Open Access
Review  |   June 2023
Gene Therapy for Retinal Degenerative Diseases: Progress, Challenges, and Future Directions
Author Affiliations & Notes
  • Selina Drag
    Ocugen, Inc., Malvern, Pennsylvania, United States
  • Farokh Dotiwala
    Ocugen, Inc., Malvern, Pennsylvania, United States
  • Arun K. Upadhyay
    Ocugen, Inc., Malvern, Pennsylvania, United States
  • Correspondence: Arun K. Upadhyay, Ocugen, Inc., 11 Great Valley Parkway, Malvern, PA 19355, USA; [email protected]
  • Footnotes
     SD and FD contributed equally to this work and are co-first authors.
Investigative Ophthalmology & Visual Science June 2023, Vol.64, 39. doi:https://doi.org/10.1167/iovs.64.7.39
  • Views
  • PDF
  • Share
  • Tools
    • Alerts
      ×
      This feature is available to authenticated users only.
      Sign In or Create an Account ×
    • Get Citation

      Selina Drag, Farokh Dotiwala, Arun K. Upadhyay; Gene Therapy for Retinal Degenerative Diseases: Progress, Challenges, and Future Directions. Invest. Ophthalmol. Vis. Sci. 2023;64(7):39. https://doi.org/10.1167/iovs.64.7.39.

      Download citation file:


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

      ×
  • Supplements
Abstract

Since long before the first approval of gene therapy for retinal disease, ocular gene therapy has captured the hopes of patients, clinicians, and scientists alike. Indeed, the retina provides a unique system for studying and treating ocular diseases, and it holds the distinction as the first tissue targeted by an approved gene therapy for inherited disorders in the United States. There are many methods for addressing genetic diseases in the eyes using a wide range of potential delivery systems and vectors. However, despite the immense progress over the last several decades, both old and new challenges remain, such as the long-term effects of treatments, immunogenicity, targeting, and manufacturing. This review provides a discussion of the history of ocular gene therapy, the various gene therapy approaches, methods to deliver a gene directly to ocular tissues (including both routes of administration and vectors), challenges to ocular gene therapy, the current clinical trial landscape, and future directions of the field.

Genetic diseases were once formidable and devastating because, until recently, they were considered untreatable.1 Gene therapy has provided a unique opportunity to treat and even cure these diseases, offering hope to the millions of people either affected by inherited disorders or carrying disease-causing mutations.2 Its history is rich and complicated, dating back 7 decades to the first observations of viral gene transfer, and has involved great scientific advancements, including improvements in vectors and vector design, and complications, such as serious adverse events and patient deaths.3,4 The earliest gene therapies used several different viral vector-based platforms, and both adenovirus and lentivirus were shown to effectively transduce retinal cells.58 However, 2 clinical studies in the late 1990s suffered serious adverse events: cytokine storms, multiple organ failures, T-cell leukemia, oncogene activation, and even death.3,4 These complications slowed the progress of research and development as researchers looked for answers and solutions to these risks. New technologies continue to provide innovative new treatment platforms, such as the clustered regulatory interspaced palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system and optogenetics.9,10 
Luxturna from Spark Therapeutics, for the treatment of Leber congenital amaurosis (LCA) type 2, showed safety and efficacy in phase I/II clinical trials1113 and was the first US Food and Drug Administration (FDA)-approved gene therapy (2017) for an inherited disorder in the United States.14,15 Yet, despite this landmark achievement, no other ocular gene therapies have been approved in the United States. This may be in part due to the biological challenges to efficient transduction and minimizing immunogenicity or the practical challenges in scaling up manufacturing and reaching the target tissues.1618 Improvements to production methods, administration, and drug design and targeting can all influence the success of gene delivery to the retina in future trials, and ocular gene therapy studies thus far have demonstrated the potential for success.18 
The retina has long been considered an excellent target for gene therapy. It offers advantages, such as an enclosed, immune-privileged site protected by the blood-retina barrier.19 Due to the small size of the retina and a lack of cellular proliferation in adulthood, only small amounts of the vector are required to treat retinal diseases.15 Already established surgical procedures or clinical practices can be used to administer ocular gene therapy products.1,15 Easy real-time ocular monitoring by optical coherence tomography and fundus imaging allows monitoring the drug effects in both animal models and clinical trial participants.15 
Despite these advantages, emerging evidence suggests that sites, such as the eye and central nervous system, once considered immune privileged, are more vulnerable to adverse immune reactions than previously believed.2022 Several studies have observed antidrug antibody responses and inflammation in the eyes due to immune cell infiltration following retinal gene therapy.21,2327 Continuing work in improving vector systems may offer alternatives to the current popular systems for gene delivery.9,2729 
History of Gene Therapy Development for Retinal Diseases
Although the history of gene therapy goes back to the 1950s, retinal gene therapies have been studied only since the 1990s. In 1994, the first attempts to identify a vector for retinal gene delivery occurred, using adenovirus-based systems in mice.5,6 These studies showed efficient transduction of the retinal pigmented epithelium (RPE) via subretinal injection; however, photoreceptor transduction was refractory and required higher treatment doses. In 1996, Bennett et al. went on to show successful gene therapy using an adenoviral vector in the rd1 mouse model of recessive retinal degeneration.30 Around that same time, other groups used an HIV-based lentiviral or adeno-associated virus (AAV)-based vectors to demonstrate efficient photoreceptor and RPE transduction.7,8 These studies also noted that younger animals treated before the onset of degeneration demonstrated improved gene transfer to photoreceptors.15 
In 1999, gene therapy in general suffered a setback due to the tragic death of the gene therapy patient, Jesse Gelsinger, who was enrolled in a gene therapy trial to treat ornithine transcarbamylase deficiency. Later, 5 of 20 patients with severe combined immunodeficiency (SCID)-X1, treated with retrovirus gene therapy, developed T-cell leukemia within 2 to 6 years following treatment.3,4 These tragedies slowed gene therapy development as researchers, clinicians, and regulators grappled with the risks associated with this technology. Despite the serious setbacks in these clinical trials, advancements continued, with the first gene therapy products Gendicine, Oncorine, and Cerepro being approved for clinical use.4,31 An overview of the history of retinal gene therapy is provided in Figure 1
Figure 1.
 
History of the development of retinal gene therapy (created with BioRender.com).
Figure 1.
 
History of the development of retinal gene therapy (created with BioRender.com).
Gene Therapy Approaches for Retinal Diseases
Retinal gene therapy approaches vary based on the nature of the mutation (Fig. 2Table 1) and maybe gene replacement/augmentation, silencing/editing the mutated gene, or supplying a gene that affects the upstream or downstream pathways from the defective gene to improve cellular function (as in modifier therapy). Retinal gene therapies include the use of diverse vectors (Table 2) and routes of administration (Table 3). 
Figure 2.
 
Gene therapy approaches. There are four major approaches to gene therapy (left to right). Gene replacement or augmentation is when a functional copy of a damaged, non-functional gene is added to augment the production of functional protein. In gene editing, mutations in a gene are corrected or expression of the mutated protein is reduced to alter a diseased state. Gene silencing uses the RNAi mechanism to eliminate the aberrant expression of the targeted pathogenic protein in acquired diseases. Modifier gene therapies provide a modifier gene that can affect pathways downstream or upstream from the damaged gene, allowing for the expression of multiple genes to be altered with a single treatment (created with BioRender.com).
Figure 2.
 
Gene therapy approaches. There are four major approaches to gene therapy (left to right). Gene replacement or augmentation is when a functional copy of a damaged, non-functional gene is added to augment the production of functional protein. In gene editing, mutations in a gene are corrected or expression of the mutated protein is reduced to alter a diseased state. Gene silencing uses the RNAi mechanism to eliminate the aberrant expression of the targeted pathogenic protein in acquired diseases. Modifier gene therapies provide a modifier gene that can affect pathways downstream or upstream from the damaged gene, allowing for the expression of multiple genes to be altered with a single treatment (created with BioRender.com).
Table 1.
 
Gene Therapy Approaches
Table 1.
 
Gene Therapy Approaches
Gene Replacement Therapy
The gene replacement is a direct approach that supplies a functional copy of a damaged or nonfunctional gene to augment the production of functional protein. This therapy addresses the missing role of the damaged gene without changing it. Gene replacement is best suited for monogenic recessive inherited diseases, for example, mutations in the CEP290 gene, is one of the most common causes (15–20%) of LCA.32 Luxturna (Spark Therapeutics, Inc.) provides a functional RPE65 gene to patients with LCA with RPE65 mutations (5–10%). However, despite the apparent success of Luxturna in the clinic, there has been some question regarding its durability of effect and effectiveness across patients with different genetic backgrounds, as well as the limitations, such as a need for remaining tissue prior to treatment and deciding how to quantify the effects of treatment.33 Several other products using gene replacement therapy are also in clinical trials (Table 4). 
Retinal diseases are caused by a wide range of mutations in many different genes with varied inheritance patterns, such as autosomal dominant, autosomal recessive, and X-linked. For example, in retinitis pigmentosa (RP), more than 3000 mutations in approximately 70 genes have been implicated in disease pathogenesis, and, in many cases of RP, a genetic basis cannot be identified.34 In addition, not all gene mutations can be addressed using a gene replacement approach, including dominant mutations, large genes which cannot be packaged in currently utilized delivery vectors, and polygenic conditions. Gene replacement for each mutation would prove costly and limit patient access. 
Gene Silencing
Gene silencing with small interfering RNA (siRNA) or microRNA (miRNA) that target VEGF, are currently in development for the treatment of age-related macular degeneration (AMD), glaucoma, and several other ocular diseases.3538 Several clinical trials using targeted gene silencing techniques are in progress (Table 4).3941 However, no clinical trials for this approach in the ocular space have progressed past phase III, as this technique faces several challenges, such as RNA instability, poor bioavailability, and nonspecific targeting leading to off-target effects.35 
Gene Editing
In gene editing, mutations in a gene are corrected or the expression of the mutated protein is reduced to alter a diseased state. Several gene editing techniques have been developed, including CRISPR/ Cas9, transcription activator-like effector nucleases (TALENs), zinc finger nucleases (ZFNs), and homing endonucleases or meganucleases.42 Of these, the most well-known gene editing technique is the CRISPR/Cas9 system has shown potential in gene therapy. CRISPR/Cas9 is a two-component system that involves a guide RNA specific to the gene of interest and an endonuclease that induces a site specific double-stranded DNA to allow for genetic modification.9 This allows for a permanent and precise modification or removal of the mutation associated with a particular disease.43 While addressing mutations in a single gene, CRISPR is not effective to patients without a known genetic diagnosis. 
Gene therapies using CRISPR/Cas9 technology are in clinical trials (Table 4). In the retinal space, EDIT-101 (Editas Medicine, NCT03872479) is a gene therapy to treat LCA type 10, targeting the IVS26 mutation that causes improper splicing between exons 26 and 27 leading to a prematurely truncated, nonfunctional CEP290 gene. This treatment uses CRISPR/Cas9 to cause deletions or inversions of the IVS26 mutation both of which restore correct splicing and CEP290 function.4345 Prime editing (PE) is an upcoming technique that uses reverse transcriptase and Cas9 to repair genome mutations with the potential to correct multiple mutations as well as small insertions and deletions.43 However, although this is a promising technique for the future treatment of genetic disorders, it still faces some of the same challenges as traditional gene therapy, particularly the sheer volume of genes involved in disease pathogenesis. 
Modifier Gene Therapy
The existence of modifier genes, or genes that may affect the expression of other genes, particularly mutant genes, without affecting healthy phenotypes, has been known since the early 1940s,46 and development of therapies to apply these modifier functions to disease has grown rapidly. The severity of the phenotype caused by some gene mutations can be influenced by these modifier genes.4749 Modifier genes can affect pathways downstream or upstream from multiple defective genes, thereby addressing a clinical phenotype without the need for a genetic diagnosis and in a mutation-agnostic manner. Modifier therapies rely heavily on identification of modifier genes involved in a particular disease phenotype, which may prove challenging and possibly costly.46 
Development of modifier gene therapies has expanded rapidly for systemic diseases, including neuromuscular disease, cystic fibrosis, spinal muscular atrophy, and cancer, as well as for retinal disease. Previous studies have shown that modifier genes, such as nuclear hormone receptors, have the potential to “reset” various networks related to retinal disease phenotypes, such as photoreceptor development, phototransduction, metabolism, cone cell development, inflammation, and cell survival, thus restoring the homeostasis in the retina to a healthy state.47,5053 One modifier gene therapy, OCU400 (Ocugen, Inc.), is a nuclear hormone receptor-based gene therapy currently in clinical trials to treat retinitis pigmentosa (NCT05203939). This gene agnostic approach has the potential to significantly reduce the need to develop a product for every mutation, thus reducing development and commercialization costs, while expanding the patient reach to address this unmet medical need in rare genetic disease spaces. 
Gene Delivery Approaches in the Retinal Space
Gene delivery to the retina involves several factors, such as the optimal route of administration, the size of the gene, the immunogenicity and specificity of the vector, and manufacturing complications, including the cost and the ability to scale up production. Various viral and non-viral vectors provide multiple options (Fig. 3, Table 2).15 
Figure 3.
 
Gene Therapy Vectors may be Either Viral or Non-Viral, Each With its own Advantages and Risks (Created With BioRender.com).
Figure 3.
 
Gene Therapy Vectors may be Either Viral or Non-Viral, Each With its own Advantages and Risks (Created With BioRender.com).
Table 2.
 
Gene Therapy Vectors
Table 2.
 
Gene Therapy Vectors
Viral Vector Delivery
Adenoviral vectors derived from the human Ad2 and Ad5 serotypes were some of the first viral delivery systems to be used in retinal gene therapy research.5,29 These viruses show good safety when administered to target tissues, do not integrate in the recipient genome, successfully transduce retinal cells, and can carry genes of around 30 kb long.4,29,5456 However, these vectors tend to be rapidly cleared due to the presence of neutralizing antibodies from pre-existing immunity.4,29,5456 Second generation adenovirus vectors have early gene regions (E2a, E2b, or E4) deleted to reduce viral replication and immunogenicity in the host cells.29,5761 Despite these advances, adenovirus vectors are typically not used for retinal gene therapy and targeting retinoblastoma with oncolytic adenovirus VCN-01is the only Ad vectored clinical trial in the ocular field.62,63 The AAV vectors are a common vector choice in ocular and non-ocular gene therapies, offering benefits such as a long duration of transgene expression, extremely low risk of insertional mutagenesis, only a mild inflammatory response, and low possibility of germline transmission.29,54 AAV vectors offer a high number of tissue-specific serotypes, including retina-specific serotypes AAV1, 2 (Luxturna), 4, 5, 6, 7, 8, and 9,64 for the treatment of LCA, is based on an AAV2 vector system, and AAV4 and AAV5 are also commonly used for retinal gene therapy due to their specificity to retina and RPE in animal models.14,65 Serotypes may also influence which cell types are transduced, for example, all the retina-specific serotypes are known to transduce RPE, their ability to transduce photoreceptors varies by AAV.64 Recombinant AAV (rAAV) vectors have been developed to combine desirable tropisms from multiple serotypes,66 or to reduce immune responses or antibody neutralization.29 Recent developments use dual/multiple vector strategy29,67 that bypasses the 4.7 kb maximum size restriction for AAVs utilizing inteins and exteins to join multiple peptide products into the large functional protein in the host cells.6872 There are four notable approvals of gene therapy products using AAV vector systems: Glybera, Luxturna, Zolgensma, and Hemgenix of which only Luxturna (voretigene neparvovec-rzyl), is a retinal gene therapy product to treat LCA. Retroviruses and lentiviruses have been used in several gene therapy products, specifically RetinoStat (Oxford BioMedica, OXB-201) against wet (neovascular)- AMD (NCT01301443), and in stem cell therapy.3 However, they carry risks for insertional mutagenesis and germline transmission and may elicit more inflammatory responses than AAVs.54,56,73 
Non-Viral Delivery
Non-viral gene delivery has been studied less extensively due to questionable transduction efficiency, durability of effect, and ability to reach the therapeutic expression levels. The simplest of the non-viral methods is physical delivery, such as the injection of naked plasmid DNA, siRNA, mRNA, or miRNA,74 which shows limited uptake due to quick degradation.75 Aptamer-based therapies, such as Macugen (pegaptanib; Eyetech/Pfizer) that received approval for clinical use have since fallen out of favor as other, more effective therapies reached the market.76,77 Zimura (avacincaptad pegol; IVERIC bio Inc.), a complement C5 protein inhibitor is in clinical trials for the treatment of geographic atrophy (NCT04435366 and NCT05536297) and Stargardt disease 1 (NCT03364153).77 Other potential methods have included the use of electroporation, gene guns, ultrasounds, and magnetofection, each using physical methods to deliver the target genes to the appropriate locations.75 Additionally, modifications of RNA treatments during production, such as use of chemical methods of delivery, can greatly increase their longevity.78 
Chemical methods of non-viral gene delivery are appealing for their reduced immunogenicity, ease of scaling, reduced expenses for production, and increased payload size.79 Inorganic nanoparticles may include metals such as iron or gold, inorganic cations including magnesium or calcium ions, or ceramics such as phosphate or carbonate salts.75,80,81 Single-molecule DNA nanoparticles compacted by polyethylene glycol (PEG)-substituted lysine peptides (CK30-PEG) have been used to carry payloads up to 20 kb in size,82,83 with several groups demonstrating their safety in BALB/c mice and rds+/− Rho-/- or Abca4-/- mouse models of retinal degeneration.8286 Similarly, lipid-based transfection systems have shown success in their ability to transfer the target genes into retinal cells in several studies, and multiple lipid-based drugs for eye diseases are already available in the clinic35,79,8791 and for the delivery of CRISPR or ribonucleoproteins for base editing.92,93 Niosomes, which are comprised of uncharged single-chain surfactant and cholesterol94,95 have also shown limited success potential as non-viral gene delivery systems96101 due to improved transfection efficiency, biocompatibility, and cellular uptake, in addition to effects on intracellular trafficking.102 Polymer systems, such as chitosan, hyaluronic acid, polyethylenimine (PEI), poly(amidoamine) (PAMAM), PEG, poly (lactic-co-glycolic acid) (PLGA), and poly(L-lysine) (PLL), have been studied in ocular gene therapy.35,75 
Gene Therapy Challenges
Some of the very traits that make the retina an ideal candidate for gene therapies (i.e. immune privilege) can also introduce additional challenges to gene delivery, such as identification of the disease-causing gene or mutation, ensuring targeted delivery of the product, appropriate route of administration, feasibility in the clinic, and immune responses to the product that may exacerbate already fragile tissue. Simply delivering the product to the desired tissue can be a physical challenge, both due to the isolated nature of the eyes and the delicate nature of diseased tissue. These challenges remain for all products and clinical trials, and several different approaches have been developed to address them. 
Identification of Disease-Causing Genes
One of the biggest problems facing traditional gene therapy and the gene editing approach is their reliance on accurate genetic diagnosis and the prohibitive cost of generating gene-specific therapies. In many inherited retinal diseases, such as RP, hundreds of mutations in many genes may lead to the same clinical phenotypes, limiting the use of a single gene therapy product to only those patients with a confirmed genetic diagnosis in that gene. However, many patients lack a genetic diagnosis altogether; the gene or genes responsible for their disease phenotype have not been identified, removing the possibility of a traditional gene therapy approach. 
Mutation Agnostic Approaches to Gene Therapy
Modifier gene therapy has the potential to alter the retinal disease state even in the absence of a genetic diagnosis or in the absence of a gene therapy specific to the mutated gene in one of these patients.47,5053 These therapies affect various networks related to retinal disease phenotypes rather than directly relying on replacing or modifying the diseased gene and have the potential to “reset” these networks to restore homeostasis of a healthy retina. As with other gene therapies, vector selection and route of administration can influence the efficiency of transduction and effectiveness of the therapy, as well as influencing any immune responses, and so careful design of delivery is just as important using this model while offering a wider potential for treatment. 
In addition to modifier therapy and gene editing, therapies that focus on protection of degenerating photoreceptors and RPE cells do not rely on knowledge of a disease-causing genetic mutation. Neuroprotective factors that support the survival of retinal cells, such as glial cell-derived neurotrophic factor (GDNF), ciliary neurotrophic factor (CNTF), brain-derived neurotrophic factor (BDNF), basic fibroblast growth factor, and pigment epithelium-derived factor (PEDF) have shown promise in mouse models of retinal degeneration.67,103105 Although repeated administration of these factors directly may lead to inflammation and patient compliance issues, a stable expression of these factors in the retina using a gene therapy approach may offer a similar advantage as modifier therapy in promoting a healthy state in the retina.67 Supporting the promise of this method, AAV delivery of BDNF and its receptor105 or CNTF106 promoted survival and function of retinal ganglion cells in models of optic nerve crush or retinal ganglion cell degeneration in a model for experimental glaucoma, respectively. Similarly, PEDF offers anti-angiogenic and anti-inflammatory effects that may be useful in wet AMD, and a combination treatment of AAV-delivered PEDF with a microRNA to inhibit VEGF successfully reduced choroidal neovascularization in a mouse model of AMD.107 MicroRNAs themselves have similar influences as neuroprotective factors on protection of photoreceptors and pathogenesis of retinal disease, and several different microRNAs have been tested in rodent models for their ability to regulate retinal physiology in models for diseases such as AMD.67,107109 
Optogenetics, a tool that involves the delivery of light-sensitive microbial opsins to retinal cells, may be particularly useful in advanced cases of retinal disease in which photoreceptors have severely degenerated. It has already shown promise in preclinical rodent and nonhuman primate (NHP) models, as well as advancing to the clinical trial stage for several products, and provides new photosensitive genes, such as channel rhodopsin, halorhodopsin, and melanopsin, to existing neural networks.67 However, optogenetics still require optimization to allow for complex visual processing and to increase the sensitivity of the proteins that are currently used.10,15 Despite this challenge, optogenetic gene therapies are currently in clinical trials (Table 4). 
Gain-of-Function Mutations
Another difficult challenge is addressing toxic gain-of-function mutations, as observed in autosomal dominant RP and mutations to RHO. Similarly, a dominant negative mutation can lead to toxic loss of function. In cases of these toxic gain or loss of function, additional gene silencing methods can been attempted, including the use of short hairpin RNAs and allele-specific ribozymes, but this would lead to complete silencing of the gene; the challenge remains to provide a version of the gene that is not susceptible to the silencing treatment.15 Gene editing techniques, such as the CRISPR/Cas9 systems, could also provide solutions to these mutations. 
Effective Targeting
One of the greatest challenges to gene therapy is ensuring the product reaches the tissue or cells of interest. For example, the blood-retina barrier (BRB) can prevent a gene therapy given systemically from reaching the retina. Additionally, the gene therapy product must be able to transduce the diseased cells themselves. The use of specific vectors or cell-specific promoters can increase the efficiency of targeted interactions, and directly administering the product to the target tissue can increase its effectiveness. 
Vector Selection and Targeted Engineering
Adenoviruses and AAVs are known to have tissue-specific tropisms that make certain serotypes attractive for development of retinal gene therapy products.14,29,56 Due to their tissue-specific tropisms, some of the most common AAV serotypes in use for retinal gene therapy include AAV2, AAV4, AAV5, and AAV8, as mentioned previously.26,110 The use of recombinant or engineered vector capsids can further increase tissue-specific targeting by allowing more precise control over the various elements in a capsid, and vector modifications. Selection of cell-type-specific promoters, further refines targeting to the affected cell types.29,111 
RPE cells can be targeted using AAV1, AAV4, or AAV6 vectors or with the engineered AAV2-7m8 vector, and photoreceptors and Müller glial cells are targeted by serotypes AAV2, AAV5, AAV6, AAV7, AAV8, and AAV9 as well as engineered AAV2-7m8 and AAV8BP2 vectors.56,112116 One group recently used a promoter engineering approach to create an extensive library of 230 AAVs that each contained a different synthetic promoter designed specifically to target cell types.111 
Route of Delivery
Once the appropriate vector design has been determined, researchers still have the challenge of finding the optimal route to deliver the gene therapy to the tissue. Gene therapies can be delivered to the retina by ocular or systemic routes (Fig. 4, Table 3), although generally it is preferred to use a route that will bring the therapy as close to the target tissue as possible, such as subretinal injections for outer retina targets and intravitreal injections for inner retina targets.56 Systemic administration has the advantage of convenience, but the lack of specific targeting can lead to nonspecific effects in non-ocular tissues, reduced bioavailability in the target tissue, and an increase in the risk of immunogenicity as more of the body is exposed to the therapy.35 Ocular administration restricts a gene therapy's effect to the target tissue and reduces immunogenicity. Invasive ocular delivery methods offer more targeted delivery and therefore increase the bioavailability of drug products, but these methods increase the risk of complications, such as infection, retinal detachment, and hemorrhage. These procedures typically require skilled and experienced surgeons for successful administration.117 Noninvasive delivery methods offer drug delivery with fewer procedure-related complications and easier administration but with reduced bioavailability.35,118 Therefore, determining the optimal route of delivery for a specific gene therapy product is critical for efficient transduction and minimized immune response in patients. 
Figure 4.
 
Ocular routes of administration may be invasive or non-invasive (created with BioRender.com).
Figure 4.
 
Ocular routes of administration may be invasive or non-invasive (created with BioRender.com).
Table 3.
 
Ocular Routes of Administration
Table 3.
 
Ocular Routes of Administration
Noninvasive Delivery
Noninvasive delivery methods include topical administration, such as eye drops, iontophoresis, ultrasonics, transdermal systems, and contact lenses. These methods avoid the risk of surgically induced damage to parts of the eye but may also reduce the bioavailability of the therapeutic. Of the noninvasive methods, topical administration is the most used.35,118 This method avoids complications of first-pass metabolism and has the advantage of easy administration, sometimes by the patients themselves. However, barriers, such as the pre-corneal tear film, the structure of the cornea, limited volume, lacrimal drainage system, reflex tearing, and aqueous outflow of the eye, reduce the bioavailability of the product to around 5% or less.35,118,119 Iontophoresis uses a low electrical current to allow easier penetration of ionic drugs into tissues. Placement of the electrode can target different regions of the eye but care should be taken as higher currents can cause tissue damage.35,119121 Ultrasonic devices use a sound field above 20 kHz to improve penetration of a product through skin and eyes, whereas transdermal patches use controlled longer release that may be beneficial for chronic ocular disease.35,119 Finally, contact lenses may be soaked in a drug solution before application to the eyes, thus serving as an alternative to eye drops and improving the sustained release of transdermal patches. Contact lenses tend to have better compliance and fewer systemic side effects than other methods, but they are limited by the solubility of the product, the time needed to soak the lens, and the amount of drug that is discarded following soaking.35,122 
Uncommon Invasive Routes of Delivery for Gene Therapy
Some invasive delivery methods less commonly used in gene therapy include ocular implants, subtenon, and subconjunctival injections.35,118 Subtenon injections have been used to treat diabetic macular edema and surgery-related choroidal detachments, but they are occasionally associated with increases in intraocular pressure.123125 Subconjunctival injections are dependent on the size of the particles. Although they demonstrate improved bioavailability over topical administration, systemic absorption may occur.35 Biodegradable and non-biodegradable ocular implants have been used in several drug treatments for conditions such as diabetic macular edema and posterior uveitis, among others, and provide a method for sustained release of poorly soluble steroids.118,126 
Subretinal Injection
Subretinal drug is administered between the retinal photoreceptor cells and the RPE layer, requires less vector than intravitreal injections to achieve a therapeutic effect on the retina.127 However, this route of administration can further damage an already damaged retina, leading to retinal or RPE detachment from underlying layers, hemorrhage, and changes in retinal pigmentation.35,127,128 Injections affecting the thinner and more fragile fovea carry greater risk than other retinal regions.15 However, subretinal injections have not been associated with severe inflammation following treatment with AAV8, AAV2, or AAV5, increasing its appeal as a route of administration.19,26 The use of corticosteroids is typical to reduce procedure-related inflammation and immune responses to the vector.27 Targeted engineering of vector capsids may also alleviate inflammation and the use of robotic devices may alleviate the drift and error of human surgeons in the operating room.19 
Intravitreal Injection
Intravitreal injections are the least invasive of these surgical administration methods for gene therapy products targeting the inner retina. Anti-VEGF therapies for AMD and diabetic retinopathy typically use intravitreal injections, for the repeated injections. Despite these advantages, intravitreal injections also carry risks of adverse reactions, including inflammation, endophthalmitis, increased intraocular pressure, retinal detachment, hemorrhage, and cataracts, and due to the repeated use of these injections in various treatments, patient compliance can become an issue.119,127,129 Most gene therapy viral vectors given intravitreally, fail to adequately transduce photoreceptors and RPE, due to the inner limiting membrane that divides the vitreous from the retina.1,15,119,127 However, development of recombinant vectors AAV2.GL and AAV2.NN improve the retinal delivery of a gene therapy product after intravitreal injection.56,66 Increased non-ocular biodistribution of ranibizumab or bevacizumab have been observed with the intravitreal route causing systemic adverse events like ecchymosis, gastrointestinal and vaginal hemorrhage, hematoma, increased blood pressure, cerebrovascular accident, myocardial infarctions, iliac artery aneurysm, and death.26,129 Intravitreal injection is also known to be more immunogenic than subretinal injection.27 Adverum Biotechnologies terminated the development of ADVM-022, an intravitreal AAV-based aflibercept gene therapy for AMD and DME due to dose-limiting toxicity in multiple patients.130 
Suprachoroidal Injection
Suprachoroidal injections, administered between the choroid and sclera, do not require vitreoretinal surgeries, and show better bioavailability than intravitreal injections.1,131,132 This injection requires a skilled surgeon and must be performed in an operating room. Procedure-related adverse events include suprachoroidal hemorrhage, endophthalmitis, choroidal tears, changes in choroidal blood flow, inflammation, and retinal detachment.131 
Immunogenicity and Retinal Toxicity
Despite many advancements in the overall safety profiles and quality assurance of gene therapy products, there remain concerns regarding the safety of gene therapy products, particularly at high doses. Although there have been safety issues observed in gene therapy trials outside the eyes, few serious adverse events directly related to recombinant AAV gene therapies have been reported in ocular gene therapy trials.26 The combination of the procedure and the retinal gene therapy drug carries the potential for toxicity to the retina, which is already fragile due to the disease.128,133 Adverse events include inflammatory responses, increases in intraocular pressure, loss of retinal layers, decrease in electroretinography (ERG) amplitudes, antidrug antibody (ADA) responses to vector, and toxicity in the photoreceptors and RPE layers.23,26,134138 Elevated immune responses due to defects in blood-tissue barriers lead to production of neutralizing antibodies that limit the effectiveness of gene therapy.133,139 Adenoviral vectors tend to provoke rapid neutralizing antibody and immunogenic responses, but these responses are reduced with improved targeting of the tissues of interest.4 Vector sequences like CpG sequences, and CMV and CAG promoters can also lead to inflammatory immune reactions, which can trigger the activation of microglia in the retina and may exacerbate degeneration.26,140 Immunogenicity against the viral vector that limits gene therapy efficacy is a major concern.4,21,25,26,141 and is one of the biggest exclusion criteria in clinical trials.14,26 In addition to neutralizing antibodies, cell based immune responses have often been observed at higher doses (above 1 × 1011 vg) in many ocular gene therapy trials.25,26 Future products will need to optimize the balance between the effective dose level and the potential for toxic effects. However, Luxturna's success demonstrates that successful gene therapy for retinal disease is possible. 
In the clinical setting, screening patients for presence of neutralizing antibodies for the gene therapy vector can improve outcomes.14,26,142 Alterations to the product itself, by changing immunogenic sequences, would alter how a product will interact with immune cells in vivo. Assessments of immunogenicity both before and after use in a clinical setting, must be continuously monitored and143 modulating CD4+ T cell and B cell responses may assist in reducing ADAs.144 For example, immature dendritic cells (iDCs) are antigen presenting cells (APCs) that often present self-antigens without stimulating T cell responses, which may allow for tolerance of peptide sequences that could be harnessed in biotherapeutic treatments.143145 Predicting and removing epitope sequences that can stimulate T or B cells offers another opportunity to reduce immunogenicity.144,145 Epitopes can also be shielded by polymers, such as PEG, XTEN, or PAS, or by methylating, glycosylating, or bio conjugating biotherapeutics, to effectively hide the surface epitopes of the product from the immune system or inducing tolerance to the antigen.145,146 
Besides altering the product directly, co-treatment with anti-inflammatory or anti-NAb substances may reduce the immune response to a gene therapy. Minocycline treatment has been shown to reduce inflammatory cytokine expression in the retina.147 Administration of the IgG-cleaving endopeptidase imlifidase (IdeS) or co-administration of a synthetic vaccine particle containing rapamycin with AAV treatment have both been shown to reduce immune cell activation and decrease titers of anti-AAV antibodies.148,149 Corticosteroid treatments are also often used to minimize the effects of inflammation and cytokine expression that occur because of administration procedures, reducing the immune response in the ocular space.150,151 Future gene therapy vectors and clinical studies may rely on one of these methods or a combination of approaches to improve the safety profile and efficacy of a product by reducing or eliminating immunogenicity, making safe and effective treatments with few adverse effects possible. 
Animal Model Selection
Rodent models are often appealing for proof-of-concept and efficacy studies, as many of the diseases under study have genetic rodent models available. However, there are other cases in which genetic models are not available or may not fully reflect a disease, such as the physical damage models sometimes used for AMD studies. These models may include laser ablation of Bruch's membrane, as in the choroidal neovascularization model in rats, or oxygen-induced retinopathy models. Although these models mimic some hallmarks of the disease, they do not fully capture human pathophysiology and viral vector serotype affinity for certain tissues may not translate well from small mammal models to NHPs or humans.14 The closest model to a human is NHP, which has eye anatomy and physiology like humans and may provide the closest response to what could be expected in the clinic.152 However, the use of NHP models may have serious concerns, including ethical issues, limited availability of animals, and the high cost of performing NHP studies. Several studies in the Luxturna trials failed to show the same effects in humans as had been observed in animal models, such as a lack of improvement in full field electroretinograms in humans that had been seen in dogs and a decline in vision improvement that had not been observed in the animal models.15 Researchers must consider how closely the model resembles the human counterpart in anatomy (such as size, volume, and critical structures including the macula), physiology, and sequence homology between the target human protein of interest and the model's homolog, which can impact factors such as immunogenicity. 
Manufacture of Gene Therapy Products
Beyond efficacy and safety concerns, gene therapy products have several challenges in manufacturing and production cost of the final clinical products. Although AAVs are most widely used vectors in retinal gene therapy, they remain difficult and costly to produce.1618 Production methods are difficult to scale from the bench to a commercial size, often with lower yields and efficiency.17 Manufacturing methods face complications, such as ensuring product potency, purity, and quality. There are considerable challenges in characterization testing, such as separating empty and full capsids.153 Aside from manufacturing challenges, AAV gene therapy products also face challenges in clinical applications. Although quite stable, AAVs often must be stored at -80°C for long term. Viral vectors may need to be diluted to the required dosage at the clinical site, requiring the facility to have the appropriate biological safety cabinets systems in place. Stability of gene therapy products must be closely monitored, as once the product is thawed, there is limited time for clinicians to use it. 
Ongoing Gene Therapy Clinical Development and Approved Gene Therapy for Retinal Diseases
Luxturna (voretigene neparvovec-rzyl; Spark Therapeutics, Inc.) was the first gene therapy approved in the United States to treat biallelic RPE65 mutation-associated retinal dystrophy. Since this first approval, 24 additional cell and gene therapies have been approved by the FDA for other non-retinal diseases (fda.gov). Presently, there are more than 40 active clinical trials or follow ups for ocular diseases using AAV vectors alone.154 In the United States, over 1000 gene therapy studies are active for a wide range of diseases and conditions. Of these, more than 30 have indications for retinal diseases and are either actively recruiting or enrolling by invitation (Table 4). Many of the gene therapy trials in progress use gene replacement platforms with AAV vectors. However, more studies are considering the use of CRISPR/Cas9 systems, such as Editas Medicine, which is targeting LCA10 caused by CEP290 mutations, or modifier therapy, such as Ocugen, Inc., which has a product (OCU400) in clinical trials for retinitis pigmentosa associated with mutations in NR2E3, RHO, and CEP290 genes. The OCU400 modifier gene product has the potential to address multiple genetic mutations associated with RP and LCA and could be a potential mutation agnostic therapy if it demonstrates success in human clinical trials. 
Table 4.
 
Summary of Active Gene Therapy Clinical Studies with Ocular Disease Indications
Table 4.
 
Summary of Active Gene Therapy Clinical Studies with Ocular Disease Indications
The Future of Gene Therapy
Over the last 25 years, great strides have been made in retinal gene therapy, and each new development leads toward improved products with better safety and efficacy. However, there is still a significant need for these products to reduce the high healthcare costs, economic burden, and costs to affected individuals and families and to improve patient well-being and quality of life.155 As the field continues to grow, research and drug developers are likely to focus on ways to improve the vector design to improve efficiency, open additional vector targets, and increase overall product safety. Additionally, standardized methods for manufacturing may reduce variability of products between batches which would also be likely to improve safety and efficacy. 
An important consideration for future gene therapy product development is finding and implementing methods to treat genetic disease in the absence of a genetic diagnosis or to treat multiple mutations with a single product. These methods would address two persistent problems in gene therapy: the high cost of production and the need to identify the genetic basis of a disease from individual to individual. A product to address both issues would allow for greater distribution among patient populations and improved cost to drug developers. Identification of mutation agnostic modifier genes and protective factors that may modulate the various phenotypes of genetic retinal diseases and continued development of optogenetic technology will open new avenues for treatment across a wide range of conditions. 
Improvements to vector design are also likely to focus on aspects such as reducing immunogenicity, improving target specificity, and improving transduction efficiency. Although AAVs have been the vector of choice in recent years, less immunogenic options, such as engineered viral vector capsids, lipid-based nanoparticles or polymer systems, may prove important to future vector design. Recombinant vectors already use some of these modifications, and future products are likely to continue this trend. Future gene therapies may also find ways around the payload size limitations of vectors, such as use of non-viral techniques or dual or triple transduction strategies. 
As gene therapy grows in use, availability of appropriate materials for viral vector purification and harvest will need to increase. In addition, standardized methods will continue to develop and become accepted across manufacturers to improve product quality and cost-effectiveness. Some of the current limitations for product quality and efficiency are being addressed as new technologies emerge, such as a shift from traditional quantitative PCR (qPCR) methods to droplet digital PCR (ddPCR) methods during in-process and release testing, changes in vector production strategies from a standard triple plasmid transfection model to a plasmid-free production system, such as tetracycline-enabled self-silencing adenovirus (TESSA), that may reduce the possibility of in-process contaminants, and changes from batch production to a perfusion-based continuous production process to improve yield. Changes in the vectors of choice may also require shifts in acceptable manufacturing practices and adaptation of existing technology. 
With the advances that have so far been made in retinal gene therapy and the progress likely to be made in the coming years, retinal therapy remains a powerful tool for the greater gene therapy field, providing innovative solutions to complex problems and modeling gene therapy in a closed system. Success in treating retinal diseases, such as the approval of Luxturna, offers hope for technology, bringing a possibility of treatment, improved quality of life, and even a cure to thousands of patients worldwide. Regardless of the research yet to be done, it is a focus on the well-being and outcomes of patients that will continue to drive the many improvements yet to be made, and both researchers and drug developers should keep this in mind as they move the field forward. 
Acknowledgments
This research did not receive specific funding but was performed as part of the employment of the authors by Ocugen, Inc. Figures were generated using BioRender.com. 
Author Contributions: S.D. wrote the manuscript, did the research, and generated tables and figures. F.D. and A.U. provided critical feedback and editing for the manuscript. All authors made substantial contributions in the article preparation at the following stages: (1) the conception and design of the manuscript and interpretation of data, (2) drafting and revising the article for important intellectual content, and (3) final approval of the version to be submitted. Figures were generated using licensed BioRender.com software. 
Disclosure: S. Drag, Ocugen, Inc. (E, I); F. Dotiwala, Ocugen, Inc. (E, I); A.K. Upadhyay, Ocugen, Inc. (E, I) 
References
Tan TE, Fenner BJ, Barathi VA, et al. Gene-based therapeutics for acquired retinal disease: opportunities and progress. Front Genet. 2021; 12: 795010. [CrossRef] [PubMed]
Hanany M, Rivolta C, Sharon D. Worldwide carrier frequency and genetic prevalence of autosomal recessive inherited retinal diseases. Proc Natl Acad Sci USA. 2020; 117: 2710–2716. [CrossRef] [PubMed]
Tamura R, Toda M. Historic overview of genetic engineering technologies for human gene therapy. Neurol Med Chir (Tokyo). 2020; 60: 483–491. [CrossRef] [PubMed]
Wirth T, Parker N, Yla-Herttuala S. History of gene therapy. Gene. 2013; 525: 162–169. [CrossRef] [PubMed]
Bennett J, Wilson J, Sun D, Forbes B, Maguire A. Adenovirus vector-mediated in vivo gene transfer into adult murine retina. Invest Ophthalmol Vis Sci. 1994; 35: 2535–2542. [PubMed]
Li T, Adamian M, Roof DJ, et al. In vivo transfer of a reporter gene to the retina mediated by an adenoviral vector. Invest Ophthalmol Vis Sci. 1994; 35: 2543–2549. [PubMed]
Ali RR, Reichel MB, Thrasher AJ, et al. Gene transfer into the mouse retina mediated by an adeno-associated viral vector. Hum Mol Genet. 1996; 5: 591–594. [CrossRef] [PubMed]
Miyoshi H, Takahashi M, Gage FH, Verma IM. Stable and efficient gene transfer into the retina using an HIV-based lentiviral vector. Proc Natl Acad Sci USA. 1997; 94: 10319–10323. [CrossRef] [PubMed]
Redman M, King A, Watson C, King D. What is CRISPR/Cas9? Arch Dis Child Educ Pract Ed. 2016; 101: 213–215. [CrossRef] [PubMed]
Prosseda PP, Tran M, Kowal T, Wang B, Sun Y. Advances in ophthalmic optogenetics: approaches and applications. Biomolecules. 2022; 12: 269. [CrossRef] [PubMed]
Maguire AM, Russell S, Chung DC, et al. Durability of voretigene neparvovec for biallelic RPE65-mediated inherited retinal disease: phase 3 results at 3 and 4 years. Ophthalmology. 2021; 128: 1460–1468. [CrossRef] [PubMed]
Maguire AM, Russell S, Wellman JA, et al. Efficacy, safety, and durability of voretigene neparvovec-rzyl in RPE65 mutation-associated inherited retinal dystrophy: results of phase 1 and 3 trials. Ophthalmology. 2019; 126: 1273–1285. [CrossRef] [PubMed]
Ciulla TA, Hussain RM, Berrocal AM, Nagiel A. Voretigene neparvovec-rzyl for treatment of RPE65-mediated inherited retinal diseases: a model for ocular gene therapy development. Expert Opin Biol Ther. 2020; 20: 565–578. [CrossRef] [PubMed]
Keeler AM, Flotte TR. Recombinant adeno-associated virus gene therapy in light of luxturna (and zolgensma and glybera): where are we, and how did we get here? Annu Rev Virol. 2019; 6: 601–621. [CrossRef] [PubMed]
Trapani I, Auricchio A. Seeing the light after 25 years of retinal gene therapy. Trends Mol Med. 2018; 24: 669–681. [CrossRef] [PubMed]
Dobrowsky T, Gianni D, Pieracci J, Suh J. AAV manufacturing for clinical use: insights on current challenges from the upstream process perspective. Current Opinion in Biomedical Engineering. 2021; 20: 100353. [CrossRef]
Moleirinho MG, Silva RJS, Alves PM, Carrondo MJT, Peixoto C. Current challenges in biotherapeutic particles manufacturing. Expert Opin Biol Ther. 2020; 20: 451–465. [CrossRef] [PubMed]
Srivastava A, Mallela KMG, Deorkar N, Brophy G. Manufacturing challenges and rational formulation development for AAV viral vectors. J Pharm Sci. 2021; 110: 2609–2624. [CrossRef] [PubMed]
Ladha R, Caspers LE, Willermain F, de Smet MD. Subretinal therapy: technological solutions to surgical and immunological challenges. Front Med (Lausanne). 2022; 9: 846782. [CrossRef] [PubMed]
Reichel FF, Dauletbekov DL, Klein R, et al. AAV8 can induce innate and adaptive immune response in the primate eye. Mol Ther. 2017; 25: 2648–2660. [CrossRef] [PubMed]
Ail D, Ren D, Brazhnikova E, et al. Systemic and local immune responses to intraocular AAV vector administration in non-human primates. Mol Ther Methods Clin Dev. 2022; 24: 306–316. [CrossRef] [PubMed]
Ampie L, McGavern DB. Immunological defense of CNS barriers against infections. Immunity. 2022; 55: 781–799. [CrossRef] [PubMed]
Timmers AM, et al. Ocular inflammatory response to intravitreal injection of adeno-associated virus vector: relative contribution of genome and capsid. Hum Gene Ther. 2020; 31: 80–89. [CrossRef] [PubMed]
Matet A, Kostic C, Bemelmans A-P, et al. Evaluation of tolerance to lentiviral LV-RPE65 gene therapy vector after subretinal delivery in non-human primates. Transl Res. 2017; 188: 40–57.e44. [CrossRef] [PubMed]
Ren D, Fisson S, Dalkara D, Ail D. Immune responses to gene editing by viral and non-viral delivery vectors used in retinal gene therapy. Pharmaceutics. 2022; 14: 1973. [CrossRef] [PubMed]
Varin J, Morival C, Maillard N, Adjali O, Cronin T. Risk mitigation of immunogenicity: a key to personalized retinal gene therapy. Int J Mol Sci. 2021; 22: 12818. [CrossRef] [PubMed]
Verdera HC, Kuranda K, Mingozzi F. AAV vector immunogenicity in humans: a long journey to successful gene transfer. Mol Ther. 2020; 28: 723–746. [CrossRef] [PubMed]
Bloquel C, Bourges JL, Touchard E, Berdugo M, BenEzra D, Behar-Cohen F. Non-viral ocular gene therapy: potential ocular therapeutic avenues. Adv Drug Deliv Rev. 2006; 58: 1224–1242. [CrossRef] [PubMed]
Bulcha JT, Wang Y, Ma H, Tai PWL, Gao G. Viral vector platforms within the gene therapy landscape. Signal Transduct Target Ther. 2021; 6: 53. [CrossRef] [PubMed]
Bennett J, Tanabe T, Sun D, et al. Photoreceptor cell rescue in retinal degeneration (rd) mice by in vivo gene therapy. Nat Med. 1996; 2: 649–654. [CrossRef] [PubMed]
Bischoff JR, Kim DH, Williams A, et al. An adenovirus mutant that replicates selectively in p53-deficient human tumor cells. Science. 1996; 274: 373–376. [CrossRef] [PubMed]
Kumaran N, Moore AT, Weleber RG, Michaelides M. Leber congenital amaurosis/early-onset severe retinal dystrophy: clinical features, molecular genetics and therapeutic interventions. Br J Ophthalmol. 2017; 101: 1147–1154. [CrossRef] [PubMed]
Stingl K, Kempf M, Jung R, et al. Therapy with voretigene neparvovec. How to measure success? Prog Retin Eye Res. 2023; 92: 101115. [CrossRef] [PubMed]
Tsang SH, Sharma T. Autosomal dominant retinitis pigmentosa. Adv Exp Med Biol. 2018; 1085: 69–77. [CrossRef] [PubMed]
Jiang J, Zhang X, Tang Y, Li S, Chen J. Progress on ocular siRNA gene-silencing therapy and drug delivery systems. Fundam Clin Pharmacol. 2021; 35: 4–24. [CrossRef] [PubMed]
Saw PE, Song EW. siRNA therapeutics: a clinical reality. Sci China Life Sci. 2020; 63: 485–500. [CrossRef] [PubMed]
Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998; 391: 806–811. [CrossRef] [PubMed]
Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature. 2001; 411: 494–498. [CrossRef] [PubMed]
Salminen A, Kauppinen A, Hyttinen JM, Toropainen E, Kaarniranta K. Endoplasmic reticulum stress in age-related macular degeneration: trigger for neovascularization. Mol Med. 2010; 16: 535–542. [CrossRef] [PubMed]
Hoy SM . Patisiran: first global approval. Drugs. 2018; 78: 1625–1631. [CrossRef] [PubMed]
Padda IS, Mahtani AU, Parmar M. Small interfering RNA (siRNA) based therapy. In: StatPearls (2022).
Gaj T, Sirk SJ, Shui SL, Liu J. Genome-editing technologies: principles and applications. Cold Spring Harb Perspect Biol. 2016; 8: a023754. [CrossRef] [PubMed]
da Costa BL, Levi SR, Eulau E, Tsai YT, Quinn PMJ. Prime editing for inherited retinal diseases. Front Genome Ed. 2021; 3: 775330. [CrossRef] [PubMed]
Maeder ML, Stefanidakis M, Wilson CJ, et al. Development of a gene-editing approach to restore vision loss in Leber congenital amaurosis type 10. Nat Med. 2019; 25: 229–233. [CrossRef] [PubMed]
Quinn J, Musa A, Kantor A, et al. Genome-editing strategies for treating human retinal degenerations. Hum Gene Ther. 2021; 32: 247–259. [CrossRef] [PubMed]
Genin E, Feingold J, Clerget-Darpoux F. Identifying modifier genes of monogenic disease: strategies and difficulties. Hum Genet. 2008; 124: 357–368. [CrossRef] [PubMed]
Cutting GR . Modifier genes in Mendelian disorders: the example of cystic fibrosis. Ann N Y Acad Sci. 2010; 1214: 57–69. [CrossRef] [PubMed]
Hamilton BA, Yu BD. Modifier genes and the plasticity of genetic networks in mice. PLoS Genet. 2012; 8: e1002644. [CrossRef] [PubMed]
Nadeau JH . Modifer genes in mice and humans. Nat Rev. 2001; 2: 165–174. [CrossRef]
Walsh MB, Janzen E, Wingrove E, et al. Genetic modifiers ameliorate endocytic and neuromuscular defects in a model of spinal muscular atrophy. BMC Biol. 2020; 18: 127. [CrossRef] [PubMed]
Cruz NM, Yuan Y, Leehy BD, et al. Modifier genes as therapeutics: the nuclear hormone receptor Rev Erb alpha (Nr1d1) rescues Nr2e3 associated retinal disease. PLoS One. 2014; 9: e87942. [CrossRef] [PubMed]
Li S, Datta S, Brabbit E, et al. Nr2e3 is a genetic modifier that rescues retinal degeneration and promotes homeostasis in multiple models of retinitis pigmentosa. Gene Ther. 2020; 28: 223–241. [CrossRef] [PubMed]
Haider NB, Zhang W, Hurd R, et al. Mapping of genetic modifiers of Nr2e3 rd7/rd7 that suppress retinal degeneration and restore blue cone cells to normal quantity. Mamm Genome. 2008; 19: 145–154. [CrossRef] [PubMed]
Goncalves GAR, Paiva RMA. Gene therapy: advances, challenges and perspectives. Einstein (Sao Paulo). 2017; 15: 369–375. [CrossRef] [PubMed]
Han IC, Burnight ER, Ulferts MJ, et al. Helper-dependent adenovirus transduces the human and rat retina but elicits an inflammatory reaction when delivered subretinally in rats. Hum Gene Ther. 2019; 30: 1371–1384. [CrossRef] [PubMed]
Planul A, Dalkara D. Vectors and gene delivery to the retina. Annu Rev Vis Sci. 2017; 3: 121–140. [CrossRef] [PubMed]
Gorziglia MI, Kadan MJ, Yei S, et al. Elimination of both E1 and E2 from adenovirus vectors further improves prospects for in vivo human gene therapy. J Virol. 1996; 70: 4173–4178. [CrossRef] [PubMed]
Engelhardt JF, Ye X, Doranz B, Wilson JM. Ablation of E2A in recombinant adenoviruses improves transgene persistence and decreases inflammatory response in mouse liver. Proc Natl Acad Sci USA. 1994; 91: 6196–6200. [CrossRef] [PubMed]
Amalfitano A, Hauser MA, Hu H, Serra D, Begy CR, Chamberlain JS. Production and characterization of improved adenovirus vectors with the E1, E2b, and E3 genes deleted. J Virol. 1998; 72: 926–933. [CrossRef] [PubMed]
Osada T, Yang XY, Hartman ZC, et al. Optimization of vaccine responses with an E1, E2b and E3-deleted Ad5 vector circumvents pre-existing anti-vector immunity. Cancer Gene Ther. 2009; 16: 673–682. [CrossRef] [PubMed]
Gao GP, Yang Y, Wilson JM. Biology of adenovirus vectors with E1 and E4 deletions for liver-directed gene therapy. J Virol. 1996; 70: 8934–8943. [CrossRef] [PubMed]
Gregory SM, Nazir SA, Metcalf JP. Implications of the innate immune response to adenovirus and adenoviral vectors. Future Virology. 2011; 6: 357–374. [CrossRef] [PubMed]
Pascual-Pasto G, Bazan-Peregrino M, Olaciregui NG, et al. Therapeutic targeting of the RB1 pathway in retinoblastoma with the oncolytic adenovirus VCN-01. Sci Transl Med. 2019; 11: eaat9321. [CrossRef] [PubMed]
Rodrigues GA, Shalaev E, Karami TK, Cunningham J, Slater NKH, Rivers HM. Pharmaceutical development of AAV-based gene therapy products for the eye. Pharm Res. 2018; 36: 29. [CrossRef] [PubMed]
Le Meur G, Lebranchu P, Billaud F, et al. Safety and long-term efficacy of AAV4 gene therapy in patients with RPE65 Leber congenital amaurosis. Mol Ther. 2018; 26: 256–268. [CrossRef] [PubMed]
Pavlou M, Schön C, Occelli LM, et al. Novel AAV capsids for intravitreal gene therapy of photoreceptor disorders. EMBO Mol Med. 2021; 13: e13392. [CrossRef] [PubMed]
Carrella S, Indrieri A, Franco B, Banfi S. Mutation-independent therapies for retinal diseases: focus on gene-based approaches. Front Neurosci. 2020; 14: 588234. [CrossRef] [PubMed]
Shah NH, Muir TW. Inteins: nature's gift to protein chemists. Chem Sci. 2014; 5: 446–461. [CrossRef] [PubMed]
Zhi S, Chen Y, Wu G, et al. Dual-AAV delivering split prime editor system for in vivo genome editing. Mol Ther. 2022; 30: 283–294. [CrossRef] [PubMed]
Tornabene P, Trapani I, Centrulo M, et al. Inclusion of a degron reduces levelsof undesired inteins after AAV-mediated proteintrans-splicing in the retina. Mol Ther Methods Clin Dev. 2021; 23: 448–459. [CrossRef] [PubMed]
McClements ME, Barnard AR, Singh MS, et al. An AAV dual vector strategy ameliorates the Stargardt phenotype in adult Abca4(-/-) mice. Hum Gene Ther. 2019; 30: 590–600. [CrossRef] [PubMed]
Tong A . Some genes are too large to fit into AAV vectors. UCB, Novartis back $57M idea to deliver them in parts instead. Endpoints News. 2022. Available at: https://endpts.com/some-genes-are-too-large-to-fit-into-aav-vectors-ucb-novartis-back-57m-idea-to-deliver-them-in-parts-instead/.
Arsenijevic Y, Berger A, Udry F, Kostic C. Lentiviral vectors for ocular gene therapy. Pharmaceutics. 2022; 14: 1605. [CrossRef] [PubMed]
Matoba T, Egashira K. Anti-inflammatory gene therapy for cardiovascular disease. Curr Gene Ther. 2011; 11: 442–446. [CrossRef] [PubMed]
Oliveira AV, Rosa da Costa AM, Silva GA. Non-viral strategies for ocular gene delivery. Mater Sci Eng C Mater Biol Appl. 2017; 77: 1275–1289. [CrossRef] [PubMed]
Zhu G, Chen X. Aptamer-based targeted therapy. Adv Drug Deliv Rev. 2018; 134: 65–78. [CrossRef] [PubMed]
Nimjee SM, White RR, Becker RC, Sullenger BA. Aptamers as therapeutics. Annu Rev Pharmacol Toxicol. 2017; 57: 61–79. [CrossRef] [PubMed]
Kim YK . RNA therapy: rich history, various applications and unlimited future prospects. Exp Mol Med. 2022; 54: 455–465. [CrossRef] [PubMed]
Patel S, Ryals RC, Weller KK, Pennesi ME, Sahay G. Lipid nanoparticles for delivery of messenger RNA to the back of the eye. J Control Release. 2019; 303: 91–100. [CrossRef] [PubMed]
Xiang C, Tenkumo T, Ogawa T, et al. Gene transfection achieved by utilizing antibacterial calcium phosphate nanoparticles for enhanced regenerative therapy. Acta Biomater. 2021; 119: 375–389. [CrossRef] [PubMed]
Hu J, Kovtun A, Tomaszewski A, et al. A new tool for the transfection of corneal endothelial cells: calcium phosphate nanoparticles. Acta Biomater. 2012; 8: 1156–1163. [CrossRef] [PubMed]
Cai X, Nash Z, Conley SM, Fliesler SJ, Cooper MJ, Naash MI. A partial structural and functional rescue of a retinitis pigmentosa model with compacted DNA nanoparticles. PLoS One. 2009; 4: e5290. [CrossRef] [PubMed]
Farjo R, Skaggs J, Quiambao AB, Cooper MJ, Naash MI. Efficient non-viral ocular gene transfer with compacted DNA nanoparticles. PLoS One. 2006; 1: e38. [CrossRef] [PubMed]
Cai X, Conley SM, Nash Z, Fliesler SJ, Cooper MJ, Naash MJ. Gene delivery to mitotic and postmitotic photoreceptors via compacted DNA nanoparticles results in improved phenotype in a mouse model of retinitis pigmentosa. FASEB J. 2010; 24: 1178–1191. [CrossRef] [PubMed]
Han Z, Banworth MJ, Makkia R, et al. Genomic DNA nanoparticles rescue rhodopsin-associated retinitis pigmentosa phenotype. FASEB J. 2015; 29: 2535–2544. [CrossRef] [PubMed]
Han Z, Conley SM, Makkia RS, Cooper MJ, Naash MI. DNA nanoparticle-mediated ABCA4 delivery rescues Stargardt dystrophy in mice. J Clin Invest. 2012; 122: 3221–3226. [CrossRef] [PubMed]
Amadio M, Pascale A, Cupri S, et al. Nanosystems based on siRNA silencing HuR expression counteract diabetic retinopathy in rat. Pharmacol Res. 2016; 111: 713–720. [CrossRef] [PubMed]
Bonilla L, Espina M, Severino P, et al. Lipid nanoparticles for the posterior eye segment. Pharmaceutics. 2021; 14: 90. [CrossRef] [PubMed]
Liu HA, Liu YL, Ma ZZ, Wang JC, Zhang Q. A lipid nanoparticle system improves siRNA efficacy in RPE cells and a laser-induced murine CNV model. Invest Ophthalmol Vis Sci. 2011; 52: 4789–4794. [CrossRef] [PubMed]
Ryals RC, Patel S, Acosta C, McKinney M, Pennesi ME, Sahay G. The effects of PEGylation on LNP based mRNA delivery to the eye. PLoS One. 2020; 15: e0241006. [CrossRef] [PubMed]
Sahu B, Chug I, Khanna H. The ocular gene delivery landscape. Biomolecules. 2021; 11: 1135. [CrossRef] [PubMed]
Jang HK, Jo DH, Lee SN, et al. High-purity production and precise editing of DNA base editing ribonucleoproteins. Sci Adv .2021; 7: eabg2661. [CrossRef] [PubMed]
Zuris JA, Thompson DB, Shu Y, et al. Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat Biotechnol. 2015; 33: 73–80. [CrossRef] [PubMed]
Kazi KM, Mandal AS, Biswas N, et al. Niosome: a future of targeted drug delivery systems. J Adv Pharm Technol Res. 2010; 1: 374–380. [PubMed]
Chen S, Hanning S, Falconer J, Locke M, Wen J. Recent advances in non-ionic surfactant vesicles (niosomes): fabrication, characterization, pharmaceutical and cosmetic applications. Eur J Pharm Biopharm. 2019; 144: 18–39. [CrossRef] [PubMed]
Al Qtaish N, Gallego I, Villate-Beitia I, et al. Niosome-based approach for in situ gene delivery to retina and brain cortex as immune-privileged tissues. Pharmaceutics. 2020; 12: 198. [CrossRef] [PubMed]
Durak S, Rad ME, Yetisgin AA, et al. Niosomal drug delivery systems for ocular disease-recent advances and future prospects. Nanomaterials (Basel). 2020; 10: 1191. [CrossRef] [PubMed]
Mashal M, Attia N, Martínez-Navarrete G, et al. Gene delivery to the rat retina by non-viral vectors based on chloroquine-containing cationic niosomes. J Control Release. 2019; 304: 181–190. [CrossRef] [PubMed]
Mashal M, Attia N, Puras G, Martínez-Navarrete G, Fernández E, Pedraz JL. Retinal gene delivery enhancement by lycopene incorporation into cationic niosomes based on DOTMA and polysorbate 60. J Control Release. 2017; 254: 55–64. [CrossRef] [PubMed]
Puras G, Mashal M, Zárate J, et al. A novel cationic niosome formulation for gene delivery to the retina. J Control Release. 2014; 174: 27–36. [CrossRef] [PubMed]
Villate-Beitia I, Gallego I, Martínez-Navarrete G, et al. Polysorbate 20 non-ionic surfactant enhances retinal gene delivery efficiency of cationic niosomes after intravitreal and subretinal administration. Int J Pharm. 2018; 550: 388–397. [CrossRef] [PubMed]
Al Qtaish N, Gallego I, Paredes AJ, et al. Nanodiamond integration into niosomes as an emerging and efficient gene therapy nanoplatform for central nervous system diseases. ACS Appl Mater Interfaces. 2022; 14: 13665–13677. [CrossRef] [PubMed]
Birch DG, Bennett LD, Duncan JL, Weleber RG, Pennesi ME. Long-term follow-up of patients with retinitis pigmentosa receiving intraocular ciliary neurotrophic factor implants. Am J Ophthalmol. 2016; 170: 10–14. [CrossRef] [PubMed]
Pardue MT, Allen RS. Neuroprotective strategies for retinal disease. Prog Retin Eye Res. 2018; 65: 50–76. [CrossRef] [PubMed]
Osborne A, Khatib TZ, Songra L, et al. Neuroprotection of retinal ganglion cells by a novel gene therapy construct that achieves sustained enhancement of brain-derived neurotrophic factor/tropomyosin-related kinase receptor-B signaling. Cell Death Dis. 2018; 9: 1007. [CrossRef] [PubMed]
Pease ME, Zack DJ, Berlinicke C, et al. Effect of CNTF on retinal ganglion cell survival in experimental glaucoma. Invest Ophthalmol Vis Sci. 2009; 50: 2194–2200. [CrossRef] [PubMed]
Askou AL, Alsing S, Benckendorff JNE, et al. Suppression of choroidal neovascularization by AAV-based dual-acting antiangiogenic gene therapy. Mol Ther Nucleic Acids. 2019; 16: 38–50. [CrossRef] [PubMed]
Wang S, Koster KM, He Y, Zhou Q. miRNAs as potential therapeutic targets for age-related macular degeneration. Future Med Chem. 2012; 4: 277–287. [CrossRef] [PubMed]
Berber P, Grassmann F, Kiel C, Weber BH. An eye on age-related macular degeneration: the role of microRNAs in disease pathology. Mol Diagn Ther. 2017; 21: 31–43. [CrossRef] [PubMed]
McClements ME, Steward H, Atkin W, et al. Tropism of AAV vectors in photoreceptor-like cells of human iPSC-derived retinal organoids. Transl Vis Sci Technol. 2022; 11: 3. [CrossRef] [PubMed]
Jüttner J, Szabo A, Gross-Scherf B, et al. Targeting neuronal and glial cell types with synthetic promoter AAVs in mice, non-human primates and humans. Nat Neurosci. 2019; 22: 1345–1356. [CrossRef] [PubMed]
Castle MJ, Turunen HT, Vandenberghe LH, Wolfe JH. Controlling AAV tropism in the nervous system with natural and engineered capsids. Methods Mol Biol. 2016; 1382: 133–149. [CrossRef] [PubMed]
Dalkara D, Byrne LC, Klimczak RR, et al. In vivo-directed evolution of a new adeno-associated virus for therapeutic outer retinal gene delivery from the vitreous. Sci Transl Med. 2013; 5: 189ra176. [CrossRef]
Allocca M, Mussolino C, Garcia-Hoyos M, et al. Novel adeno-associated virus serotypes efficiently transduce murine photoreceptors. J Virol. 2007; 81: 11372–11380. [CrossRef] [PubMed]
Boye SE, Alexander JJ, Boye SL, et al. The human rhodopsin kinase promoter in an AAV5 vector confers rod- and cone-specific expression in the primate retina. Hum Gene Ther. 2012; 23: 1101–1115. [CrossRef] [PubMed]
Vandenberghe LH, Bell P, Maguire AM, et al. AAV9 targets cone photoreceptors in the nonhuman primate retina. PLoS One. 2013; 8: e53463. [CrossRef] [PubMed]
Huang P, Narendran S, Pereira F, et al. The learning curve of murine subretinal injection among clinically trained ophthalmic surgeons. Transl Vis Sci Technol. 2022; 11: 13. [CrossRef]
Awwad S, Mohamed Ahmed AHA, Sharma G, et al. Principles of pharmacology in the eye. Br J Pharmacol. 2017; 174: 4205–4223. [CrossRef] [PubMed]
Gaudana R, Ananthula HK, Parenky A, Mitra AK. Ocular drug delivery. AAPS J. 2010; 12: 348–360. [CrossRef] [PubMed]
Perez VL, Wirostko B, Korenfeld M, From S, Raizman M. Ophthalmic drug delivery using iontophoresis: recent clinical applications. J Ocul Pharmacol Ther. 2020; 36: 75–87. [CrossRef] [PubMed]
Gote V, Sikder S, Sicotte J, Pal D. Ocular drug delivery: present innovations and future challenges. J Pharmacol Exp Ther. 2019; 370: 602–624. [CrossRef] [PubMed]
Nguyen DCT, Dowling J, Ryan R, McLoughlin P, Fitzhenry L. Pharmaceutical-loaded contact lenses as an ocular drug delivery system: a review of critical lens characterization methodologies with reference to ISO standards. Cont Lens Anterior Eye. 2021; 44: 101487. [CrossRef] [PubMed]
Yang YH, Hsu WC, Hsieh YT. Anterior migration of triamcinolone acetonide after posterior subtenon injection for macular edema predisposes to intraocular pressure elevation. Curr Eye Res. 2021; 46: 689–693. [CrossRef] [PubMed]
Nilforushan N, Abolfathzadeh N, Najafzadeh F, Samimifar D. Subtenon triamcinolone injection to treat serous choroidal detachment after glaucoma surgery: a case series. Eur J Ophthalmol. 2022; 33: 319–323. [CrossRef] [PubMed]
Raghava S, Hammond M, Kompella UB. Periocular routes for retinal drug delivery. Expert Opin Drug Deliv. 2004; 1: 99–114. [CrossRef] [PubMed]
Kim HM, Woo SJ. Ocular drug delivery to the retina: current innovations and future perspectives. Pharmaceutics. 2021; 13: 108. [CrossRef] [PubMed]
Peng Y, Tang L, Zhou Y. Subretinal injection: a review on the novel route of therapeutic delivery for vitreoretinal diseases. Ophthalmic Res. 2017; 58: 217–226. [CrossRef] [PubMed]
Jacobson SG, Acland GM, Aguirre GD, et al. Safety of recombinant adeno-associated virus type 2-RPE65 vector delivered by ocular subretinal injection. Mol Ther. 2006; 13: 1074–1084. [CrossRef] [PubMed]
Falavarjani KG, Nguyen QD. Adverse events and complications associated with intravitreal injection of anti-VEGF agents: a review of literature. Eye (Lond). 2013; 27: 787–794. [CrossRef] [PubMed]
Adverum. (GlobeNewswire, 2021).
Naftali Ben Haim L, Moisseiev E. Drug delivery via the suprachoroidal space for the treatment of retinal diseases. Pharmaceutics. 2021; 13: 967. [CrossRef] [PubMed]
Kansara V, Muya L, Wan CR, Ciulla TA. Suprachoroidal delivery of viral and nonviral gene therapy for retinal diseases. J Ocul Pharmacol Ther. 2020; 36: 384–392. [CrossRef] [PubMed]
Bucher K, Rodriguez-Bocanegra E, Dauletbekov D, Fischer MD. Immune responses to retinal gene therapy using adeno-associated viral vectors - Implications for treatment success and safety. Prog Retin Eye Res. 2021; 83: 100915. [CrossRef] [PubMed]
Song L, Bower JJ, Llanga T, Salmon JH, Hirsch ML, Gilger BC. Ocular tolerability and immune response to corneal intrastromal AAV-IDUA gene therapy in New Zealand white rabbits. Mol Ther Methods Clin Dev. 2020; 18: 24–32. [CrossRef] [PubMed]
Song C, Conlon TJ, Deng WT, et al. Toxicology and pharmacology of an AAV vector expressing codon-optimized RPGR in RPGR-deficient Rd9 mice. Hum Gene Ther Clin Dev. 2018; 29: 188–197. [CrossRef] [PubMed]
Dufour VL, Cideciyan AV, Ye GJ, et al. Toxicity and efficacy evaluation of an adeno-associated virus vector expressing codon-optimized RPGR delivered by subretinal injection in a canine model of X-linked retinitis pigmentosa. Hum Gene Ther. 2020; 31: 253–267. [CrossRef] [PubMed]
Khabou HC; Pacot L; Fisson S; Dalkara D. Dosage thresholds and influence of transgene cassette in adeno-associated virus-related toxicity. Human Gene Therapy. 2018; 29: 1235–1241. [CrossRef] [PubMed]
Xiong W, Wu DM, Xue Y, et al. AAV cis-regulatory sequences are correlated with ocular toxicity. Proc Natl Acad Sci USA. 2019; 116: 5785–5794. [CrossRef] [PubMed]
Lee S, Kang IK, Kim JH, et al. Relationship between neutralizing antibodies against adeno-associated virus in the vitreous and serum: effects on retinal gene therapy. Transl Vis Sci Technol. 2019; 8: 14. [CrossRef]
Rogers GL, Suzuki M, Zolotukhin I, et al. Unique roles of TLR9- and MyD88-dependent and -independent pathways in adaptive immune responses to AAV-mediated gene transfer. J Innate Immun. 2015; 7: 302–314. [CrossRef] [PubMed]
Ronzitti G, Gross DA, Mingozzi F. Human immune responses to adeno-associated virus (AAV) vectors. Front Immunol. 2020; 11: 670. [CrossRef] [PubMed]
Gorovits B, Clements-Egan A, Birchler M, et al. Pre-existing antibody: biotherapeutic modality-based review. AAPS J. 2016; 18: 311–320. [CrossRef] [PubMed]
Rosenberg AS, Sauna ZE. Immunogenicity assessment during the development of protein therapeutics. J Pharm Pharmacol. 2018; 70: 584–594. [CrossRef] [PubMed]
Pratt KP . Anti-drug antibodies: emerging approaches to predict, reduce or reverse biotherapeutic immunogenicity. Antibodies (Basel). 2018; 7: 19. [CrossRef] [PubMed]
Zinsli LV, Stierlin N, Loessner MJ, Schmelcher M. Deimmunization of protein therapeutics - Recent advances in experimental and computational epitope prediction and deletion. Comput Struct Biotechnol J. 2021; 19: 315–329. [CrossRef] [PubMed]
Zhang P, Jain P, Tsao C, Wu K, Jiang S. Proactively reducing anti-drug antibodies via immunomodulatory bioconjugation. Angew Chem Int Ed Engl. 2019; 58: 2433–2436. [CrossRef] [PubMed]
Scholz R, Sobotka M, Caramoy A, Stempfl T, Moehle C, Langmann T. Minocycline counter-regulates pro-inflammatory microglia responses in the retina and protects from degeneration. J Neuroinflammation. 2015; 12: 209. [CrossRef] [PubMed]
Leborgne C, Barbon E, Alexander JM, et al. IgG-cleaving endopeptidase enables in vivo gene therapy in the presence of anti-AAV neutralizing antibodies. Nat Med. 2020; 26: 1096–1101. [CrossRef] [PubMed]
Meliani A, Boisgerault F, Hardet R, et al. Antigen-selective modulation of AAV immunogenicity with tolerogenic rapamycin nanoparticles enables successful vector re-administration. Nat Commun. 2018; 9: 4098. [CrossRef] [PubMed]
Bonfiglio V, Reibaldi M, Macchi I, et al. Preoperative, intraoperative and postoperative corticosteroid use as an adjunctive treatment for rhegmatogenous retinal detachment. J Clin Med. 2020; 9: 1556. [CrossRef] [PubMed]
Wu JS, Lin CJ, Hwang JF, Chen SN. Influence of systemic steroids on subretinal fluid after scleral buckle surgery for macula-off retinal detachment. Retina. 2011; 31: 99–104. [CrossRef] [PubMed]
Wozar F, Seitz I, Reichel F, Fischer MD. Importance of nonhuman primates as a model system for gene therapy development in ophthalmology. Klin Monbl Augenheilkd. 2022; 239: 270–274. [PubMed]
Arruda VR, Samelson-Jones BJ. Obstacles and future of gene therapy for hemophilia. Expert Opin Orphan Drugs. 2015; 3: 997–1010. [CrossRef] [PubMed]
Tian B, Bilsbury E, Doherty S, et al. Ocular drug delivery: advancements and innovations. Pharmaceutics. 2022; 14: 1931. [CrossRef] [PubMed]
Gong J, Cheung S, Fasso-Opie A, et al. The impact of inherited retinal diseases in the United States of America (US) and Canada from a cost-of-illness perspective. Clin Ophthalmol. 2021; 15: 2855–2866. [CrossRef] [PubMed]
Caracciolo G, Pozzi D, Caminiti R, et al. Enhanced transfection efficiency of multicomponent lipoplexes in the regime of optimal membrane charge density. J Phys Chem B. 2008; 112: 11298–11304. [CrossRef] [PubMed]
Mochizuki S, Nishina K, Fujii S, Sakurai K. The transfection efficiency of calix[4]arene-based lipids: the role of the alkyl chain length. Biomater Sci. 2015; 3: 317–322. [CrossRef] [PubMed]
Liao HW, Yau KW. In vivo gene delivery in the retina using polyethylenimine. Biotechniques. 2007; 42: 285–286, 288. [CrossRef] [PubMed]
Mastorakos P, Kambhampati SP, Mishra MK, et al. Hydroxyl PAMAM dendrimer-based gene vectors for transgene delivery to human retinal pigment epithelial cells. Nanoscale .2015; 7: 3845–3856. [CrossRef] [PubMed]
Kurosaki T, Uematsu M, Shimoda K, et al. Ocular gene delivery systems using ternary complexes of plasmid DNA, polyethylenimine, and anionic polymers. Biol Pharm Bull. 2013; 36: 96–101. [CrossRef] [PubMed]
Figure 1.
 
History of the development of retinal gene therapy (created with BioRender.com).
Figure 1.
 
History of the development of retinal gene therapy (created with BioRender.com).
Figure 2.
 
Gene therapy approaches. There are four major approaches to gene therapy (left to right). Gene replacement or augmentation is when a functional copy of a damaged, non-functional gene is added to augment the production of functional protein. In gene editing, mutations in a gene are corrected or expression of the mutated protein is reduced to alter a diseased state. Gene silencing uses the RNAi mechanism to eliminate the aberrant expression of the targeted pathogenic protein in acquired diseases. Modifier gene therapies provide a modifier gene that can affect pathways downstream or upstream from the damaged gene, allowing for the expression of multiple genes to be altered with a single treatment (created with BioRender.com).
Figure 2.
 
Gene therapy approaches. There are four major approaches to gene therapy (left to right). Gene replacement or augmentation is when a functional copy of a damaged, non-functional gene is added to augment the production of functional protein. In gene editing, mutations in a gene are corrected or expression of the mutated protein is reduced to alter a diseased state. Gene silencing uses the RNAi mechanism to eliminate the aberrant expression of the targeted pathogenic protein in acquired diseases. Modifier gene therapies provide a modifier gene that can affect pathways downstream or upstream from the damaged gene, allowing for the expression of multiple genes to be altered with a single treatment (created with BioRender.com).
Figure 3.
 
Gene Therapy Vectors may be Either Viral or Non-Viral, Each With its own Advantages and Risks (Created With BioRender.com).
Figure 3.
 
Gene Therapy Vectors may be Either Viral or Non-Viral, Each With its own Advantages and Risks (Created With BioRender.com).
Figure 4.
 
Ocular routes of administration may be invasive or non-invasive (created with BioRender.com).
Figure 4.
 
Ocular routes of administration may be invasive or non-invasive (created with BioRender.com).
Table 1.
 
Gene Therapy Approaches
Table 1.
 
Gene Therapy Approaches
Table 2.
 
Gene Therapy Vectors
Table 2.
 
Gene Therapy Vectors
Table 3.
 
Ocular Routes of Administration
Table 3.
 
Ocular Routes of Administration
Table 4.
 
Summary of Active Gene Therapy Clinical Studies with Ocular Disease Indications
Table 4.
 
Summary of Active Gene Therapy Clinical Studies with Ocular Disease Indications
×
×

This PDF is available to Subscribers Only

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

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

×