD) study was designed to examine the effect of a modified
vision after 2 years versus 4% of the control group, and the dosing regimen for ranibizumab consisting of three initial
mean change in visual acuity was +6.6 letters in the treated injections at monthly intervals followed by mandated doses at
patients versus 14.9 in the control group (Fig. 148.8). three month intervals.38 A mean gain in visual acuity was seen
The results of the ANCHOR (anti-VEGF antibody for the in the treatment groups at month 3 (+4.3 letters in 0.5 mg
treatment of predominantly classic choroidal neovas- group vs 8.7 in sham group), but was not sustained through
cularization in AMD) trial demonstrated that ranibizumab is month 12. One-year results revealed that the mean change in
superior to PDT for predominantly classic CNV. This study visual acuity was a loss of 1.6 and 0.2 letters in patients treated
randomized 423 patients to receive verteporfin PDT and sham with 0.3-, and -0.5 mg of ranibizumab respectively, compared
injection or sham PDT and ranibizumab 0.3 or 0.5 mg. The to a loss of 16.3 letters in the control group (Fig. 148.10). More
patients received ranibizumab or sham injections monthly. patients treated with ranibizumab avoided moderate vision loss
Verteporfin or sham PDT was administered upon study entry (90% in 0.5 mg group versus 49% in control group), confirming
and could be repeated every 3 months based on evidence of the benefit of ranibizumab treatment. However, fewer patients
leakage on angiography. The results at month 12 revealed that gained 3 or more lines of vision (13% in 0.5 mg group vs 10%
96% of patients treated 0.5 mg ranibizumab avoided moderate in sham group) as compared to the pivotal Phase III trials
vision loss compared to 64% of patients treated with PDT.36 MARINA and ANCHOR. These results suggest that quarterly
Furthermore, 40% of patients treated with ranibizumab 0.5 mg, dosing may not be as effective as monthly injections. The PIER
gained three or more lines of vision versus 6% in the PDT study remained ongoing at the time of this writing. 1967
RETINA AND VITREOUS

individually tailored dosing may be similarly efficacious to


monthly dosing for ranibizumab.

BEVACIZUMAB

Key Features
• Full-length anti-VEGF antibody
• Off-label use for neovascular AMD
• Intravitreal injection
• No randomized trials for AMD completed to date

Bevacizumab (Avastin) became the first anti-VEGF drug


licensed for clinical use when it was approved for the treatment
of metastatic colorectal cancer in February 2004. It is a
FIGURE 148.10. Mean change in visual acuity over 1 year of the PIER humanized, full-length monoclonal antibody (149 kDa) which
trial of ranibizumab for neovascular AMD which involved quarterly binds all isoforms of VEGF-A and was developed for intra-
SECTION 10

dosing. venous administration. Primate studies of retinal penetration


From Lucentis Prescribing Information. Genentech, Inc. after intravitreal injection using radiolabeled antibody
preparations demonstrated that full-length antibody failed to
penetrate the internal limiting membrane while an antibody
The Phase I/II, open-label, single-center PrONTO fragment diffused through all layers of the retina40 (Fig. 148.11).
(Prospective Optical Coherence Tomography Imaging Patients Such data indicating a molecular weight limitation for retinal
with Neovascular AMD Treated with Intraocular Lucentis) penetration as well as concerns regarding potential cytotoxicity
Study conducted at the Bascom Palmer Eye Institute was also related to the Fc portion of antibodies provided the rationale for
ongoing at the time of this writing and involved a modified the development of ranibizumab for neovascular AMD.
dosing protocol using three initial injections of 0.5 mg However, in light of positive data from clinical trials of
ranbizumab at monthly intervals followed by additional doses pegaptanib and ranibizumab for neovascular AMD, the off-label
as indicated.39 Strict criteria for additional dosing in this study use of bevacizumab for this condition was initiated by clinicians
were predetermined and included visual acuity, ocular prior to the approval of ranibizumab.
coherence tomography (OCT), and fluorescein angiography An early open-label, single-center study of systemically
findings. At 1 year, patients in this study gained an average of administered bevacizumab in nine patients with neovascular
9.3 letters. The mean number of injections was 5.5, although AMD suggested visual benefit with mean increase in vision at
there was significant variability among patients, and the mean 12 weeks as well as a mean decrease in central retinal thickness
time to first retreatment was 4.3 months. Although there is no by OCT.41 There was also some improvement in these
control group for comparison, these findings suggest that measures noted in the fellow eyes of the study patients. A mild

FIGURE 148.11. Microautoradiographic


images of the distribution of (a) 125I-rhuMab
HER2 (full-length antibody against HER2) and
(b) 125I-rhuMab VEGF Fab (antibody fragment
against VEGF) after intravitreal injection in
rhesus monkey. In (a), the full-length antibody
remains at the level of the inner limiting
membrane. In (b), the antibody fragment
penetrates through the entire neural retina to
the level of the RPE.
Courtesy of Genentech, Inc. See Mordenti J,
Cuthbertson RA, Ferrara N, et al. Comparisons of the
intraocular tissue distribution, pharmacokinetics, and
safety of 125I-labeled full-length and Fab antibodies in
rhesus monkeys following intravitreal administration.
Toxicol Pathol. Sep–Oct 1999; 27(5):536–544.

a b
1968
Anti-VEGF and Other Pharmacologic Treatments for Age-Related Macular Degeneration

RISC for a new class of therapeutic agents. Various companies have


siRNA developed siRNAs targeting VEGF or its receptors and shown
efficacy in preclinical models.
Bevasiranib sodium (formerly Cand5) is an siRNA directed
against VEGF developed by Acuity Pharmaceuticals. The Phase
I clinical trial in patients with neovascular AMD indicated that
the drug was safe and well tolerated when delivered intra-
vitreally at doses up to 3.0 mg (Prenner JL, Thompson JT, Miller
DG, et al: An open label study for the evaluation and
tolerability of five dose levels of intravitreous VEGF siRNA
(Cand5) in patients with wet age-related macular degeneration.
Paper presented at: American Academy of Ophthalmology
Annual Meeting, Chicago, IL, 14–18 Oct 2005.). The Phase II
CARE Study (Cand5 anti-VEGF RNAi Evaluation) tested three
doses of bevasiranib in 129 patients. Preliminary results sug-
gested safety as well as efficacy at all doses.49 Phase III trials are
expected to begin in 2007.
Sirna-027 is another siRNA molecule which targets vascular

CHAPTER 148
mRNA degradation
endothelial growth factor receptor-1 (VEGFR-1) developed by
Sirna Therapeutics. The Phase I dose escalation trial involved
26 patients with neovascular AMD. At 8 weeks after a single
injection, 96% of these patients showed stabilization of vision
with 23% experiencing visual improvement of three or more
FIGURE 148.12. In the process of RNA interference, short interfering lines.50 Decreases in central retinal thickness measured by
RNAs (siRNAs) associate with RISC (RNA-induced silencing complex) OCT were also observed. Phase II studies are anticipated.
to trigger selective degradation of particular RNA transcripts.
VEGF Trap
Another novel molecule designed to block VEGF-mediated
angiogenesis is VEGF Trap (Regeneron Pharmaceuticals), which
increase in systolic blood pressure that was felt to be drug- contains immunoglobulin domains of both VEGFR-1 and
related was observed at 6 weeks. VEGFR-2 fused to the constant region of human IgG.51 It
Due to concerns regarding potential systemic toxicities of functions as a high-affinity soluble receptor that binds and
intravenously administered bevacizumab, intravitreal use was neutralizes both VEGF and PlGF. The Phase I study involved
adopted. Several short term reports of intravitreal bevacizumab doses ranging from 0.05 to 4 mg which were administered as a
for the treatment of patients with neovascular AMD suggest a single intravitreal injection to 21 patients with neovascular
beneficial effect on visual acuity and retinal thickness.42–45 The AMD. At 6 weeks, 95% of the patients had stabilization of
largest of these series was a retrospective study of 266 eyes of vision (loss of ≤15 letters).52 The mean change in visual acuity
266 patients, the majority (69.7%) of whom were considered to for all patients was a gain of 4.8 letters at 6 weeks. In the
have had an inadequate response to other treatments.44 These highest dose groups (2 and 4 mg), the mean improvement in
patients were treated with three injections of 1.25 mg visual acuity was 13.5 letters. Again, the visual acuity results
bevacizumab at monthly intervals. Three-month follow-up were accompanied by decreases in OCT measurements of
data was available for 141 eyes and revealed visual acuity central retinal thickness. A Phase II study is in progress.
improvement (halving of the visual angle) in 38.3% of patients
as well as progressive decrease in mean central macular thick- OTHER ANTIANGIOGENIC AGENTS
ness measurements by OCT (340 mm baseline to 213 mm at
month 3).
No apparent safety concerns have been detected in these
ANECORTAVE ACETATE
studies with intravitreal doses up to 2.5 mg. Additionally, histo- Given the complexity of the angiogenic process, drugs which
logic and electrophysiologic studies in rabbits have shown no affect an array of factors rather than one molecule may be
evidence of toxicity after intravitreal bevacizumab in doses up desirable. The degradation of capillary basement membrane
to 2.5 mg.46,47 However, longer-term, prospective, randomized and extracellular matrix is an essential step in the migration
studies are necessary to fully evaluate the safety and efficacy of endothelial cells during neovacularization and occurs
of bevacizumab as a treatment for neovascular AMD. through the action of proteases such as urokinase-type plasmi-
nogen activator (uPA) and matrix metalloproteinases.53 The
angiostatic cortisene anecortave acetate (Retaane, Alcon, Inc.) is
OTHER ANTI-VEGF AGENTS a cortisol derivative which has been chemically modified to
RNA Interference eliminate all glucocorticoid activity, thereby eliminating ocular
A powerful new technology for inhibiting the function of side effects such as glaucoma and cataracts. It has been shown
selected genes known as RNA inference (RNAi) promises a to upregulate the expression of plasminogen activator inhibitor-1
new generation of anti-VEGF drugs. This strategy exploits a (PAI-1), thereby supressing the action of uPA, which is
cellular phenomenon in which a small, double-stranded RNA necessary for the activation of various other proteases.54 By
molecule (short interfering RNA, siRNA) associates with a inhibition of this proteolytic cascade which is downstream of
RNA–protein complex called RISC (RNA-induced silencing endothelial cell stimulation, anecortave acetate may potentially
complex), resulting in selective degradation of an mRNA block angiogenesis initiated by a variety of signals.
transcript containing a sequence homologous to the siRNA49 In a randomized trial, three different doses (3, 15, 30 mg) of
(Fig. 148.12). Synthesis of siRNA molecules tailored to silence anecortave acetate administered as a posterior juxtascleral
expression of chosen genes is possible and offers the potential injection at 6 month intervals were compared to placebo in 128 1969
RETINA AND VITREOUS

anecortave acetate 15 mg versus placebo, and 84% of


anecortave-treated patients had stabilization of vision versus
50% of those in the placebo group. The results appeared
sustained through the 24 month follow-up with 73% of
anecortave-treated patients maintaining vision (<3 lines lost)
versus 47% of the placebo group (Kaiser PK, Schaffer HI, Zilliox
P, et al: Posterior juxtascleral depot of anecortave acetate for
subfoveal CNV in AMD: 24-month outcomes. Paper presented
at: American Academy of Ophthalmology Annual Meeting,
Anaheim, CA, 17 Nov 2003). No ocular or systemic safety
issues were idenitified. However, concerns regarding the results
of this trial included the decrease in apparent efficacy with the
a highest studied dose and the loss of patients between the 6 and
12 month time points. Overall, 41% of patients exited the study
between 6 and 12 months, with the highest proportion of
patient dropout occurring in the 15 mg group (48.5%).
The Phase III clinical trial of anecortave acetate for neo-
vascular AMD compared the 15 mg dose to verteporfin
SECTION 10

photodynamic therapy in 530 patients with predominantly


classic subfoveal choroidal neovascualrization.56 There was no
statistical difference between the percentage of patients losing
<3 lines of vision (responders) in the anecortave acetate
and PDT groups (45% and 49%, respectively, p = 0.43)
(Fig. 148.14). The confidence interval, however, ranged from
–13.2% favoring PDT to +5.4% favoring anecortave acetate, and
thus the results failed to meet the previously established
noninferiority margin of seven percentage points based on the
b results of the TAP (Treatment of Age-Related Macular
Degeneration with Photodynamic Therapy) Study. However,
FIGURE 148.13. (a) Average change in logMAR visual acuity through an analysis of the subgroup of patients for whom drug reflux
month 12 of patients with subfoveal neovascularization due to AMD at the time of administration was controlled and who received
treated with anecortave acetate vs placebo. (b) Proportion of patients the second injection within the 6 month treatment window
losing <3 lines of logMAR vision. revealed improved outcomes which would have met the
From D’Amico DJ, Goldberg MF, Hudson H, et al: Anecortave acetate as noninferiority criterion.
monotherapy for treatment of subfoveal neovascularization in age-related
Ongoing clinical trials of anecortave acetate include a study
macular degeneration: twelve-month clinical outcomes. Ophthalmology 2003;
investigating three dosage regimens (15 and 30 mg every
110:2372–2383; discussion 2384–2375.
6 months, 15 mg every 3 months) and the Anecortave Acetate
Risk Reduction Trial (AART), which will study the efficacy of
patients with subfoveal choroidal neovascularization due to anecortave acetate 15 mg in preventing wet AMD in patients
AMD.55 The primary outcome of change in logMAR visual with high-risk dry AMD over a 4 year period.
acuity at 12 months showed a statistically significant difference
favoring the 15 mg dose of anecortave acetate compared to
placebo. The mean change in the anecortave group was a loss of SQUALAMINE LACTATE
1.3 lines versus 3.1 lines for the placebo group (Fig. 148.13). Another molecule with a unique mechanism of antiangiogenic
Stabilization of vision (loss of <3 logMAR lines) was seen in activity is squalamine lactate (Evizon, Genaera Corp.) an
79% of the anecortave (15 mg) treated patients compared to aminosterol which potently inhibits growth factor-induced
53% of those receiving placebo. A majority of patients in this proliferation and migration of endothelial cells.57 Squalamine
study had predominantly classic lesions (78.5% in the ane- has been shown to cause redistribution of calmodulin within
cortave acetate 15 mg group and 86.7% of the placebo group), endothelial cells resulting in disruption of various intracellular
and the treatment benefit appeared slightly more pronounced signaling pathways involving calcium.58 In addition to blocking
when this subgroup of patients was analyzed separately. The growth factor mediated signaling, squalamine may also
difference in visual acuity was 2.4 logMAR lines in favor of downregulate integrin expression and induce cytoskeletal

FIGURE 148.14. Per protocol analysis of


percentage of patients losing <15 letters in
study of anecortave acetate vs verteporfin PDT
in patients with predominantly classic CNV due
to AMD (95% confidence limits for difference
between PDT and anecortave on right).
Reprinted from Slakter JS, Bochow TW, D’Amico DJ,
et al: Anecortave acetate (15 milligrams) versus
photodynamic therapy for treatment of subfoveal
neovascularization in age-related macular
degeneration. Ophthalmology 2006; 113(1):3–13 with
permission from the American Academy of
Ophthalmology.

1970
Anti-VEGF and Other Pharmacologic Treatments for Age-Related Macular Degeneration

FIGURE 148.15. Mechanism of action of


squalamine lactate.
Courtesy of Genaera Corp.

CHAPTER 148
rearrangements in endothelial cells59 (Fig. 148.15). Preclinical animal models.67–69 Therefore, a Phase I clinical trial of PEDF
studies have suggested the efficacy of squalamine in inhibiting gene transfer therapy using intravitreal injection of an
ocular neovascularization in rodent and primate models.60,61 adenoviral vector expressing human PEDF (AdPEDF.11) was
Several Phase II studies evaluating intravenous squalamine initiated for patients with advanced wet AMD.70 The initial
treatment for wet AMD are ongoing. The largest Phase II trial dose escalation phase has been completed with no evidence of
is a randomized, multi-center, double-masked, controlled study serious adverse events and no dose-limiting toxicities in 28
involving 108 patients, almost half of whom had active occult subjects. Twenty-five percent of patients experienced mild,
lesions.62 Two different doses of squalamine (40 and 20 mg) are transient ocular inflammation and 21% had increases in
given as weekly intravenous infusions for the first 4 weeks and intraocular pressure which responded to topical therapy.
monthly thereafter. PDT is permitted at the discretion of the Additional treatment of patients with less advanced neovascular
investigator. Preliminary results at 24 weeks revealed that 83% AMD is planned.
of the patients receiving the 40 mg dose lost <15 letters of
visual acuity compared to 71% of the control group. Five percent COMBINATION THERAPY
of patients in the 40 mg group and none in the control group
gained 15 or more letters. In those subjects with a fellow- With the availability of multiple modalities for the treatment of
affected eye (~60%), 11% of the 40 mg group gained ≥15 letters wet AMD, the use of two or more of these agents in
in the fellow eye versus none in the control group. Infusion site combination is now possible. With goals of improved visual
reactions were the most common treatment-related adverse outcomes and a reduction in frequency of treatments, several
events, and no ocular or systemic safety concerns have strategies for combination therapy have been and continue to be
arisen. A Phase III trial is ongoing, and another Phase II trial explored.
investigating higher doses is planned. Increasing evidence supports inflammation as a key
component in AMD pathogenesis, providing a rationale for
the use of intravitreal triamcinolone acetonide in the treatment
PIGMENT EPITHELIUM-DERIVED FACTOR of neovascular AMD. The inhibitory effect of steroids on
Of the several endogenous inhibitors of angiogenesis that macrophages which secrete angiogenic factors and their ability
have been found in ocular tissues, pigment epithelium derived to reduce leukocyte adhesion and migration might have
factor (PEDF) is among the most potent inhibitors of endo- beneficial effects in AMD.71 However, the use of intravitreal
thelial cell migration in vitro.63 The downregulation of PEDF triamcinolone as monotherapy for AMD has proved subop-
has been demonstrated in a mouse model of ischemic timal. In a randomized trial of a single 4 mg dose of intravitreal
retinopathy,63 and patients with proliferative retinopathy from triamcinolone acetonide in 151 eyes of 139 patients with neo-
diabetes or retinal vein occlusion have been shown to have vascular AMD, eyes receiving placebo were noted to have
decreased vitreous levels of PEDF.64 Since PEDF also appears to significantly more growth of the neovascular membrane com-
serve a neurotrophic function in the retina,65 therapeutic use of pared to those receiving triamcinolone at 3 months after treat-
this growth factor may have additional beneficial effects on ment.72 However, no difference was noted at 12 months, and
photoreceptor survival in AMD. Additionally, some preclinical there was no effect of intra-vitreal triamcinolone on visual
evidence suggests that PEDF could enhance the effect of acuity loss at 12 months. Another prospective, nonrandomized
verteporfin photodynamic therapy by increasing endothelial study using 25 mg of intravitreal triamcinolone in 115 patients
cell apoptosis within choroidal neovascular membranes.66 The and 72 controls with exudative AMD demonstrated a visual
efficacy of PEDF gene transfer using an adenoviral vector in acuity benefit at 1 and 3 months follow-up, but not at sub-
inhibiting ocular neovascularization has been demonstrated in sequent follow-ups through a mean of 6 months.73 These 1971
RETINA AND VITREOUS

findings reveal that the effect of an intravitreally delivered


steroid when used as monotherapy is transient. However, the
use of intravitreal steroids in combination with photodynamic
therapy might have more sustained benefits.
The combination of photodynamic therapy and intravitreal
triamcinolone has shown promising results in early studies.
A small pilot study investigated verteporfin PDT immediately
followed by 4 mg of intravitreal triamcinolone in 26 patients
with neovascular AMD.74 One year results demonstrated a
mean visual acuity improvement of 2.4 lines in patients who
had not received any prior treatment, and an improvement of
0.44 lines in those patients who had been previously treated
with PDT. Retreatment rates were also low, at about 1.2 for both
groups. Ten patients (38.5%) developed an increase in intra-
ocular pressure to greater than >24 mmHg. A larger,
prospective, noncomparative case series of 184 patients with
neovascular AMD, the majority with subfoveal CNV, evaluated
the results of photodynamic therapy followed by intravitreal
SECTION 10

injection of 25 mg triamcinolone within 16 h.75 Over the mean


follow-up of 43 weeks, the mean increase in vision was 1.22
Snellen lines and the mean number of required treatments was
1.21. Topical glaucoma therapy was required in 25% of patients
and 1% required glaucoma surgery. Cataract progression was
observed in 48.73% of phakic eyes. Randomized trials studying
various doses and preparations of triamcinolone acetonide in
combination with verteporfin photodynamic therapy are
ongoing.
Evidence demonstrating increased VEGF expression after
PDT provides one explanation for improved results with PDT
combined with triamcinolone compared to PDT alone and
suggests that combining PDT with anti-VEGF agents should
have similar or superior visual results without steroid-related
FIGURE 148.16. Schematic showing process of angiogenesis which
side effects. Trials examining the use of anti-VEGF agents in
involves extracellular matrix degradation, endothelial cell proliferation
combination with verteporfin PDT are ongoing. The FOCUS and migration, tube formation, elongation, and remodeling. Numerous
(RhuFab V2 Ocular Treatment Combining the Use of Visudyne mediators are involved in this cascade providing various targets for
to Examine Safety) study is a Phase I/II trial investigating pharmacotherapy.
the combination of ranibizumab and PDT versus PDT alone
for predominantly classic CNV76. One year results revealed that
90% of patients treated with the combination therapy avoided FUTURE DIRECTIONS
moderate vision loss compared to 68% of patients treated with
PDT alone. The proportion of patients gaining 15 or more NEW TARGETS
letters was 24% in the combination group compared to 5% in
the PDT-alone group. At month 12, a mean of 0.3 additional In addition to targeting VEGF mRNA or protein, the angiogenic
PDT treatments was required in the combination group versus stimulus provided by VEGF may be blocked by interfering
a mean of 2.4 additional treatments in those receiving PDT with the signaling events induced by activated VEGF receptors
alone. (Fig. 148.16). Various protein kinase C (PKC) isoenzymes are
A lyophilized preparation of ranibizumab was used for this activated by VEGFR binding and are important mediators in
study and initially given at day 7 after PDT. When a higher the resulting angiogenic and permeability responses.79 PKC
incidence of uveitis was detected in the combination group, the inhibition has been shown to reduce retinal and CNV in both
timing of the intra-vitreal injection was moved to 30 days after rodent and porcine models.80,81 A PKC beta-inhibitor
PDT. A second Phase II open-label study using the currently ruboxistaurin mesylate (Eli Lilly and Co.) has been shown to
approved preparation of ranibizumab 0.5 mg administered the reduce sustained moderate vision loss in patients with
same day as verteprofin PDT showed no safety concerns at the moderate to severe nonproliferative diabetic retinopathy.82
3 month time point.77 Given the proven benefit of ranibizumab These agents may also prove to be beneficial in neovascular
monotherapy versus PDT for predominantly classic CNV, it AMD.
possible that the favorable results of combination therapy with Similarly, VEGFRs belong to the family of tyrosine kinases
ranibizumab and PDT were mainly due to ranibizumab. and inhibition of this enzymatic function disrupts the down-
Additional trials investigating pegaptanib in combination with stream signaling cascade. Numerous pharmacologic inhibitors
PDT are ongoing. of tyrosine kinases have been synthesized and this class of
A Phase II trial of three doses of squalamine (10, 20, 40 mg) drugs has been used with great success in oncology as aberrant
combined with verteporfin PDT compared to PDT alone in tyrosine kinase activity appears to be pathogenic in certain
45 subjects also suggested an advantage to combination malignancies.83,84 Preclinical data suggest the efficacy of
therapy. At 29 weeks, 90% of patients in the 40 mg + PDT tyrosine kinase inhibitors in models of ocular neovasculari-
group had stable vision (loss of ≤15 letters) compared to 71% zation.85 Vatalanib (PTK787, Novartis Pharmaceuticals) is an
receiving PDT alone. The mean change in visual acuity was a orally administered tyrosine kinase inhibitor with activity
gain of 0.4 letters in the 40 mg combination group versus a loss against all three VEGF receptors as well as PDGFR-b, and
1972 of 4.8 letters in those treated with PDT only.78 c-kit.86 A Phase I/II trial of vatalanib in combination with
Anti-VEGF and Other Pharmacologic Treatments for Age-Related Macular Degeneration

FIGURE 148.17. Results of combination


therapy with anti-VEGF aptamer (pegaptanib)
and anti-PDGFR-b antibody (APB5) in mouse
model of laser-induced CNV. Mice were treated
starting at day 7 post-injury. The combination of
VEGF and PDGF pathway inhibition resulted in
the smallest area of CNV.
Reprinted from Jo N, Mailhos C, Ju M, et al: Inhibition
of platelet-derived growth factor B signaling enhances
the efficacy of anti-vascular endothelial growth factor
therapy in multiple models of ocular
neovascularization. Am J Pathol 2006;
168(6):2036–2053 with permission from the American
Society for Investigative Pathology.

CHAPTER 148
verteporfin PDT in patients with subfoveal choroidal strategies but also in creating improved delivery methods.
neovascularization due to AMD is underway. Repeated intra-vitreal injections present a disadvantage in
Although VEGF certainly plays a causal role in ocular neovas- terms of risk of endophthalmitis and high frequency of
cularization, other molecules also modulate the angiogenic treatments. Systemic delivery of agents allows for bilateral
stimulus in the eye. The Tie (Tyrosine kinase with Immuno- treatment but also substantially increases the potential for
globulin and Epidermal growth factor homology) receptors are nonocular toxicities. The juxtascleral administration of
also tyrosine kinase receptors expressed on endothelial cells anecortave acetate given at 6 month intervals offers an
which play a role in vascular development.87 Their ligands advantage with regard to safety and convenience, and thereby
are the angiopoietins, of which there are at least four.57 The justifies its potential use in the prevention as well as treatment
interaction of Tie-2 and angiopoietin-1 (Ang-1) is thought to of advanced AMD (Fig. 148.18).
induce maturation of vessels by promoting the association of The development of sustained-release delivery platforms for
pericytes with endothelial cells. Ang-2 serves as an antagonist, intra vitreal or transcleral administration will provide an
destabilizing vessels and allowing endothelial cells to respond to increased level of safety and perhaps also efficacy as long-term,
angiogenic stimuli.88 Increased expression of Ang-2 has been continuous levels of therapeutic agents will be achieved. A
demonstrated in mouse models of retinal neovascularization89 fluocinolone acetonide implant (Retisert, Bausch and Lomb,
and blockade of Tie-2 can inhibit experimental retinal Inc.) is currently available for the treatment of chronic, non-
neovascularization.90 These molecules may serve as future infectious posterior uveitis,93 and injectable forms of both fluo-
targets in the development of antiangiogenic drugs. cinolone and dexamethasone intravitreal implants are currently
Another factor important for vessel maturation is platelet- being investigated in clinical trials. The transcleral delivery of
derived growth factor-B (PDGF-B), which has been shown to pegaptanib via poly(lactic-co-glycolic)acid (PLGA) microspheres
play a critical role in the recruitment of mural cells to endo- has been studied in a rabbit model.94 Biomechanical devices
thelial cells.91 Preclinical studies in several models of ocular for transscleral delivery are also under development. These
angiogenesis have demonstrated that inhibition of PDGF-B in types of delivery methods may also allow for multiple pharma-
addition to VEGF-A appears to be more efficacious at preventing cologic agents to be delivered simultaneously, easily enabling
and regression ocular neovascularization than inhibition of combination therapy.
VEGF-A alone92 (Fig. 148.17). These data suggest that pericyte The future of pharmacologic therapy for AMD appears
loss induced by PDGF-B blockade destabilizes neovasculature promising. An array of drugs is in development and in ongoing
and increases susceptibility to anti-VEGF treatment. Such clinical trials. Our therapeutic approach will likely become
combination therapy may lead to improved visual outcomes for multifaceted as treatment options increase, and combinations
a broader spectrum of patients. of various agents lead to offer improved visual outcomes. The
identification and treatment of patients at highest risk for
vision loss early in the disease process is a goal that may be
DRUG DELIVERY realized soon. Continued progress in the understanding and
The challenge of developing new pharmacologic agents for management of macular degeneration will one day reduce the
the treatment of AMD lies not only in identifying efficacious blinding impact of this disease. 1973
RETINA AND VITREOUS

FIGURE 148.18. The posterior juxtascleral


injection procedure used for delivery of
anaecortave acetate.
From D’Amico DJ, Goldberg MF, Hudson H, et al:
Anecortave acetate as monotherapy for treatment of
subfoveal neovascularization in age-related macular
degeneration: twelve-month clinical outcomes.
Ophthalmology 2003; 110:2372–2383; discussion
2384–2375.
SECTION 10

REFERENCES
1. Ferrara N: VEGF and the quest for tumour 12. Shweiki D, Itin A, Soffer D, Keshet E: 21. Baffi J, Byrnes G, Chan CC, Csaky KG:
angiogenesis factors. Nat Rev Cancer Vascular endothelial growth factor induced Choroidal neovascularization in the rat
2002; 2:795–803. by hypoxia may mediate hypoxia-initiated induced by adenovirus mediated
2. Ide AG, Baker NH, Warren SL: angiogenesis. Nature 1992; 359:843–845. expression of vascular endothelial growth
Vascularization of the Brown Pearce rabbit 13. Miller JW, Adamis AP, Shima DT, et al: factor. Invest Ophthalmol Vis Sci 2000;
epithelioma transplant as seen in the Vascular endothelial growth factor/vascular 41:3582–3589.
transparent ear chamber. Am J Roentgenol permeability factor is temporally and 22. Lebherz C, Maguire AM, Auricchio A, et al:
1939; 42:891–899. spatially correlated with ocular Nonhuman primate models for diabetic
3. Michaelson IC: The mode of development angiogenesis in a primate model. Am J ocular neovascularization using AAV2-
of the vascular system of the retina with Pathol 1994; 145:574–584. mediated overexpression of vascular
some observations on its significance for 14. Pierce EA, Avery RL, Foley ED, et al: endothelial growth factor. Diabetes 2005;
certain retinal disorders. Trans Ophthalmol Vascular endothelial growth factor/vascular 54:1141–1149.
Soc UK 1948; 68:137–180. permeability factor expression in a mouse 23. Cui JZ, Kimura H, Spee C, et al: Natural
4. Folkman J. Tumor angiogenesis: model of retinal neovascularization. Proc history of choroidal neovascularization
therapeutic implications. N Engl J Med Natl Acad Sci USA 1995; 92:905–909. induced by vascular endothelial growth
1971; 285:1182–1186. 15. Tolentino MJ, Miller JW, Gragoudas ES, et factor in the primate. Graefes Arch Clin Exp
5. Senger DR, Galli SJ, Dvorak AM, et al: al: Vascular endothelial growth factor is Ophthalmol 2000; 238:326–333.
Tumor cells secrete a vascular permeability sufficient to produce iris neovascularization 24. Yi X, Ogata N, Komada M, et al: Vascular
factor that promotes accumulation of and neovascular glaucoma in a nonhuman endothelial growth factor expression in
ascites fluid. Science 1983; 219: primate. Arch Ophthalmol 1996; choroidal neovascularization in rats.
983–985. 114:964–970. Graefes Arch Clin Exp Ophthalmol 1997;
6. Ferrara N, Henzel WJ: Pituitary follicular 16. Tolentino MJ, Miller JW, Gragoudas ES, et 235:313–319.
cells secrete a novel heparin-binding al: Intravitreous injections of vascular 25. Husain D, Ryan AM, Cuthbertson RA, et al:
growth factor specific for vascular endothelial growth factor produce retinal Vascular endothelial growth factor (VEGF)
endothelial cells. Biochem Biophys Res ischemia and microangiopathy in an adult expression is correlated with choroidal
Commun. Jun 15 1989; 161:851–858. primate. Ophthalmology 1996; neovascularization in a monkey model
7. Keck PJ, Hauser SD, Krivi G, et al: Vascular 103:1820–1828. [ARVO Abstract]. Invest Ophthalmol Vis Sci
permeability factor, an endothelial cell 17. Adamis AP, Shima DT, Tolentino MJ, et al: 1997; 38:S501.
mitogen related to PDGF. Science 1989; Inhibition of vascular endothelial growth 26. Krzystolik MG, Afshari MA, Adamis AP, et
246:1309–1312. factor prevents retinal ischemia-associated al: Prevention of experimental choroidal
8. Leung DW, Cachianes G, Kuang WJ, etal: iris neovascularization in a nonhuman neovascularization with intravitreal anti-
Vascular endothelial growth factor is a primate. Arch Ophthalmol 1996; 114:66–71. vascular endothelial growth factor antibody
secreted angiogenic mitogen. Science 18. Aiello LP, Pierce EA, Foley ED, et al: fragment. Arch Ophthalmol 2002;
1989; 246:1306–1309. Suppression of retinal neovascularization in 120:338–346.
9. Ferrara N, Gerber HP, LeCouter J: The vivo by inhibition of vascular endothelial 27. Kwak N, Okamoto N, Wood JM,
biology of VEGF and its receptors. Nat Med growth factor (VEGF) using soluble VEGF- Campochiaro PA: VEGF is major stimulator
2003; 9:669–676. receptor chimeric proteins. Proc Natl Acad in model of choroidal neovascularization.
10. Ishida S, Usui T, Yamashiro K, et al: Sci USA 1995; 92:10457–10461. Invest Ophthalmol Vis Sci 2000;
VEGF164-mediated inflammation is 19. Adamis AP, Miller JW, Bernal MT, et al: 41:3158–3164.
required for pathological, but not Increased vascular endothelial growth 28. Frank RN, Amin RH, Eliott D, et al: Basic
physiological, ischemia-induced retinal factor levels in the vitreous of eyes with fibroblast growth factor and vascular
neovascularization. J Exp Med 2003; proliferative diabetic retinopathy. Am J endothelial growth factor are present in
198:483–489. Ophthalmol 15 1994; 118:445–450. epiretinal and choroidal neovascular
11. Plate KH, Breier G, Weich HA, Risau W: 20. Aiello LP, Avery RL, Arrigg PG, et al: membranes. Am J Ophthalmol 1996;
Vascular endothelial growth factor is a Vascular endothelial growth factor in ocular 122:393–403.
potential tumour angiogenesis factor in fluid of patients with diabetic retinopathy 29. Lopez PF, Sippy BD, Lambert HM, et
human gliomas in vivo. Nature 1992; and other retinal disorders. N Engl J Med al:Transdifferentiated retinal pigment
1974 359:845–848. 1994; 331:1480–1487. epithelial cells are immunoreactive for
Anti-VEGF and Other Pharmacologic Treatments for Age-Related Macular Degeneration

vascular endothelial growth factor in 42. Avery RL, Pieramici DJ, Rabena MD, et al: 57. Sills AK, Jr., Williams JI, Tyler BM, et al:
surgically excised age-related macular Intravitreal bevacizumab (Avastin) for Squalamine inhibits angiogenesis and solid
degeneration-related choroidal neovascular neovascular age-related macular tumor growth in vivo and perturbs
membranes. Invest Ophthalmol Vis Sci degeneration. Ophthalmology 2006; embryonic vasculature. Cancer Res 1998;
1996; 37:855–868. 113:363–372 e365. 58:2784–2792.
30. Gragoudas ES, Adamis AP, Cunningham 43. Rich RM, Rosenfeld PJ, Puliafito CA, et al: 58. Chen Q, Williams JI, Anderson MG, et al:
ET, Jr, et al: Pegaptanib for neovascular Short-term safety and efficacy of Identification of the intracellular target in
age-related macular degeneration. N Engl J intravitreal bevacizumab (Avastin) for endothelial cells for squalamine as
Med 2004; 351:2805–2816. neovascular age-related macular calmodulin. Available:
31. VISION Clinical Trial Group:Year 2 efficacy degeneration. Retina 2006; 26:495–511. http://www.genaera.com/abstracts/
results of 2 randomized controlled clinical 44. Spaide RF, Laud K, Fine HF, et al: 2000_pas_03.html.
trials of Pegaptanib for neovascular age- Intravitreal bevacizumab treatment of 59. Garcia CA, Quiroz-Mercado H, Uwaydat S,
related macular degeneration. choroidal neovascularization secondary to et al: A Phase I/II Trial of Intravenous
Ophthalmology 2006; 113:1508e–1508.e25. age-related macular degeneration. Retina Squalamine Lactate for Treatment of
32. D’Amico DJ, Patel M, Adamis AP, et al: 2006; 26:383–390. Choroidal Neovascularization in Age
Pegaptanib sodium for neovascular age- 45. Bashshur ZF, Bazarbachi A, Schakal A, et Related Macular Degeneration (ARMD).
related macular degeneration: two-year al: Intravitreal bevacizumab for the Invest Ophthalmol Vis Sci 2004; 45: ARVO
safety results of the two prospective, management of choroidal E-abstract 2362.
multicenter, controlled clinical trials. neovascularization in age-related macular 60. Ciulla TA, Criswell MH, Danis RP, et al:
Ophthalmology 2006; 113:992–1001, degeneration. Am J Ophthalmol 2006; Squalamine lactate reduces choroidal

CHAPTER 148
e1001–e1006. 142:1–9. neovascularization in a laser-injury model in
33. Hurwitz H, Fehrenbacher L, Novotny W, et 46. Bakri SJ, Cameron JD, McCannel CA, et al: the rat. Retina 2003; 23:808–814.
al: Bevacizumab plus irinotecan, Absence of histologic retinal toxicity of 61. Genaidy M, Kazi AA, Peyman GA, et al:
fluorouracil, and leucovorin for metastatic intravitreal bevacizumab in a rabbit model. Effect of squalamine on iris
colorectal cancer. N Engl J Med 2004; Am J Ophthalmol 2006; 142:162–164. neovascularization in monkeys. Retina
350:2335–2342. 47. Shahar J, Avery RL, Heilweil G, et al: 2002; 22:772–778.
34. Kabbinavar F, Hurwitz HI, Fehrenbacher L, Electrophysiologic and retinal penetration 62. Genaera Corp: Genaera announces interim
et al: Phase II, randomized trial comparing studies following intravitreal injection of Evizon Phase II (MSI-1256F–209) data.
bevacizumab plus fluorouracil bevacizumab (Avastin). Retina 2006; Available: http://www.genaera.com/
(FU)/leucovorin (LV) with FU/LV alone in 26:262–269. pressreleases/March%201,%202006_(2).pdf
patients with metastatic colorectal cancer. 48. Marano RJ, Rakoczy PE: Treatments for Mar 1, 2006.
J Clin Oncol 2003; 21:60–65. choroidal and retinal neovascularization: a 63. Dawson DW, Volpert OV, Gillis P, et al:
35. Rosenfeld PJ, Brown DM, Heier JS et al. focus on oligonucleotide therapy and Pigment epithelium-derived factor: a potent
Marina Study Group. Ranibizumab for delivery for the regulation of gene function. inhibitor of angiogenesis. Science 1999;
neovascular age-related macular Clin Experiment Ophthalmol 2005; 285:245–248.
degeneration. N Engl J Med 2006; 33:81–89. 64. Spranger J, Osterhoff M, Reimann M, et al:
355:1419–1431. 49. Acuity Pharmaceuticals. Acuity Loss of the antiangiogenic pigment
36. Brown DM, Kaiser PK, Michels M et al.; Pharmaceuticals reports positive initial epithelium-derived factor in patients with
ANCHOR Study Group. Ranibizumab Phase II results for bevasiranib (Cand5) in angiogenic eye disease. Diabetes 2001;
versus verteporfin for neovascular age- wet AMD. Available: 50:2641–2645.
related macular degeneration. N Engl J http://www.acuitypharma.com/press/releas 65. Tombran-Tink J, Barnstable CJ: PEDF: a
Med. 2006; 355:1432–1444. e13.pdf Jun 1, 2006. multifaceted neurotrophic factor. Nat Rev
37. Brown DM, Shapiro H, Schneider S, Group 50. Sirna Therapeutics. Available: Neurosci 2003; 4:628–636.
AS: Subgroup analysis of first-year results http://www.sirna.com/wt/page/ocular Aug 66. Young TA, Nakazawa T, Sobrin L, et al:
of ANCHOR: a Phase III double-masked, 4, 2006. Modulation of apoptosis following
randomized comparison of ranibizumab 51. Holash J, Davis S, Papadopoulos N, et al: combination PEDF and photodynamic
and verteporfin photodynamic therapy for VEGF-Trap: a VEGF blocker with potent therapy for choroidal neovascularization in
predominantly classic choroidal antitumor effects. Proc Natl Acad Sci USA the rat model. Invest Ophthalmol Vis Sci
neovascularization related to age-related 2002; 99:11393–11398. 2004; 45:ARVO E -abstract 2231.
macular degeneration. Invest Ophthalmol 52. Nguyen QD, Shah SM, Browning D, et al: 67. Mori K, Gehlbach P, Ando A, et al:
Vis Sci 2006; 47:ARVO E-abstract 2963. Results of a Phase I, dose-escalation, Regression of ocular neovascularization
38. Genentech, Inc: Preliminary results from a safety, tolerability, and bioactivity study of in response to increased expression of
Phase IIIb study showed patients with wet intravitreous VEGF Trap in patients with pigment epithelium-derived factor. Invest
AMD treated with Lucentis quarterly neovascular age-related macular Ophthalmol Vis Sci 2002; 43:2428–2434.
experienced a 16-letter benefit over the degeneration. Invest Ophthalmol Vis Sci. 68. Mori K, Gehlbach P, Yamamoto S, et al:
control group at one year. Available: 2006; 47:ARVO E-abstract 2144. AAV-mediated gene transfer of pigment
http://www.gene.com/gene/news/press- 53. Das A, McGuire PG. Retinal and choroidal epithelium-derived factor inhibits choroidal
releases/display.do?method=detail&id=974 angiogenesis: pathophysiology and neovascularization. Invest Ophthalmol Vis
7 2 May 2006. strategies for inhibition. Prog Retin Eye Res Sci 2002; 43:1994–2000.
39. Fung AE, Lalwani GA, Rosenfeld PJ et al. 2003; 22:721–748. 69. Saishin Y, Silva RL, Saishin Y, et al:
An optical coherence tomography-guided, 54. Penn JS, Rajaratnam VS, Collier RJ, Clark Periocular gene transfer of pigment
variable dosing regimen with intravitreal AF: The effect of an angiostatic steroid on epithelium-derived factor inhibits choroidal
ranibizumab (Lucentis) for neovascular neovascularization in a rat model of neovascularization in a human-sized eye.
age-related macular degeneration. retinopathy of prematurity. Invest Hum Gene Ther 2005; 16:473–478.
Am J Ophthalmol 2007; 143:566–583 Ophthalmol Vis Sci 2001; 42:283–290. 70. Campochiaro PA, Nguyen QD, Shah SM,
40. Mordenti J, Cuthbertson RA, Ferrara N, et 55. D’Amico DJ, Goldberg MF, Hudson H, et al: et al: Adenoviral vector-delivered pigment
al: Comparisons of the intraocular tissue Anecortave acetate as monotherapy for epithelium-derived factor for neovascular
distribution, pharmacokinetics, and safety treatment of subfoveal neovascularization age-related macular degeneration: results
of 125I-labeled full-length and Fab in age-related macular degeneration: of a Phase I clinical trial. Hum Gene Ther
antibodies in rhesus monkeys following twelve-month clinical outcomes. 2006; 17:167–176.
intravitreal administration. Toxicol Pathol Ophthalmology 2003; 110:2372–2383; 71. Ciulla TA, Walker JD, Fong DS, Criswell
1999; 27:536–544. discussion 2384–2375. MH: Corticosteroids in posterior segment
41. Michels S, Rosenfeld PJ, Puliafito CA, et al: 56. Slakter JS, Bochow TW, D’Amico DJ, et al: disease: an update on new delivery
Systemic bevacizumab (Avastin) therapy for Anecortave acetate (15 milligrams) versus systems and new indications. Curr Opin
neovascular age-related macular photodynamic therapy for treatment of Ophthalmol 2004; 15:211–220.
degeneration twelve-week results of an subfoveal neovascularization in age-related 72. Gillies MC, Simpson JM, Luo W, et al: A
uncontrolled open-label clinical study. macular degeneration. Ophthalmology randomized clinical trial of a single dose of
Ophthalmology 2005; 112:1035–1047. 2006; 113:3–13. intravitreal triamcinolone acetonide for 1975
RETINA AND VITREOUS

neovascular age-related macular 79. Donnelly R, Idris I, Forrester JV: Protein 87. Sato TN, Tozawa Y, Deutsch U, et al:
degeneration: one-year results. Arch kinase C inhibition and diabetic Distinct roles of the receptor tyrosine
Ophthalmol 2003; 121:667–673. retinopathy: a shot in the dark at kinases Tie-1 and Tie-2 in blood vessel
73. Jonas JB, Degenring RF, Kreissig I, et al: translational research. Br J Ophthalmol formation. Nature 1995; 376:70–74.
Exudative age-related macular 2004; 88:145–151. 88. Hanahan D: Signaling vascular
degeneration treated by intravitreal 80. Fabbro D, Ruetz S, Bodis S, et al: morphogenesis and maintenance. Science
triamcinolone acetonide. A prospective PKC412—a protein kinase inhibitor with a 1997; 277:48–50.
comparative nonrandomized study. Eye broad therapeutic potential. Anticancer 89. Hackett SF, Ozaki H, Strauss RW, et al:
2005; 19:163–170. Drug Des 2000; 15:17–28. Angiopoietin 2 expression in the retina:
74. Spaide RF, Sorenson J, Maranan L: 81. Saishin Y, Silva RL, Saishin Y, et al: upregulation during physiologic and
Photodynamic therapy with verteporfin Periocular injection of microspheres pathologic neovascularization. J Cell
combined with intravitreal injection of containing PKC412 inhibits choroidal Physiol 2000; 184:275–284.
triamcinolone acetonide for choroidal neovascularization in a porcine model. 90. Hangai M, Moon YS, Kitaya N, et al:
neovascularization. Ophthalmology 2005; Invest Ophthalmol Vis Sci 2003; Systemically expressed soluble Tie2
112:301–304. 44:4989–4993. inhibits intraocular neovascularization. Hum
75. Augustin AJ, Schmidt-Erfurth U: Verteporfin 82. PKC-DMES Study Group. Effect of Gene Ther 2001; 12:1311–1321.
therapy combined with intravitreal ruboxistaurin in patients with diabetic 91. Hellstrom M, Kalen M, Lindahl P, et al:
triamcinolone in all types of choroidal macular edema: thirty-month results of Role of PDGF-B and PDGFR-beta in
neovascularization due to age-related the randomized PKC-DMES clinical recruitment of vascular smooth muscle
macular degeneration. Ophthalmology trial. Arch Ophthalmol 2007; cells and pericytes during embryonic blood
2006; 113:14–22. 125:318–324. vessel formation in the mouse.
SECTION 10

76. Heier JS, Boyer DS, Ciulla TA et al.; 83. Maki RG. Gastrointestinal Stromal Tumors Development 1999; 126:3047–3055.
FOCUS Study Group. Ranibizumab Respond to Tyrosine Kinase-targeted 92. Jo N, Mailhos C, Ju M, et al: Inhibition of
combined with verteporfin photodynamic Therapy. Curr Treat Options Gastroenterol platelet-derived growth factor B signaling
therapy in neovascular age-related macular 2004; 7:13–17. enhances the efficacy of anti-vascular
degeneration: year 1 results of the FOCUS 84. O’Brien SG, Guilhot F, Larson RA, et al: endothelial growth factor therapy in
Study. Arch Ophthalmol 2006; Imatinib compared with interferon and multiple models of ocular
124:1532–1542. low-dose cytarabine for newly diagnosed neovascularization. Am J Pathol 2006;
77. Schmidt-Erfurth UM, Gabel P, Hohman T, chronic-phase chronic myeloid 168:2036–2053.
Group PS. Preliminary results from an leukemia. N Engl J Med 2003; 93. Jaffe GJ, Martin D, Callanan D, et al:
open-label, multicenter, Phase II study 348:994–1004. Fluocinolone acetonide implant (Retisert)
assessing the effects of same-day 85. Unsoeld AS, Junker B, Mazitschek R, et al: for noninfectious posterior uveitis:
administration of ranibizumab (Lucentis) Local injection of receptor tyrosine kinase Thirty-four week results of a multicenter
and verteporfin PDT (PROTECT study). inhibitor MAE 87 reduces retinal randomized clinical study. Ophthalmol
Invest Ophthalmol Vis Sci 2006; 47:ARVO neovascularization in mice. Mol Vis 2004; 2006; 113:1020–1027.
E-abstract 2960. 10:468–475. 94. Carrasquillo KG, Ricker JA, Rigas IK,et al:
78. Genaera Corp: Genaera presents positive 86. Thomas AL, Morgan B, Horsfield MA, Controlled delivery of the anti-VEGF
prelimiary clinical results for Evizon for et al: Phase I study of the safety, aptamer EYE001 with poly(lactic-co-
treatment of age-related macualr tolerability, pharmacokinetics, and glycolic)acid microspheres. Invest
degeneration at the annual AAO meeting. pharmacodynamics of PTK787/ZK 222584 Ophthalmol Vis Sci 2003; 44:290–299.
Available: http://www.genaera.com/ administered twice daily in patients with
pressreleases/October%2019,%202005.pdf advanced cancer. J Clin Oncol 2005;
Oct 19, 2005. 23:4162–4171.

1976
CHAPTER
Surgical Treatments of Neovascular Age-Related
149 Macular Degeneration
Sunil K. Srivastava and Cynthia A. Toth

Overview Major Complications


Surgical therapy for neovascular age-related macular Full Macular Translocation
degeneration (AMD) continues to evolve with refinements in Retinal detachment
technique and integration with developments in pharmacologic Proliferative vitreoretinopathy
therapy. Currently available surgical treatments for neovascular Recurrence of CNV or subretinal hemorrhage
AMD may provide useful options for salvage of vision when Cystoid macular edema
medical treatments are not enough. Some of the techniques Hypotony
discussed such as subretinal hemorrhage displacement, and Glaucoma
macular translocation with 360-degree retinectomy may be Epiretinal membrane
effective in restoring vision in patients with lesions not generally Keratopathy
treatable with medical therapy. Other therapies such as RPE Note that many of the more common, but not all, complications reported with
transplantation are in early stages of promising development. The each surgery are included on this list.
indications and techniques and evolution of the surgical
therapies for AMD are discussed in this chapter.
SUBMACULAR SURGERY
The treatment of neovascular age-related macular degeneration
(AMD) has included laser photocoagulation, photodynamic The goal of submacular surgery is to remove the choroidal
therapy, radiation, use of pharmacologic agents, and in some neovascularization (CNV) and hemorrhage if present and
cases surgical intervention. Surgical therapies have been to re-establish the normal anatomy between retina and RPE.
utilized primarily for neovascular AMD and also in few cases The poor visual outcomes associated with the Macular
of geographic atrophy. The surgeries for AMD presented here: Photocoagulation Study and the encouraging results in early
submacular surgery, hemorrhage displacement, limited or full submacular surgery case series generated interest in these
macular translocation, and transplantation of retinal pigment procedures.1–3
epithelium (RPE) will continue to evolve, affected by the rapid The surgeon considers lesion size and location when
evolution of novel pharmacologic therapies. determining a site for CNV removal through a posterior retino-
tomy. During vitrectomy the posterior hyaloid is separated. A
retinotomy is created and balanced salt solution is injected
Major Complications beneath the retina or a pick may be passed over the CNV to
Submacular Surgery separate retinal connections. The surface of the membrane is
Retinal detachment grasped with forceps and it is removed from the subretinal space
Proliferative vitreoretinopathy (Fig. 149.1). If the membrane is not free, attachments are cut
Vitreous hemorrhage with a blade or scissors. Most importantly, intraocular pressure
Cataract formation
is elevated prior to and during removal of the CNV, then
Persistence or recurrence of CNV
lowered once hemostasis is assured. For subretinal hemorrhage
Displacement of Subretinal Hemorrhage (clot) removal, subretinal tissue plasminogen activator (tPA)
Retinal detachment
12.5–50 mg is sometimes injected for thrombolysis 30–45 min
Proliferative vitreoretinopathy
Vitreous hemorrhage
prior to hemorrhage removal. Blood is then aspirated or pulled
Cataract formation from the subretinal space. A fluid to air or gas exchange is
Recurrence of subretinal hemorrhage performed and the patient is positioned upright or face down for
Persistence or recurrence of CNV the first postoperative day.2–7
Limited Macular Translocation
The Submacular Surgery Trials (SSTs), randomized prospec-
Retinal detachment tively clinical trials, compared submacular surgery to obser-
Proliferative vitreoretinopathy vation for two groups of AMD patients, those with large or
Recurrence of CNV after laser or other therapy poorly demarcated, new subfoveal CNV (group N)6 and those
Cataract formation with predominantly hemorrhagic lesions (group B).7 For group
Macular fold N, at 2 years the surgery and observation groups had no
Macular hole significant difference in results. A successful outcome (defined
Subretinal hemorrhage as <1 line best corrected vision loss) occurred in 41% and 44%,
Scleral perforation respectively. In both groups at 24 months there was a 2 line
1977
RETINA AND VITREOUS

DISPLACEMENT OF SUBMACULAR
IOP elevated during CNV removal Subretinal HEMORRHAGE
forceps
Retinotomy away from fovea The prognosis for visual acuity is poor for submacular
Grasp CNV Retina hemorrhage associated with neovascular AMD,10–12 especially
beneath retina with larger and thicker hemorrhages which cause photoreceptor
damage from clot contraction, direct iron toxicity, and the
blockade of metabolic products exchange.13,14 In light of the
potential for damage to retina and RPE during clot extraction,
even with tPA, Heriot (presentation at American Academy of
Ophthalmology Vitreoretinal Update, San Francisco, CA, 1997)
proposed subretinal hemorrhage displacement. He described
CNV RPE injection of tPA into the vitreous cavity, followed by injection
of a gas bubble with the goal of clot lysis and then displacement
Avoid RPE-Bruchís damage by with positioning. Although subretinal hemorrhage was suc-
grasping anterior surface of CNV cessfully displaced using this method, with some improvement
in visual acuity,15,16 it was unclear whether intravitreal tPA
reached the subretinal space, since others displaced blood with
SECTION 10

FIGURE 149.1. Submacular surgery technique for removal of intravitreal injections of gas alone and prone positioning.17
choroidal neovascularization. The focal retinotomy is located away
Alternately, Haupert et al described successful hemorrhage
from the fovea with attention to limiting subretinal instrument contact
with retinal pigment epithelium and choroid. displacement after pars plana vitrectomy with subretinal tPA
injection followed by fluid to air or gas exchange and prone
postoperative positioning in 11 patients.18 Two series followed
with a similar technique that added postoperative supine
median vision loss and median acuity dropped from 20/100 to positioning for the first hour and head down19 or upright
20/400. Analysis of vision-related quality of life scores revealed positioning for blood displacement (Fig. 149.2).20 Patients in all
higher scores in the SST surgery group; however, based on three series had either partial or complete displacement of
the visual acuity findings, submacular surgery could not be the blood from the subfoveal space with early vision improve-
recommended for patients with AMD and new subfoveal ment in the majority of eyes. With follow-up averaging from 3
lesions. Given these results, submacular surgery is currently to 17 months, hemorrhage recurred in 4–28% of eyes. Unlike
not indicated for the treatment of CNV associated with AMD. surgery for removal of subretinal hemorrhage, there was only
one retinal detachment in the 57 eyes in these three series.18–20
Key Feature Final visual acuity decreased compared to best postoperative
Submacular surgery trials (SST) showed no visual benefit with acuity though some patients received photodynamic therapy or
surgery when compared to observation for patients with new pegaptanib sodium injection.19,20 These studies are limited by
subfoveal CNV secondary to AMD. For predominantly hemorrhagic their small numbers and lack of control group (Fig. 149.3).
lesions secondary to AMD the SST found a reduced risk of severe In surgical therapy of subretinal hemorrhage, the earlier
vision loss with surgery when compared to observation, but a high submacular surgical techniques led to less invasive approaches
risk of rhegmatogenous retinal detachment in the surgery group. with the addition of adjuvants such as photodynamic or anti-
VEGF therapy after hemorrhage displacement. Submacular
In SST group B (hemorrhagic lesions) the surgical technique surgery in randomized trials showed no significant improve-
was similar to that described above, with both blood and CNV
removed whenever possible.7 The surgeon chose whether to
use tPA (used in 38% of Group B cases). At 24 months, the
majority, 56% of surgery eyes and 59% of observation eyes, Subfoveal blood After subretinal tPA and
experienced loss of >2 lines acuity, and 18% of eyes in each before displacement fluid-air exchange
group improved at least 2 lines. In contrast to earlier case series
which had shown a trend toward better visual acuity when
subretinal blood was removed within 1 week,5 the SST did not
show improvement in acuity in the surgery group compared
to observation; however, there was no cutoff with regards to the
length of time that the hemorrhage was present, nor was there
documentation of good visual acuity before the hemorrhage.
Compared to the observation group, surgery eyes had higher
rates of retinal detachments (16% vs 2%) and of cataract surgery
(44% vs 6%), but a lower rate of severe vision loss (greater than
6 lines) (21% vs 36%).7 It is difficult to recommend submacular
surgery for submacular hemorrhage in AMD given the high rates Fovea Vitreous gel Fovea Displaced Air
of complications and the pressure of other treatment options. blood
The observation of poor vision after intervention in both of
these AMD surgery groups in the SST is thought likely due to
FIGURE 149.2. Illustration of hemorrhage displacement after injection
damage to the retina and underlying RPE by CNV, hemorrhage,
of subretinal tPA, fluid–air exchange and upright positioning.
surgical manipulation and postoperative absence of RPE in Hemorrhage displacement with upright positioning based on research
the surgical bed.8,9 The development of alternate therapies for and diagrams presented by Marcin Stopa, MD, Duke University Eye
neovascular AMD has further diminished interest in submacular Center Resident’s Day, June 24, 2005.
surgery alone.
1978
Surgical Treatments of Neovascular Age-Related Macular Degeneration

a b c

CHAPTER 149
d e f

FIGURE 149.3. Subretinal hemorrhage displacement. (a) Preoperative color fundus photograph
and (b) late fluorescein angiogram of an 8-day-old subretinal hemorrhage. Preoperative distance
visual acuity was 20/400 in this eye with no previous treatment. (c) Color fundus photograph and
(d) early and (e) late fluorescein angiograph 1 week after hemorrhage displacement by vitrectomy,
subretinal tPA injection and fluid–air exchange. At this exam the patient received photodynamic
therapy and subsequently this eye was treated with two injections of intravitreal bevacizumab.
Color fundus photograph (f) and late fluorescein angiogram (g) 9 months later when visual acuity
was 20/400.

ment compared to observation. Though the rate of severe vision healthier site of RPE and choriocapillaris to prevent further
loss was lower with surgical removal of hemorrhage than with degeneration and possibly recover vision. Since the initial report
observation, the rate of retinal detachment was high. Given the of Machemer,22 the technique of macular translocation has
poor natural history of submacular hemorrhage, and the low evolved into two techniques: limited macular translocation
rate of complications after intravitreal or subretinal injection of (LMT) or full macular translocation with 360o retinectomy
tPA and air or gas displacement, this therapy is a reasonable (MT360, also known as retinal rotation).
consideration, though timing and coordination with use of
adjuvant anti-VEGF or other therapy awaits results of further
study. A meta-analysis of submacular surgery procedures LIMITED MACULAR TRANSLOCATION
provides a useful background for this decision making, though LMT relocates the retina by pinning detached retina against
techniques such as hemorrhage displacement are considered the eye wall using a partial air or gas fill to create a downward
together with hemorrhage removal in the analysis.21 shift in the detached fovea and inferior retina to a site of
healthier RPE and choriocapillaris; the shift is accentuated by
MACULAR TRANSLOCATION also folding the choroid and sclera in the superotemporal
quadrant in the bed of the retinal detachment.23,24 Horizontal
In contrast to submacular surgery or hemorrhage displacement, mattress sutures preplaced in the superior temporal sclera are
macular translocation surgery seeks to relocate the macula to a tied to infold the sclera only after completion of vitrectomy and
1979
RETINA AND VITREOUS

Scleral imbrication Translocation during upright


sutures positioning after surgery

Gas fill 1
2

Retinal fold

Translocation of fovea from


original site (1) to site 2
SECTION 10

FIGURE 149.4. Illustration of limited macular translocation technique


with partial retinal detachment and displacement of subretinal fluid FIGURE 149.5. Microperimetry field with good fixation (pale blue
and of the fovea with partial fluid–gas exchange along with scleral dots) 2 years after limited macular translocation surgery and laser
infolding. treatment of the choroidal neovascular membrane (which became
juxtafoveal after the retina was shifted by the limited macular
translocation surgery).

detachment of temporal retina through injection of subretinal


fluid. Then a partial fluid to air exchange is performed.
Translocation occurs after surgery when the patient is Translocation during surgery
positioned upright. The bubble holds the retina against the Retina is detached and Fovea moved to new site 2
imbrication and subretinal fluid accumulates inferiorly shifting cut 360-degrees at the ora serrata from old site 1
the retina. After reattachment, an inferior retinal fold is seen
early in the postoperative period (Fig. 149.4).
LMT changes the CNV location from subfoveal to juxtafoveal
or extrafoveal, depending on distance of translocation, with Translocation
laser photocoagulation typically applied to the CNV early after 2
surgery (Fig. 149.5).25 Foveal displacement with LMT averages
1200–1600 mm depending on the technique and series. In the 1
largest series reported (102 patients), effective translocation off
the CNV was seen in 60% of patients and mean visual acuity
was unchanged with 40% of eyes gaining >2 Snellen lines, 29%
of eyes unchanged and 31% losing >2 Snellen lines.25 CNV
recurred in 35% of eyes after effective macular translocation and
10 eyes developed retinal detachments. A retrospective com- Subfoveal hemorrhage Hemorrhage removed
with RPE tear
parison of LMT versus photodynamic therapy found comparable
postoperative visual acuities (both 20/200) but vision loss
greater with photodynamic therapy.26 The improvement in new FIGURE 149.6. Illustration summarizing macular translocation with
therapies for subfoveal, small CNV may reduce efforts to relocate 360° retinectomy surgery. The fovea is shifted onto a new RPE bed
CNV to an extrafoveal location for treatment. If small subfoveal during surgery and is held in place with silicone oil during the early
scars persist after alternate therapy and diminish acuity, LMT postoperative period.
might then be considered for relocation of the fovea.

within months. Extraocular muscle surgery is performed at the


FULL MACULAR TRANSLOCATION WITH 360° time of translocation or in a separate surgery.
RETINECTOMY Translocation case series have included subfoveal CNV, RPE
Machemer initially proposed MT360 surgery in the era of tears, disciform scars, hemorrhages, previous photocoagulation,
submacular surgery development. Though recognized as com- and photodynamic therapy. With median follow-up ranging
plex surgery, MT360 allows > 3000 mm foveal movement, from 10 to 21 months, seven of nine studies showed stabili-
enough to translocate off large hemorrhage, large CNV, or zation or improvement of vision after macular translocation
fibrosis. The retinal rotation produces torsional diplopia, surgery29 and distance acuity improved in 52–66% of patients
treated by Eckardt et al with extraocular muscle surgery to in three series.27,28,30,31 At 1 year after MT360, prospective studies
counter-rotate the globe.27 MT360 surgery currently includes a have also shown improvement in reading speed, color vision,
thorough vitrectomy followed by injection of subretinal fluid contrast, near acuity,32 critical print size,33 and in vision-related
to totally detach the retina. The peripheral retina is cut at the quality of life.34
ora serrata, subretinal membrane and/or blood is removed, and Retinal detachment rates after macular translocation were
the retina is translocated, usually superiorly, off the CNV bed highest in early case series (up to 43%).29 Other complications
and pinned in the new location with perfluorocarbon liquid include cystoid macular edema, CNV recurrence, hypotony,
(Fig. 149.6). Retinectomy margins are photocoagulated and the epiretinal membranes, and keratopathy.29 In the meta-analysis
1980 perfluorocarbon is exchanged for silicone oil which is removed of surgery for AMD, estimated rates of complications were
Surgical Treatments of Neovascular Age-Related Macular Degeneration

resulted in good vision recovery in patients after photodynamic


Treatment of Neovascular AMD therapy38 and after anti-VEGF therapy (Fig. 149.8).
1st eye: Is patient eligible for non-surgical treatment ?
Consider No Yes TRANSPLANTATION OF RETINAL PIGMENT
displacement of blood Non-surgical treatment EPITHELIUM
2nd eye: Is patient eligible for non-surgical treatment ? Work on transplantation of healthy RPE into the subfoveal
No space of eyes with AMD has followed from reports of poor
Yes acuity after submacular surgery and of improved acuity after
Start non-surgical treatment
macular translocation onto a healthier RPE bed. The pigment
cell transplantation techniques are combined with conventional
Is impairment visually significant
submacular surgery in eyes with neovascular AMD. In one
and not responding to treatment ?
technique, RPE cells are first harvested from irrigation of the
Yes No nasal subretinal space and are directly transplanted onto
the site of CNV removal.39 Alternately, an adjacent RPE patch
or a choroidal–RPE patch excised from the superior periphery
Consider MT360 Continue other treatment is placed beneath the fovea (Fig. 149.9).40–44 These techniques
or displacement of blood are still early in their evolution and results have been limited

CHAPTER 149
with some notable recovery of acuity despite complications
FIGURE 149.7. Decision-making algorithm for treating AMD with such as graft failure, fibrosis, and retinal detachment. Refine-
vision loss.
ments in the surgery and improvements in the generation/
harvesting of RPE grafts will hopefully make this a viable
highest for macular translocation surgery.21 Despite these option for patients.39–44
complication rates, in a prospective 4 year study of 64 patients,
Toth found that acuity of 20/80 or better was maintained in Key Feature
46–51% of eyes from 1 through 4 years after MT360 with RD In light of the complexity of surgery, macular translocation with
rate below 8% in this series.31 360° peripheral retinectomy could be considered for the second
In light of the risks of complications, and the potential for eye with vision loss from hemorrhagic or neovascular AMD when
vision gain, the decision whether to pursue MT360 is complex. primary therapies fail or are ineffective.
Nonsurgical therapies now display improved results with low
rates of complications.35–37 Thus in cases where patients fail The techniques described have been centered on manage-
primary therapies or when there is no effective treatment ment of neovascular AMD and not geographic atrophy.
MT360 is a reasonable salvage therapy, but one would not likely Although submacular surgery provides no advantage to eyes
consider surgical intervention until medical therapies have with geographic atrophy, macular translocation surgery or
first been utilized (Fig. 149.7), particularly since MT360 has RPE transplantation techniques could be useful in cases of

a b c

d e f

FIGURE 149.8. Macular translocation in an eye that has already received juxtafoveal laser photocoagulation, photodynamic therapy and
pegaptanib injection. (a) Preoperative color fundus photograph, (b) late angiogram and (c) OCT scan of right eye show a fibrotic subfoveal lesion
with almost no leakage. The visual acuity at distance was 20/200, and at near was J16 with +4 add. The patient had received four treatments
with photodynamic therapy in the 20/800 fellow eye. (d) Color fundus photograph, (e) late fluorescein angiographic frame, and (f) OCT after
macular translocation surgery in the right eye. Visual acuity recovered to 20/50 distance and 20/40 near with the patient resuming normal reading
tasks. 1981
RETINA AND VITREOUS

loss of RPE, if the retina and choriocapillaris could maintain


Site of lasered margins function or recover after the surgery. In small series, patients
excised retina RPE and choroid with geographic atrophy have recovered acuity after macular
RPE-choroid transplant
translocation, though later some eyes have developed recurrent
RPE-choroid transplant unfolded under fovea
in bed of removed CNV
geographic atrophy in a pattern echoing the preoperative
(retina discarded)
lesion.45,46 It may become possible to combine surgery –
Forceps CNV bed whether translocation or transplantation – with adjuvant
therapies to prevent recurrent atrophy in the future. Until then,
response to these therapies provides new insight into the pro-
gression of atrophic disease and should be cautiously pursued.
As treatment for neovascular AMD shifts from laser
photocoagulation to intravitreal delivery of antineovascular
drugs, long-term sustained-delivery devices will likely be
developed. Adaptation of current devices which allow sustained
delivery of corticosteroids47–49 or new devices will provide sus-
tained intravitreal delivery of pharmacologic agents for AMD
and reduce the risks associated with multiple intravitreal
injections.
SECTION 10

Enlarged retinotomy Intraoperative temporary The first line of therapy for neovascular AMD primarily
perfluorocarbon bubble involves medical and laser therapy. Surgical techniques such
Bed where CNV removed
transplant after submacular surgery
before fluid-air exchange as hemorrhage displacement, macular translocation, and RPE
cell transplantation play a role in the treatment paradigm
(Fig. 149.9) of neovascular AMD. Developments in surgical
FIGURE 149.9. Illustration of RPE–choroid transplant surgery. technique, adjuvants (such as growth factors), and drug therapy
Choroidal neovascularization is removed as in Figure 149.1. The will have a significant impact on the evolution of future surgical
RPE–choroid transplant is harvested from the mid-periphery, avoiding treatments of AMD.
large choroidal vessels with retina removed from the surface. The
transplant is placed into the subretinal space through an enlarged
retinotomy and is unrolled to flatten beneath the retina and an
intravitreal perfluorocarbon bubble.

REFERENCES
1. Macular Photocoagulation Study Group: membranes. Ophthalmology 1994; plasminogen activator injection and
Laser photocoagulation of subfoveal 101:1099–1111. pneumatic displacement. Ophthalmology
recurrent noevascular lesions in age-related 9. Nasir MA, Sugino I, Zarbin MA: Decreased 1999; 106:1900–1906.
macular degeneration: results of a choriocapillaris perfusion following surgical 17. Daneshvar H, Kertes PJ, Leonard BC,
randomized clinical trial. Arch Ophthalmol excision of choroidal neovascular Peyman G: Management of submacular
1991; 109:1232–1241. rmembranes in age-related macular hemorrhage with intravitreal sulfur
2. Thomas MA, Dickinson JD, Melberg NS, degeneration. Br J Ophthalmol 1997; hexafluoride: a pilot study. Can J
et al. Visual results after surgical removal of 81:481–489. Ophthalmol 1999; 34:385–388.
subfoveal choroidal neovascular membranes. 10. Scupola A, Coscas G, Sourbrane G, 18. Haupert CL, McCuen BW II, Jaffe GJ, et al:
Ophthalmology 1994; 101:1384–1396. Balestrazzi E: Natural history of macular Pars plana vitrectomy, subretinal injection
3. Thomas MA, Grand MG, Williams DF, et al: subretinal hemorrhage in age-related of tissue plasminogen activator and fluid-
Surgical management of subfoveal macular degeneration. Ophthalmologica gas exchange for displacement of thick
choroidal neovascularization. 1999; 213:97–102. submacular hemorrhage in age-related
Ophthalmology 1992; 99:952–968. 11. Avery RL, Fekrat S, Hawkins BS, Bressler macular degeneration. Am J Ophtalmol
4. De Juan E, Machemer R: Vitreous surgery NM: Natural history of subfoveal subretinal 2001; 131:208–215.
for hemorrhagic and fibrous complications hemorrhage in age-related macular 19. Olivier S, Chow DR, Packo KH, et al:
of age-related macular degeneration. Am J degeneration. Retina 1996; 16:183–189. Subretinal recombinant tissue plasminogen
Ophthalmol 1988; 105:25–29. 12. Bennett SR, Folk JC, Blodi CF, Klugman M: activator injection and pneumatic
5. Lewis H: Intraoperative fibrinolysis of Factors prognostic of visual outcome in displacement of thick submacular
submacular hemorrhage with tissue patients with subretinal hemorrhage. Am J hemorrhage in age-related macular
plasminogen activator and surgical drainage. Ophthalmol 1990; 109:33–37. degeneration. Ophthalmology 2004;
Am J Ophthalmol 1994; 5:559–568. 13. Glatt H, Machemer R: Experimental 6:1201–1208. Erratum. Ophthalmology
6. Submacular Surgery Trials Research Group: subretinal hemorrhage in rabbits. Am J 2004; 9:1640.
Surgery for subfoveal choroidal Ophthalmol 1982; 94:762–773. 20. Singh RP, Patel C, Sears JE: Management
neovascularization in age-related macular 14. Toth CA, Morse LS, Hjelmeland LM, Lander of subretinal macular haemorrhage by
degeneration: ophthalmic findings: SST MBD: Fibrin directs early retinal damage direct administration of tissue plasminogen
report no. 11 Ophthalmology 2004; after experimental subretinal hemorrhage. activator. Br J Ophthalmol 2006;
11:1967–1980. Arch Ophthalmol 1991; 109:723–729. 4:429–431.
7. Submacular Surgery Trials Research Group: 15. Schulze SD, Hesse L: Tissue plasminogen 21. Falkner CI, Leitich H, Frommlet F, et al: The
Surgery for hemorrhagic choroidal activator plus gas injection in patients with end of submacular surgery for age-related
neovascular lesions of age-related macular subretinal hemorrhage caused by age- macular degeneration? A meta-analysis.
degeneration: ophthalmic findings: SST related macular degeneration: predictive Graefes Arch Clin Exp Ophthalmol 2007;
report no. 13. Ophthalmology 2004; variables for visual outcome. Graefes Arch 245:490–501.
11:1993–2006. Clin Exp Ophthalmol 2002; 240:717–720. 22. Machemer R, Steinhorst UH: Retinal
8. Grossniklaus HE, Hutchinson AK, Capone A, 16. Hassan AS, Johnson MW, Schneiderman separation, retinotomy and macular
et al: Clinicopathologic features of TE, et al: Management of submacular relocation. II. A surgical approach for age-
1982 surgically excised choroidal neovascular hemorrhage with intravitreous tissue related macular degeneration? Graefes
Surgical Treatments of Neovascular Age-Related Macular Degeneration

Arch Clin Exp Ophthalmol 1993; 32. Toth CA, Lapolice DJ, Banks AD, epithelium translocation in patients with
231:635–641. Stinnett SS: Improvement in near visual subfoveal neovascular membranes. Br J
23. De Juan E, Lowenstein A, Bressler NM, function after macular translocation surgery Ophthalmol 2004; 88:110–113.
et al: Translocation of the retina for with 360o peripheral retinectomy. Graefes 42. Van Meurs JC, Van Den Biesen PR:
management of subfoveal chroidal Arch Clin Exp Ophthalmol 2004; Autologous retinal pigment epithelium and
neovascularization. II: a preliminary report 242:541–548. choroid translocation in patients with
in humans. Am J Ophthalmol 1998; 33. Fujikado T, Asonuma S, Ohji M, et al: exudative age-related macular
125:635–646. Reading ability after macular translocation degeneration: short-term follow-up. Am J
24. Lewis H, Kaiser P, Lewis S, Estafanous M: surgery with 360o retinotomy. Am J Ophthalmol 2003; 136:688–695.
Macular translocation for subfoveal Ophthalmol 2002; 134:849–856. 43. Stanga PE, Kychenthal A, Fitzke FW, et al:
choroidal neovascularization in age-related 34. Cahill MT, Stinnett SS, Banks AD, et al: Retinal pigment epithelium translocation
macular degeneration: a prospective study. Quality of life after macular translocation after choroidal neovascular membrane
Am J Ophthalmol 1999; 128:135–146. with 360 degree peripheral retinectomy for removal in age-related macular
25. Fujii GY, De Juan E, Pieramici DJ, et al: age related macular degeneration. degeneration. Ophthalmology 2002;
Inferior limited macular translocation for Ophthalmology 2005; 112:144–151. 109:1492–1498.
subfoveal chroidal neovascularization 35. Blinder KJ, Bradley S, Bressler NM, et al: 44. MacLaren RE, Bird AC, Sathia PJ,
secondary to age-related macular Effect of lesion size, visual acuity, and Awylard GW: Long-term results of
degeneration: 1 year visual outcome and lesion composition on visual acuity change submacular surgery combined with macular
recurrence report. Am J Ophthalmol 2002; with and without verteporfin therapy for translocation of the retinal pigment
134:69–74. choroidal neovascularization secondary to epithelium in neovascular age-related

CHAPTER 149
26. Pawlak D, Glacet–Bernard A, Papp M, et al: age-related macular degeneration: TAP and macular degeneration. Ophthalmology
Limited macular translocation compared VIP report no. 1. Am J Ophthalmol 2003; 2005; 112:2081–2087.
with photodynamic therapy in the 136:407–418. 45. Eckardt C, Eckardt U: Macular
management of subfoveal choroidal 36. Gragoudas ES, Adamis AP, Cunningham ET, translocation in nonexudative age-related
neovascularization in age-related macular et al: Pegaptanib for neovascular age- macular degeneration. Retina 2002;
degeneration. Am J Ophthalmol 2004; related macular degeneration. N Engl J 22:786–794.
137:880–887. Med. 2004; 351:2805–2816. 46. Cahill MT, Mruthyunjaya P, Bowes
27. Eckardt C, Eckardt U, Conrad HG: Macular 37. Heier JS, Antoszyk AN, Pavan PR, et al: Rickman C, Toth CA: Recurrence of retinal
rotation with and without counterrotation of Ranibizumab for treatment of neovascular pigment epithelial changes after macular
the globe in patients with age-related age-related macular degeneration: a phase translocation with 360 degrees peripheral
macular degeneration. Graefes Arch Clin I/II multicenter, controlled, multidose study. retinectomy for geographic atrophy. Arch
Exp Ophthalmol 1999; 237:313–325. Ophthalmology 2006; 113:633–642.e1–4. Ophthalmol 2005; 123:935–938.
28. Toth CA, Freedman SF: Macular 38. Park CH, Toth CA: Macular translocation 47. Jaffe GJ, Martin DF, Callanan D, et al:
translocation with 360° peripheral surgery with 360o peripheral retinectomy Fluocinolone acetonide implant (Retisert)
retinectomy: impact of technique and following ocular photodynamic therapy of for noninfectious posterior uveitis: thirty-
surgical experience on visual outcomes. choroidal neovascularization. Am J four week results of a multicenter
Retina 2001; 21:293–303. Ophthalmol 2003; 136:830–835. randomized clinical study. Ophthalmology
29. Cahill M, Toth CA: Macular translocation 39. Binder S, Krebs I, Hilgers RD, et al: 2006; 113:1020–1027.
surgery. In: Stephen JR, ed. Retina. Outcome of transplantation of autologous 48. Tan DT, Chee SP, Lim L, et al: Randomized
4th edn. Philadelphia, PA: Mosby; 2004. retinal pigment epithelium in age-related clinical trial of Surodex steroid drug
30. Pertile G, Claes C: Macular translocation macular degeneration: a prospective trial. delivery system for cataract surgery:
with 360° retinotomy for management of Invest Ophthalmol Vis Sci 2004; anterior versus posterior placement of two
age-related macular degeneration with 45:4151–4160. Surodex in the eye. Ophthalmology 2001;
subfoveal choroidal neovascularization. 40. Stanga PE, Kychenthal A, Fitzke FW, et al: 108:2172–2181.
Am J Ophthalmol 2002; 134:560–565. Retinal pigment epithelium translocation 49. Srivastava S, Mruthyunjaya P, Wiser J,
31. Mruthyunjaya P, Stinnett SS, Toth CA: after choroidal neovascular membrane et al: Intravitreal sustained-release
Change in visual function after macular removal in age-related macular fluocinolone acetonide device to treat
translocation surgery with 360° peripheral degeneration. Ophthalmology 2002; severe experimental uveitis. Association for
retinectomy for neovascular age-related 109:1492–1498. Research in Vision and Ophthalmology.
macular degeneration. Ophthalmology 41. Van Meurs JC, ter Averst E, Hofland LJ, Orlando, Florida; 2005:3536.
2004; 111:1715–1724. et al: Autologous peripheral retinal pigment

1983
CHAPTER

150 Acute Idiopathic Maculopathy


K. Bailey Freund, Catherine B. Meyerle, and Lawrence A. Yannuzzi

BACKGROUND senting visual acuities better than those typical for AIM at the
level of 20/60 to 20/100. A subretinal exudate may be present
Acute idiopathic maculopathy (AIM) is a rare disorder first in some cases. In one reported patient, the exudate had a
described by Yannuzzi and associates in 1991.1 The nine peculiar fluffy, white appearance suggestive of inflammatory
patients in this original series experienced sudden unilateral cells or debris.5 In a second patient, the exudate had a grayish,
visual loss secondary to an exudative maculopathy following a more solid appearance resembling subretinal fibrin seen in
flu-like illness. The initial term was unilateral AIM, but further central serous chorioretinopathy.
clinical experience revealed that this entity can present Whereas all the original cases were unilateral, three sub-
bilaterally.2 The preferred term today, therefore, is AIM. sequent cases with bilateral involvement have been reported.
In one case, macular involvement and acute loss of vision
DEMOGRAPHICS of the fellow eye occurred bilaterally within days of the
original presentation. In the other two cases, the lesion in the
AIM affects young adults with a mean age of 32 years (range fellow eye was eccentric to the macula, smaller in size, and
15–45 years) and has no sex predilection. It is uncommon, as asymptomatic.
demonstrated by a survey of the Macula Society, with most The natural course of AIM for most patients is complete
members of this organization seeing no more than one case per resolution of the exudative changes and near-complete resto-
year.3 Of reported patients, the majority are of white race. ration of the visual acuity (20/25 or better) within several weeks.
One reported patient was seen in the first trimester of Stigmata of the disease process persist after its resolution in
pregnancy. Another patient was examined 4 weeks post partum. the form of a bull’s eye type lesion. Specifically, the pigment
One patient was infected with human immunodeficiency virus epithelial atrophic change corresponds to the previous neuro-
but did not meet the clinical criteria for the acquired immuno- sensory detachment and the irregular hyperpigmentation is
deficiency syndrome. Of course, with small number of patients reflective of antecedent pigment epithelial disturbance. In the
and without a matched control group, any description of cases with subretinal exudation, this material resolved in
epidemiologic features is preliminary. advance of the overlying neurosensory detachment and did not
seem to influence the natural course of the disorder. In the cases
CLINICAL FEATURES with papillitis, this resolved in tandem with the exudative
macular changes.
AIM patients present with sudden onset of severe vision loss to To date, there have been no recurrent episodes in the cases
the level of 20/200 or worse with central metamorphopsia or a studied. However, the long-term visual outcome may be affected
central scotoma. A viral prodrome is typical and an association by development of choroidal neovascularization (CNV) and
between AIM and coxsackie virus has been reported.4 We have disciform scarring secondary to the retinal pigment epithelial
seen an AIM patient with positive acute coxsackie titers disturbance, which has occurred in two cases to date.
(unpublished data).
On examination, patients have a quiet anterior segment. In ANGIOGRAPHY
the posterior pole, acutely, one sees a neurosensory retinal
detachment with irregular margins overlying a smaller, grayish The fluorescein angiographic findings in the acute stage of
plaque at the level of the retinal pigment epithelium (RPE) AIM characteristically demonstrate early hyperfluorescence at
(Fig. 150.1). Other signs of inflammation such as vitreous cell, the level of the RPE lesion. Late stage hyperfluorescence also
papillitis, phlebitis, or intraretinal hemorrhage may also occur. occurs, corresponding to the combined subretinal staining
The mild vitreous cell is best appreciated with a contact lens. external to the RPE and pooling within the overlying neuro-
Papillitis (defined as disk hyperemia and fluorescein angio- sensory retinal detachment. This late stage pattern is similar
graphic staining) has been reported in a few cases as a subtle to the homogenous leakage seen beneath a serous pigment
obscuration of the disk margins. The retinal veins in these cases epithelial detachment. In the resolved state, a bull’s eye pattern
were somewhat prominent and tortuous from secondary of central hypofluorescence surrounded by a ring of hyperfluor-
obstructive change or a mild phlebitis. In one case, a relative escence is observed corresponding to the typical RPE changes of
afferent pupillary defect was noted (R Janigian and C Smith, the healed lesion. In the cases with papillitis, fluorescein
unpublished data, 1995). Intraretinal hemorrhage may be mis- angiography revealed disk staining in all three reported cases
taken for subretinal neovascularization by the less experienced and mild perivenous staining in one instance.
observer. Generally, the lesion is in the foveal center, but may Three cases were studied with indocyanine green angio-
present eccentrically. Patients with eccentric lesions have pre- graphy during the acute phase of their presentations. The studies 1985
RETINA AND VITREOUS

a b c
SECTION 10

d e

FIGURE.150.1. (a) Color photograph shows an exudative detachment of the neurosensory retina, temporal to the center of the macula. There is
a neurosensory retinal elevation overlying an irregular thickening at the level of RPE. Visual acuity is 20/60. (b) Late fluorescein angiogram reveals
complete staining of the neurosensory retinal detachment. (c) Color photograph of the resolved state shows the characteristic atrophy of the
RPE surrounding a clump of pigment epithelial hyperplasia, corresponding to the neurosensory retinal detachment and the pigment epithelial
lesion respectively. Visual acuity is 20/20. (d) Color photograph of the fellow eye reveals a shallow alteration to the retina near the disk. There is
an irregular, grayish thickening to the underlying RPE. (e) Fluorescein angiogram of the fellow eye shows staining of the acute lesion.
(a–e) From Freund KB, Yannuzzi LA, Barile GR, et al: The expanding clinical spectrum of unilateral acute idiopathic maculopathy. Arch Ophthalmol 1996; 114:555–559.
Copyright 1996, American Medical Association.

were unremarkable except for some mild hypofluorescence CNV that spontaneously infarcts and regresses. Indocyanine
corresponding to the exudative detachment, which was seen green angiography of AIM patients, however, has not demon-
best in the late phase of the study. There was no late subretinal strated early vascular hyperfluorescence or late leakage con-
staining suggestive of CNV in any of these patients. sistent with CNV.7 In addition, indocyanine green angiography
can help differentiate AIM from central serous chorioretino-
DIFFERENTIAL DIAGNOSIS pathy. Indocyanine green angiography of AIM patients does not
demonstrate the mid-phase foci of choroidal hyperfluorescence
The differential diagnosis of AIM includes numerous degener- suggestive of choroidal hyperpermeability that has been
ative, infiltrative, inflammatory, and infectious disorders, such described in central serous chorioretinopathy.7
as idiopathic CNV, serous detachment of the RPE, central serous A recent case report describes the optical coherence tomo-
chorioretinopathy, Harada’s disease, serpiginous choroidopathy, graphy (OCT) and multifocal electroretinogram (ERG) findings
posterior scleritis, acute posterior multifocal placoid pigment in a patient with AIM.8 The authors conclude that the acute
epitheliopathy (APMPPE), and secondary placoid syphilitic OCT findings were consistent with thickening at the outer
retinitis. A recent case report described an acute toxoplama retina and RPE, and that these outer edematous changes
retinitis masquerading as AIM,6 but the patient had satellite resolved as visual acuity improved. Additionally, the reduced
lesions and a marked retinal vasculitis not typical of AIM. amplitudes in multifocal ERG were suggestive of outer retinal
Many patients with AIM are originally suspected of having dysfunction as the cause of visual loss. These OCT and multi-
central serous chorioretinopathy or idiopathic CNV. In fact, the focal ERG findings indicative of outer retina and RPE pathology
authors have seen two AIM patients misdiagnosed with help exclude masquerading diseases that primarily involve
idiopathic CNV and treated with photodynamic therapy. These inner retina.
patients eventually did recover their central acuity to the level Careful clinical biomicroscopy is essential to distinguish
typical for AIM patients, but their visual recovery was delayed AIM from masquerading APMPPE. Although AIM shares
by several months. The typical flu-like illness accompanied by several clinical and demographic characteristics with APMPPE,
evidence of inflammation (vitreous cells, papillitis) helps to there are many features of AIM that exclude the diagnosis of
distinguish AIM from idiopathic CNV. Acute titers for APMPPE. These include the presence of a solitary macular lesion
coxsackie virus also help in making the diagnosis. with a neurosensory retinal detachment, intraretinal hemorr-
Indocyanine green angiography may be useful in dis- hages, and early hyperfluorescence of an acute AIM lesion on
tinguishing AIM from other entities. Some believe that AIM, in fluorescein angiography. These features have not been reported
1986 those patients without inflammation, may represent occult in APMPPE.
Acute Idiopathic Maculopathy

TREATMENT is appropriate, and any patients presenting atypically with, for


example, exuberant inflammatory findings or lack of sponta-
Since this disorder is self-limited and most patients ultimately neous resolution should undergo appropriate diagnostic testing.
recover excellent visual acuity, it does not appear that treatment
of the acute lesion is necessary. Perhaps of interest is that one CONCLUSION
of the patients in the original report who was treated with
systemic corticosteroids ultimately developed milder pigmen- AIM is a distinct entity with characteristic clinical and angio-
tary changes in the fovea than is typical.1 graphic features. The frequent association with a flu-like illness,
vitreous cells, and in some cases, papillitis and subretinal
CAUSE exudation is highly suggestive of an inflammatory disorder. The
recently reported association with coxsackie virus also suggests
AIM involves an inflammatory process of the RPE and, to a an inflammatory pathogenesis. Fortunately, the natural course
lesser degree, the optic nerve. An association with coxsackie of the disease is spontaneous, rapid resolution with recovery of
virus has recently been reported in two patients.4 The authors excellent visual acuity. The recognition of this disorder is
have also seen an AIM patient with positive acute coxsackie important to eliminate unnecessary and even potentially harm-
titers (unpublished data). Of course, the clinical circumstances ful treatment such as laser photocoagulation or photodynamic
of each individual case should dictate what medical evaluation therapy of subretinal tissue misdiagnosed as CNV.

CHAPTER 150
REFERENCES
1. Yannuzzi LA, Jampol LM, Rabb MF, 4. Beck A, Jampol L, Glaser D, et al: Is 7. Yannuzzi LA, Slakter JS, Sorenson JA, et al:
et al: Unilateral acute idiopathic coxsackie virus the cause of unilateral Digital indocyanine green videoangiography
maculopathy. Arch Ophthalmol 1991; acute idiopathic maculopathy? Arch and choroidal neovascularization. Retina
109:1411–1416. Ophthalmol 2004; 122:121–123. 1992; 12:191–223.
2. Freund KB, Yannuzzi LA, Barile GR, et al: 5. Fish RH, Territo C, Anand R: 8. Aggio F, Farah M, Meirellee R, et al:
The expanding clinical spectrum of Pseudohypopyon in unilateral acute STRATUSOCT and multifocal ERG in
unilateral acute idiopathic maculopathy. idiopathic maculopathy. Retina 1993; unilateral acute idiopathic maculopathy.
Arch Ophthalmol 1996; 114:555–559. 13:26–28. Graefes Arch Clin Exp Ophthalmol 2006;
3. Hanutsaha P, Yannuzzi LA, Freund KB: 6. Lieb DF, Scott IU, Flynn HW, et al: Acute 244:510–516.
The occurrence of uncommon intraocular acquired toxoplasma retinitis may present
inflammatory diseases: a survey of the similarly to unilateral acute idiopathic
Macula Society. Retina 1996; maculopathy. Am J Ophthalmol. 2004;
16:437–439. 137:940–942.

1987
CHAPTER

151 Idiopathic Polypoidal Choroidal Vasculopathy


Chiara M. Eandi, Christina M. Klais, K. Bailey Freund, and Lawrence A. Yannuzzi

More than two decades ago, a peculiar hemorrhagic disorder of isolated island within the central macula. It is generally thought
the macula, polypoidal choroidal vasculopathy (PCV) was first that PCV originates in the inner choroid.
described (see Yannuzzi LA: Idiopathic polypoidal choroidal vascu- Lafaut et al reported light microscopic findings in submacular
lopathy. Presented at the Annual Macular Society Meeting, Miami, tissue removed from an eye with PCV in AMD.15 They described
FL, 1982) This entity has also been designated by Kleiner and a sub-RPE, intra-Bruch’s fibrovascular membrane containing
associates as ‘posterior uveal bleeding syndrome’1,2 and by Stern several dilated thin-walled vessels under diffuse drusen. These
and co-workers as ‘multiple recurrent retinal pigment epithelium abnormal vessels are lined by a thin endothelium with occa-
(RPE) detachments in black women’.3 In 1990, Yannuzzi and sional pericytes. Some lesions were associated with islands of
colleagues suggested the term idiopathic polypoidal choroidal lymphocytic infiltration.
vasculopathy (IPCV) because the pathogenesis was unknown, In spite of increasing case experience, the pathogenesis of the
the primary abnormality involved the choroidal circulation, and IPCV is still unknown. Traditionally, IPCV has not been linked
the characteristic lesion was an inner choroidal vascular network to inflammatory processes; however, Ciardella et al reported a
of vessels ending in an aneurismal bulge or outward projection, link between PCV and chorioretinal inflammation seen in three
visible clinically as a reddish-orange, spheroid, polyp-like struc- cases. They identified typical clinical findings of PCV after pre-
ture.4 This entity has characteristic morphological features that vious inflammatory disease.8 Immunohistochemical findings in
distinguish it from other exudative maculopathies. Clinically it a case with PCV demonstrated both T and B lymphocytes in the
was associated with multiple, recurrent, serosanguineous choroid and fibrovascular tissue.17
detachments of the RPE and neurosensory retina secondary to There have been references to the association of PCV with
leakage and bleeding from the peculiar choroidal vascular lesion other ocular disorders. Ross et al reported a correlation between
(Fig. 151.1). retinal macroaneurysms and PCV in two hypertensive black
In recent years, indocyanine green (ICG) angiography has females.6 They proposed a relation between the retinal vascular
been used to detect and characterize the PCV abnormality with changes in retinal macroaneurysms and hypertensive retino-
enhanced sensitivity and specificity,5,6 expanding further the pathy such as vascular remodeling, aneurysmal dilation, and
spectrum of PCV. This allows us to describe the pathogenesis, focal vascular constriction to the characteristic choroidal
clinical manifestations, demographic profile, fluorescein and ICG lesions in PCV. A recent study confirmed that choroidal blood
angiographic findings, natural course, modalities of treatment, flow increases significantly with increasing blood pressure.18
and visual prognosis in patients with PCV with greater detail Untreated hypertension was found to be a risk factor in
and precision.7,8 development and progression of choroidal vascular pathologies.
Other potential pathogenic factors include the possibility of a
PATHOGENESIS peculiar choroidal tumor, vascular malformation, or systemic
hypertension. In spite of these observations, the association of
Although the pathogenesis of PCV is not completely clear, new PCV, inflammation or other ocular disorders is still inconclusive
information is now available that might help in our under- and must be investigated further.
standing of its possible causative factor and clinical nature. PCV
has been first described in African-American females.3,9 However, DEMOGRAPHIC FEATURES
Asian patients are also at risk for developing PCV, and white
patients can definitely develop the disorder as well.7,9–11 This pre-
disposition for pigmented races contrasts the relative immunity
AGE
to age-related macular degeneration (AMD) and disciform scarring PCV is usually diagnosed in patients between the age of 50 and
seen in these individuals.12–14 Lafaut et al suggested a possible 65 years, but the age of diagnosis can range from 20 to 80 years.
coexistence of AMD and PCV, a finding certainly supported by The average age of onset for all affected patients from the
our own clinical experience.15 In spite of multiple, recurrent sero- literature is 60.1 years. Caucasian subjects usually present the
sanguineous macular detachments, significant fibrous prolifera- disease at an older age.19
tion typical of end-stage neovascularized AMD is unusual in PCV.16
Although there is no available clinical pathologic confir-
mation, in essence the vascular abnormality in PCV appears to SEX
be a singular lesion with tubular and polypoidal components Previously, it was thought that PCV exclusively affects women.
that vary in size. The vascular lesion itself appears to be singular In recent studies it has been demonstrated that females are
and progressive. It does not occur solely in the peripapillary involved predominantly over males by a ratio of approximately
region as originally believed, but it may also present as an 4.7:1. Among Asians, men seem to be more frequently affected.16 1989
RETINA AND VITREOUS

FIGURE 151.1. (a) The fluorescein angiogram


shows a large serosanguineous detachment of
the pigment epithelium that extends in the
inter-papillo-macular region and a subfoveal
hemorrhage with an adjacent hyperfluorescent
spot (white arrow). (b) The corresponding ICG
angiogram reveals the exact location of the
vascular choroidal polypoidal abnormality in the
peripapillary area (black arrow).

a b

RACE orange, bulging, polyp-like dilations in the peripapillary and


Individuals of African-American and Asian descent are at higher macular area. Vitreous hemorrhage, relatively minimal fibrous
SECTION 10

risk of developing PCV.3,16 as this distinct disorder seems to scarring, absence of drusen, retinal vascular disease, pathologic
preferentially affect pigmented individuals by a ratio of 4.2:1. myopia, and signs of intraocular inflammation are accepted
However, more recent reports have shown that PCV occurs clinical features. Iida et al demonstrated that retinal micro-
broadly in patients of other racial descents than it has been angiopathy may occur in a chronic macular detachment
demonstrated in the past.16,20–23 secondary to polypoidal CNV.31
Lafaut et al studied the prevalence of PCV in Caucasians with
occult choroidal neovascularization (CNV). In a consecutive
series of 374 eyes with occult CNV, 4% were diagnosed with THE VASCULAR LESION
PCV by ICG angiographic findings.10 Pauleikhoff et al diagnosed In order to define the vascular lesion it is essential to use ICG
PCV in 13.9% of 101 consecutive German patients with pigment angiography because it images the active neovascularization,
epithelium detachment (PED).24 Other investigators diagnosed whereas it does not show leakage at the site of serous pigment
PCV in 85% among a consecutive series of Caucasians pre- epithelial elevations (Fig. 151.1). Although the lesion of PCV is
senting with large exudative or hemorrhagic PEDs in the absence invariably localized to the choroidal vascular network, other
of drusen.25 Yannuzzi et al diagnosed PCV in 13 (7.8%) of 167 characteristics of the lesion often differ. Significant variance is
consecutive Caucasian patients with presumed neovascular observed in size and location to the optic disk, fovea, and cross
AMD.19 Scassellati-Sforzolini et al found a prevalence of 9.8% section of the retina.
in Italian patients with newly diagnosed neovascular AMD.11
PCV has also been reported in Irish,26 French,27 and German28
subjects. SIZE
The prevalence of PCV among Chinese patients with AMD PCV usually categorized into small, medium or large lesions.
was found to be 9.3%.29 In this patient series, the most common The width of the lesions varies depending on the affected
clinical finding at presentation was subretinal hemorrhage vascular channels. With involvement of outer choroidal vessels,
(63.6%) followed by exudative neurosensory detachment the polypoidal lesions appear larger in size. Such lesions are
(59.1%), and hemorrhagic PED (59.1%). There was a predo- easily diagnosed on biomicroscopy, especially when atrophic
minance for males (68.4%). Most of the polypoidal lesions were RPE is overlying; whereas angiography is needed to detect
at the macula (63.6%) and unilateral (84%). In contrast, a vascular lesions of small dimension. Teitawa et al reported large
Japanese study reported a much higher prevalence of PCV vascular networks expanding across the vascular arcade in 12 of
among patients with newly diagnosed neovascular AMD.30 60 consecutive eyes diagnosed with PCV.32
They studied 164 eyes with PEDs and CNV and detected PCV When the middle choroidal vasculature is affected, polypoidal
in 59% of the eyes. In 70% of the patients, the PED had a lesions appear smaller. In this case it is more challenging to
hemorrhagic component. Other investigators confirmed the diagnose on clinical examination, and the preferential method
predilection for male gender in the Japanese population, as well for diagnosis is ICG angiography.4,5,33
as unilateral involvement.16
Recently, we found PCV in 14 (14%) of 100 consecutive newly
diagnosed cases with neovascular AMD. They all presented LOCATION
occult or minimally classic lesion on fluorescein angiography, Choroidal vascular lesions of PCV are usually located in the
but the polyp-like CNV was visible when studied with ICG peripapillary area (Figs 151.2 and 151.3), although recent
angiography (see Eandi CM, Iranmanesh R, Garuti S, et al: The evidence suggests that lesions could also be found in the
nature and frequency of neovascular age related macular central macula (Figs 151.4 and 151.5) and in the midperiphery
degeneration. ARVO 2005; Abstract 3311). (Fig. 151.6).34,35 Lesions can be localized to a single location in
the fundus or can be widespread involving more than one site.
CLINICAL FINDINGS Lafaut et al detected polypoidal lesion in the macula in 22 of
45 eyes, in the peripapillary area in 16 of 45, under the
PCV is a bilateral disease.4 Most patients with the evidence of temporal vascular arcade in six of 45, and in the midperiphery
PCV in one eye eventually develop similar lesions in the fellow in six of 45 eyes.10 Yannuzzi and associates described a case of
eye; however, several patients with PCV have been followed up PCV isolated in the midperiphery of the temporal fundus
for more than 10 years and so far demonstrate no evidence of associated with recurrent vitreous hemorrhage.35
the condition in the other eye. The lesion is characterized by Due to limited reports on clinocopathologic relationship
1990 the presence of dilated, choroidal vascular channels ending in between PCV lesions and retinal layers the precise location of
Idiopathic Polypoidal Choroidal Vasculopathy

a b c

FIGURE 151.2. (a) Color photograph of a patient with IPCV. A choroidal abnormality in the peripapillary area shows a barely evident vascular
pattern and polypoidal changes. (b) The fluorescein angiogram reveals the vascular abnormality because of the large size of the vascular

CHAPTER 151
element’s abnormality and the overlying pallor or atrophy of the pigment epithelium. (c) The corresponding ICG angiogram more clearly
delineates the vascular abnormality with branching inner choroidal vessels ending in polypoidal choroidal lesions.

FIGURE 151.3. (a) Patient with IPCV shows


the vascular abnormality, along with the
reddish-orange elevation, beneath a PED.
(b) The corresponding fluorescein angiogram
highlights the vascular lesion in the peripapillary
area and the serosanguineous detachment of
the pigment epithelium that extends to the
inferior and temporal macular area. (c) The
early ICG study accentuates the polypoidal
choroidal vascular changes. The reddish-
orange lesion is shown to represent a ring of
polypoidal lesions that project anteriorly into
the subpigment epithelial space. (d) The late-
stage ICG study shows staining of the margins
b
of the polypoidal lesions with relative fading of
the branching vessels (washout).
a

c d

the polypoidal vascular lesion in the cross-sectional structure neous macular detachments, fibrous proliferation resulting in
remains unknown. Optical coherence tomography (OCT) studies typical plaque characteristics of end-stage neovascular AMD is
were able to localize the polypoidal lesions under Bruch’s not seen in eyes with PCV. On the other hand, this may present
membrane36 but further studies are required to correlate the difficulties with the diagnosis of PCV when the lesion is
histologic finding and the results of OCT imaging. inactive. In some cases, polypoidal lesions have evolved with
resolution of the serosanguineous changes, and ICG angio-
NATURAL COURSE graphy reveals a nonspecific plaque which can be interpreted as
CNV of the usual type. Still, some patients may develop chronic
The natural course of PCV is becoming better understood as the atrophy and cystic degeneration of the fovea associated with
knowledge of PCV expands. This disease often follows a severe vision loss. Others may experience vitreous hemorrhage
remitting–relapsing course, and clinically, it is associated with or secondary CNV with disciform scarring and central vision
chronic, multiple, recurrent serosanguineous detachments of loss (see Yannuzzi LA: Idiopathic polypoidal choroidal vasculo-
the neurosensory retina and the RPE with long-term preser- pathy. Presented at the Annual Macular Society Meeting, Miami,
vation of good vision. Despite multiple recurrent serosangui- FL, 1982).2,13,17,37 Massive spontaneous choroidal hemorrhage 1991
RETINA AND VITREOUS

a b c

FIGURE 151.4. (a) Color photograph of a patient with IPCV who presented with chronic serous lipid exudation in the macula, simulating central
serous chorioretinopathy. (b) The ICG angiogram reveals the vascular abnormality and leaking polypoidal lesion. (c) The same patient after
photocoagulation of leaking polypoidal lesions shows resolution of the exudative changes.
SECTION 10

FIGURE 151.5. (a) The red-free photograph


shows a small serosanguineous RPE
detachment in the center of the fovea with
pigmentary defects surrounding. (b) The ICG
angiogram reveals a cluster of actively leaking
polypoidal vessels in the corresponding area.

a b

FIGURE 151.6. (a) ICG composite late


angiogram of a patient with a midequatorial
PCV lesion presenting as a subretinal
hemorrhage. (b) A magnified image of the
lesion reveals the polypoidal vascular
abnormality beneath the blood.

a b

is a rare but severe complication. Despite immediate drainage rysms.6 Smith et al reported a patient with PCV and concurrent
procedure the visual outcome is poor.38 sickle cell disease.39 Other cases of PCV have been described in
Uyama et al followed 14 eyes of 12 consecutive patients with eyes with melanocytoma of the optic nerve, and central retina
PCV for at least 2 years without any treatment.16 They demon- vein occlusion.40,41 Neither of these case reports nor our own
strated that 50% of the eyes had a favorable course. In the clinical experience demonstrates sufficient evidence finding an
remaining half, the disorder persisted for a long time with association between PCV and retinal vascular ischemic diseases,
occasional recurrent hemorrhages and leakage, resulting in chorioretinal inflammation, or sickle cell disease.
macular degeneration and vision loss. Eyes with cluster of When the lesion increases in size, it usually does so by three
grape-like polypoidal dilations of the vessels had a high risk for proposed mechanisms. The lesion may enlarge by simple vessel
severe visual loss. hypertrophy, by conversion of the lesion into the advancing
No underlying systemic factor has ever been associated with edge of a vascular channel, and by unfolding of a cluster of
this disorder, except for recent evidence suggesting a possible aneurysmal elements and subsequent transformation into
1992 association of PCV to hypertension and acquired macroaneu- enlarging, vascular, tubular components. The latter mechanism
Idiopathic Polypoidal Choroidal Vasculopathy

is usually apparent on clinical examination as a large, reddish- nature and frequency of neovascular age related macular
orange subretinal mass corresponding to a cluster of aneurysmal degeneration. ARVO 2005; Abstract 3311).
elements that, on ICG angiography, project anteriorly from the
inner choroid toward the outer retina. With time, these mass- DIFFERENTIAL DIAGNOSIS
like lesions flatten out and expand tangentially in their plane.
The overlying RPE may show signs of variable atrophy. The reddish-orange, subretinal masslike lesion seen in PCV is
composed of multiple, polypoidal elements that extend from the
DIAGNOSIS PCV vascular lesion in the plane of the inner choroid, beneath
the detached pigment epithelium toward the outer retina. The
The appearance of the vessels in PCV often depends on their differential diagnosis of such a reddish-orange lesion under the
location in the posterior pole. In patients with juxtapapillary retina includes choroidal hemangioma, metastasis from
lesions, the vascular channels may follow a radial, arching carcinoid syndrome, or more rarely, renal cell carcinoma, pos-
pattern and may be interconnected with smaller spanning terior scleritis, choroidal osteoma, and even CNV. ICG angio-
branches which are more evident and numerous at the edges of graphy is useful in differentiating the reddish-orange mass as it
the lesion. In patients with PCV limited to the macula, a demonstrates an incomplete ring of polypoidal lesions emanating
vascular network often arises in the macula and follows an oval from the PCV vascular lesion. The cluster of polypoidal
distribution pattern. elements may be seen stereoscopically to extend toward the
ICG angiography is useful to image the PCV choroidal overlying retina. The other mimicking orange mass-like entities

CHAPTER 151
abnormality.5 In the initial phases of ICG videoangiograms, are not associated with these dilated inner choroidal vessels
when the larger choroidal vessels fill with dye, a distinct and polypoidal vascular elements beneath a pigment epithelial
network of vessels within the choroid becomes visible. Early in detachment.
the course of the ICG study, the larger vessels of the PCV Two decades ago, PCV was considered an entity with a
network start to fill before the retinal vessels, but the area distinct set of demographic characteristics, risk factors, natural
within and immediately surrounding the network is relatively course, clinical interpretation, and outcome from AMD.
hypofluorescent compared with the uninvolved choroid. The Features like Caucasian origin, macular location, presence of
vessels of the network appear to fill more slowly than the retina drusen, frequent recurrences, rapid rate of progression, disci-
vessels. The network of vessels with ICG angiography generally form scarring, and poor visual prognosis were considered typical
appear to be more numerous than one would expect from of AMD and used to differentiate the two entities.13 Since the
clinical examination. Shortly after the network can be identified definition of PCV has expanded over the past two decades, the
on the ICG angiogram, small hyperfluorescent ‘polyps’ become diagnosis is no longer restricted to those specific demographic
visible within the choroid. attributes or to a specific retinal location.10,35 In addition, it
During the mid-phase of the angiogram, however, the size of becomes more evident from the increasing volume of reports
the lesions approximates the choroidal excrescences observed about PCV and our own clinical experience that the patients
clinically. They appear to leak slowly and the surrounding hypo- may have manifestations attributable to PCV and AMD. For
fluorescent area becomes increasingly hyperfluorescent. example, some of our patients diagnosed with AMD undergoing
The late phase of angiogram is associated with a reversal of ICG guided-laser treatment in the past had an exceptional and
the pattern of fluorescence observed previously. The area sur- unexpected good outcome. Later, on reviewing their records, we
rounding the lesion becomes hyperfluorescent and the center of determined that the patients with better results were actually
the lesion demonstrates hypofluorescence. The size of the lesion patients with PCV-type CNV rather than a typical form of
seems to influence the reversal pattern. The lesions that are less AMD.
than 0.5-disk diameter in size appear to have intense uniform Presently, we believe that PCV represents a subtype of CNV
fluorescence, whereas internal details seem to be visible in in AMD. PCV has also been reported in association with dry
larger polypoidal structures.5 These findings, in turn, suggest AMD.44 However, some characteristics distinguish PCV from
presence of an internal architecture.5 other types of CNV observed in AMD. Vascular proliferative
The very late stages of ICG angiography demonstrate dis- changes associated with nonpolypoidal CNV tend to produce
appearance of the fluorescence from the lesions thus defining small caliber vessels that are associated with grayish colored
the term ‘washout’ (Fig. 151.3d). The ‘washout’ is only seen in membrane and grayish discoloration of the overlying retina.
nonleaking lesions, whereas the leaking lesions remain These vessels are not easily detected clinically.21,45 In contrast,
hyperfluorescent. Varying degrees of hypopigmentation of the eyes with vascular changes typical for PCV form a network of
RPE overlying an involved area may enhance visualization of vessels ending with saccular polypoidal lesions which have a
the abnormal vessels, yet the late ICG staining results from the redish-orange color and evident with slit lamp biomicroscopy
intrinsic characteristics of the lesion rather than from RPE unless they are covered by overlying blood or exudates.4,9 Other
alterations.5 angiographic and histopathologic characteristics tends to
OCT has also proved to be useful in the diagnosis of PCV. distinguish other types of CNV from PCV. AMD is associated
Iijima and co-workers were able to demonstrate the presence of with diminishing choroidal thickness and with signs of stromal
the polypoidal lesions with OCT.36,42 They also proposed that fibrosis without any changes in the choriocapillaris size or
OCT may be able to differentiate the orange-red lesions in PCV evidence of inflammatory changes.46 These findings were not
from serous PEDs. Otsuji et al were able to demonstrate with observed clinocopathological reports on PCV.15,17 Moreover,
OCT the presence of dome-like elevation of the RPE, and poorly defined CNV secondary to AMD is clinically associated
nodular appearance beneath the RPE in 14 patients with PCV23. with indistinct subretinal thickening and is not manifested by
Giovannini et al studied with OCT four eyes with PCV and the detectable choroidal vascular channels terminating in
found in all cases a characteristic hyper-reflectivity in the polyp-like structures.
choroidal layers.43 The use of OCT is of importance in these Fluorescein and ICG angiography can be used to distinguish
patients, as we recently reported. Although the polypoidal CNV the two types of vascular abnormalities. In both, CNV is
eyes seldom had associated CME (7%), a dome-like pigment characterized by diffuse late staining plaque.12 In contrast,
epithelium detachment was detected in 13 of 14 (93%) patients early-phase ICG angiography reveals PCV as a prominent
with PCV (see Eandi CM, Iranmanesh R, Garuti S, et al: The vascular network which becomes ‘washed out’ during the late 1993
RETINA AND VITREOUS

phase. The late phase is also distinguished by a typical outline vision outcomes.52–54 Macular translocation is another surgical
of the nonleaking large choroidal vessels. If the vessels leak, approach, but most patients diagnosed with PCV have relatively
staining in the walls of aneurysmal lesions and exudation into large vascular lesions and the surgical procedure has a number
the surrounding choroid and subretinal space is observed.5 of serious complications.55 Low-dose external beam irradiation
The natural course and clinical location of PCV are different does not appear to offer dramatic results, neither beneficial nor
from those of CNV. Subfoveal CNV tends to organize into a harmful37,56; on the other hand, it has been demonstrated that
fibrotic or disciform scar leading to severe macular damage and radiation therapy for AMD is a possible triggering factor for the
vision loss.21 PED seen in eyes with PCV virtually never forms development of PCV.57
a fibrotic scar,4 whereas PED associated with occult CNV in Transpupillary thermotherapy may show promise in treating
AMD usually has a poor prognosis.47 However, the spontaneously occult CNV,58 but the value in PCV is not known. Diode laser
involuted or regressed lesion looks like a well-delineated plaque photocoagulation has also been successfully used for the
of occult CNV, even with ICG angiography. treatment of PCV.20
Some eyes with PCV may present purely exudative changes Recent studies reported encouraging results without compli-
masquerading chronic decompensation of the RPE, a variant cations after photodynamic therapy with verteporfin in subfoveal
of central serous chorioretinopathy (Fig. 151.4).33 PCV lesions PCV lesions.59–65 Spaide et al reported on improvement in
presenting with symptoms of central serous chorioretinopathy visual acuity in 56% of patients undergoing photodynamic
are usually small in size. Polypoidal lesions may resemble small therapy, while in 31% of the patients the vision was stable, and
PEDs clinically and on fluorescein angiography. The diagnosis in 12% a decrease in visual acuity was noted.59 Quaranta et al
SECTION 10

becomes especially challenging in a patient with chronic central found beneficial functional results in two cases of PCV treated
serous chorioretinopathy presenting lipid deposits in the central with PDT with verteporfin.63
macula due to subfoveal polypoidal lesion. The principle of We recently reported stabilization or improvement on visual
differentiating a small serous PED from a polypoidal lesion acuity in 80% of eyes with PCV treated with ICG guided PDT
is with ICG angiography. Late staining of the PED is seen treatment. In our series of 30 eyes with PCV, the PDT treat-
with fluorescein angiography and hypofluorescence with ICG ment was applied only on the active lesion visible with ICG
imaging. In contrast, the polypoidal lesion is usually hyperfluor- angiography, possibly reducing the collateral damage to the
escent with ICG angiography because of its vascular nature. It choriocapillaries. Angiographic and tomographic findings con-
is important to consider that the PCV lesion may exist under a firmed the resolution of the neovascular process in all cases (see
PED. The portion of the PED overlying the polypoidal lesion Eandi CM, Freund KB, Ober MD, et al: Minimally selective
will be hypofluorescent, and, if there is leakage, ICG dye may treatment for neovascular age related macular degeneration.
pool into the subpigment epithelial space. However, the Annual Meeting of the Retina Society, Coronado, CA; 2005).
majority of patients with PCV present with serosanguineous Further trials of photodynamic therapy with verteporfin for PCV
detachment of the RPE and neurosensory retina. These findings are required to address long-term efficacy and safety issues.
imply the presence of new blood vessel formation or CNV as
causative factor.21 CONCLUSION
TREATMENT PCV appears to be a distinct clinical entity involving the
choroidal circulation. The vascular abnormality is in the inner
Treatment for PCV is not yet well established. If the choroid, composed of two fundamental elements, a dilated
characteristic vascular lesion of PCV is observed, a conservative network of vessels terminating in multiple areas of aneurysmal
approach to management is recommended unless the lesion is swelling in a polypoidal configuration. The polypoidal lesion
associated with persistent or progressive exudative change itself accounts for the episodic leakage and bleeding seen in
threatening central vision. In this case conventional thermal these patients. In patients with serosanguineous detachment of
laser treatment of the leaking polypoidal choroidal abnormality the pigment epithelium, particularly those with increased risk
may be successful in causing resolution of the serosanguineous factors like African-American or Asian race, ICG angiography
manifestation.20,25,48 Yuzawa et al reported on visual should be performed to evaluate the choroidal vascular
improvement after conventional laser treatment of the entire abnormality in an attempt to establish a more definitive
PCV complex in nine of 10 eyes.49 However, no randomized, diagnosis. If the characteristic vascular lesion of PCV is seen, a
controlled studies have been performed to prove the efficacy or conservative approach to management should be entertained
safety of laser treatment. Moreover, when the CNV extends unless there is a persistent or progressive exudative change that
beneath the center of the fovea thermal laser treatment is not is threatening central macula. In that event, there may be a
indicated due to damage of the overlying neurosensory retina rationale for photocoagulation treatment of leaking aneurysmal
and subsequent lead to reduced central vision.50 It is possible or polypoidal components within the vascular lesion, but not
that the loss of central vision due to the subfoveal treatment the entire vascular complex. Photodynamic therapy seems to be
may exceed the damage produced by the natural course of PCV, a promising therapeutic modality; however, randomized clinical
although this is not known with any certainty. Conversely, it is trials are needed to establish the efficacy and safety of the PDT
not uncommon for patients with untreated subfoveal involve- treatment in the management of these patients.
ment with PCV to experience severe visual loss.
Due to limitations and uncertainty of the value of thermal SUMMARY
laser coagulation and the potential devastating outcome of
untreated subfoveal lesions, alternative modalities of treatment PCV seems to be a distinct clinical entity that should be differ-
are currently being evaluated. These include vitrectomy and entiated from other forms of CNV associated with AMD and other
submacular removal of polypoidal vessels and subretinal blood. known choroidal degenerative, inflammatory, and ischemic dis-
In some cases, vitrectomy is required to clear the media and orders. The principal abnormalities in PCV, notably the branching
recover the vision. Shiraga et al reported anatomic success in vascular network and polypoidal structures at the borders of the
the surgical treatment of submacular hemorrhage associated lesion, seem to be unique to this entity. In patients with sero-
with PCV.51 However, the value of submacular surgery has sanguineous detachment of the RPE, especially in those with
1994 been questioned in AMD because of the recurrences and poor increased risk factors, such as pigmented race, ICG angiography
Idiopathic Polypoidal Choroidal Vasculopathy

should be performed to evaluate the choroidal vascular abnor- event, thermal lasercoagulation of leaking aneurysmal or poly-
mality in an attempt to establish a more definitive diagnosis. poidal components within the vascular lesion may be a rationale.
When ICG angiography confirms the characteristic vascular Reports on photodynamic therapy for PCV demonstrating
polyp-like lesion, a conservative management approach should encouraging results, but randomized clinical trials are required
be considered, unless there is a persistent or progressive exudative to establish this therapeutic modality in the management of
change threatening the fovea and the central vision. In that patients with PCV.

REFERENCES
1. Kleiner RC, Brucker AJ, Johnson RL: 18. Polak K, Polska E, Luksch A, et al: 33. Yannuzzi LA, Freund KB, Goldbaum M, et al:
Posterior uveal bleeding syndrome. Choroidal blood flow and arterial blood Polypoidal choroidal vasculopathy
Ophthalmology 1984; 91(Suppl 9):110. pressure. Eye 2003; 17:84–88. masquerading as central serous
2. Kleiner RC, Brucker AJ, Johnson RL: 19. Yannuzzi LA, Wong DW, Sforzolino MS, chorioretinopathy. Ophthalmology 2000;
Posterior uveal bleeding syndrome. Retina et al: Polypoidal choroidal vasculopathy 107:767–777.
1990; 10:9–17. and neovascularized age-related macular 34. Moorthy RS, Lyon AT, Rabb MF, et al:
3. Stern RM, Zakov N, Zegarra H, et al: degeneration. Arch Ophthalmol 1999; Idiopathic polypoidal choroidal
Multiple recurrent serous sanguineous 117:1503–1510. vasculopathy of the macula.
retinal pigment epithelial detachments in 20. Gomez-Ulla F, Gonzalez F, Torreiro MG: Ophthalmology 1998; 105:1380–1385.

CHAPTER 151
black women. Am J Ophthalmol 1985; Diode laser photocoagulation in idiopathic 35. Yannuzzi LA, Nogueira FB, Spaide RF, et al:
100:560–569. polypoidal choroidal vasculopathy. Retina Idiopathic polypoidal choroidal
4. Yannuzzi LA, Sorenson J, Spaide RF, 1998; 18:481–483. vasculopathy: a peripheral lesion. Arch
Lipson B: Idiopathic polypoidal choroidal 21. Green WR, McDonnel PJ, Yeo JH: Ophthalmol 1998; 116:382–383.
vasculopathy. Retina 1990; 10:1–8. Pathological features of senile macular 36. Iijima H, Iida T, Imai M, et al: Optical
5. Spaide RF, Yannuzzi LA, Slakter JS, et al: degeneration. Ophthalmology 1985; coherence tomography of orange-red
Indocyanine green videoangiography of 92:615–627. subretinal lesions in eyes with idiopathic
idiopathic polypoidal choroidal 22. Okubo A, Sameshima M, Uemara A: polypoidal vasculopathy. Am J Ophthalmol
vasculopathy. Retina 1995; 15:100–110. Clinicopathological correlation of polypoidal 2000; 129:21–26.
6. Ross RD, Gitter GA, Cohen C, Shoemaker choroidal vasculopathy revealed by 37. Spaide RF, Guyer DR, McCormick B, et al:
KS: Idiopathic polypoidal choroidal ultrastructural study. Br J Ophthalmol 2002; External beam radiation therapy for
vasculopathy associated with retinal arterial 86:1093–1098. choroidal neovascularization.
macroaneurysm and hypertensive 23. Otsuji T, Takahasi K, Fukushima I, Uyama M: Ophthalmology 1998; 105:24–30.
retinopathy. Retina 1996; 16:105–111. Optical coherence tomographic findings of 38. Yang SS, Fu AD, McDonald HR, et al:
7. Yannuzzi LA, Ciardella AP, Spaide RF, et al: idiopathic polypoidal choroidal Massive spontaneous choroidal
The expanding clinical spectrum of vasculopathy. Ophthalmic Surg Lasers hemorrhage. Retina 2003; 23:139–144.
idiopathic polypoidal vasculopathy. Arch 2000; 31:210–214. 39. Smith RE, Wise K, Kingsley RM: Idiopathic
Ophthalmol 1997; 115:478–485. 24. Pauleikhoff D, Löffert D, Spital G, et al: polypoidal choroidal vasculopathy and
8. Ciardella AP, Donsoff IM, Huang SJ, et al: Pigment epithelial detachment in the sickle cell retinopathy. Am J Ophthalmol
Polypoidal choroidal vasculopathy. Surv elderly. Clinical differentiation, natural 2000; 129:105–111.
Ophthalmol 2004; 49:25–37. course and pathogenic implications. 40. Bartlett HM, Willoughby B, Mandava N:
9. Perkovich BT, Zakov ZN, Berlin LA, et al: Graefe’s Arch Clin Exp Ophthalmol 2002; Polypoidal choroidal vasculopathy in a
An update on multiple recurrent 240:533–538. patients with melanocytoma of the optic
serosanguineous retinal pigment epithelial 25. Ahuja RM, Stanga PE, Vingerling JR, et al: nerve. Retina 2001; 21:396–399.
detachments in black women. Retina 1990; Polypoidal choroidal vasculopathy in 41. Katsimpris JM, Petropoulas IK,
10:18–26. exudative and hemorrhagic pigment Pharmakakis NM, Pournaras CJ: Idiopathic
10. Lafaut BA, Leyes AM, Snyders B, et al: epithelium detachments. Br J Ophthalmol polypoidal choroidal vasculopathy
Polypoidal choroidal vasculopathy in 2000; 84:479–484. associated with central retinal vein
Caucasians. Graefe’s Arcg Clin Exp 26. Lip PL, Hope-Ross MW, Gibson JM: occlusion. J Fr Ophtalmol 2003;
Ophthalmol 2000; 238:752–759. Idiopathic polypoidal choroidal 26:489–492.
11. Scassellati-Sforzolini B, Mariotti C, Bryan vasculopathy: a disease with diverse 42. Iijima H, Imai M, Gohodo T, et al: Optical
R, et al: Polypoidal vasculopathy in Italy. clinical spectrum and systemic coherence tomography of idiopathic
Retina 2001; 21:121–125. associations. Eye 2000; 5:695–700. polypoidal choroidal vasculopathy. Am J
12. Yannuzzi LA, Slakter JS, Sorenson JA, et al: 27. Mohand-Said M, Nodarian M, Salvanet- Ophthalmol 1999; 127:301–305.
Digital indocyanine videoangiography and Bouccara A: Idiopathic polypoidal choroidal 43. Giovannini A, Amato GP, D’Altobrando E,
choroidal neovascularization. Retina 1992; vasculopathy: 2 case report. J Fr Giuliani M: Optical coherence tomography
12:191–223. Ophthalmol 2002; 25:517–521. (OCT) in idiopathic polypoidal choroidal
13. Ferris FL III: Senile macular degeneration: 28. Schneider U, Gelisken F, Inhoffen W: vasculopathy. Doc Ophthalmol 1999;
a review of epidemiological features. Am J Clinical characteristics of idiopathic 97:367–371.
Epidemiol 1983; 118:213–221. polypoidal choroidal vasculopathy. 44. Lois N: Idiopathic polypoidal choroidal
14. Capone A Jr, Wallace RT, Meredith TA: Ophthalmologe 2001; 98:1186–1191. vasculopathy in a patient with atrophic
Symptomatic choroidal neovascularization 29. Kwok AKH, Lai TYY, Chan CWN, et al: age-related macular degeneration. Br J
in blacks. Arch Ophthalmol 1994; Polypoidal choroidal vasculopathy in Ophthalmol 2001; 85:1011–1012.
112:1091–1097. Chinese patients. Br J Ophthalmol 2002; 45. Gass JDM. Stereoscopic atlas of macular
15. Lafaut BA, Aisenbrey S, van den Broecke 86:892–897. diseases. St Louis: CV Mosby; 1997.
C, et al: Polypoidal choroidal vasculopathy 30. Imaizumi H, Takeda M: Knobby-like 46. Arnold JJ, Sarks SH, Killingworth MC,
pattern in age-related macular choroidal neovascularization accompanied Sarks JP: Reticular pseudodrusen. Retina
degeneration. A clinicopathologic with retinal pigment epithelial detachment. 1995; 15:183–191.
correlation. Retina 2000; 20:650–654. Nippon Ganka Gakkai Zasshi 1999; 47. Pauleikhoff D, Radermacher M, Spital, et al:
16. Uyama M, Wada M, Nagai Y, et al: 103:527–537. Visual prognosis of second eyes in patients
Polypoidal choroidal vasculopathy: natural 31. Iida T, Yannuzzi LA, Freund KB, et al: with unilateral late exudative age-related
history. Am J Ophthalmol 2002; Retinal angiopathy and polypoidal macular degeneration. Graefe’s Arch Clin
133:639–648. choroidal vasculopathy. Retina 2002; Exp Ophthalmol 2002; 240:539–542.
17. MacCumber MW, Dastgheib K, Bressler 22:455–463. 48. Guyer DR, Yannuzzi LA, Ladas I, et al:
NM, et al: Clinicopathological correlation of 32. Tateiwa H, Kuroiwa S, Gaun S, et al: Indocyanine green guided laser
the multiple recurrent serosanguineous Polypoidal choroidal vasculopathy with photocoagulation of focal spots at the
retinal pigment epithelium detachments large vascular network. Graefe’s Arch Clin edge of plaque of choroidal vascularization.
syndrome. Retina 1994; 14:143–152. Exp Ophthalmol 2002; 240:354–361. Arch Ophthalmol 1996; 114:693–697. 1995
RETINA AND VITREOUS

49. Yuzawa M, Mori R, Haruyama M: A study 55. Fujii GY, Pieramici DJ, Humayun MS, et al: 61. Chan WM, Lam DS, Lai TY, et al:
of laser photocoagulation for polypoidal Complications associated with limited Photodynamic therapy with verteporfin for
choroidal vasculopathy. Jpn J Ophthalmol macular translocation. Am J Ophthalmol symptomatic polypoidal choroidal
2003; 47:379–384. 2000; 130:751–762. vasculopathy: one-year results of a
50. Gass JDM: Biomicroscopical and 56. Marcus D, Sheils W, Johnson M, et al: prospective case series. Ophthalmology
histopathologic considerations regarding External beam irradiation of subfoveal 2004; 111:1576–1584.
the feasibility of surgical excision of choroidal neovascularization complicating 62. Quaranta M, Mauget-Faysse M, Coscas G:
subfoveal neovascular membranes. Am J age-related macular degeneration: one- Exudative idiopathic polypoidal
Ophthalmol 1994; 118:285–298. year results of a prospective, double- vasculopathy and photodynamic therapy
51. Shiraga F, Matsuo T, Yokoe S, et al: masked, randomized clinical trial. Arch with verteporfin. Am J Ophthalmol 2002;
Surgical treatment of submacular Ophthalmol 2001; 119:171–180. 134:277–280.
hemorrhage associated with idiopathic 57. Spaide RF, Leys A, Herrman-Delemazure B, 63. Hussain N, Hussan A, Natarajan S: Role of
polypoidal choroidal vasculopathy. Am J et al: Radiation associated choroidal photodynamic therapy in polypoidal
Ophthalmol 1999; 128:147–152. neovasculopathy. Ophthalmology 1999; choroidal vasculopathy. Indian J
52. Merill PT, LoRusso FJ, Lomeo MD, et al: 106:2254–2260. Ophthalmol 2005; 53:101–104.
Surgical removal of subfoveal choroidal 58. Reichel E, Berrocal AM, Ip M, et al: 64. Lee SC, Seong YS, Kim SS, et al:
neovascularization in age-related macular Transpupillary thermotherapy of occult Photodynamic therapy with verteporfin for
degeneration. Ophthalmology 1999; subfoveal choroidal neovascularization in polypoidal choroidal vasculopathy of the
106:782–789. patients with age-related macular macula. Ophthalmologica 2004;
53. Thomas MA, Grand MG, Williams DF, et al: degeneration. Ophthalmology 1999; 21:193–201.
Surgical management of subfoveal 106:1908–1914. 65. Rogers AH, Greenberg PB, Martidis A,
SECTION 10

choroidal neovascularization. 59. Spaide RF, Donsoff I, Lam DL, et al: Puliafito CA: Photodynamic therapy of
Ophthalmology 1992; 99:952–968. Treatment of polypoidal choroidal polypoidal choroidal vasculopathy.
54. Yazawa M, Isomae T, Mori R, Shimade H, vasculopathy with photodynamic therapy. Ophthalmic Surg Lasers Imaging 2003;
Utsunomiya. I. Surgical excision versus Retina 2002; 22:529–535. 34:60–63.
laser photocoagulation for subfoveal 60. Silva RM, Figueira J, Cachulo ML, et al:
choroidal neovascularization membranes Polypoidal choroidal vasculopathy and
with age-related macular degeneration: photodynamic therapy with verteporfin.
comparison of visual outcomes. Jpn J Graefes Arch Clin Exp Ophthalmol 2005;
Ophthalmol 2001; 45:192–198. 243:973–979.

1996
CHAPTER

152 Angioid Streaks


Eric Chen, Allen C. Ho, and David R. Guyer

HISTORICAL PERSPECTIVE is no gender or racial predilection. Angioid streaks are almost


always bilateral, and can be distinguished from blood vessels as
they lie beneath the retina and above the choroidal vasculature.
Key Features Angioid streaks are within two disk diameters of the optic nerve
• Angioid streaks are irregular breaks in a calcified and in 27% of cases and are widespread in 73% of patients.6 They
thickened Bruch’s membrane usually taper as they extend away from the disk, and typically
• They radiate outward from the optic nerve in a pattern similar do not go past the equator. The streaks can be wide or narrow
to retinal vasculature and can vary in number from one to many. There are often
• They are almost always bilateral, and there is no gender or associated peripapillary chorioretinal changes.
racial predilection The color of the streaks can range from red to dark brown,
• They are not present at birth but may progress over time and is often dependent on the pigmentation of the fundus and
• Associated findings include optic disk drusen, peau d’orange, degree of overlying retinal pigment epithelium (RPE) atrophy;
retinal hemorrhage, CNV, and macular degeneration they can appear gray if fibrovascular tissue is present. In one
• Systemic diseases are present in 50% of patients; most study,6 32% of streaks were gray, 23% were red, and 14% had
common are PXE, Paget’s disease, and the pigment proliferation with a blackish coloration. Hyperpigmen-
hemoglobinopathies tation or atrophy of the RPE may occur at the margin of the
streak.
The streaks themselves are usually asymptomatic, but
Angioid streaks were first described by Doyne1 in 1889, and in associated complications may cause vision loss. Subretinal
1892 Knapp2 coined the term as he thought that they resembled hemorrhage or choroidal rupture may occur even after minor
blood vessels. It was not until 1917 that Kofler3 correctly stated trauma.7 However, the main cause of vision loss in these
that the main pathologic changes were at the level of Bruch’s patients is CNV, RPE detachment, and macular degeneration
membrane. This finding was confirmed histopathologically in (Fig. 152.2). In one series,6 macular degeneration was observed
the late 1930s.4 in 40 (72%) of 56 cases; 32 patients had exudative maculopathy,
The recognition of angioid streaks is important because they and eight patients had atrophic maculopathy. In another study
can be associated with choroidal neovascularization (CNV) and of 110 cases, the occurrence of exudative macular degeneration
macular degeneration and can herald the presence of systemic was associated with the length of the streak, the distance of the
disorders, such as pseudoxanthoma elasticum (PXE), Paget’s streak from the fovea, and the diffuse or ‘cracked eggshell’ type
disease, and the hemoglobinopathies (Table 152.1). of streak.8 A case of an RPE tear associated with angioid streaks
has also been reported.9
Other associated findings include peau d’orange changes
TABLE 152.1 Common Systemic Findings Associated with (Fig. 152.3), light margins along streaks, peripheral focal lesions
Angioid Streaks
(salmon spots), hemorrhage, paired red spots along the streaks,
Finding % and disk drusen. In one series,6 these lesions were observed in
61%, 57%, 44%, 35%, 19%, and 10% of patients, respectively.
PXE 34 Some of these lesions, such as disk drusen, salmon spots, and
Paget’s disease 10 peau d’orange changes, are exclusively or more commonly
noted in cases of angioid streaks associated with PXE.
Hemoglobinopathy 6
Idiopathic 50 ANCILLARY TEST FINDINGS
From Clarkson JG, Altman RD: Angioid streaks. Surv Ophthalmol 1982;
26:235–246. Many authors6,10–13 have observed early hyperfluorescence of
the streaks with late staining (see Fig. 152.1). Others,6,13,14 how-
ever, have reported hypofluorescence of the streaks with hyper-
CLINICAL FINDINGS fluorescence at the margins, which stain late; hypofluorescence
is caused by separation of the underlying choriocapillaris and
Angioid streaks are irregular, spoke-like, and curvilinear streaks subsequent nonperfusion.13 In some cases, fluorescein angio-
that radiate outward from the peripapillary area in all directions graphy and red-free images may reveal angioid streaks not
(Fig. 152.1). They are not present at birth, but have been docu- observed clinically.10,15 The classic findings of associated CNV,
mented as early as childhood, with subsequent growth.5 There RPE detachments, and serous or hemorrhagic detachments may 1997
RETINA AND VITREOUS

FIGURE 152.1. (a–c) Angioid streaks are


irregular, curvilinear, reddish brown streaks that
radiate outward from the peripapillary area.
(d) Fluorescein angiography reveals
hyperfluorescence of the streaks.

a
c
SECTION 10

b d

FIGURE 152.2. This patient with angioid


streaks (a) has the characteristic ‘plucked-
chicken’ skin lesions (b) of PXE. (c) and (d) The
patient experienced CNV, which occurs in a
high percentage of patients with angioid
streaks.

a b

c d

be observed on fluorescein angiography or indocyanine green NATURAL HISTORY


angiography (ICGA), which may be superior in visualizing
occult CNV.16 The peau d’orange changes, which may represent Although patients with angioid streaks are usually asymp-
focal defects of Bruch’s membrane and the choriocapillaris, tomatic early in the course of the condition, visual loss usually
produce hypofluorescent areas on fluorescein angiography and a occurs with time. In one report,17 vision of 20/200 or worse was
speckled pattern in the midperiphery on ICGA.13 noted in most eyes after 50 years of age, while in another
Usually, visual fields, color testing, ERG, EOG, and dark series,18 greater than 50% of patients had an initial visual acuity
1998 adaptation tests are all normal. of 20/40 or better, but more than half of the patients were legally
Angioid Streaks

FIGURE 152.3. (a) Following trauma with a


bungee cord, a patient with angioid streaks had
multiple areas of subretinal hemorrhage with
central macula involvement, with a drop of
visual acuity to 20/200. Color montage photos
show spontaneous resolution of the
hemorrhage and unmasking of a choroidal
rupture nasal to fixation at one (b), two (c), and
3 months (d) after initial presentation, with an
ultimate improvement in vision to 20/30.

a b

CHAPTER 152
c d

blind at an average follow-up of 3.6 years. In another study of 131 patients with angioid streaks had PXE. Many other sys-
of 29 cases, 66% of patients had a visual acuity of 20/200 temic disorders have been associated with angioid streaks, but
or worse. some of these associations may be coincidental (Table 152.2).
The cause of this vision loss is macular degeneration or CNV, PXE, Paget’s disease, and the hemoglobinopathies remain the
or both (see Fig. 152.2). Three separate studies have reported most commonly found associated systemic diseases.
macular degeneration in ~70% of patients with angioid
streaks.6,19,20 Macular degeneration has even been observed in a
14-year-old patient.19 PSEUDOXANTHOMA ELASTICUM
Unlike age-related macular degeneration, the exudative form PXE is a form of systemic elastorrhexis that mainly affects the
is more common than the atrophic type of maculopathy in skin, eyes, gastrointestinal system, and heart.23 Females are
patients with angioid streaks.18,21 However, the exudative form affected twice as often as males. Patients usually are diagnosed
is found less frequently in patients with angioid streaks and sickle in the third to fourth decades of life. The inheritance of this rare
cell disease than in patients with other associated systemic disorder is usually autosomal recessive but may be autosomal
conditions.22 Macular degeneration is not always associated dominant, and the underlying genetic defect has been localized
with a foveal angioid streak, and it does not occur in all patients to chromosome 16 and a mutation in the transport protein
with a streak through the fovea.10 ABC-C6.24,25 It has been thought that the condition is caused
Piro and associates18 studied 62 patients with angioid streaks by elastic fiber abnormalities with secondary calcification. How-
and found that 86% had CNV in at least one eye. Bilateral CNV ever, the earliest finding in PXE was determined by cytoimmuno-
was noted in 49% of patients, and 37% of patients had uni- chemistry and X-ray analysis to be an accumulation of
lateral CNV. Five fellow eyes of 22 patients with unilateral CNV polyanions in the dermis.26 These authors speculate that the
developed CNV during a mean follow-up period of 18 months. polyanions attract calcium, which causes mineralization. Thus,
Mansour and co-workers8 reported that the CNV was associ- the basic defect in PXE may not be one of calcium or elastin
ated with streak length, streak distance from the fovea, and the metabolism but rather of glycosaminoglycans and glyco-
diffuse type of streak in their series. proteins. These glycosylated molecules may attach to elastic
Minor trauma may cause subretinal hemorrhage, often with fibers, mineralize, and cause abnormal collagen synthesis.
macular involvement (Fig. 152.3).4,7,10,22 Patients should be The characteristic skin change in PXE is a redundant waxy,
warned to avoid trauma and contact sports and to monitor their yellow, papule-like lesion, which commonly affects the neck,
vision regularly with an Amsler grid for the onset of CNV. face, abdomen, axillary areas, inguinal regions, periumbilical
area, and oral mucosa (see Fig. 152.2). This skin lesion looks
SYSTEMIC ASSOCIATIONS like a ‘plucked chicken’. Skin biopsy results reveal elastic tissue
staining of the deep dermis, often with calcification. Lebwohl
Clarkson and Altman23 found an associated systemic disease in and associates27 performed biopsies on scar and normal flexural
50% of the 50 patients in their series. PXE, Paget’s disease, and skin in patients suspected of having PXE without the typical
hemoglobinopathy were diagnosed in 34%, 10%, and 6% of the clinical skin findings. Six of 10 scar biopsy results showed
cases, respectively (see Table 152.1). Piro and associates18 noted fragmentation of elastic tissue, and three normal flexural skin
PXE in 61% of their patients and no systemic disease in 35% biopsy results showed signs of PXE. These authors concluded
of the patients. Federman and colleagues13 reported PXE in 30 that scar biopsy is useful in suspected cases of PXE without the
(54%) of their 56 patients. In 1941, Scholz20 reported that 59% characteristic clinical skin lesions. 1999
RETINA AND VITREOUS

Strandberg syndrome in their honor. Angioid streaks are present


TABLE 152.2. Other Systemic Disorders Associated with
in ~85% of patients with PXE.19,23 The streaks usually occur
Angioid Streaks
in early adulthood but can occur in patients as young as age
Acromegaly 8 years.5
Ehlers–Danlos syndrome
The peau d’orange changes represent a diffuse mottling of
the RPE in the temporal midperiphery and consist of multiple
Facial angiomatosis yellowish RPE lesions that have the appearance of the ‘skin of
Heterotopic calcification with hyperphosphatemia an orange’ (see Fig. 152.4). These lesions may be observed
before the occurrence of the angioid streaks.31 They are usually
Idiopathic thrombocytic purpura
seen in association with PXE but occasionally can be present in
Multiple hamartoma syndrome with uterine cancer cases of Paget’s disease or sickle hemoglobinopathy.23
Ocular melanocytosis
The salmon spot31 is a multifocal, yellow, atrophic, and
peripheral RPE lesion that appears ‘punched-out’ like a
Lead poisoning histoplasmosis syndrome spot. Macular drusen and atypical
Familial polyposis drusen may be present in up to 75% of cases.21 The presence of
all five varieties of pattern dystrophy in patients with PXE and
Hypo- and abetalipoproteinemia
angioid streaks has been reported by Agarwal,32 and Kadri and
Chronic familial hyperphosphatemia colleagues15 described intraretinal bands from the optic disk,
SECTION 10

Diabetes peripheral retinal degeneration, and bilateral retinal detach-


ments in one case. Multiple small crystalline bodies may be
Hemochromatosis present in 75% of patients in the midperipheral or juxta-
Acquired hemolytic anemia papillary regions.10 These crystalline bodies are associated with
atrophic RPE changes and may resemble a comet when RPE
Hypercalcinosis
atrophy occurs in a tail-like configuration next to it. Vascular
Hyperphosphatemia abnormalities, including chorioretinal arteriovenous communi-
Myopia cations, have also been described.33
Optic disk drusen are found clinically or echographically in
Neurofibromatosis up to a quarter of patients with PXE and angioid streaks.
Senile elastosis Coleman and co-workers34 stated that the incidence of disk
drusen is 20–50 times greater in patients with PXE than in
Tuberous sclerosis
normal individuals. Shields and associates6 found disk drusen
Diffuse lipomatosis in 10% of the patients in their series, and Pierro and
Dwarfism colleagues35 observed them in 21.6% of their 58 patients.
Calcium-containing macromolecules may attach to the elastic
Epilepsy fibers at the cribriform plate, disrupt axonal flow, and produce
Nephrolithiasis disk drusen, which may be the first manifestation of ocular
PXE.34 The peau d’orange and other pigmentary lesions may
Congenital dyserythropoietic anemia (CDA III)
precede angioid streaks, which may not develop until the third
Trauma to fourth decades of life. The angioid streaks then lead to CNV,
disciform scarring, and macular degeneration.

Other systemic findings in PXE include cerebral ischemia, PAGET’S DISEASE


cerebrovascular accidents, intracranial aneurysms, claudication, Paget’s disease or osteitis deformans is a chronic, progressive
hypertension, myocardial infarction, and gastrointestinal connective tissue disorder that involves the collagen matrix of
hemorrhage with or without ulceration. The gastrointestinal bone.23 Osteoclastic activity with an osteoblastic reaction
hemorrhage may be life-threatening, can occur in up to 15% of occurs. The condition may be due to a slow virus related to
patients, and may occur before the skin or eye findings. measles or to the respiratory syncytial virus36 or may be trans-
The first ocular findings in PXE were reported in 1903.28 The mitted in an autosomal dominant manner. Males and females
first report of the association between PXE and angioid streaks are equally affected. Systemic findings include an enlarged bone
was in 1929 by Groenblad29 and Strandberg.30 The ocular and mass affecting the pelvis, skull, spine, humeri, and femora,
cutaneous findings of PXE are referred to as the Groenblad– extraskeletal calcifications of the skin and arteries, pain,

FIGURE 152.4. (a and b) The peau d’orange


lesion is a diffuse mottling of the RPE in the
temporal midperiphery. These changes appear
as multiple, yellowish retinal pigment epithelial
lesions that have the appearance of an orange
skin ( peau d’orange).

a b
2000
Angioid Streaks

secondary osteoarthritis, neurologic damage, cardiac disease, conus, blue sclera, ectopis lentis, and retinal detachment, while
decreased hearing, hyperparathyroidism, and claudication. The systemic findings include cardiovascular disease, diaphragmatic
diagnosis is made by finding an elevated serum alkaline hernias, and diverticuli of the gastrointestinal and respiratory
phosphatase level, increased urinary calcium and total peptide tracts.
hydroxyproline levels, and characteristic radiographic findings. The other systemic conditions associated with angioid streaks
The first associations between Paget’s disease and angioid probably represent coincidental findings. These disorders include
streaks were made by Verhoeff in 192837 and Rowland in acromegaly,54 facial angiomatosis,55 heterotopic calcification
1929.38 Angioid streaks are found in 8–15% of patients with with hyperphosphatemia,56 idiopathic thrombocytic purpura,57
Paget’s disease.20,23 However, others have suggested that the multiple hamartoma syndrome with uterine cancer,58 ocular
association may be less common. Dabbs and Skjodt39 found melanocytosis,59 lead poisoning,60 familial polyposis,61 hypo-
only one (1.4%) of 70 patients with angioid streaks to also have and abetalipoproteinemia,62 chronic familial hyperphosphatemia,63
Paget’s disease. diabetes, hemochromatosis, acquired hemolytic anemia,
Clarkson and Altman23 studied 50 patients with active myopia, senile elastosis, tuberous sclerosis, hypercalcinosis,23
Paget’s disease. Angioid streaks were found in seven patients. hyperphosphatemia,56 neurofibromatosis,64 diffuse lipomatosis,65
Those with angioid streaks were found to have had a longer dwarfism,66 epilepsy,67 nephrolithiasis,68 congenital dyserythro-
duration of Paget’s disease, a high alkaline phosphatase level, poietic anemia (CDA III),69 and trauma1 (see Table 152.2).
more disease sites seen on X-ray film, and increased urinary
hydroxyproline excretion. In addition, six of the seven patients PATHOPHYSIOLOGY

CHAPTER 152
had skull involvement.
Occasionally, patients with Paget’s disease may have peau Although the pathogenesis of angioid streaks remains con-
d’orange lesions and other RPE changes.10,23 Visual loss is troversial, the elastic lamina of Bruch’s membrane seems to be
usually caused by CNV, but optic atrophy, sometimes secondary primarily affected, with secondary degeneration of the chorio-
to bony compression, may also occur. capillaris and RPE with disease progression. In PXE and Paget’s
disease, Bruch’s membrane may become calcified and brittle.
Adelung70 studied the lines of force in the eye resulting from
HEMOGLOBINOPATHIES traction of the ocular muscles pulling the eye around the fixed
The first association of angioid streaks with sickle cell disease site of the optic nerve, leading to the peripapillary origin and
was made in 1959 by Lieb and co-workers.40 In a selected radial extension of cracks occurring in a brittle Bruch’s mem-
population of patients with sickle cell disease, five (6%) of 69 brane. Calcification is observed in other parts of the body in
patients were found to have streaks.41 Condon and Serjeant42 both PXE and Paget’s disease, and thus this mineralization may
did not find any patients with angioid streaks in their study of be a common mechanism.
76 patients with homozygous sickle cell disease (sickle cell The cause of angioid streaks in sickle cell disease is even
anememia or hemoglobin SS disease). In another study of 124 more confusing. Calcification is not common in this disorder;
patients with hemoglobin SS disease, no patients with streaks studies have conflicted as to whether a diffuse elastic degener-
were observed.43 However, five (1.4%) of 356 patients in another ation is present in patients with sickle cell disease. Another
series44 had angioid streaks, and in Clarkson and Altman’s theory is that iron deposition in Bruch’s membrane may occur
report23 six patients with hemoglobin SS disease and one from hemolysis. This iron deposition could cause mineraliz-
patient with hemoglobin SC disease, had angioid streaks. The ation of Bruch’s membrane. However, only one study4
frequency of angioid streaks appears to be higher in elderly documented iron staining of Bruch’s membrane. Other studies
patients45; of 60 elderly patients with hemoglobin SS disease, could not confirm these findings. Jampol and associates71 stated
13 (22%) had streaks, whereas only three (2%) of 150 younger that calcification may be more important than iron deposition.
patients were observed to have angioid streaks. Another hypothesis for the occurrence of angioid streaks in
Hamilton and associates46 reported 21 of 242 patients with patients with sickle cell disease stated that the lesions occur
hemoglobin SS disease to have angioid streaks. Their patients from impaired nutrition caused by sickling and stasis.72 Small-
had good prognoses, as only two individuals had macular vessel occlusion could also be important in the pathogenesis.46
disease.
Angioid streaks have also been reported in association with PATHOLOGIC FEATURES
other hemoglobinopathies besides hemoglobin SS disease, such
as hemoglobin SC disease47; hereditary spherocytosis48; sickle The first histopathologic studies of angioid streaks were
trait (hemoglobin AS)49; b-thalassemia major, minor, and inter- reported in the late 1930s.4 Basophilia and calcification of a
media50; hemoglobin H disease51; and sickle cell thalassemia.52 thickened Bruch’s membrane were observed in both reports.
Overall, angioid streaks appear in 1–2% of patients with Breaks in Bruch’s membrane correlated with the clinical sites of
hemoglobinopathies, with the incidence increasing with age. the streaks. Elastic degeneration was noted, and some breaks
Complications, such as CNV or macular degeneration, are were invaded by fibrovascular tissue from the choroid. Thus,
uncommon with angioid streaks associated with sickle cell angioid streaks appear to be linear cracks in a thickened,
disease. degenerated, and calcified Bruch’s membrane.
It appears that the histopathologic appearance is similar
regardless of the associated systemic condition.23,71 Iron
OTHER SYSTEMIC ASSOCIATIONS deposition has not been found in cases of associated sickle cell
PXE, Paget’s disease, and the hemoglobinopathies are most disease except for one report.4,46,71,73 Dreyer and Green73 studied
commonly associated with angioid streaks. However, many the histopathologic features of 32 eyes from 21 cases. Two
other associated systemic conditions have been reported. patients had PXE, five patients had Paget’s disease, and 14
Angioid streaks have been found in patients with the patients had no systemic disorder. These authors confirmed
Ehlers–Danlos syndrome,53 a rare autosomal dominant connec- that angioid streaks are defects in the thickened, calcified elastic
tive tissue disorder with hyperextensible skin and hyperflexible layer of Bruch’s membrane. They found that the earliest change
joints caused by an abnormality in the synthesis and meta- was a break in the elastic and collagenous layers of Bruch’s
bolism of collagen. Ocular findings include high myopia, kerato- membrane. Fibrovascular ingrowth occurred in some of the 2001
RETINA AND VITREOUS

breaks. Secondary changes included thickening of the basement follow-up up to 6 months. Fujii et al82 described inferior limited
membrane of the RPE, RPE atrophy, choriocapillaris damage, macular translocation for angioid streak-associated subfoveal
photoreceptor loss, RPE hypertrophy or hyperplasia, serous CNV; two of four eyes had a final vision of 20/80 or better, with
retinal detachment, and disciform scar formation. CNV and one of these suffering from a retinal detachment, while another
serous retinal detachment were observed in two cases, and eye had extrafoveal and subfoveal recurrence at 8 weeks and
disciform scars were present in eight eyes. Salmon spots were 9 months, respectively.
found to be isolated breaks in Bruch’s membrane with fibro- Photodynamic therapy (PDT), using an intravenous photo-
vascular ingrowth. sensitive drug (verteporfin) that is activated with infrared laser,
Jensen74 studied Bruch’s membrane in PXE using histo- is another therapeutic option with mixed results in treating
chemical, ultrastructural, and X-ray microanalytic techniques. angioid streak-associated CNV. Karacorlu et al83 reported eight
He found two types of calcification: hydroxyapatite and eyes treated successfully, with short-term cessation of leakage
CaHPO4. He also observed a ‘thready’ material in the mem- on fluorescein angiogram and no deterioration in visual acuity
brane and an increased amount of acid mucopolysaccharide. over a mean of 8.7 months, while Shaikh and associates84
He stated that malformed collagen may be the underlying treated 11 eyes and found conversion of a CNV to a fibrous
abnormality in PXE. disciform lesion in nine eyes and lesion enlargement in seven of
these eyes.
TREATMENT Menchini and colleagues85 evaluated 48 eyes with baseline
vision of 20/200 or greater and found that most had no or
SECTION 10

As even trivial trauma can precipitate hemorrhage in patients limited vision loss after 1 year and suggested PDT could be used
with angioid streaks, safety glasses should be considered. In to limit or delay visual loss, while another report by Ladas et al86
addition, these patients should not engage in contact sports. with 24 eyes found unsatisfactory anatomic and functional
Low-vision aids may be useful, and in some cases, genetic results with PDT, even when re-treatments were performed
counseling should be considered. Prophylactic treatment of earlier than the conventional time of 3 months. Nineteen of the
angioid streaks should not be performed, as patients are 24 eyes had a final best-corrected visual acuity of 20/400 or
generally asymptomatic and treatment may induce CNV, but worse. Browning and co-workers87 found PDT seemed to limit
patients should be advised to use an Amsler grid regularly to visual loss in most patients through the first 12 months of
allow early detection of CNV. follow-up; in 16 eyes with subfoveal CNV, 12 lost fewer than
Although there have been no prospective randomized con- eight letters and 14 of the 16 lost fewer than 15 letters on
trolled trials of different treatment options for CNV associated ETDRS charts.
with angioid streaks, it seems reasonable to consider treatment Costa et al88 also described a novel technique of selective
of CNV to slow down progression of the CNV, decrease size of occlusion of subfoveal CNV in five patients with photo-
the central scotoma, or reduce a patient’s metamorphopsia. thrombosis of the neovascular ingrowth site with large-spot,
Successful outcomes with laser photocoagulation have been lower-intensity 810-nm laser to ICG concentrated in vascular
reported with well-defined juxta- or extrafoveal CNV, although lesions. They demonstrated rapid induction of CNV hypo-
the recurrence rate may be higher with CNV associated with perfusion within 1 h, and improvement of visual acuity and
angioid streaks than with CNV associated with other macular partial restoration of retinal architecture by optical coherence
disorders. Singerman and Hatem75 treated eight eyes with extra- tomography up to 12 months after treatment.
foveal CNV with laser photocoagulation. Improved or stabilized
vision was noted in seven of the eight cases. Recurrences DIFFERENTIAL DIAGNOSIS
occurred in four eyes. Piro and associates18 found a poor
response to laser photocoagulation in their series because of Only 39% of cases in one series6 were correctly diagnosed as
recurrences. Brancato and colleagues76 treated 13 such eyes and having angioid streaks before referral. Twenty-three percent of
stated that laser photocoagulation should be performed. Van these cases were misdiagnosed as age-related macular degener-
Eijk and Oosterhuis77 successfully treated seven of 15 eyes. ation. Other disorders that were confused with the angioid streaks
Eight eyes had subfoveal recurrences. Gelisken and associates78 included choroidal sclerosis, myopia and lacquer cracks, histo-
treated 30 such eyes. Sixteen of the eyes had stable or improved plasmosis, toxoplasmosis, retinal vasculitis and papilledema,
vision postoperatively. Twelve of the remaining 14 cases and traumatic hemorrhage. Other entities to consider in the
retained 20/200 vision or better during a mean follow-up period differential diagnosis include choroidal rupture and choroidal
of 3.4 years. Eleven untreated fellow eyes developed macular folds (Table 152.3).
degeneration and the loss of central vision.
In the largest series, Pece and co-workers79 treated 66 eyes
and found a significant decrease in vision in the first year, but
no significant change thereafter. They concluded laser treat- TABLE 152.3. Differential Diagnosis of Angioid Streaks
ment could end CNV and help stabilize visual acuity or slow
down visual loss, although a high frequency of recurrences (77% Age-related macular degeneration
of eyes) demanded intense clinical and angiographic follow-up, Choroidal sclerosis
mostly in the first 3 months after treatment.
The lack of treatment for subfoveal CNV led to developments Myopic lacquer cracks
in the surgical treatment of CNV in angioid streaks. Thomas Histoplasmosis
and associates80 first reported four eyes that underwent surgical
Toxoplasmosis
removal of subfoveal membranes, with a mean loss of one line
of vision and visual outcome at 20/200 or worse at 7 months; Retinal vasculitis and papilledema
recurrent neovascularization occurred in one eye. They pos- Traumatic hemorrhage
tulated that diffuse abnormalities in Bruch’s membrane seen in
angioid streaks made it difficult to preserve underlying RPE Choroidal rupture
after CNV removal. Adelberg and colleagues81 reported only one Choroidal folds
2002 eye out of five improving after surgery, with no recurrences at
Angioid Streaks

CONCLUSIONS (OR OVERVIEW) trivial trauma can cause significant subretinal hemorrhage, and
the occurrence of CNV and macular degeneration can cause
Angioid streaks are distinctive fundus lesions that represent devastating visual loss. Treatment options for angioid streaks
breaks in a calcified and thickened Bruch’s membrane. They are have shown mixed results. While laser photocoagulation,
an important diagnosis for the ophthalmologist to make, as submacular surgery, and PDT may all be attempted in an effort
they are associated with systemic disorders and their resultant to slow down progression of the CNV, decrease size of the
complications in 50% of cases; the most common diseases seen central scotoma, or reduce a patient’s metamorphopsia, long-
include PXE, Paget’s disease, and the hemoglobinopathies. term prognosis remains guarded, as the recurrence rate with
Angioid streaks themselves are usually asymptomatic, but any treatment is high.

Summary Boxes
Pseudoxanthoma Elasticum
• Usually diagnosed in the third to fourth decade of life, typically autosomal recessive, defect on chromosome 16 and protein ABC-C6
• Characteristic ‘chicken skin’ with redundant waxy, yellow, papule-like lesions
• Life-threatening complications include GI bleeding, cardiovascular and cerebrovascular disease
• Angioid streaks are present in ~85% of patients with PXE
• Other ocular associations include optic disk drusen, peau d’orange

CHAPTER 152
Paget’s Disease
• Chronic, progressive connective tissue disorder; osteoclastic bone activity with an osteoblastic reaction
• Enlargement of the skull, kyphoscoliosis, deafness, and deformities of long bones
• Angioid streaks present in <2% of patients
Hemoglobinopathies
• Angioid streaks have been found in 1–2% of patients with hemoglobinopathies, and have been observed with many varieties of
hemoglobinopathy
• Frequency of angioid streaks increases with age
• Complications such as macular degeneration and choroidal neovascular membranes are uncommon

REFERENCES
1. Doyne RW: Choroidal and retinal changes. 13. Federman JL, Shields JA, Tomer TL: 23. Clarkson JG, Altman RD: Angioid streaks.
The result of blows on the eyes. Trans Angioid streaks. II. Fluorescein angiographic Surv Ophthalmol 1982; 26:235–246.
Ophthalmol Soc UK 1889; 9:128. features. Arch Ophthalmol 1975; 93:951–962. 24. McKusick VA: Heritable disorders of
2. Knapp H: On the formation of dark angioid 14. Gass JDM, Clarkson JG: Angioid streaks connective tissue. 4th edn. St Louis:
streaks as an unusual metamorphosis of and disciform macular detachment in CV Mosby; 1972:475–520.
retinal hemorrhage. Arch Ophthalmol 1892; Paget’s disease (osteitis deformans). Am J 25. Struk B, Cai L, Zach S, et al: Mutations of
21:289–292. Ophthalmol 1973; 75:576–586. the gene encoding the transmembrane
3. Kofler A: Beitraege zur Kenntnis der 15. Kadri W, Rosen E, Harcourt B: Intraretinal transporter protein ABC-C6 cause
angioid Streaks (Knapp). Arch Augenheilkd changes in the Groenblad–Strandberg pseudoxanthoma elasticum. J Mol Med
1917; 82:134–149. syndrome. Br J Ophthalmol 1973; 2000; 78:282–286.
4. Hagedoorn A: Angioid streaks. Arch 57:588–592. 26. Walker ER, Frederickson RG, Mayes MD:
Ophthalmol 1939; 21:746–774; 935–965. 16. Arvas S, Akar S, Yolar M, et al: Optical The mineralization of elastic fibers and
5. Mansour AM, Ansari NH, Shields JA, et al: coherence tomography (OCT) and alterations of extracellular matrix in
Evolution of angioid streaks. angiography in patients with angioid pseudoxanthoma elasticum. Arch Dermatol
Ophthalmologica 1993; 207:57–61. streaks. Eur J Ophthalmol 2002 1989; 125:70–76.
6. Shields JA, Federman JL, Tomer TL, 12:473–481. 27. Lebwohl M, Phelps RG, Yannuzzi L:
Annesley WH Jr: Angioid streaks. I. 17. Groenblad E: ‘Angioid streaks’ – Diagnosis of pseudoxanthoma elasticum by
Ophthalmoscopic variations and diagnostic pseudoxanthoma elasticum. scar biopsy in patients without
problems. Br J Ophthalmol 1975; De Zusammenhang zwischen diesen characteristic skin lesions. N Engl J Med
59:257–265. gleichzeitig auftretenden Augen- und 1987; 317:347–350.
7. Fine SL: Angioid streaks. Int Ophthalmol Hautveran-derungen. Acta Ophthalmol 28. Hallopeau L: Nouvelle note sur un cas de
Clin 1977; 17:173–182. 1932; 1(Suppl):1–114. pseudoxanthome elastique. Ann Dermatol
8. Mansour AM, Shields JA, Annesley WH, 18. Piro PA, Scheraga D, Fine SL: Angioid Syph (Paris) 1903; 4:595–596.
et al: Macular degeneration in angioid streaks. Natural history and visual 29. Groenblad E: Angioid streaks –
streaks. Ophthalmologica 1988; 197:36–41. prognosis. In: Fine SL, Owens S, eds. pseudoxanthoma elasticum; vorlaeufige
9. Lim JI, Lam S: A retinal pigment epithelium Management of retinal vascular and Mitteilung. Acta Ophthalmol 1929; 7:329.
tear in a patient with angioid streaks. Arch macular disorders. Baltimore: Williams & 30. Strandberg J: Pseudoxanthoma
Ophthalmol 1990; 108:1672–1674. Wilkins; 1983:136–139. elasticum. Z Haut Geschlechtski 1929;
10. Gass JDM: Stereoscopic atlas of macular 19. Connor PJ, Juergens JL, Perry HO, et al: 31:689.
diseases, diagnosis and treatment. 3rd edn. Pseudoxanthoma elasticum and angioid 31. Shimizu K: Mottled fundus in association
St Louis: CV Mosby; 1987. streaks. A review of 106 cases. Am J Med with pseudoxanthoma elasticum. Jpn J
11. Gass JDM: Pathogenesis of disciform 1961; 30:537–543. Ophthalmol 1961; 5:111–113.
detachment of the neuroepithelium. VI. 20. Scholz RO: Angioid streaks. Arch 32. Agarwal A, Patel P, Gass JD: Spectrum of
Disciform detachment secondary to Ophthalmol 1941; 26:677–695. pattern dystrophy in pseudoxanthoma
heredodegenerative, neoplastic, and 21. Verhoeff FH: Histological findings in a case elasticum. Arch Ophthalmol 2005;
traumatic lesions of the choroid. Am J of angioid streaks. Br J Ophthalmol 1948; 123:923–928.
Ophthalmol 1967; 63:689–711. 32:531–544. 33. Secretan M, Zografos L, Guggisberg D:
12. Smith JL, Gass JDM, Justice J Jr: 22. Archer AB, Logan WC: Angioid streaks. In: Chorioretinal vascular abnormalities
Fluorescein fundus photography of angioid Krill AE, ed. Hereditary retinal and choroidal associated with angioid streaks and
streaks. Br J Ophthalmol 1964; diseases. Philadelphia: JB Lippincott; pseudoxanthoma elasticum. Arch
48:517–521. 1976:851–909. Ophthalmol 1998; 116:1333–1336.
2003
RETINA AND VITREOUS

34. Coleman K, Ross MH, McCabe M, et al: 54. Howard GM: Angioid streaks and 74. Jensen OA: Bruch’s membrane in
Disk drusen and angioid streaks in acromegaly. Am J Ophthalmol 1963; pseudoxanthoma elasticum. Histochemical,
pseudoxanthoma elasticum. Am J 56:137–139. ultrastructural, and X-ray microanalytical
Ophthalmol 1991; 112:166–170. 55. Kalina RE: Facial angiomatosis with angioid study of the membrane and angioid streak
35. Pierro L, Brancato R, Minicucci M, et al: streaks. Association of angioid streaks with areas. Graefes Arch Clin Exp Ophthalmol
Echographic diagnosis of Drusen of the a component of the Sturge–Weber 1977; 203:311–320.
optic nerve head in patients with angioid syndrome. Arch Ophthalmol 1970; 75. Singerman LJ, Hatem GF: Laser
streaks. Ophthalmologica 1994; 84:528–531. treatment of choroidal neovascular
208:239–242. 56. McPhaul JJ Jr, Engel F: Heterotopic membranes in angioid streaks. Retina
36. Altman RD: Paget’s disease. In: Talbot MD, calcification: hyperphosphatemia and 1981; 1:75–85.
ed. Clinical rheumatology. 1st and 2nd angioid streaks of the retina. Am J Med 76. Brancato R, Menchini U, Pece A, et al:
edns. New York: Elsevier/North-Holland; 1961; 31:488–492. Laser treatment of macular subretinal
1st edn, 1978:184–190; and 2nd edn, 57. Yatzkan DN: Angioid streaks of the fundus neovascularizations in angioid streaks.
1981:198–206. in association with posthemorrhagic Ophthalmologica 1987; 195:84–87.
37. Verhoeff FH: The nature and pathogenesis amaurosis. Am J Ophthalmol 1957; 77. Van Eijk AW, Oosterhuis JA:
of angioid streaks in the ocular fundus. 43:219–222. Kryptonlaserbehandlung von
Transactions of the section on 58. Allen BS, Fitch MH, Smith JG Jr: Multiple gefabneubildungen bei angioiden streifen.
ophthalmology of the American Medical hamartoma syndrome. A report of a new Klin Monatsbl Augenheilkd 1987;
Association. 1928:243. case with associated carcinoma of the 191:443–448.
38. Rowland WD: Bilateral caerulean cataract. uterine cervix and angioid streaks of the 78. Gelisken O, Hendrikse F, Deutman AF: A
Am J Ophthalmol 1933; 16:61–62. eyes. J Am Acad Dermatol 1980; long-term follow-up study of laser
SECTION 10

39. Dabbs TR, Skjodt K: Prevalence of angioid 2:303–308. coagulation of neovascular membranes in
streaks and other ocular complications of 59. Awan KJ: Ocular melanocytosis and angioid streaks. Am J Ophthalmol 1988;
Paget’s disease of bone. Br J Ophthalmol angioid streaks. J Pediatr Ophthalmol 105:299–303.
1990; 74:579–582. Strabismus 1980; 17:300–304. 79. Pece A, Avanza P, Galli L, Brancato R:
40. Lieb WA, Geeraets WJ, Guerry D III: Sickle- 60. DeSimone S, DeConciliis V: Strie angioidi Laser photocoagulation of choroidal
cell retinopathy: ocular and systemic della retina. Arch Ottalmologia 1958; neovascularization in angioid streaks.
manifestations of sickle-cell disease. Acta 62:161. Retina 1997; 17:12–16.
Ophthalmol (Copenh) Suppl 1959; 58:1–45. 61. Awan KJ: Familial polyposis and angioid 80. Thomas MA, Dickinson JD, Melberg NS,
41. Geeraets WJ, Guerry D III: Angioid streaks streaks in the ocular fundus. Am J et al: Visual results after surgical removal of
and sickle cell disease. Am J Ophthalmol Ophthalmol 1977; 83:123–125. subfoveal choroidal neovascular
1960; 49:450–570. 62. Duker JS, Belmont J, Bosley TM: Angioid membranes. Ophthalmology 1994;
42. Condon PI, Serjeant GR: Ocular findings in streaks associated with 101:1384–1396.
homozygous sickle cell anemia in Jamaica. abetalipoproteinemia. Arch Ophthalmol 81. Adelberg DA, Del Priore LV, Kaplan HJ:
Am J Ophthalmol 1972; 73:533–543. 1987; 105:1173–1174. Surgery for subfoveal membranes in
43. Majekodunmi SA, Akinyanju OO: Ocular 63. Iancu TC, Almagor G, Savir H: Ocular myopia, angioid streaks, and other
findings in homozygous sickle cell disease abnormalities in chronic familial disorders. Retina 1995; 15:198–205.
in Nigeria. Can J Ophthalmol 1978; hyperphosphatasemia. J Pediatr 82. Fujii GY, Humayun MS, Pieramici DJ, et al:
13:160–162. Ophthalmol Strabismus 1980; 17:220–223. Initial experience of inferior limited macular
44. Nagpal K, Asdourian G, Goldbaum M, 64. McWilliam RJ: On the histology of angioid translocation for subfoveal choroidal
et al: Angioid streaks and sickle streaks. Trans Ophthalmol Soc UK 1951; neovascularization resulting from causes
haemoglobinopathies. Br J Ophthalmol 71:243–249. other than age-related macular
1976; 60:31–34. 65. Bonamour MG: Stries angioides de la retina degeneration. Am J Ophthalmol 2001;
45. Condon PI, Serjeant GR: Ocular findings in et maladie de Launois–Bensaube 131:90–100.
elderly cases of homozygous sickle cell (lipomatose diffuse). Bull Soc Ophtalmol 83. Karacorlu M, Karacorlu S, Ozdemir H,
disease in Jamaica. Br J Ophthalmol 1976; 1959; 406. Mat C: Photodynamic therapy with
60:361–364. 66. Yoneyama T, Yoneyama K: Angioid streaks verteporfin for choroidal neovascularization
46. Hamilton AM, Pope FM, Condon PI, et al: and dwarfism. J Clin Ophthalmol 1960; in patients with angioid streaks. Am J
Angioid streaks in Jamaican patients with 14:1013–1022. Ophthalmol 2002; 134:360–366.
homozygous sickle cell disease. Br J 67. Blobner F: Angioid streaks and epilepsy. 84. Shaikh S, Ruby AJ, Williams GA:
Ophthalmol 1981; 65:341–347. Klin Monatsbl Augenheilkd 1935; 95:12. Photodynamic therapy using verteporfin
47. Condon PI, Serjeant GR: Ocular findings in 68. Fabre B, Bayle P, Bazex J, et al: for choroidal neovascularization in angioid
hemoglobin SC disease in Jamaica. Am J Pseudoxanthoma elasticum and streaks. Am J Ophthalmol 2003; 135:1–6.
Ophthalmol 1972; 74:921–931. nephrolithiasis. J Eur Acad Dermatol 85. Menchini U, Virgili G, Introini U, et al:
48. McLane NJ, Grizzard WS, Kousseff BG, Venereol 2005; 19:212–215. Outcome of choroidal neovascularization in
et al: Angioid streaks associated with 69. Sandstrom H, Wahlin A, Eriksson M, et al: angioid streaks after photodynamic
hereditary spherocytosis. Am J Ophthalmol Angioid streaks are part of a familial therapy. Retina 2004; 24:763–771.
1984; 97:444–449. syndrome of dyserythropoietic anaemia 86. Ladas ID, Georgalas I, Rouvas AA, et al:
49. Gerde LS: Angioid streaks in sickle cell trait (CDA III). Br J Haematol 1997; Photodynamic therapy with verteporfin of
hemoglobinopathy. Am J Ophthalmol 1974; 98:845–849. choroidal neovascularization in angioid
77:462–464. 70. Adelung JC: Zur genese der angioid streaks: conventional versus early
50. Gartaganis S, Ismiridis K, Papageorgiou O, streaks (Knapp). Klin Monatsbl Augenheilkd retreatment. Eur J Ophthalmol 2005;
et al: Ocular abnormalities in patients with 1951; 119:241–250. 15:69–73.
b thalassemia. Am J Ophthalmol 1989; 71. Jampol LM, Acheson R, Eagle RC, et al: 87. Browning AC, Chung AK, Ghanchi F, et al:
108:699–703. Calcification of Bruch’s membrane in Verteporfin photodynamic therapy of
51. Daneshmend TK: Ocular findings in a case angioid streaks with homozygous sickle choroidal neovascularization in angioid
of haemoglobin H disease. Br J Ophthalmol cell disease. Arch Ophthalmol 1987; streaks: one-year results of a prospective
1979; 63:842–844. 105:93–98. case series. Ophthalmology 2005;
52. Goldberg MF, Charache S, Acadio I: 72. Hogan JF, Heaton CL: Angioid streaks and 112:1227–1231.
Ophthalmologic manifestations of sickle systemic disease. Br J Dermatol 1973; 88. Costa RA, Calucci D, Cardillo JA, et al:
cell thalassaemia. Arch Intern Med 1971; 89:411–416. Selective occlusion of subfoveal choroidal
128:33–39. 73. Dreyer R, Green WR: The pathology of neovascularization in angioid streaks by
53. Green WR, Friedman-Kien A, Banfield WG: angioid streaks: a study of twenty-one using a new technique of ingrowth site
Angioid streaks in Ehlers–Danlos syndrome. cases. Trans Pa Acad Ophthalmol treatment. Ophthalmology 2003;
Arch Ophthalmol 1966; 76:197–204. Otolaryngol 1978; 31:158–167. 110:1192–1203.

2004
CHAPTER

153 Ocular Histoplasmosis Syndrome


Nancy M. Holekamp, Lawrence S. Halperin, Levent Akduman, and R. Joseph Olk

The ocular histoplasmosis syndrome (OHS) causes permanent lesions had positive skin test results, but only 4.4% of those
loss of central vision in 2000 young adults per year. The who had positive results had typical fundus lesions. Only one
organism Histoplasma capsulatum has been suspected as a individual had a disciform macular lesion. We conclude that
cause of granulomatous uveitis for many years. In 1951, Krause OHS is relatively uncommon, even in endemic areas. Typically,
and Hopkins1 reported a patient with atrophic chorioretinal 2–12% of the population in an endemic area has ocular findings
lesions with pigment change and hemorrhage. A histoplasmin consistent with OHS, and only 1 in 1000 experiences
skin test result was positive, and a chest radiograph film maculopathy.
showed calcified lung nodules. In 1960, Woods and Wahlen2 Davidorf and Anderson11 examined 353 school-aged children
published a report of a collection of patients who were residents after an acute epidemic infection on Earth Day in 1970. Forty-
of an area endemic for histoplasmosis and had clear vitreous, one percent of the children became acutely ill from histo-
peripheral atrophic spots, disciform macular lesions, positive plasmosis infection, and 85% had elevated serum titers,
histoplasmin skin test results, and pulmonary calcifications. compared with 10% of controls. Two years later, all children
The four signs of OHS are peripheral punched-out who had the infection had a positive skin test result, compared
chorioretinal lesions, juxtapapillary atrophic pigmentary with 23.4% of controls. However, the incidence of fundus
changes, disciform macular changes, and a clear vitreous. lesions was similar in infected children when compared with
This chapter reviews the epidemiology, clinical findings, controls, and this brought the association of histoplasmosis
evaluation, and treatment of OHS. infection and OHS into question. A more likely explanation is
that a 2 year period after exposure to histoplasmosis is not suffi-
EPIDEMIOLOGY cient time to assess the occurrence of OHS lesions. Anand and
colleagues reported observation of active and inactive retinal
Most individuals with OHS are between 30 and 40 years of age. lesions during an outbreak of histoplasmosis (Anand R, Luby JP,
Fundus scars may occur with equal frequency among blacks and Metrikin D, et al: Acquired histoplasma chorioretinitis. Paper
whites, although maculopathy is very rare in blacks. Baskin and presented at the Annual Meeting of the Retina Society, Santa Fe,
associates3 reported six blacks with OHS and macular lesions, NM, 6–10 Sep 1995).
five of whom had positive skin test results. Feman and Tilford12 found that six of eight patients with
positive systemic histoplasmosis cultures had chorioretinal
scars consistent with OHS. This again lends support to the
EPIDEMIOLOGIC STUDIES concept that H. capsulatum causes OHS.
The epidemiology of OHS has been studied by many
investigators and yet the relationship between H. capsulatum
infection and the manifestations of ocular disease remains an FELLOW EYE
enigma. A review of several of these reports provides varying Patients with a disciform process in one eye have posterior
aspects of the debate. atrophic lesions in the fellow eye ~25–59% of the time.13,14 If
Braunstein and co-workers4 reported 15 individuals in the one eye has a choroidal neovascular membrane (CNVM), and
United Kingdom who had the clinical findings of OHS but in a focal atrophic spot in the posterior pole is present in the
whom all skin test results were negative. These investigators second eye, that eye has between 8% and 27% chance of acquir-
concluded that there could be a different causative factor in the ing a CNVM in 3 years.13–17 More histoplasmosis spots in the
United Kingdom. posterior pole may lead to increased chances of activation. If no
Ellis and Schlaegel5 found the Mississippi and Ohio river macular atrophic spot is present in the posterior pole of the
valleys to be highly infected; 80% of the adult population second eye, there is less than 5% chance of a CNVM developing
had positive skin test results. This area includes 80 million in 5 years; however, new spots may develop. Close follow-up is
individuals, involving Missouri, Illinois, Indiana, Kentucky, needed for the fellow eye, especially if posterior histoplasmosis
Tennessee, and Mississippi (Fig. 153.1). Wheat and colleagues6 spots are present. An individual with bilateral macular
collected worldwide patterns of skin sensitivity to histoplasmin. histoplasmosis spots has 5% chance of a CNVM developing in
Feman and associates7 noted that 60% of the Tennessee popu- one eye within 5 years.
lation had positive skin test results, but only 44 new cases of The macular photocoagulation study (MPS) showed that
visual loss resulting from OHS occurred in a 6 month period. there is 9% risk of CNVM formation in 5 years in the fellow eye
Smith and Ganley and co-workers8–10 studied 842 individuals if a patient with OHS has an extrafoveal or juxtafoveal CNVM
in Walkersville, Maryland, of whom 60% had positive skin test in one eye.18 The study also showed that histo spots of any type
results. One-hundred percent of patients with highly suspicious in the macula tripled the risk for later development of CNVM. 2005
SECTION 10 RETINA AND VITREOUS

FIGURE 153.1. This world map demonstrates areas where histoplasmosis infection is endemic.
Reprinted from Ellis FD, Schlaegel TF: The geographic localization of presumed histoplasmic choroiditis. Am J Ophthalmol 1973; 75: 953–956.

Key Features CNVMs were preceded by an atypical histo spot in the macula
• If one eye has a CNVM, and the fellow eye has a macular histo in more than 75% of the cases. Eighty-one percent of all
spot, the fellow eye has an 8 to 27% chance of developing a patients in the MPS retained 20/20 vision in at least one eye in
CNVM in 3 years. 5 years and risk of legal blindness was very low (8% in bilateral
• CNVMs are preceded by a macular histo spot in more than and 1% in unilateral patients).
75% of cases.
• Laser photocoagulation is appropriate treatment for extrafoveal HISTOPATHOLOGIC FEATURES
and some juxtafoveal CNVM due to OHS, but not subfoveal
CNVM.
• The most common complication of laser photocoagulation for
ORGANISM AND SYSTEMIC INFECTION
CNVM due to OHS is recurrence. The recurrence rate exceeds H. capsulatum is a fungus that is present in its yeast form. The
30%. fungus is carried and deposited by droppings from chickens,
• No controlled trial has proven the efficacy of oral steroids in pigeons, blackbirds, and bats. The birds are not infected but
OHS. Today, oral steroids are rarely used for CNVM. carry the fungus on their feathers. Bats, however, are infected.
• Subfoveal CNVM have a poor prognosis, with only 15% In humans, the fungus is first inhaled into the lungs and is
spontaneously recovering visual acuity to 20/40. then disseminated into the blood stream. Acute pneumonitis
• Potential treatments for subfoveal CNVM due to OHS include with fever may ensue and an acute, life-threatening disease
photodynamic therapy, intraocular corticosteroids, submacular may rarely occur. Pulmonary granulomas form and heal with
surgery, and anti-VEGF drugs. They may be used alone or in calcification, and these may be seen on chest radiography. The
combination. systemic forms of disease rarely have ocular manifestations.
• The SST suggests that submacular surgery may be considered
for subfoveal CNVM when visual acuity is less than 20/100.
• At present, there are no published reports of anti-VEGF
OCULAR HISTOPATHOLOGIC FEATURES
therapy for CNVM due to OHS. However, this form of therapy There are several reports of H. capsulatum in eyes with OHS.19,20
2006 will likely prove beneficial for this disease. However, Roth’s20 report was refuted by Gass and Zimmerman.21
Ocular Histoplasmosis Syndrome

a b

CHAPTER 153
c d

FIGURE 153.2. Histopathology of histoplasmosis infection,


demonstrating organisms present in the cytoplasm of endothelial cells
using different staining techniques. (a) phase-contrast, (b) periodic acid
Schiff, (c) higher power of b, (d) Gomori methanamine silver, (e) Gomori
methanamine silver with 90 minute exposure.
From Scholz R, Green WR, Kutys R, et al: Histoplasma capsulatum in the eye.
Ophthalmology (1984; 91:1100–1104).

Ocular histopathologic studies have shown H. capsulatum in Subretinal pigment epithelium neovascularization extended
disseminated histoplasmosis, often in immunocompromised beyond the disciform process. The juxtapapillary changes resulted
hosts (Fig. 153.2).22–26 Some of these patients exhibited clinical from loss of RPE, loss of photoreceptors, and discontinuities in
signs of OHS as well.20,27 These cases and others have been well Bruch’s membrane. The peripheral lesions showed loss of the RPE,
summarized.26 scarring, and occasional lymphocytic infiltration (Fig. 153.3).
Meredith and associates28 reported cracks in Bruch’s membrane Pavan and Margo29 reported a patient with fundus findings
related to the growth of choroidal blood vessels into the subretinal suggestive of OHS in which histopathologic examination of the
space. The overlying retina showed loss of outer layers and excised membrane revealed the following: intense staining with
cystic degeneration. The retinal pigment epithelium (RPE) was visible edges in the late phases of fluorescein angiography and a
lacking in the center of the lesion but was clumped at the edges. well-circumscribed granuloma containing some eosinophiles. 2007
RETINA AND VITREOUS

EXPERIMENTAL ANIMAL MODEL


Smith and co-workers developed an experimental animal model
involving the injection of live fungus via the carotid artery.30–32
Most animals had positive skin test results and early acute
choroiditis resulting from mononuclear cell and macrophage
infiltration with phagocytosis of yeast. Lymphocytes were found
in the choroid, and damage to Bruch’s membrane occurred. Six
weeks later, yeast was rarely found. Atrophic lesions resulted
from loss of RPE associated with altered Bruch’s membrane
and lymphocytes in the choroid. Subclinical lesions occurred
in areas that were not detectable by clinical examination or
fluorescein angiography, but lymphocytes were seen under the
retina and RPE. Disappearing lesions occurred where lesions
were at one time visible but became invisible. There were lym-
phocytes in the choroid, but the RPE and retina were normal.
Chronic choroiditis occurred, with organisms disappearing in
6 weeks. This may be why amphotericin B has no effect on the
SECTION 10

disease. No macular disciform lesions developed. These


laboratory data support clinical findings. Disseminated
histoplasmosis causes acute lesions with yeast particles. Later,
chronic OHS shows lesions without yeast, but inflammatory
cells may still be present. Doubts concerning the cause and
effect of H. capsulatum and OHS could be explained by these
experimental data.

IMMUNOLOGY
Several theories exist that attempt to explain the pathogenesis
of OHS, including the following: FIGURE 153.3. Histopathologic specimen from a case of ocular
1. Immunologic responses to previous infection causes OHS. histoplasmosis syndrome (OHS). Discontinuities in Bruch’s membrane
The macular lesion may be a hypersensitivity reaction to (between arrows in bottom figure) allow choroidal vessels (asterisk in
dead organisms elsewhere in the eye. bottom figure) to grow in the subretinal or subretinal pigment epithelial
2. Macular disease in OHS is the result of re-infection with space.
histoplasmosis. From Meredith TA, Green WR, Key SN, et al: Ocular histoplasmosis:
clinicopathologic correlation of 3 cases. Surv Ophthalmol 1977; 22:189, 1977.
3. Some vascular decompensation in the choroid is
responsible for the findings in OHS.
All these studies support the concept that OHS is an
immunologic disorder. Interestingly, lymphocytes are found in
LYMPHOCYTE HYPERREACTIVITY scars and lesions in OHS and they also are found in disciform
Schlaegel and colleagues33 found altered skin test reactivity to lesions of age-related macular degeneration. Age-related
certain antigens in patients with OHS. Lymphocytes are macular degeneration is increasingly being thought of as arising
hyperreactive in patients with disciform lesions in OHS, and from an underlying chronic inflammatory disorder. Unfor-
some investigators believe there is an increased rate of macular tunately, there is no animal model for macular lesions in OHS
subretinal hemorrhage after histoplasmin skin testing. to study these questions further.
Others34,35 found that lymphocyte transformation may corre-
late with OHS activity. Patients with disciform lesions have CLINICAL CHARACTERISTICS
stimulated lymphocytes in comparison with patients with only
peripheral scars and controls. Brahmi and associates36 found See Table 153.1 for a listing of clinical characteristics of the
acute histoplasmin infection to be different from OHS in terms ocular histoplasmosis syndrome.
of T-cell studies. Lymphocyte marker CD38 was significantly
higher than controls in four patients with OHS-like syndrome
from the United Kingdom, which may correlate with poor T-cell PUNCHED-OUT CHORIORETINAL LESIONS
function in making these patients more susceptible to various Smith and co-workers41 found one to four punched-out chorio-
stimuli.37 retinal lesions per eye, juxtapapillary changes in 28% of eyes,
and bilateral changes in 62% of patients with OHS. Macular
lesions are rare but more visually devastating.
HISTOCOMPATIBILITY ANTIGENS The atrophic spots are small, irregular, roundish lesions in
Meredith and co-workers,38 in the Walkersville study, found the midperiphery and posterior pole. They may have pigment
no increase in human leukocyte antigen (HLA)-B7 for peri- on the edge or in the center and range from 0.3 to 0.7 disk
pheral lesions only but did find an association with HLA- diameter. Choroidal vessels may be seen throughout the
DRw2. There was a definite association between HLA-B7 and atrophic lesions (Fig. 153.4). Watzke and Claussen42 found that
macular lesions. Braley and colleagues39 found that 78% of atrophic lesions change shape, size, and pigmentation over
patients with OHS and macular disciform lesions were HLA- time.
B7-positive compared with 20% of controls. Godfrey and associ- Linear streaks of chorioretinal lesions of variable length,
ates40 found that 54.8% of macular lesions were HLA-B7 width, and pigmentation may form (Fig. 153.5). They are
2008 positive. usually equatorial, parallel to the ora, and average 3 clock hours
Ocular Histoplasmosis Syndrome

FIGURE 153.4. (a) Fundus photograph of


peripheral, punched-out lesions typical of OHS.
(b) Fundus photograph of atrophic macular
lesions.

a b

TABLE 153.1. Clinical Characteristics of the Ocular


Histoplasmosis Syndrome

CHAPTER 153
Punched-out chorioretinal lesions
Juxtapapillary chorioretinal atrophy
Choroidal neovascularization
No vitritis
No pigment epithelial detachment
Disseminated choroiditis – rare

FIGURE 153.6. Juxtapapillary changes include retinal pigment


epithelial hypertrophy and atrophy.

suggesting that longer follow-up leads to identification of more


new lesions.

JUXTAPAPILLARY CHORIORETINAL ATROPHY


The juxtapapillary changes in OHS are probably due to
juxtapapillary choroiditis that goes unrecognized, and this leads
to chorioretinal changes around the disk (Fig. 153.6). Various
investigators have found that 85–94% of patients with OHS
have juxtapapillary changes.13,33,48 The atrophic juxtapapillary
FIGURE 153.5. Fundus photograph shows the peripheral linear streak changes may lead to choroidal neovascularization that can
of histoplasmosis spots. decrease central vision (Fig. 153.7).49 Lewis and co-workers48
found that 3.8% of patients with juxtapapillary changes
experienced a juxtapapillary CNVM, and Cantrill and Burgess50
in length. Bottoni and colleagues43 found streaks in five patients found that 15% of patients with symptoms from a juxta-
who also had CNVMs. Fountain and Schlaegel44 found streaks papillary CNVM in one eye had a juxtapapillary CNVM in the
in 5% of their patients with OHS, but this was a skewed fellow eye. Gass and Wilkinson47 found that 10% of all CNVMs
population. in OHS were juxtapapillary. Gutman13 reported that 58% of
New lesions may form in areas previously seen to be normal patients with these types of lesions had visual acuity of 20/200
on clinical examination and fluorescein angiography. Schlaegel or worse, indicating that juxtapapillary CNVM may lead to
and associates45 said 26% of spots were newly developed over significant central vision loss.
5 years of follow-up, whereas other studies cited 9% and
16.6%.13,42,46 Gass and Wilkinson47 found only 1 out of 81
patients in whom a new peripheral lesion developed, but MACULAR LESION
Lewis and Schiffman14 re-evaluated 99 of their patients with The classic macular lesion associated with OHS is a well-
longer follow-up and found that 19% acquired new lesions, defined CNVM with serous detachment of the retina, a small 2009
RETINA AND VITREOUS

FIGURE 153.7. In this eye, juxtapapillary


changes led to the development of choroidal
neovascular membrane (CNVM). (a) The fundus
photograph shows evidence of gray subretinal
lesions along with subretinal blood, both of
which are signs of CNVM. (b) An early frame of
a fluorescein angiogram shows early
hyperfluorescence. (c) A later frame shows
massive leakage from the new vessel
membrane. (d) In another patient, a
juxtapapillary CNVM causes extensive retinal
striae.

a b
SECTION 10

c d

FIGURE 153.8. Macular lesion of OHS.


(a) Fundus photograph of a CNVM, with a gray
membrane, subretinal hemorrhage, subretinal
fluid, and pigment ring. (b) An early frame of a
fluorescein angiogram with early
hyperfluorescence demonstrates a juxtafoveal
CNVM.

a b

amount of subretinal hemorrhage and hard lipid exudate, and a between 200 and 2500 mm from the center of the FAZ. Lewis
pigment halo surrounding the active lesion (Fig. 153.8). and co-workers48 found that 69% of patients with extrafoveal
Symptoms of a CNVM include decreased vision, metamor- CNVMs had visual acuity of 20/40 or better at presentation.
phopsia, and blurring. Micropsia occasionally may be a
subjective complaint. Juxtafoveal CNVMs
Generally, the CNVM occurs at the edge of an old healed Juxtafoveal CNVMs have either a hyperfluorescent edge
chorioretinal scar in the macula, termed a ‘histo spot’. Melberg 1–200 mm from the center of the FAZ, with or without block-
and colleagues have identified this scar as the ‘ingrowth site’ of age (blood, pigment) through the center of the FAZ, or a
the neovascularization as it is associated with a discontinuity CNVM 200–2500 mm from the center, with blood or blocked
in Bruch’s membrane on histopathologic examination.51,52 fluorescence within 200 mm of the center. Lewis and co-
Macular lesions can develop in previously normal retina. It is workers48 found that 71% of patients with juxtafoveal CNVMs
more common, however, that reactivation of old lesions from OHS had visual acuity measuring 20/200 or worse.
accounts for new CNVMs, with a 20–23% activation rate.47,48 Gutman13 found that if the CNVM was inside the FAZ, there
CNVMs are classified by their location in relation to the was a 63% chance that visual acuity would be less than 20/200,
center of the foveal avascular zone (FAZ). The designations of but a 25% chance if the CNVM was outside the FAZ. Olk and
extrafoveal, juxtafoveal, and subfoveal are described. co-workers17 found that 65% of patients with juxtafoveal
membranes had a visual outcome of 20/200 or worse. Poor
Extrafoveal CNVMs prognostic clinical signs include subretinal blood, poor initial
Extrafoveal CNVMs have an edge of hyperfluorescence or vision, membrane close to the center of the FAZ, and large
2010 blockage of fluorescence, as shown on fluorescein angiography, CNVM size. Steroids have no effect on juxtafoveal lesions.
Ocular Histoplasmosis Syndrome

a b c

CHAPTER 153
d e f

FIGURE 153.9. (a) Histoplasmic choroiditis causes yellow choroidal infiltrates with a surrounding pigment ring. Fluorescein angiography
demonstrates early blocked fluorescence (b) and the intermediate (c) and late (d) staining of the lesion. These lesions can become atrophic with
time (e) and ultimately may develop secondary reactive hyperplasia of the retinal pigment epithelium (f).

Subfoveal CNVMs predominantly classic. There may be surrounding atrophy of


Subfoveal CNVMs have active leakage under the center of the the RPE, thought to be due to chronic fluid accumulation in
FAZ. Fourteen to sixteen percent of these patients recover visual untreated cases.
acuity of 20/40 spontaneously without laser treatment,17,48,53
and laser treatment through the center of the FAZ would almost
certainly decrease visual acuity to the 20/200 level immediately. DISSEMINATED CHOROIDITIS
A pilot study of laser treatment for subfoveal neovascular Disseminated choroiditis usually occurs in immunocom-
membranes in OHS has shown no benefit from the laser promised patients. The lesions present as yellow, circum-
treatment in eyes with new or recurrent subfoveal CNVMs scribed, elevated choroidal infiltrates with fuzzy edges and a
that are 3.5 disc areas or less in size and with vision between surrounding ring of pigment (Fig. 153.9). The foci of choroiditis
20/40 and 20/320.54 Therefore, these patients should not be may develop into a CNVM, with serous retinal detachment,
considered for thermal laser treatment. However, untreated hemorrhage, and retinal striae. Conway and colleagues57 have
50% or more of these eyes end up with very poor vision.17,47,55 shown that 93% of patients with macular histochoroiditis were
A favorable natural history has been predicted by the following: at least stabilized with systemic steroids.
age less than 30 years, small CNVM, and good fellow eye.
A Poor prognosis untreated has been found in eyes with initially
excellent vision and a CNVM that involved more than 50% of OPTIC DISC EDEMA
the FAZ. Thirty-six percent of patients less than 30 years of age Optic disc edema occurs rarely in patients with OHS.58,59 The
had visual acuity of 20/40 or better, establishing that the greater juxtapapillary findings in OHS may originate from undetected,
the area of FAZ covered by the CNVM, or the further the CNVM transient papillitis.
is beyond the center of the FAZ, the worse the prognosis.
EXOGENOUS HISTOPLASMIC
VITRITIS AND OHS ENDOPHTHALMITIS
The presence of vitritis almost always rules out the diagnosis of One case of exogenous histoplasmic endophthalmitis after
OHS. Any time vitreous cells are present, entities other than cataract extraction has been reported.60
OHS must be considered.
VITREOUS HEMORRHAGE
PIGMENT EPITHELIAL DETACHMENTS There has been one report of a patient with OHS in whom a
Gass56 and others believe that pigment epithelial detachments CNVM developed and resulted in breakthrough vitreous
are very rare in OHS. Occult and minimally classic CNVM hemorrhage.61 OHS can be considered in the differential
are also rare. The most prevalent type of CNVM in OHS is diagnosis of vitreous hemorrhage. 2011
RETINA AND VITREOUS

a b c

FIGURE 153.10. (a) Fundus photograph of a CNVM (inferior to the fovea) and histoplasmosis spots (superior to the fovea). (b) and (c)
Fluorescein angiograms of histoplasmosis spots. (b) shows early hypofluorescence and (c) shows late staining. Note how the new vessel
membrane leaks fluorescein dye and develops fuzzy margins late in the study. The histoplasmosis spots show staining of the sclera but stay
sharply demarcated.
SECTION 10

TABLE 153.2. Symptoms and Signs of Choroidal TABLE 153.3. Differential Diagnosis of the Ocular
Neovascularization Histoplasmosis Syndrome

Metamorphopsia Multifocal choroiditis and panuveitis


Visual blurring Birdshot choroidopathy
Serous detachment of retina Diffuse unilateral subacute neuroretinopathy
Subretinal hemorrhage Acute posterior multifocal placoid pigment epitheliopathy
Hard lipid exudate Vogt–Koyanagi syndrome
Pigment halo surrounding lesion Behçet’s disease

SUBRETINAL HEMORRHAGE
choroidal vessels and CNVM. There may be a halo of hypoflu-
Subretinal hemorrhage from CNVM due to OHS tends to be orescence around the CNVM which corresponds to a reactive
smaller than similar lesions from age-related macular layer of RPE cells trying to contain or ‘cocoon’ the neovascular
degeneration. Generally, they can be observed and will undergo tissue.
spontaneous resorbtion. However, subretinal hemorrhage from
a CNVM due to OHS requiring evacuation has been reported.62 DIFFERENTIAL DIAGNOSIS
FLUORESCEIN ANGIOGRAPHY The types of differential diagnoses are listed in Table 153.3.

INDICATIONS MULTIFOCAL CHOROIDITIS AND PANUVEITIS


Patients with no subjective complaints or findings of peripheral (PSEUDO-PRESUMED OHS)
or macular chorioretinal scars do not require fluorescein Dreyer and Gass63 reported on 28 patients with multifocal
angiography. The indications for fluorescein angiography choroiditis, old punched-out lesions, juxtapapillary atrophic
include symptoms of a CNVM, such as visual loss, metamor- changes, and CNVMs. Most patients were from nonendemic
phopsia, or blurring; clinical signs of CNVM; and the need to areas and had vitritis and decreased electroretinogram signals.
evaluate the fellow eye of a patient with OHS to check for the Of 16 patients who underwent skin testing, only 5 tested
presence of macular chorioretinal scars that would indicate an positive for histoplasmosis. This syndrome is especially difficult
increased risk to that eye (Table 153.2). It is vital to obtain a to differentiate from birdshot choroidopathy and diffuse
fluorescein angiogram in any patient with new symptoms. unilateral subacute neuroretinopathy. Deutsch and Tessler64
reported similar findings, however 43% of their patients were
black. The choroidal punched-out lesions in pseudo-presumed
FINDINGS OHS are smaller than those in OHS, some of the spots
The histoplasmosis ‘spots’ show early hypofluorescence with represent an active choroiditis, and vitritis may be present.
faint late staining (Fig. 153.10). These areas can change to early Systemic steroids may help the active lesions in pseudo-
staining with late leakage over time. Gass56 believes the presumed OHS.
punched-out lesions fluoresce but that this is due to scleral
reflection and not actual leakage of dye. The RPE and
choriocapillaris between the histoplasmosis lesions are normal BIRDSHOT (VITILIGINOUS) CHOROIDOPATHY
(this is not the case in age-related macular degeneration). Birdshot choroidopathy presents in an older age group than
Acute, yellow choroidal lesions stain with fluorescein.56 does OHS. Choroidal lesions are creamy, active spots without
CNVM stains early in a lacy pattern and leaks late, with the atrophy or pigmentation, and there are no juxtapapillary changes.
margins of the lesion becoming blurred. Subretinal fluid may Optic disc pallor occurs, but choroidal neovascularization is
collect dye late in the study. Subretinal blood allows fluor- rare. The electroretinogram is depressed, and fluorescein
2012 escence of retinal vessels but blocks fluorescence of the angiography shows disc leakage and cystoid macular edema.
Ocular Histoplasmosis Syndrome

treated scar. Others have reported similar cases.69,70


DIFFUSE UNILATERAL SUBACUTE Prophylactic laser photocoagulation is not recommended, as it
NEURORETINOPATHY is not useful in preventing CNVM formation.
Diffuse unilateral subacute neuroretinopathy is a unilateral
parasitic infestation. Early in the disease, there are clusters of
white spots, vitritis, and RPE changes between white lesions. AMPHOTERICIN B
The lesions are evanescent and change with the location of the Amphotericin B has been tried in the past in an attempt to
parasite. Late in its course, optic atrophy and arterial narrowing obliterate a histoplasmosis infection. Makley and co-workers67
develop. found no treatment benefit. Most investigators believe that
OHS lesions are probably sterile, inflammatory lesions contain-
ing no organisms, and therefore, antifungal agents play no role
ACUTE POSTERIOR MULTIFOCAL PLACOID in the treatment of OHS.
PIGMENT EPITHELIOPATHY
Usually occurring after upper respiratory infection, acute
posterior multifocal placoid pigment epitheliopathy leads to ANTIHISTAMINES
clustered lesions in the posterior pole, often with severe loss of Antihistamines were at one time thought to play a regulatory
vision. The lesions disappear after a brief period, usually with role for choroidal capillaries and have been tried in OHS
return of good vision. macular disciform lesions but without success.

CHAPTER 153
VOGT–KOYANAGI SYNDROME 6-MERCAPTOPURINE
The Vogt–Koyanagi syndrome is a granulomatous uveitis with 6-Mercaptopurine has been tried, but with no benefit.
infiltrative choroidal lesions. Exudative retinal detachment is
frequently present. Systemic symptoms such as tinnitus, MACULAR PHOTOCOAGULATION STUDY –
deafness, poliosis, vitiligo, and headache occur. OHS AND EXTRAFOVEAL CNVM
The ocular histoplasmosis section of the MPS was a multi-
BEHÇET’S DISEASE center, controlled clinical study, designed to answer specific
Behçet’s disease includes severe vasculitis with panuveitis. questions concerning whether laser treatment would prevent
Aphthous oral ulcers and genital lesions usually accompany the visual loss from CNVMs. The CNVM includes hyperfluor-
attacks of uveitis. escence, blood, pigment, and blocked fluorescence on
fluorescein angiography. Extrafoveal CNVMs are defined as
TREATMENT having hyperfluorescence on fluorescein angiography
200–2500 mm from the center of the FAZ (Fig. 153.11). The
diagnosis of OHS requires at least one atrophic chorioretinal
STEROIDS scar. Entrance into the MPS required visual acuity of 20/100 or
No controlled trial has proven the efficacy of steroids in OHS. better. Juxtapapillary CNVMs were included in the study only if
Many years ago, Schlaegel and colleagues45 considered systemic treatment would spare at least 1.5 clock hours of nerve fiber
steroids to be appropriate for acute flare-ups and possibly for layer in the maculopapular bundle.
long-term use to prevent visual loss. Schlaegel65,66 believed that Results of the MPS71 showed that at 24 months, a 6 line
steroids, if used, should be given in high doses (prednisone visual loss had occurred in 50% of the group receiving no
60–100 mg/day) and tapered very slowly over 1–2 yr. Makley treatment and in 22% of the treated group. At 36 months, the
and co-workers67 found some benefit if steroids were used early results were 45% for the group receiving no treatment and 10%
in the course of macular lesions. These studies were performed for the treated group.72 The effectiveness of laser treatment
before other treatment alternatives were available. Today, when compared with controls was present in all subgroups at all
steroids are rarely used for CNVM. However, some early cases stages of follow-up. Burgess73 concluded that any CNVM
of OHS may present with choroditis at the site of a macular completely outside the FAZ should be treated with laser.
‘histo spot’ without concomitant CNVM. It can be reasonable Brown and colleagues performed an analysis of the cost-
to try local corticosteroids (subtenon or intravitreal) or a short effectiveness of laser photocoagulation for extrafoveal CNVM
course of systemic corticosteroids in these cases, with close due to OHS and found it to be highly cost-effective from a
follow-up looking for the development of CNVM. patient preference-based point of view.74

DESENSITIZATION MACULAR PHOTOCOAGULATION STUDY –


OHS AND JUXTAFOVEAL CNVM
Schlaegel and associates45 desensitized OHS patients by giving
small doses of histoplasmin antigen subcutaneously. They The MPS75 also looked at juxtafoveal CNVM. The results of the
found that there was no difference between treated and control juxtafoveal group of the MPS at 1 year of follow-up showed that
groups. Some investigators report that skin testing or the risk of a six-line visual loss was 24.8% in the group who
desensitization may exacerbate macular lesions in OHS. received no treatment compared with 6.6% in the treated group.
The results were 27.9% versus 8.5%, respectively, at 5 year
follow-up.76 Untreated eyes were at much greater risk of a
LASER TREATMENT OF INACTIVE MACULAR six-line visual loss between 1 and 5 years of follow-up
LESIONS examination than were treated eyes at an unadjusted relative
Gitter and Cohen68 attempted to prevent CNVM formation in risk of 2.60 and 4.26 after adjusting for both visual acuity
fellow eyes by laser treatment of inactive macular lesions. Their and hypertension at the baseline.
study contained no controls, but no complications were Fine and associates77 separated the juxtafoveal CNVM in
reported. Later, one of the patients experienced a CNVM in a the MPS study into two types defined as the following: A 2013
RETINA AND VITREOUS

a b c
SECTION 10

d e f

FIGURE 153.11. (a) Schematic representation of an extrafoveal CNVM demonstrates the measurement from the center of the foveal avascular
zone to the edge of the CNVM, in this case 200 mm away. (b) Fundus photograph of a CNVM. (c) Fluorescein angiogram demonstrates early,
lacy hyperfluorescence. (d) Middle frame of the angiogram shows leakage of dye. (e) Posttreatment photograph. (f) Fluorescein angiogram
shows obliteration of the new vessel membrane. Note normal hyperfluorescence at the edge of a laser scar.
(a) From Poliner LS, Olk RJ: Ocular histoplasmosis syndrome. Semin Ophthalmol 1987; 2:238.

juxtafoveal CNVM was defined in this study as having a hyper- MACULAR PHOTOCOAGULATION STUDY –
fluorescent edge 1–200 mm from the center of the FAZ or a OHS (SUMMARY)
CNVM 200–2500 mm from the center, with blood or blocked
fluorescence within 200 mm of the center (Fig. 153.12). The current recommendation for the evaluation and treatment
Proximity to the fovea is crucial in juxtafoveal CNVMs. of extrafoveal, juxtafoveal, and peripapillary (nonsubfoveal)
Outside the FAZ, the chances of maintaining visual acuity of CNVMs resulting from OHS is to follow the guidelines from
20/40 or better is 60% compared with 15% if the CNVM is the MPS trial. Juxtafoveal CNVMs should be treated according
inside the FAZ. If the CNVM was within 200 mm of the center to the MPS guidelines as long as a good margin of laser
and the foveal side was treated including 100 mm of the (>100 mm) can be applied on the foveal edge without involving
uninvolved FAZ, those eyes had less risk (5% versus 25%) of the center of the fovea. Juxtafoveal CNVMs that would require
severe vision loss than those left untreated or treated with a laser treatment into the center of the fovea should be con-
wider border on the foveal side.78 sidered for alternative treatments generally considered for
subfoveal CNVM as discussed below. CNVMs on the nasal side
MACULAR PHOTOCOAGULATION STUDY – of the optic nerve rarely need treatment, since they usually
OHS AND PERIPAPILLARY CNVM undergo spontaneous involution. The most common compli-
cation of laser photocoagulation for CNVM due to OHS is
Patients with juxtapapillary membranes could be entered into recurrence, usually attributable to inadequate coverage or
the MPS–OHS study if treatment would allow 1.5 clock hours intensity of the treatment. Other complications are rare
of the optic disc to be left untreated. The CNVM was ineligible (Table 153.4). Because the recurrence rate exceeds 30% CNVMs
if it extended under the center of the FAZ. In the group of treated with laser photocoagulation should be followed with
patients with peripapillary or large (>750 mm nasal extrafoveal frequent examination and daily home use of the Amsler grid.
or juxtafoveal CNVMs, 14% of the treated eyes versus 41% of
the untreated eyes experienced 6 lines or more vision loss in LASER TREATMENT TECHNIQUE
3 years; in those with large nasal CNVM, the numbers were 9%
versus 54%, respectively.79 This branch of the MPS used the
krypton red laser (647 nm). There are several advantages of
GENERAL GUIDELINES
krypton red laser, including less lens scatter, less absorption by There are some general rules that may guide laser
xanthophyll, and less inner retina damage.80 Hemoglobin does photocoagulation of choroidal neovascular membranes.
not absorb red laser at all. There are several disadvantages to The role of anesthesia in laser photocoagulation of CNVMs
krypton red. This laser can crack Bruch’s membrane more is to provide comfort during the procedure and to increase the
easily than can argon green, but the incidence of internal margin of safety when treating near the fovea. Topical
limiting membrane wrinkling is higher with argon. These anesthesia may be adequate for treatment in the cooperative
advantages and disadvantages are created because the red laser patient with an extrafoveal CNVM. For juxtafoveal CNVMs, we
2014 is absorbed by melanin in the RPE and choroid. recommend retrobulbar anesthesia in all but the most
Ocular Histoplasmosis Syndrome

FIGURE 153.12. (a) Schematic representation


of juxtafoveal CNVM inside 200 mm from the
center of the foveal avascular zone. Photograph
(b) and angiograms (c and d) show early
(c) and middle (d) frames from the patient
shown in FIGURE 153.7 after krypton red laser
treatment. Note that choroidal vessels are
visible through the laser scar.
(a) From Poliner LS, Olk RJ: Ocular histoplasmosis
syndrome. Semin Ophthalmol 1987; 2:238.

a b

CHAPTER 153
c d

arm. A fixation light for the patient’s fellow eye helps to


TABLE 153.4. Complications of Laser Treatment
stabilize the treatment eye. The patient should be introduced to
Recurrence of choroidal neovascular membrane the sound of the laser before treatment actually begins.
Treatment should be guided by an early frame of a recent
Retinal pigment epithelial rips
fluorescein angiogram. One should use a computer monitor (for
Acute choroidal hemorrhage digital angiograms), a table-top viewer (for film angiograms)
Nerve fiber layer field defects adjacent to the laser slit lamp.
Various fundus contact lenses are available for macular
Premacular fibroplasia treatment. It must be remembered that some lenses reverse and
invert the surgeon’s view of the fundus (Mainster, Rodenstock).
The fluorescein angiogram must be properly oriented so that
the surgeon does not confuse the anatomy.
cooperative patients. If the patient squeezes the lids, Bell’s If bleeding occurs during the laser treatment, intraocular
phenomenon can move the eye superiorly, thereby risking pressure should be increased with the contact lens and then
photocoagulation of the fovea during treatment of a CNVM laser treatment applied over the bleeding site, preferably with
superior to fixation. Further, juxtafoveal CNVMs should be argon green.
treated with krypton red or diode laser (805 nm), and this Klein and colleagues81 did not avoid retinal vessels in the
frequently causes some discomfort if local anesthesia is not treatment area. However, it is generally recommended that
given. heavy, long-duration burns not be placed directly over retinal
A recent fluorescein angiogram is essential for adequate vessels, especially if the vessel supplies the central fovea.
treatment. It has been shown that CNVMs may grow, especially Sabates and colleagues82 found that hemorrhage obscures the
those closer to the fovea, in the course of several days. The complete extent of the CNVM and absorbs the laser energy,
fluorescein angiogram being used to guide treatment should be thus sparing the CNVM. Intense treatment over areas of
less than 72 h old, especially with juxtafoveal lesions. More subretinal hemorrhage is not indicated, as energy absorption is
latitude may be taken with extrafoveal CNVMs that are greater quite superficial and will cause damage to the retina.
than 200 mm from the center of the FAZ.
The fluorescein angiogram is essential for mapping the FAZ
as well as the CNVM. The use of the aiming beam to map
ARGON GREEN LASER FOR EXTRAFOVEAL
fixation is not a reliable way to define the FAZ. Because of CNVM
eccentric fixation or distortion of the fovea, one may be fooled First, the CNVM is outlined with burns of 100 mm size and
into treating the fovea if the aiming beam is used to determine 0.1 s duration. Gass83 recommends covering any pigment ring
fixation. and choroidal neovascularization as shown by an early frame of
The following procedural details may be helpful. Both the the fluorescein angiogram. On the foveal side, 100–200 mm
surgeon and the patient should be seated comfortably at the spots of 0.2 s duration are delivered in a confluent fashion.
laser. An elbow rest should be used to stabilize the surgeon’s These should be heavy white spots. The remainder of the lesion 2015
RETINA AND VITREOUS

a b c

FIGURE 153.13. (a–c) Schematic representation of the treatment of an extrafoveal CNVM.


(a–c) From Poliner LS, Olk RJ: Ocular histoplasmosis syndrome. Semin Ophthalmol 1987; 2:238.

FIGURE 153.14. (a and b) Schematic


representation of the treatment of a juxtafoveal
CNVM.
(a and b) From Poliner LS, Olk RJ: Ocular
SECTION 10

histoplasmosis syndrome. Semin Ophthalmol 1987;


2:238.

a b

is covered confluently with heavy laser burns of 200–500 mm


spots and 0.5 s duration. The CNVM should be covered 100 mm
INCIDENCE
past the edge of the CNVM. On the foveal side, if the treatment The MPS reviewed the incidence of recurrent CNVMs.85,86 In
will not enter the FAZ, the treatment should extend 100 mm. the OHS-juxtafoveal segment of the study, 31% of the treated
If the edge of the CNVM is close to the edge of the FAZ, eyes had recurrent CNVMs, and 65% of these recurrences were
coverage of the CNVM by a full 100 mm beyond the edge is not amenable to further treatment. The remainder were not
required (Fig. 153.13). retreatable because of subfoveal extension of the CNVM. This
compares with 59% of treated eyes with age-related macular
degeneration and 33% of eyes with idiopathic CNVM.
KRYPTON RED LASER FOR JUXTAFOVEAL Recurrence after surgical removal of CNVM in OHS has been
CNVM reported between 38 to 44%.87,88 Two-thirds of recurrences
The krypton red laser should be used with large spots (at least were subfoveal and were either observed or surgically removed.
200 mm) and long-duration (at least 0.2 s) to decrease the The remaining third was amenable to laser photocoagulation
chance of rupturing Bruch’s membrane. The retina will whiten (i.e., extrafoveal or juxtafoveal) and did much better than those
less with krypton red than with argon green. It is not necessary observed or surgically removed.
to treat blood or blocked fluorescence (Fig. 153.14).
First, the foveal side of the lesion is treated with 200 mm
spots of 0.2 s duration. The remainder of the lesion is treated TIME COURSE
with 200–500 mm confluent burns of 0.5 s duration. Most recurrences were noted within 12 months after treatment,
1. If hyperfluorescence is more than 100 mm from the with the majority occurring within the first 6 months.
center of the FAZ, and if blood or blocked fluorescence
is present, treatment extends 100 mm into the blocked
fluorescence, but the center of the FAZ is not treated. CAUSE
2. If hyperfluorescence is within 200 mm and if no blocked Several factors may contribute to the recurrence of CNVMs
fluorescence is present, laser treatment should cover after treatment: inadequate coverage or intensity of treatment,
the hyperfluorescence, and treatment does not need to the presence of blood or pigment within 200 mm of the center of
extend 100 mm past the edge. the FAZ, and location of the CNVM less than 200 mm from the
center. Further, the growth of an independent CNVM may occur
RECURRENT CNVM IN OHS FOLLOWING during the posttreatment follow-up period. This is believed to be
LASER PHOTOCOAGULATION unrelated to treatment. Several risk factors have been statistically
associated with recurrences, including hypertension, cigarette
smoking, proximity to fovea, young age, and female gender.
DEFINITION In relation to the original CNVM, recurrences can occur on
After laser photocoagulation, interpretation of the post- the margin of the treatment, in the center of the treatment scar,
treatment angiogram can be one of the most challenging facets contiguous to the scar, from a feeder vessel originating from the
in the treatment of macular disease.84 A persistent CNVM original CNVM, or as a completely new lesion greater than
shows hyperfluorescence (representing choroidal neovascular- 250 mm from the margin of the laser scar (Fig. 153.15). Ninety
ization) within 6 weeks of treatment. A recurrent CNVM is percent of marginal recurrences are on the foveal side of the
defined as leakage adjacent to or within the laser scar occurring treatment, indicating that recurrences are largely responsible for
2016 more than 6 weeks after treatment. visual loss from OHS after laser treatment.
Ocular Histoplasmosis Syndrome

better in 31% of the cases at 10.5 months follow-up. Macular


function after removal of CNVM depends on functional
photoreceptors, RPE, and choriocapillaris. Akduman and
associates92 showed that visual improvement correlated with
perfusion of subfoveal choriocapillaris after submacular surgery
in OHS. Seventy-one percent of the eyes with perfused
subfoveal choriocapillaris had visual improvement postop-
eratively versus only 14% of those not perfused. These initially
encouraging case series led to the submacular surgery trial (SST)
for OHS.

THE SUBMACULAR SURGERY TRIALS


(GROUP H)
The SSTs were a group of randomized prospective clinical trials
funded by the National Institutes of Health whose purpose was
to determine if submacular surgery was beneficial for certain
types of CNVM. The SST Group H was designed to compare

CHAPTER 153
surgical removal versus observation of subfoveal CNVM that
were either idiopathic or associated with ocular histo-
plsmosis.102 Eligible patients had new or recurrent subfoveal
CNVM and visual acuity of 20/50 to 20/800 in the study eye.
A successful outcome was defined as a 24 month visual acuity
better or no more than one line (seven letters) worse than at
baseline.102
Of the 225 patients enrolled, 113 were assigned to observ-
ation and 112 to surgery. Forty-six percent of the eyes in the
observation arm and 55% in the surgery arm had a successful
outcome (success ratio 1.18; 95% confidence interval
0.89–1.56). Median visual acuity at the 24 month examination
was 20/250 for eyes in the observation arm and 20/160 for
eyes in the surgery arm. In a subgroup of eyes with initial
visual acuity worse than 20/100, surgery was more successful;
76% of 41 eyes in the surgery arm versus 50% of 40 eyes in the
observed arm at the 24 month examination had a successful
outcome (success ratio 1.53; 95% confidence interval
1.08–2.16)
Complications were more frequently detected in the eyes
undergoing submacular surgery. Four percent of eyes in the
surgery arm experienced rhegmatogenous retinal detachment.
FIGURE 153.15. Schematic examples of locations of recurrent Twenty-four percent of eyes developed postvitrectomy cataract,
CNVMs. all in patient over the age of 50 years. Recurrent CNVM
From Sorenson JA, Yannuzzi LA, Shakin JL: Recurrent subretinal developed in 58% of surgically treated eyes by the 24 month
neovascularization. Published courtesy of Ophthalmology (1985; 92:1059–1074). follow-up exam.
The SST Goup H Study Group concluded that the small
TREATMENT OF SUBFOVEAL CNVM benefit of submacular surgery for CNVM due to OHS was less
than the trial was designed to detect. However, they did
Laser photocoagulation of subfoveal CNVMs has been shown to recommend submacular surgery for the subgroup of eyes with
be of no benefit in a pilot study.54 Therefore, subfoveal laser visual acuity worse than 20/100. Other factors to consider when
treatment of CNVMs is contraindicated. contemplating submacular surgery include the small risk of
retinal detachment, probably cataract among older patients, and
the need for additional treatment if recurrent CNVM were to
MANAGEMENT OF SUBFOVEAL CNVM occur.
Subfoveal CNVM has active leakage under the center of the The SST Group H also examined the health-related quality-
FAZ. Natural history studies suggest that only 14–16% of these of-life outcomes among patients who were randomized to
patients recover visual acuity of 20/40 spontaneously without observation or surgical removal of the CNVM.103 Trained
treatment.17,50,53 Laser photocoagulation is contraindicated. interviewers who were masked to treatment assignment
Fortunately, there has been a rapid advance in recent years of administered three questionnaires by telephone at baseline
new, alternative treatments for subfoveal CNVM. and 6, 12, and 24 months: The National Eye Insitute Visual
Function Questionanaire (NEI-VFQ), the 36- Item Short-Form
Health Survey (SF-36), and the Hospital Anxiety and
SURGICAL REMOVAL OF CNVM Depression Scale (HADS). Vision-targeted quality of life
Between 1992 and 1997, there were numerous reports on improved more after submacular surgery than with observation,
surgical removal of subfoveal CNVM in OHS.62,52,89–101 Various supporting a possible small overall benefit of surgery.
series reported initial visual improvement or stabilization of Submacular surgery techniques have been successfully used
visual acuity in 37–83% of the cases (Fig. 153.16).90,92,93,98,100,101 to treat nonsubfoveal CNVM due to OHS. Atebara and
Thomas and co-workers93 reported visual acuity of 20/40 or colleagues104 studied results of surgical removal of large 2017
RETINA AND VITREOUS

FIGURE 153.16. (a and b) Preoperative color


photograph and fluorescein angiogram of a
patient with subfoveal CNVM secondary to
OHS. Preoperative vision is 20/60. (c and d)
Postoperative color photograph and fluorescein
angiogram of the same patient. Postoperative
vision is 20/40.

a b
SECTION 10

c d

non-age-related macular degeneration peripapillary CNVM, CNVM due to causes other than AMD.105 For the 13 patients
most of which (17/19) were due to OHS. Seventy-nine percent in the study, underlying etiologies included pathologic myopia,
of those were also subfoveal. These authors reported ocular histoplasmosis, angioid streaks, and idiopathic causes.
improvement of preoperative median visual acuity of 20/200 to The authors concluded that PDT with verteporfin was well
20/40 at 7 months follow-up. Submacular surgery has also been tolerated in non-AMD eyes, and treatment could successfully
employed to remove subfoveal hemorrhage that may occur on result in cessation of leakage with one or more sessions.
rare occasions from CNVM in OHS.62 However, a larger, Phase III randomized, prospective, controlled
The era of submacular surgery allowed for histopathologic clinical trial on the use of photodynamic therapy for CNVM due
analysis of the CNVM associated with OHS and increased our to ocular histoplasmosis was never done.
understanding of the pathogenesis of non-AMD choroidal In September 2004, Rosenfeld and colleagues published the
neovascularization. The excised CNVM consists of similar 2 year results of a prospective open-label three-center,
cellular and extracellular components, namely RPE cells, uncontrolled clinical study of PDT with verteporfin in patients
vascular endothelium, erythrocytes, chronic inflammatory with CNVM due to ocular histoplasmosis.106 Twenty-six
cells, cells resembling fibroblasts, collagen fibrils, fibrin, and patients with baseline visual acuity between 20/40 and 20/200
fragments of Bruch’s membrane, as in CNVMs secondary to were studied. At 24 months, 22 patients were available for
other disorders, with the exception of basal laminar drusen examination. Median visual improvement from baseline in
seen only in age-related macular degeneration.95,100 However, visual acuity was 6 letters. Ten patients (45%) gained 7 or more
clinical correlation with the histopathology in the OHS suggests letters of visual acuity while four (18%) patients lost 8 or more
that the new vessels from the choriocapillaris grow through a letters, including two patients (9%) who lost at least 15 letters.
focal abnormality in Bruch’s membrane and RPE into the There was no leakage on the fluorescein angiogram in 85% of
subsensory retinal space, unlike age-related macular degener- the evaluable lesions. No serious ocular events were noted.
ation in which a diffuse abnormality of RPE is present and the The results of the above prospective study corroborated an
CNVM grows under the RPE.52 RPE is engulfed by the CNVM earlier retrospective review published by Busquets et al in June
in age-related macular degeneration, whereas it partially engulfs 2003.107 Thirty-eight patients with CNVM due to OHS that
and delineates the CNVM in OHS. Therefore, a focal rather were treated with PDT using verteporfin were studied. On
than a diffuse abnormality of RPE in OHS may help explain average, OHS patients who received treatment developed 0.88
better surgical results in OHS than other disorders associated line of visual improvement. Visual acuity improved or stayed
with CNVM formation. the same in 69% of eyes, improved by at least two lines in 44%,
and improved by more than four lines in 22%. This
retrospective series had variable follow-up and included some
PHOTODYNAMIC THERAPY FOR SUBFOVEAL eyes (38%) that had undergone prior submacular surgery.
CNVM DUE TO OHS Nevertheless, the authors concluded that PDT with verteporfin
In March 2000, Sickenberg, Schmidt–Erfurth, and Miller may be beneficial in patients with CNV secondary to OHS.
published a nonrandomized, multicenter, open-label, dose- Colleagues at the same institution then reported the use of
escalation Phase I and II clinical trial of single or multiple PDT with verteporfin for juxtafoveal CNVM due to OHS.108
2018 sessions of photodynamic therapy (PDT) with verteporfin for Twenty-three eyes were retrospectively studied. With variable
Ocular Histoplasmosis Syndrome

FIGURE 153.17. (a) A 23-year-old patient with


ocular histoplasmosis presented with a
juxtafoveal CNVM causing metamorphopsia
and visual acuity of 20/25. (b) There is late
hyperfluorescence on the fluorescein
angiogram.

a b

FIGURE 153.18. (a) The patient refused


thermal laser and failed photodynamic therapy
and pegaptanib injection. After one injection of
bevacizumab there was complete resolution of

CHAPTER 153
symptoms, improvement of vision to 20/15, and
inactivity of the CNVM for 6 months. (b) The
fluorescein angiogram shows staining of the
inactive CNVM, but no leakage.

a b

follow-up, vision improved by more than 3 Snellen lines in pressure and cataract. Complications required the removal of
30%, remained within 2 lines of baseline in 52%, and worsened the drug delivery implant in eight eyes.
by 3 lines or more in 18%. Other smaller retrospective case Thus, it appears that intraocular corticosteroids potentially
series on PDT with Verteporfin for subfoveal and juxtafoveal offer stabilization and perhaps some improvement in eyes
CNVM due to OHS suggested similar outcomes.109,110 In with CNVM and OHS, but the complications of cataract and
summary, PDT with verteporfin for CNVM due to OHS is a glaucoma limit the desirability of this treatment.
safe and moderately beneficial treatment option.
While PDT combined with intravitreal triamcinolone
injection has been suggested to be more beneficial than PDT ANTI-VEGF THERAPY AND CNVM DUE TO
alone for CNVM due to AMD, there are no published reports in OHS
the peer-reviewed literature of this treatment modality for At the time of this writing, there are no published reports of
CNVM due to OHS. anti-VEGF therapy for CNVM due to ocular histoplasmosis.
The authors of this chapter have anecdotal experience with
pegaptanib and bevacizumab, and believe that this form of
INTRAOCULAR CORTICOSTEROIDS FOR therapy (along with ranibizumab) will likely prove beneficial
CNVM DUE TO OHS for this disease. The following case history will serve as an
Corticosteroids have some inherent properties which may be example:
considered antiangiogenic. Thus, several investigators have Case #1. A 23-year-old white male presented with blurred
looked at the effect of the intraocular use of corticosteroids for vision and distortion of 1 week duration in the left eye. He was
treating CNVM due to OHS. In October 2003, Rechtman and known to carry the diagnosis of ocular histoplasmosis. In fact,
colleagues retrospectively reported their experience with one year ago, he presented with identical symptoms and was
intravitreal triamcinolone acetonide injections for subfoveal felt to have choroiditis of a juxtafoveal macular histo spot. A
and juxtafoveal CNVM.111 Ten eyes (five subfoveal and five short course of oral steroids showed no benefit. He refused
juxtafoveal) with a mean follow-up of 17 months were treated thermal laser and subsequently underwent photodynamic
with one or more injections. Thirty percent of eyes gained therapy with verteporfin. The symptoms resolved completely
at least 5 letters, 20% lost 5–14 letters, and 50% maintained within 2 weeks.
stable visual acuity. Interestingly, reported side effects included At this recurrence one year later, visual acuity was 20/25.
transient intraocular pressure elevation and mild cataract Again, there was hyperfluorescence of the juxtafoveal macular
development, well known complications of intravitreal histo spot (Fig. 153.17). PDT with verteporfin was performed
corticosteroids. with no improvement. In order to mimic the successful result
Holekamp and colleagues reported on the use of long-term obtained one year earlier, oral corticosteroids were started and
intraocular delivery of fluocinolone acetonide in treating 14 PDT with verteporfin was performed a second time. Because of
eyes with non-AMD CNVM.112 Seven eyes had ocular histo- worsening symptoms and increased fluorecsein angiographic
plasmosis. Average follow-up was 33 months. Ten of 14 eyes leakage, an intravitreal injection of pegaptanib was given. One
demonstrated involution of CNVM or inhibition of recurrent week later there was continued worsening of vision and
CNVM. Ten eyes had stable or improved visual acuity. Median angiographic leakage. Intravitreal bevacizumab was given.
initial visual acuity was 20/64. Median final visual acuity There was immediate and complete resolution of symptoms
was 20/40. However, all eyes developed elevated intraocular with 6 months follow-up (Fig. 153.18). 2019
RETINA AND VITREOUS

FIGURE 153.19. (a) An Amsler grid is used for


diagnosis of CNVMs. This grid can be used at
home by patients at risk for the development
of new vessel membranes. (b) An artist’s
demonstration of metamorphopsia and relative
scotoma as seen by a patient with a new
CNVM.

a b
SECTION 10

CONCLUSION surgery appears to be of benefit in eyes with subfoveal CNVM


in OHS when visual acuity is less than 20/100. PDT with
OHS is a relatively common disease in the endemic areas of the verteporfin offers the possibility of stabilization and occasional
Ohio and Mississippi river valleys. Patients with macular improvement in some eyes, but this has not been shown defini-
chorioretinal scars are at risk for the development of choroidal tively in a large, randomized clinical trial. Intraocular corti-
neovascularization that can threaten central vision. Individuals costeroids have a biologic effect on CNVM in OHS, but the
with a macular ‘histo spots’ or a history of CNVM should self- complications of glaucoma and cataract make this form of
monitor vision regularly with an Amsler grid (Fig. 153.19). therapy less attractive. Finally, anti-VEGF therapy has not yet
Laser photocoagulation is recommended for extrafoveal and been well-studied in this disease, but is a potentially promising
juxtafoveal CNVMs secondary to OHS. Laser photocoagulation treatment for the future.
is not recommended for subfoveal CNVM in OHS. Submacular

REFERENCES
1. Krause AC, Hopkins WG: Ocular 12. Feman SS, Tilford RH: Ocular findings in 23. Klintworth GK, Hollingsworth AS,
manifestations of histoplasmosis. Am J patients with histoplasmosis. JAMA 1985; Lusman PA, Bradford WD: Granulomatous
Ophthalmol 1951; 34:564. 253:2534. choroiditis in a case of disseminated
2. Woods AC, Wahlen HE: The probable role 13. Gutman FA: The natural course of active histoplasmosis: histologic demonstration of
of benign histoplasmosis in the etiology of choroidal lesion in the presumed ocular Histoplasma capsulatum in choroidal
granulomatous uveitis. Am J Ophthalmol histoplasmosis syndrome. Trans Am Acad lesions. Arch Ophthalmol 1973; 90:45.
1960; 49:205. Ophthalmol Soc 1979; 77:515. 24. Craig EL, Suie T: Histoplasma capsulatum
3. Baskin MA, Jampol LM, Huamonte FU, et 14. Lewis ML, Schiffman JC: Long-term follow- in human ocular tissue. Arch Ophthalmol
al: Macular lesions in blacks with the up of the second eye in ocular 1974; 91:285.
presumed ocular histoplasmosis syndrome. histoplasmosis. Int Ophthalmol Clin 1983; 25. Schwarz J, Salfelder K, Viloria JE:
Am J Ophthalmol 1980; 89:77. 23:125. Histoplasma capsulatum in vessels of the
4. Braunstein RA, Rosen DA, Bird AC: Ocular 15. Sawelson H, Goldberg RE, Annesley WH, choroid. Ann Ophthalmol 1977; 9:633.
histoplasmosis syndrome in the United Tomer TL: Presumed ocular histoplasmosis 26. Scholz R, Green WR, Kutys R, et al:
Kingdom. Br J Ophthalmol 1974; 58:893. syndrome. Arch Ophthalmol 1976; 94:221. Histoplasma capsulatum in the eye.
5. Ellis FD, Schlaegel TF: The geographic 16. Gass JDM: Pathogenesis of disciform Ophthalmology 1984; 91:1100.
localization of presumed histoplasmic detachment of the neuroepithelium. Am J 27. Khalil MK: Histopathology of presumed
choroiditis. Am J Ophthalmol 1973; 75:953. Ophthalmol 1967; 63:661. ocular histoplasmosis. Am J Ophthalmol
6. Wheat LJ, French MLV, Kohler RB, et al: The 17. Olk RJ, Burgess DB, McCormick PA: 1982; 94:369.
diagnostic laboratory test for histoplasmosis. Subfoveal and juxtafoveal subretinal 28. Meredith TA, Green WR, Key SN, et al:
Ann Intern Med 1982; 97:680. neovascularization in the presumed ocular Ocular histoplasmosis: clinicopathological
7. Feman SS, Podgorski SF, Penn MK: histoplasmosis syndrome-Natural history. correlation of 3 cases. Surv Ophthalmol
Blindness from presumed ocular Ophthalmology 1984; 91:1592. 1977; 22:189.
histoplasmosis in Tennessee. 18. Macular Photocoagulation Study Group: 29. Pavan PR, Margo CE: Submacular
Ophthalmology 1982; 89:1295. Five-year follow-up of fellow eyes of neovascular membrane and focal
8. Smith RE, Ganley JP: An epidemiologic individuals with ocular histoplasmosis and granulomatous inflammation.
study of presumed ocular histoplasmosis. unilateral extrafoveal or juxtafoveal Ophthalmology 1996; 103:586.
Trans Am Acad Ophthalmol Otolaryngol choroidal neovascularization. Arch 30. Smith RE: Natural history and reactivation
1971; 75:994. Ophthalmol 1996; 114: 677. studies of experimental ocular
9. Smith RE, Ganley JP: Presumed ocular 19. Hoefnagels KLJ, Pijpers PM: Histoplasma histoplasmosis in a primate pool. Trans Am
histoplasmosis. I: Histoplasmin skin test capsulatum in a human eye. Am J Ophthalmol Soc 1982; 80:695.
sensitivity in cases identified during a Ophthalmol 1967; 63:715. 31. Smith RE, Dunn S, Jester JV: Natural history
community survey. Arch Ophthalmol 1972; 20. Roth AM: Histoplasma capsulatum in the of experimental histoplasmic choroiditis in the
87:245. presumed ocular histoplasmosis syndrome. primate. I: clinical features. Invest Ophthalmol
10. Ganley JP, Smith RE, Knox DL, Comstock Am J Ophthalmol 1977; 84:293. Vis Sci 1984; 25:801.
GW: Presumed ocular histoplasmosis. III: 21. Gass JDM, Zimmerman LE: Histopathologic 32. Smith RE, Dunn S, Jester JV: Natural
Epidemiologic characteristic of people with demonstration of Histoplasma capsulatum. history of experimental histoplasmic
peripheral atrophic scars. Arch Ophthalmol Am J Ophthalmol 1978; 85:725. choroiditis in the primate. II:
1973; 89:116. 22. Macher A, Rodriques MM, Kaplan W, et al: histopathologic features. Invest Ophthalmol
11. Davidorf FH, Anderson JD: Ocular lesions Disseminated bilateral chorioretinitis due to Vis Sci 1984; 25:810.
in the Earth Day histoplasmosis epidemic. Histoplasma capsulatum in a patient with 33. Schlaegel TF, Weber JC, Helveston E, et al:
2020 Trans Am Acad Ophthalmol Otolaryngol the acquired immunodeficiency syndrome. Presumed histoplasmic choroiditis. Am J
1974; 78:876. Ophthalmology 1985; 92:1159. Ophthalmol 1967; 63:919.
Ocular Histoplasmosis Syndrome

34. Check IJ, Diddie KR, Jay WM, et al: the feasibility of surgical excision of neovascular maculopathy-Three-year
Lymphocyte stimulation by yeast phase subfoveal neovascular membranes. Am J results. Arch Ophthalmol 1986; 104:594.
Histoplasma capsulatum in presumed Ophthalmol 1994; 118:285. 73. Burgess DB: Ocular histoplasmosis
ocular histoplasmosis. Am J Ophthalmol 53. Kleiner RC, Ratner CM, Enger C, et al: syndrome. Ophthalmology 1986; 83:967.
1979; 87:311. Subfoveal neovascularization in the ocular 74. Brown GC, Brown MM, Sharma S, et al:
35. Ganley JP, Nemo GJ, Comstock GW, Brody histoplasmosis syndrome. Retina 1988; Incremental cost-effectiveness of laser
JA: Lymphocyte transformation in 8:225. therapy for choroidal neovawscularization
presumed ocular histoplasmosis. Arch 54. Fine SL, Wood WJ, Isernhagen RD, et al: associated with histoplasmosis. Retina
Ophthalmol 1981; 99:1424. Laser treatment for subfoveal neovascular 2000; 20:331–337.
36. Brahmi Z, Wheat J, Rubin RH, et al: membranes in ocular histoplasmosis 75. Macular Photocoagulation Study Group:
Humoral and cellular immune response in syndrome: results of a pilot randomized Krypton laser photocoagulation for
ocular histoplasmosis. Ann Ophthalmol clinical trial. Arch Ophthalmol 1993; 111:19. neovascular lesions of ocular
1985; 17:440. 55. Klein ML, Fine SL, Knox DL, Patz A: histoplasmosis. Arch Ophthalmol 1987;
37. Hodgkins PR, Lane AC, Chisholm IH, et al: Follow-up study in eyes with choroidal 105:1499.
Immunophenotyping in presumed ocular neovascularization caused by presumed 76. Macular Photocoagulation Study Group:
histoplasmosis-like retinopathy. Eye 1995; ocular histoplasmosis. Am J Ophthalmol Laser photocoagulation for juxtafoveal
9:56. 1977; 83:830. choroidal neovascularization. Five-year
38. Meredith TA, Smith RE, Braley RE, et al: 56. Gass JDM: Stereoscopic atlas of macular results from randomized clinical trials. Arch
The prevalence of HLA-B7 in presumed disease. 3rd edn. St. Louis: CV Mosby; 1987. Ophthalmol 1994; 112:500.
ocular histoplasmosis in patients with 57. Conway MD, Olk RJ, Burgess D, et al: Oral 77. Fine SL, Olk RJ, Murphy RP, Hillis A:

CHAPTER 153
peripheral atrophic scars. Am J Ophthalmol steroid therapy histoplasmic choroiditis- Current status of clinical trials to evaluate
1978; 86:325. Natural history and visual prognosis. Paper argon (blue-green) laser and krypton (red)
39. Braley RE, Meredith TA, Aaberg TM, et al: presented at the Sixteenth Annual Meeting laser photocoagulation in the treatment of
The prevalence of HLA-B7 in presumed of Macula Society, Feb. 24–27. Naples, FL; ocular histoplasmosis. Int Ophthalmol Clin
ocular histoplasmosis. Am J Ophthalmol 1993:78. 1983; 23:79.
1978; 85:859. 58. Beck RW, Sergott RC, Barr CC, Annesley 78. Macular Photocoagulation Study Group:
40. Godfrey WA, Sabates R, Cross DE: WH: Optic disc edema in the presumed Laser photocoagulation for neovascular
Association of presumed ocular ocular histoplasmosis syndrome. lesions nasal to the fovea. Results from
histoplasmosis with HLA-B7. Am J Ophthalmology 1984; 91:183. clinical trials for lesions secondary to ocular
Ophthalmol 1978; 85:854. 59. Husted RC, Shock JP: Acute presumed histoplasmosis or idiopathic causes. Arch
41. Smith RE, Ganley JP, Knox DL: Presumed histoplasmosis of the optic nerve head. Br Ophthalmol 1995; 113:56.
ocular histoplasmosis. II: patterns of J Ophthalmol 1975; 59:409. 79. Macular Photocoagulation Study Group:
peripheral and peripapillary scarring in 60. Pulido JS, Folberg R, Carter KD, et al: The influence of treatment extent on the
persons with nonmacular disease. Arch Histoplasma capsulatum endophthalmitis visual acuity of eyes treated with krypton
Ophthalmol 1972; 87:251. after cataract extraction. Ophthalmology laser for juxtafoveal choroidal
42. Watzke RC, Claussen RW: The long-term 1990; 97:217. neovascularization. Arch Ophthalmol 1995;
course of multifocal choroiditis (presumed 61. Kranias G: Vitreous hemorrhage secondary 113:190.
ocular histoplasmosis). Am J Ophthalmol to presumed ocular histoplasmosis 80. Mainster MM: Wavelength selection in
1981; 91:750. syndrome. Ann Ophthalmol 1985; 17:295. macular photocoagulation. Ophthalmology
43. Bottoni FG, Deutman AF, Aandekerk AL: 62. Ibanez HE, Williams DF, Thomas MA, et al: 1986; 93:952.
Presumed ocular histoplasmosis syndrome Surgical management of submacular 81. Klein ML, Fine SL, Patz A: Results of argon
and linear streak lesions. Br J Ophthalmol hemorrhage. A series of 47 consecutive photocoagulation in presumed ocular
1989; 73:528. cases. Arch Ophthalmol 1995; 113:62. histoplasmosis. Am J Ophthalmol 1978;
44. Fountain JA, Schlaegel TF: Linear streaks 63. Dreyer RF, Gass JDM: Multifocal choroiditis 86:211.
of the equator in the presumed ocular and panuveitis. Arch Ophthalmol 1984; 82. Sabates NF, Lee KY, Sabates R: Early
histoplasmosis syndrome. Arch Ophthalmol 102:1776. argon laser photocoagulation of presumed
1981; 99:246. 64. Deutsch TA, Tessler HH: Inflammatory histoplasma maculopathy. Am J
45. Schlaegel TF, Cofield DD, Clark G, Weber pseudohistoplasmosis. Ann Ophthalmol Ophthalmol 1977; 84:172.
JC: Photocoagulation and other therapy for 1985; 17:461. 83. Gass JDM: Photocoagulation of macular
histoplasmic choroiditis. Trans Am Acad 65. Schlaegel TF: Ocular histoplasmosis. New lesions. Trans Am Acad Ophthalmol
Ophthalmol Otolaryngol 1968; 72:355. York: Grune & Stratton; 1977. Otolaryngol 1970; 75:580.
46. Miller SA, Stevens TS, de Venecia G: De 66. Schlaegel TF: Corticosteroids in the 84. Sorenson JA, Yannuzzi LA, Shakin JL:
novo lesions in presumed ocular treatment of ocular histoplasmosis. Int Recurrent subretinal neovascularization.
histoplasmosis-like syndrome. Br J Ophthalmol Clin 1983; 23:111. Ophthalmology 1985; 92:1059.
Ophthalmol 1976; 60:700. 67. Makley TA, Long JW, Suie T: Therapy of 85. Macular Photocoagulation Study Group:
47. Gass JDM, Wilkinson CP: Follow-up study chorioretinitis presumed to be caused by Recurrent choroidal neovascularization
of presumed ocular histoplasmosis. Trans histoplasmosis. Int Ophthalmol Clin 1975; after argon laser photocoagulation for
Am Acad Ophthalmol Otolaryngol 1972; 15:181. neovascular maculopathy. Arch Ophthalmol
76:672. 68. Gitter KA, Cohen G: Photocoagulation of 1986; 104:503.
48. Lewis ML, Van Newkirk MR, Gass JDM: active and inactive lesions of presumed 86. Macular Photocoagulation Study Group:
Follow-up study of presumed ocular ocular histoplasmosis. Am J Ophthalmol Persistent and recurrent neovascularization
histoplasmosis syndrome. Ophthalmology 1975; 79:428. after krypton laser photocoagulation for
1980; 87:390. 69. Boucher MC, Dumas J, Labelle P, et al: neovascular lesions of ocular
49. Meredith TA, Aaberg TM: Hemorrhagic Prophylactic argon laser photocoagulation histoplasmosis. Arch Ophthalmol 1989;
peripapillary lesions in presumed ocular of the second eye in presumed ocular 107:344.
histoplasmosis. Am J Ophthalmol 1977; histoplasmosis syndrome. Can J 87. Melberg NS, Thomas MA, Dickinson JD, et
84:160. Ophthalmol 1987; 22:266. al: Managing recurrent neovascularization
50. Cantrill HL, Burgess DB: Peripapillary 70. Fine SL, Patz A, Orth DH, et al: Subretinal after subfoveal surgery in presumed ocular
neovascular membranes in presumed neovascularization developing after histoplasmosis syndrome. Ophthalmology
ocular histoplasmosis. Am J Ophthalmol prophylactic argon laser photocoagulation 1996; 103:1064.
1980; 89:192. of atrophic macular scars. Am J 88. Berger AS, Conway MD, Del Priore LV, et
51. Melberg NS, Thomas MA, and Burgess DB: Ophthalmol 1976; 82:352. al: Submacular surgery for subfoveal
The Surgical removal of subfoveal 71. Macular Photocoagulation Study Group: choroidal neovascular membranes in
neovascular membranes. Ingrowth site as a Argon laser photocoagulation for ocular presumed ocular histoplasmosis. Arch
predictor of visual outcome. Retina 1996; histoplasmosis. Arch Ophthalmol 1983; Ophthalmol 1997; 115:991.
16:190–195. 101:1347. 89. Reddy VM, Zamora RL, Kaplan HJ:
52. Gass JD: Biomicroscopic and 72. Macular Photocoagulation Study Group: Distribution of growth factors in subfoveal
histopathologic considerations regarding Argon laser photocoagulation for neovascular membranes in age-related 2021
RETINA AND VITREOUS

macular degeneration and presumed ocular presumed ocular histoplasmosis. Int choroidal neovascularization in pathologic
histoplasmosis syndrome. Am J Ophthalmol 1993; 17:43. myopia, ocular histoplasmosis syndrome,
Ophthalmol 1995; 120:291. 99. Saxe SJ, Grossniklaus HE, Lopez PF, et al: angioid streaks, and idiopathic causes.
90. Maas S, Deutman AF, Bandhoe F, et al: Ultrastructural features of surgically excised Arch Ophthalmol 2000; 118:327–336.
Surgical removal of subretinal neovascular subretinal neovascular membranes in the 106. Rosenfeld PJ, Saperstein DA, Bressler NM
membranes. Eur J Ophthalmol 1995; 5:48. ocular histoplasmosis syndrome. Arch et al: Photodynamic therapy with
91. Desai VN, Del Priore LV, Kaplan HJ: Ophthalmol 1993; 111:88. verteporfin in ocular histoplasmosis:
Choriocapillaris atrophy after submacular 100. Thomas MA, Grand MG, Williams DF, et al: uncontrolled, open-label 2-year study.
surgery in presumed ocular histoplasmosis Surgical management of subfoveal Ophthalmology 2004; 111:1725–1733.
syndrome. Arch Ophthalmol 1995; 113:408. choroidal neovascularization. 107. Busquets MA, Shah GK, Wickens J et al:
92. Akduman L, Del Priore LV, Desai V, et al: Ophthalmology 1992; 99:952. Ocular photodynamic therapy with
Perfusion of the subfoveal choriocapillaris 101. Berger AS, Kaplan HJ: Clinical experience verteporfin for choroidal neovascularization
affects visual recovery after submacular with the surgical removal of subfoveal secondary to ocular histoplasmosis
surgery in presumed ocular histoplasmosis neovascular membranes. Short-term syndrome. Retina 2003; 23:299–306.
syndrome. Am J Ophthalmol 1997; 123:90. postoperative results. Ophthalmology 1992; 108. Shah GK, Blinder KJ, Hariprasad SM et al:
93. Thomas MA, Dickinson JD, Melberg NS, et 99:969. Photodynamic therapy for juxtafoveal
al: Visual results after surgical removal of 102. Hawkins BS, Bressler NM, Bressler SB, et choroidal neovascularization due to ocular
subfoveal choroidal neovascular membranes. al: Surgical removal versus observation for histoplasmosis syndrome. Retina 2005;
Ophthalmology 1994; 101:1384. subfoveal choroidal neovascularization, 25:26–32.
94. Bynoe LA, Chang TS, Funata M, et al: either associated with the ocular 109. Postelmans L, Pasteels B, Coquelet P et al:
Histopathologic examination of vascular histoplasmosis syndrome or idiopathic. I. Photodynamic therapy for subfoveal classic
SECTION 10

patterns in subfoveal neovascular Ophthalmic findings from a randomized choroidal neovascularization related to
membranes. Ophthalmology 1994; clinical trial: Submacular Surgery Trials punctuate inner choroidopathy (PIC) or
101:1112. (SST) Group H Trial: SST Report No. 9. presumed ocular hiestoplasmosis-like
95. Grossniklaus HE, Hutchinson AK, Capone Arch Ophthlamol 2004; 122:1597–1611. syndrome (POHS-like). Ocul Immunol
A Jr, et al: Clinicopathologic features of 103. Hawkins BS, Miskala PH, Bass EB, et al: Inflamm 2005; 13:361–366.
surgically excised choroidal neovascular Surgical removal versus observation for 110. Lui JC, Boldt CH, Folk JC : Photodynamic
membranes. Ophthalmology 1994; subfoveal choroidal neovascuclarization, therapy of subfoveal and juxtafoveal
101:1099. etiher associated with the ocular choroidal neovascularization in ocular
96. Wind BE, Sobol WM: Surgical management histoplasmosis syndrome or idiopathic. II. histoplasmosis syndrome : a retrospective
of a long-standing subfoveal neovascular Quality-of-life findings from a randomized case series. Retina 2004; 24:863–870.
membrane secondary to ocular clinical trial: SST Group H Trial: SST Report 111. Rechtman E, Allen VD, Danis RP et al:
histoplasmosis. Ophthalmic Surg 1993; No. 10. Arch Ophthlamol 2004; Intravitreal triamcinolone for choroidal
24:36. 122:1616–1628. neovascularization in ocular histoplasmosis
97. Coscas G, Meunier I: Chirurgie des 104. Atebara NH, Thomas MA, Melberg NS, et al: syndrome. Am J Ophthalmol 2003;
membranes neovasculaires sous- Surgical removal of large non-AMD 136:739–741.
retiniennes maculaires. J Fr Ophtalmol peripapillary choroidal neovascular 112. Holekamp NM, Thomas MA, Pearson A:
1993; 16:633. membranes. Ophthalmology 1996; 103:125. The safety profile of long-term, high-dose
98. Peyman GA, Lee KJ, Nelson NC Jr, et al: 105. Sickenberg M, Schmidt-Erfurth U, Miller intraocular corticosteroid delivery. Am J
Bimanual technique of subfoveal JW et al: A preliminary study of Ophthalmol 2005; 139:421–428.
neovascular membrane removal in photodynamic therapy using verteporfin for

2022
CHAPTER

154 Pathologic Myopia


Adrienne W. Scott and Sharon Fekrat

PREVALENCE
Overview
Pathologic, or degenerative myopia, has been defined as Myopia is the most common refractive error and affects ~25% of
refractive error more negative than –6 D, and is a leading cause adults in the United States.6 Various demographic characteris-
of blindness in Asian countries. It is a multifactorial disorder with tics have been shown to be associated with high myopia. Female
genetic, environmental, and socioeconomic etiologies. The gender, younger age, earlier onset of myopia, and family history
pathogenesis of pathologic myopia is not known. Defective
are all risk factors found to be associated with an increased like-
collagen fibril formation results in biomechanically weak sclera
and progressive globe elongation. Numerous complications, such
lihood of progressive myopia. Increased income and educational
as posterior vitreous detachment (PVD) and abnormal level have been associated with high levels of myopia, likely due
vitreoretinal interface, retinal detachment (RD), posterior to their correlation with increased levels of near work.6
staphyloma, macular hole, lacquer cracks (spontaneous breaks in Myopia prevalence varies among racial demographics. One
Bruch’s membrane), subretinal hemorrhage, lead to visual study estimated prevalence of myopia in African-Americans in
disability. The most common visually threatening complication of the United States at 13%.7 Recently, the Los Angeles Latino Eye
pathologic myopia is choroidal neovascularization (CNV). Study estimated an overall prevalence of myopia in adult Latinos
Myopia-related CNV lesions are usually self-limited, and are in one community at 16.8%, with estimated prevalence of high
smaller and less exudative than CNV observed in age-related myopia in this community at 2.4%.8 A very high prevalence of
macular degeneration (AMD). Argon laser has been used to treat
myopia has been reported among Asians. Fredrick reported the
extrafoveal CNV with a high rate of posttreatment recurrence.
Photodynamic therapy (PDT) has been established as a overall prevalence of myopia to be as high as 70–90% in Asian
treatment to preserve visual acuity for subfoveal myopic CNV. populations.9 While 0.2–0.4% of the US population was found to
Antiangiogenic therapy is a promising treatment option. have refractive error greater than ⫺7.9 D,7 the prevalence of
Surgical treatments to displace the myopic CNV have also pathologic myopia in Asian countries has been reported to be as
been utilized in attempts to salvage remaining visual acuity in high as 1%.4
these eyes.
Anticholinergic therapy is being studied as a way to slow
myopic degeneration. As yet, there is no proven method to
PATHOGENESIS
prevent the axial elongation and the myriad resulting posterior
It is well accepted that pathologic myopia is characterized by
segment complications described in pathologic myopia.
progressive scleral thinning with localized ectasia. The exact
pathogenesis of pathologic myopia, however, is not entirely
understood. It is postulated that myopic sclera is biomechan-
INTRODUCTION ically weak and has been demonstrated to alter its composition
and rigidity in response to environmental and visual stimuli.
Myopia, or nearsightedness, is the most common ocular con- Remodeling of extracellular matrix by matrix metalloproteinases
dition. Myopia occurs when an object image is formed anterior and their tissue inhibitors leads to abnormalities in collagen
to the retinal surface, due to an excessively steep corneal curva- fiber bundles and a reduction in the size of individual collagen
ture, an abnormally spherical lens, an increase in lenticular fibers in myopic eyes.10 Myopia has been induced in animal
index of refraction as in nuclear sclerosis, or more commonly, models by defocusing the retinal image, triggering a cascade of
as a result of increased axial length.1 events that leads to axial elongation.11 Cholinergic pathways
Pathologic, myopia occurs when progressive increase in axial have been studied as mediators of scleral elongation associated
length causes excessive thinning of the sclera, retina, retinal with myopia. Other neurotransmitters such as dopamine, vaso-
pigment epithelium (RPE) and choroid, resulting in varying active intestinal peptide, and glucagon have also been implicated
degrees of visual disability. Pathologic myopia has been assigned as possible mediators of scleral growth.12
varying definitions in the ophthalmic literature. Duke-Elder
defined pathologic myopia as myopia accompanied by ASSOCIATIONS
degenerative posterior segment changes.2,3 Pathologic myopia
has also been defined as high myopia accompanied by visual A variety of ocular morbidities such as low-tension glaucoma,
dysfunction.3,4 The American Academy of Ophthalmology Basic amblyopia, strabismus, early cataract formation, image mini-
Clinical Science Course defines pathologic myopia as refractive fication, decreased contrast sensitivity, and visual field defects
error greater than ⫺6 D, with axial length in excess of have all been associated to occur in association with pathologic
26.5 mm.5 myopia. Stickler syndrome, Wagner syndrome, Ehlers–Danlos
2023
RETINA AND VITREOUS

syndrome, and Marfan’s syndrome are hereditary conditions


that have been associated with axial high myopia.

COMPLICATIONS
The fundus changes observed in conjunction with pathologic myo-
pia most notably lead to visual deterioration and blindness in
highly myopic individuals. A trademark of the myopic posterior
segment clinical examination is the tessellated, or tigroid fundus,
in which choroidal vessels are evident below the thinned,
atrophic retina and RPE (Fig. 154.1). This and other abnormal
posterior segment findings in pathologic myopia were described
in a large, retrospective, histopathologic examination of enucleated
eyes with degenerative myopia by Grossniklaus and Green.13
Myopic configuration of the optic nerve head was the most fre-
quently associated finding, present in 37.7% of eyes. Posterior
staphyloma and vitreous degeneration were the next most
frequently observed findings.13
SECTION 10

Pathologic myopic retinopathy is particularly visually


devastating, as it is irreversible, often bilateral, and affects
FIGURE 154.2. RD resulting from a macular hole (black arrow) in a
individuals at younger ages during their productive years.14 The patient with pathologic myopia.
major visually threatening complications of myopic retinopathy Photo courtesy of Brooks W. McCuen, MD.
are discussed below.

POSTERIOR VITREOUS DETACHMENT RETINAL DETACHMENT


Highly myopic eyes are characterized by premature posterior A break in the retina causes liquified vitreous fluid to travel under-
vitreous detachment (PVD). The incidence of PVD in highly neath the retina into the subretinal space, detaching the retina.
myopic patients has been strongly correlated with increased RRD may result from a retinal tear, retinal dialysis, macular
axial length, age, and degree of myopia compared to controls.15 hole, or as a result of acute PVD as described previously. In the
A PVD is often preceded by vitreous syneresis, and the general population, the retinal detachment incidence is less
formation of fluid pockets within the vitreous gel.1 Acute PVD than 1%, RRD occurs more frequently in eyes with increased
may produce photopsia and a symptomatic ‘floater’, in which axial length.3 This increased likelihood of RRD in high myopes
shadows are cast on the retinal surface as a result of vitreous is due to peripheral retinal abnormalities such as lattice
condensation and opacification. Detachment of the posterior degeneration, increased frequency of PVD, and an abnormal
vitreous may also cause acute vitreous hemorrhage if retinal vitreoretinal interface caused by increased axial length.
vessels are avulsed at the time of posterior vitreous separation. RRD as a result of macular hole formation is more likely to
A PVD may cause retinal traction and lead to a retinal break as occur in eyes with pathologic myopia (Fig. 154.2). One study
well as the most common surgical complication of high reported 6.3% of highly myopic eyes to develop an asymptomatic
myopia-rhegmatogenous retinal detachment (RRD). macular hole, confirmed by ocular coherence tomography (OCT).16
High myopia also predisposes eyes to RRD after cataract sur-
gery. Highly myopic eyes undergoing cataract surgery are more
likely to develop a postoperative retinal detachment when
compared to controls.17 Pars plana vitrectomy (PPV) with or
without scleral buckling, with or without gas or silicone oil
tamponade may be used to repair RRD.

OPTIC NERVE
Globe elongation leads to characteristic optic nerve changes. The
tilted optic nerve is often seen in eyes with pathologic myopia.
Thinning of the retina and RPE leads to the peripapillary tem-
poral crescent observed to surround the optic nerve head (Fig.
154.1). A recently described optic nerve finding is peripapillary
detachment. Freund and colleagues reported peripapillary detach-
ment as an elevated, yellow-orange lesion inferior to the optic disk
seen in highly myopic eyes.18 OCT demonstrated a localized reti-
nal pigment epithelial detachment corresponding to the lesion.
Peripapillary detachment was not observed to be visually signifi-
cant. In another study, peripapillary detachment was present in
as many as 4.9% of highly myopic eyes and was associated with
glaucomatous optic nerve defects in 71% of eyes.19
FIGURE 154.1. Typical fundus appearance of pathologic myopia. This
patient has 20/60 vision with –20 D correction. Note tilted optic nerve POSTERIOR STAPHYLOMA
with peripapillary crescent, tigroid fundus appearance, attenuated
vessels, and chorioretinal atrophy. Localized, progressive globe elongation and resulting scleral ecta-
2024 Photo courtesy of Srilaxmi Bearelly, MD. sia leads to the development of a posterior staphyloma, another
Pathologic Myopia

logic myopia may be called a Fuchs spot. Foerster also described


hemorrhagic lesions in myopic eyes which may describe a dif-
ferent stage in myopic CNV formation.26,27 It has been observed
that CNV in persons younger than 50 is seen most often in
conjunction with pathologic myopia.28

NATURAL HISTORY OF MYOPIC CNV


The natural history of myopic CNV is variable. Yoshida and col-
leagues reported a generally poor prognosis for these untreated
eyes with vision decreasing to worse than 20/200 at 10-year
follow-up.29 The same group also reported favorable character-
istics of highly myopic eyes with CNV, which include younger
FIGURE 154.3. OCT in a patient with pathologic myopia. Visual acuity
is 20/200 with a spherical equivalent of –22 D. Note the posterior age at onset, smaller CNV lesion size, and juxtafoveal location.
staphyloma, and the hyperreflectivity of the tissues underlying the Degree of myopia did not appear to influence prognosis in this
atrophic retina. A partial PVD overlies the macula. study.30 Myopic CNV appears to be a self-limited process with low
Photo courtesy of James Kesler, MD, FACS. activity and rapid absorption. It is, however, the development of
chorioretinal atrophy around the myopic CNV that has been

CHAPTER 154
hallmark of degenerative myopia (Fig. 154.3). The prevalence of observed to enlarge, involve the fovea in some cases, and cause
staphyloma formation correlates with axial length. Posterior a further decrease in visual acuity.24,31
staphyloma may cause visual field deficits, and central visual
acuity may be affected if the staphyloma involves the macula. MEDICAL TREATMENTS OF MYOPIC CNV
Increased likelihood of retinal detachment and formation of
macular hole have been associated with posterior staphyloma Treatment modalities aimed toward treating CNV related to
formation in a study by Oie and colleagues.20 age-related macular degeneration (AMD) have been applied to
the treatment of CNV in myopic eyes. CNV does, however,
LACQUER CRACKS differ between these two groups in that most CNV lesions in
association with pathologic myopia are classic, less exudative,
Excessive tissue stress in pathologic myopia may also result in and smaller as compared to CNV lesions in AMD.3 Laser
lacquer crack formation – focal, linear breaks in Bruch’s mem- photocoagulation was the primary available treatment option
brane. Lacquer cracks may cause spontaneous subretinal hemor- for myopia-associated CNV prior to the 1990s.3,19,32–36 Laser
rhage (Fig. 154.4) at the time of formation, and may produce photocoagulation has shown short-term efficacy in limiting the
symptoms of acute photopsia, metamorphopsia, or scotoma.1 A extension of the extrafoveal or juxtafoveal CNV. A long-term
later complication of lacquer crack formation is the development study comparing the natural history of myopic CNV with laser
of secondary CNV. treatment found that the visual acuity was preserved only after
the first 2 years of follow-up.30 At 5-year follow-up, there was no
MYOPIC CNV significant difference in visual acuity between treated and
untreated eyes. The CNV is subject to recur along the border of
Macular CNV occurs in 5–10% of eyes that are highly myopic21,22 the laser scar in treated eyes, with recurrence rates reported as
and is the most common visually threatening complication of high as 31–72%.3,26–30,32–35 In addition to the immediate
pathologic myopia.3,23–25 In its later stages, CNV due to patho- scotoma formation caused by laser treatment, further loss of
central visual acuity may result in laser-treated myopic eyes
with laser scar expansion, a process known as ‘atrophic creep’.3
Photodynamic therapy (PDT) with verteporfin has been a
major treatment option in eyes with myopia-related subfoveal
CNV. PDT involves the administration of an intravenous
photosensitive verteporfin dye which selectively accumulates in
the CNV lesion.3 Diode laser is then used to activate the
accumulated dye and cause occlusion of the choroidal
neovascular vessels. Two-year data from the Verteporfin in
Photodynamic (VIP) Therapy Study Group showed maintained
visual benefit of the PDT-treated group as compared to the
placebo therapy group at 2-year follow-up.37 Other studies have
corroborated the VIP data, observing that PDT can increase the
chance of visual stabilization or improvement compared with
placebo.38 Intravitreal corticosteroids are also being considered
in myopic CNV therapy in conjunction with PDT.39 Though PDT
has been shown to have a treatment benefit in highly myopic CNV
lesions, as with thermal laser treatment, the potential exists for
posttreatment RPE damage, and posttreatment laser expansion of
chorioretinal atrophy in the myopic eye after PDT treatment.3
Angiogenic growth factors such as vascular endothelial growth
factor (VEGF) are implicated in the pathogenesis of ocular neo-
FIGURE 154.4. Subretinal hemorrhage in a myopic eye. This –11.5 D vascularization in the human eye.40,41 As in the case of neovas-
myopic patient experienced a decrease in vision to 20/80 from cular AMD, VEGF inhibition is being studied as a treatment
submacular hemorrhage. modality in myopia-related CNV.42 Clinical trials are currently
Photo courtesy of Srilaxmi Bearelly, MD. underway to evaluate anti-VEGF therapy in pathologic myopia. 2025
RETINA AND VITREOUS

SURGICAL TREATMENTS FOR MYOPIC CNV High myopia is a multifactorial disorder, with environmental,
socioeconomic, and genetic factors playing a role in myopic
Surgical removal of CNV in pathologic myopia has been degeneration. An X-linked form of pathologic myopia has been
performed.43,44 No significant improvement in visual acuity has described,51 and Young and associates have identified a chromo-
been reported, and surgical extraction of myopic CNV has been somal locus for autosomal dominant high myopia.52 Discovery
associated with high recurrence rates and postsurgical RPE of other genes involved in high myopia may make gene therapy
atrophy.3,43–46 a potential area for future treatment.
Another surgical intervention, macular translocation, is a com-
plex surgical procedure first described as a treatment for macular Key Features
degeneration by Machemer and Steinhorst.3,46 Macular trans- • Tigroid, or blond fundus, with choroidal vessels visible
location involves moving the overlying sensory retina from the underneath
subfoveal CNV bed to a new location overlying healthy RPE.3 • Tilted optic nerve with peripapillary atrophy
Proliferative vitreoretinopathy has been a frequent post- • Peripapillary detachment
operative complication.3 Further study is needed to establish the • Chorioretinal atrophy
role of macular translocation in the treatment of myopic CNV. • Posterior vitreous detachment
• Retinal detachment
TREATMENT • Lacquer cracks
• Lattice degeneration (spontaneous breaks in Bruch’s
SECTION 10

The treatments mentioned previously are all dedicated to miti- membrane)


gating the visual disability due to complications of pathologic • Cobblestone degeneration
myopia. There is no known cure for the globe elongation respon- • Fuchs’ spot (RPE hyperplasia in response to CNV)
sible for the complications experienced by those with pathologic • Scleral thinning
myopia. Prevention of myopic degeneration has been attempted • Peripheral retinal holes
with anticholinergic agents, such as atropine47,48 and pirenzip- • Macular holes causing retinal detachment
ine,49 that have been studied as possible therapeutic agents to • Choroidal neovascularization
slow scleral growth in humans.
Scleral reinforcement surgery has been attempted to arrest the
axial globe elongation which occurs in pathologic myopia.1,50 Treatment Options for Myopic CNV
The clinical benefit of these procedures has as yet been • Observation
unproven in a randomized, controlled clinical trial. • Photodynamic therapy with or without intravitreal steroid
Refractive surgery procedures, such as laser in situ kerato- injection (FDA approved for subfoveal CNV)
mileusis (LASIK), photorefractive keratectomy (PRK), and radial • Argon laser (used for extrafoveal CNV, high rate of CNV
keratotomy (RK), have achieved emmetropia in high myopes recurrence and expansion of RPE scarring after treatment)
through alterations of corneal architecture. Phakic refractive • Transpupillary thermotherapy
lenses and cataract surgery have also been utilized to minimize • Angiogenesis inhibition
myopic refractive error. These procedures, however, do not eli- • Surgical extraction of CNV lesion
minate the myriad posterior segment complications that • Macular translocation
threaten the visual acuity of high myopes.

REFERENCES
1. Pruett RC: Pathologic myopia. In: Albert et al: Myopic refractive error in adult Cataract surgery as a risk factor for retinal
and Jakobiec’s principles and practice of Latinos: the Los Angeles Latino Eye Study. detachment in very highly myopic eyes.
ophthalmology. 2nd edn. Philadelphia, PA: IOVS 2006; 47:1845–1852. Ophthalmology 2003; 110:2355–2361.
WB. Saunders Ca 2000; 878–882. 9. Fredrick DR: Myopia. BMJ 2002; 18. Freund KB, Ciardella AP, Yannuzzi LA, et al:
2. Duke-Elder S: Pathologic refractive errors. 324:1195–1199. Peripapillary detachment in pathologic
System of ophthalmology. vol V. 10. Rada JA, Shelton S, Norton TT: The sclera myopia. Arch Ophthalmol 2003;
Ophthalmic optics and refraction. Louis: and myopia. Exp Eye Res 2006; 121:197–204.
Mosby; 1970:297–373. 82:185–200. 19. Shimada N, Ohno-Matsui K, Yoshida T, et al:
3. Tano Y: LIX Edward Jackson Memorial 11. Wallman J: Retinal control of eye growth and Characteristics of peripapillary detachment
Lecture. Pathologic myopia: where are we refraction. Prog Ret Res 1993; 12:133–153. in peripapillary myopia. Arch Ophthalmol
now? Am J Ophthalmol 2002; 12. Crewther DP: The role of photoreceptors in 2006; 124:46–52.
134:645–660. the control of refractive state. Prog Retina 20. Oie Y, Ikuno Y, Fujikado T, Tano Y: Relation of
4. Tokoro T: On the definition of pathologic Eye Res 2000; 19:421–457. posterior staphyloma in highly myopic eyes
myopia in group studies. Acta Ophthalmol 13. Grossniklaus HE, Green WR: Pathologic with macular hole and retinal detachment.
1988; 66:107–108. findings in pathologic myopia. Retina 1992; Jpn J Ophthalmol 2005; 49:530–532.
5. American Academy of Ophthalmology: 12:127–133. 21. Curtin BJ: Basic science and clinical
Pathologic myopia. In: Retina and vitreous, 14. Shih YF, Ho TC, Hsiao CK, et al: Visual management. In: Curtin BJ, ed. The
basic and clinical science course, 2003; outcomes for high myopic patients with or myopias. Philadelphia: Harper & Row;
San Francisco: American Academy of without myopic maculopathy: a 10-year 1985:237–245.
Ophthalmology 2003; 74. follow up study. Br J Ophthalmol 2006; 22. Grossniklaus HE, Green WR: Pathologic
6. Eye Disease Prevalence Research Group: 90:546–550. findings in pathologic myopia. Retina 1992;
The prevalence of refractive errors among 15. Ye J, Zhou C, Du H, et al: Study on the 12:127–133.
adults in the United States, Western posterior vitreous detachment in patients 23. Avila MP, Weiter JJ, Jalkh AE, et al: Natural
Europe, and Australia. Arch Ophthalmol with high myopia. Zhongguo Yi Xue Ke Xue history of choroidal neovascularization in
2004; 122:495–505. Yuan Xue Bao 1999; 21:472–477. degenerative myopia. Ophthalmology 1984;
7. Sperduto RD, Seigel D, Roberts J, 16. Coppe AM, Ripandelli G, Parisi V, et al: 91:1573–1581.
Rowland M: Prevalence of myopia in the Prevalence of asymptomatic macular holes 24. Fried M, Siebert A, Meyer-Schwickerath G:
United States. Arch Ophthalmol 1983; in highly myopic eyes. Ophthalmol 2005; A natural history of Fuchs’ spot. A long
101:405–407. 112:2103–2109. term follow-up study. Doc Ophthalmol
2026 8. Tarczy-Hornoch K, Ying-Lai M, Varma R, 17. Ripandelli G, Scassa C, Parisi V, et al: 1981: 28:215–221.
Pathologic Myopia

25. Hampton GR, Kohen D, Bird AC: Visual 35. Pece A, Brancato R, Avanza P, et al: Laser neovascularization in highly myopic
prognosis of disciform degeneration in photocoagulation of choroidal patients. Br J Ophthalmol 2001;
myopia. Ophthalmology 1983; 90:923–926. neovascularization in pathologic myopia: 85:1041–1043.
26. Foerster R: Ophthalmologische beitrage. long-term results. Int Ophthalmol 1994; 44. Uemura A, Thomas MA: Subretinal surgery
Berlin: Enslin; 1862:55. 18:339–344. for choroidal neovascularization in patients
27. Soubrane G, Coscas G: Choroidal 36. Secretan M, Kuhn D, Soubrane G, et al: with high myopia. Arch Ophthalmol 2000;
neovascular membrane in degenerative Long-term visual outcome of choroidal 118:344–350.
myopia. In: Schachat AP, Murphy RP, neovascularization in pathologic myopia: 45. Haemlin N, Glacet-Bernard A, Brindeau C,
Ryan SJ, eds. Retina. Los Angeles: Mosby; natural history and laser treatment. Eur J et al: Surgical treatment of subfoveal
1994:1149. Opthalmol 1997; 7:307–316. neovascularization in myopia: macular
28. Cohen SY, Laroche A, Leguen Y, et al: 37. Verteporfin in Photodynamic Therapy (VIP) translocation versus surgical removal. Am J
Etiology of choroidal neovascularization in Study Group: Verteporfin therapy of Ophthalmol 2002; 133:530–536.
young patients. Ophthalmology 1996; subfoveal choroidal neovascularization in 46. Machemer R, Steinhorst UH: Retinal
103:1241–1244. pathologic myopia: 2- year results of a separation, retinotomy, and macular
29. Yoshida T, Ohno-Matsui K, Ohtake Y, et al: randomized clinical trial-VIP report no. 3. relocation. II. A surgical approach for
Myopic choroidal neovascularization: a Ophthalmology 2003; 110:667–673. macular degeneration. Ophthalmology
10-year follow-up. Ophthalmology 2003; 38. Schnurrbusch UE, Jochmann C, 1999; 101:1384–1396.
110:1297–1305. Wiedemann P, et al: Quantitative 47. Bedrossian RH: The effect of atropine on
30. Hayashi K, Ohno-Matsui K, Yoshida T, et al: assessment of the long-term effect of myopia. Ophthalmology 86:713–719.
Characteristics of patients with a favorable photodynamic therapy in patients with 48. Shih YF, Hsiao CK, Chen CJ, et al: An

CHAPTER 154
natural course of myopic choroidal pathologic myopia. Graefe’s Arch Clin Exp intervention trial on efficacy of atropine and
neovascularization. Graefe’s Arch Clin Exp Ophthalmol 2005; 243:829–833. multi-focal glasses in controlling myopic
Ophthalmol 2005; 243:13–19. 39. Potter MJ, Szabo SM, Ho T: Combined progression. Acta Ophthalmol Scand 1999;
31. Kojima A, Ohno-Matsui K., Teramukai S, photodynamic therapy and intravitreal 79:233–236.
et al: Factors associated with the triamcinolone for the treatment of 49. Siatkowski RM, Cotter S, Miller JM, et al:
development of chorioretinal atrophy myopic choroidal neovascularization in a US pirenzapine study group. Safety and
around choroidal neovascularization in 13-year-old girl. Graefe’s Arch Clin Exp efficacy of 2% pirenzapine ophthalmic gel
pathologic myopia. Graefe’s Arch Clin Exp Ophthalmol 2005; 21:1–3. in children with myopia: a 1-year,
Ophthalmol 2004; 242:114–119. 40. Adamis AP, Miller JW, Bernal MT, et al: multicenter, double-masked, placebo-
32. Jalkh AE, Weiter JJ, Trempe CL, et al: Increased vascular endothelial growth controlled parallel study. Arch Ophthalmol
Choroidal neovascularization in factor levels in the vitreous of eyes with 2004; 122:1667–1674.
degenerative myopia: role of laser proliferative diabetic retinopathy. Am J 50. Coroneo MT, Beaumont JT, Hollows FC:
photocoagulation. Ophthalmic Surg 1987; Ophthalmol 1994; 118:445–450. Scleral reinforcement in the treatment of
18:721–725. 41. Aiello LP, Avery RL, Arrigg PG, et al: pathologic myopia. Aust N Z J Ophthalmol
33. Soubrane G, Pison J, Bornert P, et al: Vascular endothelial growth factor in ocular 1998; 16:317–320.
Neovaisseaux sous-retiniens de la myopie fluid of patients with diabetic retinopathy 51. Gregg FM, Feinberg EB: X-linked
degerative: resultants de la and other retinal disorders. N Engl J Med pathologic myopia. Ann Ophthalmol 1992;
photocoagulation. Bull Soc Ophthalmol Fr 1994; 331:1480–1487. 24:310–312.
1986; 86:269–272. 42. Nguyen QD, Shah S, Tatlipinar S, et al: 52. Paluru PC, Nallasamy S, Devoto D, et al:
34. Brancato R, Menchini U, Pece A, et al: Dye Bevacizumab suppresses choroidal Identification of a novel locus on 2q for
laser photocoagulation of macular neovascularization caused by pathologic autosomal dominant high-grade myopia.
subretinal neovascularization in myopia. Br J Ophthalmol 2005; IOVS 2005; 46:2300–2307.
pathological myopia. A randomized study 89:1368–1370.
of three different wavelengths. Int 43. Ruiz-Moreno JM, de la Vega C: Surgical
Ophthalmol 1997; 7:339–344. removal of subfoveal choroidal

2027
CHAPTER

155 Idiopathic Macular Hole


Justin L. Gottlieb

Advances in vitreoretinal imaging techniques and success in toid degeneration, coalescence of cystic spaces, and the
surgical repair have stimulated great interest in the pathophysi- development of a full-thickness hole was an important develop-
ology and natural history of macular holes. In particular, optical ment in the understanding of macular hole development, as it
coherence tomography (OCT) provides unique imaging of the emphasized that trauma was not a necessary precedent event.24
vitreomacular interface that has refined the understanding of Current concepts of macular hole formation focus on the role
macular hole formation. In the 25 years since Kelly and of the vitreomacular interface. As early as 1924, Lister stated
Wendel’s landmark publication describing the successful the importance of the vitreous in the pathogenesis of macular
repair of macular holes with vitrectomy surgery,1 the surgical hole,25 noting traction of fibrous bands in the vitreous. How-
technique continues to be refined with improved anatomic and ever, he was puzzled by his own clinical observations of a lack
visual outcomes. of visible tractional vitreous bands in cases of macular holes.26
Several series from the 1960s described signs of vitreomacular
EPIDEMIOLOGY AND RISK FACTORS traction contributing to macular hole formation.27–30
Avila and Jalkh concluded that persistent vitreous-to-macula
Macular holes have been reported in association with many traction was important in the formation of macular holes, as
causes, including trauma,2 laser treatment,3 retinal vascular 53% of their cases of macular holes did not have a posterior
disease,4,5 retinal detachment repair,6,7 lightening,8 and vitreous detachment demonstrable on biomicroscopic exami-
electrocution,9 but the vast majority of cases are age-related and nation.31 Similarly, Akiba et al described the progression of
idiopathic, unrelated to other antecedent events or other ocular premacular hole lesions to fully developed holes without the
disease. The Eye Disease Case-Control Study Group reported occurrence of a posterior vitreous detachment.32 In contrast,
that 72% of idiopathic holes occurred in women and more than McDonnell et al noted a complete vitreous separation in all
50% in patients 65–74 years of age10 The observed increased macular hole cases and in all cases observed to progress from
risk in females is poorly understood and explanations only premacular hole lesions to full-thickness macular hole,33
speculative. Investigating the increased incidence of macular concluding that the act of vitreous separation from the macula
hole in females, the case study report found that estrogen use was critical to the formation of a full-thickness hole. It is
was protective but did not find an association with prior interesting that each group concluded that vitreous traction on
hysterectomy. The observation of macular holes among siblings the macula was important in the pathogenesis of macular holes,
within four different families has also suggested a possible but arriving at this conclusion from seemingly contradicting
genetic component in the formation of macular holes.11 evidence.
The risk of full-thickness macular hole formation in the The current concept of macular hole pathogenesis has grown
fellow eye is estimated to be ~10–15%.12–15 Normal fellow eyes from the understanding that vitreofoveal traction is central to
with posterior vitreous detachment appear to be at a very low the development of and progression to a full-thickness macular
risk of macular hole development.14,16 However, in fellow eyes hole. In 1988, Gass31 and Johnson and Gass35 proposed a new
with persistent vitreofoveal attachments (as evidenced by hypothesis of macular hole formation, including a classification
optical coherence tomography) 11% of fellow eyes developed a scheme for idiopathic holes and precursor lesions. This
full-thickness macular hole over a 2 year period of observation.17 classification scheme has guided the description of macular hole
development and theories of pathogenesis since that time. Gass
PATHOGENESIS proposed that contraction of prefoveolar cortical vitreous
resulted in tangential traction. This traction resulted in a
Theories from the early nineteenth century of the pathogenesis predictable progression through multiple stages of development
of macular hole focused on trauma,18 although contemporary (Fig. 155.1). Stage 1 is the earliest biomicroscopic sign of an
reports find that greater than 80% are idiopathic.19 Early impending macular hole and results from a foveolar or foveal
histological description of full-thickness hole noted cystic detachment. Ophthalmoscopically, it appears as a yellow dot
intraretinal changes.20,21 These changes were usually assumed (stage 1a) or yellow ring (stage 1b), respectively (Fig. 155.2).
to be due to ocular trauma, supporting the prevailing theories Progressive tangential traction may result in a small full-
of the era. thickness break in the neurosensory retina. These holes were
Other early theories of macular hole formation suggested that described as eccentric can-opener shaped tears or small central
macular cysts may form atraumatically and degenerate into defects (stage 2) (Fig. 155.3). These small holes usually prog-
macular holes.22,23 Kuhnt implicated a degenerated fovea as ressed to larger 400-500 mm holes with or without operculum
the cause of macular hole and termed this disorder retinitis (stage 3) (Fig. 155.4) and finally may involve the separation of
atrophicans sive rarificans centralis.22 The implication of cys- the posterior vitreous (stage 4) (Fig. 155.5). 2029
RETINA AND VITREOUS

FIGURE 155.1. Schematic diagram of stages


VC of development of idiopathic macular hole
based on biomicroscopic observations as
proposed by Gass. See text for detail.
A. NORMAL FOVEA Reproduced from Gass JDM: Reappraisal of
biomicroscopic classification of stages of
development of a macular hole. Am J
Ophthalmol 1995; 119:752–759.

B. STAGE 1-A IMPENDING HOLE C. STAGE 1-B IMPENDING HOLE


SECTION 3

D. STAGE 1-B OCCULT HOLE E. STAGE 1-B OCCULT HOLE

F. STAGE 2 HOLE G. STAGE 2 HOLE

H. STAGE 3 HOLE I. STAGE 4 HOLE

FIGURE 155.2. (a) Color fundus photograph of


stage 1b hole. Note the central yellow ring.
(b) Optical coherence tomography of stage 1
hole. Note the foveolar detachment. Visual
acuity was 20/25. This eye later developed a
full-thickness macular hole.

a b

FIGURE 155.3. (a) Color fundus photograph of


stage 2 macular hole. The hole is full-thickness
but a diameter less than 400 mm. (b) Optical
coherence tomograph of stage 2 macular hole.
Note the vitreous traction on the operculum
which is not yet detached from the retina.

a b

Dr Gass later provided a revision and update of the bio- prefoveolar condensed vitreous cortex bridging the hole. The
microscopic classification.36 In this reappraisal, Gass proposed occult hole may then become visible after an early separation or
that the yellow ring of a stage 1b hole is due to centrifugal an eccentric can-open-like tear in the condensed prefoveolar
displacement of retinal receptors after a dehiscence at the vitreous cortex. The prefoveolar cortex may then form a prehole
2030 umbo. The occult hole may be hidden by the semiopaque pseudo-operculum. This scheme helped to unify surgical
Idiopathic Macular Hole

FIGURE 155.4. (a) Color fundus photograph of


a stage 3 hole. This hole is at least 400 mm in
diameter and has an overlying pseudo-
operculum. (b) Optical coherence tomography
of stage 3 hole. In comparison to Figure 155.3b,
the operculum is clearly detached form the
retina surface and attached to the posterior
vitreous.
a b

stages of vitreous separation. They were able to demonstrate


initial stages of vitreous separation in the peripheral portions of
the macula which then spread throughout the macula with the
hyaloid remaining focally adherent to the foveola and the optic
disk. A distinct convexity was observed, suggesting antero-
posterior traction from the vitreous (Figs 155.2b, 155.3b, 155.4b,

CHAPTER 155
and 155.6). Other investigators have confirmed the ability of OCT
to detect the perifoveal vitreous detachment.41,42,43
Johnson and co-workers44 utilized B-scan ultrasonography
and vitreoretinal surgical observations of eyes with stage 1 or
stage 2 macular holes. They demonstrated the presence of a
shallow, localized detachment of perifoveal vitreous typically
extending to the temporal vascular arcades, supporting the
findings of Gaudric. The elegant findings of investigators using
OCT40–42 and B-scan ultrasonography14,44 suggest a modified
FIGURE 155.5. Color fundus photograph of stage 4 hole. This hole theory of macular hole pathogenesis implicating age-related
was present for at least 1 year and the visual acuity had decreased to posterior vitreous detachment beginning in the perifoveal region
20/200. with abnormally strong persistent vitreofoveal adherence. This
theory implicates anteroposterior traction, not the tangential
traction as suggested by Gass.34,36,45 Smiddy and Flynn46
observations of restoration of foveal architecture and sometimes present a unifying theory incorporating observations of early
near normal function, which seemed at odds with the earlier cystic degeneration, attempted wound healing by Müller and
scheme of pathogenesis in which foveal opercula containing glial cells creating tangential traction, and later anteroposterior
photoreceptors were thought to develop (Fig. 155.1). traction as clearly seen in OCT imaging. They suggest that it is
possible that the weakened or dehisced central fovea may be a
MACULAR HOLE IMAGING AND primary event, followed by attempted repair by focal pro-
PATHOGENESIS liferation of Müller and glial cells, and persistent anteropos-
terior traction at the foveola finally resulting in an irreversible
The classification scheme and description of pathogenesis development of full-thickness hole.
provided by Dr Gass was based primarily on biomicroscopic Staging systems utilizing the observations from OCT have
observations. Other diagnostic studies may be used to help been developed. These generally retain the Gass biomicroscopic
establish the diagnosis of macula hole, although the bio- stages 1A/1B through four as a basis.47
microscopic appearance remains the primary means of
diagnosis. Fluorescein angiography may show focal hyper- NATURAL HISTORY
fluorescence in stage 1 and stage 2 holes.34 However, a similar
pattern may be present in lesions mimicking macular holes, There is a wide range of variation in the stated progression of
such as epiretinal membrane with pseudohole. B-scan ultra- pre-hole (stage 1 or macular ‘cyst’) lesions to full-thickness hole
sonography is an excellent means of imaging the posterior formation. The Vitrectomy for Prevention of Macular Hole
vitreous hyaloid and may help determine the presence of Study Group48 reported that 40% of eyes with stage 1 lesions
persistent vitreous attachment to the macula.14,37,38 It is rarely randomized to observation progressed to full-thickness macular
utilized to determine the presence of macular hole in clinical hole over 2 years. This occurred in an average time of 4.1
practice and except for the determination of posterior vitreous months after diagnosis. Other studies are mostly small and
detachment, is not useful for staging macular holes. retrospective. Rates of progression from premacular hole lesions
Ocular coherence tomography (OCT) is a medical imaging to full-thickness holes range from ~10 to 70%.35,36,48,49
technology that has improved the imaging of both the Generally, spontaneous resolution of stage 1 lesions is asso-
neurosensory retina and vitreoretinal interface.39 OCT has ciated with a vitreofoveal separation with foveal reattachment
especially improved visualization of very shallow detachments and a normal biomicroscopic appearance, or may demonstrate
of the posterior hyaloid over the macula. Even with contact lens an inner lamellar hole.36
biomicroscopy this shallow detachment over the parafovea and Visual symptoms in stage 1 or early stage 2 lesions include
macula is most often imperceptible. Information gained from metamorphopsia and reduced visual acuity. The visual acuity of
OCT evaluation of macular hole formation in evolution has eyes with stage 1 macular holes may help to predict the
provided new understandings of the sequence of events leading progression to full-thickness hole formation. The visual acuity
from vitreofoveal traction to full-thickness macular hole of most stage 1 lesions ranges from 20/25 to 20/80. In one
formation. Gaudric et al40 utilized OCT to study eyes with retrospective series, if the visual acuity was 20/40 or better, 23%
macular holes as well at the fellow eyes to detect the initial progressed to a macular hole within 2 years, whereas, if the 2031
SECTION 3 RETINA AND VITREOUS

FIGURE 155.6. Schematic diagram of macular hole formation based on


optical coherence tomography observations.
Reproduced from Gaudric A, Haouchine B, Massin P: Macular hole
formation. New data provided by optical coherent tomography. Arch
Ophthalmol 1999; 117:744–751.

visual acuity was 20/50 or worse, 89% progressed to macular is an epiretinal membrane and pseudohole.53 Also simulating a
hole.19 The Vitrectomy for Prevention of Macular Hole Study full-thickness macular hole may be a lamellar macular hole54 or
Group similarly found that stage 1 macular holes with best- chronic cystic macular edema.
corrected visual acuity of 20/50 to 20/80 had a 66% rate of Premacular hole lesions are often misdiagnosed. Gass
progression, while eyes with best-corrected visual acuity of and Joondeph55 reported that only one of 18 patients referred
20/25 to 20/40 had a 30% risk of progression to full-thickness with a diagnosis of stage 1 lesion actually had such a
macular hole.48 lesion. The other misdiagnosed lesions included aborted
The majority of stage 2 holes continue to progress to stage 3 macular hole, stage 2 holes, stage 3 holes, and unrelated
or stage 4 holes.50,51 Stage 3 macular holes have central visual lesions. Other lesions simulating a ‘pre-hole’ lesion may
acuity loss to the 20/80 to 20/200 level.51 There is a strong co- include central serous chorioretinopathy with a central yellow
relation between macular hole diameter and visual acuity.51,52 spot,55 the early yellow lesion of solar retinopathy,55 a central
Similarly, the hole diameter (and visual acuity) is closely druse in age-related macular degeneration,55 or a pseudo-
correlated to duration of symptoms. operculum.56

DIFFERENTIAL DIAGNOSIS MANAGEMENT OF MACULAR HOLES


Because of the preservation of peripheral vision and the relative
Key Features rarity of bilateral central visual loss, some have questioned the
• Full Thickness Hole necessity for surgical intervention for repair of macular holes.57
• Epiretinal membrane and pseudohole However, the improvements in surgical techniques and results
• Lamellar hole and a concomitant decrease in overall surgical complications of
• Cystic macular edema vitreoretinal surgery have led most vitreoretinal surgeons to
• Central serous chorioretinopathy with central yellow spot readily recommend surgical repair for affected eyes. Most retina
• Solar retinopathy specialists now recommend surgery for symptomatic stage 2, 3,
• Macular druse or 4 macular holes of limited duration with visual acuity of at
• Pseudo-operculum least 20/50 or worse.
Appreciating the role of vitreomacular traction in macular
hole formation led to the first attempts at macular hole repair
Several lesions may simulate a full-thickness macular hole or a through pars plana vitrectomy surgery. In 1991, Kelly and
precursor macular hole lesion (stage 1a or 1b). The most Wendel reported on pars plana vitrectomy, removal of the
2032 common lesion to be mistaken for a full-thickness macular hole posterior cortical vitreous, and strict facedown positioning after
Idiopathic Macular Hole

gas–fluid exchange for repair of macular hole.1 The original visual acuity improvement by two or more lines occurred in
technique of Kelly and Wendel remains the basis of surgical 42% (73% of anatomically successful eyes). In a second report,
technique today. Wendel and co-authors reported improved success with 73%
The critical component of the surgical repair macular holes is anatomic success and 55% improvement by two or more lines.60
the induction of a posterior vitreous detachment (Fig. 155.7). The authors noted greater success in holes present for less than
This is confirmed intraoperatively by the identification of a 6 months. The greater success with holes present for shorter
Weiss ring and the visualization of the more opaque posterior duration has been confirmed by a number of other authors.61,62
cortical vitreous face of the vitreous. The posterior cortical Most reports of visual and anatomic results are small and
vitreous may be identified with the use of a soft silicone uncontrolled. The definitions of anatomic and visual success
extrusion cannula. With gentle aspiration, the silicone tip will differ among the many reports. Two multicenter, randomized
engage the posterior cortical vitreous and induce a bend in the clinical trials provide information regarding success rates. Kim
tip similar to a ‘fish-strike’ on a fishing line.1 Many surgeons and colleagues reported the results of surgery for stage 2
simply use the vitrectomy cutting instrument on aspiration macular holes.63 There was no statistically significant difference
mode to engage and elevate the posterior cortical vitreous, prior in ETDRS chart visual acuity between the surgical and observed
to carefully completing the vitrectomy to the vitreous base. groups. However, the Bailey–Love word-reading test did show a
Epiretinal membranes are then generally excised using a barbed significant difference between the two groups (20/78 vs 20/135,
microvitreoretinal blade, diamond-dusted silicone scraper P = 0.006).
and/or fine intraocular forceps. Indirect ophthalmoscopy of the In a second report from the Vitrectomy for Treatment of

CHAPTER 155
retina is performed to carefully inspect for iatrogenic retinal Macular Hole Study Group, Freeman and colleagues reported
tears, most often associated with the process of vitreous the results of surgery versus observation for stages 3 and 4
separation.58 A fluid–air exchange is performed, followed by a macular holes.64 They report a 69% closure rate, as compared
second fluid aspiration at least 10 min after the initial fluid–air with a 2% spontaneous closure rate in the observation group.
exchange. The second aspiration allows for removal of the The surgically repaired eyes had a statistically significant better
preretinal fluid accumulating from the residual vitreous skirt, visual acuity at 6 months, both by ETDRS chart testing (20/115
ciliary body, and anterior-segment structures.59 The air is then versus. 20/166, P ≤ 0.004) and by Bailey–Love word-reading test
exchanged with a longer acting, nonexpansile concentration of (20/155 vs 20/166, P=<0.01).
gas. Face-down positioning is required postoperatively to A 2 year randomized clinical trial from Moorefields Eye
provide maximum tamponade of the gas upon the surface of the Hospital compared surgery to observation for full-thickness
macula and to stimulate closure of the macular hole. macular holes of less than or equal to 9 months duration.65
In the initial report of successful macular hole surgery by They additionally compared whether the addition of autologous
Kelly and Wendel, 58% of the holes were successfully closed and serum improved results in the surgical arm. They reported an

a b c

FIGURE 155.7. Basic steps in vitrectomy


surgery for macular hole. (a) Engagement of
posterior cortical vitreous (CV) with a soft-
tipped extrusion cannula during active
aspiration – ‘the fish-strike sign’. (b) Stripping
of the CV. (c) Completion of the posterior
vitrectomy with the vitreous cutter. (d) Fluid–air
exchange. (e) Completion of fluid–air exchange
with removal of fluid over the optic nerve.
(a–e) Glaser BM, Michels RG, Kupprman BD,
et al: Transforming growth factor-b2 for the
treatment of full-thickness macular holes. A
prospective randomized study. Ophthalmology
1992; 99:1162–1173. Courtesy of
Ophthalmology

d e
2033
RETINA AND VITREOUS

11.5% spontaneous closure rate with little or no visual acuity Closure rates as high as 88–100% have been reported.71–75 How-
change of those eyes at 24 months. The surgical group had an ever, no prospective, randomized trials have been performed to
overall closure rate of 80.6%, with 45% of eyes achieving a compare standard procedures with and without ILM peeling. A
visual acuity of 20/40 or greater. Autologous serum did not retrospective comparative study of hole closure rate and
affect the anatomic or visual results. postoperative visual acuities did not demonstrate a statistical
difference in outcomes.76
CONTROVERSIES IN MACULAR HOLE The identification and peeling of the ILM is technically
REPAIR SURGERY challenging. Adjuvants to improve visualization of the ILM
include indocyanine green dye (ICG), trypan blue dye, and
triamcinolone acetonide.
SURGICAL ADJUVANTS Indocyanine green dye is instilled onto the surface of the
Most surgeons do not currently use adjuvant agents in the retina in an air-or fluid-filled eye. The ICG selectively stains the
surgical repair of macular holes. Such adjuvants are generally ILM, and the ILM may be more easily identified and elevated
applied to the surface of the macular hole at the conclusion of with the use of diamond-dusted silicone cannula or with fine
vitrectomy surgery and fluid–air exchange and still require face- end-grasping forceps (Fig. 155.9). Retinal and retinal pigment
down positioning after an initial period of prone positioning. epithelial toxicity77–79 have been reported and questions have
Glaser et al reported initial success with intravitreal trans- arisen about potentially diminished visual acuity in eyes
forming growth factor beta (TGF-b) derived from bovine bone exposed to intravitreal ICG dye.80 Others report excellent
SECTION 3

with higher anatomic closure rate compared with placebo.66 results with the use of adjunctive-ICG without evidence of
However TGF-b derived from recombinant DNA did not result toxicity.81 No randomized, prospective trials have been
in similar success.67 performed.
Other blood-derived adjuvants, such as autologous Trypan blue stains both the ILM and epiretinal membranes.
serum,65,68 autologous concentrated platelets,69 and whole The staining of the ILM is less intense than the staining
blood70 have also been utilized. Randomized clinical trials using achieved with ICG.82,83 There have been no reports of toxicity
autologous serum have not demonstrated improved closure associated with trypan blue.
rates or visual results.65,68 Similarly, platelet concentrates and Triamcinolone acetonide can be injected intraoperatively
whole blood have not proven effective in clinical trials. onto the surface of the macula. While it does not stain the
internal membrane, it adheres to the surface and may facilitate
identification of the ILM.84,85
INTERNAL LIMITING MEMBRANE PEEL
Currently, the greatest controversy in surgical technique for
macular hole is the role of internal limiting membrane (ILM) DURATION OF FACE-DOWN POSITIONING
peeling. The necessity, preferred technique, and potential Currently most vitreoretinal surgeons recommend strict face-
complications, including toxicity of adjuvant agents are far from down positioning for at least 1 week postoperatively. Comp-
definitively established. liance with positioning seems to increase the success rates of
Techniques for removal of the ILM involve establishing an hole closure.60 Studies of decreased duration suggest that
elevated edge of the ILM and then peeling the ILM from around successful closure of holes can occur with 0–4 days of
the macular hole. Establishing an initial edge may be accom- positioning.86,87 The shorter duration of positioning depends on
plished with the use of a barbed microvitreoretinal blade71 or
with the use of fine intraocular end-grasping forceps to ‘pinch’
and elevate the ILM.72 The ILM peel is most often performed
in a circular motion around the hole (‘maculorhexis’) (Fig.
155.8).73
Removal of the ILM appears to increase the rate of macular
hole closure and perhaps improve the final visual acuity.

FIGURE 155.8. Peeling the internal limiting membrane with forceps in


a circular fashion around the macular hole – ‘macculorhexis’. FIGURE 155.9. Intraoperative photograph of peeling of the internal
Reproduced from Sjaarda RN Thompson JT: Macular hole. In: Ryan SJ, limiting membrane after staining with indocyanine green dye.
2034 Wilkinson CP, eds. Retina. 4th edn. Elsevier Moseby; 2006. Photograph courtesy of Mathew W MacCumber, MD, PhD.
Idiopathic Macular Hole

a very complete vitreous removal in a pseudophakic eye and ment (1–5% in most reports, but up to 14% reported).74–76,87,93,95
successful ILM peeling.87 Postoperative retinal detachments are most typically inferiorly
The use of silicone oil for postoperative tamponade has been located and associated with small retinal tears at the vitreous
advocated for patients unable to position. The visual acuity and base. Visual-field loss (up to 20%),96,97 may be due to excessive
closure rates are better among eyes undergoing surgery with gas infusion pressures and retinal dehydration during fluid–air
tamponade compared with silicone oil.88,89 exchange.98,99 Elevated intraocular pressures are common after
vitrectomy surgery with intraocular gas infusions100 and angle
closure in the unoperated eye during face-down positioning has
COMPLICATIONS OF MACULAR HOLE been reported.101
SURGERY
SUMMARY
Key Features
• Iatrogenic retinal tears Full-thickness macular holes are most often unilateral, age-
• Retinal detachment related, and idiopathic. Visual symptoms include central
• Cataract metamorphopsia and reduced visual acuity. Improved under-
• Visual-field defect standing of the pathophysiology of macular holes has occurred
• Ocular hypertension through careful biomicroscopic observations and through the
improved visualization of the vitreoretinal interface provided by

CHAPTER 155
• Endophthalmitis
• Angle-closure glaucoma OCT. Vitreoretinal surgical techniques continue to evolve, and
there remain questions as to the best surgical approach to
macular holes. However, surgery most often results in suc-
The most common complication of macular hole surgery is cessful closure of the macular hole and improvement of visual
cataract formation.90,91,92 Phakic patients undergoing macular function. Vision-targeted health status questionnaires demon-
hole surgery should be advised that cataract extraction surgery strate that successful macular hole closure is associated with
will likely be required within 1–2 years after macular hole significant improvement in patients’ vision-related quality of
surgery. Other surgical complications include intraoperative life, even when there is only modest improvement in visual
iatrogenic retinal tears (3–17%),74,93,94 postoperative retinal detach- acuity.102

REFERENCES
1. Kelly NE, Wendel RT: Vitreous surgery for Graefes Arch Clin Exp Ophthalmol 1996; 26. Aaberg TM: Macular holes: A review. Surv
idiopathic macular holes: results of a pilot 234:241–245. Ophthalmol 1970; 15:139–162.
trial. Arch Ophthalmol 1991; 109:654–659. 14. Fisher YL, Slakter JS, Yannuzzi LA, Guyer 27. Reese AB, Jones IS, Cooper WC: Macular
2. Knapp H: Ueber isolerte zerreissungen der DR: A prospective natural history study and changes secondary to vitreous traction. Am
aderhaut in folge von traumen auf dem kinetic ultrasound evaluation of idiopathic J Ophthalmol 1967; 64:544–549.
augapfel. Arch Augenklinik 1869; 1:6–29. macular holes. Ophthalmology 28. Aaberg TM, Blair CJ, Gass JDM:
3. Vendantham V: Optical coherence 1994;101:5–11. Macular holes. Am J Ophthalmol 1970;
tomography findings in macular hole due to 15. Guyer DR, de Bustros S, Diener–West M, 69:555–562.
argon laser burn. Arch Ophthalmol 2006; Fine SL: Observations on patients with 29. Yoshioka H: Clinical studies on macular
124(2):287–288. idiopathic macular holes and cysts. Arch holes. III. On the pathogenesis of the senile
4. Cohen SM, Gass JDM: Macular hole Ophthalmol 1992; 110:1264–1268. macular hole. Acta Soc Ophthalmol Jpn
formation following severe hypertensive 16. Akiba J, Quiroz MA, Trempe L: Role of 1968; 72:575–584.
retinopathy. Arch Ophthalmol 1994; posterior vitreous detachment in idiopathic 30. Jaffe, NS: Vitreous traction at the posterior
112:878–879. macular holes. Ophthalmology 1990; pole of the fundus due to alterations in the
5. Amemiya T, Yoshida H: Macular hole in 97:1610–1613. vitreous posterior. Tran Am Acad
diabetic maculopathy. Ophthalmologica 17. Niwa H, Tersasaki H, Ito Y, Mikake Y: Ophthalmol Otolaryngol 1967; 71:642–652.
1978; 177:188–191. Macular hole development in fellow eyes 31. Avila MP, Jalkh AE, Murakami K, et al:
6. Brown GC: Macular hole following with unilateral macular hole. Am J Biomicroscopic study of the vitreous in
rhegmatogenous retinal detachment repair. Ophthalmol 2005; 140:370–375. macular breaks. Ophthalmol 1983;
Arch Ophthalmol 1988; 106:765–766. 18. Kipp CJ: Macular holes: a clinical 90:1277–1283.
7. Runge PE, Wyhinny GJ: Macular hole contribution. Tran Am Ophthalmol Soc 32. Akiba J, Quiroz MA, Trempe CL: Role of the
secondary to pneumatic retinopexy: case 1908; 11:518–528. posterior vitreous detachment in idiopathic
report. Arch Ophthalmol 1988; 106:586–587. 19. McDonnell PJ, Fine SL, Hillis AI: Clinical macular holes. Ophthalmology 1990;
8. Campo RV, Lewis RS: Lightening-induced features of idiopathic macular cysts and 97:1610–1613.
macular hole. Am J Ophthlamol 1984; holes. Am J Ophthalmol 1982; 93:777–786. 33. McDonnell PJ, Fine SL, Hillis AI: Clinical
97:792–794. 20. Fuchs E: Zur veranderung der macula lutea features of idiopathic macular cysts and
9. Chavanne H: Pseudo–trou maculaire par nach contusion. Ztschr Augenheilk 1901; holes. Am J Ophthalmol 1982; 93:777–786.
electrocution. Bull Soc Ophthalmol Fr 1958; 6:181–186. 34. Gass JDM: Idiopathic senile macular hole.
271–275. 21. Coats G: The pathology of macular holes. Its early stages and pathogenesis. Arch
10. Risk factors for idiopathic macular holes. Roy London Hosp Report 1907; 17:69–96. Ophthalmol 1988; 106:629–639.
The Eye Disease Case–Control Study 22. Kuhnt H: Ueber eine eigenthumliche 35. Johnson RN, Gass JDM: Idiopathic
Group. Am J Ophthalmol 1994; Veranderung der Netzhaut as maculam macular holes. Observations, stages of
118:754–761. (retinitis atrophicans sive rareficans formation, and implications for surgical
11. Lalin SC, Chang S, Flynn H, et al: Familial centralis). Z Augneheilk 1900; 3:105–112. intervention. Ophthalmology 1988;
idiopathic macular holes. Am J Ophthalmol 23. Tower P: Observations on hole in the 95:917–924.
2004; 138:608–611. macula. Ophthalmologica 1954; 36. Gass JDM: Reappraisal of biomicroscopic
12. Ezra E, Wells JA, Gray RH, et al: Incidence 40(suppl):1–60. classification of stages of development of a
of idiopathic full-thickness macular holes in 24. Aaberg TM, Blair CJ, Gass JDM: Macular macular hole. Am J Ophthalmol 1995;
fellow eyes. A 5-year prospective natural holes. Amer J Ophthalmol 1970; 119:752–759.
history study. Ophthalmology 1998; 69:640–650. 37. Dagel PU, Smiddy WE, Byrnes SF et al:
105:353–359. 25. Lister W: Holes in the retina and their Macular hole syndromes: echographic
13. Lewis ML, Cohen SM, Smiddy WE, Gass clinical significance. Br J Ophthalmol 1924; findings with clinical correlation.
JD: Bilaterality of idiopathic macular holes. 8:1–20. Ophthalmology 1994; 101:815–821. 2035
RETINA AND VITREOUS

38. Glacet–Bernard A, Zourdani A, Perrenoud F, caused by a pseudo–operculum after peeling. Ophthalmology 2000;
et al: Stage 3 macular hole: role of optical vitreofoveal separation. Retina 1992; 107:1939–1949.
coherence tomography and B-scan 12:242–244. 72. Smiddy WE, Feuer W, Cordahi G: Internal
ultrasonography. Am J Ophthalmol 2005; 57. Fine SL: Vitreous surgery for macular hole limiting membrane peeling in macular hole
139:814–819. in perspective. Is there an indication? surgery. Ophthalmology 2001;
39. Hee MR, Puliafito CA, Wong C et al: (Editorial). Arch Ophthalmol 1991; 108:1471–1478.
Optical coherence tomography of macular 109:635–636. 73. Mester V, Kuhn F: Internal limiting
holes. Ophthalmology 1995; 102:748–756. 58. Sjaarda RN, Glaser BM, Thompson JT, membrane removal in the management of
40. Gaudric A, Haouchine B, Massin P, et al: et al: Distribution of iatrogenic retinal full-thickness macular holes. Am J
Macular hole formation. New data provided breaks in macular hole surgery. Ophthalmol 2000; 129:769–777.
by optical coherent tomography. Arch Ophthalmology 1995; 102:1387–1392. 74. Park DW, Sipperly JO, Sneed SR, et al:
Ophthalmol 1999; 117:744–751. 59. Rubin JS, Thompson JT, Sjaarda RN, et al: Macular hole surgery with internal-limiting
41. Chauhan DS, Antcliff RJ, Rai PA, et al: Efficacy of fluid–air exchange during pars membrane peeling and intravitreous air.
Papillofoveal traction in macular hole plana vitrectomy. Retina 1995; 15:291–294. Ophthalmology 1999; 106:1392–1397.
formation: the role of optical coherence 60. Wendel RT, Patel AC, Kelly NA et al: 75. Olsen TW, Sternberg P Jr, Capone A Jr,
tomography. Arch Ophthalmol 2000; Vitreous surgery for macular holes. et al: Macular hole surgery using thrombin-
118:32–38. Ophthalmology 1993; 100:1671–1676. activated fibrinogen and selective removal
42. Mori K, Abe T, Yoneya S: Dome shaped 61. Thompson JT, Sjaarda RN, Lansing MB: of the internal limiting membrane. Retina
detachment pf premacular vitreous cortex The results of vitreous surgery for chronic 1998; 18:322–329.
in macular hole development. Ophthalmic macular holes. Retina 1997; 17:493–501. 76. Margherio RR, Margherio AR, Williams GA,
Surg Lasers 2000; 31:203–209. 62. Willis AW, Garcia–Cosio JF: Macular hole et al: Effect of perifoveal tissue dissection
SECTION 3

43. Chan A, Duker JS, Schuman JS, Fujimoto surgery. Comparison of longstanding in the management of acute idiopathic full-
JG: Stage 0 macular holes. Observations versus recent macular holes. thickness macular holes. Arch Ophthalmol
by optical coherence tomography. Ophthalmology 1996; 103:1811–1114. 2000; 118:495–498.
Ophthalmology 2004; 111:2027–2032. 63. Kim JW, Freeman WR, Azen SP et al: 77. Sippy BD, Englebrecht NE, Hubbard GB,
44. Johnson MW, Van Newkirk MR, Meyer KA: Prospective randomized trial of vitrectomy et al: Indocyanine green effect on cultured
Perifoveal vitreous detachment is the or observation for stage 2 macular holes. human retinal pigment epithelial cells :
primary pathogenic event in idiopathic Vitrectomy for Macular Hole Study Group. implications for macular hole surgery. Am J
macular hole formation. Arch Ophthalmol Am J Ophthalmol 1996; 121:605–614. Ophthalmol 2001; 132:433–435.
2001; 119:215–222. 64. Freeman WR, Azen SP, Kim JW, et al: 78. Gandorfer A, Haritoglou C, Kampic A:
45. Gass JDM: Müller cell cone, an overlooked Vitrectomy for treatment of full- thickness Retinal damage from indocyanine green in
part of the anatomy of the fovea centralis: stage 3 or 4 macular holes. Results of a experimental macular surgery. Invest
hypotheses concerning its role in the multicenter randomized clinical trial. The Ophthalmol Vis Sci 2003; 44:316–323.
pathogenesis of macular hole and Vitrectomy for Treatment of Macular Hole 79. Haritoglou C, Gandorfer A, Gass CA, et al:
foveomacular retinoschisis. Arch Study Group. Arch Ophthalmol 1997; Indocyanine green-assisted peeling of the
Ophthalmol 1999; 117:821–823. 115:11–21. internal limiting membrane in macular hole
46. Smiddy WE, Flynn HW Jr: Pathogenesis of 65. Ezra E, Gregor ZJ for the Moorefields surgery affects visual outcome: a
macular holes and therapeutic implications. Macular Hole Surgery Group: Surgery for clinicopathologic correlation. Am J
Am J Ophthalmol 2004; 137:525–537. idiopathic full-thickness macular hole. Two- Ophthalmol 2002; 134:836–841.
47. Altaweel M, Ip M: Macular hole: improved year results of a randomized clinical trial 80. Gass CA, Haritoglou C, Schaumberger M,
understanding of pathogenesis, staging, comparing natural history, vitrectomy, and Kampic A: Functional outcomes of macular
and management based on optical vitrectomy plus autologous serum: hole surgery with and without indocyanine
coherence tomography. Sem Ophthalmol Moorefields Macular Hole Study Group green-assisted peeling of the internal
2003; 18:58–66. Report No. 1. Arch Ophthalmol 2004; limiting membranes. Graefes Arch Clin Exp
48.De Bustros S: Vitrectomy for prevention of 122:224–236. Ophthalmol 2003: 241:716–720.
macular holes. Results of a randomized 66. Smiddy WE, Glaser BM, Thompson JT, 81. Da Mata AP, Burk SE, Foster RE, et al:
multicenter clinical trial. Vitrectomy for et al: Transforming growth factor-beta 2 Long-term follow-up of indocyanine green
prevention of macular hole study group. significantly enhances the ability to flatten assisted peeling of the retinal internal
Ophthalmology 1994; 101:105–1059. the rim of subretinal fluid surrounding limiting membrane during vitrectomy
49. Guyer DR, de Bustros S, Diener–West M, macular holes: preliminary results of a surgery for idiopathic macular hole repair.
Fine SL: The natural history of idiopathic multicenter prospective randomized study. Ophthalmology 2004; 111:2246–2253.
holes and cysts. Arch Ophthalmol 1992; Retina 1993; 13:296–301. 82. Li K, Wong D, Hiscott P, et al: Trypan blue
110:1264–1268. 67. Thompson JT, Smiddy WS, William GA, staining of internal limiting membrane and
50. Hickichi T, Toshida A, Akiba J, et al: et al: Comparison of recombinant epiretinal membrane during vitrectomy:
Prognosis of stage 2 macular holes. Am J transforming growth factor beta-2 and visual results and histopathological findings.
Ophthalmol 1995; 119:571–575. placebo as an adjuvant for macular hole Br J Ophthalmol 2003; 87:216–219.
51. Kim JW, Freeman WR, Azen SP, et al: surgery. Ophthalmology 1998; 83. Azad RV, Pal N, Vashisht N, et al: Efficacy
Prospective randomized trial of vitrectomy 105:700–706. of 0.15% trypan blue for staining and
or observation for stage 2 macular holes. 68. Banker AS, Freeman WR, Azen SP, et al: removal of the internal limiting membrane,
Am J Ophthalmol 1996; 121:605–614. A multicentered clinical study of serum as epiretinal membranes, and the posterior
52. Sjaarda RN, Frank DA, Glaser BM, et al: adjuvant therapy for surgical treatment of hyaloid during pars plana vitrectomy.
Assessment of vision in idiopathic macular macular holes. Vitrectomy for Macular Hole Retina 2005; 25:676.
hole with macular microperimetry using the Study Group. Arch Ophthalmol 1999; 84. Horio N, Horiguchi M, Yamamoto N:
scanning laser ophthalmoscope. 117:1499–1502. Triamcinolone-assisted internal limiting
Ophthalmology 1993; 100:1513–1518. 69. Paques M, Chastang C, Mathis A, et al: membrane peeling during idiopathic
53. Allen AW, Gass JDM: Contraction of a Effect of autologous platelet concentrate in macular hole surgery. Arch Ophthalmol
perifoveal epiretinal membrane simulating a surgery for idiopathic macular hole: results 2005; 123:96–99.
macular hole. Am J Ophthalmol 1976; of a multicenter, double-masked, 85. Shah GV, Rosenblatt BJ, Blinder KJ, et al:
82:684–691. randomized trial. Platelets in macular hole Triamcinolone-assisted internal membrane
54. Smiddy WE, Gass JDM: Masquerades of surgery group. Ophthalmology peeling. Retina 2005; 25:972–975.
macular holes. Ophthalmic Surg 1995; 1999;106:932–938. 86. Krohn J: Duration of face-down positioning
26:16–24. 70. Hoerauf H, Kluter H, Joachimmeyer E, after macular hole surgery; a comparison
55.Gass JDM, Joondeph BC: Observations et al: Results of vitrectomy and the no- between 1 week and 3 days. Acta
concerning patients with suspected touch-technique using autologous Ophthalmol Scan 2005; 83:289–292.
impending macular holes. Am J adjuvants in macular hole treatment. Int 87. Tornambe PE, Poliner L, Grote K: Macular
Ophthalmol 1990; 109:638–646. Ophthalmol 2001; 24: 151–159. hole surgery without face-down
56. Gass JDM, VanNewkirk M: Xanthic 71. Brooks HL Jr: Macular hole surgery with positioning: a pilot study. Retina 1997;
2036 scotoma and yellow foveolar shadow and without internal limiting membrane 17:179–185.
Idiopathic Macular Hole

88. Lai JC, Stinnett SS, McCuen BW: after vitrectomy for macular holes and 98. Welch JC: Dehydration injury as a possible
Comparison of silicone oil versus gas epiretinal membranes. Am J Ophthalmol cause of visual defect after pars plana
tamponade in the treatment of idiopathic 2004; 137:250–257. vitrectomy for macular hole. Am J
full-thickness macular holes. 93. Banker AS, Freeman WR, Kim JW, et al: Ophthalmol 1997; 124:698–699.
Ophthalmology 2003; 110:1170–1174. Vision-threatening complications of surgery 99. Hirata A, Yonemura N, Hasamura T, et al:
89. Couvillion SS, Smiddy WE, Flynn HW Jr, for full-thickness macular holes. Vitrectomy Effect of infusion pressure on visual field
et al: Outcomes of surgery for idiopathic for macular hole study group. defects after macular hole surgery. Am J
macular hole: a case-control study Ophthalmology 1997; 104:1442–1452. Ophthalmol 2000; 130:611–616.
comparing silicone oil with gas tamponade. 94. Sjaarda RN, Glaser BM, Thompson JT, 100. Anderson NG, Fineman MS, Brown GC:
Ophthalmic Surg Lasers Imaging 2005; et al: Distribution of iatrogenic retinal Incidence of intraocular pressure spike and
36:365–371. breaks in macular hole surgery. other adverse events after vitreoretinal
90. Leonard RE II, Smiddy WE, Flynn HW Jr, Ophthalmology 1995; 102:1387–1392. surgery. Ophthalmology 2006; 113:42–47.
Feuer W: Long–term visual outcomes in 95. Park SS, Marcus DM, Duker JS, et al: 101. Bansal A, Salmon JF, Malhotra R, et al:
patients with successful macular hole Posterior segment complications after Delayed acute angle closure after macular
surgery. Ophthalmology 1997; vitrectomy for macular hole. hole surgery. Eye 2003; 17:779–781.
104:1648–1652. Ophthalmology 1995; 102:775–781. 102. Tranos PG, Ghazi-Nouri SMS, Rubin GS, et
91. Cheng L, Azen SP, El-Bradley MH, et al: 96. Pendergast SD, McCuen BW II: Visual field al: Visual function and subjective
Duration of vitrectomy and post-operative loss after macular hole surgery. perception of visual ability after macular
cataract in the vitrectomy for macular hole Ophthalmology 1996; 103:1069–1077. hole surgery. Am J Ophthalmol 2004;
study. Am J Ophthalmol 2001; 97. Boldt HC, Munden PM, Folk JC, Mehaffey 138:995–1002.

CHAPTER 155
132:881–887. MG: Visual field defects after macular hole
92. Thompson JT: The role of patient age and surgery. Am J Ophthalmol 1996;
intraocular gas use in cataract progression 122:371–381.

2037
CHAPTER

156 Choroidal and Retinal Folds


Thomas R. Friberg and Jenny Y. Yu

INTRODUCTION
Folds of the choroid and retina are typically a sign of an ocular
disease or disorder. Choroidal folds develop secondary to
biomechanical stresses, for instance, from an extraocular mass
in the orbit pressing upon the globe, from thickening of the
choroid from hypotony, choroidal effusion, or inflammation, or
from stresses secondary to a growing choroidal tumor. Retinal
folds may be seen over tight choroidal folds but are more
typically a result of proliferation along the superficial retinal
layers, such as seen in macular pucker, proliferative retinopathy,
or in scarring from choroidal neovascularization (CNV). Both
choroidal and retinal folds can be seen upon biomicroscopy but
fluorescein angiography outlines choroidal folds most drama-
tically. Retinal folds, on the other hand, are well demonstrated
by optical coherence tomography (OCT) or on high-power
biomicroscopy. The clinical presence of choroidal or retinal
folds needs to be explained, as the underlying disease which
created them may need to be addressed. Both types of folds can
be symptomatic with retinal folds creating metamorphopsia.
This chapter details the biomechanical principles which create
chorioretinal folds so they can be better understood.
Many disorders are associated with the formation of folds
in the choroid and retina. For descriptive purposes, folds pri-
marily involving the choroid, with the overlying retina only
secondarily affected, are classified as choroidal folds, whereas
folds occurring exclusively within the layers of the sensory
retina are termed retinal folds (Fig. 156.1). Choroidal folds are
often a sign of orbital or ocular disease, but they may develop
after surgery on the eye or orbit or may occur idiopathically.1–6
Retinal folds found without the presence of choroidal folds
develop most commonly in macular pucker, but they can also be
present in association with uveitis, in response to certain
medications,7 and as a result of proliferative vitreoretinopathy.

CHOROIDAL FOLDS DESCRIPTION,


PATHOGENESIS, AND SYMPTOMS
Choroidal folds are undulations of the choroid, Bruch’s
membrane, and pigment epithelium, with the overlying retina
wrinkled to a lesser extent.1 They develop secondary to
mechanical stresses produced within these tissues. Because the FIGURE 156.1. (Top) Retinal folds involve the superficial layers of the
retina and are usually secondary to tangential forces present on the
choroid consists of a plexus of randomly oriented blood vessels,
retinal surface. The underlying RPE (cuboidal cells) and choroid are
an interstitium of connective tissue, and multiple fluid-filled not characteristically folded. (Bottom) Choroidal folds are
spaces, it has intrinsic elasticity and sponginess. In addition, characterized by undulations of the choroid, Bruch’s membrane, and
Bruch’s membrane and the overlying retinal pigment epithe- overlying retinal pigment epithelial layers. The sensory retina is folded
lium (RPE) are firmly bonded to the inner surface of choroid at in the deeper layers, but these pleats do not extend completely to the
the choriocapillaris. This architecture is roughly analogous to superficial layers. Hence, retinal vessels are typically not included in
that of a sponge onto which a thin elastic sheet has been fused. the folding process.
2039
RETINA AND VITREOUS

On funduscopic examination, choroidal folds appear as alter-


nating dark and light streaks. Often, their appearance is subtle,
especially if the fundus is lightly pigmented and the folds have
not undergone any chronic changes. Choroidal folds are best
viewed clinically by indirect ophthalmoscopy, as the field of
view is usually large enough to allow observation of the entire
fold pattern.
Histopathologic sections through choroidal folds demon-
strate the reason for their striated appearance. Typically, Bruch’s
membrane and the RPE are tightly folded within the troughs of
the folds (as viewed from the vitreous), whereas at the crests,
the change in curvature is more gradual.4 Hence, the melano-
FIGURE 156.2. As an analogy, the formation of choroidal folds may cytes within the choroid and the RPE are compressed into a
be compared with folds developing along the surface of a smaller-than-normal region in the troughs, whereas over the
compressed sponge onto which an elastic membrane has been glued. crest of the folds, these cells are stretched apart. This change in
The sponge represents the choroid, whereas the membrane pigment density across the folds creates the alternating dark
represents Bruch’s membrane and the RPE. and light striae seen clinically.
SECTION 10

When such a sponge is compressed, folds form along its surface FLUORESCEIN ANGIOGRAPHY AND
(Fig. 156.2). Analogously, if compressive forces of sufficient CHOROIDAL FOLDS
magnitude are induced in the choroid, it will be forced into Sodium fluorescein leaks quickly out of the fenestrations of the
pleats or folds, as will Bruch’s membrane, the pigment choroidal vessels and stains the choroid. However, because the
epithelium, and the sensory retina. Hence, choroidal folds are a RPE effectively filters out most of this potential background
manifestation of biomechanical stresses present within the choroidal fluorescence, the choroid appears gray rather than
choroid, rather than being a sign of a particular disease.2 white on angiography. In the troughs of the choroidal folds in
Symptoms of choroidal folds depend on the degree of folding which the RPE cells are compressed, the RPE becomes a more
of the overlying retina, particularly at the fovea, and the amount efficient filter. Conversely, when the choroid and RPE are
of induced refractive error associated with the development of stretched, the concentration of melanin granules per unit area
the choroidal folds. Retinal folds running through the fovea in is reduced. The net effect is that choroidal folds appear
association with choroidal folds may cause metamorphopsia dramatically as alternating dark and light striae on fluorescein
and a reduction of visual acuity that cannot be eliminated by angiography (Fig. 156.3).9
corrective lenses. Since choroidal folds are commonly found
when the choroid is thickened or the globe has been flattened
posteriorly, axial hyperopia is typical. If the globe is deformed OCT AND CHOROIDAL FOLDS
more anteriorly from an intraorbital tumor, induced astigmatic OCT is a relatively new imaging tool for the posterior segment
errors along with the hyperopia are common.8 Incidental equa- of the eye.10 Similar to ultrasound B-mode imaging, a cross-
torial folds created by encircling procedures to repair retinal sectional image is generated by measuring the backscattered or
detachments do not cause distortion or refractive errors them- backreflected light from the tissue within the eye. Based on
selves, but the scleral buckle in this situation produces relative the optical properties of the underlying tissue, the layers of the
myopia. retina and choroid are distinguished. The choroid appears

a b

FIGURE 156.3. (a) Fundus appearance of choroidal folds caused by an extraconal tumor located within the superior temporal quadrant of the
orbit. (b) On fluorescein angiography, the folds are much more dramatic, appearing as alternating light and dark streaks. Note that the retinal
vessels are not distorted by the underlying choroidal folds.
2040
Choroidal and Retinal Folds

FIGURE 156.4. (a) Fundus photo of the right


eye in a woman with hyperopia and idiopathic
choroidal folds. (b) Cross-sectional image with
false color through the posterior pole of the
same eye. The troughs and crests of the highly
reflective RPE layer indicate choroid folds in the
macula.
Courtesy of T Friberg, MD; UPMC Eye Center.

CHAPTER 156
a

darker in comparison to the highly reflective RPE above. The folds develop parallel to the boundaries of the tumor (Fig. 156.5).
cross-section image provided by OCT can easily detect the Similarly, choroidal folds may develop at the edge of large
undulating appearance of the RPE (Fig. 156.4). The elastic choroidal detachments. The folds in these cases are not exten-
properties of the choroid are different from those of the sensory sive because only local compressive stresses are produced. Folds
retina. Hence the retina can, to some degree, internally adjust typically do not form over the tumor surface, at which point the
(creep) along the underlying choroidal folds. This is evident on choroid is stretched rather than compressed. Occasionally,
the OCT when the inner surface of the retina is only mildly however, folds develop over regions of localized choroidal
undulating as opposed to the folds at the level of the chorio- hemorrhage.12
capillaris, Bruchs membrane, and RPE. Used in conjunction
with fluorescein angiography, OCT can differentiate retinal
folds from choroidal folds in various retinal pathology involving ORBITAL TUMORS
the macula and periphery.11 Intraconal Orbital Tumors
Tumors located within the muscle cone, such as cavernous
CHOROIDAL FOLDS AND SPECIFIC hemangiomas, metastatic neoplasms, and optic nerve meningio-
DISORDERS mas, can press on the globe posteriorly, producing exophthalmos,
flattening of the globe, and shifting of the refractive error toward
hyperopia.8 In addition, such a tumor often displaces the optic
CHOROIDAL TUMORS AND CHOROIDAL nerve to one side, inducing stresses within the globe at the disk
DETACHMENTS (Fig. 156.5a). The choroid on one side of the optic nerve is com-
When a choroidal tumor such as a melanoma, metastatic pressed, whereas, on the opposite side, the choroid is stretched,
carcinoma, or choroidal hemangioma grows, it compresses the often resulting in a parabolic fold pattern with the nerve head
adjacent choroid and may force it into folds. In this instance, located among the folds (Fig. 156.6).8
2041
RETINA AND VITREOUS

FIGURE 156.5. (a) An expanding choroidal


tumor compresses the choroid along its
boundaries, forming folds parallel to the tumor’s
perimeter. (b) Folds seldom form over the tumor
surface because the choroid is stretched
uniformly over the mass, as seen in this vertical
cross-section.

a b
SECTION 10

FIGURE 156.6. (a) Intraconal orbital tumors


typically displace the optic nerve, compressing
the choroid in the direction of the nerve head
displacement, and placing the choroid under
tension on the opposite side. The result is a
choroidal fold pattern, the convex side of which
is directed away from the nerve head (arrow).
(b) Extraconal tumors buckle the wall of the
globe equatorially, producing a curvilinear fold
pattern with its convex side directed toward the
optic nerve.
(a and b) From Friberg TR: The etiology of choroidal
folds. Graefes Arch Clin Exp Ophthalmol 1989;
227:459–464.
a b

Extraconal Orbital Tumors DISORDERS OF THE OPTIC NERVE HEAD


An extraconal tumor may press directly on the extraocular As the optic nerve head swells, it expands centrifugally, com-
muscles and Tenon’s capsule, as well as on the sclera. The pressing the peripapillary choroid and often creating choroidal
forces generated tend to buckle the wall of the globe (see Fig. folds concentric to the disk margins (Fig. 156.9). Thus, con-
156.6b), creating choroidal folds. Because of anterior segment centric folds are a sign of papilledema,13 disk edema, optic nerve
distortion, astigmatic refractive errors are commonly induced. head drusen, or a tumor within the nerve head. In these cases,
With respect to the fold pattern, the convex side usually points the abnormality of the optic nerve is usually quite obvious,
toward the posterior pole and optic nerve, but the nerve head is whereas the folds themselves are subtle.
usually located outside the region of folds (Fig. 156.7 and
156.8).8 Typical extraconal tumors associated with choroidal
folds include mucoceles, dermoids, tumors of the lacrimal SCLERAL BUCKLING PROCEDURES
gland, and orbital meningiomas. Encircling scleral buckling procedures are commonly associated
with choroidal striae (Fig. 156.10a,b). In this case, the folds are
Orbital Tumors and the Pattern of Folds confined to the vicinity of the buckle and are oriented perpen-
Although historically the choroidal fold pattern was said to have dicular to the plane of the encircling element. The encirclement
no relevance in localizing a tumor within the orbit,1 in fact the reduces the surface area available to the choroid, compressing
pattern of folds often reflects the location of the orbital patho- the tissue together within the plane of the buckling element. In
logic condition. The tumor is often located along the axis of addition, radial buckling elements can also produce folds by
symmetry drawn through the pattern of choroidal folds.8 The compression (Fig. 156.10c,d). Folds associated with retinal surgery
location as indicated by the folds is only approximate, however, are typically prominent during the immediate postoperative
and must be corroborated by more sophisticated evaluations, period and fade over time. Furthermore, choroidal folds caused
such as computed tomography or B-scan ultrasonography. by buckling are smoothed out if the intraocular pressure rises.
2042
Choroidal and Retinal Folds

CHAPTER 156
c

b d

FIGURE 156.7. (a and b) An intraconal tumor (in this case, an orbital hemangioma) has displaced the left globe and optic nerve superiorly
(arrow), as seen in computed tomograms. (c) The associated choroidal folds are primarily located superior to the optic nerve, whereas some
folds radiate from the disk and extend into the macula. (d) A B-scan ultrasonogram demonstrates characteristic flattening of the posterior sclera.
(a–d) From Friberg TR, Grove AS: Choroidal folds and refractive errors associated with orbital tumors. An analysis. Arch Ophthalmol 1983; 101:598–603. Copyright
1983, American Medical Association.

CHORIORETINAL SCARS AND CNV hyperopia is secondary to the development of choroidal


Dense, fibrotic chorioretinal scars from trauma or intense thickening and fold formation, rather than being the cause of
choroidal inflammation contract as the scar tissue matures. the folds.
Forces from this contraction pull on the surrounding choroid
and may produce folds directed radially toward the center of the
scar (Fig. 156.11). Scarring associated with CNV can also HYPOTONY
produce radially oriented folds.14 Hypotony or low intraocular pressure results in radial expan-
sion of the choroid toward the vitreous cavity. Hypotony may
result from an obvious cause such as uveitis, trauma, or filtra-
FOLDS ASSOCIATED WITH CHOROIDAL tion surgery, particularly when 5-fluorouracil or mitomycin C
THICKENING are used.15,16 Choroidal folds developing shortly after anterior
The choroid may thicken as a result of hypotony, inflammation, segment surgery suggest a wound leak or the presence of a
the uveal effusion syndrome, or venostasis such as that found cyclodialysis cleft.
after a central retinal vein occlusion. As its thickness increases,
the surface area available to the choriocapillaris, Bruch’s
membrane, and the RPE is reduced because the choroid must INFLAMMATION
expand inward toward the center of the eye, being confined Inflammatory diseases of the orbit and sclera such as Graves’
by the sclera. Ultimately, the choroid may be forced into exophthalmopathy, orbital pseudotumors, and orbital myositis
redundant folds (Fig. 156.12). Additionally, because the internal can flatten the sclera and cause scleral and choroidal thick-
diameter of the globe has been decreased, the refractive error ening. Choroidal compression and choroidal fold formation
shifts toward hyperopia. It is important to remember that this may be the result. In diseases such as inflammatory bowel
2043
RETINA AND VITREOUS

a
SECTION 10

FIGURE 156.8. (a and b) Choroidal folds caused by an extraconal tumor (in this case, a meningioma) that has displaced the left globe inferiorly
and temporally as seen on computed tomography. (c) A curvilinear fold pattern typically associated with an extraconal tumor is seen on the
fluorescein angiogram, with the convex side of the pattern directed toward the optic nerve head. (d) A B-scan ultrasonogram shows equatorial
flattening of the sclera by the meningioma.
(a–d) From Friberg TR, Grove AS: Choroidal folds and refractive errors associated with orbital tumors. An analysis. Arch Ophthalmol 1983; 101:598–603. Copyright
1983, American Medical Association.

disease, the choroid itself may show signs of inflammation


and may develop folds from uveitis.17 Sometimes, serous
retinal detachments develop in association with the folds, pre-
senting a confusing picture. Successful treatment of the
inflammatory component will lead to resolution of the folds in
most cases.

UVEAL EFFUSION
Eyes with idiopathic choroidal folds and concomitant serous
detachments may appear similar to eyes with the uveal effusion
syndrome. Typically, this syndrome is characterized by poor
vision, a thickened or detached choroid and ciliary body, and an
extensive serous retinal detachment that dominates the clinical
appearance.18 In some cases, however, the choroid may be
thickened uniformly without a significant serous detachment,
and vision may be maintained. In these cases, choroidal folds
may be the first sign of an abnormality. Decompression of the
FIGURE 156.9. Swelling of the optic nerve from papilledema or tumor vortex veins and excision of scleral flaps has, in some severe
compresses the adjacent choroid, producing choroidal folds cases, led to resolution of the choroidal detachments and folds
concentric to the disk. with improvement of visual acuity.18,19
2044
Choroidal and Retinal Folds

CHAPTER 156
a b

c d

FIGURE 156.10. (a) Choroidal folds secondary to an encircling scleral buckle. (b) The choroid is compressed in the x direction along the plane
of the buckle and stretched in the y direction, producing folds oriented perpendicular to the induced compressive stresses. (c) A radially placed
scleral buckle element produces choroidal folds by compressing the adjacent choroid. (d) The abrupt change in scleral contour (arrow), as seen
on cross-section, creates these compressive forces.
(a and b) From Friberg TR: The etiology of choroidal folds. Graefes Arch Clin Exp Ophthalmol 1989; 227:459–464.

IDIOPATHIC FOLDS space seen between the optic nerve and its meninges.20 Occa-
Choroidal folds may develop in one or both eyes of a patient sionally, serous retinal detachments develop (Fig. 156.13).
with no apparent ocular or orbital disorder. These patients are Patients usually retain good vision, and in some individuals the
usually male and often present with acquired hyperopia of 3 D folds disappear spontaneously.17 The cause of these alterations
or less. Computed tomographic findings in these eyes may is unclear. Possibly, drainage through the choroidal venous
show thickening of the sclera, flattening of the posterior pole, plexus and into the vortex veins is compromised from
enlargement of the optic nerve image, and the presence of a structural alterations within the scleral wall. In other cases,
2045
RETINA AND VITREOUS

posterior scleritis or other local inflammation may have initially


produced the folds, which then persist long after the inflam-
mation resolves. Alternatively, idiopathic choroidal folds and
acquired hyperopia have been described in intracranial
hypertension.14

IMPLICATIONS OF CHOROIDAL FOLDS


Choroidal folds are a sign of an ocular disorder or disease.
Hence, the major consideration when presented with a patient
with choroidal folds is to determine the cause of the folds. Of
particular importance is an investigation to rule out the possi-
bility of an orbital or choroidal tumor or intracranial hyper-
tension.21 B-scan ultrasonography and computed tomography
are usually indicated, particularly in patients with unexplained
unilateral folds.
If choroidal folds remain for months, the RPE in the troughs
may undergo hypertrophy and hyperplasia from being mech-
SECTION 10

anically compacted. Even if the folds resolve, this linear


pigmentation may persist. RPE atrophy and apparent fractures
FIGURE 156.11. A dense chorioretinal scar draws the surrounding
of Bruch’s membrane may also be seen along the folds, especially
choroid inward toward its center as the scar tissue matures and
contracts. Radially oriented folds are produced.

a b

FIGURE 156.12. (a) Fluorescein angiogram of choroidal folds associated with hypotony and secondary choroidal thickening. (b) In these cases,
the choroidal fold pattern is typically random, as the folds develop secondary to rather uniform swelling of the choroid.

a b c

FIGURE 156.13. (a) Fluorescein angiogram of the right eye in a woman with mild hyperopia and acquired chorioretinal folds. (b) There is leakage
of dye from the choriocapillaris, which pools within the fovea in the late-phase films. A serous retinal detachment is present overlying the area of
2046 leakage. (c) Left eye of the same patient. Fluorescein angiography shows randomly oriented folds and retinal pigment epithelial window defects
in the peripapillary region, at the fovea, and temporal to the macula.
Choroidal and Retinal Folds

CHAPTER 156
c

FIGURE 156.14. (a–c) Retinal pigment epithelial clumping and atrophy (arrows) can occur along choroidal folds, making the folds appear similar
to atrophic angioid streaks, both on clinical examination and particularly on fluorescein angiography.
(a–c) From Friberg TR, Grove AS: Choroidal folds and refractive errors associated with orbital tumors. An analysis. Arch Ophthalmol 1983; 101:598–603. Copyright
1983, American Medical Association.

in older patients (Fig. 156.14). Such changes also occur along


angioid streaks, which are sometimes confused with choroidal
folds.8 As with angioid streaks, CNV can develop along
choroidal folds (Fig. 156.15).22 If CNV develops, treatment
should be initiated using strategies commonly used for neo-
vascular age-related macular degeneration. Macular folds of the
choroid should therefore be eliminated, if possible, to help avoid
permanent visual loss.

Key Features
• Undulations of the choroid, Bruch’s membrane, and pigment
epithelium as a result of compressive stresses developing
along within the plane of the choroid. Dramatic patterns of
hypo- and hyperfluorescent streaks are often seen upon
fluorescein angiography.
• Can be associated with decreased vision and the development
of CNV.

RETINAL FOLDS
Retinal folds are commonly associated with epiretinal membrane
formation. Such membranes exert stress on the underlying
FIGURE 156.15. CNV may develop along choroidal folds, as
demonstrated by this trapezoidal choroidal neovascular net found
retina and promote wrinkle formation, leading to macular
along a fold running through the macula. In this case, the choroidal pucker and symptoms of metamorphopsia. These cellophane-
folds developed secondary to an orbital tumor. like membranes can best be seen on careful biomicroscopy of
From Friberg TR, Grove AS: Subretinal neovascularization and choroidal folds. the fundus. Alternatively, in certain cases of rhegmatogenous 2047
Ann Ophthalmol 1980; 12:245–250. retinal detachment, proliferative membranes grow along the
RETINA AND VITREOUS

retinal surface, creating star folds and retinal striae. In both of the sulfonamides should promote resolution of the retinal
instances, surgical removal of the membranes may be necessary. folds in these cases.6
Shallow superficial retinal folds may develop when choroidal
folds are lacking whenever there is relative choroidal thickening,
particularly in association with uveal inflammation. Reduction Key Features
of the surface area available to the retina from the radial dis- • Most typically present secondary to epiretinal membrane
placement of the choroidal surface is a likely cause. Retinal formation
folds in conjunction with transient myopia may also be associ- • May show a radial pattern if secondary to scar formation from
ated with the administration of sulfonamides. In this case, macular degeneration
swelling of the ciliary body and vitreous traction may play a • May be associated with metamorphopsia
role.6 Elimination of the uveal inflammation or discontinuation

REFERENCES
1. Newell FW: Choroidal folds. Am J pressure. Am J Ophthalmol 2001; 17. Ernst BB, Lowder CY, Meisler DM,
Ophthalmol 1973; 75:930–942. 131:158–159. Gutman FA: Posterior segment
2. Friberg TR: The etiology of choroidal folds. 10. Norton EWD: A characteristic fluorescein manifestations of inflammatory bowel
Graefes Arch Clin Exp Ophthalmol 1989; angiographic pattern in choroidal folds. disease. Ophthalmology 1991;
SECTION 10

227:459–464. Proc R Soc Med 1969; 62:119–128. 98:1272–1280.


3. Cangemi FE, Trempe CL, Walsh JB: 11. Hee MR, Izatt JA, Swanson EA, et al: 18. Schepens CL, Brockhurst RJ: Uveal
Choroidal folds. Am J Ophthalmol 1978; Optical coherence tomography of the effusion: clinical picture. Arch Ophthalmol
86:380–387. human retina. Arch Ophthalmol 1995; 1963; 70:189–201.
4. Wolter JR: Parallel horizontal choroidal 113:325–332. 19. Ward RC, Gragoudas ES, Pon DM,
folds secondary to an orbital tumor. Am J 12. Kokame GT, de Leon MD, Tanji T: Serous Albert DM: Abnormal scleral findings in
Ophthalmol 1974; 77:669–673. retinal detachment and cystoid macular uveal effusion syndrome. Am J Ophthalmol
5. Bullock JD, Egbert PR: The origin of edema in hypotony maculopathy. Am J 1988; 106:139–146.
choroidal folds: a clinical, histopathological Ophthalmol 2001; 131:384–386. 20. Dailey RA, Mills RP, Stimac GK, et al:
and experimental study. Doc Ophthalmol 13. Morgan CM, Gragoudas ES: Limited The natural history and CT appearance of
1974; 37:261–293. choroidal hemorrhage mistaken for a acquired hyperopia with choroidal folds.
6. Hertle RW, Leahey AB, Bloom S, et al: choroidal melanoma. Ophthalmology 1987; Ophthalmology 1986; 93:1336–1342.
Chorioretinal folds and a macular hole 94:41–46. 21. Jacobson DM: Intracranial hypertension
secondary to craniofacial surgery. Ophthal 14. Gass JDM: Radial chorioretinal folds: a sign and the syndrome of acquired hyperopia
Plast Reconstr Surg 1990; 6:278–282. of choroidal neovascularization. Arch with choroidal folds. J Neuroophthalmol
7. Ryan EH, Jampol LM: Drug-induced acute Ophthalmol 1982; 99:1016–1018. 1995; 15:178–185.
transient myopia with retinal folds. Retina 15. Altan T, Temel A, Baubeck T, Kazokoglu H: 22. Friberg TR, Grove AS: Subretinal
1986; 6:220–223. Hypotonic maculopathy after trabeculectomy neovascularization and choroidal folds.
8. Friberg TR, Grove AS: Choroidal folds and with post-operative use of 5-fluorouracil. Ann Ophthalmol 1980; 12:245–250.
refractive errors associated with orbital Ophthalmologica 1994; 208:318–320.
tumors. An analysis. Arch Ophthalmol 16. Stamper RL, McMenemy MG,
1983; 101:598–603. Lieberman MF: Hypotonous maculopathy
9. Griebel SR, Kosmorsky GS: Choroidal folds with subconjunctival 5-fluorouracil. Am J
associated with increased intracranial Ophthalmol 1992; 114:544–545.

2048
Acute Posterior Multifocal Placoid Pigment
CHAPTER
Epitheliopathy, Serpiginous Choroiditis, and
157 Relentless Placoid Chorioretinitis
Strutha C. Rouse, II and Lee M. Jampol

ACUTE POSTERIOR MULTIFOCAL PLACOID recurrent cases have been noted. These recurrent cases must be
PIGMENT EPITHELIOPATHY distinguished from serpiginous choroiditis (SPC), which may
be difficult initially.
APMPPE most often affects young men and women between
INTRODUCTION the ages of 20 and 50 years.3,4 Symptoms include the rapid
Acute posterior multifocal placoid pigment epitheliopathy onset of central or paracentral visual loss. Bilateral onset is the
(APMPPE) was first described in three, otherwise healthy rule but the second eye may be involved a few days or weeks
women by Gass in 1968.1 Although not the first to report the later. Photopsias can also be present and the visual acuity
constellation of clinical findings,2 Gass gave us a definitive maybe as low as counting fingers. About one-third of patients
description of the disease that speculated on its pathogenesis. will report an antecedent viral illness,1 often an upper respir-
atory infection. Other cases have followed vaccinations10 or
bacterial infections.11
CLINICAL PRESENTATION In the great majority of patients, the visual prognosis for
Typically, APMPPE causes central vision loss in one or both APMPPE is good, with most individuals spontaneously recover-
eyes in young adults with the development of gray, white, or ing vision without treatment or significant sequela. Lewis and
yellow, flat plaques that are predominantly located in the Gass both reported that the majority of patients regain vision
posterior pole at the level of the outer retina and retinal pigment of 20/40 or better.3,4 However, residual visual complaints are
epithelium (RPE) (Fig. 157.1). Lesions are usually about one disk common as elucidated by two studies,12,13 retrospectively
diameter in size. Fresh lesions may appear for several weeks. reviewing 75 eyes. Patients described scotomas (33%), meta-
Resolution of the visual symptoms begins within 2–4 weeks. morphopsias (21%), decreased vision (16%), floaters (5%), and
The acute lesions evolve into well-demarcated, lightly pigmented chronic redness (2%). Long-term vision loss may be related to
scars (Fig. 157.2). In addition, APMPPE may manifest a mild choroidal neovascularization, extensive RPE changes, or foveal
vitreitis and occasionally mild anterior chamber inflammation. involvement.14 Pagliarini et al12 noted features that may be
Usually, APMPPE is a nonrecurring syndrome,3–9 although rare associated with poor visual outcomes in APMPPE such as foveal

a b

FIGURE 157.1. (a) Fundus photograph of the right and (b) left eye of a patient with active APMPPE demonstrating deep, white, plaque-like
lesions in the macula. 2049
RETINA AND VITREOUS

a b
SECTION 10

FIGURE 157.2. (a) Fundus photograph of the right and (b) left eye of another patient demonstrates inactive APMPPE showing well-demarcated,
lightly pigmented scars in the outer retina.

involvement at initial presentation, older age at presentation demonstrates the first signs of healing and inactivity, a trans-
(>60 years), unilateral disease, longer interval between first and mission defect without late staining. Slowly, these changes
second eye involvement (>6 months) and recurrent disease. progress to involve the center of the plaque.

FLUORESCEIN ANGIOGRAPHY INDOCYANINE GREEN ANGIOGRAPHY


Fluorescein angiography for APMPPE has a characteristic Indocyanine green (ICG) angiography has been increasingly
pattern. Lesions demonstrate early hypofluorescence during utilized in recent years due to improvement of digital imaging
disease activity and these lesions show hyperfluorescence in the systems and enhanced image resolution. ICG has demonstrated
late frames with persistent staining for 20–30 min15 (Fig. 157.3). value in characterizing possible choroidal disorders, such as
Rarely, large choroidal vessels can be observed under these APMPPE. Several investigators have reported the ICG charac-
areas of hypofluorescence.16 teristics of APMPPE lesions, which substantiate the idea of
The inactive, healed phase of APMPPE displays a different primary choriocapillaris involvement.7,17–21 Acute lesions of
angiographic pattern. Lesions show early hyperfluorescence APMPPE show early hypofluorescence during ICG angiography
representing atrophic defects in the RPE with little to no with the borders of these lesions more sharply delineated in the
fluorescence in the later phase of the angiogram. late phase. The lesions are rounded and tend to be confluent.
In 1973, Annesley et al15 studied the evolution of angiographic ICG angiography may demonstrate more hypofluorescent areas
changes by performing consecutive fluorescein angiograms than are seen on clinical examination (Fig. 157.4). Inactive,
during follow-up visits. The peripheral portion of the lesion healed lesions display early hypofluorescence on ICG with

a b

FIGURE 157.3. (a) Early phase of fluorescein angiography in acute APMPPE demonstrates blockage of fluorescence. (b) Persistent staining is
2050 present in these lesions late in the angiogram (same patient as Fig. 157.1).
Acute Posterior Multifocal Placoid Pigment Epitheliopathy, Serpiginous Choroiditis, and Relentless Placoid Chorioretinitis

layer and inner and outer photoreceptor segments in healing


and inactive lesions suggesting photoreceptor damage early in
this disease course. Backscatter at the level of the outer retina
likely represents acute photoreceptor cell body swelling with
subsequent loss. Degenerative changes of the RPE and intra-
cellular edema, however, were visualized as increased back-
scatter of the underlying choroid. Similar OCT findings are
seen in patients with vascular occlusive disease. This supports
the vascular hypothesis in APMPPE, indicating intracellular
edema and inflammatory changes in the outer retina from
ischemic damage.25,26

ASSOCIATED OCULAR AND SYSTEMIC


FINDINGS
Vitreitis is seen in half the patients with APMPPE and papillitis
FIGURE 157.4. ICG angiogram of the same patient in Figure 157.1, and retinal vasculitis have been reported.27–32 Allee and Marks33
described bilateral central retinal vein occlusions that developed

CHAPTER 157
during the active phase of APMPPE illustrates multiple
hypofluorescent areas. Although some of these spots correspond to ~1 month after the onset of APMPPE. Disk edema and nerve
the photographic lesions, there are more spots apparent, during ICG fiber layer hemorrhages were present 1 week prior to the vein
angiography. occlusions suggestive of a peripapillary vasculitis leading to disk
swelling and compression of the central retinal vein. De Souza
and colleagues30 reported a case of APMPPE that displayed
better depiction late in the angiogram. However, these regions signs of retinal vasculitis with subsequent retinal neovascular-
are more irregularly shaped, smaller in size, and less confluent ization and vitreous hemorrhage 6 years after presentation.
than acute lesions.17 Other reports describe more extensive inflammatory processes,
Persistent hypofluorescence was observed by Cimino et al22 including peripheral corneal thinning,34 corneal stromal
in healed APMPPE lesions secondary to choroidal nonperfusion infiltrates,35 episcleritis, iridocyclitis,28 and choroidal vasculitis.
and chorioretinal atrophy. These findings are consistent with APMPPE has also been described with diffuse serous retinal
the current theory of choroidal occlusive vasculitis in APMPPE. detachments23,36,37 and hemorrhagic retinal detachments.29
It is likely that nonperfusion in conjunction with blockage of Neurological abnormalities in association with APMPPE may
fluorescence by edematous, damaged RPE may be present in include headache1,38–41,42 and cerebrospinal fluid (CSF) pleo-
active lesions. The inactive, healed spots are smaller in dimen- cytosis,27,38–41,43–46 which usually improve spontaneously.
sion and more irregular in shape. These changes would be However, some patients can experience episodes of transient
explained by resolution of inflammatory changes of the RPE. cerebral ischemia,27,42 producing stroke-like symptoms, optic
neuritis,6,31,33,34,47 transient hearing loss,48 and permanent focal
deficits43,45 due to vasculitic changes. APMPPE has been
OPTICAL COHERENCE TOMOGRAPHY described in association with presumed cerebral vasculitis in a
The novel technique of optical coherence tomography (OCT) dozen cases.27,39,42,43 Ten of these individuals suffered a stroke
has been applied to patients with APMPPE. Using standard and deaths have occurred. In one case, histopathology of the
OCT, Garg and Jampol23 documented backscatter at the outer brain demonstrated granulomatous inflammation of medium
retina that corresponded to the placoid lesions. This case report size arteries49 while biopsy from the tibialis anterior muscle was
also described serous detachment with reflective material suggestive of vasculitis in another.43 Comu et al42 described
within the subretinal fluid (Fig. 157.5). They proposed that this three patients with APMPPE and neurologic manifestations.
represented proteinaceous material within the serous fluid or Visual disturbance and headache were shared by all three and
damaged edematous retinal epithelium. they had a normal radiologic scan initially but CSF pleocytosis.
Scheufele et al24 illustrated similar findings in active and One patient developed recurrent strokes involving different
healed APMPPE lesions with better clarity using ultrahigh- vascular distributions that required immunosuppressants for
resolution OCT. They observed thinning of the outer neural presumed cerebral vasculitis.

FIGURE 157.5. (a) Color photo and (b) OCT of


right eye of patient with APMPPE at
presentation. Note the increased reflectance at
the level of the outer retina that corresponded
to lesions seen clinically in this patient. (c) OCT
of left eye. Note serous detachment and
b reflective material within the fluid.

a c
2051
RETINA AND VITREOUS

APMPPE has also been associated with other systemic con- substantial evidence of APMPPE-like changes in association
ditions including erythema nodosum,50,51 group A streptococcal with ocular and systemic vasculitic conditions, which impli-
infection,11 clear cell renal cell carcinoma,52 thyroiditis,34 cates an inflammatory process of the choriocapillaris. Diseases
pulmonary tuberculosis,53 ulcerative colitis,54 Wegener’s granu- like Wegener’s granulomatosis55 and ulcerative colitis54 have
lomatosis,55 and systemic vasculitis.56,57 Some of these associ- both been reported in association with APMPPE-like changes.
ations may be coincidental but others may indicate systemic Van Buskirk et al50 first reported the fluorescein angiographic
triggers for the APMPPE response as described below or different changes of APMPPE as a primary vascular abnormality of the
manifestations of a systemic vasculitis. choroid. They described the hypofluorescence as impaired
The etiology of APMPPE remains a subject of debate. A viral filling of the choriocapillaris. Many investigators who support
illness may be an immunologic trigger for some individuals. this idea feel the placoid lesions represent focal infarcts of the
Azar et al58 and most recently Thomson et al59 both described RPE caused by choroid vascular disease. Hayreh’s studies of
cases of APMPPE occurring with culture-proven adenovirus the choroidal vasculature support the hypothesis of a vascular
infections. Recombinant hepatitis B vaccination, reported by pathologic process.64 He believed the angiographic appearance
Brezin,10 caused two patients to develop APMPPE between of APMPPE lesions denoted occlusions of precapillary choroidal
3 days and 2 weeks after a booster shot. Despite the lack of arterioles.
histologic evidence, the possibility of a virus or vaccination Deutman and colleagues16 reported the appearance of large
causing APMPPE supports an immune-mediated scheme. Some choroidal vessels seen under areas of hypofluorescence. This
investigators have speculated that molecular mimicry could evidence suggested that diseased RPE could not fully account for
SECTION 10

play a significant role in damage to the RPE or occlusion of the the lack of choroidal fluorescence but rather suggested chorio-
choriocapillaris. In a patient who is genetically predisposed to capillaris nonperfusion. They surmised that APMPPE lesions
immune dysregulation, environmental triggers may precipitate signify choroidal ischemia with subsequent RPE involvement.
APMPPE. A case of atypical APMPPE occurred in a patient with
clear-cell renal-cell carcinoma; high levels of circulating
immune complexes accompanied the fundus findings.52 This MANAGEMENT/THERAPY
possibly implicates immune complexes in tandem with the No therapy has been proven to affect the short-term or long-
complement cascade in the development of choroidal vascular term outcome of APMPPE. Corticosteroids are sometimes used
occlusions. This theory is reinforced by a documented case of with the rationale that this may reduce the impairment of
APMPPE after acute group A streptococcal infection with an central vision and speed healing. This has not been proven.
observed rise in anti-DNAase B antibody titers.11 Interestingly, However, in cases where central nervous system inflammation
a patient with APMPPE in the course of ulcerative colitis was is present with APMPPE, immunosuppressive agents have been
documented in 2001.54 Mathura et al described a patient who utilized to treat these patients with success in some cases.42–44
had an episode of APMPPE and then, years later, developed
multifocal choroiditis, distinctly different from the earlier
lesions.60 Thus, a patient with immune (or genetic) dysregula- DIFFERENTIAL DIAGNOSIS
tion may manifest at different times as different white spot The majority of patients with APMPPE are easily diagnosed
syndromes. based on the classic clinical findings and course. However,
Human leukocyte antigens (HLAs) B7 and DR2 have been other inflammatory processes of the choroid should be con-
associated with APMPPE.61 Two cousins were described with sidered when contemplating the diagnosis of APMPPE. Serpiginous
recurrent APMPPE and both were found to have HLA DR2, choroiditis (SPC), multifocal choroiditis, multiple evanescent
promoting the concept of an inherent propensity to acquire this white dot syndrome, relentless placoid chorioretinitis (RPC),
disease.62 Undoubtedly, other immune regulatory genes will be diffuse unilateral subacute neuroretinitis, choroidal neoplasms,
involved.63 birdshot chorioretinopathy, and others can resemble APMPPE.
Additionally, infections like syphilis and granulomatous diseases
like sarcoidosis can resemble APMPPE lesions. The individual,
PATHOPHYSIOLOGY acute lesions of APMPPE, serpiginous, and relentless are not
Gass believed that the abnormalities in APMPPE were at the clearly distinguishable at times. However, the clinical course
level of the RPE.1 He felt that the early blockage of fluorescein and setting usually allows diagnosis.
was due to acute physiologic alterations in the RPE that
accounted for the opalescent appearance. The late staining
typical of this disorder could be explained by the uptake of CONCLUSION
fluorescein into damaged pigment epithelium. The RPE APMPPE presents typically in early adulthood and affects the
eventually recovers along with some of the photoreceptors, central vision. The visual symptoms begin to resolve over
allowing the return of visual function, despite areas of signifi- 2–4 weeks. APMPPE has a characteristic fluorescein angio-
cant pigment clumping or atrophy. graphic pattern and clinical course. The prognosis for visual
Others agreed that edematous RPE cells absorb the under- recovery is good and the exact pathologic process involved is
lying choroidal fluorescence but hypothesized that, during the still debatable. To date, there is no proven treatment.
acute process, an infectious or immunologic insult occludes the
choriocapillaris, sparing the larger vessels.27 SERPIGINOUS CHOROIDITIS
Further supporting the idea of a primary RPE process, the
electroculogram (EOG) findings are remarkably impaired during
the acute phase of the disease. However, the electroretinogram
INTRODUCTION
(ERG) responses are normal. Fishman concluded that these SPC is a rare, chronic, recurring, usually bilateral disease,65–69
findings demonstrated RPE dysfunction.5 If the choriocapillaris which has also been called geographic choroiditis,70,71 geographic
were primarily involved, both EOG and ERG responses would choroidopathy,70,72 geographic helicoid peripapillary choroido-
be extinguished. pathy,65,69,70,73 helicoid peripapillary chorioretinal degeneration,72
At the present time, most investigators currently attribute and macular geographic helicoid choroidopathy.74 The disease
2052 the hypofluorescence to hypoperfusion of the choroid. There is has been reported in whites,75 Asians,74 African-Americans,76
Acute Posterior Multifocal Placoid Pigment Epitheliopathy, Serpiginous Choroiditis, and Relentless Placoid Chorioretinitis

and Hispanics.66 Men are affected slightly more frequently than area, and visual field examination reveals small, central or para-
women.77 The onset of SPC is usually between ages 30 and central scotomas, either absolute or relative.80 In the early stages
70 years,68,78 although affected patients in their 20s have of the episode, the scotomas are more frequently absolute.76
occasionally been reported.76,79 Amsler grid visual field testing reveals scotomas that correspond
precisely with the clinical lesions.73 Acute peripapillary or
macular geographic lesions appear clinically as gray to grayish-
CLINICAL PRESENTATION yellow placoid lesions, owing to outer retinal or retinal pigment
Patients with SPC usually report painless blurring of epithelial thickening with translucency (Figs 157.6 to 157.8).80
vision.76,79,80 Vision loss can vary, depending on the affected Peripapillary lesions, when present, are discoid, often showing

FIGURE 157.6. (a) Active focus of recurrent


SPC (arrows). (b) Late-phase angiogram shows
leakage from two foci of choroiditis.
From Jampol LM, Orth D, Daily MJ, Rabb MF:
Subretinal neovascularization with geographic
[serpiginous] choroiditis. Am J Ophthalmol 1979;
88:683–689.

CHAPTER 157
a b

a b

FIGURE 157.7. (a) Active initial lesion of macular SPC. (b) Same patient 3 years and several recurrences later with serpentine extension.
From Mansour AM, Jampol LM, Packo KH, Hrisomalos NF: Macular serpiginous choroiditis. Retina 1988; 8:125.

a b c

FIGURE 157.8. (a) Unusual-shaped lesion of macular SPC. (b) Early-phase angiogram shows hypofluorescence. (c) Late-phase angiogram
shows staining of lesion.
Courtesy of Kirk Packo, MD. 2053
SECTION 10 RETINA AND VITREOUS

FIGURE 157.9. End-stage scarring of inactive SPC.


FIGURE 157.10. Inactive lesion of macular SPC.
From Mansour AM, Jampol LM, Packo KH, Hrisomalos NF: Macular serpiginous
choroiditis. Retina 1988; 8:125.
multiple confluent old scars and finger-like projections
(Fig. 157.9; see also Figs 157.6 and 157.7).81 These projections
may completely envelop small areas with normal appearance.
The EOG is unaffected.82,83 When extensive disease is
present,83 however, ERG values have been shown to fall to
70–80% of normal for total retinal illumination and to 20–30%
of normal for local macular illumination.82
Individual acute lesions normally last from weeks to
months.68,80 Resolution of the lesion results in either pigmen-
tary loss or hyperplastic mottling of the RPE, variable atrophy of
the choriocapillaris and large choroidal vessels, and scarring
(Figs 157.10 to 157.12; see also Figs 157.6 to 157.9).69,81
Characteristically, patients show evidence of multiple recur-
rences at intervals of several months to years during follow-
up.66,80 Recurrent lesions most frequently spread centrifugally
from the disk, preferentially moving toward the macula (see
Figs 157.6, 157.7, 157.10, and 157.11), although some cases
spread from the macula to the disk.69,78,80 New lesions are
often, but not always, contiguous with the borders of previous
scars.69,78,80 These new lesions are believed to be genuine recur-
rences rather than a simple progression of previous lesions,
owing to the prolonged period between the resolution of one
episode and the subsequent recurrence.69,78,80
Although blurred vision is usually the patient’s chief com-
plaint, this is frequently not noticed until the fovea becomes
involved.73,74,76,81 Many patients show evidence of previous
lesions at the time of their first examination, representing extra-
foveal disease that was previously active.66,73,76 For the patient FIGURE 157.11. Finger-like inactive lesion that eventually reached the
presenting with foveal involvement, vision may be rapidly and fovea.
permanently lost; finger-counting vision is frequently encoun- From Jampol LM, Orth D, Daily MJ, Rabb MF: Subretinal neovascularization with
tered in these patients.69,78 geographic [serpiginous] choroiditis. Am J Ophthalmol 1979; 88:683–689.
Commonly, lesions may present in the macula without pre-
ceding peripapillary activity, a variant known as macular SPC
(see Figs 157.7 to 157.9). These cases characteristically present
early in their course, owing to the early loss of central vision.66 FLUORESCEIN ANGIOGRAPHY
Other portions of the posterior pole, and even the periphery, are During the early phase of fluorescein angiography, the lesions
uncommon but occasional sites of involvement.78 are hypofluorescent (see Fig. 157.8b).65,73,80,82 The borders of
Virtually all patients eventually show bilateral disease; how- these lesions may hyperfluoresce throughout the angiography,
ever, it is uncommon to see bilateral simultaneous onset of representing leakage from the surrounding choriocapillaris. The
the disease.69 Additionally, the course and complications of the hypofluorescence of the interior represents either blockage by
disease are not necessarily the same for both eyes; one eye may swollen RPE cells or nonperfusion of the choriocapillaris, or a
show primary peripapillary involvement, whereas the other may combination of the two. Later in the angiogram, the inner
2054 show macular SPC.73 portions of a lesion may show spotty hyperfluorescence, which
Acute Posterior Multifocal Placoid Pigment Epitheliopathy, Serpiginous Choroiditis, and Relentless Placoid Chorioretinitis

CHAPTER 157
a b

FIGURE 157.12. (a) Peripapillary SPC with choroidal neovascularization at its temporal margin. (b) Angiography shows choroidal neovascular
membrane.
From Jampol LM, Orth D, Daily MJ, Rabb MF: Subretinal neovascularization with geographic [serpiginous] choroiditis. Am J Ophthalmol 1979; 88:683–689. Reprinted
from Elsevier Science.

with time becomes more intense and may spread within the
previously hypofluorescent regions (see Figs 157.6b and
ASSOCIATED OCULAR AND SYSTEMIC
157.8c).82,84 The areas of active inflammation eventually stain.73
FINDINGS
Over a period of several weeks, the active lesion shows pig- Vitreitis is seen in up to a one-third of individuals with
mentary atrophy and clumping, with variable destruction of the SPC.69,78,80 A mild, nongranulomatous, anterior uveitis has been
choroid. The atrophy is usually much more pronounced than reported but is much less common.91
APMPPE. Angiography shows mottled hyperfluorescence due to Generally, the optic nerve is not affected,69,73 although Wu and
pigment clumping.78 Late staining, representing leakage of dye colleagues described a patient with temporal sectorial atrophy of
from damaged choriocapillaris at the periphery of the lesion, is the optic nerve.81 However, photocoagulation had been per-
also observed.85–88 formed in this patient. Fujisawa and co-workers reported a
patient with evidence of recurrent disease who developed ‘optic
neuritis’.84
ICG ANGIOGRAPHY Choroidal neovascularization is a common complication of
Ciulla and Gragoudas have reported the results of ICG angio- SPC, and it may be more common in women than in men (see
graphy in one patient with an acute lesion in one eye and an Fig. 157.12).77 In a 1982 study of 53 patients, Blumenkranz and
inactive lesion in the fellow eye.89 Their findings were similar associates reported that 13% (seven patients) had active choroidal
to those with fluorescein angiography, except that the borders of neovascularization.77 Five of these patients demonstrated bilat-
both active and inactive lesions were less well defined and did eral SPC, but only two showed evidence of bilateral choroidal
not show late staining. neovascularization, demonstrating that the course of the
Giovanni and associates reported their findings in 17 patients disease can vary between eyes in the same patient. Since then,
with SPC.90 In one patient, they found evidence of a possible Gass has reported that the incidence of choroidal neovascular-
active area of choroidal involvement extending beyond the ization may be as high as 25%,69 although Nussenblatt has
borders of a lesion noted clinically and by fluorescein angio- reported an incidence of just 10%.92
graphy. Additionally, resolution of choroidal late staining in The time course for the development of the choroidal neo-
subacute lesions was noted before this resolution on fluorescein vascularization is uncertain; the asymptomatic nature of SPC
angiography. In contrast to the findings of Ciulla and before foveal involvement often allows the disease to progress,
Gragoudas,89 the healed lesions behaved similarly on both ICG so that neovascularization may be seen at the first examination.68
and fluorescein angiography, although the authors found clearer Neovascular membranes have been reported to occur as soon as
delineation of the atrophic choroid with ICG.90 7 months after presentation with acute SPC.93 As with other
Giovanni and associates also noted two cases with possible entities, hemorrhage, exudate, and retinal thickening may be
de novo lesions in patients with dormant SPC.90 In one, multi- associated with choroidal neovascularization in SPC. Some of
focal hyperfluorescent lesions were seen bilaterally on ICG, these neovascular membranes have been treated successfully
although one eye had not shown clinically active disease. In the with thermal laser photocoagulation or surgery (see section
second patient, ICG revealed late hypofluorescent spots; this, on Management/Therapy), whereas others have shown spon-
however, could represent blockage. These authors did not change taneous regression.93
their treatment strategy based on the findings in these two Retinal and optic disk neovascularization has been reported
patients and noted that further follow-up would be required to with SPC.77,93–95 Retinal edema and localized serous detach-
determine if these lesions represent early disease activity.90 ment (usually in association with choroidal neovascularization) 2055
RETINA AND VITREOUS

of the neurosensory retina may occasionally be seen.68,69,91 in eight patients with SPC. None of the patients was found to
Wojno and Meredith also reported localized RPE detachment.94 have rheumatologic or vascular disorders on follow-up.67
Their proposed mechanism suggests that irritation of the RPE
and Bruch’s membrane decreases the adherence of the two
layers, allowing fluid buildup beneath the RPE. The detached HISTOPATHOLOGY
RPE cells may then show pump dysfunction, resulting in a Histopathologic findings, including atrophy of the chorio-
serous retinal detachment. These serous detachments overlie capillaris, photoreceptors, and RPE, were reported by Wu and
RPE detachments and extend slightly beyond their borders.94 colleagues.81 Fibrocellular scars lined the inner portion of
Macular involvement is one of the most common compli- Bruch’s membrane in the regions of RPE and photoreceptor cell
cations of SPC; the disease may originate in the macula,66 or a atrophy. The choriocapillaris was acellular. A diffuse and focal
peripapillary lesion may spread to the macula.69,78,80 Foveal infiltration of lymphocytes was seen in the choroid. Infiltration
involvement causes a rapid decrease in central vision and has a was most prominent at the borders of RPE atrophy and
poor prognosis for recovery. Disciform macular scars, with or lymphocytic infiltration was also present in the venous walls.81
without active subretinal membranes, have been reported in as
many as 26% of patients.77
Retinal vasculitis, usually in the form of periphlebitis, has ETIOLOGY
been observed in patients with SPC.76,80,91,96 Occasional branch Maumenee, in his 1970 Edward Jackson Memorial Lecture to
vein77,96 or combined branch vein and artery occlusions have the American Academy of Ophthalmology and Otolaryngology,
SECTION 10

been described.91 In one report of a patient with SPC and asso- stated that the entity of SPC ran a spectrum that included as its
ciated branch vein occlusion with retinal periphlebitis, the patient two end points vascular abiotrophy and choroidal inflamma-
had ipsilateral optic nerve head drusen; the authors speculated tion.100 Since then, the theory of vascular abiotrophy, perhaps
that the drusen could have contributed to venous stasis and genetic and similar to various hereditary choroidal atrophies,
subsequent vein occlusion.96 The retinal vasculitis and vascular has not been substantiated. As recently as 1981, however,
occlusion could be secondary to a spillover of choroidal inflam- Richardson and colleagues suspected a degenerative cause in
mation or an antigen-antibody reaction of the retinal vessel wall. their analysis of a case of SPC associated with nongenetic
Erkkila and associates reported a 54.5% prevalence of unilateral extrapyramidal dystonia.72
HLA-B7 in Finnish patients with SPC,95 which is greater than Compelling evidence supporting an inflammatory cause of
that of the general Finnish population (24.3%).79,97 HLA-B7 was SPC has been set forth by Baarsma and Deutman82 in support
found in all three patients with choroidal neovascularization of Schlaegel (1969)71: an edematous zone surrounds the active
and in one patient who showed neovascularization of the disk.95 lesion, and lesions expand through this edematous zone. The
Other HLA associations have not be proved.79,95 One case of variable time course of bilateral involvement suggests that SPC
familial SPC has been reported that demonstrated autosomal is not of a degenerative origin. Degenerative disorders rarely
dominant inheritance with complete penetrance.94 show restoration of eyesight to any marked degree, unlike that
In most reports, no systemic disease has been detected in sometimes seen in SPC. Familial occurrence of SPC is rare.82
patients with SPC.76,79,91,95 Some studies, however, have found The inflammatory etiology is supported by the occasional
singular associated systemic manifestations, many of which are presence of anterior uveitis, retinal vasculitis, vitreitis,68 and
probably chance associations. optic disk neovascularization.94
Edelston and colleagues have reported two cases of biopsy- An immune response induced by a tissue or microbial antigen
proven sarcoidosis in patients who developed acute lesions could play a role in this inflammatory etiology. Recurrences
resembling those of SPC late in the course of their disease could be caused by reactivation of the immunologic reaction95
(21 and 51 years after diagnosis of sarcoidosis).98 In one patient, or by an underlying infection.
the episode represented apparent reactivation of choroiditis In support of an immunologic etiology, Erkkila and associates
in one eye, which had been quiet from age 29 until age discovered an increased prevalence of HLA-B7 (previously dis-
70 years. The lesions in these patients showed the cussed).95 These authors have also described a decline in com-
characteristic changes of SPC on fluorescein angiography and plement factor C3 in two patients.95 Broekhuyse and colleagues
demonstrated the typical helicoid progression of SPC during described the presence of immune responsiveness of SPC
follow-up.98 patients to retinal photoreceptor protein.101 Immune reactivity
Richardson and colleagues report a case of SPC and nongenetic may depend on release of autoantigens and leakage of S-antigen
dystonia (simultaneous contraction of antagonistic muscle through the blood–retina barrier at the level of the RPE.
groups) believed to be acquired secondary to remote trauma, an Attempts to demonstrate a microbial cause of SPC have been
unknown degenerative process, or microinfarction secondary to unrevealing. An infectious etiology or trigger remains appealing,
migraine.72 These authors cited two other cases that may however, because recurrences could indicate reactivation of
suggest a predisposing degenerative link between SPC and the infection, as seen with toxoplasmosis or herpesviruses. A
neurologic disorders, including a young man with SPC and a tubercular allergic cause has been proposed for patients with a
relative absence of arm swing during walking and an SPC history of tuberculosis or a positive Mantoux reaction.80 Schatz
patient whose sister had multiple sclerosis.72 and co-workers reported a single case of active SPC lesions
A case of celiac disease associated with SPC and autoimmune concurrent with both maxillary sinusitis and ‘influenza’.65
thrombocytopenic purpura has been reported.99 Laatikainen and Erkkila noted one patient who had viral
Vascular endothelial injury releases von Willebrand factor meningitis before the first attack and three patients with
(vWF), which complexes with factor VIII. Factor VIII is necess- increased antistreptolysin titers but without evidence of focal
ary for normal clotting cascade activity; its deficiency results in infection.80 One patient had been receiving estrogen-type
hemophilia. vWF increases platelet adhesion. Elevated levels of hormonal therapy for menopausal symptoms for 2 years before
vWF have been associated with several disorders that manifest onset of symptoms.80 These four patients were part of a group
vascular occlusion as a primary component, including poly- of nine for whom systemic antibody titers were negative for a
myalgia rheumatica, Raynaud’s phenomenon, and scleroderma. variety of viruses or other infectious entities. Mansour and
King and associates reported elevated factor VIII-vWF antigen colleagues also found no evidence of preceding viral illnesses.66
2056
Acute Posterior Multifocal Placoid Pigment Epitheliopathy, Serpiginous Choroiditis, and Relentless Placoid Chorioretinitis

To the contrary, Gass described one patient who developed marked infiltration of the vitreous. Visual loss with toxoplas-
bilateral SPC after herpes zoster ophthalmicus.102 Yet, Akpek mosis may be secondary to direct foveal involvement, macular
and colleagues cited postmortem histologic findings of an eye edema, or media opacities.96 Toxoplasmosis may be seen any-
with a long history of SPC and were unable to isolate herpetic where in the fundus. Additionally, the scars of toxoplasmosis
viral genomic fragments (herpes simplex, Epstein–Barr virus, are much more atrophic than those of SPC, indicating more
cytomegalovirus, and herpes zoster) in the infiltrating lympho- extensive destruction of the retina, RPE, and choriocapillaris.96
cytes or choroidal tissues, using PCR amplification.103 Systemic diseases may cause choroidal ischemia in the pos-
Friberg has reported elevated Toxoplasma gondii titers in a terior pole. These include hypertensive vascular disease, systemic
patient with SPC with branch vein occlusion and bilateral lupus erythematosus, polyarteritis nodosa, preeclampsia,
periphlebitis, but he was cautious in pointing out that serum eclampsia, disseminated intravascular coagulation, and throm-
Immunoglobulin (Ig) levels are often elevated for months to botic thrombocytopenic purpura.66 The fluorescein pattern in
years after an active toxoplasmosis infection and that serum these conditions may resemble that of SPC, but the clinical course
IgM levels, which would have been indicative of an active is different. Marked atrophy of the RPE and choroid as seen with
primary infection, were not obtained.96 SPC does not occur with these choroidal ischemic syndromes.

PATHOPHYSIOLOGY MANAGEMENT/THERAPY
Choroidal vascular occlusion could be the mechanism through Various antiinflammatory and immunosuppressive agents have

CHAPTER 157
which either an autoimmune disease or an infectious agent been used to treat SPC. Most efforts do not appear to halt
could incite inflammation and tissue damage. Although King recurrence of the disease,76 although these therapies may be
and associates did not find an immunologic association for tried when macular vision is threatened. Systemic prednisone,
SPC, they suggested that vascular occlusion is the most 60–80 mg/day, is a commonly prescribed therapeutic regimen,
probable mechanism of tissue damage. Areas of normal retina but it has not been proved to affect the recurrence rate or the
can be found immediately adjacent to affected areas, which is long-term outcome of the disease.76,80 Many experienced
suggestive of a precisely defined loss of choroidal filling. This observers, including one of the authors (LMJ), believe that the
cannot be explained by blockage of fluorescence from inflam- acute lesions of SPC heal more rapidly with systemic,
mation at choroid or RPE alone.67 Mansour and colleagues, periocular, or intraocular corticosteroids and that maintenance
however, found evidence of hyperfluorescence in an atrophic steroids can prevent recurrences.
laser spot in an active lesion of macular SPC.66 The laser spot, Araujo et al used oral cyclosporine at 3–5 mg kg–1 day–1 along
from treatment of previous subretinal neovascularization, with varying doses of prednisone to treat seven patients with
provided a window defect through involved RPE and suggested active SPC and to analyze the effects on the rate of recur-
intact underlying choroidal fluorescence.66 Thus, the hypofluor- rence.106 The median duration of treatment was 3 years, ranging
escence seen in the angiogram of an active lesion may be due at from 1.3 to 5 years; five patients demonstrated remission and
least in part to RPE opacity and not to underlying choroidal avoided recurrences while on this regimen. However, only two
vascular occlusion.66 of the seven patients actually achieved drug-free remissions (2.5
and 1.6 years) after cyclosporine therapy for 3.5 and 2.5 years,
respectively. There were no serious complications reported in
DIFFERENTIAL DIAGNOSIS this series related to the long-term use of cyclosporine.
APMPPE and SPC both present as flat geographic yellowish Hooper and Kaplan described triple immunosuppressive
white lesions of the outer retina and RPE with similar fluor- therapy, with low doses of azathioprine (Imuran), cyclosporine,
escein angiographic patterns. Most notably, hypofluorescence and prednisone in combination.107 All five of their patients
during the acute stages of the disease is seen with both showed resolution of active choroidal infiltrates within 2 weeks
lesions.66,82,96,104 In APMPPE, however, most lesions largely of beginning therapy. Recurrence was immediate in one patient
resolve within weeks,69 and recurrences are uncommon more when therapy was discontinued but ceased when the therapy
than several months after disease onset. A viral prodrome is was resumed. The authors suggest a synergistic effect between
often seen before ocular manifestations in patients with the three agents used. Nussenblatt, however, remarked in his
APMPPE. SPC invariably shows recurrences over periods of discussion of the article that although this regimen may be
many months or years after disease onset,66,73,82 and a viral effective for long-term immunosuppression, the short-term
prodrome is not seen. The scarring in APMPPE is generally effects demonstrated may be due to the effects of the steroids
limited to pigment mottling.80,82,104 Choroidal atrophy is alone or may even represent the natural resolution of the
usually not prominent in APMPPE;66,80,83 it is the rule with disease.108 Alkylating agents, such as cyclophosphamide or
SPC. Patients with APMPPE and macular involvement have a chlorambucil, in conjunction with oral steroid seemed effective
much more favorable visual outcome.66,73,105 Visual acuity, even at controlling the acute inflammation and allowing long-term
after foveal involvement, often recovers dramatically in patients drug-free remission in another report.109 Seven of the nine
with APMPPE (although this may be delayed), but recovery is patients managed to reach a drug-free remission after 15–96
rare in patients with SPC.73 APMPPE patients are generally months of treatment (median, 78 months). However, the side
younger, with simultaneous multifocal bilateral involvement effects were considerable, including nausea, and fatigue. One
throughout the posterior pole;66,73 in contrast, SPC patients are patient developed transitional epithelial carcinoma of the
most frequently middle-aged, with activity of the disease in bladder 3 years after succession of treatment, related to the
each eye generally occurring sporadically. Choroidal neovas- cyclophosphamide therapy.
cularization has been seen in APMPPE, but it occurs much less Secchi and co-workers have reported improved visual acuity
frequently than with SPC.73 in seven patients treated with cyclosporine as monotherapy,
Like those of SPC, the recurrent lesions of retinal toxoplas- with recurrence in one of three patients in whom therapy was
mosis usually begin at the margin of an inactive scar.96 How- discontinued.110 Using similar doses of cyclosporine as Secchi’s
ever, only one-third of SPC patients show evidence of vitreous group, Leznoff and colleagues treated 18 patients with
cells, whereas patients with acute toxoplasmosis usually show nonmicrobial uveitis. Of these, three carried a diagnosis of SPC.
2057
RETINA AND VITREOUS

Two showed no improvement after 4 months of therapy each, includes the development of multiple, new lesions, growth of
despite the addition of prednisone and azathioprine in one previous lesions and continued inflammation of some older
patient and prednisone in the second. The third patient showed lesions over many months (5–24 months). The disease process
dramatic improvement after the addition of prednisone and eventually involves all areas of the retina, including anterior to
azathioprine. This patient had the shortest duration of disease the equator.
(0.2 years vs 1 and 2 years in the nonresponders).111 RPC affects patients between 20 and 50 years old. Concurrent
The long-term outcome of five patients treated with inflammation of both eyes occurs in the majority of patients
interferon alpha-2a was described by Sobaci and colleagues.112 with frequent foveal involvement. Central vision can be poor
The eight eyes in this report had vision-threatening SPC that but recovery is sometimes seen, despite foveal RPE changes.115,116
failed to respond to steroid-cyclosporine or chlorambucil. The Only two out of 11 eyes, in Jones and associates’ case series,
authors found that all lesions were quiescent after this had a final vision worse than 20/40.115 No gender predilection
treatment and no recurrences were noted during the follow-up was noted.
period (16–48 months). Early flu-like symptoms were the only The individual lesions of RPC are similar to those seen with
adverse reaction observed and recovery of vision or stability of APMPPE. However, the lesions demonstrate growth in size and
useful vision was documented. This suggests that interferon contiguous relapses over several months.115 Recurrence is a
alpha-2a may be an option for the treatment of SPC but further classic feature of SPC in which a serpentine extension of existing
studies are necessary. lesions is routinely seen. For APMPPE, recurrences have been
Cases of SPC complicated by extrafoveal choroidal neovas- clearly documented but remain a rare clinical finding.8
SECTION 10

cular membranes with attendant hemorrhage, exudate, or


serous retinal detachment have been successfully treated with
laser photocoagulation.66,68 For subfoveal choroidal neovascular FLUORESCEIN ANGIOGRAPHY
membranes, present management could include photodynamic The fluorescein angiography of patients with RPC resembles
therapy, intraocular triamcinolone, or antivascular endothelial those of APMPPE or SPC. Lesions are hypofluorescent in the
growth factor agents. Improvement in visual acuity may occur early phase due to blockage or choriocapillaris nonperfusion.
after treatment, but lesions located in the foveal avascular zone Later, hyperfluorescence develops, starting at the borders of the
have a poor prognosis with treatment.77 Schatz and McDonald lesion, with uniform late staining of the underlying fibrotic scar
noted that new lesions in patients with SPC should be assessed and sclera.20
carefully to determine whether they represent fresh inflam-
mation or choroidal neovascular membranes.73 Navajas et al
cited treatment of a peripapillary choroidal neovascular mem- ICG ANGIOGRAPHY
brane secondary to SPC utilizing ICG-mediated photothrombosis ICG angiography findings reported with RPC are again similar
with intravitreal triamcinolone. This case report demonstrated to APMPPE or SPC.20,90,117,118–122 The ICG angiographic pattern
recovery of vision from 20/200 to 20/20 over a 10-week course, is characterized by hypofluorescent areas initially that persist
which was maintained up to 1 year of follow-up.113 into the late phase.
Surgical removal of choroidal neovascular membranes, either
peripapillary or submacular, has shown some benefit in specific
groups of patients but has never been systematically studied in
ASSOCIATED OCULAR AND SYSTEMIC
SPC because of its rare nature. Given the extensive RPE and FINDINGS
choroidal damage from SPC, it would unlikely be of much Vitreitis is a frequent manifestation seen with RPC. Both Lyness’
benefit. and Jones’ description of this entity reported that over half of
their patients presented with vitreitis.8,115 Also noted were
episcleritis,115 keratic precipitates,115 and optic disk swelling,33
MONITORING DISEASE ACTIVITY all of which support an inflammatory process at least within the
Amsler grid testing is an excellent means for assessing the ocular tissues if not a more global inflammatory disease.
activity of SPC or the effectiveness of therapy in patients with No consistent systemic disorder has been demonstrated in
SPC because scotomas can be mapped precisely on the grid.114 patients with RPC. However, one of Jones’ patients had herpes
Activity of the disease or secondary choroidal neovascular- keratitis.115 Some patients who subsequently developed RPC
ization may affect the Amsler test. had upper respiratory tract infections that were treated with
antibiotics. A case series in 1984 by Lyness et al described
RELENTLESS PLACOID CHORIORETINITIS ‘recurrent APMPPE’ in seven patients; three of these individuals
used antimicrobial agents on multiple occasions with con-
current episodes of blurred vision and characteristic lesions
INTRODUCTION consist with RPC. The authors concluded that an antibiotic
RPC is a newly recognized disease process that resembles could possibly act as an antigenic trigger for this disease.8 Other
APMPPE and SPC in many respects.115 Unlike these two systemic findings seen in association with RPC included
diseases, RPC shows a progressive, relentless course with the erythema nodosum, thyroiditis, and aseptic meningitis, which
widespread development of dozens to hundreds of inflamma- all have been previously described with APMPPE or recurrent
tory areas. Previous cases, called multifocal SPC,116 ampiginous APMPPE.8,38,34,50,51,58,80,115
choroiditis,92,117 or recurrent APMPPE,8 may represent RPC.
PATHOPHYSIOLOGY
CLINICAL PRESENTATION The individual lesion of RPC resembles APMPPE and SPC.
Patients with RPC develop multiple placoid, white lesions at the Fluorescein and ICG angiography both suggest an element of
level of the outer retina and RPE115 (Fig. 157.13). Over the nonperfusion of the choriocapillaris and the ERG and EOG were
course of several weeks, the acute lesions first grow in size and extinguished in one patient with RPC.115 Choroidal ischemia
then evolve to inactive pigmented chorioretinal scars. However, from an unknown trigger is the most widely accepted theory for
2058 all the patients demonstrate a recurrent, recalcitrant course that both APMPPE and SPC and may also be involved in RPC.
Acute Posterior Multifocal Placoid Pigment Epitheliopathy, Serpiginous Choroiditis, and Relentless Placoid Chorioretinitis

CHAPTER 157
b
a

c d

FIGURE 157.13. (a) Fundus photograph demonstrating the acute


presentation of RPC with multiple deep retinal lesions. (b) Within
weeks, new lesions appeared and growth of old lesions was apparent.
(c and d) Photos show light pigmentation of the outer retinal scar
around the fovea. (e) Months later, there is subretinal fibrosis and
atrophic scarring throughout the fundus.

2059
RETINA AND VITREOUS

MANAGEMENT/THERAPY CONCLUSION
With only limited experience to date, the best management for APMPPE, SPC, and RPC all represent idiopathic outer retinal
RPC patients is not known. Corticosteroids have been used and choroidal inflammatory processes. They are usually clini-
with rapid resolution of active lesions and improvement in cally distinguishable based on specific characteristics including
vision. Yet, it is difficult to conclude that steroids affected the age of onset, course, visual outcome, systemic associations,
long-term outcome, given the natural history of spontaneous and recurrence patterns. Given the clinical and angiographic
healing of individual lesions. Antiviral agents also have been similarities among these entities, they may share a common
tried in a small number of patients but without obvious benefit immune dysregulation.
to date. Other immunosuppressive agents, such as cyclosporine,
have also been used,115 but recurrence often is seen after ACKNOWLEDGMENTS
immunosuppression is tapered.
Charles J Bock Jr, MD assisted with the earlier version of the chapter
on SPC.
DIFFERENTIAL DIAGNOSIS
The present work was supported in part by an unrestricted grant from
See sections on APMPPE and/or Serpiginous Choroiditis. Research to Prevent Blindness, Inc, New York City.

REFERENCES
SECTION 10

1. Gass JDM: Acute posterior multifocal 15. Annesley WH, Tomer TL, Sheilds JA: 27. Holt WS, Regan CDJ, Trempe C: Acute
placoid pigment epitheliopathy. Arch Multifocal placoid pigment epitheliopathy. posterior multifocal placoid pigment
Ophthalmol 1968; 80:177. Am J Ophthalmol 1973; 76:511. epitheliopathy. Am J Ophthalmol 1976;
2. Bonnin P, Lavat P: In connection with a 16. Deutman AF, Lion F: Choriocapillaris 81:403.
case of Gass placoid epitheliopathy in nonperfusion in acute multifocal placoid 28. Gass JDM: Inflammatory diseases of the
1860. Bull Soc Ophthalmol Fr 1983; 83:49. pigment epitheliopathy. Am J Ophthalmol retina and choroid. In: Stereoscopic atlas of
3. Gass JDM: Acute posterior multifocal 1977; 84:652. macular diseases: diagnosis and treatment.
placoid pigment epitheliopathy: 17. Schneider U, Inhoffen W, Gelisken F: 3rd edn. St Louis: CV Mosby; 1987.
a long-term follow-up study. In: Fine SL, Indocyanine green angiography in a case of 29. Isashiki M, Koide H, Yamashita T, et al:
ed. Management of retinal vascular and unilateral recurrent posterior acute Acute posterior multifocal placoid pigment
macular disorders. Baltimore: Williams & multifocal placoid pigment epitheliopathy. epitheliopathy associated with diffuse
Wilkins; 1983. Acta Ophthalmol Scand 2003; 81:71. retinal vasculitis and late haemorrhagic
4. Lewis RA, Martonyi CL: Acute posterior 18. Dhaliwal RS, Maguire AM, Flower RW, macular detachment. Br J Ophthalmol
multifocal placoid pigment epitheliopathy: a et al: Acute posterior multifocal placoid 1986; 70:255.
recurrence. Arch Ophthalmol 1975; 93:235. pigment epitheliopathy. An indocyanine 30. De Souza S, Aslanides IM, Altomare F:
5. Fishman GA, Rabb MF, Kaplan J: Acute green angiographic study. Retina 1993; Acute posterior multifocal placoid pigment
posterior multifocal placoid pigment 13:317. epitheliopathy associated with retinal
epitheliopathy. Arch Ophthalmol 1974; 19. Howe LJ, Woon H, Graham EM, et al: vasculitis, neovascularization, and
92:173. Choroidal hypoperfusion in acute posterior subhyaloid hemorrhage. Can J Ophthalmol
6. Kirkman TH, Ffytche TJ, Sanders MD: multifocal placoid pigment epitheliopathy. 1999; 34:343.
Placoid pigment epitheliopathy with retinal An indocyanine green angiography study. 31. Abu El-Asrar AM, Aljazairy AH: Acute
vasculitis and papillitis. Br J Ophthalmol Ophthalmol 1995; 102:790. posterior multifocal placoid pigment
1972; 56:875 20. Van Liefferinge T, Sallet G, De Laey JJ: epitheliopathy with retinal vasculitis and
7. Bohlender T, Weindler J, Ratzkova A: Indocyanine green angiography in cases of papillitis. Eye 2002; 16:642.
Indocyanine green angiography in acute inflammatory chorioretinopathy. Bull Soc 32. Hector RE: Acute posterior multifocal
posterior multifocal placoid pigment Belge Ophthalmol 1995; 257:73. placoid pigment epitheliopathy. Am J
epitheliopathy. Klin Monatsbl Augenheilkd 21. Yuzawa M, Kawamura A, Matsui M: Ophthalmol 1978; 86:424.
1998; 212:170. Indocyanine green videoangiographic 33. Allee SD, Marks SJ: Acute posterior
8. Lyness AL, Bird AC: Recurrences of acute findings in acute posterior multifocal multifocal placoid pigment epitheliopathy
posterior multifocal pigment epitheliopathy. placoid pigment epitheliopathy. Acta with bilateral central retinal vein occlusion.
Am J Ophthalmol 1984; 98:203. Ophthalmol 1994; 72:128. Am J Ophthalmol 1998; 126:309.
9. Damato BE, Nanjiani M, Foulds WS: Acute 22. Cimino L, Auer C, Herbort CP: Sensitivity 34. Jacklin HN: Acute posterior multifocal
posterior multifocal placoid pigment of indocyanine green angiography for the placoid pigment epitheliopathy and
epitheliopathy: a follow up study. Trans follow-up of active inflammatory thyroiditis. Arch Ophthalmol 1977; 95:995.
Ophthalmol Soc UK 1983; 103:517. choriocapillaropathies. Ocul Immunol 35. Oh KT, Park DW: Acute posterior multifocal
10. Brezin AP, Massin-Korobelnik P, Boudin M, Inflamm 2000; 8:275. placoid pigment epitheliopathy with corneal
et al: Acute posterior multifocal placoid 23. Garg S, Jampol LM: Macular serous stromal infiltrates. Am J Ophthalmol 1998;
pigment epitheliopathy after hepatitis B detachment in acute posterior multifocal 125:556.
vaccine. Arch Ophthalmol 1995; 113:297. placoid pigment epitheliopathy. Retina 36. Bird AC, Hamilton AM: Placoid pigment
11. Lowder CY, Foster RE, Gordon SM, et al: 2004; 24:650. epitheliopathy presenting with bilateral
Acute posterior multifocal placoid pigment 24. Scheufele TA, Witkin AJ, Schocket LS, et al: serous retinal detachments. Br J
epitheliopathy after acute group A Photoreceptor atrophy in acute posterior Ophthalmol 1972; 56:881.
streptococcal infection. Am J Ophthalmol multifocal placoid pigment epitheliopathy 37. Wright BE, Bird AC, Hamilton AM:
1996; 122:115. demonstrated by optical coherence Placoid pigment epitheliopathy and
12. Pagliarini S, Piguet B, Ffytche TJ, et al: tomography. Retina 2005; 25:1109. Harada’s disease. Br J Ophthalmol 1978;
Foveal involvement and lack of visual 25. Lofoco G, Ciucci F, Bardocci A, et al: 62:609.
recovery in APMPPE associated with Optical coherence tomography findings in a 38. Savino PJ, Weinberg RJ, Yassin JG, et al:
uncommon features. Eye 1995; 9:42. case of acute multifocal posterior placoid Diverse manifestations of acute posterior
13. Vianna R, van Egmond J, Priem H, et al: pigment epitheliopathy (AMPPPE). Euro J multifocal placoid pigment epitheliopathy.
Natural history and visual outcome in Ophthalmol 2005; 15:143. Am J Ophthalmol 1974; 77:659.
patients with APMPPE. Bull Soc Belge 26. Cruz-Villegas V, Puliafito CA, Fujimoto JG: 39. Sigelman J, Behrens M, Hilal S: Acute
Ophthalmol 1993; 248:73. Retinal vascular diseases. In: Schuman JS, posterior multifocal placoid pigment
14. Quillen DA, Davis JB, Gottlieb JL, et al: The Puliafito CA, Fujimoto JG, eds. Optical epitheliopathy associated with cerebral
white dot syndromes. Am J Ophthalmol coherence tomography of ocular diseases. vasculitis and homonymous hemianopia.
2060 2004; 137:538. Thorofare, NJ: Slack incorporated; 2004. Am J Ophthalmol 1979; 88:919.
Acute Posterior Multifocal Placoid Pigment Epitheliopathy, Serpiginous Choroiditis, and Relentless Placoid Chorioretinitis

40. Bullock JD, Thomas ER, Fletcher RL: adenovirus type 5 infection. Am J 78. Jampol LM: The retina: inflammatory
Cerebrospinal fluid abnormalities in acute Ophthalmol 1975; 80:1003. diseases. In: Miller S, ed. Clinical
posterior multifocal placoid pigment 59. Thomson SPS, Roxburgh STD: Acute ophthalmology. Bristol: Wright; 1987:186.
epitheliopathy. Am J Ophthalmol 1977; 84:45. posterior multifocal placoid pigment 79. Laatikainen L, Erkkila H: A follow-up study
41. Fishman GA, Baskin M, Jednick N: Spinal epitheliopathy associated with adenovirus on serpiginous choroiditis. Acta Ophthalmol
fluid pleocytosis in acute posterior infection. Eye 2003; 17:542. 1981; 59:707.
multifocal placoid pigment epitheliopathy. 60. Mathura JR, Jampol LM, Daily MJ: 80. Laatikainen L, Erkkila H: Serpiginous
Ann Ophthalmol 1977; 9:33. Multifocal choroiditis and acute posterior choroiditis. Br J Ophthalmol 1974; 58:777.
42. Comu S, Verstraeten T, Rinkoff JS, et al: multifocal placoid pigment epitheliopathy 81. Wu JS, Lewis H, Fine SL, et al:
Neurological manifestations of acute occurring in the same patient. Arch Clinicopathologic findings in a patient
posterior multifocal placoid pigment Ophthalmol 2004; 122:1881. with serpiginous choroiditis and treated
epitheliopathy. Stroke 1996; 27:996. 61. Wolf MD, Folk JC, Panknen CA, et al: choroidal neovascularization. Retina 1989;
43. Bewermeyer H, Nelles G, Huber M, et al: HLA-B7 and HLA-DR2 antigens and acute 9:292.
Pontine infarction in acute posterior posterior multifocal placoid pigment 82. Baarsma GS, Deutman AF: Serpiginous
multifocal placoid pigment epitheliopathy. epitheliopathy. Arch Ophthalmol 1990; (geographic) choroiditis. Doc Ophthalmol
J Neurol 1993; 241:22. 108:698. 1976; 40:269.
44. Althaus C, Unsold R, Figge C, et al: 62. Kim RY, Holz FG, Gregor Z, et al: Recurrent 83. Chisholm IH, Gass JDM, Hutton WL: The
Cerebral complications in acute posterior acute multifocal placoid pigment late stage of serpiginous (geographic)
multifocal placoid pigment epitheliopathy. epitheliopathy in two cousins. Am J choroiditis. Am J Ophthalmol 1976; 82:343.
Ger J Ophthalmol 1993; 2:150. Ophthalmol 1995; 119:660. 84. Fujisawa C, Fujiwara H, Hasegawa E, et al:

CHAPTER 157
45. Smith CH, Savino PJ, Beck RW, et al: 63. Jampol LM, Becker KG: White spot The cases of serpiginous choroiditis
Acute posterior multifocal placoid pigment syndromes of the retina: a hypothesis [Japanese, English abstract]. Nippon
epitheliopathy and cerebral vasculitis. Arch based on the common genetic hypothesis Ganka Gakkai Zasshi 1978; 82:135.
Neurol 1983; 40:48. of autoimmune/inflammatory disease. Am J 85. Deutman AF: Choriocapillaris filling
46. Ryan SJ, Maumenee AE: Acute posterior Ophthalmol 2003; 135:376. patterns in health and disease. Trans
multifocal placoid pigment epitheliopathy. 64. Hayreh SS: Segmental nature of the Ophthalmol Soc UK 1968; 100:553.
Am J Ophthalmol 1972; 74:1066. choroidal vasculature. Br J Ophthalmol 86. Fitzgerald PJ, Robertson DM: Acute
47. Jenkins RB, Savino PJ, Pilkerton AR: 1975; 59:631. posterior multifocal placoid pigment
Placoid pigment epitheliopathy with 65. Schatz H, Maumenee AE, Patz A: epitheliopathy. Arch Ophthalmol 1973;
swelling of the optic discs. Arch Neurol Geographic helicoid peripapillary 89:373.
1973; 29:204. choroidopathy: clinical presentation and 87. Alvi NP, Fishman GA: Granulomatous
48. Clearkin LG, Hung SO: Acute posterior fluorescein angiographic findings. Trans Am uveitis presenting with acute posterior
multifocal placoid pigment epitheliopathy Acad Ophthalmol Otolaryngol 1974; 78:747. multifocal placoid pigment epitheliopathy.
associated with transient hearing loss. 66. Mansour AM, Jampol LM, Packo KH, Doc Ophthalmol 1995; 89:347.
Trans Ophthalmol Soc UK 1983; 103:562. Hrisomalos NF: Macular serpiginous 88. Dick DJ, Newman PK, Richardson J, et al:
49. Wilson CA, Choromokos EA, Sheppard R: choroiditis. Retina 1988; 8:125. Acute posterior multifocal placoid pigment
Acute posterior multifocal placoid pigment 67. King DG, Grizzard WS, Sever RJ, Espinoza epitheliopathy and sarcoidosis. Br J
epitheliopathy and cerebral vasculitis. Arch L: Serpiginous choroidopathy associated Ophthalmol 1988; 72:74.
Ophthalmol 1988; 107:796. with elevated factor VIII-von Willebrand 89. Ciulla TA, Gragoudas ES: Serpiginous
50. Van Buskirk EB, Lessell S, Friedman E: factor antigen. Retina 1990; 10:97. choroiditis. Int Ophthalmol Clin 1996; 36:135.
Pigment epitheliopathy and erythema 68. Jampol LM, Orth D, Daily MJ, Rabb MF: 90. Giovanni A, Ripa E, Scassellati-Sforzolini B,
nodosum. Arch Ophthalmol 1972; 85:369. Subretinal neovascularization with et al: Indocyanine green angiography in
51. Deutman AF, Oosterhuis JA, BoenTan TN, geographic (serpiginous) choroiditis. Am J serpiginous choroidopathy. Eur J
et al: Acute posterior multifocal placoid Ophthalmol 1979; 88:683. Ophthalmol 1996; 6:299.
pigment epitheliopathy: pigment 69. Gass JDM: Specific choroidal diseases 91. Masi RJ, O’Connor R, Kimura SJ: Anterior
epitheliopathy or choriocapillaritis. Br J causing disciform macular detachment. In: uveitis in geographic or serpiginous
Ophthalmol 1972; 56:863. Gass JDM, ed. Stereoscopic atlas of choroiditis. Am J Ophthalmol 1978; 86:228.
52. Parmeggiani F, Costagliola C, D’Angelo S, macular diseases: diagnosis and treatment. 92. Nussenblatt RB, Whitcup SM, Palestine
et al: Clear cell renal cell carcinoma 3rd edn. St Louis: CV Mosby; 1987:136. AG: Serpiginous choroidopathy. In: Uveitis:
associated with bilateral atypical acute 70. Green WR: The uveal tract. In: Spencer fundamentals and clinical practice. 2nd
posterior multifocal placoid pigment WH, ed. Ophthalmic pathology: an atlas edn. St Louis: CV Mosby; 1996:364–370.
epitheliopathy. Oncology 2004; 66:502 and textbook. 3rd edn. Philadelphia: WB 93. Laatikainen L, Erkkila H: Subretinal and
53. Anderson K, Patel KR, Webb L, et al: Saunders; 1986:1994. disc neovascularization in serpiginous
Acute posterior multifocal placoid pigment 71. Schlaegel TF Jr: Metastatic nonsuppurative choroiditis. Br J Ophthalmol 1982; 66:326.
epitheliopathy associated with pulmonary uveitis. In:Schlaegel TF Jr, ed. Essentials of 94. Wojno T, Meredith TA: Unusual findings in
tuberculosis [letter]. Br J Ophthalmol 1996; uveitis. Boston: Little Brown; 1969:77. serpiginous choroiditis. Am J Ophthalmol
80:186. 72. Richardson RR, Cooper IS, Smith JL: 1982; 94:650.
54. Di Crecchio L, Parodi MB, Saviano S, et al: Serpiginous choroiditis and unilateral 95. Erkkila H, Laatikainen L, Jokinen E:
Acute posterior multifocal placoid pigment extrapyramidal dystonia. Ann Ophthalmol Immunological studies on serpiginous
epitheliopathy and ulcerative colitis: a 1981; 13:15. choroiditis. Graefes Arch Clin Exp
possible association. Acta Ophthalmol 73. Schatz H, McDonald HR: Geographic Ophthalmol 1982; 219:131.
Scand 2001; 79:319. helicoid peripapillary choroidopathy 96. Friberg TR: Serpiginous choroiditis with
55. Chiquet C, Lumbroso L, Denis P, et al: (serpiginous choroiditis). In: Ryan SJ, branch vein occlusion and bilateral
Acute posterior multifocal placoid pigment Schachat AP, Murphy RB, Patz A, eds. periphlebitis. Arch Ophthalmol 1988;
epitheliopathy associated with Wegener’s Retina. St Louis: CV Mosby; 1989:705. 106:585.
granulomatosis. Retina 1999; 19:309. 74. Hardy RA, Schatz H: Macular geographic 97. Huuskonen MS, Tilikainen A, Alanko K:
56. Hsu CT, Harlan JB, Goldberg MF, et al: helicoid choroidopathy. Arch Ophthalmol HLA-B18 antigens and protection from
Acute posterior multifocal placoid pigment 1987; 105:1237. pulmonary fibrosis in asbestos workers.
epitheliopathy associated with a systemic 75. Sveinsson K: Helicoidal peripapillary Br J Dis Chest 1979; 73:253.
necrotizing vasculitis. Retina 2003; 23:64. chorioretinal degeneration. Acta 98. Edelston C, Stanford MR, Graham EM:
57. Matsuo T, Horikoshi T, Nagai C: Acute Ophthalmol 1979; 57:69. Serpiginous choroiditis: an unusual
posterior multifocal placoid pigment 76. Weiss H, Annesley WH Jr, Shields JA, et al: presentation of ocular sarcoidosis.
epitheliopathy and scleritis in a patient with The clinical course of serpiginous Br J Ophthalmol 1994; 78:70.
pANCA-positive systemic vasculitis. Am J choroidopathy. Am J Ophthalmol 1979; 99. Mulder CJJ, Pena AS, Jansen J,
Ophthalmol 2002; 133:566. 87:133. Oosterhuis JA: Celiac disease and
58. Azar P, Gohd RS, Waltman D, et al: Acute 77. Blumenkranz MS, Gass JDM, Clarkson JG: geographic (serpiginous) choroidopathy
posterior multifocal placoid pigment Atypical serpiginous choroiditis. Arch with occurrence of thrombocytopenic
epitheliopathy associated with an Ophthalmol 1982; 100:1773. purpura. Arch Intern Med 1983; 143:842. 2061
RETINA AND VITREOUS

100. Maumenee AE: Clinical entities in uveitis: in serpiginous choroiditis. Ophthalmology choroidopathy in North India. Am J
an approach to the study of intraocular 1991; 98:951. Ophthalmol 2002; 134:47.
inflammation. XXVI Edward Jackson 109. Akpek EK, Jabs DA, Tesseler HH, et al: 117. Bouchenaki N, Cimino L, Auer C, et al:
Memorial Lecture. Am J Ophthalmol Successful treatment of serpiginous Assessment and classification of choroidal
1970; 69:1. choroiditis with alkylating agents. vasculitis in posterior uveitis using
101. Broekhuyse RM, Van Herck M, Pinckers Ophthalmology 2002; 109:1506. indocyanine green angiography. Klin
AJ, et al: Immune responsiveness to retinal 110. Secchi AG, Tognon S, Maselli C: Monatsbl Augenheilkd 2002; 219:243.
S-antigen and opsin in serpiginous Cyclosporine-A in the treatment of 118. García-Sáenz MC, Gili Manzanaro P,
choroiditis and other retinal diseases. Doc serpiginous choroiditis. Int Ophthalmol Bañuelos J, et al: Indocyanine green
Ophthalmol 1988; 69:83. 1990; 14:395. angiography in chorioretinal inflammatory
102. Gass JDM: Serpiginous choroiditis. In: 111. Leznoff A, Shea M, Binkley KE, et al: diseases. Arch Soc Esp Oftalmol 2003;
Gass JDM, ed. Stereoscopic atlas of Cyclosporine in the treatment of 78:675.
macular diseases: diagnosis and treatment. nonmicrobial inflammatory ophthalmic 119. Giovannini A, Mariotti C, Ripa E, et al:
4th edn. St Louis: Mosby-Year Book; disease. Can J Ophthalmol 1992; 27:302. Indocyanine green angiography findings in
1997:158–165. 112. Sobaci G, Bayraktar Z, Bayer A: Interferon serpiginous choroidopathy. Br J
103. Akpek EK, Chan CC, Shen D, et al: Lack of alpha-2a treatment for serpiginous Ophthalmol 1996; 80:536.
herpes virus DNA in choroidal tissues of a choroiditis. Ocular Immunol Inflam 2005; 120. Salati C, Pantelis V, Lafaut BA, et al:
patient with Serpiginous choroiditis. 13:59. A 8 months indocyanine green
Ophthalmology 2004; 111:2071. 113. Navajas EV, Costa RA, Farah ME, et al: angiographic follow-up of a patient with
104. Neetens A, Burvenich H: Presumed Indocyanine green-mediated serpiginous choroidopathy. Bull Soc Belge
inflammatory maculopathies. Trans photothrombosis combined with intravitreal Ophtalmol 1997; 265:29.
SECTION 10

Ophthalmol Soc UK 1978; 98:160. triamcinolone for the treatment of choroidal 121. Squirrell DM, Bhola RM, Talbot JF:
105. Pham-Duy T, Miszalok V: Choroiditis neovascularization in serpiginous Indocyanine green angiographic findings in
geographica. Klin Monatsbl Augenheilkd choroiditis. Eye 2003; 17:563. serpiginous choroidopathy: evidence of a
1983; 183:278. 114. Duke-Elder S, Perkins E: Inflammations of widespread choriocapillaris defect of the
106. Araujo AA, Wells AP, Dick AD, et al: Early the uveal tract: uveitis. In: Duke-Elder S, peripapillary area and posterior pole. Eye
treatment with cyclosporine in serpiginous ed. System of ophthalmology. Diseases of 2001; 15:336.
choroidopathy maintains remission and the uveal tract. St Louis: CV Mosby; 122. Vabres G: Study of an atypical unilateral
good visual outcome. Br J Ophthalmol 1966:246. choroidopathy of the posterior pole with
2000; 84:979. 115. Jones EB, Jampol LM, Yanuzzi LA, et al: centrifugal serpiginous development
107. Hooper PL, Kaplan HJ: Triple agent Relentless placoid chorioretinitis: a new (clinical, angiofluorographic and functional
immunosuppression in serpiginous entity or an unusual variant of serpiginous aspects). Ann Ocul (Paris) 1972; 205:927.
choroidopathy. Ophthalmology 1990; chorioretinitis? Arch Ophthalmol 2000;
97(Suppl):109. 118:931.
108. Nussenblatt RB: Discussion of Hooper PL, 116. Gupta V, Agarwal A, Gupta A, et al: Clinical
Kaplan HJ: triple agent immunosuppression characteristics of serpiginous

2062
CHAPTER

158 Multiple Evanescent White Dot Syndrome


Howard F. Fine, Antonio P. Ciardella, and John A. Sorenson

INTRODUCTION DEMOGRAPHICS
The multiple evanescent white dot syndrome (MEWDS) was Most patients are myopic7 females in roughly the third decade
first described in 1984 by Jampol and associates.1 Within the of life. The reported age range of cases is 108 to 679 years old.
same year, Takeda and co-workers independently reported in the MEWDS does not appear to have a racial predilection.
Japanese literature, the clinical findings of four patients with
presentations indistinguishable from MEWDS; they proposed CLINICAL FINDINGS
the term acute disseminated retinal pigment epitheliopathy.2
The subjective, demographic, clinical, fluorescein angiographic, The presenting visual acuity ranges from 20/20 to 20/400.
and electrophysiological findings of MEWDS represent a dis- Some patients may have an afferent papillary defect. The ante-
tinct clinical entity. rior segment examination is normal except for the occasional
presence of mild iritis. The most striking findings on posterior
SUBJECTIVE COMPLAINTS slit-lamp biomicroscopic examinations are numerous small
white spots, ~100–200 mm in size and located at the level of the
The usual presenting symptom is the acute onset of decreased retinal pigment epithelium (RPE) or deep retina. They are most
or blurred vision often accompanied by dark or black spots in prominently seen encircling the posterior pole and in the
the periphery. Virtually all patients also note photopsia, perifoveal region, but the fovea is spared (Figs 158.1 to 158.3).
described as flickering or shimmering lights. The photopsia The spots are discrete and are not associated with overlying
sometimes seems to originate from temporal scotomas. Com- exudative changes. They are sometimes migratory, clearing in
monly, patients describe a preceding ‘flu-like’ illness.3,4 Rarely, one area while appearing in another, over a period of several
patients report headaches5 accompanying the visual symptoms days. If spots are large in size, the center of the spot may resolve
or ocular discomfort.6 before the periphery.10 Usually, the lesions resolve entirely,

FIGURE 158.1. (a) Color photograph of the


fundus in a patient with MEWDS. Note the
scattered deep retinal spots in the posterior
pole. (b) Color photograph of the nasal
posterior retina of the same patient. Additional
spots are evident extending toward the
midperipheral retina. (c) Early fluorescein
angiogram reveals multiple pinpoint dots, most
of which correspond to the white lesions.
(d) The late-stage angiogram reveals no
significant leakage associated with the
a b punctate hyperfluorescent dots. Disk edema is
evident. The left eye was completely normal
clinically and angiographically.

c d

2063
RETINA AND VITREOUS

FIGURE 158.2. (a) Color photograph of a


patient with MEWDS reveals scattered lesions
in the posterior pole, most prominently evident
in the inferior and superior nasal juxtapapillary
regions. There were a few cells in the posterior
vitreous. (b) Color photograph of the same
patient 4 days later. Note the disappearance of
the white spots in the nasal juxtapapillary
regions and the appearance of new spots in the
temporal macula. No vitreous cells were noted
at this time.
a b

FIGURE 158.3. (a) Color photograph of a


patient with MEWDS. Note the prominent deep
retinal or retinal pigment epithelial lesions, or
both. These are more conspicuously evident
SECTION 10

than in the patient in Figure 158.1. (b) A


monochromatic photograph of the same
patient highlights the white spots in the fundus.
(c) Early fluorescein angiogram in this patient
reveals the characteristic hyperfluorescent dots
in the region of the pigment epithelium.
(d) Late-stage fluorescein angiogram reveals
staining of the RPE and outer retina. In this
patient, there has been alteration of the
b posterior blood–retinal barrier from extensive
a neuroretinitis involving the pigment epithelium.

c d

leaving behind a characteristic foveal RPE granularity in most


cases (Fig. 158.4). Other findings sometimes include a few cells
in the posterior vitreous,11 mild blurring of the disk margin, and
isolated areas of perivascular sheathing. When seen, the cellular
reaction in the posterior vitreous is mild and transient in the
early stages of the disease.

ATYPICAL MANIFESTATIONS
While atypical, cotton wool spots have been noted in
MEWDS.12 Prior to the typical appearance of macular spots,
MEWDS rarely may first manifest with a large peripapillary
outer retinal lesion13 or geographic circumpapillary discolora-
tion.14 MEWDS can result in midperipheral, peripapillary, and
even macular chorioretinal scarring, typically in patients with
recurrent or bilateral disease. One report describes a patient
diagnosed with MEWDS who developed transient reddish-
brown spots after resolution of the acute MEWDS lesions.15
MEWDS can occasionally be seen with multiple, large lesions, FIGURE 158.4. Color photograph of a patient with MEWDS. Note the
up to 1000 mm in size, and granular, pigmented, placoid lesions granular appearance of the macula, which is characteristic of this
in association with a panuveitis.16 Patients with recurrent disease and is exceedingly prominent in this patient.
2064
Multiple Evanescent White Dot Syndrome

FIGURE 158.5. (a) Humphrey’s visual field of a


24-year-old woman with MEWDS involving the
left eye 1 week after the onset of symptoms.
The field demonstrates marked visual loss in
the entire visual field, most profound temporally
(giant blind spot). The visual acuity was 20/40.
(b) Visual field of the same patient 4 months
later. The field is now nearly normal, although
the patient still complained of dim vision. The
visual acuity had returned to 20/20 and the
white spots were completely gone. Eight
months after the initial onset of symptoms, this
patient still complained of dim vision in the left
eye.
a b

MEWDS can develop choroidal neovascularization.17,18 This phase of the illness, the Ganzfield electroretinogram (ERG)

CHAPTER 158
may cause some diagnostic confusion with multifocal choroidi- a-wave and early receptor potential amplitudes were profoundly
tis (MFC), especially after the acute phase of the disease has decreased. This suggests that photoreceptors are involved
resolved.19 during active disease. These normalize with resolution of the
disease.26 Additionally, S-cone reductions are more pronounced
VISUAL FIELDS than in the L- or M-cone systems on Ganzfield ERG.27 The
multifocal ERG, which reflects the cone activity of the central
The visual field findings are variable in MEWDS, ranging from retina, demonstrates a supranormal response in acute disease of
normal to a generalized depression (Fig. 158.5). Large blind less than 2 weeks duration, which may become subnormal or
spots are a frequent finding. In his original description, Jampol normalize subsequently. Similar multifocal ERG findings have
noted optic nerve edema clinically in some patients. Arcuate been observed in other deep retinal inflammatory conditions.28
scotomas, cecocentral scotomas, and central depression have also Electrooculography results have also been reported to be mildly
been reported.20 The field loss is sometimes exaggerated when abnormal, suggesting involvement of the RPE during acute
compared with the clinical findings in the retina and at the disk. disease.29 Abnormal foveal densitometry has been reported for a
patient with MEWDS who had normal ERG findings.30
FLUORESCEIN ANGIOGRAPHY
CLINICAL COURSE
The fluorescein angiography findings in MEWDS are charac-
terized by early punctate hyperfluorescent spots, 50–500 mm in Most patients with MEWDS experience a relatively short course
diameter, at the level of the RPE, often corresponding to the with recovery of vision within 3–10 weeks. Usually, the vision
white dots observed clinically.21 Classically, these punctate returns to 20/30 or better, with the majority of patients regain-
spots assume a wreath-shaped pattern. The late-phase fluores- ing 20/20 vision. The white spots also disappear during this
cein angiogram reveals staining in the area of the spots and the time; however, the orange foveal granularity usually persists.
optic nerve head (Figs 158.1c,d and 158.3c,d). Late capillary Some patients will have visual symptoms for much longer
leakage in the perifoveal area and focal areas of vasculitis are periods following resolution of the white spots. Even though
occasionally noted. Capillary leakage of the optic nerve head visual acuity returns, some patients are aware of visual field
can be present. The spots often resolve angiographically within defects, photopsia, or dim vision for extended periods.
4–6 weeks and are thought to represent inflammatory lesions at Bilateral involvement is uncommon31–33 and has been re-
the level of the photoreceptors, RPE, and choroid. ported as late as 4 years after initial presentation in the con-
tralateral eye.34 When both eyes are involved, one eye is usually
INDOCYANINE GREEN ANGIOGRAPHY involved minimally and is asymptomatic. One reported patient
was symptomatic in both eyes simultaneously.35 This patient
On indocyanine green (ICG) angiography, hypofluorescent spots experienced visual loss in the right eye after 7 weeks of vision
are seen (Fig. 158.6), which may be more extensive and may loss in the left eye. Recurrences of MEWDS are uncommon.36
persist longer than spots observed on fluorescein angiography. Choroidal neovascularization is a rare but potential late
Peripapillary hypofluorescence on ICG may account for the sequelae.37,38
enlarged blind spot and other field abnormalities observed
secondary to choroidal hypoperfusion of the optic nerve and PATHOPHYSIOLOGY
peripapillary area.22 In an evidence-based review of chorioretinal
diseases, the use of ICG for the diagnosis and management of MEWDS is a distinct clinical syndrome. Since it has an acute
MEWDS was ‘recommended with moderately strong supporting onset and is sometimes preceded by a ‘flu-like’ illness, a viral
evidence’.23 ICG may be particularly useful in cases with subtle cause seems likely. As Gass proposed for acute zonal occult
clinical findings and absent fluorescein angiographic changes. outer retinopathy, the disease may manifest first at the optic
ICG can also be used to follow the clinical course.24 nerve margin due to the lack of surrounding neuroepi-
thelium.39,40
ELECTROPHYSIOLOGY Clinically and angiographically, the disease process involves
the RPE and outer retina. ICG findings suggest that early MEWDS
The electrophysiologic findings were described for three of the involves the RPE and photoreceptors, but with periphlebitis, all
original patients reported to have MEWDS.25 During the acute layers of the retina may be involved.41 The ERG abnormalities
2065
RETINA AND VITREOUS

a b c
SECTION 10

d e f

FIGURE 158.6 (a) Color photograph of a


patient with acute onset MEWDS. Only a few
yellowish spots are visible temporally. (b) The
spots are seen better with a red-free light.
(c) Midphase fluorescein angiography
demonstrates a few hyperfluorescent spots and
staining of the disc margin. (d) Late-phase
fluoroscein angiography shows marked staining
of the optic nerve. (e) Late-phase indocyanine
green (ICG) study shows a hypofluorescent ring
around the optic nerve. (f and g) There is also
evidence of multiple hypofluorescent spots at
the posterior pole and in the midperiphery.
g h (h) Goldmann’s visual field demonstrates an
enlarged blind spot. (i) Late-phase ICG study
4 months later demonstrates resolution of the
hypofluorescent ring aroung the optic nerve.
(j) Goldmann’s visual field on the same day
shows resolution of the enlarged blind spot.
From Yannuzzi LA, Flower WR, Slakter JS (eds):
Indocyanine Green Angiography. St Louis, Mosby Year
Book, 1997.

i j

suggest a metabolic disturbance of the RPE–photoreceptor DIFFERENTIAL DIAGNOSIS


complex.
One case has been described with increased serum levels of MEWDS must be differentiated from other inflammatory dis-
total IgG and IgM during the acute phase.42 MEWDS has been orders of the RPE, choroid, or retina. The primary differential
reported in association with recent varicella infection,43 diagnosis includes the white dot syndromes: MFC, punctate
increased CSF protein levels,44 and vaccinations for hepatitis inner choroidopathy (PIC), acute idiopathic blindspot enlarge-
A45 and B.46 One study documented a 3.7-fold increase in ment syndrome (AIBSE), acute posterior multifocal placoid
association with HLA-B51.47 Hormonal factors play a role in pigment epitheliopathy (APMPPE), acute retinal pigment epi-
pathogenesis, owing to the gender imbalance and high rate of theliitis (ARPE), birdshot retinochoroidopathy, toxoplasmosis,
2066 oral contraception usage.48 diffuse unilateral subacute neuroretinitis (DUSN), acute
Multiple Evanescent White Dot Syndrome

macular neuroretinopathy (AMN), and primary intraocular early hypofluorescent spots which stain late on fluorescein
lymphoma. In most instances, the diagnosis of MEWDS can be angiography.61
made confidently on clinical and angiographic examination,
and other disorders can be excluded.
ACUTE IDIOPATHIC BLIND SPOT
ENLARGEMENT
MFC AND PIC Fletcher and colleagues described a syndrome occurring
Several papers have described a group of patients that can primarily in young females consisting of blind spot enlargement
probably best be lumped together as cases of MFC. They include without optic disk edema.62 The acute idiopathic blind spot
the following: PIC,49 MFC and panuveitis,50 MFC associated enlargement (AIBSE) syndrome has features suggesting retinal
with subretinal fibrosis,51 and recurrent MFC. The patients dysfunction as its cause, including the presence of the scotoma,
described in these papers are similar. As in MEWDS, they are steep borders of the scotoma, and prolonged recovery time with
usually young females who present with acute visual loss in one photostress testing. Hamed and colleagues suggested that
eye and spots in the fundus. Gass and Callanan posited that patients with AIBSE represent a subset of those with MEWDS,
MEWDS, MFC, PIC, and AIBES are, in fact, manifestations of presenting for examination after the retinal spots have faded.63
a single disease spectrum.52 More recently, Singh and associates suggested that MEWDS is
PIC was described by Watzke and associates in 1984. Ten one of several conditions that may be characterized by blind
moderately myopic women (aged 21–37 years) presented with spot enlargement.64 Some cases of AIBSE, but not all, may be

CHAPTER 158
blurred vision, paracentral scotomas, or light flashes. The due to MEWDS. Volpe and colleagues reviewed 27 cases of
characteristic fundus lesions are small gray-yellow spots AIBSE and suggest that the primary factor that distinguishes
~100–200 mm in diameter at the level of the RPE and inner AIBSE from MEWDS is that the visual field abnormalities do
choroids scattered throughout the posterior pole. Many lesions not resolve in AIBSE.65 Patients with AIBSE can present with
had an overlying serous detachment. Neither vitreous nor photopsia; visual field defects; and have abnormal funduscopic,
anterior chamber inflammation is present. The fluorescein angiographic, and electroretinographic findings.
angiogram reveals early hyperfluorescence and late staining or
leakage into an overlying sensory retinal detachment. Eight of
10 patients had bilateral involvement, although symptoms were ACUTE POSTERIOR MULTIFOCAL PLACOID
usually unilateral. The lesions developed into atrophic scars. PIGMENT EPITHELIOPATHY
Some scars became pigmented over time. Four of 10 patients APMPPE can also present with rapid loss of vision in a young
eventually experienced subretinal neovascularization in patient.66 However, this condition is usually bilateral and the
association with perifoveal scars. Dreyer and Gass described 28 fundus lesions are characteristic. Multiple, flat gray-white
patients with MFC and panuveitis.53 Besides the characteristic lesions are present at the level of the RPE. These lesions are
RPE lesions resembling those seen in the presumed ocular much larger than the spots of MEWDS. The lesions are hypo-
histoplasmosis syndrome, these patients also had vitreous cells fluorescent in the early-stage fluorescein angiogram and stain
and anterior segment inflammation. Many of these patients late. Extensive RPE alterations develop as lesions resolve.
experienced peripapillary or subfoveal subretinal neovas-
cularization. Cantrill and Folk reported five female patients in
whom progressive subretinal fibrosis developed in association ACUTE RETINAL PIGMENT EPITHELIITIS
with MFC.54 Doran and Hamilton55 and Palestine and co- ARPE was described by Krill and Deutman in 1972.67 Chittum
workers56 had previously reported similar cases. Morgan and and Kalina more recently reported the findings of eight patients
Shatz described a similar group of female patients with MFC with ARPE.68 This condition is also characterized by acute loss
who had multiple recurrences.57 of vision in a young patient. The ages of the first nine reported
All these papers describe a MFC that is generally seen in patients ranged from 18 to 45 years.69 Three of the nine
young myopic females. The disorder can be chronic and patients had bilateral involvement. The acute findings have
recurrent. Although often presenting with unilateral symptoms, been described as discrete clusters of dark spots surrounded by
findings are usually bilateral. The RPE and inner choroidal hypopigmented halos. These spots are localized to the peri-
spots vary in size but are generally larger than those seen in foveal region. Fluorescein angiography can reveal blockage of
MEWDS. Papillitis, retinal phlebitis, vitritis, anterior uveitis, choroidal fluorescence from the pigmented center with a
cystoid macular edema, and subretinal neovascularization can surrounding zone of hyperfluorescence corresponding to the
all be part of the disease process. One or more of these hypopigmented halo. Gradual resolution of the fundus findings
manifestations may predominate in a given patient. Following with complete recovery of vision within 7–10 weeks is typical.
resolution of the spots in the acute stage, there is some degree Subtle RPE alterations may remain.
of permanent RPE and choroidal damage, which can be
atrophic, pigmentary, or cicatricial in nature. Since many
disorders may mimic this idiopathic condition, a full medical BIRDSHOT RETINOCHOROIDOPATHY
work-up is needed, especially to rule out treatable infectious or Birdshot retinochoroidopathy or vitiliginous chorioretinitis is
inflammatory conditions such as syphilis or sarcoidosis. characterized by white or depigmented spots at the level of the
A common host susceptibility factor or even a common RPE scattered throughout the fundus.70,71 The older age at
pathogenesis may exist between MEWDS and the MFC presentation, chronic course, bilateral involvement, and signi-
spectrum of disease. Several cases have been observed ficant vitreous involvement all help to easily distinguish this
of patients with MEWDS who subsequently develop MFC,58–60 syndrome from MEWDS.
and several cases are described of patients with a previous
diagnosis of MFC who develop MEWDS. Both MEWDS and
MFC typically affect young myopic women. However, unlike TOXOPLASMOSIS
MEWDS, MFC is usually associated with bilateral involvement, Another cause of retinochoroiditis that must be differentiated
deep choroidal inflammatory (rather than deep retinal/RPE) from MEWDS is toxoplasmosis. Ocular toxoplasmosis can
lesions, prominent vitritis and anterior chamber reaction, and present with multifocal gray-white lesions at the level of the 2067
RETINA AND VITREOUS

deep retina and RPE, with little or no overlying vitreous who initially presented with transient lesions of the posterior
reaction.72 These lesions typically resolve slowly and can recur. pole, simulating MEWDS. Eventually, these patients were
The diagnosis of punctate outer retinal toxoplasmosis is dif- diagnosed with primary intraocular lymphoma due to the
ficult to confirm. The presence of satellite lesions and positive persistence or expansion of retinal lesions or of vitreous cellular
serologic findings for toxoplasmosis can help to make the infiltration.78,79
diagnosis in the appropriate clinical setting.
CONCLUSIONS
DIFFUSE UNILATERAL SUBACUTE In summary, MEWDS is a unilateral, idiopathic, and inflam-
NEURORETINITIS matory disease with a constellation of well-described subjective,
The early stage of DUSN can be characterized by recurrent clinical, fluorescein angiographic, and electrophysiologic
crops of evanescent gray-white lesions at the level of the outer features. It occurs most often as a unilateral phenomenon,
retina and RPE.73 The chronic course of DUSN with eventual predominantly in young, myopic females. The most charac-
development of visual loss, optic atrophy, diffuse RPE de- teristic manifestation is that of outer retinal or superficial
generation, and retinal vessel narrowing easily differentiates pigment epithelial white spots, often 100–200 mm in size, and
this disorder from MEWDS. DUSN is caused by a nematode usually confined to the posterior pole or midretinal areas. These
that can sometimes be visualized in the subretinal space.74 fleeting disturbances are migratory in nature and evident on
fluorescein angiography with a wreath-like distribution of early
SECTION 10

punctate hyperfluorescent spots which stain late. A persistent


ACUTE MACULAR NEURORETINOPATHY orange foveal granularity and mild papillitis are hallmarks of
AMN is another cause of loss of vision and paracentral the disorder. Similarly, there are characteristic electroretino-
scotomas in young patients.75,76 This disorder can be bilateral graphic findings and visual field loss. The field changes exceed
and is characterized by cloverleaf, wedge-shaped, grayish lesions what might be expected on the bases of the clinical findings in
in the macular area, probably in the outer retinal layers. Interes- the retina and at the optic nerve. The natural course is generally
tingly, patients have been reported with AMN and MEWDS in benign with spontaneous disappearance of the lesions and
the same eye, suggesting some similarity in their origins.77 improvement of the visual function. MEWDS is often clinically
differentiated from other categorized intraocular inflammatory
diseases. Although there is considerable information available
PRIMARY INTRAOCULAR LYMPHOMA on the subjective, demographic, clinical, angiographic, and
Primary intraocular lymphoma is well known to masquerade as electrophysiologic features of this disease, a causative agent is
numerous other ocular inflammatory syndromes, and MEWDS still unknown. Further clinical research is needed to identify the
is no exception. Several cases have been reported of patients precise pathogenesis.

REFERENCES
1. Jampol LM, Sieving PA, Pugh D, et al: 10. Huang J, Spaide R: Appearance of brown 18. Oh KT, Christmas NJ, Russell SR: Late
Multiple evanescent white dot syndrome. areas after resolution of the acute phase of recurrence and choroidal neovascularization
I. Clinical findings. Arch Ophthalmol 1984; multiple evanescent white dot syndrome. in multiple evanescent white dot syndrome.
102:671–674. Retina 2004; 24:814–816. Retina 2001; 21:182–184.
2. Takeda M, Kimura S, Tamiya M: Acute 11. Gass JD, Hamed LM: Acute macular 19. Kozielec GF, Wyhinny GJ, Jampol LM:
disseminated retinal pigment neuroretinopathy and multiple evanescent Evolution of distinct chorioretinal scars in
epitheliopathy. Folia Ophthalmol Jpn 1984; white dot syndrome occurring in the same recurrent MEWDS. Retina 2001;
35:2613. patients. Arch Ophthalmol 1989; 21:180–182.
3. Jampol LM, Sieving PA, Pugh D, et al: 107:189–193. 20. Nakao K, Isashiki M: Multiple evanescent
Multiple evanescent white dot syndrome. 12. Dodwell DG, Jampol LM, Rosenberg M, white dot syndrome. Jpn J Ophthalmol
I. Clinical findings. Arch Ophthalmol 1984; et al: Optic nerve involvement associated 1986; 30:376–384.
102:671–674. with the multiple evanescent white-dot 21. Gass JDM: Stereoscopic atlas of macular
4. Slusher MM, Weaver RG: Multiple syndrome. Ophthalmology 1990; diseases: diagnosis and treatment. 3rd edn.
evanescent white dot syndrome. Retina 97:862–868. St Louis: Mosby; 1987:510–511.
1988; 8:132–135. 13. Ray S, Loewenstein J: Atypical 22. Yen MT, Rosenfeld PJ: Persistent
5. Meyer RJ, Jampol LM: Recurrences and manifestation of multiple evanescent white indocyanine green angiographic findings in
bilaterality in the multiple evanescent dot syndrome with large peripapillary multiple evanescent white dot syndrome.
white-dot syndrome. Am J Ophthalmol lesion. Arch Ophthalmol 2003; Ophthalmic Surg Lasers 2001; 32:156–158.
1986; 101:388–389. 121:1794–1796. 23. Stanga PE, Lim JI, Hamilton P: Indocyanine
6. Kimmel AS, Folk JC, Thompson HS, 14. Luttrull JK, Marmor MF, Nanda M: green angiography in chorioretinal
Strnad LS: The multiple evanescent white- Progressive confluent circumpapillary diseases: indications and interpretation: an
dot syndrome with acute blind spot multiple evanescent white-dot syndrome. evidence-based update. Ophthalmology
enlargement. Am J Ophthalmol 1989; Am J Ophthalmol 1999; 128:378–380. 2003; 110:15–21.
107:425–426. 15. Huang J, Spaide R: Appearance of brown 24. Cimino L, Auer C, Herbort CP: Sensitivity
7. Asano T, Kondo M, Kondo N, et al: High areas after resolution of the acute phase of of indocyanine green angiography for the
prevalence of myopia in Japanese patients multiple evanescent white dot syndrome. follow-up of active inflammatory
with multiple evanescent white dot Retina 2004; 24:814–816. choriocapillaropathies. Ocul Immunol
syndrome. Jpn J Ophthalmol 2004; 16. Khurana RN, Albini T, Dea MK, et al: Inflamm 2000; 8:275–283.
48:486–489. Atypical presentation of multiple 25. Sieving PA, Fishman GA, Jampol LM,
8. Olitsky SE: Multiple evanescent evanescent white dot syndrome involving Pugh D: Multiple evanescent white dot
white-dot syndrome in a 10-year-old child. granular lesions of varying size. Am J syndrome. II. Electrophysiology of the
J Pediatr Ophthalmol Strabismus 1998; Ophthalmol 2005; 139:935–937. photoreceptors during retinal pigment
35:288–289. 17. Wyhinny GJ, Jackson JL, Jampol LM, epithelial disease. Arch Ophthalmol 1984;
9. Lim JI, Kokame GT, Douglas JP: Multiple Caro NC: Subretinal neovascularization 102:675–679.
evanescent white dot syndrome in older following multiple evanescent white-dot 26. Slusher MM, Weaver RG: Multiple
patients. Am J Ophthalmol 1999; syndrome. Arch Ophthalmol 1990; evanescent white dot syndrome. Retina
2068 127:725–728. 108:1384–1385. 1988; 8:132–135.
Multiple Evanescent White Dot Syndrome

27. Yamamoto S, Hayashi M, Tsuruoka M, et al: 44. McCollum CJ, Kimble JA: Peripapillary 61. Bryan RG, Freund KB, Yannuzzi LA, et al:
S-cone electroretinograms in multiple subretinal neovascularization associated Multiple evanescent white dot syndrome in
evanescent white dot syndrome. Doc with multiple evanescent white-dot patients with multifocal choroiditis. Retina
Ophthalmol 2003; 106:117–120. syndrome. Arch Ophthalmol 1992; 2002; 22:317–322.
28. Feigl B, Haas A, El-Shabrawi Y: Multifocal 110:13–14. 62. Fletcher WA, Imes RK, Goodman D,
ERG in multiple evanescent white dot 45. Fine L, Fine A, Cunningham ET Jr: Multiple Hoyt WF: Acute idiopathic blind spot
syndrome. Graefes Arch Clin Exp evanescent white dot syndrome following enlargement. A big blind spot syndrome
Ophthalmol 2002; 240:615–621. hepatitis A vaccination. Arch Ophthalmol: without optic disc edema. Arch Ophthalmol
29. Laatikainen L, Immonen I: Multiple 2001; 119:1856–1858. 1988; 106:44–49.
evanescent white dot syndrome. Graefes 46. Baglivo E, Safran AB, Borruat FX: Multiple 63. Hamed LA, Schatz NJ, Glaser JS, Gass JD:
Arch Clin Exp Ophthalmol 1988; evanescent white dot syndrome after Acute idiopathic blind spot enlargement
226:37–40. hepatitis B vaccine. Am J Ophthalmol without optic disc edema. Arch Ophthalmol
30. Keunen JE, van Norren D: Foveal 1996; 122:431–432. 1988; 106:1030–1031.
densitometry in the multiple evanescent 47. Borruat FX, Herbort CP, Spertini F, 64. Singh K, de Frank MP, Shults WT,
white-dot syndrome. Am J Ophthalmol Desarnaulds AB: HLA typing in patients Watzke RC: Acute idiopathic blind spot
1988; 105:561–562. with multiple evanescent white dot enlargement. A spectrum of disease.
31. Aaberg TM, Campo RV, Joffe L: syndrome (MEWDS). Ocul Immunol Ophthalmology 1991; 98:497–502.
Recurrences and bilaterality in the multiple Inflamm 1998; 6:39–41. 65. Volpe NJ, Rizzo JF 3rd, Lessell S: Acute
evanescent white-dot syndrome. Am J 48. Gass JD, Hamed LM: Acute macular idiopathic blind spot enlargement
Ophthalmol 1985; 100:29–37. neuroretinopathy and multiple evanescent syndrome: a review of 27 new cases.

CHAPTER 158
32. Jost BF, Olk RJ, McGaughey A: Bilateral white dot syndrome occurring in the same Arch Ophthalmol 2001; 119:59–63.
symptomatic multiple evanescent white-dot patients. Arch Ophthalmol 1989; 66. Gass JDM: Acute posterior multifocal
syndrome. Am J Ophthalmol 1986; 107:189–193. placoid pigment epitheliopathy. Arch
101:489–490. 49. Watzke RC, Packer AJ, Folk JC, et al: Ophthalmol 1968; 80:177.
33. Meyer RJ, Jampol LM: Recurrences and Punctate inner choroidopathy. Am J 67. Krill AE, Deutman AF: Acute retinal pigment
bilaterality in the multiple evanescent Ophthalmol 1984; 98:572–584. epitheliitus. Am J Ophthalmol 1972;
white-dot syndrome. Am J Ophthalmol 50. Dreyer RF, Gass DJ: Multifocal choroiditis 74:193–205.
1986; 101:388–389. and panuveitis. A syndrome that mimics 68. Chittum ME, Kalina RE: Acute retinal
34. Hernandez Artola F, Garay Aramburu G, ocular histoplasmosis. Arch Ophthalmol pigment epitheliitis. Ophthalmology 1987;
Llorca Pellicer S: Multiple evanescent 1984; 102:1776–1784. 94:1114–1119.
white-dot syndrome: bilateralization four 51. Cantrill HL, Folk JC: Multifocal choroiditis 69. Deutman AF: Acute retinal pigment
years later. Arch Soc Esp Oftalmol 2002; associated with progressive subretinal epitheliitis. Am J Ophthalmol 1974;
77:393–396. fibrosis. Am J Ophthalmol 1986; 78:571–578.
35. Jost BF, Olk RJ, McGaughey A: Bilateral 101:170–180. 70. Ryan SJ, Maumenee AE: Birdshot
symptomatic multiple evanescent white-dot 52. Callanan D, Gass JD: Multifocal choroiditis retinochoroidopathy. Am J Ophthalmol
syndrome. Am J Ophthalmol 1986; and choroidal neovascularization 1980; 89:31–45.
101:489–490. associated with the multiple evanescent 71. Gass JD: Vitiliginous chorioretinitis. Arch
36. Aaberg TM, Campo RV, Joffe L: white dot and acute idiopathic blind spot Ophthalmol 1981; 99:1778–1787.
Recurrences and bilaterality in the multiple enlargement syndrome. Ophthalmology 72. Doft BH, Gass DM: Punctate outer retinal
evanescent white-dot syndrome. Am J 1992; 99:1678–1685. toxoplasmosis. Arch Ophthalmol 1985;
Ophthalmol 1985; 100:29–37. 53. Dreyer RF, Gass DJ: Multifocal choroiditis 103:1332–1336.
37. Wyhinny GJ, Jackson JL, Jampol LM, and panuveitis. A syndrome that mimics 73. Gass JD, Scelfo R: Diffuse unilateral
Caro NC: Subretinal neovascularization ocular histoplasmosis. Arch Ophthalmol subacute neuroretinitis. J R Soc Med 1978;
following multiple evanescent white-dot 1984; 102:1776–1784. 71:95–111.
syndrome. Arch Ophthalmol 1990; 54. Cantrill HL, Folk JC: Multifocal choroiditis 74. Gass JD, Braunstein RA: Further
108:1384–1385. associated with progressive subretinal observations concerning the diffuse
38. Oh KT, Christmas NJ, Russell SR: Late fibrosis. Am J Ophthalmol 1986; unilateral subacute neuroretinitis syndrome.
recurrence and choroidal neovascularization 101:170–180. Arch Ophthalmol 1983; 101:1689–1697.
in multiple evanescent white dot syndrome. 55. Doran RM, Hamilton AM: Disciform 75. Bos PJ, Deutman AF: Acute macular
Retina 2001; 21:182–184. macular degeneration in young adults. neuroretinopathy. Am J Ophthalmol 1975;
39. Gass JD, Agarwal A, Scott IU: Acute zonal Trans Ophthalmol Soc UK 1982; 102(Pt 80:573–584.
occult outer retinopathy: a long-term 4):471–480. 76. Rush JA: Acute macular neuroretinopathy.
follow-up study. Am J Ophthalmol 2002; 56. Palestine AG, Nussenblatt RB, Parver LM, Am J Ophthalmol 1977; 83:490–494.
134:329–339. Knox DL: Progressive subretinal fibrosis 77. Gass JD, Hamed LM: Acute macular
40. Ray S, Loewenstein J: Atypical and uveitis. Br J Ophthalmol 1984; neuroretinopathy and multiple evanescent
manifestation of multiple evanescent white 68:667–673. white dot syndrome occurring in the same
dot syndrome with large peripapillary 57. Morgan CM, Schatz H: Recurrent multifocal patients. Arch Ophthalmol 1989;
lesion. Arch Ophthalmol 2003; choroiditis. Ophthalmology 1986; 107:189–193.
121:1794–1796. 93:1138–1147. 78. Fahim DK, Bucher R, Johnson MW: The
41. Ikeda N, Ikeda T, Nagata M, et al: Location 58. Callanan D, Gass JD: Multifocal choroiditis elusive nature of primary intraocular
of lesions in multiple evanescent white dot and choroidal neovascularization lymphoma. J Neuroophthalmol 2005;
syndrome and the cause of the associated with the multiple evanescent 25:33–36.
hypofluorescent spots observed by white dot and acute idiopathic blind spot 79. Shah GK, Kleiner RC, Augsburger JJ, et al:
indocyanine green angiography. Graefes enlargement syndrome. Ophthalmology Primary intraocular lymphoma seen with
Arch Clin Exp Ophthalmol 2001; 1992; 99:1678–1685. transient white fundus lesions simulating
239:242–247. 59. Kozielec GF, Wyhinny GJ, Jampol LM: the multiple evanescent white dot
42. Chung YM, Yeh TS, Liu JH: Increased Evolution of distinct chorioretinal scars in syndrome. Arch Ophthalmol 2001;
serum IgM and IgG in the multiple recurrent MEWDS. Retina 2001; 119:617–620.
evanescent white-dot syndrome. Am J 21:180–182.
Ophthalmol 1987; 104:187–188. 60. Oh KT, Christmas NJ, Russell SR: Late
43. Laatikainen L, Immonen I: Multiple recurrence and choroidal
evanescent white dot syndrome. Graefes neovascularization in multiple evanescent
Arch Clin Exp Ophthalmol 1988; white dot syndrome. Retina 2001;
226:37–40. 21:182–184.

2069
CHAPTER

159 Acute Zonal Occult Outer Retinopathy


Alan C. Bird

INTRODUCTION CLINICAL FINDINGS


The condition was probably reported initially by neuro- Characteristically, the fundus changes are those of the
ophthalmologists when patients were recognized as having a big precipitating disorder, with inner choroidal infiltrates or
blind spot without disk swelling or any other retinal abnor- scarring if it is PIC. In some cases, the fundi may be normal and
malities.1,2 The field loss was reported to be persistent over it is assumed that MEWDS was the cause given and the retinal
many months. It was then identified that in some, it was changes may be very limited in this disorder even a few weeks
progressive over a period extending temporally around the after its onset. Evidence of MEWDS may be evident on ICG
macula. Electro-physiological testing showed that the loss was angiography when the fundus appears to be normal on
due to retinal dysfunction. ophthalmoscopy. In the area of field loss, the retina appears
Gass was the first to recognize and characterize the nature normal including autofluorescence imaging. Visual fields show
of the disorder which he named acute zonal occult outer the characteristic loss.19,20 Both rod and cone sensitivities may
retinopathy (AZOOR) which he described in 1993 in his be reduced without there being an increase of time to recovery
Donder’s lecture.3 He reported 13 patients, mostly young from bleach.
women, who had rapid self limited loss of visual fields,
photopsia, minimal ophthalmoscopic changes. Spontaneous ERG FINDINGS
recovery could occur during the first 5 years, but in its absence
signs of retinal degeneration developed. The evidence that the ERG tests show that retinal dysfunction accounts for the field
loss was due to outer retinal dysfunction was based on the loss.21,19 The most sensitive test in flicker ERG in which the
interpretation of electrophysiological (ERG) abnormalities. implicit time is invariably prolonged. The additional changes
Gass, and subsequently others, described AZOOR as being include photopic transient ERGs with abnormal b-wave ampli-
associated with various inflammatory inner choroidal inflam- tude and increased implicit time the reduced scotopic rod
matory disease and retinopathies of unknown etiology responses were abnormal in nine eyes, and abnormal pattern
including the multiple evanescent white dot syndrome ERG. The light induced rise of ocular potential on electro-
(MEWDS),3–8 acute macular neuroretinopathy (AMN),9–11 oculography (EOG) is usually reduced to an extent not
multifocal choroidopathy,12–13 punctate inner choroidopathy explained by reduced photoreceptor responses.
(PIC)14,15 or pseudopresumed ocular histoplasmosis syndrome
(pseudo-POHS),16–18 PIC being the most common. It is con- CLINICAL COURSE
sidered that AZOOR was a complication of disorders that cause
inner choroidal inflammation rather than being intrinsic to the The long term course has been well recorded.21 Those who
primary disorder. present with unilateral AZOOR may have affection of the other
eye after weeks. Progression of the deficit has been recorded by
CLINICAL PRESENTATION both psychophysics and ERG testing over a period of a few
weeks followed by stability.
The vast majority of patients are female, healthy, in their In a proportion of cases, there is recovery of function over a
mid-thirties although the condition has been reported in the period of months or years shown both on visual field and ERG
seventh decade of life. The sex predilection is determined by testing although recovery is rarely complete. If after 5 years
that of the precipitating disorders. The condition presents with there is no recovery, widespread retinal changes occur that
progressive visual loss over a period of weeks or a few months. correspond with the field loss with retinal thinning and changes
The visual field loss most frequently involves blind spot at the level of the retinal pigment epithelium (RPE).
enlargement extending in an arcuate manner into the superior
and inferior temporal quadrants. Photopsia extending over the PATHOGENESIS
whole of the affected field is a prominent symptom described
as ‘scintillating’ or ‘shimmering’ lights. The visual acuity may The abolished EOG light rise and reduced photoreceptor cell,
or may not be affected. The severity is variable and visual acuity responses on ERG testing show that the abnormality is largely
as bad as counting fingers has been recorded. The field defect at the level of the RPE–photoreceptors complex. The greater
increases in size within a few weeks or months before reduction in EOG’s light rise than is easily explained by photo-
stabilizing. In most cases, the visual field loss and decrease in receptor functional loss implies the RPE affection is likely to be
visual acuity are asymmetric and some have unilateral intrinsic to the disorder. Autofluorescence images obtained with
symptoms only. a scanning laser ophthalmoscope are derived from lipofuscin 2071
RETINA AND VITREOUS

in the retinal pigment epithelium.22,23 The presence of firmed by Spaide et al.25 Candida famata was isolated from
lipofuscin is thought to reflect the metabolic activity of the RPE conjunctival exudates of a patient diagnosed with AZOOR.26
which is determined largely by the rate of turnover of photo- Immunological tests revealed the presence of antigen-specific
receptor outer segments. The finding of normal autofluor- T lymphocytes by using C. famata as a challenge, and enzyme-
escence distribution early in disease implies that there is no linked immunosorbent assay (ELISA) analysis showed the
significant loss of photoreceptors, and the presence of normal presence of specific antibodies against this yeast in the patient’s
outer segment and retinoid turnover. That the loss is due to cell blood. Delayed hypersensitivity by use of a skin test was also
dysfunction and not cell loss might be expected from the positive.
normal appearance of the retina on clinical examination. The Alternatively, it was considered that antibodies to retina may
loss of b-wave amplitude in some cases that is greater than have been induced either by the infective agent or by release
a-wave loss suggest that, in these, there is posttransduction of antigens from the retina. However, Jacobson found no
abnormality as well. indication that patients with AZOOR as a group have auto-
The mechanism leading to malfunction of the retinal antibodies that recognize a specific cell type in sections of
RPE–photoreceptor–epithelium complex in AZOOR and the human or rat retina. None of the AZOOR sera/IgGs produced
causal relationship between the initiating event and the specific labeling of rat retinal cells.20 No similar antibodies
widespread function loss are still unknown. Why patients with reactive with a specific retinal antigen were found in AZOOR.
multifocal choroiditis or punctate inner choroidopathy,15
presumed ocular hystoplasmosis syndrome (POS), MEWDS are TREATMENT
SECTION 10

at high risk of developing AZOOR is not understood. An


autoimmune etiology or toxic mechanism precipitated possibly As a result of finding hypersensitivity to C. famata antifungal
by a virus has been proposed by some. It was suggested that treatment was tried and it was reported that there were
persistence of virus may be the cause. Tiedeman24 found improvements in several clinical symptoms, including
evidence of persistent EBV infection in 10 patients with multi- fundoscopic analysis. This observation has not been repeated,
focal choroiditis and panuveitis. His findings were not con- and the general view is that treatment is unhelpful.21

REFERENCES
1. Rosenberg ML, O’Connor P, Carter J: syndrome. Arch Ophthalmol 1989; 18. Elliott JH, Jackson DJ: Presumed
Idiopathic unilateral disk edema. The big 107:194–198. histoplasmatic maculopathy: clinical course
blind spot syndrome. J Clin 9. Singh K, de Frank M, Shults WT, Watzke RK: and prognosis in nonphotocoagulated
Neuroophthalmol 1984; 4:181–184. Acute idiopathic blind spot enlargement: a eyes. Int Ophthalmol Clin 1975; 15:29–30.
2. Fletcher WA, Imes RK, Goodman D, spectrum of disease. Ophthalmology 1991; 19. Francis PJ, Marinescu A, Fitzke FW, et al:
Hoyt WF: Acute idiopathic blind spot 98:497–502. Acute zonal occult outer retinopathy:
enlargement. A big blind spot syndrome 10. Bos PJM, Deutman AF: Acute macular towards a set of diagnostic criteria. Br J
without optic disk edema. Arch Ophthalmol neuroretinopathy. Am J Ophthalmol 1975; Ophthalmol 2005; 89:70–73.
1988; 106:44–49. 80:573–584. 20. Gass JD, Agarwa A, Scott IU: Acute zonal
3. Gass JDM: Acute zonal occult outer 11. Gass JDM, Hamed L: Acute macular occult outer retinopathy: a long-term
retinopathy. J Cl Neuro-ophthalmology neuroretinopathy and multiple evanescent follow-up study. Am J Ophthalmol 2002;
1993; 13:79–97. white dot syndrome occurring in the same 134:329–339.
4. Jampol LM, Sieving PA, Pugh D, et al: patients. Arch Ophthalmol 1989; 21. Jacobson SG, Morales DS, Sun XK, et al:
Multiple evanescent white dot syndrome. 107:189–193. Pattern of retinal dysfunction in acute zonal
I. Clinical findings. Arch Ophthalmol 1984; 12. Khorram KD, Jampole LM, Rosenberg MA: occult outer retinopathy. Ophthalmology
102:671–674. Blind spot enlargement as a manifestation 1995; 102:1187–1198.
5. Sieving PA, Fishman Ga, Jampol LM, of multifocal choroiditis. Arch Ophthalmol 22. von Rückmann A, Fitzke FW, Bird AC:
Pugh D: Multiple evanescent white dot 1991; 109:1403–1417. Distribution of fundus autofluorescence
syndrome. II. Electrophysiology of the 13. Morgan CM, Schatz H: Recurrent multifocal with a scanning laser ophthalmoscope.
photoreceptors during retinal pigment choroiditis. Ophthalmology 1986; Br J Ophthalmol 1995; 79:407–412.
epithelial disease. Arch Ophthalmol 1984; 93:1138–1147. 23. von Rückmann A, Fitzke FW, Bird AC:
102:675–679. 14. Watzke RC, Packer AJ, Folk JC, et al: In vivo fundus autofluorescence in macular
6. Aaberg TM, Campo RV, Joffe L: Recurrence Punctate inner choroidopathy. Am J dystrophies. Arch Ophthalmol 1997;
and bilaterality in the multiple evanescent Ophthalmol 1984; 98:572–584. 115:609–615.
white dot syndrome. Am J Ophthalmol 15. Eldem B, Cumhur S: Punctate inner 24. Tiedeman JS: Epstein Barr viral antibodies
1985; 100:29–37. choroidopathy and its differential diagnosis. in multifocal choroiditis and panuveitis.
7. Callanan D, Gass JDM: Multifocal choroiditis Ann Ophthalmol 1991; 23:153–159. Am J Ophthalmol 1987; 103:659–663.
and choroidal neovascularisation 16. Gass JDM, Wilkinson CP: Follow-up study 25. Spaide RF, Sugin S, Yannuzzi LA,
associated with the multiple evanescent of presumed ocular histoplasmosis. Trans DeRosa JT: Epstein Barr virus antibodies in
white dot and acute idiopathic blind spot Am Acad Ophthalmol Otolaryngol 1972; multifocal choroiditis and panuveitis.
enlargement syndrome. Ophthalmology 76:672–694. Am J Ophthamol 1991; 112:410–413.
1992; 99:1678–1685. 17. Gutman FA: The natural cause of active 26. Carrasco L, Ramos M, Galisteo R, et al:
8. Hamed LM, Glaser JS, Gass JDM, choroidal lesions in the presumed ocular Isolation of Candida famata from a patient
Schatz NJ: Protracted enlargement of the histoplasmosis syndrome. Trans with acute zonal occult outer retinopathy.
blind spot in multiple evanescent white dot Ophthalmol Soc 1979; 777:515–541. J Clin Microbiol 2005; 43:635–640.

2072
CHAPTER

160 Macular Epiretinal Membranes


Darin R. Haivala and David W. Parke II

INTRODUCTION reported in the Blue Mountains Eye Study to occur in 13.5% of


patients over a 5 year time period.11
Iwanoff first described the abnormal proliferation of cellular Nonidiopathic ERMs have been associated with a wide
membranes on the inner retinal surface in 1865.1 Since this variety of vitreo-retinal diseases including retinal vascular
initial description, surface proliferation in the macular region disease, vitreo-retinal inflammatory conditions, postoperative
has been subsequently described as epimacular membrane, (particularly scleral buckle) and posttraumatic states, inherited
macular pucker, cellophane maculopathy, preretinal macular and congenital posterior segment anomalies and syndromes,
fibrosis, wrinkling of the internal limiting membrane, among and intraocular tumors. Inflammation and/or vascular leakage
others.2–8 Although histologically similar to epiretinal prolifera- appear to be common mediators. Retinal vascular diseases
tion elsewhere associated with proliferative vitreoretinopathy, it associated with increased vascular permeability and ERMs
is clinically considered a separate entity based on its location, include diabetic and hypertensive retinopathy, venous occlusive
clinical features, and clinical course. Most commonly, macular disease, angiomas, telangiectasis, as well as others (Fig. 160.1).12–23
epiretinal membranes (ERMs) are asymptomatic or cause mild Intraocular inflammation resulting in an inflammatory cellular
symptoms of metamorphopsia and/or modest decreased central infiltrate in the vitreous may also be associated with ERM
acuity. A minority of these membranes can cause sufficient formation. Any infectious or noninfectious ocular condition
macular distortion and macular edema to induce clinicians to resulting in intermediate or posterior uveitis may be causative.
recommend ERM removal via pars plana vitrectomy (PPV). Common examples include both toxoplasmosis and pars planitis
(Fig. 160.2).13,24-29 Ocular trauma, including both blunt and
penetrating injuries, particularly if there is associated vitreous
EPIDEMIOLOGY hemorrhage (as well as nearly all ocular surgery) may lead to
While ERM formation has been associated with and results ERM formation or to worsening of preexisting ERMs.30–38
from a variety of primary intraocular diseases, the vast majority Although cataract surgery has been reported to be associated
are considered to be idiopathic, unassociated with other sys- with new ERMs in 9% of eyes, it is likely that a substantial per-
temic or ocular disease. They are found most frequently over centage of these membranes were preexisting and undetected,
the age of 50, and several large clinical studies have noted a or were preexisting and worsened during the perioperative
clinical prevalence of between 7% and 11.8%.9,10 Most of these period.
are asymptomatic, with many being extrafoveal in location. Epiretinal membrane formation is not uncommon following
There appears to be no significant gender predilection and successful retinal detachment surgery, occurring in 4–8% of
20–30% are bilateral. Posterior vitreous detachments are eyes. Retinal breaks both before and after laser or cryo-
present in up to 90% of clinically significant ERMs. (The retinopexy are frequently associated with subsequent retinal
absence of a posterior vitreous detachment in the presence of an surface membrane proliferation (Fig. 160.3).4,30–38 Among the
‘ERM’ should alert the clinician to the probability of the less common associations with ERM are retina tumors
vitreomacular traction syndrome). Second eye involvement was (particularly vascular tumors) and retinitis pigmentosa.

FIGURE 160.1. (a) Acute CRVO. (b) ERM


development 1 year later.

a b
2073
RETINA AND VITREOUS

PATHOGENESIS AND PATHOLOGY


The cellular origin of epiretinal membranes has long been
debated, and almost every possibility has been considered. Iwanoff,
in 1865, implicated the endothelial cell in the formation of the
membranes.1 Manschot, in 1958, believed that the membrane
was an extension of the Müller cell processes,39 whereas Wolter
considered the cells to originate from fibroblasts in the vascular
connective tissue.40 Smith suggested that the cells in the mem-
brane originated from the pigmented or nonpigmented cells of
the pars ciliaris, retinal pigment epithelium (RPE) cells, meso-
dermal elements of the vascular system, normal vitreous cells,
inflammatory cells within the vitreous, or retinal glial cells.41 In
1962, Kurz and Zimmerman believed that the cells originated
from migration of RPE cells.42 Ashton later suggested that the
cells originated from a transformation of vascular mesenchymal
cells into fibroblasts,43 and in 1969, von Gloor proposed that
hyalocytes were the cells of origin.44
SECTION 10

The advent of vitreous surgery has provided an opportunity


to advance our understanding of the clinicopathologic relation-
ships of epiretinal membranes. Numerous authors have
attempted to characterize the cells of origin and correlate this
FIGURE 160.2. ERM after exogenous endophthalmitis. with the ERM etiology. Many studies have determined that the
predominant cells types in idiopathic ERMs removed at surgery
include glial cells or fibrous astrocytes and (particularly with
clinically severe membranes) elements of retinal pigment
epithelial cells, fibrocytes, and macrophages. This is consistent
Key Features with a theory of idiopathic ERM pathogenesis first advanced by
Causes of Macular ERMs Roth and Foos.
• Idiopathic Foos suggested that the glial cells found in the thin,
• Retinal vascular disease idiopathic membranes were derived from the glial cells of the
• Diabetic retinopathy superficial retina (fibrous astrocytes and Muller cells) that had
• Retinal vein occlusion migrated through breaks in the internal limiting membrane
• Perifoveal telangiectasia syndrome (ILM) to proliferate on the retinal surface.45 These ILM breaks
• Other were hypothesized to be associated with acute separation of the
• Posterior segment inflammatory disease posterior vitreous from the macular region (where it has rela-
• Idiopathic posterior uveitis tively tight adherence). This hypothesis was subsequently sup-
• Sarcoidosis ported by Bellhorn and associates.46
• Pars planitis ERMs occurring following retinal breaks and detachment are
• Other felt to have a different pathogenesis. In these situations, retinal
• Posttraumatic pigment epithelial (RPE) cells gain access to the vitreous cavity
• Postoperative through the retinal break and settle on the macular surface,
• Retinal reattachment surgery subsequently developing a membrane. These membranes are
• Treatment of retinal break architecturally enhanced by the presence of fibrocytes and macro-
• Retinal break phages, stimulated in part by the inflammation associated with
• Intraocular tumors vitreous hemorrhage and/or surgical repair.
• Angiomas Most published reports on the ultrastructure of epiretinal
• Other macular membranes have been on vitrectomy specimens in
• Miscellaneous elderly patients. Vinores and colleagues studied the ultra-
• Retinitis pigmentosa structural and electron immunocytochemical characterization
• Combined hamartoma of the RPE of cells in epiretinal membranes. Their work suggests that both
RPE cells and retinal glial cells are most likely to be the major

FIGURE 160.3. (a) ERM following cryotherapy


of a peripheral retinal break. (b) OCT showing
highly reflective ERM, corrugation of retinal
surface, loss of foveal depression, and
increased central macular thickness.

a b

2074
Macular Epiretinal Membranes

participants in the pathogenesis of epiretinal membranes.47 the 20/200 level may be seen in a small number of patients
Smiddy and co-workers reported on membranes removed in (~5%).64–66 Other complaints may include monocular diplopia
children and young adults. They noted that myofibroblasts, and macro or micropsia.
myoblastic differentiation of RPE cells and fibrous astrocytes, Clinicians will encounter cases of patients with substantially
and new collagen formation tended to be found more frequently decreased vision but clinically unimpressive ERMs. In such
in younger patients.48 The progressive tangential and antero- situations, substantial macular edema or (more rarely) a lamellar
posterior traction exerted by ERMs is likely due to the myofibro- or full-thickness macular hole may be present. On the other
plast characteristics of the membrane which have been hand, a clinically impressive ERM but with a central pseudo-
determined on immunohistochemical staining to contain actin, hole may result in preservation of good central visual acuity.
transforming growth factor, and fibronectin.49 Optical coherence tomography (OCT) assists substantively in
The dispersion of RPE cells on the retinal surface has been the evaluation of these patients.
demonstrated clinically and experimentally in cases of retinal
breaks and detachment.50–54 However, the finding of RPE cells CLINICAL ASSESSMENT
in idiopathic membranes is more difficult to explain. Smiddy
and colleagues suggested that the RPE cells gain access to the Evaluation of an eye with a suspected ERM begins first with
retinal surface by various methods including migration through testing of central visual acuity and central visual fields (such as
occult breaks, by inactivation of developmental rests of RPE via an Amsler grid). Retinal distortion or macular heterotopia
cells already on the surface of the retina, through transforma- may induce metamorphopsia and Amsler grid abnormalities.

CHAPTER 160
tion from other cell types, or via transretinal migration.55 The Indirect ophthalmoscopy and slit-lamp biomicroscopy consti-
exact mechanism or combination of mechanisms is yet to be tute the next major element in the evaluation process. The
elucidated. Stern and co-workers56 suggested that the contrac- clinical findings differ greatly depending on the severity of the
tive forces of the membranes were related to their constituent membrane and whether it has undergone significant contrac-
cell types and not dependent on intercellular collagen, as sug- tion. The membrane may be very fine and translucent without
gested by previous investigators.57 an identifiable edge. These may be identifiable only on careful
Collagen is a component of all surgically removed epiretinal contact lens biomicroscopy and can be as subtle as a fine ‘sheen’
membranes. The abundance of collagen in some membranes in the macular region. There may be blunting or an irregularity
and the presence of fibrocytic and macrophage-like cells suggest of the foveal light reflex. These finding may be best appreciated
that vitreous hyalocytes may be a significant premetaplasia cell on red-free or monochromatic green or blue light. Vascular
of origin in these collagen membranes.58 tortuosity may be more evident on fluorescein angiography than
on clinical inspection (Fig. 160.4). In such cases, visual acuity
CLINICAL FINDINGS: SYMPTOMATOLOGY may be determined principally by the amount of associated
macular edema or macular detachment.
Symptoms associated with epiretinal membrane formation vary More extensive membranes may take on an opaque appear-
greatly depending on the location and characteristics of the ance and obscure underling retinal vasculature. They may be
membrane as well as the amount of underlying macular archi- pigmented, particularly in the case of membrane formation
tectural disruption. Patients with membranes that are very thin following the treatment of a retinal break or retinal detachment,
or outside the central macula are oftentimes completely asymp- severe intraocular inflammation, or vitreous hemorrhage.67–69
tomatic. It is not uncommon for these patients to have normal More severe membranes may be contractile and result in iden-
or near-normal visual acuity. Subtle complaints of blurring of tifiable striae at the level of the internal limiting membrane and
vision or metamorphopsia may develop with increasing trac- inner retina, or true retinal folds. The contraction of the mem-
tion, membrane opacification, or macular edema, and the onset brane may also result in traction on surrounding retinal vessels
is often insidious. These patients typically remain very stable resulting in abnormal distortion and a tethering effect. It is
with only 10–25% of patients losing one or two lines of vision not uncommon for the major vasculature to have a more
over a 2 year time period.11,23,59–62 Of those patients with idio- ‘straightened’ appearance than that of the fellow eye. Contrac-
pathic membranes, two-thirds will have 20/30 or better vision tion may also result in macular ectopia and traction. The
and 85% will display a visual acuity of 20/70 or better.8,63 At the resulting macular heterotopia may cause diplopia. Contraction
opposite end of the spectrum are those patients with very thick may also lead to foveolar detachment.
central membranes associated with significant underlying Small intraretinal hemorrhages as well as evidence of pre-
retinal architectural distortion, subtle detachment of the sumed axoplasmic stasis or ischemia may be seen as a result of
posterior pole, and/or macular edema. Visual acuity at or below ERM-induced vascular traction. The axoplasmic stasis can

FIGURE 160.4. (a) Idiopathic ERM.


(b) Fluorescein image demonstrating
combination of vascular tortuosity and
straightening.

a b
2075
RETINA AND VITREOUS

manifest itself as cotton-wool spots as well as larger areas of


Key Features
inner retinal whitening.70 Additional biomicroscopic findings
Value of OCT in Macular ERMs
may include macular edema with cystic changes, subfoveal
• Diagnostic confirmation
retinal pigment epithelial changes, and macular pseudoholes
• Poor visualization
which have been reported in up to 8–20% of eyes with epiretinal
• Topographic localization
membranes.71–74 Epiretinal membranes are found in up to 30%
• Vitreoretinal relationships
of patients with true full-thickness macular holes and these
• Associated vitreomacular traction
must be differentiated from pseudoholes.23,75
• Points of attachment
A posterior vitreous separation is present in up to 80–90% of
• Multilaminar membranes
patients with epiretinal membrane, but the true vitreoretinal
• Identify coincident pathology
relationships may be difficult to discern clinically.8,23,59,60,63,66,76
• Macular holes
Margherio et al noted that the vitreoretinal interface relation-
• Macular pseudoholes
ships were more accurately evaluated at the time of vitrectomy
• Macular edema
surgery by observing the effects of surgical manipulation on the
• Age-related macular degeneration
underlying retina.77 In some cases, large areas of vitreous col-
• Assist in preoperative patient counseling
lapse noted intraoperatively with an adherent posterior hyaloid
• Evaluation of postoperative status
were misinterpreted as a true vitreous separation preoperatively.
• Macular thickness
In eyes with a partial vitreous separation, spontaneous mem-
SECTION 10

• Membrane regrowth
brane avulsion with the complete detachment of the posterior
vitreous has been reported.78 The spontaneous ‘peeling’ of an
epiretinal membrane has also been described in patients with a can assist in postoperative management by assessing macular
complete vitreous separation. An edge of the ERM may ‘scroll’ thickness and membrane re-growth.
upon itself, contracting to the other edge of the membrane Fluorescein angiography, although still useful in evaluating
resulting in spontaneous visual improvement.23,79 selected ERM cases, has been partially supplanted by OCT. The
Any eye with an ERM and without evidence of a clear pos- principal utility of angiography in the setting of an ERM is to
terior vitreous separation must be carefully inspected clinically rule out the concomitant presence of retinal vascular disease or
and by OCT for evidence of the vitreomacular traction syn- of underlying pigment epithelial pathology. In some ERM cases
drome (VMTS) (Fig. 160.5). In the VMTS, a partially detached with intraretinal hemorrhages, it may be impossible to rule out
posterior hyaloid retains attachments in the posterior pole an associated small branch retinal vein occlusion by ophthal-
resulting in traction upon the macular region. VMTS frequently moscopy alone. Similarly, it may be necessary to perform
coexists with an epiretinal membrane. Complete surgical fluorescein angiography to rule out the presence of a choroidal
management therefore requires not only removal of VMTS- neovascular membrane in the patient with substantial intra-
induced traction, but also removal of any associated epiretinal retinal and/or submacular fluid. Finally, stereo fundus photo-
tissue. graphy and angiography may assist in delineation of vitreoretinal
Attention must always be paid to the peripheral retina as relationships, ERM borders, and degree of retinal vascular dis-
well. The new diagnosis of an ERM, particularly if purely uni- tortion in the difficult-to-examine patient.
lateral, pigmented, or associated with pigmented vitreous cells,
must stimulate a careful examination for a peripheral retinal
break. This is not only an etiologic consideration, but impor-
tant in ultimate management plans.
OCT can play an important role in the clinical assessment of
eyes with ERMs (Fig. 160.6). OCT can not only detect ERMs,
but also assist in topographic localization, identification of
vitreoretinal relationships (such as in the vitreomacular traction
syndrome), detection of macular holes, and quantitation of
macular thickness and macular volume. In addition to value in
clinical characterization, OCT has therapeutic value in pre-
operative planning. The co-existence of a macular hole, the
presence of a bilaminar ERM, or knowledge of substantive
macular edema may lead the surgeon to modify his or her FIGURE 160.6. OCT showing typical ERM features including highly
approach. It also has considerable prognostic value in coun- reflective ERM, multiple points of retinal attachment, loss of foveal
seling patients as to the eye’s likely visual potential. Finally, it depression, and increased retinal thickness.

FIGURE 160.5. (a) Fundus photograph


demonstrating loss of central foveal depression.
(b) OCT demonstrating central vitreomacular
traction and traction retinal elevation.

a b
2076
Macular Epiretinal Membranes

TREATMENT brane has gained popularity. Indocyanine green, trypan blue,


and triamcinolone acetonide have been used to help visualize
The vast majority of patients who present with epiretinal mem- epiretinal tissue to facilitate its complete removal.84-91
brane will remain stable if observed over a period of time.11,60,66,80 Once an edge has been elevated, the ‘peeling’ of the tissue can
From the standpoint of the surgeon and the patient, this is be advanced using intraocular forceps, regrasping the mem-
characteristically not an inexorably progressive disease. brane near the retinal surface while advancing across the pos-
However, visual function may progressively deteriorate in some terior pole. This should be carried out in a tangential fashion,
patients, due to progressive membrane thickening, retinal limiting anterior traction. Care should be taken to minimize
distortion, macular edema, lamellar hole formation, or retinal traction on a very cystic macula to mitigate the risk of ‘unroofing’
pigment epithelial change. Episodes of intraocular inflammation a large central cyst. It is not uncommon for a seemingly
or intraocular surgery may be followed by alterations in ERM localized ERM to extend well beyond the major vascular arcades.
architecture or the amount of associated macular edema. The extent of more peripheral membranectomy should depend
Therefore, it is impossible to predict for any patient what will on ease of removal and associated pathology. After the mem-
be the course of any specific ERM. This renders decisions brane has been elevated, it is not uncommon to see superficial
difficult regarding surgery and the timing of surgery. In cases retinal whitening in the area where the membrane has been
with significant or progressive vision loss, debilitating removed. It is important to differentiate this whitening from
metamorphopsia or diplopia, surgical intervention should be residual epiretinal tissue in the cases of multilayered mem-
considered. Most surgeons reserve surgery for those patients with branes. As always, care should be taken to ensure that peri-

CHAPTER 160
vision reduction at least to the 20/50–20/60 level, although pheral or mid-peripheral retinal breaks have not occurred with
earlier surgery may be considered for those with debilitating instrument changes or with traction on the retina by extensive
symptoms or specific visual needs. The goals of surgical membranes.
intervention include the removal of all epimacular tissue and
relief of all macular traction with the subsequent resolution of RESULTS OF SURGERY
underlying-traction-induced retinal folds, macular edema, as
well as traction-induced axoplasmic stasis.81 In a majority of patients, the macular surface architecture is
Once the surgeon and the patient have elected to proceed greatly improved in the immediate postoperative period, though
with surgery, conventional 20-, 23-, or 25-gauge vitrectomy is short-term vision reduction beyond the preoperative level is not
carried out. A core vitrectomy is completed and the posterior uncommon (Figs 160.7 and 160.8). While immediate and signifi-
cortical vitreous should be elevated and excised in those cases cant improvement in acuity may occur, it often takes 4–6 weeks
where a complete vitreous separation is not present. (As noted for the patient’s vision to return to the preoperative level, with
earlier, posterior vitreous separation is present in the vast subsequent improvement over the ensuing 3-6 months. This
majority of ERM cases. Anteroposterior traction indicates that course can be modulated by factors most commonly including
the vitreomacular traction syndrome is present.) Attention is cystoid macular edema, cataract formation, and ERM regrowth.
then turned to the macular surface. A prominent edge of the Numerous authors have attempted to identify prognostic
epiretinal tissues can often be identified and this provides a con- indicators of postoperative vision in vitrectomy for treatment of
venient location in which to engage the ERM. Using a barbed ERMs. Indicators that have been mentioned are (1) preoperative
micro-vitreoretinal blade, bent 25-gauge needle, or retinal pick, visual acuity, (2) duration of diminished acuity before surgery,
the edge of the membrane can be ‘engaged’ in a tangential, (3) the presence of preoperative cystoid macular edema, (4) the
‘dragging’ fashion, elevating the membrane edge. An alternative age of the patient, (5) the thickness of the epiretinal membrane,
method involves using an intraocular diamond dusted or end- (6) idiopathic versus nonidiopathic epiretinal membranes, and
grasping forceps to ‘pinch’ the membrane and initiate removal. (7) the presence of RPE window defects on fluorescein angiography.
In cases where a defined edge is not identifiable, the same Trese and colleagues reported that transparent membranes had
‘dragging’ maneuver can be used to ‘snag’ the membrane a better visual prognosis than opaque ones. They also believed
resulting in the elevation of a piece of the membrane that can that the presence of cystoid macular edema was a poor prog-
be further advanced. Additional tools, such as the Tano Diamond nostic sign.62 However, Rice and associates found that eyes with
Dusted Membrane Scraper,82,83 can be very helpful in ‘snagging’ thin, transparent membranes had a poorer prognosis than eyes
the membrane in cases where an identifiable edge is not present. with thicker membranes.92 Several additional studies found no
Recently, the use of surgical adjuncts to help visualize the mem- association between the transparency of the membrane and the

a b c

FIGURE 160.7. (a) ERM following treatment of a retinal detachment. (b) Fluorescein angiography showing ERM and retinal edema.
(c) Appearance following vitrectomy with removal of ERM. 2077
RETINA AND VITREOUS

FIGURE 160.8. (a) Idiopathic ERM before


surgery. (b) Idiopathic ERM after vitrectomy
showing mild residual retinal fold.

a b
SECTION 10

final postoperative vision.69 (Ferencz JR, Mussbaum JJ, Thompson, in a recent review of eyes with 20/50 or better
Richards SC, et al: Predictive variables in vitrectomy for idio- preoperative acuity, reported that mean postoperative
pathic epimacular membranes. Presented at Schepens Alumni pseudophakic acuity improved to 20/32.110 Some patients will
Meeting, Boston, MA, 9 Jun 1990.) Other studies have also describe a relief of metamorphopsia or diplopia which appears
suggested that the presence of cystoid macular edema is a poor more rewarding to them than their return of Snellen acuity.111
prognostic sign.62,92,93,94,95,96 However, larger series have shown
no relationship between the presence of preoperative cystoid SURGICAL COMPLICATIONS
macular edema and the ultimate visual prognosis.97,98,99,100 Two
studies have suggested that even in eyes with severe degrees of The most common postoperative complication following
macular edema, epiretinal membrane peeling may significantly vitrectomy for epiretinal membrane is progressive nuclear
improve visual acuity.101,102 sclerotic cataract in the phakic patient. This has been reported
It is worth noting that all of these series were performed in 12% to nearly 70% of patients.93,99,109,94,95,100,96,112 It is not
before the advent of intravitreal triamcinolone acetonide uncommon for patients to experience a gradual improvement in
injection, which is utilized as a surgical adjunct by many vision follow vitrectomy surgery, followed by an insidious
surgeons when substantial preoperative cystoid macular edema decline as the nuclear sclerosis progresses. The final best-
is identified. It is well recognized that even total removal of corrected visual acuity may very well be following cataract
ERMs (clinically and by postoperative OCT) may not achieve extraction. For this reason, some surgeons routinely recom-
normal macular thickness in all eyes. mend combined phacoemulsification, posterior chamber
The presence of portions of the internal limiting membrane intraocular lens implantation and ERM removal in patients
in surgical specimens has been evaluated as a possible prognos- with any preexisting nuclear sclerosis. The cataract surgery is
tic factor in epiretinal macular membrane surgery. Sivalingam performed first, followed by the vitrectomy surgery. Even
and associates found that surgical specimens from eyes con- patients without preexisting nuclear sclerosis are generally told
taining long segments of internal limiting membrane had a less to expect more rapid progression of lens changes with a likeli-
favorable visual prognosis.103 Other series did not find a similar hood of cataract surgery within several years.
correlation.104,105 Some surgeons prefer to remove ILM as a From the opposite perspective, patients with visually signifi-
routine part of ERM surgery cases, with one preliminary study cant cataracts and ERMs judged to be clinically insignificant
suggesting a lower incidence of membrane recurrence.106 should be informed that cataract surgery may carry with it a
Nonidiopathic ERMs (particularly those following retinal higher postoperative incidence of cystoid macular edema and
reattachment surgery) have been demonstrated to have an over- worsening of the ERM.
all worse prognosis than idiopathic ERMs. This is correlated Peripheral retinal tears are encountered in up to 6% of
with antecedent macular damage related to underlying pathol- cases.38,93,108,99,109,96 Tears are likely related to the insertion of
ogy such as a prior macula off retinal detachment or macular instruments through the vitreous base with resultant peripheral
ischemia or severe cystoid macular edema secondary to retinal retinal traction. In cases where the posterior cortical vitreous
vascular disease. A recent paper studying ERM removal in eyes remains attached, inducing a vitreous separation can also lead
following retinal detachment repair by Council and co-authors to peripheral retinal tears. When identified, these tears should
noted that 88% of eyes experienced an improvement in acuity be treated with either transcleral cryotherapy or laser photo-
postoperatively with 65% of eyes 20/60 or better; 6.7% of eyes coagulation, and occasionally fluid–air or gas exchange.
had a recurrent retinal detachment.107 Posterior retinal breaks resulting from iatrogenic traction are
In those patients undergoing surgery for removal of idiopathic uncommon. Late retinal detachments have been reported in up
ERMs, 80–90% of patients will gain 2 Snellen lines.93,108,99 A to 7% of patients undergoing vitrectomy surgery and are largely
2 line gain was seen in 65–80% of patients with membranes related to missed peripheral tears or subsequent contraction of
associated with previous retinal tear or detachment.108,99,109 While the vitreous base or incarcerated vitreous with resultant tear
the Snellen acuity improvement may be limitied in some, the formation.93,108,112
metamorphopsia is improved in most. In a series of 328 eyes, The recurrence of significant epiretinal tissue following
Margherio, Cox, and Trese demonstrated visual acuity improve- vitrecomy is seen in up to 5% of patients, depending on the
ment of at least 2 lines in 74% of patients. Twenty-four percent underlying etiology.93,108,99,96 Whether the use of dyes and
were unchanged, and 2% were noted to have a reduction in their substances to identify the membrane and/or internal limiting
preoperative visual acuity level.99 Twenty-five to fifty percent of membrane peeling will reduce this risk of recurrence is unclear.
2078 eyes will achieve a final acuity of 20/40 or better.93,99,109,96 Only a small percentage of membrane regrowths become visually
Macular Epiretinal Membranes

significant. Other complications including endophthalmitis, associated with a wide variety of ocular conditions generally
retinal phototoxicity, choroidal neovascularization, macular involving inflammation or retinal vascular leakage or prior
hole, and visual field defects have also been described.113 retinal detachment. Surgical intervention should be considered
when warranted by visual symptomatology. A discussion
CONCLUSION regarding realistic postsurgical outcomes and well as the
potential risk of vitrectomy needs to be carried out prior to
Macular epiretinal membranes most commonly represent an proceeding with surgery. Vitrectomy with membrane removal
idiopathic growth of transparent tissue across the macular results in symptomatic improvement in a vast majority of
region. The majority of these membranes are not visually sig- patients and plays a very important role in managing patients
nificant and do not require treatment. A minority of ERMs are with this relatively common condition.

REFERENCES
1. Iwanoff A: Beitrage zur normalen und vitrectomy. Trans Ophthalmol Soc UK 1985; 32. Winthrop SR, Cleary PE, Minckler DS et al:
pathologischen anatommie des auges. 104:285–296. Penetrating eye injuries: a histopathological
Grafes Arch Clin Exp Ophthalmol 1865; 17. Carney MD, Jampol LM: Epiretinal review. Br J Ophthalmol 1980; 64:809–817.
11:135–170. membranes in sickle cell retinopathy. 33. Green WR, Kenyon KR, Michels RG et al:
2. Roth AM, Foos RY: Surface wrinkling Arch Ophthalmol 1987; 105:214–217. Ultrastructure of epiretinal membranes

CHAPTER 160
retinopathy in eyes enucleated at autopsy. 18. Laatikainen L, Immonen I, Summanen P: causing macular pucker after retinal
Trans Am Acad Ophthalmol Otolaryngol Peripheral retinal angioma-like lesion and reattachment surgery. Trans Am
1971; 75:1047–1059. macular pucker. Am J Ophthalmol 1989; Ophthalmol Soc 1979; 99:63–77.
3. Francois J, Verbraeken H: Relationship 108:563–566. 34. Jahn CE, Minich V, Moldaschel S et al:
between the drainage of the subretinal 19. McDonald HR, Schatz H, Johnson RN et al: Epiretinal membranes after extracapsular
fluid in retinal detachment surgery and Vitrectomy in eyes with peripheral retinal cataract surgery. J Cataract Refract Surg
the appearance of macular pucker. angioma associated with traction macular 2001; 27:753–760.
Ophthalmologica 1979; 179:111–114. detachment. Ophthalmology 1996; 35. Saran BR, Brucker AJ: Macular epiretinal
4. Tanenbaum HL, Schepens CL, Elzeneiny I, 103:329–335; discussion 34–35. membrane formation and treated retinal
Freeman HM: Macular pucker following 20. Moriarty BJ, Acheson RW, Serjeant GR: breaks. Am J Ophthalmol 1995;
retinal detachment surgery. Arch Epiretinal membranes in sickle cell disease. 120:480–485.
Ophthalmol 1970; 83:286–293. Br J Ophthalmol 1987; 71:466–479. 36. Lincoff HA, McLean JM: Cryosurgical
5. Tannenbaum HL, Schepens CL, 21. Nicoletti VG, Nicoletti R, Ferrara N et al: treatment of retinal detachment. Am J
Elzeneiny I, Freeman HM: Macular pucker Diabetic patients and retinal proliferation: Ophthalmol 1966; 61:1227–1234.
following retinal detachment surgery. and evaluation of the role of vascular 37. Lobes LAJ, Burton TC: The incidence of
A biomicroscopic study. Can J Ophthalmol endothelial growth factor (VEGF). macular pucker after retinal detachment
1969; 4:20–23. Exp Clin Endocrinol Diabetes 2003; surgery. Am J Ophthalmol 1978; 85:72–77.
6. Jaffe NS: Macular retinopathy after 111:209–214. 38. Michels RG: Vitretomy for macular pucker.
separation of vitreoretinal adherence. 22. Schwartz PL, Fastenberg DM, Shakin JL: Ophthalmology 1984; 91:1384–1388.
Arch Ophthalmol 1967; 78:585–591. Management of macular puckers 39. Manschot WA: Persistent hyperplastic
7. Maumenee AE: Further advances in the associated with retinal angiomas. primary vitreous: special reference to
study of the macula. Arch Ophthalmol Ophthalmol Surg 1990; 21:550–556. preretinal glial tissue as a pathological
1967; 78:151–165. 23. Gass JDM: Stereoscopic atlas of macular characteristic and to the development of
8. Wise GN: Preretinal macular fibrosis (an disease. St Louis: Mosby; 1997. the primary vitreous. Arch Ophthalmol
analysis of 90 cases). Trans Ophthalmol 24. Augustin AJ, Spigtznas M, Koch F et al: 1958; 59:188.
Soc UK 1972; 92:131–140. Indicators of oxidative tissue damage 40. Wolter JR: Glia of human retina. Am J
9. Mitchell P, Smith W, Chey T et al: and inflammatory activity in epiretinal Ophthalmol 1959; 48:370.
Prevalence and associations of epiretinal membranes of proliferative diabetic 41. Smith TR: Pathologic findings after retina
membranes. The Blue Mountains Eye Study, retinopathy, proliferative vitreoretinopathy, surgery. In: Schepens C, ed. Importance
Australia. Ophthalmology 1997; 1022–1040. and macular pucker. Ger J Ophthalmol of the vitreous body in retina surgery with
10. Klein R, Klein BE, Wang Q et al: The 1995; 4:47–51. special emphasis on reoperations.
epidemiology of epiretinal membranes. 25. Charteris DG, Hiscott P, Grierson I et al: St Louis: Mosby; 1960.
Trans Am Ophthalmol Soc 1994; 92: Proliferative vitreoretinopathy. Lymphocytes 42. Kurz GH, Zimmerman LE: Vagaries of the
403–425. in epiretinal membranes. Ophthalmology retinal pigment epithelium. Int Ophthalmol
11. Fraser-Bell S, Guzowski M, Rochtchina E 1992; 99:1364–1367. Clin 1962; 2:441.
et al: Five-year cumulative incidence and 26. Kiryu J, Kita M, Tanabe T et al: Pars plana 43. Ashton N: Oxygen and the growth and
progression of epiretinal membranes: the vitrectomy for epiretinal membrane development of retinal vessesl: in vivo and
Blue Mountains Eye Study. Ophthalmology associated with sarcoidosis. Jpn J in vitro studies. The XX Francis I. Proctor
2003; 110:34–40. Ophthalmol 2003; 47:479–483. Lecture. Am J Ophthalmol 1966; 62:412.
12. Lyle DJ: Detachment of the internal limiting 27. McDonald HR, De Bustros S, Sipperley JO: 44. von Gloor BP: Mitotic activity in the
membrane of the retina (hyaloid Vitrectomy for epiretinal membrane with cortical vitreous cells (hyalocytes) after
membrane). Trans Am Ophthalmol Soc Candida chorioretinitis. Ophthalmology photocoagulation. Invest Ophthalmol 1969;
1934; 39:201–217. 1990; 97:466–469. 8:633
13. Appiah AP: Secondary causes of 28. Naoi N, Sawada A: Effect of vitrectomy on 45. Foos RY: Vitreoretinal juncture-Simple
premacular fibrosis. Ophthalmology 1989; epiretinal membranes after endogenous epiretinal membranes. Graefes Arch CLin
96:389–392. fungal endophthalmitis. Jpn J Ophthalmol Exp Ophthalmol 1974; 189:231–250.
14. Wise GN: Macular changes after venous 1996; 40:434–438. 46. Bellhorn MB, Friedman AH, Wise GN,
obstruction. Arch Ophthalmol 1957; 29. Repka MX, Green WR: Epiretinal Henkind P: Ultrastructure and
58:544–557. membrane: an unusual complication of clinicopathologic correlation of idiopathic
15. Tang S, Le-Ruppert KC, Gabel VP: childhood optic neuritis. J Pediatr preretinal macular fibrosis. Am J
Proliferation and activation of vascular Ophthalmol Strabismus 1996; 33:124–127. Ophthalmol 1975; 79:366–373.
endothelial cells in epiretinal membranes 30. Hagler WS, Aturaliya U: Macular puckers 47. Vinores SA, Campochiaro PA, Conway BP:
from patients with proliferative diabetic after retinal detachment surgery. Br J Ultrstructural and electron
retinopathy. An immunohistochemistry and Ophthalmol 1971; 55:451–457. immunocytochemical characterization of
clinical study. Ger J Ophthalmol 1994; 31. Michels RG, Gilbert HD: Surgical cells in epiretinal membranes. Invest
3:131–136. management of macular pucker after retinal Ophthalmol VIs Sci 1990; 31:14–28.
16. Barry PJ, Hiscott PS, Grierson I et al: reattachment surgery. Am J Ophthalmol 48. Smiddy W, Michels RG, Gilbert HD, Green
Reparative epiretinal fibrosis after diabetic 1979; 88:925–929. WR: Clinicopathologic study of idiopathic 2079
RETINA AND VITREOUS

macular pucker in children and young 69. Robertson DM, Buettner H: Pigmented 89. Feron EJ, Veckeneer M, Parys-van
adults. Retina 1992; 12:232–236. preretinal membranes. Am J Ophthalmol Ginderdeuren R et al: Trypan blue staining
49. Hui YN, Goodnight R, Zhang HJ et al: 1977; 83:824–829. of epiretinal membranes in proliferative
Glial epiretinal membranes and contraction. 70. Arroyo JG, Irvine AR: Retinal distortion and vitreoretinopathy. Arch Ophthalmol 2002;
Immunohistochemical and morphological cottonwool spots associated with epiretinal 120:141–144.
studies. Arch Ophthalmol 1988; 106: membrane contraction. Ophthalmology 90. Hillenkamp J, Saika P, Gora F et al:
1280–1285. 1995; 102:662–668. Macular function and morphology after
50. Campochiaro PA, Kaden IH, Vidaurri-Leal J, 71. Klein BR, Hiner CJ, Glaser BM et al: peeling of idiopathic epiretinal membrane
Glaser BM: Cryotherapy enhances Fundus photographic and fluorescein with and without the assistance of
intravitreal dispersion of viable retinal angiographic characteristics of indocyanine green. Br J Ophthalmol 2005;
pigment epithelial cells. Arch Ophthalmol pseudoholes of the macula in eyes with 89:437–443.
1985; 103:434–436. epiretinal membranes. Ophthalmology 91. Haritoglou C, Eibl K, Schaumberger M
51. Machemer R, Laqua H: Pigment epithelium 1995; 102:768–774. et al: Functional outcome after trypan
proliferation in retinal detachment (massive 72. Martinez JM, Smiddy WE, Kim J et al: blue-assisted vitrectomy for macular
preretinal proliferation). Am J Ophthalmol Differentiating macular holes from macular pucker: a prospective, randomized,
1975; 80:1–23. pseudoholes. Am J Ophthalmol 1994; comparative trial. Am J Ophthalmol
52. Machemer R, Van Horn D, Aaberg TM: 762–767. 2004; 138(1):1–5.
Pigment epithelial proliferation in human 73. Massin P, Paques M, Masri H et al: Visual 92. Rice TA, de Bustros S, Michels RG, et al:
retinal detachment with massive periretinal outcome of surgery for epiretinal Prognostic factors in vitrectomy for
proliferation. Am J Ophthalmol 1978; membranes with macular pseudoholes. epiretinal membranes of the macula.
85:181–191. Ophthalmology 1999; 106:580–585. Ophthalmology 1986; 93:602–610.
SECTION 10

53. Newsome DA, Rodrigues MM, 74. Smiddy WE, Gass JDM: Masquerades of 93. McDonald HR, Verre WP, Aaberg TM:
Machemer R: Human massive periretinal macular holes. Ophthalm Surg 1995; Surgical management of idiopathic
proliferation. In vitro characteristics of 26:16–24. epiretinal membranes. Ophthalmology
cellular components. Arch Ophthalmol 75. Johnson RN, Gass JDM: Idiopathic 1986; 93:978–983.
1981; 99:878–880. macular holes: observations, stages of 94. Michels RG: Vitreous surgery for macular
54. Wallo IHL, Miller SA: Preretinal membrane formation, and implications for surgical pucker. Am J Ophthalmol 1981;
by retinal pigment epithelium. Arch intervention. Ophthalmology 1988; 92:628–639.
Ophthalmol 1978; 96:1643–1646. 95:917–924. 95. Michels RG: A clinical and hisopathologic
55. Smiddy WE, Maguire AM, Green WR, et al: 76. Fine S: Idiopathic preretinal macular study of epiretinal membranes affection the
Idiopathic epiretinal membranes. fibrosis. Int Ophthalmol Clin 1977; macula and removed by vitreous surgery.
Ophthalmology 1989; 96:811–821. 17:183–189. Trans Am Ophthalmol Soc 1982;
56. Stern WH, Fisher SK, Anderson DH, et al: 77. Margherio RR, Trese MT, Margherio AR, 100:580–656.
Epiretinal membrane formation after Cartright K: Surgical management of 96. de Bustros S, Thompson JT, Michels RG
vitrectomy. Am J Ophthalmol 1982; vitreomacular traction syndromes. et al: Vitrectomy for idiopathic epiretinal
93:757–772. Ophthalmology 1989; 96:1437–1445. membranes causing macular pucker.
57. Daicker B, Guggenheim R: 78. Greven CM, Slusher MM, Weaver RG: Br J Ophthalmol 1988; 72:692–695.
Rasterelektronenmikroskopische Epiretinal membrane release and posterior 97. Margherio RR: Epiretinal membrane.
untersuchungen an fibrosen und fibro- vitreous detachment. Ophthalmology 1988; (Discussion). Ophthalmology 1984;
gliosen epiretinalen fibrplasien. Graefes 95:902–905. 91:1387–1388.
Arch Clin Exp Ophthalmol 1978; 79. Mulligan TG, Daily MJ: Spontaneous 98. von Gunten S, Pournaras CJ, de Gottrau P,
207:229–242. peeling of an idiopathic epiretinal Brazitikos P: Facteurs prognostiques du
58. Trese MT, Chandler DB, Machemer R: membrane in a young patient. Arch traitement chirurgical des membranes
Macular pucker II. Ultrastructure. Graefes Ophthalmol 1992; 110:1367–1368. epiretiniennes. Klin Monatsbl Augenheilkd
Arch Clin Exp Ophthalmol 1983; 221:16–26. 80. Noble KG, Carr RE: Idiopathic preretinal 1994; 204:309–312.
59. Scudder JM, Eifrig DE: Spontaneous gliosis. Ophthalmology 1982; 89:521–523. 99. Margherio RR, Cox MS Jr, Trese MT et al:
surface wrinkling retinopathy. Ann 81. Reeser FH, Aaberg TM: Vitreous humor. Removal of epimacular membranes.
Ophthalmol 1975; 7:333–341. In: Records RE, ed. Physiology of the Ophthalmology 1985; 92:1075–1083.
60. Sidd RJ, Fine Sl, Owens SL et al: Idiopathic human eye and visual system. Hagerstown, 100. Pesin SR, Olk RJ, Grand MG et al:
preretinal gliosis. Am J Ophthalmol 1982; MD: Harper and Row; 1979. Vitrectomy for premacular fibroplasia.
94:44. 82. Johnston RL: Modification of the Tano Prognostic factors, long-term follow-up,
61. Spitznas M, Leuenberger R: Die primare diamond dusted scraper. Retina 2000; and time course of visual improvement.
epiretinale gliose. Klin Monatsbl Augeneilkd 20:427. Ophthalmology 1991; 98:1109–1114.
1977; 171:410. 83. Kuhn F, Mester V, Berta A: The Tano 101. Maguire AM, Margherio RR, Dmuchowski C:
62. Trese MT, Chandler DB, Machemer R: diamond dusted membrane scraper: Preoperative flourescein angiographic
Macular pucker. Prognostic criteria. indications and contraindications. features of surgically removed idiopathic
Graefes Arch Clin Exp Ophthalmol 1983; Acta Ophthalmol Scand 1998; epiretinal membranes. Retina 1994;
221:12–15. 76:754–755. 14:411–416.
63. Hirokawa H, Jalhk AE, Takahashi M et al: 84. Sakamoto H, Yamanaka I, Kubota T, et al: 102. Ma SS, Barloon S, Maberley AL, Kestle J:
Role of the vitreous in idiopathic preretinal Indocyanine green assisted peeling of the Effect of macular edema on surgical visual
macular fibrosis. Am J Ophthalmol 1986; epiretinal membrane in proliferative outcome in eyes with idiopathic epiretinal
101:166–169. vitreoretinopathy. Graefes Arch Clin Exp membrane. Can J Ophthalmol 1996;
64. Kuriyama S, Matsumura M, Harada T et al: Ophthalmol 2003; 241: 204–207. 31:183–186.
Surgical techniques and reattachments 85. Perrier M, Sebag M: Epiretinal membrane 103. Sivalingam A, Eagle RC Jr, Duker JS, et al:
rates in retinal detachment due to surgery assisted by trypan blue. Am J Visual prognosis correlated with the
macular hole. Arch Ophthalmol 1990; Ophthalmol 2003; 135:909–911. presence of internal-limiting membrane in
1089:1559. 86. Fraser EA, Cheema RA, Roberts MA: histiophathologic specimens obtained from
65. Wise GN: Clinical features of idiopathic Triamcinolone acetonide-assisted peeling epiretinal membrane surgery.
preretinal macular fibrosis. Am J of retinal internal limiting membrane for Ophthalmology 1990; 97:1549–1552.
Ophthalmol 1975; 79:349. macular surgery. Retina 2003; 23:883–884. 104. Kampik A, Green WR, Michels RG,
66. Wiznia RA: Natural history of idiopathic 87. Kimura H, Kuroda S, Nagata M: Nase PK: Ultrastructureal features of
preretinal macular fibrosis. Ann Ophthalmol Triamcinolone acetonide-assisted peeling progressive idiopahtic epiretinal
1982; 14:876–878. of the internal limiting membrane. Am J membrane removed by vitreous
67. Dellaporta A: Macular pucker and Ophthalmol 2004; 137:172–173. surgery. Am J Ophthalmol 1980;
peripheral retinal lesions. Trans Am 88. Burk SE, Da Mata AP, Snyder ME et al: 90:797–809.
Ophthalmol Soc 1973; 71:329–340. Indocyanine green assisted peeling of the 105. Smiddy WE, Maguire AM, Green WR, et al:
68. Laqua H: Pigmented macular pucker. Am J retinal internal limiting membrane. Idiopathic epiretinal membranes.
2080 Ophthalmol 1978; 86:56–58. Ophthalmology 2000; 107:2010–2014. Ophthalmology 1989; 96:811–821.
Macular Epiretinal Membranes

106. Park DW, Dugel PU, Garda J et al: Macular detachment. Arch Ophthalmol 1988; membrane peel surgery. Br J Ophthalmol
pucker removal with and without internal 106:758–760. 2006; 90: 559–562.
limiting membrane peeling : pilot study. 109. Poliner LS, Olk RJ, Grand MG et al: 112. de Bustros S, Thompson JT, Michels RG
Ophthalmology 2003; 110: 62–64. The surgical management of premacular et al: Nuclear sclerosis after vitrectomy for
107. Council MD, Shah GK, Lee HC et al: Visual fibroplasia. Arch Ophthalmol 1988; idiopathic epiretinal membrane. Am J
outcomes and complications of epiretinal 106:761–764. Ophthalmol 1988; 105:160–164.
membrane removal secondary to 110. Thompson JT: Epiretinal membrane 113. McDonald HR, Johnson RN, Ai E, et al:
rhegmatogenous retinal detachment. removal in eyes with good visual acuities. Macular epiretinal membrane. In: Ryan SJ,
Ophthalmology 2005; 112: 1218–1221. Retina 2005; 25: 875–882. ed. Retina 4th edn. Philadelphia, PA
108. de Bustros S, Rice TA, Michels RG et al: 111. Ghazi-Nouri SM, Tranos PG, Rubin GS Mosby; 2006.
Vitrectomy for macular pucker after et al: Visual function and quality of life
treatment of retinal tears or retinal following vitrectomy and epiretinal

CHAPTER 160

2081
CHAPTER

161 Acute Macular Neuroretinopathy


Lucy H. Y. Young and Donald J. D’Amico

Acute macular neuroretinopathy was first described in 1975 by When patients are tested with the Amsler grid, they can
Bos and Deutman.1 In their six patients, this entity was charac- usually outline parafoveal negative scotomas, frequently in the
terized by a sudden onset of paracentral scotomas and the shape of teardrops, corresponding exactly with the fundus
presence of cloverleaf, wedge-shaped, darkish red lesions centered lesions (Fig. 161.1).9,11–13,16 In patients whose fundus lesions are
around the fovea. Only a small number of cases have been less prominent, examination with red-free light may be useful
added subsequently. in highlighting the lesions. As mentioned by Guzak and
Characteristically, visual acuity remains normal or is slightly colleagues,4 this enhancement is probably related to the color of
reduced.1–22 The chief complaint is usually related to the sudden the lesion rather than to the surface changes in the retina.
appearance of paracentral scotomas. This rare macular disease Although Bos and Deutman1 suggested that the fundus lesions
can occur unilaterally or bilaterally, and the majority of patients seen in their six patients were located in the inner retina,
are young adults, mostly women. Many of these patients are thus the name acute macular neuroretinopathy, subsequent
taking birth control pills.1,3–5,11–15,17–20 The visual disturbance is reports relying on biomicroscopic examination, stereophoto-
often preceded by a bout of a flu-like syndrome, such as influenza, graphs, fluorescein angiography, and electrodiagnostic testing
pharyngitis, or enteritis.1,4,5,9,11,12,14,19–22 indicate that the acute macular neuroretinopathy lesions are

a b

c d e

FIGURE 161.1. This 42-year-old woman presented with a 3-week history of a flu-like syndrome before the onset of paracentral scotomas in the
left eye. Her past medical history was significant, for she had undergone hysterectomy and oophorectomy 2 years previously and had since
been using estradiol transdermal (Estroderm) patch. Her visual acuity was 20/20 in the left eye. (a) Note the darkish red lesions centered around
the fovea. (b) These lesions are better appreciated with red-free light. (c and d) Angiography results were normal. (e) Amsler grid testing showed
two paracentral scotomas.
(a–e) Courtesy of Jack F. Bowers, MD. 2083
RETINA AND VITREOUS

located in the outer retinal layers.9,11,13 Scanning laser ophthal- fosfestrol (an estrogen preparation) for a prostatic carcinoma at
moscopy has been shown to be useful in enhancing the visibility the time of the onset of visual symptoms.6
of the macular lesions.17,18 In addition to the deep, reddish O’Brien and co-workers8 reported three patients in whom
brown lesions, a few small, superficial retinal hemorrhages may acute macular neuroretinopathy developed after acute hyper-
be present in the macula.2,9 The retinal vasculature, nerve fiber tension caused by intravenous sympathomimetics. Guzak and
layer, retinal pigment epithelium, and optic disk are normal, colleagues4 reported two cases, and both of their patients experi-
and there are no vitreous cells. In symptomatic patients with enced macular neuroretinopathy after intravenous injections of
normal fundus examination, it is important to bear in mind epinephrine for adverse reactions to contrast agents. However,
that the onset of visual symptoms of acute macular neuro- blood pressure readings were not reported in these two cases.
retinopathy may occur few days before retinal lesions can be The temporal relationship between the development of acute
visualized and thus close observation can be helpful.19 macular neuroretinopathy and the use of intravenous epine-
Fluorescein angiography studies are essentially normal. phrine suggests an association with either the hypertensive epi-
Questionably dilated perimacular capillaries without leakage sodes or the sympathomimetics themselves. However, this would
and subtle hypofluorescence corresponding to the fundus lesions not explain the other cases reported to date. Acute macular
have been reported.1,2,9 Similarly, no abnormalities are observed neuroretinopathy has also been reported in two patients treated
with indocyanine green angiography.20 In two case reports, with epinephrine for acute shock.23 In another case report
optical coherence tomography (OCT) demonstrated a band of associated with epinephrine administration, multiple blood
hyperreflectivity at the outer retinal layer.21,22 pressure recordings were taken and they were all normal.16
SECTION 10

Electrical testing, such as electroretinogram and electroocu- Gass and Hamed3 reported acute macular neuroretinopathy
logram studies, of patients afflicted with acute macular neuro- and multiple evanescent white dot syndrome occurring in the
retinopathy is normal.1,7,9,12 This is not unexpected because same patients. They compared the clinical features of acute
both tests measure mass physiologic responses and are not macular neuroretinopathy and multiple evanescent white dot
sensitive to pathologic processes confined to the macula alone. syndrome and suggested that these two rare syndromes may be
To address this issue, Sieving and associates13 recorded the early related pathogenetically and causally.
retinal potential responses from the posterior fundi in one The milder forms of acute macular neuroretinopathy, i.e.,
patient with unilateral acute macular neuroretinopathy. The when the characteristic macular lesions are not prominent, may
early retinal potential is generated in the photoreceptor outer be mistaken for acute retinal pigment epitheliitis24–27 because
segments, and by restricting the recording to the posterior both entities cause loss of central vision in young adults.
fundus, the investigators made the recording more sensitive to Table 161.1 lists the differential diagnostic features of these two
localized lesions in the macula. They found a reduced ampli- entities. The peculiar macular lesions may also be mistaken for
tude in the affected eye when compared with the normal fellow subretinal hemorrhage; this is probably what happened in the
eye. This recording was done 13 months after the initial onset case reported by Weinberg and Nerney.15 No recurrence of this
of visual disturbances. A follow-up early retinal potential entity has been reported.
measurement 7 months later, or 20 months after the onset of
symptoms, showed similar amplitude reduction. Their findings
TABLE 161.1 Differential Diagnostic Features of Pigment
would suggest that the abnormality is located in the outer Epitheliitis and Acute Macular Neuroretinopathy
segments. Multifocal electroretinogram (ERG) has been shown
to be more useful in the evaluation of acute macular neuro- Feature Pigment Acute Macular
retinopathy and its findings support an outer retinal layer Epitheliitis24–27 Neuroretinopathy
involvement.20–22 Ophthalmoscopy Clusters of round, Red-brown
Although acute macular neuroretinopathy is associated with dark lesions wedges
minimal, if any, decline in visual acuity, patients with this surrounded by
disorder rarely report improvement of symptoms. The para- hypopigmented halos
central scotomas may become less dense but usually persist Lesion distribution Macular Macular
despite resolution of the fundus lesions. Most of the follow-up
examinations reported to date range from months to a few years Location Retinal pigment Sensory retina
epithelium
only.1,2,4–6,9,11–16,22 However, one patient5 with bilateral involve-
ment was followed up for 9 years. This patient had resolution Laterality Usually unilateral Either
of paracentral scotomas in the right eye but persistence of a Associated vitritis None None
large scotoma in the left eye 2 years later. During the following
7 years, the large paracentral scotoma remained but slowly Diminished visual Moderate Mild
acuity
became smaller.
Both the histopathologic features and the pathogenesis of Sex predilection None Female
these rare macular lesions are unknown. Although a large Age of onset Young adults Young adults
number of reported patients experienced a flu-like syndrome
before the onset of their visual symptoms, a relationship to viral Associated None Often preceded
infection has not yet been established. It has also been sug- by a flu-like
systemic
gested that there is an association with oral contraceptives infection
because the condition affects predominantly healthy young syndrome
women, many of whom are taking this medication. However,
since the majority of women with this condition are of child- Visual recovery Complete in Minimal
7–10 weeks
bearing age, it is highly likely that the use of birth control pills
is merely coincidental; in addition, cases have been reported Cause Unknown Unknown
without birth control pill use. Furthermore, acute macular Fluorescein Normal or halos of Normal
neuroretinopathy has been reported in male patients, although angiography hyperfluorescence
it is interesting to note that one of them was being treated with
2084
Acute Macular Neuroretinopathy

REFERENCES
1. Bos PJM, Deutman AF: Acute macular 10. Putteman A, Toussaint D, Deutman AF: 19. Douglas IS, Cockburn DM: Acute macular
neuroretinopathy. Am J Ophthalmol 1975; Neuroretinopathie maculaire aigue. Bull neuroretinopathy. Clin Exp Optom 2003;
80:573. Soc Belge Ophtalmol 1982; 199-200:35. 2:121.
2. Gass JDM: Stereoscopic atlas of macular 11. Rait JL, O’Day J: Acute macular 20. Amin P, Cox TA: Acute macular
diseases: diagnosis and treatment. 2nd edn. neuroretinopathy. Aust N Z J Ophthalmol neuroretinopathy. Arch Ophthalmol 1998;
St Louis: CV Mosby; 1977:304. 1987; 15:337. 116:112.
3. Gass JDM, Hamed LM: Acute macular 12. Rush JA: Acute macular neuroretinopathy. 21. Feigl B, Haas A: Optical coherence
neuroretinopathy and multiple evanescent Am J Ophthalmol 1977; 83:490. tomography (OCG) in acute macular
white dot syndrome occurring in the same 13. Sieving PA, Fishman GA, Salzano T, neuroretinopathy. Acta Ophthalmol Scand
patients. Arch Ophthalmol 1989; 107:189. Rabb MF: Acute macular neuroretinopathy: 2000; 78:714.
4. Guzak SV, Kalina RE, Chenoweth RE: early receptor potential change suggests 22. Chan WM, Liu DTL, Tong JP, et al:
Acute macular neuroretinopathy following photoreceptor pathology. Br J Ophthalmol Longitudinal findings of acute macular
adverse reaction to intravenous contrast 1984; 68:229. neuroretinopathy with multifocal
media. Retina 1983; 3:312. 14. van Herck M, Leys A, Missotten L: Acute electroretinogram and optical coherence
5. Miller MH, Spalton DJ, Fitzke FW, Bird AC: macular neuroretinopathy. Bull Soc Belge tomography. Clin Exp Ophthalmol 2005;
Acute macular neuroretinopathy. Ophtalmol 1984; 210:119. 33:439.
Ophthalmology 1989; 96:265. 15. Weinberg RJ, Nerney JJ: Bilateral 23. Leys M, Van Slycken S, Koller J, Van de
6. Nagasawa N, Hommura S: A case of acute submacular hemorrhages associated with Sompel W: Acute macular neuroretinopathy

CHAPTER 161
macular neuroretinopathy: an optical an influenza syndrome. Ann Ophthalmol after shock. Bull Soc Belge Ophtalmol
consideration on the peculiar features of 1983; 15:710. 1991; 241:95.
fundus oculi. Acta Soc Ophthalmol Jpn 16. Desai UR, Sudhamathi K: Intravenous 24. Krill AE, Deutman AF: Acute retinal pigment
1982; 86:2044. epinephrine and acute macular epitheliitis. Am J Ophthalmol 1972; 74:193.
7. Neetens A, Burvenich H: Presumed neuroretinopathy. Arch Ophthalmol 1993; 25. Deutman AF: Acute retinal pigment
inflammatory maculopathies. Trans 111:1206. epitheliitis. Am J Ophthalmol 1974; 78:571.
Ophthalmol Soc UK 1978; 98:160. 17. Tingsgaard LK, Sander B, Larsen M: 26. Friedman MW: Bilateral recurrent acute
8. O’Brien DM, Farmer SG, Kalina RE, Leon JA: Enhanced visualization of acute macular retinal pigment epitheliitis. Am J
Acute macular neuroretinopathy following neuroretinopathy by spectral imaging. Acta Ophthalmol 1975; 79:567.
intravenous sympathomimetics. Retina Ophthalmol Scand 1999; 77:592. 27. Eifrig DE, Knobloch WH, Moran JA: Retinal
1989; 9:281. 18. Gandorfer A, Ulbig MW: Scanning laser pigment epitheliitis. Ann Ophthalmol 1977;
9. Priluck JA, Buettner H, Robertson DM: ophthalmoscope findings in acute macular 9:639.
Acute macular neuroretinopathy. Am J neuroretinopathy. Am J Ophthalmol 2002;
Ophthalmol 1978; 86:775. 133:413.

2085
CHAPTER

162 Toxoplasmosis
Neal P. Barney, Timothy J. Daley, and C. Stephen Foster

Toxoplasmosis is one of the leading causes of infectious retinitis.1 Key Features


It is produced by the coccidian parasite Toxoplasma gondii, and 1. The majority of cases of ocular toxoplasmosis are congenital.
the cat is its definitive host. Humans and other animals act as 2. The diagnosis is based on clinical findings.
intermediate hosts. Toxoplasmosis infection is particularly 3. The definitive diagnosis requires demonstration of tachyzoites
serious and potentially fatal in congenital disease as well as in in ocular tissues
immunocompromised patients. 4. Numerous treatment protocols have been used with success.
T. gondii is ubiquitous in nature. The parasite has three forms: Most utilize two antimicrobial drugs.
tachyzoite, bradyzoite, and sporozoite.2 The tachyzoite is the 5. Complications include:
infectious form of the parasite; it multiplies intracellularly and • Choroidal neovascularization
causes host cell death and the release of more tachyzoites. The • Retinochoroidal vascular anastamosis
host’s immune response causes the tachyzoite to transform into • Branch retinal vein occlusion
the slowly dividing bradyzoite, the encysted intracellular form • Cystoid macular edema
of the parasite found in tissue cysts. These cysts may remain in • Retinal detachment
any tissue, such as the retina, for years without provoking an • Cataracts
immune response in the host. When the cysts rupture, parasites • Glaucoma (most common complication)
are released into the surrounding tissues, resulting in a recur-
rence of clinical disease. The sporozoite forms from oocysts,
which are produced exclusively in the intestinal enterocytes of reported in infants are microphthalmia, enophthalmos, ptosis,
cats. Millions of oocysts are released into the environment for nystagmus, choroidal colobomas, and strabismus.7
2–3 weeks after primary infection until the cat becomes Several reports of acquired ocular toxoplasmosis suggest that
immune.3 The oocysts undergo sporulation within a few days this route may actually be more important and more prevalent
and may remain infectious for up to 2 years. than previously thought.8–11 A population study in southern
Human infection may occur by either the congenital or the Brazil reported a high prevalence of acquired ocular toxoplas-
acquired route. The acquired disease can occur by the ingestion mosis in subjects older than 12 years.12 Similarly, a study after
of either sporozoites or tissue cysts in inappropriately cooked an outbreak of toxoplasmosis on Victoria Island, in British
infected meat and is usually asymptomatic in immuno- Columbia, Canada reported that 21% of confirmed cases of new
competent persons. Infected livestock are a prominent source of infection had ocular disease.13
infection for humans. Recent evidence also suggests the disease Ocular toxoplasmosis frequently presents as a focal necro-
can be acquired through inhalation of spores and ingestion of tizing retinitis, usually adjacent to a large, atrophic chorio-
contaminated drinking water. Infection can also be acquired retinal scar (Fig. 162.1), which is often located in the macula
through contaminated blood transfusions and organ trans- in congenital cases. The scar is typically yellow-white and
plants.4 Congenital infection occurs through transplacental unevenly pigmented. Acquired cases of toxoplasmosis may
transmission of tachyzoites from a mother infected just before present as a unilateral focal chorioretinitis with no preexisting
or during pregnancy to the developing fetus.5 The severity of retinal scars in either eye.14 The areas of retinitis are the result
congenital infection is highest when acquired during the first of tissue cysts bursting and releasing bradyzoites that transform
trimester of pregnancy, although the frequency of transmission into tachyzoites, which in turn invade neighboring cells. These
to the fetus is greatest during the third trimester when contact destructive lesions are usually larger than one disk diameter
of the maternal and fetal circulations is more likely to occur. and appear as soft, white, fluffy infiltrates surrounded by retinal
Once maternal immunity has developed, it is believed that all edema with subjacent choroiditis.15 When the tachyzoites come
future fetuses are protected from the development of congenital under increasing attack by the host’s immune response, they
toxoplasmosis. gradually transform back into bradyzoites. Inflammatory
cells will be found in the vitreous overlying the active lesion
CLINICAL FEATURES (Fig. 162.2). Perivascular inflammatory exudates are frequently
present around retinal vessels peripheral to an area of active
The majority of cases of ocular toxoplasmosis are congenital. In inflammation. In cases where the retinal vessel transverses the
one study of 300 cases of congenital toxoplasmosis in newborns, active toxoplasmic lesion, regression of the vascular infiltrates
ocular lesions were present in 76%, neurologic involvement was may take longer to recede or may even fail to disappear.16
present in 51%, intracranial calcifications were present in 32%, Patients will often complain of pain, blurred vision, floaters,
and microcephaly or hydrocephalus was present in 26%.6 Among and photophobia. More than a third of patients with active
the clinical manifestations of congenital ocular toxoplasmosis ocular toxoplasmosis will have macular involvement resulting 2087
SECTION 10 RETINA AND VITREOUS

FIGURE 162.3. Toxoplasma retinitis without typical choroidal


involvement. Note the multifocal nature of the lesions, with an area of
active retinitis at the 11 o’clock position. Note also the associated
vasculitis.

FIGURE 162.1. Atrophic chorioretinal scar with surrounding retinal


pigment epithelial proliferation; a chorioretinal scar quite typical of
Toxoplasma chorioretinitis. However, note also the fresh satellite lesion
at the edge of the pigmented chorioretinal scar (at the 11 o’clock
position) threatening the fovea.

FIGURE 162.4. Toxoplasma papillitis with vitreal inflammatory cells


anterior to the optic nerve, making visualization of the optic nerve
slightly difficult. The ELISA for Toxoplasma antibodies gave the
following results: The IgG titer was 1:496; the IgM titer at the time this
photograph was taken was 1:64; 2 weeks later, the anti-Toxoplasma
IgM antibody titer was 1:256.

FIGURE 162.2. Multiple atrophic and hyperpigmented chorioretinal


scars from previous toxoplasmosis. Note the fresh area of COMPLICATIONS
chorioretinitis with overlying vitreal inflammatory cells and vitreal
‘haze’. Secondary complications arising from ocular toxoplasmosis
include choroidal neovascularization,24 retinochoroidal vascular
anastomosis,25 and branch retinal artery26 and vein occlusion,27,28
in severe visual loss.17,18 Ocular toxoplasmosis may also present cystoid macular edema, retinal detachment, cataracts, secondary
as gray-white punctate lesions in the outer retina and retinal glaucoma, and optic atrophy.17 The most frequent complication
pigment epithelium (Fig. 162.3).19 Occasionally, patients will is secondary glaucoma. There is a well-known association
present initially with severe unilateral papillitis, macular hard between Fuchs’ heterochromic iridocyclitis and ocular toxo-
exudates distributed in a star fashion, and vitreal inflammation plasmosis, but the reasons for this are unclear.29
(Fig. 162.4).20,21 Abnormal intraocular pressure has also been
associated with active disease.22 Active toxoplasmosis HISTOPATHOLOGIC AND IMMUNOLOGIC
simultaneously involving both the retina and the optic nerve is FEATURES
unusual but has been documented clinically in an immuno-
competent patient using magnetic resonance imaging.21 There Histopathologically, there is necrosis of the involved retina
has also been a report of active ocular toxoplasmosis presenting with destruction of the retinal architecture and the underlying
as hemorrhagic retinochoroiditis in an otherwise healthy choroid.30 Since the parasite has a propensity for attacking
2088 adult.23 neural tissue, tissue cysts and trophozoites are usually found in
Toxoplasmosis

the superficial layers of the retina within the area of necrosis necessary for the resolution of active disease, antibody may be
(Figs 162.5 and 162.6). The infiltrate consists predominantly of important in establishing a state of immunity in the host.37
lymphocytes, macrophages, and epitheloid cells, with plasma Antibodies directed against the cytoplasmic protein F3G338 can
cells found in the periphery of the lesion. protect passively immunized mice from a lethal challenge of
Cell-mediated immunity is felt to be the major defense T. gondii.39
mechanism against Toxoplasma infection.31 Several purified
antigens from tachyzoites have been identified, including the OCULAR TOXOPLASMOSIS IN THE
major surface protein antigen p30, which can participate IMMUNOCOMPROMISED HOST
in antibody-dependent complement-mediated lysis of the
tachyzoite.32,33 In patients with ocular toxoplasmosis, the Although T. gondii is a common opportunistic pathogen with a
cellular immune response appears to be directed predominantly penchant for attacking the central nervous system in immuno-
against the cell surface protein p22.34 Additionally, gamma compromised patients,40–42 including those with acquired
interferon and tumor necrosis factor both appear to play an immunodeficiency syndrome (AIDS),43–45 ocular toxoplasmosis
important role in inducing and maintaining the interconversion appears to be uncommon. Ocular toxoplasmosis occurs in ~1%
of the organism’s tachyzoite form to the bradyzoite form during of patients with AIDS.46 Systemically, these patients are prone
active infection.35 When neutralizing doses of antibodies to to the development of Toxoplasma encephalitis, myocarditis, or
either gamma interferon or tumor necrosis factor are given to pneumonitis.
animal models with chronic infection, free tachyzoites and an AIDS-related Toxoplasma retinochoroiditis has several

CHAPTER 162
increased number of cysts are observed.36 However, some evi- atypical clinical manifestations, including bilateral involvement
dence also indicates that part of the disease may be mediated by and single or multifocal discrete lesions47–49 or diffuse areas of
an autoimmune mechanism directed against certain retinal retinal necrosis (Fig. 162.7).50 Furthermore, unlike immuno-
antigens. competent individuals, who rarely have more than one focus of
The role of the humoral response to toxoplasmosis is unclear. active retinal disease, even if there are multiple retinochoroidal
Whereas an intact cellular component of the immune system is scars, immunocompromised individuals can have multiple
active lesions in one or both eyes.51 Vascular sheathing may be
present, and prominent inflammatory reactions in the vitreous
and anterior chambers are common (Fig. 162.8). Lesions may
occur adjacent to retinal blood vessels and are usually not asso-
ciated with preexisting retinochoroidal scars. These findings
suggest that the ocular lesions in AIDS patients may be the
result of newly acquired disease or from organisms that have
disseminated to the eye from extraocular sites. The retina can
be infected simultaneously by both T. gondii and cytomegalo-
virus in patients with AIDS.52,53 Anti-Toxoplasma immuno-
globulin G (IgG) antibodies are present in these patients,
although IgM antibodies against Toxoplasma are uncommon.
Infection usually produces a full-thickness retinal necrosis,
but early necrosis may be confined to either the inner or the
outer layers. Large numbers of trophozoites and cysts with
scanty inflammatory reaction are seen within the necrotic
retina and optic nerve head. Although T. gondii organisms may
involve anterior segment structures and uveal tissue in patients
with AIDS, parasites are rarely found in these structures in
FIGURE 162.5. Toxoplasma feline retinochoroiditis. Histopathologic
appearance of the retina and choroid. Note the Toxoplasma cysts
(arrowheads).

FIGURE 162.7. Diffuse retinitis with probable underlying choroiditis in


FIGURE 162.6. Same specimen as in Figure 162.5. Note the an immunocompromised patient with toxoplasmosis. Note the
Toxoplasma cyst (arrowhead). overlying vitreal inflammatory cellular response with vitreal ‘haze’. 2089
RETINA AND VITREOUS

the serum level of anti-Toxoplasma titers and the activity of the


ocular disease in recurrent ocular toxoplasmosis.59
The most widely used serologic methods for detecting anti-
Toxoplasma antibodies are the Sabin–Feldman test, the
complement fixation test, the agglutination tests, the indirect
immunofluorescence assay (IFA), and the enzyme-linked
immunosorbent assay (ELISA). Although the Sabin–Feldman
dye test represents the standard against which all other tests are
measured, it is no longer performed routinely because it requires
the constant maintenance of virulent organisms in the
laboratory with the associated risks to laboratory personnel.
The IFA test, which has largely replaced the Sabin–Feldman
dye test, has become the most widely used test for the detection
of anti-Toxoplasma IgG or IgM antibodies.60 Although it is easy
to perform, the interpretations of its results can be misleading.
False-negative IgM titers may occur because of competitive
inhibition by anti-Toxoplasma IgG if present simultaneously.61
False-positive IgM titers can occur because of the presence of
SECTION 10

rheumatoid factor.62 Rheumatoid factor, an anti-IgG IgM


FIGURE 162.8. Fluorescein angiogram in a patient with active antibody, can bind to specific anti-Toxoplasma IgG. Finally, the
toxoplasmosis. Note not only the dye accumulation in the two foci presence of antinuclear antibody may produce false-positive
representing the areas of active Toxoplasma chorioretinitis but also the IgM and IgG titers.63 This occurs because Toxoplasma organ-
papillitis with dye staining of the nerve head and the associated retinal isms possess antigens that are indistinguishable from those
vasculitis with late vascular staining. found in human leukocyte nuclei using immunologic laboratory
testing techniques. Because IgM titers in ocular toxoplasmosis
otherwise healthy patients.54 Toxoplasma retinochoroiditis may become significant only in cases of acquired retinochoroiditis,9,64
rarely progress to panophthalmitis and orbital cellulitis in the specificity limitations of the IFA tests are restricted, for
patients with AIDS.55 Finally, not only do patients with AIDS practical purposes, to false-positive IgG titers produced by the
develop more extensive disease, but lesions are more likely to presence of antinuclear antibodies.
reactivate if treatment is discontinued.56 Commercially available IFA tests for Toxoplasma begin with
a 1:16 serum dilution because specificity decreases when more
DIAGNOSIS concentrated dilutions are used. Since any titer of antibody is
considered significant in establishing the diagnosis of
The definitive diagnosis of ocular toxoplasmosis requires the Toxoplasma retinochoroiditis, it is logical to expect a significant
demonstration of tachyzoites in ocular tissues.57 Cytologic percentage of false-negative titers using this serologic method.
evaluation of vitrectomy specimens to detect the presence of One report cited three documented cases of Toxoplasma
Toxoplasma organisms is especially useful in patients with retinochoroiditis with positive Sabin–Feldman dye titers and
necrotizing retinitis where the cause for the retinitis is negative immunofluorescent titers (titer <1:16).65
uncertain.58 The presence of cysts in tissue samples, however, The double-sandwich ELISA for anti-Toxoplasma antibodies
only suggests, but does not prove acute infection. Toxoplasma eliminates IgM specificity problems associated with the IFA
retinochoroiditis is, therefore, usually a clinical diagnosis based test.66,67 False-positive reactions resulting from rheumatoid
on a compatible lesion in the fundus and positive serologic factor and false-negative reactions because of competing IgG are
results for anti-Toxoplasma antibodies. Additionally, other causes avoided because only IgM of the test serum adheres to plates
of focal exudative retinitis, including syphilis, tuberculosis, precoated with anti-IgM antiserum. The ELISA for anti-
sarcoidosis, cytomegalovirus, and fungal infections, must be Toxoplasma antibodies is as specific and sensitive as the
excluded. Sabin–Feldman dye test. Therefore, in cases where ocular toxo-
Because of their ease of performance and their relatively high plasmosis is strongly suspected despite negative immunofluo-
sensitivity and specificity, serologic methods are often preferred rescent titers, a Sabin-Feldman titer or an ELISA titer should be
for the diagnosis of toxoplasmosis. Serologic diagnosis is com- obtained before excluding the diagnosis of toxoplasmosis.
plicated by the high prevalence of T. gondii-specific antibodies Some experts advocate measuring anti-Toxoplasma antibody
in the human population persisting for years and therefore titers in intraocular fluids,68 or even tears,69 but this has not
frequently reflecting past infection. Classic serodiagnosis of an been widely adopted at the present time. Polymerase chain
acute systemic infection requires the demonstration of a reaction (PCR) techniques have been used to detect the presence
seroconversion, a significant rise in specific antibody titers in of toxoplasma genes in ocular tissues and fluids.70,71 However,
paired sera taken 4–6 weeks apart, or the presence of anti- its precise role in the diagnosis of ocular toxoplasmosis remains
Toxoplasma IgM antibody in a single serum sample. If to be defined by controlled clinical trials. It is quite expensive,
retinochoroiditis develops subsequently, it is presumed to be not widely available, and requires special equipment and highly
acquired. However, because systemic toxoplasmosis infection is specialized technical skills to perform properly. Although, with
often subclinical in its manifestations, the diagnosis of acquired the advent of real-time PCR and its advantages of higher sensi-
ocular toxoplasmosis may be missed or confused with tivity, improved quantitation, and fewer manual manipulations
reactivation of the congenital type if ocular involvement occurs required, it could prove to be useful in determining disease
months or years after a systemic infection. Because sera are burden, therapy efficacy, and epidemiology of the disease.72
often submitted late after infection, it is difficult to detect an
antibody titer rise. Establishing the presence of Toxoplasma THERAPY
antibodies in a patient’s serum is considered essential in the
diagnosis of Toxoplasma retinochoroiditis. In this case, any titer Since available agents against T. gondii are ineffective against
2090 of antibody is significant because no correlation exists between tissue cysts, the major aim of therapy has been to stop the
Toxoplasmosis

multiplication of tachyzoites during episodes of active retino-


TABLE 162.1. Recommended Therapy for Ocular
choroiditis. It is important for the clinician to realize this
Toxoplasmosis
and to inform the patient that drug therapy does not totally
eradicate the parasite. Clinical Features Recommended Treatment
Complicating matters is that little is known about the
Peripheral lesion: Clindamycin, 300 mg PO tid or qid
natural history of active disease. There have been surprisingly mild to moderate and
few prospective, randomized, placebo-controlled clinical trials vitreal cells sulfadiazine, 1 g initially then
for the treatment of ocular toxoplasmosis in immunocompetent 500 mg PO qid
individuals. Interestingly, none of them demonstrated that
Peripheral lesion: As above
short-term drug therapy was effective in treating active toxo- moderate to severe plus
plasmic retinochoroiditis, reducing recurrent disease, or vitreal cells prednisone 1 mg/kg PO/day with
shortening duration of disease.73 taper based on clinical response
This is in contrast to numerous animal studies, which have
Juxtamacular lesion Clindamycin and sulfadiazine as above
found antimicrobial agents to be highly efficacious for active plus
toxoplasmosis treatment. In fact, some newer drugs such as pyrimethamine, 50 mg bid during the
atovaquone and azithromycin, have even been shown to reduce first day then 25 mg PO/day
the number of tissue cysts in animal models.74 Furthermore, plus
while treatment efficacy has been difficult to determine in folinic acid, 5 mg PO three times a week

CHAPTER 162
prednisone use based on vitreal cells,
immunocompetent patients, antimicrobial therapy can be highly
as in the case of peripheral lesions
effective in patients with AIDS. Long-standing active infections
rapidly become inactive with treatment in these individuals.
Finally, observational experience suggests that therapy is
beneficial in immunocompetent patients, despite difficulties to
quantify their efficacy. Most clinicians agree that antimicrobial toxoplasmosis has not been ruled out. Several cases of
therapy is warranted in active disease. fulminant ocular toxoplasmosis have been described after the
Toxoplasma organisms lack the transmembrane transport use of corticosteroids alone.79,80 Other recommendations for
systems for physiologic folates and thus synthesize this sub- ocular toxoplasmosis therapy in various situations are
stance instead. Folic acid antagonists inhibit this biochemical summarized in Table 162.1.
pathway and prevent the organism from replicating by Standard antitoxoplasmosis drug regimens can usually
impairing DNA synthesis.75 A combination of sulfadiazine control AIDS-related ocular lesions. Treatment is given until
(1 g PO qid for first day, then 500 mg PO qid for 2–8 weeks) 4–6 weeks after resolution of all signs and symptoms. However,
with pyrimethamine (50 mg PO bid for first day, then 25 mg PO as many as 20% of immunocompromised patients with toxo-
qid for 2–8 weeks) and folinic acid (5 mg PO three times a week) plasmosis may fail to respond to medical therapy.43 A
has classically been considered to be the most effective treatment combination of pyrimethamine and clindamycin was reported
for toxoplasmosis.76 If necessary, repeated courses of drugs may to be more effective than either pyrimethamine alone or in
be administered. However, many toxic side effects occur with combination with sulfadiazine for patients with AIDS.81
this regimen, most notably bone marrow depression with Toxoplasmosis in patients with AIDS frequently recurs when
subsequent hematologic complications. Weekly white blood cell medical treatment is discontinued.49,82,83 It may be necessary to
counts and platelet counts are therefore mandatory in these continue treatment indefinitely to maintain control of the
patients. disease, but adequate maintenance treatment regimens have
A less toxic means of treatment consists of clindamycin not been established. Zidovudine (azidothymidine (AZT) anta-
(300 mg PO tid or qid) in combination with sulfadiazine gonizes the anti-Toxoplasma activity of pyrimethamine both
(500 mg PO qid). Patients are generally treated for 4–6 weeks on in vivo and in vitro and may have to be discontinued.84 Co-
this drug regimen. However, diarrhea, colitis, and pseudomem- trimoxazole has been employed in AIDS patients sensitive to
branous colitis remain potential problems with clindamycin use. pyrimethamine, but this has been reported to be associated with
Co-trimoxazole (trimethoprim 160 mg with sulfamethoxazole treatment failure.85 Comanagement with an infectious disease
800 mg) given twice daily for 2–6 weeks either alone or in specialist is recommended when treating these patients.
combination with clindamycin (300 mg PO qid) has also been Atovaquone (Mepron), 750 mg tid, plus clarithromycin (Biaxin),
suggested for the treatment of toxoplasmosis in humans.77,78 500 mg bid, has been used for long-term suppressive therapy of
The side effects are similar to those of the sulfonamides. recurrent toxoplasmosis in selected individuals with AIDS,
Anterior uveitis associated with ocular toxoplasmosis may be especially in the context of sensitivity to the more classic anti-
treated with topical corticosteroids and cycloplegic agents along toxoplasma agents. Azithromycin (Zithromax) and Spiramycin
with concurrent systemic antitoxoplasmosis therapy. Increased (Rovamycine) are additional agents with antitoxoplasma
intraocular pressure can be managed with standard glaucoma activity that have had limited use thus far with promising
medical therapy. results.86,87
The addition of systemic corticosteroids to the therapeutic The role of laser photocoagulation in the treatment of active
regimen may diminish the degree of collateral damage from the ocular toxoplasmosis and for prophylaxis against the spread of
inflammatory response. Some experts advocate adding pred- lesions is limited.88 Although photocoagulation can destroy
nisone 12–24 h after initiating the antiparasitic therapy if the Toxoplasma cysts and tachyzoites, Toxoplasma cysts can reside
Toxoplasma lesion is in the posterior pole, threatening the optic in ophthalmoscopically normal-appearing retina. Furthermore,
nerve head, or if the vitreal response is extreme, starting with photocoagulation of active lesions can be complicated by retinal
40–80 mg of prednisone with breakfast daily for 1 week, then or vitreous hemorrhage or even by retinal detachment.
rapidly tapering the prednisone before discontinuing the anti- Vitrectomy and lensectomy can be performed to remove vitre-
toxoplasmosis therapy. Under no circumstances, however, ous and lens opacities in these patients.89 Patients undergoing
should systemic corticosteroids be used without concurrent surgery should probably be treated preoperatively with anti-
antitoxoplasmosis therapy, nor should they be used in patients Toxoplasma agents, and the medications should be continued
with necrotizing retinitis where the diagnosis of ocular postoperatively. 2091
RETINA AND VITREOUS

REFERENCES
1. Perkins, ES: Ocular toxoplasmosis. Br J elevated intraocular pressure: a protects mice against lethal infection with
Ophthalmol 1973; 57:1–17. retrospective study. J Glaucoma 2005; toxoplasma gondii. J Immunol 1984;
2. Krick JA, Remington JS: Toxoplasmosis in 14:3–10. 133:2818–2820.
the adult – an overview. N Engl J Med 23. Baglivo E, Safran AB: Haemorrhagic 40. Cohen SN: Toxoplasmosis in patients
1978; 298:550–553. toxoplasmic retinochoroiditis: description of receiving immunosuppressive therapy.
3. van Knapen F: Toxoplasmosis, old stories an unusual clinical presentation. Br J JAMA 1970; 211:657–660.
and new facts. Int Ophthalmol 1989; Ophthalmol 2003; 87:1051–1052. 41. Ruskin J, Remington JS: Toxoplasmosis in
13:371–375. 24. Fine SL, Owens SL, Haller JA, et al: the compromised host. Ann Intern Med
4. Holland GN: Reconsidering the Choroidal neovascularization as a late 1976; 84:193–199.
pathogenesis of ocular toxoplasmosis. complication of ocular toxoplasmosis. Am 42. Ryning FW, Mills J: Pneumocystis carinii,
Am J Ophthalmol 1999; 128:502–505. J Ophthalmol 1981; 91:318–322. toxoplasma gondii, cytomegalovirus and
5. Swartzberg JE, Remington JS: 25. Kennedy JE, Wise GN: Retinochoroidal the compromised host. West J Med 1979;
Transmission of toxoplasma. Am J Dis vascular anastomoses in uveitis. Am J 130:18–34.
Child 1975; 129:777–779. Ophthalmol 1971; 71:1221–1225. 43. Wong B, Gold JW, Brown AE, et al:
6. Couvreur J, Desmonts G: Congenital and 26. Braunstein RA, Gass JD: Branch artery Central-nervous-system toxoplasmosis in
maternal toxoplasmosis. A review of 300 obstruction caused by acute toxoplasmosis. homosexual men and parenteral drug
congenital cases. Dev Med Child Neurol Arch Ophthalmol 1980; 98:512–513. abusers. Ann Intern Med 1984; 100:36–42.
1962; 4:519–530. 27. Rose GE: Papillitis, retinal neovascularisation 44. Luft BJ, Brooks RG, Conley FK et al:
SECTION 10

7. de Jong PT: Ocular toxoplasmosis; and recurrent retinal vein occlusion in Toxoplasmic encephalitis in patients with
common and rare symptoms and signs. toxoplasma retinochoroiditis: a case report acquired immune deficiency syndrome.
Int Ophthalmol 1989; 13:391–397. with uncommon clinical signs. Aust N Z J JAMA 1984; 252:913–917.
8. Teutsch SM, Juranek DD, Sulzer A, et al: Ophthalmol 1991; 19:155–157. 45. Araujo FG, Remington JS: Toxoplasmosis
Epidemic toxoplasmosis associated with 28. Gentile RC, Berinstein DM, Oppenheim R, in immunocompromised patients. Eur J
infected cats. N Engl J Med 1979; et al: Retinal vascular occlusions Clin Microbiol 1987; 6:1–2.
300:695–699. complicating acute toxoplasmic 46. Jabs DA, Green WR, Fox R, et al: Ocular
9. Akstein RB, Wilson LA, Teutsch SM: retinochoroiditis. Can J Ophthalmol 1997; manifestations of acquired immune
Acquired toxoplasmosis. Ophthalmology 32:354–358. deficiency syndrome. Ophthalmology 1989;
1982; 89:1299–1302. 29. Toledo de Abreu M, Belfort R Jr, Hirata PS: 96:1092–1099.
10. Silveira C, Belfort R, Jr., Burnier M, Jr., et al: Fuchs’ heterochromic cyclitis and ocular 47. Friedman AH: The retinal lesions of the
Acquired toxoplasmic infection as the cause toxoplasmosis. Am J Ophthalmol 1982; acquired immune deficiency syndrome.
of toxoplasmic retinochoroiditis in families. 93:739–744. Trans Am Ophthalmol Soc 1984;
Am J Ophthalmol 1988; 106:362–364. 30. Jabs DA: Ocular toxoplasmosis. Int 82:447–491.
11. Couvreur J, Thulliez P: Acquired Ophthalmol Clin 1990; 30:264–270. 48. Schuman JS, Friedman AH: Retinal
toxoplasmosis of ocular or neurologic site: 31. Williams DM, Grumet FC, Remington JS: manifestations of the acquired immune
49 cases. Presse Med 1996; 25:438–442. Genetic control of murine resistance to deficiency syndrome (AIDS):
12. Glasner PD, Silveria C, Kruszon-Moran D, toxoplasma gondii. Infect Immun 1978; cytomegalovirus, candida albicans,
et al: An unusually high prevalence of 19:416–420. cryptococcus, toxoplasmosis and
ocular toxoplasmosis in southern Brazil. 32. Kasper LH, Crabb JH, Pfefferkorn ER: Pneumocystis carinii. Trans Ophthalmol
Am J Ophthalmol 1992; 114:136–144. Purification of a major membrane protein of Soc UK 1983; 103(Pt 2):177–190.
13. Burnett AJ, Shortt SG, Isaac-Renton J, toxoplasma gondii by immunoabsorption 49. Heinemann MH, Gold JM, Maisel J:
et al: Multiple cases of acquired with a monoclonal antibody. J Immunol Bilateral toxoplasma retinochoroiditis in a
toxoplasmosis retinitis presenting in an 1983; 130:2407–2412. patient with acquired immune deficiency
outbreak. Ophthalmology 1998; 33. Kasper LH, Crabb JH, Pfefferkorn ER: syndrome. Retina 1986; 6:224–227.
105:1032–1037. Isolation and characterization of a 50. Parke DW 2nd, Font RL: Diffuse
14. Ronday MJ, Luyendijk L, Baarsma GS, monoclonal antibody-resistant antigenic toxoplasmic retinochoroiditis in a patient
et al: Presumed acquired ocular mutant of toxoplasma gondii. J Immunol with AIDS. Arch Ophthalmol 1986;
toxoplasmosis. Arch Ophthalmol 1995; 1982; 129:1694–1699. 104:571–575.
113:1524–1529. 34. Nussenblatt RB, Mittal KK, Fuhrman S, 51. Holland GN: Ocular toxoplasmosis: a
15. Tabbara KF: Ocular toxoplasmosis: et al: Lymphocyte proliferative responses global reassessment. Part II. Disease
toxoplasmic retinochoroiditis. Int of patients with ocular toxoplasmosis to manifestations and management. Am J
Ophthalmol Clin 1995; 35:15–29. parasite and retinal antigens. Am J Ophthalmol 2004; 137:1–17.
16. Theodossiadis P, Kokolakis S, Ladas I, Ophthalmol 1989; 107:632–641. 52. Cochereau-Massin I, LeHoang P, Lautier-
et al: Retinal vascular involvement in acute 35. Bohne W, Heesemann J, Gross U: Frau M, et al: Ocular toxoplasmosis in
toxoplasmic retinochoroiditis. Int Induction of bradyzoite-specific human immunodeficiency virus-infected
Ophthalmol 1995; 19:19–24. toxoplasma gondii antigens in gamma patients. Am J Ophthalmol 1992;
17. Friedmann CT, Knox DL: Variations in interferon-treated mouse macrophages. 114:130–135.
recurrent active toxoplasmic Infect Immun 1993; 61:1141–1145. 53. Pivetti-Pezzi P, Accorinti M, Tamburi S,
retinochoroiditis. Arch Ophthalmol 1969; 36. Denkers EY, Gazzinelli RT: Regulation and et al: Clinical features of toxoplasmic
81:481–493. function of T-cell-mediated immunity during retinochoroiditis in patients with acquired
18. Stanford MR, Tomlin EA, Comyn O, toxoplasma gondii infection. Clin. immunodeficiency syndrome. Ann
et al: The visual field in toxoplasmic Microbiol. Rev 1998; 11:569–588. Ophthalmol 1994; 26:73–84.
retinochoroiditis. Br J Ophthalmol 2005; 37. Hafizi A, Modabber FZ: Effect of 54. Zimmerman LE: Ocular pathology of
89:812–814. cyclophosphamide on toxoplasma gondii toxoplasmosis. Surv Ophthalmol 1961;
19. Doft BH, Gass JD: Outer retinal layer infection: reversal of the effect by passive 6:832–856.
toxoplasmosis. Graefes Arch Clin Exp immunization. Clin Exp Immunol 1978; 55. Moorthy RS, Smith RE, Rao NA:
Ophthalmol 1986; 224:78–82. 33:389–394. Progressive ocular toxoplasmosis in
20. Folk JC, Lobes LA: Presumed toxoplasmic 38. Naot Y, Remington JS: Use of enzyme- patients with acquired immunodeficiency
papillitis. Ophthalmology 1984; 91:64–67. linked immunosorbent assays (ELISA) for syndrome. Am J Ophthalmol 1993;
21. Borruat FX, Kapoor R, Sanders MD: detection of monoclonal antibodies: 115:742–747.
Simultaneous retinal and optic nerve experience with antigens of toxoplasma 56. Holland GN, Engstrom RE, Jr., Glasgow BJ,
lesions in toxoplasmosis: the advantages of gondii. J Immunol Methods 1981; et al: Ocular toxoplasmosis in patients with
magnetic resonance imaging. Br J 43:333–341. the acquired immunodeficiency syndrome.
Ophthalmol 1993; 77:450–452. 39. Sharma SD, Araujo FG, Remington JS: Am J Ophthalmol 1988; 106:653–667.
22. Westfall AC, Lauer AK, Suhler EB, et al: Toxoplasma antigen isolated by affinity 57. Remington JS, Miller MJ, Brownlee I:
Toxoplasmosis retinochoroiditis and chromatography with monoclonal antibody IgM antibodies in acute toxoplasmosis.
2092
Toxoplasmosis

II. Prevalence and significance in 68. Davis JL, Feuer W, Culbertson WW, et al: ocular toxoplasmosis. Ophthalmology
acquired cases. J Lab Clin Med 1968; Interpretation of intraocular and serum 1992; 99:920–925.
71:855–866. antibody levels in necrotizing retinitis. 79. Nicholson DH, Wolchok EB: Ocular
58. Greven CM, Teot LA: Cytologic Retina 1995; 15:233–240. toxoplasmosis in an adult receiving long-
identification of toxoplasma gondii from 69. Lynch MI, Cordeiro F, Ferreira S, et al: term corticosteroid therapy. Arch
vitreous fluid. Arch Ophthalmol 1994; Lacrimal secretory IgA in active posterior Ophthalmol 1976; 94:248–254.
112:1086–1088. uveitis induced by toxoplasma gondii. Mem 80. Sabates R, Pruett RC, Brockhurst RJ:
59. Schlaegel TF Jr: Ocular toxoplasmosis and Inst Oswaldo Cruz 2004; 99:861–864. Fulminant ocular toxoplasmosis. Am J
par planitis. New York: Grune & Stratton; 70. Aouizerate F, Cazenave J, Poirier L, et al: Ophthalmol 1981; 92:497–503.
1978:138–172. Detection of toxoplasma gondii in aqueous 81. Rolston KV, Hoy J: Role of clindamycin in
60. Kelan AE, Ayllon-Leindl L, Labzoffsky NA: humour by the polymerase chain reaction. the treatment of central nervous system
Indirect fluorescent antibody method in Br J Ophthalmol 1993; 77:107–109. toxoplasmosis. Am J Med 1987;
serodiagnosis of toxoplasmosis. Can J 71. de Boer JH, Verhagen C, Bruinenberg M, 83:551–554.
Microbiol 1962; 8:545. et al: Serologic and polymerase chain 82. Luft BJ, Conley F, Remington JS, et al:
61. Pyndiah N, Krech U, Price P, et al: Simplified reaction analysis of intraocular fluids in the Outbreak of central-nervous-system
chromatographic separation of diagnosis of infectious uveitis. Am J toxoplasmosis in western Europe and
immunoglobulin M from G and its Ophthalmol 1996; 121:650–658. North America. Lancet 1983; 1:781–784.
application to toxoplasma indirect 72. Contini C, Seraceni S, Cultrera R, et al: 83. Levy RM, Bredesen DE, Rosenblum ML:
immunofluorescence. J Clin Microbiol Evaluation of a real-time PCR-based assay Neurological manifestations of the acquired
1979; 9:170–174. using the lightcycler system for detection immunodeficiency syndrome (AIDS):

CHAPTER 162
62. Fuccillo DA, Madden DL, Tzan N, et al: of toxoplasma gondii bradyzoite genes in experience at UCSF and review of the
Difficulties associated with serological blood specimens from patients with literature. J Neurosurg 1985; 62:475–495.
diagnosis of toxoplasma gondii infections. toxoplasmic retinochoroiditis. Int J Parasitol 84. Israelski DM, Tom C, Remington JS:
Diagn Clin Immunol 1987; 5:8–13. 2005; 35:275–283. Zidovudine antagonizes the action of
63. Araujo FG, Barnett EV, Gentry LO, et al: 73. Stanford MR, See SE, Jones LV, et al: pyrimethamine in experimental infection
False-positive anti-toxoplasma fluorescent- Antibiotics for toxoplasmic with Toxoplasma gondii. Antimicrob Agents
antibody tests in patients with antinuclear retinochoroiditis: an evidence-based Chemother 1989; 33:30–34.
antibodies. Appl Microbiol 1971; systematic review. Ophthalmology 2003; 85. Holliman RE: Toxoplasmosis and the
22:270–275. 110:926–931; quiz 931–932. acquired immune deficiency syndrome.
64. Asbell PA, Vermund SH, Hofeldt AJ: 74. Huskinson-Mark J, Araujo FG, Remington J Infect 1988; 16:121–128.
Presumed toxoplasmic retinochoroiditis in JS: Evaluation of the effect of drugs on the 86. Chang HR: The potential role of
four siblings. Am J Ophthalmol 1982; cyst form of toxoplasma gondii. J Infect Dis azithromycin in the treatment of
94:656–663. 1991; 164:170–171. prophylaxis of toxoplasmosis. Int J STD
65. Weiss MJ, Velazquez N, Hofeldt AJ: 75. Giles CL: Pyrimethamine (Daraprim) and AIDS 1996; 7(Suppl 1):18–22.
Serologic tests in the diagnosis of the treatment of toxoplasmic uveitis. 87. Hacker M, Richter R, Gumbel H, et al:
presumed toxoplasmic retinochoroiditis. Surv Ophthalmol 1971; 16:88–91. Toxoplasmosis retinochorioiditis, a therapy
Am J Ophthalmol 1990; 109:407–411. 76. Smith RL, Nozik RA: Toxoplasmic comparison between spiramycin and
66. Naot Y, Desmonts G, Remington JS: IgM retinochoroiditis. In: Smith RE, Nozik RA, pyrimethamine/sulfadiazine. Klin Monatsbl
enzyme-linked immunosorbent assay test eds. Uveitis: a clinical approach to Augenheilkd 1998; 212:84–87.
for the diagnosis of congenital toxoplasma diagnosis and management. Baltimore: 88. Ghartey KN, Brockhurst RJ:
infection. J Pediatr 1981; 98:32–36. Williams & Wilkins; 1989:128–134. Photocoagulation of active toxoplasmic
67. Tomasi JP, Schlit AF, Stadtsbaeder S: 77. Norrby R, Eilard T, Svedhem A, et al: retinochoroiditis. Am J Ophthalmol 1980;
Rapid double-sandwich enzyme-linked Treatment of toxoplasmosis with 89:858–864.
immunosorbent assay for detection of trimethoprim–sulphamethoxazole. Scand J 89. Fitzgerald CR: Pars plana vitrectomy for
human immunoglobulin M anti-toxoplasma Infect Dis 1975; 7:72–75. vitreous opacity secondary to presumed
gondii antibodies. J Clin Microbiol 1986; 78. Opremcak EM, Scales DK, Sharpe MR: toxoplasmosis. Arch Ophthalmol 1980;
24:849–850. Trimethoprim–sulfamethoxazole therapy for 98:321–323.

2093
Retinal Manifestations of the Acquired
CHAPTER
Immunodeficiency Syndrome: Diagnosis and
163 Treatment
Michelle Tarver-Carr, M.D., Ph.D. and James P. Dunn, M.D.

OVERVIEW defense against infections. Macrophages, however, are not lysed


in the viral cycle but instead escort the virus to vulnerable host
Acquired immunodeficiency syndrome (AIDS) is a potentially tissues such as the central nervous system.5
fatal multisystem disorder in which immunity is severely HIV infection is diagnosed by assaying viral p24 anti-
compromised as a result of infection with the human genemia, viral culture, or amplification of viral nucleic acids by
immunodeficiency virus (HIV). AIDS is defined by the Centers polymerase chain reaction. People with suspected HIV infection
for Disease Control as a CD4+ T-lymphocyte count of less are screened with enzyme-linked immunosorbent assay
than 200 cells/mL or specific opportunistic disease, including (ELISA), which detects core HIV proteins and surface
cytomegalovirus (CMV) retinitis.1 According to the World glycoproteins. While the ELISA is very sensitive, a positive
Health Organization, ~ 38 million people worldwide were result must be confirmed by Western blot analysis. Moreover,
infected with HIV in a cross-sectional analysis in 2003. In 2004 disease progression is monitored by the viral load of HIV since
alone, 4.8 million new infections developed.2 the HIV-1 RNA assay has been shown to predict the clinical
This chapter will focus on the retinal manifestations of AIDS, progression of HIV-associated disease.6 Advanced HIV disease
particularly noninfectious microvasculopathy and CMV retinitis. is associated with many ocular and systemic manifestations.
Other less common viral, bacterial, parasitic, and fungal infec-
tions affecting the retina will also be discussed. Lastly, diag- NONINFECTIOUS RETINAL
nostic methods in cases in which the cause of the ocular disease VASCULOPATHY
is not clinically evident will be described, along with current
treatments for the opportunistic infections and HIV itself.
BACKGROUND AIDS RETINOPATHY
EPIDEMIOLOGY OF HIV INFECTION Noninfectious HIV microangiopathy, also called ‘background
AND AIDS HIV retinopathy’, is the most common ophthalmic manifes-
tation of HIV/AIDS. It is uncommon in those with CD4+ cell
In 2004, there were 38 730 new cases of HIV with 14.1 cases of counts greater than 200 cells/mL but occurs in a majority of
AIDS per 100 000 in the United States. There are currently ~ patients with CD4+ counts less than 50 cells/mL, and therefore
950 000 individuals with AIDS living in the US, translating to serves as a marker for advanced immunosuppression.7
a prevalence rate of 136.7/100 000 of HIV and 168.8/100 000 of The retinopathy is characterized by cottonwool spots
AIDS or HIV.1 The number of HIV cases in the United States (Fig. 163.1), which occur in 25–50% of patients with AIDS,
has increased linearly over time with almost half of the patients intraretinal hemorrhages, white-centered hemorrhages, retinal
being diagnosed with AIDS within 12 months after becoming capillary microaneurysms, telangiectasias, and capillary
HIV seropositive.3 HIV is transmitted by (1) sexual contact, nonperfusion.8 Affected patients are usually asymptomatic.
(2) inoculation from infected sources (especially the sharing of Cotton-wool spots are a result of microinfarctions and edema in
contaminated needles by injection drug users), and (3) exposure the retinal nerve fiber layer; the histopathologic findings (cytoid
to infected blood, tissues, and milk in utero, during or after bodies) correspond to the reversible feathery whitening seen
birth. At 2–4 weeks after infection many patients are clinically. They are not pathognomonic, since they also occur in
asymptomatic; however, ~ 50–70% of patients develop an acute other conditions such as hypertension, diabetes, severe anemia,
retroviral syndrome typified by myalgias, lymphadenopathy, lupus, and leukemia.9 As in diabetes, the earliest histopatho-
diarrhea, and other viral symptoms.4 logic finding in noninfectious HIV-related microvasculopathy is
Viral invasion and replication occur through attachment of loss of capillary pericytes. Vascular sludging from hypergamma-
HIV to the CD4+ receptors on CD4+ T-lymphocytes, globulinemia, immune complex deposition on the vascular
macrophages, and other dendritic cells. The viral envelope fuses walls, and local release of cytotoxic substances have all been
with the cellular membrane, leading to insertion and uncoating postulated as causative factors for the hemorrhages.10 The
of the viral nucleocapsid. HIV reverse transcriptase forms a cotton-wool spots occur in the posterior pole, often near the
complementary DNA strand from HIV RNA which inserts into optic disc. There is no associated vitritis or increased vascular
the host genome using HIV integrase. Activation of the CD4+ permeability. The time to regression is 6–12 weeks.11
T-lymphocyte causes transcription and translation of viral Cumulative loss of retinal ganglion cells manifests itself as an
proteins and viral assembly. The viral particles bud from the cell asymptomatic decrease in visual acuity, color vision, and
surface and mature upon cleavage of viral polyproteins by HIV contrast sensitivity.12,13 With highly active antiretroviral
protease. This process promotes apoptosis of CD4+ cells, therapy (HAART), the lesions can disappear, leaving a normal-
decreasing the number of T-lymphocytes available to mount a appearing fundus, although studies of patients with AIDS show 2095
RETINA AND VITREOUS

70

Incidence per 1,000 person years


60
50
40
30
20
10
0
1992 1993 1994 1995 1996 1997
Calendar Year

FIGURE 163.2. Incidence of CMV retinitis over time standardized by


age, race, and CD4+ T-lymphocyte distribution.
SECTION 10

Reference: CDC Surveillance for AIDS-defining opportunistic illnesses, 1992–97.

FIGURE 163.1. HIV microvasculopathy. Note the cottonwool spots


and intraretinal hemorrhage in the right eye of this 36-year-old woman
with AIDS and a CD4+ count persistently below 10 cells/mL. She had compromised patients, reactivation may lead to clinically
CMV retinitis in the fellow eye. significant disease following hematogenous dissemination. The
retina is the most commonly affected tissue, occurring in
75–80% of patients with reactivation22,23; the gastrointestinal
a global decrease in the number of optic nerve fibers resulting tract, lung, adrenal glands, and nervous system are also
from axonal degeneration in the absence of infection. common sites of clinical disease. Most patients with AIDS-
The retinal hemorrhages in background retinopathy may related CMV retinitis have CD4+ count less than 50 cells/mL.24
appear as including flame-shaped, dot-blot or punctuate Histological samples of CMV retinitis demonstrate intra-
peripheral intraretinal hemorrhages; occasionally Roth spots cytoplasmic inclusion bodies in vascular endothelial cells.25
(hemorrhages with a white central region) are seen. The hemor- The infection appears to spread by cell-to-cell transmission.
rhages are not associated with a coagulopathy or a bleeding CMV retinitis was one of the first opportunistic infections
diathesis. Fluorescein angiography may reveal areas of micro- reported as a complication of AIDS. In the era before HAART,
aneurysms, telangiectasias, and capillary loss. up to 30% of patients with AIDS developed CMV retinitis.23
In patients taking HAART, the incidence of CMV retinitis has
declined dramatically to ~ 20–25% of the pre-HAART era rate
RETINAL MACROVASCULOPATHY (Fig. 163.2).26
Branch or central retinal vein occlusions, branch retinal artery
occlusion, and ischemic maculopathy have all been described in
the literature in persons with HIV/AIDS.14–17 In one study of DESCRIPTION
nearly 2500 consecutive persons with HIV infection, 1.3% of In untreated patients, CMV retinitis spreads relentlessly
patients were noted to have retinal vascular occlusion, of whom throughout the retina, causing full- thickness necrosis and
48% had central retinal vein occlusion (CRVO), 27.3% branch blindness. Lesions may be single or multiple and unilateral or
retinal vein occlusion (BRVO), 12.1% hemitretinal vein bilateral, affecting any part of the retina. Symptoms may
occlusion (HRVO), and 3% central retinal artery occlusion. The include visual field loss, decreased visual acuity, floaters, and
presence of microvasculopathy was associated with a higher risk photopsias, but it is not uncommon for patients to be
of macrovasculopathy.18 In contrast to noninfectious micro- completely asymptomatic.27,28 The presence of subjective
vasculopathy, visual loss from these macrovasculopathies can scotomata varies with the location of the lesions. In patients
be severe and permanent. The risk of retinal vein occlusion in with CD4+ counts of less than 50 cells/mL, the prevalence of
those with HIV-infection is significantly higher than in their previously undiagnosed CMV retinitis detected on screening
age-matched healthy peers. The cause of the retinal has been reported to be as high at 13–18%.24 Eye pain and
macrovasculopathy is unclear; however, it is presumed that redness are not typical features. CMV retinitis may appear as
abnormalities of retinal blood are contributing factors.19,20 intraretinal yellow necrotic lesions with retinal hemorrhages
(fulminant/edematous retinitis), as less densely necrotic
CYTOMEGALOVIRUS RETINITIS without hemorrhage (indolent/granular retinitis), or some
combination of the two. Both types, however, are characterized
Human cytomegalovirus (CMV) is an enveloped, double- by a dry-appearing, granular border (Fig. 163.3). There is often
stranded DNA herpes virus. It is most often transmitted by a mild vitritis and anterior chamber inflammation.27 The
contact with persons shedding the virus in their urine, sputum, disease progresses outward in a brushfire pattern, leaving
breast milk, semen, or cervical secretions. The virus can also be behind an atrophic, avascular, full-thickness retinal necrosis.
transmitted by solid organ or bone marrow transplantation or The disease is described according to the location within the
through blood transfusions. Between 40 and 100% of healthy retina: zone 1 is within 3000 mm of the center of the fovea or
adults have antibodies to CMV, confirming prior exposure. 1500 mm of the optic nerve, zone 2 is located from the periphery
Primary CMV infection is usually asymptomatic, although 5% of zone 1 to the ampulla of the vortex veins, and zone 3 from
of newly infected individuals may manifest a mononucleosis- the periphery of zone 2 to the ora serrata. Zone 1 lesions are
like syndrome.21 The virus is latent in immunocompetent generally considered immediately sight-threatening and usually
2096 individuals with residence in the bone marrow. In immuno- require more urgent therapy than do more peripheral lesions.
Retinal Manifestations of the Acquired Immunodeficiency Syndrome: Diagnosis and Treatment

should be considered in the differential diagnosis of CMV


retinitis, especially in patients with neurologic abnormalities.

DIAGNOSIS
CMV retinitis is usually diagnosed clinically. The presence of
antibodies to CMV indicates prior infection but is neither
indicative of concurrent clinical disease nor predictive of future
disease. Reactivation of CMV is confirmed by positive cultures
for CMV from blood, urine, or throat washings or from
polymerase chain reaction (PCR) amplification of CMV DNA.30
An elevated CMV viral load may be the most predictive risk
factor for end-organ disease. In atypical cases, endoretinal
biopsy taken at the junction of necrotic and actively infected
tissue may be necessary. The pathognomonic finding is the
presence of intranuclear and intracytoplasmic inclusion bodies
with an ‘owl’s eye’ appearance, or positive CMV staining by
immunohistochemistry; electron microscopy demonstrates

CHAPTER 163
FIGURE 163.3. CMV retinitis with granular appearance.
viral particles.

Natural History COMPLICATIONS


If CMV retinitis remains untreated, the lesions spread Retinal Detachment
inexorably to cause total retinal necrosis. The borders of the Untreated CMV retinitis causing full-thickness retinal necrosis
lesion advance on average one disc diameter every 6 weeks in a may result in a retinal detachment within 3–6 months of
centrifugal pattern.29 The older portion of the advancing lesion diagnosis.29 In the pre-HAART era, rhegmatogenous retinal
becomes gliotic scar tissue. Optic atrophy can also be observed detachment occurred at a rate of 25% per patient-year in
late in the disease from the diffuse retinal damage. Vision loss patients treated with anti-CMV therapy, and detachments
may occur from retinal necrosis involving the macula, remain a major cause of vision loss in CMV retinitis. Lesions
rhegmatogenous retinal detachment, secondary optic nerve located at the peripheral retina where the vitreous body is
involvement, or exudative swelling along an active border adherent to the retina are more likely to be associated with a
adjacent to the fovea. retinal detachment than those lesions located in the posterior
pole. Eyes with larger lesions and a longer duration of active
retinitis also have a higher risk of retinal detachment. While the
DIFFERENTIAL DIAGNOSIS retina can usually be reattached surgically, the ultimate visual
In the early stage of CMV retinitis it may be difficult to acuity is usually worse than the preoperative vision before the
distinguish cottonwool spots from foci of viral infection. detachment. It is presumed that vitreous fluid egresses through
Cottonwool spots do not cause blurred vision, floaters, or the breaks in the necrotic retina, rendering photocoagulation
photopsias and are not associated with vitritis or anterior ineffective at promoting adhesion between the retina and the
uveitis; they usually have a feathery border rather than the choriocapillaris. Scleral buckling and vitrectomy with gas
granular border of CMV retinitis. Repeating the ophthalmic tamponade are often ineffective in treating CMV-related
exam after 1-2 weeks will aid in making the diagnosis, as detachments. Long-term tamponade with oil is often necessary
cotton-wool spots will not progress in size. due to the number of holes in atrophic or gliotic retina.31,32
As CMV retinitis progresses, it may resemble other infec- Exudative retinal detachments are observed in eyes with
tions of the retina. Toxoplasmic retinochoroiditis can mimic involvement of zone 1 and peripapillary lesions.27 These
CMV retinitis, but usually presents with a creamy border, detachments usually resolve with treatment of the retinitis and
prominent vitritis, and the absence of intralesional hemor- vision may improve substantially (Fig. 163.4).33
rhages. Herpetic infections of the retina (e.g., acute retinal
necrosis and progressive outer retinal necrosis) are less common Immune Recovery Uveitis
than CMV retinitis and have a clinical appearance that is Immune recovery uveitis (IRU), also known as immune
distinctly different. Bacterial and fungal infections, including recovery vitritis, was first described in patients with inactive
syphilitic retinitis and aspergillosis or candida retinitis, may CMV retinitis who had immune reconstitution following
also mimic CMV retinitis. Finally, intraocular lymphoma initiation of HAART. The reported incidence ranges widely from

FIGURE 163.4. CMV retinitis with exudative


retinal detachment (a) before and (b) after
treatment.

a b
2097
RETINA AND VITREOUS

0.109 cases per person-year to 0.83 cases per person-year.34,35


While differences in the case definition partially explain the
TREATMENT OPTIONS
difference in incidence, another factor is thought to be changes
in the treatment of CMV retinitis; patients initially treated with CMV Retinitis Treatment Overview
cidofovir may be at higher risk for IRU. Immune recovery
Screening
uveitis usually occurs 2–16 weeks after the elevation of the
• Patients with CD4<100 cells/mL for periodic opthalmological
CD4+ lymphocyte counts, which typically is noted 2.5–8
evaluation; more urgent in patients with visual symptoms
months after starting HAART.36
Affected patients develop a painless decrease in visual acuity Diagnosis
and floaters without any redness. The vision loss is generally • Clinical presentation: single or multiple lesions with dry,
mild, in the range of 20/40 to 20/80. The symptoms often begin granular-appearing border; can occur with or without retinal
within a few weeks or months after the CD4+ counts increase hemorrhage
above 100 cells/mL.37 Examination reveals moderate to severe Management
vitritis, papillitis, cystoid macular edema, and/or epiretinal • Zone 1 disease: treat urgently with intravenous anti-CMV agent
membranes. Uncommon complications include vitreous hemor- • Insertion of ganciclovir implant in those with lesions
rhage, retinal neovascularization, optic disc neovascularization, immediately adjacent to the optic nerve or fovea
vitreomacular traction syndrome, proliferative vitreoretino- • Monitor renal function, hematological indices, and toxicity
pathy, posterior and anterior subcapsular cataracts, posterior profiles to determine appropriate treatment
SECTION 10

synechiae, and angle closure glaucoma.38,39 The clinical findings • Patients with CMV retinitis (even if inactive) and persistent
may vary depending on ocular co-morbidities; for instance, eyes immunosuppression should be examined monthly
that have had a vitrectomy with silicone oil tamponade will not Prevention
have visible vitritis but instead will have cystoid macular edema • HAART for patients with CD4+ count less than 200 cells/mL
or spillover cells in the anterior chamber.37 to reduce the risk of CMV retinitis
The cause of immune recovery syndrome is not known. • Secondary prophylaxis with valganciclovir in those with
Immune reconstitution is thought to occur after depletion of CMV retinitis until CD4 counts consistently higher than
CD4+ lymphocytes is halted, allowing the surviving clones to 100 cells/mL while on HAART
expand their population.40 It has been suggested that the
recovering immune system reaches a threshold above which the
body is able to mount an ocular inflammatory response to CMV It is essential to realize that CMV retinitis is part of systemic
antigens in the eye. Some patients, however, may lack the CMV and HIV infection and that treatment must take into
clones capable of responding to cytomegalovirus even though account the nonocular effects of disease; the best outcome is
they have elevated CD4+ T-lymphocyte counts. Eyes with obtained in patients treated with both anti-CMV therapy as
inactive retinitis involving over 30% of the retinal area are at a well as HAART. In the pre-HAART era, patients with AIDS with
higher risk of developing immune recovery uveitis when cytomegaloviremia had a higher risk of end-organ CMV disease
compared to eyes with less extensive involvement, presumably and mortality.45,46 The mechanisms by which CMV increases
because those eyes with a larger area of CMV retinitis may mortality includes transactivation of HIV, altering the tropism
generate a greater antigenic stimulus, prompting a more of HIV, production of interleukins, chemokine receptors, and
aggressive inflammatory response.41 This reaction is analogous interference with natural killer cells, thereby inhibiting the
to the necrotizing lymphadenitis observed with Mycobacterium host’s ability to clear viruses.47 Kempen et al showed ~30%
avium-complex infections after the initiation of HAART reduction in mortality in patients treated with systemic anti-
therapy.42 As immune recovery progresses, the production of CMV therapy, after adjusting for HAART.48 The widespread use
CMV antigens stops and the inflammatory reaction subsides.40 of HAART has not only changed the course of AIDS, but has
Limited histologic and immunohistochemical studies of epi- also reduced the rates of vision loss in those with CMV retini-
retinal membranes found in this syndrome have shown a tis. Those with HAART-induced immune recovery have half the
predominately T-lymphocyte population. Surprisingly, higher risk of visual acuity loss.49 This decrease has been attributed to
levels of CD4+ T-lymphocyte levels were not associated with a reduction in progression (96% reduction), contralateral eye
the severity of the inflammation. Some investigators have involvement (82%) and associated retinal detachments (88%) in
suggested that variations in the incidence of IRU may also be the HAART era.50–52 Patients with unilateral CMV retinitis are
due to different virus strains which may stimulate different at a significantly higher risk of developing second-eye retinitis
degrees of inflammation or have different replication rates. of the order of 26.1% per person-year, almost all within the first
There may also be differences in the number of clones capable few months after diagnosis, despite systemic CMV treatment.
of responding to CMV, even if the CD4+ counts are the same.43 Those on treatment with anti-CMV therapy and HAART with
Vision loss typically results from the cystoid macular edema immune recovery are significantly less likely to develop
or the formation of epiretinal membranes and vitreomacular contralateral eye involvement.53 However, cytomegalovirus
traction. Less commonly, branch retinal vessel occlusion and infection worsens the prognosis in patients with AIDS; CMV
retinal neovascularization are the cause. Successful treatment viral load and active disease are associated with an increased
of IRU has not been demonstrated in a randomized clinical mortality despite the use of HAART.54
trial; however, a few small cohort studies and case reports have There are four drugs that have been FDA approved for the
shown that treatment with periocular or systemic corticos- treatment of CMV retinitis-ganciclovir, valganciclovir, f os-
teroids may result in improved vision.41,44 On the other hand, carnet, and cidofovir. All of these drugs work by inhibiting the
Zegans found that the vitreous cells resolved and the vision activity of CMV DNA polymerase that is essential for
improved within 6 weeks with or without antiinflammatory replication of the virus (a fifth approved drug, fomvirsen, is an
therapy.37 Re-activation of CMV retinitis following corticos- antisense oligonucleotide that works by a different mechanism,
teroid therapy appears to be very rare. It is unclear whether but the drug was voluntarily taken off the market in 2003 due
resumption of anti-CMV therapy or longer use of anti-CMV to poor sales). All of these drugs are virostatic, not virocidal.
therapy prior to discontinuation in patients with immune The dosing strategy for systemic medications involves an
2098 recovery reduces the risk of IRU. induction period of 2–3 weeks, followed by indefinite, lower-
Retinal Manifestations of the Acquired Immunodeficiency Syndrome: Diagnosis and Treatment

TABLE 163.1. Medication Regimen for the Treatment of CMV Retinitis.

Drug Induction Maintenance Laboratory Tests Toxicity Considerations

Ganciclovir
Intravenous 5 mg/kg IV bid 5 mg/kg IV qd Complete blood Anemia, Dose adjustment for
(Cytovene) µ 14 days count twice a thrombocytopenia, renal disease
not used for 1 g po tid week during neutropenia Avoid other bone marrow
induction 2000 g induction, weekly suppressing
Oral N/A per 0.1 mL during maintenance medications
(Cytovene) 2000 mg intraocular Serum creatinine Cytokine support)
Implant per 0.1mL injection monthly (granulocyte colony-
(Vitrasert) intraocular qweek Complete blood stimulating factor)
Intraocular injection twice count every for neutropenia
injections weekly 1–2 weeks; serum (absolute neutrophil
creatinine monthly count <500/mm3)
None for intraocular
or implant
administration
alone

CHAPTER 163
Valganciclovir 900mg po 900 mg po Complete blood Bone marrow Avoid other bone
(Valcyte) bid µ 21 days qday count weekly suppression, marrow suppressing
nausea, diarrhea medications
Cytokine support for
neutropenia
Foscarnet 90 mg/kg bid 90–120 mg/kg Serum creatinine, Nephrotoxicity, Dose adjustment for
Intravenous µ 14 days or qday potassium, hypokaemia, renal disease
(Foscavir) 60 mg/kg tid 2400 g magnesium, hypocalcemia, IV prehydration with 1
Intraocular 2400 mg qweek calcium, hypomagnesemia, L 0.9% NaCl
injections biweekly phosphorous genital ulcers, Slow infusion if
biweekly during anemia, headache hypocalcemia
induction, weekly symptoms
during maintenance; Electrolyte replacement
hemoglobin
Cidofovir 5 mg/kg qweek 3–5 mg/kg IV Serum creatinine, urinary Nephrotoxicity, IV prehydration with 1 liter
Intravenous µ 14days q2weeks protein, complete proteinuria, Fanconi- 0.9% NaCl
(Visitide) blood count before like syndrome, Oral probenecid 2 g po
each infusion hypocalcemia, 3 h before, 1 g po 2 h
neutropenia, uveitis, after and 1g 8 h after
hypotony dose
Discontinue if IOP < 50%
of baseline
Discontinue if dipstick
proteinuria > 2+ or
serum Cr increases by
0.5 mg/dL
Fomvirsen 330 g 330 mcg None Increased IOP, uveitis Withdrawn from the
(Vitravene) intraocular intraocular market as of 2003
qweek

frequency maintenance therapy that must be lifelong in tration of ganciclovir at a dosage of 5 mg/kg twice a day during
patients with persistent immunosuppression (Table 163.1). the induction period initially halts retinal cell necrosis and
In patients with immune recovery (generally defined as a reduces the viral load of CMV. Maintenance therapy at a dosage
recovery in the CD4+ count by more than 50 cells/mL or to a of 5 mg kg–1 day–1 is then continued, but the need for daily
level greater than 100 cells/mL for at least 3–6 months), discon- intravenous mandates the need for a permanent indwelling
tinuation of maintenance therapy in those with inactive CMV catheter and the associated risks of line infection and sepsis.
retinitis is usually possible, thereby avoiding the cost and Maintenance therapy with oral ganciclovir was effective in
toxicity of treatment.55,56 Some investigators have suggested controlling retinitis temporarily and obviated the need for
that HIV viral load is a better predictor of progression than permanent catheterization,58 but bioavailability is poor (less
CD4+ counts.57 In light of these findings, there is no consensus than 7%) and the risk of progression of retinitis is high.
on when the maintenance therapy can be discontinued. Consequently, vision-threatening lesions (e.g., zone 1 disease)
Ganciclovir is the most commonly used medication for the are poor candidates for oral maintenance therapy.
treatment of CMV retinitis and can be administered intra- Valganciclovir is an oral prodrug which is hydrolyzed to
venously, orally, or intraocularly via an injection or an implant. ganciclovir in the gastrointestinal mucosa and has better bio-
Activation of ganciclovir requires initial phosphorylation by a availability (61%) than oral ganciclovir. The plasma levels
CMV-encoded kinase produced by the UL97 gene, followed by obtained with valganciclovir are comparable to that obtained
additional phosphorylation by host cellular enzymes to the with intravenous ganciclovir, making it a viable option for both
active drug, ganciclovir triphosphate, that inhibits CMV DNA induction and maintenance therapy at dosages of 900 mg twice
polymerase. In 80–90% of patients, the intravenous adminis- a day and 900 mg once daily, respectively.59,60 2099
RETINA AND VITREOUS

Intravitreous injections of both ganciclovir and foscarnet


have been used off-label to treat CMV retinitis. The usual
dosage for ganciclovir is 2 mg in 0.1 ml and for foscarnet
2400 mg in 0.1 mL. The injections must be given twice weekly
as induction therapy and once weekly thereafter, a regimen that
is usually not practical for patients in the long term. As with the
ganciclovir implant, the use of intravitreal injections does not
prevent second-eye or visceral CMV disease. On the other hand,
intravitreal injections can be very useful in obtaining rapid
control of zone 1 retinitis before more definitive therapy can be
instituted, and can serve as a bridge between systemic therapies
when toxicity requires temporary cessation. Risks include
subconjunctival hemorrhage, pain, acute glaucoma, infection,
vitreous hemorrhage, endophthalmitis, and retinal detachment.
Cidofovir is an acyclic nucleotide analog that was found in a
series of clinical trials to be effective at preventing the prog-
ression of CMV retinitis. Its activity is 10–100 times greater
FIGURE 163.5. Placement of ganciclovir implant in the vitreous cavity.
than other available anti-CMV medications. The induction
SECTION 10

regimen is 5 mg/kg once a week for 2 weeks, followed by


3–5 mg/kg every two weeks. Its prolonged antiviral effect allows
The ganciclovir implant is a surgically-implanted, sustained- for intermittent intravenous administration to control the
release, 4.5 mg ganciclovir pellet attached to a nonerodable disease, so that permanent catheterization is not needed.
strut. The pellet is coated with polyvinyl alcohol (which is However, the use of cidofovir is limited by potentially
permeable to ganciclovir) and ethylene vinyl acetate (which is irreversible nephrotoxicity through dose-dependent damage to
impermeable). The release rate of about 2 mg/h is determined proximal tubular cells of the kidney and by the development of
by the thicknesses of the coatings of the two polymers, anterior uveitis in 24–59% of patients receiving the drug.65–67
providing release of drug for about 7–8 months. The implant is Another 25% of patients developed hypotony presumed to be
placed in the vitreous cavity through a 5.5 mm pars plana secondary to changes in the ciliary epithelium and a reduction
incision made 3.5–4 mm posterior to the limbus; prolapsed of aqueous humor production. Injections of intravitreous
vitreous is excised, but an internal vitrectomy is not cidofovir may result in irreversible hypotony and for this reason
necessary.61 The strut attached to the implant is trimmed to a should never be used. The anterior uveitis is characterized by
length of about 2 mm and a hole is placed in the end of the strut pain, ciliary injection, posterior synechiae formation, and
with a 27- or 30-gauge needle. A double-armed 8–0 nylon or decreased visual acuity, and occurs on average after approx-
prolene suture is placed through this hole and the device is imately five doses of the medication. The uveitis usually
centered in the wound (Fig. 163.5). A stable intraocular con- responds to topical steroids and cycloplegia.65 The risk of
centration of ganciclovir is obtained within 24–48 h and is anterior uveitis appears to be reduced by the concomitant use of
roughly fourfold higher than that obtained with intravenous probenecid, which is thought to inhibit secretion of cidofovir by
therapy. In a randomized clinical trial of 188 patients with the ciliary body and therefore decreased intraocular drug levels.
active CMV retinitis and AIDS, the sustained release Clinical resistance to anti-CMV therapy is estimated to occur
ganciclovir implant decreased the risk of progression threefold in 20–30% of patients after one year of therapy. Mutations at
when compared to intravenous ganciclovir. However, the risk of the level of the UL97 gene result in the reduced uptake of
systemic CMV disease as well as involvement of the con- phosphorylated ganciclovir into host cells and cause low-level
tralateral eye was significantly higher than that observed in resistance that is often responsive to foscarnet or cidofovir.
patients receiving intravenous therapy.62 The implantation Mutations at the level of the UL54 gene cause high level resist-
procedure typically causes a transient decrease in visual acuity ance that may be resistant to all three medications. Resistance
from astigmatism to 20/80 on average which resolves within was associated with prior intravenous cidofovir use or prior oral
the first 4 weeks of treatment. The most significant risks are ganciclovir use.68 Phenotypic resistance is determined by
associated with the surgical complications. The presence of analysis of viral cultures, whereas genotypic resistance is deter-
silicone oil does not appear to interfere with the functioning of mined by the use of PCR techniques using blood or vitreous
the ganciclovir implant.63 specimens; there is a good correlation between resistance
Following ganciclovir implant placement, the risk of retinal determined from blood specimens and those obtained from the
detachment was 11.9%, endophthalmitis was 1.7%, and vitreous. It is important to distinguish resistant CMV retinitis
vitreous hemorrhage was 7.8%.62,64 Endophthalmitis may occur from progression of disease due to poor compliance or
through introduction of organisms into the eye at the time of inadequate drug penetration because the former is associated
surgery or subsequently through incomplete wound closure at with reduced survival and poorer visual outcomes.69
the intrascleral location of the suture tab. Because of these
potential complications, the implant is avoided if immune Treatment Effectiveness
reconstitution is expected unless zone 1 is involved. Resolution of CMV retinitis is recognized by the loss of satellite
Foscarnet, a pyrophosphate analog, also inhibits CMV DNA lesions, disappearance of vascular sheathing, and the formation
polymerase but does not require phosphorylation. It is compa- of a well-demarcated lesion borders. Systemic therapy may take
rably effective to ganciclovir in the treatment of CMV retinitis. several weeks or more to control retinitis and therefore may not
However, its tedious intravenous administration schedule be adequate in the initial treatment of lesions near the fovea or
coupled with its side effect profile, especially nephrotoxicity, has optic nerve. In these cases, immediate intravitreous injections
made it a less desirable first-line therapy. Because foscarnet does of ganciclovir or foscarnet, followed by placement of a gan-
not require phosphorylation, it may be effective in the treat- ciclovir implant, may be more effective. Secondary prophylaxis
ment of CMV retinitis resistant to ganciclovir as a result of with valganciclovir is then indicated to reduce the risk of
2100 mutations in the UL97 gene. second-eye and visceral CMV disease. In all patients with CMV
Retinal Manifestations of the Acquired Immunodeficiency Syndrome: Diagnosis and Treatment

retinitis, initiation or maximization of HAART is critical in with resumption of secondary prophylaxis in the presence of
achieving long-term control of retinitis. immunological failure (generally considered a decrease in the
Studies have shown that plasma CMV loads >400 copies/mL CD4+ count to less than 75 cells/mL). Routine use of primary
and leukocyte CMV loads >400 copies/106 leukocytes are prophylaxis with anti-CMV therapy, including valganciclovir, is
associated with an increased risk of CMV retinitis progression. not recommended because of the high cost, potential toxicity,
Unfortunately, the sensitivity and positive predictive value for and concerns about the development of drug-resistant disease.
these markers were low, limiting their global clinical utility.70
Measurement of the CMV plasma load may aid in rapidly OTHER VIRAL INFECTIONS
excluding CMV resistance and identifying those patients
requiring more detailed and time-consuming resistance testing.
PROGRESSIVE OUTER RETINAL NECROSIS
Salvage Therapy Progressive outer retinal necrosis (PORN) was the second most
Over time, most patients with CMV retinitis relapse with common infectious retinopathy in patients with AIDS before
progression of the disease when treated with systemic therapy the advent of HAART, occurring in 2% of patients with AIDS
alone. Progression is much less common in eyes treated with with a median CD4+ count of 21 cells/mL.74 It is generally
the ganciclovir implant because of the high-intraocular levels associated with reactivation of herpes zoster virus, although
obtained, but will invariably occur when the implant is herpes simplex virus (HSV-1) has also been reported to cause
exhausted of drug after 7–8 months if immune recovery is not PORN (Fig. 163.6). The retinitis can start in the macula or in

CHAPTER 163
established. Therefore, the use of HAART in all patients with the periphery with patchy, multifocal outer retinal lesions
CMV retinitis is highly desirable. Reactivation is documented coalescing rapidly throughout the retina in the absence of
with the reappearance of opacification of the border of the lesion vascular or vitreous inflammation. Severe visual loss from the
and advancement of the lesion borders or the development of diffuse retinal necrosis, optic atrophy, and retinal detachment
new lesions. Predictors of clinical relapse include HAART occurs in up to 70% of patients.75 Progression is extremely
failure, weak lymphoproliferative responses to CMV, and a low rapid, occurring over days or even hours. Some patients have
nadir HIV viral load.71 Progression of previously inactive lesions severe vision loss despite minimal or absent retinitis as a result
(‘breakthrough’ or ‘relapse’) in the first year of treatment is most of early optic nerve involvement; in these patients, the onset of
likely due to poor intraocular drug levels as a result of re- pain with vision loss may be a key finding. Treatment with
establishment of the blood–retinal barrier. Subsequent intravenous acyclovir alone results in a final vision of no light
progression may be due to drug resistance. Various strategies perception (NLP) in 67% of affected eyes within 1 month.74
have been used to manage the relapse but there is limited While there is no standard recommendation for treating PORN,
evidence to guide the clinician. Reinduction therapy with a combination therapy appears to result in better visual out-
higher dose of drug or by switching from one drug to another, comes. A regimen of intravitreal foscarnet and ganciclovir as
combined therapy with intravenous ganciclovir and foscarnet well intravenous administration of both drugs was associated
has been shown to be twice as effective as either drug alone in with 45% of patients having a vision of 20/80 or better. This
delaying further progression72; however, the added toxicity of same study suggested that laser demarcation may result in
the second drug makes combination therapy difficult to better outcomes as well due to a lower risk of retinal detachment.76
administer and reduces quality of life. In cases in which drug Indefinite maintenance therapy with valacyclovir is necessary
resistance is the cause of progression, a change from ganciclovir in patients with persistent immunosuppression. However, as
to foscarnet or cidofovir may be necessary. In patients who are with CMV retinitis and all viral retinitides, the development of
unable to tolerate systemic treatment, intravitreal injections or immune recovery by HAART is probably the most important
the ganciclovir implant are options. The ganciclovir implant factor in maintaining long-term control of the disease.
produces intravitreous drug levels high enough to overcome
some cases of low-level ganciclovir resistance (UL97 mutations)
but will not control retinitis with UL54 mutations. ACUTE RETINAL NECROSIS
Acute retinal necrosis (ARN) is an aggressive and devastating
retinitis characterized by a fulminant panuveitis with well-
INVESTIGATIONAL AGENTS demarcated confluent areas of full-thickness retinitis, choroiditis,
The dramatic drop in the incidence of CMV retinitis has
reduced the interest in developing new anti-CMV agents;
nonetheless, several drugs are being examined in early clinical
trials. Maribavir is a proposed drug that inhibits the UL97 gene
product prohibiting terminal DNA processing and hence
arresting nuclear egress of CMV particles. It has shown activity
in vitro in suppressing CMV retinitis. The drug does not need
intracellular activation and shows promise for retinitis resistant
to ganciclovir or foscarnet. Some of the side effects include
headache, dysgeusia, diarrhea, and nausea which are dose
related. Tomeglovir another orally administered drug still in the
early trial phase has also proven to have efficacy against
resistant strains of CMV.73

PREVENTION
The inhibition of viral replication instigated by HAART coupled
with the reconstitution of the immune system of patients with
AIDS has led to a dramatic decrease in morbidity and mortality.
Periodic CD4+ counts must be monitored in these patients FIGURE 163.6. PORN radiating in posterior pole. 2101
RETINA AND VITREOUS

infiltrates within choroidal vessels. The choroiditis takes from


TABLE 163.2. American Uveitis Society Criteria for Diagnosing
6 weeks to 4 months to resolve after treatment with double
ARN
strength trimethoprim-sulfmethoxazole. It is usually a marker
Required Clinical Criteria for disseminated pneumocystosis. In the era of HAART and
Rapid progression of disease in absence of therapy
systemic prophylaxis with systemic therapy such as TMP-SMX,
P. carinii choroidopathy has virtually disappeared.
One or more foci of retinal necrosis with discrete borders in
peripheral retina Toxoplasmosis Retinochoroiditis
Circumferential spread Toxoplasmosis retinochoroiditis was the third most common
infectious retinopathy of AIDS in the pre-HAART era, occurring
Evidence of occlusive vasculopathy and arteriolar involvement
in 1% of patients.7 The retinitis is characterized by yellow-white
Prominent vitreous inflammation and anterior chamber reaction areas of retinal necrosis with smooth, nongranular borders most
Supporting Clinical Criteria often involving the posterior pole in the absence of hemorrhages.
It is associated with a severe vitritis. In immunocompromised
Optic neuropathy/ atrophy patients, ocular involvement is much less common than central
Scleritis nervous system toxoplasmosis. The ocular involvement is often
atypical in patients with AIDS. The various presentations range
pain
from acute anterior uveitis to panophthalmitis with a secondary
SECTION 10

orbititis.80 Lesions are more frequently multifocal and bilateral


than in immunocompetent. Those who are infected have IgG
and papillitis. Varicella zoster virus and herpes simplex virus antibodies to T. gondii at the time of diagnosis. Treatment con-
have both been associated with this disease. The retinitis is sists of trimethoprim-sulfmethoxazole; maintenance therapy is
marked by deep retinal whitening, limited hemorrhage hemor- required because recurrences are frequent in patients without
rhage, and a rapid progression over days to weeks. Dense vitritis immune recovery but may be discontinued in patients whose
produces fibrotic bands that exert traction on necrotic retina CD4+ count is consistently above 200 cells/mL. For patients
leading to complex retinal detachments in 75% of patients and allergic to sulfa drugs, azithromycin, atovaquone, and clinda-
blindness in 64% of patients within 2–3 months. Unlike mycin with pyrimethamine/leucovorin may be effective.81,82
PORN, the CD4+ count is usually greater than 60 cells/mL in Primary prophylaxis for toxoplasmosis with TMP-SMX is given
these patients. The American Uveitis Society has criteria to those with CD4+ counts less than 200 cells/mL.
established for making the diagnosis of ARN (Table 163.2).77
Late in the course of the disease, retinal breaks and detach-
ments in the area of necrosis are common. Proliferative vitreo- FUNGAL DISEASE
retinopathy often accompanies the retinal atrophy in the Endogenous endophthalmitis can be caused by a number of
end-stages of the disease. The treatment for ARN in AIDS fungal infections.
patients is similar to that advocated in non-HIV patients.
Therapy includes intravenous acyclovir followed by indefinite Cryptococcus Neoformans
oral therapy with acyclovir, valacyclovir, or famciclovir. While Cryptococcus neoformans is a common cause of meningitis
those immunocompetent patients with ARN are sometimes with central nervous system disease in patients with AIDS.
treated with oral corticosteroids to reduce vitreous inflam- Chorioretinitis and endophthalmitis are both thought to result
mation, this approach is controversial in HIV patients because from direct invasion of the organism usually through neural
of concerns about additional immunosuppression; short-term tissue (e.g., optic nerve). Ocular symptoms often precede the
prednisone therapy probably does not put the patient at neurological disease.83 Treatment consists of amphotericin B
significant risk. Laser demarcation along the posterior border of during the induction phase and oral fluconazole for mainte-
the retinitis is indicated to reduce the risk of retinal nance therapy. Although many patients with AIDS also use
detachment. illegal drugs intravenously and Candidal infections are
associated with intravenous drug use, Candida endophthalmitis
NONVIRAL RETINAL INFECTIONS is uncommon in AIDS patients. Endogenous endophthalmitis
caused by Aspergillus fumigatus is also very rare in the AIDS
population.84 A systematic review of 77 AIDS patients with
PARASITIC INFECTIONS coccidiomycosis also found no cases of endogenous
Pneumocystis carinii Pneumonia endophthalmitis.85
Pneumocystis carinii pneumonia (PCP) was in the pre-HAART In 1987, the Centers for Disease Control (CDC) modified the
era the most common AIDS-defining condition in HIV-infected case definition for AIDS to include extrapulmonary histo-
patients, usually developing in patients with a CD4+ count less plasmosis in a person who is HIV positive. Most cases of
than 200 cells/mL. Patients at risk for developing PCP are placed histoplasmosis in immunosuppressed individuals are dissemi-
on prophylactic therapy with trimethoprim-sulfmethoxazole nated. The chorioretinitis that results is characterized by
(TMP-SMX), or dapsone, or dapsone plus leucovorin and aero- perivascular retinal lesions with minimal inflammation.
solized pentamidine. Prophylactic treatment with aerosolized Choroidal infection with H. capsulatum is rare.86
pentamidine alone is associated with an increased risk of
Pneumocystis carinii choroidopathy, characterized by pale, Paracoccidioidomycosis
multifocal, cream- to orange-colored plaques ranging in size Paracoccidioidomycosis is a granulomatous infection pro-
from 300–3000 mm deep in the mid-peripheral and posterior minent in Latin America. Although, it is the most frequent
choroid.78 Except in some cases of subfoveal involvement, systemic mycosis in Brazil, it is infrequently seen in patients
vision is typically preserved because neither the retina nor the with AIDS. The disease characteristically begins in the
vitreous are involved.79 Histopathologically, the choroidopathy oropharynx and disseminates to lymphoid tissue. In immuno
is characterized by eosinophilic, acellular, frothy, vacuolated compromised patients, it can result from reactivation of a
2102
Retinal Manifestations of the Acquired Immunodeficiency Syndrome: Diagnosis and Treatment

quiescent lesion since cell-mediated immunity keeps the can mimic CMV retinitis.90 The diagnosis may be made by
infection dormant. Choroidal granulomas and endophthalmitis endoretinal biopsy but more often treatment is initiated based
have been described. This fulminant infection can cause a total on a systemic workup, including spinal tap, neuroimaging, and
retinal detachment with marked vitritis and iridocyclitis and in brain biopsy. Survival is poor in affected patients.
one case resulted in enucleation.87
In general, the treatment for fungal endophthalmitis is
intravenous amphotericin B since most patients also have DIAGNOSTIC TECHNIQUES FOR
systemic fungemia. Because intravenous amphotericin has poor DETERMINING CAUSE OF RETINAL
intravitreal penetration, vitrectomy and intravitreal ampho- LESIONS
tericin B are recommended adjunct therapy for sight-
threatening endophthalmitis. In complex cases, where the clinical picture is atypical for any
disease, a diagnostic biopsy of the choroid, vitreous, or retina
may be necessary. Vitreous sampling is most appropriate if
BACTERIAL there is a brisk vitritis. At the time of a pars plana vitrectomy,
Syphilitic (luetic) retinitis is a complication of central nervous the vitreous washings in the cassettes can be plated for cultures
system involvement not exclusive to patients with AIDS. and cytology to further tailor treatment. If there is a choroidal
However, it may have a very fulminant course in patients with infiltrate, a choroidal biopsy can be performed. In the operating
patients with AIDS, resulting in blindness from retinal room, a complete peritomy is performed with isolation of the

CHAPTER 163
necrosis, vasculitis, and papillitis. The retinitis can manifest as four recti muscles. A half-thickness scleral trapdoor dissection
a localized large, pale yellow, placoid subretinal macular lesion is performed with preplaced sutures, allowing rapid closure of
or as a necrotizing retinitis resembling CMV retinitis, ARN and the wound, and is surrounded by cautery. Choroid only is
toxoplasmosis retinitis.88 The diagnosis is based on the clinical removed and the wound is then closed. Some advocate per-
appearance and positive serology. Because HIV-positive patients forming a pars plana vitrectomy prior to choroidal sampling to
frequently have a false-negative serum rapid plasma reagin aid in the maintenance of intraocular pressure. Endoretinal
(RPR) test, the more specific fluorescent treponemal antibody biopsy can be performed at the time of repair of rhegma-
absorbed (FTA-ABS) test should also be drawn.88 The treatment togenous retinal detachment in patients with viral retinitis. The
consists of 4 weeks of intravenous penicillin, with the same sample is taken from the margin of healed and active retinitis.
dosage used for treatment of neurosyphilis. The need for these surgical diagnostic techniques is fortunately
Hematogenous dissemination of Mycobacterium tuberculosis uncommon, as most retinal manifestations of AIDS are
to the choroid results in single or multiple round, yellow-white diagnosed clinically (Fig. 163.8).
elevated lesions with indefinite borders ranging from 0.5 to
3.0 mm in diameter in the posterior pole and midperiphery DRUG TREATMENT
(Fig. 163.7). Only a few cases have been described in the
literature.89 The diagnosis is difficult to establish the diagnosis
in the absence of systemic disease, and treatment is often
HIV THERAPY
undertaken presumptively, based on the clinical appearance, the The goals for therapy of HIV include suppression of viral
patient’s history of TB exposure and treatment even in the replication, preservation of immune function, with main-
absence of a positive chest radiograph. Tuberculous choroiditis tenance of quality of life without sacrificing a reduction in HIV-
responds well to systemic therapy with rifampin, ethambutol, related morbidity and mortality. The Department of Health and
isoniazid, and pyrazinamide. Human Services recommends treatment of HIV with a
combination of a nonnucleoside reverse transcriptase inhibitor
TUMORS or protease inhibitor with two nucleoside reverse transcriptase
inhibitors for the initiation of therapy in treatment-naïve
HIV-associated primary lymphoma of the choroid is extremely patients. This regimen has been termed highly active anti-
rare. In the reported cases, it appears as a confluent yellowish- retroviral therapy (HAART). Treatment for HIV using HAART
white retinochoroidal infiltrate with perivascular sheathing that

FIGURE 163.7. Tuberculosis nodule in the choroid. FIGURE 163.8. Toxoplasmosis scar, inactive.
2103
RETINA AND VITREOUS

Non-nucleoside Reverse Transcriptase Inhibitors


TABLE 163.3. Antiretroviral Regimens Recommended for
Non-nucleoside reverse transcriptase inhibitors are a struc-
Treatment of HIV Infection in Anti-Retroviral Naïve Patients
(Reference: JAMA (2004) 292: 251-65.) turally diverse group of agents that function as noncompetitive
inhibitors of HIV-1 reverse transcriptase and do not exhibit
Recommended Components activity against HIV-2 strains. These agents are associated with
Non-nucleoside reverse Efavirenz a hypersensitivity reaction that affects the entire class of agents.
transcriptase inhibitors Nevirapine These medications also interfere with the cytochrome P450
metabolic pathways.
Protease inhibitors Lopinavir + ritonavir
Atazanavir + low-dose ritonavir
Saquinavir + low-dose ritonavir Protease Inhibitors
Indinavir + low-dose ritonavir In 1995, inhibitors of HIV protease were first introduced and
completely altered the natural history of HIV by decreasing
Nucleoside reverse Zidovudine or tenofovir) +
transcriptase inhibitors (lamivudine or emtricitabine) morbidity and mortality when combined with other anti-
Didanosien + emtricitabine retrovirals.94 Because of their short half-life, co-administration
of ritonavir, which inhibits the cytochrome P450-mediated
Alternative Components
metabolism of these compounds, is advised. Protease inhibitors
Protease inhibitors Fosamprenavir + low-dose are often associated with gastrointestinal intolerance and
Nucleoside reverse ritonavir metabolic abnormalities.
SECTION 10

transcriptase inhibitors Atazanavir


Nelfinavir
Abacavir + lamivudine
Fusion Inhibitors
Didanosine + lamivudine This is the newest class of antiretroviral agents that works by
Didanosie + tenofovir blocking cellular invasion. It inhibits fusion of the viral envelope
Stavudine + lamivudine glycoprotein (gp41) with the host cell membrane. There is only
Zidovudine + abacavir one fusion inhibitor presently approved for HIV infection treat-
Reference: JAMA (2004) 292: 251–65. ment and it is administered as repeated subcutaneous injections.

OCULAR COMPLICATIONS OF OTHER


MEDICATIONS IN HIV
detailed in Table 163.3 is typically initiated when CD4+ cell
counts drop below 200 cells/mL or constitutional symptoms Treatment of the opportunistic infections associated with AIDS
develop. No conclusive benefit has been demonstrated for may cause ocular side effects. Rifabutin, cidofovir, and
starting the medication at CD4+ counts above 300 cells/mL. By fomivirsen may all induce uveitis.95 Ethambutol, used to treat
tracking the CD4+ count as well as the HIV-RNA viral load, tuberculosis and Mycobacterium avium infections, may cause
response to HAART can be determined. Current treatment optic neuropathy. Finally, AIDS neuropathy can affect sexual
recommendations are specified below.91 function, necessitating the use of phosphodiesterase inhibitors
(e.g., sildenafil) for erectile dysfunction. These drugs cause
Nucleoside Reverse Transcriptase Inhibitors inhibition of the nitrous oxide pathway and have been linked to
These were the first agents used to treat HIV infection and anterior ischemic optic neuropathy.96
remain integral to the management. They are competitive
inhibitors of reverse transcriptase, active against HIV-1 and SUMMARY
HIV-2, prohibiting budding of the virus from the cell. The
agents must be phosphorylated by host cell enzymes in the AIDS is a syndrome that affects all organ systems with frequent
cytoplasm to become active. Once active they are incorporated retinal findings. In particular, CMV retinitis is an AIDS-
into the proviral DNA and cause termination of the chain. defining illness as well as a prognostic indicator for mortality in
These compounds have a narrow therapeutic index secondary patients with AIDS. While CMV retinitis is the most common
to their affinity for DNA polymerases. Currently, there are eight opportunistic infection in patients with AIDS, noninfectious
available agents. They are associated with myopathy, hepatic retinal microvasculopathy is the most prevalent finding. Other
steatosis, and polyneuropathy. Macular edema has been opportunistic pathogens can destroy retinal and/or choroidal
described in a patient treated with zidovudine.92 Didanosine tissue. Aggressive treatment with specific antiviral, antibiotic,
has been linked to optic neuritis and atrophy of the retinal antifungal, or antiparasitic drugs and HAART have resulted in
pigment epithelium especially in children. Hence, quarterly reduced ocular morbidity and, in some cases, reduced mortality.
indirect ophthalmoscopy should be performed in children on Nonetheless, visual loss remains a major cause of impaired
this medication.93 quality of life in these patients.

REFERENCES
1. CDC: HIV/AIDS surveillance report. Atlanta, 5. Pantaleo G, Graziosi C, Fauci AS: New 8. Freeman WR, Lerner CW, Mines JA, et al:
Georgia: US Department of Health and concepts in the immunopathogenesis of A prospective study of the ophthalmologic
Human Services, Public Health Service: human immunodeficiency virus infection. findings in the acquired immune deficiency
CDC; 1997:(Vol 9, no. 1). N Engl J Med 1993; 328:327–335. syndrome. Am J Ophthalmol 1984;
2. UNAIDS: 2004 Report on the Global AIDS 6. Vlahov D, Graham N, Hoover D, et al: 97:133–142.
Epidemic: executive summary. Joint United Prognostic indicators for AIDS and 9. Rosenberg PR, Uliss AE, Firedland GH, et al:
Nations Programme on HIV AIDS; 2004. infectious disease death in HIV-infected Acquired immunodeficiency syndrome:
3. CDC: HIV/AIDS surveillance report 2004. injection drug users: plasma viral ophthalmic manifestations in ambulatory
Vol. 16. Atlanta, Georgia: US Department of load, CD4+ cell count. JAMA 1998; patients. Ophthalmology 1983; 90:874–878.
Health and Human Services: CDC; 2005. 279:35–40. 10. Engstrom RE, Holland GN, Hardy WD,
4. Sooqoor M, Daar ES: Primary human 7. Jabs DA: Ocular manifestations of HIV et al: Hemorheologic abnormalities in
immunodeficiency virus type 1. Curr infection. Trans Am Ophthalmol Soc 1995; patients with human immunodeficiency
2104 HIV/AIDS Rep 2005; 2:55–60. 93:623–683. virus infection and ophthalmic
Retinal Manifestations of the Acquired Immunodeficiency Syndrome: Diagnosis and Treatment

microvasculopathy. Am J Ophthalmol 1990; 27. Studies of Ocular Complications of AIDS 43. Holland GN: Immune recovery uveitis. Ocul
109:153–161. Research Group in collaboration with the Immunol Inflamm 1999; 7:215–221.
11. Mansour AM, Li H, Segal E: Picture AIDS Clinical Trials Group: 44. Henderson HWA, Mitchell SM: Treatment of
resembling hemicentral retinal vein Foscarnet–ganciclovir cytomegalovirus immune recovery vitritis with local steroids.
occlusion in the acquired immunodeficiency Retinitis Trial 5. Clinical features of Br J Ophthalmol 1999; 83:540–545.
syndrome: is it related to cytomegalovirus? cytomegalovirus retinitis at diagnosis. Am J 45. Spector SA, Hsia K, Crager M, et al:
Ophthalmologica 1996; 210:108–111. Ophthalmol 1997; 124:141–157. Cytomegalovirus (CMV) DNA load is an
12. Quiceno JI, Capparelli E, Sadun AA, et al: 28. Wei LL, Park SS, Skiest DJ: Prevalence of independent predictor of CMV disease and
Visual dysfunction without retinitis in visual symptoms among patients with survival in advanced AIDS. J Virol 1999;
patients with acquired immunodeficiency newly diagnosed cytomegalovirus retinitis. 73:7027–7030.
syndrome. Am J Ophthalmol 1992; Retina 2002; 22:278–282. 46. Jabs DA, Gilpin AMK, Min Y-I, et al: For
113:8–13. 29. Holland GN, Pepose JS, Pettit TH, et al: studies of ocular complications of aids
13. Sadun AA, Pepose JS, Madigan MC, Acquired immune deficiency syndrome. research group. HIV and CMV viral lad and
et al: AIDS-related optic neuropathy : Ocular manifestations. Ophthalmology clinical outcomes in patients with AIDS and
a histological, virological and ultrastructural 1983; 90:859–873. CMV retinitis: the Monoclonal Antibody
study. Graefes Arch Clin Exp Ophthalmol 30. McCann JD, Margolis TP, Wong MG, et al: Cytomegalovirus Retinitis Trial. AIDS 2002;
1995; 233:387–398. A sensitive and specific polymerase chain 16:877–887.
14. Yassur Y, Biedner B, Fabrikant M: Branch reaction-based assay for the diagnosis of 47. Griffiths PD: CMV as a cofactor enhancing
retinal-artery occlusion in acquired cytomegalovirus retinitis. Am J Ophthalmol progression of AIDS. J Clin Virol (2006)
immunodeficiency syndrome prodrome. 1995; 120:219–226. 35: 489–92.

CHAPTER 163
Ann Ophthalmol 1988; 20:191–195. 31. Davis JL, Serfass MS, Lai MY, et al: 48. Kempen JH, Jabs DA, Wilson LA, et al:
15. Friedman SM, Mrgo CE: Bilateral central Silicone oil in repair of retinal detachments Mortality risk for patients with
retinal vein occlusions in a patient with caused by necrotizing retinitis in HIV cytomegalovirus retinitis and acquired
acquired immunodeficiency syndrome. infection. Arch Ophthalmol 1995; immune deficiency syndrome. Clin Infect
Clinicopathologic correlation. Arch 113:1401–1409. Dis 2003; 37:1365–1373.
Ophthalmol 1995; 113:1184–1188. 32. Azen SP, Scott IU, Flynn HW Jr, et al: 49. Thorne JE, Jabs DA, Kempen JH, et al:
16. Mansour AM, Rodenko G, Dutt R: Half-life Silicone oil in the repair of complex retinal Incidence of and risk factors for visual
of cotton wool spots in the acquired detachments. A prospective observational acuity loss among patients with AIDS and
immunodeficiency syndrome. Int J STD multicenter study. Ophthalmology 1998; cytomegalovirus retinitis in the era of highly
AIDS 1990; 1:132–33. 105:1587–1597. active antiretroviral therapy. Ophthalmology
17. Park KL, Marx JL, Rao NA: Noninfectious 33. Gangan PA, Besen G, Munguia D et al: 2006; 113:1432–1440.
branch retinal vein occlusion in human Macular serous exudation in patients with 50. Kempen JH, Jabs DA, Dunn JP, et al:
immunodeficiency virus-positive patients. acquired immunodeficiency syndrome and Retinal detachment risk in cytomegalovirus
Retina 1997; 17:162–165. cytomegalovirus retinitis. Am J Ophthalmol retinitis related to the acquired immune
18. Dunn JP, Yamashita A, Kempen JH, Jabs 1994; 118:212–219. deficiency syndrome. Arch Ophthalmol
DA: Retinal vascular occlusion in patients 34. Karavellas MP, Plummer DJ, MacDonald JC, 2001; 119:33–40.
infected with human immunodeficiency et al: Incidence of immune recovery vitritis 51. Jabs DA, Van Natta ML, Thorne JE, et al:
virus. Retina 2005; 25:759–766. in cytomegalovirus retinitis patients Course of cytomegalovirus retinitis in the
19. Yung CW, Harris A, Massicotte S, et al: following institution of successful highly era of highly active antiretroviral therapy. 1.
Retinal blood flow indices in patients active antiretroviral therapy. J Infect Dis Retinitis progression. Ophthalmology 2004;
infected with human immunodeficiency 1999; 179:697–700. 111:2224–2231.
virus. Br J Ophthalmol 1996; 80:723–727. 35. Kempen JH, Min YI, Freeman WR, et al: 52. Jabs DA, Van Natta ML, Thorne JE, et al:
20. Dejaco-Ruhswurm I, Kiss B, Rainer G, et al: Risk of immune recovery uveitis in patients Course of cytomegalovirus retinitis in the
Ocular blood flow in patients infected with with AIDS and cytomegalovirus retinitis. era of highly active antiretroviral therapy. II.
human immunodeficiency virus. Am J Ophthalmology 2006; 113:684–694. Second eye involvement and retinal
Ophthalmol 2001; 132:719–725. 36. Karavellas MP, Lowder CY, MacDonald JC, detachment. Ophthalmology 2004;
21. Horwitz CA, Henle W, Henle G, et al: et al: Immune recovery vitritis associated 111:2232–2239.
Heterophil-negative infectious with inactive cytomegalovirus retinitis. 53. Kempen JH, Jabs DA, Wilson LA, et al:
mononucleosis and mononucleosis-like A new syndrome. Arch Ophthalmol 1998; Incidence of cytomegalovirus (CMV)
illnesses. Laboratory confirmation of 43 116:169–75. retinitis in second eyes of patients with the
cases. Am J Med 1977; 63:947–957. 37. Zegans ME, Walton RC, Holland GN, et al: acquired immune deficiency syndrome and
22. Hoover DR, Peng Y, Saah A, et al: Transient vitreous inflammatory reactions unilateral CMV retinitis. Am J Ophthalmol
Occurrence of cytomegalovirus retinitis associated with combination antiretroviral 2005; 139:1028–1034.
after human immunodeficiency virus therapy in patients with AIDS and 54. Jabs DA, Holbrook JT, Van Natta ML, et al:
immunosuppression. Arch Ophthalmol cytomegalovirus retinitis. Am J Ophthalmol Risk factors for mortality in patients with
1996; 114:821–827. 1998; 125:292–300. AIDS in the era of highly active antiretroviral
23. Gallant JE, Moore RD, Richman DD, et al: 38. Karavellas MP, Song M, MacDonald JC, therapy. Ophthalmology 2005;
Incidence and natural history of et al: Long-term posterior and anterior 112:771–779.
Cytomegalovirus disease in patients with segment complications of immune recovery 55. Whitcup SM, Fortin E, Lindblad AS, et al:
advanced human immunodeficiency virus uveitis associated with cytomegalovirus Discontinuation of anticytomegalovirus
disease treated with zidovudine. The retinitis. Am J Ophthalmol 2000; 130:57–64. therapy in patients with HIV infection and
Zidovudine Epidemiology Study Group. 39. Nguyen QD, Kempen JH, Bolton SG, et al: cytomegalovirus retinitis. JAMA 1999;
J Infect Dis 1992; 166:1223–1227. Immune recovery uveitis in patients with 282:1633–1637.
24. Baldassano V, Dunn JP, Feinberg J, AIDS and cytomegalovirus retinitis after 56. Wohl DA, Kendall MA, Owens S, et al: The
Jabs DA: Cytomegalovirus retinitis and low highly active anti-retroviral therapy. Am J safety of discontinuation of maintenance
CD4 T-lymphocyte counts. New England J Ophthalmol 2000; 129:634–639. therapy for cytomegalovirus (CMV) retinitis
Med 1995; 333:670. 40. Nussenblatt RB, Lane HC: Human and incidence of immune recovery uveitis
25. Egbert PR, Pollard RB, Gallagher JG, immunodeficiency virus disease: changing following potent antiretroviral therapy. HIV
et al: Cytomegalovirus retinitis in patterns of intraocular inflammation. Am J Clin Trials 2005; 6(3):136–146.
immunosupressed hosts. II. Ocular Ophthalmol 1998; 125:374–382. 57. Lin DY, Warren JF, Lazzeroni LC, et al:
manifestations. Ann Intern Med 1980; 41. Karavellas MP, Azen SP, MacDonald JP, et Cytomegalovirus retinitis after initiation of
93:664–670. al: Immune recovery vitritis and uveitis in highly active antiretroviral therapy in HIV
26. Jacobson MA, Stanley H, Holtzer C, AIDS: clinical predictors, sequelae , and infected patients: natural history and clinical
et al: Natural history and outcome of new treatment outcomes. Retina 2001; 21:1–9. predictors. Retina 2002; 22:268–277.
AIDS-related cytomegalovirus retinitis 42. Llatjos M, Romeu J, Clotet B, et al: A 58. Drew WL, Ives D, Lalezari JP, et al: Oral
diagnosed in the era of highly active distinctive cytologic pattern for diagnosing ganciclovir as maintenance treatment for
antiretroviral therapy. Clin Infect Dis 2000; tuberculous lymphadenitis in AIDS. J AIDS cytomegalovirus retinitis in patients with
30:231–233. 1993; 6:1335–1338. AIDS. New Engl J Med 1995; 333:615–620. 2105
RETINA AND VITREOUS

59. Martin DF, Sierra-Madero J, Walmsley S, failures of potent antiretroviral therapy. and review. Clin Infect Dis 1992;
et al: A controlled trial of valganciclovir as AIDS 2000; 14:173–180. 14:1069–1073.
induction therapy for cytomegalovirus 72. Combination foscarnet and ganciclovir 84. Denning DW, Follansbee SE, Scolaro M,
retinitis. N Engl J Med 2002; therapy vs monotherapy for the treatment et al: Pulmonary aspergillosis in the
346:1119–1126. of relapsed cytomegalovirus retinitis in acquired immunodeficiency syndrome.
60. Lalezari J, Lindley J, Walmsely S, et al: patients with AIDS: The Cytomegalovirus N Engl J Med 1991; 324:654–662.
A safety study of oral valganciclovir retreatment trial. The studies of ocular 85. Fish DG, Ampel NM, Galgiani JN, et al:
maintenance treatment of cytomegalovirus complications of AIDS research group in Coccidioidomycosis during human
retinitis. J Acquir Immune Defic Syndr collaboration with the AIDS clinical trials immunodeficiency virus infection: a review
2002; 30:392–400. group. Arch Ophthalmol 1996; 114:23–33. of 77 patients. Medicine 1990; 69:384–391.
61. Sandorn GE, Anand R, Torti RE, et al: 73. Emery VC, Hassan–Walker AF: Focus on 86. Macher A, Rodrigues MM, Kaplan W, et al:
Sustained-release ganciclovir therapy for new drugs in development against human Disseminated bilateral chorioretinitis due to
treatment of cytomegalovirus retinitis: cytomegalovirus. Drugs 2002; histoplasma capsulatum in a patient with
use of an intravitreal device. Arch 62:1853–1858. the acquired immunodeficiency syndrome.
Ophthalmol ;992) 110:188–195. 74. Engstrom RJ, Holland GN, Margolis TP, Ophthalmology 1985; 92:1159–1164.
62. Musch DC, Martin DF, Gordon JF, et al: et al: The progressive outer retinal necrosis 87. Finamore LP, Muccioli C, Martins MC, et al:
Treatment of cytomegalovirus retinitis with syndrome. A variant of necrotizing herpetic Ocular and central nervous system
a sustained release ganciclovir implant. retinopathy in patients with AIDS. paracoccidioidomycosis in a pregnant
N Engl J Med 1997; 337:83–90. Ophthalmology 1994; 101:1488–1502. woman with acquired immunodeficiency
63. Martidis A, Danis RP, Ciulla TA: Treating 75. Ciulla TA, Rutledge BK, Morley MG, et al: syndrome. AJO 2002; 134:456–459.
cytomegalovirus retinitis-related retinal The progressive outer retinal necrosis 88. McLeish WM, Pulido JS, Holland S, et al:
SECTION 10

detachment by combining silicone oil syndrome: successful treatment with The ocular manifestations of syphilis in
tamponade and ganciclovir implant. combination antiviral therapy. Ophthalmic the human immunodeficiency virus type
Ophthalmic Surg Lasers 2002; 33:135–139. Surg Lasers 1998; 29:198–206. 1-infected host. Ophthalmology 1990;
64. Charles NC, Freisberg L: Endophthalmitis 76. Scott IU, Luu KM, Davis JL: Intraviteal 97:196–203.
associated with extrusion of a ganciclovir antivirals in the management of patients 89. Recillas-Gispert C, Ortega-Larrocea G,
implant. Am J Ophthalmol 2002; with acquired immunodeficiency syndrome Arrelanes-Garicia L, et al: Chorioretinitis
133:273–275. with progressive outer retinal necrosis. secondary to Mycobacterium tuberculosis
65. Ambati J, Wynne KB, Angerame MC, Arch Ophthalmol 2002; 120:1219–1222. in acquired immune deficiency syndrome.
Robinson MR: Anterior uveitis associated 77. Holland GN: Standard diagnostic criteria Retina 1997;: 17:437–439.
with intravenous cidofovir use in patients for the acute retinal necrosis syndrome. 90. Matzkin DC, Samovits TL, Rosenbaum PS:
with cytomegalovirus retinitis. Br J Executive Committee of the American Simultaneous intraocular and orbital non-
Ophthalmol 1999; 83:1153–1158. Uveitis Society. Am J Ophthalmol 1994; Hodgkin lymphoma in the acquired immune
66. Akler ME, Johnson DW, Burman WJ, et al: 117:663–667. deficiency syndrome. Ophthalmology 1994;
Anterior uveitis and hypopyon after 78. Shami MJ, Freeman WR, Friedberg D, et al: 101:850–855.
intravenous cidofovir for the treatment of A multicenter study of pneumocystis 91. Rathbun RC, Lockhart SM, Stephens JR:
cytomegalovirus retinitis. Ophthalmology choroidopathy. Am J Ophthalmol 1991; Current HIV treatment guidelines-An
1998; 105:651–657. 112:15–22. overview. Current Pharmaceutical Design
67. Davis JL, Taskintua I, Freeman WR, et al: 79. Rao NA, Zimmerman PL, Boyer D, et al: 2006; 12:1045–1063.
Iritis and hypotony after treatment with A clinical, histopathologic, and electron 92. Lafeuillade A, Aubert L, Chaffanjon P, et al:
intravenous cidofovir for cytomegalovirus microscopic study of Pneumocystis carinii Optic neuritis associated with
retinitis. Arch Ophthalmol 1997; choroiditis. Am J Ophthalmol 1989; dideoxinosine. Lancet 1991; 337:615.
115:733–737. 107:218–228. 93. Whitcup SM, Butler KM, Caruso R, et al:
68. Smith IL, Taskintuna I, Rahhal FM, et al: 80. Fardeau C, Romand S, Rao NA, et al: Retinal toxicity in human immunodeficiency
Clinical failure of CMV retinitis with Diagnosis of toxoplasmic retinochoroiditis virus-infected children treated with
intravitreal cidofovir is associated with with atypical clinical features. Am J 2,3-dideoxinosine. Am J Ophthalmol 1992;
antiviral resistance. Arch Ophthalmol 1998; Ophthalmol 2002; 134:196–203. 113:1–7.
116:178–185. 81. Bosch–Driessen LH, Verbraak FD, 94. Palella FJ, Delaney KM, Moorman AC, et al:
69. Jabs DA, Martin BK, Formann MS, et al: Suttorp–Schulten MSA, et al: A prospective, Declining morbidity and mortality among
Cytomegalovirus resistance to ganciclovir randomized trial of pyrimethamine and patients with advanced human
and clinical outcomes of patients with azithromycin vs pyrimethamine and immunodeficiency virus infection. HIV
cytomegalovirus retinitis. Am J Ophthalmol sulfadiazine for the treatment of ocular Outpatient Study Investigators. N Engl J
2003; 135:26–34. toxoplasmosis. Am J Ophthalmol 2002; Med 1998; 338:853–860.
70. Jabs DA, Martin BK, Forman MS, Ricks 134:34–40. 95. Arevalo JF, Russack V, Freeman WR: New
MO: Cytomegalovirus (CMV) blood DNA 82. Kovacs JA: Efficacy of atovaquone in ophthalmic manifestations of presumed
load, CMV retinitis progression, and treatment of toxoplasmosis in patients rifabtin-related uveitis. Opthalmic Surg
occurrence of resistant CMV in patients with AIDS. The NIAID-Clinical Center Lasers 1997; 28:321–324.
with CMV retinitis. JID 2005; 192:640–649. Intramural AIDS Program. Lancet 1992; 96. Egan R, Pomeranz H: Sildenafil (Viagra)
71. Torriani FJ, Freeman WR, MacDonald JC, 340:637–638. associated anterior ischemic optic
et al: CMV retinitis recurs after stopping 83. Crump JRC, Elner SG, Elner VM, et al: neuropathy. Arch Ophthalmol 2000;
treatment in virological and immunological Cryptococcal endophthalmitis : case report 118:291–292.

2106
CHAPTER

164 Acute Retinal Necrosis


Janet L. Davis, Gregory M. Fox, and Mark S. Blumenkranz

Initially described in Japan in 1971 by Urayama and colleagues retinal infection with varicella-zoster virus, PORN is charac-
and termed Kirisawa’s uveitis, acute retinal necrosis (ARN) is a terized by the initial appearance of multifocal areas of outer
dramatic uveitic syndrome that presents with vitreous inflam- retinal opacification (with little other intraocular inflam-
mation, retinal periarteritis, optic neuropathy, and confluent mation), rapid progression of the lesions to confluence with
peripheral necrotizing retinal infiltrates.1–7 This important full-thickness retinal necrosis, and poor response to antiviral
clinical syndrome has since been documented to represent the therapy.23,24
consequences of acute infection of the retina and associated
ocular tissues by certain types of Herpesvirus hominis.8,9 In
order to promote clarity of definition, the American Uveitis Terminology
Society has suggested that the term acute retinal necrosis be • ARN syndrome
used to designate only those cases that conform to a well- • Peripheral retinal necrosis
described clinical syndrome, characterized by peripheral retinal • Rapid, circumferential spread
necrosis with discrete borders, rapid progression with circum- • Occlusive vasculopathy
ferential spread in untreated eyes, occlusive vasculopathy with • Optional: optic neuropathy, vitreitis or iritis, episcleritis, or
arteriolar involvement, and prominent inflammation in the pain
vitreous and anterior chamber. Optic neuropathy or atrophy, • Necrotizing herpetic retinopathy
scleritis, and pain are well-described features in some patients • Retinal necrosis due to herpes simplex or varicella-zoster
but are not necessary to make the diagnosis.2,10 virus
The demonstration of a viral cause for this syndrome has • Not necessary to meet criteria for ARN syndrome
led to the important development of specific pharmacologic • Specify virus if known
therapies; however, late retinal detachment continues to be a • Progressive outer retinal necrosis
serious complication.11,12 Fortunately, modern vitreoretinal • Colloquial term for necrotizing herpetic retinopathy with a
surgical techniques continue to improve the prognosis for paucity of inflammatory signs
retinal reattachment and for recovery of visual acuity.14 Prompt • Usually in immunocompromised patients
treatment with antiviral agents and prophylactic retinal photo- • ARN-like retinitis
coagulation may also lessen the risk of disease in the fellow eye • Features of ARN syndrome
and the incidence of late retinal detachment in the initially • Causative agent is cytomegalovirus, toxoplasmosis,
involved eye.15,16 Experimental animal studies have shed further Epstein–Barr virus, or syphilis
light on the mode of viral transmission between the eye and the • Other infectious and noninfectious causes of confluent
central nervous system and on the contribution of immuno- retinal inflammation
regulatory events to the pathophysiology of this disease.17,18
In selected cases in which optic neuropathy seems to be
responsible for the large proportion of visual-field and visual- EPIDEMIOLOGY
acuity loss, optic nerve sheath decompression has been reported
to be worth considering.19 Initially described in Asia,2 ARN has also been documented
Since the original descriptions of ARN, atypical cases of in both Europe3,20–22 and North America.4–9 Limited prevalence
herpetic retinitis have also been described that do not fit the data are available for this disease, but the incidence does appear
classic syndrome, including more indolent forms of necrotizing to be rising. There were no reported cases before 1971, although
retinitis in some patients with varicella.20–22 The American it now seems likely that previously cases either were not
Uveitis Society suggests that any cases of herpetic retinitis that recognized or were misclassified as other forms of posterior
do not fit the clinical syndrome of ARN be designated as uveitis. By 1982, there were 41 cases reported in the world
necrotizing herpetic retinopathy. If the specific viral cause is literature,6 and by 1996, more than 150 articles had been
known, the causative viral name can be added as a modifier.10 devoted to the syndrome. An epidemiologic study of uveitis in
Although typical features of ARN have been reported in patients Switzerland found that cases of ARN represented 2.5% of cases
with human immunodeficiency virus (HIV) infection, herpetic of uveitis seen at the uveitis clinic of the Hospital Jules Gonin
retinitis in patients with the acquired immunodeficiency syn- between January 1990 and March 1993. The incidence of ARN
drome (AIDS) frequently present with atypical features. An for the referral area studied was calculated to be 4.25 per
important devastating syndrome of necrotizing retinitis in 1 000 000 inhabitants per year.25
patients with AIDS has been described by the term progressive Men contract ARN more often than women, and more adults
outer retinal necrosis (PORN). Believed to be largely caused by are affected than children. In one report, 63% of patients were 2107
RETINA AND VITREOUS

male and 37% were female; none was younger than 13 years of However, the difference does not retain statistical significance
age.6 Since that report, an 11-year-old girl with HIV infection, after analysis in a chi-squared test with Yates correction for the
acquired at birth from her infected mother developed an number of HLA antigens that were tested.42 Klok and asso-
atypical case of necrotizing retinitis after contracting chicken- ciates43 identified the presence of anticardiolipin antibodies
pox.26 The disease may affect one or both eyes, with the initial in the sera of seven out of 24 patients with ARN versus 10%
presentation being unilateral in approximately two-thirds of of healthy control patients. The importance of anticardiolipin
cases.6,27 When the involvement is bilateral, there is generally antibodies in the pathogenesis of ARN in these patients is
a delay between involvement of the first and the second eyes, uncertain.
ranging from 1–6 weeks.2,28 The timing of involvement of the In addition to differences in underlying genetic predisposition, it
second eye is consistent with the theories on the propagation is likely that other host factors and viral factors may play a role in
and transfer of virus from one eye to another via the optic nerve the development of ARN and may influence the incidence of
and central nervous system. Delays as long as 3028 and 3429 unfavorable clinical outcomes. Recent periocular trauma has been
years between involvement of the first and the second eye have reported to precede the onset of ARN in a few patients.44,45 Clinical
been reported in patients not treated with acyclovir during signs such as diffuse arteritis, at the optic disc or beyond the area
involvement of the first eye. The risk of development of disease of retinitis, reduced electroretinographic a- and b-waves, elevated
in the fellow eye in patients not treated with intravenous levels of circulating immune complexes, and larger zones of
acyclovir has been estimated to be as high as 65% 2 years after necrosis and exudation are associated with an especially poor
the onset of the disease.15 prognosis, suggesting that the immunologic response to the
SECTION 10

The disease was initially described in otherwise healthy virus or intrinsic factors may strongly affect the outcome.46
patients,1–7 and immunocompetence has been judged to be one
criterion for establishing the diagnosis.13 This characteristic has CLINICAL FEATURES
distinguished ARN from other forms of opportunistic retinitis
in patients with incompetent immune systems or in those with Although the most dramatic and visually devastating aspect of
concurrent herpetic disease of the central nervous system.30,31 this disease is the confluent necrotizing retinitis from which the
Subsequent reports have documented that immunosuppressed syndrome draws its name, most ocular tissues are concurrently
patients may experience a clinical syndrome of ARN that is affected. The disease can be divided into two different phases:
identical to that seen in immunocompetent patients and is (1) the acute herpetic phase, which lasts ~4–8 weeks; and (2)
readily distinguished from other types of opportunistic retinitis, the late cicatricial phase, occurring after the onset of the disease
including cytomegalovirus (CMV) retinitis.32–34 and characterized by the organization of the vitreous and devel-
opment of large retinal tears, retinal detachment, and pro-
HOST FACTORS liferative vitreoretinopathy (PVR).

Most cases of ARN probably represent primary infection of the


retina with virus reactivated from dormant sites, including gan- DISEASE TYPES
glionic tissue. Serum neutralizing antibodies to the herpes zoster Increased awareness and recognition of ARN have enabled
varicella virus group are present in up to 95% of adults,1,35 and clinicians to make increasingly subtle distinctions between
it is likely that host factors predispose certain individuals to the ARN, as defined by the American Uveitis Society,10 and other
development of the syndrome of ARN. For example, although in variants of necrotizing retinitis. These variants include a ‘mild
excess of 3 million persons each year experience chickenpox, to form which is more indolent in character and not associated
date there have been 10 patients reported with ARN as a with retinal detachment’,46 and a severe, rapidly progressive
short–term complication of varicella.20,21,26,36 Similarly, although syndrome of outer retinal necrosis associated with little vitritis,
ARN has been reported to occur as an early complication of seen in patients with HIV infection, that is distinct from CMV
herpes zoster in a small number of patients, this is also believed retinitis.23,24,34,47 Moreover, there is some evidence to suggest
to be an extremely infrequent problem in immunocompetent that the disease course and clinical profile of patients with ARN
patients with cutaneous herpes zoster.37,38 In one prospective may differ, depending on whether the causative agent is herpes
randomized series of patients with herpes zoster ophthalmicus simplex virus (HSV) or the herpes varicella virus group.22
(HZO), none of the 71 patients treated with either acyclovir or
placebo experienced ARN, although in another large series,
10–33% of patients with HZO demonstrated other forms of ACUTE HERPETIC PHASE
viral dissemination.39 In contrast, the risk of a patient with HIV Presenting Symptoms
infection and HZO developing ARN is likely substantial. Sellitti The onset of ARN is frequently heralded by the development
and associates40 cited an incidence of ARN in 17% of patients of ocular pain, diminished vision, floaters, and external ocular
with HIV infection and HZO. The majority of these patients injection. The patient may give a history of recent or remote
developed bilateral retinal necrosis, and all patients presented herpes zoster or varicella infection in the majority of cases or
with retinal necrosis within 9 months of the onset of HZO. may occasionally volunteer no history of prior herpetic infection.
It is likely that patients who experience ARN may have an
underlying immunogenetic predisposition to the disease. Holland Anterior Segment Examination
and colleagues41 demonstrated a statistically significant increase The conjunctiva is invariably injected with limbal flush, epis-
in the frequency of human leukocyte antigen (HLA) -DQw7 in cleritis, or scleritis. Anterior chamber cell and flare are frequent
patients with ARN (55%) versus control patients (19%) (P in conjunction with granulomatous keratinous precipitates (Fig.
< 0.0004; relative risk 5.2). In this study, the HLA phenotype 164.1). Although a plasmoid aqueous or mild fibrin strands
Bw62,DR4 was also more frequent than in the controls (16% vs may be seen, iris nodules or hypopyon rare. One clinical feature
2.6%; relative risk 7.49). Moreover, the HLA phenotypes of that may assist in the differentiation of the early phase of ARN
patients may have some effect on the severity of the clinical from other causes of acute granulomatous anterior uveitis is the
disease. Matsuo and Matsuo42 measured an increased frequency frequent occurrence of ocular hypertension, which is also seen
of HLA-DR9 in their patients with fulminant ARN (50%) as in herpes zoster keratouveitis. Herpetic corneal epithelial
2108 compared with their patients with mild ARN (0%) (P < 0.05). dendrites or disciform corneal lesions are not encountered,
Acute Retinal Necrosis

CHAPTER 164
FIGURE 164.1. Granulomatous keratic pzrecipitates are seen in a slit- FIGURE 164.2. Nummular lesion of ARN superonasal and inferonasal
lamp photomicrograph of a middle-aged man with the early phases of to the optic nerve in a middle-aged man with associated herpes
acute retinal necrosis (ARN). zoster involving the upper extremity.

although some patients with concurrent HZO may tion, depending on their stage of evolution. Although usually
demonstrate typical vesicular lesions in the distribution of the seen in the mid-periphery and preequatorial regions, small
affected fifth dermatome. The lens is typically unaffected aside nummular lesions may also be seen in the posterior retina,
from the presence of inflammatory precipitates on its surfaces, generally sparing the macula (Fig. 164.2). In distinction to the
which are responsive to steroid therapy. If untreated, posterior granular white dots frequently associated with CMV inclusion
synechiae and pupillary seclusion may occur in addition to disease of the retina, the earliest lesions of ARN are not
complicated cataract formation. prominently associated with venules or perivenous hemorrhage.
With progression of the syndrome, the granular and nummular
Vitreous Examination lesions increase in size and coalesce to form confluent zones of
Vitreous cellular infiltration and protein exudation are charac- full-thickness retinal necrosis. These lesions, which are
teristic features of the acute phases of the syndrome. In the typically situated in the retinal periphery, may occupy as little
earliest stages, the inflammatory infiltrate in the vitreous may as 1–2 clock hours or may extend to completely encircle the
be relatively mild, increasing as cellular immunity to the virus retina over 360° (Fig. 164.3). The mature lesions have a yellow
develops. The vitreous cellular infiltrate is composed predomi- to white homogeneous color with a somewhat jagged or dentate
nantly of mononuclear cells including lymphocytes and plasma posterior border in portions extending more posteriorly (Fig.
cells, paralleling the infiltrates seen in other ocular tissues.1 A 164.4). They obstruct visualization of the underlying retinal
viral cytopathic effect has been demonstrated from culture of pigment epithelium and choroid. The lesions end abruptly at
vitreous specimens obtained at the time of enucleation during the ora serrata anteriorly and do not appear to parallel or
the acute phase of the disease.8,74 Polyclonal anti-HSV otherwise respect the retinal vasculature, as is frequently seen
antibodies have been identified from vitrectomy specimens in in CMV inclusion disease. The retinal lesions that are seen
patients with the active phase of the disease.13 Aqueous early in the disease arise abruptly and may progress in size and
specimens from patients with the disease similarly demonstrate number over approximately a 2-week period in untreated patients.
antibodies to either HSV-1 and the herpes zoster varicella virus The lesions then begin a period of regression characterized by
group in ratios suggesting local intraocular antibody loss of retinal opacification, thinning, and pigmentary scarring
production.8,13,14,70,72,90 High levels of cytokines such as (Fig. 164.5). The loss of opacification, which correlates with the
interleukin (IL)-6 have also been found in the vitreous in resolution of the inflammatory and viral infiltration of the
patients with ARN.48 retina, may initially be seen as perivascular curvilinear lucencies
within zones of confluent necrosis. This has been termed a
Retina Examination Swiss-cheese appearance (Fig. 164.6). Over the course of several
Retina examination may reveal two characteristic features: ensuing weeks, these zones of retinal lucency enlarge in
confluent zones of opaque necrotizing retinitis predominantly conjunction with the development of pigmentary changes in the
involving the periphery and generalized retinal arteritis asso- retinal pigment epithelium and neural retina.
ciated with capillary vasoocclusion. The pigmentary changes that characterize the convalescent
phase of ARN are most prominent initially at the posterior
Necrotizing retinitis margins of the lesions and progress outward in a centrifugal
The earliest retinal lesions are subtle, isolated retinal opacities fashion coincident with the development of lucencies. The
that may assume a patchy, granular or nummular configura- character of the pigmentary response associated with the 2109
SECTION 10 RETINA AND VITREOUS

FIGURE 164.3. Typical lesions of ARN involving the periphery. Note


the confluent white–yellow appearance with an irregular scalloped
posterior margin and a sharp transition between involved and
FIGURE 164.5. The same region as shown in Figure 164.4, ~4 months
uninvolved portions.
later, demonstrates clearing of ocular media opacity and better
visualization of retinal vascular detail. Note the atrophic pigmented
zone corresponding to the area of prior necrosis with some fibroglial
proliferative change overlying the center of scar.

FIGURE 164.4. Area of confluent necrotizing retinitis with early


depigmentation at the posterior margin signifies the beginning of the
resolution phase.
FIGURE 164.6. Midresolution phase of ARN with the development of
curvilinear perivascular lucencies within zones of necrosis before the
development of late pigmentary changes (Swiss-cheese appearance).
resolution of ARN is similar to that associated with other forms
of viral retinitis, including rubella and CMV, and is distinct
from either the heavily pigmented hyperplastic response typical its presence beneath areas of opaque retina, and the presence
of toxoplasmosis or the atrophic scalloped appearance asso- of shifting subretinal fluid or xanthochromia.
ciated with ocular histoplasmosis. In zones in which there has
been particularly severe retinal inflammation and underlying Retinal vasculitis
choroidal inflammation, the degree of postnecrotic thinning The second major feature of retinal involvement in this disorder
and retinal pigment epithelial atrophy may be prominent. In is the development of severe vasoocclusive changes predomi-
some patients in the convalescent phase, giant retinal pigment nantly involving the arterial system. The presence of arteritis,
epithelial tears can develop, leaving large areas devoid of retinal rather than phlebitis, and the associated peripheral vaso-
pigment epithelium.49 In other zones in which the necrotizing occlusion seen in this disease help to distinguish it from CMV
retinitis was more mild, it may be difficult to identify the infection of the retina on ophthalmoscopic grounds. Although
original margins of involvement in the later phases of the occasionally seen in ocular toxoplasmosis and syphilis, retinal
disease. In some instances, the acute phase of the disease may arteritis is particularly striking and severe in ARN and was
be accompanied by the development of a peripheral exudative recognized to be a characteristic feature of herpes zoster infec-
retinal detachment. This may be distinguished from a rheg- tion of the retina even before the widespread recognition of the
2110 matogenous or complex detachment by the lack of retinal holes, syndrome ARN in the English-language literature (Fig. 164.7).50
Acute Retinal Necrosis

CHAPTER 164
FIGURE 164.8. Patient with confirmed herpes zoster infection of the
retina producing ARN. Note the prominent hemorrhagic component
FIGURE 164.7. Artist’s representation of a patient with the classic suggestive of concomitant retinal venous obstruction.
features of ARN, including confluent necrotizing retinitis, posterior
nummular infiltrates, and retinal arteritis (with involved vessels seen in
yellow).

Fluorescein angiographic and histopathologic studies on eyes


with ARN confirm that the arteritis is not confined to the
retinal vessels and may be seen in virtually all tissues studied,
including the iris, ciliary body, choroid, and optic nerve. The
ophthalmoscopic features of retinal periarteritis include
opacification and refractile changes in the walls of the larger
retinal arterioles; ophthalmoscopically visible nonperfusion and
obliteration of the smaller, more distal ramifications; and retinal
capillary nonperfusion best demonstrated by fluorescein
angiography.
The retinal opacification seen in this syndrome is distinct
from the cloudy swelling commonly associated with retinal
arteriolar obstruction, although the latter may contribute to this
phenomenon in addition to the cellular infiltration seen
histopathologically. Despite the presence of peripheral retinal
capillary nonperfusion, retinal, optic nerve, and iris neovascu-
larization are distinctly uncommon in this disease, as con-
trasted with other forms of vasoocclusive retinopathies.51,52 The
FIGURE 164.9. Fluorescein angiogram of a patient with ARN.
reasons for this observation are unknown but may reflect the
Courtesy of Jay Duker.
concurrent development of retinal and retinal pigment epithelial
necrosis in conjunction with nonperfusion, resulting in
decreased production of intraocular angiogenic factors. Although
large zones of intraretinal hemorrhage are not a characteristic (Fig. 164.10). To date, despite the early marked inflammatory
feature of ARN, they may occur in patients in association with changes seen in the choroid, herpesvirus particles have been
zones of phlebitis or frank venous occlusion. Rarely, the fundus identified only within the choriocapillaris and choroid by in situ
appearance may be that of a combined central retinal artery and DNA hybridization and immunohistochemical stains in a blind
vein occlusion when there has been severe involvement of the eye with burned-out disease.53 Thus, the majority of early
optic nerve and associated vasculature (Fig. 164.8). choroidal changes are likely immunocytopathologic rather than
the effect of direct viral infection.
Choroidal Involvement
The choroidal vasculature is actively involved in this syndrome. Optic Nerve Examination
Before the development of opaque confluent retinal lesions, it is Optic neuropathy is a frequent and serious complication of
possible to identify more subtle opacifications in the outer retina ARN. Some degree of optic disk swelling is seen in most cases
and underlying choroid and pigment epithelium. Early frame and may be associated with engorgement of the disk capillaries,
angiograms document the presence of focal areas of choroidal swelling of the neural tissue, and distention of the optic nerve
hypoperfusion with intense late staining suggestive of infil- sheath (Fig. 164.11).19,54 In some instances, this may result in
tration and ischemia (Fig. 164.9). These frequently presage profound vision loss. The mechanism of vision loss associated
overlying zones of necrotizing retinitis. In histopathologic with optic neuropathy in ARN remains somewhat uncertain. It
specimens, the choroidal thickness may be increased three- to may reflect one of multiple causes, including direct viral infec-
fourfold in conjunction with diffuse plasma cell, macrophagic, tion of the optic nerve,8,17,19 optic nerve ischemia secondary to
and lymphocytic stromal infiltrates and perivascular cuffing vasooclusion,8 or the result of compression by distention of the 2111
RETINA AND VITREOUS

Key Features: Signs and Symptoms of Acute Phase of


ARN
• Symptoms: Pain, blurred vision, floaters, constricted field
Signs:
• Anterior segment hyperemia and iritis
• Vitreous cells and flare
• Peripheral confluent retinal necrosis
• Satellite lesions
• Optic nerve edema or pallor
• Retinal vascular occlusions
• Associated findings
• RPE scarring anterior to the active border
• Serous retinal detachment
• Rhegmatogenous retinal detachment (uncommon)
• Recent or current meningitis or encephalitis
SECTION 10

FIGURE 164.10. Histopathologic specimen demonstrates marked over the course of 1–3 months, depending on the severity of the
choroidal stromal thickening and lymphocytic infiltration including initial infection and other factors. After resolution of the active
larger choroidal vessels. H & E µ100.
phase of the disease, secondary cicatricial changes occur, which
frequently lead to the development of late retinal tears, retinal
detachment, and loss of useful visual function.
The phase of active viral replication is believed to be controlled
by normal host humoral and cellular immune mechanisms. In
patients not treated with oral acyclovir, corticosteroids, or other
pharmacologic agents, the time course for resolution of the
active phase may be 6–12 weeks. In patients treated with oral
acyclovir, this time phase may be shortened to 4–6 weeks,
although to date no prospective studies confirming this have
been performed. It is believed that the breakdown in the
blood–ocular barrier and secondary cellular and humoral infil-
tration of the vitreous contribute to the development of late
organizational changes in the vitreous body. Collagenous and
cellular membranes develop, composed of pigment epithelial
cells and fibroblastic elements develop (Fig. 164.12).28,55
Additionally, secondary growth of pigment epithelial, glial, and
fibroblastic membranes on the surface of the retina and con-
tinuous with the organized vitreal membranes occurs in the
periphery. The concurrence of peripheral retinal thinning,
membrane formation, and vitreous contraction contributes to
the development of severe PVR frequently seen in patients in
the cicatricial stages of ARN (Figs 164.13 and 164.14).
FIGURE 164.11. Photographs of severe optic nerve involvement in a Irrespective of acyclovir therapy, late retinal detachment occurs
patient with ARN. Visual acuity is reduced to light perception despite in 75–86% of eyes with ARN.7,11
the lack of obvious macular involvement. Media opacity secondary to Prophylactic photocoagulation during the acute phase of the
vitritis is not consistent with the level of vision dysfunction. disease may reduce the risk of late retinal detachment in some
patients.13,14,17 Although retinal detachment in this condition
has been associated with a generally unfavorable prognosis,2,14
nerve sheath.19 Optic nerve sheath fenestration has been vitreoretinal surgical therapy has improved the outlook (see
proposed as a method of therapy for patients who demonstrate section on Management of Retinal Detachment). Other late
optic nerve sheath distention by neuroradiologic imaging studies. cicatricial complications of the syndrome include iris atrophy,
This has been associated with dramatic visual improvement in cataract, ciliary body fibrosis and hyposecretion, visually signi-
some instances. In one clinical series, 47% of eyes in patients ficant vitreous opacities, macular pucker, and giant tears of the
with ARN demonstrated neuropathy severe enough to warrant retinal pigment epithelium.49
consideration of surgical intervention.54 Optic neuropathy in
ARN that is sufficiently severe to warrant consideration of
surgical optic sheath decompression has been defined as (1) the OTHER TYPES OF NECROTIZING RETINITIS
presence of an afferent pupillary defect not consistent with Necrotizing Herpetic Retinopathy, Varicella
retinal findings, (2) poor correlation between retinal findings
Associated
and visual acuity and visual field, and (3) sudden deterioration
of visual acuity not corresponding to retinal changes. In addition to the classic form of ARN, patients may exhibit
both more benign and more fulminant degrees of ocular invol-
vement in retinal infections caused by the varicella-zoster
CICATRICIAL PHASE group. Matsuo and co-workers46 described six patients with
With or without therapy, ARN appears to be a self-limited disease, isolated mid-peripheral nonconfluent retinal infiltrates and
2112 with resolution of the acute inflammatory changes occurring serologic evidence of herpes zoster varicella infection in whom
Acute Retinal Necrosis

CHAPTER 164
FIGURE 164.12. Zone of retinal pigment epithelial proliferation and
migration underlying thinned necrotic peripheral retina. Note the FIGURE 164.14. Retinal folds in stage C2 proliferative
presence of the nonpigmented epiretinal membrane on the surface of vitreoretinopathy associated with ARN. Note the pigmented epiretinal
the necrotic retina. H & E µ250. membranes in the lower left corner.

Acute Retinal Necrosis Associated with Cutaneous


Herpes Zoster
Typical ARN has also been associated with cutaneous herpes
zoster, reflecting reactivation of latent virus rather than primary
infection as in varicella. In contrast to necrotizing herpetic
retinopathy associated with varicella, patients with ARN and
cutaneous herpes zoster demonstrate typical features including
confluent peripheral necrosis, severe vitritis, optic nerve invol-
vement, and late retinal detachment.37–40 The time interval
between cutaneous eruption and ARN ranges from 5 days to
3 months and has been unilateral in eight of nine cases reported
in immunologically competent patients to date. Slightly less
than half of the cases reported to date have involved cranial
nerve V or VII, with the remainder of sites being remote from
the eye. However, in patients with AIDS, five cases have been
reported of ARN in patients with AIDS and history of prior
HZO. The time interval between cutaneous eruption and ARN
in these patients has ranged between 0 and 9 months, and four
out of five patients developed bilateral ARN.40 Patients with
AIDS and cutaneous zoster remote from the eye have also been
reported to develop typical ARN.59

Progressive Outer Retinal Necrosis


Although typical ARN has been reported in immunocompro-
mised patients, including those with AIDS,33–35 a subgroup of
FIGURE 164.13. Large tear in the inferior posterior retina of a patient
patients experience a more fulminant, rapidly progressive
with ARN.
necrotizing retinopathy, distinct from either CMV retinitis or
typical ARN and consistent with herpes zoster infection of the
retina. Early disease in PORN syndrome is characterized by
the ocular involvement did not progress to retinal detachment. multifocal deep retinal opacifications. These lesions quickly
In these patients, the role of early acyclovir treatment in the coalesce and progress to total full-thickness retinal necrosis over
prevention of more severe phases of the disease is unclear. More a short period, and early retinal detachment develops in the
indolent manifestations of retinal involvement have also been majority of patients. In patients with PORN, little or no inflam-
reported in other patients with varicella.20,21,56–58 Necrotizing matory reaction is seen in the vitreous and the anterior chamber,
herpetic retinopathy associated with varicella appears to be and occlusive vasculitis is generally not seen. Fluorescein angio-
limited predominantly to adults rather than children who graphy demonstrates full-thickness retinochoroiditis with
contract the disease despite more than 90% of all cases of associated choroidal leakage, pruning of the retinal vasculature
varicella occuring between the ages of 1 and 14 years.58 and zones of frank nonperfusion.60
Necrotizing herpetic retinopathy associated with varicella PORN can be easily distinguished from CMV retinitis. CMV
occurs within 3 months of the onset of primary infection retinopathy is characterized by slow progression, and the visual
(ranging from 5 to 40 days) and is believed to be limited loss associated with CMV corresponds directly with the area
primarily to adults, to involve fewer quadrants, to have less of retina involved with the infection or with direct optic nerve
vitritis, and infrequently to lead to retinal detachment.21 involvement. In contrast, the lesions in PORN are rapidly 2113
RETINA AND VITREOUS

progressive and are multifocal in presentation, and the retinal including the herpes zoster varicella virus group,1,24,69,76 herpes
dysfunction in PORN syndrome is widespread, leading to severe simplex, types 1 and 2,1,44,47,70 and CMV in several, rare, single
visual loss. The role of varicella-zoster virus group in patients case reports.75–78 The herpes zoster varicella virus group is believed
with PORN is strongly suggested by the frequent history of to account for the majority of cases and the typical syndrome.
cutaneous herpes zoster infection in affected patients. In one Herpesvirus hominis organisms, although exhibiting different
report, antecedent cutaneous zoster occurred in 67% of patients immunocytochemical staining characteristics, appear morpho-
for whom a history was available, and oral acyclovir was being logically similar by electron microscopy. The herpes zoster
taken by 32% for active cutaneous disease or for antiviral varicella viruses are relatively large, measuring ~150–200 nm
prophylaxis at the time PORN was diagnosed.61 DNA ampli- when fully enveloped by an outer lipid coat with a central core
fication using the polymerase chain reaction of a paraffin- of double-stranded DNA measuring ~100 nm. The central core
embedded transscleral eyewall biopsy specimen and an is covered by an icosahedral capsid composed of 162 tubular
enucleation specimen from a patient with PORN has successfully capsomers, which, in turn, are covered by the lipid bilayered
identified herpes zoster virus DNA, and histologic studies envelope (Figs 164.15 and 164.16). The DNA contains
showed intranuclear inclusion in choroidal cells consistent with ~125 000 bp and weighs 80 MDa. At least five families of
viral particles.62 Despite the apparently clear viral cause of this varicella-zoster virus glycoproteins have been identified by
syndrome, potent antiviral medications have failed to preserve immunohistochemical studies that represent the primary
visual acuity for most of these patients. Some case reports have markers for both humoral and cell-mediated immunity. The
described success in treatment of patients with PORN syndrome varicella-zoster group is highly cell associated and spreads from
SECTION 10

using several combinations of antiviral medications, including cell to cell by direct contact. These viruses are fastidious and are
intravenous ganciclovir and oral acyclovir,63 intravitreal
ganciclovir and oral sorivudine,64 lifelong high-dose intravenous
foscarnet alone,65 or intravenous foscarnet and ganciclovir.66
Overall, the visual prognosis is poor. Unlike other patients with
ARN and like patients with CMV retinitis, patients with PORN
often develop disease reactivation along edges of previous
retinal infection, even in those who show some initial response
to antiviral medications.67 Laser prophylaxis may be of some
benefit.68 The overall poor visual prognosis for patients with
PORN is attributable to a variety of factors, including frequent
total necrosis of the retina, retinal detachments, and optic
neuropathy. Intravitreal administration of antiviral medication
may improve prognosis.80

Key Features: Signs and Symptoms of the Cicatricial


Phase of ARN
• Symptoms: Reduced vision, persistent floaters, constricted
visual field
• Signs:
• Retinal atrophy
• Scarring of RPE and choroid
FIGURE 164.15. Transmission electron micrograph of viral particles in
• Vascular nonperfusion the necrotic retina of a middle-aged man with ARN. Note the
• Optic atrophy complete and incomplete viral particles, including the central
• Epiretinal membrane nucleocapsid and outer lipid envelope. µ87 000.
• Rhegmatogenous retinal detachment (common)

PATHOPHYSIOLOGY

VIRAL CAUSE
Although initial reports noted the similarities between the
clinical findings of ARN and those of both Behçet’s disease and
certain viral infections, the viral cause of this syndrome was not
definitively established until 1982.8 Several lines of evidence
implicate the Herpesvirus hominis class of viral infection as
the cause of this syndrome. These data include direct viral
culture,7,59,69,74 detection of viral antigens in intraocular fluid
specimens by direct or indirect immunofluorescence,34,65
elevated or serially increasing intraocular or serum antibody
titers to herpesviruses,70–72 immunocytochemical staining of
viral antigens in fixed tissue from biopsy or enucleation
specimens,8,23,72,73,74 Herpesvirus hominis particles in
gluteraldehyde-fixed retinal tissue,8,12,24,25,75 herpesvirus DNA
amplified by polymerase chain reaction,25 and clinical disease
coincident with or immediately following herpes infection in FIGURE 164.16. Another portion of the retina seen in Figure 164.15
other sites.21–23,39 Several members of the Herpesvirus hominis demonstrates a large number of tightly packed viral particles in
2114 family have been implicated in the pathogenesis of this disease, varying stages of assembly. µ30 000.
Acute Retinal Necrosis

difficult to culture, having been successfully transferred from intractable pain with chronic retinal detachment demonstrated
ocular specimens to human embryonal lung fibroblasts and extensive atrophy and degeneration of the outer retinal layers
human embryonal kidney cells in vitro.1,36,79 and pigment epithelium, occlusion of the retinal blood vessels,
HSV-1 and -2 are responsible for oral and genital ulcerations and massive choroidal lymphocytic infiltration and engorgement.
in humans. The virus is also transmitted by direct contact Membranes associated with PVR demonstrated a prominent
between skin surfaces and mucous membranes. The virus pigment epithelial component with intraretinal and subretinal
consists of four major components, a centrally located core cysts and subretinal ossification. No viral particles were
surrounded by three concentric structures; capsid, tegumen, identified.28,55
and envelope. The fully enveloped particle has an approximate Eyes in the acute phases of the disease show widespread pro-
diameter of 150–200 nm, with the envelope composed of lipid found changes in virtually all ocular structures. Granulomatous,
and protein components conveying the antigenicity of the virus keratic precipitates line the corneal endothelium, and chronic
and thereby dictating the host immunologic responses. and acute inflammatory cells infiltrate the iris and ciliary body,
Both the herpes zoster varicella virus group and the HSVs in which perivascular lymphocytic cuffing is evident in con-
code for two unique herpes-specific enzymes, an isofunctional junction with focal edema of the iris root and adjacent ciliary
deoxynucleoside kinase (herpes-specific thymidine kinase) and muscle. Chronic inflammatory cells are seen infiltrating the
a herpes-specific DNA polymerase. These enzymes are coded by sclera, accounting for the pain and scleral injection commonly
the herpesvirus genome and are not normally encountered in seen clinically. The histopathologic hallmarks of ARN are (1)
mammalian cells.80 Acyclovir (9(2-hydroxy-ethoxy)methylgua- retinal vasoocclusion, (2) sharply demarcated zones of necrotizing

CHAPTER 164
nine) is selectively phosphorylated by the herpes-coded thymidine retinitis, (3) choroidal lymphocytic infiltration and thickening,
kinase to its monophosphate form. Mammalian cellular enzymes and (4) optic neuropathy.
are then capable of converting acyclovir monophosphate to
acyclovir triphosphate. This represents the active antiviral form
of the drug, which becomes a selective substrate and inhibitor RETINAL VASOOCCLUSION
of herpesviral DNA polymerase. As a result, acyclovir inhibits Both large- and small-caliber arterioles demonstrate perivascular
viral replication in two ways: first, by prevention of the incorpo- cuffing, endothelial cell swelling, and fibrinoid or thrombotic
ration of deoxynucleotide triphosphates into herpesvirus DNA occlusion to varying degrees.1 In some instances, the lumina
and, second, by the incorporation of altered nucleotide analogs may be nearly or totally occluded through a combination of
(Fig. 164.17). Since uninfected mammalian cells do not contain endothelial swelling, red blood cell and platelet thrombi, and
the virus-specific enzyme that converts acyclovir to its fibrin (Figs 164.18 and 164.19). Despite the fact that the retinal
monophosphate form, this sequence of inhibitory events is not and choroidal vascular changes are a conspicuous feature of the
initiated in nonvirally infected cells, and the drug is therefore disease, to date no viral particles or antigen has been detected
relatively nontoxic contrasted with other antiviral agents such within retinal vessel walls, suggesting that the changes seen are
as adenine arabinoside.107 immunologically mediated.

HISTOPATHOLOGY CONFLUENT NECROTIZING RETINITIS


Histopathologic studies in the acute phases of ARN have been Another characteristic feature of ARN is the abrupt demar-
performed on a relatively small number of eyes, either after cation of necrotic and normal retina. Areas of relatively well
enucleation or on retinal biopsy specimens taken during preserved retina containing inflammatory infiltrates and
vitrectomy.1,24,35,76,77 Initial reports based on eyes undergoing hemorrhages in the inner and outer layers may be immediately
either vitrectomy for late retinal detachment or enucleation for

FIGURE 164.18. Histophathologic specimen of thrombotic vascular


FIGURE 164.17. Structural formula of acyclovir, a substituted purine occlusion in ARN. Note engorgement of the underlying choroidal
nucleoside that is active against various members of the Herpesvirus vessel as well as severe perivascular cuffing and occlusion of the
hominis family. lumen in the vessel by erythrocytes. H & E µ25. 2115
SECTION 10 RETINA AND VITREOUS

FIGURE 164.20. Cross-section of the optic nerve from a patient with


FIGURE 164.19. Cowdry’s A intranuclear eosinophilic nuclear inclusions
ARN. Note the central zone of necrosis. H & E µ100.
in the retina in a zone of severe necrosis. Note the marked swelling of
endothelial cells and the encroachment on lumen by fibrinoid change
seen adjacent to a zone of inclusion bodies. H & E µ400.
ELECTRON MICROSCOPIC AND
IMMUNOCYTOLOGIC FEATURES
adjacent to other zones of full-thickness necrosis with partial or Viral particles found within individual retinal cells demonstrate
complete obliteration of the normal retinal architecture. High- nucleocapsids of 80–100 nm, which can be seen undergoing
power views disclose the presence of eosinophilic intranuclear assembly within retinal cell nuclei to form poorly enveloped
inclusion bodies in infected retinal cells characteristic of herpes virus. These measure 150–200 nm in diameter and are adherent
infection. These changes, termed Cowdry A inclusions, contain to retinal cell membranes (Figs 164.15 and 164.16). The virus
complete and incomplete viral particles and marginally displace appears most abundant in the inner retinal layers and in the
the host nuclear chromatin, which is basophilic in H & E- transition zone between necrotic and preserved retina. In areas
stained sections. The pigment epithelial proliferation and of greatest cytopathic effect, there is marked disruption of
migration from the normal monolayer through necrotic retina retinal tissue, with bare nuclei and fragmented cell membranes
and onto the surface are frequently associated with reactive present. Viral antigens have been identified by immunocyto-
gliosis and epiretinal membrane formation. These represent the chemical techniques employing a variety of murine monoclonal
sites of future retinal tears in response to contracting preretinal antibodies and avidin–biotin–peroxidase methods specific for
and transvitreal membranes. Pigmented macrophages are the major GP98/GP62 varicella-zoster glycoprotein complex.
commonly encountered, and cytomegaly is not a conspicuous Viral antigens have been detected in transitional zones between
feature. normal and necrotic retina, retinal pigment epithelial cells, and
inner and outer retinal cells, but not in retinal vascular endo-
thelium, ciliary body, or optic nerve. However, immunoglobulins
G and M, fibrinogen, and complement component C3c have
CHOROIDAL THICKENING AND been detected in retinal arteriolar walls and in surrounding
LYMPHOCYTIC INFILTRATION retinal tissue adjacent to zones of zoster antigens.76
Another feature that differentiates ARN from CMV retinitis
histologically is the relatively severe thickening and lympho- EXPERIMENTAL ACUTE RETINAL
cytic infiltration of the choroid seen in the former condition. NECROSIS IN AN ANIMAL MODEL
The infiltrate is mononuclear, consisting predominantly of
lymphocytes and plasma cells with associated occlusion of the Although it is well established that the primary pathogenetic
choriocapillaris and, to a lesser extent, the larger choroidal event of ARN is Herpesvirus hominis infection of the retina, a
vessels. Areas of granulomatous inflammation may be seen as number of issues remain unresolved, including the route of
well. In a blind eye with ARN, varicella-zoster virus DNA was transmission to the eye, the mechanism of bilateral disease in
detected in the choroid and choriocapillaris by in situ hybridiza- some patients, and the underlying mechanisms responsible for
tion and by immunohistochemical stains in mononuclear cells the variability and severity of the disease. The development of a
with eosinophilic inclusions.53 murine model of herpes simplex retinitis has provided some
insights into these questions.17,18,81,82
It has been shown that intracameral injection of HSV-1 (KOS
strain) in BALB/c mice produces the picture of acute hemor-
OPTIC NEUROPATHY rhagic necrotizing retinitis. Interestingly, the retinitis does not
Involvement of the optic nerve is variable, ranging from patchy occur in the injected eye but rather in the contralateral (non-
areas of chronic inflammation along the pia to broad zones of injected) eye 9–10 days after injection and is completed over the
neuronal necrosis, plasma cell infiltration, and vascular course of 12–48 h.82,83 ARN develops only in immunocompetent
occlusion (Fig. 164.20). To date, viral particles have not been animals and only when viral titers in the contralateral eye
identified in the optic nerves available for study, although there exceed a threshold level (4 log10 plaque-forming units). This
is experimental evidence to suggest that transmission from confirms that viral infection is essential for the development of
one eye to another in bilateral cases may occur via the optic this syndrome. After injection into the anterior chamber of one
2116 nerve.17,18 eye, HSV reaches the contralateral eye in two waves. The first,
Acute Retinal Necrosis

which fails to replicate, arrives within 24 h. The second arrives


TABLE 164.1. Differential Diagnosis of Acute Retinal Necrosis
within 5–7 days and reaches the eye after retrograde spread
down the optic nerve from the brain. Within 24–48 h of the Syphilitic neuroretinitis
arrival of the second wave of replicating virus, the characteristic
Cytomegalovirus retinitis
picture of hemorrhagic and necrotizing retinitis develops.83
In this murine model, in addition to viral infection, Epstein–Barr virus retinitis
immunologic factors play an important role in the development Toxoplasmic retinochoroiditis
of ARN. It is known that, paradoxically, immunocompetence is
a prerequisite for the development of ARN in several different Candida albicans endophthalmitis
regards. First, immunoincompetent mice (athymic) do not Acute multifocal hemorrhagic retinal vasculitis
develop the characteristic fundus picture of ARN, although viral
Behçet’s disease
replication takes place in the contralateral eye at a comparable
time and in similar quantity to immunocompetent animals. Sarcoidosis
Second, ARN does not develop unless there is suppression of Progressive outer retinal necrosis
virus-specific delayed hypersensitivity. This phenomenon is
termed anterior chamber-associated immune deviation and is Primary intraocular lymphoma
genetically determined in certain strains of mice in which
humoral antiviral antibody production remains unaffected.

CHAPTER 164
These findings are consistent with a report on a patient with
ARN caused by HSV who had documented evidence of bilateral progression from multifocal areas of deep retinal opacification
high-intensity optic tract lesions involving the lateral geniculate to complete retinal necrosis, and the commonly very poor
ganglia, temporal lobes, and midbrain seen on magnetic response to antiviral therapies seen in patients with PORN.
resonance imaging (MRI). This, along with concurrent gene- Sarcoidosis, Candida albicans endophthalmitis, and Behçet’s
ralized malaise and fever, suggests a neural route of spread.1 disease can generally be distinguished from ARN by the clinical
The requirement for immunocompetence appears to be based history. It has been recognized that some patients with
on the participation of precursor cytotoxic T cells in the intraocular lymphoblastic lymphoma may present with retinal
development of retinal necrosis. T cells harvested from lymph hemorrhages, pseudovasculitis, and infiltrates mimicking
nodes ipsilateral to the injected eye and from the contralateral ARN.85–87 Bilateral retinal necrosis has also been described
eye proliferate in vitro when exposed to viral antigens and are as a complication of X-linked lymphoproliferative disease, and
believed to mediate the destruction of retinal cells infected by Epstein–Barr virus genomic DNA within the eye was shown by
HSV.18 It has also been shown that HSV-specific effector T cells, the polymerase chain reaction.88
when promptly injected into the contralateral eye of animals
undergoing intracameral injection of HSV, will prevent the
development of ARN, although injection of effector cells delayed LABORATORY EVALUATION
by more than 1 day after virus injection will not. Moreover, Baseline laboratory examination of patients with suspected
HSV-1-specific immune effector cells restimulated with virus in ARN can help establish the diagnosis and facilitate treatment.
vitro are more effective in producing cytotoxicity to HSV-1- Suggested studies are outlined Key Features: Diagnosis. Patients
infected target cells than are HSV-1-specific immune effector should have a complete blood count, sedimentation rate
cells that are not restimulated.83 determination, and basic serologic studies, including a Venereal
Disease Research Laboratory (VDRL) test, fluorescent tre-
DIFFERENTIAL DIAGNOSIS ponemal antibody absorption test, and rheumatoid factor and
antinuclear antibody determinations to exclude systemic
vasculitis or coagulopathy. Consideration should be given to
OPHTHALMOSCOPY serologic testing for HIV infection. Abnormal platelet hyper-
Because of its characteristic fundus appearance, the diagnosis aggregation in response to adenosine diphosphate admini-
of ARN can be established in the healthy immunocompetent stration has been detected in patients with ARN.89 Quantitative
patient on ophthalmoscopic grounds, which include (1) the antibody level testing for herpesvirus in acute and chronic sera
presence of granulomatous anterior uveitis, (2) vitreous cellular from patients with ARN is relatively unhelpful in determining
reaction, (3) retinal arteritis, (4) multiple patchy or confluent a specific causative diagnosis in this syndrome. In one study,
areas of necrotizing retinitis with or without associated only 39% of cases had a diagnostic increase or decrease in
hemorrhage predominantly located in the retina periphery, and herpes group viral antibody levels on serial sampling.90 Serum
(5) optic disk swelling of variable degree. creatinine, blood urea nitrogen determinations, as well as
The conditions to be considered in the differential diagnosis urinalysis, should be performed to determine the patient’s
of ARN are listed in Table 164.1. The clinical syndromes most suitability for acyclovir administration, and a chest radiograph
commonly misdiagnosed as ARN in healthy patients are syphilitic study and a purified protein derivative test should be performed
neuroretinitis and acute multifocal hemorrhagic retinal to exclude tuberculous disease, which could contraindicate oral
vasculitis.51,84 The former can be distinguished on the basis of corticosteroid therapy.
appropriate serologic testing, and the latter by the presence of a Optional studies should be performed in specialized
greater degree of retinal hemorrhages, initial venous involve- circumstances, including a computed tomography (CT) scan or
ment, and a subsequent clinical course that includes a failure to MRI study in patients with clinical evidence of severe optic
respond to acyclovir, a failure to develop retinal detachment, nerve dysfunction or central nervous system symptoms such
and the likelihood of ocular neovascularization. In some as headache, meningismus, or altered mental status. MRI evi-
instances, the differentiation between ARN and CMV retinitis dence of optic nerve tract, ganglionic, or cerebral disease may
or toxoplasmic retinochoroiditis may be problematic in have an impact on the dosage and duration of acyclovir therapy.
immunocompromised patients. PORN can be readily distin- Consideration should be given to lumbar puncture in patients
guished from ARN syndrome because of the absence of vitritis with either neurologic symptoms or an abnormal neuroradiologic
and anterior chamber inflammation, the extremely rapid imaging study. Appropriate cerebrospinal fluid cultures, antibody 2117
RETINA AND VITREOUS

testing, and cytologic studies should be performed only in rare of HSV (ED50 of 0.1–1.6 mM) and, to a lesser extent, the herpes
instances to exclude other causes of central nervous system zoster varicella group (ED50 of 3–4 mM) to acyclovir, the drug
disease and retinal infiltration such as syphilis or lymphoblastic theoretically is effective in ARN associated with either agent.104
lymphoma. Because ARN is generally not believed to be caused by strains of
Anterior chamber or vitreous specimens have been obtained CMV, which is typically more resistant to acyclovir (ED50 of
in patients with ARN that helped to determine the causative 200 mM), more toxic agents, including vidarabine104 and
viral agent. Assays are used to detect viral antibody levels in the ganciclovir104 or foscarnet,105 are not generally required, unless
intraocular fluids with appropriate recalculations for serum there is evidence of drug resistance or associated HIV infection.
antibody levels by the Goldmann–Wittmer coefficient.23,70,71,91–95 After intravenous administration of 5 mg/kg of acyclovir in
In one study, the rate of identifying viral cause in ARN via humans, peak plasma levels of 30–40 mM are reached with a
intraocular antibody production determinations was 89%.96 half-life of 2–4 h. These levels contrast with a peak concen-
However, the detection of multiple positive quotients for anti- tration of 8.7 mM in the serum and 3.3 mM in the aqueous
bodies within a single eye can make interpretation problematic.96 humor after five oral 400–mg doses in volunteers undergoing
The detection of herpesviral DNA in specimens from eyes with cataract extraction.106,107 Because there is a significant corre-
ARN and eyes with PORN has also been reported utilizing the lation between plasma and aqueous concentrations of the drug,
polymerase chain reaction to amplify minute quantities of viral it can be inferred that intraocular concentrations of acyclovir
DNA from specimens as small as 50 mL.63,97–101 When available are considerably higher after intravenous administration than
and when intraocular surgery is otherwise warranted, these after oral administration. At present, initial therapy with intra-
SECTION 10

tests on intraocular fluids are certainly indicated. Rarely, in venous acyclovir for a minimum of 1 week is recommended.
complex cases – such as in patients with underlying Because active viral particles have been identified in eyes with
immunocompromise or atypical findings, or in those refractory ARN subsequent to intravenous acyclovir therapy,11 and the
to antiviral therapy – diagnostic vitrectomy or retinal biopsy, or greatest period of risk for involvement of the fellow eye is within
both, may be indicated. In addition to conclusively establishing the first 6 weeks after presentation of the first eye, oral acyclovir
the diagnosis, this may permit testing for antiviral should be administered at a dosage of 2–4 g daily for 4–12 weeks
sensitivities.1,24,48,70,73,102 Diagnostic vitrectomy has been combined after completion of intravenous acyclovir as a precautionary
with infusion of intravitreal acyclovir in a concentration of measure. The role of oral acyclovir in higher dose ranges (4 g
10–40 mg/mL.12 daily) as an alternative treatment to intravenous acyclovir
remains unsettled and awaits further study.
Key Features: Diagnosis Intravenous acyclovir is generally administered at a total
• Ophthalmoscopy dosage of 1500 mg m–2 day–1 based on calculation of the body
• Serology: limited benefit due to high exposure in general index in square meters from the height and weight; 500 mg m–2
population day–1 is administered every 8 h. Oral acyclovir is available in
• MRI with contrast: perform if encephalitis suspected either 200-mg or 800-mg strength, which is administered five
• Lumbar puncture: perform if meningitis suspected; include viral times daily in dosages ranging from 1000 mg to 4 g daily,
diagnostic studies depending on the severity of the disease process and the age and
• Intraocular diagnostics: perform to discriminate between weight of the patient. Studies of antiviral sensitivities of the
herpes simplex and varicella zoster; include CMV, herpes zoster varicella virus group isolated from the vitreous
toxoplasmosis, Epstein–Barr virus, if atypical of a patient with ARN indicated in vitro susceptibility of the
• PCR: best in first 2 weeks of disease isolate to both acyclovir (ED50 of 5.3 mM) and dehydroxy-
• Antibodies: permit diagnosis in some cases after 2 weeks phenylglycol (ED50 of 4.7 mM).
• HIV test Acyclovir has a large therapeutic index, and complications
• Syphilis serology reported to date have been relatively minor. They include local
irritation at the intravenous entry site and, rarely, reversible
elevation of serum creatinine concentration, presumably
CURRENT THERAPY through crystallization. Central nervous system toxic reactions,
including delirium, tremors, abnormal electroencephalograms,
Current management of ARN consists of five primary com- as well as elevation of liver transaminase levels, have been re-
ponents in the acute phase of the disease, in addition to ported, although also rarely and in complex cases in which a
treatment of retinal detachment if it occurs in the cicatricial causal relationship has not been clearly established. To date,
phase of the disease. These five components are (1) antiviral acyclovir has not been found to be carcinogenic, mutagenic, or
therapy, (2) antiinflammatory therapy, (3) antithrombotic therapy, teratogenic.107
(4) retinal detachment prophylaxis, and (5) possible optic nerve In one clinical series, treatment of patients with ARN with
therapy. 1500 mg m–2 day–1 of acyclovir in conjunction with aspirin and
corticosteroids resulted in regression of active retinal lesions
of presumed viral origin beginning, on average, 3.9 days after
ANTIVIRAL THERAPY initiation of therapy. Lesions completely regressed on an
The demonstration of herpesvirus particles in eyes with ARN average of 32.5 days after initiation of therapy, and no eye
facilitated the development of specific treatment regimens. Both developed new retinal lesions or progressive optic nerve
intravitreal12,80 and intravenous acyclovir12,16 have been involvement 48 h or more after initiation of therapy. Acyclovir
reported to be of benefit in the treatment of this disorder. treatment does not appear to ameliorate vitritis or retard the
rate of retinal detachment in advanced cases, although it may
Acyclovir do so in cases treated at an earlier time, before the development
Acyclovir remains the drug of choice for the initial treatment of of severe confluent lesions.11,46 Retrospective analysis of
ARN and has selective advantages over other purine and patients treated both before the era of acyclovir therapy and
pyrimidine nucleoside antimetabolites with antiviral activity in subsequent to the induction of acyclovir suggests a benefit of
vitro, including phosphonoformate, bromovinyldeoxyuridine, treatment with regard to prevention of new lesions in the fellow
2118 and fluoroiodoaracytosine.69 Because of the relative sensitivity eye. Of 31 patients treated with acyclovir, 87.1% were disease-
Acute Retinal Necrosis

free in the fellow eye at the time of last examination, in contrast In patients who are unable to tolerate acyclovir or who
to only 30.4% of fellow eyes in patients not treated with demonstrate clinical disease progression suggestive of acyclovir
acyclovir. Two years after treatment using multivariate drug resistance, consideration may be given to administration
methods, the cumulative proportion of patients who remained of intravenous ganciclovir as an alternative form of therapy.
disease-free in the fellow eye was 75.3% for the group treated Recommended guidelines for treatment with ganciclovir at
with acyclovir and 35.1% for the group not treated with present are 5 mg/kg every 12 h for 7 to 14 days by intravenous
acyclovir.15 Since the patients involved in this study were infusion. Patients undergoing ganciclovir administration should
evaluated retrospectively and treated at different times and at have a twice-weekly complete blood count with differential and
multiple different institutions, these data should be interpreted platelet count to identify early signs of hematopoietic sup-
with appropriate caution (Figs 164.21 and 164.22). pression. The dosage of ganciclovir should be reduced in
patients with evidence of impaired creatinine clearance, and the
drug should be discontinued on signs of severe leukopenia
(<1000 cells/mL) or thrombocytopenia.78,104,108
In addition to oral and intravenous routes, acyclovir may also
be administered as an intravitreal infusate during the course of
vitrectomy. Concentrations of 10–40 mg/mL have been reported to
be safe.12,108 This form of treatment may be effective as an adjunct
to oral and intravenous therapy in patients requiring vitrectomy

CHAPTER 164
during the acute phase of viral replication within the eye. Direct
intravitreal injection of ganciclovir 2 mg or foscarnet 2.4 mg
during the acute phases of the disease appears to improve outcome
compared to treatment with systemic medication only.80

Other Antiviral Agents


Famciclovir
Recently, congeners of acyclovir have been developed that have
more favorable pharmacokinetic parameters after oral adminis-
tration. Famciclovir is a synthetic acyclic guanine derivative
prodrug that is metabolized to penciclovir after oral adminis-
tration. Penciclovir is active against HSV and herpes zoster
FIGURE 164.21. Life table analysis of lesions treated with and virus and has been used successfully in immunocompetent
without acyclovir and the resultant cumulative risk of disease patients with shingles and recurrent genital infection (Fig.
development in the fellow eye. The broken line represents untreated
164.23).109
patients with unilateral disease over time. The group of patients
treated with acyclovir is less likely than the group not treated to
develop ARN in the fellow eye (P = 0.0013). Valacyclovir
Reprinted with permission from Elsevier Science Palay DA, Sternberg P, Davis J, Valacyclovir is the L-valyl ester of acyclovir, which is given as an
et al: Decrease in the risk of bilateral ARN by acyclovir therapy. Am J Ophthalmol oral prodrug that undergoes rapid first-pass metabolism to yield
1991; 112:250–255. acyclovir and L-valine. Its principal advantage over oral
acyclovir is its greater bioavailability (Fig. 164.24). Three daily
1000-mg doses of valacyclovir are believed to be as effective
as five daily 800-mg doses of acyclovir in the treatment of
cutaneous herpes zoster infection.110 There remain few
published data on the effectiveness of either famciclovir or
valacyclovir in the treatment of ARN, but anecdotal data seem
promising.

ANTIINFLAMMATORY THERAPY
Inflammatory cells, including cytotoxic lymphocytes, are
believed to play a major role in both the primary phases of the
syndrome, when they may contribute to both retinal necrosis
and vasculitis,17,18,111–114 and secondarily, when vitreous cellular
infiltrates contribute to organization of the vitreous that
predisposes to late retinal detachment.1–9 As a result, anti-
inflammatory therapy is an important component of treatment
for this disease. Administration of oral prednisone or equivalent
corticosteroids in doses of 0.5–1.5 mg kg–1 day–1 is recommended
in the acute phases of the syndrome during the period of
greatest intraocular inflammation. Because steroids are known
to enhance viral replication under selected conditions, the
initiation of steroid therapy should be delayed 24–48 h after
administration of intravenous acyclovir. However, in cases in
which the severity of intraocular inflammation is great,
FIGURE 164.22. The same region as shown in Figure 164.2 after particularly with optic nerve involvement, consideration may
10 days of intravenous administration of acyclovir. Note the nearly be given to earlier institution and increase of the dosage to
complete resolution of nummular lesions nasal to the optic nerve. 2 mg kg–1 day–1 of steroid therapy. The exact duration of steroid 2119
RETINA AND VITREOUS

N O
N
O N
NH
NH2

O N O N
H3C N NH2 O
N NH2

O
Valacyclovir
O CH3 Hydrolysis by
valacyclovir hydrolase

O O

N
Famciclovir NH
SECTION 10

NH2

OH HO N N NH2
O
O
O

N L-valine Aciclovir
NH
FIGURE 164.24. Structural formula of valacyclovir, a prodrug
converted to acyclovir and L-valine by hydrolase.

HO N N NH2

markedly swollen, nearly obliterating the vascular lumina in


HO some patients.1,76 These events are believed to be secondary to
humoral and cell-mediated mechanisms and contribute to
stasis and vascular thrombosis in patients with ARN. This may
Penciclovir result in vision loss from both optic nerve dysfunction and
retinal infarction. In addition to treatment with antiinflam-
FIGURE 164.23. Structural formula of famciclovir and its active matory agents, both antiplatelet agents and anticoagulation
metabolite penciclovir.
have been previously recommended.11,115 Platelet hyper-
aggregation has been detected in patients with ARN and may be
treated with aspirin, 500–650 mg daily. The benefits of more
aggressive forms of anticoagulation, including both heparin and
warfarin, do not at present appear to warrant the use of such
forms.
therapy is dictated by the severity of the intraocular inflam-
mation and any associated complications of therapy. In patients RETINAL DETACHMENT PROPHYLAXIS
in whom there is no contraindication to steroid therapy,
treatment with prednisone should continue for 6–8 weeks, with Retinal detachment continues to be the most serious late
gradual tapering of the dose in order to reduce both the anterior complication of ARN, even with the prompt administration of
and the posterior segment intraocular inflammation. At present, acyclovir and steroid therapy. Peripheral argon laser photo-
no other form of antiinflammatory therapy, including coagulation to demarcate zones of retinitis during the acute
cyclosporine or various antimetabolites, has been shown to be phase of the disease has been recommended as a potentially
of benefit in this syndrome. Topical treatment with pred- effective prophylactic measure if the media will permit such
nisolone in conjunction with cycloplegic agents should also be treatment.16,68 In one retrospective clinical series,16 12 eyes in
given with dosage dictated by the severity of anterior segment 10 patients underwent prophylactic laser photocoagulation and
inflammation. No benefit has been demonstrated for the use of two (17%) retinal detachments developed. During the same
topical acyclovir in this syndrome. time, four of six (66%) patients not receiving photocoagulation
experienced retinal detachment as a complication of ARN.
Because it is likely that eyes with less severe forms of ARN have
ANTITHROMBOTIC THERAPY the clearest media and are therefore more likely to be able to
Periarteritis and intraluminal narrowing are conspicuous undergo prophylactic photocoagulation when compared with
features of ARN on both clinical and histopathologic studies. more severely affected eyes, it cannot be definitively concluded
Viral particles have not been demonstrated in the retinal that the difference in these two rates is solely the result of
2120 vascular endothelial cells, although the cells themselves may be photocoagulation. Nonetheless, it seems likely that photoco-
Acute Retinal Necrosis

agulation does have some prophylactic value against the devel-


opment of retinal detachment and it should be administered MANAGEMENT OF RETINAL DETACHMENT
in all patients at the earliest possible stage. Recommended Retinal detachment continues to be a difficult problem in
treatment guidelines at present include the placement of three patients with late-stage ARN. In initial series of patients treated
rows of argon, krypton, or dye laser at the junctional zones appropriately with acyclovir, corticosteroids, and aspirin, 11 of
demarcating peripheral confluent necrosis from normal retina. 13 eyes experienced retinal detachment (84.6%).11 This is com-
When possible, photocoagulation should be extended into zones parable to a 75% rate of retinal detachment seen before the
of necrosis, in which the retina tears usually occur. The benefit introduction of acyclovir therapy.7
of photocoagulation in isolated thumbprint lesions in the Retinal detachment complicating ARN is particularly severe.
posterior segment is less certain. In some instances, longer The majority of retinal detachments occur within 3 months
wavelengths and retrobulbar anesthesia may be helpful in of the onset of symptoms. In one review, 41 of 55 eyes (75%)
permitting successful applications in eyes with media opacities. affected by the virus experienced retinal detachment. The time
Despite photocoagulation, it can be estimated that up to 50% of interval ranged from 1 to 10 months, with only four of 41 eyes
eyes with ARN will develop retinal detachment. experiencing a retinal detachment more than 3 months after
One alternative strategy for retinal detachment prophylaxis the onset of the syndrome.9 In a series of 12 patients (13 eyes)
has been the use of vitrectomy combined with endophoto- treated with intravenous acyclovir, 83.6% of eyes experienced
coagulation and scleral buckling.12,14,114 One difficulty encoun- retinal detachment an average of 59 days after initiation of
tered in the performance of a vitrectomy in such eyes is the therapy, with a range of 30–145 days.11 Eyes with retinal

CHAPTER 164
tight adherence between the nondetached vitreous gel and the detachment complicating ARN have a more unfavorable visual
atrophic, necrotic peripheral retina. Additionally, eyes with and anatomic prognosis for several reasons. The retinal breaks
ARN-associated severe intraocular inflammation may develop contributing to the rhegmatogenous components of these
severe postoperative ocular hypertension, fibrin response, and detachments are often multiple, large, and posteriorly located,
choroidal detachment after vitrectomy and scleral buckling.115 either in necrotic retina or at the junction between normal and
The prophylactic benefit of combined vitrectomy and scleral necrotic retina (Figs 164.25 and 164.13). PVR is frequent, and
buckling with endolaser remains uncertain.12,14,117,118 In one eyes that demonstrate severe preexisting intraocular inflam-
series, three of four eyes undergoing such prophylactic surgery mation and vascular compromise are predisposed to fibrin
experienced retinal detachment despite this therapy and appeared response, choroidal detachment, and ocular hypertension (Fig.
to have a more unfavorable course than eyes not undergoing 164.14).5,15,118
prophylactic vitrectomy and photocoagulation.118 PVR is a frequent occurrence in eyes with ARN that expe-
rience retinal detachment. In one series, 72.7% of eyes with
retinal detachment demonstrated stage C1 or greater PVR.12
TREATMENT OF ACUTE OPTIC NEUROPATHY The high frequency with which PVR occurs in this group of
In addition to the zones of frank neural necrosis seen histopatho- patients can be understood in view of the pathophysiology of
logically,19,54 it has been suggested that in some instances optic
nerve sheath distention and compression of the optic nerve may
contribute to the visual dysfunction associated with ARN. In
one series, eight eyes in six patients with optic neuropathy
believed to be secondary to ARN underwent optic nerve sheath
decompression in addition to intravenous acyclovir therapy.
These patients had a more favorable long-term visual prognosis
than did patients with lesser degrees of neuropathy not under-
going decompression.54 Clinical experience with this form of
therapy remains limited, but it appears to be a potentially useful
way to reduce visual disability in this condition in conjunction
with appropriate antiviral, antiinflammatory, and antithrombotic
therapy. This diagnosis may be established on the basis of CT
or MRI scanning in patients with clinical or visual evidence of
optic neuropathy in association with ARN.

Key Features: Treatment


• Antiviral therapy
• Intravenous acyclovir*
• Oral valacyclovir†
• Intravitreal ganciclovir and/or foscarnet*‡
• Combination antiviral therapy with intravenous foscarnet
plus intravenous acyclovir or intravenous ganciclovir*‡
• Or, appropriate treatment for atypical cases:
cytomegalovirus, toxoplasmosis
• Antiinflammatory treatment
• Oral or extraocular corticosteroids* FIGURE 164.25. Artist’s representation of retinal detachment in a
• Retinal detachment prophylaxis patient with ARN, including a large superior retinal tear at the junction
• Laser demarcation* of involved and noninvolved retina as well as multiple smaller pinpoint
• Early vitrectomy, membrane peeling and endolaser† defects in areas of postnecrotic zones. Note the presence of fixed
*Case series support use. folds radiating through the posterior pole from the periphery

Case reports support use. superotemporally. Reprinted from Blumenkranz MS, Clarkson J, Culbertson
‡ W, et al: Vitrectomy for retinal detachment associated with ARN. Am J
Recommended in patients who are immunocompromised.
Ophthalmol 1088; 106:426–429. 2121
RETINA AND VITREOUS

the disease, including marked thinning of the peripheral retina


and migration of pigment epithelial cells through necrotic
retina onto the retina surface associated with metaplasia and
epiretinal membrane formation. These changes occur in
conjunction with cellular infiltration and secondary fibrotic
changes in the vitreous, including adherence to the peripheral
retina. It is known that the breakdown of the blood–ocular
barrier encountered in patients with uveitis contributes to the
development of PVR, through liberation of serum-derived growth
factors within the eye.119 It is also known that fibrin, which is
known to be more common in eyes undergoing retinal reattach-
ment surgery for ARN,118 is capable of producing mesenchymal
transformation of pigment epithelial cells, which may further
contribute to the development of PVR.120 As a result of these
factors, retinal detachment repair in patients with ARN has
been more difficult than other forms of retinal reattachment,
although success rates appear to be improving with improved
vitreoretinal surgical techniques.
SECTION 10

Before 1982, retinal reattachment was attempted in only 18


of 41 eyes, with only four of 18 procedures being successful
(22%).7 Employing a combination of scleral buckling and
vitrectomy in selected instances, Clarkson and colleagues118
reported an improved success rate, with 13 of 16 eyes that
experienced retinal detachment from ARN requiring operation
and seven retinas remaining reattached (53.8%). Similar results
have been reported by other authors employing a combination FIGURE 164.26. Artist’s representation of peripheral demarcating
of vitreoretinal surgery and scleral buckling.121 photocoagulation, after vitrectomy and fluid–gas exchange in a patient
with retinal holes and detachment secondary to ARN, seen in
Other reports suggest that a combination of vitrectomy,
Figure 164.25.
lensectomy, internal fluid–gas exchange, and endolaser treatment Reprinted from Blumenkranz MS, Clarkson J, Culbertson W, et al: Vitrectomy for
without scleral buckling may provide a more favorable anatomic retinal detachment associated with ARN. Am J Ophthalmol 1988; 106:426–429.
and visual outcome in eyes with retinal detachment com-
plicating ARN (Fig. 164.26).14 In one series, eyes undergoing this
procedure were compared with eyes undergoing conventional
scleral buckling in association with vitrectomy. Overall, a com-
bined final success rate of 93.8% (15 of 16 eyes) was reported.
Complete retinal reattachment with one operation was
achieved for all eyes not previously subjected to a prophylactic
procedure.14,118 In this series of patients, perioperative com-
plications, including ocular hypertension (75%), fibrin response
(87.5%), and choroidal detachment (62.5%), were common in
patients undergoing scleral buckling (Fig. 164.27). Without
scleral buckling and cryotherapy, only one of eight eyes (12.5%)
experienced fibrin response and none had choroidal detachment
or ocular hypertension (intraocular pressure greater than 35
mm Hg). The rate of reoperation and the final visual-acuity rate
also appeared to be more favorable in those patients undergoing
vitrectomy without scleral buckling, with 87.5% of patients
achieving a visual acuity of 20/200 or better and 62.5%
achieving a visual acuity of 20/70 or better. These visual-acuity
results contrasted with only 25% of patients undergoing scleral FIGURE 164.27. Slit-lamp photograph of a pupillary fibrin response in
buckling combined with vitrectomy achieving a visual acuity of a patient who underwent lensectomy, vitrectomy, scleral buckling, and
20/200 or better. This may be related to the higher rate of need endolaser treatment for an ARN-related retinal detachment.
for retinal reoperation and use of silicone oil in this group.118
Silicone oil, or other tamponades, and retinotomy may be
required in some eyes with severe proliferative retinopathy.
However, the use of silicone oil in primary retinal detachment
associated with ARN does not seem necessary in the majority
of cases.

REFERENCES
1. Lewis ML, Culbertson WW, Post JD, et al: Jpn J Clin Ophthalmol 1971; 5. Price FW, Schlaegel TF: Bilateral ARN. Am
Herpes simplex virus type I. A cause of the 25:607–619. J Ophthalmol 1980; 89:419–424.
ARN syndrome. Ophthalmology 1989; 3. Wilkerson D, Aaberg TM, Reeser FH: 6. Sternberg P, Knox DL, Finkelstein D, et al:
96:875–878. Necrotizing vaso-occlusive retinitis. Am J Acute retinal necrosis syndrome. Retina
2. Uragama A, Yamada N, Sasaki Y, Ophthalmol 1977; 84:209–219. 1982; 2:145–151.
et al: Unilateral acute uveitis with 4. Young NJA, Bird AC: Bilateral ARN. Br J 7. Fisher JP, Lewis ML, Blumenkranz MS,
2122 retinal periarteritis and detachment. Ophthalmol 1978; 62:581–590. et al: The acute retinal necrosis syndrome.
Acute Retinal Necrosis

Part 1. Clinical manifestations. 28. Ezra E, Pearson RV, Etchells DE, Gregor 48. deBoer JH, van Haren MA, de Vries
Ophthalmology 1982; 89:1309–1316. ZJ: Delayed fellow eye involvement in Knoppert WA, et al: Analysis of IL-6 levels
8. Culbertson WW, Blumenkranz MS, Haines H, acute retinal necrosis syndrome. Am J in human vitreous fluid obtained from
et al: The acute retinal necrosis syndrome. Ophthalmol 1995; 120:115–117. uveitis patients, patients with proliferative
Part 2. Histopathology and etiology. 29. Falcone PM, Brockhurst RJ: Delayed onset intraocular disorders and eye bank eyes.
Ophthalmology 1982; 89:1317–1325. of bilateral acute retinal necrosis syndrome: Curr Eye Res 1992; 11:181–186.
9. Gorman BD, Nadel AJ, Coles RS: Acute a 34-year interval. Ann Ophthalmol 1993; 49. Fox GM, Blumenkranz MS: Giant retinal
retinal necrosis. Ophthalmology 1982; 25:373–374. pigment epithelial tears in acute retinal
89:809–814. 30. Egbert P, Pollard RB, Galligher JG, Merigan necrosis. Am J Ophthalmol 1993;
10. Holland GN, the Executive Committee of T: Cytomegalovirus retinitis in 116:302–306.
the American Uveitis Society: Standard immunosuppressed hosts. Natural history 50. Brown RM, Mendis U: Retinal arteritis
diagnostic criteria for the acute retinal and effects of treatment with adenine complicating herpes zoster ophthalmicus.
necrosis syndrome. Am J Ophthalmol arabinoside. Ann Intern Med 1980; Br J Ophthalmol 1973; 57:344–346.
1994; 117:663–667. 93:655–664. 51. Blumenkranz MS, Kaplan HJ, Clarkson JG,
11. Blumenkranz MS, Culbertson WW, 31. Bloom JN, Katz JI, Kaufman HE: Herpes et al: Acute multifocal hemorrhagic retinal
Clarkson JG, et al: Treatment of the ARN simplex retinitis and encephalitis in an adult. vasculitis. Ophthalmology 1988;
syndrome with intravenous acyclovir. Arch Ophthalmol 1977; 95:1798–1799. 95:1663–1672.
Ophthalmology 1986; 93:296–300. 32. Friberg TR, Jost BF: Acute retinal necrosis 52. Han DP, Abrams GW, Williams GA:
12. Peyman GA, Morton FG, Uninsky E, et al: in an immunosuppressed patient. Am J Regression of disc neovascularization by
Vitrectomy and intravitreal antiviral drug Ophthalmol 1984; 98:515–517. photocoagulation in the ARN syndrome.

CHAPTER 164
therapy in ARN syndrome. Arch 33. Jabs DA, Schachat AP, Liss R, et al: Retina 1988; 8:244–246.
Ophthalmol 1984; 102:1618–1621. Presumed varicella-zoster retinitis in 53. Rummelt V, Wenkel H, Rummelt C, et al:
13. Culbertson WW, Clarkson JG, Blumenkranz immunocompromised patients. Retina Detection of varicella zoster virus DNA and
MS, Lewis ML: Acute retinal necrosis. Am J 1987; 7:9–13. viral antigen in the late stage of bilateral
Ophthalmol 1983; 96:683–685. 34. Chambers RB, Derick RJ, Davidorf FH, acute retinal necrosis syndrome. Arch
14. Blumenkranz MS, Clarkson JG, Culbertson et al: Varicella-zoster retinitis in human Ophthalmol 1992; 110:1132–1136.
WW, et al: Vitrectomy for retinal immunodeficiency virus infection. Arch 54. Sergott RC, Anand R, Belmont JB, et al:
detachment associated with ARN. Am J Ophthalmol 1989; 107:960–961. Acute retinal necrosis neuropathy. Clinical
Ophthalmol 1988; 106:426–429. 35. Englund JA, Balfour HH: Varicella and profile and surgical therapy. Arch
15. Palay DA, Sternberg P, Davis J, et al: herpes zoster. In: Hoeprich PD, Jordan MC, Ophthalmol 1989; 107:692–696.
Decrease in the risk of bilateral ARN by eds. Infectious diseases. 4th edn. 55. Topilow H, Nussbaum JJ, Freeman HM,
acyclovir therapy. Am J Ophthalmol 1991; Philadelphia: JB Lippincott; 1989. et al: Bilateral ARN. Clinical and
112:250–255. 36. Matsuo T, Ohno A, Matsuo N: Acute retinal ultrastructural study. Ophthalmology 1982;
16. Sternberg P, Han DP, Yeo JH, et al: necrosis following chickenpox in pregnant 100:1901–1908.
Photocoagulation to prevent retinal woman. Jpn J Ophthalmol 1988; 32:70–74. 56. Barondes MJ, Tellez F, Siegel A: Acute
detachment in ARN. Ophthalmology 1988; 37. Browning DJ, Blumenkranz MS, Culbertson retinal necrosis after chickenpox in a
95:1389–1393. WW, et al: Association of varicella-zoster healthy adult. Ann Ophthalmol 1992;
17. Atherton SS, Kanter MY, Streilein JW: ACAID dermatitis with ARN syndrome. 24:335–336.
requires early replication of HSV-1 in the Ophthalmology 1987; 94:602–606. 57. Ehongo-Bidime A, Perleux A, Zanen A:
injected eye. Curr Eye Res 1990; 10:75–80. 38. Yeo JH, Pepose JS, Stewart JA, et al: Post-varicella acute retinal necrosis. Bull
18. Streilein JW, Igietseme JU, Atherton SS: Acute retinal necrosis syndrome following Soc Belge Ophthalmol 1995; 255:61–68.
Evidence that precursor cytotoxic T cells herpes zoster dermatitis. Ophthalmology 58. Blumenkranz MS: Discussion of
mediate acute necrosis in HSV-1 infected 1986; 93:1418–1422. chickenpox-associated ARN syndrome.
retinas. Curr Eye Res 1990; 10:81–86. 39. Cobo LM, Foulks GN, Liesegant T, et al: Ophthalmology 1991; 98:1645–1646.
19. Sergott RC, Belmont JB, Savino PJ, et al: Oral acyclovir in the treatment of acute 59. Hellinger WC, Bolling JP, Smith TF,
Optic nerve involvement in the ARN herpes zoster ophthalmicus. Campbell RJ: Varicella-zoster virus retinitis
syndrome. Arch Ophthalmol 1985; Ophthalmology 1986; 93:763–770. in a patient with AIDS-related complex:
103:1160–1162. 40. Sellitti TP, Huang AJ, Schiffman J, Davis JL: case report and brief review of the acute
20. Culbertson WW, Brod RD, Flynn HW, et al: Association of herpes zoster ophthalmicus retinal necrosis syndrome. Clin Infect Dis
Chickenpox-associated ARN syndrome. with acquired immunodeficiency syndrome 1993; 16:208–212.
Ophthalmology 1991; 98:1641–1646. and acute retinal necrosis. Am J 60. Walton R, Byrnes G, Chan C, et al:
21. Matsuo T, Kuyama M, Matsuo N: Acute Ophthalmol 1993; 116:297–301. Fluorescein angiography in the progressive
retinal necrosis as a novel complication of 41. Holland GN, Cornell PJ, Park MS, et al: An outer retinal necrosis syndrome. Retina
chickenpox in adults. Br J Ophthalmol association between retinal necrosis 1996; 16:393–398.
1990; 74:443–444. syndrome and HLA-DQw7 and phenotype 61. Engstrom RE, Holland GN, Margolis TP,
22. Matsuo T, Morimoto K, Matsuo N: Factors Bw62,DR4. Am J Ophthalmol 1989; et al: The progressive outer retinal necrosis
associated with poor visual function in 108:370–374. syndrome. A variant of necrotizing herpetic
ARN. Br J Ophthalmol 1991; 75:450–454. 42. Matsuo T, Matsuo N: HLA-DR9 associated retinopathy in patients with AIDS.
23. Margolis TP, Lowder CY, Holland GN, et al: with the severity of acute retinal necrosis. Ophthalmology 1994; 101:1488–1502.
Varicella-zoster retinitis in patients with the Ophthalmologica 1991; 203:133–137. 62. Greeven CM, Ford J, Stanton C, et al:
acquired immunodeficiency syndrome. Am 43. Klok AM, Geertzen R, Rothova A, et al: Progressive outer retinal necrosis
J Ophthalmol 1991; 112:119–131. Anticardiolipin antibodies in uveitis. Curr secondary to varicella zoster virus in
24. Forster DJ, Dugel PU, Frangieh GT, et al: Eye Res 1992; 11:209–213. acquired immune deficiency syndrome.
Rapidly progressive outer retinitis in 44. Thompson WS, Culbertson WW, Smiddy Retina 1995; 15:14–20.
patients with the acquired WE, et al: Acute retinal necrosis caused by 63. Morley MG, Duker JS, Zacks C: Successful
immunodeficiency syndrome. Am J reactivation of Herpes simplex virus type 2. treatment of rapidly progressive outer
Ophthalmol 1990; 110:341–348. Am J Ophthalmol 1994; 118:205–211. retinal necrosis in the acquired
25. Tran VT, Auer C, Guex-Crosier Y, et al. 45. Rosenbaum JT, Tammaro J, Robertson JE immunodeficiency syndrome. Am J
Epidemiological characteristics of uveitis in Jr: Uveitis precipitated by nonpenetrating Ophthalmol 1994; 117:264–265.
Switzerland. Int Ophthalmol 1994–1995; ocular trauma. Am J Ophthalmol 1991; 64. Pinnolis MK, Foxworthy D, Kemp B:
18:293–298. 112:392–395. Treatment of progressive outer retinal
26. Friedman SM, Mames RN, Sleasman JW, 46. Matsuo T, Nakayama T, Koyama T, et al: A necrosis with sorivudine. Am J Ophthalmol
Whitcup SM: Acute retinal necrosis after proposed mild type of ARN syndrome. Am 1995; 119:516–517.
chickenpox in a patient with acquired J Ophthalmol 1988; 105:579–583. 65. Holland GN: The progressive outer retinal
immunodeficiency syndrome. Arch 47. Duker JS, Nielsen JC, Eagle RC, et al: necrosis syndrome. Int Ophthalmol 1994;
Ophthalmol 1993; 111:1607–1608. Rapidly progressive ARN secondary to 18:163–165.
27. Saari KM, Boke W, Manthey KF, et al: Bilateral herpes simplex virus, type I. 66. Moorthy R, Weinberg D, Teich S, et al:
ARN. Am J Ophthalmol 1982; 93:403–411. Ophthalmology 1990; 97:1638–1643. Management of varicella zoster virus 2123
RETINA AND VITREOUS

retinitis in AIDS. Br J Ophthalmol 1997; 84. Duker JS, Blumenkranz MS: Diagnosis and choroidal biopsies in intraocular
81:189–194. management of the ARN syndrome. Surv inflammation. Am J Ophthalmol 1995;
67. Johnston WH, Holland GN, Engstrom RE, Ophthalmol 1991; 35:327–343. 119:597–611.
Rimmer S: Recurrence of presumed 85. Ridley ME, McDonald HR, Sternberg P, 103. Hirsch MS, Schooley RT: Treatment of
varicella-zoster virus retinopathy in patients et al: Retinal manifestations of ocular herpes virus infections (second of two
with acquired immunodeficiency syndrome. lymphoma (reticulum cell sarcoma). parts). N Engl J Med 1983; 309:1034–1039.
Am J Ophthalmol 1993; 116:42–50. Ophthalmology 1992; 99:1153–1160. 104. Holland GN, Sakamoto MJ, Hardy D, et al:
68. Cooper H, Beek P: Spontaneous regression 86. Kohno T, Uchida H, Inomata H: Ocular CMV Retinopathy Study Group: treatment
and successful laser prophylaxis in manifestations of adult T-cell leukemia/ of cytomegalovirus retinopathy in patients
progressive outer retinal necrosis syndrome. lymphoma. A clinicopathologic study. with acquired immunodeficiency syndrome.
Am J Ophthalmol 1996; 121:723–724. Ophthalmology 1993; 100:1794–1799. Arch Ophthalmol 1984; 104:794.
69. Pepose JS, Biron K: Antiviral sensitivities of 87. Gass JD, Trattler HL: Retinal artery 105. Jacobson MA, Drew WL, Feinberg J, et al:
the ARN syndrome virus. Curr Eye Res obstruction and atheromas associated with Foscarnet therapy for ganciclovir-resistant
1987; 6:201–205. non-Hodgkin’s large cell lymphoma cytomegalovirus in patients with AIDS.
70. Sarkies N, Gregor Z, Forsey T, Darougar S: (reticulum cell sarcoma). Arch Ophthalmol J Infect Dis 1991; 163:1348–1351.
Antibodies to herpes simplex virus type I in 1991; 109:1134–1139. 106. Hung SO, Patterson A, Rees PJ:
intraocular fluids of patients with ARN. Br J 88. Grossniklaus HE, Aaberg TM, Purnell EW, Pharmacokinetics of oral acyclovir (Zovirax)
Ophthalmol 1986; 70:81–84. et al: Retinal necrosis in X-linked in the eye. Br J Ophthalmol 1984;
71. Margolis T, Irvine AR, Hoyt WF, Hyman R: lymphoproliferative disease. Ophthalmology 68:192–195.
Acute retinal necrosis syndrome presenting 1994; 101:705–709. 107. Laskin OL: Acyclovir. Pharmacology and
with papillitis and arcuate neuroretinitis. 89. Ando F, Kato M, Goto S, et al: Platelet clinical experience. Arch Intern Med 1984;
SECTION 10

Ophthalmology 1988; 95:937–940. function in bilateral ARN. Am J Ophthalmol 144:1241–1246.


72. Suttorp-Schulten MS, Zaal MJ, Luyendijk 1983; 96:27–32. 108. Pulido JS, Palacio MN, Peyman GA, et al:
BS, et al: Aqueous chamber tap and 90. Pepose JS, Flowers B, Stewart JA, et al: Toxicity of antiviral drugs. Ophthalmic Surg
serology in ARN. Am J Ophthalmol 1989; Herpesvirus antibody levels in the etiologic 1984; 15:666–669.
108:327–328. diagnosis of the acute retinal necrosis 109. Perry C, Wagstaff A: Famciclovir: a review
73. Souchi S, Ozawa H, Matsuhashi M, et al: syndrome. Am J Ophthalmol 1992; of its pharmacologic properties and
Demonstration of varicella-zoster virus 113:248–256. therapeutic efficacy in herpesvirus
antigens in the vitreous aspirates of 91. Carney MD, Peyman GA, Goldberg MF, infections. Drugs 1995; 50:396–415.
patients with ARN syndrome. et al: Acute retinal necrosis. Retina 1986; 110. Perry C, Faulds D: Valaciclovir: a review of
Ophthalmology 1988; 95:1394–1398. 6:85–94. its antiviral activity pharmacokinetic
74. Culbertson WW, Blumenkranz MS, Pepose 92. Matsuo T, Nakayama T, Koyama T, et al: properties, and therapeutic efficacy in
JS, et al: Varicella-zoster is a cause of the Cytological and immunological study of the herpesvirus infections. Drugs 1996;
ARN syndrome. Ophthalmology 1986; aqueous humor in acute retinal necrosis 52:754–772.
93:559–569. syndrome. Ophthalmologica 1987; 111. Whittum JW, McCulley JP, Niederkorn JT,
75. Rochat C, Herbort CP: Acute retinal 195:38–44. Streilein JW: Ocular disease induced in
necrosis syndrome. Klin Monatsbl 93. Matsuo T, Nakayama T, Koyama T, et al: mice by anterior chamber inoculation of
Augenheilkd 1994; 204:440–449. Mild type acute retinal necrosis syndrome herpes simplex virus. Invest Ophthalmol Vis
76. Rugger-Brandle E, Roux L, Leuenberger involving both eyes at three-year interval. Sci 1984; 25:1065–1073.
PM: Bilateral ARN: identification of the Jpn J Ophthalmol 1987; 31:455–460. 112. Cousins SW, Gonzalez A, Atherton S:
presumed infectious agent. Ophthalmology 94. Immunen I, Laatikainen L, Linnanvuori K: Herpes simplex retinitis in the mouse:
1984; 91:1648–1658. Acute retinal necrosis syndrome treated clinicopathologic correlations. Invest
77. Madhavan HN, Biswas J, Gopal L, Sharma with vitrectomy and intravenous acyclovir. Ophthalmol Vis Sci 1989; 30:1485.
T: Investigations in the virological etiology Acta Ophthalmol 1989; 67:106–108. 113. Igietseme JU, Calzada PJ, Gonzalez AR,
in acute retinal inflammation. Indian J Med 95. de Boer JH, Luyendijk L, Rothova A, et al: et al: Protection of mice from herpes
Res 1995; 101:233–237. Detection of intraocular antibody simplex virus-induced retinitis by in vitro-
78. Schmitz K, Fabricius EM, Popescu M: production to herpesviruses in acute retinal activated immune cells. J Virol 1989;
Cytomegalovirus retinitis in AIDS. necrosis syndrome. Am J Ophthalmol 63:4808–4813.
Effectiveness and complications of long- 1994; 117:201–210. 114. Igietseme JU, Streilein JW, Miranda F, et al:
term therapy [German with English abstract]. 96. Davis JL, Feuer W, Culbertson WW, Mechanisms of protection against herpes
Ophthalmologe 1992; 89:416–426. Pflugfelder SC: Interpretation of intraocular simplex virus type I-retinal necrosis by in
79. Whitley RJ: Varicella-zoster virus. Infectious antibody levels in necrotizing retinitis. vitro-activated T lymphocytes. J Virol 1991;
diseases and their etiologic agents. Part III. Retina 1995; 15:233–240. 65:763–768.
In: Mandell GL, Goudlas RG, Bennet JE, 97. Kumano Y, Manabe J, Hamamoto M, et al: 115. Ando F, Kato M, Goto S, et al: Platelet
eds. Principles and practice of infectious Detection of varicella-zoster virus genome function in bilateral ARN. Am J Ophthalmol
diseases. 3rd edn. New York: Churchill having a Pst1 site in the ocular sample 1983; 96:27–32.
Livingstone; 1991:1153–1159. from a patient with acute retinal necrosis. 116. Carney MD, Peyman GA, Goldberg MF, et al:
80. Scott IU, Luu KM, Davis JL. Ophthalmic Res 1995; 27:310–316. Acute retinal necrosis. Retina 1984; 6:85.
Clinicopathologic reports, case reports, 98. Sado K, Kimura T, Hotta Y, et al: Acute 117. Blumenkranz MS, Clarkson J, Culbertson
and small case series: Intravitreal antivirals retinal necrosis syndrome associated with WW, et al: Visual results and complications
in the management of patients with aquired herpes simplex keratitis. Retina 1994; after retinal reattachment in the ARN
immunodeficiency syndrome with 14:260–263. syndrome: the influence of operative
progressive outer retinal necrosis. Arch 99. Thompson WS, Culbertson WW, Smiddy technique. Retina 1989; 9:170–174.
Ophthalmol 2002; 20:1219–1222. WE, et al: Acute retinal necrosis caused by 118. Clarkson JG, Blumenkranz MS, Culbertson
81. Whittum JW, McCulley JP, Niederkorn JT, reactivation of herpes simplex virus type 2. WW, et al: Retinal detachment following the
Streilein JW: Ocular disease induced in Am J Ophthalmol 1994; 118:205–211. ARN syndrome. Ophthalmology 1984;
mice by anterior chamber inoculation of 100. Mitchell SM, Fox JD, Tedder RS, et al: 91:1665–1668.
herpes simplex virus. Invest Ophthalmol Vis Vitreous fluid sampling and viral genome 119. Campochiaro P, Jerdan JA, Glaser BM:
Sci 1984; 25:1065–1073. detection for the diagnosis of viral retinitis Serum contains chemoattractants for
82. Cousins SW, Gonzalez A, Atherton S: in patients with AIDS. J Med Virol 1994; human retinal pigment epithelial cells. Arch
Herpes simplex retinitis in the mouse: 43:336–340. Ophthalmol 1984; 102:1376–1379.
clinicopathologic correlations. Invest 101. Pavesio CE, Mitchell SM, Barton K, et al: 120. Vidaurri JS, Glaser BM: Effect of fibrin on
Ophthalmol Vis Sci 1989; 30:1485. Progressive outer retinal necrosis (PORN) in retinal pigment epithelial cells. Arch
83. Igietseme JU, Streilein JW, Miranda F, et al: AIDS patients: a different appearance of Ophthalmol 1984; 102:1376–1379.
Mechanisms of protection against herpes varicella-zoster retinitis. Eye 1995; 121. McDonald HR, Lewis H, Kreiger AE, et al:
simplex type 1-retinal necrosis by in vitro- 9:271–276. Surgical management of retinal detachment
activated T lymphocytes. J Virol 1991; 102. Rutzen AR, Ortega-Larrocea G, Dugel PU, associated with the ARN syndrome. Br J
2124 65:763–768. et al: Clinicopathologic study of retinal and Ophthalmol 1991; 75:455–458.
CHAPTER

165 Ocular Syphilis


C. Stephen Foster and Richard R. Tamesis

Syphilis is a sexually transmitted, chronic, systemic infection is almost exclusively sexual. Transmission requires a break in
caused by the spirochete Treponema pallidum. Primary the skin, but T. pallidum can penetrate intact mucous mem-
infection is followed by an incubation period of ~3 weeks, branes. The primary mode of transmission is through sexual
usually succeeded by the appearance of a primary skin or intercourse, but transmission during oral sexual practices may
mucous membrane lesion, the chancre. This lesion, which may also occur. Transmission after blood transfusions is, in essence,
appear from 8 days to 6 weeks after infection with T. pallidum, unheard of in civilized societies today because of the screening
is usually painless and associated with regional lymph node of blood and blood products for transfusion. The likelihood of
enlargement. The chancre typically heals within a few weeks. transmission of an infectious dose of T. pallidum from an
The secondary stage of syphilis ensues. Symptoms of this stage infected to a noninfected individual during sexual intercourse is
(fever, malaise, headache, generalized lymph node enlargement, undoubtably multifactorial. One study based on a placebo-
and rash) generally appear within a few weeks or, at most, a few controlled trial of antibiotic efficacy in aborting syphilis in
months after the primary chancre has disappeared. This known contacts, however, suggested a 30% incidence of trans-
spirochetemic stage of the disease then subsides, even without mission with a single sexual encounter.3 Few organisms are
antibiotic therapy, and the infection becomes ‘latent’. needed to survive, proliferate, and produce disease. It is
Individuals with historic or serologic evidence of syphilis but estimated that the ID50 inoculum is 57 spirochetes.4
with no clinical manifestations by definition have latent
syphilis. Secondary syphilitic relapses may develop during this
state of latency. Approximately one-third of untreated cases will INCIDENCE
progress to tertiary syphilis, with syphilitic inflammatory With the exception of two periods of a rising incidence of
lesions of the heart, aorta, brain, kidney, bone, eye, or skin. syphilis, the incidence of this disease has been steadily
decreasing since 1940. Infant deaths resulting from syphilis
CAUSE and new admissions of patients with syphilitic psychoses have
fallen 99% since 1940 in the United States, and the total
The organism responsible for syphilis, T. pallidum, was number of cases of late and latent syphilis has fallen 98% since
discovered by Schaudinn and Hoffman of Hamburg in 1905 in 1943. A decrease of 98% in the number of congenital syphilitic
inflammatory lesions from a patient with syphilis. This cases has occurred since 1941.1 An increase in the incidence
organism is a thin, spiral-shaped parasite for whom the only of syphilis in World War II was noted in all Western countries;
known natural host is Homo sapiens. Other mammals can be this incidence rapidly fell in the 1950s. A smaller rise occurred
infected with the organism. The origins are unknown, and between 1971 and 1980, which can be accounted for primarily
several hypotheses exist regarding the development of syphilis by the increasing incidence of syphilis in the homosexual
in humans. Two main theories, one tracing the development community. By 1988, there were 40 275 new cases of primary
from the tropics and the other tracing the development from and secondary syphilis reported in the United States. That
native Americans, are most commonly espoused. The first clear number had fallen to 16 500 in the 1995 Summary of
descriptions of clinical evidence of syphilis were recorded at the Notifiable Diseases, and to 7980 in 2004.4,5 Age-related data
end of the fifteenth century, when a pandemic known as the show early syphilis to be concentrated in young adults, with
Great Pox, as distinguished from smallpox, swept over Europe many more male than female cases reported.2 The reported
and Asia.1 Warfare in the fifteenth century, including the seige incidence is higher among nonwhites than among whites,
of Naples by Charles VIII of France, was associated with the higher in urban areas than in rural areas, higher in the southern
spread of what today is known as syphilis among soldiers, and southwestern United States, and higher in those states
prostitutes, and other women who followed the warriors. The with large urban populations. Grassly and coworkers used
disease became known as the French disease among Italians time series of annual disease reports for 68 US cities to
and the Italian disease among the French. It supposedly demonstrate marked 8–11 year cycles in syphilis incidence from
received its present name from a poem written in 1530 by the 1980s, which had previously been attributed to changes
Fracastoro about an infected shepherd named Sifilis.2 in sexual behavior.6 Instead, they showed that the dynamics
of syphilis infection can be explained by the ‘susceptible–
EPIDEMIOLOGY infected–recovered’ (SIR) model for microparasitic infections.7,8
Syphilis stimulates significant but imperfect immunity
following recovery from infection. In the SIR model, oscillations
TRANSMISSION in incidence of disease are produced by prolonged immunity
With the notable exception of transplacental transmission from following infection combined with a short infection period. 2125
an infected mother to her fetus, the transmission of T. pallidum Cycles occur because major epidemics exhaust their supply of
RETINA AND VITREOUS

FIGURE 165.1. Iris nodule, syphilitic uveitis. Note the true mass in the
SECTION 10

substance of the iris periphery at the 8–9 o’clock position.


FIGURE 165.2. Sector retinitis, syphilitic uveitis, retinal vasculitis, and
retinitis. Note the sector of the retina, beginning at the disk and
extending superiorly, with infiltrate retinitis and associated retinal
vasculitis.
susceptible individuals. The number of individuals in at-risk
groups then gradually increases, eventually providing enough
numbers for the next major outbreak.

OCULAR MANIFESTATIONS iris), iritis papulosa (with the iris roseati increasing in size to
resemble a papule), and iritis nodosa (with increasing size of iris
papulosa forming a yellow–red nodule) may be relatively restric-
UVEA ted to cases of syphilitic uveitis that are associated with classic
Syphilis was believed to be a common cause of iritis before the secondary syphilis with extraocular manifestations. Although it
antibiotic era. In the decade from 1970 to 1980 in a large is typically thought of syphilis as being a ‘granulomatous’ dis-
referral uveitis practice, Schlaegel and Kao estimated that only ease and, hence, imagine that the iritis of syphilis should be
1.1% of their uveitis cases were secondary to syphilis.9 However, a ‘granulomatous’ one with mutton-fat keratic precipitates on
they emphasized one of the most important points for the corneal endothelium, in fact approximately half of the
ophthalmologists to remember; they did not initially suspect patients exhibit no keratic precipitates or fine keratic preci-
syphilis in most of their 28 patients who were ultimately shown pitates on the endothelium. One case with iritis nodosa has
to have this disease, and many of their patients had a previously been reported,10 and the iris lesion in the right eye of
nonreactive Venereal Disease Research Laboratory (VDRL) test. this patient, shown in Figure 165.1, is a good example of
The authors emphasized, once again, the ‘great imitator’ syphilitic uveitis nodosa.
capabilities of syphilis and also stressed the importance of the Inflammatory cells may accumulate in the vitreous body to
fluorescent treponemal antibody absorption (FTA-ABS) test in varying degrees (including nearly to the point of vitreal
the routine screening of all patients with intraocular opacification) in syphilitic panuveitis or posterior uveitis. It
inflammation. If this test had not been used in their inves- may be difficult to evaluate the choroid and retina in such
tigations, three-fourths of their syphilitic iritis cases would have instances, but if and when the retina can be seen, sectors or
gone undiagnosed. This point is reemphasized as a result of an foci of active choroiditis will usually be found. Placoid
experience with 25 of 1020 new uveitis referral cases seen chorioretinitis may occur in immunocompetent11,12 or in
between 1 Jan 1983, and 30 Jan 1989.10 This 2.45% portion of human immunodeficiency virus (HIV)-infected individuals.13
new uveitis referral population had previously undiagnosed The outer retina, retinal pigment epithelium, and choroid
syphilis as the cause of their chronic or recurrent intraocular are affected, and the lesions tend to present in the posterior
inflammation. More than one-third of these patients had a pole. Vitreal cells are typical, and a serous detachment
nonreactive serum VDRL test; all had a positive FTA-ABS test secondary to an outpouring of serum and inflammatory cells
and, on further testing, a positive microhemagglutination assay- may produce the unusual appearance of a macular or retinal
T. pallidum (MHA-TP) test. All had total resolution of their ‘pseudohypopyon’.12 A diffuse retinitis or neuroretinitis without
uveitis with systemic intravenous penicillin therapy at doses choroiditis can also occur, as can papillitis. Disk edema in a
adequate for neurosyphilis. patient with periphlebitis and cells in the vitreous,14 central
The iritis of syphilis has no remarkably distinguishing retinal vein occlusion with disk edema,15 or even simple disk
features. Although some authors have stated that ocular mani- edema (caused by a gumma of the optic nerve)16 should
festations of syphilis are more likely to arise in the secondary probably be sufficient reason for considering the possibility of
stage of acquired syphilis, it is emphasized that in almost every syphilis and requesting an FTA-ABS test. Indeed, a healthy
instance in a referral practice, the patients have had latent dictum for all ophthalmologists to follow would be that any
syphilis with no clinical manifestations prompting a suspicion patient with iritis and papillitis should be considered to have
of this disease. In addition, although the older literature divided syphilis unless proved otherwise. An example of sectoral
syphilitic iritis into three types according to iris features, that in neuroretinitis secondary to undiagnosed syphilis is shown in
only one of the cases showed an iris pathologic condition that Figure 165.2, and an example of undiagnosed syphilitic
prompted a specific suspicion for syphilis. It is suspected that multifocal chorioretinitis is shown in Figure 165.3. Figure
2126 iritis roseati (with small dilated collections of capillaries in the 165.4 depicts syphilitic uveitis and associated papillitis.
Ocular Syphilis

TABLE 165.1. Causes of Interstitial Keratitis

Bacterial
Syphilis
Leprosy
Tuberculosis
Chlamydia
Lyme agent
Protozoal
Acanthamoeba
Malaria
Trypanosomiasis
Leishmaniasis

CHAPTER 165
FIGURE 165.3. Syphilitic multifocal choroiditis. Multifocal Other
chorioretinal lesions, now healed, with scarring in a patient with Cogan’s syndrome
previous active syphilis with multifocal choroiditis.
Sarcoidosis
Lymphoma
Viral
Herpes simplex
Herpes zoster
Epstein–Barr virus
Rubella
Rubeola
Vaccinia
Variola
Mumps
Helminthic
Cysticercosis
Onchocerciasis

FIGURE 165.4. Syphilitic uveitis with papillitis. Note the swelling of


the optic nerve and the hazy view of the nerve secondary to the
associated inflammatory cells in the vitreous anterior to the disc.
interstitial keratitis (Table 165.1), but syphilis may be the
second most common cause after herpes simplex virus.24
Approximately 10% of cases of interstitial keratitis secondary
NEUROOPHTHALMIC MANIFESTATIONS to syphilis are associated with acquired syphilis; 90% are seen
In addition to the possibility of papillitis previously described, with congenital syphilis. Syphilitic interstitial keratitis may be
optic neuritis,17–19 optic perineuritis,20,21 and papilledema22 diffuse and generalized, or it may be localized. When localized,
have been described as ocular manifestations in patients with the area affected is commonly a sector of cornea (Fig. 165.5).
ocular syphilis. Zambrano and associates reported the The keratitis may be subtle, and in the photophobic patient
occurrence of bilateral syphilitic optic neuritis in a bisexual man with minimal circumlimbal injection, patience and practice are
with AIDS; this patient’s visual acuity deteriorated over a period required to discover the subtle, diffuse patina of tiny, tan
of ~12 h from 20/20 bilaterally to total bilateral blindness.23 inflammatory cells in the affected area of the corneal stroma. As
Toshniwal reported optic perineuritis, characterized by swollen the inflammatory process increases, the density of the
optic discs without raised intracranial pressure and visual dys- inflammatory cell population rises, and the ‘infiltrate’ is easier
function, in a patient with secondary syphilis and a complaint to see, as is the associated corneal edema. An associated iritis
of recurrent headache.21 with or without keratic precipitates may develop, and peripheral
corneal neovascularization may ensue. Untreated, the keratitis
may progress to involve the entire cornea, with progressive
CORNEA AND SCLERA neovascularization of the stroma, enormous photophobia, and
Syphilis can cause corneal inflammation, and unlike the multi- discomfort and decreased visual acuity for the patient. The
faceted uveitis presentations possible with syphilis, syphilitic inflammatory process may slowly regress over the ensuing
keratitis is typically an interstitial keratitis (i.e., a nonulcerative 2 years, leaving a scarred cornea with emptied or ‘ghost’ stromal
and nonsuppurative inflammation of the corneal stroma). vessels (Fig. 165.6). One or both eyes may be affected. In
Untreated, this stromal keratitis is frequently accompanied congenital syphilitic keratitis, both eyes are affected, either
by stromal neovascularization. A variety of agents may cause simultaneously or sequentially, in more than 75% of patients.18 2127
SECTION 10 RETINA AND VITREOUS

FIGURE 165.5. Sector interstitial keratitis and luetic, old, inactive


sector interstitial keratitis with stromal scarring in the inferonasal
quadrant in a patient with previously treated syphilis.

Syphilitic scleritis may be nodular or diffuse. We have never


seen a case of necrotizing scleritis secondary to syphilis.
Syphilitic scleritis has no distinguishing features and, hence, is FIGURE 165.6. Luetic sector interstitial keratitis with ghost vessels in
diagnosed only because the ophthalmologist has cleverly the deep corneal stroma (extremely difficult to capture on film and
included, as routine, an FTA-ABS test as part of the diagnostic reproduce).
survey in all patients with scleritis, interstitial keratitis, or
uveitis.

DIAGNOSIS
Syphilis may be definitively diagnosed by direct or indirect
techniques. Direct techniques include the darkfield exam-
ination, in which exudate from a suspected syphilitic lesion
is examined by microscopy with a darkfield technique. The
corkscrew-shaped T. pallidum motile organisms are seen
directly. Fluorescein-labeled anti-T. pallidum antibodies may
be used in an immunofluorescence analysis of exudate or
material taken from a suspected syphilitic lesion. The antibody
will stick to syphilitic organisms in the exudate or specimen
and will be seen by the characteristic apple-green fluorescence
when the specimen is examined under the fluorescence
microscope. Detection of spirochetes in tissue specimens can
be accomplished through special staining techniques, including
the Warthin–Starry method. The spirochetes are seen directly in
the tissue specimens as shown in Figure 165.7.
Indirect techniques depend on serologic studies, including FIGURE 165.7. Spirochetes (arrow) in the cornea, as demonstrated
Treponema-specific and nontreponemal tests. These nontre- by silver stain, after penetrating keratoplasty in a patient with
ponemal tests detect antibodies directed against lipoidal secondary syphilis and active interstitial keratitis.
antigens. The primary nontreponemal test used in the United
States is the VDRL. This test is typically positive in patients
with active syphilis and negative in patients with successfully opinion, lies in monitoring the response to treatment. A
treated syphilis. In patients with latent syphilis, however, the persistent fall in VDRL titers after treatment provides essential
serum VDRL test is only ~70% sensitive. As discussed before, evidence of an adequate response to therapy. Polymerase chain
35% of Schlaegel and Kao’s cases of ocular syphilis would have reaction (PCR) testing for T. pallidum DNA is the most
been misdiagnosed if the authors had relied solely on the VDRL sensitive and specific test, but it is currently not economically
as the diagnostic test for syphilis.8 More than a third of the feasible for routine analyses.25
patients have nonreactive serum VDRL tests.9 These findings
dramatically emphasize the importance of never relying on the
VDRL as the sole screening test for the possibility of ocular EVALUATION FOR ASYMPTOMATIC
syphilis. The standard treponemal tests for syphilis in the NEUROSYPHILIS
United States are the FTA-ABS and the MHA-TP. The FTA-ABS Patients with positive serologic tests for syphilis should have
test is 98% sensitive, even in latent syphilis. This test will their cerebrospinal fluid (CSF) examined for VDRL titers, total
remain positive for life, regardless of whether the patient has protein, and cell counts including a differential count. Without
been treated. The VDRL titer, in contrast, reflects the systemic a positive CSF VDRL test, an elevated white blood cell count
2128 activity of the disease, and hence, its major value, in our with a predominance of lymphocytes in the CSF or an elevated
Ocular Syphilis

total protein level is indicative of neurologic involvement, and patients with ocular syphilis should be hospitalized and treated
the patient should be treated accordingly as for neurosyphilis. using 18-24 million units per day of intravenous aqueous
crystalline penicillin G (3–4 million units every 4 h or con-
tinuous infusion) for 10–14 days. An alternative regimen con-
EVALUATION FOR HIV sisting of 10–14 days of procaine penicillin 2.4 million units IM
There are reports in the literature of HIV-positive young adult once daily with plus probenecid 500 mg orally four times a day
patients with concurrent ocular syphilis.26 This is not sur- can be used if compliance with therapy can be ensured. Some
prising given the similar mode of transmission in both disease infectious disease specialists also administer benzathine
entities. There is evidence that syphilis may pursue a more penicillin 2.4 million units IM once a week up to 3 weeks after
aggressive course in patients who are concurrently infected with completion of these neurosyphilis treatment regimens. The
HIV, rendering standard therapy for primary and secondary Jarisch–Herxheimer reaction, which is due to lysis of spiro-
syphilis inadequate.27 In view of these reports, we believe that chetes and release of endotoxin-like antigens triggering an
all patients with ocular syphilis should now be evaluated for autoimmune crisis, can develop in HIV-positive and secondary
HIV and vice versa. The Centers for Disease Control recom- stage patients. Patients should be informed about this possible
mends that an asymptomatic patient with a positive tre- adverse reaction. An acute febrile reaction frequently accom-
ponemal confirmatory test and whose VDRL titer exceeds 1:32 panied by headache, myalgia and other flu-like symptoms
or alternatively whose CD4 Th count is below 350 should be occurs within the first 24 h after any therapy for syphilis and
evaluated for neurosyphilis.28 HIV-infected patients and those may induce early labor or cause fetal distress in pregnant

CHAPTER 165
patients with ocular, tertiary and neurosyphilis should have women. This concern should not, however, prevent or delay
a reassessment of the CSF by VDRL every 3 months. A source therapy. The use of aspirin or steroids may prevent this.32
of substantial concern arises with reports of proven29 or sus- Patients can become re-infected. For this reason, all sexual
pected syphilis30 in seronegative HIV-infected individuals. partners must be screened and treated. HIV testing and sexually
Clinical suspicion and penicillin therapeutic trials have been transmitted disease counseling should also be offered.
enlightening in these instances. Emergence of molecular diag-
nostic techniques (e.g., polymerase chain reaction technology)
makes it possible now to more definitively establish evidence of PATIENTS WITH PENICILLIN ALLERGY
microbial causes of uveitis,31 and we believe increasing use of Some authors have recommended doxycycline,33 tetracycline,
this technology on aqueous and vitreous specimens will clarify or erythromycin in the treatment of penicillin-allergic patients
the cause as microbial in many instances of unusual or atypical with syphilis. It is as yet unclear whether this therapeutic
ocular inflammation. approach is sufficient therapy in most patients with neuro-
syphilis, and little to no experience exists with these treatment
TREATMENT regimens in patients with ocular syphilis. Some investigators
believe that ‘penicillin-allergic’ patients should be carefully
evaluated and possibly desensitized and then treated with
PATIENTS WITHOUT PENICILLIN ALLERGY penicillin. Many patients with penicillin ‘allergy’ in fact do not
Patients with ocular syphilis should be treated in the same way have a true allergy at all. Careful allergy testing is advocated in
that patients with tertiary syphilis are treated. This philosophy patients with syphilis who claim to have a penicillin allergy.
has been adopted for two reasons: (1) the blood–ocular barrier is Such patients who have negative skin test results can safely
as much a hindrance to adequate spirocheticidal doses of undergo the aforementioned penicillin treatment program with
penicillin as is the blood–brain barrier and (2) some evidence close in-hospital monitoring. In addition, although penicillin
exists suggesting that patients with syphilitic ocular inflam- allergy desensitization can be lengthy, costly, and complex, this
mation may have central nervous system syphilis without alternative for treating the individual with ocular syphilis or
VDRL-positive CSF. We therefore recommend that the assist- neurosyphilis who has been proved to be allergic to penicillin is
ance of an infectious disease expert be obtained and that all not without merit.

REFERENCES
1. Poitevin M, Collart P, Bolgert M: Syphilis in 8. Grenfell BT, Bjørnstad ON, Kappey J: 15. Primo S: Central vein occlusion in a young
1986. J Clin Neuroophthalmol 1987; 7:11. Travelling waves and spatial hierarchies in patient with seropositive syphilis. J Am
2. Fracastoro: Le mal franais ou syphilis. measles epidemics. Nature 2001; 414:695. Optom Assoc 1990; 61:896.
Extrait du livre de ‘contagionibus et 9. Schlaegel TF Jr, Kao SF: A review (1970– 16. Smith JL, Byrne SF, Cambron CR:
contagiosis morbis’ (1546) avec 1980) of 28 presumptive cases of syphilitic Syphiloma/gumma of the optic nerve and
commentaires de A. Fournier. 1869. uveitis. Am J Ophthalmol 1982; 93:412. human immunodeficiency virus
3. Martin JP: Conquest of general paresis. 10. Tamesis RR, Foster CS: Ocular syphilis. seropositivity. J Clin Neuroophthalmol 1990;
BMJ 1972; 2:159. Ophthalmology 1990; 97:1281. 10:175.
4. Centers for Disease Control and Prevention: 11. Gass JDM, Braunstein RA, Chenoweth RG: 17. Graveson GS: Syphilitic optic neuritis.
Summary-Cases of specified notifiable Acute syphitic posterior placoid J Neurol Neurosurg Psychiatry 1950; 13:216.
disease-United States. MMWR 1996; choridretinitis. Ophthalmology 1990; 18. Walsh FB: Syphilis of the optic nerve.
44:11. 97:1288. Ophthalmology 1956; 60:39.
5. Centers for Disease Control and Prevention: 12. Ouano DP, Brucker AJ, Saran BR: Macular 19. Weinstein JM, Lexow SS, Ho P, Spickards
Primary and secondary syphilis – United pseudohypopyon from secondary syphilis. A: Acute syphilitic optic neuritis. Arch
States, 2003-2004. MMWR 2006; 55;10: Am J Ophthalmol 1995; 119:372. Ophthalmol 1981; 99:1392.
269–273. 13. Dodds EM, Lowder CY, Boskovich SA, 20. Rush JA, Ryan EJ: Syphilitic optic
6. Grassly NC, Fraser C, Garnett GP: Host et al: Simultaneous syphilitic necrotizing perineuritis. Am J Ophthalmol 1981; 91:404.
immunity and synchronized epidemics of retinitis and placoid chorioretinitis in 21. Toshniwal P: Optic perineuritis with
syphilis across the United States. Nature acquired immune deficiency syndrome. secondary syphilis. J Clin Neuroophthalmol
2005; 433:417. Retina 1995; 15:354. 1987; 7:6.
7. Anderson RM, May RM: Infectious diseases 14. Halperin LS, Berger AS, Grand MG: 22. Schatz NJ, Smith JL: Non-tumor causes of
of humans: dynamics and control. Oxford Syphilitic disc edema and periphlebitis the Foster-Kennedy syndrome. J Neurosurg
University Press; 1992. [photo essay]. Retina 1990; 10:223. 1967; 27:37. 2129
RETINA AND VITREOUS

23. Zambrano W, Perez GM, Smith JL: Acute 27. Johns DR, Tierney M, Felsenstein D: 30. Halperin LS: Neuroretinitis due to
syphilitic blindness in AIDS. J Clin Alteration in the natural history of seronegative syphilis associated with
Neuroophthalmol 1987; 7:1. neurosyphilis by concurrent infection with human immunodeficiency virus. J Clin
24. Duke-Elder SS, Leigh AG: Diseases of the the human immunodeficiency virus. N Engl Neuroopthalmol 1992; 12:171.
outer eye. System of ophthalmology series. J Med 1987; 316:1569. 31. deBoer JH, Verhagen C, Bruinenberg M,
St Louis: CV Mosby; 1965:790. 28. Marra CM, Maxwell CL, Smith SL, et al: et al: Serologic and polymerase chain
25. Noordhoek GT, Wolters EC, de Jonge MEJ, Cerebrospinal fluid abnormalities in reaction analysis of intraocular fluids in the
van Embden JDA: Detection by polymerase patients with syphilis: association with diagnosis of infectious uveitis. Am J
chain reaction of Treponema pallidum in clinical and laboratory features. J Infect Dis Ophthalmol 1996; 121:650.
cerebrospinal fluid from neurosyphilis 2004; 189:369. 32. Brown ST. Adverse reactions in syphilis
patients before and after antibiotic 29. Hicks CB, Benson PM, Lypton GP, et al: therapy. J Am Venere Dis Assoc 1967;
treatment. J Clin Microbiol 1991; 29:1976. Seronegative secondary syphilis in a 3:172.
26. Passo MS, Rosenbaum JT: Ocular syphilis in patient infected with the human 33. Tramont EC: Syphilis in the AIDS era.
patients with human immunodeficiency virus immunodeficiency virus with Kaposi N Engl J Med 1987; 316:1600.
infection. Am J Ophthalmol 1988; 106:1. sarcoma. Ann Intern Med 1987; 107:492.
SECTION 10

2130
CHAPTER

166 Subretinal Fibrosis and Uveitis Syndrome


Tina Scheufele, Jay S. Duker, David R. Guyer, and Evangelos S. Gragoudas

HISTORICAL PERSPECTIVE patients were taking oral contraceptives.8 However, all of these
associations are probably unrelated to the ocular condition.
The subretinal fibrosis and uveitis syndrome is a rare distinct
posterior uveitis, which was first described by Palestine and SYMPTOMS
associates in 1984.1 These authors described three patients
with the unusual findings of progressive subretinal fibrosis and Patients usually complain of acute vision loss, often with meta-
uveitis. The condition most commonly occurs in healthy, morphopsia. Scotomas and photopsias also may be reported.
young, myopic females, usually with only minimal signs of
ocular inflammation. Early in the disorder, multiple small, OBJECTIVE FINDINGS
whitish-yellow retinal pigment epithelial or choroidal lesions
are observed in the posterior pole and mid-periphery. In the The vision loss may be very mild or severe, depending on
later stages of the disease, progressive subretinal fibrosis occurs. the stage of the disease at which the patient presents (see
The condition usually becomes bilateral, but often initially Table 166.1). Early in the disease course, the visual acuity can
presents unilaterally. The first histopathologic and immuno- range from 20/20 to 20/400.6,8 As the disease progresses and
histopathologic features of this condition were subsequently subretinal fibrosis develops, the visual acuity may be decreased
described by the same group.2,3 severely to counting-fingers, light perception, or even no light
When it was first being described, the subretinal fibrosis and perception.10–13 The condition is usually bilateral (45–100% of
uveitis syndrome was given many different names and was cases)1–11; however, the fellow eye is often asymptomatic
often grouped together with other chorioretinal inflammatory initially and may not become involved until months later. For
diseases. Most commonly, it was grouped with multifocal example, in one series of five patients, one case remained
choroiditis, with some believing that this was a rarely observed unilateral, one case presented with bilateral involvement, and
late stage in the spectrum of this disease.4–6 It was also three cases that were initially unilateral involved the second eye
classified as a ‘disciform macular degeneration of young adults’ within 3–6 months.6
by Doran and Hamilton.7 However, in retrospect, many of these Slit-lamp examination may reveal an anterior or posterior
patients probably had choroidal neovascular membranes that uveitis (Fig. 166.1).1–11 Usually, however, the inflammation is
lead to disciform scarring, rather than the subretinal fibrosis mild when present. A mild chronic vitritis is often associated
syndrome.7 Now, it is considered a distinctly separate entity with transient, multiple, small (100–500 mm), round, discrete,
from multifocal choroiditis, punctate inner choroidopathy, and yellowish white lesions of the retinal pigment epithelium
other chorioretinal inflammatory diseases. or choriocapillaris in the posterior pole and mid-periphery
(Fig. 166.1).1–11 These lesions may fade or enlarge and coalesce
PATIENT CHARACTERISTICS to create multiple areas of whitish subretinal fibrosis
(Fig. 166.2; see also Fig. 166.1).1–11 The progression of
The subretinal fibrosis and uveitis syndrome occurs predomi-
nantly in young, healthy, myopic females. The patients are
usually less than 35 years of age.1–11 In one series of 11 patients,
the age range was 24–43 years, with a mean age of 30.2 years.8 TABLE 166.1. Findings in the Subretinal Fibrosis and Uveitis
The disease has been described in patients as young as 6 years Syndrome
and as old as 76 years.4,5,9 In several series, all of the patients Anterior uveitis
were female,1,2,6,8,10 although a meta-analysis of all large
published series found that 86% of all reported cases were Vitritis
female.11 The patients usually have a myopic refractive Multifocal choroiditis
error.7,9,11 In Morgan and Schatz’s series,8 10 of 11 patients were
Subretinal fibrosis
myopic, with a range of –2.75 to –8.50 D. No racial predilection
has been identified.11 Systemic evaluation of these patients is Optic disc edema
almost always unremarkable.1–9 Cases have been reported in Serous and hemorrhagic macular detachment
patients with Reiter’s syndrome; migraine; atopy; schizoid syn-
drome; gastric bypass surgery; positive skin, serologic, and Cystoid macular edema
biopsy findings of histoplasmosis; and a positive purified Choroidal neovascularization
protein derivative test for tuberculosis.1–9 In one series, six of 11
2131
RETINA AND VITREOUS

a b c

FIGURE 166.1. This 26-year-old woman presented with decreased vision, redness, and pain in both eyes. (a) Slit-lamp examination revealed
conjunctival injection, anterior chamber reaction, posterior synechiae, and a cataract. (b and c) Examination of the fundus showed subretinal
fibrosis.

FIGURE 166.2. (a–d) The extensive subretinal


fibrosis that occurs in this syndrome is
SECTION 10

illustrated in this patient.


Courtesy of Robert Nussenblatt, MD.

a b

c d

subretinal fibrosis may occur over months to years, and relapses subretinal fibrosis shows late staining.1,2,6 Optic disk leakage
and recurrences are common. and macular edema may be present. CNV is commonly
Other findings may include optic disk edema, serous and observed later in the disease course.5,6,8,11
hemorrhagic macular detachment, macular hole, cystoid Electroretinographic signals may be depressed.1,2 The
macular edema, and choroidal neovascularization (CNV).1–11 electrooculogram may be markedly decreased1,2 or normal.6,8
CNV may occur in up to 44% of patients at some time during Three patients in one series8 had normal color vision testing
the course of the disease.5,6,8,11 In one study, CNV occurred 2–6 results.
months after the onset of the disease, during the fibrotic stage
of the disease.5–8 NATURAL HISTORY
FLUOROANGIOGRAPHIC AND In all but one series,8 the visual prognosis of these patients was
ELECTROPHYSIOLOGIC FINDINGS poor. Many of these patients had final visual acuities of 20/200
to counting-fingers in the more severely affected eye.5,6,8,10,11
Retinal pigment epithelial window defects or mottled hyper- Two case reports documented visual decline to no light
fluorescence of the acute lesions are observed by fluorescein perception.12,13 This poor visual prognosis was confirmed in all
2132 angiography in the early stages of this disorder.1–11 The of the other reports except for the series of Morgan and Schatz.8
Subretinal Fibrosis and Uveitis Syndrome

FIGURE 166.3. (a) This 18-year-old woman


presented with the subretinal fibrosis and
uveitis syndrome and underwent a chorioretinal
biopsy. (b) Histopathologic examination
revealed a thickened choroid that contained
numerous lymphocytes and plasma cells.
Connective tissue replaced the normal retina.
(c) Histopathologic study in another patient
with the subretinal fibrosis and uveitis
syndrome revealed thick, fibrous tissue
interposed between the markedly gliotic retina
and the choroid.
Courtesy of Chi-Chao Chan, MD and Robert
Nussenblatt, MD.

a b c

CHAPTER 166
These authors noted a final vision of 20/20 to 20/70 in patients PATHOPHYSIOLOGY
treated with oral or periocular corticosteroids and vision of
20/400 in patients who were not treated. The beneficial effect of This condition is probably due to a localized autoimmune
steroids or other immunosuppressants is still controversial. antibody-mediated inflammatory process that destroys the
Although some have noted an improvement in vision and retinal pigment epithelium and may cause massive subretinal
halting of the fibrotic response when steroids were started early fibrosis.1–3,6,12 These patients have no evidence of systemic
in the disease course, most authors agree that once subretinal disease. Antibodies against the photoreceptors and the retinal
fibrosis occurs, the response to corticosteroids is minimal at pigment epithelium have been detected in the serum of affected
best and the visual prognosis is generally poor.5,6,10,11 The eye patients.12 These antibodies, likely produced by the infiltrating
affected first usually has the worst final visual acuity, except in plasma cells, lead to destruction of the retinal pigment
cases where a subfoveal CNV develops in the second eye.5,6,10 epithelium and formation of fibrotic tissue.3,12 Activated Müller
Recurrences are common in this disorder and have been cells likely stimulate retinal gliosis and further subretinal
documented to range from two to seven recurrences per patient. fibrosis.3

HISTOPATHOLOGIC AND TREATMENT


IMMUNOHISTOPATHOLOGIC FINDINGS
Once subretinal fibrosis occurs, there appears to be no beneficial
Histopathologic studies of chorioretinal biopsies from patients treatment.6,10,11 Although controversial, steroids may be useful
with this condition demonstrated a uveal inflammatory infil- early in the disease. Some have documented visual
trate, retinal gliosis, and subretinal fibrosis (Fig. 166.3). 2,3,12,13 improvement and halting of the fibrotic response when steroids
The inflammatory infiltrate was composed of plasma cells and were started during the acute phase.5,6,8 However, many more
lymphocytes.2,3,12 The subretinal space and choroid contained cases have been reported where patients showed no improve-
complement C3, immunoglobulin G, epithelioid histiocytes, ment and their disease continued to progress.10–13 In one series,
multinucleated giant cells, noncaseating granulomas, and there was a dramatic response to systemic and periocular
fibrin.2,3,12 Stains for bacteria and fungi were negative, and corticosteroids.8 All nine treated patients showed visual
polymerase chain reaction (PCR) did not detect any herpes improvement after treatment (final visual acuity 20/20 to
viruses.3,12 20/70), whereas the two untreated patients had a poor visual
Whereas T-cells normally make up 90% of the peripheral prognosis (visual acuity of 20/400).8 In this same series, one
blood lymphocytes, T- and B-cells were found in equal numbers patient developed a CNV that appeared to respond to steroid
in the iris, ciliary body, and choroid of patients with this treatment. In another case series, six of 18 patients responded
condition, indicating a large increase in the number of B-cells.3 with an improvement in visual acuity after treatment with
Within the T-cell population, there was a relative increase in steroids, but steroids failed to prevent the subfoveal progression
the number of T-helper cells, whose functions include B-cell of CNV in the one patient who declined laser treatment for
recruitment and granuloma formation.3 Some authors have an extrafoveal CNV.5 Thus, it seems reasonable to consider
proposed an association of the subretinal fibrosis and uveitis steroid treatment for acute cases but not for cases in which
syndrome with sympathetic ophthalmia, on the basis of subretinal fibrosis has already occurred.10 Other immuno-
histologic similarities.12,13 However, the predominance of suppressants have been tried as well, but experience using these
T-cells in sympathetic ophthalmia and B-cells in the subretinal for the subretinal fibrosis and uveitis syndrome is limited.
fibrosis syndrome argues against a close association.12,13 Cyclophosphamide and azathioprine have each been used in
By light microscopy, the retina was gliotic and markedly one reported case, but failed to halt the progression of disease in
attenuated due to loss of photoreceptors and thinning of the these two patients, who both already had extensive subretinal
bipolar cell layer.3 The Müller cells were thickened and fibrosis at the time of treatment.3,13
expressed class II antigen, suggestive of active proliferation.3
The retinal pigment epithelium was absent and replaced by DIFFERENTIAL DIAGNOSIS
amorphous connective tissue.3 Electron microscopy showed
that this subretinal tissue contained retinal pigment epithelial The acute phase of this condition can mimic many entities,
cells.3 It has been proposed that both the retinal pigment although the disease is easy to identify once progressive
epithelial cells and the Muller cells are responsible for the subretinal fibrosis occurs (Table 166.2). Acutely, the differential
subretinal fibrosis.3 diagnosis includes sarcoidosis, presumed ocular histoplasmosis 2133
RETINA AND VITREOUS

acute retinal pigment epitheliitis, acute macular neuro-


TABLE 166.2. Differential Diagnosis in the Subretinal Fibrosis
retinopathy, and inflammatory pseudohistoplasmosis. Recently,
and Uveitis Syndrome
a few authors proposed that this condition may, in fact, be a
Sarcoidosis variant of sympathetic ophthalmia based on histopathologic
Presumed ocular histoplasmosis syndrome
similarities; however, the immunohistopathology is very
different in these two conditions and both of the patients
Tuberculosis described in these case reports had prior intraocular surgery.12,13
Syphilis The clinical history, inflammatory reaction, and progression to
subretinal fibrosis can distinguish the subretinal fibrosis and
Birdshot chorioretinopathy
uveitis syndrome from each of these conditions. In the later
Toxoplasmosis stages of this disease, choroidopathies with CNV and disciform
Fungal infections
scarring, such serpiginous choroidopathy, may be confused with
this entity.
Acute posterior placoid pigment epitheliopathy
Serpiginous choroidopathy CONCLUSIONS
Multiple evanescent white dot syndrome
The subretinal fibrosis and uveitis syndrome is a condition that
Diffuse unilateral subacute neuroretinitis generally affects young, otherwise healthy, myopic females in
SECTION 10

Punctate outer retinal toxoplasmosis which progressive subretinal fibrosis occurs some time after the
onset of a posterior uveitis and choroiditis. CNV and macular
Sympathetic ophthalmia edema may also occur. The visual prognosis is generally poor.
Acute retinal pigment epitheliitis Corticosteroid treatment may be beneficial during the acute
stages of the disorder, and chemotherapy may be considered in
Acute macular neuroretinopathy
severe cases. However, once subretinal fibrosis occurs, the
Inflammatory pseudohistoplasmosis visual prognosis and response to treatment is generally poor.
The disease is probably caused by a localized autoimmune
reaction in which antibodies destroy the retinal pigment
epithelium and produce subretinal fibrosis. The condition is
now considered a distinctly separate entity from multifocal
syndrome, tuberculosis, syphilis, birdshot chorioretinopathy, choroiditis and panuveitis, punctate inner chorioretinopathy,
toxoplasmosis, fungal infections, acute posterior multifocal and other chorioretinal inflammatory diseases. Although the
placoid pigment epitheliopathy, serpiginous choroidopathy, acute stages of this condition may be confused with other types
multiple evanescent white dot syndrome, punctate inner of choroiditis, the progressive subretinal fibrosis observed
choroidopathy, diffuse unilateral subacute neuroretinitis, during the late stages distinguishes this disorder from most
punctate outer retinal toxoplasmosis, sympathetic ophthalmia, other diseases.

REFERENCES
1. Palestine AG, Nussenblatt RB, Parver LM, ocular histoplasmosis. Arch Ophthalmol diffuse subretinal fibrosis syndrome.
Knox DL: Progressive subretinal fibrosis 1984; 102:1776–1784. Ophthalmology 1996; 103:1100–1105.
and uveitis. Br J Ophthalmol 1984; 6. Cantrill HL, Folk JC: Multifocal choroiditis 11. Kaiser PK, Gragoudas, ES: The subretinal
68:667–673. associated with progressive subretinal fibrosis and uveitis syndrome. Int
2. Palestine AG, Nussenblatt RB, Chan CC, fibrosis. Am J Ophthalmol 1986; Ophthalmol Clinics 1996; 36:145–152.
et al: Histopathology of the subretinal 101:170–180. 12. Wang RC, Zamir E, Dugel PU, et al:
fibrosis and uveitis syndrome. 7. Doran RML, Hamilton AM: Disciform Progressive subretinal fibrosis and
Ophthalmology 1985; 92:838–844. macular degeneration in young adults. blindness associated with multifocal
3. Kim MK, Chan CC, Belfort R, et al: Trans Ophthalmol Soc UK 1982; granulomatous chorioretinitis.
Histopathologic and immunohistopathologic 102:471–480. Ophthalmology 2002; 109:1527–1531.
features of subretinal fibrosis and uveitis 8. Morgan C, Schatz H: Recurrent multifocal 13. Lim W, Chee S, Sng I, et al:
syndrome. Am J Ophthalmol 1987; choroiditis. Ophthalmology 1986; Immunopathology of progressive subretinal
104:15–23. 93:1138–1143. fibrosis: a variant of sympathetic
4. Gass JDM, Margo CE, Levy MH: 9. Nozik RA, Dorsch W: A new ophthalmia. Am J Ophthalmol 2004;
Progressive subretinal fibrosis and chorioretinopathy associated with anterior 138:475–477.
blindness in patients with multifocal uveitis. Am J Ophthalmol 1973;
granulomatous chorioretinitis. Am J 76:758–762.
Ophthalmol 1996; 122:76–85. 10. Brown J, Folk JC, Reddy, CV, et al: Visual
5. Dreyer RF, Gass JDM: Multifocal choroiditis prognosis of multifocal choroiditis,
and panuveitis: a syndrome that mimics punctuate inner choroidopathy, and the

2134
CHAPTER

167 Diffuse Unilateral Subacute Neuroretinitis


J. Michael Jumper, H. Richard McDonald, Robert N. Johnson, Arthur D. Fu, and
Everett Ai

INTRODUCTION In the nearly three decades since the original description,


dozens of papers have been written to identify the likely
As he did with many other retinal inflammatory diseases, pathogens, report cases in other regions of the world and
J Donald M Gass meticulously investigated and described the describe the findings of newer diagnostic tests performed
condition he would eventually name diffuse unilateral sub- on patients with DUSN. However, the observations of
acute neuroretinitis (DUSN). For 15 years prior to the ground- Dr Gass and his colleagues have withstood the test of time.
breaking paper in 1978, he and his colleagues had recognized a
“unilateral retinal wipe-out syndrome” in which healthy young EPIDEMIOLOGY
individuals developed: “(1) insidious, usually severe loss of
peripheral and central vision; (2) vitritis; (3) diffuse and focal There have been over 200 cases of DUSN published in the
pigment epithelial derangement with relative sparing of the literature to date, in the form of six case series each containing
macula; (4) narrowing of the retinal vessels; (5) optic atrophy; 4–78 patients with the rest being case reports of one to three
(6) increased retinal circulation time; and (7) subnormal patients (Table 167.1). DUSN is a rare condition and incidence
electroretinographic findings.”1 Further observation led to the data is not available. It has been described to occur in the
discovery of the earlier findings including: “mild optic nerve Southeast,2,6 upper Midwest,5,8 Northeast9,10 and Northwest11,12
head edema, multifocal areas of active chorioretinitis and United States. Other countries with reported cases include
occasionally, iridocyclitis.”1 Later that year, Dr Gass and Canada,13 France,14 Scotland,15 Switzerland,16 Germany,17,18
colleagues described 12 additional cases of DUSN, two in Senegal,19 Liberia,20 India,21 and China.22 de Souza and
whom a motile subretinal nematode was observed.2 Over the coworkers from Brazil were the first to report cases outside of
next 5 years, he further studied his and other reported cases3–6 the United States or Caribbean Islands.23 Recently, large series
which led to the hypothesis that at: have been published from Venezuela and Brazil.24–26
(1) at least two different nematodes can cause this Meta-analysis of the peer-reviewed literature (209 patients,
syndrome 216 eyes) reveals an average age at diagnosis of 17.5 years (range
(2) there are endemic geographic regions for the different sized 2–84 years) with slight male predominance (60%). Sixty-six
nematodes and percent of patients are Latino, 27% are Caucasian, 5% are Black
(3) laser photocoagulation of the worm can halt disease and 2% are Asian. In 53% of cases, the right eye is involved.
progression.7 Bilateral disease has been reported in 7 patients (4%).2,24,27,28

TABLE 167.1. Meta-Analysis of Diffuse Unilateral Subacute Neuroretinitis

Author No. of Patients M:F Ratio Average Age Worm


(No. of Eyes) (Range) Identified

Gass et al2 37 (38) 22:15 16 (8–41) 2/38


7
Gass and Braunstein 10 6:4 25.5 (13–65) 10/10
Gass et al45 4 2:2 10.5 (4–16) 1/4
24
Cortez et al 78 (82) 45:33 16.7 (7–64) 33/82
25
Souza et al 12 8:4 15.4 (7–36) 4/12
de Garcia et al26 22 12:10 15.6 (5–37) 22/22
Garcia et al43 4 4:0 14.2 (9–23) 4/4
Case reports* 42 (44) 26:16 23.9 (2–84) 38/44

Total 209 (216) 125:84 17.5 years (2–84) 114/216 (52%)


*References 3–6, 8–13, 16, 17, 19, 20, 22, 23, 27, 28, 30–32, 35, 37, 38, 42, 44, 46–49.
2135
RETINA AND VITREOUS

TABLE 167.2. Clinical Features of Diffuse Unilateral Subacute


Neuroretinitis

Early Stage
Vision loss
Afferent pupillary defect
Anterior chamber cells
Hypopyon (rare)
Vitritis
Optic disk swelling
Optic atrophy (rare)
Retinal arterial narrowing
Intraretinal perivascular exudation
Multifocal evanescent gray-white outer retinal lesions
SECTION 10

Localized serous retinal detachment (rare)


Focal intraretinal hemorrhage (rare)
Choroidal neovascularization (rare)
Subretinal or intraretinal tracks FIGURE 167.1. Outer retinal whitening, retinal vasculitis and arteriolar
Motile worm: preretinal (rare), intraretinal, or subretinal narrowing in a patient with early stage DUSN.

Late Stage
Severe vision loss from mild to severe is always present. Half of patients have mild
Central scotoma optic disk swelling. Optic atrophy has also been described in
the early stage of DUSN.
Peripheral visual field abnormalities
Early fundus features include retinal arteriole narrowing,
Relative afferent pupillary defect intraretinal perivascular exudation, pigment epithelial depig-
Optic atrophy mentation and recurrent, multifocal evanescent gray-white
lesions in the outer retina (Fig. 167.1). Lesions vary in size
Retinal vascular attenuation with or without sheathing from 100 to 1200 mm. The retinitis is typically found in one
Retinal pigment epithelial derangement sector of the fundus and has been attributed to an inflammatory
reaction against byproducts of the worm. These lesions can
Subretinal macular or peripapillary mass from choroidal
neovascularization (rare)
provide a clue as to the worm’s location. The retinitis resolves
in 7–10 days with minimal or no residual retinal changes. The
presence and amount of retinal whitening is dependent on
CLINICAL FEATURES the activity of the organism. Localized serous retinal detach-
ment, choroidal neovascularization, cystoid macular edema
DUSN typically affects children or young adults (see and focal intraretinal hemorrhages have been described but are
Table 167.2). Usually, there are no associated systemic symptoms less common.2 Progressive arteriolar narrowing, optic atrophy,
although cutaneous and neural larva migrans have been vitreous degeneration and pigment epithelial changes (that
described in a few patients.8,29 The vision loss, while profound, can simulate multifocal choroiditis or an inherited retinal
is often found incidentally due to the insidious nature of the degeneration) follow in the subacute phase.
disease. In Gass and associates’ original series, half of patients A motile intraretinal, subretinal and, in rare cases, intra-
are unaware of vision loss until the late stages of the disease.2 vitreal worm has been described in 52% of all cases in the
Of symptomatic patients, the presenting complaint is often literature (Fig. 167.2). These are commonly of two different
vision loss, central or paracentral scotoma.2 Ocular discomfort lengths depending on the region of the world the disease is
is a less common complaint. The occurrence of symptoms have contracted. In the Southeastern United States, Caribbean,
been reported to range from 1 day to 4 years prior to Africa, and South America, the worm is usually 400–1000 mm
presentation.3,26 The clinical features of DUSN manifest as in length whereas longer (1500–2000 mm) organisms are found
early and late stages. in the upper Midwest United States, Canada, China, and
Europe. The diameter of the worm typically measures one-
twentieth of its length but an organism measuring 550 mm by
EARLY STAGE 150 mm has been described.12 It is usually nonsegmented,
Visual acuity in the early stage of DUSN ranges from 20/20 to white, glistening and tapered at both ends. Exposure to the
hand motions, the majority being 20/200 or worse. The vision examination light can cause movement in which the worm is
loss is often out of proportion to the visible changes of the optic propelled by coiling and uncoiling or wriggling in a snake-like
nerve or retina. An afferent pupillary defect was present in 36 of manner. The worm often disappears and reappears over time.
37 patients in Gass’ original series, regardless of stage.2 More than one worm has been identified in the same eye.24
In the early stage of DUSN, the external and slit-lamp eye Living organisms have been identified in the eye several years
examination is often normal. Ciliary flush, keratic precipitates, after onset of symptoms and a thorough search for a worm
anterior chamber cell and flare have been described. Two should be carried out on all patients. Occasionally, the worm is
2136 patients presented with hypopyon.2,30 Vitreous inflammation only identified with fundus photography.
Diffuse Unilateral Subacute Neuroretinitis

a b c

FIGURE 167.2. The 42-year-old Caucasian man from the upper Midwest United States with a 1 month history of blurred left vision (20/100)
presented with (a) mild vitritis, deep retinal gray-white lesions and a motile, subretinal worm measuring 1000 mm in length located inferotemporal
to the fovea (black arrow). (b) Fluorescein angiography revealed multiple hyperfluorescent spots corresponding to areas the worm was seen

CHAPTER 167
migrating. (c) The patient was treated with argon laser photocoagulation (white arrow). Vision returned to 20/30 and remained stable.
Photographs courtesy of George A Williams, MD.

LATE STAGE
In the late or inactive stage of DUSN, vision is typically 20/200
or worse with a dense central scotoma. Visual field deficits can
include the periphery. Optic atrophy and vascular attenuation
(with or without sheathing) are prominent features (Fig. 167.3).
There can be little correlation between the optic nerve changes
and vision such that some patients with severe atrophic
changes can maintain good vision and vice versa. The severity
of arterial and optic nerve changes are usually consistent with
one another. Vascular changes and pigment epithelial alteration
can vary between different quadrants of an eye. While pigment
epithelial disruption is a common feature of DUSN, pigment
migration into the retina (bone spicules) is less common and
not widespread when present. Greater pigment epithelial
alteration has been attributed to the larger worm. A subretinal
mass associated with choroidal neovascularization has been
described in the macula and around the optic nerve.2

DIAGNOSTIC FEATURES FIGURE 167.3. Optic atrophy and vascular narrowing in a patient
with 3/200 visual acuity due to late stage DUSN.
ANGIOGRAPHY
Fluorescein angiography in the early stage of DUSN reveals
normal retinal and choroidal blood flow (see Table 167.3). Optic Table 167.3. Angiographic Features of Diffuse Unilateral
disk leakage is often present in eyes with nerve swelling on Subacute Neuroretinitis
clinical examination. Peripheral retinal vascular changes are Normal retinal and choroidal vascular filling times in the early stage
rarely seen in patients with active retinitis. Cystoid macular
Delayed retinal circulation time in the late stage
edema has been described. The focal grey-white retinal lesions
are hypofluorescent in the early phase of the angiogram and Optic disk leakage
stain later in the course of the study. Pigment epithelial Early blockage and late staining of focal retinitis when present
alterations are visible as window defects but may be subtle
and detected only when comparing the diseased to normal eye Window defect from pigment epithelial alterations
(Fig. 167.4). Cystoid macular edema (rare)
In the late stage of DUSN, retinal circulation times may be
delayed. The hyperfluorescence from pigment epithelial atrophy
is most prominent around the optic nerve and in the
midperipheral retina. The macula usually only demonstrates the study. There was also ill-defined hyperfluorescence in the
mottled hyperfluorescence except in cases of disciform scar macular area in both phases of the disease. The authors suggest
formation. that choroidal infiltration was the cause of the angiographic
Serial indocyanine green (ICG) angiographic findings have pattern.
been described in one patient with DUSN in which a worm was
identified.31 Findings in both the early and late stage of the
disease include hypofluorescence of the clinically visible lesions ELECTROPHYSIOLOGY
in the early and intermediate phases of the study with per- Electroretinographic (ERG) abnormalities of only the affected
sistent changes of only a few of the lesions in the late frames of eye were present in all but one patient in Gass and Scelfo’s 2137
RETINA AND VITREOUS

FIGURE 167.4. Color fundus photograph


(a) and fluorescein angiogram (b) of retinal
pigment epithelial changes due to subretinal
movement of a worm in a case of DUSN.

a b

original series.1 The unaffected eye is normal in all electro- In the late stages of the disease when pigment epithelial
physiologic parameters. In the affected eye, both rod and cone derangement, vascular narrowing and optic atrophy are the
function is typically abnormal, with a reduction of the b:a-wave predominant features of the disease, the differential diagnosis
ratio, suggestive of proportionately inner retinal injury. includes multifocal choroiditis with panuveitis, central or
SECTION 10

Electrooculography was noted as subnormal in 16 of 29 in the ophthalmic artery obstruction, optic neuropathy from a com-
original series. pressive orbital lesion, Behçet’s disease, large cell lymphoma,
Electrophysiologic deterioration is progressive as suggested inactive acute retinal necrosis, atypical retinitis pigmentosa,
by a case report of a 9-year-old Caucasian boy with serial retained ferrous-containing metallic foreign body or post-
electrophysiologic testing over 4 years.32 In this patient (who traumatic chorioretinopathy. Careful history and serial
had poor vision at presentation), pattern visual-evoked potential ophthalmic examinations will help to differentiate the
and pattern ERG, indicators of visual pathways and macular conditions listed above from DUSN.
photoreceptor function, respectively, both progressed from
nearly extinguished to undetectable over a year. Full-field ERG PATHOPHYSIOLOGY
demonstrated progressive inner retinal dysfunction as suggested
by reduced b-wave amplitudes and additional photoreceptor It is accepted that DUSN is a form of ocular larval migrans and
involvement, but, in keeping with Gass and associates’ findings, that vision loss is associated with the parasite movement
never became completely extinguished.32 through and under the retina. The clinical and electrophy-
Multifocal electroretinography (mfERG) was described in one siology data suggest damage to both the retina and optic
patient in early stage DUSN with good vision.9 The decreased nerve.32 Proposed mechanisms of vision loss include: (1) direct
foveal response density as well as the increased parafoveal and toxic effects of the worm’s excretory–secretory proteins or
perifoveal waveform amplitudes fully recovered after successful (2) a result of the host inflammatory reaction to the parasite
laser ablation of the parasite. (3) mechanical damage caused by movement of the worm or
(4) an autoimmune reaction somehow initiated by the
infection.2,7,32
OTHER STUDIES The toxic effect of the worm has been described as both local
There has been no consistently positive serum, urine or stool and diffuse. The local effects are manifest in the focal, evane-
assays in patients with DUSN. In an analysis by Gass and scent retinal lesions that seem to coincide with the worm’s
coworkers, serum antibody titers to Toxocara canis by enzyme- migration while the diffuse effects are manifest in the rapid
linked immunosorbent assay (ELISA) was present in five of 14 vision loss and global retinal and optic nerve dysfunction.7
eyes in which the smaller worm was identified.7 None of these While no specific causative toxin has been identified in DUSN,
titers met the Centers for Disease Control definition of a eosinophil-derived neurotoxin and major basic protein have
positive test. Eosinophilia is rare in DUSN. been implicated in neural larval migrans with Baylisascaris
Scanning laser ophthalmoscopy (SLO) has been used to study procyonis.33
four eyes of three patients with DUSN.28 The authors used the The exact identity of the parasite in most cases is not known.
argon blue (488 nm wavelength) laser to provide high contrast Multicellular parasites exist in two distinct phyla: nemahel-
for identification of the worm and helium–neon (633 nm) laser minthes (roundworms or nematodes) and platyhelminthes
energy to perform perimetry and identify scotoma associated (flatworms, containing classes trematoda and cestoda). A large
with damage associated with the worm. They suggest that SLO number of helminthes have the potential to cause ocular
may be more sensitive and better tolerated than clinical larval migrans. Species implicated in DUSN include several
examination or fundus photography in detecting the often nematodes: Toxocara canis, Baylisascaris procyonis, Ancylostoma
elusive parasite.28 caninum, Ascaris lumbricoides and Dirofilaria spp.8,29,34–38 One
trematode, Alaria mesocercaria, has been identified as causing
DIFFERENTIAL DIAGNOSIS DUSN.12
At least two distinct worms are responsible for DUSN.
The differential diagnosis for DUSN is broad and varies Investigators have speculated that the smaller (400–1000 mm)
depending on the stage at presentation. In the early stage of worm found in Southeast US, the Caribbeans, and South
the disease, optic nerve swelling and vitritis may mimic America is Toxocara canis or Ancylostoma caninum. While
intermediate uveitis or optic neuritis. The outer retinal lesions serologic evidence supports the diagonis of T. canis in some
can be mistaken for multiple evanescent white dot syndrome, cases, the majority do not have positive serology, eosinophilia or
acute posterior multifocal placoid pigment epitheliopathy, granuloma characteristic of toxocariasis.6,7,37
multifocal choroiditis and panuveitis, serpiginous choroiditis, Baylisascaris procyonis has been implicated in DUSN due to
multifocal outer retinal toxoplasmosis, sarcoidosis, syphilis, the larger (1500–2000 mm) nematode found in the upper
2138 Behçet’s disease or large-cell lymphoma. Midwest US, Canada, China and Europe. Investigators have
Diffuse Unilateral Subacute Neuroretinitis

found retinal lesions consistent with DUSN in animal models successful in a case when treatment of the entire worm would
experimentally infected with B. procyonis eggs.39,40 Goldberg jeopardize the fovea (Fig. 167.5).21
and coworkers describe a case of DUSN in which the patient Medical therapy for DUSN has produced mixed results.
had serologic evidence of Baylisascaris exposure and necropsy Rubin was the first to report using a combination of oral
of several raccoons around the patient’s home revealed adult prednisone (40 mg/day for 3 weeks) and thiabendazole (2 g/day
B. procyonis in the intestines and infective eggs in the feces of for 5 days) in a 31 year-old woman with early stage DUSN and
8/12 (67%) animals tested. Concurrent ocular and neural larval progressive vision loss.6 Three months after treatment the
migrans have been described in two children with a history of vision improved from 20/200 to 20/60 and there was reduction
pica living in an area endemic for raccoons. Both had high of scotoma size and intraocular inflammation. Subsequently,
cerebrospinal fluid and serum titers to B. procyonis. Gass and Braunstein reported patients treated with thiaben-
The parasite has been successfully removed via eye wall dazole (with or without steroids) or diethylcarbamazine citrate
resection41 and vitrectomy12,35,38 in a total of four patients. in which the worm remained viable. They postulated that the
Identification of surgically removed specimens has proven chal- drug could not cross the blood–retinal barrier at larvacidal
lenging. Gass described features of Ancylostoma caninum but concentrations. A later study by Gass and coworkers suggest
exact identification could not be made. de Souza and coworkers that thiabendazole could be effective in some patients,
described the findings of a 630 µ 30 mm worm with morphologic especially those with moderate or severe vitritis.45 One patient
features most consistent with T. canis but because of poor treated with ivermectin had continued nematode movement
fixation, the definite identification remains uncertain.35 Others and subsequently required laser photocoagulation.42

CHAPTER 167
who have reviewed photomicrographs of this case have Recently, success has been achieved with a 1 month course
suggested the organism is Ancylostoma.29 A larger worm of high dose (400 mg/day) albendazole (without corticosteroids)
measuring 4500 mm was lost in transport to the parasitology in patients with the small worm variant of DUSN.25 The
lab.38 McDonald and coworkers removed and identified a authors report improved inflammation, stable or improved
pathogen as Alaria mesocercaria.12 This is clearly not the cause vision and resolution of worm motility. Cortez and colleagues
of the majority of DUSN cases as the shape (550 µ 190 mm) and treated six patients in which a motile worm was identified
movement differs greatly. with a 10-day course of albendazole (600 mg/day). The worm
remained viable in half (three) of the patients, necessitating
TREATMENT laser.

Laser photocoagulation of the motile worm has been shown to CONCLUSION


halt disease progression in DUSN.5,7,10,12,17,20,23,42–44 In 1978,
Raymond and coworkers were the first to report the use of DUSN represents a form of ocular larva migrans that is likely
xenon (one patient) and argon (one patient) laser photo- caused by many different helminthes and leads to global retinal
coagulation to destroy a motile worm. Vision improvement and and optic nerve dysfunction. Early recognition of the disease is
resolution of inflammation was noted in both patients.5 essential to maintaining, and in some cases, improving vision.
Improved visual acuity with laser photocoagulation has since A detailed search for the motile larvae (including serial
been confirmed in early stage DUSN. Garcia reported four biomicroscopy and color fundus photography) should be carried
cases treated with laser relatively early in the course of the out in all stages of the DUSN as destruction of the worm can
disease. The two patients treated within a month of symptom halt disease progression. While most patients are otherwise
onset had the best outcomes.43 In a large series of patients healthy, cutaneous, visceral or neural larva migrans may occur.
with late stage DUSN, vision stabilization was achieved in 19 Laser photocoagulation should be considered as first-line
of 22 patients, with improvement in only one.26 There have therapy in patients in whom the motile larvae are identified,
been no reported complications of laser for DUSN. While provided treatment will spare the central macula. For the 50%
confluent treatment of the larvae has most often been of patients in whom a worm cannot be found, a month-long
employed, treatment of the advancing end of the worm has been course of albendazole should be considered.

a b c

FIGURE 167.5. The 29-year-old Caucasian man from Northern California with a 3 week history of right transient visual obscurations. Initial vision
was 20/20 in the right eye with optic nerve swelling and a trace afferent pupillary defect. Serial examination over 6 weeks led to the discovery of
a (a) motile subretinal worm within the macula (magnified in inset). (b) Examination after initial thermal laser phototcoagulation (white arrow)
revealed the worm had moved superotemporal to the fovea. A second session of laser (black arrow) was performed. The motile tail end of the
worm was not treated due to proximity to the fovea. (c) After destruction of the worm, the vision stabilized at 20/25. 2139
RETINA AND VITREOUS

REFERENCES
1. Gass JD, Scelfo R: Diffuse unilateral 17. Kuchle M, Knorr HL, Medenblik-Frysch S, 33. Moertel CL, Kazacos KR, Butterfield JH,
subacute neuroretinitis. J R Soc Med 1978; et al: Diffuse unilateral subacute et al: Eosinophil-associated inflammation
71:95–111. neuroretinitis syndrome in a German most and elaboration of eosinophil-derived
2. Gass JD, Gilbert WR Jr, Guerry RK, likely caused by the raccoon roundworm, proteins in 2 children with raccoon
Scelfo R: Diffuse unilateral subacute Baylisascaris procyonis. Graefes Arch Clin roundworm (Baylisascaris procyonis)
neuroretinitis. Ophthalmology 1978; Exp Ophthalmol 1993; 231:48–51. encephalitis. Pediatrics 2001; 108:E93.
85:521–545. 18. Meyer-Riemann W, Petersen J, Vogel M: 34. Ashton N: Larval granulomatosis of the
3. PARSONS HE: Nematode chorioretinitis; An attempt to extract an intraretinal retina due to Toxocara. Br J Ophthalmol
report of a case, with photographs of a nematode located in the papillomacular 1960; 44:129–148.
viable worm. AMA Arch Ophthalmol 1952; bundle. Klin Monatsbl Augenheilkd 1999; 35. de Souza EC, Nakashima Y: Diffuse
47:799–800. 214:116–119. unilateral subacute neuroretinitis. Report of
4. Price JA Jr, Wadsworth JA: An intraretinal 19. Rasquin F, Waterschoot MP, Termote H, transvitreal surgical removal of a subretinal
worm. Report of a case of macular Carlier Y: Diffuse unilateral subacute nematode. Ophthalmology 1995;
retinopathy caused by invasion of the retina neuroretinitis in Africa. Ocul Immunol 102:1183–1186.
by a worm. Arch Ophthalmol 1970; Inflamm 2006; 14:59–62. 36. Kazacos KR, Vestre WA, Kazacos EA,
83:768–770. 20. Carney MD, Combs JL. Diffuse unilateral Raymond LA: Diffuse unilateral subacute
5. Raymond LA, Gutierrez Y, Strong LE, et al: subacute neuroretinitis. Br J Ophthalmol neuroretinitis syndrome: probable cause.
Living retinal nematode (filarial-like) 1991; 75:633–635. Arch Ophthalmol 1984; 102:967–968.
SECTION 10

destroyed with photocoagulation. 21. Myint K, Sahay R, Mon S, et al: ‘Worm in 37. Oppenheim S, Rogell G, Peyser R: Diffuse
Ophthalmology 1978; 85:944–949. the Eye’ – The Rationale for treatment of unilateral subacute neuroretinitis. Ann
6. Rubin ML, Kaufman HE, Tierney JP, DUSN in South India. Br J Ophthalmol 2006; Ophthalmol 1985; 17:336–338.
Lucas HC: An intraretinal nematode 90:1125–1127. 38. Yamamoto S, Hayashi M, takeuchi S:
(a case report). Trans Am Acad Ophthalmol 22. Cai J, Wei R, Zhu L, et al: Diffuse unilateral Surgically removed submacular nematode.
Otolaryngol 1968; 72:855–866. subacute neuroretinitis in China. Arch Br J Ophthalmol 1999; 83:1088.
7. Gass JD, Braunstein RA: Further Ophthalmol 2000; 118:721–722. 39. Akao N, Hayashi E, Sato H, et al: Diffuse
observations concerning the diffuse 23. de Souza EC, da Cunha SL, Gass JD. retinochoroiditis due to Baylisascaris
unilateral subacute neuroretinitis syndrome. Diffuse unilateral subacute neuroretinitis in procyonis in Monglian gerbils. J Parasitol
Arch Ophthalmol 1983; 101:1689–697. South America. Arch Ophthalmol 1992; 2003; 89:174–175.
8. Mets MB, Noble AG, Basti S, et al: Eye 110:1261–1263. 40. Kazacos KR, Raymond LA, Kazacos EA,
findings of diffuse unilateral subacute 24. Cortez R, Denny JP, Muci-Mendoza R, Vestre WA: The raccoon ascarid.
neuroretinitis and multiple choroidal et al: Diffuse unilateral subacute A probable cause of human ocular larva
infiltrates associated with neural larva neuroretinitis in Venezuela. Ophthalmology migrans. Ophthalmology 1985;
migrans due to Bbaylisascaris procyonis. 2005; 112:2110–2114. 92:1735–1744.
Am J Ophthalmol 2003; 135:888–890. 25. Souza EC, Casella AM, Nakashima Y, 41. Gass JDM: Stereoscopic atlas of macular
9. Martidis A, Greenberg PB, Rogers AH, Monteiro ML: Clinical features and diseases. 3rd edn. St Louis: CV Mosby;
et al: Multifocal electroretinography outcomes of patients with diffuse unilateral 1987:474–475.
response after laser photocoagulation of a subacute neuroretinitis treated with oral 42. Casella AM, Farah ME, Belfort R Jr:
subretinal nematode. Am J Ophthalmol albendazole. Am J Ophthalmol 2005; Antihelminthic drugs in diffuse unilateral
2002; 133:417–419. 140:437–445. subacute neuroretinitis. Am J Ophthalmol
10. Sivalingam A, Goldberg RE, Augsburger J, 26. de Garcia CA, Gomes AH, Vianna RN, 1998; 125:109–111.
Frank P: Diffuse unilateral subacute et al: Late-stage Diffuse Unilateral 43. Garcia CA, Gomes AH, Garcia Filho CA,
neuroretinitis. Arch Ophthalmol 1991; Subacute Neuroretinitis: Photocoagulation Vianna RN: Early-stage diffuse unilateral
109:1028. of the Worm does not Improve the Visual subacute neuroretinitis: improvement of
11. Goldberg MA, Kazacos KR, Boyce WM, Acuity of Affected Patients. Int Ophthalmol vision after photocoagulation of the worm.
et al: Diffuse unilateral subacute 2006; 26:39–42. Eye 2004; 18:624–627.
neuroretinitis. Morphometric, serologic, and 27. de Souza EC, Abujamra S, Nakashima Y, 44. Matsumoto BT, Adelberg DA, Del Priore LV:
epidemiologic support for Baylisascaris as Gass JD: Diffuse bilateral subacute Transretinal membrane formation in diffuse
a causative agent. Ophthalmology 1993; neuroretinitis: first patient with documented unilateral subacute neuroretinitis. Retina
100:1695–1701. nematodes in both eyes. Arch Ophthalmol 1995; 15:146–149.
12. McDonald HR, Kazacos KR, Schatz H, 1999; 117:1349–1351. 45. Gass JD, Callanan DG, Bowman CB: Oral
Johnson RN: Two cases of intraocular 28. Moraes LR, Cialdini AP, Avila MP, therapy in diffuse unilateral subacute
infection with Alaria mesocercaria Elsner AE: Identifying live nematodes in neuroretinitis. Arch Ophthalmol 1992;
(Trematoda). Am J Ophthalmol 1994; diffuse unilateral subacute neuroretinitis by 110:675–680.
117:447–455. using the scanning laser ophthalmoscope. 46. Myint K, Sahay R, Mon S, et al: The Indian
13. Yuen VH, Chang TS, Hooper PL: Diffuse Arch Ophthalmol 2002; 120:135–138. case of live worm in Diffuse Unilateral
unilateral subacute neuroretinitis syndrome 29. Gass JDM: Stereoscopic atlas of macular Subacute Neuroretinitis. Eye 2006;
in Canada. Arch Ophthalmol 1996; diseases. 4th edn. St Louis: Mosby; 20:612–613.
114:1279–1282. 1997:622–628. 47. Cialdini AP, de Souza EC, Avila MP:
14. Salvanet-Bouccara A, Troussier H: Diffuse 30. Muccioli C, Belfort R Jr: Hypopyon in a The first South American case of diffuse
unilateral subacute neuroretinitis. Apropos patient with presumptive diffuse unilateral unilateral subacute neuroretinitis caused by
of a case. J Fr Ophtalmol 1987; subacute neuroretinitis. Ocul Immunol a large nematode. Arch Ophthalmol 1999;
10:667–672. Inflamm 2000; 8:119–121. 117:1431–1432.
15. Kinnear FB, Hay J, Dutton GN, Smith HV: 31. Vianna RN, Onofre G, Ecard V, et al: 48. Harto MA, Rodriguez-Salvador V, Avino JA,
Presumed ocular larva migrans presenting Indocyanine green angiography in diffuse et al: Diffuse unilateral subacute
with features of diffuse unilateral subacute unilateral subacute neuroretinitis. Eye 2006; neuroretinitis in Europe. Eur J Ophthalmol
neuroretinitis. Br J Ophthalmol 1995; 20:1113–1116 1999; 9:58–62.
79:1140–1141. 32. Audo I, Webster AR, Bird AC, et al: 49. Gass JD: Subretinal migration of a
16. Bernasconi OR, Piguet B. Unilateral diffuse Progressive retinal dysfunction in diffuse nematode in a patient with diffuse unilateral
subacute neuroretinitis. Klin Monatsbl unilateral subacute neuroretinitis. Br J subacute neuroretinitis. Arch Ophthalmol
Augenheilkd 1997; 210:327–328. Ophthalmol 2006; 90:793–4. 1996; 114:1526–1527.

2140
CHAPTER

168 Frosted Branch Angiitis


George N. Papaliodis, Carol M. Lee, and Henry J. Kaplan

Key Features with ‘frosted branch like appearance’ of the retinal vasculature
• Rare clinical entity without an established cause which must
in patients with lymphomas or leukemias, and similarly distinct
be distinguished from secondary frosted branch angiitis with
from patients with an associated viral or autoimmune disease
identifiable etiologies
in which the frosted branch appearance is a clinical sign of
• Bilateral retinal vasculitis with profound vascular sheathing
disease severity.72
found in young, otherwise healthy patients
• Treatment of choice (after excluding infectious etiologies) is DIAGNOSTIC TESTS
oral corticosteroids with improvement in vascular sheathing
and resultant visual acuity
Fluorescein angiography shows normal blood flow without
evidence of occlusion or stasis but with late staining and leakage
of dye from affected vessels (Fig. 168.2). Additional fundus
INTRODUCTION findings may include intraretinal hemorrhages, punctate hard
exudates, macular edema, and serous exudative detachments
Frosted branch angiitis was first described in 1976 by Ito and of the macula or periphery. The retina can appear diffusely
co-workers in a 6-year-old healthy boy with severe sheathing of thickened and edematous. It is postulated that these thick
all the retinal vessels, resembling the frosted branches of a tree.1 perivascular infiltrates may be a result of immune complex
Between 1976 and 2005 there have been 57 total cases2–9 deposition along the vessel walls.29,34,35
reported in the literature with some degree of uncertainty to the Electrophysiologic testing shows a reduction in the amplitude
absolute number, given the confusion in the literature regard- of the electroretinogram, which remains reduced even after
ing the use of the term ‘frosted branch angiitis’.2–29 The initial convalescence. In contrast, the visual-evoked response is initially
clinical presentation of this rare entity may be similar to that reduced but may return to normal.5,8 Visual-field testing reveals
of other more common retinal vasculitides,30–33 and the term concentric constriction or relative central defects that improve
‘frosted branch angiitis’ has been utilized to describe any con- after clinical resolution of the vasculitis.
dition which manifests with severe retinal vascular sheathing. Laboratory investigations have not revealed a single causative
It is unclear if this condition represents a distinct clinical agent in this disease. Moreover, as this entity must be dis-
syndrome or a sign of severe vasculitis/phlebitis.72 The term tinguished from pathologic processes which present as an
‘frosted branch angiitis’ has been used synonymously with exaggerated sheathing of retinal vessels, any condition which
‘diffuse acute retinal periphlebitis’.6 can cause a retinal vasculitis should be included in the differ-
ential diagnosis. Included among the normal laboratory studies
CLINICAL CHARACTERISTICS from case reports in the literature were: complete blood cell
count and differential; erythrocyte sedimentation rate; skin test
Acute frosted branch angiitis is most commonly a bilateral for tuberculin delayed-type hypersensitivity; determination of
retinal vasculitis seen in young, otherwise healthy patients in serum antibodies for syphilis, Lyme, toxoplasmosis, human
the age range of 3–36 years and present with a chief complaint immunodeficiency virus (HIV), herpes simplex, herpes zoster,
of decreased visual acuity. Unilateral cases have similarly been and cytomegalovirus (CMV); urine and blood cultures for
reported in the literature.14,15,20,29 Vision is usually profoundly both bacteria and virus; antinuclear antibody determination;
affected, with the majority of reported cases presenting with rheumatoid factor; serum protein electrophoresis; immuno-
20/200 vision or worse (range 20/20 to light perception). An electrophoresis; and determination of serum angiotensin-
antecedent viral illness has been reported in some cases. On converting enzyme levels. Chest radiograph films, lumbosacral
examination, both anterior chamber and vitreous inflammation X-ray films, cerebrospinal fluid examinations, and computed
are present in almost all cases without pars plana deposits. The tomographic scans or magnetic resonance imaging of the head
appearance of bilateral retinal phlebitis and arteritis extending and orbits have all been normal. One patient demonstrated an
from the posterior pole to the periphery with uninterrupted increased serum antistreptolysin O titer.6
severe sheathing of all the vessels, established the term ‘frosted
branch angiitis’ (Fig. 168.1). Prominent sheathing of the retinal MANAGEMENT RECOMMENDATIONS
veins is characteristic of this entity, but the retinal arteries may
similarly be involved (more commonly noted in younger Frosted branch angiitis responds to systemic steroids with a
patients). rapid resolution of the vascular sheathing, retinal hemorrhages,
Patients with frosted branch angiitis as described by Ito were and exudative neurosensory retinal detachment (Fig. 168.3).
young and otherwise healthy. This is in contrast to patients Systemic steroids (initial dose 80–100 mg oral prednisone in 2141
RETINA AND VITREOUS

a b
SECTION 10

c d

FIGURE 168.1. (a and b) The most frequent presentation of frosted branch angiitis is bilateral extensive venous sheathing, resembling the
frosted branches of a tree. There is mild pallor of the left optic disk. (c and d) Bilateral retinal phlebitis extends from the posterior pole to the
periphery with uninterrupted severe sheathing of all the vessels. Extensive perivenous exudates associated with intraretinal hemorrhage are
noted in this patient. (Left figures, right eye; right figures, left eye.)

FIGURE 168.2. The fluorescein angiogram


demonstrates normal venous flow in the early
phase (left), with diffuse staining of the vein
walls in the late phase (right).

adults for 10 days) should be initiated once treatable causes in better within 2–3 weeks after initiation of treatment. Others
the differential diagnosis are excluded (most notably infectious have reported complications prohibiting full visual recovery
etiologies which may worsen in the context of steroid therapy). including a fibrotic macular scar,6 and central retinal vein
The steroids should subsequently be tapered over several weeks. occlusion.19 Remote sequelae have included a horseshoe tear in
Funduscopic sequelae include attenuation of both the arteries one patient and multiple bilateral branch vein occlusions in
and the veins, the development of sharply demarcated atrophic another. It is not known whether these complications are
lesions in the periphery, and the deposition of yellow subretinal adverse sequelae of frosted branch angiitis or are fortuitous in
deposits in areas of previously detached retina. their occurrence.6
Recovery to normal vision usually occurs, with a range of The natural history of the disease without the use of oral
2142 20/15 to –20/40, the majority of cases improving to 20/20 or steroids is not known. One report has shown that the frosted
Frosted Branch Angiitis

FIGURE 168.3. Systemic steroids are associated with the rapid resolution of the vascular sheathing, intraretinal hemorrhages, and exudative

CHAPTER 168
neurosensory retinal detachments. A residual scar in the right macula (left figure) of this patient resulted in a permanent decrease in vision to
20/300.

branch angiitis resolved spontaneously without the use of oral one case.31 Another study reported a case of frosted branch
agents and with only topical steroid drops for an anterior uveitis angiitis due to a relapsing acute lymphoblastic leukemia involv-
over a course of 1 week without any permanent retinal sequelae.7 ing the central nervous system that resolved with appropriate
systemic chemotherapy and local radiotherapy.36 Case reports
DIFFERENTIAL DIAGNOSIS have similarly described secondary frosted branch angiitis in
patients with Behçet’s disease,10 herpes simplex virus (types 1
Although no specific causative agent has been identified in and 2),12,24 CMV retinitis,33–35 toxoplasmosis,13 Harada’s
frosted branch angiitis, treatable causes of retinal vasculitis disease,16 rapidly progressive glomerulonephritis,17 aseptic
should be excluded before the initiation of corticosteroid treat- meningitis,25 and Crohn’s disease.27
ment (Table 168.1). Conditions associated with a predominant The ocular manifestations of sarcoidosis occur in 15–33% of
periphlebitis include tuberculosis, Eales’ disease, sarcoidosis, cases.36–39 Posterior segment involvement is seen frequently
multiple sclerosis, and HIV infection, whereas systemic lupus (14–28%), usually in association with anterior segment disease.
erythematosus (SLE) and syphilis have been associated with a The perivascular sheathing of ocular sarcoid is seen in the
predominant inflammation of the arterial tree. Retinal vasculitis midperiphery, usually without vascular occlusion, and affects
has been reported as a consequence of Lyme borreliosis.30 A mainly the veins. Severe perivascular sheathing appears as
workup for a presumed diagnosis of frosted branch angiitis ‘candle wax drippings’. Acute periphlebitis may be accompanied
revealed ultimately a diagnosis of ocular large cell lymphoma in by intraretinal hemorrhages and edema. Occasionally, branch or
central retinal vein occlusion may occur, with subsequent
peripheral retinal or disk neovascularization.
TABLE 168.1 Differential Diagnosis of Frosted Branch Angiitis
The vitritis of sarcoidosis consists of clumps of vitreal inflam-
matory cells – ’snowballs’ or ‘strings of pearls’. These clumps of
Disease Laboratory Tests cells may remain adherent to the posterior vitreous face and
may appear as a sheet. Sarcoid granuloma can be seen in the
Sarcoidosis Chest radiograph film, serum calcium and
phosphorus levels, serum angiotensin deep retina or choroid or on the optic nerve head. Although
converting enzyme determination the diagnosis of sarcoid should be excluded by a chest CT scan
and determinations of serum angiotensin-converting enzyme
Multiple sclerosis Magnetic resonance imaging, cerebrospinal
and serum calcium, the associated ophthalmic findings should
fluid examination
help differentiate ocular sarcoidosis associated vasculitis from
Pars planitis frosted branch angiitis.
Eales’ disease Retinal venous sheathing is observed in 10–20% of patients
with multiple sclerosis40–45 and is seen either as active periph-
Tuberculosis Chest radiograph film, tuberculin skin test lebitis, with white patchy cuffs surrounding the blood vessel, or
Syphilis Venereal Disease Research Laboratory test, as venous sclerosis. Periphlebitis in multiple sclerosis is occa-
fluorescent treponemal antibody test sionally accompanied by pars planitis and frequently resolves
Lyme disease Lyme serology, ELISA, Western blot analysis spontaneously without visual symptoms or hemorrhages. Rarely,
severe periphlebitis can lead to peripheral retinal ischemia and
Systemic lupus Antinuclear antibody, anti DNA determinations neovascularization. The perivascular infiltrates have been ident-
erythematosus
ified histopathologically to be composed of segmental lympho-
AIDS HIV antibody determination plasmacytic infiltrates within and around the retinal veins.40,44
Bone marrow Complete blood count with differential, The diagnosis of multiple sclerosis can be made readily by
tumefaction vitreous biopsy, cerebrospinal fluid magnetic resonance imaging or cerebrospinal fluid examination
examination, magnetic resonance imaging with oligoclonal banding. Additionally, the fluffy perivenous
Abbreviations: ELISA, enzyme linked immunosorbent assay; AIDS, acquired
cuffs in multiple sclerosis are quite small, focal, and inter-
immunodeficiency syndrome; HIV, human immunodeficiency virus. rupted, in contrast to the widespread sheathing of the vessels in
frosted branch angiitis. 2143
RETINA AND VITREOUS

Retinal periphlebitis can be found in association with they also appeared occluded and extremely attenuated. This
peripheral uveitis or pars planitis.46–48 Snowbanks consisting of obliterative feature was confirmed on fluorescein angio-
fibroglial proliferation and vascular exudation are seen over a graphy.30,57 In the laboratory workup, patients seronegative for
broad area of the inferior peripheral retina; retinal edema, Lyme disease may have positive results with the more sensitive
premacular fibroplasia, and cystoid macular edema are also Western blot analysis.57 A careful history should be taken to
observed. The peripheral terminal branches of the retinal veins identify any exposure to ticks or to reveal any systemic com-
are cuffed with white inflammatory material, differentiating plaints. The presence of associated systemic findings, in areas
this entity from the diffuse periphlebitis of frosted branch endemic for Lyme disease, and the more occlusive nature of the
angiitis. periphlebitis should help in differentiating this from frosted
Tuberculosis commonly produces focal perivenous sheathing branch angiitis.
in the peripheral venules, which only occasionally involves Retinal vasculitis is a common ophthalmic manifestation of
the central retinal vein.49,50 Eales’ disease, which has been SLE. Although the most common findings are cotton-wool
associated with tuberculosis,51 characteristically produces early spots with or without intraretinal hemorrhages, almost 30% of
capillary closure in the periphery of multiple quadrants of the patients with SLE will have retinal vasculitis with micro-
retina, with sheathing of the peripheral retinal veins and intra- angiopathic small-vessel occlusion most often affecting the
retinal hemorrhages. Neovascularization of the retinal periphery arterioles.62 Occasionally, severe retinal occlusive disease can
results from the ensuing ischemia, usually at the clearly defined occur, affecting both the arteries and the veins in association
border between perfused and nonperfused retina. The charac- with anticardiolipin antibodies,63 with subsequent peripheral
SECTION 10

teristic picture of extensive peripheral capillary nonperfusion in retinal neovascularization. Retinal arterial inflammation is also
Eales’ disease and the focal perivenular sheathing in tuber- seen in segmental periarteritis of the retina, in which whitish
culosis should help differentiate these entities from frosted plaques are scattered along the main arterial branches.
branch angiitis. Surprisingly, fluorescein leakage is seen from the venous but not
The vasculitis of ocular syphilis is generally a periarteritis from the arterial wall.64 It can be distinguished from frosted
with arteriolar sheathing, exudates, and intraretinal and pre- branch angiitis by the generally more limited involvement of
retinal hemorrhages,52–55 although isolated venous periphlebitis the retinal vascular tree.
has also been described.55 The arteriolitis may become occlu- The perivasculitis in acquired immunodeficiency syndrome
sive, with eventual sclerosis of the involved arterioles and the (AIDS) retinopathy should also be included in the differential
development of peripheral retinal neovascularization. Other diagnosis of frosted branch angiitis.65–69 Diffuse perivasculitis of
more common manifestations of posterior syphilis include the far-peripheral veins and focal periarteritis can be seen in
neuroretinitis, chorioretinitis, and papillitis. The course of ocular patients with AIDS or AIDS-related complex67,68,70 without
syphilis has been noted to be more aggressive in patients with concomitant infectious retinopathy. These conditions are more
associated HIV disease, and as such it is recommended that commonly seen, however, with an associated infectious com-
patients being evaluated or treated for ocular syphilis be tested ponent such as CMV retinopathy (Fig. 168.4).33,35,66 Frosted
for HIV infection.53,56 Because ocular syphilis can mimic many branch angiitis has been described in AIDS patients accompany-
diseases, patients with a vasculitis resembling frosted branch ing small areas of CMV retinopathy. Classically, CMV lesions
angiitis should have antibody tests for syphilis (the Venereal appear as creamy yellow-white retinal opacifications generally
Disease Research Laboratory test and the fluorescent trepo- following the vessel walls and often accompanied by variable
nemal antibody test). amounts of intraretinal hemorrhage. The vessel walls may be
Retinal periphlebitis has also been described in patients with narrowed, sheathed, or occluded.60 In these patients, treatment
Lyme borreliosis.30,57 Lyme disease is a tick-borne infection that for CMV infection provided resolution of the vasculitic com-
causes a constellation of systemic complaints and manifes- ponent of their retinopathy. The perivascular infiltrates seen in
tations usually within the dermatologic, rheumatologic, cardiac, CMV-related frosted branch angiitis may be due to direct
and central nervous systems.30,57–61 Ocular manifestations are infection by the CMV of the retinal vessel walls.34 However, this
generally uncommon, and these usually present in patients infiltrate may also be a result of immune complex deposition
with either established systemic disease or systemic complaints within the vascular wall similar to that hypothesized in
referable to Lyme disease. The most frequent signs include conventional cases of frosted branch angiitis.
conjunctivitis, optic neuritis and neuropathy, papilledema, and A case of endogenous Fusarium endophthalmitis diagnosed
interstitial keratitis.58–61 In the reported cases of retinal vascu- by vitrectomy was reported in an intravenous drug user who
litis with Lyme disease, the arteries and veins were sheathed but initially presented with frosted branch angiitis and vitritis.71 In

a b c

FIGURE 168.4. (a) Severe sheathing of the vessels is present adjacent to a patch of CMV retinitis along the superotemporal arcade of the left
eye. (b) This sheathing extends to the periphery, giving a frosted branch appearance. (c) Two weeks after induction with antiviral medications,
2144 the sheathing is resolving.
Frosted Branch Angiitis

this case, the presence of worsening vitritis and the social be established by a vitreous biopsy if there is no obvious
history were helpful in obtaining a correct diagnosis. systemic evidence of disease.
Secondary vasculitis with frosted branch appearance has been
described in patients with ocular toxoplasmosis.13,14,21 The CONCLUSIONS
vasculitis from toxoplasmosis is more commonly an arteritis in
the vicinity of the active focus of retinitis. In summary, frosted branch angiitis is a bilateral inflammation,
Finally, bone marrow tumefactions such as leukemia and in healthy young individuals, of the retinal arteries and veins,
non-Hodgkin’s lymphoma (i.e., reticulum cell sarcoma) should with the veins being more severely affected. Unilateral cases
be considered in the presence of diffuse retinal vascular have been also described. No cause has been identified which is
sheathing.31,32 Intraretinal hemorrhages and cotton-wool spots distinctly different from patients who have a frosted branch
are more frequently seen in leukemia, whereas retinal cell like appearance from underlying infectious or autoimmune
sarcoma can present as a diffuse vitritis, often with accompany- mediated diseases. Vision is profoundly diminished in the acute
ing retinal pigment epithelial abnormalities such as multifocal, phase, with dramatic improvement after oral corticosteroid
yellow, subretinal infiltrates or pigment epithelial detachments. administration. The differential diagnosis is extensive, but
Underlying the vitreal inflammation may be a diffuse retinal frosted branch angiitis can frequently be distinguished by its
vasculitis resembling frosted branch angiitis. The diagnosis can unique clinical presentation and course.

REFERENCES

CHAPTER 168
1. Ito Y, Nakano M, Kyu N, et al: Frosted 16. Matsuo T, Kanata N, Itami M: Frosted 30. Smith JL, Winward KE, Nicholson DF, et al:
branch angiitis in a child. Jpn J Clin branch angiitis associated with Harada Retinal vasculitis in Lyme borreliosis. J Clin
Ophthalmol 1976; 30:797. disease-like manifestations recurs Neuroophthalmol 1991; 11:7.
2. Sakanishi Y, Kanagami S, Ohara K: Frosted 10 years later. Jpn J Ophthalmol 31. Ridley ME, McDonald HR, Sternberg P, et al:
retinal angiitis in children. Jpn J Clin 2002; 46:682. Retinal manifestations of ocular lymphoma
Ophthalmol 1984; 38:803. 17. Gupta A, Narang S, Gupta V, et al: Frosted (reticulum cell sarcoma). Ophthalmology
3. Yamane S, Nishiuchi T, Nakagawa Y, et al: branch angiitis associated with rapidly 1992; 99:1153.
A case of frosted branch angiitis of the progressive glomerulonephritis. Indian J 32. Kim TS, Duker JS, Hedges TR: Retinal
retina. Folia Ophthalmol Jpn 1985; Ophthalmol 2002; 50:317. angiopathy resembling frosted branch
36:1822. 18. Huerva V, Puig T, Sanchez MC, et al: A new angiitis in a patient with relapsing acute
4. Higuchi K, Maeda K, Uji T, et al: A case of case of acute idiopathic frosted branch lymphoblastic leukemia. Am J Ophthalmol
infantile uveitis with frosted branch angiitis. angiitis in Europe. Eur J Ophthalmol 2002; 1994; 117:806.
Jpn Rev Clin Ophthalmol 1985; 79:2660. 12:127. 33. Geier SA, Nasemann J, Klauss V, et al:
5. Watanabe Y, Takeda N, Adachi-Usami E: 19. Kaburaki T, Nakamura M, Nagasawa K, Frosted branch angiitis in a patient with the
A case of frosted branch angiitis. Br J et al: Two cases of frosted branch angiitis acquired immunodeficiency syndrome. Am
Ophthalmol 1987; 71:553. with central retinal vein occlusion. Jpn J J Ophthalmol 1992; 113:203.
6. Kleiner RC, Kaplan HJ, Shakin JL, et al: Ophthalmol 2001; 45:628. 34. Spaide RF, Vitale AT, Toth IR, et al: Frosted
Acute frosted retinal periphlebitis. Am J 20. Fine HF, Smith JA, Murante BL, et al: branch angiitis associated with
Ophthalmol 1988; 106:27. Frosted branch angiitis in a child with cytomegalovirus retinitis. Am J Ophthalmol
7. Vander JF, Masciulli L: Unilateral frosted HIV infection. Am J Ophthalmol 2001; 1992; 113:522.
branch angiitis [letter; comment]. Am J 131:394. 35. Rabb MF, Jampol LM, Fish RH, et al:
Ophthalmol 1991; 112:477. 21. Ysasaga JE, Davis J: Frosted branch Retinal periphlebitis in patients with
8. Hamed LM, Fang EN, Fanous MM, et al: angiitis with ocular toxoplasmosis. Arch acquired immunodeficiency syndrome with
Frosted branch angiitis: the role of Ophthalmol 1999; 117:1260. cytomegalovirus retinitis mimics acute
systemic corticosteroids. J Pediatr 22. Biswas J, Raizada S, Gopal L, et al: frosted retinal periphlebitis. Arch
Ophthalmol Strabismus 1992; 29:312. Bilateral frosted branch angiitis and Ophthalmol 1992; 110:1257.
9. Nakai A, Saika S: A case of frosted branch cytomegalovirus retinitis in acquired 36. Obenauf CD, Shaw HE, Sydnor CF, et al:
retinal angiitis in a child. Ann Ophthalmol immunodeficiency syndrome. Indian J Sarcoidosis and its ophthalmic
1992; 24:417. Ophthalmol 1999; 47:195. manifestations. Am J Ophthalmol 1978;
10. Reynders S, Dewachter A, de Vriese AS: 23. Luo G, Yang P, Huang S, et al: A case 86:648.
A case of secondary frosted branch angiitis report of frosted branch angiitis and its 37. Jabs DA, Johns CJ: Ocular involvement in
in Behçet’s disease. Bull Soc Belge visual electrophysiology. Doc Ophthalmol chronic sarcoidosis. Am J Ophthalmol
Ophthalmol 2005; 298:41. 1998–99; 97:135. 1986; 102:297.
11. Liu Y, Ma Z, Tso MO, Zhang Q: Frosted 24. Markomichelakis NN, Barampouti F, 38. Spalton DJ, Sanders MD: Fundus changes
branch angiitis secondary to macular Zafirakis P, et al: Retinal vasculitis with a in histologically confirmed sarcoidosis. Br J
choroidal neovascularization in a Chinese frosted branch angiitis-like response due to Ophthalmol 1981; 65:348.
woman. Jpn J Ophthalmol 2005; 49:228. herpes simplex virus type 2. Retina 1999; 39. Smith JA, Foster CS: Sarcoidosis and its
12. Shenoy R, Elagib EN, Al-Siyabi H: 19:455. ocular manifestations. Int Ophthalmol Clin
Frosted retinal branch angiitis in an 25. Johkura K, Hara A, Hattori T, et al: Frosted 1996; 1:109.
immunocompetent adult due to herpes branch angiitis associated with aseptic 40. Arnold AC, Pepose JS, Hepler RS, et al:
simplex virus. Indian J Ophthalmol 2001; meningitis. Eur J Neurol 2000; 7:241. Retinal periphlebitis and retinitis in multiple
49:56. 26. Seo MS, Woo JM, Jeong SK, et al: sclerosis. Ophthalmology 1984; 91:255.
13. Oh J, Huh K, Kim SW: Recurrent secondary Recurrent unilateral frosted branch angiitis. 41. Barnford CR, Ganley JP, Sibley WA, et al:
frosted branch angiitis after toxoplasmosis Jpn J Ophthalmol 1998; 42:56. Uveitis, perivenous sheathing and multiple
vasculitis. Acta Ophthalmol Scand 2005; 27. Sykes SO, Horton JC: Steroid responsive sclerosis. Neurology 1978; 28:119.
83:115. retinal vasculitis with a frosted branch 42. Rucker CW: Sheathing of the retinal veins
14. Diaz-Valle D, Diaz-Rodriguez E, Diaz-Valle T, appearance in Crohn’s disease. Retina in multiple sclerosis: review of the pertinent
et al: Frosted branch angiitis and later 1997; 17:451. literature. Mayo Clin Proc 1972; 47:335.
peripheral retinochoroidal scar in a patient 28. Quillen DA, Stathopoulos NA, 43. Vine AK: Severe periphlebitis, peripheral
with acquired toxoplasmosis. Eur J Blankenship GW, et al: Lupus associated retinal ischemia, and preretinal
Ophthalmol 2003; 13:726. frosted branch periphlebitis and exudative neovascularization in patients with multiple
15. Agrawal S, Agrawal J, Agrawal TP: maculopathy. Retina 1997; 17:449. sclerosis. Am J Ophthalmol 1992; 113:28.
Unilateral frosted branch angiitis with 29. Sugin SL, Henderly DE, Friedman SM: 44. Kerrison JB, Flynn T, Green WR: Retinal
vitreous hemorrhage. Indian J Ophthalmol Unilateral frosted branch angiitis. Am J pathologic changes in multiple sclerosis.
2001; 49:269. Ophthalmol 1991; 111:682. Retina 1994; 14:445.
2145
RETINA AND VITREOUS

45. Birch MK, Barbosa S, Blumhardt LD, et al: 55. Lobes LA, Folk JC: Syphilitic phlebitis 65. Holland GN, Pepose JS, Pettit TH, et al:
Retinal venous sheathing and the blood- simulating branch vein occlusion. Ann Acquired immune deficiency syndrome –
retinal barrier in multiple sclerosis. Arch Ophthalmol 1981; 13:825. ocular manifestations. Ophthalmology
Ophthalmol 1996; 114:34. 56. McLeish WM, Pulido JS, Holland S, et al: 1983; 90:859.
46. Brockhurst RJ, Schepens CL, Okamura ID, The ocular manifestations of syphilis in the 66. Jabs DA, Green WR, Fox R, et al: Ocular
et al: Uveitis II. Peripheral uveitis: clinical human immunodeficiency virus type manifestations of acquired immune
description, complications, and differential I-infected host. Ophthalmology 1990; deficiency syndrome. Ophthalmology 1989;
diagnosis. Am J Ophthalmol 1966; 97:196. 96:1092.
49:1257. 57. Karma A, Seppala I, Mikkila H, et al: 67. Kestelyn P, Van de Perre P, Rouvroy D, et al:
47. Wetzig RP, Chan CC, Nussenblatt RB, et al: Diagnosis and clinical characteristics of A prospective study of the ophthalmologic
Clinical and immunopathological studies of ocular Lyme borreliosis. Am J Ophthalmol findings in the acquired immune deficiency
pars planitis in a family. Br J Ophthalmol 1995; 119:127. syndrome in Africa. Am J Ophthalmol 1985;
1988; 72:5. 58. Winward KE, Smith JL, Culbertson WW, 100:230.
48. Malinowski SM, Pulido JS, Folk JC: Long- et al: Ocular Lyme borreliosis. Am J 68. Kestelyn P, Lepage P, Van de Perre P:
term visual outcome and complications Ophthalmol 1989; 108:651. Perivasculitis of the retinal vessels as an
associated with pars planitis. 59. Winterkorn JMS: Lyme disease: neurologic important sign in children with AIDS-related
Ophthalmology 1993; 100:818. and ophthalmic manifestations. Surv complex. Am J Ophthalmol 1985; 100:614.
49. Fountain JA, Werner RB: Tuberculous Ophthalmol 1990; 35:191. 69. Holland GN: Acquired immunodeficiency
retinal vasculitis. Retina 1984; 4:48. 60. Rosenbaum JT, Rahn DW: Prevalence of syndrome and ophthalmology: the first
50. Helm CJ, Holland GN: Ocular tuberculosis. Lyme disease among patients with uveitis. decade. Am J Ophthalmol 1992; 114:86.
Surv Ophthalmol 1993; 38:229. Am J Ophthalmol 1991; 112:462. 70. McClusky PJ, Wakefield D: Posterior uveitis
SECTION 10

51. Renie WA, Murphy RP, Anderson KC, et al: 61. Breeveld J, Kuiper H, Spanjaard L, et al: in the acquired immunodeficiency system.
The evaluation of patients with Eales’ Uveitis and Lyme borreliosis. Br J Int Ophthalmol Clin 1995; 35:1.
disease. Retina 1983; 3:243. Ophthalmol 1993; 77:480. 71. Gabriele P, Hutchins RK: Fusarium
52. Crouch ER, Goldberg MF: Retinal 62. Lanham JG, Barrie T, Kohner EM, et al: endophthalmitis in an intravenous drug
periarteritis secondary to syphilis. Arch SLE retinopathy: evaluation by fluorescein abuser. Am J Ophthalmol 1996; 122:119.
Ophthalmol 1975; 93:384. angiography. Ann Rheum Dis 1982; 41:473. 72. Kleiner RC: Frosted branch angiitis: clinical
53. Tamesis RR, Foster CS: Ocular syphilis. 63. Jabs DA, Fine SL, Hochberg MC, et al: syndrome or clinical sign? Retina 1997;
Ophthalmology 1990; 97:1281. Severe retinal vaso-occlusive disease in 17:370.
54. Schlaegel RF, Kao SF: A review systemic lupus erythematosus. Arch
(1970–1980) of 28 presumptive cases of Ophthalmol 1986; 104:558.
syphilitic uveitis. Am J Ophthalmol 1982; 64. Orzalesi N, Ricciardi L: Segmental retinal
93:412. periarteritis. Am J Ophthalmol 1971; 72:55.

2146
CHAPTER

169 Retinal Manifestations of the Rheumatic Diseases


Amitabh K. Bharadwaj, Carl D. Regillo, and E. Mitchel Opremcak

Rheumatologic disorders are a collection of inflammatory function. In rheumatologic diseases, inflammation of these
diseases which are typically multisystemic with protean mani- supportive structures and milieu results in tissue and organ
festations. Rheumatologic diseases include the arthritides, the dysfunction.
connective tissue diseases, and the vasculitides (Table 169.1). The eye, perhaps more than any other organ, maintains a
Despite the many clinical and pathologic presentations which unique connective tissue environment. Corneal keratocytes
exist, these diseases are so named because of the involvement secrete both type II and type IV collagen, as well as chondroitin
of these supportive structures. It is not uncommon for the eyes and keratan sulfate.1,4 This connective tissue combination
to be inflamed in many of these rheumatologic syndromes; results in strong tissue that becomes transparent as a result of
indeed, ocular inflammation is often considered a major diag- unique lamellar fiber orientation and active endothelial cell
nostic criterion for establishing a clinical diagnosis in several of dehydration. Hyalocytes in the vitreous produce hyaluronic acid
these disorders. and type II collagen, which forms a clear gel, allowing
An accurate understanding of the connective tissue proper is transmission of light, and provides support for the globe and
required to appreciate ocular involvement in rheumatic diseases. retina.1,5 The retina and choroid possess connective tissue and
Connective tissues and structures are the matrix that support a complex vascular system that is composed of type III and type
individual cells, tissues, and organs. This ground substance is IV collagen.1 These circulations are critical for retinal function
produced by specialized connective tissue cells that secrete and general nutrition of the eye. Each of these ocular tissues
various fibers (collagens, reticulin, and elastin), as well as a group performs critical functions in the visual system and is
of mucopolysaccharides called proteoglycans.1 Hyaluronic acid, exquisitely sensitive to inflammation.
chondroitin sulfate, dermatan sulfate, keratan sulfate, and Different connective tissues and structures within the eye
heparin sulfate are the common polysaccharides found in the can become inflamed in the various rheumatic diseases.
connective tissue proteoglycans.2 Collagen fibers can also be Wegener ’s granulomatosis, rheumatoid arthritis (RA), and
further subdivided by their polypeptide structure into six types polyarteritis nodosa (PAN) can affect the cornea and produce
(collagen types I to VI).3 Each tissue and organ maintains a peripheral ulcerative keratopathy. Reiter’s syndrome is defined
distinct connective tissue environment by varying these by the triad of urethritis, arthritis, and inflammation of the
individual components, thereby effecting optimal structure and conjunctiva and iris. Ankylosing spondylitis, Reiter’s syndrome,
psoriatic arthritis, and the chronic inflammatory bowel diseases
produce an acute iridocyclitis. Juvenile rheumatoid arthritis
(JRA) commonly produces a chronic iridocyclitis. It is
TABLE 169.1. Rheumatic Diseases With Ocular Involvement
important, therefore, to evaluate all patients who present with
Arthritides ocular inflammation for symptoms and signs of an underlying
Rheumatoid arthritis rheumatologic disease.
Seronegative spondyloarthropathies (HLA-B27 associated) Often, rheumatic diseases present in the eye before the onset
Ankylosing spondylitis
Reiter’s syndrome
of significant systemic involvement. The eye may even be the
Psoriatic arthritis primary target of several diseases such as Behçet’s syndrome,
Inflammatory bowel disease Reiter’s disease, and ankylosing spondylitis. The importance of
Juvenile rheumatoid arthritis performing a careful review of systems and physical examin-
Connective Tissue Diseases ation cannot be underestimated. Particular attention should be
Systemic lupus erythematosus paid to the skin, joints, central nervous system, lungs, gastro-
Progressive systemic sclerosis intestinal tract, and kidneys. Often, involvement of these
Polymyositis and dermatomyositis systems can lead the ophthalmologist to establish the existence
Sjögren’s syndrome of an underlying rheumatic disease as the cause for the ocular
Relapsing polychondritis inflammation. For example, iritis in a patient complaining of
Vasculitides large-joint arthritis could represent JRA, systemic lupus erythe-
Polyarteritis nodosa matosus (SLE), Wegener’s granulomatosis, or Behçet’s disease.
Churg–Strauss syndrome Oral ulcers, malaise, skin rash, and uveitis may be found in
Wegener’s granulomatosis SLE, Behçet’s disease, and sarcoidosis. Genital–urethral pain
Hypersensitivity vasculitis
Lymphomatoid granulomatosis and uveitis may represent Behçet’s disease, PAN, or Reiter’s
Giant-cell arteritis syndrome. Laboratory evaluation and consultation with an
Behçet’s disease internist or a rheumatologist can help confirm the presence of
a rheumatologic disease and result in diagnosis and the 2147
RETINA AND VITREOUS

initiation of proper systemic therapy. Local ocular therapy in associated underlying rheumatic disease.9 While the differential
rheumatic disorders without attention to the underlying diagnosis also includes conditions such as Wegener’s granulo-
systemic process universally results in suboptimal control of the matosis, relapsing polychondritis, and PAN, up to 30% of such
ocular inflammation and risks potentially life-threatening patients will have RA.
complications of uncontrolled systemic disease. Posterior scleritis is not as common as anterior scleral
In summary, rheumatic diseases provide an opportunity for inflammation. This condition is defined as scleritis posterior to
the ophthalmologist to interface with both the patient and the the equator of the eye, and in one series accounted for only 2%
internist. The expertise of the ophthalmologist in determining of all cases.10,11 It is important to recognize, however, that
the specific ocular tissue involved and the rheumatologist’s posterior scleritis is much more difficult to detect than anterior
knowledge of the systemic manifestations can not only scleritis. In one report of 30 eyes enucleated for ocular inflam-
facilitate the proper diagnosis but can also provide optimal care mation, 40% had previously undetected posterior scleritis.12
for these diseases of vision, which are often life-threatening. Posterior scleritis can be unilateral and is often associated
with a profound decrease in visual acuity. There is pain and
RHEUMATOID ARTHRITIS tenderness with motion or palpation. On biomicroscopic
examination, the anterior segment is often normal or may show
only a narrow angle resulting from displacement by the
Key Features posterior structures.
• Chronic idiopathic systemic inflammatory disease The fundus examination in posterior scleritis reveals
SECTION 10

• More common in females choroidal thickening or choroidal nodules overlying the area of
• Malaise, fatigue, weight loss, and arthralgia scleritis. Secondary choroidal folds and effusions may develop.
• Typically involves small joints of the wrist and hand The retina may demonstrate secondary striae and exudative
• Anterior segment findings more common retinal separations (Fig. 169.1).11 A high index of suspicion is
• Posterior scleritis may secondarily cause retinal striae and often required to make a clinical diagnosis of posterior scleritis.
exudative retinal detachment Ultrasonography or computed tomography can support this
diagnosis by showing thickening of the sclera and choroid.
There may be fluid in the contiguous Tenon space. Fluorescein
RA is a chronic systemic inflammatory disease of unknown angiography illustrates the choroidal and retinal striae. A
cause, producing a distinct form of polyarticular and symmetric characteristic linear pattern of alternating hypo- and hyper-
arthritis. It is more common in women (3:1) and typically fluorescent streaks can be seen as a result of folds in the retinal
begins between the ages of 30 and 40 years.6 RA is thought to pigment epithelial layer (see Fig. 169.1b). Multifocal, punctate,
have a strong autoimmune pathogenesis. Patients with RA have hyperfluorescent choroidal lesions can also be noted and may
immunoglobulin (Ig) M, IgG, and IgA antibodies that are evolve into areas of exudative retinal detachment. The retinal
directed against the Fc portion of IgG.7 The resulting immune circulation is typically unaffected.
complexes are postulated to mediate both the articular and the Once a clinical diagnosis of posterior scleritis is established,
extraarticular manifestations of the disease through activation a search for an underlying cause is in order. A general physical
of complement. examination and review of systems can help establish extra-
Patients with RA complain of malaise, fatigue, weight loss, ocular involvement. Patients with the characteristic deforming
and arthralgia. The joint disease is symmetric and often involves arthritis associated with RA seldom present a diagnostic
the small joints of the wrist and hand, excluding the distal challenge. Mild anemia, elevation of the erythrocyte sedimenta-
interphalangeal joints.1 Morning stiffness is characteristic. tion rate (ESR), and a positive result for rheumatoid factor in
Extraarticular involvement in RA, including pleurisy, neuro- serum may support the clinical diagnosis. Without obvious
pathy, and ocular inflammation, is thought to be secondary to systemic findings, scleritis may be the initial manifestation of
systemic vasculitis and may represent a change from a local occult rheumatic disease.
joint disease to a more serious systemic form of RA. Therapy for rheumatoid scleritis should be directed at
The most common anterior segment findings in RA are controlling the underlying systemic disease.10 Local ocular
keratoconjunctivitis sicca, marginal keratitis, peripheral ulcer- therapy and regional steroids should be used with caution and
ative keratopathy, and anterior scleritis.8 Uveitis and direct only as adjuncts to systemic treatment. Regional steroids
involvement of the retina are rare. The retina can, however, be should be used only in extenuating circumstances, as they have
involved secondarily after the development of posterior scleritis. been reported to cause scleral melting and ocular perforation.
Forty-six percent of patients with scleritis will have an Mild cases can often be effectively managed by nonsteroidal

FIGURE 169.1. Posterior scleritis in a patient


with rheumatoid arthritis with secondary
thickening of the choroid in the posterior pole
and peripapillary area. (a) Chorioretinal striae in
the macula. (b) Alternating hypofluorescent and
hyperfluorescent linear streaks correspond to
folding of the retinal pigment epithelium.

a b
2148
Retinal Manifestations of the Rheumatic Diseases

antiinflammatory agents. Indomethacin (50–150 mg/day) has occurs typically between the ages of 2 and 5 years. The cause of
been reported to be particularly effective for scleritis.9 The goal JRA is unknown, but it is thought to be a primary autoimmune
of early aggressive therapy is to prevent permanent damage disease. Children with the pauciarticular form of JRA have the
from the disease. In more severe cases, immunosuppressive highest risk for the development of ocular inflammation.20 Over
drugs such as methotrexate, azathioprine, or one of the newer 80% of such children are ANA positive.
biologic agents should be considered. The biologic agents are Chronic iridocyclitis develops in 5–17% of children with
designed to block cytokines or cytokine receptors, and include JRA.21 Eye disease may precede the development of arthritis by
medicines that decrease the activity of tumor necrosis factor- several years. Ocular involvement may be insidious because of
alpha (etanercept, adalimumab, and infliximab) and an the lack of symptoms and ocular signs early in the disease. JRA-
interleukin-1 receptor antagonist (anakinra).13,14 Oral prednisone associated iridocyclitis is typically chronic and bilateral (70%).
may be used adjunctively as a bridge to a steroid-sparing agent. The inflammation is usually nongranulomatous and involves
The ocular and systemic prognoses depend in part on primarily the iris and ciliary body. Chronic inflammation
establishing the proper diagnosis and detecting the underlying commonly results in band keratopathy (41%), posterior
rheumatic disease. In a report by Foster and associates, the synechiae, glaucoma (19%), and cataract formation (42–92%).22
development of necrotizing scleritis forbode a more severe form In JRA the retina and choroid are involved to a much lesser
of RA.15 They noted an 8-year mortality rate of 20% in patients extent than the anterior segment. Cystoid macular edema may
with this extraarticular involvement. develop in patients with JRA. As a result of chronic cyclitis,
organization and fibrosis of the anterior vitreous may occur,

CHAPTER 169
SERONEGATIVE resulting in further media opacification. Cyclitic membrane
SPONDYLOARTHROPATHIES formation and ocular hypotony can develop spontaneously after
standard cataract surgery. True retinitis, retinal vasculitis, or
(HLA-B27-ASSOCIATED) choroiditis is uncommon in JRA.
JRA can be diagnosed in children with a characteristic arthritic
Key Features and ocular picture. The diagnosis can be further supported by
• Ankylosing spondylitis, Reiter’s syndrome, psoriatic arthritis, documenting a positive antinuclear antibody (ANA) level (79%)
and arthritis associated with inflammatory bowel disease and a negative result for rheumatoid factor in serum.23 Topical
• Acute, recurrent, anterior uveitis corticosteroids and cycloplegics are the mainstay of therapy for
• Posterior segment findings: vitritis, papillitis, cystoid macular children with JRA-associated ocular inflammation. Oral
edema, vasculitis, and epiretinal membrane nonsteroidal antiinflammatory agents are useful in controlling
both the ocular and systemic symptoms.24 Regional corti-
costeroids can be used for ocular inflammation but are difficult
The group of collagen vascular diseases have in common to deliver in this age group and often require general anesthesia.
spondylitis and a strong association with HLA-B27. Ankylosing Methotrexate, cyclosporine, or other immunosuppressive agents
spondylitis, Reiter’s syndrome, psoriatic arthritis, and arthritis should be considered in patients whose eyes cannot be quieted
associated with chronic inflammatory bowel disease (Crohn’s by the above methods. Oral corticosteroids (1 mg kg–1 day–1)
disease and ulcerative colitis) compose this group. Patients with may be helpful while transitioning to a steroid-sparing agent.
these disorders experience an acute, alternating, recurrent, Due to the monitoring that is needed while on, immuno-
nongranulomatous iridocyclitis. This anterior segment inflam- suppressive agents these potentially toxic medicines should be
mation can be severe, with hypopyon and posterior synechiae used under the direction of physicians who have experience
formation. Secondary glaucoma and cataract are not uncommon. with their use.
These collagen vascular diseases do not have marked posterior The overall visual prognosis in JRA is poor. Seventy-five
segment involvement in the form of primary choroiditis, percent of children with JRA have moderate or severe
retinitis, or retinal vasculitis. Cystoid macular edema can occur inflammation with loss of vision due to glaucoma, cataract, or
with prolonged or severe cases of anterior uveitis, and this may phthisis.1,25 Lens and vitreous opacification should be
respond to therapies designed to address the iridocyclitis. addressed, preferably via pars plana lensectomy and vitrectomy,
Crohn’s disease has been reported with uveitis and severe to afford better control of the inflammation and prevent cyclitic
bilateral obliterative retinal vasculitis.16–18 In one series, 29 of membrane formation and hypotony.15,26
166 (17%) patients with HLA-B27-associated uveitis had
posterior segment manifestations including vitritis (93%), SYSTEMIC LUPUS ERYTHEMATOSUS
papillitis (83%), vasculitis (24%), cystic macular edema (38%),
and epiretinal membrane (17%).19 In this report, the retinal
vasculitis was responsive to corticosteroids or cyclophospha- Key Features
mide therapy, or a combination of these medicines. • Most patients are women around child-rearing age
• Malaise, fatigue, anorexia, and lowgrade fever
• Arthritis, facial rash, Raynaud’s phenomenon, and oral ulcers
JUVENILE RHEUMATOID ARTHRITIS may be present
• Anterior findings: keratoconjunctivitis sicca, scleritis, and
Key Features keratitis
• Childhood disease with multisystemic involvement • Posterior findings: cotton wool spots, vascular occlusions,
• Ocular inflammation may precede or follow arthritis retinal vasculitis, neovascularization, choroidopathy
• Chronic, bilateral iridocyclitis
• Pauciarticular arthritis is a risk factor for development of ocular SLE is a collagen vascular disease with protean manifestations.
inflammation Ninety percent of patients with SLE are women at or around
• Cyclitic membranes may form after standard cataract surgery child-rearing age.27 The pathogenesis of SLE appears to be an
autoimmune systemic necrotizing vasculitis. Patients with SLE
JRA is a multisystemic childhood disease associated with have ANAs and elevated levels of circulating immune com-
chronic arthritis. JRA is much more common in girls and plexes that appear to play a role in the disease.27 Immune 2149
RETINA AND VITREOUS

complexes composed of these autoantibodies and DNA have PROGRESSIVE SYSTEMIC SCLEROSIS
been found in the walls of inflamed blood vessels and in the
areas of fibrinoid necrosis.
Clinically, patients with SLE present with malaise, fatigue, Key Features
• Autoimmune disorder with multisystemic fibrosis
anorexia, and low-grade fever. On examination, they may have
• Often women in the fourth decade of life
arthritis, facial rash, alopecia, and pleurisy.28 Raynaud’s pheno-
• Anterior segment findings are most frequent, including
menon, oral ulcers, and central nervous system complaints are
keratoconjunctivitis sicca
also common. Laboratory evaluation may reveal anemia, an
• Posterior findings similar to those in SLE
elevated ESR, the presence of ANAs, proteinuria, and a false-
positive Venereal Disease Research Laboratory (VDRL) result.
Lupus nephritis and central nervous system involvement Progressive systemic sclerosis (PSS), or scleroderma, is a chronic
represent serious and potentially fatal complications of SLE. autoimmune disorder resulting in inflammation and fibrosis of
SLE may involve the eye in up to 50% of cases, depending on the skin and other organs. Women in the fourth decade of life
the series and the nature of the clinic reporting the findings.29 are affected more often than men (4:1).38 The immune defect in
Anterior segment findings include keratoconjunctivitis sicca, PSS is not completely understood, but circulating immune
scleritis, and keratitis. The retina and choroid may be primarily complex deposition, vasculitis, and secondary fibrosis of the
involved; however, it is important to separate lupus-associated vessels are thought to produce the typical clinical picture.
retinal vasculitis from the secondary retinal and choroidal Clinically, patients may present with scleroderma or may
SECTION 10

changes of SLE-mediated hypertension and anemia. Severe manifest other symptoms of the CREST syndrome (calcinosis,
hypertension can occur in lupus as a result of nephritis. Raynaud’s phenomenon, esophageal symptoms, sclerodactyly,
Arteriolar narrowing, intraretinal hemorrhages, exudate, and and telangiectasia).
disk edema are characteristic of hypertensive retinopathy. The eye is commonly involved in scleroderma. Keratocon-
The mechanism of primary lupus retinopathy is unknown junctivitis sicca occurs in up to 70% of patients with PSS as a
but is thought to be secondary to circulating immune com- result of lacrimal gland fibrosis.39 Filamentary keratitis, eyelid
plexes found in the disease. Ten percent of patients with SLE edema, and conjunctival shrinkage can also be noted. Patients
also have lupus anticoagulant antibodies that are known to with PSS have also been seen to have fundus findings similar to
increase the incidence of thrombosis. The relationship between those in SLE. Intraretinal hemorrhages, cotton wool spots, and
this factor and lupus retinopathy is unclear but provocative.30 retinal vasculitis, as well as choroidal infarctions, have been
Retinal manifestations of SLE are a result of focal ischemia and reported.40
necrotizing retinal vasculitis. Three fundus presentations have The diagnosis can be established by the characteristic clinical
been described.31–33 The most common form is focal ischemia picture of the scleroderma, with or without the other CREST
resulting in multiple cotton wool spots. Intraretinal hemorr- findings. These systemic findings in association with retinal
hages and mild disk edema can also be associated with this microvascular infarctions and retinal vasculitis support the
form of lupus retinopathy. A second form of retinopathy noted clinical diagnosis. Laboratory testing may reveal the presence of
in SLE is a severe retinal vasoocclusive disease without evidence ANAs with a speckled pattern. Fluorescein angiography can be
of retinal vasculitis.32 Retinal infarction and hemorrhage can used to document the cotton wool spots and define the extent
result in severe and sudden loss of vision. The third form of choroidal nonperfusion. Long-term systemic therapy has not
of retinopathy in SLE is proliferative lupus retinopathy been useful in controlling PSS, and therapy is chiefly sup-
(Fig. 169.2a,b). Retinal vasculitis and secondary ischemia portive.41 Localized scleroderma has a relatively good prognosis.
produce neovascularization of the optic nerve and elsewhere in Systemic involvement has a worse prognosis, with an 80%
the retina.33 This neovascularization can result in vitreous 10-year mortality rate.
hemorrhage and even retinal detachment.
The choroidal circulation may be involved in this process as DERMATOMYOSITIS AND POLYMYOSITIS
well (see Fig. 169.2c,d).34,35 Choroidal infarction, exudative
changes, central serous chorioretinopathy, and subretinal
neovascular membranes have been reported with SLE. Focal Key Features
choroidal vessel injury with secondary dysfunction of the • Diffuse inflammatory myopathies
overlying retinal pigment epithelial cells is believed to be one of • Children and adults may be affected
the mechanisms in this form of ocular disease.35 • In dermatomyositis, lilac discoloration and edema of eyelids
SLE is a clinical diagnosis. The American Rheumatologic • Cotton wool spots, intraretinal hemorrhages, venous
Association defines the disease by the presence of four of the 14 engorgement, disk edema, and optic atrophy have been
major symptoms and signs. Laboratory testing may support the reported in dermatomyositis
clinical diagnosis by revealing the presence of ANAs, elevated
circulating immune complexes, proteinuria, anemia, and a
false-positive VDRL test result. Fluorescein angiography may Dermatomyositis and polymyositis are autoimmune forms of
help define the extent of retinal involvement and may assist in inflammatory diffuse myopathy. The pathogenesis of these
differentiating secondary hypertensive retinopathy from the diseases appears to be mediated via microvascular damage from
true retinal vasculitis noted in lupus retinopathy. immune complex formation. Patients with dermatomyositis
Therapy for SLE varies according to the severity of the show ischemic necrosis and loss of capillary beds in the
systemic symptoms. It is important to note that lupus may perifascicular region of the involved muscles.42 Both children
present in the eye 1–5 years before the onset of other systemic and adults are affected by this disease.
findings.36 Lupus retinopathy may respond to systemic therapy, Ocular involvement in dermatomyositis includes a lilac
including nonsteroidal antiinflammatory agents, oral corti- discoloration and edema of the eyelids, conjunctivitis, iritis,
costeroids, hydroxychloroquine, gold, and cyclophosphamide.37 blepharoptosis, scleritis, uveitis, and extraocular muscle
Proliferative lupus retinopathy can be treated with panretinal paralysis.43 Cotton wool spots, intraretinal hemorrhages, venous
laser ablation to help control the consequences of ocular engorgement and disk edema, and optic atrophy have been
2150 neovascularization. observed primarily in childhood dermatomyositis.44
Retinal Manifestations of the Rheumatic Diseases

FIGURE 169.2. (a and b), Peripheral retinal


vasculitis in a patient with systemic lupus
erythematosus with areas of intraretinal
hemorrhage, retinal nonperfusion, and
neovascularization on fluorescein angiography.
(c and d) Another patient with systemic lupus
erythematosus with focal areas of serous
elevation of the retinal pigment epithelium and
sensory retina as a result of lupus-associated
choroidopathy.

a b

CHAPTER 169
c d

Corticosteroids (1 mg kg–1 day–1) have proved helpful in RELAPSING POLYCHONDRITIS


managing these diseases. Methotrexate and azathioprine have
been used for refractory cases or when complications of steroid
Key Features
use develop.45 The prognosis is better for childhood forms of the
• Collagen vascular disease involving cartilaginous tissues
disease, with a 90% 5-year survival rate.1 Adult-onset disease
• Bilateral ear pinna inflammation and nasal cartilage destruction
fares worse, with a 53% survival at 5 years.
• Anterior segment findings: episcleritis, scleritis, or conjunctivitis
• Anterior uveitis common
SJÖGREN’S SYNDROME • Posterior scleritis may secondarily affect retina and choroid

Key Features Relapsing polychondritis is a collagen vascular disease affecting


• Key findings are keratoconjunctivitis sicca and xerostomia primarily the cartilaginous tissues. The cause is unknown but
• More common in postmenopausal women appears to be an autoimmune disease directed against type II
• Anterior segment findings are typical collagen.50 Patients with polychondritis have circulating anti-
• Chorioretinal involvement is unclear type II collagen antibodies. These autoantibodies are thought to
play a mediating role in the recurrent, granulomatous inflam-
mation noted in this disease. Relapsing polychondritis occurs in
Sjögren’s syndrome is a constellation of clinical disorders that adults aged 20–60 years and affects both men and women.
have in common keratoconjunctivitis sicca and xerostomia.46 Systemically, patients will have bilateral ear pinna inflam-
Twenty-five percent to 50% of patients with RA have Sjögren’s mation and nasal cartilage destruction (72%).51 Nasal septal
syndrome. This syndrome is more common in postmenopausal damage typically results in a saddle nose deformity. Secondary
women. Exocrine glands, including the lacrimal gland, demon- otitis, vertigo, tinnitus, and deafness may be noted. A non-
strate infiltration with lymphocytes and secondary fibrosis. erosive polyarthralgia may also bring the patient to medical
These patients have antinucleoprotein antibodies; anti-SSA attention. Laryngeal and tracheal ring cartilage may be involved
(anti-Ro) and anti-SSB (anti-La). and can cause acute respiratory distress as a result of upper
Patients with Sjögren’s syndrome complain of dryness of the airway collapse.
mouth and eyes. Other manifestations of primary Sjögren’s Ocular involvement in relapsing polychondritis may occur in
syndrome include pneumonitis, renal tubular acidosis, poly- as many as 20–50% of cases.52 This involvement is typically in
myositis, and gastritis. the form of episcleritis, scleritis, or conjunctivitis. Anterior
Keratoconjunctivitis sicca is the most common ocular uveitis can be found in 20% of patients. Primary retinal or cho-
finding.47 Secondary filamentary keratitis can also be roidal involvement is unusual. Retinal pigment epithelial and
problematic. In one series, eight patients with a clinical picture sensory retinal detachments as well as choroidal thickening can
compatible with Sjögren’s syndrome had anterior or inter- be noted as a result of localized posterior scleritis.53
mediate uveitis but did not have chorioretinal involvement.48 In The diagnosis can be established by the pathognomonic
another report, two patients with progressive retinal vasculitis bilateral ear pinna inflammation. Biopsy of involved cartilage
were found to have anti-SSA antibodies, suggesting a Sjögren- demonstrates the characteristic granulomatous chondritis.
like syndrome.49 Their disease was unresponsive to cortico- Relapsing polychondritis is a potentially fatal disease with a
steroid, immunosuppressive, and panretinal photocoagulation 30% mortality rate.54 Dapsone (50–200 mg/day) is often used
therapy. first in patients with mild symptoms and no cardiac or 2151
RETINA AND VITREOUS

pulmonary involvement. Prednisone (0.5–1 mg kg–1 day–1) is diagnosis. Laboratory tests may show elevated white blood cell
highly effective, and may be added in cases that do not respond and eosinophil counts, decreased complement, elevated
well to dapsone alone. Dapsone does not appear to be as circulating immune complexes, and negative rheumatoid factor
effective in patients with severe necrotizing forms of scleritis.54 and ANA determinations. Hepatitis B surface antigen has been
Destructive changes in the cartilage are not reversible, and the found in up to 70% of patients with PAN. Angiography in
goal of therapy should be to limit further disease progression. patients with abdominal pain may reveal aneurysmal dilatation
of the hepatic and renal arteries.
POLYARTERITIS NODOSA PAN is a potentially fatal disease. Without therapy, it carries
an 80–90% 5-year mortality rate. Corticosteroids reduce this
mortality rate to 50%.59 The combination of corticosteroids
Key Features with cyclophosphamide (1–2 mg kg–1 day–1) results in an 80% 5-
• Multisystemic disease associated with a necrotizing vasculitis year survival rate. After disease control is achieved, the steroid
• Fatigue, myalgia, weight loss, fever, arthralgia, and testicular may be tapered. Patients may require high doses of intravenous
pain steroids and cyclophosphamide early in the course to gain
• Peripheral ulcerative keratitis and mild iritis can occur control of severe disease.60
• Most common ocular findings are choroidal and retinal
vasculitis WEGENER’S GRANULOMATOSIS
• High mortality rate if not treated early
SECTION 10

Key Features
PAN is a multisystemic disease associated with a necrotizing • Necrotizing granulomatous vasculitis
vasculitis. Medium- to small-sized vessels are characteristically • Epistaxis, rhinorrhea, sinusitis, otitis, chronic cough, and
affected with all stages of necrosis noted in the involved saddle nose deformity
tissues.55 The cause of PAN is unknown. Thirty percent to 70% • Pulmonary, renal, and CNS lesions possible
of patients with PAN have antihepatitis B antibodies.56 The • Most common ocular findings are proptosis and orbital pain
significance of this virus in the pathogenesis has not been • Scleritis, peripheral ulcerative keratitis, conjunctivitis, or
established. This is a rare disease that affects 20- to 50-year-old dacryocystitis may occur
adults. Men are affected more frequently than are women (3:1). • Posterior scleritis may cause secondary chorioretinal
Systemically, patients with PAN may note fatigue, myalgia, thickening
weight loss, fever, arthralgia, and testicular pain. The kidneys,
liver, and gastrointestinal and central nervous systems are
commonly involved. Renal involvement is one of the more Wegener’s granulomatosis is a multisystem disease associated
serious and potentially fatal complications of PAN. Abdominal with a necrotizing granulomatous vasculitis.61 The cause is
pain from intestinal infarction and headaches from central unknown. It is uncommon and occurs between the ages of 40
nervous system vasculitis are also serious and potentially life- and 50 years. Men are affected more often than are females
threatening complications of this disease. (3:2). Immune complex formation and deposition are thought
Ten to 20% of patients with PAN will have ocular to result in vasculitis of small- to middle-sized vessels. Acute
involvement.57 Peripheral ulcerative keratitis and mild iritis can and chronic lesions can be found simultaneously in the
be found in this disease. A mild vitritis may also be noted. The involved tissues and organs.
most common ocular findings in PAN are choroidal and retinal Characteristically, Wegener’s granulomatosis involves the
vasculitis.58 Fundus examination will show retinal vasculitis upper and lower respiratory tracts.62 Epistaxis, rhinorrhea,
with associated intraretinal hemorrhages, cotton wool spots, sinusitis, otitis, chronic cough, and saddle-nose deformity are
and retinal edema (Fig. 169.3). Central retinal artery occlusion frequently observed. Chronic pulmonary infiltrates, nodules,
and optic atrophy have also been reported in PAN. Patients with and cavitary lesions are found in the lungs. Wegener’s granu-
PAN may also experience anterior or posterior scleritis. lomatosis also commonly affects the kidneys and the central
Posterior scleritis will manifest with pain and chorioretinal nervous system. A granulomatous glomerulitis can be found in
folds similar to those in RA. 80% of patients with this collagen vascular disorder.
Any patient with occlusive retinal vasculitis should be Wegener’s granulomatosis may involve the eye in 40–50% of
examined for evidence of systemic findings that may help cases.63 Eye involvement may precede other organ involvement.
establish the presence of a collagen vascular disease. Often, a Proptosis and orbital pain from a pseudotumor are the most
biopsy of an involved artery or lesion will demonstrate a common findings. Scleritis, peripheral ulcerative keratitis, con-
hemorrhagic vasculitis and fibrinoid necrosis and establish the junctivitis, and dacryocystitis are also frequently noted ocular

FIGURE 169.3. (a) Localized area of


necrotizing retinal vasculitis associated with
polyarteritis nodosa. (b) The patient refused
immunosuppressive therapy and lost central
acuity in the left eye over a 5-month period
because of progressive ischemic retinal
vasculitis. Note the development of vitritis as
well as further retinal involvement.

a b
2152
Retinal Manifestations of the Rheumatic Diseases

manifestations of Wegener’s vasculitis. Posterior scleritis in subsequent destruction and fragmentation of the internal
Wegener’s granulomatosis behaves similarly to scleritis in other elastic membrane.
rheumatologic disorders and presents with decreased vision and Patients with GCA commonly complain of fever, weight loss,
pain. Secondary chorioretinal thickening and striae can be seen malaise, and headaches. Jaw claudication may occur. The most
both clinically and on fluorescein angiography. Although uveitis, common ocular complication of GCA is ischemic optic
retinitis, and retinal vasculitis have been reported, intraocular neuropathy.68 The larger vessels supplying the optic nerve
disease is rare. Retinitis and retinal vasculitis in Wegener’s become inflamed and effect an infarction of the optic nerve.
granulomatosis may present as a geographic area of retinal Rarely, the retinal vessels may be involved, producing a branch
edema and intraretinal hemorrhage (Fig. 169.4), which may or central retinal artery occlusion.69 Attenuation of the retinal
increase in size and can be difficult to distinguish from a arterioles, disk pallor, and optic atrophy may develop late.
secondary opportunistic or viral retinitis. Primary uveitis or retinitis is uncommon.
Wegener’s granulomatosis should be suspected in patients The diagnosis can be made in the clinical setting of an older
with these ocular findings and respiratory, renal, or central patient with malaise, fever, weight loss, headache, and sudden
nervous system involvement. The diagnosis can be supported loss of vision in one or both (65%) eyes as a result of ischemic
by finding pneumonitis or cavitary lesions on chest radiograph optic neuropathy. The diagnosis can be supported by finding a
film. Laboratory testing will demonstrate an elevated white markedly elevated ESR and/or C-reactive protein level. A
blood cell count, ESR, and serum IgA. Antineutrophilic cyto- temporal artery biopsy will demonstrate a granulomatous
plasmic antibodies (ANCAs) have been found in this disease vasculitis with infiltration of the vessel wall with mononuclear

CHAPTER 169
and have proved useful in advancing a diagnosis of Wegener’s cells, histiocytes, and giant cells and loss of the internal elastic
granulomatosis.64 Although failure of ANCA to return to membrane.
normal may predict an unfavorable clinical course, absolute Untreated, this disease has a poor prognosis with irreversible
titers do not appear to correlate with disease severity.65 Biopsy loss of vision secondary to ischemic optic neuropathy and death
of the involved tissue often establishes the diagnosis by from coronary or cerebral vasculitis.70 Therapy should be
revealing a granulomatous vasculitis. initiated as soon as GCA is suspected in order to prevent
Wegener’s granulomatosis is a serious and potentially fatal bilateral optic nerve involvement. Prednisone (1 mg kg–1 day–1)
disease. Therapy should be designed to address both the ocular is the treatment of choice in GCA, and maintenance doses may
and the systemic inflammation. Without therapy, the average be required for 1–2 years when the disease may remit.
survival is 5 months, with an 80% mortality rate by 1 year.66
Corticosteroids prolong survival to 12.5 months. Cyclophos- BEHÇET’S DISEASE
phamide (1–2 mg kg–1 day–1) in combination with corti-
costeroids is the treatment of choice, with a 90% remission rate.
Maintenance therapy may be required for 1–2 years. Key Features
• Systemic necrotizing vasculitis
GIANT-CELL ARTERITIS • Most prevalent in Asia and the Middle East
• Chronic prodrome of malaise, fever, and sore throat may occur
• Classic triad: recurrent aphthous oral ulcers, genital ulcers,
Key Features and uveitis
• Medium- to large-vessel systemic vasculitis
• Erythema nodosum is common
• Average age 70 years
• Typical occlusive retinal vasculitis with intraretinal hemorrhage
• Fever, weight loss, malaise, headaches, jaw claudication
and edema
• Most common ocular manifestation is ischemic optic
neuropathy
• Less commonly branch or central retinal artery occlusion Behçet’s disease is a systemic necrotizing vasculitis with diverse
manifestations. The cause is unknown but is thought to have a
strong autoimmune component. Human leukocyte antigen
Giant-cell arteritis (GCA), or temporal arteritis, is a systemic (HLA) associations have been established in other countries for
vasculitis that involves medium- to large-sized muscular Behçet’s disease but have not proved useful in the United
arteries.67 The cause is unknown. GCA affects an older popula- States.71 HLA-B51, HLA-B12, and HLA-B27 have all been
tion with an average age of 70 years. The pathophysiology of associated with certain forms of Behçet’s disease. The disease is
this collagen vascular disease appears to be a panarteritis. much more common in Asia and the Middle East.72 In the
Affected blood vessels demonstrate a mononuclear cell infiltra- United States, there is an equal gender distribution, and the
tion and giant-cell formation within the vessel wall, with disease is found in 20- to 40-year-old adults.

FIGURE 169.4. (a) An area of retinitis and


retinal vasculitis associated with biopsy-proven
Wegener’s granulomatosis. (b) The fluorescein
angiogram illustrates an area of segmental
vascular staining and leakage of dye into the
vitreous.

a b
2153
RETINA AND VITREOUS

Behçet’s vasculitis involves small- to medium-sized vessels. edema often result from the chronic inflammation. Choroidal
A perivascular infiltrate with polymorphonuclear neutrophils vessels may be involved and show delayed filling or localized
and mononuclear cells can be found around the veins and choroidal infarction.
arteries. This is often associated with vessel thrombosis and Treatment of Behçet’s disease is characteristically challenging.
tissue hemorrhage. The disease is relapsing and may persist for many years.
Clinically, there may be a 6- to 10-year prodrome with Behçet’s disease can have explosive exacerbations after periods
recurrent or chronic malaise, fever, and sore throat.73 The of relative inactivity or remission. Central nervous system
classic triad of recurrent aphthous oral ulcers (100%), genital involvement may be fatal in up to 40% of treated cases. Mild
ulcers (84%), and uveitis (66%) establishes the clinical diagnosis forms may be controlled initially with prednisone and
of Behçet’s disease. Erythema nodosum (66%), arthritis (66%), colchicine (0.6 mg PO bid).79 Usually, Behçet’s disease requires
and meningoencephalitis (22%) are also common. The gastro- more aggressive therapy. Periocular steroids can assist in
intestinal, renal, pulmonary, and cardiovascular systems may controlling severe bouts of ocular inflammation. Chlorambucil
also be involved and are considered ‘minor’ findings in this (0.1–0.15 mg kg–1 day–1) or cyclophosphamide (1–2 mg kg–1
syndrome. day–1) has been the mainstay of therapy.80 Cyclophosphamide
The eye may be the first or predominant organ involved in has fewer systemic side effects and therefore has been in favor
Behçet’s disease. More characteristically, uveitis follows the for the treatment of ocular Behçet’s disease in the United
other systemic findings by 1–3 years. Systemic and ocular States. Cyclosporine (4 mg kg–1 day–1), alone or in combination
symptoms typically wax and wane. Patients will often present with low-dose corticosteroid therapy, has also proved effective in
SECTION 10

with an acute loss of vision associated with a bilateral (80%) controlling this disease and has become the treatment of choice
uveitis.74 The ocular inflammation may be severe and relapsing. at some centers.81,82 Therapy may be required for several years.
A nongranulomatous iridocyclitis with hypopyon, posterior Careful monitoring for side effects and complications of
synechiae, and hyphema is common. On fundus examination, immunosuppressive therapy is required. Even with treatment,
there may be a severe vitritis, disk edema, and attenuation of up to 74% of patients lose useful visual acuity.
the arterioles.75 An essential finding in Behçet’s disease is the
presence of an occlusive retinal vasculitis with surrounding SUMMARY
intraretinal hemorrhage and retinal edema (Fig. 169.5). Cystoid
macular edema, cataract, glaucoma, and retinal detachment can The rheumatic diseases are a collection of inflammatory
occur as secondary complications of the uveitis and retinal disorders that have multisystemic involvement and protean
vasculitis.76 manifestations. Ocular inflammation may occur in many of
The complete form of Behçet’s disease consists of oral ulcers, these syndromes. The retina and choroid may be affected
genital ulcers, uveitis, and nonulcerative skin lesions. Several primarily as a result of retinal or choroidal vasculitis. Retinal
systems have been proposed for diagnosing partial or ischemia, cotton wool spots, intraretinal hemorrhages, retinal
incomplete forms of Behçet’s disease.77 Behçet’s disease is more edema, and retinal vasculitis are the typical findings in many of
prevalent in Japan, leading the Japanese to classify this disease these diseases. Posterior scleritis may result in secondary
into four forms: (1) complete-all four major findings, chorioretinal involvement.
(2) incomplete-three major findings or uveitis with one other The ophthalmologist should be aware of the association
major finding, (3) suspect-two major findings, and (4) possible- between ocular inflammation and the various rheumatologic
one major finding. disorders. Many of these diseases either involve the eye
The presence of other minor findings associated with the primarily or present initially in the eye, and can precede the
multisystemic involvement can assist in the diagnosis. involvement of other organ systems by several years. A careful
Laboratory testing may help by demonstrating an elevated ESR, review of systems and general physical examination should be
elevated levels of C-reactive protein, immune complexes, and a performed to ascertain systemic involvement. The prognosis for
positive ANA determination. HLA typing can also be helpful in many of these vision- and life-threatening disorders depends on
incomplete forms of Behçet’s disease. Properdin factor B, serum accurate diagnosis and the institution of appropriate therapy.
lysozyme, and a1-acid glycoprotein are also elevated in Behçet’s Until the pathophysiology and exact causes of the rheuma-
disease.78 Pathergy and dermatographia, although much extolled, tologic diseases are defined, antiinflammatory and immuno-
have not proved to be helpful measures for detecting Behçet’s suppressive agents remain the mainstays of therapy. As such,
disease in the United States. Fluorescein angiography can be the rheumatic diseases are best managed by a multidisciplinary
employed to help document the ischemic retinal vasculitis and approach, not infrequently negotiated by the ophthalmologist
follow up response to therapy. Cystoid macular edema and disk and the temper of the eye.

FIGURE 169.5. (a and b) Two patients with


Behçet’s disease illustrate the profound
involvement of the retinal vessels. A bilateral
occlusive retinal vasculitis results in intraretinal
edema, hemorrhage, and retinal nonperfusion.
Progressive damage to the sensory retina
results in marked attenuation of the arterioles.

a b
2154
Retinal Manifestations of the Rheumatic Diseases

REFERENCES
1. Rodnan GP, Schumacher RH: The 24. Olson NY, Lindsley CB, Godfrey WA: 46. Manthorpe R, Frost-Larson K, Isager H,
connective tissues: structure, function and Nonsteroidal anti-inflammatory drug et al: Sjögren’s syndrome. Allergy 1981;
metabolism. In: Rodnan GP, Schumacher therapy in chronic childhood iridocyclitis. 36:139.
RH, eds. Primer on the rheumatic diseases. Am J Dis Child 1988; 142:1289. 47. Brown SI, Grayson M: Marginal furrows: a
8th edn. Atlanta: Arthritis Foundation; 1983. 25. Kanski JJ: Uveitis in juvenile rheumatoid characteristic corneal lesion of rheumatoid
2. Hascall VC, Hascall GK: Proteoglycans. In: arthritis: incidence, clinical features and arthritis. Arch Ophthalmol 1968; 79:563.
Hay ED, ed. Cell biology of extracellular prognosis. Eye 1988; 2:641. 48. Rosenbaum JY, Bennett RM: Chronic
matrix. New York: Plenum; 1981. 26. Diamond JG, Kaplan HL: Lensectomy and anterior and posterior uveitis and primary
3. Sanberg LB, Gray WR, Franzblau C, eds: vitrectomy for complicated cataract Sjögren’s syndrome. Am J Ophthalmol
Elastin and elastic tissue. New York: secondary to uveitis. Arch Ophthalmol 1987; 104:346.
Plenum; 1977. 1978; 96:1798. 49. Farmer SG, Kinyoun MD, Nelson JL, et al:
4. Friend J: Physiology of the cornea. In: 27. Mintz G, Fraga A: Arteritis in systemic Retinal vasculitis associated with autoan-
Smolin G, Thoft RA, eds. Cornea. 2nd edn. lupus erythematosus. Arch Intern Med tibodies to Sjögren’s syndrome A antigen.
Boston: Little, Brown; 1987. 1965; 116:55. Am J Ophthalmol 1985; 100:814.
5. Fine BS, Yanoff M: The vitreous body. In: 28. Estes D, Christian CL: The natural history 50. Foidart JM, Abe S, Marin GR, et al:
Fine BS, Yanoff M, eds. Ocular histology. of systemic lupus erythematosus by Antibodies to type II collagen in relapsing
2nd edn. New York: Harper & Row; 1979. prospective analysis. Medicine 1971; 50:85. polychondritis. N Engl J Med 1978;
6. Kelgren HJ: Epidemiology of rheumatoid 29. Baehr G, Klemperer R, Schifrin A: A diffuse 299:1203.

CHAPTER 169
arthritis. In: Dutker JJR, Alexander WRM, disease of the peripheral circulation (usually 51. McAdam LP, O’Hanllan MA, Bluestone R,
eds. Rheumatic diseases. Baltimore: associated with lupus erythematosus and et al: Relapsing polychondritis: prospective
Williams & Wilkins; 1968. endocarditis). Trans Assoc Am Phys 1935; study of 23 patients and a review of the
7. Torrigiana G, Roitt IM: Antiglobulin factors 50:139. literature. Medicine 1976; 55:193.
in sera from patients with rheumatoid 30. Levine SR, Crofts JW, Lesse GR, et al: 52. Isaak BL, Liesang TJ, Michet CJ: Ocular
arthritis and normal subjects. Ann Rheum Visual symptoms associated with the and systemic findings in relapsing
Dis 1977; 3:315. presence of a lupus anticoagulant. polychondritis. Ophthalmology 1986;
8. Barr CC, Davis H, Culbertson WW: Ophthalmology 1988; 95:686. 93:681.
Rheumatoid scleritis. Ophthalmology 1981; 31. Gold DH, Morris DA, Henkind P: Ocular 53. Magargal LE, Donoso LA, Goldberg RE,
88:1269. findings in systemic lupus erythematosus. et al: Ocular manifestations of relapsing
9. Waston PG, Hayreh SS: Scleritis and Br J Ophthalmol 1972; 56:800. polychondritis. Retina 1981; 1:96.
episcleritis. Br J Ophthalmol 1976; 60:163. 32. Gold D, Feiner L, Henkind P: Retinal arterial 54. Hoang-Xuan T, Foster CS, Rice BA:
10. Benson WE, Sheilds JA, Tasman W, et al: occlusive disease in systemic lupus Scleritis in relapsing polychondritis:
Posterior scleritis: a cause of diagnostic erythematosus. Arch Ophthalmol 1977; response to therapy. Ophthalmology 1990;
confusion. Arch Ophthalmol 1979; 95:1580. 97:892.
97:1482. 33. Vine AK, Barr CC: Proliferative lupus 55. Christain CL, Sargent JS: Vasculitis
11. Singh G, Guthoff R, Foster CS: Observation erythematosus. Arch Ophthalmol 1988; syndromes, clinical and experimental
on long-term follow-up of posterior 106:230. models. Am J Med 1976; 61:385.
scleritis. Am J Ophthalmol 1986; 101:570. 35. Cunningham ET Jr, Alfred PR, Irvine AR: 56. Gocke DJ, HSU K, Morgan C, et al:
12. Fraunfelder FT, Watson PG: Evaluation of Central serous chorioretinopathy in patients Association between polyarteritis and
eyes enucleated for scleritis. with systemic lupus erythematosus. Australia antigen. Lancet 1970;
Br J Ophthalmol 1976; 60:227. Ophthalmology 1996; 103:2081. 2:1149.
13. O’Dell JR: Therapeutic strategies for 36. Wong K, Everett A, Jones JV, et al: Visual 57. Stillerman ML: Ocular manifestations of
rheumatoid arthritis. N Engl J Med 2004; loss as the initial symptom of systemic diffuse collagen disease. Arch Ophthalmol
350:2591. lupus erythematosus. Am J Ophthalmol 1951; 45:239.
14. Olsen NJ, Stein CM: New drugs for 1981; 92:238. 58. Goar EL, Smith LS: Polyarteritis nodosa
rheumatoid arthritis. N Engl J Med 2004; 37. Neumann R, Foster CS: Corticosteroid- of the eye. Am J Ophthalmol 1952;
350:2167. sparing strategies in the treatment of retinal 35:1619.
15. Foster CS, Forstot SL, Wilson LA: Mortality vasculitis in systemic lupus erythematosus. 59. Fauci AS, Doppman JL, Wolff SM:
rate in rheumatoid arthritis patients Retina 1995; 15:206. Cyclophosphamide-induced remissions in
developing necrotizing scleritis or 38. Maricq HR, LeRoy EC: Progressive advanced polyarteritis nodosa. 1978;
peripheral ulcerative keratitis. systemic sclerosis: disorders of the Am J Med 64:890.
Ophthalmology 1984; 91:1253. microcirculation. Clin Rheum Dis 1979; 60. Colmegna I, Maldonado-Cocco JA:
16. Salmon JF, Wright JP, Bowen RM, et al: 5:81. Polyarteritis nodosa revisited. Curr
Granulomatous uveitis in Crohn’s disease. 39. Horan EC: Ophthalmic manifestations of Rheumatol Rep 2005; 7:288.61. Goodman
Arch Ophthalmol 1989; 107:718. progressive systemic sclerosis. GC, Churg J: Wegener’s granulomatosis:
17. Duker JS, Brown GC, Brooks L: Retinal Br J Ophthalmol 1969; 53:388. pathology and review of the literature. Arch
vasculitis in Crohn’s disease. 40. Pollack IP, Becker B: Cytoid bodies of the Pathol 1954; 58:533.
Am J Ophthalmol 1987; 103:664. retina in a patient with scleroderma. 62. Robin JB, Schanzlin DJ, Meisler DM, et al:
18. Ruby AJ, Jampol LM: Crohn’s disease and Am J Ophthalmol 1962; 54:655. Ocular involvement in the respiratory
vascular disease. Am J Ophthalmol 1990; 41. Medsger TA, Masi AT, Rodnan GP, et al: vasculitides. Surv Ophthalmol 1985;
110:349. Survival with systemic sclerosis 30:127.
19. Rodriguez A, Akova YA, Pedroza-Seres M, (scleroderma): a life-table analysis of 309 63. Bullen CL, Liesegang TJ, McDonald TJ,
Foster CS: Posterior segment ocular patients. Ann Intern Med 1971; 75:369. et al: Ocular complications of Wegener’s
manifestations in patients with 42. Kissel JT, Mendell JR, Rammohan KW: granulomatosis. Ophthalmology 1983;
HLA-B27-associated uveitis. Microvascular deposition of complement 90:279.
Ophthalmology 1994; 101:1267. membrane attack complex in 64. Pulido JS, Goeken JA, Nerad JA, et al:
20. Kanski JJ: Juvenile arthritis and uveitis. dermatomyositis. N Engl J Med 1986; Ocular manifestations of patients with
Surv Ophthalmol 1990; 34:253. 314:329. circulating antineutrophilic cytoplasmic
21. Kanski JJ, Shun-Shin GA: Systemic uveitis 43. Harrison SM, Frenkel M, Grossman BJ, et al: antibodies. Arch Ophthalmol 1990;
syndromes in childhood: an analysis of Retinopathy in childhood dermatomyositis. 108:845.
340 cases. Ophthalmology 1984; 91:1247. Am J Ophthalmol 1973; 76:786. 65. Power WJ, Rodriguez A, Neves RA, et al:
22. Giles CL: Uveitis in childhood. Part I: 44. Bruce GM: Retinitis in dermatomyositis. Disease relapse in patients with ocular
anterior. Ann Ophthalmol 1989; 21:13. Trans Am Ophthalmol Soc 1938; 36:282. manifestations of Wegener granulomatosis.
23. Kanski JJ: Anterior uveitis in juvenile 45. Oddis, CV: Idiopathic inflammatory Ophthalmology 1995; 102:154.
rheumatoid arthritis. Arch Ophthalmol 1977; myopathies: a treatment update. Curr 66. Fauci AS, Haynes BF, Katz P: The
95:1794. Rheumatol Rep 2003; 5:431. spectrum of vasculitis: clinical, pathologic, 2155
RETINA AND VITREOUS

immunologic and therapeutic 72. Aoki K, Fujioka K, Katsumata H, et al: ulceration and Behçet’s syndrome.
considerations. Ann Intern Med 1978; Epidemiologic studies on Behçet’s disease J Clin Pathol 1980; 33:269.
89:660. in Hokkaido district. [Japanese] 79. Hijakata K, Masuda K: Visual prognosis in
67. Huston KA, Hunder GC, Lie JT, et al: J Clin Ophthalmol 1971; 25:2239. Behçet’s: effects of cyclophosphamide
Temporal arteritis. A 25-year 73. Chajek T, Fainaru M: Behçet’s disease: and colchicine. Jpn J Ophthalmol 1978;
epidemiological, clinical and pathological report of 41 cases and a review of the 22:506.
study. Ann Intern Med 1978; 88:162. literature. Medicine 1975; 54:179. 80. O’Duffy JD, Robertson DM, Goldstein NP:
68. Keltner JL: Giant-cell arteritis. 74. Michelson JB, Chisari VF: Behçet’s Chlorambucil in the treatment of uveitis and
Ophthalmology 1982; 89:1101. disease. Surv Ophthalmol 1982; 26:190. meningoencephalitis of Behçet’s disease.
69. Whitfield JH, Bateman M, Cooke WT: 75. James DG, Spiteri MA: Behçet’s disease. Am J Med 1984; 76:75.
Temporal arteritis. Br J Ophthalmol 1963; Ophthalmology 1982; 89:1279. 81. Nussenblatt RB, Palastine AG, Chan CC:
47:555. 76. Colvard DM, Robertson DM, O’Duffy JD: Effectiveness of cyclosporin therapy in
70. Cullen JF, Colier JA: Ophthalmic The ocular manifestations of Behçet’s Behçet’s disease. Arthritis Rheum 1985;
complications of giant cell arteritis. Surv disease. Arch Ophthalmol 1977; 95:1813. 28:671.
Ophthalmol 1976; 20:247. 77. Behçet’s Disease Research Committee of 82. Whitcup SM, Salvo EC Jr, Nussenblatt RB:
71. Numaga J, Kazumasas M, Mochizuki M, Japan: Behçet’s disease: guide to Combined cyclosporine and corticosteroid
et al: An HLA-D region restriction fragment diagnosis of Behçet’s disease. therapy for sight-threatening uveitis in
associated with refractory Behçet’s Jpn J Ophthalmol 1974; 18:291. Behçet’s disease. Am J Ophthalmol 1994;
disease. Am J Ophthalmol 1988; 78. Lehner T, Adinolfi M: Acute phase proteins, 118:39.
105:528. C9, factor B and lysozyme in recurrent oral
SECTION 10

2156
CHAPTER

170 Retinopathy Associated with Blood Anomalies


John I. Loewenstein and Demetrios Vavvas

ANEMIA Vision is not affected unless there are changes at the macula,
and such changes are unusual. Retinopathy resolves with
treatment of the underlying anemia.
Key Features The picture as a whole is suggestive, but not diagnostic, of
• Superficial flame-shaped hemorrhages and cotton wool spots anemia. It usually is not possible to determine the type of
are typically seen anemia from the fundus findings. Some authors feel that white-
• Approximately one-third of anemic patients may have some centered hemorrhages are more common in pernicious anemia
retinopathy than in other types of anemia2; others state that a picture of
• Prevalence of retinopathy increases with severity of anemia or numerous preretinal hemorrhages, together with white-centered
thrombocytopenia hemorrhages, is characteristic of aplastic anemia,3 but systematic
• Retinal abnormalities in anemia without accompanying quantitative studies have not been undertaken. Two case reports
thrombocytopenia are less common, unless the anemia is link moderately severe iron deficiency anemia and retinal
profound. arterial occlusive events.4,5 With only two cases, it is difficult to
• The combination of thrombocytopenia and anemia makes prove causality. One case of Fanconi anemia, a rare chromosomal
retinopathy more likely. abnormality, has been reported with widespread vasculopathy,
• Vision is not affected unless macula is involved including areas of central and peripheral leakage, areas of
• Treat the underlying condition nonperfusion and neovascularization.6 Angioid streaks have
been reported to occur with familial dyserythropoietic anaemia
type III.7 Anemia associated with malaria has been reported to
Fundus findings in anemia may include hemorrhages, cotton wool have high incidence of retinopathy, but cerebral malaria without
spots, and retinal edema. The hemorrhages are typically super- anemia can also have fundus changes, and the severity of
ficial and flame shaped, but they may also have a white center fundus changes may be a predictor of worse outcome.8
(Figs 170.1 and 170.2). Rarely, preretinal or vitreous hemor- There is controversy in the literature regarding the most
rhage or a macular star may occur. The retinal vessels are important factors related to the prevalence of retinopathy in
usually normal, although pale arterioles and dilated veins may anemia. Age, sex, type of anemia, hematocrit values, and platelet
be seen.1 Fluorescein angiography may reveal an increased retinal counts have all been invoked, but only a few quantitative studies
transit time. have been performed. Foulds3 studied 30 cases of pernicious

FIGURE 170.1. Intraretinal hemorrhages and cotton wool spots in a FIGURE 170.2. Intraretinal hemorrhages, white-centered hemorrhage,
case of severe anemia. and cotton wool spots in a case of aplastic anemia. 2157
RETINA AND VITREOUS

anemia. He found retinopathy in all patients with a hemoglobin cytopenia was more likely to yield retinopathy. They found that
concentration of less than 5 g/dL, in one-third of patients with a more profound anemia was required to produce cotton wool
hemoglobin concentration of 5–6 g/dL, and in no patients with a spots than was required to produce hemorrhages.
hemoglobin concentration of greater than 6 g/dL. On this basis, Carraro and associates13 studied 226 consecutive patients with
he states that retinopathy is most likely associated with a anemia alone, thrombocytopenia alone or both. They excluded
hemoglobin concentration of less than 6 g/dL. Wise and patients with leukemia, diabetes and hypertension and liver
colleagues1 stated that a hemoglobin concentration of 2.5 g/dL cirrhosis. All patients had dilated eye exams by ophthalmologists.
or less is usually associated with retinopathy. Merin and Freund9 Most common fundus findings were superficial flame-shaped or
studied 89 patients with anemia in Africa. The majority of their discrete hemorrhages. There were few patients with subhyaloid
patients had anemia secondary to nutritional deficiency and hemorrhages and one with disk edema. As a whole, 28% of the
hookworm infestation. None of their patients with a hemo- patients had retinopathy findings. Among patients with both ane-
globin level greater than 8 g/dL had retinopathy, whereas 46% mia and thrombocytopenia the incidence of retinopathy ranged
of those with a hemoglobin concentration of less than 8 g/dL from ~10% with the presence of mild anemia and thrombocyto-
showed hemorrhages or ‘exudates’, or both. Aisen and penia and increased to 100% in the presence of combined severe
associates10 examined 35 anemic patients and 35 age- and sex- anemia and severe thrombocytopenia. In severe anemia (less than
matched controls. Twenty percent of the anemic patients had 8 g/dL) only, 70% of patients had retinopathy findings, and in
hemorrhages or cotton wool spots, but there was no correlation severe or very severe thrombocytopenia alone (<50 µ 109/L),
with the hematocrit value. Rubenstein and co-workers11 studied 45% of patients (four of nine) had retinopathy findings.
SECTION 10

two groups of patients with anemia. Group 1 consisted of Merin and Freund,9 in their study of anemia in Africa, could
67 patients, each having a hemoglobin concentration of less find no cases of retinopathy in children, despite very low hemo-
than 12 g/dL or a platelet count of less than 100 000, or both. A globin levels. This has led to speculation that age is a factor in
variety of diseases was present in this group, including many the prevalence of retinal abnormalities in anemia. Aisen and
cases of leukemia. Some of the leukemic patients were receiving associates,10 however, could not confirm a correlation between
chemotherapy. It was demonstrated in these patients that, for a age and retinopathy in their population. These two studies also
given platelet level, a lower hematocrit value was more likely to suggested that hemorrhages and cotton wool spots in anemia were
be associated with retinal hemorrhages. Similarly, for a given more common in males than in females. Aisen and associates
hemoglobin concentration, more severe thrombocytopenia was subjected this hypothesis to statistical analysis and failed to find
more likely to be associated with retinal hemorrhage. The data significance despite the trend. The Carraro et al study,13 which is
for group 1 are summarized in Table 170.1. the largest and most carefully performed to date, failed to find
Group 2 consisted of 123 hemophiliacs and 42 patients with an association between age or sex and retinopathy in multi-
Cooley’s anemia. All patients in this group had a hemoglobin variate analysis. They did not, however, include children.
concentration of less than 8 g/dL at some point in the course of In summary, a precise level of anemia at which retinal abnor-
disease, but none had thrombocytopenia. No patient in this malities will occur cannot be given. Several studies have shown
group had retinal hemorrhages. Not all members of group 2 had that prevalence of retinopathy increases with severity of anemia.
a dilated fundus examination by an ophthalmologist, whereas Retinal abnormalities in anemia without accompanying thrombo-
all members of group 1 did have such an evaluation. It is cytopenia are less common, unless the anemia is profound. The
therefore possible that the number of retinal hemorrhages in combination of thrombocytopenia and anemia makes retino-
the patients in group 2 was underestimated. Since many pathy more likely. Young children may be less likely to show
leukemic patients were included in group 1, it is not possible to retinal changes with anemia, unless it is caused by leukemia.
determine from their data whether the relationship among Venous tortuosity may be a feature of anemic retinopathy,10
anemia, thrombocytopenia, and retinopathy holds true for but there is no correlation with the hematocrit value.14
nonleukemic cases. Holt and Gordon-Smith12 examined a large The pathophysiology of retinopathy in anemia is poorly under-
number of patients with a variety of blood diseases. They stood. It is possible that dilatation of retinal vessels occurs as a
studied 33 patients with leukemia and found that 18 of them response to retinal hypoxia in profound anemia. The resulting
had retinopathy, which was more likely with increasing anemia increase in transmural pressure, or perhaps the hypoxia itself, may
and thrombocytopenia. These authors did not report platelet damage vascular walls. Thrombocytopenia might contribute by
levels in their other cases of anemia, but they did give the retarding healing. This scheme might explain vascular leakage
impression that the combination of anemia and thrombo- that produces hemorrhage and edema. Cotton wool spots are
infarcts of the nerve fiber layer of the retina. It is possible that
they are produced by relative hypoxia in anemia; vascular spasm
might explain their focal nature.
TABLE 170.1. Data for Group 1 in Rubenstein and Co-Workers'
Study LEUKEMIA
Hemoglobin Patients Patients with
Platelets/mm2 g/dL (n) Retinal Hemorrhage
Key Features
<50 000 <8 10 7
• Up to 40% of the patients may have ocular findings
8–12 14 5
>12 1 0 • Hemorrhages (flame, white-centered, preretinal, and vitreous)
and cotton wool spots are the most common findings
50 000–100 000 <8 7 3 • Venous dilatation, irregularity, and vessel sheathing may be seen
8–12 8 2
• Leukemic infiltrates can occur (white clumps or masses in the
>12 5 0
retina/optic nerve)
>100 000 <8 19 2 • Microaneurysms and retinal neovascularization are sometimes
8–12 3 0 seen in chronic leukemia
Data from Rubenstein RA, Yanoff M, Albert DM: Thrombocytopenia, anemia, • Serous detachment of the retina may occur as a result of
and retinal hemorrhage. Am J Ophthalmol 1968; 65:435. choroidal infiltration
2158
Retinopathy Associated with Blood Anomalies

leukemia. Serous detachment of the retina may occur as a result


• Retinopathy is more common in adults than in children and
of choroidal infiltration.15–17
more prevalent in myeloid leukemia versus lymphoid
In general, the presence of retinal abnormalities in leukemia
• HTLV-1 leukemia has a somewhat different clinical picture that
has not been well correlated with the white cell count. Autopsy
includes pigmentary retinopathy, lymphoma-like picture, uveitis,
studies show a high agonal white cell count in patients with
and vasculitis
leukemic infiltration,18 but the relationship between the white
• Treat underlying pathology
cell count and infiltration was difficult to demonstrate in most
clinical studies. A recent study of leukemic patients in Malaysia
The fundus in leukemia may show venous dilatation, tortuosity, however, demonstrated an association between hemorrhages
and irregularity, and vessels may show abnormal color and with white centers and the white blood count, whereas hemor-
sheathing. Flame, white-centered, preretinal and vitreous hemor- rhages without white centers were found to correlate with low
rhages; cotton wool spots; and leukemic infiltrates may occur platelet counts.19
(Figs 170.3 to 170.5). The latter appear as white clumps or The anemia, thrombocytopenia, and hyperviscosity that may
masses in the retina. Microaneurysms, venous occlusions, and accompany leukemia probably account for many of the retinal
neovascularization are sometimes seen, typically in chronic findings. Retinal hemorrhages are likely the result of anemia
and thrombocytopenia. As discussed in the section on anemia,
for a given platelet level, a lower hematocrit value is more likely
to be associated with retinal hemorrhages.11,12 Similarly, for a

CHAPTER 170
given hemoglobin level, more severe thrombocytopenia is more
likely to be associated with retinal hemorrhage. A prospective
study of retinopathy in leukemia by Guyer and colleagues20 as
well as a study by Reddy and Jackson19 demonstrated signifi-
cantly lower platelet counts in patients with retinal hemorrhages
in comparison with those without such lesions, regardless of
the type of leukemia. In these studies, patients with acute
lymphocytic leukemia and retinal hemorrhages showed lower
hematocrit levels than did those without retinal hemorrhages.
This relationship was not demonstrated in patients with myeloid
leukemia. In patients with acute nonlymphocytic leukemia, the
presence of anemia was related to the presence of white-centered
hemorrhages in Guyer’s study20 but not in Reddy and Jackson’s.19
Hemorrhages (with or without white centers) and cotton wool
spots were more common in adults than in children in Guyer’s
study but not in Reddy and Jackson’s. There was no correlation
between fundus findings and mean leukocyte counts in Guyer’s
study. Karesh and co-workers,21 in a prospective evaluation of
the fundi of patients with myeloid leukemia, demonstrated
significantly lower platelet counts in patients with retinopathy
than in those without fundus lesions. They found no difference
in hematocrit levels and white cell counts in groups with and
without retinopathy. They did not do a separate analysis for
FIGURE 170.3. Large leukemic infiltrate. different types of retinal lesions. Most of their patients with
From Schachat AP, Markowitz JA, Guyer DR, et al: Ophthalmic manifestations of retinopathy had retinal hemorrhages.
leukemia. Arch Ophthalmol 1989; 107:698. Copyright 1989, American Medical Venous dilatation, tortuosity, and irregularity seen in leukemia
Association. may be caused by a combination of anemia and hyperviscosity.

a b

FIGURE 170.4. (a) Intraretinal and preretinal hemorrhages in a case of acute lymphatic leukemia. Right eye. (b) Left eye of the patient in (a). 2159
SECTION 10 RETINA AND VITREOUS

a b

FIGURE 170.5. (a) Retinal vascular dilatation and tortuosity in a case of secondary polycythemia. Right eye. (b) Left eye of the patient in (a).

Venous occlusions are likely to result from hyperviscosity. Cotton serous retinopathy cases represented a chance occurrence of two
wool spots are caused by occlusion of precapillary arterioles, conditions, a manifestation of leukemia, or were limited presen-
but the reason for these occlusions in leukemia has not been tations of serous retinal detachment secondary to infiltration
established. Ischemia secondary to anemia, direct occlusion by of the choroid. Kuwabara and Aiello34 described the pathologic
leukemic cells, occlusion by platelet-fibrin aggregates, or sludging findings in a case of leukemic miliary nodules of the retina.
resulting from hyperviscosity may all be factors. In their prospec- Human T-cell lymphotropic virus type 1 (HTLV-1) is a retro-
tive study, Guyer and colleagues20 did not find an association virus that can cause leukemia. The virus is endemic in southwest
between hematologic variables and cotton wool spots. Japan, some Caribbean countries, sub-Saharan Africa, and parts
Microaneurysms are seen in cases of chronic leukemia and of the Middle East. It has been seen in Melanesia, the south-
may be related to hyperviscosity.22,23 In Jampol and colleagues’23 eastern United States, and South America. Transmission of the
study of 25 patients with chronic leukemia, eight individuals virus is similar to that of human immunodeficiency virus.
had microaneurysms. Infection with HTLV-1 may produce several clinical pictures.35
Neovascularization may also be seen in cases of chronic The most common are transverse myelopathy and leukemia/
leukemia.24 It is usually accompanied by capillary closure.25 lymphoma. It may also produce pulmonary alveolitis, Sjögren’s
The white centers of some hemorrhages seen in leukemia may syndrome, and polymyositis.
consist of leukemic cells and debris, platelet-fibrin aggregates, About 10% of patients with transverse myelopathy and HTLV-1
or septic emboli.26 infection have a pigmentary retinopathy.36,37 Some of these
The prevalence of retinal involvement in leukemia varies widely cases show a progressive, widespread retinal degeneration indis-
in the literature. Almost all studies suffer from their retrospective tinguishable from retinitis pigmentosa. The electroretinogram
nature and a variety of selection biases. Schachat and associates27 in these cases is markedly abnormal. Other patients have a
performed a prospective ophthalmic study of patients with
newly diagnosed leukemia. A summary of the prevalence of retinal
findings in their patients is given in Table 170.2. They found a 5% TABLE 170.2. Prevalence of Retinal Findings in Schachat and
prevalence of visual loss caused by vitreoretinal disease in those Associates’ Study
patients in whom a reliable acuity determination was possible.
Another prospective study of 288 patients found similar results, Retinal Finding Myeloid Lymphoid Total
with about a third of the patients showing retinal findings. A low Patients (%) Patients (%) Affected (%)
percentage (~ 1.5%) had neuro-ophthalmic findings. Ocular Intraretinal 33 13 24
findings were more common in adults than in children (50% vs hemorrhage
16%) and more prevalent in myeloid leukemia versus lymphoid Cotton wool 24 6 16
(40% vs 30%).28 Karesh and co-workers21 prospectively evaluated spot
the fundi of patients with myeloid leukemia and found that 53%
of their subjects had retinopathy. Five of 53 patients in their White-centered 13 8 11
hemorrhage
study had visual loss in one or both eyes related to macular
hemorrhage. Central vein 7 0 4
Alterations in the retinal pigment epithelium caused by occlusion
leukemia have been reported by several authors.29–32 The appear- Vitreous 4 0 2
ance is one of pigment clumping or a reticular pattern of pigment hemorrhage
change. Pathologic examination demonstrates leukemic infiltra-
Leukemic 3
tion of the choroid and retina, with destruction and hyperplasia infiltrate
of the pigment epithelium.
From Schachat AP, Markowitz JA, Guyer DR, et al: Ophthalmic manifestations of
There are reports of bilateral central serous retinopathy in leukemia. Arch Ophthalmol 1989; 107:697. Copyright 1989, American Medical
acute lymphocytic leukemia as well as other serous retina Association.
2160 detachments.15–17,33 It is difficult to know whether the central
Retinopathy Associated with Blood Anomalies

sectoral or regional atrophy of the retina and choroid that is clinical signs of hyperviscosity begin with a hematocrit value
nonprogressive. Fukushima and associates38 demonstrated that greater than 50%. Wise and colleagues1 noted that there is a
T cells responsive to HTLV-1-infected cells are cross-reactive linear relationship between viscosity and hematocrit value up
with retinal antigens, suggesting a possible mechanism for the to a hematocrit of 50%. When the level is greater than this,
retinal degeneration. viscosity increases exponentially with the rising hematocrit
Retinal infiltration similar to that seen in large-cell lym- value. Blood flow in retinal vessels is typically laminar, and
phoma may occur with HTLV-1 leukemia/lymphoma. Kumar Foulds noted that this may be responsible for the proportion-
and colleagues39 described such a patient who presented with ality of whole-blood viscosity to the shear rate; thus, whole-blood
deep retinal and subretinal infiltrates. Chorioretinal biopsy viscosity is greater when blood flow is slower, and hyper-
demonstrated large atypical mononuclear cells located primarily viscosity, therefore, results in greater changes in the venous
in the retinal pigment epithelium, but focally involving the system.
neurosensory retina. In polycythemia, the fundus typically shows dark, dilated,
HTLV-1 infection has been associated with anterior uveitis, tortuous veins (Figs 170.6 and 170.7). The disk is usually hyper-
vitreous infiltration, and uveitis with retinal perivasculitis.40,41 Evi- emic and swollen, intraretinal hemorrhages are frequently seen,
dence that the virus may be causative of these forms of uveitis and there may be retinal edema. Central or branch vein retinal
is presented in two studies. Nakao and co-workers42 demonstrated occlusions may occur. Patients may lose vision because of retinal
that 41% of patients with nonspecific uveitis had positive titers edema or vein occlusion. Abnormalities are almost always
to HTLV-1, compared with ~23% of patients with other ocular present in both eyes; the patient with a central vein occlusion

CHAPTER 170
disorders. The latter percentage did not differ significantly from in one eye and a normal fellow eye rarely has underlying
the general population in this endemic area. polycythemia.
Mochizuki and associates43 found a 35% seroprevalence of Bilateral vein occlusion in polycythemia secondary to
HTLV-1 in nonspecific uveitis patients in an endemic area, Eisenmenger syndrome has been reported.46 However, primary
versus 16% in patients attending the eye clinic who did not have polycythemia is more likely to produce retinopathy than is
uveitis. The typical clinical picture included iritis, midvitreous secondary polycythemia, probably because of higher red cell
inflammation, and retinal vasculitis. In this study, proviral DNA counts and hyperviscosity in the former.1 Treatment will reverse
was detected by polymerase chain reaction in inflammatory cells most of the findings unless venous occlusion has occurred.
from the anterior chambers of seven of nine seropositive patients
with uveitis. The inflammatory cells from patients with other DYSPROTEINEMIAS
forms of uveitis were tested, and none were positive.
A mother and son who were both infected with HTLV-1 devel-
oped white-yellow intraretinal lesions and vitreous opacities Key Features
without vitreous cells or anterior uveitis. The lesions did not • Retinopathy is usually bilateral
respond to systemic steroid administration, but later resolved • Veins are dark, dilated, and tortuous; disk is hyperemic
spontaneously.44 • Intraretinal hemorrhages and cotton wool spots are seen
Cotton wool spots and Cytomegalovirus infection of the • Central or branch vein retinal occlusion may be found, as in
retina have also been reported with this virus.40 A compre- polycythemia
hensive review of the ocular findings in HTLV-1 can be found • Neovascularization of the retina or iris and neovascular
in Buggage.45 glaucoma can occur
• May also see serous retinal detachments and Purtscher-like
POLYCYTHEMIA retinopathy
• A paraneoplastic degenerative retinopathy has been described
Foulds2 pointed out that whole-blood viscosity is considerably in Waldenström’s macroglobulinemia
influenced by the number of red cells present. He found that

a b

FIGURE 170.6. (a) Retinal vascular dilatation and tortuosity, with intraretinal hemorrhage, in a case of hyperviscosity due to dysproteinemia.
Right eye. (b) Left eye of the patient in (a). There is disk hyperemia in addition to vascular dilatation, tortuosity, and intraretinal hemorrhage. 2161
RETINA AND VITREOUS

a b
SECTION 10

FIGURE 170.7. (a) Waldenström’s macroglobulinemia with venous dilatation and beading, as well as intraretinal hemorrhage. Right eye. (b) Left
eye of the patient in (a).

Dysproteinemia that results in hyperviscosity of the serum HEMORRHAGIC DISORDERS


from a variety of causes can produce a dramatic retinopathy.47
Carr and Henkind47 reported similar retinal findings in cases of The retina is not typically involved in hemorrhagic disorders
hyperviscosity caused by cryomacroglobulinemia, hypergam- unless there is ocular trauma. Thrombocytopenia is an exception,
maglobulinemia, Waldenström’s macroglobulinemia, chronic particularly if there is accompanying anemia (as described earlier).
lymphoctyic leukemia with macroglobulinemia, and multiple Idiopathic thrombocytopenic purpura rarely results in retino-
myeloma with hyperglobulinemia. Waldenström’s macroglobu- pathy.7 Thrombotic thrombocytopenic purpura may cause retinal
linemia may be the most likely dysproteinemia to produce reti- hemorrhages and serous retinal detachment; the relative roles of
nopathy, possibly because the large size of the protein molecules thrombosis, hypertension, and renal disease in producing the reti-
in this condition lead to very high viscosity levels.1 Holt and nal findings are uncertain.57,58 Fundus appearance compatible
Gordon-Smith,12 however, found no correlation between total with Purtscher’s retinopathy has been reported in patients with
serum protein and retinopathy in their series. Rather, they thrombotic thrombocytopenic purpura.59
thought that the presence of retinopathy correlated with the
degree of anemia found in their patients. They noted that their APPROACH TO THE PATIENT WITH
patients, as well as others reported in the literature with RETINAL HEMORRHAGES OF UNKNOWN
Waldenström’s macroglobulinemia and retinopathy, exhibited CAUSE
severe anemia.
In retinopathy of dysproteinemia, the retinal veins are dark, The patient who presents with retinal hemorrhages can be a diag-
dilated, and tortuous; intraretinal hemorrhages and cotton wool nostic dilemma, as the list of conditions involved in the differ-
spots are seen; and the disk is hyperemic. The retinal hemorrhages ential diagnosis is long. The first task is to decide whether the
may be of the superficial flame or deep punctate type and extend hemorrhages are intraretinal, subretinal, or preretinal. Intraretinal
to the periphery. Retinal edema may occur. Central or branch hemorrhages are of two types: dot and blot, and flame. Dot and
vein retinal occlusion may be found, as in polycythemia. Visual blot hemorrhages are located deep in the retina and are confined
loss may be due to vascular occlusion or retinal edema. Abnor- by the anteroposterior orientation of the rods and cones, bipolar
malities are usually found bilaterally. cells, and Müller’s cells. When viewed end-on through the ophthal-
Retinal microaneurysms may be seen in more chronic cases.48 moscope, they appear as round, red dots or somewhat larger
Exudative retinal detachment has been described in multiple round ‘blots’. Flame hemorrhages are located in the superficial
myeloma.49,50 Neovascularization of the retina or iris with vitreous retina and are confined by the mediolateral, arcing orientation
hemorrhage or neovascular glaucoma can occur. Fibrous of the nerve fiber layer. When viewed with the ophthalmoscope,
proliferation has been described. they are flame shaped, with feathery borders. Flame hemorrhages
Retinopathy may be reversible with treatment of the underlying tend to occur mainly in the posterior portion of the retina. In
disease if frank vein occlusion has not occurred.51 the periphery, hemorrhages appear as dots and blots regardless
More recently a paraneoplastic retinopathy in a patient with of their level in the retina.60 Intraretinal hemorrhages occur in
Waldenström’s macroglobulinemia has been reported with findings a wide variety of disorders, including many systemic diseases and
of deteriorating vision, declining electroretinogram and presence retinal venous occlusions. Subretinal hemorrhages are amorphous
of serum antibodies against photoreceptor proteins.52 in shape and are deep to the retinal vessels. These hemorrhages
Cases of serous detachment of the retinal pigment epithelium occur in trauma or with subretinal neovascularization (e.g., from
and neurosensory retina have been reported in patients with age-related maculopathy). Preretinal hemorrhages may also be
paraproteinemia.53–55 The relationship of the paraproteinemia amorphous, or they may be boat shaped, with a horizontal
to development of these serous leaks is uncertain since patients upper border and a curved lower border (caused by settling of red
were on steroids. cells). These hemorrhages cover retinal vessels. Preretinal hemor-
Purtscher-like retinopathy picture has also been reported in rhages occur with retinal neovascularization (e.g., in diabetic
hepatitis C-associated cryoglobulinemia.56 retinopathy or sickle cell disease), vitreous detachment or retinal
2162
Retinopathy Associated with Blood Anomalies

breaks, and trauma. Occasionally, they are seen in other dis- other disorders. They are, however, a sign of chronicity. Arteriolar
orders (e.g., vein occlusion without neovascularization, leukemia, narrowing and sclerotic changes suggest systemic hypertension.
or subarachnoid hemorrhage). Flame hemorrhages are more frequent in hypertension, with dot
Bilateral intraretinal hemorrhages pose the greatest challenge and blot lesions more common in diabetes, but both lesions
to the differential diagnosis. Unilateral intraretinal hemorrhages occur in either disorder as well as in vein occlusion and retino-
are most frequently due to venous occlusive disease. Bilateral pathy associated with blood disorders. Cotton wool spots or
findings suggest systemic disease as the cause. The distribution white-centered hemorrhages often accompany retinal hemor-
of the hemorrhages may be helpful. If they occur mainly in the rhages, and they may be seen in so many disorders that they are
posterior fundus, systemic disease is likely. Extension to the far of little help in the differential diagnosis. A thorough medical
periphery suggests venous occlusive disease. Confinement to history, general physical examination, complete blood count,
the peripapillary retina suggests optic nerve disease (including and blood glucose determination will reveal the cause of
papilledema). Accompanying findings may be helpful in the bilateral, posterior intraretinal hemorrhages in the majority of
differential diagnosis. Venous dilatation suggests obstructed cases. Study of serum protein levels may be useful when hyper-
flow, which may be due to venous occlusion or hyperviscosity. viscosity is suspected. Fluorescein angiography may be useful in
Microaneurysms are the hallmark of diabetic retinopathy, but demonstrating subtle abnormalities of the retinal and choroidal
they occur with hypertension, venous occlusion, leukemia, and vasculature.

REFERENCES

CHAPTER 170
1. Wise GN, Dollery CT, Henkind P: The retinal systemic and local treatment. Leuk Res 31. Inkeles DM, Friedman AH: Retinal pigment
circulation. New York: Harper & Row; 1971. 2003; 27:557–559. epithelial degeneration, partial retinal
2. Foulds WS: Blood is thicker than water. 17. Miyamoto K, Kashii S, Honda Y: Serous atrophy and macular hole in acute
Some haemorrheological aspects of ocular retinal detachment caused by leukemic lymphocytic leukemia. Graefes Arch Clin
disease. 50th Bowman lecture. Eye 1987; choroidal infiltration during complete Exp Ophthalmol 1975; 194:253.
1:343. remission. Br J Ophthalmol 2000; 32. Verbraak FD, van den Berg W, Bos PJM:
3. Foulds WS: The ocular manifestations of 84:1318–1319. Retinal pigment epitheliopathy in acute
blood diseases. Trans Ophthalmol Soc UK 18. Robb RM, Ervin LD, Sallan SE: A leukemia. Arch Ophthalmol 1991; 111:111.
1963; 83:345. pathologic study of eye involvement in 33. Burns CA, Blodi FC, Williamson BK: Acute
4. Imai E, Kunikata H, Udono T, et al: Branch acute leukemia of childhood. Trans Am lymphocytic leukemia and central serous
retinal artery occlusion: a complication of Ophthalmol Soc 1978; 76:90. retinopathy. Trans Am Acad Ophthalmol
iron-deficiency anemia in a young adult 19. Reddy SC, Jackson N: Retinopathy in Otolaryngol 1965; 69:307.
with a rectal carcinoid. Tohoku J Exp Med acute leukaemia at initial diagnosis: 34. Kuwabara T, Aiello L: Leukemic miliary
2004; 203:141–144. correlation of fundus lesions and nodules in the retina. Arch Ophthalmol
5. Matsuoka Y, Hayasaka S, Yamada K: haematological parameters. Acta 1964; 72:494.
Incomplete occlusion of central retinal Ophthalmol Scand 2004; 82:81–85. 35. Spalton DJ, Nicholson F: HTLV-I infection in
artery in a girl with iron deficiency anemia. 20. Guyer DR, Schachat AP, Vitale S, et al: human disease. Br J Ophthalmol 1991;
Ophthalmologica 1996; 210:358–360. Leukemic retinopathy. Ophthalmology 75:174.
6. Bahar I, Weinberger D, Kramer M, Axer- 1989; 96:860. 36. Nakao K, Ohba N, Isahiki M, et al:
Siegel R: Retinal vasculopathy in Fanconi 21. Karesh JW, Goldman EJ, Reck K, et al: A Pigmentary retinal degeneration in patients
anemia: a case report. Retina 2005; prospective ophthalmic evaluation of with HTLV-1 associated myelopathy. Jpn J
25:799–800. patients with acute myeloid leukemia: Ophthalmol 1989; 33:383.
7. Sandstrom H, Wahlin A, Eriksson M, et al: correlation of ocular and hematologic 37. Ohba N, Nakao K, Isahiki Y: HTLV-1
Angioid streaks are part of a familial findings. J Clin Oncol 1989; 7:1528. associated retinochoroidal degeneration.
syndrome of dyserythropoietic anaemia 22. Duke JR, Wilkinson CP, Sigelman S: Retinal Jpn J Ophthalmol 1996; 40:71.
(CDA III). Br J Haematol 1997; 98:845–849. microaneurysms in leukemia. Br J 38. Fukushima A, Veno H, Fujimoto S: HTLV-1
8. Beare NA, Southern C, Chalira C, et al: Ophthalmol 1968; 52:368. and retinal antigens recognized by T cells.
Prognostic significance and course of 23. Jampol LM, Goldberg MF, Busse B: Nippon Ganka Gakkai Zasshi 1994; 98:157.
retinopathy in children with severe malaria. Peripheral retinal microaneurysms in 39. Kumar SR, Gill PS, Wagner DG, et al:
Arch Ophthalmol 2004; 122:1141–1147. chronic leukemia. Am J Ophthalmol 1975; Human T-cell lymphotrophic virus type
9. Merin S, Freund M: Retinopathy in severe 80:242. 1-associated retinal lymphoma. A
anemia. Am J Ophthalmol 1968; 66:1102. 24. Morse PH, McReady JL: Peripheral retinal clinicopathologic report. Arch Ophthalmol
10. Aisen ML, Bacon BR, Goodman AM, et al: neovascularization in CML. Am J 1994; 112:954.
Retinal abnormalities associated with Ophthalmol 1971; 72:975. 40. Ohba N, Matsumoto M, Sakeshima M,
anemia. Arch Ophthalmol 1983; 101:1049. 25. Schachat AP: The leukemias and et al: Ocular manifestations in patients
11. Rubenstein RA, Yanoff M, Albert DM: lymphomas. In: Ryan SJ, Ogden TE, infected with human T-lymphotrophic
Thrombocytopenia, anemia, and retinal Schachat AP, eds. Retina. St Louis: virus type 1. Jpn J Ophthalmol
hemorrhage. Am J Ophthalmol 1968; 65:435. CV Mosby; 1989. 1989; 33:1.
12. Holt JM, Gordon-Smith EC: Retinal 26. Kincaid MC, Green WR: Ocular and orbital 41. Sasaki K, Morooka I, Inomata H, et al:
abnormalities in diseases of the blood. Br J involvement in leukemia. Surv Ophthalmol Retinal vasculitis in human T-cell
Ophthalmol 1969; 53:145. 1983; 27:211. lymphotrophic virus type I associated
13. Carraro MC, Rossetti L, Gerli GC: 27. Schachat AP, Markowitz JA, Guyer DR, et al: myelopathy. Br J Ophthalmol 1989; 73:812.
Prevalence of retinopathy in patients with Ophthalmic manifestations of leukemia. 42. Nakao K, Matsumoto M, Ohba N:
anemia or thrombocytopenia. Eur J Arch Ophthalmol 1989; 107:697. Seroprevalence of antibodies to HTLV-1 in
Haematol 2001; 67:238–244. 28. Reddy SC, Jackson N, Menon BS: Ocular patients with ocular disorders. Br J
14. Fletcher ME, Farber MD, Cohen SB, et al: involvement in leukemia – a study of 288 Ophthalmol 1991; 75:76.
Retinal abnormalities associated with cases. Ophthalmologica 2003; 217:441–445. 43. Mochizuki M, Watanabe T, Yamaguchi K,
anemia. [letter] Arch Ophthalmol 1984; 29. Clayman HM, Flynn JJ, Koch K, et al: et al: Uveitis associated with human T-cell
102:358. Retinal pigment epithelial abnormalities in lymphotrophic virus type I. Am J
15. Kincaid MC, Green WR, Kelley JS: Acute leukemic disease. Am J Ophthalmol 1972; Ophthalmol 1992; 114:123.
ocular leukemia. Am J Ophthalmol 1979; 74:416. 44. Hayasaka S, Takatori Y, Noda S, et al:
87:698. 30. Jakobiec F, Behrens M: Leukemic retinal Retinal vasculitis in a mother and her son
16. Paydas S, Soylu MB, Disel U, et al: Serous pigment epitheliopathy, with report of a with human T-lymphotrophic virus type 1
retinal detachment in a case with chronic unilateral case. J Pediatr Ophthalmol 1975; associated myelopathy. Br J Ophthalmol
lymphocytic leukemia: no response to 12:10. 1991; 75:566.
2163
RETINA AND VITREOUS

45. Buggage RR: Ocular manifestations of 51. Schwab PJ, Okun E, Fahey JL: Reversal of 56. Myers JP, Di Bisceglie AM, Mann ES:
human T-cell lymphotropic virus type 1 retinopathy in Waldenström’s Cryoglobulinemia associated with
infection. Curr Opin Ophthalmol 2003; macroglobulinemia by plasmapheresis. Purtscher-like retinopathy. Am J
14:420–425. Arch Ophthalmol 1960; 64:515. Ophthalmol 2001; 131:802–804.
46. Rodriguez N, Eliott D: Bilateral central 52. Sen HN, Chan CC, Caruso RC, et al: 57. Lambert SR, High KA, Cotlier E, et al:
retinal vein occlusion in Eisenmenger Waldenstrom’s macroglobulinemia- Serous retinal detachments in thrombotic
syndrome. Am J Ophthalmol 2001; associated retinopathy. Ophthalmology thrombocytopenic purpura. Arch
132:268–269. 2004; 111:535–539. Ophthalmol 1985; 103:1172.
47. Carr RE, Henkind P: Retinal findings with 53. Cohen SM, Kokame GT, Gass JDM: 58. Percival SPB: Ocular findings in thrombotic
serum hyperviscosity. Am J Ophthalmol Paraproteinemias associated with serous thrombocytopenic purpura (Moschcowitz’s
1963; 56:23. detachments of the retinal pigment disease). Br J Ophthalmol 1970; 54:73.
48. Gates RF, Richards RD: Macroglobulinemia epithelium and neurosensory retina. Retina 59. Power MH, Regillo CD, Custis PH:
with unusual vascular changes. Arch 1996; 16:467. Thrombotic thrombocytopenic purpura
Ophthalmol 1960; 77:64. 54. Zamir E, Chowers I: Central serous associated with Purtscher retinopathy. Arch
49. Ashton N: Ocular changes in multiple chorioretinopathy in a patient with Ophthalmol 1997; 115:128.
myelomatosis. Arch Ophthalmol 1965; cryoglobulinaemia. Eye 1999; 60. Ballantyne AJ, Michaelson IC: Textbook of
73:487. 13(Pt 2):265–266. the fundus of the eye. 2nd edn. Baltimore:
50. Franklin RM, Kenyon KR, Green WR, et al: 55. Ogata N, Ida H, Takahashi K, et al: Occult Williams & Wilkins; 1970.
Epibulbar IGA plasmacytoma occurring in retinal pigment epithelial detachment in
multiple myeloma. Arch Ophthalmol 1982; hyperviscosity syndrome. Ophthalmic Surg
100:451. Lasers 2000; 31:248–252.
SECTION 10

2164
CHAPTER

171 Posterior Segment Sarcoidosis


Anita G. Prasad, Daniel Wee, and Russell N. Van Gelder

preponderance up to 2:1, while others report equal incidence


Overview between the genders. The disease is most typically diagnosed in
Sarcoidosis is a multiorgan inflammatory disease of unknown the third decade.2
etiology that is a frequent cause of ocular inflammation. The Several studies have attempted to identify risk factors for
histologic hallmark of disease is the noncaseating granuloma. development of sarcoidosis. The predominant risk factor in the
Posterior segment findings include vitritis, choroiditis, vasculitis, Isle of Man study was living in close proximity to an individual
retinal ischemia, and retinal neovascularization. The differential
with known sarcoidosis.3–6 Husband–wife pairs with disease have
diagnosis for sarcoidosis is broad; definitive diagnosis requires
biopsy, although certain laboratory tests such as CT scanning,
been identified frequently. There is a familial association in
gallium scanning, and ACEs have adjunctive diagnostic value. first- and second-degree blood relatives as well, although this
Treatment requires corticosteroid medications and/or appears stronger in Caucasian cases than in African-American
immunomodulation, and frequently requires systemic treatment, cases.7 Nurses have a sevenfold higher lifetime risk of sarcoidosis
particularly if organs besides the eye are involved. than the average population.8 These findings suggest a trans-
missible agent responsible for at least triggering, if not causing,
chronic disease. Consistent with this, organ transplantation
studies have suggested that the disease may be transferred with
Key Features diseased organs.9–11 In one study, an individual with a known
• Vitritis history of sarcoidosis served as a bone marrow donor for his
• Choroiditis brother, who did not have disease. Despite aggressive immuno-
• Vasculitis modulation post-transplant, the sibling developed sarcoidosis
• Retinal ischemia within several weeks of the transplant. Sarcoidosis has also
• Retinal neovascularization been associated with cardiac transplantation from an affected
donor. Interestingly, sarcoid-positive recipients typically redevelop
disease in naïve organs.12 The estimated rate of redevelopment
Sarcoidosis is a chronic, multiorgan inflammatory disease of of pulmonary sarcoidosis in lung transplant recipients is close
unknown etiology. Sarcoidosis can be a very frustrating disease to 80%.
for the patient and clinician alike. The pathogenesis of the Several candidate pathogens have been proposed as causative
disease is largely unknown, risk factors for its acquisition are for sarcoidosis. L-form mycobacteria are cell wall-less bacteria
not well understood, its clinical manifestations are protean, that have been found in a subset of lymph nodes affected with
clinical diagnosis is challenging, treatments are nonspecific, sarcoidosis13; myobacterial DNA has been detected sporadically
and prognosis is variable. As sarcoidosis is a relatively common in lymph nodes from confirmed sarcoidosis cases as well.14,15
cause of uveitis (in both the anterior and posterior segment), it Other studies have suggested an association with human herpes
belongs on the differential diagnosis in many cases. It is virus 8 (HHV-8) with sarcoidosis in Europe,16 although this asso-
important for the clinician to be aware of the varied clinical ciation has not been confirmed in several other populations.17,18
appearances of sarcoidosis and to be facile with the diagnostic Most recently, members of the genus Propionibacterium, particu-
workup and management of this disease. larly Propionibacterium granulosum, have been found with high
frequency in lymph node biopsies of patients with proven sar-
EPIDEMIOLOGY coidosis.15,19 To date, however, Koch's postulates have not been
fulfilled for any infectious organism in sarcoidosis.
The incidence and prevalence of sarcoidosis vary markedly with In addition to sporadic cases, familial variants of sarcoidosis
geographic location. Because strict diagnostic criteria are not (with juvenile onset) are well documented. Blau syndrome is a
always followed in epidemiologic studies of sarcoidosis, accurate multisystem granulomatous disease presenting in childhood.
estimates of disease rates are not available. Older data from the Unlike sarcoidosis, Blau syndrome typically spares the lungs; how-
United States suggest an annual incidence in African-Americans of ever, uveitis can be a presenting complaint and the clinical appear-
82 per 100 000 person-years, compared with 8 per 100 000 person- ance of Blau syndrome can be identical to sarcoidosis. Patients
years in the Caucasian population.1 It is believed that Latino- will often present with juvenile arthritis. Recently, Blau syndrome
Americans are more commonly affected than Caucasians. The has been shown to be due to mutations in CARD15/NOD2, a
disease is also common in northern Europe, with an incidence gene involved in regulation of inflammation and apoptosis (and
of 20 per 100 000 in the United Kingdom, and 24 per 100 000 also associated with Crohns’ disease).20,21 However, sarcoidosis
in Sweden.2 There is no firm consensus on the relative rates of (and in particular the uveitis associated with sarcoidosis) does
disease between the genders; some studies have suggested a female not appear to be associated with these mutations.22,23 2165
RETINA AND VITREOUS

them to make a diagnosis; biopsy will typically demonstrate


septal panniculitis.
Central nervous system sarcoidosis occurs in ~5% of patients
with systemic sarcoidosis, and is most commonly associated with
cranial neuropathy, which may be multiple or bilateral. Facial
nerve involvement is most common. Optic neuropathy occurs in
~3% of patients with systemic sarcoidosis. Rarely, more gener-
alized aseptic meningitis may be the presenting sign of sarcoidosis.
In addition to these chronic presentations of sarcoidosis, the
disease can occasionally present acutely. Lofrgren syndrome is
characterized by acute fever, arthralgias, erythema nodosum, and
bilateral hilar adenopathy. Heerfordt–Waldenström syndrome
(also called uveo-parotid fever) is the combination of fever, parotid
enlargement, and uveitis.

RETINAL INVOLVEMENT
In most studies, the eye is reported to be involved in ~20–25%
SECTION 10

FIGURE 171.1. Sarcoid granulomas in the choroids from an


enucleated eye. of cases of sarcoidosis,25 although some studies suggest rates as
high as 60%. Of those patients with ocular involvement, posterior
segment disease is thought to occur in ~25%.26 Sarcoidosis is
HISTOPATHOLOGY thought to be responsible for between 4% and 8% of uveitis cases
in referral centers.27,28 Ocular involvement may be the initial
The defining feature of sarcoidosis is the formation of the non- manifestation of sarcoidosis, and patients may present with
caseating granuloma (Fig. 171.1). These granulomas may be found classic uveitic symptoms of redness, pain, photophobia, floaters,
in nearly any organ of the body, but typically target the lungs, or scotomas, or they may be asymptomatic.29,30 There are no
thoracic lymph nodes, skin, and eyes. The granulomas are pre- specific signs unique to the eye; the disease can present in the
dominantly composed of epithelioid multinucleated giant cells, anterior segment, as intermediate uveitis, in the posterior pole,
which are essentially aggregated macrophages. The predominant or as panuveitis. Although posterior segment disease is typically
lymphocytic cell type is the CD4+ T-cell, which is found in the associated with anterior segment inflammation, retinal findings
periphery of the granuloma. Smaller numbers of CD8+ T-cells may occur in isolation. Anterior segment disease can present as
and B-cells are also present within the granuloma. The histo- nongranulomatous or granulomatous inflammation. The latter
pathology is consistent with an exaggerated immune response is typified by large, ‘mutton fat’ keratic precipitates as well as iris
in target organs. The predominant inflammatory response is nodules. Posterior synechia are commonly seen. The anterior
Th1 mediated, with a preponderance of interferon gamma and segment can show significant ciliary flush or conjunctival injec-
interleukin-2, as well as production of tumor necrosis factor tion; it can also be relatively quiet even with active anterior
and interleukin-6 by macrophages. Progression of granulomas segment disease.
culminates in tissue damage, fibrosis, and end organ destruction. The most common posterior segment findings in sarcoidosis are
The noncaseating granuloma is not specific for sarcoidosis, vitritis, choroidal punched-out lesions, snowball lesions, cystoid
however. Noncaseating granulomas can also be seen in response macular edema, periphlebitis, and epiretinal membrane.29 In cases
to hypersensitivity pneumonitis (including berylliosis, asbestosis, associated with ischemia, neovascularization can develop. Isolated
or silicosis), mycobacterial infections, or fungal infection. Diag- or optic nerve granulomas are also typical of the disease.
nosis is considered definitive when the noncaseating granuloma The intermediate uveitis associated with sarcoidosis can be
is seen in the setting of characteristic clinical findings. isolated, or can be associated with punched-out choroidal lesions
or retinal granulomas. Approximately 10% of sarcoidosis presents
SYSTEMIC CLINICAL FINDINGS as an isolated intermediate uveitis,31 and this entity must be
considered in the differential diagnosis of isolated intermediate
Sarcoidosis can affect nearly any organ in the body. The lung is uveitis.
the most frequently involved organ, with greater than 90% of The choroidal lesions are often the principal posterior segment
patients affected. The next most commonly affected organs finding in sarcoidosis. The lesions may be large, isolated granu-
include the lymph nodes, skin, eyes, and liver, each affected in lomas (Fig. 171.2), or can be small, clustered yellowish lesions
approximately one-quarter of patients.24 Pulmonary findings range (Fig. 171.3). The lesions are most commonly found in the
from asymptomatic discovery on routine chest radiograph, to inferior equatorial retina. By themselves, these lesions rarely lead
severe pulmonary fibrosis and restrictive lung disease. A stand- to visual dysfunction unless they are localized to the macula. They
ardized staging scheme is employed in the reading of chest radio- may be associated, however, with panuveitis, vitritis, or cystoid
graphs of patients with sarcoidosis. Bilateral hilar adenopathy macular edema. Angiographically, the lesions will typically show
constitutes stage 1 disease. Parenchymal involvement with hilar early blockade of fluorescence, with late staining, typical of
adenopathy defines stage 2; in stage 3 disease, parenchymal inflammatory lesions in the posterior pole. Resolved lesions
involvement is seen without hilar adenopathy. In stage 4 disease, may show only RPE window defects (Fig. 171.4). With treatment,
pulmonary fibrosis is seen, which may be accompanied by choroidal granulomas secondary to sarcoidosis can resolve com-
bronchiectasis. pletely, without pigment epithelial change; however, chronic
The skin is involved in an estimated 18–32% of cases. The lesions may show evidence of scarring (Fig. 171.4). Rarely,
most typical skin lesion is erythema nodosum. These are typi- sarcoidosis lesions may resemble serpiginous choroiditis lesions.32
cally elevated red lesions often found on the lower extremity Retinal vasculitis may be the predominant finding in sarcoi-
(such as the shins). These are a nonspecific finding and can be dosis involving retina. Vasculitis is typically limited to the venous
identified in a number of hypersensitivity conditions. Since these system, with sparing of the arteries, although arterial involvement
2166 lesions do not contain granulomas, it is not worthwhile to biopsy has been reported. The location can be peripheral, or can be in
Posterior Segment Sarcoidosis

the posterior pole (Fig. 171.5). Extensive perivascular exudates


are called candle wax drippings or ‘taches de bougies’. In severe
cases, the vasculitis can lead to branch retinal vein occlusions
which, in turn, may lead to ischemia and neovascularization.
Complications from ocular manifestations of posterior segment
sarcoidosis are common. Cystoid macular edema has been noted
in approximately three-fourths of patients with posterior seg-
ment sarcoidosis, cataract in about half, and glaucoma in about
one-third. Retinal ischemia and neovascularization are reported
to occur in 16% and 11%, respectively.29

DIFFERENTIAL DIAGNOSIS
The differential diagnosis for posterior segment inflammation
containing features seen in sarcoidosis is remarkably broad. Iso-
lated intermediate uveitis may be due to Lyme disease, syphilis,
tuberculosis, pars planitis, multiple sclerosis, or intraocular
lymphoma. Disease associated with retinal or choroidal granu-

CHAPTER 171
lomas has a similar differential diagnosis, but also includes bird-
shot chorioretinopathy, multifocal choroiditis with panuveitis,
acute posterior multifocal placoid pigment epitheliopathy
(APMPPE), cat scratch disease, toxoplasmosis, toxocariasis,
FIGURE 171.2. Resolving solitary choroidal granuloma in a patient Vogt–Koyanagi–Harada syndrome, sympathetic ophthalmia, bru-
with sarcoidosis.
cellosis, Whipple's disease, and ocular candidiasis. With marked
phlebitis, collagen vascular diseases (including Wegener’s granulo-
matosis, systemic lupus erythematosus, Behcet’s disease, Eales'
disease, and frosted branch angiitis) should also be considered. If
the patient is immunocompromised, cytomegalovirus (CMV)
retinitis and other HIV-associated diseases must be considered.

LABORATORY TESTING
Unfortunately, there is no highly specific and sensitive labora-
tory test for the diagnosis of sarcoidosis, except for tissue
biopsy. It is always worth examining the conjunctiva carefully
for the presence of granulomas, which have been reported to
occur in as many as 40% of sarcoidosis patients.26 In older
patients with either chest radiograph abnormalities or elevated
angiotensin converting enzyme (ACE) levels, the yield on
nondirected conjunctival biopsy may exceed 50%33; in other
populations, the yield is lower.34 Probably, the most effective
means for finding tissue to make a definitive biopsy is
performing CT scan of the chest; in one recent series of elderly
women with uveitis typical for either sarcoidosis or lymphoma,
FIGURE 171.3. Multiple small granulomas associated with 57% had hilar adenopathy on CT scan, and 82% of these were
sarcoidosis. found to have sarcoidosis on biopsy or bronchoscopy. However,

a b

FIGURE 171.4. (a,b) Inactive peripheral choroidal lesions with window retinal pigment epithelial defects secondary to sarcoidosis. 2167
RETINA AND VITREOUS

threatening conditions with similar clinical appearances are ruled


out. This will typically involve placing a PPD and obtaining a
chest radiograph for evaluation of tuberculosis, testing FTA-ABS
and/or RPR for syphilis, and possibly other serologic or blood tests
for specific infections such as Lyme disease, toxoplasmosis, or
toxocariasis. A careful examination for conjunctival granulomas
or enlarged lacrimal gland should be performed; if found, this
may be the easiest site for biopsy. If this workup is negative,
consideration may be given to chest CT to identify hilar adeno-
pathy; alternatively, ACE and gallium scanning may be per-
formed (although the negative predictive value for such testing
may be limited). In the setting of abnormal ACE or gallium scan,
a nondirected conjunctival biopsy may be acquired.
If a diagnosis of sarcoidosis is made in the setting of positive
chest radiograph, positive chest CT, or positive review of systems
for shortness of breath or chronic cough, consideration should
be given to obtaining pulmonary function tests. Given a positive
workup, a complete blood count and comprehensive metabolic
SECTION 10

FIGURE 171.5. Circinate subretinal granulomas with focal panel including hepatic and renal function tests should be
periphlebitis (taches du bougie).
obtained, both to seek possible signs of systemic sarcoidosis and
to help tailor therapy.
before commencing with this test, one should consider carefully Specific ocular testing may be required to fully characterize
whether one is prepared to follow the CT with bronchoscopy or the ophthalmic manifestations of disease. Fluorescein angiogra-
mediastinoscopy; in many cases, it may not be essential to phy and optical coherence tomography may be of great value in
make a definitive diagnosis in order to guide therapy. characterizing and following cystoid macular edema. Angiography
The ACE test measures serum levels of the hydrolase enzyme can also indicate areas of occult vasculitis, and can demonstrate
involved in blood pressure regulation. It is present on the optic nerve head leakage. ICG angiography reveals several potential
luminal surface of vascular endothelial cells, as well as in cells patterns of choroidal disease, including focal hypo- and hyper-
of the macrophage–monocyte system. The ACE level is thought fluorescence and late choroidal vessel leakage which are relatively
to be a measure of total granuloma load on the body. The test, nonspecific but may be useful in following disease activity.43,44
unfortunately, is neither highly sensitive nor highly specific.
Sensitivity varies in different studies between approximately TREATMENT
40% and 80%.35–38 The specificity is somewhat higher, and has
been estimated to be as high as 90%. This yields a positive
predictive value for an elevated ACE level of ~84–90%. However, Treatment Options
negative predictive values are very poor. In addition, patients • Local
who are taking ACE inhibitors will have depressed levels, as will • Topical corticosteroids (i.e., prednisolone acetate)
patients who are already on anti-inflammatory (such as corti- • Periocular corticosteroids (i.e., triamcinolone acetonide)
costeroid) treatment. Other diseases to consider in the setting • Cycloplegics (homatropine 5%, scopolamine 0.25%)
of elevated ACE level include miliary tuberculosis, atypical • Systemic
mycobacterial infections, asbestosis and silicosis, primary biliary • Corticosteroids
cirrhosis, and hyperthyroidism. • Methotrexate
A number of other tests have been advocated for the diagnosis • Azathioprine
of sarcoidosis, including serum lysozyme, serum calcium, and • Cyclosporin A
cutaneous anergy. All suffer from low sensitivity and specificity. • Infliximab
Human Leukocyte Antigen (HLA) typing is not useful in the • Adalimumab
diagnosis of sarcoidosis, although weak disease susceptibility has • Alkylating agents (cyclophosphamide, chlorambucil)
been associated with HLA-B8 and HLA-DRB1.39–41 Gallium
scanning measures the uptake of gallium-67, which localizes to
areas of active inflammation. This may be useful in distinguish- In initiating therapy for posterior segment sarcoidosis, the primary
ing lymphoma from sarcoidosis; in the former, peripheral lymph consideration is whether the patient has systemic involvement
node uptake is increased, while in the latter bilateral hilar uptake warranting treatment; this will dictate whether the first approach
is noted. The finding of increased uptake in the lacrimal and must include systemic medication. Typically, pulmonologists
parotid glands (the panda sign) has been seen between 60% and will not treat stage 1 disease but will treat stage 2 or higher dis-
87% of the time in sarcoidosis patients.35,36 Specificity is ease. The mainstay of treatment of sarcoidosis is corticosteroid
relatively high. The combination of a positive gallium scan and therapy; for posterior segment disease, the treatment options
elevated ACE level has high specificity (>90%) for sarcoidosis; include systemic administration, periocular injection, and intra-
however, this does not serve as a substitute for tissue biopsy for ocular administration. For bilateral or sight-threatening disease,
definitive diagnosis. systemic corticosteroids are frequently employed, unless the
The older literature refers to the Kveim–Siltzbach reaction.42 patient has absolute contraindications to this treatment (such as
In this test, intradermal injection of a tissue suspension derived brittle diabetes or severe psychiatric disease). Cycloplegia should
from the spleen of a sarcoidosis patient results in local develop- be employed to reduce the risk of posterior synechia formation.
ment of sarcoid granuloma. The lesions typically form 2–6 weeks With rare exceptions, posterior segment sarcoidosis will not
after injection. The inciting antigen in this preparation is respond to topical medication; however, topical corticosteroids are
unknown. The test is not used in routine clinical settings. a mainstay of treatment for the anterior segment inflammation
In cases where sarcoidosis is clinically suspected, the following that often accompanies posterior segment inflammation. Peri-
2168 approach may be considered. First, common or medically ocular corticosteroids (typically triamcinolone acetonide admin-
Posterior Segment Sarcoidosis

istered 20–40 mg either applied beneath Tenon’s capsule or 2 years, and may be appropriate for patients with chronic pos-
retroseptally through inferior or superior approaches) offer the terior segment inflammation.47,48
benefits of sustained continuous release and high intraocular con- As sarcoidosis is typically a chronic condition, patients will
centrations. However, this approach is associated with iatrogenic frequently require long-term treatment, which may require
cataract formation, steroid responsive ocular hypertension, cos- initiation of treatment with steroid sparing agents. All classes of
metic concerns, and rarely injury to the globe or its circulation. immunomodulatory drugs, including antimetabolites (methotrex-
Recently, intravitreal corticosteroid administration has been used ate, mycophenolate mofetil, and azathioprine), anti-T-cell activa-
in small numbers of patients for the treatment of refractory tion factors (cyclosporine A, tacrolimus), biologics (infliximab and
cystoid macular edema and isolated choroidal granulomas asso- adalimumab), and alkylating agents (cyclophosphamide and
ciated with sarcoidosis, with encouraging short-term results.45,46 chlorambucil), have been utilized.49–51 Unfortunately, no well-
The recently approved fluocinolone acetonide intravitreal implant controlled prospective clinical trials on the efficacy of these treat-
releases a constant dose of corticosteroid intraocularly for over ments has been performed to date.

REFERENCES
1. Bresnitz EA, Strom BL: Epidemiology 16. Di Alberti L, Porter SR, Piatelli A, et al: 32. Edelsten C, Stanford MR, Graham EM:
of sarcoidosis. Epidemiol Rev 1983; Human herpesvirus 8 and sarcoidosis. Serpiginous choroiditis: an unusual
5:124–156. Lancet 1998; 351:1589–1590. presentation of ocular sarcoidosis. Br J

CHAPTER 171
2. Milman N, Selroos O: Pulmonary 17. Knoell KA, Hendrix JD, Jr., Stoler MH, et al: Ophthalmol 1994; 78:70–71.
sarcoidosis in the Nordic countries Absence of human herpesvirus 8 in 33. Hershey JM, Pulido JS, Folberg R, et al:
1950–1982. Epidemiology and clinical sarcoidosis and crohn disease granulomas. Non-caseating conjunctival granulomas in
picture. Sarcoidosis 1990; 7:50–57. Arch Dermatol 2005; 141:909–910. patients with multifocal choroiditis and
3. Ayres JG: Epidemiology of sarcoidosis in 18. Maeda H, Niimi T, Sato S, et al: Human panuveitis. Ophthalmology 1994;
the Isle of Man. Thorax 1987; 42:911–911. herpesvirus 8 is not associated with 101:596–601.
4. Hills SE, Parkes SA, Baker SB: sarcoidosis in Japanese patients. Chest 34. Spaide RF, Ward DL: Conjunctival biopsy in
Epidemiology of sarcoidosis in the Isle of 2000; 118:923–927. the diagnosis of sarcoidosis. Br J
Man. 2. Evidence for space-time clustering. 19. Hiramatsu J, Kataoka M, Nakata Y, et al: Ophthalmol 1990; 74:469–471.
Thorax 1987; 42:427–430. Propionibacterium acnes DNA detected in 35. Power WJ, Neves RA, Rodriguez A, et al:
5. Parkes SA, Baker SB, Bourdillon RE, et al: bronchoalveolar lavage cells from patients The value of combined serum angiotensin-
Epidemiology of sarcoidosis in the Isle of with sarcoidosis. Sarcoidosis Vasc Diffuse converting enzyme and gallium scan in
Man. 1. A case controlled study. Thorax Lung Dis 2003; 20:197–203. diagnosing ocular sarcoidosis.
1987; 42:420–426. 20. Wang X, Kuivaniemi H, Bonavita G, et al: Ophthalmology 1995; 102:2007–2011.
6. Parkes SA, Baker SB, Bourdillon RE, et al: CARD15 mutations in familial 36. Baughman RP, Shipley R, Eisentrout CE:
Incidence of sarcoidosis in the Isle of Man. granulomatosis syndromes: a study of the Predictive value of gallium scan,
Thorax 1985; 40:284–287. original Blau syndrome kindred and other angiotensin-converting enzyme level, and
7. Rybicki BA, Iannuzzi MC, Frederick MM, families with large-vessel arteritis and bronchoalveolar lavage in two-year follow-
et al: ACCESS Research Group Familial cranial neuropathy. Arthritis Rheum 2002; up of pulmonary sarcoidosis. Lung 1987;
aggregation of sarcoidosis. A case-control 46:3041–3045. 165:371–377.
etiologic study of sarcoidosis (ACCESS). 21. Miceli-Richard C, Lesage S, Rybojad M, 37. Lieberman J, Sastre A: An angiotensin-
Am J Respir Crit Care Med 2001; et al: CARD15 mutations in Blau syndrome. converting enzyme (ACE) inhibitor in human
164:2085–2091. Nat Genet 2001; 29:19–20. serum. Increased sensitivity of the serum
8. Edmondstone WM: Sarcoidosis in nurses: 22. Schurmann M, Valentonyte R, Hampe J, ACE assay for detecting active sarcoidosis.
is there an association? Thorax 1988; et al: CARD15 gene mutations in Chest 1986; 90:869–875.
43:342–343. sarcoidosis. Eur Respir J 2003; 22:748–754. 38. Klech H, Kohn H, Kummer F, Mostbeck A:
9. Burke WM, Keogh A, Maloney PJ, et al: 23. Martin TM, Doyle TM, Smith JR, et al: Assessment of activity in sarcoidosis.
Transmission of sarcoidosis via cardiac Uveitis in patients with sarcoidosis is not Sensitivity and specificity of 67gallium
transplantation. Lancet 1990; associated with mutations in NOD2 scintigraphy, serum ACE levels, chest
336:1579–1579. (CARD15). Am J Ophthalmol 2003; roentgenography, and blood lymphocyte
10. Heyll A, Mecjenstock G, Aul C, et al: 136:933–935. subpopulations. Chest 1982; 82:732–738.
Possible transmission of sarcoidosis via 24. Siltzbach LE, James DG, Neville E, et al: 39. Dubaniewicz A, Szczerkowska Z, Hoppe A:
allogeneic bone marrow transplantation. Course and prognosis of sarcoidosis Comparative analysis of HLA class I
Bone Marrow Transplant 1994; 14:161–164. around the world. Am J Med 1974; antigens in pulmonary sarcoidosis and
11. Padilla ML, Schilero GJ, Teirstein AS: 57:847–852. tuberculosis in the same ethnic group.
Donor-acquired sarcoidosis. Sarcoidosis 25. Jabs DA, Johns CJ: Ocular involvement in Mayo Clin Proc 2003; 78:436–442.
Vasc Diffuse Lung Dis 2002; 19:18–24. chronic sarcoidosis. Am J Ophthalmol 40. Hedfors E, Lindstrom F: HLA-B8/DR3 in
12. Muller C, Briege J, Haller M, Vogelmeier C, 1986; 102:297–301. sarcoidosis. Correlation to acute onset
Bittman I, Welz A Furst H, Dienemann H, 26. Obenauf CD, Shaw HE, Sydnor CF, disease with arthritis. Tissue Antigens
et al: Sarcoidosis recurrence following lung Klintworth GK: Sarcoidosis and its 1983; 22:200–203.
transplantation. Transplantation 1996; ophthalmic manifestations. Am J 41. Olenchock SA, Heise ER, Marx JJ Jr, et al:
61:1117–1119. Ophthalmol 1978; 86:648–655. HLA-B8 in sarcoidosis. Ann Allergy 1981;
13. Cantwell AR, Jr.: Histologic observations of 27. Rodriguez A, Calonge M, Pedroza-Seres M, 47:151–153.
variably acid-fast pleomorphic bacteria in et al: Referral patterns of uveitis in a tertiary 42. Teirstein AS: Kveim antigen: what does it
systemic sarcoidosis: a report of 3 cases. eye care center. Arch Ophthalmol 1996; tell us about causation of sarcoidosis?
Growth 1982; 46:113–125. 114:593–599. Semin Respir Infect 1998; 13:206–211.
14. Ikonomopoulos JA, Gourgoulis VG, 28. Perkins ES: Epidemiology of uveitis. Trans 43. Matsuo T, Itami M, Shiraga F:
Zacharatos PV, et al: Multiplex polymerase Ophthalmol Soc UK 1976; 96:105–107. Choroidopathy in patients with sarcoidosis
chain reaction for the detection of 29. Rothova A: Ocular involvement in observed by simultaneous indocyanine
mycobacterial DNA in cases of tuberculosis sarcoidosis. Br J Ophthalmol 2000; green and fluorescein angiography. Retina
and sarcoidosis. Mod Pathol 1999; 84:110–116. 2000; 20:16–21.
12:854–862. 30. Rothova A, Alberts C, Glasius E, et al: Risk 44. Wolfensberger TJ, Herbort CP: Indocyanine
15. Ishige I, Usui Y, Takemura T, Eishi Y: factors for ocular sarcoidosis. Doc green angiographic features in ocular
Quantitative PCR of mycobacterial and Ophthalmol 1989; 72:287–296. sarcoidosis. Ophthalmology 1999;
propionibacterial DNA in lymph nodes of 31. Graham EM, Edelsten C: Intermediate 106:285–289.
Japanese patients with sarcoidosis. Lancet uveitis and sarcoidosis. Dev Ophthalmol 45. Larsson J, Hvarfner C, Skarin A: Intravitreal
1999; 354:120–123. 1992; 23:106–110. triamcinolone in two patients with refractory 2169
RETINA AND VITREOUS

macular oedema in sarcoid uveitis. Acta 48. Jaffe GJ, Martin D, Callanan D, et al: 50. Callejas-Rubio JL, Ortego-Centeno N,
Ophthalmol Scand 2005; 83:618–619. Fluocinolone Acetonide Uveitis Study Lopez-Perez L, Benticuaga MN: Treatment
46. Chan WM, Lim E, Liu DT, et al: Intravitreal Group Fluocinolone acetonide implant of therapy-resistant sarcoidosis with
triamcinolone acetonide for choroidal (Retisert) for noninfectious posterior uveitis adalimumab. Clin Rheumatol 2006;
granuloma in sarcoidosis. Am J Ophthalmol thirty-four-week results of a multicenter 25:596–7.
2005; 139:1116–1118. randomized clinical study. Ophthalmology 51. Sweiss NJ, Welsch MJ, Curran JJ,
47. Jaffe GJ, Ben-Nun J, Guo H, et al: 2006; 113:1028–1034. Ellman MH: Tumor necrosis factor inhibition
Fluocinolone acetonide sustained drug 49. Baughman RP, Lower EE: Steroid-sparing as a novel treatment for refractory
delivery device to treat severe uveitis. alternative treatments for sarcoidosis. Clin sarcoidosis. Arthritis Rheum 2005;
Ophthalmology 2000; 107:2024–2033. Chest Med 1997; 18:853–864. 53:788–791.
SECTION 10

2170
CHAPTER

172 Sickle-Cell Retinopathy


Fina C. Barouch, George K. Asdourian, and John I. Loewenstein

Sickling hemoglobinopathies are caused by the presence of one retina. The constellation of retinal abnormalities observed in
or a combination of abnormal hemoglobins in red blood cells. these patients constitutes the retinopathy of sickle cell.4,14–24
Normal hemoglobin, which is referred to as hemoglobin A The posterior segment abnormalities can be divided into the
(HbA), is composed of two a-peptide and two b-peptide chains.1 following categories:
A mutation can lead to a single amino acid substitution in the • Optic disk changes
sixth position of the b-chain (valine for glutamic acid), • Posterior retinal and macular vascular occlusions
producing hemoglobin S (HbS), whereas a different substitution • Chronic macular changes
at the same position (lysine for glutamic acid) will produce • Choroidal vascular occlusions
hemoglobin C (HbC). The various hemoglobins can occur in • Nonproliferative retinal changes
combinations resulting in hemoglobins such as hemoglobin AS • Proliferative retinal changes
(sickle-cell trait), hemoglobin SS (sickle-cell disease or anemia),
hemoglobin SC (sickle-cell hemoglobin C disease), and others. OPTIC DISK CHANGES
A mutation can also induce failure or inadequate production of
one of the two peptide chains (a or b) (a- and b-thalassemia), Similar to vascular occlusions elsewhere, the small vessels on
which in combination with sickle hemoglobin, leads to sickle- the surface of the optic disk can exhibit intravascular occlusions
cell thalassemia (S-thal) disease. Sickle cell is the most common (disk sign of sickling).13 These occlusions are seen ophthal-
hemoglobinopathy affecting humans, with ~8% of African– moscopically as dark red intravascular spots (Fig. 172.1) and
Americans having the gene for HbS. Other less common geno- probably represent plugs of deoxygenated erythrocytes occurring
types of sickle-cell disease have also been described. in the small surface vessels of the disk. Fluorescein angiography
Normal red blood cells are round or oval, flexible, and can (Fig. 172.2) shows linear or Y-shaped segments of hypofluores-
squeeze through capillaries. Under conditions of hypoxia and cence corresponding to these red spots but does not reveal any
other metabolic conditions (pH, temperature), the red blood substantial impairment of blood flow in these vessels.
cells containing HbS become rigid and adopt an elongated These occlusions are transient and do not produce clinically
sickle-shaped configuration.1 This occurs because of the detectable visual impairment. Although disk changes have been
formation of intracellular aggregates of long polymers in a found in patients with hemoglobin SS, hemoglobin SC, and
crystalline gel. With recurring sickling, the membranes of the hemoglobin S-thal diseases, they are most common in patients
red blood cells become damaged and can no longer assume a with hemoglobin SS disease (29%).13
normal configuration on reoxygenation and become irreversibly
sickled. As the sickle cells increase in number in the circulation, Key Features
they increase the viscosity of the blood and lead to sluggish • Disk sign of sickling,
blood flow, erythrocytic aggregation, increased adhesion to the • vascular occlusions,
vascular endothelium, and eventual vasoocclusion of the vessel. • “salmon patch” hemorrhages,
In the fundus, the peripheral retina and the macula are the • “black sunbursts,”
areas most susceptible for vascular occlusions. The vascular • “iridescent spots,”
occlusions usually occur in the arterioles, especially in patients • neovascularization,
with a large number of irreversibly sickled cells, since these rigid • vitreous hemorrhage
red blood cells cannot enter the capillaries. Using fluorescein
angiography, prolonged transition times of blood have been well
documented in the peripheral retina of sickle-cell patients.2,3
The systemic and ocular manifestations of the different Optic disk neovascularization is uncommon in patients with
hemoglobinopathies do not go hand in hand. Although patients sickle-cell retinopathy but has been documented in patients with
with hemoglobin SS disease manifest the worst systemic symp- SC disease, hemoglobin SS, and hemoglobin SA.25,26 Scatter
toms, patients with hemoglobin SC and hemoglobin S-thal peripheral retinal photocoagulation has been found to be effec-
hemoglobinopathies exhibit the most severe ocular complica- tive in stimulating regression of disk neovascularization.25
tions. The reason for this discrepancy is poorly understood.
The sickling process can produce vasoocclusion and second- POSTERIOR RETINAL AND MACULAR
ary tissue changes in all the vascular structures of the eye, VASCULAR OCCLUSIONS
including the conjunctiva (conjunctival sickling sign),4–6 the iris
(iris atrophy and neovascularization),7,8 the choroid (occlusion Acute major retinal vascular occlusions – central and branch
of the posterior ciliary vessels),9–12 the optic disk,13 and the retinal artery occlusions and peripapillary and macular arte- 2171
RETINA AND VITREOUS

riolar occlusions – although infrequent, have been reported


(Fig. 172.3).27–36 These occlusions occur more commonly in
patients with hemoglobin SS disease, although their occurrence
in various other sickling genotypes has been reported. The occlu-
sions can occur in one eye or simultaneously in both eyes and
result in complete loss of vision or debilitating central and para-
central scotoma. Arterial occlusions in sickle-cell patients have
also been reported as a complication of retrobulbar anesthesia,29,37
after ocular compression during photocoagulation,38 during air-
plane travel,18 after trauma,39 and with extreme dehydration.11
Retinal venous occlusions – central and branch retinal vein
occlusions – are very uncommon in patients with sickle-cell
disease and one should suspect other underlying causes for their
occurrence in these patients.

CHRONIC MACULAR CHANGES (SICKLING


FIGURE 172.1. The disk sign of sickling. Blocked small vessels are MACULOPATHY)
SECTION 10

seen as dark spots or lines.


From Goldbaum MH, Jampol LM, Goldberg MF: The disk sign in sickling
Apart from the acute vascular occlusions of the macular region,
hemoglobinopathies. Arch Ophthalmol 1978; 96:1597. Copyright 1978, American chronic vascular changes of the posterior pole have been
Medical Association. reported in patients with hemoglobin SS, hemoglobin SC,
hemoglobin S-thal, and hemoglobin AS diseases. They occur in
~30% of patients with sickle-cell disease.19–21,40–43 These
occlusions are insidious, progressive, and clinically difficult to
detect. They consist of alterations in the normal architecture of
FIGURE 172.2.
Fluorescein the fine vasculature of the macula, perimacular region, and
angiography of the region of the horizontal raphe lying temporal to the macula.
optic disk shows They may be transient with a continuous remodeling of the
several plugged macular vasculature. Careful direct ophthalmoscopy, contact
vessels (arrows). lens examination, and fluorescein angiography are necessary to
From Goldbaum MH, detect these abnormalities. Microaneurysms, dark and enlarged
Jampol LM, Goldberg MF: segments of terminal arterioles representing occluded
The disk sign in sickling precapillary arterioles, hairpin-shaped vascular loops, an
hemoglobinopathies. Arch
abnormal foveal avascular zone (FAZ) with adjacent areas of
Ophthalmol 1978;
96:1599. Copyright 1978,
capillary nonperfusion (pathologic avascular zones (PAZs)), and
American Medical areas of retinal depression (Figs 172.4 and 172.5) may be seen.44
Association. Sanders and co-workers found the mean largest diameter of the
FAZ in patients with hemoglobin SC and hemoglobin SS disease
to be 1.0 mm compared with 0.61 mm in normal subjects.40
However, there was no significant difference in the FAZ
diameters within the sickle-cell group in regard to the degree of
retinopathy, type of sickle-cell disease, or the visual acuity. The
areas of retinal depression are best seen by direct ophthal-
moscopy and are thought to be the result of atrophy of the inner
retina (secondary to capillary closures) (Fig. 172.6). These
depressed areas produce a concavity that appears as a dark
retinal area with a bright central reflex.

FIGURE 172.3. (a) Several cotton wool spots


in a patient with sickle-cell disease. (b) Macular
infarction in a patient with sickle-cell
thalassemia.
(b) From Asdourian GK, Goldberg MF, Rabb MF:
Macular infarction in sickle cell B+ thalassemia. Retina
1982; 2:155.

a b
2172
Sickle-Cell Retinopathy

NONPROLIFERATIVE RETINAL CHANGES


Changes in the retina referred to as nonproliferative or back-
ground sickle retinopathy include venous tortuosity; salmon-
patch hemorrhage, schisis cavity, and iridescent spots; the black
sunburst; and other nonproliferative sickle changes.

VENOUS TORTUOSITY
Although venous tortuosity (Fig. 172.7) was one of the first
ophthalmoscopic signs of sickling to be described, it is neither
pathognomonic nor of any diagnostic value, since it occurs in a
great number of other ocular diseases. It has been reported in up
to 47% of patients with hemoglobin SS disease and 32% of
patients with hemoglobin SC disease.18 The tortuosity may
be due to arteriovenous shunting that occurs in the retinal
periphery of these patients.

CHAPTER 172
SALMON-PATCH HEMORRHAGE, SCHISIS
CAVITY, AND IRIDESCENT SPOTS
Salmon-patch hemorrhages are preretinal or superficial intra-
retinal hemorrhages that occur mainly in the mid-peripheral
retina, adjacent to a retinal arteriole.47 Because of their
peripheral location, they do not produce any visual symptoms.
FIGURE 172.4. The left macula of a patient with sickle-cell These hemorrhages are thought to occur after sudden arteriolar
hemoglobin C (SC) disease with an enlarged FAZ.
occlusions, with subsequent ‘blowout’ of the vessel wall,
presumably from ischemic necrosis.29 These hemorrhages are
round or oval and are initially bright red (Fig. 172.8). Over
several days, however, they acquire an orange-red coloration and
Even when the innermost arcades of the FAZ are occluded, have been referred to as salmon patches or salmon hemorrhages
patients are usually asymptomatic and the visual acuity, color (Fig. 172.9). Histologic studies have shown these hemorrhages
vision, and central visual fields may be normal. However, with to be limited by the internal limiting membrane, although some
more sophisticated clinical tests and static perimetry, absolute may dissect internally into the vitreous cavity or into the retina
and relative scotomas can be detected that correspond to these and the subretinal space (Fig. 172.10).48 With the resorption of
areas of vascular abnormalities. Furthermore, these macular the hemorrhages, the retina may resume a normal appearance
vascular changes may be the cause of unexplained visual loss or at the site of the hemorrhage, may be marked by a subtle retinal
amblyopia in young patients with sickle-cell disease. dimple (usually highlighted by the light reflection from the
internal limiting membrane), or more frequently, may show a
CHOROIDAL VASCULAR OCCLUSIONS schisis cavity with multiple glistening yellow spots (Fig. 172.11).
The schisis cavity represents the space created by the resorption
Choroidal vascular occlusions are reported to occur in patients of the intraretinal portion of the hemorrhage, whereas the
with sickle hemoglobinopathies, but their occurrence is rare and glistening refractive bodies in the schisis cavity, referred to as
clinically difficult to document. Condon and colleagues described iridescent spots, represent macrophages that are filled with iron
three patients with unusual chorioretinal degeneration and and blood breakdown products (Fig. 172.12).
suggested that this may be the sequelae of posterior ciliary
artery occlusions.12 Dizon and associates reported on the
clinical findings of a patient with hemoglobin SS disease that THE BLACK SUNBURST
suggested a posterior ciliary artery occlusion.9 They also reported Round or ovoid black chorioretinal scars, ranging in size from
the histopathology of three eyes in patients with hemoglobin SS 0.5 to 2 disk diameters and characteristically located in the equa-
and hemoglobin SC disease that may have sustained small torial fundus, are called black sunbursts (Fig. 172.13).2,18,21,49
posterior ciliary artery occlusions. Clinically, acute posterior They usually have stellate or spiculate borders caused by peri-
ciliary vessel occlusions appear as white, circumscribed vascular accumulation of pigment. They are sometimes associ-
triangular patches at the level of the retinal pigment epithelium ated with iridescent spots. Because of their peripheral location,
(RPE). Eventually, these areas heal with a mottled appearance of these lesions do not produce any visual symptoms.
the RPE. Acute choroidal infarctions have been well docu- On fluorescein angiography, these lesions are hypo- and
mented after feeder vessel laser photocoagulation of peripheral hyperfluorescent, denoting changes in the RPE (Fig. 172.14).
retinal neovascularization.45 Histologically, these scars represent focal areas of RPE hyper-
Peripheral spontaneous chorioretinal neovascularization has trophy, hyperplasia, and migration (Fig. 172.15).48 Occasionally,
also been reported. Liang and Jampol reported a patient with there is a distinct perivascular localization of the pigment in the
hemoglobin SS anemia who developed peripheral chorioretinal sensory retina.
neovascularization in the center of a black sunburst that was
thought to be the result of massive mid-peripheral retinal
hemorrhage.46 Choroidal neovascularization has also been OTHER NONPROLIFERATIVE SICKLE CHANGES
implicated in the formation of angioid streaks and black Mottled geographic flat, dark brown areas have been reported in
sunbursts,10 although the evidence to support the relationship the posterior pole and the fundus periphery (Fig. 172.16).21,50
is not very strong. These lesions are not associated with any hemorrhagic changes 2173
RETINA AND VITREOUS

a b
SECTION 10

c d

FIGURE 172.5. (a) Fluorescein angiography of the macular area shows an occluded vessel. (b) Fluorescein angiography shows a PAZ, as well
as microaneurysmal formation of the perifoveal capillaries. (c) Fluorescein angiography shows an enlarged FAZ. (d) Fluorescein angiography of
the macular area shows an enlarged FAZ, microaneurysmal formation, and hairpin loops.
(a–c) From Asdourian GK, Nagpal KC, Busse B, et al: Macular and perimacular vascular remodeling in sickling hemoglobinopathies. Br J Ophthalmol 1976; 60:431.)

or vascular occlusions, are transient, and disappear over a was reported to be 2.6% in SS patients,21 14% in S-b-thal
period of time. Angioid streaks have also been reported to occur patients,19 and 32.8% in SC patients.20 The development of PSR
in 22% of adult patients with hemoglobin SS disease and less is also age dependent, with the highest risk period of 20–34
frequently in patients with hemoglobin SC disease.51–53 Their years in SC disease and 40–50 years in SS disease.22 PSR has
presence is age dependent, occurring in less than 2% of hemo- also been reported in patients with other hemoglobinopathies
globin SS patients less than 40 years of age.22 They are rarely such as hemoglobin AS24 and AC.23
associated with loss of central vision secondary to choroidal The development of PSR follows a relatively orderly
neovascularization. Epiretinal membranes54 and macular holes55 sequential course. On the basis of longitudinal clinical studies
have also been reported. and with serial fluorescein angiographies, the development
of PSR has been classified by Goldberg14 into five stages
PROLIFERATIVE RETINAL CHANGES (Fig. 172.17):
1. Peripheral arteriolar occlusions
Proliferative sickle retinopathy (PSR) is the most severe ocular 2. Peripheral arteriolar–venular anastomoses
complication of sickle-cell disease, since it can lead to severe 3. Neovascular proliferation
visual disability secondary to vitreous hemorrhage or retinal 4. Vitreous hemorrhage
detachment. Although neovascularization of the disk and the 5. Retinal detachment
temporal macula have been reported,26,56 PSR is over-
whelmingly a peripheral retinal disease. PSR occurs more
frequently in the temporal retina and tends to be more rapidly STAGE I: PERIPHERAL ARTERIOLAR
progressive in children and adolescents than adults. It is more OCCLUSIONS
prevalent in patients with hemoglobin SC and hemoglobin Stage I is the earliest abnormality that can be visualized in the
S-thal disease than in those with hemoglobin SS disease. In a fundus periphery and can be seen even in children with no other
2174 study of selected patients from Jamaica, the incidence of PSR evidence of PSR. These occlusions are arteriolar rather than
Sickle-Cell Retinopathy

CHAPTER 172
FIGURE 172.6. Color photograph of the right macula shows the FIGURE 172.8. Acute preretinal hemorrhage. The hemorrhage is
retinal depression sign. bright red. Anterior to the hemorrhage, a black sunburst lesion is seen.

FIGURE 172.7. The left fundus of patient with sickle-cell (SS) disease FIGURE 172.9. Same lesion as in Figure 172.8, 4 weeks later. The
shows vascular tortuosity. hemorrhage is pink (salmon patch) with a surrounding schisis cavity.

STAGE II: PERIPHERAL ARTERIOLAR-


venular and result in the failure of the dependent capillary bed VENULAR ANASTOMOSES
to fill with resultant anteriorly located avascular zones. The With the arteriolar occlusions, a vascular remodeling process
occluded arterioles can be seen as dark red lines but eventually ensues, with some vessels remaining occluded and others
turn into white ‘silver-wire’ vessels (Fig. 172.18). Fluorescein demonstrating partial or complete reopening. As the blood is
angiography clearly delineates the occluded vessels and the diverted from the occluded arterioles to the nearest venules,
surrounding avascular and abnormal capillary bed (Fig. 172.19). arteriolar–venular anastomoses develop. Anterior to these 2175
RETINA AND VITREOUS

growth factors such as vascular endothelial growth factor and


basic fibroblast growth factor.57,58 These neovascular fronds
initially are small, lie flat on the surface of the retina, may be
mistaken for microaneurysms or telangiectasia, and are difficult
to detect ophthalmoscopically (Fig. 172.21). With time, these
neovascular tufts grow, acquire a characteristic fan-shaped
appearance resembling the marine invertebrate Gorgonia
flabellum, and are known as sea fan neovascularization
(Fig. 172.22).18
Small sea fans usually have a single feeding arteriole and a
draining venule. As they grow in size and number they acquire
additional feeding and draining vessels and may show pro-
gressive circumferential growth leading to neovascularization of
the entire retinal periphery. Fluorescein angiography shows
leakage of dye from these tufts (Fig. 172.23).
With time, these neovascular tufts may become fibrotic and
can be pulled into the vitreous cavity, appearing as elevated
neovascular tissue.
SECTION 10

STAGE IV: VITREOUS HEMORRHAGE


FIGURE 172.10. Histopathologic appearance of an acute superficial
retinal hemorrhage (salmon patch). Peripheral neovascular tufts can bleed spontaneously or second-
From Romayananda N, Goldberg MF, Green RW: Histopathology of sickle cell ary to a variety of conditions such as trauma. Spontaneous
retinopathy. Trans Am Acad Ophthalmol Otolaryngol 1973; 77:OP652-OP676. bleeding occurs secondary to contraction of the vitreous adja-
cent to the neovascular tufts or due to traction of vitreous bands
and membranes resulting from previous vitreous hemorrhages.
With PSR, the risk factors for vitreous bleeding are hemoglobin
SC disease, more than 60° of perfused sea fans, and the
presence of old blood in the vitreous.59

STAGE V: RETINAL DETACHMENT


Vitreous traction and fibrous membranes can also produce
traction on the sea fan and adjacent retina, resulting in traction
retinal detachment or traction retinoschisis.60 Localized
traction detachments may remain stationary or may progress
posteriorly. Full-thickness retinal breaks can occur due to
traction and lead to total rhegmatogenous retinal detachment.
Exudative retinal detachments are rare and may resolve after
photocoagulation of the neovascularization.61
PSR may progress rapidly in some patients, however in a
majority of patients PSR appears to progress slowly and severe
visual dysfunction is relatively uncommon. This may be due to
the tendency of the neovascular tufts to involute or undergo
autoinfarction62 (reported to occur in 20–60% of eyes with PSR)
(Fig. 172.24). Involution may occur because of strangulation of
the neovascular tufts by fibroglial tissue, whereas autoinfarction
is thought to be secondary to a spontaneous occlusion of the
arteriolar feeding vessel.
Because PSR is progressive and is the major cause of visual
morbidity in these patients (12% of eyes showing evidence of
FIGURE 172.11. Same lesion as in Figure 172.8, 6 weeks later. A visual disability), especially in young persons, obliteration of the
schisis cavity is seen with multiple iridescent spots.
neovascular tissue should be accomplished before a major
vitreous hemorrhage has occurred.63

arteriolar–venular changes, the retina remains devoid of any TREATMENT OF PSR


perfusion. On fluorescein angiography, there is no evidence of
any leakage of dye from these anastomoses, since they do not The aim of therapy is to eradicate new vessels, thus eliminating
represent true neovascularization (Fig. 172.20). the complications of vitreous hemorrhage and retinal detach-
ment.64 The mainstay of treatment is argon laser photocoag-
ulation64–68 although cryotherapy may be useful when vitreous
STAGE III: NEOVASCULAR PROLIFERATION hemorrhage prevents visualization of the retina. Fluorescein
At the interface of vascular and avascular retina, new blood angiography may be useful to localize areas of peripheral leakage
vessels arise from the arteriolar–venular anastomoses and grow prior to treatment.
peripherally into the preequatorial ischemic retina. The growth The neovascular tufts can be directly treated with photo-
of these new blood vessels is thought to be in response to coagulation if the neovascularization is flat on the retina.
2176
Sickle-Cell Retinopathy

CHAPTER 172
FIGURE 172.12. Histopathologic appearance of a schisis cavity containing proteinaceous material and hemosiderin-laden macrophages (clinical
iridescent spots).
From Romayananda N, Goldberg MF, Green RW: Histopathology of sickle cell retinopathy. Trans Am Acad Ophthalmol Otolaryngol 1973; 77:655.

FIGURE 172.14. Fluorescein angiogram of a black sunburst shows


areas of focal hyperfluorescence and hypofluorescence.
From Asdourian G, Nagpal KC, Goldbaum M, et al: Evolution of the retinal black
FIGURE 172.13. The black sunburst sign. sunburst in sickling hemoglobinopathies. Br J Ophthalmol 1975; 59:715.
From Goldberg MF: Retinal vaso-occlusion in sickling hemoglobinopathies. Birth
Defects 1976; 12:475.

Another technique favored in the past that has been shown to a 360° peripheral circumferential manner (Fig. 172.28).66,67,71
reduce vitreous hemorrhage and visual loss is feeder vessel Both of these techniques show evidence of neovascularization
treatment, whereby the feeder arterioles and subsequently the regression with minimal complications.
draining venules of the neovascularization are directly photo- Patients with nonclearing vitreous hemorrhages, tractional or
coagulated until the blood flow is obliterated (Fig. 172.25).65 rhegmatogenous retinal detachments, or epiretinal membranes
Feeder vessel treatment is now rarely performed due to com- can be treated with vitrectomy or scleral buckling.72,73 However,
plications such as choroidal hemorrhages, choroidal ischemia, these surgical procedures can be associated with both ocular and
and choroidal neovascularization that are associated with the systemic complications. Ocular complications include, anterior
intense burns needed to obliterate the vessels (Fig. 172.26).45,69,70 segment ischemia (particularly from encircling scleral buckles)74
Most physicians currently favor scatter photocoagulation and intraoperative hemorrhages with secondary glaucoma.
applied in either a localized area68 – confined to the area These complications have stimulated the preoperative use of
anterior to perfused neovascular fronds (Fig. 172.27) – or in prophylactic exchange transfusions or erythrophoresis.75 How-
2177
SECTION 10 RETINA AND VITREOUS

FIGURE 172.16. Fundus photograph shows a mottled brown area.

ever, the risks involved with these transfusion procedures –


acquired infections (human immunodeficiency virus, non-A,
non-B hepatitis) – have stimulated a reevaluation of this rou-
tine. At present, improved vitreoretinal techniques and the use
of pre- and postoperative oxygen and intraoperative measures
(avoiding excessive manipulation of extraocular muscles,
avoiding encircling scleral buckles, limiting the use of intra-
ocular gases) to reduce complications have almost eliminated
the need for preoperative exchange transfusions. Systemic
complications include thromboembolic complications such as
pulmonary or cerebral embolism or thrombosis. Patients who
need vitreoretinal surgery should be evaluated by experienced
medical personnel to decrease the systemic complications of
surgery.

FIGURE 172.15. Histopathologic appearance of a black sunburst


shows marked proliferation of the RPE.
From Romayananda N, Goldberg MF, Green RW: Histopathology of sickle cell
retinopathy. Trans Am Acad Ophthalmol Otolaryngol 1973; 77:657.

2178
Sickle-Cell Retinopathy

CHAPTER 172
FIGURE 172.18. Stage I PSR. Peripheral arteriolar occlusions are
seen as ‘silver-wire’ vessels.

FIGURE 172.17. Classification of PSR.


From Goldberg MF: Classification and pathogenesis of proliferative sickle
retinopathy. Am J Ophthalmol 1971; 71:654. Copyright from Elsevier Science.

FIGURE 172.19. Stage I PSR. Fluorescein angiography shows the


occluded peripheral vessels with the adjacent avascular retina.

2179
SECTION 10 RETINA AND VITREOUS

FIGURE 172.21. Early retinal neovascularization, which may be


FIGURE 172.20. Stage II PSR. Fluorescein angiography shows the
mistaken for microaneurysms or telangiectasia.
arteriolar–venular anastomoses with the adjacent avascular retina.

FIGURE 172.22. (a) Fluorescein angiography


of characteristic sea fan neovascularization.
(b) The sea fan neovascularization shows
evidence of leakage of dye. Inferior to the
neovascularization, the arteriolar–venular
anastomosis is seen with early
neovascularization.

a b

FIGURE 172.23. (a) Sea fan neovascularization


with a single feeder vessel and two draining
venules. (b) Sea fan neovascularization with
multiple feeder arterioles and draining venules.

a b

2180
Sickle-Cell Retinopathy

FIGURE 172.24. (a and b) Autoinfarcted sea


fan neovascularization. (c) A perfused, leaking
sea fan neovascularization. (d) Same area
several days later shows lack of perfusion.
(c and d) From Nagpal KC, Patrianakos D, Asdourian
GK, et al: Spontaneous regression (autoinfarction) of
PSR. Am J Ophthalmol 1975; 80:886. Copyright from
Elsevier Science.

a b

CHAPTER 172
c d

FIGURE 172.25. Feeder vessel photocoagulation. Both feeder


arterioles and draining venules have been photocoagulated. The
neovascular tuft is not treated.

2181
RETINA AND VITREOUS

FIGURE 172.26. (a) Feeder vessel


photocoagulation. A localized hemorrhage is
noted adjacent to the sea fan
neovascularization. (b) Anteriorly located
triangular gray areas represent areas of
choroidal ischemia. (c) Heavy treatment has
resulted in breaks in Bruch’s membrane (dark
areas in the center of photocoagulation spots).
(d) Same patient as in (c). Choroidal
neovascularization has occurred at the site of
the break in Bruch’s membrane. (e–h) Same
patient as in (c). Rapid-sequence fluorescein
angiography shows choroidal
neovascularization occurring after heavy
photocoagulation, which resulted in breaks in
Bruch’s membrane. Center arrows show filling
a b of main choroidal vascular trunk before retinal
arterial filling (open arrows). Lower arrows show
another but smaller neovascular tuft.
(e–h) From Galinos SO, Asdourian GK, Woolf MB,
SECTION 10

et al: Choroido-vitreal neovascularization after argon


laser photocoagulation neovascularization. Arch
Ophthalmol 1975; 93:524. Copyright 1975, American
Medical Association.

c d

e f

g h

2182
Sickle-Cell Retinopathy

FIGURE 172.27. (a) Fluorescein angiogram


demonstrates a leaking sea fan
neovascularization. (b) Complete regression of
the lesion occurred after scatter
photocoagulation.
(a and b) Reprinted from Rednam KR, Jampol LM,
Goldberg MF: Scatter retinal photocoagulation for
proliferative sickle cell retinopathy. Am J Ophthalmol
1982; 93:594. Copyright from Elsevier Science.

a b

CHAPTER 172
FIGURE 172.28. Fundus drawing illustrates the technique of
peripheral circumferential retinal scatter photocoagulation.
From Cruess AF, Stephens RF, Magargal LE, et al: Peripheral circumferential
retinal scatter photocoagulation for treatment of proliferative sickle retinopathy.
Ophthalmology 1983; 90:273.

2183
RETINA AND VITREOUS

REFERENCES
1. Beutler E: Disorders of hemoglobin 21. Condon PI, Serjeant GR: Ocular findings in anesthesia. Am J Ophthalmol 1982;
structure: sickle cell anemia and related homozygous sickle cell anemia in Jamaica. 93:573–577.
abnormalities. In: Lichtman MA, Beutler E, Am J Ophthalmol 1972; 73:533–543. 39. Sorr EM, Goldberg RE: Traumatic central
Kipps TJ, et al, eds. Williams Hematology. 22. Condon PI, Serjeant GR: Ocular findings of retinal artery occlusion with sickle cell trait.
7th edn. New York: McGraw-Hill; elderly cases of homozygous sickle-cell Am J Ophthalmol 1975; 80:648–652.
2006:668–700. disease in Jamaica. Br J Ophthalmol 1976; 40. Sanders RJ, Brown GC, Rosenstein RB,
2. Goldberg MF: Retinal vaso-occlusion in 60:361–364. Magargal L: Foveal avascular zone
sickling hemoglobinopathies. Birth Defects 23. Moschandreou M, Galinos S, Valenzuela R, diameter and sickle cell disease. Arch
Orig Artic Ser 1976; 12:475–515. et al: Retinopathy in hemoglobin C trait Ophthalmol 1991; 109:812–815.
3. Welch RB: Fluorescein angiography in (AC hemoglobinopathy). Am J Ophthalmol 41. Asdourian GK, Nagpal KC, Busse B, et al:
sickle-cell retinopathy and von 1974; 77:465–471. Macular and perimacular vascular
Hippel–Lindau disease. Int Ophthalmol Clin 24. Nagpal KC, Asdourian GK, Patrianakos D, remodelling sickling haemoglobinopathies.
1977; 17:137–154. et al: Proliferative retinopathy in sickle cell Br J Ophthalmol 1976; 60:431–453.
4. Goodman G, Von Sallmann L, Holland MG: trait. Report of seven cases. Arch Intern 42. Lee CM, Charles HC, Smith RT, et al:
Ocular manifestations of sickle-cell Med 1977; 137:325–328. Quantification of macular ischaemia in
disease. AMA Arch Ophthalmol 1957; 25. Kimmel AS, Magargal LE, Tasman WS: sickle cell retinopathy. Br J Ophthalmol
58:655–682. Proliferative sickle retinopathy and 1987; 71:540–545.
5. Paton D: The conjunctival sign of sickle-cell neovascularization of the disc: regression 43. Stevens TS, Busse B, Lee CB, et al:
SECTION 10

disease. Arch Ophthalmol 1961; 66:90–94. following treatment with peripheral retinal Sickling hemoglobinopathies; macular and
6. Paton D: The conjunctival sign ox scatter laser photocoagulation. Ophthalmic perimacular vascular abnormalities. Arch
sickle-cell disease. Further observations. Surg 1986; 17:20–22. Ophthalmol 1974; 92:455–463.
Arch Ophthalmol 1962; 68:627–632. 26. Ober RR, Michels RG: Optic disk 44. Goldbaum MH: Retinal depression sign
7. Galinos S, Rabb MF, Goldberg MF, Frenkel M: neovascularization in hemoglobin SC indicating a small retinal infarct. Am J
Hemoglobin SC disease and iris atrophy. disease. Am J Ophthalmol 1978; Ophthalmol 1978; 86:45–55.
Am J Ophthalmol 1973; 75:421–425. 85:711–714. 45. Goldbaum MH, Galinos SO, Apple D, et al:
8. Goldberg MF, Tso MO: Rubeosis iridis and 27. Asdourian GK, Goldberg MF, Rabb MF: Acute choroidal ischemia as a complication
glaucoma associated with sickle cell Macular infarction in sickle cell B+ of photocoagulation. Arch Ophthalmol
retinopathy: a light and electron thalassemia. Retina 1982; 2:155–158. 1976; 94:1025–1035.
microscopic study. Ophthalmology 1978; 28. Merritt JC, Risco JM, Pantell JP: Bilateral 46. Liang JC, Jampol LM: Spontaneous
85:1028–1041. macular infaction in SS disease. J Pediatr peripheral chorioretinal neovascularisation
9. Dizon RV, Jampol LM, Goldberg MF, Juarez Ophthalmol Strabismus 1982; 19:275–278. in association with sickle cell anaemia. Br J
C: Choroidal occlusive disease in sickle cell 29. Jampol LM, Condon P, Dizon-Moore R, et al: Ophthalmol 1983; 67:107–110.
hemoglobinopathies. Surv Ophthalmol Salmon-patch hemorrhages after central 47. Gagliano DA, Goldberg MF: The evolution
1979; 23:297–306. retinal artery occlusion in sickle cell disease. of salmon-patch hemorrhages in sickle cell
10. McLeod DS, Goldberg MF, Lutty GA: Arch Ophthalmol 1981; 99:237–240. retinopathy. Arch Ophthalmol 1989;
Dual-perspective analysis of vascular 30. Fine LC, Petrovic V, Irvine AR, Bhisitkul RB: 107:1814–1815.
formations in sickle cell retinopathy. Arch Spontaneous central retinal artery 48. Romayanada N, Goldberg MF, Green WR:
Ophthalmol 1993; 111:1234–1245. occlusion in hemoglobin sickle cell disease. Histopathology of sickle cell retinopathy.
11. Stein MR, Gay AJ: Acute chorioretinal Am J Ophthalmol 2000; 129:680–681. Trans Am Acad Ophthalmol Otolaryngol
infarction in sickle cell trait. Report of 31. Al-Abdulla NA, Haddock TA, Kerrison JB, 1973; 77:OP642–OP676.
a case. Arch Ophthalmol 1970; Goldberg MF: Sickle cell disease 49. Asdourian G, Nagpal KC, Goldbaum M,
84:485–490. presenting with extensive peri-macular et al: Evolution of the retinal black sunburst
12. Condon PI, Serjeant GR, Ikeda H: Unusual arteriolar occlusions in a nine-year-old boy. in sickling haemoglobinopathies. Br J
chorioretinal degeneration in sickle cell Am J Ophthalmol 2001; 131:275–276. Ophthalmol 1975; 59:710–716.
disease. Possible sequelae of posterior 32. Conrad WC, Penner R: Sickle-cell trait and 50. Nagpal KC, Goldberg MF, Asdourian G,
ciliary vessel occlusion. Br J Ophthalmol central retinal-artery occlusion. Am J et al: Dark-without-pressure fundus lesions.
1973; 57:81–88. Ophthalmol 1967; 63:465–468. Br J Ophthalmol 1975; 59:476–479.
13. Goldbaum MH, Jampol LM, Goldberg MF: 33. Kabakow B, Van Weimokly SS, Lyons HA: 51. Nagpal KC, Asdourian G, Goldbaum M, et al:
The disc sign in sickling hemoglobinopathies. Bilateral central retinal artery occlusion; Angioid streaks and sickle
Arch Ophthalmol 1978; 96:1597–1600. occurrence in a patient with cortisone- haemoglobinopathies. Br J Ophthalmol
14. Goldberg MF: Classification and treated systemic lupus erythematosus, 1976; 60:31–34.
pathogenesis of proliferative sickle sickle cell trait, and active pulmonary 52. Hamilton AM, Pope FM, Condon PI, et al:
retinopathy. Am J Ophthalmol 1971; tuberculosis. AMA Arch Ophthalmol 1955; Angioid streaks in Jamaican patients with
71:649–665. 54:670–676. homozygous sickle cell disease. Br J
15. Goldberg MF: Sickle cell retinopathy. 34. Acacio I, Goldberg MF: Peripapillary and Ophthalmol 1981; 65:341–347.
In: Tasman W, Jaeger EA, eds. Duane’s macular vessel occlusions in sickle cell 53. Paton D: Angiod streaks and sickle cell
clinical ophthalmology. Philadelphia: anemia. Am J Ophthalmol 1973; anemia: a report of two cases. Arch
Lippincott Willaims & Wilkins; 2004:1–45. 75:861–866. Ophthalmol 1959; 62:852–858.
16. Goldberg MF: Natural history of untreated 35. Khwarg SG, Feldman S, Ligh J, 54. Carney MD, Jampol LM: Epiretinal
proliferative sickle retinopathy. Arch Straatsma BR: Exchange transfusion in membranes in sickle cell retinopathy.
Ophthalmol 1971; 85:428–437. sickling maculopathy. Retina 1985; Arch Ophthalmol 1987; 105:214–217.
17. Penman AD, Talbot JF, Chuang EL, et al: 5:227–229. 55. Raichand M, Dizon RV, Nagpal KC, et al:
New classification of peripheral retinal 36. Weissman H, Nadel AJ, Dunn M: Macular holes associated with proliferative
vascular changes in sickle cell disease. Simultaneous bilateral retinal arterial sickle cell retinopathy. Arch Ophthalmol
Br J Ophthalmol 1994; 78:681–689. occlusions treated by exchange 1978; 96:1592–1596.
18. Welch RB, Goldberg MF: Sickle-cell transfusions. Arch Ophthalmol 1979; 56. Frank RN, Cronin MA: Posterior pole
hemoglobin and its relation to fundus 97:2151–2153. neovascularization in a patient with
abnormality. Arch Ophthalmol 1966; 37. Roth SE, Magargal LE, Kimmel AS, et al: hemoglobin SC disease. Am J Ophthalmol
75:353–362. Central retinal-artery occlusion in 1979; 88:680–682.
19. Condon PI, Serjeant GR: Ocular findings in proliferative sickle-cell retinopathy after 57. Cao J, Mathews MK, McLeod DS, et al:
sickle cell thalassemia in Jamaica. Am J retrobulbar injection. Ann Ophthalmol 1988; Angiogenic factors in human proliferative
Ophthalmol 1972; 74:1105–1109. 20:221–224. sickle cell retinopathy. Br J Ophthalmol
20. Condon PI, Serjeant GR: Ocular findings in 38. Klein ML, Jampol LM, Condon PI, et al: 1999; 83:838–846.
hemoglobin SC disease in Jamaica. Am J Central retinal artery occlusion without 58. Kim SY, Mocanu C, McLeod DS, et al:
Ophthalmol 1972; 74:921–931. retrobulbar hemorrhage after retrobulbar Expression of pigment epithelium-derived
2184
Sickle-Cell Retinopathy

factor (PEDF) and vascular endothelial 64. Goldberg MF: Treatment of proliferative 70. Galinos SO, Asdourian GK, Woolf MB, et al:
growth factor (VEGF) in sickle cell retina sickle retinopathy. Trans Am Acad Choroido-vitreal neovascularization after
and choroid. Exp Eye Res 2003; Ophthalmol Otolaryngol 1971; 75:532–556. argon laser photocoagulation. Arch
77:433–445. 65. Condon P, Jampol LM, Farber MD, et al: Ophthalmol 1975; 93:524–530.
59. Jacobson MS, Gagliano DA, Cohen SB, A randomized clinical trial of feeder vessel 71. Cruess AF, Stephens RF, Magargal LE,
et al: A randomized clinical trial of feeder photocoagulation of proliferative sickle cell Brown GC: Peripheral circumferential retinal
vessel photocoagulation of sickle cell retinopathy. II. Update and analysis of risk scatter photocoagulation for treatment of
retinopathy. A long-term follow-up. factors. Ophthalmology 1984; 91:1496–1498. proliferative sickle retinopathy.
Ophthalmology 1991; 98:581–585. 66. Farber MD, Jampol LM, Fox P, et al: Ophthalmology 1983; 90:272–278.
60. Goldberg MF: Retinal detachment A randomized clinical trial of scatter 72. Jampol LM, Green JL Jr, Goldberg MF,
associated with proliferative retinopathies photocoagulation of proliferative sickle cell Peyman GA: An update on vitrectomy
(sickle cell disease, retrolental fibroplasia retinopathy. Arch Ophthalmol 1991; surgery and retinal detachment repair in
and diabetes mellitus). Isr J Med Sci 1972; 109:363–367. sickle cell disease. Arch Ophthalmol 1982;
8:1447–1457. 67. Jampol LM, Farber M, Rabb MF, 100:591–593.
61. Durant WJ, Jampol LM, Daily M: Exudative Serjeant G: An update on techniques of 73. Morgan CM, D’Amico DJ: Vitrectomy
retinal detachment in hemoglobin SC photocoagulation treatment of surgery in proliferative sickle retinopathy.
disease. Retina 1982; 2:152–154. proliferative sickle cell retinopathy. Eye Am J Ophthalmol 1987; 104:133–138.
62. Nagpal KC, Patrianakos D, Asdourian GK, 1991; 5(Pt 2):260–263. 74. Ryan SJ, Goldberg MF: Anterior segment
et al: Spontaneous regression 68. Rednam KR, Jampol LM, Goldberg MF: ischemia following scleral buckling in sickle
(autoinfarction) of proliferative sickle Scatter retinal photocoagulation for cell hemoglobinopathy. Am J Ophthalmol

CHAPTER 172
retinopathy. Am J Ophthalmol 1975; proliferative sickle cell retinopathy. Am J 1971; 72:35–50.
80:885–892. Ophthalmol 1982; 93:594–599. 75. Koshy M, Weiner SJ, Miller ST, et al:
63. Stephens RF: Proliferative sickle cell 69. Condon PI, Jampol LM, Ford SM, Serjeant Surgery and anesthesia in sickle cell
retinopathy: the disease and a review of its GR: Choroidal neovascularisation induced disease. Cooperative study of sickle cell
management. Ophthalmic Surg 1987; by photocoagulation in sickle cell disease. diseases. Blood 1995; 86:3676–3684.
18:222–231. Br J Ophthalmol 1981; 65:192–197.

2185
CHAPTER

173 Traumatic Retinopathy


Melvin D. Rabena, Dante J. Pieramici, and Mark W. Balles

The retina, marvelous in its design and function, is a fragile Key Features: Contusion (Closed Globe Injuries)
tissue, easily injured and possessing only modest capabilities • When a blunt force strikes the eye the globe undergoes
for repair. The mechanisms of traumatic injury of the retina are mechanical changes in its anteroposterior and equatorial
numerous and can be categorized into those associated with dimension
blunt force (contusion/rupture), those resulting from sharp or • Forces generated at attachment sites cause retinal dialysis,
lacerating forces, those resulting from acceleration/deceleration tears, and detachments
forces, and other more complex mechanisms. The most com-
mon injuries of the retina involve contusion related trauma
(closed globe) and to understand the diffuse nature of these
injuries requires an appreciation of the mechanical changes the CHOROIDAL RUPTURE
globe undergoes following blunt impact (Fig. 173.1). A choroidal rupture is a tear in the choroid, Bruch’s membrane,
Our ability to manage severe ocular injuries has certainly and retinal pigment epithelium (RPE)1 and was first described
improved in the past 50 years; however, involvement of the by von Graefe in 1854.2 Choroidal ruptures may occur
retina continues to be an ominous sign, portending a poor anteriorly, where they are usually parallel to the ora serrata, or
visual prognosis. posteriorly, where they are usually crescentic and oriented
around the optic nerve (Fig. 173.2). Anterior choroidal ruptures
CONTUSION (BLUNT, CLOSED GLOBE) are thought to occur directly at the site of impact, while
posterior pole choroidal ruptures result indirectly from the
When a blunt force strikes the eye, the eyeball is shortened in movements of the eyewall.
the anteroposterior dimension and elongated equatorially. As Identification of a choroidal rupture acutely may be difficult
the lens–iris diaphragm and the anterior vitreous base are because of co-existing anterior segment or vitreous opacities
anchored to the eyewall, portions of these structures are forced (i.e., hyphema). Even in cases of clear ocular media, posterior
posteriorly, while their sites of attachment to the eyewall move segment hemorrhage, which may be subretinal, retinal, or
equatorially. This results in additional forces generated at the preretinal, is often present obscuring the rupture (Fig. 173.3). In
attachment sites leading to characteristic contusion injuries such cases, one or more ruptures are almost always present,
such as a retinal dialysis. More complex less well understood only to be identified with subsequent clearing of the blood or
interactions between the movement of the eyewall and the with fluorescein angiographic testing.
vitreous gel lead to posteriorly located retinal injuries such as Most patients presenting with choroidal ruptures do not
a traumatic macular hole or choroidal rupture. regain vision to the 20/40 level.3 Visual outcome is often

FIGURE 173.1. Closed globe injury.


(a) Dimensional changes in the eye following
contusion (blunt) injury. The anteroposterior
dimension of the globe shortens (large arrow)
as it expands equatorially (smaller arrows).
(b) Forces generated at the lens–iris diaphragm
and vitreous base attachment site at the point
of maximum equatorial expansion.

a b

2187
RETINA AND VITREOUS

FIGURE 173.2. Choroidal rupture.


(a) Fundus photograph showing multiple
concentric choroidal ruptures, one through the
fovea. (b–d) Corresponding red-free fundus
photograph, late-phase fluorescein angiogrpahy
and ocular coherence tomography.
b

a
SECTION 10

c d

FIGURE 173.3. Choroidal rupture. (a) Acutely


after injury, retinal and subretinal hemorrhage
and commotio retinae obscure the peripapillary
choroidal rupture from view. (b) The same
patient, 2 months later. After clearing of the
hemorrhage, the peripapillary, concentric
choroidal rupture can be seen.

a b

difficult to predict at presentation but poor final vision is COMMOTIO RETINAE


associated with foveal location of the rupture, multiple rup- First described in 1873,6 commotio retinae (Berlin’s edema)
tures, and poor presenting visual acuity.3 Choroidal ruptures occurs after blunt injury to the eye and is characterized by
heal with time through the process of fibrous and fibrovascular decrease in central and/or peripheral vision and a transient gray-
proliferation. Choroidal neovascularization may be a late com- white patchy discoloration and opacification of the outer
plication of a choroidal rupture occurring months or years neurosensory retina (Fig. 173.4). The opacification may be
following the initial injury. The development of choroidal neo- accompanied by retinal, subretinal, or preretinal hemorrhage
vascularization is associated with more posteriorly located and choroidal rupture.7 Central and peripheral vision tends to
choroidal ruptures and older aged patients. Laser photo- improve as the retinal whitening resolves; however, in other
coagulation,4 photodynamic therapy,5 or anti-VEGF therapy of cases, central vision loss may be permanent, associated with
choroidal rupture-associated choroidal neovascularization RPE changes and pigment migration.8 The peripheral pigmentary
can be beneficial in minimizing visual loss. changes when present, can mimic retinitis pigmentosa.9,10
Fluorescein angiography typically shows no alteration in
retinal vascular permeability.11,12 Experimentally produced
commotio retinae in owl monkeys has shown disruption of the
Key Features: Choroidal Rupture
photoreceptor outer segments and acute damage to the
• Characterized by a tear in the choroid, Bruch’s membrane,
photoreceptor cells.13 This was followed by pigment migration
retinal pigment epithelium
to the ganglion cell layer and, in severe injuries, thinning of the
• Anterior choroidal ruptures are parallel to the ora serrata
• Posterior choroidal ruptures are crescentic oriented around the
outer plexiform and outer nuclear layers with variable loss of
optic nerve
the photoreceptor outer segments. No evidence of intracellular
• Poor final visual outcome is associated with fovea location of
or extracellular retinal edema was seen in this model, which
rupture, multiple ruptures and poor presenting visual acuity
appears clinically identical to human patients with this
• Choroidal neovascularization can be a late complication
condition.
(months or years later) of choroidal ruptures
The visual prognosis is good in extrafoveal or mild injuries
2188 and the vision tends to recover in a vast majority of cases
Traumatic Retinopathy

(Fig. 173.5). Ocular coherence tomography reveals findings


similar to nontraumatic holes, although at times, a more
ragged/irregular appearance to the edges of the hole are present.
Sometimes, traumatic macular holes may close spontaneously,
particularly in younger patients, and a period of observation is
reasonable before deciding on surgery in these cases. Vitrectomy
with or without peeling of the internal limiting membrane
(ILM) can result in hole closure in over 90% of the cases with
visual improvement of two or more lines in more than three out
of four patients and more than half of the patients achieving
20/50.17 Eyes with associated macular RPE atrophy or choroidal
injury have a poorer visual prognosis and may have only limited
improvement after surgery.18,19 Progression to retinal
detachment rarely occurs as macular holes are usually stable
lesions.20

Key Features: Traumatic Macular Hole

CHAPTER 173
• Accounts for less than 10% of macular hole cases
• Traumatic macular holes typically ranges in size from 0.2 to 0.5
disk diameters
FIGURE 173.4. Commotio retinae in the extramacular retina. The • Vitrectomy with or without ILM peeling can result in hole
retinal whitening affects the outer retina and is seen in this patient in closure (90% of cases) and improvement in visual acuity
association with retinal hemorrhage. One month later, the retinal • Progression to retinal detachment is unlikely
whitening had resolved with some pigment migration into the • Spontaneous closure of traumatic macular holes has been
neurosensory retina.
reported, suggesting a role for initial observation

(60%).14 Permanent vision loss may occur if there is significant


damage in the fovea or there are associated retinal injuries such
CHORIORETINITIS SCLOPETARIA
as a choroidal rupture or macular hole formation. There is no Chorioretinitis sclopetaria is characterized by a rupture of the
treatment of proven benefit for this condition. retina and choroid after nonpenetrating ocular trauma in which
a high-velocity projectile usually a shotgun or BB pellet, strikes
or passes tangential to the globe.21 The first case caused by a
Key Features: Commotio Retinae shotgun, was described by Goldzeiher22 in 1901. The concus-
• Transient gray-white patchy discoloration and opacification of sive force of the injury causes full-thickness rupture of the
outer neurosensory retina retina, Bruch’s membrane, and choroid with associated
• Commotio retinae is not associated with extracellular edema hemorrhage and is followed by retraction of these tissues to
but with disruption of photoreceptor outer segments expose the bare sclera (Fig. 173.6).21 It is relatively rare,
• Visual acuity tends to recover except in cases where there is especially during peacetime, as it is most often the result of gun
significant damage to the fovea related injuries.23
Both the posterior pole and the peripheral retina may be
involved, with marked choroidal and retinal hemorrhages
TRAUMATIC MACULAR HOLE associated with tears in these layers. The sclera remains intact
Trauma is the mechanism of macular hole formation in ~10% and may be visible on ophthalmoscopy but may be obstructed
of cases.15 Macular holes may be present immediately following acutely by overlying vitreous hemorrhage and intraretinal and
the contusion injury or may take days to weeks to form. The subretinal hemorrhage. Retinal detachment occurs only rarely,
pathophysiologic mechanism(s) underlying traumatic macular and with resolution of the hemorrhage the lesion typically heals
hole formation are unclear but in most cases, traumatic holes and develops irregular border with the formation of white
probably result from similar, albeit more abrupt, tractional fibrous scars and associated pigment migration and
vitreofoveal interface changes similar to idiopathic macular proliferation.24 The visual prognosis is usually poor owing to
holes.16 Traumatic macular holes range in size from 0.2 to 0.5 the initial severity of the injury, however cases of excellent
disk diameters and may be round or oval in appearance visual return have been reported.25 Vitrectomy may be used to

a b c

FIGURE 173.5. Traumatic macular hole. (a) Color fundus photograph and (b) ocular coherence tomography revealing small full-thickness
macular hole less than 500 mm in diameter caused by contusion injury to the eye. (c) Anterior segment photograph shows zonular fiber dialysis. 2189
RETINA AND VITREOUS

FIGURE 173.6. Chorioretinitis sclopetaria.


(a) This 16-year-old boy noted decreased vision
after being shot with a BB gun from 4 ft away.
Vision was 8/200. Note the entry wound in the
left lower lid just above the inferior orbital rim.
Subconjunctival hemorrhage is also visible.
(b) Fundus photographs of the inferonasal
quadrant in this patient show the acute
appearance of choroidal and retinal rupture
with preretinal hemorrhage typical in
chorioretinitis sclopetaria. The sclera remains
intact. (c) Computed tomography scan shows
the location of the BB at the orbital apex. Note
that the globe appears intact, without the ‘flat
a b tire’ sign seen in perforating injuries of the
globe. (d) Seven months later, the patient’s
vision has improved to 20/70. The retina has
remained attached without surgical
intervention, and a white scar with pigment
proliferation and intraretinal pigment migration
SECTION 10

along its margins is present in the inferonasal


quadrant.

c d

remove nonclearing vitreous hemorrhage26 or repair retinal Avulsion of the anterior vitreous base in association with
detachment; however, surgery is not often required. detachment always indicates a history of significant ocular
trauma. Retinal detachments associated with retinal dialysis
Key Features: Chorioretinitis Sclopetaria and other traumatic retinal breaks tend to progress slowly and
• Nonpenetrating ocular trauma in which a high velocity may not present for months or years following the injury.
projectile (i.e., shotgun or BB pellet) strikes or passes Retinal tears associated with ocular trauma may be treated
tangential to the globe with cryoretinopexy or laser retinopexy. Traumatic retinal
• Concussive force causes a full-thickness rupture of the retina, detachments can be treated using scleral buckling or vitrectomy
Bruch’s membrane and choroid techniques. Proliferative vitreoretinopathy (PVR) is the most
• Lesions typically heal spontaneously and only rarely lead to common reason for surgical failure in the management of
retinal detachment trauma related retinal detachment. Cases of retinal detachment
associated with PVR generally require more advanced vitreo-
retinal surgical techniques including; vitrectomy, membrane
dissection, retinectomy, and use of long-active intraocular gases
TRAUMATIC RETINAL TEARS, DIALYSIS, AND
or silicone.
DETACHMENT
As mentioned above movement of the eyewall relative to the Key Features: Traumatic Retinal Tears, Dialysis, and
vitreous following blunt trauma creates tractional forces at Detachment
the vitreous base and pars plana. Blunt trauma to the eye can • Retinal dialysis is characterized by a break or separation at the
cause retinal tears, dialysis, and retinal detachment. Traumatic anterior edge of the ora serrata and are most common in the
retinal detachments account for ~15% of all detachments inferotemporal quadrant
and tend to occur in a much younger patient population than • Retinal detachments following trauma may occur months to
do nontraumatic detachments. Traumatic detachment is years following the trauma
four times more common in males than in females, compared • PVR is the most common reason for surgical failure in the
with a similar incidence between the sexes for nontraumatic management of trauma related retinal detachment
detachments.27,28
Retinal dialysis is the most common type of retinal break
associated with traumatic retinal detachment. It occurs most ACCELERATION/DECELERATION INJURY
commonly in the inferotemporal quadrant (66% of detachments)
followed by the superonasal quadrant (14% of detachments). A
retinal dialysis is characterized by a break or separation at the
OPTIC NERVE EVULSION
anterior edge of the ora serrata and it differs from a retinal tear Evulsion of the optic nerve is the rarest form of traumatic optic
as the vitreous remains attached to the posterior edge of the neuropathy. The optic nerve may be partially or completely
dialysis.29,30 In the same series, giant retinal tears accounted for separated from the globe. In total evulsion the vitreous and
16% of detachments, retinal flap tears accounted for 11%, and retina separate from the optic disk and the lamina cribosa is
2190 tears in an area of lattice accounted for 8% of detachments.28 detached from the choroid and sclera. Partial evulsion involves
Traumatic Retinopathy

partial disruptions of laminar–scleral connections. Cases can resolve quickly and are generally gone by 1 month following
involve severe orbital trauma;31,32 however, other cases of partial birth.40 They can mimic retinal hemorrhages seen in the shaken
evulsion have been reported after seemingly minor trauma.33,34 baby syndrome and should be considered in the differential
Fundus examination shows total or partial absence of the optic diagnosis.
disk however immediately after injury the disk is often obscured
by overlying vitreous hemorrhage. Partial evulsion may mimic RETINAL TRAUMA ASSOCIATED WITH
an optic nerve pit in appearance. Serous macular detachment OPEN GLOBE INJURIES
has been reported after partial evulsion injury.33
Fluorescein angiography may demonstrate either normal, Open globe injuries (full-thickness eyewall defect present) are
partial, or absent retinal vascular filling. Anterior segment neo- classified as lacerating if they are the result of sharp force
vascularization may develop in rare cases as a result of posterior
segment ischemia. Computed tomography or magnetic reso-
nance imaging usually demonstrates an intact nerve sheath.26
Presumed mechanisms of injury include extreme rotation
and anterior globe displacement, sudden marked increase in
intraocular pressure after traumatic compression in which the
optic nerve is pneumatically disinserted and sudden elevation of
intraorbital pressure stretches optic nerve until it is evulsed

CHAPTER 173
from its scleral insertion.35

Key Features: Optic Nerve Evulsion


• In total evulsion the vitreous and retina separate from the optic
disk and the lamina cribrosa is detached from the choroid and
sclera
• Partial evulsion involves partial disruptions of laminar–scleral
connections.
• Presumed mechanisms of injury include:
• Extreme rotation and anterior globe displacement
• Sudden increase in intraocular pressure after traumatic
compression
• Sudden elevation of intraorbital pressure
FIGURE 173.7. Shaken baby syndrome. This 1-month-old child was
hospitalized in the neonatal intensive care unit with subarachnoid and
SHAKEN BABY SYNDROME subdural hemorrhages due to child abuse. The fundus photograph of
the right eye taken at that time demonstrates a large, bean-shaped
Shaken baby syndrome presents as retinal hemorrhages and macular subinternal limiting membrane hemorrhage. A glistening light
cotton-wool spots in an abused infant who may have sustained reflex is present on its surface, and white, intraretinal patches are seen
violent shaking; direct eye, head, or chest trauma; or in the macula temporal to the hemorrhage. Similar findings were
choking.6,36 Papilledema and vitreous hemorrhage may occur in present in the fellow eye.
association, along with subdural or subarachnoid hemorrhage
or cerebral contusion. Ocular injury may be present in up to
30–40% of abused children.36–38 Retinal manifestations are
most common (Fig. 173.7) and may appear similar to Terson’s
syndrome, Purtscher’s retinopathy, or central retinal vein occlu-
sion.6 Other rare retinal manifestations include; subhyaloidal
hemorrhage (hemorrhagic macular cyst) and circular perima-
cular folds. The prognosis is poor, with children suffering from
the sequelae of their intracranial injuries as well as ocular visual
loss, which may be secondary to macular scarring, vitreous
hemorrhage, retinal detachment,39 or from amblyopia. Retinal
reattachment surgery or vitrectomy for nonclearing vitreous
hemorrhage may be beneficial.26
Intraretinal, subretinal, pretinal, and vitreous hemorrhages
may occur following either vaginal or caesarean birth in over
one-third of newborns (Fig. 173.8). These hemorrhages tend to

Key Features: Shaken Baby Syndrome


• Characterized by retinal hemorrhages and cotton wool spots
• Papilledema, vitreous hemorrhage, subarachnoid hemorrhage
or cerebral contusion may also occur
• Prognosis is poor, with children suffering from intracranial
injuries and ocular visual loss
• Intraretinal, subretinal, pretinal, and vitreous hemorrhages may
occur following either vaginal or caesarean birth in over 1/3 of
newborns but these tend to resolve by the first month FIGURE 173.8. Intraretinal hemorrhages following birth. Fundus
following birth photograph of newborn with multiple intraretinal hemorrhages some
with white centers. 2191
RETINA AND VITREOUS

(i.e., nail injury) or rupture if the eyewall opens as a result of


blunt force.41
A ruptured globe occurs when a blunt object of significant
force strikes the eyewall. This causes typical deformation
changes in the eyewall and marked increase in the intraocular
pressure. When significant blunt force is applied, or in cases of
inherent or acquired eyewall weakness and lesser forces, the
eyewall will rupture or burst at its weakest points. The most
common locations are just posterior to the rectus muscle
insertions where the sclera is thinnest, at the equator, at the site
of previous surgical incisions, or at the limbus. Ruptured globes
carry a poor prognosis as the injuries typically result in diffuse
ocular trauma with posteriorly located wounds.42
Lacerating open globe trauma is sub-classified into penetrat-
ing, perforating, and intraocular foreign body (IOFB) injuries.
Penetrating wounds are the result of sharp forces that result in
full-thickness defect entrance wounds. They tend to carry a
favorable prognosis unless the wounds are large and extend
SECTION 10

posteriorly. IOFB injuries are essentially penetrating injuries in


which the lacerating object remains lodged into the eye. They
also may carry a favorable prognosis if the foreign body is small
and sharp in nature. IOFB injuries must be identified acutely as
they carry a higher risk of traumatic endophthalmitis, FIGURE 173.9. Perforating intraocular foreign body injury. This
particularly if the IOFB is not removed urgently. CT scan and/or patient suffered perforating pellet injury to the left eye. An intraocular
foreign body (pellet) lodged in the orbit resulting after passing through
B-scan testing should be performed in all cases of open globe the globe causing diffuse injury. Note the intraocular air (closed arrow)
injury when one cannot visualize the posterior segment. and foreign body (open arrow).
Perforating injuries are through-and-through injuries of the eye
(Fig. 173.9). They are often the result of missile injuries and the
perforating object generally comes to rest in the orbit. The exit
site for perforating injuries is often the posterior segment of the endothelial damage,44,45 complement-induced granulocyte
eye, and these injuries carry a very poor prognosis. aggregation,46 air embolism associated with crushing chest
injuries,44 and fat embolism associated with long bone frac-
Key Features: Open Globe Injuries tures47 have all been proposed as possible mechanisms. Patients
• Open globe injuries are classified as lacerating if they are the with acute pancreatitis may have associated fat emboli and
result of sharp force (i.e., nail injury) or rupture if the eyewall present with a Purtscher-like retinopathy.48 The complement
opens as a result of blunt force system is also activated in acute pancreatitis, as well as in
• Ruptured globes carry a poor prognosis severe trauma,26 which lends support to the theory of retinal
• Lacerating open globe trauma is subclassified into penetrating, intravascular clotting as the mechanism of Purtscher ’s
perforating, and intraocular foreign body (IOFB) injuries retinopathy.

Key Features: Purtscher’s Retinopathy


MISCELANEOUS MECHANISM OF RETINAL • Characterized by multiple patches of superficial retinal
INJURY whitening and retinal hemorrhages surrounding the optic nerve
• Theories to explain the pathogenis of Purtscher’s retinopathy
PURTSCHER’S RETINOPATHY include:
Purtscher described patches of superficial retinal whitening, • Retinal vascular injury with endothelial damage
intraretinal hemorrhages, and disk edema in five patients after • Complement-induced granulocyte aggregation
severe compression injury to the head in 1910.43 The charac- • Air embolism associated with crushing chest injuries
teristic fundus changes may be seen immediately and may • Fat embolism associated with long bone fractures
continue to progress for 1–2 day following trauma.21 Presenting
visual acuity may range from 20/20 to count fingers and
findings typically are bilateral although unilateral cases have
TERSON’S SYNDROME
been described.21 Terson’s syndrome is the term used to describe the association
The characteristic retinal findings in Purtscher’s retinopathy of retinal and vitreous hemorrhage with subarachnoid and
are multiple patches of superficial retinal whitening and retinal subdural hemorrhage, first described by Terson in 1900.49
hemorrhages surrounding the optic nerve. The disk may appear Approximately 20% of patients with spontaneous or
normal initially, but an afferent pupillary defect may be present, posttraumatic subarachnoid hemorrhage will present with
and later optic disk edema followed by atrophy may develop. intraocular hemorrhage.50 These hemorrhages are typically
Fluorescein angiography may show leakage of dye in the region located between the ILM and the rest of the neurosensory retina
of the white retinal patches, retinal and disk edema, venous and may occasionally break into the vitreous cavity. The blood
staining, and areas of capillary nonperfusion. The retinal usually clears spontaneously; however, in cases of nonclearing
hemorrhages and white patches resolve over several months, vitreous hemorrhage, vitrectomy may be beneficial.51
although the patient may be left with some loss of vision The mechanism of intraocular hemorrhage in Terson’s
secondary to pigmentary macular changes and optic atrophy.44 syndrome is thought to be due to significant hemorrhage from
Several theories have been advanced to explain the patho- epipapillary and peripapillary capillaries that rupture after a
2192 genesis of Purtscher’s retinopathy. Retinal vascular injury with sudden increase in venous pressure resulting from acute
Traumatic Retinopathy

elevation of intracranial pressure after intracranial hemor-


Key Features: Valsalva Retinopathy
rhage.49 The hemorrhage may spread to the subretinal space,
• Retinal hemorrhages observed in association with heavy lifting,
within the retina, the subretinal limiting membrane space, the
coughing, vomiting, vigorous sexual activity or straining during
subhyaloid space, or vitreous cavity.52–55
bowel movement
• Characterized by well-circumscribed, round or dumbbell-
shaped red elevation beneath the internal limiting membrane or
Key Features: Terson’s Syndrome intraretinal hemorrhage in or near the fovea
• Term used to describe retinal and vitreous hemorrhage
associated with subarachnoid and subdural hemorrhage
• Hemorrhages are located between the internal limiting
membrane and the neurosensory retina but may break into the
vitreous cavity
• Prognosis for visual return is generally good, however,
vitrectomy may be indicated for some cases

VALSALVA RETINOPATHY

CHAPTER 173
Duane first used the term Valsalva hemorrhagic retinopathy
in 197256 to describe the retinal hemorrhages observed in
association with heavy lifting, coughing, vomiting, vigorous
sexual activity or straining during bowel movement.57
Patients may note a sudden decrease in vision associated with
these activities resulting in Valsalva retinopathy. Typical fundus
findings include a well-circumscribed, round or dumbbell-
shaped red elevation beneath the ILM or intraretinal
hemorrhage in or near the fovea. There may be associated
vitreous hemorrhage or dissection of blood beneath the retina.
The blood is initially bright red with a prominent light reflex
on the surface, but it may turn yellow after several days or
weeks (Fig. 173.10). A fluid level may develop early with settling FIGURE 173.10. Valsalva maculopathy. This 22-year-old man noted
of red blood cells.6 The visual prognosis is good, with most decreased vision and a central scotoma immediately after a coughing
episode 3 weeks before presentation. Visual acuity was 20/50. Note
patients returning to normal vision.
the bilobed appearance of the preretinal hemorrhage, now yellow
The presumed mechanism of retinal hemorrhage is rupture because of hemolysis. Along the inferior margin of the hemorrhage,
of superficial retinal capillaries owing to a sudden increase in blood can be seen breaking through the internal limiting membrane
retinal venous pressure after the rapid increase in intrathoracic and extending into the vitreous cavity inferiorly. One month later, the
or intraabdominal pressure associated with a Valsalva hemorrhage had almost completely resolved, and vision improved
maneuver, such as coughing or vomiting. to 20/20.

REFERENCES
1. Aguilar JP, Green WR: Choroidal rupture. 9. Bastek JV, Foos RY, Heckenlively J: 18. Rubin JS, Glaser BM, Thompson JT,
A histopathologic study of 47 cases. Retina Traumatic pigmentary retinopathy. Am J et al: Vitrectomy, fluid-gas exchange and
1984; 4:269–275. Ophthalmol 1981; 92:621–624. transforming growth factor beta-2 for
2. von Graefe A: Zwei Falle von Ruptur der 10. Cogan DG: Pseudoretinitis pigmentosa: the treatment of traumatic macular
Choroidia. Graefes Arch Ophthalmol 1854; report of two traumatic cases of recent holes. Ophthalmology 1995;
1:402. origin. Arch Ophthalmol 1969; 81:45–53. 102:1840–1845.
3. Ament CS, Zacks DN, Lane AM, et al: 11. Pulido JS, Blair NP: The blood–retinal barrier 19. Garcia-Arumi J, Corcostegui B, Cavero L,
Predictors of visual outcome and choroidal in Berlin’s edema. Retina 1987; 7:233–236. et al: The role of vitreoretinal surgery in the
neovascular membrane formation after 12. Hart JCD, Frank HJ: Retinal opacification treatment of posttraumatic macular hole.
traumatic choroidal rupture. Arch after blunt non-perforating concussional Retina 1997; 17:372–377.
Ophthalmol 2006; 7:957–66. injuries to the globe: A clinical and retinal 20. Aaberg TM, Blair CJ, Gass JDM:
4. Fuller B, Gitter KA: Traumatic choroidal fluorescein angiographic study. Trans Macular holes. Am J Ophthalmol 1970;
rupture with late serous detachment of the Ophthalmol Soc UK 1975; 95:94–100. 69:555–562.
macula: Report of successful argon laser 13. Sipperley JO, Quigley HA, Gass JD: 21. Elliot D, Avery RL: Issues in ocular trauma-
treatment. Arch Ophthalmol 1973; Traumatic retinopathy in primates: The Nonpenetrating posterior segment trauma.
89:354–355. explanation of commotio retinae. Arch Ophthalmol Clin North America 1995;
5. Harissi-Dagher M, Sebag M, Gauthier d, Ophthalmol 1878; 96:2267–2273. 8:647–667
Marcil G, et al: Photodynamic therapy in 14. Eagling EM: Ocular damage after blunt 22. Goldzieher W: Beitrag zur Pathologie der
young patients with choroidal trauma to the eye: its relationship to the orbitalen Schussverletzungen. Z Augenh
neovascularization following traumatic nature of the injury. Br J Ophthalmol 1974; 1901; 6:277.
choroidal rupture. Am J Ophthalmol 2005; 58:126–140. 23. Richards RD, West CE, Meisels AA:
139:726–8. 15. Aaberg TM: Macular holes. Surv Chorioretinitis sclopetaria. Trans Am
6. Berlin R: Zur sogenannten Commotio Ophthalmol 1970; 15:139:162. Ophthalmol Soc 1968; 66:214–232.
retinae. Klin Monatsbl Augenheilkd 1873; 16. Gass JDM: Stereoscopic atlas of macular 24. Richards RD, West CE, Meisels AA:
1:42–78. diseases. St Louis: CV Mosby; 1970:94–96. Chorioretinitis sclopetaria. Am J
7. Gass JDM: Stereoscopic atlas of macular 17. Johnson RN, McDonald HR, Lewis H, et al: Ophthalmol 1968; 66:852–860.
diseases: diagnosis and treatment. 3rd edn. Traumatic macular hole: observations, 25. Martin DF, Awh CC, McCuen BW, et al:
St Louis: CV Mosby, 1987:170, 552–565. pathogenesis and results of vitrectomy Treatment and pathogenesis of traumatic
8. Duke-Elder S: System of ophthalmology. surgery. Ophthalmology 2001; chorioretinal rupture sclopetaria. Am J
St Louis: CV Mosby, 1972:165. 108:853–857. Ophthalmol 1994; 117:190–200. 2193
RETINA AND VITREOUS

26. Williams DF, Mieler WF, Williams GA: 39. Harcourt B, Hopkins D: Permanent 48. Inkeles DM, Walsh JB: Retinal fat emboli as
Posterior segment manifestations of ocular chorioretinal lesions in childhood of a sequelae to acute pancreatitis. Am J
trauma. Retina 1990; 10:S35–S44. suspected origin. Trans Ophthalmol Soc Ophthalmol 1975; 80:935–938.
27. Cox MS, Schepens CL, Freeman HM: UK 1973; 93:199–205. 49. Terson A: De l’hemorrhagie dans le corps
Retinal detachment due to ocular 40. Emerson MV, Pieramici DJ, Stoessel KM, vitre au cours de l’hemorrhagie cerebrale.
contusion. Arch Ophthalmol 1966; et al: Incidence and rate of disappearance Clin Ophthalmol 1900; 6:309.
76:678–685. of retinal hemorrhage in newborns. 50. Shaw HE Jr, Landers MB III, Sydnor CF:
28. Goffstein R, Burton TC: Differentiating Ophthalmology 2001; 108:36–39. The significance of intraocular
traumatic from nontraumatic retinal 41. Kuhn F, Morris R, Witherspoon CD: hemorrhages due to subarachnoid
detachment. Ophthalmology 1982; Birmingham eye trauma terminology hemorrhage. Ann Ophthalmol 1977;
89:361–368. (BETT): terminology and classification of 9:1403–1405.
29. Ross WH: Traumatic retinal dialyses. mechanical eye injuries. Ophthalmol Clin 51. Clarkson JG, Flynn HW Jr, Daily MJ:
Arch Ophthalmol 1981; 99:1371–1374. North Am 2002; 15:139–143. Vitrectomy in Terson’s syndrome. Am J
30. Scott JD: Retinal dialysis. Trans 42. Pieramici DJ, Au Eong KG, Sternberg P Jr, Ophthalmol 1980; 90:549–552.
Ophthalmol Soc UK 1997; 97:33–35. et al: The prognostic significance of a 52. Khan SG, Frenkel M. Intravitreal
31. Lister W: Some concussive changes met system for classifying mechanical injuries hemorrhage associated with rapid increase
with in military practise. Br J Ophthalmol of the eye (globe) in open-globe injuries. in intracranial pressure. J Neurosurg 1974;
1924; 8:305. J Trauma 2003; 54:750–54. 41:167–176.
32. Caiger H: Ocular injuries resulting from the 43. Purtscher O: Angiopathia retinae 53. Muller PJ, Deck JHN: Intraocular and optic
war. Trans Ophthalmol Soc UK 1941; traumatica. Lymphorrhagien des nerve sheath hemorrhage in cases of
61:54–73. Augengrundes. Graefes Arch Clin Exp sudden intracranial hypertension.
SECTION 10

33. Delaney WV, Geiss M: Partial evulsion of Ophthalmol 1912; 82:347–371. J Neurosurg 1974; 41:160–166.
the optic nerve. Ann Ophthalmol 1988; 44. Burton TC: Unilateral Purtscher’s 54. Toosi SH, Malton M: Terson’s syndrome-
20:371–372. retinopathy. Ophthalmology 1980; significance of ocular findings. Ann
34. Chow AY, Goldberg MF, Frenkel M: 87:1096–1105. Ophthalmol 1987; 19:7–12.
Evulsion of the optic nerve in association 45. Kelley JS: Purtscher’s retinopathy related 55. Weingeist TA, Goldman EJ, Folk JC, et al:
with basketball injuries. Ann Ophthalmol to chest compression by safety belts: Terson’s syndrome. Clinicopathologic
1971; 72:969–971. Fluorescein angiographic findings. Am J correlations. Ophthalmology 1986;
35. Assaf AA: Traumatic retinal detachment. Ophthalmol 1972; 74:278–283. 93:1435–1442.
J Trauma 1985; 25:1085–1089. 46. Jacob HS, Craddock PR, 56. Duane TD: Valsalva hemorrhagic
36. Harley RD: Ocular manifestations of child Hammerschmidt DE, Moldow CF: retinopathy. Trans Am Ophthalmol Soc
abuse. J Pediatr Ophthalmol Strabismus Complement induced granulocyte 1972; 70:298–313.
1980; 17:5–13. aggregation. An unsuspected mechanism 57. Markovits AS: Sudden visual loss
37. Friendly DS: Ocular manifestation of the of disease. N Engl J Med 1980; associated with sexual activity. Arch
physical child abuse. Trans Am Acad 302:789–794. Ophthalmol 1996; 114: 106.
Ophthalmol Otolaryngol 1971; 75:318–332 47. Urbanek J: Uber Fettembolie des Auges.
38. Jensen AD: Ocular clues to child abuse. Graefes Arch Clin Exp Ophthalmol 1934;
J Pediatr Ophthalmol 1971; 8:270. 131:147–173.

2194
CHAPTER

174 Photic Retinopathy


Martin A. Mainster and Michael E. Boulton

Light can cause photomechanical, photothermal, or photo-

Spectral dependence of phototoxicity or absorption


chemical retinal damage.1–4 Photochemical (actinic) retinal 1
injury occurs at retinal temperature elevations too low for tissue
UV-blue phototoxicity
coagulation (photocoagulation).5–10 It is generally termed photic Lipofuscin
retinopathy or retinal phototoxicity when it occurs without 0.8 All-trans-retinal
exogenous photosensitizers. Photic retinopathy is produced Cytochrome c
experimentally by prolonged intense light exposures ranging Porphyrin
0.6
from seconds to hours at illuminances exceeding normal Xanthophyll
environmental levels that would probably be well tolerated if
experienced only briefly. Solar eclipse and welding arc injuries 0.4 Blue-green phototoxicity
are caused by retinal phototoxicity. Photochemical retinal
damage has also provided valuable insight into the molecular
biology of retinal degeneration.11,12 This chapter summarizes 0.2 Melinin

current understanding of the pathogenesis and clinical con-


sequences of photic retinopathy. 0
UV Violet Blue Green Yellow Orange Red
<400 400-440 440-500 500-570 570-590 590-610 610-700
OPTICAL INTERACTIONS AND
PROTECTION 350 400 450 500 550 600 650 700
Wavelength (nm)
Key Features FIGURE 174.1. Acute UV-blue type,9,55,245 lipofuscin,24,57 and all-trans-
• Thermal (photocoagulation) and photochemical (phototoxicity) retinal phototoxicity rise rapidly in the violet (400–440 nm) part of the
effects can damage the retina spectrum, where porphyrin24,246 and cytochrome c oxidase24,247
• Absorption spectra describe how effectively chromophores phototoxicities peak and macular xanthophyll protection declines.248
such as melanin or lipofuscin absorb light of different The action spectrum of the blue-green type retinal phototoxicity peaks
wavelengths at 500 nm, similar to scotopic luminous efficiency (sensitivity),
because rhodopsin mediates both processes. The absorption spectra
• Action spectra describe the relative effectiveness of
of rhodopsin in rod photoreceptors and melanopsin in light-sensitive
wavelengths in causing acute retinal phototoxicity ganglion cells are similar in form but peak at ~500 nm249 (blue-green)
• Photosensitizers are molecules that generate reactive oxygen and 480 nm (blue),67–69 respectively. Blue-green type phototoxicity in
species upon light absorption experimental studies occurs at lower retinal irradiances than UV-blue
• The cornea and crystalline lens shield the retina from UV type phototoxicity.26 The absorption spectrum for xanthophyll
radiation decreases rapidly with decreasing wavelength from its peak
absorption at 460 nm.44–47 Melanin absorption, shown for adults over
50 years of age, increases with decreasing wavelength in the visible
The retina is potentially vulnerable to damage from ultraviolet and near-UV part of the spectrum, similar to that of lipofuscin
radiation (UV; l < 400 nm), visible light (400 nm < l < 700 nm), absorption.22–24 The spectral dependence of phototoxicity and
and infrared radiation (IR; l > 700 nm). Violet (400–440 nm) absorption are shown, but ordinate (y-axis) units are arbitrary, so the
relative magnitudes of different curves are not directly comparable in
and blue (440–500 nm) light comprise the shorter wavelength
this figure.
portion of the visible spectrum.13,14 Retinal effects depend on
the wavelength (nm), power (W), duration (s), lateral extent
(cm), and location (macular vs elsewhere) of optical radiation
exposures.9,10,15–17 Photochemical effects also depend on oxygen retinal light absorbers include lipofuscin, rhodopsin, cone photo-
tension18,19 and body temperature.7,20,21 The magnitude of a pigments, melanopsin, porphyrins, and cytochrome c oxidase.
retinal exposure is usually specified in terms of its irradiance Absorption spectra describe how effectively tissue chromo-
(power density in W/cm2) or fluence (energy density in J/cm2). phores capture photons at different wavelengths. Absorption
Thermal and photochemical retinal damage occur when pho- spectra for molecules such as hemoglobin or rhodopsin have
tons are absorbed by chromophores, the light absorbing com- peaks at specific wavelengths, with lower absorption at adjacent
ponents of biomolecules. Melanin, hemoglobin, and macular wavelengths. Alternatively, absorbers such as melanin and lipo-
xanthophyll are the most effective retinal light absorbers. Other fuscin have absorption spectra that increase steadily with
2195
RETINA AND VITREOUS

decreasing wavelength (increasing photon energy).22–24 Figure pigment epithelium (RPE) cells.37,38 It is composed of the
174.1 presents examples of both patterns. When photon carotenoid pigments lutein and zeaxanthin, which can limit
absorption induces damaging reactive oxygen species, spectra photosensitization reactions in the macula either by reducing
that describe the effectiveness of photon capture at different the amount of incoming short wavelength light or by directly
wavelengths are known as action spectra. Action spectra charac- quenching reactive oxygen species generated by optical
terize how effectively different optical radiation wavelengths radiation.38–40 Xanthophyll density declines rapidly with
produce photochemical effects.25 increasing retinal eccentricity from its foveolar peak, except for
Photon absorption excites electrons in target molecules to a ring of increased extrafoveolar density that is more common
higher energy states. Excited molecules release their excess in women and may be related to the anatomy of the foveolar
energy in different ways. Energy can be converted into increased depression.37,41–43 Light absorption by xanthophyll also decreases
average molecular vibrational motion, raising retinal tempera- rapidly with decreasing wavelength from its peak absorption
ture and causing photocoagulation when tissue temperature at 460 nm.44–47 Thus, macular xanthophyll protection is least
elevation is sufficiently high.4 Vibrational energy transferred to effective in the potentially hazardous violet and UV parts of the
neighboring molecules diffuses heat beyond the directly irradiated spectrum,14,48 an issue of increased significance when the
area in a process known as heat conduction. Alternatively, if crystalline lens is removed in cataract surgery. Studies of how
photon energy is coupled effectively to chemical bonds in target macular xanthophyll varies with retinal aging and AMD have
molecules, excess molecular electronic energy from photon produced inconsistent results.49–52
absorption can break those bonds, producing photochemically
SECTION 10

induced structural changes in the molecules.26 MECHANISMS OF PHOTIC INJURY


Reactive oxygen species in the eye can be formed as a result
of (1) the interaction of ionizing radiation with biological
molecules, (2) an unavoidable byproduct of cellular respiration, Key Features
(3) synthesis in phagocytic cells such as neutrophils and macro- • Photic retinopathy is mediated by either retinal photopigments
phages, or (4) a respiratory burst during phagocytosis. Reactive (rod, cone or ganglion) or retinal photosensitizers (such as
oxygen species are highly reactive oxygen radicals with the lipofuscin, cytochrome c oxidase and all-trans-retinal)
capacity to modify and damage cell membranes, proteins, • The action spectrum of blue-green phototoxicity peaks around
carbohydrates, and nucleic acids. Most reactive oxygen species 500 nm (blue-green) because it is mediated by the rod
(e.g., hydroxyl radicals, superoxide anions, and lipid hydro- photopigment rhodopsin
peroxides) have an unpaired electron in their outer orbital and • UV-blue phototoxicity increases with decreasing wavelength,
are referred to as free radicals. In addition, there are oxygen so UV radiation is more hazardous than violet light which is
species in which electron pairing is normal but the molecule is more hazardous than blue light
in an excited state (e.g., singlet oxygen and hydrogen peroxide). • UV-blue phototoxicity requires 100 times more retinal
Singlet oxygen is the lowest excited state of the di-oxygen irradiance (power density) than blue-green phototoxicity
molecule, its lifetime in solution is in the microsecond range, • International light hazard standards incorporating photic
and it reacts readily with membrane lipids. Reactive oxygen retinopathy are based on acute UV-blue phototoxicity
species can react either directly with target tissues (type-1
photochemical or free radical reactions) or with molecular
oxygen to produce singlet oxygen or superoxide which then
reacts with target tissues (type-2 photochemical or photo- CLASSIC PHOTOTOXICITIES
dynamic reactions).26–28 Molecules that produce reactive oxygen
species upon light absorption are referred to as photosensitizers. There are two classic types of photic retinopathy. Blue-green
Photochemical damage to retinal cells can lead to cellular retinal phototoxicity in rodents was discovered by Werner Noell
dysfunction or death. The latter normally occurs by a process in 1966.5,20 Its action spectrum is similar to scotopic visual
termed apoptosis, which is a common cell death pathway not only sensitivity,53,54 which peaks at 500 nm (blue-green), because
for photic retinopathy but also conditions such as age-related rhodopsin mediates both processes. Blue-green phototoxicity
macular degeneration (AMD) and retinitis pigmentosa.11 Nor- is also termed ‘class 1’, ‘Noell-type’ or ‘white light’ photic
mal cellular homeostasis balances cell proliferation and death. retinopathy. Damage is typically located in the photoreceptor
Apoptosis removes unneeded, injured, or diseased cells, span- layer or both the photoreceptor and RPE layers.5
ning a broad complex of interrelated mechanisms for signaling The second classic type of phototoxicity was discovered in
and producing cell death.11,29 Analyses of molecular mechan- young primates by William Ham and his colleagues in
isms of rodent retinal phototoxicity provide insights into the 1976.9,16,26,55–57 UV-blue phototoxicity has an action spectrum
genetics of inherited retinal disorders.11,12 that increases with decreasing wavelength, similar to that of
Ocular media protect the retina from potentially harmful lipofuscin which may be one of its primary mediators.23,24,57
optical radiation. The cornea shields internal ocular structures Thus, UV radiation is more hazardous than violet light, which
by blocking UV radiation below 300 nm.30 The crystalline in turn is more hazardous than blue light. UV-blue photo-
lens protects the retina from UV radiation between 300 and toxicity is also termed ‘class 2’, ‘Ham-type’, or ‘blue light
400 nm,30–32 except in young eyes with clear crystalline lenses hazard’ retinal phototoxicity. Damage is typically located in the
that transmit some UV around 320 nm to the retina.30 The RPE or both the RPE and photoreceptor layers.9,58
aging crystalline lens attenuates an increasing amount of visible UV-blue phototoxicity requires ~100 times more retinal
light, particularly at shorter violet and blue wavelengths.30–34 irradiance than blue-green phototoxicity.16,26,59,60 UV-blue photo-
Additional extraretinal protection is provided by eyebrow sha- toxicity is the basis for the international consensus standard
dowing, corneal reflection of light not incident perpendicular to Al phototoxicity function used to estimate acute industrial
its surface (Fresnel’s law), and pupillary, aversion, squint, and retinal phototoxicity risks.61 Blue-green and UV-blue photic
blink responses.35,36 retinopathies are archetypes of photopigment-mediated and
Macular xanthophyll is a yellowish pigment in the cone axon photosensitizer-mediated retinal photoxicity damage mech-
and inner plexiform layers of the macula that reduces the anisms, respectively.9,16,20,26,59 Figure 174.1 illustrates the action
2196 short wavelength light exposure of photoreceptors and retinal spectra of the classic retinal phototoxicities.
Photic Retinopathy

PHOTOPIGMENT-MEDIATED DAMAGE toxicity susceptibility than rods in rodents87 and greater suscep-
There are three types of retinal photopigments: (1) cone photo- tibility in pigeons and primates.15,88 These latter observations
receptor photopigments that provide photopic (bright light) and are not consistent with the greater resistance of cones than rods
mesopic (intermediate light) vision,62,63 (2) rhodopsin in rod to photic injury in a histopathologic study of human solar
photoreceptors responsible for mesopic and scotopic (dim light) retinopathy,89 possibly because of the relatively small number of
vision,53,54 and (3) melanopsin in blue-light sensitive retinal photoreceptors per RPE cell in the primate foveola.90 Repeated
ganglion cells that modulate circadian photoentrainment, intermittent blue or green light stimulation in primates causes
pupillary function, and possibly conscious vision.64–67 Cone selective blue or green cone damage, respectively.91 Photo-
photoreceptors in humans have absorption maxima around sensitive retinal ganglion cells which modulate circadian
426 nm (blue), 530 nm (green), and 552 or 557 nm (red).62,63 rhythmicity92–95 may also be susceptible to phototoxicity
Rod and light-sensitive retinal ganglion photoreceptors have peak mediated by their photopigment melanopsin.4
spectral sensitivities of ~ 500 nm (blue-green)53,54 and 480 nm
(blue),67–69 respectively.
A photoreceptor photopigment consists of an 11-cis retinal PHOTOSENSITIZER-MEDIATED DAMAGE
chromophore molecule bound to a transmembrane opsin The retina is vulnerable to oxidative stress because of its high
protein in a photoreceptor outer segment disk.70 Differences oxygen and light levels. Its internal defenses include: (1) circadian
in rod, cone, and retinal ganglion photopigment absorption shedding of damaged photoreceptor outer segment disks,
maxima results from different amino acid sequences in the (2) enzymatic antioxidants, such as superoxide dismutase,

CHAPTER 174
opsin molecule adjacent to the 11-cis retinal binding site.71 catalase, and glutathione peroxidase, (3) nonenzymatic anti-
When a photopigment molecule absorbs a photon, 11-cis retinal oxidants including a-tocopherol, ascorbate, lutein, and zeaxanthin,
is converted to all-trans retinal, initiating a conformational and (4) light-absorbing molecules such as melanin in the
change in opsin and a sequence of transient photopigment RPE and choroid or macular xanthophyll in the inner
states that include metarhodopsin II (activated rhodopsin).70,72 retina.16,24,26,77,96–98 Retinal phototoxicity may be mediated by
Metarhodopsin II catalyzes the activation of transducin, which photosensitizers such as lipofuscin, all-trans-retinal, cyto-
continues the phototransduction cascade that converts absorbed chrome c oxidase, and the porphyrins.24,99–101 As shown in
photon energy into neurochemical signals.73 Metarhodopsin II Figure 174.1, absorption spectra for these photosensitizers
decays through hydrolysis, releasing all-trans retinal from its increase with decreasing wavelength or peak typically in the
opsin binding site. All-trans-retinal is reduced to all-trans- near-UV or violet part of the spectrum.
retinol, and then re-isomerized and oxidized back to its original Lipofuscin accumulates with aging in the RPE because
11-cis retinal in a complex series of steps involving the RPE for undegradable endproducts of either photoreceptor phagocytosis
rods and Muller cells for cone photoreceptors.74,75 In general, a or autophagy of spent intracellular organelles such as mito-
functional retinoid cycle is needed for both retinal photorecep- chondria and endoplasmic reticulum cannot be removed effec-
tion and phototoxicity. tively from cells. Lipofuscin may impair antioxidant activity,
Most experimental studies of photopigment-mediated retinal produce phototoxic damage or mechanically compromise
phototoxicity use rodent models and protocols that differ in cellular function.24,102,103 It is a photoinducible generator of
retinal irradiance, exposure duration, source wavelength, animal reactive oxygen species that can damage RPE cells.24 Lipofuscin
species, antioxidant status, and circadian timing.5,11,12,20,71,76,77 is comprised of many fluorophores, including the pyridinium
Studies using knockout mice have demonstrated apoptotic bisretinoid A2E, which has received considerable recent
pathways for photic damage that may or may not involve attention. A2E is a retinoid cycle byproduct, formed from
phototransduction.78,79 phosphatidylethanolamine and two molecules of all-trans-
Light damage in mice requires effective retinoid recycling. retinaldehyde.104 The action spectrum of A2E peaks around
The RPE65 protein is needed for the production of 11-cis 430 nm in the violet part of the spectrum. (cf Fig. 174.1)104,105
retinal. RPE65–/– knockout mice without this protein are pro- A2E and its derivatives are only weakly photoreactive compared
tected against photic retinopathy.11,71 Photochemical damage to lipofuscin and thus account for only a small fraction of the
thresholds depend on RPE65 protein levels in mice but not in total photoreactivity of lipofuscin.57,106
rats.12 The susceptibility of rats to photic injury has a circadian All-trans-retinal is a potent intraretinal photosensitizer
rhythm, with dark adaptation increasing the risk of damage.80 produced during the normal retinoid cycle. It plays an essential
Many other molecular, pharmacological, and environmental role in A2E and lipofuscin formation.71 Increased all-trans-
factors influence rodent retinal phototoxicity.11,71 retinal concentration during prolonged bright light exposure107
Most rodent retinas adapt to ambient illumination, a may cause photic retinopathy.71 Other potential intraretinal
phenomenon known as photostasis.81–84 For example, changing photosensitizers include porphyrin containing molecules such
from bright to dim environments upregulates rhodopsin and as hemoglobin and enzymes, including cytochrome c oxidase.
increases rod rhodopsin regeneration rates, rod outer segment Cytochrome c oxidase is an important respiratory chain enzyme
lengths, and rhodopsin concentration in rod outer segments.82 located in mitochondria and photoreceptors and may contribute
The reverse is true for changes from a dim to a bright environ- to short wavelength light-induced mitochondrial damage in
ment. A significant finding is that local rhodopsin concentra- the RPE.108 The role of melanin as a photosensitizer or
tion across a rodent retina is inversely proportional to local photoprotective agent in the RPE remains unclear but there is
illuminance.82 Autophagic degradation of opsin may contribute evidence that it can become a photosensitizer in older
to downregulation of rhodopsin when ambient illumination is adults.24,60,109–111
increased, possibly reducing the risk of retinal phototoxicity.85
Bleached rhodopsin is more thermally labile than the CLINICAL SYNDROMES
unbleached photopigment, so bright sustained illumination
that increases the percentage of bleached photopigment could Solar and operating microscope exposures performed on eyes
contribute to increased opsin misfolding86 and phototoxic prior to enucleation for malignant melanoma have proven that
injury.8 these light sources can cause phototoxic injuries89,112,113 at retinal
Cone photopigment-mediated phototoxicity data are limited. temperature elevations too low for thermal damage.4,114–116
Experimental studies have shown that cones have lower photo- Welding arc and solar maculopathies are similar, as are operating 2197
SECTION 10 RETINA AND VITREOUS

FIGURE 174.3. Welding arc injuries typically produce yellowish-white


FIGURE 174.2. An acute solar injury produces a yellowish-white
foveolar lesions similar to those seen in solar maculopathy, as
foveola lesion, as seen in this 15-year-old female with 20/70 visual
illustrated in this 17-year-old male 3 days after a welding arc
acuity.
exposure.
Photograph courtesy of Raj Lucas, MD and the Medical Photographic Imaging
Centre, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia.

Key Features
• Clinical photic retinopathy is an acute injury that occurs when
retinal defenses are overwhelmed by brilliant light exposures
• The four well identified clinical examples of photic retinopathy
are solar, welding, operating microscope and endoilluminator
injuries
• Solar and welder’s maculopathies share similar clinical
features, as do operating microscope and endiolluminator
injuries
• The primary sites and mediators of clinical photic injuries are
as yet undetermined
• Most large epidemiologic studies fail to support a significant
role for environment light exposure in AMD
• Minimizing the duration and intensity of operating microscope
and endoilluminator exposures reduces the risk of photic
retinopathy

microscope and endoilluminator injuries.4 The role of light in FIGURE 174.4. Optical coherence tomograms after welding arc and
AMD has yet to be proven despite decades of investigation.4,48 solar injuries typically show foveal abnormalities affecting the outer
neural retina and RPE, as seen in this 17-year-old male with a
localized hyporeflective defect 8 months after a welding arc exposure.
SOLAR AND WELDER’S MACULOPATHY Photograph courtesy of Raj Lucas, MD., and the Medical Photographic Imaging
Centre, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia.
The solar disk forms a 160 mm diameter retinal image, ~20%
of the area of the 350 mm diameter foveola.114,117 Unassisted
observation of the sun at its zenith with a 3 mm pupil diameter several weeks. Months later, there may be foveal distortion,
produces only a 4°C retinal temperature rise, well below the lamellar defects, pigment mottling, a macular hole or no appa-
10°C temperature elevation needed for threshold retinal rent damage.121 Lesion severity depends on viewing circum-
photocoagulation.114,118 Thus, solar retinopathy is usually stances and an individual’s defense mechanisms. The worst
caused by retinal phototoxicity, not photocoagulation. Solar injuries occur with prolonged observation and good fixation, as
observation with a dilated 7 mm pupil or telescope-assisted when the solar disk is foveated through a defective optical filter.
solar inspection can cause significantly higher retinal tem- Foveal RPE defects can be demonstrated on fluorescein angio-
perature increases and photocoagulation injuries.1,114,118 grams after severe solar injuries, but angiograms are often
Solar injuries can be severe or mild.119,120 Visual acuity is normal.121,126 Optical coherence tomography typically shows
typically decreased after an injury to 20/40 to 20/200, usually foveal abnormalities affecting the outer neural retina and
returning to 20/20 to 20/40 over 6 months.121,122 Injuries have RPE.124,126–128
been reported after sungazing during drug abuse123,124 and Histopathologic studies of the eyes of volunteers who gazed
hypoglycemia.125 They can occur with or without eclipses.4 at the sun prior to ocular enucleation for choroidal melanoma
Pupil dilation during an eclipse makes eclipse observation show predominantly rod and cone photoreceptor damage,
particularly hazardous.1 There are several safe methods for including vesiculation and fragmentation of outer segment
viewing solar eclipses.1,117 lamellae, mitochondrial swelling and nuclear pyknosis.89,112
Acute solar injuries produce yellowish-white foveolar lesions, Cones are more resistant to damage than rods, perhaps
2198 as illustrated in Figure 174.2.112,121 Lesions gradually fade over accounting for good visual acuity after many solar injuries.89
Photic Retinopathy

damage.30,144 UV may also be responsible for the retinal injury


from an intense flash due to a short-circuiting high-tension
electric circuit.145

OPERATING MICROSCOPE AND


ENDOILLUMINATOR MACULOPATHY
Intense visible light from operating microscopes can produce
retinal lesions that are usually oval shaped and 1–2 optic disk
diameters in lateral extent.146,147 The long axis of the lesion
often has the same orientation as the filament of the operating
microscope’s lamp.148 During cataract surgery, lesions typically
occur in the inferior macula because of microscope tilt and
illumination positioning.149,150 Injuries occur after cataract,
cornea, glaucoma, and retinal procedures.146,147,151–155 Fiberoptic
endoilluminators in vitreoretinal surgery can produce similar
lesions in different retinal locations,21,156,157 as shown in
Figure 174.5.

CHAPTER 174
Operating microscope maculopathy rates have declined with
improved, faster cataract surgery techniques, but injuries can
still occur in brief procedures.158 Acute lesions usually have a
yellowish-white appearance that fades quickly over several days.
They can be associated with a transient serous detachment of
the neural retina. Pigment mottling, RPE degeneration, and
FIGURE 174.5. The endoilluminator lesion in the temporal macula of
occasionally choroidal neovascularization may develop over
this 68-year-old male is accompanied by cystoid macular edema after months to years at injury sites.4,159 Fluorescein angiography is
epiretinal membrane surgery. useful when a lesion is suspected but not ophthalmoscopically
Fluorescein angiographic image courtesy of Giovanni Staurenghi, MD, apparent.4 Damage was present in the RPE and photoreceptor
Department of Clinical Science Luigi Sacco, University of Milan, Italy. layers of a volunteer’s eye after it was exposed to an intense
operating microscope exposure before ocular enucleation for a
choroidal melanoma.113 Experimental primate studies have
RPE damage can be extensive112 or limited and scattered.89 reported similar findings.160–162 Combined clinical and histo-
Combined clinical and histopathological evidence to date does pathological evidence to date does not permit determination of
not permit determination of whether solar/welding arc whether operating microscope/endoilluminator maculopathy is
maculopathy is a photoreceptor versus RPE injury or a a primarily photoreceptor versus RPE injury or a photopigment-
photopigment-mediated versus photosensitizer-mediated mediated versus photosensitizer-mediated process.
process. The size, location, and severity of operating microscope
The term foveomacular retinitis was originally used to describe lesions determine their visual effects.4,159 Immobilization and
foveal abnormalities resembling solar retinopathy. These higher blood oxygen levels potentially increase the risk of retinal
conditions include whiplash and blunt ocular injuries,129–131 injuries in patients undergoing general rather than local
although similar findings have been reported in people with no anesthesia.18,163 The hazard of a particular light exposure is also
prior history of photic or mechanical trauma.132,133 Foveo- higher with (1) elevated core body temperature,7 (2) increased
macular retinitis was first described during World War II, with chorioretinal pigmentation (absorption of light by pigmented
additional reports between 1966 and 1973.134–137 tissues potentially elevates local chorioretinal temperature),
Experimental photic retinopathy is enhanced by higher core and (3) systemic photosensitizing medications,164,165 including
body temperatures,7,20,21 so the risk of retinal phototoxicity may hydroxychloroquine, hydrochlorothiazide, furosemide, allopurinol,
be increased by light exposures associated with exercise, infec- and the benzodiazepines.166 Diabetes mellitus and hypertension
tion, or warm environments. Increased chorioretinal pigmenta- may also increase injury risks.148,167
tion increases chorioretinal temperature elevations from solar Operating microscope hazard can be decreased by reducing
observation, perhaps increasing the risk or extent of photic illumination, minimizing coaxial exposure duration, and dis-
injury. High local irradiance from direct sungazing places the continuing systemic photosensitizing medications preoper-
foveola at greatest risk for damage from solar observation, atively, when possible.116,160,165,168–170 Positioning a patient’s eye
but solar retinopathy can occur in young sunbathers who in downgaze is potentially useful,165 as is tilting the operating
deny sungazing,138,139 possibly due to Henle layer fiberoptic microscope appropriately,150 using corneal occluders,171–173 and
channeling of short-wavelength photons from perifoveal regions avoiding supplemental oxygen usage and/or elevated patient
to the center of the fovea.46 Fiberoptic transmission increases core body temperatures.
and xanthophyll protection decreases below 450 nm where the Operating microscopes produce little UV radiation,116,174,175
risk of UV-blue type retinal phototoxicity increases.45,46 so UV-blocking filters are of limited value in reducing injury
Photokeratitis is a common welding arc accident, but a foveal risks.176 Some microscopes produce more short wave-
injury as shown in Figures 174.3 and 174.4 is rare.1,115,140 length visible light than others,116 increasing the risk of
Welding arc maculopathy has the same clinical appearance and photosensitizer-mediated UV-blue type phototoxicity. Filtering
course as solar retinopathy.115,141–143 Welder’s maculopathy usually out light with wavelengths below 450 nm reduces the risk of
occurs in young workers at risk for retinal injury because of acute UV-blue type photic retinopathy,168,170,177 but some blue
clear ocular media, occupational inexperience, and inadequate light is useful for surgery and detecting inner retinal
protective filters. Violet and blue light from a welding arc may abnormalities.168,178–180 Filtering out wavelengths greater than
cause most damage, but 320 nm UV transmitted through the 700 nm eliminates unnecessary IR that could thermally
crystalline lens of younger eyes may contribute to the enhance photochemical damage.168,181 2199
RETINA AND VITREOUS

Studies of the relationship between cataract surgery and Additionally, in vivo fundus autofluorescence measurements of
AMD have produced differing results.182–192 Positive patients with nonneovascular AMD and retinitis pigmentosa
studies189,191,192 may have been confounded by the possibility show focal regions of lipofuscin hyperfluorescence prior to the
that cataract surgery was performed for vision loss due to occurrence of RPE atrophy.197 Nonetheless, lipofuscin concen-
AMD rather than cataract.191,193 The AREDS study found no trations are highest adjacent to rather than in the center of the
correlation between cataract surgery and AMD after checking retina, where age-related photoreceptor loss but not AMD is
for the retinal status of all subjects prior to surgery.193,194 If there most prominent.196,223 (3) There are striking similarities in the
is a correlation between AMD and cataract surgery, it is retinal abnormalities caused by AMD and repetitive
probably due to trauma or other sequellae of intraocular surgery experimental acute phototoxicity.213 214,226,227 This similarity is
and operating microscope illumination on aged susceptible not surprising, however, because the retina and choroid have
maculas.189,191,192 only a limited repertoire of responses to injury, inflammation,
and senescence.
An important failing of the phototoxicity-AMD hypothesis is
AGE RELATED MACULAR DEGENERATION the lack of supportive epidemiological evidence despite two
Geographic RPE atrophy and choroidal neovascularization cha- decades of careful investigation. Two large population-based
racterize the nonneovascular ‘dry’ and neovascular ‘wet’ forms studies did find a weak correlation,228,229 but four other large
of AMD, respectively. RPE lipofuscin accumulation increases studies did not,208,230–232 including a recent study by Taylor,208
with aging in a spatial pattern consistent with rod photo- who had initially identified an association between light expo-
SECTION 10

receptor density, possibly contributing to the pathogenesis of sure and AMD in the Waterman study.228 Additionally, three
both types of AMD.195–197 Increased lipofuscin may (1) com- large case-control studies failed to show that AMD is correlated
promise RPE cell function mechanically, (2) promote expression with environmental light exposure,233–235 one of which actually
of angiogenic factors, or (3) act as a photoinducible generator of found sunlight exposure to be lower in AMD subjects than
reactive oxygen species.24,102,103 Lipofuscin accumulation and controls.234 This lack of evidence is due either to (1) difficulty in
photic retinopathy both probably involve direct and indirect accurately estimating a subject’s cumulative light exposure,
oxidative stress.24,57,198,199 retrospectively, (2) variability in genetic susceptibility, (3) poten-
Rod photoreceptors and RPE cell numbers gradually decline tially obfuscating factors such as differences in the age at which
with retinal aging,102,195 accompanied by increasing thickness subjects experience bright environmental light exposure, or
and hydraulic resistance of Bruch’s membrane200 and decreas- (4) the possibility that phototoxicity is not a significant factor in
ing choriocapillaris vascular diameter.201 Rod photoreceptor loss retinal aging or AMD.
is more prominent in retinal aging and AMD than cone Most ocular UV exposure comes from reflective terrain below
loss.202–204 The density of RPE cells in the peripheral/equatorial or viewing the horizontal sky.25,236 There is strong evidence that
retina decreases with increasing age, but the RPE appears to be environmental UV exposure is a significant risk factor for
relatively protected in the macular region.205 Anatomic and cataractogenesis.237–239 There is additional evidence that UV
physiologic factors beyond ordinary aging conspire to cause exposure, particularly in early life, may increase the risk of
AMD, which is a complex multifactorial process affected by many ocular melanoma.240–242 Using UV-blocking sunglasses and
factors including nutrition, smoking, and genetics.202,206–211. IOLs as well as brimmed hats reduces both those risks242,243
Oxidative stress may injure the RPE and choriocapillaris in Photocarcinogenesis differs from retinal phototoxicity because
AMD and possibly aging, potentially producing chronic local it involves primarily UV-B (280 < l < 315 nm) induced damage
inflammation.207 to DNA molecules240,244 that is not induced by the longer wave-
Phototoxicity from environmental light exposure is a poten- length UV-A (315 < l < 400 nm) and visible optical radiation
tial but unproven cause of AMD.16,24,57,77,103,212–216 One origin that causes retinal phototoxicity.
for this phototoxicity-AMD hypothesis is a 1920 study which If sunlight is a risk factor in retinal aging and AMD, groups
reported that AMD occurred less frequently in cataractous that might potentially benefit from wearing sunglasses and
eyes.212 Later studies showed, however, that the risk of AMD is brimmed hats include: (1) young, lightly pigmented people,
increased in cataractous eyes.189,192 It has been speculated that particularly in warm climates, (2) individuals taking photo-
if light is a factor in the pathogenesis of AMD, then blue-green sensitizing medications, (3) people with malabsorption
type phototoxicity is more likely to be involved than UV-blue syndromes or other problems contributing to malnutrition, and
phototoxicity because photopigment-mediated damage occurs (4) aphakes or pseudophakes. Contemporary IOLs that block
at lower retinal irradiances.217 There are many other theories UV or UV+visible light provide adequate photocarcinogenic
for the pathogenesis of AMD, including those positing the protection against ocular melanoma. Conversely, they provide
primary mechanism to be (1) RPE dysfunction with subsequent 20% less acute phototoxicity protection than a 53-year-old
basal laminar deposits,206 (2) impaired choroidal perfusion,218 crystalline lens that does not prevent AMD.48 Thus, if retinal
(3) genetic defects,219 and (4) retinoid deficiency.204,220 These phototoxicity is a retinal aging or AMD risk factor, pseudo-
theories are of course not mutually exclusive. phakes should wear sunglasses in bright environments regard-
The phototoxicity-AMD hypothesis remains popular despite less of their IOL chromophores.48 Conventional sunglasses are
its limitations. (1) Retinal illumination studies show that cen- not safe for directly viewing the sun.117,168
tral retinal exposure is higher than peripheral exposure under
Ganzfield illumination conditions221 and that superior peri- CONCLUSION
pheral retinal illumination is higher than inferior peripheral
retinal exposure in bright environments.222 Retinal damage is Experimental studies have identified photopigment-mediated
worst centrally in people with AMD,204 but there is no signi- (e.g., Noell-type blue-green) and photosensitizer-mediated (e.g.,
ficant difference between superior and inferior retinal photo- Ham-type UV-blue) photic retinopathies. Clinical photo-
receptor loss with aging,203 and age-related photoreceptor loss toxicities can be divided into solar/welding arc and operating
is most pronounced adjacent to rather than in the center of microscope/endoilluminator injuries. Injuries localized pri-
the retina (possibly due to protective macular pigment).202–204,223 marily to the RPE or photoreceptor layers have been observed,
(2) Lipofuscin accumulates with aging in the RPE, increasing but clinical phototoxicity studies often show that both layers are
2200 the risk of retinal phototoxicity in older adults.102,224,225 affected, due either to multifocal nature of photic effects or the
Photic Retinopathy

suprathreshold exposure of one layer that directly or indirectly responsible for photosensitizer-mediated retinal phototoxicities.
damages the other. Photopigments and photosensitizers are International standards for light exposure in older adults may
distributed throughout the retina, so clinical injuries currently need to be reconsidered because they are based largely on light
cannot be reliably classified as being due to photopigment- exposure studies performed in young primates.
mediated or photosensitizer-mediated processes. Appropriate protective measures are available for solar and
Photopigment-mediated photic retinopathies have been welding arc maculopathies. Minimizing the intensity and dura-
studied most extensively in nocturnal rodent models, where the tion of operating microscope and endoilluminator exposures
primary photopigment is rhodopsin. Nonetheless, it is possible reduces the risk of patient injuries. There is no proof at this
that cone photopigments and light-sensitive ganglion cell time that environmental light exposure is a significant risk fac-
melanopsin may cause clinical phototoxic damage during tor in AMD, but UV exposure is implicated in cataractogenesis
bright, sustained exposures. Lipofuscin in the RPE, all-trans- and possibly ocular melanoma so it is prudent to wear sun-
retinal in the photoreceptor layer and mitochondrial enzymes glasses and a brimmed hat in bright environments such as
throughout the retina such as cytochrome c oxidase may be beaches and skiing slopes.

REFERENCES

CHAPTER 174
1. Sliney DH, Wolbarsht ML: Safety with 17. Sliney DH, Mellerio J, Gabel VP, 32. Mellerio J: Yellowing of the human lens:
lasers and other optical sources: a Schulmeister K: What is the meaning of nuclear and cortical contributions. Vision
comprehensive handbook. New York: threshold in laser injury experiments? Res 1987; 27:1581–1587.
Plenum; 1980. Implications for human exposure limits. 33. Weale RA: Age and the transmittance of
2. Mainster MA: Finding your way in the Health Phys 2002; 82:335–347. the human crystalline lens. J Physiol 1988;
photoforest: laser effects for clinicians. 18. Crockett RS, Lawwill T: Oxygen 395:577–587.
Ophthalmology 1984; 91:886–888. dependence of damage by 435 nm light in 34. Xu J, Pokorny J, Smith VC: Optical density
3. Marshall J: Structural aspects of laser- cultured retinal epithelium. Curr Eye Res of the human lens. J Opt Soc Am A 1997;
induced damage and their functional 1984; 3:209–215. 14:953–960.
implications. Health Phys 1989; 56:617–624. 19. Ruffolo JJ Jr, Ham WT Jr, Mueller HA, 35. Sliney DH: Eye protective techniques for
4. Mainster MA, Turner PL: Retinal injuries Millen JE: Photochemical lesions in the bright light. Ophthalmology 1983;
from light: mechanisms, hazards and primate retina under conditions of elevated 90:937–944.
prevention. In: Ryan SJ, Hinton DR, blood oxygen. Invest Ophthalmol Vis Sci 36. Stamper DA, Lund DJ, Molchany JW,
Schachat AP, Wilkinson P, eds. Retina. 4th 1984; 25:893–898. Stuck BE: Human pupil and eyelid
edn. London: Elsevier; 2006:1857–1870. 20. Noell WK, Walker VS, Kang BS, Berman S: response to intense laser light: implications
5. Noell WK: Possible mechanisms of Retinal damage by light in rats. Invest for protection. Percept Mot Skills 2002;
photoreceptor damage by light in Ophthalmol 1966; 5:450–473. 95:775–782.
mammalian eyes. Vision Res 1980; 21. Rinkoff J, Machemer R, Hida T, Chandler D: 37. Snodderly DM, Auran JD, Delori FC: The
20:1163–1171. Temperature-dependent light damage to the macular pigment. II. Spatial distribution in
6. Kuwabara T, Gorn RA: Retinal damage by retina. Am J Ophthalmol 1986; 102:452–462. primate retinas. Invest Ophthalmol Vis Sci
visible light. An electron microscopic study. 22. Boulton M, Docchio F, Dayhaw-Barker P, 1984; 25:674–685.
Arch Ophthalmol 1968; 79:69–78. et al: Age-related changes in the 38. Beatty S, Boulton M, Henson D, et al:
7. Friedman E, Kuwabara T: The retinal morphology, absorption and fluorescence Macular pigment and age related macular
pigment epithelium. IV. The damaging of melanosomes and lipofuscin granules of degeneration. Br J Ophthalmol 1999;
effects of radiant energy. Arch Ophthalmol the retinal pigment epithelium. Vision Res 83:867–877.
1968; 80:265–279. 1990; 30:1291–1303. 39. Kirschfeld K: Carotenoid pigments: their
8. Mainster MA: Destructive light adaptation. 23. Rozanowska M, Jarvis-Evans J, possible role in protecting against
Ann Ophthalmol 1970; 2:44–48. Korytowski W, et al: Blue light-induced photooxidation in eyes and photoreceptor
9. Ham WT Jr, Mueller HA, Sliney DH: Retinal reactivity of retinal age pigment. In vitro cells. Proc R Soc Lond B Biol Sci 1982;
sensitivity to damage from short generation of oxygen-reactive species. 216:71–85.
wavelength light. Nature 1976; J Biol Chem 1995; 270:18825–18830. 40. Khachik F, Bernstein PS, Garland DL:
260:153–155. 24. Boulton M, Rozanowska M, Rozanowski B: Identification of lutein and zeaxanthin
10. Lawwill T: Three major pathologic Retinal photodamage. J Photochem oxidation products in human and monkey
processes caused by light in the primate Photobiol B 2001; 64:144–161. retinas. Invest Ophthalmol Vis Sci 1997;
retina: a search for mechanisms. Trans Am 25. Sliney DH:. How light reaches the eye and 38:1802–1811.
Ophthalmol Soc 1982; 80:517–579. its components. Int J Toxicol 2002; 41. Berendschot TT, van Norren D: Macular
11. Wenzel A, Grimm C, Samardzija M, 21:501–509. pigment shows ringlike structures. Invest
Reme CE: Molecular mechanisms of light- 26. Mellerio J: Light effects on the retina. In: Ophthalmol Vis Sci 2006; 47:709–714.
induced photoreceptor apoptosis and Albert DM, Jakobiec FA, eds. Principles 42. Liew SH, Gilbert CE, Spector TD, et al:
neuroprotection for retinal degeneration. and practice of ophthalmology. Central retinal thickness is positively
Prog Retin Eye Res 2005; 24:275–306. Philadelphia: W. B. Saunders Company; correlated with macular pigment optical
12. Reme CE: The dark side of light: rhodopsin 1994: 1326–1345. density. Exp Eye Res 2006; 82:915–920.
and the silent death of vision the proctor 27. Girotti AW: Photodynamic lipid peroxidation 43. Delori FC, Goger DG, Keilhauer C, et al:
lecture. Invest Ophthalmol Vis Sci 2005; in biological systems. Photochem Bimodal spatial distribution of macular
46:2671–2682. Photobiol 1990; 51:497–509. pigment: evidence of a gender relationship.
13. Pokorny J, Smith VC, Verriest G, Pinckers 28. Glickman RD: Phototoxicity to the retina: J Opt Soc Am A Opt Image Sci Vis 2006;
AJLG: Congenital and acquired color vision mechanisms of damage. Int J Toxicol 2002; 23:521–538.
defects. New York: Grune & Stratton; 1979. 21:473–490. 44. Pease PL, Adams AJ, Nuccio E: Optical
14. Mainster MA: Intraocular lenses should 29. Leist M, Jaattela M: Four deaths and a density of human macular pigment. Vision
block UV radiation and violet but not blue funeral: from caspases to alternative Res 1987; 27:705–710.
light. Arch Ophthalmol 2005; 123:550–555. mechanisms. Nat Rev Mol Cell Biol 2001; 45. Werner JS, Donnelly SK, Kliegl R: Aging
15. Sykes SM, Robison WG Jr, Waxler M, 2:589–598. and human macular pigment density.
Kuwabara T: Damage to the monkey retina 30. Boettner EA, Wolter JR: Transmission of Appended with translations from the work
by broad-spectrum fluorescent light. Invest the ocular media. Invest Ophthalmol 1962; of Max Schultze and Ewald Hering. Vision
Ophthalmol Vis Sci 1981; 20:425–434. 1:776–783. Res 1987; 27:257–268.
16. Mainster MA: Light and macular 31. van Norren D, Vos JJ: Spectral 46. Mainster MA: Henle fibers may direct light
degeneration: a biophysical and clinical transmission of the human ocular media. toward the center of the fovea. Lasers Light
perspective. Eye 1987; 1:304–310. Vision Res 1974; 14:1237–1244. Ophthalmol 1988; 2:79–86. 2201
RETINA AND VITREOUS

47. Handelman GJ, Snodderly DM, Krinsky NI, photoreceptor. J Neurosci 2001; 84. Daly GH, DiLeonardo JM, Balkema NR,
et al: Biological control of primate macular 21:6405–6412. Balkema GW: The relationship between
pigment. Biochemical and densitometric 65. Thapan K, Arendt J, Skene DJ: An action ambient lighting conditions, absolute dark-
studies. Invest Ophthalmol Vis Sci 1991; spectrum for melatonin suppression: adapted thresholds, and rhodopsin in black
32:257–267. evidence for a novel non-rod, non-cone and hypopigmented mice. Vis Neurosci
48. Mainster MA: Violet and blue light blocking photoreceptor system in humans. J Physiol 2004; 21:925–934.
intraocular lenses: photoprotection versus 2001; 535:261–267. 85. Reme CE, Wolfrum U, Imsand C, et al:
photoreception. Br J Ophthalmol 2006; 66. Hankins MW, Lucas RJ: The primary visual Photoreceptor autophagy: effects of light
90:784–792. pathway in humans is regulated according history on number and opsin content of
49. Beatty S, Murray IJ, Henson DB, et al: to long-term light exposure through the degradative vacuoles. Invest Ophthalmol
Macular pigment and risk for age-related action of a nonclassical photopigment. Vis Sci 1999; 40:2398–2404.
macular degeneration in subjects from a Curr Biol 2002; 12:191–198. 86. Surgucheva I, Ninkina N, Buchman VL,
Northern European population. Invest 67. Dacey DM, Liao HW, Peterson BB, et al: et al: Protein aggregation in retinal cells
Ophthalmol Vis Sci 2001; 42:439–446. Melanopsin-expressing ganglion cells in and approaches to cell protection. Cell Mol
50. Delori FC, Goger DG, Hammond BR, et al: primate retina signal colour and irradiance Neurobiol 2005; 25:1051–1066.
Macular pigment density measured by and project to the LGN. Nature 2005; 87. Cicerone CM: Cones survive rods in the
autofluorescence spectrometry: 433:749–754. light-damaged eye of the albino rat.
comparison with reflectometry and 68. Hattar S, Lucas RJ, Mrosovsky N, et al: Science 1976; 194:1183–1185.
heterochromatic flicker photometry. J Opt Melanopsin and rod-cone photoreceptive 88. Marshall J, Mellerio J, Palmer DA: Damage
Soc Am A Opt Image Sci Vis 2001; systems account for all major accessory to pigeon retinae by moderate illumination
18:1212–1230. visual functions in mice. Nature 2003; from fluorescent lamps. Exp Eye Res 1972;
SECTION 10

51. Berendschot TT, van Norren D: On the age 424:76–81. 14:164–169.


dependency of the macular pigment optical 69. Qiu X, Kumbalasiri T, Carlson SM, et al: 89. Hope-Ross MW, Mahon GJ, Gardiner TA,
density. Exp Eye Res 2005; 81:602–609. Induction of photosensitivity by Archer DB: Ultrastructural findings in solar
52. Berendschot TT, Willemse-Assink JJ, heterologous expression of melanopsin. retinopathy. Eye 1993; 7(Pt 1):29–33.
Bastiaanse M, et al: Macular pigment and Nature 2005; 433:745–749. 90. Snodderly DM, Sandstrom MM, Leung IY,
melanin in age-related maculopathy in a 70. Burns ME, Baylor DA: Activation, et al: Retinal pigment epithelial cell
general population. Invest Ophthalmol Vis deactivation, and adaptation in vertebrate distribution in central retina of rhesus
Sci 2002; 43:1928–1932. photoreceptor cells. Annu Rev Neurosci monkeys. Invest Ophthalmol Vis Sci 2002;
53. Wald G: Human vision and the spectrum. 2001; 24:779–805. 43:2815–2818.
Science 1945; 101:653–658. 71. Rozanowska M, Sarna T: Light-induced 91. Sperling HG, Johnson C, Harwerth RS:
54. Griswold MS, Stark WS: Scotopic spectral damage to the retina: role of rhodopsin Differential spectral photic damage to
sensitivity of phakic and aphakic observers chromophore revisited. Photochem primate cones. Vision Res 1980;
extending into the near ultraviolet. Vision Photobiol 2005; 81:1305–1330. 20:1117–1125.
Res 1992; 32:1739–1743. 72. Wenzel A, Oberhauser V, Pugh EN Jr, et al: 92. Provencio I, Jiang G, De Grip WJ, et al:
55. Ham WT Jr, Ruffolo JJ Jr, Mueller HA, The retinal G protein-coupled receptor Melanopsin: an opsin in melanophores,
Guerry D 3rd: The nature of retinal radiation (RGR) enhances isomerohydrolase activity brain, and eye. Proc Natl Acad Sci USA
damage: dependence on wavelength, independent of light. J Biol Chem 2005; 1998; 95:340–345.
power level and exposure time. Vision Res 280:29874–29884. 93. Van Gelder RN: Non-visual ocular
1980; 20:1105–1111. 73. Arshavsky VY, Lamb TD, Pugh EN Jr: G photoreception. Ophthalmic Genet 2001;
56. Reme CE, Grimm C, Hafezi F, et al: Why proteins and phototransduction. Annu Rev 22:195–205.
study rod cell death in retinal Physiol 2002; 64:153–187. 94. Berson DM, Dunn FA, Takao M:
degenerations and how? Doc Ophthalmol 74. Saari JC: Biochemistry of visual pigment Phototransduction by retinal ganglion cells
2003; 106:25–29. regeneration: the Friedenwald lecture. Invest that set the circadian clock. Science 2002;
57. Margrain TH, Boulton M, Marshall J, Ophthalmol Vis Sci 2000; 41:337–348. 295:1070–1073.
Sliney DH: Do blue light filters confer 75. Lamb TD, Pugh EN Jr: Dark adaptation and 95. Menaker M: Circadian rhythms. Circadian
protection against age-related macular the retinoid cycle of vision. Prog Retin Eye photoreception. Science 2003; 299:213–214.
degeneration? Prog Retin Eye Res 2004; Res 2004; 23:307–380. 96. Feeney L, Berman ER: Oxygen toxicity:
23:523–531. 76. Rapp LM: Retinal phototoxicity. In: membrane damage by free radicals. Invest
58. Ham WT Jr, Ruffolo JJ Jr, Mueller HA, et al: Chang LW, Dyer RS, eds. Handbook of Ophthalmol 1976; 15:789–792.
Histologic analysis of photochemical neurotoxicology. New York: Dekker; 1995. 97. Ham WT Jr, Mueller HA, Ruffolo JJ Jr, et al:
lesions produced in rhesus retina by short- 77. Reme C, Reinboth J, Clausen M, Hafezi F: Basic mechanisms underlying the
wave-length light. Invest Ophthalmol Vis Light damage revisited: converging production of photochemical lesions in the
Sci 1978; 17:1029–1035. evidence, diverging views? Graefes Arch mammalian retina. Curr Eye Res 1984;
59. Kremers JJ, van Norren D: Two classes of Clin Exp Ophthalmol 1996; 234:2–11. 3:165–174.
photochemical damage of the retina. 78. Hao W, Wenzel A, Obin MS, et al: Evidence 98. Snodderly DM: Evidence for protection
Lasers Light Ophthalmol 1988; 2:41–52. for two apoptotic pathways in light-induced against age-related macular degeneration
60. ACGIH Gorgels TG, Van Norren D: Two retinal degeneration. Nat Genet 2002; by carotenoids and antioxidant vitamins.
spectral types of retinal light damage occur 32:254–260. Am J Clin Nutr 1995; 62:1448S-1461S.
in albino as well as in pigmented rat: no 79. Jacobson SG, McInnes RR: Blinded by the 99. Pautler EL, Morita M, Beezley D:
essential role for melanin. Exp Eye Res light. Nat Genet 2002; 32:215–216. Hemoprotein(s) mediate blue light damage
1998; 66:155–162. 80. Vaughan DK, Nemke JL, Fliesler SJ, et al: in the retinal pigment epithelium.
61. ACGIH Threshold limit values for chemical Evidence for a circadian rhythm of Photochem Photobiol 1990; 51:599–605.
substances physical agents: biological susceptibility to retinal light damage. 100. Gorgels TG, van Norren D: Ultraviolet and
exposure indices. Cincinnati: American Photochem Photobiol 2002; 75:547–553. green light cause different types of damage
Conference of Governmental Industrial 81. Penn JS, Williams TP: Photostasis: regulation in rat retina. Invest Ophthalmol Vis Sci
Hygienists; 1997. of daily photon-catch by rat retinas in 1995; 36:851–863.
62. Merbs SL, Nathans J: Absorption spectra response to various cyclic illuminances. 101. Grimm C, Wenzel A, Williams T, et al:
of human cone pigments. Nature 1992; Exp Eye Res 1986; 43:915–928. Rhodopsin-mediated blue-light damage to
356:433–435. 82. Williams TP, Squitieri A, Henderson RP, the rat retina: effect of photoreversal of
63. Stockman A, Sharpe LT, Merbs S, Nathans Webbers JP: Reciprocity between light bleaching. Invest Ophthalmol Vis Sci 2001;
J: Spectral sensitivities of human cone intensity and rhodopsin concentration 42:497–505.
visual pigments determined in vivo and in across the rat retina. J Physiol 102. Feeney-Burns L, Hilderbrand ES, Eldridge S:
vitro. Methods Enzymol 2000; 1999; 516(Pt 3):869–874. Aging human RPE: morphometric analysis
316:626–650. 83. Williams TP, Webbers JP, Giordano L, of macular, equatorial, and peripheral cells.
64. Brainard GC, Hanifin JP, Greeson JM, et al: Henderson RP: Distribution of photon Invest Ophthalmol Vis Sci 1984; 25:195–200.
Action spectrum for melatonin regulation in absorption rates across the rat retina. 103. Marshall J: The ageing retina: physiology or
2202 humans: evidence for a novel circadian J Physiol 1998; 508(Pt 2):515–522. pathology. Eye 1987; 1(Pt 2):282–295.
Photic Retinopathy

104. Sparrow JR, Nakanishi K, Parish CA: The 126. Kaushik S, Gupta V, Gupta A: Optical 148. Khwarg SG, Linstone FA, Daniels SA, et al:
lipofuscin fluorophore A2E mediates blue coherence tomography findings in solar Incidence, risk factors, and morphology in
light-induced damage to retinal pigmented retinopathy. Ophthalmic Surg Lasers operating microscope light retinopathy. Am
epithelial cells. Invest Ophthalmol Vis Sci Imaging 2004; 35:52–55. J Ophthalmol 1987; 103:255–263.
2000; 41:1981–1989. 127. Garg SJ, Martidis A, Nelson ML, 149. Brod RD, Olsen KR, Ball SF, Packer AJ:
105. Sparrow JR, Parish CA, Hashimoto M, Sivalingam A: Optical coherence The site of operating microscope light-
Nakanishi K: A2E, a lipofuscin fluorophore, tomography of chronic solar retinopathy. induced injury on the human retina. Am J
in human retinal pigmented epithelial cells Am J Ophthalmol 2004; 137:351–354. Ophthalmol 1989; 107:390–397.
in culture. Invest Ophthalmol Vis Sci 1999; 128. Jorge R, Costa RA, Quirino LS, et al: 150. Pavilack MA, Brod RD: Site of potential
40:2988–2995. Optical coherence tomography findings in operating microscope light-induced
106. Lamb LE, Simon JD: A2E: a component of patients with late solar retinopathy. Am J phototoxicity on the human retina during
ocular lipofuscin. Photochem Photobiol Ophthalmol 2004; 137:1139–1143. temporal approach eye surgery.
2004; 79:127–136. 129. Grey RH: Foveo-macular retinitis, solar Ophthalmology 2001; 108:381–385.
107. Mainster MA: Retinol transport and retinopathy, and trauma. Br J Ophthalmol 151. Boldrey EE, Ho BT, Griffith RD: Retinal
regeneration of human cone photopigment. 1978; 62:543–546. burns occurring at cataract extraction.
Nat New Biol 1972; 238:223–224. 130. Kelley JS, Hoover RE, George T: Whiplash Ophthalmology 1984; 91:1297–1302.
108. Godley BF, Shamsi FA, Liang FQ, et al: maculopathy. Arch Ophthalmol 1978; 152. Brod RD, Barron BA, Suelflow JA:
Blue light induces mitochondrial DNA 96:834–835. Phototoxic retinal damage during refractive
damage and free radical production in 131. Abebe MT, De Laey JJ: Foveomacular surgery. Am J Ophthalmol 1986; 102:121–123.
epithelial cells. J Biol Chem 2005; retinitis as a result of ocular contusion. Bull 153. Khwarg SG, Geoghegan M, Hanscom TA:

CHAPTER 174
280:21061–21066. Soc Belge Ophtalmol 1992; 243:171–175. Light-induced maculopathy from the
109. Lawwill T: Effects of prolonged exposure of 132. Kuming BS: Foveomacular retinitis. Br J operating microscope. Am J Ophthalmol
rabbit retina to low-intensity light. Invest Ophthalmol 1986; 70:816–818. 1984; 98:628–630.
Ophthalmol 1973; 12:45–51. 133. Jacobs NA: Foveomacular retinitis. Br J 154. Robertson DM, Feldman RB: Photic
110. Hoppeler T, Hendrickson P, Dietrich C, Ophthalmol 1987; 71:563. retinopathy from the operating room
Reme C: Morphology and time-course of 134. Cordes FC: A type of foveomacular retinitis microscope. Am J Ophthalmol 1986;
defined photochemical lesions in the rabbit observed in the US Navy. Am J Ophthalmol 101:561–569.
retina. Curr Eye Res 1988; 7:849–860. 1944; 27:803–816. 155. Mares-Perlman JA, Brady WE, Klein BE,
111. Rozanowska M, Korytowski W, 135. Ritchey CL, Ewald RA: Sun gazing as the et al: Diet and nuclear lens opacities. Am J
Rozanowski B, et al: Photoreactivity of cause of foveomacular retinitis. Am J Epidemiol 1995; 141:322–334.
aged human RPE melanosomes: a Ophthalmol 1970; 70:491–497. 156. Fuller D, Machemer R, Knighton RW:
comparison with lipofuscin. Invest 136. Kerr LM, Little HL: Foveomacular retinitis. Retinal damage produced by intraocular
Ophthalmol Vis Sci 2002; 43:2088–2096. Arch Ophthalmol 1966; 76:498–504. fiber optic light. Am J Ophthalmol 1978;
112. Tso MO, La Piana FG: The human fovea 137. Marlor RL, Blais BR, Preston FR, 85:519–537.
after sungazing. Trans Am Acad Ophthalmol Boyden DG: Foveomacular retinitis, an 157. Michels M, Lewis H, Abrams GW, et al:
Otolaryngol 1975; 79:OP788–OP795. important problem in military medicine: Macular phototoxicity caused by fiberoptic
113. Green WR, Robertson DM: Pathologic epidemiology. Invest Ophthalmol 1973; endoillumination during pars plana
findings of photic retinopathy in the human 12:5–16. vitrectomy. Am J Ophthalmol 1992;
eye. Am J Ophthalmol 1991; 112:520–527. 138. Gladstone GJ, Tasman W: Solar retinitis 114:287–296.
114. White TJ, Mainster MA, Wilson PW, Tips JH: after minimal exposure. Arch Ophthalmol 158. Kleinmann G, Hoffman P, Schechtman E,
Chorioretinal temperature increases from 1978; 96:1368–1369. Pollack A: Microscope-induced retinal
solar observation. Bull Math Biophys 1971; 139. Yannuzzi LA, Fisher YL, Krueger A, Slakter J: phototoxicity in cataract surgery of short
33:1–17. Solar retinopathy: a photobiological and duration. Ophthalmology 2002;
115. Naidoff MA, Slinkey DH: Retinal injury from geophysical analysis. Trans Am Ophthalmol 109:334–338.
a welding arc. Am J Ophthalmol 1974; Soc 1987; 85:120–158. 159. Postel EA, Pulido JS, Byrnes GA, et al:
77:663–668. 140. Terrien F: De trouble visuel provoqué par Long-term follow-up of iatrogenic
116. Sliney DH, Armstrong BC: Radiometric l’électricité. Arch Ophthalmol 1902; phototoxicity. Arch Ophthalmol 1998;
analysis of surgical microscope lights for 22:692–696. 116:753–757.
hazards analyses. Appl Opt 1986; 141. Uniat L, Olk RJ, Hanish SJ: Welding arc 160. Irvine AR, Wood I, Morris BW: Retinal
25:1882–1889. maculopathy. Am J Ophthalmol 1986; damage from the illumination of the
117. Mainster MA: Solar eclipse safety. 102:394–395. operating microscope. An experimental
Ophthalmology 1998; 105:9–10. 142. Fich M, Dahl H, Fledelius H, Tinning S: study in pseudophakic monkeys. Arch
118. Mainster MA, White TJ, Tips JH, Wilson PW: Maculopathy caused by welding arcs. Ophthalmol 1984; 102:1358–1365.
Retinal-temperature increases produced by A report of 3 cases. Acta Ophthalmol 161. Jaffe GJ, Wood IS: Retinal phototoxicity
intense light sources. J Opt Soc Am 1970; (Copenh) 1993; 71:402–404. from the operating microscope: a
60:264–270. 143. Lucas RS, Harper CA, McCombe MF, protective effect by the fovea (letter). Arch
119. Dhir SP, Gupta A, Jain IS: Eclipse retinopathy. et al: Optical coherence tomography Ophthalmol 1988; 106:445–446.
Br J Ophthalmol 1981; 65:42–45. findings in Welder’s maculopathy. Retina 162. Parver LM, Auker CR, Fine BS:
120. Jacobs NA, Headon M, Rosen ES: Solar 2007; 27 (in press). Observations on monkey eyes exposed to
retinopathy in the Manchester area. Trans 144. Sliney DH: Defining biologic exposures to light from an operating microscope.
Ophthalmol Soc UK 1985; 104(Pt light. In: Cronly-Dillon J, Rosen ES, Ophthalmology 1983; 90:964–972.
6):625–628. Marshall J, eds. Hazards of light. New York: 163. Jaffe GJ, Irvine AR, Wood IS, et al: Retinal
121. Gass JDM: Stereoscopic atlas of macular Pergamon; 1986. phototoxicity from the operating
diseases. 3rd edn. St Louis: Mosby-Year 145. Gardner TW, Ai E, Chrobak M, Shoch DE. microscope. The role of inspired oxygen.
Book; 1987. Photic maculopathy secondary to Ophthalmology 1988; 95:1130–1141.
122. MacFaul PA: Visual prognosis after solar short-circuiting of a high-tension electric 164. Mauget-Faysse M, Quaranta M, Francoz N,
retinopathy. Br J Ophthalmol 1969; current. Ophthalmology 1982; et al: Incidental retinal phototoxicity
53:534–541. 89:865–868. associated with ingestion of
123. Schatz H, Mendelblatt F: Solar retinopathy 146. Fishman GA: Light-induced maculopathy photosensitizing drugs. Graefes Arch Clin
from sun-gazing under the influence of from surgical microscopes during cataract Exp Ophthalmol 2001; 239:501–508.
LSD. Br J Ophthalmol 1973; 57:270–273. surgery. In: Ernest JT, ed. The 1985 year 165. Manzouri B, Egan CA, Hykin PG:
124. Steinkamp PN, Watzke RC, Solomon JD: book of ophthalmology. St Louis: Mosby- Phototoxic maculopathy following
An unusual case of solar retinopathy. Arch Year Book; 1985:177–180. uneventful cataract surgery in a
Ophthalmol 2003; 121:1798–1799. 147. Michels M, Sternberg P, Jr: Operating predisposed patient. Br J Ophthalmol
125. Aiello LP, Arrigg PG, Shah ST, et al: Solar microscope-induced retinal phototoxicity: 2002; 86:705–706.
retinopathy associated with hypoglycemic pathophysiology, clinical manifestations 166. Ferguson J: Photosensitivity due to drugs.
insulin reaction. Arch Ophthalmol 1994; and prevention. Surv Ophthalmol 1990; Photodermatol Photoimmunol Photomed
112:982–983. 34:237–252. 2002; 18:262–269. 2203
RETINA AND VITREOUS

167. Li S, Lam TT, Fu J, Tso MO: Systemic degeneration following bilateral cataract 202. Curcio CA, Millican CL, Allen KA, Kalina
hypertension exaggerates retinal photic surgery. Ophthalmic Surg Lasers 1998; RE: Aging of the human photoreceptor
injury. Arch Ophthalmol 1995; 113:521–526. 29:286–294. mosaic: evidence for selective vulnerability
168. Mainster MA, Ham WT Jr, Delori FC: 187. Shuttleworth GN, Luhishi EA, Harrad RA: of rods in central retina. Invest Ophthalmol
Potential retinal hazards. Instrument and Do patients with age related maculopathy Vis Sci 1993; 34:3278–3296.
environmental light sources. and cataract benefit from cataract surgery? 203. Panda-Jonas S, Jonas JB, Jakobczyk-
Ophthalmology 1983; 90:927–932. Br J Ophthalmol 1998; 82:611–616. Zmija M: Retinal photoreceptor density
169. McIntyre DJ: Phototoxicity. The eclipse 188. Wong TY: Cataract surgery in patients with decreases with age. Ophthalmology 1995;
filter. Ophthalmology 1985; 92:364–365. cataract and age related macular 102:1853–1859.
170. Landry RJ, Miller SA, Byrnes GA: Study of degeneration: do the benefits outweigh the 204. Curcio CA: Photoreceptor topography in
filtered light on potential retinal photic risks? Br J Ophthalmol 2000; ageing and age-related maculopathy. Eye
hazards with operation microscopes used 84:1337–1338. 2001; 15:376–383.
for ocular surgery. Appl Opt 2002; 189. Klein R, Klein BE, Wong TY, et al: The 205. Del Priore LV, Kuo YH, Tezel TH: Age-
41:802–804. association of cataract and cataract related changes in human RPE cell density
171. O’Brien DP, Francis IC: The corneal quilt: surgery with the long-term incidence of and apoptosis proportion in situ. Invest
a protective device designed to reduce age-related maculopathy: the Beaver Dam Ophthalmol Vis Sci 2002; 43:3312–3318.
intraoperative retinal phototoxicity. eye study. Arch Ophthalmol 2002; 206. Spraul CW, Lang GE, Grossniklaus HE,
Ophthalmic Surg 1994; 25:191–194. 120:1551–1558. Lang GK: Histologic and morphometric
172. Kraff MC, Lieberman HL, Jampol LM, 190. Armbrecht AM, Findlay C, Aspinall PA, et al: analysis of the choroid, Bruch’s membrane,
Sanders DR: Effect of a pupillary light Cataract surgery in patients with age- and retinal pigment epithelium in
occluder on cystoid macular edema. related macular degeneration: one-year postmortem eyes with age-related macular
SECTION 10

J Cataract Refract Surg 1989; 15:658–660. outcomes. J Cataract Refract Surg 2003; degeneration and histologic examination of
173. Nevyas HJ, Nevyas JY: Surgical corneal 29:686–693. surgically excised choroidal neovascular
light occluder made of black HEMA. 191. Wang JJ, Klein R, Smith W, et al: Cataract membranes. Surv Ophthalmol 1999;
Ophthalmic Surg 1985; 16:696–698. surgery and the 5-year incidence of late- 44(Suppl 1):S10–S32.
174. Jampol LM, Kraff MC, Sanders DR, et al: stage age-related maculopathy: pooled 207. Zarbin MA: Current concepts in the
Near-UV radiation from the operating findings from the Beaver Dam and Blue pathogenesis of age-related macular
microscope and pseudophakic cystoid Mountains eye studies. Ophthalmology degeneration. Arch Ophthalmol 2004;
macular edema. Arch Ophthalmol 1985; 2003; 110:1960–1967. 122:598–614.
103:28–30. 192. Freeman EE, Munoz B, West SK, et al: 208. McCarty CA, Mukesh BN, Fu CL, et al: Risk
175. Keates RH, Genstler DE: UV radiation. Is there an association between cataract factors for age-related maculopathy: the
Ophthalmic Surg 1982; 13:327. surgery and age-related macular Visual Impairment Project. Arch Ophthalmol
176. Robertson DM, McLaren JW: Photic degeneration? Data from three population- 2001; 119:1455–1462.
retinopathy from the operating room based studies. Am J Ophthalmol 2003; 209. Seddon JM, Ajani UA, Sperduto RD, et al:
microscope. Study with filters. Arch 135:849–856. Dietary carotenoids, vitamins A, C, and E,
Ophthalmol 1989; 107:373–375. 193. Ferris FL 3rd: Discussion of a model of and advanced age-related macular
177. Keates RH, Armstrong PF: Use of a short spectral filtering to reduce photochemical degeneration. Eye Disease Case-Control
wavelength filter in an operating damage in age-related macular Study Group. Jama 1994; 272:1413–1420.
microscope. Ophthalmic Surg 1985; degeneration. Trans Am Ophthalmol Soc 210. Tso MO: Pathogenetic factors of aging
16:40–41. 2004; 102:95. macular degeneration. Ophthalmology
178. Delori FC, Gragoudas ES, Francisco R, 194. Martin DF, Gensler G, Klein BEK, et al: The 1985; 92:628–635.
Pruett RC: Monochromatic effect of cataract surgery on progression to 211. van Leeuwen R, Ikram MK, Vingerling JR,
ophthalmoscopy and fundus photography. advanced AMD (abstract number 1907). et al: Blood pressure, atherosclerosis, and
The normal fundus. Arch Ophthalmol 1977; ARVO annual meeting. Ophthalmology the incidence of age-related maculopathy:
95:861–868. AfRiVa, translator. FL: Fort Lauderdale; the Rotterdam Study. Invest Ophthalmol Vis
179. Ducrey NM, Delori FC, Gragoudas ES: 2002:76. Sci 2003; 44:3771–3777.
Monochromatic ophthalmoscopy and 195. Dorey CK, Wu G, Ebenstein D, et al:. Cell 212. van der Hoeve J: Eye lesions produced by
fundus photography. II. The pathological loss in the aging retina. Relationship to light rich in ultraviolet rays: senile cataract,
fundus. Arch Ophthalmol 1979; lipofuscin accumulation and macular senile degeneration of the macula. Am J
97:288–293. degeneration. Invest Ophthalmol Vis Sci Ophthalmol 1920; 3:178–194.
180. James RH, Bostrom RG, Remark D, 1989; 30:1691–1699. 213. Ts’o MO, La Piana FG, Appleton B:
Sliney DH: Handheld ophthalmoscopes for 196. Delori FC, Goger DG, Dorey CK: Age- The human fovea after sungazing. Trans
hazard analysis: an analysis. Appl Opt related accumulation and spatial Am Acad Ophthalmol Otolaryngol 1974;
1988; 27:5072–5076. distribution of lipofuscin in RPE of normal 78:OP-677.
181. Michels M, Dawson WW, Feldman RB, subjects. Invest Ophthalmol Vis Sci 2001; 214. Mainster MA: Solar retinitis, photic
Jarolem K: Infrared. An unseen and 42:1855–1866. maculopathy and the pseudophakic
unnecessary hazard in ophthalmic devices. 197. Sparrow JR, Boulton M: RPE lipofuscin and eye. J Am Intraocul Implant Soc 1978;
Ophthalmology 1987; 94:143–148. its role in retinal pathobiology. Exp Eye Res 4:84–86.
182. Oliver M: Posterior pole changes after 2005; 80:595–606. 215. Young RW: A theory of central retinal
cataract extraction in elderly subjects. Am 198. Winkler BS, Boulton ME, Gottsch JD, disease. In: Sears ML, ed. New directions
J Ophthalmol 1966; 62:1145–1148. Sternberg P: Oxidative damage and age- in ophthalmic research. New Haven, CT:
183. Blair CJ, Ferguson J Jr: Exacerbation of related macular degeneration. Mol Vis Yale University Press; 1981:237–270.
senile macular degeneration following 1999; 5:32. 216. Burkle A: Mechanisms of ageing. Eye 2001;
cataract extraction. Am J Ophthalmol 1979; 199. Beatty S, Koh H, Phil M, et al: The role of 15:371–375.
87:77–83. oxidative stress in the pathogenesis of age- 217. van Norren D, Vos H: Sunlight and age-
184. van der Schaft TL, Mooy CM, de Bruijn WC, related macular degeneration. Surv related macular degeneration. Arch
et al: Increased prevalence of disciform Ophthalmol 2000; 45:115–134. Ophthalmol 1990; 108:1670–1671.
macular degeneration after cataract 200. Hillenkamp J, Hussain AA, Jackson TL, 218. Friedman E: A hemodynamic model of the
extraction with implantation of an et al: The influence of path length and pathogenesis of age-related macular
intraocular lens. Br J Ophthalmol 1994; matrix components on ageing degeneration. Am J Ophthalmol 1997;
78:441–445. characteristics of transport between the 124:677–682.
185. Pollack A, Marcovich A, Bukelman A, choroid and the outer retina. Invest 219. Fan BJ, Tam PO, Choy KW, et al: Molecular
Oliver M: Age-related macular degeneration Ophthalmol Vis Sci 2004; 45:1493–1498. diagnostics of genetic eye diseases. Clin
after extracapsular cataract extraction with 201. Ramrattan RS, van der Schaft TL, Mooy CM, Biochem 2006; 39:231–239.
intraocular lens implantation. et al: Morphometric analysis of Bruch’s 220. Curcio CA, Owsley C, Jackson GR: Spare
Ophthalmology 1996; 103:1546–1554. membrane, the choriocapillaris, and the the rods, save the cones in aging and age-
186. Pollack A, Bukelman A, Zalish M, et al: choroid in aging. Invest Ophthalmol Vis Sci related maculopathy. Invest Ophthalmol Vis
2204 The course of age-related macular 1994; 35:2857–2864. Sci 2000; 41:2015–2018.
Photic Retinopathy

221. Pflibsen KP, Pomerantzeff O, Ross RN: 231. Delcourt C, Carriere I, Ponton-Sanchez A, 240. Singh AD, Rennie IG, Seregard S, et al:
Retinal illuminance using a wide-angle et al: Light exposure and the risk of age- Sunlight exposure and pathogenesis of
model of the eye. J Opt Soc Am A 1988; related macular degeneration: the uveal melanoma. Surv Ophthalmol 2004;
5:146–150. Pathologies Oculaires Liées à l’Age (POLA) 49:419–428.
222. Sliney DH: Geometrical gradients in the study. Arch Ophthalmol 2001; 241. Li W, Judge H, Gragoudas ES, et al:
distribution of temperature and absorbed 119:1463–1468. Patterns of tumor initiation in choroidal
ultraviolet radiation in ocular tissues. Dev 232. Clemons TE, Milton RC, Klein R, et al: melanoma. Cancer Res 2000;
Ophthalmol 2002; 35:40–59. Risk factors for the incidence of Advanced 60:3757–3760.
223. Jackson GR, Owsley C, Curcio CA: Age-Related Macular Degeneration in the 242. Vajdic CM, Kricker A, Giblin M, et al: Sun
Photoreceptor degeneration and Age-Related Eye Disease Study (AREDS) exposure predicts risk of ocular melanoma
dysfunction in aging and age-related AREDS report no. 19. Ophthalmology 2005; in Australia. Int J Cancer 2002;
maculopathy. Ageing Res Rev 2002; 112:533–539. 101:175–182.
1:381–396. 233. Risk factors for neovascular age-related 243. Rosenthal FS, Phoon C, Bakalian AE,
224. Feeney-Burns L, Berman ER, Rothman H: macular degeneration. The eye disease Taylor HR: The ocular dose of ultraviolet
Lipofuscin of human retinal pigment case – Control Study Group. Arch radiation to outdoor workers. Invest
epithelium. Am J Ophthalmol 1980; Ophthalmol 1992; 110:1701–1708. Ophthalmol Vis Sci 1988; 29:649–656.
90:783–791. 234. Darzins P, Mitchell P, Heller RF: Sun 244. Gilchrest BA, Eller MS, Geller AC, Yaar M:
225. Weiter JJ, Delori FC, Wing GL, Fitch KA: exposure and age-related macular The pathogenesis of melanoma induced by
Retinal pigment epithelial lipofuscin and degeneration. An Australian case-control ultraviolet radiation. N Engl J Med 1999;
melanin and choroidal melanin in human study. Ophthalmology 1997; 104:770–776. 340:1341–1348.

CHAPTER 174
eyes. Invest Ophthalmol Vis Sci 1986; 235. Khan JC, Shahid H, Thurlby DA, et al: Age 245. ACGIH: Threshold limit values and
27:145–152. related macular degeneration and sun biological eposure indices. Cincinnati, OH:
226. Marshall J: Ageing changes in human cones. exposure, iris colour, and skin sensitivity to American Conference of Governmental
Acta XXIII Concilium Ophthalmologicum sunlight. Br J Ophthalmol 2006; 90:29–32. Industrial Hygienists; 2000.
(Kyoto) 1978; 1:375–378. 236. Sliney DH: Photoprotection of the eye – UV 246. Anderson HL: Building molecular wires
227. Borges J, Li ZY, Tso MO: Effects of repeated radiation and sunglasses. J Photochem from the colours of life: conjugated
photic exposures on the monkey macula. Photobiol B 2001; 64:166–175. porphyrin oligomers. Chem Commun 1999;
Arch Ophthalmol 1990; 108:727–733. 237. Midelfart A: Ultraviolet radiation and 9:2323–2330.
228. Taylor HR, West S, Munoz B, et al: The cataract. Acta Ophthalmol Scand 2005; 247. Kaderbhai MA, Hopper DJ, Akhtar KM,
long-term effects of visible light on the eye. 83:642–644. et al: A cytochrome c from a lupanine-
Arch Ophthalmol 1992; 110:99–104. 238. McCarty CA, Taylor HR: A review of the transforming Pseudomonas putida strain is
229. Tomany SC, Cruickshanks KJ, Klein R, epidemiologic evidence linking ultraviolet expressed in Escherichia coli during
et al: Sunlight and the 10-year incidence of radiation and cataracts. Dev Ophthalmol aerobic cultivation and efficiently exported
age-related maculopathy: the Beaver Dam 2002; 35:21–31. and assembled in the periplasm. Appl
Eye Study. Arch Ophthalmol 2004; 239. Delcourt C, Carriere I, Ponton-Sanchez A, Environ Microbiol 2003; 69:4727–4731.
122:750–757. et al: Light exposure and the risk of 248. Wyszecki G, Stiles WS: Color Science. 2nd
230. Hirvela H, Luukinen H, Laara E, et al: Risk cortical, nuclear, and posterior subcapsular ed. New York: Wiley; 1982.
factors of age-related maculopathy in a cataracts: the Pathologies Oculaires Liees 249. Wald G, Brown PK: Human rhodopsin.
population 70 years of age or older. a l’Age (POLA) study. Arch Ophthalmol Science 1958; 127:222–226.
Ophthalmology 1996; 103:871–877. 2000; 118:385–392.

2205
CHAPTER

175 Radiation Retinopathy


Sayjal J. Patel and Andrew P. Schachat

Key Features number and size, as well as location most likely have a large
• Retinopathy is typically delayed. If it occurs, the onset is
influence. Normal eyes indirectly or directly exposed to radia-
typically in the first 1–3 years after treatment
tion for nonocular tumors may manifest different effects of radia-
• Retinal manifestations of radiation vary with dose of radiation
tion compared to eyes with tumors being treated directly. Local
used, fraction number, size of tumor, location of tumor, and the
damage due to the effects of the tumor may be difficult to differen-
use of external beam versus plaque therapy
tiate from radiation damage alone. Chemotherapy used concur-
• Early histological findings are usually focal loss of capillary
rently with radiation or systemic diseases such as diabetes can
endothelial cells and pericytes with subsequent areas of retinal
further confound the interpretation of retinopathy seen after
capillary nonperfusion that can lead to neovascularization
radiation.
Studies using angiography and histopathology have been pivotal
in an attempt to understand the effects of radiation on the retina.
INTRODUCTION Most of these studies have concentrated on findings after external
beam radiation, presumably since external beam radiation is
Radiation has been used therapeutically for the treatment of most common. The doses used in these studies vary and there-
neoplastic lesions for over 100 years. According to del Regato, in fore the dose–effect relationship findings are difficult to
1896, 3 weeks after reading about Roentgen's discovery of ‘X-rays’, compare and interpret.
Grubbe was the first to use radiation to treat breast cancer.1 In the In 1958, histologic studies of primate eyes previously exposed
1930s, Moore and Stallard began using radiation for the treat- to external beam radiation by Cibis et al showed the acute
ment of ocular tumors.2,3 Ocular complications following radia- sequelae of radiation therapy primarily involved the outer reti-
tion were described by Stallard in 1933 subsequent to treatment for nal layer and the photoreceptors, specifically the rods, and the
retinoblastoma and retinal capillary hemangioma.3 He described delayed sequelae involved the inner retina and retinal vascula-
retinal hemorrhages, exudative changes in a circinate pattern, ture.4 The acute changes were noted as early as 19 min following
optic disk edema and optic atrophy occurring over a period of radiation exposure. The dose used in this study is much higher
~20 months postradiation. Since these early reports, many than the typical doses used today.
reports of retinopathy following radiation to various tumors of In 1970, Hayreh was the first to describe the fluorescein charac-
the head and neck have been described.4–16 teristics of radiation retinopathy followed over time in three
The onset of radiation retinopathy is usually delayed and patients with choroidal melanoma.15 All three patients had been
generally does not appear for months or years after initial expo- treated with plaque radiation at a dose of 80 Gy to the tumor
sure to radiation. Characteristics of radiation retinopathy include apex. Over a 22 month to 4 year follow-up, he noted a variety
retinal microaneurysms and intraretinal hemorrhages, telangiec- of changes in the inner and outer retina including obliteration
tasia, perivascular sheathing, cotton wool spots, exudate, macular of retinal capillary vessels and the choriocapillaris as well as
edema, retinal pigment epithelial atrophy, optic disk edema and areas of retinal ischemia.
atrophy. The appearance can mimic diabetic retinopathy and there In 1980, Egbert et al described the histological changes seen
is a similar underlying cause – microvascular leakage and occlu- in 17 human eyes with retinoblastoma after receiving 35–60 Gy
sion. Areas of ischemia can lead to retinal, optic disk or iris of external beam radiation.17 They described the arteriolar and
neovascularization which can further develop into vitreous hemor- capillary walls as thickened due to a fine fibrillary material with
rhage, retinal detachment or glaucoma. The primary cause of fibrin deposition within the walls and outside the vessel walls.
vision loss is usually due to exudative or ischemic maculopathy They postulated that the occlusion of the vessels could be due to
or optic neuropathy. The severity, although multifactorial, is largely narrowing of the vessel lumen. Brown et al, in 1982, found areas
dependent on dose, frequency, location as well as total area treated. of retinal capillary obliteration in all eyes studied with fluorescein
Patients with diabetes or patients undergoing chemotherapy are angiography after cobalt-60 plaque radiation and external beam
at increased risk for developing radiation retinopathy.11,12 radiation.12 They suggested the diagnosis of radiation retino-
pathy should be questioned if retinal capillary nonperfusion is
PATHOPHYSIOLOGY not demonstrated.
In 1987, Irvine et al reported the clinical and histological
Ionizing radiation can be used therapeutically in two general changes seen in 11 primates after radiation levels of up to 35 Gy.18
forms: external beam radiation (teletherapy, or radiation from a Early findings were focal loss of capillary endothelial cells and
distance) and plaque radiation (brachytherapy, or radiation from pericytes. As more capillaries became incompetent, areas of retinal
a nearby source). Many variables may have an effect on the capillary nonperfusion followed. Nonperfusion led to intra-
ocular manifestations of radiation therapy. Total dose, fraction retinal neovascularization and eventual neovascular glaucoma. 2207
RETINA AND VITREOUS

Elevated levels of a vasoproliferative factor were found in an eye infarctions, and focal retinal pigment epithelial changes.9–12,14 If
with rubeosis while lower levels were found in an eye without any of these findings are within 3 mm of the foveola, the term
rubeosis. Interestingly, intravitreal proliferative retinopathy was radiation maculopathy is used.9 In patients with choroidal
not noted. melanoma, local tumor effects may resemble changes similiar
In 1991, Archer et al described their observations after to radiation related retinopathy such as macular edema and intra-
enucleating seven human eyes that had received external beam retinal hemorrhages. The similarity in findings may make it
radiation for various head and neck tumors with radiation levels difficult to distinguish between these two possible causes.
up to 55 Gy.13 With the help of trypsin digest and electron Just as the manifestations of radiation vary with dose, fraction
microscopy, a preferential loss of vascular endothelial cells and number, size, location, and external beam versus plaque therapy, so
preservation of pericytes was demonstrated. Early structural does the incidence. In one study of 96 patients with large uveal
changes in capillaries such as outpouchings, fusiform dilations, melanoma who underwent plaque radiation, the 5-year incidence
and microaneurysms were described. With further damage, of maculopathy was 52%.5 Gunduz et al studied 1300 patients
fenestrated endothelial cells were seen in both the small caliber treated with plaque radiation for posterior uveal melanoma.9
and larger caliber vessels. There was atrophic retinal parenchyma Their overall incidence of radiation maculopathy was 43%. A 5%
surrounding these vessels with hypertrophy of glial cells and incidence of nonproliferative retinopathy was seen after 1 year
degeneration of neurons. They postulated that endothelial cell and 42% after 5 years. The most important predictor of nonprolif-
damage from radiation and free radical exposure was the inciting erative retinopathy in their study was the posterior location of
factor leading to a chain of events. These events included the the tumor. Guyer et al reported a radiation maculopathy incidence
SECTION 10

activation of the clotting cascade, capillary occlusion and forma- of 89% in their study of 218 patients who had received external
tion of incompetent capillary collaterals. beam radiation for paramacular choroidal melanomas.10 The
In 1992, Krebs et al described their histological findings after earliest and most common sign was macular edema, found in
examining an eye that had been enucleated 13 years following 61% of the patients at 1 year and 87% at 3 years. In their study of
60 Gy of external beam radiation and subsequent chemotherapy 32 patients with radiation retinopathy, Brown et al found hard
for embryonal rhabdomyosarcoma.19 ERG studies done prior to exudates in 85% of those treated with plaque radiation compared
enucleation showed severely reduced cone responses and no rod to 38% in those treated with external beam radiation.12 The pos-
responses. They concluded that radiation damage enhanced by sibility of leakage of serum products from necrotic tumor tissue
chemotherapy may have damaged the retinal vasculature and as a cause of the disparity in incidence could not be excluded. In
pigment epithelium with a secondary loss of photoreceptors. the group treated with plaque radiation, microaneurysms were seen
They noted that rods seemed to be more affected than cones. in 75%, intraretinal hemorrhages were seen in 65%, telangiectasia
Histologic studies of three primate eyes previously treated with were seen in 35%, cotton wool spots were seen in 30% and 20%
iodine-125 brachytherapy were done in 1999 by Robertson et al.20 had vascular sheathing. In the group treated with external beam
A theoretical choroidal tumor with apical height of 5 mm was radiation, microaneurysms and intraretinal hemorrhages were
assumed so that 80 Gy was to be delivered to the apex. Revised seen in 81% and 88% respectively, 38% had telangiectasia and
dose calculations revealed 5 mm apical doses ranged from 64 to cotton wool spots, and vascular sheathing was seen in 25%.
82 Gy and the inner scleral doses ranged from 280 to 420 Gy. In addition to the previously mentioned manifestations of radia-
Their findings showed an atrophic retina, retinal pigment epithe- tion retinopathy, other rare presentations of radiation retinopathy
lium, and choroid with macrophage invasion of the tissues in the including central retinal artery occlusion or central retinal vein
area overlying the site of the radioactive plaque. Trypsin digest occlusion have also been reported.12,15 Maberley et al have also
studies further confirmed capillary remodeling with loss of postulated that radiation may be a contributing factor to
endothelial cells and pericytes. idiopathic perifoveal telangiectasia.23
In 2003, Sefau et al described the histopathologic changes in
the choroidal circulation seen in three enucleated human eyes
from three patients after external beam radiation.21 The dose for PROLIFERATIVE RADIATION RETINOPATHY
the first patient treated for congenital hemangioma was unknown. The proliferative manifestations of radiation retinopathy include
In the second patient, the total dose used was 57.6 Gy followed any neovascularization of the retina or optic disk. Brown et al
with chemotherapy to treat malignant mesenchymoma. The third reported a higher rate of neovascularization after treatment with
patient had retinoblastoma initially treated with 36 Gy and a external beam radiation (43%) compared to plaque radiation
year later with 30 more Gy due to persistence of the tumor. (5%).12 Subsequent reports have not shown this disparity. Guyer
Findings similar to those seen in paving stone degeneration such et al reported an incidence of 6% after external beam radiation
as extensive loss of the retinal pigment epithelium and chorio- and Gunduz et al observed a proliferative retinopathy incidence
capillaris with adhesion of the degenerated retina to Bruch's of 1% at 1 year and 8% at 5 years after plaque radiation.9,10 In a
membrane were noted. study of 558 patients treated with external beam radiation for
choroidal melanoma, Gragoudas et al reported a proliferative
CLINICAL FEATURES retinopathy incidence of 3%.8 In addition to the possible manifes-
tations described above, an atypical case of subretinal neovascu-
larization originating from telangiectatic vessels has been
NONPROLIFERATIVE RADIATION reported.24 Spaide et al have also identified unusual vascular,
RETINOPATHY bleb-like choroidal complexes located at the outer border of
Most cases of radiation retinopathy are diagnosed between 1 choroidal neovascularization.25 They have proposed the term
and 3 years after treatment with rare cases reported as early as 'radiation-associated choroidal neovasculopathy' (RACN) to
1 month and as late as 15 years. The most common time of identify their observations.
onset is of course dose related but changes are infrequent before
a year and often become apparent at 18–24 months.9,11,12,22 In
general, the nonproliferative manifestations of radiation retino- DOSIMETRY
pathy consist of focal capillary closure, irregular dilation of sur- Dosimetry varies widely based on the method of radiation
rounding vessels, microaneurysms, intraretinal hemorrhages, delivery (plaque versus external beam), the type of lesion, and
2208 cotton wool spots, retinal exudates, retinal edema, nerve fiber layer location of therapy. Standard fractionation for external beam
Radiation Retinopathy

radiation is typically 1.8–2.0 Gy of radiation in single daily in four of seven eyes that had received chemotherapy in
doses until the total dose desired is achieved. The benefits of addition to external beam radiation.12 All four eyes progressed
fractionated radiation therapy had been established a few years to total blindness. Parsons et al observed four of 10 patients
prior to Moore and Stallard's use of radiation to treat ophthalmic treated with external beam radiation that developed radiation
tumors.1 Hyperfractionation refers to even smaller fractions retinopathy compared to two of two that were treated with
applied more than once daily over the same treatment time. external beam radiation in addition to chemotherapy.16
The theoretical benefits of hyperfractionation for the retina are With regard to diabetes and radiation retinopathy, Brown et
to allow normal retinal tissue to repair single-strand DNA al observed the only patient to develop proliferative retinopathy
breaks and limit late damage to double-strand breaks.22 There in the plaque radiation group was known to have diabetes. The
is also a potential to deliver a higher total radiation dose which minimal dose shown to induce foveal damage was 45 Gy,
may limit late sequelae. Randomized trials have not been whereas excluding this patient would have raised the minimal
undertaken to prove any clinical benefit of this approach. dose to 100 Gy.12 Parsons et al described a diabetic patient with
Reports of the incidence of retinopathy vary from 35% to 56% no evidence of retinopathy and normal visual acuity prior to
with doses above 45 Gy.8,12,16,22 Brown et al have reported an treatment for advanced skin cancer of the central face with
incidence of 85% with doses between 70 and 80 Gy.12 A specific external beam radiation.16 He subsequently developed radiation
threshold dose that will result in retinopathy has not been retinopathy in both eyes resulting in hand-motion vision by 47
determined. Factors such as varying radiation fractionation and months after treatment. Other large studies following plaque
hyperfractionation techniques, retinal dose measurement tech- therapy for melanoma have supported the theory that diabetes

CHAPTER 175
niques and co-morbid disease make establishing this threshold increases the risk of retinopathy following radiation.9
difficult.
In a review of patients who had undergone external beam DIFFERENTIAL DIAGNOSIS
radiation with varying fractionation schedules for various head
and neck tumors, Amoaku et al reported the threshold dose for Because the clinical features of radiation retinopathy are similar
radiation retinopathy was between 25 and 30 Gy.11 The mean to those seen in other retinal vasculopathies, it is important to
follow-up was 8 years. Parsons et al investigated radiation-induced elicit a thorough history and review any prior medical records
retinopathy according to total radiation dose and fraction size available. If a history of radiation exposure is obtained, impor-
both retrospectively from data in the literature and also prospec- tant details such as the date, dose, frequency and area of radiation
tively in 64 patients receiving external beam radiation for various should be reviewed. Information about the fields will allow an
extracranial head and neck tumors.16 A sigmoid dose–response estimate of whether the eye was or was not likely included in
curve was created indicating that with fraction sizes less than or the field or radiation.
equal to 2 Gy, total dose of 30 Gy seemed to be the threshold The clinical manifestations of diabetic retinopathy and hyper-
prior to the development of retinopathy. In a prospective study tensive retinopathy can be difficult to distinguish from radiation
of 42 patients with Graves ophthalmopathy, Robertson et al retinopathy. Other retinal vascular abnormalities such as arterial
observed the development of retinopathy in three patients or venous occlusive disease and telangiectasia must be differ-
within 3 years.7 The dose received by all patients was 20 Gy entiated. Ocular ischemic syndrome can also mimic radiation
delivered in 10 doses of 2 Gy. Their results were confounded by retinopathy although the intraretinal hemorrhages are generally
the fact that retinal microvascular abnormalities were observed found in the midperipheral retina whereas in radiation retinopa-
prior to radiation in one patient and the other two had factors thy, intraretinal hemorrhages are generally found diffusely
known to be associated with retinopathy such as uveitis, systemic throughout the retina or in the localized area of treatment.
hypertension and abnormal glucose levels. More recently, Monroe
et al identified 186 patients undergoing external beam radiation TREATMENT
for various head and neck tumors and followed them prospec-
tively for a median of 6.7 years.22 Their studies indicated the The natural history of radiation maculopathy is characterized
threshold for retinopathy was 40 Gy with standard fractionation. by progressive retinal damage with eventual resolution of edema
The threshold for retinopathy was 50 Gy with a hyperfraction- as ischemia develops. Proliferative retinopathy can result in
ation schedule. Hyperfractionation reduced the incidence of vitreous hemorrhage, retinal detachment or neovascular glaucoma
retinopathy by more than half compared to standard fractionation and loss of vision is often due to ischemia or optic neuropathy.
(13% vs 37%) in those patients who received more than 50 Gy. The optimal treatment of radiation retinopathy is not well
In the Brown et al study of radiation retinopathy, 20 eyes had established. Because of the similarities to diabetic retinopathy
been treated with plaque radiation and 16 eyes with external and branch vein occlusion, data from the Branch Retinal
beam radiation.12 They reported a higher mean dose of radiation Vein Occlusion Study (BRVO) and Early Treatment Diabetic
with plaque radiation was needed compared to external beam Retinopathy Study (ETDRS) has been extrapolated to study the
radiation therapy to produce foveopathy. The mean dose with use of laser photocoagulation for the treatment of macular
plaque radiation was 150 Gy, whereas a mean of 49 Gy was edema and proliferative retinopathy after radiation.26–28
needed with external beam radiation. In their study of plaque Hykin et al retrospectively studied the effect of focal laser- on
radiation therapy for posterior melanoma, Gunduz et al found a radiation-induced macular edema with a treatment group of 19
significantly higher risk for nonproliferative retinopathy with patients and a matched observation group of 23 patients.27 Their
doses above 57 Gy to the fovea.9 The median dose to the fovea results showed an initial 6-month modest improvement in
in those developing retinopathy was 70 Gy.9 visual acuity and resolution of macular edema in the treatment
group; however, at 2-year follow-up, no significant difference in
visual acuity was found compared to the observation group.
OTHER ASSOCIATIONS Other techniques to treat radiation maculopathy have been
Independent factors thought to exacerbate radiation retinopathy proposed such as the use of oral pentoxifylline to treat capillary
are chemotherapy and systemic conditions such as diabetes, nonperfusion, intravitreal triamcinolone acetonide or verteporfin
hypertension and collagen diseases.11 These conditions may photodynamic therapy, and hyperbaric oxygen.29–32 These treat-
decrease the threshold dose required to develop radiation ments have either been described in small cases series or have
retinopathy. For example, Brown et al found radiation retinopathy not proven to have lasting effects. 2209
RETINA AND VITREOUS

In a retrospective study of 14 eyes of 10 patients with


TABLE 175.1. Finger Classification of Radiation Retinopathy
proliferative radiation retinopathy, Kinyon et al reported a
91% rate of resolution after panretinal photocoagulation for Stage 1 (Findings Are Limited to Outside the Macula)
proliferative radiation retinopathy.28 The visual acuity, however,
went from an initial median of 20/90 to 20/400 after an average Cotton wool spots
follow-up of 75 months. Significant contributing factors to Retinal hemorrhages
visual loss included optic neuropathy, macular ischemia, and Retinal microaneursyms
vitreous hemorrhage.
Recently, Finger et al have proposed a four-stage classification Ghost vessels
for radiation retinopathy (Table 175.1).26 Based on this classi- Exudate
fication, they studied the effects of sector scatter laser photoco-
Uveal effusion
agulation through an interventional case series in two groups of
patients that had been previously treated with plaque radiation Choroidal atrophy
for posterior choroidal melanoma. The first group of 45 patients Choroidopathy
had signs of radiation retinopathy and the second group of 16
patients did not have any clinical evidence of radiation Retinal ischemia <5 DA
retinopathy but were considered 'high risk' based on the Stage 2 (Findings from Stage 1 but Found Within the Macula)
posterior location of the tumor.26 Radiation retinopathy in the
SECTION 10

Stage 3 (Any Stage 1–2 in Addition to the Following)


first group was defined as any iris or retinal neovascularization
or any stage-1 finding, with the exception of isolated ghost Retinal vascularization
vessels, uveal effusion, or chorioretinal atrophy. They reported Macular edema
a regression rate (defined as disappearance or regression of
initial treatment indications) of 64% and a loss of three lines or Stage 4 (Any Stage 1–3 in Addition to the Following)
more in visual acuity in 47% after a mean follow-up of 48 Vitreous hemorrhage
months. In a third of the patients who lost three lines of vision,
Retinal ischemia >5 DA
the cause was due to maculopathy. In the second group, three
patients developed radiation retinopathy which regressed with
further treatment. No patients lost more than three lines of
vision due to radiation maculopathy. The authors suggested
that sector scatter laser photocoagulation may regress radiation use of these growth factor inhibitors to treat permeability-
retinopathy under their proposed classification and preserve related pathology in radiation retinopathy, or to achieve
vision. The findings, however, were not statistically significant regression of new vessels. However, it is not likely to alter the
and further prospective studies were recommended. tendency to develop progressive ischemia or optic neuropathy,
With new treatments targeting vascular endothelial growth so the vision outcomes may not be good. Prospective studies
factors now available, perhaps future studies will involve the will be necessary.

REFERENCES
1. del Regato JA: The unfolding of American radiotherapy for posterior uveal melanoma. 18. Irvine AR, Wood IS: Radiation retinopathy
radiotherapy. Int J Radiat Oncol Biol Phys Arch Ophthalmol 1999; 117:609–614. as an experimental model for ischemic
1996; 35:5–14. 10. Guyer DR, Mukai S, Egan KM, et al: proliferative retinopathy and rubeosis iridis.
2. Moore RF: Choroidal sarcoma treated by Radiation maculopathy after proton beam Am J Ophthalmol 1987; 103:790–797.
the intraocular insertion of radon seeds. irradiation for choroidal melanoma. 19. Krebs IP, Krebs W, Merriam JC, et al:
Br J Ophthalmol 1930; 14:145–152. Ophthalmology 1992; 99:1278–1285. Radiation retinopathy: electron microscopy
3. Stallard HB: Radiant energy as (a) a 11. Amoaku WMK, Archer DB: Cephalic of retina and optic nerve. Histol Histopathol
pathogenic (b) a therapeutic agent in radiation and retinal vasculopathy. Eye 1990; 1992; 7:101–110.
ophthalmic disorders. Br J Ophthalmol 4:195–203. 20. Robertson DM, Garretson BR, de Venecia GB,
Monograph Suppl 1933; VI:70. 12. Brown GC, Shields JA, Sanborn G, et al: et al: Clinicopathologic correlation of
4. Cibis PA, Brown DVL: Retinal changes Radiation retinopathy. Ophthalmology radiation retinopathy in the rhesus monkey
following ionizing radiation. Am J 1982; 89:1494–1501. induced by iodine-125 episcleral plaque.
Ophthalmol 1955; 49:84–88. 13. Archer DB, Amoaku WM, Gardiner TA: Ophthalmic Pract 1999; 17:86–92.
5. Puusaari I, Heikkonen J, Kivela T: Ocular Radiation retinopathy – clinical, 21. Sefau SO, Dorey MW, Brownstein S, et al:
complications after iodine brachytherapy histopathological, ultrastructural and Radiation-induced chorioretinal degeneration:
for large uveal melanomas. Ophthalmology experimental correlations. Eye 1991; a clinicopathological report of three cases.
2004; 111:1768–1777. 5:239–251. Can J Ophthalmol 2003; 38:57–62.
6. Subramanian PS, Bressler NM, Miller NR: 14. Amoaku WM, Archer DB: Fluorescein 22. Monroe AT, Bhandare N, Morris CG,
Radiation retinopathy after fractionated angiographic features, natural course and Mendenhall WM: Preventing radiation
stereotactic radiotherapy for optic nerve treatment of radiation retinopathy. Eye retinopathy with hyperfractionation. Int J
sheath meningioma. Ophthalmology 2004; 1990; 4:195–203. Radiat Oncol Biol Phys 2005; 61:856–864.
111:565–567. 15. Hayreh SS: Post radiation retinopathy: 23. Maberley DAL, Yannuzzi LA, Gitter K, et al:
7. Robertson DM, Buettner H, Gorman CA, a fluorescence fundus angiographic study. Radiation exposure: a new risk factor for
et al: Retinal microvascular abnormalities in Br J Ophthalmol 1970; 54:705–714. idiopathic perifoveal telangiectasis.
patients treated with external radiation for 16. Parsons JT, Bova FJ, Fitzgerald CR, et al: Ophthalmology 1999; 106:2248–2253.
Graves ophthalmopathy. Arch Ophthalmol Radiation retinopathy after external beam 24. Boozalis GT, Schachat AP, Green WR:
2003; 121:652–657. irradiation: analysis of time-dose factors. Int Subretinal neovascularization from the
8. Gragoudas ES, Li W, Lane AM, et al: Risk J Radiat Oncol Biol Phys 1994; 30:765–773. retina in radiation retinopathy. Retina 1987;
factors for radiation maculopathy and 17. Egbert PR, Donaldson SS, Moazed K, 7:156–161.
papillopathy after intraocular irradiation. Rosenthal AR: Visual results and ocular 25. Spaide RF, Leys A, Herrmann-Delemazure B,
Ophthalmology 1999; 106:1571–1578. complications following radiotherapy for et al: Radiation-associated choroidal
9. Gunduz K, Shields CL, Shields JA, et al: retinoblastoma. Arch Ophthalmol 1978; neovasculopathy. Ophthalmology 1999;
Radiation retinopathy following plaque 96:1826–1830. 106:2254–2260.
2210
Radiation Retinopathy

26. Finger PT, Kurli M: Laser photocoagulation 29. Gupta P, Meisenberg B, Amin P, 32. Stanford MR: Retinopathy after irradiation
for radiation retinopathy after ophthalmic Pomeranz HD: Radiation retinopathy: the and hyperbaric oxygen. J R Soc Med 1984;
plaque radiation therapy. Br J Ophthalmol role of pentoxifylline. Retina 2001; 77:1041–1043.
2005; 89:730–738. 21:545–547.
27. Hykin PG, Shields CL, Shields JA, 30. Shields CL, Demirci H, Dai V, et al:
Arevalo JF: The efficacy of focal laser Intravitreal triamcinolone acetonide for
therapy in radiation-induced macular radiation maculopathy after plaque
edema. Ophthalmology 1998; radiotherapy for choroidal melanoma.
105:1425–1429. Retina 2005; 25:868–874.
28. Kinyoun JL, Lawrence BS, Barlow WE: 31. Bakri SJ, Beer PM: Photodynamic therapy
Proliferative radiation retinopathy. Arch for maculopathy due to radiation
Ophthalmol 1996; 114:1097–1100. retinopathy. Eye 2005; 19:795–799.

CHAPTER 175

2211
CHAPTER

176 Retinal Toxicity of Systemic Medications


Patrick D. Williams and David Callanan

Key Features: BOX 176.1 Systemic medications that cause retinal


• Systemic medications can produce posterior segment toxicity toxicity
through varying mechanisms
• Phenothiazines
• Direct toxicity to the retina
• Direct toxicity to the RPE • Thioridazine
• Vascular damage • Chlorpromazine
• Accumulation in tissue • Quinolines
• Cystoid macular edema • Chloroquine
• The American Academy of Ophthalmology has issued
• Hydroxychloroquine
recommendations for hydroxychloroquine screening
(see Box 176.2) • Drugs that cause crystal deposition
• Documented toxicity is rare • Talc
• Tamoxifen
• Canthaxanthine

INTRODUCTION • Methoxyflurane
• Cisplatin
Medications that are toxic to the retina can produce variable • Interferon
presentations and clinical courses. The retinal changes can be
• Didanosine
subtle or dramatic, and the damage can be benign or devas-
tating. The retina can demonstrate toxicity in numerous ways, • Niacin
including cystoid macular edema, damage to the retinal pigment • Clofazamine
epithelium (RPE), damage directly to the retina, and accumula- • Desferoxamine
tion of the substance in tissue. Knowledge of the known toxi- • Methanol
cities can help the practitioner to decipher the sometimes
• Quinine
unusual clinical presentations and treat patients accordingly.
• Antiepileptics
• Vigabatrin
PHENOTHIAZINES • Lamotrigine

The phenothiazine class of compounds is used to treat


psychiatric conditions such as acute psychosis. Chlorpromazine CLINICAL FEATURES
(Thorazine) and thioridazine (Mellaril) are known to cause Patients typically complain of blurred vision, dyschromatopsia
retinopathy with the high doses sometimes used to treat acutely (brownish or reddish vision), and nyctalopia with acute
psychotic patients. Chlorpromazine is used to treat intractable thioridazine toxicity.3–5 The visual symptoms can be variable,
hiccoughs as well as psychiatric diseases. and acute toxicity may present with a normal retina or a granular
stippling of the RPE with or without pigment clumping. More
chronic disease can cause patients to develop geographic
THIORIDAZINE ‘nummular’ areas of atrophy of the RPE and retina (Fig. 176.1).1
Early cessation of the medication may allow for some return of
visual acuity.6 The retina and RPE changes usually progress,
GENERAL INFORMATION suggesting atrophy of previously damaged cells.1,6
Thioridazine has been used for acute psychosis since the 1950s.
A dose of greater than 800 mg/day of thioridazine is more likely
to produce toxicity, and 2 weeks of high-dose therapy is com- ANCILLARY TESTING
monly required for retinopathy.1 A high daily dosage is Ancillary testing can confirm thioridazine toxicity, although it
considered to be a greater risk factor than cumulative dosage. is unclear if it can reveal preclinical disease. The fluorescein
Although toxicity has been reported with chronic use at doses angiogram shows irregularity of the RPE initially, with severe
less than 800 mg/day, the incidence is extremely low.2 atrophy of the RPE and choriocapillaris in the late stages of
2213
RETINA AND VITREOUS

extensively until after World War II. The toxic side effects of
chloroquine and hydroxychloroquine became apparent with the
increasing use for rheumatoid diseases, as these diseases
required higher doses and chronic treatment. The first reported
case of chloroquine retinopathy was a case report of pigmentary
retinopathy of unknown origin associated with systemic lupus
erythematosus.18

CHLOROQUINE
Chloroquine can cause retinal damage with long-term daily use.
Daily doses exceeding 250 mg and cumulative doses between
100 and 300 g are typically required to affect the posterior
segment.19 The daily dose may be more important than the
cumulative dose. Doses at or below 3–3.5 mg kg–1 day–1 appear
to prevent toxicity.20,21 Maintaining a low daily dose appears to
control the risk of toxicity, even with very high cumulative
FIGURE 176.1. Right eye of patient that used Mellaril (thioridazine) for
doses.22
SECTION 10

many years with nummular areas of hypopigmented RPE.

HYDROXYCHLOROQUINE
1
toxicity. Fluoroscein angiogram changes can be more striking Hydroxychloroquine has largely supplanted chloroquine for the
than clinical findings. Visual fields, electroretinogram, and treatment of rheumatoid diseases, in no small part due to its
electrooculogram are variably affected. Visual fields can have improved safety profile. Although hydroxychloroquine toxicity
varying ring or paracentral scotomata or constriction.4 Electro- has been well documented, the incidence of hydroxychloroquine
retinography can be normal or can have photopic and scotopic retinal toxicity is much lower than that of chloroquine.23–25 The
amplitude reductions.6,7 Electroretinography changes, like visual accepted safe dose is regarded as 6.5 mg kg–1 day–1. Below this
symptoms, are reversible with quick cessation of medication. threshold, the incidence of retinopathy is rare, but it has been
reported.26 At doses below 6.5 mg kg–1 day–1, 6 years of treat-
ment is typically required before any retinopathy is appreciated.
PATHOPHYSIOLOGY The most recent estimate of the incidence of toxicity at normal
The toxicity of thioridazine may be due to its piperidyl side doses for greater than 6 years duration was 0.5%.27 Patients
chain. The only other phenothiazine with a piperidyl side chain using low-dose hydroxychloroquine therapy with massive
was NP-207, a drug never marketed. Severe retinopathies, cumulative doses can still avoid retinopathy.28
similar to thioridazine toxicity, were found in early clinical
trials.8 Pathology specimens of eyes with thioridazine toxicity
show initial atrophy and disorganization of the photoreceptors CLINICAL FEATURES
followed by loss of the choriocapillaris and RPE.9 Both The most common early symptoms of toxicity are paracentral
thioridazine and chlorpromazine bind to melanin and show scotomata. As toxicity progresses, patients can also report a loss
accumulation in the melanin granules of pigmented tissues.10–12 of acuity, nyctalopia, and photophobia. Dyschromatopsia can
Phenothiazine toxicity may involve dopamine metabolism in occur as well.29 Toxicity is not limited to the retina. Poliosis,
the eye.13 Dopamine regulates melatonin, which is involved in subepithelial corneal deposits, decreased corneal sensitivity, and
retinal physiology and prevents light toxicity. even sixth cranial nerve palsies may develop.29 These findings
were much more common with chloroquine than what is
CHLORPROMAZINE reported now with hydroxychloroquine.
Early disease may not reveal retinal changes on biomicro-
scopy. The earliest signs include loss of the foveal reflex and
CLINICAL FEATURES nonspecific changes in the macular RPE.30 In the later stages,
Chlorpromazine causes hyperpigmentation of tissues, including there is granularity of the RPE in the classic bull’s-eye pattern.31
sun-exposed skin and conjunctiva.14–16 Corneal deposits and a Bull’s-eye maculopathy consists of a circular area of RPE hyper-
characteristic cataract can develop. Retinopathy associated with pigmentation centered on the fovea. An oval area of RPE hypo-
chlorpromazine is rare. When it occurs, it presents as a diffuse pigmentation around the circle creates the appearance of a bull’s
hyperpigmentation of the RPE. The changes are finer and more eye (Fig. 176.2). Subsequent arteriolar narrowing, optic disk
diffuse than those of thioridazine toxicity. Patients may have pallor, and peripheral RPE degeneration develop as well, giving
arteriolar attenuation and optic nerve pallor. Alternatively, an appearance similar to retinitis pigmentosa (Fig. 176.3).
chlorpromazine toxicity may present with more well-defined There is no treatment besides drug cessation. Unfortunately,
patches of depigmentation.17 Very large doses (~1–2 g/day) are the toxicity can often progress even after the drug is stopped. It
required for retinopathy.14–16 will eventually stabilize, but the central acuity can be sig-
nificantly affected. In some cases of early toxicity, visual acuity
QUINOLINES has improved after stopping therapy.

GENERAL INFORMATION ANCILLARY TESTING


The quinoline derivative antimalarials include both chloro- There is still controversy about whom and how to monitor
quine and hydroxychloroquine (Plaquenil). Chloroquine was these patients for early signs of toxicity. As stated above,
first used to treat malaria in the mid-twentieth century. It hydroxychloroquine toxicity is exceedingly rare in doses under
2214 was first discovered in Germany in 1934, but was not used 6.5 mg kg–1 day–1 and less than 5 years duration. Other factors,
Retinal Toxicity of Systemic Medications

toxicity.34,35 The standard Pseudoisochromatic Plates part 2


and the AO HRR tests are more reliable predictors of retinal
toxicity.35 A baseline color vision examination may be beneficial
in males to later differentiate inherited color deficiencies from
equivocal cases of toxicity. Also, color vision testing can be use-
ful in elderly patients, in which accurate perimetry is difficult.

Amsler Grid
The Amsler grid is widely used for evaluation of early hydroxy-
chloroquine toxicity, and is recommended by the AAO.32 It is
inexpensive and easy to administer, making it ideal for clinic
and home use. The scotomata produced on Amsler grid cor-
relate well with those found on perimetry.36 In patients with
normal perimetry, the Amsler grid can sometimes reveal relative
scotomata.37
Modifications in the Amsler grid may improve its sensitivity.
Red Amsler grid testing yields a higher incidence of scotomata,
FIGURE 176.2. Classic bull’s-eye pattern of retinopathy due to
but the sensitivity is questionable due to a lack of confirmation

CHAPTER 176
chloroquine toxicity.
of many positive tests.38,39 These may be early toxic changes or
may be false-positive results. Recently, the threshold Amsler
grid has been proposed as a test more sensitive than even the
red Amsler grid.38 The patient looks through cross-polarizing
filters at a standard Amsler grid, thereby reducing contrast sen-
sitivity and eliciting milder relative scotomata. The sensitivity
of threshold Amsler grid testing is much higher than that of
either the standard Amsler grid or the red Amsler grid.38 As
with the red Amsler grid, the true specificity is difficult to
ascertain.

Perimetry
Perimetry has become commonplace in the evaluation of
hydroxychloroquine patients. Both kinetic (Goldmann) and
static (Humphrey) perimetry have been used to evaluate
hydroxychloroquine and chloroquine toxicity. Humphrey 10–2
visual fields allow focused examination of macular function.40
The sensitivity and specificity of Humphrey static perimetry
has been evaluated and is accepted as reasonable for decisions
FIGURE 176.3. Advanced chloroquine toxicity mimicking retinitis whether to continue or withdraw medication.41 Red target peri-
pigmentosa with bone spicules and arteriolar narrowing.
metry appears to be more sensitive than white target perimetry
(91.3% and 78%, respectively), but the specificity of red target
perimetry is worse (57.8% and 84%, respectively). The AAO
however, can influence the risk of toxicity. In 2002, the currently recommends either Amsler grid testing or Humphrey
American Academy of Ophthalmology (AAO) published a 10–2 perimetry to monitor patients taking hydroxychloroquine.32
guideline for hydroxychloroquine screening.32 Not only do dose
and duration of therapy affect toxicity risk, but liver or renal Fluoroscein Angiogram
disease, obesity, concomitant retinal disease, and age over 60 Fluoroscein angiography can highlight the changes seen on
may play a role. Liver or renal disease can affect chloroquine ophthalmologic exam (Fig. 176.4). It is not mandatory for
and hydroxychloroquine levels because these drugs are cleared
by both organs. Obesity is a risk factor because neither drug
is stored in fat cells. People in which fat contributes a large
percentage of total body weight must be placed on relatively
lower doses.
The clinical examination is the basis of all ophthalmologic
testing for chloroquine and hydroxychloroquine toxicity.32 The
AAO recommends a baseline examination, although the efficacy
has not been proven. Routine examinations should be main-
tained based on risk stratification. Unfortunately, early toxicity
may be missed by only relying on clinical examination. Con-
sidering the rare incidence of hydroxychloroquine toxicity, there
has been much debate over the costs and benefits of extensive
ancillary screening.

Color Vision
There are no changes in color vision specific to chloroquine or
hydroxychloroquine retinopathy, but nonspecific changes may
occur with disease.33 In general, Ishihara color plates and the FIGURE 176.4. Fluoroscein angiogram highlighting bull’s eye
Farnsworth D-15 panel have poor sensitivity for early retinopathy due to chloroquine toxicity. 2215
RETINA AND VITREOUS

routine evaluation, but can help differentiate toxicity from other


maculopathies.32 BOX 176.2 Screening for hydroxychloroquine toxicity
• Dosage less than 6.5 mg/kg lean body weight considered
Electrooculogram safe for 5 years
Early reports suggested that the electrooculogram (EOG) may be • Risk factors for retinopathy include:
a useful tool in chloroquine toxicity screening.42,43 An Arden • Liver or renal disease
ratio of less than 1.85 appeared to signify toxicity. EOG has a
poor specificity, however, and active episodes of rheumatologic • Obesity
disease can also alter the EOG.44 The most recent review of • Concomitant retinal disease
EOG findings suggested a sensitivity of 50% and a specificity • Use greater than 5 years
of 54%.45 • Age greater than 60 years
• Recommended ancillary testing:
Multifocal Electroretinogram
Multifocal electroretinogram (mfERG) is a newer screening • Amsler grid or 10–2 perimetry for everyone screened
modality for hydroxychloroquine toxicity. Decrease in mfERG • Red test objects can improve sensitivity but may
amplitudes have been demonstrated in documented hydroxy- reduce specificity
chloroquine toxicity, consistent with the decline in visual • Fluoroscein angiography can assist when concomitant
acuity.46 More interestingly, changes in mfERG have been noted retinal disease is present
SECTION 10

in patients with no other evidence of toxicity.47–49 The signifi- • Baseline color vision testing can separate congenital
cance of these changes is unknown, as there is no comparison deficits from acquired retinopathy
to known parameters. Abnormalities were noted in patients
• mfERG can confirm retinopathy
with less than 5 years of hydroxychloroquine use, suggesting an
early subclinical toxicity that was reversible with discontinua- • May reveal preclinical disease
tion of the medication. Normal mfERG examinations have also
been found in patients taking the medication for over 10 years,
however.49 Therefore, there are likely some individual differ-
TALC
ences in mfERG responses, just as there are individual dif- General Information
ferences in frank toxicity. Multifocal ERG may become standard Talc is the common name for hydrous magnesium silicate. It is
in hydroxychloroquine screening, but more information is an inert filler used in oral medication such as methylphenidate
needed to interpret abnormal results when no other abnor- (Ritalin) and can cause crystalline retinopathy in intravenous
malities are found. drug abusers.55–57 With intravenous injection, crystals smaller
than 7 mm can pass through the lungs. The large talc crystals
are trapped in the lungs, but pulmonary shunt vessels develop
PATHOPHYSIOLOGY with repeated injections.57 The crystals can then enter the
The pathophysiology of chloroquine and hydroxychloroquine arteriolar system and embolize to the eye. This likely explains
toxicity is, for the most part, unknown.50 Chloroquine is con- why chronic intravenous drug abuse is usually required before
centrated in the RPE and uvea and is bound by melanin. Due to retinopathy develops.
the fact that the toxicity appears to be concentrated in the
macula, some have hypothesized a direct effect on cone cells. Clinical Features
Chloroquine toxicity affects lysosomal metabolism in the Patients can be asymptomatic with talc crystalline retino-
neurosensory retina and RPE.51 In a rat model, lysosomal pH pathy.56 The crystals appear reflective and migrate to the end
rises with in vivo administration of chloroquine. Numerous arterioles (Fig. 176.5). With persistent crystal deposition, an
membranous cytoplasmic bodies can be seen in retinal neurons, ischemic retinopathy develops.56,58,59 Ischemia is confined to
including cone cells but not rod cells. RPE metabolism is areas with capillary nonperfusion due to emboli. Other sequelae
affected, with increase in lysosomal associated organelles and of ischemia, such as microaneurysms, cotton wool spots, and
lysosomal accumulation of rod outer segments. The difference collateral vessels may also be present. Neovascularization of the
in toxicity between chloroquine and hydroxylchloroquine may
be due to the difference in effect on lysosomal metabolism.52 In
vitro administration of chloroquine to RPE cells causes greater
accumulation of lipofuscin than does administration of
hydroxychloroquine. Lipofuscin accumulation is increased by
elevated lysosomal pH and oxidative breakdown of phago-
cytosed material.
Changes in lipid metabolism are also present in the neuro-
sensory retina.53,54 An accumulation of lipids, which can cause
direct neuronal damage, has been documented with chloro-
quine toxicity. Based on comparisons to electroretinographic
findings in a rat model, lipidosis is not likely the direct cause of
toxicity.

CRYSTALLINE RETINOPATHIES
Several chemicals can cause deposition of crystals in the retina.
These include talc, tamoxifen, canthaxanthin, and methoxy-
flurane. In the majority of these cases, the patient is unaware of FIGURE 176.5. Talc crystals in the right eye of a patient with chronic
the presence of these crystals and they are found on a routine intravenous drug use. The patient had developed severe hypertension
2216 dilated retinal exam. and suffered an intracranial hemorrhage as well.
Retinal Toxicity of Systemic Medications

disk or elsewhere can develop, leading to vitreous hemorrhage


and retinal detachment.58 Vision loss can also occur due to
macular fibrosis.60 Animal model of talc retinopathy suggests
that the ischemic response is similar to other vascular diseases
such as diabetes and sickle cell retinopathy.61–63 Retinal photo-
coagulation to the ischemic retina can prevent complications of
proliferative disease, just as in other vascular retinopathies.64

TAMOXIFEN
General Information
Tamoxifen is an estrogen receptor antagonist typically used in
treating estrogen-receptor positive breast cancer. The number of
cases with tamoxifen retinopathy has greatly declined since the
daily dose was lowered to 10–20 mg/day, but retinopathy is still
possible.65–67 The classic descriptions of tamoxifen retinopathy
were associated with doses of 180 mg/day in the treatment of
FIGURE 176.6. Right eye of a patient that used canthaxanthin for
metastatic disease.68,69 While tamoxifen retinopathy can

CHAPTER 176
several months. There are numerous tiny crystals that are extremely
happen at standard doses, it is rare. There is no evidence to reflective around the fovea.
support routine screening of patients taking tamoxifen at Photo courtesy of Barbara Blodi, MD.
standard doses.70,71 Even if patients develop retinal crystals,
there is typically little change in retinal function.
Ancillary Testing
Clinical Features Ten-degree static perimetry can show reduced sensitivity in
Patients develop white, glistening crystals in the inner plexi- patients with retinopathy, but patients taking canthaxanthine
form layer and nerve fiber layer. Varying degrees of pigmentary without clinical toxicity have normal visual fields.83 Electro-
changes may also be present. With high doses, patients may physiology studies have not been consistent across studies, but
lose vision due to direct retinal toxicity as well as macular some alterations have occurred clinically and in animal
edema and optic neuropathy.72 Patients can have improvement models.84–87
in vision with termination of tamoxifen therapy.73
Pathophysiology
Ancillary Testing Based on histopathology, the crystals settle in the nerve fiber
Electroretinography demonstrates a decrease in the photopic layer and can be found throughout the retina, even though
and scotopic a- and b-wave amplitudes.73 Fluoroscein clinically they are only apparent in the macula.88 The crystals
angiography can highlight the cystoid macular edema. Optical are actually canthaxanthine–lipoprotein complexes in which
coherence tomography also demonstrates the cystoid spaces as only the central portion can be appreciated clinically.
well as photoreceptor disruption.74

Pathophysiology METHOXYFLURANE
Small intracellular lesions (3–10 mm in diameter) in the macula Methoxyflurane is an inhalational anesthetic that metabolizes
and larger extracellular crystals in the paramacular region into oxalate crystals. With extended use, renal failure ensues
(30–35 mm in diameter) have been described by electron due to the oxalate deposition in the kidney. A flecked retinal
microscopy.69 These lesions were assumed to be the clinically appearance with copious small yellow-white punctate lesions in
apparent crystals and seemed to represent the products of the posterior pole and mid-periphery has been described
axonal degeneration. (Fig. 176.7).89 Toxicity due to chronic illicit abuse of
methoxyflurane has also been reported.90 Cotton wool spots,
large retinal crystals in a periarteriolar pattern, and crystals in
CANTHAXANTHINE the RPE were clinically apparent. Histopathology has
General Information demonstrated oxalate crystals in the inner retina and RPE, as
Canthaxanthine has been used as an oral tanning agent, but its well as throughout the body.91
use seems to have diminished since the reports of deposits in The relationship between retinal changes in methoxyflurane
the retina. It is a naturally occurring carotenoid also used to toxicity and primary oxaluria is unknown. Both flecked retina
treat vitiligo and disorders involving photosensitivity. It also can and crystalline retinopathy have been reported in primary
be found in common foods and has been suggested to be toxic disease.92–95 Unlike methoxyflurane toxicity, pigmented patches
with large dietary ingestion.75 The first reports of retinopathy were in the RPE have been described in primary oxaluria.
in the European literature.76–79 In cumulative doses greater than
60 g, a majority of patients have clinically apparent retinopathy.79,80 CISPLATIN
Clinical Features Cisplatin has been used for the treatment of malignant nervous
In high doses for chronic use, canthaxanthine produces a cha- system tumors as well as metastatic breast cancer and is
racteristic crystalline retinopathy with an annular distribution commonly used in concert with other chemotherapeutic agents.
of glistening crystals.76–78,80,81 The ring of crystals is localized in It binds directly to DNA to inhibit normal DNA functioning.
the macula and is centered on the fovea (Fig. 176.6). Most Nephrotoxicity, ototoxicity, and peripheral neuropathy are all
patients with clinically apparent crystals are asymptomatic. common. Transient blurred vision is also common in high-dose
Patients can, however, develop metamorphopsia or decreased intravenous infusion.96
vision. The retinopathy is reversible, with slow resolution of the The platinum in cisplatin is believed to cause direct retinal
crystals over years.82 toxicity when administered directly into the carotid artery.97–99 2217
RETINA AND VITREOUS

FIGURE 176.7. Left eye of a patient with retinal oxalosis due to


SECTION 10

methoxyflurane anesthesia.
Photo courtesy of Daniel Albert, MD.

FIGURE 176.8. Left eye of a patient treated with interferon for


hepatitis C. Cotton wool spots are apparent.
It has also been documented in an inadvertently high dose given
intravenously.100 These patients can develop a pigmentary
retinopathy with severe vision loss in the eye ipsilateral to the
carotid infusion.97,98 The a-wave is attenuated and the b-wave begins within the first few months of therapy and the retinal
is lost on electrophysiology. Histopathology reveals normal signs fade with cessation of interferon.103–110 Cystoid macular
photoreceptors, but a splitting of the outer plexiform layer, edema has been reported and severe, permanent vision loss can
which may explain the loss of the b-wave on ERG.100 occur due to arteriolar occlusion.108,109 Retinal changes
Another form of retinopathy associated with cisplatin use commonly occur in asymptomatic patients, suggesting the need
involves an ischemic maculopathy and optic neuropathy.97,99 for ophthalmologic screening concurrent with therapy.106,110
Patients can present with arteriolar attenuation, cotton wool
spots, retinal hemorrhages, and macular exudates. Optic nerve
head edema may be present, and these patients can have PATHOLOGY
significant visual loss. Permanent vision loss may be due to Based on analysis of intravitreal interferon in the rabbit eye,
the optic nerve disease, as patients with only retinopathy interferon retinopathy appears to have an immune-mediated
had improvement in both symptoms and signs.99 BCNU etiology.111 Complement pathway activation and immune
(carmustine) may account for some of the toxicity, as it can complex deposition may be the underlying cause.
produce a similar retinopathy without concomitant cisplatin
treatment.97,101 DIDANOSINE
INTERFERON The 2„,3„ dideoxyinosine (didanosine) is an antiretroviral
medication used in the treatment of HIV. Pancreatitis and
peripheral neuropathy are well described complications.112 It
GENERAL INFORMATION can cause peripheral RPE atrophy in children without changes
Interferon alpha is a common adjuvant in the treatment of in central visual acuity.113–115 The toxicity appears to be dose
malignant melanoma as well as renal cell carcinoma, dependent and is common in the pediatric population (7.0%).113
lymphoma, and leukemia. Alone or in combination with Didanosine is also associated with visual field deficits in
ribavirin, interferon is now the standard of care for treatment of adults.116 In a report of two cases, an annular scotoma occurred
hepatitis C. An ischemic retinopathy can develop in patients in one patient and a mild increase in the physiologic blind spot
taking interferon, in some cases causing serious visual decline. occurred in the other. Both patients had abnormal EOG (Arden
Many of the descriptions of interferon retinopathy are in the ratio < 1.6 in both eyes of both patients), suggesting diffuse
Japanese language literature.102 RPE dysfunction.
Histopathologic examination has revealed loss of the RPE
with surrounding hypertrophy or hypopigmentation.114 Partial
CLINICAL FINDINGS loss of the choriocapillaris and neurosensory retina were present
The earliest reports of interferon retinopathy in the English in the areas of RPE loss. Electron microscopy demonstrated
literature were in cancer patients receiving combination chemo- lamellar inclusion and cytoplasmic bodies in the RPE, sug-
therapy.103 Patients developed common sequelae of retinal gesting that the primary toxicity is at the level of the RPE.
ischemia, including capillary nonperfusion, cotton wool spots,
retinal hemorrhages, and occlusive disease (Fig. 176.8). Sub- NIACIN
sequently, corroborating reports have described ischemic retino-
pathy in both cancer treatment and hepatitis C treatment.104–107
A similar retinopathy develops in patients undergoing treat-
GENERAL INFORMATION
ment for hepatitis C, suggesting that the pathophysiology is Nicotinic acid (Niacin) is both a vitamin and a medication that
2218 similar regardless of the underlying disease.104,106 Retinopathy reduces cholesterol levels when ingested orally in large doses.117
Retinal Toxicity of Systemic Medications

It causes spectrum alterations in both lipid and cholesterol riminophenazines being studied now as possible new agents for
metabolism and is relatively inexpensive. Common side effects, use in leprosy and tuberculosis.126
such as flushing, rise in uric acid levels, and gastritis have
curtailed its use in exchange for other cholesterol reducing
medications.118 An extended-release formulation of nicotinic ANCILLARY TESTING
acid (Niaspan) has been introduced as an alternative for The angiogram in the above mentioned HIV patients shows an
lowering of cholesterol levels.119 The side effect profile is annular area of RPE depigmentation in the macula. Electro-
improved in comparison to Niacin, and no reports of macular retinography shows a decrease in the amplitude of the b-wave
edema have been published. under both scotopic and photopic conditions. There is also a
delay in the implicit time.135,136
CLINICAL FEATURES
Cystoid macular edema is a rare but well known complication PATHOPHYSIOLOGY
of high-dose niacin use and can be associated with blurred Riminophenazines accumulate in the intracellular space of
vision.112–123 Most reports have documented CME associated a mononuclear phagocytic cells and also in adipose tissue. The
dosage of 3 g/day, but CME has been reported with as little as mechanism of injury to the RPE or retina is unknown. In two
1.5 g/day.124 The clinical appearance of CME is similar to the of the well documented cases, the patients received more than
edema caused by numerous other etiologies, however no other 40 g total of clofazimine. The dosage used was 200 mg/day in

CHAPTER 176
retinal changes are typically found with niacin maculopathy. one patient and 300 mg/day in the other.135,136
The edema rapidly improves after discontinuation of niacin
with resolution and improvement in vision commonly within DEFEROXAMINE
2 weeks.120–123
GENERAL INFORMATION
ANCILLARY TESTING This drug is most commonly used for treatment of
Fluoroscein angiography does not reveal any leakage.120–123 hemosiderosis. Excessive levels of iron can lead to cardiac
Thus, nicotinic acid maculopathy fits with retinitis pigmentosa failure; so the treatment is often life-saving. The most common
and Goldman–Favre disease as causes of ‘angiographically predisposing factor is repetitive blood transfusions in patients
negative’ cystoid macular edema. Optical coherence tomo- with thalassemia or aplastic anemia.138–141 The drug chelates or
graphy does, however, demonstrate cystoid spaces in the outer binds copper, iron, aluminum, and zinc. The iron present in
plexiform layer.125 The author postulated that this finding con- normal hemoglobin is not chelated. Patients with renal failure
firms the cystic nature of the toxicity and makes other possible and high aluminum levels are also sometimes treated with
etiologies, such as muller cell engorgement, less likely. Minor deferoxamine.142,143 Deferoxamine can be given subcutaneously,
disruptions in the blood-ocular barrier may cause leakage too intramuscularly, or sometimes intravenously.
slow for fluoroscein angiography, but still result in edema.
CLINICAL FEATURES
CLOFAZIMINE
Patients present with an acute onset of decreased vision,
nyctalopia, ring scotoma, or color vision abnormalities as early
GENERAL INFORMATION as 7–10 days after treatment.144–148 Patients can also develop
Clofazimine is one of a class of drugs known as rimino- symptoms after several months of treatment. The fundus may
phenazines.126 This type of drug was developed for treatment of appear normal initially, but later develops a salt and pepper-like
Mycobacterium tuberculosis. They have since been tried in RPE abnormality (Fig. 176.9). Haimovici et al and Gass have
other mycobacterial diseases, including Mycobacterium leprae reported a loss of transparency or opacification of the outer
and Mycobacterium avium complex (MAC).127,128 Clofazimine retina and RPE in the early stages of toxicity.144,149 This is
has been approved by the World Health Organization for the
treatment of leprosy and in particular dapsone-resistant cases.
It is currently under increased study for use in multidrug
resistant tuberculosis as well as systemic or discoid lupus
erythematosus.129,130

CLINICAL FEATURES
The most common side-effect is a brownish discoloration of the
skin that occurs in 75–100% of patients after a few weeks of
treatment. It also causes brownish discoloration of the
conjunctiva, tears, and thin superficial lines in the cornea.131,132
There are only a few brief reports of pigmentary retinal changes
in patients with leprosy treated with clofazimine.132–134 There
are three reports of a bull’s eye type of retinopathy in patients
treated with clofazimine.135–137 All of these occurred in HIV
positive individuals. At least one eye in these patients had
evidence of an infectious retinitis. All of the patients died
within months of the detection of the retinopathy and there was
no pathologic specimen obtained. Patients that are HIV positive FIGURE 176.9. Right eye of patient with aplastic anemia. After
and treated with clofazimine may have an increased risk for receiving multiple transfusions, the patient developed hemosiderosis
retinal toxicity and should be monitored. There are other treated with deferoxamine. Mottling of the RPE is shown. 2219
RETINA AND VITREOUS

accompanied by late hyperfluorescent staining in the macula. methanol toxicity. When methanol is ingested, an anion gap
This then progresses to RPE hypopigmentation and mottling on metabolic acidosis occurs with acute renal failure. Fomepizole is
the angiogram and a decrease in the late hyperfluorescence. a competitive alcohol dehydrogenase inhibitor with demonstrated
Haimovici also reported patients with peripapillary, papillo- efficacy in methanol poisoning.158 Electophysiologic testing sug-
macular, and paramacular changes similar to those described gests that it benefits retinal toxicity as well as systemic toxicity.159
above. Gonzalez and co-workers reported two patients treated
with deferoxamine who presented with vitelliform lesions.150 It
is not known whether the vitelliform lesions were present prior CLINICAL FEATURES
to treatment with deferoxamine. Both patients were 70 years Patients develop blurred vision and scotomata within hours of
old so a preexisting condition can not be ruled out. Neither ingestion.160,161 Optic nerve hyperemia and posterior retinal
patient improved after discontinuation of deferoxamine. One of edema are present. If the toxicity is not reversed, permanent
the patients had an abnormal ERG and showed more typical vision loss and optic atrophy ensue.
RPE mottling 1 year after the drug was stopped. Patients can
also develop a high-frequency sensorineural hearing loss.146,147
The visual symptoms and hearing loss often diminish when PATHOPHYSIOLOGY
the drug is stopped, but not all patients fully recover. Optic Methanol metabolizes into formic acid, which is the cause of
neuropathy has been reported as well.144,145,147,151 the neurotoxicity.162,163 It is a mitochondrial toxin that causes
direct damage to the ganglion cells and optic nerve. Electro-
SECTION 10

physiologic and histopathologic examinations have suggested a


ANCILLARY TESTING direct effect to the RPE as well as the neurosensory retina.
The ERG is often abnormal with increased implicit times and
a- and b-wave diminution under either scotopic or photopic QUININE
conditions.144,152,153 The EOG shows a reduced light peak to
dark trough or Arden ratio. The EOG may serve as an early Quinine is an antimalarial now used in the treatment of muscle
detector of toxicity or monitoring test.153 An audiogram often cramps and restless leg syndrome. It is the active ingredient in
shows high-frequency loss.146,147 The ERG and EOG changes tonic water, which was used by sailors as a malaria prophylactic.
can also improve following discontinuation of deferoxamine. Toxicity can be both acute and chronic.164–167
The syndrome of acute toxicity is known as cinchonism,
from the natural source of quinine – the cinchona bark.
PATHOPHYSIOLOGY Cinchonism consists of headache, dizziness, hearing loss,
Light and electron microscopy evaluation of the eyes from a nausea, and tremor.168 The pupils can be poorly responsive
patient with deferoxamine toxicity showed RPE degeneration.154 during unconsciousness, and vermiform movements of the
The patient had decreased vision and a subnormal EOG. The pupils have been described.166 Patients can lose consciousness
changes in the RPE included loss of microvilli from the apical and, if they survive, awake blind. While vision may return,
surface, patchy depigmentation, vacuolation of the cytoplasm, patients may be left with permanent, severe visual field con-
and swelling with calcification of mitochondria. Bruch’s mem- striction. During the acute phase, the retinal arterioles can be
brane overlying these degenerated RPE cells was abnormally normal with mild venous distention. Within weeks to months,
thickened with accumulation of elastic fibers and collagen. At the arterioles become severely constricted and the optic nerve
least two teams of investigators have suggested that deferoxamine becomes atrophic.
may increase the transport of copper, out of cells and raise the There has been some debate regarding the mechanism of
extracellular concentration of copper.151,155 Measurements of retinal toxicity. The classic theory included ischemia due to
copper levels in the cerebrospinal fluid of a few patients are vascular constriction.165–167 More recent studies have suggested
compatible with this hypothesis. A model of deferoxamine a direct retinal toxicity, however.165–169 Electrophysiology,
toxicity in the albino rat demonstrated increased toxicity with including ERG, EOG, and VEP, can be abnormal in the acute
light and oxygen exposure.156 This was postulated to be related phase of toxicity, and ERG and histopathology are suggestive of
to increased free radical production. There is no further diffuse damage to neurosensory retina. Even without signs of
evidence to support this hypothesis. The exact cause of the RPE quinine toxicity, ERG demonstrates a transient decrease in
toxicity from deferoxamine remains unknown. It may be a photoreceptor function at therapeutic doses.170
direct effect of the drug or a secondary effect from chelation of Despite the evidence of direct retinal toxicity, recent reports
divalent metal ions. One study in albino rats showed a have proposed the treatment of blindness due to cinchonism
reduction in toxicity as measured by electrophysiology when it with vasodilating agents.171 The results have not been tested in
was conjugated with hydroxyethyl starch.157 There are at least comparative trials.
two new agents, deferiprone and deferasirox, under study as
replacements for deferoxamine or as combination therapy.138,139 ANTIEPILEPTICS
The use of these chelating agents is necessary to prevent the
dire consequences of iron overload. It is hoped that their side
effects can be minimized. Any patient treated with deferoxamine
VIGABATRIN
should be carefully monitored for visual changes. Vigabatrin is an irreversible inhibitor of GABA transaminase
used in the treatment of pediatric and adult seizures.172,173 It is
METHANOL known to cause visual field defects at therapeutic doses.174–176
The peripheral retina can be atrophic and the optic disk can
exhibit nasal or diffuse pallor.177,178 The visual loss can be
GENERAL INFORMATION transient or permanent. Toxicity can occur within the first few
Methanol toxicity is commonly due to accidental poisoning in years of therapy. Although a dose dependent toxicity was found
alcoholics. Improper distillation allows for methanol formation in one study, no dose response or risk factors for retinopathy
as well as ethanol. Inhalation and skin contact can also induce were found in another.179,180
2220
Retinal Toxicity of Systemic Medications

Visual field changes can be asymptomatic, and, in children,


accurate peripheral visual testing can be dfficult.181 ERG, EOG,
LAMOTRIGINE
and VEP changes correlate with the visual field changes, Lamotrigine is another antileptic with documented changes
however, and can be used for routine evaluation.182–184 Reduc- in electrophysiology.185 No clear cases of toxicity have been
tions in the cone b-wave and oscillatory potentials are well identified, but one patient on high-dose lamotrigine therapy had
documented. transient visual field changes.

REFERENCES
1. Meredith TA, Aaberg TM, Willerson WD: 20. Mackenzie AH: Dose refinements in long- 38. Almony A, Garg S, Peters RK, et al:
Progressive chorioretinopathy after receiving term therapy of rheumatoid arthritis with Threshold Amsler grid as a screening tool
thioridazine. Arch Ophthalmol 1978; 96:1172. antimalarials. Am J Med 1983; 75:40. for asymptomatic patients on
2. Hamilton JD: Thioridazine retinopathy 21. Ochsendorf FR, Runne U: Chloroquine: Hydroxychloroquine therapy. Br J
within the upper dosage limit. consideration of maximum daily dose Ophthalmol 2005; 89:569.
Psychosomatics 1985; 26:823. (3.5 mg/kg ideal weight) prevents 39. Pluenneke AC, Blomquist PH: Utility of red
3. Weekley RD, Potts AM, Reboton J, et al: retinopathy. Dermatology 1996; 192:382. amsler grid screening in a rheumatology
Pigmentary retinopathy in patients receiving 22. Mackenzie AH, Scherbel AL: A decade of clinic. J Rheumatol 2004; 31:1754.
high doses of a new phenothiazine. Arch chloroquine maintenance therapy: rate of 40. Hart WM Jr, Burde RM, Johnston GP,

CHAPTER 176
Ophthalmol 1960; 64:65. administration governs incidence of Drews RC: Static perimetry in chloroquine
4. Connell MM, Poley BJ, McFarlane JR: retinotoxicity. Arthritis Rheum 1968; 11:496. retinopathy. Perifoveal patterns of visual
Chorioretinopathy associated with 23. Weiner A, Sandberg MA, Gaudio AR, et al: field depression. Arch Ophthalmol 1984;
thioridazine therapy. Arch Ophthalmol Hydroxychloroquine retinopathy. Am J 102:377.
1964; 71:816. Ophthalmol 1991; 112:528. 41. Easterbrook M, Trope G: Value of
5. Siddall, JR: The ocular toxic findings with 24. Coyle JT: Hydroxychloroquine retinopathy. Humphrey perimetry in the detection of
prolonged and high dosage chlopromazine Ophthalmology 2001; 108:243. early chloroquine retinopathy. Lens Eye
intake. Arch Ophthalmol 1965; 74:460. 25. Levy GD, Munz SJ, Paschal J, et al: Toxic Res 1989; 6:255.
6. Marmor MF: Is thioridazine retinopathy Incidence of hydroxychloroquine 42. Arden GB, Friedmann A, Kolb H:
progressive? Relationship of pigmentary retinopathy in 1207 patients in a large Anticipation of chloroquine retinopathy.
changes to visual function. Br J multicenter outpatient practice. Arthritis Lancet 1962; 1:1164.
Ophthalmol 1990; 74:739. Rheum 1997; 40:1482. 43. Kolb H: Electro-oculogram findings in
7. Miyata M, Imai H, Ishikawa S, et al: 26. Mavrikakis M, Papazoglou S, Sfikakis PP, patients treated with antimalarial drugs.
Changes in human electroretinography et al: Retinal toxicity in long-term Br J Ophthalmol 1965; 49:573.
associated with thioridazine administration. hydroxychloroquine treatment. Ann Rheum 44. Reijmer CN, Tijssen JG, Kok GA,
Ophthalmologica 1980; 181:175. Dis 1996; 55:187. van Lith GH: Interpretation of the electro-
8. Goar EL, Fletcher MC: Toxic 27. Mavrikakis I, Sfikakis PP, Mavrikakis E, et al: oculogram of patients taking chloroquine.
chorioretinopathy following the use of The incidence of irreversible retinal toxicity Doc Ophthalmol 1980; 48:273.
NP 207. Trans Am Ophthalmol Soc 1956; in patients treated with hydroxychloroquine: 45. Neubauer AS, Samari-Kermani K, Schaller U,
54:129. a reappraisal. Ophthalmology 2003; et al: Detecting chloroquine retinopathy:
9. Miller, FS III, Bunt-Millam AH, Kalina RE: 110:1321. electro-oculogram versus colour vision.
Clinical-ultrastructural study of thioridazine 28. Johnson MW, Vine AK: Hydroxychloroquine Br J Ophthalmol 2003; 87:902.
retinopathy. Ophthalmology 1982; 89:1478 therapy in massive total doses without 46. So SC, Hedges TR, Schuman JS,
10. Potts AM: The concentration of retinal toxicity. Am J Ophthalmol 1987; Quireza ML: Evaluation of
phenothiazines in the eye of experimental 104:139. hydroxychloroquine retinopathy with
animals. Invest Ophthalmol 1962; 1:522. 29. Bernstein HN: Ophthalmic considerations multifocal electroretinography. Ophthalmic
11. Potts AM: Further studies concerning the and testing in patients receiving long-term Surg Lasers Imaging 2003; 34:251.
accumulation of polycyclic compounds on antimalarial therapy. Am J Med 1983; 47. Lai TY, Chan WM, Li H, et al: Multifocal
uveal melanin. Invest Ophthalmol 1964; 75:25. electroretinographic changes in patients
3:399. 30. Henkind P, Carr RE, Siegel IM: Early receiving hydroxychloroquine therapy. Am J
12. Potts AM: The reaction of uveal pigment in chloroquine retinopathy: clinical and Ophthalmol 2005; 140:794.
vitro with polycyclic compounds. Invest functional findings. Arch Ophthalmol 1964; 48. Teoh SC, Lim J, Koh A, et al: Abnormalities
Ophthalmol 1964; 3:405. 71:157. on the multifocal electroretinogram may
13. Fornaro P, Calabria G, Corallo G, Picotti GB: 31. Easterbrook M: Chloroquine retinopathy. precede clinical signs of
Pathogenesis of degenerative retinopathies Arch. Ophthalmol 1991; 109:1362. hydroxychloroquine retino-toxicity. Eye
induced by thioridazine and other 32. Michael F M, Carr RE, Easterbrook M, et al: 2006; 20:129.
antipsychotics: a dopamine hypothesis. Mieler and American Academy of 49. Maturi RK, Yu M, Weleber RG: Multifocal
Doc Ophthalmol 2002; 105:41. Ophthalmology: recommendations on electroretinographic evaluation of long-term
14. DeLong SL, Poley BJ, McFarlane JR: screening for chloroquine and hydroxychloroquine users. Arch
Ocular changes associated with long-term hydroxychloroquine retinopathy: a report by Ophthalmol 2004; 122:973.
chlorpromazine therapy. Arch Ophthalmol the American Academy of Ophthalmology. 50. Bernstein H, Zvaifler N, Rubin M, et al:
1965; 73:611. Ophthalmology 2002; 109:1377. The ocular deposition of chloroquine.
15. Siddal JR: The ocular toxic findings with 33. Easterbrook M: Ocular effects and safety of Invest Ophthalmol 1963; 2:384.
prolonged and high dosage chlorpromazine antimalarial agents. Am J Med 1988; 85:23. 51. Mahon GJ, Anderson HR, Gardiner TA, et al:
intake. Arch Ophthalmol 1965; 74:460. 34. Carr RE, Henkind P, Rothfield N, Siegel IM: Chloroquine causes lysosomal dysfunction
16. Oshika T: Ocular adverse effects of Ocular toxicity of antimalarial drugs. Long- in neural retina and RPE: implications for
neuropsychiatric agents: incidence and term follow-up. Am J Ophthalmol 1968; retinopathy. Curr Eye Res. 2004; 28:277.
management. Drug Safety 1995; 12:256. 66:738. 52. Sundelin SP, Terman A: Different effects of
17. Zelickson AS, Zeller HC: A new and 35. Vu BL, Easterbrook M, Hovis JK: Detection chloroquine and hydroxychloroquine on
unusual reaction to chlorpromazine. JAMA of color vision defects in chloroquine lysosomal function in cultured retinal
1964; 188:394. retinopathy. Ophthalmology 1999; 106:1799. pigment epithelial cells. APMIS 2002;
18. Cambiaggi A: Unusual ocular lesions in a 36. Easterbrook M: The use of Amsler grids in 110:481.
case of systemic lupus erythematosus. early chloroquine retinopathy. 53. Duncker G, Schmiederer M, Bredehorn T:
Arch Ophthalmol 1957; 57:451. Ophthalmology 1984; 91:1368. Chloroquine-induced lipidosis in the rat
19. Tobin DR, Krohel GB, Rynes RL: 37. Easterbrook M: The sensitivity of Amsler retina: a functional and morphological
Hydroxychloroquine: seven-year grid testing in early chloroquine retinopathy. study. Ophthalmologica 1995;
experience. Arch Ophthalmol 1982; 100:81. Trans Ophthalmol Soc UK 1985; 104:204. 209:79. 2221
RETINA AND VITREOUS

54. Duncker G, Bredehorn T: Chloroquine- 77. Boudreault G, Cortin P, Corriveau LA, et al: 95. Zak TA, Buncic R: Primary hereditary
induced lipidosis in the rat retina: functional La rétinopathie à la canthaxanthine. I. oxalosis retinopathy. Arch Ophthalmol
and morphological changes after Etude clinique de 51 consommateurs. 1983; 101:78.
withdrawal of the drug. Graefes Arch Clin Can J Ophthalmol 1983; 18:325 96. Wilding G, Caruso R, Lawrence TS, et al:
Exp Ophthalmol 1996; 234:378. 78. Cortin P, Boudreault G, Rousseau AP, et al: Retinal toxicity after high-dose cisplatin
55. Atlee WE: Talc and cornstarch emboli in La rétinopathie à la canthaxanthine. 2. therapy. J Clin Oncol 1985; 3:1683.
eyes of drug users. JAMA 1972; 219:49. Facteurs prédisposants. Can J Ophthalmol 97. Miller DF, Bay JW, Lederman RJ, et al:
56. Tse DT, Ober RR: Talc retinopathy. Am J 1984; 19:215. Ocular and orbital toxicity following
Ophthalmol 1980; 90:624. 79. Metge P, Mandirac-Bonnefoy C, Bellaube P: intracarotid injection of BCNU (carmustine)
57. Schatz H, Drake M: Self-injected retinal Thésaurismose rétinienne à la and cisplatinum for malignant gliomas.
emboli. Ophthalmology 1979; 86:468. canthaxanthine. Bull Mem Soc Fr Ophthalmology 1985; 92:402.
58. Brucker AJ: Disk and peripheral retinal Ophtalmol 1984; 95:547. 98. Kupersmith MJ, Seiple WH, Holopigian K,
neovascularization secondary to talc and 80. Ros AM, Leyon H, Wennersten G: et al: Maculopathy caused by intra-
cornstarch emboli. Am J Ophthalmol 1979; Crystalline retinopathy in patients taking an arterially administered cisplatin and
88:864. oral drug containing canthaxanthine. intravenously administered carmustine.
59. Friberg TR, Gragoudas ES, Regan CDJ: Photodermatol 1985; 2:183. Am J Ophthalmol 1992; 113:435.
Talc emboli and macular ischemia in 81. Lonn LI: Canthaxanthin retinopathy. Arch 99. Khawly JA, Rubin P, Petros W, et al:
intravenous drug abuse. Arch Ophthalmol Ophthalmol 1987; 105:1590. Retinopathy and optic neuropathy in bone
1979; 97:1089. 82. Harnois C, Samson J, Malenfant M, et al: marrow transplantation for breast cancer.
60. Sharma MC, Ho AC: Macular fibrosis Canthaxanthine retinopathy: anatomic and Ophthalmology 1996; 103:87.
associated with talc retinopathy. Am J functional reversibility. Arch Ophthalmol 100. Katz BJ, Ward JH, Digre KB, et al:
SECTION 10

Ophthalmol 1999; 128:517. 1989; 107:538. Persistent severe visual and


61. Jampol LM, Setogawa T, Rednam KRV, et al: 83. Harnois C, Cortin P, Samson J, et al: electroretinographic abnormalities after
Talc retinopathy in primates: a model of Static perimetry in canthaxanthin intravenous cisplatin therapy.
ischemic retinopathy. I. Clinical studies. maculopathy. Arch Ophthalmol 1988; J Neuroophthalmol 2003; 23:132.
Arch Ophthalmol 1981; 99:1273. 106:58. 101. Shingleton BJ, Bienfang DC, Albert DM,
62. Kaga N, Tso MOM, Jampol LM, et al: Talc 84. Goralczyk R, Barker FM, Buser S, et al: et al: Ocular toxicity associated with high-
retinopathy in primates: a model of Dose dependency of canthaxanthin dose carmustine. Arch Ophthalmol 1982;
ischemic retinopathy. II. A histopathologic crystals in monkey retina and spatial 100:1766.
study. Arch Ophthalmol 1982; 100:1644. distribution of its metabolites. Invest 102. Hayasaka S, Nagaki Y, Matsumoto M,
63. Kaga N, Tso MOM, Jampol LM: Talc Ophthalmol Vis Sci 2000; 41:1513. Sato S: Interferon associated retinopathy.
retinopathy in primates: a model of 85. Weber U, Kern W, Novotny GE, et al: Br J Ophthalmol 1998; 82:323.
ischemic retinopathy. III. An electron Experimental carotenoid retinopathy. I. 103. Guyer DR, Tiedeman J, Yannuzzi LA, et al:
microscopic study. Arch Ophthalmol 1982; Functional and morphological alterations of Interferon-associated retinopathy. Arch
100:1649. the rabbit retina after 11 months dietary Ophthalmol 1993; 111:350.
64. Hanscom TA: Indirect treatment of carotenoid application. Graefes Arch Clin 104. Kawano T, Shegehira M, Uto H, et al:
peripheral retinal neovascularization. Am J Exp Ophthalmol 1987; 225:198. Retinal complications during interferon
Ophthalmol 1982; 93:88. 86. Weber U, Michaelis L, Kern W, Goerz G: therapy for chronic hepatitis C. Am J
65. Pavlidis NA, Petris C, Briassoulis E, et al: Experimental carotenoid retinopathy. II. Gastroenterol 1996; 91:309.
Clear evidence that long-term, low-dose Functional and morphological alterations 105. Schulman JA, Liang C, Kooragayala LM,
tamoxifen treatment can induce ocular of the rabbit retina after acute et al: Posterior segment complications in
toxicity. A prospective study of 63 patients. canthaxanthin application with small patients with hepatitis C treated with
Cancer 1992; 69:2961. unilamellar phospholipid liposomes. interferon and ribavirin. Ophthalmology
66. Chang T, Gonder JR, Ventresca MR: Low- Graefes Arch Clin Exp Ophthalmol 1987; 2003; 110:437.
dose tamoxifen retinopathy. Can J 225:346. 106. Esmaeli B, Koller C, Papadopoulos N,
Ophthalmol 1992; 27:148. 87. Arden GB, Oluwole JO, Polkinghorne P, Romaguera J: Interferon-induced
67. Noureddin BN, Seoud M, Bashshur Z, et al: et al: Monitoring of patients taking retinopathy in asymptomatic cancer
Ocular toxicity in low-dose tamoxifen: a canthaxanthin and carotene: an patients. Ophthalmology 2001;
prospective study. Eye 1999; 13:729. electroretinographic and ophthalmological 108:858.
68. Kaiser-Kupfer MI, Lippman ME: Tamoxifen survey. Hum Toxicol 1989; 8:439. 107. Hejny C, Sternberg P, Lawson DH, et al:
retinopathy. Cancer Treat Rep 1978; 88. Daicker B, Schiedt K, Adnet JJ, Retinopathy associated with high-dose
62:315. Bermond P: Canthaxanthin retinopathy. interferon alfa-2b therapy. Am J
69. Kaiser-Kupfer MI, Kupfer C, Rodrigues MM: An investigation by light and electron Ophthalmol 2001; 131:782.
Tamoxifen retinopathy. A clinicopathologic microscopy and physicochemical analysis. 108. Tokai R, Ikeda T, Miyaura T, et al:
report. Ophthalmology 1981; 88:89. Graefes Arch Clin Exp Ophthalmol 1987; Interferon-associated retinopathy and
70. Heier JS, Dragoo RA, Enzenauer RW, et al: 225:189. cystoid macular edema. Arch Ophthalmol
Screening for ocular toxicity in 89. Bullock JD, Albert DM: Fleck retina: 2001; 119:1077.
asymptomatic patients treated with appearance secondary to oxalate crystals 109. Kiratli H, Irkee M: Presumed interferon-
tamoxifen. Am J Ophthalmol 1994; from methoxyflurane anesthesia. Arch associated bilateral macular arterial branch
117:772. Ophthalmol 1975; 93:26. obstruction. Eye 2000; 14:920.
71. Noureddin BN, Seoud M, Bashshur Z, et al: 90. Novak MA, Roth AS, Levine MR: Calcium 110. Jain K, Lam WC, Waheeb S, et al:
Ocular toxicity in low-dose tamoxifen: a oxalate retinopathy associated with Retinopathy in chronic hepatitis C patients
prospective study. Eye 1999; 13:729. methoxyflurane abuse. Retina 1988; 8:230. during interferon treatment with ribavirin.
72. Ashford AR, Donev I, Tiwari RP, et al: 91. Albert DM, Bullock JD, Lahav M, et al: Br J Ophthalmol 2001 85:1171.
Reversible ocular toxicity related to Flecked retina secondary to oxalate 111. Kertes PJ, Britton WA, Addison DJ, et al:
tamoxifen therapy. Cancer 1988; 61:33. crystals from methoxyflurane anesthesia: Toxicity of intravitreal interferon alpha-2b
73. McKeown CA, Swartz M, Blom J, et al: clinical and experimental studies. Trans Am in the rabbit. Can J Ophthalmol 1995;
Tamoxifen retinopathy. Br J Ophthalmol Acad Ophthalmol Otolaryngol 1975; 79:817. 30:355.
1981; 65:177. 92. Wells CG, Johnson RJ, Qingli L, et al: 112. Perry CM, Balfour JA: Didanosine. An
74. Martine MF, Joel G, Maddalena QE: Optical Retinal oxalosis: a clinicopathological update on its antiviral activity,
coherence tomography in tamoxifen report. Arch Ophthalmol 1989; 107:1638. pharmacokinetic properties and therapeutic
retinopathy. Breast Cancer Res Treat 2006; 93. Meredith TA, Wright JD, Gammon JA, et al: efficacy in the management of HIV disease.
99:117. Ocular involvement in primary Drugs 1996; 52:928.
75. Sharkey JA: Idiopathic canthaxanthine hyperoxaluria. Arch Ophthalmol 1984; 113. Whitcup SM, Butler KM, Caruso R, et al:
retinopathy. Eur J Ophthalmol 1993; 3:226. 102:584. Retinal toxicity in human immunodeficiency
76. Cortin P, Corriveau LA, Rooseau AP, et al: 94. Fielder AR, Garner A, Chambers TL: virus-infected children treated with 2„,3„-
Maculopathie en pailletes d’or. Can J Ophthalmic manifestations of primary dideoxyinosine. Am J Ophthalmol 1992;
2222 Ophthalmol. 1982; 17:103–106. oxalosis. Br J Ophthalmol 1980; 64:782. 113:1.
Retinal Toxicity of Systemic Medications

114. Whitcup SM, Butler KM, Pizzo PA, 137. Forster DJ, Causey DM, Rao NA: Bull’s eye retinopathy using the albino rat.
Nussenblatt RB: Retinal lesions in children retinopathy and clofazimine. Ann Intern Ophthalmologica 1990; 201:32.
treated with dideoxyinosine. N Engl J Med Med 1992; 116:876. 157. Gehlbach PL, Purple RL, Hallaway PE,
1992; 326:1226. 138. Greenberg PL: Myelodysplastic syndromes: Hedlund BE: Polymer conjugation reduces
115. Whitcup SM, Dastgheib K, Nussenblatt RB, iron overload consequences and current deferoxamine induced retinopathy in an
et al: A clinicopathologic report of the chelating therapies. J Natl Compr Canc albino rat model. Invest Ophthalmol Vis Sci
retinal lesions associated with didanosine. Netw 2006; 4:91. 1993; 34:2871.
Arch Ophthalmol 1994; 112:1594. 139. Kattamis A, Ladis V, Berdousi H, et al: Iron 158. Megarbane B, Borron SW, Trout H, et al:
116. Cobo J, Ruiz MF, Figueroa MS, et al: chelation treatment with combined Treatment of acute methanol poisoning
Retinal toxicity associated with therapy with deferiprone and with fomepizole. Intensive Care Med 2001;
didanosine in HIV-infected adults. AIDS deferioxamine: a 12-month trial. Blood 27:1370.
1996; 10:1297. Cells Mol Dis 2006; 36:21. 159. Essama Mbia JJ, Guerit JM, Haufroid V,
117. Parsons WB Jr, Flinn JH: Reduction in 140. Pippard MJ: Iron metabolism and iron Hantson P: Fomepizole therapy for reversal
elevated blood cholesterol levels by large chelation in the thalassaemia disorders. of visual impairment after methanol
doses of nicotinic acid: preliminary report. Haematologica 1990; 75(Suppl 5):66. poisoning: a case documented by visual
JAMA 1957; 165:234. 141. Silliman CC, Peterson VM, Mellman DL, evoked potentials investigation. Am J
118. Carlson LA: Nicotinic acid: the broad- et al: Iron chelation by deferoxamine in Ophthalmol 2002; 134:914.
spectrum lipid drug. A 50th anniversary sickle cell patients with severe transfusion- 160. Krolman GM, Pidde WJ: Acute methyl
review. J Intern Med 2005; 258:94. induced hemosiderosis: a randomized, alchohol poisoning. Can J Ophthalmol
119. Capuzzi DM, Guyton JR, Morgan JM, et al: double-blind study of the dose-response 1968; 3:270.

CHAPTER 176
Efficacy and safety of an extended-release relationship. J Lab Clin Med 1993; 122:48. 161. Ingemansson SO: Clinical observations on
niacin (Niaspan): a long-term study. Am J 142. Simon P, Ang KS, Cam G, et al: ten cases of methanol poisoning. Acta
Cardiol 1998 82:74U. Desferrioxamine, aluminium, and dialysis. Ophthalmol 1984; 62:15.
120. Gass JDM: Nicotinic acid maculopathy. Lancet 1983; 2:1489. 162. Murray TG, Burton TC, Rajani C, et al:
Am J Ophthalmol 1973; 76:500. 143. Yokel RA, Ackrill P, Burgess E, et al: Methanol poisoning. A rodent model with
121. Millay RH, Klein ML, Illingworth DR: Prevention and treatment of aluminum structural and functional evidence for
Niacin maculopathy. Ophthalmology 1988; toxicity including chelation therapy: status retinal involvement. Arch Ophthalmol 1991;
95:930. and research needs. J Toxicol Environ 109:1012.
122. Jampol LM: Niacin maculopathy. Health 1996; 48:667. 163. Treichel JL, Murray TG, Lewandowski MF,
Ophthalmology 1988; 95:1704. 144. Haimovici R, D’Amico DJ, Gragoudas ES, et al: Retinal toxicity in methanol poisoning.
123. Callanan D, Blodi BA, Martin DF: Macular Sokol S: The expanded clinical spectrum of Retina 2004; 24:309.
edema associated with nicotinic acid deferoxamine retinopathy. Ophthal 2002; 164. Horgan SE, Williams RW: Chronic retinal
(niacin). JAMA 1998; 279:1702. 109:164. toxicity due to quinine in Indian tonic water.
124. Fraunfelder FW: Ocular side effects from 145. Lakhanpal V, Schocket SS, Jiji R: Eye 1995; 9:637.
herbal medicines and nutritional Deferoxamine (Desferal)-induced toxic 165. Bacon P, Spalton DJ, Smith SE: Blindness
supplements. Am J Ophthalmol 2004; retinal pigmentary degeneration and from quinine toxicity. Br J Ophthalmol
138:639. presumed optic neuropathy. 1988; 72:219.
125. Spirn MJ, Warren FA, Guyer DR, et al: Ophthalmology 1984; 91:443. 166. Canning CR, Hague S: Ocular quinine
Optical coherence tomography findings in 146. Olivieri NF, Buncic JR, Chew E, et al: Visual toxicity. Br J Ophthalmol 1988; 72:23.
nicotinic acid maculopathy. Am J and auditory neurotoxicity in patients 167. Brinton GS, Norton EW, Zahn JR,
Ophthalmol 2003; 135:913. receiving subcutaneous deferoxamine Knighton RW: Ocular quinine toxicity. Am J
126. Reddy VM, O’Sullivan JF, Gangadharam infusions. N Engl J Med 1986; 314:869. Ophthalmol 1980; 90:403.
PR: Antimycobacterial activities of 147. Orton RB, de Veber LL, Sulh HM: Ocular 168. Wolf LR, Otten EJ, Spadafora MP:
riminophenazines. J Antimicrob Chemother and auditory toxicity of long-term, high- Cinchonism: two case reports and review
1999; 43:615. dose subcutaneous deferoxamine therapy. of acute quinine toxicity and treatment.
127. Biswas SK: Chemotherapy of leprosy. Can J Ophthalmol 1985; 20:153. J Emerg Med 1992; 295.
J Indian Med Assoc 2004; 102:695. 148. Rubinstein M, Dupont P, Doppee JP, et al: 169. Buchanan TAS, Lyness RW, Collins AD,
128. Tomioka H: Present status and future Ocular toxicity of desferrioxamine. Lancet et al: An experimental study of quinine
prospects of chemotherapeutics for 1985; 1:817. blindness. Eye 1987; 1:522.
intractable infections due to 149. Gass JDM: Stereoscopic atlas of macular 170. Lochhead J, Movaffaghy A, Falsini B, et al:
Mycobacterium avium complex. Curr Drug diseases: diagnosis and treatment. 4th edn. The effect of quinine on the electroretino-
Discov Technol 2004; 1:255. Mosby, St Louis; 1997. grams of children with pediatric cerebral
129. Bezerra EL, Vilar MJ, da Trindade Neto PB, 150. Gonzales CR, Lin AP, Engstrom RE, malaria. J Infect Dis 2003; 187:1342.
Sato EI: Double-blind, randomized, Kreiger AE: Bilateral vitelliform maculopathy 171. Barrett NA, Solano T: Quinine ocular
controlled clinical trial of clofazimine and deferoxamine toxicity. Retina 2004; toxicity: treatment of blindness using
compared with chloroquine in patients with 24:464. therapy for vasospasm. Anaesth Intensive
systemic lupus erythematosus. Arthritis 151. Blake DR, Winyard P, Lunec J, et al: Care 2002; 30:234.
Rheum 2005; 52:3073. Cerebral and ocular toxicity induced by 172. Uldall P, Alving J, Gram L, Hogenhaven H:
130. Schraufnagel DE: Tuberculosis treatment desferrioxamine. Q J Med 1985; 56:345. Vigabatrin in childhood epilepsy: a 5-year
for the beginning of the next century. Int J 152. Arden GB, Wonke B, Kennedy C, follow-up study. Neuropediatrics 1995;
Tuberc Lung Dis 1999; 3:651. Huehns ER: Ocular changes in patients 26:253.
131. Kaur I, Ram J, Kumar B, et al: Effect of undergoing long-term desferrioxamine 173. The Canadian Vigabatrin Study Group,
clofazimine on eye in multibacillary leprosy. treatment. Br J Ophthalmol 1984; 68:873. Guberman A, Bruni J: Long-term open
Indian J Lepr 1990; 62:87. 153. Hidajat RR, McLay JL, Goode DH, multicentre, add-on trial of vigabatrin in
132. Walinder PE, Gip L, Stempa M: Corneal Spearing RL: EOG as a monitor of adult resistant partial epilepsy. Seizure
changes in patients treated with desferrioxamine retinal toxicity. Doc 2000; 9:112.
clofazimine. Br J Ophthalmol 1976; 60:526. Ophthalmol 2004; 109:273. 174. Eke T, Talbot JF, Lawden MC: Severe
133. Ohman L, Wahlberg I: Letter: ocular side- 154. Rahi AH, Hungerford JL, Ahmed AI: Ocular persistent visual field constriction
effects of clofazimine. Lancet 1975; 2:933. toxicity of desferrioxamine: light associated with vigabatrin. BMJ 1997;
134. Yawalkar SJ, Vischer W: Lamprene microscopic histochemical and 314:180.
(clofazimine) in leprosy. Basic information. ultrastructural findings. Br J Ophthalmol 175. Lawden MC, Eke T, Degg C, et al: Visual
Lepr Rev 1979; 50:135. 1986; 70:373. field defects associated with vigabatrin
135. Craythorn JM, Swartz M, Creel DJ: 155. Pall H, Blake DR, Winyard P, et al: Ocular therapy. J Neurol Neurosurg Psychiatry
Clofazimine-induced bull’s eye retinopathy. toxicity of desferrioxamine – an example of 1999; 67:716.
Retina 1986; 6:50. copper promoted auto-oxidative damage? 176. Wild JM, Martinez C, Reinshagen G,
136. Cunningham CA, Friedberg DW, Carr RG: Br J Ophthalmol 1989; 73:42. Harding GF: Characteristics of a unique
Clofazimine-induced generalized retinal 156. Good PA, Claxson A, Morris CJ, Blake DR: visual field defect attributed to vigabatrin.
degeneration. Retina 1990; 10:131. A model for desferrioxamine-induced Epilepsia 1999; 40:1784. 2223
RETINA AND VITREOUS

177. Buncic JR, Westall CA, Panton CM, et al: lack of correlation with dose. Epilepsia electrophysiology and visual fields in
Characteristic retinal atrophy with 2006; 47:311. epileptics: a controlled study with a
secondary ‘inverse’ optic atrophy identifies 181. Daneshvar H, Racette L, Coupland SG, discussion of possible mechanisms. Doc
vigabatrin toxicity in children. et al: Symptomatic and asymptomatic Ophthalmol 2002; 104:195.
Ophthalmology 2004; 111:1935. visual loss in patients taking vigabatrin. 184. Harding GF, Spencer EL, Wild JM,
178. Russell-Eggitt IM, Mackey DA, Taylor DS, Ophthalmology 1999; 106:1792. et al: Field-specific visual-evoked
et al: Vigabatrin-associated visual field 182. Besch D, Kurtenbach A, Apfelstedt-Sylla E, potentials. Identifying field defects in
defects in children. Eye 2000; 14:334. et al: Visual field constriction and vigabatrin-treated children. Neurology
179. Malmgren K, Ben-Menachem E, Frisen L: electrophysiological changes associated 2002; 58:1261.
Vigabatrin visual toxicity: evolution and with vigabatrin. Doc Ophthalmol 2002; 185. Arndt CF, Husson J, Derambure P, et al:
dose dependence. Epilepsia 2001; 42:609. 104:151. Retinal electrophysiological results in
180. Kinirons P, Cavalleri GL, O’Rourke D, et al: 183. Comaish IF, Gorman C, Brimlow GM, et al: patients receiving lamotrigine monotherapy.
Vigabatrin retinopathy in an Irish cohort: The effects of vigabatrin on Epilepsia 2005; 46:1055.
SECTION 10

2224
CHAPTER

177 Retinitis Pigmentosa and Allied Diseases


Eliot L. Berson

Retinitis pigmentosa (RP) is the name applied to a group of function in the area of the scotoma. When a patient reports
hereditary retinal degenerations that affect ~1 in 4000 people ‘night blindness’ in the era of the electric light bulb, most often
worldwide.1–6 In the state of Maine in the United States, genetic this symptom is related to impaired cone function. Because the
types by families were found to be 19% autosomal dominant, cones are used most of the time, patients can lose all their rod
19% autosomal recessive, 8% X-chromosome-linked, 8% function and not be aware of visual loss unless they are trying
undetermined, and 46% isolates with only one affected member to see under starlight or moonlight conditions.11
in a given family.5 In England, the X-linked type has been The rapid expansion of knowledge about RP and allied night-
reported to account for 16% of families and the autosomal blinding diseases precludes a comprehensive review in a single
recessive type for 7%.4 The condition can rarely be inherited by chapter. This chapter provides a framework for evaluating
a digenic or mitochondrial mode of transmission and uni- patients with these diseases (Table 177.1), information on some
parental isodisomy has also been reported.7–10 Taking into of the genes that, when mutated, cause these diseases, and a
account that most isolate cases are inherited by an autosomal summary of treatments for some forms.
recessive mode but some are X-linked and that most dominant
and X-linked families have two or more affected members, one SYMPTOMS AND SIGNS
can estimate that 30–40% of cases are inherited as an auto-
somal dominant trait, 50–60% as an autosomal recessive trait,
and ~10–15% as an X-linked trait in North America. Key Features: Retinitis Pigmentosa – Clinical Findings
To understand the functional abnormalities that occur in • Estimated prevalence – 1 in 4000 worldwide.
these diseases, it is first important to know that the normal • Modes of transmission – dominant, recessive, X-linked,
human retina has ~120 million photoreceptors; ~93% are rods digenic, mitochondrial, uniparental isodisomy.
and 7% are cones. Although cone density is highest in the • Symptoms – abnormal adaptation, night blindness, loss of
central macula (Fig. 177.1), more than 90% of the cones are mid-peripheral and then far-peripheral visual field, tunnel
outside the central 10°. A patient with normal cone function vision, eventual loss of visual acuity.
and absent rod function has normal visual acuity and a full • Signs on ocular examination – posterior subcapsular cataracts,
visual field even with a small (I-4e) white test light in the vitreous cells, attenuated retinal vessels, bone spicule pigment
Goldmann perimeter. A patient with absent cone function and around the mid-periphery, waxy pallor of the optic discs,
normal rod function has reduced visual acuity with a 1.2° cystoid macular edema in some cases.
diameter central scotoma but still retains a full peripheral visual • Reduced and delayed full-field electroretinograms.
field with a I-4e white test light. A patient with RP with a mid-
peripheral scotoma has, by definition, lost both cone and rod

FIGURE 177.1. Distribution of rods and cones


in the normal human retina. Corresponding
perimetric angles from the fovea at 0° are
given.
After Osterberg: From Pirenne MH: Vision and the Eye.
London, Chapman & Hall, 1967.

2225
RETINA AND VITREOUS

Patients with RP characteristically develop night blindness and lose far-peripheral field, and eventually lose central vision often
difficulty with mid-peripheral visual field in adolescence; as their by age 60. Signs on ocular examination include narrowed retinal
condition progresses, they develop a tendency to blue blindness, vessels, depigmentation of the retinal pigment epithelium, intra-
retinal bone spicule pigmentation, waxy pallor of the optic
disks, and vitreous cells.12–20 Posterior subcapsular cataracts
develop in many cases,21 and some patients show cystoid macular
TABLE 177.1. Retinitis Pigmentosa and Some Allied Diseases edema.22,23 Refractive errors, including astigmatism and myopia,
are common.24,25 The characteristic bone spicule pigment is
Autosomal dominant forms of retinitis pigmentosa typically distributed around the mid-peripheral fundus (Fig.
Autosomal recessive forms of retinitis pigmentosa
Sex-linked (X-chromosome-linked) forms of retinitis pigmentosa 177.2a) in the zone where rods are normally in maximal con-
Isolated (simplex) forms of retinitis pigmentosa centration. Histopathologic studies of autopsy eyes have shown
Progressive cone–rod degeneration loss of photoreceptors as well as photoreceptors with shortened
Atypical retinitis pigmentosa including sector, pericentral, or absent outer segments.26–30
paravenous, and unilateral forms
Some syndromes or diseases of which retinitis pigmentosa is a
part ELECTRORETINOGRAMS
Bassen–Kornzweig syndrome (abetalipoproteinemia)
Refsum disease In 1945, Karpe discovered that patients with advanced RP had
Familial isolated vitamin E deficiency very small or nondetectable (<10 mV) ERGs.31 Subsequently, it
SECTION 10

Usher syndrome types I, II, and III was shown that patients with early RP could have subnormal
Laurence–Moon and Bardet–Biedl syndromes but easily detectable ERGs.32–35 Responses were not only reduced
Kearns–Sayre syndrome in amplitude but also delayed in b-wave implicit times,36–39 and
Hereditary cerebroretinal degenerations including Batten disease
Olivopontocerebellar atrophy
these ERG changes could be detected in some instances many
Cockayne syndrome years before diagnostic abnormalities were visible on fundus
Alström disease examination.20,37 Figure 177.3 illustrates representative full-field
Congenital amaurosis of Leber ERGs from a normal subject and four children ages 9–14 years
Choroideremia with early RP. Rod responses to dim blue light under dark-
Gyrate atrophy of the choroid and retina adapted conditions (left column) are reduced in all genetic types
Retinitis punctata albescens
Clumped pigmentary retinal degenerations (enhanced S-cone and when detectable are delayed in b-wave implicit times, as
syndrome in some cases) designated by horizontal arrows. Cone responses to 30-cps (i.e.,
Stationary forms of night blindness 30 Hz) white flickering light (right column) are normal or
reduced in amplitude and normal or delayed in b-wave implicit

a b c

FIGURE 177.2. Fundus


photographs of
moderately advanced
retinitis pigmentosa (a),
moderately advanced
choroideremia (b), gyrate
atrophy of the choroid
and retina (c), Oguchi
disease without dark
adaptation (d), and
fundus albipunctatus (e).

d e
2226
Retinitis Pigmentosa and Allied Diseases

CHAPTER 177
FIGURE 177.3. Electroretinogram (ERG) responses for a normal FIGURE 177.4. ERG responses of a normal subject and four patients
subject and four patients with retinitis pigmentosa (RP) (ages 13, 14, with sector or stationary retinal disease. Horizontal arrows (column 1)
14, and 9 years). Responses were obtained after 45 min of dark designate the range of normal rod b-wave implicit times, and the
adaptation to single flashes of blue light (left column) and white light vertical bar defining this range (mean ± SD) has been extended
(middle column). Responses (right column) were obtained to a 30-cps through responses of patients with sector retinitis pigmentosa (RP).
(or 30-Hz) white flickering light. Calibration symbol (lower right) Responses (middle column) from a patient with Oguchi’s disease are
signifies 50 msec horizontally and 100 mV vertically. Rod b-wave interrupted by reflex blinking, so the latter part cannot be illustrated.
implicit times in column 1 and cone implicit times in column 3 are Cone implicit times in column 3 are designated with arrows. SNB,
designated with arrows. Auto. Rec., autosomal recessive. a, peak of stationary night blindness.
cornea-negative a-wave; b, peak of cornea-positive b-wave. From Berson EL: Retinitis pigmentosa and allied diseases: Electrophysiologic
Modified from Berson EL: Retinitis pigmentosa and allied diseases: findings. Published courtesy of Trans Am Acad Ophthalmol Otolaryngol
Electrophysiologic findings. Published courtesy of Trans Am Acad Ophthalmol 81:OP659-OP666, 1976.
Otolaryngol 81:OP659-OP666, 1976.

times. In most cases, cone b-wave implicit times (displayed by Studies of patients with widespread progressive forms of RP
arrows in the right column) are so delayed that a phase shift with detectable rod ERGs and delayed cone b-wave implicit
occurs between the stimulus artifacts (designated by the vertical times have demonstrated that cone b-wave implicit time to
lines) and the corresponding response peaks. Each stimulus flash white flicker is inversely proportional to the log amplitude of
elicits the next-plus-one response in contrast to the normal. In the dark-adapted rod b-wave to blue light. In these patients,
the mixed cone–rod responses to single flashes of white light cone b-wave implicit time did not vary with the amplitudes of
under dark-adapted conditions (middle column), the cornea- the dark-adapted cone b-waves. A 10-fold reduction in rod
negative a-wave generated by the photoreceptors is reduced in amplitude was associated with ~5.5 msec slowing in cone
amplitude in all genetic types, pointing to the involvement of b-wave implicit times.39 These results would suggest that it is
the photoreceptors in these early stages.37 the loss of rod photoreceptors among remaining cones that
The subnormal responses with delayed b-wave implicit times apparently leads to abnormal rod–cone interaction,40 which can
seen in widespread progressive forms of RP contrast with the account in large part for the delays in cone b-wave implicit
subnormal responses with normal b-wave implicit times seen times seen in progressive forms of RP.
in self-limited sector RP (Fig. 177.4). For example, a father and The ERG can be used to identify not only which patients
son with dominantly inherited sector RP, separated in age by have widespread progressive forms of RP but also which
almost 30 years, have comparably reduced amplitudes and patients are normal. Relatives of patients with RP, age 6 years or
normal b-wave implicit times. These patients usually have an over, with normal rod and cone ERG amplitudes and implicit
area of intraretinal pigment confined to one or two quadrants in times have not been observed to develop this disease at a later
the periphery of each eye, with loss of peripheral rods and cones time.20,37,38,41
and consequent reductions in both rod and cone amplitudes.
Rod b-wave implicit times are within the normal range (desig- CARRIERS OF X-LINKED RP
nated by the vertical bars), and cone b-wave implicit times are
also within the normal range, as each stimulus elicits the suc- Female carriers of X-linked RP can show a patch of bone spicule
ceeding response, as seen in the normal. The ERGs recorded pigmentation in the periphery or an abnormal tapetal reflex in
from patients with sector RP are comparable to those recorded the macula; among carriers of childbearing age, less than half
from patients with large peripheral chorioretinal scars.36,38 show diagnostic abnormalities on fundus examination. ERG 2227
RETINA AND VITREOUS

testing can be used as an aid in detecting female carriers of of RP. For example, full-field ERGs have been recorded over a
X-linked RP.42,43 ERG testing of obligate carriers has shown that 20-year interval in young patients in a family with a dominant
more than 90% have abnormal responses that either are form. Amplitudes decreased over this period, and patients age
reduced in amplitude to single flashes of blue or white light 13 years or under with normal fundi and abnormal ERGs in
under dark-adapted conditions or are delayed in cone b-wave 1967 showed fundus changes of RP and further reductions in
implicit times, or both, in one or both eyes. A few older obligate their ERG amplitudes in 1977 (Fig. 177.6).44 Examination of
carriers with visual symptoms have had very small or even some members of this family showed further loss in 1987.38
nondetectable full-field ERGs (Fig. 177.5).42 Daughters of Signal averaging with a bipolar artifact reject buffer and
obligate carriers have had either normal ERGs or abnormal bandpass filtering has extended the range of detectability of
ERGs similar to those recorded from obligate carriers. responses from affected patients. ERGs can now be recorded
The abnormal ERGs of carriers of X-linked RP contrast with that are as low as 1 mV to 0.5-Hz flashes of white light with
the normal full-field ERG amplitudes and normal fundi observed signal averaging alone and, with bandpass filtering and signal
in obligate female carriers of autosomal recessive disease.42 averaging, as low as 0.05 mV to 30-Hz white flicker. Full-field
Once carrier females of X-linked RP have been detected by ERG function could be monitored with at least one test
ophthalmoscopy or full-field ERG testing, or both, they would criterion in 90% of an outpatient population with a visual-field
know that they have a 50% chance of having an affected son and diameter greater than 8°, thereby making this technique useful
a 50% chance of having a carrier daughter with each childbirth. in defining the natural course of the disease. ERGs in an adult
Female carriers of X-linked RP can have a slowly progressive male with X-linked RP are illustrated to show changes in ERG
SECTION 10

retinal degeneration, although the natural course remains to be function over a 2-year period (Fig. 177.7).20,45,46
defined. Some female carriers have had considerable loss of Among 94 patients, ages 6–49 years, with the common forms
visual field and substantial reductions in ERG amplitudes by of RP, full-field ERGs declined significantly over a 3-year
age 70 years. interval in 66 of 86 patients (77%) with detectable responses at
baseline. Patients lost on average 16% of remaining full-field
NATURAL COURSE OF RP ERG amplitude per year to single flashes of white light (95%
confidence limits 13.1–18.6%) and 18.5% of remaining ampli-
The ERG provides a quantitative measure of remaining retinal tude per year to 30-Hz white flicker (95% confidence limits
function and therefore can aid in defining the natural course 15.1–21.5%). Patients lost on average 5.2% of remaining foveal
cone ERG amplitude per year, indicating that loss of retinal
function was primarily extrafoveal in these patients. They lost
on average 4.6% of remaining visual-field area per year in the
Goldmann perimeter with a V-4e white test light, whereas
visual acuity and dark-adaptation thresholds remained rela-
tively stable. Bone spicule pigment increased in 54% of patients
for whom comparisons could be made over a 3-year interval,
suggesting that observation of increased pigmentation as a
means of following this condition was not as sensitive as full-
field ERG testing.45
Caution must be exercised in applying these population ERG
results to predict longitudinal patterns in individual patients,
since standard deviations derived from standard errors
indicated considerable variation around the mean for these
patients.45 However, these results, describing the natural course
on a quantitative basis, provide a frame of reference for
planning interventions in similar populations to stabilize or
slow the course of RP, particularly if monitored with full-field
ERG testing.

ABNORMAL ROD ERG DIURNAL RHYTHM


IN RP
The changes that occur in RP can be considered not only in
terms of the course over years but also in terms of abnormalities
in rod function that can occur over the course of a day.
Abnormal rod ERG diurnal rhythms have been observed in
light-entrained patients with dominant RP; these patients had
abnormal reductions in rod b-wave sensitivity 11⁄2 h and 8 h
after light onset (Fig. 177.8). The abnormal reductions in rod
b-wave sensitivity at 9:30 a.m. and 4:00 p.m. raised as one
FIGURE 177.5. ERG responses from a normal subject and four possibility that these patients with dominant RP shed
obligate carriers of sex-linked retinitis pigmentosa (RP). Pt (patient) 15, abnormally large fractions of rod outer segments throughout the
age 39 years; Pt 8, age 41; Pt 4, age 51; Pt 22, age 70. Stimulus
light period and that their rods were slow to renew their pre-
onset is designated by ‘vertical hatched lines’ for columns 1 and 2
and ‘vertical shock artifacts’ for column 3. Cornea-positivity is upward
light-onset outer segment length. Abnormal diurnal rhythms
deflection. Arrows in column 3 designate cone b-wave implicit times. have also been reported in patients with autosomal recessive
Reprinted from Am J Ophthalmol, vol 87, Berson EL, Rosen JB, Simonoff EA, and isolate forms of RP. The pathogenetic mechanism that
Electroretinographic testing as an aid in detection of carriers of X-chromosome- leads to these abnormal rod ERG diurnal rhythms remains to
2228 linked retinitis pigmentosa, pp 460–468, Copyright 1979, from Elsevier Science. be defined.47
Retinitis Pigmentosa and Allied Diseases

1967 1977

Blue-Green White White (30 cps) Blue-Green White White (30 cps)

Normal Normal

V1-92, age 6 V1-92, age 15

V1-90, age 11 V1-90, age 20

CHAPTER 177
V1-86, age 13 V1-86, age 22

V1-84, age 17 V1-84, age 26

a b

FIGURE 177.6. (a) ERGs recorded in 1967 for a normal subject and four affected members from a family with dominant retinitis pigmentosa
with reduced penetrance. One to three responses to the same stimulus are represented. Stimulus onset is designated by ‘vertical hatched lines’
in columns 1 and 2 and ‘vertical shock artifacts’ in column 3; cornea-positivity is upward deflection; arrows in column 3 designate cone implicit
times. Calibration symbol (lower right) designates 50 msec horizontally for columns 1 and 2 and 25 msec for column 3 and 50 mV vertically for
column 1 and 100 mV for columns 2 and 3. (b) ERGs recorded in 1977 for a normal subject and four affected members from the same family as
in a for comparison with the ERGs recorded in 1967. Calibration symbol (lower right) designates 50 msec horizontally and 100 mV vertically for all
tracings.
(a and b) From Berson EL, Simonoff EA: Dominant retinitis pigmentosa with reduced penetrance: Further studies of the electroretinogram. Arch Ophthalmol
97:1286–1291, 1979. Copyright 1979, American Medical Association.

MOLECULAR GENETIC STUDIES OF RP among 17 of 148 unrelated patients with autosomal dominant
RP from across North America (Fig. 177.9). The nucleotide base
Key Features: Retinitis Pigmentosa – Summary of change was a cytosine-to-adenine transversion (i.e., CCC to
Molecular Genetic Findings CAC) in codon 23, corresponding to a substitution of histidine
• Most frequent known causes – mutations in the rhodopsin, for proline in the twenty-third amino acid of rhodopsin. Only
USH2A, or RPGR gene. clinically affected relatives showed this gene defect in the
• Variable severity of disease at a given age among patients with families studied. These results, coupled with the fact that
the same gene defect. proline-23 is highly conserved among normal opsins, suggested
• Causative genes broadly classified – those affecting the that this point mutation affected a critical amino acid in
phototransduction cascade, the retinoid cycle, photoreceptor rhodopsin and that this mutation could be the cause of one
structure, or other aspects of photoreceptor and retinal form of autosomal dominant RP. This mutation was designated
pigment epithelial cell biology. as rhodopsin, proline-23-histidine (i.e., Pro23His).48
• Some 40–50% of cases have an unknown molecular genetic Subsequent studies have revealed more than 100 additional
basis. mutations in the rhodopsin gene.49–50 Only one mutation has
been found in a given family, and each mutation has segregated
perfectly with the disease in the families so far studied. All
More than 100 gene loci that cause RP have been mapped or patients with rhodopsin gene mutations so far tested have had
identified; a current list of genes responsible for this group of abnormal rod ERGs. None of these mutations has been found
diseases is maintained on the RetNet site at http://www.sph. in unrelated normal subjects.
uth.tmc.edu/Retnet/sum-dis.htm. Estimated proportions of The site of the mutation in the rhodopsin gene has impli-
cases of RP, excluding syndromic RP other than Usher syn- cations with respect to the clinical severity of the disease. A
drome, caused by identified genes are listed in Table 177.2. The group of 17 patients from separate families with Pro23His
most common forms of RP are due to either mutations in (mean age 37 years) retained a mean visual acuity of 20/26,
the rhodopsin gene, the Usher IIA gene, or the RPGR gene. The compared with 20/37 for the group of eight patients from
genes listed in this table account for ~50–60% of the cases of separate families with Pro347Leu (mean age 32 years). Visual-
RP in North America. field area was on average 3463 deg2 in the Pro23His group to a
The first gene abnormality found to cause RP was a single- V-4e white test light (normal >11 399 deg2) and 1224 deg2 in
base substitution in codon 23 of the rhodopsin gene found the Pro347Leu group. Mixed cone plus rod ERG responses to 2229
RETINA AND VITREOUS

TABLE 177–2. Estimated Proportions of Cases of Retinitis


Pigmentosa (RP) Caused by Identified Genes
(Excluding Syndromic RP Except for Usher Syndrome)
(Genes currently identified account for 50–60% of cases of RP)

Inheritance Pattern/Gene Percent of all RP (including


Usher Syndrome)

Autosomal Dominant (~40% of all cases of RP)


1. RHO (rhodopsin) (3q) 10%
2. RP1 (8q) 2.2%
3. PRPF31 (19q) 2.0%
4. PRPF3 (1q) 1.6%
5. peripherin/RDS (6p) ~1%
6. FSCN2 (17q) ~1%
7. PRPF8 (17p) 0.8%
8. IMPDH1 (7q) 0.8%
9. NRL (14q) 0.4%
FIGURE 177.7. Computer-averaged full-field ERGs from a 26-year-old 10. CRX (19q) 0.4%
man with X-linked retinitis pigmentosa obtained in response to 11. CA4 (carbonic anhydrase) ? frequency in USA (~2% in UK)
SECTION 10

10-msec flashes of white light at 16 000 ftL presented at 0.5 Hz (17q)


(n = 64), 30 Hz (n = 256), and 42 Hz (n = 256) at baseline (year 0) and 12. PAP1 (7p) <1%
at 2-year follow-up (year 2). Three consecutive response averages are 13. SEMA4A (semaphorin) (1q) <1%
superimposed in each case. ‘Vertical broken lines’ denote flash onset Autosomal Recessive (~50% of all cases of RP, including
for the 0.5-Hz condition and the onset of one of the trains of flashes isolates)
for the 30-Hz and 42-Hz conditions. 14. usherin (1q) ~10% (based on analysis of all
Reprinted from Am J Ophthalmol, vol 99, Berson EL, Sandberg MA, Rosner B, 72 exons in USH2A gene)
et al, Natural course of retinitis pigmentosa over a three-year interval, 15. PDE6B (4p) 2%
pp 240–251, Copyright 1985, from Elsevier Science. 16. PDE6A (5q) 1.5%
17. CNGA1 (4p) 1%
18. RPE65 (1p) 1%
19. myosin VIIa (11q) ~1% (Usher IB)
20. LRAT (4q) <1%
21. NR2E3 (15q) <1%
22. MERTK (2q) <1%
23. harmonin (11p) <1% (Usher IC)
24. CDH23 (10q) <1% (Usher ID)
25. PCDH15 (10q) <1% (Usher IF)
26. VLGRI (5q) <1% (Usher IIC)
27. clarin-1 (3q) <1% (Usher IIIA)
28. SAG (arrestin) (2q) ~0.5%
29. CRALBP (15q) ~0.5%
30. RHO (rhodopsin) (3q) ~0.5%
31. TULP1 (6q) ~0.5%
32. ABCA4 (i.e., ABCR) (1p) <1%
33. RGR (10q) <1%
34. CRB1 (1q) <1%
35. CERKL (2q) <1%
36. CNGB1 (16q) <1%
37. SANS (17q) <1% (Usher 1G)
39. RP1 (8q) <1%
X-linked (~10% of all cases of RP)
39. RPGR (Xp21.1) 8% (RPGR and RP2 combined
40. RP2 (Xp11.3) account for ~90% of XLRP)
Digenic (only a few families described to date)
41. ROM1 (11q) and <1%
peripherin/RDS (6p)
Mitochondrial (only one family, with Usher type III, described to
date)
42. MTTS2 <1%
Note: Percentages are approximate and are based on the breakdown of RP
cases according to genetic type reported by Fishman (Arch Ophthalmol
96:822–826, 1978.), Macrae (Birth Defects: Original Article Series 18:175–185,
1982), and Bunker et al. (Am J Ophthalmol 97:357–365, 1984) and on the
FIGURE 177.8. Mean log Ithreshold–1 of rod b-wave V = kl n function assumptions that all isolate cases are autosomal recessive and that about one
versus time of day for 5 light-entrained normal subjects and 5 light- third of cases of Usher syndrome type I (approximately 6% of all cases of RP)
entrained patients with dominant retinitis pigmentosa (RP). Subjects are caused by defects in myosin VIIa. The values are calculated based on
were dark-adapted for 1 h before 9:30 a.m., 4 p.m., and 9 p.m. test frequency of cases in a published series multiplied by the proportion of RP with
sessions. log Ithresholds–1 were based on criterion amplitudes averaging that inheritance pattern (e.g., dominant rhodopsin mutations were found in
90/363 cases of ADRP; ADRP accounts for about 40% of all cases of RP;
28.8 mV for normal subjects and 10.6 mV for patients with retinitis
accordingly, the percentage of cases caused by dominant rhodopsin mutations
pigmentosa. ‘Error bars’ designate ±SEM derived from pooled is 90/363 = 24.8% µ 0.4 = 9.92 ≈ 10%). Table prepared 4/15/06. A current
estimates of population variances obtained from analyses of variance. listing of genes causing retinitis pigmentosa and allied hereditary retinal
From Sandberg MA, Baruzzi CM, Hanson AH III, Berson EL: Rod ERG diurnal diseases can be accessed on the world-wide web
rhythm in some patients with dominant retinitis pigmentosa. Invest Ophthalmol at:http://www.sph.uth.tmc.edu/RetNet.
2230 Vis Sci 29:494, 1988. © Association for Research in Vision and Ophthalmology.
Retinitis Pigmentosa and Allied Diseases

CHAPTER 177
FIGURE 177.9. Nucleotide sequence of codons 20–26 of the human rhodopsin gene derived from leukocyte DNA of a normal individual (N79)
and of five representative patients with autosomal dominant retinitis pigmentosa included in this study and identified by their molecular genetic
numbers AD12, AD160, AD133, AD87, and AD126. The normal subject and patient AD12 show the normal sequence, whereas the other four
patients are heterozygous for the cytosine-to-adenine transversion within codon 23 (CCC to CAC). In these four patients with this mutation, the
single base change can be seen as a band marked in brackets.
From Berson EL, Rosner B, Sandberg MA, et al: Ocular findings in patients with autosomal dominant retinitis pigmentosa and a rhodopsin gene defect [Pro23His].
Arch Ophthalmol 109:92, 1991. Copyright 1991, American Medical Association.

single 0.5-Hz flashes of white light were on average 14.4 mV in times (Fig. 177.10) and are folded in three dimensions, with the
the Pro23His group, almost 10-fold larger when compared with first and seventh transmembrane segments in close proximity.
the mean amplitude of 1.7 mV in the Pro347Leu group. Cone Loops on the intradiscal side near the amino terminal tail are
isolated responses to 30-Hz flicker were also 10-fold larger on thought to be involved in the folding of the molecule, whereas
average for the Pro23His group (i.e., 5.5 mV) compared with the some loops on the cytoplasmic side appear to interact with
Pro347Leu group (i.e., 0.5 mV). The ERG and other clinical transducin as the first step in the phototransduction cascade.
findings taken together would support the idea that patients The normally folded rhodopsin molecule forms a pocket for
with Pro23His have on average a less severe disease at a given vitamin A, which is bound to a lysine residue at position 296,
age than those with rhodopsin, Pro347Leu.51 A study of the designated by the letter K in the seventh transmembrane
longitudinal course of the disease among patients with these segment (Fig. 177.11). Mutations resulting in abnormal amino
mutations has shown that patients with Pro23His have, on acids in the intradiscal or intramembranous domain (e.g.,
average, a slower course than patients with Pro347Leu.52 Pro23His) may interfere with the folding of rhodopsin and
Although patients with the Pro23His mutation retain more thereby modify the capacity of the pocket to hold vitamin A
retinal function on average than patients with the Pro347Leu with consequent instability of opsin in the outer segment
mutation, considerable variability in clinical expression exists membranes. Mutations involving the intracytoplasmic domain
at a given age among patients with these mutations, particularly (e.g., Pro347Leu) may result in abnormal transport of nascent
among the group with Pro23His. This variability in clinical rhodopsin to the outer segments. The reason mutations involv-
expression among patients with the same gene defect has raised ing the rod system eventually also lead to cone photoreceptor
the possibility that some factor or factors other than the gene cell death is not known. Studies of transgenic mice with these
defect itself are responsible for the expression of this condition. mutations as well as evaluation of cultured cell systems with
This could be the result of a modifier gene or environmental these mutations should help to define the mechanisms by
factors, or both. Risk factor analyses of patients with a given which these mutations lead to photoreceptor cell death.53
mutation and varying severity of disease may help to identify Mutations in the human retinal degeneration slow (RDS)
ameliorating or aggravating factors that may be affecting the gene on the short arm of chromosome 6 have been reported in
course of this condition with possible implications for therapy.20 other families with autosomal dominant RP.54,55 All patients so
The rhodopsin (RHO) gene is ~7 kilobases (kb) in length far studied have had abnormal rod and cone ERGs. This gene
with five exons; the gene normally encodes 348 amino acids encodes for the protein peripherin, thought to help maintain
in the rhodopsin protein. Normal rhodopsin molecules are normal outer segment structure.56 An abnormality in this gene
thought to traverse the rod outer segment membrane seven is also known to be responsible for a retinal degeneration in a 2231
RETINA AND VITREOUS

mouse model called rds. Abnormalities in the human RDS gene


have been reported to be a cause not only of RP but also of
retinal pattern dystrophy, retinitis punctata albescens, macular
dystrophy, butterfly shaped macular dystrophy, and cone-rod
dystrophy.57–60 Even among relatives who share the same
mutant RDS allele various phenotypes have been reported,
including RP, pattern dystrophy, and fundus flavimaculatus.60
Phenotypic variability may be due to an unexplained variation
in the relative involvement of rods and cones and in the
presence of yellow or yellow–white deposits at the level of the
retinal pigment epithelium seen in only some patients.
Genes that cause RP can be subclassified into those that
affect (1) the phototransduction cascade (RHO, PDE6A, PDE6B,
CNGA1, CNGB1, and SAG), (2) the retinoid cycle (ABCA4,
RLBP1, RPE65, LRAT, and RGR), (3) photoreceptor structure
(RDS/peripherin, ROM-1, FSCN2, TULP1, CRB1, RP1, and
USH2A), (4) cell–cell interactions (SEMA4A, CDH23, PCDH15,
USH1C, MASS1, USH3A, and RP2), (5) RNA intron-splicing
SECTION 10

FIGURE 177.10. Model of the normal rhodopsin protein in a rod outer factors (PRPF31, PRPF8, PRPF3, and PAP1), (6) intracellular
segment disk membrane. Each letter signifies an amino acid residue,
transport of proteins (MYO7A and SANS), (7) maintenance of
using the standard single-letter code. A, alanine; R, arginine;
N, asparagine; D, aspartic acid; C, cysteine; E, glutamic acid; cilia or ciliated cells with possible role in trafficking (BBS1,
Q, glutamine; G, glycine; H, histidine; I, isoleucine; L, leucine; BBS2, ARL6, BBS4, BBS5, MKKS, BBS7, TTC8, PTHB1, and
K, lysine; M, methionine; F, phenylalanine; P, proline; S, serine; RPGR), (8) regulation of carbon dioxide/bicarbonate balance
T, threonine; W, tryptophan; Y, tryosine; V, valine. The protein, which (CA4), (9) phagocytosis (MERTK), and (10) other yet-to-be-
contains 348 amino acids, traverses the outer segment disk defined functions of the photoreceptors and retinal pigment
membrane seven times, so that different portions of the molecule are epithelium (CERKL, BBS10, and IMPDH1).61
in the cytoplasm, within the outer segment membrane, or in the
intradiscal space. The lysine residue (K) in the seventh transmembrane
domain is covalently bound to vitamin A aldehyde (i.e., 11-cis-retinal). TREATMENT OF THE COMMON FORMS
Glycosylation sites are labeled near the N-terminal of the protein. A OF RP
disulfide bond between two cysteines in separate intradiscal loops is
indicated by a line.
Reprinted from American Journal of Ophthalmology, Vol III, Berson EL, Rosner Key Features: Retinitis Pigmentosa – Treatment of
B, Sandberg MA, et al: Ocular findings in patients with autosomal dominant Common Forms
retinitis pigmentosa and rhodopsin, proline-347, pp 614–623, Copyright 1991, • Vitamin A palmitate 15 000 IU/day for nonpregnant adults with
from Elsevier Science. normal liver function.
• Avoidance of high dose vitamin E supplementation such as
400 IU/day.
• Adults already taking vitamin A palmitate should also eat
1–2 three-ounce servings per week of omega-3 rich fish (i.e.,
salmon, tuna, mackerel, herring, or sardines) of which DHA is a
major constituent.
• Adults taking vitamin A for the first time should take DHA
supplementation (1200 mg/day) for 2 years to shorten the
interval for vitamin A to achieve its benefit. After 2 years, they
should continue vitamin A palmitate, stop DHA gelcaps, and
eat 1–2 three-ounce servings per week of omega-3 rich fish.
• Potential treatment effect – 20 additional years of vision for the
average patient who starts this regimen in their mid-30s.
• Beta-carotene is not a suitable substitute for vitamin A
palmitate in the context of this treatment as it is not
predictably converted into vitamin A.

Many treatments have been attempted without proven benefit


for the common forms of RP.62–69 These include various vita-
mins and minerals, vasodilators, tissue therapy with placental
extract, cortisone, cervical sympathectomy, injections of a
hydrolysate of yeast RNA, ultrasound, transfer factor, dimethyl
sulfoxide, ozone, muscle transplants, and subretinal injections
of fetal retinal cells. A study of patients evaluated before and
FIGURE 177.11. Schematic representation of a normal rhodopsin after receiving electrical stimulation, autotransfused ozonated
molecule folded in three dimensions to form a pocket to hold the
blood, and ocular surgery in Cuba showed that this intervention
vitamin A-derived chromophore (11-cis-retinal), which is covalently
attached to a lysine residue, designated by the letter K in the seventh provided no benefit; the results raised the possibility that this
transmembrane segment. intervention was aggravating the course of the disease.69
Modified from Applebury ML: Molecular genetics. Insight into blindness. Claims of success with one or another treatment by patients
Reprinted by permission from Nature vol 343, pp 316–317; Copyright © 1990 with RP that are based solely on subjective reporting of
2232 Macmillan Magazines Limited. improved visual function should be interpreted with caution.
Retinitis Pigmentosa and Allied Diseases

Spontaneous fluctuations in acuity and field are well known in Rates of decline for visual field area both for all randomized
this condition. Given the slow course of the disease without patients and for the higher-amplitude cohort showed similar
treatment, it will usually require several years to assess whether trends (see Table 177.3), although the differences were not
or not any proposed treatment has an effect on stabilizing or statistically significant. No significant differences were observed
slowing the course of the disease. The problem of assessing among groups with respect to rates of decline of visual acuity.
treatments may be further complicated by the genetic hetero- For a subset of 125 patients with the lowest intervisit variability
geneity of this condition and the stage of disease at which treat- (i.e., ≤5%), mean rates of decline of remaining visual field area
ment is initiated. per year were group A, 3.4%; group trace, 7.3%; group A + E,
None of these attempts at treatment was conducted with a 4.0%; and group E, 5.3%. Comparisons of specific groups
randomized, controlled, double-masked protocol, which is showed that the rate of decline of visual field area for group A
necessary to avoid possible patient or examiner biases. Most of differed significantly (p=0.03) from that of group trace, whereas
these studies were performed without ERG data as an end-point
for evaluating efficacy, so that the amount of remaining retinal
function could not be quantitated in an objective manner.
Preliminary data obtained while conducting a natural history
study of the course of RP revealed that those patients self-
treating with a separate capsule of vitamin A or vitamin E or
both were losing less retinal function than those not self-

CHAPTER 177
treating with these supplements.70 This prompted a random-
ized, controlled, double-masked trial among 601 adult patients
to determine whether supplementation with vitamin A or
vitamin E, alone or in combination, would halt or slow the
progression of the common forms of RP including Usher syn-
drome type II. Patients were randomly assigned to one of
four treatment groups, as follows: vitamin A, 15 000 IU/day
plus vitamin E, 3 IU/day (Group A); vitamin A, 75 IU/day plus
vitamin E, 3 IU/day (Group Trace); vitamin A, 15 000 IU/day
plus vitamin E, 400 IU/day (Group A + E); and vitamin A,
75 IU/day plus vitamin E, 400 IU/day (Group E). The main
outcome variable was the 30-Hz cone flicker ERG.70–72
Mean annual rates of decline of remaining ERG amplitude
were slowest for the group taking 15 000 IU/day of vitamin A
and fastest for the group taking 400 IU/day of vitamin E. These
rates were observed among all randomized patients as well as
among a subgroup of 354 patients with slightly higher initial
ERG amplitudes (i.e., ≥ 0.68 mV) who could be followed more
reliably and who were designated ‘the higher amplitude cohort’.
Mean annual rates of decline of remaining 30-Hz ERG ampli-
tude among this cohort were as follows: group A, 8.3%; group
trace, 10%; group A + E, 8.8%; and group E, 11.8%. These
results are summarized in Table 177.3. The treatment effect is
plotted by year in Figure 177.12. Comparison of those taking
vitamin A 15 000 IU/day versus those not on this dose revealed
that the beneficial effect of vitamin A was significant at the
P =.01 level for all randomized patients and at the P <.001
level for the higher-amplitude cohort. Comparison of those
taking vitamin E 400 IU/day versus those not on this dose
revealed no significant treatment effect among all randomized
patients but did show an adverse effect among those in the
higher amplitude cohort (p=0.04).

TABLE 177.3. Mean Rates of Decline in Visual Function by


Treatment Group Among Patients with the Common Forms of
Retinitis Pigmentosa Supplemented with Vitamins A and E*
FIGURE 177.12. Mean change from baseline over 6 years in 30-Hz
All Randomized Patients Higher Amplitude Cohort ERG amplitude in the higher amplitude cohort by treatment group
Test A Trace A+E E A Trace A+E E (top), by vitamin A main effect (middle); and by vitamin E main effect
30 Hz* 6.1 7.1 6.3 7.9 8.3 10.0 8.8 11.8 (bottom). Sample sizes for years 1 through 6, respectively, were
n = 171, n = 167, n = 168, n = 164, n = 123, and n = 59 for patients
0.5 Hz* 8.7 9.6 9.1 10.6 8.1 9.4 8.4 10.9 receiving vitamin A, 15 000 IU/day; and n = 178, n = 182, n = 172,
n = 171, n = 125, and n = 64 for patients receiving vitamin A,
Field Area* 5.6 5.9 6.2 6.3 6.3 7.2 7.3 7.8
75 IU/day. Sample sizes for years 1-6, respectively, were n = 178,

Visual Acuity 1.1 0.9 0.7 0.9 0.8 0.8 0.7 0.7 n = 177, n = 173, n = 168, n = 122, and n = 61 for the patients on
vitamin E, 400 IU/day; and n = 171, n = 172, n = 167, n = 167,
*Percent decline in remaining function per year

Letters lost per year n = 126, and n = 62 for patients receiving vitamin E, 3 IU/day.
(Modified from Berson EL, Rosner B, Sandberg MA, et al.: Arch Ophthalmol From Berson EL, Rosner B, Sandberg MA, et al: A randomized trial of vitamin A
1993; 111:761–772; copyright 1993, American Medical Association. and vitamin E supplementation for retinitis pigmentosa. Arch Ophthalmol
111:761–772, 1993. Copyright 1993, American Medical Association. 2233
RETINA AND VITREOUS

the rates of decline of the A + E and E groups did not differ In older adults, long-term vitamin A supplementation has
significantly from that of the group trace. Therefore, for this been associated with a decrease in bone density and up to a 1%
subset of 125 patients representing all genetic types, a increased risk of hip fractures.78,79 Therefore, postmenopausal
significant beneficial effect of vitamin A supplementation on women and men aged 49 and over who are taking vitamin A
visual field area could be detected.73 should be advised to consult with their physician about their
The optimal total intake of vitamin A appeared to be bone health and be followed regularly in this regard. Vitamin A
~18 000 IU/day; that is, a supplement of 15 000 units plus a should not be given to patients with RP who have had renal
regular diet of ~3500 units of vitamin A per day resulted in the transplants as they have excessive renal re-absorption of vitamin
smallest ERG decline (Fig. 177.13). Vitamin A intake greater A and, therefore are more susceptible to toxicity. Patients on chronic
than 18 380 IU/day provided no greater benefit. Vitamin A doxycycline should not take vitamin A because the combination
intake of 25 000 IU/day or greater is potentially toxic over the has been associated with increased intracranial pressure.
long term.74–76 While conducting the clinical trial on vitamins A and E, data
No case of toxicity in adults in good general health on this were obtained raising the possibility that docosahexaenoic acid
dose has been reported.70–77 No significant differences in treat- (DHA) in addition to vitamin A could further slow the course of
ment effects were noted among different genetic types. The pre- RP. This led to a second randomized, controlled, double-masked
cise mechanism by which vitamin A supplementation provides trial for the typical forms of RP. Patients were randomly assigned
its benefit is not known. It has been speculated that vitamin A to DHA capsules (1200 mg/day) or control fatty acid capsules;
rescues remaining cones, thereby explaining how one supple- all participants were given vitamin A palmitate 15 000 IU/day.
SECTION 10

ment may help a group of patients many of whom have dif- In this trial the main outcome variable was the total point score
ferent rod-specific gene defects. Vitamin E may lead to an for the 30–2 program of the Humphrey field analyzer. Among all
adverse effect on the course of RP by inhibiting the absorption participants taken together, DHA supplementation by capsules
or transport of vitamin A.70 did not, on average, slow the course of RP over a 4-year
Based on the results of this trial, it is recommended that interval.80 However, a subgroup analysis showed that those
most adults with the common forms of RP should take 15 000 already taking vitamin A who also ate an omega-3 rich diet
units of vitamin A palmitate daily under the supervision of their (≥0.2 g/day) of which DHA is a major constituent, had a
ophthalmologists and avoid high-dose supplements of vitamin 40–50% slowing of annual decline in visual field sensitivity.81
E such as the 400 units used in this trial. It is also recom- This omega-3 rich diet consisted of two or more 3-ounce
mended that patients continue on a regular diet without servings of oily fish per week (i.e., salmon, tuna, herring,
specifically selecting foods containing high levels of preformed mackerel, or sardines). DHA is thought to facilitate the release
vitamin A. The palmitate form of vitamin A is recommended, of vitamin A from its carrier protein (interphotoreceptor
as this was the form used in this study. Other forms might be retinoid binding protein, IRBP) in the subretinal space. It has
suitable, but some are probably not, for example beta-carotene, been estimated that an average patient age 37 with typical RP
which is not predictably converted to vitamin A from one already on vitamin A palmitate 15 000 IU/day who maintains
patient to another. this oily fish DHA-rich diet would be expected to lose virtually
As a precaution, patients should be advised to have a all central visual field sensitivity by age 78, whereas an average
pretreatment assessment of fasting serum vitamin A and liver patient who eats less than two servings per week would be
function and annual evaluations thereafter. Because of the expected to lose all central visual field sensitivity by age 59.81 In
potential for birth defects, women who are pregnant or planning summary, whereas vitamin A alone is thought to provide seven
to become pregnant are advised not to take this dose of vitamin additional years of vision, vitamin A with an oily fish diet
A. Because patients under 18 years were not evaluated, no formal provides almost 20 years of visual preservation for the average
recommendation for patients under this age can be made.70 patient with RP who starts this regimen in their mid-30s.
About 3 months after starting an oily fish diet (thereby
allowing sufficient time for red blood cell (RBC) turnover),
patients with RP should have a measurement of their fasting
RBC DHA to confirm that the RBC DHA level is at least 4% of
total RBC fatty acids as it has been reported that such patients
have, on average, a slower rate of decline of visual field
sensitivity than those with lower levels.81 If the RBC DHA level
is not at least 4%, patients should be advised to consult with
their physician on how to best reach this level through food. If
the level is 4% or greater, this test should be repeated annually
thereafter.
Although high DHA supplementation (1200 mg/day) has no
benefit over a 4-year interval among patients taking vitamin A,80
it has been reported that this dose can shorten the interval for
vitamin A to achieve its benefit among those taking vitamin A
for the first time.81 Therefore, adults with RP starting vitamin
FIGURE 177.13. Mean ± SE decline from baseline in 30-Hz ERG A for the first time should be advised to take vitamin A
amplitude by total vitamin A intake (diet plus capsules) irrespective of palmitate 15 000 IU/day and DHA capsules 600 mg twice per
randomization assignment for all patients in the higher-amplitude day for 2 years (i.e., three 200 mg capsules both morning and
cohort. The mean decline was calculated as the mean of screening night with meals). After 2 years, patients should discontinue
and baseline minus the mean of all follow-up visits by quintile of total
DHA supplementation by capsules because no evidence was
vitamin A intake averaged over all visits. Sample sizes were 69, 72,
74, 65, and 74 for the lowest to highest quintiles of total vitamin A found for continued benefit and because a slight tendency
intake. Vertical bars indicate SEs. toward adversity on ocular function was observed over the long-
From Berson EL, Rosner B, Sandberg MA, et al: A randomized trial of vitamin A term among patients on high DHA with vitamin A.81 After
and vitamin E supplementation for retinitis pigmentosa. Arch Ophthalmol stopping DHA capsules after 2 years, patients should continue
2234 111:761–772, 1993. Copyright 1993, American Medical Association. the vitamin A and eat two 3-ounce servings per week of omega-
Retinitis Pigmentosa and Allied Diseases

3 rich fish. About 3 months thereafter, RBC DHA levels should Patients with this condition are treated with a low-fat diet
be checked as described above. It should be noted that com- and supplements of the fat-soluble vitamins A, E, and K.
bining an omega-3 rich diet with DHA capsules did not provide Vitamin A supplementation has been shown to restore elevated
any additional benefit. dark-adaptation thresholds and reduced ERG responses to
Sources of vitamin A palmitate and DHA capsules as well as normal in two patients in the early stages (Fig. 177.14).93,94
laboratories that perform measurements of RBC DHA are main- More advanced cases have not responded, but in one such case,
tained on the website of the Foundation Fighting Blindness in which the retina was examined after the death of the patient,
(www.blindness.org). widespread loss of photoreceptor cells was observed.95 Vitamin
A therapy may not maintain retinal function over the long
RP ASSOCIATED WITH HEREDITARY term, as patients have been reported in whom vitamin A levels
ABETALIPOPROTEINEMIA have been restored to normal and yet the retinal degeneration
has appeared to progress.96 Since these patients have low serum
vitamin E levels, supplementation with vitamin E in addition to
Key Features: Retinitis Pigmentosa – Treatment of Rare vitamin A has been advocated with reported stabilization of
Forms retinal function.97–101
• Hereditary abetalipoproteinemia (Bassen–Kornzweig syndrome)
– low-fat diet and vitamins A, E, and K. RP ASSOCIATED WITH REFSUM DISEASE

CHAPTER 177
• Refsum disease – low-phytol, low-phytanic acid diet while
maintaining body weight. Refsum disease is a recessively inherited condition in which the
• Familial isolated vitamin E deficiency – oral vitamin E patient accumulates exogenous phytanic acid.102,103 Findings
supplementation. include a peripheral neuropathy, ataxia, an increase in cerebro-
spinal fluid protein with a normal cell count, and RP. All
patients have elevated serum phytanic acid. Some cases have
In 1950, Bassen and Kornzweig described an 18-year-old girl, anosmia, neurogenic impairment of hearing, electrocardiogram
born of first cousins, who had a malabsorption syndrome, a abnormalities, and skin changes resembling ichthyosis. The
generalized retinal degeneration, a diffuse neuromuscular fundus can appear granular with areas of depigmentation
disease similar to Friedreich ataxia, and a peculiar crenation of around the periphery with a subnormal ERG in the early stages
the RBCs, now called acanthocytosis. In 1956, low serum or can show more typical RP with a nondetectable ERG in more
cholesterol (<50 mg/dL) was observed.82–86 Soon thereafter, an advanced stages.104
absence of low-density plasma lipoproteins or so-called beta- A defect exists in the conversion of phytanic acid to ∂-hydroxy
lipoproteins was found, and the term abetalipoproteinemia was phytanic acid, specifically in the introduction of a hydroxyl
assigned to this recessively inherited disorder.87–89 Other classes group on the ∂-carbon of phytanic acid (Fig. 177.15).105 The
of lipoproteins have also been found to be abnormal.90 pathogenesis appears to involve accumulation of phytanic acid
Patients with hereditary abetalipoproteinemia can assimilate in a variety of tissues, including the retinal pigment epithelium.106
fat into the intestinal mucosa, but a defect exists in its removal Treatment consists of restricting not only animal fats and
from this site because of the lack of chylomicra. Intestinal milk products (i.e., foods that contain phytanic acid) but also
biopsies have revealed normal-sized villi filled with lipid green leafy vegetables containing phytol.107 Success of treat-
droplets that are essentially triglycerides. Mutations in the gene ment depends on the patient’s maintaining his or her body
encoding a microsomal triglyceride transfer protein have been weight; if body weight becomes reduced, phytanic acid is
found in patients with this condition.91 It appears that the liver released from tissue stores, resulting in an increase of phytanic
and then the retina become depleted of vitamin A. Abnormal acid in serum and exacerbation of symptoms. Refsum reported
ERGs have been reported in children in whom the fundi were two patients whose serum phytanic acid levels were lowered to
still normal.92 The original case described by Bassen and normal and who showed improvement in motor nerve conduc-
Kornzweig showed multiple white dots in the early stages, but tion velocity, some relief of ataxia, and return of the cerebro-
by age 31 years, the patient developed multiple areas of pigment spinal fluid protein to normal. Moreover, the RP and hearing
epithelial cell atrophy. In other cases, the typical intraretinal impairment did not progress; one of these patients was followed
pigment associated with RP has been noted in the retinal for 10 years and the other for many years.108 One young adult
periphery. with a mild form of this disorder has been followed on a low-

FIGURE 177.14. Full-field ERGs to red (top)


and blue (middle) light, equal for rod vision, and
brighter white stimulus (bottom) from a patient
with hereditary abetalipoproteinemia (dark-
adapted). Responses in the left column were
obtained before vitamin A therapy, those in
middle column at 6 h, and those on right at 24 h
after vitamin A therapy. Two to three responses
to the same stimulus are superimposed. Light
stimulus begins with each trace. Calibration
(lower right) signifies 0.06 mV vertically and 60
msec horizontally.
From Gouras P, Carr RE, Gunkel RD: Retinitis
pigmentosa in abetalipoproteinemia: Effects of vitamin
A. Invest Ophthalmol Vis Sci 10:784, 1971. ©
Association for Research in Vision and Ophthalmology.

2235
RETINA AND VITREOUS

FIGURE 177.16. Full-field ERGs from a normal subject and a patient


with a mild form of Refsum disease before and 4 years after treatment
SECTION 10

with a low-phytol, low-phytanic acid diet. Pretreatment responses


were recorded at age 31 years.
From Berson EL: Electroretinographic findings in retinitis pigmentosa. Jpn J
Ophthalmol 31:327, 1987.

OTHER FORMS OF RP
Some 15–20% of individuals with RP have associated hearing
loss, sometimes referred to as Usher syndrome in recognition of
the British ophthalmologist, C. H. Usher, who observed that
this condition was recessively inherited.111,112 Patients with
Usher syndrome type I typically have night blindness in the first
or second decade of life, profound congenital deafness (i.e.,
>70 dB loss at all frequencies) with unintelligible speech, and
vestibular ataxia, whereas those with Usher syndrome type II
usually report night blindness in the second to fourth decades,
have a partial high-tone hearing loss with intelligible speech,
and do not show ataxia.113 Patients with Usher syndrome type
II have abnormalities in the cilia of sperm and in the connecting
cilia in many remaining photoreceptors in autopsy eyes.114,115
FIGURE 177.15. Phytanic acid, its immediate precursors and
Patients with Usher syndrome type III have RP with onset of
metabolites, and site of enzyme defect in Refsum disease (Rd).
From Eldjarn L, Stokke O, Try K: Biochemical aspects of Refsum disease and
hearing loss in mid-adulthood that can progress to profound
principles for the dietary treatment. In Vinken PJ, Bruyn GW [eds]. Handbook of deafness; some have vestibular ataxia.116 All forms of Usher
Clinical Neurology. Amsterdam, North Holland, 1976, pp 519–541. syndrome appear to be slowly progressive over the long term.
Patients with Usher syndrome type II were included in the clinical
trial of vitamin A70 and DHA80,81 and, therefore, adult patients
phytol, low-phytanic acid diet for 4 years; full-field ERGs, are being treated with vitamin A palmitate 15 000 IU/day and
reduced ~75% below normal before commencement of this diet, an omega-3 rich diet as described earlier in this chapter.
have remained about the same over this period (Fig. 177.16). Molecular genetic analyses have revealed gene loci for Usher
Long-term effects of this diet on retinal function continue to be syndrome type I (further subdivided into forms B through F); for
studied.38 Usher syndrome type II (forms A through D); and for Usher
syndrome type III (form A only so far detected) (see Table
FAMILIAL ISOLATED VITAMIN E 177.2). The authors who originally reported Usher syndrome
DEFICIENCY type IA have subsequently found that this form does not exist
as initially described.117
Another rare recessively inherited form of ataxia associated Some 2–6% of congenitally deaf children will develop Usher
with RP has recently been termed familial isolated vitamin E syndrome type I. If a child presents with profound deafness and
deficiency. These patients present in adulthood with Friedreich- a balance disorder manifested by late onset of walking, usually
like ataxia, dysarthria, hyporeflexia, and decreased propriocep- after 15 months of age, the possibility of Usher syndrome type
tive and vibratory sensation as well as markedly decreased I should be considered.118 The diagnosis of RP as part of Usher
serum vitamin E levels. In later stages, patients can develop the syndrome can be made in early life with ERG testing. No tests
fundus changes of RP and abnormal ERGs. Molecular genetic of retinal function are yet available to identify carriers of Usher
analysis has revealed a mutation in the alpha-tocopherol- syndrome.
transfer protein (alpha-TTP) gene. Oral administration of The Laurence–Moon–Bardet–Biedl syndrome includes RP,
vitamin E restored serum vitamin E levels to normal and mental retardation, polydactylism, truncal obesity, and hypo-
appeared to halt or slow the progression of the neurologic gonadism as the most frequent features.119–122 Some authors
abnormalities and RP in three patients followed for 1, 4, and have subdivided this syndrome into two disorders: poly-
2236 10 years, respectively.109,110 dactylism is found primarily in patients with the Bardet–Biedl
Retinitis Pigmentosa and Allied Diseases

form, whereas neurologic findings are typically observed in Leber congenital amaurosis has been produced in mice with
patients with the Laurence–Moon form.123–124 However, some knockout of the RPGRIP1 gene, and rod structure and function
patients have been described with both polydactylism and have been restored with subretinal injection of the RPGRIP1
neurologic abnormalities and therefore would seem to qualify gene also carried in an AAV vector.141 It remains to be
for inclusion in both syndromes.125,126 Variability of clinical established whether gene therapy can be safely used to reverse
expression is well known, and some patients may have this retinal malfunction in humans with Leber congenital
condition without mental retardation or polydactylism.127 amaurosis with remaining photoreceptor cells.
Genetic heterogeneity also exists, as the condition is caused by Retinitis punctata albescens can be associated with some
at least 12 different genes (BBS1, BBS2, ARL6, BBS4, BBS5, signs and symptoms characteristic of RP.142 Patients usually
MKKS, BBS7, TTC8, PHTB1, BBS10, BBS11, and BBS12). These present with profound adaptational problems and gradual loss
genes in the aggregate account for only about half of the cases of peripheral vision. Examination with a direct ophthalmoscope
of this syndrome. Renal disease can be a part of this reveals multiple punctate white deposits in the macula and
syndrome,127 and therefore, patients should have their blood around the mid-periphery at the level of the pigment epithe-
pressure and urine checked periodically, with appropriate lium, for which this condition was named. Patients have retinal
treatment instituted for their renal disease if detected. arteriolar attenuation and some develop round areas of atrophy
Retinal degeneration is the most common feature of the of the retinal pigment epithelium as well as intraretinal bone
Laurence–Moon–Bardet–Biedl syndrome, occurring in ~90% of spicule pigment in the midperiphery. This raises the possibility
cases.127 The macula is often involved early, leading to the that this condition is a variant of RP.143 This form of retinal

CHAPTER 177
suggestion that these patients have an inverse form of RP. The degeneration may affect the eyes of a patient asymmetrically;
fundus in early life may be granular without pigment formation, therefore, these patients often receive a neurologic evaluation in
so that the diagnosis is established only after ERG testing.128 A search of an intracranial abnormality before it is realized that
child born with polydactylism to normal parents should be a the visual loss is due to a widespread retinal degeneration. Full-
suspect for this syndrome. Once this syndrome is identified in field ERGs of such patients are invariably abnormal with reduc-
one individual, parents would know that they have as much as tions in amplitude and delays in implicit times. The condition
a 25% chance with each succeeding childbirth of having another is usually slowly progressive, although the course appears to
child with this condition. The inheritance of the Bardet–Biedl vary from one individual to another. This condition has been
syndrome can be unusual as mutations in two unlinked thought to be inherited by an autosomal recessive mode, but a
Bardet–Biedl genes at one locus have been detected in some dominant mode of transmission can occur. Therefore, ERG
families and expression of this condition may be modified by testing of relatives of affected patients is recommended to help
a mutation at a second locus that may not alone cause establish the mode of transmission. A null mutation in the
disease.129–131 No treatment is currently known for the retinal peripherin/RDS gene has been reported in one family with this
degeneration associated with this syndrome. condition with a dominant mode of transmission144; other
Congenital amaurosis of Leber is an autosomal recessive families with an autosomal recessive mode of transmission
disorder associated with severe reduction in vision near birth have shown mutations in the cellular retinaldehyde-binding
and very reduced ERGs.132–133 Patients characteristically have protein (CRALBP) gene.145–147
hyperopia and nystagmus, and fundus examination reveals Another atypical form of RP has been designated as
granularity, white flecks, or intraretinal bone spicule pigment, “progressive cone–rod degeneration.”148 Patients with this form
or some combination of these. Carrier parents have a 25% present with reduced visual acuity, photophobia, color deficiency,
chance with each childbirth of having a child affected with this and night deficiency and show signs of macular degeneration,
condition. Mental retardation has been reported in some retinal arteriolar attenuation, and in some cases, intraretinal
patients, possibly secondary to visual impairment. Leber con- bone spicule pigment in the peripheral fundus. Dark-adaptation
genital amaurosis involving the retina and pigment epithelium testing shows elevated final dark-adaptation thresholds across
should not be confused with Leber optic atrophy, a maternally the fundus, and full-field ERGs show a profound loss of cone
inherited condition involving the optic nerve associated in some function and some reduction in rod function. This condition is
families with an abnormality in mitochondrial DNA.134,135 usually inherited by an autosomal recessive mode but can also
Abnormalities in 12 genes have been so far identified as be inherited by an autosomal dominant mode. Mutations have
causes of Leber congenital amaurosis. Genes so far implicated been found in the retina-specific ATP-binding cassette trans-
are GUCY2D, CRB1, IMPDH1, RPE65, AIPL1, RDH12, porter (ABCA4) gene,149 the same gene that has been found to
RPGRIP1, CRX, TULP1, LRAT, CEP290, and LCA5. These be abnormal in patients with juvenile macular degeneration
genes account for more than half of the known cases. In one (Stargardt disease).150–152 No treatment is yet known for pro-
form of this condition mutations in GUCY2D (retinal guanylate gressive cone–rod degeneration.
cyclase 1) suggest that cGMP production in photoreceptors is Other atypical forms of RP include pericentral, paravenous,
abolished. Consequently, photoreceptor excitation would be or unilateral RP. The pericentral form is characterized by a near
expected to be impaired due to closure of cGMP-gated cation mid-peripheral scotoma extending from the 5–30° isopter (Fig.
channels with hyperpolarization of the photoreceptors. It has 177.17) (in contrast to typical RP where the field loss initially
been proposed that the cGMP concentration in photoreceptor extends from the 20–40° isopter), single flash (0.5 Hz) ERGs
cells cannot be restored to the dark level, leading to a situation greater than 100 mV and 30-Hz cone ERGs greater than 15 mV
equivalent to constant light exposure during photoreceptor initially (Fig. 177.18), and a normal or nearly normal final dark
development.136 A model for this disease exists in the rd/rd adaptation threshold in the periphery. This form progresses at a
chicken, which is functionally blind at hatching, with an slower rate than typical RP; in later life they can lose central
extinguished ERG, absent guanylate cyclase activity, and a vision but they retain considerable peripheral vision.153 The
mutation in the guanylate cyclase gene.137 Mutations in paravenous form is usually characterized by slightly reduced
another gene, designated as RPE65, affect vitamin A full-field ERGs and intraretinal pigment and atrophy of the
metabolism in the retinal pigment epithelium.138,139 A canine pigment epithelium confined to the distribution of the retinal
model of this condition with an RPE65 gene deficiency has been veins in each eye. In some cases the macula degenerates as well
rescued with subretinal injection of the RPE65 gene carried by (Fig. 177.19). Patients with paravenous disease may lose central
an attenuated adenoviral (AAV) vector.140 Another model of vision but, like pericentral RP, they retain peripheral vision into 2237
RETINA AND VITREOUS

FIGURE 177.17. Fundus photographs for a


patient with pericentral retinitis pigmentosa to
show an increase in pericentral bone-spicule
pigmentation over an 18-year interval.
Reprinted from Am J Ophthalmol, vol 140, Sandberg
MA, Gaudio AR, Berson EL. Disease course of patients
with pericentral retinitis pigmentosa, pp 100–106,
Copyright 2005 from Elsevier Science.

FIGURE 177.18. Visual fields to the V-4e white


test light and full-field electroretinograms to
0.5 Hz and 30 Hz flashes spanning 22 years
from a patient with pericentral retinitis
pigmentosa to illustrate disease progression.
Reprinted from Am J Ophthalmol, vol 140, Sandberg
SECTION 10

MA, Gaudio AR, Berson EL. Disease course of patients


with pericentral retinitis pigmentosa, pp 100–106,
Copyright 2005 from Elsevier Science.

FIGURE 177.19. Fundus photographs of the right eye of a patient at age 22 years (left) and at age 39 years (right) with pigmented paravenous
retinochoroidal atrophy. At age 22 years, the fundus showed paravenous intraretinal bone spicule deposits overlying areas of pigment epithelial
and choroidal atrophy with some disturbance of the retinal pigment epithelium in the macula. At age 39 years, progression can be seen with
more paravenous pigment and some clumped pigment in the macula.
Reprinted from Am J Ophthalmol, vol 141, Choi JY, Sandberg MA, Berson EL. Natural course of ocular function in pigmented paravenous retinochoroidal atrophy,
pp 763–765, Copyright 2006 from Elsevier Science.

later life.154 Unilateral RP is characterized by fundus changes of No evidence exists that patients with pericentral, paravenous,
RP in one eye with no evidence of RP in the fellow eye. Full-field or unilateral RP will benefit from vitamin A treatment.
ERGs are substantially reduced in the affected eye and normal An estimated 0.5% of patients with RP have a clumped pat-
in the fellow eye. Patients with unilateral RP have progressive tern of pigmentation in the mid-peripheral fundus, sometimes
disease in the affected eye but have not been observed to develop referred to as ‘clumped pigmentary degeneration’. These patients
a generalized form of RP in the fellow eye at a later time. characteristically have hyperopia, night blindness, and reduced
Pericentral RP can be inherited by a recessive or dominant mode and delayed full-field ERGs. Humphrey static perimetry with
while paravenous and unilateral RP have been found charac- blue stimuli on a yellow background to evaluate short-wave (S)
2238 teristically in patients with no family history of this condition. blue-cone function and white stimuli on a white background to
Retinitis Pigmentosa and Allied Diseases

evaluate function mediated by all three cone types has revealed further increased and the visual fields become constricted at a
that some of these patients with clumped pigment have rela- time when ERGs are profoundly reduced or nondetectable and
tively enhanced blue-cone function and such patients are extensive choroidal atrophy and clumped pigment are visible
therefore considered to have the enhanced S-cone syndrome.155 around the peripheral fundus (see Fig. 177.2b). Males usually
Molecular genetic analyses have revealed mutations in the retain little, if any, central vision beyond the age of 60 years.158,159
NR2E3 gene which is thought to encode a ligand-dependent Obligate female carriers of this X-linked disease may demon-
transcription factor156 or in the NRL gene which encodes a basic strate fundus changes that include patchy depigmentation of
motif-leucine zipper protein.157 Patients with enhanced S-cone the retinal pigment epithelium and coarse pigment granularity
syndrome have been shown to have shared mutations in the or even pigment clumps in the periphery. However, carriers
NR2E3 gene with patients with Goldmann–Favre syndrome. typically retain normal visual acuities and normal final dark-
Patients with NR2E3 mutations appear to inherit this condition adapted rod thresholds.159–161 Generally, carriers have been thought
by an autosomal recessive mode while those with NRL muta- to have a nonprogressive course, but carriers with severe disease
tions can inherit this condition by a dominant or recessive have been described.159 In contrast to the carrier state of
mode. Patients with the enhanced S-cone syndrome appear to X-linked RP, ERGs of carriers of choroideremia usually are
have a slowly progressive disease with gradual loss of peripheral normal,159 although reduced amplitudes have been reported in
field. No treatment is yet known. some cases.159,161
Study of a patient with choroideremia with a large deletion
CHOROIDEREMIA helped to assign the choroideremia gene to a small segment of

CHAPTER 177
the long arm of the X-chromosome, specifically in the Xq21
Young males with choroideremia characteristically have normal band.162,163 The gene (CHM) responsible for this condition,
visual acuities, minimally increased dark-adaptation thresholds, encodes Rab escort protein 1 (REP-1).164 Many mutations in this
and full visual fields to large test lights at a time when granu- gene have been described subsequently.165,166 Loss of the REP-1
larity and depigmentation of the retinal pigment epithelium are protein is thought to compromise Rab protein prenylation; the
seen around the fundus periphery. ERGs are reduced in ampli- lack of prenylation, in turn, would affect protein trafficking
tude with delays in b-wave implicit time (Fig. 177.20). In more inside the retinal pigment epithelium or choroidal cells triggering
advanced stages in adulthood, dark-adaptation thresholds are the degenerative process in this condition.167–169 An animal
model of this condition has been created and is under study.170
Interfamilial and intrafamilial variability of clinical expres-
sion is known to exist in this disease. The natural course of
choroideremia on a year-to-year basis is being investigated.
Correlations, if any, between specific mutations and clinical
expression of the disease remain to be completed. No treatment
is yet known.

GYRATE ATROPHY OF THE CHOROID AND


RETINA

Key Features: Gyrate Atrophy of the Choroid and Retina


– Diagnosis and Management
• Symptoms – night blindness, loss of peripheral field, eventual
loss of visual acuity.
• Ocular findings - myopia, peripheral chorioretinal atrophy,
reduced electroretinograms.
• Biochemical findings - hyperornithinemia, hyperornithinuria,
hypolysinemia.
• Treatment – low-protein, low-arginine diet in all cases; vitamin
B6 supplementation for the B6-responsive form.

Gyrate atrophy of the choroid and retina is a rare chorioretinal


degeneration inherited by an autosomal recessive mode of
transmission.171,172 Patients usually report night deficiency and
loss of peripheral vision in adolescence. Ocular findings include
myopia, constricted visual fields, elevated dark-adaptation
FIGURE 177.20. ERG responses of four males with choroideremia. thresholds, very small or nondetectable ERG responses, and
Cone flicker responses to white 30-cps flicker remained detectable in chorioretinal atrophy distributed circumferentially around the
the oldest male when rod responses to blue light were not detectable. peripheral fundus (see Fig. 177.2c) and sometimes near the
Normal responses are shown for comparison. Time of stimulus onset optic disk. In addition to the ocular findings, abnormalities in
is designated by ‘vertical hatched lines’ in columns 1 and 2 and electroencephalograms, muscle and hair morphology, and
vertical shock artifacts in column 3. Arrows and vertical bar on rod mitochondrial structure in the liver have been reported.173
responses to blue light show the range of normal b-wave implicit
Enlargement, coalescence, and posterior extension of areas of
times. Responses to single flashes of white light show reduced a- and
b-wave amplitudes in all males. Arrows on white flicker responses atrophy have been observed in young patients within 2 years.174
show delayed implicit times in the affected males. Patients develop cataracts in young adulthood and often require
Reprinted from Sieving PA, Niffenegger AB, Berson EL, Am J Ophthalmol, surgery. If untreated, patients usually become virtually blind
vol 101, Electroretinographic findings in selected pedigrees with choroideremia, between the ages of 40 and 55 years as a consequence of
pp 361–367, Copyright 1986, from Elsevier Science. extensive chorioretinal atrophy.175 2239
RETINA AND VITREOUS

Patients with this condition have plasma ornithine concen- (251 and 313 mmol/L) in the other pair. These younger siblings
trations elevated 10- to 20-fold above normal176,177 due to a had significantly less atrophy than their elder siblings when
deficiency of ornithine aminotransferase (OAT) activity (Fig. they reached or approached the same age at which their elders
177.21).178–180 Affected individuals cannot convert ornithine to had begun the diet. Thus, there is evidence that a low-protein,
pyrroline-5-carboxylic acid (PCA); this deficiency can be low-arginine diet can slow progression of the chorioretinal
detected in extracts of cultured skin fibroblasts. Patients have degeneration, but only a few patients have so far been studied.
virtually no OAT activity in contrast to normal persons, where- The goal of treatment is to maintain plasma ornithine levels
as carrier parents have a partial deficiency. Cultured cells from as close to normal as possible. Since some patients may respond
some patients have shown increased OAT activity when the to supplementation with pyridoxine (vitamin B6), all patients
cofactor for OAT, namely pyridoxal phosphate or vitamin B6, is are initially given a trial of this vitamin to determine to what
added to the media in increased concentration. Plasma lysine181 extent, if any, it will lower plasma ornithine levels. Both
and glutamate and glutamine,182 as well as serum and urine pyridoxine-responsive and nonresponsive patients are then
creatine,183 have also been found to be reduced. More than placed on a low-protein, low-arginine diet. Based on plasma
60 mutations have been discovered in the OAT gene on ornithine levels, biochemical control has been classified as good
chromosome 10 in affected patients.184 to excellent (<200 mmol/L), fair (200 to 400 mmol/L), or poor
Since arginine is a precursor of ornithine, and since arginine, (>400 mmol/L). In the management of children, expertise is
but not ornithine, is a constituent of food protein, it has been required to be certain that growth and development remain
suggested that dietary restriction of protein and arginine will normal while lowering ornithine levels with a low-protein diet.
SECTION 10

reduce plasma ornithine levels in these patients.185 OAT- An arginine-free essential amino acid mixture (e.g., Cyclinex-1
deficient mice produced by gene targeting develop a retinal or -2 [Ross Laboratories], depending on the patient’s age) is used
degeneration over several months that is amenable to treatment to provide sufficient nitrogen and meet essential amino acid
postweaning with an arginine-restricted diet.186 requirements. In adults, a low-protein diet is also likely to result
The hyperornithinemia associated with human disease has in amino acid deficiency. Thus, adults should also be placed on
been lowered toward normal with a low-protein, low-arginine an arginine-free essential amino acid mixture. Lysine supple-
diet in all cases so far studied185,187,188 and with vitamin B6 (300 mentation may be necessary, depending on plasma levels. As a
to 500 mg/day) in some cases.189–191 However, extreme protein precaution, all patients are placed on a multivitamin prepara-
restriction (10–15 g/day) with substantial lowering of plasma tion with minerals. In addition to a regular ocular examination,
ornithine, accomplished under supervision in the hospital, has all patients on this treatment regimen should have their amino
been difficult to achieve at home, and therefore, many of the acid and protein levels monitored periodically.194
patients have followed modified (20–35 g/day) protein restric-
tion with slight rises in their plasma ornithine levels. Some OTHER RETINAL DEGENERATIONS
investigators have reported improvements in visual acuity,
visual fields, dark-adaptation thresholds, or ERGs in patients Retinal degenerations involving the photoreceptors across the
with gyrate atrophy after onset of either the diet or vitamin retina can exist in association with other systemic findings. If a
B6,187 whereas others have not documented any significant patient presents with ophthalmoplegia and retinal degeneration,
improvement in visual function despite substantial reductions the Kearns–Sayre syndrome should be considered. Abnormalities
in plasma ornithine concentrations.191 Improvement in muscle include ptosis, chronic progressive external ophthalmoplegia, a
morphology after creatine supplementation has been reported disturbance of the retinal pigment epithelium, ERGs usually
in several patients.192 reduced in amplitude, and respiratory distress. Some patients
The effect of severe arginine restriction has been evaluated in develop heart block and may require a pacemaker.195,196 The
two pairs of siblings younger than 10 years who were followed diagnosis is confirmed by the observation of ragged red fibers on
for 5–7 years.193 The plasma ornithine levels were reduced to muscle biopsy. This syndrome has been associated with mito-
approximately the normal range (106 and 121 mmol/L) in one chondrial DNA mutations; treatment of one patient with
pair of siblings and reached twice the upper limit of normal coenzyme Q10 and succinate resulted in clinical improvement

FIGURE 177.21. Pathways of ornithine


metabolism.
From Weleber RG, Kennaway GN, Buist NR: Gyrate
atrophy of the choroid and retina. Int Ophthalmol
4:23–32, 1981. Reprinted by permission of Kluwer
Academic Publishers.

2240
Retinitis Pigmentosa and Allied Diseases

of respiratory distress.197 The value, if any, of this treatment for Stationary night blindness can be inherited by an autosomal
retinal malfunction remains to be established. dominant, autosomal recessive, or X-linked mode. Some patients
Patients who present with a widespread loss of photoreceptor can have a normal fundus appearance. Two forms, Oguchi disease
function with abnormal ERGs and symptoms of cerebral and fundus albipunctatus, have characteristic fundus abnor-
deterioration may have a cerebroretinal degeneration, grouped malities (see Fig. 177.2d and e). It is the normality or near-
under the overall heading of neuronal-ceroid lipofuscinosis, or normality of the cone system in the full-field ERGs that allows
Batten disease. This group of diseases, which are recessively separation of most stationary forms of night blindness from
inherited, is characterized by accumulation of lipopigments practically all forms of night blindness associated with the early
(lipofuscin and ceroid) in neurons and other cell types. Batten stages of RP (see Fig. 177.4).
disease can be subdivided into an infantile form (psychomotor The original classification of these diseases was based on the
deterioration by age 2 years, ataxia, microcephaly, granular family of origin (e.g., Nougaret type) or by the initial description
inclusions on conjunctival biopsy), a late-infantile form (seizures, of the ERG waveforms associated with the condition (e.g., Riggs
rapid mental deterioration in early childhood, curvilinear inclu- or Schubert–Bornschein type), or by the ophthalmologist who
sions on biopsy), a juvenile form (mental deterioration begin- first described the disease (e.g., Oguchi disease). These diseases
ning around age 6 years, bull’s-eye maculopathy, fingerprint are now being reclassified by molecular genetic analysis.207
inclusions on conjunctival biopsy), and an adult-onset form Missense mutations in three different genes encoding mem-
(seizures, slow dementia, abnormal inclusions best seen on bers of the rod phototransduction cascade have been reported to
muscle biopsy). cause dominantly inherited stationary night blindness. Specifi-

CHAPTER 177
Molecular genetic studies have revealed that the juvenile cally, the mutations are the Ala292Glu and Gly90Asp muta-
onset form of Batten disease results from defects in the CLN3 tions in the rhodopsin gene;208,209 the His258Asp mutation in
gene on chromosome 16p; the majority of patients have a 1.2-kb the beta-subunit of rod phosphodiesterase gene;210 and the
deletion in this gene on at least one chromosome.198 The gene Gly38Asp mutation in the alpha-subunit of rod transducin
encodes a novel protein of unknown function. Studies have gene.211 The latter mutation has been seen only in the descen-
suggested that the mechanism of apoptosis is involved in the dants of Jean Nougaret. Whereas the Ala292Gly rhodopsin
demise of both neurons and photoreceptors.199 No treatment mutation apparently causes a complete loss of rod function (i.e.,
exists for the retinal degeneration in this condition. Riggs type),208 the other mutations cause an incomplete loss of
Patients with olivopontocerebellar atrophy can present with a rod function.211,212
history of tremors, ataxia, and dysarthria and show findings of Some evidence exists from psychophysical and ERG testing
oculomotor impairment and retinal degeneration. This con- that the rods are constitutively active in the dark in some forms
dition is inherited by a dominant mode with variable pene- of stationary night blindness. In the rhodopsin, Gly90Asp
trance. Some patients have retinal arteriolar attenuation, a mutation, psychophysical measures with Stiles two-color incre-
diffuse disturbance of the retinal pigment epithelium, and ment threshold technique show the equivalent of light adapta-
very reduced ERGs. Some patients may show only neurologic tion in the dark owing to rhodopsin activation in darkness.208
findings and have normal fundi and normal retinal function, The ERG abnormalities in patients with the Gly38Asp muta-
whereas others have had abnormal ERGs with no diagnostic tion in the alpha-subunit of rod transducin can be simulated by
findings on fundus examination and no history of neurologic light-adapting the normal retina, compatible with the idea that
disease. Of all the known forms of spinocerebellar atrophy, if these patients have mutant rod transducin that is constitutively
there is associated photoreceptor degeneration, almost all are active in the dark.212 Evidence that patients with stationary night
due to mutations in the SCA-7 gene. In general, each succeeding blindness have rod photoreceptors despite their night blindness
generation is more severely affected and has an earlier onset is supported by fundus reflectometry studies that have shown
and increase in severity is more common if the condition normal levels of rhodopsin and normal rhodopsin kinetics.213
is inherited from the father. This condition is presently Oguchi disease is a recessively inherited form of stationary
untreatable.200–202 night blindness. Patients usually require 2–12 h to attain nor-
Other rare syndromes associated with retinal degeneration mal dark-adapted rod thresholds and show a characteristic
include Alström disease and Cockayne syndrome. Patients with change from a golden brown color of the fundus in the light-
Alström disease have RP with profound loss of vision in the first adapted state (see Fig. 177.2d) to a fundus of normal color in
decade of life and very reduced ERGs. Systemic findings include the dark-adapted state (the ‘Mizuo phenomenon’). After 1 h of
diabetes mellitus, obesity, deafness, renal failure, baldness, and dark adaptation, patients with Oguchi disease have no rod
hypogenitalism.203,204 Patients with Cockayne syndrome show b-wave in response to dim blue light flashes, a cornea-negative
extensive loss of vision by the second decade of life and have RP, response to white light, and a normal cone response to 30-Hz
dwarfism, deafness, mental deterioration, and premature aging. white flicker. Following complete dark-adaptation after 12 h,
No treatments are known for these conditions.205,206 some patients have a normal rod b-wave amplitude and normal
rod b-wave implicit time, but only in response to one or two
STATIONARY FORMS OF NIGHT flashes of light.214 Apparently, the test flash used to elicit the
BLINDNESS ERG, although relatively dim, can be intense enough to light-
adapt the rod system. Molecular genetic analyses have shown a
null mutation, Asn309(1-bp del), in the gene encoding arrestin
Key Features: Stationary Forms of Night Blindness in some patients of Japanese descent.215 Several null mutations
• Inherited by a dominant, recessive, or X-linked mode.
have also been described in the gene encoding rhodopsin kinase
• Clinical findings – normal appearing fundi or golden brown in some patients of Eastern European descent.216 Both arrestin
fundi (Oguchi disease) or white deposits around the mid- and rhodopsin kinase participate in the deactivation of photo-
periphery (fundus albipunctatus). activated rhodopsin. The lack of either arrestin or rhodopsin
• Electroretinograms – impaired rod function after 45 min of dark kinase would be expected to have the same physiologic effect,
adaptation, normal or nearly normal cone function. namely the prolongation of photoactivated rhodopsin with a
• Long-term visual prognosis relatively good compared to consequent abnormality in rod sensitivity.
retinitis pigmentosa. Fundus albipunctatus is another form of recessively inherited
stationary night blindness.217,218 Patients have yellow-white 2241
RETINA AND VITREOUS

deposits in the deep retina outside the macula (see Fig. 177.2e). axial myopia as the only finding.225 Patients with X-linked
Some develop macular degeneration in later life. Patients with congenital nyctalopia have been further subdivided into the
fundus albipunctatus have a delay in rod visual pigment and complete or incomplete form. In the complete form, the rod a-
foveal cone pigment regeneration as monitored with fundus wave is normal and the rod b-wave is absent and responses are
reflectometry.217,219 Changes in visual pigment levels parallel mediated by the cone system. This form appears to be caused
the prolonged cone and rod limbs of the dark-adaptation curve. by abnormalities in the NYX gene, whose gene product appears
Full-field ERGs have been found to be normal with respect to to be involved in ON-bipolar cell signaling.226,227 In the incom-
both cone and rod amplitudes and b-wave implicit times after plete form rod ERG responses, although reduced in amplitude,
full dark-adaptation.218 The findings in fundus albipunctatus are still measurable to dim blue light, and the cone ERG is also
suggest an abnormality in visual pigment regeneration second- subnormal. The incomplete form appears due to mutations in
ary to some abnormality in the relationship between the photo- the CACNAIF gene; patients with this condition have ERG
receptors and the pigment epithelium. The composition of the abnormalities due to malfunction of both the ON- and OFF-
yellow–white lesions and their relationship to the abnormality bipolar pathways. Whereas patients with the complete form have
in visual pigment regeneration are not known. Mutations in the moderate or high myopia, those with the incomplete form are
RDH5 gene compromising the function of 11 cis-retinol-dehydro- thought to have mild myopia or slight hyperopia.228 Whereas
genase have been found to be the cause of this condition.220,221 the complete form appears to be stationary, the long-term course
Another form of stationary night blindness is congenital of patients with the incomplete form remains to be established.
nyctalopia with myopia. This condition (i.e., Schubert– Congenital night blindness associated with a negative ERG
SECTION 10

Bornschein type) is inherited by either an X-linked or an waveform has also been described in some patients with
autosomal recessive mode.222–224 The extent of the myopia is mutations in the GRM6 gene encoding the glutamate receptor
characteristically –3.5 to –14.5 D. Patients cannot attain mGluR6. This receptor is present only in the synapses of the
normal dark-adapted rod thresholds and have fundus findings ON-cell bipolar dendrites and it mediates synaptic transmission
of myopia. Their rod ERG b-wave responses to dim blue light from rod and cone photoreceptors to this second order neuron.
are nondetectable, and they show a characteristic cornea- Patients are night blind at an early age and have acuities that are
negative response to white light in the dark-adapted state and normal or slightly reduced. In addition to reduced b-waves, ERG
normal or nearly normal cone responses to 30-Hz white flicker responses to saw-tooth flickering light indicate a reduced ON-
(see Fig. 177.4). The preservation of the a-wave from the rod response and a nearly normal OFF-response. The condition
photoreceptors with loss of the b-wave from neurons proximal appears inherited by an autosomal recessive mode and is
to the photoreceptors suggests some abnormality in intraretinal thought to be stationary or very slowly progressive.229
transmission of the response from the rod photoreceptors to The ERG abnormality in congenital nyctalopia with myopia
proximal retinal cells. ERG amplitudes are reduced in has been reported as an acquired defect in patients with
congenital nyctalopia with myopia compared with those from cutaneous malignant melanoma, who report an acute onset of
normal emmetropes; this probably occurs because of the known night blindness with selective reduction in the rod b-wave
reductions of ERG amplitude seen in patients with moderate response (Fig. 177.22).230 Development of an antibody to the

FIGURE 177.22. Full-field ERG responses from a normal subject, from the right (OD) and left (OS) eyes of a patient with malignant melanoma,
and from a patient with congenital stationary night blindness with myopia (CSNB). Stimulus onset is designated by the ‘vertical hatched lines’ in
columns 1, 2, and 3 and the ‘vertical lines’ superimposed on the responses in column 4. Two or three consecutive responses are illustrated, and
cornea-positivity is an upward deflection. The peak of the cornea-negative a-wave, generated by photoreceptors, and the peak of the cornea-
positive b-wave, generated by activity of cells proximal to the photoreceptors, are designated in the response of the normal subject to single
flashes of white light. Calibration symbol (lower right) designates 50 msec horizontally, 200 mV vertically for the top recording in column 3, and
100 mV vertically for all other tracings. Both patients lack the cornea-positive b-wave from the rod system in columns 1, 2, and 3 in contrast to
the normal, while retaining normal cone amplitudes (≥50 mV) in their responses to 30-Hz white flickering light in column 4.
Reprinted from Berson EL, Lessell S, Am J Ophthalmol, vol 106, Paraneoplastic night blindness with malignant melanoma, pp 307–311, Copyright 1988, from Elsevier
2242 Science.
Retinitis Pigmentosa and Allied Diseases

melanoma that also reacts to retinal cells with an interruption in the less astigmatic eye.25 Uncorrected myopia can be a cause
in rod signal transmission has been considered as a probable of night blindness, and therefore it is important to establish
mechanism to account for the sudden onset of night blindness that the patient has the correct prescription. Reading vision
in these patients. should be tested and lenses prescribed as needed to ensure that
Another form of congenital night blindness is that seen in the patient can read at least 8-point print. After an assessment
patients with impaired cone adaptation and elevated final rod of acuity and refractive error, a visual field should be performed
thresholds which has been termed ‘bradyopsia’.231 Patients with with either the Goldmann perimeter or the Humphrey field
this condition have mutations in the genes encoding RGS9 or analyzer. If the Goldmann perimeter is used, it is recommended
its anchor protein R9AP with consequent slow photoreceptor to test with both a small (e.g., I-4e or II-4e) and large (e.g., IV-4e
deactivation. RGS9 specifically deactivates the G-proteins or V-4e) white test light, as many patients do not have sufficient
(transducins) in the rod and cone phototransduction cascades. remaining function to detect the small light. If the Humphrey
It is anchored to photoreceptor membranes by the transmem- field analyzer is used, use of the size III or size V white target is
brane protein R9AP which enhances RGS9 activity up to advised using both the 30–2 and 60–2 programs to assess the
70-fold. Patients present in childhood with photophobia and central and the mid-peripheral visual fields. Color vision testing
have difficulty adapting to sudden changes in luminance levels should then be performed, preferably without dilation and
under photopic conditions. Visual acuity is either normal or under standard lighting conditions. The Farnsworth panel D15
moderately subnormal and can fluctuate. Patients have diffi- and the Ishihara plates are adequate for initial screening for red
culty seeing moving objects especially with low contrast despite and green color deficiencies; a tritan axis of confusion (i.e., blue

CHAPTER 177
full visual fields. Fundus findings are unremarkable. Diminu- blindness) can occur in patients with RP. After these tests,
tion in cone 30-Hz flicker ERG responses to successive flashes applanation pressures are measured; elevated pressures (>35
of light should suggest the possibility of this condition. Findings mm Hg) can result in diminished ERG amplitudes.
in patients followed for as long as 25 years suggest that this Pupils are then maximally dilated with phenylephrine hydro-
condition is stationary. chloride and cyclopentolate hydrochloride and the patient patched.
It is of interest that stationary forms of night blindness, with After 30–45 min of dark adaptation, final dark-adaptation
impaired rod function in early life but no evidence of rod cell thresholds are measured to establish whether the patient has
death, are not associated with intraretinal pigment migration or impaired rod function. After dark-adaptation testing, full-field
attenuation of the retinal vessels, whereas these latter findings ERG testing is performed. If the responses are easily detected
are characteristically seen in patients with rod cell degeneration without computer averaging, the test can be accomplished
and RP. Although the stationary or near stationary forms of within 10–15 min per eye; if computer averaging is required, the
night blindness are rare, it is important to identify these con- testing may take 20–30 min per eye. This sequence of
ditions when patients present with night blindness on a retinal evaluations helps to ensure that the patients have their dark-
basis, as the long-term prognosis in these patients is relatively adaptation and ERG testing before more extensive light
good, in contrast to the progressive retinal degeneration of rods and exposure associated with ophthalmoscopy that might light-
cones seen in patients with night blindness associated with RP. adapt the patient and thereby affect the dark-adaptation and
ERG measurements.
APPROACH TO THE PATIENT After the results of dark-adaptation and ERG testing are
obtained, patients are examined with pupils fully dilated, at
Patients with known or suspected RP or an allied disease should which time the refractive error can be confirmed by retinoscopy
be asked to provide a history of the age of onset of night blind- and glasses modified, if necessary. A slit-lamp examination and
ness, visual-field loss, and loss of visual acuity. Often symptoms direct and indirect ophthalmoscopy are performed; fine retinal
of night blindness reflect problems in adaptation under dim deposits (as, e.g., those seen in retinitis punctata albescens) and
photopic conditions, and therefore suggest cone malfunction. slight retinal arteriolar attenuation may be appreciated with
Patients with a sudden onset of night blindness, particularly direct but not indirect ophthalmoscopy. In the case of patients
after the age of 50 years, should be considered suspects for with cataracts, a retinal acuity can be obtained with a retinal
a paraneoplastic process (i.e., nonocular malignancy with potential acuity meter or equivalent instrument to compare
associated retinal malfunction), or so-called cancer-associated retinal acuity with distance acuity.
retinopathy.232–234 Difficulties in reading may reflect a general- In the case of isolate males with RP when X-linked disease is
ized loss of cone function or loss of cone function confined to suspected, the patient’s mother should be evaluated, if possible,
the macula. with both a fundus examination and ERG testing, to determine
The medical history should include information on whether she shows signs of a carrier state of X-linked disease
operations and illnesses as well as current medications and and, if affected, how much retinal function remains. Carriers of
nutritional supplements. Intestinal surgery or liver disease may X-linked RP have abnormal full-field ERGs in one or both eyes
have affected the absorption or metabolism of vitamin A. in more than 90% of cases, whereas carriers of autosomal reces-
Chronic intake of some medications (e.g., hydroxychloroquine, sive disease have normal fundi and normal ERG amplitudes.
phenothiazines) may have compromised retinal function. The Establishment of a diagnosis of X-linked disease can be helpful
medical history should include information on the status of in determining the visual prognosis of a male patient with RP,
hearing, as some 10–15% of patients with RP have a high-tone since untreated males with X-linked disease often become vir-
partial hearing loss, and some 5% have profound congenital tually blind by age 30–45 years, whereas males with autosomal
deafness. A history of polydactyly, mental retardation, or renal recessive disease often become virtually blind after age 50–60 years.
disease, if elicited, should raise the possibility of an association If the diagnosis of a common form of RP is established, adult
of these abnormalities with a retinal degeneration. A detailed patients should be advised that their condition is treatable with
family history is always warranted to determine whether other vitamin A palmitate.70–73 Before treatment is initiated, eligible
members of the family are affected with the retinal degeneration patients should have a fasting serum vitamin A level and liver
and to determine whether or not consanguinity exists in the function profile performed to be certain these values are normal
parents, as this raises the likelihood of a hereditary disease. before starting vitamin A. Information should be provided as to
The ocular examination should include a refraction; some available sources of the correct dose of vitamin A palmitate (i.e.,
50% of patients with RP have 1 or more diopters of astigmatism 15 000 IU/day for adults). (For current sources of vitamin A 2243
RETINA AND VITREOUS

palmitate see www.blindness.org). The patient should be subjective improvement without measurable change in acuity
advised that higher doses provide no additional benefit and that within 1 month. Topical dorzolamide 2% can provide benefit.
doses of 25 000 IU/day or greater are potentially toxic over the Intravitreal steroid injections have also been used for severe
long term. All patients should be advised of the possible adverse edema. The value of these treatments over the longer term for
effect of high-dose (i.e., ≥ 400 IU/day) vitamin E supplemen- such patients remains to be established.235,236
tation.70 Adult patients already taking vitamin A should be For patients under age 40 years with autosomal recessive RP,
advised to eat two 3-ounce servings of oily fish per week of serum lipoprotein electrophoresis and serum phytanic acid can
which DHA is a major constituent. Adult patients starting be assessed after fasting for 12 h if Bassen–Kornzweig syndrome
vitamin A for the first time should be encouraged to take DHA or Refsum disease is suspected. A fasting serum vitamin A level
gelcaps 600 mg twice a day for 2 years at which time they can be obtained if nutritional deficiency or intestinal malabsorp-
should stop the DHA supplement, continue to take vitamin A, tion is suspected. A serologic test for syphilis should be performed
and begin to eat two 3-ounce servings of oily fish (e.g., salmon, if this diagnosis is suspected, as RP has been associated with
tuna, mackerel, or sardines of which DHA is a major con- this disease. A fasting serum vitamin E level should be
stituent) per week. Once oily fish is begun, they should have a considered in patients with the signs of RP who also report the
fasting red blood cell DHA (RBC DHA) level 3 months after onset of ataxia in adulthood, as some success has been reported
starting consumption of oily fish (i.e., to give time for the red with vitamin E supplementation for this rare disorder.
blood cells to renew themselves) to confirm that this level is at Discussion of results of the ocular examination with the
least 4% of total RBC fatty acids. Patients should have a fasting patient, and with the patient’s family if indicated, must be
SECTION 10

serum vitamin A, liver function profile, and RBC DHA individualized for each case; but in any event, every effort
annually thereafter. Whereas vitamin A alone is thought to slow should be made to answer the patient’s questions faithfully and
the annual rate of decline of remaining retinal function by 20%, provide a clear description of not only what abnormalities exist
the combination of vitamin A plus an oily fish diet is thought but also how much function remains. In this regard, the
to slow the amount of remaining function by an additional measurement of any ERG function is an advantage, as the
40–50%.81 Precautions with regard to the safe use of vitamin A patient can be shown in positive terms how much function
are reviewed earlier in this chapter in the section on The remains. The use of computer averaging and narrow bandpass
Treatment of Retinitis Pigmentosa. filtering is particularly important, as most patients with
At the initial examination, every effort should be made to remaining vision have detectable cone ERGs with use of this
establish the diagnosis; consider the best possible optical devices specialized technology (which can detect responses as low as
to maximize remaining vision; explain the genetic implications, 0.05 mV) but will show nondetectable responses with conven-
where indicated; and offer referral to appropriate psychologic, tional full-field ERG systems (which can detect responses only
social, and community resources. In most cases, follow-up as low as 10 mV). The demonstration of remaining retinal func-
examinations are conducted every 2 years to help determine the tion has prognostic implications. For example, assuming an
course of the disease as monitored by visual acuity, visual fields, exponential rate of decline of remaining cone ERG function, the
and ERGs (computer averaged in most cases); to adjust glasses average patient with 1.3 mV at age 32 would be expected to
and provide low-vision aids, if required; and to consider whether retain some useful vision (i.e., greater than or equal to 0.05 mV)
cataract surgery is indicated. Cataract surgery is usually until age 63 without treatment or until age 83 with vitamin A
deferred in the case of RP until a time when the patient can- supplementation plus an oily fish diet. A patient with 2.6 mV at
not read with either eye. A trial with dilating drops (e.g., age 32 would be expected to retain useful vision until age 69
homatropine 2% taken at dusk) may be attempted to see without treatment or until age 89 with vitamin A and oily fish.
whether the patient can meet her or his visual needs without Knowledge of the amount of remaining cone function quan-
surgery. Cataract extraction has the greatest potential for tified by full-field ERG testing is therefore helpful in estimating
improving vision among those patients who have large enough the long-term visual prognosis.237
cataracts to impair visualization of the fundus with the It is important to impress on the patient that these con-
ophthalmoscope, detectable ERGs with computer averaging as a ditions in general progress at a very slow rate. Remarks such as
measure of some remaining retinal function, and evidence of “Your vision may last for 2 years or 30 years” should be avoided,
retinal acuity that is better than distance acuity. as the patient hears only the more severe prognosis and does
not understand that in most instances he or she may well retain
Key Features: Retinitis Pigmentosa – Other Treatment vision for 20 to 40 years or more after the initial evaluation,
Considerations particularly in the case of RP or choroideremia. It is often useful
• Cataracts – surgical removal when patient can no longer read to hear what the patient believes he or she was told previously
with the better eye. and to try to address any misconceptions. For example, some
• Cystoid macular edema – oral Diamox, Acular drops, patients believe that retinal dystrophy and degeneration are
dorzolamide drops, intravitreal steroids in some cases. ‘different diseases’ and erroneously conclude that none of the
• Reduced doses of vitamin A palmitate for children aged physicians agree on the diagnosis.
6–18 years. Literature provided to the patient at the time of the visit is
• Night deficiency – night vision pocketscope to alleviate the extremely important, as most patients cannot assimilate all the
symptom of night blindness. information given to them verbally. Printed information
• Other optical aids – closed circuit television in selected cases, describing their condition, patterns of inheritance, reasons why
avoidance of bifocals in patients with constricted visual fields. they should be followed up over time by their ophthalmologist,
the significance of molecular genetic studies, and the like
should be available for distribution in the office. Patients may
Some 10–25% of patients with RP develop cystoid macular wish to receive copies of the results of their ocular examin-
edema. In some cases the edema can be subclinical and only ations; visual displays of their ERG responses as part of this
detected with optical coherence tomography (OCT). Acetazo- report can be helpful in demonstrating to patients the amount
lamide and Acular drops have been advocated for patients with of retinal function remaining.
RP with cystoid macular edema; some given these medications When a child is detected with RP or an allied retinal degen-
2244 have shown an improvement in acuity and others have claimed eration, parents should be provided with a full written report. If
Retinitis Pigmentosa and Allied Diseases

the parents agree, then the child should be provided with sufficient
verbal information at the time of the examination to allow the
child to understand her or his own symptoms. For example, if
the child is night deficient, the child should be advised to be
careful when engaging in activities under conditions of dim
illumination. If the child has a deficiency in side vision, the
child should be advised that it may be difficult if not unsafe to
play certain sports (e.g., baseball or hockey). A child may well be
legally blind due to loss of acuity and yet still be able to read
with magnifying lenses; if this child has sufficient visual field,
he or she may well be able to continue functioning in a regular
school. It is psychologically helpful to both the child and the
parents to emphasize how much visual function remains rather
than how much has been lost and to defer conclusions about
long-term visual prognosis until the child has been examined
several times over 2-year intervals. In the case of children aged
6–18 years with the common forms of RP and normal serum
vitamin A and liver function, reduced doses of vitamin A

CHAPTER 177
palmitate adjusted for age and weight may be considered on a
trial basis with the agreement of the parents and close follow-up
by the pediatrician and ophthalmologist. As a general guideline,
for children above the fifth percentile in weight for a given age,
sex, and height, 5 000 IU/day of vitamin A palmitate may be
tried for those between ages 6 and 10 years, 10 000 IU/day for
those between 10 and 15 years of age, and 15 000 IU/day
thereafter. Sources of reduced doses of vitamin A palmitate can
be found at www.blindness.org. As noted earlier in this chapter
in the section on treatment of RP, beta-carotene is not a suitable
substitute for vitamin A palmitate in the context of this
treatment as it is not predictably converted into vitamin A.
Children should also be encouraged to eat 1–2 servings of oily
fish per week. However, it must be re-emphasized that patients
younger than age 18 were not included in our randomized trials
and therefore no formal recommendations can be made.

NIGHT VISION POCKETSCOPE


Advances in electro-optical technology have resulted in night
vision devices that allow patients with RP, choroideremia,
stationary night blindness, and allied diseases to use their cones
to function under scotopic (starlight or moonlight conditions)
or, if necessary, under dim photopic conditions that exist at
night near streetlights or in a slightly darkened room.238,239
These light-amplifying devices have been incorporated into a
monocular pocketscope or binoculars that can be used as an aid
to alleviate the symptom of night blindness. The instruments
contain bright-point-source protection so that a patient can
view a dimly illuminated scene in the presence of automobile FIGURE 177.23. Top, Model LV6015 night vision pocketscope
headlights or street lights. Figure 177.23 illustrates a hand-held adapted for patients with night blindness (top and left). The instrument
monocular pocketscope. The instrument provides a patient can also be used when held in the hand (right). Middle, Schematic
with a 40-degree field of view and gives the patient her or his diagram of a generation III night vision pocketscope. The pocketscope
best daylight vision at night in one eye. contains a light-emitting diode to provide supplementary illumination
Candidates for these devices should have a best-corrected within 3 m and is powered by two AA batteries. It weighs 13.8 oz and
vision of better than 20/200 and a central visual-field diameter is 4.5 inches long µ 2 inches wide µ 2.25 inches high. Bottom, Single-
stage image intensifier tube, 3 cm in length, contained in the night
of greater than 20° in at least one eye. Patients considering a
vision pocketscope.
monocular device should also have good mobility in daylight Top, From ITT Night Vision, Inc., Roanoke, VA. Middle and bottom, from Berson
with one eye patched. Patients who depend heavily on a far- EL: In Faye EE [ed]: Clinical Low Vision. Boston, Little, Brown, 1976.
temporal crescent for mobility are not good candidates; nor are
patients who, because of a central scotoma, cannot view the
output screen in these devices. Patients should be advised to particularly since patients can view the reading material as
assess the value of the device for their routine activities before white on a black background, thereby reducing glare. For those
making a decision as to whether or not to obtain it. who have difficulty tracking a line, a ruler or cutout window
may be of value. Patients with night deficiency should be
OTHER OPTICAL AIDS encouraged to carry a small penlight. Magnifying lenses put
into a glasses frame are also helpful. Bifocals are to be avoided
Some patients with RP have reduced acuity. A closed-circuit in patients with very constricted visual fields, as the lower
television for magnification is helpful in many instances, segment may cut the visual field such that patients find them 2245
RETINA AND VITREOUS

less than useful. In the author’s experience, field wideners have ophthalmoscope, but under age 30 years, ERG testing is indi-
had limited if any value for patients with RP and allied cated if the patient wants to be certain whether any relatives are
conditions. affected. The ERG identifies those who are affected and those
who are normal; in families with RP, patients aged 6 years and
GENETIC COUNSELING over with normal ERGs would not be expected to develop the
condition at a later time.
If RP is dominantly inherited (i.e., three consecutive genera- Recent advances with molecular genetic techniques
tions with father-to-son transmission), each affected patient has undoubtedly will facilitate genetic counseling. For example,
a one in two chance with each childbirth of having a child of some 20–25% of patients with autosomal dominant RP in the
either sex with this condition. If RP is inherited by an autosomal United States have rhodopsin gene mutations detectable
recessive mode (i.e., at least two comparably affected female through analysis of leukocyte DNA. Patients identified through
siblings or male and female siblings comparably affected, with analysis of DNA would have an ocular examination including
normal parents, or an isolate case with a family history of ERG testing to confirm the diagnosis and to determine the
consanguinity), an affected patient has about a one in 80 chance amount of remaining retinal function, as variable clinical
of marrying a carrier with this condition and the carrier has a expression at a given age is known to exist among patients with
one in two chance of passing on the abnormal gene. Therefore, the same rhodopsin gene mutation. Patients with dominant RP
the chance of a patient with an autosomal recessive form of RP due to rhodopsin Pro23His can differ by up to 1000-fold in their
having an affected child is ~1 in 160 with each childbirth (1/80 ERG amplitudes at a given age; therefore definition of severity
SECTION 10

µ 1/2). If a male has X-linked RP or choroideremia, all of his of disease cannot be determined simply by a molecular genetic
sons will be normal and all of his daughters will be carriers. If a analysis. Leukocyte DNA analysis should be considered not
woman is a carrier of X-linked RP or choroideremia, she has a only for families with a known dominant mode of transmission
50% chance of having an affected son and a 50% chance of over three consecutive generations but also for families with
having a carrier daughter with each childbirth. Patients with transmission over two consecutive generations or for isolate
isolate (simplex) RP (i.e., no known affected family members) cases with presumably normal parents. Some of these patients
can be considered to be recessive, although exceptions exist. All may have a rhodopsin gene mutation described in dominant
offspring of males or females with autosomal recessive RP are pedigrees, thereby helping to establish that the mode of
carriers of this condition; carriers enjoy normal vision but have inheritance is dominant. It is important to establish the correct
about a one in 320 chance of having an affected child with each genetic type in families with RP, not only for the sake of
childbirth (i.e., one in 80 chance of marrying a carrier and, if providing accurate genetic counseling but also as a guideline in
married to a carrier, one in four chance of having an affected establishing long-term visual prognoses of affected patients, as
child; 1/80 µ 1/4 = 1/320). In the case of Usher syndrome type patients with dominantly inherited disease generally retain
I, Usher syndrome type II, Leber congenital amaurosis, or other vision longer than those with autosomal recessive or X-linked
autosomal recessive diseases, once a couple has one affected disease.
child they have a one in four chance with each succeeding child-
birth of having another affected child. PSYCHOLOGIC AND VOCATIONAL
In families with only one affected male, RP can be inherited COUNSELING
by an autosomal recessive or X-linked mode; ERG testing and
fundus examination of the mother can help to determine Patients identified with progressive retinal degenerations often
whether she is a carrier of X-linked disease. A careful family need psychologic counseling to help adjust to the knowledge
history and examination of relatives of an affected patient with that they can expect to lose their vision over time. Initial
ERG testing can aid in establishing this mode of transmission. expressions of anger, frustration, despair, or depression are nor-
It is important to recognize that RP may skip generations and mal reactions in such patients, and they may need counseling
yet be transmitted by a dominant mode; the dominant mode in order to come to terms with their disease. Similarly, parents
with variable penetrance should be suspected in patients who of affected patients may feel profound guilt when they learn
have large cone ERGs with substantial delays in implicit time about the genetic implications of the disease and may need
under age 20 years. psychologic counseling. Patients and their family members
If a pedigree with multiple affected members is inconclusive frequently benefit from a support group where they can talk to
as to whether the disease is transmitted by a recessive or a others who have similar problems. Patients may also need
dominant mode, a digenic mode of inheritance should be con- guidance in selecting an appropriate vocation that they can
sidered. In digenic transmission, two unlinked mutations (neither pursue with their present vision and can continue to pursue in
of which results in RP) cause this disease in patients with both the event their vision fails. To this end, guidance provided by a
mutations. Affected individuals can have asymptomatic parents trained specialist can be extremely helpful. It is very important
but a 25% chance of having an affected child with each birth. that the patient be advised to return every 1–2 years to see an
The first example of digenic transmission of RP involved a muta- ophthalmologist. Patients with these diseases need periodic
tion in the peripherin/RDS gene on chromosome 6 (Leu185Pro) contact with their ophthalmologists to receive information on
and one or another mutation in the rod outer segment mem- the course of their disease, particularly if they are being treated
brane 1 (ROM1) gene on chromosome 11; both genes encode with vitamin A plus an omega-3 rich fish diet; to obtain,
proteins responsible for maintaining photoreceptor outer wherever possible, optical aids to maximize use of remaining
segment structure. The discovery of this mode of transmission vision; and in some cases, to receive psychologic support.
provides an explanation on a molecular genetic basis for the
pattern of inheritance described clinically as dominant RP with SUNGLASSES
reduced or incomplete penetrance.7
Patients often state that their children have had normal General agreement exists that patients with hereditary degen-
routine eye examinations and therefore must not have RP or an erations of the retina should avoid excessive exposure to bright
allied disease. Some young people may have these diseases and sunlight outdoors until more is known about whether light
yet appear normal on routine ocular examination. By age stress will modify the course of these conditions.240,241 Two
2246 30 years, more than 90% of patients can be diagnosed with the patients with RP who wore an opaque scleral contact lens over
Retinitis Pigmentosa and Allied Diseases

one eye for 8–10 h per day for 5 years showed comparable example a deficiency in the RPE65 gene results in loss of the
degeneration in both eyes, suggesting that this type of protec- isomerase needed for production of 11-cis-retinal in the retinal
tion from light did not alter the course of their disease.241 Some pigment epithelium; this deficiency leads to a form of Leber
patients have expressed a preference for orange photochromic congential amaurosis or early onset recessive RP. Gene therapy
sunglasses (e.g., Corning CPF 550) with tinted side shields or has been achieved in animal models of this condition140,246,247
dark amber sunglasses (e.g., NoIR brand worn over an existing and trials of human therapy have begun. In dominantly
prescription) with tinted side shields. However, no particular inherited mutations or so-called ‘gain of function mutations’,
type of sunglasses has been shown to alter the course of retinal the encoded amino acid sequence is altered and can result in
degeneration. Until more is known, patients should be advised accumulation of abnormal proteins leading to toxicity in retinal
to select sunglasses for outdoor use that provide maximal com- cells. In this instance treatment is aimed at eliminating the
fort without compromising vision. mutant gene with the expectation that the normal copy of the
gene will encode sufficient normal protein. Research is being
SOME FUTURE DIRECTIONS WITH done involving ribosome-based or RNAi-based therapy to reduce
REGARD TO THERAPY expression or inactivate specific abnormal alleles.248 With appro-
priate preliminary data continued study of nutritional interven-
tions is also warranted.
Key Features: Retinitis Pigmentosa – Some Future Other strategies to promote photoreceptor survival include
Directions that May Lead to Treatments attempts to interfere with apoptosis and thereby prevent

CHAPTER 177
• Gene therapy for early stages of Leber congenital amaurosis. photoreceptor degeneration or use of a calcium channel blocker
• Neuroprotection with intravitreal administration of ciliary to counter the effects of cyclic GMP elevation that occurs in
neurotrophic factor. some degeneration with consequent opening of cGMP-gated
• Treatment with channel blockers or drugs that interfere with channels. In the former case some success has been achieved
apoptosis. with animal models249,250 but none so far in humans. In the
• Microchip implant technology. latter case an initial claim of success with a calcium channel
• Stem cell therapy. blocker in an rd mouse model of retinal degeneration251 was not
• Risk factor analyses of genetically defined populations confirmed by further studies of this channel blocker, namely
followed over time may reveal additional ameliorating factors D-cis-diltiazem, in this and other animal models.252–254 Some
with possible implications for treatment. research has been done with embryonic stem cells in mice and
rats that were found to integrate into the host retina with
Some preliminary evidence exists that neuroprotection with a reported protection of host retinal neurons.255,256 It is not
ciliary neurotrophic growth factor (CNTF) will substantially known whether stem cells will have clinical applications in the
slow the course of retinal degeneration in animal models of RP. treatment of these diseases.
A phase I study in humans with RP that used encapsulated cell ERG testing, using computer averaging and narrow bandpass
technology with intravitreal sustained release of CNTF showed filtering, provides a basis for objectively monitoring the course
that CNTF could be well-tolerated over 6 months with a few of retinal degeneration in most patients with RP and allied
patients reporting improved vision.242 This has set the stage for diseases. Variability of clinical severity at a given age among
a phase II–III study. Preliminary work has also been done with patients with the same gene defects suggests that a modifier
implantation of light sensitive microchips on or under the retina gene or some factor(s) other than the gene defects alone may be
in patients with advanced RP; it is not yet established whether involved in the clinical expression of some forms of these
patients can achieve useful vision with these devices.243,244 diseases.257 Evaluation of subgroups of patients with known
Retinal transplantation of photoreceptor cells under the retina gene defects prospectively over a period of years may reveal
has also been advocated, but no convincing data exists that the ameliorating or aggravating factors with possible implications
transplanted cells form viable synaptic connections with the for therapy. Collaborations between scientists and clinicians
inner retina.245 should enhance the likelihood of developing new treatments for
Increased knowledge of the gene abnormalities and these diseases.258
biochemical defects involved in these conditions provide new
opportunities to discover additional therapies for hereditary
retinal diseases. In the case of recessively inherited mutations ACKNOWLEDGMENTS
which usually result in ‘loss of function’, patients typically develop This work is supported in part by the National Eye Institute, EY00169,
disease due to loss of a critical protein, and gene replacement Bethesda, MD and in part by the Foundation Fighting Blindness, Owings
therapy could result in restoration of the missing protein. For Mills, MD.

REFERENCES
1. Ammann F, Klein D, Franceschetti A: 5. Bundy S, Crews SJ: Wishes of patients mitochondrial MTTS2 gene. Am J Hum
Genetic and epidemiological investigation with retinitis pigmentosa concerning Genet 1999; 64:971–85.
of pigmentary degeneration of the retina genetic counseling. J Med Genet 1982; 9. Rivolta C, Berson EL, Dryja TP: Paternal
and allied disorders in Switzerland. 19:317–318. uniparental heterodisomy with partial
J Neurol Sci 1965; 2:183–196. 6. Bunker CH, Berson EL, Bromley WC, et al: isodisomy of chromosome 1 in a patient
2. Boughman JA, Conneally PM, Nance WE: Prevalence of retinitis pigmentosa in Maine. with retinitis pigmentosa without hearing
Population genetic studies of retinitis Am J Ophthalmol 1984; 97:357–365. loss and a missense mutation in the Usher
pigmentosa. Am J Hum Genet 1980; 7. Kajiwara K, Berson EL, Dryja TP: Digenic syndrome type II gene USH2A. Arch
32:223–235. retinitis pigmentosa due to mutations at the Ophthalmol 2002; 120:1566–1571.
3. Hu DN: Genetic aspects of retinitis unlinked peripherin/RDS and ROM1 loci. 10. Thompson DA, McHenry CL, Li Y, et al:
pigmentosa in China. Am J Med Genet Science 1994; 264:1604–1608. Retinal dystrophy due to paternal isodisomy
1982; 12:51–58. 8. Mansergh FC, Millington-Ward S, Kennan for chromosome 1 or chromosome 2, with
4. Jay M: On the hereditary of retinitis A, et al: Retinitis pigmentosa and homoallelism for mutations in RPE65 or
pigmentosa. Br J Ophthalmol 1982; progressive sensorineural hearing loss MERTK, respectively. Am J Hum Genet
66:405–416. caused by a C12258A mutation in the 2002; 70:224–229.
2247
RETINA AND VITREOUS

11. Berson EL: Visual function testing: clinical retinitis pigmentosa and allied diseases. rhodopsin gene defect (Pro23His). Arch
correlations. In: Tasman W, Jaeger E, eds. Ophthalm Genet 2005; 26:91–100. Ophthalmol 1991; 109:92–101.
Foundations of clinical ophthalmology. 31. Karpe G: The basis of clinical 51. Berson EL, Rosner B, Sandberg MA, et al:
Philadelphia: J.B. Lippincott; 1991:1–10; electroretinography. Acta Ophthalmol 1945; Ocular findings in patients with autosomal
reprinted in J Clin Neurophys 11:472–481, 23(Suppl):1–118. dominant retinitis pigmentosa and
1994; updated Vol. 2, Chapter 14. 32. Goodman G, Gunkel RD: Familial rhodopsin, proline-347-leucine. Am J
Hagerstown, MD: Lippincott Williams & electroretinographic and adaptometric Ophthalmol 1991; 111:614–623.
Wilkins; 2001: 1–12. studies in retinitis pigmentosa. Am J 52. Berson EL, Rosner B, Weigel-DiFranco C,
12. Donders FC: Beiträge zur pathologischen Ophthalmol 1958; 46:142–172. et al: Disease progression in patients with
Anatomie des Auges. Graefes Arch Clin 33. Gouras P, Carr RE: Electrophysiological dominant retinitis pigmentosa and
Exp Ophthalmol 1855; 1:139; 1857; studies in early retinitis pigmentosa. Arch rhodopsin mutations. Invest Ophthalmol Vis
2:106–118. Ophthalmol 1964; 72:104–110. Sci 2002; 43:3027–3036.
13. Leber T: Die Pigmentdegeneration der 34. Berson EL, Gouras P, Gunkel RD: Rod 53. Hargrave PA, O’Brien PJ: Speculations on
Netzhaut und mit ihr verwandte responses in retinitis pigmentosa, dominantly the molecular basis of retinal degeneration
Erkrankungen. In: Graefe A, Saemisch T, inherited. Arch Ophthalmol 1968; 80:58–67. in retinitis pigmentosa. In: Anderson RE,
Hess C, eds. Handbuch der Gesamten 35. Berson EL, Gouras P, Gunkel RD, et al: Hollyfield JG, LaVail MM, eds. Retinal
Augenheilkunde, 2nd edn. Leipzig: Dominant retinitis pigmentosa with reduced degenerations. Boca Raton, FL; CRC;
W Engelmann; 1916:1076–1225. penetrance. Arch Ophthalmol 1969; 1991:517–518.
14. Bell J: Retinitis pigmentosa and allied 81:226–234. 54. Farrar GJ, Kenna P, Jordan S, et al: A
diseases of the eye. In: Pearson K, ed. 36. Berson EL, Gouras P, Hoff M: Temporal three-base-pair deletion in the peripherin-
Treasury of human inheritance. London: aspects of the electroretinogram. Arch RDS gene in one form of retinitis
SECTION 10

Cambridge University; 1922; 2:1–28. Ophthalmol 1969; 81:207–214. pigmentosa. Nature 1991; 354:478–480.
15. Franceschetti A, François J, Babel J: 37. Berson EL: Retinitis pigmentosa and allied 55. Kajiwara K, Hahn L, Mukai S, et al: Mutations
Les Héredodégenerescences Chorio- diseases: Electrophysiologic findings. Trans in the human retinal degeneration slow
rétiniennes. Paris: Masson; 1963. Am Acad Ophthalmol Otolaryngol 1976; gene in autosomal dominant retinitis
16. Duke-Elder S, Dobrie JH: Diseases of the 81:659–666. pigmentosa. Nature 1991; 354:480–483.
retina, Vol. 10. In: Duke-Elder S, ed. 38. Berson EL: Electroretinographic findings in 56. Molday RS: Peripherin/rds and rom-1:
System of ophthalmology. St. Louis: CV retinitis pigmentosa. Jpn J Ophthalmol Molecular properties and role in
Mosby; 1967. 1987; 31:327–348. photoreceptor cell degeneration. In:
17. Marmor MF, Aguirre G, Arden G, et al: 39. Birch DG, Sandberg MA: Dependence of Osborne N, Chader G, eds. Progress in
Retinitis pigmentosa, a symposium on cone b-wave implicit time on rod amplitude retinal and eye research. Oxford:
terminology and methods of examination. in retinitis pigmentosa. Vision Res 1987; Pergamon; 1994:271–299.
Ophthalmology 1983; 90:126–131. 27:1105–1112. 57. Weleber RG, Carr RE, Murphey WH, et al:
18. Pruett RC: Retinitis pigmentosa: Clinical 40. Sandberg MA, Berson EL, Effron MH: Phenotypic variation including retinitis
observations and correlations. Trans Am Rod-cone interaction in the distal human pigmentosa, pattern dystrophy, and fundus
Ophthalmol Soc 1983; 81:693–735. retina. Science 1981; 212:829–831. flavimaculatus in a single family with a
19. Heckenlively JR: Retinitis pigmentosa. 41. Berson EL: Retinitis pigmentosa, the deletion of codon 153 or 154 of the
Philadelphia: JB Lippincott; 1988. electroretinogram, and Mendel’s laws. peripherin/RDS gene. Arch Ophthalmol
20. Berson EL. Retinitis pigmentosa. The Trans Pa Acad Ophthalmol Otolaryngol 1993; 111:1531–1542.
Friedenwald lecture. Invest Ophthalmol Vis 1973; 26:109–113. 58. Fossarello M, Bertini C, Galantuomo MS,
Sci 1993; 34:1659–1676. 42. Berson EL, Rosen JB, Simonoff EA: et al: Deletion in the peripherin/RDS gene
21. Heckenlively JR: The frequency of posterior Electroretinographic testing as an aid in in two unrelated Sardinian families with
subcapsular cataract in the hereditary detection of carriers of X-chromosome- autosomal dominant butterfly-shaped
retinal degenerations. Am J Ophthalmol linked retinitis pigmentosa. Am J macular dystrophy. Arch Ophthalmol 1996;
1982; 93:733–738. Ophthalmol 1979; 87:460–468. 114:448–456.
22. Fetkenhour CL, Choromokos E, Weinstein 43. Fishman GA, Weinberg AB, McMahon TT: 59. Nakazawa M, Kikawa E, Chida Y, et al:
J, et al: Cystoid macular edema in retinitis X-linked recessive retinitis pigmentosa: Autosomal dominant cone-rod dystrophy
pigmentosa. Trans Am Acad Ophthalmol Clinical characteristics of carriers. Arch associated with mutations in codon 244
Otolaryngol 1977; 83:515–521. Ophthalmol 1986; 104:1329–1335. (Asn244His) and codon 184 (Tyr184Ser) of
23. Fishman GA, Maggiero JM, Fishman M: 44. Berson EL, Simonoff EA: Dominant retinitis the peripherin RDS gene. Arch Ophthalmol
Foveal lesions seen in retinitis pigmentosa. pigmentosa with reduced penetrance: 1996; 114:72–78.
Arch Ophthalmol 1977; 95:1993–1996. Further studies of the electroretinogram. 60. Apfelstedt-Sylla E, Theischen M, Rüther K,
24. Sieving PA, Fishman GA: Refractive errors Arch Ophthalmol 1979; 97:1286–1291. et al: Extensive intrafamilial and interfamilial
of retinitis pigmentosa patients. Br J 45. Berson EL, Sandberg MA, Rosner B, et al: phenotypic variation among patients with
Ophthalmol 1978; 52:625–633. Natural course of retinitis pigmentosa over autosomal dominant retinal dystrophy and
25. Berson EL, Rosner B, Simonoff EA: Risk a three-year interval. Am J Ophthalmol mutations in the human RDS/peripherin
factors for genetic typing and detection in 1985; 99:240–251. gene. Br J Ophthalmol 1995; 79:28–34.
retinitis pigmentosa. Am J Ophthalmol 46. Andréasson SOL, Sandberg MA, Berson 61. Hartong D, Berson EL, Dryja TP: Retinitis
1980; 89:763–775. EL: Narrow-band filtering for monitoring pigmentosa. Lancet 2006; 368:1795–1809.
26. Kolb H, Gouras P: Electron microscopic low-amplitude cone electroretinograms in 62. Biro I: Therapeutic experiments in cases of
observations of human retinitis pigmentosa retinitis pigmentosa. Am J Ophthalmol retinitis pigmentosa. Br J Ophthalmol 1939;
dominantly inherited. Invest Ophthalmol Vis 1988; 105:500–503. 23:332–342.
Sci 1974; 13:487–498. 47. Sandberg MA, Baruzzi CM, Hanson AH III, 63. Gordon DM: The treatment of retinitis
27. Szamier RB, Berson EL: Retinal et al: Rod ERG diurnal rhythm in some pigmentosa with special reference to the
ultrastructure in advanced retinitis patients with dominant retinitis pigmentosa. Filatov method. Am J Ophthalmol 1947;
pigmentosa. Invest Ophthalmol Vis Sci Invest Ophthalmol Vis Sci 1988; 29:494–498. 30:565–580.
1977; 16:947–962. 48. Dryja TP, McGee TL, Reichel E, et al: A 64. Campbell DA, Harrison R, Tonks EL:
28. Szamier RB, Berson EL, Klein R, et al: Sex- point mutation of the rhodopsin gene in Retinitis pigmentosa: Vitamin A serum
linked retinitis pigmentosa: ultrastructure of one form of retinitis pigmentosa. Nature levels in relation to clinical findings. Exp
photoreceptors and pigment epithelium. 1990; 343:364–366. Eye Res 1964; 3:412–426.
Invest Ophthalmol Vis Sci 1979; 49. Dryja TP, McGee TL, Hahn LB, et al: 65. Chatzinoff A, Nelson E, Stahl N, et al:
18:145–160. Mutations within the rhodopsin gene in Eleven-CIS vitamin A in the treatment of
29. Bunt-Milam AH, Kalina RE, Pagon RA: patients with autosomal dominant retinitis retinitis pigmentosa: A negative study. Arch
Clinical-ultrastructural study of a retinal pigmentosa. N Engl J Med 1990; Ophthalmol 1968; 80:417–419.
dystrophy. Invest Ophthalmol Vis Sci 1983; 323:1302–1307. 66. Bergsma DR, Wolf ML: A therapeutic trial of
24:458–469. 50. Berson EL, Rosner B, Sandberg MA, et al: vitamin A in patients with pigmentary retinal
30. Ben-Arie-Weintrob Y, Berson EL, Dryja TP: Ocular findings in patients with autosomal degeneration: A negative study. In: Landers
2248 Histopathologic-genotypic correlations in dominant retinitis pigmentosa and a MA, Wolbarsht ML, Laties AM, et al, eds.
Retinitis Pigmentosa and Allied Diseases

Retinitis pigmentosa. New York: Plenum; 85. Jampel RS, Falls HF: Atypical retinitis 103. Klenk E, Kahlke W: Über das Vorkommen
1976; 197–209. pigmentosa, acanthocytosis and der 3,7,11,15-tetramethylhexadecansäure
67. Katznelson LA, Khoroshilova-Maslova IP, heredodegenerative neuromuscular (Phytansäure) in den Cholesterinestern und
Eliseyeva RF: A new method of treatment disease. Arch Ophthalmol 1956; anderen Lipoidfraktionen der Organe bei
of retinitis pigmentosa/pigmentary 59:818–820. einem Krankheitsfall unbekannter Genese:
abiotrophy. Ann Ophthalmol 1990; 86. Salt HB, Wolff OH, Lloyd JK, et al: On Verdacht auf Heredopathia atactica
22:167–172. having no betalipoprotein. A syndrome polyneuritiformis (Refsum syndrome).
68. Duke-Elder S, Dobrie JH: System of comprising A-beta-lipoproteinemia, Hoppe Seylers Z Physiol Chem 1963;
ophthalmology. In: Duke-Elder S, ed. acanthocytosis and steatorrhea. Lancet 333:133–139.
Diseases of the retina. St. Louis: CV 1960; 2:325–329. 104. Berson EL: Nutrition and retinal
Mosby; 1967; 10:574–622. 87. Druez G, Lamy M, Frézal J, et al: degenerations: Vitamin A, taurine, ornithine
69. Berson EL, Remulla JFC, Rosner B, et al: L’acanthocytose: Ses rapports avec and phytanic acid. Retina 1982; 2:236–255.
Evaluation of patients with retinitis l’absence congénitale de

You might also like