Open Access
Perspective  |   July 2018
Inherited Retinal Degenerations: Current Landscape and Knowledge Gaps
Author Affiliations & Notes
  • Jacque L. Duncan
    Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
  • Eric A. Pierce
    Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
  • Amy M. Laster
    Foundation Fighting Blindness, Columbia, MD, USA
  • Stephen P. Daiger
    Human Genetics Center, School of Public Health, and Ruiz Department of Ophthalmology and Visual Science, The University of Texas Health Science Center, Houston, TX, USA
  • David G. Birch
    Rose-Silverthorne Retinal Degenerations Laboratory, Retina Foundation of the Southwest, Dallas, TX, USA
  • John D. Ash
    Department of Ophthalmology, University of Florida, Gainesville, FL, USA
  • Alessandro Iannaccone
    Center for Retinal Degenerations and Ophthalmic Genetic Diseases, Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
  • John G. Flannery
    Vision Science, the Helen Wills Neuroscience Institute, the Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
  • José A. Sahel
    Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
    Institut de la Vision-Sorbonne Université, Inserm, CNRS-Paris, France
  • Donald J. Zack
    Departments of Ophthalmology, Neuroscience, Molecular Biology and Genetics, and Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
  • Marco A. Zarbin
    Institute of Ophthalmology and Visual Science, Rutgers-New Jersey Medical School, Rutgers University, Newark, NJ, USA
  • Correspondence: Jacque L. Duncan, MD, Department of Ophthalmology, University of California, San Francisco, 10 Koret Way, K113, San Francisco, CA 94143-0730, USA. e-mail: jacque.duncan@ucsf.edu 
Translational Vision Science & Technology July 2018, Vol.7, 6. doi:https://doi.org/10.1167/tvst.7.4.6
  • Views
  • PDF
  • Share
  • Tools
    • Alerts
      ×
      This feature is available to authenticated users only.
      Sign In or Create an Account ×
    • Get Citation

      Jacque L. Duncan, Eric A. Pierce, Amy M. Laster, Stephen P. Daiger, David G. Birch, John D. Ash, Alessandro Iannaccone, John G. Flannery, José A. Sahel, Donald J. Zack, Marco A. Zarbin, and the Foundation Fighting Blindness Scientific Advisory Board; Inherited Retinal Degenerations: Current Landscape and Knowledge Gaps. Trans. Vis. Sci. Tech. 2018;7(4):6. https://doi.org/10.1167/tvst.7.4.6.

      Download citation file:


      © ARVO (1962-2015); The Authors (2016-present)

      ×
  • Supplements
Introduction
Inherited retinal degenerations (IRDs) represent a diverse group of progressive, visually debilitating diseases that can lead to blindness in which mutations in genes that are critical to retinal function lead to progressive photoreceptor cell death and associated vision loss. IRDs are genetically heterogeneous, with over 260 disease genes identified to date.1 The development of treatments and cures to modify the rate of disease progression has been limited to date, with some success of neurotrophic factor therapy and gene therapies reported from clinical trials.211 The best example of treatment success is gene augmentation therapy for IRD caused by mutations in the RPE65 gene, which recently received US Food and Drug Administration (FDA) approval, which in fact represented the first FDA-approved gene therapy (GT) for any genetically inherited disease.49 Recent developments in the IRD field have advanced understanding of the mechanisms responsible for vision loss, creating new opportunities to intervene in the course of disease by developing new therapeutic approaches. In 2013, a Delphi-style gathering of IRD experts led to the identification, by consensus, of top priorities to advance therapeutic efforts for IRDs, including the need for systematic genotyping, improved standardization of visual function testing, development of more rigorous and widespread data collection protocols, and increased data sharing.12 This document summarizes more recent advances in the IRD field and outlines specific knowledge gaps. These knowledge gaps present opportunities for further investigation to enable development of therapies that may slow down or prevent vision loss, or restore vision, in affected patients. 
Atrophic age-related macular degeneration (AMD) is included among the target inherited retinal diseases of interest because first, understanding AMD may contribute to understanding of inherited macular diseases, and second, understanding of the genetics and mechanism of inherited macular degenerations may contribute to understanding of AMD. 
Recent Advances in IRD Research
The development of treatments for IRDs requires basic and translational research that leads to improved understanding of the nature and causes of these diseases. Brief summaries of recent advances in IRD research are included here. 
Genetic Causes of Disease
Notable progress has been made identifying the genetic causes of IRDs, with over 260 disease genes identified to date.1 By sequencing the coding regions of these disease genes via panel based genetic testing, it is currently possible to identify the genetic cause of disease for approximately two thirds of patients with IRDs1315 and up to 85% of children with IRDs.16 Additional mutations can be identified using whole genome sequencing.17 Active research programs in multiple centers are directed toward identifying the genetic causes of disease in the one third of patients who do not have identifiable mutations in the presently known IRD disease genes. This includes discovery of additional novel disease genes, and identification of noncoding mutations, including structural variants (SVs) in the genome.18 Additionally, the identification and characterization of modifier genes, which themselves do not cause disease but “modify” the disease severity caused by other disease causing mutations is in its infancy, but has great potential for identifying new targets and approaches for treatment. Lastly, it may be worthwhile to evaluate patients with unilateral disease for somatic mutations19,20 or other potential causes of retinal degeneration such as posterior uveitis,21 acute zonal occult outer retinopathy,22 or medication toxicity.23,24 
Disease Pathogenesis
Identification of the genetic causes of IRDs has led to improvements in our understanding of retinal biology in general, and in some cases to our understanding of disease pathogenesis. For example, several cell death mechanisms including apoptosis and necrosis have been shown to be activated in different genetic forms of IRD.25,26 Delineation of the genetic causes of syndromic ciliopathies has led to improved understanding of photoreceptor cell structure, and the importance of cellular transport processes such as intraflagellar transport in IRDs.27,28 Studies of the noncell autonomous nature of cone cell death in rod-cone degenerations has led to recognition of metabolic and oxidative stress in photoreceptor dysfunction and death.29,30 These studies have also identified supportive factors such as RdCVF and NRF2 that could be used to develop nongene specific treatments that may be beneficial to groups of disorders that are caused by mutations in a variety of different genes and that could potentially also help at later stages of the disease process. 
Technical advances in the modeling of disease have facilitated improved understanding of pathophysiology and basic mechanisms of IRDs to identify novel targets for therapy and provide proof of concept for therapeutic strategies. The use of induced pluripotent stem cells (iPSCs) to model disease has provided a platform to study IRDs that do not have a relevant animal model or for which the human mutations have not been recapitulated in an animal model.31 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).32 
Disease Progression
Consensus guidelines for the care of patients with IRDs can be viewed at the American Academy of Ophthalmology Clinical Education Guidelines portal.33 Recent developments that have advanced the retinal degenerations field in clinical structure and function have related to novel technologies that enables improved assessment of retinal structure and function. Optical coherence tomography (OCT) provides noninvasive, objective assessment of retinal structure. The axial resolution is 5 μm with commercially available OCT systems,34 and the outer retinal layers including the outer nuclear layer, the external limiting membrane, the inner segments, the inner segment/outer segment junction or ellipsoid zone (EZ), the outer segments, and the retinal pigment epithelium (RPE) can be measured in eyes with IRD. Numerous studies have established a relationship between visual field parameters and structure on OCT.3540 The objective, quantitative, and high-resolution nature of OCT measures combined with correlation to functional measures has created interest in using the EZ area as a potential clinical trial outcome measure.41 In addition, swept-source OCT penetrates deeper into the choroid to facilitate optical coherence tomography angiography (OCTA),42 which may provide insight into how retinal and choroidal vasculature is affected in eyes with different forms of retinal degeneration. Choroidal abnormalities may play a role in disease pathogenesis in conditions like choroideremia and gyrate atrophy, and may be a target for potential drug delivery. 
Since photoreceptors degenerate in IRDs with consequent visual loss, imaging photoreceptors in living eyes noninvasively could provide insight into rod and cone structure during disease progression and in response to experimental therapies. However, most clinical imaging modalities do not have adequate resolution to visualize individual photoreceptors at the cellular level. Adaptive optics is a strategy to measure aberrations in light exiting the eye that reduce image resolution and prevent images of photoreceptors at the cellular level.34,4345 Adaptive optics scanning laser ophthalmoscopy (AOSLO) captures confocal light wave-guided by photoreceptors with intact inner and outer segments,44,46,47 while split-detector AOSLO images nonconfocally wave-guided light, to visualize cones with only inner segments.4855 Split detector systems can also be used to see inner retinal structures56 and RPE cells using dark field imaging.57 Adaptive optics systems are not widely available but are valuable research tools to improve understanding of photoreceptor survival in eyes with IRD, and may have potential as a sensitive, objective outcome measure of safety and efficacy in clinical trials for IRD patients.58 
Continuing a long history of studies of the course of disease for specific IRDs,5967 several multicenter natural history studies have been initiated, including the ProgStar6874 and RUSH2A trials sponsored by the Foundation Fighting Blindness Clinical Research Institute. These studies have required standardization of equipment and procedures among sites. Registries for patients with rare inherited eye diseases represent an important development that provides a valuable resource for research into genotype-phenotype correlations and for investigators seeking to identify patients who may be eligible to participate in clinical trials.75 My Retina Tracker®, now the largest patient-driven registry in the IRD community, continues to amass genetic data that is accessible to researchers globally.75 
Neuroprotective Agents
Therapies that may slow photoreceptor degeneration due to a range of genetic causes have also been investigated. Vitamin A and docosahexaenoic acid have been demonstrated to provide modest reductions in the rate of disease progression in patients with retinitis pigmentosa (RP).2,3,7682 Oral valproic acid was reported to slow visual field progression in a case series of RP patients,83 but a randomized clinical trial of valproic acid treatment in patients with autosomal dominant RP showed no significant difference between patients treated with valproic acid and placebo (https://www.clinicaltrials.gov/ct2/show/results/NCT01233609?term=01233609&rank=1&sect=X01256#all). 
Advances in high-throughput screening have accelerated the pace of identifying cellular targets and candidate neuroprotective agents. Oxidative damage has been implicated in photoreceptor degeneration,84 and N-acetylcysteine (NAC)85 and N-acetylcysteine amide (NACA)86 have been shown to prevent retinal degeneration in preclinical studies of RP. A clinical trial of NAC in RP patients is in progress (NCT0306302). Nonspecific neurotrophic factor therapy with ciliary neurotrophic factor (CNTF) has been shown to slow photoreceptor degeneration in a number of animal models, but did not demonstrate visual function benefit in human clinical trials of patients with early or advanced RP.87 However, neurotrophic factors that reduce photoreceptor susceptibility to oxidative stress and promote cone outer segment regeneration, such as rod-derived cone viability factor (RdCVF) show promise in preserving cone photoreceptors in eyes with RP in preclinical models,88 and clinical trials are planned in patients with RP. Various protein kinase inhibitors have also been shown to slow retinal degeneration in various mouse and rat models of IRD. 
Gene and Genetic (Gene-Specific) Therapies
Based on progress in understanding the genetic causes of IRDs, significant effort has been directed toward developing gene augmentation therapies for specific genetic forms of IRD. Reports of success of clinical trials of gene augmentation therapy for RPE65- and CHM-associated retinal degeneration suggest the great potential of gene therapies for the treatment of IRDs.10,89,90 The recent FDA approval of gene augmentation therapy for RPE65-associated IRD is an important milestone and suggests that similar approaches can be used for the treatment of many other genetic forms of IRD. For example, preclinical studies support the broad use of gene therapies for the treatment of IRDs, with proof-of-concept of benefit reported in at least 24 genetic forms of disease.91113 Genome editing, including CRISPR-based therapies, and genetically directed pharmacologic therapies, including antisense oligonucleotides, premature termination codon read-through strategies, base editing, and RNA editing are also promising approaches for genetic forms of disease that may be not amenable to gene augmentation therapies.84,112,114120 
Regenerative Medicine
Building on data from studies of retinal development, there is now great interest in the use of retinal stem cells for the study and treatment of IRDs. Recent studies show that instead of or in addition to integration of donor cells into the host retina, transplanted photoreceptor precursors into animal models of retinal degenerative disease demonstrated transfer of cytoplasmic material from donor to recipient photoreceptors.59,121126 Transplanted donor cell integration and material transfer between transplanted and host cells may both underlie the therapeutic benefits associated with transplantation therapy. It may also be possible to take advantage of material transfer between cells for therapeutic purposes, such as stimulation of host Müller cells to differentiate into photoreceptor cells.127 
Another approach to transplantation involves iPSC-derived organoids, which enhances the possibility of autologous transplants.128,129 Retina organoids can be used for disease modeling, as well as for therapeutic purposes.130,131 There are many efforts underway to develop in vitro models of degenerative retinal disease, which will enable high throughput screening of possible therapies, as well as facilitate understanding of disease mechanisms. The National Eye Institute announced the 3-D retina organoid challenge (https://www.nih.gov/news-events/news-releases/nih-solicits-next-generation-retina-organoids-prize-competition). Erin Lavik, Sc.D., won the “ideation phase” of this competition. Lavik's team proposed using a method to screen print tissue models. Her group will create layers of the various types of retinal neurons that can be derived in a lab from adult stem cells. The method would allow the layers to be correctly oriented to mimic the structure of the human retina. 
Approximately 20 early phase clinical trials (www.clinicaltrials.gov) involving cell-based therapy for degenerative retinal disease are underway. Therapeutic targets include the advanced atrophic form of AMD,132 choroidal neovascularization associated with AMD, Stargardt disease, RP, and atrophy associated with high myopia. Therapeutic cells under study in these trials include autologous bone marrow-derived stem cells, human retinal progenitor cells, embryonic stem cell-derived RPE, iPSC-derived RPE, and human CNS stem cells.133 
As a note of caution, there is evidence of severe visual loss including complete blindness after ocular injection of autologous adipose derived “stem cells” from unregulated clinics in the United States.134 There were no clinical trials that supported these treatments and, thus, it remains of utmost importance to wait for the results of formal clinical trials that test the safety and efficacy of these emerging new treatments before patients should undergo any such treatment in a clinical setting. 
Visual Prosthetics
For patients with IRD there have traditionally been no effective treatments to restore vision. In 2013, an epiretinal prosthetic device received human use device (HUD) exemption from the FDA for use in patients with near total vision loss due to RP. Since then, over 200 patients world-wide have received the Argus 2™ device from Second Sight Medical Products, Inc. (Sylmar, CA) with some evidence of improved visual function and performance.135139 Other retinal prostheses that target various regions of the visual pathway are under development or are in clinical trials including the electronic retinal implant Alpha AMS,140,141 Intelligent Retinal Implant System (IRIS V2 (NCT02670980), Suprachoroidal Retinal Prosthesis (NCT01603576), and the PRIMA high-resolution photovoltaic retinal prosthetic system (NCT03333954). 
Optogenetics provides the opportunity to confer novel light-sensing properties to inner retinal neurons that normally have nonimage forming light sensitivity such as bipolar cells and retinal ganglion cells. The strategy involves transfecting inner retinal neurons with a gene encoding a light-sensitive protein such as channelrhodoposin-2 or halorhodopsin. Optogenetic proteins can be sensitive to wavelengths of natural light (Allergan RST-001, NCT02556736) or genetically red-shifted to decrease the potential for light damage by bright white light. In this case, an external visual interface (goggles) transforms external light stimuli into signal that activates the transduced retinal ganglion cells with the appropriate (infrared) wavelength (GenSight Biologics GS030, NCT03326336; GenSight Biologics SA, Paris, France). 
This combination of GT and electronics could offer cell specificity, as well as stimulation of a large number of neurons via viral vector-based transfection technology, delivered by intravitreal injection. The light-sensing receptors may also be built into actual artificial retina implants.142,143 
Specific Knowledge Gaps That Should Be Addressed to Advance the Field
The goal of this analysis of gaps in knowledge regarding IRDs is to identify priority areas for research that will accelerate progress toward development of treatments and cures for IRDs. 
Genetics of IRDs (GE)
The goals of this Research Priority Area include: 
  1.  
    Identification of the genetic causality of all forms of IRDs
  2.  
    Integrate comprehensive genetic testing into clinical care for patients with IRDs
  3.  
    Identify the genetic risk factors for AMD
Identification of the genetic cause of disease is an important part of clinical care for patients with IRDs. Many times, the phenotype of a condition can be ambiguous and molecular genetic diagnosis leads to the accurate clinical diagnosis. A genetic diagnosis can identify potential treatment options for patients, inform them about the potential risk of disease to family members, and identify the potential risk that other organ systems may be affected in syndromic diseases. An important goal in the IRD field is for molecular genetic diagnostic testing to become a routine part of clinical care, and for testing to be accessible, affordable, and accurate.12 A related goal is to improve the sensitivity of testing such that pathogenic or likely pathogenic mutations causing IRDs are identified in a substantial fraction of cases (at least 95%). 
Specific Knowledge Gaps
  • 1.  
    Identify the remaining “elusive” genetic causality of IRDs
  •  
    • a.  
      Identify remaining IRD disease genes
    • b.  
      Identify noncoding mutations in IRD disease genes
As described above, the genetic cause of disease can be found for approximately two thirds of patients via sequencing of the coding regions of known IRD disease genes. The genetic causality for the remaining one third of patients needs to be identified. Based on results of ongoing studies by multiple research groups, it is apparent that the genetic causes of disease in these patients and families will be distributed between novel IRD disease genes and noncoding mutations, including SVs in known disease genes. 
  • 2.  
    Incorporate improved sequencing methods into research and diagnostic testing, including long-read sequencing, single-molecule sequencing, advanced methods to detect copy number variants (CNVs) and other types of SVs, and methods to reconstruct extended haplotypes.
  • 3.  
    Improve the ability to determine which rare variants are damaging, potentially pathogenic, and likely disease-causing to resolve variants of uncertain significance (VUSs), using bioinformatics and computational approaches, collaborative data sharing, and functional assays, including in vitro, cell, and animal based assays.
  • 4.  
    Identify genetic modifiers of disease severity through studies of cohorts of patients with mutations in the same gene but varied disease severity. Many IRDs have variable penetrance, age of onset, progression, and clinical consequences. Even patients who share the same disease gene or mutation may differ substantially in penetrance and clinical expression. While in some cases primary genotype-phenotype correlations have been reported, it is hypothesized that additional genetic modifiers of disease severity exist. Identification of genetic factors modifying clinical consequences may reveal shared disease pathways and novel treatment targets.
  • 5.  
    Improve access to molecular genetic diagnostic testing, test result evaluation, and genetic counseling, including improved payment mechanisms and more widespread coverage of testing costs. This includes the adoption of faster, more accurate, and less expensive methods to identify mutations. Further need to improve communication of test results to patients. Genetic counseling is also an important part of care for patients with IRDs, to help navigate the genetic testing process and interpret the results.
  • 6.  
    Improve contribution of anonymous genetic data to public databases. Continue to support patient data registries such as My Retinal Tracker.
  • 7.  
    Identify additional genetic factors contributing to atrophic AMD, with attention to understudied ethnic/geographic populations, and to genetic factors contributing to specific clinical features and endophenotypes, that is, intermediate disease states and associated biomarkers.
Cell and Molecular Mechanisms of Retinal Disease (CMM)
The goals of this Research Priority Area include: 
  1.  
    Improve our understanding of the mechanisms by which mutations in IRD disease genes cause dysfunction and death of retinal cells so that improved therapies to prevent vision loss can be developed.
  2.  
    Delineate pathways that link mutations in multiple different genes to common disease mechanisms and molecular pathways, with the goal of identifying potentially common therapeutic targets that are applicable to groups of genetic forms of IRD.
  3.  
    Determine mechanisms and pathways by which modifier genes and environmental factors modulate the disease impact of IRD-causing mutations, since such understanding could also identify new targets and pathways for therapeutic intervention.
Specific Knowledge Gaps
  • 1.  
    Improve understanding of the pathways leading to retinal cell dysfunction and death, including:
  •  
    • a.  
      The roles of different cell death pathways such as apoptosis, necrosis, and a distinct form of programmed cell death dependent on poly-ADP-ribose (PAR) overactivation termed parthanatos in IRDs
    • b.  
      The roles of different cell stresses such as oxidative stress and endoplasmic reticulum stress in IRDs
    • c.  
      The roles of inflammation, autoimmunity, and retinal microglia in IRDs
    • d.  
      The role of autophagy in IRDs
    • e.  
      The role of mitochondria dysfunction and energy metabolism in IRDs
  • 2.  
    Improve understanding of regulation of gene expression in retinal cells
  •  
    • a.  
      Investigate how gene expression is altered in different genetic forms of IRD and with aging
    • b.  
      Identify the regulators that control rod and cone differentiation, which could inform efforts to reprogram rod cells to become more cone-like and vice-versa for therapeutic purposes
    • c.  
      Identify the gene expression patterns in all retinal cell types through improved RNA-sequencing methods, including single cell transcriptome analyses
    • d.  
      Define role of epigenetic factors in modulating retinal/photoreceptor health and disease
  • 3.  
    Improve understanding of the metabolism of retinal cells, and how this is altered in disease
  •  
    • a.  
      Identify the unique aspects of photoreceptor and RPE cell metabolism that affect the responses of these cells to genetic disease, and thus are potential therapeutic targets
    • b.  
      Improve understanding of the metabolic interactions of retinal cell types
  • 4.  
    Improve understanding of the consequences of therapeutic drug delivery, including retinal detachments for delivery of therapies and impact on photoreceptor synaptic connectivity
  • 5.  
    Develop and characterize additional animal models of IRDs, including:
  •  
    • a.  
      Genetically modified rodent models of specific genetic forms of IRD
    • b.  
      Nonrodent models of retinal degeneration
    • c.  
      Cone-rich models of disease
    • d.  
      Large animal models of disease
    • e.  
      Determine which animal models best model human disease in terms of both mechanism of disease and which are best for predicting therapeutic efficacy and safety
  • 6.  
    Improve understanding of the noncell autonomous aspects of retinal neurodegeneration
  • 7.  
    Establish model systems for the study of macular degenerations, including AMD
  •  
    • a.  
      Animal and cell models of inherited macular degenerations
    • b.  
      Cell-based models of AMD, such as iPSC-derived RPE cells
  • 8.  
    Identify the molecular events responsible for retinal remodeling during different stages of retinal degeneration, including:
  •  
    • a.  
      Interactions of retinal cells with Müller cells, microglia, and RPE cells
    • b.  
      The impact of these interactions on formation or remodeling of synaptic connections
    • c.  
      Reactive gliosis and loss/remodeling of inner retinal neurons
  • 9.  
    Determine the mechanisms that maintain synapses between photoreceptor and bipolar cells, and that promote new synapse formation. Such understanding will be crucial for the development of regenerative medicine-based treatment approaches.
Clinical-Structure and Function (CL)
The goals of this Research Priority Area include: 
  1.  
    To develop and apply new technology to measure structure and function in IRDs.
  2.  
    To establish relationships between measures of retinal functional test and retinal structure, with the goals of understanding the relationship between genotype and clinical phenotype.
  3.  
    To identify outcome measures or biomarkers to demonstrate change over a relatively short time period spanning no more than 2 to 3 years.
Specific Knowledge Gaps
  • 1.  
    To improve and facilitate new approaches to clinical care for IRD patients and families, including access to care, education of nonexpert clinicians, shared standards for clinical tests, and support for expenses. This includes methods of dissemination to retinal specialists and other eye care professionals of advancements in IRD research.
  • 2.  
    To understand factors modifying penetrance and clinical expression of IRDs.
  •  
    • a.  
      Continue natural history studies of specific disease-causing genes to document the spectrum of clinical consequences. Evaluate mutation-specific variation within these natural history studies.
    • b.  
      Evaluation of possible environmental modifying factors such as diet, smoking, exercise, and sunlight exposure in natural history studies.
  • 3.  
    To identify the earliest manifestations of degeneration.
  •  
    • a.  
      Identify primary cells affected in response to mutations that cause different IRDs.
    • b.  
      To develop and validate reliable outcome measures, endpoints, and/or biomarkers that may be important targets to monitor and modify as initiating processes in degeneration.
    • c.  
      Improved understanding of the relationship between photoreceptor structure and functional vision, including how many photoreceptors and other retinal neurons need to be restored to provide useful vision.
    • d.  
      Improved evaluation of the retinal periphery: earliest changes often occur in the midperiphery. Current structural imaging methods including OCT and AOSLO are limited primarily to the central retina. Improvements in imaging speed and eye movement compensation may facilitate evaluation of midperipheral disease.
    • e.  
      Improved evaluation of rods. Most clinical tests to monitor function are cone mediated, despite the fact that many mutations in RP affect primarily rods. Rod perimetry and microperimetry should be further developed and used to characterize early degeneration. High resolution measures of retinal function during degeneration and responses to therapies need to be developed.
    • f.  
      Improved evaluation of the RPE to determine the relationship between RPE and photoreceptor death. Current methods (shortwave length AF or IR AF) only measure substructures within RPE cells (melanosome or lipofuscin) rather than RPE cell number and shape. Newer imaging techniques, such dark field adaptive optics, hold promise for imaging RPE, but need further refinement.
    • g.  
      Improved evaluation of retinal and choroidal vasculature: utilize OCT angiography to detect and investigate early vascular abnormalities in the retinal and choroidal circulation to correlate perfusion changes with photoreceptor and RPE degeneration.
    • h.  
      Improved image acquisition, processing and analysis, including:
    •  
      • i.  
        Automated OCT segmentation that can more accurately segment the images specific to IRDs. With increased density of B-scans for en-face imaging, manual correction becomes impractical.
      • ii.  
        Better software to easily align multiple structural and functional data such as fundus photography, AF, en face OCT, OCTA, and AO.
      • iii.  
        Development of commercially available instruments that can be deployed and used in multicenter trials.
  • 4.  
    Develop Patient Reported Outcome measures (PROs):
  •  
    • a.  
      Tailored to and validated in specific diseases based on mutation, mechanism, patient age, and stage of progression.
    • b.  
      Assess impact of treatment on patients who have participated in clinical trials.
    • c.  
      Natural history studies provide an opportunity to develop and validate PROs for specific diseases.
  • 5.  
    Improved assessments of vision in patients with advanced disease.
  •  
    • a.  
      Multiluminance mobility tests, pupillometry, and full-field stimulus threshold testing are some examples.
Novel Medical Therapies (NMT)
The goals of this Research Priority Area include: 
  1.  
    To develop drug therapies that protect retinal function and structure in IRDs.
  2.  
    To create and develop improved animal models of human disease to evaluate the effects of NMTs.
  3.  
    To develop better functional testing of drug effectiveness.
  4.  
    To develop novel drug delivery systems.
Specific Knowledge Gaps
  • 1.  
    Study design rigor to improve repeatability of preclinical studies. Lack of reproducibility is largely responsible for basic science developments not moving forward into translational studies.144148
  • 2.  
    Develop targeted high throughput phenotypic drug screening tools relevant to human IRDs.
  • 3.  
    Develop treatments that target different pathways involved in IRD pathogenesis including but not limited to ER stress, proteostasis, or protein editing, alternative splicing, including antisense oligonucleotides to alter splicing of key exons.
  • 4.  
    Develop treatments that promote regeneration or reprogramming of Müller cells, ganglion cells, or RPE cells.
  • 5.  
    Improved drug delivery: develop platforms for sustained delivery methods and formulations to enable penetration and long-term sustained delivery to target cells with minimal off-target side effects.
  • 6.  
    Advancing therapies to clinical trials
  •  
    • a.  
      Develop appropriate animal models of human IRD for testing approaches/compounds.
    • b.  
      Determine optimal patient population to test pan therapeutic treatments.
    • c.  
      Consideration of study of post-mortem samples from patients treated with novel therapies.
Gene Therapy (GT)
The goals of this Research Priority Area include: 
  1.  
    To develop and optimize viral and/or nonviral gene delivery systems for use in the treatment of IRDs.
  2.  
    To demonstrate efficacy and safety using preclinical models in preparation for human trials.
Specific Knowledge Gaps
  • 1.  
    Develop gene editing approaches that can correct multiple genes/classes of mutations, including:
  •  
    • a.  
      Treatment of diseases due to mutations in large genes
    • b.  
      Treatment of diseases due to dominant gain-of-function mutations
    • c.  
      Evaluate and address limitations including efficiency and off target effects
  • 2.  
    Translation from preclinical models to humans:
  •  
    • a.  
      Identify optimal large animal models for preclinical studies
    • b.  
      Identify optimal promoters for expression of therapeutic transgenes at desired levels in appropriate retinal cell types
    • c.  
      Genetic isoform/splice variants expression should be carefully evaluated (i.e., Myo7A in mouse versus human)
    • d.  
      Inflammation varies by species and by vector (i.e., AAV2 vs. AAV4 or AAV5)
    • e.  
      IP issues with specific types of vectors (i.e., AAV8)
  • 3.  
    Careful assessment of GT trial results:
  •  
    • a.  
      Effects of retinal detachment: characterize photoreceptors and synaptic connectivity after detachment
    • b.  
      Evaluate safety and efficacy results
    • c.  
      Evaluate the impact of inflammation and autoimmunity on efficacy results
    • d.  
      Consideration of study of post-mortem samples from patients treated with GT
  • 4.  
    Improved delivery of treatments on a long-term basis
  •  
    • a.  
      Because naturally occurring AAV and lentiviral serotypes are unable to cross existing physical barriers in the retina, subretinal injections are the standard method by which vectors are administered to photoreceptors and RPE cells, the major targets of GT. Subretinal injections have limitations including:
    •  
      • i.  
        Detachment of the retina away from the underlying RPE
      • ii.  
        Delivery of AAV or lentiviral vectors is limited to the area of the retinal detachment
    • b.  
      Intravitreal injections, which do not require damage or detachment of the retina, represent a less invasive and safer approach by which vector could potentially be delivered across the entire retinal surface, while avoiding complications associated with subretinal injection. Challenges associated with intravitreal injection include:
    •  
      • i.  
        High intravitreal viral titers may cause inflammation; it is necessary to manage the immune-mediated inflammatory response treatments can induce
      • ii.  
        Dense vitreous in young subjects is barrier; improve penetration for pan-retinal distribution without increasing clearance of vectors
Regenerative Medicine
The goals of this Research Priority Area include: 
  1.  
    To develop strategies that provide functional rescue or replacement of degenerating or dead retinal cells that can slow and prevent vision loss or restore vision.
Specific Knowledge Gaps
  • 1.  
    Improved production of cells:
  •  
    • a.  
      Developing cells that will survive in hostile environment of advanced degeneration
    • b.  
      Improve understanding of effects of host tissue status on transplant survival and integration
    • c.  
      Understand role of and need for adequate immunosuppression; autologous versus xenograft
  • 2.  
    Clarify the mechanism by which transplanted cells can provide benefit to the host retina
  •  
    • a.  
      Distinguish clearly between material transfer between transplanted and host cells and integration of transplanted cells into the host retina
  • 3.  
    Develop ways to track and improve transplant survival
  •  
    • a.  
      Demonstrate that stem cells persist after implantation with better means of imaging implanted cells
    • b.  
      Distinguish effects of transplant survival from fusion of stem cells with native surviving cells
    • c.  
      Consideration of study of post-mortem samples from patients treated with GT
  • 4.  
    Develop ways to reprogram Müller cells and/or RPE cells that persist despite advanced photoreceptor degeneration
  • 5.  
    Identify improved large animal models to determine if xenograft will be safe and effective in patients
  • 6.  
    Learn from ongoing clinical trials and preclinical studies:
  •  
    • a.  
      Survival of transplants in primates to study fate
    • b.  
      Remodeling in eyes that have received transplanted cells in non-human primates
Addressing the knowledge gap detailed above will likely require projects that incorporate components of multiple project areas because the gaps are synergistic. Making progress to address the gaps will require multipronged, multidisciplinary approaches. Many of the gaps listed pertain to more sections than the ones listed above, and impactful research projects will address gaps and problems from multiple different approaches. 
References
RetNet. Available at: http://www.sph.uth.tmc.edu/RetNet/. Accessed July 9, 2018.
Berson EL, Rosner B, Sandberg MA, et al. A randomized trial of vitamin A and vitamin E supplementation for retinitis pigmentosa. Arch Ophthalmol. 1993; 111: 761–772.
Berson EL, Rosner B, Sandberg MA, et al. Clinical trial of docosahexaenoic acid in patients with retinitis pigmentosa receiving vitamin A treatment. Arch Ophthalmol. 2004; 122: 1297–1305.
Bennett J, Wellman J, Marshall KA, et al. Safety and durability of effect of contralateral-eye administration of AAV2 gene therapy in patients with childhood-onset blindness caused by RPE65 mutations: a follow-on phase 1 trial. Lancet. 2016; 388: 661–672.
Cideciyan AV, Aleman TS, Boye SL, et al. Human gene therapy for RPE65 isomerase deficiency activates the retinoid cycle of vision but with slow rod kinetics. Proc Natl Acad Sci U S A. 2008; 105: 15112–15117.
Jacobson SG, Cideciyan AV, Ratnakaram R, et al. Gene therapy for Leber congenital amaurosis caused by RPE65 mutations: safety and efficacy in 15 children and adults followed up to 3 years. Arch Ophthalmol. 2012; 130: 9–24.
Weleber RG, Pennesi ME, Wilson DJ, et al. Results at 2 years after gene therapy for RPE65-deficient Leber congenital amaurosis and severe early-childhood-onset retinal dystrophy. Ophthalmology. 2016; 123: 1606–1620.
Bainbridge JW, Mehat MS, Sundaram V, et al. Long-term effect of gene therapy on Leber's congenital amaurosis. N Engl J Med. 2015; 372: 1887–1897.
Bainbridge JW, Smith AJ, Barker SS, et al. Effect of gene therapy on visual function in Leber's congenital amaurosis. N Engl J Med. 2008; 358: 2231–2239.
Edwards TL, Jolly JK, Groppe M, et al. Visual acuity after retinal gene therapy for choroideremia. N Engl J Med. 2016; 374: 1996–1998.
MacLaren RE, Groppe M, Barnard AR, et al. Retinal gene therapy in patients with choroideremia: initial findings from a phase 1/2 clinical trial. Lancet. 2014; 383: 1129–1137.
Thompson DA, Ali RR, Banin E, et al. Advancing therapeutic strategies for inherited retinal degeneration: recommendations from the Monaciano Symposium. Invest Ophthalmol Vis Sci. 2015; 56: 918–931.
Consugar MB, Navarro-Gomez D, Place EM, 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; 17: 253–261.
Wang F, Wang H, Tuan HF, et al. Next generation sequencing-based molecular diagnosis of retinitis pigmentosa: identification of a novel genotype-phenotype correlation and clinical refinements. Hum Genet. 2014; 133: 331–345.
Wang X, Wang H, Sun V, et al. Comprehensive molecular diagnosis of 179 Leber congenital amaurosis and juvenile retinitis pigmentosa patients by targeted next generation sequencing. J Med Genet. 2013; 50: 674–688.
Taylor RL, Parry NRA, Barton SJ, et al. Panel-based clinical genetic testing in 85 children with inherited retinal disease. Ophthalmology. 2017; 124: 985–991.
Ellingford JM, Barton S, Bhaskar S, et al. Whole genome sequencing increases molecular diagnostic yield compared with current diagnostic testing for inherited retinal disease. Ophthalmology. 2016; 123: 1143–1150.
Bujakowska KM, Fernandez-Godino R, Place E, et al. Copy-number variation is an important contributor to the genetic causality of inherited retinal degenerations. Genet Med. 2017; 19: 643–651.
Mukhopadhyay R, Holder GE, Moore AT, Webster AR. Unilateral retinitis pigmentosa occurring in an individual with a germline mutation in the RP1 gene. Arch Ophthalmol. 2011; 129: 954–956.
Weller JM, Michelson G, Juenemann AG. Unilateral retinitis pigmentosa: 30 years follow-up. BMJ Case Rep. 2014; 2014.
Lotery AJ, McBride MO, Larkin C, Sharkey JA. Pseudoretinitis pigmentosa due to sub-optimal treatment of neurosyphilis. Eye (Lond). 1996; 10: 759–760.
Jacobson SG, Morales DS, Sun XK, et al. Pattern of retinal dysfunction in acute zonal occult outer retinopathy. Ophthalmology. 1995; 102: 1187–1198.
Lam RW, Remick RA. Pigmentary retinopathy associated with low-dose thioridazine treatment. Can Med Assoc J. 1985; 132: 737.
Mesquita LRC, da Fonseca MLG, da Silva RM, Morizot EH. Panretinal ritonavir-induced retinopathy: a report of long-term use. Retin Cases Brief Rep. 2018. Available at: https://doi.org/10.1097/ICB.0000000000000733. Accessed March 15, 2018.
Chinskey ND, Besirli CG, Zacks DN. Retinal cell death and current strategies in retinal neuroprotection. Curr Opin Ophthalmol. 2014; 25: 228–233.
Murakami Y, Notomi S, Hisatomi T, et al. Photoreceptor cell death and rescue in retinal detachment and degenerations. Prog Retin Eye Res. 2013; 37: 114–140.
Bujakowska KM, Liu Q, Pierce EA. Photoreceptor cilia and retinal ciliopathies. Cold Spring Harb Perspect Biol. 2017; 9.
May-Simera H, Nagel-Wolfrum K, Wolfrum U. Cilia—the sensory antennae in the eye. Prog Retin Eye Res. 2017; 60: 144–180.
Campochiaro PA, Mir TA. The mechanism of cone cell death in retinitis pigmentosa. Prog Retin Eye Res. 2018; 62: 24–37.
Wong-Riley MT. Energy metabolism of the visual system. Eye Brain. 2010; 2: 99–116.
Tucker BA, Scheetz TE, Mullins RF, et al. Exome sequencing and analysis of induced pluripotent stem cells identify the cilia-related gene male germ cell-associated kinase (MAK) as a cause of retinitis pigmentosa. Proc Natl Acad Sci U S A. 2011; 108: E569–E576.
Duong TT, Vasireddy V, Ramachandran P, et al. Use of induced pluripotent stem cell models to probe the pathogenesis of choroideremia and to develop a potential treatment. Stem Cell Res. 2018; 27: 140–150.
Recommendations on Clinical Assessment of Patients with Inherited Retinal Degenerations-2016. American Academy of Ophthalmology Clinical Education/Guidelines/Clinical Statements; 2016. Available at: https://www.aao.org/clinical-statement/recommendations-on-clinical-assessment-of-patients. Accessed July 9, 2018.
Morgan JI. The fundus photo has met its match: optical coherence tomography and adaptive optics ophthalmoscopy are here to stay. Ophthalmic Physiol Opt. 2016; 36: 218–239.
Birch DG, Locke KG, Wen Y, Locke KI, Hoffman DR, Hood DC. Spectral-domain optical coherence tomography measures of outer segment layer progression in patients with X-linked retinitis pigmentosa. JAMA Ophthalmol. 2013; 131: 1143–1150.
Birch DG, Wen Y, Locke K, Hood DC. Rod sensitivity, cone sensitivity, and photoreceptor layer thickness in retinal degenerative diseases. Invest Ophthalmol Vis Sci. 2011; 52: 7141–7147.
Hood DC, Lazow MA, Locke KG, Greenstein VC, Birch DG. The transition zone between healthy and diseased retina in patients with retinitis pigmentosa. Invest Ophthalmol Vis Sci. 2011; 52: 101–108.
Hood DC, Lin CE, Lazow MA, Locke KG, Zhang X, Birch DG. Thickness of receptor and post-receptor retinal layers in patients with retinitis pigmentosa measured with frequency-domain optical coherence tomography. Invest Ophthalmol Vis Sci. 2009; 50: 2328–2336.
Hood DC, Ramachandran R, Holopigian K, Lazow M, Birch DG, Greenstein VC. Method for deriving visual field boundaries from OCT scans of patients with retinitis pigmentosa. Biomed Opt Express. 2011; 2: 1106–1114.
Lazow MA, Hood DC, Ramachandran R, et al. Transition zones between healthy and diseased retina in choroideremia (CHM) and Stargardt disease (STGD) as compared to retinitis pigmentosa (RP). Invest Ophthalmol Vis Sci. 2011; 52: 9581–9590.
Csaky K, Ferris F3rd, Chew EY, Nair P, Cheetham JK, Duncan JL. Report from the NEI/FDA Endpoints Workshop on age-related macular degeneration and inherited retinal diseases. Invest Ophthalmol Vis Sci. 2017; 58: 3456–3463.
Gao SS, Jia Y, Zhang M, et al. Optical coherence tomography angiography. Invest Ophthalmol Vis Sci. 2016; 57: OCT27–36.
Litts KM, Cooper RF, Duncan JL, Carroll J. Photoreceptor-based biomarkers in AOSLO retinal imaging. Invest Ophthalmol Vis Sci. 2017; 58: BIO255–BIO267.
Miller DT, Williams DR, Morris GM, Liang J. Images of cone photoreceptors in the living human eye. Vision Res. 1996; 36: 1067–1079.
Roorda A, Duncan JL. Adaptive optics ophthalmoscopy. Annu Rev Vis Sci. 2015; 1: 19–50.
Roorda A. Adaptive optics ophthalmoscopy. J Refract Surg. 2000; 16: S602–S607.
Roorda A, Williams DR. Optical fiber properties of individual human cones. J Vision. 2002; 2: 404–412.
Scoles D, Flatter JA, Cooper RF, et al. Assessing photoreceptor structure associated with ellipsoid zone disruptions visualized with optical coherence tomography. Retina. 2016; 36: 91–103.
Scoles D, Sulai YN, Langlo CS, et al. In vivo imaging of human cone photoreceptor inner segments. Invest Ophthalmol Vis Sci. 2014; 55: 4244–4251.
Tu JH, Foote KG, Lujan BJ, et al. Dysflective cones: visual function and cone reflectivity in long-term follow-up of acute bilateral foveolitis. Am J Ophthalmol Case Rep. 2017; 7: 14–19.
Wang Q, Tuten WS, Lujan BJ, et al. Adaptive optics microperimetry and OCT images show preserved function and recovery of cone visibility in macular telangiectasia type 2 retinal lesions. Invest Ophthalmol Vis Sci. 2015; 56: 778–786.
Aboshiha J, Dubis AM, Cowing J, et al. A prospective longitudinal study of retinal structure and function in achromatopsia. Invest Ophthalmol Vis Sci. 2014; 55: 5733–5743.
Langlo CS, Erker LR, Parker M, et al. Repeatability and longitudinal assessment of foveal cone structure in Cngb3-associated achromatopsia. Retina. 2017; 37: 1956–1966.
Dubis AM, Cooper RF, Aboshiha J, et al. Genotype-dependent variability in residual cone structure in achromatopsia: toward developing metrics for assessing cone health. Invest Ophthalmol Vis Sci. 2014; 55: 7303–7311.
Scoles D, Sulai YN, Cooper RF, et al. Photoreceptor inner segment morphology in best vitelliform macular dystrophy. Retina. 2017; 37: 741–748.
Rossi EA, Granger CE, Sharma R, et al. Imaging individual neurons in the retinal ganglion cell layer of the living eye. Proc Natl Acad Sci U S A. 2017; 114: 586–591.
Scoles D, Sulai YN, Dubra A. In vivo dark-field imaging of the retinal pigment epithelium cell mosaic. Biomed Opt Express. 2013; 4: 1710–1723.
Talcott KE, Ratnam K, Sundquist SM, et al. Longitudinal study of cone photoreceptors during retinal degeneration and in response to ciliary neurotrophic factor treatment. Invest Ophthalmol Vis Sci. 2011; 52: 2219–2226.
Aleman TS, Han G, Serrano LW, et al. Natural history of the central structural abnormalities in choroideremia: a prospective cross-sectional study. Ophthalmology. 2017; 124: 359–373.
Birch DG, Anderson JL, Fish GE. Yearly rates of rod and cone functional loss in retinitis pigmentosa and cone-rod dystrophy. Ophthalmology. 1999; 106: 258–268.
Cideciyan AV, Swider M, Aleman TS, et al. ABCA4-associated retinal degenerations spare structure and function of the human parapapillary retina. Invest Ophthalmol Vis Sci. 2005; 46: 4739–4746.
Lenassi E, Saihan Z, Cipriani V, et al. Natural history and retinal structure in patients with Usher syndrome type 1 owing to MYO7A mutation. Ophthalmology. 2014; 121: 580–587.
Roesch MT, Ewing CC, Gibson AE, Weber BH. The natural history of X-linked retinoschisis. Can J Ophthalmol. 1998; 33: 149–158.
Sumaroka A, Matsui R, Cideciyan AV, et al. Outer retinal changes including the ellipsoid zone band in usher syndrome 1B due to MYO7A mutations. Invest Ophthalmol Vis Sci. 2016; 57: OCT253–261.
Berson EL. Long-term visual prognoses in patients with retinitis pigmentosa: the Ludwig von Sallmann lecture. Exp Eye Res. 2007; 85: 7–14.
Berson EL, Sandberg MA, Rosner B, Birch DG, Hanson AH. Natural course of retinitis pigmentosa over a three-year interval. Am J Ophthalmol. 1985; 99: 240–251.
Sandberg MA, Rosner B, Weigel-DiFranco C, McGee TL, Dryja TP, Berson EL. Disease course in patients with autosomal recessive retinitis pigmentosa due to the USH2A gene. Invest Ophthalmol Vis Sci. 2008; 49: 5532–5539.
Kong X, Strauss RW, Cideciyan AV, et al. Visual acuity change over 12 months in the prospective progression of atrophy secondary to Stargardt Disease (ProgStar) Study: ProgStar report number 6. Ophthalmology. 2017; 124: 1640–1651.
Kong X, Strauss RW, Michaelides M, et al. Visual acuity loss and associated risk factors in the retrospective progression of Stargardt Disease Study (ProgStar report no. 2). Ophthalmology. 2016; 123: 1887–1897.
Schonbach EM, Ibrahim MA, Kong X, et al. Metrics and acquisition modes for fixation stability as a visual function biomarker. Invest Ophthalmol Vis Sci. 2017; 58: BIO268–BIO276.
Schonbach EM, Wolfson Y, Strauss RW, et al. Macular sensitivity measured with microperimetry in Stargardt disease in the progression of atrophy secondary to Stargardt Disease (ProgStar) Study: report no. 7. JAMA Ophthalmol. 2017; 135: 696–703.
Strauss RW, Ho A, Munoz B, et al. The natural history of the progression of atrophy secondary to Stargardt Disease (ProgStar) Studies: design and baseline characteristics: ProgStar report no. 1. Ophthalmology. 2016; 123: 817–828.
Strauss RW, Munoz B, Ho A, et al. Progression of Stargardt disease as determined by fundus autofluorescence in the retrospective progression of Stargardt Disease Study (ProgStar report no. 9). JAMA Ophthalmol. 2017; 135: 1232–1241.
Strauss RW, Munoz B, Ho A, et al. Incidence of atrophic lesions in Stargardt disease in the progression of atrophy secondary to Stargardt Disease (ProgStar) Study: report no. 5. JAMA Ophthalmol. 2017; 135: 687–695.
My Retina Tracker. Available at: https://www.myretinatracker.org/. Accessed July 9, 2018.
Berson EL, Rosner B, Sandberg MA, et al. Clinical trial of lutein in patients with retinitis pigmentosa receiving vitamin A. Arch Ophthalmol. 2010; 128: 403–411.
Berson EL, Rosner B, Sandberg MA, et al. Further evaluation of docosahexaenoic acid in patients with retinitis pigmentosa receiving vitamin A treatment: subgroup analyses. Arch Ophthalmol. 2004; 122: 1306–1314.
Bok D. The retinal pigment epithelium: a versatile partner in vision. J Cell Sci Suppl. 1993; 17: 189–195.
Massof RW, Finkelstein D. Supplemental vitamin A retards loss of ERG amplitude in retinitis pigmentosa. Arch Ophthalmol. 1993; 111: 751–754.
Berson EL, Weigel-DiFranco C, Rosner B, Gaudio AR, Sandberg MA. Association of vitamin A supplementation with disease course in children with retinitis pigmentosa. JAMA Ophthalmol. 2018; 136: 490–495, https://doi.org/10.1001/jamaophthalmol.2018.0590.
Klaver CCW, Thiadens A. Vitamin A for children with retinitis pigmentosa: an unresolved mystery. JAMA Ophthalmol. 2018; 136: 496–497, https://doi.org/10.1001/jamaophthalmol.2018.0658.
Hoffman DR, Hughbanks-Wheaton DK, Spencer R, et al. Docosahexaenoic acid slows visual field progression in X-linked retinitis pigmentosa: ancillary outcomes of the DHAX Trial. Invest Ophthalmol Vis Sci. 2015; 56: 6646–6653.
Clemson CM, Tzekov R, Krebs M, Checchi JM, Bigelow C, Kaushal S. Therapeutic potential of valproic acid for retinitis pigmentosa. Br J Ophthalmol. 2011; 95: 89–93.
Campochiaro PA, Strauss RW, Lu L, et al. Is there excess oxidative stress and damage in eyes of patients with retinitis pigmentosa? Antioxid Redox Signal. 2015; 23: 643–648.
Lee SY, Usui S, Zafar AB, et al. N-Acetylcysteine promotes long-term survival of cones in a model of retinitis pigmentosa. J Cell Physiol. 2011; 226: 1843–1849.
Schimel AM, Abraham L, Cox D, et al. N-acetylcysteine amide (NACA) prevents retinal degeneration by up-regulating reduced glutathione production and reversing lipid peroxidation. Am J Pathol. 2011; 178: 2032–2043.
Birch DG, Bennett LD, Duncan JL, Weleber RG, Pennesi ME. Long-term follow-up of patients with retinitis pigmentosa (RP) receiving intraocular ciliary neurotrophic factor implants. Am J Ophthalmol. 2016; 170: 10–14, https://doi.org/10.1016/j.ajo.2016.07.013.
Leveillard T, Sahel JA. Rod-derived cone viability factor for treating blinding diseases: from clinic to redox signaling. Sci Transl Med. 2010; 2: 26ps16.
Hohman TC. Hereditary retinal dystrophy. Handb Exp Pharmacol. 2017; 242: 337–367.
Russell S, Bennett J, Wellman JA, 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; 390: 849–860.
Jomary C, Vincent KA, Grist J, Neal MJ, Jones SE. Rescue of photoreceptor function by AAV-mediated gene transfer in a mouse model of inherited retinal degeneration. Gene Ther. 1997; 4: 683–690.
Ali RR, Sarra GM, Stephens C, et al. Restoration of photoreceptor ultrastructure and function in retinal degeneration slow mice by gene therapy. Nat Genet. 2000; 25: 306–310.
Griffey M, Macauley SL, Ogilvie JM, Sands MS. AAV2-mediated ocular gene therapy for infantile neuronal ceroid lipofuscinosis. Mol Ther. 2005; 12: 413–421.
Batten ML, Imanishi Y, Tu DC, et al. Pharmacological and rAAV gene therapy rescue of visual functions in a blind mouse model of Leber congenital amaurosis. PLoS Med. 2005; 2: e333.
Tam LC, Kiang AS, Kennan A, et al. Therapeutic benefit derived from RNAi-mediated ablation of IMPDH1 transcripts in a murine model of autosomal dominant retinitis pigmentosa (RP10). Hum Mol Genet. 2008; 17: 2084–2100.
Pawlyk BS, Bulgakov OV, Liu X, et al. Replacement gene therapy with a human RPGRIP1 sequence slows photoreceptor degeneration in a murine model of Leber congenital amaurosis. Hum Gene Ther. 2010; 21: 993–1004.
Sun X, Pawlyk B, Xu X, et al. Gene therapy with a promoter targeting both rods and cones rescues retinal degeneration caused by AIPL1 mutations. Gene Ther. 2010; 17: 117–131.
Simons DL, Boye SL, Hauswirth WW, Wu SM. Gene therapy prevents photoreceptor death and preserves retinal function in a Bardet-Biedl syndrome mouse model. Proc Natl Acad Sci U S A. 2011; 108: 6276–6281.
Mao H, James TJr, Schwein A, et al. AAV delivery of wild-type rhodopsin preserves retinal function in a mouse model of autosomal dominant retinitis pigmentosa. Hum Gene Ther. 2011; 22: 567–575.
Boye SE, Boye SL, Pang J, et al. Functional and behavioral restoration of vision by gene therapy in the guanylate cyclase-1 (GC1) knockout mouse. PLoS One. 2010; 5: e11306.
Koch S, Sothilingam V, Garcia Garrido M, et al. Gene therapy restores vision and delays degeneration in the CNGB1(-/-) mouse model of retinitis pigmentosa. Hum Mol Genet. 2012; 21: 4486–4496.
Wert KJ, Sancho-Pelluz J, Tsang SH. Mid-stage intervention achieves similar efficacy as conventional early-stage treatment using gene therapy in a pre-clinical model of retinitis pigmentosa. Hum Mol Genet. 2014; 23: 514–523.
Guziewicz KE, Zangerl B, Komaromy AM, et al. Recombinant AAV-mediated BEST1 transfer to the retinal pigment epithelium: analysis of serotype-dependent retinal effects. PLoS One. 2013; 8: e75666.
Seo S, Mullins RF, Dumitrescu AV, et al. Subretinal gene therapy of mice with Bardet-Biedl syndrome type 1. Invest Ophthalmol Vis Sci. 2013; 54: 6118–6132.
Deng WT, Dinculescu A, Li Q, et al. Tyrosine-mutant AAV8 delivery of human MERTK provides long-term retinal preservation in RCS rats. Invest Ophthalmol Vis Sci. 2012; 53: 1895–1904.
Ramachandran PS, Bhattarai S, Singh P, et al. RNA interference-based therapy for spinocerebellar ataxia type 7 retinal degeneration. PLoS One. 2014; 9: e95362.
Pellissier LP, Quinn PM, Alves CH, et al. Gene therapy into photoreceptors and Müller glial cells restores retinal structure and function in CRB1 retinitis pigmentosa mouse models. Hum Mol Genet. 2015; 24: 3104–3118.
Zhong H, Eblimit A, Moayedi Y, et al. AAV8(Y733F)-mediated gene therapy in a Spata7 knockout mouse model of Leber congenital amaurosis and retinitis pigmentosa. Gene Ther. 2015; 22: 619–627.
Choi VW, Bigelow CE, McGee TL, et al. AAV-mediated RLBP1 gene therapy improves the rate of dark adaptation in Rlbp1 knockout mice. Mol Ther Methods Clin Dev. 2015; 2: 15022.
Mookherjee S, Hiriyanna S, Kaneshiro K, et al. Long-term rescue of cone photoreceptor degeneration in retinitis pigmentosa 2 (RP2)-knockout mice by gene replacement therapy. Hum Mol Genet. 2015; 24: 6446–6458.
Scalabrino ML, Boye SL, Fransen KM, et al. Intravitreal delivery of a novel AAV vector targets ON bipolar cells and restores visual function in a mouse model of complete congenital stationary night blindness. Hum Mol Genet. 2015; 24: 6229–6239.
Garanto A, Chung DC, Duijkers L, et al. In vitro and in vivo rescue of aberrant splicing in CEP290-associated LCA by antisense oligonucleotide delivery. Hum Mol Genet. 2016; 25: 2552–2563.
Zhang W, Li L, Su Q, Gao G, Khanna H. Gene therapy using a miniCEP290 fragment delays photoreceptor degeneration in a mouse model of Leber congenital amaurosis. Hum Gene Ther. 2018; 29: 42–50.
Fritsche LG, Igl W, Bailey JN, et al. A large genome-wide association study of age-related macular degeneration highlights contributions of rare and common variants. Nat Genet. 2016; 48: 134–143.
Suzuki K, Tsunekawa Y, Hernandez-Benitez R, et al. In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration. Nature. 2016; 540: 144–149.
Ruan GX, Barry E, Yu D, Lukason M, Cheng SH, Scaria A. CRISPR/Cas9-mediated genome editing as a therapeutic approach for Leber congenital amaurosis 10. Mol Ther. 2017; 25: 331–341.
Li Y, Li T, Li JZ, Wu QS. (2R, 3S)-Pinobanksin-3-cinnamate ameliorates photoreceptor degeneration in Pde6(rd)(10) mice. Cutan Ocul Toxicol. 2017; 36: 273–277.
Nudelman I, Rebibo-Sabbah A, Cherniavsky M, et al. Development of novel aminoglycoside (NB54) with reduced toxicity and enhanced suppression of disease-causing premature stop mutations. J Med Chem. 2009; 52: 2836–2845.
Moosajee M, Tracey-White D, Smart M, et al. Functional rescue of REP1 following treatment with PTC124 and novel derivative PTC-414 in human choroideremia fibroblasts and the nonsense-mediated zebrafish model. Hum Mol Genet. 2016; 25: 3416–3431.
Gaudelli NM, Komor AC, Rees HA, et al. Programmable base editing of A*T to G*C in genomic DNA without DNA cleavage. Nature. 2017; 551: 464–471.
Barber AC, Hippert C, Duran Y, et al. Repair of the degenerate retina by photoreceptor transplantation. Proc Natl Acad Sci U S A. 2013; 110: 354–359.
Pearson RA, Barber AC, Rizzi M, et al. Restoration of vision after transplantation of photoreceptors. Nature. 2012; 485: 99–103.
MacLaren RE, Pearson RA, MacNeil A, et al. Retinal repair by transplantation of photoreceptor precursors. Nature. 2006; 444: 203–207.
Singh MS, Charbel Issa P, Butler R, et al. Reversal of end-stage retinal degeneration and restoration of visual function by photoreceptor transplantation. Proc Natl Acad Sci U S A. 2013; 110: 1101–1106.
Huisingh C, McGwin GJr, Neely D, et al. The association between subretinal drusenoid deposits in older adults in normal macular health and incident age-related macular degeneration. Invest Ophthalmol Vis Sci. 2016; 57: 739–745.
Santos-Ferreira T, Llonch S, Borsch O, Postel K, Haas J, Ader M. Retinal transplantation of photoreceptors results in donor-host cytoplasmic exchange. Nat Commun. 2016; 7: 13028.
Sanges D, Simonte G, Di Vicino U, et al. Reprogramming Müller glia via in vivo cell fusion regenerates murine photoreceptors. J Clin Invest. 2016; 126: 3104–3116.
Eiraku M, Takata N, Ishibashi H, et al. Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature. 2011; 472: 51–56.
Assawachananont J, Mandai M, Okamoto S, et al. Transplantation of embryonic and induced pluripotent stem cell-derived 3D retinal sheets into retinal degenerative mice. Stem Cell Rep. 2014; 2: 662–674.
Llonch S, Carido M, Ader M. Organoid technology for retinal repair. Dev Biol. 2018; 433: 132–143.
Parfitt DA, Lane A, Ramsden CM, et al. Identification and correction of mechanisms underlying inherited blindness in human iPSC-derived optic cups. Cell Stem Cell. 2016; 18: 769–781.
da Cruz L, Fynes K, Georgiadis O, et al. Phase 1 clinical study of an embryonic stem cell-derived retinal pigment epithelium patch in age-related macular degeneration. Nat Biotechnol. 2018; 36: 328–337.
Zarbin M. Cell-based therapy for degenerative retinal disease. Trends Mol Med. 2016; 22: 115–134.
Kuriyan AE, Albini TA, Townsend JH, et al. Vision loss after intravitreal injection of autologous “stem cells” for AMD. N Engl J Med. 2017; 376: 1047–1053.
Dagnelie G, Christopher P, Arditi A, et al. Performance of real-world functional vision tasks by blind subjects improves after implantation with the Argus(R) II retinal prosthesis system. Clin Exp Ophthalmol. 2017; 45: 152–159.
da Cruz L, Dorn JD, Humayun MS, et al. Five-year safety and performance results from the Argus II Retinal Prosthesis System Clinical Trial. Ophthalmology. 2016; 123: 2248–2254.
Geruschat DR, Richards TP, Arditi A, et al. An analysis of observer-rated functional vision in patients implanted with the Argus II Retinal Prosthesis System at three years. Clin Exp Optom. 2016; 99: 227–232.
Ho AC, Humayun MS, Dorn JD, et al. Long-term results from an epiretinal prosthesis to restore sight to the blind. Ophthalmology. 2015; 122: 1547–1554.
da Cruz L, Coley BF, Dorn J, et al. The Argus II epiretinal prosthesis system allows letter and word reading and long-term function in patients with profound vision loss. Br J Ophthalmol. 2013; 97: 632–636.
Edwards TL, Cottriall CL, Xue K, et al. Assessment of the electronic retinal implant alpha AMS in restoring vision to blind patients with end-stage retinitis pigmentosa. Ophthalmology. 2018; 125: 432–443.
Liu Y, Hu H, Liang M, et al. Regulated differentiation of WERI-Rb-1 cells into retinal neuron-like cells. Int J Mol Med. 2017; 40: 1172–1184.
Barrett JM, Berlinguer-Palmini R, Degenaar P. Optogenetic approaches to retinal prosthesis. Vis Neurosci. 2014; 31: 345–354.
Busskamp V, Picaud S, Sahel JA, Roska B. Optogenetic therapy for retinitis pigmentosa. Gene Ther. 2012; 19: 169–175.
Begley CG, Ellis LM. Drug development: raise standards for preclinical cancer research. Nature. 2012; 483: 531–533.
Goodman SN, Fanelli D, Ioannidis JP. What does research reproducibility mean? Sci Transl Med. 2016; 8: 341ps312.
Ioannidis JP. Why most published research findings are false. PLoS Med. 2005; 2: e124.
Mullard A. Reliability of “new drug target” claims called into question. Nat Rev Drug Discov. 2011; 10: 643–644.
Prinz F, Schlange T, Asadullah K. Believe it or not: how much can we rely on published data on potential drug targets? Nat Rev Drug Discov. 2011; 10: 712.
×
×

This PDF is available to Subscribers Only

Sign in or purchase a subscription to access this content. ×

You must be signed into an individual account to use this feature.

×