Вы находитесь на странице: 1из 271

Vitreous Microsurgery

FIFTH EDITION
Vitreous Microsurgery
FIFTH EDITION

Steve Charles, MD
Vitreoretinal Surgeon and Founder
Charles Retina Institute
Clinical Professor of Ophthalmology
University of Tennessee
Memphis, Tennessee
Adjunct Professor of Ophthalmology
Columbia College of Physicians and Surgeons
New York City, New York

Jorge Calzada, MD
Vitreoretinal Surgeon
Charles Retina Institute
Clinical Instructor in Ophthalmology
University of Tennessee
Memphis, Tennessee

Byron Wood, MS
Chief Photographer and Director of Graphics
Charles Retina Institute
Memphis, Tennessee
Senior Executive Editor: Jonathan W. Pine, Jr.
Senior Product Manager: Emilie Moyer
Senior Manufacturing Manager: Benjamin Rivera
Marketing Manager: Lisa Lawrence
Design Coordinator: Joan Wendt
Production Service: SPi Technologies

© 2011 by LIPPINCOTT WILLIAMS & WILKINS, a WOLTERS KLUWER business


Two Commerce Square
2001 Market Street
Philadelphia, PA 19103 USA
LWW.com

All rights reserved. This book is protected by copyright. No part of this book may be reproduced in any form by any means, including
photocopying, or utilized by any information storage and retrieval system without written permission from the copyright owner, except for
brief quotations embodied in critical articles and reviews. Materials appearing in this book prepared by individuals as part of their official
duties as U.S. government employees are not covered by the above-mentioned copyright.

Printed in China

Library of Congress Cataloging-in-Publication Data


Charles, Steve.
Vitreous microsurgery / Steve Charles, Jorge Calzada, Byron Wood. —5th ed.
p. ; cm.
Includes bibliographical references and index.
Summary: “Written by a pioneering leader in the development of vitreoretinal surgical techniques and instruments, Vitreous Microsur-
gery is a comprehensive how-to guide to all vitreoretinal procedures. This thoroughly updated Fifth Edition describes many new techniques
and refinements of established procedures and includes new chapters and new illustrations. More than 170 three-dimensional full-color
illustrations—many by the Charles Retina Institute’s resident medical artist, Byron Wood—enable surgeons to clearly visualize the tech-
niques. The focus of the text is on the decision making process a surgeon goes through in evaluating the best course of treatment for his/her
patient undergoing vitreous surgery. The book describes in detail clinically proven methods of managing the anterior and posterior segment
vitreous surgery patient in a systematic manner. The text is organized in a building block approach with general methodology preceding its
application to specific disease states. The book stresses algorithms for intra-operative decision making, relying on knowledge of physical
principles and performed in the order of ascending risk. A companion website included with purchase offers the fully searchable text and
an online image bank”—Provided by publisher.
ISBN 978-1-60831-503-1 (hardback)
1. Vitreous body—Surgery. 2. Microsurgery. I. Calzada, Jorge. II. Wood, Byron. III. Title.
[DNLM: 1. Vitreous Body—surgery. 2. Eye Diseases—surgery. 3. Microsurgery—methods. WW 250 C477v 2011]
RE501.C48 2011
617.7’46059—dc22
2010018076

Care has been taken to confirm the accuracy of the information presented and to describe generally accepted practices. However, the
authors, editors, and publisher are not responsible for errors or omissions or for any consequences from application of the information in
this book and make no warranty, expressed or implied, with respect to the currency, completeness, or accuracy of the contents of the pub-
lication. Application of the information in a particular situation remains the professional responsibility of the practitioner.
The authors, editors, and publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accor-
dance with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government
regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert
for each drug for any change in indications and dosage and for added warnings and precautions. This is particularly important when the
recommended agent is a new or infrequently employed drug.
Some drugs and medical devices presented in the publication have Food and Drug Administration (FDA) clearance for limited use in
restricted research settings. It is the responsibility of the health care provider to ascertain the FDA status of each drug or device planned for
use in their clinical practice.

To purchase additional copies of this book, call our customer service department at (800) 638-3030 or fax orders to (301) 223-2320. Inter-
national customers should call (301) 223-2300.

Visit Lippincott Williams & Wilkins on the Internet: at LWW.com. Lippincott Williams & Wilkins customer service representatives are
available from 8:30 am to 6 pm, EST.
10 9 8 7 6 5 4 3 2 1
The Fifth Edition is dedicated to the quest to
better serve humankind.
Preface

In keeping with the approach taken with perspective in collaboration with Gary Fanning, MD, and
previous editions, the fifth edition is designed Jay Mattingly, MD, leading experts on this subject.
as a technology book rather than an academic Kenneth Fung, MD, MBA made a substantial contribu-
analysis of the literature, a statistical review of tion to the chapter on retinal vein occlusions.
outcomes, or a series of clinical cases and images. This edi- The retinopathy of prematurity chapter has been com-
tion is intended to be fine grained with emphasis on every pletely rewritten to address new examination, laser guide-
nuance of surgical technique and tool technology. lines, anti-VEGF therapy, and changing, more conservative
Surgical and medical retina strategies have under- indications for surgery.
gone significant paradigm shifts since the fourth edition All chapters have been updated, and new references
was written. The fifth edition includes new chapters on have been included. All illustrations have been updated; the
the Constellation vitrectomy system, uveitis, retinal com- majority of illustrations have been updated to a 25-gauge
plications of permanent keratoprosthesis, and anti-VEGF approach instead of a 20-gauge approach, and many new
therapy. illustrations have been included.
Many techniques and parameters unique to 25-gauge It is planned that this text will go forward with an update
sutureless vitrectomy are discussed in detail. The authors approximately every 5 years with my able coauthors, associates,
use 25-gauge sutureless technique for all cases, and micro- and great friends Byron Wood, MS and Jorge Calzada, MD.
incisional 23-, 25-, or 27-gauge vitrectomy is growing in
popularity on a worldwide basis. Steve Charles, MD
The section on anesthesia for vitreoretinal surgery
has been completely rewritten from an anesthesiologist’s

vii
Acknowledgm ents

This book is made possible because Lippincott because of his input. It is a great pleasure to work with great
Williams & Wilkins invited us to write a fifth friends who are dedicated to producing the highest quality
edition based, in part, on extensive sales of the work.
fourth edition as well as Chinese, Portuguese, I would also like to thank my wonderful, loyal, and
Turkish, and planned Spanish editions. I would like to thank hardworking office and OR staff who provided clinical sup-
the many surgeons, libraries, and universities worldwide port, allowing me to perform over 700 vitrectomies per year
that purchased the previous edition. We are very fortunate for the last 30 years, which ultimately drove the concepts
to have worked with Jonathan Pine and Emily Moyer of described in this book.
Lippincott Williams & Wilkins to produce this larger and I would like to thank Alcon Laboratories for working
improved textbook. with me over the years to develop most of the wonderful
I would like to especially thank my incredible coau- tools and systems described in this text. The engineers, man-
thors, Byron Wood, MS and Jorge Calzada, MD. Byron Wood agers, and craftsmen have enabled surgeons to provide the
has created the illustrations, as he has for four previous edi- best possible care to the patients.
tions; these are even more detailed, anatomically and surgi- I would like to thank my three amazing daughters—
cally accurate, and beautiful to view as art. Jordi Calzada has Kelli Ross, MD, Kerri Charles, MD, JD, and Marci Charles—
done a superb job of restructuring all references as well as for their love and their patience with their often preoccupied
adding new ones. In addition, many concepts were refined father.

viii
Contents

Preface vii Chapter 8


Acknowledgm ents viii Combined Phacoemulsification Vitrectomy
Procedures 98
SECTION I Chapter 9
Pre surg ical Evaluat io n an d 25-Gauge Vitrectomy 103
Im ag in g 1 Chapter 10
Chapter 1 20-Gauge Sutured Wound Construction
Presurgical Evaluation 2 and Closure 112

SECTION II SECTION III

Surg ical Te ch n o lo g y an d In -Offi ce Pro ce d ure s 117

Te ch n iq ue s 17 Chapter 11
Laser Photocoagulation 118
Chapter 2
Surgical Systems, Tools, and Operating Chapter 12
Room 18 Pneumatic Retinopexy and Office
Fluid-Gas Exchange 121
Chapter 3
Constellation Architecture 32
SECTION IV
Chapter 4
Visualization and Illumination 37 Dise ase -Sp e cifi c
Man ag e m e n t 125
Chapter 5
Anesthesia and Operating Room Chapter 13
Efficiency 40 Prophylaxis of Retinal Breaks and Scleral
Buckling 126
Chapter 6
General Posterior Segment Techniques 45 Chapter 14
Vitrectomy for Retinal Detachment 131
Chapter 7
Vitrectomy Techniques and Technology Chapter 15
for Anterior Segment Problems 76 Giant Breaks 138

ix
x Contents

Chapter 16 Chapter 26
Proliferative Vitreoretinopathy 144 Retinopathy of Prematurity and Pediatric
25-Gauge Vitrectomy 225
Chapter 17
Epimacular Membranes and Vitreomacular Chapter 27
Traction Syndrome 157 Pediatric Traction Retinal Detachments 231

Chapter 18 Chapter 28
Macular Holes 164 Management of Vitreoretinal
Complications Associated with
Chapter 19 Keratoprosthesis 236
Diabetic Retinopathy 171
Chapter 29
Chapter 20 Inadvertent Penetration of the Eye 239
Venous Occlusive Diseases 189
Chapter 30
Chapter 21 Management of Suprachoroidal
Treatment of Choroidal Neovascular Hemorrhage 241
Membranes 193
Chapter 31
Chapter 22 Complications of Vitreoretinal Surgery 244
Intravitreal Injection Technique 204
Chapter 32
Chapter 23 Surgical Self-Education 249
Trauma 205
Chapter 24 Index 253
Endophthalmitis 216

Chapter 25
Surgical Management of the Uveitis
Patient 221
SEC TIO N I
Pre surg ical Evaluat io n an d Im ag in g

1
C H AP TER
1
PRESURGICAL EVALUATION

Vitreous surgery has been applied successfully used, the requirement for postoperative examinations, and
to a wide spectrum of ocular diseases. The com- limitations on activity.
plex set of biologic, systemic risk, technique, The surgeon must take personal responsibility for obtain-
and technology considerations confronting the ing financial support for the economically disadvantaged
surgeon creates a difficult decision-making environment. patient and act as a consumer advocate concerning facility
Rather than emphasizing a search for good prognosis or easy fees. It is unconscionable to turn away a patient or permit the
cases, the goal must be to avoid bilateral or irreversible visual facility to do so because of the patient’s financial status.
disability. Many potential vitreous surgery patients have pro- It is the surgeon’s responsibility to organize the schedul-
found socioeconomic problems as a result of blindness and/ ing process so that the preoperative assessment or logistical/
or systemic disease. A significant attempt to understand the bureaucratic delay does not affect the prognosis. Waiting
patient’s medical and socioeconomic situation is essential to lists are highly inappropriate for patients with significant
effective and compassionate health care delivery. visual problems. If surgery is performed within a few days of
Since the introduction of the first edition of this book, the original examination, the surgeon will be more acutely
the scope of vitreous microsurgery has expanded greatly. aware of significant aspects of the patient’s history, findings,
Whereas vitrectomy surgery was initially considered only and imaging during surgery.
on the most advanced and complicated clinical situations,
we can safely perform 25-gauge vitrectomy with minimal
discomfort to the patient and, most importantly, with low SYSTEMIC FACTORS
surgical complication rates. This allows the use of vitrectomy
for epimacular membranes (EMMs) with relatively good Ag e
preoperative visual acuity, relieving patients’ symptoms and Age is a consideration at both extremes of the timeline. While
improving functional outcomes in ways that would not have age cannot be used as an arbitrary criterion, it is clear that
been possible in decades past. both the neonate with severe pulmonary problems and the
The surgeon must examine both eyes personally to elderly patient with multisystem systemic disease have higher
reach the best clinical decision. The surgeon must develop anesthesia risks. Surgery on the premature infant can be
an open, direct, and warm relationship with the patient delayed until pulmonary function and body weight increase,
before surgery can be considered. It is probably better to dis- allowing safer anesthesia. Most vitreoretinal surgery today is
cuss specific statistical results of surgery on similar patients performed under local anesthesia with anesthesia monitoring,
rather than to discuss the details of a specific methodology. but children and certain adults require general anesthesia.
Rather than using qualitative words such as “excellent” or Sensory deprivation from poor vision may contribute to
“poor,” using a percentage success rate is a more meaning- disorientation and agitation in certain elderly patients. Uni-
ful way to communicate. The frequency of the most impor- lateral visual loss is well tolerated in many elderly patients
tant complications should be mentioned as well as the range because of less demanding occupational and recreational
and mean of postoperative vision in functional and numeric needs. While it is the surgeon’s obligation to determine
terms. All available family members and the surgeon’s sec- ocular status and needs, medical consultants and anesthesia
retary, technician, assistant, or nurse should witness this team can help determine and manage systemic risk.
conversation. A complex, legalistic permission document is
far less important than meaningful, compassionate, under-
Diab e t e s
standable, personal communication with the patient. The
patient should be informed about postoperative positioning, As diabetes is common in vitrectomy patients, the vitreous
pain, medications, operating time, type of anesthesia to be surgeon must be quite familiar with the complications of this

2
CHAPTER 1 ■ Presurgical Evaluation 3

multisystem disease. An individualized approach is superior OCULAR FACTORS


to arbitrary surgical criteria. The presence of a systemic infec-
tion is a contraindication to vitrectomy. All ocular conditions Examination of the vitrectomy patient embodies the same
except endophthalmitis and severe glaucoma can wait until a steps, but with different emphasis, that required for other
systemic infection has been treated. ophthalmic specialties. A problem-oriented approach with
End-stage renal disease decreases life span and creates emphasis on specific factors that have a high impact on deci-
difficult socioeconomic as well as medical problems. The sion making is more effective than a nonstructured, “routine”
stable transplant patient can undergo successful vitrectomy, examination.
although immunosuppressant drugs create increased infection
risk, especially with general anesthesia. The dialysis patient
has increased risk with general anesthesia risk and may have Visual Fun ct io n Te st in g
increased bleeding with vitrectomy. While surgical conser-
vatism is indicated for patients with end-stage renal disease, Visual function testing can be the most difficult component
many carefully selected patients can benefit from vitrectomy. of previtrectomy evaluation unless approached properly.
Careful, precise measurement of visual acuity is more rel-
evant than an array of laboratory tests.
Card io p ulm o n ary Dise ase
Specific guidelines in the area of cardiopulmonary disease
Visual Acuit y
again must be replaced by an individualized approach with
reliance upon medical and anesthesia consultations. Although The inherent subjectivity of visual acuity testing demands
local anesthesia is safer than general, there remains some random presentation of stimuli, multiple repetitions, and
risk associated with local anesthesia, pointing to the need sophisticated examiners. Total coverage of the other eye with
for careful medical assessment and a monitored approach by the patient’s palm, not fingers, pressed against the bridge of
an anesthesiologist or a certified registered nurse anesthetist the nose is essential to determine light perception. Specifi-
(CRNA) with anesthesiology supervision. Pulse oximetry, cally, the examiner cannot ask, “Can you see the light?” but
EKG, continuous blood pressure monitoring, and frequent rather should say, “Tell me when the light goes on and off.”
blood glucose assessments are mandatory. The patient must describe the direction of randomly pre-
The recent trend of vitrectomy surgery moving from sented motion when testing for “hand motion.” Multiple
the hospital setting, where subspecialized consultants are repetitions increase reliability because of problems stem-
readily available, to the ambulatory surgery center requires ming from patient guessing. The patient’s neurological/psy-
the surgeon to be aware of the anesthesia risk of each chological status and educational level may interfere with
patient. We feel that high-risk patients require hospital- accurate testing.
based surgeries. The light perception determination is of particular sig-
nificance because patients without light perception should
never have vitrectomy unless it is clearly understood that the
Ob e sit y an d Ob st ruct ive Sle e p Ap n e a
procedure is to prevent the spread of infection or, in rare
Syn d ro m e
instances, for cosmetic preservation of the globe. Dense
A major practical problem that has become more prevalent opaque media requires a bright testing light such as that
in the recent years is morbid obesity and associated obstruc- from an indirect ophthalmoscope at 7.5 volts.
tive sleep apnea. These patients require surgical tables that The finger-counting method is fraught with error
can withstand extreme weights. In addition, surgical bed because of the great variance in finger size, position, color,
designs that have an off-centered pedestal to allow room for and illumination. It is preferable to use a printed, 20/200-
the surgeon’s legs can tip over with morbidly obese patients. sized “E” on a white card. This can be presented in random
These patients may not be able to lie flat during surgery due orientation with multiple repetitions, and the distance can
to orthopnea, requiring either delaying elective surgery until be measured to determine acuity as a fraction (e.g., 1/200).
the patient loses enough weight to safely undergo the pro- A patient with 1/200 vision typically cannot see well enough
cedure or operating on patients on uncomfortable angles for to walk, while a 5/200 patient typically is ambulatory, and an
the surgeon if the bed is positioned with the head elevated individual with 9/200 vision can read large print with mag-
enough for adequate ventilation. nification. Placing all these patients into the finger-counting
Pickwickian, morbidly obese, patients can become category is misleading.
hypoxic postoperatively. If an inexperienced certified nurse Flat screen display, computer-driven vision testing
assistant increases the concentration of inspiratory oxygen devices are accurate and rapid ways of testing visions of
to improve orthopnea, the oxygen-driven respiratory drive 20/400 or better and can replace more traditional projector
(since these patients have chronic hypercapnia that desensi- charts (1). The printed charts with special illumination used
tizes the CO2 chemoreceptors) can be suppressed and lead by the Early Treatment of Diabetic Retinopathy Study (2) are
to immediate postoperative hypoxia. very accurate but cumbersome.
4 SECTION I ■ Presurg ical Evaluation and Im aging

Current refraction must be in place for visual acuity posterior 30 degrees of retina. However, many patients with
testing at the 20/400 or better level. Pinholes are difficult for normal retinas cannot respond in a positive manner to this
retinal patients to use and may reduce contrast sensitivity test, greatly reducing its value.
in the presence of retinal disease, resulting in underestimat-
ing visual function. Near vision testing is essential because
Co lo r Discrim in at io n
of hereditary myopia and induced myopia from nuclear
sclerosis and encircling buckles. Patients with recent total retinal detachments and many
long-term, low detachments have excellent color discrimi-
nation with large targets. Because 90% of the cones are extra-
Co n t rast Se n sit ivit y
macular, patients with large macular scars have normal color
The ability to detect changes in contrast is a significant function with large targets. Standardized color testing meth-
measure of macular function. Not infrequently a patient’s ods are highly dependent on 20/200 or better visual acuity,
perception of visual changes and visual recovery is more negating their value in opaque media cases. Hemorrhagic
dependent on contrast sensitivity than on visual acuity, since vitreous acts as a red filter, altering color discrimination.
most real world scenarios involve reduced contrast. We do
not routinely perform formal contrast sensitivity testing on
Mad d o x Ro d
our patients. On the other hand, an adequate history from
the clinician can gather information regarding visual func- Detection of the orientation of a Maddox rod is said to cor-
tion in reduced light that can help orient surgical decision relate with good postoperative function in opaque media
making in patients with macular disease, even when formal cases. Patients with recent total detachments and advanced
contrast sensitivity testing is not performed. glaucoma can provide accurate answers to this test, thereby
eliminating its value. Certain inoperable patients respond
positively to this test, further decreasing its value.
Me t am o rp h o p sia
Amsler grid evaluation can help explain a patient’s visual
Two -Po in t Discrim in at io n
complaints and can assist in the recommendation of sur-
gery in patients with EMMs and macular striae. No objective Two-point sources of light (transilluminators) can be
evaluation has been developed for metamorphopsia analysis, moved laterally, initially touching and then progressing to
but adequate history and proper Amsler grid testing can 1-m distance. Reporting of “two lights” at close distance
demonstrate the presence of visual distortion that may be (< 2.5 cm) corresponds with better retinal function. Patients
visually disabling and that can be improved with surgery. with recent or partial retinal detachments can respond nor-
We do not recommend using only visual acuity testing for mally to this test, and light scatter can cause false negatives,
determination of need for macular surgery, since many high- dramatically reducing the value of this test.
functioning patients with relatively good visual acuity can
have disabling, correctable metamorphopsias.
Lase r In t e rfe ro m e t ry
Dense cataracts and vitreous hemorrhage prevent visualiza-
Pro je ct io n an d Fie ld Te st in g
tion of laser interference fringes. Although laser interferom-
Marked opaque media creates light scatter and might cause etry can be used with minimally opaque media, it measures
a false impression of being able to perceive the direction of vernier-type acuity, with only moderate correlation with
projection, while recent retinal detachments can have nor- Snellen acuity.
mal projection. Occasionally, an advanced glaucoma patient
with opaque media will respond only in the temporal field,
but frequently, the patient indicates a larger field due to light VALUE OF PSYCHOPHYSICAL TESTING
scattering.
In general, patients with a positive response to all of the above
psychophysical tests show better postoperative results and,
En t o p t ic Ph e n o m e n o n
conversely, those with a negative response typically achieve
The patient should be able to see the shadows of the reti- poor visual function (3). The subjective nature of these tests
nal vessels if a focal light source (transilluminator) is rubbed and the many instances of false positives and false negatives
against the closed lids. Care must be taken because these greatly decrease their value. Examiners may use these meth-
lights generate heat. If the light source is turned on after ods to seek out good prognosis cases, some of which do not
skin contact is made and the skin contact is never broken, really require surgery, but a negative response to these tests
heat dissipation is usually adequate. Many patients report frequently indicates an operable retinal detachment demand-
the shadows as appearing like leaves or cracks, and there ing immediate vitrectomy. At present, the authors use only
appears to be excellent correlation with attachment of the visual acuity testing to assess function.
CHAPTER 1 ■ Presurgical Evaluation 5

exposed iris vessels due to prior cataract surgery or certain


PUPIL TESTING
lens implants are usually quite easy to differentiate from iris
neovascularization. Topical phenylephrine decreases the vis-
An afferent pupillary defect discovered with the swinging
ibility of iris neovascularization due to vasoconstriction.
flashlight test can be of great value in preoperative evaluation.
While significant trabecular meshwork neovasculariza-
Glaucoma, retinal detachment, ischemic optic neuropathy, and
tion can be present with minimal iris neovascularization, this
optic nerve trauma are the most frequent causes of an abnor-
is somewhat unusual. Because gonioscopy is time-consuming
mal response. If one afferent system (retina and optic nerve)
and may damage the corneal epithelium, the iris is usually
and one efferent system (third nerve, ciliary ganglion, and
used as an indicator of anterior segment neovascularization,
iris sphincter) are intact, the test can have value. Bilateral iris
but gonioscopy detection of trabecular meshwork neovascu-
disease eliminates the utility of this test. As in psychophysical
larization is the most sensitive and relevant indicator of vas-
methods, a negative response may indicate an operable retinal
cular endothelial growth factor (VEGF) levels in the anterior
detachment rather than an inoperable eye. Large macular dis-
chamber. Cells and flare are present in the anterior chamber
ciform scars can cause a moderate afferent pupillary defect.
of many patients requiring vitrectomy and are at most a rela-
tive contraindication. Fibrin syndrome usually contraindi-
cates all but emergency vitrectomies for endophthalmitis.
SLIT LAMP EXAMINATION
Le n s Op acit ie s
A problem-oriented examination is far more effective than
a “routine” examination. Specific, subtle details can have a Blood or pigment on the anterior or posterior surface of the
major impact on presurgical decision making. lens or lens implant may necessitate lens or implant removal
for adequate visualization. Many vitreous hemorrhage
cases have posterior subcapsular cataracts. The lens should
Co rn e a be removed if significant posterior subcapsular changes
Epithelial erosions are common in diabetics and some are present to provide excellent surgical visualization and
patients who have had recent surgery. Corneal edema may because these changes virtually always rapidly progress post-
preclude the excellent visualization required for vitrectomy. operatively. Cortical changes, if moderate, can be tolerated.
Unless vitreous-corneal touch, lens-corneal touch, or glau- Moderate nuclear sclerosis will permit adequate visualiza-
coma to be treated by vitrectomy is present, the net effect of tion and progresses more slowly than posterior subcapsular
vitrectomy will be to further decrease endothelial function. opacities following vitrectomy.
Guttata and low endothelial cell counts indicate the possibil-
ity of postoperative corneal edema and suggest more rigid In t rao cular Pre ssure
vitrectomy indications. Blood staining, scarring, or edema, if Indentation (Schiotz) tonometry is generally inadequate
very extensive, is an indication for penetrating keratoplasty, and outdated for the measurement of intraocular pressure
Descemets stripping endothelial keratoplasty (DSEK), or (IOP). Applanation tonometry is adequate but is inaccurate
endoscopic surgery. Vitrectomies can often be performed by with irregular corneas. The Tonopen is more accurate with
working around central corneal or lenticular opacities. irregular corneas and ocular rigidity problems, causes less
corneal trauma, and does not result in fluorescein in the tear
film and anterior chamber. Tonopen tonometry is easier than
Iris-An t e rio r Ch am b e r
applanation tonometry for patients with marked blepharos-
The extent of pupillary dilation is important if the lens is to pasm and can be used with a contact lens in place.
be retained. On occasion, a clear lens lensectomy will be nec- Low pressure (hypotony) has an undeserved bad repu-
essary in an emergency vitrectomy only to allow the removal tation. Most low pressures are due to reparable wound leaks
of anterior chamber or perilenticular blood or fibrin. or reparable retinal detachments with increased uveal-scleral
Iris and trabecular neovascularization is subtle in its outflow. Low pressure does not cause phthisis; rather, phthisis
early stages but of extreme importance. The iris surface over- may cause hypotony. Phthisis might best be defined as ocular
lying the sphincter should be examined using a magnifica- collapse from a cyclitic membrane. In no instance should
tion of 24× to 40× prior to dilation. Large vessels, ectropion hypotony delay an otherwise indicated vitrectomy. Ciliary
uveae, and peripheral anterior synechia are late changes and body shutdown is an overrated, theoretical cause of hypot-
are not required for a diagnosis of iris neovascularization. ony, which occurs only after massive destruction of the ciliary
Capillaries on the iris or trabecular surface indicate activity, epithelium by cryopexy, ultrasound, laser, or infection.
while larger vessels will persist after regression of the capil- Elevated IOP has many causes, and a complete dis-
lary activity. Examiners with red-green color discrimination cussion is beyond the scope of this book. Pupillary block,
problems find it difficult to recognize iris neovascularization hemolytic (erythroclastic), and phacolytic glaucoma are
and make this condition a negative factor in the decision to treated by vitrectomy. Neovascular glaucoma (NVG) can
become a vitreoretinal surgeon. Dilated stromal vessels or be made worse by vitrectomy unless anti-VEGF agents,
6 SECTION I ■ Presurg ical Evaluation and Im aging

panretinal photocoagulation (PRP) or retinal reattachment opaque or semiopaque vitreous. A common tendency is for
can be accomplished during the procedure, thereby decreas- the clinician to give up and describe the opaque media case
ing VEGF levels. as “no view,” “no retinal detail,” or “red reflex.”
Vitreous consists of a three-dimensional (3D) matrix of
collagen fibers suspended in hyaluronic acid. The normal
SLIT LAMP BIOMICROSCOPY OF shape of the vitreous is roughly spherical with a small ante-
THE RETINA rior concavity for the lens. The continuity of the multilay-
ered outer surface (cortex) is the most essential concept in
An understanding of vitreoretinal anatomy and the forces developing an understanding of surgical anatomy. The vitre-
acting on the retina is essential in the evaluation of potential ous base is not a space-occupying structure but a zone of
vitrectomy cases. In large part, vitreous drawing is a use- adherence between retina and vitreous. The vitreous base
less exercise; the traction forces are evident from retinal con- represents a zone connecting the anterior vitreous cortex
tours and visibility of the vitreous is far better at the time (AVC) and the PVC.
of surgery. Small retinal breaks, macular edema, macular Posterior vitreous detachment (PVD or posterior
holes, EMMs, subretinal neovascular membranes, and subtle vitreous separation) occurs in approximately 70% of the
neovascularization are best observed with slit lamp biomi- population and can be thought of as a normal process
croscopy. Cloudy media decreases the value of slit lamp bio- although it is a factor in many vitreoretinal problems. Cata-
microscopy. Plano-concave (flat, Goldman) contact lenses ract surgery and other anterior segment procedures such as
with antireflective coatings provide much better lateral refractive lens exchange, ocular trauma, vitreous, myopia,
resolution than 78- to 90-diopter (D) lenses and eliminate aging, and hereditary processes increase the likelihood of
corneal asphericity. In addition, flat or three-mirror contact PVD. Syneresis is traditionally described as the development
lenses provide better axial resolution, better depth of field, of cavities within the vitreous and vitreous “collapse,” which
and improved stereopsis than 78- to 90-D lenses and wide- then causes separation from the internal limiting membrane
field contacts, although the noncontact lenses are easier to (ILM) of the retina. A better way to think of PVD is that vit-
use and somewhat more comfortable for the patient. reous collagen becomes less adherent to certain parts of the
retina, central vitreous collagen fibers cross-link and clump,
and saccadic motion facilitates a separation from the retina.
INDIRECT OPHTHALMOSCOPY Cavities do not passively form in the vitreous. Collagen
clumping creates this appearance. Collapse exerts minimal
An understanding of the vitreoretinal anatomy and the forces force because the vitreous collagen is nearly neutrally buoy-
acting on the retina is a necessity in clinical decision mak- ant in hyaluronan and the high water content of vitreous.
ing. The examiner cannot be a passive observer of vitreous Degenerative changes (syneresis), hemorrhage, inflam-
“bands” but must grasp the concepts of the continuum of the mation, thermal effects, and cellular migration and prolif-
anterior and posterior vitreous cortex (PVC) and the forces eration can cause the shortening of vitreous surfaces, which
causing retinal contour changes. is best termed “hypocellular vitreous contraction.” Dense
The indirect ophthalmoscope is essential to vitreoreti- hemorrhagic vitreous has been described as “organized” by
nal examination because of the stereopsis, wide field, and many, while it should more correctly be termed “opacified.”
contrast obtained even with nonoptimal media. It should be “Organization” is a specific term applied to the proliferation
used on its brightest setting (7.5 volts) in most instances. The of fibroblasts. It is usually observed only with severe ocular
20-D lenses have greater light-gathering ability and increased trauma or retinal necrosis.
contrast compared to the 28-D lenses, which are better for Clear vitreous is fully capable of creating sufficient trac-
small pupils and gas bubbles. If possible, the pupil should be tion force to cause a total traction retinal detachment (TRD).
widely dilated, the room dark, and the examiner well dark- Many examiners refer to “vitreous bands” in their analysis of
adapted. Continued dark adaptation, by keeping the entire vitreoretinal pathology. Discrete vitreous bands are markedly
examining unit dimly illuminated, is far more efficient than overemphasized and typically represent only a more opaque
waiting for dark adaptation to occur during each examina- section of the continuous PVC. The clear contiguous por-
tion. Noncognitive repositioning of the hand holding the con- tions of PVC often create a tremendous force on the retina,
densing lens allows prism effect to enable peripheral viewing, demonstrating that “band”-oriented vitreous description or
reduced astigmatism, and viewing around opacities. surgical approach is useless.
As interaction between a few cells and the vitreous col-
lagen fibers causes contraction, the PVC pulls away from the
VITREOUS CONFIGURATION posterior retinal surface but remains adherent to the vitreous
base. The vitreous is normally more adherent to the optic
The examiner must make a specific effort to look at rather nerve head, macula, and retinal vessels. Abnormal adher-
than through the vitreous with the indirect ophthalmoscope. ence is common in areas of neovascularization, chorioretinal
Much can be learned from the geometric configuration of scarring, trauma sites, and prior photocoagulation. Further
CHAPTER 1 ■ Presurgical Evaluation 7

vitreous contraction will create a conical configuration of vitreous and should be described and managed accordingly.
the PVC because of the posterior adherence areas. Epiretinal The retina can frequently be seen through semiopaque vit-
membranes (ERMs) may occur at the attachment sites but reous. It is useful to grade vitreous hemorrhage from 1+ to
also occur independently of the vitreous in the form of pro- 4+ or clear, semiopaque, and opaque, to permit subsequent
liferative vitreoretinopathy (PVR) star folds, EMMs, or the examiners to assess the rate of clearing. Fresh blood is bright
ridge or sheetlike ERMs seen in proliferative diabetic retin- red; the color of older hemorrhages becomes less saturated
opathy and retinopathy of prematurity (ROP). Frequently, red and finally becomes yellow or gray.
the vitreous will separate from the retinal surface in some
areas, leaving scattered zones of adherence connected by
the PVC. A large fenestration in the PVC may occur at the RETINAL CONFIGURATION
macula. Posterior vitreous separation is often accompanied
by a Weiss ring, which is an elliptical condensation of PVC The transition from retinal to vitreoretinal surgery necessi-
formerly adherent to the optic nerve head. Posterior vitreous tates a change in examination goals. A compulsive, ritualis-
separation is somewhat illusory in that an additional layer or tic quest for retinal breaks must be replaced in large part by
layers of vitreous often remain adherent to the retina in the an approach that emphasizes understanding of the forces
presence of an apparent PVD. acting on the retina. Retinal breaks eliminate the 0.8 mm Hg
Saccadic motion of the eye and a change in head posi- pressure gradient normally existing across the retina, result-
tion can help differentiate between flaccid PVC with retinal ing in a convex contoured rhegmatogenous retinal detach-
adherence and taut PVC causing traction on the retina. Sac- ment. Damage to the retinal pigment epithelium (RPE)
cadically induced motion can create dynamic traction capa- pump mechanism or increased vascular permeability also
ble of creating a retinal break but incapable of causing TRD. results in loss of the transretinal pressure gradient, which
The traction referred to in this book might more correctly be produces a convex configuration of the retina termed exuda-
termed “static traction.” tive retinal detachment. In contrast, the transretinal pressure
The conical surface of the PVC in diabetic vitreous gradient causes the concave shape of the retinal surface in
hemorrhages, TRDs, or penetrating trauma may have one TRD cases. A concave configuration with the presence of
or more apices created by zones of adherence. The most one or more retinal breaks means the normal transretinal
typical apex of the single point cone is the optic nerve. pressure gradient is still present and proves the presence of
Two-point conical PVC surfaces are usually attached to the traction. In contrast, convex retinal detachment with no vis-
optic nerve and a vascular arcade (most frequently super- ible retinal break must have a rhegmatogenous or exudative
otemporal). It is essential to understand that the PVC must component.
bridge between these vitreoretinal attachment points. Nasal Perpendicular or oblique traction on the retina is best
location of the apex of the conical surface usually precludes recognized by alterations in retinal contour. Steep changes
traction detachment of the macula. The apices can be acute in retinal contour result only from PVC adherence and trac-
angles or broad truncations of the conical PVC, indicating tion at the apex of the elevated retina. Broader zones of PVC
small or larger zones of adherence, respectively. If the vitre- adherence create truncated cones (mesa-like) of retinal ele-
ous is taut, the shape of the PVC apex can be thought of vation. Ridge-like changes in the retinal surface result from
as a mirror image of the shape of the traction detachment. perpendicular (vitreous sheet) or ERM traction. Star folds
Single acute angle apices result in simple conical traction and EMMs result from traction directed inward. Macular
detachments. Ringlike apices along the arcades and disk holes result from forces due to ILM elasticity, fenestrated
create ringlike traction detachments. A broad area of adher- ERMs, and/or residual vitreous and are directed outward.
ence of the contracted PVC to the retina causes “tabletop” Retinal incarceration in a trauma or drainage site results in
traction detachments. The continuity of the PVC between retinal foreshortening and must be relieved by retinal resec-
each apex must always be kept in mind. If the retina can be tion so that the retina can conform to the RPE.
seen in some areas but not others, the shape of the retina
in the visible areas can be used to extrapolate the contour
as well as attached or detached configuration of adjacent RETINAL DETACHMENT
nonvisible retina. MECHANISM CONFIGURATION
Exudative Convex
Rhegmatogenous Convex
VITREOUS CLARITY Traction Concave

Some assessment of the age of a vitreous hemorrhage should DIRECTION OF RETINAL SURFACE
be made. Discrete blood clots must go through thrombolysis TANGENTIAL TRACTION FINDING
and dispersion phases before clearing can occur. Subposte- Inward (centripetal) Star folds, EMM
rior vitreous detachment (sub-PVD) and preretinal hemor- Outward (centrifugal) Macular hole
rhage clear much more rapidly than hemorrhage within the
8 SECTION I ■ Presurg ical Evaluation and Im aging

The ERMs in PVR are typically less opaque than the


RETINAL BREAKS
membranes associated with diabetic TRD or ROP. For this
reason, they are frequently overlooked, and such misnomers
The detection of retinal breaks assists in surgical planning
as “retinal stiffening” are applied. Retinal stiffening in most
but can be more difficult in the patient with decreased vitre-
disease processes is from periretinal membrane proliferation,
ous clarity. In contrast to scleral buckling surgery, the view
even if the ERMs or subretinal membranes are transparent
is always far better in the operating room than in the office.
and difficult to see. Every fixed fold or star fold must have
The reward for prolonged office examination of the patient
an ERM, subretinal membrane, or incarceration as its cause
can be examiner fatigue and patient discomfort.
unless the problem is developmental. The surgical approach
Retinal breaks are frequently located adjacent to abrupt
can only be planned by determination of the location of these
changes in retinal contour associated with vitreous traction.
membranes. The exact indications for removal are discussed
If the retina is concave, it is certain that these breaks have
in the chapters on appropriate disease states.
played a minor role in the retinal detachment. As any break
may contribute to postvitrectomy detachments, all breaks
must be recognized at the time of vitrectomy and treated.
Retinal breaks are frequently located adjacent to retinal-
FOREIGN BODY LOCALIZATION
RPE adherence areas or previous retinopexy sites acted
Localization of an intraocular foreign body (IOFB) has
on by vitreous traction. A drawing and description of the
become somewhat of an end unto itself in the workup of
position of all retinal breaks should be made preoperatively
trauma cases. Vitreous surgery has radically changed the
and reviewed moments before or during surgery to avoid
approach to IOFB patients. For this reason, there is little
overlooking areas needing retinopexy. Too much emphasis
need for extensive localization studies in most instances.
is placed on the poor prognosis of so-called “combined”
If the fundus cannot be seen, in most cases, vitrectomy will
traction and rhegmatogenous retinal detachments associated
be performed and accurate visualization of the foreign body
with diabetic retinopathy.
will become possible intraoperatively. The relative inaccu-
racy of preoperative localization of foreign bodies near the
ocular wall can contribute to mismanagement.
RETINAL NEOVASCULARIZATION The nature of the injury causing the foreign body assists
in localization. Most steel foreign bodies are created by ham-
Active neovascularization of the disk (NVD) or neovascular- mering and seldom have the velocity to double penetrate the
ization “elsewhere” (NVE) can be thought of as a marker for globe. For this reason, they may be assumed in most cases
the presence of VEGF in the vitreous cavity. Assessment of to be intraocular and managed with vitrectomy and forceps
neovascularization is important not because of intraopera- removal. In contrast, most shotgun injuries are double per-
tive bleeding, which is usually easy to control, but because forating because of the high velocity, and even if a lead pel-
it correlates with postoperative NVG and fibrovascular pro- let is intraocular, it need not be removed for 10 to 14 days.
liferation on the AVC and is an absolute indication for initial Computerized axial tomography is expensive and only mod-
or supplemental PRP and anti-VEGF therapy. Large vessels erately accurate, while magnetic resonance imaging (MRI)
in an ERM without active capillary budding remain pres- is contraindicated because a ferrous foreign body will be
ent even after PRP and anti-VEGF agents markedly decrease moved by the magnetic field. Foreign body localization tech-
VEGF levels. These larger vessels are more impressive but niques not utilizing a contact lens are extremely inaccurate
are not as significant as capillary activity. and can give false information. Foreign body localization
methods using a contact lens have the risks of infection and
prolapse of ocular contents from the pressure of application
EPIRETINAL MEMBRANE of the contact lens. Ultrasonic methods, although better, are
difficult to interpret when the foreign body is near the ocular
Since “fibrosis” in the vitreous cavity is only present in the wall. This will be discussed further in the section on diag-
context of severe trauma or inflammation, this term should nostic ultrasound. Anteroposterior and lateral x-rays of the
rarely be used. It should be recognized that an ERM is usu- orbit are important, however, to determine the presence and
ally contiguous with the PVC in areas not having a posterior number of IOFBs but are not as useful in localization.
vitreous separation in the context of diabetic TRD. ERMs
should be examined for color: a brownish pigment may
indicate RPE etiology, while white coloration points more DIAGNOSTIC ULTRASOUND
toward a glial mechanism although RPE cells dedifferentiate
and lose pigment over time. It is essential to assess the rela- Diagnostic ultrasound has greatly improved the management
tionship of the ERM to any retinal elevation. Broad areas of of the opaque media patient. This method, however, is best
ERM, when contracted, cause larger areas of retinal elevation utilized in the hands of the potential surgeon rather than in
than do very small epicenters of ERM. a diagnostic laboratory setting. The surgeon has the entire
CHAPTER 1 ■ Presurgical Evaluation 9

clinical picture in mind at the time of ultrasonic examination, displayed as double, while far-field targets are displayed as
making possible better integration of the ultrasonic diagno- spread out laterally. This is inescapable without using com-
sis into the thought process. If the ultrasound equipment puted sonography, which is currently not available in oph-
is located in the surgeon’s primary examining room, it will thalmic ultrasound systems.
be utilized much more frequently and can be done without Axial resolution is a function of frequency, with higher
charge if needed. Technicians should not be used for what frequencies resulting in better axial resolution. Higher fre-
should be thought of as acoustic ophthalmoscopy. quencies are absorbed more by biologic structures, mandat-
ing higher powers to obtain the same small echo sensitivity.
The risk of cataract formation determines the maximum
Clin ical Aco ust ic Ph ysics
power limit that can safely be utilized. In practice, this com-
An understanding of the physical principles involved in the promise results in the use of 10- to 20-MHz ultrasound with
interaction between ultrasonic energy and biologic materi- about 0.15-mm axial resolution, which is more than an order
als is essential for accurate ultrasonic diagnoses. Ophthalmic of magnitude better than lateral resolution (Fig. 1.3). Axial
ultrasonography utilizes pulse-echo reflection ultrasound. resolution is degraded when the wide beam is reflected from
Brief pulses of ultrasonic energy having a 10-MHz, or curved surfaces such as the crest of a TRD.
greater, center frequency are repeated at a rate of 1 to 5 kHz, The greatest reflection of ultrasound is obtained when
allowing time for the same transducer to receive the reflected the beam strikes the interface perpendicularly. Beams reflect-
echoes. Knowledge of the average speed of ultrasonic energy ing tangentially from the equatorial ocular wall result in little
propagation through tissue (~1,540 m/s) permits the real- reflection back to the transducer. With accurate representa-
time calculation and display of the distance between the tion of echo amplitudes, an entire circular eye cross section
transducer and the echo-producing structure as a dimen- cannot be displayed.
sion on the two-dimensional (2D) display. Acoustic energy Because the speed of sound is greater in denser tissues
is reflected and refracted at interfaces between materials of such as the lens, structures behind the lens are displayed as
different acoustic densities (Fig. 1.1). being closer, and refraction occurs at the edge of the lens
If the face of the piezoelectric transducer crystal has a (Fig. 1.4). Dense structures such as lens, intraocular lens
small radius of curvature, the result is a point focus with (IOL), IOFB, and scleral buckles have multiple internal
poor depth of field. The length of the eye (~25 mm) requires reflections and display evenly spaced false echoes of decreas-
a weaker focusing to obtain an appropriate depth of field. ing amplitude behind the structure (Fig. 1.5). The echoes
This wide beam width (3 mm at the 6-dB point) creates produce paradoxical movement with transducer reposition-
poor lateral resolution (Fig. 1.2): Targets in the near field are ing, which aids in their recognition. Dense structures such as

Fig ure 1 .1 ■ Materials of different acoustic densities reflect and refract acoustic energy at their
interfaces.
10 SECTION I ■ Presurgical Evaluation and Im aging

Fig ure 1 .2 ■ Poor lateral resolution results from the inherently wide ultrasound beam .

Fig ure 1 .3 ■ 10-MHz ultrasound produces axial resolution ten tim es greater than the lateral
resolution.
CHAPTER 1 ■ Presurgical Evaluation 11

Fig ure 1 .4 ■ Multiple, evenly spaced false echoes result from internal reflection with an IOL.

Fig u re 1 .5 ■ Structures p osterior to d ense tissue ap p ear closer than their actual location
because of increased speed of sound in denser m aterials.
12 SECTION I ■ Presurgical Evaluation and Im aging

calcified cyclitic membranes, IOLs, and foreign bodies create or 3D slices using perspective, shading, motion parallax,
significant shadowing behind them because of the absorp- and various other digital graphics technologies. Because the
tion of acoustic energy. image data set is acquired from a single vantage point, struc-
The absorption of ultrasonic energy as it passes twice tures with surfaces that are more tangential than perpendic-
through the tissue results in distant echoes being displayed ular to the scan beam will be invisible or depicted with far
with relatively less amplitude. Electronically increasing the less reflectivity. Current 3D ultrasonic imaging techniques
gain for distant targets can compensate for this absorption. are of minimal value in vitreoretinal disease and best used
This is called time-varied gain. for tumor volume determination.
Electronics that automatically display interfaces such
as cornea, lens capsule, retina, and sclera have resulted in
Re al-Tim e
diagnostic inaccuracies. Increasing the amplitude and clip-
ping the peaks to display interfaces means that all echoes Real-time is defined as appearing virtually continuous and
are displayed as equal amplitude. With this approach, the instantaneous to the human observer. It is accomplished by
vitreous and retina can easily be confused. Similarly, elec- using a scanning rate of 10 to 30 scans per second. Real-
tronic differentiation to detect interfaces eliminates the lower time ultrasound has many advantages in presurgery decision
amplitude echoes within the lens, vitreous, subretinal fluid, making. As opposed to a static scan captured with digital
suprachoroidal space, and tumors. frame store, continuous tomography is possible by moving
the transducer. As the transducer is moved with respect to
the eye, the examiner can observe the screen and the relation-
A-Scan Ve rsus B-Scan
ship of the probe to the eye and integrate multiple 2D scans
Time amplitude ultrasonography (A-scan) was the origi- into a 3D mental picture of the intraocular contents utilizing
nal method of ultrasonography but has virtually no util- proprioceptive awareness of the transducer position. Any
ity in the opaque media workup. A-scan information is attempt to reconstruct this picture from a protocol-driven
one-dimensional (1D) and is analogous to a “needle in approach using individual images taken at discrete positions
the haystack” approach. Extremely experienced A-scan is much more difficult and inaccurate.
ultrasonographers can spatially integrate the 1D informa- Real-time ultrasound is of great value in detecting
tion and obtain some limited value. The typical clinician, rigidity or, conversely, mobility of ocular structures. The
however, has far more difficulty with this approach. Quan- PVC, when detached from the retinal surface in a PVD con-
titative A-scan echography has been overemphasized as figuration, is highly mobile. Loose vitreous apices attached
being capable of a tissue diagnosis. A-scan echo amplitude at a single point to the retina are quite mobile but become
is highly dependent on the angle at which the ultrasonic immobile if they are taut with resultant TRD. Rhegmatog-
beam strikes the reflecting structure. Oblique angles cause enous retinal detachments are somewhat less mobile than
marked attenuation of the reflected echo. Highly convo- the detached vitreous but have definite undulation motion
luted retinal detachments will have areas of high and low during saccadic motion of the eye. Retinal detachments with
reflections. For this reason, there is a large sampling error PVR have much less mobility than a typical rhegmatogenous
in this 1D approach. retinal detachment (4). Giant breaks have marked retinal
mobility (5).
With real-time capability, moving patients and even
B-Scan
those with nystagmus can easily be examined. In fact,
Sector-type B-scan ultrasonography is a 2D approach in it is advantageous to elicit repeated ocular motion in
which a knifelike slice or plane of acoustic section is made all directions to decrease the sampling error intrinsic to
through the tissue, as opposed to the 1D needle approach 2D scanning and to test the rigidity and mobility of ocular
of A-scan. Echoes are displayed as intensity-modulated pix- structures.
els. As with A-scan ultrasonography, the best reflections are Preretinal hemorrhage can be seen to flow from one
from those structures roughly perpendicular to the beam. position to the other, and dislocated lenses, implants, and
For this reason, the cornea, anterior and posterior lens, and foreign bodies can be seen to move with saccadic motion,
retinal and scleral surfaces are the best-displayed structures. changing gaze, or patient repositioning (supine or seated).
The equatorial sclera and lens are seen less well, except Real-time adds the fourth dimension (time) to the 3D image
when the eye is rotated or approached from different angles, reconstructed in the examiner’s cerebral cortex. When the
emphasizing the need to move the eye and probe during the posterior vitreous has detached from the retina, it will lie in
examination. contact with the retina when the patient is in the supine
position. Saccadic motion can then demonstrate with
real-time ultrasound that the apparent adherence is only
Th re e -Dim e n sio n al
gravity-induced contact of the vitreous with the retina. The
Slow rotation of a sector scan will produce a conical volume saccadic-induced motion of preretinal hemorrhages can help
of information that can be displayed as a conical 3D image differentiate them from disciform scars.
CHAPTER 1 ■ Presurgical Evaluation 13

Gray Scale are measured in ultrasound by the use of an assured


speed of sound, increased tissue density causes more
Gray scale B-scan relates echo amplitude (acoustic reflectivity
distant echoes to be displayed as being closer to the
or absorption) to the brightness of specific pixels on the
transducer.
display. The vector positional data from the sector scan are
5. Displays have only a 20- to 30-dB dynamic range,
displayed as a sector on the screen; very reflective echoes
while ultrasound interpretation of ocular tissue
have white dots on the screen, less reflective echoes have a
produces 60- to 100-dB dynamic range. For this
dimmer shade of white, and absence of echo is displayed as
reason, high gain must be used for minimal vitreous
black. Simply stated, the more sound reflected from the ocu-
hemorrhage or normal vitreous surfaces, while the
lar structure, the more light emitted from the corresponding
gain must be turned down to avoid saturating high-
area of the screen. Earlier ultrasound units had static scan-
amplitude echoes such as IOLs.
ning with bistable storage cathode ray tubes (CRTs), and the
echo amplitude information was therefore not displayed two-
dimensionally. Combined A-scan was utilized to obtain a lim- Ve ct o r Scan n in g
ited, 1D impression of echo amplitude. If a combined A-scan
The process of electronically removing a line from a real-time
approach was used and the A-scan intersected the retina
B-scan and displaying it in the typical time amplitude, A-scan
obliquely, it might be called vitreous. The pattern recognition
manner is called vector scanning or simply simultaneous
capabilities of the examiner, however, can, in effect, join the
A-scan and B-scan. It makes possible the combined A-scan
dotted lines between the highly reflective areas using a gray
approach with its wide dynamic range combined with the
scale B-scan. With low noise and high dynamic range signal
real-time, gray scale, 2D topographic information. It is of
processing, even diffuse red blood cells and normal vitreous
more value in looking at internal tumor architecture than it
can be seen. Long-term retinal detachment causes atrophy,
is in the typical opaque media previtrectomy workup.
decreasing acoustic reflectivity just as cellular proliferation in
the vitreous can increase reflection, making amplitude alone
an insufficient criterion to distinguish retina from vitreous. Im ag e Arch ivin g an d Vie win g
When gray scale is coupled with real-time (6), it permits
Hard copy images can be used to establish to the reimburse-
a further appreciation of retinal structures, such that diagno-
ment or legal community that an ultrasound examination
sis is made in a pattern recognition manner rather than by
was performed. Rarely, prior digital or hard copy images can
itemization of individual diagnostic criteria. The authors con-
be compared with the current on-screen image to determine
sider a real-time, gray scale B-scan to be an absolute necessity
if a change has occurred. The authors again emphasize that
for the workup of the opaque media patient (7,8).
ultrasound is a key component of the clinician’s examina-
tion, not a test, archival, or photo opportunity.
Te ch n ical Lim it at io n s
Diagnostic ultrasound has certain limitations: Exam in at io n Me t h o d s
1. The typical semifocused B-scan transducer creates a Contact scanning on the eyelid is used in the majority of
beam that is 2 to 3 mm in width, and as a result, the opaque media examinations before vitrectomy. A monolithic
target is displayed widened in the lateral orientation. blob of highly viscous acoustic coupling material is applied
A target is imaged before and after the beam center to the probe before application to the upper lid for contact
is lined up with the target during the sector scan. scans. The patient is asked to look multiple times in every
Highly focused beams improve lateral resolution but direction, and the scan is made in the anteroposterior direc-
markedly decrease the depth of focus. tion both through the lens for the classic picture and then
2. Axial resolution is a function of the frequency of the outside the lens for better resolution and to aid in 3D think-
ultrasound. Higher frequency ultrasound, however, ing. It is best that this examination be performed in the stan-
is absorbed more by tissue, thus requiring dangerous dard examining room and that an ultrasound unit is always
power levels to achieve the same sensitivity or imag- available. Putting the unit on a wheeled cart can also increase
ing limited to the anterior segment. Practical 20-MHz utilization. Ultrasound should be used at every visit in which
retinal B-scan technology is now available. the patient has opaque media, because the retinal situation
3. Beam inclination to a reflecting “surface” markedly can change very rapidly from one visit to the next (9,10).
decreases the amplitude of the reflected echo. The
complex topography of vitreoretinal disease pre-
vents quantitative echography because of this beam OPTICAL COHERENCE TOMOGRAPHY
inclination–induced decrease in echo amplitude.
4. Denser structures such as a nuclear sclerotic cataract, Optical coherence tomography (OCT) (11) is an imaging
IOL, or calcified tissue absorb sound but increase the technique that produces high-resolution cross-sectional
speed of sound within the tissue. Because distances images of optical reflectivity. It is based on the principle of
14 SECTION I ■ Presurgical Evaluation and Im aging

within thickened neurosensory retina. OCT images that


topographically display retinal thickness can be produced.
Topographic maps and cross-sectional OCT images can be
displayed using a false color scheme thought by some to
facilitate interpretation; however, this approach produces
false interfaces; gray scale images allow better visualization
of detail and facilitate correct interpretation.
OCT has become an invaluable tool in evaluating
EMMs (12) (Fig. 1.6), partial- and full-thickness macular
Fig ure 1 .6 ■ OCT im ag e of EMM.
holes (13) (Fig. 1.7), diffuse and cystoid macular edema,
vitreomacular traction, submacular fluid, macular schisis,
low-coherence interferometry where distance information macular cysts, and choroidal neovascular membranes (14).
concerning various ocular structures is extracted from time Spectral domain OCT often demonstrates clinically relevant
delays of reflected signals. OCT systems use light waves pathology not visible with contact lens slit lamp biomicros-
emitted by a superluminescent diode operating at 840 to copy or angiography. It has become an essential part of mac-
870 nm and between 200 microwatts and 1 milliwatt to ular assessment when macular surgery is being considered.
construct the images. This is analogous to the application The authors prefer the Heidelberg Spectralis HRA-OCT
of sound waves in B-scan ultrasonography. The use of light to other available systems for many reasons. The Heidelberg
waves enables OCT to achieve an optical axial resolution of Spectralis HRA-OCT produces 40,000 A-scans per second
7 mm and a digital depth resolution of 3.5 mm using the and the best OCT resolution available. This device also has
Heidelberg Spectralis HRA-OCT spectral domain OCT. The TruTrack, which tracks eye movements and sets the OCT
initial systems were time domain (Zeiss Stratus) and used a scan line on the same position on the reference images or
moving mirror in the reference arm which required much scanning laser ophthalmoscope (SLO). The combined SLO
longer acquisition times, resulting in motion blur and less produces 15 dB better signal-to-noise ratio than fundus
scan lines. Less scan lines degrade the image quality and pro- camera–based angiography systems. In addition, the Heidel-
duce more artifacts. Spectral domain OCT systems (Fourier berg Spectralis HRA-OCT produces very high-quality auto-
domain) use a spectrometer analyzed by Fast Fourier Trans- fluorescence images using tracking and signal averaging (Fig.
formation instead of a moving mirror in the reference arm. 1.8). The 3D reconstruction enables overlapping of SLO and
OCT images can be presented as either cross-sectional OCT images. OCT image XY location is simultaneously dis-
images or topographic maps. Although autosegmentation played on infrared, autofluroscence (AF), or SLO angiogram
algorithms can produce topographic maps of isolated retina images by using a clickable cursor.
or RPE (retinal pigment epithelium), this has not been dem- We recommend that retina surgeons learn to analyze
onstrated to be of clinical value except for following RPE spectral domain OCT the way a radiologist reads a brain
loss or drusen volume in clinical trials. Cross-sectional or MRI: All scans should be seen by the physician to avoid
B-mode imaging is accomplished by acquiring a sequence missing pathology that a single line scan printout may not
of 40,000 A-scans (Heidelberg Spectralis HRA-OCT) across show. The best place to examine OCT data is at the com-
a section of retina. puter terminal where all image raw data are stored rather
Retinal thickness can be assessed longitudinally using than from an image database where only a few scans per ses-
serial OCT images. The presence of cystic spaces can be sion are stored, based on the opinion of the technician, or,
detected by the presence of focal areas of very low reflectivity even less, from a single scan printout.

Fig ure 1 .7 ■ OCT im age of m acular hole.


CHAPTER 1 ■ Presurgical Evaluation 15

that in retinitis pigmentosa. Therefore, a patient who had


a vitreous hemorrhage after a successful PRP would have a
nonrecordable bright flash ERG, causing some examiners to
think that surgery was not indicated. If a protocol mentality
allows the bright flash ERG to be utilized in a patient with
clear media just because his or her initial ERG testing was
not recordable, permanent damage to the retina can result
from the bright strobe.
Because of the many false positives and false negatives
described above and the possibility of technical laboratory
error, bright flash ERG has very little value in the preopera-
tive evaluation. Certainly, if the ERG is recordable and other
clinical factors suggest surgery, the patient has a better prog-
nosis. Having a better prognosis, however, in many clinical
situations is not the reason to have a vitrectomy.

VISUAL EVOKED POTENTIAL


Fig ure 1 .8 ■ Tracking and im age averaging on the Spectralis
produce high-quality autofluorescence im ages. The visual evoked potential (VEP) also has limited value
because of difficult interpretation, equipment complexity,
false negatives, and false positives. As with ERG testing, a
patient with severely decreased preoperative VEP might not
BRIGHT FLASH be considered for vitrectomy, when in reality, he or she could
ELECTRORETINOGRAPHY have useful vision after vitrectomy if a retinal detachment is
repaired.
Bright flash electroretinography was originally developed in
the context of recording the human early receptor poten-
tial (ERP) (15). It was discovered that bright strobes could References
overcome the light-absorbing property of dense vitreous 1. Timberlake GT, Mainster MA, Schepens CL. Automated visual
hemorrhage and allow the elicitation of an electroretinogram acuity testing. Am J Ophthalmol 1980;90:369.
(ERG) in these densely opaque media patients. There are 2. NAS-NRC Committee on Vision and Recommended Standard
Procedures for the Clinical Measurement and Specification of
many problems with this method, however (16,17). Many
Visual Acuity: Report of Working Group 39. Adv Ophthalmol.
ophthalmologists incorrectly think that a retinal detachment 3. Michels RG, Ryan SJ. Preoperative evaluation of patients for
will still have a recordable ERG that can be used to deter- vitreous surgery. In Gitter KA, ed. Current concepts of the vitreous
mine retinal viability. However, because of conduction prob- including vitrectomy. St. Louis: C.V. Mosby, 1976:121–128.
lems, a total retinal detachment of any age does not produce 4. Han DP, Lewandowski M, Mieler WF. Echographic diagnosis
of anterior hyaloid fibrovascular proliferation. Arch Ophthalmol
a recordable ERG. Similarly, some examiners believe that
1991;109:842–846.
a nonrecordable bright flash ERG means that a vitrectomy 5. Genovesi-Ebert F, Rizzo S, Chiellini S, et al. Echographic study
should not be done. In most cases, the remaining part of of the vitreoretinal interface in giant retinal tears. Ophthalmo-
the clinical and ultrasound examinations will reveal a total logica 1998;212(Suppl 1):89–90.
detachment, which requires immediate vitrectomy. In con- 6. Charles S, Griffith J, Lindgren W. Unpublished paper, 1974.
7. Capeans C, Santos L, Touriño R, et al. Ocular echography in
trast, many surgeons wish to do cases that have a recordable
the prognosis of vitreous hemorrhage in type II diabetes mel-
bright flash ERG. While this is certainly easier for the sur- litus. Int Ophthalmol 1997–1998;21:269–275.
geon, if these are unilateral moderate vitreous hemorrhages, 8. Kumar A, Verma L, Jha SN, et al. Ultrasonic errors in analysis of
these are just the cases that may not require vitrectomies. Use vitreous hemorrhage. Indian J Ophthalmol 1990;38:162–163.
of this test, therefore, as a prognostic means must be thought 9. Jack RL. Ultrasonographic ocular evaluation prior to vitrectomy.
In Irvine AR, O’Malley C, eds. Advances in vitreous surgery.
of in terms of the complete clinical picture.
Springfield: Charles C. Thomas Company, 1976:100–112.
Although it is true that a decreased B-wave correlates 10. Jack RL, Hutton WL, Machemer R. Ultrasonography and
with ischemic inner retinal layers, this is also not a valu- vitrectomy. Am J Ophthalmol 1978;78:265.
able criterion because the small portion of the retina around 11. Huang D, Swanson EA, Lin CP, et al. Optical coherence tomog-
the macula can be well perfused and capable of good vision raphy. Science 1991;254:1178–1181.
12. Wilkins JR, Puliafito CA, Hee MR, et al. Characterization of
postvitrectomy. Further, false negatives occur in extremely
epiretinal membranes using optical coherence tomography.
dense vitreous hemorrhage cases because even the bright Ophthalmology 1996;103:2142–2151.
strobes used cannot penetrate the ocular media sufficiently. 13. Hee MR, Puliafito CA, Wong C, et al. Optical coherence tomog-
Extensive PRP may cause a nonrecordable ERG similar to raphy of macular holes. Ophthalmology 1995;102:748–756.
16 SECTION I ■ Presurgical Evaluation and Im aging

14. Michels S, Rosenfeld PJ, Puliafito CA, et al. Systemic bevaci- 16. Fuller DG, Knighton RW, Machemer R. Bright flash
zumab (Avastin) therapy for neovascular age-related macular electroretinography for the evaluation of eye with opaque vit-
degeneration twelve-week results of an uncontrolled open-label reous. Am J Ophthalmol 1975;80:214.
clinical study. Ophthalmology 2005;112:1035–1047. 17. Fuller D, Knighton R, Machemer R. Bright flash ERG. In: Irvine
15. Galloway NR. Early receptor potential in the human eye. AR, O’Malley C, eds. Advances in vitreous surgery. Springfield:
Br J Ophthalmol 1967;51:21. Charles C. Thomas Company, 1976:97–99.
SEC TIO N II
Surg ical Te ch n o lo g y an d Te ch n iq ue s

17
C H AP TER
2
SURGICAL SYSTEMS, TOOLS,
AND OPERATING ROOM

High-technology vitreous surgery requires troubleshoot, improvise, and perform better and faster in the
intensive and continuous attention to the equip- operating room (OR). A surgeon should be able to set up all
ment and operating environment. Equipment, machines, set all parameters and modes, and connect, prime,
supplies, training, staffing, maintenance, and stor- and test all tools without the help of a nurse or technician.
age of the equipment are ultimately the surgeon’s responsibil- This knowledge reduces dependency on OR personnel and
ity. The requirements for effective high-consequence surgery resultant anxiety if key people are not available.
should outweigh logistical considerations. Blaming problems
on equipment, companies, nurses, technicians, administra-
tors, or managed care is irresponsible and ineffective.
OPERATING ROOM ENVIRONMENT
Vitreoretinal surgery has undergone many engineer-
ing advances in the four decades since it was first intro-
Sch e d ulin g
duced. Most of the important developments resulted from
an intuitive rather than a physics-based design approach. By An attempt should be made to schedule vitrectomies when
contrast, medical school curriculums emphasize a biology- the surgeon and team are not too rushed or heavily bur-
based approach to diagnosis and therapy. The intent of this dened mentally. If this means early, late, day-off, or week-
chapter is to provide a physics and engineering framework end surgery, it must be done. Some surgeons become tense if
for understanding the essential techniques and technologies they are heavily committed for time following their surgery
used in vitreoretinal surgery today. time. As some vitrectomies can be lengthy, especially during
There are many similarities between the task domain the learning phase, time must be available to facilitate con-
of the pilot and of the surgeon. Pilots are required to under- centration on the surgery rather than a subsequent event.
stand the engineering principles behind the propulsion, A designated OR is preferable to ad hoc room utilization as it
hydraulic, pneumatic, avionics, and electrical systems, facilitates equipment accessibility, maintenance, and stock-
while surgeons often use tools and machines with little or ing and storage of disposables.
no understanding of how they work. The physics of flight is
very complex but is relatively well understood by both engi-
In st rum e n t St o rag e
neers and pilots. Tissue cutting, turbulence, friction, state
change in materials, and fracture propagation are so complex The approach in which instruments are assembled from
that a precise mathematical description of the underlying various sources just prior to the case is inadequate for high-
physics is impossible. Nevertheless, valid assumptions about technology vitreous surgery. It is not optimal to share these
the physical principles involved and estimation of the math- specialized instruments with other surgeons and proce-
ematical parameters can and should be made. Vitreoretinal dures. A tray that is equipped for all basic vitreous, retinal,
surgery requires a wider variety of techniques and technolo- and microsurgical methods should be prepared and stored
gies than cataract surgery, because it is used to treat a broader together. The removal of dense lenses, intraocular lenses
array of disease entities and anatomic configurations using a (IOLs), and large intraocular foreign bodies through the
greater array of technologies. Knowledge of the engineering limbus requires that the appropriate instruments be present
concepts involved in vitreoretinal surgery can facilitate better on the basic vitrectomy tray. Similarly, scleral buckling tech-
selection of tools and techniques as well as infusion, suc- niques may be required and probably should not dictate the
tion, duty cycle, and cutting rate parameters. Surgeons who use of a second tray. An identical sterile backup tray should
understand vitreoretinal surgical systems can problem solve, be available at all times in the OR or adjacent storeroom.
18
CHAPTER 2 ■ Surgical System s, Tools, and Op erating Room 19

All fragile instruments such as intraocular scissors, Vid e o Re co rd in g


forceps, contact lenses, and the like must be kept sterile at
While certain cases may have teaching value, video recording
all times in transparent peel-packs stored in well-labeled
should not become a time- and concentration-consuming
trays, cabinets, or boxes. These instruments are best stored
consideration in the OR. Centering the microscope and
in a lockable cabinet in the OR or a movable cabinet that is
producing the best video opportunity should not consume
taken to the OR before use. They should be stored in spe-
costly labor dollars or a surgeon’s mental focus. At best, it
cific places in the cabinet to permit daily inventory control
is a moderately efficient way to teach and is often used for
and maintenance. If not stored in a single cabinet, the instru-
promotional purposes. 3-CCD cameras provide better color
ments tend to become lost and unavailable at a critical time
purity and image quality than 1-CCD cameras but require
in vitrectomy.
over three times as much light. Firewire (IEEE 1394) or
All disposable materials such as packs, patient drapes,
DVCAM is currently the optimal connection scheme between
microscope drapes, sutures, needles, IOLs, gases, and tub-
the camera and the digital recording systems. Recording can
ing sets should be stored in sufficient numbers in a spe-
be on hard drives or DVDs. Currently, the best image quality
cific place near the OR. In this way, faulty or inadvertently
is obtained with AVI files, although MPEG is a reasonable
contaminated materials and instruments can be instantly
alternative for DVDs as it requires less storage space. Direct-
replaced without having to be ordered from a central supply
to-edit (DTE) systems are far more efficient than systems
area. The many steps in a complex procedure should not
requiring capturing the video at a time subsequent to the
be delayed while waiting for instruments or materials to be
surgical procedure. DTE permits the first review of the video
delivered.
using fast forward rather than normal speeds.
A fast para-acetic acid (Steris) and/or gas plasma (Sterad)
as well as a fast cycle steam autoclave should be immedi-
ately available to the OR personnel to reduce case turnover
Tissue Cut t in g
time as well as resterilization of inadvertently contaminated
equipment if sufficient backups are not available. An external force placed on an object will cause it to move,
unless the object is constrained. A force placed on a con-
strained object will cause an internal reaction called stress
Pre surg ical Eq uip m e n t Te st in g
that resists the external force. Tensile (pulling) and compres-
All equipment should be set up and tested prior to the sive stresses occur normal (perpendicular) to the cross-sec-
administration of anesthesia. If needed vitrectomy, lensec- tional plane of an object. Guy wires on a suspension bridge
tomy, phacoemulsification, endophotocoagulation, operat- are subject to tensile stress while the road surface is subject
ing microscope, or scissors equipment is not functional, the to compressive stress by a traveling car. Forces that act in
case must be postponed. Infusion fluid should be run copi- the plane as opposed to normal to the plane are called shear
ously through all tubing to remove any bubbles or particulate stress and can be identified as transverse or torsional (twist-
material. Only after all equipment is tested and the surgeon ing). A rivet between two sliding metal plates may be subject
is present can the team begin local or general anesthesia. to transverse shear. The drive train of a car has a number of
torsional stresses when the throttle is rapidly depressed.
Force placed on a constrained object induces defor-
Op e rat in g Ro o m Pe rso n n e l
mation in relation to the amount, duration, and speed of
Preferably, a single surgical technician or nurse should be onset of the external force. The proportion of the amount of
responsible along with the surgeon for all instruments as deformation per unit length is known as strain. Tensile and
well as for the ordering, maintenance, and inventory of dis- compressive stresses will cause a lengthening or shortening
posable materials. Preferably, this individual should also of an object. Transverse shear stress causes bending, and tor-
function in the office environment to enhance information sional shear stress causes a twisting deformation (Fig. 2.1).
transfer, efficiency, and patient confidence. This same indi- Relatively small amounts of stress cause small amounts of
vidual can assist in the recording and compilation of preop- strain. After the stress is removed, the object will return to its
erative, intraoperative, and postoperative data for outcome original size and shape in a process known as elastic strain.
testing, medical records, and billing. Participation in the Increasing amounts of stress will eventually cause a perma-
follow-up care provides personal evidence of the impact of nent deformation in the object that persists after the stress
surgical success and failure. This individual should then be is removed. In material such as steel, the permanent defor-
in command of the remainder of the team and should be mation is caused by changes in the crystalline structure of
responsible for backup personnel in their absence. A friendly, the metal. Eventually, small cracks appear that interconnect,
cooperative atmosphere with a sense of humor is conducive leading to rupture and failure of the metal.
to the team-play attitude, which is as necessary in the OR as Biological tissues, particularly in the eye, display more
it is for a successful sports team. This is preferable to a tense, complicated characteristics. An externally applied force can
angry, chauvinistic, find-someone-to-blame attitude unfor- cause deformation, with the possibility of elastic strain. With
tunately so prevalent in ORs. continuing force, a “flow” may occur, such as that seen in
20 SECTION II ■ Surgical Technology and Techniques

diameter of the cutter by the thickness of the outer “needle”


wall. Tissue larger than the diameter of the port must be
elastically deformed to pass through the port.
Sharpness can be defined as high pressure per unit area
created by a blade with a thin cross section. Knives work
by sharpness, while scissors are never “sharp” or “dull.”
It is not known if microscopic surface roughness facili-
tates cutting or increases undesirable tissue displacement.
The tip of the microvitreoretinal (MVR) blade, although
originally developed for making the sclerotomies (Devel-
oped by Steve Charles, September 1976), is occasionally
used to incise intraocular tissue including ERM. Angled
knives have been developed to delaminate ERMs but have
not been widely adopted because of the risk of iatrogenic
retinal breaks.
Fig ure 2 .1 ■ Force p laced on a constrained object ind uces Inertial cutting utilizes a rapidly moving cutting element
deform ation in relation to the am ount, duration, and speed of impacting against tissue at rest and is the mode of action
onset of the external force. of the phacoemulsifier. The term “emulsification,” however,
is inaccurate as this refers to overcoming surface (interfa-
cial) tension to produce small droplets of a liquid otherwise
a viscous fluid. This combination of elasticity and viscous immiscible in water. It is probable that vitreous cutters do
flow is termed viscoelasticity. For example, it is common for not achieve velocities sufficient for significant inertial cut-
the scleral buckling effect from a sponge exoplant to appear ting effects.
larger 1 or 2 days later. This is caused by viscoelastic changes Lasers and the peak electron avalanche knife (PEAK)
in the sclera. work by producing vaporization of tissue, which inherently
As noted above, the remarkable complexity of tissue produces bubbles and acoustic (remote, mechanical) effects.
cutting prevents an exact physics-based understanding and Colloquial terms such as “ablation” and “disruption” are mis-
mathematical representation. The author has found it useful leading as they are not physics-based terms. Lasers produce
to define several types of tissue cutting in order to develop power densities sufficient to vaporize tissue or water by utiliz-
and utilize more effective techniques and devices: elongation, ing spatial or temporal coherence. Temporal coherence means
shear, sharpness, inertial, vaporization, and enzyme-assisted. that power is delivered in a very short interval, whereas spatial
Ideal cutting can be defined as separating tissue into two coherence means the energy is delivered to a very small vol-
pieces without producing any remote forces or physical or ume. Spatial coherence inherently produces a small volume
chemical effects. of tissue vaporization. Therefore, tissue cutting is dependent
Elongation simply means to apply force to tissue or a upon a contiguous line of spots or propagation of a tear that
tissue interface until the tissue fails, tears, or breaks. This occurs with Yittrium Aluminium, garnet (YAG) laser capsu-
is analogous to the methodology used in tensile strength lotomy. ERMs and vitreous are very elastic as well as resis-
testing. Membrane peeling is an example of this mode of tant to fracture propagation. As a result of these properties,
action. Epiretinal membranes (ERMs) are thought to be it requires very high-power densities to produce significant
approximately 100 times stronger than retina, which limits tissue cutting. High-power densities produce many bubbles
the application of this method to cases with low adherence as well as a propagating acoustic pressure wave, which can
between the retina and the membrane. damage tissue remote to the intended site. The erbium YAG
Shear can be defined as moving two square-edged cut- laser requires shielding from the retina, produces bubbles,
ting edges past each other with the target tissue interposed. requires vitrectomy system–like fluidics, and removes tissue
Scissors implement shear type cutting with nonparallel at a very slow rate (1). Femtosecond lasers used for LASIK
blades that push the tissue forward as the blades close. This flap creation and other anterior segment applications utilize
concept can be referred to as an “exclusive shear.” Scissors very high temporal coherence and inherently broadband
cut at a point that advances toward the tips with blade clo- (white) light but are not applicable to ERM removal.
sure. Vitreous cutters also work primarily by shear. How- Enzyme-assisted approaches to vitreoretinal surgery
ever, in contrast to scissors, vitreous cutters cut on a line such as microplasmin-assisted vitrectomy have been stud-
rather than a point. Vitreous cutters utilize parallel cutting ied for over three decades but have not been shown to be
edges and, therefore, do not produce the “squeeze out” safe and effective at this time. Enzymatic methods have
force produced by scissors. Vitreous cutters may be termed been directed at liquefying the vitreous or for separation of
“inclusive” shears. Vitreous cutters require a pressure gradi- the posterior vitreous cortex from the retina (2). Problems
ent across the port to cause fluid or tissue to move into the include the following: (a) potential for damage to the retina,
port. The actual cutting edges are recessed from the outside lens, or zonules; (b) inflammation; (c) potential for prion,
CHAPTER 2 ■ Surgical System s, Tools, and Op erating Room 21

viral, or bacterial contamination from biological materials; retinal breaks. A baseline incidence of aphakic-like retinal
(d) the need to inject the enzyme at least 30 minutes before detachments will exist as long as vitreous is removed using
surgery; and (e) complicated, costly preparation processes. suction force. Factors other than suction force such as lattice
Autologous plasmin enzymes are obtained from the patient’s degeneration, preexisting retinal breaks, and vitreous incar-
own plasma and, thus, will not have prion or viral contami- ceration in the sclerotomies also cause postvitrectomy retinal
nation and appear to be safe with respect to damage to ocu- detachments.
lar tissues.
Infusion Fluidics
Surgica l Fluidics
Pars plana vitrectomy requires both infusion and aspiration;
A pressure difference across the suction port of a vitrec- there are many parallels between infusion and aspiration flu-
tomy instrument is necessary to cause substances to enter idics because they are influenced by the same physical prin-
the port. The term “passive egress” is incorrect because ciples. Resistance to fluid flow is determined by the internal
there is no physical difference between transorifice pressure diameter of a lumen, length of tubing, and flow restrictions
(TOP, delta P) achieved by high infusion pressure and that in the cannula. Fluidic resistance is proportional to the
achieved by negative pressures (vacuum) on the inside of fourth power of the diameter (Hagen-Poiseuille equation)
the port. Semirigid materials such as ERM and lens nucleus and linearly related to the length. The impact of diameter
must be deformed to pass through a suction port in response is significant because of the fourth-power relationship and
to a TOP gradient. Markedly low TOP will prolong the pro- clinically relevant because of the transition from 20-gauge
cess of vitreous removal; large gradients create vitreoretinal (0.89 mm) to 23-gauge (0.75 mm) or 25-gauge (0.5 mm)
traction and retinal breaks. technology in recent years.
In general, a maximum suction setting of 150 mm The resistance of the infusion cannula is more than that
Hg should be used for 20-gauge systems and 650 mm Hg produced by 84 inches of connected tubing. Ohm’s law (volt-
for 25-gauge systems when removing vitreous. The lowest age = current × resistance [E = IR]) is mathematically equiva-
suction force that enables vitreous removal in a reasonable lent to Ohm’s law for fluid flow (pressure gradient = flow ×
time should be used to reduce vitreoretinal traction. The resistance). Fluidic resistance is an advantage in the aspira-
safest way to clear air lock and plugged tubing is by flush- tion system. By contrast, fluidic resistance is a disadvantage
ing the system outside the eye and not by increasing the in the infusion system because it produces a pressure drop
suction force. After the vitreous has been removed, higher between the infusion pressure and the intraocular pressure
suction force can be used to carefully remove adherent or (IOP), which must be compensated for. The pressure drop
previously delaminated ERMs. Suction-induced vitreoretinal occurs only during flow and increases with higher flow rates
traction is a key factor in the production of intraoperative (Fig. 2.2). It is typically 20 mm Hg or more during core

Fig ure 2 .2 ■ Intraoperative IOP


is d eterm ined b y infusion p res-
sure that is reduced by resistance
th roug h in fusion system wh ile
flow is occurring m inus vacuum
level at console reduced by resis-
tance in suction system including
cutter port interm ittent closing.
22 SECTION II ■ Surgical Technology and Techniques

vitrectomy. Conversely, the infusion pressure is equal to the infusion cannula must be inserted in a relatively firm eye so
IOP when there is no flow, that is, during membrane peel- that it penetrates completely through the choroid and nonpig-
ing, scissors segmentation or delamination, and endophoto- mented pars plana epithelium, as opposed to just the sclera.
coagulation. The highest flow rate and subsequent pressure Care must be taken to choose a location away from a preexist-
drop occurs with the 20-gauge fragmenter, excessively low ing choroidal or suprachoroidal hemorrhage or known abnor-
cutting rates, and nontapered extrusion cannulas. mality of the pars plana (trauma scar, persistent hyperplastic
primary vitreous (PHPV)). The cannula should be visualized
with the operating microscope or an indirect ophthalmo-
Effects of Low IOP
scope, not an unaided eye prior to use to avoid subretinal or
Excessively low IOP during vitrectomy is common in part suprachoroidal infusion from failure to visualize transparent
because of undue concern about the effects of higher pres- tissue over the cannula tip. The infusion cannula tubing must
sure. Low IOP causes pupillary constriction for unknown be adhesively fastened to the drape with a significant service
reasons; miosis often results in a need to perform potentially loop to prevent cannula dislocation from ocular rotation to
complication-producing techniques such as using iris retrac- see the periphery. The cannula must be placed close to the
tors and injecting intraocular epinephrine. Low IOP permits 3 o’clock or 9 o’clock position so that it will not be displaced
deformation of the corneal dome from the force of a handheld by the lower lid or speculum during ocular rotation.
irrigating contact lens; the author prefers an IOP of 45 mm Hg
unless there is low arterial perfusion pressure as occurs in
Infusion System Technologies
children or in some instances with general anesthesia. The
most significant complication of low IOP is bleeding from All systems available prior to the development of the Accu-
resected vessels, most commonly in diabetic traction retinal rus (Alcon, Inc., Ft. Worth, Texas) used gravity-based infu-
detachment cases or those with elevated neovascularization. sion systems. Gravity-based systems have three significant
Suprachoroidal hemorrhage during vitrectomy is virtually disadvantages: There is no digital readout of infusion pres-
always iatrogenic, caused by inadvertent suprachoroidal infu- sure, and surgeons cannot mentally convert inches (or
sion with secondary shearing of the short posterior ciliary ves- centimeters) of water to millimeters of mercury; IV poles
sels. Hypotony is 100% preventable during vitrectomy, which cannot be controlled by the surgeon; and motorized IV
is not the case with cataract surgery because of the need to poles are slower than the VGFI (vented gas-forced infu-
open the eye for IOL implantation. Suprachoroidal infusion is sion) and the more advanced Alcon Constellation Vision
preventable by careful attention to surgical technique during System pressurized infusion/aspiration system. The older
infusion cannula placement and rapid recognition of displace- gas-forced infusion is better than gravity-based systems
ment of the infusion cannula into the suprachoroidal space. because gas-forced infusion readily produces a direct digital
readout. The VGFI implemented on the Accurus was even
better because it allowed rapid decrease as well as increase
Effects of High IOP
of infusion pressure via surgeon foot pedal command. The
Excessively high IOP during vitrectomy can lead to the Constellation Vision System has servo-controlled IOP com-
occlusion of the central retinal artery and corneal edema. pensation (not infusion pressure). The Constellation Vision
Corneal edema is more likely to occur when the endothelial System calibrates the resistance of the infusion system dur-
cell count is low as a result of previous intraocular surgery, ing push-priming, measures actual flow using a proprietary
trauma, or Fuchs’ dystrophy. Corneal edema occurs almost sensing system, calculates fluid Ohm’s law in real time, and
instantaneously when the IOP is excessive but takes substan- adjusts infusion pressure to produce the selected IOP with
tially longer to resolve when the IOP is normalized. Fortu- ±2 mm Hg.
nately, corneal edema is an early warning sign of excessive
IOP before the retinal vessels come into view.
Asp irat io n Fluid ics an d Vit re o us Cut t in g
Once the retinal vessels are in view, the retinal vessels
can be monitored for interrupted flow. Pars plana vitrectomy requires both infusion and aspiration;
Intentionally elevated IOP is a useful technique to con- there are many parallels between infusion and aspiration flu-
trol bleeding when segmenting or delaminating vascularized idics because they are influenced by the same physical prin-
ERMs, resecting scar tissue, or removing a subretinal chor- ciples. Resistance to fluid flow is determined by the internal
oidal neovascular membrane. Rapid foot pedal control is diameter of a lumen or port, length of tubing, cannula or
essential when using transient high IOP to control bleeding. tool, and the cutter port opening and closing, cyclically
obstructing the port. Fluidic resistance is proportional to the
fourth power of the diameter (Hagen-Poiseuille equation)
Sa fety Preca utions for Infusion Ca nnula
and linearly related to the length. The impact of diameter
Pla cement
is very significant because of the fourth-power relation-
Wound construction is discussed extensively in another tuto- ship and clinically relevant because of the transition from
rial but several safety points are important to emphasize. The 20-gauge (0.89 mm) to 23-gauge (0.75 mm) or 25-gauge
CHAPTER 2 ■ Surgical System s, Tools, and Op erating Room 23

Fig ure 2 .3 ■ High cutting rates


cause p ort-b ased flow lim iting ,
which increases fluidic stability.

(0.5 mm) technology in recent years. The resistance of the producing desirable port-based flow limiting reduce the
cutter inner needle and the infusion port limit the flow far travel of uncut vitreous collagen fibers through the port.
more than the 84 inches of connected tubing. Ohm’s law, 25 gauge provides more resistance than 23 gauge because
voltage = current × resistance (E = IR), is mathematically fluidic resistance is proportional to the fourth power of the
equivalent to Ohm’s law for fluid flow; pressure gradient = diameter. Surgeons often incorrectly believe that 25-gauge
flow × resistance. vitrectomy is “inefficient” or produces insufficient flow rates
Port-based flow limiting is a term coined by the prin- when, in fact, it is safer because of less pulsatile vitreoretinal
cipal author that encompasses both the flow limiting result- traction.
ing from smaller diameter cutters and that caused by higher Port-based flow limiting relies on the same physical
cutting rates and biased closed duty cycles. Higher cutting principles as high-vacuum, low-flow phaco as first imple-
rates cyclically interrupt the flow through the port, thereby mented on Alcon MicroFlare ABS and MicroTaper ABS
increasing fluidic resistance (Fig. 2.3). High cutting rates, phaco systems, now the standard of care. High-vacuum,
and therefore higher fluidic resistance at the port, are ben- low-flow phaco produces better anterior chamber stability
eficial for all cases and all tasks because they increase flu- and decreased fluid surge after occlusion break, which are
idic stability, which, in turn, decreases pulsatile vitreoretinal directly analogous to the advantages of port-based flow lim-
traction on both detached (retinal motion) and attached iting for posterior vitrectomy.
retina and therefore iatrogenic retinal breaks. The principal
author refers to the amount of fluid that passes through the
Th e Ro le o f Vit re ct o m y Te ch n iq ue
port during an open-close cycle as “pulse flow.” High cut-
ting rates produce many small-volume pulses with much Phaco technique is largely based on using aspiration to move
less remote (far field) effects, that is, pulsatile vitreoretinal lens material away from the lens capsule (to prevent capsu-
traction, than those that occur using lower cutting rates. lar defects and vitreous loss). In marked contrast, the vitre-
Small pulse flow means that the vitreous does not have time ous cutter port should be moved to the vitreous rather than
to produce remote effects because of the Force = Mass × the vitreous pulled to the port using excessive flow rates
Acceleration (F = MA) relationship. Higher cutting rates (Fig. 2.4). Phaco surgeons must consciously focus on mov-
do not cut collagen fibers better; this is because the veloc- ing the port to the vitreous because their phaco experience
ity of the cutter does not increase with higher cutting rates teaches them the opposite approach.
on pneumatic cutters. In addition, port-based flow limit- Higher flow rates from larger diameter cutters are not
ing decreases surge, and therefore iatrogenic retinal breaks, more (or less) efficient; efficiency is defined as the volume of
after sudden elastic deformation of the dense ERM or scar vitreous removed per volume of infusion fluid. Similarly, effi-
tissue through the port. High cutting rates in addition to ciency is not a function of cutting rate; efficiency is entirely
24 SECTION II ■ Surgical Technology and Techniques

Fig ure 2 .4 ■ Cutting while pulling away increases vitreoretinal Fig u re 2 .5 ■ Cutting wh ile ad van cing shears tissue in its
traction from vacuum force at cutter port. orig inal position, which m inim izes vitreoretinal traction.

driven by technique. Keeping the port constantly immersed costs. Cleanup of any tool with a lumen that includes cutters,
in vitreous produces efficiency. scissors, forceps, and cannulas has the potential of creating a
The author refers to the optimal technique as continu- TASS (toxic anterior segment syndrome)-like inflammation
ous engage and advance vitrectomy. Current emphasis on from biological materials from previous patients, enzymes
efficiency and faster operating times can result in the unin- used in ultrasonic cleaning, autoclave water impurities, and
tended consequence of pulling the cutter back while aspirat- the like. In addition, vitreous cutters, scissors, and forceps,
ing, greatly increasing vitreoretinal traction (Fig. 2.5). especially in smaller form factors (23 and 25 gauge), have
fragile cutting and gripping surfaces that are damaged by
Ph ysics o f Vit re ct o m y cleaning and sterilization process.
The author developed the InnoVit dual actuation
Vitreous is a very complex tissue with low homogeneity; the scheme to eliminate the spring used to open the port after
physical properties vary widely from patient to patient and the pressure pulse on the diaphragm closed the port. Elimi-
disease to disease and change dramatically as the vitrectomy nation of the spring increased cutting rates and cutter veloc-
progresses. Vitreous hyaluronan acts as a Non-Newtonian, ity at the time of closure. The InnoVit utilized a limited angle
pseudoplastic fluid similar to viscoelastic agents in the ante- rotary cutting scheme rather than an axial (guillotine) cut-
rior chamber resisting deformation into the cutter port. Early ting action. The UltraVit developed for the new Alcon Con-
in the vitrectomy, surgeons often believe that “nothing is stellation system uses a diaphragm-based, dual actuation,
happening” when in fact hyaluronan is being removed, and axial cutting design (Fig. 2.6). Duty cycle is defined as the
they may react by unsafely increasing flow rates, usually by percentage of port open time versus total time. Lower duty
decreasing the cutting rate. Hyaluronan acts as a dampening cycle results in more port-based flow limiting, and there-
agent reducing vitreoretinal traction from pulsatile flow fore fluidic stability, and less pulsatile vitreoretinal traction.
through the port. Hyaluronan is diluted as the vitrectomy
progresses, decreasing the dampening effect, clearly an issue
because vitreous cutting close to the retina is typically per-
formed after core vitrectomy. Furthermore, infusion fluid
changes the electrochemical properties of the vitreous, dra-
matically decreasing its viscosity. It is of interest that vitre-
ous viscosity is reduced by a factor of five in minutes after
removing it from the eye or enucleating an animal eye.

Cut t e r Te ch n o lo g y
Pneumatic cutters are much lighter and more compact than
electric cutters, thereby improving dexterity (Weber-Fechner
law) and decreasing hand fatigue. Although this is commonly
misunderstood, disposable tools actually reduce per-case
costs because they eliminate cleaning, rinsing, drying, wrap- Fig u re 2 .6 ■ Cutaway view of Alcon UltraVit 5,000 cp m
ping, sterilization, storage, replacement, and spare parts cutter.
CHAPTER 2 ■ Surgical System s, Tools, and Op erating Room 25

Higher duty cycle produces greater flow and more pulsatile the Alcon Constellation (Fig. 2.7). Vacuum response to a foot
vitreoretinal traction, suitable only for core vitrectomy. The pedal command to decrease vacuum is far more important
Alcon Constellation UltraVit currently cuts at 5,000 cuts per from a safety perspective than a command to increase vac-
minute and has variable duty cycle control, enabling the uum. Response time is driven by many factors: size of vac-
control of port-based flow limiting independent of cutting uum chamber in the cassette, proportional valve(s) response
rates. time, embedded controller, and use of a real-time operating
system. The Alcon Accurus had the first real-time operating
system and distributed embedded processing resulted in a
Asp irat io n Te ch n o lo g y
25-millisecond response time compared to ten times lon-
The VISC (vitreous infusion suction cutter), RotoExtractor ger for the closest competitor. The Constellation Vision Sys-
Vitreophage, and other early vitrectomy machines used a tem has flow control using flow sensing and servo feedback
syringe operated by the assistant to produce aspiration. This control and is twice as fast as the Accurus. This technology
method was highly imprecise and even dangerous because produces rapid, nonpulsatile control unlike the pulsatile,
of stiction in the syringe and control by someone other than slower flow control produced by a peristaltic pump system.
the surgeon. Conor O’Malley and Ralph Heinz developed In addition to flow control, there is a flow-limiting mode;
the Ocutome 800 that utilized on-off foot pedal control of these systems will increase safety near the retinal surface,
preset vacuum, which was a huge improvement. The author especially with mobile retina. Port-based flow limiting pro-
developed linear (proportional) vacuum for the Ocutome duced by high cutting rates (5,000), smaller lumens (23,
8000 working with engineers at CooperVision. The Ocular 25, and 27 gauge), and now variable duty cycle control
Connection Machine (OCM) system that the senior author is instantaneous, while console-based flow control must
developed with InnoVision had extremely fast fluidics but interact through two-way pass of the fluidic signal through
was never commercialized; fortunately, much of the technol- 84 inches of compliant tubing.
ogy was incorporated into the Alcon Accurus system, and It has long been incorrectly taught that cutting rates
the entire package is implemented and improved upon on should be reduced when removing dense ERMs, scar tis-
sue, and lens material or performing core vitrectomy. The
surgeon reaction time from seeing a visual target to mov-
ing a foot pedal is approximately 400 milliseconds because
of visual and cognitive processing, generation of a motor
response, propagation through the spinal cord and leg, and
contraction of the lower leg muscles. The surgeon reaction
time is more than an order of magnitude longer than the
25 milliseconds it takes a modern venturi-based system to
respond to a foot pedal command. The senior author has
calculated that approximately thirty times the volume of the
fluid contained between the tip and the port goes through
the port in the time that occurs between when the surgeon
decides to lift the pedal and when the vacuum is actually
reduced at the cutter port.

Cutter Movement
Many surgeons habitually pull back the cutter while suc-
tion is applied, which greatly increases vitreoretinal traction.
Some surgeons have recommended pulling the cutter away
from vitreous while cutting, but this technique is dangerous
because the force created by probe movement is added to
the force created by suction. This is particularly dangerous
when slow cutter speeds, relatively high suction levels, or
malfunctioning cutters are used. In cataract or penetrating
keratoplasty, vitrectomy cutting while pulling the probe out
of the vitreous has the added disadvantage of creating air-
lock as air is introduced into the system. If material becomes
caught in the port, the surgeon should not jerk the probe
back (“burned-hand” reflex) but should wait and have an
assistant squeeze the suction tubing or, better yet, use the
Fig ure 2 .7 ■ Alcon Constellation Vision System . foot pedal reflux mode.
26 SECTION II ■ Surgical Technology and Techniques

With the Accurus or especially the Constellation, Vacuum systems controlled by the surgeon’s foot offer
accurately controlled low suction force allows the port a major advantage over surgeon’s hand or assistant control.
to be turned toward the retina or 90 degrees rather than Suction force is analogous to the pressure one places on a
180 degrees away. The probe should be advanced toward knife or scissors to cut and should be directly controlled by
the tissue to cut, just as one would use any other power the surgeon. The maximum vitreoretinal traction is under
tool. Excessive suction must be applied if the port is turned constant control with a controlled vacuum system. Control-
anteriorly, away from the vitreous to be cut. The combina- ling the vacuum with a button on the cutter causes inadver-
tion of the high-performance cutters, advanced fluidics, tent hand movements and operator fatigue. Foot-controlled
and especially high-speed cutting achieves the ultimate solenoid valves interrupt the connection of the suction port
goal of cutting tissue in its original position. Surgeons to a collection bottle at a preset vacuum level but create an
accustomed to the vitreous being drawn to the port at abrupt onset of suction and inability to decrease suction
first may think that these systems are not cutting, but later without stopping the process. Better control is made possible
they learn to move the cutter to the tissue to be removed. by having the foot pedal proportional to the suction force.
The authors refer to the optimal approach as continuous Simply stated, pedal position correlates with suction force.
engage and advance, emphasizing the need to take the port Proportional suction control, frequently called linear suc-
to the vitreous, never pull back while cutting, and avoid a tion (3,4), permits continuous optimization of the suction
start-stop approach. force rather than using the circulating nurse for resetting the
level of suction. A preset maximum should be utilized, with
a constant use of just enough suction to remove vitreous as
Port Confi gura tion
judged visually. Readout of this vacuum-produced force is
Large ports decrease the force per unit area but allow the available on the console but seldom observed because the
entry of larger tissue pieces without cutting or deformation correct force is judged by viewing vitreous flowing into the
of the tissue. Ports closer to the tip are advantageous only port and unwanted retinal movement.
when high cutting rates and high-performance cutter/flu- A distinct advantage of linear suction is the control over
idic systems permit safe operation near the retina. Although the rate of increase, and especially decrease, of the suction
most ERMs are removed by delamination or forceps mem- force. The suction force is gradually increased until vitre-
brane peeling, cutter operation near the retina is especially ous movement into the port is just achieved. Fast response
advantageous in removing ERM in diabetic traction detach- time is essential for proportional (linear) suction systems.
ment cases using conformal cutter delamination or foldback The Constellation and Accurus systems have been opti-
delamination. mized to decrease the vacuum rapidly (25 ms) in response
to a foot pedal command to reduce suction, without the
need to stop cutting. This precise control requires match-
Self-Sha rpening
ing dynamic port resistance, cutting rate, tubing resistance,
Higher force-to-weight ratio pneumatic actuators are pre- and console fluidics. A real-time operating system imple-
ferred over electric actuators to drive self-sharpening cutters. mented on a dedicated fluidics processor provides guaran-
Self-sharpening cutters remain sharp throughout the proce- teed response time. The Accurus system and especially the
dure. Any self-sharpening cutter must be disposable because Constellation system coupled with the high-speed cutters
self-sharpening results in loss of metal and, ultimately, fail- enable safe dissection with the probe virtually touching the
ure of the probe. retinal surface.
There has been an evolution of systems utilized to con-
trol suction force for vitreous surgery. Manually operated
Vitreous Cutter Design Issues
syringes controlled by the assistant provided excessive fluc-
tuation of suction force because of syringe friction and a vol- The initial electric motor–powered, nondisposable vitreous
ume/flow rather than pressure control. Manually operated cutters such as the Machemer-Parel VISC and the Douvas
syringes caused large swings in TOP and excessive vitreo- RotoExtractor utilized continuous rotary motion and non-
retinal traction. Use of the mechanical syringe drive offered disposable cutters. This combination often produced marked
better control because of its mechanical advantage. Peristal- winding and vitreoretinal traction as the cutters began to have
tic pump systems primarily control flow rather than pres- wear-induced poor apposition of the shearing surfaces. Non-
sure and typically undergo transient peaks of high TOP as disposable axial cutters such as the O’Malley-Heinz Berkley
materials are impacted in the port. As the material deforms Bioengineering Ocutome eliminated the winding problem
and moves rapidly through the port, this excessive pulse of noted above but did not solve the wear-induced poor cutting
“residual” suction force is transmitted to the surrounding vit- problem. Disposable, pneumatic, axial cutters, also referred
reous, creating undue vitreoretinal traction. Venturi pumps to as guillotine cutters, such as the Alcon Accurus and Alcon
are safer than peristaltic or scroll pump–based vitrectomy Constellation UltraVit are self-sharpening, which ensures
systems and reduce pulsatile (i.e., peristaltic) vitreoretinal that all vitreous collagen fibers flowing through the port are
traction. cut if cutting rates are sufficiently high.
CHAPTER 2 ■ Surgical System s, Tools, and Op erating Room 27

The initial vitreous cutters utilized brush-type direct


current (DC) motors that are too costly to be disposable. The
nondisposable Ocutome utilized a bellows pneumatic actua-
tor, which dramatically reduced the weight and size of the
cutter system from hundreds of grams to 22 g. Diaphragm
(Accurus, Constellation UltraVit), dual-activated piston
(InnoVit), and Alcon Constellation UltraVit dual-actuated
diaphragm cutters weigh less than 10 g. Pneumatic actuators
produce roughly ten times the force per unit mass and force
per unit volume compared to electrodynamic actuators.
Fatigue is reduced and tactile sensing and dexterity increased
with lighter finger and hand loading. Shorter handles reduce
torque on the hand due to cable/tubing friction on the drape
as well as unintentional pulling by the assistant.
Pneumatic actuators have much lower moving mass than Fig ure 2 .8 ■ 25-gauge infusion cannula.
direct electromagnetic actuators such as solenoids. Higher
moving mass reduces acceleration, assuming a constant force
Infusion Ca nnula s
(F = MA, where F is force, M is mass, and A is acceleration).
Swash plate cams driven by a continuous rotary motor have Although an infusion sleeve can be slipped over a 20-gauge
velocities limited by the cam follower “floating” as well as cutter to restore the full function probe concept, this is only
by spring return force limitations. Although some advocates rarely useful for selected anterior vitrectomy scenarios. Infu-
have stated that the motion of a cam-based cutter is a triangle sion cannulas through the pars plana create far less turbu-
wave, this is impossible, because it would require infinite lence and decrease fluid throughput (Fig. 2.8). Separation
acceleration when the cutter reverses direction. Electromag- of the infusion function from the vitreous cutter decreases
netic actuator forces are limited by the heat produced by I2R its size and enhances flexibility. Proportional (linear) suction
(current squared times resistance) losses in the motor wind- connected to various extrusion cannulas (Chapter 4) is far
ings. Electrically driven cutters become noticeably hot when more efficient with separate infusion and does not require
driven at high cutting rates. The spring return on axial cutters the presence of the vitrectomy instrument in the eye to
reduces the velocity as the port approaches closure (Hooke’s provide infusion. The wide separation of the infusion and
law, F = −KY, where F is force, K is the spring constant, and egress ports decreases turbulence and operating time when
Y is the displacement). The Alcon InnoVit cutter (US Patent removing blood products from the eye. Internal fluid-air/
5,176,628) utilizes dual pneumatic actuation to eliminate the gas exchange and internal drainage of subretinal fluid (SRF)
spring slowdown problem and a gear-driven cutter to pro- (Chapter 4) are similarly more efficient with a separate
duce limited rotary motion, which eliminates winding. Rotary infusion system. If a retinal break is present or occurs dur-
motion allows the port to be closer to the tip because an over- ing vitrectomy, the SRF will increase when exposed to the
shoot distance is not needed for the moving cutter element probe-located infusion of the full function probe, while the
to slow down and reverse direction. The Alcon Constellation separate infusion system approach results in decreased SRF
UltraVit cutter utilizes a dual-actuated diaphragm cutter that if the cutter is brought near the break. The infusion system
eliminates the spring, eliminates the friction of the InnoVit is placed without the vitrectomy probe, following the “first
piston seals, and enables variable duty cycle control. in, last out” rule to provide infusion and pressure control
throughout the case.
Infusion into the suprachoroidal or subretinal space is
In fusio n Sit e Op t io n s
the only complication unique to the infusion cannula sys-
Infusion instruments, as well as other vitreous surgery tem. These complications are manageable but, more impor-
instruments, can enter the eye through the limbus, ciliary tantly, preventable by careful technique (Chapter 4). Longer
body, or pars plana. (4 mm) cannulas than the 2-mm cannulas originally recom-
mended reduce the chance of inadvertent suprachoroidal
or subretinal infusion and do not strike the lens, unless the
Full Function Probes
cannula is pushed by excessive rotation of the eye, causing
The generic term “vitreous infusion suction cutter” (VISC) contact with the lids, or if the surgical assistant pulls on the
implies the full function probe concept. While having all tubing by mistake.
functions on one probe was an original goal in the design of
vitrectomy instruments (5–7), in almost all situations, sepa-
Alt e rn at ive In fusio n De vice s
ration of functions is more flexible and permits smaller inci-
sions (8,9). Restated, three-port vitrectomy is preferred to A 30-degree bend in a blunt, end-opening, handheld can-
one-port vitrectomy for virtually all situations. nula permits access to the anterior chamber via a pars plana
28 SECTION II ■ Surgical Technology and Techniques

entry site over the bridge of the patient’s nose (10). These
cannulas can be used for infusion when the pars plana can-
not be visualized, as in most ROP cases, preexisting supra-
choroidal hemorrhages, pars plana abnormalities, and some
cases of anterior vitrectomy.
Sharp infusion needles risk inadvertent puncture of
ocular structures and damage to the cutter. The bevel can
leak fluid when the tip is near the wound and can infuse into
the choroid or cornea. Scalp vein needles (butterfly needles,
winged infusion sets) and silicone tubing can inadvertently
contact the corneal endothelium and should be avoided.

In fusio n Fluid
Infusion fluids that include a bicarbonate buffer, dextrose,
and glutathione in addition to the usual components of
Fig u re 2 .9 ■ En d oillum in ator elim inates lig h t scatter that
Ringer’s solution should be considered mandatory in all cases
would occur with transcorneal illum ination and allows the light
(Alcon Laboratories BSS Plus) (11). The tubing and connec- source to be m oved around for optim al focal, d iffuse, specular,
tors must be handled with meticulous, sterile technique. and retro illum ination.
Bubbles should be prevented by flushing, and care should
be taken to avoid negative pressure in infusion bottles. Cer-
tain surgeons use lactated Ringer’s solution or balanced salt than the cutter and allows illumination of any area of interest.
solution (BSS) rather than BSS Plus in a misguided attempt Retro, focal, and specular illumination with the endoillumi-
to save money. High labor cost due to slow surgery, not infu- nator allows better visualization of clear vitreous and pro-
sion fluid, is the most significant cost driver, yet it receives duces much less scattered light (glare) than transcorneal slit,
less emphasis because it points back to the surgeon. Glucose chandelier, or torpedo illumination. Safety is increased by
was initially added to the infusion fluid before the advent of the ability to illuminate peripheral vitreous during mem-
frequent blood glucose testing during surgery. Twenty years brane or vitreous removal. Diathermy can be added to the
ago, the procedure was to utilize intravenous dextrose 5% disposable endoilluminator, which is ideal for diabetic cases
in water to prevent hypoglycemia during local or general still using 20-gauge systems. Various divergence angles can
anesthesia. The principal author has not added anything be used for a spot or flood light effect. Wide-angle illumina-
to the infusion fluid for over 25 years and never observes tion devices produce more uniform illumination for video
significant posterior subcapsular changes. The safety of the but make visualization of clear vitreous more difficult. Wide-
cumulative dose of antibiotics or epinephrine in the infusion angle illumination using a standard 78-degree endoillumina-
bottle has not been established. The principal author has tor is essential when using wide-angle visualization systems
performed over 26,000 vitrectomies, with only three cases for peripheral viewing in rhegmatogenous retinal detach-
of postoperative endophthalmitis. The author (S.C.) had ment, proliferative vitreoretinopathy (PVR), and giant break
two cases of endophthalmitis in 1975; one case had severe cases.
oral sepsis, and multilayer drapes were used, permitting the
patient to exhale on the operative site. The second case had Erg o n o m ic Issue s
an infected suprapubic cystotomy and, in retrospect, should
have been postponed until the systemic infection had been All hand instruments should be held in three fingertips. Scis-
successfully treated. The third case occurred when no sub- sors and forceps should rest on the web of tissue extend-
conjunctival antibiotics were used at the end of the case, an ing from the base of the thumb to the second digit, while
experiment that was immediately terminated. The authors vitreous cutters should be shorter to permit greater angular
never add epinephrine or any other agent to the infusion motion. Fingertips are softer and have better tactile sensa-
fluid because of the risk of contamination and toxicity and tion than the bony portions of the fingers. Triangulation of
the possibility of the incorrect dose or agent. instruments between three fingertips and this web is the
most stable grip and occupies approximately 35 mm of han-
dle length. Any handle or cable extending beyond this grip
En d o illum in at io n
moves the center of gravity behind the grip point and cre-
Full function probes utilize a coaxial fiberoptic illumination ates torque on the fingers. Extension beyond 35 mm gives
system. While this is vastly superior to microscope illumina- dangerous leverage to an assistant inadvertently striking the
tion, it is inflexible and illuminates only the cutter tip and handle or pulling on the cable, tubing, or fiber. All tools
surrounding vitreous. A separate endoilluminator (Fig. 2.9) should have a concavity or another means of axially con-
may be used bimanually with intraocular instruments other straining the grip. Axially constraining the grip allows the
CHAPTER 2 ■ Surgical System s, Tools, and Op erating Room 29

surgeon to loosen the grip and reduce fatigue without fear of temporomandibular joint discomfort. Zoom, microscope
the probe slipping. Consistent tip to grip distance facilitates light on-off, and XY reversal for wide-angle systems must
cerebellar learning (so-called muscle memory) for the sur- be controlled on the foot pedal. Speech recognition systems
geon and reduces bumping the retina. An hourglass-shaped have been used to control microscope position but are ill
probe (Developed by Steve Charles, September, 1976) suited to control any of the real-time tasks. An additional
wedges between the fingers, reducing the force required to observer tube can be utilized for teaching other surgeons,
prevent slippage in either direction. technicians, or nurses. High-quality television in the OR is
The lighter the probe the better; a heavier electrically very advantageous for team coordination and in-room teach-
driven cutter creates excess loading of the proprioceptive ing, but recording is overutilized.
sense (Weber-Fechner law) and decreases sensitivity. Excess
weight coupled with excessive length is extremely muscle
En d o p h o t o co ag ulat io n
fatiguing and tremor inducing. Anxiety and caffeine accen-
tuate natural tremor as well. Pneumatically driven, dispos- Endophotocoagulation was initially developed for retin-
able, self-sharpening, hourglass-shaped, small, lightweight opexy and panretinal photocoagulation (PRP) (14). The
probes are the best design compromise. Nonslip surface of principal author developed a technique of coagulating
the cutter allows easier grip by the surgeon and improves retinal surface bleeders in diabetic cases with the 20-gauge
control of the instrument with less firmness of grasp. Chang end-aspirating endophotocoagulation probe but
now uses 25-gauge laser probes for hemostasis without
aspiration. Although some surgeons prefer the laser indi-
OPERATING MICROSCOPE rect ophthalmoscope (LIO) to endophotocoagulation, the
LIO can cause cornea, lens, and iris damage; longer operat-
An operating microscope with power zoom, focus, and ing time; and surgeon neck strain. The far periphery can be
two-axis power translational (XY) or rotational (pitch, yaw) visualized extremely well with wide-angle viewing systems;
movement is necessary for vitrectomy (Fig. 2.10) (12). using an LIO is unnecessary and has no advantages in the
Coaxial stereo assistant’s microscopes are essential for vit- majority of cases. A 532-nm, diode-pumped, frequency up-
rectomy. The power XY system can be controlled by a converted laser is currently the best choice for endophoto-
three-axis, six-direction chin switch (13), but this can cause coagulation.

Fig ure 2 .1 0 ■ Zeiss operating m icro-


scop e with m otorized XY m ovem ent,
stereo observer scope, em bedded 3-CCD
television cam era, and sterile controls for
releasing brakes to enable m anual posi-
tioning of the m icroscope.
30 SECTION II ■ Surgical Technology and Techniques

Diat h e rm y The surgeon’s hands rest on the patient’s face during


vitrectomy and not directly on the “wrist rest.” The wrist rest
Diathermy is the term used to describe the use of radio frequency
is used primarily to create a trough in conjunction with the
(RF) energy to produce heat and thereby coagulate proteins.
drape, permitting continuous aspiration of fluid runoff from
Although intraocular diathermy can be used for retinopexy,
the eye. The trough is a necessity to protect the foot pedals
the primary application is hemostasis. Bipolar means all the RF
from water damage, to contain contaminated fluids, and to
energy must pass between two intraocular electrodes within
catch dropped instruments. A high position of the wrist rest
the eye rather than between one electrode and patient ground.
will result in retinal or lens damage if the patient turns his or
Bipolar is safer than monopolar because RF might propagate
her head from side to side.
through the optic nerve with monopolar diathermy. The Alcon
The microscope should usually be placed on the patient’s
disposable bipolar endoilluminator and its predecessor, the tis-
right side to create a tent with the drape under which the
sue manipulator, have a small antenna as one electrode and the
anesthesia personnel can visualize the airway and EKG leads.
tool outer shank as the return path. This small antenna can be
An assistant can be located on the right or left side of the
thought of as “focused” diathermy as is preferred to the standard
patient, but the left side is preferable because of access to the
intraocular bipolar with a symmetrical electrode configuration.
console and instruments. Hand instruments are best kept on
The principal author found the disposable bipolar endoillumi-
a Mayo stand between the surgeon and the assistant on the
nator to be an ideal “second” instrument during the delamina-
left, with a back table placed behind for other surgical tools
tion of diabetic traction retinal detachments before switching
and disposables. The vitrectomy system and 532-nm diode-
exclusively to 25-gauge surgery for all cases.
pumped laser are stacked on a cart at the patient’s left hand,
with a sterile shelf or Mayo stand in front for the associated
OPERATING ROOM SETUP handpieces and tubing. This stand should be placed over
the patient’s chest with the bipolar handpiece, contact lens,
The operating table and head support must be very rigid contact lens suction, and infusion tubing, after the patient is
and stable and have minimal thickness between the patient’s prepped and draped. The Mayo stand over the patient’s chest
head and the surgeon’s knees. The surgeon must sit up creates a tent that allows the anesthesiologist to visualize the
very straight, stretching to reach the microscope in order to chest wall movement, abdomen, airway, and EKG leads. This
decrease all-too-common cervical and lumbar spine prob- setup is used regardless of which eye is to be operated upon,
lems caused by leaning and bending forward. The ideal thus permitting more rapid and consistent setting up.
surgeon chair or stool should have back support and no Placing the equipment on a rack over the patient pre-
armrests, and the shape and elasticity of the seat should exert vents the surgeon from viewing the console display. In addi-
no pressure on the sciatic nerves or perineum. The chair or tion, access to the patient is limited in the case of a medical
stool must have a foot pedal or, even better, a full circle foot emergency or restless patient. A modern vitrectomy system
control for height adjustment by the surgeon. such as the Alcon Constellation Vision System (Fig. 2.11) is

Fig ure 2 .1 1 ■ Standardized OR


config uration enab les rap id and
efficient setup and takedown.
CHAPTER 2 ■ Surgical System s, Tools, and Op erating Room 31

best placed at the patient’s left hip. Dual xenon light sources St e rilizat io n Syst e m s
are embedded in the Constellation system.
The steam autoclave, when properly utilized, kills bacteria,
Microscope drapes are essential to prevent fibers, wires,
fungi, parasites, and prions (transmissible spongiform enceph-
tubing, and instruments from being contaminated. The micro-
alopathy, bovine spongiform encephalopathy). Vaporized
scope is handled by many people and is directly over the
hydrogen peroxide (Sterad) and washer disinfecting, spori-
patient, and it is therefore a potential source of contaminated
cidal (Steris) systems cause less damage to certain instruments
particles falling on the eye.
than the steam autoclave but probably do not kill prions.
Powder-free gloves should be used on all cases. Spe-
cific care must be taken for patients with latex allergy. Care
should be taken to prevent cotton fibers, plastic particles, Surg ical Effi cie n cy
and cellulose material from touching the instruments. The
Reduced operating, setup, cleanup, and turnover time
authors almost never touch the eye with cellulose sponges or
reduces labor costs. Standardization and stable staffing
cotton-tipped applicators to avoid particulate matter being
reduce training time and associated costs, errors, and mate-
left in the eye. Surgeon masks with an integral plastic flap
rials waste. Intuitive user interfaces allow the utilization of
to prevent fogging are easier on the skin than foam rubber
less-skilled staff and reduce errors and surgeon frustration.
or adhesives. It has been reported that there is a higher inci-
It is the surgeon’s responsibility to lead by example and
dence of facial basal cell carcinoma in surgeons who taped
encourage a team approach rather than blaming, criticizing,
their mask for many years.
and becoming angry.
Use of a single-piece drape with an integral fluid collec-
tion trough is the most efficient method of draping. The drape
should be transparent over the airway and face for safety. The References
drape should be thicker than typical drapes to avoid inadver- 1. Banko A. Apparatus for removing blood clots, cataracts, and
tent perforation. Head drapes and other drapes are not needed, other objects from the eye. United States Patent 3,732,858.
saving time and money. Drapes without an opening for the eye Filed February 14, 1969.
should be used and a cut made in the drape after the drape 2. Machemer R, Buettner H, Norton EW, et al. Vitrectomy: a
is adhesively applied with the lids open. The flaps created by pars plana approach. Trans Am Acad Ophthalmol Otolaryngol
1971;75:813.
this method are then folded over the lid margins and held in 3. Charles S, Wang C. Linear suction control system for the vitre-
place by the speculum to keep cilia and lid bacteria away from ous cutter. Arch Ophthalmol 1981;99:1613.
the operative site. The skin must be degreased and dry, or 4. Charles S, Wang C. Motorized gas injector for vitreous surgery.
the drape will not adhere. The drape must first be applied to the Arch Ophthalmol 1981;99:1398.
superior orbital ridge, then the nasal canthus, and finally the 5. Peyman GA, Dodick NA. Experimental vitrectomy. Arch Oph-
thalmol 1971;86:548.
zygoma and outer portion of the orbital ridge in order to have 6. Douvas NG. The cataract roto-extractor (a preliminary report).
a complete seal. A complete seal prevents water from running Trans Am Acad Ophthalmol Otolaryngol 1973;77:792.
into the patient’s hair and ears and, more importantly, prevents 7. Federman JL, Cook K, Bross R, et al. Intraocular microsurgery 1:
exhaled air from reaching the eye. The patient’s exhaled air is a new instrumentation (SITE). Ophthalmic Surg 1976;7:82.
sterility risk and causes fogging of the contact lens. Tincture of 8. O’Malley C, Heintz RM. Vitrectomy via the pars plana—a new
instrument system. Trans Pac Coast Otoophthalmol Soc Annu
benzoin can be used to increase drape adherence. Meet 1972;53:121–137.
9. O’Malley C, Heintz RM. Vitrectomy with an alternative instru-
ment system. Am Ophthalmol 1975;7:585–588.
Pre p Te ch n iq ue 10. May DR. Anterior chamber infusion with the 30 degree bent
Povidone-iodine (Betadine 5%) prep should be used even if needle. Ocutome Fragmatome Newsletter 1979;4:4.
11. Edelhauser HF, VanHorn DL, Schultz RO, et al. Compara-
the patient has an iodine allergy. Betadine drops should be tive toxicity of intraocular irrigating solutions on the corneal
used for the cul-de-sac before and immediately after surgery. endothelium. Am J Ophthalmol 1976;81:473.
Lash trimming is no longer performed because it has been 12. Parel J-M, Machemer R, Aumayr WA. New concept for vitreous
reported that it increases bacteria loading in the conjunctival surgery for automated operating microscope. Am J Ophthalmol
cul-de-sac, emphasizing the need to cover the lashes with the 1974;77:161.
13. Charles S, McCarthy C, Eichenbaum DA. Chin operated switch
drape. The corneal epithelium should be covered with the lids for motorized three axis microscope movement. Am J Ophthal-
or irrigated frequently with BSS to markedly reduce the need mol 1975;80:150.
for scraping. The authors almost never need to remove the 14. Charles S. Endophotocoagulation. Retina 1981;1(2):117–120.
corneal epithelium. There is little, if any, evidence that vis-
coelastics preserve the corneal epithelium better than BSS.
C H AP TER
3
CONSTELLATION ARCHITECTURE

The Constellation is the culmination of over fragmenter, silicone injector (VFC), and a halogen light
three decades of evolutionary development of source. 25-gauge and later 23-gauge tools were developed
vitreous cutters and fluidics, new and improved for the Accurus platform and are now in their third genera-
tools, illumination sources, phaco technology, tion. A nonintegrated EyeLite 532-nm diode-pumped laser
systems integration, efficiency systems, and advanced user and later a nonintegrated xenon source were developed to
interface design. The principal author is the architect of the use with the Accurus system.
Alcon Constellation Vision System. The Constellation incorporates new, improved imple-
The vitreous infusion suction cutter (VISC) and Roto- mentations of all the concepts incorporated in the OCM as
Extractor were single-port, large-incision, so-called full well as the Accurus and adds many new capabilities. The
function, slow-speed, rotary, electric cutters with aspiration vitreous cutter is the pivotal component of any vitreoretinal
provided by a syringe operated by the assistant. The Berkley surgery system; the Constellation UltraVit cutter utilizes dual
Bioengineering Ocutome 800, developed by Conor O’Malley pneumatic actuation (no spring return axial cutter) such as
and Ralph Heinz, was the first three-port, 20-gauge (0.89 the InnoVit and currently operates at 5,000 cuts per minute
mm) system and had the first lightweight, pneumatic probe (Fig. 3.1). Because it is diaphragm driven, it eliminates the
and surgeon foot pedal–controlled, on-off aspiration, a major friction of the InnoVit piston, provides the familiar axial cut-
advance. Berkley Bioengineering subsequently was acquired ter format, and scales better to 23-, 25-, and even 27-gauge
by Coopervision, and Coopervision was later acquired by form factors. The cutter is driven using a proprietary variable
Alcon. The Coopervision Ocutome 8000, developed by duty cycle technology dependent on the dual actuation sys-
Carl Wang, his engineering team, and the author, had the tem. Variable duty cycle control enables the use of a biased
first linear suction (now used on all vitrectomy and phaco closed approach to produce increased port-based flow limit-
machines), an integrated light source, and a connected ing when working in close proximity to the retina, so-called
fragmenter. The MidLabs MVS system, developed by Carl vitreous shaving. Alternatively, port-based flow limiting can
Wang and the author, had the first disposable pneumatic be decreased by using a biased open approach enabling
cutter, a crucial improvement over reusable cutters with greater flow rates when doing core vitrectomy. The aspi-
low-performance cutting. After the original MidLabs sys- ration system has an extremely low latency response time
tem was acquired by Alcon Laboratories, the author started to a foot pedal command for vacuum decrease or increase
InnoVision and began the development of the Ocular Con- because of a new, triple proportional valve aspiration sys-
nection Machine (OCM). The OCM had a dual actuation tem and cassette design. Sensor-based, fast-response, digital
InnoVit cutter with limited angle rotary cutting at 1,500 flow control and flow limiting facilitate working safely near
cuts per minute, linear diathermy, tool ID, an articulated the retinal surface. The aspiration system provides continu-
arm with integrated tubing management, servo-controlled ous linear (proportional) reflux as well as microreflux for all
intraocular pressure (IOP), a graphical user interface with aspiration tools.
soft keys, integrated xenon illuminator, integrated frag- The Constellation utilizes a real-time operating system
menter, auto–gas mixing, auto fluid-air exchange valving, and distributed processor architecture to ensure reliability
and power scissors. The InnoVision OCM technology was as well as 2× faster response time than the Accurus. It has
never commercialized and was later acquired by Alcon Lab- a switched Ethernet architecture with 42 printed circuit
oratories, and I became a consultant for Alcon. Many of the boards, a Pentium, five microprocessors, and many Field
OCM concepts were improved upon and incorporated into Programmable Gate Arrays and uses over 600,000 lines of
the highly successful Accurus system, which included an code. The electronics have power backup for situations such
advanced graphical user interface with soft keys and global as the cord being pulled out or power failure in the operating
functions, VGFI (vented gas-forced infusion), integrated room (OR) to facilitate resumption of surgery.

32
CHAPTER 3 ■ Constellation Architecture 33

Fig u re 3 . 1 ■ Cu t away view


of UltraVit 5,000 cp m vitreous
cutter showing d ual d iap hrag m
actuation.

Integrated pressurized infusion using a two-chamber


system and servo control of the IOP is unique to the Con-
stellation and is especially valuable in high-flow scenarios
such as removal of dislocated dense lens fragments using
the fragmenter with 23- or 25-gauge infusion. IOP compen-
sation will reduce the sudden IOP decrease and resultant
bleeding after dense epiretinal membrane deforms through
the cutter port. It is likely that IOP control will enable the
use of lower average infusion pressures. The infusion system
has automatic bottle-out warning and enables changing the
bottle with no interruption in fluid flow or bubbles.
The system can be configured with one or two dual-port
xenon illumination sources to facilitate the use of illuminated
tools, chandeliers, and Torpedoes (Fig. 3.2). The new xenon
illuminator design is more efficient and produces longer, 400-
hour lamp life. The xenon optical system produces greater
than 25 lumens using 23- and 25-gauge fibers at 200 hours.
Radio frequency identification (RFID) connectors on the illu-
mination tools automatically adjust the initial xenon source
intensity depending on specific tool characteristics: light
throughput, typical working distance, and divergence angle.
This set point is 8 to 10 lumens, optimal light intensity for all
tools in 20, 23, and 25 gauges. The surgeon can increase illu-
mination, if needed, to the maximum FDA-allowed output.
RFID in the tool connectors activates parameter and
mode setup, decreasing the setup time as well as workload and
training requirements for the circulator. RFID also activates a
new time-saving, push-prime system for the vitreous cutter
and any connected extrusion tools. A sterile articulated arm
incorporating a tubing management system enables priming
and testing of all infusion and aspiration components before Fig ure 3 .2 ■ One or two dual-port xenon illum inations system s
the patient is anesthetized, prepped, and draped, significantly can supply m ultiple endoillum inated devices with over 25 lum ens
decreasing the setup time and eliminating the need for a Mayo of white light.
34 SECTION II ■ Surgical Technology and Techniques

Fig ure 3 .3 ■ Diagram of the em bedded PurePoint 532-nm laser with a thin-disk laser engine for
im proved control and reliability.

stand. Embedded wizards and video help facilitate faster circulator. The Constellation graphical user interface eliminates
setup even when using less experienced personnel. the need for a separate display and controls.
The Constellation can be configured with an embedded An auto infusion valve replaces the stopcock used for
PurePoint 532-nm laser. The PurePoint laser is a novel, fluid-air exchange, eliminating delay while air travels through
advanced, thin-disk 532-nm solid-state laser (Fig. 3.3). The 84 inches of tubing and bubbles. The auto infusion valve can
thin-disk laser engine reduces thermal lensing, which is the be controlled by the surgeon’s foot pedal or the scrub tech
change in the index of refraction of the Nd:Vanadate lasing (Fig. 3.4) using the Constellation sterile user interface.
medium as it heats up. The thin-disk design produces a more Auto gas syringe fill system enables filling the syringe from
constant laser output, especially with higher powers and firing attached tanks of SF6 and C3F8 reduce gas wastage, ensures
rates because of thermal stability. The lithium triborate frequency sterility, and eliminates the need for the circulator to assist the
upconversion crystal (1,064–532 nm), thermal electric cooler, scrub tech. A software applet calculates how much air to add
and all optomechanical components are solder mounted in a to the gas to produce the desired concentration of gas in air.
fixed, mechanically and thermally stable position. A separate The VFC power silicone injector supports simultane-
foot pedal controls the laser power and standby/ready/standby ous aspiration and has RFID to automatically configure the
control with voice verification, eliminating dependence on the Constellation mode.

Fig ure 3 .4 ■ Auto gas fill calculates the d esired g as concentration and elim inates the need of
a circulator to operate the gas tank regulator.
CHAPTER 3 ■ Constellation Architecture 35

Fig ure 3 .5 ■ Power forceps offer linear control and disposable tip s.

Advanced phaco technology including Ozil torsional supported by single-pedal linear control of power forceps
phaco is embedded in the Constellation to support both to grasp and stabilize epiretinal during the initial segment of
combined phacoemulsification-vitrectomy (phaco-vit) pro- pedal travel followed by control of the power scissors with
cedures and phaco only procedures. further depression of the pedal.
The Constellation has proportional control of new, The RFID system and a barcode reader wand (Fig. 3.7)
higher frequency, 1.5-MHz, sinusoidal diathermy system support the end-of-case transmission to a wireless printer
producing 10 Watts maximum. Higher frequency diathermy for inventory control, cost accounting, and billing. Turnover
produces a more focused lesion possibly reducing retinal time is greatly reduced because of RFID, barcode reader,
damage. automated push-prime system, testing and setup, and the
Power forceps with linear (proportional) control and sterile articulated arm system. Surgical parameters, laser log,
power scissors with single-cut and multicut modes sup- tool, and consumable use data are generated automatically
port disposable Alcon Grieshaber disposable DSP forceps and printed on wireless printer for analysis or incorporation
and scissors tips (Figs. 3.5 and 3.6). Bimanual surgery is into an operative note.

Fig ure 3 .6 ■ Power scissors have single-cut and m ulticut m odes and have d isposab le tips.
36 SECTION II ■ Surgical Technology and Techniques

Fig u re 3 .7 ■ Barcod e read er wand allows the inp ut of d isp osab les used for instrum en t
program m ing, inventory control, and inform ation m anag em ent.
C H AP TER
4
VISUALIZATION AND
ILLUMINATION

Optimal visualization is required for vitreoretinal Wide-angle visualization should be used for all
surgery. Many surgeons utilize wide-angle visu- rhegmatogenous retinal detachment cases, proliferative vit-
alization systems for all cases because of habit, reoretinopathy, and giant breaks when viewing the periph-
video considerations, or the perception that a ery. On occasion, wide-angle visualization is necessary to
second surgeon is required to hold a contact lens. A plano visualize the peripheral portion of the posterior vitreous
(flat) contact lens provides significantly greater lateral and cortex when operating large tabletop diabetic traction reti-
axial (depth) resolution than a noncontact (BIOM, Oculus nal detachments. Wide-angle viewing is ideal to examine
Optikgeräte GmbH, Wetzlar-Dutenhofen, Germany; EIBOS, the periphery for iatrogenic retinal breaks after the comple-
Möller-Wedel, Wedel, Germany) or contact-based (Volks tion of vitrectomy and for removal of peripheral intraocular
Reinverting Operating Lens System, Volk Optical, Inc., foreign bodies or lens fragments, especially at the inferior
Mentor, Ohio; AVI Panoramic Wide-Angle Viewing System, vitreous base. Contact-based wide-angle visualization (Volk,
Advanced Visual Instruments, Inc., New York) wide-angle AVI) provides 10 degrees greater field of view than noncon-
visualization system. A plano contact lens should be used for tact viewing (BIOM) and eliminates all corneal asphericity,
all macular and traction retinal detachment surgeries. Surgi- which is common after cataract surgery, LASIK, photorefrac-
cal technicians usually do an excellent job of holding the tive keratectomy, limbal relaxing incisions, radial keratotomy,
contact lens if they are comfortably seated, view through a astigmatic keratotomy, penetrating keratoplasty, pterygium
stereo observer tube, and are treated with respect. Assistant surgery, and corneal lacerations. Noncontact wide-angle
surgeons frequently mentally focus on the surgical procedure visualization systems require much greater ocular rota-
instead of positioning the contact and require reminders to tion than contact-based systems for viewing the periphery,
center the lens. Sewed-on contact lenses were developed to thereby increasing the 25-gauge tool flex problem. The most
eliminate the need for an assistant to hold the lens but cre- recent generation of articulated and flexible laser probes as
ate many problems. Bleeding and bubbles under the lens, well as stiffer vitreous cutters, scissors, forceps, and endoil-
use of expensive viscoelastics, cost of sutures, suboptimal luminators virtually eliminate the 25-gauge tool flex issue.
centering, and damage to the conjunctiva are all potential
problems with sewed-on lenses. Sewed-on contact lenses
are inappropriate for 23/25-gauge, sutureless, transconjunc- ILLUMINATION
tival surgery because of inherent conjunctival damage and
bleeding. Self-stabilizing contact lenses are useful if a steady- Wide-angle visualization requires some form of wide-angle
handed assistant is not available, but they still require fre- illumination. The principal author prefers the standard
quent recentering by the assistant. Prism lenses can be used 25-gauge, 78-degree endoilluminator to chandelier or Tor-
for viewing the periphery but slow down the surgery and pedo devices. Focal illumination, specular illumination,
can lead to lens or retinal damage because of paradoxical and retroillumination are better for visualizing clear, color-
movement. Indirect ophthalmoscope viewing for vitrectomy less vitreous, internal limiting membrane (ILM), and shiny
is grossly inferior to operating microscope-endoilluminator epiretinal membranes than diffuse illumination produced by
viewing. The author does not agree with the use of micro- chandeliers and Tornambe Torpedoes (Insight Instruments,
scope-mounted slit lamp illumination during vitrectomy Stuart, Florida and Alcon Laboratories, Fort Worth, Texas).
because of light scattering by the cornea and lens as well as Slit lamp biomicroscopy was developed to provide focal illu-
other issues. mination, retroillumination, and specular illumination to

37
38 SECTION II ■ Surgical Technology and Techniques

better visualize transparent structures. Specular illumination because of corneal disease or cataract, the use of optimal
is used when looking for ILM or shiny epiretinal mem- illumination and visualization technology is an essential goal
branes, similar to the way an oblique light source is used in all cases.
to inspect a shiny surface after sanding, painting, cleaning,
or waxing. Retroillumination is produced by reflected light
from the white sclera transmitted through the retina, retinal PHOTOTOXICITY
pigment epithelium, and choroid; it is therefore more use-
ful in lightly pigmented eyes. Another very effective way to Phototoxicity has become a potential issue because of the
produce retroillumination is to reflect endoilluminator light availability of high-brightness xenon and mercury vapor
from the metal surface of the vitreous cutter; this technique sources. Xenon light sources do not burn out suddenly; the
cannot be performed with a chandelier or Tornambe Tor- light output decreases over time, creating potential risk of
pedo. The author uses this technique consciously, but it is excessive light levels if the power is not reduced after lamp
likely that many surgeons use this technique instinctively. replacement. Chandeliers, illuminated infusion cannulas,
Focal illumination is utilized by positioning the endoillumi- illuminated tools, and small-diameter tools produce less
nator near the port of the vitreous cutter, scissors, or forceps; light output for a given light source intensity because of
the surgeon should be careful to use minimal light intensity inherent losses or larger divergence angles. The light source
when near the macula. intensity must be reduced if the surgeon chooses to utilize a
Some surgeons use a chandelier or Torpedo to enable 20-gauge standard endoilluminator with high light through-
bimanual surgery, typically using forceps in one hand and put for macular surgery immediately after doing a case using
scissors or a vitreous cutter in the other, although a pic or low–light throughput tools or operating a dense vitreous
microvitreoretinal (MVR) blade can be used as well. Scis- hemorrhage in a darkly pigmented fundus requiring higher
sors produce a push-out force, which increases with each light source power settings. The best practice is to start every
use if they are reusable scissors and further increases with case, particularly macular surgery, with low light intensity
the reuse of disposable scissors. Bimanual surgery is often and increase the intensity gradually until sufficient illumina-
used to offset this push-out force using forceps in one hand tion is reached. The latest generation of 25-gauge tools have
for epiretinal membrane stabilization and scissors or the vit- high light throughput; the author typically sets his xenon
reous cutter in the other. The author uses disposable curved source at approximately15%.
scissors for both segmentation and delamination and rarely ICG and other stains are chromophores and, there-
performs bimanual surgery. fore, dramatically increase the risk of phototoxicity, espe-
The use of indocyanine green and other staining agents cially when using high-intensity xenon and mercury vapor
for the ILM as well as triamcinolone particulate marking of sources. Phototoxicity is additive to the well-known chemi-
the vitreous is driven, in part, by the use of wide-angle dif- cal toxicity of the dye molecule in addition to pH, osmolar-
fuse illumination sources that reduce a surgeon’s ability to ity, and diluent toxicity issues.
view transparent, colorless structures such as clear vitreous, Video is another factor in phototoxicity; because CCD
the ILM, and thin, transparent epiretinal membranes. The cameras are not as sensitive as the surgeon’s eye, some sur-
dynamic range of charge-coupled-device (CCD) cameras geons use 30/70 beam splitters to produce more light for the
and displays for surgical video is 2 to 3 log units (f-stops), video channel, which produces the unintended consequence
while the surgeon’s eye is 7 log units (f-stops), which, in of requiring higher illumination levels to optimize the sur-
part, drive the use of wide-angle illumination systems to geon’s view. Single-chip CCD cameras are more than three
provide uniform illumination for video. The objective is safe, times more sensitive than three-chip cameras but produce
effective, rapid surgery; making a video is far less important. inferior color quality. Most surgeons use three-chip cameras,
A large-screen display in the operating room does, however, which require higher illumination levels. The optimal micro-
enhance technician and nurse interest, involvement, effi- scope objective has a focal length of 175 mm. Although
ciency, and education. Large flat-screen displays are ideal for 150-mm objectives produce 1 log unit (f-stop) more light,
teaching visiting surgeons, residents, and fellows. contamination of tools, tubing, wires, and fibers due to lim-
Vitreous without hemorrhage, ILM, epiretinal mem- ited clearance between the microscope and the eye is likely.
branes, and the retina are all colorless; the argument that A 200-mm objective requires 1 f-stop more light than a
green or yellow light improves the ability to see these struc- 175-mm objective and should not be used.
tures is without merit. Green light improves the contrast of
red structures (blood and blood vessels), but this is never
a problem. Yellow light makes visualization of the macular ENDOSCOPE SYSTEMS
xanthophyll nearly impossible, which is crucial in many
cases of macular surgery, diabetic traction retinal detach- Endoscope systems are rarely, if ever, needed in vitreous sur-
ment, and difficult trauma cases with displaced maculas. gery. They can theoretically be of value to dissect, drain sub-
Although there are clinical situations in which vitreo- retinal fluid (SRF), and laser peripheral retinal detachments
retinal surgery must be performed with a suboptimal view in phakic eyes with small pupils. If capsular fibrosis limits
CHAPTER 4 ■ Visualization and Illum ination 39

the view, it can be dissected with the scissors to enlarge the Kim SR, Nakanishi K, Itagaki Y, Sparrow JR. Photooxidation of
opening, eliminating the need for an endoscope. The authors A2-PE, a photoreceptor outer segment fluorophore, and protec-
tion by lutein and zeaxanthin. Exp Eye Res 2006;82:828–839.
rarely use iris retractors because of iris damage, bleeding,
Koch FH, Schmidt HP, Mönks T, et al. The retinal irradiance and
and postoperative inflammation. If the pupil is small, the spectral properties of the multiport illumination system for vitre-
periphery can be seen by using wide-angle viewing, care- ous surgery. Am J Ophthalmol 1993;116:489–496.
ful positioning of the head and eye, scleral depression, and Kraushar MF, Harris MJ, Morse PH. Monochromatic endoillu-
sphincterotomies (in aphakic eyes). Endophotocoagulation mination for epimacular membrane surgery. Ophthalmic Surg
1989;20:508–510.
of the ciliary body can be performed using scleral depression
Kuhn F, Morris R, Massey M. Photic retinal injury from endoillumi-
or with endoscopic viewing. GRIN rod endoscopes have 15× nation during vitrectomy. Am J Ophthalmol 1991;111:42–46.
more pixels than fused coherent fiber bundle–based systems Lawwill T. Three major pathologic processes caused by light in the
but have limited optical resolution because of low modu- primate retina: a search for mechanisms. Trans Am Ophthalmol
lar transfer function (MTF) microoptics. Combining white Soc 1982;80:517–579.
Maia M, Haller JA, Pieramici DJ, et al. Retinal pigment epithelial
light illumination, laser endophotocoagulation, and a fluid
abnormalities after internal limiting membrane peeling guided
channel can increase the utility of an endoscope. The fluid by indocyanine green staining. Retina 2004;24:157–160.
channel plugs easily if used for aspiration and is seldom, if McDonald HR, Irvine AR. Light-induced maculopathy from the
ever, needed for infusion. The authors find little practical operating microscope in the extracapsular cataract extraction and
value for endoscopic vitreous surgery. intraocular lens implantation. Ophthalmology 1983;90:945–951.
Meyers SM, Bonner RF. Retinal irradiance from vitrectomy endoil-
luminators. Am J Ophthalmol 1982;94:26–29.
Bibliogra phy Michels M, Lewis H, Abrams GW, et al. Macular phototoxicity
caused by fiberoptic endoillumination during pars plana vitrec-
Andonegui Navarro J, Marcuerquiaga Arriaga J. Xenon light tomy. Am J Ophthalmol 1992;114:287–296.
induced phototoxic lesions [Article in Spanish]. Arch Soc Esp Miller SA, Landry RJ, Byrnes GA. Endoilluminators: evaluation of
Oftalmol 2000;75:117–120. potential retinal hazards. Appl Opt 2004;43:1648–1653.
Azzolini C, Brancato R, Venturi G, et al. Updating on intraoperative Noell WK, Walker VS, Kang BS, Berman S. Retinal damage by light
light-induced retinal injury. Int Ophthalmol 1994–1995;18:269– in rats. Invest Ophthalmol Vis Sci 1966;5:450–473.
276. Poliner LS, Tornambe PE. Retinal pigment epitheliopathy after
Birngruber R, Gabel VP. Thermal versus photochemical damage in macular hole surgery. Ophthalmology 1992;99:1671–1677.
the retina—thermal calculations for exposure limits. Trans Oph- [Comment in Ophthalmology 1993;100:1604–1605.]
thalmol Soc UK 1983;103:422–427. Remé CE. The dark side of light: rhodopsin and the silent
Charles S. Illumination and phototoxicity issues in vitreoretinal death of vision. The Proctor lecture. Invest Ophthalmol Vis Sci
surgery. Retina 2008;28:1–4. 2005;46:2671–2682.
Charles S. Retinal pigment epithelial abnormalities after macular Sparrow JR, Zhou J, Ben-Shabat S, et al. Involvement of oxidative
hole surgery [Letter to the Editor]. Retina 1993;13:176. mechanisms in blue-light-induced damage to A2E-laden RPE.
Donovan M, Carmody RJ, Cotter TG. Light-induced photoreceptor Invest Ophthalmol Vis Sci 2002;43:1222–1227.
apoptosis in vivo requires neuronal nitric-oxide synthase and gua- van den Biesen PR, Berenschot T, Verdaasdonk RM, et al. Endoil-
nylate cyclase activity and is caspase-3-independent. J Biol Chem lumination during vitrectomy and phototoxicity thresholds. Br J
2001;276:23000–23008. [Epub ahead of print 2001 Mar 8.] Ophthalmol 2000;84:1372–1375.
Fisher PL, Suh DW, Rapp LM. Evaluation of retinal susceptibility to Williams TP, Howell WL. Action spectrum of retinal light-damage
light damage in pigmented rats supplemented with beta-carotene. in albino rats. Invest Ophthalmol Vis Sci 1983;24:285–287.
Curr Eye Res 1996;15:219–223. Wu WC, Hu DN, Roberts JE. Phototoxicity of indocyanine green
Fuller D, Machemer R, Knighton RW. Retinal damage produced by on human retinal pigment epithelium in vitro and its reduction
intraocular fiber optic light. Am J Ophthalmol 1978;85:519–537. by lutein. Photochem Photobiol [Epub ahead of print September
Ham WT Jr, Mueller HA, Ruffolo JJ Jr, et al. Action spectrum for 2004.]
retinal injury from near-ultraviolet radiation in the aphakic mon- Yanagi Y, Inoue Y, Jang WD, Kadonosono K. A2e mediated pho-
key. Am J Ophthalmol 1982;93:299–306. totoxic effects of endoilluminators. Br J Ophthalmol 2006;90:
Harwerth RS, Sperling HG. Prolonged color blindness induced 229–232.
by intense spectral lights in rhesus monkeys. Science 1971;174: Yanagi Y, Iriyama A, Jang WD, Kadonosono K. Evaluation of the
520–523. safety of xenon/bandpass light in vitrectomy using the A2E-laden
Keller C, Grimm C, Wenzel A, et al. Protective effect of halot- RPE model. Graefes Arch Clin Exp Ophthalmol 2007;245:677–
hane anesthesia on retinal light damage: inhibition of meta- 681. [Epub ahead of print 2006 Oct 17.]
bolic rhodopsin regeneration. Invest Ophthalmol Vis Sci 2001;42: Zilis JD, Machemer R. Light damage in detached retina. Am J Oph-
476–480. thalmol 1991;111:47–50.
C H AP TER
5
ANESTHESIA AND OPERATING
ROOM EFFICIENCY

for general anesthesia is the patient who insists upon it,


PREOPERATIVE EVALUATION
although these patients will be rare if properly informed
and reassured by a sympathetic surgical team. Nitrous oxide
Regardless of the type of anesthesia contemplated for
should be avoided not only because of the potential inter-
vitreoretinal (VR) surgery, the patient should undergo a thor-
action with intraocular gas bubbles but also because it has
ough preoperative evaluation prior to the procedure. Under
been shown in large, randomized multicenter trials to result
most circumstances, this evaluation should occur well before
in a 30% higher incidence of nausea and vomiting.
the day of surgery so that required treatment can be per-
formed in advance to help ensure that the patient is in opti-
mal condition prior to surgery. Specific investigations, such
as chest X-ray, ECG, and blood chemistries, should be per- MONITORING DURING SURGERY
formed only when dictated by the findings of thorough his-
tory and physical examinations. So-called “screening labs” Regardless of the type of anesthesia used, the patient must
are not indicated when the appropriate history and physical be carefully monitored during surgery. Appropriate monitor-
examinations are negative. ing begins with the continuous presence of an anesthesiolo-
gist or certified registered nurse anesthetist during the entire
procedure. If sedation is being given, it is not in the patient’s
GENERAL VERSUS LOCAL ANESTHESIA best interest to have the surgeon or circulating nurse moni-
toring the patient, as may be the case in a brief procedure
Both general and local anesthetic techniques are accept- performed under strictly local anesthesia without sedation.
able for VR surgery; however, the authors prefer to do the Basic monitoring includes continuous ECG, noninvasive
vast majority of their cases using monitored local anesthesia blood pressure, and pulse oximetry. End-tidal CO2 monitor-
for a variety of reasons: (a) local anesthesia offers increased ing is additionally essential during general anesthesia and
safety for patients, especially those in high-risk categories, can also be helpful during local anesthesia. Core tempera-
(b) local anesthesia saves time and reduces cost, and (c) local ture monitoring is indicated during longer procedures under
anesthesia provides rapid recovery and prolonged analgesia, general anesthesia to help ensure that thermal preservation
both of which are especially important in the outpatient procedures are successful and to help in monitoring for
population. the rare occurrence of malignant hyperthermia. In diabetic
Not all patients are appropriate candidates for VR patients, the ability to monitor blood glucose in the intraop-
surgery under local anesthesia. Immature, mentally defi- erative and perioperative periods is also important in order
cient, claustrophobic, and uncooperative patients are best to recognize and treat extremes of both hyperglycemia and
managed with general anesthesia. Patients with language hypoglycemia.
barriers, however, can frequently be managed extremely well
with local anesthesia if a competent translator can be found.
Estimated surgical time is an additional consideration when BLOOD PRESSURE CONSIDERATIONS
choosing general versus local anesthesia. Surgeons requiring DURING GENERAL ANESTHESIA
more than 90 minutes for a given VR procedure should con-
sider general anesthesia over local anesthesia, as patients may It is common for VR surgeons to become angry if the patient
become restless and uncomfortable when asked to lie com- moves at all during surgery. An unintended consequence
pletely still for such long periods. An additional indication of this tendency is for the anesthesia provider to maintain
40
CHAPTER 5 ■ Anesthesia and Operating Room Efficiency 41

deeper levels of anesthesia to prevent movements, which this drug only remains sterile in a syringe for a maximum
may result in low enough systemic blood pressures to com- of 12 hours, which may be a consideration in developing
promise cerebral, myocardial, and retinal perfusions. During countries. For VR surgery, the emphasis must be placed
VR surgery, intraocular pressure (IOP) should be controlled on balancing patient comfort and satisfaction while provid-
in the 35 to 45 mm Hg range. Ocular ischemia and central ing the most stable conditions for surgery. In general, this
retinal artery occlusion can occur if low systemic blood pres- means using small doses of rapid-onset, short-acting drugs
sures are allowed to persist during the procedure. To ensure given continuously with very careful monitoring of effect.
adequate levels of general anesthesia and immobility of the The goals are to assist the patient in lying perfectly still for
patient, adequate, monitored muscular relaxation combined 60 to 90 minutes without falling asleep, to enhance analge-
with processed EEG (i.e., bispectral analysis) monitoring sia, and to provide a measure of amnesia. These are not eas-
should be considered so that excessively deep levels of gen- ily achieved, but they can be accomplished in most patients
eral anesthesia can be avoided. by an experienced and knowledgeable anesthesia team.

SEDATION DURING LOCAL PSYCHOLOGICAL PREPARATION


ANESTHESIA FOR LOCAL ANESTHESIA

In general, patients having VR surgery under local In preparing patients for VR surgery under some form of
anesthesia should have minimal sedation, most of which local anesthesia, it is important to give them specific details
should be given at the time of the block. Patients should about the experience so that they will suffer no surprises.
not be sedated too deeply during VR surgery for a num- They need to know about the drape and about not being
ber of reasons. In the first place, airway obstruction may able to see during the procedure. They also need to know
occur, requiring manual support and interruption of the that plenty of fresh air will be provided for them under the
procedure. This has been described as AWAC (anesthesia drape and that breathing under the drape will not be a prob-
without airway control). Secondly, respiratory movements lem. This is the perfect opportunity to discuss the patient’s
during sleep or near sleep often result in magnified move- fears, such as claustrophobia, positional dyspnea, positional
ments of the head, which greatly hinder the progress of the pain, and similar concerns. One may discover during these
surgeon who is seeing these movements magnified 20 to discussions that a particular patient might be better man-
40 times through the operating microscope. Thirdly, some aged with general anesthesia.
patients become quite talkative and social when overly The patient should also be given a realistic estimate of
sedated. It may be impossible for them to quit talking and the length of the procedure and the need for lying extremely
moving despite the most vigorous admonitions to do so. still. Almost anyone can lie still for 30 to 45 minutes, but for
The only way to manage these patients is to stop all seda- longer procedures the patient must be reassured that short
tion completely or to convert to general anesthesia. Finally, “time-outs” can be arranged to allow for some movement.
patients who are asleep or nearly asleep are prone to awak- Patients must also be aware that an anesthesia pro-
ening suddenly and being totally disoriented, resulting in vider will be constantly present and dedicated to monitor-
movements, which can be devastating, even in the hands of ing their condition and to act as liaison with the rest of the
the finest surgeon. The goal of sedation should be control team. It is extremely important for the anesthesia provider
of anxiety, rather than true sedation with decreased aware- and surgeon to communicate freely during the procedure,
ness of surroundings. both with each other and with the patient. Simple means
Judicious amounts of sedatives and/or opioid agents for communication with minimal movement, such as hand-
can be helpful during local anesthesia for VR surgery, espe- holding or handheld signaling devices, give the patient a
cially in the patient who is very apprehensive or slightly feeling of comfort in knowing that it is possible to alert the
claustrophobic. Brevital, thiopental, midazolam, propofol, team to a problem while not jeopardizing the surgical field.
alfentanil, remifentanil, ketamine, and others have been If the patient cannot speak English, it is imperative to have
promoted to provide good operating conditions and accept- a translator in the room who is fluent in the patient’s native
able patient sedation for a variety of procedures performed language.
under local anesthesia. Brevital, thiopental, and alfentanil
have been largely supplanted by propofol. Remifentanil
has a higher profile of nausea and vomiting than the other CHOICE OF LOCAL ANESTHESIA
narcotics. Ketamine will cause nystagmus and probably
increases IOP. Although the regional block should pre- There are essentially four types of local anesthesia commonly
vent ocular movements, ketamine is a questionable choice used in ophthalmic surgery: topical, retrobulbar, peribulbar,
unless a mentally challenged or combative patient had to and sub-Tenon’s. Topical anesthesia is useful in a variety of
be done under regional anesthesia. The cost of propofol has operations, but it has limitations in VR surgery due to the
significantly decreased since the drug became generic, but need for complete akinesia during many VR procedures,
42 SECTION II ■ Surgical Technology and Techniques

such as macular surgery and membrane peeling. The terms


REBLOCKING DURING THE
“retrobulbar” and “peribulbar” are confusing and imprecise,
and they should perhaps be replaced by the terms “intra-
PROCEDURE
conal” and “extraconal,” which more accurately describe
Sometimes local anesthesia must be supplemented during
the intended location of the needle in the orbit. These tech-
surgery. This can occasionally be accomplished with topi-
niques carry a risk, albeit small, of major complications,
cal anesthesia, but the authors most commonly supplement
such as ocular perforation, bleeding, and brainstem anes-
intraoperatively by placing a flexible cannula into Tenon’s
thesia, but both are very useful for VR surgery, providing
space and injecting additional local anesthetic. An additional
excellent akinesia, anesthesia, and prolonged postoperative
intraconal injection can also be performed by placing the
analgesia. Sub-Tenon’s anesthesia offers an increased level
needle between Tenon’s capsule and the sclera to enter the
of safety over intraconal and extraconal techniques. Sub-
intraconal space. Most often, reblocking is necessary when
Tenon’s anesthesia may not be appropriate for patients who
the block has been inadequate, when the patient is undergo-
have had previous scleral buckling, as scleral perforation
ing a reoperation, and when the procedure is prolonged.
with a sub-Tenon’s cannula has been reported in such a
patient.
The two main local anesthestics used for retrobulbar
anesthesia are lidocaine and bupivacaine. Although bupiva- FACIAL NERVE BLOCKS
caine (Marcaine) has a longer duration of action, it has sig-
nificant cardiotoxicity (1). For the usual macular surgeries Separate facial nerve blocks are rarely indicated, especially
and retinal detachment surgeries, the duration of action of if a well-performed extraconal or high-volume intraconal
lidocaine is sufficient for painless surgery. On the other hand, block is used. Avoiding a facial nerve block spares the patient
a longer and more complicated case, such as combined pha- a painful injection and prevents the bleeding, swelling,
covitrectomy for a diabetic with dense cataract and complex and other complications that occasionally accompany these
traction retinal detachment, may take some surgeons longer blocks. If the patient is a marked “squeezer,” the orbicularis
than 90 minutes, including the time for setting up and prim- occuli can be easily and effectively blocked by inserting a
ing the machine. These cases are best blocked with bupiva- 0.5-inch 30-gauge needle transconjunctivally into the lower
caine or done under general anesthesia. lid just beneath the orbicularis and injecting about 1.5 mL
of local anesthetic.

TECHNIQUE FOR INTRACONAL


ANESTHESIA SOURCES OF PAIN DURING
VITREORETINAL SURGERY
A 27-gauge needle is preferred to larger needles and blunt
so-called “retrobulbar” needles that cause much more pain Local anesthesia needs to be quite complete if the experience
when going through the collagenous lid septum. In addition, is to be pain free. Manipulation of the iris, ciliary body, and
retrobulbar needles often penetrate the septum abruptly sclera can all be painful, especially if blunt instruments are
after considerable force is applied and may then perforate being used. Thermal stimulation is also an important source
the eye. The conventional 1.5-inch needle is too long for of discomfort. Cryopexy is very painful, more so than laser
many orbits and should be replaced by a 1.25-inch needle or even radio-frequency cautery (bipolar diathermy). Lasers
to avoid impaling the optic nerve in the orbital apex. The in the near-infrared range are more painful than the argon
entry point should be at the outer “corner” of the orbital rim, laser at 514 nm or the diode-pumped, frequency-doubled
not at the outer 1/3, inner 2/3 junction in order to reduce CW YAG laser at 532 nm. Some patients also experience
potential damage to the eye and inferior oblique muscle. The pain or discomfort during extreme globe rotations dur-
needle should be directed on a straight, not curved, path ing vitrectomy. As one or more of these modalities may be
along a plane intersecting the visual axis. The authors use employed during VR surgery, it is important that the patient
2% plain lidocaine without epinephrine to reduce the risk receives adequate anesthesia.
of arrhythmias and hypertension and avoid using bicarbon-
ate because of reports and personal experience with lateral
rectus paralysis for months after surgery. The authors recom- CARBON DIOXIDE ISSUES
mend applying pressure on the entire orbit with the palm of
the hand immediately after withdrawing the needle to reduce Patients lying awake under the drape frequently complain
bleeding and disperse the anesthetic agent. The authors do that they “cannot get enough air.” As pulse oximetry rou-
not use Wydase because approximately half of the random- tinely records normal oxygen saturation in these patients,
ized trials show no effect while the others show minimally their complaints are frequently attributed to anxiety. In fact,
faster onset and the agent is likely to cause severe damage if CO2 often builds up under the drape, resulting in hypercar-
injected into the eye. bia and a feeling of air hunger. This may be noted by a rise in
CHAPTER 5 ■ Anesthesia and Operating Room Efficiency 43

the baseline if capnography is being used, even though the Scle ral Buckle s
peak expired CO2 may be normal or only slightly elevated.
Many presenting for scleral buckling procedures will be
An easy solution to this problem is to ensure adequate air or
high myopes. These patients have long axial lengths, often
oxygen supplementation near the patient’s nose and mouth
accompanied by posterior staphylomas and scleral thinning.
as well as active evacuation of the exhaled gases by way
Sub-Tenon’s cannula techniques might be considered in
of a large-bore vacuum line placed under the drapes. The
these patients to lessen the risk of perforation.
vacuum line also facilitates cooling, which can be an issue
Regional anesthesia for scleral buckling procedures may
as well.
be complicated by the fact that the orbital retractor can cause
significant orbital rim pain even in the presence of complete
ocular anesthesia. Additionally, with traction of the extraoc-
AIR/ GAS AND GENERAL ANESTHESIA ular muscles, the oculocardiac reflex may occur. Most com-
monly, the resulting bradycardia will return to normal when
If gas and/or air are introduced into the eye during VR
traction is released, and the reflex will diminish over time.
surgery, nitrous oxide should be turned off at least 10
Intravenous atropine is more effective than glycopyrrolate in
minutes beforehand and fresh gas flow into the anesthesia
blocking the reflex, but its use is associated with the higher
machine should be increased to ensure adequate wash-
incidence of subsequent tachyarrhythmias. Local anesthetic
ing out prior to introduction of the gas. Failure to do so
injection may block the bradycardia, but the reflex is also
results in a smaller-than-desired gas bubble within the eye
seen in the presence of a complete block.
and lower-than-desired IOP postoperatively when nitrous
Patients who have had previous scleral buckles and
oxide diffuses out of the bubble. Conversely, if a patient has
present for another procedure may be difficult to block.
a bubble in the eye from a previous procedure, nitrous oxide
As the buckling may slightly elongate the eye, one must be
should be avoided from the beginning in order to prevent
aware of an increased danger for perforation. As scarring
the expansion of the bubble by diffusion of nitrous oxide
occurs, normally “safe” procedures may become less safe,
into it, thus raising IOP. In fact, patients must be warned to
and ocular perforation has been reported with sub-Tenon’s
alert physicians to the presence of the bubble should they
anesthesia in a patient with a previous scleral buckle.
require emergency surgery for a nonophthalmic condition.

An t ico ag ulat io n Issue s


ANESTHETIC CONSIDERATIONS FOR
In their practice, the authors virtually never stop anticoagu-
SPECIFIC PROCEDURES lation or antiplatelet therapy prior to VR surgery, although
it is wise to ensure that the patient taking warfarin com-
En d o p h t h alm it is pounds has an international normalized ratio (INR) in the
Endophthalmitis is an acute situation in which cultures must therapeutic range (generally 2–3). Stopping anticoagulants
be taken and therapy instituted as quickly as possible. In risks causing morbidity or mortality from a variety of causes,
many situations, cultures and even core vitrectomy can be including stroke, myocardial infarction, pulmonary embo-
performed under topical anesthesia. If general anesthesia is lism, and deep venous thrombosis. In the authors’ opinion,
required, surgery cannot be delayed to allow the stomach to the dangers of intraoperative hemorrhage are grossly over-
empty. emphasized when compared to the dangers of stopping
therapeutic anticoagulation. Properly performed VR surgery
is safe on patients with therapeutic INRs (2,3). Use of can-
Th e Op e n Glo b e nula techniques for local anesthesia greatly reduces the risk
Each patient must be thoroughly evaluated, as the choice of hemorrhage in these patients, as does the use of short
of anesthesia will depend on the extent of the injury and (1.25 inches) 27-gauge needles placed in the less vascu-
the ability of the patient to cooperate. Often, initial wound lar areas of the orbit (i.e., avoiding the superior half of the
closure can be accomplished under topical and intracam- orbit in general and especially the superonasal quadrant) for
eral anesthesia. In cooperative patients with limited damage, orbital blocks.
orbital regional anesthesia can be safely used, provided that
the person performing the block has had sufficient experi-
Po st o p e rat ive Pain
ence, uses limited volumes of anesthetic, and injects very
slowly (i.e., 1 mL every 30–60 seconds) while closely watch- One source of postoperative pain is the injection of antibi-
ing the eye. When general anesthesia is required, the issue of otics and steroids into the periocular tissues at the end of
whether or not to use a depolarizing muscle relaxant arises. the procedure. This pain can be reduced by injecting these
As there are advocates on both sides of this issue, the choice substances into the sub-Tenon’s space with a cannula if con-
must be left to the anesthesia provider, who will make a junctival incisions have been made, which is not the case
decision based on the total clinical picture. with 25-gauge, sutureless surgery. In addition, injection of a
44 SECTION II ■ Surgical Technology and Techniques

long-acting local anesthetic, such as bupivacaine, at the end choice of anesthesia technique must be based on the needs
of the procedure with a flexible cannula can greatly reduce of the patient, the requirements of the surgeon, and the
postoperative pain. This is especially important in the occa- skills of the anesthesia provider, ever keeping in mind that
sional patient who requires general anesthesia for VR surgery our ultimate goal is a satisfied patient with a good visual
and those undergoing scleral buckles. outcome.

SUMMARY References
1. Marwick PC, Levin AI, Coetzee AR. Recurrence of cardiotoxic-
The vast majority of VR procedures can be safely, comfort- ity after lipid rescue from bupivacaine-induced cardiac arrest.
ably, and efficiently performed under local anesthesia with Anesth Analg 2009;108:1344–1346.
minimal sedation. Compared with general anesthesia, prop- 2. Dayani PN, Grand MG. Maintenance of warfarin anticoagulation
for patients undergoing vitreoretinal surgery. Arch Ophthalmol
erly performed and monitored local anesthesia offers the 2006;124:1558–1565.
patient an increased level of safety, reduced recovery times, 3. Fu AD, McDonald HR, Williams DF, et al. Anticoagulation with
and prolonged postoperative pain relief. Nonetheless, the Warfarin in vitreoretinal surgery. Retina 2007;27:290–295.
C H AP TER
6
GENERAL POSTERIOR SEGMENT
TECHNIQUES

trabecular meshwork and lens damage compared to other


STRATEGIES
solutions. The authors do not add glucose, epinephrine, or
antibiotics to the fluid to reduce the chances of using incor-
All surgery requires a strategy, game plan, or algorithm before
rect agents or concentrations as well as to reduce the risk
being undertaken. The complexity of high-technology vit-
of corneal, lens, and retinal toxicities. Plastic infusion fluid
reoretinal surgery requires extensive preoperative planning.
containers should be avoided because air diffuses through
The surgical algorithm must include an expected series of
the plastic during storage, causing bubbles. Two-component
events but must contain branches to allow for the contin-
infusate systems should be designed so that the substance in
gencies and discoveries that occur in the course of surgery.
the actual infusion container is safe if the second component
The main branches of the algorithm are determined by the
is inadvertently omitted, as is the case with BSS Plus.
expected techniques, such as lens removal, endophotoco-
agulation, silicone oil, gas, scleral buckling, etc. Equipment,
disposables, gas, oil, buckles, etc. must be available for any
possible additional techniques driven by the individual case IMPORTANCE OF VISUALIZATION
and any intraoperative findings or complications.
Optimal visualization is essential to safe vitrectomy. While it
is obviously desirable to preserve the corneal epithelium and
SPECULUM the lens, neither one is as important as the ability to perform
a safe vitrectomy. This simple principle is often overlooked
A heavy wire speculum is an effective means of retracting with adverse consequences.
the eyelids. Larger bladed specula that lift the lids may inter-
fere with the contact lens and infusion cannula. The drape
Pup illary Dilat io n
should not have a precut hole; it should be placed with the
lids open and cut so the flaps completely cover both lid mar- The preoperative administration of topical adrenergic
gins using the speculum to hold them in place. Covering (phenylephedrine 2.5%–10%) and anticholinergic (tropic-
the lashes and lid margins is done to reduce the chance of amide 1%, cyclopentolate 1%, hyoscine 0.25%, or homa-
endophthalmitis and to prevent cilia from being entrapped tropine 2.5%) dilating drops is critical to safe vitrectomy.
in tissue. Aspirating speculums reduce fluid runoff under Minimization of hypotony and mechanical iris trauma usu-
the drape or onto the foot pedals, surgeon’s feet, and floor ally allow the preoperative dilation to be sustained through-
but tend to be bulkier. out the case. If pupillary constriction occurs, a small pulse
of intraocular epinephrine can redilate the pupil. This epi-
nephrine should be labeled “for injection,” contain minimal
INFUSION FLUID amounts of sodium bisulfite antioxidant, and be diluted to
1:10,000 (1). Intracardiac injection preparations, although
High-quality infusion fluid containing dextrose, glutathione, labeled as containing no preservative, contain too much
and a bicarbonate buffer should be used for all cases. This sodium bisulfite and are potentially toxic to the corneal
should be freshly prepared just prior to the operation. High- endothelium. Surgical pupillary sphincterotomies should be
quality commercial solutions (Balanced Salt Solution, BSS avoided unless the above steps fail and the fundus cannot be
Plus, Alcon Laboratories) should be utilized in all cases. Use seen adequately for safe surgery. Iris surgery leads to postop-
of BSS Plus reduces corneal endothelial cell loss as well as erative glare, photophobia, and cosmetic problems, as well
45
46 SECTION II ■ Surgical Technology and Techniques

as increased postoperative inflammation. Iris retractors and best scleral tunnel and is less likely to result in an inadvertent
sutures cause iris trauma, inflammation, longer operating suprachoroidal infusion than a flatter (5 degree) itrajectory.
time, and increased cost and should only be used if there
is no other option for surgical visualization. Contact wide-
angle visualization systems with a trained assistant obviate Sta biliza tion of the Eye
the need for iris retractors, iris surgery, and intracameral epi- Traditionally, surgeons have attempted to stabilize the eye to
nephrine in almost all cases. offset translational and rotational forces acting on the eye
during trocar-cannula insertion. An alternative approach
Le n s Re m o val is to allow the eye to move deeper into the orbit but pre-
vent eye rotation by rigidly enforcing the correct geometric
If lens removal is necessary, phaco or lensectomy should be relationship between the trocar and the eye (4). Using the
performed before proceeding with vitrectomy, unless the forefinger on the surgeon’s hand to triangulate the trocar
lens is subluxated or dislocated. Relative miosis may occur support on the insertion hand facilitates better control of
as the vitrectomy progresses, and early lens removal permits hand and trocar attitude. Some surgeons have described this
better access to the periphery and prevents unnecessary technique as pressing down on the eye; while this is true,
iris surgery. The view is always better in aphakic or pseu- the key point is to maintain the correct insertion angle and
dophakic eyes, and safe, effective vitrectomy should never direction by continuously modifying the handle angle while
be compromised to preserve the lens, even if it is relatively allowing the eye to freely move. Initially, this hand holds the
clear. Saving the anterior lens capsule until the end of the cotton-tip applicator used to displace the conjunctiva, but
operation in lensectomy cases hinders the view and offers no the cotton-tip applicator is dropped after the trocar impales
significant advantages. Forceps removal of the lens capsule the conjunctiva against the sclera, maintaining the displaced
in lensectomy cases facilitates total capsule removal without position. Incidentally, the authors use the same two-handed
iris touch and resultant miosis and inflammation. approach when performing intravitreal injections and ret-
robulbar blocks.
Wound Construction Issues for Microincisiona l The surgeon should allow a large service loop before
Vitrectomy Surgery adhesively fastening the infusion line to the drape to avoid
traction and inadvertent suprachoroidal infusion or can-
A transconjunctival, sutureless vitrectomy technique was nula pullout during ocular rotation. Unless a filtering bleb
introduced by Eugene de Juan, to reduce pain, inflamma- or other conjunctival conditions prevent this approach, the
tion, and operating time, although from the outset, concern sclerotomies should be placed to facilitate the greatest pos-
regarding hypotony and endophthalmitis was expressed sible angular intraocular access.
with this technique, as with sutureless, small-incision cata- The infusion cannula port is traditionally placed infer-
ract surgery (2). de Juan recommends leaving vitreous in otemporally near the lower border of the lateral rectus, but
the wound to reduce the incidence of wound leaks, but the it is better to move it closer to the 3 o’clock or 9 o’clock
authors disagree with this approach because leaving vitreous position to avoid contact with the lower lid. The supero-
in the sclerotomies increases the likelihood of vitreous wick nasal sclerotomy should be placed on a virtual line from
(and therefore increases the likelihood of endophthalmitis) the lowest point of the bridge of the nose to the center
and increases the likelihood of subsequent hypocellular con- of the pupil, facilitating better angular movement. Similarly,
traction of the collagen fibers and retinal breaks immediately the superotemporal incision should be placed on a virtual
posterior to the sclerotomies (3). In the authors’ opinion, vit- line extending from the lowest point of the supraorbital rim
reous wicks are more likely to be a factor in endophthalmitis to the center of the pupil. The position of the 25-gauge can-
than hypotony with retrograde flow of tear film into the eye. nulas may require modification in the presence of a filtering
bleb, an area of previous trauma with a sclera wound, or if
Angula ted Wound Construction there is underlying choroidal effusion or hemorrhage.
The authors initially made the sclerotomies 3.5 mm
Although the authors initially used straight-in wound con- posterior to the limbus but have found that moving them to
struction for 25-gauge vitrectomy, they changed to angulated 4.0 mm posterior to the limbus reduces the chance of air in
wound construction after the technique was introduced for the anterior chamber of phakic or pseudophakic eyes during
23-gauge vitrectomy. The purpose of angulated wound con- fluid-air exchange (FAX).
struction is to create a scleral tunnel that functions as a flap
valve, similar to no-stitch cataract surgery incision. It is not,
however, considered a biplanar wound; the change in trocar
Conjunctiva l Displa cement
insertion angle is to avoid intraocular contact. The wound
already has been made at the point of trocar insertion. Some The concept of conjunctival displacement for sutureless,
surgeons call the uptilt in insertion angle supination, which 25-gauge surgery was developed by de Juan; some sur-
is incorrect. A single plane, 15 degree incision produces the geons using 23-gauge systems have abandoned conjunctival
CHAPTER 6 ■ General Posterior Segm ent Techniques 47

displacement (in the authors’ opinion, unwisely) because should avoid filtering blebs, previous conjunctival wounds,
various techniques to stabilize the globe made displacement areas of pars plana scarring, high choroidals, or suprachoroidal
difficult. Tornambe Torpedoes (Insight Instruments, Stuart, hemorrhage to ensure that the cannula extends into the vitre-
Florida) and other chandelier systems are inserted without ous cavity and not in the suprachoroidal space. If required,
conjunctival displacement, which removes vitreous at the site, contiguous placement of all cannulas on the same hemisphere
or without angulated wound construction; both a lack of con- or quadrant can be performed. Additionally, the infusion port
junctival displacement or failure to perform angulated wound with some vitrectomy systems (Accurus and Constellation
construction increases the likelihood of endophthalmitis (5). Vision System, Alcon, Fort Worth, Texas) can be moved from
one cannula to another to facilitate tool access to pathology.
Several early studies demonstrate an apparent increase
Reduction of Wound Lea ks
in endophthalmitis rates after 23-gauge and 25-gauge surger-
The senior author first reported the use of FAX to reduce ies. The Wills study has been widely quoted, but the surgeons
wound leaks after 25-gauge straight-in wound construc- involved in the study now believe that the factors resulting
tion, but with the adoption of scleral tunnel wound con- in the apparent increase in endophthalmitis are no longer
struction, the author currently utilizes the FAX technique to significant (9,10). Although hypotony is considered a factor
reduce wound leaks only in patients with a high degree of for endophthalmitis, the author believes that vitreous wicks
myopia or Marfan syndrome or in other patients with thin and omission of subconjunctival antibiotics are more impor-
sclera. tant issues and are bigger risk factors for the development
Logically, wounds made during 20-gauge (0.89-mm of endophthalmitis. Some vitreoretinal surgeons stopped
tools) and 23-gauge (0.75-mm tools) surgeries are signifi- using subconjunctival antibiotics after cataract surgeons
cantly larger than wounds made during 25-gauge (0.5-mm abandoned the practice; however, topical antibiotics achieve
tools) surgery, increasing the likelihood of wound leaks and minimum inhibitory concentration antibiotic levels in the
making wound construction more challenging. Tools used anterior chamber but not in the vitreous cavity of phakic or
in 23-gauge surgery were developed to address tool stiffness pseudophakic eyes. Harry W. Flynn, as well as the author,
and flow restriction issues associated with the first-genera- recommend using both an antibiotic effective against com-
tion 25-gauge tools (6). Subsequent generations of 25-gauge mon Gram-positive organisms and one effective against
tools have increased tool stiffness and reduced flow restric- virulent Gram-negative organisms. The authors prefer
tion; therefore, the authors do not see an advantage of administering 20 mg tobramycin subconjunctivally and cef-
23-gauge surgery. tazidime 50 mg; vancomycin 25 mg is a good alternative to
When withdrawing the cannulas at the end of a case, the ceftazidime if the patient is allergic to penicillin. Injection in
surgeon should take care to avoid wound eversion. Cannulas the inferior cul-de-sac may help the surgeon avoid reflux of
should be withdrawn slowly on the same trajectory as they the aminoglycoside into the eye and, therefore, the possibil-
were inserted, because the sclera is relatively inelastic and ity of retinal toxicity.
the goal is to close the scleral tunnel. Scleral tunnel closure
is also dependent on leaving the infusion set at least 25 mm
Hg, which will cause the intraocular pressure (IOP) to press VITRECTOMY OBJECTIVES
on the inside of the wound. The surgeon should use point
pressure with smooth forceps, not a cotton-tip applicator, on Vitreous removal requires a thorough understanding of
the scleral tunnel to press on the outside of the tunnel, simi- surgical anatomy and must proceed in a systematic fashion.
larly to how pressure is placed when withdrawing a needle The vitreous body should be thought of in terms of discrete
or IV line from the antecubital vein. The conjunctiva should surfaces that are removed in a specific order. The goals should
be repositioned while point pressure is applied on the tun- not be band cutting, obtaining a view on the posterior pole, or
nel. Some surgeons withdraw the cannulas over the endoil- core vitrectomy only but should be directed at management
luminator. Withdrawing on the insertion trajectory, leaving of the underlying vitreoretinal process. When vitrectomy
the infusion turned on, and point pressure on the tunnel are was performed using rotating cutters with syringe-controlled
far more important in achieving tunnel closure, though. suction, the surgeon was able to remove much of the vitreous
If the wounds leak, then they should be sutured with- without moving the instrument from the center of the vitre-
out hesitation with a single, transconjunctival absorbable ous cavity. This occurred because of excessive suction and
polyglycolic suture such as 8-0. PolySorb. PolySorb has cutter winding, which drew the vitreous inward; however,
been associated with less irritation than polyglactin (Vicryl, such traction is now recognized to be dangerous. From this
Novartis, Basel, Switzerland) (7). It is not necessary to open traction-induced central relocation of the vitreous arose the
the conjunctiva to suture the sclerotomy. Some surgeons erroneous concept of “core vitrectomy.” In fact, many eyes
unwisely use sutured-on contact lens support rings, which with sufficient vitreoretinal disease to require vitrectomy do
can damage the conjunctiva (8). not even have a vitreous “core.” Recent trauma, rarely recent
Transconjunctival 23-gauge and 25-gauge, sutureless retinal detachments, and macular hole cases may have rela-
vitrectomies allow flexibility in cannula location. The surgeon tively normal vitreous requiring core vitrectomy.
48 SECTION II ■ Surgical Technology and Techniques

Fig ure 6 .1 ■ Penetration through


the opaque PVC should be m ade
nasally or into an area known to
have PVD and no d etachm ent if
p ossible as determ ined b y ultra-
sound or ophthalm oscopy.

Better cutters, high cutting rates, fast fluidic controllers, requiring trans–pars plana vitrectomy usually have total
and proportional (linear) suction control facilitate cutting vitreoretinal contact, partial posterior vitreous detachment
the vitreous without moving it from its original position. The (PVD) with a conical vitreous configuration, or total PVD
surgeon accustomed to low-performance systems may at first with a frontal plane configuration. Entry into the PVC should
be confused by the better systems’ minimal pulsatile traction be made in an area nasally (Fig. 6.1) or preferably known
and may falsely conclude that they are not working. to have a PVD by ophthalmoscopy or ultrasound. The por-
If the eye is aphakic at the commencement of vitrec- tions of PVC extending between areas of vitreoretinal adher-
tomy, the anterior vitreous cortex (AVC) should be removed ence are known as bridging and are areas where tangential
first, starting centrally and progressing peripherally. Any traction occurs. All portions of the PVC not in contact with
attachments to anterior segment wounds or the iris should the retina, that is, both the conical surface and the bridging
be removed before proceeding posteriorly. In phakic eyes, portions, must be removed to relieve traction. However, the
the AVC is frequently adherent to the posterior lens cap- vitreous “skirt,” which is the anterior edge of the truncated
sule, making removal difficult without lens damage. A clear cone, must be treated differently. Because retinal breaks can
AVC not causing traction should be avoided in eyes with- be caused by surgically induced traction on the skirt, only
out retinal detachment to reduce lens damage. Similarly, the enough skirt should be removed to allow satisfactory surgi-
AVC should be retained in pseudophakic eyes to reduce the cal visualization and to prevent the superior skirt from cov-
chances of fogging due to condensation on the intraocular ering any portion of the view when the patient is upright.
lens (IOL) in eyes that have had a YAG capsulotomy. If blood products are incorporated in the layers of the skirt,
Pars plana infusion cannulas permit the surgeon to they should be carefully debulked to diminish postoperative
change sides between the vitrectomy instrument and the hemolytic and erythroclastic glaucoma.
endoilluminator, thereby gaining access to the whole pos-
terior curve of the lens. Removal of the AVC in a phakic eye
requires direct microscope viewing and coaxial plus endoil- EPIRETINAL MEMBRANES
lumination without a contact or wide-angle system to avoid
lens damage. A combined nasal and temporal approach is The PVC may include new collagen and glial cells on its sur-
best for AVC removal in phakic eyes. Eyes with fibrovascu- face at sites of vitreoretinal adherence in addition to hypocel-
lar proliferation on or near the pars plana or a significant lular contraction of preexisting vitreous collagen. This type
inflammatory component should have lensectomy with for- of pathoanatomy is usually referred to as an epiretinal mem-
ceps capsule removal as a precaution against the formation brane (ERM). The treatment of ERM is a challenging and
of a cyclitic membrane at the lens-AVC interface. essential component of management or vitreoretinal cases.
After removal of the AVC, the second objective is usu- The approach to ERM varies with the disease; specific tech-
ally the removal of the posterior vitreous cortex (PVC). Eyes niques will be discussed with the specific diseases.
CHAPTER 6 ■ General Posterior Segm ent Techniques 49

Forceps membrane peeling, scissors segmentation, and of the product by reducing the number of steps or processes.
scissors delamination are all useful approaches to ERM. The A central theme of this book is to present a simplified, faster,
goal of ERM surgery is to reattach the retina while mini- less-step, less-tool approach to vitreoretinal surgery.
mizing recurrences and complications. If the membrane is At present, the authors use end-grasping forceps for all
loosely adherent and can be removed without iatrogenic epiretinal and subretinal membrane peeling and fine curved
retinal breaks or bleeding, forceps membrane peeling is scissors for all segmentation and delamination. Pics and bent
the best approach. In diabetes, retinopathy of prematurity needles are not needed for EMM dissection and increase
(ROP), and some trauma and proliferative vitreoretinopathy the potential for retinal trauma. End-grasping forceps and
(PVR) cases, the membrane is usually quite adherent, requir- curved scissors are the only dissection tools used by the
ing scissors segmentation and/or delamination. Excellent authors for virtually all vitreoretinal surgery. The following
visualization is necessary for ERM dissection. section describes a unified approach to tool and technique
selection for vitreoretinal surgery that was developed for use
by novice and advanced surgeons alike.
Sim p lifi e d Ep ire t in al Me m b ran e Disse ct io n
Current approaches to vitreoretinal surgery utilize a wide
variety of techniques and technologies. The techniques for Man ag e m e n t o f Ep ire t in al Me m b ran e s
PVR, proliferative diabetic retinopathy (PDR), ROP, epimac-
Membra ne Peeling
ular membranes (EMMs), macular holes, and other causes of
vitreoretinal traction typically vary considerably with the dis- Robert Machemer, developed ERM peeling only 2 years
ease process. Fibrin syndrome, corneal problems, cataract, after he developed pars plana vitrectomy. He used a bent
light toxicity, postanesthesia complications, postoperative 23-gauge needle to peel macular puckers, which we now call
pain, lid and conjunctival hyperemia, and edema are more EMMs. His technique, although effective, required a sharp
frequent after longer procedures. Postoperative atelectasis, needle tip adjacent to the retina surface. Conor O’Malley,
infection, pulmonary embolism, and increased length of soon thereafter introduced the pic—a rounded tip instru-
stay have been associated with longer operating times in ment, safer for membrane peeling. The principal author
the surgical literature. The principal author has observed a developed forceps membrane peeling using end-grasping
relationship between number of tools used, longer operating forceps and a pinch peeling, en face technique because
times, and worse surgical outcomes. A unified, all-disease membranes slip off the needles and pics and because for-
approach to tools, techniques, and algorithms could theo- ceps were still required to remove the membrane from the
retically reduce costs associated with longer operating times, eye. A single-step, end-grasping forceps membrane peeling
staff training time, setup time, and instrument acquisition approach is safer and faster than initiating peeling with a
and maintenance. Advancements in manufacturing technol- blade, pic, or membrane scraper and subsequently removing
ogy generally decrease the cost while increasing the quality the ERM with forceps (Fig. 6.2). End-grasping forceps are

Fig ure 6 .2 ■ Mem brane peeling


with end-grasping forceps does not
require entering into the potential
space between the retina and the
m em b ran e or fin d in g an ed g e;
therefore, it is m uch less likely to
d am age the retina than a p ic or
an approach using a forceps blade
under the retina.
50 SECTION II ■ Surgical Technology and Techniques

used with both blades on the anterior surface of the ERM, Anslem Kampnik, MD, has emphasized that ILM
a safer approach than attempting to place one blade under peeling in EMM surgery produces a reduction in recur-
the membrane risking retinal damage similar to peeling with rence rates. The authors have been following his recom-
needles, microvitreoretinal (MVR) blades, or pics. Using pics mendations by peeling ILM in all EMM cases for several
or MVR blades requires finding or constructing an “edge,” years; they agree with his observation and believe that
which is a potentially dangerous maneuver. Finding the an additional benefit is the elimination of residual striae,
outer perimeter of an ERM can be difficult with thin, so- thereby producing better visual outcomes and faster
called “glassy,” membranes; retinal surface damage can result improvement.
from using this approach. Making an edge involves using an
MVR blade to slice through the ERM, a potentially danger- Staining and Particulate Marking
ous maneuver if the patient moves or the visualization is not Many surgeons still use ICG staining of the ILM, although its
optimal. The senior author developed the MVR blade and use is decreasing. The authors have never used ICG because
was an early adopter of pics but has used only end-grasping of concerns about the toxicity of the agent and vehicle and
forceps membrane peeling for more than three decades. An because it is simply unnecessary. One factor driving danger-
end-grasping forceps technique requires perfect alignment ous and unnecessary ICG use is using noncontact optical
of the forceps blades at the tip, which cannot be maintained systems such as the BIOM and EIBOS systems that decrease
with blades with long axial gripping surfaces, particularly both axial and lateral resolutions. An additional factor driv-
with multiple cleaning and sterilization cycles for reusable ing ICG use is using inadequate forceps that are incapable of
tools or, worse yet, reuse of disposable forceps. Smaller end grasping.
23-gauge and 25-gauge forceps are more vulnerable to dam- Triamcinolone particulate marking, often incorrectly
age during cleaning and sterilization; the authors use dispos- referred to as staining, is not specific to ILM or ERMs. The
able 25-gauge Grieshaber Revolution DSP ILM (Alcon, Ft. optimal use for triamcinolone is for visualizing the vitre-
Worth, Texas) forceps for all ERMs, PVR membranes, and ous. Triamcinolone is more often required with a Tornambe
internal limiting membrane (ILM) peeling without pics or Torpedo (Alcon) or a chandelier illumination system that
MVR blades. The senior author developed conformal forceps produces diffuse illumination, making visualization of the
so that the blade tips would have the same radius of curva- vitreous, ILM, and ERMs difficult. Triamcinolone is useful
ture as the retinal surface in contrast to the Eckardt forceps when learning vitrectomy and for the identification of the
(Dutch Ophthalmic USA, Exeter, New Hampshire) that have PVC anatomy in complex cases.
sharp corners that tend to grasp retina. The DSP ILM forceps
are small enough that the radius of curvature is not a signifi-
Scissors Segmenta tion a nd Dela mina tion
cant issue.
Yasuo Tano, MD, developed the diamond-dusted mem- The principal author developed scissors segmentation of
brane scraper, which some surgeons have adopted. The ERMs more than three decades ago to address the issue
authors prefer the end-grasping forceps technique because of iatrogenic retinal tears caused when attempting to peel
it is less likely to damage the retinal surface, especially in adherent ERMs, which are common in diabetic traction
smaller 23-gauge and 25-gauge form factors that produce retinal detachment cases. Segmentation is used to reduce
greater force per unit area. tangential force on the retina by separating an ERM into
“epicenters,” (Fig. 6.3) better termed vascularized adherence
sites. Segmentation is performed by placing one blade of the
Interna l Limiting Membra ne Peeling
scissors under the ERM, between the retina and the ERM,
ILM peeling was introduced for macular hole surgery and and the other blade above the ERM. Although this was ini-
has been widely accepted; it is generally agreed that it tially performed with “vertical scissors,” curved scissors are
improves closure rates. A few papers suggested that ILM far superior for segmentation. Because blade width is much
produced worse visual outcome despite hole closure; greater than blade thickness, vertical scissors require more
the authors believe that this is due to indocyanine green space between the retina and the ERM than curved scissors
(ICG) toxicity, not ILM peeling. The standard justification (Fig. 6.4). Segmentation is now primarily used for access seg-
for ILM peeling is to remove “tangential traction.” Successful mentation to expose the delamination plane. Using curved
ILM peeling guarantees removal of vitreomacular traction, scissors expedites transitioning from access segmentation to
residual PVC not contiguous with the detached vitreous, delamination without tool exchange.
and any ERM that might be present. The principal author The principal author developed scissors delamination
believes that an additional and critical role for ILM peeling 2 years after developing segmentation to address the issue of
is increasing retinal compliance by approximately 50% so residual ERM after segmentation. Residual ERMs in diabetic
that the inward-directed surface tension force from a gas traction retinal detachment cases resulted in small areas of
bubble can almost immediately reapproximate the inner chronic retinal elevation that ultimately caused atrophic
margins of the hole. retinal holes and late rhegmatogenous retinal detachments.
CHAPTER 6 ■ General Posterior Segm ent Techniques 51

Fig u re 6 .3 ■ Scissors seg m en-


tation of ERMs reduces tangential
traction b ut leaves ep icen ters,
wh ich m ay b leed o n th e cu t
ed g es. The b lood acts as a sub -
strate for glial recurrence. Residual
ERM m ay cause residual elevation
and late retinal detachm ent from
atrop h ic holes. Seg m entation is
used when the m em brane is too
ad h erent to p eel an d the retina
too thin for delam ination.

Scissors delamination is performed by inserting both for conformal delamination. Curved scissors are preferred
scissors blades in the potential space between the ERM and the to “horizontal” scissors because the curvature of the scissors
retina and severing the attachment points (Fig. 6.5). Simplisti- matches the curvature of the retina, reducing the likelihood of
cally, segmentation means to cut up the ERM, and delamina- impaling the retina with scissors tips. The same design concept
tion means to cut it off. Initially, delamination was performed is used for skis—the tips curve up. If the scissors are intro-
with “horizontal” scissors, actually 135 degrees. For almost duced under the ERM with the blades wide open and then
two decades, the senior author has used only curved scissors closed, attachment points will be ripped instead of sheared,

Fig ure 6 .4 ■ Fine curved scissors


are better than vertical scissors for
segm entation because blade width
is greater than blade thickness and
the curve is conform al with retinal
contour, reducing iatrogenic reti-
nal breaks.
52 SECTION II ■ Surgical Technology and Techniques

Fig u re 6 .5 ■ Scissors d elam i-


n ation of ERMs is the p referred
m eth od of rem ovin g ad h eren t
m em b ranes as it p erm its com -
p lete rem oval of ERM.

which causes retinal breaks (Fig. 6.6). When the scissors are cut, pull back slightly, move laterally to the next attachment
introduced under the ERM with the blades closed and then point, advance slightly, and cut again (Fig. 6.7). It is not nec-
opened similar to the technique used for blunt dissection in essary to lift or stabilize the ERM when disposable scissors or
general surgery, attachment points will be ripped instead of reasonably new reusable scissors are used. Because an ERM is
sheared, creating retinal breaks. The optimal technique is to taut and there is a pressure gradient across the retina, the ERM
open the scissors just enough to engage an attachment point, will scroll up and separate from the retina without lifting.

Fig ure 6 .6 ■ The shear point of


scissors m oves forward as the scis-
sors close, creating an exclusion
force that can tear the retina.
CHAPTER 6 ■ General Posterior Segm ent Techniques 53

Fig u re 6 .7 ■ Th e scissors are


m oved back and forth while m ak-
ing sm all cuts with the tips barely
op en ing ; th e ERM will scroll up
b ecause of in h eren t elasticity,
rarely requiring lifting.

Inside-Out Versus Outside-In Technique PVC. When most or all the ERM has been delaminated using
Robert Machemer, used an outside-in technique for bent an inside-out direction, creation of PVD will be accomplished
needle membrane peeling, and many surgeons emulate this as well or it will be irrelevant because there are no longer any
technique when using delamination. A better approach for posterior attachment points.
end-grasping forceps membrane peeling, scissors segmenta-
tion, and scissors delamination is to initiate the dissection
Bima nua l Surgery
centrally and peel, segment, or delaminate outward. Inside-
out dissection is safer because the retina is thicker and Bimanual surgery is a poor term; surgeons have always used
100 times stronger centrally than in the periphery, especially both hands. The point of bimanual surgery is to accomplish
in ischemic patients and postpanretinal photocoagulation; what the author calls “forceps stabilization of epiretinal
the retina is redundant centrally in traction retinal detach- membranes,” using forceps to offset the scissors push-out
ment cases, and the view is better centrally. force (Fig. 6.8). Scissors cut at a point that starts near the
pivot point and moves toward the tip, creating undesirable
push-out force. By contrast, a vitreous cutter cuts on a curved
En Bloc Dissection
line, which makes manufacturing much more difficult and
En bloc dissection, as initially described, utilized an out- requires vacuum for imbrications into the port but eliminates
side-in technique and advocated using the PVC to lift the push-out force. An unintended consequence of bimanual
ERM. Putting traction on the PVC results in peripheral retina surgery is to inadvertently lift the ERM, thereby creating iat-
breaks while the surgeon is focusing on the ERM dissection. rogenic retinal breaks. Making small delamination cuts with
En bloc is a poor term when applied to ERM dissection; it scissors almost closed obviates the need to lift the ERM and
is only meaningful in cancer surgery where the notion is to visualize the attachment points. Bimanual surgery requires
minimize the dispersion of cancer cells. A better way to think the use of either illuminated tools, which are not available in
about this issue is to forget the outdated notion that the stan- 23-gauge or 25-gauge systems, or a chandelier or Torpedo,
dard surgical algorithm is first performing core vitrectomy, which produces diffuse illumination, making visualization of
then creating a PVD, and finally dissecting ERMs. If a pos- vitreous, ILM, and transparent membranes more difficult.
terior vitreous separation PVD is not present in a diabetic
traction retinal detachment (TRD) case, proceed directly to
Viscodissection
an inside-out delamination starting at or near the optic nerve
head. It is not necessary to remove all ERM in one piece as Viscodissection is performed by injecting a viscoelastic in
suggested by the term en bloc; it is preferable to segment the potential space between the ERM and the retina. Viscodis-
in several pieces, each of which will be contiguous with the section is seldom performed because it has not been shown
54 SECTION II ■ Surgical Technology and Techniques

Fig ure 6 .8 ■ Illum inated forceps


allow stab ilization of the ERM to
offset p eelin g , seg m en tation , or
delam ination forces.

to be advantageous and it increases cost and operating time. believe that using excessive diathermy on retinal surface
Viscodissection pressurizes the potential space between the bleeders can result in subclinical retinal necrosis and late
ERM and the retina, which can result in a hydraulic retinal atrophic holes. On occasion, larger vessels may require dia-
break if the ERM is tightly adherent. If the ERM is adher- thermy before or after transection, especially in delayed
ent with no gap between the ERM and the retina, the vis- trauma surgery and when performing retinectomy.
cocannula may create a retinal break during introduction.
Viscoelastic substances significantly decrease the interfacial
St ab ilize d Surg e ry
tension of silicone oil, increasing the likelihood of emulsifi-
cation. It is virtually impossible to remove the viscoelastic; Retinal movement in response to fluidic or mechanical dis-
the viscous material potentially increases glial recurrence section forces is often an indication that retinal tearing may be
rates by retaining cells, fibronectin, and cytokines such as imminent. Ideal vitreous and ERM removal would not cause
vascular endothelial growth factor (VEGF), basic fibroblast any force on the retina but is impossible as described above.
growth factor, and transforming growth factor beta. Many techniques have been developed that, although pro-
moted for a variety of reasons, primarily function by reduc-
ing surgically induced retinal motion: (a) flow limiting using
Power Scissors a nd Forceps
high cutting rates and 25-gauge fluidics, (b) forceps stabiliza-
Power scissors and power forceps address the problem of tion of ERM (bimanual surgery), (c) perfluorocarbon liquids
inadvertent motion of the tool tip during manual actuation. used during ERM dissection, and (d) viscodissection.
A potentially offsetting issue is inadvertent hand motion due Perfluorooctane has twice the specific gravity of bal-
to whole leg motion while moving the foot pedal. Older gen- anced salt solution. Therefore, it produces twice the gravita-
eration power scissors and power forceps were too heavy and tional force on the retina (11).
did not have high-fidelity proportional control; the Constel- In addition, inertial stabilization, because of the F =
lation (Alcon) power scissors and forceps have solved these MA relationship, offsets pulsating fluidic forces. Perfluo-
design issues and also have a single foot pedal control mode rocarbon liquids are immiscible in balanced salt solution,
for bimanual surgery. blood, and subretinal fluid (SRF) and are therefore used
because of surface tension properties (interfacial tension) to
unfold giant breaks and remove SRF in all types of retinal
Hemosta sis
detachments (Fig. 6.9). The interfacial tension effect also
Endolaser is better than diathermy for treating the severed prevents transretinal fluid flow (uveal-scleral outflow) and
vessel ends of attachment points after delaminating ERMs tends to reduce retinal edema while the perfluoron (PFO)
in diabetic traction retinal detachment surgery. The authors is present.
CHAPTER 6 ■ General Posterior Segm ent Techniques 55

Fig u re 6 . 9 ■ PFO st ab ilizes


the retina to offset forces due to
m em brane peeling while floating
out subretinal fluid through ante-
rior retinal breaks.

Ep ire t in al Me m b ran e Disse ct io n that excessive retinal surface damage or bleeding will result
Flo wch art if delamination is used. This is sometimes the case with
atrophic, long-standing, diabetic traction retinal detach-
Removal of all apparent vitreoretinal traction should pre-
ments because of ischemia and prior extensive panretinal
cede ERM dissection unless there is limited separation of
photocoagulation (PRP).
the posterior cortical vitreous from the retina. The princi-
If the retina is pushed away by the scissors or if peel-
pal author developed the concept of inside-out membrane
ing is causing excessive retinal movement, forceps stabiliza-
delamination preceding removal of the posterior cortical
tion of the ERM can be used. If retinal mobility is excessive,
vitreous for cases without apparent PVD. Abrams and
PFO or air stabilization of the retina can be used; this is
Williams (12) subsequently recommended an outside-in
typically required in retinal detachment, giant break, and
approach for delamination before the cortex is removed,
PVR cases. Fast cutting and 25-gauge fluidics produce port-
which they termed “en bloc.” In their paper, they rec-
based flow limiting that greatly increases fluidic stability
ommended using traction on the vitreous to lift up the
by reducing pulse flow, thereby reducing pulsatile retinal
ERM. This approach may create peripheral retinal breaks
movement as well as surge after sudden deformation of
because of counter traction and is not recommended
ERM through the port.
by the authors. Many now call the inside-out delamina-
After all vitreoretinal traction and all apparent ERM-
tion before posterior cortical vitreous separation method
induced retinal foreshortening has been managed, inter-
“en bloc” even though that is not how it was originally
nal or direct transscleral needle drainage of SRF should
described.
be initiated in cases with a rhegmatogenous component.
The first stage of ERM dissection is to carefully test
After retinal movement in the direction of attachment
the adherence of the ERM to the retina. In general, EMM,
ceases, internal FAX should be started while continuing
macular hole or ILM, and PVR have less adherence than
to drain SRF. If the retina does not completely reattach
PDR or ROP. If the adherence is low, inside-out forceps
or subretinal air appears, the remaining traction should
membrane peeling is the best approach. If the adherence is
be managed by further vitrectomy, peeling, segmenta-
high, inside-out delamination is the next step. Access seg-
tion, delamination, subretinal traction removal, or, lastly,
mentation can be used to facilitate finding a tissue plane
retinectomy. The silicone soft-tip cannula allows the aspi-
for delamination. In general, almost all ERM is delami-
ration of SRF and preretinal hemorrhage while minimiz-
nated using an inside-out orientation in PDR cases. The
ing trauma to the retina and is the preferred tool for FAX
procedure is halted at the segmentation step if it is thought
by the authors.
56 SECTION II ■ Surgical Technology and Techniques

Fig ure 6 .1 0 ■ Conform al cutter


d elam in atio n rem o ves ep ireti-
nal tissue without rem oving the
underlying retina.

Cut t e r De lam in at io n The tool can then be moved, lifting the membrane or vitreous
away from the retina (13). The suction force can, however,
Modern high-speed microincisional vitrectomy cutters allow
cause retinal traction, and certain membranes are difficult to
safe, direct delamination of epiretinal tissue with the cutter.
imbricate into a port. Forceps peeling is preferable in most
Cutter delamination permits continuous aspiration of blood
instances except for PVD creation in macular hole cases.
from the dissection surface, improving surgical visualiza-
tion, decreases surgical time by minimizing instrument han-
dover, decreases surgical costs, and decreases intraoperative “Ch o p st icks” Me m b ran e Pe e lin g
hemorrhage from the hypotony that occurs during replacing If a flexible ERM has been partially lifted from the retina
instruments on open cannulas. surface by peeling, removal can be completed by holding
Two techniques of cutter delamination can be performed: the membrane between the endoilluminator and the vitrec-
conformal cutter delamination and foldback cutter delami- tomy probe. However, if excessive pressure is utilized with
nation. Conformal cutter delamination involves placing the this bimanual method, the instruments may slip past one
cutter opening directly against the edge of the epiretinal tissue another (14). Outward spreading of a pair of blunt tools
and modulating the angle of attack and suction until the cut- such as the probe and the endoilluminator can be used to
ter removes the epiretinal tissue without removing the under- separate the PVC or an ERM from the retina by blunt dissec-
lying retina (Fig. 6.10). Segmentation can also be performed tion. The blunt dissection, spreading method is very useful
with the cutter by placing the cutter directly over the valleys of in PVR cases.
retinal folds, between retinal attachment points, where clear-
ance from the retina is the highest, and removing the bridg-
ing tractional tissue. Foldback delamination involves placing Vacuum Cle an in g –Ext rusio n
the cutter above the epiretinal tissue, with the cutter opening When an opening is first made in the PVC, nonclotted
facing in the direction of the leading edge of the dissection blood, hemoglobin products, and erythroclasts tend to
and allowing the epiretinal tissue to “foldback” into the cutter flow out the opening into the vitreous cavity, especially if
(Fig. 6.11). This is best performed when there is a free mobile suction is turned off and on repeatedly. This is often falsely
edge to the epiretinal tissue that can move toward the cutter. perceived as active bleeding and should be removed by
continuous vacuum cleaning (Fig. 6.12) before continu-
Suct io n Me m b ran e Pe e lin g o r Po st e rio r
ing with PVC truncation. This approach ensures excellent
Vit re o us De t ach m e n t Cre at io n
visualization and avoids the flow of blood products to the
If a loose edge of a flexible ERM or PVC requires peeling, it can lens, cornea, and trabecular meshwork.
be held in the port of the vitrectomy cutter using the suction- The vacuum cleaning method utilizes a soft-tip cannula
only mode of the foot pedal or a soft-tip extrusion cannula. and a nonpulsatile, controlled pressure gradient across
CHAPTER 6 ■ General Posterior Segm ent Techniques 57

Fig ure 6 .1 1 ■ Foldback delam i-


n ation is b est p erform ed wh en
there is a free m obile edge to the
ep iretin al tissue th at can m ove
toward the cutter.

the needle port. Keeping the cannula away from the infu- fluted) needle, in which egress is controlled by the surgeon’s
sion port minimizes turbulence. If console or foot pedal forefinger over the exit port on the side of the handle (15).
controlled, this method has been referred to as “extrusion” This has been incorrectly termed passive egress. Whether
(38). The key to understanding extrusion is to remember these techniques are considered “active” or “passive” is
these features: (a) end opening, (b) low, precisely controlled irrelevant; precise control of transorifice pressure is the key
pressure gradient, and (c) nonpulsatile. The original and concept. The extrusion method utilizes a 20- or, preferably,
now-obsolete form of vacuum cleaning utilized the flute (not 25-gauge soft-tip cannula, vitreous cutter on suction-only

Fig u re 6 .1 2 ■ After m akin g


an op en in g in th e PVC, sub -
PVD b lood p rod ucts sh ould b e
rem o ved b y extrusion with a
soft-tip cannula b efore continu-
ing with PVC truncation.
58 SECTION II ■ Surgical Technology and Techniques

mode or an end-aspirating laser probe if the surgeon elects vitreous). This immiscibility is best referred to as interfacial
to use 20-gauge technology connected to the console-based surface tension.
suction system, with foot control by the surgeon (16). Pro- Surface tension is a physical property of liquids that
portional (linear) suction control allows greater control over depends on intermolecular attraction forces within the liquid
the extrusion process and has supplanted the flute needle for itself. Whereas deep in a liquid all molecules are attracted to
vacuum cleaning. Proportional suction applied to a cannula neighboring molecules in all directions, in the surface of a
is very effective for removing blood products, substances to liquid, molecules are only attracted to those in the plane of
be exchanged, and SRF. Extrusion allows for reflux using the the surface and underneath it, but not above the surface. This
foot pedal. causes surface liquid molecules to form a “film” of closely
Vacuum cleaning–extrusion should be utilized only packed molecules. The energy required to break this film is
when there is a region of nonclotted blood in the vitreous termed surface tension. Different liquids will have different sur-
cavity, the sub-PVD space, or on the retinal surface. The can- face tensions, which depend on the chemical characteristics
nula should never touch the vitreous or retinal surface but of their molecules and their intermolecular attraction forces.
may be held very close to the retina if the transorifice pres- Liquids with high electrostatic intermolecular attraction
sure is kept low. Intentionally lowered IOP can be used to forces, such as water (surface tension of 73 dynes/cm), have
induce bleeding, so that the source of the hemorrhage can higher surface tension than nonpolar liquids, such as silicone
be located and treated. oil (surface tension of 20 dynes/cm). Perfluorocarbon liquids
The Chang end-aspirating endolaser probe is ideal for have intermediate surface tension properties (50 dynes/cm).
surgeons using 20-gauge technology for removal of prereti- When two immiscible liquid phases interact, the particular
nal blood and active bleeding while treating surface bleeders tension at the interphase is called interfacial tension. The sur-
and vascular attachment points and performing PRP in dia- face tension or interfacial tension values of clinical significance
betic cases. The authors only perform 25-gauge surgery. for vitreous surgery are water/air (or gas) = 73 dynes/ cm, sili-
cone oil/water = 40 dynes/cm, and perfluorooctane/water =
50 dynes/cm. As mentioned above, vitreous, retina, BSS, and
aqueous humor behave as water.
EXCHANGES AND SUBRETINAL
Surface tension management of rhegmatogenous
FLUID DRAINAGE retinal detachments is based on creating a “liquid surface”
interphase at the level of a retinal hole (or holes). This inter-
Surface Te n sio n Man ag e m e n t an d Exch an g e s
phase, by virtue of its surface tension, creates a film over the
Air, various inert gases (SF6, C3F8) (17–20), silicone oil, hole that prevents the passage of fluid through the hole and
and perfluorocarbon liquids are utilized primarily because restores the transretinal pressure gradient (Fig. 6.13). This
they are immiscible in water (balanced salt solution, retina, enables the RPE cells to absorb and drain the SRF, resolving

Fig ure 6 .1 3 ■ Interfacial tension


allows restoration of the transreti-
nal pressure gradient.
CHAPTER 6 ■ General Posterior Segm ent Techniques 59

the retinal detachment. The term “tamponade” is meaning- retinal breaks with endophotocoagulation, and then perform
less from a physics perspective. It is derived from French air-silicone exchange (ASX) for PVR cases. They are often
and means “to plug” and not “to press.” If a gas or silicone surprised when the retina is detached on the first postopera-
bubble surface is not in direct contact with a retinal hole, tive day; this is because the interfacial tension of air is greater
it will not “seal” the hole, and the rhegmatogenous compo- than that of silicone oil and the force of residual traction is
nent will not be adequately managed. The interfacial tension greater than the interfacial tension of oil. Emulsification is
of an air or gas bubble “seals” the hole even when it is not in related to many factors, but the most numerically significant
contact with the RPE. parameter is interfacial tension, which is not related to vis-
Density (specific gravity) determines if a substance will cosity explaining why the emulsification rate of 1,000 cs oil
sink or float and the force it will produce at the retinal con- is same as 5,000 cs oil.
tact surface. Air, gases, and silicone oil are less dense than There are many ways to think about interfacial tension
saline or vitreous and migrate opposite to the gravity vec- agents: Does the agent float (air, gas, and silicone oil) or sink
tor, and they rise inside the eye. Perfluorocarbon liquids are (PFO)? Does the agent absorb over time (air, gas) or remain in
almost twice as dense as saline or vitreous and, therefore, place (PFO, oil)? Can the patient see through the bubble (sili-
migrate to the lowest part of the eye. cone oil) or not (air, gas, and PFO)? Can the substance be left
Air, SF6, and C3F8 have identical intraoperative proper- in the eye long term (oil) or only medium term (gas, PFO)?
ties (21,22), although they are different with respect to bub- Factors in silicone oil emulsification include steep bub-
ble expansion and duration in the eye. Gases such as C3F8 ble radius due to an incomplete fill and the use of viscoelas-
(23–29) produce greater duration in the eye because the gas- tics or postoperative bleeding or inflammation, all of which
eous diffusion rate of these larger molecules is much slower decrease interfacial tension. Nystagmus is another factor in
than nitrogen in the blood. Abrams showed that an empiri- emulsification because it increases the shear rate, in essence
cal isoexpansive mixture of each inert gas with air could be spinning off small bubbles.
determined so that complete intraocular filling could be used
without increased postoperative pressure. The isoexpansive
Te rm in o lo g y
concentration is approximately 25% for SF6 and 18% for
C3F8. Nitrous oxide anesthesia should not be used with air Logical terminology is essential in a procedural context as well.
or any of the inert gases as it can cause rapid intraoperative English language convention is that word order is relevant;
expansion of a preexisting bubble or marked postoperative FAX means replace fluid with air. Often, surgeons incorrectly
shrinkage of the bubble. call this gas-fluid exchange, which actually means remove
gas and replace it with fluid. The preferred naming conven-
tion is to use IDS for internal drainage of SRF, FAX for fluid-
In t e rfacial Te n sio n Ag e n t s
air exchange, AGX for air-gas exchange, ASX for air-silicone
Exchange techniques are utilized in vitreoretinal surgery to exchange, PGX for perfluorooctane-gas exchange, PSX for
inject or remove interfacial tension agents: air and longer perfluorooctane-silicone exchange, etc.
acting gases, perfluorocarbon liquids, and silicone oil after
the vitreous has been removed. Although these substances
Drain ag e o f Sub re t in al Fluid
are often called tamponade agents, this is a poor term.
Using correct terms from physics and chemistry promotes Total FAX and near total IDS are utilized by most vitreo-
the understanding of surgical concepts. For example, some retinal surgeons when repairing retinal detachments using
surgeons inject gas after EMM surgery thinking that it will vitrectomy. The principal author invented IDS over three
press on and eliminate retinal folds, which is simply not the decades ago in order to drain SRF through a preexisting or
case. Interfacial tension is not related to viscosity or density. iatrogenic retinal break or drainage retinotomy rather than
Air and air-gas mixtures produce greater interfacial tension through the sclera, choroid, and RPE, risking bleeding and
(70 nM/M) than silicone oil (40–45 nM/M) or perfluo- retinal incarceration. Unlike cryopexy during scleral buck-
rooctane (50 nM/M). The silicone-PFO interface produces ling, vitrectomy using internal drainage and FAX permits
only 5 to 7 nM/M interfacial tension, explaining why these postreattachment retinopexy, which in turn enables precise
substances become mixtures over time. Density determines location of the endolaser without overtreatment and treat-
where the bubble goes; interfacial tension determines what ing both the retina and the RPE, which results in stronger
it does. Many surgeons have said that PFO cannot be used adherence. In addition, internal drainage, when coupled
with 25-gauge surgery because it is too viscous, which is with FAX (Fig. 6.14), enables the assessment of residual vit-
incorrect; PFO is less viscous than BSS, although it has reoretinal traction, a maneuver the principal author refers to
almost twice the density demonstrating that density and as the reattachment experiment. An assessment of residual
viscosity are not related. PFO has approximately the same vitreoretinal traction can determine the need for interface
interfacial tension as silicone oil, although oil has over 1,000 vitrectomy, additional forceps peeling of ERMs, scissors seg-
times greater viscosity. Most surgeons use FAX coupled mentation or delamination, or retinectomy, all performed
with internal drainage of SRF to attach the retina, treat the “under” air.
60 SECTION II ■ Surgical Technology and Techniques

Fig u re 6 .1 4 ■ FAX req uires a


soft-tip or altern ative extrusion
cannula to rem ove fluid while air is
injected through the infusion can-
nula powered by the low-p ressure
air source on the console.

The first IDS was performed using the vitreous cutter, would be better controlled by using the console and foot
which is still a useful technique in some cases. Soon thereaf- pedal, a technique he called “extrusion.” The author imme-
ter, the principal author developed the flute needle, a nonta- diately adopted extrusion because it prevented inadvertent
pered, end-opening, blunt cannula with a port on the handle motion of the tip when covering or uncovering the port on
to control fluid egress with the surgeon’s fingertip (hence the handle and enabled more precise vacuum control as well
the term flute, like the musical instrument). This cannula as foot pedal–controlled reflux.
was also used to remove free blood products using a tech- Sandy Grizzard and Harry Flynn independently devel-
nique called “vacuum cleaning” similar to cleaning the bot- oped various flexible or soft-tip cannulas that are preferable
tom of a swimming pool. Many surgeons called this “passive to rigid cannulas for all applications (Figs. 6.15 and 6.16).
egress,” which is incorrect; IOP can produce a substantial Soft-tip cannulas are less likely to damage the retina, optic
pressure difference across the port relative to atmospheric nerve, RPE, or choroid due to patient movement or posi-
pressure, which is no safer than so-called active suction. tioning errors caused by suboptimal visualization. Soft-tip
Conor O’Malley soon thereafter suggested that fluid egress cannulas should be inserted through the retina break to

Fig ure 6 .1 5 ■ Straight soft-tip extrusion cannula. Fig ure 6 .1 6 ■ Angulated soft-tip extrusion cannula.
CHAPTER 6 ■ General Posterior Segm ent Techniques 61

Fig u re 6 .1 7 ■ In tern al d rain -


age can be perform ed through a
p reexisting retinal b reak or m id-
peripheral drainage retinotom y.

a position just above the RPE to reduce the likelihood of spot of diathermy can be made using the disposable bipolar
retinal incarceration in the port. Small-diameter, especially diathermy before making a small, round retinotomy with the
25 gauge, soft-tip cannulas are very flexible and will flex to single cut mode of the vitreous cutter. The diathermy needle
conform to the RPE surface when inserted through a reti- can itself be used to punch through the retina while cauter-
nal break or retinotomy. Soft-tip, 25-gauge cannulas always izing, leading to a small, round hole without hemorrhage. The
provide sufficient flow rates when used with proportional hole should be made rather posterior, away from retinal ves-
(linear) vacuum with a preset maximum of 650 mm Hg. sels but outside the temporal arcades. The soft-tip cannula can
IDS should be initiated before starting FAX to prevent SRF then be used to drain all the SRF through the retinotomy.
from being displaced posteriorly by the buoyancy of the air.
Internal drainage can be performed through a preexisting reti-
Th e Re at t ach m e n t Exp e rim e n t
nal break or midperipheral drainage retinotomy well away from
the macula and vascular arcades (Fig. 6.17). Internal drainage Internal (simultaneous) FAX combined with IDS is an excel-
should be continued during FAX to prevent the posterior dis- lent test for relief of traction on the retina. This can be
placement of subretinal fluid; the view is often transiently lost thought of as the reattachment experiment. If during this
until bubbles coalesce, necessitating very stable positioning of procedure the retina does not completely attach, further
the soft-tip cannula. Removal of the flap from flap tears not vitreous removal, forceps membrane peeling, scissors seg-
only eliminates traction but also reduces the likelihood of reti- mentation, scissors delamination, subretinal surgery, retine-
nal incarceration in the soft-tip cannula. It is best to perform ctomy, or scleral buckling is necessary. Surface tension
a slow yet continuous drainage of the SRF. This promotes the stabilizes the retina and allows visualization of mechanical
migration of all SRF toward the area of drainage and maxi- factors preventing reattachment. If all these techniques fail,
mizes drainage efficiency. Fast aspiration leads to reattachment this test serves to indicate inoperability.
of the area of drainage with peripheral areas of persistent SRF If subretinal air appears, it is indicative of residual trac-
that may not be contiguous with the area of drainage. tion that must be treated as outlined above. Direct needle
drainage of SRF posterior to a scleral buckle tied after the
subretinal air appears will remove subretinal air, but a poste-
Dra ina ge Retinotomy
rior drainage retinotomy is usually a better choice.
Although the senior author developed IDS and independently Total intraoperative reattachment eliminates postopera-
codeveloped “relaxing” retinotomy or retinectomy, drainage tive questions concerning leakage through the tear or the rate
retinotomies were infrequently used for many years. More of RPE pumping of SRF. In addition, retinopexy performed after
recently, small drainage retinotomies have been utilized safely reattachment allows for more precise localization and less risk
and effectively when the tear could not be adequately visualized of RPE migration and permits treatment of the retina as well as
or adequately reached with the drainage cannula. A small the RPE, ensuring better adherence. The guesswork method
62 SECTION II ■ Surgical Technology and Techniques

Fig u re 6 .1 8 ■ Fog g ing occurs


when the warm air bubble com es
into contact with th e relatively
cooler IOL, causing condensation.

of using a small expanding bubble without completion of SRF completed around all retinal breaks (Fig. 6.19). It is essential
drainage leaves many questions unanswered at the time of vit- to use an isoexpansive gas concentration as developed by
rectomy and, consequently, lowers the success rate. Gary Abrams to prevent high IOP or undersize bubbles.
Sulfur hexafluoride (SF6) should be mixed with air to
achieve a 25% concentration; 18% is appropriate for C3F8. It
In t rao cular Le n s Fo g g in g Durin g
is dangerous to estimate the volume of the air bubble in the
Fluid -Air Exch an g e
vitreous cavity and inject a higher gas concentration. This
Fogging is not specific to silicone IOLs, although they have a inaccurate method solves no problems and can result in very
greater thermal mass than polymethyl methacrylate (PMMA) high IOP and central retinal artery occlusion or inadequate
or acrylic IOLs and have a higher posterior capsular opaci- postoperative bubble size. Some surgeons use partial FAX
fication rate than acrylic IOLs. Fogging occurs when a YAG and higher gas concentrations, which are inaccurate and
laser capsulotomy has been performed and the cataract or prone to high IOP or inadequate bubble errors. The air-gas
vitreous surgeon removes the AVC, enabling the warm air concentration should be carefully determined; many errors
bubble to come into contact with the relatively cooler IOL have occurred by confusing cubic centimeters with percent,
causing condensation. PFO is ideal when fogging of the a potential disaster if a different size syringe is used than that
IOL occurs (Fig. 6.18). If fogging occurs, the air should used when the percent calculation was done.
be removed and the retina attached with PFO, followed by
endophotocoagulation around all retinal breaks, and finally
Liq uid Pe rfl uo ro carb o n Te ch n iq ue s
by placing the soft-tip cannula in the optic nerve cup to per-
form a PFO-gas exchange. Often, a few drops of PFO will Liquid perfluorocarbon (perfluorooctane, PFO) can be used
be retained if fogging occurs during the exchange, but these to remove SRF in cases without PVR or giant breaks. There
can be removed at a later time if necessary. Alternatively, ASX are several pros and cons of using PFO compared to IDS,
could be performed; condensation will disappear when the although the techniques can be combined. PFO should be
oil reaches the posterior surface of the IOL, and IDS fol- injected slowly, near the retinal surface, preferably into the
lowed by endophotocoagulation to all breaks can then be optic nerve cup or nasally to prevent damage to the macula.
completed under oil. The MedOne 25-gauge, dual-bore PFO injection cannula per-
mits fluid egress as the PFO is injected to maintain a normal
IOP (Fig. 6.20). The cannula must be retracted as the PFO
Air-Gas Exch an g e
level rises to avoid loss of PFO through the fluid egress port.
AGX is performed after IDS combined with FAX is used PFO is best injected by placing a full 7.5-mL vial in
to attach the retina and endophotocoagulation has been Alcon VFC MedOne dual-bore cannula.
CHAPTER 6 ■ General Posterior Segm ent Techniques 63

Fig u re 6 .1 9 ■ AGX p rod uces


an isoexp ansive concentration of
g as, sig n ificantly red ucing p ost-
operative variation in bubble size
and elevated IOP.

The VFC system from the Accurus and Constellation SRF and infusion fluid float anteriorly when PFO is
systems is ideal to control infusion of PFO into the eye. The injected into the vitreous cavity, which often results in SRF
surgeon should be cautious to keep the maximum infusion being displaced anterior to the most anterior retinal break
pressure set at 10 mm Hg and should continuously visual- unless a giant retinal break is present. Anterior displacement
ize the fundus during the VFI injection of PFO. A similar of SRF can be managed by extending a retinal break to the
technique can be used to inject tissue plasminogen activa- ora with scissors or the vitreous cutter, making a very small
tor into the subretinal space in cases of massive subretinal drainage retinotomy right at the ora, or by inserting a soft-tip
hemorrhage. cannula through a retinal break and carefully removing the
SRF without removing PFO.

Fig u re 6 .2 0 ■ Th e d ual-b ore


cannula m ust be retracted as the
PFO level rises to avoid loss of PFO
through the fluid egress port.
64 SECTION II ■ Surgical Technology and Techniques

Fig ure 6 .2 1 ■ Triam cinolone is


injected into the air bubble, which
reduces the foreign body reaction
that can occur with PFO.

Removal of the PFO should be made carefully, under fill is utilized and air turned on after total PFO fill; a very
air infusion, to prevent any layer of aqueous fluid (BSS, SRF) thin air bubble at the pars plana prevents PFO loss while
that may be above the PFO to enter the subretinal space. removing the cannulas. Triamcinolone 4 mgm (Alcon Tries-
Sometimes, a small amount of SRF can become trapped in a ence, preservative free) is then injected into the air bubble
doughnut configuration anterior to the retinal tear and may and the cannulas are removed (Fig. 6.21). Use of triamcino-
be difficult to perceive until the PFO is removed and the lone reduces the foreign body reaction that can occur with
surgeon notices the presence of posterior SRF. To achieve PFO left in the eye for 14 to 16 days until the laser treatment
complete removal of PFO and SRF, the removal of PFO areas are pigmented. Triamcinolone is removed when the
should begin anteriorly, trying to remove any amount of PFO is removed at 14 to 16 days using the 25-gauge, three-
BSS above the PFO meniscus. The tip of the aspiration can- port technique; removal of triamcinolone prevents steroid
nula should slowly be moved to the location of the retinal glaucoma and cataract.
tear, and aspiration should be continued until the PFO-air
meniscus is clearly observed. If any SRF above the PFO is
Gian t Re t in al Bre aks
present, the surgeon can remove it at this time immediately
above the PFO meniscus through the retinal tear. This is David Wong emphasizes that a full fill with PFO with the
critical in giant retinal tear cases to prevent slippage. Once infusion fluid turned off prevents posterior slippage of the
the PFO-air meniscus has been identified and the presence giant break. Exchange of PFO for air to enable AGX or ASX
of BSS or SRF has been ruled out or removed, the surgeon must be done precisely to prevent slippage; the soft-tip can-
can proceed to drain the rest of the PFO at the level of the nula for PFO removal must be positioned above the PFO-
optic nerve head. fluid interface just below the air-fluid interface at all times
so that all BSS (containing SRF) is removed before any PFO.
Obviously the cannula must be slowly advanced posteriorly
Me d ium -Te rm Pe rfl uo ro n
as the exchange is done.
The principal author has used medium-term PFO to repair
selected inferior retinal detachments and giant retinal breaks
Silico n e In fusio n
since PFO became available. The technique allows the patient
to stand, sit, fly, and even work and drive if there is good ASX is preferable to fluid-silicone exchange because air
vision in the other eye. The technique involves completing helps keep silicone out of the anterior chamber. The prin-
the vitrectomy and removing all traction followed by attach- cipal author uses 25-gauge, transconjunctival vitrec-
ing the retina with PFO and then endophotocoagulation sur- tomy in all cases and performs ASX by injecting 1,000
rounding all retinal breaks and suspicious areas. A total PFO cs oil through one cannula with a Alcon cannula while
CHAPTER 6 ■ General Posterior Segm ent Techniques 65

Fig ure 6 .2 2 ■ Following FAX and IDS and usually retinop exy, a silicone injector such as the
Alcon VFC is used to inject silicone oil through a short, thin-wall cannula while air through the
infusion cannula m aintains IOP and air egress occurs with an extrusion cannula behind the lens/
IOL or in the anterior cham ber (A/ C) of aphakic eyes. Injection is stop ped and the infusion line
clam ped with a hem ostat when silicone enters the infusion cannula.

allowing air to escape through the other, open cannula head so that the cannula used for extraction is at the highest
(Fig. 6.22). The air infusion cannula line is clamped when point. Repeated fluid-air-fluid exchanges may be needed to
oil refluxes up the infusion tubing and the oil injection is displace any silicone oil that may be adherent to the zonules.
continued, carefully monitoring the IOP until all the air This technique allows complete removal of all silicone oil
comes out the open cannula in phakic or IOL eyes. Silicone through the 25-gauge cannulas.
oil is filled up to the pupillary plane in aphakic eyes. Air
egress is made possible in phakic eyes and IOL eyes by posi-
Silico n e Re o p e rat io n s
tioning the eye and patient’s head so that the open cannula is
at the highest point, although an air bubble can be removed The principal author has reoperated all silicone cases for
by extrusion with the soft-tip cannula if care is taken to stay EMM or redetachment without removing the oil for over
out of the oil. 25 years. Before 25-gauge vitrectomy was developed, the
author used a short, silicone infusion tubing–cannula device
attached to the Alcon Constellation VFC to infuse silicone oil
Silico n e Re m o val
as SRF is removed. The principal author developed another
Silicone oil should not be removed unless the retina is 100% method that works well with 25-gauge silicone reopera-
attached, all retinal breaks are surrounded by confluent laser, tions, a two-port, sequential, inject-oil-and-remove-SRF
and, preferably, three rows of 360-degree laser have been method using the Alcon 25G silicone injection cannula. The
applied. It is not necessary to remove oil in older patients, techniques utilized for surgery without removing the oil will
especially if an PC is present and silicone does not enter be described in the next section.
the anterior chamber. The authors use 25-gauge, three-port
technique to remove silicone oil by placing a 0.25-inch-
In t e rface Vit re ct o m y
long segment of silicone tubing (fragmenter tubing) on the
Alcon Constellation VFC and placing it over the outside of The principal author developed a concept known as inter-
one of the cannula hubs (Fig. 6.23). The infusion maintains face vitrectomy that is based on performing a variety of tech-
the IOP, and the other cannula is left open to allow addi- niques with air, PFO, or silicone oil in the eye. Vitrectomy,
tional egress of small silicone bubbles. Complete silicone oil forceps membrane peeling, scissors segmentation or delam-
removal is made possible by positioning the eye and patient’s ination, diathermy, and retinectomy all work well with
66 SECTION II ■ Surgical Technology and Techniques

Fig u re 6 .2 3 ■ Silicone oil can


be com pletely rem oved by attach-
ing a 0.25-inch-long segm ent of
silicone tubing to the Alcon Con-
stellation VFC and p lacing it over
the outside of one of the cannula
hubs.

these agents in the eye. The term “interface” was selected to of endophotocoagulation “under” air over three decades
emphasize that the vitreous cutter must be placed outside ago, and Stanley Chang introduced membrane peeling
the air, PFO, or silicone oil bubble. Air and silicone oil float “under” PFO for PVR cases many years ago as well. Inter-
in infusion fluid but PFO sinks; it is crucial to understand face vitrectomy prevents an increase of SRF during traction
that there is always a fluid layer at the retinal surface and removal, allows realistic assessment of remaining traction,
the retina is over 90% water as well as immiscible in air, confines bleeding to the interface, and stabilizes the retina.
PFO, or oil. The principal author introduced the concept Air stabilizes the retina by spring dampening (Fig. 6.24),

Fig ure 6 .2 4 ■ Air stabilizes the


retina by spring dam pening.
CHAPTER 6 ■ General Posterior Segm ent Techniques 67

Fig u re 6 . 2 5 ■ Silico n e o il
stab ilizes th e retin a b y viscous
dam pening.

silicone oil by viscous dampening (Fig. 6.25), and PFO by and ERM removal without plugging the vitreous cutter by
inertial (F = MA) and gravitational (high specific gravity) using 600–650mm Hg vacuum.
effects (Fig. 6.26). The author uses 25-gauge vitrectomy for Prior to the development of FAX by the senior author
these cases and is very careful to avoid applying vacuum (30), fluid was withdrawn through a single needle, which was
while the port is in silicone oil. Moderate amounts of subreti- then used to inject gas into the collapsed eye after turning a
nal silicone can be removed in conjunction with retinectomy stopcock. AGX is usually best performed at the end of the case,

Fig u re 6 .2 6 ■ PFO stab ilizes


th e retin a b y in ertial (F = MA)
an d g ravitation al (h ig h sp ecific
gravity) effects.
68 SECTION II ■ Surgical Technology and Techniques

Fig u re 6 .2 7 ■ Forcep s m em -
brane peeling, scissors segm enta-
tion or delam ination, retinectom y,
laser, etc. can be perform ed with-
out rem oving silicone oil.

after FAX and retinopexy. Similarly, most surgeons perform The air-vitreous interface facilitates visualization of
FAX and retinopexy before ASX. Some surgeons recommend residual vitreous after an exchange is performed. The senior
fluid-silicone exchange and omit the FAX step. The fluid- author often places the vitreous cutter port just posterior to
silicone exchange approach can make visualization as well as the air-vitreous interface and performs additional vitrectomy
complete reattachment for retinopexy more difficult. under air (Fig. 6.28). This can be done with PFO and sili-
Fluid-perfluorocarbon exchange is preferred to FAX cone oil as well, which is termed “interface vitrectomy.”
before perfluorocarbon placement in retinal detachment, Forceps membrane peeling, scissors segmentation and
PVR, and giant break cases. PGX or PSX is usually performed delamination, subretinal surgery, retinectomy, laser endopho-
after laser retinopexy. tocoagulation, and foreign body removal can all be performed
The authors never remove silicone oil when perform- under air if it is not an aphakic eye with striate keratopathy.
ing reoperations as forceps membrane peeling, scissors Fogging of IOLs if a YAG capsulotomy has been performed
segmentation or delamination, retinectomy, laser, etc. are is a serious limitation of this method. The authors used to
effective “under” silicone (Fig. 6.27). Silicone is infused to refrigerate the BSS Plus prior to all vitrectomies except those
replace silicone lost through the sclerotomies. The suction with IOLs because hypothermia reduces inflammation, isch-
(vacuum) is used at the highest setting (600–650mm Hg), emic damage, endothelial damage, iris bleeding, and light
and scissors segmentation or delamination, forceps mem- toxicity, but low fluid volumes associated with current vitrec-
brane peeling, retinectomy, subretinal surgery, and endo- tomy technology dramatically reduce the advantages. Silicone
photocoagulation work in the usual manner. lenses fog much more than acrylic or PMMA lenses because
they have a higher thermal inertia. Prior YAG capsulectomy
and intraoperative removal of the AVC create the fogging
Vit re ct o m y Un d e r Air
problem by allowing air access to the IOL surface. IOLs fog
Certain portions of the vitreous surgery procedure can because they have significant thermal inertia and are cooled
be completed after FAX (31). At times, continuous severe by room temperature infusion fluid, and the air in the eye
bleeding cannot be controlled with combined extrusion and is saturated with water vapor. Viscoelastics can be injected
bipolar diathermy or endophotocoagulation. If internal FAX against the posterior surface of the lens to reduce fogging, but
is performed in such an instance, the bubble will confine this creates a morphed image, increases cost, and increases
the blood to a small space so that overall visualization is silicone emulsification. The authors use a soft-tip cannula as
improved and diathermy or endophotocoagulation of the a windshield wiper until the procedure is completed or the
vessel can be completed. The air-vitreous interface is quite fogging abates. PFC liquids such as PFO completely elimi-
visible, which facilitates the removal of residual vitreous by nate this problem by facilitating removal of SRF and perform-
placing the cutter port barely into the vitreous. ing endophotocoagulation before PFO-gas exchange.
CHAPTER 6 ■ General Posterior Segm ent Techniques 69

Fig ure 6 .2 8 ■ Th e air-vitreous


in terface facilitates visualization
and rem oval of resid ual vitreous
after an exchange is perform ed.

Po st o p e rat ive Po sit io n in g The reduced atmospheric pressures intrinsic to air


travel or rapid ground travel to higher altitudes dictate
Postoperative positioning requirements will depend on the
marked caution when a bubble is present. Bubble expansion
location of the tears being treated and lens status. A phakic
from air travel causes a severe increase in IOP and can cause
patient can develop a gas cataract from contact of the gas to
vascular occlusion (32,33). Air travel is best prohibited while
the posterior lens surface. While this cataract can be transi-
an air or gas bubble is present, although clinically it has been
tory, once it persists for longer than 48 hours, it may become
observed that a bubble less than 10% of ocular volume can
permanent. The first requirement is that the location of the
be tolerated. Ground travel should be used to prevent this
treated tear is at the highest position in the eye to allow con-
severe problem. Patients should be cautioned to avoid rapid
tact with the gas. Once the laser retinopexy becomes pig-
ground travel from low to high altitudes.
mented, further head positioning is not required to keep the
retina attached. Once this occurs on a pseudophakic patient,
strict head positioning can be relieved. If the patient is pha- Sub re t in al Fluid Re m o val
kic, forward head tilt (depending on the remaining size of
Indica tions for Dra ina ge of Subretina l Fluid
the bubble) is required to keep the gas away from the poste-
rior lens surface. The debate over the necessity of drainage of SRF in sclera
Making an analogy to a “cast on a broken arm” can buckling has raged for years and will not be settled by this
enhance patient acceptance. Phrases such as “put the bub- book. Drainage of SRF is essential in difficult retinal detach-
ble on the trouble” and “longer duration bubbles result in ment cases because it instantaneously determines the need
higher success rates” help the patient gain understand- for subsequent surgical steps. It is not always possible for
ing. Numerous pillows facilitate comfortable positioning. even the most experienced surgeons to correctly determine
A comparison can be made to sleeping with crossed arms the exact steps to accomplish retinal reattachment from the
on a school desk and a table at home used with a pil- preoperative examination. The principal author refers to SRF
low for padding. Wingback chairs and sofas are similar drainage with FAX as the “reattachment experiment” because
to the car, bus, or airplane seating position and can be it frequently helps determine the need for further vitreous
used to explain the required position. Some surgeons removal, peeling, segmentation, delamination, retinectomy,
recommend expensive special chairs or support systems, or scleral buckling.
which the authors believe to be unnecessary in most Removal of virtually all SRF before fluid-air-gas or
cases. These devices require immobility, which increases fluid-air-silicone exchange increases the likelihood of injec-
muscle cramping, psychological stress, and deep venous tion of the correct volume of isoexpansive gas mixtures
thrombosis. or silicone oil. It is virtually impossible for the surgeon to
70 SECTION II ■ Surgical Technology and Techniques

accurately estimate the volume of the subretinal space or of retina in the sclera, prevents choroidal bleeding, reduces
vitreous cavity. Expanding gas mixtures should not be used RPE damage, and is the technique that should be used dur-
with total fluid-gas exchange because of the risk of central ing pars plana vitrectomy.
retinal artery occlusion due to high pressure. Similarly, if the
volume of isoexpansive gas or silicone oil required is over-
estimated, central retinal artery occlusion can result. If the Direct Needle Tra nssclera l Dra ina ge of Subretina l
volume of gas or oil is underestimated, the area of contact Fluid for Sclera l Buckling
with the surface tension management agent may be insuf- The senior author developed direct (transscleral) needle
ficient, causing the procedure to fail. drainage of SRF to reduce the complications of the scleral
The principal author introduced the concept of internal cutdown method during scleral buckling. Direct needle
(transretinal) drainage of SRF and postreattachment retin- drainage has been shown in a randomized trail (Cairns) to
opexy in 1973. Prior to that time, retinopexy was performed eliminate the problem of retinal incarceration in the drain
before the vitrectomy and expanding gas bubbles were used site (>3% with the scleral cutdown method), allow more
to slowly reattach the retina postoperatively. Postreattach- complete drainage, and reduce hypotony, without adding
ment retinopexy probably reduces the dispersion of retinal any complications. Direct needle drainage of SRF can be
and RPE cells, potentially reducing PVR recurrences. Postre- performed through the conjunctiva, a buckle, or a choroi-
attachment retinopexy allows accurately placed, controlled dal detachment (edema). This method is performed using a
intensity retinopexy to the retina as well as the RPE. Over- 25- to 27-gauge, 0.5-inch, disposable needle on a tubercu-
treatment is a significant factor in PVR recurrences and fibrin lin syringe with the plunger removed. A transparent needle
syndrome. hub is preferred because it allows early visualization of fluid
Internal drainage methods and exchanges allow the egress (34). A site is selected that has the highest retinal
benefits of postreattachment retinopexy; accurate air, gas, detachment by viewing through the operating microscope
and silicone volumes; and the reattachment experiment to using the endoilluminator and contact lens. Indirect oph-
be used on a routine basis. thalmoscopy can be used if an operating microscope is not
being utilized. The bevel of the needle is always oriented
away from the retina to prevent retinal entrapment. The tip
Externa l Versus Interna l Dra ina ge
of the needle is kept away from the sclera by intentional
External drainage of SRF requires an opening through the “shanking” until the entry point is located. The needle is then
sclera, choroid, and RPE. By contrast, internal or transretinal oriented in a more tangential manner and advanced slowly
drainage requires no eye wall incision. Internal drainage through the sclera until a slight proprioceptive “pop” is felt
enables complete drainage of the SRF, prevents incarceration similar to venipuncture (Fig. 6.29). At this point, a brown

Fig u re 6 .2 9 ■ Direct n eed le


d rain ag e of SRF is in itiated b y
inserting 25- to 27-gauge, 0.5-inch
needle (attached to a 1-m L syringe
with p lung er rem oved ) throug h
the sclera slightly anterior to the
g reatest h eig h t of th e retin al
d etach m en t. A p ro p rio cep tive
“pop” will be felt when the needle
is through the sclera.
CHAPTER 6 ■ General Posterior Segm ent Techniques 71

Fig u re 6 .3 0 ■ Th e n eed le is
ad van ced th roug h the ch oroid
an d RPE un til it can b e seen
under the retina. A brown “pencil
p oint” of RPE tissue can be seen
just before the RPE is penetrated.
Rap id , sm all-am p litu d e m o ve-
m ent of the retina and shifting of
SRF toward the n eed le ind icate
that flow is occurring.

pencil point–like structure may be seen in the subretinal of the retina will indicate fluid egress. The detachment may
space, which is the RPE stretched over the needle tip. The increase near the drainage site as the pseudoplastic mass
needle is then advanced slightly under direct visualization of SRF shifts toward the egress lumen. The needle should
through the RPE until the silvery sheen of the needle is seen be angulated slightly until it is tangential to the retina after
under the retina (Fig. 6.30). At this point, the retina begins to the initial placement. The needle should be held immobile
tremble due to turbulence in the needle lumen. The needle until all the SRF is gone (Fig. 6.31). Pigment typically comes
cannot be visualized if the SRF is turbid, but the trembling through the needle hub as the last fluid drains.

Fig u re 6 . 3 1 ■ Th e n e e d le
sh ould b e h eld im m ob ile un til
all SRF is d rained and the retina
drapes over the needle; pigm ent
often ap p ears in SRF in the clear
n eed le h u b wh en d rain ag e is
com p leted.
72 SECTION II ■ Surgical Technology and Techniques

ENDODIATHERMY Lase r En d o p h o t o co ag ulat io n


Xenon arc light energy can be delivered to the retinal surface
Although neovascularization (NVE) is frequently encountered with the endophotocoagulator (37). The prototype was
in diabetic vitrectomy, intraoperative bleeding is seldom of an adapter to the Zeiss xenon photocoagulator. The first
great consequence. Because endocoagulation can cause tis- commercial endophotocoagulator was an adapter to the
sue shrinkage, retinal breaks, nerve fiber layer damage, and Clinitex Log III portable xenon arc photocoagulator (38).
optic atrophy, it should be reserved for large vessels that A xenon endophotocoagulator has high beam divergence
are to be transected or that actually bleed during surgery. and reliability problems and is no longer used by most
When extensive networks of NVE are associated with ERM, surgeons.
it is best to control bleeding during the dissection by using Laser endophotocoagulation was developed later
transient elevation of IOP. Vascular attachment points can (Peyman, Fleischman, Charles, and Landers; all indepen-
then be treated as needed with endocoagulation. Frequently, dently and simultaneously) and is preferable to xenon
vascularized ERM has only a few connections to the retinal because of less beam divergence, better reliability, and more
arterial system, which are only recognized and treated after rapid firing. Virtually all compact, laser photocoagulator sys-
delamination of the ERM. Use of preoperative bevacizumab tems can be utilized for endophotocoagulation.
in proliferative retinopathies has also decreased the problem Near IR (infrared) diode lasers (Tano) are 20% to 30%
of intraoperative hemorrhage. efficient in converting electrical to light energy in contrast to
Every attempt should be made to prevent and control ion lasers (argon) at 0.0001% or less. For this reason, they
bleeding; blood can form a substrate and stimulus for post- use standard, single-phase electrical power instead of high-
operative cellular proliferation. As it is not always possible current, three-phase power. Because of their efficiency, they
to safely control bleeding, it is fortunate that the aphakic, are far more reliable, are more compact, and do not require
vitrectomized eye rapidly clears postoperative hemorrhage. internal or external water cooling. The near IR wavelength
Elevated new vessels are best coagulated with diathermy. is the major disadvantage of these lasers because it is much
Unipolar diathermy, which was used originally, has been more difficult to judge the correct power threshold and the
supplanted by bipolar diathermy because of the latter’s lesion is deeper.
greater safety (35,36). If unipolar diathermy is applied near Diode-pumped frequency up-converted, continuous,
the optic nerve, radio frequency energy can travel through 532-nm lasers are ideal for all operating room and most
the optic nerve, causing optic atrophy and permanent blind- office photocoagulation. They have all the above described
ness. Unimanual bipolar diathermy (UBD) functions well if advantages of diode laser but use an ideal wavelength for
care is taken to maintain at least 1 mm distance from the hemoglobin absorption and xanthophyll avoidance.
optic nerve. By controlling bleeding with transient increases The endophotocoagulator is used primarily for the
in IOP, UBD can be very effective in treating vascular attach- treatment of bleeding from surface NVE, for retinopexy, and
ment points. for PRP. Endophotocoagulation should be used for surface
Bimanual bipolar diathermy (BBD) (37) can function bleeding from specific sites, usually after scissors delamina-
through any two intraocular instruments or one intraocular tion of ERMs. When using the endophotocoagulator, the
and one extraocular instrument by the simple attachment of endoilluminator is usually held in the opposite hand to pro-
unshielded wires and clips. Insulated microclips or connec- vide diffuse illumination. 20-gauge illuminating laser probes
tors on the instrument handles can be used for BBD (Charles). (Chang) were developed to solve this problem. When there
BBD clips are ideal for 25-gauge surgery. This method has is acute bleeding, it is recommended that the suction can-
been virtually supplanted by the disposable bipolar endoil- nula or vitreous cutter be alternated with the laser probe
luminator for 20-gauge surgery, which combines diathermy to remove blood and facilitate precise coagulation. The
with the endoilluminator. This device is ideal for coagulat- 20-gauge aspirating endophotocoagulator probe (Chang)
ing vascular attachment points during delamination and seg- can be used to aspirate SRF during endophotocoagulation as
mentation of ERM. The disposable bipolar endoilluminator reported by the principal author.
is typically utilized in conjunction with the vitreous cutter The endophotocoagulator is well suited for PRP con-
or fine curved scissors. It is not necessary to interrupt the comitant with vitreous surgery for diabetic retinopathy,
procedure in order to place a specialized instrument in the venous occlusive disease, hemoglobinopathies, and telangi-
eye, and bleeding can be washed away continuously using ectasia. As discussed in the chapter on diabetic retinopathy,
the cutter to permit excellent visualization of the bleeding endo-PRP can bring about a decreased incidence of neovas-
vessel during dissection. By intentionally allowing the IOP cular glaucoma, AVC fibrovascular proliferation, and recur-
to become very low, bleeding can be stimulated so that its rent flat NVE leading to postoperative bleeding. The only
source can be identified and coagulated. Although bipolar limitation of endophotocoagulation is that elevated retina
diathermy can be used for retinopexy, there is little effect cannot be treated.
at the RPE level. Endophotocoagulation is better suited to In contrast to endocryopexy, endophotocoagulation is
retinopexy than is endodiathermy. a noncontact method. Therefore, dispersion of RPE cells,
CHAPTER 6 ■ General Posterior Segm ent Techniques 73

choroidal bleeding, retinal tears, and the increased wound choroidal hemorrhage due to the combination of engorged
size associated with endocryopexy are avoided. Endophoto- choroidal vessels. Postreattachment retinopexy will ensure
coagulation lesions are between 600 and 1,000 mm in diam- better visualization, so that all breaks can be identified and
eter, depending on the distance from the tip of the probe iatrogenic breaks can be treated. Completion of internal
to the retinal surface, the beam divergence, and the power fluid-gas exchange and IDS will confine any RPE cells mobi-
setting. If the retina is detached, endophotocoagulation lized by retinopexy to the area of the break and possibly
must be preceded by FAX and IDS, which bring the retina decrease the incidence of PVR.
and RPE into contact to permit energy absorption. For focal Transscleral cryopexy in vitrectomy has long been vir-
treatment of retinal breaks, the continuous mode is utilized tually replaced by laser endophotocoagulation and laser
to treat in a confluent manner (painting) around the breaks. indirect ophthalmoscope (LIO). Cryopexy disperses live
This technique minimizes the possibility of undertreat- RPE and glial cells and causes more inflammation and PVR.
ment or overtreatment, which frequently occurs with the Endocryopexy is an unsafe, contact-based method that
placement of discrete photocoagulation spots in rows. Peri- requires an enlarged opening and is fortunately no longer
odically during treatment, small amounts of SRF will shift used by most surgeons. This method causes increased cel-
posteriorly, making repeated internal drainage necessary to lular proliferation and choroidal hemorrhages. As discussed
permit retinopexy. previously, endophotocoagulation now performs the tasks
The endophotocoagulator should never be utilized in formerly handled by endocryopexy. Endophotocoagulation
air (gas) when there is blood on its tip, or damage to the is used far more commonly than transscleral retinopexy.
probe may result. PRP under air to areas of retina that had Transscleral retinopexy is used primarily for scleral buckling
been detached before surgery is a common cause of fibrin without vitrectomy.
syndrome because of overtreatment.
A microscope filter attenuates the laser energy in the
Tran sscle ral Diat h e rm y
optic path of the operating microscope during endophotoco-
agulation and ensures continued dark adaptation and safety. Transscleral diathermy probably creates less proliferation
The fovea and optic nerve must be avoided, and all treat- than cryopexy but causes more scleral damage. It can be used
ment must be initiated at low energy with stepwise increases with fiberoptic transillumination to find and treat any breaks,
until the desired effect is achieved. except in the macular and peripapillary areas. The small
The endophotocoagulator can be used to dilate the probe fits under buckles more easily than a cryoprobe.
pupil by treating the posterior surface or margin of the iris Because of the thermal and electrical insulating proper-
sphincter. Endophotocorepexy is of special use when iris ties of an air (gas) bubble, diathermy causes larger retinal
NVE prohibits sphincterotomy with the vitrectomy lesions and less scleral damage in an air (gas)-filled eye than
instrument. in a fluid-filled eye. Endophotocoagulation is preferred in
The ciliary processes can be endophotocoagulated in virtually all instances.
glaucoma cases, with subsequent decrease in pressure. The
senior author first reported endocyclophotocoagulation but
is concerned that it is significantly overutilized by cataract
surgeons in conjunction with phacoemulsification. Many
INTRAOPERATIVE FLUORESCEIN
glaucoma experts share this concern based on the observa- ANGIOSCOPY
tion that the apparent benefits are short term and postopera-
tive CME increases. Intravenous sodium fluorescein dye (3.0 mL of 25%) can be
administered during vitreous surgery after the retina is visu-
alized (31). The dye can then be made to fluoresce by put-
ting a fluorescent interference-type exciter filter in the path
RETINOPEXY of the endoilluminator light source. The sources of bleeding,
especially in reoperation, can be readily identified.
All retinal breaks, except macular holes, peripapillary
breaks, and retinotomies for submacular surgery, should be
In je ct io n o f An t ib io t ics an d St e ro id s
treated with some form of retinopexy. This policy is neces-
sary because of the impossibility of predicting which retinal Subconjunctival antibodies should be injected in all patients
break will result in detachment, and it is justified because of at the end of surgery. A 30- or 32-gauge needle can be used
the relative safety of retinopexy. to inject the antibiotics in the inferior cul-de-sac. This tech-
Although it may be contrary to the tendency of the nique has low risk of sclera perforation. Antibiotics effective
scleral buckling surgeon, retinopexy should be used only against Gram-positive penicillinase producers and Gram-
after vitrectomy, surgical dissection, IDS, FAX, and comple- negative organisms should be used (the authors currently use
tion of SRF drainage have reattached the retina. Performed a combination of cefazolin and tobramycin and replace the
at the beginning of the operation, retinopexy may cause cefazolin with vancomycin in penicillin-allergic patients).
74 SECTION II ■ Surgical Technology and Techniques

This use of antibiotics is necessary with the extensive


SUMMARY
amount of tubing connections, instrumentation, and irrigat-
ing fluid utilized in vitreous surgery. It is thought, however,
This chapter provides a discussion of the basic building
that the myelinolytic effect of aminoglycoside antibiotics
blocks of successful posterior vitrectomy. Each step must be
may contribute to an occasional transient neuritis-like facial
understood, and the proper equipment must be available
pain after surgery. This lasts several weeks, can be managed
so that complications or unexpected findings can be man-
medically, and always disappears. Most “scleral buckle pain”
aged safely, rapidly, and effectively. Even the infrequent
is probably caused by this mechanism.
vitreous surgeon who tries to limit himself or herself to
A subconjunctival steroid (triamcinolone acetonide
“easier” cases can find the unexpected and is obliged to use
20 mg in 0.5 mL or dexamethasone 4 mg) should be injected
these “advanced” techniques to safely complete the surgery.
in every case unless the patient is an infant, has a macular
Implicit in understanding the building blocks is using them
hole, or is known to be a steroid glaucoma responder. Perio-
in the appropriate sequence.
cular injections are more effective than systemic or topical
steroids and are medically safer than systemic steroids. Intra-
vitreal steroids can be used in conjunction with vitrectomy.
References
The duration of action is shorter (~1 month) than in the
nonvitrectomized eye (3 months). There is a 30% to 50% 1. Edelhauser HF, Hyndiuk RA, Zeeb A, Schultz RO. Cor-
risk of steroid glaucoma and a significant cataract risk, and neal edema and the use of epinephrine. Am J Ophthalmol
1982;93(3):327–333.
nonclosure of macular holes has been reported. 2. Kunimoto DY, Kaiser RS, Wills Eye Retina Service. Incidence
of endophthalmitis after 20- and 25-gauge vitrectomy. Oph-
thalmology 2007;114:2133–2137. [Epub 2007 Oct 4.]
BUCKLING ANTICIPATED OR 3. Martidis A, Chang TS. Sutureless 25-gauge vitrectomy: risky or
PREVIOUS BUCKLE PRESENT rewarding? Ophthalmology 2007;114:2131–2132.
4. Rizzo S, Genovesi-Ebert F, Vento A, et al. Modified incision in
25-gauge vitrectomy in the creation of a tunneled airtight scle-
The operating microscope is useful for all scleral buckling rotomy: an ultrabiomicroscopic study. Graefes Arch Clin Exp
procedures, even if vitrectomy is not planned. The authors do Ophthalmol 2007;245:1281–1288. [Epub 2007 Feb 21.]
not currently perform vitrectomy buckles; primary vitrectomy 5. Mason JO III, Yunker JJ, Vail RS, et al. Incidence of endophthal-
produces better outcomes with less complications and shorter mitis following 20-gauge and 25-gauge vitrectomy. Retina
2008;Jul 28.
operating times. If, on the other hand, a decision is made to 6. López-Guajardo L, Vleming-Pinilla E, Pareja-Esteban J, Teus-
perform combined vitrectomy-buckle, conjunctival incision Guezala MA. Ultrasound biomicroscopy study of direct and
should be performed to expose the sclera as explained in the oblique 25-gauge vitrectomy sclerotomies. Am J Ophthalmol
sclera buckling chapter. Better visualization reduces inadver- 2007;143:881–883.
tent scleral perforation and damage to the muscles and ocular 7. Inoue M, Shinoda K, Shinoda H, et al. Two-step oblique inci-
sion during 25-gauge vitrectomy reduces incidence of postop-
blood supply and improves conjunctival closure and patient erative hypotony. Clin Exp Ophthalmol 2007;35:693–696.
comfort. Practice with the microscope improves surgical skills. 8. Asheesh T, Shah GK, Fang A. Visual outcomes with 23-gauge
If an existing buckle is to be revised or removed, it should be transconjunctival sutureless vitrectomy. Retina 2008;28:258–262.
exposed under the microscope by dissection with a hockey 9. Gupta OP, Ho AC, Kaiser PK, et al. Short-term out-
stick–type blade. A small suction cannula can be used to aspi- comes of 23-gauge pars plana vitrectomy. Am J Ophthalmol
2008;146:193–197. [Epub 2008 Jun 10.]
rate routine bleeding, which usually subsides with minimal 10. Chen E. 25-gauge transconjunctival sutureless vitrectomy. Curr
diathermy. Bipolar diathermy coagulation should be used Opin Ophthalmol 2007;18:188–193.
only for major bleeding as these techniques increase conjunc- 11. Landers MB. Removal of blood from the retinal surface in pars
tival and episcleral scarring if used excessively. Bipolar forceps plana vitrectomy. Am J Ophthalmol 1978;86:427.
produce more focused diathermy than the “eraser.” 12. Abrams GW, Williams GA. “En bloc” excision of diabetic mem-
branes. Am J Ophthalmol 1987;103(3):302–308.
Excellent exposure can be obtained by using a microret- 13. Charles S (Developer). Suction forceps membrane peeling.
inal retractor without a slot. The slot on the Schepens retrac- Presented at Wilmer Vitrectomy Course, The Johns Hopkins
tor allows prolapse of orbital tissues into the surgical field. School of Medicine, Baltimore, May 1976.
The assistant’s hand is kept out of the surgical field because of 14. Charles S (Developer). Chopsticks membrane peeling. Pre-
the right angulated handle and can exert a lateral pull rather sented at Wilmer Vitrectomy Course, The Johns Hopkins
School of Medicine, Baltimore, May 1976.
than a twisting force, resulting in reduced hand fatigue. If 15. Wang CT, Charles S. Microsurgical instrumentation for vitrec-
scleral buckling elements are to be removed, it is best to tomy, Part 1. J Clin Eng 1983;8(4):321.
place the infusion cannula, verify placement through the 16. O’Malley C (Developer). Extrusion method. Ocutome Frag-
microscope, and turn on the infusion before buckle removal matome Newsletter 1978;3(1):3.
to prevent hypotony. Care must be taken when rotating the 17. Fineberg E, Machemer R, Sullivan P. SF6 for retinal detach-
ment surgery, a preliminary report. Mod Probl Ophthalmol
eye temporally and inferiorly so as not to rotate the interior 1974;12:173.
portion of the infusion cannula posteriorly into the periph- 18. Fineberg E, Machemer R, Sullivan P, et al. Sulfur hexafluoride
eral retina when the cannula contacts the lid. in owl monkey vitreous cavity. Am J Ophthalmol 1975;79:67.
CHAPTER 6 ■ General Posterior Segm ent Techniques 75

19. Machemer R. Intravitreal injection of sulfur hexafluoride gas 29. Lincoff A, Lincoff H, Iwamoto T, et al. Perfluoro-n-butane.
(SF6). In: Freeman HM, Hirose T, Schepens CL, eds. Vitreous A gas for a maximum duration retinal tamponade. Arch Oph-
surgery and advances in fundus diagnosis and treatment. New thalmol 1983;101:460.
York: Appleton-Century-Crofts, 1977:421–422. 30. Aaberg TM, Abrams GW, Edelhauser HF. Intraocular sulfur
20. Bourgeois JE, Machemer R. Results of sulfur hexafluoride gas hexafluoride: experimental and clinical correlation. In: Inter-
in vitreous surgery. Am J Ophthalmol 1983;96:405. national Symposium on New and Controversial Aspects of
21. Killey F, Edelhauser H, Aaberg TM. The effects of intraocular Vitreoretinal Surgery, Texas Medical Center, Houston, Texas,
sulfur hexafluoride and freon gas on intraocular pressure and St. Louis: C.V. Mosby, 1977:393–397.
vitreous volume. Arch Ophthalmol 1978;96:521. 31. S. Charles. unpublished data, 1974.
22. Miller B, Lean JS, Miller H, Ryan SJ. Intravitreal expanding gas 32. Fuller D. Flying and intraocular gas bubbles (letter). Am J
bubble: a morphologic study in the rabbit eye. Arch Ophthalmol Ophthalmol 1981;91:276.
1984;102:1708. 33. Dieckert JP, O’Connor PS, Schacklett DE, et al. The effects of
23. Chang S, Lincoff H, Coleman J, et al. Perfluorocarbon gases in air travel on eyes with intraocular gas. Presented at the Annual
vitreous surgery. Ophthalmology 1985;92:651. Meeting, American Academy of Ophthalmology, Atlanta, GA,
24. Lincoff A, Haft D, Liggett P, Reifer C. Intravitreal expansion of October 2, 1985.
perfluorocarbon bubbles. Arch Ophthalmol 1980;98(9):1646. 34. Charles S. Controlled drainage of subretinal and choroidal
25. Lincoff A, Kreissig I. Intravitreal behavior of perfluorocarbons. fluid. Retina 1985;5(4):233.
Surv Ophthalmol 1981;2:17. 35. Machemer R. Letter to the editor. Am J Ophthalmol 1977;
26. Lincoff H, Coleman J, Kreissig J, et al. The perfluorocarbon 83:282.
gases in the treatment of retinal detachment. Am Acad Ophthal- 36. Schepens C. Letter to the editor. Am J Ophthalmol 1978;
mol 1983;90:546. 85:574.
27. Lincoff H, Kressig J. Intravitreal behaviour of perfluorocarbons. 37. Charles S, White J, Dennison C, Eichenbaum D. Bipolar biman-
Dev Ophthalmol 1981;2:17. ual intraocular diathermy. Am J Ophthalmol 1976;81:101.
28. Lincoff H, Mardirossian J, Lincoff A, et al. Intravitreal longevity of 38. O’Malley P. Portable xenon arc light coagulator. Br J Ophthalmol
three perfluorocarbon gases. Arch Ophthalmol 1980;98:1610. 1973;57(12).
C H AP TER
7
VITRECTOMY TECHNIQUES AND
TECHNOLOGY FOR ANTERIOR
SEGMENT PROBLEMS

Although developed for posterior vitreoretinal incision for a one-incision technique (Chapter 2). This
surgery, the vitrectomy instrument has wide- method is easy to use but results in a return to the turbulent
spread application to anterior segment surgery fluid flow observed with full function probes (Fig. 7.1). Use
(1–8). Every anterior segment microsurgeon of the infusion sleeve should be limited to vitrectomy in soft
must have vitrectomy techniques and equipment at his or her eyes with no sideport, a rare situation.
fingertips to manage vitreous loss during cataract surgery and A bimanual approach (10) with a blunt 30-degree bent
other common problems. The anterior segment microsurgeon infusion cannula as the infusion device is used if the pars
with extensive knowledge of corneal physiology, aqueous plana cannot be seen, as well as for retinopathy of prema-
dynamics, intraocular lenses (IOLs), cataract complications, turity (ROP) (5). This technique allows the independently
and microsurgical techniques need not rely on a posterior movable infusion to hydrate and mobilize the lens material
segment surgeon to manage all anterior segment vitreous for aspiration. By using 25-gauge infusion, the infusion nee-
problems. Similarly, an anterior segment surgeon should not dle can be interchanged with a cutter to provide better access
delve into complex vitreoretinal problems just because the to the posterior chamber and periphery.
technology is available. The demands are so complex that it is The authors encourage anterior segment surgeons to
a very unique individual who is an expert in both arenas. Per- develop bimanual surgery dexterity by using bimanual irri-
haps the most important issue to convey to anterior segment gation and aspiration techniques for lens cortex removal.
surgeons is that proper management of vitreous is critical for The surgeon can use the bimanual I/A irrigation tip during
the prevention of postoperative retinal detachments. anterior vitrectomy as well. If this instrument is used, the
decrease in caliber (and irrigation flow capacity) should be
compensated by decreasing the aspiration flow limit of the
VITRECTOMY INSTRUMENT CHOICE vitrectomy system, otherwise collapse of the anterior cham-
ber can occur during surgery.
The large size and fluid flow characteristics of full function Unsupported butterfly needles or silicone tubing should
probes make them as inappropriate for anterior segment not be used for anterior chamber infusion because they may
surgery as they are for posterior segment surgery. Dispos- damage the endothelium and offer no help in accessing the
able, small, lightweight, pneumatic, 23- or 25-gauge cutters, posterior chamber for manipulation of cortex. Sew-on cor-
high-speed cutting, and fast proportional (linear) suction neal infusion cannulas do not permit changing the active
control are a necessity for anterior segment surgery, just as instrument from one hand to the other and can cause cor-
they are for vitreoretinal procedures (Chapter 2). neal damage via mechanical trauma or fluid directed against
endothelium.

INFUSION SYSTEMS FOR ANTERIOR


VITRECTOMY ULTRASONIC FRAGMENTATION

Infusion can be provided by the infusion sleeve (9), which Aspirating ultrasonic fragmenters such as the Alcon Accurus
slips over the vitrectomy instrument through a limbal four-crystal device and the Constellation permit 20-gauge
76
CHAPTER 7 ■ Vitrectom y Techniques and Technology for Anterior Segm ent Problem s 77

Fig u re 7 .1 ■ In fusion sleeves


in crease in fu sio n flu id tu rb u -
lence, resulting in endothelial cell
dam age.

incisions to be used for relatively dense cataracts (11,12). venturi-based vacuum systems are far superior to the other
The 20-gauge aspirating fragmenters require a separate approaches because they reduce vitreoretinal traction and
infusion cannula identical to modern vitrectomy systems. ocular collapse. A vitrectomy system with proportional
The phacoemulsifier is analogous to the full function vitrec- (linear) suction control of a venturi source offers the most
tomy approach and similarly requires a larger incision and precise control over suction level currently available.
creates more turbulence. Leaving the fragmenter lumen open to the atmosphere
Current 25- and 23-gauge vitrectomy systems lack a (incorrectly called “passive egress”) is inappropriate because
fragmenter in both gauge diameters. The only available frag- precision, foot control, and reflux capability are lost. The tub-
menter at the time of this writing is 20 gauge. On the other ing can be squeezed to create reflux if the iris or retina becomes
hand, modern fluidics in both 25- and 23-gauge systems impacted in the port. Certain vitrectomy systems (Accurus
allow removal of soft nuclei with the vitrectomy probe alone. and Constellation Vision System) offer foot pedal–controlled
Only harder nuclei require the creation of a 20-gauge wound reflux. Foot pedal control is also available with the vitrectomy
to introduce the fragmenter. mode of phaco systems such as the Alcon Legacy and Infinity.
Modern phaco technique(s) utilizing high suction and
In fusio n So urce s limited flow rates are not used with current fragmenters
because of the 20-gauge needle configuration.
Infusion can be accomplished with the same options used
with the vitrectomy instrument: bimanual, with 20-gauge,
30-degree bent, blunt cannula, infusion sleeve, sew-on infu- In t e rm it t e n t Ve rsus Co n t in uo us
sion cannula or 25-gauge trocar-cannula system. So n ifi cat io n an d Asp irat io n
The metal infusion sleeve, which has the potential of
Bursts of intermittent sonification alternated with aspira-
dampening the sonic energy, is not intended for use with
tion have been recommended for fragmenter use, but they
the fragmenter. Bimanual infusion with the angulated, blunt
prolong the procedure and cause unnecessary heating of the
cannula can be kept away from the suction port, decreasing
sclera (13). Aspiration without sonification is ineffective and
turbulence, and may be used for selective mobilization and
time consuming. Luminal fluid flow is the most effective
hydration of lens material in certain cases.
means of dissipating frictional heat generated by the frag-
menter in the wound. Squirting fluid on the needle shank
Vacuum So urce s
externally does not cool the internal scleral wound. By com-
Syringes, peristaltic pumps, or venturi-based systems can bining continuous sonification with continuous aspiration, a
provide suction for the vitrectomy probe or aspirating safe and rapid lensectomy can be performed using the con-
fragmenter. For the reasons discussed in Chapter 2, tinual luminal fluid flow for scleral cooling.
78 SECTION II ■ Surgical Technology and Techniques

Lim b al Ve rsus Pars Plan a Ap p ro ach the anterior segment; VEGF causes neovascularization of the
fo r An t e rio r Vit re ct o m y iris and trabecular meshwork. For this theoretical reason and
to reduce posterior subcapsular cataract rates, the anterior
Elective anterior vitrectomy during routine cataract removal
vitreous cortex should be avoided in diabetic vitrectomies
should be reserved only for selected pediatric cataracts or
in which the lens is left in place, unless it is opaque. The
for those cases in which the anterior vitreous cortex and an
indications for elective clear lens and anterior vitreous cortex
opaque posterior capsule resistant to polishing are firmly
removal without IOL implantation in the diabetic patient will
adherent. Complete posterior capsulectomy from the lim-
be explained in a subsequent chapter.
bal approach almost always results in anterior vitrectomy
because of the approach angle and posterior displacement of
De ve lo p m e n t o f Pars Plan a Le n se ct o m y
the posterior capsule from the infusion system.
While the limbal approach to anterior vitrectomy is more The development of vitrectomy and phacoemulsification has an
comfortable for the anterior segment surgeon, it creates more intertwined and interesting history. The first vitrectomy infusion,
endothelial and iris trauma than pars plana surgery. Limbal suction cutter was patented by Anton Banko in 1969 in response
anterior vitrectomy should be reserved for intraoperative cata- to vitreous complications of early phacoemulsification. Banko
ract surgery complications if the surgeon is not comfortable had learned of the need for a vitreous cutter because he devel-
with sideport infusion combined with pars plana vitrectomy. oped the fluidics for Kelman’s early phacoemulsifier. Machemer
Peripheral anterior synechia frequently form at the limbal developed the trans-PPV procedure in 1970 and shortly there-
wounds postoperatively. The pars plana approach allows more after performed lensectomy with the vitreous suction cutter
complete removal of peripheral membranes and vitreous with- (VISC). It was soon discovered that the vitreous cutter would
out iris trauma (see “Pupillary Membranes”). If conversion to not handle significant nuclear sclerosis. Girard developed the
posterior vitrectomy becomes necessary, the limbal approach fragmenter with Sparta in 1972 as a phacoemulsifier alternative
is inappropriate because the angulated instruments cause without the coaxial infusion sleeve. One of the authors (S.C.)
striate keratopathy. Most pupillary or retro-IOL membranec- was the first advocate of PPL using the Girard fragmenter with
tomies should be done through the pars plana. Translimbal aspiration. Girard advocated vitrectomy with his unit and later
bimanual lensectomy should be used for radiation cataracts in recommended using the fragmenter for routine cataract sur-
retinoblastoma patients and in some cases of severe iris-retinal gery. The author believes that ultrasonic vitrectomy and PPL for
adherence when pars plana entry is unsafe. routine cataract surgery are unsafe. Shock adapted a dental unit
to cataract surgery just as Kelman had done at an earlier date
but, like Girard, could not use a coaxial infusion sleeve, which
PARS PLANA LENSECTOMY had been patented by Kelman. The Shock technique required
a large, intentionally leaky wound and was used with infusion
In d icat io n s through the fragmenter needle rather than suction. Machemer
used the Shock system with a large, leaky pars plana incision
Pars plana lensectomy (PPL) is not indicated for elective
for cases with nuclear sclerosis too great for the VISC.
cataract removal unless the lens is subluxated or dislocated.
The obligatory interruption of the anterior vitreous cor-
Co n ve n t io n al Frag m e n t e r Te ch n iq ue s
tex increases retinal complications (cystoid macular edema
[CME] and retinal detachment), and it is not compatible with Current practice for PPL with the fragmenter begins after
endocapsular IOL implantation. The principal role of PPL is standard 25-, 23-, or 20-gauge port placement. The sur-
in combination with indicated pars plana vitrectomy (PPV). geon may attempt lensectomy with the vitreous cutter if the
Many phakic, proliferative vitreoretinopathy (PVR) lens does not appear too dense. Once a decision to perform
(Chapter 9) and giant break (Chapter 10) cases often require lensectomy with a 20-gauge fragmenter is done, the con-
PPL even if the lens is clear. Many trauma cases require junctiva and sclera should be opened as described in the
removal of the lens because of associated lens damage or 20-gauge wound construction chapter.
ciliary body damage. The next step in conventional lensectomy is to incise
Aphakic eyes allow better visualization, better approach the equatorial lens capsule with the microvitreoretinal
to the anterior vitreous cortex and peripheral vitreous, and eas- (MVR) blade. The author used the fragmenter to incise the
ier fluid-air or fluid-gas exchange; eliminate the substrate for lens capsule before endocapsular lensectomy was developed.
cyclitic membrane formation; and allow faster egress of cells, The fragmenter method was initiated to avoid the stress that
protein, fibrin, fibronectin, hemorrhage, and growth factors. capsular incision with the MVR blade creates on the zonules.
Clear lenses should not be removed in a vitrectomy performed Many surgeons advocate penetration of the nucleus with the
for proliferative diabetic retinopathy (PDR) because of the MVR blade. This step is unnecessary if the nucleus is soft and
increased risk of neovascular glaucoma unless there is fibrin creates excessive stress on the zonules if the nucleus is hard.
syndrome, there is a high probability of postoperative bleed- After incising the lens capsule, the fragmenter is used in
ing, or silicone is used for large retinectomies. It is thought what phaco surgeons would call a sculpting mode to remove
that the anterior vitreous cortex and the lens act as a barrier lens material. The author has advocated sculpting in a plane
to diffusion of vascular endothelial growth factor (VEGF) to parallel to the iris, starting on the temporal side adjacent to
CHAPTER 7 ■ Vitrectom y Techniques and Technology for Anterior Segm ent Problem s 79

the entry site. An initial thick sheet of cortex and nucleus complete capsulectomy is recommended to decrease post-
is removed while being careful to avoid the cortex near the operative inflammation and remove the substrate for cyclitic
anterior and posterior lens capsules. The posterior capsule membrane formation. Most surgeons use the vitreous cutter to
is much weaker than the anterior capsule and is usually not remove the capsule. The senior author has recommended end-
intact by the time the posterior cortex has been removed. opening forceps to remove the capsule since the late 1970s.
Some surgeons recommend alternating aspiration and The author has used the diamond-coated forceps developed
sonification. Unlike the phacoemulsifier, the fragmenter has no with Grieshaber since they became available in the late 1980s.
infusion sleeve and must rely on fluid flow through the lumen Zonulorhexis is performed in a circular motion identical to
for cooling. The needle has approximately 0.003 inches of lon- capsulorhexis. Care must be taken to avoid engaging the vitre-
gitudinal movement and generates significant frictional heat. ous so as to prevent undue force on the retina. Using the vitre-
By contrast, the author has always recommended continuous ous cutter to remove lens capsule frequently damages the iris,
and simultaneous sonification and aspiration. The constant causes miosis, prolongs operating time, may cause bleeding,
fluid flow cools the needle and therefore the sclera. If white, and leaves residual lens material. Residual capsule and lens
particulate matter (lens milk) appears at the needle tip, the sur- material cause inflammation and may lead to cyclitic mem-
geon must release the foot pedal immediately to avoid scleral branes and closure of peripheral iridectomies, especially the
burns. If the needle becomes clogged, it should be back flushed inferior peripheral iridectomy necessary in aphakic silicone
with a syringe filled with balanced salt solution (BSS) with the oil cases.
ultrasound activated after double-checking to determine that
the needle is outside the eye. Aspiration of saline from a cup
Frag m e n t e r Pe rfo rm an ce
is never effective, nor is it necessary to clean the needle with a
stylet or replace the needle if it becomes plugged. The original fragmenter handpieces sold by Berkley Bioengi-
neering, Coopervision, Sparta, Fibrasonics, MidLabs, Storz,
and Alcon were all made by Fibrasonics. These fragmenters
In fusio n Op t io n s
were useful but had less power than the phacoemulsifier and
Many surgeons recommend placing a separate infusion into hence more difficulty with dense nuclear sclerosis. Alcon
the lens rather than using the pars plana infusion cannula. If introduced a four-crystal fragmenter in the early 1990s that
the lens is soft, infusion directly into the lens is not necessary. uses the same ultrasonics as the Legacy and Infinity pha-
If the lens is hard, infusion into the lens will not reach the coemulsifiers. The Alcon Accurus and Constellation frag-
temporal side where the sculpting must begin. Infusion into menters (Fig. 7.2) will handle dense nuclear sclerosis just as
the lens tends to force lens material through ever-present well as the phacoemulsifier.
defects in the posterior capsule. For this reason, the authors
use only the pars plana infusion cannula for lensectomy
Ph aco e m ulsifi cat io n wit h Pars Plan a
cases except during hydrodissection.
Vit re ct o m y
As both phacoemulsification and vitrectomy techniques have
Le n s Cap sule Re m o val
become more sophisticated, performing safe combined pha-
In the presence of anterior PVR or severe intraocular inflam- co-vitrectomy technique is a possibility for select patients.
mation, placement of an IOL should not be performed and This issue is discussed further in Chapter 8.

Fig u re 7 .2 ■ Alco n Co n stel-


lat io n frag m en t er an d p h aco
probes.
80 SECTION II ■ Surgical Technology and Techniques

An t e rio r Le n s Cap sule Re t e n t io n an d Le n s defect frequently leads to capsular tears that extend into
Im p lan t at io n in t h e Sulcus the anterior capsule. Extension of capsular tears was a
frequent problem in phaco surgery until the continuous
The late Ron Michels was long an advocate of preserving the
capsulorhexis was developed. Cataract surgery has seen a
anterior lens capsule until the end of the vitrectomy to reduce
progression from intracapsular to extracapsular surgery and,
damage to the endothelium and trabecular meshwork induced
ultimately, phacoemulsification. Phaco has evolved from
by erythrocytes, infusion fluid, and turbulence. Kokame and
anterior chamber phaco to iris plane phaco to endocapsular
Blankenship reported retention of the anterior lens capsule to
surgery. Continuous capsulorhexis has dramatically reduced
permit the implantation of posterior chamber lens in the ciliary
problems with capsular tears for the cataract surgeon.
sulcus at the end of the vitrectomy. They recommended per-
forming an anterior capsulotomy after lens implantation. Some
En d o cap sular Le n se ct o m y Surg ical Se q ue n ce
phaco surgeons have been critical of this procedure because
endocapsular implantation has better results than ciliary sulcus One of the authors (S.C.) has been using posterior capsu-
placement in elective cataract surgery. The authors have had lorhexis, cortical cleaving hydrodissection, and sculpting
very good results with this method with no cases of decen- since 1994 and has found these techniques of great value
tration. Capsular fibrosis occurs rapidly if the capsule is in in lensectomy. This approach is referred to as endocapsular
contact with silicone oil. The authors have found that polish- lensectomy.
ing the capsule increases the chance of preserving a clear cap- Endocapsular lensectomy begins with placement of the
sule. The authors use Acrysof three-piece foldable acrylic lens ports as described previously for conventional lensectomy.
from Alcon through a 2.2-mm keratome, clear corneal inci- The endoilluminator is essential to stabilize the eye and
sion with implantation in the sulcus, anterior to the retained provide a controllable red reflex. The endoilluminator can
anterior capsule after the vitrectomy. One should avoid the use be touched to the lens capsule in various locations to aid
of silicone lenses as they absorb silicone oil and have marked in visualization without the light toxicity associated with
condensation problems during fluid-air (gas) exchange. Phaco- retina-reflective red reflex methods. The sclerotomy for the
vitrectomy has virtually eliminated the need for this method. fragmenter is made superotemporally for the capsulorhexis,
hydrodissection, sculpting, and cortex-aspiration tools.
A central posterior capsulorhexis is made with vitre-
En d o cap sular Le n se ct o m y Me t h o d
ous cutter (Fig. 7.3) after a limited anterior vitrectomy. The
Conventional lensectomy, as described previously, has many vitrectomy is performed to prevent vitreoretinal traction
of the attributes of endocapsular phaco except that it starts from the fragmenter. Cortical cleaving hydrodissection is
with a risky, equatorial capsular incision. This equatorial the next step. A blunt, 27-gauge cannula attached with a

Fig u re 7 .3 ■ Posterior cap su-


lorh exis is p erform ed with th e
vitreous cutter after anterior vit-
rectom y to p revent eng ag em ent
of vitreous in the fragm enter.
CHAPTER 7 ■ Vitrectom y Techniques and Technology for Anterior Segm ent Problem s 81

Fig u re 7 .4 ■ Cortical cleaving


h yd ro d issect io n is p erfo rm ed
with a b lun t 27-g aug e can n ula
attached to a 3- to 5-m L syringe
via short length of tub ing.

short length of tubing to a 5-mL syringe operated by the nuclear cleaving or rotation is then initiated at the temporal
assistant is used for this step (Fig. 7.4). If endocapsular margin of the nucleus to avoid damaging the capsule with
lensectomy is attempted with the 25-gauge cutter without the fragmenter. The Alcon 20-gauge, four-crystal fragmenter
the fragmenter, hydrodissection can be performed with a is used to sculpt the nucleus, epinucleus, and finally the
27-gauge needle advanced through the 25-gauge cannula to cortex (Fig. 7.5). This fragmenter has the same ultrason-
the edge of the posterior capsulectomy. Sculpting without ics as the Legacy. The anterior cortex is avoided to prevent

Fig ure 7 .5 ■ The nucleus, then


ep in ucleus, an d then cortex are
sculp ted with the frag m enter.
82 SECTION II ■ Surgical Technology and Techniques

Fig u re 7 .6 ■ I/ A is p erform ed
with th e vitreous cutter so th at
cortex can b e cut and asp irated
after u sin g g en tle su ctio n t o
rem ove from capsular bag.

damaging the anterior capsule. The vitreous cutter is used incision. The hydrophobic foldable acrylic multipiece lens
for aspirating the remaining cortex. The vitreous cutter is (Alcon Acrysof) is currently being utilized because of the
better than classic irrigation/aspiration (I/A) tools because low incidence of capsular opacification. Scleral tunnel or
of the cutting capability that acts like chopping. The ante- clear cornea incisions can be used (Fig. 7.7), and the lens
rior capsule is polished after aspirating the cortex (Fig. 7.6). is positioned in the ciliary sulcus, over the intact anterior
Almost any posterior chamber lens could theoretically be capsule. Silicone lenses, as previously stated, should not
utilized, but foldable lenses offer the advantage of a smaller be used because they have severe condensation problems

Fig ure 7 .7 ■ A foldable Acrysof


IOL is in serted th roug h a clear
corneal or scleral tunnel incision.
CHAPTER 7 ■ Vitrectom y Techniques and Technology for Anterior Segm ent Problem s 83

Fig u re 7 .8 ■ Zo n ulorh exis is


p erform ed in a circular fash ion
with th e in tern al lim itin g m em -
b ran e (ILM) o r en d -g rasp in g
forceps.

during fluid-air exchange, have higher posterior capsular traction (Fig. 7.9). When vitreous enters the fragmenter,
opacification rates, and absorb silicone oil. Multipiece the vitrectomy probe, not the fragmenter, should be used to
(three-piece) foldable acrylic lenses are preferable in the remove the vitreous. The fragmenter should never be used
sulcus to single-piece lenses, because the thickness of the for emergency or elective vitrectomy.
haptics in the latter might rub against the posterior iris and
cause postoperative iritis and pigment dispersion. Sub luxat e d Le n se s Durin g Vit re ct o m y
Subluxated lenses are best managed by completing the
Pars Plan a Cap sulo t o m y Aft e r Le n se ct o m y
vitrectomy through the pars plana before approaching the
Capsulotomy is used if the intent is complete capsule removal. lens. Posterior dislocation is frequently seen and presents
An MVR blade is placed through the sclerotomy and used to no problem with the pars plana approach. After the vitre-
incise the anterior and posterior capsules centrally to create ous surrounding the lens is removed, the 20-gauge aspirat-
an “edge.” The fragmenter almost always ruptures the pos- ing ultrasonic fragmenter can safely be used to complete
terior capsule, eliminating the need for posterior capsulo- the lensectomy as described above (Fig. 7.10). Rhexis and
tomy. The forceps method of total capsule removal requires hydrodissection are very difficult if the lens is subluxated or
an edge. If the lensectomy is accomplished with the vitre- dislocated and may be omitted.
ous cutter without the need for enlargement to a 20-gauge
incision, a 25- or 27-gauge needle can be used through the Po st e rio r Dislo cat io n o f Le n s Mat e rial Durin g
25-gauge cannula to incise the anterior lens capsule. Cat aract Surg e ry
Surgica l Psychodyna mics
Pars Plan a Cap sule ct o m y Aft e r Le n se ct o m y
Cataract surgery has been one of the most frequently per-
End-grasping forceps should be used to remove the anterior
formed surgical procedures worldwide for over a century.
and posterior capsules after capsulotomy. Iris contact should
While inexperience is known to cause a higher complication
be avoided. A circular, zonulorhexis approach is better than
rate, high surgical volumes and phenomenal success rates
pulling across the eye because it reduces traction on the
can also cause surgeons to become complacent and make
peripheral retina (Fig. 7.8).
judgment errors when complications do occur. Busy sched-
ules, observers, and video may contribute to faulty decision
Avo id an ce o f Vit re o us in t h e Frag m e n t e r
making when the capsule ruptures and lens material and the
The fragmenter emulsifies formed vitreous gel but does vitreous exchange locations. High success rates, outpatient
not emulsify collagen fibers, giving a false impression of surgery, no stitch, no patch, emmetropia, and topical
vitrectomy. The vitreous fibers remain intact, and suction anesthesia elevate patient expectations unrealistically, which
applied with the fragmenter creates dangerous vitreoretinal can place more pressure on the surgeon.
84 SECTION II ■ Surgical Technology and Techniques

Fig u re 7 .9 ■ Rem oval of th e


p h aco p rob e can cause vitre-
ous p rolap se in to th e an terior
cham ber.

Ea rly Recognition a nd Ma na gement of Defects


the surgeon needs to notice subtle indirect changes during
in the Lens Ca psule
cataract surgery that can alert to the presence of a poste-
Modern microscopes enhance the red reflex, allowing early rior capsular tear and vitreous prolapse. These are (a) sud-
recognition of capsular defects. Very commonly, the posterior den, unexpected deepening of the lens or iris diaphragm,
capsular defect is not recognized by direct visualization until (b) difficulty in rotating the lens inside the capsular bag,
later in the surgery, and early recognition is important to (c) difficulty in manipulating and moving lens fragments,
prevent further complication of the procedure. Therefore, and (d) minor decentration of the lens nucleus. Any of these

Fig ure 7 .1 0 ■ Sub luxated lens


m aterial can b e safely rem oved
with the fragm enter only after all
vitreous has been rem oved.
CHAPTER 7 ■ Vitrectom y Techniques and Technology for Anterior Segm ent Problem s 85

signs should suggest either instability of the lens support by a vitreous cutter causes virtually no inflammation, while
structures or presence of vitreous intermingled with the marked inflammation is the rule after sponge vitrectomy.
remaining lens fragments. If a posterior capsular tear is sus- Mechanical damage to the iris caused by contact with the
pected, the surgeon should immediately decrease all the sponge as it swells and is lifted appears to be the cause of
phaco fluidic parameters (lower the height of the infusion this inflammation, as could the tugging of vitreous fibers on
bottle, lower the vacuum, and lower the aspiration flow rate) a miotic pupil, and the unnecessary and prolonged intraocu-
and proceed operating in “slow motion.” lar manipulation required to finish a surgery using inappro-
Most surgeons feel the urge to quickly withdraw the priate equipment and techniques. One of the authors (S.C.)
phaco tip from the eye when they suspect that a posterior has also observed cellulose material on the anterior vitreous
capsular tear has occurred, but doing so acts as a suction cortex after sponge vitrectomy has been performed. One can
device that would guarantee immediate vitreous prolapse speculate that this retained material causes inflammation in
into the anterior chamber. The surgeon must admit that the addition to that caused by iris trauma. Testing for vitreous
defect has occurred rather than rationalize because of the can be accomplished by injecting air into the anterior cham-
psychological factors described previously. When a capsular ber via the sideport incision and looking for fragmentation
defect is recognized, the first actions should be to leave the of the bubble. Alternatively, a single drop of sterile fluores-
phaco tip inside the eye (with the pedal on position zero) cein from a newly opened ampule can be used to stain the
and inject a viscoelastic into the capsular defect using the vitreous. Injection of small quantities of triamcinolone sus-
sideport incision, creating a barrier between the posterior pension (Kenalog) in the anterior chamber through one of
capsule and the anterior vitreous cortex. High-viscosity, the wounds can also aid in highlighting the prolapsed vitre-
cohesive viscoelastics injected into the defect can serve as a ous, as the steroid particles are adsorbed by the vitreous.
temporary barrier, enabling removal of remaining lens mate- This technique permits the visualization of very thin vitre-
rial and removal of the phaco tip from the eye without caus- ous strands that might be missed otherwise and that cause
ing further vitreous loss. If the lens implant can be placed vitreoretinal traction. Although theoretically this technique
over the defect and posterior to the lens material, it can itself might decrease postoperative inflammation, this is not the
function as a barrier. intended consequence and is only a potential side benefit.
Many surgeons focus exclusively on the prevention or Vitrectomy with a high-quality vitreous cutter is the
management of posterior dislocation of lens material rather preferred method of managing vitreous that presents in the
than the more serious matter of reducing vitreoretinal traction anterior chamber. Alcon builds high-quality cutters for use
and subsequent retinal detachment. Any maneuver designed with their phaco systems such as the Legacy and Infinity
to prevent posterior dislocation that increases vitreoretinal trac- which now has a 23-guage Ultravit, 2500 cut/minute cutter.
tion should not be employed. Kelman described the use of a These cutters should be operated at the highest possible cut-
needle inserted through the pars plana to prevent lens material ting rate and low vacuum/flow (Fig. 7.12). Posterior vitreous
from falling posteriorly. This method ignores the pressure that surgeons use vacuum control in addition to flow control to
must be placed on the eye to place the needle and the anterior reduce vitreoretinal traction. The anterior segment machines
movement of the vitreous that occurs without a barrier. The frequently utilize peristaltic pumps, which do not directly
next section discusses the management of vitreous that pro- control the vacuum. The best procedure is to use a very
lapses through the capsular defect. This discussion intention- low flow rate and vacuum settings to reduce traction on the
ally precedes the discussion of the management of dislocated retina. The cutter should be advanced or held stationary
lens material because reduction of vitreoretinal traction and during vitrectomy, never retracted. Pulling the cutter back
retinal detachment prevention are the most important issues. while vacuum is applied dramatically increases vitreoretinal
traction (Fig. 7.13). The introduction of the cutter through
the original cataract incision will cause excessive fluid egress
Vitreous Loss
due to the mismatch of size and diameter of the cutter to
As previously stated, use of the phacoemulsifier to remove the phaco wound. It is preferable to perform bimanual vit-
vitreous is a dangerous step that should never be under- rectomy with two limbal paracenteses separated 90 degrees
taken. Phaco probes liquefy hyaluronic acid but do not cut apart (one at each side of the larger phaco wound): one for
collagen fibers. Use of a large-bore needle to aspirate “liquid” the sleeveless vitreous cutter and the second for an infusion
vitreous should be avoided because of the obligate vitreoreti- cannula. It might be necessary to place temporary nylon
nal traction. The theoretical “pockets” of liquid vitreous are sutures on the original phaco wound to stop any fluid and
more difficult to locate than the fountain of youth. vitreous egress through that wound while the vitrectomy is
Cellulose sponge vitrectomy as developed by Kasner performed. Sideport infusion is preferable to “dry” vitrec-
has been an obsolete and dangerous method for two decades tomy because it prevents hypotony and therefore reduces
in spite of the important role it played before machine vit- the chance of choroidal hemorrhage. Air should be used
rectomy. A cellulose sponge causes significant traction on the instead of infusion fluid to keep the vitreous from hydrat-
retina as the sponge is lifted to transect the adherent vitreous ing and coming forward. The air helps to delineate the sur-
(Fig. 7.11) and because of wicking. Removal of all vitreous face of the vitreous and keep it confined by surface tension.
86 SECTION II ■ Surgical Technology and Techniques

Fig ure 7 .1 1 ■ Cellulose sponge


vitrectom y or testing for vitreous
causes excessive vitreoretinal trac-
tion because of lifting to cut and
wicking and should never be used.
The sponge also causes m echani-
cal traum a to the iris, resulting in
postoperative inflam m ation.

Sweeping the wound for vitreous is dangerous because of the vitreous cavity. If lens material falls posteriorly, there
the vitreoretinal traction it causes. is a natural tendency for the surgeon to chase it with the
phaco probe. The phaco probe gives the appearance of vit-
reous emulsification but does not sever the collagen fibers
Disloca ted Lens Ma teria l
(Fig. 7.14). The surgeon must consciously stop, relax, and
Phacoemulsifiers, lens loops, and irrigation should never plan before performing any further maneuvers. The best
be utilized in an attempt to extract lens material from plan is usually to let the material fall posteriorly and focus

Fig u re 7 .1 2 ■ Vitreous in the


an t e rio r ch am b er sh o u ld b e
rem oved with a hig h-quality vit-
reous cutter using the m axim um
cuttin g rate availab le an d low
vacuum / flow rate.
CHAPTER 7 ■ Vitrectom y Techniques and Technology for Anterior Segm ent Problem s 87

Fig u re 7 .1 3 ■ Pulling b ack d uring low-sp eed cuttin g with suction ap p lied causes severe
vitreoretinal traction.

Fig ure 7 .1 4 ■ The phacoem ulsifier does not cut collagen fibers; it only disperses the gel, giving
the illusion of vitrectom y. The phaco p robe should never be introduced into the vitreous cavity
in order to prevent dangerous vitreoretinal traction.
88 SECTION II ■ Surgical Technology and Techniques

Fig ure 7 .1 5 ■ Lens loops cause


sig nificant vitreoretinal traction if
used in the vitreous cavity.

on vitreous cleanup and IOL implantation. Lens loops can If the pupil is large, the cornea is clear, and the
put significant traction on the retina and cause retinal breaks surgeon and available staff are optimum for posterior vitrec-
and detachments (Fig. 7.15). Foulds, and subsequently tomy, immediate intervention may be undertaken. In most
Machemer, used a saline stream directed at the retina to cre- instances, though, it is preferable to perform posterior vit-
ate experimental retinal detachments. There is a significant rectomy and removal of lens material at a second procedure.
risk of retinal breaks if saline irrigation is used in an attempt This procedure should be performed when the cornea is clear,
to move the lens material anteriorly (Fig. 7.16). the wound is sealed, and the pupil is well dilated. The timing

Fig ure 7 .1 6 ■ BSS irrig ation to


m ove the lens m aterial anteriorly
can cause retinal breaks.
CHAPTER 7 ■ Vitrectom y Techniques and Technology for Anterior Segm ent Problem s 89

can be from several days to weeks later. If there is a moderate (proportional) suction is increased gradually until the lens
amount of cortex, no inflammation, no glaucoma, and no material is picked up. The lens material is then moved away
lens-corneal touch, a vitrectomy may not be necessary. from the retina and the foot pedal is used to activate soni-
Posterior vitrectomy requires a surgeon specifically fication in midvitreous. Proportional fragmentation power
trained in posterior vitrectomy techniques and a sophis- is used to adjust the power until sufficient sculpting occurs
ticated vitrectomy system. An infusion cannula placed without bouncing of the lens material. If the fragmenter
through the pars plana is essential. An assistant-supported, drills into the lens, the endoilluminator is used to push the
irrigating, corneal contact lens is easier and faster to use than lens material off the tip. Alternatively, the endoilluminator
a sutured-on contact lens. Wide-angle visualization systems can be used to crush or “chop” the fragment that is speared
increase cost, complexity, and the learning curve, although on the fragmenter tip. This process is continued until all lens
they provide an excellent view of the periphery (unneces- fragments are removed.
sary in anterior vitrectomy). A fiberoptic endoilluminator is Perfluorocarbon (PFC) liquids (Chang) were intro-
essential for all cases. Light reflexes from the cornea prevent duced to vitreoretinal surgery for unfolding giant breaks,
the surgeon from having an optimal view if coaxial illumina- draining subretinal fluid, and stabilizing the retina during
tion is used. Iris retractors increase inflammation and may the dissection of epiretinal membranes. PFC liquids can also
cause a distorted pupil after surgery. Some surgeons have be used to float the lens material away from the retina, allow-
advocated indirect ophthalmoscopic visualization, but an ing aspiration-fragmentation or phacoemulsification to be
inverted view and the need to support the lens with one performed anteriorly (Fig. 7.17). This method increases cost
hand make this approach dangerous. and may require a subsequent procedure to remove residual
All vitreous should be removed before removing any PFC liquids. The PFC liquid method is safe but unneces-
dislocated lens material. Many surgeons have the miscon- sary, unless there is extremely dense nuclear sclerosis that
ception that lens material can damage the retina if it falls should not have been managed with phaco in the first place.
posteriorly. Inappropriate techniques, not the lens, damage It may be useful for surgeons with limited posterior vitrec-
the retina. It is dangerous and unnecessary to leave a layer tomy experience.
of vitreous under the lens material until it is removed. Some It is imperative to evaluate the retinal periphery at the end
cortex may be removed with the vitreous cutter, but dense of removal of dropped lens fragments. The retinal periphery
nuclear material requires the phacofragmenter. The Alcon may be impossible to fully evaluate for retinal tears preopera-
four-crystal fragmenter utilizes the same drive electronics tively due to the presence of the lens fragments and dispersed
and piezo driver as the Infinity phaco probe and, similarly, is cortex. Despite what a referring cataract surgeon tells the con-
able to handle the majority of nuclear sclerosis cases. sulting vitreoretinal specialist, if vitreous loss occurred dur-
After removal of the vitreous, the fragmenter is intro- ing the cataract surgery, we should assume the presence of
duced and moved to the surface of the lens material. Linear peripheral retinal tears until otherwise demonstrated.

Fig ure 7 .1 7 ■ PFC liquids such


as PFO can be used to float a hard
nucleus into the anterior cham -
ber enabling rem oval with a lens
loop .
90 SECTION II ■ Surgical Technology and Techniques

Ha rd Disloca ted Lens Ma teria l permits removal of all peripheral cortex and capsule. One of
the authors (S.C.) has used pars plana lensectomy with total
Hard dislocated nuclei can usually be crushed between the capsule romoval many pediatric cataracts since 1975 and
endoilluminator and the aspirating 20-gauge ultrasonic frag- has never had a postoperative retinal detachment. The con-
menter (14,15). After the pieces are made smaller, they can ventional I/A approach results in a fibrous ring often includ-
be removed with the fragmenter. If the fragmenter becomes ing iris, neocortex, ciliary body, pars plana, and peripheral
plugged, it can be removed from the eye and back flushed vitreoretinal traction. This configuration can lead to retinal
with sonification as often as required. detachment with even minor trauma. Phaco and IOL implan-
If the bimanual crush technique is not effective because tation often with primary posterior capsulorhexis and ante-
of a dense, black nucleus, internal simultaneous fluid-gas rior vitrectomy can be used in children of appropriate age, if
exchange can be performed if PFC liquids are not available. there is a normal-sized eye and no glaucoma or vitreoretinal
The endoilluminator should then be removed and the sclero- problem (20–23).
tomy plugged with a scleral plug. The surgeon’s other hand
can then make a razor blade-scissors limbal incision of 90
to 110 degrees to remove the nucleus. The iris will prolapse TRAUMATIC CATARACTS
unless the infusion is turned off after the section is made. An
assistant can elevate the cornea and the lens can be removed Most traumatic cataracts are either subluxated or have had
with a 1-mm cryoprobe without touching the endothelium. interruption of the anterior vitreous cortex. Vitrectomy
Perfluoron (PFO) can be used to eliminate the need for a instruments should be used to remove all vitreous and soft
cryoprobe. The wound is then closed with a running shoe- lens material using standard pars plana techniques. After vit-
lace or X-type 8-0 or 9-0 monofilament nylon suture. This rectomy, the fragmenter can be utilized if denser lens material
method has not been needed for decades. is present. The retinal periphery should be always examined
at the end of lensectomies for subluxated lenses, since the
Intra ocula r Lens Impla nta tion original contusive force that created zonular damage could
have created a retinal dialysis or giant retinal tear.
Some retinal surgeons are opposed to lens implantation in
cases of posterior dislocation of lens material for unclear rea-
sons. The authors recommend lens implantation if certain PUPILLARY MEMBRANES
conditions are met. If the posterior capsule can support an
IOL, it can be placed in the bag with the haptics rotated Multiple tissues contribute to the formation of pupillary
away from the capsular defect. If the posterior capsule will membranes. There is a wide range in density from an opaque
not support an IOL, the IOL can be implanted in the ciliary hyaloid to extremely dense calcified membranes. These
sulcus anterior to the anterior lens capsule. membranes may be postsurgical or posttraumatic or may
If the capsule is not sufficient to support the IOL, an occur spontaneously. A range of approaches to these prob-
anterior chamber lens can be used. Anterior chamber lenses lems, depending on density, is required (24). Neodymium
are contraindicated if there is a low endothelial cell count, YAG laser capsulectomy can be utilized for thin or mod-
significant open angle glaucoma, or uveitis. Judgment is erately dense pupillary membranes. The extravagant term
required to set the level of cell counts and severity of glau- “anterior segment reconstruction” is inappropriate. As dis-
coma that represent contraindications. cussed earlier, the pars plana approach is preferable to the
If there is insufficient capsular support for a posterior limbal approach in managing most pupillary membranes.
chamber lens and low corneal endothelial cell counts or signif-
icant glaucoma prevents the use of an anterior chamber lens, a
sutured lens can be used. This technique requires practice and
Me m b ran o t o m y (Discissio n )
can result in significant intraoperative and postoperative com- Simple membranotomy (discission) or neodymium YAG
plications, including suprachoroidal hemorrhage as well as late laser techniques will suffice for a thin membrane such as
complications such as endophthalmitis and suture breakage. lens capsule. The MVR blade can be utilized translimbally
or through the pars plana for the task. If membranectomy
is required, pars plana membranotomy serves as the initial
CONGENITAL AND PEDIATRIC stage to provide an edge on which the vitrectomy instrument
CATARACTS can imbricate and cut. As an alternative, 20- to 25-gauge
scissors can be used to segment the membrane and free it up
PPL is suitable for cataracts occurring in infants and young from the zonules and any scarring that is present.
children. Extracapsular cataract extraction technique in this
age group uniformly results in capsular clouding requiring
Me m b ran e ct o m y
discission or YAG capsulotomy (16–19). Limbal anterior vit-
rectomy (17) reduces late vitreoretinal traction and detach- After an edge is made with the MVR blade, the vitrectomy
ment. A pars plana approach has less corneal problems and instrument can be used to remove a moderately dense lens,
CHAPTER 7 ■ Vitrectom y Techniques and Technology for Anterior Segm ent Problem s 91

iris, or fibrous material. The infusion cannula should be In fusio n So urce s fo r Clo se d
placed only if the pars plana is visible through the membrane An t e rio r Vit re ct o m y
to verify infusion at the correct location. The best approach
Unless the presence of dense hyphema, cataract, or pupillary
if the infusion site cannot be visualized is bimanual, using
membranes dictates the use of the bimanual, angulated can-
a 20-gauge, 30-degree angulated, blunt infusion cannula,
nula approach, the pars plana infusion cannula is the best
through the superonasal pars plana.
method for elective anterior vitrectomy. If more extensive
vitrectomy is required, the pars plana infusion cannula
De n se Me m b ran e ct o m y should be placed after the initial bimanual infusion tech-
nique and removal of anterior opacities has been used to
If the vitrectomy cutter using moderate suction levels can-
obtain a view of the pars plana. If preoperative ultrasound
not cut a dense membrane, scissors should be utilized. The
examination demonstrates attached retina with normal pars
membrane is cut into many small triangular pieces that can
plana anatomy, the 25-gauge infusion cannula can be placed
be removed with the vitrectomy instrument. Bipolar biman-
in a standard fashion prior to anterior vitrectomy.
ual diathermy (Chapter 4) can be used to control bleed-
ing from these membranes. Care should be taken to avoid
excessive tissue removal; a 4- to 5-mm opening is usually Vit re o co rn e al To uch
sufficient if the iris is incorporated into the membrane. Off-
center opening can be used to avoid areas of corneal astig- Anterior vitrectomy can cause complete clearing of corneal
matism or opacity. All tissues adherent to the ciliary body edema due to vitreous contact, if rigid case selection is uti-
can be removed to reduce hypotony and phthisis. lized (25). It is essential to obtain a history showing that
While an 8-mm opening resembles a dilated pupil, it the cornea was once clear after lens removal, subluxation,
causes glare, discomfort, reduced vision, and cosmetic prob- or dislocation (26) and that endothelial cell counts are
lems postoperatively. Occasionally, a calcified membrane will reasonable. In this way, surgery for corneal edema primar-
be resectable outside the calcified zone or crushable with a ily due to endothelial trauma, which will not respond to
sclerectomy punch. Limited anterior vitrectomy should then subsequent vitrectomy, can be avoided. Long-term touch
follow to prevent subsequent pupillary block or corneal with retrocorneal membrane formation is equally unrespon-
touch. sive to anterior vitrectomy. The pars plana approach prevents
further endothelial trauma, in contrast to a limbal approach.
Vitrectomy should be completed behind the iris and using
ANTERIOR VITRECTOMY the 25-guage infusion cannula to minimize anterior cham-
ber turbulence and reduce endothelial damage.
Anterior vitrectomy is useful for a wide variety of anterior
segment problems: aphakic pupillary block, vitreous touch, Ap h akic Pup illary Blo ck
CME with vitreous to the wound, vitreous loss at cataract
surgery, aphakic keratoplasty, secondary IOL implantation, While simple slashing of the anterior vitreous cortex or YAG
and aphakic trabeculectomy. Each of these will be discussed laser vitreolysis will suffice in some cases of pupillary block,
individually, but the methodology is similar and will be dis- more frequently an anterior vitrectomy is required (27). The
cussed in the following sections. pars plana approach decreases iris and endothelial damage
in these shallow chamber cases.
Tran slim b al An t e rio r Vit re ct o m y
Ciliary Blo ck Glauco m a
The simplest and most frequent form of anterior vitrec-
tomy is that required for vitreous loss at cataract surgery or Trans-PPL and trans-PPV will alleviate ciliary block (malig-
aphakic keratoplasty. The vitrectomy probe is placed in the nant, aqueous misdirection) glaucoma in every case. The
central anterior vitreous space and used with minimal move- creation of an unobstructed passage of fluid from the vit-
ment. The iris, endothelium, and especially the peripheral reous cavity through the anterior vitreous, zonule or lens
vitreous are avoided. Slight increased suction is required, capsule diaphragm, and iris is curative for this condition.
compared to closed vitrectomy, because there is no infu- A stepwise approach can be utilized to guarantee this fluid
sion to contribute to transorifice pressure. Care should be passage. Core vitrectomy can be first performed, followed by
taken to avoid air into the cutter port as it increases vacuum anterior vitrectomy. In the setting of pseudophakia, a periph-
requirements, thereby decreasing safety. A rapid cutting rate eral iridectomy from the posterior pars plana approach can
is to be used to reduce vitreoretinal traction and improve be performed. Deepening of the anterior chamber and ref-
fluidic stability. The approach in penetrating keratoplasty is ormation of the angle may be required if a flat chamber has
similar, but more extensive vitrectomy is usually required to been present for over 24 hours. It is advisable to deepen the
prevent postoperative corneal vitreous touch. Closure on an chamber with BSS instead of a viscoelastic, since reflatten-
air bubble is best to avoid residual strands to the wound. ing of the chamber implies that the aqueous misdirection
92 SECTION II ■ Surgical Technology and Techniques

has not been relieved. Viscoelastics may artificially maintain Hypotony also causes striate keratopathy and miosis. Air
anterior chamber depth despite persistence of the misdirec- should be used instead of BSS to eliminate hydration of
tion syndrome. If the iridectomy does not repair the malig- the vitreous as well as prevent vitreous incarceration in the
nant glaucoma, explantation of the IOL may be required. wound. If a sideport is present, infusion should be used
Aqueous misdirection in a phakic patient may require vit- through this site to reduce turbulence. If “one-handed”
rectomy-lensectomy, since it is impossible to create anterior technique was used without a sideport, a sideport for infu-
vitrectomy and iridectomy without violating the lens capsule sion is shoule be placed.
and creating a cataract.
Cyst o id Macular Ed e m a
Ap h akic Filt e rin g Pro ce d ure s
There is no need to perform anterior vitrectomy if vitreous is
Most aphakic filtering procedures fail because of closure of in the cataract wound and there is no CME. If only a strand
the filtration site by episcleral tissue proliferation. Occa- of vitreous is seen in contact with the wound, laser YAG
sionally, vitreous can plug a filtration site internally. On vitreolysis might be sufficient to sever a strand rubbing on
occasion, this can be recognized at the time of aphakic the iris and help relieve CME. If CME is present without vit-
trabeculectomy and managed with the vitrectomy instru- reous in the wound, vitrectomy is unlikely to improve the
ment. The sleeve is quite useful in these cases because edema (28,29). Topical Nevanac should be tried for several
the incision is already large and the eye too soft to place months when postoperative CME is present. Topical predni-
the infusion cannula or make a second incision. Vitrec- solone or Durezol should be used as well unless the patient
tomy plays a role in the management of other glaucoma is a steroid responder. If topical treatment is not effective
problems such as phacolytic glaucoma, as discussed and the patient is not a steroid responder, retrobulbar, juxta-
elsewhere. scleral triamcinolone or intravitreal triamcinolone should be
tried before PPV is considered. Retrobulbar steroids prob-
ably diffuse through the sclera, but posterior juxta-scleral
Vit re o us Lo ss at Cat aract Surg e ry
delivery produces a higher drug level at the macula. Topi-
If vitreous presents in the anterior chamber during cata- cal and subconjunctival treatments target the source of the
ract surgery, the vitrectomy instrument can be used with inflammation in postoperative CME (Irvine Gass syndrome)
sideport air infusion (Fig. 7.18) for anterior vitrectomy. So- but produce 1/100 the anterior chamber drug level in phakic
called dry vitrectomy is not advised because of unavoid- or pseudophakic eyes (two-compartment eyes). Leakage of
able hypotony and risk of suprachoroidal hemorrhage. the perifoveal capillary bed persists after vitrectomy, and the

Fig ure 7 .1 8 ■ Bim anual vitrec-


tom y utilizing a 23-gauge vitreous
cutter and BSS infusion throug h
a 21-g aug e can n ula is th e p re-
ferred m ethod of anterior vitreous
rem oval.
CHAPTER 7 ■ Vitrectom y Techniques and Technology for Anterior Segm ent Problem s 93

visual improvement is usually modest. It is probable that be avoided. Cataract surgery without vitrectomy may result
the visual improvement results primarily from clearing the in pupillary or cyclitic membranes and pupillary block. It is
media. It appears that vitrectomy rarely causes CME, and not necessary to “quiet” eyes with steroids before vitrectomy
rarely a successful case will develop recurrent CME in spite and lensectomy. Vitrectomy reduces, rather than increases,
of having the vitreous removed. Because of the modest differ- inflammation and the risk of phthisis.
ences between vitrectomized and nonvitrectomized eyes in Iris contact and retractors should be avoided to reduce
the national collaborative study (30) and the risk of aphakic inflammation. Scleral depression can be used to remove all
retinal detachment and endothelial damage, caution must be peripheral lens material with the forceps method without
applied in the utilization of vitrectomy for CME (31–33). iridectomy. Subconjunctival repository steroids are essen-
If surgery is undertaken, the infusion cannula can be tial in these cases unless the patient is a steroid responder.
used superotemporally and the vitrectomy instrument inf- The authors rarely use systemic steroids because of the well-
erotemporally. In this way, the fibers from iris border to a known medical risk factors.
superior cataract wound can be removed without dangerous The authors strongly urge surgeons to avoid pha-
sweeping techniques. All vitreous adherence to the iris should coemulsification in severe uveitis cases. Capsule retention
be eliminated because this may play a role in inflammation and the IOL result in a two-compartment eye with increased
and CME. Low suction force should be used to reduce post- retention of prostaglandins, complement, cells, and other
operative aphakic retinal detachment. Vitreous scissors can be components of the inflammatory process in the vitreous cav-
used to sever iris-to-wound vitreous fibers. Subconjunctival ity, thereby increasing CME.
steroids are essential at the end of surgery unless the patient The lens capsule should be removed after lensectomy
is a steroid glaucoma responder. Posterior juxta-scleral triam- using end-grasping forceps to reduce lens-related inflam-
cinolone injected at the end of the case maximizes the dose to mation and prevent retro-iris membranes, which increase
the posterior segment while reducing the risk of steroid glau- hypotony, phthisis, and vitreoretinal traction.
coma compared to subconjunctival or peribulbar steroids. Juxta-scleral triamcinolone injected at the end of the
case maximizes the dose to the posterior segment while
reducing the risk of steroid glaucoma compared to subcon-
Ap h akic Ke rat o p last y
junctival or peribulbar steroids.
Successful aphakic keratoplasty can be threatened by vitre-
ous touch, pupillary block, and iris touch to the host-donor
interface. Anterior, open sky vitrectomy can be used without INTRAOCULAR LENSES
infusion in these cases. Care should be taken to avoid pull-
ing the probe back during cutting to avoid vitreoretinal trac- Many IOL problems can be managed effectively with a vitrec-
tion and air ingestion. Chamber deepening by pressing on tomy. At times, a normal-functioning IOL must be removed
the midiris with a small iris spatula through the pars plana in the context of posterior vitreoretinal surgery. It is essential
wound can deepen the chamber without placing an instru- that the implant surgeon be familiar with the vitrectomy-
ment in the angle. based management of postoperative problems.

Re t ro le n t al In t rao cular Le n s Me m b ran e s


PHACOLYTIC GLAUCOMA
The YAG laser can manage most retro-IOL membranes. Pars
Macrophages laden with ingested lens material clogging the plana or limbal membranotomy (discission) is infrequently
trabecular meshwork are best managed with vitrectomy. indicated. Denser membranes require trans–pars plana
Gentle suction with a small, blunt cannula near the trabe- membranectomy with the vitrectomy instrument. While the
cular meshwork can enhance the removal process. Careful infusion sleeve can be utilized, it increases probe diameter
irrigation of the angle can be utilized as well. Frequently, and reduces access to the membrane. Standard infusion can-
cortex is entrapped in the peripheral cortical vitreous, usu- nula placement is preferred to maintain adequate intraocular
ally inferiorly. Care should be taken to remove this material pressure during the surgery. Membranotomy with the scis-
without vitreoretinal traction. sors, needle, or MVR blade must precede membranectomy
to provide an edge. Occasionally, dense membranes will
require scissors to radially segment and circumferentially
UVEITIS dissect the membrane away from the ciliary body and iris.

Cataracts secondary to severe uveitis are usually best


Dislo cat e d In t rao cular Le n s Re p o sit io n in g
removed by a trans-PPL and trans-PPV approach. Vitrec-
tomy with lens removal for uveitis usually results in the Posterior chamber lens implants occasionally dislocate into
deposition of inflammation on the posterior lens capsule the vitreous cavity. On very rare occasion, positioning and
and little, if any, improvement in CME and therefore should miotics can return the implant to proper position without
94 SECTION II ■ Surgical Technology and Techniques

Fig ure 7 .1 9 ■ Com plete vitrec-


tom y sh ould p reced e IOL rep o-
sitioning to p revent vitreoretinal
traction.

surgery. If this approach is unsuccessful, vitrectomy is forceps (34) should be used to grasp the IOL optic with
required. Surgical manipulation of a dislocated IOL without the endoilluminator providing illumination and additional
prior vitrectomy causes vitreoretinal traction and should be support for the lens. The implant can be placed in the intact
avoided. portion of the capsule, the ciliary sulcus, or in the ante-
Complete vitrectomy with the infusion cannula, rior chamber (Fig. 7.20). Rotation of the lens in the cap-
vitrectomy probe, contact lens or wide-angle viewing sys- sular bag away from the defect that caused the dislocation
tem, and endoilluminator should precede IOL reposition- can be effective in certain cases (Fig. 7.21). Ciliary sulcus
ing to avoid vitreoretinal traction (Fig. 7.19). End-grasping placement can be used if the anterior and posterior capsules

Fig ure 7 .2 0 ■ The IOL can b e


placed in the capsule if sufficiently
intact, in th e sulcus if sufficient
cap sule is p resent, or in the ante-
rior cham ber if no Fuchs d ystro-
phy or significant glaucom a.
CHAPTER 7 ■ Vitrectom y Techniques and Technology for Anterior Segm ent Problem s 95

Fig u re 7 .2 1 ■ Rotation of the


IOL in to the cap sular b ag with
the hap tics away from the cap -
sular d efect can occasion ally b e
effective.

are fused together and there is sufficient capsular support Ciliary sulcus suturing through positioning holes was
(Fig. 7.22). Anterior chamber relocation of certain IOLs can reported by the principal author but is seldom possible or
be utilized if there is no glaucoma and the endothelial cell indicated today. Sutures can be passed around the haptics for
counts are good. Vaulting of plate IOLs and silicone IOLs ciliary sulcus suturing. This method is complex and requires
limits the value of this method. A peripheral iridectomy with experience and careful planning and results in a significant
the vitreous cutter should be used in all cases to prevent incidence of late complications such as suture breakage and
pupillary block. endophthalmitis.

Fig u re 7 . 2 2 ■ Ciliary su lcu s


placem ent can be used if there is
sufficient cap sular sup p ort.
96 SECTION II ■ Surgical Technology and Techniques

Suturing of the IOL to the iris with McCannel sutures Ep it h e lial In g ro wt h


may be performed in certain situations. To perform this
Vitrectomy instruments can permit removal of the fibrous
technique, the IOL should be grasped with the forceps and
tissue, anterior vitreous cortex, and lens remnants in the
the optic passed through the pupil into the anterior cham-
treatment of epithelial ingrowth (35). Pretreatment of the iris
ber, while keeping the haptics below the iris. Pharmaco-
with laser photocoagulation aids in identification of the tis-
logic constriction of the pupil with carbachol in the anterior
sue and its removal. It does not kill all abnormal cells but can
chamber will keep the IOL in the pupil capture position.
be advantageous in defining the extent of involvement. The
The absence of a functional pupil sphincter makes the iris
wound should be explored and the edges excised. An iridec-
suturing of IOLs significantly more difficult. Once the IOL is
tomy with a 1- to 2-mm margin should then be performed
stable, a viscoelastic can be injected into the anterior cham-
with Vannas scissors. The wound should be closed tightly
ber to push the iris posteriorly and delineate the contour of
with a running shoelace, 8-0 monofilament nylon suture.
the haptics. Suturing with a long, straight or curved spatu-
Internal fluid-air exchange (Chapter 4) should be performed
lated needle with 10-0 Prolene can be then performed by
by injecting air through the infusion system while allowing
passing the needle through a limbal paracentesis, through
fluid egress with the vitrectomy instrument. Transcorneal
the iris, underneath the haptic, through the iris, and out
and transscleral cryopexies are then applied over affected
of the eye through the limbus. An exit paracentesis is not
areas with a 2-mm margin. The thermal insulating effect of
required. Mobilization of the loops of Prolene with a Kuglen
the air bubble causes more uniform destruction of abnormal
hook toward a central paracentesis directly above the haptic
tissue with less scleral damage.
position and out through the limbus allows the surgeon to
The cytotoxin, 5-fluorouracil, has been used in con-
tie a knot that can be then reintroduced into the eye, secur-
junction with the surgical approach by some investigators in
ing the position of the haptic. This technique can be per-
the hope of inhibiting regrowth of epithelial cells. Its long-
formed in both haptics, if there is total luxation of the IOL,
term efficacy is uncertain. Fortunately, ingrowth through
or can be performed on a single haptic, if the contralateral
cataract wounds is extremely rare because of small incision
haptic is stable.
techniques and advanced wound construction.

Dislo cat e d In t rao cular Le n s Re m o val


On occasion, marked inflammation or retinal problems will References
necessitate removal of a dislocated IOL. A total vitrectomy 1. Michels RG. Anterior segment and vitreoretinal surgery through
should be performed as described previously and the lens the pars plana, Part I. Ann Ophthalmol 1976;8:1353.
optic grasped with forceps. The IOL should be brought 2. Michels RG. Anterior segment and vitreoretinal surgery through
anteriorly and a scleral plug placed in the endoilluminator the pars plana, Part V. Ann Ophthalmol 1976;8:1497.
opening. A limbal or clear corneal section of appropriate 3. Michels RG, Stark WJ. Vitrectomy technique in anterior segment
surgery. Trans Am Acad Ophthalmol Otolaryngol 1976;81:382.
size should be made with the surgeon’s other hand using 4. Taylor HR, Michels RG, Stark WJ. Vitrectomy methods in ante-
the diamond knife-scissors technique also used for very rior segment surgery. Ophthalmic Surg 1979;10(10):25.
dense nuclei and large intraocular foreign bodies. The IOL 5. Michels RG. Anterior segment applications of vitrectomy tech-
can be passed from the 25-gauge forceps to a second forceps niques. Trans Ophthalmol Soc UK 1978;98(4):458.
such as 20-gauge diamond-coated forceps through the lim- 6. Stark WJ, Michels RG. Anterior segment surgery using instru-
ments designed for pars plana vitrectomy. Trans Penn Acad
bal incision for explantation. After the IOL is removed, the Ophthalmol Otolaryngol 1981;34:27.
limbal wound is closed with a running shoelace or X-type 7. Michels RG, Paton D. Results of radical anterior vitrectomy: a
10-0 monofilament nylon suture. Smaller diameter sutures preliminary report of 26 cases. Ophthalmic Surg 1970;1:33.
cause less astigmatism but require care to prevent wound 8. Charles S. Anterior segment vitrectomy. In: Carroll D, ed. Sur-
leaks during vitrectomy. gery of the eye. New York: Churchill Livingstone, 1988.
9. Charles S. The Charles anterior segment infusion sleeve. Ocu-
tome Fragmatome Newsl 1978;3(2):6.
In t rao cular Le n s Re m o val Be fo re 10. May D. Closed vitrectomy for vitreous prolapse during cataract
extraction. Ocutome Fragmatome Newsl 1979;4(2):2.
Tran s–Pars Plan a Vit re ct o m y 11. Girard LJ, Hawkins RS. Cataract extraction by ultrasonic aspi-
Occasionally, IOLs should be removed because of fibro- ration vitrectomy by ultrasonic aspiration. Trans Am Acad Oph-
thalmol Otolaryngol 1974;78:50.
vascular membranes from diabetic anterior vitreous cortex 12. Charles S. Trans-pars plana and posterior chamber lensectomy
fibrovascular proliferation, trauma, or uveitis. The hap- with the Girard phacofragmenter and automated suction. In:
tics should be cut with scissors with viscoelastics used to Emery J, ed. Current concepts in cataract surgery, selected pro-
maintain the anterior chamber. The haptics can be left in ceedings of the Fifth Biennial Cataract Surgical Congress. St. Louis:
the eye if they are surrounded by a fibrous capsule and C.V. Mosby; 1978.
13. Charles S. Trans-pars plana lensectomy update. Ocutome Frag-
cannot be rotated out with forceps. Multiple X-type 9-0 matome Newsl 1980;5(3).
or 10-0 monofilament nylon sutures facilitate tight wound 14. Michels RG, Shockett DE. Vitrectomy techniques for removal
closure. of cataract lens material. Arch Ophthalmol 1977;95(10):1767.
CHAPTER 7 ■ Vitrectom y Techniques and Technology for Anterior Segm ent Problem s 97

15. Morse PH. Intracapsular cataract extraction at the time of 26. Snip RC, Kenyon KR, Green WR. Retrocorneal fibrous
vitrectomy. Ophthalmic Surg 1979;10(3):65. membrane in the vitreous touch syndrome. Am J Ophthalmol
16. Parks MM, Hiles DA. Management of infantile cataracts. Am J 1975;79(2):233.
Ophthalmol 1967;63:10. 27. Irvine A. Pars plana vitrectomy for malignant and aphakic
17. Parks MM. Posterior lens capsulectomy during primary cata- pupillary block glaucoma. Trans Pac Coast Otoophthalmol Soc
ract surgery in children. Ophthalmology 1983;90:344. 1977;58:189.
18. Calhoun JH. Cutting-aspiration instruments. Int Ophthalmol 28. Orth DH, Henry MD. Management of Irvine Gass Syndrome
Clin 1977;17(4):103. using argon laser photocoagulation and pars plana vitrectomy.
19. Chrousos GA, Parks MM, O’Neill JF. Incidence of chronic glau- Presented at the Bicentennial Cataract Surgical Congress,
coma, retinal detachment and secondary membrane surgery in Miami Beach, Florida, February 1977.
pediatric aphakic patients. Ophthalmology 1984;91:1238. 29. Rice TA, Michels RG. Vitreous wick syndrome-current surgical
20. Hamill MB, et al. Pediatric cataracts. Curr Opin Ophthalmol management. Am J Ophthalmol. 1978;85:656.
1999;10(1):4–9. 30. Fung WE. Vitrectomy for chronic aphakic cystoid macular
21. Malukiewicz-Wisniewska G, et al. Intraocular lens implanta- edema. Ophthalmology 1985;92:1102.
tion in children and youth. J Pediatr Ophthalmol Strabismus 31. Pendergast SD, et al. Vitrectomy for chronic cystoid macular
1999;36(3):129–133. edema. Am J Ophthalmol 1999;128(3):317–323.
22. Simons BD, et al. Surgical technique, visual outcome, and com- 32. Ikeda T, et al. Vitrectomy for cystoid macular edema with
plications of pediatric intraocular lens implantation. J Pediatr attached posterior hyaloid membrane in patients with diabe-
Ophthalmol Strabismus 1999;36(3):118–124. tes. Br J Ophthalmol 1999;83(1):12–14.
23. Zwaan J, et al. Pediatric intraocular lens implantation. Surgical 33. Holekamp NM. Treatment of pseudophakic CME. Ocul Immu-
results and complications in more than 300 patients. Ophthal- nol Inflamm 1998;6(2):121–123.
mology 1998;105(1):112–118. 34. Wilson DL. A new intraocular foreign body retriever. Ophthal-
24. Treister G, Machemer R. Pars plana approach for pupillary mic Surg 1975;6(4):64.
membranes. Arch Ophthalmol 1978;96(6):1014. 35. Stark WJ, Michels RG, Maumenee AE, et al. Surgical manage-
25. Wilkinson CP, Ramsey JJ. Closed vitrectomy for the vitreous ment of epithelial ingrowth. Am J Ophthalmol 1978;85(6):772.
touch syndrome. Am J Ophthalmol 1980;90(3):304.
C H AP TER
8
COMBINED
PHACOEMULSIFICATION
VITRECTOMY PROCEDURES

Many surgeons now combine phacoemulsifica- dissection, drainage of subretinal fluid through retina breaks,
tion with vitrectomy for a variety of reasons. This or retinopexy is required. Posterior subcapsular cataracts
approach adds significant complexity but is indi- interfere with visualization more than nuclear sclerotic cata-
cated in certain situations. There is a widespread racts. If the surgery is elective, as is typically the case with
but incorrect notion that vitrectomy inevitably leads to cata- macular surgery, cataract surgery can be performed 1 month
ract. It is widely appreciated that vitrectomy leads to progres- before vitrectomy if the cataract is likely to interfere with
sion of preexisting nuclear sclerosis, likely due to ascorbic visualization during vitrectomy. A potential problem with
acid depletion and resultant permanent increase in the partial pre–pars plana vitrectomy (pre-PPV) cataract surgery is the
pressure of oxygen in the vitreous cavity by 7 to 12 mm Hg accurate determination of axial length. The Zeiss IOL Master
(Holekamp, Chang, Steffanson). BSS Plus (Alcon) has been uses the retinal pigment epithelium (RPE) for measurement
available for three decades (Edelhauser) and has eliminated in contrast to A-scan ultrasound that uses the ILM. Mea-
the development of posterior subcapsular cataract during surement from the RPE is not a problem with epimacular
the procedure, yet many surgeons use BSS or, even worse, membranes (EMMs) or vitreomacular traction syndrome;
lactated Ringer’s solution. Other surgeons choose inappro- however, the A-scan ultrasound axial length may be reduced
priately to add a variety of compounds including bicarbon- when these conditions are present or increased when mac-
ate, dextrose, antibiotics, and epinephrine, all of which can ular holes are present. Fixation is an additional issue with
lead to posterior subcapsular cataract, especially if infrequent macular disease; it may be difficult to determine if the axial
mixing errors do not occur. Bicarbonate is unnecessary and length is measured in the fovea or an extramacular region
inappropriate to use with BSS Plus because it is correctly buff- with either technology.
ered without additives. Dextrose was added three decades Cataract surgery performed as a separate procedure
ago when diabetic patients were often markedly hyperglyce- after PPV permits more accurate axial length measurement.
mic during surgery, but the widespread availability of serum The procedure can be performed before or shortly after PPV
glucose monitoring during surgery has eliminated this issue. if cataract interferes with examination of the retina and a
Contact of the posterior lens with a gas bubble over a period retinal detachment (RD) is present. For this scenario, the
of several days leads to posterior subcapsular cataract; this options are phaco-vit or pars plana lensectomy with com-
occurs only if the patient does not maintain the correct posi- plete removal of the posterior capsule. Although endocapsu-
tion and when there is discontinuity in the anterior vitreous lar lensectomy with retention of the anterior capsule followed
cortex. Patient education is very important. Some of what is by intraocular lens (IOL) implantation in the ciliary sulcus
described as poor compliance by the patient should be attrib- is possible, this typically results in severe capsular opacifica-
uted to poor patient education. The vast majority of younger tion. Therefore, the anterior capsule could not be utilized as
patients with a clear lens undergoing vitrectomy will retain a barrier for silicone oil posterior retention. The lensectomy,
a clear lens for decades if BSS Plus without additives is used complete removal of the capsule with forceps and inferior
and gas bubble contact with the lens is avoided. In short, iridectomy, approach has been advocated for these cases, but
cataract is not inevitable after vitrectomy. phaco-vit approach is now favored because it enables the
Optimal visualization is essential for vitrectomy, espe- preservation of a silicone oil barrier and optimal correction
cially if epiretinal or internal limiting membrane (ILM) of aphakia.

98
CHAPTER 8 ■ Com bined Phacoem ulsification Vitrectom y Procedures 99

COMPARISON AMONG SEQUENTIAL PHACO AND VITRECTOMY, COMBINED


PHACO-VITRECTOMY, AND PARS PLANA LENSECTOMY/ CAPSULECTOMY
SEQUENTIAL PHACO COMBINED PHACO-PPV PARS PLANA LENSECTOMY/
AND PPV CAPSULECTOMY
Refractive Outcome Good if axial length can be Difficult to predict, especially in Worst; effective lens position
determined silicone oil and EMM, MH cases difficult to determine except for ACL
Difficulty Level Easiest Difficult (successful vitrectomy requires Moderately difficult
near perfect phaco outcomes to maintain
corneal clarity and pupillary dilation)
Best Suitable for Isolated macular pathology Nonelective cases with significant RDs with anterior PVR (stage D),
and elective cases cataracts (e.g., RD with proliferative markedly inflamed eyes
vitreoretinopathy [PVR] stage C or less)
IOL Considerations Usually not an issue except IOLs can be subluxated forward when Best to leave eye aphakic and only
cannot use silicone lenses gases are used, leading to IOL optic consider secondary IOL at a later
capture in pupil date
Surgical Equipment Usually not an issue Must use a vitrectomy machine with May require enlargement of one
combined phaco and vitrectomy 25-gauge sclerotomy to a 20-gauge
capabilities, otherwise needs two size for fragmenter in hard nuclei
separate machines
Cost Usually not an issue Significantly higher costs (viscoelastic, Only additional surgical cost is
IOL, time, surgical packs). May be cost marginal (the use of the fragmenter)
prohibitive in the ambulatory surgery
center setting

ACL, anterior chamber lens; EMM, epimacular membrane; MH, macular hole.

[IOP] maintenance, nonleaking wounds, rigid aspiration


SILICONE OIL ISSUES
system), which are equally essential for vitrectomy.
Phaco and vitrectomy are both safer and more effective
Silicone oil has a different index of refraction than vitreous.
when a closed system without leaks is utilized. The principal
Because the posterior surface of most IOLs is convex, instead
author has long recommended using 45 mm Hg, unless per-
of plano, the usual IOL power calculations cannot be used.
fusion pressure is compromised, just as Mackool advocates
Although many surgeons think silicone oil must be removed
elevated infusion pressure for phaco. Two-handed methods
after a certain number of months, this is simply not true. The
are best for both procedures both for optimal access and
incidence of silicone oil–related glaucoma is approximately
for surgical manipulation. A spatula, chopper, or nucleus
11% (1), silicone oil is not toxic to the retina, and corneal com-
manipulator is used to hold back the capsule and manipu-
plications are even less common. A not uncommon scenario is
late and position lens material similar to using the endoil-
PPV plus silicone oil to accomplish reattachment, removal in
luminator to hold back retina or peel vitreous with bimanual
several months followed immediately by redetachment, and
spreading technique.
reoperation with replacement of the silicone oil. There is sim-
High vacuum, a flared tip, and low flow (for sculpting
ply no need to remove oil contained behind a posterior cham-
the nucleus) promote efficiency without surge. This concept
ber lens; the refractive effect is approximately four diopters (D),
is the same as the principal author’s port-based flow-limiting
and contrary to popular belief, oil does not cause decreased
concept for PPV. The author developed linear (proportional)
vision. If there is a full fill behind a posterior chamber intraoc-
control of vacuum for vitrectomy, and this was subsequently
ular lens (PCL), change in focus with changing head position
applied to all phaco machines for aspiration control of both
as well as emulsification is minimized. IOL calculations must
flow and vacuum. Unfortunately, many PPV surgeons make
anticipate the ultimate retention or removal of oil. There is a
little use of linear control, fully depressing the pedal and
greater impetus to removal of oil in younger patients.
changing vacuum settings on the console. Mackool empha-
sizes avoiding position 2 (aspiration) or 3 (aspiration and
GENERAL PHACOEMULSIFICATION ultrasound) unless actually intending to remove material at
CONCEPTS that time. The surgeon may elect to engage the “continuous
irrigation” mode in the Constellation and Infinity machines,
Many key phaco concepts are similar to vitrectomy princi- which maintains constant irrigation despite foot pedal posi-
ples. Richard Mackool and others emphasize the importance tion and avoids unintended position 0 of the foot position.
of maintaining optimal visualization, frequent adjustment of The continuous irrigation mode can be toggled on and off
microscope focal plane, and stability (intraocular pressure when entering and exiting the eye to save irrigation fluid.
100 SECTION II ■ Surgical Technology and Techniques

There are also essential differences between phaco and repetition of the hydrodissection maneuver. Viscodissection
PPV; the phaco tip should be positioned near the center of is then accomplished by injecting Viscoat beneath the
the chamber and lens fragments mobilized to the phaco tip anterior capsule for 1,800 opposite the phaco incision. In
by the use of an appropriate aspiration flow rate to protect eyes with evidence of zonular laxity, viscodissection should
the capsule and iris. In marked contrast, the vitreous cutter be performed for 360 degrees.
should be brought to the vitreous; vitreous should not be The nucleus can now be rotated in order to verify that
drawn to the port by high vacuum. the hydrodissection or viscodissection has been successful;
however, this maneuver is not mandatory if the surgeon is
confident that separation has been successful.
GENERAL PHACOEMULSIFICATION A Mackool chopper or other chopper or spatula is
PRINCIPLES introduced through the sideport incision, and the phaco
tip is inserted through the primary incision without the
A small, tapered (reduced incision width), nonleaking need to fixate the latter with forceps. Viscoelastic material
sideport incision should be constructed. This can be done is aspirated from the surface of the lens in order to prevent
with the Alcon I-Knife. The entry should be created at a obstruction of the phaco tip by OVD. The nucleus is then
location that will be 2 to 2.5 clock hours from the phaco sculpted and divided by either chopping or other nuclear
incision prior to making the latter incision. The separation division methods. Mackool prefers to sculpt a central bowl
between the sideport and the keratome incision should be of approximately two phaco tips (2 mm) in width prior
refined to allow for surgeons “comfortable hand position.” to performing phaco chop, and he stabilizes the nucleus
The globe can be stabilized with an index finger placed during sculpting by placing the Mackool chopper over
against the nasal conjunctiva during the creation of these the edge of the equatorial nucleus opposite the phaco
incisions. incision.
For microincisional surgery, the temporal clear corneal Note that a nonleaking phaco incision is extremely
incision should be made with a 2.2-mm Alcon metallic ker- important to reduce intraocular turbulence and excessive
atome with a superior bevel (diamond knives can result in fluid flow through the eye. Rotation of instruments utilizing
inaccurate incision size if not inserted and removed at the the incisions as if they were an oarlock prevents distortion
exact same meridian). The initial entry should be made with of the cornea and compression of the phaco sleeve against
the blade at approximately 10-degree angle to the cornea. the vibrating tip (this reduces the risk of incision burn). The
After entering the cornea for 0.25 to 0.5 mm, the angle of authors prefer the Alcon 450 mini-flared tip with enhanced
the blade is lowered so that it is parallel to the cornea. When Ultrasleeve and torsional phacoemulsification.
the blade has penetrated the cornea so that the 2-mm mark Chopping of the nucleus is an excellent, efficient
on its anterior surface reaches the external incision, the tip of technique with the exception of nuclei that are elastic or
the blade is directed slightly downward (parallel to the iris) extremely dense (red-black nuclei). In the former case, high-
and the anterior chamber is entered. vacuum levels can be used to remove the peripheral lens
The anterior capsule should be stained with trypan blue, material until only a small central nuclear plate remains. The
and this can be done efficiently with one injection from a latter can then be elevated with a spatula and removed.
syringe that sequentially delivers air, trypan blue, and then Red-black nuclei are best divided into two heminuclei
BSS to remove as much of the trypan blue as possible from the by sculpting a long, very deep central trough prior to crack-
chamber prior to viscoelastic injection. Sequential injections ing with a spatula and phaco tip.
of Viscoat (anterior) and Provisc or other hyaluronic acid– Whenever possible, use both instruments to rotate the
based ophthalmic viscoelastic device (OVD) posterior to the nucleus and/or quadrants in order to reduce the amount of
Viscoat is done to replace the air with clear viscous material. stress placed on the lens zonules. Try to avoid the placement
The continuous curvilinear capsulorhexis (ccc) should of the phaco tip in the peripheral regions of the posterior
be initiated centrally with a bent needle, forceps, or cystotome. chamber, and after one or two nuclear quadrants have been
Capsulorhexis forceps (Mackool Microincisional Capsulor- removed, a spatula should be placed posterior to the phaco
hexis Forceps) are then used to create the ccc. It is important tip during subsequent nucleus removal in order to prevent
to regrasp the elevated flap at a position that is relatively close anterior movement of the lens capsule (infusion misdirec-
to the margin of the advancing tear during the creation of the tion syndrome) and possible capsule aspiration.
capsulorhexis. A 900 I/A tip with a small (0.25 mm) port permits
Hydrodissection using a flat 25-gauge cannula to ele- access to cortex for 360 degrees. In this manner, cortex can
vate the anterior capsule prior to injection of BSS results in be removed with the port of the I/A tip facing anteriorly.
the separation of lens material from the capsule (capsular- Note that the PPV can be performed prior to inser-
cortical cleavage). Hydrodissection is performed in at least tion of the IOL. This is highly desirable as a more uniform
two locations, usually 180 degrees apart. After each injec- view of the peripheral vitreous cavity is obtained when not
tion, the nucleus should shift forward and should be repo- looking through the edges of the optic after completion of
sitioned by gentle depression with the cannula prior to the vitrectomy. The only risk of delaying the introduction
CHAPTER 8 ■ Com bined Phacoem ulsification Vitrectom y Procedures 101

of the IOL is the possibility of inadvertent capsular tear vitrectomy procedure, a small posterior capsulectomy can be
during anterior vitrectomy, which is prevented by the performed with the vitreous cutter to improve visualization
presence of the IOL creating tension on the capsule and of the retina after the IOL is well positioned. The vitreous
diminishing its mobility. The capsular sac is then filled cutter is ideal for the construction of a posterior continuous
with Provisc prior to injecting the single-piece acrylic IOL curvilinear capsulorhexis. This rarely leads to problems other
via a Monarch injector and D cartridge. Do not remove than potential fogging of the IOL if air is used as vitreous
the viscoelastic until the haptics have released from the infusion or allowing silicone oil access to the AC. YAG laser
optic. If necessary, their release can be facilitated by using capsulotomy is ineffective in the presence of silicone oil.
a chopper to place traction on a haptic while the optic is Higher levels of postoperative inflammation are typical
stabilized with the I/A tip. After the haptics have opened, in phaco-vitrectomy cases. Very frequent dosing of topical
the viscoelastic can be aspirated with the 900 I/A tip, fol- steroids in the early postoperative period (e.g., every hour)
lowed by stromal hydration of both the primary and the with a rapid taper to qid after 1 or 2 weeks postoperatively
sideport incisions. often prevents inflammatory complications.
Even if the visual potential of an eye is poor, there are
still benefits of placing an IOL in an eye that will require
PHACO-VITRECTOMY TECHNIQUES long-term silicone oil retention is planned. The IOL can
form a barrier to the forward movement of the silicone and
Combined phacoemulsification with intraocular lens (IOL) prevent emulsification glaucoma and keratopathy. In addi-
implantation and vitrectomy is a safe technique when used tion, aphakic eyes with long-term silicone oil fill contain-
in the appropriate patients (2). The threshold for the use of ing an inferior RD may develop neovascular glaucoma and
iris expander is lower than for standard phaco. For example, require Ahmed valve placement. In these eyes, if the eye is
a case with a pupil size of 4 mm or less may be done without aphakic, the silicone oil can exit the vitreous cavity through
problems as standard phaco since anterior chamber infu- the valve into the subconjunctival space, causing an inad-
sion during phaco maintains pupillary size but may develop equate intraocular oil fill that, in turn, causes progression of
marked miosis when the anterior chamber infusion is ter- the previously contained RD.
minated at the end of the phaco. Early use of iris expanders
prevents severe miosis and a very difficult vitrectomy. If iris
expanders are used, do not remove until the completion of PHACOEMULSIFICATION PRINCIPLES
the vitrectomy. FOR SILICONE OIL–FILLED EYES
Iris prolapse through the cataract incision leads to mio-
sis during vitrectomy. Keratome wounds should be made 2 × Because silicone oil “floats” over aqueous, oil will frequently
2 mm, which is longer than some phaco surgeons use, in push forward on the posterior lens capsule causing something
order to prevent iris prolapse. similar to “positive pressure” during phaco. This is likely the
Staining of the anterior lens capsule with trypan blue is main cause of complications during the procedure. A tight seal
usually required. The red reflex can often be poor in cases of the cataract incision around the phaco probe significantly
that need combined phaco-vitrectomy approach, usually decreases turbulence and intraoperative incision leaks and sta-
from coexisting vitreous hemorrhage or RD. The need for bilizes capsular position and chamber depth. The height of the
capsular staining may be underestimated if the only criterion infusion bottle with gravity-fed infusion should be elevated to
used is the appearance of the lens. Since adequate continu- the maximum to maintain lens capsule inflation during the
ous curvilinear capsulorhexis (CCC) is critical in combined case. If a small bubble of silicone oil enters the AC through
phaco-vitrectomy approach, it is best to use trypan blue intact zonules, it is best to ignore it. All cortical fragments must
staining in the majority of cases. be removed without damaging the capsule. In standard phaco,
Viscoelastic can be left in after phaco to maintain cham- one may leave small cortical fragments without problems. If
ber depth and pupil size. In the setting of combined vit- a cortical fragment is left in a silicone oil case, it may not be
rectomy, IOP elevation in the early postoperative period is reabsorbed as expected and can cause inflammation and oil
rare. If silicone oil is to be used at the end of the vitrectomy, emulsification. It is best to use bimanual I/A or angulated, that
the surgeon should remove the viscoelastic from the ante- is, 90-degree, I/A tip to remove subincisional cortex. If a small
rior chamber immediately before the oil fill, and it should be posterior capsular tear occurs during the last phase of the I/A,
replaced with air. This prevents oil emulsification from the immediate introduction of an OVD or IOL can seal the hole and
viscoelastic interaction. prevent further oil bubbles from coming into the AC. If a large
The cataract incision should be sutured with 10-0 nylon capsular tear is encountered, insertion of a multipiece acrylic
in all phaco-vitrectomy cases. This prevents failure of the IOL into the ciliary sulcus and capture of the lens optic through
self-sealing clear corneal incision mechanism if hypotony the capsulorhexis can be done to isolate the AC from the oil; if
occurs during the vitrectomy or postoperatively. this cannot be done, it may be best to avoid IOL placement and
If the posterior lens capsule is not clear at the end of proceed with total capsular removal to prevent leaks of oil into
the phaco and this interferes with visualization during the the AC that can lead to silicone oil glaucoma and keratopathy.
102 SECTION II ■ Surgical Technology and Techniques

Fig u re 8 .1 ■ A layer of aq ue-


ous wedged between the IOL and
the silicone oil can create m ultiple
refractive surfaces and p rism atic
effects that can b e im p ossib le to
predict or correct with an IOL.

silicone oil interface). This neutralizes the refractive interface


ISSUES REGARDING INTRAOCULAR
between IOL and silicone and places all the refractive power
LENS CALCULATIONS FOR SILICONE on the front surface of the IOL. This design may be difficult
OIL–FILLED EYES to obtain and is usually made of polymethyl methacrylate
(PMMA) and is not available with modern, foldable acrylic
Silicone oil impedes ultrasound A-scan measurements by material. Another source of error in refractive outcomes in
slowing of ultrasound waves and decreasing the intensity of silicone oil eyes relates to the completeness of the oil fill. If
the posterior spike. If media clarity allows measurements to there is any degree of underfill, a layer of aqueous can wedge
be made with the IOL Master (which utilizes laser optical between the IOL and the silicone oil (Fig. 8.1). This creates
measurements instead of ultrasound), this is the best option. multiple refractive surfaces and prismatic effects that can be
Otherwise, calculations based on the contralateral eye mea- impossible to predict or correct with an IOL. If the goal is
surements, prior refractions, etc. must be used. Therefore, long-term fill with silicone oil, the IOL can be calculated for
do biometry with IOL Master soon after initial surgery with an emmetropic or mildly myopic result. If the goal is even-
silicone prior to the onset of cataract and other possible reti- tual removal of the silicone oil, placement of an IOL that is
nal complications such as pucker or recurrent RD, which appropriate for the eye after oil removal should be done.
may affect measurement. The majority of the cases that require silicone oil and pha-
Because the refractive index of silicone oil is higher than coemulsification with IOL placement are incapable of achiev-
water, the refractive power of the IOL/silicone interface is ing excellent acuity due to the underlying retinal pathology;
lower than the refractive power of the IOL/vitreous interface there is normally no need to achieve a precise refractive result.
(the refractive power is proportional to the ratio between In the event of major refractive surprises or if the visual acu-
refractive indexes on both sides of the refractive interface, ity happens to be very good, the IOL can always be replaced
based on Snell’s law). In effect, silicone oil diminishes the within the first month or two after implantation.
power of the IOL, creating a hyperopic outcome. The higher
the lens power of the IOL placed, the higher the total loss of
its power in a silicone oil–filled eye, since the difference is References
based on a percentage loss of power. For this reason, highly 1. Al-Jazzaf AM, Netland PA, Charles SM. Incidence and manage-
myopic eyes tend to be closer to emmetropia than hyperopic ment of elevated intraocular pressure after silicone oil injection.
eyes when IOLs are placed in silicone oil–filled eyes, because J Glaucoma 2005;14:40–46.
2. Hwang JU, Yoon YH, Kim DS, et al. Combined phacoemulsi-
they require lower power IOLs. Theoretically, the IOL design fication, foldable intraocular lens implantation, and 25-gauge
that provides the most reliable refractive results in silicone transconjunctival sutureless vitrectomy. J Cataract Refract Surg
oil–filled eyes is a planoconvex lens (the plano side faces the 2006;32:727–731.
C H AP TER
9
25-GAUGE VITRECTOMY

Sutureless, transconjunctival 25-gauge vitrectomy Patients with prior or anticipated glaucoma-filtering


was introduced by DeJuan and colleagues (1,2) procedures are ideal candidates for 25-gauge surgery.
and has rapidly become an accepted tool in the Patients with severe dry eyes, ocular surface disorders, and
vitreoretinal surgery armamentarium. Suture- scarred conjunctiva are excellent candidates for 25-gauge
less vitrectomy is dependent on two concepts: (a) smaller surgery as well.
diameter instruments and therefore smaller sclerotomies and
(b) conjunctival displacement before making the transcon-
junctival sclerotomies so that the conjunctiva covers the scle- TROCAR-CANNULA SYSTEM
rotomy after surgery is completed. Trocars are used to place
flexible, thin-wall cannulas, resulting in 23.5-gauge scleroto- The purpose of the trocar is to make a 25-gauge sclerotomy
mies. The cannulas are used to maintain alignment between and enable simultaneous insertion of flexible 23.5-gauge self-
the offset conjunctival incision and the sclerotomy and facili- retaining cannula that fits over the trocar (Fig. 9.1). The con-
tate finding the small incisions when inserting tools. junctival incision is intentionally displaced from the scleral
incision so that the two incisions will not be aligned after
cannula withdrawal and the conjunctiva will cover the scle-
CASE SELECTION rotomy (Fig. 9.2). Repeated insertion and withdrawal of tools
is accomplished through the cannula, which maintains the
Initially it was thought that 25-gauge, sutureless vitrectomy alignment of the conjunctival and scleral incisions and pro-
was only indicated for epimacular membranes, macular holes, tects the wounds. The conjunctiva should be displaced using a
vitreomacular traction, and surgery for retinal venous occlu- cotton-tip applicator by gently moving it anteriorly or circum-
sion. Early on, many surgeons thought 25-gauge surgery was ferentially or some combination thereof (Fig. 9.3). An attempt
inappropriate for vitreous hemorrhages, rhegmatogenous should be made to avoid conjunctival and scleral vessels
retinal detachments, proliferative vitreoretinopathy (PVR), to reduce postoperative subconjunctival hemorrhages. The
diabetic traction retinal detachments, or giant breaks. Experi- Alcon noncoring trocar is based on a modified microvitreoret-
ence has shown, however, that 25-gauge surgery is ideal for inal (MVR) blade and requires much less insertion force than
vitreous hemorrhages, rhegmatogenous retinal detachments, competitive hypodermic needle-based, coring-type designs.
PVR, and giant breaks and is applicable for diabetic traction A prospective, consecutive clinical series by the author shows
retinal detachments as well. Few surgeons now believe that that the Alcon system using fluid-air exchange demonstrates
branch vein decompression (sheathotomy) and radial optic significantly less hypotony than published results with the
neurotomy (RON) are effective procedures, making 25-gauge coring-type trocar. The Alcon trocar cannula usually requires
surgery for these procedures a moot point except in the con- no rotation when inserted. If some resistance is encountered,
text of vitrectomy alone for the reduction of macular edema small-amplitude back and forth rotation will facilitate inser-
via vascular endothelial growth factor (VEGF) reduction in the tion. The cotton-tip applicator should be handed back to the
macula and increased oxygen tension in the vitreous cavity. scrub technician or dropped so that the forefinger of this hand
Dislocated lens material (dropped nucleus) and removal can be used to guide the trocar cannula during insertion. The
of intraocular foreign bodies (IOFBs) require one 20-gauge trocar should be aimed toward a virtual point about 2 mm
sclerotomy because dense membranes have too much flow anterior to the center of the eye or about 2 mm posterior to
resistance for small-lumen instruments and IOFBs are too the lens. The incisions should be made 4 mm posterior to the
large. The technique of combining two 25-gauge suture- limbus (Fig. 9.4) unless there is a pars plana abnormality from
less incisions with one 20-gauge sutured incision has been retinopathy of prematurity (ROP), trauma, or pars planitis; a
described as 20/25 vitrectomy by the authors. large choroidal detachment; suprachoroidal hemorrhage; or
103
104 SECTION II ■ Surgical Technology and Techniques

Fig ure 9 .1 ■ A 25-gauge trocar cannula from Alcon.

high retinal detachment that would necessitate making the


FLUIDICS
incisions through the pars plicata. The 25-gauge incisions
may be made 3 mm posterior to the limbus in aphakic eyes.
Ohm’s law for fluids is directly analogous to Ohm’s law for
The inferotemporal incision should be made just below the
electricity and teaches that pressure (gradient) is equal to the
3 or 9 o’clock position to reduce bleeding and pain but as far
resistance times the flow. Resistance to flow is proportional
as possible from the lower lid so that it will not be displaced if
to the fourth power of the inner diameter of the lumen. The
the eye is rotated inferiorly. The superonasal incision is usually
much higher resistance of 25-gauge cutters and infusion
made on a virtual line from the lowest point of the bridge of
cannulas was initially thought by many to be a major dis-
the nose extending through the center of the pupil and then
advantage. It turns out that increasing infusion pressure to
plugged with a special plug made for 25-gauge cannulas. This
50 to 60 mm Hg while flow is occurring (dynamic state) and
location reduces tool flexion issues and facilitates peripheral
lowering it to 35 to 45 mm Hg (static state) when using for-
and anterior access. The purpose of the plug is to prevent vit-
ceps, pics, scissors, or the endophotocoagulator (static state)
reous prolapse or fluid loss through the port while making the
solves the problem. The Alcon Constellation pressurized
third incision. The superotemporal incision is usually made on
infusion system and the Accurus Vented Gas Forced Infusion
a virtual line extending from the lowest point of the supraor-
(VGFI) allow rapid and accurate switching between static
bital rim through the center of the pupil again to reduce tool
and dynamic infusion pressures, and the Constellation pres-
flexion and facilitate anterior and peripheral access.
sure compensation system directly addresses this problem.

Fig u re 9 .2 ■ Wound leaks are p revented b y conjunctival disp lacem ent with resp ect to the
0.5-m m scleral incision.
CHAPTER 9 ■ 25-Gauge Vitrectom y 105

Fig u re 9 .3 ■ Circu m feren tial


or anterior conjunctival disp lace-
m ent with a cotton-tip actuator
is done before entry of the trocar
cannula.

The 23/25/27-gauge cutters have more resistance than advantageous because of port-based flow limiting, which
20-gauge cutters because of the smaller bore coaxial inner results in decreased pulsatile vitreoretinal traction. The
needle as well as the cutter intermittently closing the port smaller bore of the 23/25/27-gauge cutters also produces
during the open-close cycle, thus requiring greater com- port-based flow limiting (Fig. 9.5). The 25-gauge cutter pro-
pensation with respect to the vacuum settings used with duces one half the pulse flow of the 2,500-cpm 20-gauge cut-
20-gauge vitrectomy. The authors use 650 mm Hg maxi- ters and therefore has 2× greater fluidic stability. In addition,
mum setting and proportional (linear) suction and the Con- port-based flow limiting reduces surge when dense epireti-
stellation system. Fast cutting with 20-gauge cutters is also nal membrane (ERM) suddenly deforms through the port

Fig u re 9 . 4 ■ In fero t em p o ral


trocar-cannula entry should be 3.5
m m p osterior to the lim b us just
inferior to the 3 or 9 o’clock p osi-
tion. Sup eronasal trocar-cannula
entry should b e m ad e 3.5 m m
p osterior to the lim bus on a vir-
tual line extending from the low-
est point of the bridge of the nose
through the center of the p up il.
Sup erotem p oral trocar-can n ula
entry should b e m ad e 3.5 m m
p osterior to the lim bus on a vir-
tual line extending from the low-
est p oint of the sup raorb ital rim
through the center of the pupil.
106 SECTION II ■ Surgical Technology and Techniques

Fig ure 9 .5 ■ The sm aller lum en


of a 25-g aug e cutter p rod uces
p ort-b ased flow lim iting like fast
cuttin g , wh ich in creases fluid ic
stab ility an d red uces surg e an d
iatrog en ic b reaks after sud d en
elastic d eform ation of the ERM
through the port.

analogous to occlusion break surge in phaco procedures. Fast Multifunction tools such as the end-aspirating laser
cutting has the additional advantage of reducing vitreoretinal probe (Chang) and the disposable bipolar endoilluminator
traction by minimizing collagen fiber travel through the port. are not available in a 25-gauge diameter.
The authors recommend using a cutting rate of 5,000 cpm
To o l Fle xio n
for all 25-gauge tasks and cases and have found that core
vitrectomy times are very reasonable when using 650 mm Tool flexion is the most significant complaint about 25-gauge
Hg vacuum. systems, but this problem can be overcome using certain tech-
niques. The second-generation Alcon endoilluminators and
endolaser probes are 58% stiffer than the first-generation tools.
CUTTER DESIGN OPTIONS Careful positioning of the sclerotomies reduces tool flexion.
The superonasal incision should be placed aligned with the
Pneumatic disposable cutters weigh about 1/14 as much as lowest point on the bridge of the nose. The superotemporal
available electric cutters, thereby increasing dexterity, reduc-
ing fatigue, and reducing flexion of the shaft. Pneumatic
cutters vary (optimize) the duty cycle as a function of cut-
ting rate, which is an advantage compared to the fixed duty
cycle of electric cutters, because of the wide range of materi-
als properties encountered during vitrectomy (from air to
perfluorooctane to vitreous to dense ERM).

TOOL ISSUES

Disposable forceps and scissors are advantageous for 25-gauge


surgery (3). The advantages of disposable 25-gauge instru-
ments include elimination of sterilization and cleaning issues
(Toxic Anterior Segment Syndrome (TASS)-like inflammation
from retained biological material and enzymes from ultrasonic
instrument cleaning) and the inevitable damage that occurs Fig ure 9 .6 ■ Disposable 25-gauge Internal Lim iting Mem brane
to fragile reusable instruments. The authors use Alcon DSP forceps from Alcon are used to p eel ERMs, p osterior vitreous cor-
disposable forceps (Fig. 9.6) and scissors (Fig. 9.7). tex, and internal lim iting m em brane (ILM).
CHAPTER 9 ■ 25-Gauge Vitrectom y 107

maintain the eye on the primary position or close to the


primary position with proper rotation of the instruments
around the pivot point of the sclerotomy and cannula. Contact
wide-angle lenses decrease the need for extreme eye rotation.
Scleral depression by the scrub technician or assistant can
provide peripheral access without probe-induced ocular rota-
tion. Contact-based wide-angle visualization systems (Volk,
AVI) require the surgeon to maintain the eye near the primary
position and rotating the instruments around the sclerotomies
instead of using translational forces to rotate the eye.
Using a 23-gauge approach reduces tool flexion but raises
significant questions about wound leaks, which demands
more challenging wound construction and leak issues.

Fig ure 9 .7 ■ Disposable 25-gauge curved scissors from Alcon COMBINED 25-GAUGE AND
are used for segm entation and delam ination. 20-GAUGE VITRECTOMY

Certain limitations of 25-gauge vitrectomy can be over-


incision should be placed at a low point on the brow, typically come by enlarging one port to 20 gauge for certain tasks
just above the 3 or 9 o’clock meridian. The inferotemporal (4). One sclerotomy can be enlarged for fragmenter use for
incision should be placed just below the 3 or 9 o’clock merid- lensectomy (Fig. 9.8) or removal of dislocated lens material
ian. The patient’s head can be rotated toward the target pathol- (Fig. 9.9), or use of the Machemer-Parel diamond-coated for-
ogy during surgery to better view the periphery. The patient ceps for intraocular foreign body removal. The technique var-
can cooperate when using local anesthesia and the anesthesi- ies depending on the indications previously described, but in
ologist can help when general anesthesia is used. general, an 8-0 nylon sclerotomy suture cut on the knot was
A significant part of the tool flexion experienced by used for the 20-gauge sclerotomy and an 8-0 Biosorb conjunc-
surgeons is technique dependent. Proper technique should tival suture cut on the knot was used at the inflection point

Fig ure 9 .8 ■ A 20/ 25 endocapsular lensectom y requires higher infusion pressure and judicious use of
linear suction to prevent sudden hypotony.
108 SECTION II ■ Surgical Technology and Techniques

Fig ure 9 .9 ■ A 20/ 25 rem oval


of d islocated len s m aterial m ust
be done after perform ing a com -
p lete vitrecto m y. A 2 5 -g au g e
infusion port requires higher infu-
sion pressure and judicious use of
linear suction to p revent sudden
hyp otony with a 20-g auge frag -
m enter.

of an L-shaped mini-conjunctival flap. Utilizing one 20-gauge or certain illuminated instruments for the so-called bimanual
sclerotomy with two 25-gauge sclerotomies to address the surgery, although the authors do not use these methods. Use
mechanical properties of dense lens material or the physical of 20-gauge fragmenter with 25-gauge infusion requires eleva-
size of IOFBs (Fig. 9.10) was found to be safe and effective. The tion of the infusion pressure and proper technique with the
20/25 technique can also be used for 20-gauge conformal for- fragmenter, avoiding aspiration when there is no lens frag-
ceps, end-gripping diamond-coated forceps, the Chang end- ment occluding the tip of the instrument. This prevents ocular
aspirating laser probe, the disposable bipolar endoilluminator, collapse from the mismatch of inflow and outflow of fluid.

Fig ure 9 .1 0 ■ A 20/ 25 rem oval


of IOFB with Machem er-Parel dia-
m ond-coated forceps from Alcon
Grieshaber should be done after
com p lete vitrectom y to red uce
vitreoretinal traction.
CHAPTER 9 ■ 25-Gauge Vitrectom y 109

Wound leaks can lead to hypotony, resulting in choroidal


VISUALIZATION
detachments, bleeding, vitreous volume enhancement, or
reoperations to suture the wounds. Postoperative vitreous
Smaller diameter fibers can increase light losses, which can
wicks can occur with wound leaks and increase the risk
be compensated for by using one of the new xenon sources,
of endophthalmitis. Intraoperative wound leaks through
such as the Accurus Xenon Illuminator. The Accurus Xenon
the cannula can result in bleeding, vitreous prolapse with
Illuminator has ISO standard filtering to produce a safe
secondary vitreoretinal traction, miosis due to hypotony, or
white-appearing light with low aphakic hazard function
even retinal prolapse. The author initially raised the issue
(AHF). The second-generation endoilluminators have twice
of conjunctival antibiotics gaining access to the vitreous
as much light throughput, have much larger divergence
cavity through unsutured sclerotomies, resulting in retinal
angle, and are twice as stiff. The Alcon Constellation Vision
toxicity, and anecdotal reports have substantiated this con-
System incorporates dual two-port xenon illuminators that
cern. De Juan has advocated allowing vitreous to plug the
are more efficient than first-generation xenon systems and
wounds, while the author introduced the idea of using flu-
have longer lamp life.
id-air exchange to reduce wound leaks. Initially the author
recommended partial fluid-air exchange, but this left the
inferotemporal sclerotomy unprotected when the patient
WOUND LEAK ISSUES was seated or standing; total fluid-air exchange (Fig. 9.12)
was later used for all cases not requiring gas or silicone oil
Postoperative wound leaks have always created the if there was a concern about wound leaks. Proper oblique
greatest concern about sutureless, 23/25-gauge technol- sclera wound construction has decreased the need for fluid-
ogy. Oblique wound construction produces a scleral tun- air exchanges, and the authors currently do this technique
nel that is self-closing when the intraocular pressure is only rarely.
normal (Fig. 9.11); turning the infusion off at the end of Wound leaks are more common in high myopes,
the case makes no sense. Repositioning the conjunctiva Marfan’s syndrome, and other patients with connective tis-
with a cotton-tip applicator after cannula removal is essen- sue disorders and thin sclera. Many surgeons have noted
tial but not sufficient; firm pressure should be applied with the role of vitreous incarceration in 20-gauge wounds as
the closed forceps over the scleral tunnel created by oblique a factor in postvitrectomy retinal detachment. The senior
wound construction to close the outer wall of the tunnel. author has emphasized that the surface tension effect of

Fig ure 9 .1 1 ■ Obliq ue wound construction p roduces a scleral tunnel, which is self-closing when the
intraocular pressure is norm al.
110 SECTION II ■ Surgical Technology and Techniques

Fig ure 9 .1 2 ■ Fluid-air exchange can be utilized at the end of cases not requiring g as or silicone oil,
if there is a concern about wound leaks.

Fig ure 9 .1 3 ■ The conjunctiva m ust b e rep ositioned after the cannulas are rem oved so that the
conjunctival incision is not overlying the sclerotom y. The sclera should be sup ported while slowly with-
drawing the cannulas to prevent air, gas, or silicone loss and vitreous prolapse.
CHAPTER 9 ■ 25-Gauge Vitrectom y 111

an air bubble can reduce or eliminate wound leaks and


CANNULA WITHDRAWAL
prevent vitreous wicks until fibrin seals the wound just as
gas bubbles are used to “seal” retinal breaks and macular
AND WOUND CLOSURE
holes. The author has long suggested that an air bubble in
Slow withdrawal of tools from cannulas reduces acute
the anterior chamber is the best way to keep vitreous out
vitreoretinal traction; think of the tool in the cannula as
of the anterior chamber and away from the wounds after
a syringe with plunger being pulled back. Supporting the
rupture of the posterior capsule in cataract surgery. Air is
sclera while removing the cannulas reduces wound eversion,
also an excellent way to intraoperatively seal wound leaks
pressure on the globe, and the likelihood of vitreous pro-
in trauma cases. The authors recommend removing as
lapse. The wounds should not be massaged after cannula
much peripheral vitreous as possible in 20-gauge as well as
removal; the conjunctiva should be gently moved back (Fig.
23/25-gauge cases.
9.13) into its original position. If a wound leak is detected, a
single 8-0 Biosorb suture can be placed through conjunctiva
and sclera to close these small wounds.
POSTOPERATIVE ENDOPHTHALMITIS
AND RETINAL DETACHMENTS
SUMMARY
Several surgeons have been concerned about a possible
increased risk of postoperative endophthalmitis and retinal 25-gauge, sutureless vitrectomy requires modification of tech-
detachments (5) after 25-gauge vitrectomy. The rationale for niques and settings but offers the advantage of better fluidic sta-
postoperative endophthalmitis would be postoperative entry bility, faster visual improvement, much better patient comfort,
of bacteria into the globe through unsutured wounds. Since much less conjunctival damage, and, less importantly, tedium.
the authors have started performing 25-gauge vitrectomy as The time saved by not having to suture an infusion cannula in
the standard surgery in their practice, they have only had place, make or close conjunctival incisions, and suture sclero-
one case of postoperative endophthalmitis in over 700 cases. tomies is largely offset by taking more time for the vitrectomy.
This anecdotal case is not sufficient for them to presume an
increased risk of endophthalmitis in their practice. In terms References
of prophylaxis, all patients undergoing 25-gauge vitrectomy
have the standard meticulous prepping with povidone-iodine 1. Fujii GY, De Juan E Jr, Humayun MS, et al. Initial experience
using the transconjunctival sutureless vitrectomy system for vit-
as well as postoperative injection of subconjunctival antibi- reoretinal surgery. Ophthalmology 2002;109(10):1814–1820.
otics. In addition, patients are instructed to remove the eye 2. Fujii GY, De Juan E Jr, Humayun MS, et al. A new 25-gauge
patch upon returning home from surgery and start topical instrument system for transconjunctival sutureless vitrectomy
antibiotics every hour while awake immediately postopera- surgery. Ophthalmology 2002;109(10):1807–1812.
tively, attempting to maintain significant antibiotic concen- 3. Capone A. 25 gauge pars plana vitrectomy: advances in instru-
mentation and broader indications. Program and abstracts of the
trations in the ocular surface to decrease the conjunctival American Society of Retina Specialists 23rd Annual Meeting; July
bacterial load that may migrate through unsutured wounds. 16–20, 2005, Montreal, Canada.
The authors have not noticed an increase of postopera- 4. Charles ST. 20/25 vitrectomy: combined 25 and 20-gauge vitrec-
tive retinal detachments after 25-gauge vitrectomies in their tomy. Program and abstracts of the American Society of Retina Spe-
practice. Previous mentions of post–25-gauge vitrectomy cialists 23rd Annual Meeting, July 16–20, 2005, Montreal, Canada.
5. Williams DF. The incidence of rhegmatogenous retinal detach-
retinal detachments may be related to the surgical learning ment (RRD) following 20 g versus 25 g vitrectomy. Program
curve associated with a new procedure rather than to a flaw and abstracts of the American Society of Retina Specialists 23rd
intrinsic to the equipment and techniques. Annual Meeting, July 16–20, 2005, Montreal, Canada.
C H AP TER
10
20-GAUGE SUTURED WOUND
CONSTRUCTION AND CLOSURE

Whereas 25-gauge vitrectomy is usually risk of exposure of scleral sutures and make visualization of
performed transconjunctivally with trocar- the sclerotomies difficult because of the anterior flap.
cannula systems, 20-gauge vitrectomy usually
requires conjunctival incision, exposure of TRACTION SUTURES
the anterior sclera, and sclera closure and conjunctival
reapproximation with sutures. Despite performing nearly all Traction sutures under the muscles are of no value in vitrec-
of their vitrectomies with 25-gauge techniques, the authors tomy because the instruments move the eye. Only if scleral
still find the need for creation of 20-gauge incisions. These buckling is anticipated should traction sutures be used, and
may be required, for example, to introduce a 20-gauge then they should be placed beneath the muscles directly,
fragmenter handpiece to remove pieces of dropped dense using a short-handled fenestrated muscle hook with cham-
nucleus in the vitreous or to introduce diamond-coated 20- fered hole. Large sutures (0 to 2-0 silk or cotton) do not cut
gauge forceps to remove intraocular foreign bodies. This the muscle tendons and provide a good grip for the assis-
chapter will focus on techniques of creation and closure of tant. Only the muscles absolutely required for access to the
20-gauge incisions. sclera should be trapped to reduce postoperative pain, pto-
sis, and strabismus. The superior rectus should be avoided
if possible to avoid or reduce vertical strabismus as well as
CONJUNCTIVAL INCISIONS
ptosis secondary to levator and Mueller’s muscle damage. It
is never necessary to remove muscles for scleral buckling or
Care should be taken to avoid highly scarred or vascular
trauma repair in the author’s experience.
areas, filtering blebs, setons, and flaps for sutured intraoc-
ular lenses (IOLs). Two temporal incisions are made, one
superotemporal for the cutter, scissors, forceps, fragmenter, 20-GAUGE SCLERAL INCISIONS
laser, etc. and another inferotemporal for the infusion can-
nula. They should be limbus-parallel, 1 mm posterior to the Sclerotomies for the endosurgical instruments will be focal
limbus, and at the superior and inferior border of the lateral points for the remainder of the operation (1). If they are
rectus. The superonasal incision is usually for the endoil- incorrectly placed or constructed, the entire course of the
luminator and should be limbus-parallel, 1 mm posterior operation can be adversely influenced. The incisions should
to the limbus, and centered at the superior border of the be placed in the anterior pars plana to reduce the risk of
medial rectus. A small radial extension of the limbus-parallel damage to the peripheral retina. Contrary to customary
incisions at one end resulting in an L-shaped flap facilitates teaching, bleeding of the ciliary body is infrequent and rarely
better exposure and single suture closure. serious when it does occur. Except in children, patients with
For any vitreous surgery in which scleral buckling is an abnormal pars plana, and microphthalmic patients, the
anticipated, it is best to make a continuous incision 1 mm sclerotomies should be placed 3 mm posterior to the limbus
posterior to the limbus including each quadrant requiring if the lens is absent or to be removed and 4 mm posterior
buckling. A limbal peritomy tends to bleed under the con- if the lens is to be retained (Fig. 10.1). Some surgeons use
tact lens, causes conjunctival retraction and redundancy 3.5 mm for all cases as the authors do for 25-gauge cases.
problems in the postoperative course, and makes postopera- The authors use 25-gauge, sutureless, transconjunctival sur-
tive contact lens fitting difficult and dellen more common. gery for almost all cases; the following techniques apply only
Incisions more than 1 mm behind the limbus increase the to sutured 20-gauge surgery.
112
CHAPTER 10 ■ 20-Gaug e Sutured Wound Construction and Closure 113

Fig ure 1 0 .1 ■ All sclerotom ies are m ad e with MVR b lad e, 3 m m p osterior to the lim b us if
aphakic or lens is to be rem oved and 4 m m p osterior to the lim bus if lens is to be retained.

The disposable microvitreoretinal (MVR) blade is ideal is unnecessary because the tip of a high-quality MVR blade is
for making 20-gauge sclerotomies. The blade is lancet shaped typically as sharp as a 30-gauge needle and provides a correct
to ensure that the incision is centered at the initial entry point size incision if a subsequent fluid injection is required.
(Fig. 10.2). The blade is 1.4 mm in width, which makes a lin- The sharpness of a new MVR blade usually prevents
ear incision that becomes 0.89 mm (20 gauge) in diameter the nonpigmented pars plana epithelium and choroid from
when deformed to a circular shape by the instruments. The being pushed into the eye without penetration. The pos-
nontapered, 20-gauge shank and 25-mm length correspond sibility of penetrating the sclera but not the choroid or pars
to the dimensions of the other endosurgical instruments. plana epithelium should be a reminder to use these blades
Stilettos, myringotomy blades, illuminated stab needles, for one case only. To verify proper penetration, the blade
20-gauge needles, and transilluminators are unnecessary should be seen through the operating microscope. Under
with this method. Prefirming prior to use of the MVR blade no circumstances should the naked eye be used to check
the cannula nor should the 20-gauge cannula be used for
infusion without inspection (although this is commonly
done with 25-gauge infusion cannulas). If the eye is soft, a
20-gauge needle should then be placed through the initial
MVR incision and infusion fluid injected with the needle
tip visible. This step will decrease the chances of supra-
choroidal and subretinal infusion. If excessive bleeding
through the sclerotomy occurs, it can be treated by exter-
nal-internal bipolar diathermy using a scleral plug as the
internal contact or by placing one blade of the bipolar in
the sclerotomy and the other on the scleral surface adjacent
to the bleeder.

20-GAUGE INFUSION CANNULA


PLACEMENT
Fig ure 1 0 .2 ■ MVR blade has a lancet tip that m akes a sym -
m etrical incision around the initial entry p oint, 1.4 m m b lad e Unless there is severe fibrovascular proliferation or some
width to m ake a 0.89-m m incision for 20-g aug e tools; shaft is other obstruction discussed above at this region of the pars
20 gauge to m ake a round opening from the linear incision. plana, the infusion cannula is placed near the inferior border
114 SECTION II ■ Surgical Technology and Techniques

of the lateral rectus. In this position, it does not hit the nose, condensing lens can be used for visualization. To see the cannula,
lids, or speculum or interfere with the surgical instruments. it is usually necessary to push it gently toward the center of the
Because it decreases the incidence of choroidal infusion eye, being careful to avoid the lens, if it is a phakic eye. If the tip
and directs infusion fluid further away from the lens than appears shiny and clear of overlying tissue, then infusion may
does the 2-mm cannula, the 4-mm cannula should be used be turned on. Observing the cannula with the unaided eye and
in all cases. Unless it is carelessly directed anteriorly, the endoilluminator has insufficient magnification and can result in
4-mm cannula will not damage the lens. A 6-mm cannula inadvertent suprachoroidal or subretinal infusion.
can be used for thick choroid cases but can bump the lens. If tissue is seen over the cannula, the corrective action
The authors make the sclerotomy for the infusion cannula depends on the status of the lens (Fig. 10.3). If the eye is
before placing the cannula retention suture. By placing the inci- aphakic or will be at the end of the operation, the MVR blade
sion before the suture, each edge of the wound can be grasped is inserted from the opposite side of the eye to incise the tis-
with 0.12 forceps and stabilized during suture passage. The sue over the cannula internally, so that it will retract down
tension applied to the sclera makes possible a deep suture bite over the entire interior portion of the cannula. This tech-
and wound eversion without opening the deepest part of the nique is also effective if a choroidal infusion is present from
incision can be used to judge scleral thickness. The suture bites previous problems or occurs during surgery from disloca-
should be at least three-fourth scleral depth and 1 mm long so tion of the cannula. If the eye is to remain phakic, the can-
that they will not fall off the tabs of the infusion cannula. They nula should be removed and a 20-gauge needle inserted to
should be widely spaced to ensure imbrication of the cannula repressurize the globe and compress the choroid against the
into the sclera, similar to a buckle. A braided, flexible suture sclera before reinserting the cannula.
(6-0 silk) is easier to place over the cannula footplates than is a If the pars plana is not visible preoperatively, the pars
springy nylon or polypropylene suture. Some surgeons use the plana infusion cannula cannot be turned on at this point.
cannula retention suture for wound closure. The authors are Infusion should begin using an angulated 30-degree,
opposed to this method because it compromises imbrication of 20-gauge blunt infusion cannula placed through the supra-
the cannula into the sclera as well as tight wound closure. Vicryl nasal sclerotomy (Fig. 10.4). If the lens is absent or is to
sutures fray when used for infusion cannula retention and cause be removed, the cannula can be visualized by indenting the
significant and long-lasting inflammation (Vicryl scleritis). cannula toward the pupil. When the anterior segment opac-
After insertion, the cannula should always be inspected ity has been removed facilitating a view of the cannula, the
with the operating microscope before the infusion is turned surgeon can then turn on the infusion cannula before pro-
on. If the pupil is very small, the indirect ophthalmoscope and ceeding with the rest of the vitrectomy.

Fig ure 1 0 .3 ■ If tissue is seen over infusion cannula, it should be incised with MVR blade. The
blade is inserted from the opposite side of the eye if the eye is aphakic or lens is to be rem oved
and from the sam e sid e if the lens is to be retained.
CHAPTER 10 ■ 20-Gaug e Sutured Wound Construction and Closure 115

Fig ure 1 0 .4 ■ If the infusion cannula cannot be seen with the operating m icroscope, infusion
is initiated with the blunt, 20-gauge, 30-degree angulated infusion cannula. Infusion is changed
to the sew-in cannula after the anterior opacities are rem oved and the cannula becom es visible.

Basics o f In fusio n Can n ula Use limbus in eyes to remain phakic and 3 mm for aphakic eyes
or 3.5 mm with 25-gauge systems. This incision is plugged
The infusion cannula should be placed as soon as the sclera
with the endoilluminator held backhand as the MVR blade is
is exposed. The infusion should remain until just prior to
removed to maintain intraocular pressure (IOP) rather than
conjunctival closure. Any infusion system should be visual-
the unnecessary step of placing a scleral plug.
ized before use. This first-in, last-out approach is essential to
The third scleral incision is primarily for the vitreous
prevent hypotony leading to intraocular or suprachoroidal
cutter, fragmenter, scissors, forceps, endophotocoagulator,
choroidal hemorrhage as well as miosis.
etc. It is placed along an imaginary line extending from the
lowest part of the bridge of the lateral aspect of the supraor-
bital rim through the center of the pupil, the usual distance
INSTRUMENT SCLEROTOMIES from the limbus. Vitreoretinal instruments bump the brow
or nose and have difficulty reaching the anterior chamber if
After the infusion cannula is inspected, the infusion is entered superiorly or nasally. Using the active instruments in
turned on with the infusion pressure at 45 mm Hg in adults the hand corresponding to the eye being operated on (right
or 35 mm Hg in children or patients with extremely low per- eye—right hand; left eye—left hand) and having the instru-
fusion (blood) pressure. Use of the Alcon vented gas forced ments enter the eye at a point corresponding to the lowest
infusion (VGFI) system is better than placing the bottle a point of the nose and temporal orbital rim allow better access
certain distance above the eye because it gives a direct digital to all areas in the globe. Many surgeons limit their capabili-
readout of the infusion pressure. Starting the infusion before ties by always using the active instruments in their dominant
the second and third incisions are made inflates the uvea hand. There are certain maneuvers that involve exchanging
against the sclera, permitting the other sclerotomies to be the active instrument to the opposite hand for better access
made through the choroid, and nonpigmented ciliary epi- to the pathology. This is frequently the case with endophoto-
thelium because they are pushed against the sclera like an coagulation to peripheral retinal breaks, scissors delamina-
inner tube in a tire. This approach also prevents the miosis tion, submacular surgery, and drainage of subretinal fluid
and bleeding that result from hypotony. through peripheral breaks.
The second scleral incision, usually for the endoillumi- In all cases, the placement of sclerotomies should avoid
nator, is made superonasally. It is best made near along an previous incisions, areas of known fibrovascular proliferation
imaginary line extending from the lowest part of the bridge on the pars plana, and dense vascularity. Most attempts to use
of the nose through the center of the pupil, 4 mm from the previous sclerotomies result in wound leaks, tearing of the
116 SECTION II ■ Surgical Technology and Techniques

Fig ure 1 0 .5 ■ Running shoelace 8-0 or 9-0 nylon sutures with ends cut flush with knot elim i-
nate blebs and scleritis associated with absorbable sutures and p roduce tight closure.

sclera, and excessive bleeding and may lead to fibrovascular the tensions between loops, and has fewer ends to protrude
proliferation. This is not the case with 25-gauge surgery. through the conjunctiva. The ends should be cut right on
the knot with a sharp blade under operating microscope
visualization by moving the suture toward the blade. The
WOUND CLOSURE AND ends can be buried in the wound to prevent conjunctival
POSTOPERATIVE MEDICATION erosion, but this makes a watertight wound more difficult
to attain.
Wo un d Clo sure
Cryopexy, diathermy, or thermal cautery should not be CONJUNCTIVA AND TENON’S
applied to pars plana incisions. These modalities damage
CAPSULE
the sclera and predispose it to poor healing, poor closure,
inflammation, vascularization, and tissue ingrowth. Any form
The conjunctiva and Tenon’s capsule are closed with inter-
of retinopexy is inappropriate at pars plana sites because the
rupted 6-0 plain gut, 8-0 Biosorb, or equivalent sutures
scleral incisions are anterior to the retina. If there is exces-
trimmed on the knot in a single layer. The Tenon’s capsule
sive bleeding, minimal bipolar diathermy can be applied, but
should not be sutured to the muscle insertions because this
an instrument through the wound or closure by the suture
decreases the lid fissure, limits ocular motility postopera-
will stop most bleeding. Absorbable sutures are not used in
tively, and makes reoperation more difficult. Longer lasting
the sclera by the authors because their inelasticity facilitates
sutures are very irritating to the patient and are unneces-
wound leakage during the operation, postoperatively, and
sary. Conjunctival closure with bipolar diathermy (coapta-
during reoperations. The author (S.C.) has observed many
tion) can cause conjunctival shrinkage and fistulas and is
filtering blebs from the use of absorbable sutures by other
less secure than suturing.
surgeons. Wound leaks may cause hypotony and predispose
to tissue ingrowth. Vicryl scleritis causes considerable post-
operative inflammation for many weeks. The best closure is Reference
achieved with running shoelace 8-0 to 10-0 monofilament 1. O’Malley C, Heintz RM. Vitrectomy via the pars plana, a
nylon sutures (Fig. 10.5). A running or X-type suture can new instrument system. Trans Pac Coast Otoopthalmol Soc
be placed more rapidly than interrupted sutures, equalizes 1972;53:121.
SEC TIO N III
In -Offi ce Pro ce d ure s

117
C H AP TER
11
LASER PHOTOCOAGULATION

PASCAL LASER PROPHYLAXIS FOR RETINAL


BREAKS
The Pascal method of photocoagulation allows the delivery
of a predetermined pattern by scanning the placement of the Though roughly 70% of the population develops a posterior
laser spots and controlling the emission of the laser light to vitreous detachment (2), only about 4% of the population
high degrees of accuracy and precision. This laser system has retinal breaks (3). Of these patients, only about 6 in
(PASCAL; OptiMedica Inc.) provides 532-nm light through 10,000 develop retinal detachments. Patients go on to
a multimode step index optical fiber to an integrated gal- develop retinal detachment after retinopexy in about 2% of
vanometer-based optical scanner housed within a slit lamp cases, with the complication rate of retinopexy remaining
biomicroscope. The exit surface of the fiber is telecentrically exceedingly low and difficult to measure (4). Determin-
imaged through the scanning system onto the retina, provid- ing the need for treatment is multifactorial and complex
ing a variety of spot sizes with nominally top-hat intensity at best.
profiles. At the aerial image plane of the slit lamp micro- The most conservative position is that only symp-
scope, the laser spots measure 60, 100, 200, and 500 µm in tomatic flap tears should be treated (5). However, many
diameter, all at the same numerical aperture. Different core large horseshoe tears, which all surgeons would agree
diameter fibers are used to produce the different spots. Pulse need treatment, are asymptomatic as many surgeons
durations from 10 to −1,000 ms are available. These optical have discovered in examinations carried out before laser-
pulses have 10 µs rise and fall times and a temporal power assisted in situ keratomileusis (LASIK) and even routine
stability of greater than 90%. A touch-screen graphic user examinations (6).
interface is used to control treatment parameters, includ- Clinical characteristics in favor of treatment include
ing the spot size, laser power, pulse duration, and pattern larger breaks, flap tears instead of round holes, breaks outside
geometry. Once the treatment parameters are appropriately lattice, superior location, and evidence of vitreous traction (7).
selected, a foot pedal is used to activate the laser. The Pascal Larger breaks allow greater transhole flow, potentially exceed-
Photocoagulator enables the physician to deliver multiple ing the capacity of the retinal pigment endothelium (RPE)
laser lesions with a single footswitch depression by automat- pump to stabilize a subclinical retinal detachment. Round
ing the emission of laser light with as much as 56 pulses tears are thought to be less likely to have traction, but they are
within half a second. often noted to have traction at the time of vitrectomy. Opercu-
The PASCAL laser utilizes short-duration pulses lated breaks are least likely to have vitreous traction (8). Rolled
(20–30 ms) to limit thermal diffusion (1). Anterior thermal edges are said to be indicative of tangential traction, but the
diffusion can create damage to the nerve fiber layer. Poste- internal limiting membrane (ILM) is elastic, and surgically
rior thermal diffusion heats the choroid, causing pain and resected normal retina immediately rolls inward. Superior
even choroidal edema (effusion). Lateral thermal diffu- breaks are probably slightly more significant because grav-
sion is the most observable and clinically significant issue ity will decrease the likelihood of a stable subclinical retinal
because it results in “RPE creep,” enlargement of lesions detachment.
over time causing loss of central visual function. The PAS- Pigmentation indicates chronicity, not adherence; there-
CAL laser utilizes a three-galvanometer system to rapidly fore, pigmentation is only a relative contraindication (9).
produce a precise pattern with programmable interlesion Other factors favoring treatment include a history of reti-
spacing. Lighter intensity, smaller, more numerous spots nal detachment in the other eye, a family history of retinal
with precise spacing produce the best outcomes and can detachment, and physically active careers and/or sports (10).
be produced in a shorter time because of the scanning Surgeons cannot predict which patients will be struck by an
system. air bag, experience a serious fall, or suffer another type of
118
CHAPTER 11 ■ Laser Photocoag ulation 119

trauma suggesting that trying to guess who needs laser by to the break. If posterior capsular opacification or cortical
evaluating lifestyle is problematic. cataract makes visualization anterior to the break impossible
Socioeconomic situations suggesting that the patient is even with scleral depression, the anterior treatment can be
less likely to return for follow-up should also be taken into continued to the ora at both ends of the break or laser retin-
account. Most surgeons believe that anticipated LASIK, cata- opexy can be combined with cryopexy.
ract removal, or vitrectomy surgery is a reason to be aggres- The presence of subretinal fluid around a tear (subclini-
sive about treating asymptomatic retinal holes and similar cal retinal detachment) requires enclosure of the total area
low-risk breaks. A high percentage of the population will of the subretinal fluid with at least three rows of laser. These
ultimately have cataract surgery, suggesting the need to treat patients often require laser treatment from ora to ora for
lower risk breaks in most patients (11). complete enclosure, and this is best accomplished with laser
indirect ophthalmoscopy with sclera depression.
The PASCAL laser (OptiMedica, Santa Clara, California)
COMPARISON WITH CRYOPEXY produces a precision pattern of shorter duration spots.
The pattern results in greater spacing uniformity. Using 20- to
Unlike laser, cryopexy disperses living RPE cells, possibly 30-ms–duration burns results in significantly less pain from
increasing the risk of proliferative vitreoretinopathy (PVR) thermal diffusion to the choroid while not increasing treat-
and epimacular membranes. The PVR and epimacular mem- ment time. The arc pattern works well for retinal breaks.
branes that are said to be complications of treating retinal The laser indirect ophthalmoscope (LIO) is useful for
breaks could actually be also directly related to the retinal wheelchair patients and patients with spinal deformities,
break causing the retinal glial cells and RPE cells to have such as severe osteoporosis or scoliosis. The LIO is also ideal
loss of contact inhibition (12). Cryopexy produces inflam- for treating through gas bubbles using trial-and-error head/
mation and exudation but no immediate adherence, while bubble positioning to optimize focus and access to the breaks.
laser produces moderate, immediate adherence and no exu- The LIO is ideal for operating room use on children or for
dation. In addition, cryopexy is more painful and produces treatment of the contralateral eye during retinal detachment
conjunctival damage as well (13). surgery often under general anesthesia. LIO treatment is eas-
ily utilized with scleral depression.
Retrobulbar blocks are seldom needed with laser retin-
TECHNIQUE opexy. There is an increased risk of globe penetration in
myopic patients with thin sclera, who, of course, are the very
Spot spacing is a judgment issue in determining the optimal patients with a higher incidence of retinal breaks.
number of rows. Wide spacing (underlap) raises the issue of If a patient has difficulty with pain during laser treat-
subretinal fluid leakage between the spots, while overlapping ment, the authors prefer subconjunctival or peribulbar
results in areas of overtreatment. Most surgeons use approxi- anesthesia with lidocaine rather than retrobulbar block.
mately three rows of confluent spots. Many surgeons do not This avoids the risks of retrobulbar injection and provides
treat round holes within lattice degeneration unless breaks adequate analgesia, although total anesthesia is not often
outside lattice are present that require laser. While the use of accomplished.
discrete circular spots is the standard of care, movement of the Patients can return to full activity 14 days after laser treat-
laser using a painting technique increases uniformity of the ment because tensile strength reaches the maximum level at
thermal effect, although painting potentially produces more this time. Often, patients are counseled unnecessarily to avoid
pain due to heat diffusion. work, housekeeping, and exercise for extended periods.
Selecting optimal power is also a judgment issue. The longevity of the population, high expectations of
Undertreatment may not produce enough pigmentation medical treatment, and the impossibility of trauma predic-
to subsequently validate treatment adequacy, while the tion make treatment of most retinal tears advisable. The
so-called heavy treatment may produce excessive inflamma- advances in surgical equipment and techniques reduce
tion, possibly leading to PVR and epimacular membranes. the likelihood of complications. The short recovery period
It is crucial to completely surround the retinal break. Many makes treatment acceptable to active and working patients.
patients receive insufficient treatment anterior to the break.
Three-mirror contact lenses (150 degrees field of view) are
the contact lenses most widely used to treat retinal breaks, References
but various wide-angle (>130 degrees) lenses can be used for 1. Jain AM, Blumenkranz MS, Paulus Y, et al. Effect of pulse
all but the most peripheral breaks if care is taken to ensure duration on size and character of the lesion in retinal photoco-
treatment anterior to the break. The Eisner (Crystal Lake, agulation. Arch Ophthalmol 2008;126(1):78–85.
Illinois) scleral depressor fits over the 3-mirror lens and 2. Snead MP, Snead DR, Mahmood AD, et al. Vitreous detachment
and the posterior hyaloid membrane: a clinicopathological
works well, although it is somewhat more uncomfortable study. Eye (Lond) 1994;8(Pt 2):204–209.
and generally underutilized. Ensuring maximal dilation of 3. Murakami K, Jalkh AE, Avila MP, et al. Vitreous floaters.
the pupil is important to enable adequate treatment anterior Ophthalmology 1983;90:1271–1276.
120 SECTION III ■ In-Office Procedures

4. Pollak A, Oliver M. Argon laser photocoagulation of 9. Morse PH, Eagle RC Jr. Pigmentation and retinal breaks. Am J
symptomatic flap tears and retinal breaks of fellow eyes. Br J Ophthalmol 1975;79(2):190–193.
Ophthalmol 1981;65:469–472. 10. Davis MD. Natural history of retinal breaks without detach-
5. Kazahaya M. Prophylaxis of retinal detachment. Semin Oph- ment. Arch Ophthalmol 1974;92:183–194.
thalmol 1995;10(1):79–86. 11. Robertson DM, Buettner H. Pigmented preretinal membranes.
6. Wilkinson CP. Evidence-based analysis of prophylactic treat- Am J Ophthalmol 1977;83:824–829.
ment of asymptomatic retinal breaks and lattice degeneration. 12. Glaser BM, Vidaurri-Leal J, Michels RG, et al. Cryotherapy
Ophthalmology 2000;107(1):12–15. during surgery for giant retinal tears and intravitreal disper-
7. Combs JL, Welch RB. Retinal breaks without detachment: nat- sion of viable retinal pigment epithelial cells. Ophthalmology
ural history, management, and long-term follow-up. Trans Am 1993;100(4):466–470.
Ophthalmol Soc 1982;80:64–97. 13. Kramer SG, Benson WE. Prophylactic therapy of retinal breaks.
8. Byer NE. What happens to untreated aymptomatic retinal Surv Ophthalmol 1977;22(1):41–47.
breaks, and are they affected by posterior vitreous detach-
ments? Ophthalmology 1998;105(6):1045–1049.
C H AP TER
12
PNEUMATIC RETINOPEXY
AND OFFICE FLUID-GAS
EXCHANGE

Air was used by Ohm and Rosengren over a The authors’ current approach to pneumatic retinopexy
half century ago for the repair of retinal detach- involves avoiding cryotherapy unless an individual small tear
ments. Dominguez (1), and subsequently Hilton is seen that can be treated with a single cryo spot. Avoidance
and Grizzard (2), reported the use of in-office of cryotherapy decreases the stimulus for PVR. If a larger tear
injection of expanding gas for the repair of primary, rheg- is seen and pneumatic retinopexy is performed, injection of
matogenous retinal detachment. Initially, cryoretinopexy was C3F8 gas without cryotherapy is the authors’ preferred course
applied before gas injection, while others used laser retin- of action. If reattachment is successful, secondary laser retin-
opexy after reattachment, analogous to the postreattachment opexy to the retinal tear can be performed on the attached
retinopexy concept developed by the principal author for retina. If the gas injection is unsuccessful at retinal reattach-
vitreoretinal surgery (3–9). ment, the authors proceed to final surgical repair with vitrec-
Retinal reattachment surgery outcome data are very dif- tomy, and the added insult of cryo is avoided.
ficult to interpret because of the large number of variables, vast
array of surgical options, and combinations of techniques, as
well as the relatively low number of cases per surgeon. Some SURGICAL SEQUENCE
surgeons state that pneumatic retinopexy causes proliferative
vitreoretinopathy (PVR), and in spite of this, they inject gas after The authors use topical viscous lidocaine anesthesia applied
scleral buckling. While it is clear that pneumatic retinopexy first as a drop and then with a sterile applicator at the intended
should not be used for cases with PVR or vitreous traction, pars plana injection site. Retrobulbar or peribulbar anesthesia is
it is not known with statistical accuracy what the indications used in most cases, followed by cryopexy to the retinal breaks.
should be. It is clear that single, superior retinal breaks are the The patient should be prepped with Betadine 5% after
ideal cases for pneumatic retinopexy, but these cases can also anesthesia is achieved. A sterile speculum is required to pre-
be repaired with very high success rates by scleral buckling or vent the needle from contacting the nonsterile lid margins
vitrectomy, gas, and laser. Pneumatic retinopexy costs less than and lashes. The surgeon should use sterile gloves, sterile
scleral buckling or vitrectomy-based repair. Some advocate technique, and a filter when drawing up the gas.
the use of 360-degree laser retinopexy in these cases, which The authors routinely use C3F8 gas rather than SF6
raises the issue of potential stimulation of PVR (10). The prin- because it expands three to four times (its original size)
cipal author has utilized pneumatic retinopexy in a wide vari- compared to two times for SF6 (11–18). Greater expansion
ety of cases since 1988 but uses the technique less frequently means that less gas can be injected and therefore less para-
in recent years because of failures due to progressive contrac- centesis will be required.
tion of the vitreous, new retinal breaks, unpredictability, and Injection of gas is best performed with the patient lying
PVR. Patients with medical problems combined with simple, on his or her side (Fig. 12.1), not supine, seated, leaning over
superior retinal detachments are the best candidates for pneu- the examining table, or at the slit lamp. Multiple bubbles (fish
matic retinopexy. The advent of transconjunctival, sutureless, eggs) are completely preventable if the injection is performed
25-gauge vitrectomy allows many of the advantages of pneu- at the highest point of the eye, with the needle advanced just
matic retinopexy to be retained but in the context of the better past the pars plana epithelium. By using this method, all
outcomes afforded by vitrectomy, gas, and laser. gas is injected into the original bubble, preventing multiple
121
122 SECTION III ■ In-Office Procedures

opinion that higher volumes give higher success rates but


demand paracentesis. The authors use paracentesis in all
cases. Immediately after the injection, the reclining patient
chair is tilted up to the seated position at the slit lamp with
the speculum remaining in place. Sitting the patient up has
the added advantage of rolling the bubble away from the
injection site, eliminating the need for an applicator stick to
prevent leakage. A 30-gauge or 27-gauge needle is advanced
parallel and adjacent to the limbus through the inferior clear
cornea into the anterior chamber. Oblique entry ensures a
self-sealing wound. Needle placement over the iris instead
of the pupil reduces the risk of lens damage. The patient
usually reports no light perception immediately after the
injection, but light perception returns as soon as the para-
centesis reduces the intraocular pressure.
Fig ure 1 2 .1 ■ Gas injection is perform ed at the highest point
If the breaks could not be treated before gas injection
of the eye with the patient lying on his or her side. because of a bullous detachment, retinopexy can be per-
formed 1 or 2 days after the gas injection when the subretinal
bubbles (Fig. 12.2). A 32-gauge needle should be used to fluid has pumped out. The laser indirect ophthalmoscope
reduce leakage of gas after the injection. The smaller gauge of is preferred to the 3-mirror contact for postreattachment
the needle favors a slower rate of injection due to increased retinopexy (Fig. 12.3). If there are concerns about the abil-
resistance. Slower gas injection creates less fish egg bubbles. ity to visualize the breaks after reattachment, preattachment
A tuberculin or 3-mL syringe is used for the gas injection. retinopexy with transscleral cryopexy can be utilized.
Repetitive in and out movement of the needle into the eye
is avoided if the patient is on his or her side, decreasing the
risk of endophthalmitis. By placing the needle only 3 mm IN-OFFICE FLUID-GAS EXCHANGE
into the eye, inadvertent contact with the lens and retina is
reduced. It is difficult for a patient on his or her side to raise Postvitrectomy retinal detachment without traction or PVR
the head and bring the needle into contact with the retina. or hemorrhage patients can be managed by in-office fluid-gas
Approximately 0.3 to 0.6 mL of gas is injected, although the exchange using isoexpansive concentrations of C3F8 (16%)
most appropriate volume is controversial. It is the authors’ or SF6 (25%) (Fig. 12.4). The injection needle is positioned

Fig u re 1 2 . 2 ■ In sertin g t h e
30-g auge needle just b arely into
the eye ensures having a sing le
bubble.
CHAPTER 12 ■ Pneum atic Retinopexy and Office Fluid -Gas Exchange 123

Fig u re 1 2 .3 ■ Retin o p exy is


perform ed with transconjunctival
cryop exy b efore gas injection or
using the laser in direct op hthal-
m oscope as shown.

3 to 4 mm posterior to the limbus at the highest part of the


eye as in pneumatic retinopexy. The needle is advanced only
3 mm into the eye to avoid multiple bubbles. This 32-gauge
needle is connected via a short piece of tubing to a 30- to
60-mL air- or gas-filled syringe operated by the assistant.
A second 25- to 30-gauge needle, depending on the viscosity
of the intraocular fluid, is positioned 4 mm posterior to the
limbus, at the lowest portion of the eye. This needle is con-
nected to a tuberculin syringe, with the plunger removed,
to act as a handle. The open end of this needle is positioned
over a waste can to collect the fluid. The exchange is contin-
ued until gas appears in the egress needle. The egress needle
is withdrawn and the intraocular pressure adjusted using the
gas syringe and tactile assessment of the IOP with a sterile
applicator stick.
Phakic patients postvitrectomy can develop posterior
subcapsular cataracts after fluid-gas exchange if strict face-
down positioning is not followed. This can make second-
ary laser retinopexy very difficult. Cryotherapy to the retinal
tear prior to the fluid-gas exchange should be considered in
phakic postvitrectomy patients.

References
1. Dominguez A. Cirugia precoz y ambulatoria del desprendimento
de retina. Arch Soc Esp Oftamol 1985;48:47–54.
2. Hilton GF, Grizzard WS. Pneumatic retinopexy: a two step out-
Fig u re 1 2 .4 ■ In -office two -n eed le (30-g aug e in jectio n , patient operation without conjuctival incision. Ophthalmology
25-g aug e eg ress) fluid -g as exchang e is p erform ed in p atients 1986;93:626.
wh o d evelop retinal d etachm en t after vitrectom y. Th is tech - 3. McDonald HR, Abrams GW, Irvine AR, et al. Management of
niq ue ensures a com plete fill with an isoexp ansive concentration subretinal gas following attempted pneumatic retinal reattach-
of long-acting gas. ment. Ophthalmology 1987;94:319 –326.
124 SECTION III ■ In-Office Procedures

4. Hilton GF, Kelly NE, Salzano TC, et al. Pneumatic retinopexy. 12. Constable IJ, Swann DA. Vitreous substitution with gases. Arch
A Collaborative report of the first 100 cases. Ophthalmology Ophthalmol 1975;93:416.
1987;94:307–314. 13. DeJuan E Jr, McCuen B, Tiedeman J. Intraocular tamponade
5. Roy FH. Master techniques in ophthalmic surgery. Williams and and surface tension. Surv Ophthalmol 1985;30:47.
Wilkins, 1995: 1118–1119. 14. Stinson TW III, Donion JV Jr. Interaction of intraocular air and
6. Wilkinson CP, Rice TA. Michels retinal detachment. Mosby, SF6 with nitrous oxide: a computer simulation. Anesthesiology
1997:596–612. 1982;56:385.
7. Brinton DA, Hilton GF. Pneumatic retinopexy. Ophthalmol Clin 15. Lincoff H, Coleman J, Kreissig I, et al. The perfluorocarbon
North Am 1994;7:1. gases in the treatment of retinal detachment. Ophthalmology
8. Vygantas CM, Peyman GA, Daily MJ, Ericson ES. Octafluo- 1983;90:546.
rocyclobutane and other gases for vitreous replacement. Arch 16. Fineberg E, Machemer R, Sullivan P. SF6 for retinal detach-
Ophthalmol 1973;90:235. ment surgery. A preliminary report. Mod Probl Ophthalmol
9. Norton EWD. Intraocular gas in the management of selected retinal 1974;12:173.
detachments. Trans Acad Ophthalmol Otolaryngol 1973;77:OP-85. 17. Lincoff HA, Maisel JM, Licoff A. Intravitreal disappear-
10. Hilton GF, Tornambe PE. Pneumatic retinopexy: an analysis ance rates of four perfluorocarbon gases. Arch Ophthalmol
of intraoperative and postoperative complications. Retina 1984;102:928.
1991;11:285—294. 18. Lowe MA, McDonald HR, Campo RV, et al. Pneumatic retin-
11. Lincoff H, Mardirossian J, Lincoff A, et al. Intravitreal longevity of opexy. Surgical results. Arch Ophthalmol 1988;106:1672.
three perfluorocarbon gases. Arch Ophthalmol 1980;98:1610.
SEC TIO N IV
Dise ase -Sp e cifi c Man ag e m e n t

125
C H AP TER
13
PROPHYLAXIS OF RETINAL BREAKS
AND SCLERAL BUCKLING

spaced-out lesions. Many authors recommend treatment


PREVENTION OF RETINAL
only if the retinal breaks are symptomatic (1–10). Relying
DETACHMENT on the patient can be effective in a population of highly edu-
cated people but is less reliable in patients having less edu-
It is probable that the per capita incidence of retinal detach-
cation and more socioeconomic problems. Large breaks are
ment has decreased over the past three decades. The replace-
typically more significant than small breaks. Retinal detach-
ment of intracapsular cataract surgery, first by extracapsular
ment in the fellow eye or another location in the same eye
surgery and then by phacoemulsification with endocapsular
or a family history of detachment is a relative indication for
intraocular lenses, has certainly contributed to the decline.
treatment. Patients for whom cataract surgery is planned,
The widespread use of indirect ophthalmoscopy and retin-
athletes, or certain careers with risk of high G-forces or non-
opexy for prophylaxis of retinal breaks has probably had a
availability of care may indicate the need for prophylactic
major impact as well. It is also likely that the increased use
treatment as well. Retinal breaks outside lattice are more sig-
of protective eyewear has made a contribution.
nificant than breaks inside lattice. Superior breaks are prob-
ably more significant than inferior breaks. Pigment around
IS EVIDENCE-BASED TREATMENT OF breaks does not indicate adherence to the retinal pigment
RETINAL DETACHMENT POSSIBLE? endothelium but does indicate chronicity.
Laser can be used to “wall-off,” better termed “laser
The increasing number of retinal specialists per capita coupled delimiting,” a retinal detachment which is small enough that a
with fewer detachments per patient-year, variable pathology, triple row of laser will not significantly impair the visual field.
and significantly more treatment options has virtually guaran- On occasion, laser-delimited detachments will spontaneously
teed that the treatment of retinal detachment will never move reattach. Laser confinement of relatively small retinal detach-
to an evidence-based paradigm because of the statistical com- ments has remarkably good long-term results and is almost
plexity. Therapeutic options include scleral buckling, vitrec- certainly underutilized because of habit and economics.
tomy, and pneumatic retinopexy. Gas choices include air, C3F8,
and SF6. Buckle options include sponges versus “hard” silicone, SCLERAL BUCKLING
drainage versus nondrainage, encircling versus segmental,
radial versus circumferential, etc. Retinopexy choices include Case Se le ct io n
cryotherapy versus laser (transscleral or laser indirect ophthal-
moscope) versus diathermy. Many of these therapies are used Given the success of modern microincisional vitrectomy for
in combination, making the analysis even more complex. repair of simple and complex retinal detachments, the ques-
tion of when to recommend either procedure is still heavily
debated (11). Even in this era of evidence-based medicine, it
PROPHYLACTIC RETINOPEXY is exceedingly difficult to create a clinical trial that accounts
for the myriad of variables present in surgical techniques
Laser is preferred to cryotherapy for prophylactic retinopexy by individual surgeons. The authors do not perform sclera
because there is less pain and potentially less proliferative buckling in combination with vitrectomy, since there is no
vitreoretinopathy (PVR). Low to moderate intensity con- additive benefit in their view of combining both procedures,
fluent lesions with fewer rows are preferred by the authors although the risks and complications of both techniques are
over the very common method of using many rows of heavy, nonoverlapping and therefore additive.
126
CHAPTER 13 ■ Prophylaxis of Retinal Breaks and Scleral Buckling 127

The authors currently only recommend sclera buckling Bucklin g wit h Vit re o us Surg e ry
in young phakic patients with simple retinal detachments,
Scleral buckling is used with many variations for retinal
anterior tears, and no PVR. The presence of PVR requires
reattachment (13,14). A rapid, simplified form of scleral
vitrectomy and obviates sclera buckling.
buckling is preferred by the authors for all scleral buckling
(Fig. 13.2). Scleral buckling is significantly overutilized in
Sp o n g e s Ve rsus Hard Silico n e conjunction with vitreous surgery. Scleral buckling causes
The smooth surface and relative incompressibility of “hard” a very significant incidence of strabismus, ptosis, pain, con-
silicone make it superior to sponges for scleral buckling in junctival damage, and refractive error. Scleral buckling is not
the author’s opinion. Higher extrusion and infection rates indicated for giant break surgery, PVR, or routine rhegmatog-
make sponges less desirable (12). Sponges create a higher enous retinal detachments. The elimination of vitreous trac-
buckle immediately under the sutures and a lower buckle tion coupled with complete intraoperative reattachment by
between the sutures. This potentially creates a higher inci- vitrectomy has eliminated, in the authors’ opinion, the need
dence of radial folds. The area of lesser buckling effect for combined buckle-vitrectomy.
between sutures corresponds with regions of bulging under
the conjunctiva, which creates a dellen-like effect leading to En circlin g Ban d s
exposure of the buckle (Fig. 13.1).
Prophylactic scleral buckling with an encircling band can
then be thought of as making a new ora serrata to treat
Im p lan t s Ve rsus Exp lan t s
peripheral vitreoretinal traction preceding retinal detach-
While scleral dissection is an acceptable method of scleral ment. Encircling bands for prophylactic buckling were used
buckling, it is rarely performed currently because of its frequently in the early days of vitrectomy (15). This approach
inflexible, time-consuming nature. The original justifica- is not used today because of better cutters, techniques, flu-
tion for scleral dissection was the ability to avoid scleral idics, and dissection methods. Local anesthesia, outpatient
damage from full-thickness diathermy. Although burying surgery, cost containment as well as avoidable complications
the buckle under a flp reduces extrusion, it increases intru- such as pain, strabismus, refractive error, conjunctival dam-
sion, operating time, inflexibility, and the risk of scleral age, and ptosis all contribute to the virtual elimination of
perforation. prophylactic buckling.

Fig ure 1 3 .1 ■ Silicone sponges bulg e between m attress sutures at the end of buckle, which
causes conjunctival erosion (dellen form ation), which lead s to exposure of buckle and extrusion.
In addition, b uckle is flexible, which results in a lower buckle between m attress sutures, leading
to radial folds and unsupported retinal breaks.
128 SECTION IV ■ Disease-Sp ecific Managem ent

Fig u re 1 3 . 2 ■ A sim p lified


approach saves time, reduces com-
plexity, and reduces complications.

Circum fe re n t ial Exp lan t s The explant is trimmed from a larger piece of silicone
for a custom fit if a standard width implant will not work.
Because of their narrow configuration, bands alone are usu-
In every case, the explant width is made so that the outer
ally not utilized to treat specific retinal breaks. If a circum-
surface of the explant conforms to the contour of the globe
ferential explant can cover the posterior extent of a retinal
after tying up the sutures. Off-shelf explant with sutures
break, it is utilized in preference to a radial explant. Circum-
placed 1 to 3 mm wider than the explant will not provide
ferential explants require less exacting localization, do not
the correct effect. If a large ratio of implant width to suture
distort the macula, and cover a broader extent of vitreous
spread is utilized, the explant will be only partially imbri-
base pathology. Posterior breaks are managed with vitrec-
cated into the sclera. This external protuberance wears
tomy techniques. The principal author has not used radial
a hole in the conjunctiva by creating a dellen-like effect,
buckles or sponges for over 25 years in the buckling alone
is similar to that observed with sponges, and provides a
or vitrectomy setting.
less substantial buckling effect. A slight chamfering of the
Monofilament (5-0) nylon sutures are utilized with a
cut edges of the silicone seems to provide a lesser chance
single circumferential posterior scleral bite. In contrast to
of scleral or conjunctival erosion. Two or three mattress
radial suture bites, the circumferential bite can be quite long
sutures per quadrant provide the most consistent buck-
without reducing the posterior extent of the buckle. The sin-
ling effect and reduce the chance of extrusion or sutures
gle circumferential posterior scleral bite reduces by one half
pulling through the sclera. Explants without grooves are
the chances of perforating the retina, as compared to paired
preferred because they do not fold along the groove and
bites. This posterior bite is always placed 3 mm and prefer-
bands are almost never combined with buckles using the
ably 5 mm posterior to the most posterior aspect of the most
authors’ technique.
posterior break. A too-anterior positioning of the buckle
The circumferential buckle method described above
causes many reoperations after scleral buckling procedures.
is used by the authors for all scleral buckling. All detached
All anterior scleral bites are placed circumferentially in
quadrants are buckled, direct needle drainage of SRF is used
the scleral condensation, conforming to the rectus muscle
exclusively, and bands, sponges, and radial buckles are never
insertions. This provides an area of thicker sclera for greater
used. An attempt is made to avoid the superior rectus as
permanence. This muscle ring also conforms to the ora ser-
mentioned earlier and only muscles absolutely necessary to
rata; therefore, a scleral suture bite placed here in a circum-
exposure are engaged with traction sutures. Minimal retin-
ferential orientation cannot perforate the retina (Fig. 13.3).
opexy is used. Transscleral diode laser retinopexy is alter-
Extending all circumferential buckles to the ora serrata pre-
native to cryopexy; however, it cannot be used in highly
vents the anterior leakage of subretinal fluid (SRF) associated
pigmented patients and it is more difficult to judge treat-
with narrow bands or buckles placed more posteriorly.
ment intensity.
CHAPTER 13 ■ Prophylaxis of Retinal Breaks and Scleral Buckling 129

Fig ure 1 3 .3 ■ Anterior b ites of m attress suture should b e m ad e in thicker sclera associated
with the rectus m uscle insertions. The thicker sclera red uces p ostop erative p ull-through, and the
location corresponds to the ora so that p erforation will not dam age the retina. Placing all sutures
this anterior prevents SRF from leaking anteriorly.

Fig ure 1 3 .4 ■ Scleral buckling with encircling tire is used for PVR. A band is not necessary, and
the ends of the buckle are joined by a butt joint. All im brication is accom plished by using two to
three m attress sutures per quadrant.
130 SECTION IV ■ Disease-Sp ecific Managem ent

Scle ral Bucklin g wit h a Bro ad References


En circlin g Buckle 1. Foos RY. Posterior vitreous detachment. Trans Am Acad
The authors no longer utilize scleral buckling for PVR. For Ophthalmol Otolaryngol 1972;76:480.
2. Linder B. Acute posterior vitreous detachment and its retinal
those surgeons who believe in using a buckle, they recom- complications. Acta Ophthalmol (supp)(KBH) 1966;87:1.
mend a 360-degree moderate height, broad buckle with the 3. Foos RY. Anatomic and pathologic aspects of the vitreous body.
suturing techniques described for circumferential buckles. Trans Am Acad Ophthalmol Otolaryngol 1973;77:171.
Encircling sponges can result in anterior or posterior leak- 4. Foos RY. Tears of the peripheral retina: pathogenesis, incidence,
age. As PVR is a diffuse process, no attempt is made to cus- and classification in autopsy eyes. Mod Probl Ophthalmol
1975;15:68–81.
tomize the buckle contour; instead, a uniformly high, broad 5. Byer NE. Clinical study of retinal breaks. Trans Am Acad
encircling buckle is utilized (Fig. 13.4). Ophthalmol Otolaryngol 1967;71:461–473.
The posterior circumferential bites are made as posteri- 6. Byer NE. The natural history of asymptomatic retinal breaks.
orly as possible without compression of vortex veins, and the Ophthalmology 1982;89:1033–1039.
circumferential anterior bite is placed in the scleral conden- 7. Byer NE. Long term natural history of lattice degeneration of
the retina. Ophthalmology 1989;96:1369–1401.
sation at the muscle ring. The usual suture spread is 10 to 12 8. Byer NE. Cystic retinal tufts and their relationship to retinal
mm, with the usual tire being 6 to 9 mm in width. Imbrica- detachment. Arch Ophthalmol 1981;99:1788–1790.
tion will then cause the buckle to be flush with the surface 9. Byer NE. Natural history of posterior vitreous detachment with
of the globe. The ends are joined with two interrupted 5-0 early management as the premier line of defense against retinal
nylon sutures, with long bites through the implant mate- detachment. Ophthalmology 1994;101:1503–1513.
10. McHugh DA, Schwartz S, Dowler JG, et al. Diode laser contact
rial, and the knot is then buried. No band, encircling suture, transscleral retinal photocoagulation: a clinical study. Br J
or overlapping is utilized so as to provide a consistently Ophthalmology 1995;79:1083–1087.
smooth internal buckle contour. The direct needle drainage 11. Schwartz SG, Flynn HW. Primary retinal detachment: scleral
technique is used for the intraoperative elimination of SRF buckle or pars plana vitrectomy? Curr Opin Ophthalmol 2006
before the encircling tire is anchored in place. Jun;17(3):245–250.
12. Russo CE, Ruiz RS. Silicone sponge rejection; early and late
complications in retinal detachment surgery. Arch Ophthalmol
Scle ral De fe ct s 1971;85:647.
13. Hahn YS, Lincoff A, Lincoff H, Kreissig I. Infection
Thin sclera can be covered in most cases with a circumferen- after sponge implantation for scleral buckling. Am J
tial scleral buckling or encircling tire previously described. Ophthalmol1979;87:180.
14. Devenyi RG, de Carvalho Nakamura H. Combined scleral
Preserved sclera and fascia lata add a degree of complexity, buckle and pare plana vitrectomy as a primary procedure for
infection risk, and risk of dehiscence and are rarely utilized. pseudophakic retinal detachments. Ophthalmic Surg Lasers
Full-thickness defects, unless extremely large, can usually be 1999;30(8):615–618.
repaired by suturing good tissue edges together rather than by 15. Desai UR, Strassman IB. Combined pars plana vitrectomy
oversewing. This approach creates a scleral shortening effect and scleral buckling for pseudophakic and aphakic retinal
detachments in which a break is not seen preoperatively.
that may be beneficial with PVR or wound-related cellular Ophthalmic Surg Lasers 1997;28(9):718–722.
proliferation. A small leaking area can be handled with vari-
ous tissue adhesives. Sponges appear to be more erosive to
the sclera than hard silicone and are therefore not utilized.
C H AP TER
14
VITRECTOMY FOR RETINAL
DETACHMENT

visualization is essential for peripheral vitreous removal and


INDICATIONS
endolaser and frequently obviates the need for sclera depres-
sion vitrectomy. Contact-type wide-angle visualization (Volk or
The use of vitrectomy for retinal detachment repair is
AVI) provides a 10-degree wider field of view than noncontact
increasing (1) because of a number of factors: more surgeons
systems and eliminates all corneal asphericity due to cataract
are trained in vitrectomy, better cutters and fluidics, wide-
surgery, Limbal Relaxing Incision (LRI), Radial Keratotomy
angle visualization, better methods for drainage of subretinal
(RK), laser-assisted in situ keratomileusis, Penetrating Kerato-
fluid (SRF) and retinopexy are available, and fellows are less
plasty (PK), trauma, keratoconus, pterygium, and pterygium
likely to be taught scleral buckling. The decision between
removal. In addition, contact-based wide-angle visualization
vitrectomy, scleral buckling, and pneumatic retinopexy is a
markedly reduces the need for ocular rotation compared to
very complex issue with few, if any, scientific answers. In gen-
noncontact visualization (BIOM, EIBOS), thereby reducing
eral, vitrectomy, gas, and laser should be used when vitreous
tool flex with 25- and 27-gauge pars plana vitrectomy.
opacities or tractions make it unlikely that scleral buckling
All traction should be removed from each retinal break.
alone will succeed. Relative advantages of scleral buckling
It is crucial to remove traction on the anterior margin as
include less cataract progression and 80% to 90% success
well as from the more apparent flap. Amputation of the flap
rates in selected detachments. Advantages of vitrectomy
ensures removal of this component of the traction.
include no refractive change; no damage to the extraocular
If the breaks or tears can be easily seen after fluid-air
muscles or levator, therefore no strabismus or ptosis; and
exchange, they can be used for internal drainage of SRF.
much less postoperative pain and conjunctival damage.
If access to the breaks is difficult, a posterior drainage reti-
Many surgeons, including the authors, use vitrectomy, gas,
notomy or perfluorocarbon liquids are required. Drainage
and laser in most pseudophakic detachments (2). Many sur-
retinotomy can be initiated by using a disposable bipolar
geons are inclined to use vitrectomy, gas, and laser for supe-
endoilluminator or 25-gauge bipolar diathermy to coagu-
rior detachments and buckling for inferior detachments.
late, weaken, and mark the site (Fig. 14.1). Outside the
Evidence of traction on breaks, especially large horseshoe
arcades, at the most posterior extent of the SRF is often a
tears, suggests the need for vitrectomy as does the pres-
good location; nasal is better than temporal and superior is
ence of avulsed vessels. Vitreous blood or significant opaci-
better than inferior. Single-cut mode with the vitreous cutter
ties require vitrectomy to visualize the retinal breaks. The
will produce an ideal small, round retinotomy (Fig. 14.2).
authors rarely use scleral buckling and never combine vit-
The soft-tip cannula is preferred for internal drainage of SRF
rectomy with scleral buckling. Their current indications for
(Fig. 14.3). Internal drainage of SRF should precede fluid-air
sclera buckling are young phakic patients with clear lenses,
exchange, especially with the use of preexisting peripheral
retinal detachments with anterior tears, and the absence of
retinal breaks for drainage; this sequence prevents posterior
proliferative vitreoretinopathy.
shift of the SRF. If intraocular lens (IOL) fogging occurs dur-
ing fluid-air exchange, the air should be removed and per-
fluorooctane (PFO) used to displace the SRF anteriorly out
SURGICAL SEQUENCE through the retinal breaks (Fig. 14.4). Endolaser retinopexy is
then performed under PFO followed by PFO-gas exchange or
It is essential to remove as much peripheral vitreous as possi- medium-term PFO surface tension management. Fogging can
ble without damaging the lens or creating new retinal breaks. be anticipated if a YAG capsulotomy has been performed and
Scleral depression is very useful for this purpose. Wide-angle the anterior vitreous cortex has been disrupted or removed.
131
132 SECTION IV ■ Disease-Sp ecific Managem ent

Fig ure 1 4 .1 ■ Bip olar d iatherm y is used to m ark the site p rior to m aking d rainag e retino-
tom y to facilitate finding the site after retina reattached by internal d rainage of SRF and fluid-air
exchange.

Fig ure 1 4 .2 ■ Sing le-cut m od e on a 20-g aug e or 25-g aug e cutter results in a sm all, round
drainage retinotom y.
CHAPTER 14 ■ Vitrectom y for Retinal Detachm ent 133

Fig ure 1 4 .3 ■ The soft-tip extrusion cannula can be used for drainage of SRF.

Fig ure 1 4 .4 ■ Liquid perfluorocarbon such as Alcon PFO displaces SRF anteriorly out through
retinal breaks into the anterior vitreous cavity. PFO should be used if fluid-air exchange results in
IOL fogg ing .
134 SECTION IV ■ Disease-Sp ecific Managem ent

Fig ure 1 4 .5 ■ Redrainage facili-


tates com plete rem oval of SRF.

Complete removal of SRF can be accomplished by around the retinal tear or retinotomy can then be performed
sequential drainage. After the initial drainage of SRF, there safely with sharp demarcation of laser uptake (Fig. 14.6).
may be a thin layer of diffuse SRF that may be imperceptible. The authors use 25-gauge, sutureless, transconjunctival
This SRF often recollects underneath the drainage retinotomy technique for all vitrectomy for retinal detachment. Advan-
after approximately 30 seconds. At this time, redrainage can tages of 25-gauge technique over 20-gauge technique include
remove most of the remaining SRF (Fig. 14.5). This tech- (a) port-based flow limiting that produces greater fluidic
nique is best repeated until no SRF recollects. Laser treatment stability and therefore less retinal motion, (b) less pain, (c) less

Fig ure 1 4 .6 ■ Complete removal


of SRF im p roves the up take of
endolaser retinopexy around drain-
age retinotom y.
CHAPTER 14 ■ Vitrectom y for Retinal Detachm ent 135

inflammation, and (d) less conjunctival damage (important for management. Removal of the hyaluronan gel dramatically
patients with glaucoma filtering procedures and surface disor- reduces the viscosity as aqueous humor fills the former vit-
ders). A 25-gauge, soft-tip cannula is used for simultaneous reous cavity. This reduces the pressure gradient the retinal
drainage of SRF and fluid-air exchange unless PFO is used. pigment epithelium pump can achieve across the retina. Sub-
The 25-gauge, soft-tip cannula is excellent for air-gas exchange clinical retinal detachments rarely remain stable after vitrec-
or PFO-gas exchange after endolaser to all retinal breaks. tomy; even very small holes usually result in a total retinal
Although many surgeons perform 360-degree laser in vit- detachment in hours to days. Laser and cryo retinopexy sites
rectomy for retinal detachment and even macular hole cases, reach maximum tensile strength at 10 to 14 days; SF6 bubbles
it is not known whether this reduces postoperative retinal often last 7 to 14 days, which means a full fill or isoexpansive
detachment. Some surgeons use laser indirect ophthalmos- SF6 is required to avoid the use of the longer duration C3F8.
copy (LIO) instead of endolaser. The LIO approach could the-
oretically increase nuclear sclerosis as well as iris and corneal
damage from laser energy absorption. It is possible that light MEDIUM-TERM PERFLUORON
scatter from the cornea and lens could cause macular damage
The authors have been using medium-term PFO for infe-
as well. The only advantage to the LIO approach is less chance
rior retinal detachments and giant breaks since 2002 in a
of bumping the lens during peripheral endolaser treatment.
prospective, consecutive series. The advantages are as fol-
Lens bump can be eliminated by scleral depression and flex-
lows: (a) the patients can be seated, reclining, or supine,
ible or articulated laser probes as well as by using the laser
not face down; (b) compared to scleral buckles, there is no
probe in both the left and the right hand for better access.
induced refractive error, no strabismus, no ptosis, no pain,
It has not been shown that scleral buckling is needed
and no inflammation; and (c) the patients can fly on an air-
when using vitrectomy for retinal detachment repair, and the
plane. Disadvantages are as follows: (a) two procedures are
authors believe that it is widely overutilized. The authors
required, the second one to remove the PFO (this is often
do not use scleral buckling in vitrectomy cases in order to
done with silicone oil as well) and (b) residual PFO droplets
reduce induced refractive error, strabismus, ptosis, pain,
in the anterior chamber and former vitreous cavity. Drop-
conjunctival swelling, and conjunctival damage.
lets of PFO adhere to the zonules and pars plana and are
common even after very compulsive surgical removal. Some
SURFACE TENSION MANAGEMENT investigators believe that PFO is toxic, but the authors have
seen no evidence of toxicity and the patients have excellent
All rhegmatogenous retinal detachments and traction visual function. Some patients develop mild inflammatory
retinal detachments with one or more retinal breaks require deposits on the retina and the lens, but these disappear
gas, medium-term PFO, or silicone oil surface tension in a few weeks after PFO removal. Intraocular pressure

Fig ure 1 4 .7 ■ Medium-term PFO


requires com pulsive, 360-degree
rem oval of vitreous traction using
wide-angle visualization.
136 SECTION IV ■ Disease-Sp ecific Managem ent

Fig u re 1 4 .8 ■ PFO in jectio n


o ver t h e o p t ic n erve wit h a
25-gauge dual-bore cannula.

elevation has been seen by the authors in patients who have macular holes, (f) 5,000 cs has less emulsification than 1,000
PFO migration into the anterior chamber. This technique, cs oil, and (g) inferior peripheral iridectomy is needed in eyes
similar to using vitrectomy plus gas and laser, requires com- with in-the-bag posterior chamber lenses and intact capsules.
pulsive, 360-degree removal of vitreous traction with empha- Unlike gas bubbles, silicone oil does not expand and therefore
sis on traction to all retinal breaks (Fig. 14.7). Adequate can be used if the patient must fly. Positioning is required with
peripheral vitrectomy requires wide-angle visualization and breaks below the horizontal meridian if retinopexy has been
may require scleral depression. After vitrectomy, PFO is performed to inferior breaks. The patient can lie on his or her
injected over the optic nerve with a MedOne double-bore side for temporal and nasal retinal breaks. Retinopexy can be
cannula that enables infusion fluid to leak through the exter- omitted for macular holes causing retina detachment; large
nal lumen of the cannula to equilibrate the intraocular pres- breaks and retinectomies likely to cause PVR if treated; and
sure (Fig. 14.8). Confluent laser is applied with the Alcon breaks not well seen during vitrectomy that can be treated
25-gauge articulating laser probe to each retinal break and later when there is no SRF, retinal edema, or inflammation.
suspicious area. If SRF does not completely disappear, a very This technique is referred to as using “rhegmatogenous con-
small drainage retinotomy can be made and SRF aspirated, finement” for “retinopexy avoidance.”
being careful to not remove PFO. If vitreous traction is seen Air-silicone exchange rather than fluid-silicone
after PFO injection, it can be removed “under” PFO by care- exchange is used by the authors. It is done as described in
fully keeping the cutter port outside the bubble of PFO. This Chapters 4 and 5 using 25-gauge technique.
technique is called “interface” vitrectomy by the authors and
is similar to vitrectomy “under” air or “under” silicone oil.
SUMMARY

SILICONE OIL Vitrectomy as first surgery monotherapy for rhegmatog-


enous retinal detachment has significant advantages over
Silicone oil is essential for most cases of PVR and is often indi- scleral buckling (3) with respect to strabismus, refractive
cated for giant retinal breaks and large retinal defects. Silicone effects, ptosis, pain, conjunctival hyperemia, and chemosis.
oil has less intrafacial tension (surface tension) than air or gas Vitrectomy requires advanced tools such as wide-angle
but does not absorb, and therefore it remains in the eye indefi- visualization, PFO, and fast cutting. Vitrectomy approaches
nitely. Many physicians incorrectly believe (a) silicone oil is toxic require strict attention to the peripheral vitreous and espe-
and must be removed in a few months, (b) silicone oil reduces cially traction on flap tears. A 25-gauge vitrectomy offers
vision, (c) glaucoma is common with oil and most glaucoma better fluidic stability than 20- or 23-gauge vitrectomy and
in patients with silicone oil is oil related, (d) positioning is less patient discomfort, less conjunctival damage, less sub-
not necessary with silicone oil, (e) silicone oil is effective for conjunctival hemorrhage, and less chemosis.
CHAPTER 14 ■ Vitrectom y for Retinal Detachm ent 137

References 3. Brazitikos PD, Androudi S, Christen WG, Stangos NT. Primary


pars plana vitrectomy versus scleral buckle surgery for the treat-
1. Kapran Z, Acar N, Altan T, et al. 25-Gauge sutureless vitrectomy ment of pseudophakic retinal detachment: a randomized clinical
with oblique sclerotomies for the management of retinal detach- trial. Retina 2005 Dec;25(8):957–964.
ment in pseudophakic and phakic eyes. Eur J Ophthalmol 2009
Sep-Oct;19(5):853–860.
2. Arya AV, Emerson JW, Engelbert M, et al. Surgical management
of pseudophakic retinal detachments: a meta-analysis. Ophthal-
mology 2006 Oct;113(10):1724–1733.
C H AP TER
15
GIANT BREAKS

Giant retinal breaks are traditionally defined as


SURGICAL SEQUENCE AND
breaks extending for greater than 90 degrees.
Because of the propensity of the retina to fold
TECHNIQUES
over, surgical management of these detachments
Pat ie n t Ed ucat io n
was very difficult until the advent of vitrectomy and intraoc-
ular gas. The next major advance occurred when Chang ini- Patients should be psychologically prepared for 1 to 3 weeks
tiated the application of perfluorocarbon (PFO) liquids to in the face-down position after surgery, unless medium-term
giant break surgery (1). It is now relatively straightforward PFO liquids (2–3 weeks) are used instead of gas. Patients
to achieve surgical success in most cases. The long-term suc- treated with silicone oil must be positioned as well if the
cess rate is determined by the incidence of proliferative vit- break extends below the horizontal meridian.
reoretinopathy (PVR). Although vitreous dynamics, trauma,
and hereditary peripheral retinal changes play a role, the
In cisio n s
exact pathogenesis of giant breaks remains unclear. The high
incidence of associated PVR presumably relates to the expo- The authors use 25-gauge vitrectomy, laser, internal drain-
sure of vast areas of retinal pigment epithelium (RPE) and age of subretinal fluid (SRF), and air-silicone exchange
retinal glial cells to the vitreous matrix. RPE cells migrate (ASX) or air-gas exchange (AGX) in all giant break cases.
along the vitreous and retinal surfaces. The exposed edge of Intraoperative use of perfluoron (PFO) allows adequate
retina presumably enables glial cells to migrate to the retinal repositioning of the folded retina in large retinal tears.
surface as well. Vitreous is virtually never observed on the Medium-term PFO prevents slippage of the giant break but
photoreceptor side of the retina in these cases. Inward fold- requires two procedures and cannot be used for superior
ing of the retina is caused by hypocellular contraction of the giant breaks. The patient must lie on his or her side for
vitreous, the inherent elasticity of the internal limiting mem- nasal or temporal breaks and be seated, standing, or semi-
brane (ILM), and PVR. Although one can occasionally dis- reclining for inferior breaks with the medium-term PFO
tinguish between giant breaks and giant disinsertions, this technique.
differentiation does not seem to play a role in management
strategies or prognosis.
Man ag e m e n t o f t h e Le n s
Phaco-vit and IOL insertion should be used if there is
CASE SELECTION significant cataract preventing sufficient visualization. In
the past, giant retinal tears were considered indications
Quadrantic giant breaks can rarely be managed effectively for pars plana lensectomy, but the authors do not find a
with scleral buckling alone if there is minimal folding of the need for lensectomy at this time with modern vitrectomy
retina. Mild degrees of folding can be managed by adding techniques.
expanding C3F8 gas to the scleral buckling modality. Most
giant breaks with retinal folding are best managed with vit-
Vit re ct o m y
rectomy and PFO liquids. Giant breaks combined with PVR,
vitreous to a wound, dislocated lenses, or vitreous hemor- Vitreous removal should be performed using very low suc-
rhage are absolute indications for vitrectomy, regardless of tion force with proportional suction control and high cut-
the size of the break. ting rates (5,000 cuts per minute) because of marked retinal

138
CHAPTER 15 ■ Giant Breaks 139

Fig ure 1 5 .1 ■ Peripheral vitreous


must be removed for 360 degrees
as close as possible to the retinal
surface. The anterior flap of the
giant break should be rem oved.
A 25-gauge vitrectom y is ideal for
giant breaks because of greater
fluidic stability.

mobility. The vitreous is very rarely attached to the posterior laser retinopexy to both the retina and the RPE. PFO liquids
edge of the giant break. The authors use 25-gauge technique are surface tension management agents similar to silicone
for all giant break cases because the greater fluidic stabil- oil, air, and gas. Silicone and gas float because their density is
ity makes removal of vitreous near the highly mobile retina less than infusion fluid. PFO liquids are denser than infusion
much safer. fluid and retina and migrate to the lowest part of the eye.
The vitreous must be trimmed to the anterior retinal Retina is less dense than PFO liquids and therefore floats
surface and away from the anterior flap to prevent vitreous to its original position, limited only by vitreoretinal traction
entrapment in the SRF drainage cannula and to prevent late and its inherent stiffness. Similarly, SRF is less dense than
vitreous traction from causing redetachment (Fig. 15.1). It is PFO liquids and floats up through the giant break and into
better to remove the detached nonpigmented pars plana epi- the anterior vitreous cavity. The authors use PFO from Alcon
thelium anterior to the break during removal of the periph- for all cases.
eral vitreous. PFO liquid should be injected on the anterior side of the
retina (Fig. 15.2), near the optic nerve with a MedOne dual-
bore 25-gauge viscous fluid injection cannula. Because the
Fluid -Air-Gas Exch an g e
endoilluminator is plugging one sclerotomy and both PFO
Internal drainage of SRF and fluid-air exchange can be per- and balanced salt solution (BSS) are incompressible, BSS Plus
formed without PFO liquids in selected cases with smaller must be allowed to egress through the dual-port cannula to
breaks and little or no folding. Internal drainage of SRF prevent increasing the intraocular pressure. The PFO should
must precede fluid-air exchange and continue during the be injected until the retina is unfolded and all SRF is gone.
exchange. A soft-tip extrusion cannula should be positioned PFO must reach the pars plana when injection is complete and
just anterior to the edge of the giant break. The retina will all SRF is gone (Fig. 15.3). David Wong has taught us that a
be gently pulled into position as the SRF is drained. Rotating full fill with PFO with the infusion fluid turned off prevents
the eye toward the giant break enables drainage of the vitre- posterior slippage of the giant break. Exchange of PFO for
ous fluid simultaneous with the SRF and facilitates complete air to enable AGX or ASX must be performed precisely to
drainage of SRF and better repositioning of the retina. prevent slippage; the soft-tip cannula for PFO removal must
be positioned above the PFO-fluid interface at all times so
that all saline solution (containing SRF) is removed before any
Pe rfl uo ro carb o n Liq uid s
PFO. The cannula must be slowly advanced posteriorly as the
PFO liquids permit removal of all SRF and position the ret- exchange is done (Fig. 15.4). The BSS-PFO interface is easier
ina in a nonfolded state near its original position, permitting to see than with perfluorodecalin.
140 SECTION IV ■ Disease-Sp ecific Managem ent

Fig ure 1 5 .2 ■ Perfluoron (PFO) is slowly injected near the optic nerve on the anterior surface
of the retina, which displaces SRF anteriorly and unfolds the flap.

Fig ure 1 5 .3 ■ PFO injection is continued until the entire vial has been injected and the PFO is
anterior to the giant break.
CHAPTER 15 ■ Giant Breaks 141

Fig u re 1 5 . 4 ■ Th e so ft -t ip
cannula for PFO rem oval m ust be
p osition ed ab ove th e PFO-fluid
interface at all tim es so that all
saline solution (containing SRF) is
rem oved before any PFO.

If PFO liquids are used when giant breaks and PVR is less than the combined stiffness of the retina and PVR
coexist, care must be taken to avoid PFO liquids from membranes (2–4).
entering the subretinal space by removing all epiretinal
membrane by inside-out forceps membrane peeling with
Re t in o p e xy
the Alcon 25-gauge DSP forceps (Fig. 15.5). PFO liquids
will enter the subretinal space when the difference between Confluent laser retinopexy should be used at the posterior
forces due to specific gravity and intrafacial tension effects margin of the retinal break (Fig. 15.6). Multiple spots with

Fig ure 1 5 .5 ■ Ep iretinal m em -


b ranes should b e rem oved with
inside-out forcep s p eeling , using
conform al, d iam on d -coated , or
DSP forcep s.
142 SECTION IV ■ Disease-Sp ecific Managem ent

Fig u re 1 5 .6 ■ Confluent laser end op hotocoag ulation should b e ap p lied in rows on the
posterior edge of the break and extended to the ora at the two ends of the break.

interspersed untreated spaces necessitate a larger treatment area cause posterior slippage. Laser retinopexy must be applied
and create the problem of leakage of SRF between the spots. Care before this slippage occurs in order to position the laser cor-
should be taken to extend the laser (treatment) around the ends rectly. The PFO-air exchange prior to AGX or ASX must be
of the break to the pars plana to avoid SRF leakage. A 532-nm, performed by placing the extrusion cannula right behind the
diode-pumped, frequency up-converted laser is the authors’ lens or IOL and staying just below the air-BSS + SRF inter-
preference (5–7). Treatment intensity is more difficult to judge face as it moves back. In this way, the PFO is removed last
with red or near infrared laser energy. Cryo is thought to cause and posterior slippage minimized. The eye should be rotated
increased PVR (8–13) and retinal slippage compared to laser. toward the break as the last few drops of PFO are removed
Transscleral diathermy can damage the sclera but can be very to ensure removal of all SRF. The patient must be positioned
effective in an air-filled eye, although it is no longer utilized. prone immediately after surgery and continuously in order
Transscleral laser is less predictable, is not feasible in darkly for the retina to move back to a nonredundant position. On
pigmented patients, and, like cryo and diathermy, requires some occasions, permanent folds will occur. Retinal folds do
exposure of the scleral surface (14). Endophotocoagulation is not represent a serious problem unless the macula is involved
the preferred mode for all vitrectomy cases. Laser indirect oph- or very large areas of RPE are exposed. Damage to the retina
thalmoscope may cause damage to the cornea, iris, and lens. and/or RPE occurring in a prolonged attempt to eliminate
folds may unnecessarily stimulate PVR.
Po st o p e rat ive Surface Te n sio n Man ag e m e n t It is tempting to allow the patient to position part-
time or position other than prone if gas or silicone oil is
The options for postoperative surface tension management used or to shorten the prone period. These compromises, in
include medium-term perfluorocarbon liquids, silicone, and the authors’ opinion, reduce success rates with gas and oil.
gas. The advantage of medium-term PFO is that the slippage Medium-term PFO allows all patients to be supine for sleep-
that occurs at the time of PFO-gas or PFO-silicone exchange ing, and they can be semireclining with an inferior break and
is avoided. The minor disadvantage is that a second surgi- on their side for nasal or temporal breaks.
cal procedure is required to remove the PFO. PFO is ideal
for medium-term tamponade but must be used off-label in
the United States. Gas will absorb, eliminating the need for RESULTS
removal. Gas expands with air travel, bubble size decreases
with time, and the view is problematic for the patient and the With the previously described methodology, about 95% of
physician. Silicone is the best option for PVR combined with cases can be successfully reattached in the operating room
giant breaks. The exchange from PFO to gas or silicone can and remain reattached for the first 3 weeks. However, the
CHAPTER 15 ■ Giant Breaks 143

long-term results range from 50% to 90%, depending 4. Verstraeten T, Williams GA, Chang S, et al. Lens-sparing
on PVR, surgical techniques, and case selection (15,16). vitrectomy with perfluorocarbon liquid for the primary treat-
ment of giant retinal tears. Ophthalmology 1995;102:17.
PVR can be managed by reoperation with the approach
5. Charles S. Endophotocoagulation. Retina 1981;1:117.
described earlier. Epimacular membranes (macular pucker) 6. Yoon YH, Marmour MF. Rapid enhancement of retinal adhesion
occur in a significant number of cases and can be managed by laser photocoagulation. Ophthalmology 1988;95:1385.
effectively with substantial visual recovery after vitrectomy 7. Powell JO, Bresnick GH, Yanoff M, et al. Ocular effects of argon
revision and end-opening, inside-out forceps membrane laser radiation. II. Histopathology of chorioretinal lesions.
Am J Ophthalmol 1971;71:1267.
peeling.
8. Campochiaro PA, Kaden IH, Vidaurri-Leal JS, Glaser BM. Cry-
otherapy enhances viable intravitreal dispersion of retinal pig-
ment epithelial cells. Arch Ophthalmol 1984;103:434.
MANAGEMENT OF THE FELLOW EYE 9. Kreissig I, Lincoff H. Mechanism of retinal attachment after
cryosurgery. Trans Ophthalmol Soc UK 1975;95:148.
The high incidence of bilaterality in the nontraumatic, 10. Johnson RN, Irvine AR, Wood IS. Endolaser, cryopexy, and
genetically determined cases of giant breaks is of great retinal reattachment in the air-filled eye: a clinicopathologic
concern (17). The potential for giant break in the other correlation. Arch Ophthalmol 1987;105:231.
11. Jaccoma EH, Conway BP, Campochiaro PA. Cryotherapy
eye raises the question of prophylaxis to the other eye. causes extensive breakdown of the blood retina barrier. A com-
It is known that retinal breaks and even giant breaks fre- parison with argon laser photocoagulation. Arch Ophthalmol
quently occur at the posterior margin of previous retin- 1985;103:1728.
opexy marks. It is well known that hypocellular vitreous 12. Campochiaro PA, Kaden IH, Vidaurri-leal J, Glaser BM. Cryo-
contraction can pull the retina away from a high, broad therapy enhances intravitreal dispersion of viable retinal pig-
ment epithelial cells. Arch Ophthalmol 1985;103:434.
scleral buckle. The authors currently laser retinal breaks 13. Glaser BM, Vidaurri-leal J, Michels RG, Campochiaro PA. Cry-
in the other eye but do not perform 360-degree retinopexy otherapy during surgery for giant retinal tears and intravitreal
or scleral buckling. dispersion of viable pigment epithelial cells. Ophthalmology
1993;100:466.
References 14. Haller JA, Lim J, Goldberg MF. Pilot trial of transscleral diode
laser retinopexy in retinal detachment surgery. Arch Ophthalmol
1. Chang S, Lincoff H, Zimmerman NJ, Fuchs W. Giant retinal 1993;111:952.
tears: surigical techniques and results using perfluorocarbon 15. Freeman HM, Schepens CL, Couvillion GC. Current manage-
liquids. Arch Ophthalmol 1989;107:761. ment of giant retinal breaks 11. Trans Am Acad Ophthalmol
2. Glaser BM, Carter JB, Kupperman BD, Michels RG. Perfluo- Otolaryngol 1970;74(1):59.
octane in the treatment of giant retinal tears with PVR. Oph- 16. Machemer R, Allen AW. Retinal tears 180 degrees and greater.
thalmology 1991;98:1613. Management with vitrectomy and vitreoretinal gas. Arch Oph-
3. Darmakusma IE, Glaser BM, Sjaarda RN, et al. The use of per- thalmol 1976;94(8):1340.
fluoro-octane in the management of giant retinal tears without 17. Freeman HM. Fellow eyes of giant retinal breaks. Mod Probl
PVR. Retina 1994;14:323. Ophthalmol 1979;20:267.
C H AP TER
16
PROLIFERATIVE
VITREORETINOPATHY

Proliferative vitreoretinopathy (PVR) can be the “P” out of PVR. Like a keloid, PVR is excessive healing or
thought of as a reparative or scarring mechanism. scarring. Scarring is biologically identical to healing; scarring
Contact inhibition is lost because of mechanical is basically a functional description. PVR should be thought
disruption of a tissue such as the retinal pigment of as a hypocellular interaction between various cell types
epithelium (RPE), and the response is migration, contraction, and the extracellular matrix (ECM). RPE cells, glial cells, and
and moderate proliferation of cells. A defect in the internal inflammatory cells have been shown to be involved in PVR.
limiting membrane (ILM) can be created by vitreous trac- Better understanding of the disease process leads to better
tion, a retinal break, or membrane dissection. Glial cells then treatment strategies; excessive retinopexy leads to PVR, and
migrate, proliferate to some extent, and contract the retinal repeated surgical interventions with brief intervals between
surface as if to repair the retina-ILM defect. Retinal breaks procedures are common causes of PVR. Lengthy procedures,
“heal” as a result of this mechanism as has been shown in extensive retinopexy, iris trauma (retractors), combined
macular hole surgery and retinotomies for submacular sur- phaco and intraocular lens (IOL) implantation, and exces-
gery. Similarly, if the retina is separated from the RPE, the sive peripheral “membrane peeling” instead of retinectomy
RPE cells migrate to the retinal surface and the posterior are all factors in iatrogenic PVR. Unlike cancer surgery or
surface of the posterior vitreous cortex (PVC). Monocytes draining an abscess, the goal is minimal tissue destruction,
can gain access to the retinal surface from the iris and cili- not removal of all pathologic tissue. The PVR surgeon should
ary body capillaries as a result of ocular inflammation. It is think like a plastic surgeon, not a cancer surgeon.
known that these cells can migrate, proliferate, and contract Tangential shortening of the cells occurs prior to col-
on the retinal surface (1–13). Glial, RPE, and monocytic cells lagen production, which may be thought of as a late stabi-
share in common the migration along an existing substrate lization phase. The generation of basement membrane and
or to a new tissue surface. As cellular migration occurs, intra- collagen can be thought of as a recreation of Bruch’s mem-
cellular contraction of the cells occurs, creating tangential brane or the ILM, as if to duplicate the process that occurs in
traction on the retina. All healing mechanisms studied have embryonic development.
demonstrated a microtubule, smooth muscle-like contrac- PVR can be localized and create isolated star folds, fixed
tion process occurring with many cells acting in concert. The folds, subretinal changes, or epimacular membranes (16– 18).
myofibroblasts have coated pits, which have specific recep- It can be more widespread, which gave rise to the no longer
tor sites for collagen, fibrin, and elastin. These pits contain used term, “massive periretinal proliferation.” If the prolif-
fibronectin, which allows the cells to attach to collagen fibers eration of glial or RPE cells extended onto the contiguous
(14,15). Growth factors (TGF-b, etc.), metalloproteinases, posterior surface of the PVC, the older term “massive vitre-
fibronectin, and receptors for collagen, elastin, and fibrin are ous retraction” would have been used. At this time, there is
components of the migration and contraction phenomenon. no proof of the role of hyalocytes in the pathogenesis.
Proliferation (mitotic activity) is of much lesser importance. Understanding surgical anatomy is a crucial component
This process can be thought of as hypocellular periretinal of surgical intervention. Core vitrectomy is actually removal
scarring, analogous to hypocellular vitreous contraction. PVR of confluent PVC and anterior vitreous cortex (AVC) in the
is somewhat of a misnomer; there is minimal mitotic activity frontal plane (FP); there is no spherical core vitreous in these
in PVR, and antimetabolites have been shown to be ineffec- cases. It is important to recall that the vitreous base has no
tive; in fact, the disease is more like a keloid. To emphasize the volume and cannot be “shaved” as surgeons often state; it is
hypocellular nature of this process, the authors suggest taking a zone of adherence between the peripheral vitreous and the

144
CHAPTER 16 ■ Proliferative Vitreoretinopathy 145

retina. Often surgeons state that they perform vitreous base behavior, especially if the patient has another functional eye,
dissection; this is simply not the case, although so-called has medical problems, or is very elderly. Iris neovasculariza-
shaving of the peripheral vitreous close to the retina surface tion, glaucomatous cupping, and severe uveitis can influence
is essential. Circumferential vitreous collagen fibers shortened the decision of operability as well.
by hypocellular contraction pull the equator inward; these Cases with extremely recent proliferative activity and an
fibers are contiguous with the vitreous base and rarely remov- inflammatory component have a worse prognosis than inac-
able by peeling. Anterior-posterior–oriented vitreous col- tive cases (19). In such cases, a period of observation and
lagen formerly contiguous with the vitreous base shortened subconjunctival steroids should precede surgery (19; R.G.
by hypocellular contraction causes what the principal author Michels, personal communication). A definite life cycle as
initially identified as anterior loop traction; resection of this described by S. J. Ryan characterizes this reparative or scar-
component of anterior PVR is essential to reduce the anterior ring process. Recurrent proliferation is reduced if reoperation
pull on equatorial retina. Epiretinal membranes (ERMs) are is delayed until the proliferation stabilizes and inflammation
always behind to the posterior margin of the vitreous base; is minimal. Proliferative inactivity can be judged by the pres-
peeling peripheral PVR membranes is a misnomer as well. ence of minimal cells and flare, waning conjunctival hyper-
Scarring in the subretinal space can occur in many emia, comfort, and lack of further progression of ERMs.
physical configurations, as discussed later (see “Subreti-
nal Proliferation”). A placoid configuration can create an
SURGICAL SEQUENCE AND TECHNIQUES
inverted star fold configuration. Subretinal bands (strands)
occur presumably as tubes of RPE cells proliferate along a
Man ag e m e n t o f t h e Le n s o r In t rao cular Le n s
fibrin strand; these then contract, causing an extended den-
dritic configuration. If the band is circumferential at the The crystalline lens may be retained in most cases of poste-
midretinal level, it can create an annular configuration with rior PVR but may require removal in the presence of anterior
a resultant closed cone retinal detachment. PVR to allow adequate dissection of the anterior vitreous and
prevent cyclitic membrane formation. Endocapsular lensec-
tomy should be utilized if the eye is extremely inflamed;
CASE SELECTION otherwise, phaco-vit should be used to facilitate posterior
containment of silicone oil.
Vit re ct o m y Ve rsus Scle ral Bucklin g If a posterior chamber IOL is present, it should be
retained. Selected anterior chamber and iris plane lenses must
Vitrectomy should be thought of as a mechanical approach
be removed. Implant removal can occasionally result in an
to a mechanical problem caused by retinal traction. It has no
intraoperative hemorrhage or corneal and iris damage and
known prophylactic benefit in the prevention of PVR nor is
compromise the surgical result. Corneal incisions, viscoelas-
it an approach to be utilized only after several scleral buckles
tics, and haptic cutting minimize the incidence of intraop-
are tried. The authors utilize 25-gauge vitrectomy techniques
erative hemorrhage. Cut haptics can be left if bleeding is
without scleral buckling for practically all cases of retinal
anticipated because of fibrous scarring around the haptic.
detachment secondary to PVR.
The hypotony that occurs during lens removal can theoreti-
cally cause choroidal hemorrhage.
Op e rab ilit y
Extensive star folds, even in a closed cone (funnel) configuration, COMPARTMENTALIZATION
are usually operable. If extensive membrane peeling has been
performed, the recurrent proliferation may be more adherent to Serum components and inflammation play a role in the cau-
the retinal surface. Excessive retinopexy probably causes RPE sation and acceleration of PVR. A source tissue produces
and glial reproliferation, while retinal surface dissection proba- growth factors, which then target substrate tissues. The tar-
bly causes glial proliferation. If extensive subretinal placoid pro- get tissue (retina) is in the same compartment as the source
liferation is present, this is often inoperable. Redundant retina in PVR, but on remote surfaces in proliferative diabetic retin-
can simulate diffuse subretinal placoid proliferation, making opathy (vascular endothelial growth factor [VEGF], iris and
clinical assessment difficult. The presence of large retinal breaks trabecular meshwork). Viscoelastics, fibrin, inflammation,
and extensive surface proliferation was thought of as inoperable and blood can be thought of as culture media enhancing PVR.
before the advent of epiretinal and subretinal dissection, inter- The lens or IOL creates a two-compartment eye, prolonging
nal drainage of subretinal fluid (SRF), retinectomy, and silicone the cell culture–like environment and providing a migration
oil. In addition to the mechanically inoperable configurations, substrate. Removal of the lens in PVR cases increases the
there are many patient-based and biologic decisions related to egress rate of cytokines, cells, and serum components such
operability. If the patient has an apparently operable mechani- as fibronectin through the trabecular meshwork. Similarly,
cal configuration but has had many recurrences after vitrec- gases and silicone produce sequestration of cells and factors
tomy, surgery might be contraindicated because of biologic at the retinal-bubble interface, enhancing PVR.
146 SECTION IV ■ Disease-Sp ecific Managem ent

Fig u re 1 6 .1 ■ Th e AVC an d
PVC un d erg o h yp ocellular con -
traction of th e collag en fib ers
resulting in confluence and an FP
sheet. The FP com ponent should
be rem oved before forceps m em -
brane peeling of star folds.

VITRECTOMY can be done with the vitreous cutter, if broad, or delamination


scissors, if narrow (Fig. 16.2). The 25-gauge ILM forceps can
The AVC is removed first, along with all vitreous attachments to be used to peel posterior ERMs in an anterior direction to the
cataract and trauma wounds, and the iris. Very low suction force posterior edge of the vitreous base. Blunt dissection with the
using the proportional suction control should be utilized to pre- vitreous cutter and endoilluminator can be used to separate
vent retinal breaks from suction-induced vitreoretinal traction. vitreous and ERM from peripheral retina. Delamination scis-
The highest cutting rates should be used to increase fluidic sors can be used to delaminate or segment the circumferen-
stability. The PVC is frequently in contact with the AVC and tial component (Fig. 16.3).
removable in a single step. A posterior vitreous detachment
is almost always present in PVR cases. Hypocellular vitreous Ep ire t in al Me m b ran e s
contraction causes the PVC and AVC to come together in a
“frontal plane” configuration (Fig. 16.1). “Core vitrectomy” is a Membrane peeling can cause retinal breaks and hemorrhage
misnomer; a core configuration does not exist in PVR eyes. implicated in the recurrence of cellular migration, contrac-
tion, and proliferation. Often, the ERMs peel easily with none
of these problems and allow complete release of tangential
An t e rio r Pro life rat ive Vit re o re t in o p at h y traction (Fig. 16.4). End-grasping forceps membrane
Anteroposterior vitreous fibers normally extend from the peeling is preferable to using pics or forceps with one blade
retina at the posterior edge of the vitreous base to the pars under the ERM because the end-grasping forceps contact
plana, ciliary body, and iris. Frequently, these fibers undergo the ERM, not the retina. Use of forceps without pics reduces
hypocellular vitreous contraction and pull the equatorial ret- the incidence of iatrogenic retinal breaks. The principal author
ina anteriorly into a circumferential fold. The author (S.C.) refers to this approach as inside-out forceps membrane peel-
first described this phenomenon as anterior loop contraction ing; it is the same direction of dissection used for epimacular
in 1975. A circumferential equatorial ring of ERM or con- membranes, ILM, as well as scissors segmentation and delami-
tracted equatorial vitreous is typically present in PVR. Hypo- nation. The senior author initially developed diamond-coated,
cellular contraction of the AVC and PVC is also common 20-gauge forceps for membrane peeling, then developed
in PVR cases. Collectively, these structures are known conformal forceps, and more recently transitioned to Alcon
as anterior PVR. They are frequently incorrectly termed 25-gauge DSP ILM forceps with conformal tips. The senior
“vitreous membranes” or the “vitreous base.” Anterior PVR author has not used pics since 1978. Alcon 25-gauge
must be relieved to allow retinal reattachment. DSP ILM forceps are ideal for this technique. The end-
Scleral depression by the assistant facilitates anterior PVR grasping forceps should be applied to the apparent epicenter
dissection. The radial component of anterior PVR dissection of a star fold, or the outer margin only if clearly visible. It is
CHAPTER 16 ■ Proliferative Vitreoretinopathy 147

Fig ure 1 6 .2 ■ The radial (anter-


op osterior) com p onen t of ante-
rior PVR is also called anterior loop
traction. It can b e resected with
curved scissors if it is not b road
enough to perm it transaction with
the vitreous cutter. The 25-gauge
cutter is p referred b ecause the
com p act tip facilitates access.

usually best to use the forceps to peel toward the periphery Retinectomy may be required if forceps membrane peeling,
because central retina is much stronger than peripheral segmentation, or delamination of the ERM causing a star
retina. Peripheral ERMs may extend to the vitreous base; once fold, fixed fold, or zone of retinal-retinal adherence cannot
lifted from the retina surface, they can be removed with the be completed. A specific attempt must be made to release
vitreous cutter often while removing anterior loop traction. the ERM causing circumferential traction at the equator by
If the membranes are tightly adherent, they should be seg- using scissors segmentation or delamination. If this cannot
mented and/or delaminated with 25-gauge curved scissors. be safely accomplished because of tight adherence to the

Fig u re 1 6 .3 ■ Curved scissors


can be used to segm ent or delam -
inate ERM and condensed circum -
ferential fib ers on the eq uatorial
surface of the retina.
148 SECTION IV ■ Disease-Sp ecific Managem ent

Fig ure 1 6 .4 ■ Inside-out forceps


m em b ran e p eelin g with Alcon
25-gauge DSP ILM forceps is used
to rem ove ERMs. Pics, viscodissec-
tion, and forceps with one b lad e
under the ERM are m ore likely to
dam age the retinal surface.

retinal surface, this portion of ERM can be left intact and visualized and the sharp angulations have been converted to
supported by the very broad and high buckle or retinectomy rounded contours, dissection is complete.
performed. Frequently, dense ERM can be delaminated from
Sub re t in al Pro life rat io n
the retinal surface with curved delamination scissors with
the blades parallel to the retinal surface. Segmentation of the Many anatomic forms of subretinal proliferation occur, just
denser portions of an ERM using the curved scissors is very as there is variability in the configuration of ERMs. The term
effective in releasing traction and tends to be underutilized “strands” is a misnomer because of the frequent occurrence
(Fig. 16.5). When the entire surface of the retina can be of placoid elements.

Fig u re 1 6 .5 ■ Insid e-out seg -


m en tation of d en se ERM with
curved scissors is very effective
and tends to be underutilized.
CHAPTER 16 ■ Proliferative Vitreoretinopathy 149

Fig ure 1 6 .6 ■ Dendritic subreti-


nal p roliferation can b e rem oved
with forceps through a preexisting
retinal defect.

Placoid subretinal proliferation, if moderate in extent, internal drainage of SRF, fluid-air exchange, and completion
is managed by attempting internal drainage of SRF, fluid- of SRF drainage. The diamond-coated, end-opening forceps,
air exchange, and completion of SRF drainage. If the retina or 25-gauge DSP forceps are pushed through the retina over
becomes attached, subretinal surgery, and/or scleral buckling the densest part of the subretinal proliferation (Fig. 16.7)
are not required. If there is extensive proliferation, subretinal and used to grasp and remove the tissue (Fig. 16.8). Care
forceps removal is required. must be taken to avoid entrapping the retina in the forceps
Dendritic proliferation can usually be managed by or damaging the RPE or choroid. Frequently, the dendrite
forceps removal (Fig. 16.6) or segmentation, followed by will break, releasing the traction.

Fig ure 1 6 .7 ■ Dendritic subret-


inal p roliferation can be rem oved
with forceps after punch-through
retin o to m y an d sp re ad in g t o
enlarg e the op ening and g rasp
the “strand.”
150 SECTION IV ■ Disease-Sp ecific Managem ent

Fig ure 1 6 .8 ■ Sequential regrasping and use of the endoillum inator as a fulcrum enable long
subretinal dend rites to be com pletely rem oved.

Extension of dendritic proliferation in a rather posterior, anterior to the circumferential cut, reducing postoperative
circumferential configuration gives rise to an annular ring con- hypotony and peripheral PVR recurrences. Unless retina
figuration. After completion of the vitrectomy and inside- is incarcerated in a trauma wound or transscleral drain-
out forceps membrane peeling, the scissors are placed in age site from prior scleral buckling, retinectomy should be
the subretinal space through an existing retinal break or a performed with air, perfluoron (PFO), or silicone oil in the
punch-through retinotomy is made with the forceps in the eye. The author uses the term “incremental retinectomy” to
retina over the subretinal annulus. Scissors allow the sur- make the point that it is usually better to do just enough
geon to transect the annulus at a considerable distance from retinectomy to reattach the retina, which is best accom-
the retinal defect used to gain entry to the subretinal space. plished with a surface tension agent in the eye. If the retinec-
The endoilluminator can be used to illuminate the subretinal tomy reaches 270 degrees, it is usually better to extend for a
space and to contact and palpate the retina to determine if full 360 degrees because the remaining unresected quadrant
the traction has been alleviated. At times, it is necessary to usually contracts postoperatively.
transect the annulus in several locations. If extensive mem-
brane is present in the subretinal space, a 90-degree or greater Th e Re at t ach m e n t Exp e rim e n t
circumferential retinectomy can be performed to allow visu-
alization and access to the subretinal space, although this The reattachment experiment is defined as the combination of
is rarely required. Direct puncture (punch-through) retin- internal drainage of SRF, fluid-air exchange, and completion
otomies can allow access to very posterior subretinal pro- of internal drainage of SRF in order to determine the need
liferation. Internal drainage of SRF, fluid-air exchange, and for further dissection. In contrast to the normally low tran-
completion of SRF drainage followed by endophotocoagula- sretinal pressure gradient, a somewhat higher gradient is cre-
tion and air-silicone exchange are required in most of these ated, forcing a moderately stiffened retina against the RPE.
severe PVR cases. Air infusion through the infusion cannula and SRF aspiration
through the soft-tip cannula should be utilized for this step.
The authors use the Alcon 25-gauge soft-tip extrusion can-
RETINECTOMY nula for drainage of SRF and all exchanges except silicone
oil and PFO injection and silicone oil removal. On occa-
For three decades, the principal author has advocated cir- sion, this step will uncover posterior areas of ERM not seen
cumferential retinectomy rather than Machemer’s technique before the retinal cone was opened up. Forceps membrane
of relaxing retinotomy. Retinectomy removes all tissue peeling, delamination, or segmentation of these now visible
CHAPTER 16 ■ Proliferative Vitreoretinopathy 151

Fig ure 1 6 .9 ■ Interface vitrectom y is defined as positioning the cutter port just p osterior to air,
perfluoron (PFO), or silicone oil so that residual vitreoretinal traction can be rem oved.

membranes can be performed under air, further releasing strength and contraction forces of periretinal membranes
tangential traction. exceed the strength of the retina and the normal transretinal
On occasion, further vitreoretinal traction will be pressure gradient by a factor of approximately 100.
uncovered by the reattachment experiment. This is an ideal
situation for interface vitrectomy (vitrectomy under air). The
Pe rfl uo ro carb o n Liq uid s
port is positioned in the remaining vitreous, outside the air
bubble with the port midway between the retina and the vit- PFO can be used to stabilize the retina during ERM dis-
reous (Fig. 16.9). Moderate to small amounts of subretinal section. The term “tamponade” is inappropriate; PFO is
air appearing during the reattachment experiment indicate immiscible in fluid and therefore immiscible in the retina
that the traction has not been completely eliminated. Until and SRF. It provides 1.67× inertial and gravitational stabi-
the remaining traction on the retina is relieved, it is impos- lization. The MedOne dual-bore 25-gauge cannula is ideal
sible to remove subretinal air unless the eye is refilled with for injecting PFO. The optimal method for injecting PFO
infusion fluid. It is better to leave the subretinal air in place is to attach a short length of tubing between the dual-bore
and proceed with further peeling, segmentation or delami- cannula and a syringe filled with half PFO and half BSS.
nation, subretinal surgery, or retinectomy. The reattachment The BSS eliminates dead space, enabling use of all the PFO.
experiment, if successful, should be followed by laser treat- If a giant break is present, PFO will enter the subretinal space
ment of all retinal breaks and then air-gas exchange or air- if insufficient ERM has been removed. PFO-air exchange fol-
silicone exchange. lowed by air-silicone exchange is usually a better approach
At times, retinal defects will enlarge greatly or sponta- than PFO-silicone exchange. PFO-air exchange should be
neous defects will occur as the reattachment experiment is performed by placing the tip of the soft-tip cannula just
performed. This will allow the release of tangential traction below the air-liquid interface and slowly advancing the
on foreshortened retinas and permit retinal conformation cannula posteriorly following the interface down to the
with the pigment epithelium. These defects occasionally are retina. Removing all liquid vitreous, SRF, and infusion fluid
extensive, leading to inoperability, but in many cases, they before PFO is removed prevents posterior slippage of giant
are helpful in allowing retinal reattachment. breaks and reaccumulation of SRF as the PFO-air exchange
The basic mechanical problem in PVR retina is retinal is performed.
foreshortening combined with the requirement of the retina to Steep folds can result from retinal incarceration in the
conform to the inner surface of the rigid eye wall. The tensile sclera at the prior sclerotomies, trauma sites, or previous
152 SECTION IV ■ Disease-Sp ecific Managem ent

Fig ure 1 6 .1 0 ■ If the reattachm ent experim ent using internal drainage of SRF and fluid-air
exchange results in subretinal air, additional vitrectom y, forceps m em brane peeling, segm entation,
or delam ination should be perform ed to reattach the retina. If the subretinal air persists, increm en-
tal retinectom y should be used. All tissue anterior to the circum ferential cut should be rem oved to
reduce hypotony and reproliferation, which distinguishes this technique from relaxing retinotom y.

drainage sites. If a single fold occurs, a retinal cut made the retinal reattachment experiment should be attempted
perpendicular to the long axis of the fold will release the after the completion of periretinal membrane dissection and
traction analogous to a plastic surgeon releasing a scar. Mul- stopped immediately if subretinal air appears. Incremental
tiple steep folds radiating from an epicenter of periretinal retinectomy with endodiathermy applied to retinal vessels
membrane may require retinectomy including or surround- to be transected should be alternated with incremental addi-
ing the epicenter. In general, radial folds require circumfer- tional drainage of SRF. This process should cease only when
ential cuts just as the typical equatorial, circumferential fold the retina is so rigid that it cannot be mechanically unfolded
requires a radial cut. or it becomes completely reattached (20).
Diffuse retinal foreshortening that cannot be managed by The contributing factors in achieving intraoperative reat-
membrane peeling, segmentation, or delamination requires tachment include surface tension of the fluid-air interface, size
large circumferential retinectomies (Fig. 16.10). Other sur- of the retinal breaks, retinal stiffness, and the contour of the
geons use the term “relaxing retinotomies,” indicating that eye wall. When periretinal membrane dissection is completed
no retina is removed with their approach. The retinectomy and the reattachment experiment applied, further mechani-
approach was developed by the author to remove all tissue cal forces on the retina may become apparent. Mechanically
anterior to the circumferential “relaxing” retinotomy. The holding the retina in position as reattachment is attempted
advantages of complete removal are less hypotony from epi- can be useful at times (21). This can be accomplished with
ciliary tissue, less iris neovasculization from ischemic tissue, the endoilluminator, vitreous cutter, or a soft-tip cannula.
and less anterior movement of a silicone oil bubble due to
contraction of the circumferential ring of pars plana tissue.
Scle ral Bucklin g
Cases requiring large circumferential retinectomies are man-
aged similar to giant breaks with PVR using long-term sili- The principal author has not used buckles for PVR cases
cone oil tamponade instead of scleral buckling. for almost two decades and never revises buckles if pres-
The need for large retinectomies frequently cannot be ent. Buckles and buckle revision increase pain, inflamma-
anticipated in the office. Excessive retinectomies can result tion, refractive error, hypotony, strabismus, ptosis, operating
from underestimation of the effectiveness of periretinal time, and labor cost due to longer procedures. High scleral
dissection methods. Large retinectomies are needed only buckles coupled with marked retinal foreshortening may
after failure of the reattachment experiment. In all cases, push the posterior retina away from the RPE.
CHAPTER 16 ■ Proliferative Vitreoretinopathy 153

The patient must be informed about possible corneal problems,


VISCOELASTICS AS VITREOUS
glaucoma, and the possibility of silicone removal.
SUBSTITUTES Silicone oil is used for rhegmatogenous confinement
for breaks not identified during surgery and new breaks due
Viscoelastics are non-Newtonian fluids and have exceedingly
to recurrent PVR. The authors prefer fluid-air exchange fol-
low surface tension (tamponade) effects. They are pseudo-
lowed by air-silicone exchange to fluid-silicone exchange
plastic and will slowly deform and pass through a small retinal
unless IOL fogging occurs. IOL fogging is due to contact
break very easily. In addition, viscoelastics act as a sustained
between the air and the IOL; therefore, YAG capsulotomy
release medium for cells and cytokines. For these reasons,
or surgical capsule rupture and disruption of the AVC is
viscoelastics have no place in the PVR management.
required. PFO is an excellent alternative to fluid-air exchange
if IOL fogging occurs. Some surgeons believe incorrectly that
SILICONE OIL IOL fogging is due to the use of silicone IOLs; it occurs with
all IOLs but is more common with silicone lenses because of
Silicone oil was first utilized without vitreous surgery higher Posterior Capsular Opacification (PCO) and therefore
techniques as a surface tension and dissection device, pro- higher YAG rates and greater thermal mass.
ducing ERM peeling by forcing the retina back with con- Silicone oil should be used for rhegmatogenous confine-
current drainage of SRF (22–27). This method will not ment often without retinopexy when retinectomy is required
work with more extensive ERM or subretinal proliferation. for areas of highly adherent ERMs. The authors often utilize
Recent studies with improved silicone oil (28–37) have not retinectomy without retinopexy when reoperating cases ini-
shown the retinal toxicity noted in previous years. Infe- tially operated by other surgeons that created a large number
rior peripheral iridectomy as developed by Ando allows of retinal breaks attempting to peel highly adherent mem-
aqueous humor to pass from the ciliary processes into branes. Long-term surface tension management is a crucial
the anterior chamber to supply metabolites to the corneal tool in these difficult cases. Excessive retinopexy causes PVR
endothelium (38). recurrences and fibrin syndrome; permanent silicone oil elim-
Aqueous access to the cornea reduces silicone keratop- inates the need for retinopexy, hence the terms “rhegmatog-
athy. Keratopathy occurs in less than 10% of cases with long- enous confinement” and “retinopexy avoidance.” There is an
term oil (S. Charles, data presented at the Vitreous Society, undue emphasis on silicone oil removal, which often results
1996, using Adatomed 5,000 cs). Reduction of lower molec- in redetachment. Silicone oil complications are minimal when
ular weight components, by using higher average molecu- the oil is confined behind an IOL. The emulsification and
lar weight and therefore higher viscosity, and purification to complication rates for 1,000 cs and 5,000 cs oil are equal but
remove metal ions dramatically reduce silicone emulsifica- injection and removal are 5× faster with 1,000 cs oil.
tion, corneal changes, and glaucoma. Many surgeons incorrectly believe that removal of sili-
The physical effects of silicone are based on its inter- cone oil is required to reoperate for PVR or epimacular mem-
facial (surface) tension resulting from its immiscibility with branes. The authors find it advantageous to operate “under oil”
water (aqueous). The ability to restore a transretinal pressure just as they operate under air and PFO. This technique is called
gradient is a function of the size and shape of the retinal interface vitrectomy. Vitreous removal, forceps membrane peel-
break and the tangential forces (stiffness) on the retina. Vis- ing, retinectomy, and drainage of SRF are all effective under oil,
cosity is not a factor in the transretinal pressure gradient, air, or PFO. Advantages include less surgical trauma, much less
and the silicone-aqueous interfacial tension is 25 dyne/cm, operating time, and realistic assessment of residual forces on
which is much less than that of the air (gas) fluid interface, the retina. It is crucial to recall that silicone oil has one third
which is 70 dyne/cm. the surface tension of gas or air; often retinas attached under
Long-term rhegmatogenous confinement (tamponade) air in the operating room are noted to be partially detached
may obviate the need for retinopexy, therefore reducing repro- under oil on the first postoperative day. Two-port, 25-gauge
liferation in response to tissue destruction (39). Long-term technique is ideal for reoperation “under” silicone oil. The
tamponade limits the rhegmatogenous component from algorithm is to sequentially remove SRF and top up the oil
contraction-created breaks and those missed at the time of with the MedOne 25-gauge viscous fluid injection cannula
surgery. Silicone may act to prevent the wetting of the retinal when the eye gets soft; then repeat the process until the ret-
surface by cytokines implicated in reproliferation (J. Lean, ina is attached and the IOP is normal.
personal communication). Silicone can reduce reproliferation
by retinopexy avoidance but appears to have no direct role in
suppressing or stimulating reproliferation. Silicone may also RETINAL SUTURES, TACKS,
increase reproliferation because of sequestration of cells and AND INTENTIONAL INCARCERATION
cytokines at the silicone-retinal interface and prevention of
access of pharmacologic agents to the retina surface. Permanent fixation of the retina to the sclera has been utilized
Retinectomy and multiple previous procedures using to prevent inward curling or posterior sliding of large retinal
gas are relative indications for using silicone in PVR. flaps or the edges of retinectomies. These methods have the
154 SECTION IV ■ Disease-Sp ecific Managem ent

Fig ure 1 6 .1 1 ■ After all of the SRF has been d rained, all retinal defects are surrounded by three
rows of confluent endophotocoagulation.

risk of retinal or choroidal hemorrhage, tissue damage, and Re t in o p last y


the creation of retinal breaks. Retinal tacks are more safely
Retinoplasty using a synthetic adhesive such as cyanoacrylate
and easily utilized than sutures or intentional retinal incar-
(46,47) could theoretically replace all vitreous substitutes
ceration. They can be repositioned as well as removed later.
and retinopexy. An ideal retinal patch should be very flex-
Different materials have been used for the construction of
ible, more elastic than the retina, less permeable than the
retinal tacks: polyacetal (40), steel (41), titanium (42,43),
retina, and not adhere to the RPE. Aqueous humor (the ideal
and certain alloys (H.G.). Sutures and incarceration are time
vitreous substitute) could be used in place of gas or silicone
consuming and frequently place additional traction on the
if a retinal patch were available.
retina. No permanent fixation method can prevent surface
contraction and detachment. Tacks cause bleeding, reprolif-
eration, distortion, and secondary breaks and have not been
utilized by the principal author for 30 years. RESULTS

Results are highly dependent on case selection, surgical meth-


RETINOPEXY ods, and surgical experience. Using conventional techniques
of vitreous surgery, about 50% to 85% of cases achieved
After the reattachment experiment has been successfully com- long-term retinal reattachment (48–50). With the previously
pleted, endophotocoagulation can be applied to retinal breaks. described methods and case selection criteria, 90% of cases
Postreattachment retinopexy (S.C.) permits treatment of both can be repaired surgically, with over 74% remaining attached
the retina and the RPE for stronger adherence. It may limit over the long term (51,52). About 50% of the cases sustain
the migration of RPE cells implicated in PVR. Because retin- improved vision better than 5/200, but many cases require
opexy is implicated in PVR formation (44,45) and inflamma- more than one procedure (53).
tion can cause retinal-retinal adherence, retinopexy should be
limited to the retinal breaks or suspected break areas. Panreti-
COMPLICATIONS
nal photocoagulation has never been used by the authors for
PVR because it can cause reproliferation and fibrin syndrome.
Pro life rat ive Vit re o re t in o p at h y Re curre n ce
Similarly, the authors never use many rows of treatment but
rather surround retinal defects with a moderately wide row of A recurrence of PVR with secondary retinal separation is
confluent laser (Fig. 16.11) unless silicone is used for retin- the most frequent complication. In approximately one half
opexy avoidance in highly active or inflamed cases. of the recurring cases, further forceps membrane peeling
CHAPTER 16 ■ Proliferative Vitreoretinopathy 155

air-silicone exchange, and endolaser photocoagulation can 10. Campochiaro PA, Jerdan JA, Glaser BM. Serum contains
cause successful reattachment. chemoattractants for human retinal pigment epithelial cells.
Arch Ophthalmol 1984;102:1830.
Although the use of intraocular steroids and 5-fluo-
11. Campochiaro PA, Glaser BM. Platelet-derived growth factor is
rouracil and its derivatives (54–61) for PVR cases has been chemotactic for human retinal pigment epithelial cells. Arch
described, therapeutic efficacy has not been proven. It appears Ophthalmol 1985;103:576.
that the migration phase of the RPE cells is more important 12. Pastor JC. Proliferative vitreretinopathy: an overview. Surv
than the proliferation phase in the development of PVR. As Ophthalmol 1998;43(1):3–18.
13. Campochiaro PA. Pathogenis mechanisms in proliferative vit-
these drugs have a short half-life, better delivery systems are
reoretinopathy. Arch Ophthalmol 1997;115:237–241.
required than those presently available. Cytotoxic agents have 14. Kohno T, Sorgente N, Ryan SJ. Fibronectin distribution at
low therapeutic ratios, which is a significant problem, espe- the vitreoretinal interface. Invest Ophthalmol Vis Sci 1983;
cially when coupled with the highly variable drug elution rate 24(Suppl.):240.
of sustained-release delivery systems. Other systemic medica- 15. Kohno T, Sorgente N, Patterson R, et al. Fibronectin and
Laminin distribution in bovine eye. Jpn J Ophthalmol 1983;
tions, such as colchicine, have no proven use in the treatment
27:496.
or prevention of PVR and should be avoided. Further under- 16. Anderson DH, Stern WH, Fisher SK, et al. The onset of pigment
standing of the molecular processes of cellular and pericellular epithelial proliferation following retinal detachment. Invest Oph-
contraction might eventually lead to directed pharmacologic thalmol 1981;21:10.
therapy in the future (akin to anti-VEGF therapy in choroidal 17. Machemer R. Pathogenesis and classification of massive pen-
retinal proliferation. Br J Ophthalmol 1978;62:737.
neovascularization). Toxic medications should not be utilized
18. Machemer R, Aaberg TM, Freeman HM, et al. An updated clas-
in human patients on a “what if” approach. sification of retinal detachment with proliferative vitreoretin-
opathy. Am J Ophthalmol 1991;112:159–165.
19. Charles S. Presentation at 11th Annual Estelle Doheny Eye
In fl am m at io n Foundation Conference. Los Anvitreouses, CA, September
Because retinal breaks are exclusively treated with laser 1979.
20. Han DP. Relaxing rentinotomies and retinectomies. Surgical
endocoagulation, the usual exudative detachment and fibrin results and predictors of visual outcome. Arch Ophthalmol 1990;
syndrome associated with two or three rows of cryopexy are 108(5):694–697.
eliminated. A very rare patient will experience severe inflam- 21. Glaser BM. A new method of treating giant tears without
mation and transient iris neovascularization and go on to patient rotation with the use of intraocular gas and silicone
(develop) phthisis bulbi. This rare complication could be from oil. Presentation at Annual Meeting of American Academy of
Ophthalmology. Atlanta, GA, October 2, 1985.
anterior segment necrosis syndrome, sympathetic uveitis, or 22. Cibis PA, Becker B, Okun E, et al. The use of liquid silicone in
some yet unexplained etiology but is usually due to exces- retinal detachment surgery. Arch Ophthalmol 1962;68:590.
sive retinopexy or operating on inflamed eyes. Subconjunc- 23. Cibis PA. Vitreous transfer and silicone injections. Trans Am
tival repository steroids without systemic steroids are used in Acad Ophthalmol Otolaryngol 1964;68:983.
every PVR case unless the patient is a steroid responder. 24. Okun E. Intravitreal surgery utilizing liquid silicone: a long-
term followup. Trans Pac Coast Otolaryngol Ophthalmol Soc
1968;49:141.
References 25. Okun E, Arribas NP. Therapy of retinal detachment compli-
cated by massive preretinal fibroplasia (long-term followup
1. Machemer R, Van Horn D, Aaberg TM. Pigment epithelial of patients treated with intravitreal liquid silicone). In: New
proliferation. Am J Ophthalmol 1978;85:181. Orleans Academy of Ophthalmology Symposium on Retina and
2. Van Horn DL, Aaberg TM, Modene R, et al. Glial cell prolif- Reena Surgery. St. Louis: Mosby, 1969:278–293.
eration in human retinal detachment with massive periretinal 26. Scott JD. The treatment of massive vitreous retraction by the
proliferation. Am J Ophthalmol 1977;84(3):383. separation of preretinal membranes using liquid silicone. Mod
3. Machemer R. Role of the pigment epithelium in vitreous Probl Ophthalmol 1975;15:285.
pathology. Trans Ophthalmol Soc UK 1975;95:402. 27. Scott JD. A rationale for the use of liquid silicone. Trans
4. Machemer R, Laqua H. Pigment epithelium proliferation in Ophthalmol Soc UK 1977;97:235.
retinal detachment (massive penretinal proliferation). Am 28. Labelle P, Okun E. Ocular tolerance to liquid silicone: an
J Ophthalmol 1975;80:1. experimental study. Can J Ophthalmol 1972;7:199.
5. Machemer R. Massive periretinal proliferation (MPP). 1. Pigment 29. Ober RR, Blanks JC, Ogden TE. Experimental retinal tolerance
epithelial proliferation. Mod Prob Ophthalmol 1975;15:227. to liquid silicone. Retina 1983;3:77.
6. Mandelhorn M, Machemer R, Fineberg E, et al. Proliferation 30. Ober RR, Ryan SJ, Minckler DS, et al. Ocular tolerance to liq-
and metaplasia of intravitreal retinal pigment epithelium cell uid silicone: an experimental study. Invest Ophthalmol Vis Sci
autotransplants. Am J Ophthalmol 1975;80:227. 1980;19(Suppl.):47.
7. Laqua H, Machemer R. Clinical-pathological correlation in mas- 31. Meredith TA, Lindsey DT, Edelhauser HF, et al. Electroretino-
sive periretinal proliferation. Am J Ophthalmol 1975;80:913. graphic studies following vitrectomy and intraocular oil injec-
8. Laqua H, Machemer R. Glial cell proliferation in retinal tion. Br J Ophthalmol 1985;69:254.
detachment (massive penretinal proliferation). Am J Ophthal- 32. Momirov D, Van Lith GHM, Zivojnovic R. Electroretinogram
mol 1975;80:602. and electro-oculograms of eyes with intravitreously injected
9. Campochiaro PA, Jerdan JA, Cardin A, et al. Vitreous aspi- silicone oil. Ophthalmologica 1983;186:183.
rates from patients with proliferative vitreoretinopathy stimu- 33. Freeman KD, Gregor ZJ. Electrophysiological responses after
late retinal pigment epithelial cell migration. Arch Ophthalmol vitrectomy and intraocular tamponade. Trans Ophthalmol Soc
1985;103(9):1403–1405. UK 1985;104:129.
156 SECTION IV ■ Disease-Sp ecific Managem ent

34. Foerster M, Esser J, Laqua H. Silicone oil and its influence on 49. Machemer R. Massive periretinal proliferation: a logical approach
electrophysiologic findings. Am J Ophthalmol 1985;99:201. to therapy. Trans Am Ophthalmol Soc 1977;75:556.
35. Abrams GW, Azen SP, McCuen BW II et al. Vitrectomy with 50. Sternberg P, Machemer R. Results of conventional vitreous
silicone oil or long acting gas in eyes with severe PVR: results surgery for proliferative vitreoretinopathy. Am J Ophthalmol
of additional and long-term follow-up. Silicone Study report 1985;100:141.
11. Arch Ophthalmol 1997;115(3):335–344. 51. Charles S. Methodology and research on proliferative vitreo-
36. Diddie KR, Azen SP, Freeman HM, et al. Anterior proliferative retinopathy. Presented at Retina Sodety Meeting. Cleveland,
vitreoretinopathy in the silicone study. Silicone Study Report OH, November 1985.
Number 10. Ophthalmology 1996;103(7):1092–1099. 52. Charles S. Vitreous surgery for proliferative vitreoretinopathy.
37. Vitrectomy with silicone oil or pleurofluorocarbon gas in eyes Presented at Vitreous Society Meeting. Orlando, FL, October
wit severe PVR: results of a randomized clinical trial. Silicone 1985.
report 2. Arch Ophthal 1992;110(6):780–792. 53. Lewis H, Aaberg TM. Causes of failure after repeat vitrectomy
38. Ando F. Intraocular hypertension resulting from pupillary for recurrent proliferative vitreoretinopathy. Am J Ophthalmol
block by silicone oil. Am J Ophthalmol 1985;99:87. 1991;15;111(1):15–19.
39. Charles S. Vitrectomy for retinal detachment. Trans Ophthalmol 54. Machemer R, Sugita G, Tano Y. Treatment of intraocular pro-
Soc UK 1980;100(4):542. liferations with intravitreal steroids. Trans Am Ophthalmol Soc
40. Ando F, Kondo J. A plastic tack for the treatment of reti- 1979;77:171.
nal detachment with giant tear (letter). Am J Ophthalmol 55. Tano Y, Chandler D, Machemer R. Treatment of intraocular
1983;95:260. proliferation with intravitreal injection of triamcinolone ace-
41. De Juan E, Hickingbotham D, Machemer R. Retinal tacks. Am tonide. Am J Ophthalmol 1980;90:810.
J Ophthalmol 1985;99:272. 56. Chandler DB, Rozakis G, De Juan E, et al. The effect of triam-
42. Aaberg TM. Presentation at Duke Advanced Vitrectomy Course. cinolone acetonide on a refined experimental model of prolif-
Durham, NC, April 1985. erative vitreoretinopathy. Am J Ophthalmol 1985;99:686.
43. Aaberg TM. Presentation at American Retina Society Meeting. 57. Stern WH, Lewis GP, Erickson PA, et al. Fluorouracil therapy
Cleveland, OH, November 1985. of proliferative vitreoretinopathy after vitrectomy. Am J Oph-
44. Campochiaro PA, Koden IH, Vidaurri-Leal JS, et al. Cryother- thalmol 1983;96:32.
apy enhances intravitreal dispersion of viable retinal pigment 58. Blumenkranz MS, Ophir A, Claflin AL, et al. Fluorouracil for
epithelial cells. Arch Ophthalmol 1984;103:434. the treatment of massive periretinal proliferation. Am J Oph-
45. Glaser BM, Vidaurri-Leal J, Michels RG, et al. Cryotherapy thalmol 1982;94:458.
during surgery for giant retinal tears enhances dispersion of 59. Santana M, Wiedemann P, Kinmani M, et al. Daunomycin in
viable retinal pigment epithelial cells. Ophthalmology 1993 the treatment of experimental proliferative vitreoretinopathy-
Apr;100(4):466–470. retinal toxicity of intravitreal daunomycin in the rabbit. Graefes
46. Hida T, Sheta SM, Proia AD, et al. Experimental transvitreal Arch Clin Exp Ophthalmol 1984;221:210.
cyanoacrylate retinopexy in a primate model. Am J Ophthalmol 60. Wiedemann P, Sorgente N, Bekhor C, et al. Daunomycin in
1987 Jun 15;103(6):782–789. the treatment of experimental proliferative vitreoretinopathy-
47. McCuen BW II, Hida T, Sheta SM. Transvitreal cyanoacrylate effective doses in vitro and in vivo. Invest Ophthalmol Vis Sci
retinopexy in the management of complicated retinal detach- 1985;26:719.
ment. Am J Ophthalmol 1987 Aug 15;104(2):127–132. 61. Wiedemann P, Sorgente N, Kirmani M, et al. Daunorubicin in
48. Machemer R, Laqua H. Alogical approach to the treatment of mas- the treatment of experimental MPP-effective doses in vitro and
sive periretinal proliferation. Ophthalmology 1978;85(6):584. in vivo. Invest Ophthalmol Vis Sci 1983;24(Suppl.):241.
C H AP TER
17
EPIMACULAR MEMBRANES
AND VITREOMACULAR
TRACTION SYNDROME

Membranes on the macular surface can result unproven. Although the terms “macular pucker” and “surface
from several pathogenic mechanisms with the wrinkling retinopathy” emphasize retinal distortion, some
common theme of tissue damage and subse- patients have marked improvement in postoperative vision in
quent repair (1–5). Epimacular membranes spite of persistent retinal distortion and metamorphopsia.
(EMMs) are hypocellular, largely collagen structures. EMMs
are also called macular puckers, cellophane maculopathy,
surface wrinkling retinopathy, and premacular fibrosis. Each HISTORY
of these names has certain deficiencies, hence the currently
most widely accepted name, EMMs. The typical EMM patient experiences a relatively rapid loss
of vision accompanied by metamorphopsia over a period
of several weeks, followed by relative stabilization of visual
PATHOGENESIS function. In spite of this typical history, it is common prac-
tice for doctors to advise a patient with a recent history of
The so-called idiopathic type of EMM is caused by glial visual loss to, for example, the 20/50 level that he or she
migration and proliferation from a defect in the internal should wait until the vision is reduced to 20/80 or worse
limiting membrane (ILM) created by a posterior vitreous before considering surgery. In fact, the vision will usually
separation (6). Retinal breaks, retinopexy, photocoagulation, stabilize at a visual level at or near that noted on initial pre-
inflammation, and vascular disease (7) can lead to glial pro- sentation. Because visual results are better with better pre-
liferation (8–12) on the retinal surface. Retinal pigment epi- operative vision and shorter duration, it is better practice to
thelial cells (13,14) can migrate through a retinal break and make a decision on surgical intervention on the first visit.
proliferate on the retinal surface just as they do in proliferative
vitreoretinopathy (PVR). EMMs can be thought of as localized
glial or retinal pigment epithelium (RPE)–induced PVR. CASE SELECTION

As with all surgical procedures, the decision to operate is a


ETIOLOGY OF VISUAL LOSS multifactorial process based on symptoms, extent of visual
loss, visual needs, status of the other eye, age, duration,
Hypocellular contraction of the EMM causes nonrhegmatog- medical status, and the presence of other ocular diseases.
enous elevation of the macula thought by the authors to be There is no substitute for ethical, sound clinical judgment in
responsible for a major fraction of the associated visual loss. making the decision to operate.
Fluid under the macula is universally seen on optical coher- The principal author’s visual acuity threshold for sur-
ence tomography (OCT). Reversible macular edema secondary gery has moved from 20/200 to 20/40 in selected cases, as
to macular separation from the fluid pumping mechanism of the methodology has improved. A patient with preoperative
the RPE contributes to visual loss as well. Although it is widely vision of 20/40 can and should be operated if the patient
stated that traction on the ILM can produce macular edema, it is is significantly symptomatic, is in good health, is relatively
unclear what the mechanism would be and the concept remains young, and understands the issues. Specific visual acuity

157
158 SECTION IV ■ Disease-Sp ecific Managem ent

levels are less important than symptomatology and impact with marked adherence of the vitreous to the macula (28–30).
on activities of daily living for recommendation of surgery This entity is known as vitreomacular traction syndrome.
for EMMs. Duration is a relative rather than absolute crite- Spectral domain OCT invariably demonstrates vitreomacu-
rion because cases of 10 years’ duration have had significant lar traction in these cases. When operating on these cases,
visual improvement following surgery. The visual improve- care must be taken to avoid tearing the fovea by imbrica-
ment in long-duration cases is presumably because the mini- tion of the vitreous into the port of the vitreous cutter. Fine
mal amount of subretinal fluid present in these cases leads curved scissors can be used to delaminate the PVC from the
to minimal irreversible photoreceptor degeneration, just as fovea prior to any removal of the vitreous (Fig. 17.1).
is the case in central serous retinopathy. Macular edema,
except in the vascular disease subgroup, is probably sec- No n rh e g m at o g e n o us Pro life rat ive
ondary to macular elevation, typically reversible and not a Vit re o re t in o p at h y
contraindication to vitreoretinal surgery. Knowledge that the
patient had poor vision before the membrane occurred is an Some patients have multiple star folds from PVR in addition
absolute contraindication to surgery. The slow recovery of to an epimacular component. Removing these additional
vision after retinal reattachment surgery coupled with the epiretinal membranes is a stimulus for recurrent PVR and
typical 1-month onset of EMM makes it difficult to make a is unnecessary unless they are causing macular elevation or
surgical decision in this situation. Patients with severe hered- distortion.
itary photoreceptor degeneration or a previous central reti-
nal artery occlusion frequently have wrinkling of the retinal Ne e d fo r Vit re ct o m y at t h e Tim e
surface without an epiretinal membrane because of marked o f Me m b ran e Pe e lin g
decrease in retinal thickness. Surgery is contraindicated in
The principal author initially suggested the concept of
these situations (15–27).
membrane peeling without vitreous removal but stopped
advocating this approach after several hundred cases
because the patients complain bitterly of floaters and
SURGICAL SEQUENCE
in-office fluid-gas exchange cannot be performed if the
AND TECHNIQUES patient develops a retinal detachment. If the vitreous has
been removed, a postoperative retinal detachment can be
Vit re o m acular Tract io n Syn d ro m e
managed by in-office, two-needle, fluid-gas exchange and
The posterior vitreous cortex (PVC) is rarely adherent laser retinopexy. Anterior vitreous cortex removal is prob-
to typical EMMs. Some patients have macular elevation ably correlated with an increased incidence of posterior
secondary to hypocellular contraction of the PVC combined subcapsular cataract probably related to fluid turbulence.

Fig u re 1 7 .1 ■ Curved scissors


are used to resect the PVC in vitre-
om acular traction syndrom e cases
to p revent avulsion of the fovea
caused by im brication of the taut
PVC into the cutter.
CHAPTER 17 ■ Epim acular Mem branes and Vitreom acular Traction Syndrom e 159

Fig ure 1 7 .2 ■ Inside-out forceps


peeling is initiated at the epicenter,
which is the densest portion of the
EMM and the focal point of striae.
Pics and looking for an ed g e can
result in retinal d am ag e an d are
not required with this m ethod.

Avoiding the anterior vitreous cortex may reduce post- for surface grasping and must have precise alignment of the
operative posterior subcapsular cataract. blade tips. Eckardt developed effective end-opening forceps
that are preferred to diamond-coated forceps because the
diamonds are recessed from the tips of the forceps blades.
Ep ire t in al Me m b ran e Re m o val
Conformal forceps were then developed by the princi-
Machemer developed the concept of membrane peeling in pal author because Eckardt-type forceps have square cor-
1972 soon after his introduction of vitrectomy. Originally, ners that can tear the nerve fiber layer or cause bleeding.
peeling was performed with a bent needle. O’Malley sub- The tips of conformal forceps have a radius of curvature
sequently developed the concept of using a rounded, angu- which matches that of the retina. The authors use 25-gauge
lated instrument he called a pic to perform the peeling. The Alcon disposable, DSP ILM forceps (Fig. 17.3) for all cases.
principal author and the late Ron Michels popularized the The Alcon 25-gauge MVR pic is used by some surgeons if the
pic method. Bent needles and pics require the presence of membrane is very smooth and taut (so-called glassy), but
a visible outer margin of the EMM, frequently called an the authors rarely find this step necessary when using DSP
“edge,” unless a slit is made in the membrane. Searching
for an edge creates the risk of making a retinal break. The
principal author developed the concept of inside-out for-
ceps membrane peeling initially because of the difficulty of
finding an edge in certain cases. In contrast to Machemer’s
outside-in membrane peeling method, inside-out peeling
is initiated by surface grasping the EMM with end-opening
forceps (Fig. 17.2). Although the senior author formerly rec-
ommended making a slit in the apparent center of the EMM
using the microvitreoretinal (MVR) blade or a sharp-tipped
pic, this is not necessary with the Alcon 25-gauge ILM DSP
forceps. The center of the membrane can be identified by
noting the orientation of radial striae, the most elevated
retinal region, the most opaque region of the membrane,
and the relative movement of the membrane with respect
to the retina induced by lateral movement of the forceps or
MVR blade tip. End-opening forceps with diamond coating Fig u re 1 7 .3 ■ Alcon 25-g aug e ILM forcep s are id eal for
were developed by the senior author and Hans Grieshaber rem oving EMMs.
160 SECTION IV ■ Disease-Sp ecific Managem ent

Fig ure 1 7 .4 ■ Inside-out forceps


m em b ran e p eeling is p erform ed
tan g ential to the retinal surface,
u sin g a circular m o tion wh ile
observing the fovea so that foveal
avulsion will not occur.

forceps. Membrane peeling should be accomplished by over 1,600 consecutive cases; all other series report a 5%
moving the forceps tangentially along the surface of the to 6% incidence of retinal breaks. The peripheral retina
retina in a circular fashion (Fig. 17.4). If the membrane should be inspected at the end of the case, and all retinal
tears, it can be regrasped without removing any membrane breaks with elevated edges should be managed with fluid-
from the forceps because microteeth will penetrate several air exchange, air-gas exchange with 25% SF6, and laser
layers of membrane and facilitate removal of the membrane endophotocoagulation around the break(s). Laser alone can
through the pars plana as well. The surgeon should always be used if the edges are not elevated, but there is an increased
observe the fovea during the peeling process rather than risk of immediate postoperative retinal detachment. Scleral
focusing on the forceps in order to prevent tearing the fovea. buckling is not required in these cases.
Areas of stronger adherence to the ILM can be detected by
noting fine fibers being lifted from the retinal surface dur- Man ag e m e n t o f Co e xist e n t Cat aract
ing the peeling process. If prominent striae are still noted
after peeling the epiretinal membrane, ILM peeling with the Cataract surgery should be performed prior to vitreoretinal
25-gauge DSP forceps must be performed. Kampik intro- surgery if a visually significant cataract is present. Although
duced the idea that ILM peeling reduces the incidence of this approach increases cost and subjects the patient to an
recurrent membranes, and the authors have validated this additional procedure, it enables a superior view at vitrec-
observation. Scissors delamination with fine curved scis- tomy and optimal refractive status. EMM surgery requires
sors rather than peeling is utilized if strong adherence to excellent visualization and should not be attempted if
the fovea, vessels, or any region of the retina is noted during 3+ or greater nuclear sclerosis or a significant posterior
the peeling process (Fig. 17.5). If there are marked folds, subcapsular cataract is present. The fact that vitreous sur-
the blunt, polished end of the vitrectomy instrument can be gery causes progression of preexisting nuclear sclerotic
used to gently push the retinal folds into better position, a cataract in a large number of cases should be taken into
method referred to by the principal author as “burnishing.” account while considering when to remove the lens. Opti-
Moderate-sized peeled or delaminated membrane pieces mal refractive outcomes are obtained when cataract sur-
should be removed through the pars plana with the forceps. gery incorporating all of the latest technology developed to
If the membrane is very large or dense, it should be removed enable emmetropia is utilized. Although phaco-vit is now
with the vitrectomy probe. recommended by the authors and many other surgeons for
many clinical scenarios for the management of vitreoretinal
disease combined with cataract or in anticipation of nuclear
Man ag e m e n t o f Re t in al Bre aks
sclerosis progression, it is difficult to obtain optimal refrac-
No posterior retinal breaks occurred in the principal tive outcomes. It is often a better approach to perform vitre-
author’s prospective, consecutive, single-surgeon series of oretinal surgery first if the view is adequate and to have an
CHAPTER 17 ■ Epim acular Mem branes and Vitreom acular Traction Syndrom e 161

Fig u re 1 7 . 5 ■ If t h e EMM
is extrem ely ad h eren t, scisso rs
delam ination with curved scissors
should b e used to avoid tearin g
the retina.

experienced cataract surgeon perform refractive phaco later Rh e g m at o g e n o us Re t in al De t ach m e n t


when nuclear sclerosis progression becomes significant.
The author’s series had a similar incidence of postoperative
retinal detachment to that reported by other authors (5%).
This complication is related to inadvertent force on the vitre-
VISUAL RESULTS ous base during vitreous removal, instrument introduction
forces, and incarceration of vitreous in the sclerotomies. This
About 40% of the patients in the senior author’s series
complication can be decreased by using high-quality cut-
had improved vision to 20/40 or better. Approximately
ters, low suction force (<80 mm Hg), highest possible cut-
56% improved to 20/80 or better. Over 85% of the patients
ting rates, and avoiding pulling the cutter away when cutting
improved two lines or greater in visual acuity. It is noted that
is activated. The retinal periphery should be inspected with
those with greater visual loss (1/200 to 5/200) preoperatively
the indirect ophthalmoscope at the end of the case and any
improve to approximately the 20/200 level postoperatively.
retinal breaks treated with endolaser, laser indirect ophthal-
Those in the 20/200 region preoperatively typically return to
moscope (LIO), or cryopexy and fluid-gas exchange with
the 20/40 to 20/50 level postoperatively. Those with 20/50
air or 25% SF6, if the edges are elevated. Alternatively, the
vision preoperatively usually return to the 20/20 to 20/25
contact wide-angle lens can be utilized to perform limited
level. Visual results have improved significantly since ILM
peripheral vitrectomy with simultaneous observation of the
peeling was combined with epiretinal membrane peeling in
retinal periphery for presence of retinal tears. If found, endo-
all cases.
photocoagulation can be performed.

Pro life rat ive Vit re o re t in o p at h y Re curre n ce


COMPLICATIONS
If PVR was successfully repaired prior to the development of
Re t in al Bre aks a membrane, it can be stimulated to recur by EMM surgery.
Usually, the PVR-induced redetachment can be successfully
No posterior retinal breaks were caused in the author’s series repaired with repeat vitrectomy and silicone oil, but little
of over 1,100 consecutive vitrectomies for EMM. All other improvement in pre-EMM surgery vision is then obtained.
authors have reported an incidence of 5% to 7%. Although
the author’s series was not randomized to outside-in versus
Cyst o id Macular Ed e m a
inside-out peeling, this marked difference probably indicates
that inside-out forceps membrane peeling is a safer method The author’s series noted a preoperative cystoid macular
than using pics and outside-in peeling. edema (CME) incidence of 3.2%. This incidence was based
162 SECTION IV ■ Disease-Sp ecific Managem ent

on clinical observation, as routine fluorescein angiography The observed increase in the incidence of nuclear
was not performed. The McDonald et al. series reports a 3% sclerosis associated with EMM surgery is thought by the
incidence of CME. authors to result from increased oxygen tension in the former
vitreous cavity after vitrectomy. The incidence of progression
of nuclear sclerosis has been reported to range from 10% to
En d o p h t h alm it is
100%. This wide variation could be accounted for by many
The author’s series of over 1,400 cases had no cases of post- factors, including follow-up period, definition of clinical sig-
operative endophthalmitis, while the literature reports an nificance, postoperative refraction, operating time, intraop-
average incidence of 0.7%. The author uses a one-piece sur- erative cataract surgery, light source UV and IR content, and
gical drape that is folded over the lid margins and a micro- unknown factors.
scope drape in all cases. Subconjunctival Tobramycin and
Ancef are injected at the end of the case. High-quality infu-
sion fluid (Alcon BSS Plus) is used in all cases. Absorbable References
sclerotomy sutures are not used. 1. Kampik A, Kenyon KR, Michels RG, et al. Epiretinal and
vitreous membranes: comparative study of 56 cases. Arch Oph-
thalmol 1981;99:1445–1454.
Re curre n ce o f Ep im acular Pro life rat io n 2. Pearlstone AD. The incidence of idiopathic preretinal macular
gliosis. Ann Ophthalmol 1985;17:378.
Approximately 1.6% of treated EMM cases resulted in clini- 3. Scudder MJ, Eifrig DE. Spontaneous surface wrinkling retin-
cally significant recurrent EMMs (31–34) in the author’s opathy. Ann Ophthalmol 1982;94:44.
series of over 1,100 cases. Successful reoperation can usu- 4. Sidd RJ, Fine SL, Owens SL, et al. Idiopathic preretinal gliosis.
ally be performed with sustained visual improvement. The Am J Ophthalmol 1982;94:44.
5. Wise GN. Clinical features of idiopathic preretinal macular
recurrence rate emphasizes the reparative nature of the pro-
fibrosis. Am J Ophthalmol 1975;79:349.
cess and the damage to the retinal surface associated with 6. Roth AM, Foos RY. Surface wrinkling retinopathy in eyes
membrane peeling. All patients probably have some repair of enucleated at autopsy. Trans Am Acad Ophthalmol Otolaryngol
the retinal surface after membrane peeling. The criterion for 1971;75:1047.
defining a recurrence has not been established in the litera- 7. Wise GN. Clinical features of idiopathic preretinal macular
fibrosis. Am J Ophthalmol 1975;79:349.
ture. In some patients, the folds disappear completely and a
8. Kenyon KR, Michels RG. Ultrastructure of epiretinal membrane
recurrence can easily be determined if folds recur. In other removed by pars plana vitreoretinal surgery. Am J Ophthalmol
patients, the recurrent membrane causes macular elevation 1977;83(6):815.
with minimal striae and the membrane is more difficult to 9. Clarkson SG, Green WR, Massof D. A histopathologic review of
visualize. Decreased visual function is usually the best indi- 168 cases of preretinal membrane. Am J Ophthalmol 1977;84:1.
10. Green WR, Kenyon KR, Michels RG, et al. Ultrastructure of
cator of clinically significant recurrences if the patient had
epiretinal membranes causing macular pucker following reti-
initially experienced gradual visual improvement. Visually nal reattachment. Trans Ophthalmol Soc UK 1979;99:63.
significant cataract must be ruled out before the visual loss 11. Kampik A, Green WR, Michels RG, et al. Ultrastructural fea-
can be attributed to the macula. tures of idiopathic progressive epiretinal membrane removed
by vitreous surgery. Am J Ophthalmol 1981;90:797.
12. Michels RG. A clinical and histopathological study of epiretinal
Re t in al Wh it e n in g membranes affecting the macula and removed by vitreous sur-
gery. Trans Am Ophthalmol Soc 1982;80:580.
Immediate postoperative retinal whitening occurs at the 13. Laqua H. Pigmented macular pucker. Am J Ophthalmol 1978;
removal site in a significant percentage of cases. This disap- 86(1):56.
pears spontaneously in several days and does not seem to 14. Lindsey PS, Michels RG, Luckenbach M, et al. Ultrastructure
of epiretinal membrane causing retinal starfold. Ophthalmology
affect the visual outcome. It is probable that this phenom- 1983;90:578.
enon represents ganglion cell axoplasmic flow disruption. 15. Michels RG. Surgical treatment of macular pucker. In: Stirpe
Michels noted that retinal whitening is present preopera- M, Convers M, Blankenship G, et al., eds. Advances in vitreoreti-
tively in a significant number of cases presumably because of nal surgery. Filia, Rome; 1991.
traction on the nerve fiber layer exerted through the ILM. 16. Shea M. The surgical management of macular pucker. Can J
Ophthalmol 1979;2:110.
17. Wilkinson CP. Recurrent macular pucker. Am J Ophthalmol
1979;88(6):1029.
Cat aract 18. Machemer R. A new concept for vitreous surgery: two instru-
Posterior subcapsular cataracts after vitrectomy are largely ment techniques in pars plana vitrectomy. Arch Ophthalmol
1974;92:407–441.
avoidable. Posterior subcapsular cataracts can be caused by
19. Trese MT, Chandler DB, Machemer R. Macular Pucker: prognos-
using low-quality infusion fluids such as lactated Ringer’s tic criteria. Graefes Arch Clin Exp Ophthalmol 1983;221:12–15.
solution rather than glutathione bicarbonate Ringer’s solu- 20. Margherio RR, Cox MS, Trese MT, et al. Removal of epimacular
tion (Alcon BSS Plus). membranes. Ophthalmology 1985;92:1075–1083.
CHAPTER 17 ■ Epim acular Mem branes and Vitreom acular Traction Syndrom e 163

21. McDonald HR, Verre WP, Aaberg TM. Surgical management 28. Smiddy WE, Michels RG, Glaser BM, et al. Vitrectomy for
of idiopathic epiretinal membranes Ophthalmology 1986;93: macular traction caused by incomplete vitreous separation.
978–983. Arch Ophthalmol 1988;106:624–628.
22. de Bustros S, Thompson JT, Michels RG, et al. Vitrectomy for 29. Melberg NS, Williams DF, Balles MW, et al. Vitrectomy for
idiopathic epiretinal membranes causing macular pucker. Br J vitreomacular traction syndrome with macular detachment.
Ophtalmol 1988;72(9):692–695. Retina 1995;15:192–197.
23. Rice TA, de Bustros S, Michels RG, et al. Prognostic factors in 30. Bellhorn MB, Friedman AH, Wise GN, et al. Ultrastructural
vitrectomy for epiretinal membranes of the macula. Ophthal- and clinicopathologic correlation of idiopathic preretinal mac-
mology 1986;93:602–610. ular fibrosis. Am J Ophthalmol 1975;79:366–373.
24. Charles S. General posterior segment techniques. In: Charles S, 31. Michels RG. Vitreous surgery for macular pucker. Am J Oph-
ed. Vitreous microsurgery. Baltimore: Williams & Wilkins; thalmol 1981;92:628.
1987:98. 32. Michels RG. Surgery of macular pucker. In: Fine SL, Owens
25. Margherio RR. Discussion of Michels RG: vitrectomy for macu- SL, eds. Management of retinal vascular and macular disorders.
lar pucker. Ophthalmology 1984;91:1387–1388. Baltimore: Williams & Wilkins; 1983:120–130.
26. Gass JDM. Steroscopic atlas of macular diseases, 4th ed., St Louis: 33. Michels RG. Vitrectomy for macular pucker. Ophthalmology
Mosby; 1997:288–299, 938, 940, 944. 1984;91:1384.
27. Barr CC, Michels RG. Idiopathic nonvascularized epiretinal 34. Michels RG. Macular pucker. In: Fraunfelder FT, Roy FH, eds.
membranes in young patients:report of six cases. Ann Ophthal- Current ocular therapy, 2nd ed. Philadelphia: W.B. Saunders;
mol 1982;14:335–341. 1984:440–442.
C H AP TER
18
MACULAR HOLES

Until 1991, it was thought that macular holes represents a serious problem in discussing these cases with
were untreatable and their pathogenesis was vitreoretinal surgeons.
unknown. In that year, Kelly and Wendel devel- Surgical success should be defined as clinical disap-
oped the concept of using vitrectomy and fluid- pearance of the hole, reconstitution of the foveal anatomy
gas exchange to treat these patients (1). Initially there was on OCT, and marked improvement in vision.
great skepticism about the treatment, but the facts prevailed.
The initial goal was to “seal” the hole much as is done for
rhegmatogenous retinal detachment and eliminate the cuff INDICATIONS FOR MACULAR
of subretinal fluid that surrounds the hole. When it was HOLE SURGERY
noted that many holes actually disappeared after surgery, and
patients obtained near-normal vision, the skeptics again did Freeman et al. have shown that smaller, more recent holes
not believe it. Fortunately, it is now accepted that complete have the best prognosis. Size is much more important than
disappearance of the hole is the usual outcome after surgery. duration with respect to closure rates; duration plays no
role if size is controlled for. Duration probably plays a role
in visual success in successful cases. Most surgeons do
PATHOGENESIS not suggest repairing holes secondary to chronic macular
edema from diabetic retinopathy, venous occlusive disease,
It was widely believed that a posterior vitreous detachment cystoid macular edema after cataract surgery, or secondary
(PVD) pulled a full-thickness piece of tissue out of the mac- to uveitis. Traumatic macular holes are a complex decision-
ula, probably as a result of or during a saccade. Electron making process because of the high likelihood of associ-
microscopy of the so-called operculum, which occurs in ated photoreceptor, retinal pigment epithelium (RPE), and
these cases, has shown that few photoreceptors are present optic nerve damage and because many will spontaneously
(2,3). Gass published a hypothesis for the pathogenesis (4,5) close within 1 to 2 months. If there is good evidence that
of macular holes that has remained viable with minimal the macular hole is the only significant damage, these cases
modifications since the introduction of high-resolution opti- can be considered for surgery after a reasonable period of
cal coherence tomography (OCT). The concept is that radial observation.
vitreous fibers remaining on the perimacular surface after
apparent posterior vitreous separation contract and slowly
tear the macula in a circumferential fashion. The Mueller OCULAR COHERENCE TOMOGRAPHY
cells may play a role as well according to Gass. Many observ-
ers have noted that vitreous is attached to the optic nerve OCT is invaluable in the evaluation of macular hole patients
after an apparent posterior vitreous separation characterized both preoperatively and after surgery. It can be difficult to
by the presence of a prepapillary (Weiss) ring. Because of distinguish partial-thickness holes from full-thickness holes
these observations, the event might better be termed “delam- clinically. Some holes are very small at the internal limiting
ination of the posterior vitreous cortex.” The senior author membrane (ILM) level but much larger at the level of the outer
terms the cuff of fluid “the traction cuff.” Sjaarda has shown retina; others have the opposite configuration. Vitreomacular
using scanning laser ophthalmoscope (SLO) microperimetry traction and epimacular membranes can be seen with care-
that the actual extent of scotoma extends far beyond the mar- ful OCT examination. Some holes have no cuff, are ellipti-
gin of the cuff (6). The vitreous that is attached to the inner cal, and are caused by typical epimacular membranes rather
margin of the macular hole is rarely contiguous with the than vitreous. These holes may be thought of as secondary
vitreous attached to the midperipheral retina. Terminology holes and round holes with a cuff above as primary or
164
CHAPTER 18 ■ Macular Holes 165

classic holes. The Gass classification system using Stage I, etc. overlie a large, full-thickness outer layer hole producing an
is no longer used by the authors now that OCT is available. appearance suggesting a macular cyst. Spectral domain OCT
Macular holes are now classified as partial thickness or full is essential in the evaluation of macular disease. Time domain
thickness. Diameter is a major factor in driving outcomes OCT is no longer adequate. Charteris has shown that 30%
and can be accurately measured with OCT. Whether there to 50% of partial-thickness outer macular holes (Stage I on
is a PVD or not is irrelevant clinically as an attempt must be Gass’ Classification) spontaneously close if observed over
made to create a PVD in all cases and ILM peeling removes the long term. A multicenter clinical trial done before the
any adherent posterior vitreous cortex. Some patients have availability of OCT or ILM peeling did not demonstrate a
apparent closure after surgery but modest visual recovery; benefit of operating on a partial-thickness hole to prevent
some of these cases have edema or subretinal fluid as shown progression to a full-thickness hole. Neither ultrasound nor
by Kaiser, while others have a persistent defect in the outer OCT can predict whether a partial-thickness hole will prog-
retina or disruption on the outer segment layer of the fovea ress to become a full-thickness hole, and status of the other
on spectral domain OCT. eye is not helpful because holes are bilateral less than 10%
of the time.
Size of the macular hole is the only preoperative factor
MACULAR HOLE SURGERY that has been shown to drive surgical closure rates; duration
is not a determining factor if size is controlled for. Although
Macular holes were untreatable until Neil Kelly devel- duration has an influence on visual outcomes, assessment of
oped the concept of core vitrectomy followed by fluid-gas the subhole RPE using spectral domain OCT and confocal
exchange using an isoexpansive mixture of air and SF6 or autofluorescence will probably prove to be more effective in
C3F8 gas. His goal was to reattach the cuff of subretinal fluid predicting visual outcomes in longer duration holes. Some
around a full-thickness macular hole and therefore eliminate studies have shown reasonable visual outcomes after operat-
the relative scotoma that surrounds the absolute scotoma. ing macular holes of relatively long duration.
Serendipitously he soon discovered that the macular hole Macular holes originating from macular cysts due to
often closed with remarkable improvement in vision. Many chronic macular edema typically have relatively poor visual
leading surgeons initially discounted his discovery, but for- outcomes because of macular ischemia related to underly-
tunately it has become the standard of care. The prevailing ing diabetic retinopathy or retinal vein occlusion. Similarly,
thought at the time was that substantial neural tissue was macular holes arising from chronic inflammatory macular
avulsed by a PVD; an operculum was often seen, validating edema have a relatively poor visual prognosis.
this notion. Examination of surgically removed operculums Traumatic macular holes can spontaneously close in the
using the electron microscope revealed very little neural first 4 to 6 weeks suggesting that a period of observation
tissue; most of the operculum proved to be glial tissue, should precede the determination of operability. An affer-
explaining why substantial visual improvement was pos- ent pupillary defect (APD) should be considered a relative
sible. Subsequently, OCT demonstrated restoration of near- contraindication to surgical repair because it indicates asso-
normal or normal foveal anatomy in successfully operated ciated optic nerve damage. As mentioned above, assessment
cases. Clearly, “closure” of macular hole is quite different of the subhole RPE using spectral domain OCT and confocal
than the term “closure” when used in the context of retinal autofluorescence will probably prove to be more effective in
detachment repair. predicting visual outcomes. Presence of a choroidal rupture
Much emphasis is placed on the substantial work of in the papillomacular bundle is a relative contraindication to
Don Gass, which involved clinical observation of the evolu- repair of a traumatic macular hole; often these patients will
tion of macular holes, theoretical considerations concerning have an APD.
the pathogenesis, and a classification system. Although all The authors’ management of partial-thickness holes
surgeons agree that the posterior vitreous cortex is somehow has changed over the years. The senior author began
involved and the elasticity of the ILM plays a role, the patho- operating lamellar holes after Arthur Willis introduced
genesis of macular holes remains unknown. Presurgical clas- the concept of macular hole prevention surgery. Macular
sification, even using spectral domain OCT, is incapable of hole prevention surgery was no longer performed after
reliably determining if residual vitreous cortex is adherent to the results of the Macular Hole Study and the Chart-
the retinal surface rendering the classification system virtu- eris paper were published. The authors’ current practice
ally useless. Macular holes are three times as common in is to determine the need for surgery on partial-thickness
females as they are in males, but there is no explanation for holes based on symptoms, particularly metamorphopsias
this interesting observation. from the epiretinal membranes (ERMs) that commonly
OCT is essential to determine if the hole is partial thick- surround lamellar macular holes. The goal is to improve
ness or full thickness; not infrequently clinical examination visual function, not prevent a full-thickness hole because
is inadequate to detect a very small, full-thickness hole at progression to a full-thickness hole cannot be predicted.
the base of a large-diameter partial-thickness hole. Occa- The authors have determined in recent years that surface
sionally, a very small, inner layer, full-thickness hole will tension management using air or SF6 combined with ILM
166 SECTION IV ■ Disease-Sp ecific Managem ent

peeling is required to restore normal or near-normal foveal on OCT; edema-mediated retinal thickening coupled with
anatomy and improve or eliminate symptoms when operat- the elasticity of the ILM causes eversion of the hole mar-
ing on symptomatic partial-thickness holes. gins. Elimination of the edema facilitates restoration of near-
normal foveal anatomy, resolution of the edge eversion, and
approximation of the hole margins. The bubble dries out the
HOLE CLOSURE MECHANISMS retinal surface, which probably signals the astrocytes to heal
the hole days after approximation of hole margins via the
The conventional explanation of the role of surgical steps in lateral surface tension effect.
surgical hole closure is incomplete and, to an extent, prob-
ably incorrect. It is often stated that the role of PVD creation
is to eliminate vitreous traction on the macula, but, in fact, POSTERIOR VITREOUS DETACHMENT
the posterior vitreous is usually attached to the optic nerve CREATION METHODS
head, nasal, and midperipheral retina but not attached to
the macula or retina within the temporal arcades. Care- Many surgeons use a lateral (tangential) motion of the extru-
ful PVD creation reduces the likelihood of inferior retinal sion cannula or vitreous cutter to create a PVD. This method
breaks caused by interaction of the bubble with residual creates shear force at the vitreous base, potentially leading to
vitreous similar to what may occur with pneumatic retin- iatrogenic retinal breaks. A better method is to position the
opexy. The role of core vitrectomy is to enable the exchange vitreous cutter at the nasal, superior and inferior disk margins
of vitreous for a large gas bubble and to ensure that a thin with the port oriented away from the center of the disk and
layer of vitreous does not prevent contact of the gas bubble pull anteriorly (toward the cornea) using the vacuum-only
with the hole. mode (Fig. 18.1). The anterior-pull, disk-margin method
Virtually all surgeons agree that ILM peeling improves safely, reliably, and quickly produces a PVD using 25-gauge
closure rates, but why this is true has a complex answer. ILM cutters. It is a misconception that higher flow rates help pro-
peeling ensures removal of tangential traction due to resid- duce PVDs or that 25-gauge cutters cannot produce a PVD;
ual vitreous on the retinal surface, which, although rare, can it is all about technique.
occur even with an apparent PVD as evidenced by a Weiss
ring. In addition, ILM peeling guarantees successful removal
of ERMs that are occasionally present. ILM peeling increases INTERNAL LIMITING MEMBRANE
retinal elasticity by over 50%, which enables lateral surface PEELING
tension forces to close the hole as soon as the bubble comes
into contact with the hole. It is also likely that ILM peel- Unfortunately, many surgeons equate ILM peeling with
ing also initiates mechanical signaling to the astrocytes to indocyanine green (ICG) staining, staining with alterna-
heal the hole margins days after it is closed by lateral surface tive dyes, or particulate marking with triamcinolone. ICG
tension. has been shown to be toxic to the retina and RPE and may
Most surgeons use the term “tamponade” when describ- also be associated with increased intraoperative phototox-
ing the mechanism of action of air, gas, and silicone oil bub- icity (7). Many papers using ICG for ILM peeling report
bles. To some, tamponade means to “press,” but tamponade the typical 90% closure rate but unacceptable visual out-
is from the French and means to “seal.” It is more accurate comes. One factor driving dangerous and unnecessary
and descriptive to use the term “surface tension manage- ICG use is using noncontact optical systems such as the
ment” or “interfacial tension management” to describe the BIOM and EIBOS, which decrease both axial resolution
function of the bubble. It is obvious that a bubble elimi- and lateral resolution, making it more difficult to visual-
nates transhole flow, which is the reason it is used in retinal ize the ILM. There is anecdotal evidence for triamcino-
detachment repair. An additional and crucial function of the lone reducing closure rates and particles being trapped
bubble as Reppucci has pointed out is the lateral surface ten- within the hole or in the subretinal space. The authors use
sion effect. Surface tension forces act along an interface of Alcon 25-gauge DSP ILM end-grasping forceps (Fig. 18.2)
two immiscible substances effectively pulling the surface but never use pics, MVR blades, or membrane scrapers.
inward. Surface tension makes a droplet nearly spherical as Pics, MVR blades, and membrane scrapers often result in
it falls away from a faucet and a soap bubble spherical. Rep- unnecessary retinal surface damage as attempts are made
pucci describes the bubble as bridging the hole, but the key to find or construct an “edge.” Forceps are still required to
concept is that the bubble pulls the hole margins together actually peel the ILM when pics, scrapers, or MVR blades
immediately after surgery as soon as the patient is positioned are used to construct an edge, making this a two-step
with bubble in contact with the hole. Yet another function approach. An additional factor driving ICG use is using
of the bubble, as the senior author has pointed out, is the inadequate forceps incapable of true end grasping. For-
prevention of transretinal flow (uveal-scleral outflow). Tor- ceps used for ILM peeling should be end grasping and
nambe has introduced the hydration hypothesis making 25 or 23 gauge and should not be asymmetric or built
a note of the marked edema surrounding the hole as seen with a gripping surface extending more than 120 mm
CHAPTER 18 ■ Macular Holes 167

Fig ure 1 8 .1 ■ After core vitrectom y, a PVD is created using suction-only m ode on the vitreous
cutter, p ulling anteriorly (not tang entially) over the op tic nerve. Peeling sh ould extend to
the m id p erip hery, and the p erip heral retina should b e exam ined for p eeling-ind uced retinal
breaks.

Fig ure 1 8 .2 ■ Conform al forcep s, 20-gauge DSP forceps, or 25-gauge DSP forceps (preferred)
are used to peel residual vitreous, epim acular m em brane, and ILM. ILM is peeled 360 degrees;
pics, m em b rane scrapers, and ICG staining are not required with this technique.
168 SECTION IV ■ Disease-Sp ecific Managem ent

Fig ure 1 8 .3 ■ Com plete fluid-air exchange is followed b y air-gas exchang e.

along the blades. Reusable forceps rapidly lose the ability Some surgeons advocate silicone oil for patients who
to grip at the leading edge of the gripping surface as the cannot or will not position after surgery. This is a seriously
surface is eroded and the blades are warped outward by flawed concept because silicone oil has roughly one third the
use, cleaning, and sterilization. Reuse of disposable for- surface tension of a gas-fluid (actually gas-retina) interface.
ceps rapidly degrades the ability to grip at the leading A silicone oil bubble to a significant extent conforms to the
edge as well. hole and RPE in the base of the hole, reducing the likelihood
of closure.
Vitrectomy produces accelerated nuclear sclerosis
SURFACE TENSION MANAGEMENT progression rates in approximately 90% of cases, although
it does not produce cataract in cases with clear lenses
The principal author used C3F8 for many years thinking preoperatively, rarely the case with macular hole patients.
incorrectly that glial cells pulled the hole together and longer As Nancy Holekamp has shown, nuclear sclerosis progres-
term surface tension management would improve outcomes sion is most likely due to a permanent increase in oxy-
but converted to SF6 in 2006 after learning of Reppucci’s gen tension of 7 to 12 mm Hg secondary to removal of
surface tension concept and observing that holes that were ascorbate. Ascorbate, a powerful antioxidant, is actively
not closed on the first postoperative day rarely, if ever, closed secreted into the vitreous cavity, producing nine times the
(Fig. 18.3). Proof that glial cells do not pull the hole together serum level.
follows from the observations that normal or near-normal Combining cataract surgery with vitrectomy for macu-
foveal anatomy without a glial scar is the typical postopera- lar holes or even removing lenses with minimal or no cata-
tive OCT appearance (Fig. 18.4). ract is advocated by some surgeons, since, in their opinion,
Positioning is controversial, in part because of seman-
tics and also from inappropriate pandering to the patients
suggesting that positioning is not necessary with “their”
technique. If patients remained supine continuously after
surgery, the hole would never close because surface ten-
sion from bubble contact is required to pull the hole mar-
gins together. It is unknown how long it takes for enough
adherence to occur that exposure to fluid would not cause
the hole to reopen, but it is probably 1 to 3 days. Fur-
ther positioning is necessary in phakic eyes to prevent gas Fig ure 1 8 .4 ■ OCT after m acular hole surgery dem onstrates
cataract. near-norm al foveal anatom y with no glial scar.
CHAPTER 18 ■ Macular Holes 169

cataracts will progress after vitrectomy. The authors do not


POSTOPERATIVE POSITIONING
recommend phaco-vitrectomy technique for macular holes
because it produces less precise refractive outcomes and
Most surgeons recommend full-time or near full-time face-
increases the likelihood of anterior intraocular lens (IOL)
down positioning for the duration of a reasonably sized
subluxation postoperatively. Gas bubbles also increase
bubble (8). The authors believe in full-time, face-down
the likelihood of iris capsule adhesions (synechia) during
positioning for 1 week. The patients are allowed to walk,
the postoperative course. As described in Chapter 8, elective
work, read, watch TVs placed on the floor, ride in cars (not
macular surgery is best performed independently of phaco
drive), and use the treadmill or stationary bicycle. Although
surgery. If a cataract precludes adequate visualization for
the patients often ask, a head-down position does not inter-
ILM peeling, an experienced cataract surgeon can per-
fere with eating, showering, reading, or using the restroom.
form phacoemulsification with IOL implantation weeks
Face-down driving has consequences that extend beyond
prior to vitrectomy. IOL calculations in macular disease
vitreoretinal surgery.
are best done with the IOL Master instead of A-scan ultra-
sonography, since the former calculates axial length to the
RPE and is unaffected by macular holes or ERMs as the BIOLOGIC MODIFIERS
latter is.
Fibrin, serum, autologous platelet concentrate, thrombin,
whole blood, cryo precipitates, and transforming growth
ARCUATE RETINOTOMY factor beta (bovine, recombinant, and autologous) have
been used to accelerate glial proliferation in the hole (9–11).
Aneesh Nikhra suggested that a relaxing incision in the Sterile endophthalmitis, uveitis, inflammatory prolifera-
retina could be used to repair large, failed macular holes, tive vitreoretinopathy, and bacterial endophthalmitis have
and the principal author developed the arcuate retinotomy been reported after use of these agents. The authors and
procedure. This technique is never used in primary surgery many other surgeons no longer use these agents because
cases. Alcon disposable 25-gauge curved DSP scissors were of unproven efficacy and the risk factors noted previously.
used to make an arcuate retinotomy temporal to the macula. Freeman et al. showed no effect of serum in a controlled
The cut was made by separating the nerve fibers with special clinical trial.
care to avoid transecting nerve fibers. The arc length of the
retinotomy was 100 degrees centered around the temporal
horizontal meridian (Fig. 18.5). The arcuate retinotomy was RESULTS
made approximately 250 mm from the edge of the macu-
lar hole. Internal drainage of subretinal fluid was performed Results have been reported as high as 100% for small series
repeatedly to dry the macular hole. Drying the submacu- with cases selected for small, short-duration holes. The
lar RPE was necessary to allow surface tension to retain the authors believe that 90% is a reproducible success rate with
bridge of temporal retina in the appropriate position after SF6; 1 week of face-down positioning; and cortex, ERM, and
it was moved inward with the 25-gauge soft-tip cannula. ILM peeling (12).
Repositioning of the temporal retinal bridge resulted in a
vertically oval macular hole and widening of the retinotomy
at the end of surgery. The horizontal diameter of the hole References
markedly decreases at this point. Air-gas exchange with C3F8 1. Kelly NE, Wendel RT. Vitreous surgery for idiopathic macu-
or SF6 was the final step. In the authors’ series, which has lar holes. Results of a pilot study. Arch Ophthalmol 1991;109:
been submitted for publication, they achieved an 83% mac- 654–659.
ular hole closure rate in these patients who otherwise would 2. Madreperla SA, McCuen BW II, Hickinbotham D, et al. Clini-
copathologic correlation of surgically removed macular hole
be deemed inoperable. opercula. Am J Ophthalmol 1995;120:197–207.
3. Ezra E, Munro PM, Charteris DG, et al. Macular hole opercula.
Ultrastructural features and clinicopathological correlation.
Arch Ophthalmol 1997;115(11):1381–1387.
4. Gass JD. Idiopathic senile macular hole. Its early stages and
pathogenesis. Arch Ophthalmol 1991;106:654–659.
5. Gass JD. Reappraisal of biomicroscopic classification of
stages of development of a macular hole. Am J Ophthalmol
1995;119:752–759.
6. Sjaarda RN, Frank DA, Glaser BM, et al. Assessment of vision
in idiopathic macular holes with macular microperimetry using
the scanning laser ophthalmoscope. Ophthalmology 1993;
100(10):1513–1518.
Fig ure 1 8 .5 ■ OCT after arcuate retinotom y for large m acular 7. Gandorfer A, Aritoglou C, Kampik A. Toxicity of Indocyanine
hole showing retinotom y site. Green in vitreoretinal surgery. In: Meyer CH, ed. Vital dyes
170 SECTION IV ■ Disease-Sp ecific Managem ent

in vitreoretinal surgery. Developments in ophthalmology. Karger: 10. Ligget PE, Skolik SA, Horio B, et al. Human autologous serum
Basel, 2008:69–81. for the treatment of full thickness macular holes: preliminary
8. Tornambe PE, Poliner LS, Grote K. Macular hole surgery study. Ophthalmology 1995;102:1071–1076.
without face down positioning. A pilot study. Retina 1997;17: 11. Thompson JT, Smiddy WE, Williams GA, et al. Comparison of
179–185. recombinant transforming growth factor beta-2 and placebo
9. Glaser BM, Michels RG, Kupperman BD, et al. Transforming as an adjuvant agent for macular hole surgery. Ophthalmology
growth factor-beta 2 for the treatment of full thickness mac- 1998;105:700–706.
ular holes. A prospective randomized study. Ophthalmology 12. Thompson JT, Sjaarda RN, Lansing MB. The results of vitreous
1992;99:1162–1173. surgery for chronic macular hole. Retina 1997;17:493–501.
C H AP TER
19
DIABETIC RETINOPATHY

image to background ratio via rejection of light scattered by


MEDICAL ISSUES
the cornea, lens, and vitreous. In addition, lasers are more
efficient in stimulating the fluorescein fluorophore than
Diabetes is increasing in prevalence in the American population
filtered noncoherent light. The use of contact wide-angle
for many reasons (1,2). Diabetics are living longer because of
angiography lenses permits the diagnosis of preequatorial
dialysis, kidney and pancreas transplants, improved cardio-
retinal neovascularization that was previously difficult to
vascular management, and pharmacologic advances. The
diagnose (Fig. 19.1).
availability of high-calorie, high-carbohydrate, high-fat,
Optical coherence tomography has not only allowed
and high-sugar foods; fast food restaurants; cultural atti-
more precise diagnosis of diabetic macular edema but
tudes toward eating; poor role models; large serving sizes;
also permitted detailed observation of the vitreomacu-
and many other factors contribute to the problem (3). Joslin
lar interface and the presence of traction on the macular
observed that in 1912, there was no diabetes in the Pima
area that can be potentially relieved by vitrectomy surgery
Indian population; the incidence is now almost 70%. Fried-
(Fig. 19.2).
man and others have attributed this to the “thrifty gene,”
which enabled survival of man with “occasional” eating
rather the three-square meals that many believe we should
eat. It is of interest that beef, pork, egg, fast food, and dairy SCREENING
business interests support the nutrition community and
make very large political donations. If dieticians, teachers, Up to 70% of the known diabetic population in America
parents, doctors, nurses, ministers, and coaches eat improp- does not get annual eye exams. Many studies have utilized
erly, there are no positive role models. The diabetic, antiath- various methods to screen this population as well as monitor
erosclerosis, anticancer, weight-loss, longevity, feel-good, patients with no retinopathy or preclinically significant mac-
look-good, and fitness diet are virtually the same. If the doc- ular edema (pre-CSME) nonproliferative diabetic retinopa-
tor eats properly, exercises, and educates the family and staff thy (NPDR) at their last exam. It appears that color images
to do the same, a positive model is created for all. Everyone are somewhat more sensitive than monochrome. Digital
who comes into contact with the patient and family should cameras have decreased in cost while increasing in quality
support better health habits. and have almost replaced standard film and Polaroid images.
Nonmydriatic fundus cameras have proven to be very effec-
tive for diabetic screening. Screeners, optometrists, general
IMAGING ophthalmologists, and retinal specialists can evaluate the
images, if properly trained.
Fluorescein angiography was instrumental in understand-
ing the stages in the pathogenesis of diabetic retinopathy
and remains a valuable diagnostic tool to determine the RETINAL THICKNESS MEASUREMENT
extent and location of capillary nonperfusion, presence of
neovascularization, and management of diabetic macular Spectral domain ocular coherence tomography (OCT)
edema. The confocal, laser scanning, digital imager pro- (Heidelberg Spectralis or equivalent) is used to evaluate
vides 15 dB better signal/background (contrast and reso- macular thickness in patients with NPDR. Although the
lution), eliminates the bright flash that increases patient studies demonstrated excellent sensitivity to macular edema,
comfort, and has a faster frame rate when compared to the real issue is whether it is beneficial to perform focal laser
digital or film-based angiography systems. Better image before CSME is present on clinical biomicroscopic examina-
quality is achieved because confocal imaging increases the tion. This issue will be addressed in the next section.
171
172 SECTION IV ■ Disease-Sp ecific Managem ent

Fig u re 1 9 .2 ■ OCT im ag e of vitreom acular traction in a


diab etic eye.

Ph arm aco t h e rap y o f Diab e t ic Macular Ed e m a:


Co rt ico st e ro id s
Since the introduction of intravitreal triamcinolone for
the treatment of uveitic cystoid macular edema (6), it has
Fig ure 1 9 .1 ■ The Staurenghi 230 SLO lens perm its angiogra- become clear that steroids have a direct effect on the reti-
phy of preequatorial neovascularization. nal vascular endothelium, decreasing leakage and restoring
the blood retinal barrier. This discovery has led to the wide-
spread use of intravitreal steroids for macular edema due to
multiple causes, including diabetic retinopathy and retinal
NONPROLIFERATIVE RETINOPATHY vein occlusions. Recent reports from the Intravitreal Steroid
Injection Studies–Diabetic Macular Edema (7) (ISIS-DME)
The Early Treatment Diabetic Retinopathy Study (ETDRS) have shown that intravitreal injections of triamcinolone
has contributed greatly to our management of patients (Kenalog) result in significant visual improvement (three or
with NPDR (4,5). The ETDRS defined CSME as (a) thick- more lines) in 38% of patients. Subgroup analysis revealed
ening of the retina at or within 500 µm of the center of that 62% of patients with cystoid macular edema improved
the macula, (b) hard exudates at or within 500 µm of the at least three lines of vision, whereas only 9% of those with
center of the macula if associated with thickening of adja- noncystoid macular edema had a similar response. The
cent retina, or (c) an area of retinal thickening one disk macular edema recurred in approximately 50% of patients 6
diameter or larger, part of which is within one disk diam- months after the injection. Complications noted were steroid
eter of the center of the macula. It should be emphasized glaucoma in approximately a third of patients. Intravitreal
that the ETDRS was based on the clinical perception of steroids have other important complications in addition to
macular edema. The majority of the physicians, including steroid glaucoma. Posterior subcapsular cataract formation
the authors, do not treat pre-CSME patients. Pre-CSME is a well-recognized complication of steroids and should be
patients should probably be treated based on upcom- considered whenever injections are to be performed in pha-
ing cataract surgery, poor result in the other eye, marked kic individuals. The authors do not use steroids in the man-
peripheral capillary nonperfusion, upcoming panretinal agement of diabetic macular edema because of the untenable
photocoagulation (PRP), or an estimation that the patient steroid glaucoma and cataract rates.
was unlikely to return for follow-up visits. Most surgeons The widespread use of intravitreal injections of ste-
use the Goldmann (flat) contact lens or macular lenses roids has been followed by various drug delivery systems
and start with a 60- to 100-µm, 100-milliwatts, and 20- that promise to prolong the release of steroids with slow-
to 30-ms spot. Many surgeons now use lighter treatment release devices implanted in the vitreous. Currently, there
compared to the Diabetic Retinopathy Study (DRS) and are two types of drug delivery steroid implants in clinical tri-
ETDRS. Some surgeons primarily treat microaneurysms als: biodegradable implants (e.g., Osurdex dexamethasone-
and microangiopathy noted to leak on angiography, oth- polyacticglycolic acid) and nonbiodegradable (e.g., Retisert
ers primarily use a light grid in the thickened, leaking fluocinolone implant). Both types of implants have potential
area, and most treat both. Preferred lasers include argon advantages and disadvantages. Biodegradable implants have
green (514 nm), diode or flash tube–pumped diode lasers the advantage of requiring one implantation (that can be
(532 nm), and krypton yellow (577 nm). Infrared (IR) performed in the office with a 22-gauge injection applicator)
diode (>800 nm) lasers are not recommended because of but may have nonlinear release kinetics, whereas nonbiode-
absorption in the choroid and pain. The authors prefer gradable implants, although having linear release kinetics,
a 532-nm, diode-pumped, frequency up-converted con- require more complex surgical implantation and subsequent
tinuous wave YAG laser on the PASCAL platform. removal unless the implant is left in place. The Retisert clinical
CHAPTER 19 ■ Diabetic Retinopathy 173

trials reported a 90% incidence of steroid glaucoma and a 34% authors present their current perspective of management of
incidence of glaucoma filtering procedures. The risk/benefit diabetic macular edema, with the understanding that this
and cost/benefit analyses of these implants in comparison with protocol may change in the near future as clinical research is
repeated intravitreal injections of triamcinolone suggest that presented. Combination therapy is an appealing concept but
the implants are too costly, in addition to causing an unaccept- is appropriate in some instances and not in others. Combina-
ably high rate of steroid glaucoma and cataracts. tion chemotherapy in oncology is utilized because the agents
Because of steroid glaucoma and steroid-induced cata- have narrow windows between effective and toxic drug lev-
racts, the authors do not use intravitreal steroids for diabetic els as well as to provide multiple barriers to the evolution of
macular edema and use a combination of anti-VEGF therapy cancer cells. Combination therapy for infectious disease is
(Avastin), PASCAL laser, and topical nonsteroidal agents utilized in severe infections when the infectious agent has
(Nepafenac, Alcon). not been identified and delayed treatment would produce
bad outcomes. Combination therapy for infectious disease
creates multiple barriers for evolution of the infectious agent
Ph arm aco t h e rap y o f Diab e t ic Macular
but unfortunately leads to higher incidence of resistance.
Ed e m a: Vascular En d o t h e lial Gro wt h
Focal and/or PRP laser plus anti-VEGF therapy is very effec-
Fact o r An t ag o n ist s
tive in diabetic retinopathy and can be broadly defined as
Macular edema is caused by vascular endothelial growth fac- combination therapy. Topical nonsteroidal (Nepafenac) ther-
tor (VEGF) (8–10), the same agent that causes retinal neo- apy in combination with laser and anti-VEGF compounds
vascularization in diabetes and choroidal neovascularization is effective for diabetic macular edema because of multiple
in age-related macular degeneration. VEGF downregulates mechanisms, VEGF, and inflammation.
the tight junctions of the endothelium of the retinal ves-
sels, causing breakdown of the blood-retinal barrier, and
therefore leakage of fluid and macromolecules into the reti- PROLIFERATIVE RETINOPATHY
nal intercellular space. Ischemia leading to VEGF produc-
tion may be a factor in certain macular edema cases. For The DRS randomized, multicenter clinical trial proved that
this reason, patients with macular edema not responding to PRP is effective for patients with PDR (12–15). Many sur-
direct focal treatment of leaking microaneurysms, or areas of geons now treat before the DRS treatment criteria are reached
leakage on fluorescein angiography, may respond to treating (16), in part because of the subsequent ETDRS study data.
areas of ischemia. Heavy grid photocoagulation probably has The DRS criteria are based on high-risk characteristics as
little effect in reducing neovascularization but significantly compared to standard photographs. The authors use light
reduces central visual fields and therefore reading speeds treatment with small spot size (100 to 200 µm), using 20- to
and often causes patients with excellent Snellen acuity to 30-ms duration to reduce thermal diffusion, with the spots
complain that they “cannot see.” placed one spot size apart. Many surgeons treat using a very
The introduction of anti-VEGF therapy has led to a new large number of intense, large, 100-ms duration spots and
mode of therapy for macular edema secondary to NPDR as see the patient in follow-up in 3 months. Some of this behav-
well as for venous occlusive disorders. Bevacizumab (Avas- ior is unfortunately driven by the so-called global period for
tin) is an anti-VEGF antibody currently approved by the FDA Medicare reimbursement. It is probably better practice to use
for systemic therapy of cancer. Intravitreal Avastin is widely a moderate number of spots and reexamine the patients in
used for the treatment of choroidal neovascularization in age- 1 month. Some surgeons have a protocol mindset with respect
related macular degeneration. Avastin is also being used for to PRP treatment. This approach may result in inappropriate
macular edema secondary to Branch Retinal Vein Occlusion vitrectomy or peripheral cryo when the “protocol has been
(BRVOs) (11) and Central Retinal Vein Occlusion (CRVOs) completed” and active neovascularization is present.
as well as for retinal neovascularization in proliferative dia- It is better to perform PRP fill-in, combined with anti-
betic retinopathy (PDR). The initial published results as well VEGF therapy, if there is any neovascularization and the
as the authors’ results demonstrate that anti-VEGF therapy patient is greater than 1-month posttreatment. The authors
combined with laser should become standard therapy in the believe that this combined, incremental approach to therapy
management of diabetic macular edema and PDR. reduces treatment-related complications and discomfort. If
the patients present with florid neovascularization, it is better
to use anti-VEGF therapy at the initial visit combined with
Man ag e m e n t o f Diab e t ic Macular Ed e m a
PASCAL PRP. PRP with PASCAL using 30-ms, less intense
The recent advances in pharmacotherapy and surgical ther- burns does not destroy ischemic inner retina raising ques-
apy for diabetic macular edema, added to the well-known tions about efficacy, but studies have shown identical out-
and time-proven approaches with laser photocoagulation, comes with less pain. The principal author points out that
provide the clinician multiple therapeutic possibilities. the destruction of RPE and photoreceptors results in trans-
Although there are no trials that clearly indicate which com- synaptic degeneration of the bipolar, horizontal, amacrine,
bination and sequence of therapies should be employed, the and ultimately ganglion cells similar to what occurs in
174 SECTION IV ■ Disease-Sp ecific Managem ent

retinitis pigmentosa. Combining PASCAL PRP with Avastin at 1-month intervals until the blood clears or surgery is
combines the benefit of rapid response to Avastin with performed. Ultrasonic evidence of posterior pole detach-
the durable response of PRP without the disadvantage of ment requires immediate vitrectomy. The usual question of
complications secondary to longer duration, more intense duration of a vitreous hemorrhage plays a less important role
burns. in the surgical decision-making process than other factors. If
The authors virtually never use a retrobulbar block it does not appear that near-term clearing will occur, bilat-
for laser treatment. Red and IR lasers cause more pain than eral hemorrhage requires surgery on the eye with the best
532-nm lasers. Durations greater than 30 ms cause more visual prognosis. Vitreous hemorrhage in a patient with only
pain because of thermal diffusion; a duration of 30 ms on one eye as well as the better eye of bilateral cases should be
PASCAL is a better choice for PRP. Larger spot sizes and operated on to improve visual function. Those patients with
higher powers cause more pain, light scattering, and poten- shortened lifespan and multisystem disease need immediate
tially more nuclear sclerosis and occult macular photic visual rehabilitation for emotional and social reasons. Sub-
damage. posterior vitreous detachment and preretinal hemorrhages
clear more rapidly than does hemorrhage in the vitreous cor-
tex. For this reason, patients with bilateral or only-eye sub-
VITRECTOMY PVD or preretinal hemorrhage can be followed up for as long
as the patient’s emotional and social needs permit. If one
Better medical management and laser photocoagulation eye has macular ischemia and the other, better, eye devel-
should significantly reduce the need for vitrectomy for the ops a vitreous hemorrhage, vitrectomy may be indicated to
complications of PDR. Vitrectomy, when indicated, has an improve the patient’s overall visual function.
excellent prognosis when appropriate patient selection and
techniques are utilized (17).
Tract io n Re t in al De t ach m e n t
TRD can be diagnosed by ophthalmoscopic or ultrasonic
CASE SELECTION examination. If the macula is detached, vitreous surgery
should be performed within 3 weeks, unless there are medi-
It is useful to divide blindness from diabetic retinopathy cal contraindications. If there is active neovascularization, it
into two subgroups: those cases requiring immediate sur- is better to perform PRP before vitrectomy if possible. Because
gery and those in which surgery is elective. Traction retinal of extensive exudation and fibrous proliferation, panretinal
detachment (TRD) involving the macula (MTRD), neovas- cryopexy should not be utilized. If vitrectomy indications
cular glaucoma (NVG), and anterior vitreous cortex fibro- are present, endo-PRP can be combined with vitrectomy. If
vascular proliferation (AVCFVP) are permanently blinding vitrectomy is postponed until PRP-induced or spontaneous
if left untreated. By contrast, the visual potential probably involution of neovascularization occurs, the incidence of
does not change in the treatment of vitreous or preretinal postoperative NVG and AVCFVP is dramatically reduced.
hemorrhage if there is substantial delay before surgery is Recent studies demonstrate that intravitreal anti-VEGF
performed. therapy with bevacizumab can precipitate TRDs in patients
with severe neovascularization (18). These patients should
be followed closely after anti-VEGF therapy and the surgeon
Vit re o us He m o rrh ag e
should be ready to proceed to vitrectomy if progression to
Early experimental work incorrectly concluded that vitreous TRD is evidenced.
hemorrhage caused neovascularization via organization of Because of the relatively high rate of biologic complica-
the blood clot. Vitreous hemorrhage is a result of neovas- tions and medical risk factors, vitrectomy is not indicated in
cularization rather than the cause. Although long-standing extramacular TRD. This is true even if progression toward
vitreous hemorrhage can deposit iron on many intraocular the macula or a similar condition in the other eye seems to
structures, there is usually no retinal damage from a vitre- “threaten” the macula. It is safer to operate on actual, rather
ous hemorrhage. Retinal detachment, macular damage, isch- than predicted, visual loss. The rate of progression of extra-
emia, and optic nerve function will determine the ultimate macular TRD to include the macula is about 15% per year
visual outcome when long-standing vitreous hemorrhages (19,19a,20). After several years, progression to MTRD stabi-
are removed, not the hemorrhage per se. lizes at a cumulative rate of about 30%, and there are many
If the other eye has good vision, a unilateral hemorrhage patients with 5 to 10 years duration extramacular TRDs sur-
can be followed indefinitely with ultrasound, unless retinal rounding the macula with good vision that never required
detachment, anterior vitreous cortex (AVC) neovasculariza- surgery.
tion, or iris neovascularization occurs. An eye without prior Cataract surgery can result in anterior movement of the
PRP is at greater risk for the development of neovascular vitreous with progression of extramacular TRD to macular
complications and must be watched more closely. B-scan involvement. Once again, vitreous surgery should only be
ultrasonography should be repeated at each visit, preferably performed if the macula actually becomes elevated (21).
CHAPTER 19 ■ Diabetic Retinopathy 175

MACULAR EDEMA, CYSTS, SURGICAL SEQUENCE


AND SUBMACULAR EXUDATES AND TECHNIQUES

Hilel Lewis et al. first reported vitrectomy with peeling of Vitreous surgery for hemorrhage or TRD requires a planned
taut posterior vitreous cortex (PVC) from diffuse macula sequence of surgical steps, with multiple branches depend-
edema (DME) eyes not responsive to focal laser photocoagu- ing on different intraoperative scenarios. As in all vitreous
lation. SD OCT is very effective at determining the presence surgery, a full complement of sterile tools and materials must
of vitreoretinal traction and measuring macular thickness. be immediately available.
Many surgeons now believe that vitrectomy in DME cases
without vitreomacular traction can reduce macular edema
similar to the observations in vitrectomy without branch An e st h e sia
vein decompression for BRVO and vitrectomy without radial The frequency of cardiovascular and renal disease in the dia-
optic neurotomy in CRVO. The principal author has postu- betic patient requires careful preoperative evaluation by the
lated that vitrectomy or separation of the PVC could allow primary care physician or internist and utilization of cardi-
greater egress of VEGF, known to be causative of macular ologists, endocrinologists, and other consultants as needed.
edema. Steffanson and later Holekamp have shown that vit- The anesthesiologist should review the preoperative medi-
rectomy produces higher oxygenation in the vitreous cavity cal evaluation. Diabetic vitrectomies can be performed in the
(22), which may improve macular edema as well. Peeling of ambulatory surgery center setting if systemic disease is stable,
the internal limiting membrane can improve diabetic mac- an anesthesiologist is available and the patients systemic spe-
ular edema and should be considered in patients who are cialists (nephrologists, cardiologists) give adequate consent
resistant to laser and pharmacologic treatment (23). Some for surgery. Patients that are not controlled systemically and
surgeons have reported removal of submacular exudates or require vitrectomy surgery that cannot be delayed should
draining macular cysts, but there is scant evidence of efficacy be operated in a hospital outpatient setting. It is essential
for either procedure at this time and some concern about to have MD anesthesia function in an immediate availability
retinal damage caused by the procedures. and supervisory role if CRNA anesthesia is utilized. Proximity
to the hospital ensures availability of cardiologists, endocri-
nologists, pulmonary specialists, and intensive care facilities
CONTRAINDICATIONS as well as providing higher facility fees than free-standing
ambulatory surgery centers, thereby facilitating access to best
The absence of light perception indicates glaucomatous technology. An intravenous line, EKG, pulse oximetry, blood
optic atrophy, ischemic optic neuropathy, or extensive retinal pressure monitoring, and oxygen mask with suction hose to
vascular occlusive disease and contraindicates vitreous sur- prevent hypercapnia must be utilized in all cases. The anes-
gery. Corneal opacity; corneal, lid, or conjunctival infection; thesiologist or nurse anesthetist should make liberal use of
and the inability to withstand local anesthesia are relative intraoperative serum glucose monitoring. Operating times
contraindications. are always less than 1 hour and usually about 30 minutes,
Iris neovascularization can be an indication for surgery which is compatible with local anesthesia and the associated
in diabetic TRDs (24–26). Intravitreal bevacizumab has been reduction in nausea, vomiting, and medical complications
shown by the authors to cause regression of anterior seg- of general anesthesia. Minimal, if any, sedation is used after
ment neovascularization and revert the progression toward the block, which is performed with a 27-gauge, 1.25-inch
NVG (27). Anti-VEGF therapy for NVG requires permanent needle at the outer “corner” of the orbit.
control of VEGF production through retinal reattachment
and PRP. Vitrectomy in an aphakic eye with active iris neo- In cisio n s
vascularization will result in rapid progression of postop-
erative NVG unless intravitreal bevacizumab and PRP are The authors use 25-gauge techniques for all cases, although
performed preoperatively and intraoperatively. 23-gauge methods have similar advantages and results.
Cases of several years’ duration may exhibit extensive
white vessels and retinal atrophy. If the temporal arcades are
Man ag e m e n t o f t h e Le n s
not perfusing the macula, there is no need for vitreous sur-
gery because visual improvement will not occur. If the retina Lens removal correlates with an increased incidence of post-
is extremely atrophic but PRP has not previously been per- operative NVG but prevents AVCFVP (28–30). The AVC and
formed, this is an indication that limited visual improvement lens apparently act as barriers to the anterior diffusion of
will occur from reattachment. There are, however, cases of 3 VEGF, and therefore their presence reduces the incidence of
to 4 years’ duration that have improved to ambulatory vision NVG. A specific attempt should be made to leave the AVC, if
levels after vitrectomy. Late cases with atrophic retina produc- the lens is retained during vitrectomy in an effort to reduce
ing little VEGF have a lesser incidence of NVG and AVCFVP. the development of posterior subcapsular cataract.
176 SECTION IV ■ Disease-Sp ecific Managem ent

Contact lenses for aphakic correction should be uti- acrylic sulcus lens can be implanted anterior to the anterior
lized judiciously because of decreased corneal sensitivity capsule. An anterior chamber lens can be implanted if there
and infection risk but are effective in a surprising number of is minimal likelihood of bleeding and the capsule is not suit-
patients. Spectacle correction is surprisingly well tolerated able for a posterior chamber lens.
in these patients. Implantation of posterior chamber lenses
after endocapsular lensectomy (ECL) or phacoemulsifica- Vit re ct o m y
tion tends to keep the AVC and posterior capsule intact, may
decrease NVG, offers the best optical outcome, and can be The continuity of the PVC is a critical concept in the
used in all but the most complicated cases. understanding and planning for the vitrectomy process.
ECL with a 20-gauge ultrasonic fragmenter is preferable The PVC will be completely adherent to the retina, par-
to phacoemulsification combined with vitrectomy if there is tially detached, or completely detached from the retina. The
significant fibrin syndrome or neovascularization. Preopera- “core” vitrectomy misconception stems from earlier days
tive anti-VEGF therapy as well as injection of anti-VEGF when high suction and slow cutting pulled vitreous into the
agents at the end of the case reduce the complications and central portion of the eye. Complete sectioning or truncation
enable more frequent use of phaco-vit. Phaco combined with of the PVC (Fig. 19.3), rather than “band cutting” or “core
vitrectomy reduces the number of procedures. If the phaco vitrectomy,” must be understood before surgical success can
is performed at the beginning of the procedure, pupillary be obtained. These concepts apply whether the vitreous is
dilation and corneal clarity may suffer, thereby reducing sur- opaque, semiopaque, or clear.
gical visualization. If phaco and intraocular lens (IOL) inser-
tion are performed after the vitrectomy, unavoidable periods Pro ce d ure if Po st e rio r Vit re o us
of hypotony will increase bleeding. If significant cataract is De t ach m e n t Is Pre se n t
noted at the preoperative evaluation, best practice is usually
to send the patient to a cataract surgeon for phaco and IOL. If the PVC is detached from the retina, a central opening in
If the preoperative view is judged to be adequate to per- the vitreous should be created and linear extrusion with the
form vitrectomy, phaco and IOL insertion can be performed vitreous cutter or a 25-gauge, soft-tip cannula performed if
approximately 1 month after vitrectomy. If the preoperative any subvitreous erythroclastic or hemolytic material is pres-
view was judged to be adequate but it was determined dur- ent. This step is also known as vacuum cleaning or extru-
ing vitrectomy that the view was inadequate, endocapsular sion. When a clear effluent is obtained from this fenestration,
fragmenter lensectomy with retention of the anterior cap- the opening should be enlarged with the cutter until only a
sule is currently the author’s preference. If sufficient capsule small “skirt” at the confluence of the AVC and PVC remains.
was retained and bleeding is unlikely, a foldable, multipiece Particular care must be taken to trim the superior “skirt”

Fig u re 1 9 .3 ■ Trun cation o f


PVC relieves so-called anterop os-
terior traction on diabetic traction
detachm ents.
CHAPTER 19 ■ Diabetic Retinopathy 177

Fig u re 1 9 .4 ■ Lin ear suction


with a straight 25-gauge, soft-tip
cannula should be used to rem ove
all p reretin al b lo od p rod u cts,
p erm it b etter visualization an d
less p ostop erative erythroclastic
g laucom a, and enab le p hotoco-
ag ulation without d am ag ing the
retina.

if opaque, because it can hang down postoperatively and absence of this information, the first opening should be
obscure the seated patient’s view. Linear suction with a straight made nasally in the midperiphery to avoid the macula and
25-gauge, soft-tip cannula should be performed to remove in an area easily treatable if a retinal break occurs. After
all preretinal blood products, permit better visualization an opening is made, extrusion (vacuum cleaning) through
and less postoperative erythroclastic glaucoma, and enable the opening must be continued until a clear effluent is
photocoagulation without damaging the retina (Fig. 19.4). If obtained. It is at this time that the novice vitreous surgeon
a complete posterior vitreous detachment (PVD) is present, could mistake voluminous old blood trapped behind the
there is no perpendicular or oblique force on the retina, but vitreous for active bleeding. When the retina is visualized
tangential force from epiretinal membranes (ERMs) can cause through the opening, a safe circumferential truncation of
TRD. Vascular attachment points of the ERM to the retina the PVC can proceed from this point and extend for 360
should be treated with the bipolar diathermy or endophoto- degrees. It is not necessary to make multiple openings in
coagulator only if they bleed intraoperatively or appear active. the PVC or to dissect the layers of the PVC in “onion skin”
The Alcon 20-gauge disposable bipolar endoilluminator and fashion. The “skirt” must be trimmed as described above
25-gauge bipolar diathermy are ideal for this technique. and the portion connected to the retina trimmed down
to near the retinal surface. Any areas of PVC connected
to two or more retinal points should be sectioned with
Pro ce d ure if Part ial Po st e rio r Vit re o us
the vitrectomy instrument, if there is sufficient clearance
De t ach m e n t Is Pre se n t
for the tip, or with the 25-gauge, curved segmentation/
If only a partial PVD has occurred, the vitreous will be delamination scissors. If these bridging areas of PVC are
adherent to the retina at one or more epicenters. Typically, vascularized, they can be precoagulated with the dispos-
the optic nerve and vessels serve as attachment points able bipolar cautery.
because of glial proliferation. As the vitreous contracts,
these attachment points become the apex (apices) of the
Pro ce d ure if No Po st e rio r Vit re o us
now-conical PVC. This is the most common vitreous
De t ach m e n t Is Pre se n t
configuration encountered in PDR. It is critical in these
patients to section or truncate the PVC to completely In some diabetics, the entire PVC is adherent to the retina. In
eliminate any anteroposterior traction (Fig. 19.5). Vitre- the unlikely event that the PVC is minimally adherent, mem-
ous bands do not exist as such but are actually more vis- brane peeling can be performed. Caution must be exercised
ible portions of the PVC continuum. The PVC penetration in attempting to peel the PVC in a single sheet because reti-
is initiated in an area indicated by preoperative ultrasound nal breaks can occur in sites remote from the forceps grasp
or indirect ophthalmoscopy to have attached retina. In the site. Areas of photocoagulation can cause adherence of the
178 SECTION IV ■ Disease-Sp ecific Managem ent

Fig u re 1 9 .5 ■ Im b rication of
taut PVC into the port of the cut-
ter can cause posterior or periph-
eral retinal b reaks. It is b etter to
sever the PVC with scissors.

PVC to the retina as well as the choroid and sclera. If strongly latter method, developed by the principal author, was later
adherent zones are present, the PVC should be allowed termed “en-bloc” when described by others using a less safe,
to remain in these locations and the scissors or vitreous outside-in direction of dissection. Even worse, the advocates
cutter used to sever all tangential traction (Fig. 19.6). PVC of en-bloc dissection, as originally described, suggest that
truncation must be completed in every case but can be per- intentional traction on the vitreous can facilitate dissection.
formed with scissors in the case of shallow PVDs or following Using the vitreous to lift the ERM has substantial risk of
inside-out delamination if a TRD is present (Fig. 19.7). This causing peripheral retinal breaks from counter traction.

Fig u re 1 9 .6 ■ In sid e-out d el-


am in atio n o f ERM co n tig uo us
with PVC.
CHAPTER 19 ■ Diabetic Retinopathy 179

Fig ure 1 9 .7 ■ Rem oval of PVC


and ERM after inside-out delam i-
nation (en bloc).

DIABETIC TRACTION RETINAL Occasionally the PVC is so taut that it is better to truncate
DETACHMENT some regions with scissors rather than the vitreous cutter.
Smaller cutters (25 or 23 gauge) usually obviate the need
Understanding surgical anatomy is crucial to a success- for this step. Wide-angle illumination systems such as chan-
ful surgical approach; the central element is the continuity deliers and Tornambe Torpedos reduce the ability of the
of the PVC and the role of vitreoretinal (so-called AP trac- surgeon to visualize transparent PVC, necessitating the use
tion) and tangential, ERM-driven traction. Three often used of triamcinolone particulate marking. Similarly, wide-angle
phrases indicate a poor grasp of surgical anatomy and surgi- visualization systems (BIOM, EIBOS, Volk, AVI) reduce lat-
cal principles: (a) core vitrectomy, (b) resection of traction eral and axial resolution and, just as chandeliers and Torpe-
“bands,” and (c) “membrane peeling or stripping.” Let us does, hinder visualization of the transparent PVC.
start with the incorrect concepts of core vitrectomy and trac- Truncation of the PVC should be followed by manage-
tion bands. There is no “core” vitreous and there are no trac- ment of ERMs unless no PVD is present. If a PVD has not
tion bands; the entire transparent PVC is the cause of TRD, occurred, inside-out delamination of ERM starting at or near
not a region of vitreous rendered opaque by chronic vitreous the disk extending out along the arcades should be initi-
hemorrhage or a so-called traction band. It follows that the ated rather than vigorously and dangerously trying to create
first and most important surgical goal is 360-degree trun- a PVD. “En bloc dissection,” as initially described, utilized
cation of the conical (or truncated cone if table-top TRD) outside-in technique and advocated using the PVC to lift the
PVC midway between the outer margin of ERM attachment ERM. Putting traction on the PVC results in peripheral reti-
areas and the periphery. Secondly, the incorrect notion of nal breaks while the surgeon is focusing on the ERM dissec-
membrane peeling (stripping) must be addressed. ERMs in tion. En bloc is a poor term when applied to ERM dissection;
the vast majority of diabetic TRD cases are highly adherent; it is only meaningful in cancer surgery when the notion is to
attempts to peel or strip the membranes using pics and simi- minimize the dispersion of cancer cells. A far better way to
lar tools usually result in iatrogenic retinal breaks. The prin- think about this issue is to forget the outdated notion that
cipal author developed scissors segmentation of ERMs and the standard surgical algorithm is first performing core vit-
subsequently scissors delamination of ERMs to address the rectomy, then creating a PVD, and finally peeling ERMs. If a
strong adherence problem. PVD is not present in a diabetic TRD case, it is better to go
Truncation of the PVC should be performed by using directly to inside-out delamination starting at or near the
the highest possible cutting rates and lowest vacuum while optic nerve head. It is not necessary to remove all ERM in
applying the port to the surface of the PVC. It is dangerous one piece as suggested by the term en bloc; it is preferable to
and inefficient to attempt to do core vitrectomy and pull the use access segmentation, cutting ERM into several segments,
vitreous to the port using excessive vacuum and flow rates. each of which will be contiguous with the PVC. When most
180 SECTION IV ■ Disease-Sp ecific Managem ent

Fig ure 1 9 .8 ■ Access segmenta-


tion develops a plane for inside-out
delam ination. Forceps m em brane
p eeling , scissors seg m entation,
and scissors delamination are usu-
ally best performed in an inside-out
direction because central retina
is stronger than m ore peripheral
retina, the retina is redundant cen-
trally, and edges are hard to find,
creating the risk of iatrogenic retinal
breaks.

or all the ERM has been delaminated using an inside-out and delamination means to cut it off. Initially, delamination
direction, PVD creation will be accomplished as well or it was performed with so-called horizontal scissors, actually
will be irrelevant because there are no longer any posterior 135 degrees. For almost two decades, the principal author used
attachment points. only curved scissors for what is termed “conformal delamina-
tion.” The 25-gauge Alcon DSP curved scissors have been used
for all cases since they became available (Fig. 19.9). Curved
Scisso rs Se g m e n t at io n an d De lam in at io n
scissors are preferred to “horizontal” scissors because the
Scissors segmentation of ERMs is used to reduce tangential curvature of the scissors matches the curvature of the retina,
force on the retina by separating ERM into so-called epicen- reducing the likelihood of impaling the retina with scissors tips.
ters, better termed “vascularized adherence sites.” Segmenta- The same design concept is used for skis, the tips curved up.
tion is performed by placing one blade of the scissors under If the scissors are introduced under the ERM with the blades
the ERM, between the retina and the ERM, and the other blade wide open and then closed, attachment points will be ripped
anterior to the ERM. Although this was initially performed with
so-called vertical scissors, curved scissors are far superior for
segmentation because blade width is much greater than blade
thickness; “vertical” scissors require more space between the
retina and the ERM than curved scissors. The principal author
largely abandoned segmentation after developing delamina-
tion; segmentation is now primarily used for access segmen-
tation to expose the delamination plane (Fig. 19.8). Using
curved scissors expedites transitioning from access segmenta-
tion to delamination without tool exchange.
The senior author developed scissors delamination
2 years after developing segmentation to address the issue of
residual ERM after segmentation. Residual ERMs in diabetic
TRD cases resulted in small areas of chronic retinal elevation,
which ultimately caused atrophic retinal holes and late rheg-
matogenous retinal detachments. Scissors delamination is per-
formed by inserting both scissors blades in the potential space
between the ERM and the retina and severing the attachment Fig ure 1 9 .9 ■ Curved scissors are the best tool for both seg-
points. Simplistically, segmentation means to cut up the ERM m entation and inside-out conform al delam ination.
CHAPTER 19 ■ Diabetic Retinopathy 181

Fig ure 1 9 .1 0 ■ Conform al cut-


ter delam ination is p erform ed by
rotating the cutter around the lon-
gitudinal axis to control the angle
of attack; ERM is fed into a p ort
that is p ositioned ap p roxim ately
9 0 d eg rees fro m th e tan g en t
plane to local retinal contour.

instead of sheared, which causes retinal breaks. If the scissors behind the leading edge, allowing vacuum to cause the ERM
are introduced under the ERM with the blades closed and then to fold back into the port (Fig. 19.11). With this technique,
opened similar to the technique used for blunt dissection in the ERM protects the retina. Smaller cutters, 23 gauge or
general surgery, attachment points will be ripped instead of preferably 25 gauge, are ideal for these techniques as well as
sheared, creating retinal breaks. The optimal technique is to segmentation of PVC between vitreoretinal adherence sites
open the scissors just enough to engage an attachment point, because the port is close to the tip; the tip has smaller diam-
cut, pull back slightly, move laterally to the next attachment eter, reducing axial and lateral setback from the target ERM.
point, advance slightly, and cut again. It is not necessary to lift
or stabilize the ERM if disposable scissors or reasonably new
reusable scissors are used. Because ERM is elastic and there is a In sid e -Out Ve rsus Out sid e -In Te ch n iq ue
pressure gradient across the retina, the ERM will scroll up and
Machemer used outside-in technique for bent needle mem-
separate from the retina without lifting.
brane peeling, and many surgeons unfortunately emulate this
technique when using delamination. A far better approach
Cut t e r De lam in at io n o f Ep ire t in al Me m b ran e s for end-grasping forceps membrane peeling, scissors seg-
mentation, and scissors delamination is to initiate the dis-
Vitreous cutters are increasingly being used for removal
section centrally and peel, segment, or delaminate outward.
and segmentation of ERMs in diabetic TRD cases, but they
Outside-in dissection is safer because the retina is thicker
cannot replace scissors delamination for all cases. There
and 100× stronger centrally than in the periphery, especially
are three distinct techniques for using scissors in diabetic
in ischemic and post-PRP patients, the retina is redundant
TRD surgery: (a) conformal cutter delamination, (b) fold-
centrally in TRD cases, and the view is better centrally.
back delamination, and (c) segmentation of PVC segments
between adherence sites. Conformal cutter delamination is
performed by placing the port at the outer margin of the
Visco d isse ct io n
ERM and feeding, not sucking or pushing the ERM into
the port (Fig. 19.10). It is conformal because the angle of Viscodissection is performed by injecting a viscoelastic in the
attack is continuously adjusted so that the port is rotated potential space between the ERM and the retina. Viscodissec-
away from the retinal surface as the cutter is moved along tion is seldom performed today because it has not been shown
the concavities and convexities of the TRD. This technique is to be advantageous and it increases cost and operating time.
used for relatively rigid, thick ERMs not candidates for fold- Viscodissection pressurizes the potential space between the
back delamination. Foldback delamination involves plac- ERM and the retina, which can result in a hydraulic retinal
ing the cutter port on the anterior surface of the ERM just break if the ERM is tightly adherent. If the ERM is very
182 SECTION IV ■ Disease-Sp ecific Managem ent

Fig u re 1 9 .1 1 ■ Fold b ack d el-


am in ation involves p lacin g th e
cutter p ort on the anterior sur-
face of the ERM just b ehind the
leading edge allowing vacuum to
cause the ERM to fold b ack into
the port.

adherent with no gap between the ERM and the retina, the He m o st asis
visco cannula may create a retinal break during introduction.
Endolaser is better than diathermy for treatment of the
Viscoelastic substances significantly decrease the interfacial
severed vessel ends of vascular attachment points after
tension of silicone oil, increasing the likelihood of emulsifi-
delaminating ERMs in diabetic TRD surgery (Fig. 19.12).
cation. It is virtually impossible to remove the viscoelastic;
The authors believe that using diathermy on retinal sur-
the viscous material potentially increases glial recurrence
face bleeders can result in subclinical retinal necrosis and
rates by retaining cells, fibronectin, and cytokines such as
late atrophic holes. On occasion, larger vessels may require
VEGF, basic fibroblast growth factor, and TGF-β.

Fig ure 1 9 .1 2 ■ Focal 25-g auge


laser is id eal for coag ulating vas-
cular attachm ents after delam ina-
tion because of sm all spot size.
CHAPTER 19 ■ Diabetic Retinopathy 183

diathermy before or after transsection, especially when the oil is confined behind an IOL. The emulsification and
performing retinectomy. silicone oil glaucoma rate is less than 5%.

An cillary Ep ire t in al Me m b ran e Man ag e m e n t


Pan re t in al Ph o t o co ag ulat io n
Te ch n iq ue s
PRP should be performed on all attached areas when operat-
Multifunction tools such as the disposable, 20-gauge bipolar
ing diabetic TRD cases. However, PRP should not be per-
endoilluminator and the Chang end-aspirating laser probe
formed on areas that were detached preoperatively because
were developed because of the need of more than two func-
the ever-present thin layer of subretinal fluid (SRF) after
tions and the limitation of having only two hands. Illuminated
fluid-air exchange and internal drainage of SRF coupled with
instruments have been developed largely to enable so-called
retinal edema results in overtreatment and fibrin syndrome.
bimanual surgery, which is better termed “forceps stabiliza-
tion of ERMs.” The general concept is that scissors produce
An t i–Vascular En d o t h e lial Gro wt h a push-out force and pics produce stress, which can tear
Fact o r Th e rap y the retina during the dissection of ERM. The purpose of the
forceps is to offset these unwanted forces. Bimanual technique
Preoperative bevacizumab (off-label in the United States,
is often incorrectly used to lift the ERM, which often results in
Avastin) rapidly causes regression of neovascularization in vir-
retinal tears. The author seldom uses bimanual surgery.
tually all cases, dramatically reducing intraoperative bleeding.
Viscodissection can create retinal breaks by pressurizing
Caution is required, however, because rapid cicatrization can
the potential space between the ERM and the retina and then
occur, resulting in increased TRD. This effect can be avoided
hydraulically rupturing the 100× weaker retina. Viscodissection
in almost all instances by operating between 4 and 7 days after
has not been shown to improve outcomes, and viscoelastics
injection. As Avery has pointed out, it is crucial to reinject
can cause retention of cytokines, cells, fibrin, and blood at the
Avastin at the end of the case to avoid rebound. The principal
retinal surface, which may lead to glial reproliferation. The
author recommends injecting Avastin at the end of all diabetic
authors do not recommend using viscodissection. As men-
vitrectomies recalling that the anti-VEGF effect of PRP may
tioned previously, all of authors’ complicated diabetic TRDs are
take longer than Avastin to take effect and PRP and Avastin are
operated using 25-gauge techniques, and multifunction tools
synergistic. Endo-PRP decreases NVG and AVCFVP in high-
are currently not available for 25-gauge instruments. Bimanual
risk cases but is probably not required in cases with com-
techniques require chandelier illumination, which is usually
pletely regressed neovascularization. TRD cases are at higher
unnecessary for successful repair of the most severe TRDs.
risk for neovascularization than are hemorrhage cases, but
unfortunately endophotocoagulation cannot be safely utilized
Re t in e ct o m y
on elevated retina. PRP should not be performed on formerly
detached retina because a thin layer of SRF always remains, If extremely dense ERM is firmly adherent to atrophic, ele-
the retina is often edematous, and overtreatment is inevitable. vated retina, it is often advisable to perform retinectomy,
Overtreatment results in fibrin syndrome. removing membrane and retina together. Retinectomy is
often best done under air with the cutter also being used for
fluid-air exchange and internal drainage of SRF. If the retine-
Silico n e Oil
ctomy is small, it can be surrounded by endolaser. If large or
Silicone oil should be used for rhegmatogenous confinement multiple retinectomy areas are present, air-silicone exchange
usually without retinopexy when retinectomy is required is a better choice than gas. If silicone oil is used, retinopexy
for areas of highly adherent ERMs affixed to atrophic retina. avoidance is practiced, as this reduces tissue damage and
This is more often the case with long-standing extramacular glial reproliferation. Laser can be performed in a fractional
TRDs that finally developed a retinal break and progressed to dose staged manner in the postoperative course if removal
include the macula necessitating surgery. Midperipheral and of the oil is anticipated. In many, if not most, cases, the best
nasal areas are more likely to require retinectomy. The authors practice is to allow the oil to remain in place indefinitely.
often utilize retinectomy without retinopexy when reoperat-
ing cases initially operated by other surgeons who created
a large number of retinal breaks attempting to peel highly SURFACE TENSION MANAGEMENT
adherent membranes. Long-term surface tension management
is a crucial tool in these difficult cases. Excessive retinopexy Fluid-gas exchange should only be utilized if there are atro-
causes glial recurrences and fibrin syndrome; permanent sili- phic or iatrogenic retinal breaks. The author prefers SF6 to
cone oil eliminates the need for retinopexy, hence the terms C3F8 because fibrin may form around long-term bubbles in
“rhegmatogenous confinement” and “retinopexy avoidance.” diabetic TRD patients. If retinal breaks are noted at surgery, all
There is an undue emphasis on silicone oil removal, which ERM should be delaminated followed by simultaneous fluid-
often results in redetachment. Silicone oil complications are air exchange and internal drainage of SRF. The Alcon 25-gauge
minimal and incidentally equal for 1,000 and 5,000 cs when J-wand is an ideal tool for simultaneous internal drainage of
184 SECTION IV ■ Disease-Sp ecific Managem ent

SRF, fluid-air exchange, and removal of preretinal blood. The in 1976 in the hope that it could be used as a vitreous
25-gauge silicone soft-tip cannula also allows drainage of SRF substitute in the diabetic patient. It was evaluated because of
with minimum trauma to the intraocular tissues. Portions of high oxygen capacity. Unfortunately, it was discovered that
the retina that were attached intraoperatively should not be it caused severe damage to the inferior cornea when left in
treated with PRP because this increases the likelihood of fibrin place for months.
syndrome due to overtreatment resulting from residual SRF
and retinal edema. Intraocular air (gas) is only utilized if a
retinal break is suspected or seen. The only role of air (gas) VISUALIZATION
is to restore the transretinal pressure gradient via surface ten-
sion. Air (gas) surface tension management has no role in the Wide-angle visualization is ideal for rhegmatogenous reti-
treatment of TRD without retinal breaks (no tear, no air). nal detachments, proliferative vitreoretinopathy, and giant
retinal breaks but disadvantageous for most diabetic TRDs.
Flat irrigating contact lenses (Machemer) provide much bet-
SILICONE OIL SURFACE TENSION ter axial (depth) resolution and lateral resolution than wide-
MANAGEMENT angle optics and are preferred for most diabetic vitrectomies.
Wide-angle illuminations such as the chandelier and Torpedo
Silicone should be utilized when large or multiple retinal do not permit focal-, specular-, and retro-illumination tech-
breaks or retinectomies are present to eliminate the need for niques, which are essential to visualize transparent vitreous.
retinopexy and reduce reproliferation, since it allows long-
term (or indefinite) surface tension management of retinal
breaks. Internal fluid-air exchange with the console air INFUSION FLUID
source and internal drainage of SRF with linear extrusion
should precede air-silicone exchange. Before the availability of intraoperative serum glucose moni-
Silicone oil acts as a barrier to the anterior diffusion of toring, best practice was to use an intravenous infusion of
VEGF and markedly reduces anterior segment neovascular- dextrose 5% in water to prevent unrecognized hypoglyce-
ization if no inferior iridectomy is present. Because silicone mia. Serum glucose levels were often in the 300 to 500 mg%
recompartmentalizes the eye, fibrovascular proliferation can range, necessitating adding dextrose to the infusion fluid.
occur at the retina-silicone interface (Fig. 19.13). For this The author has not added dextrose or anything else to the
reason, extensive PRP is our best hope for these cases. Sili- infusion fluid since BSS Plus (glutathione bicarbonate Ring-
cone may prevent oxygen diffusion from well-perfused to er’s solution) became available and never observes intraop-
ischemic retinal areas, thus causing visual loss. The princi- erative or early postoperative posterior subcapsular cataract.
pal author used perfluorocarbon liquids in an animal study Smaller fluid volumes associate with three-port systems,

Fig u re 1 9 . 1 3 ■ Silico n e o il
and g as cause recom p artm ental-
ization, wh ich seq uesters VEGF,
fibrin, cells, and cytokines at the
retinal-bubble interface.
CHAPTER 19 ■ Diabetic Retinopathy 185

better fluidics, and better techniques, and now 25-gauge vessels, or sclerotomies. Approximately 50% of phakic cases
fluidics have improved the situation as well. develop immediate postoperative vitreous hemorrhages
(44). In aphakic cases, this hemorrhage will typically clear in
1 to 2 weeks, but the phakic cases can take several months.
RETINOPEXY If ultrasound indicates that the retina is attached, and there
is excellent vision in the other eye, no reoperation is neces-
All breaks require retinopexy unless they are macular, papil- sary. If ultrasound shows the retina to be detached, immedi-
lomacular bundle, or peripapillary in location, in spite of ate reoperation is indicated. If the patient is bilaterally blind,
evidence that an occasional untreated break will not result in emotional and social needs dictate the need for reoperation.
detachment. The laser endophotocoagulator is used to treat It is advisable to operate on the eye with the highest visual
all breaks unless they are very extensive, indicating the need potential whether it is a first operation or reoperation.
for long-term silicone oil for rhegmatogenous confinement. A full vitrectomy setup with standard three-port
25-gauge vitrectomy is preferable to washouts in most
instances. In this way, ERM, persistent vitreous traction to neo-
SCLERAL BUCKLING vascular tufts, and bleeding vessels can be managed and endo-
PRP combined. Two-needle, in-office fluid-fluid exchange can
Although a scleral buckle could potentially support any reti- be used if medical conditions do not permit surgery under
nal breaks that cannot reach the RPE after dissection, internal monitored local anesthesia. If any neovascularization is pres-
fluid-air exchange, and internal drainage of SRF, retinectomy ent, endo-PRP should be combined with the procedure.
is preferred. Prophylactic encircling bands have not been used
by one of the authors (S.C.) since the early 1980s because of
improved vitreous cutters, fluidics, and techniques. Po st o p e rat ive Cat aract
If a visually significant cataract occurs in the best or
only-vision eye, it should be removed using phacoemulsi-
RESULTS fication and PCL implantation. If the cataract occurs in the
poorer vision eye, the patient can be observed with ultra-
Greater than 80% of diabetic TRD patients managed in the sound if medical status does not warrant surgery.
previously described manner will sustain visual improvement
with vision greater than 5/200 (31–33). Ninety-seven percent
of the retinas of the patients are attached at the 2-week post- Eryt h ro clast ic (He m o lyt ic) Glauco m a
operative visit, but even after reoperation, 5% of the patients Erythroclastic (hemolytic) glaucoma is best prevented by blunt
are blind (34) from AVCFVP and glial recurrence with sec- cannula extrusion with the 25-gauge soft-tip cannula, trim-
ondary retinal detachment (35–37). The incidence of glial ming of the vitreous skirt, and coagulation of all bleeding ves-
recurrence is less with delamination than with segmentation. sels. If the pressure exceeds 25 mm Hg, topical alpha-agonists
Glial recurrence never occurs in truncation-only cases. and beta-blockers will usually control the pressure. On rare
Some aphakic patients with attached retinas ultimately occasions, reoperation may be needed to control the pressure.
become blind from NVG in spite of careful management.
NVG correlates with the presence of active retinal neovascu-
larization. AVCFVP causes permanent blindness in some of Ne o vascular Co m p licat io n s
the phakic cases (38). Just as retinal neovascularization is the most significant
Some of the patients with attached retinas do not have complication of the unoperated PDR eye, NVG and AVCFVP
improved vision because of photoreceptor damage and reti- (AHFVP, RLNV) are the most severe problems in the postvit-
nal ischemia. Some of the successfully operated cases ulti- rectomy eye. An understanding of the pathogenesis is neces-
mately become blind from ischemic optic neuropathy. Some sary to reduce and manage neovascular complications.
become blind from open angle glaucoma. Case selection has Anterior segment and AVC neovascularization are due to
a large impact on success rate, but the goal is to help every- VEGF released from hypoxic but noninfarcted retina (45–49).
one possible, not to improve the success rate by elimination PRP is successful in reducing VEGF by causing the destruc-
of difficult cases. Patients with good results at 6 months typi- tion of hypoxic retinal areas, transient release of an inhibitor
cally have excellent long-term success (39–43). substance (50), and increased choroidal oxygenation of the
retina (51). Trabecular meshwork neovascularization with-
out peripheral anterior synechia or apparent iris neovascu-
COMPLICATIONS larization can cause severe glaucoma. It is no longer thought
that anterior segment neovascularization is secondary to a
He m o rrh ag e
circulatory disturbance or that iris neovascularization some-
Immediate postoperative intraocular hemorrhage can occur how migrates to the trabecular meshwork. Although vitrec-
from ERM vascular attachment points, nontreated new tomy can induce changes in the oxygen distribution in the
186 SECTION IV ■ Disease-Sp ecific Managem ent

globe, this observation does not explain the transmissibility If any neovascularization is seen in the postoperative
of ocular neovascularization from human vitreous specimens course, immediate intravitreal bevacizumab and PRP should
to bioassay systems, which can only be explained by VEGF. be performed. It is not advisable to wait for pressure eleva-
Intravitreal bevacizumab has been shown by the authors tion, which may obscure the view and lead to irreversible
to cause regression of anterior segment neovascularization NVG. While PRP may not affect the intraocular pressure, it
and should be used as necessary to treat active iris neovas- decreases fibrin release and hemorrhage from the iris vessels
cularization in diabetic patients. This treatment should be that contribute to the phthisis process. Although on occa-
combined with adequate PRP for long-term control of VEGF sion iris neovascularization will disappear spontaneously or
production and recurrence of neovascularization. stabilize, it is better to treat all cases of iris neovasculariza-
tion with bevacizumab and PRP.
If the eye pressure exceeds 25 mm Hg, topical timolol
THE BARRIER CONCEPT may be effective and can be used in combination with bri-
monidine, latanoprost, and topical carbonic anhydrase
VEGF encounters sequential barriers in its anterior diffusion inhibitors. If topical treatment cannot keep the pressure in
en route to ocular egress through the trabecular meshwork the mid-20s, glaucoma surgery may be required. Presumably
(Fig. 19.14). In nonoperated eyes, Neovascularization (NVE) because of poor perfusion, diabetics have poor tolerance for
and Neovascularization of the disc (NVD) occur along the elevated pressure. Filtering procedures are effective in some
back surface of the PVC. If vitrectomy has removed the PVC, of these patients but have a tendency to cause hypotony with
neovascularization occurs along the back surface of the AVC. resultant repeated intraocular bleeding.
AVCFVP as first reported by one of the authors (S.C.) was Cyclodestructive procedures can be performed if the
previously incorrectly thought to be due to “fibrovascular patient cannot tolerate an operating room procedure. Cryo-
ingrowth” from the sclerotomies. therapy on bare sclera, for 6 clock hours, 4 to 5 mm poste-
In aphakic eyes or when present in high concentrations, rior to the limbus to avoid the trabecular meshwork seems
VEGF encounters the trabecular meshwork barrier, causing to have best results. The treatments are held at 80°C for
NVG. Iris neovascularization serves to indicate the presence 1 minute. Although this can be quite effective in controlling
of VEGF in the anterior segment. Trabecular meshwork neo- the pressure using a single treatment, many of these patients
vascularization, however, has a direct role in NVG. If a suc- go on to further fibrin release, cyclitic membrane formation,
cessful filtering procedure is performed in a diabetic, aphakic, fibrovascular proliferation, and phthisis bulbi. Transscleral
vitrectomized eye, anterior segment neovascularization will Nd-YAG laser cyclodestruction has largely replaced cyclocry-
frequently disappear. This is analogous to the disappearance otherapy, since ocular surface damage and inflammation are
of NVE and NVD after removal of the PVC by vitrectomy. In greatly diminished, and does not require intraocular sur-
these filtered cases, neovascularization occurs on the inside gery. When possible, endocyclophotocoagulation combined
of the bleb, which can be thought of as the final barrier. with PRP is better than cryotherapy, transscleral laser, and

Fig u re 1 9 .1 4 ■ Seq uential b arriers


to anterior d iffusion of VEGF. Barriers
also act as sub strates for neovascular
and fibrovascular p roliferation.
CHAPTER 19 ■ Diabetic Retinopathy 187

ultrasonic cyclodestructive procedures because of reduced 2. Klein R, Klein BE, Moss SE, et al. The Wisconsin Epidemio-
inflammation, less pain, and better visual prognosis. logic Study of Diabetic Retinopathy. III. Prevalence and risk of
diabetic retinopathy when age at diagnosis is 30 or more years.
Arch Ophthalmol 1984;102:527–532.
3. The Diabetes control and complications trial research group.
An t e rio r Vit re o us Co rt e x Fib ro vascular Progression of retinopathy with intensive versus conventional
Pro life rat io n treatment in the Diabetes Control and complications trial.
Ophthalmology 1995;102:647–661.
If AVCFVP develops, VEGF and other cytokines will cause 4. Chew EY, Klein ML, Ferris FL, et al. Association of elevated
cellular migration and proliferation on the AVC. The mem- serum lipid levels with retinal hard exudates in diabetic retin-
brane causes a characteristic ring-like equatorial TRD fol- opathy. Early treatment of Diabetic retinopathy Study report
lowed by total retinal detachment. This configuration can be 22. Arch Ophthalmol 1996;114:1079–1084.
noted on ultrasound and must be operated on immediately. 5. Early treatment of diabetic retinopathy study research group.
Early photocoagulation for diabetic retinopathy: ETDRS report
The retrolental, retro-IOL, or cyclitic membrane should be 9. Ophthalmology 1991;98:766–785.
detected as early as possible by looking obliquely at the slit 6. Antcliff RJ, Spalton DJ, Stanford MR, et al. Intravitreal triamci-
lamp to ensure early treatment. Treatment requires ECL, nolone for uveitic cystoid macular edema: an optical coherence
removal of the capsule and cyclitic membrane with applica- tomography study. Ophthalmology 2001;109:765–772.
tion of the bipolar diathermy to the resected edges, internal 7. Pollack JA, ISIS Trial Group. Steroid for Diabetic Macular
Edema: The ISIS trial. Presentation in the Retina Annual Sub-
fluid-air exchange, and long-term silicone surface tension specialty Day, American Academy of Ophthalmology Annual
management. Extensive PRP is necessary to reduce further Meeting, Chicago, IL, October 2005.
neovascularization. 8. Vinores SA, Derevjanik NL, Ozaki H, et al. Cellular mecha-
nisms of blood-retinal barrier dysfunction in macular edema.
Doc Ophthalmol 1999;97(3–4):217–228.
Rh e g m at o g e n o us Re t in al De t ach m e n t 9. Ozaki H, Hayashi H, Vinores SA, et al. Intravitreal sustained
release of VEGF causes retinal neovascularization in rabbits
Peripheral rhegmatogenous retinal detachment is rela- and breakdown of the blood-retinal barrier in rabbits and
tively infrequent in diabetic cases. If retinal detachment primates. Exp Eye Res 1997;64(4):505–517.
10. Funatsu H, Yamashita H, Sakata K, et al. Vitreous levels of vas-
occurs, it is usually related to retinal breaks missed at the cular endothelial growth factor and intercellular adhesion mol-
time of the original surgery or vitreous incarceration in the ecule 1 are related to diabetic macular edema. Ophthalmology
wounds. Postvitrectomy retinal detachments usually can- 2005;112(5):806–816.
not be managed with scleral buckling alone. A greater suc- 11. Rosenfeld PJ, Fung AE, Puliafito CA. Optical coherence tomog-
cess rate is usually obtained by using a vitrectomy revision raphy findings after an intravitreal injection of bevacizumab
(avastin) for macular edema from central retinal vein occlu-
approach with a search for residual traction or glial recur- sion. Ophthalmic Surg Lasers Imaging 2005;36(4):336–339.
rence, internal drainage of SRF, internal fluid-air exchange, 12. The Diabetic Retinopathy Vitrectomy Study Research Group.
focal endophotocoagulation, and air-gas or air-silicone Early Vitrectomy for severe vitreous hemorrhage in diabetic
exchange. retinopathy: two year results of a randomized trial–DRVS
Study report 2. Arch Ophthalmol 1985;103:1644–1652.
13. Diabetic Retinopathy Vitrectomy Study Research Group. Early
Glial Re curre n ce vitrectomy for severe proliferative diabetic retinopathy in eyes
with useful vision: results of a randomized trial: DRVS report
Epiretinal surgery, especially with peeling, can result in a recur- 3. Ophthalmology 1988;95:1307–1320.
rence of glial proliferation. Contrary to previous teaching, no 14. Diabetic Retinopathy Study Research Group. Early vitrectomy
vitreous substrate (often called “scaffold”) is required for a glial for severe vitreous hemorrhage. Four-year results of a random-
ized trial: DRVS report 5. Arch Ophthalmol 1990;108:958–964.
recurrence. Glial tissue can proliferate directly on the retinal 15. Doft BH, Blankenship GW. Single versus multiple treatment
surface. Fibrin from ERM epicenters can constitute a bridge- sessions of argon laser panretinal photocoagulation for prolif-
like substrate along which glial tissue can reproliferate. erative diabetic retinopathy. Ophthalmology 1982;89:772–779.
Glial recurrences are managed with scissors delami- 16. The Diabetic Retinopathy Study Group. Preliminary report
nation and internal drainage of SRF, internal fluid-air on the effects of photocoagulation therapy. Am J Ophthalmol.
1976;81:383–396.
exchange, endophotocoagulation, and long-term silicone oil 17. Machemer R, Buettner H, Norton EW, et al. Vitrectomy: a pars
for rhegmatogenous confinement if there is a rhegmatoge- plana approach. Trans Am Acad Ophthalmol 1971;75:813–820.
nous component. Frequently, retinectomy is required. These 18. Arevalo JF, Maia M, Flynn HW Jr, et al. Tractional retinal detach-
membranes are tightly adherent to the retina and cannot be ment following intravitreal bevacizumab (Avastin) in patients
treated with membrane peeling. with severe proliferative diabetic retinopathy. Br J Ophthalmol
2008;92(2):213–216. [Epub 2007 Oct 26.]
19. Cohen HB, McMeel W, Franks EP. Diabetic traction detach-
References ment. Arch Opthalmol 1979;97:1268.
20. Flinn C, Charles S. The natural history of diabetic extramacu-
1. Klein R, Klein BE, Moss SE, et al. The Wisconsin epidemiologic lar traction detachment. Arch Ophthalmol 1981;99:66.
study of diabetic retinopathy. II. Prevalence and risk of diabetic 21. Hykin PG, Gregson RM, Stevens JD, et al. Extracapsular cata-
retinopathy when age at diagnosis is less than 30 yrs. Arch Oph- ract extraction in proliferative diabetic retinopathy. Ophthal-
thalmol 1984;102:520–526. mology 1993;100:394–399.
188 SECTION IV ■ Disease-Sp ecific Managem ent

22. Holekamp NM, Shui YB, Beebe D. Lower intraocular oxygen 37. Michels RG. Vitrectomy for complications of diabetic
tension in diabetic patients: possible contribution to decreased retinopathy. Arch Ophthalmol 1978;96:237.
incidence of nuclear sclerotic cataract. Am J Ophthalmol 38. Charles S. Vitreous surgery for diabetic traction detachment.
2006;141(6):1027–1032. Presented at Frontiers in Ophthalmology. Phoenix, Arizona,
23. Hartley KL, Smiddy WE, Flynn HW Jr, et al. Pars plana vit- February 18, 1982.
rectomy with internal limiting membrane peeling for diabetic 39. Rice TA, Michels RG. Long-term anatomic and functional
macular edema. Retina 2008;28(3):410–419. results of initially-successful vitrectomy for diabetic retinopa-
24. Blankenship GW. Preoperative iris rubeosis and diabetic vitrec- thy. Am J Ophthalmol 1980;90:297.
tomy results. Ophthalmology 1980;87:176. 40. Blankenship CW. Stability of pars plana vitrectomy results
25. Scuderi 1, Blumenkranz M, Blankenship G. Regression of dia- for diabetic retinopathy complications, a comparison of five-
betic rubeosis iridis following successful surgical reattachment year and six-month postvitrectomy findings. Arch Ophthalmol
of the retina by vitrectomy. Retina 1982;2:193. 1981;99:1009.
26. Little HL. Rubeosis indis after vitrectomy for complications 41. Rice TA, Michels RG, Palmer L. Late results of initially-successful
of diabetic retinopathy. In: Little HL, Jack RL, Patz A, et al., vitrectomy in diabetes. Dev Ophthalmol 1981;2:286.
eds. Diabetic retinopathy. New York: Thieme-Stratton, Inc.; 42. Blankenship GW, Machemer R. Long-term diabetic vitrec-
1983:315–340. tomy results, report of 10 year follow-up. Ophthalmology
27. Lupinacci AP, Calzada JI, Rafieetery M, et al. Clinical out- 1985;92:503.
comes of patients with anterior segment neovascularization 43. Blankenship GW. Pars plana vitrectomy for diabetic retinopa-
treated with or without intraocular bevacizumab. Adv Ther thy, a report of 8 years’ experience. In: S. Karger AG, ed. Mod-
2009;26(2):208–216. [Epub 2009 Feb 20.] ern problems in ophthalmology. Basel, Switzerland: Medical and
28. Blankenship G, Cortez R, Machemer R. The lens and pars Scientific Publishers; 1979:376–386.
plana vitrectomy for diabetic retinopathy complications. Arch 44. Schachat AP, Oyakawa RT, Michels RG, et al. Complications
Ophthalmol 1979;97:1263. of vitreous surgery for diabetic retinopathy. II. Postoperative
29. Rice TA, Michels RG, Maguire MG, et al. The effects of complications. Ophthalmology 1983;90:522.
lensectomy on the incidence of ins neovascularization and 45. Glaser BM, D’Amore PA, Michels RG, et al. Demonstration of
neovascular glaucoma after vitrectomy for diabetic retinopathy. vasoproliferative activity from mammalian retina. J Cell Biol
Am J Ophthalmol 1983;95:1. 1980;84:298.
30. Glaser BM. Relationship of cataract extraction and rubeosis in 46. Glaser BM, D’Amore PA, Michels RG, et al. The demonstration
diabetes mellitus. Ophthalmology 1983;90:819. of angiogenic activity from ocular tissues: preliminary report.
31. Blankenship GW, Machemer R. Pars plana vitrectomy for the Ophthalmology 1980;87:440.
management of severe diabetic retinopathy, an analysis of 47. Glaser BM, D’Amore PA, Lutty GA, et al. Chemical mediators
results five years after surgery. Ophthalmology 1978;85(6):553. of intraocular neovascularization. Trans Ophthalmol Soc UK
32. Michels RG. Vitrectomy for the complication of diabetic retin- 1980;100:369.
opathy. Arch Ophthalmol 1978;96(2):237. 48. Glaser BM, D’Amore PA, Michels RG. The effects of human
33. Aaberg T. Clinical results in vitrectomy for diabetic traction intraocular fluid on vascular endothelial cell migration: cor-
retinal detachment. Am J Ophthalmol 1979;88(2):246. relation with intraocular neovascularization. Ophthalmology
34. Diabetic retinopathy Study Research Group. Four risk factors 1981;88:986.
for severe visual loss in diabetic retinopathy: the third report 49. Fett JW, Strydom DJ, Lobb RR, et al. Isolation and characteriza-
from the DRS. Arch Ophthalmol 1979:97:654–655. tion of angiogenin, an angiogenic protein from human carci-
35. Rice TA, Michels RG. Complications of vitrectomy. In: Little noma cells. Biochemistry 1985;24:5480–5486.
HL, Jack RL, Patz A, et al., eds. Diabetic retinopathy. New York: 50. Glaser BM, Campochiaro PA, Davis JL, et al. Retinal epithelial
Thieme-Stratton, Inc.; 1983:315–340. cells release an inhibitor of neovasculanzation. Arch Ophthal-
36. Michels RG. Vitreous surgery in proliferative diabetic retinopa- mol 1985;103:1870.
thy. In: Shimizu K, Oosterhuis LA, eds. Acta XXIII Concilium 51. Wolbarsht ML, Landers MB. The rationale of photocoagulation
Ophthalmologicurm, Part 1, Amsterdam/Oxford: Excerpta Med- therapy for proliferative diabetic retinopathy—a review and a
ica; 1979:420. model. Ophthalmic Surg 1980;11:235.
C H AP TER
20
VENOUS OCCLUSIVE DISEASES

Very little progress has been made in the past structural changes. Prior to the widespread use of intravitreal
two decades in elucidating the precise patho- triamcinolone, it was thought that steroid glaucoma only
genesis of central retinal vein occlusion (CRVO); occurred in about 6% of the population referred to as steroid
controversy still exists about the mechanism of responders. This notion was based on studies using topi-
arterial compression of the vein as well as the role of throm- cal 1.0% prednisolone acetate. Steroid glaucoma has been
bosis. There is less controversy about the pathogenesis of shown to occur in approximately 30% of patients receiving
branch retinal vein occlusion (BRVO); compression of the intravitreal triamcinolone (3); unfortunately, efficacy is pro-
vein by a branch artery is widely accepted, but again the role portional to the steroid glaucoma and cataract side effects.
of thrombosis is unclear. Prior to the use of intravitreal triamcinolone, it was thought
Although many physicians order a battery of lab tests, that steroid glaucoma was reversible after the drug was with-
only homocysteine and anticardiolipin antibodies have been drawn, but many patients receiving intravitreal triamcinolone
shown in meta-analysis to be associated with retinal vein develop permanent glaucoma. Steroid cataracts occur in at
occlusion (RVO) (1). Systemic hypertension, atherosclerotic least 50% of the patients receiving intravitreal triamcinolone
cardiovascular disease, and associated risk factors are risk (4). The authors do not use intravitreal triamcinolone for
factors for RVO as well. The role of systemic disease does not RVO patients, but controversy exists among physicians still
lead to a systemic treatment for RVO, although it emphasizes using this therapy about whether the drug should be used in
the role of wellness-based approaches to prevention. patients with a personal or family history of glaucoma. Kena-
Systemic anticoagulation does not prevent RVO; many log use is off-label in the United States; the manufacturer
patients presenting with vein occlusions are adequately anti- sent a warning letter to the retinal community in 2007. Some
coagulated (2). Similarly, there is no randomized clinical surgeons filter and resuspend Kenalog, while others decant
trial evidence that systemic anticoagulation is effective in the the diluent to reduce the adverse effects related to preser-
treatment of RVOs. vatives. Triesence (Alcon Laboratories, Ft. Worth, Texas), a
preservative-free triamcinolone, received FDA approval in
2007 and addresses the preservative toxicity issue.
PHARMACOTHERAPY Dexamethasone (Decadron) can be used to treat macu-
lar edema secondary to RVO but has a very short duration
Historical treatments of interest include topical administra- of action, with an approximate half-life of 4 hours (5). Short
tion of potassium iodide and pilocarpine, antihyperlipidemic duration of action reduces the chances of cataract and steroid
agents, anticoagulants, fibrinolytic agents, hyperosmotic glaucoma, but the only potential clinical benefit is to reduce
agents, carbogen inhalation, and isovolemic hemodilution. retinal thickness a few days prior to laser treatment. Sustained-
These treatments have been demonstrated to be largely inef- release dexamethasone (Ozurdex, Allergan), was recently
fective and therefore abandoned. introduced into the US market.The duration of action is lon-
The modern era of pharmacotherapy for RVO began ger than intravitreal triamcinolone, with a single treatment
in the 1990s with intravitreal triamcinolone acetonide producing statistically significant best corrected visual acuity
(Kenalog, Bristol Myers Squibb) injections to treat macu- improvements 90 days after treatment. Kuppermann et al.
lar edema. Many observers noted marked decrease in reti- (6) have shown that the dexamethasone molecule is less toxic
nal thickness by optical coherence tomography but modest than triamcinolone without preservative when tested against
increase in best corrected visual acuity presumably because human retinal neuronal cells, glial cells, and retinal pigment
of ischemia-induced neuronal loss and permanent macular epithelial cells in tissue culture (7,8).

189
190 SECTION IV ■ Disease-Sp ecific Managem ent

An intravitreal fluocinolone 0.59 mg sustained-release 3 months, with a 25% improvement in central macular thick-
device (Retisert, Bausch & Lomb, St. Louis, Missouri) is FDA ness (14). Case reports of intravitreal Avastin for CRVO have
approved for posterior uveitis and has been used for the treat- shown potential benefit of this treatment in short-term follow-
ment of macular edema by some surgeons. Unfortunately, up (15). Ranibizumab (Lucentis) is the only FDA-approved
the Retisert produces a 90% glaucoma incidence, and at least anti-VEGF compound other than the minimally effective
30% of the patients ultimately require a glaucoma filtering pegaptanib sodium (Macugen), but neither is approved for
procedure (9). This level of adverse effects is simply not jus- the RVO indication. The RVO study arm of the Ranibizumab
tifiable when treating macular edema secondary to RVO. for Edema of the Macula in Diabetes (READ) demonstrated
Nonsteroidals are effective when used topically for a doubling of the visual angle after three monthly injections
macular edema after cataract surgery. Postoperative macular (16). The authors have observed excellent results from using
edema (Irvine-Gass syndrome) is inflammatory in nature, intravitreal Avastin on all BRVOs and CRVOs at the initial
while macular edema in RVO patients is primarily driven visit and every month until all intraretinal hemorrhage, axo-
by vascular endothelial growth factor (VEGF) and increased plasmic stasis, and edema have regressed as recommended
hydrostatic pressure. There is little direct evidence that by Richard Spaide (17). Often these cases have complete
inflammation is a significant factor in macular edema, but the resolution of the BRVO or CRVO appearance even without
observation that intravitreal steroids reduce macular edema evidence of collaterals. It is of interest that VEGF creates a
in RVO suggests a role for inflammatory mediators. Ketoro- CRVO picture in animal models (17).
lac tromethamine 0.5% (Acular, Allergan, Irvine, California) Intracameral and intravitreal Avastin injection results in
and diclofenac 0.1% (Voltaren, Novartis Ophthalmics) sup- rapid and dramatic regression of iris neovascularization sec-
press the anterior segment inflammation that causes postop- ondary to CRVO; effects are seen within 48 hours and last at
erative macular edema but have minimal direct effect on the least 4 weeks (18, 19). Typically, if no cicatricial closure of the
macula. Bromfenac 0.09% (Xibrom, Ista Pharmaceuticals, iridocorneal angle was seen on gonioscopy, there is a marked
Irvine, California) and nepafenac (Nevanac, Alcon Labora- decrease in intraocular pressure as well, presumably because
tories, Ft. Worth, Texas) have been shown to reduce mac- vessels on the surface of the trabecular meshwork regress
ular edema secondary to retinal vascular disease in several and the viscosity of the aqueous humor decreases markedly
observational series as well as to be effective for postcataract (much less flare). Avastin and presumably any anti-VEGF
surgery of macular edema (10,11). Nepafenac is a prodrug compound must be used with panretinal photocoagulation
that has been shown to produce significant drug levels at the for sustained effect on anterior segment and retinal neovas-
macula in phakic and pseudophakic eyes (12). cularization excluding choroidal neovascular membranes.
Proliferative diabetic retinopathy, RVOs, and retinopa- Avastin is very effective prior to focal, grid, or sector
thy of prematurity all produce high levels of intravitreal photocoagulation for RVOs to decrease retinal thickness and
VEGF (Lloyd Paul Aiello), which is not the case for retinal decrease laser fluence requirements. The anti-VEGF agent
detachments or proliferative vitreoretinopathy (PVR) (13). should be injected approximately 1 week prior to laser.
VEGF increases vascular permeability and is a powerful
angiogenic signaling ligand. Anti-VEGF therapy is effective
for macular edema as well as neovascularization secondary LASER THERAPY
to RVO and diabetic retinopathy. Bevacizumab (Avastin)
is a safe and effective anti-VEGF compound, has low cost, Focal, sector, or light grid laser photocoagulation can be
and is widely used for retinal vascular disorders worldwide, used for macular edema secondary to BRVO, hemi–vein
albeit off-label in the United States. Although some physi- occlusion, or CRVO. Treatment with Avastin approximately
cians have expressed concerns about long-term VEGF sup- 1 week prior to laser has dramatically improved results in
pression, this has not proven to be a problem in the 6 to the author’s opinion, although there is no randomized con-
7 years of anti-VEGF compounds use. Initially, there was trolled trial evidence at this time.
concern that an anti-VEGF antibody would cause uveitis, Panretinal photocoagulation is very effective in treating
but this has not proven to be a problem either. The typical iris and retinal neovascularization secondary to hemi-RVO
intravitreal dose, 1.25 mg, is approximately 1/500 the sys- or CRVO but, as pointed out above, is best used combined
temic dose used for cancer. Cancer patients using this drug with anti-VEGF therapy. Steve Schwartz has coined the term
every 2 week intravenously with a much higher dose than “targeted retinal photocoagulation (TRP)” to describe the
is used in the eye have a doubling of the thromboembolic notion of ablation of angiographically proven midperiph-
event rate. Although this has been a matter of concern, there eral ischemia and transition zones to treat diffuse diabetic
is no evidence of increased thromboembolic event rates after macular edema (20). Wessing and Shimizu have advocated
intravitreal injection. this concept for years. The authors believe that this concept
Most physicians inject Avastin approximately every is applicable to RVOs as well.
2 months based on OCT measurements of macular thick- The PASCAL laser from OptiMedica in Santa Clara,
ness. Intravitreal Avastin for BRVO demonstrates visual California, typically uses 30-ms pulses to reduce thermal
acuity improvements from 20/200 at baseline to 20/100 at diffusion, anteriorly to the nerve fiber layer, laterally causing
CHAPTER 20 ■ Venous Occlusive Diseases 191

retinal pigment epithelial “creep,” and posteriorly to the Furthermore, the results of this surgery are difficult to
choroid, causing pain. Although this concept has most often evaluate because the studies were small and because of the
been applied to diabetic macular edema, the authors believe lack of analysis between time after onset of the occlusion and
that it is applicable to RVOs as well. perfusion status prior to surgery. Figueroa has demonstrated
High-power laser pulses were shown by McAllister that capillary reperfusion does not improve after branch vein
and Constable (21) to create retinal-choroidal anastomo- decompression (30).
sis, which seemed to reduce macular edema. Initially oth- Radial optic neurotomy for CRVO was developed by
ers had enthusiasm for this approach, but many physicians Opremcak (31), and a similar procedure, laminar puncture,
noted the development of large disciform scars, vitreous was developed by D’Amico (32). D’Amico performed a care-
hemorrhages, and retinal detachments and abandoned this ful internal review board–controlled trial and determined
procedure. McAllister and Constable advocated 100-ms that the procedure was ineffective, and again there is no
pulses directed at retinal veins; subsequently, Leonard randomized clinical trial evidence of efficacy. In addition to
(22) proposed 1-second burns and avoiding the veins and these issues, there is a significant complication rate and the
reported a decreased incidence of these complications. procedure has been largely abandoned.

References
VITRECTOMY-BASED THERAPY
1. Janssen MC, den Heijer M, Cruysberg JR, et al. Retinal vein
occlusion: a form of venous thrombosis or a complication
Vitrectomy for vitreomacular traction was introduced by of atherosclerosis? A meta-analysis of thrombophilic factors.
Hilel Lewis (23) for diabetic macular edema but is effec- Thromb Haemost 2005;93(6):1021–1026.
tive for carefully selected cases of RVO as well. Fluorescein 2. Mruthyunjaya P, Connor T, Wirostko WJ, et al. Central retinal
angiography to evaluate macular capillary nonperfusion vein occlusion in patients on long-term coumadin anticoagula-
is essential, especially on CRVO cases. The presence of tion. Retina 2006;26(3):285–291.
3. Jonas JB, Degenring R, Kreissig I, et al. Intraocular pressure
an afferent pupillary defect is a relative contraindication elevation after intravitreal triamcinolone acetonide injection.
to pars plana vitrectomy. Optical coherence tomography, Ophthalmology 2005;112:593–598.
especially spectral domain optical coherence tomography 4. Thompson JT. Cataract formation and other complications of
such as Heidelberg Spectralis, has resulted in diagnosing intravitreal triamcinolone for macular edema. Am J Ophthalmol
a much higher incidence of vitreomacular traction than 2006;141(4):629–637.
5. Kwak HW, D’Amico DJ. Evaluation of the retinal toxicity and
is noted even with contact lens examination. Timing is pharmacokinetics of dexamethasone after intravitreal injec-
important in these cases; vitreomacular traction with tion. Arch Ophthalmol 1992;110:259–266.
macular cysts has better visual outcomes if surgery is per- 6. Kuppermann BD, Blumenkranz MS, Haller JA, et al. Random-
formed after a one-month trial of Avastin and nepafenac. ized controlled study of an intravitreous dexamethasone drug
The authors recommend internal limiting membrane delivery system in patients with persistent macular edema.
Arch Ophthalmol 2007;125:309–317.
removal in these cases to ensure complete elimination of 7. Nabih M, Peyman GA, Tawakol ME, Naguib K. Toxicity of
vitreomacular traction. high dose intravitreal dexamethasone. Int Ophthalmol 1991:15:
Vitrectomy in patients without vitreomacular traction is 234–235.
effective in diabetic macular edema. An increasing number 8. Narayanan R, Mungcal JK, Kenney MC, et al. Toxicity of tri-
of surgeons believe that pars plana vitrectomy is effective amcinolone acetonide on retinal neurosensory and pigment
epithelial cells. Invest Ophthalmol Vis Sci 2006;47:722–728.
for macular edema in RVO patients as well. The mechanism 9. Jaffe GJ, Martin D, Callanan D, et al. Fluocinolone Acetonide
in both disease processes is almost certainly similar because Uveitis Study group. Fluocinolone acetonide implant (Retisert)
pars plana vitrectomy permanently increases oxygen tension for noninfectious posterior uveitis; thirty-four-week results
by 7 to 12 mm Hg as shown in the laboratory by Stefansson of a multicenter randomized clinical study. Ophthalmology
(24), and later Chang, and clinically by Holekamp (25–26). 2006;113:1020–1027.
10. Gross JG. Xibrom (bromfenac) reduces cystoid macular edema
Increased oxygen tension is probably the mechanism by associated with vein occlusions, macular epiretinal membrane,
which “sheathotomy” and radial optic neurotomy seem to diabetic retinopathy and age related macular degeneration.
be effective. Poster F-3 presented at: Annual Meeting of the Retina Society.
Branch vein decompression was developed by the Boston, MA, USA, September 27–30, 2007.
first author and subsequently rediscovered and renamed 11. Hariprasad SM, Callanan D, Gainey S, et al. Cystoid and dia-
betic macular edema treated with nepafenac 0.1%. J Ocul Phar-
“sheathotomy” by Opremcak (27). Seitz and Green have macol Ther 2007;23(6):585–590.
both emphasized that there is no sheath; the artery and vein 12. Walters T, Raizman M, Ernest P, et al. In vivo pharmacokinet-
have a common adventitia (28–29). The author abandoned ics and in vitro pharmacodynamics of nepafenac, amfenac,
this procedure after 15 cases but, in retrospect, should ketorolac, and bromfenac. J Cataract Refract Surg 2007;33:
have done a randomized clinical trial. Most surgeons have 1539–1545.
13. Aiello LP, Avery RL, Arrigg PG, et al. Vascular endothelial
now abandoned this procedure as well. Perceived visual growth factor in ocular fluid of patients with diabetic retin-
improvement is probably due to the natural history as well opathy and other retinal disorders. N Engl J Med 1994;331:
as increased oxygen tension from pars plana vitrectomy. 1480–1487.
192 SECTION IV ■ Disease-Sp ecific Managem ent

14. Rabena M, Pieramici DJ, Castellarin AA, et al. Intravitreal 23. Lewis H, Abrams GW, Blumenkranz MS, Campo RV. Vitrectomy
bevacizumab (Avastin) in the treatment of macular edema for diabetic macular traction and edema associated with poste-
secondary to branch retinal vein occlusion. Retina 2007;27: rior hyaloidal traction. Ophthalmology 1992;99(5):753–9.
419–425. 24. Stefansson E. The therapeutic effects of retinal laser treatment
15. Iturralde D, Spaide FR, Meyerle CB, et al. Intravitreal beva- and vitrectomy. A theory based on oxygen and vascular physi-
cizumab (Avastin) treatment of macular edema in central ology. Acta Ophthalmol Scand 2001;79(5):435–440.
retina vein occlusion: a short term study. Retina 2006;26: 25. Barbazetto IA, Liang J, Chang S, et al. Oxygen tension in the
279–284. rabbit lens and vitreous before and after vitrectomy. Exp Eye
16. Campochiaro PA, Hafiz G, Shah SM. Ranibizumab for macular Res 2004;78(5):917–924.
edema due to retinal vein occlusions; implication of VEGF as a 26. Holekamp NM, Shui YB, Beebe DC. Vitrectomy surgery increases
critical stimulator. Mol Ther 2008. oxygen exposure to the lens: a possible mechanism for nuclear
17. Ferrara DC, Koizumi H, Spaide RF. Early bevacizumab treat- cataract formation. Am J Ophthalmol 2005;139:302–310.
ment of central retinal vein occlusion. Am J Ophthalmol 27. Opremcak EM, Bruce RA. Surgical decompression of branch
2007;144:864–871. retinal vein occlusion via arteriovenous crossing sheathotomy.
18. Grisanti S, Biester S, Peters S, et al. Intracameral bevacizumab Retina 1999;19:1–5.
for iris rubeosis. Am J Ophthalmol 2006;142:158–160. 28. Seitz R. Die Netzhautgefäße. Stuttgart, Germany: Georg Thieme
19. Iliev ME, Dommig D, Wolf-Schnurrbursch U, et al. Intravitreal Verlag, 1962.
bevacizumab (Avastin) in the treatment of neovascular glau- 29. Green WR, Chan CC, Hutchins GM, Terry JM. Central retinal
coma. Am J Ophthalmol 2006;142:1054–1056. vein occlusion: a prospective histopathologic study of 29 eyes
20. Schwartz S. Ultra-wide angle angiographically targeted retinal in 28 cases. Trans Am Ophthalmol Soc 1981;79:371–422.
photocoagulation (TRP) combined with anti-VEGF pharmaco- 30. Figueroa MS, Torres R, Alvarez MT. Comparative study
therapy for the treatment of retinal vascular macular edema. of vitrectomy with and without vein decompression for
The Macula Society 30th Annual Scientific Program. June 2, branch retinal vein occlusion: a pilot study. Eur J Ophthalmol
2007. 2004;14(1):40–47.
21. McAllister IL, Constable IJ. Laser-induced chorioretinal venous 31. Opremcak EM, Bruce RA, Lomeo MD, et al. Radial optic neu-
anastomosis for non-ischemic central retinal vein occlusion: rotomy for central retinal vein occlusion: a retrospective pilot
evaluation of the complications and their risk factors. Am J study of 11 consecutive cases. Retina 2001;21:408–415.
Ophthalmol 1998;126:219–229. 32. D’Amico DJ, Lit ES, Viola F. Lamina puncture for central reti-
22. Leonard BC, Coupland SG, Kertes PJ, Bate R. Long-term nal vein occlusion: a pilot study. Arch Ophthalmol 2006;124:
follow-up of a modified technique for laser-induced chorioret- 972–977.
inal venous anastomosis in nonischemic central retinal vein
occlusion. Ophthalmology 2003;110(5):948–954.
C H AP TER
21
TREATMENT OF CHOROIDAL
NEOVASCULAR MEMBRANES

The management of choroidal neovascular were combination with other therapies (like laser) can be
(CNV) membranes in age-related macular entertained.
degeneration (AMD), ocular histoplasmosis
syndrome, myopia, and uveitis has advanced
greatly in the past few years. In general terms, there has been FOCAL THERMAL LASER ABLATION
a progression from nihilistic observation to laser ablation
to submacular surgery, photodynamic therapy (PDT) with The goal of laser therapy is to achieve complete ablation of
and without intravitreal steroids to modern anti–vascular the CNV complex. Laser therapy produces concurrent over-
endothelial growth factor (VEGF) therapy. lying retinal ablation with an immediate scotoma.
Many potential treatments have been promoted by Photocoagulation of juxtafoveal lesions has been shown
positive pilot studies ultimately followed by randomized to produce greater than 50% recurrence rates, frequently
clinical trials showing no significant efficacy. Examples of on the foveal side (3). Many patients present with subfoveal
this sequence include alpha one interferon, thalidomide, lesions; therefore, laser treatment of these lesions was inves-
brachytherapy, anecortave acetate, transpupillary thermo- tigated in the Macular Photocoagulation Study (MPS) as well.
therapy for occult CNV, and external beam radiation. It was shown that treated patients suffer an immediate loss of
Pegaptanib sodium (Macugen) and PDT (Visudyne) greater than three lines of vision but have slightly better vision
therapy resulted in modest slowing in the rate of visual loss after 18 months compared to patients who did not receive laser
but rarely resulted in visual improvement. Ranibizumab therapy. Few physicians ever became comfortable with causing
(Lucentis), a humanized Fab fragment of an anti-VEGF immediate loss of central vision, and this treatment is no lon-
antibody, was the first medication to achieve reliable visual ger performed since Lucentis and Avastin became available.
gains in a significant percentage of patients. Visual results Laser treatment can still be considered for extrafoveal
in several pivotal multicenter, randomized clinical trials CNVs if they are at a substantial distance from the fovea and
were simply outstanding compared to the results previously the total size of the lesion (and the secondary scotoma) is small.
achieved with earlier therapies: 95% of patients avoided In other words, the smaller and more peripheral the lesion is,
moderate visual loss (2 ETDRS lines or 25 letters), 30% to the more amenable it is to laser therapy. It is important for the
40% of the patients experienced improved vision, and 75% lesion to have clearly defined borders on angiography to guar-
achieved true stability. Phillip Rosenfeld introduced the con- antee that all the CNV is ablated whenever laser is preformed.
cept off-label use of bevacizumab (1,2) (Avastin), a related, The advantage of laser above anti-VEGF therapy is
full-length anti-VEGF antibody, as a significantly less expen- single-session treatment with long-term control. The authors
sive alternative to Lucentis. Retrospective, prospective, and, often recommend injection of Avastin or Lucentis concur-
more recently, small randomized trials and a large amount rently with focal laser therapy for extrafoveal lesions with the
of anecdotal experience using Avastin have demonstrated hopes of decreasing recurrence rates.
similar outcomes to Lucentis. The National Eye Institute–
sponsored Comparisons of Age-Related Macular Degenera-
tion Treatments Trials (CATT) is a head-to-head comparison
PREVENTION STRATEGIES
of Lucentis to Avastin and has both protocol and PRN treat-
Die t ary Sup p le m e n t at io n
ment arms for both drugs.
Most patients are currently treated with monotherapy Oral antioxidants such as zinc, beta-carotene, zeaxanthin,
with either bevacizumab or ranibizumab. There are patients lutein, vitamin A, vitamin E, selenium, and others have been
193
194 SECTION IV ■ Disease-Sp ecific Managem ent

advocated for the prevention of AMD and treatment of early use of PDT as monotherapy has largely been abandoned for
stages of AMD or atrophic (dry) AMD. The Age Related Eye choroidal neovascularizations.
Study showed that zinc plus antioxidants reduced AMD The authors currently do not recommend PDT therapy
progression risk by 26% in patients with intermediate AMD. in the management of CNVs, since they aim to avoid any
The Beaver Dam Eye Study looked at the dietary and smok- further injury to the choriocapillaris, which is often dam-
ing history of individuals with AMD compared to matched aged with PDT therapy.
normals and concluded that spinach and, to a lesser extent,
other dark leafy green vegetables slightly reduce the progres-
ANTI-VEGF THERAPY STRATEGIES
sion to AMD (4). Smoking was found to have a doubling
effect on the incidence of AMD (5). The authors predict that
The injection frequency selected for the pivotal Lucentis trials
some specific antioxidants may be proven to have minor
was based on preclinical pharmacokinetic data and assump-
value if given to individuals at genetic risk for AMD (drusen).
tions about the length of time untreated CNV membranes
It is likely that this effect will be greatest in patients who do
remain active. This strategy resulted in patients having an
not eat dark leafy green vegetables, other vegetables, whole
average of 21 injections over 24 months. Rosenfeld introduced
grains, and fruits. Beta carotene has been shown to increase
the concept of treat and observe based on time domain opti-
the risk of lung cancer in smokers in a Swedish study (6). At
cal coherence tomography (OCT) evidence of subretinal fluid
this time, the authors counsel the children of patients with
(SRF), Pigment epithelial detachment (PED), or retinal edema
AMD, drusen patients, and patients with early AMD to stop
(Prospective OCT Imaging of Patients with Neovascular AMD
smoking and eat spinach and other dark leafy green veg-
Treated with Intraocular Ranibizumab (PrONTO) Study).
etables at least five times per week.
More recently, Spaide et al. introduced the concept of treat and
The Age Related Eye Disease Study (AREDS) 2 Study is
extend; generally, the notion is to give a “booster shot” after
underway, which is structured to assess the effects of oral sup-
the CNV is not leaking based primarily on OCT evidence and
plementation of macular xanthophylls (lutein and zeaxanthin)
a 6-week rather than a 1-month appointment. If the lesion is
and/or long-chain omega-3 fatty acids (docosahexaenoic acid
not leaking at the 6-week appointment, another injection is
and eicosapentaenoic acid) on the progression to advanced
given and the patient is scheduled to return in 8 weeks. If the
AMD. An additional goal of the study is to assess whether
lesion remains inactive at this point, no injection is given and
forms of the AREDS nutritional supplement with reduced zinc
follow-up is done at 1- to 2-month intervals. More recently,
and/or no beta-carotene work as well as the original supple-
many physicians, including the authors, use clinical decision
ment in reducing the risk of progression to advanced AMD.
making and include vision, visual complaints, exudates, and
hemorrhage in addition to spectral domain OCT (Spectralis)
Plasm ap h e re sis to determine the need for retreatment. Fluorescein angiogra-
phy (confocal using Spectralis), formerly the primary tool in
A randomized trial of plasmapheresis did not reach the pri-
determining the need for treatment, is still used anytime there
mary endpoint, and there is no scientific rationale for this
is a visual complaint or decreased vision and SD OCT fails to
treatment. Unfortunately, many patients have been treated
reveal the etiology. Indocyanine green angiography is used
using this unproven, expensive treatment.
less often but remains a valuable tool.

Lase r Tre at m e n t o f Druse n


COMBINATION THERAPY
Laser treatment of drusen has been shown to cause faster dis-
appearance of drusen than the spontaneous disappearance Combination therapy is an appealing concept but is appro-
rate (7–9). The reader is reminded that drusen are “associ- priate in some instances and not in others. Combination che-
ated with” AMD; they do not “cause” AMD. Threshold treat- motherapy in oncology is utilized because the agents have
ment has been shown to result in higher CNV rates in two US narrow windows between effective and toxic drug levels as
studies now investigating subthreshold treatment (10,11). well as to provide multiple barriers to the evolution of cancer
Two large randomized clinical trails failed to show a benefit cells. Combination therapy for infectious disease is utilized
of subthreshold laser treatment (Friberg, Fine CAPT). in severe infections when the infectious agent has not been
identified and delayed treatment would produce bad out-
comes. Combination therapy for infectious disease creates
Ph o t o d yn am ic Th e rap y
multiple barriers for the evolution of the infectious agent but
PDT was arguably the first therapy other than focal laser unfortunately leads to higher incidence of resistance.
therapy available for the treatment of choroidal neovascu- Focal and/or panretinal photocoagulation laser plus
larizations (12–20). Since the introduction of anti-VEGF anti-VEGF therapy is very effective in diabetic retinopathy
therapy, the use of PDT has decreased dramatically. There are and can be broadly defined as combination therapy. Topical
still physicians who recommend the use of PDT for certain nonsteroidal (Nevanac) therapy in combination with laser and
patients, often in combination with anti-VEGF therapy. The anti-VEGF compounds is effective for diabetic macular edema
CHAPTER 21 ■ Treatm ent of Choroidal Neovascular Mem branes 195

because of multiple mechanisms: VEGF and inflammation. other hand, do cause foveal damage and are amenable to
Combination therapy has been advocated for CNV, but in the hemorrhage displacement. The authors recommend per-
authors’ view, it has very little place with current therapeu- forming spectral domain OCT on all of these patients,
tic options in view of the extraordinary success of Lucentis since it is often difficult to determine the presence of
and Avastin monotherapy. PDT requires intravitreal steroids, an underlying retinal pigment epithelium (RPE) detach-
which results in an approximately 30% incidence of steroid ment or sub-RPE hemorrhage that could give the clini-
glaucoma and approximately 90% incidence of cataract while cal impression of massive macular elevation. If the OCT
only modestly reducing the need for anti-VEGF therapy. demonstrates a subfoveal RPE detachment or sub-RPE
Induction with Lucentis or Avastin followed by main- hemorrhage with a thin overlying subretinal hemorrhage,
tenance with pegaptanib sodium (Macugen) is based on the the authors do not recommend surgical displacement and
notion that pan-isoform suppression (Lucentis, Avastin) may treat with anti-VEGF monotherapy as described previ-
result in neuronal loss but suppressing only the so-called ously. Only those patients in whom the OCT demonstrates
pathological 165 isoform would not. Long-term pan-isoform thick subfoveal hemorrhage should be treated with surgi-
suppression has not been demonstrated to have risk in cal displacement.
patients. Another important clinical consideration for surgical
Vitrectomy to enable epiretinal radiation with a Stron- displacement is the duration between first symptoms and
tium 90 probe (NeoVista) is currently being studied but surgery. The natural history of massive submacular hemor-
requires anti-VEGF therapy to be effective, produces cata- rhages is first to appear with dark red submacular blood that
ract in 90% of the AMD patients because of the vitrectomy, changes in color over the course of weeks to a slate gray
reduces the effectiveness of the anti-VEGF agent by eliminat- appearance as the blood is dehemoglobinized. By the time
ing the inherent sustained-release action of vitreous, creates the hemorrhage appears gray, the clot is frequently too dense
retinal detachment risk, and potentially will produce radia- and hard to be amenable for displacement, and often there is
tion retinopathy. already photoreceptor damage. Given these considerations,
it is important to proceed with surgical displacement within
2 weeks of initial symptoms, while the hemorrhage is still
Vit re ct o m y Surg e ry fo r Sub m acular
dark red in clinical appearance.
He m o rrh ag e Disp lace m e n t
The surgical sequence is as follows: A core vitrectomy
Submacular hemorrhages have been surgically removed is performed with 25-gauge sutureless technique, a poste-
since the early 1990s. These cases have widely varying rior vitreous separation is carefully created if not present,
outcomes based on the underlying pathology and surgical and tPA is injected into the clot using a 38-gauge polyamide
trauma involved (21–23). Some investigators have injected cannula (developed for macular translocation surgery). The
tissue plasminogen activator (tPA) under the retina at the tPA can be injected using the viscous fluid injection system
time of surgery and wait approximately 45 minutes for of the Accurus and Constellation vitrectomy machines to
apparent liquefaction of the clot before aspiration (24–27). allow the surgeon to use foot control of injection pressure.
Others inject tPA into the vitreous approximately 24 hours The VFC system should be set to a maximum of 12 mm Hg
before surgical removal in an attempt to liquefy the blood to prevent overpressuring the subretinal space. A slow, con-
clot (28). It has been reported that tPA can be injected into trolled injection is then achieved and a pocket of subretinal
the vitreous cavity in the office followed by a gas injection tPA resembling an exudative retinal detachment is created
and face-down positioning (29). Hilel Lewis has shown that underneath the macula and further expanded below the
tPA does not cross the primate retina (30). This less invasive inferior vascular arcade to create a path for the hemorrhage
method was thought to displace the blood away from the to be easily displaced.
macula; however, there is no randomized clinical trial evi- Total fluid-air exchange is performed, and the patient is
dence of efficacy. The authors no longer recommend intra- kept in the supine position for 45 minutes after surgery. The
vitreal tPA. patient is then placed in the seated position so that the air
The authors have positive experience with a technique bubble will displace the liquefied blood clot inferiorly. Verti-
developed by Hilel Lewis to displace submacular hemor- cal position should be maintained for 48 to 72 hours to keep
rhage from underneath the fovea, with the aim of decreas- the displaced hemorrhage away from the fovea while the
ing foveal photoreceptor damage caused directly from the injected SRF is reabsorbed. Timing is important; blood clots
submacular hemorrhage. Thin submacular hemorrhages over 2 weeks old should not be operated. Surgical aspiration
(those that do not have mass effect and do not cause foveal of subretinal blood results in damage to the photoreceptor
elevation) do not require surgery and can be managed with outer segments and Retinal Pigment Epithelium (RPE) apical
anti-VEGF monotherapy, as the hemorrhage will invariably processes and should be avoided. Intravitreal Avastin should
clear spontaneously and the final visual outcomes are not be used to treat underlying CNV membranes after successful
worsened by the presence of thin submacular hemorrhage. displacement of the blood.
Massive submacular hemorrhages (those that cause Surgical success should be defined as only thin subfo-
macular elevation from submacular mass effect), on the veal hemorrhage on postoperative day one.
196 SECTION IV ■ Disease-Sp ecific Managem ent

membranes present for over 6 months have almost no chance


SUBMACULAR SURGERY
of visual improvement after membrane removal because of
AND MACULAR TRANSLOCATION irreversible photoreceptor damage. Type I membranes seen
in AMD cases are under the RPE and cannot be removed
In 1991, Thomas and Kaplan reported submacular surgery
without removing the overlying RPE. Type II membranes,
for subretinal neovascular membranes secondary to pre-
as seen in idiopathic and histoplasmosis cases, are on the
sumed ocular histoplasmosis syndrome (31). Subsequently,
anterior surface of the RPE and can have a reasonable prog-
these authors and many other surgeons applied their tech-
nosis. Anti-VEGF therapy is the first-line treatment for all
nique to idiopathic submacular membranes as well as
CNV membranes, but submacular surgery can be effective
membranes secondary to AMD and many other disease pro-
for larger subfoveal, nonleaking membranes.
cesses. In 1993, the principal author developed a simplified
AMD cases have a very poor visual prognosis after mem-
approach to submacular surgery, which will be described
brane removal. Approximately 20% of the AMD patients
herein along with a discussion of indications in the context
have improved vision, 60% have unchanged vision, and
of the negative results of the Submacular Surgery Trial and
20% have worse vision after submacular surgery. Poor vision
the advent of effective anti-VEGF therapy.
after submacular surgery in these patients can be explained
Although the initial article referred to removal of “neo-
by several factors: (a) diffuse, ongoing RPE disease, (b) high
vascular” membranes, it is actually a fibrovascular scar that
incidence of recurrences, (c) photoreceptor and RPE damage
is removed. It is common for the scar (membrane) to be
from scar and hemorrhage, (d) surgical removal of the RPE,
significantly larger than the “net” seen on the angiogram
and (e) absence of the choriocapillaris. The membrane must
presumably because of vascular regression in some areas
be located between the retina and the RPE, not under the
(Fig. 21.1).
RPE, in order to permit surgery without inadvertent removal
of a segment of RPE. Most, if not all, AMD cases have type I
membranes, which are under the RPE. The authors have not
INDICATIONS FOR SUBMACULAR performed submacular surgery on patients with AMD since
SURGERY FOR SUBFOVEAL MEMBRANES 1994. Determination that the patient has AMD is not made
by age alone but by the presence of drusen in the affected
Presumed photoreceptor and RPE viability are an absolute and/or other eye. A 50-year-old patient with advanced
requirement when considering submacular surgery. The drusen has “AMD,” while a 70-year-old patient with a uni-
membrane acts as a diffusion barrier between the photore- lateral membrane and no drusen in either eye probably has
ceptors and the RPE and leads first to degeneration of the an idiopathic CNV membrane. The Submacular Surgery
outer segments and ultimately the inner segments. Free iron Trial results were released in November 2004 and did not
from blood in the subretinal space for an extended period of demonstrate a benefit for any AMD cases with vision better
time is toxic to the photoreceptor inner segments. Cases with than 20/100.
Idiopathic cases have the best prognosis, while his-
toplasmosis is the most frequent indication in the cen-
tral United States. The rare myopic patient with a recent,
medium to large well-defined lesion can benefit from sub-
macular surgery. Selected angioid streaks and trauma cases
are candidates for surgery as well.

SURGICAL SEQUENCE

A core vitrectomy is performed (Fig. 21.2) to facilitate pas-


sage of the instruments and to enable fluid-air exchange.
An aggressive attempt to create a posterior vitreous detach-
ment (PVD) in submacular surgery cases is unnecessary and
a potentially dangerous step. Many surgeons believe that
forceful creation of a PVD is a required step, while in fact it
may increase the chance of retinal detachment and/or dam-
age the optic nerve. The principal author has had no rheg-
matogenous detachments or retinotomy-related problems in
a series of over 250 cases with no aggressive attempt to make
Fig ure 2 1 .1 ■ It is com m on for the fibrovascular scar (neovas- a PVD.
cular m em brane) to b e significantly larger than the “net” seen Thomas and Kaplan recommended injection of balanced
on angiography. salt solution (BSS) under the retina to create a small retinal
CHAPTER 21 ■ Treatm ent of Choroidal Neovascular Mem branes 197

Fig ure 2 1 .2 ■ Core vitrectom y


is p erfo rm ed with n o sp ecific
attem p t to m ake a PVD.

detachment before removing the membrane (1). The senior preventing damage from shearing photoreceptors from the
author (S.C.) observed that this step occasionally results in an RPE in an area larger than the membrane.
acute hydraulic foveal hole. The author has learned that other A very small retinotomy is made with the 25-gauge
surgeons have experienced this problem as well. For this rea- microvitreoretinal (MVR) blade at the outer margin of
son, the principal author has omitted the BSS injection step the membrane (Fig. 21.3), usually in the superotemporal
since 1992. In addition to preventing hydraulic rupture of quadrant. The retinotomy is made by teasing the nerve
the fovea, omitting BSS injection has the added advantage of fibers apart along the orientation of the nerve fibers rather

Fig u re 2 1 .3 ■ A nonm od ified


MVR blade is used to create a reti-
notom y by teasing the nerve layer
ap art. The retinotom y should be
m ade at the outer edge of the CNV
m em brane. If the CNV m em brane
extends from the fovea und er the
p ap illom acular (PM) b un d le, a
radial incision should be m ade in
the PM bundle. There is no need
to create a retinal detachm ent by
injecting BSS, thereby avoiding a
hydraulic m acular hole.
198 SECTION IV ■ Disease-Sp ecific Managem ent

Fig u re 2 1 . 4 ■ En d -g rasp in g
forcep s are used to g rasp th e
subfoveal m em brane at the outer
edge on the top surface. Pics and
forcep s b lad es sh ou ld n o t b e
p laced und er the retina to avoid
avulsion of th e RPE. Th e m em -
b ran e should b e rotated slig htly
to test for adherence to the retina
and to free up the m em brane.

than cutting across the fibers. This concept is similar to the retina away from the membrane and toward the RPE.
the making of incisions along Langer’s lines in facial plas- Suction should not be applied to the retinotomy because
tic surgery. Diathermy is not applied to the retinotomy site of the possibility of shearing damage to the photoreceptors
because bleeding is not an issue, as retinal vessels are never and RPE.
transected. Diathermy causes an unnecessary nerve fiber After the membrane is removed from the subreti-
layer defect and may stimulate scarring. nal space, it is retained in the grasp of the forceps and
The 25-gauge forceps are used to grasp the anterior used to gently push the retinotomy margins back together
surface of the outer margin of the membrane (Fig. 21.4). (Fig. 21.6) and express fluid from the subretinal space.
Forceps used with one blade under the membrane are more The membrane is then moved to the anterior vitreous
likely to strip the RPE or damage the choriocapillaris in the space and approximated to the port of the vitreous cutter.
authors’ opinion. Pics are never used because of the risk of The membrane is then removed with the vitreous cutter
RPE avulsion and bleeding from choriocapillaris trauma. (Fig. 21.7) using coaxial illumination from the operating
Grasping of the anterior surface of submacular membranes microscope. Dragging the membrane through the periph-
with end-opening forceps was developed to reduce damage eral vitreous and pars plana for removal with the forceps
to the RPE in an analogous manner to that developed for may create vitreoretinal traction and subsequent retinal
epiretinal membranes. The intraocular pressure (IOP) is ele- detachment. There is no need to submit the membrane to
vated to 60 mm Hg using the Constellation alternative infu- pathology, as the histologic appearance of these lesions is
sion feature, using another console air source, or increasing well known.
the bottle height prior to removing the membrane to reduce Fluid-air exchange is used for surface tension manage-
bleeding. The membrane is rotated slowly to determine if ment for the retinotomy site to eliminate the need for laser
there is excessive adherence to the retina or RPE. Exces- retinopexy. Laser retinopexy creates a scotoma from nerve
sive adherence is a common problem in patients with prior fiber layer damage and increases the chance of a new neovas-
photocoagulation, but much less frequent since anti-VEGF cular membrane at the laser site. The exchange is performed
therapy became available. Changing the direction of pull- using the vitreous cutter for fluid egress and the Landers
ing can usually resolve this problem, although 25-gauge fine or equivalent high minus lens for visualization in phakic
curved scissors are occasionally necessary to delaminate the or pseudophakic eyes. The Constellation or equivalent air
membrane from the retina or RPE. The membrane should be pump is used as an air source.
removed very slowly to reduce retinal tearing and to enable The IOP should be maintained at higher than normal
close observation of the retina and RPE to prevent damage levels during wound closure to prevent bleeding. Conjunc-
(Fig. 21.5). If the retina becomes more elevated during tival closure and subconjunctival antibiotics and steroids are
removal, the endoilluminator can be used to gently push used in the manner described elsewhere in the text.
CHAPTER 21 ■ Treatm ent of Choroidal Neovascular Mem branes 199

Fig ure 2 1 .5 ■ Rem ove the m em brane slowly to reduce retinal tearing and RPE avulsion. Scissors
can be used to sever the adherent areas if necessary.

Fig ure 2 1 .6 ■ Use the m em brane to gently reap proxim ate the retinotom y and exp ress SRF.
200 SECTION IV ■ Disease-Sp ecific Managem ent

Fig u re 2 1 . 7 ■ Re m o ve t h e
m em brane with the vitreous cut-
ter; d o n ot d rag in throug h the
p erip heral vitreous. Coaxial illu-
m ination from the m icroscop e is
sufficient for this step in the ante-
rior vitreous.

Macular Degeneration (ARMD) patients during submacular


RESULTS
surgery. It has been shown in numerous studies that RPE
removal will lead to choriocapillaris atrophy (36–41). While
Underlying, ongoing pathology, surgical damage to the RPE
partial RPE regeneration may occur in some areas, other areas
and retina, and recurrent membranes determine the outcome.
develop choriocapillaris atrophy and resultant disorganized
Careful case selection, as described previously, is essential in
photoreceptors. If one could insert new RPE cells during the
producing better outcomes. The membrane recurrence rate has
submacular surgical procedure, perhaps the inevitable atro-
been reported from 25% (the principal author’s series) to 45%.
phy could be prevented, or at least minimized.
An occasional patient will develop an extrafoveal recurrence
It is not difficult to imagine the inherent problems of
and benefit from laser photocoagulation. Reoperation is indi-
RPE transplantation. Complex issues include transplanted
cated for a well-defined subretinal membrane in patients who
RPE cell viability and function, the need for lifelong immune
had visual improvement after previous successful surgery.
suppression to prevent rejection, choriocapillaris viabil-
ity, and RPE cell adherence to Bruch’s membrane. For over
25 years, researchers have been investigating these and the
TRANSPLANTATION OF RETINAL many other complex issues regarding RPE transplantation.
PIGMENT EPITHELIUM News stories in the media have caused an excitement among
patients, and therefore it is very important for physicians to
Although tremendous advances have been made in the treat- be educated in order to counsel their patients effectively.
ment of retinal diseases, macular degeneration still results in In 1975, researchers discovered that autotransplanted
poor visual outcomes in most patients and there is no treat- RPE cells underwent metaplasia after their injection into the
ment for dry AMD. It has been postulated that a contributing vitreous cavity. The RPE cells first transformed into mac-
factor to the poor visual outcome following removal of CNV rophages and then later into spindle-shaped cells with col-
membranes in macular degeneration patients is atrophy of lagen production (42,43).
the subfoveal choriocapillaris (32). It has been reported that In 1989, physicians described a pars plana approach for
the area of atrophy can continue to enlarge 1 year after sur- the transplantation of autologous RPE cells from a periph-
gery. The stimulus for the choriocapillaris atrophy may be eral chorioretinal biopsy to prepare Bruch’s membrane at the
the failure of the RPE to repopulate the surgical bed (33). posterior pole of the same eye (44).
The extent of perfusion in the fovea is related to the visual In 1991, Peyman reported his technique for RPE trans-
prognosis and therefore of great importance (34,35). plantation in two patients with extensive subfoveal scar-
Unfortunately, tightly integrated RPE cells are removed ring secondary to macular degeneration (45). His technique
along with subfoveal neovascular membranes in Age Related involved the preparation of a large retinal flap encompassing
CHAPTER 21 ■ Treatm ent of Choroidal Neovascular Mem branes 201

the macula and the arcades, removal of the submacular scar, The latest development in the field of RPE transplantation
and replacement of the RPE cells using either an autologous involves the cotransplantation of intact sheets of fetal retina
pedicle graft or homologous RPE cells and Bruch’s mem- with RPE (52). Investigators at the University of Louisville
brane. One patient, who had undergone a pedicle graft, transplanted intact cografts into the subretinal space of RCS
had an improvement of visual acuity from count fingers to rats. After 6 to 7 weeks, transplanted photoreceptors, with
20/400 at 14 months. The other patient developed encap- the support of the cografted RPE cells, developed fully in
sulation of his homologous graft without any improvement organized, parallel layers in the subretinal space. They con-
in vision. cluded that such transplants have the potential to benefit
In 1992, scientists in Japan reported on the histology of retinal diseases with dysfunctional RPE and photoreceptors.
transplanted RPE cells in New Zealand White rabbits (46). A tremendous amount of RPE transplantation research
They found that by 1 week, the transplanted cells had formed has been conducted and continues to be undertaken. While
a monolayer. By 3 weeks, grafted RPE cells had formed api- this is an exciting area of research with tremendous potential
cal microvilli and tight junctions with adjacent cells. Their benefits, we must remember that currently this remains an
contact with Bruch’s membrane appeared to be composed of area of research, not treatment.
basal infoldings that were well formed. Their findings dem-
onstrated the functional appearance of the transplanted RPE
cells. The same year, a group of researchers reported that MACULAR TRANSLOCATION
RPE transplants stabilized retinal vasculature and prevented
neovascularization in the RCS (Royal College of Surgeons) Lindsey and Finklestein first reported macular transloca-
rat (47). Another study demonstrated that the transplanta- tion for the purpose of studying the relationship between
tion of normal RPE cells reversed pathological changes in the macula and the submacular RPE (53). The principal
the photoreceptors that had already occurred by the time of author developed the concept of macular translocation for
transplantation in the RCS rat (48). the treatment of submacular CNV. Both retinal rotation and
In 1994, a Swedish group led by Algvere published small flap translocation were developed and investigated in a
their results on RPE transplantation in patients with exu- Hanover pig model with the late Scott Langdon. It was deter-
dative ARMD performed in Sweden with RPE harvested by mined that there was a high incidence of retinal detachment
investigators from Columbia University (49). Fetal RPE was and proliferative vitreoretinopathy (PVR). This work was
placed beneath the neurosensory retina after the removal of reported at the Bascom Palmer Eye Institute Annual Alumni
submacular neovascularization in five patients with ARMD. meeting in 1987. The principal author stated at the meeting
The preoperative vision in all five patients was very poor. that this procedure should not be undertaken in humans
Surgical complications included cystoid macular edema and because of retinal detachment and PVR risk. Machemer, who
macular pucker. Microperimetry demonstrated that all five was present at the presentation, later reported performing
patients were able to fixate over the area of the RPE graft this work in humans. Subsequently, DeJuan, Tano, Toth,
immediately after surgery, but an absolute scotoma devel- Lewis, Eckardt, and others began clinical studies. All of
oped in this region within several months. There is no this work confirmed the original concerns of the principal
evidence that the transplanted RPE cells survived in the author, which were retinal detachment and PVR. In addition
subretinal space. It is noteworthy that these patients did not to retinal detachment and PVR, macular holes, new CNV
receive any immune suppression. membranes at the BSS injection sites, hemorrhage, cyclo-
While progress was being made in transplantation tech- version, diplopia, phthisis, multiple reoperations, macular
niques, the topic of rejection was also being studied. In 1997, folds, and decreased or unchanged vision in spite of macu-
Algvere’s group published another study comparing the fate lar translocation were reported (54–57). The authors do not
of fetal (13–20 weeks of gestational age) transplanted RPE believe that this procedure is indicated because of the high
cells in the subretinal space of five patients with fibrovascu- incidence of complications.
lar membranes with those transplanted in four patients with DeJuan developed a scleral resection method and later
atrophic ARMD (50). In patients with disciform lesions, all an imbrication method called “limited macular translocation”
grafts were rejected over a 6-month period. In patients with (58). Complications associated with this method include ret-
nonexudative disease, however, three out of four transplants inal detachment, PVR, hemorrhage, macular hole, new CNV,
showed little change in shape or size at 12 months. Visual phthisis, multiple reoperations, marked astigmatism, aniso-
acuity remained stable in these patients. The authors con- coria, diplopia, ptosis, enophthalmos, and failure to improve
cluded that human RPE allografts are not invariably rejected vision in spite of translocation. The authors do not believe
in the subretinal space and that an intact blood-retinal bar- that this method is indicated because of unacceptable com-
rier is likely to protect against rejection. More recent stud- plication rates. Hilel Lewis has developed an outpouching
ies demonstrate a slow but significant effect of the systemic method using clips that may prove to be more effective than
immune system in the subretinal space, and therefore sci- DeJuan’s technique of scleral imbrication, although most of
entists are cautioning investigators against considering the the same complications will not be reduced by this tech-
subretinal space to be immunologically privileged (51). nique change.
202 SECTION IV ■ Disease-Sp ecific Managem ent

After approximately a 2-year period of intense interest 17. Miller JW, Walsh AW, Kramer M, et al. Photodynamic therapy
and promotion of this technique, the vast majority of sur- of experimental choroidal neovascularization using lipoprotein-
delivered benzopophyrin. Arch Ophthalmol 1995;113:810–818.
geons have given up this procedure. In spite of extensive
18. Kramer M, Miller JW, Michaud N, et al. Liposomal benzopor-
early experimental work, the principal author has never phyrin derivative verteporfin photodynamic therapy: selective
elected to perform this procedure on humans. treatment of choroidal neovascularization in monkeys. Oph-
thalmology 1996;103:427–438.
19. Husain D, Miller JW, Michaud N, et al. Intravenous infusion of
References liposomal benzoporphin derivative for photodynamic therapy
of experimental choroidal neovascularization. Arch Ophthalmol
1. Michels S, Rosenfeld PJ, Puliafito CA, et al. Systemic bevaci- 1996;114:978–985.
zumab (Avastin) therapy for neovascular age-related macular 20. Weishaupt KR, Gomer CJ, Dougherty TJ. Identification of
degeneration twelve-week results of an uncontrolled open-label singlet oxygen as the cytotoxic agent in photo-inactivation of
clinical study. Ophthalmology 2005 Jun;112(6):1035–1047. murine tumor. Cancer Res 1976;36:2326–2329.
2. Avery RL, Pieramici DJ, Rabena MD, et al. Intravitreal bevaci- 21. Steinhorst UH, Theischen M, Winter R. Subretinal lavage: a
zumab (Avastin) for neovascular age-related macular degen- technique of continuous irrigation for removal of traumatic sub-
eration. Ophthalmology 2006 Mar;113(3):363–372. e5. [Epub macular hemorrhage. Ophthalmologica 1997;211(6):399–401.
2006 Feb 3.] 22. Capone A. Submacular surgical procedures. Int Ophthalmol
3. Macular Photocoagulation Study Group. Laser photocoagula- Clin 1995;35(4):83–93.
tion of subfoveal neovascular lesions in age-related macular 23. Ibanez HE, Williams DF, Thomas MA, et al. Surgical man-
degeneration: results of a randomized clinical trial. Arch Oph- agement of submacular hemorrhage. Arch Ophthalmol
thalmol 1991;109:1220–1231. 1995;113(1):62–69.
4. Klein BE, Brady WE, Palta M. Association between antioxidant 24. Humayun M, Lewis H, Flynn HW, et al. Management of sub-
and zinc intake and the 5-year incidence of early age-related macular hemorrhage associated with retinal arterial macroan-
maculopathy in the Beaver Dam Eye Study. Am J Epidemiol eurysms. Am J Ophthalmol 1998;126(3):358–361.
1998;148(2):204–214. 25. Claes C, Zivojnovic R. Efficacy of tissue plasminogen activator
5. Klein R, Klein BE, Moss SE. The Beaver Dam Eye Study. Rela- in subretinal hemorrhage removal. Bull Soc Belge Ophthalmol
tionship of smoking to the incidence of age-related maculopa- 1996;261:115–118.
thy. Am J Epidemiol 1998;147(2):103–110. 26. Hawkins WR. Intraocular fibrinolysis of submacular hem-
6. De Luca LM, Ross SA. Beta-carotene increases lung cancer inci- orrhage with tPA and surgical drainage. Am J Ophthalmol
dence in cigarette smokers. Nutr Rev 1996;54(6):178–180. 1994;118(5):559–568.
7. Figueroa MS, Regueras A, Bertrand J. Laser photocoagulation to 27. Lim JI, Drews-Botsch C, Sternberg P, et al. Submacular hemor-
treat macular soft drusen in ARMD. Retina 1994;14(5):391–396. rhage removal. Ophthalmology 1995;102(9):1393–1399.
8. Wetzig PC. Photocoagulation of drusen-related macular degen- 28. Chaudhry NA, Mieler WF, Han DP, et al. Preoperative use of
eration: a long-term outcome. Trans Am Ophthalmol Soc 1994;92: tissue plasminogen activator for large submacular hemorrhage.
299–303. Ophthalmic Surg Lasers 1999;30(3):176–180.
9. Olk RJ, Friberg TR, Stickney KL, et al. Theraputic benefits of 29. Johnson MW. Pneumatic displacement of submacular hemor-
infared (810 nm) diode laser macular grid photocoagulation in rhage. Curr Opin Ophthalmol 2000;11(3):201–206.
prophylactic treatment of nonexudative ARMD. Ophthalmology 30. Kamei M, Misono K, Lewis H. A study of the ability of tPA to
1999;106(11):2082–2090. diffuse into the subretinal space after intravitreal injections in
10. The Choroidal Neovascularization Prevention Trial Research rabbits. Am J Ophthalmol 1999;128(6):739–746.
Group. Choroidal neovascularization in the Choroidal Neo- 31. Thomas MA, Kaplan HJ. Surgical removal of sub-foveal neo-
vascularization Prevention Trial. Ophthalmology 1998;105(8): vascularization in POHS. Am J Ophthalmol1991;111:1–7.
1364–1372. 32. Castellarin A, Nasir M, Sugino IK, et al. Decreased chorio-
11. The Choroidal Neovascularization Prevention Trial Research capillaris perfusion following surgical excision of choroidal
Group. Laser treatment in eyes with large drusen. Short term neovascular membranes in age-related macular degeneration.
effects seen in a pilot randomized clinical trial. Ophthalmology Br J Ophthalmol 1997;81:481–489.
1998;105(1):11–23. 33. Castellarin A, Nasir M, Sugino IK, et al. Progressive choriocap-
12. Treatment of Age-related Macular Degeneration With Photo- illaris atrophy after surgery for age-related macular degenera-
dynamic Therapy (TAP) Study Group. Photodynamic ther- tion. Retina 1998;18:143–149.
apy of subfoveal choroidal neovascularization in age-related 34. Akduman L, Del Priore LV, Desai VN, et al. Perfusion of the
macular degeneration with verteporfin. Arch Ophthalmol subfoveal choriocapillaris affects visual recovery after submac-
1999;117:1329–1345. ular surgery in the presumed ocular histoplasmosis syndrome.
13. Schmidt-Erfurth U, Hasan T, Gragoudas E, et al. Vascular tar- Am J Ophthalmol 1997;123:90–96.
geting in photodynamic occlusion of subretinal vessels. Oph- 35. Pollack JS, Del Priore LV, Smith ME, et al. Postoperative abnor-
thalmology. 1994;101:1953–1961. malities of the choriocapillaris in exudative age-related macu-
14. Allison BA, Waterfield E, Richter AM, et al. The effects of lar degneration. Br J Ophthalmol 1996;80:314–318.
plasma lipoproteins on in vitro tumor cell killing and in vivo 36. Korte GE, Reppucci V, Henkind P. RPE destruction causes
tumor photosensitization with benzoporphyrin derivative. choriocapillaris atrophy. Invest Ophthal Vis Sci 1984;25:1135–
Photochem Photobiol 1991;54:709–715. 1145.
15. Schmidt-Erfurth U, Hasan T, Schomacker K, et al. In vivo 37. Kuwabara T, Ishikawa Y, Kaiser-Kupfer MI. Experimental
uptake of liposomal bezoporphyrin derivative and photo- model of gyrate atrophy in animals. Ophthalmology 1981;88:
thrombosis in experimental corneal neovascularization. Lasers 331–334.
Surg Med 1995;17:178–188. 38. Takeuchi M, Itagaki T, Takahashi K, et al. Changes in the inter-
16. Young LH, Howard MA, Hu LK, et al. Photodynamic therapy of mediate stage of retinal degeneration after intravitreal injection
pigmented choroidal melanomas using a liposomal preparation of of ornithine. Nippon Ganka Gakkai Zasshi. Acta Soc Ophthalmol
bezoporphyrin derivative. Arch Ophthalmol 1996;114:186–192. Jpn (Tokyo) 1993;97:17–28.
CHAPTER 21 ■ Treatm ent of Choroidal Neovascular Mem branes 203

39. Valentino A, Kaplan HJ, Del Priore LV, et al. Retinal pigment 49. Algvere PV, Berglin L, Gouras P, et al. Transplantation of fetal
epithelium repopulation in monkeys after submacular surgery. retinal pigment epithelium in age-related macular degenera-
Arch Ophthalmol 1995;113:932–938. tion with subfoveal neovascularization. Graefes Arch Clin Exp
40. Del Priore LV, Kaplan HJ, Hornbeck R, et al. Retinal pigment Ophthalmol 1994;232(12):707–716.
epithelium debridement as a model for the pathogenesis 50. Algvere PV, Berglin L, Gouras P, et al. Transplantation of RPE
and treatment of macular degeneration. Am J Ophthalmol in age-related macular degeneration: observations in disciform
1996;122:629–643. lesions and dry RPE atrophy. Graefes Arch Clin Exp Ophthalmol
41. Del Priore LV, Hornbeck K, Kaplan HJ. Debridement of the 1997;235(3):149–158.
pig retinal epithelium in vivo. Arch Ophthalmol 1995;113: 51. Zhang X, Bok D. Transplantation of retinal pigment epithelial
939–944. cells and immune response in the subretinal space. Invest Oph-
42. Mueller-Jensen K, Machemer R, Azarnia R. Autotransplanta- thalmol Vis Sci 1998;39(6):1021–1027.
tion of retinal pigment epithelium in intravitreal diffusion 52. Aramant RB, Seiler MJ, Ball SL. Successful cotransplantation
chamber. Am J Ophthalmol 1975;80:530–537. of intact sheets of fetal retina with retinal pigment epithelium.
43. Mueller-Jensen K, Mandelcorn MS. Membrane formation by Invest Ophthalmol Vis Sci 1999;40(7):1557–1564.
autotranplanted retinal pigment epithelium. Mod Probl Oph- 53. Bressler NM, Finklestein D, Sunness JS, et al. Retinal pigment
thalmol 1975;15:228–234. epithelial tears through the fovea with preservation of good
44. Lane C, Boulton M, Marshall J. Transplantation of retinal pigment visual acuity. Arch Ophthalmol 1990;108(12):1694–1697.
epithelium using a pars plana approach. Eye 1989;3:27–32. 54. American Academy of Ophthalmology. Macular translocation.
45. Peyman GA, Blinder KJ, Paris CL, et al. A technique for reti- Ophthalmology 2000;107(5):1015–1018.
nal pigment epithelium for age related macular degeneration 55. Lewis H, Kaiser PK, Lewis S, et al. Macular translocation for
secondary to extensive subfoveal scarring. Ophthalmol Surg subfoveal choroidal neovascularization in ARMD: a prospec-
1991;22(2):102–108. tive study. Am J Ophthalmol 1999;128(2):135–146.
46. Yamaguchi K, Yamaguchi K, Young RW, et al. Vitreoretinal sur- 56. Ohji M, Fujikado T, Saito Y, et al. Foveal translocation:
gical technique for transplanting retinal pigment epithelium in a comparison of two techniques. Semin Ophthalmol 1998;
rabbit retina. Jpn J Ophthalmol 1992;36(2):142–150. 13(1):52–62.
47. Seaton AD, Turner JE. RPE transplants stabilize retinal vascula- 57. Ninomiya Y, Lewis JM, Hasegawa T, et al. Retinotomy and
ture and prevent neovascularization in the RCS rat. Invest Oph- foveal translocation for surgical management of subfoveal chor-
thalmol Vis Sci 1992;33(1):83–91. oidal neovascular membranes. Am J Ophthalmol 1996;122(5):
48. Lavail MM, Li L, Turner JE, et al. Retinal pigment epithelial 613–621.
cell transplantation in RCS rats: normal metabolism in rescued 58. DeJuan E, Vander JF. Effective macular translocation without
photoreceptors. Exp Eye Res 1992;55(4):555–562. scleral imbrication. Am J Ophthalmol 1999;128(3):380–382.
C H AP TER
22
INTRAVITREAL
INJECTION TECHNIQUE

Never inject if blepharitis or conjunctivitis is Betadine should be used for all patients because the
present; external infections should be treated risk of endophthalmitis is greatly reduced. The risk of topi-
intensively for several days before injecting and cal allergic reactions is exaggerated; an allergy to shellfish
the patient examined at the slit lamp before is not sufficient to avoid povidone iodine, nor is mild skin
injecting to verify eradication of the infection. irritation. It is important to use sterile saline (EyeStream) to
Never stop anticoagulants before intravitreal injec- wash off lids and lashes after the procedure.
tions or vitrectomy; the risk of bleeding is exception- A sterile, bladed speculum must be used to expose the
ally low, bleeding is rare and minimal, and the risk of injection site as well as contain the lashes. The patient must
thromboembolic events is increased. Many patients as be supine at elbow height for the surgeon to maximize inject-
well as their medical doctors and nurses stop anticoagu- ing ophthalmologist ergonomics. Measure 3.5 mm posterior
lants before injections and surgery even though they were to the limbus using the Luer Lok on a sterile tuberculin
not instructed to do so, increasing thromboembolic risk, syringe without the needle. The tip of the index finger on
potentially creating a scenario in which an anti–vascular the opposite hand should be placed against the needle hub
endothelial growth factor compound is wrongly blamed for and used to steer the 30 or 32 gauge needle and maintain a
a stroke or myocardial infarction (1). trajectory perpendicular to a tangent plane at the injection
The technician and injecting ophthalmologist should point. The patient should be given a fixation point on the
wear a mask because of the proximity of the nose and mouth ceiling, the room should be silent, and no one should move
bacteria to the needle when adjusting the quantity of drug in or enter or leave the room during the injection to eliminate
the tuberculin syringe. Gloves should be worn by the techni- stimuli for saccades.
cian and injecting ophthalmologist largely for patient percep- Indirect ophthalmoscopy should be performed after the
tion. Sterile gloves are also advantageous if the sterile speculum injection to verify circulation rather than intraocular pres-
is manipulated to improve access to the injection site. sure measurement because of the risk of contamination. The
Subconjunctival lidocaine is not required; the authors authors use topical antibiotics after injection (Vigamox q2h
believe that perforation risk and the pain of anesthetic on the day of injection and qid for 3 days) but no pretreat-
injection exceed the value. Lidocaine jelly (2%) in prefilled ment with topical antibiotics.
syringes should be applied before 5% povidone iodine
(Betadine) both to improve patient comfort and because
Reference
multiuse packaging could theoretically become contaminated.
The povidone iodine should be applied to lashes, lid mar- 1. Charles S, Rosenfeld PJ, Gayer S. Medical consequences of stop-
gins, injection site, and cul-de-sac. Povidone iodine swabs ping anticoagulant therapy before intraocular surgery or intravit-
real injections. Retina. 2007;27(7):813-815.
are used on both upper and lower lids.

204
C H AP TER
23
TRAUMA

Vitreous surgery techniques have greatly The vitreous has been referred to as a “scaffold” (2,3), but
improved the management of ocular trauma. this is a substrate. Substrate better describes the manner in
Work-related activity, home maintenance, auto- which cellular migration and proliferation occur on the vit-
mobile and motorcycle accidents, fireworks, reous collagen matrix. More accurately, it is critical to rec-
hunting, violence, and recreational activity create a contin- ognize that proliferation usually occurs along the anterior
ued threat of severe ocular injury. The vast range of objects vitreous cortex (AVC) and posterior vitreous cortex (PVC),
and velocities implicated in ocular trauma lead to the wide although a foreign object can make a path through the vit-
spectrum of injuries seen (1). In this chapter, the emphasis reous along which apparent transvitreal proliferation can
will be placed on several stereotypic subgroups. occur. As the retina is an ideal substrate for proliferation,
it is not necessary to implicate the vitreous when epiretinal
cellular migration and proliferation occur.
WOUND-RELATED CELLULAR
MIGRATION/ PROLIFERATION
TIMING OF VITRECTOMY
Any interruption of tissue results in proliferation of the inter-
rupted cell groups. Planar cell groups respond to loss of contact Immediate vitrectomy in penetrating ocular trauma cases
inhibition by a migration/proliferation of the cells adjacent to should be avoided unless certain types of intraocular for-
the interruption. Misalignment of the proliferating plane may eign bodies (IOFBs) are present. The experience of the Iraq
result in reduplication of the original tissue layer. Migration/ war has shown that primary closure of entry wounds with
proliferation ceases when contact inhibition is restored by con- delayed removal of the IOFB can have good outcomes (4).
tinuity of the new cell group with similar cells representing the Arterial bleeding, choroidal swelling, leaking wounds, striate
margin of defect. This new structure can be called a membrane; keratopathy, corneal edema, and lack of preparation add to
however, it is actually a reparative extension of previously nor- the difficulty of immediate vitrectomy (5,6). More impor-
mal tissues. The widely used term “fibrovascular ingrowth” tantly, penetrating trauma frequently occurs in the young
implies that wound-related cellular proliferation originates from patient not having a posterior vitreous detachment (PVD).
extraocular tissues. The severe proliferation that occurs in blunt Without adequate vitreoretinal separation, iatrogenic retinal
trauma after choroidal rupture (retinitis sclopetaria) is clinically breaks, difficult surgery, and postoperative contraction of
indistinguishable from “fibrovascular ingrowth.” There is little residual vitreous can ensue.
direct evidence of extraocular origin for the majority of cases Typically, the hemorrhage and inflammation that accom-
of wound-related cellular proliferation. Because the reparative pany trauma induce a PVD in 7 to 14 days, allowing safer,
mechanism stems from tissue disruption and destruction, the more effective vitreous surgery. Cellular proliferation starts at
additional damage of retinopexy should be avoided unless nec- 10 to 14 days, making this the ideal time to intervene (7,8). If
essary for the treatment of a retinal defect. Silicone oil can be the retina can be seen, these cases can be watched at weekly
used for what the authors term “retinopexy avoidance” in the intervals and vitrectomy avoided if cellular proliferation
setting of severe trauma with retinal breaks. does not occur. Cases with opaque media require vitrectomy
at this 10- to 14-day point because further delay could lead
to cellular proliferation, which is initially difficult to deter-
SUBSTRATES FOR PROLIFERATION mine with B-scan ultrasound. Decreased vitreous mobility
observed on ultrasound examination indicates hypocel-
Cellular proliferation occurs on preexisting support sub- lular vitreous collagen contraction and/or early cellular
strates such as the cornea, retina, vitreous, lens, and iris. proliferation.
205
206 SECTION IV ■ Disease-Sp ecific Managem ent

INTRAOCULAR FOREIGN BODIES Tim in g o f Surg e ry


All large, toxic, biologic, or sharp IOFBs should be removed as
Vitreous surgery allows excellent visualization; prevention soon as the patient can be safely taken to the operating room.
of postoperative transvitreal proliferation; and removal of This approach decreases secondary mechanical trauma, rapid
blood, lens materials, and organisms if present. Bronson toxicity, and endophthalmitis. Plastic, glass, and lead shotgun
or giant magnet foreign body removal has virtually disap- pellets can be observed until vitrectomy is indicated for other
peared because of the widespread availability of vitreous reasons. Although late-night and weekend vitreous surgery
surgery training, technology, techniques, and the Machemer- creates logistical and cost problems, it decreases the risk of
Parel diamond-coated IOFB forceps (Fig. 23.1). Intraocular endophthalmitis and toxic damage and should be undertaken
magnets can be used to pick up foreign bodies for transfer if possible.
to diamond-coated forceps and removal, although this is
seldom necessary (9–15).
Surg ical Se q ue n ce an d Te ch n iq ue s

In e rt Ve rsus To xic Fo re ig n Bo d ie s Wound Repa ir

Most iron- and copper-containing foreign bodies should Any visible corneal or scleral wound should be sutured
be removed immediately (16,17). Stainless steel, alumi- before proceeding with the vitrectomy. Running shoelace
num, and lead foreign bodies are much less toxic and monofilament nylon sutures distribute stress evenly, are
can be tolerated in selected cases. Indication for removal elastic and well tolerated, and may be rapidly placed. Silk
of these less toxic materials must be individualized on a sutures are inelastic and lead to wound leaks during the vit-
clinical basis. Occasionally, iron- and copper-containing rectomy, while absorbable sutures are inelastic and not per-
foreign bodies will be overlooked and only discovered manent. Interrupted sutures can cause striate keratopathy
after they have become encapsulated. If no evidence of and take longer to place. Small sutures (10-0) are used for
toxicity is seen in these cases, frequent follow-up should central cornea, 9-0 for midcornea, and 8-0 for peripheral
be undertaken rather than surgery. Clinical examination cornea or sclera.
of adjacent structures (cornea, iris, and lens) is probably Surgical judgment should guide the decision-making
a better indication of toxicity than electroretinography. process concerning excision of prolapsed tissue. Viable-
Plastic materials can be left in place in selected cases. appearing iris or ciliary body in a very recent injury can
Submacular foreign bodies and those embedded in the be irrigated and repositioned, while any sign of infection
optic nerve should be managed on an individual basis or tissue destruction indicates excision. Retinopexy should
because of the extreme hazards of removal. Any exog- not be used anterior to the muscle ring (ora serrata) and
enous biologic material such as vegetable matter should should be applied only to definite retinal breaks located
be removed immediately because of the risk of infection posteriorly. Excessive and unnecessary retinopexy leads to
and inflammation. Endogenous cilia and bone fragments greater wound-related cellular proliferation and inflamma-
are usually well tolerated and need not be removed unless tion. Endolaser retinopexy should be applied only to specific
vitrectomy is performed for other reasons or they appear breaks identified during vitrectomy.
to be the cause of inflammation. Exploration of/for a posterior wound should be done
only if pressure on the globe can be completely avoided.
The vitreous and retina can be prolapsed from a posterior
wound by surgical manipulation. If vitrectomy is completed
first, the location of the wound will be known, and the eye
can be softened and filled with air (gas) before proceeding
with posterior wound repair in the rare instance that it is
thought to be necessary. Most posterior wounds are self-
sealing, and wound closure does not decrease the incidence
of wound-related cellular proliferation. Retinopexy increases
the incidence of wound-related cellular proliferation and
should be avoided.

Conjunctiva l Incisions
The authors currently use 25-gauge sutureless vitrectomy for
all trauma cases and open the conjunctiva selectively only
to repair scleral wounds and to construct a one clock-hour
Fig ure 2 3 .1 ■ Diam ond -coated foreign body forceps prevent incision when a superotemporal 25-gauge wound is enlarged
dropping and grasp-induced m ovem ent of the IOFB. to 20 gauge or larger for IOFB removal.
CHAPTER 23 ■ Traum a 207

Sclerotomies it is tempting to remove the foreign body as soon as it is seen,


it is far better to complete the vitrectomy first. A big advan-
The incision for the vitrectomy instrument, infusion cannula, tage of the vitrectomy approach is the avoidance of removal-
and endoilluminator should be made in the usual position, induced vitreous traction. The need for a relatively complete
3 mm posterior to the limbus if the lens is to be removed vitrectomy before a foreign body removal is absolute.
or 4 mm posterior to the limbus if not. Extra care must be
taken to avoid choroidal infusion in trauma cases because
hypotony and choroidal edema are frequently present. Foreign Body Remova l
The incision for the foreign body removal should usu- It is not recommended that an extraocular magnet be used
ally be the superotemporal vitrectomy instrument incision, in conjunction with vitrectomy; the advantage of a slow,
enlarged after the vitreous is removed using a diamond or controlled removal without vitreous traction would be lost.
disposable knife. Some surgeons have recommended intraocular magnets,
Lens Remova l but the authors have not used these devices because of the
availability of the diamond-coated forceps. When all the vit-
If the lens is clear, it should be allowed to remain unless a reous around the IOFB has been removed, the superotempo-
very large foreign body requires translimbal removal. Many ral incision is enlarged to accommodate the largest outside
small, localized traumatic cataracts do not progress and diameter of the diamond IOFB forceps.
therefore may not require removal. The vitrectomy instru-
ment should be used for anterior vitrectomy if any vitreous Moderately Sized Foreign Bodies
is in the anterior chamber or capsular bag. The aspirating After the foreign body is grasped in the forceps and
fragmenter is faster than vitrectomy instruments for lensec- brought to the anterior vitreous area, its size can be better
tomy but should never be applied to the vitreous. If vitreous assessed. If the foreign body will not fit safely through the
enters the capsular bag, it should be removed with a vit- wound, but is not larger than 6 mm in its smallest diameter,
rectomy instrument and the lensectomy completed with the the scleral wound should be enlarged with a knife (Fig. 23.2).
fragmenter. In most cases, the capsule should be removed It is best to plug the other sclerotomy so that the surgeon’s
with the diamond-coated or end-grasping forceps. hand is free to enlarge the wound. A knife at the opposite
end of the circumferential sclerotomy from the shank of the
Vitrectomy
forceps should be used to extend the sclerotomy while stabi-
The vitreous may be clear or have significant hemorrhage in lizing the other end of the wound with the shank of the for-
acute IOFB cases, but in either instance, it must be completely ceps. Using this approach, it is not necessary to release the
removed. The first goal is to remove enough vitreous to grasp of the foreign body, and a large, leaky incision need not
remove all vitreous attachments to the foreign body. Although be present while searching for and picking up the IOFB.

Fig u re 2 3 .2 ■ The free h and


should use a d iam ond or d isp os-
ab le knife to enlarg e sclerotom y
for foreign bodies too large to go
through 20-gauge m icrovitreoret-
inal (MVR) incision. This should be
d on e after g rasp in g th e foreig n
body and bringing it near the pars
plana to avoid a wound leak while
grasping the IOFB.
208 SECTION IV ■ Disease-Sp ecific Managem ent

Fig u re 2 3 .3 ■ A secon d p air


of forcep s is used to reg rasp a
long, thin IOFB so that it can be
rem oved along its long axis.

Cylindrical Foreign Bodies Large Foreign Bodies


Long, cylindrical, small-diameter foreign bodies such as If the foreign body is brought to the anterior vitreous
wire frequently lie tangential to the retinal surface. The initial and appears too large to be removed through the pars plana,
pickup of the foreign body will then result in the IOFB being translimbal removal should be performed (Fig. 23.4). The
nearly perpendicular to the shank of the forceps. If it were grasp on the foreign body need not be lost as the lens is
removed with this orientation, the scleral opening would have removed with the aspirating fragmenter in the surgeon’s other
to be unnecessarily large. It is therefore best to use a second hand. A cataract-type limbal section is then initiated with a
forceps to bimanually transfer or regrasp the foreign body to blade, again in the surgeon’s other hand. The infusion system
permit removal along the long axis of the IOFB (Fig. 23.3). prevents hypotony during the initial incision but will cause

Fig ure 2 3 .4 ■ Very larg e IOFBs


sh o u ld b e re m o ved t h ro u g h
the lim b us after vitrectom y and
len sectom y, g rasp in g th e IOFB
an d b rin g in g it in to th e an te-
rior cham b er without d ang erous
regrasping.
CHAPTER 23 ■ Traum a 209

Fig u re 2 3 . 5 ■ En cap su lat ed


IO FBs sh o u ld b e rem o ved b y
perform ing com plete vitrectom y,
then incising the capsule with an
MVR blade.

iris prolapse as the wound is enlarged. If the pupil is large and Encapsulated Foreign Bodies
iridectomy is not required, the infusion should be turned off The fibrous capsule must be incised to safely remove
to avoid iris prolapse. If an iridectomy is required, the infu- an encapsulated IOFB (Fig. 23.5). The vitrectomy should be
sion-induced iris prolapse can be used to the surgeon’s advan- completed first and all vitreous traction to the foreign body
tage, permitting ab-externo iridectomy with one hand. When site severed. The capsular incision should be performed with
the incision is large enough, the IOFB should be passed retro- 20-gauge scissors (Fig. 23.6). A cruciate incision will allow
grade through the wound and removed with a second pair of the IOFB to be completely free before the IOFB forceps are
forceps through the limbal wound. The wound should then used to remove the fragment (Figs. 23.7 and 23.8). Stan-
be closed with a running shoelace, 8-0 nylon suture. dard removal techniques as described previously are used,

Fig u re 2 3 .6 ■ The top of the


cap sule should be rem oved with
the scissors.
210 SECTION IV ■ Disease-Sp ecific Managem ent

Fig ure 2 3 .7 ■ The foreign body


should b e g ently m oved to free
up adhesions.

depending on the size of the foreign body. Subretinal lOFBs people, it may not be possible to remove a portion of the
are removed with diamond-coated forceps (Fig. 23.9). PVC on the macula and optic nerve; however, all connec-
After the IOFB has been removed, the wound should be tions between these areas and to the peripheral retina must
closed with a running shoelace, 8-0 nylon suture, leaving a be severed. Theoretically, it would be beneficial to remove
20-gauge–size opening around a scleral plug. the entire PVC, but this is not possible in many young people
The vitrectomy should then be completed, removing without damaging the optic nerve or retina. Any subsequent
any fibrin, capsular material, or hemosiderin left in the cellular proliferation along these remaining portions of PVC
residual vitreous or at the IOFB site. In acute cases in young can be managed, if necessary, at a later date.

Fig ure 2 3 .8 ■ The IOFB is


rem oved with diam ond-coated
forceps after m ob ilization.
CHAPTER 23 ■ Traum a 211

Fig ure 2 3 .9 ■ Subretinal IOFBs


are rem o ve d wit h d iam o n d -
coated forcep s throug h a retinal
break or retinotom y.

Subretinal surgery is occasionally required in late trauma The purpose of postoperative surface tension management
cases, but the majority of these membranes are highly vascu- is to prevent aqueous humor from passing through nonvisi-
lar and should be left in place. ble retinal defects, new traction-induced breaks, and defects
that are intentionally untreated. Retinopexy avoidance is
an excellent approach for the prevention of proliferative
Retinopexy
vitreoretinopathy (PVR) and fibrovascular proliferation.
Laser retinopexy should only be performed if a definite reti-
nal break is seen, not for prophylaxis along trauma contact
Sclera l Buckling
and wound sites. If the break is located contiguous with
the optic nerve, papillomacular bundle, or macula, it is not The authors find no benefit in placing buckle elements for
treated because breaks in these areas almost never result in trauma cases, either for primary repair or for secondary
detachment, and retinopexy in these areas can decrease cen- retinal detachments. These complex cases are usually best
tral vision. repaired with vitrectomy.
Segmental buckling can be used for peripheral
detachments/breaks but is unnecessary for posterior
Surfa ce Tension Ma na gement
detachments/breaks. Circumferential segmental explants
Internal fluid-air exchange and subsequent air-gas exchange are preferable to radial buckles in all instances. The
should be used if a definite retinal break is present. Inter- authors no longer buckle trauma eyes. Vitreous traction
nal drainage of subretinal fluid (SRF) should be performed from initial IOFB impact, egress of the foreign body, or
simultaneously with internal fluid-air exchange, if detach- vitreous removal creates a relatively high incidence of
ment exists. Postreattachment endolaser retinopexy can then dialyses and aphakic-like retinal breaks. These can be dif-
be used to treat the break(s). ficult to recognize at the time of surgery, thus emphasizing
Although perfluorocarbon (PFC) liquids have been rec- the need for a careful search and air-silicone exchange in
ommended for IOFB removal and management of coexisting difficult cases. Late traction from vitreous incarceration in
retinal detachment, the authors have not found these agents the sclerotomies plays a role in postoperative detachment
to be necessary in most cases. as well.
Air-gas exchange with SF6 after postreattachment endo-
laser retinopexy should be used for small to moderate retinal
Antibiotic-Steroid Thera py
defects.
Large retinal defects or especially severe trauma should Subconjunctival antibiotic therapy against both Gram-
be managed by air-silicone exchange without retinopexy. positive and Gram-negative organisms should be utilized.
212 SECTION IV ■ Disease-Sp ecific Managem ent

Subconjunctival repository steroids such as triamcinolone posterior and the anterior retina and vitreous are normal.
should be used to decrease postoperative fibrin formation An opening should then be made through the PVC nasally
and scarring. or in any area known to have attached retina. Vacuum
cleaning–extrusion may be necessary through this initial
opening to remove free blood products in the sub-PVD
DOUBLE PENETRATING INJURY space. When an adequate view of the retina is obtained,
vitrectomy can proceed by enlarging this opening in a
High-velocity objects from shotgun and explosive injuries circumferential fashion. If retinal detachment is present,
typically enter the eye anteriorly and exit posteriorly, creat- extremely low suction force should be used. If a retinal
ing two ocular wounds (18). Low-velocity injuries such as break is seen, vitrectomy should be continued with inter-
hammering metal-on-metal typically cause IOFBs (19–25). mittent drainage of SRF. Layer-by-layer removal of the vit-
reous increases the chance of retinal breaks and is more
In it ial Re p air time-consuming than the full-thickness, circumferential
method described previously.
Any corneal or anterior scleral wound should be closed as All vitreous attached to the exit wound should be
soon as the patient is seen, and any visible wound should be removed if it can be accessed without further damage to
closed before attempting further exploration. Great care must the retina. The vitreous applied to the surface of the wound
be taken to avoid tissue prolapse from surgical manipula- should be allowed to remain because its removal can create
tion. Running shoelace monofilament nylon sutures should bleeding, retinal breaks, wound leaks, and enhancement of
be utilized as described above. Posterior exploration is rarely cellular proliferation (Fig. 23.10). At the 14-day point, it is
indicated and should be done only if it can be done without extremely unusual for an exit wound to leak. As in vitrec-
exerting any pressure on the globe. tomy for diabetes, the goal of cone truncation is to eliminate
the anteroposterior traction, which is the critical element in
Tim in g o f Vit re ct o m y successful management of these cases.

Vitrectomy should be performed between 7 and 14 days


unless angle closure from lens swelling or endophthalmi- Surfa ce Tension Ma na gement
tis is an issue. Ultrasonic evidence of retinal detachment Fluid-air exchange is utilized as in other clinical situations
does not necessarily indicate early vitrectomy. Delay for 7 to when a retinal break is present. Internal drainage of SRF fol-
14 days permits PVD to occur, decreases choroidal swelling, lowed by fluid-air exchange and completion of SRF drain-
decreases bleeding, and provides better corneal clarity. age should be performed if detachment is present. Air can
also be utilized to confine bleeding and to help seal leaky
Surg ical Se q ue n ce an d Te ch n iq ue s wounds by a surface tension effect. The use of PFC liquids,
gas, and silicone is described earlier.
Lens Ma na gement
Clear lenses or small, localized cataracts should be allowed to Retinopexy
remain. Any central, posterior subcapsular opacity warrants
lensectomy because this will almost invariably progress after Laser endophotocoagulation is used only if retinal breaks
vitrectomy. Wounds at the ciliary body–pars plana level per- are apparent. Most posterior exit wounds do not give rise to
mit wound-related cellular proliferation to proceed along the retinal detachment, and retinopexy serves only to increase
AVC-lens interface, creating a cyclitic membrane. Although wound-related cellular proliferation.
prophylactic lens removal is not indicated, these cases must
be watched weekly postoperatively for any evidence of cyclitic Sclera l Buckling
membrane growth. The endocapsular fragmenter lensectomy
technique should be utilized, with care taken to avoid vitre- The authors no longer use prophylactic encircling bands.
ous in the fragmenter. The vitrectomy instrument should be This change was made because buckles increase operating
used to remove any vitreous in the anterior chamber or cap- time, postoperative pain, refractive error, strabismus, ptosis,
sular bag. Diamond-coated or end-grasping forceps should be and cost and may develop late intrusion into the sclera.
used for capsular removal. Iris surgery should be performed
only if absolutely necessary to see posteriorly; overzealous
removal creates postoperative inflammation and glare. CORNEAL-SCLERAL LACERATION

The great variability of corneal-scleral lacerations makes


Vitrectomy
generalization difficult, but certain principles merit discus-
The AVC should be removed to decrease the chance of sion. Microsurgery has advanced the success rate in these
cyclitic membrane formation unless the trauma is very cases, as has vitrectomy technology.
CHAPTER 23 ■ Traum a 213

Fig u re 2 3 .1 0 ■ Con e trun ca-


tion (rem oval of PVC) elim inates
the substrate for transvitreal p ro-
liferation and reduces the chances
of retinal detachm ent.

Wo un d Re p air cases must be observed very frequently for evidence of vitre-


ous “orientation” and loss of vitreous mobility. Taut vitreous
As discussed previously, running monofilament sutures are
with stress lines directed toward a wound is associated with a
best for closing corneal-scleral lacerations. Running sutures
high incidence of retinal detachment. If this condition coexists
distribute stress evenly, stimulate less vascularization, leak
with opaque media, frequent ultrasound examinations, includ-
less, and are placed rapidly. All knots should be buried if
ing the periphery, are necessary to rule out retinal detachment.
interrupted sutures are used.
In virtually all instances of taut vitreous with hypocellular gel
The scleral portion should be closed by sequential sutur-
contraction, vitrectomy will be necessary. If the vitreous is clear,
ing, exploration, and further suturing. Unless extremely large
the retina must be observed frequently for signs of detachment
pieces of sclera are absent, primary closure is preferable to
and/or cellular proliferation. True vitreous “organization” is a
scleral grafting. As these cases frequently develop wound-
late sign and should not be allowed to occur.
related cellular proliferation, scleral resection-like shortening
that occurs from primary closure of a defect can be beneficial.
COMPLICATIONS
Tim in g o f Le n s Re m o val
In fe ct io n
Many factors relate to the question of whether lens removal
should occur at the time of primary surgical repair or later. Immediate surgical repair with subconjunctival antibiotic
Hypotony-induced choroidal edema, striate keratopathy, prophylaxis and removal of all necrotic exteriorized tissue
and miosis make safe lens removal difficult at the time of leads to a surprisingly low incidence of endophthalmitis.
initial repair. Arterial bleeding is also common, as is surgical- If suspected endophthalmitis does occur, it should be man-
induced corneal stromal swelling. Delayed lensectomy can aged as described elsewhere in this book.
be easier because the lens imbibes water, sliding endothelial
cells have closed the cornea, the pupil may dilate better, and
Co rn e al Op acifi cat io n
arterial bleeding has ceased. If ideal circumstances permit
lensectomy at the time of primary repair, this, of course, Modern wide-angle contact lens systems have nearly elimi-
would avoid two trips to the operating room. nated the need for penetrating keratoplasty and temporary
intraoperative keratoprosthesis in the management of severe
intraocular trauma with corneal lacerations or opacifications.
Ro le o f Po st e rio r Vit re ct o m y
A single 1-mm area of clear cornea is sufficient for adequate
Lacerations anterior to the ora serrata (muscle ring) do not cre- viewing of the retina for vitrectomy if a trained assistant is
ate a need for posterior vitrectomy in the early stages. These holding the contact lens. The coupling agent also neutralizes
214 SECTION IV ■ Disease-Sp ecific Managem ent

Fig u re 2 3 .1 1 ■ Op en -sky vit-


rectom y is p referred to th e use
of tem porary ep ikeratop rosthesis
if corneal op acification p revents
sufficient visualization.

the irregular astigmatism created by the corneal wound and vitrectomy and removal of lens material as well. Other forms of
improves visualization in comparison to noncontact indirect traumatic glaucoma are well beyond the scope of this book.
visualization systems. If total corneal opacification is encoun-
tered, a temporary keratoprosthesis (26) could be utilized to References
permit vitrectomy. This device allows visualization without a 1. Goldblum D, Frueh BE, Koerner F. Eye injuries caused by cow
fundus contact lens and allows replacement with the original horns. Retina 1999;19(4):314–317.
blood-stained cornea or a donor button if permanent opac- 2. Cleary PE, Minckler DS, Ryan SJ. Ultrastructure of traction
ity is suspected. The principal author prefers, however, to retinal detachment in rhesus monkey eyes after a posterior
penetration ocular injury. Am J Ophthalmol 1980;90:829.
trephine the cornea and perform the entire procedure “open 3. Cleary PE, Ryan SJ. Experimental posterior penetrating eye
sky” (Fig. 23.11), that is, lens (remnant) removal, vitrectomy, injury in the rabbit 11. Histology of wound, vitreous, and ret-
IOFB removal, epiretinal and subretinal dissection, instilla- ina. Br J Ophthalmol 1979;63:312.
tion of silicone oil through the corneal opening, and suturing 4. Colyer MH, Weber ED, Weichel ED, et al. Delayed intraocular
of the donor button. This approach is faster than use of the foreign body removal without endophthalmitis during Opera-
tions Iraqi Freedom and Enduring Freedom. Ophthalmology
temporary epikeratoprosthesis. It allows removal of very large 2007;114(8):1439–1447. [Epub 2007 Feb 28.]
IOFBs, gentle dissection of epiciliary tissue, bimanual surgery, 5. Ryan SJ, Allen AW. Pars plana vitrectomy in ocular trauma. Am
subretinal dissection, aspiration of all intraocular fluid, and J Ophthalmol 1979;88:483.
easy instillation of silicone. Although endolaser can easily be 6. Ryan SJ. Results of pars plana vitrectomy in penetrating ocular
used open sky, many of these cases are candidates for retin- trauma. Int Ophthalmol 1978;1:5.
7. Ryan SJ. Guidelines in the management of penetrating ocular
opexy avoidance using medium- to long-term silicone. Per- trauma with emphasis on the role and timing of pars plana
fluoron (PFO) can also be used open sky and provides inertial vitrectomy. Int Ophthalmol 1979;1:105.
and gravitational stabilization compared to air. PFO removal 8. DeJuan E, Sternberg P, Michels RG, et al. Timing of vit-
will allow air to fill the eye at the end of the case, enabling sili- rectomy after penetrating ocular injuries. Ophthalmology
cone to be poured into the eye through the corneal incision. 1984;91:1072.
9. Chiquet C. Intraocular foreign bodies. Factors influencing final
visual outcome. Acta Ophthalmol Scand 1999;77(3):321–325.
Glauco m a 10. Coday MP. Nailing down the diagnosis: imaging intraocular
foreign bodies Arch Ophthalmol 1999;117(4):548.
Erythroclastic (hemolytic) glaucoma can be managed effectively 11. De Souza S, Howcroft MJ. Management of posterior segment
by vitrectomy (27) if medical management fails. “Phacogenic intraocular foreign bodies: 14 years’ experience. Can J Ophthalmol
1999;34(1):23–29.
glaucoma” is a general term including phacolytic and lens- 12. Chiquet C, Zech J, Gain P, et al. Visual outcome and prognostic
induced pupillary block. If unresponsive to medical treatment, factors after magnetic extraction of posterior segment foreign
these forms of glaucoma can be managed effectively by bodies in 40 cases. Br J Ophthalmol 1998;82(7):801–806.
CHAPTER 23 ■ Traum a 215

13. Kozielec GF, To K. Penetrating eye injury from a metal wedge. 20. Mandelcorn MS. Results after vitrectomy for trauma. Can J
Ophthalmic Surg Lasers 1999;30(1):59–60. Ophthalmol 1977;12(1):34.
14. Azad R, Sharma VR, Mitra S, et al. Triple procedure in pos- 21. Benson WE, Machemer R. Severe perforating injuries treated
terior segment intraocular foreign body. Indian J Ophthalmol with pars plana vitrectomy. Am J Ophthalmol 1976;B1(6):728.
1998;46(2):91–92. 22. Michels RG. Early surgical management of penetrating ocu-
15. Pavlovic S, Schmidt KG, Tomic Z, et al. Management of intra- lar injuries involving the posterior segment. South Med J
ocular foreign bodies impacting or embedded in the retina. 1976;69(9):1175.
Aust N Z J Ophthalmol 1998;26(3):241–246. 23. Conway BP, Michels RG. Vitrectomy techniques in the man-
16. Michels RG. Surgical management of non-magnetic intraocular agement of selected penetrating ocular injuries. Ophthalmology
foreign bodies. Arch Ophthalmol 1975;93(10):1003. (Rochester) 1978;85(6):560.
17. Michels RG. Closed vitrectomy in trauma: selected intraocu- 24. Michels RG, Conway BP. Vitreous surgery techniques in penetrat-
lar foreign bodies. In: Freeman HM, ed. Vitreous surgery and ing ocular trauma. Trans Ophthalmol Soc UK 1978;98(4):472.
advances in fundus diagnosis and treatment. New York: Appleton- 25. Abrams GW, Topping TM, Machemer R. The effect of vitrec-
Century-Crofts; 1977:335–344. tomy on intraocular proliferation following perforating injuries
18. Cleary PE, Ryan SJ. Vitrectomy in penetrating eye injury. in rabbit eyes. Arch Ophthalmol 1978;96:521.
Results of a controlled trial of vitrectomy in an experimental 26. Landers MB, Foulks G, Landers DM, et al. Temporary keratopros-
posterior penetrating eye injury in the rhesus monkey. Arch thesis for pars plana vitrectomy. Am J Ophthalmol 1981;91:615.
Ophthalmol 1981;99:287. 27. Brucker AJ, Michels RG, Green WR. Pars plana vitrectomy in
19. Hutton WL, Snyder WR, Vaiser A. Vitrectomy in the treatment of the management of blood-induced glaucoma with vitreous
ocular perforating injuries. Am J Ophthalmol 1976;B1(6):733. hemorrhage. Ann Ophthalmol 1978;10:1427.
C H AP TER
24
ENDOPHTHALMITIS

A broad definition of endophthalmitis includes in the management of these patients and require infectious
any severe intraocular inflammation. Toxic sub- disease consultation. Metastatic infection accounts for appro-
stances, necrotic tumors, noninfectious uveitis, ximately 8% of endogenous bacterial endophthalmitis. At
and infarction can create the clinical picture of times, vitrectomy is indicated in this patient group but is
vitreitis, hypopyon, and ocular pain. quite difficult because of the associated anesthesia risk and
Infectious endophthalmitis can be of bacterial, fungal, the potential bilaterality of the disease. Eyes with endog-
or parasitic etiology. Vitreous surgery reduces the number of enous endophthalmitis have increased penetration of sys-
organisms; reduces the intravitreal load of neutrophils, mac- temic antibiotics through breakdown of the blood-retinal
rophages, lymphocytes, and soluble mediators; enhances the barrier, and intravenous antibiotics may be sufficient to treat
penetration and diffusion of antibiotics; and aids in identifi- the disease if the intraocular inflammation is not severe. On
cation of the pathogen. Late complications related to cellular the other hand, whenever bilateral endogenous endophthal-
proliferation on the vitreous matrix are reduced as well. mitis is diagnosed and rapid progression of the diseases is
Early diagnosis and treatment are of paramount impor- noticed or one of the eyes has progressed to severe visual
tance when managing a patient with endophthalmitis. It is loss, the authors often recommend vitrectomy for the less
strongly recommended that all ocular surgery postoperative involved eye to prevent bilateral blindness. Adequate coordi-
patients be examined on the first postoperative morning nation with the infectious disease consultants is paramount.
as well as immediately if the patient complains of pain or If the patient has a known systemic infection, the appropri-
decreased vision. If the physician cannot personally exam- ate intravitreal antibiotic therapy can be chosen. If the infec-
ine the patient, immediate and definite arrangements can tious agent has not been identified, vitrectomy can permit
be made with another qualified physician. The welfare of isolation of the bacteria and guide systemic treatment.
the patient is always the surgeon’s responsibility. Once
endophthalmitis is suspected, one can examine the patient
often and take definitive action when indicated. While the Exo g e n o us En d o p h t h alm it is
more common signs of endophthalmitis are well known
Ocular trauma (2,3) accounts for approximately 20% of bac-
by physicians, less frequent signs are also important. These
terial endophthalmitis (4,5). Ocular surgery (6–8) accounts
include chemosis, lid edema, fibrin membrane formation on
for the vast majority (~70%) as it facilitates the introduction
the intraocular lens (IOL), and retinal hemorrhages.
of organisms into the eye. Management of trauma cases usu-
ally requires vitreous surgery and may involve removal of
intraocular foreign bodies. While conventional surgical wis-
ETIOLOGIC SUBGROUPS dom suggests removal of any implanted materials if infection
occurs, this logic does not apply to the IOL. Removal of an
En d o g e n o us En d o p h t h alm it is
IOL, especially in an endophthalmitis case, has a risk of iris
Endogenous endophthalmitis accounts for a minority of avulsion, endothelial trauma, intraocular bleeding, choroidal
cases (1). Predisposing conditions include immune defi- expulsive hemorrhage, and retinal detachment. Vitrectomy
ciency, immune suppression, diabetes mellitus, chronic renal with intraocular antibiotics without implant removal can be
failure, IV drug abuse, and patients receiving hyperalimenta- successful in a high percentage of cases. This is probably
tion. These patients can develop endophthalmitis without because the smooth surface of the lens implant coupled with
prior ocular disease. Such cases may be bilateral, increasing the high fluid throughput of vitrectomy removes all organ-
both the impact on the patient and the management diffi- isms on the lens surface, but organisms may remain in the
culties. Systemic workup and therapy play an extensive role peripheral capsular bag.

216
CHAPTER 24 ■ Endophthalm itis 217

Fortunately, the incidence of acute postoperative meshwork damage, chronic inflammation, CME, corneal
endophthalmitis remains relatively low. Extracapsular cata- endothelial damage, and iris damage (fixed pupil and tran-
ract surgery with or without an IOL insertion carries an inci- sillumination defects).
dence of 0.072% (9). Vitrectomy (0.051%) and penetrating Causes in general include drugs and devices, process
keratoplasty (0.11%) have a far less risk than the insertion issues, and surgical issues. Device and drug factors include
of a secondary IOL (0.30%). Sutureless clear corneal inci- poor-quality infusion solutions (Cytosol and others), intraoc-
sions and temporal placement of phacoemulsification inci- ular lidocaine, intraocular epinephrine (pH, dose, preser-
sions may have an increased risk of postoperative infectious vative, stabilizing agents: bisulphites or metabisulphites),
endophthalmitis (10). The most frequent organisms include intraocular antibiotics (pH, concentration, dose), intraocu-
coagulase-negative Staphylococcus, Staphylococcus aureus, Strep- lar Kenalog (active drug and preservative), and poor-quality
tococcus species, and Gram-negative bacteria. viscoelastics. Process issues include contamination within
Delayed-onset, post–cataract surgery endophthalmitis steam sterilizer (filters, water chambers, and the inside of
is most often caused by Propionibacterium acnes, coagulase- autoclave), denatured viscoelastics from resterilized cannu-
negative Staphylococcus, or Candida. In the setting of P. acnes las, bacterial endotoxins from Gram-negative bacteria killed
endophthalmitis, inflammatory plaque and associated capsule when cannulas are autoclaved, and contamination of ultra-
must be removed at a minimum; in most instances, the IOLmust sonic cleaner and water baths with enzymes or detergents.
be removed as well (11). Devices designated for single use should not be reused.
The incidence of trauma-related endophthalmitis varies Additional process causes include endotoxins from tap water,
with or without the presence of a retained foreign body. Follow- detergent or chemical residue on instruments, and incom-
ing a penetrating injury, the incidence of endophthalmitis ranges plete cleaning of reusable cannulas. Surgical factors include
from 3.2% to 7.4%. With a retained foreign body, the incidence retained lens cortex, iris trauma, iris retractors, viscoelastics,
jumps to 6.9% to 13% (metallic—7.2%, nonmetallic—7.3%, intracameral agents, IOLs, and endocyclophotocoagulation.
organic—6.3%). The most frequent organisms include Bacillus There are many advantages of disposable forceps, scis-
sp., Staphylococcus sp., and Streptococcus sp. sors, pics, and cannulas: small-diameter 25- or 23-gauge
Streptococcus, coagulase-negative Staphylococcus, and tools are easily damaged during cleaning and sterilization,
Haemophilus are the most common organisms in delayed-onset optimal gripping, and cutting performance beginning to end
filtering bleb–related cases. Immediate vitrectomy interven- of every case, and there is no risk of contamination (Transmis-
tion is mandatory in these often rapidly deteriorating cases. sible Spongiform Encephalopathy (TSE)/Bovine Spongiform
Therapeutic intravitreal injections with anti–vascular Encephalopathy (BSE)/prions, bioburden, hepatitis, HIV/
endothelial growth factor (VEGF) compounds are wide- AIDS, denatured proteins, bacterial endotoxins). Scissors and
spread but fortunately produce a low incidence of postinjec- forceps all have a lumen, and because the IOP is greater than
tion endophthalmitis when meticulous sterile technique is atmospheric pressure, proteins are forced into the lumen.
utilized. Fortunately, the incidence of endophthalmitis after Disposable tools result in a reduced cost per case because of
intravitreal triamcinolone injections, although higher than the elimination of cleanup, sterilization, packaging, storage,
anti-VEGF compounds, appears to be low (~0.1%–0.9%) backup, and inventory costs including labor and materials.
(12,13). The initial workup should include an anterior cham-
ber tap if there is any suspicion of bacterial endophthal-
mitis; it will be negative if TASS is the cause. Vitreous tap
TASS should be performed if there is any vitreous clouding or
clumped inflammatory deposits. If an infectious etiology is
Toxic anterior chamber syndrome must be distinguished suspected, treat with tap and inject intravitreal antibiotics;
from infectious endophthalmitis because the management vitrectomy is unnecessary. If the surgeon has a significant
and causation implications are vastly different. The inci- level of confidence in the diagnosis of TASS, frequent topical
dence after cataract, glaucoma, and corneal surgery is 0.1% antibiotics and close observation are usually enough to con-
to 2.0%, increasing (14) worldwide, and occurring in clus- trol inflammation and recuperate vision. On the other hand,
ters because of institutional outbreaks. Toxic Anterior Seg- a surgeon will never be faulted for performing a vitreous tap
ment Syndrome (TASS) usually is diagnosed within a few and intravitreal antibiotic injection if the diagnosis of infec-
hours after surgery; all patients have blurred vision, severe tious endophthalmitis is considered.
limbus-to-limbus corneal edema (surgical trauma produces
localized edema), 4+ flare, possible hypopyon, fibrin, mini-
mal conjunctiva or episcleral hyperemia, and minimal or ANTERIOR CHAMBER VERSUS
no pain (75%–80% of endophthalmitis patients have pain). VITREOUS ASPIRATION
Although TASS typically presents on the first postopera-
tive day, virulent bacterial endophthalmitis can also present Vitreous taps have a much higher incidence of positive
on the first postoperative day. Outcomes include glaucoma cultures than anterior chamber taps (15). Both types of
due to Peripheral Anterior Synechiae (PAS) and trabecular taps risk pain, wound disruption, intraocular hemorrhage,
218 SECTION IV ■ Disease-Sp ecific Managem ent

and retinal detachment. Because of these problems, many unscrew the syringe with the vitreous sample and proceed
clinicians recommend performing the tap in the operating to sequentially screw the syringes with the medications and
room or a minor procedure room. Vitreous samples obtained inject all the antibiotics and steroids necessary without per-
at the time of therapeutic vitrectomy have higher yields and forming a second penetration of the sclera. Great care should
are safer than vitreous taps. The authors use tap and inject be taken to lock the surgeon’s hand holding the hemostat
intravitreal antibiotics to manage most cases. Busy office and to the patient’s face to avoid injury to the eye if the patient
operating room schedules and the concern about contamina- unexpectedly moves his or her head.
tion of other cases can lead to a delay between the suspected
diagnosis and the performance of the tap. The rapid progres-
sion of this disease does not warrant any delay in the onset TIMING OF VITRECTOMY
of therapy. Given the typical delay between the diagnosis
of endophthalmitis and the start of vitrectomy, the authors Vitreous surgery is not required for every case of infectious
nearly always choose to perform an injection of intravitreal endophthalmitis. Patients with acute-onset postoperative
antibiotics immediately after the diagnosis of endophthal- endophthalmitis and vision of hand motion or better may
mitis even if the situation warrants vitrectomy. Taps should be treated with tap and injection of intravitreal antibiotics
only be done if they are accomplished immediately and with alone, as shown by the Endophthalmitis Vitrectomy Study
the intent of not performing vitreous surgery at that time. (EVS) study. If the vision is worse than hand motion, vitre-
Suspicion of the operating room environment, IOL, or surgi- ous surgery should be undertaken immediately, regardless of
cal materials as an etiologic agent demands an epidemiologic the hour of the day or night.
approach to the workup. In general, the goal should be to Advanced cases with corneal decompensation cannot
initiate treatment immediately. have vitreous surgery under safe conditions because of the
visualization requirements. Open-sky vitrectomy would be
fraught with complications and is not recommended. Imme-
TECHNIQUE FOR IN-OFFICE diate intracameral antibiotics offer the best hope in this poor
VITREOUS TAP AND INJECTION prognosis group and in patients with medical problems pre-
OF INTRAVITREAL ANTIBIOTICS venting surgical intervention.
EVS findings do not apply to endophthalmitis cases
Anesthesia with a peribulbar injection of lidocaine and topi- that are endogenous, delayed onset postoperative, traumatic,
cal viscous lidocaine is necessary for this procedure. Retrob- or filtering bleb related.
ulbar anesthesia in the office without intravenous sedation
in a very anxious patient with acute ocular pain is usually SURGICAL SEQUENCE AND
not recommended. TECHNIQUES
Sterile techniques should be utilized the same way that
intravitreal injections are performed, with 5% iodine on the The operating room should be alerted to prepare for a
conjunctival surface and eyelids and the use of a bladed dirty case with all appropriate isolation and postsurgical
speculum to cover the lashes. The main difference in tech- cleanup precautions. This should not be used as an excuse
nique revolves around these factors: (a) multiple medications for delay of therapy, however. At times, it is best to operate
are usually injected (typically vancomycin, ceftazidime, and in a nonophthalmic operating room with only the minimal
dexamethasone), (b) these medications should not be mixed equipment required for the case.
in the same syringe since they can precipitate, (c) a success- Monitored local anesthesia (MAC) is appropriate in
ful vitreous tap requires a larger caliber needle, usually 25 these cases. Intravenous antibiotics should be started at this
gauge or larger, and (d) the acutely inflamed eye is very dif- time if a presurgical medical treatment plan has not been
ficult to anesthetize using topical anesthetics. utilized, but the role of systemic antibiotics is controversial.
A vitreous tap and injection of all the required medi- If a cataract wound, surgical wound, or ruptured filter-
cations can be performed with a single needle penetration ing bleb is present, it must be secured before the initiation
of the sclera. As the first step, a 3-mL syringe with a short of vitreous surgery. Absorbable sutures should be removed
25-gauge needle is firmly grasped at the hub of the needle
and replaced with 10-0 or 9-0 monofilament nylon sutures.
with a locking mosquito hemostat, perpendicular to the axis Careful attention to a tight wound prior to vitrectomy can
of the needle. The needle is introduced into the pars plana decrease subsequent problems.
and gentle suction is applied to the embolus, withdrawing
the vitreous sample. In a nonvitrectomized eye, it is usu-
Tran s–Pars Plan a Ve rsus Tran slim b al Ap p ro ach
ally difficult to withdraw more than 0.1 mL of vitreous. In a
previously vitrectomized eye, if the tip of the needle is intro- As many endophthalmitis cases can and should be handled
duced beyond the vitreous base, it is easy to withdraw up to by predominantly anterior segment surgeons, the lim-
0.3 mL of liquid vitreous. The surgeon can then firmly hold bal approach can be considered. Unfortunately, the limbal
the hemostat that is immobilizing the hub of the needle and approach prevents adequate visualization for posterior
CHAPTER 24 ■ Endophthalm itis 219

vitrectomy and causes more corneal and iris trauma. effective dose should be used. Intraocular antibiotics should
Translimbal vitrectomy should be reserved for the novice always be used if an IOL is present. Antibiotics diluted in the
surgeon only in a true emergency. Translimbal vitrectomy infusion fluid are not recommended because of the toxicity
is virtually impossible in patients with posterior chamber question and the difficulty in assessing total retinal dose with
IOLs, which represents the biggest subgroup of endophthal- this method. Vancomycin 1.0 mg/0.1 mL and ceftazidime
mitis cases. 2.25 mg/0.1 mL are the most commonly used agents today.
It has been shown by D’Amico et al. (20) that pharmacists
mix antibiotics more accurately than nurses and the nurses
Vit re ct o m y are far more accurate than physicians. Many toxicity cases
The vitrectomy instrument should be used with the lowest are probably due to incorrect concentrations and/or volumes
possible suction force, with the highest cutting rate, preferably being injected. Intraocular dexamethasone 0.4 mg/0.1 mL
5,000 cuts per minute, and proportional suction control to has been recommended by many investigators and has been
reduce the chance of tearing necrotic retina and iris. A 25-gauge shown to produce better outcomes (21).
vitrectomy is ideal for this application, but 23-gauge vitrec- The antibiotic should then be injected slowly in the mid-
tomy can be used as well. Utilize the highest possible cutting vitreous cavity through a cannula with a 25-gauge needle. If
rate, and never pull the cutter away from the retina while suc- two to three separate injections are utilized, as is usually the
tion is being applied. The anterior vitreous cortex should be case, these can be applied sequentially in the same wound
removed first, with special care taken to avoid iris contact and with this method. Mixing the antibiotics in the same syringe
peripheral vitreoretinal traction. The iris tissue can be quite is poor practice and results in dilution and precipitation.
necrotic and may be easily shredded or avulsed. On occasion,
hypotony will lead to oozing from iris vessels, requiring bipo- Sub co n jun ct ival An t ib io t ics an d St e ro id s
lar diathermy. If a fibrin membrane covers the anterior surface
After removal of 25-gauge cannulas, subconjunctival antibi-
of the IOL, it can be removed through a peripheral iridectomy
otics can be considered. Vancomycin 25 mg and ceftazidime
constructed with the vitreous cutter. Viscoelastic can also be
100 mg are the most widely used agents at the time of this
injected through a small limbal paracentesis to displace fibrin
writing. Detailed discussion of antibiotic options is left to
from the surface of the intraocular lens. Limbal incisions fre-
other publications because of their rapidly changing status.
quently leak, causing hypotony and miosis.
It is recommended that an antibiotic that is primarily for
Endophthalmitis is one of the rare situations in which
Gram-positive organisms and effective on penicillinase pro-
only a “core” vitrectomy should be done to avoid traction
ducers be combined with an antibiotic that is primarily for
on the potentially necrotic retina. At least 50% of the
Gram-negative organisms. Substances released from inflam-
formed vitreous is removed with this approach. The dense
matory cells, as well as bacteria, cause severe tissue destruc-
initial vitreous aspirate should be removed for smear,
tion and inflammation. The inflammation suppression role
culture, and sensitivity testing (16,17). Blood culture bottles
of steroids is thought to far outweigh the potential enhance-
have been shown by Joondeph and Flynn et al. (18) to be
ment of infection. If the case is strongly thought to be fungal
equally effective at achieving a positive culture compared to
in origin, then steroids should probably be omitted.
inoculating culture plates and tubes in the operating room. The
Although the authors recommend using prophylactic
endoilluminator is essential to adequate visualization. Vacuum
subconjunctival antibiotics routinely after vitrectomy sur-
cleaning (extrusion) and membrane peeling should never be
gery (whether for the treatment of endophthalmitis or other-
utilized in these cases because of the necrotic retina. Creation
wise), subconjunctival antibiotics do not alter the course of
of an intraoperative posterior vitreous detachment can also
endophthalmitis adequately treated with intravitreal antibi-
lead easily to iatrogenic retinal tears. If there is severe retinal
otic injections or surgery (22).
necrosis, the retina will appear white and rough surfaced with
the vessels appearing dull. Such retinas are extremely prone to
retinal breaks and can be seen to move with the probe several To p ical An t ib io t ics
millimeters away from the retina, even with the mild pulsatile Topical antibiotics and steroids are typically used in all cases,
suction force of the vitrectomy instrument. primarily to inhibit potential, associated lid, cul-de-sac, bleb,
and wound infections (23). Care should be taken to prevent
cross contamination by doctors, nurses, and technicians tak-
USE OF ANTIBIOTICS ing care of other patients.

In t racam e ral An t ib io t ics


RESULTS
Intraocular antibiotics are used in virtually all cases (19).
There is currently less disagreement in the literature about The causative organisms and pretreatment delay constitute
safe doses of intraocular antibiotics than in previous years. the most important factors in therapeutic success. If cases
Because of the risk of retinal toxicity, the lowest published with mild pathogens that could have been managed with
220 SECTION IV ■ Disease-Sp ecific Managem ent

subconjunctival and systemic antibiotics are operated upon, 8. Solomon A, Ticho U, Frucht-Pery J. Late onset bleb associ-
the success rate appears to improve. Even with the best man- ated endophthalmitis following glaucoma filtering surgery
with or without antifibrotic agents. J Ocul Pharmacol Ther
agement, a significant percentage of these eyes will be lost. In
1999;15(4):283–293.
view of the overall poor prognosis in endophthalmitis cases, 9. Cottingham AJ, Forster RK. Vitrectomy in endophthalmitis;
major emphasis should be placed on prevention. Meticulous results of study using vitrectomy, intraocular antibiotics, or a
preparation of the operative field is vital, including a drape combination of both. Arch Opthalmol 1976;94:2078.
that covers the lashes and lid margins, Betadine prep, micro- 10. Nagaki Y, Hayasaka S, Kadoi C, et al. Bacterial endophthal-
mitis after small-incision cataract surgery. Effect of incision
scope draping, use of the highest quality infusion fluids (Alcon
placement and intraocular lens type. J Cataract Refract Surg
BSS, BSS Plus), and extreme care if tubing or any other instru- 2003;29(1):20–26.
ments with a lumen or cavity are resterilized. Subconjunctival 11. Clark WL, Kaiser PK, Flynn HW Jr, et al. Treatment strate-
antibiotics are used at the end of vitrectomy cases, which may gies and visual acuity outcomes in chronic postoperative
decrease the incidence of this dreaded complication. Propionibacterium acnes endophthalmitis. Ophthalmology
1999;106(9):1665–1670.
12. Westfall AC, Osborn A, Kuhl D, et al. Acute endophthal-
mitis incidence: intravitreal triamcinolone. Arch Ophthalmol
COMPLICATIONS 2005;123(8):1075–1077.
13. Moshfeghi DM, Kaiser PK, Scott IU, et al. Acute endophthalmi-
Corneal edema, glaucoma, and retinal detachment can be tis following intravitreal triamcinolone acetonide injection. Am
J Ophthalmol 2003;136(5):791–796.
seen after otherwise successful endophthalmitis manage- 14. Kim SY, Park YH, Kim HS, Lee YC. Bilateral toxic anterior
ment with or without vitrectomy. Prompt therapy is prob- segment syndrome after cataract surgery. Can J Ophthalmol
ably the single most important factor in the prevention of 2007;42(3):490–491.
these nonspecific complications. 15. Results of the Endophthalmitis Vitrectomy Study. Arch Oph-
Retinal breaks and detachment related to retinal necro- thalmol 1995;113(12):1479–1496.
16. Frederick AR. A modification of the Ocutome setup to permit
sis and surgically induced retinal traction occur in at least the sterile collection of intraocular specimens. Ocutome Frag-
10% of the cases. Postoperative follow-up includes periph- matome Newsletter No. 3,1978:2.
eral retinal examination by indirect ophthalmoscopy because 17. Smith RE. A technique for collecting uncontaminated speci-
of the relatively high frequency of retinal breaks. mens for culture during vitrectomy for endophthalmitis. Ocu-
tome Fragmatome Newsletter No. 3, 1978:2.
18. Joondeph BC, Flynn HW, Miller D, Joondephet HC. A new cul-
References ture method for infectious endophthalmitis. Arch Ophthalmol
1989;107(9):1334–1337.
1. Romero CF, Rai MK, Lowder CY, Adal KA. Endogenous 19. Peyman GA, Vashine DW, Crouch ER, Herbst RW. Clinical use
endophthalmitis: case report and brief review. Am Fam Physi- of intravitreal antibiotics to treat bacterial endophthalmitis.
cian 1999;60(2):510–514. Trans Am Acad Ophthalmol Otolaryngol 1974;8:862.
2. Brinton GS, Topping TM, Hyndiuk RA, et al. Post-traumatic 20. D’Amico DJ, Caspers-Velu L. Comparitive toxicity of intravit-
endophthalmitis. Arch Ophthalmol 1984;102:547. real aminoglycoside antibiotics. Am J Ophthalmol 1985;100(2):
3. Forster RK. Endophthalmitis. In: Duane TD, ed. Clinical oph- 264–275.
thalmology, Vol. 4. New York: Harper & Row, 1981:1–20. 21. Park SS, Vallar RV, Hong CH, et al. Intravitreal dexamethasone
4. Abu el-Asrar AM. Post-traumatic endophthalmitis: causative effect on Vancomycin elimination in endophthalmitis. Arch
organisisms and visual outcome. Eur J Ophthalmol 1999;9(1): Ophthalmol 1999;117(8):1058–1062.
21–31. 22. Smiddy WE, Smiddy RJ, Ba’Arath B, et al. Subconjunctival
5. Meredith TA. Posttraumatic endophthalmitis. Arch Ophthalmol antibiotics in the treatment of endophthalmitis managed with-
1999;117(4):520–521. out vitrectomy. Retina 2005;25(6):751–758.
6. Forster RK. Etiology and diagnosis of bacterial post-operative 23. Bannerman TL, Rhoden DL, McAllister SK, et al. The source of
endophthalmitis. Ophthalmology 1978;85:320. coagulase-negative staphylococci in the EVS. Arch Ophthalmol
7. Schmitz S, Dick HB, Krummenauer F, Pfeiffer N. Endophthal- 115(3):357–361.
mitis in cataract surgery: results of a German study. Ophthal-
mology 1999;106(10):1869–1877.
C H AP TER
25
SURGICAL MANAGEMENT
OF THE UVEITIS PATIENT

for cutting to begin before aspirating on the syringe, thereby


VITREORETINAL SURGERY
preventing peripheral retinal tears. The surgeon can then
IN THE MANAGEMENT OF direct the sampling of the vitreous to the area that has the
UVEITIC DISORDERS most apparent cellular infiltration. This increases the yield
of the vitreous biopsy and increases safety since the retina is
Vitrectomy is recommended in certain situations for the visualized at all times during the dry aspiration. As soon as
management of uveitic diseases. Vitrectomy may be needed the eye appears to become soft, the surgeon can direct the
for diagnostic purposes as well as therapeutic purposes. assistant to stop manual aspiration and then disengages the
cutter action. Immediately the fluid infusion can be initiated
Diag n o st ic Vit re ct o m y and the intraocular pressure normalized. Appropriate order
of steps is important to decrease the potential for iatrogenic
Indica tions retinal tears.
The three main indications for diagnostic vitrectomy in Some surgeons prefer to use air infusion during the
uveitis are (a) severe vitreitis that precludes visualiza- undiluted vitreous biopsy, trying to decrease intraopera-
tion of the fundus and diagnosis is not clear, (b) suspi- tive hypotony. The authors’ main objection to this tech-
cion of intraocular lymphoma, and (c) retinochoroiditis of nique is that it compromises visualization during the
unclear etiology that threatens vision in the short term and vitrectomy and prevents adequate positioning of the cutter
(d) cystoid macular edema (CME) unresponsive to medical on the most pathological area of the eye. Once the undi-
management. luted vitreous sample has been obtained, standard vitrec-
tomy can continue with normal pars plana infusion. The
vitrectomy cassette can then be sent directly to the labora-
Vitreous Sa mpling Techniques
tory or a syringe can be used to aspirate from the cassette
Whenever diagnostic vitrectomy is performed, great care to be sent.
should be taken with the vitreous sample, which, after all, is As in all vitrectomy cases, great care should be taken
the main goal of the surgery. There are variations in the sur- at the end of the surgery to visualize the retinal periph-
gical technique that can provide adequate vitreous samples. ery and verify the absence of peripheral retinal tears. The
The authors will present the techniques that they find pref- creation of a posterior vitreous detachment (PVD) dur-
erable in their hands. Surgery begins by placing the infer- ing surgery has potential pitfalls. If there are any areas of
otemporal 25-gauge cannula and attaching to it the infusion retinitis, the likelihood of retinal tears during PVD creation
cannula in the off position (without infusion). Following the is high. On the other hand, patients with dense vitreitis
placement of the superior 25-gauge cannulas, removal of who undergo core vitrectomy will have postoperative PVDs
undiluted vitreous sample can begin. For this step, the aspi- and can often bitterly complain of floaters. Surgical safety
ration line coming from the vitrectomy probe is disconnected should override minor issues such as postoperative float-
from the Constellation vitrectomy system and attached to a ers. The authors do not recommend performing PVDs rou-
syringe. With adequate visualization with the endoillumina- tinely during diagnostic vitrectomies, but do explain to
tor and plano irrigating contact lens, the surgeon engages patients the possibility of floaters. As long as the patient
the cutting action of the vitrectomy probe and immediately understands that the reason for the surgery is to prevent
after orders the assistant to begin gentle aspiration with the blindness, issues such as floaters can be placed in their
syringe on the line. It is important to have the assistant wait appropriate perspective.
221
222 SECTION IV ■ Disease-Sp ecific Managem ent

La bora tory Studies low on the authors’ initial differential diagnosis have led the
authors to recommend PCR testing for herpes simplex virus,
The authors cannot overemphasize the importance of varicella zoster virus, and cytomegalovirus whenever there
personal communication with the pathologist prior and is sufficient sample to run these tests after vitreous biopsy.
following diagnostic vitrectomies. Given the small volumes Again, direct communication with the pathologist is the
of the authors’ samples, protocol-based testing can poten- authors’ best tool to increase efficient utilization of valuable
tially waste highly valuable samples on unimportant tests. and scant vitreous material.
Since clinical laboratory techniques are constantly changing
and improving, the vitrectomy surgeon may be unaware of
the availability of certain tests that can provide the needed Re t in al Bio p sy
diagnosis. The authors strongly advocate personally explain- There are clinical scenarios that require retinal and even
ing to the pathologist the differential diagnosis and following choroidal biopsy for final diagnosis. The potential risks of
the pathologist’s recommendations regarding handling of the postoperative retinal detachment and intraoperative hemor-
material and prioritizing of studies. rhage should limit the frequency of these complex cases.

Cytology and Flow Cytom etry for Diagnosis


of Intraocular Lym phom a Indica tions
The diagnosis of intraocular B-cell lymphoma requires The indication for performing a retinal biopsy is progressive
vitreous biopsy for cytology and flow cytometry analysis (1). undiagnosed relentless retinitis that cannot be diagnosed
Whereas many surgeons in the past have delayed diagnostic with less invasive means. The availability of PCR testing for
vitrectomy in lieu of empirical steroid therapy and/or neu- viral infections and modern flow cytometry and gene rear-
roimaging with lumbar puncture, the authors routinely per- rangement studies as discussed previously have decreased
form 25-gauge diagnostic vitrectomies on elderly patients the need for retinal biopsies. The authors usually only per-
with new-onset vitreitis if a syndromic clinical diagnosis is form a retinal biopsy in the setting of bilateral retinitis in
unclear. Steroid therapy risks partial clinical improvement which one eye has progressed to profound vision loss and
with delayed diagnosis. Neuroimaging and lumbar puncture the contralateral eye has good vision but is in danger. In this
should be recommended on patients with headaches or neu- circumstance, retinal biopsy of the blind eye has little risk of
rological signs or symptoms (2), but they are of low yield worsening the clinical condition and can potentially provide
on patients who have isolated vitreitis without neurological a diagnosis that can save the good eye.
complaints. Cytological examination should be done by an
experienced cytopathologist, and no delay in the transfer
of the sample should occur, since the lymphoma cells can Techniques for Retina l Biopsy
die and render a biopsy useless if they are left out waiting Follow the steps mentioned previously for vitreous biopsy
for someone to pick up the sample to take it to the labo- (the authors recommend to always send vitreous for labora-
ratory. Since the authors communicate with the pathologist tory analysis in cases where retinal biopsies are performed).
prior to every diagnostic vitrectomy for possible lymphoma, The next step is the selection of the biopsy site. The surgeon
the pathologist comes into the operating room and receives should remember that biopsies of necrotic retinal tissue are
the undiluted sample for immediate processing before the usually not diagnostic. The best location for biopsy is at the
authors finish the core vitrectomy, increasing the yield of the leading edge of the retinitis, where viable retinal cells with
study. pathological changes can increase diagnostic yield. If there
is no leading edge of disease, perhaps from diffuse retinal
Gene Rearrangem ent Studies for Lym phom a Diagnosis involvement, the most comfortable location for biopsy can
Modern cytogenetic studies can be performed on vit- be selected. Immunohistochemical staining of necrotic tissue
reous lymphocytes whenever the diagnosis of intraocular can still be positive for viral antigens.
lymphoma is unclear. Experienced laboratories can provide Retinal biopsies are technically easier when performed
a final lymphoma diagnosis with only a few cells analyzed around the posterior pole. The authors usually choose a site
based on gene rearrangement studies (3). just peripheral to the vascular arcades or nasal to the optic
nerve. The authors first perform three rows of retinal laser
Polym erase Chain Reaction for Infectious Diseases surrounding the designated biopsy site and later cut the reti-
Polymerase chain reaction (PCR) testing can confirm the nal biopsy free hand with 25-gauge scissors. Retrieval of the
presence of viral DNA in the setting of unusual viral retinitis biopsy tissue can sometimes be challenging. The authors
such as atypical acute retinal necrosis syndrome, progressive prefer to remove a 25-gauge cannula, perform a small con-
outer retinal necrosis syndrome, and cytomegalovirus retini- junctival incision for exposure of the sclerotomy site, and
tis (4). Despite the authors’ opinions against protocol-based enlarge the sclerotomy with a 20-gauge microvitreoretinal
testing, personal experiences with misdiagnosed viral retini- blade. 25-G ILM forceps can then be used to grasp the reti-
tis in which PCR testing was not performed since they were nal tissue and retrieve it atraumatically from the eye through
CHAPTER 25 ■ Surgical Managem ent of the Uveitis Patient 223

the 20-gauge incision, which is later sutured. End-grasping recommend laser demarcation in necrotizing retinitis for
forceps should not be used because they have a larger the following reasons: (a) if done during active disease,
grasping platform and can damage the retinal tissue. the surgeon may underestimate the final extent of the reti-
A similar technique can be performed to remove chor- nal necrosis, (b) if the area of retinal necrosis is small, the
oidal tissue, but it may require endodiathermy of the chor- risk of detachment is low, and (c) on the other hand, those
oid surrounding the biopsy site to decrease intraoperative patients with extensive retinal necrosis who have a high risk
and postoperative hemorrhage. Following retinal biopsy of progressing to retinal detachment have such compromise
on previously attached retina, the authors perform fluid-air of visual fields that the surgeons do not advocate further
exchange to decrease postoperative retinal detachment. encroachment of their field of vision by laser photocoagula-
The fixation media should be chosen after discussion tion. Rhegmatogenous retinal detachments that present after
with the pathologist to verify that appropriate special stains retinal necrosis are usually relatively easy surgical repairs,
are not ruined. which, if done, promptly may not damage visual function
significantly. The authors do, therefore, thoroughly explain
to patients the risk of retinal detachment and the need for
Th e rap e ut ic Vit re ct o m y
immediate communication with them in case of visual
Vitrectomy should be seen as a potential treatment strategy changes suggestive of retinal detachment.
in the therapy of severe posterior uveitis. The most common
scenarios are described in the following sections. Technical Considerations for Repair of Retinal
Necrosis–Associated Retinal Detachm ents
These retinal detachments usually have broad areas
Adjuva nt Thera py in the Ma na gement of
of retinal holes and atrophy that extend posteriorly. Scleral
Intermedia te Uveitis
buckling of these large holes is usually impossible. The
Vitrectomy surgery can positively affect the course of chronic authors perform 25-gauge vitrectomy and fluid-air exchange
idiopathic intermediate uveitis (pars planitis) (5). Patients with with simultaneous drainage of subretinal fluid through the
unresponsive CME can have decreased edema and improve- retinal holes to achieve retinal reattachment. The authors
ment in vision following vitrectomy (6). The explanation for usually do not require perfluorooctane or posterior drainage
the improved clinical course in pars planitis following vit- retinotomies given the presence of posterior retinal defects.
rectomy is not well understood. The authors postulate that Use of long-term silicone oil rhegmatogenous confinement is
a vitreous antigen may be the target of the immune response usually required for permanent retinal reattachment. Periph-
and that vitrectomy may debulk this antigen and improve eral vitrectomy should be undertaken with great care, at the
the clinical inflammation. Considering that 25-gauge vitrec- highest cut rate possible and with the lowest suction that
tomy is a safe procedure when done by an experienced sur- is sufficient for vitreous removal, since iatrogenic retinal
geon, the authors strongly support the idea of progressing to tears are easily created on these atrophic retinas. Intraop-
complete pars plana vitrectomy before immunosuppressive erative laser retinopexy versus postoperative laser treatment
therapy is considered. The risk profile of a well-performed is decided based on the clinical likelihood of proliferative
vitrectomy is much better than the systemic complications vitreoretinopathy. The authors tend to defer laser retinopexy
associated with immunosuppressive therapy. whenever there are any remaining inflammatory signs in the
eye until total resolution of inflammation is determined. This
decreases the stimulus for PVR and limits the extent of the
Ma na gement of Complica tions of Severe Uveitis
laser treatment only to the areas that absolutely require it.
Vitreoretinal surgery may be required for the management
of complications of posterior segment uveitis. The following Traction Retinal Detachm ents in Interm ediate
scenarios are the most important clinically. and Posterior Uveitis
Severe pars planitis can develop neovascularization of
Rhegm atogenous Retinal Detachm ents the disc (NVD) and retinal periphery (8). Similar to NVDs
in Necrotizing Retinitis in diabetes, contraction of the NVD with attached posterior
Viral retinitis that develops retinal necrosis can frequently hyaloid can develop tractional retinal detachments. Traction
complicate with large retinal holes in the areas of the retinal retinal detachment without retinal neovascularization can
necrosis that progress to rhegmatogenous retinal detachment. also be seen in any uveitic condition that develops severe
vitreous inflammation in an eye without preexisting PVD.
Prophylactic Retinopexy Around Attached This may occur in pars planitis, toxoplasmosis, toxocariasis,
Necrotic Retina and sarcoidosis.
Many authors have suggested performing laser demar-
cation posterior to the areas of retinal necrosis to decrease Technical Considerations
the likelihood of ulterior retinal detachments (7). Despite The surgical goal for repair of traction retinal
acknowledging this technique, the authors do not routinely detachments in uveitis, as in any traction retinal detachment,
224 SECTION IV ■ Disease-Sp ecific Managem ent

is relief of all traction vectors that are acting on the retina Im p lan t at io n o f Drug De live ry Syst e m s
without creation of retinal tears in the process. Contraction of
At the time of this writing, there are two classes of medications
NVDs in pediatric patients can present as an unusual traction
that are available on implantable drug delivery formulations:
detachment where the papillomacular bundle and the nasal
intravitreal steroids and intravitreal ganciclovir. The specific
retina immediately adjacent to the optic nerve adhere to each
indications for the use of these implants are beyond the
other, covering the underlying optic nerve head. An inexpe-
scope of this book.
rienced examiner may assume that the origin of this traction
configuration is the peripheral retina and attempt to repair
this configuration through risky peripheral vitrectomy. These
detachments are best repaired by careful scissors dissection at
the posterior pole where the NVD contraction occurred. LENS MANAGEMENT
Peripherally originated vitreous traction, as in toxo- IN SEVERE UVEITIS
cariasis or toxoplasmosis, can create peripheral retinal folds
that reach the optic nerve. These folds may not be readily There has been a push for increased use of immunosup-
apparent through the vitreous opacification, but the surgeon pressants in severe uveitis to allow phacoemulsification with
should infer their location based on the contours of the sur- intraocular lens (IOL) placement in the setting of cataracts.
rounding retina to avoid iatrogenic retinal tears. Sometimes While a patient with mild uveitis can undergo safe IOL
the vitreous traction takes the configuration of broad pos- placement with topical steroids only, the authors believe that
terior attachment centered on the optic nerve with a cone patients with severe uveitis and cataracts are best served by
of posterior hyaloid creating anteroposterior traction. Cone performing pars plana vitrectomy, lensectomy, and capsulec-
truncation, similar to diabetic vitrectomies, can release this tomy. This avoids the possibility of lens-induced uveitis,
traction, but the authors always proceed to peel the posterior chronic CME, posterior synechiae and secondary glaucoma,
vitreous from the retinal surface to decrease postoperative and cyclitic membrane formation. Immunosuppressants
epimacular membranes and macular striae from contraction should not be seen as a therapy for aphakia, which creates a
of the posterior hyaloids left over the macula. single-chamber eye that allows easier access of topical medi-
cations and prevents synechiae and chronic CME. Aphakic
Vitreom acular Interface Disorders spectacles or contact lenses have a much safer risk profile
Epimacular membranes, vitreomacular traction syn- than antimetabolites and high-dose steroids.
drome, and macular holes are frequent in the setting of
severe uveitis. There is no significant difference in surgical
approach to these macular disorders compared to nonuveitis
References
eyes. The surgeon should only attempt whenever possible to
defer vitrectomy until clinical quiescence of the inflamma- 1. Davis JL. Diagnostic testing of vitrectomy specimens. Am
tion is observed. J Ophthalmol 2005;140(5):822–829.
2. Herrlinger U, Schabet M, Bitzer M, et al. Primary central
Many uveitic patients can develop CME, and the vitre- nervous system lymphoma: from clinical presentation to
ous surgeon can be presented with the question of whether diagnosis. J Neurooncol 1999;43(3):219–226.
to recommend vitrectomy surgery in the patient with CME. 3. White VA, Gascoyne RD, Paton KE. Use of the polymerase
Certain issues should be analyzed: (a) Is medical therapy chain reaction to detect b- and t-cell gene rearrangements in
optimized to control the inflammation? The main cause for vitreous specimens from patients with intraocular lymphoma.
Arch Ophthalmol 1999;117:761–765.
CME in uveitis is diffuse breakdown of the blood-retinal bar- 4. Knox CM, Chandler D, Short GA, Margolis TP. Polymerase
rier due to inflammatory mediators. Other than the specific chain reaction-based assays of vitreous samples for the diagno-
setting of intermediate uveitis where, as mentioned previ- sis of viral retinitis. Use in diagnostic dilemmas. Ophthalmology
ously, vitrectomy can improve the inflammatory reaction 1998;105(1):37–44.
and decrease CME, the best course of treatment for uveitic 5. Becker M, Davis J. Vitrectomy in the treatment of Uveitis. Am
J Ophthalmol 2005;140(6):1096–1105.
CME is medical control of the inflammation. (b) Is there any 6. Wiechens B, Nölle B, Reichelt JA. Pars-plana vitrectomy in
tractional component to the CME? Modern spectral domain cystoid macular edema associated with intermediate uveitis.
optical coherence tomography (OCT) can help answer this Graefe’s Arch Clin Experimen Ophthalmol 2001;474–481.
question. The presence of macular traction on OCT, based 7. Han DP, Lewis H, Williams GA, et al. Laser photocoagula-
not only on the physical presence of a membrane but also on tion in the acute retinal necrosis syndrome. Arch Ophthalmol
1987;105:1051–1054.
the observation of sharp contour changes on the macula due 8. Kalina PH, Pach JM, Buettner HM, Robertson DM. Neo-
to traction, suggests that vitrectomy with membrane peeling vascularization of the disc in pars planitis. Retina 1990;10:
can help control CME. 269–273.
C H AP TER
26
RETINOPATHY OF PREMATURITY
AND PEDIATRIC 25-GAUGE
VITRECTOMY

Retinopathy of prematurity (ROP) is a disease of screening services and (b) timely and appropriate laser
of premature, low–birth-weight infants that is treatment for active ROP disease. Vitrectomy surgery should
characterized by the presence of peripheral avas- be viewed as salvage therapy for selected cases.
cular retina, neovascularization at the junction
of the vascular to the avascular retina, and cicatricial traction
on the retina with subsequent traction retinal detachments. NORMAL RETINAL DEVELOPMENT
ROP has a distinct epidemiology worldwide that depends
on the availability of sophisticated neonatal intensive care Re t in al Vascular De ve lo p m e n t
units (NICUs), trained ophthalmologists for screening and
To understand the pathophysiology of ROP, we must start
treatment, adequate coordination of services, and obstet-
with a basic framework of normal retinal vascular develop-
rical care. In general, ROP is not a significant problem in
ment. Mesenchymal spindle cells grow from the optic nerve
countries where the lack of intensive neonatal services leads
toward the retinal periphery and differentiate centrifugally
to poor survival of very low–birth-weight infants, as in cer-
into retinal blood vessels. Vasculature development starts at
tain countries in sub-Saharan Africa. Moving up in the eco-
the optic nerve at gestational age 16 weeks. Retinal blood
nomic development, middeveloped countries, such as many
vessels reach the nasal ora serrata at week 36. Infants born
countries in Latin America and Eastern Europe, have a high
before full retinal vascular development have peripheral reti-
incidence of severe ROP, given that NICUs are available, but
nas devoid of fully developed blood vessels.
tight postnatal oxygen control, sufficiently trained ophthal-
mic consultants, and adequate coordination of services are
difficult to provide. Whereas ROP had decreased in inci- PATHOPHYSIOLOGY
dence in the United States as neonatal services had improved
OF RETINOPATHY OF PREMATURITY
and coordination with ophthalmologists trained in screen-
ing and treatment of ROP had been achieved, two issues are
Vascular En d o t h e lial Gro wt h Fact o r
increasing the incidence of the disease: (a) increased sur-
in No rm al Re t in al Vascular De ve lo p m e n t
vival of extremely low–birth-weight infants, who commonly
develop ROP, and (b) increased incidence of multiple preg- Vascular endothelial growth factor (VEGF) is one of the major
nancies (twins, triplet, quadruplet pregnancies), in large part molecules involved in retinal vascular development and is
due to aggressive fertility treatments and in vitro fertilization critical in the pathophysiology of ROP. Normal VEGF pro-
(1). A more recent problem is that ophthalmologists willing duction is required for vascular development. Both phases
and able to treat ROP are becoming less common due to of ROP have dysregulation of VEGF.
fears of liability and due to the practical concerns of coordi-
nating an ROP service.
Dire ct io n al Vasculo g e n e sis
The basic premise that this chapter will try to convey is
that, beyond the specific technical details of vitreous micro- An important aspect of retinal vascular development is its
surgery, the most cost effective and reliable way to decrease “directionality.” In other words, vessel development has to
ROP blindness is determined by (a) adequate coordination progress from the nerve toward the periphery. Although the
225
226 SECTION IV ■ Disease-Sp ecific Managem ent

biological details of vessel development are beyond the scope vitreous VEGF leads to retinal neovascularization. In ROP,
of this book, some simple concepts should be understood. the localization of the neovascularization is at the border of
The VEGF gene promoter has “hypoxia response elements” the vascular and avascular retina.
that bind hypoxia-induced factors (2). This creates a feed-
back loop that increases VEGF production in hypoxic tissues
De ve lo p m e n t o f Tract io n
and decreases VEGF levels as tissue oxygen levels increase.
Re t in al De t ach m e n t
This relation between hypoxia and VEGF also plays a role in
diabetic retinopathy, sickle cell retinopathy, and other isch- Cicatricial regression of the retinal neovascularization leads
emic retinopathies. In the developing retina, areas absent of to retinal traction and secondary traction retinal detachment.
blood vessels are relatively hypoxic in comparison to vascu- An understanding of the vectors of traction is important to
larized retina. This creates a gradient of VEGF concentrations surgical relief of traction and repair of the retinal detach-
that provide the “directional signal,” akin to chemotaxis, for ments. The origin of all traction vectors is the circumferen-
blood vessel development. tial ring of retinal neovascularization at the ridge between
the vascular and the avascular retina. From this point, the
traction, as in the anterior loop traction (radial vitreous
collagen fibers) in proliferative vitreoretinopathy (PVR), is
directed toward the anterior vitreous base, the lens, and the
diametrically opposite neovascular ridge. Since these infants
do not have a posterior vitreous detachment, another vector
that can play a role in traction is directed from the ridge to
the posterior pole and optic nerve.

MANAGEMENT OF THE ROP INFANT


Ph ase 1 o f ROP: Arre st o f No rm al
The CryoROP study demonstrated that ROP was amenable to
Vascular De ve lo p m e n t
successful therapy through ablation of the peripheral avascu-
The association of ROP and O2 became clear after 10,000 lar retina (4). It defined threshold disease as retinal neovas-
babies had vision loss in the 1940s due to aggressive O2 cularization (stage 3 ROP), in 5 contiguous or 8 total clock
therapy. In 1956, clinical trials demonstrated 3× risk of hours and plus disease, and it determined that cryotherapy
ROP in babies with O2 supplementation (3). The incidence for this level of disease decreased blindness approximately
of ROP decreased after O2 therapy was strongly discour- 25%. Since the CryoROP study, despite the absence of large
aged in the late 1950s, but NICU mortality increased multicenter, randomized trials, laser therapy has largely
concurrently. replaced cryotherapy. A significant caveat of the CryoROP
The intrauterine environment has a PaO 2 of 22 to trial was that it was not directed toward determining differ-
24 mm Hg, whereas the human adult has an arterial PaO2 of ent clinical levels of disease progression and lumped zone 1
70 to 90 mm Hg. In essence, the intrauterine environment is disease infants with zone 2 disease infants. It is now clear that
relatively hypoxic. In addition, many very low–birth-weight zone 1 disease, since by definition has a larger proportion of
premature babies have pulmonary disease that requires avascular to vascularized retina and occurs in the most imma-
hyperoxic therapy for survival. This hyperoxic environment ture of babies, has a worse prognosis than zone 2 disease and
provides retinal oxygenation of the thin premature avascular therefore requires earlier intervention. The Early Treatment
retina through the subjacent choriocapillaris and abolishes of Retinopathy of Prematurity Study (ETROP) trial demon-
the hypoxic-driven VEGF gradient required for normal retinal strated that simpler and earlier guidelines for therapy, based
vascular development. mostly on the presence of plus disease (retinal blood vessel
dilation and tortuosity at the posterior pole) and/or neovas-
cularization at zone 1, led to improved outcomes (5). Plus
Ph ase 2 o f ROP: Re t in al Ne o vascularizat io n
disease in particular has become the main focus of clinical
Weeks after birth, when growth of the eye and retina increase determination of vascularly active ROP disease. Despite a
oxygen demand and the choriocapillaris cannot supply degree in subjectivity is accepted in the diagnosis of plus dis-
enough oxygen to keep up with this increased demand, the ease, the biological implication is clear. Since the junctional
peripheral avascular and ischemic retina becomes hypoxic. neovascular ridge acts as an arteriovenous shunt, increased
While the initial phase of ROP depended on decreased shunting through the ridge increases retinal venous blood
VEGF production, the second phase of ROP is caused by flow and congestion. Venous congestion at the posterior pole
pathologically high levels of VEGF secondary to peripheral can then be used as a clinical surrogate for determining the
retinal hypoxia. As in other forms of proliferative retin- magnitude of shunting through the ridge and likelihood of
opathies, such as diabetes, the increased concentration of progression of the disease.
CHAPTER 26 ■ Retinopathy of Prem aturity and Pediatric 25-Gauge Vitrectom y 227

The single important scenario where the clinician should Techniques


not wait for overt plus disease to recommend peripheral laser
retinal ablation is the infant with zone 1 disease. Zone 1 The authors perform their laser therapies with the near-
disease can progress through stages that are not the typical infrared 810-nm laser. The advantage of the near-infrared laser
clinical ROP stages: (a) line, (b) ridge, (c) neovascularization. is decreased absorption through blood vessels in the tunica
Particularly, the neovascularization, rather than growing in a vasculosa lentis, which can potentially lead to cataract for-
perpendicular plane to the retina, can grow along the retinal mation. The goal of therapy is near confluent laser to all
surface and can be very difficult to diagnose. The clinician the avascular retina. It is imperative to perform 360-degree
should always follow the retinal vessels from the nerve to indirect ophthalmoscopy at the end of the laser treatment
the avascular retina and look for the normal dichotomous to confirm that all the avascular retina is treated and no
branching. Nondichotomous branching, with vessels grow- gaps in the laser are present. The sclera depressor is used
ing in a different direction other than centrifugally in the both to manipulate the globe and rotate it to the required
posterior retina, often signifies retinal neovascularization. direction and to depress and expose the far peripheral retina
The speed of progression of disease from zone 1 with flat for laser treatment. Scleral depression should be gentle to
neovascularization to plus disease and later to traction retinal decrease oculocardiac reflex and progressive media opaci-
detachment can be very fast, often taking 2 to 3 weeks. If the fication during the laser. The authors rarely need to repeat
clinician fails to diagnose the presence of nonelevated neo- laser treatments after a single session of near confluent laser.
vascularization and waits for the development of plus dis- The exception is APROP, where the extent of the avascular
ease in these infants with posterior disease, peripheral laser retina can be covered by the nonelevated neovasculariza-
ablation may not stop the progression of retinal detachment. tion. These infants require reexamination a week after the
This is the previously called “Rush disease,” which implies a laser therapy, since regression of the neovascularization can
retrospective diagnosis of fast progression to retinal detach- expose untreated avascular retina that may require ablation.
ment. The concept that the clinician should now have is Postoperatively, the authors treat all eyes with topical antibi-
aggressive posterior ROP (APROP), which is characterized otics and steroids (Tobradex) and mydriatics (homatropine
by severe vessel immaturity (zone 1 or posterior zone 2) with 2%) for 2 weeks.
flat neovascularization. In contrast to the retrospective “Rush
disease,” APROP has prognostic implication and mandates Man ag e m e n t o f St ag e 4a Re t in o p at h y
immediate laser ablation to the avascular retina. o f Pre m at urit y
The concept of lens-sparing vitrectomy for retinal detach-
Lase r Tre at m e n t fo r Re t in o p at h y ments in ROP that spare the macula (stage 4a ROP)
o f Pre m at urit y has been popularized in recent years, with the goal of
improved visual outcomes (6). The authors maintain a
Indica tions
sense of caution toward stage 4a vitrectomy. The authors
As mentioned previously, the indication for laser treatment feel that many eyes that may have good visual poten-
in ROP is zone 2 disease with plus disease (regardless of the tial without vitrectomy are being operated on and that
extent of neovascularization) and zone 1 neovascularization, the risks of vitrectomy, namely, iatrogenic retinal tears
regardless of the presence of plus disease. with rhegmatogenous retinal detachments and systemic
complications from general anesthesia in these delicate
infants—including death, should not be minimized. In
Anesthesia Considera tions addition, many inexperienced surgeons are feeling pushed
The authors prefer to do laser treatments with light, moni- to recommend surgery for 4a ROP.
tored intravenous sedation, and analgesia with opiates
and without endotracheal intubation in the NICU when-
Issues with Exuda tive Versus
ever possible. Efficient laser techniques are important to
Tra ctiona l Retina l Deta chment Dia gnosis
decrease the duration of treatment and likelihood of sys-
temic problems during laser therapy. Unfortunately, often Whereas tractional retinal detachments are the typical and
the babies require intubation due to the severity of their most ominous type of detachment in ROP, not infrequently
cardiopulmonary disease and intraoperative oxygen desat- will exudative retinal detachments be observed. These
uration and bradycardia. Still, in many babies, intubation detachments are convex in appearance and, in ROP, tend to
can be avoided if the laser therapy is started and tolerance form in the peripheral retina, below areas of active plus dis-
of the procedure is determined. The authors require all ease. It is often very difficult to determine if a small amount
babies to be either in the operating room with an experi- of subretinal fluid represents early tractional 4a detachment
enced pediatric anesthesiologist or at the NICU, with an or exudative detachment. Exudative detachments do not
available ventilator at bedside and constant supervision by require surgery, since they are self-limited by their nature as
the neonatology team. the vascular activity of the disease subsides.
228 SECTION IV ■ Disease-Sp ecific Managem ent

Progression of 4a Retina l Deta chment The authors currently do not recommend vitrectomy
for total closed-funnel retinal detachments with epiretinal
The natural history of stage 4a retinal detachments is not membranes. Recent open-funnel detachments in which
clearly established in the literature. There are 4a detach- epiretinal membrane dissection is not required and careful
ments that can be nonprogressive and do not require sur- vitrectomy can relieve all transvitreal traction, on the other
gery. It is likely that earlier gestational age at the time of hand, the authors do think should be operated on.
diagnosis of tractional 4a retinal detachment as well as active
vascular dilatation with plus disease at the time of diagnosis
may be associated with the higher likelihood of progression Re t in o p at h y o f Pre m at urit y Lase r Failure s
to macular detachment. The authors’ anecdotal experience makes them believe that
The authors do not recommend early stage 4a vitrec- true ROP laser failures, that is, after timely and adequate
tomy for all infants. They advocate close observation upon laser therapy by an experienced surgeon, are uncommon.
the diagnosis of peripheral retinal detachment, and they only Although severe immaturity with zone 1 disease (APROP)
perform vitrectomy in cases where the tractional detachment can progress to retinal detachment despite well-performed
is progressive and threatens the macula. As in all their pedi- laser, many cases of laser failure are due to late treatment,
atric vitrectomies, they perform 25-gauge techniques. insufficient treatment, or both. The likelihood of “laser fail-
ure” is very likely low when laser is administered before
Man ag e m e n t o f 4b Re t in o p at h y significant growth of neovascularization, since ROP traction
o f Pre m at urit y detachment progression is dependent on the contraction of
preexisting extraretinal proliferation, as in diabetic traction
The most important indication for vitrectomy in ROP is reti- retinal detachments. In the authors’ view, the main reasons
nal detachment with macular involvement (stage 4b ROP). for laser failure are (a) lack of adequate follow-ups during the
As mentioned previously, the authors perform all their sur- screening stage, for example, inpatients lost during hospital
geries using 25-gauge techniques. The technical details will transfers and outpatients not brought by parents to clinic
be discussed later in this chapter. visits as scheduled, and (b) misdiagnosis of APROP. This
leads the authors to the discussion of “The ROP Service.”
Man ag e m e n t o f St ag e 5 Re t in o p at h y
o f Pre m at urit y Th e Re t in o p at h y o f Pre m at urit y Se rvice
The most important problem with total retinal detachment Since most failures of therapy are caused by nonpathological
(stage 5 ROP) is the poor visual prognosis of these patients. causes, the best protection against ROP blindness is a well-
The senior author has published a large series (7) of stage 5 run ROP service.
vitrectomy that has determined very poor visual outcomes
(only 4% of patients have final visual acuity 5/200 or bet-
Retinopa thy of Prema turity Service
ter). Although these data largely preceded the introduction
of peripheral retinal ablation and had a high proportion of An ROP service should have these characteristics:
vascularly active eyes, they underscore the difficulties of suc-
1. Multiple physicians trained in screening and laser
cessful functional retinal reattachment. A number of problems
therapy, to provide adequate cross-coverage and be
plague vitrectomy for total retinal detachments in ROP: (a)
able to provide timely therapy regardless of an indi-
high frequency of postoperative epiretinal reproliferation after
vidual physician’s schedule
epiretinal membrane peeling, (b) diffuse retinal foreshortening
2. At least one or two retina surgeons knowledgeable in
after infantile retinal detachment (maturation in the detached
medical therapy and surgical therapy of ROP
configuration), (c) high likelihood of surgical failure after iat-
3. Open communication with the neonatology services
rogenic retinal breaks, and (d) amblyopia ex anopsia, since
of all NICUs served by the ROP service
retinal reattachment may not occur until after an interlude of
4. Ancillary staff that keeps track of all babies in a ser-
detachment during a critical cortical developmental period.
vice. Again, overlapping and redundant tracking
The issue of lens-sparing versus lens removal vitrec-
lists decrease the likelihood of loss of follow-ups
tomy depends more on the anatomic configuration of the
5. Adequate coordination between outpatient clinics
retinal detachment and the surgical space available between
and neonatology wards
the lens and the retina for successful removal of all traction.
6. Adequate attending physician supervision of all
In other words, if the space allows for safe entry of the sur-
these activities
gical instruments and accomplishment of all surgical goals
without removal of the lens, the authors certainly avoid per- Unfortunately, this level of workforce coordination is very
forming a lensectomy. Unfortunately, certain retinal detach- difficult to provide in many midsized or smaller cities in the
ments simply are not amenable to surgical repair without United States and almost impossible in many developing
removal of the lens. countries. The authors run an ROP service that consists of
CHAPTER 26 ■ Retinopathy of Prem aturity and Pediatric 25-Gauge Vitrectom y 229

seven NICUs spread in multiple hospitals in the metropolitan adequate ROP management, and these costs are significantly
Memphis region, one designated attending surgeon coordi- lowered by using intravitreal injection therapy. Still, there are
nating all activities, two surgeons experienced in pediatric ret- many caveats of neonatal anti-VEGF therapy: (a) potential
ina, screening services facilitated by two vitreoretinal surgery systemic side effects and (b) potential for rapid progression to
fellows, one board-certified pediatric ophthalmologist with traction detachment as in diabetic retinopathy.
ROP expertise, and two secretarial staff assisting in updating
the ROP rounding list and constantly communicating with
Man ag e m e n t o f t h e Ch ild wit h
the NICUs and outpatient parents. The authors are proud
In ad e q uat e Pre o p e rat ive Lase r
to say that despite the large size of their service, they only
need to perform one or two ROP vitrectomies a year from the As suggested previously, many infants who come to an ROP
local NICU population that they follow. On the other hand, referral practice have inadequate laser at the time of exami-
the authors perform more vitrectomies on patients from the nation, often with coexisting retinal detachment. This is one
hospitals outside their metropolitan region. of the most difficult situations in a referral practice, since
there are no large studies that demonstrate the best course
of action. The authors’ approach, whenever possible, is to
Te le m e d icin e in Re t in o p at h y o f Pre m at urit y
perform laser therapy to any part of the avascular retina that
The development of the Retcam system for pediatric retinal is still attached to attempt to decrease the vascular activity of
photography has kindled interest in the idea of telemedi- the eye and perform the vitrectomy 4 to 7 days following the
cine in ROP precisely to increase the coverage area of ROP laser. This is another setting in which combination therapy
services from metropolitan regions to hospitals outside of with anti-VEGF medications can provide a way to stop the
these regions where skilled and willing ophthalmologists are progression of the disease.
difficult to find. In the authors’ view, the most appropriate
application for ROP telemedicine is screening of neonates
in satellite locations (suburban or rural) too far from a cen- 25-GAUGE PEDIATRIC VITRECTOMY
tralized ROP service to provide a skilled examiner but close
enough for a timely transfer of a patient in need of laser ther- After the discussion of ROP, the authors will now focus on
apy. After all, the diagnosis of prethreshold ROP is useless if the technical aspects of pediatric vitrectomy. Since the incor-
an ophthalmologist skilled in ROP laser therapy is immedi- poration of 25-gauge technique as the authors’ nearly exclu-
ately unavailable. A secondary application of Retcam retinal sive system for adults, they have come to adopt 25-gauge
imaging is medicolegal protection by photographic docu- techniques exclusively in all pediatric vitrectomies, from
mentation of ROP pathology. As the technology decreases in ROP to congenital cataracts and trauma. There are certain
size and technical difficulty and costs of the equipment drop, technical considerations that need to be kept in mind when
the authors may see a stronger drive toward telemedicine in using 25-gauge technique in children that are not relevant to
ROP using skilled technicians and nurses in the NICU and adults. On the other hand, the same benefits that 25-gauge
centralized imaging interpretation centers. surgery offers adult patients are applicable to pediatric vit-
rectomies. Particularly, the lack of postoperative pain and
ocular surface discomfort is highly desirable in children.
An t i–Vascular En d o t h e lial Gro wt h Fact o r
Th e rap y in Re t in o p at h y o f Pre m at urit y
In t ro d uct io n o f Tro car Can n ulas
Since the introduction of bevacizumab into the retina surgery
field, the question of anti-VEGF therapy in ROP has been The elasticity of the pediatric sclera makes the introduction
entertained. At the time of this writing, only anecdotal reports of the 25-gauge trocar cannulas significantly more difficult
are available that suggest that bevacizumab intravitreal injec- than in adults. If any anterior segment surgery is planned in
tions in ROP are not associated with grossly obvious compli- conjunction with the vitrectomy surgery, the authors prefer
cations and may have adequate anatomic outcomes (8). While to preplace the inferotemporal infusion cannula prior to the
the jury is still out, the main issue to ponder is: Is there a first anterior segment incision. Since vitreous infusion may
need for anti-VEGF therapy given the good outcomes with create “positive pressure” to the anterior segment surgeon, the
timely and adequate laser therapy? The authors believe that authors either place a plug on the cannula or introduce the
there is a need. As mentioned previously, laser therapy, despite infusion system in the off position into the infusion cannula.
being successful, requires a highly skilled surgeon and has This avoids the introduction of the first 25-gauge cannula into
a very short window of opportunity to stop the progression a soft eye, which may be very difficult.
of ROP. If anti-VEGF therapy decreases the complexities of Whereas in adults the authors feel it is very important
therapy and expands the window of opportunity for interven- to introduce the cannulas in an oblique orientation, this
tion, many more infants could be properly treated who are type of entry may be too difficult on the pediatric sclera. The
not being treated properly today. In addition, the high costs authors prefer to introduce all 25-gauge trocar cannulas near
of lasers are barriers of entry in many parts of the world to perpendicularly to the sclera in pediatric cases. The inherent
230 SECTION IV ■ Disease-Sp ecific Managem ent

elasticity of the sclera actually closes the sclerotomies faster instrument. The surgeon can hold a plano lens on his free
in children than in adults, and the authors have never seen hand, and coaxial microscope illumination can be used to
choroidal effusions in pediatric vitrectomies during the post- see the vitreous anatomy, given the small size of the eye. The
operative period. lens has to be angled to direct the reflected light from the
A problem inherent to children is the lack of space in the microscope away from the surgeon’s view.
interpalpebral fissure for the external profile of the 25-gauge can-
nulas. This is usually managed by placing all three cannulas near
the horizontal meridian. In very small infants and those with MANAGEMENT OF EPIRETINAL
eyelid malformations, one may need to do a small canthotomy MEMBRANES
(without cantholysis) in the temporal raphe. If done properly
with a single snip of the Wescott scissors, adequate space can In ROP as well as in rhegmatogenous detachments with
be achieved with minimal difficulties. The canthotomy can be PVR, the postoperative reproliferation rate is very high, and
approximated at the end of the case with a single absorbable plain this limits anatomic postoperative outcomes. The surgeon
gut suture and is rarely noticeable after 2 weeks from surgery. should use a minimalist approach to surgery and limit the
The thin pediatric sclera does not provide enough grasp epiretinal dissection to the minimum required to relieve
of the infusion cannula to prevent torquing of the cannula retinal traction. The more a surgeon dissects, the higher the
from the weight of the infusion line. To prevent the rotation stimulus for postoperative reproliferation.
of the cannula from injuring the lens or the peripheral retina,
the surgeon should hold the cannula with bishop forceps
until the assistant secures the infusion line with Tegaderm
SCLERAL BUCKLING
in its appropriate position. For the same reason, the surgeon
The authors do not advocate scleral buckling in ROP. Given
should take great care when rotating the eye, since the eyelid
the pathophysiology of internal vitreous traction, the best
can push on the infusion cannula and alter its orientation
approach is to relieve the traction through vitrectomy tech-
with respect to the sclera, pushing it into the suprachoroidal
niques when warranted.
space, subretinal space, or the lens.
On the other hand, scleral buckles are a good way to
treat uncomplicated pediatric retinal detachments with inf-
25-Gaug e Surg e ry Wit h o ut Can n ulas erotemporal dialyses or other anterior retinal pathology. The
low likelihood of cataract formation after buckling makes
If the surgeon is approaching the anterior vitreous in a lens-
this technique attractive for these cases. On the other hand,
sparing case (so that the contralateral entry cannula cannot
if PVR is present, the authors proceed with vitrectomy with-
be used to vitrectomize the opposite anterior vitreous with-
out scleral buckling.
out lens injury), the internal profile of the 25-gauge can-
nula system may preclude the ipsilateral approach with the
vitreous cutter to the anterior vitreous. Some surgeons have References
recommended using 20-gauge techniques in these patients 1. Bergh T, Ericson A, Hillensjö T, et al. Deliveries and children
to avoid the internal profile of the 25-gauge cannula. This born after in-vitro fertilisation in Sweden 1982–95: a retrospec-
opinion does not take into consideration that the 25-gauge tive cohort study. Lancet 1999;354(9190):1579–1585.
cannulas are not required for proper functioning of the vit- 2. Kimura H, Weisz A, Kurashima Y. Hypoxia response element of
the human vascular endothelial growth factor gene mediates tran-
rectomy probe. The real need for the cannula is to perform scriptional regulation by nitric oxide: control of hypoxia-inducible
transconjunctival vitrectomy. On the other hand, after per- factor-1 activity by nitric oxide. Blood 2000;95(1):189–197.
forming a conjunctival incision, the 25-gauge cutter tip can 3. Kinsey VE. Retrolental fibroplasia: cooperative study of ret-
enter a sclerotomy without the cannula and be used for vit- rolental fibroplasia and the use of oxygen. Arch Ophthalmol
rectomy in a similar way as larger 20-gauge instruments. In 1956;56:481–543.
4. Group CF. Multicenter trial of cryotherapy for retinopathy of pre-
other words, if the cannula is in the way of the tip of the maturity: preliminary results. Arch Ophthalmol 1988;106:471–
cutter, then the surgeon can remove the cannula, perform 479.
a small conjunctival incision to expose the sclerotomy, and 5. Group ET. Final results of the early treatment for retinopathy of
introduce the cutter and endoilluminator through the scle- prematurity (etrop) randomized trial. Trans Am Ophthalmol Soc
rotomy sites. Closure will only then require reapproximation 2004;102:233–250.
6. Capone A. Lens-sparing vitreous surgery for tractional stage 4A
of the conjunctiva without sclera suturing. retinopathy of prematurity retinal detachments. Ophthalmology
2001;108(11):2068–2070.
7. Cusick MM, Charles MK, Agrón EM, et al. Anatomical and visual
Th re e -Po rt Ve rsus Two -Po rt Vit re ct o m y results of vitreoretinal surgery for stage 5 retinopathy of prema-
Whenever the space allows for three-port vitrectomy, the turity. Retina 2006;26(7):729–735.
8. Kong L, Mintz-Hittner HA, Penland RL, et al. Intravitreous beva-
authors prefer to perform surgery in this way. If needed, the cizumab as anti–vascular endothelial growth factor therapy for
surgery can be performed by two-port techniques: a cannula retinopathy of prematurity: a morphologic study. Arch Ophthal-
for infusion and a superotemporal cannula for vitrectomy mol 2008;126(8):1161–1163.
C H AP TER
27
PEDIATRIC TRACTION
RETINAL DETACHMENTS

Diverse disease processes may create traction Surg ical Se q ue n ce an d Te ch n iq ue s


retinal detachments in the pediatric age group.
Microincisional vitrectomy with 25-gauge or possible
Special approaches are required to manage
23-gauge technology is ideal for PFV. The vitrectomy instru-
these difficult problems. These young patients
ment is introduced into the lens substance (Fig. 27.1),
have many years ahead of them and require the most aggres-
except in the rare instance that there is a clear lens. If the
sive attempts at restoration of vision. Patients in the pedi-
lens is clear, occasionally it is possible to introduce just the
atric age group having retinal detachments of several years’
scissors through the pars plana without infusion and to
duration can have remarkable visual recovery, and this
transect the membrane behind the lens, which can allow
emphasizes the need to proceed with such cases. By con-
the traction detachment to resolve. More commonly, the
trast, the very young patient, especially with retinopathy of
lens is cataractous or a large retrolental membrane is pres-
prematurity, is a high medical risk patient requiring careful
ent, requiring lens removal. The vitrectomy instrument is
assessment of the risk-benefit ratio of surgery. From the ocu-
used to remove the retrolental membrane centrally, being
lar standpoint, unilateral disease in the pre-6-year-old patient
careful not to amputate a ciliary process and thus cause
has an extremely high incidence of amblyopia, diminishing
bleeding. After a large opening is made, almost to the tips
the visual impact of successful surgery.
of the ciliary processes, the curved, 25-gauge scissors are
introduced and are used to cut between ciliary processes in
PERSISTENT HYPERPLASTIC order to segment the ring. If this is not done, the circum-
PRIMARY VITREOUS, PERSISTENT ferential traction will keep the ciliary processes detached in
the future.
FETAL VASCULATURE
As the anterior portion of the stalk extending poste-
riorly is resected, bleeding will frequently occur from the
Persistent hyperplastic primary vitreous is also known as per-
remnants of the hyaloid artery system. This should be
sistent fetal vasculature (PFV). It is usually a unilateral phe-
treated with bipolar diathermy. Although the stalk can be
nomenon accompanied by a smaller eye (1,2). The almost
resected midway in the vitreous cavity, it is usually necessary
uniform incidence of amblyopia means that these cases should
to delaminate the posterior termination of the stalk away
be operated on early (3). An additional argument for early sur-
from the retinal surface. Care should be taken to avoid the
gery is the prevention of long-term traction detachment and
retina, which can be pulled up into the central portion of the
pupillary block chamber (4,5). This condition is usually recog-
stalk around the optic nerve area. If there is a small “table-
nized early in life, and the patient should be operated upon if a
top” traction detachment surrounding the optic nerve, the
traction detachment is recognized, the cataract is sufficient to
25-gauge fine, curved scissors should be used to delaminate
cause visual loss and amblyopia, or there is shallowing of the
the membrane.
anterior chamber secondary to pupillary block. The embryo-
logical explanation for the syndrome is the lack of regression
of the primary vitreous and hyaloid vasculature, although the
Re sult s
primary cause is still unclear. Bilateral cases in males are usu-
ally associated with Norrie’s syndrome. Norrie’s cases should Anatomic results are excellent in this group, well over 95%.
not be operated on because the retina is dysplastic and the Bleeding and postoperative retinal detachment are exceed-
vitreoretinal interface cannot be delineated at surgery. ingly rare.

231
232 SECTION IV ■ Disease-Sp ecific Managem ent

Fig u re 2 7 .1 ■ The vitrectom y


instrum ent is used to rem ove the
lens and retrolental m em brane.

Amblyopia is such a frequent accompaniment of this of this disease in a nonrhegmatogenous fashion. When this
disorder that aggressive treatment by early surgery, early occurs, vitrectomy is necessary.
contact lens fitting, and patching of the other eye is neces-
sary. On occasion, a patient will achieve an excellent visual
Surg ical Se q ue n ce an d Te ch n iq ue s
result, but amblyopia is more common. If aggressive contact
lens fitting and patching are not done, amblyopia is the uni- Standard 25-gauge cannula placement is performed. The vit-
form result. rectomy instrument is introduced superotemporally and the
endoilluminator superonasally. The vitreous is usually coni-
cally shaped and the posterior vitreous cortex (PVC) is con-
TOXOCARA CANIS tinuous with the tract (Fig. 27.2). Although a casual examiner
would think that this tract progresses through the vitreous
The parasite, Toxocara canis, can migrate from the choroidal cavity, in fact, posterior vitreous detachment usually occurs
vessels through the choroid and retina into the vitreous cav- with the proliferation on the vitreous surface. If resection of
ity (6). As it migrates through the eye, it creates an intense the dense tract alone is performed, the clear vitreous may cause
inflammatory reaction, which may simulate endophthalmi- the traction detachment to remain. For this reason, the vitrec-
tis or severe uveitis (7). After this initial phase, a fibrous tomy is completed first and then attention is directed to the
proliferation, more extensive than that seen with vascular stalk. Care should be taken to avoid a steep radial fold of retina
retinopathy, enters the eye. It may come from both the underlying the stalk, which is present in many cases. Scissors
posterior entrance site of the parasite and the pars plana segmentation and delamination techniques should be utilized
exit site. If this membrane, created by the inflammatory to allow the release of retinal traction (Figs. 27.3–27.5). The
reaction, does not cause traction detachment, it is not nec- principles are similar to those used for traction detachments
essary to operate. If there is a small traction detachment from diabetic retinopathy, although the proliferation is much
away from the macula, this also can be followed up without more dense and localized. Bleeding may occur from this stalk
surgery. and can be treated with bipolar diathermy, paying close atten-
These cases have a high incidence of late rhegmatog- tion to avoid the retina and optic nerve.
enous detachment secondary to long-standing traction. Scleral buckling is not utilized in these young patients
Although scleral buckling may be able to reattach some of because of the possibility of later intrusion of the encircling
these detachments, vitreous surgery with resection of the element. If a rhegmatogenous component is present, it must
stalk might be beneficial in certain cases. Occasionally, the be managed by internal drainage of subretinal fluid, fluid-
macula will become detached in the relatively early stages gas exchange, and laser endophotocoagulation.
CHAPTER 27 ■ Pediatric Traction Retinal Detachm ents 233

Fig ure 2 7 .2 ■ The vitreous is usually conically shaped and the PVC is continuous with the tract.

Fig ure 2 7 .3 ■ Multip le radial cuts are m ade in an inside-out direction.


234 SECTION IV ■ Disease-Sp ecific Managem ent

Fig ure 2 7 .4 ■ Scissors are used to delam inate the resulting segm ents.

Fig ure 2 7 .5 ■ Curved scissors are used to rem ove the d elam inated seg m ents.
CHAPTER 27 ■ Pediatric Traction Retinal Detachm ents 235

Re sult s 2. Goldberg MF. Persistent fetal vasculature (PFV): an integrated


interpretation of signs and symptoms associated with persistent
The visual outlook primarily depends upon the involvement hyperplastic primary vitreous (PHPV). LIV Edward Jackson
of the macula in the full-thickness chorioretinal destructive Memorial Lecture. Am J Ophthalmol 1997;124(5):587–626.
process. If the macula is secondarily detached but has not 3. Hunt A, Rowe N, Lam A, et al. Outcomes in persistent hyper-
plastic primary vitreous. Br J Ophthalmol 2005; 89(7):859–863.
been invaded by the parasite, excellent visual recovery is 4. Gass JDM. Surgical excision of persistent hyperplastic primary
possible. Fortunately, recurrence of postoperative inflamma- vitreous. Arch Ophthalmol 1970;83:163.
tion is not a problem in these cases. 5. Smith RE, Maumenee AE. Persistent hyperplastic primary vitre-
ous. Trans Am Acad Ophthalmol Otolaryngol 1974;78:911.
6. Hogan MJ, Kimura SJ, Spencer WH. Visceral larval migrans and
References peripheral retinitis. JAMA 1965;194:1345.
7. Stewart JM, Cubillan LD, Cunningham ET Jr. Prevalence, clini-
1. Yanoff M, Fine BS. Ocular pathology-a text and atlas. Hagerstown, cal features, and causes of vision loss among patients with ocu-
MD: Harper & Row, 1975:698. lar toxocariasis. Retina 2005;25(8):1005–1013.
C H AP TER
28
MANAGEMENT OF VITREORETINAL
COMPLICATIONS ASSOCIATED
WITH KERATOPROSTHESIS

The management of severe anterior segment and frequently require vitrectomy techniques for removal of
disease and intractable corneal opacities has the retroprosthetic membrane.
evolved rapidly in the recent years. Whereas in In addition, since the Boston keratoprosthesis never
the late 1990s there was a push for limbal stem integrates with the host cornea or sclera, there will always be
cell transplantation in conjunction with aggressive systemic a permanent open wound around the keratoprosthesis that
immunosuppression, the complications and practical diffi- may be the route for bacteria to develop endophthalmitis.
culties associated with immune therapy have decreased the The Alphacor keratoprosthesis and the osteo-odontogenic
interest in this technique. Keratoprosthetic devices have been keratoprosthesis designs are attempts to increase integra-
in development for decades. Claes Dohlman, at the Massa- tion of the device. Dohlman has noticed that placement of
chusetts Eye and Ear Infirmary, has developed and perfected a contact lens over the keratoprosthesis and chronic topical
the Boston keratoprosthetic device. His relentless pursuit of antibiotics decreases endophthalmitis rates.
technical improvements of the device has allowed the recent Glaucoma is very common after keratoprosthesis, and
popularization of the Boston keratoprosthesis and its more many surgeons recommend primary placement of Ahmed
common utilization in a variety of disorders of the cornea valve glaucoma implants (3). In eyes that have undergone
and anterior segment (1). iridectomy, lensectomy, and keratoprosthesis that also have
One may classify vitreoretinal disorders associated with an Ahmed valve, vitreous occlusion of the tip of the Ahmed
keratoprosthesis implantation into valve can require emergent vitrectomy for relief of the acute
glaucoma attack.
1. Preexisting vitreoretinal conditions
3. Disorders associated with the surgical technique of ker-
Keratoprosthesis can be associated with various vitreoretinal
atoprosthesis implantation
disorders that may require vitreous microsurgery (2). The eyes
that require keratoprosthetic implantations have commonly Keratoprosthesis implantation is a difficult procedure that
been operated on multiple times unsuccessfully and have may require, beyond replacement of the cornea, total iridec-
sustained long-standing inflammation. These eyes may har- tomy, lensectomy, or intraocular lens explantation and open-
bor preexisting retinal conditions that may be unmasked by sky vitrectomy. The iridectomy may cause intraoperative
the clear optics of the keratoprosthetic device, such as dense and postoperative vitreous hemorrhage, and the vitrectomy
epiretinal membranes, retinal detachment, or macular holes, and lens removal may cause inadvertent retinal tears with
that may require surgery with the keratoprosthesis in situ. subsequent retinal detachment. Early postoperative vitreous
hemorrhages are often difficult to manage, since although
2. Disorders secondary to the presence of the keratopros-
the most likely cause for the hemorrhage is iridectomy in
thetic device
a hypotonic eye, one cannot usually rule out intraoperative
Keratoprosthesis can commonly present with retroprosthetic retinal tear until surgery is undertaken. Another surgical
membranes that severely limit visual outcomes. These mem- complication of keratoprosthesis implantation is intraopera-
branes are usually more frequent and severe in children but tive suprachoroidal hemorrhage. Since these eyes undergo
can also present in adults. Retroprosthetic membranes are open-sky surgery, choroidal expulsive hemorrhages are a real
usually too thick and adherent to be amenable to YAG laser risk. Unfortunately, it is almost impossible to repair these
236
CHAPTER 28 ■ Managem ent of Vitreoretinal Com plications Associated with Keratoprosthesis 237

eyes even if the surgeon is able to suture the keratoprosthesis is the target, then cannula entry should be performed 3.75
quickly enough to prevent expulsion of the intraocular con- to 4 mm posterior to the edge of the optic (3 mm in case of
tents, since the retina and anterior vitreous can become smaller pediatric back plates). This entry location likely cor-
adherent to the corneal wound and the back plate of the responds to pars plicata rather than pars plana. The authors
keratoprosthetic device, making successful retinal detach- do not recommend more posterior entry since the surgeon
ment repair almost impossible. has no assurance of perfect centration of the keratoprosthetic
device. If the device is decentered, posterior entry through
the anterior retina can be unfortunately performed. If the
TECHNICAL CONSIDERATIONS surgeon can visualize the edge of the back plate, then direct
REGARDING OFFICE EXAMINATIONS measurement from this location overrides the previous dis-
IN KERATOPROSTHESIS PATIENTS cussion. The authors do not have experience with translid
keratoprostheses but would recommend a similar analysis
Office examinations of patients with keratoprosthesis can be for sclerotomy placement if confronted with the situation.
challenging. While examination of the posterior pole with The authors find the 25-gauge system excellent of surgery
an indirect ophthalmoscope can usually be accomplished, in keratoprosthesis cases, since it avoids conjunctival dissec-
examination of the retinal periphery may be more difficult. tion on these eyes that have preexisting ocular surface scar-
The authors prefer the use of high plus wide-angle lenses ring. Transconjunctival sclerotomy closure with a single 8-0
on the slit lamp for examination of the retinal periphery on Biosorb suture may be needed at the end of the surgery since
patients with keratoprosthesis, since the oblique slit beam the conjunctiva rarely covers the sclerotomies.
reduces reflections and glare from the surface of the ker-
atoprosthesis. If B-scan ultrasonography is needed due to Pe rip h e ral Re t in al Visualizat io n
vitreous hemorrhage, the examiner may need to place the an d Man ag e m e n t
transducer directly adjacent to the keratoprosthesis and
perform transscleral imaging, avoiding artifacts from the The configuration of the optic is a cylinder of 3 mm diam-
keratoprosthesis. eter, with a thickness of slightly greater than 3 mm. Direct
visualization of the posterior pole structures is usually excel-
lent with the eye on primary position. Rotation of the globe,
VITRECTOMY IN though, creates tilting of the optical cylinder and decreases
KERATOPROSTHESIS PATIENTS the optical aperture for visualization. In essence, permanent
keratoprosthetic vitrectomies require surgery in the primary
The most significant technical considerations while doing position. Given this constraint, contact-based wide-angle lens
vitrectomies in eyes with keratoprosthesis are (a) introduc- (Volk) visualization is imperative for peripheral vitrectomy.
tion of 25-gauge trocar-cannula systems, (b) peripheral reti- Even in these conditions, visualization of the far periphery
nal visualization and management, and (c) management of may be very difficult, and this is one of the reasons that rheg-
retroprosthetic membranes. matogenous detachments are difficult to repair. Endoscopic
techniques may be required for complete retinal peripheral
evaluation and laser.
In t ro d uct io n o f t h e 25-Gaug e Tro car Can n ulas
If peripheral visualization impairs adequate manage-
Eyes that require keratoprosthesis may not have clearly iden- ment of a retinal detachment, a useful technique to consider
tifiable limbal structures from which to measure posteriorly is medium-term perfluorooctane with 360-degree peripheral
the location of the pars plana. Entry into the eye should obvi- retinal endophotocoagulation. Perfluoro-octane (PFO) can
ously avoid the anterior retina but is constrained anteriorly be injected over the optic nerve and used to fill the vitreous
by the presence of the back plate of the keratoprosthesis. cavity and reattach the retina as described in Chapter 14. Pos-
The 25-gauge cannula needs to be at least 2 mm posterior to terior retinotomy should be avoided in these eyes. Once the
the edge of the back plate to allow clearance below the back retina is attached, the surgeon can proceed with laser of the
plate, required to approach the posterior side of the optic in retinal periphery circumferentially with at least three to four
case of retroprosthetic membrane removal. The only identifi- rows of confluent laser as far as visualization allows. The PFO
able structure on the anterior segment of keratoprostheses is can then be left in the vitreous cavity for 2 weeks and can be
the edge of the front plate. The back plate measures 8.5 mm later removed. If permanent posterior pole reattachment is
in diameter, but 7-mm diameter back plates are also avail- accomplished, despite peripheral subretinal fluid, this quali-
able for small pediatric eyes. It is important to remember fies as a successful repair in an eye that would otherwise have
that the edge of the back plate is not usually visible, so the proceeded to blindness. The authors discourage the use of sil-
surgeon should estimate its location from the edge of the icone oil in keratoprosthesis retinal detachments. These eyes
anterior optic. Since the radius of the front plate is 2.5 mm, are already prone to glaucoma, intraocular pressure measure-
the edge of the back plate is 1.75 mm posterior to the edge ments are impossible to perform reliably, and the silicone oil
of the optic. If 2 mm clearance posterior to the back plate can prevent fluid egress through the Ahmed valve.
238 SECTION IV ■ Disease-Sp ecific Managem ent

Fig ure 2 8 .1 ■ After an opening


is m ade with an MVR blad e, the
retroprosthetic m em brane can be
rem oved with the vitreous cutter.

Man ag e m e n t o f Re t ro p ro st h e t ic Me m b ran e s keratoprosthetic device. The authors’ goal is to eliminate


postoperative retinal detachments and vitreous hemorrhages
Retroprosthetic membranes are often very thick and adherent
and intraoperative suprachoroidal hemorrhages. Their most
to the posterior surface of the optic. Forceps, scissors, and
recent approach is placement of 25-gauge cannulas and
the vitreous cutter are usually unable to start an opening
anterior vitrectomy (limited by visualization) prior to the
on the membrane. The authors use a 25-gauge needle or
removal of the corneal button. Once the device is in place,
25-gauge microvitreoretinal (MVR) blade with the tip bent
pars plana infusion can be reinitiated to pressurize the globe.
to form a sharp pick that is introduced through the 25-gauge
The authors believe that this technique decreases iatrogenic
cannula. The sharp bent needle tip is then used to engage
retinal tears and decreases the time of globe depressurization
the membrane away from the center of the optic (in case the
and hypotony that can lead to intraoperative hemorrhages.
needle scratches the posterior surface of the device) and an
initial opening on the membrane is created. Once an edge
is found, further removal with forceps, scissors (segmenta- References
tion), or the vitreous cutter can be performed (Fig. 28.1). 1. Dohlman CH, Harissi-Dagher M, Khan BF, et al. Introduction
The authors attempt to remove the membrane beyond the to the use of the Boston keratoprosthesis. Exp Rev Ophthalmol
edge of the optic along the surface of the back plate, with 2006;1(1):41–48.
the goal of decreasing recurrence of the membrane over the 2. Subhransu Ray, Khan BF, Dohlman CH, et al. Management of
vitreoretinal complications in eyes with permanent keratopros-
optical surface of the device. thesis. Arch Ophthalmol 2002;120:559–566.
The authors are currently working with their cornea 3. Netland PA, Terada H, Dohlman CH. Glaucoma associated with
specialist colleagues to refine the initial implantation of the keratoprosthesis. Ophthalmology 1998;105(4):751–757.

.
C H AP TER
29
INADVERTENT PENETRATION
OF THE EYE

Inadvertent penetration of the globe can occur in retrobulbar injection. This position has been shown by
association with many ocular procedures. Most, Grizzard to minimize displacement and increased tension
if not all, of these incidents can be prevented on the optic nerve, which increase the risk of penetration of
with strict attention to technique. The keys to the nerve (4).
prevention are awareness of the factors likely to cause pen- Oversedation is a significant cause of patient motion
etration and constant vigilance. Many factors are associated and inadvertent penetration of the eye during the adminis-
with inadvertent penetration of the eye. They include patient tration of anesthesia. Explanation is frequently better than
movement, posterior staphylomas, myopic eyes, and poor sedation, especially for elderly patients. There is no evidence
technique (1–3). that blunt, so-called retrobulbar needles are safer than the
standard, sharp, 27-gauge, 1.25-inch needles used by the
authors. There is cadaver CT evidence that 1.5-inch needles
OFFICE INJECTIONS are too long and increase the likelihood of optic nerve dam-
age at the apex of the orbit. Sharp needles require less force
Subconjunctival injection in the lower fornix is safer than than blunt needles and are therefore less likely to advance
sub-Tenons (infra-Tenons) or so-called periocular injection. abruptly through the lid septum or cause pain and patient
Subconjunctival injection under the bulbar conjunctiva is movement. The entry point should be at the outer “corner”
potentially dangerous. Myopic eyes are more likely to be of the orbit, not the more traditional junction of the inner
penetrated than emmetropic or hyperopic eyes because of 2/3 and outer 1/3 of the lower lid.
greater ocular length, thin sclera, and staphylomas. Injec-
tions should be performed with patient supine. Viscous
lidocaine provides better anesthesia than topical anesthe- TOXICITY
sia. Both physicians’ hands should be braced against the
patient’s facial bones with the needle entering laterally and Intraocular gentamicin and other aminoglycoside antibiotics
nearly parallel to the lid margins. If the needle is directed are very toxic to the retina (5–7). Lidocaine is apparently
posteriorly, the globe can be penetrated if the patient moves relatively safe but Wydase has been shown to be very toxic
forward. to the retina (8). Many randomized trials have not demon-
strated any beneficial effect of Wydase (9,10). The authors
never use Wydase and suspect that many surgeons are
PRESURGICAL ANESTHESIA unaware of the lack of efficacy and potential hazards and use
this agent on a “routine” basis.
Topical and intraocular anesthesia is rapidly increasing in
popularity for cataract surgery. Although some straightfor-
ward core vitrectomy procedures can be performed with RECOGNITION
topical anesthesia, the extraocular muscles must be blocked
for macular and complex surgery. Peribulbar anesthesia Corneal edema occurs instantaneously when the intraocular
using a blunt, curved, flexible cannula and small conjunc- pressure is elevated to very high levels. Some surgeons have
tival incision are theoretically safer than multiple injec- reported that the patients experience marked pain, anxiety,
tions into the anterior orbital tissue. The patient should be and/or nausea if intraocular injection or penetration of the
directed to position the eye in the primary position during eye occurs. Hypotony is a variable and somewhat delayed
239
240 SECTION IV ■ Disease-Sp ecific Managem ent

finding in these cases. Hemorrhage into the orbital tissue,


SURGICAL INDICATIONS
eye wall, or eye can occur if a scleral laceration or double
penetrating injury occurs.
If vitreous hemorrhage persists over 10 days or if hypocellular
vitreous collagen contraction as evidenced by decreased vitre-
ous mobility, or a fibrous tract, or retinal detachment occurs,
IMMEDIATE ACTION vitrectomy is indicated. If the eye is No Light Perception
Vision (NLP) from optic nerve damage, surgery is not indi-
The fundus should be immediately inspected with the indi- cated. Surgery is not indicated for removal of subretinal blood
rect ophthalmoscope by the operating surgeon if there is or intravitreal anesthetic agents in the authors’ opinion.
any suspicion of inadvertent penetration. If a penetration
is recognized, elective anterior segment surgery should be
cancelled in most instances and a vitreoretinal surgeon Surg e ry Ap p ro ach
contacted immediately. An immediate view of the fundus
Standard vitrectomy methods for trauma are used as previ-
can determine if there has been a single or double pen-
ously described. Retinopexy around the penetration sites is
etration and if the macula, posterior pole, optic nerve, or
only indicated if a retinal break is observed. Exploration of
retinal vessels have been damaged. Although some sur-
the site, cryopexy, or scleral buckling is not required.
geons recommend immediate vitrectomy if an aminogly-
coside antibiotic is injected into the eye, the diffusion rate
is very rapid and it is highly likely that retinal damage will References
occur before a vitrectomy can be accomplished. The laser
1. Edge R, Navon S. Scleral perforation during retrobulbar and
indirect ophthalmoscope (LIO) is ideal to treat the pen- peribulbar anesthesia: risk factors and outcome in 50,000
etration site(s) before diffusion of vitreous hemorrhage consecutive injections. J Cataract Refract Surg 1999;25(9):
can occur. Cryopexy is less desirable than LIO because it 1237–1244.
requires pressure on the eye and a conjunctival incision 2. Ginsburg RN, Duker JS. Globe perforation associated with ret-
and probably causes more reparative scarring. Exploration robulbar and peribulbar anesthesia. Semin Ophthalmol 1993;
8(2):87–95.
of the sclera and suturing of the scleral penetration sites 3. Modarres M, Parvaresh MM, Hashemi M, Peyman GA. Inad-
are not indicated in these cases in the authors’ opinion. vertent globe perforation during retrobulbar injection in high
myopes. Int Ophthalmol 1997–1998;21(4):179–185.
4. Grizzard WS, Kirk NM, Pavan PR, et al. Perforating ocular
DELAYED VITRECTOMY injuries caused by anesthesia personnel. Ophthalmol 1991;
98(12):1757.
5. Campochiaro PA, Lim JI. Aminoglycoside toxicity in the treat-
Although some surgeons recommend immediate vitrectomy, ment of endophthalmitis. The aminoglycoside toxicity study
the authors believe that it is better to delay vitrectomy and group. Arch Ophthalmol 1994;112(1):48–53.
find vitrectomy unnecessary in most cases. Indications for 6. Peyman GA. Aminoglycoside toxicity. Arch Ophthalmol 1992;
vitrectomy include the development of a tract, retinal detach- 110(4):446.
7. Campochiaro PA, Conway BP. Aminoglycoside toxicity- a sur-
ment, or formation of an epimacular membrane. Vitreous vey of retinal specialists. Implications for intraocular use. Arch
hemorrhage should be observed frequently for the develop- Ophthalmol 1991;109(7):946–950.
ment of hypocellular collagen contraction as evidenced by 8. Gottlieb JL, Antoszyk AN, Hatchell DL, Saloupis P. The safety
decreased vitreous mobility. Ultrasonic imaging can deter- of intravitreal hyaluronidase. A clinical and histologic study.
mine the presence of hypocellular collagen contraction via Invest Ophthalmol Vis Sci 1990;31(11) 2345–2352.
9. Bowman RJ, Newman DK, Richardson EC. Is hyaluronidase
decreased vitreous mobility during saccades. B-scan can also helpful for peribulbar anaesthesia? Eye 1997;11:385–388.
determine if retinal and/or choroidal detachment is present. 10. Crawford M, Kerr WJ. The effect of hyaluronidase on peribulbar
Scleral buckling is virtually never indicated. block. Anaesthesia 1994;49:907–908.
C H AP TER
30
MANAGEMENT OF
SUPRACHOROIDAL HEMORRHAGE

Suprachoroidal hemorrhage is a devastating acute lowering of the IOP to atmospheric pressure in the
complication of intraocular surgery. This com- presence of hypertension. The bleeding is probably due to
plication is very difficult to anticipate, prevent, shearing of vessels traversing the suprachoroidal space
and manage (1–5) (Fig. 30.1). Intraoperative caused by scleral stretch. Penetration of the eye is probably
management is complex, as is the decision to intervene in frequent and often unrecognized because of suprachoroidal
the postoperative period. These cases are also called chor- hemorrhage. Hypertension and arterial disease are critical
oidal hemorrhages because blood dissects into the spongy factors in the pathogenesis. Patients should be normotensive
choroidal tissue. They are termed expulsive hemorrhages if during cataract, penetrating keratoplasty, secondary IOL,
the choroid and retina are forced out of the eye by high pres- and filtering procedures. If general anesthesia is being used,
sure in the suprachoroidal space. it is probably advisable to ask the anesthesiologist to use
neuromuscular blockade for open eye procedures to prevent
high blood pressure secondary to “bucking on the tube.”
INCIDENCE OF SUPRACHOROIDAL
HEMORRHAGE
COMPLICATIONS OF
The incidence of suprachoroidal hemorrhage in extracapsu- SUPRACHOROIDAL HEMORRHAGE
lar cataract surgery and phacoemulsification is approximately
0.15%, filtering procedures 0.15%, penetrating keratoplasty Bad outcomes in nonexpulsive cases are usually not directly
0.56%, vitrectomy 0.41%, and the principal author’s vit- due to the hemorrhage but are secondary to retinal detach-
rectomy series is 0.01% (5/26,000). Small-incision cata- ment from hypocellular collagen contraction and adherence
ract surgery does not necessarily reduce the incidence of of vitreous to anterior structures (iris, wound, capsule). Vit-
this complication as the intraocular pressure (IOP) must reoretinal traction increases in the weeks and months after
be reduced to atmospheric pressure during intraocular lens surgery after the choroidal blood absorbs and the buckle-
(IOL) insertion, although it certainly makes wound closure like effect disappears (Fig. 30.2). Many patients suffer optic
faster and safer. Clearly, the duration of low IOP is less with nerve damage secondary to the acute increase in IOP or
phacoemulsification than with intracapsular surgery. Small- possibly high intrasheath pressures.
incision surgery with self-sealing wounds construction facil-
itates rapid wound closure and normalization of the IOP.
Filtering procedures such as trabeculectomy, setons, and ACUTE MANAGEMENT
valves remain a common cause of this complication. Late
hemorrhages are common if antimetabolites (mitomycin) are It is not advisable to open the anterior chamber to implant,
used producing a sustained, very low IOP. reposition, or replace the IOL if a suprachoroidal hemorrhage
occurs. It is better not to cut down on the sclera if a hemorrhage
occurs, but instead, the focus should be on closing the wound
PATHOGENESIS AND PREVENTION with 8-0 nylon sutures as rapidly as possible. A viscoelastic
can occasionally be used to reposition the iris. The surgeon
A key factor in the pathogenesis of suprachoroidal hemor- should close the wound and delay surgery until inflammation
rhages is a high trans–arterial wall pressure gradient due to is gone if a suprachoroidal hemorrhage is recognized.

241
242 SECTION IV ■ Disease-Sp ecific Managem ent

Fig u re 3 0 .1 ■ Sup rach oroid al


h em o rrh ag e p u sh es th e retin a
centrally and anteriorly and com -
presses rem aining vitreous.

INDICATIONS FOR VITRECTOMY hemorrhage is often cited as an indication for vitrectomy.


The authors disagree with this indication and use the indi-
Rhegmatogenous or traction retinal detachment secondary cations described previously. Appositional (kissing) supra-
to hypocellular collagen contraction and adherence of vitre- choroidal hemorrhage is not an indication for surgery in the
ous to anterior structures is the most common indication authors’ opinion. There is no scientific evidence for retinal
for vitrectomy. A flat chamber is also an indication for surgi- detachment due to retinal-retinal adherence. There is usually
cal intervention. An arbitrary duration of the suprachoroidal a layer of vitreous interposed between the retinal surfaces.

Fig u re 3 0 .2 ■ Retin al d etach -


m ent m ay be caused by hypocel-
lular con traction of the vitreous
com bined with adherence to ante-
rior structures.
CHAPTER 30 ■ Managem ent of Suprachoroidal Hem orrhage 243

Fig ure 3 0 .3 ■ Scleral cut-downs


are not required because the blood
d rain s out th roug h p ars p lan a
sclerectom ies during vitrectom y.

retinal breaks and subsequent retinal breaks secondary to


SURGICAL MANAGEMENT
collagen contraction and epiretinal membrane formation.
OF SUPRACHOROIDAL HEMORRHAGE

The infusion should be initiated with a 25-gauge needle


or the long, handheld, angulated, blunt cannula (May) or SUMMARY
the end-irrigating endoilluminator if a 20-gauge case. The
suprachoroidal hemorrhage usually drains through each As in many medical problems, prevention, followed by early
pars plana sclerotomy during the early part of the vitrectomy recognition and conservative management, is the key to
(Fig. 30.3). Scleral cut-downs are not required to drain the reducing bad outcomes from suprachoroidal hemorrhage.
blood and may cause severe acute bleeding. Manipulation of
the sclera with a scleral depressor breaks up clots, allowing
faster egress of blood through the standard pars plana sclero-
References
tomies. Removal of all the blood is not necessary as the blood 1. Chu TG, Green RL. Suprachoroidal hemorrhage. Surv Ophthal-
acts as a scleral buckle and will slowly absorb in the postop- mol 1999;43(6):471–486.
2. Glazer LC, Williams GA. Management of expulsive choroidal
erative course. hemorrhage. Semin Ophthalmol 1993;8(2):109–113.
3. Beatty S, Lotery A, Kent D, et al. Acute intraoperative suprachor-
oidal haemorrhage in ocular surgery. Eye 1998;12:815–820.
MANAGEMENT OF ASSOCIATED 4. Tabandeh H, Sullivan PM, Smahliuk P, et al. Suprachoroidal
RETINAL DETACHMENT hemorrhage during pars plana vitrectomy. Risk factors and out-
comes. Ophthalmology 1999;106(2):236–242.
5. Wirostko WJ, Han DP, Mieler WF, Pulido JS. Suprachoroidal
Silicone is usually preferred to gas in conjunction with vit- hemorrhage: outcome of surgical management according to
rectomy. The purpose of silicone surface tension manage- hemorrhage severity. Ophthalmology 1998;105(12):2271–2275.
ment is rhegmatogenous confinement for unrecognized
C H AP TER
31
COMPLICATIONS OF
VITREORETINAL SURGERY

Most complications of vitreous surgery, after an contact lens. Infection of the corneal epithelial defect may
initial surgical learning phase, result from bio- occur in conjunction with soft lens usage.
logic problems associated with specific disease Endothelial cell damage is rare after vitrectomy with
states. Implicit in the discussion of management decreased turbulence three-port and 25-gauge systems, less
of complications is their recognition. The importance of fluid throughput, better lens removal techniques, and better
frequent follow-up of the complex vitreous surgery patient irrigating fluids (BSS Plus). If those patients with previous
cannot be overemphasized. In some cases, the primary oph- surgical trauma, glaucoma, or inherited endothelial dys-
thalmologist, less familiar with vitreous surgery, will not ini- trophies are excluded, postoperative corneal edema should
tiate appropriate action if complications occur. It is therefore not happen. Prolonged contact of a gas bubble or a silicone
quite important for the surgeon to follow up the patient with bubble totally filling the anterior chamber can damage the
a general ophthalmologist. Every attempt should be made to corneal endothelium and must be avoided by proper post-
educate other ophthalmologists in the recognition and man- operative positioning. Epithelial edema can be managed by
agement of these complications, but the primary responsi- topical hyperosmotic medications, although this is largely
bility rests with the surgeon. symptomatic treatment. Fortunately, sliding of endothelial
cells and the regaining of function of the remaining cells
cause clearing of corneal edema in most cases. If corneal
CORNEAL COMPLICATIONS edema persists and the eye is required for the patient’s visual
function, Descemet’s Stripping Endokeratoplasty (DSEK) or
Poor epithelial adherence can persist for weeks if the epithe- penetrating keratoplasty should be performed.
lium is removed at the time of vitrectomy (1–4). Care dur- Precipitates on the endothelium are frequently inter-
ing the prep, frequent irrigation of the cornea with balanced preted as evidence of inflammation, while in many cases,
salt solution (BSS), and shorter operating times dramatically they represent pigment released from iris and retinal pigment
reduce the need to remove the epithelium. The authors epithelium or erythroclasts.
remove the epithelium in less than 2% of cases. When neces-
sary, the epithelium should be mobilized rather than scraped.
The rounded blade used for mobilization should never touch INFLAMMATION
Bowman’s membrane. The defect should be made the same
size as the pupil, avoiding the peripheral cornea. Some sur- Most cells in the anterior chamber are erythroclasts released
geons use Gelfoam, tear substitutes, or viscoelastics during either from the vitreous lamella or as a result of intraop-
surgery, but the authors have not found these to be necessary erative or postoperative bleeding. If no retinopexy is per-
or efficacious. Bandage contact lenses or pressure patches formed, vitrectomy results in little inflammation. Iris trauma
are not necessary in the typical postoperative management. in conjunction with vitreous surgery results in inflamma-
In the rare cases requiring epithelial removal, the epithelium tion and should be avoided. Retained nucleus appears to
is healed within several days postvitrectomy and is always play a greater role in postoperative inflammation than does
healed on the 2-week office visit. If the patient develops a persistent cortex. All cases of iris neovascularization and
recurrent epithelial defect, cessation of topical medications most cases of retinal detachment have protein release in the
and taping the lid closed for 12 to 24 hours will usually anterior chamber, which is visible as “flare.” Treatment of
be sufficient. A rare patient will require a therapeutic soft the basic disease process by reattachment of the retina or

244
CHAPTER 31 ■ Com plications of Vitreoretinal Surgery 245

panretinal photocoagulation (PRP) is more effective than diabetic patients. The authors never use these agents because
topical steroids in reducing the flare due to neovasculariza- of the risk of stroke, myocardial infarction, and ketoacidosis.
tion. Anti–vascular endothelial growth factor (VEGF) com- Extreme caution should also be taken to prevent the IOP
pounds have proven to be effective in these cases. Topical from going above 30 mm Hg in the patient with vascular
and intraoperative subconjunctival, long-acting steroids are disease, systemic hypotension, and poor retinal perfusion.
used in all patients who are not steroid glaucoma responders
(5). This is primarily because any severe inflammation can
Air (Gas) Pup illary Blo ck
result in the development of a cyclitic membrane, periretinal
membranes, and, ultimately, phthisis bulbi. Steroids do not When air (gas) is utilized in the vitrectomized, aphakic or
appear to significantly retard the healing of any of the ocu- pseudophakic patient, the surface tension effect of the bub-
lar structures and should be used to reduce inflammation. ble can seal the pupil, just as it does a retinal break, resulting
The authors rarely prescribe systemic steroids for primary in a transiris pressure gradient. The continued production of
ocular conditions. Inflammation not responsive to frequent aqueous then forces the iris forward against the cornea, clos-
topical steroids is treated with repeated subconjunctival ing the angle and elevating the IOP. This can be prevented by
triamcinolone. proper postoperative positioning and typically occurs when
the instructions to patients are disregarded. In most cases,
reinforcement of the instructions given to the patient with
IRIS NEOVASCULARIZATION assistance from the office and nursing staff can reverse this
pupillary block immediately. If it is unrecognized for several
The cause and treatment of iris neovascularization have been days, the iris can become adherent to the cornea, requiring
extensively discussed in Chapter 6. Occasionally, iris neo- reoperation with a chamber deepening procedure through
vascularization will appear when retinal detachments with the pars plana. Iridectomies do not prevent this complica-
severe periretinal proliferation are operated on unsuccess- tion and are not required in most vitrectomy procedures.
fully. Intravitreal bevacizumab can be used to cause regres-
sion of the neovascularization postoperatively, followed by
Ne o vascular Glauco m a
PRP (6). If anti-VEGF therapy and PRP cause regression
of iris neovascularization, the large vessels will not disap- The treatment of neovascular glaucoma (NVG) can be sepa-
pear because of rheologic considerations. The examiner rated into components. The basic process of VEGF release
must concentrate on the presence of capillary activity and from the retina, which causes the iris neovascularization,
endothelial budding on the iris surface rather than the more must be treated by anti-VEGF agents, followed by PRP and/
impressive large vessels. Peripheral anterior synechiae and or retinal reattachment. If the pressure is highly elevated,
ectropion uveae are late changes and never disappear, even this must be managed aggressively. Timolol, brimonidine,
when capillary involution occurs. Although some clinicians and latanoprost are effective in many of these patients in
have emphasized the difficulty in differentiating iris neovas- combination with a topical carbonic anhydrase inhibitor.
cularization from dilation of preexisting stromal vessels, as a In others, these medications appear to have little effect. If
rule, this is not a difficult problem. medical management is unable to achieve pressure control
in the sub-35 mm Hg region, endocyclophotocoagulation
can be combined with endo-PRP. Trabeculectomy proce-
GLAUCOMA dures usually fail unless there is near complete involution of
neovascularization and inflammation. Ahmed valves are the
Increased intraocular pressure (IOP) from varied mecha- preferred method of filtering most of these patients. Patients
nisms is an all too frequent complication of vitreous sur- without light perception are usually taken off their pressure
gery. A high incidence of suspicion and frequent follow-up drops and treated only with topical steroids and pain medi-
is a necessity for recognizing and managing this dreaded cations. High IOP destroys the pain fibers to the eye just
complication (7). as it destroys the optic nerve; this approach eliminates the
need for enucleation. Enucleation is only necessary if there
is a risk of systemic infection from endophthalmitis, certain
Eryt h ro clast ic (He m o lyt ic) Glauco m a
tumors, or a cosmetic appearance that cannot be managed
Erythroclastic (hemolytic) glaucoma was quite frequent after with a scleral shell.
vitrectomy before vacuum cleaning–extrusion techniques
and intraoperative coagulation methods were utilized (8).
This type of glaucoma is transient and self-limited. The vast CYCLOCRYOTHERAPY
majority of cases can be treated with ocular hypotensive
medications such as topical carbonic anhydrase inhibitors, In-office cyclocryopexy is usually reserved for No Light
beta-blockers, and other agents. Extreme caution should be Perception Vision (NLP) cases. Cyclocryotherapy may
used in the administration of systemic hyperosmotic agents to cause phthisis bulbi, but it can be used for diseases that
246 SECTION IV ■ Disease-Sp ecific Managem ent

will result in phthisis bulbi without cyclocryotherapy. In the Op e n -An g le Glauco m a


vitrectomized aphakic eye, there is no matrix in the pupil-
Open-angle glaucoma (OAG) can be exacerbated by trans–pars
lary plane along which a cyclitic membrane can develop
plana vitrectomy. It appears that this type of OAG is second-
unless fibrin is present. It is important to use large doses
ary to the trauma suffered by the trabecular meshwork from
of periocular steroids in conjunction with cyclocryotherapy.
infusion fluid, cells, cytokines, protein, and debris. In this
Cyclocryotherapy can be performed on bare sclera so as to
way, it is comparable to the metabolic trauma suffered by
cause more predictable destruction of the ciliary processes as
the endothelium from infusion fluid and other intraocular
well as less postoperative conjunctival and Tenon’s capsule
agents. OAG can usually be managed by topical medications
damage and discomfort. If the cryopexy is used at −80°C and
but may require surgery. Chang has postulated that increased
confluent treatments are held for 1 minute, the effect appears
oxygen tension after vitrectomy plays a role.
to be reasonably predictable. It is better to use 180-degree
treatment and additional treatment only in reoperation situ-
ations. If the cryopexy is directed over the ciliary body with
only minimal anterior extension, there is less damage to the ENDOPHTHALMITIS
remaining functioning trabecular meshwork. If the iceball
reaches the limbus, this means it is reaching the trabecular Endophthalmitis was relatively frequent in the early days
meshwork internally and has extended too far. At this point, of vitrectomy at some institutions; it now fortunately has
the probe should be reapplied and treatment directed more become an infrequent complication. The principal author
posteriorly. Therapeutic ultrasound or transscleral 1,064-nm has had three cases of endophthalmitis in over 26,000 vit-
laser cyclodestructive procedures may be effective in lieu of rectomies. Better equipment, shortened operating times,
cyclocryotherapy in these cases. Endocyclophotocoagula- povidone iodine prep, microscope drapes, one-piece drapes,
tion is more effective and has fewer complications than cryo, and better irrigating solution (BSS Plus) have probably con-
laser, or ultrasound but requires intraocular surgery. tributed to the rarity of endophthalmitis now seen after vit-
rectomy. The authors recommend the use of subconjunctival
antibiotics with coverage for both Gram-positive and Gram-
FILTERING PROCEDURES FOR negative bacteria as the final step in surgery. Even though
NEOVASCULAR GLAUCOMA these agents will only infrequently play a role, the devastat-
ing effect of endophthalmitis should be prevented at all costs.
Trabeculectomies (9), valve, filtering shunt procedures The authors are currently using Tobramycin and Cefazolin.
(10–14), and pars plana filtering procedures (15) have been If endophthalmitis does occur in the postvitrectomy patient,
utilized with moderate success in selected patients with there is no need to perform further vitrectomy. A needle may
NVG. The Ahmed valve is the preferred procedure at this be placed through the pars plana into the vitreous cavity and
time. It appears that in the filtering procedures, there is a fluid aspirated for culture and sensitivity testing. This same
dynamic dilutional effect whereby VEGF is decreased due needle can then be used for intraocular antibiotic injection.
to increased fluid throughput. Because of this, the iris neo- Some have recommended the use of antibiotics routinely in
vascularization may involute rapidly and neovascularization the infusion fluid. The question about toxicity of these sub-
may appear in the bleb. stances, especially with cumulative dosing, coupled with the
Extensive PRP to cause involution of iris vessels should relative infrequency of endophthalmitis makes this approach
precede filtering procedures whenever possible. Frequent unnecessary and possibly dangerous.
topical or subconjunctival steroids should be used to suppress
inflammation before and after filtering procedures.
RETINAL COMPLICATIONS
St e ro id Glauco m a
Removal of the vitreous causes minimal, if any, inflam-
The efficacy of subconjunctival short-acting steroid (Decad- mation or cystoid macular edema (CME). Postvitrectomy
ron) is so great in the postvitrectomy patient that it should inflammation is caused primarily by iris trauma, retinopexy,
be universally used, unless the patient is a known steroid retinal trauma, laser treatment, agents injected into the eye,
responder. Subconjunctival Kenalog was used in most cases and residual lens material. Vitrectomy can reduce or elimi-
in earlier years, but the more recent observation that steroid nate CME by debulking inflammatory factors in the vitre-
glaucoma is often not reversible has changed this practice. ous. Vitrectomy, especially when combined with aphakia,
Intravitreal triamcinolone results in approximately 30% decompartmentalizes the eye, facilitating egress of cells,
incidence of steroid glaucoma, which is often irreversible. In proteins, and cytokines through the trabecular meshwork.
addition, the incidence of steroid cataracts is approximately Oral and topical nonsteroidal anti-inflammatory agents
90%. The incidence of steroid glaucoma after fluocinolone (Nevanac) as well as subconjunctival and topical steroids
implant (Retsisert, Bausch & Lomb) is approximately 90%, can have a beneficial effect on postvitrectomy inflammation
and over 30% of the patients require a filtering procedure. and CME (16).
CHAPTER 31 ■ Com plications of Vitreoretinal Surgery 247

Vascular Occlusio n component. Required ERM peeling, segmentation, and


delamination elicit a reparative effort with further prolifera-
The elevation of IOP associated with vitrectomy can create
tion, causing retinal elevation. Residual ERM can contract
vascular occlusion and permanent visual loss. Strict atten-
also, creating nonrhegmatogenous retinal elevation. If these
tion to intraoperative and postoperative IOP is required to
elevated areas are small, they can be observed without surgi-
prevent this severe complication. Children and adults with
cal intervention. At times, vitreous will unintentionally be
low blood pressure are at high risk for retinal and optic nerve
left, causing residual traction on the retina. If extramacular
ischemia from elevation of the IOP.
retina is elevated, it can be observed postoperatively for an
extensive period with much the same criteria as those used
Re t in al Bre aks an d De t ach m e n t at the time of the original surgery. Nonrhegmatogenous mac-
ular elevation or any elevation accompanied by iris neovas-
Retinal breaks may precede vitrectomy, occur at the time of cularization should indicate the need for reoperation with
vitrectomy, or appear in the postoperative course. Any breaks scissors segmentation and delamination methods. At times,
present at the time of vitrectomy should have been identified segmental scleral buckling is required.
and treated, but, on occasion, surgical visualization prob-
lems prevent this. Retinal breaks occurring at the time of
vitrectomy can be divided into two groups: those caused by Man ag e m e n t o f Re t in al De t ach m e n t
direct retinal trauma and those related to vitrectomy trac- Rhegmatogenous detachment repair after vitrectomy almost
tion. Any time that instruments are brought near the retinal always requires gas or silicone for surface tension manage-
surface, the potential exists for the creation of a retinal break. ment. Because vitreous removal markedly decreases the vis-
A retinal tear can be made with any intraocular instrument, cosity of the vitreous fluid, a very small break will rapidly
although it is most frequently found as a result of epiretinal result in total retinal detachment. With the use of the sur-
membrane (ERM) surgery in diabetic traction retinal detach- face tension effects of air (gas), these cases can be reattached
ment cases. Vitreous removal results in an obligatory trac- more expeditiously and safely. In virtually all instances, the
tion on the retina from the suction force on the vitreous fiber procedure is set up for full vitrectomy with the customary
framework. This mechanism is similar to that of an aphakic incisions, the endoilluminator, cutter, and tools. Any residual
retinal detachment occurring from anterior movement of the vitreous traction is removed and the visualization improved
vitreous after capsule rupture during cataract surgery. With by removal of any blood or debris. If ERM is causing reti-
this form of detachment, there are small breaks at the poste- nal foreshortening, peeling, segmentation, and delamina-
rior edge of the vitreous base. These are frequently difficult tion are utilized. After removal of the causative traction,
to recognize at the time of vitrectomy or are only of partial internal drainage of subretinal fluid (SRF), internal fluid-air
thickness at that time, becoming full thickness later. Thus, exchange, and completion of internal drainage of SRF are
there is a built-in incidence of aphakic-like retinal detach- utilized. This is followed by endolaser retinopexy unless the
ment as a result of vitreous removal by any method. More break is in the macular or peripapillary region.
direct trauma to the peripheral retina occurs from the entry
of instruments through the pars plana. Large instruments
with abrupt increases in diameter can cause dialyses, while INTRAOCULAR HEMORRHAGE
small instruments can push the vitreous base in front of
them and create dialyses or small breaks as well. Postoperative intraocular hemorrhage is rare except in the
Late retinal breaks occur from several mechanisms. diabetic patient. Diabetic patients develop postoperative
Trauma to the retina at the time of vitrectomy may result in hemorrhage approximately 50% of the time. Resected vas-
partial-thickness retinal loss with subsequent retinal break cular tissue, sclerotomy wounds, iris neovascularization, and
formation. Postoperative traction can occur from residual incomplete regression of retinal neovascularization are the
vitreous particularly that incarcerated in the sclerotomies. most frequent causes of postoperative intraocular hemor-
Any form of periretinal proliferation can cause retinal short- rhage. As discussed in this chapter, hemorrhage cases should
ening and retinal breaks from tangential traction. In some be followed up with ultrasound and reoperated only if reti-
cases, large, atrophic retinal breaks will occur without any nal detachment occurs. If the patient is blind in an only eye
evidence of traction or surgical trauma. These are seen in the or both eyes, reoperation to remove blood should be consid-
context of ischemic retinopathies and appear to be due to ered for the better eye.
retinal necrosis from vascular occlusive disease. If indicated, blood removal should be performed in all
instances using the full setup and three-port, usually 25-gauge
technique. This permits extrusion, bipolar diathermy, endo-
No n rh e g m at o g e n o us Re t in al De t ach m e n t
photocoagulation, and dissection of any significant ERM to
Periretinal migration and proliferation from glial, retinal pig- be done. If iris neovascularization is present, blood removal
ment epithelial, or wound-related cellular proliferation can should be performed to accomplish retinal reattachment
lead to elevation of the retina without a rhegmatogenous and/or completion of PRP.
248 SECTION IV ■ Disease-Sp ecific Managem ent

In all patients, it is important to emphasize that they should


CATARACT
check their vision every day and should contact the phy-
sician immediately if visual loss or pain occurs. It is criti-
Many surgical factors add to the baseline incidence of cataract
cal to instruct the office staff about the access these patients
associated with the ocular diseases requiring vitreous sur-
must be afforded to postoperative care. The complexity of
gery (17). Lens opacities are a frequent but easily manage-
postvitrectomy patients simply prohibits the use of a wait-
able postvitrectomy complication. Lens removal should be
ing list. As a rule, diabetics should be followed up at 6-week
considered to address the patient’s visual needs or if the view
to 3-month intervals until they are stable for 1 year. At this
of the retina is lost, preventing good management.
point, the interval can be lengthened.
Phaco-vit is preferred to pars plana lensectomy approach
for cases requiring combined vitreoretinal surgery.
References
SUTURE EXTRUSION 1. Perry HD, Foulks GN, Thoft RA, et al. Corneal complications
after closed vitrectomy through the pars plana. Arch Ophthal-
mol 1978;96(8):401.
On occasion, 8-0 monofilament nylon sclerotomy sutures 2. Brightvill FS, Myers FL, Bresnick GH. Postvitrectomy keratop-
will erode through the conjunctiva. This problem has been athy. Am J Ophthalmol 1978;85:651.
virtually eliminated with the advent of sutureless 25-gauge 3. Aaberg TM, Van Horn DL. Late complications of pars plana
vitrectomy. Because this is a running suture, it is quite dif- vitreous surgery. Ophthalmology (Rochester) 1978;85(2):116.
4. Kenyon KR, Stark WJ, Stone DL. Corneal endothelial degen-
ficult to remove at the slit lamp. It is better to use topical
eration and fibrous proliferation after pars plana vitrectomy.
anesthesia and the laser to melt the protruding end. Alterna- Am J Ophthalmol 1976;8(4):486.
tively, a disposable electrothermal cautery can be used if the 5. Hall AJ. Effective local treatment for severe ocular inflam-
lids are retracted securely. If loops of suture erode, they can matory disease: towards the holy grail. Clin Exp Ophthalmol
be severed with a blade or Vannas scissors at the slit lamp 2001;29(1):1.
6. Lupinacci AP, Calzada JI, Rafieetery M, et al. Clinical outcomes
in the office. The ends of 5-0 nylon scleral buckle sutures
of patients with anterior segment neovascularization treated
can be trimmed in the office, but this may result in buckle with or without intraocular bevacizumab. Adv Ther 2009;
extrusion. With the use of hard silicone explants, copious 26(2):208–216. [Epub 2009 Feb 20.]
antibiotic flushing, and a 1.0-mm conjunctival flap, infected 7. Campbell DG, Simmons RL, Tolentino FL, et al. Glaucoma
buckles are extremely rare. If infection occurs, it should be occurring after closed vitrectomy. Am J Ophthalmol 1977;
83(1):63.
managed by removal of the explant materials and irrigation
8. Brucker AJ, Michels RG, Green WR. Pars plana vitrectomy in
with saline and antibiotics. the management of blood-induced glaucoma with vitreous
hemorrhage. Am J Ophthalmol 1978;10(10):1427.
9. Herschler J, Agness D. A modified filtering operation for vascu-
FOLLOW-UP INTERVALS lar glaucoma. Arch Ophthalmol 1979;97:2339.
10. Krupin T, Kaufman P, Mandell A, et al. Filtering valve implant
Typically, vitrectomy patients are treated as outpatients and surgery for eyes with neovascular glaucoma. Am J Ophthalmol
1980;89:338.
therefore go home on the day of surgery. The first post- 11. Krupin T, Kaufman P, Mandell A, et al. Long-term results of
operative visit should be on the day after surgery, and the valve implants in filtering surgery for eyes with neovascular
subsequent exam should be in 3 weeks in most instances. glaucoma. Am J Ophthalmol 1983;95:775.
It would be unusual to experience any reason to reoperate 12. Molteno ACB, Van Rooyen MMB, Bartholomew RS. Implants for
in the first 3 weeks, and a 3-week follow-up exam discov- draining neovascular glaucoma. Br J Ophthalmol 1977;61:120.
13. Schocket SS, Lakhanpal V, Richards RD. Anterior chamber
ers the highest incidence of treatable postoperative com- tube shunt to an encircling band in the treatment of neovascu-
plications. If this period were extended to 1 month, in the lar glaucoma. Ophthalmology 1982;89:1188.
diabetic vitrectomy patient, there would be a much higher 14. Schocket SS, Nirankari VS, Lakhanpal V, et al. Anterior cham-
incidence of NVG. Most PVR recurrences occur in the first ber tube shunt to an encircling band in the treatment of neo-
3 weeks. Most rhegmatogenous postvitrectomy retinal vascular glaucoma and other refractory glaucomas; a longterm
study. Ophthalmology 1985;92:553.
detachments will also present by the 3-week mark and can 15. Sinclair SH, Aaberg TM, Meredith TA. A pars plana filtering
be effectively managed at that time. The first postoperative procedure combined with lensectomy and vitrectomy for neo-
day visit is usually sufficient to cover the acute endophthal- vascular glaucoma. Am J Ophthalmol 1982;93:185.
mitis risk. 16. Sjaarda RN, Glaser BM, Thompson JT, et al. Distribution of
If the patient is doing well at the 3-week visit, he or she iatrogenic retinal breaks in macular hole surgery. Ophthalmol-
ogy 1995;102(9):1387–1392.
can be given a 6-week visit unless the patient is a diabetic. 17. Blankenship G, Cortez R, Machemer R. The lens and pars
The diabetic patient should return in 3 weeks for further plana vitrectomy for diabetic retinopathy complications. Arch
examination for iris neovascularization and glial recurrence. Ophthalmol 1979;97(7):1263.
C H AP TER
32
SURGICAL SELF-EDUCATION

While there are many excellent courses, articles, see. It is even important to attempt an assessment of one’s
and textbooks available concerning vitreo- temperament. Vitreous surgery requires a calm but rapid
retinal surgery, improvement in judgment and and efficient approach. A surgeon who becomes very tense
surgical skills must principally come about by and inefficient in times of surgical stress has no place in vit-
self-education. The complexity of high-technology vitre- reous surgery. A person so compulsive and rigid that nec-
ous surgery on high-risk patients demands a continued essary changes in the game plan produce overwhelming
assessment of surgical and biologic results. Vitreous surgery stress probably should not be performing vitreous surgery.
requires an excellent training in microsurgery and retinal Although ego and economic factors unfortunately influence
diseases. Eye bank eyes, porcine eyes, and molded rubber some surgeons’ decisions, the pleasure is short lived if the
practice eyes can be used for surgical practice (1–6). It is results are poor, resulting in a damaged and unhappy patient
simply poor judgment to begin vitreous surgery or a new and possibly a lawsuit. It therefore becomes important to
technique on the human patient without sufficient practice. look realistically at the demands for vitreous surgery in the
After reading the available literature, visiting other surgeons, individual’s practice, with an intent to determine if certain
and attending appropriate courses, progress will be made in procedures can be done frequently enough to attain suffi-
the laboratory. When sufficient competency is obtained in cient surgical skill.
the laboratory, the beginning surgeon should assemble the
disposables and equipment required for simulated surgery.
Simulator technology has improved, gaining substantial OUTCOMES ANALYSIS
fidelity in recent years, and has the advantages that disease
states and intraoperative complications can be simulated. The The collection of preoperative, operative, and postopera-
EyeSi simulator has an excellent vitreoretinal module and is tive information is essential to the self-assessment of surgi-
highly recommended. Regardless of the presence of other cal techniques and skills. While some feel that this is the
vitreous surgeons at the same institution, it is the responsi- obligation of so-called academic institutions, it is, in fact,
bility of the beginning surgeon to go through this practice the obligation of each and every surgeon. Some surgeons
surgery approach. It is absolutely the responsibility of each are able to achieve series large enough for publication, but
surgeon to make certain that all equipment is available and each individual must produce a series for comparison. The
functioning. Unfortunately, many surgeons fall into the trap importance is not so much in contributing to the literature
of placing this responsibility upon technicians, nurses, and as in being apprised of one’s own outcomes. Outcomes
ancillary personnel. Practice surgery in the actual operating research begins with careful preoperative evaluation and
room should be repeated on the days preceding vitreous sur- prospective recording of this information. It is best to have
gery if the case in question has not been approached before a format on which these data can be recorded, and exten-
or the procedures are done infrequently. sive use of abbreviations and recognized grading systems
The great complexity of vitreoretinal surgery requires should be utilized. With the use of a scribe accompanying
an honest assessment of the surgeon’s own capabilities. It is the examiner, the information can be dictated in abbrevia-
simply inadequate to perform vitrectomy without stereopsis. tion format and recorded extremely rapidly without the
There are many areas of medicine that are less demanding unavoidable errors that occur with trying to recall this later
in the requirements for stereopsis, and the surgeon should at the time of dictation. Although it is permissible to fill
not perform vitreous surgery without stereopsis. Red-green out sheets at this time, if they are secondary to the primary
color blindness is a major handicap because it makes the charting method, details tend to be overlooked in a busy
diagnosis of iris neovascularization, small retina breaks, and practice. It is critical to determine the parameters that one
the regression of neovascularization much more difficult to wishes to follow preoperatively and postoperatively and
249
250 SECTION IV ■ Disease-Sp ecific Managem ent

to record all of this information on each patient to permit is a complication of this method. Again, a computer-based
biostatistical evaluation at a later date. approach can be quite effective but requires additional work
It is best to compile this information by disease cat- to set up, and its unavailability should not be used as an
egory so that discrete biologic groups may be identified. excuse for not having continuous data monitoring.
This compilation can be as simple as single sheets that list It is helpful to have one member of the surgical assistant
patients with a given disease state and entrance criteria, with team monitor these data with the surgeon so as to increase
columns for preoperative, operative, and postoperative find- his or her involvement and understanding. If a surgical team
ings and all complications. These sheets should be filled out member helps by abstracting operative records, it adds imp-
immediately following each day’s examinations so that miss- artial credibility and enhances surgical understanding as
ing findings can be determined at that point. The surgical well. If this same individual is involved in preoperative and
part of these forms should be filled out immediately follow- postoperative clinical photography and other special exami-
ing surgery so that it will not have the inaccuracies inher- nation techniques, it increases concern for the patient and
ent in subsequent abstraction from postoperative dictation. understanding of prognostic and management factors.
Similarly, the postoperative findings should be recorded after
each visit, preferably while the patient is still present, so that
any missing information can be obtained. CORRESPONDENCE
The computers utilized in office practice could make
this process more efficient, but useful computerized medical All postoperative follow-up information from referring doctors
record software is just becoming available and if one waits should be coded immediately into the file sheets as well as
until computer skills or capabilities are sufficient, much data on the chart. In this way, a retrospective chart review with all
and useful information will be lost. its inherent inadequacies is avoided entirely. The quarterly or
It is essential to determine average success rates from the pretalk data compilation will uncover many patients who had
data forms and to compare complication rates to published inadequate follow-up. At this time, the referring doctor should
outcomes. This should be done at least every 3 months, with be contacted by phone or postoperative follow-up forms
a stimulus being provided by a quarterly report, an upcom- should be mailed in order to complete all follow-up informa-
ing paper, or an upcoming lecture. The data then become tion. These frequent checkups serve a purpose in stimulating
very helpful in predicting the outcome of surgery for patients follow-up by the surgeon and referring physicians. Preprinted
and in self-assessment for surgical improvement. Care must follow-up forms can be made available to referring physicians,
be taken to compare similar biologic groups that are selected which can act as a stimulus for better data retrieval. Unfortu-
with given entrance criteria. For example, a vitreous hemor- nately, many practitioners do not refract other physician’s post-
rhage patient should not be compared to a traction detach- operative patients, making the visual acuity data inadequate.
ment patient with respect to visual improvement because One then must specifically inquire and encourage the use of
simple clearing of the media improves the vision in a differ- best-refracted visions.
ent manner than macular reattachment.

GROUP EDUCATION
SURGICAL DATA
The use of effective outcomes research as described previ-
It is important to dictate an extremely complete and honest ously contributes to the quality of presentations to other sur-
operative note at the end of each procedure. This should be geons and at meetings. It is important to frequently attend
done describing every aspect of the technique and all surgi- meetings with surgeons performing similar work in order to
cal findings. While some physicians use surgical forms, they upgrade medical and surgical knowledge. Unfortunately, the
are usually too stereotyped and inflexible for this complex literature is months to years behind in reporting newer meth-
type of surgery. Similarly, while photographs and drawings ods; more rapid education is possible by attending meetings.
can complement the description, carefully described surgical If all speakers emphasize this approach to outcomes research
findings are very helpful in following up the patients in the with at least a modicum of knowledge concerning biostatis-
office. This approach helps not only in outcomes research but tics, better communication is possible. Certainly, everything
also in understanding complications on an individual basis. cannot be studied in a randomized masked study, but accu-
It is suggested that in addition to the copies of the operative rate compilation of results is nevertheless mandatory.
notes in the hospital and office records, a third copy be kept
in the computer or separate files. These sequential operative
notes should then be abstracted with cross-referencing for INTERACTION WITH COMPANIES
certain findings or techniques that will benefit from subse-
quent analysis. For example, all macular hole patients who The careful analysis of results with different surgical
underwent peeling of the cortex from the optic nerve can be techniques permits better communication with medical
assessed for visual results to determine if optic nerve damage equipment manufacturing companies. This should not be
CHAPTER 32 ■ Surgical Self-Education 251

done so as to make the equipment a scapegoat for surgical References


failure but rather to provide constructive advice concern-
1. Michels RG. Intraocular fluorescein in experimental vitrectomy.
ing equipment improvement. It is usually better to go to Ophthalmic Surg 1977;8:139.
major equipment manufacturers for prototype equipment 2. Bensen WE. Vitrectomy in rabbit eyes (appendix). In: Vitrectomy,
than to local machine shops. If these devices are made in a pars plana approach. New York: Grune & Stratton, 1975.
local shops, they are not accessible to colleagues and are 3. O’Malley C. Learning surgery without risk or anxiety. Ocutome
Newsletter1977; 2(3).
not prototyped with a view to future manufacturing meth-
4. Borirak-Chanyavat S, Lindquist TD, Kaplan HJ. A cadaveric eye
ods. It is very important for the surgeon to have frequent model for practicing anterior and posterior segment surgeries.
and open communication with companies so as to cre- Ophthalmology 1995;102(12):1932–1935.
ate a climate of intellectual cooperation that encourages 5. Eckardt U, Eckardt C. Keratoprosthesis as an aid to learning sur-
equipment improvement. An extension of this attitude gical techniques on cadaver eyes. Ophthalmic Surg 1995;26(4):
358–359.
toward the surgical team and colleagues benefits all those
6. Moorehead LC. Practice vitrectomy. Arch Ophthalmol 1980;
involved. 98(7):1297–1298.
Index

Note: Page numbers followed by “f” indicate figures; page numbers followed by “t” indicate tables.

A for ciliary block glaucoma, 91–92 Blood pressure


Adjuvant therapy congenital and pediatric cataracts, 90 during general anesthesia, 40–41
in management of intermediate uveitis, 223 for cystoid macular edema, 92–93 Bright flash electroretinography, 15
Age infusion sources for, 91 B-scan ultrasonography, 12
in preoperative assessment, 2 instrumentation for, 76 Buckling. See Scleral buckling
Air intraocular lenses, 93–96, 94f, 95f
vitrectomy under, 68, 69f limbal versus pars plana approach in, 78 C
Air-gas exchange pars plana lensectomy, 78–90 Cannulas
and general anesthesia, 43 for phacolytic glaucoma, 93 extrusion
for giant retinal breaks, 139 pupillary membranes, 90–91 for subretinal fluid drainage, 60f
pupillary block and, 245 translimbal, 91 infusion, 22, 113–115, 114f, 115f
subretinal fluid drainage and, 62, 63f traumatic cataracts, 90 soft-tip
Air/silicone exchange. See Silicone infusion ultrasonic fragmentation in, 76–78 for subretinal fluid drainage, 60f
Anesthesia in uveitis, 93 trocar system of, 103–104, 104f, 105f
anticoagulation and, 43 for vitreocorneal touch, 91 withdrawal and wound closure, 111
carbon dioxide issues during, 42–43 for vitreous loss in cataract surgery, Capsulectomy
diabetic retinopathy, 175 92, 92f pars plana
for endophthalmitis, 43 Antibiotics after lensectomy, 83
facial nerve blocks, 42 in infusion fluid, 28 Capsulotomy
general intracameral pars plana
air/gas exchange and, 43 for endophthalmitis, 219 after lensectomy, 83
blood pressure during, 40–41 in posterior vitrectomy, 73–74 Carbon dioxide
versus local, 40 subconjunctival intraoperative build-up of, 42–43
intraconal for endophthalmitis, 219 Cardiopulmonary disease
technique for, 42 topical preoperative assessment in, 3
local for endophthalmitis, 219 Cataract(s)
choice of, 41–42 toxicity of, 239 after vitrectomy for epimacular
psychological preparation for, 41 Anticoagulation, 43 membranes, 162
reblocking during, 42 Aphakic keratoplasty congenital and pediatric, 90
sedation during, 41 anterior vitrectomy and, 93 postoperative
monitoring during, 40 A-scan ultrasonography, 12 in diabetic retinopathy, 185
with open globe, 43 Aspiration traumatic, 90
and operating room efficiency, 40–44 technology for in vitreoretinal surgery, 248
postoperative pain, 43–44 cutter movement, 25–26 Cataract surgery
preoperative evaluation for, 40 port configuration, 26 capsular defects in
presurgical self-sharpening, 26 recognition and management of, 84–85
for penetration of eye, 239–240 vitreous cutter design issues, 26–27 posterior lens dislocation in, 83–90, 86f–89f
for scleral buckling, 43 with ultrasonic fragmentation, 77 vitreous loss in
for vitreoretinal surgery, 42 Avastin anterior vitrectomy for, 92, 92f
Angioscopy in choroidal neovascular membranes, 193 cellulose sponge vitrectomy, 85, 86f
fluorescein Cellular migration/proliferation
in posterior vitrectomy, 73–74 B traumatic, 205
Anterior chamber aspiration Barcode reader wand, 35, 36f Cellulose sponge vitrectomy, 85, 86f
for endophthalmitis, 217–218 Barrier concept Children
Anterior segment in diabetic retinopathy, 186–187, 186f cataracts in, 90
problems of Bimanual bipolar diathermy traction retinal detachment in, 231
vitrectomy techniques and, 76–96 in posterior vitrectomy, 72 Choroidal hemorrhage, 241
Anterior vitrectomy Biologic modifiers Choroidal neovascular membranes
aphakic filtering procedures and, 92 for macular holes, 169 anti-VEGF therapy strategies, 194
aphakic keratoplasty and, 93 Bipolar diathermy systems, 30 combination therapy, 194–195
for aphakic pupillary block, 91 Bleeding. See Hemorrhage dietary supplementation for, 193–194

253
254 Index

Choroidal neovascular membranes (Continued) cysts, 175 Endophthalmitis


drusen, laser treatment of, 194 imaging for, 171, 172f anesthesia and, 43
focal thermal laser ablation, 193 infusion fluid, 184–185 anterior chamber versus vitreous aspiration,
macular translocation, 201–202 macular edema, 175 217–218
photodynamic therapy, 194 medical issues in, 171 antibiotics for, 219
plasmapheresis, 194 nonproliferative retinopathy complications of, 220
results of, 200 corticosteroids, 172–173 endogenous, 216
retinal pigment epithelium transplantation, macular edema, 172–173 epimacular membranes and, 162
200–201 vascular endothelial growth factor exogenous, 216–217
submacular surgery antagonists, 173 in-office vitreous tap and intravitreal
and macular translocation, 196, 196f proliferative retinopathy, 173–174 antibiotics injection, 218
for subfoveal membranes, 196 results of, 185 postoperative, 111
surgical sequence for, 196–198, 197f–200f retinal detachment results of, 219–220
Color discrimination ancillary epiretinal membrane management surgical sequence and techniques for
preoperative assessment of, 4 techniques, 183 trans–pars plana versus translimbal
Companies anti-vascular endothelial growth factor approach, 218–219
interaction with, 250–251 therapy, 183 vitrectomy, 219
Congenital cataracts, 90 epiretinal membranes, 181, 182f TASS, 217
Conjunctiva hemostasis, 182–183, 182f vitrectomy for, 218
displacement of, 46–47 inside-out versus outside-in technique, 181 in vitreoretinal surgery, 246
incisions of, 112 panretinal photocoagulation, 183 Endoscope systems, 38–39
in trauma, 206 retinectomy, 183 Entoptic phenomenon
and tenon’s capsule, 116 scissors segmentation and delamination, preoperative assessment of, 4
Constellation architecture 180–181, 180f, 181f Epimacular membranes
auto gas fill, 34, 34f silicone oil, 183 vitreomacular traction syndrome
barcode reader wand, 35, 36f viscodissection, 181–182 case selection, 157–158
embedded PurePoint 532-nm laser, 34, 34f retinal thickness measurement, 171 cataract, 162
power forceps, 35, 35f retinopexy, 185 coexistent cataract, 160–161
power scissors, 35, 35f scleral buckling, 185 cystoid macular edema, 161–162
UltraVit 5,000 cpm vitreous cutter, 32, 33f screening for, 171 endophthalmitis, 162
xenon illuminations systems, 33, 33f submacular exudates, 175 epimacular proliferation, 162
Contrast sensitivity surface tension management, 183–184 epiretinal membrane removal, 159–160,
in preoperative assessment, 4 surgical sequence and techniques 159f–161f
Cornea anesthesia, 175 history of, 157
preoperative slit lamp examination of, 5 incisions, 175 membrane peeling, 158–159
Corneal complications, 244 lens management, 175–176 nonrhegmatogenous proliferative
Corneal opacification, 213–214, 214f posterior vitreous detachment, 176–178, vitreoretinopathy, 158
Corneal-scleral laceration, 212–213 177f–179f pathogenesis, 157
Correspondence vitrectomy, 176, 176f proliferative vitreoretinopathy
management of, 250 traction retinal detachment, 174 recurrence, 161
Corticosteroids visualization, 184 retinal breaks, 160, 161
diabetic macular edema, 172–173 vitrectomy, 174 retinal whitening, 162
Cryopexy, 119 vitreous hemorrhage, 174 rhegmatogenous retinal detachment, 161
Cutters. See Tissue cutting Diathermy, 30 surgical sequence and techniques, 158–161
Cyclocryotherapy, 245–246 bimanual bipolar visual loss and, 157
Cystoid macular edema in posterior vitrectomy, 72 visual results, 161
anterior vitrectomy for, 92–93 transscleral, 73 Epiretinal membrane dissection
epimacular membranes and, 161–162 Direct (transscleral) needle drainage bimanual surgery, 53, 54f
Cysts of subretinal fluid, 70–71, 70f, 71f “chopsticks” membrane peeling, 56
in diabetic retinopathy, 175 Discission, 90 cutter delamination, 56, 56f, 57f
Dislocation en bloc in, 53
D lens flowchart, 55
Diabetes mellitus in cataract surgery, 83–90, 86f–89f hemostasis, 54
preoperative assessment in, 2–3 Double penetrating injury, 212 membrane peeling in, 49–50, 49f
Diabetic retinopathy Drainage retinotomy, 61 power scissors and forceps in, 54
barrier concept in proliferative vitreoretinopathy, 146–148,
anterior vitreous cortex fibrovascular prolif- E 148f
eration, 187 Education scissors segmentation and delamination in,
glial recurrence, 187 group, 250 50–53, 51f–53f
rhegmatogenous retinal detachment, 187 surgical self-education, 249–251 simplified, 49
VEGF encounters, 186, 186f Electroretinography suction, 56
complications bright flash, 15 surgery stabilization in, 54, 55f
erythroclastic (hemolytic) glaucoma, 185 Endocapsular lensectomy, 80–83, 80f–82f vacuum cleaning–extrusion, 56–58, 57f
hemorrhage, 185 Endodiathermy, 72–73. See also Diathermy viscodissection, 53–54
neovascular complications, 185–186 Endoillumination, 28, 28f Epiretinal membranes
postoperative cataract, 185 Endophotocoagulation, 29. See also Laser in diabetic retinopathy, 181, 183
contraindications, 175 endophotocoagulation preoperative assessment of, 8
Index 255

removal of, 159–160, 159f–161f anterior vitrectomy for, 93 Intraocular hemorrhage, 247
retinopathy of prematurity, 230 steroid, 246 Intraocular lens
Epithelial ingrowth, 96 in trauma, 214 dislocated
Erythroclastic (hemolytic) glaucoma, 245 Glial recurrence lens removal, 96
Extrusion method in diabetic retinopathy, 187 repositioning of, 93–96, 94f, 95f
in epiretinal membrane dissection, 56–58, 57f Gray scale ultrasonography, 13 epithelial ingrowth of, 96
Group education, 250 fogging during fluid-air exchange, 62
F implantation of, 90
Facial nerve blocks, 42 H removal of
Fluid Hemorrhage before trans–pars plana vitrectomy, 96
infusion, 28 in diabetic retinopathy, 185 retrolental membranes and, 93
diabetic retinopathy, 184–185 intraocular, 247 Intraocular lymphoma
posterior vitrectomy, 45 submacular, 195 cytology and flow cytometry for, 222
Fluid-air-gas exchange. See also Air-gas exchange suprachoroidal, 241–243 Intraocular pressure
giant retinal breaks, 139 vitreous slit lamp preoperative assessment of, 5–6
in-office, 122–123, 123f in diabetic retinopathy, 174 Intraoperative video recording, 19
Fluidics, 104–106, 106f Intravitreal antibiotics, 218
aspiration, 22–23 I Intravitreal injection technique, 204
infusion, 21–22, 21f Illumination, 37–38 Iris
surgical, 21 Indirect ophthalmoscopy anterior chamber
Fluorescein angiography preoperative, 6 slit lamp preoperative examination
in diabetic retinopathy, 171 Inflammation of, 5
Fluorescein angioscopy in proliferative vitreoretinopathy, 155 Iris neovascularization
in posterior vitrectomy, 73–74 in vitreoretinal surgery, 244–245 in vitreoretinal surgery, 245
Follow-up Infusion fluid, 28 Irrigation
intervals for, 248 diabetic retinopathy, 184–185 saline, 88, 88f
Forceps posterior vitrectomy, 45
constellation architecture, 35, 35f Infusion instruments K
power scissors and, 54 alternative, 27–28 Keratoplasty
Foreign body. See Intraocular foreign body cannulas, 27, 27f aphakic
Fragmentation enter sites for, 27 anterior vitrectomy and, 93
in pars plana lensectomy, 78–79 Infusion needles, 28 Keratoprosthesis
ultrasonic Infusion systems classification of, 236
in anterior vitrectomy, 76–78 for anterior vitrectomy, 76, 77f office examinations in, 237
Fragmenter, 79, 79f Injuries. See Trauma vitrectomy in
avoidance of vitreous in, 83, 84f In-office fluid-air-gas exchange, 122–123, 123f 25-gauge trocar cannulas, 237
Full function probes, 27 Instruments peripheral retinal visualization and
for anterior vitrectomy, 76 management, 237
G cutters, 19–22 retroprosthetic membranes, 238, 238f
General anesthesia. See Anesthesia, general ergonomic issues and, 28–29
Giant retinal breaks infusion, 27 L
case selection, 138 peak electron avalanche knife, 20 Laser endophotocoagulation, 72–73
definition of, 138 port configuration, 26 Laser interferometry, 4
management of, 143 presurgical testing of, 19 Laser photocoagulation
proliferative vitreoretinopathy and, 138 self-sharpening, 26 cryopexy and, 119
subretinal fluid drainage, 64 storage of, 18–19 PASCAL laser, 118
vitrectomy for, 138–139, 139f Intracameral antibiotics, 219 retinal breaks, prophylaxis for, 118–119
fluid-air-gas exchange in, 139 Intraconal anesthesia, 42 technique for, 119
incisions in, 138 Intraocular foreign body Laser retinopexy, 141
lens management in, 138 antibiotic-steroid therapy, 211–212 Laser therapy
patient education for, 138 conjunctival incisions, 206 for retinopathy of prematurity, 227
perfluorocarbon liquids in, 139, 140f, cylindrical, 208, 208f for venous occlusive diseases, 190–191
141, 141f encapsulated, 209–211, 209f–211f Lasers, tissue cutting, 20
postoperative surface tension inert versus toxic, 206, 206f Lens
management, 142 large, 208–209, 208f management
results of, 142–143 lens removal, 207 in diabetic retinopathy, 175–176
retinopexy, 141–142, 142f localization of, 8 in double penetrating injury, 212
Glaucoma moderately sized, 207, 207f in giant retinal breaks, 138
air (gas) pupillary block, 245 retinopexy, 211 in proliferative vitreoretinopathy, 145
ciliary block scleral buckling, 211 in severe uveitis, 224
anterior vitrectomy for, 91–92 sclerotomies, 207 removal of
erythroclastic (hemolytic), 185 surface tension management, 211 conjunctival displacement, 46–47
erythroclastic (hemolytic) glaucoma, 245 surgery for eye, stabilization of, 46
neovascular, 245 sequence and techniques for, 206–212 timing of, 213
filtering procedures for, 246 timing of, 206 in trauma, 207
open-angle, 246 vitrectomy for, 207 wound construction, 46
phacolytic wound repair, 206 wound leaks, 47
256 Index

Lens capsule defects Operating microscope, 29–30, 29f presurgical anesthesia for, 239
in recognition and management of, 84–85 diathermy, 30 recognition, 239–240
Lens loops endophotocoagulation, 29 surgical indications for, 240
vitreous traction from, 88, 88f Operating room environment, 18–29 toxicity, 239
Lens material aspiration fluidics and vitreous cutting, Perfluorocarbon liquids, 151–152, 152f
posterior dislocation of 22–23, 23f in floating of lens material, 89, 89f
in cataract surgery, 86, 87f–89f, 88–90 instrument storage, 18–19 in vitrectomy
Lens opacities personnel, 19 for giant retinal breaks, 139, 140f,
slit lamp preoperative examination and, 5 presurgical equipment testing, 19 141, 141f
Lensectomy. See Pars plana lensectomy scheduling, 18 Perfluoron, 135–136, 135f, 136f
Limbal approach tissue cutting, 19–22, 20f, 21f Peribulbar (extraconal) anesthesia, 42
in anterior vitrectomy, 78 video recording, 19 Persistent fetal vasculature, 231–232, 232f
Liquid perfluorocarbon techniques, 62–64, 63f vitrectomy technique Persistent hyperplastic primary vitreous. See
Local anesthesia. See Anesthesia, local aspiration technology, 25–27, 25f Persistent fetal vasculature
cutter technology, 24–25, 24f Phacoemulsification
M endoillumination, 28, 28f with pars plana lensectomy, 79
Macular edema infusion devices, 27–28 vitrectomy procedures, 99t
cystoid, 92–93, 161–162 infusion fluid, 28 concepts of, 99–100
diabetic, 172–173 physics of, 24 principles of, 100–101
Macular holes role of, 23–24, 24f silicone oil issues, 99
arcuate retinotomy, 169, 169f Operating room setup, 30–31, 30f for silicone oil-filled eyes, 101, 102
biologic modifiers, 169 prep technique, 31 techniques, 101
hole closure mechanisms, 166 sterilization systems, 31 Phacolytic glaucoma, 93
indications for, 164 surgical effciency, 31 Photocoagulation
internal limiting membrane peeling, 166, Ophthalmoscopy laser. See Laser photocoagulation
167f, 168 indirect Photodynamic therapy
ocular coherence tomography, 164–165 preoperative, 6 for choroidal neovascular membranes, 194
pathogenesis of, 164 Optical coherence tomography, 13–14, 14f, 15f Phototoxicity, 38
posterior vitreous detachment creation Pneumatic retinopexy, 121–122, 122f, 123f
methods, 166, 167f P Polymerase chain reaction, 222
postoperative positioning, 169 Pain Ports
results of, 169 during vitreoretinal surgery, 42 configuration of, 26
surface tension management, Panretinal photocoagulation, 183 Posterior segment surgery. See Posterior
168–169, 168f Pars plana approach vitrectomy
surgery for, 165–166 in anterior vitrectomy, 78 Posterior vitrectomy
Macular translocation, 196, 201–202 Pars plana capsulectomy under air, 68, 69f
Maddox rod after lensectomy, 83 antibiotic injection in, 73–74
in preoperative assessment, 4 Pars plana capsulotomy in corneal-scleral laceration, 213
Medical equipment companies after lensectomy, 83 endodiathermy, 72–73
interaction with, 250–251 Pars plana lensectomy epiretinal membrane dissection in, 48–58. See
Membrane peeling development of, 78 also Epiretinal membrane dissection
methods of. See Epimacular membranes; endocapsular, 80–83, 80f–82f fluorescein angioscopy, 73–74
Epiretinal membrane dissection fragmenter performance, 79, 79f infusion fluid in, 45
Membranectomy, 90–91 fragmenter techniques in, 78–79 laser endophotocoagulation, 72–73
dense, 91 indications for, 78 lens removal in, 46
Membranotomy, 90 infusion options in, 79 objectives of, 47–48, 48f
Metamorphopsia lens capsule removal in, 79 pupillary dilation in, 45–46
in preoperative assessment, 4 phacoemulsification with, 79 retinopexy, 73
Microscope sulcus in, 80 scleral buckling in, 74
operating, 29f, 29–30 Patient speculum, 45
placement of, 30 draping of, 31 stabilized surgery in, 54, 55f
prep technique for, 31 strategies of, 45
N Patient movement viscodissection in, 53–54
Needles prevention of, 40–41 visualization in, 45–47
infusion, 28 Patient preparation Post-operative follow-up, 248
Neovascular glaucoma, 245 psychological, 41 Povidone-iodine (Betadine), 31
filtering procedures for, 246 Peak electron avalanche knife, 20 Power scissors and forceps, 54
Neovascularization Pediatric cataracts, 90 Practice surgery, 249
in diabetic retinopathy, 185–186 Pediatric 25-gauge vitrectomy, 229–230 Preoperative assessment
Pediatric traction retinal detachments bright flash electroretinography in, 15
O persistent fetal vasculature, 231–232, 232f in cardiopulmonary disease, 3
Obesity, 3 results of, 235 color discrimination, 4
Obstructive sleep apnea syndrome, 3 Toxocara canis, 232, 233f, 234f, 235 contrast sensitivity, 4
Ocular coherence tomography, 164–165 Penetrating injury in diabetes mellitus, 2–3
Open globe delayed vitrectomy in, 240 entoptic phenomenon, 4
anesthesia in, 43 immediate action in, 240 indirect ophthalmoscopy in, 6
Open-angle glaucoma, 246 office injections, 239 laser interferometry, 4
Index 257

Maddox rod, 4 Retinal breaks/detachment, 8 Retinotomy


metamorphopsia, 4 diabetic retinopathy and, 174, 179–183 drainage, 61
obesity and obstructive sleep apnea epimacular membrane and, 160, 161 Retrolental membranes
syndrome, 3 laser prophylaxis for, 118–119 intraocular lens, 93
projection and field testing, 4 management of, 247 Retroprosthetic membranes, keratoprosthesis,
psychophysical testing in, 4 nonrhegmatogenous, 247 238, 238f
pupil testing, 5 postoperative endophthalmitis and, 111 Rhegmatogenous retinal detachment, 161, 187
retinal configuration, 7 prevention of, 126 in necrotizing retinitis, 223
slit lamp biomicroscopy, 6 prophylaxis of, 126
slit lamp examination in, 5–6 rhegmatogenous, 161, 187 S
two-point discrimination, 4 in necrotizing retinitis, 223 Scissors
visual acuity, 3–4 suprachoroidal hemorrhage, 243 power, 35, 35f, 54
visual function testing, 3 traction, 226 segmentation and delamination, 180–181,
vitreous clarity, 7 in intermediate and posterior uveitis, 180f, 181f
vitreous configuration, 6–7 223–224 Scleral buckling
Pressure pediatric, 231–235 anesthesia for, 43
intraocular treatment of for diabetic retinopathy, 185
slit lamp preoperative assessment of, 5–6 evidence-based, 126 for double penetrating injury, 212
Presurgical evaluation and imaging, 2–15 vitrectomy for in posterior segment techniques, 74
Projection and field testing, 4 indications for, 131 in proliferative vitreoretinopathy, 145, 152
Proliferative retinopathy medium-term perfluoron for, 135–136, prophylaxis of, 126–130, 127f–129f
diabetic retinopathy, 173–174 135f, 136f with broad encircling buckle, 129f, 130
Proliferative vitreoretinopathy silicone oil in, 136 case selection, 126–127
compartmentalization in, 145 surface tension management for, 135 circumferential explants, 128, 129f
and intentional incarceration, 153–154 surgical sequence for, 131, 132f–134f, with encircling band, 127
lens management in, 145 134–135 implants versus explants, 127
operability, 145 Retinal neovascularization sponges versus hard silicone, 127, 127f
retinal sutures, 153–154 presurgical evaluation in, 8 with vitreous surgery, 127, 128f
retinectomy for, 150–152, 151f, 152f for retinopathy of prematurity, 226 retinopathy of prematurity, 230
retinopexy in, 154, 154f Retinal pigment epithelium transplantation, subretinal fluid drainage, 70–71, 70f, 71f
retinoplasty in, 154 200–201 in trauma, 211
scleral buckling for, 145 Retinectomy, 150–152, 151f, 152f, 183 Scleral defects, 130
silicone oil, 153 Retinopathy of prematurity Scleral incisions, 112–113, 113f
surgical sequence in, 145 epiretinal membrane management, 230 Scleral laceration, 212–213
tacks, 153–154 25-gauge pediatric vitrectomy Sclerotomies
vitrectomy for three-port versus two-port in trauma, 207
anterior dissection in, 146, 147f vitrectomy, 230 wound construction and closure, 115–116
complications of, 154–155 trocar cannulas, 229–230 Silicone infusion, 64–65, 65f
epiretinal membranes in, 146–148, 148f without cannulas, 230 Silicone oil, 101, 183
results of, 154 infants intraocular lens calculation issues, 102
subretinal proliferation, 148–150, anti-vascular endothelial growth factor phacoemulsification vitrectomy
149f, 150f therapy, 229 procedures, 99
viscoelastics as vitreous substitutes, 153 4b management, 228 in proliferative vitreoretinopathy, 153
Psychological preparation with inadequate preoperative laser, 229 in vitrectomy for retinal detachment, 136
for local anesthesia, 41 laser failures, 228 Slit lamp biomicroscopy
Psychophysical testing, 4 laser treatment for, 227 preoperative, 6
Pupil service, 228–229 Slit lamp examination
preoperative assessment of, 5 stage 5 management, 228 preoperative, 5–6
Pupillary block stage 4a management, 227–228 Sonification
aphakic, anterior vitrectomy for, 91 telemedicine in, 229 with ultrasonic fragmentation, 77
Pupillary dilation pathophysiology of Sterilization systems, 31
in posterior vitrectomy, 45–46 directional vasculogenesis, 225–226 Steroid glaucoma, 246
Pupillary membranes retinal neovascularization, 226 Steroids
dense membranectomy, 91 traction retinal detachment, 226 for endophthalmitis, 219
membranectomy, 90–91 vascular development, 226 in posterior vitrectomy, 73–74
membranotomy (discission), 90 vascular endothelial growth factor, 225 Subfoveal membranes
scleral buckling, 230 submacular surgery for, 196
R vascular development, 225 Subluxation
Radio frequency identification, 33 Retinopexy lens
Real-time ultrasonography, 12 for diabetic retinopathy, 185 during vitrectomy, 83, 84f
Reattachment experiment, 61–62, 150–151, for double penetrating injury, 212 Submacular exudates
151f for giant retinal breaks, 141–142, 142f in diabetic retinopathy, 175
Retina posterior vitrectomy, 73 Submacular hemorrhage
configuration of in proliferative vitreoretinopathy, 154, 154f displacement of, 195
preoperative assessment of, 7, 7t prophylactic, 126 Submacular surgery
slit lamp biomicroscopy of, 6 in trauma, 211 and macular translocation, 196, 196f
Retinal biopsy, 222–223 Retinoplasty, 154 for subfoveal membranes, 196
258 Index

Subretinal fluid drainage Traction retinal detachment vitrectomy for, 191


air-gas exchange, 62, 63f in diabetic retinopathy, 174 Video recording
direct transscleral needle, 70–71, 70f, 71f pediatric, 231 intraoperative, 19
external versus internal, 70 Transscleral diathermy, 73 Viscodissection
fluid-air exchange and, 59, 60f Trauma in diabetic retinopathy, 181–182
giant retinal breaks, 64 cellular migration/proliferation in, 205 in posterior vitrectomy, 53–54
indications for, 69–70 complications of Viscoelastics
interface vitrectomy, 65–68, 66f–68f corneal opacification, 213–214, 214f in proliferative vitreoretinopathy, 153
interfacial tension agents, 59 glaucoma, 214 Visual acuity testing, 3–4
internal, 61, 61f infection, 213 Visual evoked potential, 15
intraocular lens fogging during, 62, 62f corneal-scleral lacerations Visual function testing, 3
liquid perfluorocarbon techniques, 62–64, 63f lens removal, timing of, 213 Visualization
medium-term perfluoron, 64, 64f posterior vitrectomy, 213 25-gauge vitrectomy, 109
postoperative positioning, 69 wound repair, 213 and illumination, 37–39
in reattachment experiment, 61–62 double penetrating injury posterior vitrectomy, 45–47
retinotomy for, 61 initial repair, 212 Vitrectomy
silicone infusion, 64–65, 65f surgical sequence and techniques, 212 under air, 68, 69f
silicone removal, 65, 66f vitrectomy, 212 cellulose sponge, 85, 86f
silicone reoperations, 65 intraocular foreign bodies and, 206–212 for diabetic retinopathy, 174, 176
soft-tip extrusion cannula, 60f substrates for proliferation, 205 for double penetrating injury, 212
surface tension management, 58–59, 58f vitrectomy for endophthalmitis, 218, 219
vitrectomy under air, 68, 69f timing of, 205 25-gauge
Suprachoroidal hemorrhage Traumatic cataracts, 90 cannula withdrawal and wound closure, 111
acute management, 241 Triamcinolone case selection, 103
complications of, 241, 242f intravitreal cutter design options, 106
incidence of, 241 venous occlusive diseases, 189 fluidics, 104–106, 106f
pathogenesis and prevention, 241 medium-term perfluoron and, 64, 64f 20-gauge combined, 107–108, 107f, 108f
retinal detachment, 243 Trocar-cannula system, 103–104, 104f, 105f, 237 postoperative endophthalmitis and retinal
surgical management of, 243, 243f retinopathy of prematurity, 229–230 detachments, 111
vitrectomy, indications for, 242 Two-point discrimination tool issues, 106–107, 106f, 107f
Surface tension management, 58–59, 58f preoperative assessment of, 4 trocar-cannula system, 103–104, 104f, 105f
for diabetic retinopathy, 183–184 visualization, 109
for double penetrating injury, 212 U wound leak issues, 109, 109f, 110f, 111
macular holes, 168–169, 168f Ultrasound interface, 65–68, 66f–68f
postoperative, 142 diagnostic keratoprosthesis, 237–238
for retinal breaks/detachment, 135 A-scan versus B-scan, 12 outcomes analysis, 249–250
in trauma, 211 B-scan, 12 phacoemulsification, 98–102
Surgical data, 250 clinical acoustic physics, 9, 9f–11f, 12 phacoemulsification and, 98–102
Surgical self-education examination methods in, 13 for retinal detachment, 131–136
companies, interaction with, 250–251 gray scale, 13 subluxated lenses during, 83, 84f
correspondence, 250 image archiving and viewing, 13 submacular hemorrhage displacement, 195
group education, 250 limitations, 13 for suprachoroidal hemorrhage, 242
outcomes analysis, 249–250 real-time, 12 techniques of
surgical data, 250 three-dimensional, 12 and anterior segment problems, 76–96
Sutures vector scanning, 13 in trauma
extrusion of fragmentation intraocular foreign bodies, 207
in vitreoretinal surgery, 248 in anterior vitrectomy, 76–78 timing of, 205
Uveitis, 93 uveitis
T lens management in, 224 diagnostic, 221–222
Tenon’s capsule, 116 vitreoretinal surgery in therapeutic, 223–224
Tissue cutting diagnostic vitrectomy, 221–222 for venous occlusive diseases, 191
elongation and, 20 drug delivery systems, 224 Vitreocorneal touch
enzyme-assisted approaches, 20–21 retinal biopsy, 222–223 anterior vitrectomy for, 91
high IOP effects, 22 therapeutic vitrectomy, 223–224 Vitreomacular interface disorders, 224
inertial cutting in, 20 Vitreomacular traction syndrome, 157–162
infusion cannula placement, 22 V Vitreoretinal complications
infusion fluidics, 21–22, 21f Vacuum cleaning–extrusion, 56–58, 57f keratoprosthesis, 236–238
infusion system technologies, 22 Vacuum systems Vitreoretinal examination
low IOP effects, 22 for anterior vitrectomy, 77 indirect ophthalmoscopy in, 6
sharpness and, 20 Vascular endothelial growth factor (VEGF) Vitreoretinal surgery
shear and, 20 antagonists anesthesia and, 42
surgical fluidics, 21 diabetic macular edema, 173 complications of
Tomography in retinal vascular development, 225 cataract, 248
ocular coherence, 164–165 Vascular occlusion corneal, 244
optical coherence, 13–14, 14f, 15f in vitreoretinal surgery, 247 cyclocryopexy, 245–246
Toxocara canis Venous occlusive diseases endophthalmitis, 246
traction retinal detachments, 232, 233f, laser therapy for, 190–191 follow-up intervals, 248
234f, 235 pharmacotherapy for, 189–190 glaucoma, 245
Index 259

inflammation, 244–245 hypocellular contraction, 6 20-gauge sutures


intraocular hemorrhage, 247 persistent hyperplastic primary, 231–232, infusion cannula placement, 113–115,
iris neovascularization, 245 232f 114f, 115f
neovascular glaucoma, 246 Vitreous bands, 6 scleral incisions, 112–113, 113f
retinal complications, 246–247 Vitreous cortex instrument sclerotomies, 115–116
suture extrusion, 248 fibrovascular proliferation and postoperative medication, 116, 116f
in uveitis management, 221–224 in diabetic retinopathy, 187 traction sutures, 112
Vitreoretinal traction Vitreous cutters. See Tissue cutting Wound leaks, 47
minimization of Vitreous hemorrhage Wound repair
in vitrectomy, 24, 24f in diabetic retinopathy, 174 in corneal-scleral laceration, 213
Vitreoretinopathy Vitreous infusion suction cutter, 27 in trauma, 206
proliferative. See Proliferative Vitreous loss
vitreoretinopathy in cataract surgery, 85–86, 86f, 87f X
Vitreous Vitreous microsurgery. See specific techniques and Xenon illuminations systems, 33, 33f
clarity of indications
preoperative assessment of, 7 Z
configuration of W Zonulorhexis
preoperative assessment of, 6–7 Wound construction and closure in anterior vitrectomy, 83, 83f
in fragmenter conjunctiva and tenon’s capsule, 116
avoidance of, 83, 84f conjunctival incisions, 112

Вам также может понравиться