Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Pharm Res ; 41(4): 807-817, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38443629

ABSTRACT

OBJECTIVE: Current gene therapy of inherited retinal diseases is achieved mainly by subretinal injection, which is invasive with severe adverse effects. Intravitreal injection is a minimally invasive alternative for gene therapy of inherited retinal diseases. This work explores the efficacy of intravitreal delivery of PEGylated ECO (a multifunctional pH-sensitive amphiphilic amino lipid) plasmid DNA (pGRK1-ABCA4-S/MAR) nanoparticles (PEG-ELNP) for gene therapy of Stargardt disease. METHODS: Pigmented Abca4-/- knockout mice received 1 µL of PEG-ELNP solution (200 ng/uL, pDNA concentration) by intravitreal injections at an interval of 1.5 months. The expression of ABCA4 in the retina was determined by RT-PCR and immunohistochemistry at 6 months after the second injection. A2E levels in the treated eyes and untreated controls were determined by HPLC. The safety of treatment was monitored by scanning laser ophthalmoscopy and electroretinogram (ERG). RESULTS: PEG-ELNP resulted in significant ABCA4 expression at both mRNA level and protein level at]6 months after 2 intravitreal injections, and a 40% A2E accumulation reduction compared with non-treated controls. The PEG-ELNP also demonstrated excellent safety as shown by scanning laser ophthalmoscopy, and the eye function evaluation from electroretinogram. CONCLUSIONS: Intravitreal delivery of the PEG-ELNP of pGRK1-ABCA4-S/MAR is a promising approach for gene therapy of Stargardt Disease, which can also be a delivery platform for gene therapy of other inherited retinal diseases.


Subject(s)
Nanoparticles , Retina , Mice , Animals , Stargardt Disease/genetics , Stargardt Disease/metabolism , Stargardt Disease/therapy , Retina/metabolism , Genetic Therapy/methods , Plasmids/genetics , DNA/metabolism , Mice, Knockout , Polyethylene Glycols/metabolism , Intravitreal Injections , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism
2.
Hum Gene Ther ; 34(13-14): 616-628, 2023 07.
Article in English | MEDLINE | ID: mdl-37227014

ABSTRACT

Adeno-associated virus (AAV)-based gene therapy has been shown to be safe and effective in numerous animal models and clinical trials for various ophthalmic diseases. Stargardt disease (STGD1; MIM #248200) is the most common autosomal recessive macular dystrophy disease, and the most common form is caused by mutations in the ABCA4 gene, a gene with 6.8 kb coding sequence. Split intein approaches increase the capacity of dual AAV gene therapy, but at the cost of reduced protein expression, which may be insufficient to achieve a therapeutic effect. In this study, we designed various dual split intein ABCA4 vectors and showed that the efficiency of expression of full-length ABCA4 protein is dependent on combinations of types and split sites of the intein system. The most efficient vectors were identified through in vitro screening, and a novel dual AAV8-ABCA4 vector was constructed and subsequently proven to express full-length ABCA4 protein at a high level, reducing bisretinoid formation and correcting the visual function of ABCA4-knockout mice. Furthermore, we evaluated therapeutic effects of different dosages by subretinal injection in mice model. Both therapeutic effects and safety were guaranteed under the treatment of 1.00 × 109 GC/eye. These results support the optimized dual AAV8-ABCA4 approach in future clinical translation for treatment of Stargardt disease.


Subject(s)
Macular Degeneration , Retinal Diseases , Mice , Animals , Stargardt Disease/genetics , Stargardt Disease/therapy , Macular Degeneration/genetics , Macular Degeneration/therapy , Genetic Therapy/methods , Mice, Knockout , Mutation , Retinal Diseases/therapy
3.
Am J Ophthalmol ; 240: 285-301, 2022 08.
Article in English | MEDLINE | ID: mdl-35248547

ABSTRACT

PURPOSE: To report on the safety of the first 5 cohorts of a gene therapy trial using recombinant equine infectious anemia virus expressing ABCA4 (EIAV-ABCA4) in adults with Stargardt dystrophy due to mutations in ABCA4. DESIGN: Nonrandomized multicenter phase I/IIa clinical trial. METHODS: Patients received a subretinal injection of EIAVABCA4 in the worse-seeing eye at 3 dose levels and were followed for 3 years after treatment. MAIN OUTCOME MEASURES: The primary end point was ocular and systemic adverse events. The secondary end points were best-corrected visual acuity, static perimetry, kinetic perimetry, total field hill of vision, full field electroretinogram, multifocal ERG, color fundus photography, short-wavelength fundus autofluorescence, and spectral domain optical coherence tomography. RESULTS: The subretinal injections were well tolerated by all 22 patients across 3 dose levels. There was 1 case of a treatment-related ophthalmic serious adverse event in the form of chronic ocular hypertension. The most common adverse events were associated with the surgical procedure. In 1 patient treated with the highest dose, there was a significant decline in the number of macular flecks as compared with the untreated eye. However, in 6 patients, hypoautofluorescent changes were worse in the treated eye than in the untreated eye. Of these, 1 patient had retinal pigment epithelium atrophy that was characteristic of tissue damage likely associated with bleb induction. No patients had any clinically significant changes in best-corrected visual acuity, static perimetry, kinetic perimetry, total field hill of vision, full field electroretinogram, or multifocal ERG attributable to the treatment. CONCLUSIONS: Subretinal treatment with EIAV-ABCA4 was well tolerated with only 1 case of ocular hypertension. No clinically significant changes in visual function tests were found to be attributable to the treatment. However, 27% of treated eyes showed exacerbation of retinal pigment epithelium atrophy on fundus autofluorescence. There was a significant reduction in macular flecks in 1 treated eye from the highest dose cohort. Additional follow-up and continued investigation in more patients will be required to fully characterize the safety and efficacy of EIAV-ABCA4.


Subject(s)
Genetic Therapy , Stargardt Disease , ATP-Binding Cassette Transporters/genetics , Atrophy , Electroretinography , Fluorescein Angiography , Genetic Therapy/methods , Humans , Infectious Anemia Virus, Equine/genetics , Ocular Hypertension , Retinal Degeneration , Stargardt Disease/therapy , Tomography, Optical Coherence , Visual Acuity
4.
Int J Mol Sci ; 23(2)2022 Jan 08.
Article in English | MEDLINE | ID: mdl-35054869

ABSTRACT

The retinal pigmented epithelium (RPE) plays a pivotal role in retinal homeostasis. It is therefore an interesting target to fill the unmet medical need of different retinal diseases, including age-related macular degeneration and Stargardt disease. RPE replacement therapy may use different cellular sources: induced pluripotent stem cells or embryonic stem cells. Cells can be transferred as suspension on a patch with different surgical approaches. Results are promising although based on very limited samples. In this review, we summarize the current progress of RPE replacement and provide a comparative assessment of different published approaches which may become standard of care in the future.


Subject(s)
Ophthalmologists , Retinal Pigment Epithelium/pathology , Translational Research, Biomedical , Clinical Trials as Topic , Humans , Macular Degeneration/therapy , Stargardt Disease/therapy
5.
Biomolecules ; 11(8)2021 08 09.
Article in English | MEDLINE | ID: mdl-34439845

ABSTRACT

Despite being the most prevalent cause of inherited blindness in children, Stargardt disease is yet to achieve the same clinical trial success as has been achieved for other inherited retinal diseases. With an early age of onset and continual progression of disease over the life course of an individual, Stargardt disease appears to lend itself to therapeutic intervention. However, the aetiology provides issues not encountered with the likes of choroideremia and X-linked retinitis pigmentosa and this has led to a spectrum of treatment strategies that approach the problem from different aspects. These include therapeutics ranging from small molecules and anti-sense oligonucleotides to viral gene supplementation and cell replacement. The advancing development of CRISPR-based molecular tools is also likely to contribute to future therapies by way of genome editing. In this we review, we consider the most recent pre-clinical and clinical trial data relating to the different strategies being applied to the problem of generating a treatment for the large cohort of Stargardt disease patients worldwide.


Subject(s)
Gene Editing/methods , Genetic Therapy , Stargardt Disease/therapy , Animals , Cell Line , Child , Clinical Trials as Topic , Humans , Mice
6.
Int J Mol Sci ; 22(4)2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33579019

ABSTRACT

Progenitor cells derived from the retinal pigment epithelium (RPECs) have shown promise as therapeutic approaches to degenerative retinal disorders including diabetic retinopathy, age-related macular degeneration and Stargardt disease. However, the degeneration of Bruch's membrane (BM), the natural substrate for the RPE, has been identified as one of the major limitations for utilizing RPECs. This degeneration leads to decreased support, survival and integration of the transplanted RPECs. It has been proposed that the generation of organized structures of nanofibers, in an attempt to mimic the natural retinal extracellular matrix (ECM) and its unique characteristics, could be utilized to overcome these limitations. Furthermore, nanoparticles could be incorporated to provide a platform for improved drug delivery and sustained release of molecules over several months to years. In addition, the incorporation of tissue-specific genes and stem cells into the nanostructures increased the stability and enhanced transfection efficiency of gene/drug to the posterior segment of the eye. This review discusses available drug delivery systems and combination therapies together with challenges associated with each approach. As the last step, we discuss the application of nanofibrous scaffolds for the implantation of RPE progenitor cells with the aim to enhance cell adhesion and support a functionally polarized RPE monolayer.


Subject(s)
Drug Carriers/chemistry , Nanofibers/chemistry , Retinal Diseases/therapy , Retinal Pigment Epithelium/transplantation , Stem Cell Transplantation/methods , Tissue Scaffolds/chemistry , Animals , Bruch Membrane/chemistry , Diabetic Retinopathy/therapy , Drug Delivery Systems/methods , Humans , Macular Degeneration/therapy , Retinal Pigment Epithelium/cytology , Stargardt Disease/therapy , Stem Cells/cytology
7.
Am J Med Genet C Semin Med Genet ; 184(3): 828-837, 2020 09.
Article in English | MEDLINE | ID: mdl-32893963

ABSTRACT

Genetic testing in a multisite clinical trial network for inherited eye conditions is described in this retrospective review of data collected through eyeGENE®, the National Ophthalmic Disease Genotyping and Phenotyping Network. Participants in eyeGENE were enrolled through a network of clinical providers throughout the United States and Canada. Blood samples and clinical data were collected to establish a phenotype:genotype database, biorepository, and patient registry. Data and samples are available for research use, and participants are provided results of clinical genetic testing. eyeGENE utilized a unique, distributed clinical trial design to enroll 6,403 participants from 5,385 families diagnosed with over 30 different inherited eye conditions. The most common diagnoses given for participants were retinitis pigmentosa (RP), Stargardt disease, and choroideremia. Pathogenic variants were most frequently reported in ABCA4 (37%), USH2A (7%), RPGR (6%), CHM (5%), and PRPH2 (3%). Among the 5,552 participants with genetic testing, at least one pathogenic or likely pathogenic variant was observed in 3,448 participants (62.1%), and variants of uncertain significance in 1,712 participants (30.8%). Ten genes represent 68% of all pathogenic and likely pathogenic variants in eyeGENE. Cross-referencing current gene therapy clinical trials, over a thousand participants may be eligible, based on pathogenic variants in genes targeted by those therapies. This article is the first summary of genetic testing from thousands of participants tested through eyeGENE, including reports from 5,552 individuals. eyeGENE provides a launching point for inherited eye research, connects researchers with potential future study participants, and provides a valuable resource to the vision community.


Subject(s)
Choroideremia/genetics , Eye Diseases, Hereditary/genetics , Retinitis Pigmentosa/genetics , Stargardt Disease/genetics , ATP-Binding Cassette Transporters/genetics , Adaptor Proteins, Signal Transducing/genetics , Choroideremia/diagnosis , Choroideremia/epidemiology , Choroideremia/therapy , Extracellular Matrix Proteins/genetics , Eye Diseases, Hereditary/diagnosis , Eye Diseases, Hereditary/epidemiology , Eye Diseases, Hereditary/therapy , Eye Proteins/genetics , Female , Genetic Testing/trends , Genetic Therapy/trends , Humans , Male , Peripherins/genetics , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/epidemiology , Retinitis Pigmentosa/therapy , Stargardt Disease/diagnosis , Stargardt Disease/epidemiology , Stargardt Disease/therapy
8.
Mol Ther ; 28(1): 293-303, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31611143

ABSTRACT

Stargardt disease (STGD) is an autosomal recessive retinal disorder caused by a monogenic ABCA4 mutation. Currently, there is no effective therapy to cure Stargardt disease. The replacement of mutated ABCA4 with a functional gene remains an attractive strategy. In this study, we have developed a non-viral gene therapy using nanoparticles self-assembled by a multifunctional pH-sensitive amino lipid ECO and a therapeutic ABCA4 plasmid. The nanoparticles mediated efficient intracellular gene transduction in wild-type (WT) and Abca4-/- mice. Specific ABCA4 expression in the outer segment of photoreceptors was achieved by incorporating a rhodopsin promoter into the plasmids. The ECO/pRHO-ABCA4 nanoparticles induced substantial and specific ABCA4 expression for at least 8 months, 35% reduction in A2E accumulation on average, and a delayed Stargardt disease progression for at least 6 months in Abca4-/- mice. ECO/plasmid nanoparticles constitute a promising non-viral gene therapy platform for Stargardt disease and other visual dystrophies.


Subject(s)
ATP-Binding Cassette Transporters/administration & dosage , ATP-Binding Cassette Transporters/metabolism , Drug Delivery Systems/methods , Genetic Therapy/methods , Lipopeptides/administration & dosage , Nanoparticles/chemistry , Rhodopsin/administration & dosage , Stargardt Disease/therapy , ATP-Binding Cassette Transporters/genetics , Animals , Cell Line , Disease Models, Animal , Humans , Lipopeptides/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Photoreceptor Cells/metabolism , Plasmids/genetics , Plasmids/therapeutic use , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Rhodopsin/genetics , Stargardt Disease/genetics , Transfection
9.
Hum Gene Ther ; 30(11): 1361-1370, 2019 11.
Article in English | MEDLINE | ID: mdl-31418294

ABSTRACT

Autosomal recessive Stargardt disease is the most common inherited macular degeneration in humans. It is caused by mutations in the retina-specific ATP binding cassette transporter A4 (ABCA4) that is essential for the clearance of all-trans-retinal from photoreceptor cells. Loss of this function results in the accumulation of toxic bisretinoids in the outer segment disk membranes and their subsequent transfer into adjacent retinal pigment epithelium (RPE) cells. This ultimately leads to the Stargardt disease phenotype of increased retinal autofluorescence and progressive RPE and photoreceptor cell loss. Adeno-associated virus (AAV) vectors have been widely used in gene therapeutic applications, but their limited cDNA packaging capacity of ∼4.5 kb has impeded their use for transgenes exceeding this limit. AAV dual vectors were developed to overcome this size restriction. In this study, we have evaluated the in vitro expression of ABCA4 using three options: overlap, transplicing, and hybrid ABCA4 dual vector systems. The hybrid system was the most efficient of these dual vector alternatives and used to express the full-length ABCA4 in Abca4-/- mice. The full-length ABCA4 protein correctly localized to photoreceptor outer segments. Moreover, treatment of Abca4-/- mice with this ABCA4 hybrid dual vector system resulted in a reduced accumulation of the lipofuscin/N-retinylidene-N-retinylethanolamine (A2E) autofluorescence in vivo, and retinal A2E quantification supported these findings. These results show that the hybrid AAV dual vector option is both safe and therapeutic in mice, and the delivered ABCA4 transgene is functional and has a significant effect on reducing A2E accumulation in the Abca4-/- mouse model of Stargardt disease.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/therapeutic use , Dependovirus/genetics , Genes, Recessive , Genetic Vectors/metabolism , Retina/pathology , Stargardt Disease/genetics , Stargardt Disease/therapy , Animals , Disease Models, Animal , Fluorescence , Fundus Oculi , HEK293 Cells , Humans , Mice, Inbred C57BL , Retina/metabolism , Retinoids/metabolism
10.
Expert Opin Biol Ther ; 19(4): 335-342, 2019 04.
Article in English | MEDLINE | ID: mdl-30686077

ABSTRACT

INTRODUCTION: Dry age-related macular degeneration (AMD) and Stargardt Macular Dystrophy (STGD1) result in vision loss due to progressive atrophy of the macula and lack of effective treatments. Numerous studies have implicated complement-associated inflammation as a contributor to both diseases. AREAS COVERED: The complement factor D inhibitor, lampalizumab, failed to halt geographic atrophy (GA) progression in phase 3 studies. The complement factor 3 (C3) inhibitor, APL-2, has shown potential to reduce GA growth in a phase 2 trial, supporting advancement to phase 3 trials. The intravenous complement factor 5 (C5) inhibitor, eculizumab, failed to halt GA progression in a phase 2 study. Another C5 inhibitor, avacincaptad pegol, is delivered by intravitreal injection, and will be studied for safety and preliminary signs of efficacy for AMD and STGD1 patients in phase 2 trials. LFG316 (C5 inhibitor) and CLG561 (properdin inhibitor) failed to halt GA progression in phase 2 studies. A phase 1 trial is evaluating the effects of combining LFG316 and CL561. Complement inhibition by gene therapy will be explored in the phase 1 trial of HMR59 in AMD patients. EXPERT OPINION: While complement inhibition has not yet demonstrated the ability to halt GA progression in a phase 3 trial, further study is warranted.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Geographic Atrophy/drug therapy , Immunoglobulin Fab Fragments/therapeutic use , Stargardt Disease/drug therapy , Clinical Trials as Topic , Complement C3/immunology , Complement C5/immunology , Complement Factor D/immunology , Genetic Therapy , Geographic Atrophy/pathology , Geographic Atrophy/therapy , Humans , Stargardt Disease/pathology , Stargardt Disease/therapy
11.
Hum Gene Ther ; 30(5): 590-600, 2019 05.
Article in English | MEDLINE | ID: mdl-30381971

ABSTRACT

The recent approval in the United States of the first adeno-associated viral (AAV) vector for the treatment of an inherited retinal degeneration validates this approach for the treatment of many other diseases. A major limiting factor continues to be the size restriction of the AAV transgene at under 5 kb. Stargardt disease is the most prevalent form of recessively inherited blindness and is caused by mutations in ABCA4, the gene that codes for ATP-binding cassette transporter protein family member 4, which has a coding sequence length of 6.8 kb. Dual vector approaches increase the capacity of AAV gene therapy, but at the cost of substantially reduced levels of target protein, which may be insufficient to achieve a therapeutic effect. Here we show that the efficacy of recombination of dual vectors is dependent on the length of DNA overlap between two transgenes. With optimized recombination, full-length ABCA4 protein is expressed in the photoreceptor outer segments of Abca4-/- mice at levels sufficient to reduce bisretinoid formation and correct the autofluorescent phenotype. These observations support a dual vector approach in future clinical trials using AAV gene therapy to treat Stargardt disease.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Dependovirus/genetics , Genetic Vectors/genetics , Phenotype , Stargardt Disease/genetics , Transgenes , ATP-Binding Cassette Transporters/metabolism , Animals , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line, Tumor , Chromatography, High Pressure Liquid , Disease Models, Animal , Humans , Mice, Knockout , Open Reading Frames , Optical Imaging , Photoreceptor Cells/metabolism , Stargardt Disease/diagnosis , Stargardt Disease/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...