Purpose: To define unmet needs in ophthalmology that can realistically be addressed in the next 5 years (2019–2025) and describe potential avenues for research to address these challenges. Methods: Outcomes of a consensus process within the European Vision institute (Brussels) are outlined. Disease areas that are discussed comprise glaucoma, retinal dystrophies, diabetic retinopathy, dry eye disease, corneal diseases, cataract and refractive surgery. Results: Unmet needs in the mentioned disease areas are discussed and realistically achievable research projects outlined. Conclusions: Considerable progress can be made in the ophthalmic field and patient-relevant outcomes in the near future.

Vision is the most important sensory function with >30% of the human brain being involved in processing visual information. Consequently, loss of vision ranks highest in the general population’s fear among all possible disabilities [1]. Ophthalmology principally is a very successful part of medicine, where numerous patients can be treated and vision can be restored or maintained in many patients. Nonetheless, there are areas where pathophysiological understanding and consequently treatment options are insufficient.

Only research and medical progress can solve these unmet needs. Relevant disease areas with high prevalence include glaucoma, retinal degenerative diseases, dry eye disease (DED), corneal dystrophies, cataract and refractive diseases.

The European Vision institute is an established “think tank” in ophthalmological research in Europe. As a result of internal discussions, we here set out to define unmet needs in ophthalmology which can realistically be achieved within the time frame of 2019–2025.

The unmet needs should guide policy makers, industry and academia in their endeavours to minimize preventable blindness.

Here we outline unmet needs in ophthalmic research in the areas of glaucoma, retinal dystrophies, diabetic retinopathy (DR), dry eye, corneal diseases and cataract as well as refractive surgery and discuss realistically achievable research projects in these areas.

Non-Intraocular Pressure-Dependent Treatments (Especially Neuroprotection)

The number of pharmacological agents available to treat glaucoma is limited by the few mechanisms of action currently utilized in glaucoma pharmacotherapy. Currently, these all target intraocular pressure (IOP) via reducing aqueous production or increasing the draining from the eye. However, we know that some patients will have disease progression even though their IOP has been sufficiently lowered [2], and similarly, some patients will not ever present with raised IOP but appear to have progressive glaucoma (“Normal-tension” glaucoma) [3]. A further subset of patients will either be resistant to topical treatments to lower the pressure in the eye or intolerant of their side effects. All these anomalies suggest that the underlying process of neurodegeneration is incompletely targeted by IOP lowering and therefore, pharmacologic strategies to protect against glaucomatous neurodegeneration in an IOP-independent manner are an unmet need. These may provide an early method of preventing the initiation of secondary neurodegenerative processes and also a lifeline for those patients who have no further treatment options.

Surrogate End Points

Progression of glaucoma is often slow, and furthermore can take many years to reliably detect and confirm on visual field testing. If follows that demonstrating the efficacy of novel treatments by showing visual field preservation is a long and expensive task, and often prohibitive in clinical trials. It is for this reason that a majority of clinical trials in glaucoma only use IOP as their outcome measure, with the visual benefit for the patient assumed via evidence from studies such as the Ocular Hypertension Treatment Study [2]. There is an unmet need for surrogate markers to represent progression of glaucoma that can be used as a substitute for visual field testing in order to shorten the duration of clinical trials and make them more cost effective, without compromising on the quality of information gained. The considerable amount of attention being given to imaging biomarkers to fulfil this role has been a promising development over the previous decade [4].

Better Outcome Measures in Clinical Trials

The outcome measures in glaucoma are currently suboptimal in a variety of ways. Visual field testing as the current “gold-standard” relies on the patient’s ability to carry out the procedure, requiring a certain amount of learning, physical and mental ability. It is also evident that there is significant delay in the appearance of visual field defects after a considerable loss of retinal nerve fibre layer, at which point neurodegenerative rescue may be less effective [5]. There is an unmet need for new endpoints that can represent the earliest stages of disease without the need to wait for damage to occur to visual function or structural pathways. Universal biomarkers that encompass patients without raised IOP are likely to be most valuable, such as objective imaging of progressive nerve fibre layer thinning, which has a clear pathophysiological involvement in the visual loss experienced in glaucoma [6]. Future methods of in vivo cellular and high-magnification imaging are exciting new avenues of investigation that are being explored in order to detect the underlying retinal ganglion cell loss, prior to manifest clinical significance [7-9].

Early Diagnosis with High Specificity and Sensitivity

The ability to reliably diagnose early glaucoma is an unmet clinical need. The crossover between ocular hypertension and glaucoma results in many patients being followed up and taking daily eyedrops for many years, which will not develop clinically significant disease in their lifetime. However, the risk is that if visual loss occurs, it is currently irreversible. Several advances have suggested that the current biomarkers can be modified to improve their sensitivity for early disease such as alternative visual field testing protocols [10] and imaging of the macular ganglion and nerve fibre layers [11]; however, novel methods have been few and far between. What is being increasingly recognised that may increase the complexity of diagnosis is that glaucoma exists within a spectrum of age-related neurodegenerative conditions including Alzheimer’s disease and Parkinson’s that are also associated with retinal nerve fibre layer thinning [12, 13], however are unlikely to benefit from the lowering of IOP.

IOP Telemetry

The use of one-off IOP measurements in clinical practice is likely to be misleading a variety of scenarios. First, significant variations in IOP occur over seconds, days or months [14]. Therefore, if the clinic appointment is scheduled at a different time of day to the previous appointment, the readings will not be comparable, and furthermore, spikes of high pressure may not be captured within clinic hours. Second, many different factors can effect this reading taken in clinic, including the methods or observer used, as well as physiological factors such as postural changes [15], exercise [16] or some systemic medications. These factors suggests there is an unmet clinical need to widen the availability of continuous IOP measuring devices [17] that will give the clinician a better idea of the range of IOPs that the optic nerve is subject to, and a more comprehensive impression of an individuals’ response to a particular treatment.

More Proven (with Long-Term Efficacy) Minimally-Invasive Glaucoma Surgery

Operations such as trabeculectomy and tube surgery have been the mainstay of surgical glaucoma management for decades. However, not all patients require such dramatic pressure reductions, and the procedures also come with risk of cataract formation, hypotony and endophthalmitis. Minimally invasive glaucoma surgery is a surgical field of interest that has been devised to augment cataract surgery [18]. Given the frequency of cataract operations, the insertion of small devices such as those which bypass the trabecular meshwork [19] or drain into the supraciliary space [20] have become popular in order to reduce the drop burden for the patient following surgery. However, given the small lumens of such devices, it is imaginable that the lifespan of these conduits is limited, and therefore long-term follow-up data is required in order to confirm that these procedures are safe and produce lasting effects. Given the anatomical and pathological variations in angle configuration, there is an unmet need for a wider variety of such surgical procedures to provide long-term IOP-lowering effects, and in more complex cases such as angle-closure glaucoma and secondary disease.

Inherited retinal degenerative diseases (retinal dystrophies; IRDs) are a genetically and phenotypically heterogeneous group of disorders that affect the function of photoreceptor cells [21]. They typically lead to severe visual impairment or blindness early in life, and are the leading cause of legal blindness among working age adults in England and Wales, having replaced DR [22]. Recently, the American Academy of Ophthalmology published recommendations for the clinical assessment of patients with IRDs [23]. Therapeutic options are currently limited, as is the case for many neurodegenerative diseases, but new therapeutic approaches including gene therapy, stem cell therapy, optogenetics and retinal prostheses are on the horizon. Many of these approaches have entered the clinical phase of development and retinal prostheses have received market approval in Europe and the United States [21, 24] (Fig. 1).

Fig. 1.

Current and planned tools for treating blinding diseases. The approximate physical sizes span 6 log units, and the numbers of tools delivered to the eye span 15 log units. The approximate sizes and numbers of tools delivered are shown below each tool. The complexity and sophistication of the tools increase with size, as does the difficulty of delivering treatment across a large retinal surface in vivo. The characteristics of the different tools are shown below the images. Reprinted by permission from [24].

Fig. 1.

Current and planned tools for treating blinding diseases. The approximate physical sizes span 6 log units, and the numbers of tools delivered to the eye span 15 log units. The approximate sizes and numbers of tools delivered are shown below each tool. The complexity and sophistication of the tools increase with size, as does the difficulty of delivering treatment across a large retinal surface in vivo. The characteristics of the different tools are shown below the images. Reprinted by permission from [24].

Close modal

To date, 271 disease genes have been identified [25]. Genetic analysis employing sequencing the coding regions of these gene via panel diagnostics allows to identify the genetic cause in approximately 2 thirds of patients [26]. There is the unmet medical need to improve the sensitivity of testing such that pathogenic mutations are identified in a more substantial fraction of cases, to incorporate improved sequencing methods into research and diagnostic testing, to improve the ability to determine which rare variants are indeed disease-causing, to identify genetic modifiers of disease severity, and to overall improve the access to molecular genetic diagnostic testing, test result evaluation, and genetic counselling, including improved payment mechanisms and more widespread coverage of testing costs [27].

Disease models are needed for the understanding of the pathophysiology and basic mechanisms of IRDs, to identify novel targets for therapy and to provide proof of concept for therapeutic strategies [27]. The most widely used mammalian animal model is the mouse. However, there are differences in retinal cell-type composition, in cell-type-specific gene expression, and in the organization of the retina at the macroscopic level that strongly limit the translation of findings to the human situation [24]. The use of induced pluripotent stem cells (iPSCs) to grow human retinal organoids has provided a platform to study IRDs for which there is no relevant animal model or for which the human mutations have not been recapitulated in an animal model [24, 27]. Further, iPSC models have proven useful in establishing proof of concept when an animal model is absent. For example, the use of iPSC to validate gene augmentation as a therapeutic strategy for choroideremia has resulted in FDA approval of a phase I/II clinical trial (NCT02341807) [27].

The recent FDA approval of gene augmentation therapy for RPE65-associated IRD using adeno-associated viruses is an important milestone and suggests that similar approaches can be used for the treatment of many other genetic forms of IRDs [28]. However, cell-specific targeting remains an unmet need, especially since the human retina contains about 100 different cell types that form specific synaptic connections and reside in functionally distinct microcircuits [29]. Along this line, it has very recently been accomplished to develop a library of 230 adeno-associated viruses, each with a different synthetic promoter, most of them driving an optogenetic tool fused to a fluorescent marker. The marker-tagged optogenetic tool enables cell types to be both identified and manipulated [30]. Also, the best way to deliver gene products to the target cells by, for example, subretinal delivery or intravitreal injection remains to be determined [21].

Although it is accepted by clinicians that IRDs are typically progressive, natural history studies are lacking. The ProgStar [31] and RUSH2A studies are examples to study the progression of disease in a multi-centre approach. For therapy development and clinical monitoring of patients, there is a need to develop and validate reliable outcome measures, endpoints, and/or biomarkers that may be important targets to monitor and modify both for in the earliest possible stages of the degenerative process and along the disease process in general [27]. Such outcome measures or biomarkers must demonstrate change over a relatively short time period spanning no >2–3 years. This also means that we need visual function tests of foveal vision other than visual acuity. In the ProgStar study, the estimate for the specific Stargardt disease patient population was 27 years to lose 3 lines on an Early Treatment of Diabetic Retinopathy Study (ETDRS) visual acuity chart [32]. Also, we need an improved understanding of the relationship between photoreceptor structure and visual function with the goal of understanding the relationship between genotype and clinical phenotype and how many photoreceptors and other retinal neurons need to be restored to provide useful vision [27].

DR is a frequent complication of diabetes and through its sight-threatening complications, that is, macular oedema and proliferative retinopathy, may lead to loss of vision. Indeed, DR is the leading cause of vision loss in working age adults [33].

Recent advances in therapy, particularly intravitreal administration of anti-angiogenic agents, have opened new therapeutic perspectives [34, 35]. However, the availability of treatment only for advanced, sight-threatening stages of the disease and its rate of success make it urgent to understand the initial alterations of DR and their progression in order to be able to treat earlier before vision loss has already occurred [36, 37]. The challenge is to treat and stop disease progression before complications develop or, at least, as soon as they are expected to occur.

The course of DR is not linear and the progression of DR varies in different individuals with the time necessary for the development of sight-threatening complications being much less in some patients than others. The activity of disease and its progress varies from patient to patient [38].

It is recognized that the duration of diabetes and the level of metabolic control determine the development of DR. However, these risk factors do not explain the great variability that characterizes the evolution and rate of progression of the retinopathy in different diabetic individuals. There are many diabetic patients who, after many years with diabetes, never develop sight-threatening retinal changes, whereas other patients progress rapidly.

We have identified 3 major phenotypes of DR progression: one, characterized by slow progression, where neurodegeneration may be the only identified alteration; a second one, characterized by occurrence of oedema, even in the absence of relevant microvascular pathology; a third one identified by increased microaneurysm turnover and the presence of active microvascular lesions [39-41]. In follow-up studies, the first phenotype only rarely progressed to sight-threatening complications, whereas the second phenotype showed a relatively high risk for development of macular oedema, and the third one, shows the higher risk for development of both macular oedema and proliferative retinopathy. This phenotype is identified on OCT angiography (OCTA) by the early presence of retinal capillary dropout (Fig. 2). Recent studies by our group indicate that capillary dropout and its extent is the only retinal alteration that is directly correlated with the severity of DR [42].

Fig. 2.

Example for capillary dropout. a–d Superficial retina plexus (SRP) and deep retina plexus (DRP) images and skeletonization for vessel density measurements, from a 64-year-old male patient of the ETDRS severity level 43–47 group, showing definite capillary dropout on the SRP and on the DRP. e A B-scan in the central macular area.

Fig. 2.

Example for capillary dropout. a–d Superficial retina plexus (SRP) and deep retina plexus (DRP) images and skeletonization for vessel density measurements, from a 64-year-old male patient of the ETDRS severity level 43–47 group, showing definite capillary dropout on the SRP and on the DRP. e A B-scan in the central macular area.

Close modal

It is becoming clear now that only a subset of patients with diabetes who develop retinal changes is expected to progress to advanced retinopathy stages and be at risk to lose functional vision during their lifetime.

Identification of biomarkers of progression of DR to vision-threatening complications is, therefore, a major need. Microaneurysm turnover calculated form fundus photography images and capillary dropout measured by OCTA are major candidates to fulfil this role.

Another need is the validation of biomarkers of visual acuity response to treatment of diabetic macular oedema. Identification and quantification of extracellular fluid in the outer retinal layers show clear potential, but further studies are needed as well as practical and improved functional testing of central vision loss [43-46].

It is now becoming clear that ischaemia, identified by retinal capillary closure, is the central alteration occurring in DR and the one that determines its progression to more severe stages of the disease [42]. Treatments addressing specifically localized ischaemia are expected to stabilize the retinopathy and may stop DR progression. Standardisation of OCTA measurements of capillary dropout and ischaemia are, therefore, crucial to allow the evaluation of potential drugs and a still unmet need.

Other unmet needs include the identification of the changes that may characterize the eyes with macular edema that will respond to anti-VEGF or steroid intravitreal injections. It is believed that the predominance of localized ischaemia or inflammation may determine those eyes that should be more responsive to these different therapies. Again, methods to identify these changes are clearly needed. Vitreous sampling at the time of the injections and rapid analysis of the sample would be ideal if sufficiently safe [47, 48]. Analysis of other fluids such as aqueous humor or tears may offer appealing alternatives if they give reliable results [49-51].

DED [52] and corneal diseases encompass the most common eye diseases where patients seek help from their ophthalmologist as well as the second most common form of blindness worldwide [53]. Although significant progress has been made in some aspects of the field, several important unmet needs need to be addressed in the next 5 years.

Dry Eye Disease

The recent Dry Eye Workshop report [52] outlines current progress in understanding the pathogenesis and therapy of DED. Important unmet needs to be addressed in the near future are as follows:

Optimized Topical Anti-Inflammatory Therapy

Several options for anti-inflammatory therapy including topical (unspecific) steroids, cyclosporine and integrin antagonists are already on the market. Nonetheless, better tolerated and more efficient therapies are needed. This may be achieved using different modes of formulation and/or new anti-inflammatory/immunomodulating agents. Progress can be expected from translation in the field of immunomodulation in neurology and internal medicine.

Pain Relief

New tools are needed for the diagnosis and treatment of peripheral and central neuropathic pain, a novel key feature in dry eye patients [52]. Topical and systemic therapies against receptors triggering and processing pain sensation are needed. Readily established systemically applied pain therapies need to be tested for efficacy in ocular pain. A key aspect here is the essential absence of negative effects on epithelial integrity and wound healing. Such compounds would also be very valuable in refractive surgery (see below).

Artificial Tears

A plethora of substances is already on the market. There exists a need for a personalized approach to substitute only those tear film layers affected in a given patient, isolated or in combination. This should be preservative free and long lasting. Here unmet needs exist in diagnosing the individual tear film deficiency as well as in an individualized approach to replace these layers affected in a given patient.

Blepharitis/Meibomian Gland Dysfunction

Meibomian gland dysfunction contributes to 80% of all DED [52]. A sufficient anti-inflammatory/lipid-modulating local therapy also allowing for release of Meibomian gland obstruction is still missing. Here again progress could be expected using immunomodulation from the fields of dermatology, neurology and rheumatology.

Biomarkers for Diagnosis and Monitoring

Identification of biomarkers and surrogate markers and the consecutive development of point-of-care and imaging devices is crucial for better and more precise diagnosing and stratifying subtypes of disease and disease activity. The use of these tools will further allow the possibility to adapt treatment options in a personalized manner to each individual patient in each individual phase of disease. In the future this should evolve in self-testing devices for use by the patient at home.

Cornea

Corneal transplantation has been revolutionized in recent years by the advent of modern lamellar surgical techniques such as descemet membrane endothelial keratoplasty and DALK [54]. These allow for the safe and efficient restoration of vision in the low-risk scenario, for example, of Fuchs dystrophy and keratoconus with very low rates of immune reactions [55]. Unmet needs exist primarily in the area of vascularized high-risk eyes and eyes with limbal stem cell deficiency.

Topical Anti(lymph)angiogenic Agents in High-Risk Eyes to Promote Graft Survival

There is compelling evidence from preclinical and early clinical studies suggesting a key role of pathologic corneal hem- and lymphangiogenesis in triggering immune reactions after high-risk transplantation [56]. Furthermore, there is clear evidence that targeting these (lymphatic) vessels can promote graft survival [56, 57]. Nonetheless, there is still no licensed topical inhibitor of (lymph)angiogenesis available for use as eye drops for the anterior eye and ocular surface [58], although certain compounds have already been tested in phase III trials [59]. In addition to promoting graft survival, several other indications for topical anti-angiogenesis exist at the ocular surface [54, 57].

Gene Therapy for Corneal Dystrophies

Corneal dystrophies lead to reduced vision, blindness and pain. Although the genetic basis is increasingly better understood [60], current treatments still rely on laser or mechanical surgery and carry the burden of high recurrence rates and costs. Gene therapy, for example, using CRISPR/Cas approaches could be an ideal solution for the treatment of the easily accessible corneal dystrophies.

Anti-inflammatory and Anti-collagenolytic Therapy, for example, in Keratoprosthesis Surgery and Rheumatic Diseases

Several systemic vasculitis- and rheumatology-associated diseases are linked to collagenolysis and melting of the peripheral cornea. The same can occur after artificial corneal transplantation (keratoprosthesis). Here an unmet need exists for topically applicable agents preventing keratolysis. Similarly, an unmet need exists for better tolerated biocorneas to relieve the worldwide donor tissue shortage (see also below [61]). Furthermore, local wound-healing modulators preventing scar formation are needed.

Neurotrophic Keratopathy: Nerve Growth Factor and More

Although with recombinant nerve growth factor eye drops a first agent to treat neurotrophic keratopathy is on the market [62], there is an unmet need for better understanding of neurotrophic keratopathy and for the development of other neurotrophic agents in treatment failures with nerve growth factor eye drops. Here again synergies with neurology might help.

Therapies against Acute and Chronic Corneal Oedema

Although there are surgical approaches for diseases leading to corneal oedema and blindness available (descemet membrane endothelial keratoplasty [54]), pharmacologic approaches are missing avoiding the need for transplantation. Recently isolated temporary corneal lymphangiogenesis was shown to lead to expedited recovery of acute cornea oedema [63]. Here an unmet need exists for the development of topical prolymphangiogenic agents to induce oedema drainage from the cornea (Fig. 3).

Fig. 3.

Transient isolated lymphangiogenesis in acute corneal oedema representing a potential new treatment strategy against acute and chronic corneal oedema (from [63]). ns, not significant. Reprinted by permission from Springer Nature on behalf of Cancer Research UK. *** p < 0.01.

Fig. 3.

Transient isolated lymphangiogenesis in acute corneal oedema representing a potential new treatment strategy against acute and chronic corneal oedema (from [63]). ns, not significant. Reprinted by permission from Springer Nature on behalf of Cancer Research UK. *** p < 0.01.

Close modal

Limbal Stem Cell Deficiency

Although the concept of (limbal) stem cell transplantation was initially developed in ophthalmology, there is still no commercially available stem cell-based therapy, especially not for bilaterally affected patients [64]. Progress has been made in characterizing stem cell phenotypes [65]. Unmet needs exist in delivering reliable stem cell therapies also for bilateral diseases. In addition, approaches to prevent UV-induced damage to stem cells also in the context of pterygium are needed [66].

Biomaterials for the Replacement of Diseased Corneal Tissue

There is a worldwide shortage of donor tissue [61]. Moreover, chronic inflammatory diseases of the cornea and the anterior surface are associated with a high rate of graft failure in almost all variations of corneal tissue transplantation/replacement. There is a high demand for biomaterials that are optically clear, inert to enzymatic degradation and swelling, and that integrate well with diseased corneal tissue.

Conjunctival Diseases

Therapies against Metastasis and Recurrence of Squamous Cell Carcinoma and Conjunctival Melanoma

There is currently no therapy against recurrent or metastasing conjunctival malignancies such as squamous cell carcinoma or melanoma. Progress could be made using approaches from oncology outside of the eye, for example, using murine models [67].

Antiinflammatory Approaches against Cicatricial Diseases of the Ocular Surface (Ocular Pemphigoid)

Several blinding diseases of the eye are associated with conjunctival scarring and inflammation, such as in ocular bullous pemphigoid or Steven Johnson syndrome. Here no effective local anti-inflammatory and/or anti-fibrotic agent is available. Progress can be made using anti-fibrotic/antiinflammatory agents from other areas of medicine.

Artificial Conjunctiva for Ocular Surface Reconstruction

Chronic (autoimmune) inflammatory diseases of the conjunctiva often lead to corneal blindness and are particularly difficult to treat. The accompanying dry eye syndrome prevents a sufficient reconstruction of the corneal surface. New techniques for the generation of conjunctival grafts with low immunologic activity (e.g., autologous cell cultures or cultures from iPSC derived conjunctival epithelial and goblet cells on immunological inert carrier membranes) need further developments before transfer into clinic.

In Cataract Surgery

A spectacular level of safety and efficacy has been achieved in the last decades. Here, realistically addressable unmet needs are as follows:

Phenotyping/genotyping of congenital cataracts

To allow for potential pharmacologic or gene therapies.

Presbyopia

Accommodative lens-filling material allowing for presbyopia free vision even after cataract surgery or pharmacologic approaches for (temporary) near sightedness is needed. Other options include laser-based or corneal shaping-based approaches against presbyopia.

Refractive Surgery

Refractive surgery has achieved an astonishing level of safety and efficacy using Excimer- and Femtosecond laser applications. Nonetheless, unmet needs exist in the following areas:

Topical Pain Relief not Affecting Epithelial Healing

This would allow for a revolution in ocular surface laser ablation avoiding potential flap-based complications.

Non-Invasive Corneal Tissue Modelling Approaches for Customized Reshaping of the Ocular Surface

For example, the use of crosslinking techniques. Here intraoperative OCT-based imaging seems important for monitoring [68].

Standardized therapy for prevention of myopia progression in children.

All authors declare no financial or competing interests or relationships.

This study has been funded by the European Vision institute, Brussels, Belgium.

1.
Scott
AW
,
Bressler
NM
,
Ffolkes
S
,
Wittenborn
JS
,
Jorkasky
J
.
Public Attitudes About Eye and Vision Health
.
JAMA Ophthalmol
.
2016
Oct
;
134
(
10
):
1111
8
.
[PubMed]
2168-6165
2.
Kass
MA
,
Heuer
DK
,
Higginbotham
EJ
,
Johnson
CA
,
Keltner
JL
,
Miller
JP
, et al
The Ocular Hypertension Treatment Study: a randomized trial determines that topical ocular hypotensive medication delays or prevents the onset of primary open-angle glaucoma
.
Arch Ophthalmol
.
2002
Jun
;
120
(
6
):
701
13
.
[PubMed]
0003-9950
3.
Collaborative Normal-Tension Glaucoma Study Group
.
The effectiveness of intraocular pressure reduction in the treatment of normal-tension glaucoma
.
Am J Ophthalmol
.
1998
Oct
;
126
(
4
):
498
505
.
[PubMed]
0002-9394
4.
Weinreb
RN
,
Kaufman
PL
.
Glaucoma research community and FDA look to the future, II: NEI/FDA Glaucoma Clinical Trial Design and Endpoints Symposium: measures of structural change and visual function
.
Invest Ophthalmol Vis Sci
.
2011
Oct
;
52
(
11
):
7842
51
.
[PubMed]
0146-0404
5.
Kuang
TM
,
Zhang
C
,
Zangwill
LM
,
Weinreb
RN
,
Medeiros
FA
.
Estimating Lead Time Gained by Optical Coherence Tomography in Detecting Glaucoma before Development of Visual Field Defects
.
Ophthalmology
.
2015
Oct
;
122
(
10
):
2002
9
.
[PubMed]
0161-6420
6.
Yu
M
,
Lin
C
,
Weinreb
RN
,
Lai
G
,
Chiu
V
,
Leung
CK
.
Risk of Visual Field Progression in Glaucoma Patients with Progressive Retinal Nerve Fiber Layer Thinning: A 5-Year Prospective Study
.
Ophthalmology
.
2016
Jun
;
123
(
6
):
1201
10
.
[PubMed]
0161-6420
7.
Yap
TE
,
Donna
P
,
Almonte
MT
, et al
Real-Time Imaging of Retinal Ganglion Cell Apoptosis.
Cells
2018
; 7 2018/06/20. DOI: .
8.
Rossi
EA
,
Granger
CE
,
Sharma
R
,
Yang
Q
,
Saito
K
,
Schwarz
C
, et al
Imaging individual neurons in the retinal ganglion cell layer of the living eye
.
Proc Natl Acad Sci USA
.
2017
Jan
;
114
(
3
):
586
91
.
[PubMed]
0027-8424
9.
Jonnal
RS
,
Kocaoglu
OP
,
Zawadzki
RJ
, et al
A Review of Adaptive Optics Optical Coherence Tomography: Technical Advances, Scientific Applications, and the Future. Invest Ophthalmol Vis Sci
2016
; 57: OCT51-68. 2016/07/15. DOI: .
10.
De Moraes
CG
,
Hood
DC
,
Thenappan
A
,
Girkin
CA
,
Medeiros
FA
,
Weinreb
RN
, et al
24-2 Visual Fields Miss Central Defects Shown on 10-2 Tests in Glaucoma Suspects, Ocular Hypertensives, and Early Glaucoma
.
Ophthalmology
.
2017
Oct
;
124
(
10
):
1449
56
.
[PubMed]
0161-6420
11.
Kim
YK
,
Ha
A
,
Na
KI
,
Kim
HJ
,
Jeoung
JW
,
Park
KH
.
Temporal Relation between Macular Ganglion Cell-Inner Plexiform Layer Loss and Peripapillary Retinal Nerve Fiber Layer Loss in Glaucoma
.
Ophthalmology
.
2017
Jul
;
124
(
7
):
1056
64
.
[PubMed]
0161-6420
12.
den Haan
J
,
Verbraak
FD
,
Visser
PJ
,
Bouwman
FH
.
Retinal thickness in Alzheimer’s disease: A systematic review and meta-analysis
.
Alzheimers Dement (Amst)
.
2017
Jan
;
6
(
6
):
162
70
.
[PubMed]
2352-8729
13.
Yu
JG
,
Feng
YF
,
Xiang
Y
, et al
Retinal nerve fiber layer thickness changes in Parkinson disease: a meta-analysis. PLoS One
2014
; 9: e85718. 2014/01/28. DOI: .
14.
Kim
JH
,
Caprioli
J
.
Intraocular Pressure Fluctuation: Is It Important?
J Ophthalmic Vis Res
.
2018
Apr-Jun
;
13
(
2
):
170
4
.
[PubMed]
2008-2010
15.
Tarkkanen
A
,
Leikola
J
.
Postural variations of the intraocular pressure as measured with the Mackay-Marg tonometer
.
Acta Ophthalmol (Copenh)
.
1967
;
45
(
4
):
569
75
.
[PubMed]
0001-639X
16.
Qureshi
IA
.
Effects of mild, moderate and severe exercise on intraocular pressure of sedentary subjects
.
Ann Hum Biol
.
1995
Nov-Dec
;
22
(
6
):
545
53
.
[PubMed]
0301-4460
17.
Ittoop
SM
,
SooHoo
JR
,
Seibold
LK
,
Mansouri
K
,
Kahook
MY
.
Systematic Review of Current Devices for 24-h Intraocular Pressure Monitoring
.
Adv Ther
.
2016
Oct
;
33
(
10
):
1679
90
.
[PubMed]
0741-238X
18.
Pillunat
LE
,
Erb
C
,
Jünemann
AG
,
Kimmich
F
.
Micro-invasive glaucoma surgery (MIGS): a review of surgical procedures using stents
.
Clin Ophthalmol
.
2017
Aug
;
11
(
11
):
1583
600
.
[PubMed]
1177-5467
19.
Resende
AF
,
Patel
NS
,
Waisbourd
M
, et al
iStent(R) Trabecular Microbypass Stent: An Update. J Ophthalmol 2016;
2016
: 2731856. 2016/07/15. DOI: .
20.
Vold
S
,
Ahmed
II
,
Craven
ER
,
Mattox
C
,
Stamper
R
,
Packer
M
, et al;
CyPass Study Group
.
Two-Year COMPASS Trial Results: Supraciliary Microstenting with Phacoemulsification in Patients with Open-Angle Glaucoma and Cataracts
.
Ophthalmology
.
2016
Oct
;
123
(
10
):
2103
12
.
[PubMed]
0161-6420
21.
Scholl
HP
,
Strauss
RW
,
Singh
MS
,
Dalkara
D
,
Roska
B
,
Picaud
S
, et al
Emerging therapies for inherited retinal degeneration
.
Sci Transl Med
.
2016
Dec
;
8
(
368
):
368rv6
.
[PubMed]
1946-6234
22.
Liew
G
,
Michaelides
M
,
Bunce
C
.
A comparison of the causes of blindness certifications in England and Wales in working age adults (16-64 years), 1999-2000 with 2009-2010
.
BMJ Open
.
2014
Feb
;
4
(
2
):
e004015
.
[PubMed]
2044-6055
23.
Duncan
, et al
Recommendations on Clinical Assessment of Patients with Inherited Retinal Degenerations – 2016. Available at: https://www.aao.org/clinical-statement/recommendations-on-clinical-assessment-of-patients#Authors. Accessed November 17, 2018.
24.
Roska
B
,
Sahel
JA
.
Restoring vision
.
Nature
.
2018
May
;
557
(
7705
):
359
67
.
[PubMed]
0028-0836
25.
RetNet
. Available at: https://sph.uth.edu/retnet/. Accessed April 26, 2019.
26.
Consugar
MB
,
Navarro-Gomez
D
,
Place
EM
,
Bujakowska
KM
,
Sousa
ME
,
Fonseca-Kelly
ZD
, et al
Panel-based genetic diagnostic testing for inherited eye diseases is highly accurate and reproducible, and more sensitive for variant detection, than exome sequencing
.
Genet Med
.
2015
Apr
;
17
(
4
):
253
61
.
[PubMed]
1098-3600
27.
Duncan
JL
,
Pierce
EA
,
Laster
AM
,
Daiger
SP
,
Birch
DG
,
Ash
JD
, et al;
and the Foundation Fighting Blindness Scientific Advisory Board
.
Inherited Retinal Degenerations: Current Landscape and Knowledge Gaps
.
Transl Vis Sci Technol
.
2018
Jul
;
7
(
4
):
6
.
[PubMed]
2164-2591
28.
Russell
S
,
Bennett
J
,
Wellman
JA
,
Chung
DC
,
Yu
ZF
,
Tillman
A
, et al
Efficacy and safety of voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65-mediated inherited retinal dystrophy: a randomised, controlled, open-label, phase 3 trial
.
Lancet
.
2017
Aug
;
390
(
10097
):
849
60
.
[PubMed]
0140-6736
29.
Azeredo da Silveira
R
,
Roska
B
.
Cell types, circuits, computation
.
Curr Opin Neurobiol
.
2011
Oct
;
21
(
5
):
664
71
.
[PubMed]
0959-4388
30.
Juettner
, et al
Targeting neuronal and glial cell types with synthetic promoter AAVs in mice, non-human primates, and humans
.
[online]
.
bioRxiv
.
2018
.
31.
Strauss
RW
,
Ho
A
,
Muñoz
B
,
Cideciyan
AV
,
Sahel
JA
,
Sunness
JS
, et al;
Progression of Stargardt Disease Study Group
.
The Natural History of the Progression of Atrophy Secondary to Stargardt Disease (ProgStar) Studies: Design and Baseline Characteristics: ProgStar Report No. 1
.
Ophthalmology
.
2016
Apr
;
123
(
4
):
817
28
.
[PubMed]
0161-6420
32.
Kong
X
,
Fujinami
K
,
Strauss
RW
,
Munoz
B
,
West
SK
,
Cideciyan
AV
, et al;
ProgStar Study Group
.
Visual Acuity Change Over 24 Months and Its Association With Foveal Phenotype and Genotype in Individuals With Stargardt Disease: ProgStar Study Report No. 10
.
JAMA Ophthalmol
.
2018
Aug
;
136
(
8
):
920
8
.
[PubMed]
2168-6165
33.
International Diabetes Federeation
(
Diabetes Atlas
ID
, editor
).
2013
. 6th ed.
Brussels, Belgium
:
International Diabetes Federation
;
2013
.[
Internet
], Available from www.idf.org/diabetesatlas
34.
Elman
MJ
,
Aiello
LP
,
Beck
RW
,
Bressler
NM
,
Bressler
SB
,
Edwards
AR
, et al;
Diabetic Retinopathy Clinical Research Network
.
Randomized trial evaluating ranibizumab plus prompt or deferred laser or triamcinolone plus prompt laser for diabetic macular edema
.
Ophthalmology
.
2010
Jun
;
117
(
6
):
1064
1077.e35
.
[PubMed]
0161-6420
35.
Mitchell
P
,
Bandello
F
,
Schmidt-Erfurth
U
,
Lang
GE
,
Massin
P
,
Schlingemann
RO
, et al;
RESTORE study group
.
The RESTORE study: ranibizumab monotherapy or combined with laser versus laser monotherapy for diabetic macular edema
.
Ophthalmology
.
2011
Apr
;
118
(
4
):
615
25
.
[PubMed]
0161-6420
36.
Wells
JA
,
Glassman
AR
,
Ayala
AR
,
Jampol
LM
,
Aiello
LP
,
Antoszyk
AN
, et al;
Diabetic Retinopathy Clinical Research Network
.
Aflibercept, bevacizumab, or ranibizumab for diabetic macular edema
.
N Engl J Med
.
2015
Mar
;
372
(
13
):
1193
203
.
[PubMed]
0028-4793
37.
Figueira
J
,
Silva
R
,
Henriques
J
,
Caldeira Rosa
P
,
Laíns
I
,
Melo
P
, et al
Ranibizumab for High-Risk Proliferative Diabetic Retinopathy: An Exploratory Randomized Controlled Trial
.
Ophthalmologica
.
2016
;
235
(
1
):
34
41
.
[PubMed]
0030-3755
38.
Cunha-Vaz
J
,
Bernardes
R
.
Nonproliferative retinopathy in diabetes type 2. Initial stages and characterization of phenotypes
.
Prog Retin Eye Res
.
2005
May
;
24
(
3
):
355
77
.
[PubMed]
1350-9462
39.
Nunes
S
,
Ribeiro
L
,
Lobo
C
,
Cunha-Vaz
J
.
Three different phenotypes of mild nonproliferative diabetic retinopathy with different risks for development of clinically significant macular edema
.
Invest Ophthalmol Vis Sci
.
2013
Jul
;
54
(
7
):
4595
604
.
[PubMed]
0146-0404
40.
Ribeiro
ML
,
Nunes
SG
,
Cunha-Vaz
JG
.
Microaneurysm turnover at the macula predicts risk of development of clinically significant macular edema in persons with mild nonproliferative diabetic retinopathy
.
Diabetes Care
.
2013
May
;
36
(
5
):
1254
9
.
[PubMed]
0149-5992
41.
Cunha-Vaz
J
,
Ribeiro
L
,
Lobo
C
.
Phenotypes and biomarkers of diabetic retinopathy
.
Prog Retin Eye Res
.
2014
Jul
;
41
:
90
111
.
[PubMed]
1350-9462
42.
Marques
IP
,
Alves
D
,
Santos
T
,
Mendes
L
,
Santos
AR
,
Lobo
C
, et al
Multimodal Imaging of the Initial Stages of Diabetic Retinopathy. Different Disease Pathways in Different Patients
.
Diabetes
.
2018
Dec
;
•••
:
db181077
.
[PubMed]
0012-1797
43.
Cunha-Vaz
J
,
Santos
T
,
Ribeiro
L
,
Alves
D
,
Marques
I
,
Goldberg
M
.
OCT-leakage: A new method to identify and locate abnormal fluid accumulation in diabetic retinal edema
.
Invest Ophthalmol Vis Sci
.
2016
Dec
;
57
(
15
):
6776
83
.
[PubMed]
0146-0404
44.
Cunha-Vaz
J
,
Santos
T
,
Alves
D
,
Marques
I
,
Neves
C
,
Soares
M
, et al
Agreement between OCT Leakage and Fluorescein Angiography to Identify Sites of Alteration of the Blood-Retinal Barrier in Diabetes
.
Ophthalmol Retina
.
2017
Sep - Oct
;
1
(
5
):
395
403
.
[PubMed]
2468-7219
45.
Santos
AR
,
Alves
D
,
Santos
T
,
Figueira
J
,
Silva
R
,
Cunha-Vaz
JG
.
Measurements of Retinal Fluid by OCT Leakage in Diabetic Macular Edema: A Biomarker of Visual Acuity Response to Treatment
.
Retina
.
2019
Jan
;
39
(
1
):
52
60
.
[PubMed]
0275-004X
46.
Santos
AR
,
Ribeiro
L
,
Bandello
F
,
Lattanzio
R
,
Egan
C
,
Frydkjaer-Olsen
U
, et al;
European Consortium for the Early Treatment of Diabetic Retinopathy (EUROCONDOR)
.
Functional and structural findings of neurodegeneration in early stages of diabetic retinopathy: cross-sectional analyses of baseline data of the EUROCONDOR project
.
Diabetes
.
2017
Sep
;
66
(
9
):
2503
10
.
[PubMed]
0012-1797
47.
Yoshimura
T
,
Sonoda
KH
,
Sugahara
M
,
Mochizuki
Y
,
Enaida
H
,
Oshima
Y
, et al
Comprehensive analysis of inflammatory immune mediators in vitreoretinal diseases
.
PLoS One
.
2009
Dec
;
4
(
12
):
e8158
.
[PubMed]
1932-6203
48.
Srividya
G
,
Jain
M
,
Mahalakshmi
K
,
Gayathri
S
,
Raman
R
,
Angayarkanni
N
.
A novel and less invasive technique to assess cytokine profile of vitreous in patients of diabetic macular oedema
.
Eye (Lond)
.
2018
Apr
;
32
(
4
):
820
9
.
[PubMed]
0950-222X
49.
Oh
IK
,
Kim
SW
,
Oh
J
,
Lee
TS
,
Huh
K
.
Inflammatory and angiogenic factors in the aqueous humor and the relationship to diabetic retinopathy
.
Curr Eye Res
.
2010
Dec
;
35
(
12
):
1116
27
.
[PubMed]
0271-3683
50.
Lee
WJ
,
Kang
MH
,
Seong
M
,
Cho
HY
.
Comparison of aqueous concentrations of angiogenic and inflammatory cytokines in diabetic macular oedema and macular oedema due to branch retinal vein occlusion
.
Br J Ophthalmol
.
2012
Nov
;
96
(
11
):
1426
30
.
[PubMed]
0007-1161
51.
Dong
N
,
Xu
B
,
Wang
B
,
Chu
L
.
Study of 27 aqueous humor cytokines in patients with type 2 diabetes with or without retinopathy
.
Mol Vis
.
2013
Aug
;
19
:
1734
46
.
[PubMed]
1090-0535
52.
Craig
JP
,
Nelson
JD
,
Azar
DT
,
Belmonte
C
,
Bron
AJ
,
Chauhan
SK
, et al
TFOS DEWS II Report Executive Summary
.
Ocul Surf
.
2017
Oct
;
15
(
4
):
802
12
.
[PubMed]
1542-0124
53.
Changing Indications in Penetrating Keratoplasty
.
A Systematic Review of 34 Years of Global Reporting. Matthaei M, Sandhaeger H, Hermel M, Adler W, Jun AS, Cursiefen C, Heindl LM
.
Transplantation
.
2017
Jun
;
101
(
6
):
1387
99
.
[PubMed]
0041-1337
54.
Hos
D
,
Matthaei
M
,
Bock
F
,
Maruyama
K
,
Notara
M
,
Clahsen
T
,
Hou
Y
,
Le
VNH
,
Salabarria
AC
,
Horstmann
J
,
Bachmann
BO
,
Cursiefen
C
. Immune reactions after modern lamellar (DALK, DSAEK, DMEK) versus conventional penetrating corneal transplantation. PRER
2019
Jul 3. pii: S1350-9462
55.
Hos
D
,
Tuac
O
,
Schaub
F
,
Stanzel
TP
,
Schrittenlocher
S
,
Hellmich
M
, et al
Incidence and Clinical Course of Immune Reactions after Descemet Membrane Endothelial Keratoplasty: Retrospective Analysis of 1000 Consecutive Eyes
.
Ophthalmology
.
2017
Apr
;
124
(
4
):
512
8
.
[PubMed]
0161-6420
56.
Bock
F
,
Maruyama
K
,
Regenfuss
B
,
Hos
D
,
Steven
P
,
Heindl
LM
, et al
Novel anti(lymph)angiogenic treatment strategies for corneal and ocular surface diseases. Bock F, Maruyama K, Regenfuss B, Hos D, Steven P, Heindl LM, Cursiefen C
.
Prog Retin Eye Res
.
2013
May
;
34
:
89
124
. 1350-9462
57.
Dietrich
T
,
Bock
F
,
Yuen
D
,
Hos
D
,
Bachmann
BO
,
Zahn
G
, et al
Cutting edge: lymphatic vessels, not blood vessels, primarily mediate immune rejections after transplantation
.
J Immunol
.
2010
Jan
;
184
(
2
):
535
9
.
[PubMed]
0022-1767
58.
Cursiefen
C
,
Colin
J
,
Dana
R
,
Diaz-Llopis
M
,
Faraj
LA
,
Garcia-Delpech
S
, et al
Consensus statement on indications for anti-angiogenic therapy in the management of corneal diseases associated with neovascularisation: outcome of an expert roundtable
.
Br J Ophthalmol
.
2012
Jan
;
96
(
1
):
3
9
.
[PubMed]
0007-1161
59.
Cursiefen
C
,
Viaud
E
,
Bock
F
,
Geudelin
B
,
Ferry
A
,
Kadlecová
P
, et al
Aganirsen antisense oligonucleotide eye drops inhibit keratitis-induced corneal neovascularization and reduce need for transplantation: the I-CAN study
.
Ophthalmology
.
2014
Sep
;
121
(
9
):
1683
92
.
[PubMed]
0161-6420
60.
IC3D classification of corneal dystrophies--edition 2. Weiss JS, Møller HU, Aldave AJ, Seitz B, Bredrup C, Kivelä T, Munier FL, Rapuano CJ, Nischal KK, Kim EK, Sutphin J, Busin M, Labbé A, Kenyon KR, Kinoshita S, Lisch W. Cornea.
2015
Feb;34(2):117-59
61.
Ophthalmologe.
2014
Nov;111(11):1027-32. doi: . [Decellularized collagen matrix from tilapia fish scales for corneal reconstruction (BioCornea)]. [Article in German] Hos D1, van Essen TH, Bock F, Chou CH, Pan HA, Lin CC, Huang MC, Chen SC, Cursiefen C, Jager MJ.
62.
Phase II Randomized
.
Double-Masked, Vehicle-Controlled Trial of Recombinant Human Nerve Growth Factor for Neurotrophic Keratitis. Bonini S, Lambiase A, Rama P, Sinigaglia F, Allegretti M, Chao W, Mantelli F; REPARO Study Group
.
Ophthalmology
.
2018
Sep
;
125
(
9
):
1332
43
.
[PubMed]
0161-6420
63.
Transient Ingrowth of Lymphatic Vessels into the Physiologically Avascular Cornea Regulates Corneal Edema and Transparency
.
Hos D, Bukowiecki A, Horstmann J, Bock F, Bucher F, Heindl LM, Siebelmann S, Steven P, Dana R, Eming SA, Cursiefen C
.
Sci Rep
.
2017
Aug
;
7
(
1
):
7227
.
[PubMed]
2045-2322
64.
Rama
P
,
Matuska
S
,
Paganoni
G
,
Spinelli
A
,
De Luca
M
,
Pellegrini
G
.
Limbal stem-cell therapy and long-term corneal regeneration
.
N Engl J Med
.
2010
Jul
;
363
(
2
):
147
55
.
[PubMed]
0028-4793
65.
Notara
M
,
Lentzsch
A
,
Coroneo
M
,
Cursiefen
C
.
The Role of Limbal Epithelial Stem Cells in Regulating Corneal (Lymph)angiogenic Privilege and the Micromilieu of the Limbal Niche following UV Exposure
.
Stem Cells Int
.
2018
May
;
2018
:
8620172
.
[PubMed]
1687-9678
66.
Notara
M
,
Behboudifard
S
,
Kluth
MA
,
Maßlo
C
,
Ganss
C
,
Frank
MH
, et al
UV light-blocking contact lenses protect against short-term UVB-induced limbal stem cell niche damage and inflammation
.
Sci Rep
.
2018
Aug
;
8
(
1
):
12564
.
[PubMed]
2045-2322
67.
Schlereth
SL
,
Iden
S
,
Mescher
M
,
Ksander
BR
,
Bosch
JJ
,
Cursiefen
C
, et al
A Novel Model of Metastatic Conjunctival Melanoma in Immune-Competent Mice
.
Invest Ophthalmol Vis Sci
.
2015
Sep
;
56
(
10
):
5965
73
.
[PubMed]
0146-0404
68.
Siebelmann
S
,
Horstmann
J
,
Scholz
P
,
Bachmann
B
,
Matthaei
M
,
Hermann
M
, et al
Intraoperative changes in corneal structure during excimer laser phototherapeutic keratectomy (PTK) assessed by intraoperative optical coherence tomography
.
Graefes Arch Clin Exp Ophthalmol
.
2018
Mar
;
256
(
3
):
575
81
.
[PubMed]
0721-832X

EVI Steering Board (in addition to authors above): Camiel Boon, Sascha Fauser, Sobha Sivaprasad, Phil Luthert, Jose Sahel, Eberhard Zrenner.

Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.