You are on page 1of 12

review article Diabetes, Obesity and Metabolism 17: 219–230, 2015.

© 2014 John Wiley & Sons Ltd

ARTICLE
REVIEW
New treatments for diabetic retinopathy
A. Das1,2 , S. Stroud1 , A. Mehta1 & S. Rangasamy3
1 Department of Surgery/Ophthalmology, University of New Mexico School of Medicine, Albuquerque, NM, USA
2 Department of Surgery, New Mexico VA Health Care System, Albuquerque, NM, USA
3 T-Gen Institute, Phoenix, AZ, USA

Diabetic retinopathy is the major cause of vision loss in middle-aged adults. Alteration of the blood–retinal barrier (BRB) is the hallmark of diabetic
retinopathy and, subsequently, hypoxia may result in retinal neovascularization. Tight control of systemic factors such as blood glucose, blood pressure
and blood lipids is essential in the management of this disease. Vascular endothelial growth factor (VEGF) is one of the most important factors responsible
for alteration of the BRB. The introduction of anti-VEGF agents has revolutionized the therapeutic strategies used in people with diabetic retinopathy,
and the use of laser therapy has been modified. In the present article, we examine the clinical features and pathophysiology of diabetic retinopathy and
review the current status of new treatment recommendations for this disease, and also explore some possible future therapies.
Keywords: blood–retinal barrier, diabetes complications, diabetes mellitus, diabetic retinopathy, vascular endothelial growth factor

Date submitted 7 June 2014; date of first decision 7 July 2014; date of final acceptance 20 August 2014

Diabetes mellitus is quickly becoming the global epidemic of into (i) non-proliferative (NPDR) and (ii) proliferative (PDR).
the 21st century. Currently, there are 382 million people with NPDR is further divided into four stages: mild, moderate,
diabetes mellitus worldwide, and this number is projected to severe and very severe (Table 1). In the mild/moderate stages,
reach 559 million by the year 2035 [1]. The estimated global microaneurysms and intraretinal haemorrhages are present in
healthcare expenditure for treating diabetes and its compli- the retina, mostly in the posterior pole. As the blood–retinal
cations was $376 bn in 2010, and this number is projected to barrier (BRB) breaks down, the plasma leaks out of the retinal
exceed $490 bn by 2030 [1]. Diabetic retinopathy, a microvas- capillaries resulting in retina oedema. The oedema most com-
cular complication of diabetes, is prevalent in ∼35% of people monly occurs in the macula, resulting in distortion of vision
with diabetes [2]. It is the leading cause of vision loss in adults and vision loss. Macular oedema is the most common cause of
aged 20–64 years in developed countries [2,3]. Although laser vision loss in diabetic retinopathy (20–25% of people with dia-
photocoagulation therapy has been the mainstay of manage- betes have vision loss after 10 years). Hard exudates that look
ment therapy in addition to control of systemic factors, the use like well-defined yellow deposits, often accompany macular
of intravitreal anti-vascular endothelial growth factor (VEGF) oedema, and these lesions represent lipid materials that leak
agents and steroids in recent years has revolutionized the man- out of the retinal microvessels. NPDR becomes severe when
agement of diabetic macular oedema. In the present review, it shows intraretinal haemorrhages in all four quadrants, or
we will discuss the pathogenesis of diabetic retinopathy and venous beading in two quadrants, or intraretinal microvascular
the treatment strategies currently available for the treatment of abnormalities in one quadrant (the so-called 4-2-1 rule) [6].
diabetic macular oedema and proliferative diabetic retinopathy The retinopathy is considered very severe if two of these features
(PDR; Table 1). are present. Of people with severe NPDR, 50% will progress to
the PDR stage in 1 year [6]. PDR is present when new retinal
vessels grow out of the retinal capillaries into the vitreous. As
Clinical Features these new vessels are fragile (of endothelial tube formation),
The earliest clinical lesions of diabetic retinopathy are microa- they often lead to preretinal and vitreous haemorrhage, causing
neurysms, or focal dilations of retinal microvessels seen as symptoms of floaters and decreased, severe vision loss. As the
deep red dots, mainly in the posterior pole. Usually these new vessels grow over the vitreous interface, other cells, such
lesions appear and disappear over time and cause no symp- as fibroblasts and glial cells, participate in forming epiretinal
toms themselves. Microaneurysms are present in almost all membranes that contract and cause traction retinal detachment
people with type 1 diabetes of 20 years duration, and in 80% with severe vision loss. Some people with PDR have growth of
of people with type 2 diabetes [4,5]. Based on the absence the neovascular tissue on the surface of the iris into the anterior
or presence of new vessels, diabetic retinopathy is classified chamber angle, causing blockage of the aqueous outflow, and a
severe type of glaucoma, called neovascular glaucoma.

Correspondence to: Arup Das, MD, PhD, Department of Surgery, Division of Ophthalmology, Pathophysiology
University of New Mexico School of Medicine, MSC10-5610, 1 University of New Mexico,
Albuquerque, NM 87131, USA. The alteration of the BRB is the hallmark of the pathogenesis of
E-mail: adas@unm.edu diabetic retinopathy. Normally, this BRB at the retinal capillary
review article DIABETES, OBESITY AND METABOLISM

Table 1. International classification of the clinical features of diabetic retinopathy

International classification of diabetic retinopathy


Severity level Ophthalmoscopic findings
No retinopathy No abnormalities
Mild non-proliferative retinopathy Microaneursyms only
Moderate non-proliferative More than just microaneursyms (retinal haemorrhages and
retinopathy hard exudates), less than severe non-proliferative diabetic
retinopathy
Severe non-proliferative retinopathy Any one of the following three features:
20 retinal haemorrhages in each of the four quadrants
Venous beating in two quadrants
Intraretinal microvascular abnormalities in one quadrant
Very severe non-proliferative Any two of the above features present
retinopathy
Proliferative retinopathy One or both of the following features:
Neovascularization
Preretinal/vitreous haemorrhage

Adapted from [72].

level comprises endothelial cell–cell junctions, basement Systemic Factor Control


membrane and pericytes that cover the vessels outside. In dia-
The beneficial effects of systemic control of blood glucose
betes, three changes happen at the BRB: (i) loss of endothelial
on retinopathy have been shown in three large, randomized
cell–cell junctions; (ii) thickening of the basement membrane;
clinical trials. The Diabetes Control and Complications Trial
and (iii) selective loss of pericytes. The breakdown of the BRB
showed that tight glucose control [glycated haemoglobin
leads to intraretinal haemorrhages, hard exudates and macular
(HbA1c) of <6% or 42.1 mmol/mol) in people with type 1
oedema. Selective pericyte loss is one of the early histopatho- diabetes prevented the development of diabetic retinopathy by
logical lesions seen in diabetic retinopathy [7]. It is not clear 76% and decreased progression of existing diabetic retinopathy
why there is selective loss of pericytes in diabetes. Normally, by 54% [12]. The United Kingdom Prospective Diabetes Study
pericytes (modified smooth muscle cells), are contractile, and (UKPDS) then showed that intensive glucose control (median
maintain the retinal capillary flow [8]. Pericyte loss leads to HbA1c of 7.0% or 53 mmol/mol versus 7.9% or 62.8 mmol/mol
focal weakening of the capillary wall as well as uninhibited in conventional glucose control group) led to a 25% reduced
focal endothelial cell proliferation that leads to formation of rate of microvascular disease, including retinopathy, in patients
microaneurysms [9]. Later, endothelial cells also die, resulting with type 2 diabetes [13]. More recently, the ACCORD Eye
in acellular capillaries and non-perfusion in the retina. Apop- Study Group [14] reported that tight glucose control in patients
tosis, or programmed cell death, is responsible for death of with type 2 diabetes (median HbA1c of 6.4% or 46.4 mmol/mol
both these cell types in diabetes. Neuronal death attributable in the intensive group versus 7.5% or 58.5 mmol/mol in the
to apoptosis may occur in the ganglion cell layer even earlier conventional group) reduced progression of diabetic retinopa-
than the vascular lesions. Such a silent death of neurons before thy by 35% over a 4-year span. A recent meta-analysis of the
the vascular lesions appear may explain the defect in dark recent clinical trials in people with type 2 diabetes concluded,
adaptation and reduced contrast sensitivity seen in people however, that intensive control of blood glucose did not prevent
with diabetes before the development of retinopathy. The the need for photocoagulation, nor the development of severe
hyperglycaemia-induced pathogenesis of diabetic retinopathy vision loss [15]. With regard to blood pressure control, the
is linked to four major biochemical pathways: (i) the increased UKPDS found that controlling blood pressure also led to a sig-
polyol pathway; (ii) increased advanced glycation end product nificant decrease in visual loss [13]. Interestingly, the ACCORD
formation; (iii) activation of protein kinase C isoforms; and (iv) Eye study showed that tight hypertension control did not sig-
the increased hexosamine pathway [10]. All these pathways nificantly decrease the incidence of diabetic retinopathy [14].
eventually lead to oxidative stress, inflammation and retinal These results point to the notion that maintaining systolic blood
vascular dysfunction. Hypoxia is the initiating factor in the pressure at <150 mmHg probably has a therapeutic threshold of
development of new retinal vessels seen in PDR. Many angio- ∼130 mmHg systolic pressure, lower than which would proba-
genic factors, such as VEGF, basic fibroblast growth factor, bly have little effect on reducing visual loss in diabetic retinopa-
insulin-like growth factor and angiopoietin-2, play a key role thy. Interestingly, a newer study, the Hypertension Intervention
in this process [11]. Normally, there is a balance of angiogenic Nurse Telemedicine Study, has shown that lowering systolic
factors and endogenous anti-angiogenic factors [e.g. Pigment blood pressure by a mean of 8 mmHg over an 18-month period
Epithelium Derived Factor (PEDF), endostatin], and this bal- led to decreased progression of diabetic retinopathy among
ance breaks down in PDR, ultimately resulting in the growth of mostly male veterans monitoring blood pressure at home and
new vessels. receiving nurse-administered telemedicine behavioural and

220 Das et al. Volume 17 No. 3 March 2015


DIABETES, OBESITY AND METABOLISM review article
medical interventions [16]. Although control of blood pressure caused by functional loss of retinal tissue and deep burns
overall probably reduces the progression of diabetic retinopa- that caused long term scarring and fibrosis. Two principles
thy, it has been shown that treatment with specific agents may help achieve this goal. One, longer laser wavelengths are used,
have additional benefits. For instance, candesartan led to a 25% 647 nm krypton red and 810 nm diode lasers, in order to reduce
lower incidence of diabetic retinopathy in people with type 1 burn intensity and avoid absorption to macular chromophores.
diabetes [17] and a 34% higher regression of diabetic retinopa- Second, the development of micropulsar techniques, which
thy in people with type 2 diabetes with mild retinopathy com- increase the delay between pulses and reduce the size of reti-
pared with placebo [18]. With regard to lipid control, it appears nal lesions by eliminating heat diffusion and lesion growth after
that LDL cholesterol and triglyceride levels are directly related treatment. In 2005, a pilot study of the subthreshold diode
to the incidence and severity of diabetic retinopathy, while micropulsar (SDM) laser, which uses an 810-nm diode laser at
HDL cholesterol level is indirectly related [19]. The Fenofibrate sub-optimum intensity applied to all areas of macular thicken-
Intervention and Event Lowering in Diabetes study showed that ing, showed that SDM laser photocoagulation was similar to
fenofibrate (200 mg/day) reduced the need for retinopathy laser previous laser treatments, without any adverse effects or evi-
treatment in people with type 2 diabetes [20]. This was further dence of iatrogenic retinal damage [27–29].
confirmed in the ACCORD Eye study, which provided evi- Further studies showed similar effects. In 2009, a prospec-
dence that fenofibrate added to simvastatin therapy in people tive randomized control trial comparing the efficacy of SDM
with type 2 diabetes slowed down the progression of diabetic laser photocoagulation with conventional argon green laser
retinopathy at 4 years [14]. Among other risk factors, anaemia
used in the modified ETDRS protocol, showed that best cor-
has been associated with progression of diabetic retinopathy
rected visual acuity (BCVA) remained unchanged at 12 months
[21]. The Early Treatment Diabetic Retinopathy Study (ETDRS)
in either treatment arm; however, laser scars were identified in
showed an association between a decrease in haematocrit and
only 13.9% of eyes treated with SDM, while laser scars were
an increase in incidence of high-risk PDR. In people with type
identified in 59% of eyes treated with conventional green laser
2 diabetes, a strong correlation was observed between diabetic
[30]. Furthermore, in a study conducted in Japan, fundus aut-
retinopathy and obstructive sleep apnoea [22]. Pregnancy has
been associated with rapid progression of diabetic retinopathy, ofluorescence, which uses fluorescence from the retina to indi-
and this has been attributed to high levels of oestrogens as well cate the health of RPE cells, was used to evaluate morphological
as serum insulin-like growth factor in pregnancy [23]. changes objectively in 24 eyes treated with SDM laser pho-
In treatment of diabetic retinopathy, the ophthalmologist tocoagulation for centre-involving clinically significant mac-
should be in close consultation with the primary care physician ular oedema. At 1-year follow-up, fundus autofluorescence
regarding tight control of blood glucose, blood pressure and remained unchanged from baseline values in the SDM laser
blood lipids. The HbA1c target should be 7%, as further lower- photocoagulation treatment arm, thus indicating that there was
ing of blood glucose may be difficult to achieve in people with no RPE cell damage [31].
long-standing type 2 diabetes, and may cause a higher number The lack of damage to deeper retinal cell layers and long-term
of cardiovascular events and mortality [14]. With regard to con- scarring, as evidenced by these studies, helped pave the way
trol of retinopathy, the aim should be a systolic blood pressure for the introduction of high-density SDM lasers in an effort
of <140 mmHg, and fenofibrates should be added to the statin to improve clinical outcomes. The aim of the high-density
therapy for optimum blood lipid control. approach was to increase lateral spread of thermally stimulated
RPE cells, thus delivering ∼900 burns throughout the macula,
but avoiding the fovea, and even involving normal-appearing
Laser Therapy areas of the retina. This hypothesis was investigated by Lavinsky
Diabetic Macular Oedema et al. [32] and reported in 2011. These authors showed that
The ETDRS in 1985 showed that the use of small, light-intensity high-density SDM laser photocoagulation led to a superior
laser burns to microaneurysms, and/or diffuse area of thick- improvement in BCVA and central macular thickness in com-
ening in a grid pattern resulted in a 50% reduction in severe parison with low-density SDM laser and conventional argon
vision loss [24]. The proposed underlying mechanisms behind laser therapy.
this approach involve increased intraocular oxygen tension, a Another newly developed mode of subthermal laser ther-
decreased production of vasoactive cytokines, primarily VEGF, apy involves creating a horse-shoe shaped macular grid
and increased phagocytosis by retinal pigment epithelial (RPE) pattern using a 577-nm yellow semi-automated patterned
cells and glial cells. Recently, it has been proposed that RPE scanning laser (PASCAL; Topcon, Capelle aan den IJssel, The
cells at the margins of burns participate in their own recovery Netherlands), which allows the physician to deliver multiple
through the modulation of various cytokines via photorecep- spots of photocoagulation simultaneously in a preset pattern.
tors [25,26]. Because of the efficacy of anti-VEGF agents for The PASCAL uses either 20 or 10-ms burns localized to the
diabetic macular oedema, the current indication of focal/grid outer retina without changes to adjacent RPE cells or inner
laser is now limited to those patients with non-centre-involving neuroretina, similarly to the SDM laser [33,34]; however,
diabetic macular oedema. unlike the invisible nature of the SDM laser burn, the burn
In recent years, treatment has shifted towards selectively from the PASCAL is able to be tracked 1 h after treatment
applying subthermal intensity to RPE cells, while sparing the by frequency domain-optical coherence tomography, which
neurosensory retina, therefore, reducing iatrogenic side effects shows localized defects at the junction of the photoreceptor

Volume 17 No. 3 March 2015 doi:10.1111/dom.12384 221


review article DIABETES, OBESITY AND METABOLISM

inner and outer segments and apical RPE. At 12 months, fre- option. Conventionally, a clinician dilates a patient’s eyes
quency domain-optical coherence tomography shows a normal and uses a slit-lamp or indirect lens technique to visualize
inner and outer segment layer with burns localized only to the the retina during photocoagulation procedures; however,
uppermost RPE layer of the retina [23]. The use of the 577-nm new technology, specifically the Optos camera (Optos,
yellow laser also gives it the ability to be titrated to a certain Dunfermline, UK), allows a clinician to visualize the retina up
power. A new navigated laser photocoagulator (NAVILAS ,
NAVILAS Laser System, Irvine, CA, USA), a prototype of
® to 200 internal degrees (sometimes without pupillary dilation),
perform fluorescein angiography and autofluorescence to map
retinal eye-tracking laser delivery system with integrated dig- sub-threshold laser burns on the retina. The PETER PAN
ital fundus imaging, allows registered image overlay and laser study also concluded that targeted retinal photocoagulation
stabilization on the retina and thus a higher rate of accuracy in could be used in conjunction with visible peripheral retinal
the treatment of diabetic retinopathy lesions [35]. The Com- ischaemia on Optos angiography [42]. The Optos device has a
bination of Anti-VEGF and Navigational Laser in Diabetic favourable safety profile and will probably continue to become
Macular Edema (CAVNAV) study, a 12-month prospective more prominent in the treatment of retinopathy [44].
cohort study, showed a reduced number of anti-VEGF injec-
tions and a higher proportion of injection-free patients with
the combination therapy of anti-VEGF drugs and NAVILAS Pharmacotherapies
laser compared with the monotherapy of anti-VEGF injections
in diabetic macular oedema [36]. Anti-VEGF Therapy
VGEF has been the most important factor that has been inves-
Proliferative Diabetic Retinopathy tigated extensively in relation to the pathophysiology retinal
neovascularization and alteration of the BRB. VEGF levels are
The Diabetic Retinopathy Study [37] showed that panretinal
photocoagulation (PRP) reduced the risk of severe visual loss significantly elevated in the vitreous of patients with diabetic
by ∼50% over 5 years. Furthermore, the preservation of vision macular oedema when compared with non-diabetic eye condi-
from PRP was shown in the ETDRS follow-up study [38]. In tions [45]. VEGF is a potent vaso-permeability factor. It affects
this procedure, stronger-intensity, large-sized burns (500 μm) endothelial tight junction proteins, resulting in extravasation of
are placed in the mid-peripheral retina in a 360 degree fashion fluid and retinal oedema. VEGF induces the phosphorylation
in one or two sessions. The treatment does not target new ves- of vascular endothelial cadherin, occludin and ZO-1, and thus
sels directly, and causes regression of new vessels in ∼6 weeks. causes a breakdown of the barrier [46]. VEGF also increases
The mechanism for new vessel regression is probably increased leukostasis in the retinal microvessels, and the sticky leukocytes
oxygen tension in the remaining retina, and thus decreased pro- may migrate via the interendothelial or transendothelial route
duction of angiogenic factors (such as VEGF). PRP laser ther- [47]. There are several drugs that target the molecule, VEGF.
apy has been established as a mainstay of treatment for PDR, but Drugs that directly inhibit the VEGF molecule include the
there are notable side effects that should be considered, includ- anti-VEGF aptamer, pegaptanib (Macugen; OSI Pharmaceuti-
ing the worsening of pre-existing macular oedema and impair- cals, Long Island, NY, USA), the monoclonal antibody fragment
ment of peripheral retina function and night vision [39]. The ranibizumab (Lucentis; Genentech, San Francisco, CA, USA),
reduction of these side effects with favourable clinical outcomes and the full-length antibody bevacizumab (Avastin; Genen-
has been (and will likely continue to be) the focus of evolv- tech). Other treatments include soluble VEGF receptor ana-
ing therapies. The administration of PRP laser therapy did not logues, VEGF-Trap (Regeneron, Tarrytown, NY, USA), small
fundamentally change until the introduction of the PASCAL in interfering RNAs bevasiranib (Opko Health, Miami, FL, USA),
recent years. The aim of this device was to shorten the proce- and rapamycin (Sirolimus; MacuSight, Union City, CA, USA).
dure time of PRP laser treatment and to decrease patient pain Anti-VEGF drugs are injected into the eye in the form of
by decreasing stimulation of the ciliary nerves in the choroid, intravitreal injections under topical anaesthesia as office pro-
while providing the same therapeutic outcomes [40]. The PAS- cedures. Currently, the indication for use of anti-VEGF agents
CAL requires a shorter duration of photocoagulation, but more in diabetic macular oedema is centre-involving diabetic macu-
power, which has indeed been shown to decrease patient pain lar oedema, while the indication for laser treatment is limited
with similar outcomes when compared with conventional PRP to non-centre-involving diabetic macular oedema.
[41]. Recently, the PETER PAN study [42] reported that a lower
power setting achieved similar results with less vision loss com-
pared with standard PRP settings. Additionally, it has been
Diabetic Macular Oedema
shown that single-session multi-spot PASCAL photocoagula- Ranibizumab is a monoclonal antibody that blocks all iso-
tion is as efficacious as multi-session single-spot photocoagula- forms of VEGF-A, and is ’affinity-enhanced’ to provide stronger
tion, and resulted in no difference in adverse macular oedema affinity to bind to VEGF-A. It is the only drug that has been
effects [43]. This finding has the potential to increase patient approved by the US Food and Drug Administration for use in
compliance and result in cost savings from reduced numbers patients with diabetic macular oedema (Figure 1). The READ-2
of visits. Study [48] compared focal/grid laser treatment with intrav-
Retina visualization technologies have also affected the itreal injection of ranibizumab only, as well as a combina-
treatment options for PDR and may have the potential to tion therapy of ranibizumab plus focal/grid laser. Three-year
make targeted retinal photocoagulation a viable treatment results of the study showed a mean improvement in visual

222 Das et al. Volume 17 No. 3 March 2015


DIABETES, OBESITY AND METABOLISM review article

Figure 1. Optical coherence tomography images of the study eye retina of a patient with diabetic macular oedema at baseline and 1, 3 and 6 months. The
patient received 0.3 mg of ranibizumab (adapted from [71]).

acuity from baseline of 10.3 letters compared with −1.6 let- focal/grid laser treatment for 12 months showed an improve-
ters in the laser group and +2.0 letters in the combination ment of eight letters with bevacizumab injections, whereas
therapy group. Two more studies also showed the beneficial the laser group lost a median of 0.5 letters [53]. Two-year
effects of intravitreal ranibizumab injections in patients with results of that study showed a similar improvement with
diabetic macular oedema. In the RISE and RIDE study [49], bevacizumab.
where patients were randomized to either two different doses Aflibercept (Eylea; Regeneron) is a soluble protein that
of intravitreal ranibizumab (0.3 and 0.5 mg) or sham injec- contains extracellular VEGF receptor sequences fused to an
tion, the proportion of patients showing >15 letter improve- immunoglobulin G molecule, and blocks all isoforms of VEGF
ment was 19% in the sham group compared with 37% in the as well as placental growth factor. The prolonged biological
0.3 mg ranibizumab group and 40% in the 0.5 mg ranibizumab activity of this drug offers the advantage of injections every
group after 36 months. Interestingly, that study reported a ben- second month rather than more frequent monthly injec-
eficial effect of ranibizumab in slowing down the progression tions. In the phase III VIVID-diabetic macular oedema and
of diabetic retinopathy and improvement of the severity of VISTA-diabetic macular oedema trials [54], patients receiv-
retinopathy. The Diabetic Retinopathy Clinical Research Net- ing aflibercept (2 mg monthly or every second month) had
work (DRCR), a National Institutes of Health-sponsored mul- a mean BCVA change from baseline of 12.5 and 11.1 letters,
ticentre, randomized clinical trial, concluded that intravitreal respectively, after 2 years, compared with a mean change from
ranibizumab with prompt or deferred laser was more effec- baseline in BCVA of 0.2 letters in patients receiving laser
tive for up to at least 1 year compared with prompt laser alone photocoagulation. Currently, the DRCR is evaluating protocol
for centre-involving diabetic macular oedema [50]. Three-year T, a head-to-head comparison of the efficacy and safety of
data from that study indicated that visual improvement was these three drugs, ranibizumab, bevacizumab and aflibercept
greater in the ranibizumab + deferred laser group (57% with in the treatment of patients with diabetic macular oedema
>10 letters improvement) than in the ranibizumab + prompt [55]. Until we have the results published from that pivotal
laser group (42% with >10 letters improvement), and thus study, ophthalmologists are left to make their own personal
suggested no benefit of earlier initiation of focal/grid laser choice of anti-VEGF drugs in patients with diabetic macular
for better visual outcome [51]. Although the first-line treat- oedema. In addition, there remains a large disconnect between
ment of centre-involving diabetic macular oedema is currently what the randomized clinical trials publish and what the oph-
anti-VEGF injections, focal/grid laser therapy can be added thalmologists actually do in their clinical practice in terms of
the number of intravitreal injections. The issue of which drug
to this anti-VEGF regimen if the oedema persists and is no
to choose and how frequently to inject after the initial three
longer improving with two consecutive injections after an ini-
monthly injections is still a matter of personal choice.
tial period of 24 weeks of monthly anti-VEGF injections [52].
Bevacizumab (Avastin; Genentech) is a full-length human-
ized monoclonal antibody, almost three times the size of Proliferative Diabetic Retinopathy
the ranibizumab molecule, which also blocks all isoforms of Anti-VEGF therapy has been found to be very effective in the
VEGF-A. It has been used as an off-label drug for the treat- rapid regression of retinal neovascularization seen in patients
ment of age-related macular degeneration, retinal vascular with PDR [56]; however, the effects of anti-VEGF agents appear
occlusions, PDR and diabetic macular oedema. Because of to be transient, and therefore PRP may still be necessary to
its much lower cost compared with other available drugs, allow more permanent regression of new vessels. Caution is
such as ranibizumab and aflibercept, bevacizumb has gained necessary in using anti-VEGF agents in those patients with
worldwide popularity among eye clinics for the treatment PDR with significant fibrovascular proliferation as these agents
of retinal vascular diseases. The BOLT study that compared may worsen traction retinal detachment. Currently, a phase
intravitreal injections of bavacizumab (1.25 mg, 6 weekly) with III prospective clinical trial is comparing prompt PRP with

Volume 17 No. 3 March 2015 doi:10.1111/dom.12384 223


review article DIABETES, OBESITY AND METABOLISM

Systemic Factor Control DIABETES

Polyol, AGE, PKC & Hexosamine


Hyperlipidemia
Pathway Activation

ROS

INFLAMMATION
Hypertension
Anti-VEGF Therapies
Growth Factors (VEGF),
(for center-involving DME)
Chemokines & Cytokines
(Ang-2, CCL2, TNF-α, IL-1β ) Steroids

Altered Blood-Retinal Barrier

Increased vascular permeability

Diabetic
Focal/Grid LASER Vitrectomy
Macular
(for non-center involving DME) (for Vitreo-macular traction)
Edema

Figure 2. Pathophysiology and current strategies of therapies in diabetic macular oedema. Systemic factor control targeting blood glucose, blood pressure
and lipids is still the ’gold standard’ treatment for diabetic retinopathy. Focal/grid laser is now indicated in those with non-centre-involving diabetic macular
oedema only. Anti-vascular endothelial growth factor (VEGF) intravitreal injections are the first line of treatment in patients with centre-involving diabetic
macular oedema. Steroids are reserved for those who are poor responders to anti-VEGF therapies. Finally, a small number of patients with vitreo-macular
traction are treated with vitrectomy surgery. Other potential novel target molecules are angiopoietin 2 (Ang-2), chemokine ligand 2 (CCL2), tumour necrosis
factor-𝛼 (TNF-𝛼) and interleukin-1𝛽 (IL-1𝛽). AGE, advanced glycation end-products; PKC, protein kinase C.

intravitreal ranibizumab and deferred PRP in patients with chemokines involved in the inflammatory cascade of diabetic
PDR [57]. macular oedema are susceptible to steroids, whereas inhibi-
tion of VEGF itself may not result in neutralization of these
Steroids molecules (Figure 2).
Inflammation plays an important role in the pathogenesis of The efficacy of intravitreal steroids in diabetic macular
diabetic retinopathy [58]. All the features of inflammation oedema has been shown in the DRCR Protocol I. In that trial,
[increased blood flow, increased vascular permeability, tissue the effect of intravitreal triamcinolone and laser was equivalent
oedema, leukostasis, microglial activation, macrophages and to that of ranibizumab and laser up to 24 weeks, and then
neutrophil infiltration, complement activation and increased the effect of triamcinolone gradually diminished because of
cytokines (VEGF, tumour necrosis factor -𝛼, interleukin)] have increased rates of cataract formation [50]. In the subgroup of
been reported in animal models as well as in human dia- patients with pseudophakia, the triamcinolone + laser treat-
betic retinopathy [59]. The inflammatory cytokines are upreg-
ment was superior to the laser-alone treatment and equivalent
ulated in the serum, vitreous and aqueous samples in sub-
to ranibizumab treatment [50].
jects with diabetic retinopathy. Inflammation can alter the BRB
In another randomized, multicentre 3-year long trial, the
by acting on the following steps in the cascade: (i) increased
expression of endothelial adhesion molecules such as ICAM1, FAME study [59], intravitreal inserts of fluocinolone acetonide
VCAM1, PECAM-1, and P-selectin; (ii) adhesion of leukocytes (0.2 and 0.5 μg/day) provided the substantial benefit of visual
to the endothelium (leukostasis); (iii) release of chemokines, improvement in patients with diabetic macular oedema; how-
and cytokines; (iv) alteration of adherens and tight junctional ever, almost all patients receiving fluocinolone acetonide had
proteins between the endothelial cells; and (v) infiltration of cataract formation, and the incidence of incisional glaucoma
leukocytes into the neuro-retina, resulting in the alteration of surgery was 4.8% (low dose) and 8.1% (high dose). Although
the BRB [58]. the US Food and Drug Administration did not approve this
drug for diabetic macular oedema because of its side effects,
Diabetic Macular Oedema it has recently been approved in UK. It is interesting to note
The beneficial effects of steroids in diabetic macular oedema that the FAME study showed enhanced benefits of using
are attributable to the fact that inflammatory cytokines and fluocinolone acetonide in chronic diabetic macular oedema

224 Das et al. Volume 17 No. 3 March 2015


DIABETES, OBESITY AND METABOLISM review article
(>3 years duration) compared with non-chronic diabetic mac- the Constellation System with ultra-speed cutters, duty cycle
ular oedema (<3 years duration) [60]. It has been hypothesized controls, improved fluidics and powerful endoillumination,
that the disease may be driven by VEGF early in the disease, and small-gauge surgery (23- and 25-gauge) has significantly
but in chronic diabetic macular oedema microenvironmen- improved visual outcomes in these patients. Extramacular trac-
tal changes necessitate the targeting of multiple mediator tion retinal detachment can be observed safely until it threatens
molecules with steroids. The beneficial effect of steroids in to involve the fovea. In patients with macula-involving trac-
diabetic macular oedema has further been reported with the tion retinal detachment, the traction can be relieved by vitrec-
use of slow-release intravitreal dexamethasone implant (Ozur- tomy using segmentation, delamination and en bloc delamina-
dex; Allergan Inc., Irvine, CA, USA) that releases sustained tion techniques. Currently, some surgeons prefer using preop-
levels of dexamethasone for 6 months [61]. Because of an erative anti-VEGF agents now 1 week before surgery so as to
increased rate of elevated intraocular pressure and cataract decrease intra-operative and postoperative haemorrhage from
formation with triamcinolone, the use of intravitreal steroids the neovascular membranes, reduce surgical time and improve
in clinical practice is currently reserved for those patients with visual acuity [64]; however, this might also increase the traction
pseudophakia who respond poorly to intravitreal anti-VEGF exerted by the epiretinal membranes.
therapy. The combination of anti-VEGF agents and steroids
may be more effective in certain patients with diabetic mac-
ular oedema who are difficult to control with anti-VEGF Future Directions
agents alone. Anti-VEGF Agents
As the VEGF level varies in the vitreous of patients with diabetic
Proliferative Diabetic Retinopathy macular oedema, it has been proposed that poor responders to
Although steroids may be effective in the treatment of neo- anti-VEGF therapy may need much higher doses of anti-VEGF
vascular age-related macular degeneration, these drugs are drugs because of their higher levels of VEGF. With this concept,
not used in retinal neovascularization, as seen in PDR. The a new study, READ-3 has examined the efficacy of ranibizumab
PRP laser is still the ’gold standard’ treatment for PDR, and at two different doses (0.5 and 2.0 mg) in patients with diabetic
anti-VEGF drugs are currently used as supplementary options; macular oedema. The 1-year results from that study, however,
this is being explored in ongoing trials, as mentioned above. show no significant difference in the effectiveness between these
All therapies for diabetic retinopathy are summarized in two different doses [65]. Other anti-VEGF approaches include
Table 2 [11–13,15,16,18,19,25,30,36–38,53,55–60]. targeting a central regulator, such as Raf kinase, mammalian
target of rapamyacin and the RTP 801 gene.

Surgical Therapies Extended Drug Delivery


Despite laser and pharmacotherapies, a small number of As the monthly intravitreal anti-VEGF injections are a treat-
patients with PDR and diabetic macular oedema undergo ment burden not only for the patients, but also for the
surgery. physicians, investigations into extended drug delivery are ongo-
ing, using bioerodable implants, bioerodable microspheres and
Diabetic Macular Oedema encapsulated cells. Recently, a phase I clinical trial using a
refillable, non-biodegradable long-term drug delivery implant
The majority of people with diabetic macular oedema benefit
for ranibizumab has been completed on 20 patients with wet
from pharmacotherapies (anti-VEGF and steroids) and laser
macular degeneration. The trial showed a constant mean
treatment. In selected patients with significant vitreo-macular
improvement of vision of 10 letters throughout 1 year [66].
traction in diabetic macular oedema, vitrectomy with peel-
ing of the inner limiting membrane can be combined with
or without indocyanine green dyes. In a multicentre DRCR Other Novel targets
study in patients with diabetic macular oedema and macu- As inflammation releases several cytokines and chemokines
lar traction, retinal thickening was reduced in most patients’ in diabetic macular oedema, it is possible that targeting
eyes, but visual acuity improved by 10 letters in 38% patients only VEGF may not be the best approach to treating dia-
only [62]. betic macular oedema. Other potential target molecules
include angiopoietin-2, tumour necrosis factor, interleukins,
Proliferative Diabetic Retinopathy proteinases, chemokines (CCL2 and CCL5) and kallikrein.

Persistent non-clearing vitreous haemorrhage and traction reti- Angiopoietin-2. Angiopoietin-2, a growth factor that binds to
nal detachment are two common indications for vitrectomy the endothelial receptor tyrosine kinase Tie-2, has been shown
surgery in PDR. The Diabetic Retinopathy Vitrectomy Study to be upregulated in retinas in an animal model of diabetes, and
showed a benefit of earlier surgery in patients with type 1 dia- increased angiopoietin-2 levels leads to increased retinal vascu-
betes and in those with active neovascularization [63]. The vit- lar permeability [67]. This pathway has been targeted in a recent
rectomy surgery involves a three-port technique, with removal ongoing clinical trial with a Tie-2 activator [angiopoietin-2
of vitreous, along with blood and endolaser photocoagula- antagonist; AKB 9778 (Aerpio Therapeutics, Blue Ash, OH,
tion similar to PRP laser treatment. The recent introduction of USA)] in patients with diabetic macular oedema.

Volume 17 No. 3 March 2015 doi:10.1111/dom.12384 225


Table 2. List of clinical trial results for prevention and intervention of diabetic retinopathy using systemic factor control, laser therapy, anti-vascular endothelial growth factor and steroid injections, and
combination therapies

226 Das et al.


Prevention/intervention of diabetic retinopathy

Systemic Laser (conventional, Combined


Outcome affected glycaemic control Blood pressure control Lipid control SDM, PASCAL) Steroid therapy Anti-VEGF therapy (laser and anti-VEGF)

Severity of HbA1c <6% in patients 144/82 mmHg mean blood Decreases in progression PASCAL success rate Triamcinolone injections led 3.9% of ranibizumab-treated
retinopathy with type 1 diabetes pressure versus of diabetic retinopathy significantly higher (28/30) to a 88% relative decrease (0.5 mg injection) patients
reduced risk of diabetic 154/87 mmHg led to a 34% (6.5 vs. 10.2%) [13] compared with in fluorescein leakage at progressed to PDR versus
retinopathy by 76%, reduction in progression of conventional laser for PDR 12 months [56] 11.5% of patients treated
review article

decreased progression diabetic retinopathy [12] Decreases in progression (20/30) [53] with sham treatment [37]
of existing retinopathy of diabetic retinopathy
by 54%, and reduced Prevents development of in patients with PASCAL multi-spot 20 ms 1% of patients receiving
development of severe diabetic retinopathy in pre-existing diabetic with mean power of ranibizumab (0.5 mg
NPDR by 47% [11] patients with type 1 retinopathy (3.1 vs. 157 mW compared with injection) underwent PRP
diabetes (25 vs. 31%) [15] 14.6%) [18] 20 ms PASCAL with mean versus 14% of patients in
HbA1c of 7.0 versus 7.9% power of 280 mW sham group [37]
led to a 25% reduced No significant effect on produced similar outcomes
rate of laser progression of diabetic [30]
photocoagulation for retinopathy in patients
retinopathy [12] with type 1 or type 2
diabetes [15,16]
Tight control decreases
progression of diabetic Regression of diabetic
retinopathy (7.3 vs. retinopathy in 19% of
10.4%) [13] participants versus 14%
taking placebo in people
with type 2 diabetes [16]
Visual acuity Decreases risk of legal Tight blood pressure control Fenofibrate has no Argon conventional laser Visual acuity is transiently Bevacizumab and Ranibizumab combined with
blindness by 16% [12] (mean pressure of significant effect reduces risk of visual acuity better with triamcinolone ranibizumab improve prompt focal laser or
144/82 mmHg) versus versus placebo [13] loss by 50% in clinically injections in the first visual acuity in diabetic deferred focal
Prevents visual acuity loss mean pressure of significant macular 4 months compared with macular oedema by an photocoagulation resulted
(23.8 vs. 26.3%) [13] 154/87 mmHg led to 47% oedema [19] focal laser therapy; focal average of 1–2 Snellen in greater improvement of
risk reduction of vision loss laser group was found to chart lines [38,60] visual acuity than focal
[12] SDM equally effective in have better visual acuity at laser alone [38]
correcting visual acuity in follow-up of 16 months and Ranibizumab improved visual
Intensive-treatment blood diabetic macular oedema 3 years. A total of 83% of acuity in diabetic macular
pressure control (mean versus conventional green patients who received 4 mg oedema from baseline by
systolic pressure of laser [55] triamcinolone injection 10.3 ± 9.1 letters versus a
117 mmHg) versus had developed cataracts at decline by 1.4 ± 14.2 letters
standard treatment blood PRP reduced the rate of 3-year follow-up [57] with sham [60]
pressure control (mean severe vision loss by 50% in
systolic pressure of PDR [25] Visual acuity is increased >5 Ranibizumab (0.5 mg
133 mmHg) led to rates of letters in 56% of eyes injection) improved visual
moderate vision loss of receiving triamcinolone acuity by ≥15 letters in
27.7% and 24.7%, versus 26% of eyes 45.7% patients versus
respectively [13] receiving placebo [58] 12.3% patients treated with
sham injection [37]
DIABETES, OBESITY AND METABOLISM

Volume 17 No. 3 March 2015


Table 2. Continued

Volume 17 No. 3 March 2015


Prevention/intervention of diabetic retinopathy

Systemic Laser (conventional, Combined


Outcome affected glycaemic control Blood pressure control Lipid control SDM, PASCAL) Steroid therapy Anti-VEGF therapy (laser and anti-VEGF)

Central retinal Fenofibrate decreases SDM equally effective as Intravitreal triamcinolone Ranibizumab (0.5 mg Ranibizumab plus focal/grid
thickness progression of macular conventional green laser injections are superior to injection) decreased laser reduced foveal
oedema by 34% (optical coherence posterior sub-tenon macular oedema by thickness more than
DIABETES, OBESITY AND METABOLISM

compared with tomography; measured capsule injections in 270.7 μm compared with a focal/grid laser alone in
placebo [18] central retina thickness of reducing foveal thickness decrease of 125.8 μm in DME [38,60]
255 vs. 248.9 μm, [59] sham groups [37]
respectively) with less
scarring [55] Intravitreal triamcinolone Ranibizumab injections
injections decreased decreases foveal thickness
PASCAL laser equally macular thickness by a less than laser therapy in
effective in treating mean of 125 μm as DME [36]
clinically significant compared with a mean
macular oedema versus decrease of 71 μm with Forty-four patients receiving
conventional laser (28/30 placebo [58] Ranibizumab (0.5 mg inj.)
vs. 27/30) [53] received macular laser
treatments versus 94
PASCAL laser reduced patients in sham group [37]
frequency domain-optical
coherence tomography by
20+/−21 um at 3 months
[24]

SDM and PASCAL


(double-frequency
neodymium YAG laser
532 nm) have equally
efficacy in reducing
diabetic macular oedema
[54]

VEGF, vascular endothelial growth factor; SDM, subthreshold diode micropulse laser; PASCAL, patterned scanning laser; HbA1c, glycated haemoglobin; NPDR, non-proliferative diabetic retinopathy; PDR, proliferative diabetic retinopathy;
PRP, panretinal photocoagulation.

doi:10.1111/dom.12384 227
review article
review article DIABETES, OBESITY AND METABOLISM

Tumour Necrosis Factor. Tumor necrosis factor-𝛼 has been when age at diagnosis is less than 30 years. Arch Ophthalmol 1984; 102:
implicated in many inflammatory diseases, including rheuma- 520–526.
toid arthritis, ankylosing spondylitis, psoriatic arthritis, plaque 5. Klein R, Klein BEK, Moss SE, Davis MD, DeMets DL. The Wisconsin Epidemiologic
psoriasis and Crohn’s disease, and a monoclonal antibody, Study of Diabetic Retinopathy. III. Prevalence and risk of diabetic retinopathy
when age at diagnosis is 30 or more years. Arch Ophthalmol 1984; 102:
infliximab, has been approved for treating these disorders. A
527–532.
double-blind, randomized, placebo-controlled, crossover study
in patients with diabetic macular oedema (persisting after 6. Early Treatment Diabetic Retinopathy Study Research Group. Fundus photo-
graphic risk factors for progression of diabetic retinopathy. ETDRS Report # 12.
two sessions of laser photocoagulation) showed significant
Ophthalmology 1991; 98(Suppl): 823–833.
improved visual acuity and reduction in retinal thickness with
7. Kuwabara T, Cogan DG. Retinal vascular patterns. VI. Mural cells of the retinal
intravenous infliximab (5 mg/kg) [68].
capillaries. Arch Ophthalmol 1963; 69: 492–502.
Chemokines. One of the most common chemokines that is sig- 8. Das A, Frank RN, Weber M et al. ATP causes retinal pericytes to contract in vitro.
nificantly elevated in serum and vitreous in diabetic retinopa- Exp Eye Res 1988; 46: 349–362.
thy has been identified as the chemokine ligand, CCL2. CCL2, 9. Frank RN. Diabetic retinopathy. N Engl J Med 2004; 350: 48–58.
also known as monocyte chemotactic protein-1, is considered 10. Brownlee M. The pathobiology of diabetic complications: a unifying mechanism.
to play an important role in vascular inflammation by induc- Diabetes 2005; 54: 1615–1625.
ing leukocyte recruitment and activation. Animal studies have 11. Das A, McGuire PG. Retinal and choroidal angiogenesis: Pathophysiology and
shown the role of the CCL2 gene in alteration of the BRB in strategies for inhibition. Prog Retin Eye Res 2003; 22: 721–748.
diabetic retinopathy [69]. A CCR2/CCR5 receptor antagonist 12. The Diabetes Control and Complications Trial Research Group. The effect
(Pfizer, New York, NY, USA) is currently being tested in an of intensive treatment of diabetes on the development and progression of
ongoing clinical trial in patients with diabetic macular oedema. long-term complications in insulin-dependent diabetes mellitus. N Engl J Med
1993; 329: 977–986.
Kallikrein–kinin Inhibitors. Intraocular activation of the plasma 13. King P, Peacock I, Donnelly R. The UK Prospective Diabetes Study (UKPDS):
kallikrein–kinin pathway may contribute to increased retinal clinical and therapeutic implications for Type 2 diabetes. Br J Clin Pharmacol
vascular permeability in many patients with diabetic macular 1999; 48: 643–648.
oedema [70]. A phase I single-ascending dose study to inves- 14. The ACCORD, Study Group ACCORD, Eye Study Group. Effects of medical
tigate the safety and tolerability of the novel plasma kallikrein therapies on retinopathy progression in type 2 diabetes. N Engl J Med 2010;
inhibitor, KVD001 (intravitreal) in people with diabetic macu- 363: 233–244.
lar oedema is in progress. 15. Boussageon R, Bejan-Angoulvant T, Saadatian-Elahi M et al. Effect of intensive
Although the anti-VEGF effect is beneficial in PDR for glucose lowering treatment on all cause mortality, cardiovascular death, and
regression of new vessels, the effect is much less robust in dia- microvascular events in type 2 diabetes: meta-analysis of randomised controlled
betic macular oedema. Multiple, frequent monthly anti-VEGF trials. Br Med J 2011; 343: d4169.

injections are needed in patients with diabetic macular oedema 16. Muir KW, Grubber J, Mruthyunjaya P, McCant F, Bosworth HB. Progression of
to reduce the macular oedema. Nevertheless, a large number diabetic retinopathy in the hypertension intervention nurse telemedicine study.
JAMA Ophthalmol 2013; 131: 957–958.
of these patients respond poorly to these anti-VEGF drugs.
For such patients, steroids may be an alternative approach as 17. Chaturvedi N, Porta M, Klein R et al. Effect of candesartan on prevention
(DIRECT-Prevent 1) and progression (DIRECT-Protect 1) of retinopathy in type 1
a second-line treatment, with careful consideration of poten-
diabetes: randomised, placebo-controlled trials. Lancet 2008; 372: 1394–1402.
tial side effects. It is possible that other molecules in addition to
18. Sjølie AK, Klein R, Porta M et al. Effect of candesartan on progression and
VEGF play a key role in the inflammatory process in diabetic
regression of retinopathy in type 2 diabetes (DIRECT-Protect 2): a randomised
macular oedema. A combination therapy with inhibitors of placebo-controlled trial. Lancet 2008; 372: 1385–1393.
these molecules combined with laser treatment or anti-VEGF
19. Benarous R, Sasongko MB, Qureshi S et al. Differential association of serum
agents may be more effective in treating macular oedema in
lipids with diabetic retinopathy and diabetic macular edema. Invest Ophthalmol
people with diabetes in the future. Vis Sci 2011; 52: 7464–7469.
20. Keech AC, Mitchell P, Summanen PA et al. Effect of fenofibrate on the need for
laser treatment for diabetic retinopathy (FIELD study): a randomised controlled
Conflict of Interest trial. Lancet 2007; 370: 1687–1697.
The authors have no conflicts of interest to declare. 21. Davis MD, Fisher MR, Gangnon RE et al. Risk factors for high-risk proliferative
diabetic retinopathy and severe visual loss: early Treatment Diabetic Retinopa-
thy Study Report #18. Invest Ophthalmol Vis Sci 1998; 39: 233–252.
References
22. West SD, Groves DC, Lipinski HJ et al. The prevalence of retinopathy in men with
1. International Diabetes Federation. Diabetes: facts and figures 2013. Avail- type 2 diabetes and obstructive sleep apnoea. Diabet Med 2010; 27: 423–430.
able from URL: http://www.idf.org/worlddiabetesday/toolkit/gp/facts-figures.
23. Klein R, Klein B. The epidemiology of diabetic retinopathy. In: Ryan SJ, ed. Retina
Accessed 19 September 2014.
II. London: Elsevier, 2013; 907–924.
2. Yau JW, Rogers SL, Kawasaki R et al. Global prevalence and major risk factors of 24. Early Treatment Diabetic Retinopathy Study Research Group. Photocoagulation
diabetic retinopathy. Diabetes Care 2012; 35: 556–564. for diabetic macular edema: early treatment diabetic retinopathy study report
3. Klein R. Overview of epidemiologic studies of diabetic retinopathy. Ophthalmic number 1. Arch Ophthalmol 1985; 103: 1796–1806.
Epidemiol 2007; 14: 179–183. 25. Duh EJ, Yang HS, Suzuma I, Miyagi M et al. Pigment epithelium-derived fac-
4. Klein R, Klein BEK, Moss SE, Davis MD, DeMets DL. The Wisconsin Epidemiologic tor suppresses ischemia-induced retinal neovascularization and VEGF-induced
Study of Diabetic Retinopathy. II. Prevalence and risk of diabetic retinopathy migration and growth. Invest Ophthalmol Vis Sci 2002; 43: 821–829.

228 Das et al. Volume 17 No. 3 March 2015


DIABETES, OBESITY AND METABOLISM review article
26. Miura Y, Treumer F, Klettner A et al. VEGF and PEDF secretions [68] over time 46. Antonetti DA, Barber AJ, Hollinger LA, Wolpert EB, Gardner TW. Vascular
following various laser irradiations on an RPE organ culture. Invest Ophthalmol endothelial growth factor induces rapid phosphorylation of tight junction
Vis Sci 2010; 51: 469. proteins occludin and zonula occluden 1. A potential mechanism for vascu-
27. Luttrull JK, Musch DC, Mainster MA. Subthreshold diode micro- pulse photoco- lar permeability in diabetic retinopathy and tumors. J Biol Chem 1999; 274:
agulation for the treatment of clinically significant diabetic macular oedema. Br 23463–23467.
J Ophthalmol 2005; 89: 74–80. 47. Joussen AM, Murata T, Tsujikawa A, Kirchhof B, Bursell SE, Adamis AP.
28. Laursen ML, Moeller F, Sander B, Sjoelie AK. Subthreshold micropulse diode laser Leukocyte-mediated endothelial cell injury and death in the diabetic retina. Am
in patients with diabetic macular edema. Br J Ophthalmol 2004; 88: 1173–1179. J Pathol 2001; 158: 147–152.
29. Sivaprasad S, Dorin G. Subthreshold diode micropulse photocoagulation for the 48. Do DV, Nguyen QD, Khwaja AA et al. Ranibizumab for edema of the Macula
treatment of diabetic macular edema. Expert Rev Med Devices 2012; 9: 189–197. in Diabetes study: 3 year outcomes and the need for prolonged frequent
30. Figueira J, Khan J, Nunes S et al. Prospective randomised controlled treatment. JAMA Ophthalmol 2013; 131: 139–145.
trial comparing sub-threshold micropulse diode laser photocoagulation 49. Brown DM, Nguyen QD, Marcus DM et al. RIDE and RISE Research Group.
and conventional green laser for clinically significant diabetic macular oedema. Long-term outcomes of ranibizumab therapy for diabetic macular edema: the
Br J Ophthalmol 2009; 93: 1341–1344. 36-month results from two phase III trials: RISE and RIDE. Ophthalmology 2013;
31. Vujosevic S, Bottega E, Casciano M et al. Microperimetry and fundus autofluo- 120: 2013–2022.
rescence in diabetic macular edema: subthreshold micropulse diode laser versus 50. The Diabetic Retinopathy Clinical Research Network. Randomized trial evalu-
modified early treatment diabetic retinopathy study laser photocoagulation. ating ranibizumab plus prompt or deferred laser or triamcinolone plus prompt
Retina 2010; 30: 908–916. laser for diabetic macular edema. Ophthalmology 2010; 117: 1064–1077.
32. Lavinsky D, Cardillo JA, Melo LA Jr et al. Randomized clinical trial evaluating 51. Diabetic Retinopathy Clinical research Network. Intravitreal ranibizumab for
mETDRS versus normal or high-density micropulse photocoagulation for dia- diabetic macular edema with prompt versus deferred laser treatment: three year
betic macular edema. Invest Ophthalmol Vis Sci 2011; 52: 4314–4323. randomized trial results. Ophthalmology 2012; 119: 2312–2318.
33. Muqit MM, Henson DB, Young LB et al. Laser tissue interactions. Ophthalmology
52. Diabetic Retinopathy Clinical Research Network; Writing Committee, Aiello LP
2010; 117: 2039.
et al. Rationale for the diabetic retinopathy clinical research network treatment
34. Muqit MM, Gray JC, Marcellino GR et al. Barely visible 10-millisecond pascal protocol for center-involved diabetic macular edema. Ophthalmology 2011;
laser photocoagulation for diabetic macular edema: observations of clinical 118: e5–e14.
effect and burn localization. Am J Ophthalmol 2010; 149: 979–986.
53. Michaelides M, Kaines A, Hamilton RD et al. A prospective randomized trial
35. Kozak I, Oster SF, Cortes MA et al. Clinical evaluation and treatment accuracy of intravitreal bevacizumab or laser therapy in the management of diabetic
in diabetic macular edema using navigated laser photocoagulator NAVILAS. macular edema (BOLT study) 12-month data: report 2. Ophthalmology 2010;
Ophthalmology 2011; 118: 1119–1124. 117: 1078–1086.
36. Barteselli G, Kozak I, El-Emam S, Chhablani J, Cortes MA, Freeman WR. 12-month 54. American Society of Retina Specialists (ASRS) News. Industry news – clinical
results of the standardised combination therapy for diabetic macular oedema: updates. Two-year results of phase 3 VISTA trial of Aflibercept for DME treatment
intravitreal bevacizumab and navigated retinal photocoagulation. Br J Ophthal-
show sustained vision improvement. Available from URL: http://www.asrs.org.
mol 2014; 98: 1036–1041.
Accessed 19 September 2014.
37. Diabetic Retinopathy Study Research Group. Photocoagulation treatment of
55. Diabetic Retinopathy Clinical Research Network. A comparative effectiveness
proliferative diabetic retinopathy. Clinical application of Diabetic Retinopathy
study of intravitreal aflibercept, bevacizumab and ranibizumab for diabetic mac-
Study (DRS) findings, DRS Report Number 8. Ophthalmology 1981; 88: 583–600.
ular edema. Available from URL: http://drcrnet.jaeb.org/Studies.aspx. Accessed
38. Chew EY, Ferris FL, Csaky KG etal. The long-term effects of laser photocoag- 4 June 2014.
ulation treatment in patients with diabetic retinopathy (the early treatment
56. Avery RL, Pearlman J, Pieramici DJ et al. Intravitreal bevacizumab (Avastin) in
diabetic retinopathy follow-up study). Ophthalmology 2003; 110: 1683–1689.
the treatment of proliferative diabetic retinopathy. Ophthalmology 2006; 113:
39. Fong DS, Girach A, Boney A. Visual side effects of successful scatter laser 1695–1705.
photocoagulation surgery for proliferative diabetic retinopathy: a literature
review. Retina 2007; 27: 816–824. 57. Diabetic Retinopathy Clinical Research Network. Prompt panretinal pho-
tocoagulation versus intravitreal ranibizumab with deferred panretinal
40. Blumenkranz MS, Yellachich D, Andersen DE et al. Semiautomated patterned
photocoagulation for proliferative diabetic retinopathy. Available from URL:
scanning laser for retinal photocoagulation. Retina 2006; 26: 370–376.
http://clinicaltrials.gov/show/NCT01489189. Accessed 19 September 2014.
41. Al-Hussainy S, Dodson PM, Gibson JM. Pain response and follow-up of patients
58. Rangasamy S, McGuire P, Das A. Diabetic retinopathy and inflammation: novel
undergoing panretinal laser photocoagulation with reduced exposure times. Eye
therapeutic targets. Middle East Afr J Ophthalmol 2012; 19: 52–59.
2008; 22: 96–99.
59. Campochiaro P, Brown DM, Pearson A et al., FAME Study Group. Sustained
42. Muqit MM, Young LB, McKenzie R et al. Pilot randomised clinical trial of Pascal
delivery fluocinolone acetonide vitreous inserts provide benefit for at least
TargETEd Retinal versus variable fluence PANretinal 20 ms laser in diabetic
retinopathy: PETER PAN Study. Br J Ophthalmol 2013; 97: 220–227. 3 years in patients with diabetic macular edema. Ophthalmology 2012; 110:
2125–2132.
43. Muqit MMK, Marcellino GR, Henson DB et al. Single-session vs multiple-session
pattern scanning laser panretinal photocoagulation in proliferative diabetic 60. Cunha-Vaz J, Ashton P, Iezzi R et al., FAME Study Group. Sustained delivery
retinopathy. Arch Ophthalmol 2010a; 128: 525–533. fluocinolone acetonide vitreous implants: long-term benefit in patients with
chronic diabetic macular edema. Ophthalmology 2014; 119: 2125–2132.
44. Muqit MM, Marcellino GR, Henson DB et al. Optos-guided pattern scan laser
(Pascal)-targeted retinal photocoagulation in proliferative diabetic retinopathy. 61. Boyer D. Ozurdex diabetic macular edema study. Paper presented at the Annual
Acta Ophthalmol 2013; 91: 251–258. Meeting of the America Academy of Ophthalmology, New Orleans, LA, 16–19
November 2013.
45. Funatsu H, Yamashita H, Ikeda T, Nakanishi Y, Kitano S, Hori S. Angiotensin II
and vascular endothelial growth factor in the vitreous fluid of patients with 62. Diabetic Retinopathy Clinical Research Network. Vitrectomy outcomes in eyes
diabetic macular edema and other retinal disorders. Am J Ophthalmol 2002; with diabetic macular edema and vitreomacular traction. Ophthalmology 2010;
133: 537–543. 117: 1087–1093.

Volume 17 No. 3 March 2015 doi:10.1111/dom.12384 229


review article DIABETES, OBESITY AND METABOLISM

63. Diabetic Retinopathy Vitrectomy Study. Early vitrectomy for severe vitreous 69. Das A, Rangasamy S, Maestas J, McGuire P. CC Chemokines play an important
hemorrhage in diabetic retinopathy. Report No. 5. Arch Ophthalmol 1990; 108: role in alteration of the blood-retinal barrier in diabetes. ARVO Abstract ARVO
958–964. Meeting, Ft Lauderdale, FL, 2011, Program# 997.
64. Ossadon P, Fagan XJ, Lifshitz T, Levy J. A review of anti-VEGF agents for 70. Feener EP. Plasma kallikrein and diabetic macular edema. Curr Diab Rep 2010;
proliferative diabetic retinopathy. Eye 2014; 28: 510–520. 10: 270–275.
65. Nguyen Q. Higher anti-VEGF approach. Presented at the World Ophthalmology 71. Chun DW, Heier JS, Topping TM et al. A pilot study of multiple intravitreal
Congress, Tokyo, Japan, 2–6 April 2014. injections of ranibizumab in patients with center-involving clinically significant
66. Rubio R. Long-acting anti-VEGF delivery: How close are we? Presented at the diabetic macular oedema. Ophthalmology 2006; 113: 1706–1712.
World Ophthalmology Congress, Tokyo, Japan, 2–6 April 2014. 72. American Academy of Ophthalmology. 2002. Available from URL:
67. Rangasamy S, Srinivasan R, Maestas J, McGuire P, Das A. A potential role http://one.aao.org/asset.axd?id=4b0447c9-24c6-411e-a888-3081553693d2.
of angiopoietin-2 in the alteration of the blood-retinal barrier in diabetic Accessed 19 September 2014.
retinopathy. Invest Ophthalmol Vis Sci 2011; 52: 3784–3791.
68. Sfikakis P, Grigoropoulos V, Emfietzoglou I et al. Infliximab for diabetic mac-
ular edema refractory to laser photocoagulation. Diabetes Care 2010; 33:
1523–1528.

230 Das et al. Volume 17 No. 3 March 2015

You might also like