Skip to main content
Cold Spring Harbor Perspectives in Medicine logoLink to Cold Spring Harbor Perspectives in Medicine
. 2023 Apr;13(4):a041292. doi: 10.1101/cshperspect.a041292

Therapeutic Gene Editing in Inherited Retinal Disorders

Jinjie Ling 1, Laura A Jenny 2, Ashley Zhou 1, Stephen H Tsang 2,3,4,5,
PMCID: PMC10071418  PMID: 36096547

Abstract

Since the development of CRISPR/Cas9 gene editing in 2012, therapeutic editing research has produced several phase 1-2a trials. Here we provide an overview of the mechanisms and applications of various gene-editing technologies including adeno-associated virus vectors, lentiviruses, CRISPR/Cas9 systems, base and prime editing, antisense oligonucleotides, short-hairpin RNAs, Cas13, and adenosine deaminase acting on RNA for the treatment of various inherited retinal diseases (IRDs). We outline the various stages of clinical trials using these technologies and the impacts they have made in advancing the practice of medicine.


The eye is an extraordinary organ that harbors several characteristics conducive for the implementation of gene therapy. The retinal–blood barrier, which excludes ocular immune cell penetration, and the presence of cytokines and signaling molecules, which promotes regulatory T-cell activation and immunosuppression, confer a unique immune-privileged state to the eye (Medawar 1948; Stein-Streilein and Taylor 2007; Caspi 2010). Anatomically, the eye consists of a limited volume of cells, and the organization of these cells into their functional layers is amenable to the delivery of gene therapy vectors. The bilateral nature of human eyes is of particular interest for clinical trials as it enables one eye to serve as the treatment recipient and the other to serve as the control. Here we provide an overview of the current applications of gene editing in the treatment of retinal diseases and include an update on promising developments in recent clinical trials and cutting-edge technologies.

STRATEGIES IN GENE THERAPY AND EDITING

Adeno-Associated Virus

Historically, the adeno-associated virus (AAV) vector has served as the delivery modality of choice for the treatment of ocular disease as it exhibits low immunogenicity and high transduction efficiency in retinal cells (Timmers et al. 2020). Although AAV-based therapies have found success in the treatment of some retinal diseases (i.e., RPE65-associated Leber congenital amaurosis [LCA] summarized in Table 1; Bainbridge et al. 2008, 2015; Maguire et al. 2008; Cideciyan et al. 2013; Russell et al. 2017; Duncan et al. 2018), the genetic payloads delivered by AAV-based therapies are restricted to ∼4.5 kb due to the small size of the AAV genome (Bainbridge et al. 2008, 2015). Thus, retinal disease-causing alterations in larger genes such as ABCA4, USH2A, and CEP290, have been difficult to correct using conventional AAV-based methods. In addition, AAV-based strategies rely on the supplementation of a wild-type (WT) copy of the mutant gene, which is not conducive to the treatment of autosomal-dominant retinal diseases. Finally, gene therapy applications of AAV vectors often require removal of the rep open reading frame (ORF) to prevent vector integration into the host genome. Thus, AAV vectors are conventionally expressed as episomes with waning therapeutic efficacy over time (Trapani and Auricchio 2018). Some groups have circumvented these obstacles by engineering dual AAV systems that carry segmented transgenes or by constructing a truncated, yet functional form of the gene of interest that is operable within the size constraints of the AAV vector (Lai et al. 2009).

Table 1.

Description of various gene-editing strategies that have been developed in recent years along with any associated ongoing clinical trials

Gene-editing strategy Diseases Clinical trial status Notes
Adeno-associated virus (AAV) vector RPE65-associated Leber congenital amaurosis (LCA) (FDA approved; NCT00999609)
CEP290-associated LCA (phase 2; NCT03872479)
CMS and FDA approved Low immunogenicity and high transduction efficiency in retinal pigmented epithelium
Therapeutic gene is limited to ∼4.5 kb in size
Difficult to apply to autosomal-dominant retinal diseases
Waning therapeutic efficacy over time
Lentiviruses Stargardt patients (NCT01367444) Phase 1-2a; terminated as sponsor decided to stop development of product Larger genetic payloads of up to ∼9 kb
Unwanted insertional mutations in host
CRISPR/Cas system RHO-adRP (preclinical)
Stargardt macular degeneration LCA
Preclinical Off-target effects
Inability to cleave single-stranded DNA (ssDNA) (Cas9)
Requires specific G-rich protospacer-adjacent motif (PAM) sequence localization (Cas9)
Inefficient in mitotically inactive cells (Cas9)
Base editing Muscular atrophy; inherited liver and skin diseases; RPE65-associated LCA; sickle cell disease; ALL; α1 antitrypsin deficiency; Stargardt disease Preclinical Independent of double-strand break (DSB) formation and homology-directed repair (HDR)-dependent DNA repair
High fidelity and efficiency
Minimal off-target mutations and indel events
Prime editing Facilitates reverse transcriptase (RT) hybridization and mutation correction on the opposing strand
Does not require HDR-mediated DNA repair or DSB formation
Limited to transition mutations
Dependent on PAM position near the desired incision site
Antisense oligonucleotides RHO-adRP (preclinical, phase 1/2; NCT04855045)
CEP290-associated LCA (preclinical)
Usher syndrome type 2 (phase 1/2; NCT03780257)
SCA7 (preclinical murine model)
Preclinical, phase 1/2 May be used in treatment of inherited retinal diseases (IRDs)
Unscalable to all mutated genes causing disease
Short-hairpin RNAs RHO-adRP (preclinical canine model) Preclinical Require delivery via a viral vector
Cas13 and ADAR Usher syndrome type 2 (preclinical) Preclinical Improved specificity and off-target effects compared to previous RNAi approaches

Lentiviruses

Lentiviruses are retroviruses with the ability to accommodate delivery of larger genetic payloads of up to ∼9 kb and have been viewed as attractive alternatives for the treatment of retinal diseases mediated by mutations in large causative genes (Coffin et al. 1997). However, lentiviral transduction results in integration of foreign DNA into the host, which can lead to unwanted insertional mutations (Trapani et al. 2014). The development of integration-deficient lentiviral vectors (IDLVs) enables expression of the lentiviral vector as a stable episome and has been successfully applied to the treatment of retinal dystrophy in a murine model (Yáñez-Muñoz et al. 2006).

DNA EDITING

CRISPR/Cas9

The CRISPR/Cas9 system is the current state-of-the-art technique for genomic surgery. Conventional applications center on a Cas9 endonuclease that is directed across the genome by a single-guide RNA (sgRNA) (Jinek et al. 2012). The sgRNA hybridizes with a complementary target DNA strand, and the Cas9 endonuclease delivers a precise molecular incision. This Cas9-mediated DNA cleavage relies on positioning near a spacer sequence or protospacer-adjacent motif (PAM), which must be directly downstream of the incision site. These DNA double-strand breaks (DSBs) subsequently activate one of two DNA repair pathways, the homology-directed repair (HDR) or nonhomologous end joining (NHEJ), which are exploited to generate a desired genetic alteration.

Cas 12

Cas12a (previously known as Cpf1) and its orthologs possess unique properties that expand on the repertoire of CRISPR/Cas applications. Compared to Cas9, they are more compact in size, generate staggered double-stranded DNA (dsDNA) breaks instead of blunt-ended dsDNA breaks, and reportedly have lower overall off-target effects (Anzalone et al. 2020). In addition, Cas12 can also cleave single-stranded DNA (ssDNA). Recent studies demonstrate that following recognition of a complementary DNA sequence, Cas12 can indiscriminately cleave surrounding ssDNA molecules, a property that has been exploited for the development of novel diagnostic technologies (Broughton et al. 2020). One key limitation of Cas9 and its orthologs is their heavy reliance on G-rich PAM sequences for localization. In contrast, Cas12a and its variants primarily recognize T-rich PAM sequences. Thus, a combination of Cas9 and Cas12 applications may expand the areas of the genome that are amenable to gene editing in retinal diseases.

CasX, also known as Cas12e, harbors many of the same features as the other Cas12 proteins in its family (Liu et al. 2019). However, it has an even lighter molecular weight with a size of only ∼980 amino acids (aa), which may facilitate efficient delivery. Furthermore, the mechanism by which the CasX protein processes and cleaves DNA differs from its predecessors. Scribe Therapeutics is optimizing this novel editor toward the treatment of several ophthalmologic diseases including retinitis pigmentosa, Stargardt macular degeneration, and LCA.

Cas14

Cas14 is a remarkably compact endonuclease identified from uncultivated archaea bacteria with a size ranging from 400 to 700 aa. In contrast to prior Cas proteins, it is guided by an RNA molecule and selectively binds and cleaves ssDNA. Strikingly, its function and localization are not dependent on the proximity of a PAM sequence, and like Cas12a, harbors potential to be used in the future for exquisite diagnostic applications.

Base Editing

CRISPR/Cas9 approaches for the correction of point mutations initially centered on the delivery of a WT Cas9 and an sgRNA directed toward the site of the point mutation followed by HDR using a functional donor sequence (Chapman et al. 2012). Unfortunately, HDR-mediated repair is highly inefficient, particularly in mitotically inactive cells. In addition, Cas9-mediated DSB formation is susceptible to widespread, inadvertent indels that may produce unwanted deleterious effects (Lin et al. 2014; Zhang et al. 2015).

Recent advancements have revealed a new class of gene-editing tools that are capable of editing ssDNA independent of DSB formation and HDR-dependent DNA repair. Termed base editors, they include cytosine base editors (CBEs), which consist of a catalytically inactivated Cas9 protein (dCas9) linked to a cytosine deaminase (rAPOBEC1), and adenine base editors (ABEs), which consist of a dCas9 linked to an adenine deaminase (Komor et al. 2016; Gaudelli et al. 2017). CBEs and ABEs perform point changes of C•G to T•A and vice versa, respectively, with high fidelity and efficiency and have been successfully applied to a variety of genetic diseases including muscular atrophy and inherited liver and skin diseases in preclinical studies (Rossidis et al. 2018; Villiger et al. 2018; Osborn et al. 2020).

Suh et al. demonstrated the utility of base editors in the treatment of RPE65-associated LCA caused by an underlying point mutation. Specifically, the group engineered a lentiviral vector containing a codon-optimized ABE variant (ABEmax) and an sgRNA directed toward a point mutation in exon 3 (c.130C > T; p.R44X) of the Rpe65 gene. In rd12 mice, lentiviral transduction via subretinal injection corrected the de novo nonsense mutation with a maximum 29% efficiency, recovered visual chromophore production, and restored retinal function as assessed by scotopic electroretinography (ERG), optomotor responses (OMRs), and visually evoked potentials (VEPs) (Suh et al. 2021). Strikingly, these improvements were achieved with minimal off-target mutations and indel events. Recently, base editing was demonstrated to be efficacious in long-lasting restoration of cone functionality and survival in mice, building on the success of AAV-mediated treatment of retinal pigment epithelium (RPE)-LCA. The patients with LCA who were treated with the AAV-mediated therapy often developed continued retinal degeneration after treatment (Cideciyan et al. 2013; Bainbridge et al. 2015). Base editing offers promising development of treatment for a variety of retinal diseases on a long-term basis (Caruso et al. 2022; Choi et al. 2022).

BEAM Therapeutics is leveraging base-editing techniques in the treatment of a variety of genetic diseases including sickle cell disease, acute lymphoblastic leukemia (ALL), α1 antitrypsin deficiency, and Stargardt disease, an inherited retinal disease caused by a wide array of mutations in the ABC4A gene (Tanna et al. 2017).

Prime Editing

While base editing is able to overcome a critical obstacle in its avoidance of DSB formation and HDR-dependent repair, its applications are limited to transition mutations and are dependent on PAM position near the desired incision site. Prime editing (PE) relies on reverse transcriptase (RT)-mediated repair to expand on the repertoire of desired mutational corrections including deletions, insertions, and all types of transitions and transversions, and, like base editing, PE does not require HDR-mediated DNA repair or DSB formation. PE consists of an RT, a pegRNA, and an SpCas9 nickase modified with an H840A mutation that causes the nickase to preferentially generate a single-strand break (SSB) along the PAM-containing strand (da Costa et al. 2021). The pegRNA contains an sgRNA directed toward the mutation site and an extension segment containing a spacer sequence, an sgRNA scaffold, a primer-binding sequence (PBS), and a reverse transcription template (RTT). The spacer sequence extends the 3′ end of the sgRNA, facilitating RT hybridization and mutation correction on the opposing strand (Fig. 1).

Figure 1.

Figure 1.

Prime editing is a novel strategy that uses Cas 9 nickase, pegRNA, and reverse transcriptase to edit target DNA. (1) The target DNA binds to the prime editing complex via protospacer-adjacent motif (PAM), and (2) the Cas 9 nickase creates a single-strand break (SSB) in the target DNA. (3) The reverse transcriptase reads the RNA and synthesizes new DNA to the edited strand. (4) The edited strand and unedited strand now have a base pair mismatch, and then a guide RNA (gRNA) directs the prime editor complex to create another SSB in the unedited strand. (5) The cell remakes the nicked strand using the edited strand as a template, creating a double-stranded section of edited DNA (in yellow). (RTT) Reverse transcription template, (PBS) primer-binding sequence.

Recently, Zhi et al. (2022) engineered a dual AAV8 split-PE system, which consists of Split-ABE- and Split-CBE-containing vectors. Split-ABE was then generated by two distinct mechanisms of splicing either by mRNA trans-splicing or intern-mediated protein slicing, the latter of which produces a Split-ABE that is more efficient in genome editing in vivo. Using this approach, the group delivered the dual AAV8 split-PE system into adult mouse retina via subretinal injection and successfully generated a G•A transversion in the Dnmt1 gene at the p.P55Q, c.G164T position. The transversion efficiency of the dual system was only 1.71%.

RNA Editing

CRISPR/Cas9 and other relevant genome surgery approaches result in permanent alterations to the patient's genomic DNA. Many patients often opt for strategies that ensure the integrity of the genome and limit permanent modifications to their underlying DNA. Thus, approaches that modulate disease processes at the RNA level are highly desirable and can address these patient-centered concerns. RNA-editing strategies include degradation of pathological mRNA molecules, modulation of pre-mRNA splicing, and, more recently, individual base editing of mRNA transcripts.

Antisense Oligonucleotides

Antisense oligonucleotides (ASOs) are synthetic small RNA molecules or nucleotide analogs that bind to complementary pre-mRNA sequences. Depending on the chemical modification made to these molecules, ASOs can initiate a variety of biochemical activities following pre-mRNA hybridization including mRNA degradation, translational obstruction, and exon exclusion/inclusion (Hammond and Wood 2011).

ASOs with the ability to degrade mRNA are of particular interest in the treatment of autosomal-dominant inherited retinal diseases (IRDs), which have been unamenable to gene replacement strategies in mouse models of retinal dystrophies (Wu et al. 2022). These ASO moieties hybridize to complementary pre-mRNA molecules and recruit ribonuclease H (RNase H1) to cleave the bound pre-mRNA molecule. Several ASO therapies centered on an mRNA degradation strategy have been developed for various diseases, and one such therapy is currently in phase 2 of clinical trials testing for the treatment of rhodopsin-mediated autosomal-dominant retinitis pigmentosa (RHO-adRP) (NCT04123626).

Other ASO moieties can facilitate skipping of pathological exon or pseudoexon containing stop codon mutations. By avoiding these harmful segments, an appropriate reading frame can be reestablished, and the production of functional protein with the required domains can be restored. ProQR Therapeutics is employing this strategy in the treatment of LCA caused by an underlying c.2991 + 1655 > G intronic mutation in the CEP290 gene and in the treatment of Usher syndrome type 2 with underlying exon 13 mutation.

Short-Hairpin RNAs

Short-hairpin RNAs (shRNAs) are small RNA molecules that initiate the intracellular RNA interference (RNAi) pathway. Specifically, the RNase III Dicer complex is recruited to process the shRNAs into small-interfering RNAs (siRNAs). These RNA molecules can then hybridize to complementary mRNA molecules and recruit the RNA-induced silencing complex (RISC) for mRNA degradation. Importantly, while siRNAs can be delivered as discrete RNA molecules, shRNAs require delivery via a lentiviral DNA vector (Lambeth and Smith 2013).

Cas13 and ADARs

Recent advances in CRISPR/Cas therapies have yielded unique Cas proteins with the capacity to precisely target and degrade RNA transcripts. Abudayyeh et al. isolated an RNA-guided, RNA endonuclease from the Leptotrichia shahii bacterium (Abudayyeh et al. 2017; Cabral et al. 2017; Sengillo et al. 2017). Previously termed C2c2, Cas13a relies on an sgRNA to localize to and target mRNA transcripts for degradation, achieving the same efficacy as previous RNAi approaches but with improved specificity and off-target effects.

While the aforementioned RNA-based therapies center on transcript degradation, recent advances in Cas13 systems have yielded the possibility of base editing at the mRNA level. These approaches employ a catalytically inactive dCas13 and an adenosine deaminase acting on RNA (ADAR). dCas13 guides the ADAR to the desired editing site to facilitate A·I trans conversions. As of now, however, ADARs are only able to facilitate A·I conversions and therefore can only correct A > G mutations.

DISEASE-SPECIFIC ADVANCEMENTS

RPE65-Associated LCA

The RPE65 gene encodes a crucial retinoid isomerase that regenerates 11-cis-retinal for rod and cone photoreceptors in the classical RPE visual cycle. Mutations in the RPE65 gene among several other genes have been extensively linked to LCA, a recessively inherited retinal disease characterized by severe vision impairment, nystagmus, and diminished ERG responses by the first year of life (Gu et al. 1997; Perrault et al. 1999).

In 2017, Voretigene neparvovec-rzyl (Luxturna) was approved by the U.S. Food and Drug Administration (FDA) as the first gene-replacement therapy for the treatment of RPE-associated LCA. Luxturna is an AAV serotype 2 (AAV2) vector that contains a WT copy of the RPE65 gene and is delivered via subretinal injection to restore RPE65 production by RPE cells (Bainbridge et al. 2008). The AAV2 vector is expressed intracellularly as a stable episome in nonmitotic RPE cells. Thus, the therapy could conceivably serve as a one-time treatment solution for patients with RPE-associated LCA. In clinical trials, Luxturna-treated patients demonstrated dramatic improvements in bilateral multi-luminance mobility testing (MLMT) compared to control patients following 1 year of treatment. Moreover, patients also reported improvements in visual field testing, best corrected visual acuity (BCVA), and overall navigational ability in both light and dark conditions (Maguire et al. 2019). These exciting clinical developments in the landmark case of Luxturna provide compelling support for future investigation and the use of gene therapy strategies in the treatment of retinal diseases.

CEP290-Associated LCA

The CEP290 gene encodes a critical regulator protein that directs ciliary trafficking and synthesis. These processes are vital to the function of the photoreceptor outer segment, which is dependent on effective transport of proteins and lipids synthesized in the photoreceptor inner segment. Mutations in CEP290 lead to a form of LCA, LCA10, which is highlighted by nystagmus, compromised ERG response, and severe vision loss by the first year of life (Perrault et al. 2007). Mutations in the CEP290 primarily exhibit an autosomal-recessive pattern of inheritance and have been implicated in ∼30% of all LCA patients. The c.2991 + 1655A > G mutation in the intervening sequence in intron 26 (IVS26) represents the most frequent underlying mutation for LCA10 and produces an mRNA with a premature stop codon (p.Cys998*). Subsequently, both a nonfunctional truncated CEP290 protein and a full length-WT CEP290 protein are produced, resulting in haploinsufficiency (Ruan et al. 2017).

Gene augmentation approaches in the treatment of CEP290-mediated LCA have been hindered by the size of the CEP290 gene (∼8 kb), which is not able to be accommodated by the size restrictions of the AAV vector. However, one group circumvented this size limitation by developing a truncated, yet fully functional version of the gene, termed miniCEP290. The truncated gene was successfully packaged and delivered by an AAV vector with demonstrable improvement in photoreceptor viability in a murine model (Zhang et al. 2018).

In the realm of gene editing, Maeder et al. (2019) developed an AAV5 vector containing a Staphylococcus aureus Cas9 (saCas9) and two sgRNAs flanking the IVS26 mutation in the CEP290 gene. This construct, termed EDIT-101, removes the IVS26-containing segment in the CEP290 gene to restore mRNA splicing and functional CEP290 protein expression. The group demonstrated that EDIT-101 was able to recover CEP290 mRNA splicing and protein function in an ex vivo, postmortem human retinal model with a productive edit rate of ∼17%. These results were recapitulated in HuCEP290 mice, a human CEP290 IVS26 knock-in mouse model. Because mouse retina harbors lower proportions of cones (∼3%), the group further evaluated the efficacy of EDIT-101 in a cynomolgus monkey nonhuman primate (NHP) model, which better recapitulates the human eye (Carter-Dawson and LaVail 1979; Curcio et al. 1987). In this NHP model, administration of EDIT-101 via subretinal injection in the perifoveal region achieved a maximum 28% productive edit rate with a vector concentration of 1 × 1012 vgml−1.

In 2019, Editas Medicine spearheaded the phase 1/2 Brilliance clinical trial (NCT03872479) to assess the safety, tolerability, and efficacy of EDIT-101 (Fig. 2) in LCA patients aged 3 and older with at least one mutation in the CEP290 gene (including c.2991 + 1655A > G) and a visual acuity no better than 0.4 LogMAR (NCT03872479). Primary outcome measures include drug toxicity and adverse effects assessed every 3 months for up to 1 year. Secondary measures including BCVA, color vision, full-field stimulus testing (FST), microperimetry, and quality of life scores, among other metrics. The efficacy component of the study consists of five subjects, three of which received a single dose of EDIT-101 at the moderate-level dose and two at the low-level dose (1.1 × 1012vg/mL and 6 × 1011vg/mL, respectively). Unfortunately, initial results have been inconclusive with only two patients demonstrating improvement in outcome measures at 6 months following treatment. Furthermore, only one patient achieved clinically meaningful improvement, an increase of 0.7logMAR in BCVA. Reported adverse effects include retinal tears and hemorrhages.

Figure 2.

Figure 2.

Mechanisms of two therapies for CEP290. (A) Mutations in CEP290 cause incorrect splicing, which results in a truncated CEP290 protein. The CEP290 protein is a crucial component of protein trafficking in cilia and results in vision loss due to the malfunction of the outer segment of the photoreceptors. (B) QR-110 is an RNA therapeutic designed by ProQR, which binds to the mutated intronic sequence in CEP290 to prevent improper splicing and allow for correct protein formation. (C) EDIT-101 is a gene-editing therapy developed by Editas Medicine, which removes the disease-causing mutation, allowing for correct splicing and full-length CEP290 formation.

Toward an RNA-editing approach, Dulla et al. (2018) developed an optimized ASO capable of restoring normal RNA splicing in LCA10 with an underlying CEP290 c.2991 + 1655A > G mutation. Also known as QR-110 and sepofarsen, this construct does not require AAV packaging but can be delivered directly via intravitreal injection (IVT). In preclinical studies, QR-110 treatment successfully corrected CEP290 c.2991 + 1655A > G mutations in human retinal organoids and demonstrated robust bioavailability in WT mice and rabbit retinas and excellent tolerability in an NHP model.

ProQR Therapeutics is currently conducting the phase 2/3 Illuminate clinical trial (NCT03913143) to assess the safety, tolerability, and efficacy of sepofarsen in the treatment of LCA10 caused by underlying CEP290 p.Cys998X mutation (Fig. 2). Thirty-six patients aged 8 and older received one treatment dose of sepofarsen at 0 mo and 3 mo and every 6 mo thereafter, up to 24 mo. The primary outcome measure is the mean change in BCVA following 12 mo of treatment, and secondary outcome measures include navigational ability, FST, ERG, ocular adverse events, and changes in photoreceptor outer/inner layer, among others. While an initial case study reported sustained visual improvement in one enrolled patient 15 mo following treatment, the Illuminate trial ultimately failed to meet primary outcome measure (Cideciyan et al. 2019). In addition, differences in secondary outcome measures between the treatment and control groups were not statistically significant. Although sepofarsen was overall well-tolerated, adverse events included retinal thinning and cystoid macular edema (CME), which were consistent with findings from phase 1/2 of the trial.

RHO-adRP

Retinitis pigmentosa (RP) is a group of rare inherited retinal disorders initiated by rod photoreceptor degeneration with progression to a wide range of disease phenotypes from mild night blindness to severe reductions in visual acuity (Verbakel et al. 2018). In the early stages of RP, patients report diminishing night vision that then progresses to deterioration of peripheral vision, termed “tunnel vision.” In later stages, significant rod photoreceptor loss subsequently leads to secondary cone photoreceptor death, which may compromise daytime visual acuity and result in complete blindness (Narayan et al. 2016).

RP can be precipitated by a compendium of defects in the photoreceptors and RPE. In particular, the rhodopsin (RHO) gene encodes a G-protein coupled receptor that serves as the primary visual pigment for rod photoreceptors (Nathans et al. 1986). Mutations in the RHO gene exhibit an autosomal-dominant hereditary pattern of retinitis pigmentosa and were the first to be associated with the development of RP. Rhodopsin-mediated autosomal-dominant retinitis pigmentosa (RHO-adRP) accounts for almost 25% of all autosomal-dominant retinitis pigmentosa (adRP) cases (Jacobson et al. 1991).

Conventional gene therapy approaches for the treatment of RHO-adRP centered on attenuating expression of the mutant RHO gene, which is directly implicated in the retinal degeneration process. QR-1123 is an ASO therapy designed to treat RHO-adRP with underlying P23H mutation in the RHO gene by targeting P23H mutant RHO transcripts for degradation (Murray et al. 2015). In preclinical studies, intravitreal injection of QR-1123 effectively attenuated P23H mutant RHO expression while preserving WT RHO expression, restored ERG response, and augmented outer nuclear layer (ONL) thickness in a P23H mutant murine model. In 2018, ProQR Therapeutics acquired QR-1123 from Ionis Pharmaceuticals and initiated a phase 1/2 Aurora clinical trial (NCT04123626) to assess the safety, tolerability, and efficacy of QR-1123 in the treatment of patients with P23H mutated RHO-adRP. Phase 1 results indicate that QR-1123 is well tolerated with an excellent overall safety profile.

One critical limitation for therapies such as QR-1123 is that they are not scalable to the over 150 reported mutations in the RHO gene that have been shown to cause RHO-adRP. Thus, a more conventional paradigm involves eliminating expression of both the patient WT and mutant RHO genes followed by gene replacement with an exogenous WT RHO gene (Meng et al. 2020). Such a strategy can be expanded to treat all forms of RHO-adRP without designing individually tailored therapies for each underlying mutation.

Cideciyan et al. (2018) developed a single AAV system containing both a WT copy of the human RHO gene and a component encoding an shRNA that targets the human RHO gene. Importantly, the group engineered the codon-modified RHO gene to be resistant to degradation by the vector-carrying shRNA (Gorbatyuk et al. 2007; O'Reilly et al. 2007; Palfi et al. 2012).

A single subretinal injection of the shRNA820 candidate in a canine model for light-induced RHO-adRP degeneration model dramatically attenuated RHO mRNA and protein expression in mutant RHO retinas. Next, the group delivered the AAV system containing the shRNA820 and the codon-modified human RHO gene and showed that the rescue RHO gene could be expressed at maximum 132% and 33%, at the mRNA and protein level, respectively, in comparison to control retinas. The group also demonstrated that vector administration could preserve retinal ONL thickness, photoreceptor cells, and the outer segment layers in treated retinas (Cideciyan et al. 2018). In 2018, IVERIC Bio acquired the rights to the therapy, now named IC-100, with plans to initiate a phase 1/2 clinical trial. However, subsequent preclinical studies in NHPs were unable to recapitulate the safety profiles previously reported in canine models and have stalled clinical trial proposals. IVERIC Bio is seeking potential collaborations as it turns its attention to other products in its portfolio. Natural history studies and IND-enabling activities for IC-100 are ongoing (Meng et al. 2020).

Using a CRISPR/Cas9-based approach in the treatment of RHO-adRP, Editas Medicine developed a dual AAV5 system to knock out and replace the mutant RHO gene. The dual system consists of a vector encoding an S. aureus Cas9 (SaCas) cRNA and a second encoding several gRNAs and a codon-optimized copy of the RHO cRNA. Widespread frameshift deletions generated by this dual system dramatically diminished RHO expression in human retinal explants, and a rescue RHO vector was sufficient to restore RHO expression similar to physiological levels (Diner et al. 2020).

USH2A

Biallelic mutations in the USH2A gene result in Usher syndrome type 2, an autosomal-recessive syndrome characterized by congenital hearing deficits and late-onset retinitis pigmentosa with late-onset, or nonsyndromic retinitis pigmentosa (nsRP) (van Wijk et al. 2004). USH2A encodes usherin, a transmembrane protein that is critical for photoreceptor outer layer and periciliary region structure (Géléoc and El-Amraoui 2020). ∼35% of USH2A mutations occur in exon 13, which can cause premature termination and inappropriate splicing resulting in dysfunctional usherin protein. Like CEP290-mediated LCA, Usher syndrome type 2 has been difficult to approach using AAV-based gene augmentation strategies due to the size of the USH2A gene.

Dulla et al. developed the novel ASO QR-421a, which promotes exon 13 skipping in the mutant USH2A gene, thereby restoring normal pre-mRNA splicing (Dulla et al. 2021). QR-421a treatment selectively induced exon 13 skipping in an induced pluripotent stem cell model derived from the photoreceptors of a patient with homozygous c.2299delG mutation in the USH2A gene. In addition, the group showed that QR-421a had high bioavailability in the photoreceptor outer layer of WT mice. Based on these promising results, ProQR Therapeutics initiated phase 1/2 Stellar clinical trial (NCT03780257) to assess the safety, tolerability, and efficacy of QR-421a (also termed ultevursen) in patients with exon 13 mutations in the USH2A gene. Results from phase 1/2 have been moderately positive. Ultevursen is well tolerated and is without significant adverse effects, and the 16 subjects that received a single dose of the therapy reported a mean 6.0 letter improvement in BCVA. ProQR has since initiated two phase 2/3 clinical trials, Sirius (NCT05158296) and Celeste (NCT05176717), with results expected in 2024.

Currently, Locanabio is developing a Cas13d-centered RNA-editing approach to promote exon 13 skipping in the USH2A gene. In preliminary studies, a single AAV vector containing a catalytically inactive Cas13d (dCas13d) and a gRNA targeting a portion of exon 13 augmented the proportion of truncated usherin protein with exon 13 exclusion in HEK293T cells (Lardelli et al. 2021).

Spinocerebellar Ataxia

Polyglutamine spinocerebellar ataxias (SCAs) represent a heterogeneous group of six autosomal-dominant, neurodegenerative diseases characterized by a constellation of symptoms including ataxia, speech difficulties, impaired gait, and motor dysfunction (Duenas et al. 2006). The six disease groups, SCA1, SCA2, SCA3, SCA6, SCA7, and SCA17, are defined by CAG (glutamate) repeat expansions in the coding region of their associated genes, ATXN1, ATXN2, ATXN3, CACNA1A, ATXN7, and TBP, respectively (Paulson et al. 2017; Buijsen et al. 2019).

In particular, SCA7 distinctively presents with pigmentary regional dystrophy and progressive visual loss beginning in the macular region and spreading to the retinal periphery (Lebre and Brice 2003). These pathological changes can be clinically monitored via optical coherence tomography (OCT), ERG, and full-field ERG (ffERG), which demonstrate retinal thinning of the retinal nerve fiber layer, diminished amplitudes, and diminished 30 Hz cone flicker amplitudes, respectively (Campos-Romo et al. 2018; Park et al. 2020).

Niu et al. (2018) successfully developed an ASO against Ataxin-7, and delivery of the ASO in a SCA7 266Q knockin mouse model via intravitreal injection dramatically attenuated Ataxin-7 expression. In addition, mice receiving the ASO therapy recovered rod and cone function. Ramachandran developed short double-stranded RNA (dsRNA) molecules to target repeat expansion of SCA7 (Ramachandran et al. 2014). In preclinical studies, this RNAi strategy significantly reduced WT and mutant Ataxin-7 expression, improved disease phenotype in a murine model of SCA7, and demonstrated minimal toxic effects.

CONCLUSION

The past decade has seen an explosion in gene therapy and editing techniques with major improvements to existing technologies. While Luxturna remains the only FDA-approved AAV2 therapy for the treatment of RPE65-mediated LCA, editing tools including CRISPR/Cas9 and its emerging novel variants have expanded the ways that we may approach the treatment of IRDs. These technologies have opened the possibility for researchers to treat IRDs with varying inheritance patterns and disease pathologies, IRDs with underlying mutations in previously inoperable genes due to size constraints, and IRDs with aberrations at the DNA and/or RNA levels. Meanwhile, existing RNA-editing techniques such as RNAi continue to be refined and optimized, and several trials assessing the efficacy of ASOs and RNA-based therapies have since been initiated with promising results. While the majority of clinical trials have failed to translate preclinical findings in patients and the recent results from ProQR's Brilliance trial for the treatment of CEP290-mediated LCA represents one such trial, gene therapy and editing techniques are still in the early stages of development. Undoubtedly, gene therapy and editing will continue to encounter challenges and several rounds of refinement in clinical trials before it is shown to be safe and effective in the treatment of many IRDs. Simultaneously, gene therapy has garnered overwhelming public support and financial backing. Cas14a-Scribe Therapeutics recently closed a 100-million-dollar series B round, and other top gene therapy companies carry robust market caps. There are more gene therapy–based clinical trials than ever before with more than 20 recruiting, enrolling by invitation, and active (not recruiting) trials designated on Clinicaltrials.gov as of July 2022. Finally, the success story of Luxturna and the profound impact that it has had on patients with RPE65-mediated LCA indicate that gene therapy and editing are viable strategies in the treatment of IRDs and will inevitably become mainstays in the way we manage patient IRDs in the future.

COMPETING INTEREST STATEMENT

S.H.T. receives financial support from Abeona Therapeutics and Emendo. He is also the founder of Rejuvitas and is on the scientific and clinical advisory board for Nanoscope Therapeutics and Medical Excellence Capital.

ACKNOWLEDGMENTS

Jonas Children's Vision Care receives support from the National Institutes of Health (grant numbers: 5P30CA013696, U01EY030580, U54OD020351, R24EY028758, R24EY027285, 5P30EY019007, R01EY018213, R01EY024698, R01EY033770, R21AG05043; the Schneeweiss Stem Cell Fund; New York State (grant number SDHDOH01-C32590GG-3450000); the Foundation Fighting Blindness New York Regional Research Center Grant (grant numbers PPA-1218-0751-COLU and TA-NMT-0116-0692-COLU); Nancy & Kobi Karp; the Crowley Family Funds; The Rosenbaum Family Foundation; Alcon Research Institute; the Gebroe Family Foundation; and unrestricted funds from Research to Prevent Blindness, New York, NY, USA.

Footnotes

Editors: Eyal Banin, Jean Bennett, Jacque L. Duncan, Botond Roska, and José-Alain Sahel

Additional Perspectives on Retinal Disorders: Genetic Approaches to Diagnosis and Treatment available at www.perspectivesinmedicine.org

REFERENCES

  1. Abudayyeh OO, Gootenberg JS, Essletzbichler P, Han S, Joung J, Belanto JJ, Verdine V, Cox DBT, Kellner MJ, Regev A, et al. 2017. RNA targeting with CRISPR-Cas13. Nature 550: 280–284. 10.1038/nature24049 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Anzalone AV, Koblan LW, Liu DR. 2020. Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime editors. Nat Biotechnol 38: 824–844. 10.1038/s41587-020-0561-9 [DOI] [PubMed] [Google Scholar]
  3. Bainbridge JW, Smith AJ, Barker SS, Robbie S, Henderson R, Balaggan K, Viswanathan A, Holder GE, Stockman A, Tyler N, et al. 2008. Effect of gene therapy on visual function in Leber's congenital amaurosis. N Engl J Med 358: 2231–2239. 10.1056/NEJMoa0802268 [DOI] [PubMed] [Google Scholar]
  4. Bainbridge JW, Mehat MS, Sundaram V, Robbie SJ, Barker SE, Ripamonti C, Georgiadis A, Mowat FM, Beattie SG, Gardner PJ, et al. 2015. Long-term effect of gene therapy on Leber's congenital amaurosis. N Engl J Med 372: 1887–1897. 10.1056/NEJMoa1414221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Broughton JP, Deng X, Yu G, Fasching CL, Servellita V, Singh J, Miao X, Streithorst JA, Granados A, Sotomayor-Gonzalez A, et al. 2020. CRISPR-Cas12-based detection of SARS-CoV-2. Nat Biotechnol 38: 870–874. 10.1038/s41587-020-0513-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Buijsen RAM, Toonen LJA, Gardiner SL, van Roon-Mom WMC. 2019. Genetics, mechanisms, and therapeutic progress in polyglutamine spinocerebellar ataxias. Neurotherapeutics 16: 263–286. 10.1007/s13311-018-00696-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Cabral T, DiCarlo JE, Justus S, Sengillo JD, Xu Y, Tsang SH. 2017. CRISPR applications in ophthalmologic genome surgery. Curr Opin Ophthalmol 28: 252–259. 10.1097/ICU.0000000000000359 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Campos-Romo A, Graue-Hernandez EO, Pedro-Aguilar L, Hernandez-Camarena JC, Rivera-De la Parra D, Galvez V, Diaz R, Jimenez-Corona A, Fernandez-Ruiz J. 2018. Ophthalmic features of spinocerebellar ataxia type 7. Eye (Lond) 32: 120–127. 10.1038/eye.2017.135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Carter-Dawson LD, LaVail MM. 1979. Rods and cones in the mouse retina. I: Structural analysis using light and electron microscopy. J Comp Neurol 188: 245–262. 10.1002/cne.901880204 [DOI] [PubMed] [Google Scholar]
  10. Caruso SM, Quinn PM, da Costa BL, Tsang SH. 2022. CRISPR/Cas therapeutic strategies for autosomal dominant disorders. J Clin Invest 132: e158287. 10.1172/JCI158287 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Caspi RR. 2010. A look at autoimmunity and inflammation in the eye. J Clin Invest 120: 3073–3083. 10.1172/JCI42440 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Chapman JR, Taylor MR, Boulton SJ. 2012. Playing the end game: DNA double-strand break repair pathway choice. Mol Cell 47: 497–510. 10.1016/j.molcel.2012.07.029 [DOI] [PubMed] [Google Scholar]
  13. Choi EH, Suh S, Foik AT, Leinonen H, Newby GA, Gao XD, Banskota S, Hoang T, Du SW, Dong Z, et al. 2022. In vivo base editing rescues cone photoreceptors in a mouse model of early-onset inherited retinal degeneration. Nat Commun 13: 1830. 10.1038/s41467-022-29490-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Cideciyan AV, Jacobson SG, Beltran WA, Sumaroka A, Swider M, Iwabe S, Roman AJ, Olivares MB, Schwartz SB, Komaromy AM, et al. 2013. Human retinal gene therapy for Leber congenital amaurosis shows advancing retinal degeneration despite enduring visual improvement. Proc Natl Acad Sci 110: E517–E525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Cideciyan AV, Sudharsan R, Dufour VL, Massengill MT, Iwabe S, Swider M, Lisi B, Sumaroka A, Marinho LF, Appelbaum T, et al. 2018. Mutation-independent rhodopsin gene therapy by knockdown and replacement with a single AAV vector. Proc Natl Acad Sci 115: E8547–E8556. 10.1073/pnas.1805055115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Cideciyan AV, Jacobson SG, Drack AV, Ho AC, Charng J, Garafalo AV, Roman AJ, Sumaroka A, Han IC, Hochstedler MD, et al. 2019. Effect of an intravitreal antisense oligonucleotide on vision in Leber congenital amaurosis due to a photoreceptor cilium defect. Nat Med 25: 225–228. 10.1038/s41591-018-0295-0 [DOI] [PubMed] [Google Scholar]
  17. Coffin JM, Hughes SH, Varmus HE. 1997. Retroviruses. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY. [PubMed] [Google Scholar]
  18. Curcio CA, Sloan KR Jr, Packer O, Hendrickson AE, Kalina RE. 1987. Distribution of cones in human and monkey retina: individual variability and radial asymmetry. Science 236: 579–582. 10.1126/science.3576186 [DOI] [PubMed] [Google Scholar]
  19. da Costa BL, Levi SR, Eulau E, Tsai YT, Quinn PMJ. 2021. Prime editing for inherited retinal diseases. Front Genome Ed 3: 775330. 10.3389/fgeed.2021.775330 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Diner BA, Dass A, Nayak R, Flinkstrom Z, Tallo T, Miller M, DaSilva J, Gotta G, Wang T, Marco E, et al. 2020. Dual AAV-based “knock-out-and-replace” of RHO as a therapeutic approach to treat RHO-associated autosomal dominant retinitis pigmentosa (RHO adRP). Editas Medicine, Cambridge MA. [Google Scholar]
  21. Duenas AM, Goold R, Giunti P. 2006. Molecular pathogenesis of spinocerebellar ataxias. Brain 129: 1357–1370. 10.1093/brain/awl081 [DOI] [PubMed] [Google Scholar]
  22. Dulla K, Aguila M, Lane A, Jovanovic K, Parfitt DA, Schulkens I, Chan HL, Schmidt I, Beumer W, Vorthoren L, et al. 2018. Splice-modulating oligonucleotide QR-110 restores CEP290 mRNA and function in human c.2991+1655A>G LCA10 models. Mol Ther Nucleic Acids 12: 730–740. 10.1016/j.omtn.2018.07.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Dulla K, Slijkerman R, van Diepen HC, Albert S, Dona M, Beumer W, Turunen JJ, Chan HL, Schulkens IA, Vorthoren L, et al. 2021. Antisense oligonucleotide-based treatment of retinitis pigmentosa caused by USH2A exon 13 mutations. Mol Ther 29: 2441–2455. 10.1016/j.ymthe.2021.04.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Duncan JL, Pierce EA, Laster AM, Daiger SP, Birch DG, Ash JD, Iannaccone A, Flannery JG, Sahel JA, Zack DJ, et al. 2018. Inherited retinal degenerations: current landscape and knowledge gaps. Transl Vis Sci Technol 7: 6. 10.1167/tvst.7.4.6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Gaudelli NM, Komor AC, Rees HA, Packer MS, Badran AH, Bryson DI, Liu DR. 2017. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551: 464–471. 10.1038/nature24644 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Géléoc GGS, El-Amraoui A. 2020. Disease mechanisms and gene therapy for Usher syndrome. Hear Res 394: 107932. 10.1016/j.heares.2020.107932 [DOI] [PubMed] [Google Scholar]
  27. Gorbatyuk M, Justilien V, Liu J, Hauswirth WW, Lewin AS. 2007. Suppression of mouse rhodopsin expression in vivo by AAV mediated siRNA delivery. Vision Res 47: 1202–1208. 10.1016/j.visres.2006.11.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Gu SM, Thompson DA, Srikumari CR, Lorenz B, Finckh U, Nicoletti A, Murthy KR, Rathmann M, Kumaramanickavel G, Denton MJ, Gal A. 1997. Mutations in RPE65 cause autosomal recessive childhood-onset severe retinal dystrophy. Nat Genet 17: 194–197. 10.1038/ng1097-194 [DOI] [PubMed] [Google Scholar]
  29. Hammond SM, Wood MJ. 2011. Genetic therapies for RNA mis-splicing diseases. Trends Genet 27: 196–205. 10.1016/j.tig.2011.02.004 [DOI] [PubMed] [Google Scholar]
  30. Jacobson SG, Kemp CM, Sung CH, Nathans J. 1991. Retinal function and rhodopsin levels in autosomal dominant retinitis pigmentosa with rhodopsin mutations. Am J Ophthalmol 112: 256–271. 10.1016/S0002-9394(14)76726-1 [DOI] [PubMed] [Google Scholar]
  31. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. 2012. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337: 816–821. 10.1126/science.1225829 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Komor AC, Kim YB, Packer MS, Zuris JA, Liu DR. 2016. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533: 420–424. 10.1038/nature17946 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Lai Y, Thomas GD, Yue Y, Yang HT, Li D, Long C, Judge L, Bostick B, Chamberlain JS, Terjung RL, et al. 2009. Dystrophins carrying spectrin-like repeats 16 and 17 anchor nNOS to the sarcolemma and enhance exercise performance in a mouse model of muscular dystrophy. J Clin Invest 119: 624–635. 10.1172/JCI36612 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Lambeth LS, Smith CA. 2013. Short hairpin RNA-mediated gene silencing. Methods Mol Biol 942: 205–232. 10.1007/978-1-62703-119-6_12 [DOI] [PubMed] [Google Scholar]
  35. Lardelli R, Roth DM, Nachtrab G, Geddes C, Wilson A, Narayan N, Villegas G, Adams R, Gibbs D, Batra R. 2021. RNA-targeting gene therapy exon-skipping strategy to treat genetic diseases. Locanabio, San Diego, CA. [Google Scholar]
  36. Lebre AS, Brice A. 2003. Spinocerebellar ataxia 7 (SCA7). Cytogenet Genome Res 100: 154–163. 10.1159/000072850 [DOI] [PubMed] [Google Scholar]
  37. Lin S, Staahl BT, Alla RK, Doudna JA. 2014. Enhanced homology-directed human genome engineering by controlled timing of CRISPR/Cas9 delivery. eLife 3: e04766. 10.7554/eLife.04766 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Liu JJ, Orlova N, Oakes BL, Ma E, Spinner HB, Baney KLM, Chuck J, Tan D, Knott GJ, Harrington LB, et al. 2019. Author correction: CasX enzymes comprise a distinct family of RNA-guided genome editors. Nature 568: E8–E10. 10.1038/s41586-019-1084-8 [DOI] [PubMed] [Google Scholar]
  39. Maeder ML, Stefanidakis M, Wilson CJ, Baral R, Barrera LA, Bounoutas GS, Bumcrot D, Chao H, Ciulla DM, DaSilva JA, et al. 2019. Development of a gene-editing approach to restore vision loss in Leber congenital amaurosis type 10. Nat Med 25: 229–233. 10.1038/s41591-018-0327-9 [DOI] [PubMed] [Google Scholar]
  40. Maguire AM, Simonelli F, Pierce EA, Pugh EN Jr, Mingozzi F, Bennicelli J, Banfi S, Marshall KA, Testa F, Surace EM, et al. 2008. Safety and efficacy of gene transfer for Leber's congenital amaurosis. N Engl J Med 358: 2240–2248. 10.1056/NEJMoa0802315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Maguire AM, Russell S, Wellman JA, Chung DC, Yu ZF, Tillman A, Wittes J, Pappas J, Elci O, Marshall KA, et al. 2019. Efficacy, safety, and durability of voretigene neparvovec-rzyl in RPE65 mutation-associated inherited retinal dystrophy: results of phase 1 and 3 trials. Ophthalmology 126: 1273–1285. 10.1016/j.ophtha.2019.06.017 [DOI] [PubMed] [Google Scholar]
  42. Medawar PB. 1948. Immunity to homologous grafted skin; the fate of skin homografts transplanted to the brain, to subcutaneous tissue, and to the anterior chamber of the eye. Br J Exp Pathol 29: 58–69. [PMC free article] [PubMed] [Google Scholar]
  43. Meng D, Ragi SD, Tsang SH. 2020. Therapy in rhodopsin-mediated autosomal dominant retinitis pigmentosa. Mol Ther 28: 2139–2149. 10.1016/j.ymthe.2020.08.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Murray SF, Jazayeri A, Matthes MT, Yasumura D, Yang H, Peralta R, Watt A, Freier S, Hung G, Adamson PS, et al. 2015. Allele-specific inhibition of rhodopsin with an antisense oligonucleotide slows photoreceptor cell degeneration. Invest Ophthalmol Vis Sci 56: 6362–6375. 10.1167/iovs.15-16400 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Narayan DS, Wood JP, Chidlow G, Casson RJ. 2016. A review of the mechanisms of cone degeneration in retinitis pigmentosa. Acta Ophthalmol 94: 748–754. 10.1111/aos.13141 [DOI] [PubMed] [Google Scholar]
  46. Nathans J, Piantanida TP, Eddy RL, Shows TB, Hogness DS. 1986. Molecular genetics of inherited variation in human color vision. Science 232: 203–210. 10.1126/science.3485310 [DOI] [PubMed] [Google Scholar]
  47. Niu C, Prakash TP, Kim A, Quach JL, Huryn LA, Yang Y, Lopez E, Jazayeri A, Hung G, Sopher BL, et al. 2018. Antisense oligonucleotides targeting mutant Ataxin-7 restore visual function in a mouse model of spinocerebellar ataxia type 7. Sci Transl Med 10: eaap8677. 10.1126/scitranslmed.aap8677 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. O'Reilly M, Palfi A, Chadderton N, Millington-Ward S, Ader M, Cronin T, Tuohy T, Auricchio A, Hildinger M, Tivnan A, et al. 2007. RNA interference-mediated suppression and replacement of human rhodopsin in vivo. Am J Hum Genet 81: 127–135. 10.1086/519025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Osborn MJ, Newby GA, McElroy AN, Knipping F, Nielsen SC, Riddle MJ, Xia L, Chen W, Eide CR, Webber BR, et al. 2020. Base editor correction of COL7A1 in recessive dystrophic epidermolysis bullosa patient-derived fibroblasts and iPSCs. J Invest Dermatol 140: 338–347.e5. 10.1016/j.jid.2019.07.701 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Palfi A, Chadderton N, McKee AG, Blanco Fernandez A, Humphries P, Kenna PF, Farrar GJ. 2012. Efficacy of codelivery of dual AAV2/5 vectors in the murine retina and hippocampus. Hum Gene Ther 23: 847–858. 10.1089/hum.2011.142 [DOI] [PubMed] [Google Scholar]
  51. Park JY, Joo K, Woo SJ. 2020. Ophthalmic manifestations and genetics of the polyglutamine autosomal dominant spinocerebellar ataxias: a review. Front Neurosci 14: 892. 10.3389/fnins.2020.00892 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Paulson HL, Shakkottai VG, Clark HB, Orr HT. 2017. Polyglutamine spinocerebellar ataxias—from genes to potential treatments. Nat Rev Neurosci 18: 613–626. 10.1038/nrn.2017.92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Perrault I, Delphin N, Hanein S, Gerber S, Dufier JL, Roche O, Defoort-Dhellemmes S, Dollfus H, Fazzi E, Munnich A, et al. 2007. Spectrum of NPHP6/CEP290 mutations in Leber congenital amaurosis and delineation of the associated phenotype. Hum Mutat 28: 416. 10.1002/humu.9485 [DOI] [PubMed] [Google Scholar]
  54. Perrault I, Rozet JM, Gerber S, Ghazi C, Leowski C, Ducroq D, Souied E, Dufier JL, Munnich A, Kaplan J. 1999. Leber congenital amaurosis. Mol Genet Metab 68: 200–208. 10.1006/mgme.1999.2906 [DOI] [PubMed] [Google Scholar]
  55. Ramachandran PS, Boudreau RL, Schaefer KA, La Spada AR, Davidson BL. 2014. Nonallele specific silencing of ataxin-7 improves disease phenotypes in a mouse model of SCA7. Mol Ther 22: 1635–1642. 10.1038/mt.2014.108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Rossidis AC, Stratigis JD, Chadwick AC, Hartman HA, Ahn NJ, Li H, Singh K, Coons BE, Li L, Lv W, et al. 2018. In utero CRISPR-mediated therapeutic editing of metabolic genes. Nat Med 24: 1513–1518. 10.1038/s41591-018-0184-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Ruan GX, Barry E, Yu D, Lukason M, Cheng SH, Scaria A. 2017. CRISPR/Cas9-mediated genome editing as a therapeutic approach for Leber congenital amaurosis 10. Mol Ther 25: 331–341. 10.1016/j.ymthe.2016.12.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Russell S, Bennett J, Wellman JA, Chung DC, Yu ZF, Tillman A, Wittes J, Pappas J, Elci O, McCague S, et al. 2017. 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 390: 849–860. 10.1016/S0140-6736(17)31868-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Sengillo JD, Justus S, Cabral T, Tsang SH. 2017. Correction of monogenic and common retinal disorders with gene therapy. Genes (Basel) 8: 53. 10.3390/genes8020053 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Stein-Streilein J, Taylor AW. 2007. An eye's view of T regulatory cells. J Leukoc Biol 81: 593–598. 10.1189/jlb.0606383 [DOI] [PubMed] [Google Scholar]
  61. Suh S, Choi EH, Leinonen H, Foik AT, Newby GA, Yeh WH, Dong Z, Kiser PD, Lyon DC, Liu DR, et al. 2021. Restoration of visual function in adult mice with an inherited retinal disease via adenine base editing. Nat Biomed Eng 5: 169–178. 10.1038/s41551-020-00632-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Tanna P, Strauss RW, Fujinami K, Michaelides M. 2017. Stargardt disease: clinical features, molecular genetics, animal models and therapeutic options. Br J Ophthalmol 101: 25–30. 10.1136/bjophthalmol-2016-308823 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Timmers AM, Newmark JA, Turunen HT, Farivar T, Liu J, Song C, Ye GJ, Pennock S, Gaskin C, Knop DR, et al. 2020. Ocular inflammatory response to intravitreal injection of adeno-associated virus vector: relative contribution of genome and capsid. Hum Gene Ther 31: 80–89. 10.1089/hum.2019.144 [DOI] [PubMed] [Google Scholar]
  64. Trapani I, Auricchio A. 2018. Seeing the light after 25 years of retinal gene therapy. Trends Mol Med 24: 669–681. 10.1016/j.molmed.2018.06.006 [DOI] [PubMed] [Google Scholar]
  65. Trapani I, Puppo A, Auricchio A. 2014. Vector platforms for gene therapy of inherited retinopathies. Prog Retin Eye Res 43: 108–128. 10.1016/j.preteyeres.2014.08.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. van Wijk E, Pennings RJ, te Brinke H, Claassen A, Yntema HG, Hoefsloot LH, Cremers FP, Cremers CW, Kremer H. 2004. Identification of 51 novel exons of the Usher syndrome type 2A (USH2A) gene that encode multiple conserved functional domains and that are mutated in patients with Usher syndrome type II. Am J Hum Genet 74: 738–744. 10.1086/383096 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Verbakel SK, van Huet RAC, Boon CJF, den Hollander AI, Collin RWJ, Klaver CCW, Hoyng CB, Roepman R, Klevering BJ. 2018. Non-syndromic retinitis pigmentosa. Prog Retin Eye Res 66: 157–186. 10.1016/j.preteyeres.2018.03.005 [DOI] [PubMed] [Google Scholar]
  68. Villiger L, Grisch-Chan HM, Lindsay H, Ringnalda F, Pogliano CB, Allegri G, Fingerhut R, Häberle J, Matos J, Robinson MD, et al. 2018. Treatment of a metabolic liver disease by in vivo genome base editing in adult mice. Nat Med 24: 1519–1525. 10.1038/s41591-018-0209-1 [DOI] [PubMed] [Google Scholar]
  69. Wu W, Tsai YT, Huang IW, Cheng CH, Hsu CW, Cui X, Ryu J, Quinn PM, Caruso S, Lin CS, et al. 2022. CRISPR genome surgery in a novel humanized model for autosomal dominant retinitis pigmentosa. Mol Ther 30: 1407–1420. 10.1016/j.ymthe.2022.02.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Yáñez-Muñoz RJ, Balaggan KS, MacNeil A, Howe SJ, Schmidt M, Smith AJ, Buch P, MacLaren RE, Anderson PN, Barker SE, et al. 2006. Effective gene therapy with nonintegrating lentiviral vectors. Nat Med 12: 348–353. 10.1038/nm1365 [DOI] [PubMed] [Google Scholar]
  71. Zhang XH, Tee LY, Wang XG, Huang QS, Yang SH. 2015. Off-target effects in CRISPR/Cas9-mediated genome engineering. Mol Ther Nucleic Acids 4: e264. 10.1038/mtna.2015.37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Zhang W, Li L, Su Q, Gao G, Khanna H. 2018. Gene therapy using a miniCEP290 fragment delays photoreceptor degeneration in a mouse model of Leber congenital amaurosis. Hum Gene Ther 29: 42–50. 10.1089/hum.2017.049 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Zhi S, Chen Y, Wu G, Wen J, Wu J, Liu Q, Li Y, Kang R, Hu S, Wang J, et al. 2022. Dual-AAV delivering split prime editor system for in vivo genome editing. Mol Ther 30: 283–294. 10.1016/j.ymthe.2021.07.011 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cold Spring Harbor Perspectives in Medicine are provided here courtesy of Cold Spring Harbor Laboratory Press

RESOURCES