Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

PRIMER

Diabetic retinopathy
Tien Y. Wong1,2, Chui Ming Gemmy Cheung1,2, Michael Larsen3, Sanjay Sharma4
and Rafael Simó5
Abstract | Diabetic retinopathy (DR) is a common complication of diabetes mellitus and is a major
cause of vision loss in middle-aged and elderly people. One-third of people with diabetes have DR.
Severe stages of DR include proliferative DR, caused by the abnormal growth of new retinal blood
vessels, and diabetic macular oedema, in which there is exudation and oedema in the central part of
the retina. DR is strongly associated with a prolonged duration of diabetes, hyperglycaemia and
hypertension. It is traditionally regarded as a microvascular disease, but retinal neurodegeneration is
also involved. Complex interrelated pathophysiological mechanisms triggered by hyperglycaemia
underlie the development of DR. These mechanisms include genetic and epigenetic factors, increased
production of free radicals, advanced glycosylation end products, inflammatory factors and vascular
endothelial growth factor (VEGF). Optimal control of blood glucose and blood pressure in individuals
with diabetes remains the cornerstone for preventing the development and arresting the progression
of DR. Anti-VEGF therapy is currently indicated for diabetic macular oedema associated with vision
loss, whereas laser photocoagulation prevents severe vision loss in eyes with proliferative DR.
These measures, together with increasing public awareness and access to regular screening for
DR with retinal photography, and the development of new treatments to address early disease stages,
will lead to better outcomes and prevent blindness for patients with DR.

Diabetes mellitus is a global epidemic with profound A pooled meta-analysis11 of 35 population-based (mostly
morbidity 1. Diabetes leads to a range of complications from developed countries) studies reported that more
grouped into macrovascular (large blood vessel) compli- than 90 million individuals are estimated to have DR, with
cations, such as cardiovascular disease and stroke, and 21 million having DME and 17 million proliferative DR.
microvascular (small blood vessel) complications, such There has been considerable progress in understand-
as kidney disease. Diabetic retinopathy (DR) is a specific ing the key pathogenetic factors involved in the develop­
microvascular complication of diabetes and is the lead- ment of DR. Although DR is traditionally regarded as a
ing cause of vision loss in the general population in many microvascular disease, growing evidence supports the
countries, including the adult working population and idea that retinal neurodegeneration is an early event in
the elderly 2–5. Patients with severe degrees of DR have the pathogenesis of DR. For example, a reduction in the
impaired quality of life, reduced physical, emotional and oscillatory potential in electroretinogram tests is the first
social well-being, and use many health-care resources6. ­m easurable change in retinal function in patients
Without treatment, DR progresses from mild, with diabetes12.
non-proliferative DR to moderate and severe non-­ Past epidemiological studies and clinical trials have
proliferative DR before the occurrence of proliferative shown that the development risk and progression of
DR, in which there is growth of abnormal new retinal DR can be reduced by controlling blood glucose levels
blood vessels. Concurrently, at any stage of retinopathy, and blood pressure2,13. Landmark clinical trials in the
Correspondence to T.Y.W.
Singapore Eye Research
with exudation and oedema at the macula (the central 1980s and 1990s have also demonstrated that timely
Institute, Singapore National focal point of the retina), patients may also develop laser photo­coagulation can prevent moderate to severe
Eye Centre, 11 Third Hospital ­diabetic macular oedema (DME) (FIG. 1; TABLE 1). vision loss in patients with DME or proliferative DR2,13–15.
Avenue, Singapore 168751, Although DR is present in approximately 30% of In the 2000s, the introduction of intraocular adminis-
Singapore.
individuals with diabetes, only 5–10% may have the tration of anti-vascular endothelial growth factor (anti-
wong.tien.yin@snec.com.sg
sight-threatening stages of proliferative DR and DME7,8. VEGF) agents and fast, non-invasive imaging in the
Article number: 16012 However, the individual lifetime risk of DR is 50–60% in form of optical coherence tomography (OCT) further
doi:10.1038/nrdp.2016.12
Published online 17 March 2016; a person with type 2 diabetes mellitus (T2DM) and up to revolutionized the management of DME, resulting in
corrected online 7 April 2016 90% in those with type 1 diabetes mellitus (T1DM)9,10. the possibility of reversing vision loss2,3,15,16. Anti-VEGF

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 1


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Author addresses Epidemiology


Prevalence and incidence of DR
1
Singapore Eye Research Institute, Singapore National DR occurs in people with either T1DM or T2DM.
Eye Centre, 11 Third Hospital Avenue, Singapore 168751, Classical epidemiological studies in the field of ophthal-
Singapore.
mology, such as the Wisconsin Epidemiologic Study
2
Duke–NUS Medical School, National University of
of Diabetic Retinopathy (WESDR), provided detailed
Singapore, Singapore.
3
Department of Clinical Medicine, University of long-term longitudinal data on the prevalence of DR,
Copenhagen, Copenhagen, Denmark. DME and proliferative DR in populations with T1DM or
4
Departments of Ophthalmology and Epidemiology, T2DM9,10,23,24. The WESDR reported that ~75% of people
Queen’s University, Kingston, Ontario, Canada. with diabetes developed DR 10 years after diagnosis, and
5
Vall d’Hebron Research Institute and CIBERDEM (ISCIII), for those with DR at baseline, approximately two-thirds
Barcelona, Spain. developed more severe stages of DR; 20% developed
proliferative DR or DME25. Incidence data from other
therapy for DME has been shown to improve quality of cohorts in people with diabetes in the United Kingdom,
life17 and reduce vision impairment 18; such therapy is Australia and Asia26–31 have become recently available,
also cost-­effective19. New clinical trials now suggest that and these studies included white people8,32–34 as well as
anti-VEGF therapy may also be useful for proliferative other racial and ethnic groups (for example, African
DR20. Finally, from a public health perspective, studies American and Hispanic people)7,35. Similarly, prevalence
have shown that regular widespread screening of per- estimates of between 20% and 50% have been reported
sons with diabetes by digital retinal photography enables in other countries, notably in Europe36,37 and Asia38–42.
the identification of patients with mild DR to facilitate Recent epidemiological studies have shown a r­ ising
early intervention21,22. prevalence of DME over time. Consequently, DME
In this Primer, we discuss trends in the epidemiol- has overtaken proliferative DR as the most common
ogy and risk factors of DR, the underlying pathophysio­ cause of vision impairment in T2DM. In the recent US
logical mechanisms, the classification and diagnosis, the National Health and Nutrition Examination Survey,
treatment options (including screening), and the impact DME was twice as common as proliferative DR among
of DR on quality of life. people with diabetes8. This trend may be related to the

Non-proliferative diabetic retinopathy   Pre-proliferative retinopathy


Microaneurysms form in the blood vessels of the Changes are increasingly severe and
eye, which can burst to leak blood widespread, and include bleeding into the retina

Fovea
centralis Macula lutea
Pupil Macula
Optic nerve
Optic disc
Central vein
of the retina
Retina Central artery
Light Sclera of the retina
Choroid

Hard
exudates

Cornea Lens Retina


Oedema
Scar tissue
Haemorrhage

Diabetic macular oedema  Proliferative retinopathy


Vascular leakage and accumulation of plasma New blood vessels and scar tissue form on
constituents in the macula the retina, causing loss of vision

Figure 1 | Clinical stages and main pathogenetic events of diabetic retinopathy. The first pathological
Nature stagePrimers
Reviews | Disease that
can be identified in a fundoscopic examination is non-proliferative diabetic retinopathy (DR), which can be divided into
mild, moderate or severe non-proliferative DR. Diabetic macular oedema can be considered a particular subtype of
non-proliferative DR in which vascular leakage involves the macula. Pre-proliferative or severe non-proliferative DR is
characterized by capillary closures and non-perfused areas. The typical fundoscopic finding is the presence of
‘cotton wool’ spots and intraretinal microvascular abnormalities. Proliferative DR, in which hypoxia has an essential role,
is the final stage of DR and its hallmark is the development of neovascularization. Haemovitreous (that is, bleeding
in the vitreous cavity) and retinal detachment are advanced complications of proliferative DR.

2 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Table 1 | Classification of diabetic retinopathy and diabetic macular oedema triglyceride) and DR has not been consistently docu-
mented in studies52,53. However, increasing evidence
Condition Clinical findings*
supports the idea that non-traditional lipid measures
Diabetic retinopathy (DR) (for example, apolipoproteins A and B) are stronger
No DR No abnormalities risk markers of DR than total cholesterol and tri-
Mild non- Microaneurysms only glyceride levels54,55. Other systemic risk factors for
proliferative DR DR include diabetic nephropathy 56, obesity 57, anae-
mia 58, and markers of systemic inflammation and
Moderate non- Any of the following: microaneurysms, retinal dot and blot
proliferative DR haemorrhages, hard exudates or cotton wool spots; no signs of endothelial dysfunction55,59.
severe non-proliferative DR Ocular risk factors for DR include previous cataract
Severe non- Any of the following: intraretinal haemorrhages (≥20 in each of surgery, which is associated with the progression of DR
proliferative DR 4 quadrants), definite venous beading (in 2 quadrants) or intraretinal and the development of DME60,61, and a myopic refract­
microvascular abnormalities (in 1 quadrant); no signs of proliferative DR ive error (that is, near sightedness), which appears to
Proliferative DR One or more of the following: neovascularization, vitreous or protect against DR62,63.
preretinal haemorrhages In terms of genetic factors, heritability estimates of
Diabetic macular oedema (DME) 25–50% have been reported in patients with T1DM
who have proliferative DR64,65; however, candidate and
No DME No apparent retinal thickening or hard exudates in the posterior pole
genome-wide association studies have discovered few
Mild DME Some retinal thickening or hard exudates in the posterior pole but new genetic markers for DR in T2DM66,67. Epigenetic
outside the central subfield of the macula (diameter 1,000 microns) mechanisms (that is, DNA methylation, histone modifi­
Moderate DME Retinal thickening or hard exudates within the central subfield of the cations of chromatin and non-coding RNAs) have been
macula, but not involving the centre of the macula suggested to play a part in the pathogenesis of DR68,69.
Severe DME Retinal thickening or hard exudates involving the centre of the macula This hypothesis is supported by observations in clinical
Based on data from REF. 106. *Findings are based on dilated ophthalmoscopy for DR and on trials of a lower risk for DR related to the persistence of
dilated binocular, stereoscopic ophthalmoscopy for DME. beneficial effects (‘metabolic memory’) in patients with
T1DM or T2DM who had previously received sustained
declining prevalence of proliferative DR, which is due intensive glycaemic treatment 70,71.
to the improved systemic management of diabetes.
Concurrently, the incidence of DR‑related vision loss has Mechanisms/pathophysiology
declined over the past three decades, most prom­inently Pathogenesis of DR
in the United States43–45. Again, these observations reflect Extensive research has provided insight into the molecu­
a combination of improved systemic management of lar pathways of DR2,15. Metabolic pathways triggered by
diabetes, enhanced public awareness and access to hyperglycaemia in diabetes, such as the polyol and the
screening 44. Nevertheless, in the context of an expanding hexosamine pathways, the de novo synthesis of diacyl­
diabetes epidemic worldwide, particularly in develop­ing glycerol72 by protein kinase C, and the production of
countries where awareness, systemic control and access free radicals and advanced glycosylation end prod-
to general health-care and eye-care services are more ucts (AGEs), seem to be central to the development of
limited, the magnitude of DR-associated ­blindness will DR73. In addition, accumulating evidence indicates that
remain substantial. neurodegeneration, neuroinflammation and renin–­
angiotensin system (RAS) activation also have impor-
Risk factors for DR tant roles in DR development 74–76. Furthermore, the
The most consistent risk factors for the development of aberrant production of mitochondria-derived reactive
DR are long duration of diabetes, hyperglycaemia and oxygen species and endoplasmic reticulum (ER) stress
hypertension9,10,23,46 (BOX 1). However, there is substantial are also involved in the pathogenesis of DR. The roles of
variation in the onset and severity of DR that cannot mitochondrial impairment and oxidative stress in the
be fully explained by hyperglycaemia and hypertension. development of DR have been reviewed extensively77. ER
Indeed, studies have shown that a proportion of patients stress results from an impairment of the folding capacity
with poor glycaemic and/or blood pressure control do of the ER, causing an accumulation of unfolded proteins
not develop DR47, whereas others with appropriate con- in the ER lumen and activation of the unfolded protein
trol develop severe stages of DR48, suggesting that other response. ER stress contributes to increased oxidative
risk factors have a role. stress as well as inflammation and apoptosis, owing to
Puberty and pregnancy are also well-known risk fac- the failure of the unfolded protein response to resolve
tors for DR, particularly in T1DM49,50. The risk of DR ER stress. This condition is likely to be involved in the
related to pregnancy may be mediated by fluctuating pathogenesis of different neuronal diseases in the brain
glucose levels, but other systemic factors (for example, and retina, including the early stages of DR78.
hypertension and pre-eclampsia, and hyperdynamic
­circulatory state) are also involved49. Development and progression
Although there are strong pathophysiological The chronopathology of DR is slow and can be separ­
­reasons to suggest that dyslipidaemia is a risk factor 51, ated into four overlapping clinical stages: retinal
the relationship between dyslipidaemia, measured using damage without any visible microvascular abnormal-
traditional lipid measures (that is, total cholesterol and ities in a fundoscopic examination; non-proliferative

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 3


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Box 1 | Risk factors for diabetic retinopathy excitatory neurotransmitter and its enhancement in
the extracellular space results in neuron death (excito­
Common and established risk factors toxicity)84. The retina synthesizes neuroprotective fac-
• Hyperglycaemia: a 1% reduction in glycated haemoglobin (HbA1c) serum levels is tors such as pigment epithelium-derived factor (PEDF),
associated with an approximately 35% reduction in risk of diabetic retinopathy (DR) somatostatin, interstitial retinol-binding protein and
development, 15–25% reduction in DR progression, 25% reduction in several neurotrophins74. These neuroprotective factors
vision-threatening DR and 15% reduction in blindness13,128,129,192. are downregulated in the diabetic retina, which might
• Hypertension: a 10 mm Hg reduction in systolic blood pressure is associated with compromise the neuroprotection against the neurotoxic
an approximately 40–50% reduction in DR progression, need for laser treatment factors involved in neurodegeneration.
and vision loss13,134.
However, not all neuroprotective factors are down-
• Diabetes duration: a duration of diabetes of ≥20 years is associated with DR regulated in the diabetic retina. For example, VEGF and
development in 50–90% of patients9,10.
erythropoietin are two well-recognized neuroprotective
• Cataract surgery: cataract surgery is associated with an increased risk of DR factors that are upregulated74. VEGF is upregulated in
progression and diabetic macular oedema development60,61.
the very early stages of the neuro­degenerative process.
• Life stages: pregnancy193–195 and puberty49,195 are associated with an increased risk The accumulation of glutamate85 and the loss of PEDF86
of DR progression.
and somatostatin87 are mech­anisms involved in VEGF
• Nephropathy: diabetic nephropathy is closely correlated with DR56. overexpression. This finding is important because
Less-common or less-established risk factors the increase in VEGF levels as a consequence of the
• Dyslipidaemia: biologically plausible but inconsistent associations between neuro­degenerative process links neuro­degeneration
dyslipidaemia and DR have been reported in different studies. Some studies suggest with early microvascular impairment (that is, blood–
DR risk is associated with increased triglyceride levels, whereas diabetic macular retinal barrier breakdown). Other factors linking
oedema risk is associated with increased low-density lipoprotein and decreased neurodegener­ation and early microvascular impair-
high-density lipoprotein levels51–53. ment are inflammation, upregulation of receptors for
• Obesity: an increased waist/hip ratio and body mass index is associated with DR risk57. AGEs (RAGE) and RAS activation74. Both erythro-
• Anaemia: a strong but uncommon association between anaemia and DR risk has poietin and its receptor are synthesized by the human
been reported58. retina (mainly in the retinal pigment epithelium) 88.
• Inflammatory markers: inconsistent associations have been reported59. Erythropoietin is a powerful neuro­protective factor,
• Ethnicity: Hispanic and South Asian populations show inconsistent associations7,8,11,32. and very high levels of this hormone are found in the
• Genetic factors: few genetic risk markers have been identified67. vitreous fluid of patients with diabetes89. Erythropoietin
is also an important stimulus for the mobilization of
endothelial progenitor cells towards injured retinal sites;
retinal microvascular changes (mild and moderate non-­ thus, erythropoietin is also involved in the remodel­ling
proliferative DR with or without DME); more advanced of damaged tissue90. VEGF and erythro­poietin over-
pre-proliferative changes (severe non-proliferative expression occur in parallel with the downregulation
DR); and proliferative DR and advanced stages of DR of neuroprotective factors and, therefore, may partly
(FIG. 1). The main pathogenetic events involved in the counteract any reduction of the neuroprotective fac-
­development of DR are highlighted in FIG. 2. tors mentioned above. However, in advanced stages
of DR the high levels of VEGF and/or erythropoietin
Retinal damage without visible microvascular abnor- could favour neovascularization and so contribute to
malities. Hyperglycaemia and associated metabolic proliferative DR90,91. In addition, erythropoietin could
pathways can lead to neurodegeneration, early micro- potentiate the effects of VEGF. Thus, overexpression of
vascular impairment and neurovascular unit impair- VEGF and ­erythropoietin is a ­double-edged sword in
ment (FIG. 3). Neurovascular unit impairment includes the pathogenesis of DR.
the alteration of neurovascular coupling, a physio­logical Finally, the increased intraretinal production of
process whereby neural activity is coupled to blood endothelin 1 that occurs in the diabetic retina can
flow and metabolism. This coupling permits the retina induce retinal neurodegeneration, contributing to the
to regulate blood flow in response to neural activity crosstalk between endothelial cells and the neuroretina.
or metabolic demands79. The two hallmarks of retinal Accordingly, the early impairment of endothelial cells
neuro­degeneration are neural apoptosis and reactive could contribute to neurodegeneration through the
glio­sis. These features were confirmed in histopatho- ­activation of endothelin receptors92,93.
logical examinations of eyes of organ donors with dia-
betes who did not show clinical DR in ophthalmological Mild and moderate non-proliferative DR and DME. The
examinations 1–2 years before death80,81. The loss of neu- first vascular changes in the retina that are observed on
ral cells results in the thinning of the retinal ganglion cell histopathology are thickening of the basement mem-
layer and nerve fibre layer, which can now be detected brane, an endothelial injury that leads to the disrup-
by spectral domain OCT82,83. tion of the tight junctions and pericyte loss15 (FIG. 2).
Extracellular glutamate accumulation and an imbal- The consequences of pericyte loss are vascular tone
ance in the retinal production of neuroprotective fac- dysregu­lation and the growth and proliferation of
tors are two of the most important pathogenetic factors endothelial cells owing to the deficit of transforming
involved in diabetes-induced retinal neurodegener­ growth ­factor‑­β produced by pericytes. These changes
ation 74 (BOX  2; FIG.  3) . Glutamate is the principal underlie the development of microaneurysms and dot

4 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

intraretinal haemorrhages, two of the earliest detect­able blood–retinal barrier breakdown. Ageing and hyper­
clinical abnormalities seen in early non-proliferative tension, two frequent conditions associated with T2DM,
DR. The thickening of the basement membrane and the also increase the expression of pro-inflammatory mol­
disruption of the tight junctions are determining fac- ecules in the retina94,95. Increased levels of inflammatory
tors in the leakage of the retinal capillaries. Although mediators may lead to an early and persistent chronic
thickened, the basement membrane is dysfunctional and inflammatory condition in the diabetic retina that results
permits the passage of the intravascular content (pro- in leukocyte activation, leukocyte adhesion to the vas-
teins, lipids, inflammatory mediators and other plasma cular endo­thelium (leukostasis) and alteration of the
constituents) to the interstitial space. VEGF and pro-in- blood–retina barrier, which then leads to increased
flammatory cytokines (IL‑1β, tumour necrosis factor, vascular perme­ability 96,97. The clinical consequence of
IL‑6, IL‑8 and monocyte chemoattractant protein 1) this process is the occurrence of hard exudates, which
produced by glial cells (Müller cells and microglia), the represent the leakage of plasma constituents and mainly
retinal pigment epithelium and macrophages also have consist of lipids and proteins. DME is a consequence of
essential roles in early microvascular impairment and this leakage in the macula region.
The main pathogenetic factors of DME are also
RPE
VEGF and pro-inflammatory cytokines. As men-
tioned above, the main source of pro-inflammatory
VEGF Macrophage cytokines is local synthesis by the retina. Circulating
Photoreceptor cytokines that are increased in T2DM could also
layer
Pro-inflammatory
increase the vascular leakage, but their contribution
OLM cytokines to DME develop­ment remains uncertain. Although
Cone Rod
ONL hypoxia is the most well-­characterized factor that
induces VEGF gene expression, chronic hyper­
OPL Horizontal glycaemia, AGEs98 and pro-inflammatory cytokines
cell
(for example, IL‑1β and IL‑6) could upregulate VEGF
mRNA expression99. This finding explains why VEGF is
Bipolar
INL cell
overexpressed in DME despite the absence of overt
hypoxia. Blood–­retina barrier disruption and vascular
Amacrine cell Müller
glial cell leakage in the macular region leads to DME p ­ rogression
from mild to severe forms (TABLE 1).

IPL Severe non-proliferative DR. In later stages of non-­


proliferative DR (that is, severe non-proliferative DR
Dot haemorrhage or pre-proliferative DR), severe endothelial damage
Ganglion cell
GCL occurs and the balance of vasoactive factors synthesized
Hard exudates by the endothelium leans towards the vasoconstrictor
NFL
agents (for example, endothelin and thromboxane A2).
ILM Pericyte Microaneurysm
In addition, nitric oxide is quenched by AGEs, which
Basement are upregulated in the retina of individuals with dia-
membrane
Tight junction Vasoconstrictor betes. Thus, there is a clear predominance of vaso­
constriction, favouring a hypoxic environment. As
Basement Tight Pericyte loss the disease progresses, endothelial cell loss becomes
membrane junction New blood
thickening failure
a generalized phenomenon, and the capillaries con-
vessel formation
sist of tubes of thickened basement membrane, which
Healthy Non-proliferative Pre-proliferative Proliferative are highly thrombogenic, and could become occluded
DR DR DR
either by platelet-fibrin aggregation or leukocytes. These
Figure 2 | Main pathogenetic events in diabetic retinopathy. Schematic representation two pathways — vasoconstriction and capillary occlu-
Nature Reviews | Disease Primers sion — are crucial in worsening the ischaemia and the
of the main pathogenetic events in the development and progression of diabetic
retinopathy (DR) depicted from healthy (left) to advanced-stage DR (right). consequence is pre-proliferative DR. In addition, angio­
Non-proliferative DR is characterized by vascular changes such as thickening of the poietin 1 receptor (TIE2) is a major regulator of vascular
basement membrane, endothelial injury that leads to the disruption of the tight junctions ­integrity and is involved in pathological angiogenesis100.
and pericyte loss, resulting in dot haemorrhages, microaneurysms and hard exudates. The main clinical finding in a fundoscopic examin­
Vascular endothelial growth factor (VEGF) and pro-inflammatory cytokine production ation of an eye with severe non-proliferative DR is
(IL‑1β, tumour necrosis factor, IL‑6, IL‑8 and monocyte chemoattractant protein 1) by the the presence of ‘cotton wool’ spots. These lesions are
retinal pigment epithelium (RPE), glial cells and macrophages also contribute to the manifest­ations of stasis in the axoplasmic flow, which,
vascular changes. Endothelial damage leads to vasoconstriction owing to vasoconstrictor
in the setting of DR, indicate areas of retinal ischaemia
release (endothelin 1 and thromboxane A2) and hypoxia in pre-proliferative DR, which is
aggravated owing to the capillary occlusion. The severe hypoxia in end-stage DR leads to or infarction. Dilated capillaries and shunts between
neovascularization. These new blood vessels tend to grow into the vitreous body and are arteries and veins can also be identified and are referred
prone to rupture. GCL, ganglion cell layer; ILM, inner limiting membrane; INL, inner to as intraretinal microvascular abnormalities. In addi-
nuclear layer; IPL, inner plexiform layer; NFL, nerve fibre layer; OLM, outer limiting tion, retinal veins appear dilated, tortuous and irregular
membrane; ONL, outer nuclear layer; OPL, outer plexiform layer. in calibre.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 5


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Proliferative DR. The end stage in the natural history or fundoscopic examination is a broad term cover-
of DR is proliferative DR and its subsequent complica- ing three types of tests: direct fundoscopy, indirect
tions. The main pathogenetic factor involved is a severe fundo­s copy and slit lamp microscopy. Other causes
hypoxia, which leads to an imbalance between angio- of retino­pathy should be excluded3,15,16, as these micro-
genic (for example, VEGF) and anti-angiogenic (for vascular lesions are not specific to diabetes and milder
example, PEDF) mediators that favours neovascular­ levels of retinopathy are common in the general popu­
ization, the hallmark of proliferative DR. The first step lation and may be related to hypertension, anaemia
for neovascularization is the proteolytic digestion of the and other factors102–104. In fact, T2DM is defined by
basement membrane, in which the proteases released by the glycaemic level (fasting level of 7.0 mg per dl)
leukocytes have an essential role. The proteolytic frag- at which the preva­lence of retinopathy signs begin
ments of the basement membrane and the angiogenic to rise sharply above the normal population level of
factors stimulated by hypoxia, especially VEGF, lead to ­approximately 10%105.
the migration and replication of endothelial cells, and Clinically, DR is classified as mild, moderate and
finally new vessels appear. These new vessels are fragile severe non-proliferative DR and proliferative DR
owing to architectural weakness, are prone to bleed- (TABLE 1), with proliferative DR further classified based
ing and tend to grow into the vitreous body in which on the location of the new vessels, either on the optic
they are eventually anchored by means of fibrovascular disc or elsewhere 72. Mild, non-proliferative DR is
­tissue. This fibrotic tissue may contract and this can defined by capillary microaneurysms and a few intra-
lead to advanced DR and tractional retinal detachment, retinal dot and blot haemorrhages (FIG. 4a,b). These
accompanied by severe vision loss101. Retinal detach- lesions can become evident years after the onset of
ment and vitreous haemorrhage constitute the advanced T1DM but are often seen at the time of diagnosis in
stage of DR and are sight-threatening conditions. T2DM106. As disease progresses to moderate and severe
non-proliferative DR stages, lesion density increases.
Diagnosis, screening and prevention Disease progression is also associated with other signs
Diagnosis reflecting capillary leakage, seen as oedema and intra-
By definition, DR is restricted to people with dia­ retinal hard exudates, or signs reflecting capillary
betes and is defined as a spectrum of retinal micro­ occlusion associated with ischaemic damage of the
vascular lesions seen on retinal examination. A retinal nerve fibre layer, seen as cotton wool spots (FIG. 4b),
venous dilation and beading, and intraretinal micro-
vascular abnormalities. Neovascularization in the ret-
Metabolic pathways induced by hyperglycaemia ina signals the onset of proliferative DR (FIG. 4c), with
a risk of vitreous haemorrhage and traction retinal
detachment, and anterior segment neovascularization
Neurovascular
unit impairment of the iris (rubeosis) or anterior chamber angle with
Early microvascular impairment: Neurodegeneration
intraocular pressure rise (neovascular glaucoma)3. The
• Abnormal haemodynamic
response risk of progression to proliferative DR is proportional
ET1 and ET
• BRB breakdown receptors to severity; 5% of mild non-proliferative DR progress
• Glutamate accumulation
(excitotoxicity) to proliferative DR within 1  year, compared with
• Imbalance in the retinal 20% of moderate and 50% of severe non-­proliferative
production of neuroprotective DR. DME is separately classified as mild, moder­
factors
ate and severe (FIG. 4d), and may occur even in mild,
• VEGF and other vasoactive agents non-proliferative DR72.
• Inflammatory mediators ↓SMS, CORT, NTFs, Fluorescein angiography — a test in which retinal
• RAGE upregulation • Neuron death PEDF and RBP photographs are taken after systemic injection of a
• RAS activation • Glial cell dysfunction ↑VEGF and EPO vegetable-­based, fluorescent dye — has traditionally
been used to locate the sources and extent of vascular
Figure 3 | Potential mechanisms linking neurodegeneration and microangiopathy
Nature Reviews | Disease Primers
in diabetic retinopathy. The metabolic pathways induced by hyperglycaemia lead to leakage, the presence of ischaemia, and in planning
neurodegeneration, early microvascular impairment and neurovascular unit impairment. laser photocoagulation therapy. However, there is now
The accumulation of glutamate in the extracellular space (that is, the intersynaptic cleft no longer a strong rationale for using standard fluo-
of retinal neurons) and the decrease in the retinal production of neuroprotective factors rescein angiography routinely, although studies using
are key events for neuron apoptosis and glial dysfunction. However, this process also wide-field fluorescein angiography are underway and
induces the overexpression of vascular endothelial growth factor (VEGF)196, which has a may change the utility of angiography 106,107.
key role in the disruption of the blood–retinal barrier (BRB). Apart from VEGF and other OCT is a newer non-contact technology that creates
vasoactive agents, there are other factors that link neurodegeneration and early high-resolution cross-sectional images of the retina to
microvascular impairment, such as the upregulation of inflammatory mediators and document 3D structural changes, such as thickening of
receptors for advanced glycosylation end products (RAGE) and activation of the renin–
different retinal layers owing to oedema, which cannot
angiotensin system (RAS). In addition, an increase in the intraretinal production of
endothelin 1 (ET1) can induce retinal neurodegeneration, thus contributing to the be readily assessed via clinical fundoscopic examin­
crosstalk between endothelial cells and the neuroretina. CORT, cortistatin; EPO, ation of retinal photography (FIG. 5). OCT has been
erythropoietin; NMDA, N‑methyl-d‑aspartate; NTFs, neurotrophins; PEDF, pigment particu­larly useful for monitoring DME progression
epithelium-derived factor; RBP, inter-photoreceptor retinoid-binding protein; over the course of intravitreal injection therapies for
SMS, somatostatin. DME (see Management below).

6 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Box 2 | Links between neurodegeneration and microvascular impairment adults114. Nevertheless, there remains a lack of consensus
on the best mode of screening. Although detailed clin­
The main molecular links between neurodegeneration and microvascular impairment ical examination by an ophthalmologist using a slit-lamp
in diabetic retinopathy (DR) are as follows: microscopy instrument remains ideal, logistical chal-
• Endothelin 1: endothelin is synthesized by endothelial cells and is a potent lenges are considerable, and it is impossible for ophthal-
vasoconstrictor that exerts its action through type A and type B endothelin receptors. mologists to screen every person with diabetes, as this
Endothelin 1 exerts neuroprotective actions on top of its vasoactive role and therefore is a highly inefficient use of resources even if adequate
has an essential role in neurovascular coupling.
trained ophthalmologists are available.
• Glutamate: glutamate is the principal excitatory neurotransmitter, and increased levels The major technical innovation in the past two
of glutamate in the extracellular space induces retinal neuron death (excitotoxicity).
­decades has been the introduction of digital retinal
The N‑methyl-d‑aspartate (NMDA) receptor is a glutamate receptor.
photo­graphy without the need for pharmacological pupil
• Somatostatin: in the eye, somatostatin is a peptide that is mainly synthesized by retinal
dilation (non-mydriatic retinal photography)110,111,115,
pigment epithelial cells and has neuroprotective and anti-inflammatory actions.
and the expansion and adoption of telemedicine to
• Cortistatin: cortistatin is very similar to somatostatin, shares its receptors,
transmit digital images between locations22,116. These
has neuroprotective actions in the retina and is synthesized in the retinal
pigment epithelium.
technologies have the potential to offer fast, systematic
large-scale solutions to DR screening to both developed
• Pigment epithelium-derived factor: this protein is a powerful anti-angiogenic factor
and developing countries. For example, nurses or tech-
with neuroprotective properties.
nicians can take a retinal photograph at the commu-
• Retinol-binding protein: interstitial or retinol-binding protein is crucial for the visual
nity or primary care setting (image capture sites), and
cycle (physiological and biochemical changes associated with perception of a light
stimulus at the retina) and is an essential carrier of nutrients for photoreceptors. transmit the retinal images via the Internet to a central
location where trained technicians can evaluate the
• Neurotrophins: neurotrophic factors (NTFs) are a family of structurally and functionally
related proteins that are essential for the growth, differentiation and survival of several photographs for signs and stages of DR (retinal grading
cell types, including retinal neurons, glial cells and endothelial cells. The main members centre). Results are then transmitted back to the image
of this family are nerve growth factor and brain-derived neurotrophic factor. capture sites, either directly to primary care physicians
• Vascular endothelial growth factor: vascular endothelial growth factor (VEGF) is or ophthalmologists. A meta-analysis demonstrated
required for normal vascular development, is also a survival factor essential for the that non-mydriatic retinal photographs taken by non-­
integrity of endothelial cells and exerts neuroprotective actions. VEGF medical technicians is associated with an 80% sensitivity
overexpression in DR leads to hyperpermeability (vascular leakage) and induces for detecting DR, well above the 60% recommended for
proliferation of capillaries in advanced-stage DR, resulting in neovascularization cost-effectiveness117. Telemedicine, national-level DR
(the hallmark of proliferative DR). screening programmes with quality assurance, is widely
• Erythropoietin: erythropoietin is abundantly synthesized by the retinal pigment practiced in the United Kingdom118. The threshold for
epithelium and has potent neurotrophic action in the retina. In addition, erythropoietin referral to subsequent examinations by an ophthal­
stimulates the mobilization of endothelial progenitor cells towards injured retinal sites. mologist can be set at varying levels of DR severity
However, when VEGF is overexpressed, erythropoietin could enhance its angiogenic according to international criteria or local guidelines
properties, contributing to the progression to proliferative DR.
and preferences118,119. The UK systemic screening pro-
grammes in particular have provided the foundation for
innovative models exploring the feasibility, safety and
In addition to these assessments, there are multiple cost-effectiveness of extending screening beyond the
early functional and structural abnormalities of the yearly screening 120,121.
retina that can be identified before definitive signs of Irrespective of screening methods, assessment of
microangiopathy have developed2. Such abnormalities patients’ best-corrected visual acuity and systemic
include subnormal electroretinographic oscillatory diabetes profile may allow determination of overall
potentials, downregulation of endothelial junction pro- compliance and general risks, and thereby influence
teins, and upregulation of markers of glial activity and the urgency of referral109. Furthermore, retinal photo­
inflammation. However, the clinical utility of assessing graphy screening only captures DR and does not screen
these early functional changes is unclear 108. for other eye conditions such as cataract or glaucoma,
which are also common in individuals with diabetes.
Screening There are three emerging advances in DR screening.
DR meets all of the criteria for screening: it is highly First, DME screening using retinal photography relies
prevalent, it is largely confined to a well-defined popu­ on an indirect assessment based on the detection of
lation (people with diabetes), it is easy to detect and it hard exudates near the fovea122 (FIG. 4d). As indicated,
is asymptomatic until advanced. Moreover, screening clinical fundoscopic examination or retinal photography
methods are non-invasive and well accepted, and there ­cannot objectively assess the thickness of the retina seen
are clear treatment modalities for early-stage disease. in DME that can be measured by OCT. Thus, an inte-
DR screening coupled with timely, pre-symptomatic grated screening programme combining photo­graphy
intervention is a rational and cost-effective strategy for screening for all people with diabetes, with selected use
the prevention of blindness109–111. Results from studies of OCT in cases in which DME is suspected122, may
have suggested that systematic, national screening pro- reduce false-positive cases (presumed DME based on
grammes for DR can reduce blindness from diabetes by photographic signs but no retinal thickening on OCT).
30–50%112–114, and this would translate to a reduction of However, the logistics and costs of having OCT in a DR
more than 10% of all cases of blindness in working-age screening programme are challenging. Second, newer

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 7


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

HbA1c (for example, from 9% to 8%) lowers the risk


a b
Haemorrhage of retinopathy by 30–40%128. In addition, the benefit of
early intensive glycaemic control can be long-lasting
Microaneurysm (metabolic memory). This effect was demonstrated by
or haemorrhage a 72–87% risk reduction in progression of DR that was
even seen 4 years after completion of the DCCT study,
when patients in the intensive treatment group had
HbA1C levels return to levels >7%130.
Microaneurysm The effect of intensive glycaemic control in T2DM
or haemorrhage was also investigated in the UKPDS129. Patients were
Cotton wool spots
randomly assigned to either intensive treatment with
c d sulfonylurea and/or insulin, or conventional treatment
with diet control for an average of 10 years. The inten-
Hard exudates Blot sive glucose control group maintained a lower HbA1c
haemorrhage level (of ~7.0%) than the conventional treatment
group (HbA1c level of 7.9%). Over 12 years, intensive
glucose control resulted in 21% reduction in risk for
DR129. Importantly, a long-term beneficial effect (meta­
New blood vessel
bolic memory) was evident, with the protective effect
formation at the Microaneurysm Hard exudates of initial intensive therapy observable 25 years after
optic disc or haemorrhage randomization (24% relative risk reduction for micro-
vascular disease)131. However, the additional benefit of
Figure 4 | Clinical signs of diabetic retinopathy on fundoscopic examination.
Nature Reviews | Disease Primers achieving even lower HbA1c levels (towards normo­
a | Mild non-proliferative diabetic retinopathy (DR) with microaneurysms and glycaemia) with intensive glycaemic therapy is less clear.
haemorrhage. b | Moderate non-proliferative DR with microaneurysms, haemorrhage The Action to Control Cardiovascular Risk in Diabetes
and cotton wool spots. c | Proliferative DR showing new blood vessels at the optic disc.
(ACCORD) 132 eye study was a subgroup of 2,856
d | Diabetic macular oedema showing hard exudates at the fovea centre.
patients within the main ACCORD study, designed to
evaluate the effects of intensive glycaemic control, inten-
photographic techniques are being tested, including sive blood pressure control and lipid management on
wide-field fundus photography, whereby larger areas of the progression of DR in patients with T2DM selected
the retina can be captured in a single image than tradi- for increased cardiovascular risk. Intensive glycaemic
tional cameras that can image only a limited part of the therapy (target HbA1c levels of 6.0% versus 7.0–7.9%)
retina123,124. Third, new automated DR detection software significantly reduced the risk of progression of DR at
to aid photographic assessment has been developed125,126. 4 years (7.3% versus 10.4% with standard therapy)132.
Although the cost-effectiveness of these technologies However, the Action in Diabetes and Vascular Disease:
has not been thoroughly tested, their use will probably Preterax and Diamicron MR Controlled Evaluation133
increase in the future. trial did not show an additional benefit of more inten-
sive glucose control compared with intensive control
Prevention (HbA1c levels of 6.5% versus 7.5%) for reduction of
Glycaemic control. In addition to screening, DR can new or worsening retinopathy over 5 years133, although
be prevented by the effective systemic control of a further 21% reduction of risk for nephro­pathy was
glycaemia and blood pressure in patients with dia­ reported in the more intensive glucose control group
betes3,15,16,127. The Diabetes Control and Complications compared with the intensive control group. The
Trial (DCCT)71,128 and the United Kingdom Prospective ADVANCE133 trial randomly assigned 11,140 patients
Diabetes Study (UKPDS)129 have provided strong evi- with T2DM and an additional risk factor for a vascu-
dence that a tighter control of glycaemia (target gly- lar event into four categories using a factorial design
cated haemoglobin (HbA1c) level of <7%) reduces the of intensive lowering of glucose (target HbA1c level of
risk of development and progression of DR in both 6.5%) versus standard glucose therapy (target HbA1c
T1DM and T2DM. Although a small risk of initial level of 7.0%), and additional routine blood pressure
worsening of retino­pathy at the onset of therapy exists, lowering versus routine blood pressure lower­ing. The
the long-term benefits outweigh this risk128. Over a ADVANCE trial did not show any beneficial effect of
7‑year period, the DCCT reported that intensive ther- either therapy on DR. Furthermore, any potential bene­
apy to maintain HbA1c levels of ~7% (compared with fit on DR should also be balanced against the increased
~9% in those with T1DM) reduced the onset of new risk of having a hypoglycaemic event in the intensive
DR by 76% in the primary intervention cohort (which glycaemic therapy group, as the intensive therapy group
recruited patients with no DR at baseline). Moreover, had an increased rate of death from any cause in the
progression of existing DR in the secondary inter- ACCORD study 133.
vention cohort (which recruited patients with pre-­
existing mild DR) was reduced by 50%128. Therefore, it Blood pressure control. In terms of controlling blood
is important to achieve glycaemic control early, prefer­ pressure in patients with diabetes who also have hyper-
ably before the onset of DR. Each per cent reduction in tension, to prevent DR, the clinical evidence is also

8 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

robust. In the UKPDS, tighter blood pressure control the control versus treated group, and its short (2‑year)
(target blood pressure of <150/85 mm Hg versus a less follow‑up. The Diabetic Retinopathy Candesartan Trials
tight control target of <180/105 mm Hg) reduced the (DIRECT) programme examined effects of candesar­
risks of DR progression and need for laser treatment tan (an angiotensin receptor blocker) in T1DM 136
by approximately one-third and vision loss by 50% and T2DM 136,137. In T1DM, 5  years of ­c andesartan
in T2DM129. However, in contrast to glycaemic con- treatment resulted in a reduction in the incidence of
trol, these benefits are not sustainable (no ‘memory’) DR compared to placebo (hazard ratio of 0.82), but
without ongoing and long-term maintenance of blood no beneficial effect on DR progression in those with
­pressure control134. pre-existing DR was observed136. In T2DM with mild
Certain blood pressure-lowering medications, par- to moderate DR, 5 years of candesartan treatment
ticularly those targeting the RAS, can exert beneficial resulted in DR regression by 34%137. An overall change
actions other than lowering the blood pressure level. towards less severe DR was observed in the candesartan
The EURODIAB controlled trial of lisinopril (an group in T2DM. The Renin Angiotensin System Study
angiotensin-converting enzyme inhibitor) in insulin-­ (RASS) evaluated the effect of enalapril (an angio­tensin
dependent diabetes (EUCLID) study investigated the convert­ing enzyme inhibitor) and losartan (an angio-
effect of lisinopril on retinopathy in non-hypertensive tensin receptor blocker) on DR ­progression in T1DM138.
patients with T1DM who were normoalbuminuric or After 5 years, the risk of DR progression was reduced
microalbuminuric. One hundred and seventeen patients by 65% with enalapril and 70% with losartan, indepen­
were randomly assigned to the placebo group and dent of blood pressure. These trials suggest that specific
103 patients were randomized to the lisinopril group. drugs that target the RAS may be superior to stand-
The results showed that lisinopril reduced the risk of ard blood ­pressure-lowering medications in reducing
DR and proliferative DR progression by 50% and 80%, retinopathy risk.
respectively 135. However, the EUCLID study had limi-
tations in terms of differences in baseline glycaemia in Lipid control. Although dyslipidaemia has also been
linked to risk of DR, the effects of statin therapy on DR
risk have been inconsistent 51. The strongest evidence
a from two independent randomized trials suggests that
Cystoid
spaces Nerve fenofibrate could reduce the risk of vision-­threatening
Inner limiting membrane Inner nuclear layer fibre layer DR in T2DM. The Fenofibrate Intervention and Event
Inner plexiform layer Lowering in Diabetes study assessed the effects of
Outer plexiform layer fenofibrate on cardiovascular outcomes in patients
with T2DM who did not take statins139. Laser treatment
Retina pigment epithelium
for DR, a pre-specified tertiary outcome, was reduced
Choroid by 37% in the fenofibrate group compared to a pla-
Outer nuclear layer
cebo group after 5 years. The number needed to treat
b Cystoid with fenofibrate to prevent laser treatment was only
spaces 17 in people with existing DR at study entry 139. In a
sub-study, progression of DR after 4.7 years was signifi­
cantly lower in the fenofibrate group compared to a
placebo group in patients with DR at baseline (14.6%
versus 3.1%)139. The ACCORD eye trial, in addition to
investigating the effect of intensive glycaemic control,
also evaluated the effect of adding fenofibrate to stat-
c ins to evaluate the effect of this combination on DR132.
Intraretinal
cysts After 4 years, the fenofibrate group had reduced pro-
gression of DR compared to standard treatment (6.5%
versus 10.2%)132. In terms of the mechanisms of action,
fenofibrate has been suggested to also influence non-­
lipid mechanisms (for example, VEGF levels)140, but
specific mechanisms remain to be fully elucidated.

Management
Figure 5 | Optical coherence tomography showing treatment outcomes
Nature Reviews for Primers
| Disease Vision loss as a consequence of severe DR can be mini­
diabetic macular oedema. Optical coherence tomography (OCT) images of a mized by intervention using intraocular anti-VEGF
patient with type 2 diabetes with diabetic macular oedema (DME). a | OCT image of
agents for DME, panretinal laser photo­coagulation
a patient with vision of 6/45 (vision of 6/6 is considered normal vision and 6/60 is
considered legal blindness in some countries) showing fluid accumulation in cystoid
for proliferative DR, and vitrectomy for vitreous
spaces before intravitreal injections of ranibizumab (an anti-vascular endothelial growth haemor­rhage and traction retinal detachment 3,14–16,109.
factor therapy) were started. b | Vision of 6/18 (that is, vision improved from 6/45 to 6/18) Intravitreal anti-VEGF therapy has also been shown to
was observed after three intravitreal injections of ranibizumab. c | Vision of 6/9 (that is, reduce the level of DR141, and its potential utility for treat-
vision improved from 6/18 to 6/9, almost normal vision) was observed after seven ment of proliferative DR has now been ­demonstrated in
intravitreal injections of ranibizumab. recent clinical trials20.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 9


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Laser photocoagulation injections are usually performed in office settings


For almost three decades, laser photocoagulation has by ophthalmologists using an aseptic technique and
been the standard ocular treatment for proliferative DR ­topical anaesthesia.
and DME14,15. The mechanism of action of laser photo- The activation of inflammatory pathways has been
coagulation is not fully understood, but one hypothesis proposed as one of the mechanisms in DME patho-
is that laser photocoagulation-induced scars alleviate genesis. Corticosteroids suppress multiple pathways of
hypoxia and affects the haemodynamics of the retinal inflammation and have been advocated as a treatment
circulation. Improved retinal oxygenation reduces neo- option for DME, particularly in cases refractory to laser
vascularization and oedema formation by reducing the photocoagulation. In addition, corticosteroids down-
production of VEGF and other factors, and by trigger­ regulate VEGF expression144 and reduce the induction
ing haemodynamic changes14. The primary goal for of VEGF by pro-inflammatory mediators. However, the
most patients receiving laser therapy is to preserve use- introduction of anti-VEGF agents confined the use of
ful vision and prevent blindness, and reversal of vision corticosteroids to selected patients, such as those with
loss is uncommon. previous cataract surgery and with chronic DME.
Two landmark trials provided the evidence for the Off-label use of triamcinolone (a long-acting
use of laser photocoagulation for DR. The Diabetic cortico­steroid) has been shown to reduce DME and to
Retinopathy Study (DRS) demonstrated that pan­ improve vision in the short-term145. The DR Clinical
retinal laser photocoagulation reduced the risk of Research Network (DRCRnet) reported that the bene-
severe vision loss from 33.0% to 13.9% at 5  years fits of intravitreal triamcinolone did not persist beyond
in patients with proliferative DR or severe non-­ 1 year, and by 2 years the visual outcome was generally
proliferative DR 142. The Early Treatment Diabetic not better than laser photocoagulation146. Importantly,
Retinopathy Study (ETDRS) demonstrated that focal considerable adverse effects (such as cataract and
laser photocoagulation reduced the risk of vision loss glaucoma) are common. In addition, there are con-
in patients with clinically significant DME from 24% cerns of increased risk of infectious endophthalmitis
to 12%, but did not eliminate this risk142,143. However, (an eye infection) owing to the immunosuppressive
only 3% of patients had a significant improvement in properties of corticosteroid and also sterile (non-­
visual acuity, which is partly explained by the ceiling infectious) endophthalmitis, which may be precipi-
effect caused by the relatively good baseline vision tated by additives in the t­ riamcinolone preparation or
in the study population. Furthermore, laser photo­ bacterial endotoxins147.
coagulation itself is associated with significant ocular Two sustained-release intravitreal implants have
side effects due to its inherent destructive nature to the recently been evaluated for DME. Ozurdex (Allergan,
retina, including induction of scotoma (that is, a blind Irvine, California, USA) is a biodegradable implant
spot), enlargement of laser scars and ­development of that is inserted using a 22‑gauge applicator and
secondary neovascularization. releases 0.7 mg of dexamethasone over 120–180 days.
In one trial, significantly more patients in the Ozurdex
Intraocular steroids group achieved ≥15 letter improvement (improve-
In the 2000s, intraocular administration of steroids and ment in ~3 lines on the standard Snellen visual acuity
anti-VEGF agents were introduced as new treatment chart) in best corrected visual acuity compared with
modalities for DME and proliferative DR. Intraocular the sham group (22.2% versus 12%, P < 0.018) over

Table 2 | Randomized controlled trials of anti-VEGF therapy for diabetic macular oedema
Trial name Number of Anti-VEGF agent Vision results (%)* Follow‑up Refs
eyes (months)
Gained Lost
RISE and RIDE 377 Ranibizumab 34–46 (12–18) 2–4 (9–10) 24 153
DR Clinical Research 854 Ranibizumab (+ laser) 28–30 (15) 2 (8) 24 146
Network Protocol I
READ‑2 126 Ranibizumab (± laser) 23 (17) 3% (6) 24 156
RESOLVE 151 Ranibizumab (± laser) 32 (10) 3 (20) 12 155
RESTORE 345 Ranibizumab (± laser) 23 (8) 1–3 (8) 12 154
BOLT 80 Bevacizumab 32 (4) 0 (14) 24 145
DA VINCI 176 Aflibercept 46 (11) NR 12 157
VIVID and VISTA 872 Aflibercept 31–41 (7.8–9.1) 0–0.7 (9–10) 12 158
DR Clinical Research 102 Aflibercept 68 1 12 159
Network Protocol T
102 Bevacizumab 42 2 12
101 Ranibizumab 50 2 12
Based on data from REF. 196. NR, not reported; VEGF, vascular endothelial growth factor. *Percentage of patients with ≥3‑line
vision gain or loss (versus laser photocoagulation therapy alone).

10 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Diabetic macular oedema visual acuity compared with ranibizumab and bevaci-
zumab. Another comparative effectiveness randomized
clinical trial conducted by the DRCRnet evaluated the
Assessment effectiveness at ≤5 years of two different regimens of
intravitreal ranibizumab combined with focal laser for
DME. Most eyes maintained vision gains obtained dur-
Mild to moderate Moderate to severe
ing treatment for the first year with little need for addi-
No centre involvement Centre involvement tional treatment after 3 years (median 13–17 injections
over 5 years)146,160. Laser photocoagulation performed
immediately at the start of intravitreal ranibizumab
Visual acuity Visual acuity treatment is not superior to delaying laser treatment
better than 20/30 20/30 or worse
for ≥4 weeks, and half of eyes in the group treated with
Treatment intravitreal ranibizumab may avoid laser for ≥5 years
failure with this approach. These results suggest that in con-
Focal or grid laser photocoagulation Anti-VEGF therapy
trast to treatment for neovascular wet (advanced-stage)
Clinical OCT age-related macular degener­ation, anti-VEGF treatment
for DME appears to allow the tapering of injection
Figure 6 | Management of diabetic macular oedema.Nature Schematic overview
Reviews of thePrimers ­frequency over time.
| Disease
management algorithm of diabetic macular oedema16. OCT, optical coherence Anti-VEGF therapy has also been used as a potential
tomography; VEGF, vascular endothelial growth factor. treatment for proliferative DR. The DRCRnet recently
presented the results of a non-inferiority trial compar-
3 years148. Retreatment is necessary in most patients; ing panretinal laser photocoagulation and intravitreal
most patients received on average four injections over ranibizumab among patients with proliferative DR20. At
the study period (3 years). Iluvien (Alimera Sciences, 2 years, mean improvement in visual acuity was similar
Alpharetta, Georgia, USA), another sustained-release between the two treatment modalities. Although longer-
intravitreal implant, provides a therapeutic effect of up term outcomes are unclear, these results do suggest that
to 36 months by delivering sustained sub-microgram anti-VEGF therapy might be an alternative treatment for
levels of fluocinolone acetonide (a corticosteroid). The proliferative DR.
implant can be injected through the back of the eye In terms of safety, most clinical trials reported a
with a 25‑gauge needle. The Fluocinolone Acetonide favourable safety profile for anti-VEGF agents. Ocular
in Diabetic Macular Edema149 study demonstrated that safety profiles based on various DME trials have been
in people with chronic DME for 3 years or longer, more consistent with anti-VEGF injections for other indica-
patients treated with Iluvien had an improvement of tions. In particular, rates of procedure-related serious
≥15 letters after 36 months treatment than in the sham adverse events, such as endophthalmitis and traumatic
group (28.7% versus 18.9%)149. cataract, are low. However, long-term intravitreal injec-
tions of anti-VEGF agents could lead to the neuro­
Intraocular anti-VEGF agents degeneration of the remaining healthy retina and to an
Angiogenesis is a key common pathway of DR, par- increased risk of circulation disturbances in the chorio­
ticularly in severe stages of DR, including DME and capillaris161. There were also concerns regarding the
proliferative DR. The development of anti-VEGF systemic safety of anti-VEGF therapy 162, particularly as
therapy has therefore been one of the most profound patients with diabetes have multiple comorbidities and
advances in ophthalmology 15,150–152. There are currently have increased cardiovascular risk163. Up to 40–50% of
three commonly used anti-VEGF agents: ranibizumab, patients with DME may need bilateral ocular therapy,
­bevacizumab and aflibercept. thus potentially increasing the drug concentration in the
For DME, pivotal clinical trials have demonstrated systemic circulation164. Although no significant safety
the superior visual acuity outcome of anti-VEGF concern has been raised from the large-scale clinical
therapy compared to laser photocoagulation for the ­trials, these studies do not have adequate power to detect
treatment of DME145,146,153–158 (FIG. 5; TABLE 2). Although small changes in the risk of rare events such as stroke or
multiple intraocular injections (between 6 and 12) over vascular death162,165. Continued vigilance from clinicians
the first 12 months are needed, there are differences in and collection of real-life data from post-marketing sur-
retreatment protocols over this period (such as fixed veillance are necessary to further clarify the safety profile
monthly dosing versus treatment based on clinical of anti-VEGF treatments in patients with diabetes.
parameters) and intensity between individual stud-
ies, leading to different levels of visual improvement Vitrectomy
reported. In 2015, the DRCRnet reported the 12‑month In a small proportion of people with advanced dis-
results of a study that compared, head-to-head, the ease, such as those with severe proliferative DR who
rela­tive efficacy and safety of the three commonly used do not respond to panretinal laser photocoagulation
anti-VEGF agents159. All three agents improved vision and intravitreal anti-VEGF therapy, or those who have
in patients who have DME involving the foveal centre, persistent vitreous haemorrhage and/or traction retinal
and were well tolerated. However, aflibercept was more detachment, ocular surgery involving the removal of the
effective in improving vision in eyes with poorer initial ­vitreous body (termed vitrectomy) may be needed3.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 11


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

There are multiple ways to measure HR‑QOL.


Principally, these include patient preferences, quality-­
n–x Elimination of diabetic retinopathy adjusted life years (QALY)-validated instruments such
(maintained health and/or vision), but as EQ‑5D and decision analyses based on these inputs173.
patient accepts shorter duration of life Patient preferences aim to quantify HR‑QOL through
the use of two theoretical constructs: the standard refer­
Current health
and/or vision ence gamble or the time trade-off technique. In the
n former model, individuals are asked to consider either
living life in their present state (in this case DR) or opt-
ing for an experimental treatment with some degree of
risk. Research based on this model demonstrates that
Diabetic retinopathy, people are willing to take a 50% risk of death to eliminate
but longer duration of life
Deterioration of their DR171. In the time trade-off model, people are asked
health and/or vision whether they are willing to live with their present con-
Remaining lifetime dition for an additional set number of years (frequently
10 years) or opt for a shorter life with their condition
Figure 7 | Time trade-off technique to evaluate quality of life. The time trade-off eliminated. Research based on this model shows that
technique is a method to elicit patient preferences. In this Nature Reviews
scenario, | Disease
a patient withPrimers patients with DR are willing to accept a 20–30% shorter
diabetic retinopathy (DR) is asked to trade-off between their current level of health for a life if their condition could be eliminated169,170 (FIG. 7).
defined period of time (n), or perfect vision or health but for a reduced length of time In terms of the use of QALY to measure HR‑QOL,
(n − x). When the patient has no preference for one of the two options — for instance,
a multi-attribute utility (with utility defined as a cardi-
7 years in perfect health and 10 years in their current state — a utility, or numerical
estimate of quality of life, is quantified (0.7 in this case). Research has shown that the
nal value that represents the strength of an individual’s
average patient with DR is willing to trade-off between 20 and 30% of their remaining life prefer­ence for a specific health outcome) instrument
to eliminate their ocular disease; their utilities are estimated at 0.7–0.8 (REFS 170,197). such as the EQ‑5D or regression equation based on visual
acuity is another way to calculate QALY174. However,
research into the impact of DR on HR‑QOL by employ-
Treatment guidelines ing EQ‑5D has produced inconsistent findings. This
With new therapies now available, guidelines on the inconsistency could be due to a lack in sensitivity of the
management of DR and DME are evolving 16,109,166,167 generic utility measures used to evaluate the specific
(FIG. 6). If DME involves the fovea centre with vision burden of DR‑induced vision loss.
loss, anti-VEGF therapy is considered the first-line treat- Estimation of utilities is also important in deriving the
ment 16,166,167. However, focal laser coagulation remains inputs for cost–utility analyses (aimed at calculat­ing
a useful mode of treatment for DME not involving the the cost per QALY), which are mathematical models to
foveal centre. Intraocular steroids could be used as a determine incremental benefits of a new treat­ment ver-
second-line therapy for cases that do not respond to sus incremental costs. Research shows that treatments
these therapies. For proliferative DR and in certain cases including laser photocoagulation and anti-VEGF ther-
of severe non-proliferative DR, panretinal laser photo­ apy both improve quality of life and are cost-­effective19.
coagulation continues to be the treatment of choice, until Intravitreal ranibizumab confers an 11% improvement
longer term results of trials using anti-VEGF therapy in quality of life and saves society money through a
are available109. reduction in falls, in rates of depression and in nursing
The choice of which anti-VEGF agent to use for home admissions, among others19.
patients with DME should be individualized to health-care
systems, patient preference and cost. No evidence exists Outlook
that specific patient characteristics, systemic conditions Despite improvements in diabetes care, increasing access
or medication use influences choice of therapy. However, to screening for diabetes and DR, and effective systemic
patients with recent cardiovascular events should be aware and ocular treatments, the public health burden of DR
of possible increased risks of stroke or vascular disease continues to be substantial. The number of people with
associated with anti-VEGF therapy, although there is no DR is likely to continue to rise owing to population
robust evidence to support this concern168. growth, ageing demographics and the expanding dia­
betes epidemic worldwide. A review using published esti-
Quality of life mates from studies from 91 countries suggests that nearly
The impact of DR and associated visual impairment on 440 million people between the ages of 20 and 79 years
health-related quality of life (HR‑QOL) is substantial. will suffer from diabetes (prevalence 7.7%) by 2030,
Patients with DR have reduced physical, emotional and largely driven by an increase in the diabetes prevalence in
social well-being, and use many health-care resources6. ­developing nations, including those in Asia and Africa1.
Numerous studies have estimated that DR reduces Although laser photocoagulation therapy has long
HR‑QOL by 20–30% when it affects the eye with worse been the standard of care for managing DME and
vision, and up to 50% if it affects the eye with better vision, prolifer­ative DR142,143, the development and application
compared with persons with diabetes without DR17,169,170. of intravitreal anti-VEGF agents has heralded a new
Patients with diabetes estimate a 60% ­reduction in treatment paradigm for DME, with the potential to
HR‑QOL if they were to develop blindness171,172. dramatically improve vision and quality of life3,14,16,159,175,

12 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

and potentially for proliferative DR20. Given that anti- As discussed in this Primer, the development of DR
VEGF agents are costly and require repeat applications, is strongly influenced by diabetes duration, and systemic
the economic burden for both patients and the society control of glycaemia and blood pressure9. Optimal con-
is substantial176. Considerable savings of 40–90% could trol of glycaemia and blood pressure is critical for pre-
potentially be achieved with a less costly drug, when less- venting DR development in primary care settings128,129,134.
costly drugs of similar efficacy are used in a personal- Although international guidelines recommend that
ized approach to manage DME177. Bevacizumab confers patients with T1DM should be screened 5 years from
greater cost-effective value among the currently available diagnosis and those with T2DM at the time of diagno-
anti-VEGF agents for DME, as ranibizumab and afliber- sis109, only half of patients receive appropriate screen-
cept are substantially more expensive than bevacizumab ing 186. Moreover, patients’ understanding of diabetes and
in some countries178. Furthermore, anti-VEGF therapy DR, the importance of systemic control, and utilization
poses considerable challenges in developing countries. of health-care services is low in the United States and
What are avenues for further research? Clinical other countries187–189. Systems that facilitate seamless
­trials are ongoing to evaluate anti-VEGF treatments for communication between primary care providers, intern-
DME eyes with very good visual acuity and for redu­ ists, optometrists and ophthalmologists are crucial to
cing DR progression (TABLE 2). Importantly, anti-VEGF ensure that diabetes care is patient-centric, holistic and
monotherapy for DME is not a cure, and new thera- comprehensive. Thus, broad, integrated public health-
peutics targeted at other mechanisms are needed179. care programmes that combine DR screening with
Apart from neovascularization, several other mech- raising awareness, public education and appropriate sys-
anisms are important in the pathogenesis of DR (that temic control of glycaemia and blood pressure are likely
is, neuro­degeneration, neuroinflammation and renin–­ to have greater impact than just the provision of tertiary
angiotensin). Experimental therapies targeting these DR care21,109,190,191. Indeed, data from western populations
pathways have been shown to have positive effects180, but are starting to show a reduction in DR progression and
more research is clearly needed in this area. Emerging vision loss based on broad measures44.
research into systemic (for example, microRNA, pro- In conclusion, DR is an increasingly common global
teomics and metabolomics) and vitreous biomarkers clinical and public health condition that exerts a con-
may uncover additional novel pathways181,182. siderable effect on patients and society. New thera-
The increasing use of advanced OCT technology pies such as anti-VEGF treatments have been a major
(for example, new higher resolution OCT) has also breakthrough in the management of vision-threatening
allowed more precise qualitative and quantitative assess- stages of DR; indeed, anti-VEGF therapies are now first-
ment of structural changes in DME183. OCT ­enables line therapies for DME. However, issues related to the
the objective monitoring of treatment response and cost-effectiveness of anti-VEGF treatment and access to
prognosti­cation (for example, disruption of outer retinal this treatment in developing nations remain. Improved
layers may indicate poor prognosis)184. Newer techno­l­ understanding of the underlying pathophysiological
ogies such as OCT angiography (which also measures mechanisms of DR to find innovative strategies for its
retinal blood flow) may facilitate the assessment of both screening and early treatment would be important to
structural and functional changes in DR185. prevent DR‑associated blindness.

1. Shaw, J. E., Sicree, R. A. & Zimmet, P. Z. Global retinopathy when age at diagnosis is 30 or more years. 18. Varma, R. et al. Visual impairment and blindness
estimates of the prevalence of diabetes for 2010 and Arch. Ophthalmol. 102, 527–532 (1984). avoided with ranibizumab in hispanic and non-
2030. Diabetes Res. Clin. Pract. 87, 4–14 (2010). 11. Yau, J. W. et al. Global prevalence and major risk hispanic whites with diabetic macular edema in the
2. Antonetti, D. A., Klein, R. & Gardner, Diabetic, T. W. factors of diabetic retinopathy. Diabetes Care 35, united states. Ophthalmology 122, 982–989 (2015).
retinopathy. N. Engl. J. Med. 366, 1227–1239 556–564 (2012). 19. Brown, G. C., Brown, M. M., Turpcu, A. & Rajput, Y.
(2012). This article is an individual participant The cost-effectiveness of ranibizumab for the
3. Cheung, N., Mitchell, P. & Wong, T. Y. Diabetic meta-analysis of 35 population-based studies treatment of diabetic macular edema. Ophthalmology
retinopathy. Lancet 376, 124–136 (2010). from around the world, reporting on the 122, 1416–1425 (2015).
4. Klein, R., Lee, K. E., Gangnon, R. E. & Klein, B. E. prevalence and risk factors of DR. 20. Writing Committee for the Diabetic Retinopathy
The 25‑year incidence of visual impairment in type 1 12. Lieth, E., Gardner, T. W., Barber, A. J. Clinical Research Network. Panretinal
diabetes mellitus. the wisconsin epidemiologic study & Antonetti, D. A. Retinal neurodegeneration: photocoagulation versus intravitreous ranibizumab
of diabetic retinopathy. Ophthalmology 117, 63–70 early pathology in diabetes. Clin. Experiment. for proliferative diabetic retinopathy: A randomized
(2010). Ophthalmol. 28, 3–8 (2000). clinical trial. JAMA 314, 2137–2146 (2015).
5. Moss, S. E., Klein, R. & Klein, B. E. The 14‑year 13. Mohamed, Q., Gillies, M. C. & Wong, T. Y. This clinical trial shows that anti-VEGF therapy is
incidence of visual loss in a diabetic population. Management of diabetic retinopathy: a systematic as effective as panretinal laser photocoagulation
Ophthalmology 105, 998–1003 (1998). review. JAMA 298, 902–916 (2007). for proliferative DR.
6. Fenwick, E. K. et al. The impact of diabetic A systematic review of all major trials of treatment 21. James, M., Turner, D. A., Broadbent, D. M., Vora, J.
retinopathy: understanding the patient’s perspective. options for DR. & Harding, S. P. Cost effectiveness analysis of
Br. J. Ophthalmol. 95, 774–782 (2011). 14. Bressler, N. M., Beck, R. W. & Ferris 3rd, F. L. screening for sight threatening diabetic eye disease.
7. Wong, T. Y. et al. Diabetic retinopathy in a multi-ethnic Panretinal photocoagulation for proliferative diabetic BMJ 320, 1627–1631 (2000).
cohort in the United States. Am. J. Ophthalmol. 141, retinopathy. N. Engl. J. Med. 365, 1520–1526 22. Shi, L. et al. Telemedicine for detecting diabetic
446–455 (2006). (2011). retinopathy: a systematic review and meta-analysis.
8. Zhang, X. et al. Prevalence of diabetic retinopathy 15. Frank, R. N. Diabetic retinopathy. N. Engl. J. Med. Br. J. Ophthalmol. 99, 823–831 (2015).
in the United States, 2005–2008. JAMA 304, 350, 48–58 (2004). 23. Klein, R., Klein, B. E., Moss, S. E., Davis, M. D.
649–656 (2010). 16. Mitchell, P. & Wong, T. Y. Management paradigms for & DeMets, D. L. The Wisconsin epidemiologic study
9. Klein, R., Klein, B. E., Moss, S. E., Davis, M. D. diabetic macular edema. Am. J. Ophthalmol. 157, of diabetic retinopathy. IV. Diabetic macular edema.
& DeMets, D. L. The Wisconsin epidemiologic study of 505–513 e1–e8 (2014). Ophthalmology 91, 1464–1474 (1984).
diabetic retinopathy. II. Prevalence and risk of diabetic A review and updated guideline on the 24. Klein, R., Klein, B. E., Moss, S. E. & Cruickshanks, K. J.
retinopathy when age at diagnosis is less than management of DME. The Wisconsin epidemiologic study of diabetic
30 years. Arch. Ophthalmol. 102, 520–526 (1984). 17. Bressler, N. M. et al. Vision-related function after retinopathy: XVII. The 14‑year incidence and
10. Klein, R., Klein, B. E., Moss, S. E., Davis, M. D. ranibizumab treatment for diabetic macular edema: progression of diabetic retinopathy and associated
& DeMets, D. L. The Wisconsin epidemiologic study of results from RIDE and RISE. Ophthalmology 121, risk factors in type 1 diabetes. Ophthalmology 105,
diabetic retinopathy. III. Prevalence and risk of diabetic 2461–2472 (2014). 1801–1815 (1998).

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 13


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

25. Klein, R., Klein, B. E., Moss, S. E. & Cruickshanks, K. J. 48. Zhang, L., Krzentowski, G., Albert, A. & Lefebvre, P. J. 73. Brownlee, M. Biochemistry and molecular cell biology
The Wisconsin epidemiologic study of diabetic Risk of developing retinopathy in Diabetes Control of diabetic complications. Nature 414, 813–820
retinopathy. XV. The long-term incidence of macular and Complications Trial type 1 diabetic patients with (2001).
edema. Ophthalmology 102, 7–16 (1995). good or poor metabolic control. Diabetes Care 24, A seminal paper in which the integrating paradigm
26. Klein, R., Knudtson, M. D., Lee, K. E., Gangnon, R. 1275–1279 (2001). of hyperglycaemia-induced overproduction of
& Klein, B. E. The Wisconsin epidemiologic study 49. Donaghue, K. C. et al. Do all prepubertal years of superoxide by the mitochondrial electron-transport
of diabetic retinopathy: XXII the twenty-five-year diabetes duration contribute equally to diabetes chain is described.
progression of retinopathy in persons with type 1 complications? Diabetes Care 26, 1224–1229 74. Simo, R. & Hernandez, C. Neurodegeneration in the
diabetes. Ophthalmology 115, 1859–1868 (2008). (2003). diabetic eye: new insights and therapeutic perspectives.
27. Stratton, I. M. et al. UKPDS 50: risk factors for 50. Olsen, B. S. et al. The significance of the prepubertal Trends Endocrinol. Metab. 25, 23–33 (2014).
incidence and progression of retinopathy in Type II diabetes duration for the development of retinopathy A comprehensive review of the mechanisms involved
diabetes over 6 years from diagnosis. Diabetologia and nephropathy in patients with type 1 diabetes. in retinal neurodegeneration, the link between
44, 156–163 (2001). J. Diabetes Compl. 18, 160–164 (2004). neurodegeneration and microangiopathy, and the
28. Younis, N., Broadbent, D. M., Vora, J. P. 51. Lim, L. S. & Wong, T. Y. Lipids and diabetic therapeutic implications based on this new concept.
& Harding, S. P. Incidence of sight-threatening retinopathy. Expert Opin. Biol. Ther. 12, 93–105 75. Tang, J. & Kern, T. S. Inflammation in diabetic
retinopathy in patients with type 2 diabetes in the (2012). retinopathy. Prog. Retin. Eye Res. 30, 343–358
Liverpool Diabetic Eye Study: a cohort study. Lancet 52. Benarous, R. et al. Differential association of serum (2011).
361, 195–200 (2003). lipids with diabetic retinopathy and diabetic macular 76. Wilkinson-Berka, J. L., Agrotis, A. & Deliyanti, D. The
29. Jones, C. D., Greenwood, R. H., Misra, A. edema. Invest. Ophthalmol. Vis. Sci. 52, 7464–7469 retinal renin–angiotensin system: roles of angiotensin
& Bachmann, M. O. Incidence and progression (2011). II and aldosterone. Peptides 36, 142–150 (2012).
of diabetic retinopathy during 17 years of a 53. Lyons, T. J. et al. Diabetic retinopathy and serum 77. Kowluru, R. A. & Chan, P. S. Oxidative stress and
population-based screening program in England. lipoprotein subclasses in the DCCT/EDIC cohort. diabetic retinopathy. Exp. Diabetes Res. 2007, 43603
Diabetes Care 35, 592–596 (2012). Invest. Ophthalmol. Vis. Sci. 45, 910–918 (2004). (2007).
30. Kawasaki, R. et al. Incidence and progression of 54. Sasongko, M. B. et al. Serum apolipoprotein AI and B 78. Zhang, S. X., Sanders, E., Fliesler, S. J. & Wang, J. J.
diabetic retinopathy in Japanese adults with type 2 are stronger biomarkers of diabetic retinopathy Endoplasmic reticulum stress and the unfolded protein
diabetes: 8 year follow‑up study of the Japan Diabetes than traditional lipids. Diabetes Care 34, 474–479 responses in retinal degeneration. Exp. Eye Res. 125,
Complications Study (JDCS). Diabetologia 54, (2011). 30–40 (2014).
2288–2294 (2011). 55. Sasongko, M. B. et al. Novel versus traditional risk 79. Riva, C. E., Logean, E. & Falsini, B. Visually evoked
31. Varma, R. et al. Four-year incidence and progression markers for diabetic retinopathy. Diabetologia 55, hemodynamical response and assessment of
of diabetic retinopathy and macular edema: the Los 666–670 (2012). neurovascular coupling in the optic nerve and retina.
Angeles Latino Eye Study. Am. J. Ophthalmol. 149, 56. Wong, C. W., Wong, T. Y., Cheng, C. Y. Prog. Retin. Eye Res. 24, 183–215 (2005).
752–761 e1‑e3 (2010). & Sabanayagam, C. Kidney and eye diseases: common 80. Carrasco, E. et al. Lower somatostatin expression
32. Kempen, J. H. et al. The prevalence of diabetic risk factors, etiological mechanisms, and pathways. is an early event in diabetic retinopathy and is
retinopathy among adults in the United States. Kidney Int. 85, 1290–1302 (2014). associated with retinal neurodegeneration.
Arch. Ophthalmol. 122, 552–563 (2004). 57. Cheung, N. & Wong, T. Y. Obesity and eye diseases. Diabetes Care 30, 2902–2908 (2007).
A meta-analysis of the prevalence of DR in the Surv. Ophthalmol. 52, 180–195 (2007). 81. Garcia-Ramirez, M. et al. Interphotoreceptor
United States. 58. Conway, B. N., Miller, R. G., Klein, R. & Orchard, T. J. retinoid‑binding protein (IRBP) is downregulated at
33. Roy, M. S. et al. The prevalence of diabetic retinopathy Prediction of proliferative diabetic retinopathy early stages of diabetic retinopathy. Diabetologia 52,
among adult type 1 diabetic persons in the United with hemoglobin level. Arch. Ophthalmol. 127, 2633–2641 (2009).
States. Arch. Ophthalmol. 122, 546–551 (2004). 1494–1499 (2009). 82. Lopes de Faria, J. M., Russ, H. & Costa, V. P. Retinal
34. Varma, R. et al. Prevalence of and risk factors for 59. Nguyen, T. T. et al. Inflammatory, hemostatic, nerve fibre layer loss in patients with type 1 diabetes
diabetic macular edema in the United States. and other novel biomarkers for diabetic retinopathy: mellitus without retinopathy. Br. J. Ophthalmol. 86,
JAMA Ophthalmol. 132, 1334–1340 (2014). the multi-ethnic study of atherosclerosis. 725–728 (2002).
35. West, S. K. et al. Diabetes and diabetic retinopathy Diabetes Care 32, 1704–1709 (2009). 83. van Dijk, H. W. et al. Early neurodegeneration in the
in a Mexican-American population: Proyecto VER. 60. Hong, T. et al. Development and progression of retina of type 2 diabetic patients. Invest. Ophthalmol.
Diabetes Care 24, 1204–1209 (2001). diabetic retinopathy 12 months after Vis. Sci. 53, 2715–2719 (2012).
36. Bertelsen, G. et al. Tromso eye study: prevalence and phacoemulsification cataract surgery. Ophthalmology 84. Pulido, J. E. et al. A role for excitatory amino acids in
risk factors of diabetic retinopathy. Acta Ophthalmol. 116, 1510–1514 (2009). diabetic eye disease. Exp. Diabetes Res. 2007, 36150
91, 716–721 (2013). 61. Squirrell, D., Bhola, R., Bush, J., Winder, S. (2007).
37. Thomas, R. L. et al. Prevalence of diabetic retinopathy & Talbot, J. F. A prospective, case controlled study 85. Kusari, J., Zhou, S. X., Padillo, E., Clarke, K. G.
within a national diabetic retinopathy screening of the natural history of diabetic retinopathy and & Gil, D. W. Inhibition of vitreoretinal VEGF elevation
service. Br. J. Ophthalmol. 99, 64–68 (2015). maculopathy after uncomplicated phacoemulsification and blood–retinal barrier breakdown in
38. Jee, D., Lee, W. K. & Kang, S. Prevalence and risk cataract surgery in patients with type 2 diabetes. streptozotocin-induced diabetic rats by brimonidine.
factors for diabetic retinopathy: the Korea National Br. J. Ophthalmol. 86, 565–571 (2002). Invest. Ophthalmol. Vis. Sci. 51, 1044–1051 (2010).
Health and Nutrition Examination Survey 62. Lim, L. S. et al. Are myopic eyes less likely to have 86. Zhang, S. X., Wang, J. J., Gao, G., Parke, K. & Ma, J. X.
2008–2011. Invest. Ophthalmol. Vis. Sci. 54, diabetic retinopathy? Ophthalmology 117, 524–530 Pigment epithelium-derived factor downregulates
6827–6833 (2013). (2010). vascular endothelial growth factor (VEGF) expression
39. Raman, R. et al. Prevalence of diabetic retinopathy 63. Man, R. E. et al. Longer axial length is protective and inhibits VEGF–VEGF receptor 2 binding in diabetic
in india: Sankara Nethralaya Diabetic Retinopathy of diabetic retinopathy and macular edema. retinopathy. J. Mol. Endocrinol. 37, 1–12 (2006).
Epidemiology and Molecular Genetics Study report 2. Ophthalmology 119, 1754–1759 (2012). 87. Cervia, D., Catalani, E., Dal Monte, M. & Casini, G.
Ophthalmology 116, 311–318 (2009). 64. Arar, N. H. et al. Heritability of the severity of diabetic Vascular endothelial growth factor in the ischemic
40. Wang, F. H. et al. Prevalence of diabetic retinopathy retinopathy: the FIND-Eye study. Invest. Ophthalmol. retina and its regulation by somatostatin.
in rural China: the Handan Eye Study. Ophthalmology Vis. Sci. 49, 3839–3845 (2008). J. Neurochem. 120, 818–829 (2012).
116, 461–467 (2009). 65. Hietala, K., Forsblom, C., Summanen, P. 88. Garcia-Ramirez, M., Hernandez, C. & Simo, R.
41. Wong, T. Y. et al. Prevalence and risk factors for & Groop, P. H. Heritability of proliferative diabetic Expression of erythropoietin and its receptor in the
diabetic retinopathy: the Singapore Malay Eye Study. retinopathy. Diabetes 57, 2176–2180 (2008). human retina: a comparative study of diabetic and
Ophthalmology 115, 1869–1875 (2008). 66. Huang, Y. C. et al. Genome-wide association study nondiabetic subjects. Diabetes Care 31, 1189–1194
42. Zheng, Y. et al. Prevalence and risk factors of diabetic of diabetic retinopathy in a Taiwanese population. (2008).
retinopathy in migrant Indians in an urbanized society Ophthalmology 118, 642–648 (2011). 89. Hernandez, C. & Simo, R. Erythropoietin produced
in Asia: the Singapore Indian eye study. 67. Sobrin, L. et al. Candidate gene association study for by the retina: its role in physiology and diabetic
Ophthalmology 119, 2119–2124 (2012). diabetic retinopathy in persons with type 2 diabetes: retinopathy. Endocrine 41, 220–226 (2012).
43. Klein, R. & Klein, B. E. Are individuals with diabetes the Candidate gene Association Resource (CARe). 90. Chen, J., Connor, K. M., Aderman, C. M. & Smith, L. E.
seeing better?: a long-term epidemiological Invest. Ophthalmol. Vis. Sci. 52, 7593–7602 (2011). Erythropoietin deficiency decreases vascular stability
perspective. Diabetes 59, 1853–1860 (2010). 68. Kowluru, R. A., Kowluru, A., Mishra, M. & Kumar, B. in mice. J. Clin. Invest. 118, 526–533 (2008).
44. Wong, T. Y. et al. Rates of progression in diabetic Oxidative stress and epigenetic modifications in the 91. Grant, M. B., Boulton, M. E. & Ljubimov, A. V.
retinopathy during different time periods: a systematic pathogenesis of diabetic retinopathy. Prog. Retin. Erythropoietin: when liability becomes asset in
review and meta-analysis. Diabetes Care 32, Eye Res. 48, 40–61 (2015). neurovascular repair. J. Clin. Invest. 118, 467–470
2307–2313 (2009). 69. Perrone, L., Matrone, C. & Singh, L. P. Epigenetic (2008).

45. Klein, R., Lee, K. E., Knudtson, M. D., Gangnon, R. E. modifications and potential new treatment targets in 92. Chou, J. C., Rollins, S. D., Ye, M., Batlle, D.
& Klein, B. E. Changes in visual impairment prevalence diabetic retinopathy. J. Ophthalmol. 2014, 789120 & Fawzi, A. A. Endothelin receptor-A antagonist
by period of diagnosis of diabetes: the Wisconsin (2014). attenuates retinal vascular and neuroretinal pathology
Epidemiologic Study of Diabetic Retinopathy. 70. Chalmers, J. & Cooper, M. E. UKPDS and the legacy in diabetic mice. Invest. Ophthalmol. Vis. Sci. 55,
Ophthalmology 116, 1937–1942 (2009). effect. N. Engl. J. Med. 359, 1618–1620 (2008). 2516–2525 (2014).
46. Ding, J. & Wong, T. Y. Current epidemiology of diabetic 71. Nathan, D. M. The diabetes control and complications 93. Minton, A. Z. et al. Endothelin B receptors contribute
retinopathy and diabetic macular edema. Curr. Diab. trial/epidemiology of diabetes interventions and to retinal ganglion cell loss in a rat model of glaucoma.
Rep. 12, 346–354 (2012). complications study at 30 years: overview. PLoS ONE 7, e43199 (2012).
47. Keenan, H. A. et al. Clinical factors associated with Diabetes Care 37, 9–16 (2014). 94. Silva, K. C., Pinto, C. C., Biswas, S. K., de Faria, J. B.
resistance to microvascular complications in diabetic 72. Wilkinson, C. P. et al. Proposed international clinical & de Faria, J. M. Hypertension increases retinal
patients of extreme disease duration: the 50‑year diabetic retinopathy and diabetic macular edema inflammation in experimental diabetes: a possible
medalist study. Diabetes Care 30, 1995–1997 disease severity scales. Ophthalmology 110, mechanism for aggravation of diabetic retinopathy
(2007). 1677–1682 (2003). by hypertension. Curr. Eye Res. 32, 533–541 (2007).

14 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

95. Xu, H., Chen, M. & Forrester, J. V. Para-inflammation 118. Scanlon, P. H. The English national screening 136. Chaturvedi, N. et al. Effect of candesartan on
in the aging retina. Prog. Retin. Eye Res. 28, programme for sight-threatening diabetic retinopathy. prevention (DIRECT-Prevent 1) and progression
348–368 (2009). J. Med. Screen. 15, 1–4 (2008). (DIRECT-Protect 1) of retinopathy in type 1 diabetes:
96. Joussen, A. M. et al. Leukocyte-mediated endothelial 119. Looker, H. C. et al. Rates of referable eye disease randomised, placebo-controlled trials. Lancet 372,
cell injury and death in the diabetic retina. in the Scottish National Diabetic Retinopathy 1394–1402 (2008).
Am. J. Pathol. 158, 147–152 (2001). Screening Programme. Br. J. Ophthalmol. 98, 137. Sjolie, A. K. et al. Effect of candesartan on progression
97. McLeod, D. S., Lefer, D. J., Merges, C. & Lutty, G. A. 790–795 (2014). and regression of retinopathy in type 2 diabetes
Enhanced expression of intracellular adhesion 120. Scanlon, P. H. et al. Development of a cost- (DIRECT-Protect 2): a randomised placebo-controlled
molecule‑1 and P‑selectin in the diabetic human effectiveness model for optimisation of the screening trial. Lancet 372, 1385–1393 (2008).
retina and choroid. Am. J. Pathol. 147, 642–653 interval in diabetic retinopathy screening. 138. Mauer, M. et al. Renal and retinal effects of enalapril
(1995). Health Technol. Assess. 19, 1–116 (2015). and losartan in type 1 diabetes. N. Engl. J. Med. 361,
98. Sone, H. et al. Vascular endothelial growth factor is This paper describes the UK DR screening 40–51 (2009).
induced by long-term high glucose concentration and programme and its cost-effectiveness. 139. Keech, A. C. et al. Effect of fenofibrate on the need for
up‑regulated by acute glucose deprivation in cultured 121. Taylor-Phillips, S. et al. Extending the diabetic laser treatment for diabetic retinopathy (FIELD study):
bovine retinal pigmented epithelial cells. Biochem. retinopathy screening interval beyond 1 year: a randomized controlled trial. Lancet 370,
Biophys. Res. Commun. 221, 193–198 (1996). systematic review. Br. J. Ophthalmol. (2015). 1687–1697 (2007).
99. Ferrara, N. Vascular endothelial growth factor: basic 122. Olson, J. et al. Improving the economic value This clinical trial shows that fenofibrate treatment
science and clinical progress. Endocr. Rev. 25, of photographic screening for optical coherence reduces DR progression in T2DM.
581–611 (2004). tomography-detectable macular oedema: 140. Wong, T. Y., Simo, R. & Mitchell, P. Fenofibrate — a
An excellent review of the basic research and a prospective, multicentre, UK study. Health Technol. potential systemic treatment for diabetic retinopathy?
clinical implications of VEGF as a key mediator Assess. 17, 1–142 (2013). Am. J. Ophthalmol. 154, 6–12 (2012).
in DR. 123. Kernt, M. et al. Assessment of diabetic retinopathy 141. Ip, M. S., Domalpally, A., Hopkins, J. J., Wong, P.
100. Takagi, H. et al. Potential role of the angiopoietin/tie2 using nonmydriatic ultra-widefield scanning laser & Ehrlich, J. S. Long-term effects of ranibizumab
system in ischemia-induced retinal neovascularization. ophthalmoscopy (Optomap) compared with ETDRS on diabetic retinopathy severity and progression.
Invest. Ophthalmol. Vis. Sci. 44, 393–402 (2003). 7‑field stereo photography. Diabetes Care 35, Arch. Ophthalmol. 130, 1145–1152 (2012).
101. Simo, R., Carrasco, E., Garcia-Ramirez, M. & 2459–2463 (2012). 142. The Diabetic Retinopathy Study Research Group.
Hernandez, C. Angiogenic and antiangiogenic factors 124. Silva, P. S. et al. Peripheral lesions identified on Photocoagulation treatment of proliferative diabetic
in proliferative diabetic retinopathy. Curr. Diabetes ultrawide field imaging predict increased risk of retinopathy. Clinical application of Diabetic
Rev. 2, 71–98 (2006). diabetic retinopathy progression over 4 years. Retinopathy Study (DRS) findings, DRS Report
102. Grosso, A., Cheung, N., Veglio, F. & Wong, T. Y. Ophthalmology 122, 949–956 (2015). Number 8. Ophthalmology 88, 583–600 (1981).
Similarities and differences in early retinal phenotypes 125. Abramoff, M. D. et al. Automated analysis of retinal This landmark clinical trial shows that panretinal
in hypertension and diabetes. J. Hypertens. 29, images for detection of referable diabetic retinopathy. laser photocoagulation prevents severe vision loss
1667–1675 (2011). JAMA Ophthalmol. 131, 351–357 (2013). in eyes with proliferative DR.
103. Klein, R., Klein, B. E., Moss, S. E. & Wong, T. Y. 126. Abramoff, M. D. et al. Automated early detection 143. Early Treatment Diabetic Retinopathy Study Research
The relationship of retinopathy in persons without of diabetic retinopathy. Ophthalmology 117, Group. Early photocoagulation for diabetic
diabetes to the 15‑year incidence of diabetes 1147–1154 (2010). retinopathy. ETDRS report number 9. Ophthalmology
and hypertension: Beaver Dam Eye Study. Trans. Am. 127. Virk, S. A., Donaghue, K. C., Wong, T. Y. & Craig, M. E. 98, 766–785 (1991).
Ophthalmol. Soc. 104, 98–107 (2006). Interventions for diabetic retinopathy in type 1 This landmark clinical trial shows that focal laser
104. Ojaimi, E. et al. Retinopathy signs in people without diabetes: systematic review and meta-analysis. photocoagulation prevents moderate vision loss
diabetes: the multi-ethnic study of atherosclerosis. Am. J. Ophthalmol. (2015). in eyes with clinically significant DME.
Ophthalmology 118, 656–662 (2011). 128. The Diabetes Control and Complications Trial 144. Nauck, M., Karakiulakis, G., Perruchoud, A. P.,
105. Wong, T. Y. et al. Relation between fasting glucose Research Group. The effect of intensive treatment of Papakonstantinou, E. & Roth, M. Corticosteroids
and retinopathy for diagnosis of diabetes: three diabetes on the development and progression of inhibit the expression of the vascular endothelial
population-based cross-sectional studies. Lancet 371, long‑term complications in insulin-dependent diabetes growth factor gene in human vascular smooth muscle
736–743 (2008). mellitus. N. Engl. J. Med. 329, 977–986 (1993). cells. Eur. J. Pharmacol. 341, 309–315 (1998).
106. Sim, D. A. et al. Patterns of peripheral retinal and This landmark clinical trial is the first to show that 145. Rajendram, R. et al. A 2‑year prospective randomized
central macula ischemia in diabetic retinopathy as tight glycaemic control prevents the development controlled trial of intravitreal bevacizumab or laser
evaluated by ultra-widefield fluorescein angiography. and progression DR in T1DM. therapy (BOLT) in the management of diabetic
Am. J. Ophthalmol. 158, 144–153 (2014). 129. UK Prospective Diabetes Study (UKPDS) Group. macular edema: 24‑month data: report 3.
107. Wessel, M. M. et al. Peripheral retinal ischaemia, Intensive blood-glucose control with sulphonylureas or Arch. Ophthalmol. 130, 972–979 (2012).
as evaluated by ultra-widefield fluorescein insulin compared with conventional treatment and risk 146. Elman, M. J. et al. Expanded 2‑year follow‑up
angiography, is associated with diabetic macular of complications in patients with type 2 diabetes of ranibizumab plus prompt or deferred laser or
oedema. Br. J. Ophthalmol. 96, 694–698 (2012). (UKPDS 33). Lancet 352, 837–853 (1998). triamcinolone plus prompt laser for diabetic macular
108. Holfort, S. K., Jackson, G. R. & Larsen, M. This landmark clinical trial shows that tight edema. Ophthalmology 118, 609–614 (2011).
Dark adaptation during transient hyperglycemia in glycaemic control prevents DR and other 147. Nelson, M. L. et al. Infectious and presumed
type 2 diabetes. Exp. Eye Res. 91, 710–714 (2010). microvascular complications in T2DM. noninfectious endophthalmitis after intravitreal
109. International Council of Ophthalmology (ICO) 130. The Diabetes Control and Complications triamcinolone acetonide injection. Retina 23,
Diabetic Eye Care Committee. ICO Guidelines Trial/Epidemiology of Diabetes Interventions and 686–691 (2003).
for Diabetic Eye Care (ICO, 2014). Complications Research Group. Retinopathy 148. Boyer, D. S. et al. Three-year, randomized,
110. Javitt, J. C. & Aiello, L. P. Cost-effectiveness of and nephropathy in patients with type 1 diabetes four sham‑controlled trial of dexamethasone intravitreal
detecting and treating diabetic retinopathy. years after a trial of intensive therapy. N. Engl. J. Med. implant in patients with diabetic macular edema.
Ann. Intern. Med. 124, 164–169 (1996). 342, 381–389 (2000). Ophthalmology 121, 1904–1914 (2014).
This paper demonstrates the cost-effectiveness The follow‑up of the landmark DCCT clinical trial 149. Campochiaro, P. A. et al. Long-term benefit of
of screening for DR. demonstrates the long-term effectiveness of tight sustained-delivery fluocinolone acetonide vitreous
111. Singer, D. E., Nathan, D. M., Fogel, H. A. glycaemic control in preventing progression of DR inserts for diabetic macular edema. Ophthalmology
& Schachat, A. P. Screening for diabetic retinopathy. in T1DM. 118, 626–635 (2011).
Ann. Intern. Med. 116, 660–671 (1992). 131. Holman, R. R., Paul, S. K., Bethel, M. A., 150. Aiello, L. P. et al. Vascular endothelial growth factor
112. Arun, C. S., Al‑Bermani, A., Stannard, K. & Taylor, R. Matthews, D. R. & Neil, H. A. 10‑year follow‑up of in ocular fluid of patients with diabetic retinopathy
Long-term impact of retinal screening on significant intensive glucose control in type 2 diabetes. N. Engl. and other retinal disorders. N. Engl. J. Med. 331,
diabetes-related visual impairment in the working age J. Med. 359, 1577–1589 (2008). 1480–1487 (1994).
population. Diabet. Med. 26, 489–492 (2009). The follow‑up of the landmark UK Prospective This translational study is the first to demonstrate
113. Backlund, L. B., Algvere, P. V. & Rosenqvist, U. Diabetes Study demonstrates long-term that higher VEGF levels occur in the vitreous in eyes
New blindness in diabetes reduced by more than effectiveness of tight glycaemic control in with proliferative DR; this result led to the
one‑third in Stockholm County. Diabet. Med. 14, preventing progression of DR in T2DM. development of anti-VEGF agents in the treatment
732–740 (1997). 132. The ACCORD Study Group & ACCORD Eye Study of DR.
114. Rohan, T. E., Frost, C. D. & Wald, N. J. Prevention Group. Effects of medical therapies on retinopathy 151. Cheung, N., Wong, I. Y. & Wong, T. Y. Ocular anti-VEGF
of blindness by screening for diabetic retinopathy: progression in type 2 diabetes. N. Engl. J. Med. 363, therapy for diabetic retinopathy: overview of clinical
a quantitative assessment. BMJ 299, 1198–1201 233–244 (2010). efficacy and evolving applications. Diabetes Care 37,
(1989). 133. The ADVANCE Collaborative Group. Intensive blood 900–905 (2014).
115. Williams, G. A. et al. Single-field fundus photography glucose control and vascular outcomes in patients with 152. Wirostko, B., Wong, T. Y. & Simo, R. Vascular
for diabetic retinopathy screening: a report by the type 2 diabetes. N. Engl. J. Med. 358, 2560–2572 endothelial growth factor and diabetic complications.
American Academy of Ophthalmology. Ophthalmology (2008). Prog. Retin. Eye Res. 27, 608–621 (2008).
111, 1055–1062 (2004). 134. UK Prospective Diabetes Study Group. Tight blood 153. Nguyen, Q. D. et al. Ranibizumab for diabetic macular
116. Cavallerano, A. A. et al. A telemedicine program for pressure control and risk of macrovascular and edema: results from 2 phase III randomized trials: RISE
diabetic retinopathy in a Veterans Affairs Medical microvascular complications in type 2 diabetes: and RIDE. Ophthalmology 119, 789–801 (2012).
Center — the Joslin Vision Network Eye Health Care UKPDS 38. BMJ 317, 703–713 (1998). This large Phase III clinical trial demonstrates
Model. Am. J. Ophthalmol. 139, 597–604 (2005). This landmark clinical trial shows that tight blood the effectiveness of anti-VEGF treatment for DME.
117. Bragge, P., Gruen, R. L., Chau, M., Forbes, A. pressure control prevents DR and vision loss 154. Mitchell, P. et al. The RESTORE study: ranibizumab
& Taylor, H. R. Screening for presence or absence in T2DM. monotherapy or combined with laser versus laser
of diabetic retinopathy: a meta-analysis. 135. Tripathi, K. EUCLID study. Lancet 350, 1102–1103 monotherapy for diabetic macular edema.
Arch. Ophthalmol. 129, 435–444 (2011). (1997). Ophthalmology 118, 615–625 (2011).

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 15


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

155. Massin, P. et al. Safety and efficacy of ranibizumab 172. Lloyd, A. et al. Health utility values associated with 188. Huang, O. S. et al. Glycemic and blood pressure
in diabetic macular edema (RESOLVE Study): diabetic retinopathy. Diabet. Med. 25, 618–624 control in an Asian malay population with diabetes
a 12‑month, randomized, controlled, double-masked, (2008). and diabetic retinopathy. Arch. Ophthalmol. 128,
multicenter phase II study. Diabetes Care 33, 173. Torrance, G. W. Measurement of health state utilities 1185–1190 (2010).
2399–2405 (2010). for economic appraisal. J. Health Econ. 5, 1–30 189. Paz, S. H. et al. Noncompliance with vision care
156. Nguyen, Q. D. et al. Two-year outcomes of the (1986). guidelines in Latinos with type 2 diabetes mellitus:
ranibizumab for edema of the mAcula in diabetes 174. Heintz, E., Wirehn, A. B., Peebo, B. B., Rosenqvist, U. the Los Angeles Latino Eye Study. Ophthalmology
(READ‑2) study. Ophthalmology 117, 2146–2151 & Levin, L. A. QALY weights for diabetic retinopathy 113, 1372–1377 (2006).
(2010). — a comparison of health state valuations with HUI‑3, 190. Aiello, L. P. et al. Assessing the effect of personalized
157. Do, D. V. et al. One-year outcomes of the DA VINCI EQ‑5D, EQ‑VAS, and TTO. Value Health 15, 475–484 diabetes risk assessments during ophthalmologic
Study of VEGF Trap-Eye in eyes with diabetic macular (2012). visits on glycemic control: a randomized clinical trial.
edema. Ophthalmology 119, 1658–1665 (2012). 175. Brown, D. M. et al. Long-term outcomes of JAMA Ophthalmol. 133, 888–896 (2015).
158. Korobelnik, J. F. et al. Intravitreal aflibercept for ranibizumab therapy for diabetic macular edema: 191. Wang, S., Tikellis, G., Wong, N., Wong, T. Y.
diabetic macular edema. Ophthalmology 121, the 36‑month results from two phase III trials: RISE & Wang, J. J. Lack of knowledge of glycosylated
2247–2254 (2014). and RIDE. Ophthalmology 120, 2013–2022 (2013). hemoglobin in patients with diabetic retinopathy.
159. The Diabetic Retinopathy Clinical Research Network. 176. Regnier, S. A., Malcolm, W., Haig, J. & Xue, W. Diabetes Res. Clin. Pract. 81, e15–e17 (2008).
Aflibercept, bevacizumab, or ranibizumab for diabetic Cost‑effectiveness of ranibizumab versus aflibercept 192. Diabetes Control and Complications Trial Research
macular edema. N. Engl. J. Med. 372, 1193–1203 in the treatment of visual impairment due to diabetic Group. Progression of retinopathy with intensive
(2015). macular edema: a UK healthcare perspective. versus conventional treatment in the Diabetes Control
This clinical trial demonstrates that different Clinicoecon. Outcomes Res. 7, 235–247 (2015). and Complications Trial. Ophthalmology 102,
anti-VEGF therapies are effective for treating DME. 177. Smiddy, W. E. Clinical applications of cost analysis of 647–661 (1995).
160. Elman, M. J. et al. Intravitreal Ranibizumab for diabetic macular edema treatments. Ophthalmology 193. Best, R. M. & Chakravarthy, U. Diabetic retinopathy
diabetic macular edema with prompt versus deferred 119, 2558–2562 (2012). in pregnancy. Br. J. Ophthalmol. 81, 249–251 (1997).
laser treatment: 5‑year randomized trial results. 178. Stein, J. D. et al. Cost-effectiveness of various 194. Klein, B. E., Moss, S. E. & Klein, R. Effect of pregnancy
Ophthalmology 122, 375–381 (2015). interventions for newly diagnosed diabetic macular on progression of diabetic retinopathy. Diabetes Care
161. Simo, R., Sundstrom, J. M. & Antonetti, D. A. Ocular edema. Ophthalmology 120, 1835–1842 (2013). 13, 34–40 (1990).
Anti-VEGF therapy for diabetic retinopathy: the role 179. Truong, A., Wong, T. Y. & Khachigian, L. M. Emerging 195. Aiello, L. P., Cahill, M. T. & Wong, J. S. Systemic
of VEGF in the pathogenesis of diabetic retinopathy. therapeutic approaches in the management of retinal considerations in the management of diabetic
Diabetes Care 37, 893–899 (2014). angiogenesis and edema. J. Mol. Med. 89, 343–361 retinopathy. Am. J. Ophthalmol. 132, 760–776 (2001).
162. Lim, L. S., Cheung, C. M., Mitchell, P. & Wong, T. Y. (2011). 196. Heier, J. S. et al. Comparison of aflibercept,
Emerging evidence concerning systemic safety of 180. Simo, R. & Hernandez, C. Novel approaches for bevacizumab, and ranibizumab for treatment of
anti‑VEGF agents — should ophthalmologists be treating diabetic retinopathy based on recent diabetic macular edema: extrapolation of data to
concerned? Am. J. Ophthalmol. 152, 329–331 pathogenic evidence. Prog. Retin. Eye Res. 48, clinical practice. JAMA Ophthalmol. 134, 95–99
(2011). 160–180 (2015). (2015).
163. Cheung, N. & Wong, T. Y. Diabetic retinopathy and 181. McAuley, A. K. et al. Vitreous biomarkers in diabetic 197. Hollands, H. et al. Reliability of the time trade-off
systemic vascular complications. Prog. Retin. Eye Res. retinopathy: a systematic review and meta-analysis. technique of utility assessment in patients with retinal
27, 161–176 (2008). J. Diabetes Compl. 28, 419–425 (2014). disease. Can. J. Ophthalmol. 36, 202–209 (2001).
164. Nicholson, B. P. & Schachat, A. P. A review of clinical 182. Zampetaki, A. et al. Angiogenic microRNAs linked
trials of anti-VEGF agents for diabetic retinopathy. to incidence and progression of diabetic retinopathy Acknowledgements
Graefes Arch. Clin. Exp. Ophthalmol. 248, 915–930 in type 1 diabetes. Diabetes 65, 216–227 (2016). This paper was supported by a National Medical Research
(2010). 183. Yeung, L., Lima, V. C., Garcia, P., Landa, G. Council Grant (STaR Award 2013).
165. Thulliez, M. et al. Cardiovascular events and bleeding & Rosen, R. B. Correlation between spectral domain
risk associated with intravitreal antivascular optical coherence tomography findings and fluorescein Author contributions
endothelial growth factor monoclonal antibodies: angiography patterns in diabetic macular edema. Introduction (T.Y.W.); Epidemiology (T.Y.W.); Mechanisms/
systematic review and meta-analysis. Ophthalmology 116, 1158–1167 (2009). pathophysiology (R.S.); Diagnosis, screening and prevention
JAMA Ophthalmol. 132, 1317–1326 (2014). 184. Maheshwary, A. S. et al. The association between (M.L.); Management (C.M.G.C.); Quality of life (S.S.); Outlook
166. Bandello, F. et al. New approaches for the treatment percent disruption of the photoreceptor inner (S.S.); overview of Primer (T.Y.W.).
of diabetic macular oedema: recommendations segment–outer segment junction and visual acuity
by an expert panel. Eye 26, 485–493 (2012). in diabetic macular edema. Am. J. Ophthalmol. 150, Competing interests statement
167. Thomas, B. J., Shienbaum, G., Boyer, D. S. & 63–67 e1 (2010). T.Y.W. is on advisory boards for Abbott, Allergan, Bayer,
Flynn, H. W. Jr. Evolving strategies in the management 185. Ishibazawa, A. et al. Optical coherence tomography Novartis, Pfizer and Solvay, and has received travel, honoraria
of diabetic macular edema: clinical trials and current angiography in diabetic retinopathy: a prospective and research support from these companies. He has no stocks,
management. Can. J. Ophthalmol. 48, 22–30 (2013). pilot study. Am. J. OphthalmoL. 160, 35–44 (2015). equity, contract of employment or named positions on company
168. Avery, R. L. & Gordon, G. M. Systemic safety of 186. Lee, P. P., Feldman, Z. W., Ostermann, J., Brown, D. S. boards. C.M.G.C. is on advisory boards for Allergan, Bayer and
prolonged monthly anti-vascular endothelial growth & Sloan, F. A. Longitudinal rates of annual eye Novartis, and has received travel, honoraria and research
factor therapy for diabetic macular edema: examinations of persons with diabetes and chronic eye support from these companies. She has no stocks, equity,
A systematic review and meta-analysis. diseases. Ophthalmology 110, 1952–1959 (2003). contract of employment or named positions on company
JAMA Ophthalmol 134, 21–29 (2015). This study demonstrates that over a 9‑year period, boards. M.L. is on advisory boards for Allergan, Bayer and
169. Brown, M. M., Brown, G. C., Sharma, S., Landy, J. only just over half of patients with chronic eye Novartis, and has received travel, honoraria and research
& Bakal, J. Quality of life with visual acuity loss from diseases, including diabetes, complied with support from these companies. He has no stocks, equity,
diabetic retinopathy and age-related macular practice guidelines. contract of employment or named positions on company
degeneration. Arch. Ophthalmol. 120, 481–484 187. Bressler, N. M. et al. Underuse of the health care boards. S.S. has served on advisory boards for Allergan, Alcon,
(2002). system by persons with diabetes mellitus and Novartis, Pfizer, Bayer and eSight. He has received travel,
170. Sharma, S. et al. Utilities associated with diabetic diabetic macular edema in the United States. honoraria or research support from these companies. He has
retinopathy: results from a Canadian sample. JAMA Ophthalmol. 132, 168–173 (2014). no stocks, equity, contract of employment or named positions
Br. J. Ophthalmol. 87, 259–261 (2003). This study, based on the US National Health and on company boards. R.S. is on advisory boards of Allergan,
171. Huang, E. S., Brown, S. E., Ewigman, B. G., Foley, E. C. Nutrition Examination Survey conducted between Novo Nordisk and Eli Lilly and Company. In addition, he has
& Meltzer, D. O. Patient perceptions of quality of life 2005 and 2008, showed that a high proportion of received travel and research support from Abbott, Sanofi-
with diabetes-related complications and treatments. people with diabetes in the United States did not Aventis and Astra Zeneca. He has no stocks, equity, contract of
Diabetes Care 30, 2478–2483 (2007). have screening. employment or named positions on company boards.

16 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
CORRECTION

CORRIGENDUM

Diabetic retinopathy
Tien Y. Wong, Chui Ming Gemmy Cheung, Michael Larsen, Sanjay Sharma and Rafael Simó
Nature Reviews Disease Primers article number: 16012; doi:10.1038/nrdp.2016.12; published online 17 March 2016
The affiliation for Sanjay Sharma has now been corrected: Departments of Ophthalmology and Epidemiology,
Queen’s University, Kingston, Ontario, Canada.

NATURE REVIEWS | DISEASE PRIMERS www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like