Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Proc Natl Acad Sci U S A ; 119(13): e2117038119, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35316139

ABSTRACT

SignificanceCanine models of inherited retinal diseases have helped advance adeno-associated virus (AAV)-based gene therapies targeting specific cells in the outer retina for treating blinding diseases in patients. However, therapeutic targeting of diseases such as congenital stationary night blindness (CSNB) that exhibit defects in ON-bipolar cells (ON-BCs) of the midretina remains underdeveloped. Using a leucine-rich repeat, immunoglobulin-like and transmembrane domain 3 (LRIT3) mutant canine model of CSNB exhibiting ON-BC dysfunction, we tested the ability of cell-specific AAV capsids and promotors to specifically target ON-BCs for gene delivery. Subretinal injection of one vector demonstrated safety and efficacy with robust and stable rescue of electroretinography signals and night vision up to 1 y, paving the way for clinical trials in patients.


Subject(s)
Genetic Diseases, X-Linked , Night Blindness , Animals , Dependovirus/genetics , Dogs , Electroretinography , Eye Diseases, Hereditary , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/therapy , Genetic Therapy , Humans , Membrane Proteins/genetics , Myopia , Night Blindness/genetics , Night Blindness/therapy
2.
Mol Ther ; 29(8): 2456-2468, 2021 08 04.
Article in English | MEDLINE | ID: mdl-33781914

ABSTRACT

The inherited childhood blindness caused by mutations in NPHP5, a form of Leber congenital amaurosis, results in abnormal development, dysfunction, and degeneration of photoreceptors. A naturally occurring NPHP5 mutation in dogs leads to a phenotype that very nearly duplicates the human retinopathy in terms of the photoreceptors involved, spatial distribution of degeneration, and the natural history of vision loss. We show that adeno-associated virus (AAV)-mediated NPHP5 gene augmentation of mutant canine retinas at the time of active degeneration and peak cell death stably restores photoreceptor structure, function, and vision with either the canine or human NPHP5 transgenes. Mutant cone photoreceptors, which failed to form outer segments during development, reform this structure after treatment. Degenerating rod photoreceptor outer segments are stabilized and develop normal structure. This process begins within 8 weeks after treatment and remains stable throughout the 6-month posttreatment period. In both photoreceptor cell classes mislocalization of rod and cone opsins is minimized or reversed. Retinal function and functional vision are restored. Efficacy of gene therapy in this large animal ciliopathy model of Leber congenital amaurosis provides a path for translation to human treatment.


Subject(s)
Calmodulin-Binding Proteins/administration & dosage , Dependovirus/genetics , Leber Congenital Amaurosis/therapy , Retinal Cone Photoreceptor Cells/pathology , Animals , Calmodulin-Binding Proteins/pharmacology , Disease Models, Animal , Dogs , Electroretinography , Genetic Therapy , Genetic Vectors/administration & dosage , Humans , Leber Congenital Amaurosis/genetics , Treatment Outcome
3.
Mol Ther ; 28(1): 266-278, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31604676

ABSTRACT

The form of hereditary childhood blindness Leber congenital amaurosis (LCA) caused by biallelic RPE65 mutations is considered treatable with a gene therapy product approved in the US and Europe. The resulting vision improvement is well accepted, but long-term outcomes on the natural history of retinal degeneration are controversial. We treated four RPE65-mutant dogs in mid-life (age = 5-6 years) and followed them long-term (4-5 years). At the time of the intervention at mid-life, there were intra-ocular and inter-animal differences in local photoreceptor layer health ranging from near normal to complete degeneration. Treated locations having more than 63% of normal photoreceptors showed robust treatment-related retention of photoreceptors in the long term. Treated regions with less retained photoreceptors at the time of the intervention showed progressive degeneration similar to untreated regions with matched initial stage of disease. Unexpectedly, both treated and untreated regions in study eyes tended to show less degeneration compared to matched locations in untreated control eyes. These results support the hypothesis that successful long-term arrest of progression with RPE65 gene therapy may only occur in retinal regions with relatively retained photoreceptors at the time of the intervention, and there may be heretofore unknown mechanisms causing long-distance partial treatment effects beyond the region of subretinal injection.


Subject(s)
Genetic Therapy/methods , Leber Congenital Amaurosis/therapy , Mutation , Retinal Degeneration/genetics , cis-trans-Isomerases/genetics , Animals , Disease Models, Animal , Dogs , Electroretinography , Female , Follow-Up Studies , Leber Congenital Amaurosis/diagnostic imaging , Photoreceptor Cells, Vertebrate/metabolism , Retina/diagnostic imaging , Retina/metabolism , Retinal Degeneration/diagnostic imaging , Treatment Outcome , Vision, Ocular
4.
Proc Natl Acad Sci U S A ; 115(12): E2839-E2848, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29507198

ABSTRACT

Mutations in the BEST1 gene cause detachment of the retina and degeneration of photoreceptor (PR) cells due to a primary channelopathy in the neighboring retinal pigment epithelium (RPE) cells. The pathophysiology of the interaction between RPE and PR cells preceding the formation of retinal detachment remains not well-understood. Our studies of molecular pathology in the canine BEST1 disease model revealed retina-wide abnormalities at the RPE-PR interface associated with defects in the RPE microvillar ensheathment and a cone PR-associated insoluble interphotoreceptor matrix. In vivo imaging demonstrated a retina-wide RPE-PR microdetachment, which contracted with dark adaptation and expanded upon exposure to a moderate intensity of light. Subretinal BEST1 gene augmentation therapy using adeno-associated virus 2 reversed not only clinically detectable subretinal lesions but also the diffuse microdetachments. Immunohistochemical analyses showed correction of the structural alterations at the RPE-PR interface in areas with BEST1 transgene expression. Successful treatment effects were demonstrated in three different canine BEST1 genotypes with vector titers in the 0.1-to-5E11 vector genomes per mL range. Patients with biallelic BEST1 mutations exhibited large regions of retinal lamination defects, severe PR sensitivity loss, and slowing of the retinoid cycle. Human translation of canine BEST1 gene therapy success in reversal of macro- and microdetachments through restoration of cytoarchitecture at the RPE-PR interface has promise to result in improved visual function and prevent disease progression in patients affected with bestrophinopathies.


Subject(s)
Bestrophins/genetics , Eye Diseases, Hereditary/therapy , Genetic Therapy/methods , Retinal Diseases/therapy , Animals , Dog Diseases/therapy , Dogs , Eye Diseases, Hereditary/diagnostic imaging , Eye Diseases, Hereditary/pathology , Eye Diseases, Hereditary/veterinary , Female , Genetic Vectors/pharmacology , Humans , Light , Male , Mutation , Retinal Detachment/diagnostic imaging , Retinal Detachment/pathology , Retinal Detachment/therapy , Retinal Diseases/diagnostic imaging , Retinal Diseases/pathology , Retinal Diseases/veterinary , Retinal Pigment Epithelium/pathology , Tomography, Optical Coherence
5.
Proc Natl Acad Sci U S A ; 115(36): E8547-E8556, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30127005

ABSTRACT

Inherited retinal degenerations are caused by mutations in >250 genes that affect photoreceptor cells or the retinal pigment epithelium and result in vision loss. For autosomal recessive and X-linked retinal degenerations, significant progress has been achieved in the field of gene therapy as evidenced by the growing number of clinical trials and the recent commercialization of the first gene therapy for a form of congenital blindness. However, despite significant efforts to develop a treatment for the most common form of autosomal dominant retinitis pigmentosa (adRP) caused by >150 mutations in the rhodopsin (RHO) gene, translation to the clinic has stalled. Here, we identified a highly efficient shRNA that targets human (and canine) RHO in a mutation-independent manner. In a single adeno-associated viral (AAV) vector we combined this shRNA with a human RHO replacement cDNA made resistant to RNA interference and tested this construct in a naturally occurring canine model of RHO-adRP. Subretinal vector injections led to nearly complete suppression of endogenous canine RHO RNA, while the human RHO replacement cDNA resulted in up to 30% of normal RHO protein levels. Noninvasive retinal imaging showed photoreceptors in treated areas were completely protected from retinal degeneration. Histopathology confirmed retention of normal photoreceptor structure and RHO expression in rod outer segments. Long-term (>8 mo) follow-up by retinal imaging and electroretinography indicated stable structural and functional preservation. The efficacy of this gene therapy in a clinically relevant large-animal model paves the way for treating patients with RHO-adRP.


Subject(s)
Dependovirus , Gene Knock-In Techniques/methods , Gene Knockdown Techniques/methods , Genetic Therapy/methods , Genetic Vectors , RNA, Catalytic , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa , Rhodopsin , Animals , Dogs , HEK293 Cells , Humans , RNA, Catalytic/biosynthesis , RNA, Catalytic/genetics , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Rhodopsin/biosynthesis , Rhodopsin/genetics
6.
Vet Ophthalmol ; 24(6): 645-652, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34709702

ABSTRACT

PURPOSE: To describe the ophthalmoscopic, in-vivo imaging, fluorescein angiography, and therapeutic photocoagulation outcome in a case of bilateral optic nerve colobomas associated with focal unilateral retinal detachment in a dog. METHODS: Pretraining eye examination of a 1.6-year-old female German shepherd service dog showed a focal juxta-papillary bullous retinal separation in the right eye. In vivo imaging and angiography were performed under general anesthesia using optical coherence tomography. Nonoverlapping diode laser burns were applied through an operating microscope adapter to selected areas along the leading margins of the detachment. RESULTS: The funduscopic examination and in-vivo imaging revealed bilateral optic nerve colobomas associated with a focal bullous detachment in the right eye. Fluorescein angiography showed absence of blood vessel leakage and absence of staining inside of the retinal elevation. Photocoagulation induced immediate changes in retinal layer reflectivity. Three months post-photocoagulation, the retinal detachment had improved and scarring of the burns was visible. One and two years post-procedure, the retinal detachment resolved. CONCLUSIONS: Optical coherence tomography (OCT) imaging provides a detailed analysis of the retinal abnormalities associated with the clinical lesion. Laser retinopexy is a valid therapeutic option to limit the extension of the detachment.


Subject(s)
Coloboma , Dog Diseases , Retinal Detachment , Animals , Coloboma/surgery , Coloboma/veterinary , Dog Diseases/diagnostic imaging , Dog Diseases/surgery , Dogs , Female , Fluorescein Angiography , Lasers , Optic Nerve/abnormalities , Optic Nerve/diagnostic imaging , Optic Nerve/surgery , Retinal Detachment/surgery , Retinal Detachment/veterinary , Tomography, Optical Coherence/veterinary
7.
Exp Eye Res ; 192: 107926, 2020 03.
Article in English | MEDLINE | ID: mdl-31931002

ABSTRACT

The objectives of the present work were to assess by spectral domain optical coherence tomography (OCT) the changes in thickness of the outer nuclear layer (ONL), the ONL + photoreceptor inner segment (IS), and the retinal thickness, as a function of age in the normal canine retina. OCT retinal scans extending from the edge of the optic nerve head (ONH) along the superior and inferior meridians were captured in both eyes of 17 normal dogs at age ranging from 4 to 119 weeks. The different parameters along the superior and the inferior regions were determined following manual segmentation using the Heidelberg Eye Explorer software. Changes in thickness with age were modeled using one-phase exponential decay models. In vivo OCT imaging results showed no interocular statistically significant differences in ONL, ONL + IS, and retinal thickness at any age. All three parameters were however found to be statistically significantly thicker in the superior vs inferior retina. A rapid thinning of the three layers occurs in both the superior and inferior retina between 4 and 12 weeks of age, before reaching a plateau at around 20 weeks of age. In conclusion, the ONL, ONL + IS, and retinal thickness of the normal canine retina decrease significantly during the first three postnatal months, and is likely attributed to an overall increase in the eye volume and tangential dispersion of the photoreceptor since early photoreceptor developmental cell death is very limited at that age. Establishment of the natural history of ONL, ONL + IS, and retinal thinning will allow a more accurate assessment of the progression of a retinal degenerative condition as well as facilitate the detection of positive rescue effect of novel retinal therapies evaluated in this large animal model.


Subject(s)
Dogs/anatomy & histology , Retina/anatomy & histology , Aging/physiology , Animals , Female , Longitudinal Studies , Male , Models, Theoretical , Optic Disk/anatomy & histology , Optic Disk/diagnostic imaging , Organ Size , Retina/diagnostic imaging , Retinal Neurons/cytology , Retinal Photoreceptor Cell Inner Segment/physiology , Tomography, Optical Coherence
8.
Vet Ophthalmol ; 23(2): 292-304, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31746146

ABSTRACT

PURPOSE: To examine the in vivo microanatomy of retinal folds and geographic lesions in dogs with acquired or inherited retinal dysplasia. MATERIAL AND METHODS: Thirteen dogs had retinal microanatomy evaluation under general anesthesia using cSLO/sdOCT; two eyes had noninherited multifocal retinal folds, five had inherited multifocal retinal folds (drd1 or drd2), and 10 geographic retinal dysplasia. Retinas from two drd2 carrier dogs were examined by histology and immunohistochemistry (IHC) after in vivo imaging. RESULTS: Retinal folds are the common feature of acquired focal/multifocal or geographic retinal dysplasia, are indistinguishable structurally from those associated with syndromic oculoskeletal dysplasia, and represent outer nuclear layer invaginations and rosettes visible by sdOCT. In dogs heterozygous for oculoskeletal dysplasia, the folds form clusters in a perivascular distribution along superior central vessels. IHC confirmed photoreceptor identity in the retinal folds. The geographic dysplasia plaques are not focally detached, but have inner retinal disorganization and intense autofluorescence in cSLO autofluorescence mode that is mainly limited to the geographic lesion, but is not uniform and in some extends beyond the plaques. CONCLUSION: We propose that the autofluorescent characteristic of the geographic lesions is associated with an inner retinal disruption associated with perivascular or infiltrating macrophages and phagocytosis of cellular debris. As well, we suggest restructuring the examination forms to distinguish the folds that are sporadically distributed from those that have a perivascular distribution as the latter likely represent carriers for drd. In this latter group, DNA testing would be a helpful tool to provide specific breeding advice.


Subject(s)
Dog Diseases/pathology , Retinal Dysplasia/veterinary , Animals , Dog Diseases/genetics , Dogs , Female , Genetic Predisposition to Disease , Male , Retinal Dysplasia/genetics , Retinal Dysplasia/pathology
9.
Mol Ther ; 25(8): 1866-1880, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28566226

ABSTRACT

X-linked retinitis pigmentosa (XLRP) caused by mutations in the RPGR gene is an early onset and severe cause of blindness. Successful proof-of-concept studies in a canine model have recently shown that development of a corrective gene therapy for RPGR-XLRP may now be an attainable goal. In preparation for a future clinical trial, we have here optimized the therapeutic AAV vector construct by showing that GRK1 (rather than IRBP) is a more efficient promoter for targeting gene expression to both rods and cones in non-human primates. Two transgenes were used in RPGR mutant (XLPRA2) dogs under the control of the GRK1 promoter. First was the previously developed stabilized human RPGR (hRPGRstb). Second was a new full-length stabilized and codon-optimized human RPGR (hRPGRco). Long-term (>2 years) studies with an AAV2/5 vector carrying hRPGRstb under control of the GRK1 promoter showed rescue of rods and cones from degeneration and retention of vision. Shorter term (3 months) studies demonstrated comparable preservation of photoreceptors in canine eyes treated with an AAV2/5 vector carrying either transgene under the control of the GRK1 promoter. These results provide the critical molecular components (GRK1 promoter, hRPGRco transgene) to now construct a therapeutic viral vector optimized for RPGR-XLRP patients.


Subject(s)
Carrier Proteins/genetics , Eye Proteins/genetics , Genes, X-Linked , Genetic Therapy , Mutation , Retina/metabolism , Retinitis Pigmentosa/genetics , Animals , Dependovirus/genetics , Disease Models, Animal , Dogs , G-Protein-Coupled Receptor Kinase 1/genetics , Gene Expression , Gene Order , Genes, Reporter , Genetic Vectors/genetics , Humans , Phenotype , Photoreceptor Cells, Vertebrate/metabolism , Primates , Promoter Regions, Genetic , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/therapy , Transduction, Genetic , Transgenes , Vision Tests
10.
Adv Exp Med Biol ; 1074: 309-315, 2018.
Article in English | MEDLINE | ID: mdl-29721958

ABSTRACT

Canine bestrophinopathy (cBest) is an important translational model for BEST1-associated maculopathies in man that recapitulates the broad spectrum of clinical and molecular disease aspects observed in patients. Both human and canine bestrophinopathies are characterized by focal to multifocal separations of the retina from the RPE. The lesions can be macular or extramacular, and the specific pathomechanism leading to formation of these lesions remains unclear. We used the naturally occurring canine BEST1 model to examine factors that underlie formation of vitelliform lesions and addressed the susceptibility of the macula to its primary detachment in BEST1-linked maculopathies.


Subject(s)
Bestrophins/deficiency , Dog Diseases/pathology , Models, Animal , Retinal Pigment Epithelium/pathology , Vitelliform Macular Dystrophy/veterinary , Animals , Bestrophins/genetics , Bestrophins/physiology , Cytoskeletal Proteins/metabolism , Dog Diseases/genetics , Dog Diseases/metabolism , Dogs , Extracellular Matrix/pathology , Eye Proteins/metabolism , Genes, Recessive , Humans , Microvilli/pathology , Monocarboxylic Acid Transporters/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Detachment/etiology , Retinal Pigment Epithelium/metabolism , Species Specificity , Symporters/metabolism , Vitelliform Macular Dystrophy/genetics , Vitelliform Macular Dystrophy/metabolism , Vitelliform Macular Dystrophy/pathology
12.
Vet Ophthalmol ; 18(6): 457-61, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25522663

ABSTRACT

OBJECTIVE: To compare the effect of commercially available solution and compounded ointment formulations of dorzolamide(2%)-timolol(0.5%) on intraocular pressure (IOP) of normal horses. ANIMALS: Eighteen clinically normal horses. PROCEDURES: A randomized, masked prospective design was used with horses divided into two equal groups. One eye of each horse was selected for topical ophthalmic treatment with either 0.2 mL of dorzolamide(2%)-timolol(0.5%) solution or 0.2 g of dorzolamide(2%)-timolol(0.5%) ointment every 12 h for 5 days. The contralateral eye of horses in both groups was untreated. Rebound tonometry was performed every 6 h starting 2 days prior to and ending 2 days after the treatment period. RESULTS: The mean IOP reduction in eyes treated with the solution or ointment formulations was 13%. Untreated eyes in both groups experienced a lesser but still statistically significant reduction in IOP. The IOP values did not return to baseline within 48 h of the last treatment. CONCLUSIONS AND CLINICAL RELEVANCE: The commercially available solution and compounded ointment formulations of ophthalmic dorzolamide(2%)-timolol(0.5%) had similar effects on IOP in normal horses. Persistent IOP reduction following cessation of treatment may indicate prolonged drug effect or acclimation of horses to tonometry.


Subject(s)
Adrenergic beta-Antagonists/administration & dosage , Intraocular Pressure/drug effects , Sulfonamides/administration & dosage , Thiophenes/administration & dosage , Timolol/administration & dosage , Administration, Ophthalmic , Adrenergic beta-Antagonists/therapeutic use , Animals , Drug Combinations , Female , Horses , Male , Ointments , Ophthalmic Solutions , Sulfonamides/therapeutic use , Thiophenes/therapeutic use , Timolol/therapeutic use , Tonometry, Ocular/veterinary
13.
Vision Res ; 218: 108379, 2024 05.
Article in English | MEDLINE | ID: mdl-38460402

ABSTRACT

Mutations in BEST1 cause an autosomal recessive disease in dogs where the earliest changes localize to the photoreceptor-RPE interface and show a retina-wide micro-detachment that is modulated by light exposure. The purpose of this study was to define the spatial and temporal details of the outer retina and its response to light with ultra-high resolution OCT across a range of ages and with different BEST1 mutations. Three retinal regions were selected in each eye: near the fovea-like area, near the optic nerve, both in the tapetal area, and inferior to the optic nerve in the non-tapetal area. The OS+ slab thickness was defined between the peak near the junction of inner and outer segments (IS/OS) and the transition between basal RPE, Bruch membrane, choriocapillaris and proximal tapetum (RPE/T). In wildtype (WT) dogs, two tapetal regions showed additional hyperscattering OCT peaks within the OS+ slab likely representing cone and rod outer segment tips (COST and ROST). The inferior non-tapetal region of WT dogs had only one of these peaks, likely ROST. In dogs with BEST1 mutations, all three locations showed a single peak, likely suggesting optical silence of COST. Light-dependent expansion of the micro-detachment by about 10 um was detectable in both tapetal and non-tapetal retina across all ages and BEST1 mutations.


Subject(s)
Retina , Tomography, Optical Coherence , Dogs , Animals , Retinal Cone Photoreceptor Cells , Vision, Ocular
14.
Elife ; 102021 10 19.
Article in English | MEDLINE | ID: mdl-34664552

ABSTRACT

Background: Adeno-associated virus (AAV)-mediated gene therapies are rapidly advancing to the clinic, and AAV engineering has resulted in vectors with increased ability to deliver therapeutic genes. Although the choice of vector is critical, quantitative comparison of AAVs, especially in large animals, remains challenging. Methods: Here, we developed an efficient single-cell AAV engineering pipeline (scAAVengr) to simultaneously quantify and rank efficiency of competing AAV vectors across all cell types in the same animal. Results: To demonstrate proof-of-concept for the scAAVengr workflow, we quantified - with cell-type resolution - the abilities of naturally occurring and newly engineered AAVs to mediate gene expression in primate retina following intravitreal injection. A top performing variant identified using this pipeline, K912, was used to deliver SaCas9 and edit the rhodopsin gene in macaque retina, resulting in editing efficiency similar to infection rates detected by the scAAVengr workflow. scAAVengr was then used to identify top-performing AAV variants in mouse brain, heart, and liver following systemic injection. Conclusions: These results validate scAAVengr as a powerful method for development of AAV vectors. Funding: This work was supported by funding from the Ford Foundation, NEI/NIH, Research to Prevent Blindness, Foundation Fighting Blindness, UPMC Immune Transplant and Therapy Center, and the Van Sloun fund for canine genetic research.


Gene therapy is an experimental approach to treating disease that involves altering faulty genes or replacing them with new, working copies. Most often, the new genetic material is delivered into cells using a modified virus that no longer causes disease, called a viral vector. Virus-mediated gene therapies are currently being explored for degenerative eye diseases, such as retinitis pigmentosa, and neurological disorders, like Alzheimer's and Parkinson's disease. A number of gene therapies have also been approved for treating some rare cancers, blood disorders and a childhood form of motor neuron disease. Despite the promise of virus-mediated gene therapy, there are significant hurdles to its widespread success. Viral vectors need to deliver enough genetic material to the right cells without triggering an immune response or causing serious side effects. Selecting an optimal vector is key to achieving this. A type of viruses called adeno-associated viruses (AAV) are prime candidates, partly because they can be easily engineered. However, accurately comparing the safety and efficacy of newly engineered AAVs is difficult, due to variation between test subjects and the labor and cost involved in careful testing. Öztürk et al. addressed this issue by developing an experimental pipeline called scAAVengr for comparing gene therapy vectors head-to-head. The process involves tagging potential AAV vectors with unique genetic barcodes, which can then be detected and quantified in individual cells using a technique called single-cell RNA sequencing. This means that when several vectors are used to infect lab-grown cells or a test animal at the same time, they can be tracked. The vectors can then be ranked on their ability to infect specific cell types and deliver useful genetic material. Using scAAVengr, Öztürk et al. compared viral vectors designed to target the light-sensitive cells of the retina, which allow animals to see. First, a set of promising viral vectors were evaluated using the scAAVengr pipeline in the eyes of marmosets and macaques, two small primates. Precise levels and locations of gene delivery were quantified. The top-performing vector was then identified and used to deliver Cas9, a genome editing tool, to primate retinas. Öztürk et al. also used scAAVengr to compare viral vectors in mice, analysing the vectors' ability to deliver their genetic cargo to the brain, heart, and liver. These experiments demonstrated that scAAVengr can be used to evaluate vectors in multiple tissues and in different organisms. In summary, this work outlines a method for identifying and precisely quantifying the performance of top-performing viral vectors for gene therapy. By aiding the selection of optimal viral vectors, the scAAVengr pipeline could help to improve the success of preclinical studies and early clinical trials testing gene therapies.


Subject(s)
Dependovirus/physiology , Gene Expression Profiling/methods , Macaca fascicularis/physiology , Retina/physiology , Transcriptome , Transduction, Genetic , Animals , Genetic Vectors
15.
Hum Gene Ther ; 31(13-14): 743-755, 2020 07.
Article in English | MEDLINE | ID: mdl-32414297

ABSTRACT

Recombinant adeno-associated viral (rAAV) vector-mediated gene therapy is being developed to treat X-linked retinitis pigmentosa (XLRP) in patients with mutations in the retinitis pigmentosa GTPase regulator (RPGR) gene. In preparation for a clinical gene therapy trial, we conducted dose range finding (DRF) studies with an AAV2 capsid with three surface tyrosine residues changed to phenylalanine (AAV2tYF) vector administered by subretinal injection in a naturally occurring RPGR-mutant canine model (XLPRA2) to compare two different human RPGR (hRPGR) transgenes and to establish a reasonable starting dose for a clinical trial. Different dose levels of two candidate vectors (0.15 mL at 1.2 × 1010-3.0 × 1012 vg/mL of rAAV2tYF-GRK1-hRPGRco or 4 × 1010-3.0 × 1012 vg/mL of rAAV2tYF-GRK1-hRPGRstb), 6.0 × 1011 vg/mL rAAV5-GRK1-hRPGRco reference vector or Vehicle were subretinally administered, and the dogs were followed for 8 weeks postdose. Ophthalmic examinations, analyses of retinal structure by in vivo imaging using confocal scanning laser ophthalmoscopy (cSLO)/optical coherence tomography (OCT) in the Lower (4.0 × 1010 vg/mL) and Lowest (1.2 × 1010 vg/mL) Doses, immunological responses by cell based assays or enzyme-linked immunosorbent assay, RPGR transgene expression, and reversal of opsin mislocalization by immunohistochemistry were performed. No sustained signs of ocular discomfort or ophthalmic complications were noted in any of the injected eyes except some in the High Dose group (3.0 × 1012 vg/mL), which showed signs of retinal detachment and inflammation. A change in fundus reflectivity suggestive of a rescue effect was seen in the High, Mid (6.0 × 1011 vg/mL), and Low (1.2 × 1011 vg/mL) Dose groups. cSLO/OCT demonstrated qualitative and quantitative evidence of rescue effect in eyes treated with the Lower Dose. Anti-hRPGR antibodies were absent, but neutralizing antibody titers against AAV2 were detected in all animals dosed with rAAV2tYF in an apparent dose-related pattern. RPGR expression was stronger for rAAV2tYF-GRK1-hRPGRco compared to rAAV2tYF-GRK1-hRPGRstb at all dose levels. Subretinal administration of rAAV2tYF-GRK1-hRPGRco and rAAV2tYF-GRK1-hRPGRstb both corrected rod and cone opsin mislocalization, two early markers of disease in the XLPRA2 canine model of RPGR-XLRP. These results support the selection and use of rAAV2tYF-GRK1-hRPGRco (AGTC-501) and guided the initial doses in clinical studies in patients with XLRP caused by RPGR mutations.


Subject(s)
Dependovirus/genetics , Eye Proteins/genetics , Genetic Diseases, X-Linked/therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Mutation , Retinitis Pigmentosa/therapy , Animals , Dogs , Female , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , Genetic Vectors/genetics , Male , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Transgenes
16.
Hum Gene Ther ; 31(3-4): 253-267, 2020 02.
Article in English | MEDLINE | ID: mdl-31910043

ABSTRACT

Applied Genetic Technologies Corporation (AGTC) is developing a recombinant adeno-associated virus (rAAV) vector AGTC-501, also designated rAAV2tYF-GRK1-hRPGRco, to treat X-linked retinitis pigmentosa (XLRP) in patients with mutations in the retinitis pigmentosa GTPase regulator (RPGR) gene. The vector contains a codon-optimized human RPGR cDNA (hRPGRco) driven by a photoreceptor-specific promoter (G protein-coupled receptor kinase 1 [GRK1]), and is packaged in an AAV2 capsid variant with three surface tyrosine residues changed to phenylalanine (AAV2tYF). We conducted a toxicity and efficacy study of this vector administered by subretinal injection in the naturally occurring RPGR mutant (X-linked progressive retinal atrophy 2 [XLPRA2]) dog model. Sixteen RPGR mutant dogs divided into four groups of three to five animals each received either a subretinal injection of 0.07 mL of AGTC-501 at low (1.2 × 1011 vector genome [vg]/mL), mid (6 × 1011 vg/mL), or high dose (3 × 1012 vg/mL), or of vehicle control in the right eye at early-stage disease. The left eye remained untreated. Subretinal injections were well tolerated and were not associated with systemic toxicity. Electroretinography, in vivo retinal imaging, and histological analysis showed rescue of photoreceptor function and structure in the absence of ocular toxicity in the low- and mid-dose treatment groups when compared with the vehicle-treated group. The high-dose group showed evidence of both photoreceptor rescue and posterior segment toxicity. These results support the use of AGTC-501 in clinical studies with patients affected with XLRP caused by RPGR mutations and define the no-observed-adverse-effect level at 6 × 1011 vg/mL.


Subject(s)
Dependovirus/genetics , Eye Proteins/genetics , Genes, X-Linked , Genetic Therapy , Genetic Vectors/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Animals , Biomarkers , Biopsy , Cell Line , Codon , Dogs , Electroretinography , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Humans , Immunohistochemistry , Mutation , Retinitis Pigmentosa/diagnosis , Tomography, Optical Coherence
SELECTION OF CITATIONS
SEARCH DETAIL