Download as pdf or txt
Download as pdf or txt
You are on page 1of 211

Any screen.

Any time.
Anywhere.
Activate the eBook version
of this title at no additional charge.

Expert Consult eBooks give you the power to browse and find content,
view enhanced images, share notes and highlights—both online and offline.

Unlock your eBook today.


1 Visit expertconsult.inkling.com/redeem Scan this QR code to redeem your
eBook through your mobile device:
2 Scratch off your code
3 Type code into “Enter Code” box

4 Click “Redeem”
5 Log in or Sign up
6 Go to “My Library”
Place Peel Off
It’s that easy! Sticker Here

For technical assistance:


email expertconsult.help@elsevier.com
call 1-800-401-9962 (inside the US)
call +1-314-447-8200 (outside the US)
Use of the current edition of the electronic version of this book (eBook) is subject to the terms of the nontransferable, limited license granted on
expertconsult.inkling.com. Access to the eBook is limited to the first individual who redeems the PIN, located on the inside cover of this book,
at expertconsult.inkling.com and may not be transferred to another party by resale, lending, or other means.
2015v1.0
Atlas of
RETINAL OCT
This page intentionally left blank
Atlas of
RETINAL OCT
Darin R. Goldman MD
Partner, Retina Group of Florida
Affiliate Associate Professor
Charles E. Schmidt College of Medicine
Florida Atlantic University
Boca Raton, FL, USA

Nadia K. Waheed MD, MPH


Assistant Professor of Ophthalmology
New England Eye Center
Tufts Medical Center
Tufts University School of Medicine
Boston, MA, USA

Jay S. Duker MD
Director, New England Eye Center
Professor and Chairman
Department of Ophthalmology
Tufts Medical Center
Tufts University School of Medicine
Boston, MA, USA

For additional content visit

Expertconsult

Edinburgh London New York Oxford Philadelphia St Louis Sydney Toronto


© 2018 Elsevier Inc. All rights reserved.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic or
mechanical, including photocopying, recording, or any information storage and retrieval system, without
permission in writing from the publisher. Details on how to seek permission, further information about
the Publisher’s permissions policies and our arrangements with organizations such as the Copyright
Clearance Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/
permissions.

This book and the individual contributions contained in it are protected under copyright by the Publisher
(other than as may be noted herein).

ISBN 978-0-323-46121-4
eISBN 978-0-323-46122-1

Notices

Knowledge and best practice in this field are constantly changing. As new research and experience
broaden our understanding, changes in research methods, professional practices, or medical treatment
may become necessary.

Practitioners and researchers must always rely on their own experience and knowledge in evaluating
and using any information, methods, compounds or experiments described herein. Because of rapid
advances in the medical sciences, in particular, independent verification of diagnoses and drug dosages
should be made. To the fullest extent of the law, no responsibility is assumed by Elsevier, authors, editors
or contributors for any injury and/or damage to persons or property as a matter of products liability,
negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas
contained in the material herein.

The
publisher’s
policy is to use
paper manufactured
from sustainable forests

For Elsevier

Content Strategist: Russell Gabbedy


Content Development Specialist: Joshua Mearns
Project Manager: Andrew Riley
Designer/Design Direction: Christian Bilbow

Printed in China
Contents

PART 1: NORMAL OPTICAL COHERENCE TOMOGRAPHY Section 10: Hydroxychloroquine Toxicity............................68


Chapter 10.1: Hydroxychloroquine Toxicity..........68
Section 1: Normal Optic Nerve.............................................1 Section 11: Vitelliform Macular Dystrophy...........................72
Chapter 1.1: Normal Optic Nerve..........................1 Chapter 11.1: Vitelliform Dystrophy......................72
Section 2: Normal Retina......................................................4 Section 12: Macular Telangiectasia......................................73
Chapter: 2.1: Time-Domain OCT...........................4 Chapter 12.1: Macular Telangiectasia..................73
Chapter: 2.2: Spectral Domain OCT......................5 Section 13: Isolated Cystoid Macular Edema......................76
Chapter: 2.3: Swept-Source OCT..........................6 Chapter 13.1: Isolated Cystoid Macular
Section 3: Normal Choroid....................................................7 Edema..............................................................76
Chapter 3.1: Normal Choroid.................................7 Section 14: Other Disorders Affecting the Macula..............78
Section 4: Normal Vitreous ..................................................9 Chapter 14.1: Angioid Streaks.............................78
Chapter 4.1: Normal Vitreous...............................9 Chapter 14.2: X-Linked Juvenile Retinoschisis.....79
Section 5: OCT: Artifacts and Errors..................................10 Chapter 14.3: Oculocutaneous Albinism..............80
Chapter 5.1: OCT: Artifacts and Errors................10 Chapter 14.4: Subretinal Perfluorocarbon............82
Chapter 5.2: OCT Angiography Artifacts..............13

PART 3: VASOOCCLUSIVE DISORDERS


PART 2: ISOLATED MACULAR DISORDERS
Section 15: Diabetic Retinopathy.........................................84
Section 6: Age-Related Macular Degeneration.................16 Chapter 15.1: Diabetic Macular Edema...............84
Chapter 6.1.1: Drusen.........................................16 Chapter 15.2: Nonproliferative Diabetic
Chapter 6.1.2: Geographic Atrophy.....................24 Retinopathy......................................................86
Chapter 6.1.3: Isolated Pigment Epithelial Chapter 15.3: Proliferative Diabetic
Detachment......................................................28 Retinopathy......................................................88
Chapter 6.2.1: Type 1 Choroidal Neovascular Section 16: Retinal Venous Occlusive Disease ..................90
Membrane........................................................30 Chapter 16.1: Branch Retinal Vein
Chapter 6.2.2: Type 2 Choroidal Neovascular Occlusion.........................................................90
Membrane........................................................32 Chapter 16.2: Central Retinal Vein Occlusion......92
Chapter 6.2.3: Type 3 Choroidal Neovascular Section 17: Retinal Arterial Occlusive Disease....................96
Membrane........................................................34 Chapter 17.1: Branch Retinal Artery
Chapter 6.2.4: Subretinal Hemorrhage................35 Occlusion.........................................................96
Chapter 6.2.5: Disciform Scar.............................36 Chapter 17.2: Central Retinal Arterial
Chapter 6.2.6: Retinal Pigment Epithelial Tear.....37 Occlusion.........................................................98
Chapter 6.2.7: Polypoidal Choroidal
Vasculopathy....................................................38
PART 4: UVEITIS AND INFLAMMATORY DISORDERS
Section 7: Vitreomacular Interface Disorders....................40
Chapter 7.1: Vitreomacular Adhesion..................40 Section 18: Noninfectious Uveitis.......................................100
Chapter 7.2: Vitreomacular Traction....................42 Chapter 18.1.1: Birdshot
Chapter 7.3: Full-Thickness Macular Hole...........47 Retinochoroidopathy.......................................100
Chapter 7.4: Lamellar Macular Hole....................50 Chapter 18.1.2: Acute Posterior Multifocal
Chapter 7.5: Epiretinal Membrane.......................52 Placoid Pigment Epitheliopathy.......................103
Section 8: Central Serous Chorioretinopathy....................55 Chapter 18.1.3: Multiple Evanescent White
Chapter 8.1: Central Serous Dot Syndrome ...............................................107
Chorioretinopathy.............................................55 Chapter 18.1.4: Serpiginous Choroiditis............109
Section 9: Myopic Degenerative Maculopathies...............59 Chapter 18.1.5: Multifocal Choroiditis
Chapter 9.1: Myopic Choroidal Neovascular and Panuveitis and Punctate Inner
Membrane........................................................59 Choroidopathy................................................111
Chapter 9.2: Myopic Macular Schisis..................62 Chapter 18.2: Vogt-Koyanagi-Harada
Chapter 9.3: Dome-Shaped Macula....................64 Disease...........................................................114
Chapter 9.4: Posterior Staphyloma......................66 Chapter 18.3: Sympathetic Ophthalmia.............116

v
Section 19: Infectious Uveitis.............................................119 PART 7: INHERITED RETINAL DEGENERATIONS
Chapter 19.1: Toxoplasmic Chorioretinitis.........119
Chapter 19.2: Acute Syphilitic Posterior Section 26: Retinal Dystrophies.........................................148
Placoid Chorioretinitis.....................................121 Chapter 26.1: Retinitis Pigmentosa....................148
Chapter 19.3: Tuberculosis................................124 Chapter 26.2: Stargardt Disease.......................149
Contents

Chapter 19.4: Posterior Scleritis........................127 Chapter 26.3: Best Disease...............................152


Chapter 19.5: Candida Chorioretinitis................129 Chapter 26.4: Cone Dystrophy..........................154
Chapter 19.6: Acute Retinal Necrosis Chapter 26.5: Malattia Leventinese
Syndrome.......................................................132 (Doyne’s Honeycomb Retinal
Dystrophy)......................................................155
PART 5: RETINAL AND CHOROIDAL TUMORS Chapter 26.6: Central Areolar Choroidal
Dystrophy.......................................................157
Section 20: Choroidal Tumors............................................134
Chapter 20.1: Choroidal Nevus.........................134 PART 8: VITREOUS DISORDERS
Chapter 20.2: Choroidal Melanoma...................135
Chapter 20.3: Solitary Choroidal Section 27: Posterior Vitreous Detachment.......................159
Hemangioma..................................................136 Chapter 27.1: Stages of Posterior Vitreous
Section 21: Retinal Tumors.................................................137 Detachment....................................................159
Chapter 21.1: Retinal Capillary Section 28: Asteroid Hyalosis.............................................162
Hemangioma..................................................137 Chapter 28.1: Asteroid Hyalosis........................162
Section 22: Retinal Pigment Epithelium Tumors...............138 Section 29: Vitreous Hemorrhage.......................................164
Chapter 22.1: Simple Hamartoma of Chapter 29.1: Vitreous Hemorrhage..................164
the RPE..........................................................138 Section 30: Vitreous Inflammation......................................166
Chapter 22.2: Combined Hamartoma of Chapter 30.1: Vitreous Inflammation..................166
the Retina and RPE........................................139
Section 23: Metastatic Choroidal Tumors.........................140 PART 9: MISCELLANEOUS RETINAL DISORDERS
Chapter 23.1: Choroidal Metastases.................140 Section 31: Peripheral Retinal Abnormalities....................170
Chapter 31.1: Tractional Retinal
PART 6: TRAUMA
Detachment....................................................170
Section 24: Mechanical Trauma.........................................142 Chapter 31.2: Rhegmatogenous Retinal
Chapter 24.1: Valsalva Retinopathy...................142 Detachment....................................................174
Section 25: Photic Maculopathy.........................................145 Chapter 31.3: Bullous Retinoschisis...................180
Chapter 25.1: Laser Maculopathy......................145 Chapter 31.4: Lattice Degeneration...................183
Chapter 25.2: Solar Maculopathy......................146 Chapter 31.5: Myelinated Nerve Fiber Layer......186

vi
Preface

Optical coherence tomography (OCT) continues to occupy an can stand alone as an independent reference. We sought to
ever-expanding role in the ophthalmic community. OCT is widely include a breadth of retinal conditions with a focus on those most
available and forms a requisite portion of the comprehensive applicable to everyday clinical practice. However, a wide array
ophthalmic evaluation, particularly as it pertains to the retina. of pathology is included to also illustrate unique, less common
Although still a relatively young technology that continues to OCT findings. Each condition is illustrated with numerous, large,
evolve, OCT has become widely accepted. This acceptance is high-quality OCT images to highlight disease pathology and aid
due to its non-invasive nature, ease of image acquisition, and in disease identification. Additional imaging modalities, such as
wealth of information that it affords. The quantity of information fundus photographs and fluorescein angiograms, are included
conveyed within a typical OCT scan is immense, which can be to supplement OCT images where appropriate.
daunting to both the beginner and experienced clinician. Atlas of Retinal OCT provides the reader with a high quality,
Atlas of Retinal OCT grew out of the success of Handbook of easy-to-follow visual aid to incorporating OCT scans into the
Retinal OCT. The Atlas expands on the images and material in evaluation and care of your patients. The atlas is designed to
the handbook, while maintaining a similar and consistent layout make OCT more comprehensible for both the novice and expert
that will be familiar to the reader. This atlas was created to serve clinician. We hope that the reader finds this to be a handy and
as a supplement to the original text, although the atlas certainly practical addition to your everyday reference armamentarium.

vii
This page intentionally left blank
Contributors

A YASIN ALIBHAI, MD NORA W. MUAKKASSA, MD


OCT Research fellow, Ophthalmology, New England Eye New England Eye Center, Tufts Medical Center, Hospital de
Center, Tufts Medical Center, Boston, Massachusetts, USA Olhos do Paraná, Curitiba, Brazil

CAROLINE R. BAUMAL, MD CARLOS A. MOREIRA NETO, MD, PhD


New England Eye Center, Tufts Medical Center, Boston, New England Eye Center, Tufts Medical Center, Hospital de
Massachusetts, USA Olhos do Paraná, Curitiba, Brazil

SHILPA DESAI, MD, FRCP EDUARDO A. NOVAIS, MD


Assistant Professor, Ophthalmology, New England Eye Department of Ophthalmology, Federal University of São
Center/Tufts University Medical Center, Boston, MA, USA Paulo, School of Medicine, São Paulo, Brazil

IVANA N. DESPOTOVIC, MD CARL REBHUN, BA


New England Eye Center, Tufts University School of Medicine, New England Eye Center, Tufts Medical Center, Tufts
Boston, MA, USA University School of Medicine, Boston, USA

JAY S. DUKER, MD LUIZ ROISMAN, MD


Director, New England Eye Center, Professor and Chairman, Department of Ophthalmology, Federal University of São
Department of Ophthalmology, Tufts Medical Center, Tufts Paulo, School of Medicine, São Paulo, Brazil
University School of Medicine, Boston, MA, USA
EDUARDO UCHIYAMA, MD
DANIELA FERRARA, MD, PhD Retina Group of Florida, Boca Raton, FL, USA
Assistant Professor of Ophthalmology, Tufts University School
of Medicine, Boston, MA, USA NADIA K. WAHEED, MD, MPH
Assistant Professor of Ophthalmology, New England Eye
DARIN R. GOLDMAN, MD Center, Tufts Medical Center, Tufts University School of
Partner, Retina Group of Florida, Affiliate Associate Professor, Medicine, Boston, MA, USA
Charles E. Schmidt College of Medicine, Florida Atlantic
University, Boca Raton, FL, USA

ix
This page intentionally left blank
Acknowledgments

A project such as this requires contributions from many different also like to thank the many co-authors who have contributed
groups and individuals to be successful. First and foremost, to various chapters throughout the atlas. Additionally, thanks
the images used in this atlas would not be possible without are due to our fellows whose archive of cases and interesting
our many patients. We are very grateful to these individuals images were invaluable to this project. Specifically, we would like
who trust their care in our hands on a daily basis. Additionally, to thank Dr. Chris Or, who provided invaluable feedback on the
we rely on the talented photographers and technical staff at final chapters. Lastly, the professionalism and expertise of the
both the New England Eye Center at Tufts Medical Center and staff at Elsevier is unmatched. We want to thank the entire team
Retina Group of Florida to obtain the majority of the included at Elsevier who were critical to the completion of this project,
OCT images. Their expertise is reflected in the volume of high in particular Russell Gabbedy, Humayra Rahman Khan, Joshua
quality images available for inclusion in this project. We would Mearns, and Andrew Riley.

Dedications
To the memory of my dear sister Candice, whose love, strength
and determination live on in all that she touched. And to my
daughter, Rona, who has added immeasurable joy to our lives.
D.R.G.

To Jujie, Memsie and Ammi, without whom none of this would


have been possible.
N.K.W.

To my colleagues at the New England Eye Center who have


assisted me in bringing innovation to eye care for over 3 decades.
J.S.D.

xi
This page intentionally left blank
SECTION 1: NORMAL OPTIC NERVE

Normal Optic Nerve


Carlos A. Moreira Neto | Carl Rebhun 1.1
Spectral domain OCT (SD-OCT) devices have two scan patterns Optic Nerve Morphology
to analyze the optic nerve head (ONH): volume scans and line
scans. SD-OCT devices also calculate optic nerve diameter, area, cup,
and rim measurements (see Fig. 1). Each measurement varies
according to age (Cavallotti et al. 2002) and ethnicity (Girkin 2008).
Volume Scans According to Budenz et al. (2007), the mean RNFL thickness in
Volume scans acquire a volumetric set of data, centered at a normal population is 100.1 µm. Thinner RNFL measurements
the ONH. It delineates the optic disc margin and optic disc were associated with older age. Caucasians had slightly thinner
surface contour and is segmented to obtain the retinal nerve RNFL thickness than Hispanics or Asians. Persons with smaller
fiber boundaries. Each device has its own scanning protocol. optic disc areas also have thinner RNFL thickness.
The Cirrus HD-OCT identifies the center of the optic disc and
creates a 3.46-mm circle on this location and calculates the Line Scans
thickness of the retinal nerve fiber layer (RNFL). The Heidelberg
Spectralis creates a cylindrical volume with a diameter of 3.4 mm Aiming to obtain a higher resolution visualization of structure and
through and around the ONH (Duker, Waheed & Goldman 2014). anatomic anomalies at the ONH, line scans provide a single or a
The Optovue RTVue’s protocol for the ONH consists of a grid series of high-resolution B-scans similar to the scans obtained
pattern with circular and radial scans that acquires a 4- × 4-mm in the macula (Fig. 3).
volume around the optic nerve. Because different machines use OCT angiography (OCTA) (Fig. 4) allowed for a greater under-
circles of different diameters around the center of the ONH, the standing of optic disc vasculature and peripapillary vessel density.
measurement of RNFL between machines is not comparable This information provides insight into the role of this vascular
(Duker et al. 2014). bed in the functioning of the RNFL.

REFERENCES
Retinal Nerve Fiber Layer Thickness (RNFL) Budenz DL, Anderson DR, Varma R, et al. Determinants of normal retinal
nerve fiber layer thickness measured by Stratus OCT. Ophthalmology.
OCT devices calculate RNFL thickness as the distance between 2007;114(6):1046–1052.
the internal limiting membrane and the outer aspect of the RNFL Cavallotti C, Pacella E, Pescosolido N, et al. Age-related changes in the
human optic nerve. Can J Ophthalmol. 2002;37(7):389–394.
(Fig. 1).
Duker JS, Waheed NK, Goldman DR. Scanning Principles. Handbook of Retinal
OCT. St Louis: Elsevier; 2014.
Ganglion Cell Complex Girkin CA. Differences in optic nerve structure between individuals of predomi-
nantly African and European ancestry: Implications for disease detection
The ganglion cell complex (GCC) consists of the thickness of and pathogenesis. Clin Ophthalmol. 2008;2(1):65–69.
three inner retinal layers: the NFL, the ganglion cell layer, and the
inner plexiform layer. The scan is centered at the fovea, and the
software presents the results as a color-coded map, comparing
to a normative database (Fig. 2).

1
ONH and RNFL OU Analysis: Optic Disc Cube 200¥200 OD OS
RNFL Thickness Map ! OD OS
350
Section 1: Normal Optic Nerve

Average RNFL Thickness 102 m X

RNFL Symmetry X

Rim Area 1.88 mm2 X


175
Disc Area 2.17 mm2 X

Average C/D Ratio 0.36 X

Vertical C/D Ratio 0.32 X


0 m
Cup Volume 0.009 mm3 X

RNFL Deviation Map


Neuroretinal Rim Thickness
m OD
800

400

0
TEMP SUP NAS INF TEMP

Disc Center (0.09,0.12) mm


RNFL Thickness
Extracted Horizontal Tomogram
m OD

200

100

TEMP SUP NAS INF TEMP


Extracted Vertical Tomogram
Diversified:
121 Distribution of Normals

S NA 95% 5% 1%

86 T N 72

I RNFL
Quadrants
RNFL Circular Tomogram 129
93
162 109
113 80

64 65 RNFL
Clock
80 70 Hours
174 74
137

FIG. 1. Normal peripapillary RNFL, neuroretinal rim thickness, and disc area measurements using SD-OCT.

2
GCC Right / OD
1.1
Thickness Map Signal Strength Index 73 NDB Reference Map
250 6 mm × 6 mm

Average Thickness Thickness (m)


Total 89.28
Superior 88.52

Normal Optic Nerve


Inferior 90.03
200 Intra Eye Difference (S-I) –1.51
FLV (%) 1.058
GLV (%) 6.029

150

T N

Fovea
100

50

p > 5% Within Normal


0 m p > 5% Borderline
p < 1% Outside Normal

FIG. 2. Normal color-coded ganglion cell complex (GCC) thickness using SD-OCT.

FIG. 3. Line scan of the ONH. FIG. 4. OCT angiograph image (3 × 3 mm) of the ONH.

3
SECTION 2: NORMAL RETINA

Time-Domain OCT
Carlos A. Moreira Neto | Carl Rebhun 2.1
The first OCT image, published by Huang et al. (1991), was REFERENCES
captured using a device that detected light echoes using time Huang D, Swanson EA, Lin CP, et al. Optical coherence tomography. Science.
1991;254(5035):1178–1181.
domain detection. In time domain OCT (TD-OCT) the reference Duker JS, Waheed NK, Goldman DR. Scanning principles. In: Handbook of
arm, with a physically moving mirror, and a sample arm undergo Retinal OCT. St Louis: Elsevier; 2014.
interference, which is used to generate an A-scan. Multiple
A-scans obtained linearly are combined to generate a cross-
sectional B-scan (Duker et al. 2014).

4
Spectral Domain OCT
Carlos A. Moreira Neto | Carl Rebhun 2.2
Summary Because the retinal pigment epithelium (RPE) is highly hyper-
reflective with OCT imaging, there is limited penetration of light
In spectral domain OCT (SD-OCT), a spectral interference beyond it, decreasing the resolution of the choroid (Schuman,
pattern between the reference beam and the sample beam is Fujimoto & Duker 2013). Normal mean central foveal thickness is
obtained simultaneously by a spectrometer and an array detec- approximately 225 ± 17 µm as measured by SD-OCT, although
tor. Unlike time domain (TD)-OCT, SD-OCT does not require this varies with age and retinal status.
a physically moving reference mirror, instead using frequency
information to produce interference patterns. This allows for REFERENCE
much faster acquisition and higher quality images than those with Schuman J, Fujimoto J, Duker J. Optical Coherence Tomography of Ocular
TD-OCT. Diseases. 3rd ed. Thorofare NJ: Slack Inc.; 2013.
The high resolution provided by SD-OCT allows for visualization
of the microscopic anatomy of the retina (Fig. 1) with more detail
than with TD-OCT.

Ganglion cell Outer


layer plexiform Outer
Inner plexiform layer nuclear
layer Inner layer Nerve
nuclear fiber
layer layer

Photoreceptors
Choriocapillaris outer segments
RPE/Bruch’s
complex
External
IS/OS/EZ Myoid limiting
zone membrane
S
3
T N

FIG. 1. Normal macula imaged using SD-OCT. IS/OS/EZ, Inner segment/outer segment/ellipsoid zone; RPE, retinal pigment epithelium.

5
Swept-Source OCT
Carlos A. Moreira Neto | Carl Rebhun 2.3
Summary penetration of the choroid (Fig. 1). In SS-OCT, a narrow-band
light source is rapidly swept through a wide range of frequencies.
Swept source OCT (SS-OCT) is a modified Fourier-domain and The interference pattern is detected on a single or small number
depth-resolved technology that offers potential advantages over of receivers as a function of time.
SD-OCT, including reduced sensitivity roll-off with imaging depth,
higher detection efficiencies, improved imaging range, and better

Inner
Ganglion
plexiform Outer cell
layer plexiform layer
Inner layer
nuclear Outer Nerve
layer nuclear fiber
layer layer

Choriocapillaris
Choroidal larger
vessels
External Myoid
limiting zone
membrane
IS/OS/ RPE/
EZ Bruch’s Choroid/
complex sclera
Photoreceptors junction
outer
segments

FIG. 1. Normal retina imaged using SS-OCT. EZ, ellipsoid zone; IS, inner segments; OS, outer segments; RPE, retinal pigment epithelium.

6
SECTION 3: NORMAL CHOROID

Normal Choroid
Carlos A. Moreira Neto | Carl Rebhun 3.1
Summary The choroid is divided into three layers, the choriocapillaris
or smaller blood vessels, Sattler’s layer, and Haller’s layer, or
Enhanced depth imaging (EDI) on commercially available OCT larger blood vessels (Fig. 2).
devices allows for higher quality images of the choroid (Fig. 1).
EDI mode moves the zero-delay line of the spectral domain REFERENCES
(SD)-OCT closer to the choroid, enabling better visualization of Margolis R, Spaide RF. A pilot study of enhanced depth imaging optical
choroidal structures and a more precise measurement of choroidal coherence tomography of the choroid in normal eyes. Am J Ophthalmol.
2009;147(5):811–815.
thickness than standard OCT scanning protocols. This is useful for Fujiwara A, Shiragami C, Shirakata Y, et al. Enhanced depth imaging spectral-
diseases such as central serous chorioretinopathy, in which the domain optical coherence tomography of subfoveal choroidal thickness in
choroidal-scleral interface may be difficult to visualize. Studies of normal Japanese eyes. Jpn J Ophthalmol. 2012;56(3):230–235.
choroidal thickness in normal subjects and those with pathologic
processes have shown a wide variation in measurements (Fujiwara
et al. 2012; Margolis & Spaide 2009).

7
Section 3: Normal Choroid

Choriocapillaris
Larger choroidal Choroid
Stroma
vessels
S S
3 3
T N
Lumens of larger blood
T N
vessels
Choroid/sclera
I I junction
A B
FIG. 1. Chorioretinal OCT image not using EDI (A) and using EDI (B).

Larger choroidal vessels

A B

FIG. 2. En face structural OCT images of choriocapillaris (A) and Haller/Sattler layers (B).

8
SECTION 4: NORMAL VITREOUS

Normal Vitreous
Nadia K. Waheed 4.1
Summary Key OCT Features
Until recently, the anatomy of the vitreous could not be imaged In OCT of a normal retina the following vitreous structures may
in vivo. With the use of OCT, a better view and understanding be observed:
of vitreous structure has become possible. Along with normal • Posterior cortical vitreous (posterior hyaloid) (Fig. 2)
structure, abnormal vitreous processes such as vitreomacular trac- • Retrohyaloid space: Created after posterior vitreous detachment
tion have been revealed (Duker et al. 2013). High dynamic range (Fig. 2).
imaging as well as enhanced vitreous imaging techniques, present • Premacular bursa: Liquid space overlying the macula, caused
on most commercially available OCT devices, allow visualization by liquefaction of the vitreous (Fig. 3).
of the fluid-filled spaces as well as the collagenous and cellular
structure of the vitreous. Secondary features of vitreous debris REFERENCE
Duker JS, Kaiser PK, Binder S, et al. The International Vitreomacular Traction
are also often identifiable on SD-OCT (Fig. 1).
Study Group classification of vitreomacular adhesion, traction, and macular
hole. Ophthalmology. 2013;120(12):2611–2619.

Posterior cortical vitreous (posterior hyaloid)

Retrohyaloidal space

S
1
N T

FIG. 2. Posterior hyaloid and retrohyaloid spaces.

Vitreous

B Premacular bursa Vitrepapillary adhesion

FIG. 1. Vitreous opacity (arrows) demonstrates shadowing on SS-OCT. FIG. 3. Premacular bursa in a normal patient using SD-OCT.

9
SECTION 5: OCT: ARTIFACTS AND ERRORS

OCT: Artifacts and Errors


Carlos A. Moreira Neto | Carl Rebhun 5.1
Artifacts can occur during image acquisition or analysis because of • The Early Treatment Diabetic Retinopathy Study (ETDRS) grid
patient, operator, or software factors. Accurate image interpreta- usually can be moved to obtain an accurate measure of the
tion depends on the quality of the image and an understanding foveal thickness.
of the various artifacts that can affect an OCT image (Duker,
Waheed & Goldman 2014).
Software Breakdown
Mirror Artifact • OCT segmentation lines are incorrectly drawn because there
is misidentification of the inner or outer retinal boundaries.
• Occurs only on spectral domain (SD)-OCT. • Vitreomacular surface disorders (epiretinal membrane, vitreo-
• Occurs when the area of interest crosses the zero-delay line macular traction) could cause inner line breakdown.
and results in an inverted image. • Outer retinal/retinal pigment epithelium disorders (age-related
• Reasons macular degeneration, cystoid macular edema) might cause
1. OCT device is pushed too close to the eye. outer line breakdown (Fig. 4).
2. Conditions in which the curvature of the retina is such that
it crosses the zero-delay line, such as retinoschisis, retinal
detachment, an elevated choroidal lesion, or high myopia
Blink Artifact
(Fig. 1). • If a patient blinks during image acquisition, loss of data occurs.
• OCT scans and volumetric maps both show black or white
bars (Fig. 5).
Vignetting
• Occurs when the iris blocks a part of the OCT beam. Motion Artifact
• Loss of signal is seen over one side of the image (Fig. 2).
• Occurs when there is movement of the eye during scan
acquisition.
Misalignment
• OCT image shows distortion or double scanning of the
• This occurs when the fovea is not centered during the volu- same area.
metric scan (Fig. 3). • Blood vessels are misaligned (Fig. 6).
• Most common reason is a patient with poor fixation or incorrect • The fovea may be duplicated.
placement of fixation target by operator. • This is much less common due to better eye tracking software
on current OCT machines.

Real image

Mirror image

Fovea

FIG. 1. Mirror artifact occurring in a high myopic eye.

FIG. 2. Vignetting: Loss of signal over the left side of the image. FIG. 3. Misalignment error. The fovea is not centered because of an
eccentric fixation.

10
5.1

OCT: Artifacts and Errors


A

FIG. 4. Software breakdown caused by choroidal neovascularization (A) FIG. 6. Motion artifact.
and geographic atrophy (B).

Blink artifact

FIG. 5. Blink artifact.

11
Out of Range Error
• Occurs when the B-scan is not centered in the preview screen,
resulting in it being shifted out of the scanning range.
• A section of the OCT scan is cut off (Fig. 7).
Section 5: OCT: Artifacts and Errors

REFERENCE
Duker JS, Waheed NK, Goldman DR. Artifacts on OCT. Handbook of Retinal
OCT. St Louis: Elsevier; 2014.

FIG. 7. Out-of-range error. Due to improper positioning of the machine


during image acquisition, the outer retina and the choroid are cut off.

12
OCT Angiography Artifacts
Carlos A. Moreira Neto | Carl Rebhun 5.2
Artifacts are very common in OCT angiography, and their Segmentation Errors (Fig. 6)
identification is important for appropriate image interpretation
(Ferrara, Waheed & Duker). • Caused by PED, macular edema, or other pathologic process
that disrupts the horizontal alignment of retinal layers.

Blockage Artifacts (Fig. 1)


Shadowing Artifacts (Fig. 7)
• Blockage artifacts are caused by lesions that affect light
penetration through ocular tissues, including both the anterior • Usually appear in choriocapillary segmentation.
and posterior segments. • Caused by PED, hemorrhage, floaters.
• Anterior segment blockage can be cause by cataract, inflam-
REFERENCES
mation, or corneal scar. Ferrara D, Waheed NK, Duker JS. Investigating the choriocapillaris and choroidal
• Posterior segment blockage can be caused by intravitreal vasculature with new optical coherence tomography technologies. Prog
hemorrhage or inflammation, floaters, intraretinal or subretinal Retin Eye Res. 2016;52:130–155.
hemorrhage, pigment epithelial detachment (PED), or large Spaide RF, Fujimoto JG, Waheed NK. Image Artifacts in Optical Coherence
Tomography Angiography. Retina. 2015;35(11):2163–2180.
drusen.

White Line Artifacts (Fig. 2)


• Caused by transverse ocular movements.
• A major cause of artifacts in OCT angiography.
False Positive Flow
• Ocular movements are in the axial direction (arterial pulsation).
• An OCT dataset may be displaced and may have enough
decorrelation to cause the appearance of flow (Ferrara, Waheed
& Duker 2016; Spaide, Fujimoto & Waheed 2015).

Quilting Defects (Fig. 3)


• Related to software correction of ocular movement.
• Caused by multiple saccades in the horizontal and vertical
directions.

False Negative Flow


• Caused by blood flow below a given threshold.
• Vessels seem absent even if they are present.
Projection Artifacts (Fig. 4)
• Superficial vessels are seen in deep and choroidal slabs when
they are not actually present in those slabs (Ferrara et al.
2016; Spaide et al. 2015).

Vessel Duplication (Fig. 5)


• Result of a breakdown in registration of the X and Y scans.
• Caused by eye movement. FIG. 1. Blockage artifact causing a focal loss of signal.

13
White line
Section 5: OCT: Artifacts and Errors

FIG. 2. White line artifact. FIG. 3. Quilting artifact.

FIG. 4. Projection artifact on deep plexus. Vessels from the superficial FIG. 5. Vessel duplication.
plexus (arrows) are seen in the deep plexus.

14
5.2

OCT Angiography Artifacts


FIG. 6. Segmentation error (green line) caused by a pigment epithelial detachment.

FIG. 7. Shadowing artifact (arrow) in the choriocapillaris segmentation.

15
SECTION 6: AGE-RELATED MACULAR DEGENERATION

Drusen
Ivana N. Despotovic | Daniela Ferrara 6.1.1
Summary OCT imaging of refractile drusen (drusenoid material containing
small refractile spherules) show hyperreflective dots (many small
Drusen are focal yellow or white deposits of extracellular debris spherules rich in calcium phosphate) and appear to be a stage
located between the retinal pigment epithelium (RPE) and of drusen regression marked by loss of RPE, thus contributing
Bruch’s membrane. They occur naturally with age and usually to the development of GA (Suzuki et al. 2015).
are asymptomatic. Drusen are the hallmark of age-related
macular degeneration (AMD) and the most common early sign
of nonexudative AMD. Esterified and unesterified cholesterol are Key Points
significant components of the lipid-rich lesions associated with
AMD (basal linear deposits and soft drusen) and comprise more
• Small drusen (“drupelets”) are less than 63 µm in diameter,
intermediate drusen are 63 to 125 µm, and large drusen are
than 40% of hard druse volume (Curcio et al. 2011). greater than 125 µm.
Drusen may range in appearance, size, and location. Hard
drusen are smaller and have distinct margins (Figs. 2, 3, 4, 5, 6,
• Small drusen are considered normal aging and do not represent
a risk for progression to advanced AMD.
10, and 12). Soft drusen are larger, mound-like elevations that
may have a diameter greater than 1000 µm, with margins that
• Subretinal drusenoid deposits (also known as reticular pseu-
dodrusen) are located above the RPE and are associated with
are not clearly defined (Figs. 1, 2, 3, 4, 7, 8, 9, and 11). A large progression to advanced AMD.
number of round and punctate cuticular drusen give a “stars
in the sky” appearance. Cuticular drusen have a spheroid or
• OCT is valuable in the differential diagnosis of drusen.

triangular shape on OCT (Fig. 12).


• Specific drusen features on OCT are investigated as biomarkers
for AMD progression.
The tomographic features of drusen on OCT have been
extensively investigated in natural history studies as potential REFERENCES
biomarkers for AMD progression, although some features have Abdelfattah NS, Zhang H, Boyer DS, et al. Drusen volume as a predictor of
yet to be validated. Among drusen characteristics defined on disease progression in patients with late age-related macular degeneration
cross-sectional OCT, drusen shape, internal reflectivity, and in the fellow eye. Invest Ophthalmol Vis Sci. 2016;57(4):1839–1846.
Casswell AG, Kohen D, Bird AC. Retinal pigment epithelial detachments in
substructures can be cited as some of the relevant biomarkers with
the elderly: classification and outcome. Br J Ophthalmol. 1985;69(6):397–
increased risk for development to advanced AMD (Yehoshua et al. 403.
2011; Veerappan et al. 2016). Drusen size and confluency have Cukras C, Agrón E, Klein ML, et al. Age-Related Eye Disease Study Research
been historically associated with the progression of AMD. More Group. Natural history of drusenoid pigment epithelial detachment in age-
recently, drusen volume has been assessed through automatic related macular degeneration: Age-Related Eye Disease Study Report No.
28. Ophthalmology. 2010;117(3):489–499. doi:10.1016/j.ophtha.2009.12.002.
OCT algorithms, which also appears to be relevant in disease Curcio CA, Johnson M, Rudolf M, et al. The oil spill in ageing Bruch membrane.
progression (Abdelfattah et al. 2016). Br J Ophthalmol. 2011;95(12):1638–1645. doi:10.1136/bjophthalmol-2011-
Drusenoid pigment epithelial detachment (PED) is formed by 300344.
the confluence of large areas of soft drusen (Figs. 7, 8, and 11) Huisingh C, McGwin G Jr, Neely D, et al. The Association between subretinal
drusenoid deposits in older adults in normal macular health and incident age-
and is part of the clinical spectrum of AMD (Casswell, Kohen, &
related macular degeneration. Invest Ophthalmol Vis Sci. 2016;57(2):739–745.
Bird 1985). The natural history of eyes containing drusenoid PED doi:10.1167/iovs.15-18316.
is characterized by a high rate of progression to both geographic Mrejen S, Sato T, Curcio CA, et al. Assessing the cone photoreceptor
atrophy (GA) and neovascular AMD (Cukras et al. 2010). mosaic in eyes with pseudodrusen and soft Drusen in vivo using adap-
Subretinal drusenoid deposits (SDDs, also known as reticular tive optics imaging. Ophthalmology. 2014;121(2):545–551. doi:10.1016/j.
ophtha.2013.09.026. [Epub 2013 Oct 30].
pseudodrusen, can be confounded with drusen, but are actually Suzuki M, Curcio CA, Mullins RF, et al. Refractile Drusen: Clinical imaging
a clinically distinct entity located above the RPE (Figs. 3, 4, and and candidate histology. Retina. 2015;35(5):859–865. doi:10.1097/
10). OCT is considered a fundamental imaging modality to identify IAE.0000000000000503.
and characterize SDD (Suzuki, Sato & Spaide 2014; Zweifel et al. Suzuki M, Sato T, Spaide RF. Pseudodrusen subtypes as delineated by
multimodal imaging of the fundus. Am J Ophthalmol. 2014;157(5):1005–1012.
2010). SDD in older eyes with a normal macular appearance, as
Veerappan M, El-Hage-Sleiman AM, Chiu SJ, et al. Optical coherence
defined by the Age-Related Eye Disease Study (AREDS) scale, tomography reflective drusen substructures predict progression to
is a risk factor for further development of AMD (Huisingh et al. geographic atrophy in age-related macular degeneration. Ophthalmology.
2016). Reduced visibility of cones overlying SDD in adaptive 2016;123(12):2554–2570.
optics images can be due to several possible causes, including Yehoshua Z, Wang F, Rosenfeld PJ, et al. Natural history of drusen morphology
in age-related macular degeneration using spectral domain optical coherence
a change in photoreceptors’ orientation, an alteration of their tomography. Ophthalmology. 2011;118(12):2434–2441.
cellular architecture, or absence of the cones themselves, imply- Zweifel SA, Spaide RF, Curcio CA, et al. Reticular pseudodrusen are subretinal
ing decreased cone photoreceptor function (Mrejen et al. 2014). drusenoid deposits. Ophthalmology. 2010;117(2):303–312.

16
6.1.1

Drusen
A

FIG. 1. (A) Color fundus photograph of soft drusen with indistinct margins. (B) Cross-sectional OCT B-scan shows soft drusen.

A B

FIG. 2. (A) Color fundus photograph of multiple large soft drusen and distinct small hard drusen. Reticular pseudodrusen are present in the superior
portion on the macula. (B) OCT B-scan shows confluent soft drusen and a few hard drusen.

17
FIG. 3. OCT B-scans of the same eye depicted in Fig. 2. Reticular
pseudodrusen are evident on the right side of the scans, in addition to
confluent soft drusen and a few hard drusen, that are also visible in
Fig. 2.
Section 6: Age-Related Macular Degeneration

S
1
N T

S
1
N T

A B

FIG. 4. (A) Color fundus photograph showing hard and soft drusen, as well as reticular pseudodrusen. (B) Red-free fundus photograph of the same
eye.

18
Cross Line Signal Strength index 54 Right/OD 6.1.1

Drusen
250m

# of Averages, 20, 20 Auto Zoom 30.00mm Scan Length

FIG. 5. En face and cross-sectional wide-field OCT B-scan of the same eye depicted in Fig. 4, showing distinct drusen.

A B

FIG. 6. (A) Color fundus photograph showing hard and soft drusen. (B) OCT B-scan shows hard and soft drusen.

19
Section 6: Age-Related Macular Degeneration

A B

FIG. 7. (A) Color fundus photograph of multiple, confluent, large, soft drusen. (B) Near-infrared imaging of the same eye. (C) OCT B-scan shows
confluent soft drusen and a drusenoid pigment epithelium detachment.

20
6.1.1

Drusen
A B C

FIG. 8. (A) Color fundus photograph with multiple, confluent, large, soft drusen. (B) Fundus autofluorescence.(C) Blue-light reflectance of the same
eye. (D) OCT B-scan showing confluent soft drusen with a drusenoid pigment epithelium detachment. A posterior hyaloid membrane is also visible.

21
Section 6: Age-Related Macular Degeneration

A B C

FIG. 9. (A) Color fundus photograph of the


fellow eye of the patient depicted in Fig. 8,
with multiple, confluent, large soft drusen.
Fundus autofluorescence (B) and blue-light
reflectance of the same eye (C). (D) OCT
D B-scan shows confluent soft drusen. A
posterior hyaloid membrane is also visible.

A C

D F

FIG. 10. (A) Color fundus photograph of hard and large, soft drusen. Reticular pseudodrusen are also evident in the superotemporal aspect of the
macula. (B) Red-free image of the same eye. (C) Near-infrared imaging, showing the reticular pattern of reticular pseudodrusen. (D) OCT B-scan
22 shows drusen and reticular pseudodrusen. (E) Fluorescein angiography showing staining drusen. (F) Indocyanine green angiography showing
hypofluorescent and hyperfluorescent patterns related to drusen and reticular pseudodrusen.
6.1.1

Drusen
FIG. 11. Wide-field 12-mm cross-sectional OCT B-scan shows large, confluent, soft drusen, with a drusenoid PED.

A B

C D

FIG. 12. (A) and (B) Color fundus photographs of hard and cuticular cluster-like drusen. (C) and (D) OCT B-scan of the same patient shows drusen
as closely packed blunted triangles.

23
Geographic Atrophy
Ivana N. Despotovic | Daniela Ferrara 6.1.2
Summary in the outer nuclear layer (Zweifel et al. 2009). En face OCT
imaging of the outer retina may visualize the areas with inner
GA is characterized by sharply demarcated, round or oval areas segment/outer segment (IS/OS) band disruption and can predict
of hypopigmentation or depigmentation of the RPE (Figs. 1, 2, 4, the progression of GA in some eyes (Nunes et al. 2013).
5, 6). Choroidal vessels are more visible within areas of GA than
in adjacent areas. The minimum lesion diameter that characterizes
a GA lesion varies in the literature, but many authors agree that Key Points
it must be at least 175 µm in diameter. The atrophy in overt GA
is not limited to the RPE alone; it involves the photoreceptors • GA is characterized on OCT by loss of the hyperreflective
and choriocapillaris as well. The sequence of pathologic events external band (attenuation of the RPE layer) and/or by the
that lead to GA is still not completely understood. GA progresses hypertransmission effect (increased OCT signal penetration
gradually, and in most cases the fovea is initially spared. Drusen into the choroid secondary to loss of outer retinal layers, RPE,
regression may be a sign of progression toward GA (Schlanitz and choriocapillaries).
et al. 2017). Based on currently revised classification schemes, • Nascent GA is a recently described entity, based on specific
GA is a manifestation of advanced AMD, even if the fovea is OCT features that precede manifestations on clinical examina-
spared, because the lesion causes irreversible loss of visual tion or other imaging modalities and portend the development
function (Ferris et al. 2013). of drusen-associated GA
Hypopigmentation is manifested by increased transmission
of the reflected OCT signal beneath the RPE layer. However, the REFERENCES
Ferris FL 3rd, Wilkinson CP, Bird A, et al. Clinical classification of age-related
so-called “hypertransmission signal” on OCT is associated with
macular degeneration. Ophthalmology. 2013;120(4):844–851. doi:10.1016/j.
marked loss of cells in the RPE, photoreceptors, and choriocapil- ophtha.2012.10.036. [Epub 2013 Jan 16].
laries layers and has been considered the tomographic definition Nunes RP, Gregori G, Yehoshua Z, et al. Predicting the progression of
of GA on OCT (Figs. 2, 3, 4, 5, and 6). geographic atrophy in age-related macular degeneration with SD-OCT en
A new entity termed “nascent GA” was recently described face imaging of the outer retina. Ophthalmic Surg Lasers Imaging Retina.
2013;44(4):344–359. doi:10.3928/23258160-20130715-06.
based on OCT findings. It has been described to be a precursor of Schaal KB, Gregori G, Rosenfeld PJ. En face optical coherence tomography
drusen-associated GA and is identified on OCT before presentation imaging for the detection of nascent geographic atrophy. Am J Ophthal-
of overt GA in the clinical examination or other imaging modalities. mol. 2017;174:145–154. doi:10.1016/j.ajo.2016.11.002. [E-pub ahead of
Nascent GA has unique tomographic characteristics related to print].
Schlanitz FG, Baumann B, Kundi M, et al. Drusen volume development over
abnormalities at the level of the inner and outer nuclear layers
time and its relevance to the course of age-related macular degeneration.
and represents a risk for GA development when associated with Br J Ophthalmol. 2017;101(2):198–203. pii: bjophthalmol-2016-308422.
pigment abnormalities and the presence of GA in the fellow eye doi:10.1136/bjophthalmol-2016-308422. [Epub ahead of print].
(Schaal, Gregori, & Rosenfeld 2017; Wu et al. 2014). Wu Z, Luu CD, Ayton LN, et al. Optical coherence tomography-defined changes
Outer retinal tubulation (ORT) contains degenerating photo- preceding the development of drusen-associated atrophy in age-related
macular degeneration. Ophthalmology. 2014;121(12):2415–2422.
receptors and is commonly found in advanced AMD. It may be Zweifel SA, Engelbert M, Laud K, et al. Outer retinal tubulation: a novel optical
identified on cross-sectional OCT scans as a hyperreflective coherence tomography finding. Arch Ophthalmol. 2009;127(12):1596–1602.
circular or ovoid structure with a hyporeflective center, located doi:10.1001/archophthalmol.2009.326.

24
6.1.2

Geographic Atrophy
A B C

FIG. 1. (A) Color fundus photograph of GA secondary to age-related macular degeneration involving the center of the fovea, with a sharply
demarcated area of RPE depigmentation with visible choroidal vessels. (B) Fundus autofluorescence shows the hypoautofluorescent area of GA
resulting from the loss of photoreceptors and retinal pigment epithelium. (C) En face OCT scan of geographic atrophy shows a large area of
hyperreflectivity with distinct margins and visible choroidal blood vessels.

A B

S
1
T N

I C D

FIG. 2. (A) and (B) Color fundus photographs of bilateral GA involving the center of the fovea in both eyes, with sharply demarcated areas of retinal
pigment epithelium (RPE) loss and visible choroidal blood vessels. (C) and (D) Cross-sectional OCT scan shows loss of the outer nuclear layer,
photoreceptors, and RPE in the area of GA, causing overall thinning of the neurosensory retina, as well as apparent loss of choriocapillaris. The OCT
signal is increased below the level of the RPE, corresponding to the so-called hypertransmission effect.

25
Macula Thickness: Macular Cube 512 ¥ 128 OD OS Macula Thickness: Macular Cube 512 ¥ 128 OD OS
500 500
275 276
400 400
274 271

300 255 280 301 246 297 300 300 270 163 250 257

260 255
200 200
275 272
Section 6: Age-Related Macular Degeneration

100 100
ILM-RPE Thickness (m) Fovea: 263, 56 ILM-RPE Thickness (m) Fovea: 264, 63

0 m 0 m
Overlay: ILM-RPE Transparency: 50% Overlay: ILM-RPE Transparency: 50%

ILM-RPE ILM-RPE

T N N T

ILM ILM
Diversified: Diversified:
Distribution Distribution
of Normals of Normals

99% 99%
95% 95%
5% 5%
1% 1%

RPE RPE
S S

Central Cube Central Cube


Cube Cube
Subfield Average Subfield Average
Volume Volume
I Thickness Thickness I Thickness Thickness
(mm3) (mm3)
(m) (m) (m) (m)

ILM-RPE 201 9.4 264 ILM-RPE 161 9.6 270

FIG. 3. Macular cube scan of both eyes of the same patient depicted in Fig. 2, showing thickness maps, en face OCT, and cross-sectional OCT
scans. Decreased thickness is evident in the center of the macula. En face OCT shows the area of GA. Cross-sectional OCT scan demonstrates loss
of outer nuclear layer, photoreceptors, and retinal pigment epithelium in the areas of GA, causing thinning of the neurosensory retina. The OCT signal
is increased below the level of the RPE, corresponding to the hypertransmission effect. ILM, Inner limiting membrane.

A B C

FIG. 4. (A) Color fundus photograph of multifocal GA involving the center of the fovea, showing RPE depigmentation with visible choroidal vessels.
In the temporal aspect of the macula, hard, soft and calcified drusen are present. (B) Fundus autofluorescence with the hypoautofluorescent areas
of GA secondary to loss of the photoreceptors and RPE. (C) En face OCT scan shows multifocal areas of GA and heterogeneous reflectivity in the
topography of drusen. (D) Cross-sectional scan shows loss of outer nuclear layer, photoreceptors, RPE, and choriocapillaries in the areas of GA, with
26 thinning of the neurosensory retina. The hypertransmission effect is present. On the left and right side of the scan, drusen are also visible.
6.1.2

Geographic Atrophy
A B C

S
1
N T

I D

FIG. 5. (A) Color fundus photograph of multifocal geographic atrophy (GA), with retinal pigment epithelium (RPE) depigmentation and visible
choroidal vessels. Hard and soft drusen are present. (B) Fundus autofluorescence with the hypoautofluorescent areas of GA secondary to loss of
photoreceptors and RPE. (C) En face OCT scan shows the multifocal areas of GA. (D) Cross-sectional OCT scan with loss of outer nuclear layer,
photoreceptors, RPE, and choriocapillaries in the areas of GA, with thinning of the neurosensory retina. A few drusen are present in the scan. Note
irregularities at the level of the photoreceptors throughout the scan.

FIG. 6. (A) Color fundus photograph of


multifocal GA, with RPE depigmentation
and visible choroidal vessels. Hard,
soft, and calcified drusen are present,
as well as reticular pseudodrusen.
(B) Near-infrared fundus reflectance
shows the areas of GA surrounded by
multiple bright spots corresponding to
pigmentary abnormalities associated
with drusen. (C) Cross-sectional
OCT scan with loss of outer nuclear
layer, photoreceptors, RPE, and
choriocapillaries in the area of GA,
with collapse of the inner retina over
the lesion and marked thinning of the
neurosensory retina, affecting the foveal
contour. The hypertransmission effect is
A B evident. Note presence of drusen and
reticular pseudodrusen, associated with
irregularity of the photoreceptor layer,
throughout the OCT scan beyond the
borders of the GA lesion.

C
T N

I 27
Isolated Pigment Epithelial Detachment
Ivana N. Despotovic | Daniela Ferrara 6.1.3
Summary with CSC) or hyperreflective (e.g., fibrovascular PED associated
with neovascular AMD). An OCT finding of layered, hyperre-
Retinal PED results from a separation between the RPE basement flective, sub-RPE lines (described as the “onion sign”) suggest
membrane and the inner collagenous layer of Bruch’s membrane. the presence of cholesterol crystal precipitation in an aqueous
PED may be idiopathic or occur in association with many chorio- environment (Pang et al., 2015). This finding is usually associated
retinal diseases, including central serous chorioretinopathy (CSC) with intraretinal hyperreflective foci (RPE and lipid-filled cells in
and AMD. In patients with AMD, PED may be associated with the retina) and is a common presentation of fibrovascular PED.
drusen, RPE hyperpigmentation, subretinal fluid, or subretinal
hemorrhage.
Although often associated with an underlying disorder, PED
Key Points
may occasionally be an isolated finding, with no known primary • PED may be associated with underlying conditions such as
diagnosis. In this case it is usually not associated with subretinal CNV or CSC.
or intraretinal fluid. The pathophysiology of isolated PED is not • Isolated PED is not associated with a known underlying condi-
completely understood. Patients can be asymptomatic if the tion and is usually asymptomatic, but should be followed
lesion does not affect the fovea (Fig. 1). An isolated PED can closely because of the potential risk for further development
resolve completely, sometimes leaving RPE atrophy. However, of CNV.
it should be monitored because of a potential risk for further • OCT is useful in the differential diagnosis of isolated PED and
development of symptomatic choroidal neovascularization (CNV). PED secondary to underlying conditions; internal reflectivity
OCT features of an isolated PED usually include a dome-shaped of PED is helpful in the differential diagnosis.
elevation of the RPE layer lining over an optically clear space
(Figs. 1, 2B, and 4) or homogenous hyperreflective material (Fig. BIBLIOGRAPHY
Pang CE, Messinger JD, Zanzottera EC, et al. The onion sign in neovascular
2A). In an isolated PED, the overlying retina is usually adherent age-related macular degeneration represents cholesterol crystals. Oph-
to the RPE and no signs of exudation or leakage are observed thalmology. 2015;122(11):2316–2326. doi:10.1016/j.ophtha.2015.07.008.
(Figs. 1–4). In contrast, a PED associated with underlying condi- [Epub 2015 Aug 19].
tions may manifest as hyporeflective (e.g., serous PED associated

FIG. 1. Cross-sectional OCT of an isolated PED.

A B

FIG. 2. Cross-sectional OCT showing bilateral isolated PED in an 87-year-old patient. (A) The PED is filled with homogeneous hyperreflective material.
Although the layer corresponding to the retinal pigment epithelium seems preserved, there is hypertransmission below the PED. No intraretinal or
subretinal fluid is present in this scan. (B) The fellow eye of the same patient. Multilobulated dome-shaped isolated PED.

28
6.1.3
Macula Thickness OU: Macular Cube 512 ¥ 128 OD OS
OD ILM-RPE Thickness Map OS ILM-RPE Thickness Map
500

Isolated Pigment Epithelial Detachment


400

300

200

100

0 m
Fovea: 249, 78 Fovea: 270, 66
!
OD OCT Fundus OD ILM-RPE Thickness OS ILM-RPE Thickness OS OCT Fundus
Diversified: 278
Distribution
274
of Normals
347
324 99%
300 340 235 313 264
266 330 248 319 313 95%
314
5%
332
1%
246

ILM-RPE OD OS
Thickness Central Subfield (m) 248 235
Volume Cube (mm3) 10.1 10
Thickness Avg Cube (m) 282 277
OD Horizontal B-Scan BScan: 78 OS Horizontal B-Scan BScan: 66

FIG. 3. Macular cube scan of the same patient depicted in Fig. 2, showing the thickness map and retinal thicknesses in subfields across the macular
area, in en face OCT and cross-sectional OCT scans. ILM, Inner limiting membrane.

A B

FIG. 4. Isolated PED in a 66-year-old patient. (A) En face OCT demonstrates PED with drusen and pigmentary changes. (B) Cross-sectional OCT
scan showing multilobulated isolated PED. Note the relative thinning of the outer nuclear layer above the PED.

29
Type 1 Choroidal Neovascular Membrane
A. Yasin Alibhai | Daniela Ferrara 6.2.1
Summary Key Points
Type 1 CNV involves the formation of abnormal blood vessels that • Type 1 CNV is classified based on its anatomic location, being
develop from the choroid, extend through Bruch’s membrane, present above Bruch’s membrane but below the RPE.
and arborize beneath the RPE. On fluorescein angiography, • On OCT, type 1 CNV manifests as PED and commonly multi-
type 1 CNV is characterized as “occult CNV,” manifesting as lobulated with variable internal reflectivity.
stippled fluorescence (leakage from undetermined source) or • On fluorescein angiography, type 1 CNV manifests as “occult
as fibrovascular PED (Fig. 1). CNV,” with stippled fluorescence or fibrovascular PED.
These neovascular lesions are commonly associated with AMD. • Type 1 CNV is commonly associated with AMD, but they can
They can, however, also develop as a result of other pathologic also be secondary to other conditions such as trauma, CSC,
processes, such as central serous chorioretinopathy (CSC). and pseudoxanthoma elasticum.
Type 1 CNV lies within the sub-RPE space and results in
elevation of the overlying RPE. The most characteristic OCT REFERENCES
Keane PA, Patel PJ, Liakopoulos S, et al. Evaluation of age-related macular
findings include irregular PEDs, occasionally with thickening of
degeneration with optical coherence tomography. Surv Ophthalmol.
the overlying RPE (Figs 2 and 3). These PEDs may present vari- 2012;57:389–414.
able internal reflectivity, ranging from hyporeflective or optically Roisman L, Zhang Q, Wang RK, et al. optical coherence tomography angi-
empty spaces to hyperreflective, usually heterogeneous internal ography of asymptomatic neovascularization in intermediate age-related
reflectivity. On OCT scans with high quality, the intravascular macular degeneration. Ophthalmology. 2016;123(6):1309–1319. doi:10.1016/j.
ophtha.2016.01.044.
lumens of the CNV complex may be documented, giving the
appearance of “stacked” material. In active, exudative CNVs, BIBLIOGRAPHY
associated subretinal and/or intraretinal fluid may also be present Freund KB, Zweifel SA, Engelbert M. Do we need a new classification for
(Keane et al. 2012). choroidal neovascularization in age-related macular degeneration? Retina.
The diagnosis of inactive, nonexudative type 1 CNVs may be 2010;30(9):1333–1349.
challenging because they can be confounded with drusenoid
PED or nonneovascular PED (Fig. 4). On OCT, they commonly
appear as small PEDs with increased internal reflectivity. The
nonexudative neovascular complex can be identified only with
OCT angiography (Roisman et al. 2016).

30
6.2.1

Type 1 Choroidal Neovascular Membrane


Early Middle Late

FIG. 1. Fluorescein angiography shows progressive hyperfluorescence, with dye accumulation within a fibrovascular PED surrounded by stippled
fluorescence with unclear margins. This is the characteristic angiographic presentation of type 1 CNV.

Sub-retinal
fluid

Irregular pigment
epithelial detachment

FIG. 2. OCT scan corresponding to the eye depicted in Fig. 1. The line scan shows an irregular PED, which is characteristic of a type 1 CNV lesion.
The heterogeneous internal reflectivity of the PED and the presence of subretinal fluid also suggest this is an active (exudative) type 1 CNV.

Sub-retinal Flat irregular pigment


fluid epithelial detachment

Pigment epithelial
detachment

FIG. 3. OCT line scan shows a small PED with relatively heterogeneous FIG. 4. OCT line scan showing a flat, irregular PED characteristic of a
internal reflectivity that is associated with subretinal fluid, characteristic of type 1 CNV lesion. Also seen is the stacked nature of the material within
active (exudative) type 1 CNV. the CNV. Presence of fluid clefts within the PED (not present here) is
very suggestive of an active type 1 lesion.

31
Type 2 Choroidal Neovascular Membrane
A. Yasin Alibhai | Daniela Ferrara 6.2.2
Summary hemorrhage, and subretinal fluid or intraretinal fluid may also
be present (Fig. 3).
Type 2 CNV extends through the subretinal space, between the
neurosensory retina and the RPE. It is characterized on fluorescein
angiography as “classic CNV,” showing in the early phases of Key Points
the angiogram the fine contour of the neovessels and in the
late stages of the angiogram marked hyperfluorescence from
• Type 2 CNV is classified based on its anatomic location, being
present below the neurosensory retina but above the RPE.
profuse leakage (Fig. 1). On OCT, type 2 CNV typically manifests
as subretinal hypereflective material (Fig. 2) (Freund et al. 2010).
• Type 2 CNV can be secondary to AMD or a result of acquired
defects in the RPE–basement membrane complex, such as
Type 2 CNV commonly occurs in association with exuda- in pathologic myopia, retinochoroiditis, trauma, choroidal
tive AMD as well as in patients with acquired defects in their hamartomas, and optic disc anomalies.
RPE–Bruch’s membrane complex, such as pathologic myopia,
retinochoroiditis, trauma, choroidal tumors, optic disc anomalies,
• Depending on the cause, type 2 CNV can occur in association
with type 1 CNV.
and other primary conditions (Shah et al. 2014). These acquired
defects provide a path of least resistance to the growth of REFERENCES
abnormal blood vessels and serve as an entry point into the Freund KB, Zweifel SA, Engelbert M. Do we need a new classification for
sub-retinal space. Depending on the cause, type 2 CNV can occur choroidal neovascularization in age-related macular degeneration? Retina.
2010;30(9):1333–1349.
in association with type 1 CNV, forming a mixed neovascular Shah VP, Shah SA, Mrejen S, et al. Subretinal hyperreflective exudation
complex. In this case, irregular PED with visible interruptions associated with neovascular age-related macular degeneration. Retina.
in the RPE layer are observed on OCT. Associated subretinal 2014;34:1281–1288.

32
6.2.2

Type 2 Choroidal Neovascular Membrane


Early Middle Late

FIG. 1. Fluorescein angiography shows in the early phase of the angiogram the fine contour of CNV above the RPE and marked hyperfluorescence in
the late phase secondary to profuse leakage typical of classic CNV.

Intraretinal cysts
CNV complex

FIG. 2. OCT scan corresponding to the eye depicted in Fig. 1. There is subretinal hyperreflective material corresponding to the type 2 CNV complex
located above split RPE. Intraretinal cysts are also present.

Sub-retinal fluid

Pigment epithelial
Type 2 CNV detachment

FIG. 3. OCT scan of a mixed CNV lesion showing the type 2 component. There is a PED with subretinal fluid.

33
Type 3 Choroidal Neovascular Membrane
A. Yasin Alibhai | Daniela Ferrara 6.2.3
Summary Key Points
Type 3 CNV results from the proliferation of abnormal blood • Type 3 CNV corresponds to RAP. This CNV subtype originates
vessels from deep within the retinal tissue. These new intraretinal within the retina and eventually evolves with chorioretinal
vessels are also known as retinal angiomatous proliferation (RAP) anastomosis.
lesions. Type 3 CNV may grow within the neurosensory retina • Type 3 CNV is secondary to exudative AMD.
or down toward the choroid, forming chorioretinal anastomosis. • On OCT, type 3 CNV is commonly associated with marked
Type 3 CNV typically occurs secondary to AMD, in eyes with intraretinal fluid accumulation and subretinal fluid.
confluent soft drusen (Fig. 1). • Atrophy of the RPE often develops upon collapse of an
On OCT, the intraretinal neovascularization is typically associ- associated PED.
ated with marked intraretinal fluid and intraretinal cystic changes,
as well as occasionally with subretinal fluid (Fig. 2). Hyperreflective BIBLIOGRAPHY
Freund KB, Ho IV, Barbazetto IA, et al. Type 3 neovascularization: the expanded
material below the neurosensory retinal also may be present.
spectrum of retinal angiomatous proliferation. Retina. 2008;28:201–211.
Chorioretinal anastomosis results in irregular PED (Fig. 3). Yannuzzi LA, Negrao S, Iida T, et al. Retinal angiomatous proliferation in
Type 3 CNVs are exquisitely responsive to anti–vascular age-related macular degeneration. Retina. 2001;21:416–434.
endothelial growth factor (VEGF) agents. Very commonly, collapse
of a PED may lead to atrophy.

Hyperreflective material,
presumed early
Intraretinal cysts neovascularization

Irregular pigment epithelial detachment

N T

FIG. 1. Color fundus image of an eye with retinal angiomatous FIG. 3. Color fundus image of an eye with retinal angiomatous
proliferation (RAP), or type 3 CNV, shows an area of sub-retinal proliferation (RAP), or type 3 CNV, shows an area of sub-retinal
hemorrhage. Reticular pseudodrusen are also present. hemorrhage. Reticular pseudodrusen are also present.

FIG. 2. OCT scan of a type 3 CNV lesion. There is an area of


Subretinal fluid RAP lesion hyperreflectivity in the outer retina overlying the PED. Intraretinal cystic
changes are also present. Reticular pseudodrusen are observed in the
surrounding macular area.

Flat irregular pigment epithelial detachment

S
1
T N

34
Subretinal Hemorrhage
A. Yasin Alibhai | Daniela Ferrara 6.2.4
Summary retina (Fig. 2). Depending on the cause, it can be associated with
additional changes—for example, pigment epithelial detachment
Subretinal hemorrhage (SRH) can be caused by various choroidal secondary to type 1 CNV.
and retinal vascular abnormalities. However, the most common
cause of those involving the macula is CNV secondary to AMD.
SRH can vary in size and distribution. The clinical appearance
is that of a dark red area with normal overlying retinal vessels
Key Points
and fairly well-demarcated margins (Fig. 1). There may also be • On OCT, a subretinal hemorrhage (SRH) appears as hyper-
associated breakthrough intraretinal or vitreous hemorrhage. reflective material underneath the neurosensory retina.
Large submacular hemorrhages tend to have a poorer prognosis • The most common cause of SRH is CNV secondary to AMD;
regardless of any attempted intervention. Damage to the photo- other causes include myopia, trauma, angioid streaks, and
receptors is a consequence of the toxic nature of blood and its histoplasmosis.
components (iron, hemosiderin, and fibrin) within the subretinal • The size of SRH can vary; larger hemorrhages carry a poorer
spaces, sheer forces created by clot retraction and physical visual prognosis.
separation from the RPE layer. This can result in atrophy and
disciform scar formation regardless of treatment approach. On
BIBLIOGRAPHY
clinical examination or fundus photographs, it is challenging to Bressler NM, Bressler SB, Fine SL. In: Schachat Andrew SP, ed. Neovascular
differentiate SRH from sub-RPE hemorrhage, although the latter (Exudative) Age-Related Macular Degeneration in Retina. Vol. II. 4th ed.
tends to be darker because of the overlying RPE. Elsevier, Mosby; 2006:[Chapter 61].
On OCT, subretinal hemorrhage appears as optically dense,
hyperreflective material accumulated under the neurosensory

FIG. 1. Color fundus image


and red-free image of subretinal
hemorrhage. Note the dark color
of the blood.

FIG. 2. OCT line scan


corresponding to the eye
depicted in Fig. 1 shows
Subretinal fluid Subretinal hemorrhage subretinal hemorrhage and
subretinal fluid with an associated
pigment epithelial detachment.

S
5 Pigment epithelial detachment
T N

35
Disciform Scar
A. Yasin Alibhai | Daniela Ferrara 6.2.5
Summary complete loss of the outer retinal layers, especially the photoreceptor
layer, is commonly seen above the disciform scar. Intraretinal cystic
A disciform scar represents the end stage of a CNV lesion. spaces can be present as a result of retinal degeneration (Fig. 2).
Fibrovascular tissue develops within Bruch’s membrane, the
subretinal space, and the subretinal pigment epithelium space.
The RPE undergoes thickening in and around the area of scar- Key Points
ring, and cystic degeneration of the overlying retina results in
photoreceptor loss (Fig. 1). Although these scars are usually • Disciform scar represents the end stage of CNV.
considered stable, disease activity may still persist in an adjacent • It appears as hyperreflective subretinal material on OCT.
CNV lesion, with recurring hemorrhage and exudation being the
BIBLIOGRAPHY
most common signs of activity. Spaide RF. Clinical manifestations of choroidal neovascularization in AMD.
On OCT, a disciform scar appears as hyperreflective tissue with In: Holz FG, Pauleikhoff D, Spaide RF, Bird AC, eds. Age-Related Macular
distinct borders, underneath the neurosensory retina. Attenuation or Degeneration. 3rd ed. Berlin, Heidelberg: Springer Berlin Heidelberg; 2013.

FIG. 1. Color fundus photograph of a subretinal


disciform scar from CNV secondary to AMD.

FIG. 2. Corresponding OCT scan


of disciform scar seen in Fig. 1.
Intraretinal cyst Hyperreflective material is present
in the subretinal space that
corresponds to the organized
subretinal scar. Note loss of the
photoreceptor layer above the
scar and degenerative intraretinal
cystic changes not associated
with CNV activity. Shadowing
Organized, subretinal underlies the scar.
scar

N T

36
Retinal Pigment Epithelial Tear
A. Yasin Alibhai | Daniela Ferrara 6.2.6
Summary RPE at the margins of an area with exposed Bruch’s membrane
and absent overlying RPE (Fig. 1). The outer neurosensory retinal
Retinal pigment epithelium (RPE) tears develop as a consequence layers may have normal appearance or may be thinned.
of pigment epithelial detachments (PED) secondary to CNV
membranes, retinal angiomatous proliferation (RAP) lesions, or
polypoidal choroidal vasculopathy (PCV). They can occur without
treatment for CNV or after thermal laser, photodynamic therapy,
Key Points
or anti–vascular endothelial growth factor (anti–VEGF) therapy. • RPE tears develop as a result of PED secondary to CNV.
The mechanisms behind an RPE tear are thought to be the • On OCT, RPE tears appear as hyperreflective tissue rolled up
result of two opposing forces: traction from CNV contraction underneath the neurosensory retina and adjacent to an area
and adhesion from the RPE that is still attached. Since an RPE with complete RPE loss.
tear involving the fovea usually results in a devastating visual
outcome; therefore prognostic markers that are predictive of
RPE tear formation have been investigated. Studies have shown BIBLIOGRAPHY
Chan CK, Abraham P, Meyer CH, et al. Optical coherence tomography-
PEDs measuring more than 400 µm in height have a higher risk measured pigment epithelial detachment height as a predictor for retinal
for evolving into RPE tears. pigment epithelial tears associated with intravitreal bevacizumab injections.
On OCT, RPE tears appear as hyperreflective tissue rolled up Retina. 2010;30:203–211.
underneath the neurosensory retina, with a free edge of “wavy”

Edge of RPE
tear
RPE bunching
Subretinal
fluid

OCT
signal
blockage Reverse
shadowing
S

T N

FIG. 1. OCT line scan through an area of retinal hemorrhage shows an RPE tear. The absence of the RPE in the region of the tear allows for deeper
penetration of the OCT signal, creating the characteristic “reverse shadowing” (or “hypertransmission”) sign. The RPE is rolled up where it is still
present, resulting in OCT signal blockage and reduced visibility of deeper structures.

37
Polypoidal Choroidal Vasculopathy
A. Yasin Alibhai | Daniela Ferrara 6.2.7
Summary documenting the entire extent of the lesion. OCT angiography
may fail to document well PCV lesions. In challenging or atypical
Polypoidal choroidal vasculopathy (PCV) manifests as a variant cases, multimodal imaging might be helpful in the differential
of type 1 CNV, more commonly occurring in African-American diagnosis of PCV, especially indocyanine-green angiography
or Asian individuals. Its cause is still not fully understood, and (ICGA) (Fig. 4).
many authors consider it as a variant of AMD. However, it can
present itself as a distinct clinical entity, without the typical clinical
features of AMD or in conjunction with typical clinical findings
Key Points
(drusen, pigmentary changes) (Fig. 1). PCV lesions are character- • PCV presents a variant of type 1 CNV.
ized by polyp-shaped vascular complexes located beneath the • OCT findings typical of PCV lesions include multiple large
RPE layer. PEDs.
Typical OCT findings include multiple large PEDs with associ- • Round-oval polyps may be adherent to the posterior surface
ated subretinal fluid. These PEDs may have, adherent to their of associated PED.
posterior surface, round-oval cavities with hyperreflective borders • A branching vascular network may be seen underlying a flat
representing the polypoidal lesions (Fig. 2). Exudate due to PCV PED adjacent to the polyp, which is particularly well visualized
lesions appear as hyperreflective intraretinal spots visible on OCT on en face structural OCT.
line scans. Often, a large PED is adjacent to a smaller, flat PED
or adjacent to elevated RPE with underlying moderately hyper- BIBLIOGRAPHY
reflective tissue, representing the branching vascular network that Alasil T, Ferrara D, Adhi M, et al. En face imaging of the choroid in polypoidal
choroidal vasculopathy using swept-source optical coherence tomography.
is thought to feed the polyps (Fig. 3). Subretinal hyperreflective Am J Ophthalmol. 2015;159(4):634–643. doi:10.1016/j.ajo.2014.12.012.
material also may be observed in some cases. En face structural Yannuzzi LA, Sorenson J, Spaide RF, et al. Idiopathic polypoidal choroidal
OCT is particularly useful in supporting the diagnosis of PCV and vasculopathy (IPCV). Retina. 1990;10:1–8.

FIG. 1. Color fundus photograph of an eye with a peripapillary PCV lesion


that has subretinal fluid accumulation. Reticular pseudodrusen are also
observed.

38
6.2.7
Subretinal
Intraretinal fluid hyperreflective
material

Polypoidal Choroidal Vasculopathy


Large PED

FIG. 2. OCT line scan corresponding to Figs. 1 and 2. The OCT shows a large PED with internal hyporeflectivity adjacent to a flat PED with
heterogeneous internal hyperreflectivity where a vascular lumen is observed. Overlying intraretinal fluid is present, and subretinal hyperreflective
material is associated with the PED.

Shallow
elevation of RPE Exudates

Large PEDs
S
1
T N

FIG. 3. OCT line scan shows multiple large PEDs adjacent to a shallow PED. The branching vascular network typically lies within the shallow RPE
elevation and supplies the polyps beneath the large PEDs.

FIG. 4. ICGA corresponding to the eye depicted in Fig. 1. Early phase


image (left) shows a branching vascular network (red circle), and late
frame image shows hyperfluorescent polyps consistent with the diagnosis
of PCV.

39
SECTION 7: VITREOMACULAR INTERFACE DISORDERS

Vitreomacular Adhesion
Darin R. Goldman 7.1
Summary Key OCT Features
During the normal evolution of age-related vitreomacular separa- • Vitreomacular adhesion may be focal or broad.
tion, areas of vitreous may remain attached to the macula (see • Vitreomacular adhesion is a benign physiologic finding (Fig.
Chapter 27.1). Vitreomacular adhesion represents a normal 4) that should be distinguished from similar, often pathologic,
physiologic state of the vitreomacular interface (Fig. 1). The states of the vitreomacular interface.
degree of adhesion can vary from focal to broad (Figs. 2 and • In a patient with a history of full-thickness macular hole in
3). This benign finding is distinguished from the pathologic state one eye, if vitreomacular adhesion is present in the fellow eye,
of vitreomacular traction (see Chapter 7.2) as a result of the lack there is a risk for future full-thickness macular hole (FTMH)
of distortion or disruption of the retinal layers and no impact on formation in that eye.
vision. The presence of vitreomacular adhesion can be helpful
to determine future risk for vitreomacular interface disorders.

S
1
N T

A I

S
S 1
1 N T
T N
I

B I

FIG. 2. Broad vitreomacular adhesion is present over the entire macula.


FIG. 1A and B. Focal vitreomacular adhesion is present overlying the The posterior face of the hyaloid blends, almost imperceptibly, with the
central macula. The posterior hyaloid is seen inserting near the edges of surface of the macula (arrows). This eye later developed vitreomacular
the fovea (arrows) without any disturbance of the normal foveal contour. traction with an FTMH.

40
FIG. 3. Broad vitreomacular adhesion beginning to release. Nasally, the 7.1
posterior hyaloid is still adherent to the macular surface (white arrows).
Temporally, the posterior hyaloid has begun to release from the macula
at a focal point (yellow arrowhead), beyond where there is release of the
adhesion (yellow arrows).

Vitreomacular Adhesion
S
1
N T

Vitreomacular adhesion releasing Focal vitreomacular adhesion

S S
3 3

T N T N

I
A
I
B

Complete release of posterior hyaloid


from macular surface

S
1 FIG. 4A–C. Normal physiologic progression of vitreomacular adhesion
N T begins with release (arrows) from the paramacular edges (A), followed by
focal adhesion over the fovea (B, arrow), and finally with complete release
C
I
(C, arrows). After complete release of vitreomacular adhesion, there is no
longer a risk for FTMH development.

41
Vitreomacular Traction
Darin R. Goldman 7.2
Summary Key OCT Features
Vitreomacular traction (VMT) syndrome falls into the broad • VMT is due to an abnormally strong focal adhesion of the
category of vitreomacular interface disorders. VMT occurs when posterior hyaloid to the macula.
the posterior hyaloid fails to separate normally from the central • VMT is distinguished on OCT by disruption of the normal
macular surface during abnormal posterior vitreous detachment macular contour at a focal point with overlying vitreous
(Figs. 1–7). The degree of traction over the fovea varies from a insertion.
pinpoint spot to a broad area. Secondary effects on the macula • VMT may resolve spontaneously or progress to visually impact-
include distortion of retinal layers, cystoid macular edema, and ful sequela, including lamellar macular hole and FTMH.
subretinal fluid (Fig. 6). Visual impairment can vary from mild to
severe. VMT often will release spontaneously; therefore an initial
period of observation is usually warranted. However, VMT also
may worsen or progress to a lamellar hole or FTMH (Figs. 4 and
5). Pharmacologic or surgical interventions should be considered
if symptoms become significantly bothersome to the patient.

S S
3 3

T N T N

I I
A B

FIG. 1A and B. Mild VMT (A) spontaneously releases over time, leaving a normal macular contour (B).

42
7.2

Vitreomacular Traction
Subretinal fluid
S S
3 3
T N T N

I I
A B

Released VMT

S
5
T N

I FIG. 2A–C. VMT with associated subfoveal fluid (A) improves


C spontaneously over time (B), with eventual complete release (C).

S S
2 4

T N T N

I I
A B

FIG. 3A–B. VMT associated with changes in the fovea similar to those in tractional schisis (A) that worsen over time (B). Intervention may be
considered, depending on the visual impact.

Large macular hole

Mild VMT

S S
4 2

T N T N

I I
A B
43
FIG. 4A and B. Mild VMT (A) may progress to a large FTMH over time (B). This evolution was not possible to predict.
Section 7: Vitreomacular Interface Disorders

S S
1 1
T N N T

I I
A B

Progressive VMT
Posterior hyaloid released
Normal macular contour

S S
1 1
T N N T

I I
C D

Lamellar macular hole

Posterior hyaloid released

S
1
N T FIG. 5A–E. VMT present in both eyes (A and B). With release of the
posterior hyaloid from the macular surface, spontaneous normalization
I
E of the macular contour (C) or development of a lamellar macular hole
(D and E) can occur.

44
7.2

Vitreomacular Traction
S S
4 1 Cystoid macular edema
T N N T

I I
A D

S S
3 3
T N T N

I I Subretinal fluid
B E

Distortion of retinal layers

S S
4 3
N T T N

I I

C F

FIG. 6A–F. Mild (A), moderate (B), and severe (C) VMT with focal adhesion over the fovea. VMT may be associated with cystoid macular edema (D),
subretinal fluid (E), and distortion of the retinal layers (F).

45
Worsening vitreomacular traction

Vitreomacular traction
Section 7: Vitreomacular Interface Disorders

S S Worsening secondary pseudo-vitelliform lesion


3 Secondary pseudo-vitelliform lesion 3
N T N T

I I
A B

Worsening vitreomacular traction

Secondary cystoid
macular edema Restoration of macular contour following pars plana vitrectomy

S S
1 7
N T N T

I I
C D

FIG. 7A–D. VMT may significantly worsen over time (A–C). This patient underwent pars plana vitrectomy with elevation of the hyaloid; postoperatively
the patient experienced a dramatic normalization of the macular contour and a significant improvement in visual acuity (D).

46
Full-Thickness Macular Hole
Darin R. Goldman 7.3
Summary Key Points
Normally, with aging there is progressive liquefaction of the • Small FTMHs have a diameter of 250 µm or less (Figs. 1–3).
vitreous body and eventual complete separation of the posterior • Medium FTMHs have a diameter between 250 and 400 µm
hyaloid from the macula during posterior vitreous detachment. Fig. 4.
FTMHs typically develop in a predisposed individual who has an • Large FTMHs have a diameter greater than 400 µm Figs. 5–6.
abnormally taut adhesion between the vitreous and the central • The presence or absence of VMT is important in classifying
macula (Figs. 1–7). In these persons, during posterior vitreous FTMHs.
detachment evolution, focal traction on the fovea results in • The size of a FTMH is determined based on the minimum hole
the development of a full-thickness defect or hole. FTMHs are width at the narrowest point in the mid-retina (Figs. 1, 4, and 5).
described based on diameter and the presence or absence of
REFERENCE
VMT according to the International Vitreomacular Traction Study Duker JS, Kaiser PK, Binder S, et al. The International Vitreomacular Traction
Classification System (Duker et al. 2013). Study Group classification of vitreomacular adhesion, traction, and macular
hole. Ophthalmology. 2013;120(12):2611–2619.

Vitreomacular traction
Perifoveal intraretinal cysts

Large
S ~200 µm S drusen
3 3
N T T N

I I

FIG. 1. A small FTMH (~200 µm diameter) with VMT. FIG. 2. A small FTMH without VMT. There are minimal perifoveal
intraretinal cysts, the paucity of which suggests this hole is of more
chronic duration. Incidentally, large drusen are present.

Posterior hyaloid Foveal


face operculum

S
7
N T

FIG. 3. A small FTMH without VMT. The posterior hyaloid face and a
foveal operculum are visible toward the top of the image.

47
Prominent intraretinal cysts Vitreomacular traction
Section 7: Vitreomacular Interface Disorders

S
3
N T 307 µm diameter – measured at the
narrowest point in the mid-retina >450 µm diameter
I
A

FIG. 5. A large FTMH with VMT.

Retinal tissue has recanalized


the full-thickness hole

S
3
N T
Hyporeflective outer retinal
cleft in the IS/OS/EZ layer
I
B

No vitreomacular traction

S
3 S
N T 3
T N
I
C I

FIG. 4A–C. (A) Medium FTMH without VMT. This image provides a good FIG. 6. A large FTMH without VMT.
example of the proper location to measure the aperture of a macular
hole—the minimum hole width at the narrowest point in the mid-retina.
Prominent intraretinal cysts on the hole edge suggest this hole is of
relatively acute onset. (B) Typical appearance 2 weeks after pars plana
vitrectomy with internal limiting membrane peeling and gas tamponade
(corresponding to A). The foveal depression has returned; however,
a hyporeflective outer retinal cavity is present. (C) Typical appearance
6 months postoperatively in which the foveal contour is well defined,
although still somewhat irregular, and the outer retinal layers have fully
returned and are continuous. EZ, Ellipsoid zone.

48
Macula Thickness: Macular Cube 512 ¥ 128 OD OS
7.3
500
293

400
348

Full-Thickness Macular Hole


300 275 324 406 386 320

329
200
287
100
ILM-RPE Thickness (m) Fovea: Not found

0 m
Overlay: ILM-RPE Transparency: 50%

ILM-RPE

ILM
Diversified:
Distribution
of Normals

99%
95%
5%
1%
RPE

Central Cube
Cube
Subfield Average
Volume
Thickness Thickness
(mm3)
(m) (m)

ILM-RPE 406 10.7 298

FIG. 7. A volumetric map illustrates an eccentric FTMH superotemporal to the fovea. This was noted in a patient after uneventful pars plana
vitrectomy and membrane peeling for an epiretinal membrane, presumably iatrogenic. No additional treatment was performed, the patient was
asymptomatic, and the hole remained stable over an extended period.

FIG. 8. This vitreomacular abnormality fits somewhere between VMT and


an early FTMH. A layer of inner retina is clearly still intact, but with time
this would be expected to progress to a full-thickness defect. This case
helps illustrate the dynamic nature of VMT and macular holes. ILM, Inner
limiting membrane; RPE, retinal pigment epithelium.
Vitreomacular traction
Thin residual intact retina

49
Lamellar Macular Hole
Darin R. Goldman 7.4
Summary Key OCT Features
A lamellar macular hole or defect results from a variety of causes, • Foveal OCT features of lamellar macular hole include the
such as abortive full-thickness macular hole (FTMH) or epiretinal following (Witkin 2006):
membrane (ERM) formation that causes partial thickness loss • Irregular foveal contour
of the inner macular layers involving the fovea. Historically, this • Defect of inner fovea
was an ill-defined clinical diagnosis; it has become more clearly • Separation of the inner and outer retinal layers
defined with OCT. The characteristic appearance on OCT is an • Lack of a full-thickness retinal defect
irregular, anvil-shaped inner foveal defect without loss of the • Lamellar macular holes typically remain stable over time, and
outer retinal layers (Figs. 1–4). Schisis-like changes also may be surgical treatment is rarely required.
seen between the inner and outer retinal layers. Associated ERMs
REFERENCE
are common. Lamellar macular hole can be distinguished from Witkin AJ, Ko TH, Fujimoto JG, et al. Redefining lamellar holes and the
“macular pseudohole” in that there is no loss of foveal tissue in vitreomacular interface: an ultrahigh-resolution optical coherence tomography
the latter (see Chapter 7.5). Visual acuity generally remains good. study. Ophthalmology. 2006;113(3):388–397.

50
FIG. 1. Common features of lamellar macular hole are depicted, including 7.4
Irregular foveal contour an irregular foveal contour, a defect of the inner fovea (between bars), a
separation between the inner and outer retina, and an intact outer retina
(lack of full-thickness hole). There is also an associated ERM present,
Epiretinal membrane which is common.
Defect of inner fovea

Lamellar Macular Hole


Intact outer Separation between inner
S
retina and outer retina
3
T N

Irregular foveal contour Irregular foveal contour

Defect of inner fovea

Separation between
S inner and outer retina S Separation between Intact outer retina
2 4 inner and outer retina
T N Intact outer retina N T

I I
A B

FIG. 2A and B. Additional typical examples of lamellar macular hole, depicting features similar to those in Fig. 1.

Anvil-shaped defect between Gap between inner and outer retina


S S
6 inner and outer retina 5 may be asymmetric
T N T N

I I

FIG. 3. In a lamellar macular hole, the defect between the inner and FIG. 4. Depending on the exact cross section imaged, the defect
outer retina often conforms to an anvil shape. This area also may have between the inner and outer retina in a lamellar macular hole may be
schisis-like clefts. asymmetric.

51
Epiretinal Membrane
Darin R. Goldman 7.5
Summary cases are mild and do not require treatment. More severe cases
with visual impairment may require surgical removal.
ERM formation is much more common than either VMT or lamellar
macular hole formation, with a prevalence greater than 30%
(Meuer et al. 2015). ERM may occur secondary to a variety of
ocular conditions, such as uveitis, retinal tear, and retinal detach-
Key OCT Features
ment. However, frequently they are idiopathic with no identifiable • OCT is very sensitive in detecting the presence of an ERM,
cause. In these cases, in which a posterior vitreous detachment which appears as a hyperreflective, thickened membrane on
is usually present, it is thought that residual posterior hyaloid the inner surface of the macula.
forms a scaffold for the proliferation of cellular material on the • An ERM may take on a corrugated or undulating contour in
surface of the macula. An ERM appears as a thin, hyperreflective cross section.
sheet overlying the internal limiting membrane Figs. 1–6. A mild • In the presence of ERM, the macular contour may appear
ERM may cause little or no distortion of the underlying retinal normal or become highly disorganized on OCT.
layers and no impact on vision. With more severe proliferation,
the macular contour can be quite disturbed, with loss of the
REFERENCE
foveal depression and resultant impairment in visual acuity Fig. Meuer SM, Myers CE, Klein BE, et al. The epidemiology of vitreoretinal interface
7. Associated visual symptoms may include metamorphopsia, abnormalities as detected by spectral-domain optical coherence tomography:
decreased visual acuity, micropsia, and monocular diplopia. Most the Beaver Dam Eye Study. Ophthalmology. 2015;122(4):787–795.

S Typical epiretinal membrane


3
T N

FIG. 1. Color photograph showing typical macular appearance of an FIG. 2. Typical OCT appearance of ERM showing a hyperreflective,
ERM with irregular sheen, loss of the foveal reflex, and distortion of the thin membrane overlying the surface of the macula. The normal smooth
normal vascular pattern. contour of the macula is replaced by corrugations of the macular surface
(white arrows). There is loss of the foveal depression (yellow arrow).

52
7.5
Indistinct epiretinal membrane

Epiretinal Membrane
Schisis-like splitting between
inner and outer retina
S
7
T N

FIG. 3. Typical ERM appearance (white arrows). Associated schisis-like FIG. 4. The ERM is somewhat indistinct (white arrows) and is associated
changes are present between the inner and outer retina (asterisk), with significant schisis between the inner and outer retinal layers, an
similar to the appearance of a lamellar macular hole. The corresponding apparent tractional effect. This could be considered a lamellar macular
thickness map (inset) shows thickening of the central macula in an hole by some classification schemes.
irregular shape.

Prominent epiretinal membrane Postoperative appearance after vitrectomy


associated with a pseudohole with membrane peeling

S S
3 3
N T N T

I I

FIG. 5. A prominent ERM (arrows) is associated with a pseudohole FIG. 6. Postoperative appearance after vitrectomy, with peeling of the
(asterisk). No loss of retinal tissue occurred, which distinguishes this ERM corresponding to preoperative Fig. 5.
entity from a lamellar macular hole.

53
500 500
400 400
300 300
Section 7: Vitreomacular Interface Disorders

200 200
100 100
0 m 0 m

+150

+75

0 m

–75

–150

FIG. 7. In addition to B-scans, thickness and difference maps can be very illustrative in depicting changes over time, although correct segmentation
is imperative. A typical ERM shown preoperatively (left) and postoperatively after vitrectomy with membrane peeling (right). The thickness maps (top)
show normalization of macular thickness within the central macula. The difference map (middle, right) shows the corresponding degree of reduced
thickness. Corresponding B-scans are also shown (bottom).

54
SECTION 8: CENTRAL SEROUS CHORIORETINOPATHY

Central Serous Chorioretinopathy


Eduardo A. Novais | Luiz Roisman 8.1
Summary Kishi 2000; Piccolino et al. 2005; Yang, Jonas, & Wei, 2013). OCT
can clearly define the presence of and gauge changes in detach-
Central serous chorioretinopathy (CSR) is a disease characterized ment of the RPE and subretinal fluid (Fig. 2). OCT angiography
by the serous detachment of the neurosensory retina that occurs provides the additional ability to visualize flow impairment of the
over an area of leakage from the choriocapillaris through the retinal choriocapillaris and detect secondary CNV (Feucht et al. 2016;
pigment epithelium (RPE) (Hussain & Gass 1998; Wang et al. 2008; Shinojima et al. 2016).
Yap & Robertson 1996). Its frequency is approximately 10 cases
per 100,000 and is sixfold higher in men than in women (Hussain
& Gass 1998; Wang et al. 2008; Yap & Robertson 1996). The OCT Key Features
age at diagnosis usually ranges from the third to the fifth decade
(Hussain & Gass 1998; Wang et al. 2008; Yap & Robertson 1996). • Serous macular detachment along with
The pathophysiology is unknown; however, the most accepted • Retinal pigment epithelium detachment
theory asserts that initial choroidal vascular hyperpermeability • Choroidal thickening
leads to secondary dysfunction of the overlying RPE (Hussain & • Outer retinal granulations (Figs 2-4)
Gass 1998; Wang et al. 2008; Yap & Robertson 1996). • Secondary choroidal neovascularization may be identified on
OCT angiography (Fig. 5)
Type A personalities, exogenous steroid use, and systemic
hypertension are the most common associations with the develop-
ment of CSR (Hussain & Gass 1998; Wang et al. 2008; Yap & REFERENCES
Feucht N, Maier M, Lohmann CP, et al. OCT angiography findings in acute
Robertson 1996). Serous retinal detachments typically resolve central serous chorioretinopathy. Ophthalmic Surg Lasers Imaging Retina.
spontaneously in most patients, and approximately 80% to 90% of 2016;47:322–327.
them return to 20/25 or better vision (Hussain & Gass 1998; Wang Hussain D, Gass JD. Idiopathic central serous chorioretinopathy. Indian J
et al. 2008; Yap & Robertson 1996). A subset of patients may Ophthalmol. 1998;46:131–137.
Iida T, Hagimura N, Sato T, et al. Evaluation of central serous chorioretinopathy
have recurrent or chronic serous retinal detachments, resulting in with optical coherence tomography. Am J Ophthalmol. 2000;129:16–20.
progressive RPE atrophy and permanent visual loss of 20/200 or Piccolino FC, de la Longrais RR, Ravera G, et al. The foveal photoreceptor
worse. In recurrent or chronic cases, choroidal neovascularization layer and visual acuity loss in central serous chorioretinopathy. Am J
(CNV) also may develop (Hussain & Gass 1998; Wang et al. 2008; Ophthalmol. 2005;139:87–99.
Shinojima A, Kawamura A, Mori R, et al. Findings of optical coherence
Yap & Robertson 1996).
tomographic angiography at the choriocapillaris level in central serous
Fluorescein angiography is currently considered the gold chorioretinopathy. Ophthalmologica. 2016;236:108–113.
standard for diagnosis, and findings vary from discrete areas Wang M, Munch IC, Hasler PW, et al. Central serous chorioretinopathy. Acta
of focal leakage to diffuse RPE defects (Hussain & Gass 1998; Ophthalmol. 2008;86:126–145.
Wang et al. 2008; Yap & Robertson 1996) (Fig. 1). OCT has Yang L, Jonas JB, Wei W. Optical coherence tomography-assisted enhanced
depth imaging of central serous chorioretinopathy. Invest Ophthalmol Vis
played an increasingly important role in the diagnosis, prognosis, Sci. 2013;54:4659–4665.
and follow-up of patients with CSR, especially considering its Yap EY, Robertson DM. The long-term outcome of central serous chorio-
non-invasive nature in comparison to FA (Iida, Hagimura, Sato, & retinopathy. Arch Ophthalmol. 1996;114:689–692.

55
Section 8: Central Serous Chorioretinopathy

A B C

D E F

G H I

FIG. 1A–I. Multimodal imaging of a patient with acute CSCR. (A) Fundus photography shows a fovea neurosensory retinal detachment. Fluorescein
angiography (B, arrow) and indocyanine green angiography indicate an “inkblot” leakage (C, white arrow). (D) Baseline OCT B-scan shows a serous
detachment of the neurosensory retina and a serous pigment epithelium detachment (PED) (yellow arrow). Baseline structural en face OCT segmented
at the inner plexiform membrane (IPM) (E) and RPE (F) show a circular hyporeflective area corresponding to signal loss secondary to subretinal fluid
blockage. (G) Follow-up OCT B-scan shows regressions of subretinal fluid with persistence of the serous PED (yellow arrow). (H) Follow-up en face
OCT segmented at the level of IPM. (I) RPE shows regression of the hyporeflective area.

56
8.1

Central Serous Chorioretinopathy


FIG. 2. OCT B-scan of a patient with central serous chorioretinopathy, associated to pigmented epithelium detachment (yellow arrow).

A B C

FIG. 3A–D. Multimodal imaging of a patient with inactive CSCR with thick choroid and large choroidal vessels. (A) Fundus autofluorescence
was able to identify the classic descending tract sign associated with areas of hyperautofluorescence and hypoautofluorescence. (B) Fluorescein
angiography shows areas of hyperfluorescence resulting from retinal pigment epithelium defect. (C) Indocyanine green angiography shows areas of
hyperpermeability. (D) On the OCT B-scan it is possible to identify a thickened choroid (yellow dotted line and yellow arrow) and dilated choroidal
vessels (white asterisks).

57
Section 8: Central Serous Chorioretinopathy

FIG. 4. OCT B-scan of a patient acute CSCR, associated with granulated changes of the outer photoreceptors layers (yellow arrow and arrowhead).

A B

C D

FIG. 5. Multimodal imaging CSCR with secondary CNV. (A) Fundus photograph shows mottled RPE without the presence of intraretinal or subretinal
hemorrhage. (B) OCT B-scan centered on the fovea shows a flat RPE detachment (white arrow) associated with little subretinal fluid and thickened
choroid and large dilated vessel (yellow arrow). (C) Mid-phase fluorescein angiography shows no noticeable leakage. (D) OCT angiogram segmented
at the choriocapillaris clearly indicates the neovascular membrane complex (yellow dashed line).

58
SECTION 9: MYOPIC DEGENERATIVE MACULOPATHIES

Myopic Choroidal Neovascular Membrane


Darin R. Goldman 9.1
Summary appearance of myopic CNV is often more subtle than that seen
in other causes of CNV and sometimes can be overlooked if this
Myopic choroidal neovascular membrane (CNV) is the most is the only modality relied on for diagnosis (Figs. 1–2).
common non–age-related macular degeneration (AMD) cause
for CNV. High myopia results in degenerative changes to Bruch’s
membrane and the retinal pigment epithelium (RPE) underly- Key OCT Features
ing the macula, which results in CNV formation in more than
10% of highly myopic eyes (Grossniklaus & Green 1992). The • Myopic CNV appears as a highly reflective, well-circumscribed,
associated exudation, hemorrhage, and/or scarring contribute round complex on OCT.
to associated visual impairment. Although the natural history • Typically, minimal associated exudation such as cystoid macular
of myopic CNV tends to be more favorable than that of CNV edema or subretinal fluid is present.
secondary to AMD, without treatment a significant portion of • Additional diagnostic modalities such as fluorescein angiog-
eyes will lose vision. Therefore myopic CNV is a major cause raphy are often required to confirm the diagnosis of myopic
for serious vision loss and blindness worldwide. With the rising CNV (Figs. 3–4).
prevalence of high myopia, this is likely to become an increasingly
encountered macular condition. Anti–vascular endothelial growth REFERENCE
factor (anti–vascular endothelial growth factor [VEGF]) therapy Grossniklaus HE, Green WR. Pathologic findings in pathologic myopia. Retina.
has emerged as the most effective treatment for myopic CNV, 1992;12(2):127–133.
although its use remains off-label in most countries. The OCT

59
Intraretinal fluid
Ill-defined
subretinal material
Section 9: Myopic Degenerative Maculopathies

Myopic CNV
Subretinal fluid

Myopic CNV
S S
2 2
T N T N

A I
B I

FIG. 1A–C. (A) Myopic CNV appears as a somewhat ill-defined


hyperreflective, dome-shaped elevation in the subretinal space between
the neurosensory retina and the RPE. Associated subretinal fluid and less
well-defined subretinal material with mixed reflectivity is present.
(B) At 1 month after initial anti-VEGF therapy, the CNV complex becomes
Myopic better defined and the associated exudation is mostly resolved with some
1
S
CNV residual intraretinal fluid. (C) At 2 months after monthly anti-VEGF therapy,
T N RPE all associated fluid has resolved. The CNV has regressed into an area
Atrophy of subretinal fibrosis, and a small area of RPE atrophy has appeared.
(Modified with permission Goldman, D. In press. Choroidal Neovascularization, Not
C I
AMD. In Duker J., Liang M [Eds.], Anti-VEGF Use in Ophthalmology.)

S S
1 1
T N
Resolution of
T N
Typical myopic CNV after single
CNV anti-VEGF
A I
B I

FIG. 2A and B. (A) Typical myopic CNV identified as a dome-shaped hyperreflective elevation between the neurosensory retina and RPE. There is no
associated exudation. (B) After treatment with a single anti-VEGF injection, the CNV complex completely resolved.

60
9.1

Myopic Choroidal Neovascular Membrane


Myopic CNV

Myopic CNV

B
A

Myopic CNV

FIG. 3A–C. Corresponds to Fig. 2. (A) Clinical appearance of myopic CNV


C is subtle, with hemorrhage best appreciated on red-free photograph (B).
(C) Fluorescein angiography reveals a type 2 leakage pattern.

Myopic CNV
Myopic CNV

A B

FIG. 4A and B. (A) Typical myopic CNV clinical appearance and corresponding fluorescein angiogram demonstrating type 2 leakage pattern (B).

61
Myopic Macular Schisis
Darin R. Goldman 9.2
Summary Key OCT Features
Myopia is one of the most common causes for macular schisis. • Schisis may occur in multiple layers of the macula; however,
Other causes include juvenile X-linked retinoschisis, optic pits, the outer layers are most commonly affected, leaving a thicker
and idiopathic. Macular schisis in the setting of high myopia is inner retina and thinner outer retina.
thought to be due to a mechanical effect as a result of abnormal • Perpendicular strands stretching within the schisis cavity are
axial elongation and progressive sclera thinning. Subsequent thought to be Müller cells.
stretching or splitting of the macular layers occurs due to the • Other associated features of myopia may be noted on OCT,
induced abnormal vitreoretinal tractional forces. A more descriptive such as staphyloma, RPE atrophy, and vitreomacular interface
term “myopic ectatic retinopathy,” representative of this presumed abnormalities (Figs. 1–5).
mechanism, has been proposed (Tsilimbaris, Vavvas, & Bechrakis
2016). The schisis plane commonly occurs between the outer REFERENCE
Tsilimbaris MK, Vavvas DG, Bechrakis NE. Myopic foveoschisis: an ectatic
plexiform layer (OPL) and Henle fiber layer (HFL). However, the
retinopathy, not a schisis. Eye (Lond). 2016;30(2):328–329.
plane may occur in the inner, middle, or outer macular layers. No
OCT-based epidemiologic studies have investigated the incidence BIBLIOGRAPHY
and prevalence of myopic macular schisis. Myopic macular schisis Gohil R, Sivaprasad S, Han LT, et al. Myopic foveoschisis: a clinical review.
appears to be more common with increasing degree of myopia/ Eye (Lond). 2015;29(5):593–601.
axial length and in the setting of posterior staphyloma. Visual Ober MD, Freund KB, Shah M, et al. Stellate nonhereditary idiopathic foveo-
macular retinoschisis. Ophthalmology. 2014;121(7):1406–1413.
acuity may be variably affected and, over time, myopic macular
schisis is slowly progressive. Treatment is considered when visual
symptoms or acuity progressively worsen. Interventions include
vitrectomy with or without internal limiting membrane peeling
and/or gas tamponade.

Portion of
posterior hyaloid

Myopic macular Myopic


schisis macular
S schisis
1
N T

A B I
Geographic atrophy

FIG. 1A and B. (A) Fundus photograph of typical myopic macular schisis with significant atrophic myopic degeneration. (B) Corresponding OCT
shows typical myopic macular schisis with a plane of separation located within the outer retinal layers.

62
9.2
INL
schisis

Lamellar macular
hole

Myopic Macular Schisis


INL OPL

OPL/HFL
schisis

S S
1 1
N T OPL/HFL N T
OPL/HFL
Myopic macular schisis schisis
schisis
I I

FIG. 2. Myopic macular schisis located within the OPL/HFL. A small FIG. 3. A typical example of myopic macular schisis with the schisis
affected area of schisis is seen within the inner nuclear layer (INL). plane located between the OPL and HFL. The INL is identified to help
with orientation. A lamellar macular hole is present, which is commonly
associated with myopic macular schisis.
Myopic macular schisis Posterior
Presumed müller cells hyaloid

Myopic
macular
schisis
A 200 m

Vitreomacular traction Posterior


hyaloid

Myopic Myopic
macular macular
B schisis schisis
200 m 200 m

FIG. 4A and B. The right eye (A) and left eye (B) show features similar FIG. 5. An additional example of myopic macular schisis affecting the
to those of myopic macular schisis in multiple layers. The posterior outer retinal layers.
hyaloid is still adherent to the macula in the right eye but is lifting off in
the left eye with associated vitreomacular traction. A tractional element
may be contributing to the schisis in this case, along with the pathologic
anatomic changes associated with myopia.

FIG. 6. Stellate nonhereditary idiopathic foveomacular retinoschisis is


an entity with an OCT appearance similar to that of myopic macular
schisis. However, the affected patient has neither myopia nor any known
hereditary predisposing risk factors.

Stellate
nonhereditary
S
1
idiopathic
N T
foveomacular
retinoschisis
I

63
Dome-Shaped Macula
Darin R. Goldman 9.3
Summary Key OCT Features
A dome-shaped macula describes an inward or convex indentation • There is inward bowing of the sclera within the central macula;
beneath the macula that is distinct from posterior staphyloma and the overlying macula follows the same contour, giving it an
evident only on optical coherence tomography (OCT) imaging. They arched structure.
occur in approximately 20% of highly myopic eyes. Although the • A vertically oriented OCT scanning plane is most likely to
pathogenesis is not clearly understood, the best theory surmises detect the presence of a dome-shaped macula, although it
that variation in scleral thickness under the macula results in a may also be evident on a horizontally oriented OCT scan
localized region where a dome-shaped, relative, inward bulge (Fig. 2).
occurs (Imamura et al. 2011). There is often an associated • When a hyporeflective subretinal cavity/cap is present, this
“cap” of subretinal fluid (hyporeflective space) in the absence usually does not signify the presence of CNV.
of concomitant CNV. This fluid may be associated with larger
domes and decreased visual acuity, is unresponsive to treatment, REFERENCES
Imamura Y, Iida T, Maruko I, et al. Enhanced depth imaging optical coher-
is typically nonprogressive, and is of unknown pathogenesis. The
ence tomography of the sclera in dome-shaped macula. Am J Ophthalmol.
macular convexity associated with dome-shaped macula is most 2011;151(2):297–302.
commonly oriented horizontally (Liang et al. 2015) and therefore Liang IC, Shimada N, Tanaka Y, et al. Comparison of clinical features in highly
is best visualized on a vertically oriented OCT B-scan (Fig. 1). myopic eyes with and without a dome-shaped macula. Ophthalmology.
Dome-shaped macula are more likely to occur in younger patients 2015;122(8):1591–1600.
with longer axial lengths.
BIBLIOGRAPHY
Caillaux V, Gaucher D, Gualino V, et al. Morphologic characterization of dome
shaped macula in myopic eyes with serous macular detachment. Am J
Ophthalmol. 2013;156(5):958–967.
Gaucher D, Erginay A, Lecleire-Collet A, et al. Dome-shaped macula in eyes
with myopic posterior staphyloma. Am J Ophthalmol. 2008;145(5):909–914.

64
FIG. 1. Vertically oriented OCT 9.3
Dome-shaped macula scan reveals an inward bulge of
the scleral in the central macula,
which is typical of dome-shaped
macula.

Dome-Shaped Macula
Inward scleral bulge

Dome-shaped macula
Subretinal fluid

S Inward scleral bulge


1
T N

A B I

Subretinal fluid Dome-shaped macula

Inward scleral bulge


C

FIG. 2A–C. (A) Color photograph in a high myope with a posterior staphyloma. (B) Horizontally oriented OCT scan reveals a dome-shaped macula
with corresponding inward bulging of the underlying sclera. Subretinal fluid is present, which did not change over a prolonged period with and without
treatment. (C) Vertically oriented OCT scan reveals the same features as in part B; however, the dome-shaped elevation is less obvious, which is
atypical.

65
Posterior Staphyloma
Darin R. Goldman 9.4
Summary Key OCT Features
Axial elongation occurs as the globe enlarges in pathologic myopia. • The normal horizontal appearance of retinal layers on OCT is
Along with this elongation, localized outward protrusions in the lost in posterior staphyloma, giving way to a posterior bowing.
globe wall termed posterior staphyloma can occur. These areas • Posterior bowing of the retina, choroid, and sclera occur at
have a steeper curvature than the surrounding globe wall. Many the edges of a staphyloma.
types of posterior staphyloma have been described (Hsiang et al. • OCT is most useful to identify both peripapillary and macular
2008), the most common being ovoid-shaped and involving the staphyloma.
central macula and optic nerve (Frisina et al, 2016). The presence • Identification of macular staphyloma should raise suspicion for
of posterior staphyloma is strongly correlated with the degree of other concomitant macular pathologic processes associated
myopia and is present in as many as 50% of eyes with pathologic with high myopia.
myopia (Ohno-Matsui et al. n.d.). Detection of posterior staphyloma
on a purely clinical basis can be difficult in milder cases. The REFERENCES
Hsiang HW1, Ohno-Matsui K, Shimada N, Hayashi K, Moriyama M, Yoshida T,
depth-resolved nature of OCT image acquisition makes this
Tokoro T, Mochizuki M, et al. Clinical characteristics of posterior staphyloma
imaging modality particularly useful for detecting staphyloma, in eyes with pathologic myopia. Am J Ophthalmol. 2008;146(1):102–110.
especially those in a peripapillary or macular location Figs. 1–2. Ohno-Matsui K, Alkabes M, Salinas C, Mateo C, Moriyama M, Cao K, Yoshida
Additional macular pathologic conditions are commonly found T, et al. Features of posterior staphylomas analyzed in wide-field fundus
in concert with staphyloma, such as schisis, CNV, epiretinal images in patients with unilateral and bilateral pathological myopia. Retina.
2016;[Epub ahead of print].
membrane, vitreomacular traction (VMT), and tractional detach-
ment. Identification of staphyloma should raise suspicion for BIBLIOGRAPHY
identifying these additional entities. Frisina R, Baldi A, Semeraro F, Cesana BM, Parolini B, et al. Morphological
and clinical characteristics of myopic posterior staphyloma in Caucasians.
Graefes Arch Clin Exp Ophthalmol. 2016;[Epub ahead of print].

66
9.4

Macular posterior staphyloma

Posterior Staphyloma
A

Posterior staphyloma

Macular posterior staphyloma

S
3 S
1
T N
T N Dome-shaped macula
B I
I

FIG. 1A and B. (A) Wide-angle OCT view of macular posterior FIG. 2. Posterior staphyloma involving the posterior pole encompassing
staphyloma. There is a point of deflection adjacent to the optic nerve the optic nerve (not pictured) and macula. There is a high radius
where the sclera bows outward along with the overlying retina and of curvature at the edges of the macula (arrowheads). Note that a
choroid (between arrows). The staphyloma is present temporal to this dome-shaped macula is also present (see Chapter 9.3), which accounts
point and extends off the area imaged. (B) Higher magnification OCT for the relatively flatter foveal contour. (With permission from Duker, J.W.,
view of same macular staphyloma. Waheed, N.K., Goldman, D.R., 2014. Handbook of Retinal OCT: Optical Coherence
Tomography, 9.1, 46-47. Saunders, Philadelphia.)

Myopic tilt

Myopic tilt

S S
3
No outward bowing or point of deflection 3
N T T N

No outward bowing or
A I
B I
point of deflection

FIG. 3A and B. Myopic tilt should be distinguished from posterior staphyloma. Myopic tilt is common with lower degrees of myopia. There is a
generalized diagonal sloping of the macula. However, distinct from posterior staphyloma, there is no outward bowing or point of deflection.

67
SECTION 10: HYDROXYCHLOROQUINE TOXICITY

Hydroxychloroquine Toxicity
Darin R. Goldman 10.1
Summary only in advanced stages of toxicity. With continued progression,
diffuse, severe photoreceptor loss develops, with secondary
Hydroxychloroquine sulfate (Plaquenil) can result in irreversible damage and disorganization of the retinal pigment epithelium
macular toxicity. This toxicity is more prevalent than historical data (RPE) Figs. 3–4. The inner retina remains relatively normal even
suggest, with rates as high as 7.5% in long-term users (Melles with advanced maculopathy.
& Marmor 2014). The risk for developing toxicity correlates with
daily dose and overall treatment duration and is more likely with
renal impairment and concomitant tamoxifen usage. The risk for Key OCT Features
hydroxychloroquine macular toxicity is less than 1% after 5 years
on suggested dosing. Daily dosage of hydroxychloroquine should • The earliest signs of hydroxychloroquine maculopathy include
remain less than 5 mg/kg of real weight (Marmor et al. 2016). parafoveal outer retinal thinning, loss of the cone outer segment
When macular toxicity from hydroxychloroquine becomes tip line, and disruption of the IS/OS/EZ.
clinically apparent, advanced disease is already present. Multiple • The inferotemporal macula is the earliest location of hydroxy-
diagnostic modalities are useful to aid in recognizing toxicity at chloroquine toxicity (Marmor 2012).
its earliest detectable point, before clinically apparent signs or • Thickness map may be more sensitive to identifying early
symptoms. These modalities include fundus autofluorescence, toxicity than structural B-scan.
visual field testing, multifocal electroretinogram (mfERG), and • “The flying-saucer sign” becomes present with more advanced
OCT. Of these, OCT is the most readily available and one of or moderate toxicity.
the most sensitive for detecting early pathologic changes.
The earliest sign of toxicity on OCT is loss of the outer retinal
REFERENCES
layers (inner segment/outer segment/ellipsoid zones [IS/OS/EZ]) Marmor MF. Comparison of screening procedures in hydroxychloroquine
in a parafoveal distribution Fig. 1. These earliest changes are toxicity. Arch Ophthalmol. 2012;130(4):461–469.
sometimes better visualized via a thickness map rather than a Marmor MF, Kellner U, Lai TY, et al. Recommendations on screening for
structural B-scan. With progressive loss of the perifoveal outer chloroquine and hydroxychloroquine retinopathy (2016 Revision). Ophthalmol-
ogy. 2016;123(6):1386–1394.
retina in moderate toxicity, the preserved central foveal tissue Melles RB, Marmor MF. The risk of toxic retinopathy in patients on long-
results in a saucer-shaped appearance or “flying-saucer” sign term hydroxychloroquine therapy. JAMA Ophthalmol. 2014;132(12):1453–
Figs. 2–4.” A bulls-eye maculopathy becomes present clinically 1460.

68
FIG. 1A and B. Early hydroxychloroquine maculopathy. (A) Signs of early 10.1
Early/mild hydroxychloroquine
maculopathy are more evident temporally (yellow arrow) than nasally (white
maculopathy
Loss of cone outer arrow). Temporally, there is attenuation of the IS/OS/EZ. Just inside this
segment tip line area is loss of the cone outer segment tip line. (B) Corresponding OCT
thickness map illustrating mild thinning of the outer retina in a parafoveal
distribution (asterisks and red box). ILM, Inner limiting membrane; RPE,

Hydroxychloroquine Toxicity
retinal pigment epithelium.

Parafoveal outer retinal attenuation


of IS/OS/EZ
S
1
N T

A I

500
250

** 400

300
303

277 295 230 266 225

* 200
287

**
252
100 ILM-RPE Thickness (µm) Fovea: 256, 64

0 µm
Overlay: ILM-RPE Transparency: 50%
High-definition mode

ILM-RPE

N T

ILM
Diversified:
Distribution
of Normals

99%
95%
5%
1%

RPE
S

Central Cube
Cube
Subfield Average
Volume
I
Thickness Thickness
(mm3)
(µm) (µm)
69
B ILM-RPE 230 9.2 256
FIG. 2. Early hydroxychloroquine
maculopathy. There is subtle outer retinal
attenuation in a parafoveal distribution.
Early/mild hydroxychloroquine maculopathy
Section 10: Hydroxychloroquine Toxicity

S Parafoveal outer retinal attenuation


3
N T

FIG. 3. Advanced hydroxychloroquine


maculopathy. There is severe outer retinal and
Advanced hydroxychloroquine maculopathy RPE loss in a parafoveal distribution (between
yellow arrowheads). As a result of significant
Severe attenuation of outer retina RPE dropout, there is localized reverse
shadowing. Thickness map (inset) shows
and RPE in parafoveal distribution global, profound macular atrophy.

223
S
3 Reverse shadowing 235

N T 210 244 164 214 202

231

I 213

70
FIG. 4A and B. Moderate to advanced hydroxychloroquine maculopathy.
Moderate-advanced hydroxychloroquine maculopathy (A) The most profound outer retinal and RPE loss is located nasally
(between arrowheads). A partial “flying-saucer” sign is evident as a 10.1
result of preservation of the outer retina in the central fovea. (B) Severe,
generalized outer macular atrophy that is quite striking is evident on the
corresponding thickness map.

Hydroxychloroquine Toxicity
Severe attenuation of Partial
outer retina and RPE “flying-saucer”
sign

S
3
N T

A I

500
229
400
231

300 229 210 161 211 210

206
200
217

100 ILM-RPE Thickness (µm) Fovea: 262, 66

0 µm
Overlay: ILM-RPE Transparency: 50%
High-definition mode

ILM-RPE

N T

ILM

Diversified:
Distribution
of Normals

99%
95%
5%
1%
RPE
S

Central Cube
Cube
Subfield Average
I Volume
Thickness Thickness
(mm3)
(µm) (µm)
ILM-RPE 161 7.8 215
71
B
SECTION 11: VITELLIFORM MACULAR DYSTROPHY

Vitelliform Dystrophy
Shilpa Desai | A. Yasin Alibhai 11.1
Summary Treatment of vitelliform dystrophy is typically monitoring
unless choroidal neovascularization (CNV) occurs. In that case,
Vitelliform dystrophy is defined as an abnormal accumulation of anti–vascular endothelial growth factor agents are indicated.
lipofuscin in or under the retinal pigment epithelium (RPE) layer
of the retina (Agrawal 2012). Vitelliform dystrophy is typically
inherited in an autosomal dominant pattern and thought to be Key Features
caused by a defect in the peripherin/retinal degeneration slow
(RDS) gene (Schatz et al. 2003).
• Vitelliform dystrophy is an abnormal accumulation of lipofuscin
in the RPE layer of the retina.
Onset is typically between 30 to 50 years of age. Patients may
report a decrease in vision or metamorphopsia. The vision loss in
• OCT shows deposits above the RPE.
vitelliform dystrophy is typically mild with many patients diagnosed
• Over time, regression of the deposits can occur and appear
as hyporeflective lucency underlying the retina.
during a screening eye examination while asymptomatic.
Retinal examination will show a yellow or pigmented deposit
• Vitelliform dystrophy can be associated with subretinal fluid
or choroidal neovascularization.
in the macula. The disease is typically bilateral, but may be
asymmetric. Fluorescein angiography will show blockage at the
• Treatment is typically monitoring or treatment of associated
CNV, if indicated.
area of lipofuscin deposition with late hyperfluorescence of the
lesion resulting from staining (Fig. 1). OCT will show deposits in REFERENCES
or beneath the RPE layer (Fig. 2). Vitelliform dystrophy can be Agrawal A. Gass’ Atlas of Macular Diseases. 5th ed. Chapter 5: Heredodys-
associated with subretinal fluid or choroidal neovascularization. trophic disorders affecting the pigment epithelium and retina. Philadelphia:
Elsevier; 2012:239–426.
The differential diagnosis of vitelliform dystrophy includes Schatz P, Magnus A, Eksandh L, et al. Macular appearance by means of OCT
other pattern dystrophies including Best disease, age-related and electrophysiology in members of two families with different mutations in
macular degeneration, choroiditis, and idiopathic choroidal RDS (the peripherin/RDS gene). Acta Ophthalmol Scand. 2003;81(5):500–507.
neovascularization.

FIG. 1. Color fundus, red-free, and fluorescein angiography images showing accumulation of lipofuscin material at the fovea.

FIG. 2. OCT scan corresponding


Pigment migration
to Fig. 1. There is a large retinal
pigment epithelium detachment
with hyperreflective material within
it corresponding to the vitelliform
lesion. PED, pigment epithelial
detachment.

Large PED containing


hyperreflective material

72
SECTION 12: MACULAR TELANGIECTASIA

Macular Telangiectasia
Carlos A. Moreira Neto | Carl Rebhun 12.1
Summary • OCT:
Macular telangiectasia manifests as retinal vascular anomalies in • Cystoid macular edema is most prominent feature, although
middle-aged or older adults. Abnormalities of the retinal capillary subretinal fluid may also be present (Fig. 2).
bed include vessel dilatation and tortuosity, aneurysms, vascular • Cystoid intraretinal edema is present that could be associated
leakage, and deposition of hard exudates. Type 1 macular tel- with subretinal fluid.
angiectasia, thought to be a form of Coats’ disease, is typically
unilateral and is more commonly seen in males. Telangiectatic Type 2
blood vessels are seen temporal to the macula and are associated • Bilateral.
with exudates. Type 2 macular telangiectasia is a bilateral disease • Microaneurysmal abnormalities are in the temporal parafovea.
with no sex predilection. There is a loss of the parafoveal retinal • RPE hyperplasia is present (Fig. 3).
transparency with ectasia of temporal capillaries. Exudation is • OCT:
rarely seen in type 2. With disease progression, crystal deposition • Area of low intraretinal reflectivity with lamellar defects at
and retinal pigment epithelium (RPE) hyperplasia can occur and multiple layers typically located just temporal to the foveal
CNV may develop. center. (Fig. 4).
• Pigment deposition and atrophy may develop with chronic
Key Points disease.
• OCTA:
Type 1 • Foveal avascular zone may become irregular.
• Male and unilateral. • Capillary drop out with reduced vascular density.
• Presence of exudation (Fig. 1). •Distortion and dragging (Fig. 5).

FIG. 1. Color fundus photograph shows


macular exudation in a patient with type 1
macular telangiectasia.

73
FIG. 2. OCT (corresponding
to the eye with type 1 macular
telangiectasia seen in Fig.
1) before (A) and after (B–D)
treatment. (A) Note subretinal
fluid and intraretinal cystic cavities
Section 12: Macular Telangiectasia

of low and medium reflectivity


Hard and hyperreflective deposits
exudates Subretinal Intraretinal within the retina, corresponding
fluid cysts to hard exudates. Visual acuity of
20/200. (B) OCT 3 months after
A treatment with focal grid laser
and one intravitreal bevacizumab
injection. Visual acuity is
20/60. (C) OCT 3 months after
second focal grid laser and one
intravitreal bevacizumab injection.
Visual acuity is 20/30. (D) OCT 3
months after (C). Visual acuity is
20/30.
Intraretinal
cysts

FIG. 3. Color fundus


photographs of a subject with
type 2 macular telangiectasia
show loss of the foveal reflex
with subtle microaneurysmal
abnormalities in the temporal
parafoveal region in both eyes.
Color photograph of the right
eye shows RPE clumping and
hyperplasia along with foveal
atrophy.

74
Loss of tissue from outer nuclear layer
12.1
leaving hyporeflective cavities

Macular Telangiectasia
Pigment epithelium
clumping

Hyporeflective intraretinal cavity

FIG. 4. OCT (corresponding to the subject in Fig. 3) in macular telangiectasia type 2. In the right eye (top image) there is loss of tissue from the outer
nuclear layer and atrophy of the IS/OS/ellipsoid junction. In the left eye (bottom image) there is a small intraretinal hyporeflective cavity.

FIG. 5. OCT angiography shows superficial


segmentation (corresponding to the right eye
of the subject in Fig. 3) in type 2 macular
telangiectasia. Note the irregular foveal
avascular zone.

75
SECTION 13: ISOLATED CYSTOID MACULAR EDEMA

Isolated Cystoid Macular Edema


Shilpa Desai | A. Yasin Alibhai 13.1
Summary Key Points
Cystoid macular edema (CME) is defined as retinal thickening • Cystoid macular edema (CME) is defined as retinal thickening
in the macula due to leakage and accumulation of fluid in the within the macula.
intracellular spaces of the macula. Symptoms of CME include • OCT shows large, hyporeflective cystic spaces located
vision loss, decreased color/contrast sensitivity, metamorphopsia, predominantly within the outer plexiform layer, although the
micropsia, or scotoma. CME can be caused by a myriad of inner plexiform and nuclear layers may also be involved.
etiologies, most typically occurring following ocular surgery. • Severe cases may include spillover of fluid into the subretinal
Intraretinal fluid accumulates preferentially within the outer space.
plexiform layer. Underlying photoreceptor function is affected
by the fluid and changed architecture, which results in vision BIBLIOGRAPHY
loss. The clinical appearance is that of many small cystic cavities Afshar AR, Fernandes JK, Patel RD, et al. Cystoid macular edema associ-
ated with fingolimod use of multiple sclerosis. JAMA Ophthalmol. 2013;
bunched together with the fovea into a petaloid arrangement (Fig. 113(1):103–107.
2). Fluorescein angiography shows petalloid leakage in the macula. Augustin A, Loewenstein A, Kuppermann BD. Macular edema. General
OCT show cystic accumulation of fluid in the macula (Fig. 3). pathophysiology. Dev Ophthalmol. 2010;47:10–26.
CME is most often self-limited. When treatment is required, it is Rotsos TG, Moschos MM. Cystoid macular edema. Clin Ophthalmol. 2008;
24:919–930.
geared towards the specific underlying cause typically including
topical and intravitreal corticosteroids.

76
FIG. 1. Color fundus photograph showing loss of the normal foveal reflex 13.1
due to cystoid macular edema in a patient status post vitrectomy for
retinal detachment repair.

Isolated Cystoid Macular Edema


Blockage of choroidal flush Petaloid pattern

A B

FIG. 2. Fluorescein angiogram corresponding to Fig. 1. Early phase (A) demonstrates blockage of choroidal flush from edema. Late phase image
(B) shows leakage in a petaloid pattern.

Multiple intraretinal cysts Multiple intraretinal cysts

N
1
T
Subretinal fluid

FIG. 3. OCT B-scan corresponding to Figs. 1 and 2. Retinal thickening FIG. 4. OCT B-scan in a patient with pars planitis and severe CME
and intraretinal fluid primarily in the outer plexiform layers is seen. shows multiple intra retinal cysts primarily in the outer plexiform layer,
but also involving the inner nuclear layer. Additionally, subretinal fluid is
present.

77
SECTION 14: OTHER DISORDERS AFFECTING THE MACULA

Angioid Streaks
Shilpa Desai | A. Yasin Alibhai 14.1
Summary CNV is the main cause for vision loss and can be treated with
anti–vascular endothelial growth factor therapy (Martinez-Serrano
Angioid streaks are linear cracks in Bruch’s membrane. They et al., 2016).
are caused by a breakdown in the collagen and elastic lamina
of Bruch’s membrane. There are a number of potential causes
of angioid streaks, including pseudoxanthoma elasticum, Ehler- Key Points
Danlos syndrome, Paget’s disease, sickle cell disease, and other
hemoglobinopathies (Paton, 1972). Angioid streaks may cause • Angioid streaks are linear cracks in Bruch’s membrane resulting
vision change in three different ways. First, the streak may travel from breakdown in the elastic and collagen lamina.
through the fovea, leading to retinal pigment epithelium (RPE) loss. • Visual symptoms are caused by secondary hemorrhage or
Mild trauma to the eye can cause rupture of the choroid leading CNV.
to submacular hemorrhage and vision loss. Finally, secondary • OCT shows breaks in Bruch’s membrane–RPE complex of
choroidal neovascularization (CNV) can cause hemorrhage and an angioid streak in cross section.
macular edema. Clinical examination reveals orange-red to • Secondary CNV, which is type 2, can be effectively detected
brown linear irregularities extending radially from the peripapil- and monitored with OCT.
lary region into the peripheral fundus. Fluorescein angiography
shows irregular hyperfluorescence along the streak secondary BIBLIOGRAPHY
to overlying RPE deposition (Fig. 1). OCT shows the angioid Martinez-Serrano MG, Rodriguez-Reyes A, Guerrero-Naranjo JL, et al. Long-
term follow-up of patients with choroidal neovascularization due to angioid
streaks in cross section as a discontinuity in Bruch’s membrane streak. Clin Ophthalmol. 2016;11:23–30.
(Fig. 2). Although no specific treatment can address angioid Paton D. The Relation of Angioid Streaks to Systemic Disease. Springfield,
streaks, any underlying systemic condition should be addressed. IL: Charles C Thomas; 1972.

FIG. 1. Color fundus and


fluorescein angiogram
photographs show RPE loss over
angioid streaks radiating from the
peripapillary zone (white arrows).
There is secondary CNV present
within the nasal macula.

FIG. 2. OCT B-scan


corresponding to Fig. 1.
Subretinal There are focal breaks in the
Type 2 CNV
fluid RPE–Bruch’s membrane (BM)
complex in the area of an angioid
streak. Subretinal fluid and
subretinal hyperreflective material
associated with a type 2 CNV
lesion are also identified.
Disruption in RPE/BM complex
S
1
N T

78
X-Linked Juvenile Retinoschisis
Jay S. Duker 14.2
Summary Key OCT Features
X-linked retinoschisis or juvenile retinoschisis (XLJR) is an inherited • Splitting (schisis) of both inner and outer retinal layers occurs
disorder affecting only males. It is caused by mutations in the (Figs. 1 and 3).
retinoschisin gene (RS-1). There is considerable variability in the • Posterior pole retinal cystic changes in both inner and outer
onset and severity of the disease. retinal layers resemble cystoid macular edema but extend
The clinical hallmarks are cystic retinal changes in the macula beyond the macula and do not result in extensive macular
(stellate maculopathy) and peripheral retinal elevations as the thickening.
result of retinoschisis (Figs. 1 and 2). Vitreous veils over the
peripheral schisis, vitreous hemorrhage, and retinal detachment
also can occur. OCT is the most useful imaging modality available
to confirm the diagnosis.

FIG. 1. X-linked schisis with OCT. Note splitting of both inner and outer FIG. 2. Corresponding color fundus photograph showing extensive
retina with no evidence of retinal detachment. peripheral retina elevation from retinoschisis.

FIG. 3. Macular OCT of a 16-year-old male patient showing typical inner and outer schisis diffusely through the posterior pole.

79
Oculocutaneous Albinism
Jay S. Duker 14.3
Summary not have nyctalopia or color vision problems. The classic clinical
finding is blunting of the foveal reflex (Fig. 2). In patients with
Ocular albinism can occur clinically in several forms. If only the OCA, it is important to rule out Hermansky-Pudlak syndrome
eyes are involved, it is termed ocular albinism, which is most and Chediak-Higashi syndrome because these disorders can be
frequently inherited as X-linked recessive. If both the skin and associated with important hematologic abnormalities.
the eyes are affected, it is referred to as oculocutaneous albinism
(OC albinism). OC albinism is most often inherited autosomal
recessively. Clinically, the fundus is always hypopigmented, but
it can be difficult in a young child to differentiate normal from
Key OCT Finding
disease. However, most affected children have nystagmus and • Absence or severe blunting of the foveal depression is present
iris transillumination (Fig. 1). Interestingly, affected individuals do (Fig. 3).

FIG. 1. Typical iris trans-illumination defects


are visualized with retro-illumination.

80
FIG. 2. Representative fundus photograph 14.3
of a patient with oculocutaneous albinism.
There is blunting of the foveal reflex and
hypopigmentation of the entire fundus.

Oculocutaneous Albinism
S S
1 1
T N T N

A I
B I

S
1
T N FIG. 3A–C. Typical OCT appearance of albinism in three different
patients. Note that the horizontal b-scans were obtained such that
C I the central fovea was bisected in all patients. However, the foveal
depression is absent as the key feature of disease.

81
Subretinal Perfluorocarbon
Jay S. Duker 14.4
Summary Key OCT Features
Perfluorocarbon (PFC) is a dense, synthetic liquid used during • There is a dome-shaped, hyperreflective space with a round
vitrectomy surgery to flatten the retina. Inadvertent migration top and flat bottom, located under the retina (Figs. 1 and 2).
of this liquid under the potential subretinal space is possible. • The internal reflectivity is comparable to the normal vitreous
OCT is the most definitive diagnostic modality for detecting cavity.
the presence of subretinal PFC, with which it has a distinct • The overlying retinal layers are bunched into a thin sheet and
appearance. It is important to differentiate this entity from other appear mostly hyperreflective.
subretinal pathologic conditions in which management can be • Reverse shadowing may be present throughout the underlying
entirely different. choroid.

82
14.4
Retina
Similar hyporeflectivity to vitreous space

Retina

Subretinal Perfluorocarbon
Perfluorocarbon liquid

S
10
Perfluorocarbon liquid
N T S
1
N T
A I

FIG. 2. PFC liquid bubble underneath the macula showing characteristic


features, including round top, flat bottom, and compressed overlying
retina.

N T

I
B

FIG. 1. (A) Structural OCT shows PFC liquid underneath the nasal
macula. (B) Thickness map is shown, centered on PFC bubble.

83
SECTION 15: DIABETIC RETINOPATHY

Diabetic Macular Edema


Nadia K. Waheed 15.1
Summary growth factor. OCT is the most important ancillary test in DME
diagnosis, monitoring treatment, and following progression.
Diabetic macular edema (DME) is characterized by thickening
and edema of the macula that can develop at any stage of
diabetic retinopathy. High blood glucose levels damage the Key OCT Features
retinal microcirculation, resulting in abnormal permeability and
ischemia. Increased vessel permeability results in fluid and lipid-
• Sub- and intra-retinal fluid accumulation are the key features
of DME that are evident on OCT (Fig. 3).
rich exudate leakage into the surrounding retina, distorting normal
retinal architecture and reduces visual acuity if near the fovea
• There is decreased reflectivity of outer retinal layers on OCT
due to increased foveal thickness.
(Figs. 1 and 2). Clinically significant macular edema has been
largely replaced by macular edema involving the center as an
• Exudates can be seen as hyper-reflective spots within the
retina.
indication for treatment in the era of anti–vascular endothelial

FIG. 1. Color fundus image shows hard


exudates, intraretinal hemorrhages,
microaneurysms, and cotton wool spots.

84
15.1

Diabetic Macular Edema


FIG. 2. Early frame fluorescein angiography highlights the microaneurysms, and late phase shows diffuse leakage across the posterior pole of the
retina.

500 OD-ILM Thickness


Diversified:
349
400 Distribution
of Normals
300 528
99%
441 559 713 537 354
200 95%
530 5%
100
331
1% Intraretinal cysts
0 m

Hard exudates

S
1
T N
Subretinal fluid

FIG. 3. OCT B-scan through the central macula shows thickening with cystic changes. The area and extent of thickening can be visualized by the
false color rendering of the thickness map over the C-scan (inset). The retinal thickness map also provides quantitative information about thickening.
Hard exudates can be seen as a hyperreflective cluster. There is also a trace of subretinal fluid with overlying distortion of the inner segment/outer
segment/ellipsoid zone. ILM, Inner limiting membrane.
85
Nonproliferative Diabetic Retinopathy
Nadia K. Waheed 15.2
Summary the development of diabetic macular edema (DME), which can
occur at any stage of the disease (Fig. 3).
Nonproliferative diabetic retinopathy (NPDR) represents the earliest
stages of retinopathy caused by diabetes. Damage to the small
blood vessels of the retina can result in the formation of nerve
Key OCT Features
fiber layer infarcts (cotton wool spots), hard exudates (Fig. 1), • Hard exudates appear as hyperreflective clusters in the retina.
and intraretinal hemorrhages. Other microvascular abnormalities, • MAs have homogeneous inner reflectivity and an outer rim of
including microaneurysms and dilated or tortuous vessels, also hyperreflectivity.
can develop (Fig. 2). Venous beading and intraretinal microvascular • Cotton wool spots manifest as areas of hyperreflectivity within
abnormalities (IRMA) can arise in severe NPDR. NDPR is classified the nerve fiber layer.
into mild, moderate, and severe based on the risk for developing
neovascularization. Vision loss in NPDR primarily occurs from

Hard exudate

T N

FIG. 1. Color fundus image and corresponding OCT scan shows hard exudate. Hard exudates appear as irregularly shaped, intraretinal,
hyperreflective lesions on OCT imaging.

86
15.2
Microaneurysm

Nonproliferative Diabetic Retinopathy


S

FIG. 2. Color fundus image and corresponding OCT scan show microaneurysms.

Intraretinal
fluid

FIG. 3. Color fundus image and corresponding OCT scan shows intraretinal fluid.

87
Proliferative Diabetic Retinopathy
Nadia K. Waheed 15.3
Summary macula-involved tractional detachment is generally permanent.
OCT is quite useful in documenting the presence of tractional
High blood glucose levels over a prolonged period damage the retinal detachments within the macula, particularly if the area
retinal microvasculature and result in ischemia. In response, factors affected is shallowly detached.
such as vascular endothelial growth factor are released from
ischemic retina, inducing the formation of new blood vessels.
These new blood vessels are often fragile and prone to leaking.
Key Points
Proliferative diabetic retinopathy (PDR) is characterized by • PDR is characterized by the formation of new retinal blood
neovascularization arising from the optic disc and retina, which vessels.
may cause preretinal and vitreous hemorrhage (Figs 1 and 2). • Neovascularization on OCT can be seen as loops of hyper-
Subsequent fibrosis of the new vessels creates tractional forces reflective blood vessels protruding into the retina.
leading to retinal detachment (Fig. 3). Vision loss may occur as • Tractional retinal detachments are defined by the presence of
a result of PDR-induced hemorrhage, but this often resolves subretinal fluid. They are distinguished by overlying tractional
as a result of reabsorption of blood. Vision loss because of bands.

FIG. 1. Neovascularization of the superior temporal arcade of the retina and optic disc is seen on both the color fundus photograph and the
accompanying fluorescein angiogram.

88
15.3

NVD

Proliferative Diabetic Retinopathy


FIG. 2. OCT scan corresponding to Fig. 1. Line scan through the area of the neovascularization of the optic disc reveals hyperreflective
neovascularization into the vitreous cavity. NVD, neovascularization of the disc.

FIG. 3. Tractional retinal detachment seen


on OCT corresponds to superotemporal
neovascularization seen on Fig. 1. There is
thickening of the retina with associated cystic
changes.

Cystic retinal changes

89
SECTION 16: RETINAL VENOUS OCCLUSIVE DISEASE

Branch Retinal Vein Occlusion


Caroline R. Baumal 16.1
Summary Key OCT Findings
Branch retinal vein occlusion (BRVO) is due to blockage of one • OCT is key to diagnosing associated macular edema, deter-
of the branches of the central retinal vein. The pathogenesis mining if the fovea is involved, and determining response to
involves mechanical compression from an atherosclerotic tributary therapy.
of the retinal artery, degenerative vascular changes, and thrombus • OCT findings include cystoid macular edema, intraretinal
formation. It is the second most common retinovascular disorder hyperreflectivity from hemorrhages, shadowing from edema
after diabetic retinopathy. The 5-year incidence is 0.6% and and hemorrhages, and subretinal fluid (Fig. 1).
increases with age. Typical age of onset is between 60 and 70 • Even with resolution of cystoid macular edema, disruption of
years, with a variable range. Risk factors include hypertension, the foveal photoreceptor layer, specifically the ellipsoid layer
cardiovascular disease, hyperlipidemia, obesity, and glaucoma. and external limiting membrane, have been associated with
Hypercoagulability may play a role in some cases. Clinical exami- a poorer visual prognosis.
nation reveals dilation and tortuosity of the obstructed branch
retinal vein associated with retinal hemorrhages. Cotton wool BIBLIOGRAPHY
Kang HM, Chung EJ, Kim YM, et al. Spectral-domain optical coherence
spots, lipid exudate, macular edema, retinal neovascularization,
tomography (SD-OCT) patterns and response to intravitreal bevacizumab
and capillary nonperfusion may be concomitant features. therapy in macular edema associated with branch retinal vein occlusion.
Retinal imaging with OCT and fluorescein angiography can aid Graefes Arch Clin Exp Ophthalmol. 2013;251(2):501–508.
in prognostication and determination of therapy. BRVO usually Lim HB, Kim MS, Jo YJ, et al. Prediction of retinal ischemia in branch retinal
has a good prognosis. Visual acuity depends on the area of retinal vein occlusion: spectral-domain optical coherence tomography study. Invest
Ophthalmol Vis Sci. 2015;56(11):6622–6629.
ischemia and status of the fovea. Treatment of the sequelae of Spaide RF, Lee JK, Klancnik JK Jr, et al. Optical coherence tomography of
BRVO (specifically macular edema or neovascularization) may branch retinal vein occlusion. Retina. 2003;23:343–347.
include anti–vascular endothelial growth factor agents, intraocular
corticosteroids, and laser photocoagulation.

90
16.1

Branch Retinal Vein Occlusion


A B

C D

E F

G H

FIG. 1(A–H). A. Fundus image of the left eye demonstrating a superotemporal branch vein occlusion with prominent retinal hemorrhages. (B) En-face
OCT scan demonstrates cystoid macular edema with hyperreflectivity secondary to intraretinal cysts. (C) B-scan demonstrates cystoid macular
edema. (D) Fundus photograph of the left eye demonstrating resolution of the hemorrhages. (E and F). OCT angiography of the superficial and deep
plexuses demonstrate vessel dilation, vessel tortuosity and capillary dropout, indicating retina ischemia. (G) OCT-B scan with angiography flow overlay.
(H) OCT-B scan of the macula showing resolution of the edema but residual external limiting membrane and possible IS/OS junction disruption in the
outer retina.

91
Central Retinal Vein Occlusion
Nadia K. Waheed | Caroline R. Baumal 16.2
Summary and neovascularization. Treatment includes maximizing control
of systemic risk factors, intravitreal injection of anti–vascular
Central retinal vein occlusion (CRVO) results from occlusion of endothelial growth factor (anti–VEGF) or corticosteroid agents, and
the central retinal vein at or proximal to the lamina cribrosa. panretinal laser photocoagulation for significant neovascularization.
The central retinal vein is the major venous drainage system
for the inner retina, and disruption of flow can result in severe
visual loss from ischemia, macular edema, and/or neovascu-
Key OCT Features
larization. Pathogenesis of CRVO is hypothesized to follow the
principles of Virchow’s triad for venous thrombosis, involving • OCT is key to diagnosing associated macular edema, evaluating
vascular wall damage, stasis of blood flow, and hyperco- the involved retinal layers, and determining response to therapy
agulability. The prevalence and 15-year incidence of CRVO is (Figs. 2 and 3).
0.2%. • Visual acuity inversely correlates with macular thickening
Risk factors include elderly age, diabetes mellitus, hyper- measured with OCT.
lipidemia, hypertension, cardiovascular disease, obesity, ocular • After treatment of macular edema, discontinuity of the photo-
hypertension, glaucoma, and hypercoagulable states. receptors (especially the ellipsoid zone) and retinal architecture
CRVO is classified as either nonischemic, which is a milder correlate with worse visual acuity.
form, or as ischemic; this has implications for prognosis and
treatment. Clinical findings include dilation and tortuosity of
BIBLIOGRAPHY
the retinal veins and retinal hemorrhages in all four quadrants Laouri M, Chen E, Looman M, et al. The burden of disease of retinal vein
(Figs. 1A, B, C), optic disc edema, cotton wool spots (nerve occlusion: review of the literature. Eye (Lond). 2011;25(8):981–988.
fiber layer infarcts), lipid exudate, cystoid macular edema, retinal Martinet V, Guigui B, Glacet-Bernard A, et al. Macular edema in central
ischemia, and later sequelae of iris and angle neovascularization retinal vein occlusion: correlation between optical coherence tomography,
angiography and visual acuity. Int Ophthalmol. 2012;32(4):369–377.
leading to glaucoma in ischemic CRVO subtypes. Fluorescein Shin HJ, Chung H, Kim HC. Association between integrity of foveal photorecep-
angiography findings include delayed venous filling, retinal tor layer and visual outcome in retinal vein occlusion. Acta Ophthalmol.
capillary nonperfusion, staining of retinal veins, retinal edema, 2011;89:e35–e40.

92
16.2

Central Retinal Vein Occlusion


A B C

S S

N T N T

D E
I I

S S

N T N T

F G
I I

N T

H
I

FIG. 1. (A–C) Color and red-free photographs of a patient presenting with mild central retinal vein occlusion and cystoid macular edema (CME). (D)
OCT B-scan reveals CME with large central cysts in the inner nuclear layer and smaller perifoveal cysts. Note the vitreomacular attachment. First anti-
VEGF injection was given. (E) At 4 weeks after anti-VEGF treatment, the central subfoveal thickness is reduced to 362 and acuity improved to 20/40.
(F) At 3 months after two anti-VEGF injections, cystoid macular edema resolved and acuity returned to 20/20. Anti-VEGF injection was given. (G) After
three anti-VEGF injections, the CME was completely resolved. At this time no treatment was given. (H) CME recurred 6 months after presentation with
a central subfoveal thickness of 655 microns. Her last anti-VEGF was 3 months before recurrence of CME.

93
Superficial Deep
Section 16: Retinal Venous Occlusive Disease

FIG. 2. En face images highlight that black cysts are more prominent in the deep retina than the superficial retina.

94
16.2

Central Retinal Vein Occlusion


A

FIG. 3. (A) Two OCT B-scans centered on the fovea. Intraretinal cysts are large centrally and decrease in size in the perifoveal region. A small amount
of subretinal fluid and an epiretinal membrane can be seen. Central foveal thickness measures 560 µm. (B) Four weeks after anti-VEGF treatment, the
cysts have resolved but the residual epiretinal membrane is apparent. (C) OCT shows abnormally widened capillary spaces even though no CME is
present.

95
SECTION 17: RETINAL ARTERIAL OCCLUSIVE DISEASE

Branch Retinal Artery Occlusion


Caroline R. Baumal 17.1
Summary Key OCT Findings
Branch retinal artery occlusion (BRAO) results from obstruction • OCT can demonstrate structural damage to the inner retina
of one of the branches of the central retinal artery. The most in BRAO.
common cause is emboli secondary to either carotid plaques or • Acutely, OCT demonstrates increased hyperreflectivity in the
cardiac. Less common, nonembolic causes include vasospasm affected inner retinal layers and retinal thickening where the
and inflammatory and hypercoagulable disorders. Medical evalu- occlusion occurred.
ation for cardiovascular and carotid disease is indicated because • Inner retinal changes in acute BRAO can shadow details of
of the association with increased morbidity and mortality. Patients the underlying outer retinal layers.
are typically 70 years or older and present with painless, unilateral, • In chronic BRAO, OCT reveals atrophy in the nerve fiber layer
partial visual loss. and inner retina, while the outer nuclear layer and adjacent
Amaurosis fugax preceding persistent loss of vision may photoreceptor/RPE layers retain their physiologic thickness.
represent emboli causing temporary occlusion of the retinal artery • OCTA can characterize flow deficits in eyes with acute BRAO
with subsequent release into the distal circulation. Location of the to differentiate the involvement of the inner retinal plexuses.
BRAO is often at the retinal artery bifurcation, where the artery
lumen is narrower. Ischemic retinal whitening and inner retinal BIBLIOGRAPHY
Chu YK, Hong YT, Byeon SH, et al. In vivo detection of acute ischemic
edema develop along the path of the occluded branch artery. damage in retinal arterial occlusion with optical coherence tomography: a
Other features include visible emboli in 60% of eyes, narrowing “prominent middle limiting membrane sign.”. Retina. 2013;33(10):2110–2117.
and boxcarring of the affected vessel, segmentation of blood flow, Coady PA, Cunningham ET, Vora RA, et al. Spectral domain optical coherence
and cotton wool spots. Clinical features are usually diagnostic in tomography findings in eyes with acute ischaemic retinal whitening. Br J
Ophthalmol. 2015;99(5):586–592.
acute BRAO, but fluorescein angiography and OCT are useful to Ritter M, Sacu S, Deak GG, et al. In vivo identification of alteration of inner
demonstrate BRAO features (see Fig. 1). OCT can be a great aid neurosensory layers in branch retinal artery occlusion. Br J Ophthalmol.
in diagnosing inner retinal atrophy and loss of the inner retinal 2012;96(2):201–207.
layer architecture after acute BRAO findings have resolved.

96
17.1

Branch Retinal Artery Occlusion


A B

C D

FIG. 1. (A) Color photograph of acute superotemporal BRAO. (B)


Fluorescein angiogram at 22 seconds shows delayed filling of the
affected artery and hypoperfusion of the affected area. (C) At 2
minutes the affected artery has filled by retrograde flow. (D) The
area of the BRAO superior to the fovea is highlighted in orange.
(E) Spectral domain OCT reveals increased hyperreflectivity of
the inner retinal and nerve fiber layers adjacent to the optic
nerve. Temporal to the nerve there is hyperreflectivity of the
E middle retinal layers (paracentral acute middle maculopathy) and
shadowing of the deeper structures and photoreceptors.

97
Central Retinal Arterial Occlusion
Nadia K. Waheed | Caroline R. Baumal 17.2
Summary to neural tissue occurs after only 90 minutes, and there is no
effective treatment for embolic causes of CRAO.
Central retinal artery occlusion (CRAO) results from obstruc- Diagnosis is determined based on history and clinical features.
tion of the central retinal artery, which is a major branch of the Fluorescein angiography shows delayed central artery filling. OCT
ophthalmic artery. The central retinal artery provides oxygen acutely reveals inner retinal thickening, hyperlucency, and loss of
and nutrients to the inner retina and the surface of the optic distinction between the inner retinal layers. Chronic CRAO shows
nerve. The cause of the occlusion is often embolic, but may also inner retinal thinning with intact retinal pigment epithelium (RPE).
be thrombotic, inflammatory, traumatic, or from vasospasm. In
patients over 70 years of age, giant cell arteritis is more likely to
be the underlying cause than in younger patients. Risk factors
Key OCT Findings
include atherosclerosis, cardiac disease, coagulopathies, age • OCT in acute CRAO demonstrates inner retinal thickening,
range between 60 and 65 years, male gender, smoking, and and the RPE, photoreceptors, and outer nuclear layer appear
diabetes mellitus. Additional risk factors include endocarditis, relatively unaffected (Figs. 1 and 2).
atrial myxoma, inflammatory diseases of the blood vessels, and • Chronic CRAO demonstrates inner retinal atrophy and loss
predisposition to blood clots. Patients typically present with of the foveal depression (Fig. 2).
sudden, acute, painless, unilateral loss of vision. Acuity is count
fingers/hand motions in 75% to 90% of eyes at presentation. BIBLIOGRAPHY
Ahn SJ, Woo SJ, Park KH, et al. Retinal and choroidal changes and visual
Fundoscopy reveals a central “cherry red spot,” surrounded by outcome in central retinal artery occlusion: An optical coherence tomography
pale ischemic retina. The pale white color is secondary to inner study. Am J Ophthalmol. 2015;159(4):667–676.
retinal ischemia; the red spot is the spared central fovea, which Chen H, Chen X, Qiu Z, et al. Quantitative analysis of retinal layers’ optical
receives its blood supply solely from the choroidal circulation. intensities on 3D optical coherence tomography for central retinal artery
occlusion. Sci Rep. 2015;5:9269. doi:10.1038/srep09269.
The artery can recanalize over time, and the inner edema clears Kapoor KG, Barkmeier AJ, Bakri SJ. Optical coherence tomography in retinal
with ensuing atrophy. Optic atrophy leads to permanent loss of arterial occlusions: case series and review of the literature. Semin Ophthalmol.
vision. The prognosis for visual recovery is poor, and over 90% 2015;30:74–79.
have final vision count fingers or worse. Irreversible damage

98
FIG. 1. Acute CRAO with perifoveal whitening 17.2
and a cherry red spot.

Central Retinal Arterial Occlusion


S
1
T N

S
1 S

N T
N T

I I

A B

FIG. 2. (A) Inner retina thickening and hyperlucency in acute CRAO, shadowing the ellipsoid and choroidal details. (B) At 1 year later there is loss of
the inner retinal architecture and no distinct foveal depression. The RPE remains intact.

99
SECTION 18: NONINFECTIOUS UVEITIS

Birdshot Retinochoroidopathy
Eduardo Uchiyama 18.1.1
Summary (CNV), and lamellar macular hole formation. In advanced cases,
diffuse retinal dysfunction can cause severe vision loss associated
Birdshot retinochoroidopathy (BSRC) is a bilateral, chronic, with retinal thinning and disruption of the inner segment/outer
idiopathic inflammatory disorder that affects predominantly the segment/ellipsoid zone (IS/IO/EZ) on OCT.
retina and choroid. It is characterized by mild anterior chamber
inflammation, vitritis, retinal vasculitis, and the presence of multiple
hypopigmented fundus lesions. BSRC is strongly associated Key OCT Features
with human leukocyte antigen (HLA)-A29 (Fig. 1). OCT is the
most common imaging modality used to monitor disease activity • CME is the most common cause of vision loss.
and response to treatment. Macular changes visible on OCT at • Retinal thinning and disruption of the IS/OS/EZ worsen as
disease progresses and correlate with vision loss.
presentation vary widely and include normal appearance, mild
disruption of retinal architecture (Figs. 2–7), and even severely • After successful treatment, OCT demonstrates resolution of
CME and, in some patients, improvement of outer retinal
edematous or atrophic changes. Detecting and monitoring cystoid
changes.
macular edema (CME) is particularly important, because this is the
most common cause of decreased vision in affected patients with • Enhanced depth imaging–OCT is useful to best reveal choroidal
thinning in long-standing cases (Figs. 4–5).
BSRC. OCT with enhanced depth imaging (EDI-OCT) may reveal
choroidal thinning in long-standing cases, in addition to macular • Similar to other inflammatory chorioretinal conditions, ERM
and CNV formation are sometimes manifest secondarily.
thinning. There is no consistent relationship between lesions
seen on OCT and lesions seen clinically or on indocyanine green
BIBLIOGRAPHY
angiography (ICGA). In addition to CME, other secondary features Uchiyama E. Birdshot retinochoroidopathy. In: Papaliodis G, ed. Uveitis: A
of BSRC associated with vision loss include epiretinal membrane Practical Guide to the Diagnosis and Treatment of Intraocular Inflammation.
(ERM) formation, retinal thinning, choroidal neovascular membrane Geneva: Springer; 2017.

Epiretinal
membrane Vitreous cell

Cystoid macular edema


S
3
N T

FIG. 1. Fundus photograph of patient with poorly controlled BSRC. FIG. 2. A patient with recently diagnosed BSRC has characteristic
Birdshot lesions are seen in the posterior pole along with perivascular OCT features, including cystoid macular edema, ERM, and vitreous cell
exudation and optic disc pallor. (yellow arrows).

100
FIG. 3. Chronic inflammation caused ERM 18.1.1
formation and associated mild cystic changes
Epiretinal membrane in this patient with BSRC. Note that this
patient has mild/early disease and has a
preserved IS/OS/EZ.

Birdshot Retinochoroidopathy
Mild cystic intraretinal
changes
Preserved IS/OS/EZ
S
3
T N

OD OS

VA = 20/200 VA = 20/400

T N N T
A

VA = 20/150 OD VA = 20/40 OS

T N N T
B

FIG. 4A and B. (A) Bilateral CME in a patient with poorly controlled BSRC (corresponding to patient in Fig. 1). Visual acuity (VA) measured 20/200
OD and 20/400 OS. (B) At 1 year after treatment with mycophenolate mofetil, CME has mostly resolved and vision improved to 20/150 OD and
20/40 OS.

101
FIG. 5A and B. (A) Bilateral CME
OD OS and subretinal fluid in a patient
Cystoid macular with BSRC. (B) At 1 month
edema after treatment with intravitreal
triamcinolone acetonide, there is
brisk resolution of both CME and
Section 18: Noninfectious Uveitis

subretinal fluid.
Cystoid macular
edema
Subretinal
fluid Subretinal
S
1 S fluid
T N 1
N T

A I
I

OD OS
After treatment with
intravitreal triamcinolone
acetonide

After treatment with


intravitreal triamcinolone
1
S
S
acetonide
T N 1
N T

B I
I

FIG. 6. Severe loss of IS/OS/EZ in a patient


with active BSRC. Visual acuity measured
Preservation of IS/OS/EZ 20/40 because of preservation of the central
IS/OS/EZ.

Severe loss of IS/OS/


EZ Patchy loss of IS/OS/
EZ

FIG. 7. Severe bilateral retinal


OD OS thinning and atrophy in a patient
with long-standing BSRC. Visual
acuity measured counting fingers
at 2 feet.

T N N T

102
Acute Posterior Multifocal Placoid
Pigment Epitheliopathy
Darin R. Goldman
18.1.2
Summary Key OCT Features
Acute posterior multifocal placoid pigment epitheliopathy • OCT abnormalities in APMPPE correlate with clinically identifi-
(APMPPE) is a poorly understood, rare, bilateral outer retinal able lesions.
and/or choroidal syndrome that predominantly affects the macula. • OCT findings in the acute phase show hyperreflectivity or
The cause is thought to be a localized inflammatory response to disruption of the outer plexiform layer contiguous through
a recent generalized viral illness. The clinical appearance is that to the RPE, which includes the photoreceptors (Fig. 1A–D,
of a multifocal array of flat, cream-colored lesions in an inkblot Fig. 2A–E).
or irregular pattern, usually centered on the macula. The clinical • OCT findings in the resolution phase may normalize or leave
course is usually self-limited, marked by an initial acute phase permanent defects in the outer nuclear layer, IS/OS/EZ, and
followed by a resolution phase. Fluorescein angiography has a RPE (Fig. 1E, Fig. 2D–F).
typical appearance with early hypofluorescence and late, irregular • Atypical OCT findings include intraretinal fluid, significant
hyperfluorescence. It often shows additional lesions that are subretinal fluid, and localized thickening of the inner retina
not seen clinically. OCT can provide useful information to help (Fig. 3).
distinguish this entity from other similar inflammatory macular
conditions. In the acute, active stage of disease, OCT shows
BIBLIOGRAPHY
multiple focal disruptions of the retinal pigment epithelium (RPE),
Goldenberg D, Habot-Wilner Z, Loewenstein A, et al. Spectral domain optical
IS/OS/EZ, and external limiting membrane (ELM), with overlying coherence tomography classification of acute posterior multifocal placoid
hyperreflectivity of the outer retina limited to the outer nuclear pigment epitheliopathy. Retina. 2012;32(7):1403–1410.
layer and outward layers. Localized thickening of the inner retina Querques G, Querques L, Bux AV, et al. High-Definition OCT Findings in
overlying active lesions has been reported. Rarely, subretinal fluid Acute Posterior Multifocal Placoid Pigment Epitheliopathy. Ophthalmic Surg
Lasers Imaging. 2010;41:e1–e6.
and intraretinal fluid within the outer retinal layers may be present, Scheufele TA, Witkin AJ, Schocket LS, et al. Photoreceptor atrophy in acute
similar to the appearance in Vogt-Koyanagi-Harada syndrome (see posterior multifocal placoid pigment epitheliopathy demonstrated by optical
Chapter 18.2). In the resolution phase, OCT findings generally coherence tomography. Retina. 2005;25(8):1109–1112.
improve with reappearance of outer retinal bands along with
lessening and eventual resolution of outer nuclear layer hyper-
reflectivity. However, permanent localized RPE irregularities and
outer retinal atrophy with loss of photoreceptors may develop.

103
FIG. 1A–E. Typical clinical
appearance (A) and fluorescein
angiography (B) of APMPPE at
presentation. (C) Corresponding
OCT in the acute phase shows
multifocal areas of disrupted IS/
Section 18: Noninfectious Uveitis

OS/EZ and ELM bands with


abnormal hyperreflectivity of
the outer retina (arrowheads)
spanning from the outer plexiform
layer to the RPE, inclusive of the
photoreceptors. There is faint
subretinal fluid associated with
each lesion (red arrows). (D) The
fellow eye shows similar findings.

A B

External limiting membrane

Multifocal disruptions of
IS/OS/EZ and ELM

Disruption of IS/OS/EZ
and ELM

104
FIG. 1A–E, cont’d. (E) At 2 weeks after presentation, OCT shows near 18.1.2
normalization of the outer retinal architecture with a restored ELM and
IS/OS/EZ band, although they remain attenuated. (Courtesy Robin A. Vora,
MD, and John Lewis, MD.)

Acute Posterior Multifocal Placoid Pigment Epitheliopathy


External limiting membrane

IS/OS/EZ

FIG. 2A–F. At presentation in


the subacute phase of APMPPE,
numerous lesions are visible
clinically (A) and many more
visible angiographically (B).
OCT shows multifocal areas of
outer retinal attenuation (yellow
circles) affecting predominantly
the photoreceptors and RPE,
with relative sparing of the outer
nuclear layer (C). At 6 months
later the active lesions are
resolved (D), leaving pigment
clumping at the level of the
RPE with a nearly complete
A B normalization of the OCT
appearance, although faint IS/OS/
EZ attenuation remains (E). This
normalization trend continues at
1 year (F). Note that the same
areas highlighted by yellow circles
Multifocal areas of outer retinal attenuation in Fig. 2C, E, and F. (Fig. 2A and
B with permission from Reichel, E.,
Duker, J., Goldman D., Vora R., Fein,
J. 2015. Handbook of retinal disease.
JP Medical, London.)

S
1
N T

C I D

S S
1 1
N T N T

E I
F I

105
FIG. 3. Atypical case of APMPPE shows significant subretinal fluid
(asterisks) and localized cystic thickening of the outer retina. These
features can resemble findings in Vogt-Koyanagi-Harada syndrome.
Section 18: Noninfectious Uveitis

*
*

Intraretinal fluid
S
1
N T

106
Multiple Evanescent White Dot Syndrome
Darin R. Goldman | Nora W. Muakkassa 18.1.3
Summary spontaneously over time as symptoms abate and white dots
resolve.
Multiple evanescent white dot syndrome (MEWDS) is thought to
be an inflammatory outer retinal disease that occurs in young,
myopic women. A flulike prodrome may occur, with symptoms
Key OCT Features
including photopsias, blurred vision, or scotomas. Findings on • Acute findings include disruption of the IS/OS/EZ.
examination may include mild vitreous cells, mild disc edema, • Inflammatory cells in the posterior vitreous, seen as hyper-
foveal granularity, and small white dots at the level of the deep reflective dots, may be seen during the acute phase.
retina. Visual field testing may reveal an enlarged blind spot. • IS/OS disruption is diffuse and does not necessarily correlate
Fluorescein angiography typically shows early wreath-like hyper- with white dots seen clinically.
fluorescence with late staining. Indocyanine green angiography • The IS/OS/EZ typically regains a normal appearance over
reveals hypocyanescent spots that are usually more numerous several months, although mild IS/OS abnormalities may persist
than the white spots noted on examination. OCT shows areas
of disruption of the inner segment/outer segment/ellipsoid zone BIBLIOGRAPHY
Li D, Kishi S. Restored photoreceptor outer segment damage in multiple
(IS/OS/EZ) throughout the posterior pole (Figs. 1 and 2). These evanescent white dot syndrome. Ophthalmology. 2009;116:762–770.
areas of disruption are more diffuse than the focal white dots Silva RA, Albini TA, Flynn HW Jr. Multiple evanescent white dot syndromes.
on funduscopic examination. Posterior vitreous cells also may J Ophthalmic Inflamm Infect. 2012;2:109–111.
be visible on OCT imaging. OCT findings typically normalize

107
Disruption of the IS/OS junction
Section 18: Noninfectious Uveitis

Vitreous cells

A Severely disrupted Mildly disrupted


B IS/OS junction IS/OS junction

IS/OS disruption

B
C Improved but persistent IS/OS junction attenuation
Improved but with persistent mild IS/OS irregularity
Resolved vitreous cell

C D Improved but still irregular IS/OS junction

FIG. 1A–C. MEWDS. (A) Late phase fluorescein angiogram reveals FIG. 2A–D. MEWDS. (A) OCT at presentation shows diffuse disruption
stippled hyperfluorescence. (B) OCT at presentation demonstrates of the IS/OS/EZ. (B) OCT through the inferior macula shows diffuse IS/
disruption of the IS/OS/EZ temporal and nasal to the fovea. (C) OCT OS disruption and inflammatory cells in the posterior vitreous, which
6 months later shows a trend toward normalization of the IS/OS appear as hyperreflective dots. (C) At 4 months later, OCT shows
appearance, although mild attenuation persists. RPE, Retinal pigment improved but persistent IS/OS attenuation. (D) At 6 months after
epithelium. (Images courtesy of Trexler Topping, MD.) presentation, OCT through the inferior macula shows resolved vitreous
cells and restoration of the IS/OS/EZ, though mild irregularity persists.
RPE, Retinal pigment epithelium. (Courtesy Jeffrey Heier, MD.)

108
Serpiginous Choroiditis
Darin R. Goldman 18.1.4
Summary choroidal ischemia associated with active lesions, which should
resolve with resolution of active disease.
Serpiginous choroiditis is an inflammatory condition of the choroid
that affects the outer retina and the RPE. The cause is unknown,
Key OCT Features
although tuberculosis should be ruled out in all cases. The clinical
course is marked by progression, regression, and recurrences. The • Active lesions feature homogeneous hyperreflectivity of the
affected eye(s) typically has a characteristic unifocal, serpentine, very outer retina that coalesces with similar subretinal material
grayish lesion located beneath the retina, typically in a peripapillary and the underlying RPE (Fig. 1).
or macular location. Over time, the active lesion regresses, leaving • Inactive lesions show generalized outer retinal and RPE atrophy.
generalized atrophy of the RPE and underlying choriocapillaris. Subretinal fibrosis also may be present (Fig. 2).
Recurrent lesions typically initiate at the edges of old inactive
BIBLIOGRAPHY
areas. Vision loss occurs as a result of atrophy underlying the Carreño E, Fernandez-Sanz G, Sim DA, et al. Multimodal imaging of macular
fovea or secondary choroidal neovascularization. serpiginous choroidopathy from acute presentation to quiescence. Ophthalmic
OCT features of active lesions include hyperreflectivity and Surg Lasers Imaging Retina. 2015;46(2):266–270.
thickening of the outer retina. Homogeneous hyperreflective Lim WK, Buggage RR, Nussenblatt RB. Serpiginous choroiditis. Surv Oph-
thalmol. 2005;50(3):231–244.
subretinal material accumulation also may occur. OCT features Punjabi OS, Rich R, Davis JL, et al. Imaging serpiginous choroidopathy with
of inactive lesions include generalized atrophy of the outer retina spectral domain optical coherence tomography. Ophthalmic Surg Lasers
and RPE. Enhanced-depth OCT imaging may be useful to detect Imaging. 2008;39(4 suppl):S95–S98.

109
Section 18: Noninfectious Uveitis

B
A

Active lesion
Normal
RPE
Inactive lesion

FIG. 1A–C. Acute serpiginous choroiditis. (A and B) Color and


red-free photographs show active choroiditis (yellow arrowheads) in
the macula emanating from older, inactive lesions that exhibit RPE
Nodular and choriocapillaris atrophy (white arrowheads). (C) OCT shows two
Irregular
Normal hyperreflectivity distinct abnormal areas: (1) acute lesion exhibits a nodular elevation of
RPE
Choriocapillaris of outer retina Abnormal hyperreflective material infiltrating the outer retina with obliteration of
C and RPE Choriocapillaris the underlying RPE and (2) inactive lesion exhibits irregular RPE and
underlying choriocapillaris.

Loss of IS/OS/EZ

Irregular
RPE
Abnormal
A B Choriocapillaris

FIG. 2A and B. At 1 week after systemic prednisone. (A) Color photograph shows disappearance of the subretinal white infiltrate (see Fig. 1) that is
replaced by developing atrophy of the RPE and choriocapillaris. (B) In the area of the previously active lesion, OCT shows loss of the inner segment/
outer segment/ellipsoid zone (IS/OS/EZ) with outer retinal atrophy, diffuse RPE irregularity with dropout, and an abnormal choriocapillaris. Note that
unaffected retina/choroid is located to the left of the arrowhead.

110
Multifocal Choroiditis and Panuveitis and
Punctate Inner Choroidopathy
Darin R. Goldman
18.1.5
Summary Key OCT Features
Multifocal choroiditis and panuveitis (MCP) and punctuate inner • Acute inflammatory lesions can best be differentiated from
choroidopathy (PIC) are idiopathic inflammatory disorders that CNVs using OCT angiography, particularly when fluorescein
primarily affect the outer retina and sub-RPE bilaterally (Spaide, angiography is inconclusive (Figs. 3 and 7).
Goldberg & Freund 2013). The two conditions exhibit many • Both inflammatory lesions and CNVs affect the outer retinal
similarities and are considered within a spectrum of the same layers and sub-RPE space, exhibiting mixed reflectivity with
disorder, with PIC being a subtype of MCP. Younger myopic dehiscence of the RPE layer. The inner segment/outer segment/
females are typically affected. MCP exhibits vitreous inflammation ellipsoid zone is often absent.
during active disease, whereas PIC typically lacks intraocular • CNV tend to be less well-defined than inflammatory lesions
inflammation. Active lesions appear as yellowish or gray circular and often have associated subretinal fluid.
subretinal irregularities. Inactive lesions appear as multifocal • Chronic or inactive lesions are well-defined hyperreflective
“punched-out” atrophic areas with pigmented borders located nodular elevations of the RPE with overlying layers intact and
throughout the fundus that can resemble those seen in presumed visible.
ocular histoplasmosis syndrome.
Secondary CNV formation is the most frequent cause of vision REFERENCE
Spaide RF, Goldberg N, Freund KB. Redefining multifocal choroiditis and
loss. The distinction between inflammatory lesion and CNV is
panuveitis and punctate inner choroidopathy through multimodal imaging.
critical but can be difficult, even with fluorescein angiography. Retina. 2013;33(7):1315–1324.
OCT, particularly OCT angiography, is extremely helpful in dif-
ferentiating between these two pathologic sequelae (Figs. 1–7). BIBLIOGRAPHY
Treatment includes systemic immunosuppression, localized Cheng L, Chen X, Weng S, et al. Spectral-domain optical coherence tomography
treatment to inflammatory lesions (steroid), and CNV (anti–vascular angiography findings in multifocal choroiditis with active lesions. Am J
Ophthalmol. 2016;169:145–161.
endothelial growth factor [anti-VEGF]). Treatment response, best Levison AL, Baynes KM, Lowder CY, et al. Choroidal neovascularisation on
monitored by OCT, can help confirm the type of underlying active optical coherence tomography angiography in punctate innerchoroidopathy
lesion. and multifocal choroiditis. Br J Ophthalmol. 2017;101(5):616–622. doi:10.1136/
bjophthalmol-2016-308806. [Epub 2016 Aug 18].

Choroidal neovascular
membrane

Organized choroidal
S S neovascular membrane
2 1
N T
Nodular RPE N T

elevation
A I
B I

FIG. 1A and B. (A) MCP with active CNV. There are two separate elevated lesions located beneath the RPE. The nasal lesion (white arrow) is
ill-defined, with loss of the RPE layer and an abnormal subretinal hyperreflective signal extending into the outer retinal layers. This area corresponds
to a CNV with associated leakage on fluorescein angiography. The temporal lesion (yellow arrow) is a well-defined nodular sub-RPE elevation with
medium reflectivity. This area did not have any definitive corresponding leakage on fluorescein angiography, suggesting no CNV. Subretinal fluid
present is also present (asterisk). (B) After treatment with anti-VEGF therapy, the temporal lesion regressed completely and the nasal lesion organized
into a well-defined hyperreflective dome-shaped elevation at the level of the RPE. (Courtesy Patrick E. Rubsamen, MD, and Eduardo Uchiyama, MD.)

111
Intraretinal fluid
Section 18: Noninfectious Uveitis

Choroidal neovascular
Subretinal
membrane and
fluid
discontinous RPE Subfoveal fibrotic pigment
epithelial detachment

S S
1
Recurrent choroidal 7
N T N T
neovascular membrane
I I

FIG. 2. At 3 months after initial anti-VEGF therapy, the patient FIG. 4. At 6 months after initial presentation, despite anti-VEGF
had recurrence active CNV with an appearance similar to the initial treatment, a subfoveal fibrotic pigment epithelial detachment developed,
occurrence (see Fig. 1). Note that the RPE is discontinuous and there is with visual acuity measuring counting fingers. (Courtesy Patrick E.
both intraretinal and subretinal fluid. (Courtesy Patrick E. Rubsamen, MD, and Rubsamen, MD, and Eduardo Uchiyama, MD.)
Eduardo Uchiyama, MD.)

Choroidal neovascular membrane Choroidal neovascular membrane

A B

FIG. 3. MCP with active CNV. OCT angiography slabs shows two distinct areas of choroidal
N T neovascularization (circles) located within the deep retinal slab. Corresponding OCT B-scan is shown
C with segmentation lines that demarcate the OCT angiography slabs (bottom). (Courtesy Patrick E.
Rubsamen, MD, and Eduardo Uchiyama, MD.)

FIG. 5. Wide-field fundus image of MCP illustrating numerous atrophic


and pigmented round lesions distributed throughout the fundus. (Courtesy
Patrick E. Rubsamen, MD, and Eduardo Uchiyama, MD.)

112
FIG. 6. Baseline OCT in a patient with inactive multifocal choroiditis. 18.1.5
(Courtesy Patrick E. Rubsamen, MD, and Eduardo Uchiyama, MD.)

Multifocal Choroiditis and Panuveitis and Punctate Inner Choroidopathy


S
6
T N

S
5 Ill-defined lesion at level of RPE
T N – OCT angiogram confirmed CNV
in this location
A B C I

Choroidal neovascular Choroidal neovascular


membrane membrane

T N

FIG. 7A–D. (A) Color photograph of an active macular lesion (circle) in MCP. (B) Fluorescein angiogram reveals hyperfluorescence of the lesion
resulting from staining but no definitive leakage, which was inconclusive regarding the presence of CNV. (C) Structural OCT B-scan shows an
ill-defined, moderately reflective lesion at the level of the RPE with obscuration of the outer retinal layers. (D) OCT angiogram slabs confirms the
presence of CNV in this location (circle). Corresponding OCT B-scan is shown with segmentation lines that demarcate the OCT angiography slab
(bottom). (Courtesy Patrick E. Rubsamen, MD, and Eduardo Uchiyama, MD.)

113
Vogt-Koyanagi-Harada Disease
Darin R. Goldman 18.2
Summary of the outer retina. These additional features help distinguish
VKH from central serous chorioretinopathy (see Chapter 8.1).
Vogt-Koyanagi-Harada (VKH) disease is a multiphase (prodromal,
acute, convalescent, and chronic recurrent phases) systemic
condition that manifests with vestibular-cochlear symptoms includ- Key OCT Features
ing hearing loss and tinnitus, dermatologic signs including vitiligo,
and prominent posterior segment findings including bilateral • The presence and height of subretinal fluid and choroidal
uveitis, serous retinal detachment, and choroidal inflammation. thickness on OCT correlate with disease activity and visual
In addition to fluorescein angiography and indocyanine green acuity, which can be useful to gauge treatment response.
angiography, OCT has proved useful as an imaging modality • Subretinal septae and RPE/choroidal folds are distinguishing
to aid in diagnosis of VKH and monitor response to systemic features of VKH that are well-visualized on OCT (Fig. 2).
treatment. In the acute phase, OCT reveals characteristic serous • Intraretinal fluid may be present in the outer retinal layers.
retinal detachments (subretinal fluid) and choroidal thickening • OCT features normalize with time (Fig. 5).
(Figs. 1 and 2). These findings correlate with active disease and,
in the convalescent phase, subretinal fluid resolves and choroidal REFERENCE
Fong AH, Li KK, Wong D. Choroidal evaluation using enhanced depth imaging
thickness decreases, both features that can be used to monitor spectral-domain optical coherence tomography in Vogt-Koyanagi-Harada
treatment response. Reduction of focal hyperreflectivity of the disease. Retina. 2011;31(3):502–509.
inner choroid in acute and convalescent stages of VKH may
correlate with choroidal inflammation/infiltration and account for BIBLIOGRAPHY
compromising choriocapillaris blood flow (Fong, Li & Wong 2011) O’Keefe GA, Rao NA. Vogt-Koyanagi-Harada Disease. Surv Ophthalmol.
(Figs. 3 and 4). Additional OCT features of VKH include subretinal 2017;62(1):1–25.
septae, RPE/choroidal folds, and intraretinal cysts in various layers

FIG. 1A and B. (A) VKH in


the acute stage with visible
serous retinal detachment in
the macula and surrounding
chorioretinal folds. (B) At 1 month
after treatment with high-dose
systemic prednisone there is near
complete resolution.

A B

Intraretinal FIG. 2A and B. Right (A) and


Subretinal left (B) eyes in the acute stage of
fluid in Intraretinal
fluid Vitreous
outer retinal fluid VKH show significant intraretinal
layers inflammation fluid accumulation located within
Subretinal the outer retina. Additional
Subretinal fluid characteristic OCT features of
septae VKH that are illustrated include
intraretinal and subretinal fluid,
RPE/Choroidal subretinal septae, a thickened
fold choroid, RPE/choroidal folds, and
S S
1 1 vitreous inflammation.
T N N T
Thickened choroid Thickened choroid RPE/Choroidal
A I B I fold

114
18.2
One week after systemic prednisone One week after systemic prednisone

Subretinal fluid with Subretinal fluid with

Vogt-Koyanagi-Harada Disease
hyperreflective flecks hyperreflective flecks

S S

T N

Irregular
N T
Irregular
Thickened choroid Thickened choroid RPE
I I

A RPE B

FIG. 3A and B. Right (A) and left (B) eyes 1 week after treatment with systemic prednisone. There is dramatic reduction of active disease features
on OCT. Subretinal fluid and choroidal thickness are both reduced from baseline but remain abnormal. Hyperreflective flecks are seen within the
subretinal fluid, and the RPE retains an irregular contour.

S S

N T N T

I Normalizing choroidal thickness I Normalizing choroidal thickness


A B

FIG. 4A and B. Right (A) and left (B) eyes 1 month after treatment, in the convalescent phase. Subretinal fluid has completely resolved. The RPE
still has an irregular contour that has improved. The choroidal thickness is near normal; its depth can be visualized in certain areas (estimated by red
bars).

FIG. 5. At 1 year after treatment, all OCT


features of disease have resolved. The RPE
has a regained a linear contour, and the
Normalized retinal features choroidal thickness has normalized.

Normalized choroidal thickness

115
Sympathetic Ophthalmia
Darin R. Goldman | Nora W. Muakkassa 18.3
Summary Key OCT Findings
Sympathetic ophthalmia is a rare, bilateral, granulomatous • Dalen-Fuchs nodules are hyperreflective nodular lesions at
uveitis resulting from autoantibodies targeting ocular tissues the level of the RPE.
after penetrating injury or surgery. Findings range from mild • Diffuse loss of the ELM and IS/OS/EZ are common, along
anterior uveitis to severe panuveitis. Additional clinical findings with serous retinal detachment and a thickened choroid, all
include mutton-fat keratic precipitates, vitritis, papillitis, and of which should improve after treatment.
serous retinal detachments. Dalen-Fuchs nodules are a classic • Permanent irregularities of the IS/OS/EZ and ELM may persist.
finding present in only 25% to 35% of cases. These nodules
are composed of subretinal RPE epithelioid cells. OCT findings
include focal nodular hyperreflective lesions at the level of the BIBLIOGRAPHY
Behdad B, Rahmani S, Montahaei T, et al. Enhanced depth imaging OCT
RPE, which correspond clinically with Dalen-Fuchs nodules (Fig. (EDI-OCT) findings in acute phase of sympathetic ophthalmia. Int Ophthalmol.
1). Additionally, there may be diffuse external limiting membrane 2015;35:433–439.
(ELM) and inner segment/outer segment/ellipsoid zone (IS/OS/EZ) Gupta V, Gupta A, Dogra MR, et al. Reversible retinal changes in the acute
loss. Subretinal fluid and choroidal thickening may be present stage of sympathetic ophthalmia seen on spectral domain optical coherence
tomography. Int Ophthalmol. 2011;31:105–110.
in cases with serous retinal detachment. After treatment, which Muakkassa NW, Witkin AJ. Spectral-domain optical coherence tomography
typically includes systemic immunosuppression, these findings of sympathetic ophthalmia with Dalen-Fuchs nodules. Ophthalmic Surg
often resolve. However, attenuated areas of the IS/OS/EZ and Lasers Imaging Retina. 2014;45(6):610–612.
ELM may persist (Fig. 2).

116
18.3

Loss of IS/OS

Vitreous debris

Sympathetic Ophthalmia
Nodular thickening
of RPE
Shadowing

A C

FIG. 1A–C. Sympathetic ophthalmia. (A) Color photograph shows media opacity secondary to vitritis, peripapillary whitening, and multiple subretinal,
white, creamy lesions. (B) Fluorescein angiogram shows early blockage and late staining of the peripapillary area and multiple lesions throughout the
posterior pole. (C) OCT shows shadowing secondary to vitritis, hyperreflective, nodular deposits at the level of the RPE, and loss of the IS/OS/EZ and
ELM. (From Muakkassa NW, Witkin AJ. 2014. Ophthalmic spectral-domain optical coherence tomography of sympathetic ophthalmia with Dalen-Fuchs nodules. Surg
Lasers Imaging Retina. 45 (6), 610–612. Reproduced with permission of SLACK Inc.)

117
FIG. 2A and B. At 10 months after
immunosuppressive therapy. (A) Color
fundus photograph shows improved vitritis.
Previous creamy, white, subretinal lesions
have resolved, leaving scattered areas of
chorioretinal atrophy. (B) OCT reveals resolved
Section 18: Noninfectious Uveitis

hyperreflective nodules along with a partially


restored IS/OS/EZ temporally. An area of
chorioretinal atrophy is visible nasally.

Chorioretinal atrophy
Hyperreflective nodules resolved

S
Partially restored IS/OS junction
3
N T

B I

118
SECTION 19: INFECTIOUS UVEITIS

Toxoplasmic Chorioretinitis
Darin R. Goldman 19.1
Summary Key OCT Features
Toxoplasmic chorioretinitis, caused by infection of the parasite • Full-thickness retinal hyperreflectivity is present in focal areas
Toxoplasma gondii, is the most common identifiable cause for of retinitis with distinct margins (Fig. 2).
posterior uveitis and focal retinitis. The clinical appearance is • White spots in vitreous and within retina can be seen.
that of focal yellow or white retinitis with overlying vitreous • Subretinal fluid may be present, usually only detectable via
inflammation. The active area of retinitis is typically adjacent to OCT.
a darkly pigmented chorioretinal scar, indicative of old disease. • Round, hyperreflective plaques overlying retinal blood vessels
The diagnosis of toxoplasmic chorioretinitis is usually determined may be seen throughout the fundus, which are strongly sug-
based on the clinical appearance alone. Unusual cases may be gestive of toxoplasmosis chorioretinitis.
difficult to differentiate from other causes of retinitis. In these
cases, additional diagnostic testing, such as serology or aqueous BIBLIOGRAPHY
Saito M, Barbazetto IA, Spaide RF. Intravitreal cellular infiltrate imaged as
sampling for polymerase chain reaction, may be helpful. OCT,
punctate spots by spectral-domain optical coherence tomography in eyes
although not definitive in its diagnostic specificity for toxoplasmic with posterior segment inflammatory disease. Retina. 2013;33(3):559–565.
chorioretinitis, can elucidate unique findings that support the
diagnosis (Figs. 1–4).

A B

FIG. 1A–C. (A) Diffuse toxoplasmic chorioretinitis with a large area of focal retinitis (circle). (B) Infrared image highlighting spheroid plaques located on
both arterioles and venules. (C) OCT in an area not involved with focal retinitis. Secondary features of active inflammation are present, including visible
white blood cells in the vitreous cavity (arrows) and a typical perivascular spheroid deposit in cross section (circle).

119
FIG. 2A and B. (A and B) Diffuse
retinal hyperreflectivity is present
in a focal area of active retinitis
due to toxoplasmosis (arrows).
The margins of involved and
uninvolved retina are distinct.
Section 19: Infectious Uveitis

S S
1 1
N T N T

A I
B I

FIG. 3. Peripapillary toxoplasmic chorioretinitis. There is a highly reflective,


thickened vitreous membrane due to localized inflammation. A subclinical
retinal detachment is present, but difficult to appreciate. The involved
area of focal retinitis is partially obscured by overlying inflammation, which
causes shadowing.

S
1
N T

A B

C D

FIG. 4A–D. (A) Color photograph and (B) fluorescein angiogram of typical toxoplasmic chorioretinitis showing an active lesion adjacent to a scar. (C
and D) OCT through the area of active retinitis shows full-thickness involvement with diffuse hyperreflectivity. (Courtesy Lana Rifkin, MD.)
120
Acute Syphilitic Posterior Placoid Chorioretinitis
Darin R. Goldman 19.2
Summary Key OCT Features
Syphilis is a rare cause of infectious uveitis caused by the • In the acute phase of ASPPC, there may be shallow subretinal
spirochete Treponema pallidum that occurs in the secondary or fluid in the fovea, which is transient, and only detectable by
tertiary stages of disease. Ocular involvement typically manifests OCT.
as posterior uveitis with chorioretinitis. A distinctive form of this • The most distinctive feature of ASPPC is patchy disruption of
condition is termed acute syphilitic posterior placoid chorioretinitis the inner segment/outer statement/ellipsoid zone (IS/OS/EZ)
(ASPPC), which exhibits a characteristic clinical appearance. with intermixed hyperreflective nodular lesions, which may
Single or multiple, round, yellowish lesions within the macula, represent thickened RPE (Fig. 1).
which involve the outer retina and retinal pigment epithelium • The external limiting membrane is typically disrupted focally
(RPE), are present in one or both eyes. Although the clinical over the nodular lesions and punctate hyperreflectivity in the
and angiographic appearance of ASPPC can be fairly distinct, choroid may be present.
OCT findings can increase diagnostic certainty in the appropriate • With appropriate and timely treatment, abnormal OCT findings
clinical context. Distinct OCT features are present in the outer parallel clinical findings and show dramatic, complete resolution
retinal and RPE layers, which typically resolve completely on within 1 to 2 months in the majority of cases (Fig. 2).
appropriate antibiotic treatment (Figs. 1–3).
BIBLIOGRAPHY
Burkholder BM, Leung TG, Ostheimer TA, et al. Spectral domain optical coher-
ence tomography findings in acute syphilitic posterior placoid chorioretinitis.
J Ophthalmic Inflamm Infect. 2014;4(1):2.
Eandi CM, Neri P, Adelman RA, et al. Acute syphilitic posterior placoid
chorioretinitis: report of a case series and comprehensive review of the
literature. Retina. 2012;32(9):1915–1941.
Pichi F, Ciardella AP, Cunningham ET Jr, et al. Spectral domain optical
coherence tomography findings in patients with acute syphilitic posterior
placoid chorioretinopathy. Retina. 2014;34(2):373–384.

121
Section 19: Infectious Uveitis

A A

B B

Disruption of IS/OS/ Nodular


ellipsoid zone hyperreflective
S lesions S
4 3
N N
T T
Acute syphilitic posterior placoid
C I chorioretinitis (ASPPC) C I

FIG. 1A–C. (A and B) Color and red-free photographs of typical ASPPC FIG. 2A–C. (A and B) Color and red-free photographs 1 month after
at presentation with visual acuity of 20/100. The borders of the lesion intravenous ceftriaxone therapy with return of visual acuity to 20/20.
are illustrated with arrowheads. (C) OCT reveals characteristic patchy (C) OCT shows normalization of the IS/OS/EZ and resolution of the
loss of the IS/OS/EZ along with hyperreflective nodular RPE lesions. hyperreflective nodular RPE lesions. (Courtesy of Lana Rifkin, MD.)
(Courtesy Lana Rifkin, MD.)

122
19.2

Acute Syphilitic Posterior Placoid Chorioretinitis


B
A

FIG. 3A–C. (A and B) In severe cases of ASPPC in which treatment is


delayed, significant subretinal fibrosis can develop. In this example, a
S Subretinal secondary choroidal neovascular membrane developed (circle) on the
3 hyperreflective Subretinal
temporal edge of old scarring many years following the acute episode.
T N
material fibrosis
(C) OCT reveals two distinct subretinal pathologic processes: subretinal
C I hyperreflective material resulting from choroidal neovascularization and
subretinal fibrosis secondary to scarring.

123
Tuberculosis
Darin R. Goldman 19.3
Summary Key OCT Features
Mycobacterium tuberculosis is a common cause of infectious • Choroidal infiltration results in homogeneous, hyporeflec-
uveitis in certain tropical countries. Choroidal granuloma, tive material present beneath the RPE in a dome-shaped
chorioretinitis, and various forms of uveitis all can be ocular configuration.
manifestations of tuberculosis. Involvement of the choroid can • Overlying subretinal fluid may be present acutely.
be localized or multifocal, mimicking various noninfectious ocular • After systemic therapy, choroidal elevation and secondary
conditions such as serpiginous choroiditis. Secondary abnormali- features of disease resolve, leaving varying degrees of RPE
ties may develop within the subretinal space. OCT is particularly destruction and deposition of hyperreflective subretinal material.
useful in identifying both subretinal and choroidal manifestations
of tuberculosis (Figs. 1 and 2).

124
19.3

Tuberculosis
A B

D E

F G

FIG. 1A–G. (A) Color photograph of choroidal granuloma secondary to tuberculosis (circle) before treatment. (B and C) OCT reveals characteristic,
dome-shaped, choroidal infiltration. Associated subretinal fluid and subretinal material with mixed reflectivity are present. (D) Color photograph 1
month after systemic therapy. (E) OCT reveals resolution of subretinal fluid and flattening of the choroidal infiltration. (F) Color photograph 3 months
after treatment. (G) OCT shows complete resolution of choroidal infiltration, with restoration of a flat RPE contour. Some RPE abnormalities remain
temporally. (Courtesy of Alay S. Banker, MD.)

125
Section 19: Infectious Uveitis

A B

C D

FIG. 2A–D. (A) Color photograph of choroidal granuloma secondary to tuberculosis. (B) OCT shows significant choroidal infiltration, mild subretinal
fluid, and hyperreflective subretinal material. (C) At 4 months after systemic therapy, the choroidal granuloma has become atrophic. (D) OCT shows
resolution of choroidal infiltration with flattening of the RPE. Significant hyperreflective subretinal material remains. (Courtesy Alay S. Banker, MD.)

126
Posterior Scleritis
Darin R. Goldman 19.4
Summary posterior scleritis, which may not be obvious by other means.
OCT is very sensitive in detecting both chorioretinal folds and
The sclera is an opaque structure lining the exterior of the globe serous retinal detachment, both common features of posterior
that provides structural support and is integral to the function of scleritis (Figs. 1 and 2). Additionally, OCT provides a particularly
various ocular structures. Posterior scleritis is the rarest subtype useful manner of monitoring therapeutic response to treatment.
of scleritis, occurring in less than 5% of all cases, defined by
inflammation of the posterior sclera. The underlying causes of
posterior scleritis are myriad and include both infectious and Key OCT Features
autoimmune processes, although no cause is identified frequently.
The condition may be bilateral, usually with an indolent onset.
• Undulations of the retinal pigment epithelium (RPE) in an
irregular, wavelike pattern indicate chorioretinal folds, which
Pain is a salient feature and visual acuity may be normal or are common in posterior scleritis.
reduced. The diagnosis of posterior scleritis is difficult to establish
because of its low incidence and ability to mimic other pathologic
• Subretinal fluid, often overlying chorioretinal folds, indicates
serous retinal detachment. This fluid may be subclinical and,
states. Fluorescein angiography, ultrasonography, and orbital thus, visible only on OCT.
radiologic imaging are useful adjunctive diagnostic tests. OCT
can provide additional supportive evidence to corroborate the BIBLIOGRAPHY
diagnosis while definitively identifying secondary features of Benson WE. Posterior scleritis. Surv Ophthalmol. 1988;32(5):297–316.

S Chorioretinal
1 folds
T N

I
A B

FIG. 1A-B. (A) Color photograph montage in posterior scleritis showing chorioretinal folds within the macula and temporal periphery. There is a
sizable choroidal detachment temporally. (B) OCT on the edge of the superior macula illustrates the typical appearance of chorioretinal folds in
posterior scleritis. The RPE has an irregular, wavelike configuration.

127
Section 19: Infectious Uveitis

A B

Chorioretinal Subretinal
1
S
fold fluid
T N

C I

T N

S
S 1
T N

I
D E I

FIG. 2A–E. (A) Color photograph in posterior scleritis does not illustrate pathologic findings because they are not readily visible clinically. (B)
Fluorescein angiography illustrates chorioretinal secondary to alternating stretching and squeezing of the RPE with resultant bands of relative
hyper reflectivity and hyporeflectivity. (C) Structural OCT identifies subclinical subretinal fluid and mild chorioretinal folds. (D) Typical appearance of
chorioretinal folds on OCT thickness map is shown (top). There are bands of varying thickness that result from the undulation of the underlying RPE
and subsequent wave-like alteration in the automated inner limiting membrane to RPE thickness that is computed to generate the thickness map. (E)
Treatment response to systemic steroids is illustrated with near resolution of chorioretinal folds and subretinal fluid (corresponding to Fig. 2C).
128
Candida Chorioretinitis
Darin R. Goldman 19.5
Summary Key OCT Features
Fungal chorioretinitis is a rare ocular disorder that may be • Dome-shaped lesion with highly reflective surface and a vari-
acquired via hematogenous dissemination in the setting of able, but significant, degree of underlying shadowing (Fig. 1B).
fungemia (endogenous) or by direct inoculation in the setting of • Extensive infection results in diffuse hyperreflective material
postoperative infection (exogenous). The most common etiologic on the surface of the retina, which may be detectable only
pathogen in cases of fungal chorioretinitis are Candida species, on OCT (Fig. 2B).
specifically Candida albicans. Although patients with endogenous • Poor signal strength is common because of media opacity
fungal chorioretinitis are often hospitalized where OCT evaluation from overlying vitreous involvement.
is impractical, ambulatory patients also can be affected. Thus • Lesions resolve following appropriate antifungal therapy, leaving
fungal chorioretinitis may be encountered in the outpatient setting, full-thickness retinal disruption and choroidal atrophy (Fig.
where OCT is readily available. In this setting, diagnosis can be 3D,E).
particularly challenging because of limitations in obtaining an
adequate specimen and length of time required for confirmatory BIBLIOGRAPHY
culture results. OCT is particularly useful to identify characteristic Adam MK, Rahimy E. Enhanced depth imaging optical coherence tomography of
endogenous fungal chorioretinitis. JAMA Ophthalmol. 2015;133(11):e151931.
features of fungal chorioretinitis, which can aid in formulating an Lavine JA, Mititelu M. Multimodal imaging of refractory Candida chorioretinitis
already challenging diagnosis (Figs. 1–3). progressing to endogenous endophthalmitis. J Ophthalmic Inflamm Infect.
2015;5(1):54.

T N

S
5 S

T N

I
B I
C

FIG. 1A–C. (A) Color photograph of a typical chorioretinal lesion resulting from Candida infection. Note that there is also background diabetic
retinopathy with exudate. (B) OCT through the macular lesion shows full-thickness retinal involvement. (C) OCT thickness map highlights the focal
lesion.

129
FIG. 2A–C. (A) Anterior segment photograph of postoperative
endophthalmitis caused by Candida parapsilosis shows a visible nidus
of infected material within the capsular bag (circle). (B) OCT at the
edge of the macula shows a sheet of hyperreflective material lining the
surface of the retina, which is a feature of extensive infection and was
not visible clinically. (C) Grocott-Gomori methenamine silver (GMS) stain
Section 19: Infectious Uveitis

of the lens capsule, which was removed at the time of vitrectomy, shows
extensive Candida parapsilosis organisms (dark material).

Hyperreflective material on surface of retina

S
5
N T

B I

130
19.5

Candida Chorioretinitis
A D

Hyperreflective material
on surface of retina

S S
1 3
N T
Full-thickness N T

hyperreflective lesion
I I
B E

Hyperreflective material on surface of retina

FIG. 3A–E. (A) Color photograph in a patient with bilateral endogenous


endophthalmitis as a result of Candida albicans. (B) OCT shows
extensive hyperreflective material on the retinal surface and a full-
S thickness hyperreflective retinal lesion. (C) Extensive shadowing is
5 present because of overlying vitreous involvement, although preretinal
N T hyperreflective lesions are still visible. (D) Chorioretinitis is resolved 6
months following antifungal treatment, leaving extensive chorioretinal
I Extensive shadowing atrophy. (E) Corresponding OCT reveals chorioretinal atrophy with
C varying degrees of retinal layer disorganization. (Courtesy of Larry S.
Halperin, MD.)

131
Acute Retinal Necrosis Syndrome
Darin R. Goldman 19.6
Summary Key OCT Features
Acute retinal necrosis syndrome is a severe, vision-threatening • In the acute phase of acute retinal necrosis syndrome, there
viral infection of the retina, most frequently caused by viruses is full-thickness, diffuse hyperreflectivity of the retina in a
in the herpetic family. The retinal necrosis typically begins in the well-defined area, corresponding to clinically evident retinal
peripheral retina in a sporadic array of opaque, white patches necrosis.
that are well circumscribed. These patches rapidly coalesce in • Subclinical overlying vitreous inflammation, subretinal fluid,
a circumferential trajectory, becoming confluent over time and or choroidal thickening may be visible on OCT.
spreading posteriorly. Associated retinal occlusive vasculitis and • Over time, retinal thinning and loss of tissue often develop in
uveitis are common features. Choroiditis and subclinical serous areas of prior retinal necrosis, despite antiviral therapy.
retinal detachment also may be present, which can be identified
using OCT. In the acute setting, clinically evident, full-thickness BIBLIOGRAPHY
Duker JS, Blumenkranz MS. Diagnosis and management of the acute retinal
retinal whitening appears on OCT as diffuse, homogeneous
necrosis (ARN) syndrome. Surv Ophthalmol. 1991;35:327–343.
hyperreflectivity with variable thickening of the retina (Fig. 1). Kurup SP, Khan S, Gill MK. Spectral domain optical coherence tomog-
The inner retina may become involved initially followed by rapid raphy in the evaluation and management of infectious retinitis. Retina.
involvement of the entire thickness of the retina. Within days to 2014;34(11):2233–2241.
weeks, the retinal layers become disorganized and generalized Murata K, Yamada W, Nishida T, et al. Sequential Optical Coherence Tomog-
raphy Images of Early Macular Necrosis Caused by Acute Retinal Necrosis
atrophy appears. The remaining retinal tissue maintains a diffusely in Non-Human Immunodeficiency Virus Patients. Retina. 2016;36(7):e55–e57.
hyperreflective signal. Over time, complete loss of retinal layers Ohtake-Matsumoto A, Keino H, Koto T, et al. Spectral domain and swept
may develop, leaving hyporeflective cavities. Secondary retinal source optical coherence tomography findings in acute retinal necrosis.
detachment is a common late complication. Although OCT findings Graefes Arch Clin Exp Ophthalmol. 2015;253(11):2049–2051.
in acute retinal necrosis syndrome are not specific, they serve
as a useful adjunct in the diagnostic evaluation.

132
19.6

Acute Retinal Necrosis Syndrome


tin tic
re cro
a
Ne
S

tin al
5

re rm
a
T N

No
I
A B

Vitreous
inflammation

Serous retinal
detachment
S S
4 2
T N
Normal T N
Acute retinal necrosis syndrome
I I following antiviral therapy
C Acute retinal necrosis syndrome D

FIG. 1A–D. (A) Wide-angle image of acute retinal necrosis syndrome involving the nasal periphery (white arrows). A satellite lesion is also present
above the optic nerve (yellow arrow). (B) The nasal periphery is imaged at the transition from normal to necrotic retina. The necrotic retina is diffusely
hyperreflective with mild thickening and overlying vitreous inflammation. (C) The satellite lesion above the optic nerve is imaged in cross section. There
is diffuse hyperreflectivity throughout the involved region of the retina with significant thickening. Associated subclinical serous retinal detachment,
choroidal enlargement, and vitreous inflammation are also present. (D) Same section as in part C 2 weeks after antiviral therapy shows less retinal
hyperreflectivity, vitreous inflammation, and choroidal thickening along with resolution of subretinal fluid. (Courtesy Eduardo Uchiyama, MD.)

133
SECTION 20: CHOROIDAL TUMORS

Choroidal Nevus
Jay S. Duker 20.1
Summary appearance of choroidal nevi. OCT provides adjunctive information
and may help confirm the diagnosis in uncertain cases.
Choroidal nevi are common, benign, typically flat, variably pig-
mented lesions of the choroid. Nevi do not usually grow but can
become increasingly pigmented, acquire drusen on the surface, Key OCT Features
and be associated with subretinal fluid, which is often detectable
only on OCT. Nevi can transform into melanoma, although this is
• Flat to minimal thickening is seen.
quite rare. Clinically, choroidal nevi appear as darkly pigmented,
• Well-defined blocking of signal occurs in the outer choroid.
round lesions located beneath the retina (Fig. 1). Typical size
• Overlying choriocapillaris is compressed but may be retina
(Fig. 2).
ranges from 1 to 4 disc diameters, although larger nevi may
occur. Choroidal nevi may be located throughout the fundus.
• Overlying retina may have cystic changes or localized subretinal
fluid.
Clinical photographs are the best method of documenting the

FIG. 1. Fundus photograph of a 3- × 2-mm


nonsuspicious choroidal nevus

FIG. 2. SD OCT line scan through a 3- × 2-mm


nonsuspicious choroidal nevus. Note intact overlying
retina and no subretinal fluid. There is compression of
the choriocapillaris. The nevus itself is located in the
Compressed outer choroid and blocks penetration of the OCT signal.
choriocapillaris

Choroidal nevus

134
Choroidal Melanoma
Jay S. Duker 20.2
Summary Key OCT Features
Choroidal melanoma is the most common primary malignancy of • Elevated choroidal mass with extensive blocking of signal
the posterior segment of the eye in adults. They are typically dark (Fig. 1).
brown to tan in color and may be completely amelanotic. They vary • Obscuration of the normal choroidal vascular pattern in the
in size, although the larger and more elevated, the more likely it is area of the tumor (Fig. 2).
to be melanoma in comparison to a nevus. They occur unilaterally • Overlying subretinal fluid is present with shaggy photoreceptors
and unifocally in nearly every instance. When they metastasize, (Fig. 1).
they typically go to the liver via hematogenous spread.

FIG. 1. OCT showing elevated transchoroidal mass,


subretinal fluid, and “shaggy” photoreceptors.

N T
I

FIG. 2. Color figure corresponding to OCT in Fig. 1


showing a small choroidal melanoma measuring 7 mm in
diameter and 2 mm in thickness.

135
Solitary Choroidal Hemangioma
Jay S. Duker 20.3
Summary photocoagulation, photodynamic therapy, and low-dose radiation
can be successful in eliminating the fluid leakage.
Solitary choroidal hemangioma is a benign lesion of the choroid
that typically presents in the fourth or fifth decade of life. These
lesions become apparent when leakage into the retina or sub- Key OCT Findings
retinal space results in symptoms or are identified during routine
ophthalmologic examination. The lesions are typically round or
• Localized choroidal mass with large inherent vasculature.
oval in shape, red in coloration and are between 3 and 4 mm
• Subretinal fluid and cystic retinal degeneration may be present
over tumor.
in thickness (Fig. 1). Overlying cystic retinal degeneration, sub-
retinal fluid, and retinal pigment epithelial changes are common.
• Tumor mass exhibits a hyporeflective internal signal
(Fig. 2).
Treatment is indicated only if central acuity is affected. Laser

FIG. 1. Color fundus photograph showing


subtle reddish mass superior to disc.

FIG. 2. OCT of solitary choroidal hemangioma,


which is located between the two arrows. Not
the generalized hyporeflectivity of the lesion.

136
SECTION 21: RETINAL TUMORS

Retinal Capillary Hemangioma


Jay S. Duker 21.1
Summary Key OCT Features
Retinal capillary hemangiomas are benign lesions that can develop • Secondary macular edema, hard exudate, and subretinal fluid
either as an isolated phenomenon or in association with Von- due to leaking peripheral retinal capillary hemangiomas can
Hippel Lindau syndrome. The lesions are round or oval and be identified (Figs. 1–3).
typically have enlarged, dilated feeding and draining retinal vessels. • Vitreous changes over capillary hemangiomas are common.
Early in their course, hemangiomas rarely cause visual issues, • The lesions are elevated and well circumscribed but show
but over time they can grow and cause both intraretinal and little internal detail.
subretinal fluid accumulation with hard exudate, both locally in
the area of the tumor and at a distance in the macula. Over time,
vitreous traction can develop with resultant retinal detachment.

Retinal
capillary
hemangioma
S
Intraretinal fluid 1
N T
and exudate Subretinal fluid
I

FIG. 1. Macular OCT in a patient with a peripheral retinal capillary FIG. 2. Peripheral OCT in the same patient as in Fig. 1 through the
hemangioma. Intraretinal fluid, subretinal fluid, and exudate are all center of the retinal capillary hemangioma. The surface of the lesion
common features that may be present. is hyperreflective, and the interface with the vitreous exhibits sites
of adhesion. Internal details are not visualized because of significant
shadowing of the signal.

A B

FIG. 3. (A) Color photographs of the macula (corresponding to Fig. 1) and (B) the peripheral lesion (corresponding to Fig. 2).

137
SECTION 22: RETINAL PIGMENT EPITHELIUM TUMORS

Simple Hamartoma of the RPE


Jay S. Duker 22.1
Summary Key OCT Features
Congenital simple hamartoma of the retinal pigment epithe- • Preretinal location with dense hyperreflectivity that results in
lium (RPE) is an uncommon, benign tumor of the RPE that is shadowing and obscuration of both the underlying retina and
present on the retinal surface. The lesions are thought to be choriocapillaris (Figs. 1 and 2).
congenital, although they typically are identified during routine • Extremely sharply demarcated borders.
ophthalmoscopy in adulthood because significant visual symp-
toms are uncommon. The location on the retinal surface may BIBLIOGRAPHY
Barnes AC, Goldman DR, Laver NV, et al. Congenital simple hamartoma of
be explained by aberrant migration during embryogenesis. The
the retinal pigment epithelium: clinical, optical coherence tomography, and
clinical appearance is suggestive, and the OCT appearance is histopathological correlation. Eye (Lond). 2014;28(6):765–766.
pathognomonic of the diagnosis. The lesions are typically jet
black, well-demarcated, and round.

FIG. 1. OCT of typical simple hamartoma of the RPE. The


lesion is located between the arrowheads and exhibits
extreme hyperreflectivity. The extent of the underlying
shadowing is shown between the arrows.

S
3
N T

A B

FIG. 2. Color (A) and red-free (B) photographs (corresponding to Fig. 1) illustrate the jet-black color and typical clinical appearance of congenital
hamartoma of the RPE.

138
Combined Hamartoma of the Retina and RPE
Jay S. Duker 22.2
Summary Key OCT Features
Combined hamartoma of the retina and RPE is a rare, benign • Full-thickness retinal and RPE disorganization is present in
hamartomatous growth that is important to distinguish from other most cases (Fig. 1).
entities that may appear similar but are actually malignant. The • Overlying fibrosis is present, often with a sawtooth pattern
clinical appearance is focal disorganization of the retina with (Figs. 1 and 2).
overlying fibrosis. The involved region includes the full thickness • Cystoid macular edema, epiretinal membrane, and tractional
of the retina and RPE. The central macula can be disrupted by retinal detachment may be present as secondary features
secondary vitreoretinal interface forces or by the lesion itself, (Fig. 1).
resulting in visual impairment. OCT is extremely useful in confirm-
ing the diagnosis and examining the affected tissues. In particular, BIBLIOGRAPHY
Xue K, Mellington F, Gout I, et al. Combined hamartoma of the retina and retinal
there is an absence of involvement of the underlying choroid.
pigment epithelium. BMJ Case Rep. 2012;doi:10.1136/bcr-2012-006944.

FIG. 1. OCT appearance of a typical combined


Sawtooth appearance of retinal surface hamartoma of the retina and RPE.
with epiretinal membrane

Cystoid macular
Disorganization of
edema
all retinal layers
and RPE

FIG. 2. Corresponding color photograph to Fig. 1.

139
SECTION 23: METASTATIC CHOROIDAL TUMORS

Choroidal Metastases
Jay S. Duker 23.1
Summary Key OCT Features
Choroidal metastases are rare but represent the most common • Dense choroidal mass with little to no visible choroidal vas-
malignancy of the posterior segment in adults. The most common culature (Figs. 1 and 2).
sites of primary lesions are the breast and lung. Metastases are • Overlying subretinal fluid is very common.
often multiple and bilateral. Treatment in the form of radiation is • Retinal pigment epithelium tends to assume a “wave-like”
often successful at halting growth of the lesions, but the systemic configuration overlying the metastases.
prognosis in most cases is poor.

140
FIG. 1. OCT of choroidal 23.1
metastasis exhibits characteristic
infiltration of the choriocapillaris.

Choroidal Metastases
Subretinal fluid

Choroidal Undulating contour


metastases of RPE

FIG. 2. Corresponding color photograph


shows the amelanotic choroidal mass just
temporal to the optic nerve.

141
SECTION 24: MECHANICAL TRAUMA

Valsalva Retinopathy
Jay S. Duker 24.1
Summary Key OCT Features
Valsalva retinopathy is caused by the rupture of superficial capillar- • Vertically oriented OCT scanning patterns are most useful.
ies within the inner retina. Inciting events include anything in which • On vertical OCT, two distinct areas are present within the
forced exhalation against a closed glottis occurs, which results sub-ILM space (Fig. 1):
in a sudden increase in intraocular venous pressure. Resultant 1. Superior hyporeflective cavity (serous component)
hemorrhage pools within the sub-inner limiting membrane (ILM) 2. Inferior hyperreflective cavity (hemorrhagic component)
space, typically overlying the macula. The acute hemorrhage • Comparing registered vertical OCT scans or volume cube
becomes layered with time because of gravitational dependence. scans over time can definitively determine the clinical course
The clinical appearance is often very characteristic. Over time, (Figs. 1 and 2).
most cases resolve spontaneously. OCT is useful in identify-
ing the location of the hemorrhage within the sub-ILM space, BIBLIOGRAPHY
Goldman DR, Baumal CR. Natural history of Valsalva retinopathy in an
which confirms the diagnosis in the appropriate clinical context.
adolescent. J Pediatr Ophthalmol Strabismus. 2014;51(2):128.
Additionally, OCT provides a precise modality for monitoring the Szelog JT, Lally DR, Heier JS. Natural history of Valsalva-induced subhyaloid
hemorrhage size and volume. It can be difficult to determine hemorrhage. JAMA Ophthalmol. 2015;133(2):e143268.
changes in hemorrhage size based on the clinical appearance
alone; therefore OCT can prove quite useful in Valsalva retinopathy.

142
24.1

Valsalva Retinopathy
A C E

T N T N T N

S S S

B I
D I
F I

FIG. 1. A 33-year-old healthy woman with Valsalva retinopathy secondary to vigorous vomiting related to cholecystitis. (A and B) At 1 week after
symptom onset, there is layered hemorrhage in the sub-ILM space. Visual acuity measured 20/200. (C and D) At 2 weeks after presentation, visual
acuity measured 20/200 and the vertical height of the hemorrhage clinically appears to have increased compared to that at presentation. However,
the OCT confirms that the total hemorrhage volume has actually contracted significantly. (E and F) At 1 month after presentation, visual acuity
measured 20/100, with continued contracture of the hemorrhage.
Continued

143
Section 24: Mechanical Trauma

G I

T N T N

S S

H I
J I

FIG. 1, cont’d. (G and H) At 2 months after presentation, visual acuity improved to 20/25 with clearance of the hemorrhage from the central fovea. (I
and J) At 4 months after presentation, visual acuity returned to 20/20, with complete resolution of hemorrhage.

S S
7 7
T N T N

A I
B I

FIG. 2. Comparison of sub-ILM hemorrhage in Valsalva retinopathy as seen on vertically oriented OCT scans at baseline (A) and 1 month after
presentation (B).

144
SECTION 25: PHOTIC MACULOPATHY

Laser Maculopathy
Jay S. Duker 25.1
Summary Key OCT Features
High-powered lasers have become common in both the com- • In acute setting, there is localized outer > inner retinal involve-
mercial and recreational setting. This provides an opportunity for ment, with hyperreflectivity involving the inner segment/outer
both accidental and purposeful exposure of the macula, which has segment/ellipsoid zone (IS/OS/EZ) and RPE (Figs. 1 and 2).
increased in frequency during the 21st century. The central macula • In the subacute or chronic setting, patchy RPE disruption and
generally receives the majority of the injury because of direct clumping are present (Fig. 3).
visualization of the laser beam. Clinically, the acute lesion appears • IS/OS/EZ normalization varies depending on severity of initial
yellow and mottled which fade quickly over weeks to months. injury and correlates with visual recovery.
The later stages of injury are nonspecific, with varying amounts
of retinal pigment epithelium (RPE) pigmentary disturbances BIBLIOGRAPHY
Wyrsch S, Baenninger PB, Schmid MK. Retinal injuries from a handheld laser
and retinal atrophy. The extent of retinal injury depends on the
pointer. N Engl J Med. 2010;363(11):1089–1091.
burden of exposure, with mild injuries affecting only the outer
retina; more severe injuries can affect the full retinal thickness.

S Near full-thickness
1 hyperreflectivity
T N
LASER maculopathy at baseline
I

FIG. 1. OCT of fairly severe acute laser maculopathy. FIG. 2. Corresponding color photograph to Fig. 1.

FIG. 3. OCT 6 weeks after the original injury.

Restoration of normal inner


retinal reflectivity

Persistent RPE and IS/OS/EZ


S
disruption
1
T N
LASER maculopathy 6 weeks after injury
I

145
Solar Maculopathy
Jay S. Duker 25.2
Summary Key OCT Features
Accidental or purposeful prolonged exposure to intense light • The inner retina is normal.
sources such as the sun or a welding arc can result in photo- • Focal disruption or loss of the retinal pigment epithelial (RPE)
chemical injury to the macula. The injury is typically bilateral, and inner segment/outer segment/ellipsoid zone (IS/IO/EZ),
symmetric, and located within the fovea. Loss of vision occurs with sharply demarcated borders of normal retina on the edges
as a central scotoma that correlates with the severity of the (Figs. 1, 2 and 3).
exposure. OCT findings can be diagnostic in the appropriate • OCT findings are generally present long-term after the initial
clinical context. Focal outer retinal “holes” in the central fovea photic exposure.
are the key finding, which can occasionally be seen in other
conditions. BIBLIOGRAPHY
Chen RW, Gorczynska I, Srinivasan VJ, et al. High-speed ultrahigh resolution
optical coherence tomography findings in chronic solar retinopathy. Retin
Cases Brief Rep. 2008;2(2):103–105.
Comander J, Gardiner M, Loewenstein J. High-resolution optical coherence
tomography findings in solar maculopathy and the differential diagnosis of
outer retinal holes. Am J Ophthalmol. 2011;152(3):413–419.

Rectangular disruption
of RPE and IS/OS/EZ

A B

FIG. 1. (A) OCT of solar maculopathy. (B) Corresponding fluorescein angiogram shows pinpoint area of hyperfluorescence.

146
25.2

Solar Maculopathy
Intact ELM Normal inner
retina

S
1
T N
Rectangular loss of
I RPE and IS/OS/EZ
A B

FIG. 2. Typical appearance of solar maculopathy. (A) OCT.


C (B) Fundus photograph. (C) Fluorescein angiogram. ELM, external
limiting membrane.

FIG. 3. Fellow eye of Fig. 2 shows similar


findings but are somewhat milder.

S
1
N T

147
SECTION 26: RETINAL DYSTROPHIES

Retinitis Pigmentosa
Shilpa Desai | A. Yasin Alibhai 26.1
Summary Key Features
Retinitis pigmentosa (RP) refers to a heterogeneous group of • Symptoms include nyctalopia, constricted visual field, and in
inherited disorders that are characterized by loss of retinal cell some cases profound vision loss.
function, preferentially in the peripheral retina. RP can have varying • Waxy pallor of the optic nerve, attenuation of the retinal vessels,
severity, age of onset, mode of inheritance, and systemic associa- and bone spicules in the peripheral retina are common clinical
tions. RP may be inherited in an autosomal dominant, autosomal findings.
recessive, or X-linked recessive fashion. The X-linked form of the • OCT findings in early/milder disease include relative preserva-
disease is typically the most severe. The disease is often secondary tion of the central retina and RPE with loss of the outer retina
to mutations in the rhodopsin gene, though some forms have been and RPE adjacent to the fovea.
linked to mutations in the RDS gene (Anasagasti et al., 2012). • OCT findings in advanced/severe disease include marked
Generally, RP is characterized by a slowly progressive loss of attenuation of all retinal layers, particularly the outer retina
night vision (nyctalopia) along with contraction of the visual field. and photoreceptors.
In later stages of the disease central acuity is affected, which may • Associated cystoid macular edema may be identified on OCT.
cause profound vision loss. Typical fundus abnormalities include
waxy pallor of the optic nerve, a tapetal-like reflex resulting from BIBLIOGRAPHY
Anasagasti A, Irigoyen C, Barandika O, et al. Current mutation discovery
changes in the retinal pigment epithelium (RPE), narrowing of
approaches in retinitis pigmentosa. Vision Res. 2012;75:117–129.
the peripheral retinal vasculature, and bone-spicule changes in Farrar GJ, Millington-Ward S, Chadderton N, et al. Gene-based therapies for
the mid-peripheral retina (Fig. 1). Definitive diagnosis requires dominantly inherited retinopathies. Gene Ther. 2012;19(2):137–144.
electrophysiologic testing. Computed tomography is useful to aid in Wolfensberger TJ. The role of carbonic anhydrase inhibitors in the management
the initial diagnosis and detecting associated macular abnormalities of macular edema. Doc Ophthalmol. 1999;97(3-4):387–397.
such as cystoid macular edema (Fig. 2). Treatment of RP is limited
at this time, although retinal prosthetic implants are available for
extremely severe cases (Farrar et al., 2012).

FIG. 1. Color fundus photograph of a patient with typical RP. There is peripheral bone
spicule deposition encroaching into the macula, optic nerve pallor, and prominent
vascular attenuation. The central retina and RPE are preserved (“central island”).

FIG. 2. OCT B-scan


corresponding to Fig. 1. There is
significant thinning of the outer
retinal layers and dropout of the
RPE involving the edges of the
macula. However, the central
fovea is spared with normal
retinal architecture.

148
Stargardt Disease
Jay S. Duker 26.2
Summary OCT provides adjunctive information regarding the location of
the pathologic lesions with respect to the RPE, which can help
Stargardt disease is the most common inheritable macular confirm the diagnosis. In cases of known Stargardt disease,
dystrophy, associated with mutations in the ABCA4 gene, which OCT is critical to detect and monitor macular atrophy that can
accounts for the majority of macular degeneration in young people. develop parafoveally or subfoveally.
The age of onset and disease severity vary, but, generally, the
longer the duration of disease, the more severe it is. The fundus
appearance is that of fleck-like deposits of yellowish material at
Key OCT Features
the level of the RPE in a pattern centered on the macula, although • There is disruption of the RPE and overlying outer retina and
the entire posterior pole may be involved (Fig. 1). Associated inner segment/outer segment/ellipsoid zone (IS/OS/EZ), which
central macular atrophy accounts for vision loss that correlates correlates with disease severity.
with its proximity to the foveal center. Fluorescein angiogra- • Associated retinal atrophy begins in a parafoveal location,
phy displays a characteristic dark choroid in the majority of spreading to involve the fovea with longer disease duration
cases, and fundus autofluorescence reveals a distinctive pattern. (initially spares central fovea) (Figs. 2–4).

A B
FIG. 1. (A) Color photograph of typical early Stargardt disease showing yellowish pisciform flecks. (B) Corresponding fluorescein angiograph shows
staining of pisciform lesions and a characteristic dark choroid.

149
Patchy loss of outer retina and RPE
Section 26: Retinal Dystrophies

S S
3
1
N T
N T

A I Moderate Stargardt disease


I

FIG. 3. Moderate Stargardt disease with patchy outer retinal and RPE
atrophy. The central fovea is spared.

N T

B I

FIG. 2. (A) Structural OCT in mild Stargardt disease shows patchy


disruption of the IS/OS/EZ band in a parafoveal location. The central
fovea is relatively intact. (B) Corresponding OCT thickness map reveals
150 mild parafoveal thinning secondary to outer retinal loss.
FIG. 4A and B. (A) Structural OCT in severe/advanced Stargardt disease 26.2
shows generalized outer retinal and RPE atrophy involving the entire macula.
(B) Corresponding thickness map best illustrates the global macular atrophy.

Stargardt Disease
y
troph
and RPE a
retinal
outer
alized
Gener
S
1
N T

Severe Stargardt disease


A I

N T

B I

151
Best Disease
Jay S. Duker 26.3
Summary Key OCT Features
Best disease is caused by a mutation in the BEST1 gene and • Vitelliform stage exhibits subretinal material that is a mix of
features multiple clinical phenotypes during different stages of both hyperreflective and hyporeflective material (Figs. 1–3).
disease. These include the vitelliform, pseudohypopyon, scrambled • Pseudohypopyon stage exhibits layering of hyporeflective
egg, and atrophic stages. Typically, there is a singular central material superiorly and hyperreflective material inferiorly, which
macular lesion present bilaterally. However, multifocal and is best identified using a vertical OCT orientation (Figs. 4–6).
asymmetric disease phenotypes are possible. OCT can identify • Scrambled egg stage exhibits a mix of RPE atrophy, pigment
representative submacular findings in Best disease, including clumping, and subretinal fibrosis.
differences among the various stages. • Atrophic stage exhibits generalized atrophy.

FIG. 1. Vitelliform stage of Best disease.


Subretinal material is a mixture of
hyperreflective and hyporeflective material.

FIG. 2. Color photograph corresponding to Fig. 1. FIG. 3. Fundus autofluorescence corresponding to Fig. 1.

152
FIG. 4. Pseudohypopyon stage of Best disease, horizontal imaging plane 26.3
through the superior component, which is hyporeflective.

Best Disease
S
N T
I

FIG. 5. Color photograph corresponding to Fig. 4.

FIG. 6. Fundus autofluorescence corresponding to Fig. 4.

153
Cone Dystrophy
Shilpa Desai, A. Yasin Alibhai 26.4
Summary Key Features
Cone dystrophies encompass a number of heritable dystrophies • Cone dystrophy refers to a wide variety of dystrophies char-
affecting the cone system. As with other heritable conditions, acterized by dysfunction in the cone system.
the age of onset, severity, and rate of progression can vary • Clinical symptoms include loss of vision, impaired color vision,
depending on the mutation type. There have been several genes central scotoma, and hemeralopia.
implicated in cone dystrophy (Renner et al. 2009). Symptoms • A bull’s-eye maculopathy is the most characteristic clinical
include visual loss, impairment in color vision, and increased visual feature.
difficulty in brightly lit conditions (hemeralopia). Visual acuity is • OCT shows loss of the outer retinal layers affecting the macula
variable but typically ranges between 20/20 and 20/200. Fundus and fovea, unlike in retinitis pigmentosa, in which there is
examination reveals a bull’s-eye pattern of RPE atrophy within often foveal sparing.
the macula, which is highlighted with fundus autofluorescence • OCT findings in advanced disease include complete atrophy
(Fig. 1). The peripheral retina is typically spared. OCT shows loss of the macula.
of the outer retinal layers affecting the macula (Fig. 2). Electro-
REFERENCE
physiologic testing is often required to confirm the diagnosis Renner AB, Fiebig BS, Weber BH, et al. Phenotypic variability and long-term
and shows a decreased or undetectable photopic and flicker follow-up of patients with known and novel PRPH2/RDS mutations. Am J
response. Ophthalmol. 2009;147:518–530.

FIG. 1. Color fundus image showing RPE loss at the fovea in a


characteristic bulls eye pattern.

FIG. 2. OCT corresponding to Fig. 1. Line scan shows outer retinal loss
through fovea and para-fovea.

Loss of outer
retina

S
1
N T

154
Malattia Leventinese (Doyne’s Honeycomb
Retinal Dystrophy)
Jay S. Duker
26.5
Summary Key OCT Features
Malattia leventinese, also referred to as dominant drusen, is • Many large, round drusen are always present (hyperreflective
caused by a known genetic defect (EFEMP1). The diagnosis is material between the retinal pigment epithelium and Bruch’s
typically made based on the clinical appearance, which includes membrane) (Figs. 1 and 2).
many variably sized, but predominantly large, drusen located • Small radial drusen, similar to cuticular drusen, may be present.
within the central macula and peripapillary region. The drusen
appear at an early age compared to drusen resulting from BIBLIOGRAPHY
age-related macular degeneration. Vision loss may occur as a Querques G, Guigui B, Levezial N, et al. Multimodal morphological and
result of the development of atrophic changes and secondary functional characterization of malattia leventinese. Graefes Arch Clin Exp
Ophthalmol. 2013;251(3):705–714.
choroidal neovascularization. OCT is used predominantly as an Zhang T, Xie X, Cao G, et al. Malattia leventinese/Doyne honeycomb retinal
adjuvant imaging modality to determine the extent and monitor dystrophy in a Chinese family with mutation of the EFEMP1 gene. Retina.
the progression of secondary features of disease. 2014;34(12):2462–2471.

Large round Small radial


drusen drusen

FIG. 1. OCT appearance of malattia leventinese with both large and small drusen.

155
Section 26: Retinal Dystrophies

FIG. 2. Wide-field fluorescein angiogram corresponding to Fig. 1


illustrates the extent of peripheral drusen.

FIG. 3. Wide-field fundus autofluorescence demonstrates extensive


peripheral RPE dropout.

S
2 Subretinal fibrosis
T N

FIG. 4. Advanced malattia leventinese with subretinal fibrosis. (Courtesy


Elias Reichel, MD.)

FIG. 5. Fundus photograph corresponding to Fig. 4 illustrates the


classical appearance of malattia leventinese. (Courtesy Elias Reichel, MD.)

156
Central Areolar Choroidal Dystrophy
Jay S. Duker 26.6
Summary Key OCT Features
Central areolar choroidal dystrophy (CACD) is an inheritable • The earliest features of disease include focal RPE disturbances
disorder affecting the macula that can resemble age-related that may resemble drusen.
macular degeneration (AMD) and is often confused for this entity. • With moderate disease, outer retinal atrophic changes are
Patients with CACD are typically younger and have a strong family present.
history of vision loss compared to those with AMD. In the earlier • In advanced stages of disease, generalized outer retinal,
stages of disease, RPE changes are present in the macula. Over RPE, and choriocapillaris atrophy develop. An absence of
time, severe RPE atrophy develops, leaving a well-circumscribed, sub-RPE deposits at this stage can help distinguish CACD
round area of tissue loss in the central macula. OCT is useful from geographic atrophy secondary to AMD (Figs. 1 and 2).
to identify key features of the disease at each stage and help
distinguish this entity from AMD. BIBLIOGRAPHY
Boon CJ, Klevering BJ, Cremers FP, et al. Central areolar choroidal dystrophy.
Ophthalmology. 2009;116(4):771–782.
Smailhodzic D, Fleckenstein M, Theelen T, et al. Central areolar choroidal
dystrophy (CACD) and age-related macular degeneration (AMD): differen-
tiating characteristics in multimodal imaging. Invest Ophthalmol Vis Sci.
2011;52(12):8908–8918.

FIG. 1. OCT of the left eye from a 53-year-old


woman who presented 25 years earlier with
decreased vision OU. At the time this OCT
was taken the visual acuity was 20/400 OU
and the findings were bilaterally symmetric.
There was a positive family history, with her
father having a similar disease and course.
This depicts the advanced stage of central
areolar choroidal dystrophy with atrophy of the
outer retina, RPE, and choriocapillaris. Note
the absence of any sub-RPE deposits that
would be more typical of geographic atrophy
secondary to AMD.

157
Section 26: Retinal Dystrophies

A B

FIG. 2. (A) Fundus photograph showing well-demarcated area of chorioretinal atrophy, corresponding to Fig. 1. (B) Fundus photograph from the
same patient 25 years earlier.

158
SECTION 27: POSTERIOR VITREOUS DETACHMENT

Stages of Posterior Vitreous Detachment


Darin R. Goldman 27.1
Summary Key OCT Features
The vitreous is a dynamic substance that ages similarly to other • Stages of PVD include stage 1: perifoveal PVD with intact
parts of the body. This aging process results in ongoing liquefac- vitreofoveal adhesion (Fig. 3); stage 2: isolated macular PVD
tion with weakening and eventual separation of vitreous adhesions with residual vitreopapillary adhesion (Fig. 4); stage 3: peripheral
to various structures in the posterior segment, including the optic PVD with residual vitreopapillary adhesion (Fig. 5); stage 4:
nerve, macula, retinal blood vessels, and peripheral retina. The complete PVD (Fig. 6) (Johnson 2005).
evolution of posterior vitreous detachment (PVD) occurs over many • Progressive stages (1–4) of PVD are all detectable via ocular
years through continuous vitreous separation from the posterior coherence tomography (OCT) (Figs. 1–7).
pole. The first stage of separation begins in the paramacular
region with eventual complete vitreopapillary separation. It is only REFERENCE
Johnson MW. Perifoveal vitreous detachment and its macular complications.
this final event, which may take years to develop, that causes
Trans Am Ophthalmol Soc. 2005;103:537–567.
typical symptoms of an acute PVD with a visible Weiss ring
clinically. Aberrant vitreous separation from posterior eye wall
structures can result in a host of pathologic conditions such
as vitreous hemorrhage, vitreomacular traction, macular hole,
epiretinal membrane, retinal tear, and retinal detachment.

Posterior Cortical Vitreous


* *
* *

S S
3 3
T N N T

A I
B I

FIG. 1A and B. OCT of a normal macula with attached posterior hyaloid. Although the vitreous cavity exhibits a generalized hyporeflective signal, the
presence of attached posterior hyaloid (lack of a posterior vitreous detachment) is evident by a diffuse, faint, grainy lightly reflective signal (asterisks).
Often, there will be a distinct demarcation (arrowheads) between layers of the posterior vitreous cortex that differ slightly in degree of reflectivity, which
aids in confirmation of the presence of attached cortical vitreous. Additionally, the edges of the posterior hyaloid are visible as thin hyperreflective
membranes (arrows), which blend almost imperceptibly with the surface of the macula.

FIG. 2. The earliest sign of posterior vitreous separation is marked by


focal separation between the posterior hyaloid and macular surface
Earliest sign of posterior vitreous separation (arrows).

S
3
N T

159
Broad vitreo-foveal adhesion

*
*
Section 27: Posterior Vitreous Detachment

S S
11 4
T N Stage 1 posterior vitreous detachment T N Stage 1 posterior vitreous detachment

A I
B I

Focal vitreo foveal adhesion

* * *
*
S S
3 3
N T
Stage 1 posterior vitreous detachment
T N
Stage 1 posterior vitreous detachment
C I
D I

FIG. 3A–D. Stage 1 posterior vitreous detachment is marked by posterior hyaloid separation from the surface of the retina surrounding the central
fovea (asterisks), while maintaining adhesions to the optic nerve and temporal to the macula. Within this stage, there is continued elevation of the
hyaloid from a flat configuration (A and B) to one that is more bowed (C and D), and the area of foveal adhesion transitions from broad to more focal.

* *
Posterior hyaloid Posterior hyaloid
* *

S S
1 3
N T Stage 2 posterior vitreous detachment T N Stage 2 posterior vitreous detachment

A I
B I

FIG. 4A and B. Stage 2 posterior vitreous detachment results in complete release of vitreomacular attachment with residual adhesion to the temporal
macula, optic nerve, and nasal periphery. At this stage, the posterior hyaloid face is easily visualized as a hyperreflective membrane above the macula
(arrowheads). The space between the posterior hyaloid and macula is generally darker or more hyporeflective (white asterisk) compared to the
reflectivity of the vitreous (yellow asterisk).

FIG. 5. OCT sectioned through the optic nerve head in stage 3 posterior
Vitreo-papillary attachment vitreous detachment. There is residual vitreopapillary attachment (arrows),
Vitreo-papillary attachment and the posterior hyaloid has released from the entire retina anteriorly.

S
1
T N Stage 3 posterior vitreous detachment

160
27.1
Homogeneous, hypo-reflective space

Homogeneous, hypo-reflective space

Stages of Posterior Vitreous Detachment


S S
3 3
T N Stage 4 posterior vitreous detachment N T Stage 4 posterior vitreous detachment

A I
B I

S
FIG. 6A–C. Stage 4, or complete posterior vitreous detachment
3 (PVD), is evident on OCT by a lack of any features of posterior
T N vitreous that have been depicted in Figs. 1 to 5. The vitreous cavity
Stage 4 posterior vitreous detachment has a homogeneous, hyporeflective appearance. Secondary features
C I associated with symptomatic stage 4 PVD, such as red blood cells in
the setting of vitreous hemorrhage, may be visible (C, arrows).

FIG. 7. Stages of PVD: Stage 1 indicates perifoveal vitreous separation


from the retina with intact vitreofoveal adhesion, stage 2 indicates
complete vitreous separation from the macula, stage 3 indicates further
separation of the vitreous from the peripheral retina while maintaining
vitreopapillary adhesion, and stage 4 indicates complete separation of the
posterior vitreous from all attachments to the posterior segment. (From
Johnson Perifoveal vitreous detachment and its macular complications. Trans Am
Ophthalmol Soc. 2005 Dec; 103: 537–567. figure 2 (stages of PVD).)
Stage 1 Stage 2

Stage 3 Stage 4

161
SECTION 28: ASTEROID HYALOSIS

Asteroid Hyalosis
Darin R. Goldman 28.1
Summary Key OCT Features
Asteroid hyalosis is a benign condition resulting in vitreous opaci- • OCT is often able to visualize macular details even in the
ties that appear as small, white, variably sized spheres (“stars setting of severe asteroid hyalosis.
in the night”) dispersed throughout the vitreous cavity with a • Asteroid hyalosis spheres appear on OCT as hyperreflective
gravity-dependent distribution(Fig. 1A). The opacities generally vertically oriented streaks of varying width, which likely cor-
produce no visual impairment for the patient; however, they can responds to the size of the sphere (Figs. 2–4).
cause difficulty in clinical visualization and diagnosis of retinal • Although physically present only within the vitreous cavity,
abnormalities. The condition is unilateral or significantly greater because of mirroring artifacts, lesions on OCT can appear
in one than the other and occurs in approximately 1 in 100 to overlie the retinal layers and choroid.
people. In some cases in which asteroid hyalosis is dense and • Shadowing effects are also characteristic of asteroid hyalosis
the macula cannot be viewed clinically, OCT can aid in visualizing and may be due to visualized flecks or those outside the frame
macular detail and any associated macular pathologic processes of image capture.
(Fig. 1B). Asteroid hyalosis has a classic appearance on OCT as
hyperreflective vertical streaks with varying width.

FIG. 1. (A) Color photograph of


Asteroid hyalosis dense asteroid hyalosis, which
obscures the ability to discern
Lamellar macular hole
details of the underlying fundus.
(B) Corresponding OCT has a
reduced signal but reveals fairly
good detail of the macula. In
this particular case, there was
concern for a possible full-
thickness macular hole; however,
OCT revealed a lamellar macular
3
S
hole without full-thickness defect.
T N
Asteroid
hyalosis
A B I

Asteroid hyalosis

Asteroid hyalosis

S S
1 4
T N T N
Asteroid hyalosis
I I

FIG. 2. Mild asteroid hyalosis with only a few lesions visible on OCT. FIG. 3. Moderate asteroid hyalosis showing characteristic OCT
appearance of hyperreflective, vertically oriented flecks.

162
FIG. 4. Severe asteroid hyalosis showing 28.1
many hyperreflective flecks. Note the
numerous areas of shadowing artifact
(right side of yellow lines). These areas are
associated with asteroid flecks located above
(white arrows, true image) and below (red

Asteroid Hyalosis
arrows, mirrored artifact) this plane.

S Asteroid hyalosis
2
T N

163
SECTION 29: VITREOUS HEMORRHAGE

Vitreous Hemorrhage
Darin R. Goldman 29.1
Summary when vitreous hemorrhage is present with sufficient visualization
of the fundus, OCT can be helpful to identify underlying macular
Spontaneous vitreous hemorrhage is a common disorder of abnormalities. Additionally, in cases of very subtle vitreous hemor-
the vitreous cavity, occurring in approximately 7 per 100,000 rhage, OCT can be useful to confirm its presence.
people (Spraul & Grossniklaus 1997). The appearance of vitreous
hemorrhage develops secondary to bleeding from normal or
neovascular blood vessels within the retina/vitreous and also Key OCT Features
may occur as a result of extension from layers underneath the
retina. The most common causes for nontraumatic, spontaneous • OCT findings in vitreous hemorrhage include visualization
vitreous hemorrhage include diabetic retinopathy, retinal tear/ of individual red blood cells, clumps of hemorrhage, and
detachment, vitreoretinal traction resulting from posterior vitreous secondary effects from shadowing (Figs. 1–4).
detachment, retinal venous occlusive disease, ruptured retinal • Individual red blood cells appear as small, densely hyper-
macroaneurysm, and exudative age-related macular degeneration. reflective spots, whereas diffuse hemorrhage appears as a
Spontaneous vitreous hemorrhage is a common cause for visual homogeneous sheet of hyperreflectivity.
impairment and often will resolve without treatment in mild cases.
More severe cases may require surgical vitrectomy. In the setting REFERENCE
of severe vitreous hemorrhage and no visualization of the fundus, Spraul CW, Grossniklaus HE. Vitreous Hemorrhage. Surv Ophthalmol.
B-scan ultrasound is the imaging modality of choice. However, 1997;42(1):3–39.

* * *
* *
*

Shadowing
artifact Vitreous
hemorrhage
S
3
N T

A B I

FIG. 1. (A) Color photograph of diabetic vitreous hemorrhage. (B) OCT visualizes the posterior hyaloid face (arrowheads) with underlying hemorrhage
that is both attached to the back of the hyaloid (asterisks) and also dispersed within the underlying vitreous (between arrows). Where the hemorrhage
attached to the posterior hyaloid is thickest, the underlying details are obscured by shadowing artifact.

Vitreous hemorrhage
Sub-hyaloid
hemorrhage

S
1
N T

A B I

FIG. 2. (A) Color photograph of diabetic vitreous hemorrhage, mostly contained in the sub-hyaloid space. (B) OCT shows mild vitreous hemorrhage
overlying the nasal macula. Temporally, there is subhyaloid hemorrhage (yellow arrow corresponds to same arrow in Fig. 2A) with underlying
shadowing artifact.

164
FIG. 3. Vitreous hemorrhage secondary to 29.1
ruptured retinal macroaneurysm. Where the
hemorrhage is denser, there is underlying
shadowing artifact (yellow arrows). Where the
hemorrhage is milder, there is no underlying
shadowing (white arrows).

Vitreous Hemorrhage
S
3
T N

S S
3 2
N T T N
Mild vitreous hemorrhage Vitreous hemorrhage
A I
B I

Red blood cells Vitreous Red blood


hemorrhage cells
streaks

S S
3 3
N T T N
Vitreous hemorrhage
C I
D I

Red blood Vitreous


cells hemorrhage streaks

FIG. 4A–E. (A) Mild vitreous hemorrhage in the setting of acute posterior vitreous
detachment might go unnoticed clinically. Small, round, hyperreflective spots correspond
to individual red blood cells (arrows). (B) More significant vitreous hemorrhage with
shadowing artifact (between arrowheads) from overlying vitreous hemorrhage, which is
S
located above the imaged frame. Individual red blood cells are also visualized (arrows). (C)
5 In addition to red blood cells (arrows), the thickness map reveals a black segmentation
N T
artifact (circle) that is due to localized signal blockage from overlying vitreous hemorrhage
E I in this region. (D and E) Both red blood cells (white arrows) and hyperreflective, vertical
streaks of vitreous hemorrhage (yellow arrows) are shown.

165
SECTION 30: VITREOUS INFLAMMATION

Vitreous Inflammation
Darin R. Goldman 30.1
Summary Key OCT Features
A wide variety of uveitides result in vitreous inflammation as • OCT images acquired in the presence of vitreous inflammation
part of their disease course. The ability to monitor inflammation will have loss of signal corresponding to the severity of vitreous
severity from visit to visit relies on an imprecise subjective clini- opacity.
cal assessment that lacks reproducibility. OCT provides a more • OCT signal intensity can be used to monitor uveitis activity
precise method to assess fluctuations in vitreous inflammation and response to treatment.
over time. The media opacity created by vitreous inflammation • Secondary effects of uveitis such as CME can be measured
impairs transmission of light. The corresponding light attenuation and monitored with precision in response to treatment.
is fairly evenly distributed and results in OCT signal degradation.
In the absence of other causes for OCT signal loss, such as poor REFERENCES
Keane PA, Karampelas M, Sim DA, et al. Objective measurement of vitre-
tear film or cataract, variation in OCT signal intensity can be
ous inflammation using optical coherence tomography. Ophthalmology.
useful in monitoring uveitis activity and response to treatment. 2014;121(9):1706–1714.
The variation in OCT signal intensity, which correlates with vitritis Zarranz-Ventura J, Keane PA, Sim DA, et al. Evaluation of objective vitritis
severity, provides a direct measure of disease activity that is grading method using optical coherence tomography: influence of phakic
objective and reproducible (Figs. 1–3). Other, more sophisticated, status and previous vitrectomy. Am J Ophthalmol. 2016;161:172–180.
methods for monitoring vitreous inflammation using OCT have
been described (Keane et al. 2014; Zarranz-Ventura et al. 2016).
Additionally, OCT is very useful to detect associated cystoid
macular edema (CME) and monitor response to treatment (Fig. 4).

Signal strength = 4/10 Signal strength = 6/10


Cystoid macular
edema

S S
3 3
N T N T
Idiopathic posterior uveitis Idiopathic posterior uveitis
A I
B I - following intravitreal steroid

FIG. 1. (A) Idiopathic posterior uveitis. The signal strength is 4/10 because of diffuse vitreous inflammation, which is evidenced by the poor overall
resolution of the image. There is also significant associated CME. (B) At 1 month after intravitreal dexamethasone implant, CME resolved and signal
strength improved to 6/10 along with improved vitreous inflammation. The improvement in signal strength and resolution of CME both indicate a
positive response to treatment.

166
30.1
500 500

400 400

Vitreous Inflammation
300 300

200 200

100 100

0 m 0 m

+150

+75

0 m

–75

–150
Overlay: OCT Fundus Transparency: 0% Overlay: ILM-RPE Difference Transparency: 0%

Extracted B-Scan

N T N T

A
FIG. 2. (A) In addition to the structural OCT B-scan (bottom), thickness maps (top) and difference map (middle, right) are very helpful to visualize
changes over time, particularly in response to treatment. Pretreatment (yellow box) thickness maps show significant central CME, which resolves after
treatment (red box). This change over time is best visualized on the difference map (middle, right).
Continued

167
500 500

400 400
Section 30: Vitreous Inflammation

300 300
Pre-Tx Post-Tx
200 200

100 100

0 m 0 m

+150

+75

0 m

–75

–150

T N T N

B
FIG. 2, cont’d. (B) A similar treatment response in the fellow eye is shown, highlighted by the thickness maps and difference map.

168
30.1

Cystoid macular
edema

Vitreous Inflammation
Subretinal
S fluid
1
N T

B I

Birdshot retinochoroidopathy
- following intravitreal steroid
S
1
N T FIG. 3. (A) Color photograph of birdshot retinochoroidopathy.
(B) Pretreatment there is significant CME and subretinal fluid. (C) After
C I treatment with intravitreal steroid, there is complete resolution of CME
and subretinal fluid.

169
SECTION 31: PERIPHERAL RETINAL ABNORMALITIES

Tractional Retinal Detachment


Darin R. Goldman 31.1
Summary finally the development of underlying subretinal fluid. Tractional
retinal detachments, in the setting of proliferative vitreoretinopathy,
Tractional retinal detachment most commonly occurs secondary to develop as a more acute process with prominent preretinal
proliferative diabetic retinopathy or proliferative vitreoretinopathy. membranes and significant subretinal fluid.
The clinical appearance is characterized by visible epiretinal and
vitreous membranes exerting tractional forces on the retinal
surface. The type and location of retinal detachment depend on
Key OCT Features
where these forces are applied and how strong they are. Isolated • Tractional retinal detachments resulting from proliferative
macular, isolated peripheral, or total retinal detachment can occur. diabetic retinopathy are characterized by multifocal areas of
The existence of a detachment is defined by the presence of traction, preretinal membranes, schisis-like changes within
subretinal fluid. Such fluid is often detectable on OCT before the retina, and areas of subretinal fluid (Figs. 1–4).
being visible clinically (Figs. 1–5). Tractional retinal detachments, • Tractional retinal detachments secondary to other causes,
in the setting of proliferative diabetic retinopathy, begin with such as proliferative vitreoretinopathy, haves more prominent
focal traction on the retina, which progressively enlarges causing preretinal membranes and substantial amounts of subretinal
thickening of the underlying retina with schisis-like changes and fluid (Fig. 5).

170


Tractional Retinal Detachment


A

*
*
*
T N
S
2
T N

Chronic diabetic tractional


B I
retinal detachment

Posterior hyaloid

Vitreous pegs

S
S
5
T N
Chronic diabetic tractional
C I retinal detachment D I

FIG. 1A–D. (A) End-stage proliferative diabetic retinopathy with atrophic tractional retinal detachment in a “wolf-jaw” ’ configuration. (B) OCT shows
multiple areas of traction (arrows) and loculated subretinal fluid (asterisks). The retina is severely attenuated and disorganized with loss of discernable
layers. (C) The posterior hyaloid is visible with pegs of vitreous inserting onto the retinal surface. (D) OCT thickness map is limited because of
segmentation errors, which are common in this setting.

171
Diffuse preretinal
membrane
Section 31: Peripheral Retinal Abnormalities

Subretinal fluid

Intraretinal fluid/
retinal disorganization
Advanced diabetic tractional
A B retinal detachment

FIG. 2A and B. (A) Advanced diabetic tractional retinal detachment involving the macula. (B) OCT reveals a diffuse membrane on the macular surface
(arrowheads), cystic intaretinal changes with retinal disorganization, a serpentine-like retinal arrangement, and significant subretinal fluid.

Focal vitreomacular
traction

S
3
N T

A B I

Focal vitreomacular traction


Schisis-like
intraretinal changes

S
3 Subretinal fluid
N T
Diabetic tractional
C D I
retinal detachment

FIG. 3A–D. (A) Proliferative diabetic retinopathy with preretinal fibrosis in the absence of any retinal detachment. (B) There is a focal area of
vitreomacular traction but no detachment, because of the lack of subretinal fluid. (C) At a later date, the preretinal fibrovascular proliferation
progressed to the point of causing a tractional retinal detachment involving the peripheral macula. (D) OCT reveals tractional separation of the retinal
layers and subretinal fluid, which defines the presence of retinal detachment.
172


Tractional Retinal Detachment


Subretinal fluid
Schisis-like
intraretinal changes

Diabetic tractional
A B retinal detachment

FIG. 4A and B. (A) Diabetic tractional retinal detachment with early “wolf-jaw” configuration. (B) Structural OCT shows the presence of subretinal fluid
nasal to the fovea and tractional thickening, or schisis-like changes, temporal to the fovea. The OCT thickness map (inset, bottom-left) is useful to
better illustrate the three-dimensional nature of the tractional effect exerted in a circumferential manner on the edges of the macula.

Diffuse preretinal
membrane

Outer retinal corrugations

Subretinal fluid

Tractional retinal detachment


A B

Postoperative apperance
C D

FIG. 5A–D. (A) Tractional retinal detachment secondary to proliferative vitreoretinopathy after unsuccessful primary rhegmatogenous retinal
detachment repair. (B) There is a thick, diffuse preretinal membrane along the surface of the macula (arrowheads) that gives the macular surface a
smooth appearance. Additionally, there is significant underlying subretinal fluid and a corrugated configuration to the outer retinal surface (arrows). (C)
After surgical repair, the preretinal membranes have been removed and the macula is reattached. A residual intravitreal gas bubble is visible superiorly.
(D) OCT shows complete resolution of the preretinal membrane with a smooth macular contour and no subretinal fluid.
173
Rhegmatogenous Retinal Detachment
Darin R. Goldman 31.2
Summary Key OCT Features
Rhegmatogenous retinal detachment (RRD) results from subretinal • Subretinal fluid is present, evidenced by a homogeneous
fluid accumulation underneath the retina after a retinal break. hyporeflective space underneath the neurosensory retina,
The detachment starves the retina of nourishment, with resultant which increases in height from nasal to temporal (Figs. 1–8).
damage to photoreceptors over time. If the central macula is • The underlying retinal pigment epithelium (RPE) remains
involved, vision loss occurs that recovers to a variable degree attached with a smooth contour, which can be visualized in
after successful reattachment. This visual recovery correlates shallow detachments.
best with presenting visual acuity but has proved difficult to • In acute detachments, retinal corrugations and cystoid edema
predict. However, OCT may provide insight into predicting and are present (Fig. 1).
monitoring visual recovery based on identifiable anatomic changes. • In chronic detachments, a relatively flat configuration and
Although these anatomic alterations are not well understood, both atrophic retinal changes are seen (Figs. 6 and 8).
qualitative and quantitative changes within the photoreceptor • Microstructural changes within the photoreceptor layers cor-
microstructure are thought to be largely responsible. Various relate with final visual acuity.
studies have attempted to clarify these suspicions using primar- • Optical density of subretinal fluid increases with duration of
ily OCT-based end points. OCT findings have been shown to detachment.
correlate with final visual acuity after macula-involving retinal • The most useful OCT output formats to determine whether
detachments, including outer nuclear layer thinning and ellipsoid subretinal fluid involves the fovea are thickness maps and
zone irregularities (Sridhar & Flynn 2014). Additionally, overall vertically oriented B-scans.
photoreceptor thickness may correlate with visual recovery after
retinal reattachment with an initial thinning followed by normaliza- REFERENCES
Leshno A, Barak A, Loewenstein A, et al. Optical density of subretinal fluid
tion, which coincides with visual recovery (Terauchi et al. 2015).
in retinal detachment. Invest Ophthalmol Vis Sci. 2015;56(9):5432–5438.
Another potentially useful parameter measurable on OCT is the Sridhar J, Flynn HW Jr. Spectral-domain optical coherence tomography
optical density of subretinal fluid, which increases with duration imaging of macula-off rhegmatogenous retinal detachment. Clin Ophthalmol.
of detachment and correlates with postoperative visual acuity 2014;8:561–566.
(Leshno et al. 2015). Terauchi G, Shinoda K, Matsumoto CS, et al. Recovery of photoreceptor inner
and outer segment layer thickness after reattachment of rhegmatogenous
retinal detachment. Br J Ophthalmol. 2015;99(10):1323–1327.

174
31.2
Vitreous cavity

Rhegmatogenous Retinal Detachment


Corrugations
Thickened
photoreceptors
S
3
T N Subretinal fluid
Retinal edema
A B I

FIG. 1A and B. (A) Color photograph of a macula-off rhegmatogenous retinal detachment within 2 weeks of symptom onset. (B) OCT shows
corrugations of the underlying retinal surface, diffuse retinal edema, and enlargement of the photoreceptors.

FIG. 2. Macula-off rhegmatogenous


Vitreous cavity detachment illustrates retinal edema,
thickened photoreceptors, and vitreous
inflammation, which suggests a longer
duration of detachment.

Vitreous
Inflammation

Retinal
edema
Thickened
S
photoreceptors
3
N T
Subretinal fluid
I

175
Section 31: Peripheral Retinal Abnormalities

Preoperative VA = 20/400 Postoperative VA = 20/40

T N T N

S S

I I
A B

FIG. 3A and B. (A) Preoperative OCT of macula-off rhegmatogenous retinal detachment with visual acuity of 20/400. (B) Postoperative OCT 1 month
after reattachment of the retina by vitrectomy. Visual acuity improved to 20/40. On reattachment, resolution of subretinal fluid, retinal edema, and
retinal corrugations are seen, with restoration of a fairly normal macular contour. The retinal thickness, mostly the result of photoreceptor integrity,
remains normal.

176
31.2

Rhegmatogenous Retinal Detachment


T N T N

S S

I I
A C

Full-thickness
retinal elevation Fovea is attached

S S
7 Intact RPE with 7
T N
smooth contour T N

B I
D I

FIG. 4A–D. Macula-involving, but fovea-on, rhegmatogenous retinal detachment. (A) Thickness map (top) illustrates the border of the detached retina
(yellow arrowheads), located inferior to the fovea. The fovea remains attached (circles). (B) Vertically oriented B-scan image best illustrates the extent
of the subretinal fluid and is useful to evaluate the status of the fovea. (C and D) At 6 months after surgical reattachment, the macula has regained a
normal appearance and the fovea is completely reattached

177
FIG. 5A and B. RRD secondary to giant retinal tear. (A) Color photograph
illustrates attached and detached retina. (B) Corresponding OCT at the border
between attached and detached retina.
Section 31: Peripheral Retinal Abnormalities

Detached
Retina

Attached
Retina

N T

B I

178
31.2

Detached
Detached retina
retina

Rhegmatogenous Retinal Detachment


Mirror
Artifact
Attached
retina Attached
retina
2
S
Intact RPE
T N

A B I

FIG. 6A and B. Chronic RRD. (A) Color photograph illustrates the absence of retinal corrugations. (B) Corresponding OCT (line in color photograph
indicates cross section) shows distinct areas of attached and detached retina. The detached retina is flat and somewhat attenuated. Note the dual
mirror artifacts with inverted images.

Detached retina
Attached
retina

Subretinal fluid

FIG. 7. The transition zone from attached retina to detached retina is illustrated. (Courtesy Netan Choudhry, MD.)

Pigmented demarcation line

A B
Preoperative VA = 20/400 Postoperative VA = 20/60
No
Foveal corrugations Foveal thickness
attenuation improved

Mirror
Artifact
Swollen photoreceptor Restored outer
S outer segments S retinal microstructure
2 1
N T
Subretinal fluid N T

C I
D I

FIG. 8A–D. Chronic RRD with presenting visual acuity of 20/400. (A) Color photograph centered on the optic nerve shows subretinal fluid in the
macula (within arrowheads). (B) Color photograph of the inferonasal periphery reveals a pigmented demarcation line, indicative of chronicity. (C)
Preoperative OCT through the fovea shows a smooth contour of the retina with significant subretinal fluid. The central fovea is attenuated, and the
photoreceptor outer segments are markedly swollen. (D) At 3 months after surgical reattachment of the retina, the foveal thickness increased to near
normal and the outer retinal bands are restored. Visual acuity returned to 20/60. Note the presence of a myopic staphyloma.

179
Bullous Retinoschisis
Darin R. Goldman 31.3
Summary Key OCT Features
Bullous senile retinoschisis involves splitting of the retina along a • OCT provides a definitive method of differentiating retinal
plane within or between a particular retinal layer or layers. Although elevations resulting from retinoschisis or retinal detachment
schisis is most commonly located in the superotemporal peripheral (Figs. 1–7).
retina, it may occur in any meridian and is often bilateral. Schisis • Schisis is defined by a separation within the retina that occurs
is typically limited to the peripheral retina but may communicate between the inner and outer retinal layers, always leaving outer
posteriorly and involve the macula. The involved retina appears retinal layers (ORLs) overlying the RPE. This is in contrast to
smoothly elevated, which may be difficult to distinguish from retinal detachment, in which the plane of separation is between
rhegmatogenous retinal detachments. This distinction is critical the RPE and neurosensory retina.
to avoid unnecessary intervention in the case of retinoschisis and • At least two distinct partitions of split retina are visible with
guide appropriate treatment in the case of retinal detachment. intervening connecting strands of stretched retinal elements,
Certain clinical signs and diagnostic evaluation such as visual thought to be Müller’s cells.
field testing or laser demarcation can help clarify the diagnosis, • Retinal separation widens from posterior to anterior.
although these methods are not definitive. OCT provides an
unequivocal method of distinguishing retinoschisis from retinal BIBLIOGRAPHY
Choudhry N, Golding J, Manry MW, et al. Ultra-widefield steering-based spectral
detachment (Figs. 1–7). Multiple, separate areas of retina should
domain optical coherence tomography imaging of the retinal periphery.
be imaged to increase the likelihood of identifying any associated Ophthalmology. 2016;123(6):1368–1374.
detachment that could be overlooked by imaging only one location. Stehouwer M, Tan SH, van Leeuwen TG, et al. Senile retinoschisis versus
Because of the typical peripheral location of retinoschisis, OCT retinal detachment, the additional value of peripheral retinal OCT scans
imaging can be cumbersome and therefore may be underutilized. (SLSCAN-1, Topcon). Acta Ophthalmol. 2014;92(3):221–227.
Orientation of the OCT imaging plane perpendicular to the elevated
area while including the retinal transition from flat to elevated
is helpful.

Outer retinal
layers present
Bullous
retinoschisis

RPE
Retinoschisis
Mirror
artifact
S
3
T N

I
A B

FIG. 1A and B. (A) Color photograph of bullous retinoschisis located in the superotemporal retinal periphery. (B) OCT obtained with the scanning
plane orthogonal to elevated retina, while still including a portion of flat retina (see yellow line, Fig. 1A). Note that in the area where the retina is most
elevated, ORLs are visible overlying the RPE, which defines schisis on OCT.

180
FIG. 2A and B. (A) Color photograph 31.3
of bullous retinoschisis located in the
inferotemporal retinal periphery. (B) OCT was
obtained with the scanning plane orthogonal
to the elevated retina, while still including a
portion of flat retina (see yellow line, Fig. 2A).

Bullous Retinoschisis
ORLs are visible overlying the RPE, which
defines schisis on OCT.

Bullous
retinoschisis

Outer retinal
layers present
Retinoschisis

B RPE

Retinoschisis widens from


nasal to temporal
Retinoschisis

Retinoschisis

Outer retinal
Flat retina layers present

1
S
2
S
RPE
N T N T
Müller’s cells
A I
B I

FIG. 3A and B. (A) Transition zone from flat retina to shallow retinoschisis is illustrated. (B) More of the area of retinoschisis is shown, which widens from
left to right (nasal to temporal). Connecting elements, thought to be Müller’s cells, are visible, and outer retinal layers are present overlying the RPE.
181
Section 31: Peripheral Retinal Abnormalities

FIG. 4. Shallow retinoschisis with interweaving retinal elements that appear stretched. (Courtesy Netan Choudhry, MD.)

Retinoschisis Retinal detachment

Transition zone

FIG. 5. Combined retinoschisis and retinal detachment. Both areas are distinctly visualized on each side of the transition zone (yellow arrow). (Courtesy
Netan Choudhry, MD.)

Retinoschisis
Retinoschisis Outer plexiform layer

Outer nuclear
layer

S S
1 1
N T N T

A I
B I

FIG. 6A and B. (A) Peripheral retinoschisis includes multiple planes of separation. (B) The peripheral retinoschisis communicates posteriorly to involve
the macula, where the plane of separation is easier to determine between the outer plexiform layer and outer nuclear layers.

FIG. 7. Wide-angle OCT imaging the retina


from ora to ora. Bullous retinoschisis is visible
temporally. (Courtesy Netan Choudhry, MD.)

Retinoschisis

182
Lattice Degeneration
Darin R. Goldman 31.4
Summary This can be overcome with a skilled photographer and coopera-
tive patient. In the future, OCT imaging modalities may include
Lattice degeneration is a common retinal disorder that affects the modifications that assist with peripheral imaging, which would
peripheral retina and increases the risk for developing a retinal make its use more practical for lattice degeneration.
tear or retinal detachment. Lattice degeneration is present to
some degree in 5% to 10% of the population, varies in exten-
siveness, and is usually asymptomatic. The presence of lattice Key OCT Features
degeneration is typically detected as an incidental finding during
routine ophthalmoscopy. The clinical appearance can vary widely • Lattice degeneration is characterized by firm vitreoretinal
adhesions on the edges with clear vitreous overlying the lesion.
but generally includes thin, well-demarcated, pigmented ovoid
patches of retina located in a circumferential orientation. Within • The involved retina typically demonstrates generalized thinning
(Figs. 2–7).
the involved area, there may be sclerotic vessels or atrophic
holes. Although usually darkly pigmented, lattice degeneration • Subclinical retinal detachment is very common, a feature best
appreciated on OCT (Figs. 4–7).
may appear hypopigmented (Fig. 1). The vitreous overlying lattice
degeneration is liquefied and, on the edges of lattice, there is • U-shaped (in cross section) overlying vitreous insertion/traction
is common (Fig. 4).
firm vitreoretinal adhesion, which has been established mostly
by histopathologic studies. The vitreoretinal interface has not
BIBLIOGRAPHY
been well studied using OCT, although OCT provides a unique Manjunath V, Taha M, Fujimoto JG, et al. Posterior lattice degeneration
opportunity to evaluate this entity in vivo. The difficulty in imaging characterized by spectral domain optical coherence tomography. Retina.
lattice degeneration with OCT is due to its peripheral location. 2011;31(3):492–496.

FIG. 1. Wide-field image illustrating characteristic appearance of lattice


degeneration. (Courtesy of Netan Choudhry, MD.)

Lattice degeneration

Firm vitreoretinal FIG. 2. The OCT imaging plane is oriented perpendicular to the short axis
adhesion of lattice degeneration. The involved retina is thin, with firm vitreoretinal
adhesion to both edges of the lattice degeneration.
Th
in
re
tin
a Firm vitreoretinal
de La adhesion
ge ttic
ne e
ra Re
tio tin
n a
th wit
ic h
kn no
es rm
s al

183
Section 31: Peripheral Retinal Abnormalities

Lattice degeneration

FIG. 3. In this example of lattice degeneration, there is broad vitreoretinal traction over the involved region that creates an appearance of localized
retinal thickening. (Courtesy of Netan Choudhry, MD.)

U-shaped vitreous
insertion

Intraretinal Subclinical retinal


cyst detachment

FIG. 4. The area imaged is just on the edge of a small area of lattice degeneration, which highlights the abnormal appearance of the overlying
vitreous. The vitreous is thickened, appearing as a broad, U-shaped, hyperreflective band (white line). Secondary features, including a small subclinical
retinal detachment and an intraretinal cyst, are also visible. (Courtesy of Netan Choudhry, MD.)

Subclinical retinal
detachment

FIG. 5. Two small areas of lattice degeneration are shown that have an atypical OCT appearance. The focal vitreoretinal traction overlying each area
has created a schisis-like effect. Splitting and elevation of the inner retinal layers are seen toward the vitreous cavity. (Courtesy of Netan Choudhry, MD.)

184
FIG. 6A–C. (A) At the edge of this area 31.4
of lattice degeneration, there is firm
vitreoretinal adhesion causing a subclinical
retinal detachment, which was not visible
clinically. (B) Many of the features of lattice
degeneration visible on OCT are illustrated.

Lattice Degeneration
(C) A retinal break is present at the edge of
vitreoretinal traction, along with a small cuff
of subclinical subretinal fluid. (Fig. 6B courtesy
Firm vitreoretinal
Netan Choudhry, MD.)
adhesion

de La
ge ttic
ne e
ra
tio
n Subclinical retinal
detachment

Thickened vitreous
band

Thin retina
B Subclinical retinal detachment

Firm
vitreoretinal
adhesion

Retinal
hole

S
4
T N Cuff of
subretinal fluid

I
C

Subclinical retinal Clear vitreous above


detachment lattice degeneration

FIG. 7. A subclinical retinal detachment is shown, associated with typical lattice degeneration. The vitreous immediately overlying the lattice
degeneration is optically empty and completely hyporeflective compared to the surrounding vitreous, which exhibits some degree of granular
reflectivity. (Courtesy Netan Choudhry, MD.)

185
Myelinated Nerve Fiber Layer
Darin R. Goldman 31.5
Summary correlates with the thickness of the mNFL. Blood vessels within
the mNFL are highlighted by the contrast in reflectivity between
Myelinated nerve fiber layer (mNFL) is a benign clinical entity that the adjacent mNFL and relatively hyporeflective vessel lumen. In
results from an embryologic developmental anomaly whereby focal distinction to other, similar-appearing pathologic entities, there is
areas of the retinal nerve fiber layer fail to lose their myelin sheath. no associated cystoid macular edema, retinal atrophy, overlying
Clinically, mNFL appears as distinct white patches on the inner vitreous inflammation, or dynamic changes over time.
retinal surface. This appearance may mimic acute pathologic retinal
conditions such as retinal edema from branch retinal artery occlusion,
acute retinal necrosis, or retinal vasculitis. Given the benign nature
of mNFL, the distinction is crucial. OCT exhibits characteristic
Key OCT Features
features that are helpful to confirm the diagnosis of mNFL (Figs. • The mNFL displays characteristic OCT features, including
1–3). These features include a homogeneous, highly reflective extreme hyperreflectivity of the involved superficial retinal layer
band in the affected area of the retina that is isolated to the most with underlying shadowing that correlates with the thickness
superficial retinal layer. Areas of mNFL vary in thickness but are of the mNFL.
thickest toward their center. The associated intense hyperreflectivity • There is absence of any associated retinal thinning or overlying
results in underlying shadowing. This shadowing causes a loss vitreous inflammation.
of the distinction to the outer retinal layers, the degree of which • The OCT appearance remains stable over time.

186
31.5

Myelinated Nerve Fiber Layer


Myelinated nerve fiber layer (mNFL)

Myelinated nerve fiber layer (mNFL)

A A

Typical mNFL

Typical mNFL

S S
3 8
N T N T

B I B I

Thin mNFL
Normal
Thicker mNFL
retinal
Distinct
border

mNFL
Retinal blood vessel

S S
4 13
N T N T

C I C I Shadowing obscures underlying retinal layers

FIG. 1A–C. (A) Color photograph of typical mNFL. The inner retina FIG. 2A–C. (A) Color photograph of the mNFL located on the inferior
is involved with some obscuration of retinal blood vessels. (B) edge of the macula. (B) Horizontally oriented OCT image toward
Horizontal OCT imaging plane illustrates characteristic intense, diffuse superior edge of the mNFL (plane corresponds to white line in A), which
hyperreflectivity of the involved region on the inner retinal surface. The captures both thin and thick areas of the mNFL with varying degrees of
underlying retinal layers are not discernable because of shadowing. (C) underlying shadowing, which correlates to the thickness of the mNFL.
Vertical OCT imaging plane transects both thinner and thicker regions Characteristic extreme, diffuse hyperreflectivity of the mNFL is present.
of the mNFL. In the thin area, underlying retinal layers are discernable, (C) Horizontally oriented OCT image within the midsection of the mNFL
whereas in the thicker area, underlying retinal layers are obscured by (plane corresponds to black line in A). Because of the thickness of the
shadowing. Note the blood vessel that is highly visible within the mNFL mNFL in this region, the underlying retina is obscured from significant
because of the contrasting reflectivity of the mNFL and vessel lumen shadowing. Note that the border between normal retina and mNFL is
(yellow arrow in A and C). distinct (yellow arrow in A and C).

187
FIG. 3A and B. (A) Color photograph of
mNFL located inferonasally. (B) OCT through
mNFL and two interspersed retinal blood
vessels.
Section 31: Peripheral Retinal Abnormalities

Myelinated nerve fiber layer (mNFL)

Retinal blood vessels

mNFL
S
3
T N

I
B

188
Index

Page numbers followed by “f ” indicate figures.

A Autoimmune process, posterior scleritis and,


127
Chediak-Higashi syndrome, 80
“Cherry red spot”, 98, 99f
Axial elongation, 66 Choriocapillaris
A-scans, multiple, 4 en face structural OCT images of, 8f
Acute central serous chorioretinopathy, 56f
outer photoreceptors layers in, 58f B infiltration of, in choroidal metastases,
141f
Acute posterior multifocal placoid pigment Chorioretinal folds, in posterior scleritis, 127f
epitheliopathy (APMPPE), 103, B-scan see Line scans Chorioretinal lesion, in Candida chorioretinitis,
104f–106f Best disease, 152, 152f–153f 129f
Acute retinal necrosis syndrome, 132, Bilateral uveitis, Vogt-Koyanagi-Harada disease Chorioretinal OCT image, 8f
133f and, 114 Chorioretinitis
Acute rhegmatogenous retinal detachment, Birdshot retinochoroidopathy, 100, 100f–102f, acute syphilitic posterior placoid, 121,
174 169f 122f–123f
Acute syphilitic posterior placoid chorioretinitis Blink artifact, 10, 11f Candida, 129, 129f–131f
(ASPPC), 121, 122f–123f Blockage artifacts, 13, 13f toxoplasmic, 119, 119f–120f
Adhesion, vitreomacular, 40, 40f–41f Blue-light reflectance, drusen and, 21f–22f tuberculosis and, 124, 126f
Age-Related Eye Disease Study (AREDS) scale, Branch retinal artery occlusion (BRAO), 96, 97f Chorioretinopathy, central serous, 55, 58f
16 Branch retinal vein occlusion, 90, 91f acute, 56f
Age-related macular degeneration (AMD), 16 BRAO see Branch retinal artery occlusion associated to pigmented epithelium
exudative, type 2 choroidal neovascular Breast, choroidal metastases of, 140 detachment, 57f
membrane and, 32 Bruch’s membrane, 16 characteristic of, 55
geographic atrophy secondary to, 25f choroidal neovascular membrane and, 30 findings of, 55
isolated pigment epithelial detachment and, Bullous retinoschisis, 180, 180f–182f inactive, with thick choroid and large
28 peripheral, 182f choroidal vessels, 57f
polypoidal choroidal vasculopathy and, 38, shallow, 182f with secondary CNV, 58f
38f–39f Bulls-eye maculopathy, 68 Choroid, normal, 7
Albinism Choroidal dystrophy, central areolar, 157,
ocular, 80
oculocutaneous, 80, 80f–81f C 157f–158f
Choroidal elevation, tuberculosis and, 124
Amaurosis fugax, 96 Choroidal granuloma, tuberculosis and, 124,
AMD see Age-related macular degeneration C-scan, 85f 125f–126f
Angiography Calcified drusen, in geographic atrophy, Choroidal hemangioma, solitary, 136, 136f
fluorescein, for multiple evanescent white dot 26f–27f Choroidal infiltration, tuberculosis and, 124,
syndrome, 107, 108f Candida albicans, 129 125f
indocyanine green, for multiple evanescent Candida chorioretinitis, 129, 129f–131f Choroidal inflammation, Vogt-Koyanagi-Harada
white dot syndrome, 107 Capillary hemangioma, retinal, 137, 137f disease and, 114
Angioid streaks, 78, 78f Ceftriaxone therapy, for acute syphilitic Choroidal mass, dense, in choroidal
Anti-vascular endothelial growth factor posterior placoid chorioretinitis (ASPPC), metastases, 140
(anti-VEGF) 122f Choroidal melanoma, 135, 135f
for central retinal vein occlusion, 92 Central areolar choroidal dystrophy, 157, Choroidal metastases, 140, 141f
for multifocal choroiditis and panuveitis and 157f–158f Choroidal neovascular membrane
punctate inner choroidopathy, 111 Central retinal arterial occlusion (CRAO), 98, myopic, 59, 60f
for myopic CNV, 59 99f clinical appearance of, 61f
Antifungal therapy, for Candida chorioretinitis, Central retinal vein occlusion (CRVO), 92, features of, 59
129 93f–95f type 2 leakage pattern of, 61f
Areolar choroidal dystrophy, central, 157, Central serous chorioretinopathy, 7, 55, 58f type 1, 30
157f–158f acute, 56f type 2, 32, 33f
Artifacts associated to pigmented epithelium type 3, 34, 34f
blink, 10, 11f detachment, 57f Choroidal neovascularization (CNV), 78, 78f
mirror, 10, 10f characteristic of, 55 isolated pigment epithelial detachment and,
misalignment, 10, 10f findings of, 55 28
motion, 10, 11f inactive, with thick choroid and large Choroidal nevus, 134, 134f
in OCT angiography, 10–13 choroidal vessels, 57f Choroidal thickening, 7
out of range error, 12, 12f isolated pigment epithelial detachment and, in acute retinal necrosis syndrome, 132
software breakdown, 10, 11f 28 Choroidal vascular pattern, obscuration of, in
vignetting, 10, 10f with secondary CNV, 58f choroidal melanoma, 135
Asteroid hyalosis, 162, 162f–163f type 1 choroidal neovascular membrane Choroidal vasculopathy, polypoidal, 38,
Atrophy, geographic, 24 and, 30 38f–39f

189
Choroiditis En face structural OCT images, 8f Fundus autofluorescence, 21f–22f, 25f–27f
in acute retinal necrosis syndrome, 132 Endogenous fungal chorioretinitis, 129 Fungal chorioretinitis, 129
multifocal, 111, 111f–113f Endophthalmitis, in Candida chorioretinitis,
serpiginous, 109, 110f
Choroidopathy, punctate inner, 111, 111f–113f
130f–131f
Enhanced depth imaging (EDI), 7, 8f G
Chronic rhegmatogenous retinal detachment, Epiretinal membrane (ERM), 52, 54f
Index

174, 179f appearance of, 53f Ganglion cell complex (GCC), 1, 3f


Cirrus HD-OCT, 1 macular, 52f Geographic atrophy, 24
Classic choroidal neovascular membrane, 32 OCT, 52f bilateral, 25f
CNV see Choroidal neovascularization postoperative, 53f macular cube scan in, 26f
Coats’ disease see Type 1 macular associated with pseudohole, 53f multifocal, with retinal pigment epithelium,
telangiectasia associated with schisis between inner and 26f–27f
Combined hamartoma, of retina and RPE, 139, outer retinal layers, 53f secondary to age-related macular
139f features of, 52 degeneration, 25f
Computed tomography, for retinitis in lamellar macular hole, 50 Giant retinal tear, rhegmatogenous retinal
pigmentosa, 148 mild, 52 detachment secondary to, 178f
Cone dystrophy, 154, 154f Epithelial tear, retinal pigment, 37, 37f Glaucoma, 90, 92
Cotton wool spots, 84f, 86 ERM see Epiretinal membrane
CRAO see Central retinal arterial occlusion
CRVO see Central retinal vein occlusion
Errors, OCT, 10–12
External limiting membrane H
Cube scan, macular, 26f in acute posterior multifocal placoid pigment
Cystic retinal degeneration, in solitary choroidal epitheliopathy, 103 Haller/Sattler layers, 8f
hemangioma, 136 in acute syphilitic posterior placoid Hamartoma
Cystoid macular edema chorioretinitis, 121 combined, of retina and RPE, 139, 139f
in combined hamartoma, of retina and RPE, simple, of retinal pigment epithelium (RPE),
139
isolated, 76, 77f F 138, 138f
Hard drusen, 16
in vitreous inflammation, 166, 166f, 169f Hard exudate, in retinal capillary hemangioma,
False negative flow, artifacts, 13 137, 137f

D False positive flow, artifacts, 13


Fibrosis
Heidelberg Spectralis, 1
Hemangioma
in combined hamartoma, of retina and RPE, retinal capillary, 137, 137f
Dalen-Fuchs nodules, 116, 117f 139 solitary choroidal, 136, 136f
Dense hyperreflectivity, in simple hamartoma, subretinal, in acute syphilitic posterior placoid Hemeralopia, 154
of retinal pigment epithelium (RPE), 138 chorioretinitis (ASPPC), 123f Hemorrhage
Depth-resolved technology, 6 Flecks, pisciform, 149, 149f subretinal, 35, 35f
Diabetes, vitreous hemorrhage and, 164f Floaters, 9, 9f vitreous, 164, 165f
Diabetic macular edema, 84, 84f–85f Fluorescein angiography, 22f diabetic, 164f
Diabetic retinopathy for BRAO, 96, 97f Hermansky-Pudlak syndrome, 80
in Candida chorioretinitis, 129f for central serous chorioretinopathy, 55 High dynamic range imaging, 9
in diabetic macular edema, 84 for CRAO, 98 Human leukocyte antigen (HLA)-A29, birdshot
nonproliferative, 86, 86f–87f for CRVO, 92 retinochoroidopathy associated with,
proliferative, 88, 88f–89f, 170, 171f–173f for multiple evanescent white dot syndrome, 100
Diffuse hyperreflectivity, of retina, in acute 107 Hyaloid membrane, posterior, 21f
retinal necrosis syndrome, 132 for posterior scleritis, 128f Hydroxychloroquine sulfate, 68
Disciform scar, 36, 36f of type 1 choroidal neovascular membrane, Hydroxychloroquine toxicity, 68, 69f–71f
Dome-shaped lesion, in Candida chorioretinitis, 30 Hyperreflectivity, in myelinated nerve fiber layer,
129 Fovea 186, 187f
Dome-shaped macula, 64, 65f changes in, VMT associated with, 43f Hypertransmission signal, 24, 25f, 27f
Dominant drusen see Malattia leventinese focal adhesion over, VMT with, 45f Hypopigmentation, 24
Doyne’s honeycomb retinal dystrophy, 155, geographic atrophy, 25f Hyporeflective internal signal, for solitary
155f–156f multifocal, 26f choroidal hemangioma, 136
Drusen, 16 secondary to age-related macular
AMD and, 16
cuticular cluster-like, 23f
degeneration i, 25f
FTMH see Full-thickness macular hole I
hard, 16, 19f Full-thickness macular hole (FTMH), 47
soft, 16, 17f–19f abortive, in lamellar macular hole, 50 Idiopathic posterior uveitis, 166f
tomographic features of, 16 eccentric, superotemporal to the fovea, 49f Immunosuppression, systemic, for multifocal
Drusenoid pigment epithelial detachment (PED), large choroiditis and panuveitis and punctate
16, 20f–21f with VMT, 48f inner choroidopathy, 111
Dystrophy, vitelliform, 72, 72f without VMT, 48f Inactive CSCR, with thick choroid and large
medium, without VMT, 48f choroidal vessels, 57f

E small, 47f
without VMT, 47f
Indocyanine green angiography, for multiple
evanescent white dot syndrome, 107
vitreomacular abnormality in, 49f Infectious process, posterior scleritis and, 127
Eccentric FTMH, superotemporal to the fovea, Full-thickness retinal disorganization, in Inferotemporal retinal periphery, bullous
49f combined hamartoma, of retina and retinoschisis in, 181f
Edema, diabetic macular, 84, 84f–85f RPE, 139 Infiltration, of choriocapillaris, in choroidal
EDI see Enhanced depth imaging Fundus, 134 metastases, 141f

190
Inner segment/outer segment (IS/OS), 24 Maculopathy Optical coherence tomography angiography,
Intraretinal cyst, in lattice degeneration, hydroxychloroquine 1, 3f
184f advanced, 70f artifacts, 13
Intraretinal hemorrhages, from NPDR, 86 early, 69f–70f Optical coherence tomography (OCT), 16
Intraretinal microvascular abnormalities, from moderate, 71f artifacts and errors of, 10–12
NPDR, 86 laser, 145, 145f B-scan, 17f–23f

Index
Ischemic central retinal vein occlusion, 92 solar, 146, 146f–147f for birdshot retinochoroidopathy, 100
Isolated cystoid macular edema, 76, 77f Malattia leventinese, 155, 155f–156f of dome-shaped macula, 64, 65f
Isolated pigment epithelial detachment, 28 Melanoma, choroidal, 135, 135f for isolated pigment epithelial detachment,
macular cube scan in, 29f Metastases, choroidal, 140, 141f 28, 28f–29f
OCT features of, 28, 28f–29f Mild epiretinal membrane, 52 scanning protocols of, 7
Mild VMT, 42f–43f, 45f spectral domain, 5

L Mirror artifact, 10, 10f


Misalignment, artifact, 10, 10f
swept- source, 6, 6f
time-domain, 4
Moderate VMT, 45f for vitreous inflammation, 166, 167f–168f
Lamellar macular hole, 50, 51f Modified Fourier-domain, 6 Out of range error, 12, 12f
causes of, 50 Motion artifact, 10, 11f Outer retinal tubulation (ORT), 24
characteristic appearance of, 50 Müller cells, 62, 63f
features of, 50, 51f
in retina, 51f
Multifocal choroiditis and panuveitis (MCP),
111, 111f–113f P
Laser maculopathy, 145, 145f Multiple A-scans, 4
Laser photocoagulation, for solitary choroidal Multiple evanescent white dot syndrome Pain, in posterior scleritis, 127
hemangioma, 136 (MEWDS), 107, 108f Panuveitis, 111, 111f–113f
Lattice degeneration, 183 Mycobacterium tuberculosis, 124, 125f–126f Pars planitis, 77f
hyporeflective, 183, 185f Myelinated nerve fiber layer, 186 Patchy disruption, in acute syphilitic posterior
intraretinal cyst in, 184f extreme hyperreflectivity in, 186, 187f placoid chorioretinitis (ASPPC), 121
subclinical retinal detachment in, 183, inferonasal, 188f Perfluorocarbon, subretinal, 82, 83f
184f–185f retinal blood vessels in, 187f–188f Peripheral retinoschisis, 182f
vitreoretinal adhesion in, 183, 183f, 185f retinal thinning in, 186 Photodynamic therapy, for solitary choroidal
vitreoretinal traction in, 183, 184f Myopic choroidal neovascular membrane hemangioma, 136
Light echoes, using time-domain OCT, 4 (CNV), 59, 60f Pigment epithelial tear, retinal, 37, 37f
Line scans, for optic nerve, 1, 3f clinical appearance of, 61f Pigmented epithelium detachment, in central
Low-dose radiation, for solitary choroidal features of, 59 serous chorioretinopathy, 57f
hemangioma, 136 type 2 leakage pattern of, 61f Pisciform flecks, 149, 149f
Lung, choroidal metastases of, 140 Myopic ectatic retinopathy, 62 Polypoidal choroidal vasculopathy, 38,
Myopic macular schisis, 62, 62f–63f 38f–39f

M Posterior cortical vitreous, 9

N Posterior hyaloid membrane, 21f–22f


Posterior scleritis, 127, 127f–128f
Macula Posterior staphyloma, 66, 67f
central, in combined hamartoma, of retina Nasal macula, PFC liquid under, 83f Posterior uveitis
and RPE, 139 Nascent geographic atrophy, 24 with chorioretinitis, 121
diabetic tractional retinal detachment Necrosis syndrome, acute retinal, 132, idiopathic, 166f
involving, 172f 133f Posterior vitreous detachment, stages of, 159,
dome-shaped, 64, 65f Neovascular membrane, choroidal 159f–161f
superotemporal aspect of, 22f type 1, 30 Premacular bursa, 9, 9f
vitreomacular tract in, 42 type 2, 32, 33f Preretinal fibrosis, proliferative diabetic
Macula-off rhegmatogenous retinal type 3, 34, 34f retinopathy with, 172f
detachment, 175f–176f Nerve fiber layer see Retinal nerve fiber layer Projection artifacts, 13, 14f
Macular cube scan Nevus, choroidal, 134, 134f Proliferative diabetic retinopathy, 88, 88f–89f
of geographic atrophy, 26f Nonischemic central retinal vein occlusion, tractional retinal detachment from, 170,
of isolated pigment epithelial detachment, 92 171f–173f
29f Nonproliferative diabetic retinopathy (NPDR), Proliferative vitreoretinopathy, tractional retinal
Macular degeneration, age-related see Age- 86, 86f–87f detachment secondary to, 170, 173f
related macular degeneration Normal vitreous, 9 Pseudodrusen, reticular, 17f
Macular edema Nyctalopia, 148 Pseudohole, ERM associated with, 53f
diabetic, 84, 84f–85f Punctate inner choroidopathy (PIC), 111,
in retinal capillary hemangioma, 137
Macular hole O 111f–113f

full-thickness, 47, 47f–49f


lamellar, 50, 51f OCT see Optical coherence tomography Q
causes of, 50 Ocular albinism, 80
characteristic appearance of, 50 Oculocutaneous albinism, 80, 80f–81f Quilting defects, artifacts, 13, 14f
features of, 50, 51f Ophthalmia, sympathetic, 116, 117f–118f
in retina, 51f
Macular schisis, myopic, 62, 62f–63f
Optic nerve, normal, 1
line scans for, 1, 3f R
Macular telangiectasia, 73 morphology of, 1, 2f
type 1, 73, 73f–74f scan patterns for, 1 Radiation, for choroidal metastases, 140
type 2, 73, 74f–75f volume scans for, 1 Reticular pseudodrusen, 16, 17f–18f, 22f

191
Retina nonproliferative, 86, 86f–87f Subretinal fibrosis, in acute syphilitic posterior
diffuse hyperreflectivity of, in acute retinal proliferative, 88, 88f–89f, 170, 171f–173f placoid chorioretinitis (ASPPC), 123f
necrosis syndrome, 132 myopic ectatic, 62 Subretinal fluid
lamellar macular hole in, 51f nonproliferative diabetic, 86, 86f–87f in acute retinal necrosis syndrome, 132
RPE and, combined hamartoma of, 139, Valsalva, 142, 143f–144f in acute syphilitic posterior placoid
139f Retinoschisis chorioretinitis (ASPPC), 121
Index

Retinal artery occlusion bullous, 180, 182f in choroidal melanoma, 135, 135f
branch, 96, 97f in inferotemporal retinal periphery, 181f in choroidal metastases, 140
central, 98, 99f peripheral, 182f in choroidal nevus, 134
Retinal blood vessels, in myelinated nerve fiber retinal detachment and, 180f–182f in posterior scleritis, 127
layer, 187f–188f shallow, 182f in retinal capillary hemangioma, 137, 137f
Retinal capillary hemangioma, 137, 137f in superotemporal retinal periphery, in rhegmatogenous retinal detachment,
Retinal detachment 180f 174
bullous retinoschisis and, 180f–182f transition zone in, 181f in solitary choroidal hemangioma, 136
in combined hamartoma, of retina and RPE, X-linked juvenile, 79, 79f in tuberculosis, 124, 125f
139 Retrohyaloid space, 9, 9f Subretinal hemorrhage, 35, 35f
rhegmatogenous, 174 Rhegmatogenous retinal detachment, 174 Subretinal perfluorocarbon, 82, 83f
acute, 174 acute, 174 Superotemporal retinal periphery, bullous
chronic, 174, 179f chronic, 174, 179f retinoschisis in, 180f
macula-involving, 177f macula-involving, 177f Swept- source OCT, 6, 6f, 9, 9f
macula-off, 175f–176f macula-off, 175f–176f Sympathetic ophthalmia, 116, 117f–118f
microstructural changes in, 174 microstructural changes in, 174 Syphilis, 121
retinal pigment epithelium in, 174 retinal pigment epithelium in, 174
secondary to giant retinal tear, 178f
subretinal fluid in, 174
secondary to giant retinal tear, 178f
subretinal fluid in, 174 T
transition zone in, 179f transition zone in, 179f
serous, Vogt-Koyanagi-Harada disease and, RPE see Retinal pigment epithelium Telangiectasia, macular, 73
114 type 1, 73, 73f–74f
subclinical, in lattice degeneration, 183,
184f–185f S type 2, 73, 74f–75f
Time-domain OCT, 4
tractional, 170 Tissue, loss of, in acute retinal necrosis
from proliferative diabetic retinopathy, 170, Scan patterns, for optic nerve, 1 syndrome, 132
171f–173f Scar, disciform, 36, 36f Toxicity, hydroxychloroquine, 68, 69f–71f
secondary to proliferative vitreoretinopathy, Schisis, 180 Toxoplasma gondii, 119
170, 173f between inner and outer retinal layers Toxoplasmic chorioretinitis, 119, 119f–120f
subretinal fluid in, 170, 172f epiretinal membrane with, 53f Tractional retinal detachment, 170
type and location of, 170 Sclera, 127 from proliferative diabetic retinopathy, 170,
Retinal necrosis syndrome, acute, 132, 133f Scleritis, posterior, 127, 127f–128f 171f–173f
Retinal nerve fiber layer (RNFL), 1 SD-OCT see Spectral domain OCT secondary to proliferative vitreoretinopathy,
thickness, 1, 2f Secondary CNV, central serous 170, 173f
Retinal occlusive vasculitis, in acute retinal chorioretinopathy with, 58f subretinal fluid in, 170, 172f
necrosis syndrome, 132 Segmentation errors, artifacts, 13, 15f type and location of, 170
Retinal pigment epithelial changes, in solitary Serous chorioretinopathy, central, 7 Transchoroidal mass, elevated, in choroidal
choroidal hemangioma, 136 Serous retinal detachment, in acute retinal melanoma, 135f
Retinal pigment epithelial tear, 37, 37f necrosis syndrome, 132 Transition zone
Retinal pigment epithelium (RPE), 5, 5f, 16 Serpiginous choroiditis, 109, 110f in bullous retinoschisis, 181f
acute posterior multifocal placoid pigment Severe VMT, 45f in rhegmatogenous retinal detachment, 179f
epitheliopathy and, 103 Shadowing artifacts, 13, 15f Trauma, mechanical, Valsalva retinopathy, 142,
loss over angioid streaks, 78, 78f “Shaggy” photoreceptors, in choroidal 143f–144f
multifocal geographic atrophy with, 24, melanoma, 135f Treponema pallidum, 121
26f–27f Shallow retinoschisis, 182f Tuberculosis, 124, 125f–126f
posterior scleritis and, 127 Signal strength, poor, and Candida Type 1 choroidal neovascular membrane, 30,
in rhegmatogenous retinal detachment, chorioretinitis, 129 31f
174 Simple hamartoma, of retinal pigment Type 1 macular telangiectasia, 73, 73f–74f
simple hamartoma of, 138, 138f epithelium (RPE), 138, 138f Type 2 choroidal neovascular membrane, 32,
Retinal separation, 180 Small FTMH, 47f 33f
Retinal thinning Soft drusen, 16, 17f–19f Type 2 leakage pattern, of myopic CNV, 61f
in acute retinal necrosis syndrome, 132 Software breakdown, artifacts, 10, 11f Type 2 macular telangiectasia, 73, 74f–75f
in myelinated nerve fiber layer, 186 Solar maculopathy, 146, 146f–147f Type 3 choroidal neovascular membrane, 34,
Retinal vein occlusion Solitary choroidal hemangioma, 136, 136f 34f
branch, 90, 91f Spectral domain OCT (SD-OCT), 5, 5f, 7, 9, 9f
central, 92, 93f–95f
Retinitis pigmentosa, 148, 148f
Split retina, 180
Staphyloma, posterior, 66, 67f U
Retinochoroidopathy, birdshot, 100, 100f–102f, Stargardt disease, 149, 149f–151f
169f Steroid, for multifocal choroiditis and panuveitis Uveitis
Retinopathy and punctate inner choroidopathy, 111 in acute retinal necrosis syndrome, 132
diabetic Streaks, angioid, 78, 78f bilateral, Vogt-Koyanagi-Harada disease and,
in Candida chorioretinitis, 129f Subfoveal fluid, VMT with associated, 43f 114
in diabetic macular edema, 84 Subretinal drusenoid deposits (SDDs), 16 idiopathic posterior, 166f

192
V Vitreoretinal adhesion, in lattice degeneration,
183, 183f, 185f
VMT see Vitreomacular traction
Vogt-Koyanagi-Harada disease, 114, 114f–115f
Vitreoretinal interface forces, in combined Vogt-Koyanagi-Harada syndrome, 103
Valsalva retinopathy, 142, 143f–144f hamartoma, of retina and RPE, 139 Volume scans, for optic nerve, 1
Vein occlusion, central retinal, 92, 93f–95f Vitreoretinal traction, in lattice degeneration, von Hippel Lindau syndrome, 137
Vessel duplication, artifacts, 13, 14f 183, 184f

Index
Vignetting, artifacts, 10, 10f
Vitelliform dystrophy, 72, 72f
Vitreoretinopathy, proliferative, tractional
retinal detachment secondary to, 170, W
Vitreomacular adhesion, 40, 40f–41f 173f
Vitreomacular traction (VMT), 42, 46f Vitreous, 159 White line artifacts, 13, 14f
with associated subfoveal fluid, 43f normal, 9 “Wolf-jaw” configuration, in tractional retinal
associated with changes in fovea, 43f Vitreous detachment, posterior, stages of, 159, detachment, 171f, 173f
degree of, 42 159f–161f
in eyes, 44f
features of, 42
Vitreous hemorrhage, 164, 165f
diabetic, 164f X
with focal adhesion, over fovea, 45f Vitreous inflammation, 166, 167f–168f
large FTMH with, 48f in acute retinal necrosis syndrome, 132 X-linked juvenile retinoschisis (XLJR), 79, 79f
mild, 42f–43f, 45f cystoid macular edema and, 166, 166f, 169f XLJR see X-linked juvenile retinoschisis
moderate, 45f idiopathic posterior uveitis and, 166f
severe, 45f optical coherence tomography for, 166,
vitreomacular abnormality in, 49f 167f–168f

193
This page intentionally left blank
This page intentionally left blank
This page intentionally left blank
Fast answers and trusted evidence

Drive better outcomes with a clinical search engine


that makes it easier to find and apply relevant knowledge.

Fast Convenient
Anticipates your query, recognizing Accessible at the patient’s bedside or
concepts and offering shortcuts on the go, making it easy to discover,
share, and apply content anywhere
Complete
Draws relevant answers from a wide Trusted
range of current, comprehensive Content from Elsevier, the
content across 30+ medical and name healthcare professionals
surgical specialties worldwide rely on

ckey.co/books

You might also like