Abstract
Retinal degenerative diseases are a major cause of blindness. Retinal gene therapy is a trail-blazer in the human gene therapy field, leading to the first FDA approved gene therapy product for a human genetic disease. The application of Clustered Regularly Interspaced Short Palindromic Repeat/Cas9 (CRISPR/Cas9)-mediated gene editing technology is transforming the delivery of gene therapy. We review the history, present, and future prospects of retinal gene therapy.
Introduction
The eye is an intricate sensory organ, loosely designed like a camera, in which the retina captures high resolution images functioning similar to the film (Fig. 1). The retina is composed of multiple layers of neurons which convert visible light images into electrical signals and transmit them to the brain (Fig. 2). The retinal photoreceptors are the primary light detecting cells (Fig. 2), whereas pigment epithelial cells play a crucial role in supporting photoreceptor cell functions including phagocytosis and regeneration of visual pigments via the visual cycle.
Figure 1.
Structure of the eye comparing to a camera. Illustration adapted from American Academy of Ophthalmology.
Figure 2.
Schematic of the eye and retina structure. The magnified area represents different cell types in the retina. Most of retinal gene therapy trials are directed to defective genes affecting photoreceptors or RPE.
Loss of vision caused by inherited or acquired retinal diseases can significantly impact on quality of life. In the past several decades, scientists and physicians have begun to unravel the underlying molecular and genetic factors contributing to the onset and progression of these diseases. This review discusses recent early-stage clinical trials and a variety of preclinical studies in which the gene editing techniques resulted in significant functional improvement in the retina. It focuses mostly on the gene therapies that have showed the most progress and the greatest clinical potential. We list all retinal gene therapy trials that have been registered in ClinicalTrial.org in Table 1.
Table 1.
Information on recent gene therapy clinical trials adapted from ClinicalTrials.gov.
| Condition | Identifier | Status | Study Title | Interventions | Sponsor | 
|---|---|---|---|---|---|
| RPE65 - Leber Congenital Amaurosis | NCT00643747 | Completed | Safety Study of RPE65 Gene Therapy to Treat Leber Congenital Amaurosis | rAAV 2/2.hRPE65p.hRPE65 (tgAAG76) | University College, London | 
| NCT00749957 | Completed | Phase 1/2 Safety and Efficacy Study of AAV-RPE65 Vector to Treat Leber Congenital Amaurosis | rAAV2-CB-hRPE65 | Applied Genetic Technologies Corp | |
| NCT00821340 | Completed | Clinical Trial of Gene Therapy for Leber Congenital Amaurosis Caused by RPE65 Mutations | rAAV2-hRPE65 | Hadassah Medical Organization | |
| NCT01496040 | Completed | Clinical Gene Therapy Protocol for the Treatment of Retinal Dystrophy Caused by Defects in RPE65 | rAAV2-hRPE65 | Nantes University Hospital | |
| NCT00481546 | Active | Phase I Trial of Gene Vector to Patients With Retinal Disease Due to RPE65 Mutations | rAAV2-CBSB-hRPE65 | University of Pennsylvania | |
| NCT00516477 | Active | Safety Study in Subjects with Leber Congenital Amaurosis | AAV2-hRPE65v2 | Spark Therapeutics | |
| NCT00999609 | Active | Safety and Efficacy Study in Subjects With Leber Congenital Amaurosis | AAV2-hRPE65v2 | Spark Therapeutics | |
| NCT01208389 | Active | Phase 1 Follow-on Study of AAV2-hRPE65v2 Vector in Subjects With Leber Congenital Amaurosis (LCA) 2 | AAV2-hRPE65v2 | Spark Therapeutics | |
| NCT02946879 | Recruiting | Long-Term Follow-Up Gene Therapy Study for Leber Congenital Amaurosis OPTIRPE65 (Retinal Dystrophy Associated with Defects in RPE65) | AAV OPTIRPE65 | MeiraGTx UK II Ltd | |
| NCT02781480 | Recruiting | Clinical Trial of Gene Therapy for the Treatment of Leber Congenital Amaurosis (LCA) | AAV RPE65 | MeiraGTx UK II Ltd | |
| MERTK-associated retinitis pigmentosa | NCT01482195 | Recruiting | Trial of Ocular Subretinal Injection of a Recombinant Adeno-Associated Virus (rAAV2-VMD2-hMERTK) Gene Vector to Patients With Retinal Disease Due to MERTK Mutations | rAAV2-VMD2-hMERTK | Fowzan Alkuraya | 
| Usher syndrome | NCT01505062 | Recruiting | Study of UshStat in Patients With Retinitis Pigmentosa Associated With Usher Syndrome Type 1B | EIAV-CMV-MYO7A (UshStat) | Sanofi | 
| NCT02065011 | Enrolling by invitation | A Study to Determine the Long-Term Safety, Tolerability and Biological Activity of UshStat® in Patients With Usher Syndrome Type 1B | EIAV-CMV-MYO7A (UshStat) | Sanofi | |
| Stargardt disease | NCT01367444 | Recruiting | Phase I/II Study of SAR422459 in Patients With Stargardt’s Macular Degeneration | EIAV-ABCA4 (SAR422459) | Sanofi | 
| NCT01736592 | Recruiting | Phase I/II Follow-up Study of SAR422459 in Patients With Stargardt’s Macular Degeneration | EIAV-ABCA4 (SAR422459) | Sanofi | |
| Choroideremia | NCT01461213 | Completed | Gene Therapy for Blindness Caused by Choroideremia | rAAV2.REP1 | University of Oxford | 
| NCT02553135 | Completed | Choroideremia Gene Therapy Clinical Trial | AAV2-REP1 | Byron Lam | |
| NCT02077361 | Completed | An Open Label Clinical Trial of Retinal Gene Therapy for Choroideremia | rAAV2.REP1 vector | Ian M. MacDonald | |
| NCT02671539 | Active | THOR - Tübingen Choroideremia Gene Therapy Trial | rAAV2.REP1 | STZ eyetrial | |
| NCT02341807 | Active | Safety and Dose Escalation Study of AAV2-hCHM in Subjects with CHM (Choroideremia) Gene Mutations | AAV2-hCHM | Spark Therapeutics | |
| NCT02407678 | Recruiting | REP1 Gene Replacement Therapy for Choroideremia | AAV-REP1 | University of Oxford | |
| NCT03496012 | Recruiting | Efficacy and Safety of AAV2-RPE1 for the Treatment of Choroideremia | AAV2-RPE1 | Nightstar Therapeutics | |
| NCT03507686 | Recruiting | A Safety Study of Retinal Gene Therapy for Choroideremia (GEMINI) | AAV2-REP1 | Nightstar Therapeutic | |
| Wet age-related macular degeneration | NCT01494805 | Completed | Safety and Efficacy Study of rAAV.sFlt-1 in Patients With Exudative Age-Related Macular Degeneration | rAAV.sFlt-1 | Lions Eye Institute | 
| NCT00109499 | Completed | Study of AdGVPEDF.11D in Neovascular Age-related Macular Degeneration (AMD) | AdGVPEDF.11D | GenVec | |
| NCT01301443 | Completed | Phase I Dose Escalation Safety Study of RetinoStat in Advanced Age-Related Macular Degeneration (AMD) (GEM) | RetinoStat (EIAV-CMV-hEndo-hAngio) | Oxford BioMedica | |
| NCT01024998 | Active | Safety and Tolerability Study of AAV2-sFLT01 in Patients with Neovascular Age-Related Macular Degeneration (AMD) | AAV2-sFLT01 | Genzyme, a Sanofi Company | |
| NCT03066258 | Recruiting | RGX-314 Gene Therapy for Neovascular AMD Trial | AAV-VEGF (RGX-314) | Regenxbio Inc. | |
| NCT01678872 | Enrolling by invitation | A Follow-up Study to Evaluate the Safety of RetinoStat® in Patients With Age-Related Macular Degeneration | RetinoStat (EIAV-CMV-hEndo-hAngio) | Oxford BioMedica | |
| Achromatopsia | NCT03278873 | Recruiting | Long-Term Follow-Up Gene Therapy Study for Achromatopsia CNGB3 | AAV - CNGB3 | MeiraGTx UK II Ltd | 
| NCT02599922 | Recruiting | Safety and Efficacy Trial of AAV Gene Therapy in Patients With CNGB3 Achromatopsia | rAAV2tYF-PR1.7-hCNGB3 | Applied Genetic Technologies Corp | |
| NCT03001310 | Recruiting | Gene Therapy for Achromatopsia (CNGB3) | AAV-CNGB3 | MeiraGTx UK II Ltd | |
| NCT02935517 | Recruiting | Safety and Efficacy Trial of AAV Gene Therapy in Patients With CNGA3 Achromatopsia | AGTC-402 | Applied Genetic Technologies Corp | |
| NCT02610582 | Active | Safety and Efficacy of a Single Subretinal Injection of rAAV.hCNGA3 in Patients with CNG3-linked Achromatopsia | rAAV.hCNGA3 | STZ eyetrial | |
| X-linked retinoschisis | NCT02317887 | Recruiting | Study of RS1 Ocular Gene Transfer for X-linked Retinoschisis | AAV RS1 | National Eye Institute (NEI) | 
| NCT02416622 | Recruiting | Safety and Efficacy of rAAV-hRS1 in Patients With X-linked Retinoschisis (XLRS) | rAAV2tYF-CB-hRS1 | Applied Genetic Technologies Corp | 
RPE65-Leber Congenital Amaurosis
RPE65 is a retinal pigment epithelial-specific protein (65 kDa), which is almost exclusively found in the retinal pigment epithelium (RPE) as a retinoid isomerase and is responsible for converting all-trans retinoid to 11-cis retinal during pigment regeneration.1–3 Absence or deactivation of RPE65 results in an accumulation of all-trans-retinyl esters, which promotes pigment degeneration, disabling the formation of visual pigments, rhodopsin and cone opsin because of a lack of 11-cis retinal.4–6 Therefore, mutations in Rpe65 associate strongly with RPE65-Leber Congenital Amaurosis (LCA2), retinitis pigmentosa (RP), and early-onset severe retinal dystrophies.7–10
The absence of this isomerase activity along with pigment degeneration and significant visual impairment were observed in Rpe65 knockout mice, mutant knock-in mice, and naturally occurring Rpe65 mutant mouse.11–13 Gene therapies using adeno-associated virus (AAV), adenovirus, and lentivirus-mediated Rpe65 delivery have all resulted in improvement in electroretinogram (ERG) response and visual function test in RPE65 deficient mice models (Fig. 3).14,15 Subretinal injection with AAV1-RPE65 to a Rpe65 knockout (Rpe65−/−) mouse model of LCA2 as early as in-utero resulted in substantial improvements in ERG responses lasting as late as 24 months of age.16 Another study in which AAV5-RPE65 was subretinally delivered to a naturally occurring Rpe65 mutant mouse model, rd12, also showed improvements in ERG responses and visual guided behaviors.17 Additional experiments with the rd12 mice receiving subretinal AAV2-RPE65 delivery attempted to establish an in vivo bioassay to evaluate the stability of vectors used in clinical trials of LCA2.18
Figure 3.
Diagrams of gene delivery vectors including adeno-associated virus (AAV), adenovirus, and second-generation lentivirus. ITR, inverted terminal repeats; Rep, Replication; Cap, Capsid; E2A/E3/E4/VA, adenovirus genes that mediate replication. LTR, long terminal repeats; ψ/RRE, Rev response element; VSV-G, vesicular stomatitis virus G protein; GAG, Group-specific antigen; pol, DNA polymerase; tat, Trans-Activator of Transcription.
Although cone photoreceptors use a pathway independent of the RPE for chromophore recycling that enables them to function in continuous bright light, studies of patients affected with LCA2 suggest that cone survival is still dependent on RPE65 isomerase activity, regardless of the residual cone activity in the absence of the enzyme.19–21 This is consistent with observations of rapid cone degeneration in Rpe65−/− and rd12 models.3,22 Self-complementary AAV-RPE65 vectors have been shown to be capable of restoring cone function and preventing cone degeneration in both rd12 mice and Rpe65 and Rhodopsin double knockout (Rpe65−/−Rho−/−) mice.23,24
The first gene therapy for LCA2 in a large animal model gained widespread attention in 2001 using a recombinant adeno-associated virus (rAAV2) vector containing Rpe65 cDNA to treat three Rpe65 mutant dogs (Fig. 3).25 In this study, a subretinal delivery of the canine Rpe65 gene carried by the rAAV2 vector, under the control of the hybrid cytomegalovirus/chicken β-actin (CBA) promoter, resulted in substantial visual improvements as assessed by ERG (Fig. 4). Follow-up studies found that subretinal delivery of recombinant AAV1, AAV4, and AAV5-mediated RPE65 expression driven by a human promoter were also capable of restoring visual function, which remained stable over time.26–33 Additional follow-ups found that cortical responses, assessed by functional magnetic resonance imaging (fMRI), were significantly improved and visually guided behavior was recovered in treated dogs, suggesting that retinal signals were correctly propagated to the visual processing centers of the brain.28,31,32,34
Figure 4.
Delivery of a viral vector via intraocular injection. Maximal 1–2 μl of viral mixture in rodents or 200 μl in large eyes can be injected into the subretinal space through a small scleral incision. A successful injection was judged by creation of a small subretinal fluid bleb.
Unlike most conventional treatment methods, the efficacy of gene therapies for LCA2 is not heavily affected by the disease stage. The previously mentioned studies, which demonstrated significant therapeutic efficacy, included animals exhibiting mid-to-late stage disease, such as dogs treated at 30 months of age and rd12 mice treated at 3 months of age.26,35 Even Rpe65−/− mice treated as late as 24 months of age resulted in mild but significant (16%) ERG improvements.36 These results indicate that an adult patient qualified for Phase I trials would have a reasonable chance of obtaining improved visual function after treatment.
Previous animal studies laid a foundation of proof-of-concept studies that allowed researchers to conduct multiple phase I/II trials which eventually led to an FDA-approved gene therapy after a successful phase III trial in late 2017/early 2018.37–43 The studies delivered an AAV2 vector carrying a normal human Rpe65 cDNA through subretinal injection. All trials reported clinically measurable visual improvement, albeit with varied magnitude and significance. In addition, no vector-related adverse events or toxic immune responses were elicited despite differences in postoperative steroid use, doses delivered, promoters used, vector specifics, and anesthesia during delivery.41 Improvement in vision was maintained 3 years after treatment,42,44 but progressively declined after 6 years because of photoreceptor degeneration in the treated retina as in the untreated retina.45 Moreover, in cases where vector was delivered subfoveally and caused a foveal detachment, patients with LCA2 typically reported no change in foveal light sensitivity but instead reported improvement extrafoveally.39,46 In a 3-year study, nearly half of the patients experienced a detached fovea caused by a vector bleb and further foveal thickness loss.44 A follow-up with optical coherence tomography (OCT) analysis suggested that the loss of thickness resulted from foveal cone loss, which can occur from potentially damaging effects of subretinal vector-mediated foveal detachment,45 suggesting that subretinal vector delivery in this locale should be approached cautiously. Notably, in this study, a few patients with improved extrafoveal function experienced a shift of their visual fixation away from the fovea into the superior-temporal retina, known as the ‘pseudo-fovea’, that coincided with the locale of the vector bleb.47 This phenomenon results in a change in cortical control of the ocular muscles such that images are positioned on this new, more light-sensitive pseudo-fovea.
In summary, human gene therapy for RPE65-LCA2 has been shown to be safe, free of serious complications, and effective at improving impaired vision, yet still needs more investigation and exploration.
MERTK-associated autosomal recessive retinitis pigmentosa
Retinitis pigmentosa (RP) is a retinal rod photoreceptor specific disease characterized by primary rod photoreceptor death and degeneration, followed by secondary cone death.48 RP is one of the most common inherited blinding retinal diseases, affecting more than one million patients worldwide.49,50 The mer receptor tyrosine kinase (MERTK) is a member of the Axl/Mer/Tyro3 receptor tyrosine kinase family and is necessary for proper phagocytosis of photoreceptor outer segments by the RPE. The MERTK-associated form of autosomal recessive retinitis pigmentosa (arRP) is caused by an absence of functional MERTK expression, leading to significant degeneration of the retina.51 This disease is very rare; it is only found in isolated populations identified in the Middle East and the Faroe Islands. Nevertheless, the profound impact it has on a patient’s quality of life has attracted the attention of the scientific community.52–54 Since then, several groups have reported numerous isolated cases of MERTK-associated arRP in other parts of the world.51,55 Studies simultaneously identifying patients with RP and homozygous mutations in Mertk conclusively linked this gene to the disease.56 Degeneration of the retina is caused by accumulation of subretinal debris of shed photoreceptor outer segments resulting from inability of the RPE to perform phagocytosis, leading to consequent apoptosis of retinal cells and progressive deterioration of visual function as evaluated by ERG.57–59
The most successful gene-replacement study for MERTK-associated arRP used a lentivirus expressing MERTK and was successful at preserving retinal structure and function, as observed by microscopy and ERG up to 7 months post-injection (Fig. 3).60 A later study used a fast-acting AAV8 Y733F capsid mutant vector as early treatment for long-term preservation of retinal function in a mouse model; this treatment method can quickly restore MERTK expression before a significant debris field can incite apoptosis in photoreceptors.61 The effect of Mertk gene therapy was shown to be improved with a co-administration of AAV expressing glial cell derived neurotropic factor (GDNF).62 Most recently, a new method, homology-independent targeted integration (HITI), was reported to treat a Royal College of Surgeons (RCS) rat, a well-established animal model for RP resulting from a homozygous 1.9-kb deletion from intron 1 to exon 2 in the Mertk gene.63 The HITI used CRISPR/Cas9-mediated gene editing to endogenously insert a wild-type exon 2 at Mertk locus. Subretinal injection of HITI-AAVs led to statistically significant increases in Mertk mRNA and protein expression levels, preservation of the retina outer nuclear layer (ONL) thickness, and significant improvement in ERG b-wave responses.63
A phase I clinical trial of six patients showed no complications that could be attributed to the gene vector and resulted in improved visual acuity in three of the patients (ClinicalTrials.gov Identifier: NCT01482195). However, at 2-year follow-up, two of these patients lost these improvements, although disease progression could have caused this. Based on the established safety profile, the trials are still recruiting to assess the efficacy of this approach, especially in the population with higher starting visual acuity.64
Usher syndrome
Usher syndrome (USH) is a heterogeneous collection of autosomal recessive disorders, causing a combination of deafness and blindness in people with an estimated three-to-six-person prevalence per 100 000 individuals.65–67 It accounts for 15–20% of RP cases, and 50% of combined blindness and deafness cases.68,69 The three clinical subtypes of USH (USH1, USH2, USH3) are distinguished by severity and the progression of hearing loss, presence or absence of vestibular dysfunction, and vision loss from RP, with USH1 being the most severe in terms of onset, extent of hearing loss, and RP.70–72 Currently, there are 11 protein-encoding genes associated with USH reported in the literature. They are considered to be important for stability and development of the inner hair bundle, photoreceptor cilium, and phagocytosis of the RPE.73–75
The most prevalent causative gene for USH is myosin VIIa (Myo7A), which encodes a critical actin-base protein functioning in the inner ear and retina.70,76 Mutations in Myo7A (USH1b gene) account for approximately 60% of all USH1 cases, causing deafness, vestibular dysfunction, and retinal degeneration with onset during childhood.71,77 MYO7A has been found to be expressed in the RPE, the photoreceptor connecting cilia and synapses. It is proposed to play a role in intracellular transport, endocytosis, and cell-cell adhesion.78–81
There are several reported mouse models containing mutations in the Myo7A gene.82 While they all display deafness and vestibular dysfunction phenotypes, their photoreceptors do not undergo degeneration as do human ones.83,84 However, shaker1 mice, which carry a mutated Myo7A gene, have been shown to exhibit retinal degeneration when exposed to cycles of bright light.85,86 This is thought to be caused by pathologies in melanosome localization, opsin transport through the collecting cilium, and dysregulation of transducin translocation.87–90
Before human trials began, some studies demonstrated both the requirement of MYO7A for the apical localization of melanosomes in human RPE cells and that RPE melanosome localization and opsin transport could be restored in the shaker1 mouse using a lentivirus containing Myo7A delivered through subretinal injection.91,92 After these findings, Oxford Biomedica UK launched a phase I clinical trial to evaluate the safety of subretinally delivered Myo7A using an equine infectious anemia virus (EAIV) with lentiviral vector (UshStat) in patients with USH1b (ClinicalTrials.gov Identifier: NCT01505062).93 This was followed by a long-term study of UshStat safety, tolerability, and biological activity (ClinicalTrials.gov Identifier: NCT02065011). Concurrently with the ongoing clinical trials, the safety profile of the EAIV-based Myo7A gene therapy was assessed in rhesus monkeys.93 However, lentiviral transduction is limited mostly to the RPE after subretinal delivery of postnatal retina. There is a clear need to effectively transduce photoreceptors in patients with USH1b as photoreceptors are the site of the earliest disease expression. Studies on mice showed that a photoreceptor mutant phenotype was corrected with HIV-MYO7A.94,95 As AAV capsid capacity is only approximately 4.7 kb, one approach is to split the full-length Myo7A cDNA (6.6kb) into two and package them separately. Depending on the design, these incomplete cDNAs are reconstituted into the full gene through recombination between internal homologous sequences, or trans-splicing, or a hybrid mix of the two strategies.96–99 Dual AAV vector delivery methods have since been used to deliver Myo7A to the subretinal space of C57BL/6 mice,100shaker1 mice,96 as well as pigs.101 These approaches have shown promising results in terms of MYO7A expression in RPE and photoreceptors, but require further investigation to establish a long-term safety profile and therapeutic efficacy before clinical trials.
Stargardt disease
Stargardt disease, an inherited form of juvenile macular degeneration, is both clinically and genetically highly heterogeneous.102 This disease is commonly caused by recessive mutations in ATP-binding cassette, sub-family A, member 4 (ABCA4) gene, which encodes a transporter protein present in photoreceptors and RPE.103 ABCA4 actively transports retinylidene phosphatidylethanol-amine, and phosphatidylethanolamine from the lumen to the cytoplasm of photoreceptor outer segments, playing an important role in the visual cycle.104 Mutations on ABCA4 gene reduce or terminate this transporter activity, leading to a buildup of potentially toxic bisretinoid compounds in the lumen and outer segment membranes of photoreceptors.105,106 The accumulation of toxic bisretinoid compounds leads to lipofuscin accumulation in the RPE,107 followed by degeneration of RPE and later of photoreceptors.108 Mice missing ABCA4 also exhibit formation of lipofuscin granules.109
There are several gene therapy approaches under investigation to treat Stargardt disease, including the use of AAV and lentiviruses. Although, similar to Myo7A, the size of ABCA4 cDNA exceeds the usual packaging capacity of AAVs (4.7 kb) for gene replacement, different procedures have attempted to surmount this challenge. Successful expression of ABCA4 using oversized AAVs in photoreceptors of Abca4−/− mice resulted in improved morphology and function of retina.110 Later, a dual AVV trans-splicing strategy that efficiently reconstituted ABCA4 gene in mice was used and demonstrated significant phenotype improvement.96 Moreover, lentivirus was also used to infect photoreceptors in Abca4−/− mice, which showed better results than AAV-based methods when subretinal injections of the vector were performed on postnatal Days 4 and 5.111 Experiments by Binley et al. achieved even more efficient photoreceptor transduction in retinae of non-human primates using EIAV lentivectors.112
In light of these good results, currently there are two ongoing Phase I/II clinical trials using gene therapy to treat Stargardt disease (ClinicalTrials.gov Identifiers: NCT01367444, NCT01736592). Since 2012, Oxford BioMedica has performed subretinal injections of EIAV lentivirus to deliver the ABCA4 gene to patients with homozygotic mutation of ABCA4 and significant visual impairment, to assess safety and tolerability of ascending doses of the virus in both the short and long term.
Choroideremia
Choroideremia is an X-linked recessive progressive retinochoroidal degenerative disease associated with mutations within the choroideremia (CHM) gene. It affects males in early life, causing night blindness, peripheral visual field loss, and in most cases, complete blindness within the first 30 years of onset.113–117 Chorioretinal atrophy, RPE degeneration, and abnormal ERG responses of retina sensitivity are seen even before patients report visual loss.116 Female carriers often present with altered ERGs and irregular areas of pigmentation on fundus. For females, this disease is generally asymptomatic in early years, although some females can present with severe disease resulting from unequal inactivation of the X chromosome.115,118,119
The CHM gene encodes Rab escort protein 1 (REP1), which recognizes Rab proteins and delivers them to Rab geranylgeranyl transferase (RabGGTs), thereby participating in intracellular vesicular transport and in modification of Rab via addition of geranylgeranyl groups. This process, referred to as prenylation, allows Rabs to attach to lipid bilayers.120,121 Additionally, REP1 escorts prenylated Rabs to their destination membrane.117 Mutations in the CHM gene lead to truncation or absence of REP1, resulting in defects in delivery of opsin to photoreceptor outer segments, in phagocytosis of photoreceptor outer segments by the RPE, and in apical migration of RPE melanosomes.122 As indicated by experimental evidence, the severity of the CHM phenotype correlates with the severity of defects in intracellular trafficking processes.123
Chm knockout mice models showed that disease pathogenesis is associated with independent Rab prenylation defects that trigger photoreceptor and RPE degeneration.124 A study also found that regardless of whether CHM was knocked out in photoreceptors or the RPE, defects in the RPE accelerate degeneration of photoreceptors.125
In vitro and animal studies with Chm knockout models have been performed using a number of AAV serotypes. Restoration of REP1 expression and function have been observed in lymphocytes, fibroblasts, and induced pluripotent stem cells (iPSCs) derived from choroideremia patients using AAV vectors in vitro.126,127 In 2013, AAV2-mediated Chm gene was used to achieve functional expression of REP1 in human cells ex vivo and ChmNull/WT female carriers in vivo.128 AAV8 has also been demonstrated to reverse the biochemical defects both in vitro and in the conditional Chm knockout mice.129
There are several clinical studies ongoing evaluating efficacy and safety of subretinal injections of AAV2-hCHM (Table 1). A completed phase I/II clinical trial (ClinicalTrials.gov Identifier: NCT01461213) showed retina sensitivity, visual improvement, and treatment safety in patients treated with subfoveal injections of AAV-REP1.130
Wet age-related macular degeneration
Wet (exudative) age-related macular degeneration (AMD) is the leading cause of blindness for people over 65 years of age. Current treatments for AMD involve inhibition of vascular endothelial growth factor (VEGF) with antibodies, RNA aptamers, or soluble receptors.131–133 VEGF is implicated in intraocular neovascularization associated with diabetic retinopathy and age-related macular degeneration, promoting damaging neovascularization in the choroidal vasculature.134,135 The current VEGF antibody treatment used in practice requires long-term repetitive intravitreal injections, which places significant financial and psychological burden on the patient.
All VEGF isoforms bind to two type III receptor tyrosine kinases, FLT1 and KDR (also known as FLK1).136,137 Several studies have shown that injecting AAV2 with full-length soluble FLT-1 (sFLT1) subretinally can safely inhibit ocular neovascularization for up to 8 and 17 months postinjection in mice and in monkeys, respectively.138,139 A study using an AAV2 vector with a chimeric soluble protein (AAV2-sFLT01) revealed that the protein was consistently expressed and was effective at managing neovascularization in a rodent model with minimum toxicity.138–141 In 2017, a phase I/II clinical trial to treat wet AMD was completed (ClinicalTrials.gov Identifier: NCT01494805), in which a single subretinal injection of rAAV.sFlt-1 into a patient’s eyes was found to be safe, highly reproducible, and may reduce ranibizumab retreatments.142,143 Intravitreal injection of AAV2-sFLT01 was also evaluated in another ongoing clinical trial (ClinicalTrials.gov Identifier: NCT01024998) and was found to be safe and well tolerated at all injected doses.144
Soluble VEGF receptors are not the only way to suppress angiogenesis in the eye. Pigment epithelium-derived factor (PEDF), produced during normal wound repair, endostatin, cleavage product of collagen VII, and angiostatin, cleavage product of plasminogen, are endogenous proteins that attenuate physiologic neovascularization.145–147 Studies have shown that AAV-driven upregulation of PEDF, endostatin, or angiostatin, resulted in suppression of laser-induced choroidal neovascularization (CNV) in mice.148–150 This led to the launch of a now completed phase I clinical trial (ClinicalTrials.gov Identifier: NCT01024998), in which a modified AAV-PEDF (AdGVPEDF.11D) was delivered intravitreally.151 The high dose treatment group showed a slightly lower neovascular lesion size than the low dose group, but the effect was not lasting, and thus not viable for management of a chronic disease such as AMD.152 Concurrently, a study reported that subretinal delivery of a EIAV lentivector encoding LacZ resulted in long-term expression of LacZ in the RPE for up to 1 year in mice. Subretinal injection of the same vector, but encoding for murine angiostatin and endostatin, resulted in suppression of laser CNV.153 This resulted in a phase I clinical study completed in 2017, as well as a long-term follow-up cohort (ClinicalTrials.gov Identifiers: NCT01301443 and NCT01678872) in which EIAV expressing endostatin and angiostatin (RetinoStat) was used to treat late-stage AMD. At completion, the trial reported safety, tolerability, and long-term therapeutic gene expression (up to 4.5 years), showing promise as a platform for chronic disease treatment. However, the treatment was not reliable in eliminating sub- and intra-retinal fluid in severe wet AMD.154
Achromatopsia
Achromatopsia (ACHM) is characterized by poor central visual acuity (<20/200), photophobia, complete color blindness, and reduced cone-mediated ERG response amplitudes, and has a prevalence of about 1 in 30 000.155 It has recently been shown that cone degeneration begins early in childhood, with deterioration progressing at a moderate rate.156 A combined 80% of all ACHM cases can be characterized by mutations in genes encoding the cone-specification channel, cyclic nucleotide gated channel α3 and β3 (CNGA3 and CNGB3), while fewer than 5% of all cases combined are caused by mutations in the cone-specific α subunit of transducin (GNAT2), activating transcription factor 6 (ATF6), and α subunit of cone-specific phosphodiesterase (PDE6C).157–159 The first gene therapy for ACHM was performed in a mouse carrying a recessive mutation in Gnat2, resulting in little to no cone-mediated ERG and poor visual acuity. Subretinal injection of AAV5 containing Gnat2 driven by the human red cone opsin promoter was shown to restore cone-mediated ERG amplitudes and cone-mediated behavior responses to the levels of the age-matched wild-type mice.160,161
Delivery of CNGA3 and GNAT2 using AAV5/AAV8-based vectors has been shown to normalize protein expression and improve vision in murine models of CNGA3 and CNGB3 forms of ACHM.162–164 Subretinal injection of AAV5-CNGB3 in ACHM-affected dogs, or AAV5-CNGA3 in diseased sheep both resulted in restoration of cone function and day vision, which lasted up to 33 months in dogs and up to 3 years in sheep.165,166 To optimize the effectiveness, studies also showed that decreasing the length of promoter and using AAV2 with single tyrosine-to-phenylalanine (YF) mutations can increase the efficiency for CNGB3 expression.167,168 These studies have resulted in initiation of several independent phase I/II clinical trials for both CNGA3- and CNGB3-linked ACHM launched in Europe by STZ eyetrial and MeiraGTx, and in the United States by AGTC (Table 1). All these approaches rely on modified rAAV2-based vectors delivered with a single subretinal injection to supplement the affected gene and aim to assess the safety and efficacy of the treatment.
X-linked juvenile retinoschisis
X-linked juvenile retinoschisis (XLRS) is the leading cause of monogenic macular dystrophy with between 1:5 000 and 1:25 000 males afflicted.169 XLRS is typically classified by localized splitting in the retina (schisis) and an unusual electronegative ERG with a preserved a-wave and a diminished b-wave.170–172 It starts with retinal presentation in early childhood, exacerbation of symptoms in teenage years, and then stabilizes during adulthood.169,173
XLRS is associated with mutations in the retinoschisin (RS1) gene, which encodes the RS1 protein (24 kDa) secreted from retinal photoreceptors with a discoidin domain that is likely to be involved in cell adhesion.174–177 It was first reported that intravitreal delivery of an AAV2 vector containing murine RS1 cDNA driven by cytomegalovirus promoter in the Rs1h knockout mice at 13 weeks of age led to visual improvements as tracked by ERG and schisis cavities out to 6 months of age.178 Subretinal delivery of AAV5 vector expressing murine opsin promoter driving human RS1 cDNA or intravitreal delivery of AAV8 vector expressing human retinoschisin promoter driving human RS1 cDNA into Rs1h-KO mice at young stage (P14 to 2 month) both showed improved retinal structure and function.179,180 However, treatment at 7 months of age improved only retinal structure and not ERG function,181 indicating a critical window of treatment. Currently, a phase I/II clinical study is being conducted to evaluate the safety and efficacy of a rAAV vector expressing retinoschisin (rAAV2tYF-CB-hRS1) delivered intravitreally in XLRS patients (ClinicalTrials.gov Identifier: NCT02416622).
CRISPR/Cas9-mediated gene and mutation-independent therapy
Although current gene therapy offers many promising treatments for various human diseases, its application is often limited to a narrow spectrum of diseases and patient population, because it can only be directed to a single gene. Similarly, in current regenerative medicine, the application of endogenous stem cells in tissue repair/regeneration represents an important method in treatment of many diseases. Promising results have been demonstrated in mouse liver, zebrafish heart, and human lens.182–184 However, as in gene therapy, endogenous stem cell treatment can be applied to only a very narrow spectrum of disease. The major challenge is that normal genetic makeup and function are required in the starting cells for tissue regeneration; if the starting cell type harbors a causal genetic mutation which renders subsequently generated cells susceptible to the same disease etiology, then regenerated cells will assume the previous cell fate.
One approach to overcome the above drawbacks is to combine the advantages of both gene therapy and regenerative medicine. The resulting method is called therapeutic cellular reprogramming. Using CRISPR/Cas9-based gene editing, this strategy switches a cell type sensitive to a mutation to a cell type that is resistant to the same mutation, with related function. Therefore, this strategy eliminates the occurrence of underlying mutation, while preserving tissue structure and function. As a result, distantly related cells can be directly converted in vivo by appropriate combinations of developmentally relevant transcription factors,185 expanding the application of conventional regenerative medicine in both disease spectrum and patient population.
The potential of therapeutic cellular reprogramming was first examined on RP. As RP is caused by mutations in over 200 genes, the therapeutic impact of conventional gene therapy is limited. Acute gene knockout of either rod determinant Nrl or its downstream transcriptional factor Nr2e3 showed successful rod to cone reprogramming in adult rod photoreceptors.186,187 The resulting cone photoreceptors demonstrated resistance to mutations in RP-specific genes on rod photoreceptors, which consequently prevented secondary cone loss. More importantly, by combining an AAV-based delivery system with CRISPR/Cas9-mediated targeted inactivation of Nrl or Nr3e3, successfully in vivo reprogramming of rod photoreceptors into cone photoreceptors with consequent retinal photoreceptor preservation and visual rescue was achieved (Fig. 5).188,189 These results indicate that therapeutic cellular reprogramming can serve as a novel treatment approach that is gene- and mutation-independent, broadening implications for genetic disease therapy.
Figure 5.
Conversion between two cell types from the same lineages: reprogramming rod photoreceptors to cone photoreceptors.126
Retinal gene therapy has always been at the forefront of human gene therapy and much progress has been made in retinal gene therapy. The successful approval of the first retinal gene therapy for LCA2 caused by RPE65 mutations has ushered in a new era in human gene therapy. The application of CRISPR/Cas9-mediated gene editing technology is transforming how the gene therapy is administered. We anticipate great progress and further approvals of retinal gene therapy products in the near future.
Conflict of interest statement
The authors declare no conflict of interest.
References
- 1. Moiseyev G, Chen Y, Takahashi Yet al. RPE65 is the isomerohydrolase in the retinoid visual cycle. Proc Nat Acad Sci USA 2005;102:12413–18. 10.1073/pnas.0503460102. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 2. Jin M, Li S, Moghrabi WNet al. Rpe65 is the retinoid isomerase in bovine retinal pigment epithelium. Cell 2005;122:449–59. 10.1016/j.cell.2005.06.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 3. Redmond TM, Yu S, Lee Eet al. Rpe65 is necessary for production of 11-cis-vitamin A in the retinal visual cycle. Nat Genet 1998;20:344–51. 10.1038/3813. [DOI] [PubMed] [Google Scholar]
 - 4. Tang PH, Buhusi MC, Ma JXet al. RPE65 Is Present in Human Green/Red Cones and Promotes Photopigment Regeneration in an In Vitro Cone Cell Model. J Neurosci 2011;31:18618–26. 10.1523/Jneurosci.4265-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 5. Zhang T, Zhang N, Baehr Wet al. Cone opsin determines the time course of cone photoreceptor degeneration in Leber congenital amaurosis. Proc Natl Acad Sci USA 2011;108:8879–84. 10.1073/pnas.1017127108. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 6. Redmond TM, Poliakov E, Yu Set al. Mutation of key residues of RPE65 abolishes its enzymatic role as isomerohydrolase in the visual cycle. Proc Nat Acad Sci USA 2005;102:13658–63. 10.1073/pnas.0504167102. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 7. Gu S, Thompson DA, Srikumari CRSet al. Mutations in RPE65 cause autosomal recessive childhood-onset severe retinal dystrophy. Nat Genet 1997;17:194–7. 10.1038/ng1097-194. [DOI] [PubMed] [Google Scholar]
 - 8. Morimura H, Fishman GA, Grover SAet al. Mutations in the RPE65 gene in patients with autosomal recessive retinitis pigmentosa or leber congenital amaurosis. Proc Natl Acad Sci USA 1998;95:3088–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 9. Marlhens F, Bareil C, Griffoin JMet al. Mutations in RPE65 cause Leber’s congenital amaurosis. Nat Genet 1997;17:139–41. 10.1038/ng1097-139. [DOI] [PubMed] [Google Scholar]
 - 10. Thompson DA, Gal A. Genetic defects in vitamin A metabolism of the retinal pigment epithelium. Dev Ophthalmol 2003;37:141–54. [DOI] [PubMed] [Google Scholar]
 - 11. Wright CB, Chrenek MA, Feng Wet al. The Rpe65 rd12 allele exerts a semidominant negative effect on vision in mice. Invest Ophthalmol Vis Sci 2014;55:2500–15. 10.1167/iovs.13-13574. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 12. Li Y, Yu S, Duncan Tet al. Mouse model of human RPE65 P25L hypomorph resembles wild type under normal light rearing but is fully resistant to acute light damage. Hum Mol Genet 2015;24:4417–28. 10.1093/hmg/ddv178. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 13. Shin Y, Moiseyev G, Chakraborty Det al. A Dominant Mutation in Rpe65, D477G, Delays Dark Adaptation and Disturbs the Visual Cycle in the Mutant Knock-In Mice. Am J Pathol 2017;187:517–27. 10.1016/j.ajpath.2016.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 14. Bemelmans AP, Kostic C, Crippa SVet al. Lentiviral gene transfer of RPE65 rescues survival and function of cones in a mouse model of Leber congenital amaurosis. PLoS Med 2006;3:e347. 10.1371/journal.pmed.0030347. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 15. Chen Y, Moiseyev G, Takahashi Yet al. RPE65 gene delivery restores isomerohydrolase activity and prevents early cone loss in Rpe65-/- mice. Invest Ophthalmol Vis Sci 2006;47:1177–84. 10.1167/iovs.05-0965. [DOI] [PubMed] [Google Scholar]
 - 16. Dejneka NS, Surace EM, Aleman TSet al. In utero gene therapy rescues vision in a murine model of congenital blindness. Mol Ther 2004;9:182–8. 10.1016/j.ymthe.2003.11.013. [DOI] [PubMed] [Google Scholar]
 - 17. Pang JJ, Chang B, Kumar Aet al. Gene therapy restores vision-dependent behavior as well as retinal structure and function in a mouse model of RPE65 Leber congenital amaurosis. Mol Ther 2006;13:565–72. 10.1016/j.ymthe.2005.09.001. [DOI] [PubMed] [Google Scholar]
 - 18. Roman AJ, Boye SL, Aleman TSet al. Electroretinographic analyses of Rpe65-mutant rd12 mice: developing an in vivo bioassay for human gene therapy trials of Leber congenital amaurosis. Mol Vis 2007;13:1701–10. [PubMed] [Google Scholar]
 - 19. Jacobson SG, Aleman TS, Cideciyan AVet al. Human cone photoreceptor dependence on RPE65 isomerase. Proc Natl Acad Sci USA 2007;104:15123–28. 10.1073/pnas.0706367104. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 20. Wang JS, Kefalov VJ. An alternative pathway mediates the mouse and human cone visual cycle. Curr Biol 2009;19:1665–9. 10.1016/j.cub.2009.07.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 21. Wang JS, Estevez ME, Cornwall MCet al. Intra-retinal visual cycle required for rapid and complete cone dark adaptation. Nat Neurosci 2009;12:295–302. 10.1038/nn.2258. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 22. Pang JJ, Chang B, Hawes NLet al. Retinal degeneration 12 (rd12): a new, spontaneously arising mouse model for human Leber congenital amaurosis (LCA). Mol Vis 2005;11:152–62. [PubMed] [Google Scholar]
 - 23. Ku CA, Chiodo VA, Boye SLet al. Gene therapy using self-complementary Y733F capsid mutant AAV2/8 restores vision in a model of early onset Leber congenital amaurosis. Hum Mol Genet 2011;20:4569–81. 10.1093/hmg/ddr391. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 24. Pang J, Boye SE, Lei Bet al. Self-complementary AAV-mediated gene therapy restores cone function and prevents cone degeneration in two models of Rpe65 deficiency. Gene Ther 2010;17:815–26. 10.1038/gt.2010.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 25. Acland GM, Aguirre GD, Ray Jet al. Gene therapy restores vision in a canine model of childhood blindness. Nat Genet 2001;28:92–5. 10.1038/88327. [DOI] [PubMed] [Google Scholar]
 - 26. Narfstrom K, Katz ML, Ford Met al. In vivo gene therapy in young and adult RPE65-/- dogs produces long-term visual improvement. J Hered 2003;94:31–7. [DOI] [PubMed] [Google Scholar]
 - 27. Narfstrom K, Seeliger M, Lai CMet al. Morphological aspects related to long-term functional improvement of the retina in the 4 years following rAAV-mediated gene transfer in the RPE65 null mutation dog. Adv Exp Med Biol 2008;613:139–46. [DOI] [PubMed] [Google Scholar]
 - 28. Bennicelli J, Wright JF, Komaromy Aet al. Reversal of blindness in animal models of Leber congenital amaurosis using optimized AAV2-mediated gene transfer. Mol Ther 2008;16:458–65. 10.1038/sj.mt.6300389. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 29. Le Meur G, Stieger K, Smith AJet al. Restoration of vision in RPE65-deficient Briard dogs using an AAV serotype 4 vector that specifically targets the retinal pigmented epithelium. Gene Ther 2007;14:292–303. 10.1038/sj.gt.3302861. [DOI] [PubMed] [Google Scholar]
 - 30. Jacobson SG, Acland GM, Aguirre GDet al. Safety of recombinant adeno-associated virus type 2-RPE65 vector delivered by ocular subretinal injection. Mol Ther 2006;13:1074–84. 10.1016/j.ymthe.2006.03.005. [DOI] [PubMed] [Google Scholar]
 - 31. Narfstrom K, Katz M, Bragadottir Ret al. Assessment of structure and function over a 3-year period after gene transfer in RPE65-/- dogs. Doc Ophthalmol 2005;111:39–48. 10.1007/s10633-005-3159-0. [DOI] [PubMed] [Google Scholar]
 - 32. Acland GM, Aguirre GD, Bennett Jet 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;12:1072–82. 10.1016/j.ymthe.2005.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 33. Narfstrom K, Bragadóttir R, Redmond TMet al. Functional and structural evaluation after AAV.RPE65 gene transfer in the canine model of Leber’s congenital amaurosis. Adv Exp Med Biol 2003;533:423–30. [DOI] [PubMed] [Google Scholar]
 - 34. Aguirre GK, Komáromy AM, Cideciyan AVet al. Canine and human visual cortex intact and responsive despite early retinal blindness from RPE65 mutation. PLoS Med 2007;4:e230. 10.1371/journal.pmed.0040230. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 35. Li X, Li W, Dai Xet al. Gene therapy rescues cone structure and function in the 3-month-old rd12 mouse: a model for midcourse RPE65 leber congenital amaurosis. Invest Ophthalmol Vis Sci 2011;52:7–15. 10.1167/iovs.10-6138. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 36. Jacobson SG, Aleman TS, Cideciyan AVet al. Identifying photoreceptors in blind eyes caused by RPE65 mutations: Prerequisite for human gene therapy success. Proc Natl Acad Sci USA 2005;102:6177–82. 10.1073/pnas.0500646102. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 37. Hauswirth WW, Aleman TS, Kaushal Set 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;19:979–90. 10.1089/hum.2008.107. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 38. Bainbridge JWB, Smith AJ, Barker SSet al. Effect of gene therapy on visual function in Leber’s congenital amaurosis. N Engl J Med 2008;358:2231–9. 10.1056/NEJMoa0802268. [DOI] [PubMed] [Google Scholar]
 - 39. Maguire AM, Simonelli F, Pierce EAet al. Safety and efficacy of gene transfer for Leber’s congenital amaurosis. N Engl J Med 2008;358:2240–8. 10.1056/NEJMoa0802315. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 40. Cideciyan AV, Aleman TS, Boye SLet al. Human gene therapy for RPE65 isomerase deficiency activates the retinoid cycle of vision but with slow rod kinetics. Proc Nat Acad Sci USA 2008;105:15112–17. 10.1073/pnas.0807027105. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 41. Russell S, Bennett J, Wellman JAet al. Efficacy and safety of voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65-mediated inherited retinal dystrophy: a randomised, controlled, open-label, phase 3 trial. Lancet 2017;390:849–60. 10.1016/S0140-6736(17)31868-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 42. Le Meur G, Lebranchu P, Billaud Fet al. Safety and Long-Term Efficacy of AAV4 Gene Therapy in Patients with RPE65 Leber Congenital Amaurosis. Mol Ther 2018;26:256–68. 10.1016/j.ymthe.2017.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 43. Weleber RG, Pennesi ME, Wilson DJet al. Results at 2 Years after Gene Therapy for RPE65-Deficient Leber Congenital Amaurosis and Severe Early-Childhood-Onset Retinal Dystrophy. Ophthalmology 2016;123:1606–20. 10.1016/j.ophtha.2016.03.003. [DOI] [PubMed] [Google Scholar]
 - 44. Jacobson SG, Cideciyan AV, Ratnakaram Ret al. Gene therapy for leber congenital amaurosis caused by RPE65 mutations: safety and efficacy in 15 children and adults followed up to 3 years. Arch Ophthalmol 2012;130:9–24. 10.1001/archophthalmol.2011.298. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 45. Jacobson SG, Cideciyan AV, Roman AJet al. Improvement and decline in vision with gene therapy in childhood blindness. N Engl J Med 2015;372:1920–6. 10.1056/NEJMoa1412965. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 46. Cideciyan AV, Hauswirth WW, Aleman TSet al. Human RPE65 gene therapy for Leber congenital amaurosis: persistence of early visual improvements and safety at 1 year. Hum Gene Ther 2009;20:999–1004. 10.1089/hum.2009.086. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 47. Cideciyan AV, Aguirre GK, Jacobson SGet al. Pseudo-fovea formation after gene therapy for RPE65-LCA. Invest Ophthalmol Vis Sci 2014;56:526–37. 10.1167/iovs.14-15895. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 48. Hartong DT, Berson EL, Dryja TP. Retinitis pigmentosa. Lancet 2006;368:1795–1809. 10.1016/S0140-6736(06)69740-7. [DOI] [PubMed] [Google Scholar]
 - 49. Parmeggiani F. Clinics, epidemiology and genetics of retinitis pigmentosa. Curr Genomics 2011;12:236–7. 10.2174/138920211795860080. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 50. Sullivan LS, Bowne SJ, Birch DGet al. Prevalence of disease-causing mutations in families with autosomal dominant retinitis pigmentosa: A screen of known genes in 200 families. Invest Ophthalmol Vis Sci 2006;47:3052–64. 10.1167/iovs.05-1443. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 51. Evans DR, Green JS, Johnson GJet al. Novel 25 kb Deletion of MERTK Causes Retinitis Pigmentosa With Severe Progression. Invest Ophthalmol Vis Sci 2017;58:1736–42. 10.1167/iovs.16-20864. [DOI] [PubMed] [Google Scholar]
 - 52. Shahzadi A, Riazuddin SA, Ali Set al. Nonsense mutation in MERTK causes autosomal recessive retinitis pigmentosa in a consanguineous Pakistani family. Br J Ophthalmol 2010;94:1094–9. 10.1136/bjo.2009.171892. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 53. Mackay DS, Henderson RH, Sergouniotis PIet al. Novel mutations in MERTK associated with childhood onset rod-cone dystrophy. Mol Vis 2010;16:369–77. [PMC free article] [PubMed] [Google Scholar]
 - 54. Ostergaard E, Duno M, Batbayli Met al. A novel MERTK deletion is a common founder mutation in the Faroe Islands and is responsible for a high proportion of retinitis pigmentosa cases. Mol Vis 2011;17:1485–92. [PMC free article] [PubMed] [Google Scholar]
 - 55. Jinda W, Poungvarin N, Taylor TDet al. A novel start codon mutation of the MERTK gene in a patient with retinitis pigmentosa. Mol Vis 2016;22:342–51. [PMC free article] [PubMed] [Google Scholar]
 - 56. Gal A, Li Y, Thompson DAet al. Mutations in MERTK, the human orthologue of the RCS rat retinal dystrophy gene, cause retinitis pigmentosa. Nat Genet 2000;26:270–1. 10.1038/81555. [DOI] [PubMed] [Google Scholar]
 - 57. LaVail MM, Sidman M, Rausin Ret al. Discrimination of light intensity by rats with inherited retinal degeneration: a behavioral and cytological study. Vision Res 1974;14:693–702. [DOI] [PubMed] [Google Scholar]
 - 58. LaVail MM, Battelle BA. Influence of eye pigmentation and light deprivation on inherited retinal dystrophy in the rat. Exp Eye Res 1975;21:167–92. [DOI] [PubMed] [Google Scholar]
 - 59. Ben-Arie-Weintrob Y, Berson EL, Dryja TP. Histopathologic-genotypic correlations in retinitis pigmentosa and allied diseases. Ophthalmic Genet 2005;26:91–100. 10.1080/13816810590968032. [DOI] [PubMed] [Google Scholar]
 - 60. Tschernutter M, Schlichtenbrede FC, Howe Set al. Long-term preservation of retinal function in the RCS rat model of retinitis pigmentosa following lentivirus-mediated gene therapy. Gene Ther 2005;12:694–701. 10.1038/sj.gt.3302460. [DOI] [PubMed] [Google Scholar]
 - 61. Deng WT, Dinculescu A, Li Qet al. Tyrosine-mutant AAV8 delivery of human MERTK provides long-term retinal preservation in RCS rats. Invest Ophthalmol Vis Sci 2012;53:1895–1904. 10.1167/iovs.11-8831. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 62. Buch PK, MacLaren RE, Durán Yet al. In contrast to AAV-mediated Cntf expression, AAV-mediated Gdnf expression enhances gene replacement therapy in rodent models of retinal degeneration. Mol Ther 2006;14:700–9. 10.1016/j.ymthe.2006.05.019. [DOI] [PubMed] [Google Scholar]
 - 63. Suzuki K, Tsunekawa Y, Hernandez-Benitez Ret al. In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration. Nature 2016;540:144–9. 10.1038/nature20565. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 64. Ghazi NG, Abboud EB, Nowilaty SRet 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;135:327–43. 10.1007/s00439-016-1637-y. [DOI] [PubMed] [Google Scholar]
 - 65. Grondahl J. Estimation of Prognosis and Prevalence of Retinitis-Pigmentosa and Usher Syndrome in Norway. Clin Genet 1987;31:255–64. [DOI] [PubMed] [Google Scholar]
 - 66. Vernon M. Ushers Syndrome - Deafness and Progressive Blindness - Clinical Cases, Prevention, Theory and Literature Survey. J Chron Dis 1969;22:133–51. 10.1016/0021-9681(69)90055-1. [DOI] [PubMed] [Google Scholar]
 - 67. Boughman JA, Vernon M, Shaver KA. Usher syndrome: definition and estimate of prevalence from two high-risk populations. J Chronic Dis 1983;36:595–603. [DOI] [PubMed] [Google Scholar]
 - 68. Millan JM, Aller E, Jaijo Tet al. An update on the genetics of usher syndrome. J Ophthalmol 2011;2011:417217. 10.1155/2011/417217. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 69. Ferrari S, Di Iorio E, Barbaro Vet al. Retinitis pigmentosa: genes and disease mechanisms. Curr Genomics 2011;12:238–49. 10.2174/138920211795860107. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 70. Petit C. Usher syndrome: from genetics to pathogenesis. Annu Rev Genomics Hum Genet 2001;2:271–97. 10.1146/annurev.genom.2.1.271. [DOI] [PubMed] [Google Scholar]
 - 71. Zina ZB, Masmoudi S, Ayadi Het al. From DFNB2 to Usher syndrome: Variable expressivity of the same disease. Am J Med Genet 2001;101:181–3. [DOI] [PubMed] [Google Scholar]
 - 72. Smith RJ, Berlin CI, Hejtmancik JFet al. Clinical diagnosis of the Usher syndromes. Usher Syndrome Consortium. Am J Med Genet 1994;50:32–8. 10.1002/ajmg.1320500107. [DOI] [PubMed] [Google Scholar]
 - 73. Williams DS. Usher syndrome: Animal models, retinal function of Usher proteins, and prospects for gene therapy. Vision Res 2008;48:433–41. 10.1016/j.visres.2007.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 74. Riazuddin S, Belyantseva IA, Giese APJet al. Alterations of the CIB2 calcium- and integrin-binding protein cause Usher syndrome type 1J and nonsyndromic deafness DFNB48. Nat Genet 2012;44:1265–71. 10.1038/ng.2426. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 75. Sahly I, Dufour E, Schietroma Cet al. Localization of Usher 1 proteins to the photoreceptor calyceal processes, which are absent from mice. J Cell Biol 2012;199:381–99. 10.1083/jcb.201202012. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 76. Well D, Blanchard S, Kaplan Jet al. Defective Myosin Viia Gene Responsible for Usher Syndrome Type 1b. Nature 1995;374:60–1. 10.1038/374060a0. [DOI] [PubMed] [Google Scholar]
 - 77. Smith RJH, Berlin CI, Hejtmancik JFet al. Clinical-Diagnosis of the Usher Syndromes. Am J Med Genet 1994;50:32–8. 10.1002/ajmg.1320500107. [DOI] [PubMed] [Google Scholar]
 - 78. Liu XR, Vansant G, Udovichenko IPet al. Myosin VIIa, the product of the Usher 1B syndrome gene, is concentrated in the connecting cilia of photoreceptor cells. Cell Motil Cytoskeleton 1997;37:240–52. doi:10.1002/(Sici)1097-0169(1997)37:3 240::Aid-Cm6 3.3.Co;2-2. [DOI] [PubMed] [Google Scholar]
 - 79. Hasson T, Heintzelman MB, Santos-Sacchi Jet al. Expression in cochlea and retina of myosin VIIa, the gene product defective in Usher syndrome type 1B. Proc Nat Acad Sci USA 1995;92:9815–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 80. Wolfrum U. The cellular function of the Usher gene product myosin VIIa is specified by its ligands. Adv Exp Med Biol 2003;533:133–42. [DOI] [PubMed] [Google Scholar]
 - 81. Weil D, Levy G, Sahly Iet al. Human myosin VIIA responsible for the Usher 1B syndrome: a predicted membrane-associated motor protein expressed in developing sensory epithelia. Proc Nat Acad Sci USA 1996;93:3232–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 82. Lillo C, Kitamoto J, Liu Xet al. Mouse models for Usher syndrome 1B. Adv Exp Med Biol 2003;533:143–50. [DOI] [PubMed] [Google Scholar]
 - 83. Hasson T, Walsh J, Cable Jet al. Effects of shaker-1 mutations on myosin-VIIa protein and mRNA expression. Cell Motil Cytoskeleton 1997;37:127–38. 10.1002/(Sici)1097-0169(1997)37:2127::Aid-Cm53.0.Co;2-5. [DOI] [PubMed] [Google Scholar]
 - 84. Libby RT, Steel KP. Electroretinographic anomalies in mice with mutations in Myo7a, the gene involved. in human Usher syndrome type 1B. Invest Ophthalmol Vis Sci 2001;42:770–8. [PubMed] [Google Scholar]
 - 85. Kong L, Li F, Soleman CEet al. Bright cyclic light accelerates photoreceptor cell degeneration in tubby mice. Neurobiol Dis 2006;21:468–77. 10.1016/j.nbd.2005.08.017. [DOI] [PubMed] [Google Scholar]
 - 86. Peng YW, Zallocchi M, Wang WMet al. Moderate Light-Induced Degeneration of Rod Photoreceptors with Delayed Transducin Translocation in shaker1 Mice. Invest Ophthalmol Vis Sci 2011;52:6421–7. 10.1167/iovs.10-6557. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 87. Tian M, Wang W, Delimont Det al. Photoreceptors in whirler mice show defective transducin translocation and are susceptible to short-term light/dark changes-induced degeneration. Exp Eye Res 2014;118:145–53. 10.1016/j.exer.2013.10.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 88. Liu XR, Ondek B, Williams DS. Mutant myosin VIIa causes defective melanosome distribution in the RPE of shaker-1 mice. Nat Genet 1998;19:117–8. 10.1038/470. [DOI] [PubMed] [Google Scholar]
 - 89. Liu XR, Udovichenko IP, Brown SDMet al. Myosin VIIa participates in opsin transport through the photoreceptor cilium. J Neurosci 1999;19:6267–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 90. Gibbs D, Azarian SM, Lillo Cet al. Role of myosin VIIa and Rab27a in the motility and localization of RPE melanosomes. J Cell Sci 2004;117:6473–83. 10.1242/jcs.01580. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 91. Hashimoto T, Gibbs D, Lillo Cet al. Lentiviral gene replacement therapy of retinas in a mouse model for Usher syndrome type 1B. Gene Ther 2007;14:584–94. 10.1038/sj.gt.3302897. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 92. Gibbs D, Diemer T, Khanobdee Ket al. Function of MYO7A in the Human RPE and the Validity of Shaker1 Mice as a Model for Usher Syndrome 1B. Invest Ophthalmol Vis Sci 2010;51:1130–5. 10.1167/iovs.09-4032. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 93. Zallocchi M, Binley K, Lad Yet al. EIAV-based retinal gene therapy in the shaker1 mouse model for usher syndrome type 1B: development of UshStat. PLoS One 2014;9:e94272. 10.1371/journal.pone.0094272. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 94. Ikeda Y, Yonemitsu Y, Miyazaki Met al. Stable Retinal Gene Expression in Nonhuman Primates via Subretinal Injection of SIVagm-Based Lentiviral Vectors. Hum Gene Ther 2009;20:573–9. 10.1089/hum.2009.009. [DOI] [PubMed] [Google Scholar]
 - 95. Bainbridge JWB, Stephens C, Parsley Ket al. In vivo gene transfer to the mouse eye using an HIV-based lentiviral vector; efficient long-term transduction of corneal endothelium and retinal pigment epithelium. Gene Ther 2001;8:1665–8. 10.1038/sj.gt.3301574. [DOI] [PubMed] [Google Scholar]
 - 96. Trapani I, Colella P, Sommella Aet al. Effective delivery of large genes to the retina by dual AAV vectors. EMBO Mol Med 2014;6:194–211. 10.1002/emmm.201302948. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 97. Yan Z, Zak R, Zhang Yet al. Inverted terminal repeat sequences are important for intermolecular recombination and circularization of adeno-associated virus genomes. J Virol 2005;79:364–79. 10.1128/JVI.79.1.364-379.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 98. Lopes VS, Boye SE, Louie CMet al. Retinal gene therapy with a large MYO7A cDNA using adeno-associated virus. Gene Ther 2013;20:824–33. 10.1038/gt.2013.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 99. Dong BA, Nakai H, Xiao WD. Characterization of Genome Integrity for Oversized Recombinant AAV Vector. Mol Ther 2010;18:87–92. 10.1038/mt.2009.258. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 100. Dyka FM, Boye SL, Chiodo VAet al. Dual Adeno-Associated Virus Vectors Result in Efficient In Vitro and In Vivo Expression of an Oversized Gene, MYO7A. Hum Gene Ther Methods 2014;25:166–77. 10.1089/hgtb.2013.212. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 101. Colella P, Trapani I, Cesi Get al. Efficient gene delivery to the cone-enriched pig retina by dual AAV vectors. Gene Ther 2014;21:450–6. 10.1038/gt.2014.8. [DOI] [PubMed] [Google Scholar]
 - 102. Lambertus S, van Huet RAC, Bax NMet al. Early-Onset Stargardt Disease Phenotypic and Genotypic Characteristics. Ophthalmology 2015;122:335–44. 10.1016/j.ophtha.2014.08.032. [DOI] [PubMed] [Google Scholar]
 - 103. Allikmets R, Singh N, Sun Het al. A photoreceptor cell-specific ATP-binding transporter gene (ABCR) is mutated in recessive Stargardt macular dystrophy. Nat Genet 1997;15:236–46. 10.1038/ng0397-236. [DOI] [PubMed] [Google Scholar]
 - 104. Molday RS, Beharry S, Ahn Jet al. Binding of N-retinylidene-PE to ABCA4 and a model for its transport across membranes. Adv Exp Med Biol 2006;572:465–70. [DOI] [PubMed] [Google Scholar]
 - 105. Wu YL, Li J, Yao K. Review: Structures and biogenetic analysis of lipofuscin bis-retinoids. J Zhejiang Univ Sci B 2013;14:763–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 106. Molday RS. Insights into the Molecular Properties of ABCA4 and Its Role in the Visual Cycle and Stargardt Disease. Prog Mol Biol Transl Sci 2015;134:415–31. 10.1016/bs.pmbts.2015.06.008. [DOI] [PubMed] [Google Scholar]
 - 107. Quazi F, Lenevich S, Molday RS. ABCA4 is an N-retinylidene-phosphatidylethanolamine and phosphatidylethanolamine importer. Nat Commun 2012;3:925. 10.1038/ncomms1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 108. Cella W, Greenstein VC, Zernant-Rajang Jet al. G1961E mutant allele in the Stargardt disease gene ABCA4 causes bull’s eye maculopathy. Exp Eye Res 2009;89:16–24. 10.1016/j.exer.2009.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 109. Mata NL, Tzekov RT, Liu Xet al. Delayed dark-adaptation and lipofuscin accumulation in abcrb/2 mice: Implications for involvement of ABCR in age-related macular degeneration. Invest Ophthalmol Vis Sci 2001;42:1685–90. [PubMed] [Google Scholar]
 - 110. Allocca M, Doria M, Petrillo Met al. Serotype-dependent packaging of large genes in adeno-associated viral vectors results in effective gene delivery in mice. J Clin Invest 2008;118:1955–64. 10.1172/Jci34316. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 111. Kong J, Kim SR, Binley Ket al. Correction of the disease phenotype in the mouse model of Stargardt disease by lentiviral gene therapy. Gene Ther 2008;15:1311–20. 10.1038/gt.2008.78. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 112. Binley K, Widdowson P, Loader Jet al. Transduction of photoreceptors with equine infectious anemia virus lentiviral vectors: Safety and biodistribution of StarGen for Stargardt disease. Invest Ophthalmol Vis Sci 2013;54:4061–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 113. Sorsby A, Franceschetti A, Joseph Ret al. Choroideremia - Clinical and Genetic Aspects. Br J Ophthalmol 1952;36:547–81. 10.1136/bjo.36.10.547. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 114. Seabra MC. New insights into the pathogenesis of choroideremia: A tale of two REPs. Ophthalmic Genet 1996;17:43–6. 10.3109/13816819609057869. [DOI] [PubMed] [Google Scholar]
 - 115. Karna J. Choroideremia - a Clinical and Genetic-Study of 84 Finnish Patients and 126 Female Carriers. Acta Ophthalmol 1986;64:1–68. [PubMed] [Google Scholar]
 - 116. Dimopoulos IS, Chan S, MacLaren REet al. Pathogenic mechanisms and the prospect of gene therapy for choroideremia. Expert Opin Orphan Drugs 2015;3:787–98. 10.1517/21678707.2015.1046434. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 117. Zinkernagel MS, MacLaren RE. Recent advances and future prospects in choroideremia. Clin Ophthalmol 2015;9:2195–2200. 10.2147/Opth.S65732. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 118. Perez-Cano HJ, Garnica-hayashi RE, Zenteno JC. CHM Gene Molecular Analysis and X-Chromosome Inactivation Pattern Determination in Two Families With Choroideremia. Am J Med Genet A 2009;149a:2134–40. 10.1002/ajmg.a.32727. [DOI] [PubMed] [Google Scholar]
 - 119. Potter MJ, Wong E, Szabo SMet al. Clinical findings in a carrier of a new mutation in the choroideremia gene. Ophthalmology 2004;111:1905–9. 10.1016/j.ophtha.2004.04.028. [DOI] [PubMed] [Google Scholar]
 - 120. Pereira-Leal JB, Seabra MC. The mammalian Rab family of small GTPases: Definition of family and subfamily sequence motifs suggests a mechanism for functional specificity in the Ras superfamily. J Mol Biol 2000;301:1077–87. 10.1006/jmbi.2000.4010. [DOI] [PubMed] [Google Scholar]
 - 121. Corbeel L, Freson K. Rab proteins and Rab-associated proteins: major actors in the mechanism of protein-trafficking disorders. Eur J Pediatr 2008;167:723–9. 10.1007/s00431-008-0740-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 122. Alory C, Balch WE. Organization of the Rab-GDI/CHM superfamily: The functional basis for choroideremia disease. Traffic 2001;2:532–43. 10.1034/j.1600-0854.2001.20803.x. [DOI] [PubMed] [Google Scholar]
 - 123. Strunnikova N, Zein WM, Silvin Cet al. Serum Biomarkers and Trafficking Defects in Peripheral Tissues Reflect the Severity of Retinopathy in Three Brothers Affected by Choroideremia. Retinal Degenerative Diseases 2012;723:381–7. 10.1007/978-1-4614-0631-0_49. [DOI] [PubMed] [Google Scholar]
 - 124. Tolmachova T, Anders R, Abrink Met al. Independent degeneration of photoreceptors and retinal pigment epithelium in conditional knockout mouse models of choroideremia. J Clin Invest 2006;116:386–94. 10.1172/Jci26617. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 125. Tolmachova T, Wavre-Shapton ST, Barnard ARet al. Retinal Pigment Epithelium Defects Accelerate Photoreceptor Degeneration in Cell Type-Specific Knockout Mouse Models of Choroideremia. Invest Ophthalmol Vis Sci 2010;51:4913–20. 10.1167/iovs.09-4892. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 126. Arland V, Barral DC, Zeng Yet al. Gene therapy for choroideremia: in vitro rescue mediated by recombinant adenovirus. Vision Res 2003;43:919–26. 10.1016/S0042-6989(02)00389-9. [DOI] [PubMed] [Google Scholar]
 - 127. Vasireddy V, Mills JA, Gaddameedi Ret al. AAV-Mediated Gene Therapy for Choroideremia: Preclinical Studies in Personalized Models. PLoS One 2013;8:e61396. 10.1371/journal.pone.0061396. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 128. Tolmachova T, Tolmachov OE, Barnard ARet 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 2013;91:825–37. 10.1007/s00109-013-1006-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 129. Black A, Vasireddy V, Chung DCet al. Adeno-associated virus 8-mediated gene therapy for choroideremia: preclinical studies in in vitro and in vivo models. J Gene Med 2014;16:122–30. 10.1002/jgm.2768. [DOI] [PubMed] [Google Scholar]
 - 130. MacLaren RE, Groppe M, Barnard ARet al. Retinal gene therapy in patients with choroideremia: initial findings from a phase 1/2 clinical trial. Lancet 2014;383:1129–37. 10.1016/S0140-6736(13)62117-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 131. Agarwal A, Rhoades WR, Hanout Met al. Management of neovascular age-related macular degeneration: current state-of-the-art care for optimizing visual outcomes and therapies in development. Clin Ophthalmol 2015;9:1001–15. 10.2147/OPTH.S74959. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 132. Brown DM, Regillo CD. Anti-VEGF agents in the treatment of neovascular age-related macular degeneration: applying clinical trial results to the treatment of everyday patients. Am J Ophthalmol 2007;144:627–37. 10.1016/j.ajo.2007.06.039. [DOI] [PubMed] [Google Scholar]
 - 133. Aiello LP, Pierce EA, Foley EDet al. Suppression of retinal neovascularization in vivo by inhibition of vascular endothelial growth factor (VEGF) using soluble VEGF-receptor chimeric proteins. Proc Natl Acad Sci USA 1995;92:10457–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 134. Kvanta A, Algvere PV, Berglin Let al. Subfoveal fibrovascular membranes in age-related macular degeneration express vascular endothelial growth factor. Invest Ophthalmol Vis Sci 1996;37:1929–34. [PubMed] [Google Scholar]
 - 135. Adamis AP, Miller JW, Bernal MTet al. Increased vascular endothelial growth factor levels in the vitreous of eyes with proliferative diabetic retinopathy. Am J Ophthalmol 1994;118:445–50. [DOI] [PubMed] [Google Scholar]
 - 136. de Vries C, Escobedo JA, Ueno Het al. The fms-like tyrosine kinase, a receptor for vascular endothelial growth factor. Science 1992;255:989–91. [DOI] [PubMed] [Google Scholar]
 - 137. Shibuya M, Yamaguchi S, Yamane Aet al. Nucleotide sequence and expression of a novel human receptor-type tyrosine kinase gene (flt) closely related to the fms family. Oncogene 1990;5:519–24. [PubMed] [Google Scholar]
 - 138. Lai CM, Shen WY, Brankov Met al. Long-term evaluation of AAV-mediated sFlt-1 gene therapy for ocular neovascularization in mice and monkeys. Mol Ther 2005;12:659–68. 10.1016/j.ymthe.2005.04.022. [DOI] [PubMed] [Google Scholar]
 - 139. Lai CM, Estcourt MJ, Himbeck RPet al. Preclinical safety evaluation of subretinal AAV2.sFlt-1 in non-human primates. Gene Ther 2012;19:999–1009. 10.1038/gt.2011.169. [DOI] [PubMed] [Google Scholar]
 - 140. Lai YKY, Shen WY, Brankov Met al. Potential long-term inhibition of ocular neovascularisation by recombinant adeno-associated virus-mediated secretion gene therapy. Gene Ther 2002;9:804–13. 10.1038/sj.gt.3301695. [DOI] [PubMed] [Google Scholar]
 - 141. Bainbridge JW, Mistry A, De Alwis Met al. Inhibition of retinal neovascularisation by gene transfer of soluble VEGF receptor sFlt-1. Gene Ther 2002;9:320–6. 10.1038/sj.gt.3301680. [DOI] [PubMed] [Google Scholar]
 - 142. Constable IJ, Pierce CM, Lai CMet al. Phase 2a Randomized Clinical Trial: Safety and Post Hoc Analysis of Subretinal rAAV.sFLT-1 for Wet Age-related Macular Degeneration. EBioMedicine 2016;14:168–75. 10.1016/j.ebiom.2016.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 143. Rakoczy EP, Lai CM, Magno ALet al. Gene therapy with recombinant adeno-associated vectors for neovascular age-related macular degeneration: 1 year follow-up of a phase 1 randomised clinical trial. Lancet 2015;386:2395–2403. 10.1016/S0140-6736(15)00345-1. [DOI] [PubMed] [Google Scholar]
 - 144. Heier JS, Kherani S, Desai Set al. Intravitreous injection of AAV2-sFLT01 in patients with advanced neovascular age-related macular degeneration: a phase 1, open-label trial. Lancet 2017;390:50–61. 10.1016/S0140-6736(17)30979-0. [DOI] [PubMed] [Google Scholar]
 - 145. Dawson DW, Volpert OV, Gillis Pet al. Pigment epithelium-derived factor: a potent inhibitor of angiogenesis. Science 1999;285:245–8. [DOI] [PubMed] [Google Scholar]
 - 146. O’Reilly MS, Boehm T, Shing Yet al. Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell 1997;88:277–85. [DOI] [PubMed] [Google Scholar]
 - 147. O’Reilly MS, Boehm T, Shing Yet al. Angiostatin: a novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cell 1994;79:315–28. [DOI] [PubMed] [Google Scholar]
 - 148. Mori K, Gehlbach P, Yamamoto Set al. AAV-mediated gene transfer of pigment epithelium-derived factor inhibits choroidal neovascularization. Invest Ophthalmol Vis Sci 2002;43:1994–2000. [PubMed] [Google Scholar]
 - 149. Mori K, Ando A, Gehlbach Pet al. Inhibition of choroidal neovascularization by intravenous injection of adenoviral vectors expressing secretable endostatin. Am J Pathol 2001;159:313–20. 10.1016/S0002-9440(10)61697-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 150. Lai CC, Wu WC, Chen SLet al. Suppression of choroidal neovascularization by adeno-associated virus vector expressing angiostatin. Invest Ophthalmol Vis Sci 2001;42:2401–7. [PubMed] [Google Scholar]
 - 151. Rasmussen H, Chu KW, Campochiaro Pet al. Clinical protocol. An open-label, phase I, single administration, dose-escalation study of ADGVPEDF.11D (ADPEDF) in neovascular age-related macular degeneration (AMD). Hum Gene Ther 2001;12:2029–32. [PubMed] [Google Scholar]
 - 152. Campochiaro PA, Nguyen QD, Shah SMet al. Adenoviral vector-delivered pigment epithelium-derived factor for neovascular age-related macular degeneration: results of a phase I clinical trial. Hum Gene Ther 2006;17:167–76. 10.1089/hum.2006.17.167. [DOI] [PubMed] [Google Scholar]
 - 153. Kachi S, Binley K, Yokoi Ket al. Equine infectious anemia viral vector-mediated codelivery of endostatin and angiostatin driven by retinal pigmented epithelium-specific VMD2 promoter inhibits choroidal neovascularization. Hum Gene Ther 2009;20:31–9. 10.1089/hum.2008.046. [DOI] [PubMed] [Google Scholar]
 - 154. Campochiaro PA, Lauer AK, Sohn EHet al. Lentiviral Vector Gene Transfer of Endostatin/Angiostatin for Macular Degeneration (GEM) Study. Hum Gene Ther 2017;28:99–111. 10.1089/hum.2016.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 155. Sharpe LT, Gegenfurtner KR. Color vision: from genes to preception. Cambridge: Cambridge University Press, 2001. [Google Scholar]
 - 156. Thiadens AA, Somervuo V, van den Born LIet al. Progressive loss of cones in achromatopsia: an imaging study using spectral-domain optical coherence tomography. Invest Ophthalmol Vis Sci 2010;51:5952–7. 10.1167/iovs.10-5680. [DOI] [PubMed] [Google Scholar]
 - 157. Chiang WC, Chan P, Wissinger Bet al. Achromatopsia mutations target sequential steps of ATF6 activation. Proc Natl Acad Sci USA 2017;114:400–5. 10.1073/pnas.1606387114. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 158. Thiadens AA, den Hollander AI, Roosing Set al. Homozygosity mapping reveals PDE6C mutations in patients with early-onset cone photoreceptor disorders. Am J Hum Genet 2009;85:240–7. 10.1016/j.ajhg.2009.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 159. Kohl S, Baumann B, Rosenberg Tet al. Mutations in the cone photoreceptor G-protein alpha-subunit gene GNAT2 in patients with achromatopsia. Am J Hum Genet 2002;71:422–5. 10.1086/341835. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 160. Alexander JJ, Umino Y, Everhart Det al. Restoration of cone vision in a mouse model of achromatopsia. Nat Med 2007;13:685–7. 10.1038/nm1596. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 161. Chang B, Dacey MS, Hawes NLet al. Cone photoreceptor function loss-3, a novel mouse model of achromatopsia due to a mutation in Gnat2. Invest Ophthalmol Vis Sci 2006;47:5017–21. 10.1167/iovs.05-1468. [DOI] [PubMed] [Google Scholar]
 - 162. Pang J, Deng WT, Dai Xet al. AAV-mediated cone rescue in a naturally occurring mouse model of CNGA3-achromatopsia. PLoS One 2012;7:e35250. 10.1371/journal.pone.0035250. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 163. Carvalho LS, Xu J, Pearson RAet 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;20:3161–75. 10.1093/hmg/ddr218. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 164. Muhlfriedel R, Tanimoto N, Schön Cet al. AAV-Mediated Gene Supplementation Therapy in Achromatopsia Type 2: Preclinical Data on Therapeutic Time Window and Long-Term Effects. Front Neurosci 2017;11:292. 10.3389/fnins.2017.00292. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 165. Komaromy AM, Alexander JJ, Rowlan JSet al. Gene therapy rescues cone function in congenital achromatopsia. Hum Mol Genet 2010;19:2581–93. 10.1093/hmg/ddq136. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 166. Banin E, Gootwine E, Obolensky Aet al. Gene Augmentation Therapy Restores Retinal Function and Visual Behavior in a Sheep Model of CNGA3 Achromatopsia. Mol Ther 2015;23:1423–33. 10.1038/mt.2015.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 167. Ye GJ, Budzynski E, Sonnentag Pet 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 2016;27:37–48. 10.1089/humc.2015.164. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 168. Ye GJ, Budzynski E, Sonnentag Pet al. Cone-Specific Promoters for Gene Therapy of Achromatopsia and Other Retinal Diseases. Hum Gene Ther 2016;27:72–82. 10.1089/hum.2015.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 169. George ND, Yates JR, Moore AT. X linked retinoschisis. Br J Ophthalmol 1995;79:697–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 170. Peachey NS, Fishman GA, Derlacki DJet al. Psychophysical and Electroretinographic Findings in X-Linked Juvenile Retinoschisis. Arch Ophthalmol 1987;105:513–6. [DOI] [PubMed] [Google Scholar]
 - 171. Minami Y, Ishiko S, Takai Yet al. Retinal changes in juvenile X linked retinoschisis using three dimensional optical coherence tomography. Br J Ophthalmol 2005;89:1663–4. 10.1136/bjo.2005.075648. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 172. Prenner JL, Antonio Capone JR, Ciaccia Set al. Congenital X-linked retinoschisis classification system. Retina 2006;26:S61–64. 10.1097/01.iae.0000244290.09499.c1. [DOI] [PubMed] [Google Scholar]
 - 173. Roesch MT, Ewing CC, Gibson AEet al. The natural history of X-linked retinoschisis. Can J Ophthalmol 1998;33:149–58. [PubMed] [Google Scholar]
 - 174. Baumgartner S, Hofmann K, Chiquet-Ehrismann Ret al. The discoidin domain family revisited: New members from prokaryotes and a homology-based fold prediction. Protein Sci 1998;7:1626–31. 10.1002/pro.5560070717. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 175. Wu WWH, Wong JP, Kast Jet al. RS1, a discoidin domain-containing retinal cell adhesion protein associated with X-linked retinoschisis, exists as a novel disulfide-linked octamer. J Biol Chem 2005;280:10721–30. 10.1074/jbc.M413117200. [DOI] [PubMed] [Google Scholar]
 - 176. Takada Y, Fariss RN, Muller Met al. Retinoschisin expression and localization in rodent and human pineal and consequences of mouse RS1 gene knockout. Mol Vis 2006;12:1108–16. [PubMed] [Google Scholar]
 - 177. Sauer CG, Gehrig A, Warneke-Wittstock Ret al. Positional cloning of the gene associated with X-linked juvenile retinoschisis. Nat Genet 1997;17:164–70. 10.1038/ng1097-164. [DOI] [PubMed] [Google Scholar]
 - 178. Zeng Y, Takada Y, Kjellstrom Set al. RS-1 Gene Delivery to an Adult Rs1h Knockout Mouse Model Restores ERG b-Wave with Reversal of the Electronegative Waveform of X-Linked Retinoschisis. Invest Ophthalmol Vis Sci 2004;45:3279–85. 10.1167/iovs.04-0576. [DOI] [PubMed] [Google Scholar]
 - 179. Min SH, Molday LL, Seeliger MWet al. Prolonged recovery of retinal structure/function after gene therapy in an Rs1h-deficient mouse model of x-linked juvenile retinoschisis. Mol Ther 2005;12:644–51. 10.1016/j.ymthe.2005.06.002. [DOI] [PubMed] [Google Scholar]
 - 180. Park TK, Wu Z, Kjellstrom Set al. Intravitreal delivery of AAV8 retinoschisin results in cell type-specific gene expression and retinal rescue in the Rs1-KO mouse. Gene Ther 2009;16:916–26. 10.1038/gt.2009.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 181. Janssen A, Min SH, Molday LLet al. Effect of late-stage therapy on disease progression in AAV-mediated rescue of photoreceptor cells in the retinoschisin-deficient mouse. Mol Ther 2008;16:1010–7. 10.1038/mt.2008.57. [DOI] [PubMed] [Google Scholar]
 - 182. Yanger K, Zong Y, Maggs LRet al. Robust cellular reprogramming occurs spontaneously during liver regeneration. Genes Dev 2013;27:719–24. 10.1101/gad.207803.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 183. Zhang RL, Han P, Yang Het al. In vivo cardiac reprogramming contributes to zebrafish heart regeneration. Nature 2013;498:497–501. 10.1038/nature12322. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 184. Lin HT, Ouyang H, Zhu Jet al. Lens regeneration using endogenous stem cells with gain of visual function. Nature 2016;531:323–8. 10.1038/nature17181. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 185. Song GQ, Pacher M, Balakrishnan Aet al. Direct Reprogramming of Hepatic Myofibroblasts into Hepatocytes In Vivo Attenuates Liver Fibrosis. Cell Stem Cell 2016;18:797–808. 10.1016/j.stem.2016.01.010. [DOI] [PubMed] [Google Scholar]
 - 186. Montana CL, Kolesnikov AV, Shen SQet al. Reprogramming of adult rod photoreceptors prevents retinal degeneration. Proc Natl Acad Sci USA 2013;110:1732–7. 10.1073/pnas.1214387110. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 187. Cheng H, Khanna H, Oh ECTet al. Photoreceptor-specific nuclear receptor NR2E3 functions as a transcriptional activator in rod photoreceptors. Hum Mol Genet 2004;13:1563–75. 10.1093/hmg/ddh173. [DOI] [PubMed] [Google Scholar]
 - 188. Zhu J, Ming C, Fu Xet al. Gene and mutation independent therapy via CRISPR-Cas9 mediated cellular reprogramming in rod photoreceptors. Cell Res 2017;27:830–3. 10.1038/cr.2017.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 189. Yu WH, Mookherjee S, Chaitankar Vet al. Nrl knockdown by AAV-delivered CRISPR/Cas9 prevents retinal degeneration in mice. Nat Commun 2017;8:14716. 10.1038/ncomms14716. [DOI] [PMC free article] [PubMed] [Google Scholar]
 





