You are on page 1of 113

Albert J.

Augustin
Editor

Intravitreal Steroids

123
Intravitreal Steroids
Albert J. Augustin
Editor

Intravitreal Steroids
Editor
Albert J. Augustin
Stdt. Klinikum Karlsruhe, Augenklinik
Karlsruhe, Germany

ISBN 978-3-319-14486-3 ISBN 978-3-319-14487-0 (eBook)


DOI 10.1007/978-3-319-14487-0
Springer Cham Heidelberg New York Dordrecht London

Library of Congress Control Number: 2015932097

Springer International Publishing Switzerland 2015


This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or
part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way,
and transmission or information storage and retrieval, electronic adaptation, computer software,
or by similar or dissimilar methodology now known or hereafter developed. Exempted from this
legal reservation are brief excerpts in connection with reviews or scholarly analysis or material
supplied specifically for the purpose of being entered and executed on a computer system, for
exclusive use by the purchaser of the work. Duplication of this publication or parts thereof is
permitted only under the provisions of the Copyright Law of the Publishers location, in its
current version, and permission for use must always be obtained from Springer. Permissions for
use may be obtained through RightsLink at the Copyright Clearance Center. Violations are liable
to prosecution under the respective Copyright Law.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
While the advice and information in this book are believed to be true and accurate at the date of
publication, neither the authors nor the editors nor the publisher can accept any legal responsibility
for any errors or omissions that may be made. The publisher makes no warranty, express or
implied, with respect to the material contained herein.

Printed on acid-free paper

Springer is part of Springer Science+Business Media (www.springer.com)


Preface

Corticosteroids are well known for their antiangiogenic, antiedematous, and


anti-inflammatory properties. Consequently, the drugs have been widely used
in the treatment of many retinal diseases since the early 1950s. Later,
intravitreal steroids emerged as an essential tool, allowing for optimal drug
efficacy when given locally. The topical or local route offers the great
advantage that there is no significant systemic side effect.
Randomized studies have properly shown that steroids are very effective
in the treatment of posterior segment diseases such as retinal vascular
occlusion, diabetic macular edema, and uveitis. This is true for both, disease
activity and visual outcome. Today because of the anti-inflammatory,
antiangiogenic, and antipermeability properties, steroids are an attractive
therapeutic option for vascular and inflammatory retinal diseases.
Recent advances in ocular drug delivery methods led to the development
of intraocular implants, which help to provide prolonged treatment with a
continuous drug release. An increasing number of ophthalmologists use
intravitreal steroids for the treatment of various posterior segment disorders,
not only when traditional therapeutic methods have failed, but more and more
as a first-line therapy.
Through careful selection of cutting edge authors and timely subjects, to
the fine details of quality editing and illustrations, we have created a most
useful book. In this book the reader is treated to an exceptional update on
current topics concerning the pathophysiology and clinical value of intravit-
real steroids.
The depth and breadth of the information presented is intended to bring
the reader the newest diagnostic and therapeutic approaches in our rapidly
advancing field.

Karlsruhe, Germany Albert J. Augustin, MD

v
Contents

1 Pathophysiology of Macular Edema: Results


from Basic Research . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Ana Bastos-Carvalho and Jayakrishna Ambati
2 Pathophysiology of Macular Edema in Diabetes,
Retinal Vein Occlusion, and Uveitis:
A Disease-Related Approach. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
Edoardo Midena and Silvia Bini
3 Devices for the Delivery of Steroids to the Eye . . . . . . . . . . . . . . 25
Raja Narayanan and Baruch D. Kuppermann
4 Imaging Before, During,
and After Steroid Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
Antonio Caimi and Giovanni Staurenghi
5 Treatment of DME with Steroids . . . . . . . . . . . . . . . . . . . . . . . . . 55
Couturier Aude and Pascale Massin
6 Intravitreal Steroids for the Treatment of Macular
Edema in Retinal Vein Occlusions . . . . . . . . . . . . . . . . . . . . . . . . 69
Eran Zunz and Anat Loewenstein
7 Treatment of Uveitis with Intraocular Steroids . . . . . . . . . . . . . 81
Lazha Talat, Filis Ismetova, Susan Lightman,
and Oren Tomkins-Netzer
8 Steroids in a Combination Strategy . . . . . . . . . . . . . . . . . . . . . . . 91
Paolo Lanzetta, Daniele Veritti, and Valentina Sarao
9 Intravitreal Steroids as a Surgical Adjunct . . . . . . . . . . . . . . . . . 105
Stanislao Rizzo, Tomaso Caporossi, and Francesco Barca

vii
Pathophysiology of Macular
Edema: Results from Basic 1
Research

Ana Bastos-Carvalho and Jayakrishna Ambati

Contents 1.1 Introduction


1.1 Introduction 1
Macular edema (ME) is the final common path-
1.2 Anatomy of the Macula 1
way of numerous retinal diseases, and ocular dis-
1.3 Definition of Macular Edema 2 orders associated with this condition are, when
1.4 The Blood-Retinal Barrier 2 considered together, a major cause of blindness
1.4.1 Inner BRB 3 in the Western world [1]. In a physiological set-
1.4.2 Outer BRB 7 ting, the blood-retinal barrier (BRB), which is
1.5 Physical Mechanisms Determining largely formed by the retinal pigment epithelium
Fluid Absorption/Retention in the and the retinal capillary endothelial cells, blocks
Retina 9
the passage of fluid and potentially harmful
1.6 Concluding Remarks 10 blood-borne molecules into the retina. When this
References 10 barrier is broken, water and proteins can enter the
retinal extra- and intracellular space, with fluid
accumulation leading to edema. Frequently, BRB
leakage occurs at the macula, causing ME and
vision loss.

1.2 Anatomy of the Macula

Anatomically, the macula (area centralis) is


defined as the central retinal region containing
more than one layer of ganglion cells and pre-
A. Bastos-Carvalho, MD (*)
senting xanthophyll pigment, which is located
Department of Ophthalmology and Visual Sciences,
University of Kentucky, 1095 VA Drive HSRB 252, mainly in the layer of the fibers of Henle and in
Lexington, KY, 40536, USA the inner nuclear layer [2]. At its center lies the
e-mail: aba253@uky.edy fovea, a 1.5 mm depression that is devoid of gan-
J. Ambati, MD glion cells and thinnest at its central area, the
Department of Ophthalmology and Visual Sciences, umbo. The macula has several anatomical fea-
University of Kentucky, 740 S. Limestone Street,
tures that diverge from the remaining retina;
Lexington, KY, 40536, USA
e-mail: jamba2@email.uky.edu, these include a high cell density with almost no
jayakrishna.ambati@uky.edu extracellular space [3], the loose arrangement of

A.J. Augustin (ed.), Intravitreal Steroids, 1


DOI 10.1007/978-3-319-14487-0_1, Springer International Publishing Switzerland 2015
2 A. Bastos-Carvalho and J. Ambati

thick fibers in Henles layer, and the avascularity Table 1.1 Conditions associated with macular edema
of its central zone, which is a watershed zone Etiology of macular edema
between the choroidal and retinal circulations Vascular diseases of the posterior segment
primarily supplied by the choriocapillaris. These Diabetic retinopathy (DR)
unique anatomical characteristics of the macula, Neovascular age-related macular degeneration (AMD)
combined with its high metabolic demand, make Retinal venous occlusion (RVO)
it a preferential site for fluid accumulation and Retinopathy of prematurity (ROP)
edema [4]. Radiation retinopathy
Hypertensive retinopathy
Macular telangiectasias type 2 (MacTel2)
Retinal arterial macroaneurysm
1.3 Denition of Macular Edema
IRVAN syndrome
Choroidal neovascularization of other causes
Macular edema is broadly defined as the retinal Inflammatory diseases
accumulation of fluid in the macula. Histologically, Postsurgical (intraocular surgery; laser procedures)
fluid accumulation may occur in the intercellular Uveitis
space or may be intracellular, causing cellular Drug induced
swelling, which most commonly occurs with Systemic drugs
Mller cells [5]. Clinically, ME can be classified Thiazolidinediones (glitazones)
as diffuse, with generalized leakage throughout Taxanes (docetaxel, paclitaxel)
the posterior pole, or focal, if discrete areas of Tamoxifen
retinal thickening are present (usually caused by Niacin (nicotinic acid)
leakage from microaneurysms or dilated capillar- Interferon-, interferon-, and interferon-
ies). This classification is mostly used in diabetic Topical drugs
ME where recognition of these forms has impor- Prostaglandin analogs
tant prognostic value [6]. In cystoid ME, clear Epinephrine
fluid-filled cystoid spaces are present in the retina, Inherited dystrophies
Autosomal dominant cystoid macular edema
most commonly in the outer plexiform and inner
(ADCMO)
nuclear layers, and they are visible by ophthal- Retinitis pigmentosa
moscopy, OCT, and/or fluorescein angiography, Tumors
which shows a typical petaloid pattern of fluores- Choroidal melanoma
cein leakage surrounding the fovea [7]. Retinal or choroidal hemangioma
Microcystic macular edema (MME) is a subtype Retinal detachment
of cystoid ME with microcysts in the inner Tractional disorders
nuclear layer. MME has often been associated Epiretinal membrane
with multiple sclerosis (with or without optic Vitreoretinal traction syndrome
neuritis) but can be found in numerous ocular Diseases of the optic nerve head
conditions [8]. ME can also be classified as acute Optic pit
or chronic, depending on the time elapsed since Optic coloboma
the diagnosis. Other clinically relevant parame- Optic neuropathy
ters in evaluating ME are the geographic extent
of the edema (i.e., the area that shows increased
retinal thickness), degree of foveal involvement, 1.4 The Blood-Retinal Barrier
signs of leakage in fluorescein angiography, asso-
ciation with retinal ischemia, and presence of The outer retina receives oxygen and nutrients
vitreous traction [9]. from the blood in the choriocapillaris, whereas
A myriad of ocular conditions can be associ- the retinal vessels, branches of the central retinal
ated with ME, summarized in Table 1.1 and dis- artery, supply the inner retinal layers. Similar to
cussed in further detail in Chap. 2. the blood-brain barrier (BBB) elsewhere in the
1 Pathophysiology of Macular Edema: Results from Basic Research 3

Fig. 1.1 Schematic


diagram of cell types that
form the inner
BRB. Retinal vascular
endothelial cells present
Mller
tight junctions that regulate cell
the permeability of the
paracellular pathway.
Pericytes closely envelop
the capillaries, and
astrocytes and Mller cells Astrocyte
extend their processes to
surround the retinal
microvasculature
Pericyte

Tight
junctions Endothelial cell

Basement membrane
Retinal capillary

central nervous system (CNS), the eye possesses a transcytosis (vesicular transport), and solute modi-
pair of mechanisms together named the blood- fication (degradation or modification of solutes
ocular barrier that allow separation between before these molecules pass the barrier) [11].
blood components and nervous tissues. This bar- Below, we elaborate on each of these components
rier has two components: the blood-aqueous bar- and their role in the inner and outer BRB.
rier (BAB) and the blood-retinal barrier (BRB).
At the level of the BAB, plasma proteins are pre-
vented from crossing into the aqueous and vitre- 1.4.1 Inner BRB
ous humor by the endothelium of the iris and
ciliary muscle capillaries, as well as by the non- The inner component of the BRB is formed by
pigmented ciliary epithelium [10]. The BRB is the the retinal capillaries, which are distributed in
major player maintaining the posterior segment two plexuses: the inner vascular plexus, at the
tissues, principally the retina and subretinal space, ganglion cell and nerve fiber layers, and the outer
in a state of relative deturgescence. It can be sub- vascular plexus, between the inner nuclear and
divided into an inner BRB (the retinal capillaries) outer plexiform layers [12]. Retinal capillaries
and an outer BRB (at the level of the retinal pig- are composed of a single layer of endothelial
ment epithelium). In this chapter, we will focus on cells (ECs), a basement membrane, and peri-
the BRB component of the ocular barrier, as it is cytes. These cells, together with the surrounding
the main factor influencing the movement of flu- glial cells, form the retinal neurovascular unit,
ids to and from the retina and subretinal space which is responsible for the meticulous regula-
and, hence, the absence or formation of ME. tion of retinal blood flow and BRB integrity [13]
As with any blood-tissue barrier, there are five (Fig. 1.1). Retinal ECs are phenotypically dis-
components at interplay in the BRB, namely, tight tinct from other capillary ECs and similar only to
junctions (TJs) between the cells of the monolayer those found in the BBB. This continuous endo-
that comprises the barrier, facilitated diffusion thelium is devoid of fenestrations and surrounded
(through channels in the plasma membrane), by a thick basement membrane; it expresses
active transport (through ATP-consuming pumps), TJs surrounding every component cell and is
4 A. Bastos-Carvalho and J. Ambati

maintained and regulated by a high pericyte-to- former to the actin cytoskeleton include ZO-1
EC ratio. Below, we elaborate further on each of and cingulin [21, 22]. Modulation of TJ protein
these features. expression and phosphorylation are two of the
A healthy BRB allows the passage of lipid- major regulators of paracellular permeability and
soluble substances [14, 15] but is impermeable to have been demonstrated in several animal models
fluorescein [16]. The BRB maintains a tightly of retinal vessel hyperpermeability. In diabetic
regulated separation between the blood compo- rats, occludin expression in retinal ECs is
nents and the retina, and selective transport of decreased [21, 23], and insulin treatment reverses
glucose, amino acids, small ions, and retinal this phenotype [24]; and in high glucose condi-
metabolites, as well as leukocytes, can take place tions, human retinal ECs decrease their expres-
through two pathways: the paracellular pathway, sion of occludin, JAM-A, and claudin-5 [25].
formed by inter-endothelial junctions, and the Additionally, occludin levels are also reduced
transcellular route, mediated by vesicles, carri- [23, 26] in models of vascular retinopathy
ers, transmembrane channels, and receptors [17]. induced by VEGF a cytokine that is present in
When one or more of these mechanisms is dis- increased quantities in the vitreous of diabetic
turbed, fluid may accumulate in the retinal inter- patients and is known to be a vascular permeabil-
stitium, resulting in edema. ity factor. Exposing retinal ECs to the corticoste-
roid hydrocortisone a drug class used in the
1.4.1.1 EC Paracellular Pathway treatment of ME of various etiologies also
Endothelial cells in the retina are tightly joined increased expression of occludin and decreased
through specialized structures: tight junctions, transport of water and solutes across the EC
adherens junctions, and gap junctions, which monolayer [27]. Similarly, in a porcine model of
together form the junctional complex. The passage branch retinal vein occlusion (BRVO), treatment
of fluid and molecules through these intercellular with triamcinolone acetonide another cortico-
spaces constitutes the paracellular pathway. Aside steroid used for management of ME caused a
from the major function of regulating capillary decrease in VEGF expression and an increase in
permeability, ECs junctions also play important occludin levels in the retina [28]. These results
roles in modulation of angiogenesis and apoptosis, suggest that the mechanism of action of cortico-
contact inhibition of proliferation, and response to steroids in ME may be a reduction in available
blood flow and leukocyte diapedesis [18]. While VEGF and a subsequent increase in occludin
both tight and adherens junctions are constituted expression and localization at the TJs.
by transmembrane proteins arranged in a zipper- Adherens junctions (AJs) are composed of
like structure along the cell border [18], the basic VE-cadherin membrane proteins and several
organization of adherens junctions is determined intracellular partners, including b-catenin, p120-
by VE-cadherin [19], whereas claudins and occlu- catenin, and plakoglobin [19]. AJs have been
din form TJs [20]. These transmembrane proteins shown to modulate TJs through VE-cadherin-
interact with cytoskeletal proteins inside the cell, dependent overexpression of claudin-5 [29], which
to which they are anchored. This allows tight and offers a possible explanation for the close associa-
adherens junctions to open and close and facili- tion and intermingling of these two types of junc-
tates intracellular signaling. tional complexes at the cell membrane.
Tight junctions (TJs) are present in high num- Gap junctions are cell membrane hemi-
bers in retinal ECs and are composed of mem- channels found in retinal ECs [30] and formed by
brane proteins, adaptor/cytoskeletal proteins, and the assembly of six connexins, whose functions
effector proteins. TJ membrane proteins that have include maintenance of the BRB and intercellular
been identified in retinal ECs include occludin (endothelial-endothelial or pericyte-endothelial)
[20], claudin-1, claudin-2, claudin-5, and junc- communication [31].
tional adhesion molecule-A (JAM-A), whereas Also worthy of note is the role of fenofi-
intracellular adaptor proteins that couple the brate a peroxisome proliferator-activated
1 Pathophysiology of Macular Edema: Results from Basic Research 5

receptor- (PPAR-) agonist widely prescribed respectively [4345]. While the previously
as a cholesterol-reducing agent in BRB per- referred transporters allow passage of nutrients
meability, since this agent has been linked to a into the retina, elimination of metabolites is
decreased risk of ME in diabetic patients [32]. achieved primarily via active efflux transport
Consistent with this clinical evidence, oral feno- systems, mostly mediated by ATP-binding cas-
fibrate supplementation ameliorates retinal vas- sette (ABC) transporters and solute carrier
cular leakage in rat and mouse models of type (SLC) transporters (reviewed in [46]).
1 diabetes [33]. Further studies are warranted
to determine the pathway by which this drug is Vesicle Transport
modulating permeability in the inner BRB of dia- The transport of macromolecules, like albumin,
betic patients. through the inner BRB, is tightly regulated so as to
maintain a low oncotic pressure in the retinal tis-
1.4.1.2 EC Transcellular Pathway sue. This type of molecules can be shuttled in a
This pathway involves the transport of molecules process called transcytosis, defined as the trans-
through the endothelial cells and may occur by port of molecules across a cell via plasmalemmal
passive diffusion or mediated via receptors, carri- vesicles, or caveolae [17]. Macromolecules are
ers, or vesicles (caveolae), the last being taken up by invagination of one of the sides of the
employed for the transport of macromolecules. cell membrane. Thereafter, fission of the invagi-
nated membrane portion forms the caveola, which
Receptors and Carriers then fuses with the opposite cell membrane and
Transport of nutrients and metabolites across the releases the vesicular contents. Macromolecules
BRB is done primarily via carriers, and these can can be carried in the fluid phase of caveolae or
mediate facilitated transport, primary active efflux, bound to receptors present at the membrane of
or secondary active influx and efflux [34]. Influx these vesicles. Albumin, insulin, lipoproteins, and
transporters allow passage of hydrophilic sub- VEGF receptors, among others, have been identi-
stances, like D-glucose, amino acids, vitamins, fied in caveolae (reviewed in [47]). Numerous
and nucleosides into the retina, and do so at the studies indicate that vesicle-mediated transcytosis
high rates necessary to sustain this tissues high may be partially responsible for the BRB hyper-
metabolic demand. One such transporter is permeability seen in condition associated with
GLUT1/SLC2A1, which transports D-glucose, ME. Indeed, VEGF has been shown to induce
the main energy source of the retina, and dehydro- transcytosis [48] and to modulate the abundance
ascorbic acid (reviewed in [35]). Regulation in the and location of vesicles in retinal endothelial cells,
expression of GLUT1 has been shown to occur in thereby modulating vascular permeability [49].
the retinal vasculature of diabetic rats by some Consistently, there is an overexpression of plasma-
groups [36, 37], but other reports suggest that the lemmal vesicle-associated protein 1 (PLVAP), a
expression of this transporter is not changed in caveola-associated protein, at the sites of leakage
retinal vessels of diabetic retinopathy in the same in DR patients and in primate VEGF-induced reti-
rat model [23] or in humans with diabetic retinop- nopathy [50]. Further, in a mouse model of
athy (DR) [38]. Interestingly, ECs of neovessels in autoimmune-induced uveitis, vesicular transport
proliferative DR do not express GLUT1 [38], plays a fundamental role in protein exudation to
which is consistent with the notion that this trans- the retina, whereas EC TJs are relatively conserved
porter is characteristic of barrier tissues [39]. [51]. The implications of transcytosis dysregula-
Other influx transporters in the retina include tion in diabetic retinopathy, not only for molecular
the L-lactic acid transporter MCT1 [40]; CRT, transport (as discussed above) but also for leuko-
which transports creatine [41, 42]; and the cyte diapedesis, are thoroughly reviewed by
amino acid transporters LAT1, system Xc, and Klaassen and colleagues [17].
CAT1, which are responsible for the transport of Recently, downregulation of the protein
large amino acids, L-cystine and L-arginine, Msfd2a in ECs has been implicated in increased
6 A. Bastos-Carvalho and J. Ambati

vascular permeability in the BBB, which is asso- several features characteristic of diabetic retinop-
ciated with increased transcytosis and no evident athy [62]. Another important signaling pathway
changes in EC TJs [52]. Future studies focusing between pericytes and ECs is the angiopoietin1
on regulation of this protein in the retina may (Ang1; ligand)/Tie2 (receptor) system. Pericytes
indicate whether this factor also has a role in express Ang1, which binds to Tie2 in ECs [63],
increased BRB permeability and ME. and this signaling promotes vessel formation and
maturation [64]. Ang1 signaling can also induce
1.4.1.3 Other Cell Types expression of occludin [65] and decrease vascular
Pericytes and glial cells can modulate the perme- leakage in ischemia [66]. Therefore, disruptions
ability of the inner BRB by transmitting regula- of this signaling pathway may lead to increased
tory signals to endothelial cells [53]. vessel leakiness. Indeed, Ang1 can block VEGF-
Pericytes are a part of the neurovascular reti- induced vessel leakage by VEGF, by prevent-
nal unit and contribute to the regulation of EC ing Src activation and consequent VE-cadherin
function and blood flow in the nervous system phosphorylation and redistribution [67]. Ang1/
[54], thereby playing an important role in the mice also display defects in embryonic vascular
inner BRB. Pericytes are separated from ECs by arrangement, including a loss of association with
a basement membrane (BM) but can communi- periendothelial cells (such as cells that differenti-
cate through holes, or gap junctions, in this lam- ate into pericytes) and with the matrix [68]. In
ina. Retinal capillaries present the highest known contrast, Ang2 induces increased permeability
EC-to-pericyte ratio (1:1) and have a pericyte by binding to Tie2 and preventing Ang1 interac-
coverage of 8595 % [55, 56], which supports the tion with this receptor. Ang2 is increased in the
notion that high EC-to-pericyte ratios are pres- vitreous of diabetic patients [69] and diabetic
ent in tight inter-endothelial junctions and blood mice, where it induces pericyte loss by apopto-
barriers [57]. Indeed, pericytes have been shown sis [70]. Further, intravitreous administration of
to decrease permeability to tracers and increase this molecule promotes vessel leakage in rats,
trans-endothelial electric cell resistance (TEER; a and treatment of human retinal ECs with Ang2
measure of EC permeability) in an in vitro model reduces expression of VE-cadherin and increases
of inner BRB [58]. Moreover, pericytes regulate monolayer permeability [71]. The data above
the expression of Mfsd2a a major BBB regu- suggests that a balance in the Ang1/Ang2/Tie2
latory protein in ECs [52], which may facili- pathway maintains vascular stability and perme-
tate BRB integrity. Importantly, the formation of ability at physiological levels, and an imbalance
EC TJs during retinal embryonic development in this system can increase vessel leakage by loss
is closely related to pericyte recruitment, both of EC-associated pericytes and disruption of tight
spatially and temporally [59]. Pericyte loss is a junctions.
hallmark of early diabetic retinopathy [60], and Retinal glial cells that participate in the neu-
some evidence suggests that this loss may induce rovascular unit include astrocytes, Mller cells,
BRB leakage in diabetes. Cocultures of ECs with and microglia. Astrocytes and Mller cells are
pericytes and glial cells show a higher TEER macroglial cells that form a sheath around the
and less permeability of tracers than ECs cul- retinal capillaries and are separated from ECs
tured alone [58]. Pericytes seem to influence both by the BL. As with pericytes, astrocytes and
paracellular and transcellular pathways. Indeed, Mller cells also enhance retinal ECs barrier
animal and in vitro models of pericyte deficiency function (as measured by TEER, inulin flow,
associated with increased permeability in the and permeability to tracers [58, 72, 73]), and the
BBB show both TJ morphological changes and recruitment of these glial cells is related to an
an increase in cytoplasmic vesicles and uptake increase in expression of ZO-1, a TJ protein, in
of biotin by endocytosis [61]. Additionally, mice developing retinal vessels [59]. Mller cell con-
with endothelium-specific ablation of PDGF-B, tributions to the BRB have been linked to their
which induces loss of capillary pericytes, develop secretion of glial cell line-derived neurotrophic
1 Pathophysiology of Macular Edema: Results from Basic Research 7

Fig. 1.2 Outer BRB Retina


constituents. Tight junctions
between the retinal pigment
epithelial (RPE) cells modulate
the flow of fluid and molecules
from the subretinal space into RPE Cell
the choroidal circulation. Other
mechanisms regulating the
passage of substances through Tight
the RPE barrier include junctions
vesicle-mediated transcytosis
Transcytosis
and transport via receptors or
carriers (not depicted)
Choroid

factor (GDNF) [74] and pigment epithelium- intercellular TJs and pump functions. The RPE
derived factor (PEDF) [75, 76]. Additionally, is a monolayer of cuboidal pigmented cells,
Mller cells clear fluid from the retinal intersti- located between the photoreceptors (PRs) in the
tium by transporting water to the bloodstream neural retina and the fenestrated vessels in the
through their perivascular processes via con- choriocapillaris. The outer retina is supplied by
stitutive integral membrane aquaporin (AQ) the choriocapillaris, which means nutrients
channels. This fluid flux is coupled with potas- from the bloodstream have to pass the RPE bar-
sium currents mediated by Kir channels [77]. rier to reach the PRs. Additionally, the end prod-
Dysfunction or dysregulation of these chan- ucts resulting from the high metabolism of the
nels may lead to fluid accumulation and retinal outer retina, as well as water and ions, are trans-
edema [78]. Consistently, Mller cell down- ported from the subretinal space to the blood-
regulation of Kir4.1 and AQ4 was reported in stream by the RPE.
animal models of retinopathies that frequently As with the inner BRB, the outer barrier has
associate with ME, including diabetes [79], both paracellular and transcellular transport com-
ocular inflammation [80, 81], and retinal venous ponents, regulated by the RPE TJs and vesicles,
occlusion [82]. Recently, intracellular edema respectively (Fig. 1.2). A possible functional and
of Mller cells has been described in ME, and regulatory link between RPE transcellular and
it may be linked to the dysregulation of AQ4, paracellular pathways has been suggested [85].
which causes osmotic swelling of the cells [83], In this work, Rajasekaran et al. report that
as well as to other factors such as the extrava- inhibiting ion channel transport, a transcellular
sation of albumin from leaky vessels (reviewed function, results in the increase of TJ permeabil-
in [5]). Intriguingly, contrary to this physiologi- ity. Importantly, as with the inner BRB, the outer
cal function of fluid absorption and transport, BRB can be disrupted in diseases associated with
under hypoxic conditions Mller cells can pro- ME, as demonstrated in subsets of patients with
mote vascular permeability via stabilization DR and subretinal leakage [86, 87] and in animal
of hypoxia-inducible factor 1- (HIF1-) and models of DR and retinopathy of prematurity
production of angiopoietin-like 4 [84], which [88, 89].
suggests that retinal glial cells exert opposing
roles in healthy and diseased states. 1.4.2.1 RPE Paracellular Pathway
Similar to the ECs paracellular pathway, the
RPE junctional complex is constituted by tight
1.4.2 Outer BRB junctions, adherens junctions, and gap junctions.
The junctional complex lies on the apical
The outer BRB is formed by the retinal pigment (PR-facing) side of RPE cells, separating this
epithelium (RPE) cells, namely, by its aspect from the basolateral side. The apical
8 A. Bastos-Carvalho and J. Ambati

membrane of the pigmented cells has long [99], endoplasmic reticulum (ER) stress [100],
microvilli that interdigitate with the overlying IL-1 [101], TNF- [102], or hepatocyte growth
PRs, the space between the RPE and the PRs factor [103] regulate several RPE TJ compo-
being filled by interphotoreceptor matrix. This nents, including claudins and occludin. The
close contact may be a factor that allows the neu- effect(s) of VEGF (a growth factor produced by
ral retina to regulate the function of the RPE by RPE cells and known to be upregulated in DR) on
modulating the expression and localization of TJ the regulation of TJs remains controversial.
proteins [90]. While Peng et al. have shown that this cytokine
As in other epithelial cells, proteins that form has minimal effects on RPE TJs [104],
TJs in the RPE can be grouped in three catego- Ghassemifar and colleagues report an increase in
ries membrane, adaptor, and effector proteins ZO-1 expression and in TEER in RPE cell lines
and include the families of claudins and cadherins, supplemented with VEGF [105], and Ablonczy
occludin, JAMs, ZO-1, and many others (for an and Crosson describe a breakdown of the outer
excellent review on RPE tight junctions, see barrier induced by apically administered VEGF
[11]). In the RPE, the most studied of these pro- in vitro [106]. In a similar line, DAmores group
teins are claudins and occludin. Both are constit- showed that soluble VEGF reduces RPE barrier
uents of TJ strands that regulate their permeability properties in RPE-EC cocultures and that these
and selectivity. properties can be partially recovered by neutral-
izing VEGF [107]. As for placental growth factor
Claudins 1 (PlGF-1), another member of the VEGF super-
Twenty-four different claudins have been identi- family, its in vitro supplementation decreases
fied so far, and they have different patterns and RPE TEER, and in vivo administration causes
levels of expression depending on the cell, tissue, sub- and intraretinal edema [108]. From the data
and species [91]. Claudins are responsible for reviewed above, there may be a role of cytokines
paracellular selectivity to small ions, and their that are overexpressed in hypoxic conditions in
properties vary between each member of the fam- increasing TJ permeability and inducing subreti-
ily and their polymerization profile [92]. In the nal accumulation of water. Fenofibrate, also dis-
human RPE, claudin-3 and claudin-19 have been cussed above in Sect. 1.4.1.1, may also decrease
identified, while the presence and role of claudin- the risk of diabetic ME by reestablishing the
16 is still a matter of controversy [11, 93, 94]. In outer BRB properties of RPE cells. Although fur-
the RPE, TJ integrity and claudin expression are ther studies are warranted, Villarroel et al. report
regulated by miRNA 204/211 [93]. that treatment with fenofibric acid reverts the dis-
ruption of the RPE monolayer and the increase in
Occludin permeability induced by high glucose and IL-1
Like claudins, occludin is a transmembrane pro- [109], while Trudeau and colleagues report that
tein present in RPE TJs. Although not essential the same agent reduces the expression of mole-
[95], occludin contributes to TJs stabilization and cules associated with blood-tissue barrier break-
barrier function, and its depletion has been down, namely, fibronectin and collagen type IV
described in a mouse model of diabetes with a [110]. Fenofibric acid also dampens ER stress,
leaky outer BRB [89]. Interestingly, unlike clau- reactive oxygen species (ROS) generation, and
dins, occludin exhibits a dynamic behavior and apoptotic signals and increases insulin-like
contributes to the constant remodeling of TJs [96], growth factor I receptor (IGF-IR)-mediated sur-
which may allow regulation of these structures. vival signals in ARPE-19 cells under hyperglyce-
mia/hypoxia conditions. These works suggest
Numerous stress stimuli that have been linked that aside from reestablishing inner BRB integ-
to the pathogenesis of diabetic retinopathy and rity, fenofibrates may also contribute for the ame-
age-related macular degeneration like high glu- lioration of ME by modulating permeability at
cose concentrations [97, 98], oxidative stress the level of the RPE.
1 Pathophysiology of Macular Edema: Results from Basic Research 9

1.4.2.2 RPE Transcellular Pathway space [117]. Channels located at the apical mem-
The transcellular pathway, also present in endo- brane, namely, the Na+/K+ ATPase and the Na+/
thelial cells, involves the diverse set of transport K+-2Cl cotransporter, transport the said cations
systems described in Sect. 1.4.1.2. Below, we into the RPE cells, which in turn facilitate Cl-
elaborate on the most relevant aspects of this transport through the cell [118, 119]. Transport
pathway in RPE cells. of this anion from the apical to the basolateral
membrane induces fluid transport in the same
Facilitated Transport direction, contributing to the absorption of sub-
The GLUT family of proteins is responsible for retinal water into the choriocapillary system.
the transcellular facilitated transport of glucose Interestingly, a decrease in Na+/K+ ATPase activ-
in the RPE. Namely, GLUT1 is expressed in the ity in RPE cells of diabetic rabbits [120, 121] and
RPE [39, 111], and its expression in these cells in primary RPE cultures [122] has been reported,
does not vary in patients with diabetic retinopa- which may be one mechanism by which retinal/
thy (DR) [38] or in animal models of diabetes subretinal accumulation of fluid occurs.
[36]. Minor gene expression of GLUT3 and Rehak and colleagues also describe downreg-
GLUT5 has also been identified in the RPE [112], ulation of Kir4.1, an apical K+ channel involved
but further studies on these isoforms are lacking. in subretinal water transport, and of aquaporin-1
Aquaporins (AQ), another family of transport- and aquaporin-4 in RPE cells of an animal model
ers, function as constitutive water channels (also of central retinal vein occlusion [123]. Treatment
mentioned in Sect. 1.4.1.3). Several members of with triamcinolone reversed the downregulation
the AQ family are expressed in the RPE [113, of Kir4.1, indicating a possible additional mecha-
114] and contribute to the transepithelial trans- nism by which steroids decrease retinal edema in
port of water, thereby preventing subretinal accu- venous occlusions.
mulation of fluid [113, 115]. Experimental
conditions that mimic diseases or states associ- Transcytosis
ated with ME (such as diabetes, retinal vein There is evidence that apical-to-basal transport of
occlusion, high glucose concentrations, and macromolecules via vesicles occurs in RPE cells
VEGF supplementation) induce a complex regu- [104]. Interestingly, deletion of caveolin-1, an
lation of AQ that may be responsible for the accu- integral component of caveolae, disturbs the
mulation of sub- or intraretinal fluid [114, 116]. retina-RPE interface by impairing the fluid/ion
homeostasis in the subretinal space [124].
Active Transport Furthermore, transcytosis has been shown to con-
RPE cells have numerous, varied functions, many tribute to the trans-RPE transport of macrophages
of which depend on ion channels (e.g., transport and microglia in DR [125]. Therefore, disruption
and metabolism of vitamin A, regeneration of of this barrier mechanism may be an additional
PRs outer segments, phagocytosis, communica- factor contributing to the pathogenesis of ME.
tion with neighboring cells, etc.). However, in the
context of outer BRB function and ME, we will
only discuss the role of ion channels and pumps 1.5 Physical Mechanisms
in the context of transcellular transport and regu- Determining Fluid
lation of water in the subretinal space. Channel- Absorption/Retention
mediated ion transport plays a fundamental role in the Retina
in the movement of water across the RPE and,
hence, in preventing its subretinal accumulation. Aside from the BRB, which markedly limits the
K+ and Cl channels have an important role in passage of fluid and macromolecules to the retina
supporting the absorption of water in the retina- and subretinal space, physical forces and anatom-
to-choroid direction and do so by controlling ical features of this tissue determine that it
the concentration of these ions in the subretinal remains in a state of deturgescence in
10 A. Bastos-Carvalho and J. Ambati

physiological conditions. The physical forces 1.6 Concluding Remarks


that drive fluid into or out of the retina obey
Starlings law in general terms, i.e., the fluid Both inner and outer components of the BRB play
movement due to filtration across the wall of a a crucial role on the pathophysiology of macular
capillary is dependent on the balance between the edema. Many efforts have been done to understand
hydrostatic pressure gradient and the osmotic the mechanisms that regulate the permeability of
pressure gradient across the capillary [126]. this barrier, both in a healthy state and in disease
Therefore, the absorption or retention of fluid in conditions. Although currently used models have
the retina is, in part, a product of the balance thus far provided important data on ME patho-
between hydrostatic pressure in the capillaries physiology, none of these closely mimics ME as it
and retinal interstitium and osmotic pressure in happens in humans. Therefore, improved models
these two compartments. As a result, in physio- that better simulate human ME are warranted for a
logical conditions where the protein content of faster development of treatments for this patho-
the retinal extracellular space is very low, fluids logic state. Additionally, it is important to consider
will tend to be absorbed into the high osmotic that ME is a common end stage for many different
pressure compartment, the capillary lumen. ocular conditions, whose pathophysiology is very
However, when a breakdown of the BRB occurs distinct. Hence, differences in the mechanisms
and protein exudation occurs, this balance may governing ME in each disease may exist. Individual
be inverted, leading to fluid accumulation in the studies focusing on specific etiologies of ME may
retina. Aside from the four classic Starling fac- provide better insight into the molecular underpin-
tors and the low permeability of the BRB, tissue nings and possible treatment avenues for each of
compliance is also a limiting factor to fluid pas- condition.
sage into the interstitium. The retina has closely
interwoven cellular processes and minimal extra- Acknowledgments We thank Flvia Carvalho for design-
cellular space [7], which may influence fluid ing the illustrations and S. Lee Ware for the constructive
suggestions on the text.
flow. Indeed, flow conductivity of the retina is
low [127] and may, to some extent, decrease the
outflow of fluid from capillaries and also from
the vitreous into the retina. Indeed, the intraocu- References
lar pressure constantly pushes water from the vit-
reous into the retina and toward the choriocapillaris 1. Pascolini D, Mariotti SP. Global estimates of visual
[128], where the oncotic pressure is high and the impairment: 2010. Br J Ophthalmol. 2012;96:
EC fenestrations allow entry of fluid. Despite this 6148.
2. Trieschmann M, van Kuijk FJ, Alexander R, et al.
continuous force, which is not opposed by the
Macular pigment in the human retina: histological
vitreoretinal interface or the internal limiting evaluation of localization and distribution. Eye
membrane (as these do not constitute barriers to (Lond). 2008;22:1327.
the passage of fluid or small molecules [16]), the 3. Dowling JE, Boycott BB. Organization of the pri-
mate retina: electron microscopy. Proc R Soc Lond
retinal interstitium is dry in healthy condition.
B Biol Sci. 1966;166:80111.
This may be due to the retinas low fluid conduc- 4. Scholl S, Kirchhof J, Augustin AJ. Pathophysiology of
tivity that limits water entry from the vitreous. macular edema. Ophthalmol J Int Ophtalmol Int J
Fluid from the vitreous, as well as water pro- Ophthalmol Z Augenheilkd. 2010;224 Suppl 1:815.
5. Wurm A, Pannicke T, Iandiev I, et al. Purinergic sig-
duced by the retinas high metabolism, is directed
naling involved in Muller cell function in the mam-
toward the subretinal space and reaches the RPE, malian retina. Prog Retin Eye Res. 2011;30:32442.
where it can be absorbed [129]. 6. Aroca PR, Salvat M, Fernandez J, Mendez I. Risk
Finally, the retina is devoid of lymphatics, factors for diffuse and focal macular edema.
J Diabetes Complications. 2004;18:2115.
therefore lacking a common route of intersticial
7. Cunha-Vaz JG, Travassos A. Breakdown of the
fluid removal in organs outside the central ner- blood-retinal barriers and cystoid macular edema.
vous system. Surv Ophthalmol. 1984;28(Suppl):48592.
1 Pathophysiology of Macular Edema: Results from Basic Research 11

8. Burggraaff MC, Trieu J, de Vries-Knoppert WA, occludin in retinal endothelial cells. Penn State Retina
Balk L, Petzold A. The clinical spectrum of micro- Research Group. Diabetes. 1998;47:19539.
cystic macular edema. Invest Ophthalmol Vis Sci. 24. Barber AJ, Antonetti DA, Gardner TW. Altered
2014;55:95261. expression of retinal occludin and glial fibrillary
9. Coscas G, Cunha-Vaz J, Soubrane G. Macular acidic protein in experimental diabetes. The Penn
edema: definition and basic concepts. Dev Ophthalmol. State Retina Research Group. Invest Ophthalmol Vis
2010;47:19. Sci. 2000;41:35618.
10. Cunha-Vaz JG. The blood-ocular barriers: past, pres- 25. Saker S, Stewart EA, Browning AC, Allen CL,
ent, and future. Doc Ophthalmol Adv Ophthalmol. Amoaku WM. The effect of hyperglycaemia on per-
1997;93:14957. meability and the expression of junctional complex
11. Rizzolo LJ, Peng S, Luo Y, Xiao W. Integration of molecules in human retinal and choroidal endothe-
tight junctions and claudins with the barrier func- lial cells. Exp Eye Res. 2014;121:1617.
tions of the retinal pigment epithelium. Prog Retin 26. Antonetti DA, Barber AJ, Hollinger LA, Wolpert EB,
Eye Res. 2011;30:296323. Gardner TW. Vascular endothelial growth factor induces
12. Gariano RF, Iruela-Arispe ML, Hendrickson rapid phosphorylation of tight junction proteins occlu-
AE. Vascular development in primate retina: compari- din and zonula occluden 1. A potential mechanism for
son of laminar plexus formation in monkey and human. vascular permeability in diabetic retinopathy and
Invest Ophthalmol Vis Sci. 1994;35:344255. tumors. J Biol Chem. 1999;274:234637.
13. Kur J, Newman EA, Chan-Ling T. Cellular and 27. Antonetti DA, Wolpert EB, DeMaio L, Harhaj NS,
physiological mechanisms underlying blood flow Scaduto Jr RC. Hydrocortisone decreases retinal
regulation in the retina and choroid in health and dis- endothelial cell water and solute flux coincident with
ease. Prog Retin Eye Res. 2012;31:377406. increased content and decreased phosphorylation of
14. Davson H, Duke-Elder WS, Maurice DM, Ross EJ, occludin. J Neurochem. 2002;80:66777.
Woodin AM. The penetration of some electrolytes and 28. McAllister IL, Vijayasekaran S, Chen SD, Yu DY.
non-electrolytes into the aqueous humour and vitreous Effect of triamcinolone acetonide on vascular endo-
body of the cat. J Physiol. 1949;108:20317. thelial growth factor and occludin levels in branch
15. Toda R, Kawazu K, Oyabu M, Miyazaki T, Kiuchi Y. retinal vein occlusion. Am J Ophthalmol. 2009;147:
Comparison of drug permeabilities across the blood- 83846. 46.e12.
retinal barrier, blood-aqueous humor barrier, and 29. Taddei A, Giampietro C, Conti A, et al. Endothelial
blood-brain barrier. J Pharm Sci. 2011;100: adherens junctions control tight junctions by
390411. VE-cadherin-mediated upregulation of claudin-5.
16. Cunha-Vaz JG. The blood-retinal barriers. Doc Nat Cell Biol. 2008;10:92334.
Ophthalmol Adv Ophthalmol. 1976;41:287327. 30. Kerr NM, Johnson CS, de Souza CF, et al.
17. Klaassen I, Van Noorden CJ, Schlingemann RO. Immunolocalization of gap junction protein con-
Molecular basis of the inner blood-retinal barrier nexin43 (GJA1) in the human retina and optic nerve.
and its breakdown in diabetic macular edema and Invest Ophthalmol Vis Sci. 2010;51:402834.
other pathological conditions. Prog Retin Eye Res. 31. Nagasawa K, Chiba H, Fujita H, et al. Possible
2013;34:1948. involvement of gap junctions in the barrier function
18. Dejana E. Endothelial cell-cell junctions: happy of tight junctions of brain and lung endothelial cells.
together. Nat Rev Mol Cell Biol. 2004;5:26170. J Cell Physiol. 2006;208:12332.
19. Giannotta M, Trani M, Dejana E. VE-cadherin and 32. Keech AC, Mitchell P, Summanen PA, et al. Effect of
endothelial adherens junctions: active guardians of fenofibrate on the need for laser treatment for dia-
vascular integrity. Dev Cell. 2013;26:44154. betic retinopathy (FIELD study): a randomised con-
20. Russ PK, Davidson MK, Hoffman LH, Haselton FR. trolled trial. Lancet. 2007;370:168797.
Partial characterization of the human retinal endothe- 33. Chen Y, Hu Y, Lin M, et al. Therapeutic effects of
lial cell tight and adherens junction complexes. Invest PPARalpha agonists on diabetic retinopathy in type
Ophthalmol Vis Sci. 1998;39:247985. 1 diabetes models. Diabetes. 2013;62:26172.
21. Klaassen I, Hughes JM, Vogels IM, Schalkwijk CG, 34. Hosoya K, Tachikawa M. Inner blood-retinal barrier
Van Noorden CJ, Schlingemann RO. Altered expres- transporters: role of retinal drug delivery. Pharm
sion of genes related to blood-retina barrier disrup- Res. 2009;26:205565.
tion in streptozotocin-induced diabetes. Exp Eye 35. Kumagai AK. Glucose transport in brain and retina:
Res. 2009;89:415. implications in the management and complications of
22. Luo Y, Xiao W, Zhu X, et al. Differential expression diabetes. Diabetes Metab Res Rev. 1999;15:26173.
of claudins in retinas during normal development 36. Badr GA, Tang J, Ismail-Beigi F, Kern TS. Diabetes
and the angiogenesis of oxygen-induced retinopathy. downregulates GLUT1 expression in the retina and
Invest Ophthalmol Vis Sci. 2011;52:755664. its microvessels but not in the cerebral cortex or its
23. Antonetti DA, Barber AJ, Khin S, Lieth E, Tarbell JM, microvessels. Diabetes. 2000;49:101621.
Gardner TW. Vascular permeability in experimental dia- 37. Fernandes R, Carvalho AL, Kumagai A, et al.
betes is associated with reduced endothelial occludin Downregulation of retinal GLUT1 in diabetes by
content: vascular endothelial growth factor decreases ubiquitinylation. Mol Vis. 2004;10:61828.
12 A. Bastos-Carvalho and J. Ambati

38. Kumagai AK, Glasgow BJ, Pardridge WM. GLUT1 autoimmune uveoretinitis. Clin Exp Immunol. 1992;
glucose transporter expression in the diabetic and 88:4737.
nondiabetic human eye. Invest Ophthalmol Vis Sci. 52. Ben-Zvi A, Lacoste B, Kur E, et al. Mfsd2a is criti-
1994;35:288794. cal for the formation and function of the blood-brain
39. Takata K, Kasahara T, Kasahara M, Ezaki O, Hirano H. barrier. Nature. 2014;509:50711.
Ultracytochemical localization of the erythrocyte/ 53. Cunha-Vaz J. The bloodretinal barrier in retinal dis-
HepG2-type glucose transporter (GLUT1) in cells of ease. Eur Ophthalmic Rev. 2009;3:1058.
the blood-retinal barrier in the rat. Invest Ophthalmol 54. Hall CN, Reynell C, Gesslein B, et al. Capillary peri-
Vis Sci. 1992;33:37783. cytes regulate cerebral blood flow in health and dis-
40. Hosoya K, Kondo T, Tomi M, Takanaga H, Ohtsuki ease. Nature. 2014;508:5560.
S, Terasaki T. MCT1-mediated transport of L-lactic 55. Frank RN, Turczyn TJ, Das A. Pericyte coverage of
acid at the inner blood-retinal barrier: a possible retinal and cerebral capillaries. Invest Ophthalmol
route for delivery of monocarboxylic acid drugs to Vis Sci. 1990;31:9991007.
the retina. Pharm Res. 2001;18:166976. 56. Chan-Ling T, Koina ME, McColm JR, et al. Role of
41. Nakashima T, Tomi M, Katayama K, et al. Blood-to- CD44+ stem cells in mural cell formation in the
retina transport of creatine via creatine transporter human choroid: evidence of vascular instability due
(CRT) at the rat inner blood-retinal barrier. J Neurochem. to limited pericyte ensheathment. Invest Ophthalmol
2004;89:145461. Vis Sci. 2011;52:399410.
42. de Souza CF, Kalloniatis M, Christie DL, 57. Shepro D, Morel NM. Pericyte physiology. FASEB J
Polkinghorne PJ, McGhee CN, Acosta ML. Creatine Off Publ Fed Am Soc Exp Biol. 1993;7:10318.
transporter immunolocalization in aged human and 58. Wisniewska-Kruk J, Hoeben KA, Vogels IM, et al. A
detached retinas. Invest Ophthalmol Vis Sci. 2012; novel co-culture model of the blood-retinal barrier
53:193645. based on primary retinal endothelial cells, pericytes
43. Tomi M, Mori M, Tachikawa M, Katayama K, Terasaki and astrocytes. Exp Eye Res. 2012;96:18190.
T, Hosoya K. L-type amino acid transporter 1-mediated 59. Kim JH, Kim JH, Yu YS, Kim DH, Kim KW.
L-leucine transport at the inner blood-retinal barrier. Recruitment of pericytes and astrocytes is closely
Invest Ophthalmol Vis Sci. 2005;46:252230. related to the formation of tight junction in developing
44. Hosoya K, Saeki S, Terasaki T. Activation of carrier- retinal vessels. J Neurosci Res. 2009;87:6539.
mediated transport of L-cystine at the blood-brain 60. Mizutani M, Kern TS, Lorenzi M. Accelerated death
and blood-retinal barriers in vivo. Microvasc Res. of retinal microvascular cells in human and experi-
2001;62:13642. mental diabetic retinopathy. J Clin Invest. 1996;
45. Tomi M, Kitade N, Hirose S, et al. Cationic amino 97:288390.
acid transporter 1-mediated L-arginine transport at 61. Daneman R, Zhou L, Kebede AA, Barres BA. Pericytes
the inner blood-retinal barrier. J Neurochem. 2009; are required for blood-brain barrier integrity during
111:71625. embryogenesis. Nature. 2010;468:5626.
46. Kubo Y. Inner blood-retinal barrier transporters: rel- 62. Enge M, Bjarnegard M, Gerhardt H, et al.
evance to diabetic retinopathy. In: Ola MS, editor. Endothelium-specific platelet-derived growth factor-
Diabetic retinopathy. Intech; 2012. p. 91108 http:// B ablation mimics diabetic retinopathy. EMBO J.
www.intechopen.com/books/diabetic-retinopathy 2002;21:430716.
47. Frank PG, Woodman SE, Park DS, Lisanti MP. 63. Davis S, Aldrich TH, Jones PF, et al. Isolation of angio-
Caveolin, caveolae, and endothelial cell function. poietin-1, a ligand for the TIE2 receptor, by secretion-
Arterioscler Thromb Vasc Biol. 2003;23:11618. trap expression cloning. Cell. 1996;87:11619.
48. Feng Y, Venema VJ, Venema RC, Tsai N, Caldwell 64. Jones N, Iljin K, Dumont DJ, Alitalo K. Tie recep-
RB. VEGF induces nuclear translocation of Flk-1/ tors: new modulators of angiogenic and lymphangi-
KDR, endothelial nitric oxide synthase, and caveo- ogenic responses. Nat Rev Mol Cell Biol. 2001;2:
lin-1 in vascular endothelial cells. Biochem Biophys 25767.
Res Commun. 1999;256:1927. 65. Hori S, Ohtsuki S, Hosoya K, Nakashima E, Terasaki
49. Hofman P, Blaauwgeers HG, Tolentino MJ, et al. T. A pericyte-derived angiopoietin-1 multimeric
VEGF-A induced hyperpermeability of blood-retinal complex induces occludin gene expression in brain
barrier endothelium in vivo is predominantly associ- capillary endothelial cells through Tie-2 activation
ated with pinocytotic vesicular transport and not with in vitro. J Neurochem. 2004;89:50313.
formation of fenestrations. Vascular endothelial 66. Zhang ZG, Zhang L, Croll SD, Chopp M.
growth factor-A. Curr Eye Res. 2000;21:63745. Angiopoietin-1 reduces cerebral blood vessel leak-
50. Hofman P, Blaauwgeers HG, Vrensen GF, age and ischemic lesion volume after focal cerebral
Schlingemann RO. Role of VEGF-A in endothelial embolic ischemia in mice. Neuroscience. 2002;
phenotypic shift in human diabetic retinopathy and 113:6837.
VEGF-A-induced retinopathy in monkeys. Ophthalmic 67. Gavard J, Patel V, Gutkind JS. Angiopoietin-1 pre-
Res. 2001;33:15662. vents VEGF-induced endothelial permeability by
51. Lightman S, Greenwood J. Effect of lymphocytic sequestering Src through mDia. Dev Cell. 2008;14:
infiltration on the blood-retinal barrier in experimental 2536.
1 Pathophysiology of Macular Edema: Results from Basic Research 13

68. Suri C, Jones PF, Patan S, et al. Requisite role of 82. Rehak M, Hollborn M, Iandiev I, et al. Retinal gene
angiopoietin-1, a ligand for the TIE2 receptor, expression and Muller cell responses after branch
during embryonic angiogenesis. Cell. 1996;87: retinal vein occlusion in the rat. Invest Ophthalmol
117180. Vis Sci. 2009;50:235967.
69. Patel JI, Hykin PG, Gregor ZJ, Boulton M, Cree 83. Pannicke T, Wurm A, Iandiev I, et al. Deletion of
IA. Angiopoietin concentrations in diabetic retinop- aquaporin-4 renders retinal glial cells more suscep-
athy. Br J Ophthalmol. 2005;89:4803. tible to osmotic stress. J Neurosci Res. 2010;88:
70. Park SW, Yun JH, Kim JH, Kim KW, Cho CH, Kim 287788.
JH. Angiopoietin 2 induces pericyte apoptosis via 84. Xin X, Rodrigues M, Umapathi M, et al. Hypoxic
alpha3beta1 integrin signaling in diabetic retinopa- retinal Muller cells promote vascular permeability
thy. Diabetes. 2014;63:305768. by HIF-1-dependent up-regulation of angiopoietin-
71. Rangasamy S, Srinivasan R, Maestas J, McGuire like 4. Proc Natl Acad Sci U S A. 2013;110:
PG, Das A. A potential role for angiopoietin 2 in the E342534.
regulation of the blood-retinal barrier in diabetic 85. Rajasekaran SA, Hu J, Gopal J, et al. Na,K-ATPase
retinopathy. Invest Ophthalmol Vis Sci. 2011;52: inhibition alters tight junction structure and permea-
378491. bility in human retinal pigment epithelial cells. Am J
72. Tout S, Chan-Ling T, Hollander H, Stone J. The role Physiol Cell Physiol. 2003;284:C1497507.
of Muller cells in the formation of the blood-retinal 86. Weinberger D, Fink-Cohen S, Gaton DD, Priel E,
barrier. Neuroscience. 1993;55:291301. Yassur Y. Non-retinovascular leakage in diabetic
73. Tretiach M, Madigan MC, Wen L, Gillies MC. Effect maculopathy. Br J Ophthalmol. 1995;79:72831.
of Muller cell co-culture on in vitro permeability of 87. Vinores SA, Gadegbeku C, Campochiaro PA, Green
bovine retinal vascular endothelium in normoxic and WR. Immunohistochemical localization of blood-
hypoxic conditions. Neurosci Lett. 2005;378:1605. retinal barrier breakdown in human diabetics. Am J
74. Nishikiori N, Osanai M, Chiba H, et al. Glial cell- Pathol. 1989;134:2315.
derived cytokines attenuate the breakdown of vascu- 88. Vinores SA, Derevjanik NL, Ozaki H, Okamoto N,
lar integrity in diabetic retinopathy. Diabetes. 2007; Campochiaro PA. Cellular mechanisms of blood-
56:133340. retinal barrier dysfunction in macular edema. Doc
75. Eichler W, Yafai Y, Keller T, Wiedemann P, Ophthalmol Adv Ophthalmol. 1999;97:21728.
Reichenbach A. PEDF derived from glial Muller 89. Xu HZ, Le YZ. Significance of outer blood-retina
cells: a possible regulator of retinal angiogenesis. barrier breakdown in diabetes and ischemia. Invest
Exp Cell Res. 2004;299:6878. Ophthalmol Vis Sci. 2011;52:21604.
76. Yafai Y, Lange J, Wiedemann P, Reichenbach A, 90. Sun R, Peng S, Chen X, Zhang H, Rizzolo LJ.
Eichler W. Pigment epithelium-derived factor acts as an Diffusible retinal secretions regulate the expression of
opponent of growth-stimulatory factors in retinal glial- tight junctions and other diverse functions of the reti-
endothelial cell interactions. Glia. 2007;55:64251. nal pigment epithelium. Mol Vis. 2008;14:223762.
77. Nagelhus EA, Horio Y, Inanobe A, et al. Immunogold 91. Tsukita S, Furuse M, Itoh M. Multifunctional strands
evidence suggests that coupling of K+ siphoning and in tight junctions. Nat Rev Mol Cell Biol. 2001;2:
water transport in rat retinal Muller cells is mediated 28593.
by a coenrichment of Kir4.1 and AQP4 in specific 92. Angelow S, Yu AS. Claudins and paracellular trans-
membrane domains. Glia. 1999;26:4754. port: an update. Curr Opin Nephrol Hypertens.
78. Reichenbach A, Wurm A, Pannicke T, Iandiev I, 2007;16:45964.
Wiedemann P, Bringmann A. Muller cells as players 93. Wang FE, Zhang C, Maminishkis A, et al.
in retinal degeneration and edema. Graefes archive MicroRNA-204/211 alters epithelial physiology.
for clinical and experimental ophthalmology. FASEB J Off Publ Fed Am Soc Exp Biol. 2010;
Albrecht Von Graefes Arch Klin Exp Ophthalmol. 24:155271.
2007;245:62736. 94. Peng S, Rao VS, Adelman RA, Rizzolo LJ.
79. Pannicke T, Iandiev I, Wurm A, et al. Diabetes alters Claudin-19 and the barrier properties of the human
osmotic swelling characteristics and membrane con- retinal pigment epithelium. Invest Ophthalmol Vis
ductance of glial cells in rat retina. Diabetes. 2006; Sci. 2011;52:1392403.
55:6339. 95. Saitou M, Fujimoto K, Doi Y, et al. Occludin-deficient
80. Pannicke T, Uckermann O, Iandiev I, Wiedemann P, embryonic stem cells can differentiate into polarized
Reichenbach A, Bringmann A. Ocular inflammation epithelial cells bearing tight junctions. J Cell Biol.
alters swelling and membrane characteristics of rat 1998;141:397408.
Muller glial cells. J Neuroimmunol. 2005;161: 96. Shen L, Weber CR, Turner JR. The tight junction
14554. protein complex undergoes rapid and continuous
81. Zhao M, Bousquet E, Valamanesh F, et al. molecular remodeling at steady state. J Cell Biol.
Differential regulations of AQP4 and Kir4.1 by tri- 2008;181:68395.
amcinolone acetonide and dexamethasone in the 97. Villarroel M, Garcia-Ramirez M, Corraliza L,
healthy and inflamed retina. Invest Ophthalmol Vis Hernandez C, Simo R. High glucose concentration
Sci. 2011;52:63407. leads to differential expression of tight junction pro-
14 A. Bastos-Carvalho and J. Ambati

teins in human retinal pigment epithelial cells. functional implications in retinal permeability.
Endocrinol Nutr Organo Soc Esp Endocrinol Nutr. Invest Ophthalmol Vis Sci. 2011;52:634854.
2009;56:538. 111. West KA, Yan L, Shadrach K, et al. Protein database,
98. Villarroel M, Garcia-Ramirez M, Corraliza L, human retinal pigment epithelium. Mol Cell Proteome
Hernandez C, Simo R. Effects of high glucose con- MCP. 2003;2:3749.
centration on the barrier function and the expression 112. Takagi H, Tanihara H, Seino Y, Yoshimura
of tight junction proteins in human retinal pigment N. Characterization of glucose transporter in cul-
epithelial cells. Exp Eye Res. 2009;89:91320. tured human retinal pigment epithelial cells: gene
99. Bailey TA, Kanuga N, Romero IA, Greenwood J, expression and effect of growth factors. Invest
Luthert PJ, Cheetham ME. Oxidative stress affects Ophthalmol Vis Sci. 1994;35:1707.
the junctional integrity of retinal pigment epithelial 113. Stamer WD, Bok D, Hu J, Jaffe GJ, McKay
cells. Invest Ophthalmol Vis Sci. 2004;45:67584. BS. Aquaporin-1 channels in human retinal pigment
100. Yoshikawa T, Ogata N, Izuta H, Shimazawa M, Hara epithelium: role in transepithelial water movement.
H, Takahashi K. Increased expression of tight junc- Invest Ophthalmol Vis Sci. 2003;44:28038.
tions in ARPE-19 cells under endoplasmic reticulum 114. Hollborn M, Dukic-Stefanovic S, Pannicke T, et al.
stress. Curr Eye Res. 2011;36:115363. Expression of aquaporins in the retina of diabetic
101. Abe T, Sugano E, Saigo Y, Tamai M. Interleukin- rats. Curr Eye Res. 2011;36:8506.
1beta and barrier function of retinal pigment epithe- 115. Baetz NW, Stamer WD, Yool AJ. Stimulation of
lial cells (ARPE-19): aberrant expression of aquaporin-mediated fluid transport by cyclic GMP
junctional complex molecules. Invest Ophthalmol in human retinal pigment epithelium in vitro. Invest
Vis Sci. 2003;44:4097104. Ophthalmol Vis Sci. 2012;53:212732.
102. Peng S, Gan G, Rao VS, Adelman RA, Rizzolo LJ. 116. Hollborn M, Rehak M, Iandiev I, et al. Transcriptional
Effects of proinflammatory cytokines on the claudin-19 regulation of aquaporins in the ischemic rat retina:
rich tight junctions of human retinal pigment epithelium. upregulation of aquaporin-9. Curr Eye Res. 2012;
Invest Ophthalmol Vis Sci. 2012;53:501628. 37:52431.
103. Jin M, Barron E, He S, Ryan SJ, Hinton DR. 117. Strauss O. The retinal pigment epithelium in visual
Regulation of RPE intercellular junction integrity function. Physiol Rev. 2005;85:84581.
and function by hepatocyte growth factor. Invest 118. Miller SS, Edelman JL. Active ion transport path-
Ophthalmol Vis Sci. 2002;43:278290. ways in the bovine retinal pigment epithelium.
104. Peng S, Adelman RA, Rizzolo LJ. Minimal effects J Physiol. 1990;424:283300.
of VEGF and anti-VEGF drugs on the permeability 119. DiMattio J, Degnan KJ, Zadunaisky JA. A model for
or selectivity of RPE tight junctions. Invest transepithelial ion transport across the isolated reti-
Ophthalmol Vis Sci. 2010;51:321625. nal pigment epithelium of the frog. Exp Eye Res.
105. Ghassemifar R, Lai CM, Rakoczy PE. VEGF differ- 1983;37:40920.
entially regulates transcription and translation of 120. MacGregor LC, Matschinsky FM. Altered retinal
ZO-1alpha+ and ZO-1alpha- and mediates trans- metabolism in diabetes. II. Measurement of sodium-
epithelial resistance in cultured endothelial and epi- potassium ATPase and total sodium and potassium
thelial cells. Cell Tissue Res. 2006;323:11725. in individual retinal layers. J Biol Chem. 1986;261:
106. Ablonczy Z, Crosson CE. VEGF modulation of reti- 40528.
nal pigment epithelium resistance. Exp Eye Res. 121. MacGregor LC, Matschinsky FM. Experimental dia-
2007;85:76271. betes mellitus impairs the function of the retinal pig-
107. Hartnett ME, Lappas A, Darland D, McColm JR, mented epithelium. Metab Clin Exp. 1986;35:2834.
Lovejoy S, DAmore PA. Retinal pigment epithelium 122. Crider JY, Yorio T, Sharif NA, Griffin BW. The
and endothelial cell interaction causes retinal pigment effects of elevated glucose on Na+/K(+)-ATPase of
epithelial barrier dysfunction via a soluble VEGF- cultured bovine retinal pigment epithelial cells mea-
dependent mechanism. Exp Eye Res. 2003;77:5939. sured by a new nonradioactive rubidium uptake
108. Miyamoto N, de Kozak Y, Jeanny JC, et al. Placental assay. J Ocul Pharmacol Ther Off J Assoc Ocul
growth factor-1 and epithelial haemato-retinal barrier Pharmacol Ther. 1997;13:33752.
breakdown: potential implication in the pathogenesis of 123. Rehak M, Drechsler F, Koferl P, et al. Effects of
diabetic retinopathy. Diabetologia. 2007;50:46170. intravitreal triamcinolone acetonide on retinal gene
109. Villarroel M, Garcia-Ramirez M, Corraliza L, expression in a rat model of central retinal vein
Hernandez C, Simo R. Fenofibric acid prevents reti- occlusion. Graefes archive for clinical and experi-
nal pigment epithelium disruption induced by mental ophthalmology. Albrecht Von Graefes Arch
interleukin-1beta by suppressing AMP-activated Klin Exp Ophthalmol. 2011;249:117583.
protein kinase (AMPK) activation. Diabetologia. 124. Li X, McClellan ME, Tanito M, et al. Loss of caveo-
2011;54:154353. lin-1 impairs retinal function due to disturbance of
110. Trudeau K, Roy S, Guo W, et al. Fenofibric acid subretinal microenvironment. J Biol Chem. 2012;
reduces fibronectin and collagen type IV overexpres- 287:1642434.
sion in human retinal pigment epithelial cells grown 125. Omri S, Behar-Cohen F, de Kozak Y, et al. Microglia/
in conditions mimicking the diabetic milieu: macrophages migrate through retinal epithelium
1 Pathophysiology of Macular Edema: Results from Basic Research 15

barrier by a transcellular route in diabetic retinopa- 128. Moseley H, Foulds WS, Allan D, Kyle PM.
thy: role of PKCzeta in the Goto Kakizaki rat model. Routes of clearance of radioactive water from the
Am J Pathol. 2011;179:94253. rabbit vitreous. Br J Ophthalmol. 1984;68:
126. Starling EH. On the absorption of fluids from the con- 14551.
nective tissue spaces. J Physiol. 1896;19:31226. 129. Marmor MF. Mechanisms of fluid accumulation in
127. Fatt I, Shantinath K. Flow conductivity of retina and its retinal edema. Doc Ophthalmol Adv Ophthalmol.
role in retinal adhesion. Exp Eye Res. 1971;12:21826. 1999;97:23949.
Pathophysiology of Macular
Edema in Diabetes, Retinal Vein 2
Occlusion, and Uveitis: A Disease-
Related Approach

Edoardo Midena and Silvia Bini

Contents
2.1 Introduction
2.1 Introduction 17
2.2 Macular Edema in Diabetes 18 Diabetic retinopathy (DR), retinal vascular dis-
2.2.1 Diabetes and Diabetic Retinopathy 18 orders (central and branch retinal vein occlu-
2.2.2 Mechanisms in DR Leading to DME 18
2.2.3 Morphological Findings in DR and DME 19 sions), and uveitis are the three major causes
2.2.4 Mediators in DME 20 of permanent visual acuity loss secondary to
2.3 Macular Edema in Retinal Vein
macular edema in the working population.
Occlusion 21 Macular edema (ME) is a common complica-
2.3.1 Definition of RVO 21 tion shared by these conditions leading to vision
2.3.2 Pathogenesis in RVO 21 loss. Nowadays, several treatment options are
2.4 Macular Edema in Uveitis 22 available for ME secondary to these clinical
2.4.1 Inflammatory Markers 22 situations. Notwithstanding ME represents a
2.4.2 Treatment Options 22 big therapeutic challenge for ophthalmologists,
References 23 due to its unpredictable therapeutic results. The
pathophysiological mechanisms leading to ME
are still poorly understood due to the complex
and multifactorial origin (Fig. 2.1).
Recent studies aim to clarify the mecha-
nisms and the molecules mainly involved in
the pathogenesis of fluid accumulation in the
outer and inner retina. Many cytokines, chemo-
kines, growth factors, and other molecules have
been investigated in human vitreous and aque-
ous humor samples. The correct understand-
E. Midena, MD, PhD (*) S. Bini, MD ing of pathways and interactions among these
Department of Ophthalmology, University of Padua, molecules in the pathogenesis of ME may lead
Via Giustiniani, n. 2, Padova 35128, Italy
e-mail: EDOARDO.MIDENA@UNIPD.IT; to the possibility of a more tailored therapeutic
silviabini85@hotmail.it approach.

A.J. Augustin (ed.), Intravitreal Steroids, 17


DOI 10.1007/978-3-319-14487-0_2, Springer International Publishing Switzerland 2015
18 E. Midena and S. Bini

Fig. 2.1 Tiny hyperreflective spots are evident both in diabetic retinopathy and macular edema. Hyperreflective
inner and outer retina (subsequently counted between the spots are considered as aggregates of activated microglial
two vertical red lines) in an eye with nonproliferative cells (for more details, please see reference number [1])

2.2 Macular Edema in Diabetes progression of DR [4, 5]. Aiello et al. recently
demonstrated that early effects of metabolic con-
2.2.1 Diabetes and Diabetic trol continue to accrue over time because of the
Retinopathy presence of metabolic memory. For this rea-
son, it is important to reach the best glycemic
DR is a common complication of both type 1 control in diabetic patients as early as possible
and type 2 diabetes mellitus (DM). It is a chronic after the development or occurrence of diabetes
progressive, potentially sight-threatening dis- mellitus [6].
order affecting the whole retinal components
associated with prolonged hyperglycemia and
other concomitant risk factors. The International 2.2.2 Mechanisms in DR Leading
Diabetes Federation (IDF) in 2006 published to DME
data showing that diabetes was affecting 246
million people worldwide. Nowadays, IDF For decades DR has been mainly considered
estimates 382 million people worldwide liv- simply a microvascular disorder, caused by
ing with diabetes. Recent data predict that by endothelial cell damage, pericyte loss, and sec-
2030, the diabetic population will raise to 552 ondary breakdown of the inner blood-retinal bar-
million [2]. Diabetic macular edema (DME) rier (BRB), leading to DME formation. Other
may occur at any stage of DR and represents factors such as hypoxia, altered blood flow, reti-
the leading cause of legal blindness among dia- nal ischemia, and inflammation are also associ-
betics. Approximately 14 % of all diabetics are ated with the progression of DR and DME [7, 8].
affected by DME [3]. Systemic risk factors pro- However, an increasing body of evidence sug-
moting the development of DME include arterial gests that neurodegeneration and retinal glial
hypertension, hyperlipidemia, and poor blood cell activation occur even before the earliest
glucose control. The control of these risk factors clinical manifestation of diabetic retinal micro-
decreases the development of ME and lowers the vasculopathy [9].
2 Pathophysiology of Macular Edema in Diabetes, Retinal Vein Occlusion, and Uveitis 19

Fig. 2.2 Hyperreflective spots (white brilliant dots) in macular edema due to radiation maculopathy. This disorder is
now recognized as being mainly inflammatory in nature

2.2.3 Morphological Findings the outer retina. Thus, the HRS may represent
in DR and DME a sign of microglial activation [1] (see Fig. 2.1).
This hypothesis has been recently confirmed
Morphological examination of DR and DME in macular edema secondary to eye irradiation
classically includes biomicroscopy and fundus (Midena et al. IOVS, 2014, ARVO e-abs# 5952),
photography, optical coherence tomography a clinical entity considered mainly inflammatory
(OCT), and fluorescein angiography. In particu- in nature (Fig. 2.2). Murakami et al. recently
lar, spectral domain OCT (SD-OCT) is largely reviewed SD-OCT aspects and measurement of
contributing to the understanding of DR patho- the retinal thickness in DME [11]. These authors
physiology. In fact it allows to analyze indi- highlighted that central retinal thickness is mod-
vidual retinal layer changes secondary to DM, estly correlated with visual impairment, whereas
both for thickness and for reflectivity aspects. it dramatically changes (reduces) after treat-
Recently, Vujosevic et al. confirmed the impor- ments compared to visual acuity. This observa-
tance of studying retinal layers by means of tion suggests that central retinal thickness may
SD-OCT not only in nonproliferative DR but be important in determining final visual acuity,
also in the preclinical stages. These changes but other probably more relevant parameters
are visible mainly in the inner retina, with the contribute to visual deterioration (i.e., external
massive involvement of Mller cells [10]. These limiting membrane integrity, which represents a
data have been recently reconfirmed and asso- sign of Mller cells morpho-functional integ-
ciated with Mller cell biomarkers (Midena rity). Murakami et al. also reported the presence
et al. IOVS, 2014, ARVO e-abs # 4429). The of intraretinal hyperreflective spots (or foci) in
same authors recently demonstrated the pres- the outer retinal layers, sometimes associated
ence of intraretinal hyperreflective spots (HRS) with serous retinal detachments. The location of
in diabetic patients with and without detectable these spots once again suggests that the patho-
retinopathy [1]. The HRS are initially present genesis of retinal fluid accumulation may be also
in the same retinal location (inner retina) where investigated at different levels of the retina, even
microglia is usually resident. Moreover, with at the interface between outer retina and retinal
the progression of retinopathy, HRS migrate to pigment epithelium [11].
20 E. Midena and S. Bini

2.2.4 Mediators in DME 2.2.4.2 The Inammatory Cascade


The other relevant mediator of DME formation
Another factor to be considered as a key factor in is the inflammatory cascade. This may be easily
the onset and progression of DR is the oxidative hypothesized considering its well-known role in
stress caused by DM. In fact, oxidative stress is vasopermeability and also looking at the efficacy
responsible for the increased generation of reac- of intravitreal steroid treatment [16, 17]. The
tive oxygen species and the decreased activity of inflammatory cascade includes a vast number of
antioxidant enzymes [12]. Many authors have cytokines and chemokines, which secondarily
contributed to the understanding of the metabolic also activate VEGF. The full and detailed inter-
pathways that lead to progression of DR and actions are still poorly understood. The media-
DME, by means of aqueous humor and vitreous tors showing the most relevant correlation with
analysis. This kind of approach is extremely rel- the presence of DME and the highest accordance
evant to clarify any molecule involved in the among the different studies seem to be interleu-
pathogenesis of DME and may suggest a targeted kin 6 (IL-6), IL-8, and monocyte chemoattractant
therapy. protein-1 (MCP-1) [1821]. IL-6 is a cytokine,
acting widely throughout the inflammatory cas-
2.2.4.1 Vascular Endothelial Growth cade, and is known to induce acute-phase reac-
Factor (VEGF) tions and increase vascular permeability. Higher
Aiello et al. demonstrated that the vascular endo- levels of IL-6 have been demonstrated in aqueous
thelial growth factor (VEGF) is increased in humor of diabetic patients compared to nondia-
human ocular fluids and plays a crucial role in betics [18]. A more recent study also demon-
ischemic retinal diseases, such as DR and retinal strated a correlation between elevated aqueous
vein occlusions [13]. The VEGF family includes IL-6 level and severity of DME, measured by
different isoforms: VEGF-A has been shown to increased thickness at OCT [22]. MCP-1 is a
be upregulated in hypoxic tissues. In DR, the loss chemotactic chemokine that induces monocyte
of retinal capillaries is believed to lead to hypoxia, and macrophage infiltration into tissue. The
which is the main stimulus for increased retinal recruitment of monocyte and macrophage to ves-
expression of VEGF-A, mediated through sel walls has been shown to promote vascular
hypoxia-inducible factors. VEGF-A has an permeability, increasing DME. Sohn et al. dem-
angiogenic role that is responsible for the pro- onstrated that aqueous MCP-1 levels were signif-
gression of DR to the proliferative stage. Apart icantly elevated in patients with DME compared
from its angiogenic role, VEGF-A increases vas- to controls and elevated MCP-1 was significantly
cular permeability [14]. Thus, the role of reduced following treatment with intravitreal
VEGF-A may be central to DME pathogenesis. steroid, associated also to a reduction of central
Moreover, several studies have demonstrated the macular thickness at OCT [20]. IL-8 is a pro-
efficacy of anti-VEGF treatment of DME, thus inflammatory and angiogenic cytokine produced
supporting the role of VEGF. However, more by endothelial and glial cells in ischemic retina
recently, Funk et al. demonstrated that while anti- [23]. Aqueous levels of IL-8 have been shown to
VEGF treatment decreases aqueous VEGF to increase with progression of DR [22] and are also
sub-physiologic levels, this effect does not induce significantly elevated in ocular fluids of diabetic
a complete resolution of DME. This study, con- patients with DME [15]. Furthermore, Sohn et al.
firmed by many others, also demonstrated the observed that IL-8 levels were unaffected follow-
important correlation between high intraocular ing intravitreal anti-VEGF or steroid treatment
cytokine levels and the presence of DME. These in patients with DME [20]. Noticeably, IL-8 has
data suggest that VEGF alone cannot be the been found positively correlated with severity
unique driving factor for the development of of DME but not ME secondary to branch retinal
DME [15]. vein occlusion [19]. If these data are confirmed,
2 Pathophysiology of Macular Edema in Diabetes, Retinal Vein Occlusion, and Uveitis 21

it may suggest that IL-8 has a specific role in lular and inflammatory reactions, leading to vas-
the genesis of diabetic macular edema and may cular dysfunction. Vascular dysfunction is
represent a new specific target for therapy [24]. responsible for the breakdown of the BRB. As in
Different studies point out that there are several DME, in RVO the release of angiogenic and
mechanisms and complex pathways leading to inflammatory mediators has a crucial role in the
the breakdown of the BRB and the subsequent alteration of BRB.
development of DME. As suggested by Owen and
Hartnett, we can talk about a pathway overlap 2.3.2.1 Inhibition of VEGF
and crosstalk, and the final common pathway As previously reported, Aiello et al. demon-
of DME still needs to be clarified [24]. More strated pathologically high levels of VEGF in
studies are required for a better understanding of intraocular fluid of patients with retinal neovas-
all molecular events. Understanding the upstream cularization secondary not only to DR but also to
factors in the pathogenesis of DME will lead to RVO. In fact, hypoxia causes increased expres-
a more targeted therapy, conceived not only as sion of VEGF, leading to increase in vascular
a unique therapy, but also as a combination of permeability. Moreover several studies have
treatments, with the goal of providing the most demonstrated the efficacy of intravitreal injec-
effective and safest way to treat DME. tions of anti-VEGF agents, thus confirming the
pathogenetic role of VEGF. Clinical trials like the
BRAVO and the CRUISE study demonstrated
2.3 Macular Edema in Retinal that in patients with RVO, the blockage of VEGF
Vein Occlusion with ranibizumab reduces macular edema and
improves vision [28, 29]. The role of inflamma-
2.3.1 Denition of RVO tion in RVO is also considered. Some reports
showed that intraocular and periocular steroid
Retinal vein occlusion (RVO) is the second most administration reduces ME in patients with RVO
common retinal vascular disorder after diabetic [3032]. Moreover, the Geneva Study Group
retinopathy [25, 26]. It is caused by vascular demonstrated the efficacy and safety of dexa-
obstruction leading to the formation of intrareti- methasone intravitreal implant in reducing the
nal hemorrhages, fluid exudation, and develop- risk of vision loss due to ME in BRVO and
ment of a variable degree of retinal ischemia. CRVO, compared to sham treatment [33].
RVO is classified as central RVO (CRVO),
hemiretinal vein occlusions, and branch RVO 2.3.2.2 Inhibition of Other
(BRVO) [27]. Inammatory Molecules
ME often occurs in the course of these vascu- Many studies have evaluated the profiles of vari-
lar disorders and is the main cause of visual ous cytokines in intraocular fluid and the relation-
impairment. Although ME is the main reason for ship between their levels and ME in RVO.
visual impairment, retinal ischemia may lead to A recent work by Sohn et al. analyzed several
neovascularization as a late complication and cytokine levels and other factors like VEGF in
may result in vitreous hemorrhage and neovascu- aqueous humor of patients with BRVO who under-
lar glaucoma. went two different intravitreal treatments: steroid
and anti-VEGF. They found aqueous levels of
IL-6, IL-8, IL-17, and VEGF significantly higher
2.3.2 Pathogenesis in RVO in the BRVO group than in the control group
before the administration of any treatment. These
The pathogenesis of ME in RVO is complex, as data agree with previous results [3437]. In the
reported for DME. When venous occlusion steroid treatment group, the levels of inflammatory
occurs, vascular damage is accompanied by cel- molecules and VEGF were significantly reduced.
22 E. Midena and S. Bini

In the anti-VEGF treatment group, only VEGF infectious and noninfectious uveitis and demon-
was significantly reduced (the reduction was more strated an increased level of IL-6, IL-8, soluble
relevant than in steroid-treated eyes). These results intercellular adhesion molecule, soluble vascular
clarify that intravitreal steroid treatment is able to cell adhesion molecule, and interferon-inducible
reduce plural inflammatory cytokines and VEGF protein-10 compared to non-uveitic patients.
levels in aqueous humor in BRVO patients, while Higher levels of these and other factors were
anti-VEGF seems to have a selective influence on found in patients with active uveitis compared to
VEGF levels but not on other cytokines [16]. quiescent uveitis. No differences in the inflam-
These data once again suggest that the inflam- matory molecule levels were found between uve-
matory cascade may have a crucial role in ME itic patients with and without CME [40]. More
pathogenesis, also in RVO. Moreover, patients recent studies have confirmed the increased lev-
developing venous occlusion have atheroscle- els of these molecules during inflammatory uve-
rotic lesions associated with chronic inflamma- itic processes, but the correct interactions and the
tion, preceding the acute event of vascular entire inflammatory cascade are not yet com-
occlusion. Thus, we may speculate that the pletely understood.
inflammatory cascade and the consequent cyto-
kine elevation in ocular fluids might precede or
be independent from VEGF expression observed 2.4.2 Treatment Options
after ischemia. However, more studies are needed
to clarify the correct interactions among all the A few studies have compared the efficacy of ste-
involved mediators. roid intravitreal treatment and anti-VEGF treat-
ment in controlling CME in uveitis. The results
show that anti-VEGF may be a supplementary
2.4 Macular Edema in Uveitis therapy for persistent CME, but steroid therapy
remains the best treatment option in reducing
Uveitis recognizes infectious and noninfectious CME at OCT and in improving visual acuity. In
causes. As in DR and RVO, cystoid macular particular VEGF has been demonstrated to play
edema (CME) is a major cause of reduced visual an important role in CME genesis in Behet dis-
acuity in patients affected by uveitis. The patho- ease, more than in any other uveitis entity [41,
genesis is strictly connected with the prolonged 42]. This may be explained because Behet
ocular inflammation and the consequent disrup- disease is mainly an occlusive vasculitis associ-
tion of BRB. CME in uveitis may persist even ated with retinal capillary non-perfusion and pos-
when intraocular inflammation is stabilized. sible secondary neovascularization [43].
Moreover, in Behet disease, IL-8 has been
demonstrated to increase as the inflammation
2.4.1 Inammatory Markers reactivates, thus suggesting that IL-8 may repre-
sent a marker of the inflammation activity in this
Several studies have demonstrated an increased uveitic process [44, 45].
release of molecules, such as interleukins, tumor Conventional immunosuppressive agents like
necrosis factors, and interferon- (IFN-), lead- intraocular methotrexate may represent a thera-
ing to a chronic inflammatory status [38]. peutic option in patients untreatable with steroid.
Therefore, the therapy of CME in uveitis has Another new option may be offered by anti-tumor
always been represented by steroid treatment: necrosis factor (TNF)- agents, but randomized
either by intravitreal administration or by sub- clinical trials still show questionable results [46].
Tenon injection, as they both provide a signifi- Many of these mediators are active not only in
cant concentration of steroid reaching the uveitis but also in many conditions leading to
posterior segment of the eye [39]. Van Kooij et al. ME, such as DM and RVO. These observations
analyzed the aqueous humor of patients with may suggest that a common pathway is involved
2 Pathophysiology of Macular Edema in Diabetes, Retinal Vein Occlusion, and Uveitis 23

in the genesis of retinal fluid accumulation in dif- 13. Aiello LP, Avery RL, Arrigg PG, Keyt BA, Jampel
HD, Shah ST, Pasquale LR, Thieme H, Iwamoto MA,
ferent clinical situations. More collaborative
Park JE, et al. Vascular endothelial growth factor in
studies are requested to correctly compare macu- ocular fluid of patients with diabetic retinopathy and
lar edema in these different clinical entities. other retinal disorders. N Engl J Med.
1994;331(22):14807.
14. Roberts WG, Palade GE. Increased microvascular
permeability and endothelial fenestration induced by
vascular endothelial growth factor. J Cell Sci.
References 1995;108:236979.
15. Funk M, Schmidinger G, Maar N, Bolz M, Benesch T,
1. Girach A, Lund-Andersen H. Diabetic macular Zlabinger GJ, Schmidt-Erfurth UM. Angiogenic and
oedema: a clinical overview. Int J Clin Pract. 2007;61: inflammatory markers in the intraocular fluid of eyes
8897. with diabetic macular edema and influence of therapy
2. WHO Report. Global prevalence of diabetes mellitus with bevacizumab. Retina. 2010;30:14129.
and its complications. Geneva; 2005. 16. Jonas JB, Sofker A. Intraocular injection of crystal-
3. The Diabetes Control and Complications Trial line cortisone as adjunctive treatment of diabetic mac-
Research Group. The effect of intensive treatment of ular edema. Am J Ophthalmol. 2001;132:4257.
diabetes on the development and progression of long- 17. Martidis A, Duker JS, Greenberg PB, Rogers AH,
term complications in insulin-dependent diabetes Puliafito CA, Reichel E, et al. Intravitreal triamcino-
mellitus. N Engl J Med. 1993;329(14):97786. lone for refractory diabetic macular edema.
4. Shamoon H. Pathophysiology of diabetes. A review Ophthalmology. 2002;109:9207.
of selected recent developments and their impact on 18. Funatsu H, Yamashita H, Ideda T, Mimura T,
treatment. Drugs. 1992;44 Suppl 3:112. Equchi S, Hori S. Vitreous levels of interleukin-6 and
5. Aiello LP, DCCT/EDIC Research Group. Diabetic vascular endothelial growth factor are related to
retinopathy and other ocular findings in the diabetes diabetic macular edema. Ophthalmology. 2003;110:
control and complications trial/epidemiology of dia- 16906.
betes interventions and complications study. Diabetes 19. Lee WJ, Kang MH, Seong M, Cho HY. Comparison
Care. 2014;37(1):1723. of aqueous concentrations of angiogenic and inflam-
6. Anderson JM, Van Itallie CM. Tight junctions and the matory cytokines in diabetic macular oedema and
molecular basis for regulation of paracellular perme- macular oedema due to branch retinal vein occlusion.
ability. Am J Physiol. 1995;269:G46776. Br J Ophthalmol. 2012;0:15.
7. Antonetti DA, Barber AJ, Khin S, Lieth E, Tarbell 20. Sohn HJ, Han DH, Kim IT, Oh IK, Kim KH, Lee DY,
JM, Gardner TW. Vascular permeability in experi- Nam DH. Changes in aqueous concentrations of vari-
mental diabetes is associated with reduced endothelial ous cytokines after intravitreal triamcinolone vs beva-
occludin content: vascular endothelial growth factor cizumab for diabetic macular edema. Am J
decreases occludin in retinal endothelial cells. Penn Ophthalmol. 2011;152:68694.
Diabetes. 1998;47:19539. 21. Sonoda S, Sakamoto T, Yamashita T, Shirasawa M,
8. Barber AJ. A new view of diabetic retinopa- Otsuka H, Sonoda Y. Retinal morphologic changes
thy: a neurodegenerative disease of the eye. Prog and concentrations of cytokines in eyes with diabetic
Neuropsychopharmacol Biol Psychiatry. 2003;27(2): macular edema. Retina. 2014;34(4):7418.
28390. 22. Oh I, Kim S, Oh J, Lee TS, Huh K. Inflammatory and
9. Vujosevic S, Midena E. Retinal layers changes in angiogenic factors in the aqueous humor and the rela-
human preclinical and early clinical diabetic retinopa- tionship to diabetic retinopathy. Curr Eye Res.
thy support early retinal neuronal and Mller cells 2010;35:111627.
alterations. J Diabetes Res. 2013;2013:905058. 23. Yoshida A, Yoshida S, Khalil AK, Ishibashi T,
10. Vujosevic S, Bini S, Midena G, Berton M, Pilotto E, Inomata H. Role of NF-kB-mediated interleukin-8
Midena E. Hyperreflective intraretinal spots in diabet- expression in intraocular neovascularization. Invest
ics without and with nonproliferative diabetic retinop- Ophthalmol Vis Sci. 1998;39:1097106.
athy: an in vivo study using spectral domain OCT. J 24. Owen LA, Hartnett ME. Soluble mediators of diabetic
Diabetes Res. DR Guidelines Expert Working Party macular edema: the diagnostic role of aqueous VEGF
Members. The Royal College of Ophthalmologists. and cytokine levels in diabetic macular edema. Curr
London. 2013;2013:491835. Epub 2013 Dec 9. Diab Rep. 2013;13(4):47680.
11. Murakami T, Yoshimura N. Structural changes in 25. Klein R, Klein BE, Moss SE, Meuer SM. The epide-
individual retinal layers in diabetic macular edema. J miology of retinal vein occlusion: the Beaver Dam
Diabetes Res. 2013;2013:920713. Review. Eye Study. Trans Am Ophthalmol Soc. 2000;98:133
12. Tarr JM, Kaul K, Chopra M, Kohner EM, Chibber 41, discussion 141133.
R. Pathophysiology of diabetic retinopathy. ISRN 26. Rehak J, Rehak M. Branch retinal vein occlusion:
Ophthalmol. 2013;2013:343560. eCollection 2013 pathogenesis, visual prognosis, and treatment modali-
Review. ties. Curr Eye Res. 2008;33:11131.
24 E. Midena and S. Bini

27. Hayreh SS, Zimmerman MB, Podhajsky P. Incidence 37. Yoshimura T, Sonoda KH, Sugahara M, Mochizuki Y,
of various types of retinal vein occlusion and their Enaida H, Oshima Y, Ueno A, Hata Y, Yoshida H,
recurrence and demographic characteristics. Am J Ishibashi T. Comprehensive analysis of inflammatory
Ophthalmol. 1994;117:42941. immune mediators in vitreoretinal diseases. PLoS
28. Campochiaro PA, Heier JS, Feiner L, et al. Ranibizumab One. 2009;4(12):e8158.
for macular edema following branch retinal vein occlu- 38. Schaap-Fogler M, Amer R, Friling R, Priel E,
sion: 6-month primary endpoint results of a phase III Kramer M. Anti-TNF- agents for refractory cystoid
study. Ophthalmology. 2010;117(6):110212. macular edema associated with noninfectious uve-
29. Brown DM, Campochiaro PA, Singh RP, et al. itis. Graefes Arch Clin Exp Ophthalmol. 2014;
Efficacy and safety of ranibizumab in the treatment of 252(4):63340.
macular edema secondary to central retinal vein 39. Bae JH, Lee CS, Lee SC. Efficacy and safety of intra-
occlusion: 6-month results of the phase III CRUISE vitreal bevacizumab compared with intravitreal and
study. Ophthalmology. 2010;117(6):112433. posterior sub-tenon triamcinolone acetonide for treat-
30. Noma H, Funatsu H, Mimura T. Changes of inflam- ment of uveitic cystoid macular edema. Retina.
matory factors after intravitreal triamcinolone aceton- 2011;13(1):1118.
ide for macular edema with central retinal vein 40. van Kooij B, Rothova A, Rijkers GT, de Groot-Mijnes
occlusion. J Ocul Pharmacol Ther. 2013;29:3635. JD. Distinct cytokine and chemokine profiles in the
31. Lin JM, Chiu YT, Hung PT, Tsai YY. Early treatment aqueous of patients with uveitis and cystoid macular
of severe cystoid macular edema in central retinal edema. Am J Ophthalmol. 2006;142(1):1924.
vein occlusion with posterior sub-tenon triamcinolone 41. Soheilian M, Rabbanikhah Z, Ramezani A, Kiavash
acetonide. Retina. 2007;27:1809. V, Yaseri M, Peyman GA. Intravitreal bevacizumab
32. Joshi L, Yaganti S, Gemenetzi M, et al. Dexamethasone versus triamcinolone acetonide for refractory uveitic
implants in retinal vein occlusion: 12-month clinical cystoid macular edema: a randomized pilot study. J
effectiveness using repeat injections as-needed. Br J Ocul Pharmacol Ther. 2010;13(2):199206.
Ophthalmol. 2013;97:10404. 42. Lasave AF, Zeballos DG, El-Haig WM, Diaz-Llopis
33. Haller JA, Bandello F, Belfort Jr R, Blumenkranz MS, M, Salom D, Arevalo JF. Short-term results of a single
Gillies M, Heier J, Loewenstein A, Yoon YH, Jacques intravitreal bevacizumab (avastin) injection versus a
ML, Jiao J, Li XY, Whitcup SM, OZURDEX single intravitreal triamcinolone acetonide (kenacort)
GENEVA Study Group. Randomized, sham- injection for the management of refractory noninfec-
controlled trial of dexamethasone intravitreal implant tious uveitic cystoid macular edema. Ocul Immunol
in patients with macular edema due to retinal vein Inflamm. 2009;13(6):42330.
occlusion. Ophthalmology. 2010;117(6):113446.e3. 43. Cekmen M, Evereklioglu C, Er H, et al. Vascular
34. Sohn HJ, Han DH, Lee DY, Nam DH. Changes in endothelial growth factor levels are increased and
aqueous cytokines after intravitreal triamcinolone associated with disease activity in patients with
versus bevacizumab for macular oedema in branch Behcet syndrome. Int J Dermatol. 2003;42:8705.
retinal vein occlusion. Acta Ophthalmol. 44. al-Dalaan A, al-Sedairy S, al-Balaa S, et al. Enhanced
2014;92(3):e21724. interleukin-8 secretion in circulation of patients with
35. Funk M, Kriechbaum K, Prager F, Benesch T, Behcets disease. J Rheumatol. 1995;22:9047.
Georgopoulos M, Zlabinger GJ, Schmidt-Erfurth 45. Hashida N, Ohguro N, Yamamoto S, Nakagawa Y,
U. Intraocular concentrations of growth factors and Tano Y. Unusual neutrophil infiltration under the soft
cytokines in retinal vein occlusion and the effect of contact lens in a patient with Behcets disease. Jpn J
therapy with bevacizumab. Invest Ophthalmol Vis Ophthalmol. 2003;47:46972.
Sci. 2009;50:102532. 46. Tempest-Roe S, Joshi L, Dick AD, Taylor SR. Local
36. Lim JW. Intravitreal bevacizumab and cytokine levels therapies for inflammatory eye disease in translation:
in major and macular branch retinal vein occlusion. past, present and future. BMC Ophthalmol.
Ophthalmologica. 2010;225:1504. 2013;13(1):39.
Devices for the Delivery
of Steroids to the Eye 3
Raja Narayanan and Baruch D. Kuppermann

Contents
3.1 Introduction
3.1 Introduction 25
3.2 Dexamethasone Drug Delivery System 26 Over the last two decades, there has been an
3.2.1 Dexamethasone 26 extensive increase in the usage of steroids to treat
3.3 Fluocinolone Acetonide Implant 27 macular edema due to diabetic retinopathy,
3.3.1 Fluocinolone 27 venous occlusive disease, ocular inflammation,
3.3.2 New-Generation Fluocinolone Device 27
and also in cases of CNV [14]. Dexamethasone
3.4 Summary and Key Points 28 is used clinically to reduce intraocular inflamma-
References 28 tion and macular edema, but its intraocular half-
life is on the order of hours so its duration of
action in free form is much less than that of
triamcinolone acetonide. However, a dexametha-
sone implant (OZURDEX, Allergan) is currently
approved by the FDA to treat patients with macu-
lar edema due to retinal vein occlusion as well as
for noninfectious posterior uveitis and for dia-
betic macular edema (DME). Fluocinolone ace-
tonide is another steroid in a FDA-approved drug
delivery system (Retisert, Bausch & Lomb) used
for the treatment of noninfectious posterior uve-
itis. Fluocinolone acetonide is also in an inject-
able drug delivery system (Iluvien, Alimera)
approved for the treatment of chronic DME in
several countries in Europe and pending approval
in the United States. Macular edema occurs due
R. Narayanan, MD to changes in retinal capillaries resulting in a
Department of Retina, L.V. Prasad Eye Institute, breakdown of the tight junctions that form the
L.V. Prasad Marg, Hyderabad, Telangana 50034, India blood-retinal barrier and subsequent increased
e-mail: narayanan@lvpei.org
retinal vascular permeability [5]. Chronic, low-
B.D. Kuppermann, MD, PhD (*) grade inflammation of the retinal microvascula-
Department of Ophthalmology,
ture appears to be a significant contributor to this
Gavin Herbert Eye Institute-UC Irvine,
850 Health Sciences Road, Irvine, CA 92697, USA process. The goals of therapy for macular edema
e-mail: bdkupper@uci.edu should be to reduce inflammation and restore

A.J. Augustin (ed.), Intravitreal Steroids, 25


DOI 10.1007/978-3-319-14487-0_3, Springer International Publishing Switzerland 2015
26 R. Narayanan and B.D. Kuppermann

Table 3.1 Relative anti-inflammatory strength of various the treatment of noninfectious uveitis affecting
corticosteroids
the posterior segment of the eye [11] and has just
Steroid Relative potency been granted approval by the FDA for the treat-
Hydrocortisone 1.0 ment of DME in aphakic patients and patients
Prednisolone 4.0 planning to undergo cataract surgery.
Methylprednisolone 5 Key features of the drug delivery system are
Triamcinolone 5 the sustained-release formulation of the
Fluocinolone 25
poly(lactic acid-coglycolic acid) (PLGA) matrix
Dexamethasone 26
material, which dissolves completely in vivo.
Betamethasone 33
Due to the lipophilic nature and the relatively
Data from: Zimmerman et al. [10]
low molecular weight of dexamethasone,
elimination is largely driven by diffusion through
blood-retinal barrier patency. Corticosteroids act the retina. There are two phases of drug release
upon most of the multiple processes in the patho- after the implant administration. The first phase
physiology of macular edema. For example, cor- provides high concentrations of dexamethasone
ticosteroids are capable of inhibiting followed by a second phase in which a low con-
prostaglandin and leukotriene synthesis as well centration of dexamethasone is released, extend-
as interfering with ICAM-1, IL-6, VEGF-, and ing the therapeutic period to 6 months.
SDF-1 pathways [6, 7]. Corticosteroids also have Dexamethasone concentrations in the retina and
been shown to decrease paracellular permeability vitreous humor reach a plateau within days of
and increase tight junction integrity both by administration and are maintained at high levels
directly restoring tight junctional proteins to their for 2 months before declining over subsequent
proper location at the cell border and by increas- months. The vitreoretinal pharmacokinetic pro-
ing the gene expression of those proteins [8, 9]. files are similar between non-vitrectomized and
Table 3.1 shows the relative anti-inflammatory vitrectomized eyes.
potency of different corticosteroids. The OZURDEX GENEVA study consisted of
The use of corticosteroids is indicated in the two identical trials, each including patients with
following ocular diseases: cystoid macular BRVO and CRVO, randomized to receive either
edema, diabetic macular edema, retinal vein sham or one of two strengths of dexamethasone
occlusion, and uveitis. implant (0.35 or 0.7 mg) at baseline [12]. The pri-
mary endpoint for the study, combining the two
identical trials (each containing patients with
3.2 Dexamethasone Drug BRVO and CRVO), was amended to time to
Delivery System achieve at least 15-letter improvement in BCVA.
This time was significantly faster in both of the
3.2.1 Dexamethasone implant-treated groups. In the subgroup analysis
of individuals with CRVO, differences in BCVA
The molecular weight for dexamethasone is existed at days 30, 60, and 90 (29 % vs. 9 % for
392.47 Da. The empirical formula is C22H29FO5. three-line gain at 60 days), but by day 180, the
The plasma half-life of parenterally adminis- treatment group had returned to baseline and the
tered dexamethasone is 34 h. The intravitreal sham group had slightly worsened. Patients
dose of free dexamethasone is typically 1 mg in receiving either the 350 g or the 700 g dose of
0.1 cc. The DEX implant (OZURDEX; Allergan, OZURDEX had a statistically significant
Inc., Irvine, CA) is a novel approach approved by increase in vision based on a three-line or better
the US Food and Drug Administration (FDA) for improvement in visual acuity compared to a sham
the intravitreal treatment of macular edema after treatment. In addition, both doses of OZURDEX
branch or central retinal vein occlusion and for were well tolerated in the studies. Less than 7 %
3 Devices for the Delivery of Steroids to the Eye 27

of patients receiving 700 or 350 g of implantation, 93 % of implanted eyes required


OZURDEX experienced an elevation of intra- cataract surgery compared to 20 % of non-
ocular pressure (IOP) greater than 35 mmHg at implanted eyes.
any time during the 6-month study, and at Pearson and colleagues also studied the effi-
6 months, less than 1 % of patients had an IOP cacy and safety results of the Retisert fluocino-
above 25 mmHg. lone acetonide intravitreal implants in 196 eyes
with persistent or recurrent DME over 4 years
[15]. Patients were randomized 2:1 to receive
3.3 Fluocinolone Acetonide 0.59-mg fluocinolone acetonide implant or stan-
Implant dard of care (SOC additional laser or observa-
tion). The primary efficacy outcome was
3.3.1 Fluocinolone 15-letter improvement in VA at 6 months which
was achieved by 16.8 % of FA implanted eyes as
Fluocinolone acetonide has an empirical formula compared to 1.4 % in the SOC group which was
of C24 H30 F2 O6 and a molecular weight of statistically highly significant, but this difference
452.49 Da. The fluocinolone acetonide intravit- was not maintained, and at the end of 3 years,
real implant (Retisert) contains 0.59 mg of the there was no statistically significant difference
medication. The rate of drug releasing initially is between the two groups.
0.6 ug/day, decreasing over the first month to a Intraocular pressure (IOP) 30 mmHg was
steady rate between 0.3 and 0.4 ug/day over recorded in 61.4 % of implanted eyes (SOC,
approximately 30 months [13]. The concentra- 5.8 %) at any time and 33.8 % required surgery
tion of fluocinolone acetonide was found to be for ocular hypertension by 4 years. Of implanted
relatively constant from the first time point, 2 h, phakic eyes, 91 % (SOC, 20 %) had cataract
through 1 year. In 2005, the FDA approved a extraction by 4 years.
fluocinolone acetonide-containing intraocular
implant (Retisert, Bausch & Lomb, Rochester,
NY) for the treatment of chronic noninfectious 3.3.2 New-Generation Fluocinolone
uveitis, affecting the posterior segment of the Device
eye. The fluocinolone implant reduced the recur-
rence rate from 62 % preimplantation to 4 %, A much smaller, non-bioerodible, extended-
10 %, and 20 % at the 1-, 2-, and 3-year time release fluocinolone acetonide injectable device
points postimplantation in the study eyes [14]. (Iluvien previously known as Medidur, Alimera,
Comparatively, there was a significant increase in Alpharetta, GA), which is injected via a 25-gauge
the recurrence rate in the fellow non-implanted injector in a clinic setting, has been investigated
eyes from 30 % preimplantation to 44 %, 52 %, for the treatment of DME. The Fluocinolone
and 59 % at the 1-, 2-, and 3-year postimplanta- Acetonide for Macular Edema (FAME) studies
tion study time points. The percentage of eyes were two phase III RCTs that collectively ran-
that required systemic medications, periocular domized 956 patients with persistent DME fol-
injections, and topical corticosteroids decreased lowing photocoagulation to receive a 0.2-g/day
from 44 %, 69 %, and 37 %, respectively, preim- implant, a 0.5-g/day implant, or a sham injec-
plantation to 7 %, 4 %, and 18 % 1-year postim- tion [16]. In both treatment groups, approxi-
plantation. At 3 years postimplantation, 78 % of mately 28 % of patients gained vision by 15
implanted eyes required ocular antihypertensive letters or more, as opposed to approximately
drops, and 40 % underwent glaucoma filtering 16 % of sham-treated patients. The benefit was
surgery compared to 36 % of fellow eyes requir- even greater in patients with chronic DME (dura-
ing antihypertensive drops and 2 % of fellow tion 3 years or more), with 34.0 % of patients
eyes requiring filtering surgery. By year 3 post- with chronic DME treated with the 0.2-g/day
28 R. Narayanan and B.D. Kuppermann

implant gaining three or more lines of vision, From screening to 6 months, the proportion of
compared to 13.4 % of patients with chronic patients with visual acuity of at least 70 ETDRS
DME in the sham-treated group. letters (in the study eye) increased from 14 to
Subsequent cataract surgery was reported in 46 % in the slow group and from 18 to 41 % in
80.0 % of patients receiving a 0.2-g/day implant, the fast group. A gain of more than 15 letters
87.2 % of patients receiving a 0.5-g/day implant, occurred in 8 % of patients in the slow group and
and 27.3 % of patients receiving the sham injec- 18 % in the fast group. Both implant formula-
tion. Increased IOP requiring incisional glau- tions were associated with improvements in mac-
coma surgery was reported in 4.8 % of patients ular thickness.
receiving a 0.2-g/day implant, 8.1 % of patients
receiving a 0.5-g/day implant, and only 0.5 % of
patients receiving a sham injection. 3.4 Summary and Key Points
The US FDA initially announced that it would
not approve the Iluvien device for DME. However, In summary, corticosteroids are the one class of
the device has received approval in several agents that act upon most of the multiple pro-
European nations and is being reconsidered for cesses in the pathophysiology of macular edema.
approval by the US FDA. However, despite their significant benefits, the
Triamcinolone acetonide is designated chemi- primary ocular adverse effects associated with the
cally as 9-fluoro-11b,16a,17,21-tetrahydroxypregna- use of steroids IOP rise and cataract are impor-
1,4-diene-3,20-dione cyclic 16,17-acetal with tant issues which may limit their use in certain
acetone. The empirical formula is C24H31FO6 and situations. The need for repeated injections may
the molecular weight is 434.50 Da. be offset by the use of long-term sustained-release
implants. The Iluvien implant is not FDA
3.3.2.1 The STRIDE Study approved in the United States but is approved for
The prospective, randomized, double-masked use in Europe and is being reconsidered for
STRIDE (Sustained Triamcinolone Release for approval by the US FDA. The other extended-
Inhibition of Diabetic Macular Edema) trial is release devices (OZURDEX and Retisert) are
assessing the safety and tolerability of the I-vation FDA approved for indications other than DME,
TA (SurModics, Irvine, Calif.) in 30 patients. The and the OZURDEX implant is currently undergo-
I-vation intravitreal implant is a titanium helical ing review by the FDA for the treatment of DME.
coil coated with TA (925 g) and the nonbiode-
gradable polymers poly(methyl methacrylate)
and ethylene-vinyl acetate. The narrow wire References
diameter of the implant allows for minimally
invasive placement through a 25-g to 30-g needle- 1. Jonas JB, et al. Intravitreal injection of crystalline cor-
tisone as adjunctive treatment of proliferative diabetic
stick. The unique helical shape of the device
retinopathy. Am J Ophthalmol. 2001;131(4):46871.
maximizes the surface area available for the 2. Martidis A, et al. Intravitreal triamcinolone for refrac-
drug-eluting portion of the implant and enables tory diabetic macular edema. Ophthalmology. 2002;
secure, sutureless anchoring of the device against 109(5):9207.
3. Antcliff RJ, et al. Intravitreal triamcinolone for uveitic
the surface of the sclera.
cystoid macular edema: an optical coherence tomog-
In the study, patients are randomized to either raphy study. Ophthalmology. 2001;108(4):76572.
a slow release (1 g/day) or fast release (3 g/ 4. Danis RP, et al. Intravitreal triamcinolone acetonide in
day), each containing 925 g of triamcinolone. exudative age-related macular degeneration. Retina.
2000;20(3):24450.
They also were stratified by baseline visual acu-
5. Ferris 3rd FL, Patz A. Macular edema. A complica-
ity and by presence or absence of prior laser tion of diabetic retinopathy. Surv Ophthalmol.
treatment. 1984;28:45261.
3 Devices for the Delivery of Steroids to the Eye 29

6. Nauck M, et al. Corticosteroids inhibit the expression system in patients with persistent macular edema.
of the vascular endothelial growth factor gene in Arch Ophthalmol. 2007;125(3):30917.
human vascular smooth muscle cells. Eur J Pharmacol. 12. Haller JA, Bandello F, Belfort Jr R, et al. Randomized,
1998;341(23):30915. sham-controlled trial of dexamethasone intravitreal
7. Tamura H, et al. Intravitreal injection of corticosteroid implant in patients with macular edema due to retinal
attenuates leukostasis and vascular leakage in experi- vein occlusion. Ophthalmology. 2010;117(6):113446.
mental diabetic retina. Invest Ophthalmol Vis Sci. 13. Jaffe GJ, Yang CH, Guo H, et al. Safety and pharma-
2005;46(4):14404. cokinetics of an intraocular fluocinolone acetonide
8. Felinski EA, Antonetti DA. Glucocorticoid regulation sustained delivery device. Invest Ophthalmol Vis Sci.
of endothelial cell tight junction gene expression: 2000;41:356975.
novel treatments for diabetic retinopathy. Curr Eye 14. Callanan DG, Jaffe GJ, Martin DF, Pearson PA,
Res. 2005;30(11):94957. Comstock TL. Treatment of posterior uveitis with a
9. Antonetti DA, et al. Hydrocortisone decreases reti- fluocinolone acetonide implant: three-year clinical trial
nal endothelial cell water and solute flux coincident results. Arch Ophthalmol. 2008;126(9):1191201.
with increased content and decreased phosphoryla- 15. Pearson PA, Cornstock TL, Ip M, et al. Fluocinolone
tion of occludin. J Neurochem. 2002;80(4): acetonide intravitreal implant for diabetic macular
66777. edema: a 3-year multicenter, randomized, controlled
10. Zimmerman T, Kooner K, Sharir M, et al. Textbook of trial. Ophthalmology. 2011;118(8):15807.
ocular pharmacology. Philadelphia: Lippincott- 16. Campochiaro PA, Brown DM, Pearson A, et al. Long-
Raven; 1997. p. 6834. term benefit of sustained-delivery fluocinolone ace-
11. Kuppermann BD, et al. Randomized controlled study tonide vitreous inserts for diabetic macular edema.
of an intravitreous dexamethasone drug delivery Ophthalmology. 2011;118(4):62635.e2.
Imaging Before, During,
and After Steroid Therapy 4
Antonio Caimi and Giovanni Staurenghi

Contents 4.5.5 Red-Free Imaging 44


4.5.6 Fundus Autofluorescence Imaging 44
4.1 Introduction 31 4.5.7 Optical Coherence Tomography 45
4.2 History of Fundus Ocular Imaging 32 4.6 Postsurgical Macular Edema 45
4.3 Macular Edema in Retinal Vein 4.6.1 Color Fundus Photography 45
Occlusions 32 4.6.2 Fluorescein Angiography 45
4.3.1 Color Fundus Photography 32 4.6.3 Infrared Imaging 45
4.3.2 Fluorescein Angiography (FA) 32 4.6.4 Red-Free Imaging 45
4.3.3 Infrared Imaging (IR) 33 4.6.5 Fundus Autofluorescence Imaging 45
4.3.4 Red-Free Imaging (RF) 37 4.6.6 Optical Coherence Tomography 47
4.3.5 Fundus Autofluorescence Imaging (FAF) 37 4.7 Macular Edema and Retinal
4.3.6 Optical Coherence Tomography (OCT) 37 Macroaneurysm 48
4.4 Diabetic Macular Edema 37 4.7.1 Color Fundus Photography 48
4.4.1 Color Fundus Photography 37 4.7.2 Fluorescein Angiography 48
4.4.2 Fluorescein Angiography 37 4.7.3 Infrared Imaging 48
4.4.3 Infrared Imaging 38 4.7.4 Red-Free Imaging 48
4.4.4 Red-Free Imaging 38 4.7.5 Fundus Autofluorescence Imaging 48
4.4.5 Fundus Autofluorescence Imaging 38 4.7.6 Optical Coherence Tomography 51
4.4.6 Optical Coherence Tomography 38 References 51
4.5 Uveitic Macular Edema 41
4.5.1 Color Fundus Photography 41
4.5.2 Fluorescein Angiography 41
4.5.3 Indocyanine Green Angiography 41 4.1 Introduction
4.5.4 Infrared Imaging 44
Macular edema complicates numerous retinal
and choroidal diseases. It could lead to perma-
nent and severe visual loss. Therefore, differen-
tial diagnosis is crucial in the management of this
A. Caimi, MD disabling condition. The following section offers
Eye Clinic, Department of Clinical Science, Luigi
Sacco, Luigi Sacco Hospital, University of Milan,
an exhaustive overview on imaging macular
Via G.B. Grassi 74, Milan 20157, Italy edema in different ocular diseases and their treat-
e-mail: antonio.caimi81@gmail.com ment with intravitreal steroids.1
G. Staurenghi, MD (*)
Eye Clinic, Department of Clinical Science, Luigi
1
Sacco, Luigi Sacco Hospital, University of Milan, Every macular edema subtype is illustrated by imaging a
via Muratori, 29, Milan 20135, Italy single numbered patient before and after treatment with
e-mail: giovanni.staurenghi@unimi.it intravitreal steroids.

A.J. Augustin (ed.), Intravitreal Steroids, 31


DOI 10.1007/978-3-319-14487-0_4, Springer International Publishing Switzerland 2015
32 A. Caimi and G. Staurenghi

4.2 History of Fundus Ocular The venous thrombosis elevates intravascular


Imaging pressure distal to the occlusion site inducing
vasodilation and venous stasis. This results in
Fundus ocular imaging was born in 1851 with the transudation of fluid into the extracellular space.
invention of the direct ophthalmoscope by In addition, upregulation of proinflammatory
Hermann von Helmholtz [1]; only in 1886, cytokines and breakdown of blood-retinal barrier
Jackman and Webster published the first attempt could facilitate intraretinal exudation [1113].
of in vivo human retinal photography [2].
A great progress in ocular imaging was reached
in 1961 when two medical students, Novotny and 4.3.1 Color Fundus Photography
Alvis, first described retinal fluorescein angiogra- (Fig. 4.1a, b)
phy (FA) [3]. This new technique allowed a better
understanding of vascular diseases of the retina. Fundus examination shows variable amount of
Furthermore in 1970, using the filters for FA, intraretinal blot and/or flame-shaped hemor-
the hyper-fluorescence coming from the fluoro- rhages, engorged retinal veins, and cotton wool
phores of lipofuscin granules in the retinal spots.
pigment epithelium was noted [4]. This brilliant The distribution of these findings depends on
observation opened the door to the modern fun- the site of the occlusion: typically one sector in
dus autofluorescence (FAF) imaging, a precious branch retinal vein occlusion (BRVO), hemiret-
tool in the study of retinal diseases. ina in hemiretinal vein occlusion (HRVO), and
Pomerantzeff opened the era of wide-field all the fundus in central retinal vein occlusion
retinal imaging in 1975 when he reported the use (CRVO).
of contact lens-based device that allowed visu- Notably, the severity of intraretinal hemor-
alization up to 148 of the retina with pupillary rhages and cotton wool spots seems to correlate to
dilation [5]. extension of retinal capillary non-perfusion [14].
In the early 1980s, Webb and associates devel- Additionally in BRVO, the arteriovenous
oped a new retinal imaging technique: the scan- crossing site of the occlusion and in CRVO optic
ning laser ophthalmoscope (SLO) [6]. nerve head swelling and hyperemia are often
Another great step forward in ocular imaging visible.
came in the 1990s [7], when optical coherence Cystoid macular edema appears as cystic
tomography was developed thanks to the efforts spaces associated with loss of physiologic foveal
of Fujimoto and Puliafito. This is a fast, noninva- reflex.
sive, radiation-free technique that provides in vivo
cross-sectional visualization of the human retina.
The attempt to integrate SLO and OCT technol- 4.3.2 Fluorescein Angiography (FA)
ogies was started in the late 1990s by Podoleanu (Fig. 4.1cf)
and colleagues [8]. This idea laid the foundation
of modern multimodal imaging concept. Dynamic fluorescein angiography shows a vari-
Today, multimodal imaging combined with able amount of retinal vein filling delay down
spectral domain OCT and wide-field imaging [9] line the site of occlusion. Hypo-fluorescent
could allow a better understanding and manage- areas that correspond to retinal capillary non-
ment of retinal and choroidal disorders. perfusion, hemorrhages, and intraretinal exu-
dates could characterize early phases. In mid
to late phases the petaloid pattern of CME is
4.3 Macular Edema in Retinal visible as pooling in the intraretinal cystoid
Vein Occlusions spaces.
Sometimes in CRVO hot disk and vascular
Macular edema is one of the leading causes of peripheral leakage from the veins are present;
visual loss in patients affected by retinal vein opto-ciliary shunt vessels could develop on the
occlusion [10]. top of the optic nerve head in long-standing
4 Imaging Before, During, and After Steroid Therapy 33

CRVO. Unlike neo-vessels, they typically do not 4.3.3 Infrared Imaging (IR)
leak on FA. (Fig. 4.1g, h)
In the chronic phase of the disease, areas of cap-
illary non-perfusion could be associated with micro- Intraretinal fluid and hemorrhages appear as
vascular abnormalities such as microaneurysms and hypo-reflective areas spreading on the site of
collateral vessels. These areas could increase over the occlusion, while cotton wool spots and
time leading to retinal, optic nerve, and/or anterior hard exudates are hyper-reflective ill-defined
segment neovascularization; then neovascular glau- lesions that follow the course of retinal nerve
coma and hemovitreous could occur. fibers.

a b

Fig. 4.1 (a) (Patient 1) Central retinal vein occlusion and intraretinal vascular abnormalities (purple arrow). (i)
(color fundus photography): intraretinal blot (orange (Patient 1) RF: cotton wool spots (yellow arrow), flame-
arrow) and flame-shaped (red arrow) hemorrhages, shaped hemorrhages (red arrow), and cystoid macular
engorged retinal veins (blue arrow), cotton wool spots edema (green arrow). (j) (Patient 2) RF: hard exudates
(yellow arrow), and optic nerve head swelling (purple (orange arrow). (k) (Patient 1) FAF: cotton wool spots
arrow). (b) (Patient 2) Branch retinal vein occlusion (yellow arrow) and blot hemorrhages (red arrow). (l)
(color fundus photography): arteriovenous crossing site (Patient 2) FAF: hard exudates (orange arrow) and cys-
(red arrow), hard exudates (yellow arrow), intraretinal toid macular edema (green arrow). (m) (Patient 1)
vascular abnormalities (orange arrow), and hyaline reti- Macular edema in central retinal vein occlusion, baseline
nal vein wall (blue arrow). (c) (Patient 1) Early FA: hot OCT: cotton wool spots (yellow arrow), intraretinal cysts
disk (purple arrow), masking effect of cotton wool spots (green arrow), and subfoveal serous neuro-retinal detach-
(yellow arrow) and flame-shaped hemorrhages (red ment (brown arrow). (n) (Patient 2) Macular edema in
arrow), and engorged retinal veins (blue arrow). (d) branch retinal vein occlusion, baseline OCT: hard exu-
(Patient 1) Late FA: extensive area of capillary non-per- dates (orange arrow), cystoid macular edema (green
fusion (green arrow), masking effect of flame-shaped arrow), and incomplete posterior vitreous detachment
hemorrhages (red arrow), diffuse breakdown of blood- (blue arrow). (o) (Patient 1) Macular edema in central
retinal barrier (brown arrow), and leakage from retinal retinal vein occlusion, OCT follow-up during 6 months
veins (sky blue arrow). (e, f) (Patient 2) 4.1e early FA and after treatment with dexamethasone intravitreal implant:
4.1f late FA: arteriovenous crossing site (red arrow), resolution of cotton wool spots, serous retinal detach-
masking effect of hard exudates (yellow arrow), intrareti- ment, and macular edema. (p) (Patient 2) Macular edema
nal vascular abnormalities (orange arrow), area of capil- in branch retinal vein occlusion, OCT follow-up during
lary non-perfusion (blue arrow), and pooling of cystoid 6 months after treatment with dexamethasone intravitreal
macular edema (green arrow). (g) (Patient 1) IR: cotton implant: normalization of foveal contour, residual macu-
wool spots (yellow arrow) and intraretinal fluid (brown lar hard exudates, and defects in inner segment/outer
arrow). (h) (Patient 2) IR: hard exudates (orange arrow) segment junction
34 A. Caimi and G. Staurenghi

c d

e f

g h

Fig. 4.1 (continued)


4 Imaging Before, During, and After Steroid Therapy 35

i j

k l

Fig. 4.1 (continued)


36 A. Caimi and G. Staurenghi

Fig. 4.1 (continued)


4 Imaging Before, During, and After Steroid Therapy 37

4.3.4 Red-Free Imaging (RF) 4.4.1 Color Fundus Photography


(Fig. 4.1i, j) (Fig. 4.2a)

It shows hemorrhages as dark areas along the According to the Early Treatment Diabetic
thrombotic vessel often in a flame-shaped distri- Retinopathy Study (ETDRS) [17], diabetic mac-
bution. As in IR imaging, cotton wool spots and ular edema (DME) is defined as retinal thicken-
hard exudates are hyper-reflective. ing or presence of hard exudates within 1 disk
diameter of the center of the macula. In order to
state a severity scale, the study defined clinically
4.3.5 Fundus Autouorescence significant macular edema (CSME) when one of
Imaging (FAF) (Fig. 4.1k, l) the following conditions occurs: retinal thicken-
ing within 500 of the center of the macula, hard
Hemorrhages and cotton wool spots appear as exudates within 500 of the center of the mac-
hypo-autofluorescent areas due to the mask- ula with adjacent retinal thickening, and retinal
ing effect on the physiologic autofluorescence thickening at least 1 disk area in size, any part of
coming from RPE. Cystoid spaces of macular which is within 1 disk diameter of the center of
edema are visible as hyper-autofluorescent the macula.
oval-shaped areas due to the shifting of mac- The Global Diabetic Retinopathy Project
ular pigment from the fovea by intraretinal Group [18] defined DME as retinal thickening or
fluid. lipid exudates in the macula basing on slit-lamp
biomicroscopy and/or stereo fundus photogra-
phy. It was classified as mild, moderate, or severe
4.3.6 Optical Coherence depending on distance of the thickening or exu-
Tomography (OCT) dates from the fovea.
(Fig. 4.1mp) Different studies used the terms focal and dif-
fuse to differentiate two subtypes of DME basing
It shows intra- and subretinal accumulation of on the area of retinal thickening [19, 20].
fluid as cystoid spaces located in all retina layers Despite these efforts, the two subtypes have
and sometimes a serous neurosensory retinal not yet been defined univocally in the literature
detachment. In BRVO and HRVO, edema is lim- [21].
ited to the portion of the macula affected, while
in CRVO, it is typically diffuse. When the CME
becomes chronic, OCT could reveal various 4.4.2 Fluorescein Angiography
amounts of retinal pigment epithelium atrophic (Fig. 4.2b, d)
changes, subfoveal fibrosis, intraretinal exuda-
tion, and epiretinal membrane. Fluorescein angiography (FA) is essential in the
study of patients with DME. It allows detecting
fluid leakage. Notably leakage itself is not syn-
4.4 Diabetic Macular Edema onymous with retinal thickening or edema [22].
FA is also useful in evaluating foveal avascu-
Diabetic macular edema (DME) is the most com- lar zone (FAZ) enlargement and ischemic macu-
mon cause of visual loss in patient affected by lopathy that could lead to permanent and severe
diabetes mellitus [15]. visual loss.
Its incidence increases with older onset of dia- According to ETDRS [23], clinically signifi-
betes and with higher grade of retinopathy sever- cant macular edema is classified into focal or dif-
ity [16]. fuse, depending on the leakage pattern.
38 A. Caimi and G. Staurenghi

Eyes with 67 % of leakage associated with 4.4.5 Fundus Autouorescence


microaneurysms were classified as focal, those Imaging (Fig. 4.2g)
with 3366 % of leakage from the same source as
intermediate, and those with <33 % as diffuse. Intraretinal cysts of DME could be detected as
Early FA phase shows hyper-fluorescent hyper-autofluorescent oval-shaped lesions sur-
lesions such microaneurysms, intraretinal micro- rounding the fovea. This appearance is probably
vascular anomalies (IRMAS), neo-vessels, and due to the displacement of xanthophyll pigment
hypo-fluorescent lesions such hard exudates, cot- by the intraretinal fluid cavities [26].
ton wool spots, and hemorrhages. Microaneurysms, IRMAS, neo-vessels, pre- or
Foveal avascular zone and non-perfused areas intraretinal hemorrhages, and hard exudates can
are easily detectable by means of dynamic be visualized as hypo-autofluorescent structures.
angiography.
Diabetic macular edema becomes evident in
mid to late phase as pooling of the cystoid intra- 4.4.6 Optical Coherence
retinal spaces with a honeycomb appearance Tomography (Fig. 4.2h, i)
(cysts in the inner nuclear layer on OCT) or pet-
aloid disposition (cysts in the outer plexiform OCT is a noninvasive helpful tool in diagnosis
layer on OCT) [24, 25]. and follow-up of patients affected by DME.
Pathologic accumulation of extracellular fluid
may result in alterations of the normal retinal
4.4.3 Infrared Imaging (Fig. 4.2e) architecture: small amount of fluid could lead to
global increase in retinal thickening, bigger one,
Intraretinal fluid and hemorrhages appear hypo- to characteristic cystoid spaces [27].
reflective, while microaneurysms and hard exu- As in histologic studies sponge-like retinal
dates appear hyper-reflective. thickening mainly occurs in the OPL of the mac-
ula; cystoid spaces are primarily present in the
inner nuclear layer (INL) and outer plexiform
4.4.4 Red-Free Imaging (Fig. 4.2f) layer (OPL) [2830].
Numerous OCT patterns of DME have been
Hard exudates, microaneurysms, and hemorrhages described [3133].
appear hypo-reflective. CME sometimes is visual- Kim and colleagues [34] proposed a compre-
ized as hyper-reflective round perifoveal areas. hensive OCT classification of DME.

Fig. 4.2 (a) (Patient 3) Clinically significant, center- with PRP due to high-risk proliferative diabetic retinopa-
involved diabetic macular edema (color fundus photogra- thy. (e) Clinically significant, center-involved, diffuse dia-
phy): intraretinal blot hemorrhages (yellow arrow) and betic macular edema (IR): hard exudates (yellow arrows)
hard exudates (orange arrows). (b) Clinically significant, and microaneurysms (red arrows). (f) RF: hard exudates
center-involved, diffuse diabetic macular edema (early (yellow arrows). (g) FAF: displacement of macular pig-
FA): foveal avascular zone enlargement (red arrow), ment by intraretinal cysts (green arrow). (h) Clinically
microaneurysms (purple arrow), and blot hemorrhages significant, center-involved, diffuse diabetic macular
(brown arrow). (c) Clinically significant, center-involved, edema baseline OCT: intraretinal cystic spaces (green
diffuse diabetic macular edema (late FA): extensive arrows) and hard exudates (yellow arrow) with back-
blood-retinal barrier rupture (yellow arrows) and late shadowing effect (purple arrow). (i) Diffuse diabetic
pooling of cystoid macular edema (green arrow). (d): macular edema, OCT follow-up during 6 months after
Severe, nonproliferative diabetic retinopathy with incom- treatment with dexamethasone intravitreal implant: pro-
plete pan-retinal photocoagulation (PRP) and diffuse gressive restoration of foveal contour and diminishing of
clinically significant macular edema (mid to late FA com- hard exudates
posite). The right eye of the patient was already treated
4 Imaging Before, During, and After Steroid Therapy 39

b c

d
40 A. Caimi and G. Staurenghi

e f g

Fig. 4.2 (continued)


4 Imaging Before, During, and After Steroid Therapy 41

According to this, five patterns can be found: snow balls, snow banking, perivascular segmen-
1. Diffuse retinal thickening characterized by tal or continuous sheathing, optic nerve head
increase in retinal thickness with reduced edema, and cystoid macular edema.
intraretinal reflectivity, particularly in the
outer retinal layers
2. Cystoid macular edema characterized by 4.5.2 Fluorescein Angiography
hypo-reflective cavities separated by highly (Fig. 4.3b, c)
reflective septa
3. Posterior hyaloid traction characterized by a Fluorescein angiography (FA) is essential in doc-
highly reflective band over the retina umenting the fundus alteration induced by under-
surface lying inflammatory process and its activity level.
4. Serous retinal detachment characterized by a Typical signs of posterior uveitis are late pool-
hypo-reflective dome-shaped space between ing of dye in cystoid macular edema, late staining
the retinal pigment epithelium and and leakage from optic nerve (hot disk) and vas-
neuro-retina culitic vessels, and diffuse leakage due to the rup-
5. Posterior hyaloidal traction and tractional ture of blood-retinal barrier.
retinal detachment resulting from a combina- The FA is useful in detecting active inflamma-
tion of pattern 3 and 4 tory foci (early hypo-fluorescence and late hyper-
Microaneurysms and dot hemorrhages appear fluorescence), choroidal neovascularization
as spots in the inner retina and hard exudates as (early hyper-fluorescence and late staining and
round lesions in the outer retinal layers; all these leakage), and progressive retinal scarring (early
lesions including cotton wool spots show hyper- hypo-fluorescence and late leakage from the bor-
reflective appearance on B-scan OCT with ders of active lesions).
back-shadowing. Furthermore, it is helpful in distinguishing
The integrity of the inner segment/outer seg- occlusive from nonocclusive vacuities; the first
ment junction [3541] and the external limiting one is characterized by areas of retinal ischemia
membrane [4042] seems to be clinically rele- sometimes surrounded by capillary abnormalities
vant in the interpretation of DME on OCT. with late leakage of dye. This process could
involve the posterior pole leading to permanent
and severe visual loss.
4.5 Uveitic Macular Edema

Cystoid macular edema represents the most fre- 4.5.3 Indocyanine Green
quent cause of vision loss in patients affected by Angiography (Fig. 4.3d)
uveitis [43]. In fact, it can be found in about 30 %
of patients [44]. It could complicate infectious, Due to its peculiar physical properties, indocyanine
autoimmune, toxic, and idiopathic uveitis [11]. green angiography (ICGA) is a precious tool in dis-
tinguishing inflammatory choriocapillaropathies
(such as white dots syndromes) and stromal choroi-
4.5.1 Color Fundus Photography ditis; [45] the first one is characterized by hypo-
(Fig. 4.3a) fluorescent areas of choriocapillary non-perfusion,
the second one by diffuse choroidal hyper-fluores-
Depending on the main site of inflammatory pro- cence in mid to late phase due to inflammatory vas-
cess, fundus examination could reveal many dif- culopathy of the bigger choroidal vessels.
ferent lesions: white dots, yellow creamy spots, In addition, ICGA findings seem to correlate
chorioretinal scars, intraretinal infiltrates, hard to visual field testing in patients affected by cho-
and/or cotton wool exudates, flame-shaped and riocapillaropathies, allowing a more accurate
dot/blot hemorrhages, retinal whitening, vitreitis, follow-up and therapeutic management [46].
42 A. Caimi and G. Staurenghi

b c

Fig. 4.3 (a) (Patient 4) Idiopathic segmental periphlebi- (f) RF: foveal irregularity (green arrow) due to macular
tis (color fundus photography): segmental perivenular edema and segmental perivenular sheathing (purple
sheathing (purple arrows) and foveal irregular appearance arrow). (g) FAF: foveal cysts (green arrow) due to macu-
(green arrow) due to macular edema. (b) Early FA: peri- lar edema. (h) Macular edema in idiopathic segmental
foveal capillary leakage (red arrow). (c) Late FA: hot disk periphlebitis, baseline OCT: cystoid macular edema
(yellow arrow), segmental perivenular sheathing (purple (green arrow), subfoveal retinal pigment epithelium irreg-
arrows), leakage from vasculitic vessels (brown arrow), ularity (yellow arrow), and incomplete posterior vitreous
diffuse breakdown of blood-retinal barrier (blue arrow), detachment (blue arrow). (i) Macular edema in idiopathic
and pooling of cystoid macular edema (green arrow). (d) segmental periphlebitis, OCT follow-up during 6 months
Mid-phase ICGA: foveal dishomogeneity (sky blue after treatment with dexamethasone intravitreal implant:
arrow) due to macular edema, no choroidal or retinal pig- progressive normalization of foveal contour, residual reti-
ment epithelium permeability abnormalities. (e) IR: nal pigment epithelium irregularity, and incomplete poste-
foveal irregularity (green arrow) due to macular edema. rior vitreous detachment
4 Imaging Before, During, and After Steroid Therapy 43

e f g

Fig. 4.3 (continued)


44 A. Caimi and G. Staurenghi

Fig. 4.3 (continued)

The late phase of ICGA imaging could distin- 4.5.5 Red-Free Imaging (Fig. 4.3f)
guish between full-thickness choroidal infiltrates
(late hypo-fluorescence) and partial thickness In red-free cotton wool spots, perivascular
(late iso-fluorescence). sheathing and hard exudates appear hyper-
Finally, ICGA is a reliable instrument in fol- reflective; intra- or preretinal hemorrhages are
lowing up Vogt-Koyanagi-Harada disease and ser- hypo-reflective. Secondary epiretinal membranes
piginous choroiditis; these diseases, in fact, could are easily detected by the presence of typical
present occult choroidal lesions and progression inner retinal folds of the surface. When the media
without clinical or FA counterparts [47, 48]. are clear, CME is usually detected as hyper-
reflective oval-shaped areas around the fovea.

4.5.4 Infrared Imaging (Fig. 4.3e)


4.5.6 Fundus Autouorescence
Infrared imaging shows a macula with irregular Imaging (Fig. 4.3g)
appearance in case of macular edema. Intraretinal
fluid and hemorrhages are hypo-reflective, while Uveitic CME in FAF appears as hyper-
hard exudates are usually hyper-reflective. autofluorescent round areas with a typical radial
4 Imaging Before, During, and After Steroid Therapy 45

distribution respect to the fovea; this aspect Patient affected by clinically significant
seems to be related to displacement of macular ME complain a decrease in visual acuity gen-
pigment by intraretinal cystoid spaces [49]. erally during 412 weeks after surgery with a
Increase FAF signal is documented also around peak of onset at 46 weeks postoperatively
active choroidal neovascularization that might [53].
complicate the course of posterior uveitis [50].
Finally, FAF imaging could show in one shot
the early atrophic alterations of the retinal pig- 4.6.1 Color Fundus Photography
ment epithelium non-detectable with color fun- (Fig. 4.4a)
dus photography and the overall RPE damage.
This is helpful in evaluating the timing and the On biomicroscopic examination loss of foveal
aggressiveness of treatment required in these depression occurs, with intraretinal cystoid
patients. spaces and often optic nerve head swelling.
Sometimes the alteration of fundus reflex and the
wrinkling of retinal surface associated to epireti-
4.5.7 Optical Coherence nal membrane are present.
Tomography (Fig. 4.3h, i)

OCT is useful in detecting uveitic macular 4.6.2 Fluorescein Angiography


edema, epiretinal membranes, and vitreoretinal (Fig. 4.4b, c)
interface abnormalities such as vitreo-macular
traction syndrome that could complicate the Early phase of FA shows leakage from the perifo-
course of the disease. veal capillaries with late pooling of cysts: typical
According to Markomichelakis and col- petaloid like appearance in the foveal region is
leagues [51], three different patterns of fluid frequently surrounded by a honeycomb pooling
accumulation could be found on OCT: pattern at posterior pole.
Diffuse macular edema (spongy appearance of
the thickened retina)
Cystoid macular edema (well-defined intra- 4.6.3 Infrared Imaging (Fig. 4.4d)
retinal cystoid spaces)
Serous retinal detachment (separation of the IR is characterized by nonhomogeneous aspect
neurosensory retina from the retinal pigment of the foveal region.
epithelium)
In addition, chronic CME could be associated
to foveal atrophy and subretinal fibrosis. 4.6.4 Red-Free Imaging (Fig. 4.4e)

RF imaging could reveal hyper-reflective round


4.6 Postsurgical Macular Edema lesions corresponding to intraretinal cysts.

Macular edema (ME) could occur after any intra-


ocular surgery. It still represents one of the major 4.6.5 Fundus Autouorescence
complications after both cataract surgery and Imaging (Fig. 4.4f)
pars plana vitrectomy.
Although the pathophysiology of this condition FAF signal is increased with the typical hyper-
is not completely understood, inflammation seems autofluorescent round-shaped cysts due to the
to play a crucial role in its development [11, 52]. shifting of pigment from the macula.
46 A. Caimi and G. Staurenghi

b c

d e f

Fig. 4.4 (a) (Patient 5) Postsurgical macular edema after and dissociated optical nerve fiber layer (brown arrows).
25 gauge pars plana vitrectomy with peeling of epiretinal (f) FAF: displacement of macular pigment by intraretinal
membrane and inner limiting membrane (color fundus cysts (green arrow). (g) Postsurgical macular edema base-
photography): foveal irregular appearance (green arrow) line OCT: wide intraretinal cystic spaces (green arrow)
due to macular edema. (b) Early FA: blood-retinal barrier and subfoveal inner segment/outer segment junction
rupture (sky blue arrows). (c) Late FA: blood-retinal bar- defects (orange arrow). (h) Postsurgical macular edema,
rier rupture (sky blue arrows), late pooling of intraretinal OCT follow-up during 6 months after treatment with
cysts (yellow arrow), and hot disk (orange arrow). (d) IR: dexamethasone intravitreal implant: progressive reduction
nonhomogeneous foveal appearance (green arrow). (e) of macular edema; at 6 months some small intraretinal
RF: nonhomogeneous foveal appearance (green arrow) cysts still persist
4 Imaging Before, During, and After Steroid Therapy 47

Fig. 4.4 (continued)

4.6.6 Optical Coherence cystoid spaces coalesce in foveal macrocysts that


Tomography could evolve into a lamellar macular hole; addi-
tionally irregularity of the inner segment/outer
OCT shows retinal thickening with cystic spaces segment junction (IS/OS), atrophic changes in
especially located in the outer nuclear layer of the subfoveal RPE, and the development of an epireti-
fovea and eventual subfoveal neuro-retinal detach- nal membrane or vitreo-macular traction could
ment [54]. In case of chronic CME, sometimes, occur.
48 A. Caimi and G. Staurenghi

4.7 Macular Edema and Retinal effect on the background fluorescence. Sometimes,
Macroaneurysm distal to the dilatation, arterial occlusion with the
corresponding area of capillary non-perfusion is
By definition, retinal macroaneurysm is an acquired visible.
dilatation of the large arterioles of the retina [55]. In the late phase, leakage from the lesion and
Clinically, they can be classified as quiescent, pooling of intraretinal cystic spaces are visible.
exudative, or hemorrhagic [56]. Both exudative
and hemorrhagic could present with macular
edema due to the increase in vascular permeabil- 4.7.3 Infrared Imaging (Fig. 4.5d)
ity of the lesion.
Infrared imaging is useful in detecting the lesion
especially when a dense hemorrhage occurs [58].
4.7.1 Color Fundus Photography In fact, the longer wavelength of IR allows a bet-
ter visualization through media opacity.
It shows saccular and fusiform dilatation of the
retinal artery with a preferential location on the
temporal side usually within the first three orders 4.7.4 Red-Free Imaging (Fig. 4.5e)
of bifurcation.
The lesion could be associated with intra- or sub- Hard exudates, cotton wool spots, and the mac-
retinal exudation; pre-, intra-, or subretinal blood; roaneurysm itself are hyper-reflective. In the
cystoid macular edema; perilesional retinal thicken- chronic phase of the disease, sometimes retinal
ing; and serous retinal detachment; venous and arte- wrinkling induced by concurrent epiretinal mem-
rial occlusions have also been reported [57]. brane is evident.

4.7.2 Fluorescein Angiography 4.7.5 Fundus Autouorescence


(Fig. 4.5b, c) Imaging (Fig. 4.5f)

In the early phase of FA, there is a rapid filling of The macroaneurysm appears as dark hypo-
the lesion. Blood and hard exudates have masking autofluorescent area along the vascular arcade.

Fig. 4.5 (a) (Patient 6) Retinal macroaneurysm (color retinal macroaneurysm, OCT follow-up during 6 months
fundus photography): arterial saccular dilatation (red after treatment with dexamethasone intravitreal implant:
arrow), perilesional hard exudates (blue arrow), and progressive decrease in retinal thickness and hard exu-
foveal and perifoveal hard exudates (yellow arrow). (b, c) dates; at 6 months subfoveal deposition of hard exudates
4.5b early FA and 4.5c late FA: filling and early leakage still persists. (g) (Patient 6) Retinal macroaneurysm
(red arrow), late diffuse leakage (blue arrow) of arterial (OCT): hyper-reflective vascular wall (brown arrow),
macroaneurysm, and pooling of cystoid macular edema medium- to high-reflectivity intravascular blood (red
(green arrow). (d) IR: foveal (blue arrow) and perifoveal arrow), hard exudates (sky blue arrow), perilesional reti-
hard exudates (yellow arrow) and arterial saccular dilata- nal thickening (orange arrow), and back-shadowing (yel-
tion (brown arrow). (e) RF: foveal irregularity (sky blue low arrow). (h) Macular edema in retinal macroaneurysm
arrow) due to macula edema, perifoveal hard exudates baseline OCT: perifoveal (purple arrow) and subfoveal
(yellow arrow), and hyper-reflectant arterial saccular dila- (orange arrow) hard exudates, intraretinal cystic spaces
tation (brown arrow). (f) FAF: foveal cysts (green arrow) (green arrow), and subfoveal neuro-retinal detachment
due to macula edema and hypo-autofluorescence arterial (blue arrow)
saccular dilatation (brown arrow). (i) Macular edema in
4 Imaging Before, During, and After Steroid Therapy 49

b c

d e f
50 A. Caimi and G. Staurenghi

Fig. 4.5 (continued)


4 Imaging Before, During, and After Steroid Therapy 51

Old pre, intra, or sub retinal bleeding usually 10. Coscas G, Loewenstein A, Augustin A, Bandello F,
Parodi MB, Lanzetta P, Mons J, de Smet M, Soubrane
present an hyper-autofluorescent appearance due
G, Staurenghi G. Management of retinal vein occlu-
to degradation of hemoglobin. sion consensus document. Ophthalmologica.
2011;226:428.
11. Johnson MW. Etiology and treatment of macular
edema. Am J Ophthalmol. 2009;147(1):1121.
4.7.6 Optical Coherence
12. Tranos PG, Wickremasinghe SS, Stangos NT,
Tomography (Fig. 4.5gi) Topouzis F, Tsinopoulos I, Pavesio CE. Macular
edema. Surv Ophthalmol. 2004;49(5):47090.
OCT usually shows an oval lesion located in the 13. Cunha-Vaz J, Coscas G. Diagnosis of macular edema.
Ophthalmologica. 2010;224 Suppl 1:27.
inner retina with hyper-reflective wall corre-
14. Hayreh SS. Prevalent misconceptions about acute
sponding to the dilatation and hypo-reflective retinal vascular occlusive disorders. Prog Retin Eye
content; it elevates the internal limiting mem- Res. 2005;24:493519.
brane and retinal ganglion cell layer [58]. 15. Bhagat N, Grigorian RA, Tutela A, Zarbin
MA. Diabetic macular edema: pathogenesis and treat-
The lesion is often surrounded by retinal
ment. Surv Ophthalmol. 2009;54(1):132.
thickening, hard exudates (as hyper-reflective 16. Aiello LP, Gardner TW, King GL, Blankenship G,
dots in the outer retina with back-shadowing), Cavallerano JD, Ferris 3rd FL, Klein R. Diabetic reti-
and blood (as hyper-reflective area with nopathy. Diabetes Care. 1998;21(1):14356.
17. Photocoagulation for diabetic macular edema. Early
back-shadowing).
Treatment Diabetic Retinopathy Study report number 1.
Macular edema and subfoveal serous retinal Early Treatment Diabetic Retinopathy Study research
detachment could be visualized. group. Arch Ophthalmol. 1985;103(12):1796806.
18. Wilkinson CP, Ferris 3rd FL, Klein RE, Lee PP,
Agardh CD, Davis M, Dills D, Kampik A,
Pararajasegaram R, Verdaguer JT, Global Diabetic
Retinopathy Project Group. Proposed international
References clinical diabetic retinopathy and diabetic macular
edema disease severity scales. Ophthalmology.
1. von Helmholtz HLF. Beschreibung eines 2003;110(9):167782.
Augenspiegels. Berlin: A Frstner Sche 19. Laursen ML, Moeller F, Sander B, Sjoelie
Verlagsbuchhandlung; 1851. AK. Subthreshold micropulse diode laser treatment in
2. Jackman WT, Webster JD. On photographing the ret- diabetic macular oedema. Br J Ophthalmol.
ina of the living human eye. Phila Photogr. 2004;88(9):11739.
1886;23:2756. 20. Kang SW, Park CY, Ham DI. The correlation between
3. Novotny HR, Alvis DL. A method of photographing fluorescein angiographic and optical coherence tomo-
fluorescence in circulating blood in the human retina. graphic features in clinically significant diabetic mac-
Circulation. 1961;24:826. ular edema. Am J Ophthalmol. 2004;137(2):31322.
4. Machemer R, Norton EW, Gass JD, Choromokos 21. Browning DJ, Altaweel MM, Bressler NM, Bressler
E. Pseudofluorescencea problem in interpretation of SB, Scott IU. Diabetic Retinopathy Clinical Research
fluorescein angiograms. Am J Ophthalmol. Network. Diabetic macular edema: what is focal and
1970;70(1):110. what is diffuse? Am J Ophthalmol. 2008;146(5):649
5. Pomerantzeff O. Equator-plus camera. Invest 55, 655.
Ophthalmol. 1975;14(5):4016. 22. Kinyoun J, Barton F, Fisher M, Hubbard L, Aiello L,
6. Webb RH, Hughes GW, Delori FC. Confocal scanning Ferris 3rd F. Detection of diabetic macular edema.
laser ophthalmoscope. Appl Opt. 1987;26(8):14929. Ophthalmoscopy versus photography-Early
7. Huang D, Swanson EA, Lin CP, Schuman JS, Stinson Treatment Diabetic Retinopathy Study report number
WG, Chang W, Hee MR, Flotte T, Gregory K, 5. The ETDRS Research Group. Ophthalmology.
Puliafito CA, et al. Optical coherence tomography. 1989;96(6):74650.
Science. 1991;254(5035):117881. 23. Focal photocoagulation treatment of diabetic macular
8. Podoleanu AG, Seeger M, Dobre GM, Webb DJ, edema. Relationship of treatment effect to fluorescein
Jackson DA, Fitzke FW. Transversal and longitudinal angiographic and other retinal characteristics at base-
images from the retina of the living eye using low coher- line: ETDRS report no. 19. Early Treatment Diabetic
ence reflectometry. J Biomed Opt. 1998;3(1):1220. Retinopathy Study Research Group. Arch Ophthalmol.
9. Staurenghi G, Viola F, et al. Scanning laser ophthal- 1995;113(9):114455.
moscopy and angiography with a wide-field contact 24. Otani T, Kishi S. Correlation between optical coher-
lens system. Arch Ophthalmol. 2005;123(2):24452. ence tomography and fluorescein angiography find-
52 A. Caimi and G. Staurenghi

ings in diabetic macular edema. Ophthalmology. 39. Otani T, Yamaguchi Y, Kishi S. Correlation between
2007;114(1):1047. visual acuity and foveal microstructural changes in
25. Bolz M, Ritter M, Schneider M, Simader C, Scholda diabetic macular edema. Retina. 2010;30(5):77480.
C, Schmidt-Erfurth U. A systematic correlation of 40. Murakami T, Nishijima K, Akagi T, Uji A, Horii T,
angiography and high-resolution optical coherence Ueda-Arakawa N, Muraoka Y, Yoshimura N. Optical
tomography in diabetic macular edema. coherence tomographic reflectivity of photoreceptors
Ophthalmology. 2009;116(1):6672. beneath cystoid spaces in diabetic macular edema.
26. Pece A, Isola V, Holz F, Milani P, Brancato Invest Ophthalmol Vis Sci. 2012;53(3):150611.
R. Autofluorescence imaging of cystoid macular 41. Shin HJ, Lee SH, Chung H, Kim HC. Association
edema in diabetic retinopathy. Ophthalmologica. between photoreceptor integrity and visual outcome
2010;224(4):2305. in diabetic macular edema. Graefes Arch Clin Exp
27. Antcliff RJ, Marshall J. The pathogenesis of edema in Ophthalmol. 2012;250(1):6170.
diabetic maculopathy. Semin Ophthalmol. 42. Murakami T, Nishijima K, Sakamoto A, Ota M, Horii
1999;14(4):22332. T, Yoshimura N. Association of pathomorphology,
28. Yanoff M, Fine BS, Brucker AJ, Eagle Jr photoreceptor status, and retinal thickness with visual
RC. Pathology of human cystoid macular edema. acuity in diabetic retinopathy. Am J Ophthalmol.
Surv Ophthalmol. 1984;28:50511. 2011;151(2):3107.
29. Tso MO. Pathology of cystoid macular edema. 43. Rothova A, Suttorp-van Schulten MS, Frits Treffers
Ophthalmology. 1982;89(8):90215. W, Kijlstra A. Causes and frequency of blindness in
30. Fine BS, Brucker AJ. Macular edema and cystoid mac- patients with intraocular inflammatory disease. Br J
ular edema. Am J Ophthalmol. 1981;92(4):46681. Ophthalmol. 1996;80(4):3326.
31. Otani T, Kishi S, Maruyama Y. Patterns of diabetic 44. Lardenoye CW, van Kooij B, Rothova A. Impact of
macular edema with optical coherence tomography. macular edema on visual acuity in uveitis.
Am J Ophthalmol. 1999;127(6):68893. Ophthalmology. 2006;113(8):14469.
32. Yamamoto S, Yamamoto T, Hayashi M, Takeuchi 45. Bouchenaki N, Cimino L, Auer C, Tao Tran V,
S. Morphological and functional analyses of diabetic Herbort CP. Assessment and classification of choroi-
macular edema by optical coherence tomography and dal vasculitis in posterior uveitis using indocyanine
multifocal electroretinograms. Graefes Arch Clin Exp green angiography. Klin Monbl Augenheilkd.
Ophthalmol. 2001;239(2):96101. 2002;219(4):2439.
33. Panozzo G, Parolini B, Gusson E, Mercanti A, 46. Cimino L, Auer C, Herbort CP. Sensitivity of indocy-
Pinackatt S, Bertoldo G, Pignatto S. Diabetic macular anine green angiography for the follow-up of active
edema: an OCT based classification. Semin inflammatory choriocapillaropathies. Ocul Immunol
Ophthalmol. 2004;19:1320. Inflamm. 2000;8(4):27583.
34. Kim BY, Smith SD, Kaiser PK. Optical coherence 47. Giovannini A, Ripa E, Scassellati-Sforzolini B,
tomographic patterns of diabetic macular edema. Am Ciardella A, Tom D, Yannuzzi L. Indocyanine green
J Ophthalmol. 2006;142(3):40512. angiography in serpiginous choroidopathy. Eur J
35. Sakamoto A, Nishijima K, Kita M, Oh H, Tsujikawa Ophthalmol. 1996;6(3):299306.
A, Yoshimura N. Association between foveal photore- 48. Giovannini A, Mariotti C, Ripa E, Scassellati-
ceptor status and visual acuity after resolution of dia- Sforzolini B. Indocyanine green angiographic find-
betic macular edema by pars plana vitrectomy. ings in serpiginous choroidopathy. Br J Ophthalmol.
Graefes Arch Clin Exp Ophthalmol. 1996;80(6):53640.
2009;247(10):132530. 49. Roesel M, Henschel A, Heinz C, Dietzel M, Spital G,
36. Alasil T, Keane PA, Updike JF, Dustin L, Ouyang Y, Heiligenhaus A. Fundus autofluorescence and spec-
Walsh AC, Sadda SR. Relationship between optical tral domain optical coherence tomography in uveitic
coherence tomography retinal parameters and visual macular edema. Graefes Arch Clin Exp Ophthalmol.
acuity in diabetic macular edema. Ophthalmology. 2009;247(12):16859.
2010;117(12):237986. 50. Haen SP, Spaide RF. Fundus autofluorescence in mul-
37. Forooghian F, Stetson PF, Meyer SA, Chew EY, Wong tifocal choroiditis and panuveitis. Am J Ophthalmol.
WT, Cukras C, Meyerle CB, Ferris 3rd 2008;145(5):84753.
FL. Relationship between photoreceptor outer seg- 51. Markomichelakis NN, Halkiadakis I, Pantelia E,
ment length and visual acuity in diabetic macular Peponis V, Patelis A, Theodossiadis P, Theodossiadis
edema. Retina. 2010;30(1):6370. G. Patterns of macular edema in patients with uveitis:
38. Maheshwary AS, Oster SF, Yuson RM, Cheng L, qualitative and quantitative assessment using optical
Mojana F, Freeman WR. The association between coherence tomography. Ophthalmology.
percent disruption of the photoreceptor inner segment- 2004;111(5):94653.
outer segment junction and visual acuity in diabetic 52. Scholl S, Augustin A, Loewenstein A, Rizzo S,
macular edema. Am J Ophthalmol. 2010; Kupperman B. General pathophysiology of macular
150(1):637. edema. Eur J Ophthalmol. 2011;21 Suppl 6:S109.
4 Imaging Before, During, and After Steroid Therapy 53

53. Loewenstein A, Zur D. Postsurgical cystoid macular 56. Lavin MJ, Marsh RJ, Peart S, Rehman A. Retinal arte-
edema. Dev Ophthalmol. 2010;47:14859. rial macroaneurysms: a retrospective study of 40
54. Sahin M, Cing AK, Gzm N. Evaluation of cystoid patients. Br J Ophthalmol. 1987;71(11):8172.
macular edema using optical coherence tomography 57. Panton RW, Goldberg MF, Farber MD. Retinal arte-
and fundus autofluorescence after uncomplicated rial macroaneurysms: risk factors and natural history.
phacoemulsification surgery. J Ophthalmol. 2013; Br J Ophthalmol. 1990;74(10):595600.
2013:376013. 58. Goldenberg D, Soiberman U, Loewenstein A,
55. Robertson DM. Macroaneurysms of the retinal arter- Goldstein M. Heidelberg spectral-domain optical
ies. Trans Am Acad Ophthalmol Otolaryngol. coherence tomographic findings in retinal artery mac-
1973;77(1):OP5567. roaneurysm. Retina. 2012;32(5):9905.
Treatment of DME with Steroids
5
Couturier Aude and Pascale Massin

Contents 5.1 Introduction


5.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . 55
5.1.1 Epidemiology. . . . . . . . . . . . . . . . . . . . . . . . 55 5.1.1 Epidemiology
5.1.2 Systemic Factors . . . . . . . . . . . . . . . . . . . . . 56
5.1.3 Laser Photocoagulation . . . . . . . . . . . . . . . . 56 Diabetic macular edema (DME) is the main
5.1.4 Intravitreal Therapy . . . . . . . . . . . . . . . . . . . 56
cause of mild to moderate vision loss in patients
5.2 Pathophysiology of DME. . . . . . . . . . . . . . 56 with diabetes. It is a major public health prob-
5.3 Efficacity of Intravitreal Steroids lem as the prevalence of diabetes is increasing.
in DME . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57 In western countries, the expected prevalence
5.3.1 Triamcinolone . . . . . . . . . . . . . . . . . . . . . . . 58
of diabetic retinopathy is close to 35 % in the
5.3.2 Dexamethasone . . . . . . . . . . . . . . . . . . . . . . 59
5.3.3 Fluocinolone Acetonide . . . . . . . . . . . . . . . . 61 diabetic population, while DME may be present
in 710 % of patients [44]. After 2 years, over
5.4 Comparison of Intravitreal Steroids
for the Treatment of DME. . . . . . . . . . . . . 63 half of patients with DME will lose two or more
lines of visual acuity (VA) [18]. Worldwide, it
Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65
has been estimated that 21 million people have
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65 DME [44].
Two clinical subtypes of DME have been
described. Focal DME is defined by localized
retinal thickening, often surrounded by exu-
date rings, resulting from leakage from micro-
aneurysms, and/or intraretinal microvascular
abnormalities. The prognosis of focal DME is
generally good, as it responds well to focal laser
photocoagulation of leaking microaneurysms
[16]. Diffuse DME is characterized by general-
ized thickening of the central macula caused by
widespread leakage from dilated capillaries in
this area. The effect of laser treatment on diffuse
DME is limited. Finally, ischemic maculopathy
C. Aude, MD (*) P. Massin, MD, PhD (*) is secondary to extended occlusion of macu-
Department of Ophthalmology, Lariboisiere,
lar capillaries. Frequently, there is combined
2 rue Ambroise Pare, 75003 Paris, France
e-mail: audecout@lrb.aphp.fr; pascale.massin@lrb.aphp.fr; pathology of focal and diffuse edema as well
pascale.massin1@gmail.com as ischemia. However, there is no international

A.J. Augustin (ed.), Intravitreal Steroids, 55


DOI 10.1007/978-3-319-14487-0_5, Springer International Publishing Switzerland 2015
56 C. Aude and P. Massin

consensus on the definition of focal and diffuse in BCVA. Approximately half of the eyes treated
macular edema, and this classification tends to with laser alone continued to have thickened reti-
be abandoned [7]. nas at year 2.
Grid laser photocoagulation has been shown
to be effective to treat DME, but with a slow
5.1.2 Systemic Factors effect and retinal scars [15], and new medical
therapeutic approaches have been developed
Risk factors for DME include duration of dia- including intravitreous injections of corticoste-
betes, poor glycemic control, hypertension, pro- roids or anti-vascular endothelial growth factors
teinuria, and hypercholesterolemia. Treatment (anti-VEGF) agents. In addition, in these eyes
of DME therefore begins with controlling these with DME, more than 15 % of patients go on los-
factors. It is now widely accepted that control ing vision despite previous laser photocoagula-
of systemic factors, which may worsen DME, tion [22].
is essential. The United Kingdom Prospective
Diabetes Study (UKPDS) trial showed that an
11 % reduction in hemoglobin A1c, from 7.9 to 5.1.4 Intravitreal Therapy
7.0 %, in type 2 diabetics reduced the risk of
microvascular complications of diabetes, includ- Intravitreal anti-VEGF agents have revolution-
ing the need for photocoagulation, by 25 % [39]. ized the treatment of DME by demonstrating
Similarly, tight blood pressure control reduced visual improvement in multiple randomized clin-
the risk of microvascular complications by 37 % ical trials [34]. However, about 23 % of patients
[40]. are nonresponders to anti-VEGF therapy [6], and
their use may be compromised because of the dif-
ficulties for a monthly follow-up.
5.1.3 Laser Photocoagulation Intravitreal steroids are used in the treatment
of DME for their anti-inflammatory, angiostatic,
Treatments for DME are evolving rapidly, and and antipermeability effects. While steroids
while it was once treated solely with laser ther- are broad-spectrum anti-inflammatory medica-
apy, today there are numerous alternative thera- tions, the anti-VEGF agents specifically target
pies that have demonstrated promise. Over the a molecule implicated in the pathogenesis of
past 30 years, the first-line therapy has been laser DME. The use of corticosteroid injections for
photocoagulation. Direct photocoagulation of treatment of DME has been widely evaluated,
leaking microaneurysms combined with grid due to their ability to inhibit pro-inflammatory
laser photocoagulation reduces the risk of moder- cells, pro-angiogenic cytokines such as IL6,
ate vision loss by 50 % in patients with DME, as and VEGF expression [33]. These studies have
reported in the large 3-year Early Treatment shown efficacy of steroids in controlling DME
Diabetic Retinopathy Study (ETDRS) clinical and potentially in improving vision [23, 24]
trial, but it often produces little or no improve- (Fig. 5.1).
ment in VA [15]. This result has been reinforced
in more recent trials by the Diabetic Retinopathy
Clinical Research Network [12]. DRCR.net study 5.2 Pathophysiology of DME
showed that among eyes with DME treated at
4-month intervals with laser as needed, 32 % DME is defined by retinal thickening involving
gained at least ten letters in BCVA from baseline or threatening the center of the macula, consecu-
to year 2, but 49 % had less than a ten-letter tive to the intraretinal accumulation of fluid in the
change in BCVA, and 19 % lost at least ten letters macular area. Although the pathogenesis of DME
5 Treatment of DME with Steroids 57

Fig. 5.1 Left eye of a 52-year-old woman with type 2 diabetes. OCT analyses show a diffuse macular edema with an
epiretinal membrane and a mild foveal serous detachment. The foveal thickness was measured at 606 m

is still not fully understood, it is mainly caused by diabetes [37]. Because activated microglia
the breakdown of the inner bloodretinal barrier, release pro-inflammatory cytokines and chemo-
but capillary non-perfusion and possibly direct kines, such as VEGF and tumor necrosis factor,
neuronal damage are also major actors. The it is likely that they further exacerbate retinal
pathophysiology of DME is complex and multi- vascular permeability in diabetes.
factorial, and inflammation has an important role Corticosteroids may be useful in the treatment
in the pathogenesis of DME. Chronic hypergly- of DME because they block leukostasis; inhibit the
cemia results in the formation of advanced glyca- expression of prostaglandins, pro-inflammatory
tion end products [38], oxidative stress [43], and cytokines, and VEGF [41]; and enhance the bar-
the activation of protein kinase C. These changes rier function of vascular tight junctions [17].
lead to the recruitment and adhesion of leuko-
cytes to the retinal vascular endothelium (leu-
kostasis) [32] and the expression of inflammatory 5.3 Efcacy of Intravitreal
factors such as intercellular adhesion molecule-1, Steroids in DME
vascular endothelial growth factor (VEGF),
tumor necrosis factor-, and interleukin-6 [19]. Treatment for DME includes treatment of the sys-
The upregulation of these factors causes endo- temic disease by maintaining tight blood sugar
thelial damage, hypoxia [27], alterations in and blood pressure control, weight loss, and the
endothelial tight junction proteins, and increased lowering of circulating triglycerides and choles-
vessel permeability. As a result, the integrity of terol. However, once there is progression to DME,
the bloodretinal barrier becomes permeable to therapy is indicated to slow the rate of vision loss
fluid, leading to vascular leakage, accumulation and to attempt to improve the long-term progno-
of fluid in the extravascular space of the retina, sis. The addition of intravitreal corticosteroid
and thickening of the macula [3]. Another aspect therapy to laser photocoagulation may help to
of chronic inflammation is related to microglial reduce further the risk of clinically significant
cells, which are chronically activated during vision loss [45].
58 C. Aude and P. Massin

5.3.1 Triamcinolone focal/grid photocoagulation (N = 330), 1-mg


IVTA (N = 256), or 4-mg IVTA (N = 254), with
Triamcinolone has been shown to reduce VEGF the option to retreat for persistent edema every
production, decrease prostaglandin production, 4 months. After 4 months, the 4-mg IVTA group
and stabilize the bloodretinal barrier [42]. This had better VA than both the laser group (P < 0.001)
action of decreasing vascular permeability has and the 1-mg IVTA group (P < 0.001). However,
led investigators to try triamcinolone in the treat- the benefit diminished thereafter, and at 1 year
ment of diffuse DME [29]. there was no significant difference between the
A few small, randomized trials have reported three groups. At the primary end point of 2 years,
improvement in VA in eyes with recalcitrant dif- the laser group had a small but significant
fuse DME treated with intravitreal triamcinolone improvement in best-corrected VA compared to
(IVTA) [1, 23, 26, 28, 30]. Yet, as a result of drug the IVTA groups (P = 0.02 comparing 1-mg
absorption, these effects are not sustained and IVTA and P = 0.002 comparing 4-mg IVTA) [12].
there is a rather risk of recurrences of DME [26]. Change in VA from baseline to 3 years was + five
In a study of 69 eyes randomized to receive letters in the laser group and 0 in each IVTA
either 4-mg IVTA injections or placebo, Gillies group [13]. It has been suggested that the acute
et al. reported an improvement of five or more anti-inflammatory action of the steroid is effec-
letters in 56 % of eyes treated with IVTA com- tive in the short term, but due to the gradual
pared with 26 % eyes in the sham group decline in concentration, it is not beneficial as a
(P = 0.006), after 2 years [23]. OCT analysis long-term therapy. Similarly to other studies, the
demonstrated a significant decrease in central eyes treated with 4-mg IVTA had significantly
macular thickness in eyes treated with IVTA higher rates of increased intraocular pressure
compared to controls. IOP-lowering medication (IOP) (33 %) and need antiglaucoma medication
was necessary in 44 % of treated eyes, and IOP (30 %) compared with the laser treatment group
elevation required surgery in two eyes. Cataract in the DRCR.net study (DRCR.net [12, 13]). The
surgery was performed in 54 % of treated eyes. cumulative probability of cataract surgery by
When compared with laser therapy however, 3 years was 31, 46, and 83 % in the laser and
the results have been inconclusive. The Diabetic 1-mg IVTA and 4-mg IVTA groups, respectively
Retinopathy Clinical Research Network (DRCR. [13]. When compared to laser therapy, these
net) conducted a trial in 840 eyes to evaluate results indicate the absence of long-term benefit
IVTA compared with focal/grid photocoagula- of IVTA and an increased risk of side effects
tion [12]. Eyes were randomized to receive either (Fig. 5.2).

66

64
Mean visual acuity (letter score)

62

60

58

56 Laser

54 1 mg IVTA

Fig. 5.2 Mean visual acuity 4 mg IVTA


at each visit according to 52
treatment group Baseline 4 months 12 months 24 months 36 months
5 Treatment of DME with Steroids 59

More recently, the DRCR.net study group con- Drug Administration (FDA) for the treatment of
ducted a comparative clinical trial in 854 eyes to macular edema due to retinal vein occlusion and
evaluate four different treatments, including intra- noninfectious posterior segment uveitis.
vitreal 0.5-mg ranibizumab combined with Two phase II trials studied the effects of the
prompt or deferred laser photocoagulation or DEX-DDS (Ozurdex; Allergan Inc, Irvine,
4-mg triamcinolone combined with focal/grid California) compared with observation, in eyes
laser, compared with sham injections combined with DME treated with laser [8, 21].
with focal/grid laser for center-involved DME In a phase II study [21], 315 eyes with persis-
[14]. The study found that intravitreal ranibi- tent macular edema, including 171 DME, were
zumab with prompt or deferred laser was more randomized to receive either 350-g DEX-DDS,
effective through at least 1 year than triamcino- 700-g DEX-DDS, or observation. After 90 days,
lone combined with laser photocoagulation or an improvement of ten letters or more was
laser alone for the treatment of DME involving observed in significantly more DME eyes treated
the central macula, although uncommonly associ- with 700-g DEX-DDS (33.3 %) and 350-g
ated with endophthalmitis. In pseudophakic eyes, DEX-DDS (21.1 %) than with observation
intravitreal triamcinolone plus prompt laser was (12.3 %; P = 0.007 vs. 700-g DEX-DDS) [20].
as effective as ranibizumab combined with prompt At day 180, a BCVA improvement of ten letters
or deferred laser photocoagulation and was more or more was seen in 30 % of eyes in the 700-g
effective than laser alone, but the triamcinolone DEX-DDS group, 19 % in the 350-g DEX-DDS
plus prompt laser arm had an increased risk of group, and 23 % in the observation group (P 0.4
intraocular pressure elevation [14]. for treated vs. observed eyes). There were also
So, several studies have shown the efficacy of significantly greater improvements in retinal
IVTA to temporarily reduce DME and increase thickness and fluorescein leakage in treated eyes
visual acuity [1, 26, 29]. The mean reduction in than observed eyes [20]. In both treatment
macular thickening reaches 85 % 3 months after groups, 15 % of eyes had an increase in IOP
injection with a mean two-line improvement of greater than 10 mmHg from baseline at any time
visual acuity [1, 23, 29]. However, recurrence of during follow-up, though the authors pointed out
DME occurs between 3 and 6 months after that these elevations generally only occurred
injection. Due to the high rate of complications once during follow-up, and only 2 % of patients
and the absence of evidence of its superiority had sustained IOP elevations at 90 days. No
over laser photocoagulation, IVTA is generally patients required IOP-lowering surgery (Fig. 5.3).
reserved for patients refractory to focal/grid More recently, Callanan et al. randomized 253
laser therapy and anti-VEGF agents [46]. eyes with DME either to treatment with DEX-
Despite the adverse events associated with ste- DDS at baseline plus laser at month 1 or sham
roids, a recent Cochrane review supported the implant at baseline plus laser at month 1 [8].
use of intravitreal steroids in the treatment of They could receive up to three additional laser
DME refractory to laser therapy [25]. IVTA has treatments and one additional DEX implant as
the advantage over laser therapy that it can be needed. The percentage of patients who gained
repeated several times, as long as IOP rise is ten letters or more at month 12 did not differ
controlled. between treatment groups (27.8 % in the combi-
nation treatment group and 23.6 % in the laser
alone group), but the percentage of patients was
5.3.2 Dexamethasone significantly greater in the combination group at
month 1 (P < 0.001) and month 9 (P = 0.007). In
The dexamethasone 700 g posterior segment patients with angiographically verified diffuse
drug delivery system (Ozurdex) is a bioerodible DME, the mean improvement in BCVA was sig-
implant delivered intravitreally using a 22-gauge nificantly greater with DEX-DDS plus laser
injector. It has been approved by the US Food and treatment than with laser treatment alone (up to
60 C. Aude and P. Massin

Fig. 5.3 Percentage of patients who P = 0.01 P = 0.4


35
achieved 10 letters or more or 15
letters or more of improvement in
best-corrected visual acuity. P values 30
represent the comparison between the
700-g group and the observation 25
group. DDS indicates drug delivery

%age patients
system 20

15

10

0
Day 90 Day 180

Observation group 700 g group2


350 g group

7.9 vs. 2.3 letters) at all time points through edema were recently published [5]. This study
month 9 (P 0.013). At month 4, patients with consists of two randomized, multicenter, sham-
verified diffuse edema who were given DEX- controlled, 3-year, phase III clinical trials con-
DDS before laser treatment were approximately ducted in 22 countries. Because the trials were
twice as likely to gain at least ten letters in BCVA identical in study design, the results were pooled
compared with patients who were given the sham for analysis. A total of 1,048 patients were ran-
injection (23.2 % vs. 11.9 %; P = 0.035). Decrease domized in a 1:1:1 ratio to study treatment with
in the area of diffuse vascular leakage measured DEX implant 0.7 mg, DEX implant 0.35 mg, or
angiographically was significantly larger with sham procedure and followed for 3 years.
DEX-DDS plus laser treatment through month Retreatment was not allowed more often than
12 (P 0.041). Increased IOP was more common every 6 months. The predefined primary efficacy
with combination treatment (16.8 % of eyes). No end point for the US Food and Drug
surgery for elevated IOP was required. Cataract Administration was achievement of 15-letter
events were more common in phakic study eyes improvement in BCVA from baseline at
in the DEX-DDS plus laser group (22.2 %) than study end.
in the laser-alone group (9.5 %; P = 0.017). These The percentage of patients with a 15-letter
results suggest that a retreatment interval shorter improvement from baseline at the year 3 was
than 6 months may be needed to provide a better greater with DEX implant 0.7 mg (22 %) and
sustained benefit. DEX implant 0.35 mg (18.4 %) than with sham
DEX-DDS has also demonstrated efficacy for (12 %, P 0.018). The mean (standard devia-
the treatment of DME in vitrectomized eyes [4]. tion) average change in BCVA from baseline
Fifty-five vitrectomized eyes with diffuse DME during the study was 3.5 (8.4) letters with DEX
were retrospectively studied. After 8 weeks, implant 0.7 mg, 3.6 (8.1) letters with DEX
30.4 % of eyes had an improvement in VA of ten implant 0.35 mg, and 2.0 (8.0) letters with sham.
letters or more. During the follow-up, 16 % Mean change in BCVA from baseline at each
developed IOP elevation. study visit was significantly greater in both DEX
The results of the phase III study evaluating implant groups compared with sham at most
the efficacy and safety of dexamethasone intra- time points during the first 15 months. However,
vitreal implant in patients with diabetic macular the improvement in BCVA provided by DEX
5 Treatment of DME with Steroids 61

implants relative to sham was reduced after interval for DEX-DDS in the treatment of
month 15, due to the development of cataract. In DME. A study directly comparing DEX-DDS
pseudophakic eyes, mean improvement in BCVA and ranibizumab in the treatment of DME is
provided by DEX implant relative to sham was underway. Recently, the European Medicines
consistent across time in the 3-year study, and Agency adopted a positive opinion on the use of
there was no reduction in treatment benefit DEX-DDS in DME patients, who are pseudopha-
observed in year 2. Mean number of treatments kic or considered insufficiently responsive to or
received over 3 years was 4.1, 4.4, and 3.3 with unsuitable for non-corticosteroid therapy.
DEX implant 0.7 mg, DEX implant 0.35 mg, and
sham, respectively. Mean average reduction in
central retinal thickness from baseline was sig- 5.3.3 Fluocinolone Acetonide
nificantly greater with DEX implant 0.7 and
0.35 mg than sham. Rates of cataract-related Two fluocinolone acetonide (FAc) intravitreal
adverse events in phakic eyes were 67.9, 64.1, devices have been studied for the treatment of
and 20.4 % in the DEX implant 0.7 mg, DEX DME: a FAc implant (Retisert; Bausch and
implant 0.35 mg, and sham groups, respectively. Lomb Inc, Rochester, NY, USA) and a FAc insert
Approximately one third of patients in each (ILUVIEN; Alimera Sciences, Inc, Alpharetta,
DEX implant group had a clinically significant GA, USA). The results of the studies examining
increase in IOP requiring treatment during the the use of FAc in the treatment of DME have
study, and only one patient (0.3 %) in each DEX been reviewed by Messenger et al. [31].
implant treatment group underwent glaucoma
incisional surgery for steroid-induced increases 5.3.3.1 FAc Implant
in IOP. Increase in IOP of 10 mmHg from Retisert contains 0.59-mg FAc and has an initial
baseline was observed in 27.7, 24.8, and 3.7 % release of approximately 0.6 g/day in the vitre-
of eyes in the DEX implant 0.7 mg, DEX implant ous. It is surgically implanted into the posterior
0.35 mg, and sham groups, respectively. In con- segment of the eye through a pars plana incision.
clusion, the DEX implant demonstrated efficacy The FAc implant received initial FDA approval
in the treatment of DME, meeting the primary for the treatment of chronic, posterior uveitis;
efficacy end points for improvement of however, more recently investigators have exam-
BCVA. The safety profile was favorable, being ined its utility in the treatment of DME [35, 36].
consistent with previous reports. In 2006 and 2011, Pearson et al. published the
In all studies, DEX-DDS treatment was well results of two trials with 197 and 196 eyes with
tolerated and had an acceptable safety profile. refractory DME randomized in a 2:1 ratio, to
Similar to IVTA, DEX-DDS has the advantage of receive either 0.59-mg FAc implant or the stan-
repeatability, as long as the IOP and cataract dard of care (SOC) (focal/grid laser photocoagu-
effects are mitigated. The advantages of DEX- lation or observation at the investigators
DDS over intravitreal triamcinolone include a discretion). Inclusion criteria were a history of
possible longer duration of action, the absence of persistent or recurrent DME with retinal thicken-
postinjection visual clouding or floater-like ing involving fixation of 1 disk area in size and
symptoms, and a lower rate of IOP increase. In an ETDRS VA of 20 letters (20/400) to 68 let-
all cases, the elevations in IOP were managed ters (20/50).
with IOP-lowering medication or observation, In the 2011 study, a 15-letter improvement
and no patient discontinued the studies because in VA was achieved in 16.8 % of implanted eyes
of IOP or had an IOP of 25 mmHg or more at at 6 months (P = 0.0012), 16.4 % at 1 year
month 12. (P = 0.1191; SOC group, 8.1 %), 31.8 % at
These studies have demonstrated the efficacy 2 years (P = 0.0016), and 31.1 % at 3 years
of DEX implant for DME. Further studies are (P = 0.1566; SOC group, 20.0 %). Moreover, the
warranted to address the optimal retreatment number of eyes with resolution of macular retinal
62 C. Aude and P. Massin

thickening was higher in the implanted eyes than and 27.8 % (high dose) of the FAc insert groups
in the SOC group at 6 months (P < 0.0001), 1 year compared with 18.9 % in the sham group
(P < 0.0001; 72 % vs. 22 %), 2 years (P = 0.016), (P = 0.018).
and 3 years (P = 0.861); and a lower rate of Interestingly, preplanned subgroup analysis
decline in diabetic retinopathy severity score demonstrated a doubling of benefit compared
occurred in the implanted group (P = 0.0207 at with sham injections in patients who reported
3 years). duration of DME 3 years at baseline. In a sub-
However, this FAc implant has side effects, analysis of patients with DME for 3 years prior
including a high rate of steroid-induced ocular to enrollment, patients in both the low-dose
comorbidities. Compared with the SOC, patients (P < 0.001) and high-dose groups (P = 0.002)
treated with the FAc implant had significantly were more likely to achieve a 15-letter gain
higher rates of cataract extraction (91 % of compared with patients in the sham group. Foveal
implanted phakic eyes compared with 20 % in thickness was significantly reduced in both low-
the SOC group). An IOP above 30 mmHg was dose and high-dose groups compared with the
recorded in 61.4 % of implanted eyes (vs. 5.8 % sham group until month 30.
in the SOC group) at any time, and 33.8 % The percentage of patients that needed two
required surgery to control elevated IOP by FAc insert injections at month 36 were 18.7 and
4 years [35]. While potentially effective for 24.2 in the low- and high-dose insert groups,
refractory DME, this implant has a substantial respectively; and 5.3 and 7.0 % respectively
risk of glaucoma and cataract progression. needed 3 study treatments at month 36.
Adverse effects were common in both low-
5.3.3.2 FAc Insert dose and high-dose insert groups. The most com-
ILUVIEN is a non-bioerodible insert implanted mon adverse event was cataract, and almost all
in the eye via injection through the pars plana phakic patients in the FAc insert groups devel-
using a 25-gauge needle. ILUVIEN contains oped cataract (81.7 and 88.7 % in the low- and
0.19 mg of FAc. It is indicated in Europe for the high-dose insert groups respectively compared
treatment of vision impairment associated with with 50.7 % in the sham group), but their visual
chronic diabetic macular edema considered benefit after cataract surgery was similar to that
insufficiently responsive to available therapies. in pseudophakic patients. The incidence of
There are two clinical trials that studied the increased IOP (37.1 and 45.5 %, in the low-dose
effects of the FAc insert on DME [911]. The and high-dose insert groups, respectively), need
Fluocinolone Acetonide for Macular Edema for IOP-lowering medication (38.4 and 47.3 %,
(FAME) studies were two large prospective, ran- respectively), incisional glaucoma surgery (4.8
domized, controlled studies that followed 956 and 8.1 %, respectively), and trabeculoplasty (1.3
eyes randomized to receive 0.2-g/day (low- and 2.5 %, respectively) was higher in the FAc
dose) or 0.50-g/day (high-dose) inserts or insert groups compared with the sham group.
sham. Subjects with persistent DME despite at The primary outcome, which was percentage
least one macular laser treatment and BCVA 19 of patients with improvement of 15 letters from
and 68 letters on ETDRS chart were included. baseline at months 24 and 36, was met in both
Based on retreatment criteria, additional study FAME trials. These results show that low-dose
drug or sham injections could be given after FAc insert provides substantial benefit to patients
1 year. with refractory DME for 3 years. The maximum
A BCVA letter score 15 letters was achieved benefit occurs at month 30, with 75 % of patients
at month 24 in 28.7 and 28.6 % in the low- and requiring only one insert to obtain this benefit
high-dose FAc insert groups respectively com- (Fig. 5.4).
pared with 16.2 % in the sham group (P = 0.002 Messenger et al. reviewed and compared the
for each) and after 3 years in 28.7 % (low dose) results of the studies of FAc devices in the treat-
5 Treatment of DME with Steroids 63

Eyes with 15 letter gain in BCVA (%) 40 pared with only 45.5 % of high-dose FAc-inserted
35 eyes and 37.1 % of low-dose FAc-inserted eyes;
finally, 33.8 % of FAc-implanted eyes required
30 incisional surgery to control increased IOP com-
25 pared with only 4.8 and 8 % of low- and high-
dose FAc-inserted eyes, respectively [31].
20
Although the improvement in BCVA may be
15 similar, the significantly lower rate of ocular
hypertension in patients treated with FAc inserts
10
makes it the preferred FAc device to treat
5 DME. In Europe, FAc insert (ILUVIEN;
Alimera Sciences, Inc, Alpharetta, GA, USA)
0
3 6 12 18 24 30 36 recently obtained authorities approval for the
Months follow-up
treatment of chronic refractory DME in patients
with absence of response to other approved
FAc implant 0.5 g/day FAc therapies.
insert
0.2 g/day FAc
insert
5.4 Comparison of Intravitreal
Fig. 5.4 Vision outcomes through month 36 in patients
with diabetic macular edema treated with Fluocinolone
Steroids for the Treatment
Acetonide (FAc): Percentage of eyes achieving 15 letter of DME
gain in best-corrected visual acuity during follow-up
between FAc devices [31] Comparing results from trials studying different
molecules is problematic because of the differ-
ences in follow-up time, patient populations, and
study design. However, large variations in rates
ment of DME [31]. Comparing the studies of may elucidate important differences in the effi-
FAc devices head-to-head is complicated cacy and rates of adverse events between fluo-
because of differences in their inclusion/exclu- cinolone, dexamethasone, and triamcinolone,
sion criteria, retreatment protocols, and follow- and Messenger et al. reviewed and compared the
up time; both FAc devices appear to have similar results of the main steroid studies for DME [31]
efficacy to treat DME. In both studies, 15 % or (Table 5.1).
more of eyes achieved a 15-letter increase in When comparing effects on VA, triamcinolone
BCVA. By 18 months, 20 % eyes treated with appears less effective than both fluocinolone and
the FAc implant or either doses of the FAc insert dexamethasone devices, with fewer patients achiev-
maintained a 15-letter increase in BCVA. By ing a BCVA improvement of 15 letters. The per-
3 years, 30 % of eyes treated with a FAc device centage of eyes achieving 15-letter improvement
achieved a 15-letter increase in BCVA with this DEX-DDS is comparable with that seen in
(Fig. 5.5). the FAc device studies and is greater than that seen
The rate of cataract progression was similar in the triamcinolone studies at similar follow-up
with the two FAc devices, with more than 80 % times [13, 21, 20, 35, 10]. Overall, these data show
of phakic eyes requiring cataract surgery after greater efficacy of the FAc and dexamethasone
4 years of treatment. However, the risk of devices compared with triamcinolone injections
increased IOP and glaucoma progression was that is unlikely attributable simply to variations in
significantly higher in the FA implant trial com- follow-up time or study design. IVTA may only
pared with the FAc insert. An increased IOP have a temporary effect on macular thickening and
occurred in 69.7 % of FAc-implanted eyes com- VA, without long-term benefit.
64 C. Aude and P. Massin

Fig. 5.5 Right eye of a 39-year old woman with diffuse tion, OCT showing a decrease of DME and improvement in
DME treated with FAc insert. (a) OCT showing diffuse cys- foveal thickening, VA was 59 letters and IOP 21 mmHg. (c,
toid DME, VA was 26 letters on ETDRS chart and IOP was d) Improvement maintened at 1 and 3 months after FAc
measured at 14 mmHg. (b) One week after FAc insert injec- insqert injection, VA was 61 letters and IOP 16 mmHg

The rate of serious adverse events was higher month 12 [8]. The follow-up period of the phase
in the FAc studies than in the triamcinolone and II dexamethasone study is short and still not
dexamethasone studies [10, 12, 20, 21, 35]. long enough to compare cataract progression
The need for cataract surgery may be higher rates [20]. However, in the phase III trial evalu-
in the FAc-treated eyes, with more than 85 % of ating DEX implant for DME, 5259 % of DEX
eyes requiring cataract surgery at 3 years com- implanted eyes required cataract surgery at
pared with 37 % of triamcinolone-injected eyes 3 years. Rates of IOP increase were higher in the
at 2 years. Cataract progression occurred in FAc-implanted study and lower in the dexa-
22 % of phakic eyes in the DEX-DDS group at methasone implant study. However, each study
5 Treatment of DME with Steroids 65

Table 5.1 Treatment of DME with steroids. Visual acuity, macular thickness, and adverse events among different
steroids for diabetic macular edema
Triamcinolone Dexamethasone FAc implant FAc insert
DRCR.net (2-year Healler et al. Pearson et al. Campochiaro et al.
follow-up) (6-month follow-up) (3-year follow-up) (3-year follow-up)
1 mg 4 mg 350 g 700 g 0.59 mg 0.2 g 0.5 g
(n = 256) (n = 254) (n = 103) (n = 105) (n = 127) (n = 375) (n = 393)
15 letter improvement in VA 15 % 16 % 15 % 18 % 31 % 33 % 32 %
15 letter loss in VA 21 % 21 % NR NR 17 % NR NR
Mean change in foveal 86 77 43 132 110 171 161
thickness (m)
Cataract surgery 23 % 51 % NR NR 91 % 80 % 87 %
Increased IOP 10 mmHg 16 % 33 % 15 % 15 % NR NR NR
from baseline
Need of IOP-lowering meds 12 % 30 % NR NR NR 38 % 47 %
Glaucoma surgery 0% 1% 0% 0% 33 % 6% 11 %
FAc fluocinolone acetonide, IOP intraocular pressure, NR not reported

reported different criteria of increased IOP, treatment of DME and was well tolerated with
making direct comparisons difficult. Of patients significant improvements in BCVA and retinal
with increased IOP, it appears that more patients thickness at 1 year [20, 21]. The phase III trial
treated with FAc device needed glaucoma sur- evaluating DEX-DDS for DME confirmed its
gery than did patients treated with triamcinolone efficacy and safety.
and dexamethasone. Visual improvement obtained with fluo-
Lastly, there was no difference in cinolone acetonide vitreous insert implants
endophthalmitis incidence, with this occurring in was counterbalanced by a high level of ocular
less than 1 % of eyes treated with steroids. hypertony and the need to perform IOP-
lowering procedure in a high percentage of
Conclusions cases at 3 years, limiting the use of this device
Many randomized clinical studies have shown to ancient, refractory DME [10, 35].
the clinical efficacy of steroid injections to
treat DME, but their use is limited by their
adverse events, including cataract progression
and intraocular pressure (IOP) increase. References
The Diabetic Retinopathy Clinical Research
Network study demonstrated the superiority 1. Audren F, Lecleire-Collet A, Erginay A, Haouchine
B, Benosman R, Bergmann JF, et al. Intravitreal tri-
of focal/grid photocoagulation, over a 3-year
amcinolone acetonide for diffuse diabetic macular
period, on intravitreal triamcinolone injections edema: phase 2 trial comparing 4 mg vs 2 mg. Am J
[12, 13]. However, in pseudophakic patients, Ophthalmol. 2006;142:7949.
the recent DRCR.net results showed a simi- 2. Audren F, Erginay A, Haouchine B, Benosman R,
Conrath J, Bergmann JF, et al. Intravitreal triamcino-
lar effect of triamcinolone plus laser com-
lone acetonide for diffuse diabetic macular oedema:
pared to ranibizumab associated plus prompt 6-month results of a prospective controlled trial. Acta
or deferred laser [14]. Still, IOP increase has Ophthalmol Scand. 2006;84:62430.
been observed in 40 % of cases and remains a 3. Bhagat N, Grigorian RA, Tutela A, Zarbin
MA. Diabetic macular edema: pathogenesis and treat-
current problem in clinical practice.
ment. Surv Ophthalmol. 2009;54(1):132.
Phase 2 clinical studies suggested that 4. Boyer DS, Faber D, Gupta S, Patel SS, Tabandeh
dexamethasone implant may be useful for H, Li XY, Liu CC, Lou J, Whitcup SM, Ozurdex
66 C. Aude and P. Massin

CHAMPLAIN Study Group. Dexamethasone intravit- plus prompt laser for diabetic macular edema.
real implant for treatment of diabetic macular edema Ophthalmology. 2010;117(6):106477. e35.
in vitrectomized patients. Retina. 2011;31(5):91523. 15. Early Treatment Diabetic Retinopathy Study Research
5. Boyer DS, Yoon YH, Belfort Jr R, Bandello F, Maturi Group. Photocoagulation for diabetic macular edema.
RK, Augustin AJ, Li XY, Cui H, Hashad Y, Whitcup Early treatment diabetic retinopathy study report
SM, Ozurdex MEAD Study Group. Three-year, ran- number 1. Arch Ophthalmol. 1985;103:1796806.
domized, sham-controlled trial of dexamethasone 16. Early Treatment Diabetic Retinopathy Study Research
intravitreal implant in patients with diabetic macular Group. Treatment techniques and clinical guidelines
edema. Ophthalmology. 2014;121(10):190414. for photocoagulation of diabetic macular edema.
Epub ahead of print. Early treatment diabetic retinopathy study report
6. Bressler SB, et al. Factors associated with changes in number 2. Ophthalmology. 1987;94(7):76174.
visual acuity and central subfield thickness at 1 year 17. Felinski EA, Antonetti DA. Glucocorticoid regulation
after treatment for diabetic macular edema with of endothelial cell tight junction gene expression:
ranibizumab. Arch Ophthalmol. 2012;130:115361. novel treatments for diabetic retinopathy. Curr Eye
7. Browning DJ, Altaweel MM, Bressler NM, Bressler Res. 2005;30(11):94957.
SB, Scott IU. Diabetic Retinopathy Clinical Research 18. Ferris FL, Patz A. Macular edema, a complication of
Network. Diabetic macular edema: what is focal and diabetic retinopathy. Surv Ophthalmol. 1984;28:
what is diffuse? Am J Ophthalmol. 2008;146(5): 45261.
64955. 19. Funatsu H, Noma H, Mimura T, Eguchi S, Hori
8. Callanan DG, Gupta S, Boyer DS, Ciulla TA, Singer S. Association of vitreous inflammatory factors with
MA, Kuppermann BD, Liu CC, Li XY, Hollander DA, diabetic macular edema. Ophthalmology. 2009;
Schiffman RM, Whitcup SM, Ozurdex PLACID Study 116(1):739.
Group. Dexamethasone intravitreal implant in combi- 20. Haller JA, Kuppermann BD, Blumenkranz MS,
nation with laser photocoagulation for the treatment of Dexamethasone DDS Phase II Study Group, et al.
diffuse diabetic macular edema. Ophthalmology. Randomized controlled trial of an intravitreous dexa-
2013;120(9):184351. methasone drug delivery system in patients with dia-
9. Campochiaro PA, Brown DM, Pearson A, FAME betic macular edema. Arch Ophthalmol.
Study Group, et al. Long-term benefit of sustained- 2010;128(3):28996.
delivery fluocinolone acetonide vitreous inserts for 21. Kuppermann BD, Blumenkranz MS, Haller JA,
diabetic macular edema. Ophthalmology. Dexamethasone DDS Phase II Study Group, et al.
2011;118(4):62635. Randomized controlled study of an intravitreous
10. Campochiaro PA, Brown DM, Pearson A, FAME dexamethasone drug delivery system in patients with
Study Group, et al. Sustained delivery fluocinolone persistent macular edema. Arch Ophthalmol.
acetonide vitreous inserts provide benefit for at least 3 2007;125(3):30917.
years in patients with diabetic macular edema. 22. Lee CM, Olk RJ. Modified grid laser photocoagula-
Ophthalmology. 2012;119(10):212532. tion for diffuse diabetic macular edema. Long-term
11. Campochiaro PA, Hafiz G, Shah SM, Famous Study visual results. Ophthalmology. 1991;98:1594602.
Group, et al. Sustained ocular delivery of fluocinolone 23. Gillies MC, et al. Intravitreal triamcinolone for refrac-
acetonide by an intravitreal insert. Ophthalmology. tory diabetic macular edema: two-year results of a
2010;117(7):13939. double masked, placebo-controlled, randomized clini-
12. Diabetic Retinopathy Clinical Research Network. A cal trial. Ophthalmology. 2006;113:15338.
randomized trial comparing intravitreal triamcinolone 24. Gillies MC, et al. Intravitreal triamcinolone prior to
acetonide and focal/grid photocoagulation for dia- laser treatment of diabetic macular edema: 24-month
betic macular edema. Ophthalmology. 2008;115(9): results of a randomized controlled trial. Ophthalmology.
14479. 2011;118:86672.
13. Diabetic Retinopathy Clinical Research Network 25. Grover D, Li TJ, Chong CC. Intravitreal steroids for
(DRCR.net), Beck RW, Edwards AR, Aiello LP, macular edema in diabetes [review]. Cochrane
Bressler NM, Ferris F, Glassman AR, Hartnett E, Ip Database Syst Rev. 2008;(1):CD005656.
MS, Kim JE, Kollman C. Three-year follow-up of a 26. Jonas JB, Kreissig I, Sfker A, Degenring
randomized trial comparing focal/grid photocoagula- RF. Intravitreal injection of triamcinolone for diffuse
tion and intravitreal triamcinolone for diabetic macu- diabetic macular edema. Arch Ophthalmol. 2003;
lar edema. Arch Ophthalmol. 2009;127(3):24551. 121(1):5761.
14. Diabetic Retinopathy Clinical Research Network, 27. Joussen AM, Murata T, Tsujikawa A, Kirchhof B,
Elman MJ, Aiello LP, Beck RW, Bressler NM, Bursell SE, Adamis AP. Leukocyte-mediated endo-
Bressler SB, Edwards AR, Ferris 3rd FL, Friedman thelial cell injury and death in the diabetic retina. Am
SM, Glassman AR, Miller KM, Scott IU, Stockdale J Pathol. 2001;158(1):14752.
CR, Sun JK. Randomized trial evaluating ranibi- 28. Lam DS, Chan CK, Mohamed S, et al. A prospective
zumab plus prompt or deferred laser or triamcinolone randomised trial of different doses of intravitreal
5 Treatment of DME with Steroids 67

triamcinolone for diabetic macular oedema. Br J co-localize with AGE receptors in the retinal vascula-
Ophthalmol. 2007;91(2):199203. ture of diabetic and of AGE-infused rats. Am J Pathol.
29. Martidis A, Duker JS, Greenberg PB, et al. Intravitreal 1997;150(2):52331.
triamcinolone for refractory diabetic macular edema. 39. United Kingdom Prospective Diabetes Study
Ophthalmology. 2002;109(5):9207. (UKPDS) Group. Tight blood pressure control and
30. Massin P, Audren F, Haouchine B, Erginay A, risk of macrovascular and microvascular complica-
Bergman JF, Benosman R, Caulin C, Gaudric tions in type 2 diabetes. BMJ. 1998;317:70313.
A. Intravitreal triamcinolone acetonide for diabetic 40. United Kingdom Prospective Diabetes Study
diffuse macular edema: preliminary results of a pro- (UKPDS) Group. Effect of intensive blood-glucose
spective controlled trial. Ophthalmology. 2004;111: control with metformin on complications in over-
21825. weight patients with type 2 diabetes. Lancet.
31. Messenger WB, Beardsley RM, Flaxel 1998;352:85465.
CJ. Fluocinolone acetonide intravitreal implant for 41. Wang K, Wang Y, Gao L, Li X, Li M, Guo
the treatment of diabetic macular edema. Drug Des J. Dexamethasone inhibits leukocyte accumulation
Devel Ther. 2013;7:42534. and vascular permeability in retina of streptozotocin-
32. Miyamoto K, Ogura Y. Pathogenetic potential of leu- induced diabetic rats via reducing vascular endothe-
kocytes in diabetic retinopathy. Semin Ophthalmol. lial growth factor and intercellular adhesion
1999;14(4):2339. molecule-1 expression. Biol Pharm Bull. 2008;
33. Nauck M, Karakiulakis G, Perruchoud AP, 31(8):15416.
Papakonstantinou E, Roth M. Corticosteroids inhibit 42. Wilson CA, Berkowitz BA, Sato Y, Ando N, Handa
the expression of the vascular endothelial growth fac- JT, De Juan Jr E. Treatment with intravitreal steroid
tor gene in human vascular smooth muscle cells. Eur reduces blood-retinal barrier breakdown due to retinal
J Pharmacol. 1998;341:30915. photocoagulation. Arch Ophthalmol.
34. Nguyen QD, et al. Ranibizumab for diabetic macular 1992;110(8):11559.
edema: results from 2 phase III randomized trials: 43. Witmer AN, Vrensen GF, Van Noorden CJ,
RISE and RIDE. Ophthalmology. 2012;119:789801. Schlingemann RO. Vascular endothelial growth fac-
35. Pearson PA, Comstock TL, Ip M, et al. Fluocinolone tors and angiogenesis in eye disease. Prog Retin Eye
acetonide intravitreal implant for diabetic macular Res. 2003;22(1):129.
edema: a 3-year multicenter, randomized, controlled 44. Yau JW, Rogers SL, Kawasaki R, Meta-Analysis for
clinical trial. Ophthalmology. 2011;118(8):15807. Eye Disease (META-EYE) Study Group, et al. Global
36. Pearson PA, Levy B, Comstock T, Fluocinolone prevalence and major risk factors of diabetic retinopa-
Acetonide Implant Study Group. Fluocinolone ace- thy. Diabetes Care. 2012;35:55664.
tonide intravitreal implant to treat diabetic macular 45. Ladas ID, Theodossiadis GP. Long-term effectiveness
edema: a 3year results of a multicenter clinical trial. of modified grid laser photocoagulation for diffuse
Invest Ophthalmol Vis Sci. 2006;47:E-Abstract 5442. diabetic macular edema. Acta Ophthalmol. 1993;
37. Rungger-Brandle E, Dosso AA, Leuenberger 71(3):3937.
PM. Glial reactivity, an early feature of diabetic 46. Martidis A, Duker JS, Greenberg PB, Rogers AH,
retinopathy. Invest Ophthalmol Vis Sci. 2000;41: Puliafito CA, Reichel E, Baumal C. Intravitreal triam-
197180. cinolone for refractory diabetic macular edema.
38. Stitt AW, Li YM, Gardiner TA, Bucala R, Archer DB, Ophthalmology. 2002;109(5):9207.
Vlassara H. Advanced glycation end products (AGEs)
Intravitreal Steroids
for the Treatment of Macular 6
Edema in Retinal Vein Occlusions

Eran Zunz and Anat Loewenstein

Contents 6.1 Introduction


6.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . 69
Retinal vein occlusion is considered a multifac-
6.2 Efficacy . . . . . . . . . . . . . . . . . . . . . . . . . . . . 70
6.2.1 Intravitreal Triamcinolone
torial disease with a complex pathogenesis that
for Macular Edema in Central often reflects locally systemic vascular and hemo-
Retinal Vein Occlusion . . . . . . . . . . . . . . . . 70 dynamic disorders [11]. Nowadays, it is generally
6.2.2 Intravitreal Triamcinolone agreed that the RVO management should target
for Macular Edema in Branch
Retinal Vein Occlusion . . . . . . . . . . . . . . . . 70
the angiogenic factors as well as the inflamma-
6.2.3 Dexamethasone Implant Treatment tory compounds of this etiologic complex. The
of Macular Edema in RVO. . . . . . . . . . . . . . 71 inflammatory reactions put in motion by retinal
6.2.4 Fluocinolone Acetonide ischemia and subsequent macular edema in RVO
Intravitreal Implant . . . . . . . . . . . . . . . . . . . 74
6.2.5 Combined Treatments . . . . . . . . . . . . . . . . . 74
are closely associated to the angiogenic mani-
6.2.6 Systematic Reviews Comparing festation of the pathology and include various
Different Treatment Modalities interrelated processes such as vasodilatation, leu-
for RVO . . . . . . . . . . . . . . . . . . . . . . . . . . . . 75 kostasis, diapedesis, increased vascular perme-
6.3 Safety Issues . . . . . . . . . . . . . . . . . . . . . . . . 75 ability, and inflammatory proteins secretion [2].
6.3.1 Intravitreal Triamcinolone . . . . . . . . . . . . . . 76 Macrophages, neutrophils, and microglia partici-
6.3.2 Dexamethasone Intravitreal Implant . . . . . . 76
pate in the concert of inflammatory responses,
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77 in which several inflammatory mediators such
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78 as prostaglandins, IL-1, TNF-alpha, VEGF, and
ICAM-1, among others, are involved [20]. These
factors promote hyperpermeability of blood ves-
sel wall, contributing to the breakdown of blood
retinal barrier, and subsequently leading to macu-
lar edema. In patients with CRVO, it was found
that ICAM-1 and VEGF levels in the vitreous
in vivo correlated with the severity of macu-
E. Zunz, MD A. Loewenstein, MD (*) lar edema [16]. The rationale of using steroids
Department of Ophthalmology, that are well known for their potent anti-inflam-
Sackler Faculty of Medicine, matory action in RVO is based on their ability,
Tel Aviv Medical Center, Tel Aviv University,
among others, to block IL-1 mRNA synthesis and
6 Weizmann St, Tel Aviv 64239, Israel
e-mail: eranzunz@gmail.com; ICAM-1-mediated leukocyte adhesion to vessel
anatl@tlvmc.gov.il walls and to inhibit VEGF expression [6, 16].

A.J. Augustin (ed.), Intravitreal Steroids, 69


DOI 10.1007/978-3-319-14487-0_6, Springer International Publishing Switzerland 2015
70 E. Zunz and A. Loewenstein

Steroids delivered into the vitreous have the change in VA was a loss of 1.2 letters in the treat-
advantage of directly targeting the edematous ment groups as opposed to 12.1 letters loss in the
macula and leaking retinal blood vessels, with a observation group. The likelihood of VA gain
stronger and more efficient action against the dis- was five times greater in the IVTA groups than
ease processes and without inducing systemic the observation group. The mean number of
side effects. There are various drug formulations, injections after 12 months was 2 in the 4 mg
delivery methods, and treatment doses protocols. group and 2.2 in the 1 mg group.
In the present overview, we will evaluate the effi- Although the subsequent 2-year extension of
cacy and safety of the most commonly used intra- the SCORE study lacked definitive data, the
vitreal steroids in current practice. authors noted that the visual acuity results after
2 years were similar to those at 12 months. As
cataract had some effect on 2 years visual acuity
6.2 Efcacy results, cataract surgery was encouraged when-
ever it was clinically indicated by the investiga-
6.2.1 Intravitreal Triamcinolone tors. The authors stated that these findings
for Macular Edema in Central support the continuation of IVTA for 2 years
Retinal Vein Occlusion when indicated.
The SCORE study demonstrated the efficacy
The first multicenter RCT that investigated the and safety of 1 mg preservative-free intravitreal
efficacy and safety of intravitreal steroids was the triamcinolone (Trivaris: Allergan) in the treat-
Standard Care vs. Corticosteroid for Retinal Vein ment of CRVO. This preservative-free formula-
Occlusion (SCORE) Study. It included 271 tion is not available in Europe, and there is no
patients with macular edema and perfused extensive and clear evidence that the off-label
CRVO. It evaluated two doses of intravitreal tri- formulation of intravitreal triamcinolone aceton-
amcinolone (preservative-free), 1 and 4 mg, com- ide, which is non-preservative free presents simi-
pared to observation [10]. Eyes were treated with lar efficacy and safety profile [2].
intravitreal triamcinolone (IVTA) every 4 months
for up to a year. The study stated three criteria for
withholding retreatment during this year, which 6.2.2 Intravitreal Triamcinolone
included significant clinical improvement (VA for Macular Edema in Branch
improved to 20/25 or better or OCT with Retinal Vein Occlusion
CMT = 225 m), significant side effect (such as
IOP elevation that required treatment), or nonsig- In a parallel RCT on patients with BRVO, the
nificant response to two consecutive injections 1 and 4 mg IVTA treatment arms were com-
after 8 months (i.e., less than 20 % reduction in pared to the standard treatment of BRVO with
retinal thickness compared to baseline and less grid laser for macular edema (as opposed to no
than five letters improvement in VA score). The treatment in the CRVO patients). The results
SCORE allowed salvage IVTA for some patients showed that 28.9 % of the grid laser eyes
with CRVO, who had significant visual loss at improved by 15 letters in VA, which was not
two consecutive visits that was considered sec- significantly different from the 1 and 4 mg
ondary to macular edema and not to other causes IVTA treatment groups in which the VA of
such as cataract [10]. 25.6 and 27.2 % of the eyes gained 15 letters
The SCORE study demonstrated a gain of VA or more, respectively. The investigators con-
letter score of 15 or more in 26.5 % of eyes cluded that triamcinolone was not superior to
treated with 1 mg IVTA, 25.6 % of eyes treated standard care (grid laser) in treating macular
with 4 mg IVTA, and 6.8 % of eyes under obser- edema secondary to BRVO. The mean change
vation alone at 1 year. At 12 months, the mean in VA at 12 months was 4.2 in laser eyes, 5.7 in
6 Intravitreal Steroids for the Treatment of Macular Edema in Retinal Vein Occlusions 71

1 mg IVTA, and 4.0 in 4 mg IVTA. Three After this period patients were eligible to a sec-
additional reasons led to the abandonment ond dexamethasone implant of 0.7 mg if BCVA
of IVTA as treatment for macular edema in was lower than 20/20 or central macular thick-
BRVO. Firstly, the 3-year results indicated that ness higher than 250 m [9].
the laser group achieved in average a better VA Improvement of 15 letters or more in visual
than the IVTA groups (12.9 letters gain in laser acuity was documented in the 0.7 mg dexametha-
group, 4.4 letters gain in 1 mg IVTA group, and sone group in 30 % of patients 60 days after the
8 letters gain in 4 mg IVTA group). Secondly, first injection and in 32 % of patients 60 days
pseudophakic eyes also failed to demonstrate after the second injection. These figures dropped
better improvement with steroids rather than to 16 and 24 % 180 days after the first and second
laser, and thirdly, adverse events such as cata- injection, respectively. The peak of visual acuity
ract formation and elevated IOP were more gain of a mean of ten letters occurred 60 days
frequent in the IVTA groups than in the laser after the implant injections. Improvement of ten
group, with the highest frequency in the 4 mg letters or more was recorded in 55 % of eyes
treatment group [21]. treated with two dexamethasone implant injec-
tions and in up to 46 % of eyes treated with only
one dexamethasone injection at day 180 (delayed-
6.2.3 Dexamethasone Implant treatment group).
Treatment of Macular
Edema in RVO 6.2.3.1 Effects of Treatment
in Different Study Groups
Dexamethasone is a potent corticosteroid capa- According to the subgroup analysis of the
ble of reducing or suppressing the mediators GENEVA trial, patients with CRVO who had
promoting macular edema. It is highly soluble received the injection at baseline returned to the
with a short half-life. To ensure a prolonged mean baseline visual acuity after 180 days but
intraocular action, a biodegradable implant was gained an average of two letters (compared to
developed (Ozurdex: Allergan). The implant baseline) at conclusion of the study after 1 year,
contains 0.7 mg dexamethasone in a polyglyco- i.e., 180 days after the second 0.7 mg Ozurdex
late-acetate coating. After being injected intra- implant injection.
vitreally, it slowly releases dexamethasone into Patients treated for CRVO, who were initially
the vitreous for up to 6 months with maximal randomized to sham and then received their first
effect 60 days after injection [2]. The implanted dexamethasone implant at day 180, did not catch
device is able to deliver a long-standing steroid, up with the early treatment group visual acu-
and thus it has the potential to reduce the fre- ity results. Not only that they did not achieve a
quency of injections with alternative or comple- mean of 2 and 1 letters change in BCVA (as
mentary treatments. compared with baseline) at day 180 and day
The GENEVA trial evaluated one to 360, respectively, but they showed only a mean
two intravitreal dexamethasone biodegradable of Four letters improvement peak (instead of
implant injections in the treatment of macular eight letters peak) 60 days after injection. This
edema in CRVO and BRVO for 12 months. The reflects a mean improvement in VA of two letters
study was conducted as a randomized controlled after 180 days as compared with no treatment
multicenter study in the first 180 days and then and of three letters as compared with delayed
as an open label in the following 180 days single dexamethasone intravitreal implant in the
extension. GENEVA trials.
Patients were randomized at baseline to either Another important finding of this study was
0.7 or 0.35 mg dexamethasone implant or sham that eyes with macular edema that were treated
injection, and they were followed up for 180 days. within the first 3 months since the onset of
72 E. Zunz and A. Loewenstein

Fig. 6.1 (a) A 55-year-old man. Severe persistent CME 6 weeks s/p Ozurdex injection. BCVA improved to 6/8.5.
d/t CRVO in only eye after multiple bevacizumab injec- IOP = 20, CMT = 286 m. (c) Complete resolution of mac-
tions and no satisfactory result. BCVA 6/15. ular edema at 11 weeks post Ozurdex injection. BCVA
CMT = 656 m. (b) Nearly complete resolution of CME 6/8.5 IOP = 17, CMT = 280 m

macular edema had a higher probability to no-treatment group. This difference was
improve as compared with eyes with longer- statistically significant [2, 9]. Along all the time
standing macular edema [8]. Upon the evidence points of the study, BRVO eyes treated with
gathered in the GENEVA trial, Ozurdex received dexamethasone 0.7 mg demonstrated a higher
the approval for use in macular edema in CRVO, mean visual gain than the sham BRVO group, the
indication for which it could be the first-line difference being statistically significant. In
treatment choice [2] (Fig. 6.1). the open-label phase of the study, 60 days
In patients with BRVO, treated eyes demon- after the second dexamethasone implant injec-
strated a mean peak of over ten letters gain at day tion, visual acuity gain peaked to a mean of ten
60 after first dexamethasone implant injection, letters as compared with eight letters gain in the
compared with five letters gain with no treat- delayed-treatment group. After 1 year, the mean
ment. At day 60 after injection, 29.6 % of BRVO gain was six letters, in both groups, the BRVO
eyes treated with dexamethasone 0.7 mg gained eyes injected twice and eyes with a single injec-
15 letters or more as compared with 12.5 % of the tion at day 180 [8]. Similar to the findings in
sham group eyes [2]. By day 180, 41.2 % of CRVO eyes, in the open-label phase of the study,
0.7 mg dexamethasone-treated eyes improved the eyes with BRVO that had received delayed
by ten letters or more versus 33 % in the treatment had a lower visual acuity gain than
6 Intravitreal Steroids for the Treatment of Macular Edema in Retinal Vein Occlusions 73

Fig. 6.2 (a) A 58-year-old woman with superotemporal dexamethasone implant (Ozurdex) OCT demonstrates
BVO. A macula at presentation with severe CME. BCVA complete resolution of CME. BCVA improved to 6/30. (c)
6/60. After single bevacizumab injection, the patient 10 weeks after Ozurdex injection, no macular edema, cen-
developed subarachnoid hemorrhage, and anti-VEGF tral retinal thickness reduced to 267 m, and BCVA
agents were contraindicated. (b) 33 days s/p intravitreal improved to 6/12

those in the initial-treatment groups [8]. Further dexamethasone intravitreal implant. Such proba-
analysis found that delaying treatment for eyes bility was reduced by 54 %, 32 %, and 12 % if the
with BRVO and macular edema reduces the treatment was delayed by 6, 3, and 1 month,
chance to achieve 15 letters gain in vision with respectively [2] (Fig. 6.2).
74 E. Zunz and A. Loewenstein

6.2.3.2 Effects of Treatment on Retinal treatment arm and followed for 6 months. The
Thickness as Measured on OCT results demonstrated no statistically significant
Studies difference in visual outcomes between groups.
In both CRVO and BRVO, 90 days after the sec- The bevacizumab eyes had a significantly thinner
ond implant injection, the mean retinal thickness central retina at the end of month 1, but this effect
change was a reduction of 263 m, while a was not evident in any of the following months of
decrease of 267 m was noted in patients who the study. As expected, the eyes with dexametha-
had received their first dexamethasone implant at sone implant had a statistically significant higher
day 180 (delayed-treatment group) [8]. IOP between months 3 and 6 [5]. Considering the
More clinical data on extended use and results scarce comparative clinical data, and despite its
of dexamethasone implants in retinal vein occlu- small sample size and the non multicenter RCT
sion have been added quite recently. A retrospec- design, this study may have a significant impact
tive case series of 51 eyes with RVO and ME on future head-to-head, larger studies comparing
examined the effectiveness of repeated dexa- anti-VEGF agents with the dexamethasone intra-
methasone intravitreal implant injections in 51 vitreal implant. A large, ongoing multicenter RCT
eyes. After 12 months of follow-up, 30 % of eyes (the COMO study (http://clinicaltrials.gov/show/
gained 15 or more letters. At 12 months BRVO NCT01427751) was designed to compare the
treated eyes had ETDRS letter gain of 5.7 and efficacy of ranibizumab versus intravitreal dexa-
CRVO eyes gained a mean of 11.5 letters com- methasone implant in eyes with early BRVO of
pared to baseline visual acuity versus 5 and 2 let- less than 90 days. It is expected to complete the
ters gain, respectively, in the GENEVA trial. It data collection for the primary outcome measure
was found that, with repeated injections, the (the change from baseline in best corrected visual
dexamethasone implants effect had a shorter acuity) only by the end of 2014. Clinical evidence
duration. In this series, the mean number of dexa- based on well-designed head-to-head RCTs of
methasone implant injections after 12 months different intravitreal substances is needed.
was 1.9, but 23 % of the eyes required three or
more injections. The minimal interval between
repeated injections was 3 months. Retreatment 6.2.4 Fluocinolone Acetonide
was performed if after an initial response to the Intravitreal Implant
first injection, the patient experienced a visual
loss of at least five letters, with macular edema The Iluvien intravitreal implant was designed to
relapse on OCT. This study concluded that, espe- release fluocinolone acetonide over a period of
cially in eyes with CRVO, dexamethasone 36 months. In 2009, Alimera Inc. initiated the
implants can be injected on as-needed protocol, FAVOR study to examine the efficacy and safety
but its outcomes remained similar to those of Iluvien for RVO. After the results were
reported by the SCORE trial [12]. A latest post reported, this treatment modality for RVO was
hoc analysis of the GENEVA trial revealed that abandoned.
visual gain in response to dexamethasone implant
injection occurred as early as 7 days postinjec-
tion, when 27.2 % of treated eyes presented with 6.2.5 Combined Treatments
a ten or more letters gain [13].
6.2.5.1 Combined Laser Grid
6.2.3.3 Anti-VEGF Versus and Dexamethasone
Dexamethasone Intravitreal Intravitreal Implants
Implants Efcacy in RVO A small prospective randomized study examined
A recent small randomized clinical trial com- the efficacy of dexamethasone implant versus
pared anti-VEGF (bevacizumab) as needed with a dexamethasone implant plus laser grid
single dexamethasone implant in 60 nonischemic 68 weeks after injection. Each treatment arm
CRVO eyes. Thirty eyes were randomized to each included 25 eyes, all of which were eligible for
6 Intravitreal Steroids for the Treatment of Macular Edema in Retinal Vein Occlusions 75

repeated dexamethasone intravitreal implant One such recent review [4] assessed eight
injection when macular edema relapsed or when RCTs that evaluated the following available treat-
there was a decrease in visual acuity. Although ments for CRVO: triamcinolone, dexamethasone,
at 6 months both groups improved the vision, the bevacizumab, pegaptanib, ranibizumab, and
combination group achieved a mean of 0.18 aflibercept. This systematic review concluded
LogMAR, while the injections-only group 0.25 that bevacizumab, triamcinolone, aflibercept, and
LogMAR. Interestingly, in the combination ranibizumab demonstrated a significant higher
treatment arm, only 12 % of patients were proportion of eyes gaining 15 or more letters
retreated for macular edema relapse at 4 months compared with sham. The results for pegaptanib
as opposed to 48 % of patients in the injections- and dexamethasone were mixed, because of
only group. At 6 months, macular edema per- safety issues, with significant cataract formation
sisted in 12 % of the eyes in the injections-only and IOP elevation under intravitreal steroids as
treatment arm and in none in the combined treat- opposed to no significantly higher adverse event
ment arm. This study demonstrated that the rate under anti-VEGF therapies [4].
combination of slow-release dexamethasone A recently published systematic review by
implant with laser grid could achieve better Glanville et al. [7] examined the effects of ranibi-
visual results and can secure longer intervals zumab or dexamethasone for both BRVO and
between injections [18]. CRVO. This study found that dexamethasone
provided a significant improvement in visual
6.2.5.2 Combined Treatment acuity in BRVO eyes but not in CRVO eyes. As
of Intravitreal Triamcinolone mentioned earlier, a head-to-head comparison
and Ranibizumab between these treatments was not possible due to
A new small but prospective study compared the different patients baseline characteristics and
treatment results in 57 eyes with CRVO that were study design [7].
assigned to either initial combined ranibizumab and Another systematic review by Pielen et al.
triamcinolone (30) or ranibizumab alone (27). [19] summarized data from 11 RCTs of different
Improvement was seen in both treatment groups dur- treatment modalities for BRVO and CRVO. This
ing the study. The only statistically significant differ- review found that in CRVO a visual improvement
ence between the groups was the mean number of of at least 13.9 letters was achieved with a mean
injections, namely, 3.42 in the combination group as of 8.8 injections of different anti-VEGF agents
compared with 4.23 in the initial ranibizumab alone. (either aflibercept, bevacizumab, or ranibi-
Differences in central subfield thickness or visual zumab), and a stabilization of vision (1.2 letters
acuity between groups were not significant [3]. compared to baseline) was achieved with a mean
of 2.2 injections of triamcinolone (either 4 mg or
1 mg). In BRVO, the ranibizumab-treated eyes
6.2.6 Systematic Reviews achieved 18.4 letters (in average 8.3 injections).
Comparing Different In both BRVO and CRVO, the Ozurdex-treated
Treatment Modalities for RVO eyes achieved a transient visual improvement
with a mean of 1.9 injections, but the comparison
Systematic reviews attempted to compare differ- is limited as the RCTs of dexamethasone did not
ent treatments available for RVO. To note that, include the PRN treatment [19].
although systematic analyses have advantages
when comparing clinical outcomes, not all RCTs
are comparable in terms of patients selection cri- 6.3 Safety Issues
teria and the measured outcomes. For this reason,
the systematic reviews emphasize in their conclu- In addition to side effects such as pain, subcon-
sions the need for large RCT for RVO comparing junctival hemorrhage, floaters, transient IOP
different treatments head to head similar to the increase, or, rarely, endophthalmitis that are com-
CATT study for AMD [14]. monly related to the procedure itself regardless of
76 E. Zunz and A. Loewenstein

the therapeutic agent, there are some adverse During 12 months of follow-up in the SCORE
effects or complications attributed to intravitreal BRAVO study, the reported cataract formation
steroids [17]. and progression rates were 35 % in the 4 mg tri-
amcinolone arm, 25 % in the 1 mg arm, and 13 %
in the standard care group. Only few cataract sur-
6.3.1 Intravitreal Triamcinolone geries were performed over the first year, but by
the end of the second year, 35 of eyes in the 4 mg
6.3.1.1 Elevated IOP group, 8 eyes in the 1 mg group, and 6 eyes in the
During the 12 months of CRVO SCORE Study, standard treatment group underwent cataract
IOP-lowering drops were prescribed for 35 % of extraction. Rates of cataract formation, progres-
4 mg IVTA patients, 20 % of 1 mg IVTA, and 8 % sion, and surgery were statistically significantly
of the patients in the observation group. Two higher in the 4 mg triamcinolone arm compared
patients who received 1 mg IVTA required tube to either the 1 mg or standard treatment arms.
shunt operation during the first year, and two of Since cataract side effects were not significantly
the patients receiving 4 mg IVTA underwent this higher in the 1 mg triamcinolone group compared
procedure during the second year. All of the oper- to laser grid (standard care), this treatment is con-
ated patients suffered from neovascular glaucoma sidered relatively safe [21].
and not from steroid-induced glaucoma [10].
In the BRVO SCORE Study, higher frequency
of IOP increase was noted in the IVTA treatment 6.3.2 Dexamethasone Intravitreal
arms, which was highest in the 4 mg arm. Implant
Elevated IOP requiring medical treatment was
found in 41 %, 8 %, and 2 % of the eyes in the The GENEVA trial included extensive safety
4 mg, 1 mg, and laser arms, respectively, while analysis concerning treatment with intravitreal
36 %, 7 %, and 0.7 % of eyes, respectively, had dexamethasone implants. The most frequent
an IOP rise of 10 mmHg or more above baseline. adverse effects were conjunctival hemorrhage
Severe IOP increase (above 35 mmHg) was and elevation of IOP. After 1 year, the cumulative
recorded in 10 % of the eyes treated with 4 mg frequency of conjunctival hemorrhage was nearly
IVTA and in less than 2 % in other treatment 25 % of cases and did not differ significantly
groups. None required glaucoma surgery by between treatment arms. Actually, excluding cat-
12 months but two patients from the 4 mg group aract formation, there were no statistically sig-
required glaucoma surgery to lower IOP (one nificant differences in frequencies of any of the
tube, one trab) [21]. ocular side effects between patients who received
one or two injections of dexamethasone implant
6.3.1.2 Cataract during the 12 months of the study. Endophthalmitis
In the CRVO SCORE Study, cataract progres- was not reported in any of the study eyes.
sion was recorded in 18 % in the nontreatment
group, 26 % of the patients in the 1 mg IVTA, 6.3.2.1 IOP Elevation
and in 33 % of patients in the 4 mg IVTA arm. It Elevated IOP rates demonstrated peaks at days
was found that only the 4 mg group had a signifi- 60 and 240, namely, 60 days after each injection.
cantly higher rate of cataract surgery (27 %) Incidence was 12.6 % after first injection and
between the months 1224 of the study com- 15.4 % after second injection. Although up to
pared to the observation (0 %) or 1 mg group 32.8 % of patients who received two dexametha-
(3 %), which implies that significant cataract sone implant injections experienced an IOP ele-
may not be expected in significantly higher rates vation equal or larger than 10 mmHg during
in eyes receiving 1 mg IVTA as compared with some point of the study, this effect was transient
no treatment strategy. No events of endophthal- and resolved 180 days after injection of the
mitis or retinal detachment occurred at 12 months implant. It should be noted that those elevations
in this study [10]. were either observed or treated topically. After
6 Intravitreal Steroids for the Treatment of Macular Edema in Retinal Vein Occlusions 77

the first injection, 25.5 % of eyes required IOP- results of VA in patients who received dexameth-
lowering medications and up to 38.8 % of the asone as monotherapy were not reported. Cataract
eyes injected twice with intravitreal dexametha- progression was not systematically recorded in
sone implant required this treatment. In total 14 this study, but 46 eyes out of 289 (15.9 %) under-
eyes required laser or surgical measures to lower went cataract extraction during the study period.
the IOP. Four of those cases developed neovascu- Concerning IOP elevations, the study reported
lar glaucoma secondary to the RVO and this was that 32.6 % of eyes sustained a 10 mmHg eleva-
the indication for IOP lowering [8]. tion or more (at any of the visits). Despite that
A retrospective case series of 51 eyes treated 29.1 % of eyes required topical IOP-lowering
with Ozurdex implants for RVO reported that agents some time during the follow-up, only
27 % of eyes suffered from an IOP increase of 4.3 % presented with elevated IOP at final fol-
more than 25 mmHg after the first injection. IOP low-up visit [1].
in almost all of these patients was controlled Another long-term study assessed the fre-
under topical treatment only [12]. quency of side effects and long-term visual prog-
nosis with dexamethasone intravitreal treatment
6.3.2.2 Cataract [15]. It included 17 patients who had participated
Cataract was reported in 29.8 % of eyes treated in the GENEVA Study. The mean follow-up was
with two injections of Ozurdex and in 10.5 % of of 50.5 months. Fourteen out of 17 patients with
eyes in the delayed-treatment group. These BRVO demonstrated an improvement of two
differences were statistically significant. lines.
Conversely, cataract formation rates were not sig- Although side effects frequencies were not
nificantly different between eyes that received mentioned per original treatment arm group in
one or no intravitreal dexamethasone implant, the original GENEVA trial, this small series
namely, between 5.7 and 7.6 % [8]. Cataract reflects real-life scenario. Seven out of 17 of the
extraction rates during study period were very eyes had two Ozurdex injections during the origi-
low and nonsignificantly different between treat- nal GENEVA trial and the other ten patients had
ment arms. only one injection. Regardless of the number of
injections, at the last follow-up (mean follow-up
6.3.2.3 Long-Term, Real-Life of 50.5 months), the average IOP levels in all the
Retrospective Data of Patients study sample was 14.7 mmHg and only one
Treated with Dexamethasone patient required topical treatment for elevated
Intravitreal Implant Monotherapy IOP above 21 mmHg throughout the follow-up
or with Combined Treatments period. This finding may suggest a favorable
Few retrospective studies are available concern- long-term rate of elevated IOP after one or two
ing long-term efficacy and safety of Ozurdex Ozurdex injections. Long-term cataract progres-
treatment. These reports are not homogenous in sion rate was 58.8 % (ten eyes) and 35.2 % (six
terms of the number of Ozurdex injections patients) underwent cataract extraction after a
received and whether combined therapy with mean period of 28.3 18.7 months [15].
anti-VEGF or laser was applied. Nevertheless,
the results provide some evaluation of the data Conclusion
regarding the safety and efficacy issues with the Without any doubt, intravitreal steroids have
treatment of dexamethasone intravitreal implants an important place in RVO treatment arma-
combined with anti-VEGF treatments. mentarium. Ozurdex that received the FDA
A multicenter retrospective review of 289 approval for treatment of macular edema in
eyes treated with a mean of 3.2 Ozurdex injec- retinal vein occlusion was shown in the
tions (29.1 % as monotherapy) and a mean inter- GENEVA trials and in more recent studies to
val between injections of 5.6 months reported be effective and safe in both BRVO and CRVO
that 59.7 % of BRVO eyes and 66.7 % of CRVO and to achieve significant visual gain espe-
eyes improved two or more lines in vision. The cially when multiple dexamethasone implants
78 E. Zunz and A. Loewenstein

were used on an as-needed basis under IOP secondary to central retinal vein occlusion. Curr Eye
control. Intravitreal triamcinolone remains a Res. 2014;39:93843.
4. Ford JA, Clar C, Lois N, Barton S, Thomas S, Court R,
valid alternative for treatment of macular Shyangdan D, Waugh N. Treatments for macular oedema
edema if other means such as anti-VEGF or following central retinal vein occlusion: systematic
Ozurdex have failed. This treatment, though, review. BMJ Open. 2014;4(2):e004120. doi:10.1136/
is not available as preservative-free formula- bmjopen-2013-004120.
5. Gado AS, Macky TA. Dexamethasone intravitreous
tion in Europe, and the preservative-contain- implant versus bevacizumab for central retinal vein
ing formula has not been evaluated for efficacy occlusion-related macular oedema: a prospective ran-
and safety in RVO [2]. domized comparison. Clin Experiment Ophthalmol.
2014. doi:10.1111/ceo.12311. [Epub ahead of print]
Steroids in RVO offer effective monother-
PubMed PMID: 24612095.
apy and are indeed capable of upgrading other 6. Glacet-Bernard A, Coscas G, Zourdani A, Soubrane
treatment modalities by inducing a compre- G, Souied EH. Steroids and macular edema from reti-
hensive and long-standing anti-inflammatory nal vein occlusion. Eur J Ophthalmol. 2011;21 Suppl
6:S3744.
and antiangiogenic effect and allow reduction
7. Glanville J, Patterson J, McCool R, Ferreira A, Gairy
of burden of intravitreal anti-VEGF injec- K, Pearce I. Efficacy and safety of widely used treat-
tions. Intravitreal steroids are also an alterna- ments for macular oedema secondary to retinal vein
tive option for patients not responding to occlusion: a systematic review. BMC Ophthalmol.
2014;21:147.
regular anti-VEGF treatments, and there is
8. Haller JA, Bandello F, Belfort Jr R, Blumenkranz MS,
some evidence that their combination with Gillies M, Heier J, Loewenstein A, Yoon YH, Jiao J,
laser in BRVO may be synergistic. Li XY, Whitcup SM, Ozurdex GENEVA Study
Our knowledge is constantly evolving with Group, Li J. Dexamethasone intravitreal implant in
patients with macular edema related to branch or cen-
the new developments in the retina field. Clinical
tral retinal vein occlusion twelve-month study results.
trials have yet to answer many questions regard- Ophthalmology. 2011;118(12):245360.
ing the way in which steroids affect the ocular 9. Haller JA, Bandello F, Belfort Jr R, Blumenkranz MS,
diseased structures as well as the normal eye Gillies M, Heier J, Loewenstein A, Yoon YH, Jacques
ML, Jiao J, Li XY, Whitcup SM, OZURDEX
structures. We need more studies that will exam-
GENEVA Study Group. Randomized, sham-
ine other combination therapies, different treat- controlled trial of dexamethasone intravitreal implant
ment protocols such as multiple Dexamethasone in patients with macular edema due to retinal vein
implant injections including PRN regimen, and occlusion. Ophthalmology. 2010;117(6):113446.e3.
10. Ip MS, Scott IU, VanVeldhuisen PC, Oden NL, Blodi
head-to-head efficacy investigations.
BA, Fisher M, Singerman LJ, Tolentino M, Chan CK,
Gonzalez VH, SCORE Study Research Group. A ran-
domized trial comparing the efficacy and safety of intra-
vitreal triamcinolone with observation to treat vision loss
References associated with macular edema secondary to central reti-
nal vein occlusion: the Standard Care vs Corticosteroid
1. Capone Jr A, Singer MA, Dodwell DG, Dreyer RF, for Retinal Vein Occlusion (SCORE) study report 5.
Oh KT, Roth DB, Walt JG, Scott LC, Hollander Arch Ophthalmol. 2009;127(9):110114.
DA. Efficacy and safety of two or more dexa- 11. Jaulim A, Ahmed B, Khanam T, Chatziralli IP. Branch
methasone intravitreal implant injections for retinal vein occlusion: epidemiology, pathogenesis,
treatment of macular edema related to retinal vein risk factors, clinical features, diagnosis, and compli-
occlusion (Shasta study). Retina. 2014;34(2): cations. An update of the literature. Retina.
34251. 2013;33:90110.
2. Coscas G, Loewenstein A, Augustin A, Bandello F, 12. Joshi L, Yaganti S, Gemenetzi M, Lightman S,
Battaglia Parodi M, Lanzetta P, Mons J, de Smet M, Lindfield D, Liolios V, Menezo V, Shao E, Taylor
Soubrane G, Staurenghi G. Management of retinal vein SR. Dexamethasone implants in retinal vein occlusion:
occlusionconsensus document. Ophthalmologica. 12-month clinical effectiveness using repeat injections
2011;226(1):428. as-needed. Br J Ophthalmol. 2013;97(8):10404.
3. Fan C, Wang Y, Ji Q, Zhao B, Xie J. Comparison of 13. Kuppermann BD, Haller JA, Bandello F, Loewenstein
clinical efficacy of intravitreal ranibizumab with and A, Jiao J, Li XY, Whitcup SM. Onset and duration of
without triamcinolone acetonide in macular edema visual acuity improvement after dexamethasone
6 Intravitreal Steroids for the Treatment of Macular Edema in Retinal Vein Occlusions 79

intravitreal implant in eyes with macular edema due macular grid laser in patients with branch retinal vein
to retinal vein occlusion. Retina. 2014;34:17439. occlusion. Am J Ophthalmol. 2014;157(3):60715.
14. Martin DF, Maguire MG, Fine SL, Ying G, Jaffe GJ, e1.
et al. Ranibizumab and bevacizumab for treatment of 19. Pielen A, Feltgen N, Isserstedt C, Callizo J, Junker B,
neovascular age-related macular degeneration: two- Schmucker C. Efficacy and safety of intravitreal ther-
year results. Ophthalmology. 2012;119(7):138898. apy in macular edema due to branch and central reti-
15. Moisseiev E, Goldstein M, Waisbourd M, Barak A, nal vein occlusion: a systematic review. PLoS One.
Loewenstein A. Long-term evaluation of patients 2013;8(10):e78538.
treated with dexamethasone intravitreal implant for 20. Scholl S, Augustin A, Loewenstein A, Rizzo S,
macular edema due to retinal vein occlusion. Eye Kupperman B. General pathophysiology of macular
(Lond). 2013;27(1):6571. edema. Eur J Ophthalmol. 2011;21 Suppl 6:S109.
16. Noma H, Funatsu H, Harino S, Mimura T, Eguchi S, 21. Scott IU, Ip MS, VanVeldhuisen PC, Oden NL, Blodi
Hori S. Vitreous inflammatory factors in macular BA, Fisher M, Chan CK, Gonzalez VH, Singerman
edema with central retinal vein occlusion. Jpn J LJ, Tolentino M, SCORE Study Research Group. A
Ophthalmol. 2011;55(3):24855. randomized trial comparing the efficacy and safety
17. Peyman GA, Lad EM, Moshfeghi DM. Intravitreal of intravitreal triamcinolone with standard care to
injection of therapeutic agents. Retina. 2009;29(7): treat vision loss associated with macular edema sec-
875912. ondary to branch retinal vein occlusion: the Standard
18. Pichi F, Specchia C, Vitale L, Lembo A, Morara M, Care vs Corticosteroid for Retinal Vein Occlusion
Veronese C, Ciardella AP, Nucci P. Combination ther- (SCORE) study report 6. Arch Ophthalmol.
apy with dexamethasone intravitreal implant and 2009;127(9):111528.
Treatment of Uveitis
with Intraocular Steroids 7
Lazha Talat, Filis Ismetova, Susan Lightman,
and Oren Tomkins-Netzer

Contents 7.1 Pathophysiology of Uveitis


7.1 Pathophysiology of Uveitis . . . . . . . . . . . . 81
Uveitis, or inflammation inside the eye, is a
7.2 Steroid Treatment in Uveitis . . . . . . . . . . . 82
relatively uncommon disease with a prevalence
7.3 Local Steroid Treatment in Uveitis. . . . . . 82 of 58.0114.5 per 100,000 persons [1, 2].
7.3.1 Intravitreal Triamcinolone Acetonide
However, it accounts for 1015 % of all causes of
(IVTA) Injection . . . . . . . . . . . . . . . . . . . . . 82
7.3.2 Extended-Release Dexamethasone blindness among people of working age in the
Implant (Ozurdex) . . . . . . . . . . . . . . . . . . . 85 developed world [3, 4]. Uveitis can be catego-
7.3.3 Fluocinolone Acetonide Intravitreal rized according to anatomical involvement (ante-
Implants . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
rior, intermediate, posterior, or panuveitis) or by
Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88 etiology (infectious, noninfectious, or neoplas-
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88 tic). While the aim of the immune system is to
control exogenous infections and promote tissue
healing, in doing so it may also result in damage
to tissue structure that, in the eye, can cause sig-
nificant functional consequences for vision. The
inflammatory response involves a myriad of
immune cells that are controlled by a balance of
proinflammatory and inhibitory cytokines.
Abnormal regulation of the immune system can
result in autoimmunity, excessive inflammatory
responses, and further tissue damage.
In order to maintain normal visual function,
the eye must remain an immune-privileged site to
L. Talat, MBChB, MPH, MSc (*)
which immune cells have restricted access and
F. Ismetova, MD, MPH
Department of Clinical Ophthalmology, function. The intact blood-ocular barriers impede
University College London/Moorfields Eye Hospital, the passage of blood constituents and cells into
162 City Road, London EC1V 2PD, UK the intraocular space. An intricate anti-
e-mail: l.talat.11@ucl.ac.uk; filis.ismetova.12@ucl.ac.uk
inflammatory and immunosuppressive microenvi-
S. Lightman, FRCP (UK), PhD FRCOphth ronment within the eye inhibits T-cell activation
O. Tomkins-Netzer, MD, PhD
and differentiation, suppresses macrophage acti-
UCL/Institute of Ophthalmology and Moorfields Eye
Hospital, 162 City Road, London EC1V 2PD, UK vation [5], inhibits natural killer cells lysis of tar-
e-mail: s.lightman@ucl.ac.uk; o.tomkins-netzer@ucl.ac.uk get cells [6], induces apoptosis of immune cells

A.J. Augustin (ed.), Intravitreal Steroids, 81


DOI 10.1007/978-3-319-14487-0_7, Springer International Publishing Switzerland 2015
82 L. Talat et al.

[7], and limits antigen presentation by downregu- increased risk of raised blood glucose levels, sys-
lation of major histocompatibility complex temic hypertension, cushingoid effects, reduced
(MHC) class II and MHC class I molecules on bone mass (steroid-induced osteopenia/osteopo-
immune and retinal cells [8]. Ocular inflammation rosis), and behavioral changes [19, 20]. In the
is T-cell regulated and animal models have indi- eye, systemic corticosteroids may cause cataract
cated that marked migration of cells into the eye formation/progression, central serous retinopa-
leads to onset of structural damage in the retina thy, or more rarely increased intraocular pressure
within 1215 days [7, 9]. In these models, inflam- (IOP). In children, systemic corticosteroid treat-
mation is mediated by T-helper-1 (Th-1) cells, ment can also rapidly result in reduced growth, as
while Th-2 cells promoted immunosuppression well as delayed puberty [21]. Indeed, the risks
and remission [1013]. Other T-cell types have associated with using systemic steroids over any
been associated with intraocular inflammation period of time have led to the increased use of
with downregulation of CD4+CD25+ regulatory locally administered steroids where possible for
T (Treg) cells and increased Th-17 cell activity the management of sight-threatening/sight-
both playing a role in the severity and resolution reducing intraocular inflammation [22].
of the inflammation [1416]. Dysfunction of
these immunosuppressive mechanisms results in
an uncontrolled intraocular immune response that 7.3 Local Steroid Treatment
can lead to irreversible structural damage and loss in Uveitis
of visual function.
While systemic corticosteroids are very effective
in controlling ocular inflammation, the options
7.2 Steroid Treatment in Uveitis for local treatment are increasing. Their advan-
tage is achieving high concentrations of the drug
Treatment of uveitis is based on the use of drugs within close proximity of the target area, while
that are directed at suppressing clinically evident minimizing the risk of systemic side effects.
inflammation, which can be administered either Corticosteroids can be administered around the
systemically or locally. The cornerstone for eye, through orbital floor or sub-tenon injections
immunosuppressive treatment is corticosteroids and can be effective in controlling intermediate
that act by multiple mechanisms including induc- or posterior uveitis using these routes [23].
ing phospholipase-A2 inhibitory proteins that Alternatively, use of intravitreal steroids or inser-
control the synthesis of inflammatory mediators tion of slow-releasing steroid implants may prove
such as prostaglandins and leukotrienes [17]. more effective under conditions when periocular
They inhibit vasodilatation, reduce vascular per- injections do not achieve adequate disease con-
meability, decrease leukocyte migration, and trol [24, 25]. In this chapter, we will describe the
reduce vascular endothelial growth factor various intravitreal steroids used to treat uveitis
(VEGF) levels [18]. Corticosteroids also sup- (Table 7.1).
press T-cell proliferation and cytokine production
and decrease activation mechanisms such as
interferon production. Systemic corticosteroids 7.3.1 Intravitreal Triamcinolone
are generally considered best for use in cases of Acetonide (IVTA) Injection
bilateral intraocular inflammation causing a
threat to vision or uveitis with concomitant 7.3.1.1 Dose and Administration
systemic disease which requires corticosteroid Triamcinolone acetonide (TA) is a water-
treatment. Though corticosteroids are highly insoluble crystalline steroid in a suspension
effective in controlling inflammation, they are which can be injected into the vitreous at a
not without attendant side effects. When given therapeutic dose of typically 2 or 4 mg.
systemically, steroids are associated with an Kenalog-40 (40 mg/mL, Bristol-Myers Squibb,
7 Treatment of Uveitis with Intraocular Steroids 83

Table 7.1 Comparison of intravitreal corticosteroids used for noninfectious uveitis


Duration Uveitis Improved
Dose of effect activity vision by Cataract Glaucoma
(mg) Half-life (months) resolve 15 letters surgery surgery
Kenalog (TA) 4 18 days 35 33 % in 51 % in 29 %in 01 % in
4 monthsa 4 weeks a 2 yearsb 1 yearc
Ozurdex 0.7 5.5 h 46 47 % in 53 % in 0.4 % in 0 % in
(dexamethasone) 2 months 3 monthsd 6 months 6 months
Retisert (FAc) 0.59 1.7 h 2430 12 % in 21 % in 80 % in 26 % in
2 yearse 2 yearse 2 yearse 2 yearse
Iluvien(FAc)f 0.019 111 days 2430
TA triamcinolone acetonide, FAc fluocinolone acetonide
a
Kok et al. [24]
b
Gillies et al. [45]
c
Vasconcelos-Santos et al. [69]
d
The dexamethasone DDS phase II study group, 2009 [54]
e
Multicenter uveitis steroid treatment (MUST) trial research group, 2011 [55]
f
Based on the use of Iluvien in diabetic eyes with CME. No data available in uveitic eyes

NJ) is the most commonly used form for off- 7.3.1.3 IVTA in Uveitis
label intravitreal injection with an advantage of IVTA was first administered in 1979 during reti-
being cheap and easily available compare to nal detachment surgery to reduce cellular prolif-
other alternative therapies. Other forms include eration [31]. These days it is widely used as a
Trivaris TM (80 mg/mL, Allergan Inc., Irvine, therapeutic option in the management of macular
CA) and Triesence (40 mg/mL, Alcon Inc., edema secondary to a variety of ocular patholo-
Fort Worth, TX), both preservative-free TA gies such as diabetic retinopathy [32], retinal
approved by the US food and drug administra- vein occlusion [33], and uveitis [34, 35]. In non-
tion (FDA) to treat ocular inflammatory dis- infectious uveitis, IVTA has been reported as
eases, although their availability in different effective in reducing cystoid macular edema
countries varies. (CME) and vitritis, including CME secondary to
Behets disease [36], Vogt-Koyanagi-Harada
7.3.1.2 Pharmacokinetics disease [37], non-TB-related serpiginous choroi-
Intravitreal injection can achieve a higher vit- ditis [38], and ankylosing spondylitis [39].
reous concentration of TA (1.2 g/mL) com- An early report of IVTA safety and efficacy
pared to that following sub-tenon injection of for CME in uveitis was a prospective pilot study
the same drug [26]. After the injection, the TA of six patients with chronic uveitis and visually
gradually settles in the inferior vitreous in significant CME, which was refractory to all sys-
most patients [27]. Once in the vitreous, it has temic agents. Clinical and angiographic resolu-
a half-life of approximately 18 days (3 days in tion of CME was evident in all patients by
eyes that underwent previous vitrectomy) with 6 weeks, together with improvement in visual
a therapeutic activity that lasts for approxi- acuity by at least two Snellen lines [40]. In a ret-
mately 3 months post injection [28]. The slow rospective study of 65 eyes with uveitis and
rate of clearance gives IVTA superiority over refractory CME managed with IVTA injections,
intravitreal injections of dexamethasone the maximum vision improvement occurred at a
sodium phosphate, which has a short half-life mean of 4 weeks during which 51 % had a sig-
of 5.5 h in the human eye [29] and is com- nificant improvement in visual acuity of at least
pletely cleared from the vitreous within 72 h two Snellen lines [24]. Additionally in pediatric
post injection [30]. patients, a clinically significant improvement in
84 L. Talat et al.

visual acuity was achieved in 56 % of 16 uveitic Thus, in children known to have experienced
eyes up to 15 months following IVTA injection steroid-induced raised IOP before, the use of
[41]. The long-term outcome of IVTA injections IVTA must be accompanied by measures to
has also been studied in uveitis secondary to ensure adequate IOP control, including surgical
Behets disease where 87 % of the eyes had intervention when needed.
complete resolution of vitritis within 6 months Owing to the cataractogenic effect of cortico-
post injection and 40 % had no relapse for up to steroids as well as that induced by intraocular
12 months [36]. inflammation, cataract progression and the even-
A single IVTA injection was found to be supe- tual need to undergo cataract surgery are com-
rior to a single intravitreal bevacizumab injection mon after IVTA injection especially if multiple
in improving the vision and reducing refractory injections are required. The risk of visually sig-
CME secondary to noninfectious uveitis 6 months nificant cataract requiring surgery post IVTA
following injection [42]. A clinical trial of 31 injection can vary but has been reported in one
eyes with CME in which patients were random- study to occur in 29 % of uveitic eyes at an aver-
ized to receive either repeat intravitreal injections age of 2 years (1234 months) post IVTA injec-
of either IVTA or bevacizumab found no signifi- tion compared to only 5 % of eyes that received a
cant difference in visual improvement between placebo injection [45]. Repeat injections increase
the groups up to 36 weeks. However, when the risk of developing significant cataract with up
adjusted for the development of cataract, the to 100 % of patients requiring cataract extraction
IVTA group had better visual outcome [43]. surgery by the fourth injection [46].
While IVTA injections allow for rapid and effec- Endophthalmitis is a rare, yet most serious
tive resolution of active uveitis and associated complication which can occur after any intravit-
macular edema, they have a relatively short half- real injection. With IVTA injection, the endo-
life [24, 28, 44], lasting several weeks to months, phthalmitis can be infective but can also be due to
limiting the duration of its therapeutic effect. noninfectious inflammation possibly secondary
Repeat injections are required usually every to a toxic reaction against triamcinolone aceton-
23 months, which increases the risk of develop- ide or its preservatives [47]. While a systematic
ing ocular side effects. review of literature from 1966 to 2004 estimated
the prevalence of all forms of endophthalmitis
7.3.1.4 Side Effects post IVTA injections to be 1.4 % (0.6 % for bac-
and Contraindications terial endophthalmitis) [48], the use of the cur-
The main side effects following IVTA injections rent preventive strategies has resulted in a lower
include increased IOP and progression of cata- incidence and recent reports found it ranging
ract. In a study examining the effect of IVTA in from 0 % to 0.21 % [5, 36, 49].
eyes with uveitis, by 4 weeks following injection, Viral retinitis following IVTA injection,
43 % of eyes had developed transient elevation in although rare, has also been reported, and a litera-
IOP of >10 mmHg [24]. A randomized clinical ture review found 30 reported cases of viral retini-
trial aimed at investigating the safety of a single tis within a mean of 4 months following intraocular
IVTA injection found increased IOP in 30 % of and periocular steroid injection [50]. Of these
cases, with almost two thirds requiring topical cases, 70 % occurred post IVTA injection with
antiglaucoma treatment for up to 8 months [45]. cytomegalovirus being the main isolated patho-
Young patients may also be vulnerable to devel- gen. Type 2 diabetes mellitus, human immune
oping increased IOP following IVTA injection deficiency virus (HIV) infection, and systemic
[22], shown in a study involving 16 eyes of pedi- immunosuppressive therapies were considered
atric patients, where an IOP increase of common comorbidities associated with the occur-
>15 mmHg occurred in 31 % of eyes [24, 41]. rence of viral retinitis post IVTA injection. IVTA
Other complications included progression of cat- should always be avoided in cases where infec-
aract (55 %) and recurrence of CME (31 %). tious uveitis has not been excluded, including
7 Treatment of Uveitis with Intraocular Steroids 85

uveitis secondary to toxoplasmosis reactivation visual acuity by at least 15 letters in 53.8 % of


that can progress to fulminant chorioretinitis fol- eyes that received the 0.7 mg implant compare to
lowing IVTA injection [51]. 16.7 % of eyes with the 0.35 mg implant and only
7 % in the observed group. In addition, improve-
ment in macular leakage on fluorescein angiogra-
7.3.2 Extended-Release phy was observed in 58 % of eyes treated with
Dexamethasone Implant the 0.7 mg implant compared to 8 % of eyes
(Ozurdex) without an implant [54]. The HURON trial, a
prospective randomized trial involving 229
7.3.2.1 Dose and Administration patients with noninfectious intermediate or pos-
The dexamethasone intravitreal implant terior uveitis, evaluated the efficacy of 0.35 and
(Ozurdex, Allergan, Inc., Irvine, CA) is a biode- 0.7 mg intravitreal dexamethasone implants in
gradable device that is inserted through the pars patients with significant vitreous haze over a
plana into the vitreous using a 23 gauge needle period of 8 weeks and then up to 26 weeks [55].
device preloaded with the implant. The implant At 8 weeks, there was a significant reduction in
itself contains a therapeutic dose of 0.7 mg pre- the vitreous haze score in 47 % of eyes receiving
servative-free dexamethasone. the 0.7 mg implant compare to 36 % and 12 % in
those receiving the 0.35 mg implant or sham
7.3.2.2 Pharmacokinetics injection, respectively. Approximately 34 % of
Dexamethasone is a potent, water-soluble, corti- the eyes with the 0.7 mg implant had reduction of
costeroid that is five times more potent than TA vitreous haze that was maintained up to 26 weeks
and less toxic to the retina, but also has a shorter postimplantation. In addition, visual acuity
half-life (3.55.5 h) and rapid clearance (72 h) improvement from baseline of 15 letters or more
when given as an intravitreal injection [30, 52]. was twice as likely to occur in treated eyes com-
Inserting the drug into an implant made of a solid pared to the sham-injected group.
bioerodable polymer allows for an extended, While these studies focused on the effective-
dual-phase release of dexamethasone, an initial ness of a single dexamethasone implant, a recent
rapid burst reaching peak concentration within observational study examined the outcome of
2 months, followed by a slower, sustained release repeated implants in eyes with noninfectious
detected for up to 6 months [52]. intermediate uveitis, posterior uveitis, and panu-
veitis [56]. In 38 eyes (63 % with repeat implan-
7.3.2.3 Dexamethasone Implants tations), there was an accumulating effect
in Uveitis resulting in continuing vision improvement and
Initially approved for the treatment of macular reduced central retinal thickness when compared
edema following retinal vein occlusion, dexa- to baseline. The relapse rate was 69 % within
methasone implants were approved by the FDA 6 months following the first implantation, and
in 2010 for the treatment of noninfectious inter- 48 % within 6 months following the second
mediate and posterior uveitis (Fig. 7.1) [53]. This implantation. A similar effect has also been
was the result of the encouraging outcomes observed in a cohort of pediatric patients with
observed in published reports by the dexametha- noninfectious uveitis [57].
sone drug delivery system (DDS) phase II study
group [54]. In the first clinical trial, 41 eyes with 7.3.2.4 Side Effects
persistent macular edema secondary to uveitis or and Contraindications
Irvine-Gass syndrome were randomly assigned Elevated IOP and cataract progression can be
to receive either a 0.35 mg, a 0.7 mg dexametha- seen in eyes treated with dexamethasone implants
sone implant or to an observation group who but are significantly less frequent when compared
received no treatment nor sham procedures. At to the rate seen with IVTA injections and fluo-
3 months, there was a significant improvement in cinolone (Retisert) implants. In the HURON trial,
86 L. Talat et al.

Fig. 7.1 Dexamethasone implant for cystoid macular intermediate uveitis that was unresponsive to systemic
edema. (a) A 22-year-old patient suffering from interme- immunosuppression and intravitreal triamcinolone. (c)
diate uveitis with cystoid macular edema (CME) result- Prior to the implant insertion, her CRT was 657 m with
ing in central retinal thickness (CRT) of 640 m and a BCVA of 6/60. (d) Four weeks following implantation,
best corrected visual acuity (BCVA) of 6/24. (b) One the CME was almost completely resolved with a CRT of
month following implantation, the CME resolves with 183 m. Although the foveal architecture returned to
CRT of 237 m and BCVA returned to 6/6. A 62-year- normal, visual function did not improve and remained at
old patient suffering from chronic CME secondary to 6/36

eyes treated with the 0.7 mg dexamethasone required glaucoma surgery [58]. Cataract devel-
implants experienced increased IOP requiring opment as an adverse effect of dexamethasone
topical antiglaucoma treatment in 838 % implant was reported in a prospective random-
[5456] of eyes with none requiring glaucoma ized clinical trial in 15 % of 62 phakic eyes within
surgery. A single case report of a child with inter- 6 months after implantation, with one eye (1.6 %)
mediate uveitis reported that he developed intrac- that required cataract extraction within the fol-
table glaucoma, which continued to progress low-up period [55, 56]. Dexamethasone implants
even following removal of the implant and should not be used in eyes with a compromised
7 Treatment of Uveitis with Intraocular Steroids 87

posterior lens capsule and in aphakic eyes due to from 51.4 % in the 34 weeks preceding implanta-
the high chance of implant migration into the tion to 6.1 % postimplantation. This was unlike
anterior chamber, which can result in significant the fellow non-implanted eyes which showed an
corneal decompensation requiring implant increase in the rate of relapse from 20.3 % preim-
removal [59]. plantation to 42.0 % postimplantation [61].
A randomized, phase IIb/III, multicenter trial
assessed the safety and effectiveness of Retisert
7.3.3 Fluocinolone Acetonide in treating noninfectious uveitis compared to
Intravitreal Implants standard therapy of systemic corticosteroid with
or without immunosuppressive agents. Eyes with
Fluocinolone acetonide (FAc) is one of the most Retisert experienced a lower rate of recurrence of
potent and selective glucocorticoids which does uveitis compared to those on standard therapy
not have a mineralocorticoid effect. FAc is avail- (18.2 % versus 63.5 %) [56].
able in two different intravitreal implant formula- The multicenter uveitis steroid treatment
tions, Retisert (Bausch and Lomb, Irvine, CA) (MUST) trial is an ongoing prospective study
and Iluvien (Alimera Sciences, Alpharetta, GA). setup to compare the visual acuity outcome in
patients with noninfectious intermediate uveitis,
7.3.3.1 Retisert posterior uveitis, or panuveitis randomized to
Dose and Administration receive either systemic immunosuppression or
Each Retisert implant consists of a pellet (a plas- Retisert insertion, in both eyes if necessary and
tic device with a flange with a semipermeable withdrawal of systemic medication [62]. The
membrane) containing 0.59 mg of FAc. The 2-year results demonstrated that use of the implant
implant is surgically implanted through a scle- achieved similar results to systemic treatment
rotomy incision in the pars plana where it is with regard to visual acuity but that control of the
secured in position with a suture to the sclera. uveitis was better at all time points in the Retisert-
The implant lasts on average up to 2.5 years treated patients. The longer-term outcome results
(30 months) when there is evidence of FAc deple- of the study are now awaited. Retisert has also
tion and in some cases recurrence of uveitis, been reported in retrospective studies to control
requiring implant replacement. inflammation and reduce the need for systemic
corticosteroids and immunosuppressives in
Pharmacokinetics patients with sympathetic ophthalmia [63, 64].
Retisert consists of a nondegradable polyvinyl
alcohol and silicone-encased tablet that releases Side Effects and Contraindications
FAc in low quantities over an extended period. Cataract is the most common side effect, with
FAc is initially released at a rate of 0.6 g/day, 80100 % of phakic eyes requiring cataract sur-
decreasing over the first month to a steady state gery after 2 years [62, 65]. The MUST trial group
between 0.3 and 0.4 g/day over approximately also found eyes with implants to have a fourfold
30 months [60]. risk of developing IOP elevation of 10 mmHg
[62] and significantly higher incidence of glauco-
Retisert in Uveitis matous optic neuropathy over the first 2 years
Retisert was approved by the FDA in 2005 for the compared with those assigned to systemic ther-
treatment of chronic, noninfectious posterior apy (23 % versus 6 %, respectively) [66]. Other
uveitis. In a double-masked, multicenter con- potential complications observed by 2 years post-
trolled clinical trial, 278 patients were random- implantation include hypotony (8 %), vitreous
ized to receive either the 0.59 mg or the 2.1 mg hemorrhage (15 %), retinal detachment (2 %),
FAc implant. Both implants were able to stabilize and endophthalmitis (1.3 %) [45].
or improve the visual acuity in 87 % of eyes and Fluocinolone implants are contraindicated in
were able to reduce the rate of uveitis relapse patients who have concomitant viral diseases of
88 L. Talat et al.

the cornea and conjunctiva such as epithelial her- while minimizing systemic absorption. Ocular
pes simplex, dendritic keratitis, vaccinia, vari- side effects remain a significant issue espe-
cella, mycobacterial infections of the eye, and cially with fluorinated steroids such as fluo-
ocular fungal disease [67]. cinolone or triamcinolone, and many patients
A study comparing treatment of uveitis with may require surgery for cataract and elevated
Ozurdex or Retisert implants found no superior- intraocular pressure. However, the visual out-
ity of either treatment in terms of uveitis control comes are good with good patient monitoring,
and visual acuity improvement. However, the and these intraocularly delivered drugs provide
Retisert implant had higher rates of cataract pro- an effective option for management of patients
gression compared to Ozurdex (100 % versus with sight threatening disease or in cases of
50 %), as well as more need for glaucoma medi- visual loss from the complications of uveitis.
cations, and surgery [68].

7.3.3.2 Iluvien References


Iluvien implants are approved in several European
countries for the treatment of chronic diabetic 1. Acharya NR, Tham VM, Esterberg E, et al. Incidence
macular edema (DME). Its effectiveness in man- and prevalence of uveitis: results from the pacific ocu-
lar inflammation study. JAMA Ophthalmol. 2013;131:
aging noninfectious uveitis has not been reported 140512.
to date. 2. Suhler EB, Lloyd MJ, Choi D, Rosenbaum JT, Austin
DF. Incidence and prevalence of uveitis in Veterans
Dose and Administration Affairs Medical Centers of the Pacific Northwest. Am
J Ophthalmol. 2008;146:8906.e8.
Iluvien is a non-erodible implant which contains 3. Nussenblatt RB. The natural history of uveitis. Int
FAc at a dose of 190 g and is injected into the Ophthalmol. 1990;14:3038.
vitreous using a preloaded device with a 25 gauge 4. Suttorp-Schulten MS, Rothova A. The possible
needle. impact of uveitis in blindness: a literature survey. Br J
Ophthalmol. 1996;80:8448.
5. Kaiser CJ, Ksander BR, Streilein JW. Inhibition of
Pharmacokinetics lymphocyte proliferation by aqueous humor. Reg
Following injection into the vitreous, the implant Immunol. 1989;2:429.
releases FAc at a rate of 0.2 g/day, reaching an 6. Apte RS, Niederkorn JY. Isolation and characteriza-
tion of a unique natural killer cell inhibitory factor
intravitreal steady-state concentration of 2.2 ng/ present in the anterior chamber of the eye. J Immunol.
mL, and continues to release FAc at a steady rate 1996;156:266773.
for up to 36 months post injection. 7. Dace DS, Chen PW, Niederkorn JY. CD4+ T-cell-
dependent tumour rejection in an immune-privileged
environment requires macrophages. Immunology.
Iluvien in Uveitis 2008;123:36777.
No studies have been published to date. 8. Sun D, Enzmann V, Lei S, Sun SL, Kaplan HJ, Shao
H. Retinal pigment epithelial cells activate uveito-
Conclusions genic T cells when they express high levels of MHC
class II molecules, but inhibit T cell activation when
Treatment of significant uveitis affecting the they express restricted levels. J Neuroimmunol. 2003;
posterior segment and causing visual loss has 144:18.
historically required the use of systemic drugs 9. Jiang HR, Lumsden L, Forrester JV. Macrophages and
which can have extensive side effects. In addi- dendritic cells in IRBP-induced experimental autoim-
mune uveoretinitis in B10RIII mice. Invest
tion, they are not always effective. The recent Ophthalmol Vis Sci. 1999;40:317785.
advances in local treatment provide clinicians 10. Takeuchi M, Yokoi H, Tsukahara R, Sakai J, Usui
with an increasing range of effective alterna- M. Differentiation of Th1 and Th2 cells in lymph
tives to systemic therapy. These methods nodes and spleens of mice during experimental auto-
immune uveoretinitis. Jpn J Ophthalmol. 2001;
deliver the therapeutic agent directly to the tar- 45:4639.
get area in the posterior segment of the eye, 11. Sugita S, Futagami Y, Smith SB, Naggar H, Mochizuki
providing a high intraocular concentration M. Retinal and ciliary body pigment epithelium
7 Treatment of Uveitis with Intraocular Steroids 89

suppress activation of T lymphocytes via transform- pharmacokinetics of triamcinolone acetonide after


ing growth factor beta. Exp Eye Res. 2006;83: a single intravitreal injection. Ophthalmology.
145971. 2003;110:6816.
12. Mochizuki M, Sugita S, Kamoi K. Immunological 29. Gan IM, Ugahary LC, van Dissel JT, van Meurs
homeostasis of the eye. Prog Retin Eye Res. 2013;33: JC. Effect of intravitreal dexamethasone on vitreous
1027. vancomycin concentrations in patients with suspected
13. de Smet MD, Taylor SR, Bodaghi B, et al. postoperative bacterial endophthalmitis. Graefes
Understanding uveitis: the impact of research on visual Arch Clin Exp Ophthalmol Albrecht Graefes Archiv
outcomes. Prog Retin Eye Res. 2011;30:45270. Klin Exp Ophthalmol. 2005;243:11869.
14. Chen L, Yang P, Zhou H, et al. Diminished frequency 30. Kwak HW, DAmico DJ. Evaluation of the retinal
and function of CD4+CD25high regulatory T cells toxicity and pharmacokinetics of dexamethasone after
associated with active uveitis in Vogt-Koyanagi- intravitreal injection. Arch Ophthalmol. 1992;110:
Harada syndrome. Invest Ophthalmol Vis Sci. 2008; 25966.
49:347582. 31. Machemer R, Sugita G, Tano Y. Treatment of intra-
15. Hamzaoui K, Hamzaoui A, Houman H. CD4+CD25+ ocular proliferations with intravitreal steroids. Trans
regulatory T cells in patients with Behcets disease. Am Ophthalmol Soc. 1979;77:17180.
Clin Exp Rheumatol. 2006;24:S718. 32. Yilmaz T, Weaver CD, Gallagher MJ, et al. Intravitreal
16. Chi W, Zhu X, Yang P, et al. Upregulated IL-23 and triamcinolone acetonide injection for treatment of
IL-17 in Behcet patients with active uveitis. Invest refractory diabetic macular edema: a systematic review.
Ophthalmol Vis Sci. 2008;49:305864. Ophthalmology. 2009;116:90211; quiz 1213.
17. Barnes PJ. Glucocorticosteroids: current and future 33. Cakir M, Dogan M, Bayraktar Z, et al. Efficacy of
directions. Br J Pharmacol. 2011;163:2943. intravitreal triamcinolone for the treatment of macular
18. Fischer S, Renz D, Schaper W, Karliczek GF. In vitro edema secondary to branch retinal vein occlusion in
effects of dexamethasone on hypoxia-induced hyper- eyes with or without grid laser photocoagulation.
permeability and expression of vascular endothelial Retina (Phila, Pa). 2008;28:46572.
growth factor. Eur J Pharmacol. 2001;411:23143. 34. Conti SM, Kertes PJ. The use of intravitreal cortico-
19. Tomkins-Netzer O, Taylor SR, Lightman S. steroids, evidence-based and otherwise. Curr Opin
Corticosteroid-sparing agents: new treatment options. Ophthalmol. 2006;17:23544.
Dev Ophthalmol. 2012;51:4756. 35. Das-Bhaumik RG, Jones NP. Low-dose intraocular
20. Friedlaender MH. Corticosteroid therapy of ocular triamcinolone injection for intractable macular
inflammation. Int Ophthalmol Clin. 1983;23:17582. oedema and inflammation in patients with uveitis.
21. Mondino BJ, Farley MK, Aizuss DH. Sectorial corneal Eye (London, England). 2006;20:9347.
infiltrates and pannus in herpes zoster ophthalmicus. 36. Park UC, Park JH, Yu HG. Long-term outcome of
Graefes Arch Clin Exp Ophthalmol Albrecht Graefes intravitreal triamcinolone acetonide injection for the
Archiv Klin Exp Ophthalmol. 1986;224:3136. treatment of uveitis attacks in Behet disease. Ocul
22. Sallam A, Taylor SR, Lightman S. Review and update Immunol Inflamm. 2014;22:2733.
of intraocular therapy in noninfectious uveitis. Curr 37. Moreker MR, Lodhi SA, Pathengay A. Role of intravit-
Opin Ophthalmol. 2011;22:51722. real triamcinolone as an adjuvant in the management of
23. Ferrante P, Ramsey A, Bunce C, Lightman S. Clinical Vogt-Koyanagi-Harada disease. Indian J Ophthalmol.
trial to compare efficacy and side-effects of injection 2007;55:47980.
of posterior sub-tenon triamcinolone versus orbital 38. Wadhwa N, Garg SP, Mehrotra A. Prospective evalua-
floor methylprednisolone in the management of pos- tion of intravitreal triamcinolone acetonide in serpigi-
terior uveitis. Clin Exp Ophthalmol. 2004;32:5638. nous choroiditis. Ophthalmol J Int Ophtalmol Int J
24. Kok H, Lau C, Maycock N, McCluskey P, Lightman Ophthalmol Z Augenheilkd. 2010;224:1837.
S. Outcome of intravitreal triamcinolone in uveitis. 39. Moschos MM, Gatzioufas Z, Margetis I. Intravitreal
Ophthalmology. 2005;112:1916 e17. triamcinolone acetonide for macular edema in HLA-
25. Roesel M, Gutfleisch M, Heinz C, Heimes B, Zurek- B27 negative ankylosing spondylitis. Case Rep
Imhoff B, Heiligenhaus A. Intravitreal and orbital Ophthalmol. 2010;1:1059.
floor triamcinolone acetonide injections in noninfec- 40. Young S, Larkin G, Branley M, Lightman S. Safety
tious uveitis: a comparative study. Ophthalmic Res. and efficacy of intravitreal triamcinolone for cystoid
2009;42:816. macular oedema in uveitis. Clin Exp Ophthalmol.
26. Inoue M, Takeda K, Morita K, Yamada M, Tanigawara 2001;29:26.
Y, Oguchi Y. Vitreous concentrations of triamcinolone 41. Sallam A, Comer RM, Chang JH, et al. Short-term
acetonide in human eyes after intravitreal or subtenon safety and efficacy of intravitreal triamcinolone ace-
injection. Am J Ophthalmol. 2004;138:10468. tonide for uveitic macular edema in children. Arch
27. Sivaprasad S, McCluskey P, Lightman S. Intravitreal Ophthalmol. 2008;126:2005.
steroids in the management of macular oedema. Acta 42. Lasave AF, Zeballos DG, El-Haig WM, Daz-Llopis
Ophthalmol Scand. 2006;84:72233. M, Salom D, Arevalo JF. Short-term results of a single
28. Beer PM, Bakri SJ, Singh RJ, Liu W, Peters 3rd intravitreal bevacizumab (avastin) injection versus a
GB, Miller M. Intraocular concentration and single intravitreal triamcinolone acetonide (kenacort)
90 L. Talat et al.

injection for the management of refractory 57. Taylor SR, Tomkins-Netzer O, Joshi L, Morarji J,
noninfectious uveitic cystoid macular edema. Ocul McLoone E, Lightman S. Dexamethasone implant in
Immunol Inflamm. 2009;17:42330. pediatric uveitis. Ophthalmology. 2012;119:2412e2.
43. Soheilian M, Rabbanikhah Z, Ramezani A, Kiavash 58. Kumari N, Parchand S, Kaushik S, Singh R. Intractable
V, Yaseri M, Peyman GA. Intravitreal bevacizumab glaucoma necessitating dexamethasone implant
versus triamcinolone acetonide for refractory uveitic (ozurdex) removal and glaucoma surgery in a child
cystoid macular edema: a randomized pilot study. with uveitis. BMJ Case Rep. 2013;Epub.
J Ocul Pharmacol Ther Off J Assoc Ocul Pharmacol 59. Bansal R, Bansal P, Kulkarni P, Gupta V, Sharma A,
Ther. 2010;26:199206. Gupta A. Wandering ozurdex implant. J Ophthal
44. Kosobucki BR, Freeman WR, Cheng L. Photographic Inflamm Infect. 2011;2:15.
estimation of the duration of high dose intravitreal tri- 60. Driot JY, Novack GD, Rittenhouse KD, Milazzo C,
amcinolone in the vitrectomised eye. Br J Ophthalmol. Pearson PA. Ocular pharmacokinetics of fluocino-
2006;90:7058. lone acetonide after retisert intravitreal implantation
45. Gillies MC, Simpson JM, Billson FA, et al. Safety of in rabbits over a 1-year period. J Ocul Pharmacol
an intravitreal injection of triamcinolone: results from Ther Off J Assoc Ocul Pharmacol Ther. 2004;20:
a randomized clinical trial. Arch Ophthalmol. 26975.
2004;122:33640. 61. Jaffe GJ, Martin D, Callanan D, Pearson PA, Levy B,
46. Sallam A, Taylor SR, Habot-Wilner Z, et al. Repeat Comstock T. Fluocinolone acetonide implant (retisert)
intravitreal triamcinolone acetonide injections in uve- for noninfectious posterior uveitis: thirty-four-week
itic macular oedema. Acta Ophthalmol (Copenh). results of a multicenter randomized clinical study.
2012;90:e3235. Ophthalmology. 2006;113:10207.
47. Lorenzo Carrero J, Gonzlez Barcia M, Prez Flores 62. Kempen JH, Altaweel MM, Holbrook JT, et al.
I. Sterile endophthalmitis after benzyl alcohol filtered Randomized comparison of systemic anti-
triamcinolone acetonide injection. Arch Ophthalmol. inflammatory therapy versus fluocinolone acetonide
2008;126:1423. implant for intermediate, posterior, and panuveitis:
48. Jager RD, Aiello LP, Patel SC, Cunningham Jr the multicenter uveitis steroid treatment trial.
ET. Risks of intravitreous injection: a comprehensive Ophthalmology. 2011;118:191626.
review. Retina (Phila, Pa). 2004;24:67698. 63. Rush RB, Goldstein DA, Callanan DG, Meghpara B,
49. Cheung CSY, Wong AWT, Lui A, Kertes PJ, Devenyi Feuer WJ, Davis JL. Outcomes of birdshot chorioreti-
RG, Lam W-C. Incidence of endophthalmitis and use nopathy treated with an intravitreal sustained-release
of antibiotic prophylaxis after intravitreal injections. fluocinolone acetonide-containing device. Am J
Ophthalmology. 2012;119:160914. Ophthalmol. 2011;151:6306.
50. Takakura A, Tessler HH, Goldstein DA, et al. Viral 64. Mahajan VB, Gehrs KM, Goldstein DA, Fischer DH,
retinitis following Intraocular or periocular cortico- Lopez JS, Folk JC. Management of sympathetic oph-
steroid administration: a case series and comprehen- thalmia with the fluocinolone acetonide implant.
sive review of the literature. Ocul Immunol Inflamm. Ophthalmology. 2009;116:5527.e1.
2014;22:17582. 65. Pavesio C, Zierhut M, Bairi K, Comstock TL, Usner
51. Rush R, Sheth S. Fulminant toxoplasmic retinocho- DW. Evaluation of an intravitreal fluocinolone aceton-
roiditis following intravitreal triamcinolone adminis- ide implant versus standard systemic therapy in non-
tration. Indian J Ophthalmol. 2012;60:1413. infectious posterior uveitis. Ophthalmology. 2010;
52. Chang-Lin J-E, Attar M, Acheampong AA, et al. 117:56775, 75.e1.
Pharmacokinetics and pharmacodynamics of a 66. Friedman DS, Holbrook JT, Ansari H, et al. Risk of
sustained-release dexamethasone intravitreal implant. elevated intraocular pressure and glaucoma in
Invest Ophthalmol Vis Sci. 2011;52:806. patients with uveitis: results of the multicenter uveitis
53. London NJ, Chiang A, Haller JA. The dexamethasone steroid treatment trial. Ophthalmology. 2013;120:
drug delivery system: indications and evidence. Adv 15719.
Ther. 2011;28:35166. 67. Inc. BL. Retisert (fluocinolone acetonide intravitreal
54. Williams GA, Haller JA, Kuppermann BD, et al. implant) 0.59 mg for intravitreal use: US prescribing
Dexamethasone posterior-segment drug delivery sys- information. 2011. Bausch & Lomb Incorporated
tem in the treatment of macular edema resulting from Rochester, NY.
uveitis or Irvine-Gass syndrome. Am J Ophthalmol. 68. Arcinue CA, Ceron OM, Foster CS. A comparison
2009;147:104854, 54.e12. between the fluocinolone acetonide (retisert) and
55. Lowder C, Belfort Jr R, Lightman S, et al. dexamethasone (ozurdex) intravitreal implants in uve-
Dexamethasone intravitreal implant for noninfectious itis. J Ocul Pharmacol Ther Off J Assoc Ocul
intermediate or posterior uveitis. Arch Ophthalmol. Pharmacol Ther. 2013;29:5017.
2011;129:54553. 69. Vasconcelos-Santos DV, Nehemy PG, Schachat AP,
56. Tomkins-Netzer O, Taylor SRJ, Bar A, et al. Treatment Nehemy MB. Secondary ocular hypertension after
with repeat dexamethasone implants results in long- intravitreal injection of 4 mg of triamcinolone aceton-
term disease control in eyes with noninfectious uve- ide: incidence and risk factors. Retina (Phila, Pa).
itis. Ophthalmology. 2014;121:164954. 2008;28:57380.
Steroids in a Combination
Strategy 8
Paolo Lanzetta, Daniele Veritti, and Valentina Sarao

Contents 8.6 Steroids and Pars Plana Vitrectomy. . . . . 100


8.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . 91 8.6.1 Diabetic Macular Edema . . . . . . . . . . . . . . . 100
8.6.2 Macular Edema Secondary to Retinal
8.2 Rationale . . . . . . . . . . . . . . . . . . . . . . . . . . . 92 Vein Occlusion . . . . . . . . . . . . . . . . . . . . . . . 100
8.2.1 Neovascular AMD . . . . . . . . . . . . . . . . . . . . 92 8.6.3 Other Indications . . . . . . . . . . . . . . . . . . . . . 100
8.2.2 Macular Edema Secondary to Diabetes
and Retinal Vein Occlusion . . . . . . . . . . . . . 92 Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100

8.3 Steroids and Laser Photocoagulation. . . . 92 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100


8.3.1 Diabetic Macular Edema . . . . . . . . . . . . . . . 92
8.3.2 Macular Edema Secondary to Retinal Vein
Occlusion . . . . . . . . . . . . . . . . . . . . . . . . . . . 94
8.4 Steroids and Photodynamic Therapy
with Verteporfin . . . . . . . . . . . . . . . . . . . . . 95
8.4.1 Neovascular AMD . . . . . . . . . . . . . . . . . . . . 95
8.4.2 Other Indications . . . . . . . . . . . . . . . . . . . . . 96
8.5 Steroids and Anti-VEGF . . . . . . . . . . . . . . 96
8.5.1 Diabetic Macular Edema . . . . . . . . . . . . . . . 96 8.1 Introduction
8.5.2 Macular Edema Secondary to Retinal
Vein Occlusion . . . . . . . . . . . . . . . . . . . . . . . 96
8.5.3 Neovascular AMD . . . . . . . . . . . . . . . . . . . . 97 Todays retinal pharmacotherapy is the culmina-
8.5.4 Other Indications . . . . . . . . . . . . . . . . . . . . . 99 tion of years of research in the fields of biology,
biotechnology, and pharmacology. Furthermore,
the advancements in the understanding of the
pathogenesis of posterior-segment diseases
allowed targeting important key points in the
P. Lanzetta, MD (*) D. Veritti, MD
pathogenic cascade more precisely. A revolution
Department of Medical and Biological
Sciences Ophthalmology, University of Udine, in retinal pharmacotherapy has started with the
P.le S.M. della Misericordia, 33100 Udine, Italy introduction of vascular endothelial growth fac-
Istituto Europeo di Microchirugia Oculare IEMO, tor (VEGF) inhibitors. Since then, anti-VEGF
Via M.A. Fiducio, 8, 33100 Udine, Italy therapy has become a paradigm in the treatment
e-mail: paolo.lanzetta@iemo.eu; of neovascular age-related macular degeneration
verittidaniele@gmail.com
(AMD), diabetic macular edema (DME), and
V. Sarao, MD macular edema secondary to retinal vein occlu-
Department of Medical and Biological
sion (RVO).
Sciences Ophthalmology, University of Udine,
P.le S.M. della Misericordia, 33100 Udine, Italy However, while intravitreal anti-VEGF
e-mail: sarao.valentina@gmail.com monotherapy offers unprecedented benefit with

A.J. Augustin (ed.), Intravitreal Steroids, 91


DOI 10.1007/978-3-319-14487-0_8, Springer International Publishing Switzerland 2015
92 P. Lanzetta et al.

regard to visual outcome, it often requires mul- 8.2.2 Macular Edema Secondary
tiple treatments, and a significant proportion of to Diabetes and Retinal Vein
patients do not respond to antiangiogenic treat- Occlusion
ment alone or develop tolerance during the
follow-up. Intraretinal accumulation of fluid is a hallmark of
The use of intravitreal steroids in association DME and macular edema secondary to RVO. It is
with other agents allows broadening the thera- usually accompanied by a blood-retinal barrier
pys spectrum of activity addressing multiple tar- (BRB) dysfunction. Steroids stabilize BRB, reduce
gets in the complex pathogenic pathway of exudation, and downregulate inflammatory stimuli.
retinal diseases. This chapter reviews the ratio- Steroids are believed to act by the induction of pro-
nale for combining intravitreal steroids to other teins called lipocortins. These proteins reduce leu-
interventions and presents a synthesis of evi- kocyte chemotaxis, control biosynthesis, and inhibit
dence available. the release of arachidonic acid from the phospho-
lipid membrane. Moreover, corticosteroids have
been shown to control gene expression of key medi-
8.2 Rationale ators. Steroids influence the expression of VEGF,
inhibit proinflammatory genes such as tumor necro-
8.2.1 Neovascular AMD sis factor-alpha (TNF-) and other inflammatory
chemokines, and induce the expression of anti-
Pathological choroidal neovascularization (CNV) inflammatory factors such as pigment-derived
due to AMD is the result of a series of subretinal growth factor (PEDF) [24]. It has been shown that,
changes in which neoangiogenesis, inflammation, besides inflammation and BRB breakdown, isch-
and cellular recruitment play a pivotal role [1]. emia is a main driver of diabetic and RVO-related
The vascular component of CNV can be macular edema. Therefore, combination therapy
advantageously targeted with anti-VEGF and with corticosteroids and anti-VEGF agents can be
anti-platelet-derived growth factor (PDGF) used to target both the inflammatory and ischemic
drugs. But, as in most biologic systems, inhi- drivers of macular edema (via vigorous VEGF inhi-
bition of one cascade upregulates alternate bition). Combining steroids with laser photocoagu-
pathways. Therefore, when treating neovascu- lation and/or surgical removal of epiretinal
lar AMD with antiangiogenic agents alone, membrane allows a better oxygenation of the retinal
there is a theoretical risk of compensatory tissue and reduce hydrostatic pressure on capillaries
upregulation of VEGF receptors and/or VEGF and venules. From a pharmacodynamic standpoint,
production. associating steroids with others agents has a strong
Corticosteroids act at numerous key points rationale to further widen the spectrum of action via
of neoangiogenesis by altering cell behavior a synergistic treatment effect. From a pharmacoki-
and cytokine expression and have been shown netic point of view, combining agents with different
to be effective in reducing inflammation and intravitreal half-lives may allow a rapid treatment
cellular recruitment, addressing the most effect associated with prolonged duration of action
important pathogenic key points in the nonvas- and reduced need of re-treatments.
cular component of CNV development.
Therefore, combination approaches can lessen
the chance of compensatory rebound of non- 8.3 Steroids and Laser
inhibited pathological pathways when directing Photocoagulation
therapy to a single target. Additionally, com-
bined approaches may not only increase overall 8.3.1 Diabetic Macular Edema
efficacy but may also reduce the potential
adverse effects by allowing fewer re-treatments Combination therapy with intravitreal triamcino-
needed. lone acetonide (IVTA) and laser photocoagulation
8 Steroids in a Combination Strategy 93

has been widely studied in many short- and long- lines occurred in 18, 29, 28, and 22 % of eyes,
term clinical trials [511]. Several reports have respectively. Compared with the sham + prompt
demonstrated that IVTA combined with macular laser group, central retinal thickness decreased on
laser treatment produced better morphological average by 31 m in the ranibizumab + prompt
and functional outcomes compared with laser laser group (p = 0.03), 28 m in the ranibizumab +
treatment alone up to 6 months of follow-up deferred laser group (p = 0.01), and 10 m in the
[57]. The first significant trial comparing laser TA + prompt laser group (p = 0.37). These results
and IVTA was reported by Avitabile et al. [8] in showed that patients treated with TA + prompt
which patients were randomized to receive IVTA laser did not experience a significant gain in
alone or combined with grid laser at 3 months or visual acuity compared to the other treatment
grid laser alone in the treatment of cystoid macu- groups at 2 years of follow-up. However, in a sub-
lar edema. At 45 days after treatment, the eyes of group analysis, among individuals who were
the two groups receiving triamcinolone had better pseudophakic at the baseline, TA + prompt laser
visual acuity and lower central macular thickness group reported an improvement in visual acuity
than those receiving photocoagulation. However, of more than 1.6 letters compared with the sham
the triamcinolone effect regressed 6 months after + prompt laser group. For eyes that were pseudo-
injection. Lam et al. [9] reported a comparative phakic at baseline in the TA plus prompt laser
randomized controlled trial. Patients with DME group, improvement in mean visual acuity was
were randomized to grid laser photocoagulation essentially the same as the groups treated with
(37 eyes), 4 mg of IVTA (38 eyes), or 4 mg of ranibizumab (+0.5 letters in ranibizumab +
IVTA combined with sequential grid laser about prompt laser group and +3.5 letters in the ranibi-
1 month later (36 eyes). After treatment, signifi- zumab + deferred laser group), suggesting that
cant central foveal thickness reductions were noted the inferior result in eyes treated with intravitreal
in both the IVTA and combined groups at all fol- TA was associated with the development of cata-
low-up visits (p = 0.01) but not in the laser group. ract. Cataract surgery was performed in the 12 %
The standardized reduction in macular thickening of phakic eyes in the sham + prompt laser and in
at 17 weeks was significantly greater in the com- the ranibizumab + prompt laser groups, in the
bined group versus the IVTA group (p = 0.007), 13 % of phakic eyes in the ranibizumab + deferred
suggesting that combined treatment might prolong laser group, and in the 55 % of patients treated
the effects of IVTA. The Diabetic Retinopathy with TA + laser. An IOP-lowering medication
Clinical Research Network has investigated the was required in the 5 % of eyes in the sham +
efficacy of intravitreal TA associated with laser prompt laser and ranibizumab + prompt laser
photocoagulation in eyes with DME involving the groups, in the 3 % of eyes in the ranibizumab +
central macula after 2 years of follow-up [11]. deferred laser group, and in the 28 % of patients
Patients were randomly assigned to one of four of the TA + laser group [11, 12]. Other studies
groups: sham injection plus prompt laser, ranibi- have demonstrated promising results of combina-
zumab 0.5 mg plus prompt laser, ranibizumab tion therapy with intravitreal injection of TA and
0.5 mg plus deferred laser, and intravitreal TA laser photocoagulation for the treatment of prolif-
4 mg plus prompt laser. At 2 years, compared with erative diabetic retinopathy (PDR) with clinically
the sham + prompt laser group, mean change in significant macular edema (CSME) [1317]. In a
visual acuity from baseline was 3.7 letters greater 12-month randomized clinical trial conducted by
in ranibizumab + prompt laser group (p = 0.03), Maia et al. [18], 44 eyes with PDR and CSME
5.8 letters greater in the ranibizumab + deferred were randomized to receive treatment with com-
laser group (p < 0.01), and 1.5 letters worse in the bined 40 mg of intravitreal triamcinolone aceton-
TA + prompt laser group (p = 0.35). The propor- ide and laser photocoagulation (n = 22) or laser
tion of patients that lost at least 15 letters was 10, photocoagulation alone (n = 22). On average,
4, 2, and 13 % of eyes, respectively, and an best correct visual acuity (BCVA) improved sig-
improvement in visual acuity by three or more nificantly (p < 0.001) in the TA and laser group
94 P. Lanzetta et al.

compared with the laser-alone group at all study 8.3.2 Macular Edema Secondary
follow-up visits. An improvement of two or more to Retinal Vein Occlusion
ETDRS lines was observed in 63.1 and 10.5 % of
eyes, respectively (p < 0.001). A significant Combining macular laser photocoagulation with
decrease in mean central macular thickness corticosteroid may increase the efficacy of treat-
occurred in the TA and laser group when com- ment for RVO-related macular edema in patients
pared with the laser-alone group at all study fol- who are not adequately responsive to laser
low-up intervals (p < 0.001). At 12 months, mean monotherapy. A combination approach of ste-
(SD) reductions in central macular thickness roid therapy with laser photocoagulation may
were 123 68 m and 65 51 m, respectively result in synergistic effects as it targets both the
(p < 0.001). Despite the beneficial effect on visual inflammatory and VEGF-related component at
acuity and macular edema, especially during the multiple different points along the pathway.
first months after initial treatment, IVTA use is Unfortunately, only a small number of studies
greatly limited by significant side effects, espe- exploring RVO combination therapies have been
cially due to frequent re-treatment need [1921]. published to date. Most are short term and retro-
Therefore, the recent availability of an intravitreal spective and/or involve small numbers of
sustained-release corticosteroid device that would patients [2325]. A case series has demonstrated
be equally effective but safer was fully justified. that TA combined with laser photocoagulation is
The results of combining dexamethasone implant an effective treatment in eyes affected by persis-
plus laser were recently examined in the PLACID tent macular edema after BRVO [25]. After
trial [22]. The purpose of this multicenter, ran- 6 months of follow-up, the combination therapy
domized study was to assess the efficacy and resulted in a significant reduction of central
safety of 0.7 mg dexamethasone intravitreal foveal thickness (p = 0.001) and improvement of
implant associated with laser photocoagulation visual acuity (p = 0.016) after a mean of 1.13
compared with laser alone for treatment of diffuse IVTA injections. It is known that grid photoco-
DME. Patients were randomized to treatment agulation has potential side effects, including
with dexamethasone implant at baseline plus laser laser scar expansion, paracentral scotoma, ele-
(n = 126) or sham implant at baseline and laser vation of central visual field thresholds, second-
(n = 127). In this 1-year trial, patients in the dexa- ary choroidal neovascularization, subretinal
methasone group were eligible for a second fibrosis, and atrophy of the outer retinal layers.
implant at 6 months and a third at 9 months if they In order to reduce the potential side effects due
met re-treatment criteria. More patients with the to laser therapy, a prospective randomized clini-
dexamethasone implant plus laser achieved a two- cal trial has published the beneficial effects of
line gain in visual acuity than with laser alone at subthreshold grid laser photocoagulation
month 1 (p < 0.001) and month 9 (p = 0.007). After (SGLT) in combination with intravitreal TA in
9 months of treatment, this difference was no lon- eyes affected by macular edema secondary to
ger statistically significant. The mean improve- BRVO [26]. Twenty-four patients were random-
ment in BCVA was significantly greater in ized to the SGLT alone (13 eyes) or in combina-
patients with diffuse DME who received the tion with IVTA (11 eyes). At 12 months, the
implant plus laser than sham plus laser (up to 7.9 mean number of lines gained was 3.4 in the
vs. 2.3 letters) at all time points through month 9 SGLT-TA group and 1.3 in the SGLT group. An
(p = 0.013). Areas of leakage and retinal thickness improvement in visual acuity by two lines
decreased more in patients treating with dexa- occurred in 91 and 62 %, respectively. The
methasone and laser than those receiving laser authors reported a more rapid reduction in the
alone. A proportion of 1.0 % of patients at month macular edema in the combined group in com-
12 experienced elevation of IOP by 10 mg Hg or parison with the SGLT group, which was main-
more and no patient required surgery for IOP tained over the follow-up. The recent availability
management. of implants with corticosteroid has allowed
8 Steroids in a Combination Strategy 95

alternative approaches to treating macular patients with CNV who underwent combined
edema secondary to BRVO with combined ther- therapy with PDT and an injection of 4 mg of
apy [2729]. The intent of a 6-month trial was TA. On average, an improvement in visual acu-
to determine whether a sustained-release corti- ity of 2.5 lines with a mean 1.24 combined treat-
costeroid injection combined with macular grid ments was registered. Significant complications
pattern laser could offer an additional benefit by can occur with this therapeutic combination,
reducing the number of intravitreal implants including cataract (in up to 57 % of patients),
while maintaining the same visual acuity gain glaucoma (in up to 40 % of patients, sometimes
and macular fluid reduction registered in patients requiring surgical intervention), and endophthal-
treated with as-needed dexamethasone [29]. In mitis (<1 % of patients) [32]. The efficacy and
this study, 50 patients with perfused macular safety of PDT-V with IVTA in occult CNV were
edema secondary to BRVO received an intravit- investigated in a prospective and uncontrolled
real dexamethasone implant at the baseline and study [33, 34]. A total of 41 patients were
were then randomized to dexamethasone implant included and followed up for 2 years. The mean
re-treatment on an as-needed basis (group 1, number of treatments needed was 1.8. Visual
n = 25 eyes) or macular grid photocoagulation acuity improved gradually in most patients, with
68 weeks after baseline followed by re-treat- mean values of 20/133, 20/84, and 20/81 at
ment with pro re nata dexamethasone implant baseline 12 and 24 months after treatment,
(group 2, n = 25 eyes). After 6 months of follow- respectively. A total of 11 of the 41 treated study
up, mean BCVA improvement was 0.23 log- eyes (26.8 %) underwent cataract surgery
MAR in group 1 and 0.15 logMAR in group 2. between 6 and 15 months after the first treat-
Central macular thickness decreased on average ment. Nine patients required local or systemic
by 33.4 m in group 1 and 21.4 m in group 2 at glaucoma therapy owing to a transient steroid-
the end of follow-up. Re-treatment with dexa- induced IOP increase. A randomized, masked,
methasone implant was given in 12 % eyes in controlled clinical trial of PDT-V and anecor-
group 1 and in 0 % in group 2 at the end of tave acetate versus PDT and sham in 136 patients
follow-up. with CNV due to AMD showed some visual
acuity benefit for the combination therapy over
the PDT and sham group (78 vs. 67 % of patients
8.4 Steroids and Photodynamic had stable visual acuity), but the effect was not
Therapy with Verteporn statistically significant [35]. Chaudhary et al.
[36] reported a prospective, randomized pilot
8.4.1 Neovascular AMD study that demonstrated effective stabilization
of visual acuity and reduced treatment frequency
A large number of publications confirmed the at 12 months with a combination of PDT-V and
positive synergic role of combining TA and PDT IVTA therapy versus PDT-V alone. A total of 30
therapy for the treatment of all types of CNV: eyes with occult or minimally classic CNV due
classic or predominantly classic, occult or mini- to AMD were randomized to receive standard
mally classic, and RAP (retinal angiomatous PDT-V alone or combined PDT-V plus 12 mg
proliferation) lesions [3039]. Several uncon- IVTA. The treatment rate was 1.13 in the PDT-V
trolled studies indicate that combined PDT-V plus IVTA group and 3.6 in the PDT-V-alone
and IVTA injection is more effective than either group. Mean ETDRS scores for the PDT-V-
treatment alone [30, 31]. Generally, these stud- alone group at 3, 6, 9, and 12 months decreased
ies have shown that combination therapy might statistically from baseline by 5.7, 8.1, 9.1, and
play a role in reducing the frequency of PDT-V 13.3 letters, respectively. However, in the PDT-V
sessions and improving visual outcome com- plus IVTA group, the ETDRS scores did not
pared with PDT alone. Spaide et al. [31] origi- decline significantly at any follow-up visit. The
nally reported a 12-month follow-up of 26 mean decline in ETDRS scores at 3, 6, 9, and
96 P. Lanzetta et al.

12 months was 1.7, 3.7, 5.3, and 1.9 letters, 8.5 Steroids and Anti-VEGF
respectively. In the combined PDT plus IVTA
group, 60 % of patients achieved either stable or 8.5.1 Diabetic Macular Edema
improved vision compared with 33.3 % of cases
in the control group. A total of 13 % of patients Various studies have reported the benefit of intra-
in the combined therapy and 53.3 % in the con- vitreal bevacizumab combined with corticoste-
trol group experienced moderate or severe vision roids for the treatment of DME [4348]. A
loss. However, in some clinical trials, patients recently published systematic review and meta-
receiving IVTA and PDT-V did not show an analysis was conducted to evaluate the short-term
improvement in visual acuity, as compared to effect of adding intravitreal bevacizumab to TA at
the PDT-V-only treatment group [3739]. A 6, 12, and 24 weeks [48]. The authors have sug-
2-year, prospective, randomized study con- gested that combination therapy did not result in
ducted on 84 patients affected by new CNV sec- any significant reduction in central thickness or
ondary to AMD has confirmed that combination gain in vision compared to treatment with bevaci-
of TA with PDT significantly reduced the num- zumab alone at any point in time. However,
ber of PDT-V re-treatments needed to obtain patients who experienced a significant gain in
permanent inactivation of CNV [39]. However, vision after treatment with bevacizumab alone
analysis of long- term visual outcomes revealed did not maintain it through the following months,
that after an initial significant benefit observed showing a decrease of its beneficial effect up to
in the first year, the combination of PDT with 24 months of follow-up. The authors suggest that
TA is associated with progressive visual decline the use of IVTA should be guarded against
during the second year of follow-up. At the because of cumulative side effects such as cata-
24-month point, the combined group showed a ract and glaucoma and potential toxicity of the
significant visual acuity worsening, reaching drug itself.
values statistically similar to the PDT-only
group (p = 0.74). A reduction of visual function
may be explained by the increased risk of devel- 8.5.2 Macular Edema Secondary
oping macular atrophy after the combination to Retinal Vein Occlusion
therapy with intravitreal TA and PDT-V [39].
In many studies, combination strategies with
anti-VEGF drugs have shown beneficial effects
8.4.2 Other Indications in treating macular edema due to retinal vein
occlusion [49, 50]. Ehrlich et al. [50] reported
As reported in previous studies, IVTA reduces the 6-month results of combined treatment
the frequency of PDT-V re-treatment that could using intravitreal bevacizumab and IVTA for
result from combining the short-term effect of the treatment of macular edema secondary to
PDT-induced CNV closure with antiangiogenic BRVO. At the end of the follow-up, the authors
and anti-inflammatory effects of intravitreal concluded that combination treatment showed
corticosteroids [40]. Combination of PDT and beneficial effects in improving structural out-
IVTA may reduce the need for re-treatment and comes such as central macular thickness, but
could be potentially useful for preserving vision had no advantage over results with intravitreal
in some patients with CNV due to angioid bevacizumab alone. Cekic et al. [51] compared
streaks or secondary to pathological myopia the three different intravitreal treatment modali-
[41, 42]. Possible adverse events of intrasurgical ties, including intravitreal bevacizumab, IVTA,
use of TA (such as rise in IOP, endophthalmitis, and the combination of both. They concluded
and cataract progression) have not been that all treatment groups had similar therapeutic
described. effects at 1 month, but at 6 months of follow-up,
8 Steroids in a Combination Strategy 97

patients treated with bevacizumab alone by 6.6 (1.7) letters in CRVO and 7.8 (2.9) in
achieved better functional results. Recently, Lee BRVO patients and in group 2 by 9.8 (1.0) ver-
et al. [52] compared the long-term efficacy of sus 9.4 (2.1) letters. A significant difference
intravitreal TA, bevacizumab, and drug combi- was only seen between CRVO patients in groups
nation injections with focal grid laser photoco- 1 and 2 at 12 months (p < 0.05). Recurrence after
agulation for the treatment of macular edema the first dexamethasone injection occurred after
secondary to BRVO. Patients received one of 3.8 (CRVO) and 3.5 months (BRVO) in group 1
the following intravitreal treatments: 4 mg TA versus 3.2 and 3.7 months in group 2. Elevated
monotherapy (n = 31), 1.25 mg bevacizumab IOP was measured in approximately 40 % and
monotherapy (n = 95), or a combination of 2 mg cataract progression requiring surgery in about
TA and 1.25 mg bevacizumab (n = 25). There 50 % of eyes after three dexamethasone injec-
was no statistically significant difference in tions. The authors concluded that combined treat-
change in visual acuity among the three groups ment showed slightly better functional outcome
at 1, 3, 6, or 24 months postoperatively. for CRVO patients at 1 year of follow-up.
However, at the 12-month follow-up, the IVTA Increased IOP and cataract progression were fre-
group showed less visual improvement than did quent and should be considered when an individ-
the other groups, with statistical significance ual treatment is planned.
(p = 0.01). A percentage of 16, 5.6, and 0 % of
participants in the three groups showed signifi-
cant visual loss of more than three lines of the 8.5.3 Neovascular AMD
Snellen chart at the final follow-up examina-
tion. These considerations suggested that The combination of steroids and VEGF inhibitors
although IVTA yielded outcomes similar to may overcome the shortcomings of anti-VEGF
those of standard grid photocoagulation, intra- monotherapy by addressing the multiple stimuli
vitreal bevacizumab in monotherapy or a drug that lead to angiogenesis, inflammation, and fibro-
combination resulted in significantly lower sis. Data available in literature have reported that
visual loss and could be used as a valid alterna- the combined approach allows to prolong the ben-
tive to grid laser photocoagulation. eficial effects of anti-VEGF monotherapy and to
Some studies were conducted to assess reduce the number of re-treatments. Some studies
whether dexamethasone intravitreal implant have investigated the combined use of anti-
0.7 mg with bevacizumab therapy can be syner- VEGFs with steroids in patients affected by CNV
gistic, providing further improvements in both secondary to AMD unresponsive to anti-VEGF
visual acuity and macular thickness, minimizing therapy. A retrospective 12-month case series has
the number of injections and increasing the dura- analyzed 25 eyes with CNV due to AMD who
tion of effects [5355]. In a prospective, consecu- received a combination of intravitreal bevaci-
tive, nonrandomized interventional case series zumab (1 mg) and TA (4 mg) [56]. Re-treatment
were included 64 eyes of patients with ME asso- with intravitreal bevacizumab or combination
ciated with RVO [54]. Patients were assigned to therapy was considered at investigator discretion
two groups: group 1 in which 38 patients (22 eyes at every follow-up visit. A trend toward progres-
with CRVO and 16 eyes with BRVO) were treated sive loss of BCVA, from 1.08 to 1.31 logMAR,
using a dexamethasone implant alone and group had been observed in the 6 months before starting
2 which consisted of 26 patients (14 cases with the combination therapy, in spite of regular treat-
CRVO and 12 cases with BRVO) and treated with ment with anti-VEGFs (p < 0.0001). After combi-
three consecutive intravitreal bevacizumab injec- nation therapy, a mean BCVA improvement of
tions at monthly intervals followed by a dexa- 0.18 (95 % CI: 0.05; 0.3) logMAR was reported
methasone implant 2 weeks later. In case of between baseline and the 12-month evaluation
recurrence, both cohorts received further dexa- (p < 0.01). OCT measurements showed a 139 mm
methasone implants. In group 1, BCVA improved (95 % CI: 76; 203) decrease in mean central
98 P. Lanzetta et al.

retinal thickness (p < 0.01). On average, patients (6.8 %) received the maximum of six possible
required 1.8 additional treatments. Five (20 %) ranibizumab injections, and 16 (36.4 %) received
cases of IOP elevation were successfully treated five injections. Almost half of the patients (45.5 %)
with topical medications. In this study, the com- required three or fewer injections of ranibizumab
bined approach seems to produce an inversion of over the study period to achieve these results, and
the tendency to visual loss in anti-VEGF nonre- 20.4 % needed one injection or fewer. This open-
sponders, and, in addition, it suggests a reduced label study provides the first demonstration of a
need for re-treatment compared to those required beneficial effect of the combination dexametha-
by anti-VEGF treatment alone. El-Matri and sone implant and ranibizumab in patients with
coauthors [57] published the outcomes of com- untreated neovascular AMD.
bined IVTA injection with intravitreal bevaci- The benefit of adding intravitreal dexametha-
zumab in treating CNV associated with large sone to the treatment of neovascular AMD with
retinal pigment epithelial detachment (PED) in anti-VEGF therapy was examined in the LuceDex
seven eyes of five patients with AMD [53]. study [60]. This is a pilot randomized prospective
Triamcinolone and bevacizumab were adminis- clinical trial in which 37 eyes were randomly
tered at 1-week interval. The combined treatment, assigned to receive combination therapy with intra-
maintaining the same time interval between the vitreal ranibizumab and dexamethasone implant
two agents, was repeated in four eyes, and the (group 1) or intravitreal ranibizumab monotherapy
number of intravitreal bevacizumab injection, (group 2). Visual acuity improvement was on aver-
after combined therapy, ranged from 1 to 3. At the age 11.1 and 5.9 ETDRS letters in groups 1 and 2,
end of the follow-up (914 months), FA showed respectively, at month 12. A gain of at least 15 let-
reduced or no leakage in all eyes, reduced foveal ters was achieved by six patients (35 %) in group 1
thickness, and PED in all eyes. and four patients (20 %) in group 2 (p = 0.50). The
A randomized controlled study evaluating the average number of treatments by month 12 was
use of the dexamethasone implant in combination 7.1 in group 1 and 6.6 in group 2. Choroidal neo-
with ranibizumab in patients with treatment- vascular membrane size decreased in group 1 sig-
resistant neovascular AMD found that the implant nificantly compared with group 2 (p < 0.05).
significantly delayed or reduced the need for Other studies have reported the beneficial effect
repeated ranibizumab injection. The treatment of a triple therapy strategy in which the adminis-
also demonstrated an acceptable safety profile tration of anti-VEGF drugs was associated with
[58]. The results of a 26-week multicenter open- PDT-V and steroids. Augustin et al. [61] investi-
label trial, performed to evaluate the dexametha- gated the efficacy and safety of triple therapy with
sone implant 0.7 mg as adjunctive therapy to PDT-V (42 J/cm2), intravitreal dexamethasone
intravitreal ranibizumab in treatment-nave sub- (800 g), and intravitreal bevacizumab (1.5 mg).
jects with subfoveal CNV secondary to AMD, One hundred and four eyes were included in the
were recently reported [59]. The use of dexameth- study; the baseline mean VA was 0.8 logMAR. A
asone implant alone resulted in statistically signifi- mean increase in visual acuity of 1.8 lines was
cant improvements in CRT from baseline as early reported at a mean follow-up of 40 weeks. Eighteen
as week 1 and continued through week 4 (p < 0.01). eyes required an additional intravitreal bevaci-
In addition, clinically significant improvements in zumab injection and five eyes necessitated a sec-
BCVA and fluorescein leakage were seen with the ond cycle of triple therapy. Bakri et al. [62]
implant alone. With the addition of ranibizumab, reviewed retrospectively the safety and efficacy of
statistically significant improvements in CRT, same-day therapy with PDT-V (25 J/cm2), intravit-
BCVA, and FA leakage were seen (p < 0.001) at real dexamethasone (200 g), and bevacizumab
every time point through the end of the study at (1.25 mg) in 31 eyes. Visual acuity improved from
week 26. Most patients (84 %) did not need rescue 0.61 to 0.58 logMAR after a mean follow-up of
ranibizumab before 4 weeks. Over the course of 13.7 months. Re-treatment was given with a mean
26 weeks, only three of the 44 patients enrolled of 2.3 anti-VEGF injections and 0.3 repeat TT
8 Steroids in a Combination Strategy 99

treatments. Ahmadieh et al. [63] evaluated the with 8.9 for the ranibizumab monotherapy group.
functional results of triple therapy with PDT-V The number of individual treatments was 12.6 in
(50 J/cm2; 600 mW/cm2), intravitreal triamcino- the TT half-fluence group and 8.9 in the ranibi-
lone (2 mg), and intravitreal bevacizumab zumab monotherapy group. A 6-month interven-
(1.25 mg) in 17 eyes. Mean VA at baseline was tional case series has investigated the effects of
0.74 logMAR. VA improved by 0.33 logMAR adjunctive use of intravitreal ranibizumab, modi-
after 24 weeks with a mean of 1.1 additional intra- fied juxtascleral triamcinolone acetonide, and
vitreal bevacizumab injections per eye. Yip et al. PDT-V with different irradiance in 25 patients
[64] reported the results of triple therapy with affected by CNV secondary to AMD and nonre-
PDT-V (50 J/cm2; 600 mW/cm2), IVTA (4 mg), sponder to anti-VEGF monotherapy [68]. The first
and intravitreal bevacizumab (1.25 mg) in 36 eyes ten eyes (cohort 1) enrolled received triple therapy
with neovascular AMD. Mean VA at baseline was (TT) with reduced fluence/reduced irradiance
1.22 logMAR, and the mean follow-up time was PDT-V (25 J/cm2, 300 mW/cm2), cohort 2 (n = 10)
14.7 months. At the end of the follow-up, mean received TT with reduced fluence/standard irradi-
improvement in VA was 0.04 logMAR. A single ance PDT-V (25 J/cm2, 600 mW/cm2), and the last
session of triple therapy led to complete resolution ten eyes included (cohort 3) were treated with TT
of angiographic evidence of CNV in 77.8 % of with standard fluence/standard irradiance PDT-V
cases. Forte et al. [65] reviewed retrospectively the (50 J/cm2, 600 mW/cm2). At the end of the follow-
efficacy of PDT-V (50 J/cm2), intravitreal dexa- up, mean BCVA change was 0.15, 0.13, and
methasone, and ranibizumab or bevacizumab in 21 0.29 logMAR, respectively. The mean number of
eyes. They reported a significant improvement of ranibizumab reinjections was 0.7 (cohort 1), 0.8
visual acuity and foveal thickness with a mean (cohort 2), and 0.5 (cohort 3). No patient experi-
follow-up of 14 months. No adverse effects were enced a severe VA loss. In one case, a moderate
observed. Ehmann et al. [66] studied prospectively visual loss was reported among patients included
the safety and efficacy of same-day PDT-V (25 J/ in cohort 3. Among all eyes enrolled, in five
cm2) and intravitreal dexamethasone (800 g). At (17 %), topical treatment was required due to IOP
1 and 7 weeks, patients received a bevacizumab 25 mmHg. In all cases, the increase in IOP
(1.25 mg) injection. Thirty-two eyes were included resolved within 3 months, and thus the medication
and followed-up for 12 months. Visual acuity was discontinued. One of the 20 phakic eyes at
improved from 0.74 to 0.53 logMAR. In the baseline required cataract surgery. The authors
RADICAL study, 162 patients were randomized confirmed that triple therapy can potentially be a
to one of four treatment arms: double therapy with safe and effective new treatment modality for
reduced fluence PDT-V (25 J/cm2) followed by patients who fail to demonstrate an appropriate
ranibizumab, reduced-fluence PDT-V (25 J/cm2) response to anti-VEGF monotherapy. Reduced
followed by ranibizumab-dexamethasone triple fluence PDT seems to be safer than standard flu-
therapy, very low-fluence PDT-V (15 J/cm2) fol- ence PDT and to warren better functional results.
lowed by ranibizumab-dexamethasone triple ther-
apy, or ranibizumab monotherapy [67]. The
24-month results showed that mean visual acuity 8.5.4 Other Indications
change from baseline was not statistically different
among the treatment groups. Mean visual acuity in Several published works reported a beneficial
the double-therapy group decreased by two role for IVTA as an adjuvant to bevacizumab for
ETDRS letters, in the TT half-fluence group the treatment of macular edema in patients with
improved by two letters, in the TT very low- astrocytic hamartomas and tuberous sclerosis
fluence group improved by 0.3 letters, and in the [69] and Coatss disease [70] and due to radiation
monotherapy group improved by 3.8 letters. retinopathy in patients with posterior uveal mela-
Through 24 months, patients in the TT half-fluence noma [71]. Severe local or systemic side effects
group had a mean of 4.2 re-treatments compared were not observed.
100 P. Lanzetta et al.

8.6 Steroids and Pars Plana CME in patients with uveitis [84]. After
Vitrectomy 12 months, CME was improved in 44 % of patient
and worsened in 12 % of cases. A mean improve-
8.6.1 Diabetic Macular Edema ment in visual acuity was noted in 28 % of eyes
after 12 months. However, development or pro-
Triamcinolone acetonide in combination with gression of cataract and increased IOP were fre-
pars plana vitrectomy with or without laser pho- quent. Cataract progressed in 85 % of the phakic
tocoagulation has recently shown beneficial patients postoperatively. Increased IOP was
effects on both anatomical and functional out- detected in 11 % 12 months after surgery.
comes in eyes with nontractional DME refractory
to anti-VEGF therapy, proliferative diabetic reti- Conclusions
nopathy, and proliferative vitreoretinopathy [72, The aim of combining intravitreal steroids to
73]. other treatments is to take advantage of syner-
gies among the complementary means of
action of different drugs and interventions to
8.6.2 Macular Edema Secondary provide similar or better efficacy outcomes
to Retinal Vein Occlusion than approved therapies while reducing treat-
ment frequency and the proportion of nonre-
The benefit of combined vitrectomy (with or sponder patients. The results presented in this
without internal limiting membrane peeling) with chapter support the use of steroids in associa-
IVTA is unclear for the treatment of macular tion with anti-VEGF agents in the treatment of
edema secondary to RVO [7476]. Vitrectomy neovascular AMD, DME, and macular edema
with sheathotomy has been combined with IVTA, secondary to RVO. However, the level of evi-
showing favorable results of 13 Snellen lines for dence is still low, and further studies are
CRVO [76, 77] and BRVO [78, 79]. RON com- needed to better elucidate which are the most
bined with vitrectomy and IVTA has also been efficacious and safe combinations and what is
described with improved VA and decreased cen- the best treatment regimen/dosage. Moreover,
tral macular thickness [80, 81]. In the combined it is needed to clarify which patients would
treatment group, in terms of VA and macular benefit the most from a combination approach
thickness, hemodilution treatment for CRVO has rather than monotherapy.
been compared with hemodilution with IVTA
with improvement [82]. Lewis et al. [83] com-
pared maculopexy assisted by gas and/or triam- References
cinolone for BRVO with macular edema. Patients
undergoing gas-assisted treatment showed 1. Schmidt-Erfurth UM, Richard G, Augustin A, et al.
Guidance for the treatment of neovascular age-related
greater VA improvement than the IVTA group.
macular degeneration. Acta Ophthalmol Scand. 2007;
Promising results have been described for triple 85:48694.
therapy with IVTA, bevacizumab, and vitrec- 2. Kang BS, Chung EY, Yun YP, et al. Inhibitory effects
tomy. However, there is a lack of large, multi- of anti-inflammatory drugs on interleukin-6 bioactiv-
center, randomized controlled trials to support ity. Biol Pharm Bull. 2001;24(6):7013.
3. Umland SP, Nahrebne DK, Razac S, et al. The inhibi-
these results [73]. tory effects of topically active glucocorticoids on
IL-4, IL-5, and interferon- production by cultured
primary CD4+ T cells. J Allergy Clin Immunol.
8.6.3 Other Indications 1997;100(4):5119.
4. Floman N, Zor U. Mechanism of steroid action in
ocular inflammation: inhibition of prostaglandin pro-
Another study has investigated the efficacy of duction. IOVS. 1977;16(1):6973.
pars plana vitrectomy with additional intraopera- 5. Tunc M, Onder HI, Kaya M. Posterior sub-Tenons
tive intravitreal TA injection on the course of capsule triamcinolone injection combined with focal
8 Steroids in a Combination Strategy 101

laser photocoagulation for diabetic macular edema. 18. Maia OO, Takahashi BS, Costa RA. Combined laser
Ophthalmology. 2005;112:108691. and intravitreal triamcinolone for proliferative dia-
6. Se WK, Sa HS, Hee YC, et al. Macular grid photoco- betic retinopathy and macular edema: one-year results
agulation after intravitreal triamcinolone acetonide of a randomized clinical trial. Am J Ophthalmol.
for diffuse diabetic macular edema. Arch Ophthalmol. 2009;147:2917.
2006;124(5):6538. 19. Park HY, Yi K, Kim HK. Intraocular pressure eleva-
7. Kang SW, Sa HS, Cho HY, Kim JI. Macular grid pho- tion after intravitreal triamcinolone acetonide injec-
tocoagulation after intravitreal triamcinolone aceton- tion. Korean J Ophthalmol. 2005;19:1227.
ide for diffuse diabetic macular edema. Arch 20. Jonas JB, Degenring R, Vossmerbauemer U, et al.
Ophthalmol. 2006;124:6538. Frequency of cataract surgery after intravitreal injec-
8. Avitabile T, Longo A, Reibaldi A. Intravitreal triam- tion of high-dosage triamcinolone acetonide. Eur J
cinolone compared with macular laser grid photoco- Ophthalmol. 2005;15:4624.
agulation for the treatment of cystoid macular edema. 21. Lang Y, Zemel E, Miller B, et al. Retinal toxicity of
Am J Ophthalmol. 2005;140:695702. intravitreal kenalog in albino rabbits. Retina.
9. Lam DS, Chan CK, Mohamed S, et al. Intravitreal tri- 2007;27:77888.
amcinolone plus sequential grid laser versus triam- 22. Callanan DG, Gupta S, Boyer DS, Ozurdex PLACID
cinolone or laser alone for treating diabetic macular Study Group, et al. Dexamethasone intravitreal
edema: six-month outcomes. Ophthalmology. 2007; implant in combination with laser photocoagulation
114:21627. for the treatment of diffuse diabetic macular edema.
10. Gillies MC, McAllister IL, Zhu M, et al. Intravitreal Ophthalmology. 2013;120(9):184351.
triamcinolone prior to laser treatment of diabetic mac- 23. Polito A, Pognuz DR, Lanzetta P, et al. Intravitreal
ular edema: 24-month results of a randomized con- triamcinolone as adjunctive treatment to laser photo-
trolled trial. Ophthalmology. 2011;118:86672. coagulation for cystoid macular edema due to branch
11. Diabetic Retinopathy Clinical Research Network, retinal vein occlusion. In: Abstracts of annual ARVO
Elman MJ, Aiello LP, Beck RW, et al. Randomized meeting, Fort Lauderdale, 14 May 2006.
trial evaluating ranibizumab plus prompt or deferred 24. Sakamoto T, Shimura M, Yasuda K, et al. Combination
laser or triamcinolone plus prompt laser for diabetic therapy for retinal vein occlusion. Ophthalmology.
macular edema. Ophthalmology. 2010;117:106477. 2010;117:1858.e13.
12. Elman MJ, Bressler NM, Qin H, et al. Expanded 25. Riese J, Loukopoulos V, Meier C, Timmermann M,
2-year follow-up of ranibizumab plus prompt or Gerding H. Combined intravitreal triamcinolone
deferred laser or triamcinolone plus prompt laser for injection and laser photocoagulation in eyes with per-
diabetic macular edema. Ophthalmology. 2011;118: sistent macular edema after branch retinal vein occlu-
60914. sion. Graefes Arch Clin Exp Ophthalmol. 2008;
13. Choi KS, Chung JK, Lim SH. Laser photocoagulation 246(12):16716.
combined with intravitreal triamcinolone acetonide 26. Fernandes JK, Patel R, Hariprasad SM. Combination
injection in proliferative diabetic retinopathy with therapy of an intravitreal dexamethasone implant
macular edema. Korean J Ophthalmol. 2007; (OzurdexTM) plus prompt focal laser for macular
21:117. edema secondary to retinal vein occlusions 12
14. Kaderli B, Avci R, Gelisken O, et al. Intravitreal month follow-up. In: Abstracts of annual ARVO
triamcinolone as an adjunct in the treatment of con- meeting, Fort Lauderdale, May 2012.
comitant proliferative diabetic retinopathy and diffuse 27. Parodi MB, Iacono P, Ravalico G. Intravitreal triam-
diabetic macular oedema. Combined IVTA and laser cinolone acetonide combined with subthreshold grid
treatment for PDR with CSMO. Int Ophthalmol. laser treatment for macular oedema in branch retinal
2007;26:20714. vein occlusion: a pilot study. Br J Ophthalmol.
15. Zacks DN, Johnson MW. Combined intravitreal injec- 2008;92(8):104650.
tion of triamcinolone acetonide and panretinal photo- 28. Cetin EN, Scanlon C, Akduman L. Outcomes of com-
coagulation for concomitant diabetic macular edema bined treatment with Ozurdex implant and grid laser
and proliferative diabetic retinopathy. Retina. 2005; photocoagulation for macular edema in branch retinal
25:13540. vein occlusion. In: Abstracts of annual ARVO meet-
16. Zein WM, Noureddin BN, Jurdi FA, et al. Panretinal ing, Fort Lauderdale, May 2012.
photocoagulation and intravitreal triamcinolone ace- 29. Pichi F, Specchia C, Vitale L, et al. Combination ther-
tonide for the management of proliferative diabetic apy with dexamethasone intravitreal implant and
retinopathy with macular edema. Retina. 2006; macular grid laser in patients with branch retinal vein
26:13742. occlusion. Am J Ophthalmol. 2014;157:60715.
17. Bandello F, Polito A, Pognuz DR, et al. Triamcinolone 30. Ruiz-Moreno JM, Schmidt-Erfurth U. Photodynamic
as adjunctive treatment to laser panretinal photocoag- therapy and high dose intravitreal triamcinolone to
ulation for proliferative diabetic retinopathy. Arch treat exudative age-related macular degeneration:
Ophthalmol. 2006;124:64350. 1-year outcome. Retina. 2006;26:60212.
102 P. Lanzetta et al.

31. Spaide RF, Sorenson J, Maranan L. Photodynamic 43. Soheilian M, Ramezani A, Obudi A, et al. Randomized
therapy with verteporfin combined with intravitreal trial of intravitreal bevacizumab alone or combined
injection of triamcinolone acetonide for choroidal with triamcinolone versus macular photocoagulation
neovascularization. Ophthalmology. 2005;112: in diabetic macular edema. Ophthalmology.
3014. 2009;116(6):114250.
32. Arias L, Garcia-Arumi J, Ramon JM, et al. 44. Ahmadieh H, Ramezani A, Shoeibi N, et al.
Photodynamic therapy with intravitreal triamcinolone Intravitreal bevacizumab with or without triamcino-
in predominantly classic choroidal neovasculariza- lone for refractory diabetic macular edema; a placebo-
tion: one-year results of randomized study. controlled, randomized clinical trial. Graefes Arch
Ophthalmology. 2006;113:224350. Clin Exp Ophthalmol. 2008;246:4839.
33. Augustin AJ, Schmidt-Erfurth U. Verteporfin photo- 45. Faghihi H, Roohipoor R, Mohammadi SF, et al.
dynamic combined with intravitreal triamcinolone in Intravitreal bevacizumab versus combined
all types of choroidal neovascularization due to age- bevacizumab-triamcinolone versus macular laser pho-
related macular degeneration. Ophthalmology. tocoagulation in diabetic macular edema. Eur J
2006;113:1422. Ophthalmol. 2008;18:9418.
34. Augustin AJ, Schmidt-Erfurth U. Verteporfin and 46. Sheth S, Rush R, Natarajan S, Gillies M. Intravitreal
intravitreal triamcinolone acetonide combination triamcinolone acetonide versus combined intravitreal
therapy for occult choroidal neovascularization in bevacizumab and dexamethasone in diffuse diabetic
age-related macular degeneration. Am J Ophthalmol. macular oedema. Clin Exp Ophthalmol. 2011;
2006;141:63845. 39(7):67381.
35. Russel SR, Hudson HL, Jerdan JA, et al. Anecortave 47. Ford JA, Lois N, Royle P, Clar C, et al. Current treat-
acetate for the treatment of exudative age-related ments in diabetic macular oedema : systematic review
macular degeneration: a review of clinical outcomes. and meta-analysis. BMJ Open. 2013;3:e002269.
Surv Ophthalmol. 2007;52:7990. 48. Goyal S, LaValley M, Subramanian ML. Meta-
36. Chaudhary V, Mao A, Hooper PL, et al. Triamcinolone analysis and review on the effect of bevacizumab in
acetonide as adjunctive treatment to verteporfin in diabetic macular edema Graefes archive for clinical
neovascular age-related macular degeneration: a pro- and experimental. Ophthalmology. 2011;249:1527.
spective randomized trial. Ophthalmology. 2007; 49. Ekdawi NS, Bakri SJ. Intravitreal triamcinolone and
114(12):21839. bevacizumab combination therapy for macular edema
37. Frimpong-Boateng A, Bunse A, Rfer F, et al. due to central retinal vein occlusion refractory to
Photodynamic therapy with intravitreal application of either treatment alone. Eye. 2007;21:112830.
triamcinolone acetonide in age-related macular 50. Ehrlich R, Ciulla TA, Moss AM, Harris A. Combined
degeneration: functional results in 54 patients. Acta treatment of intravitreal bevacizumab and intravitreal
Ophthalmol. 2009;87:1837. triamcinolone in patients with retinal vein occlusion:
38. Becerra EM, Morescalchi F, Gandolfo F, et al. Clinical 6 months of follow-up. Graefes Arch Clin Exp
evidence of intravitreal triamcinolone acetonide in the Ophthalmol. 2010;248:37580.
management of age-related macular degeneration. 51. Ceki O, Cakr M, Yazc AT, et al. A comparison of
Curr Drug Targets. 2011;12(2):14972. three different intravitreal treatment modalities of
39. Piermarocchi S, Sartore M, Lo Giudice G, Maritan V, macular edema due to branch retinal vein occlusion.
Midena E, Segato T. Combination of photodynamic Curr Eye Res. 2010;35(10):9259.
therapy and intraocular triamcinolone for exudative 52. Lee K, Jung H, Sohn J. Comparison of injection of
age-related macular degeneration and long-term cho- intravitreal drugs with standard care in macular edema
rioretinal macular atrophy. Arch Ophthalmol. secondary to branch retinal vein occlusion. Korean J
2008;126(10):136774. Ophthalmol. 2014;28(1):1925.
40. Nowak-Sliwinska P, Weiss A, Sickenberg M, et al. 53. Singer MA, Bell DJ, Woods P, et al. Effect of combi-
The role of photodynamic therapy in non-malignant nation therapy with bevacizumab and dexamethasone
and malignant eye disorders. J Anal Bioanal Tech. intravitreal implant in patients with retinal vein occlu-
2013;S1:007. doi:10.4172/2155-9872.S1-007. sion. Retina. 2012;32(7):128994.
41. Pece A, Russo G, Ricci F, Isola V, Introini U, 54. Mayer WJ, Remy M, Wolf A, et al. Comparison of
Querques G. Verteporfin photodynamic therapy com- intravitreal bevacizumab upload followed by a dexa-
bined with intravitreal triamcinolone for choroidal methasone implant versus dexamethasone implant
neovascularization due to angioid streaks. Clin monotherapy for retinal vein occlusion with macular
Ophthalmol. 2010;4:52530. edema. Ophthalmologica. 2012;228(2):1106.
42. Rishi P, Rishi E, Venkataraman A, et al. Photodynamic 55. Mayer WJ, Wolf A, Kernt M, et al. Twelve-month
monotherapy or combination treatment with intravit- experience with Ozurdex for the treatment of macular
real triamcinolone acetonide, bevacizumab or ranibi- edema associated with retinal vein occlusion. Eye.
zumab for choroidal neovascularization associated 2013;27(7):81622.
with pathological myopia. Indian J Ophthalmol. 56. Veritti D, Sarao V, Lanzetta P. Bevacizumab and tri-
2011;59:2426. amcinolone acetonide for choroidal neovasculariza-
8 Steroids in a Combination Strategy 103

tion due to age-related macular degeneration sented at: annual meeting of the American Society of
unresponsive to antivascular endothelial growth fac- Retinal Specialists, Vancouver, 28 Aug1 Sept 2010.
tors. J Ocul Pharmacol Ther. 2013;29(4):43741. 68. Veritti D, Lanzetta P. Triple therapy for anti-vascular
57. El Matri L, Chebil A, Kort F, Bouraoui R, Baklouti K, endothelial growth factor nonresponders in neovascu-
Mghaieth F. Intravitreal injection of triamcinolone com- lar age-related macular degeneration: impact of dif-
bined with bevacizumab for choroidal neovasculariza- ferent photodynamic therapy parameters.
tion associated with large retinal pigment epithelial Ophthalmologica. 2013;230(3):1317.
detachment in age-related macular degeneration. 69. Lonngi M, Gold AS, Murray TG. Combined bevaci-
Graefes Arch Clin Exp Ophthalmol. 2010;248:77984. zumab and triamcinolone acetonide injections for
58. Loewenstein A, Kuppermann BD, Weinberger D, et al. macular edema in a patient with astrocytic hamarto-
Safety and efficacy of Ozurdex (dexamethasone intra- mas and tuberous sclerosis. Ophthal Surg Lasers
vitreal implant) as adjunctive therapy to Lucentis in Imaging Retin. 2013;44(1):8590.
patients with choroidal neovascularization (CNV) sec- 70. Bhm MR, Uhlig CE. Use of intravitreal triamcino-
ondary to age-related macular degeneration (AMD). lone and bevacizumab in Coats disease with central
Paper presented at: Association for Research in Vision macular edema. Graefes Arch Clin Exp Ophthalmol.
and Ophthalmology, Fort Lauderdale, 3 May 2010. 2011;249(7):1099101.
59. Singer M, Patel SS, Uy HS, Li X-Y, Liu C-C, Whitcup 71. Shah NV, Houston SK, Markoe A, Murray
SM. Dexamethasone intravitreal implant alone and as TG. Combination therapy with triamcinolone aceton-
an adjunct to ranibizumab for the treatment of choroi- ide and bevacizumab for the treatment of severe radia-
dal neovascularization secondary to age-related mac- tion maculopathy in patients with posterior uveal
ular degeneration. Paper presented at: Retina Society melanoma. Clin Ophthalmol. 2013;7:187782.
annual meeting, Rome, 2225 Sept 2011. 72. Saeed AM. Combined vitrectomy and intravitreal
60. Ranchod TM, Ray SK, Daniels SA, Leong CJ, Ting injection versus combined laser and injection for
TD, Verne AZ. LuceDex: a prospective study compar- treatment of intractable diffuse diabetic macular
ing ranibizumab plus dexamethasone combination edema. Clin Ophthalmol. 2013;7:28397.
therapy versus ranibizumab monotherapy for neovas- 73. Koss MJ, Naser H, Sener A, et al. Combination ther-
cular age-related macular degeneration. Retina. apy in diabetic macular oedema and retinal vein
2013;33(8):16004. occlusionpast and present. Acta Ophthalmol.
61. Agustin AJ, Puls S, Offermann I. Triple therapy for 2012;90(6):5809.
choroidal neovascularization due to age-related macu- 74. Jin M, Ke Y, Zheng Z, Xiajing T. Twenty-five-gauge
lar degeneration: verteporfin PDT, bevacizumab, and vitrectomy and triamcinolone acetonide-assisted
dexamethasone. Retina. 2007;27:13340. internal limiting membrane peeling for chronic macu-
62. Bakri SJ, Couch SM, McCannel CA, Edwards lar edema associated with branch retinal vein occlu-
AO. Same-day triple therapy with photodynamic ther- sion. Retina. 2008;28:94756.
apy, intravitreal dexamethasone, and bevacizumab in 75. Uemura A, Yamamoto S, Sato E, et al. Vitrectomy
wet age-related macular degeneration. Retina. alone versus vitrectomy with simultaneous injection
2009;29:5738. of triamcinolone for macular edema associated with
63. Ahmadieh H, Taei R, Soheilian M, et al. Single- branch retinal vein occlusion. Ophthalmic Surg Lasers
session photodynamic therapy combined with Imaging. 2009;40:612.
intravitreal bevacizumab and triamcinolone for neo- 76. Opremcak EM, Rehmar AJ, Ridenour CD, et al.
vascular age-related macular degeneration. BMC Radial optic neurotomy with adjunctive intraocular
Ophthalmol. 2007;7:10. triamcinolone for central retinal vein occlusion: 63
64. Yip PP, Woo CF, Tang HH, Ho CK. Triple therapy for consecutive cases. Retina. 2006;26:30631.
neovascular age-related macular degeneration using 77. Garcia-Arumi J, Boixedera A, Martinez-Castillo V,
single-session photodynamic therapy combined with et al. Chorioretinal anastomosis alter radial optic neu-
intravitreal bevacizumab and triamcinolone. Br J rotomy for central retinal vein occlusion. Arch
Ophthalmol. 2009;93:7548. Ophthalmol. 2003;121:138591.
65. Forte R, Bonavolonta P, Benayoun Y, Adenis JP, 78. Oono Y, Tsuru H, Teruya K, et al. Long-term out-
Robert PY. Intravitreal ranibizumab and bevacizumab comes of radial optic neurotomy with or without
in combination with full-fluence verteporfin therapy adjunctive intraocular triamcinolone acetonide in cen-
and dexamethasone for exudative age-related macular tral retinal vein occlusion. In: Abstracts of annual
degeneration. Ophthalmic Res. 2011;45:12934. ARVO meeting, Fort Lauderdale, May 2008.
66. Ehmann D, Garcia R. Triple therapy for neovascular 79. Hwang JC, Gelman SK, Fine HF, Chang S, Del Priore
age-related macular degeneration (verteporfin photody- LV. Combined arteriovenous sheathotomy and intra-
namic therapy, intravitreal dexamethasone, and intravit- operative intravitreal triamcinolone acetonide for
real bevacizumab). Can J Ophthalmol. 2010;45:3640. branch retinal vein occlusion. Br J Ophthalmol.
67. Hudson H. RADICAL: 24-month results of a phase II 2010;94(11):14839.
exploratory randomized clinical trial of reduced fluence 80. Ferrari TM, Sborgia L, Furino C, et al. Radial optic
vPDT-anti-VEGF-dexamethasone in AMD. Paper pre- neurotomy, internal limiting membrane peeling and
104 P. Lanzetta et al.

intravitreal triamcinolone in the occlusion of the cen- Abstracts of annual ARVO meeting, Fort Lauderdale,
tral retinal vein occlusion. In: Abstracts of annual May 2007.
ARVO meeting, Fort Lauderdale, May 2004. 83. Lewis GM, Morris R. Treatment of cystoid macular
81. Fortunato P, Pollazzi L, Baroni M, et al. Venous edema in branch retinal vein occlusion with grid pho-
retinal flow reperfusion mechanisms following tocoagulation, intravitreal triamcinolone, and pneu-
radial optic neurotomy with adjunctive intraocular matic maculopexy. In: Abstracts of annual ARVO
triamcinolone in central retinal vein occlusion. meeting, Fort Lauderdale, May 2006.
Graefes Arch Clin Exp Ophthalmol. 2010;248: 84. Gutfleisch M, Spital G, Mingels A, Pauleikhoff D,
16773. Lommatzsch A, Heiligenhaus A. Pars plana vitrec-
82. Serny C, Mura F, Rebollo B, et al. Hemodilution with tomy with intravitreal triamcinolone: effect on uveitic
and without triamcinolone at the initial phase of vein cystoid macular oedema and treatment limitations. Br
occlusion: a retrospective comparative study. In: J Ophthalmol. 2007;91(3):3458.
Intravitreal Steroids as a Surgical
Adjunct 9
Stanislao Rizzo, Tomaso Caporossi,
and Francesco Barca

Contents
9.1 Introduction
9.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . 105
9.2 Preoperative Therapeutic Uses . . . . . . . . . 105 Corticosteroids are powerful tools for preserving
9.3 Intraoperative Use of Steroids ocular tissues due to their anti-inflammatory,
as Surgical Instrument . . . . . . . . . . . . . . . 106 anti-permeability, and anti-fibrotic properties.
9.4 Use During and After Vitrectomy: Steroids can be administered through topical
As an Adjuvant. . . . . . . . . . . . . . . . . . . . . . 107 application, intravitreal injection, and, more
9.4.1 Proliferative Vitreoretinopathy (PVR) . . . . . 107 recently, sustained-release delivery vehicles.
9.4.2 Macular Edema After Membrane Systemic administration of steroids is not useful
Peeling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
9.4.3 Chronic Refractory Uveitic Hypotony. . . . . 108 as a long-term therapeutic approach in the man-
9.4.4 Macular Edema in Vitrectomized agement of ophthalmic diseases.
Patients. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108 This class of drugs plays key roles before,
Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108 during, and after ophthalmic surgery. Alone or
as part of a combined therapeutic approach,
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109
corticosteroids reduce the patients ability to
mount a strong inflammatory response to
surgery.

9.2 Preoperative
Therapeutic Uses

Steroids are a mainstay in the management of a


wide range of inflammatory and vascular poste-
S. Rizzo, MD
rior segment disease including diabetic macular
Department of Neuroscience,
Azienda Ospedaliera Universitaria Pisana, edema (DME) [14], proliferative diabetic reti-
Viaparadisa n 2, Pisa 56124, Italy nopathy (PDR) [57], central retinal vein occlu-
e-mail: stanislaorizzo@gmail.com sion (CRVO) [810], branch retinal vein
T. Caporossi (*) F. Barca, MD occlusion (BRVO) [810], neovascular glau-
Chirurgia Oftalmica, Azienda Ospedaliera coma, chronic hypotony (phthisis), chronic uve-
Universitaria Pisana, via Paradisa n 2,
itis, pseudophakic cystoid macular edema
Pisa 56124, Italy
e-mail: tomaso.caporossi@me.com; (PCME), and neovascular age-related macular
barcaf@hotmail.com degeneration (AMD) [11].

A.J. Augustin (ed.), Intravitreal Steroids, 105


DOI 10.1007/978-3-319-14487-0_9, Springer International Publishing Switzerland 2015
106 S. Rizzo et al.

9.3 Intraoperative Use floating in the vitreous cavity (Fig. 9.2). The
of Steroids as Surgical absence of TA particles on the underlying retina
Instrument helped to identify areas where the ILM had been
successfully removed. Use of TA in peeling of
Triamcinolone acetonide (TA) is a water- the ERM has also been described as part of a
insoluble steroid molecule that has been used in double-staining technique with brilliant blue G
ophthalmic surgery as a stain to improve the visu- vital dye [16].
alization of ophthalmic structures such as the Staining of the vitreous with TA can facilitate
cortical vitreous and the internal limiting mem- its effective removal in a variety of surgical sce-
brane (ILM). Granules of TA are trapped on the narios. Sonoda et al. [17] used TA to identify
surface of the vitreous body, making it clearly residual vitreous cortex in a series of eyes that
visible under the surgical microscope. This prop- underwent vitrectomy for a variety of patholo-
erty is thought to be due to the integration of gies including diabetic retinopathy, diabetic mac-
insoluble white TA crystals into loosely orga- ular edema, branch retinal vein occlusion, and
nized collagen matrices in the vitreous [12]. rhegmatogenous retinal detachment (RRD). TA
The use of a corticosteroid as a surgical instru- helped to visualize residual vitreous cortex on the
ment was first suggested by Peyman et al. [12]
who described the use of TA to improve visual-
ization of the vitreous and the posterior hyaloid
during pars plana vitrectomy (PPV) (Fig. 9.1).
These authors showed that TA adheres to vitreous
fibrils, but not to the retina; when TA is injected
into the vitreous cavity, the crystals become
trapped in the vitreous gel, and the suspension
creates good visual contrast between the whit-
ened vitreous and the post-hyaloid space, which
contains a free-floating suspension of TA parti-
cles. Intraoperative injection of TA can be used to
demonstrate the vitreous anatomy, including the
highlighting of the invisible residual cortical
vitreous.
An extension of this mechanism is thought to Fig. 9.1 Posterior hyaloid stained with TA
be responsible for the staining of the superficial
portions of the ILM and epiretinal membrane
(ERM). Electron microscopy studies have shown
that a small layer of cells and collagen fibers
remains attached to the retina after manual vitre-
ous detachment. The latter residual material may
be responsible for the integration of TA crystals,
allowing the visualization of the ILM and ERM
[1315]. TA crystals entrapped in superficial col-
lagen fibrils facilitate the identification of the
ILM and ERM through the microscope [14]. TA
application can be repeated until the desired visu-
alization is achieved [1315].
Tognetto et al. [15] showed that TA staining is
useful in ILM peeling during vitreoretinal sur-
gery. Once peeled, the stained ILM could be seen Fig. 9.2 ILM visualization with TA during peeling
9 Intravitreal Steroids as a Surgical Adjunct 107

retinal surface after surgical posterior vitreous with proliferation of membranes on both the
separation during a PPV, especially in patients retina and the vitreous base [21].
with diabetic retinopathy. In 1980, Tano et al. [22] showed that intravit-
TA can also be used during surgical repair of real injection of dexamethasone inhibited fibro-
proliferative vitreoretinopathy (PVR). Furino blast growth in a rabbit model of intraocular
et al. [18] described the use of TA to facilitate proliferation and tractional retinal detachment.
removal of the posterior hyaloid and ERM in Later, in a model of PVR induced with macro-
patients undergoing PPV for RRD as a complica- phages, Hui and Hu [23] showed that a combina-
tion of PVR. Staining with TA improved visual- tion of daunomycin and TA was effective in
ization of the posterior hyaloid and ERM, preventing development of PVR in rabbits.
allowing more complete ERM removal in this A randomized controlled clinical trial pub-
critical scenario. TA was safe and well tolerated lished in 2008 showed no benefit of adjunctive
when used in a low concentration and rapidly TA injected into the vitreous cavity in eyes filled
removed during surgery. with silicone oil after PVR surgery. The trial was
The value of steroids and steroid-like factors conducted to investigate the role of silicone oil
to visualize prolapsed vitreous was soon recog- (frequently administered as a tamponade in sur-
nized by anterior segment surgeons. Huang et al. gery for PVR) as a vehicle for long-term postop-
[19] used a precursor of cortisol without steroid erative release of steroids. Ahmadieh and
activity (11-deoxycortisol) to visualize vitreous colleagues [21] found no benefit in outcomes
in the anterior chamber in an animal model of with the injection of 4 mg TA after surgery. The
posterior capsule rupture. The use of TA in com- retinal reattachment rate was not statistically sig-
plicated cataract surgery was found to be benefi- nificantly different between treatment and con-
cial to visualize vitreous in the anterior chamber; trol groups at 6 months follow-up.
Yamakiri et al. [20] recommended a washout of
the anterior chamber to avoid complications such
as elevation of IOP. 9.4.2 Macular Edema After
Membrane Peeling

9.4 Use During and After Recently, the effects of intraoperative and post-
Vitrectomy: As an Adjuvant operative use of steroids in membrane-peeling
surgery have been investigated by several authors.
The use of steroids as an adjuvant during or after The major findings suggest that local or systemic
vitrectomy has been suggested for numerous steroid treatment, during or after successful vit-
indications, including control of PVR, reduction rectomy combined with peeling, does not
of macular edema after membrane peeling, treat- improve anatomic and functional outcomes.
ment of chronic refractory uveitic hypotony, and TA injected during vitrectomy and membrane
treatment of macular edema in vitrectomized peeling in patients with idiopathic ERM did not
patients. affect postoperative foveal thickness or func-
tional recovery [24]. Similarly, there was no dif-
ference with or without the use of intravitreal TA
9.4.1 Proliferative in long-term anatomic and visual outcomes after
Vitreoretinopathy (PVR) vitrectomy and membrane peeling in patients
with idiopathic ERM, although there was an
PVR can occur as a complication of RRD. Surgical increased risk of IOP elevation [25].
approaches to the management of PVR include Ritter and colleagues [26] prospectively eval-
PPV, membrane peeling, endolaser application, uated the use of systemic steroids after mem-
and postoperative intraocular tamponade. Despite brane peeling in eyes with macular edema due to
these measures, PVR can recur after surgery, idiopathic ERM. Patients undergoing 23-gauge
108 S. Rizzo et al.

vitrectomy with ERM and ILM peeling were ran- group (mean 0.92 g/mL). The authors concluded
domly assigned to receive postoperative oral that faster clearance of the drug should be consid-
prednisolone. No statistically significant differ- ered when intravitreal injection of TA is used in
ences in anatomic or functional outcomes were vitrectomized eyes.
found between treated and control group at up to Similar differences in clearance rates have
3 months postoperative. been reported with VEGF [30], amphotericin B
[31], and 5-fluorouracil [32], with faster clear-
ance from vitrectomized than non-vitrectomized
9.4.3 Chronic Refractory Uveitic rabbit eyes.
Hypotony The use of sustained-release delivery modalities
may be a solution to this rapid clearance. Chang-
Long-term intraocular delivery of the steroid Lin and colleagues [33] assessed the pharmacoki-
fluocinolone acetonide (FA) has been evaluated netics of a sustained-release 0.7-mg dexamethasone
in the treatment of refractory uveitic hypotony. intravitreal implant in vitrectomized (n = 25) and
Dayani et al. [27] reported the results of com- non-vitrectomized (n = 25) rabbit eyes, showing
bined PPV, insertion of a sustained-delivery FA that the maximum concentration of dexamethasone
implant, and silicone oil infusion in 13 eyes with in the vitreous humor of non-vitrectomized eyes
refractory uveitic hypotony with a mean baseline was 791 ng/mL, compared with 731 ng/mL in vit-
IOP of 2.3 mmHg. At 6, 12, and 24 months fol- rectomized eyes at 22-day follow-up.
low-up, postoperative mean IOP was 5.9 mmHg, The CHAMPLAIN Study Group [34] evalu-
5.1 mmHg, and 5.0 mmHg, respectively. No ated the effectiveness of the 0.7-mg dexametha-
intraoperative complications were described, and sone intravitreal implant for the treatment of
the procedure was well tolerated by patients. diabetic macular edema (DME) in vitrectomized
eyes. In this prospective multicenter study, 55
patients with refractory DME (mean duration,
9.4.4 Macular Edema 43 months) and history of PPV received a single
in Vitrectomized Patients 0.7-mg implant. Statistically and clinically sig-
nificant improvements in visual acuity and cen-
Macular edema can occur as an inflammatory tral retinal thickness were shown in treated eyes
reaction after vitreoretinal surgery, including vit- during the follow-up.
rectomy. Steroid therapy is a potential approach
to treat macular edema after vitrectomy. However, Conclusions
several studies have indicated that a number of From its height of popularity a few years ago,
drugs are cleared from the posterior segment the use of intravitreal TA has exponentially
faster in vitrectomized eyes than in eyes with an decreased due to a high rate of complication.
intact vitreous. The association of corticosteroid therapy with
In the 1980s, Schindler and colleagues [28] the development of posterior subcapsular cat-
showed rapid clearance of TA from vitrectomized aracts has been well documented. Despite
rabbit eyes (average 17 days) compared with eyes these findings, these drugs are widely used
without surgery (average 41 days). therapeutically, principally to capitalize on
More recently, Chin et al. [29] compared the their ability to inhibit inflammatory responses
clearance of TA between vitrectomized (n = 42) before, during, and after surgery.
and non-vitrectomized (n = 42) rabbit eyes. The For many years, an injectable intramuscu-
concentration of TA decreased 1.5 times more rap- lar suspension of TA was adapted for ophthal-
idly in vitrectomized than non-vitrectomized eyes. mic use because no ophthalmic formulation
At 30-day follow-up, TA was detected in only one was commercially available. Recently, inject-
eye in the vitrectomized group (0.22 g/mL) com- able suspensions have become available for
pared with four eyes in the non-vitrectomized intravitreal injection, with indications from
9 Intravitreal Steroids as a Surgical Adjunct 109

the US FDA for use in sympathetic ophthal- focal/grid photocoagulation and intravitreal
mia, temporal arteritis, uveitis, and ocular triamcinolone for diabetic macular edema. Arch
Ophthalmol. 2009;127(3):24551.
inflammatory conditions unresponsive to topi- 4. Diabetic Retinopathy Clinical Research Network,
cal corticosteroids. A sustained-release dexa- Elman MJ, Aiello LP, Beck RW, et al. Randomized
methasone intravitreal implant has FDA trial evaluating ranibizumab plus prompt or deferred
approval for the treatment of macular edema laser or triamcinolone plus prompt laser for diabetic
macular edema. Ophthalmology. 2010;117(6):1064
following BRVO and CRVO and for the treat- 77.e35.
ment of noninfectious uveitis affecting the 5. Zein WM, Noureddin BN, Jurdi FA, Schakal A,
posterior segment. A 0.59-mg FA intravitreal Bashshur ZF. Panretinal photocoagulation and intra-
implant is FDA approved for the treatment of vitreal triamcinolone acetonide for the management
of proliferative diabetic retinopathy with macular
chronic noninfectious uveitis affecting the edema. Retina. 2006;26(2):13742.
posterior segment. A smaller, 190-g FA 6. Maia Jr OO, Takahashi BS, Costa RA, Scott IU,
implant is approved in some countries in Takahashi WY. Combined laser and intravitreal triam-
Europe for the treatment of vision impairment cinolone for proliferative diabetic retinopathy and
macular edema: one-year results of a randomized clin-
associated with chronic DME insufficiently ical trial. Am J Ophthalmol. 2009;147(2):2917.e2.
responsive to other available therapies. 7. Mirshahi A, Shenazandi H, Lashay A, Faghihi H,
Steroids are useful to better visualize the Alimahmoudi A, Dianat S. Intravitreal triamcinolone
vitreous, as a non-colorant precipitating stain- as an adjunct to standard laser therapy in coexisting
high-risk proliferative diabetic retinopathy and clini-
ing agent in uncomplicated membrane peeling cally significant macular edema. Retina. 2010;30(2):
and in sustained-release delivery modes in 2549.
intraocular inflammatory diseases. Steroids 8. Scott IU, Ip MS, VanVeldhuisen PC, SCORE Study
are not effective to treat macular edema in vit- Research Group, et al. A randomized trial comparing
the efficacy and safety of intravitreal triamcinolone
rectomized eyes or to visualize the ILM in with standard care to treat vision loss associated with
complex cases such as highly myopic eyes. macular edema secondary to branch retinal vein
The therapeutic use of steroids remains invalu- occlusion: the Standard Care vs Corticosteroid for
able in the treatment of inflammatory patholo- Retinal Vein Occlusion (SCORE) study report 6. Arch
Ophthalmol. 2009;127(9):111528. Erratum in: Arch
gies such as uveitis. Ophthalmol. 2009;127(12):1655.
Widening therapeutic indications and the 9. Ip MS, Scott IU, VanVeldhuisen PC, SCORE Study
availability of new longer-lasting sustained- Research Group, et al. A randomized trial comparing
release delivery modes for corticosteroids will the efficacy and safety of intravitreal triamcinolone
with observation to treat vision loss associated with
lead to new approaches to treat the widest pos- macular edema secondary to central retinal vein
sible range of ocular diseases, along with sig- occlusion: the Standard Care vs Corticosteroid for
nificant lowering of health-care costs for both Retinal Vein Occlusion (SCORE) study report 5. Arch
patients and institutions. Ophthalmol. 2009;127(9):110114. Erratum in: Arch
Ophthalmol. 2009;127(12):1648.
10. Haller JA, Bandello F, Belfort Jr R, Ozurdex GENEVA
Study Group, Li J, et al. Dexamethasone intravitreal
References implant in patients with macular edema related to
branch or central retinal vein occlusion twelve-month
1. Silva PS, Sun JK, Aiello LP. Role of steroids in the study results. Ophthalmology. 2011;118(12):245360.
11. Becerra EM, Morescalchi F, Gandolfo F, et al. Clinical
management of diabetic macular edema and prolifera-
evidence of intravitreal triamcinolone acetonide in the
tive diabetic retinopathy. Semin Ophthalmol. 2009;
management of age-related macular degeneration.
24(2):939. Review.
Curr Drug Targets. 2011;12(2):14972.
2. Diabetic Retinopathy Clinical Research Network. 12. Peyman GA, Cheema R, Conway MD, Fang
A randomized trial comparing intravitreal triamcino- T. Triamcinolone acetonide as an aid to visualization
lone acetonide and focal/grid photocoagulation for of the vitreous and the posterior hyaloid during pars
diabetic macular edema. Ophthalmology. 2008; plana vitrectomy. Retina. 2000;20(5):5545.
115(9):14479, 1449.e110. 13. Shah GK, Rosenblatt BJ, Smith M. Internal limiting
3. Diabetic Retinopathy Clinical Research Network membrane peeling using triamcinolone acetonide:
(DRCR.net), Beck RW, Edwards AR, Aiello LP, et al. histopathologic confirmation. Am J Ophthalmol.
Three-year follow-up of a randomized trial comparing 2004;138(4):6567.
110 S. Rizzo et al.

14. Horio N, Horiguchi M, Yamamoto N. Triamcinolone- idiopathic epiretinal membrane. Retina. 2012;32(5):
assisted internal limiting membrane peeling during 8926.
idiopathic macular hole surgery. Arch Ophthalmol. 25. Lai CC, Wang NK, Wu WC, et al. The long-term ana-
2005;123(1):969. tomical and visual effect of intravitreal triamcinolone
15. Tognetto D, Zenoni S, Sanguinetti G, Haritoglou C, injection during vitrectomy for the treatment of idio-
Ravalico G. Staining of the internal limiting mem- pathic macular epiretinal membrane. Cutan Ocul
brane with intravitreal triamcinolone acetonide. Toxicol. 2011;30(4):2927.
Retina. 2005;25(4):4627. 26. Ritter M, Sacu S, Matt G, et al. Use of systemic ste-
16. Shimada H, Nakashizuka H, Hattori T, Mori R, roid after successful macular surgery in eyes with
Mizutani Y, Yuzawa M. Double staining with brilliant epiretinal membrane: a randomized, controlled clini-
blue G and double peeling for epiretinal membranes. cal study. Eye (Lond). 2011;25(10):128493.
Ophthalmology. 2009;116(7):13706. 27. Dayani PN, Chow J, Stinnett SS, Jaffe GJ. Pars plana
17. Sonoda KH, Sakamoto T, Enaida H, et al. Residual vitrectomy, fluocinolone acetonide implantation, and
vitreous cortex after surgical posterior vitreous sepa- silicone oil infusion for the treatment of chronic,
ration visualized by intravitreous triamcinolone ace- refractory uveitic hypotony. Am J Ophthalmol.
tonide. Ophthalmology. 2004;111(2):22630. 2011;152(5):84956.e1.
18. Furino C, Micelli Ferrari T, Boscia F, Cardascia N, 28. Schindler RH, Chandler D, Thresher R, Machemer
Recchimurzo N, Sborgia C. Triamcinolone-assisted R. The clearance of intravitreal triamcinolone aceton-
pars plana vitrectomy for proliferative vitreoretinopa- ide. Am J Ophthalmol. 1982;93(4):4157.
thy. Retina. 2003;23(6):7716. 29. Chin HS, Park TS, Moon YS, Oh JH. Difference in
19. Huang R, Kaji Y, Fukuda S, Oshika T. Experimental use clearance of intravitreal triamcinolone acetonide
of estriol for visualizing the vitreous body in the anterior between vitrectomized and nonvitrectomized eyes.
chamber after posterior capsule rupture in animal mod- Retina. 2005;25(5):55660.
els. J Cataract Refract Surg. 2009;35(7):12605. 30. Lee SS, Ghosn C, Yu Z, et al. Vitreous VEGF clear-
20. Yamakiri K, Uchino E, Kimura K, Sakamoto T. ance is increased after vitrectomy. Invest Ophthalmol
Intracameral triamcinolone helps to visualize and Vis Sci. 2010;51(4):21358.
remove the vitreous body in anterior chamber in cata- 31. Doft BH, Weiskopf J, Nilsson-Ehle I, Wingard Jr LB.
ract surgery. Am J Ophthalmol. 2004;138(4):6502. Amphotericin clearance in vitrectomized versus non-
21. Ahmadieh H, Feghhi M, Tabatabaei H, Shoeibi N, vitrectomized eyes. Ophthalmology. 1985;92(11):
Ramezani A, Mohebbi MR. Triamcinolone acetonide 16015.
in silicone-filled eyes as adjunctive treatment for pro- 32. Jarus G, Blumenkranz M, Hernandez E, Sossi N.
liferative vitreoretinopathy: a randomized clinical Clearance of intravitreal fluorouracil. Normal and
trial. Ophthalmology. 2008;115(11):193843. aphakic vitrectomized eyes. Ophthalmology. 1985;
22. Tano Y, Chandler D, Machemer R. Treatment of intra- 92(1):916.
ocular proliferation with intravitreal injection of tri- 33. Chang-Lin JE, Burke JA, Peng Q, et al.
amcinolone acetonide. Am J Ophthalmol. 1980;90(6): Pharmacokinetics of a sustained-release dexametha-
8106. sone intravitreal implant in vitrectomized and nonvit-
23. Hui YN, Hu D. Prevention of experimental prolifera- rectomized eyes. Invest Ophthalmol Vis Sci. 2011;
tive vitreoretinopathy with daunomycin and triamcin- 52(7):46059.
olone based on the time course of the disease. Graefes 34. Boyer DS, Faber D, Gupta S, Ozurdex CHAMPLAIN
Arch Clin Exp Ophthalmol. 1999;237(7):6015. Study Group, et al. Dexamethasone intravitreal
24. Ahn JH, Park HJ, Lee JE, Oum BS. Effect of intravit- implant for treatment of diabetic macular edema in
real triamcinolone injection during vitrectomy for vitrectomized patients. Retina. 2011;31(5):91523.

You might also like