Abstract
Due to improved phenotyping and genetic characterization, the field of ‘incurable’ and ‘blinding’ inherited retinal diseases (IRDs) has moved substantially forward. Decades of ascertainment of IRD patient data from Philadelphia and Toronto centers illustrate the progress from Mendelian genetic types to molecular diagnoses. Molecular genetics have been used not only to clarify diagnoses and to direct counseling but also to enable the first clinical trials of gene-based treatment in these diseases. An overview of the recent reports of gene augmentation clinical trials by subretinal injections is used to reflect on the reasons why there has been limited success in this early venture into therapy. These first-in human experiences have taught that there is a need for advancing the techniques of delivery of the gene products - not only for refining further subretinal trials, but also for evaluating intravitreal delivery. Candidate IRDs for intravitreal gene delivery are then suggested to illustrate some of the disorders that may be amenable to improvement of remaining central vision with the least photoreceptor trauma. A more detailed understanding of the human IRDs to be considered for therapy and the calculated potential for efficacy should be among the routine prerequisites for initiating a clinical trial.
Keywords: Retinitis pigmentosa, gene therapy, molecular mechanisms, genetic retinal degenerations, Leber congenital amaurosis, RPE65, MERTK, MYO7A, ABCA4, CNGA3, CNGB3, PDE6B, RLBP1, REP1, RPGR, TULP1, NPHP5, RPGRIP1, BCM, OPN1LW, OPN1MW
1. Introduction
After a relatively long journey of clinical and scientific progress, we have arrived at a time when therapeutic strategies are not only being discussed but also initiated for previously incurable orphan genetic retinal diseases. In the past three decades, there has been an ever-increasing list of causative genes associated with inherited retinal diseases (IRDs) and this list has been continually updated in reviews (for example, Iannaccone, 2005; Bramall et al., 2010; Wright et al., 2010; Verbakel et al., 2018) in addition to websites (Retnet, https://sph.uth.edu/retnet/; European Retinal Disease Consortium, https://www.erdc.info). Many reports have now tabulated ongoing and planned clinical trials of gene-based therapies for IRDs (for example, Trapani and Auricchio, 2018; Arbabi et al., 2019; Lee et al., 2019; Ramlogan-Steel et al., 2019). Missing in the current literature, however, are detailed explanations of the criteria used for selecting the first IRDs that have entered clinical trials. Also needed are assessments of what we have learned as a result of these major efforts by patients, doctors, administrators, regulators, and sponsors, and, when not successful, analysis of cause(s) of failure. In the present work, we review and discuss criteria for targeting an IRD for a clinical trial and make suggestions about which diseases (and stages of these diseases) warrant consideration as candidates for upcoming therapy.
2. Historical perspective: Mendelian genetic percentages to genes and mutations
By the early 1980’s, data on genetic percentages of IRDs from centers in different countries were already reported. The tallies represented first attempts to understand the genetic heterogeneity of these diseases and eventually led to linkage mapping of disease loci and finally gene and mutation identification. There were differences in percentages of the Mendelian inheritance types reported (Amman et al., 1965; Panteleeva, 1969; Jay, 1972; Bird, 1975; Fishman, 1978; Hu, 1982; Boughman and Fishman, 1983). For example, a common finding was that the largest subgroup of inheritance patterns, ranging from 36–92%, in most countries was autosomal recessive (AR) or presumed AR in the case of simplex and multiplex cases. In countries other than the UK, X-linked disease was ≤10%; UK results were 25%. There was a range of contribution of autosomal dominant (AD) inheritance from 5–39% depending on the population studied. In this premolecular era, questions arose as to the reason for such potentially interesting differences, such as different genetic pools (Fishman, 1978). At least some of the X-linked variation could be attributed to whether or not examiners evaluated the ophthalmoscopic appearance of mothers and other female relatives within a family with single or multiple affected men in one generation only. The heterozygous state of X-linked RP may be detectable by ophthalmoscopy as a patchy, and often more regional, pigmentary retinopathy or a tapetal-like reflex. A full blown disease, due to unfavorable lyonization, can also occur (Bird, 1975; Cideciyan and Jacobson, 1994).
Two North American centers that specialize in IRDs and originally collaborated to perform one of the first retinal gene therapy trials (Cideciyan et al., 2008) surveyed the genetic makeup of their patient populations. One center was the Center for Hereditary Retinal Degenerations (CHRD), first established at Bascom Palmer Eye Institute at the University of Miami and then relocated to Scheie Eye Institute at the University of Pennsylvania; CHRD is primarily an adult center specializing in IRDs. At the CHRD (henceforth named the Philadelphia center) between 1983 and 2019, 1656 patients diagnosed with an IRD were clinically and molecularly evaluated (male n=877; female n=779; mean age of first visit, 36.8 years; median age of first visit, 35.5 years; range of ages at first visit, 6 months – 89 years). The second center was the Department of Ophthalmology and Vision Sciences, Hospital for Sick Children, University of Toronto, Toronto, Canada, which is primarily a children’s center and a referral center for IRDs. At the Hospital for Sick Children (named the Toronto center) between 2005 and 2016, 1148 unrelated IRD patients underwent genetic testing (male, n=688; female n=460; mean age of first visit, 16 years; median age of first visit, 10 years; range of ages at first visit, 3 months – 77 years). From this 20 year difference in the mean age of first visit between the two centers, we could expect differences in the composition of IRDs; however, we would also expect similarities. There is variability in the age at which the disease is first detected for many IRDs, and this is not necessarily directly related to age of onset of symptoms or age of disease onset. For example, in IRD families with histories of affected members in various generations, proactive members may be examined and diagnosed pre-symptomatically. Acknowledging many caveats, the two sites generally serve different patient populations and we decided to compare the results.
In a survey of 300 IRD patients evaluated before 1990 at the Philadelphia center, AR/simplex/multiplex pattern of inheritance accounted for 63% (189 patients), and for the AD inheritance, it was 23% (70 patients); these figures are similar to many other series in that era. The X-linked mode of inheritance group was 14% (41 patients) which is between the higher number reported for the UK and the very low percentages of other countries (Figure 1A). The ocular fundi of female relatives of simplex males were routinely examined at the Philadelphia center, following the observations in UK studies (Bird, 1975). The decades following 1990 to the present day witnessed the discovery of >250 causative IRD genes (Duncan et al., 2018; Retnet, https://sph.uth.edu/retnet/). Once the causal genes began to be identified, collaboration occurred with many molecular scientists interested in gene discovery and, in more recent years, samples were sent to commercial laboratories to determine the molecular diagnosis of the IRDs. With a continuous effort to the present day, the number of unique genes that cause IRDs identified within our patient population has steadily increased. The genes unique to the Philadelphia center and to the Toronto center and those common to both centers are listed (Table 1).
Fig 1.

Mendelian patterns of inheritance, resolution of genotype over time and comparison of genotypes in IRD centers. (A-C) Data from the Philadelphia IRD center. (A) Pie chart showing the Mendelian patterns of inheritance in clinically-diagnosed patients seen prior to 1990. (B) Cumulative number of unique genes identified as causal over time. (C) The proportion of patients with unresolved and resolved genotypes in the current patient population and the comparison of Mendelian patterns of inheritance for patients with resolved and unresolved molecular diagnoses. (D) Comparison of the genes identified in the Philadelphia and Toronto cohorts that each cause disease in ≥2% of patients. Common top genes are given in the center. Blue, Philadelphia cohort; Orange, Toronto cohort. (E) Comparison of the top 5 disease-causing genes in the Philadelphia and Toronto cohorts along with percentages of the cohorts that are positive for each disease-causing gene. Blue (solid and hashed), Philadelphia; Orange (solid and hashed), Toronto. Solid bars represent genes that are in the top 5 for respective center; hashed bars indicate genes that are not in the top 5 for that center but still contribute to the overall cohort.
Table 1.
Disease-causing genes observed in the Philadelphia and Toronto IRD Centers
| Genes unique to Philadelphia Center (8 genes) | Genes common to both Centers (58 genes) | Genes unique to Toronto Center (32 genes) | |||
|---|---|---|---|---|---|
| FLVCR1 | ABCA4 | CRX | PRPF31 | AHI1 | GRK1 |
| IMPDH1 | ADGRV1 | DHDDS | PRPH2 | ALMS1 | IFT140 |
| INPP5E | AIPL1 | EYS | RDH12 | ATF6 | IMPG1 |
| LCA5 | BBS1 | FAM161A | RDH5 | BBS5 | IMPG2 |
| RLBP1 | BBS2 | GNAT2 | RHO | BBS7 | OFD1 |
| RP1L1 | BBS4 | GUCA1A | RP1 | BBS8 | PDE6A |
| SCA7 | BBS6 | GUCY2D | RP2 | BBS9 | PDE6C |
| SNRNP200 | BBS10 | KCNV2 | RPE65 | BBS12 | PNPLA6 |
| C1QTNF5 | KLHL7 | RPGR | BBS16 | POC1B | |
| C2orf71 | MAK | RPGRIP1 | C8ORF37 | PRPF3 | |
| CDH23 | MERTK | SPATA7 | CA4 | PRPF8 | |
| CDHR1 | MFRP | TIMP3 | CDH3 | RD3 | |
| CEP290 | MYO7A | TOPORS | CNGA1 | SLC4A11 | |
| CERKL | NMNAT1 | TULP1 | CNNM4 | TMEM216 | |
| CHM | NPHP5 | USH1C | CYP4V2 | TRPM1 | |
| CLRN1 | NR2E3 | USH2A | DRAM2 | USH1D | |
| CNGA3 | opsin gene arraya | VMD2 | |||
| CNGB1 | PCDH15 | WDR19 | |||
| CNGB3 | PDE6B | ||||
| CRB1 | PROM1 | ||||
Opsin gene array includes the OPN1LW and OPN1MW genes and the locus control region (LCR)
Of the 1656 patients who underwent genetic testing at the Philadelphia center, about half of the patients (48%) remain without a known genotype (Figure 1C); this is similar to previously published series investigating the molecular diagnoses of IRDs (Haer-Wigman et al., 2017; Carrigan et al., 2016; Ellingford et al., 2016; Bernardis et al., 2016; Huang et al., 2015; Glöckle et al., 2014). Of the patients with resolved genotypes, 59% (509 patients) are AR/simplex/multiplex; 24% (211) have an AD mode of inheritance; and 17% (146) have X-linked disease, indicating that the inheritance pattern composition of the genotypically-resolved patients remains generally the same through 2019 as compared to 1989. For patients whose molecular diagnosis remains unresolved through 2019, the overwhelming majority (80%, 635) has AR/simplex/multiplex disease; 16% (125) have AD disease; and 4% (30) have an X-linked mode of inheritance. The observations were comparable in the Toronto cohort. This is consistent with data from other groups that analyzed smaller IRD cohorts in which unresolved genotypes are composed primarily of autosomal recessive and sporadic cases (Audo et al., 2012; Bernardis et al, 2016; Huang et al., 2015). We learned that the likelihood of identifying a causal gene was increased if the inheritance pattern was known and if the phenotype was clearly defined.
Of the genes responsible for causing ≥ 2% of the IRDs, there were 11 that were common to both the Toronto and the Philadelphia cohorts (Figure 1D). The genes that comprise at least 2% of their respective cohorts account for 77% of the IRDs in the Philadelphia cohort and 63% of IRDs in the Toronto cohort. Of the top 5 genes in each cohort, only ABCA4, the most commonly-mutated gene in both cohorts, was shared. For Philadelphia, the remaining 4 top disease-causing genes identified were RHO, RPGR, PRPH2, and RPE65, whereas for Toronto, the remaining 4 were USH2A, VMD2, CHM and CNGA3 (Figure 1E). Both cohorts have patients with mutations in all of these genes, but there are differences between the populations in genetic composition of the patients. Many factors could be contributing to this, such as the ages of the patient populations, availability and methodology of genetic testing, and biases resulting from recruitment of certain populations stemming from research interests. The top disease-causing genes identified in Philadelphia and Toronto are similar to those in other surveys of inherited eye diseases or IRDs; ABCA4 and USH2A are among the top 3 genes in other reported cohorts (Haer-Wigman et al., 2017; Bernardis et al., 2016; Huang et al., 2015; Glöckle et al., 2014). In a recent estimation of the frequency of IRD genes, ABCA4, USH2A, RPGR, RHO and PRPH2 are listed as the most prevalent disease-causing genes in the United States (Stone et al., 2017).
A logical next step from clinical and then molecular diagnostics is to use this information to design and implement gene-based clinical trials in select IRDs. And this has been occurring.
3. From molecular genetic diagnoses to subretinal gene therapies
Milestones toward ocular gene therapy were present long before IRD causes were identified (Lee et al., 2019). Given the underlying molecular causes of IRDs, gene-based therapies were launched and there is now a list of clinical treatment trials of IRDs (Trapani and Auricchio, 2018; Ramlogan-Steel et al., 2019; Arbabi et al., 2019; Lee et al., 2019). What lessons have we learned from the first attempts to treat IRDs with a known genetic cause? To seek answers to this question, we provide a perspective on each of the 10 IRDs treated to date with subretinal gene therapy (Table 2) for which information is publicly available. Within these summaries, we will mention pre-clinical proof-of-concept studies, pre-clinical safety studies, clinical efficacy outcome measures and clinical natural history studies supporting each clinical trial. Our focus in this manuscript is on gene augmentation for specific IRDs; the interesting topic of gene therapies designed to target multiple IRDs is not covered (for example, Yu et al., 2017). There is also no attempt to discuss other promising therapies, such as stem-cell approaches, optogenetics or both (for example, Garita-Hernandez et al., 2019).
Table 2.
Subretinal gene therapy trials of inherited retinal degenerations
| Mendelian Genetic Type | Disease | Gene | Cell(s) with primary genetic defect | Estimated Gene Mutation Frequency in the USa | Proof of Concept and Safetyb | Dissociation of Structure-Functionc | Clinical trial number (Phase)d |
|---|---|---|---|---|---|---|---|
| Autosomal Recessive | LCA2 | RPE65 | RPE (and cone) | 1/576,667 | c, m, nhp | + for cones + for rods |
NCT00643747 (Phase I/II)e NCT00481546 (Phase I) NCT00516477 (Phase I) NCT00821340 (Phase I)e NCT00749957 (Phase I/II)e NCT01208389 (Phase I/II) NCT01496040 (Phase I/II)e NCT00999609 (Phase III) NCT02781480 (Phase I/II)e |
| RP38 | MERTK | RPE | 1/576,667 | m, nhp | NP | NCT01482195 (Phase I) | |
| USH1B | MYO7A | Rod, cone (and RPE) | 1/216,250 | Complex (m), nhp | − for rods NP for cones |
NCT01505062 (Phase I/II) | |
| STGD | ABCA4 | Rod,cone (and RPE) | 1/10,000 | Complex (m), r, nhp | NP | NCT01367444 (Phase I/II) | |
| ACHM2 | CNGA3 | Cone | 1/576,667 | o, m, nhp | +/− for cones |
NCT02610582 (Phase I/II) NCT02935517 (Phase I/II) NCT03758404 (Phase I/II) |
|
| ACHM3 | CNGB3 | Cone | 1/346,000 | c, m, nhp | +/− for cones |
NCT03001310 (Phase I/II) NCT02599922 (Phase I/II) NCT03278873 (Phase I/II) |
|
| RP40 | PDE6B | Rod | 1/247,143 | c, m | NP | NCT03328130 (Phase I/II) | |
| RP | RLBP1 | RPE and Muller cells | 1/1,730,000 | m, nhp | +/− for rods +/− for cones |
NCT03374657 (Phase I/II) | |
| X-linked | CHM | CHM | Rod, cone (and RPE) | 1/123,571 | m | NP |
NCT01461213 (Phase I/II) NCT02341807 (Phase I/II) NCT02077361 (Phase I/II)e NCT02553135 (Phase II)e NCT02671539 (Phase II) NCT02407678 (Phase II) NCT03507686 (Phase II) NCT03496012 (Phase III) |
| XLRP (RP3) | RPGR | Rod and cone | 1/36,042 | c, m | − for foveal cones NP for extrafoveal cones NP for rods |
NCT03116113 (Phase I/II) NCT03252847 (Phase I/II) NCT03316560 (Phase I/II) |
|
| Autosomal dominant | None to date |
Estimated frequency from Stone EM, et al. Ophthalmology. 2017;124(9):1314-1331.
Animal studies: c, canine; m, murine; nhp, non-human primate; o, ovine; r, rabbit
Gradation:
− No dissociation
+/− Qualitative dissociation: Evidence for existence of photoreceptor cell of interest lacking normal function implying improvement possible
+ Quantitative dissociation: Colocalized measure of retinal photoreceptor structure and light sensitivity defining vision improvement potential
NP Not evaluated or unknown
In order of trial start date
Completed trials
3.1. RPE65-LCA:
The form of Leber congenital amaurosis (LCA) due to mutations in the RPE65 gene (responsible for the retinoid isomerohydrolase of the retinal pigment epithelium, RPE; Hamel et al., 1993, 1994; Marlhens et al., 1997; Redmond et al., 1998) was the first IRD where a pre-clinical gene therapy success was translated to the clinic. Scientific discoveries leading to understanding of the molecular pathways in the mammalian visual cycle occurred well over 20 years preceding the concept of human gene therapy trials. Of course, the discovered association of mutations in the RPE65 gene with LCA was critical (Hamel et al., 1994; Marlhens et al., 1997) and now we know that LCA is caused by many different genes and mutations therein (Kumaran et al., 2017). Preclinical studies using the canine disease model of RPE65-LCA yielded convincing proof-of-concept results; there was dramatic improvement of visual function after subretinal gene augmentation with adeno-associated virus (AAV) vector. This stimulated the interest of the more clinically-minded scientists; and, as such, translation to the clinic became imaginable (Acland et al., 2001, 2005; Cideciyan, 2010).
How did we prepare to perform this first-in-human clinical trial? Safety of the gene-vector product was studied in both affected canines and in normal non-human primates, and the data published (Jacobson et al., 2006a, 2006b). Of interest, these toxicity studies were performed without the use of systemic steroids. Efficacy outcomes to quantify the severely impaired vision with nystagmus in LCA had to be devised anew because traditional outcomes used for other eye diseases (Csaky et al., 2008) were not as helpful. Hence, the FST (full-field stimulus test) was developed (Roman et al., 2005, 2007). The natural history of the human disease was not known at the time the trials began. The advent of cross-sectional in vivo imaging by optical coherence tomography (OCT) would have permitted a pre-trial natural history to be performed but there was an urgent desire by investigators to translate to the clinic and natural history studies require patients, time and resources. What information was there about the human disease to be treated? There were brief clinical descriptions in the reports of gene causation (for example, Lotery et al., 2000) or visual function and retinal structure in rare cases (van Hooser et al., 2000). We knew that there was serious visual dysfunction and that electroretinograms (ERGs) were abnormal for rod- and cone-mediated signals at all ages; there could even be non-detectable ERGs in the first years of life (Jacobson et al., 2009a). To deliver a local subretinal gene delivery, however, it seemed necessary to have a strategy about where this injection should be placed in the retina. Although it would be far more convenient to design a trial with a standardized bleb location in every patient (thinking ahead to multi-center trials in late phase trials), targeting a region of retina without photoreceptors would not be sensible. Photoreceptor topography using OCT was thus performed (Jacobson et al., 2008a). Normal ONL thickness topography represents a composite measure of cone and rod photoreceptor nuclei: there is peak ONL thickness at the fovea due to the high cone density and a further area of high density is at 10–16° superior to the fovea, the rod hot spot (Curcio et al.,1990; Jacobson et al., 2008a; Figure 2A). In RPE65-LCA patients there was loss of photoreceptors even in the youngest patients studied and there were different patterns to the preserved ONL (Jacobson et al., 2008a). Examples of six young patients are shown (Figure 2B–G). Two 7-year-olds had abnormally thinned ONL; one patient maintained a central island of ONL surrounded by barely detectable ONL and more superior than inferior pericentral structure (Figure 2B). The other 7-year old had greater ONL thickness, albeit still reduced, and an inferior retinal island of greater abnormality (Figure 2C). Two other patients, both age 12, had thicker ONL in the superior and temporal retina compared with inferiorly. They differed in the degree of foveal and parafoveal ONL thickness (Figure 2D,E). A 13- and a 14-year old patient retained central islands of ONL. Beyond this central patch, there was very limited preservation of ONL in the scanned area in the 13-year old; but there were preserved islands beyond the arcades in the 14-year old patient (Figure 2F,G). Such findings led to the suggestion that it would be judicious for ONL mapping to be used to guide location of the therapeutic bleb (Jacobson et al., 2008a). Targeting subretinal gene delivery based on ONL mapping occurred in only one of the three initial clinical trials (Cideciyan et al., 2008).
Fig 2.

Photoreceptor layer thickness topography in young patients with RPE65-LCA (A). Left. OCT cross-sectional images through the horizontal (upper scan) and vertical (lower scan) meridians of a normal subject (icons at upper right of scans are the location of the image on a schematic fundus; ONL, highlighted in blue). Right. Normal ONL thickness topography with color scale above. Hatched area, location of the optic nerve head. (B-G) ONL thickness topography in 6 young patients with RPE65-LCA, illustrating that there can be abnormalities at these ages and differences between patients despite similar ages. All eyes are depicted as right eyes. F, fovea. Modified from Jacobson et al., 2008a, copyright by the Association for Research in Vision and Ophthalmology.
What did we learn from the first-in-human retinal therapy of an IRD? Several independent clinical trials were performed concurrently and others followed (Table 2). Common to all the trials were reports of safety issues that mainly related to the subretinal surgical procedure (Bainbridge et al., 2008; Maguire et al., 2008; Hauswirth et al., 2008; Cideciyan et al., 2008; Jacobson et al., 2012). Even in the early phase safety trials, efficacy was present in the retinal area of treatment. Was this predictable or just lucky? Evidence for dissociation of structure and function in RPE65-LCA patients across a wide age spectrum was provided before the clinical trial began. In other words, biochemical recovery of the visual cycle pathway could theoretically occur and activate residual photoreceptors (Jacobson et al., 2005).
To test this hypothesis, we determined the co-localized relationship between retinal structure and function in two patients (Cideciyan et al., 2008). Outer photoreceptor nuclear layer (ONL) thickness was 29–43% of mean normal thickness (Fig.3A). Patients with retinitis pigmentosa with degenerative photoreceptor loss but without RPE65 mutations would be expected to show sensitivity losses ranging from 0.7–1.1 log units (Figure 3B). Pretreatment conservative estimates of the loss of rod function for RPE65-LCA patients were 6.4–7.2 log units. After treatment, one patient showed a 2.3 log unit increase in rod sensitivity, which approached, but did not reach, the predictions of the simple photoreceptor degeneration (Figure 3B). Another patient showed a posttreatment visual gain of 4.8 log units and the result became no different from that expected of simpler diseases with only a degenerative component contributing to the vision loss (Figure 3B).
Fig 3.

Early effects on rod-mediated vision of gene augmentation therapy in patients with RPE65-LCA. (A) OCT cross-sectional images (grayscale) from a normal subject and two patients (P2, P1) with RPE65-LCA. The sections are taken from ~5 mm superior retina in the normal subject and P2; section in P1 is from ~5 mm inferior retina. Schematics of ONL and RPE layers are overlaid (in yellow) on the OCT and illustrate the interpretation of reduced outer retinal structure in patients compared to normal. (B) Relationship of ONL thickness and rod-mediated sensitivity loss in normal subjects (triangles) and the two patients before therapy (unfilled large symbols) and after (up to 90 days; filled large symbols) gene therapy. The effect of the therapy led to rod sensitivity increase of ~5 log units (P2) and ~2 log units (P1). Ellipse, normal variability; broken lines, model of the expected relationship between structure and function based on quantum catch. Modified from Cideciyan et al., 2008, copyright by the National Academy of Sciences.
The dissociation of structure and function in this type of LCA thus stands as a feature worth measuring in future candidate diseases before clinical trials are initiated as it directs methods used to establish outcomes. There was no consensus, however, about the longevity of the efficacy (Cideciyan et al., 2013a; Jacobson et al., 2015; Bainbridge et al., 2015; Maguire et al., 2019). Evidence from two different groups indicated that there was continued retinal degeneration and eventual localized loss of visual function after therapy (Cideciyan et al., 2013a; Jacobson et al., 2015; Bainbridge et al., 2015). A third group which was involved in the commercialization of the gene therapy product (Russell et al., 2017) used non-localized (global) visual function and functional vision measures (Deverell et al., 2017) to support the conjecture that efficacy was durable (Maguire et al., 2019). Recent studies in the dog model have shown that longevity of the efficacy depends on the number of photoreceptors present at the time of treatment. Retinas with ONL that is >63% of normal at the time of treatment show no progression of degeneration, while areas with less photoreceptors at the time of treatment show progressive degeneration (Gardiner et al., 2019). Most patients at the time of treatment have ONL thicknesses that are less than 40% of normal (Jacobson et al., 2005; Jacobson et al., 2008a; Jacobson et al., 2012).
Despite the complexities revealed by these early trials, RPE65-LCA has become a business model for pharmaceutical and biotechnology companies who would launch programs into rare-orphan IRDs and seek regulatory approval of future gene therapy products.
3.2. MERTK-RP:
A second severe IRD treated with subretinal AAV gene therapy was caused by mutations in the MERTK (MER Proto-Oncogene, Tyrosine Kinase) gene (Gal et al., 2000; D’Cruz et al., 2000). Like RPE65-LCA, this was a primary RPE disease; it was a recessive human IRD modeled by the well-investigated RCS (Royal College of Surgeons; rdy) rat with an RPE phagocytosis defect (Dowling and Sidman, 1962; LaVail, 2001). A genetically-engineered mouse model was also studied (Duncan et al., 2003). Various modes of therapy have been applied to the rodent models (LaVail, 2001), and gene augmentation was entertained as a possible treatment (Ali, 2004; LaVail et al., 2016). Proof-of-concept was performed in rodents with different viral vectors and suggested rescue of degeneration and halting of the phagocytosis defect leading to preserved ERG responses rather than loss of recordable ERGs (Vollrath et al., 2001; Smith et al., 2003; Deng et al., 2012; LaVail et al., 2016). What was the preparation for this clinical trial? Safety studies were performed in SD rats (Conlon et al., 2013); non-human primate studies were published along with the clinical trial results but very limited information was provided (Ghazi et al., 2016). Efficacy outcomes used in the trial mirrored those in the RPE65-LCA trials (Ghazi et al., 2016). Unlike in patients with RPE65-LCA, however, there was no evidence in the literature (Charbel Issa et al., 2009; Audo et al., 2018) or in baseline examinations of the six clinical trial patients indicating dissociation of function and structure (i.e. potential for improvement of vision). Considering the proof-of-concept rodent data, human natural history data, and a trial timeline for halting progression would have been of value instead of the expectation of short-term improvement. The two-year long trial (Table 2) showed no change (in either eye) of parameters used other than visual acuity: one patient was reported to have improved acuity which then regressed (attributed to cataract formation); another patient improved at the end of the trial; and a third patient reported improvement but this was relatively minimal and later lost, also complicated by cataract. These data suggest that greater attention to the candidacy of patients and the human disease for gene augmentation, along with modified outcome measures, may be needed to perform future trials in MERTK-RP. Even in such a well-researched mechanism as in this rodent model, details of the disease expression in humans cannot be simply assumed.
3.3. MYO7A-USH1B:
Proof-of-concept studies for Usher syndrome genotypes in general have been hampered by the lack of retinal degenerative disease expression in murine models; USH1B due to mutations in MYO7A was no exception. There has also been debate about the cellular site of disease in USH1B with the shaker1 mouse model showing defective melanosome distribution in the RPE (Liu et al., 1998). Is it the photoreceptors and/or RPE (Williams and Lopes, 2011)? The MYO7A gene is too large for the AAV cargo capacity, so it was delivered with an equine infectious anemia virus (EIAV)-based lentiviral vector. A light-induced degenerative phenotype was reported in the shaker1 mouse (Peng et al., 2011) and subsequent studies of safety and efficacy with the lentivirus were performed in this USH1B model. Independent of the proof-of-concept complexities, movements were made towards a gene augmentation clinical trial (Table 2). Safety studies were performed in the shaker1 murine model and in non-human primates. The macaque experiments produced uveitis to different degrees in the vector-administered eyes and there were retinal abnormalities attributed to the subretinal injection procedure (Zallocchi et al, 2014). Efficacy outcomes have been explored in detailed studies of patients with USH1B (Jacobson et al., 2009b; Jacobson et al., 2011; Sumaroka et al., 2016). The transition zones from preserved and functional retina to severe retinal degeneration with visual loss were defined by OCT and by chromatic rod and cone perimetry. These regions were suggested to be sites for subretinal injection of vector-gene (Jacobson et al., 2009b). A disease sequence was also proposed from a study of cross-sectional data (Jacobson et al., 2011); different rates of progression within the retina were noted and recommendations were made about how to monitor patients in natural history studies and treatment trials (Sumaroka et al., 2016). Evidence was previously presented that USH1B showed no dissociation of structure and function (Jacobson et al., 2008b), so a natural history of disease was indicated to determine the timeline for a trial testing an intervention. The proof-of-concept work also supported that the goal of a trial would be to slow the rate of degeneration. A trial began many years ago using subretinal injection of (EIAV)-based lentiviral vector and, to our knowledge, there has been no formal report of the results of this clinical trial of gene augmentation therapy in USH1B to date.
3.4. ABCA4-STGD:
The other disease group that had gene augmentation therapy with a lentiviral-based gene transfer vector is ABCA4-associated IRD (Kong et al., 2008; Binley et al., 2013). The molecular mechanisms and human phenotypes of this group of diseases have been well explored and therapeutic concepts other than gene augmentation have been proposed and initiated (Allikmets et al., 1997; Sparrow et al., 1999; Fishman et al., 2003; Sears et al., 2017). Prevalence of the disease group is relatively high, compared to some of the other autosomal recessive IRDs in early clinical trials (Table 2). Lipofuscin accumulation in mouse models of ABCA4-STGD has been identified but the retinal degeneration that characterizes the human disease has not been a main finding (Weng et al., 1999; Zhang et al., 2015). Safety studies in rabbits and non-human primates using subretinal EIAV-gene in macular-equivalent regions showed some uveitis compared to controls and other retinal abnormalities (Binley et al., 2013). Efficacy outcomes have been explored in detailed studies attempting to quantify the progression and estimate natural history of ABCA4-STGD with various parameters (Cideciyan et al., 2004, 2007a, 2009, 2012, 2015; Huang et al., 2014; Ervin et al., 2019). All evidence to date suggests that the goal of a gene therapy approach would be to halt progression of the disease and prevent future retinal degeneration; improvement in vision would not be expected. As such, a very clear notion of the progression rate in candidate patients and the stages of disease to include in various cohorts would be required. We are unaware of a formal report of results of the clinical trial of EIAV-gene augmentation therapy in ABCA4-STGD.
3.5. CNGA3- and CNGB3-ACHM:
The autosomal recessive IRD group achromatopsia (ACHM) has severe and retina-wide abnormality of cone photoreceptor-based visual function from birth and is known to be caused by several different genes (CNGA3, CNGB3, GNAT2, PDE6C, PDE6H and ATF6; Hassall et al., 2017; Michalakis et al., 2017; Hirji et al., 2018). CNGA3 and CNGB3 genes encode the two subunits of the cone cyclic nucleotide-gated (CNG) channel and represent the most common genetic causes of ACHM (Wissinger et al., 2001; Kohl et al., 2005). There are disturbances of visual acuity and color vision, photosensitivity and nystagmus; a small percentage of patients with autosomal recessive cone dystrophy due to CNGB3 and GNAT2 mutations have also been reported (Michaelides et al., 2003; Thiadens et al., 2010). ACHM is clinically and molecularly distinct from the X-linked IRD known as blue cone monochromacy (BCM), which is caused by mutations in the array of long- and middle-wavelength cone opsin genes. There are small and large animal models of both CNGA3 and CNGB3 mutations and proof-of-concept research has been performed for subretinal gene therapy using AAV (Carvalho et al., 2011; Komáromy et al., 2010; Banin et al., 2015; Gootwine et al., 2017). In these models, function is restored once the normal subunit is expressed in mutant cells; of note, age-dependent effects of the therapy have been documented in the CNGB3 canine model even when the number of cones are not significantly reduced (Komáromy et al., 2010, 2013) and the age effect can be abrogated with a combinatorial therapy approach (see below). Safety studies relevant to CNGB3 were performed in mice, dogs and non-human primates, and at certain dose levels, there was detectable uveitis (Ye et al., 2016a, 2016b, 2017). For CNGA3, non-human primates were studied and minimal inflammation was present at one month but not at 3 months post injection; other abnormalities were ascribed to the surgical procedure (Tobias et al., 2019). Efficacy has been considered despite the appropriate focus on safety in early trials planned or ongoing (Yang et al., 2014; Zelinger et al., 2015; Zobor et al., 2015, 2017; Langlo et al., 2016; Kahle et al. 2018). Although not specifically addressed or quantified, there are OCT scans, cone photoreceptor imaging and acuities in the literature indicating dissociation of structure and function at the fovea in patients without atrophic central retinal lesions. Qualitatively, there is potential for improvement of vision. Natural history studies of either genotype, to our knowledge, have not been reported but there are indicators (from data in the literature) that foveal ONL losses can be present in some patients while others only show apparent cone outer segment abnormalities. For one of the CNGA3 trials, preliminary safety results were provided (Reichel et al., 2017) with a submacular injection (Kahle et al., 2018). There have been no formal reports to date of the results of the ongoing CNGA3 and CNGB3 clinical trials of subretinal gene augmentation therapy.
While it is premature to consider complementary approaches to ACHM gene therapies, it is important to remember that, at least in the CNGB3 canine model, recovery of function in treatment of aged mutant animals required that cone photoreceptors be transiently deconstructed with ciliary neurotrophic factor (CNTF), i.e. to lose whatever outer segments were present and regrow new ones in the presence of both subunits of the CNG channel (Komáromy et al., 2013).
3.6. PDE6B-RP:
PDE6B was one of the first genes identified to cause forms of AR RP in mice (Bowes et al., 1990; Pittler and Baehr, 1991; Chang et al., 2007), humans (McLaughlin et al., 1993) and dogs (Aguirre et al., 1978; Farber et al., 1992; Suber et al., 1993). Rarely PDE6B mutations are associated with AD RP (Gal et al., 1994; Gao et al., 1996; Manes et al., 2014). PDE6B encodes the beta-subunit of rod cGMP-phosphodiesterase which is a key enzyme specific for phototransduction activation in rod photoreceptors (Lamb and Pugh, 2004). Consistent with the gene function, patients show early onset of night vision loss (Jacobson et al., 2007; Tsang et al., 2008; Tatour et al., 2019). Animal models show an extremely fast degeneration of the rods. Proof-of-concept studies with subretinal AAV in a murine model were mostly unsuccessful (Bennett et al., 1996) unless a slower degenerating mouse line was used and treatment initiated relatively early (Pang et al., 2008). Similarly, successful subretinal gene therapy in dogs was achievable only when injected at a very early time during post-natal development of the retina (Petit et al., 2012; Pichard et al., 2016). We are not aware of published preclinical safety studies but a human clinical trial is ongoing (Table 2). For a primary rod disease treated with gene augmentation of a rod-specific gene, clinical efficacy outcomes would have to include evaluation of rod function and rod structure but there has been no evidence of either in patients published to date (Danciger et al., 1996; Jacobson et al., 2007; Tsang et al., 2008; Ali et al., 2011; Kim et al., 2012; Tatour et al., 2019).
3.7. RLBP1-RP:
A spectrum of AR IRDs are caused by mutations in RLBP1 (Maw et al., 1997; Burstedt et al., 1999; Morimura et al., 1999; Eichers et al., 2002). Prevalence of RLBP1-RP is very low in diverse populations (Table 2); however, prevalence can be substantially higher in some remote geographical regions (Burstedt et al., 1999; Eichers et al., 2002). In addition to a range of retinal degenerative features in common with many other IRDs, RLBP1-RP patients demonstrate extreme slowing of the rate of dark-adaptation for both rod and cone photoreceptors (Burstedt et al., 2003, 2013). It has long been known that RLBP1 encodes cellular retinal-binding protein (CRALBP) which is an 11-cis-retinal blinding protein expressed in the Muller and RPE cells (Bunt-Milam and Saari, 1983); it is a critical visual cycle component. But the rod and cone dark-adaptation kinetic defects in patients have recently become more interpretable by the finding of direct CRALBP involvement in the retinal Muller cell visual cycle supporting the cones (Xue et al., 2015) as well as the canonical RPE visual cycle supporting the rods (Saari et al., 2001). Preclinical proof-of-concept studies with subretinal AAV targeted both the RPE and Muller cells in mice and showed improvements in the rate of dark adaptation in rods and cones (Choi et al., 2015). Preclinical safety studies performed in mice, rats and non-human primates showed intraocular inflammation and dose-dependent retinal thinning (MacLachlan et al., 2018).
Clinical efficacy outcomes and natural history have not been published but are reported to include measures of retinal structure and visual function including full-field dark-adaptation kinetics (Green et al. 2017; Ni et al., 2017). There have been no published reports from the ongoing clinical trial aiming to improve the rod and cone visual cycle defects and stabilize the retinal degeneration (Table 2).
3.8. REP1-CHM:
Choroideremia (CHM) has been recognized clinically for more than a century due to a distinctive funduscopic appearance in hemi- and heterozygotes (reviewed in Mitsios et al., 2018). The causative gene and understanding of pathophysiology are more recent discoveries (Cremers et al., 1990; Seabra et al., 1993; Tolmachova et al., 2006). The finding of independent site(s) of disease action (photoreceptors and/or RPE) in both the human patients (Syed et al., 2001; Jacobson et al., 2006c; Syed at al., 2013; Morgan et al., 2014) and in a murine model of this X-linked IRD adds complexity to the proof-of-concept data and how it relates to the human disease (Tolmachova et al., 2006, 2010, 2013). Safety studies were performed in wild-type mice and in vitro (Vasireddy et al., 2013). Efficacy outcomes have concentrated on central retinal parameters, considering the surgical convenience of a macular subretinal injection and the common mid- and late-disease stage showing relatively preserved central retinal structure and function but little or no peripheral vision (Heon et al., 2016). Psychophysical results have included best-corrected visual acuities and perimetry under fundus visualization (‘microperimetry’); objective results are OCT and AF imaging (MacLaren et al., 2014; Aylward et al., 2018; Dimopoulos et al., 2018; Xue et al., 2018a, 2018b; Lam et al., 2019). The natural history of CHM, whether from cross-sectional data or performed prospectively, agree that CHM is a severe IRD but there is retained (near normal) central vision into late decades of life (Heon et al., 2016; Aleman et al., 2017; Di Iorio et al., 2019). This slow progression has been cited as advantageous because it provides a wide window for administering the treatment (Sengillo et al., 2016; Dimopoulos et al., 2018: Pennesi et al., 2019; Mitsios et al., 2018), yet it complicates outcome assessments in the absence of robust natural disease history studies. The proof-of-concept work with subretinal AAV-CHM in the CHM murine model showed increased function by dark-adapted ERGs but not light-adapted ERGs when treated eyes were compared with the contralateral sham-treated eyes; there was also reduced or no change in responses at lower doses of this vector but the higher dose led to increases in ERG amplitude. Further studies are likely needed to reconcile some of the results in the treated murine model and what we know of the human disease (Tolmachova et al., 2013). Despite the many studies of CHM hemizygotes, there has not been a test of the hypothesis of potential dissociation of structure and function, as has occurred with some IRDs (Jacobson et al., 2005, 2008b, 2014; Cideciyan and Jacobson, 2019).
REP1-CHM patients have been treated in several clinical trials of subretinal gene therapy. There have been reports of improvement in visual acuity and improvements of rod and cone function (MacLaren et al., 2014; Edwards et al., 2016; Fischer et al., 2018), no significant changes between treated and untreated eyes (Dimopoulos et al., 2018; Fischer et al., 2019), and retained visual acuity in later stage disease when uninjected contralateral eyes show progression (Xue et al., 2018b). CHM has been stated to be an attractive target for gene therapy because it represents an unmet need and will reduce societal burden through decreased health care costs (Pennesi et al., 2019). Certainly, the X-linked inheritance and the ease of ophthalmoscopic diagnosis make this rare disease detectable in the clinic; mutations in the CHM gene, encoding REP1 (rab escort protein1), have been identified and the gene can be delivered to RPE and photoreceptors with AAV2 (Pennesi et al., 2019). The months-long recovery of these photoreceptor-rich foveas after the macular detachment from a central subretinal injection has been used as supportive evidence that such surgical procedures are now of limited concern and should open the door to further subretinal macular injections even in patients with normal acuities (MacLaren et al., 2014; Xue et al., 2018a, 2018b). But foveal thinning and other outer retinal alterations remain a vulnerability of the surgical procedure (Pennesi et al., 2019; Aleman et al., 2019). Efficacy outcome results from the available trial reports seem mixed with uncertainty as to what the therapy is intended to do; from a notion of improving vision, the goals of the therapy seem to have changed to stopping disease progression.
3.9. RPGR-RP:
X-linked RP is a severe and relatively common IRD (Bird, 1975; Jay, 1982; Fishman et al., 1988). Pedigree analyses and examination of heterozygotes led to the identification of many XLRP families across the world that enabled linkage and then identification of the gene (Wright et al., 1987; Meindl et al., 1996; Vervoort et al., 2000; Breuer et al., 2002). Clinical gene augmentation therapy trials have concentrated on the common XLRP subtype: mutations in the RPGR gene and especially ORF15 mutations which account for >70% of RPGR disease. There has been availability of both small and large animal disease models for RPGR-XLRP; comparisons of the various mutant mice have been made (Huang et al., 2012) and AAV gene augmentation has been performed in mutant mice (for example, Wu et al., 2015; Fischer et al., 2017). Key proof–of-concept data came from the canine RPGR-ORF15 disease model using subretinal AAV gene therapy; the results indicated rescue of photoreceptors and reversal of secondary effects, specifically retinal remodeling secondary to photoreceptor loss (Beltran et al., 2012). The experiments showed that the arrest of disease progression was feasible not only in early stages but also in later stages of retinal degeneration in the model (Beltran et al., 2015). Safety studies were performed in wild-type and mutant mice (Deng et al., 2015; Fischer et al., 2017; Song et al., 2018) and issues about instability of the ORF15 were addressed. Large animal safety studies have not been published to date. Efficacy, as with many forms of RP considered to be severe, has been characterized as having sufficiently rapid progression that it will be able to be detected on clinical trial outcomes (Martinez-Fernandez De La Camara et al., 2018). Indeed, there are sufficient objective and subjective parameters that could be used as secondary outcomes.
A highly cited outcome for the central retina of RPGR-XLRP patients is the OCT (EZ line width) and the corresponding cone-mediated visual sensitivity change that brackets the EZ line change from measurable to unmeasurable; this rate of change has been shown to be within a two-year time frame that would be acceptable to clinical trialists (Birch et al., 2013, 2015). This remarkable observation to permit monitoring change is confined to the central retina and, if trials are being designed for subretinal injection, then this injection must be macular. Also to be considered is the common macular degeneration in RPGR-XLRP patients that would make submacular injections depending on EZ width measures either not possible or not desirable (Charng et al., 2016). In addition, quantitative results at the fovea have shown a lack evidence for cone vision improvement potential (Jacobson et al., 2014).
Extracentral retinal regions with rod- and cone-mediated vision measured with chromatic psychophysical methods, however, should not be neglected and untreated (Charng et al., 2016); in other words, the surgical convenience of macular subretinal injections should not drive the clinical trial. For clinical trials aiming to halt progression of disease, a realistic duration of 4.5 years for rod vision and 6.1 years for cone vision have been estimated, and ways to try to reduce these to shorter durations have been suggested (Cideciyan et al., 2018). Natural history studies have mainly been motivated by determining the least time it would take to determine change in a clinical trial of RPGR-XLRP patients (Birch et al., 2013, 2015; Charng et al., 2016; Cideciyan et al., 2018). These may appear as widely disparate results but actually what is being reported is that early trials would have to reckon with the facts of variation within this population and if there was a surgical commitment to macular subretinal injections as a trial and central retinal parameters (mainly cone-mediated) to monitor, then excluding more than half of the patients because of macular degenerative disease and unmeasurable EZ width would need to be part of the entry criteria. A larger population, and not necessarily older cohort, would include patients with the common maculopathies but a clinical trial strategy to monitor extracentral rod and cone vision and accept a lengthier timeline to decision about efficacy (Charng et al., 2016; Cideciyan et al., 2018). The three ongoing clinical trials of RPGR-XLRP seem to have chosen the route of submacular injection of AAV-gene delivery, but no results have been reported to date (Table 2).
3.10. Summary:
The first ten subretinal gene augmentation therapy trials were performed in otherwise incurable IRDs associated with a range of visual abnormalities; all IRDs represent unmet needs and this is not a justification for treating one before another (Pennesi et al., 2019). Prevalence varied between ~1:10,000 and ~1:2,000,000 (Table 2) and was also not the main reason for performing these specific trials. What then was the reason for beginning the era of retinal gene therapy with these specific diseases? Convincing reasons for considering a human clinical trial should have been evidence in pre-clinical proof-of-concept and safety studies that the product could have a positive effect with minimum risk to the patient. Some of the ten disorders had more of such evidence, and some had less. In addition, pre-clinical proof-of-concept results would have to be matched with the potential for short term improvement of vision and/or arrest of progression that is measurable within the study duration in the patients enrolled. Was there indication that the patients (before the trials) had potential for improvement in vision? Only RPE65-LCA had definite pre-clinical evidence of improvement (Acland et al., 2001, 2005) and definite evidence obtained prior to trial onset of potential for improvement (Jacobson et al., 2005). Maybe not surprisingly, substantial improvement in vision did occur in the RPE65 trials. The only other patients studied quantitatively for potential for visual improvement among the 10 conditions were those with MYO7A-USH1B (Jacobson et al., 2008b) and RPGR-XLRP (Jacobson et al., 2014). Dissociation of structure and function was not present in the extra-foveal rods of MYO7A-USH1B and foveal cones of RPGR-XLRP. In the remaining diseases, there were no pre-trial quantitative studies in affected patients to test for structure-function dissociation that would signal the potential for improvement in vision with therapeutic intervention. In CNGA3- and CNGB3-ACHM, human phenotype studies were suggestive of retained but dysfunctional cones; however, quantitative predictions of the extent of cone function improvement remain unknown.
Was there indication that arrest of progressive retinal degeneration can be measured within the study duration? In the case of RPE65-LCA, natural history was measurable and there was evidence that the natural history was not modified despite large vision improvements (Cideciyan et al., 2013a; Jacobson et al., 2015). Data are less direct or not available in other IRDs. RPGR-XLRP has natural history data which present different time courses of degeneration depending on whether the central retinal structure or extracentral retinal function is considered. The former suggests a 2-year study duration (Birch et al., 2013) while the latter indicates 4.5–6 years of monitoring (Cideciyan at al., 2018) needed to determine slowing or halting of progression. To date, there are no reported results from the RPGR-XLRP trials. CHM was not examined in a prospective natural history study before subretinal clinical trials began, but there have been cross-sectional studies (prospective and retrospective). Most of these CHM studies emphasized the persistence of central retinal structure and function until late decades in life. Natural history of central and peripheral ABCA4-STGD disease are described (Cideciyan et al. 2009; Cideciyan et al., 2015; Strauss et al., 2019) but there have been no gene therapy results described to date.
The decision to begin most of the trials likely also had to do with the feasibility of AAV vector delivery, and the fact that the RPE65-LCA trial safely used AAV vectors for subretinal injections. Diseases with genes that could be within the carrying capacity of AAV were targeted; dual AAV vectors platforms were evolving but not ready for the clinic (Dyka et al., 2014, 2019). Some trials used EIAV based lentiviral vectors for MYO7A and ABCA4 which were too large for AAV. There was a competitive rush to initiate trials after the RPE65-LCA trial and this led to an assumption that uveitis was not a serious adverse event; systemic steroids before and after the subretinal procedure have almost become an ‘industry standard’ to dampen any potential uveitic response that would cause adverse events. This conventional wisdom needs to be reconsidered and any uveitis investigated for cause.
4. Moving towards intravitreal injections
The next series of trials should target IRDs with a goal of improving remaining central vision with the least photoreceptor trauma and the most predictable outcome, all the while seeking to improve the quality of life of the patients. There is no doubt that the RPE65-LCA trials were inspirational - not only to patients (with or without RPE65-LCA) but also to sponsors that were not previously interested enough in orphan genetic retinal diseases to invest in clinical trials of treatment. Some sponsors have assumed that RPE65-LCA provides a ‘one-size fits all’ template waiting to be followed from concept to commercialization, and the underlying principle in designing further trials has seemed to be ‘don’t change the formula that worked before’. Publications about investor expectations during the trial to predictions of cost-effectiveness after the clinical trial have stoked further interest (for example, Schimmer and Breazzano, 2015; Johnson et al., 2019). Now that there are 9 further subretinal gene augmentation trials without evidence of multi-log-unit improvements, it has become clear that choosing a disabling IRD and following an RPE65-LCA-type formula may not be sufficient to achieve success.
Although there are many apologists for the disruptive effects of surgically-induced macular detachments, this common method to deliver vector-gene to photoreceptors and RPE begs to become a relic of this early period of human gene therapy trials. Presently, the subretinal surgical technique has almost become a subspecialty unto itself. There are innovations and improvements in the method to reduce adverse events and this is to be applauded. The influx of ideas is due to the fact that the current list of gene augmentation trials has extended to include multi-center protocols and thereby multiple talented retinal surgeons (Davis, 2018; Davis et al., 2019). It has been stated that there are no alternative methods to deliver vector-gene to the outer retina (Davis et al., 2019); without further research and a detailed understanding of the diseases being treated, that will remain true. Intravitreal delivery has been stated to be only useful for inner retinal or optic nerve diseases (Davis et al., 2019). However, it is recognized that intravitreal delivery could reduce the retinal trauma from detachment that can complicate the goals of the therapy (Khabou et al., 2018; Miller and Vanderberghe, 2018). Intravitreal injections have the potential to transfect large retinal areas but experimental evidence in non-human primates indicates that penetration of current intravitreal vectors to photoreceptors would be mainly or only in the fovea (Dalkara et al., 2013; Boye et al., 2016; Khabou et al., 2018). There are problems that need to be addressed and then solved. For example, an inflammatory uveitic response to the current vectors is worth the research effort to refine the details of this mode of delivery; “more potent and better tolerated vector systems” are needed (Miller and Vandenberghe, 2018). Of course, there are also non-gene-therapy and/or non-gene-specific approaches involving systemic or ocular drugs, but these will not be considered here.
We propose a group of 6 IRDs that should be candidates for intravitreal gene augmentation therapy, once it is improved (Table 3; Figure 4). In order to evaluate which IRDs may be most appropriate candidates for intravitreal gene therapy, we considered criteria based on current knowledge of the diseases, feasibility of clinics to acquire outcome measurements and the lessons learned from gene therapy trials to date. The goal would be to improve foveal vision in general, and foveal visual acuity in specific.
Table 3.
Diseases warranting consideration of intravitreal gene augmentation therapy
| Mendelian genetic type | Disease | Gene | Estimated gene mutation frequency in the USa | Proof of conceptb | Dissociation of structure-functionc | Comments |
|---|---|---|---|---|---|---|
| Autosomal recessive | ACHM2 | CNGA3 | 1/576,667 | o, m | +/− for cones | Qualitative evidence from literature of dissociation of structure-function |
| ACHM3 | CNGB3 | 1/346,000 | c, m | +/− for cones | Qualitative evidence from literature of dissociation of structure-function | |
| EORP/LCA | TULP1 | 1/1,730,000 | m model available | + for cones | Small central island only with remaining ONL but insensitive | |
| Senior-Loken/LCA | NPHP5 | 1/346,000 | c, m | + for cones | Central island only and concern over anesthesia because of renal abnormalities and multiple previous anesthesias for renal transplants Dissociation of structure and function provend |
|
| LCA6 | RPGRIP1 | 1/865,000 | m | +/− for cones | Qualitative evidence from literature of dissociation of structure and function | |
| X-linked | BCM | OPN1LW, OPN1MW | 1/576,667 | m | +/− for cones | Qualitative evidence from literature of dissociation of structure and function |
Estimated frequency from Stone EM, et al. Ophthalmology. 2017;124(9)-1314-1331.
Animal studies: c, canine; m, murine; o, ovine
Gradation:
+/− Qualitative dissociation: Evidence for existence of cell of interest lacking normal function implying improvement possible
+ Quantitative dissociation: Colocalized measure of retinal photoreceptor structure and light sensitivity defining vision improvement potential
Sumaroka A, et al. Invest Ophthalmol Vis Sci. 2019;60(7):2551–2562; Downs LM, et al. Hum Mol Genet. 2016;25(19)4211–4226.
Fig 4.

Foveal ONL thickness and central visual function, as measured with best-corrected visual acuity (VA, represented as decimal). (A) OCT scans of a normal subject (upper panel) and patients with IRDs that show normal or close to normal ONL thickness and minimally reduced or even normal VA. (B) Patients with IRD that show foveal ONL thickness comparable to ones in panel A, but severely impaired visual function. Representative OCT scans are along the vertical meridian from inferior retina (I) to superior (S) through the fovea. ONL is highlighted in blue. Foveal ONL thickness is depicted by white double arrow in normal scan. Dashed line in ONL thickness bar graph is lower limit of normal (n= 11; ages 26–62). Dashed line in VA bar graph is normal VA (20/20 or 1.0 in decimal representation).
Given the understanding that intravitreal vector-gene delivery would be limited to foveal cones (at least initially), how can we predict which IRDs may be better targets for treatment? Are there any measurements that could be performed in the eye clinic with widely available clinical methodology that would permit such prediction? The two such parameters we identified are foveal ONL thickness and best corrected visual acuity (BCVA). Spectral domain OCTs are commonly performed in the eye clinic and a single horizontal (and possibly also a vertical scan) through the fovea should allow for a measurement of foveal ONL. For example, OCT instruments, such as the Spectralis (Heidelberg Engineering, Heidelberg, Germany), the Cirrus 6000 (Carl Zeiss AG, Oberkochen, Germany), the Avanti RTVue XR or RTVue-100 (both manufactured by Optovue, Inc., Fremont, California, USA) include manufacturer software with built-in tools to enable measurement of the foveal ONL manually, and in some devices, automatically. Given this measure of photoreceptor nuclear structure at the fovea, the other relevant measure is routine – BCVA. Foveal ONL thickness and BCVA results (represented as decimal) in normal subjects and in patients with IRDs are shown (Figure 4). IRDs which can present with normal ONL thickness and minimally reduced or even normal visual acuity are on the left (Figure 4A). The IRDs vary in severity of disease outside the fovea but share the feature of preserved foveal ONL and central visual function (as measured by acuity) at the stages depicted. The 6 retinopathies with potential for improvement are shown on the right (Figure 4B); these are not uniform in genotype or phenotype but all share near-normal foveal ONL thickness (at least in early stages of disease) but with relatively reduced visual acuity. These diseases could be considered for intravitreal gene therapy.
Retina-wide cone diseases with decreased visual acuity, such as ACHM and BCM, would be worthy of attention. The current subretinal approach to ACHM (macular injections) risks these fragile foveas. Sufficient photoreceptor structure is evident in published (and quantified) images of foveal structure in CNGA3 and CNGB3 forms of ACHM (Zelinger et al. 2015; Langlo et al., 2016, 2017). The time course of the maculopathy needs to be considered and disease stage as well as genotype need to be taken into account (Kahn et al., 2007). Given the relatively long time course of disease (Langlo et al., 2017), patients may favor waiting for development of lower-risk means to deliver a gene product in a clinical trial. There is also evidence for retained photoreceptor lamination at the fovea in BCM which suggests that this condition is also a candidate for intravitreal gene therapy (Cideciyan et al., 2013b; Sumaroka et al., 2018).
In the spectrum of IRDs that could benefit from intravitreal delivery of vector is the severe form of early-onset RP caused by mutations in the TULP1 gene (Jacobson et al., 2014). This rapidly progressive IRD can lead to an advanced level of visual loss with a residual central island even in early decades of life. Surrounding the small central island there could be little or no detectable photoreceptor lamination by OCT but foveal ONL structure can be intact. Measurements of TULP1 structure and function and comparison with data from other ciliopathies causing IRDs indicated insensitive function for the amount of photoreceptor structure remaining at theTULP1 foveas (Jacobson et al., 2014). Given the simplified clinical ‘screening’ method using foveal ONL and visual acuity described above, other early-onset IRDs at late disease stages could be candidates for treatment. Whereas a small central island of impaired vision would seem to be an unlikely target for a subretinal surgical procedure, proof of dissociation of structure and function and the availability of a less invasive intravitreal vector delivery may be worth considering.
Also deserving consideration for gene augmentation therapy are those forms of LCA that have not been approached in clinical trials to date. Two forms of LCA share the phenotype that leads to severe loss of peripheral retinal structure and function early in life but with retained central photoreceptors that are more dysfunctional than expected from OCT measures of structure. This pattern is common to patients with mutations in the RPGRIP1 and NPHP5 (IQCB1) genes. Of interest, this is also the phenotype of CEP290-LCA (Cideciyan et al., 2007b; Cideciyan et al., 2011; Cideciyan and Jacobson, 2019) which has shown evidence of visual improvement in an antisense oligonucleotide clinical trial (Cideciyan et al., 2019); the large CEP290 gene would not be a candidate for an AAV vector approach. RPGRIP1 has been studied in depth and is a key protein in the photoreceptor connecting cilium (Li, 2014; Raghupathy et al., 2017). There are murine models, and proof-of-concept research with subretinal AAV has been performed (Pawlyk et al., 2010). The form of LCA caused by NPHP5 mutations (one of the causes of Senior-Loken syndrome, SLSN) is associated with renal disease (Otto et al., 2005; Stone et al., 2011; Estrada-Cuzcano et al., 2011; Cideciyan et al., 2011). Dissociation between structure and function in NPHP5-LCA means there is opportunity for visual improvement with a gene therapy treatment. There are large and small animal models (Downs et al., 2016; Oh et al., 2017; Hanke-Gogokhia et al., 2018); and a cone-only Nphp5−/−Nrl−/− murine model has been treated with subretinal gene augmentation using an AAV vector (Hanke-Gogokhia et al., 2018). Preliminary studies in the dog NPHP5 model in which there is a dissociation of structure and function have shown recovery of cone function and improvement of rod function in response to subretinal gene augmentation (Hardcastle et al., 2018).
Although studies relating structure and function are not, to our knowledge, specifically reported in later stages of CHM with reduced visual acuity, it would also seem worthy to consider an intravitreal alternative, once available, to subfoveal injections. Improved acuity has been reported in such patients treated with exceptional surgical care not to cause excessive foveal stretching or macular hole formation; if the same result were possible with an intravitreal injection, it may eventually extend the limits of CHM gene therapy beyond institutions with a high level of surgical prowess (Lam et al., 2019). Those stages with excellent foveal ONL and visual acuity would not seem to be worthy targets for therapy (Figure 4A).
5. Honoring the commitment to resolve the unresolved genotypes
Many recent reviews of IRDs strongly suggest there should be continued pursuit of molecular genetic diagnoses because of the obvious value in understanding cause of disease as well as helping to clarify pathobiology (Duncan et al., 2016 AAO Recommendations; Duncan et al., 2018). Based on what our cohort reveals with regard to the Mendelian type of the genetically unresolved patients (Figure 1), we asked why AR/simplex/multiplex diagnoses are relatively high versus other modes of inheritance. AR disease makes up the majority of IRD cases, and although many genes that cause IRDs have been identified, it is also likely that many more genes that cause IRDs remain to be discovered. In the list of 9 genes that are ‘new or updated’ as per the RetNet database (Retnet, https://sph.uth.edu/retnet/, last update, October 8, 2019), 8 of the genes were associated with a recessive disease. It has been observed that the number of mapped and identified genes have generally increased linearly with time, and since technology aiding in gene discovery has only improved, it follows that new genes are rarer or more difficult to detect (Daiger et al., 2013). These rarer genes may account for a considerable portion of the unresolved AR/simplex/multiplex cases. Although grouped with the AR cases, some of the simplex or multiplex cases may in reality be autosomal dominant (for example, de novo or reduced penetrance) or X-linked (Schwartz et al., 2003; Branham et al., 2012; Glöckle et al., 2014; Haer-Wigman et al., 2017). Mis-categorization of inheritance pattern likely accounts for only approximately 10% of the unsolved simplex cases if our cohort is comparable to those in other reports (Haer-Wigman et al., 2017; Glöckle et al., 2014). Incomplete family history information or small family size may contribute to mis-categorization of inheritance type. The more rare, genetically heterogeneous, or unclear the phenotype, the less-likely the IRD will be molecularly resolved (Haer-Wigman et al., 2017; Bernardis et al., 2016). Also, prevalent genetic testing strategies, such as analyzing a panel of genes, single-gene testing or targeted testing of regions known to harbor mutations, may not be comprehensive enough to yield a definitive molecular diagnosis and can be cost-prohibitive to patients. Mutations in non-coding regions, large structural aberrations or variants that are not detected using current computational filtering methods likely also play a role in contributing to the high proportion of molecularly unresolved AR/simplex/multiplex patients (Cremers et al., 2018; Farrar et al., 2017). Although still imperfect, there is movement towards providing patients with more comprehensive testing options, such as whole exome sequencing, and these are becoming increasingly affordable. Resolution of molecular diagnosis is valuable for patients with IRDs as the diagnosis could affect clinical management and eligibility for future trials of treatment.
6. Future directions and conclusions
From the extensive preparation for the first human retinal gene therapy trials, there have been varying levels of preparation for later novel treatment trials for these incurable IRDs. Initially, there seemed to be reluctance by pharmaceutical and biotechnology companies to be involved in this new direction, possibly due to the tragic death of a participant in an early gene therapy clinical trial (Wilson, 2010; Smith and Byers, 2002; Somia and Verma, 2000). With time and healing, there has been greater interest. Companies are labeling themselves as gene therapy companies, and regular conferences of gene and cell therapeutics companies are now occurring throughout the year. Whereas the initial drivers for such therapies were mainly academic and clinical achievement, the field is transforming into a competitive marketplace for economic achievement (Schimmer and Breazzano, 2015; Johnson et al., 2019; Zimmerman et al., 2019).
In this manuscript, we advocated for a forward movement of therapy but one based on the realities of the initial results of IRD human clinical trials. We have learned that there are many prerequisites for entering a group of patients into a gene augmentation trial. Entry criteria should include at least the following: 1) sufficient proof-of-concept research; 2) evidence of safety preferably based on testing affected models of the disease that are faithful to the human condition being modeled and also acquiring non-human primate data; 3) a thorough understanding of the human disease expression and its natural history and not just an extrapolation from the model data; 4) informative efficacy outcomes based on the human diseases being treated even if these are not the conventional outcomes of previous non-IRD disease trials; and, 5) determining before the trial onset if the goal of the trial is an improvement of vision or a slowing of disease progression. A first step toward predicting the extent of functional improvement from treatment was recently taken using supervised machine learning (Sumaroka et al., 2019). Specifically, cone vision improvement potential in the CEP290 and NPHP5 forms of LCA was determined. Data from patients with RP and residual cone-mediated macular function were used to associate visual sensitivities to local photoreceptor structure from colocalized OCTs (Figure 5). Such predictive strategies regarding potential vision based on recordable cross-sectional OCT images could lead to measurement-based decisions of the maximal potential efficacy in clinical trials and guide decisions as to which retinal diseases (or disease stages) to be treated and whether to proceed with planned increases of dose in cohorts of a trial (Sumaroka et al., 2019).
Fig 5.

Predicted and measured sensitivities for CEP290-LCA patients with severe loss of visual acuity (VA) and fixation. The upper row shows OCT scans along the horizontal meridian across the fovea; VAs are given above the scans (LP=light perception, BLP=bare light perception, NLP=no light perception). Plots under scans are measured FST sensitivities with red and blue full-field stimuli in the dark-adapted state (symbols) and artificial intelligence predictions of localized retinal function based on residual retinal structure (lines). Gray regions depict normal cone-mediated sensitivity in dark-adapted conditions. Modified from Sumaroka et al., 2019, copyright by the Association for Research in Vision and Ophthalmology.
If there is no measurable improvement possible based on a predictive strategy such as supervised machine learning, then the intent of an intervention would likely be to slow the disease course. The category of IRDs without improvement potential must be reckoned with rather than ignored. Trials should not begin on an untested hypothesis of potential visual improvement and then be in the unenviable circumstance of not knowing when to end. If a hoped-for miracle result does not occur within weeks or months, there must be a plan. Indeed, there should be a plan before starting the trial.
Gene augmentation trials will obviously move forward at a greater pace if the targeted diseases can be treated with less risk of adverse events. Subretinal injections were an appropriate way to begin; but moving to another delivery system, such as intravitreal injections, should be sought for many of the IRDs. The inflammatory consequences of both types of delivery systems must also be accepted and then studied and understood as opposed to being masked by anti-inflammatory agents. It is obvious that further research is required and this is the investment currently needed. The next group of IRDs entering trials should not be slowed by potentially solvable problems that are simply not as immediately rewarding or interesting as making a media announcement that a sponsor is first to the finish line with a new drug.
References
- Acland GM, Aguirre GD, Ray J, Zhang Q, Aleman TS, Cideciyan AV, et al. Gene therapy restores vision in a canine model of childhood blindness. Nat Genet. 2001;28(1):92–95. [DOI] [PubMed] [Google Scholar]
- Acland GM, Aguirre GD, Bennett J, Aleman TS, Cideciyan AV, Bennicelli J, et al. Long-term restoration of rod and cone vision by single dose rAAV-mediated gene transfer to the retina in a canine model of childhood blindness. Mol. Ther 2005;12(6):1072–1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aguirre G Retinal degenerations in the dog. I. Rod dysplasia. Exp Eye Res. 1978;26(3):233–253. [DOI] [PubMed] [Google Scholar]
- Aleman TS, Han G, Serrano LW, Fuerst NM, Charlson ES, Pearson DJ, et al. Natural history of the central structural abnormalities in choroideremia: A prospective cross-sectional study. Ophthalmology. 2017;124(3):359–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aleman TS, Huckfield RM, Serrano L, Vergillio G, Pearson DJ, Uyhazi KE, et al. AAV2-hCHM subretinal delivery to the macular in choroideremia: 2 year results of an ongoing phase I/II gene therapy trial. Invest Ophthalmol Vis Sci. 2019;60(9):5713. [Google Scholar]
- Ali RR. Prospects for gene therapy. Novartis Found Symp. 2004;255:165–172; discussion 173–178. [PubMed] [Google Scholar]
- Ali S, Riazuddin SA, Shahzadi A, Nasir IA, Khan SN, Husnain T, et al. Mutations in the β-subunit of rod phosphodiesterase identified in consanguineous Pakistani families with autosomal recessive retinitis pigmentosa. Mol Vis. 2011;17:1373–1380. [PMC free article] [PubMed] [Google Scholar]
- Allikmets R, Singh N, Sun H, Shroyer NF, Hutchinson A, Chidambaram A, et al. A photoreceptor cell-specific ATP-binding transporter gene (ABCR) is mutated in recessive Stargardt macular dystrophy. Nat Genet. 1997;15(3):236–246. [DOI] [PubMed] [Google Scholar]
- Ammann F, Klein D, Franceschetti A. Genetic and epidemiological investigations on pigmentary degeneration of the retina and allied disorders in Switzerland. J Neurol Sci. 1965;2(2):183–196. [DOI] [PubMed] [Google Scholar]
- Arbabi A, Liu A, Ameri H. Gene therapy for inherited retinal degeneration. J Ocul Pharmacol Ther. 2019;35(2):79–97. [DOI] [PubMed] [Google Scholar]
- Audo I, Bujakowska KM, Léveillard T, Mohand-Saïd S, Lancelot ME, Germain A, et al. Development and application of a next-generation-sequencing (NGS) approach to detect known and novel gene defects underlying retinal diseases. Orphanet J Rare Dis. 2012;7:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Audo I, Mohand-Said S, Boulanger-Scemama E, Zanlonghi X, Condroyer C, Démontant V, et al. MERTK mutation update in inherited retinal diseases. Hum Mutat. 2018;39(7):887–913. [DOI] [PubMed] [Google Scholar]
- Aylward JW, Xue K, Patrício MI, Jolly JK, Wood JC, Brett J, et al. Retinal degeneration in choroideremia follows an exponential decay function. Ophthalmology. 2018;125(7):1122–1124. [DOI] [PubMed] [Google Scholar]
- Bainbridge JW, Smith AJ, Barker SS, Robbie S, Henderson R, Balaggan K, et al. Effect of gene therapy on visual function in Leber's congenital amaurosis. N Engl J Med. 2008;358(21):2231–2239. [DOI] [PubMed] [Google Scholar]
- Bainbridge JW, Mehat MS, Sundaram V, Robbie SJ, Barker SE, Ripamonti C, et al. Long-term effect of gene therapy on Leber's congenital amaurosis. N Engl J Med. 2015;372(20):1887–1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Banin E, Gootwine E, Obolensky A, Ezra-Elia R, Ejzenberg A, Zelinger L, et al. Gene augmentation therapy restores retinal function and visual behavior in a sheep model of CNGA3 achromatopsia. Mol Ther. 2015;23(9):1423–1433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beltran WA, Cideciyan AV, Lewin AS, Iwabe S, Khanna H, Sumaroka A, et al. Gene therapy rescues photoreceptor blindness in dogs and paves the way for treating human X-linked retinitis pigmentosa. Proc Natl Acad Sci U S A. 2012;109(6):2132–2137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beltran WA, Cideciyan AV, Iwabe S, Swider M, Kosyk MS, McDaid K, et al. Successful arrest of photoreceptor and vision loss expands the therapeutic window of retinal gene therapy to later stages of disease. Proc Natl Acad Sci U S A. 2015;112(43):E5844–E5853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bennett J, Tanabe T, Sun D, Zeng Y, Kjeldbye H, Gouras P, Maguire AM. Photoreceptor cell rescue in retinal degeneration (rd) mice by in vivo gene therapy. Nat Med. 1996;2(6):649–654. [DOI] [PubMed] [Google Scholar]
- Bernardis I, Chiesi L, Tenedini E, Artuso L, Percesepe A, Artusi V, et al. Unravelling the complexity of inherited retinal dystrophies molecular testing: Added value of targeted next-generation sequencing. Biomed Res Int. 2016;2016:6341870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Binley K, Widdowson P, Loader J, Kelleher M, Iqball S, Ferrige G, et al. Transduction of photoreceptors with equine infectious anemia virus lentiviral vectors: safety and biodistribution of StarGen for Stargardt disease. Invest Ophthalmol Vis Sci. 2013;54(6):4061–4071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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(9):1143–1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Birch DG, Locke KG, Felius J, Klein M, Wheaton DK, Hoffman DR, et al. Rates of decline in regions of the visual field defined by frequency-domain optical coherence tomography in patients with RPGR-mediated X-linked retinitis pigmentosa. Ophthalmology. 2015;122(4):833–839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bird AC. X-linked retinitis pigmentosa. Br J Ophthalmol. 1975;59(4):177–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boughman JA, Fishman GA. A genetic analysis of retinitis pigmentosa. Br J Ophthalmol. 1983;67(7):449–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bowes C, Li T, Danciger M, Baxter LC, Applebury ML, Farber DB. Retinal degeneration in the rd mouse is caused by a defect in the beta subunit of rod cGMP-phosphodiesterase. Nature. 1990;347(6294):677–680. [DOI] [PubMed] [Google Scholar]
- Boye SE, Alexander JJ, Witherspoon CD, Boye SL, Peterson JJ, Clark ME, et al. Highly efficient delivery of adeno-associated viral vectors to the primate retina. Hum Gene Ther. 2016;27(8):580–597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bramall AN, Wright AF, Jacobson SG, McInnes RR. The genomic, biochemical, and cellular responses of the retina in inherited photoreceptor degenerations and prospects for the treatment of these disorders. Annu Rev Neurosci. 2010;33:441–472. [DOI] [PubMed] [Google Scholar]
- Branham K, Othman M, Brumm M, Karoukis AJ, Atmaca-Sonmez P, Yashar BM, et al. Mutations in RPGR and RP2 account for 15% of males with simplex retinal degenerative disease. Invest Ophthalmol Vis Sci. 2012;53(13):8232–8237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Breuer DK, Yashar BM, Filippova E, Hiriyanna S, Lyons RH, Mears AJ, et al. A comprehensive mutation analysis of RP2 and RPGR in a North American cohort of families with X-linked retinitis pigmentosa. Am J Hum Genet. 2002;70(6):1545–1554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bunt-Milam AH, Saari JC. Immunocytochemical localization of two retinoid-binding proteins in vertebrate retina. J Cell Biol. 1983;97(3):703–712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burstedt MS, Sandgren O, Holmgren G, Forsman-Semb K. Bothnia dystrophy caused by mutations in the cellular retinaldehyde-binding protein gene (RLBP1) on chromosome 15q26. Invest Ophthalmol Vis Sci. 1999;40(5):995–1000. [PubMed] [Google Scholar]
- Burstedt MS, Sandgren O, Golovleva I, Wachtmeister L. Retinal function in Bothnia dystrophy. An electrophysiological study. Vision Res. 2003;43(24):2559–2571. [DOI] [PubMed] [Google Scholar]
- Burstedt M, Jonsson F, Köhn L, Burstedt M, Kivitalo M, Golovleva I.Genotype-phenotype correlations in Bothnia dystrophy caused by RLBP1 gene sequence variations. Acta Ophthalmol. 2013;91(5):437–444. [DOI] [PubMed] [Google Scholar]
- Carrigan M, Duignan E, Malone CP, Stephenson K, Saad T, McDermott C, et al. Panel-based population next-generation sequencing for inherited retinal degenerations. Sci Rep. 2016;6:33248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carvalho LS, Xu J, Pearson RA, Smith AJ, Bainbridge JW, Morris LM, et al. Long-term and age-dependent restoration of visual function in a mouse model of CNGB3-associated achromatopsia following gene therapy. Hum Mol Genet. 2011;20(16):3161–3175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang B, Hawes NL, Pardue MT, German AM, Hurd RE, Davisson MT, et al. Two mouse retinal degenerations caused by missense mutations in the beta-subunit of rod cGMP phosphodiesterase gene. Vision Res. 2007;47(5):624–633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Charbel Issa P, Bolz HJ, Ebermann I, Domeier E, Holz FG, Scholl HP. Characterisation of severe rod-cone dystrophy in a consanguineous family with a splice site mutation in the MERTK gene. Br J Ophthalmol. 2009;93(7):920–925. [DOI] [PubMed] [Google Scholar]
- Charng J, Cideciyan AV, Jacobson SG, Sumaroka A, Schwartz SB, Swider M, et al. Variegated yet non-random rod and cone photoreceptor disease patterns in RPGR-ORF15-associated retinal degeneration. Hum Mol Genet. 2016;25(24):5444–5459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choi VW, Bigelow CE, McGee TL, Gujar AN, Li H, Hanks SM, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV, Jacobson SG. Image analysis of the tapetal-like reflex in carriers of X-linked retinitis pigmentosa. Invest Ophthalmol Vis Sci. 1994;35(11):3812–3824. [PubMed] [Google Scholar]
- Cideciyan AV, Aleman TS, Swider M, Schwartz SB, Steinberg JD, Brucker AJ, et al. Mutations in ABCA4 result in accumulation of lipofuscin before slowing of the retinoid cycle: a reappraisal of the human disease sequence. Hum Mol Genet. 2004;13(5):525–534. [DOI] [PubMed] [Google Scholar]
- Cideciyan AV, Swider M, Aleman TS, Roman MI, Sumaroka A, Schwartz SB, et al. Reduced-illuminance autofluorescence imaging in ABCA4-associated retinal degenerations. J Opt Soc Am A. 2007a;24(5):1457–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV, Aleman TS, Jacobson SG, Khanna H, Sumaroka A, Aguirre GK, et al. Centrosomal-ciliary gene GEP290/NPHP6 mutations result in blindness with unexpected sparing of photoreceptors and visual brain: implications for therapy of Leber congenital amaurosis. Hum Mutat. 2007b;28(11):1074–1083. [DOI] [PubMed] [Google Scholar]
- Cideciyan AV, Aleman TS, Boye SL, Schwartz SB, Kaushal S, Roman AJ, 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(39):15112–15117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV, Swider M, Aleman TS, Tsybovsky Y, Schwartz SB, Windsor EA, et al. ABCA4 disease progression and a proposed strategy for gene therapy. Hum Mol Genet. 2009;18(5):931–941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV. Leber congenital amaurosis due to RPE65 mutations and its treatment with gene therapy. Prog Retin Eye Res. 2010;29(5):398–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV, Rachel RA, Aleman TS, Swider M, Schwartz SB, Sumaroka A, et al. Cone photoreceptors are the main targets for gene therapy of NPHP5 (IQCB1) or NPHP6 (CEP290) blindness: generation of an all-cone Nphp6 hypomorph mouse that mimics the human retinal ciliopathy. Hum Mol Genet. 2011;20(7):1411–1423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV, Swider M, Aleman TS, Feuer WJ, Schwartz SB, Russell RC, et al. Macular function in macular degenerations: repeatability of microperimetry as a potential outcome measure for ABCA4-associated retinopathy trials. Invest Ophthalmol Vis Sci. 2012;53(2):841–852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV, Jacobson SG, Beltran WA, Sumaroka A, Swider M, Iwabe S, et al. Human retinal gene therapy for Leber congenital amaurosis shows advancing retinal degeneration despite enduring visual improvement. Proc Natl Acad Sci U S A. 2013a;110(6):E517–E525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV, Hufnagel RB, Carroll J, Sumaroka A, Luo X, Schwartz SB, et al. Human cone visual pigment deletions spare sufficient photoreceptors to warrant gene therapy. Hum Gene Ther. 2013b;24(12):993–1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV, Swider M, Schwartz SB, Stone EM, Jacobson SG. Predicting progression of ABCA4-associated retinal degenerations based on longitudinal measurements of the leading disease front. Invest Ophthalmol Vis Sci. 2015;56(10):5946–5955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV, Charng J, Roman AJ, Sheplock R, Garafalo AV, Heon E, et al. Progression in X-linked retinitis pigmentosa due to ORF15-RPGR mutations: Assessment of localized vision changes over 2 years. Invest Ophthalmol Vis Sci. 2018;59(11):4558–4566. [DOI] [PubMed] [Google Scholar]
- Cideciyan AV, Jacobson SG. Leber Congenital Amaurosis (LCA): Potential for improvement of vision. Invest Ophthalmol Vis Sci. 2019;60(5):1680–1695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cideciyan AV, Jacobson SG, Drack AV, Ho AC, Charng J, Garafalo AV, et al. Effect of an intravitreal antisense oligonucleotide on vision in Leber congenital amaurosis due to a photoreceptor cilium defect. Nat Med. 2019;25(2):225–228. [DOI] [PubMed] [Google Scholar]
- Conlon TJ, Deng WT, Erger K, Cossette T, Pang JJ, Ryals R, et al. Preclinical potency and safety studies of an AAV2-mediated gene therapy vector for the treatment of MERTK associated retinitis pigmentosa. Hum Gene Ther Clin Dev. 2013;24(1):23–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cremers FP, van de Pol DJ, van Kerkhoff LP, Wieringa B, Ropers HH. Cloning of a gene that is rearranged in patients with choroideraemia. Nature. 1990;347(6294):674–647. [DOI] [PubMed] [Google Scholar]
- Cremers FPM, Boon CJF, Bujakowska K, Zeitz C. Special issue introduction: inherited retinal disease: novel candidate genes, genotype-phenotype correlations, and inheritance models. Genes (Basel). 2018;9(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Csaky KG, Richman EA, Ferris FL 3rd. Report from the NEI/FDA ophthalmic clinical trial design and endpoints symposium. Invest Ophthalmol Vis Sci. 2008;49(2):479–489. [DOI] [PubMed] [Google Scholar]
- Curcio CA, Sloan KR, Kalina RE, Hendrickson AE. Human photoreceptor topography. J Comp Neurol. 1990;292:497–253. [DOI] [PubMed] [Google Scholar]
- D’Cruz PM, Yasumura D, Weir J, Matthes MT, Abderrahim H, LaVail MM et al. Mutation of the receptor tyrosine kinase gene Mertk in the retinal dystrophic RCS rat. Hum Mol Genet. 2000;9(4):645–651. [DOI] [PubMed] [Google Scholar]
- Daiger SP, Sullivan LS, Bowne SJ. Genes and mutations causing retinitis pigmentosa. Clin Genet. 2013;84:132–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dalkara D, Byrne LC, Klimczak RR, Visel M, Yin L, Merigan WH, et al. In vivo-directed evolution of a new adeno-associated virus for therapeutic outer retinal gene delivery from the vitreous. Sci Transl Med. 2013;5(189):189ra76. [DOI] [PubMed] [Google Scholar]
- Danciger M, Heilbron V, Gao YQ, Zhao DY, Jacobson SG, Farber DB. A homozygous PDE6B mutation in a family with autosomal recessive retinitis pigmentosa. Mol Vis. 1996;2:10. [PubMed] [Google Scholar]
- Davis JL. The blunt end: surgical challenges of gene therapy for inherited retinal diseases. Am J Ophthalmol 2018;196:25–29. [DOI] [PubMed] [Google Scholar]
- Davis JL, Gregori NZ, MacLaren RE, Lam BL. Surgical technique for subretinal gene therapy in humans with inherited retinal degeneration. Retina. 2019; Epub ahead of print. [DOI] [PubMed] [Google Scholar]
- Deng WT, Dinculescu A, Li Q, Boye SL, Li J, Gorbatyuk MS, et al. Tyrosine-mutant AAV8 delivery of human MERTK provides long-term retinal preservation in RCS rats. Invest Ophthalmol Vis Sci. 2012;53(4):1895–1904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deng WT, Dyka FM, Dinculescu A, Li J, Zhu P, Chiodo VA, et al. Stability and safety of an AAV vector for treating RPGR-ORF15 X-linked retinitis pigmentosa. Hum Gene Ther. 2015;26(9):593–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deverell L, Meyer D, Lau BT, Al Mahmud A, Sukunesan S, Bhowmik J, et al. Optimising technology to measure functional vision, mobility and service outcomes for people with low vision or blindness: protocol for a prospective cohort study in Australia and Malaysia. BMJ Open. 2017;7(12):e018140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Iorio V, Esposito G, De Falco F, Boccia R, Fioretti T, Colucci R, et al. CHM/REP1 transcript expression and loss of visual function in patients affected by choroideremia. Invest Ophthalmol Vis Sci. 2019;60(5):1547–1555. [DOI] [PubMed] [Google Scholar]
- Dimopoulos IS, Hoang SC, Radziwon A, Binczyk NM, Seabra MC, MacLaren RE, et al. Two-year results after AAV2-mediated gene therapy for choroideremia: the Alberta experience. Am J Ophthalmol. 2018;193:130–142. [DOI] [PubMed] [Google Scholar]
- Dowling JE, Sidman RL. Inherited retinal dystrophy in the rat. J Cell Biol. 1962;14:73–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Downs LM, Scott EM, Cideciyan AV, Iwabe S, Dufour V, Gardiner KL, et al. Overlap of abnormal photoreceptor development and progressive degeneration in Leber congenital amaurosis caused by NPHP5 mutation. Hum Mol Genet. 2016;25(19):4211–4226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duncan JL, LaVail MM, Yasumura D, Matthes MT, Yang H, Trautmann N, et al. An RCS-like retinal dystrophy phenotype in mer knockout mice. Invest Ophthalmol Vis Sci. 2003;44(2):826–838. [DOI] [PubMed] [Google Scholar]
- Duncan JL, Bernstein PS, Birch DG, Fishman GA, Heon E, Jacobson SG, et al. 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 August 2, 2019. [Google Scholar]
- Duncan JL, Pierce EA, Laster AM, Daiger SP, Birch DG, Ash JD, et al. Inherited retinal degenerations: current landscape and knowledge gaps. Transl Vis Sci Technol. 2018;7(4):6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dyka FM, Boye SL, Chiodo VA, Hauswirth WW, Boye SE. Dual adeno-associated virus vectors result in efficient in vitro and in vivo expression of an oversized gene, MYO7A. Hum Gene Ther Methods. 2014; 25(2):166–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dyka FM, Molday LL, Chiodo VA, Molday RS, Hauswirth WW. Dual ABCA4-AAV vector treatment reduces pathogenic retinal A2E accumulation in a mouse model of autosomal recessive Stargardt disease. Hum Gene Ther. 2019; 30(11):1361–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Edwards TL, Jolly JK, Groppe M, Barnard AR, Cottriall CL, Tolmachova T, et al. Visual acuity after retinal gene therapy for choroideremia. N Engl J Med. 2016;374:1996–1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eichers ER, Green JS, Stockton DW, Jackman CS, Whelan J, McNamara JA, et al. Newfoundland rod-cone dystrophy, an early-onset retinal dystrophy, is caused by splice-junction mutations in RLBP1. Am J Hum Genet. 2002;70(4):955–964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ellingford JM, Barton S, Bhaskar S, Williams SG, Sergouniotis PI, O'Sullivan J, et al. Whole genome sequencing increases molecular diagnostic yield compared with current diagnostic testing for inherited retinal disease. Ophthalmology. 2016;123(5):1143–1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ervin AM, Strauss RW, Ahmed MI, Birch D, Cheetham J, Ferris FL 3rd, et al. A workshop on measuring the progression of atrophy secondary to Stargardt disease in the ProgStar studies: Findings and lessons learned. Transl Vis Sci Technol. 2019;8(2):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Estrada-Cuzcano A, Koenekoop RK, Coppieters F, Kohl S, Lopez I, Collin RW, et al. IQCB1 mutations in patients with Leber congenital amaurosis. Invest Ophthalmol Vis Sci. 2011;52(2):834–839. [DOI] [PubMed] [Google Scholar]
- European Retinal Disease Consortium. http://www.EDRC.info. Accessed 8 August 2019.
- Farber DB, Danciger JS, Aguirre G. The beta subunit of cyclic GMP phosphodiesterase mRNA is deficient in canine rod-cone dysplasia 1. Neuron. 1992;9(2):349–356. [DOI] [PubMed] [Google Scholar]
- Farrar GJ, Carrigan M, Dockery A, Millington-Ward S, Palfi A, Chadderton N, et al. Toward an elucidation of the molecular genetics of inherited retinal degenerations. Hum Mol Genet. 2017;26(R1):R2–R11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fischer MD, McClements ME, Martinez-Fernandez de la Camara C, Bellingrath JS, Dauletbekov D, Ramsden SC, et al. Codon-optimized RPGR improves stability and efficacy of AAV8 gene therapy in two mouse models of X-linked retinitis pigmentosa. Mol Ther. 2017;25(8):1854–1865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fischer MD, Ochakovski GA, Beier B, Seitz IP, Vaheb Y, Kortuem C, et al. Changes in retinal sensitivity after gene therapy in choroideremia. Retina. 2018;00:1–9. [DOI] [PubMed] [Google Scholar]
- Fischer MD, Ochakovski GA, Beier B, Seitz IP, Vaheb Y, Kortuem C, et al. Efficacy and safety of retinal gene therapy using adeno-associated virus vector for patients with choroideremia: A randomized clinical trial. JAMA Ophthalmol. 2019; Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fishman GA. Retinitis pigmentosa. Genetic percentages. Arch Ophthalmol. 1978;96(5):822–826. [DOI] [PubMed] [Google Scholar]
- Fishman GA, Farber MD, Derlacki DJ. X-linked retinitis pigmentosa. Profile of clinical findings. Arch Ophthalmol. 1988;106(3):369–375. [DOI] [PubMed] [Google Scholar]
- Fishman GA, Stone EM, Eliason DA, Taylor CM, Lindeman M, Derlacki DJ. ABCA4 gene sequence variations in patients with autosomal recessive cone-rod dystrophy. Arch Ophthalmol. 2003;121(6):851–855. [DOI] [PubMed] [Google Scholar]
- Gal A, Orth U, Baehr W, Schwinger E, Rosenberg T. Heterozygous missense mutation in the rod cGMP phosphodiesterase beta-subunit gene in autosomal dominant stationary night blindness. Nat Genet. 1994;7(4):551. [DOI] [PubMed] [Google Scholar]
- Gal A, Li Y, Thompson DA, Weir J, Orth U, Jacobson SG, et al. Mutations in MERTK, the human orthologue of the RCS rat retinal dystrophy gene, cause retinitis pigmentosa. Nat Genet. 2000;26(3):270–271. [DOI] [PubMed] [Google Scholar]
- Gao YQ, Danciger M, Zhao DY, Blaney J, Piriev NI, Shih J, et al. Screening of the PDE6B gene in patients with autosomal dominant retinitis pigmentosa. Exp Eye Res. 1996;62(2):149–154. [DOI] [PubMed] [Google Scholar]
- Gardiner KL, Cideciyan AV, Swider M, Dufour VL, Sumaroka A, Komáromy AM, et al. Long-term structural outcomes of late-stage RPE65 gene therapy. Mol Ther. 2019; in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garita-Hernandez M, Lampič M, Chaffiol A, Guibbal L, Routet F, Santos-Ferreira T, et al. Restoration of visual function by transplantation of optogenetically engineered photoreceptors. Nat Commun. 2019;10(1):4524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ghazi NG, Abboud EB, Nowilaty SR, Alkuraya H, Alhommadi A, Cai H, et al. Treatment of retinitis pigmentosa due to MERTK mutations by ocular subretinal injection of adeno-associated virus gene vector: results of a phase I trial. Hum Genet. 2016;135(3):327–343. [DOI] [PubMed] [Google Scholar]
- Glöckle N, Kohl S, Mohr J, Scheurenbrand T, Sprecher A, Weisschuh N, et al. Panel-based next generation sequencing as a reliable and efficient technique to detect mutations in unselected patients with retinal dystrophies. Eur J Hum Genet. 2014;22(1):99–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gootwine E, Abu-Siam M, Obolensky A, Rosov A, Honig H, Nitzan T, et al. Gene augmentation therapy for a missense substitution in the cGMP-binding domain of ovine CNGA3 gene restores vision in day-blind sheep. Invest Ophthalmol Vis Sci. 2017;58(3):1577–1584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Green J, Burstedt M, Whelan JH, He Y, Su Z, Ni X, et al. Baseline characteristics of patients with RLBP1 retinitis pigmentosa enrolled in a prospective natural history study. Invest Ophthalmol Vis Sci. 2017;58:3254.28662230 [Google Scholar]
- Haer-Wigman L, van Zelst-Stams WA, Pfundt R, van den Born LI, Klaver CC, Verheij JB, et al. Diagnostic exome sequencing in 266 Dutch patients with visual impairment. Eur J Hum Genet. 2017;25(5):591–599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hamel CP, Tsilou E, Pfeffer BA, Hooks JJ, Detrick B, Redmond TM. Molecular cloning and expression of RPE65, a novel retinal pigment epithelium-specific microsomal protein that is post-transcriptionally regulated in vitro. J Biol Chem. 1993;268(21):15751–15757. [PubMed] [Google Scholar]
- Hamel CP, Jenkins NA, Gilbert DJ, Copeland NG, Redmond TM. The gene for the retinal pigment epithelium-specific protein RPE65 is localized to human 1p31 and mouse 3. Genomics. 1994;20(3):509–512. [DOI] [PubMed] [Google Scholar]
- Hanke-Gogokhia C, Chiodo VA, Hauswirth WW, Frederick JM, Baehr W. Rescue of cone function in cone-only Nphp5 knockout mouse model with Leber congenital amaurosis phenotype. Mol Vis. 2018;24:834–846. [PMC free article] [PubMed] [Google Scholar]
- Hardcastle AJ, Sieving PA, Sahel JA, Jacobson SG, Cideciyan AV, Flannery JG, et al. Translational retinal research and therapies. Transl Vis Sci Technol 2018;7(5):8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hassall MM, Barnard AR, MacLaren RE. Gene therapy for color blindness. Yale J Biol Med. 2017;90(4):543–551. [PMC free article] [PubMed] [Google Scholar]
- Hauswirth WW, Aleman TS, Kaushal S, Cideciyan AV, Schwartz SB, Wang L, et al. Treatment of Leber congenital amaurosis due to RPE65 mutations by ocular subretinal injection of adeno-associated virus gene vector: short-term results of a phase I trial. Hum Gene Ther. 2008;19(10):979–990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heon E, Alabduljalil T, McGuigan III DB, Cideciyan AV, Li S, Chen S, et al. Visual function and central retinal structure in choroideremia. Invest Ophthalmol Vis Sci. 2016;57(9):OCT377–OCT387. [DOI] [PubMed] [Google Scholar]
- Hirji N, Aboshiha J, Georgiou M, Bainbridge J, Michaelides M. Achromatopsia: clinical features, molecular genetics, animal models and therapeutic options. Ophthalmic Genet. 2018;39(2):149–157. [DOI] [PubMed] [Google Scholar]
- Hu DN. Genetic aspects of retinitis pigmentosa in China. Am J Med Genet. 1982;12(1):51–56. [DOI] [PubMed] [Google Scholar]
- Huang WC, Wright AF, Roman AJ, Cideciyan AV, Manson FD, Gewaily DY, et al. RPGR-associated retinal degeneration in human X-linked RP and a murine model. Invest Ophthalmol Vis Sci. 2012;53(9):5594–5608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang WC, Cideciyan AV, Roman AJ, Sumaroka A, Sheplock R, Schwartz SB, et al. Inner and outer retinal changes in retinal degenerations associated with ABCA4 mutations. Invest Ophthalmol Vis Sci. 2014;55(3):1810–1822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang XF, Huang F, Wu KC, Wu J, Chen J, Pang CP, et al. Genotype-phenotype correlation and mutation spectrum in a large cohort of patients with inherited retinal dystrophy revealed by next-generation sequencing. Genet Med. 2015;17(4):271–278. [DOI] [PubMed] [Google Scholar]
- Iannaccone A The genetics of hereditary retinopathies and optic neuropathies. Comp Ophthalmol Update. 2005;6(1):39–62. [Google Scholar]
- Jacobson SG, Aleman TS, Cideciyan AV, Sumaroka A, Schwartz SB, Windsor EA, et al. Identifying photoreceptors in blind eyes caused by RPE65 mutations: Prerequisite for human gene therapy success. Proc Natl Acad Sci U S A. 2005;102(17):6177–6182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobson SG, Acland GM, Aguirre GD, Aleman TS, Schwartz SB, Cideciyan AV, et al. Safety of recombinant adeno-associated virus type 2-RPE65 vector delivered by ocular subretinal injection. Mol Ther. 2006a;13(6):1074–1084. [DOI] [PubMed] [Google Scholar]
- Jacobson SG, Boye SL, Aleman TS, Conlon TJ, Zeiss CJ, Roman AJ, et al. Safety in nonhuman primates of ocular AAV2-RPE65, a candidate treatment for blindness in Leber congenital amaurosis. Hum Gene Ther. 2006b;17(8):845–858. [DOI] [PubMed] [Google Scholar]
- Jacobson SG, Cideciyan AV, Sumaroka A, Aleman TS, Schwartz SB, Windsor EA, et al. Remodeling of the human retina in choroideremia: rab escort protein 1 (REP-1) mutations. Invest Ophthal Vis Sci. 2006c;47(9):4113–4120. [DOI] [PubMed] [Google Scholar]
- Jacobson SG, Sumaroka A, Aleman TS, Cideciyan AV, Danciger M, Farber DB. Evidence for retinal remodelling in retinitis pigmentosa caused by PDE6B mutation. Br J Ophthalmol. 2007;91(5):699–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobson SG, Cideciyan AV, Aleman TS, Sumaroka A, Windsor EA, Schwartz SB, et al. Photoreceptor layer topography in children with Leber congenital amaurosis caused by RPE65 mutations. Invest Ophthalmol Vis Sci. 2008a;49(10):4573–4577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobson SG, Cideciyan AV, Aleman TS, Sumaroka A, Roman AJ, Gardner LM, et al. Usher syndromes due to MYO7A, PCDH15, USH2A or GPR98 mutations share retinal disease mechanism. Hum Mol Genet. 2008b;17(15):2405–2415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobson SG, Aleman TS, Cideciyan AV, Roman AJ, Sumaroka A, Windsor EA, et al. Defining the residual vision in Leber congenital amaurosis caused by RPE65 mutations. Invest Ophthalmol Vis Sci. 2009a;50(5):2368–2375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobson SG, Aleman TS, Sumaroka A, Cideciyan AV, Roman AJ, Windsor EA, et al. Disease boundaries in the retina of patients with Usher syndrome caused by MYO7A gene mutations. Invest Ophthalmol Vis Sci. 2009b;50(4):1886–1894. [DOI] [PubMed] [Google Scholar]
- Jacobson SG, Cideciyan AV, Gibbs D, Sumaroka A, Roman AJ, Aleman TS, et al. Retinal disease course in Usher syndrome 1B due to MYO7A mutations. Invest Ophthalmol Vis Sci. 2011;52(11):7924–7936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobson SG, Cideciyan AV, Ratnakaram R, Heon E, Schwartz SB, Roman AJ, 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(1):9–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobson SG, Cideciyan AV, Huang WC, Sumaroka A, Roman AJ, Schwartz SB, et al. TULP1 mutations causing early-onset retinal degeneration: preserved but insensitive macular cones. Invest Ophthalmol Vis Sci. 2014;55(8):5354–5364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacobson SG, Cideciyan AV, Roman AJ, Sumaroka A, Schwartz SB, Heon E, et al. Improvement and decline in vision with gene therapy in childhood blindness. N Engl J Med. 2015;372(20):1920–1926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jay B Hereditary aspects of pigmentary retinopathy. Trans Ophthalmol Soc U K. 1972;92:173–178. [PubMed] [Google Scholar]
- Jay M On the heredity of retinitis pigmentosa. Br J Ophthalmol. 1982;66(7):405–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson S, Buessing M, O'Connell T, Pitluck S, Ciulla TA. Cost-effectiveness of voretigene neparvovec-rzyl vs standard care for RPE65-mediated inherited retinal disease. JAMA Ophthalmol. 2019; Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kahle NA, Peters T, Zobor D, Kuehlewein L, Kohl S, Zhour A, et al. Development of methodology and study protocol: Safety and efficacy of a single subretinal injection of rAAV.hCNGA3 in patients withCNGA3-linked achromatopsia investigated in an exploratory dose-escalation trial. Hum Gene Ther Clin Dev. 2018;29(3):121–131. [DOI] [PubMed] [Google Scholar]
- Kahn NW, Wissinger B, Kohl S, Sieving PA. CNGB3 achromatopsia with progressive loss of residual cone function and impaired rod-mediated function. Invest Ophthalmol Vis Sci. 2007;48(8):3864–3871. [DOI] [PubMed] [Google Scholar]
- Khabou H, Garita-Hernandez M, Chaffiol A, Reichman S, Jaillard C, Brazhnikova E, et al. Noninvasive gene delivery to foveal cones for vision restoration. JCI Insight. 2018;3(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim C, Kim KJ, Bok J, Lee EJ, Kim DJ, Oh JH, et al. Microarray-based mutation detection and phenotypic characterization in Korean patients with retinitis pigmentosa. Mol Vis. 2012;18:2398–2410. [PMC free article] [PubMed] [Google Scholar]
- Kohl S, Varsanyi B, Antunes GA, Baumann B, Hoyng CB, Jägle H, et al. CNGB3 mutations account for 50% of all cases with autosomal recessive achromatopsia. Eur J Hum Genet. 2005;13(3):302–308. [DOI] [PubMed] [Google Scholar]
- Komáromy AM, Alexander JJ, Rowlan JS, Garcia MM, Chiodo VA, Kaya A, et al. Gene therapy rescues cone function in congenital achromatopsia. Hum Mol Genet. 2010;19(13):2581–2593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Komáromy AM, Rowlan JS, Corr AT, Reinstein SL, Boye SL, Cooper AE, et al. Transient photoreceptor deconstruction by CNTF enhances rAAV-mediated cone functional rescue in late stage CNGB3-achromatopsia. Molecular Therapy. 2013; 21:1131–1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong J, Kim SR, Binley K, Pata I, Doi K, Mannik J, et al. Correction of the disease phenotype in the mouse model of Stargardt disease by lentiviral gene therapy. Gene Ther. 2008;15(19):1311–1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumaran N, Moore AT, Weleber RG, Michaelides M. Leber congenital amaurosis/early-onset severe retinal dystrophy: clinical features, molecular genetics and therapeutic interventions. Br J Ophthalmol. 2017;101(9):1147–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Langlo CS, Patterson EJ, Higgins BP, Summerfelt P, Razeen MM, Erker LR, et al. Residual foveal cone structure in CNGB3-associated achromatopsia. Invest Ophthalmol Vis Sci. 2016;57(10):3984–3995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Langlo CS, Erker LR, Parker M, Patterson EJ, Higgins BP, Summerfelt P, et al. Repeatability and longitudinal assessment of foveal cone structure in CNGB3-associated achromatopsia. Retina. 2017;37(10):1956–1966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lam BL, Davis JL, Gregori NZ, MacLaren RE, Girach A, Verriotto JD, et al. Choroideremia gene therapy phase 2 clinical trial: 24-month results. Am J Ophthalmol. 2019;197:65–73. [DOI] [PubMed] [Google Scholar]
- Lamb TD, Pugh EN Jr. Dark adaptation and the retinoid cycle of vision. Prog Retin Eye Res. 2004;23(3):307–380. [DOI] [PubMed] [Google Scholar]
- LaVail MM. Legacy of the RCS rat: impact of a seminal study on retinal cell biology and retinal degenerative diseases. Prog Brain Res. 2001;131:617–627. [DOI] [PubMed] [Google Scholar]
- LaVail MM, Yasumura D, Matthes MT, Yang H, Hauswirth WW, Deng WT, et al. Gene therapy for MERTK-associated retinal degenerations. Adv Exp Med Biol. 2016;854:487–493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee JH, Wang JH, Chen J, Li F, Edwards TL, Hewitt AW, et al. Gene therapy for visual loss: Opportunities and concerns. Prog Retin Eye Res. 2019;68:31–53. [DOI] [PubMed] [Google Scholar]
- Li T Leber congenital amaurosis caused by mutations in RPGRIP1. Cold Spring Harb Perspect Med. 2014;5(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu X, Ondek B, Williams DS. Mutant myosin VIIa causes defective melanosome distribution in the RPE of shaker-1 mice. Nat Genet. 1998;19(2):117–118. [DOI] [PubMed] [Google Scholar]
- Lotery AJ, Namperumalsamy P, Jacobson SG, Weleber RG, Fishman GA, Musarella MA, et al. Mutation analysis of 3 genes in patients with Leber congenital amaurosis. Arch Ophthalmol. 2000;118(4):538–543. [DOI] [PubMed] [Google Scholar]
- MacLaren RE, Groppe M, Barnard AR, Cottriall CL, Tolmachova T, Seymour L, et al. Retinal gene therapy in patients with choroideremia: initial findings from a phase 1/2 clinical trial. Lancet. 2014;383(9923):1129–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MacLachlan TK, Milton MN, Turner O, Tukov F, Choi VW, Penraat J, et al. Nonclinical safety evaluation of scAAV8-RLBP1 for treatment of RLBP1 retinitis pigmentosa. Mol Ther Methods Clin Dev. 2018;8:105–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maguire AM, Simonelli F, Pierce EA, Pugh EN Jr, Mingozzi F, Bennicelli J, et al. Safety and efficacy of gene transfer for Leber's congenital amaurosis. N Engl J Med. 2008;358(21):2240–2248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maguire AM, Russell S, Wellman JA, Chung DC, Yu ZF, Tillman A, et al. Efficacy, safety, and durability of voretigene neparvovec-rzyl in RPE65 mutation-associated inherited retinal dystrophy: results of phase 1 and 3 trials. Ophthalmology. 2019;126(9):1273–1285. [DOI] [PubMed] [Google Scholar]
- Manes G, Cheguru P, Majumder A, Bocquet B, Sénéchal A, Artemyev NO, et al. A truncated form of rod photoreceptor PDE6 β-subunit causes autosomal dominant congenital stationary night blindness by interfering with the inhibitory activity of the γ-subunit. PLoS One. 2014;9(4):e95768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marlhens F, Bareil C, Griffoin JM, Zrenner E, Amalric P, Eliaou C, et al. Mutations in RPE65 cause Leber's congenital amaurosis. Nat Genet. 1997;17(2):139–141. [DOI] [PubMed] [Google Scholar]
- Martinez-Fernandez De La Camara C, Nanda A, Salvetti AP, Fischer MD, MacLaren RE. Gene therapy for the treatment of X-linked retinitis pigmentosa. Expert Opin Orphan Drugs. 2018;6(3):167–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maw MA, Kennedy B, Knight A, Bridges R, Roth KE, Mani EJ, et al. Mutation of the gene encoding cellular retinaldehyde-binding protein in autosomal recessive retinitis pigmentosa. Nat Genet. 1997;17(2):198–200. [DOI] [PubMed] [Google Scholar]
- McLaughlin ME, Sandberg MA, Berson EL, Dryja TP. Recessive mutations in the gene encoding the beta-subunit of rod phosphodiesterase in patients with retinitis pigmentosa. Nat Genet. 1993;4(2):130–134. [DOI] [PubMed] [Google Scholar]
- Meindl A, Dry K, Herrmann K, Manson F, Ciccodicola A, Edgar A, et al. A gene (RPGR) with homology to the RCC1 guanine nucleotide exchange factor is mutated in X-linked retinitis pigmentosa (RP3). Nat Genet. 1996;13(1):35–42. [DOI] [PubMed] [Google Scholar]
- Michaelides M, Aligianis IA, Holder GE, Simunovic M, Mollon JD, Maher ER, et al. Cone dystrophy phenotype associated with a frameshift mutation (M280fsX291) in the alpha-subunit of cone specific transducing (GNAT2). Br J Ophthalmol. 2003;87(11)1317–1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Michalakis S, Schön C, Becirovic E, Biel M. Gene therapy for achromatopsia. J Gene Med. 2017;19(3). [DOI] [PubMed] [Google Scholar]
- Miller JW, Vandenberghe LH. Breaking and sealing barriers in retinal gene therapy. Mol Ther. 2018;26(9):2081–2082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitsios A, Dubis AM, Moosajee M. Choroideremia: from genetic and clinical phenotyping to gene therapy and future treatments. Ther Adv Ophthalmol. 2018;10:2515841418817490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morgan JIW, Han G, Klinman E, Maguire WM, Chung DC, Maguire AM, et al. High-resolution adaptive optics retinal imaging of cellular structure in choroideremia. Invest Ophthalmol Vis Sci. 2014;55(10):6381–6397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morimura H, Berson EL, Dryja TP. Recessive mutations in the RLBP1 gene encoding cellular retinaldehyde-binding protein in a form of retinitis punctate albescens. Invest Ophthalmol Vis Sci. 1999;40(5):1000–1004. [PubMed] [Google Scholar]
- Ni X, Walk M, Burstedt M, Green J, Whelan J, Su Z, et al. Full-field stimulus threshold dark-adaptation kinetics over two-year follow-up in patients with RLBP1 retinitis pigmentosa enrolled in a prospective natural history study. Invest Ophthalmol Vis Sci. 2017;58(8):3249.28660278 [Google Scholar]
- Oh A, Pearce JW, Gandolfi B, Creighton EK, Suedmeyer WK, Selig M, et al. Early-onset progressive retinal atrophy associated with an IQCB1 variant in African black-footed cats (Felis nigripes). Sci Rep. 2017;7:43918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Otto EA, Loeys B, Khanna H, Hellemans J, Sudbrak R, Fan S, et al. Nephrocystin-5, a ciliary IQ domain protein, is mutated in Senior-Loken syndrome and interacts with RPGR and calmodulin. Nat Genet. 2005;37(3):282–288. [DOI] [PubMed] [Google Scholar]
- Pang JJ, Boye SL, Kumar A, Dinculescu A, Deng W, Li J, et al. AAV-mediated gene therapy for retinal degeneration in the rd10 mouse containing a recessive PDEbeta mutation. Invest Ophthalmol Vis Sci. 2008. October;49(10):4278–4283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pawlyk BS, Bulgakov OV, Liu X, Xu X, Adamian M, Sun 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(8):993–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Panteleeva OA. [On the problem of hereditary tapetoretinal degeneration]. Vestn Oftalmol. 1969; 53–56. [PubMed] [Google Scholar]
- Peng YW, Zallocchi M, Wang WM, Delimont D, Cosgrove D. Moderate light-induced degeneration of rod photoreceptors with delayed transducin translocation in shaker1 mice. Invest Ophthalmol Vis Sci. 2011;52(9):6421–6427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pennesi ME, Birch DG, Duncan JL, Bennett J, Girach A. Choroideremia: retinal degeneration with an unmet need. Retina. 2019; Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petit L, Lhériteau E, Weber M, Le Meur G, Deschamps JY, Provost N, et al. Restoration of vision in the pde6β-deficient dog, a large animal model of rod-cone dystrophy. Mol Ther. 2012;20(11):2019–2030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pichard V, Provost N, Mendes-Madeira A, Libeau L, Hulin P, Tshilenge KT, et al. AAV-mediated gene therapy halts retinal degeneration in PDE6β-deficient dogs. Mol Ther. 2016;24(5):867–876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pittler SJ, Baehr W. Identification of a nonsense mutation in the rod photoreceptor cGMP phosphodiesterase beta-subunit gene of the rd mouse. Proc Natl Acad Sci U S A. 1991;88(19):8322–8326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raghupathy RK, Zhang X, Liu F, Alhasani RH, Biswas L, Akhtar S, et al. Rpgrip1 is required for rod outer segment development and ciliary protein trafficking in zebrafish. Sci Rep. 2017;7(1):16881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramlogan-Steel CA, Murali A, Andrzejewski S, Dhungel B, Steel JC, Layton CJ. Gene therapy and the adeno-associated virus in the treatment of genetic and acquired ophthalmic diseases in humans: Trials, future directions and safety considerations. Clin Exp Ophthalmol. 2019;47(4):521–536. [DOI] [PubMed] [Google Scholar]
- Redmond TM, Yu S, Lee E, Bok D, Hamasaki D, Chen N, et al. Rpe65 is necessary for production of 11-cis-vitamin A in the retinal visual cycle. Nat Genet. 1998;20(4):344–351. [DOI] [PubMed] [Google Scholar]
- Reichel FF, Dauletbekov DL, Klein R, Peters T, Ochakovski GA, Seitz IP, et al. AAV8 can induce innate and adaptive immune response in the primate eye. Mol Ther. 2017;25(12):2648–2660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Retnet, the Retinal Information Network. http://www.sph.uth.tmc.edu/RetNet/. Accessed 8 October 2019.
- Roman AJ, Schwartz SB, Aleman TS, Cideciyan AV, Chico JD, Windsor EA, et al. Quantifying rod photoreceptor-mediated vision in retinal degenerations: dark-adapted thresholds as outcome measures. Exp Eye Res. 2005;80(2):259–272. [DOI] [PubMed] [Google Scholar]
- Roman AJ, Cideciyan AV, Aleman TS, Jacobson SG. Full-field stimulus testing (FST) to quantify visual perception in severely blind candidates for treatment trials. Physiol Meas. 2007;28(8):N51–N56. [DOI] [PubMed] [Google Scholar]
- 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;390(10097):849–860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saari JC, Nawrot M, Kennedy BN, Garwin GG, Hurley JB, Huang J, et al. Visual cycle impairment in cellular retinaldehyde binding protein (CRALBP) knockout mice results in delayed dark adaptation. Neuron. 2001;29(3):739–748. [DOI] [PubMed] [Google Scholar]
- Schimmer J, Breazzano S. Investor outlook: Focus on upcoming LCA2 gene therapy phase III results. Hum Gene Ther Clin Dev. 2015;26(3):144–149. [DOI] [PubMed] [Google Scholar]
- Schwartz SB, Aleman TS, Cideciyan AV, Swaroop A, Jacobson SG, Stone EM. De novo mutation in the RP1 gene (Arg677ter) associated with retinitis pigmentosa. Invest Ophthalmol Vis Sci. 2003;44(8):3593–3597. [DOI] [PubMed] [Google Scholar]
- Seabra MC, Brown MS, Goldstein JL. Retinal degeneration in choroideremia: deficiency of rab geranylgeranyl transferase. Science. 1993;259(5093):377–381. [DOI] [PubMed] [Google Scholar]
- Sears AE, Bernstein PS, Cideciyan AV, Hoyng C, Charbel Issa P, Palczewski K, et al. Towards treatment of Stargardt disease: Workshop organized and sponsored by the Foundation Fighting Blindness. Transl Vis Sci Technol. 2017;6(5):6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sengillo JD, Justus S, Tsai YT, Cabral T, Tsang SH. Gene and cell-based therapies for inherited retinal disorders: An update. Am J Med Genet C Semin Med Genet. 2016;172(4):349–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith AJ, Schlichtenbrede FC, Tschernutter M, Bainbridge JW, Thrasher AJ, Ali RR. AAV-Mediated gene transfer slows photoreceptor loss in the RCS rat model of retinitis pigmentosa. Mol Ther. 2003;8(2):188–195. [DOI] [PubMed] [Google Scholar]
- Smith L, Byers JF. Gene therapy in the post-Gelsinger era. JONAS Healthc Law Ethics Regul. 2002;4(4):104–110. [DOI] [PubMed] [Google Scholar]
- Somia N, Verma IM. Gene therapy: trials and tribulations. Nat Rev Genet. 2000;1(2):91–99. [DOI] [PubMed] [Google Scholar]
- Song C, Conlon TJ, Deng WT, Coleman KE, Zhu P, Plummer C, et al. Toxicology and pharmacology of an AAV vector expressing codon-optimized RPGR in RPGR-deficient Rd9 mice. Hum Gene Ther Clin Dev. 2018;29(4):188–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sparrow JE, Parish CA, Hashimoto M, Nakanishi K. A2E, a lipofuscin fluorophore, in human retinal pigmented epithelial cells in culture. Invest Ophthalmol Vis Sci. 1999;40(12):2988–2995. [PubMed] [Google Scholar]
- Strauss RW, Kong X, Ho A, Jha A, West S, Ip M, et al. Progression of Stargardt disease as determined by fundus autofluorescence over a 12-month period: ProgStar Report No. 11. JAMA Ophthalmol. 2019;137:1134–1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stone EM, Cideciyan AV, Aleman TS, Scheetz TE, Sumaroka A, Ehlinger MA, et al. Variations in NPHP5 in patients with nonsyndromic leber congenital amaurosis and Senior-Loken syndrome. Arch Ophthalmol. 2011;129(1):81–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stone EM, Andorf JL, Whitmore SS, DeLuca AP, Giacalone JC, Streb LM, et al. Clinically focused molecular investigation of 1000 consecutive families with inherited retinal disease. Ophthalmology. 2017;124(9):1314–1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suber ML, Pittler SJ, Qin N, Wright GC, Holcombe V, Lee RH, et al. Irish setter dogs affected with rod/cone dysplasia contain a nonsense mutation in the rod cGMP phosphodiesterase beta-subunit gene. Proc Natl Acad Sci U S A. 1993;90(9):3968–3972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sumaroka A, Matsui R, Cideciyan AV, McGuigan DB 3rd, Sheplock R, Schwartz SB, et al. Outer retinal changes including the ellipsoid zone band in Usher syndrome 1B due to MYO7A mutations. Invest Ophthalmol Vis Sci. 2016;57(9):OCT253–OCT261. [DOI] [PubMed] [Google Scholar]
- Sumaroka A, Garafalo AV, Cideciyan AV, Charng J, Roman AJ, Choi W, et al. Blue cone monochromacy caused by the C203R missense mutation or large deletion mutations. Invest Ophthalmol Vis Sci. 2018;59(15):5762–5772. [DOI] [PubMed] [Google Scholar]
- Sumaroka A, Garafalo AV, Semenov EP, Sheplock R, Krishnan AK, Roman AJ, Jacobson SG, Cideciyan AV. Treatment potential for macular cone vision in Leber congenital amaurosis due to CEP290 or NPHP5 mutations: Predictions from artificial intelligence. Invest Ophthalmol Vis Sci. 2019;60(7):2551–2562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Syed N, Smith JE, John SK, Seabra MC, Aguirre GD, Milam AH. Evaluation of retinal photoreceptors and pigment epithelium in a female carrier of choroideremia. Ophthalmology. 2001;108:711–720. [DOI] [PubMed] [Google Scholar]
- Syed R, Sundquist SM, Ratnam K, Zayit-Soudry S, Zhang Y, Crawford V, et al. High-resolution images of retinal structure in patients with choroideremia. Invest Ophthalmol Vis Sci. 2013;54(2):950–961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tatour Y, Tamaiev J, Shamaly S, Colombo R, Bril E, Rabinowitz T, et al. A novel intronic mutation of PDE6B is a major cause of autosomal recessive retinitis pigmentosa among Caucasus Jews. Mol Vis. 2019;25:155–164. [PMC free article] [PubMed] [Google Scholar]
- Thiadens AA, Roosing S, Collin RW, van Moll-Ramirez N, van Lith-Verhoeven JJ, van Schooneveld MJ, et al. Comprehensive analysis of the achromatopsia genes CNGA3 and CNGB3 in progressive cone dystrophy. Ophthalmology. 2010;117(4):825–830. [DOI] [PubMed] [Google Scholar]
- Tobias P, Philipp SI, Stylianos M, Martin B, Barbara W, Felix R, et al. Safety and toxicology of ocular gene therapy with recombinant AAV vector rAAV.hCNGA3 in nonhuman primates. Hum Gene Ther Clin Dev. 2019;30(2):50–56. [DOI] [PubMed] [Google Scholar]
- Tolmachova T, Anders R, Abrink M, Bugeon L, Dallman MJ, Futter CE, et al. Independent degeneration of photoreceptors and retinal pigment epithelium in conditional knockout mouse models of choroideremia. J Clin Invest. 2006;116(2):386–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tolmachova T, Wavre-Shapton ST, Barnard AR, MacLaren RE, Futter CE, Seabra MC. Retinal pigment epithelium defects accelerate photoreceptor degeneration in cell type-specific knockout mouse models of choroideremia. Invest Ophthalmol Vis Sci. 2010;51(10):4913–4920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tolmachova T, Tolmachov OE, Barnard AR, de Silva SR, Lipinski DM, Walker NJ, et al. Functional expression of Rab escort protein 1 following AAV2-mediated gene delivery in the retina of choroideremia mice and human cells ex vivo. J Mol Med (Berl). 2013;91(7):825–837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trapani I, Auricchio A. Seeing the light after 25 years of retinal gene therapy. Trends Mol Med. 2018;24(8):669–681. [DOI] [PubMed] [Google Scholar]
- Tsang SH, Tsui I, Chou CL, Zernant J, Haamer E, Iranmanesh R, et al. A novel mutation and phenotypes in phosphodiesterase 6 deficiency. Am J Ophthalmol. 2008;146(5):780–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Hooser JP, Aleman TS, He Y, Cideciyan AV, Kuksa V, Puttler SJ, et al. Rapid restoration of visual pigment and function with oral retinoid in a mouse model of childhood blindness. PNAS. 2000;97(15):8623–8628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vasireddy V, Mills JA, Gaddameedi R, Basner-Tschakarjan E, Kohnke M, Black AD, et al. AAV-mediated gene therapy for choroideremia: preclinical studies in personalized models. PLoS One. 2013;8(5):e61396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verbakel SK, van Huet RAC, Boon CJF, den Hollander AI, Collin RWJ, Klaver CCW, et al. Non-syndromic retinitis pigmentosa. Prog Retin Eye Res. 2018;66:157–186. [DOI] [PubMed] [Google Scholar]
- Vervoort R, Lennon A, Bird AC, Tulloch B, Axton R, Miano MG, et al. Mutational hot spot within a new RPGR exon in X-linked retinitis pigmentosa. Nat Genet. 2000;25(4):462–466. [DOI] [PubMed] [Google Scholar]
- Vollrath D, Feng W, Duncan JL, Yasumura D, D'Cruz PM, Chappelow A, et al. Correction of the retinal dystrophy phenotype of the RCS rat by viral gene transfer of Mertk. Proc Natl Acad Sci U S A. 2001. October 23;98(22):12584–12589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weng J, Mata NL, Azarian SM, Tzekov RT, Birch DG, Travis GH. Insights into the function of Rim protein in photoreceptors and etiology of Stargardťs disease from the phenotype in abcr knockout mice. Cell. 1999;98(1):13–23. [DOI] [PubMed] [Google Scholar]
- Williams DS, Lopes VS. The many different cellular functions of MYO7A in the retina. Biochem Soc Trans. 2011;39(5):1207–1210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilson RF. The death of Jesse Gelsinger: new evidence of the influence of money and prestige in human research. Am J Law Med. 2010;36(2–3):295–325. [DOI] [PubMed] [Google Scholar]
- Wissinger B, Gamer D, Jägle H, Giorda R, Marx T, Mayer S, et al. CNGA3 mutations in hereditary cone photoreceptor disorders. Am J Hum Genet. 2001;69(4):722–737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wright AF, Bhattacharya SS, Clayton JF, Dempster M, Tippett P, McKeown CM, et al. Linkage relationships between X-linked retinitis pigmentosa and nine short-arm markers: exclusion of the disease locus from Xp21 and localization to between DXS7 and DXS14. Am J Hum Genet. 1987;41(4):635–644. [PMC free article] [PubMed] [Google Scholar]
- Wright AF, Chakarova CF, Abd El-Aziz MM, Bhattacharya SS. Photoreceptor degeneration: genetic and mechanistic dissection of a complex trait. Nat Rev Genet. 2010;11(4):273–84. [DOI] [PubMed] [Google Scholar]
- Wu Z, Hiriyanna S, Qian H, Mookherjee S, Campos MM, Gao C, et al. A long-term efficacy study of gene replacement therapy for RPGR-associated retinal degeneration. Hum Mol Genet. 2015; 24(14):3956–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xue K, Jolly JK, Barnard AR, Rudenko A, Salvetti AP, Patrício MI, et al. Beneficial effects on vision in patients undergoing retinal gene therapy for choroideremia. Nat Med. 2018a;24(10):1507–1512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xue K, MacLaren RE. Ocular gene therapy for choroideremia: clinical trials and future perspectives. Expert Rev Ophthalmol. 2018b;13(3):129–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xue Y, Shen SQ, Jui J, Rupp AC, Byrne LC, Hattar S,et al. CRALBP supports the mammalian retinal visual cycle and cone vision. J Clin Invest. 2015;125(2):727–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang P, Michaels KV, Courtney RJ, Wen Y, Greninger DA, Reznick L, et al. Retinal morphology of patients with achromatopsia during early childhood: implications for gene therapy. JAMA Ophthalmol. 2014;132(7):823–831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye GJ, Budzynski E, Sonnentag P, Nork TM, Miller PE, McPherson L, et al. Safety and biodistribution evaluation in CNGB3-deficient mice of rAAV2tYF-PR1.7-hCNGB3, a recombinant AAV vector for treatment of achromatopsia. Hum Gene Ther Clin Dev. 2016a;27(1):27–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye GJ, Budzynski E, Sonnentag P, Nork TM, Miller PE, Sharma AK, et al. , Safety and biodistribution evaluation in cynomolgus macaques of rAAV2tYF-PR1.7-hCNGB3, a recombinant AAV vector for treatment of achromatopsia. Hum Gene Ther Clin Dev. 2016b;27(1):37–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye GJ, Komáromy AM, Zeiss CJ, Calcedo R, Harman CD, Koehl KL, et al. Safety and efficacy of AAV5 vectors expressing human or canine CNGB3 in CNGB3-mutant dogs. Hum Gene Ther Clin Dev. 2017;28(4):197–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu W, Mookherjee S, Chaitankar V, Hiriyanna S, Kim J, Brooks M, et al. , Nrl knockdown by AAV-delivered CRISPR/Cas9 prevents retinal degeneration in mice. Nat Commun. 2017;8:14716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zallocchi M, Binley K, Lad Y, Ellis S, Widdowson P, Iqball S, et al. EIAV-based retinal gene therapy in the shaker1 mouse model for usher syndrome type 1B: development of UshStat. PLoS One. 2014;9(4):e94272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zelinger L, Cideciyan AV, Kohl S, Schwartz SB, Rosenmann A, Eli D, et al. Genetics and disease expression in the CNGA3 form of achromatopsia: steps on the path to gene therapy. Ophthalmology. 2015;122(5):997–1007. [DOI] [PubMed] [Google Scholar]
- Zhang N, Tsybovsky Y, Kolesnikov AV, Rozanowska M, Swider M, Schwartz SB, et al. Protein misfolding and the pathogenesis of ABCA4-associated retinal degenerations. Hum Mol Genet. 2015;24(11):3220–3237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zimmermann M, Lubinga SJ, Banken R, Rind D, Cramer G, Synnott PG, et al. Cost utility of voretigene neparvovec for biallelic RPE65-mediated inherited retinal disease. Value Health. 2019;22(2):161–167. [DOI] [PubMed] [Google Scholar]
- Zobor D, Zobor G, Kohl S. Achromatopsia: on the doorstep of a possible therapy. Ophthalmic Res. 2015;54(2):103–108. [DOI] [PubMed] [Google Scholar]
- Zobor D, Werner A, Stanzial F, Benedicenti F, Rudolph G, Kellner U, et al. The clinical phenotype of CNGA3-related achromatopsia: Pretreatment characterization in preparation of a gene replacement therapy trial. Invest Ophthalmol Vis Sci. 2017;58(2):821–832. [DOI] [PubMed] [Google Scholar]
