1
|
Igoe JM, Lam BL, Gregori NZ. Update on Clinical Trial Endpoints in Gene Therapy Trials for Inherited Retinal Diseases. J Clin Med 2024; 13:5512. [PMID: 39336999 PMCID: PMC11431936 DOI: 10.3390/jcm13185512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Inherited retinal diseases (IRDs) encompass a wide spectrum of rare conditions characterized by diverse phenotypes associated with hundreds of genetic variations, often leading to progressive visual impairment and profound vision loss. Multiple natural history studies and clinical trials exploring gene therapy for various IRDs are ongoing. Outcomes for ophthalmic trials measure visual changes in three main categories-structural, functional, and patient-focused outcomes. Since IRDs may range from congenital with poor central vision from birth to affecting the peripheral retina initially and progressing insidiously with visual acuity affected late in the disease course, typical outcome measures such as central visual acuity and ocular coherence tomography (OCT) imaging of the macula may not provide adequate representation of therapeutic outcomes including alterations in disease course. Thus, alternative unique outcome measures are necessary to assess loss of peripheral vision, color vision, night vision, and contrast sensitivity in IRDs. These differences have complicated the assessment of clinical outcomes for IRD therapies, and the clinical trials for IRDs have had to design novel specialized endpoints to demonstrate treatment efficacy. As genetic engineering and gene therapy techniques continue to advance with growing investment from industry and accelerated approval tracks for orphan conditions, the clinical trials must continue to improve their assessments to demonstrate safety and efficacy of new gene therapies that aim to come to market. Here, we will provide an overview of the current gene therapy approaches, review various endpoints for measuring visual function, highlight those that are utilized in recent gene therapy trials, and provide an overview of stage 2 and 3 IRD trials through the second quarter of 2024.
Collapse
Affiliation(s)
- Jane M Igoe
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Byron L Lam
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ninel Z Gregori
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Miami Veterans Administration Medical Center, Miami, FL 33125, USA
| |
Collapse
|
2
|
Aweidah H, Xi Z, Sahel JA, Byrne LC. PRPF31-retinitis pigmentosa: Challenges and opportunities for clinical translation. Vision Res 2023; 213:108315. [PMID: 37714045 PMCID: PMC10872823 DOI: 10.1016/j.visres.2023.108315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/17/2023]
Abstract
Mutations in pre-mRNA processing factor 31 cause autosomal dominant retinitis pigmentosa (PRPF31-RP), for which there is currently no efficient treatment, making this disease a prime target for the development of novel therapeutic strategies. PRPF31-RP exhibits incomplete penetrance due to haploinsufficiency, in which reduced levels of gene expression from the mutated allele result in disease. A variety of model systems have been used in the investigation of disease etiology and therapy development. In this review, we discuss recent advances in both in vivo and in vitro model systems, evaluating their advantages and limitations in the context of therapy development for PRPF31-RP. Additionally, we describe the latest approaches for treatment, including AAV-mediated gene augmentation, genome editing, and late-stage therapies such as optogenetics, cell transplantation, and retinal prostheses.
Collapse
Affiliation(s)
- Hamzah Aweidah
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhouhuan Xi
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Ophthalmology, Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - José-Alain Sahel
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leah C Byrne
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Sp S, Mitra RN, Zheng M, Chrispell JD, Wang K, Kwon YS, Weiss ER, Han Z. Gene augmentation for autosomal dominant retinitis pigmentosa using rhodopsin genomic loci nanoparticles in the P23H +/- knock-in murine model. Gene Ther 2023; 30:628-640. [PMID: 36935427 DOI: 10.1038/s41434-023-00394-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 02/13/2023] [Accepted: 02/28/2023] [Indexed: 03/21/2023]
Abstract
Gene therapy for autosomal dominant retinitis pigmentosa (adRP) is challenged by the dominant inheritance of the mutant genes, which would seemingly require a combination of mutant suppression and wild-type replacement of the appropriate gene. We explore the possibility that delivery of a nanoparticle (NP)-mediated full-length mouse genomic rhodopsin (gRho) or human genomic rhodopsin (gRHO) locus can overcome the dominant negative effects of the mutant rhodopsin in the clinically relevant P23H+/--knock-in heterozygous mouse model. Our results demonstrate that mice in both gRho and gRHO NP-treated groups exhibit significant structural and functional recovery of the rod photoreceptors, which lasted for 3 months post-injection, indicating a promising reduction in photoreceptor degeneration. We performed miRNA transcriptome analysis using next generation sequencing and detected differentially expressed miRNAs as a first step towards identifying miRNAs that could potentially be used as rhodopsin gene expression enhancers or suppressors for sustained photoreceptor rescue. Our results indicate that delivering an intact genomic locus as a transgene has a greater chance of success compared to the use of the cDNA for treatment of this model of adRP, emphasizing the importance of gene augmentation using a gDNA that includes regulatory elements.
Collapse
Affiliation(s)
- Simna Sp
- Department of Ophthalmology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Rajendra N Mitra
- Department of Ophthalmology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Min Zheng
- Department of Ophthalmology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jared D Chrispell
- Department of Cell Biology and Physiology, the University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Kai Wang
- Department of Ophthalmology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yong-Su Kwon
- Department of Ophthalmology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ellen R Weiss
- Department of Cell Biology and Physiology, the University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Zongchao Han
- Department of Ophthalmology, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Carolina Institute for NanoMedicine, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
4
|
Panikker P, Roy S, Ghosh A, Poornachandra B, Ghosh A. Advancing precision medicines for ocular disorders: Diagnostic genomics to tailored therapies. Front Med (Lausanne) 2022; 9:906482. [PMID: 35911417 PMCID: PMC9334564 DOI: 10.3389/fmed.2022.906482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/29/2022] [Indexed: 11/20/2022] Open
Abstract
Successful sequencing of the human genome and evolving functional knowledge of gene products has taken genomic medicine to the forefront, soon combining broadly with traditional diagnostics, therapeutics, and prognostics in patients. Recent years have witnessed an extraordinary leap in our understanding of ocular diseases and their respective genetic underpinnings. As we are entering the age of genomic medicine, rapid advances in genome sequencing, gene delivery, genome surgery, and computational genomics enable an ever-increasing capacity to provide a precise and robust diagnosis of diseases and the development of targeted treatment strategies. Inherited retinal diseases are a major source of blindness around the world where a large number of causative genes have been identified, paving the way for personalized diagnostics in the clinic. Developments in functional genetics and gene transfer techniques has also led to the first FDA approval of gene therapy for LCA, a childhood blindness. Many such retinal diseases are the focus of various clinical trials, making clinical diagnoses of retinal diseases, their underlying genetics and the studies of natural history important. Here, we review methodologies for identifying new genes and variants associated with various ocular disorders and the complexities associated with them. Thereafter we discuss briefly, various retinal diseases and the application of genomic technologies in their diagnosis. We also discuss the strategies, challenges, and potential of gene therapy for the treatment of inherited and acquired retinal diseases. Additionally, we discuss the translational aspects of gene therapy, the important vector types and considerations for human trials that may help advance personalized therapeutics in ophthalmology. Retinal disease research has led the application of precision diagnostics and precision therapies; therefore, this review provides a general understanding of the current status of precision medicine in ophthalmology.
Collapse
Affiliation(s)
| | - Shomereeta Roy
- Grow Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| | - Anuprita Ghosh
- Grow Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| | | | - Arkasubhra Ghosh
- Grow Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| |
Collapse
|
5
|
Wu WH, Tsai YT, Huang IW, Cheng CH, Hsu CW, Cui X, Ryu J, Quinn PMJ, Caruso SM, Lin CS, Tsang SH. CRISPR genome surgery in a novel humanized model for autosomal dominant retinitis pigmentosa. Mol Ther 2022; 30:1407-1420. [PMID: 35150888 PMCID: PMC9077379 DOI: 10.1016/j.ymthe.2022.02.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/27/2022] [Accepted: 02/07/2022] [Indexed: 11/21/2022] Open
Abstract
Mutations in rhodopsin (RHO) are the most common causes of autosomal dominant retinitis pigmentosa (adRP), accounting for 20% to 30% of all cases worldwide. However, the high degree of genetic heterogeneity makes development of effective therapies cumbersome. To provide a universal solution to RHO-related adRP, we devised a CRISPR-based, mutation-independent gene ablation and replacement (AR) compound therapy carried by a dual AAV2/8 system. Moreover, we developed a novel hRHOC110R/hRHOWT humanized mouse model to assess the AR treatment in vivo. Results show that this humanized RHO mouse model exhibits progressive rod-cone degeneration that phenocopies hRHOC110R/hRHOWT patients. In vivo transduction of AR AAV8 dual vectors remarkably ablates endogenous RHO expression and overexpresses exogenous WT hRHO. Furthermore, the administration of AR during adulthood significantly hampers photoreceptor degeneration both histologically and functionally for at least 6 months compared with sole gene replacement or surgical trauma control. This study demonstrates the effectiveness of AR treatment of adRP in the human genomic context while revealing the feasibility of its application for other autosomal dominant disorders.
Collapse
Affiliation(s)
- Wen-Hsuan Wu
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital/Columbia University Medical Center, New York, NY 10032, USA
| | - Yi-Ting Tsai
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - I-Wen Huang
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital/Columbia University Medical Center, New York, NY 10032, USA
| | - Chia-Hua Cheng
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital/Columbia University Medical Center, New York, NY 10032, USA
| | - Chun-Wei Hsu
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital/Columbia University Medical Center, New York, NY 10032, USA
| | - Xuan Cui
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital/Columbia University Medical Center, New York, NY 10032, USA
| | - Joseph Ryu
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital/Columbia University Medical Center, New York, NY 10032, USA
| | - Peter M J Quinn
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital/Columbia University Medical Center, New York, NY 10032, USA
| | | | - Chyuang-Sheng Lin
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Stephen H Tsang
- Jonas Children's Vision Care and the Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA; Edward S. Harkness Eye Institute, New York-Presbyterian Hospital/Columbia University Medical Center, New York, NY 10032, USA; Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA; Department of Pathology & Cell Biology, Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
6
|
Fenner BJ, Tan TE, Barathi AV, Tun SBB, Yeo SW, Tsai ASH, Lee SY, Cheung CMG, Chan CM, Mehta JS, Teo KYC. Gene-Based Therapeutics for Inherited Retinal Diseases. Front Genet 2022; 12:794805. [PMID: 35069693 PMCID: PMC8782148 DOI: 10.3389/fgene.2021.794805] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
Inherited retinal diseases (IRDs) are a heterogenous group of orphan eye diseases that typically result from monogenic mutations and are considered attractive targets for gene-based therapeutics. Following the approval of an IRD gene replacement therapy for Leber's congenital amaurosis due to RPE65 mutations, there has been an intensive international research effort to identify the optimal gene therapy approaches for a range of IRDs and many are now undergoing clinical trials. In this review we explore therapeutic challenges posed by IRDs and review current and future approaches that may be applicable to different subsets of IRD mutations. Emphasis is placed on five distinct approaches to gene-based therapy that have potential to treat the full spectrum of IRDs: 1) gene replacement using adeno-associated virus (AAV) and nonviral delivery vectors, 2) genome editing via the CRISPR/Cas9 system, 3) RNA editing by endogenous and exogenous ADAR, 4) mRNA targeting with antisense oligonucleotides for gene knockdown and splicing modification, and 5) optogenetic approaches that aim to replace the function of native retinal photoreceptors by engineering other retinal cell types to become capable of phototransduction.
Collapse
Affiliation(s)
- Beau J Fenner
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Graduate Medical School, Ophthalmology and Visual Sciences Academic Clinical Programme, Singapore, Singapore
| | - Tien-En Tan
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Graduate Medical School, Ophthalmology and Visual Sciences Academic Clinical Programme, Singapore, Singapore
| | | | - Sai Bo Bo Tun
- Singapore Eye Research Institute, Singapore, Singapore
| | - Sia Wey Yeo
- Singapore Eye Research Institute, Singapore, Singapore
| | - Andrew S H Tsai
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Graduate Medical School, Ophthalmology and Visual Sciences Academic Clinical Programme, Singapore, Singapore
| | - Shu Yen Lee
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Graduate Medical School, Ophthalmology and Visual Sciences Academic Clinical Programme, Singapore, Singapore
| | - Chui Ming Gemmy Cheung
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Graduate Medical School, Ophthalmology and Visual Sciences Academic Clinical Programme, Singapore, Singapore
| | - Choi Mun Chan
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Graduate Medical School, Ophthalmology and Visual Sciences Academic Clinical Programme, Singapore, Singapore
| | - Jodhbir S Mehta
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Graduate Medical School, Ophthalmology and Visual Sciences Academic Clinical Programme, Singapore, Singapore.,School of Material Science and Engineering, Nanyang Technological University, Singapore, Singapore.,Yong Loo Lin School of Medicine, Department of Ophthalmology, National University of Singapore, Singapore, Singapore
| | - Kelvin Y C Teo
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Graduate Medical School, Ophthalmology and Visual Sciences Academic Clinical Programme, Singapore, Singapore
| |
Collapse
|
7
|
Shughoury A, Ciulla TA, Bakall B, Pennesi ME, Kiss S, Cunningham ET. Genes and Gene Therapy in Inherited Retinal Disease. Int Ophthalmol Clin 2021; 61:3-45. [PMID: 34584043 DOI: 10.1097/iio.0000000000000377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
8
|
Nuzbrokh Y, Ragi SD, Tsang SH. Gene therapy for inherited retinal diseases. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1278. [PMID: 34532415 PMCID: PMC8421966 DOI: 10.21037/atm-20-4726] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/04/2020] [Indexed: 01/04/2023]
Abstract
Inherited retinal diseases (IRDs) are a genetically variable collection of devastating disorders that lead to significant visual impairment. Advances in genetic characterization over the past two decades have allowed identification of over 260 causative mutations associated with inherited retinal disorders. Thought to be incurable, gene supplementation therapy offers great promise in treating various forms of these blinding conditions. In gene replacement therapy, a disease-causing gene is replaced with a functional copy of the gene. These therapies are designed to slow disease progression and hopefully restore visual function. Gene therapies are typically delivered to target retinal cells by subretinal (SR) or intravitreal (IVT) injection. The historic Food and Drug Administration (FDA) approval of voretigene neparvovec for RPE65-associated Leber's congenital amaurosis (LCA) spurred tremendous optimism surrounding retinal gene therapy for various other monogenic IRDs. Novel disease-causing mutations continue to be discovered annually, and targeted genetic therapy is now under development in clinical and preclinical models for many IRDs. Numerous clinical trials for other IRDs are ongoing or have recently completed. Disorders being targeted for genetic therapy include retinitis pigmentosa (RP), choroideremia (CHM), achromatopsia (ACHM), Leber's hereditary optic neuropathy, usher syndrome (USH), X-linked retinoschisis, and Stargardt disease. Here, we provide an update of completed, ongoing, and planned clinical trials using gene supplementation strategies for retinal degenerative disorders.
Collapse
Affiliation(s)
- Yan Nuzbrokh
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York, NY, USA.,Jonas Children's Vision Care, New York, NY, USA.,Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, NY, USA
| | - Sara D Ragi
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York, NY, USA.,Jonas Children's Vision Care, New York, NY, USA
| | - Stephen H Tsang
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, New York, NY, USA.,Jonas Children's Vision Care, New York, NY, USA.,Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
9
|
Botto C, Rucli M, Tekinsoy MD, Pulman J, Sahel JA, Dalkara D. Early and late stage gene therapy interventions for inherited retinal degenerations. Prog Retin Eye Res 2021; 86:100975. [PMID: 34058340 DOI: 10.1016/j.preteyeres.2021.100975] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/18/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022]
Abstract
Inherited and age-related retinal degeneration is the hallmark of a large group of heterogeneous diseases and is the main cause of untreatable blindness today. Genetic factors play a major pathogenic role in retinal degenerations for both monogenic diseases (such as retinitis pigmentosa) and complex diseases with established genetic risk factors (such as age-related macular degeneration). Progress in genotyping techniques and back of the eye imaging are completing our understanding of these diseases and their manifestations in patient populations suffering from retinal degenerations. It is clear that whatever the genetic cause, the majority of vision loss in retinal diseases results from the loss of photoreceptor function. The timing and circumstances surrounding the loss of photoreceptor function determine the adequate therapeutic approach to use for each patient. Among such approaches, gene therapy is rapidly becoming a therapeutic reality applicable in the clinic. This massive move from laboratory work towards clinical application has been propelled by the advances in our understanding of disease genetics and mechanisms, gene delivery vectors, gene editing systems, and compensatory strategies for loss of photoreceptor function. Here, we provide an overview of existing modalities of retinal gene therapy and their relevance based on the needs of patient populations suffering from inherited retinal degenerations.
Collapse
Affiliation(s)
- Catherine Botto
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Marco Rucli
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Müge Defne Tekinsoy
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Juliette Pulman
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France; Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, United States; CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, F-75012, Paris, France; Fondation Ophtalmologique Rothschild, F-75019, Paris, France
| | - Deniz Dalkara
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France.
| |
Collapse
|
10
|
Maldonado R, Jalil S, Wartiovaara K. Curative gene therapies for rare diseases. J Community Genet 2021; 12:267-276. [PMID: 32803721 PMCID: PMC8141081 DOI: 10.1007/s12687-020-00480-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023] Open
Abstract
Diseases caused by alterations in the DNA can be overcome by providing the cells or tissues with a functional copy of the mutated gene. The most common form of gene therapy implies adding an extra genetic unit into the cell. However, new genome engineering techniques also allow the modification or correction of the existing allele, providing new possibilities, especially for dominant diseases. Gene therapies have been tested for 30 years in thousands of clinical trials, but presently, we have only three authorised gene therapy products for the treatment of inherited diseases in European Union. Here, we describe the gene therapy alternatives already on the market in the European Union and expand the scope to some clinical trials. Additionally, we discuss the ethical and regulatory issues raised by the development of these new kinds of therapies.
Collapse
Affiliation(s)
- Rocio Maldonado
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Sami Jalil
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Kirmo Wartiovaara
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland.
- Clinical Genetics, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
11
|
Hu ML, Edwards TL, O'Hare F, Hickey DG, Wang JH, Liu Z, Ayton LN. Gene therapy for inherited retinal diseases: progress and possibilities. Clin Exp Optom 2021; 104:444-454. [PMID: 33689657 DOI: 10.1080/08164622.2021.1880863] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Inherited retinal diseases (IRDs) comprise a heterogeneous group of genetic disorders affecting the retina. Caused by mutations in over 300 genes, IRDs result in visual impairment due to dysfunction and degeneration of photoreceptors, retinal pigment epithelium, or the choroid. Important photoreceptor IRDs include retinitis pigmentosa and Leber congenital amaurosis. Macular dystrophies include Stargardt and Best disease. Currently, IRDs are largely incurable but the landscape of treatment options is rapidly changing for these diseases which, untreated, result in severe visual impairment and blindness.Advances in DNA delivery to the retina and improved genetic diagnosis of IRDs have led to a new era of research into gene therapy for these vision-threatening disorders. Gene therapy is a compelling approach due to the monogenic nature of most IRDs, with the retina being a favourable target for administering genetic vectors due to its immunoprivileged environment, direct visibility, and multiple methods to assess sensitivity and function. Generally, retinal gene therapy involves a subretinal or intravitreal injection of a viral vector, which infects target cells to deliver a therapeutic gene, or transgene. A gene augmentation strategy introduces a functioning copy of a gene to restore expression of a mutated gene, whereas a gene-editing strategy aims to directly edit and correct the mutation. Common delivery vectors include adeno-associated virus (AAV) and lentivirus.Voretigene neparvovec-rzyl (Luxturna) became the first FDA-approved direct gene therapy in December 2017, and the Australian TGA followed suit in August 2020. More are projected to follow, with clinical trials underway for many other IRDs.This review provides an overview of gene therapy for IRDs, including current progress and challenges. A companion article in this issue details target patient populations for IRD gene therapy, and how optometrists can assist in assessing individuals who may be eligible for current and future therapies.
Collapse
Affiliation(s)
- Monica L Hu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Thomas L Edwards
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.,Department of Surgery (Ophthalmology), Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Australia
| | - Fleur O'Hare
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.,Department of Surgery (Ophthalmology), Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Australia.,Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Australia
| | - Doron G Hickey
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Jiang-Hui Wang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Zhengyang Liu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Lauren N Ayton
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.,Department of Surgery (Ophthalmology), Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Australia.,Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Australia
| |
Collapse
|
12
|
Liu J, Ding X, Fu Y, Xiang C, Yuan Y, Zhang Y, Yu P. Cyclodextrins based delivery systems for macro biomolecules. Eur J Med Chem 2020; 212:113105. [PMID: 33385835 DOI: 10.1016/j.ejmech.2020.113105] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Macro biomolecules are of vital importance in regulating the biofunctions in organisms, in which proteins (including peptides when mentioned below) and nucleic acids (NAs) are the most important. Therefore, these proteins and NAs can be applied as "drugs" to regulate the biofunctions from abnormal to normal. Either for proteins and NAs, the most challenging thing is to avoid the biodegradation or physicochemical degradation before they reach the targeted location, and then functions as complete functional structures. Hence, appropriate delivery systems are very important which can protect them from these degradations. Cyclodextrins (CDs) based delivery systems achieved mega successes due to their outstanding pharmaceutical properties and there have been several reviews on CDs based small molecule drug delivery systems recently. But for biomolecules, which are getting more and more important for modern therapies, however, there are very few reviews to systematically summarize and analyze the CDs-based macro biomolecules delivery systems, especially for proteins. In this review, there were some of the notable examples were summarized for the macro biomolecules (proteins and NAs) delivery based on CDs. For proteins, this review included insulin, lysozyme, bovine serum albumin (BSA), green fluorescent protein (GFP) and IgG's, etc. deliveries in slow release, stimulating responsive release or targeting release manners. For NAs, this review summarized cationic CD-polymers and CD-cluster monomers as NAs carriers, notably, including the multicomponents targeting CD-based carriers and the virus-like RNA assembly method siRNA carriers.
Collapse
Affiliation(s)
- Jiang Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China.
| | - Xin Ding
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Yupeng Fu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Cen Xiang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Yuan Yuan
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China
| | - Yongmin Zhang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China; Sorbonne Université, CNRS, IPCM, UMR 8232, 4 Place Jussieu, 75005, Paris, France
| | - Peng Yu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, 300457, Tianjin, China.
| |
Collapse
|
13
|
Zhou J, Shao Z, Liu J, Duan Q, Wang X, Li J, Yang H. From Endocytosis to Nonendocytosis: The Emerging Era of Gene Delivery. ACS APPLIED BIO MATERIALS 2020; 3:2686-2701. [DOI: 10.1021/acsabm.9b01131] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jie Zhou
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, People’s Republic of China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Zhentao Shao
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Jia Liu
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Qiao Duan
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Xiang Wang
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, People’s Republic of China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People’s Republic of China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, People’s Republic of China
| |
Collapse
|
14
|
Vázquez-Domínguez I, Garanto A, Collin RWJ. Molecular Therapies for Inherited Retinal Diseases-Current Standing, Opportunities and Challenges. Genes (Basel) 2019; 10:genes10090654. [PMID: 31466352 PMCID: PMC6770110 DOI: 10.3390/genes10090654] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 12/15/2022] Open
Abstract
Inherited retinal diseases (IRDs) are both genetically and clinically highly heterogeneous and have long been considered incurable. Following the successful development of a gene augmentation therapy for biallelic RPE65-associated IRD, this view has changed. As a result, many different therapeutic approaches are currently being developed, in particular a large variety of molecular therapies. These are depending on the severity of the retinal degeneration, knowledge of the pathophysiological mechanism underlying each subtype of IRD, and the therapeutic target molecule. DNA therapies include approaches such as gene augmentation therapy, genome editing and optogenetics. For some genetic subtypes of IRD, RNA therapies and compound therapies have also shown considerable therapeutic potential. In this review, we summarize the current state-of-the-art of various therapeutic approaches, including the pros and cons of each strategy, and outline the future challenges that lie ahead in the combat against IRDs.
Collapse
Affiliation(s)
- Irene Vázquez-Domínguez
- Department of Human Genetics and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands
| | - Alejandro Garanto
- Department of Human Genetics and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands.
| | - Rob W J Collin
- Department of Human Genetics and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands.
| |
Collapse
|
15
|
Genome Editing as a Treatment for the Most Prevalent Causative Genes of Autosomal Dominant Retinitis Pigmentosa. Int J Mol Sci 2019; 20:ijms20102542. [PMID: 31126147 PMCID: PMC6567127 DOI: 10.3390/ijms20102542] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/15/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023] Open
Abstract
: Inherited retinal dystrophies (IRDs) are a clinically and genetically heterogeneous group of diseases with more than 250 causative genes. The most common form is retinitis pigmentosa. IRDs lead to vision impairment for which there is no universal cure. Encouragingly, a first gene supplementation therapy has been approved for an autosomal recessive IRD. However, for autosomal dominant IRDs, gene supplementation therapy is not always pertinent because haploinsufficiency is not the only cause. Disease-causing mechanisms are often gain-of-function or dominant-negative, which usually require alternative therapeutic approaches. In such cases, genome-editing technology has raised hopes for treatment. Genome editing could be used to i) invalidate both alleles, followed by supplementation of the wild type gene, ii) specifically invalidate the mutant allele, with or without gene supplementation, or iii) to correct the mutant allele. We review here the most prevalent genes causing autosomal dominant retinitis pigmentosa and the most appropriate genome-editing strategy that could be used to target their different causative mutations.
Collapse
|
16
|
Guiding Lights in Genome Editing for Inherited Retinal Disorders: Implications for Gene and Cell Therapy. Neural Plast 2018; 2018:5056279. [PMID: 29853845 PMCID: PMC5964415 DOI: 10.1155/2018/5056279] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/18/2018] [Indexed: 12/26/2022] Open
Abstract
Inherited retinal dystrophies (IRDs) are a leading cause of visual impairment in the developing world. These conditions present an irreversible dysfunction or loss of neural retinal cells, which significantly impacts quality of life. Due to the anatomical accessibility and immunoprivileged status of the eye, ophthalmological research has been at the forefront of innovative and advanced gene- and cell-based therapies, both of which represent great potential as therapeutic treatments for IRD patients. However, due to a genetic and clinical heterogeneity, certain IRDs are not candidates for these approaches. New advances in the field of genome editing using Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and CRISPR-associated protein (Cas) have provided an accurate and efficient way to edit the human genome and represent an appealing alternative for treating IRDs. We provide a brief update on current gene augmentation therapies for retinal dystrophies. Furthermore, we discuss recent advances in the field of genome editing and stem cell technologies, which together enable precise and personalized therapies for patients. Lastly, we highlight current technological limitations and barriers that need to be overcome before this technology can become a viable treatment option for patients.
Collapse
|
17
|
LaVail MM, Nishikawa S, Steinberg RH, Naash MI, Duncan JL, Trautmann N, Matthes MT, Yasumura D, Lau-Villacorta C, Chen J, Peterson WM, Yang H, Flannery JG. Phenotypic characterization of P23H and S334ter rhodopsin transgenic rat models of inherited retinal degeneration. Exp Eye Res 2018; 167:56-90. [PMID: 29122605 PMCID: PMC5811379 DOI: 10.1016/j.exer.2017.10.023] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/25/2017] [Accepted: 10/31/2017] [Indexed: 02/07/2023]
Abstract
We produced 8 lines of transgenic (Tg) rats expressing one of two different rhodopsin mutations in albino Sprague-Dawley (SD) rats. Three lines were generated with a proline to histidine substitution at codon 23 (P23H), the most common autosomal dominant form of retinitis pigmentosa in the United States. Five lines were generated with a termination codon at position 334 (S334ter), resulting in a C-terminal truncated opsin protein lacking the last 15 amino acid residues and containing all of the phosphorylation sites involved in rhodopsin deactivation, as well as the terminal QVAPA residues important for rhodopsin deactivation and trafficking. The rates of photoreceptor (PR) degeneration in these models vary in proportion to the ratio of mutant to wild-type rhodopsin. The models have been widely studied, but many aspects of their phenotypes have not been described. Here we present a comprehensive study of the 8 Tg lines, including the time course of PR degeneration from the onset to one year of age, retinal structure by light and electron microscopy (EM), hemispheric asymmetry and gradients of rod and cone degeneration, rhodopsin content, gene dosage effect, rapid activation and invasion of the outer retina by presumptive microglia, rod outer segment disc shedding and phagocytosis by the retinal pigmented epithelium (RPE), and retinal function by the electroretinogram (ERG). The biphasic nature of PR cell death was noted, as was the lack of an injury-induced protective response in the rat models. EM analysis revealed the accumulation of submicron vesicular structures in the interphotoreceptor space during the peak period of PR outer segment degeneration in the S334ter lines. This is likely due to the elimination of the trafficking consensus domain as seen before as with other rhodopsin mutants lacking the C-terminal QVAPA. The 8 rhodopsin Tg lines have been, and will continue to be, extremely useful models for the experimental study of inherited retinal degenerations.
Collapse
Affiliation(s)
- Matthew M LaVail
- Beckman Vision Center, University of California, San Francisco, San Francisco, CA 94143-0730, USA.
| | - Shimpei Nishikawa
- Beckman Vision Center, University of California, San Francisco, San Francisco, CA 94143-0730, USA.
| | - Roy H Steinberg
- Beckman Vision Center, University of California, San Francisco, San Francisco, CA 94143-0730, USA
| | - Muna I Naash
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd., Room 2011, Houston, TX 77204-5060, USA.
| | - Jacque L Duncan
- Beckman Vision Center, University of California, San Francisco, San Francisco, CA 94143-0730, USA.
| | - Nikolaus Trautmann
- Beckman Vision Center, University of California, San Francisco, San Francisco, CA 94143-0730, USA.
| | - Michael T Matthes
- Beckman Vision Center, University of California, San Francisco, San Francisco, CA 94143-0730, USA.
| | - Douglas Yasumura
- Beckman Vision Center, University of California, San Francisco, San Francisco, CA 94143-0730, USA
| | - Cathy Lau-Villacorta
- Beckman Vision Center, University of California, San Francisco, San Francisco, CA 94143-0730, USA.
| | - Jeannie Chen
- Zilka Neurogenetic Institute, USC Keck School of Medicine, Los Angeles, CA 90089-2821, USA.
| | - Ward M Peterson
- Beckman Vision Center, University of California, San Francisco, San Francisco, CA 94143-0730, USA.
| | - Haidong Yang
- Beckman Vision Center, University of California, San Francisco, San Francisco, CA 94143-0730, USA.
| | - John G Flannery
- School of Optometry, UC Berkeley, Berkeley, CA 94720-2020, USA.
| |
Collapse
|
18
|
Moore NA, Morral N, Ciulla TA, Bracha P. Gene therapy for inherited retinal and optic nerve degenerations. Expert Opin Biol Ther 2017; 18:37-49. [DOI: 10.1080/14712598.2018.1389886] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Nicholas A. Moore
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nuria Morral
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Thomas A. Ciulla
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
- Retina Service, Midwest Eye Institute, Indianapolis, IN, USA
| | - Peter Bracha
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
19
|
Yu-Wai-Man P. Genetic manipulation for inherited neurodegenerative diseases: myth or reality? Br J Ophthalmol 2016; 100:1322-31. [PMID: 27002113 PMCID: PMC5050284 DOI: 10.1136/bjophthalmol-2015-308329] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/28/2016] [Indexed: 12/22/2022]
Abstract
Rare genetic diseases affect about 7% of the general population and over 7000 distinct clinical syndromes have been described with the majority being due to single gene defects. This review will provide a critical overview of genetic strategies that are being pioneered to halt or reverse disease progression in inherited neurodegenerative diseases. This field of research covers a vast area and only the most promising treatment paradigms will be discussed with a particular focus on inherited eye diseases, which have paved the way for innovative gene therapy paradigms, and mitochondrial diseases, which are currently generating a lot of debate centred on the bioethics of germline manipulation.
Collapse
Affiliation(s)
- Patrick Yu-Wai-Man
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK Newcastle Eye Centre, Royal Victoria Infirmary, Newcastle upon Tyne, UK NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, UK
| |
Collapse
|
20
|
Wong LL, Pye QN, Chen L, Seal S, McGinnis JF. Defining the catalytic activity of nanoceria in the P23H-1 rat, a photoreceptor degeneration model. PLoS One 2015; 10:e0121977. [PMID: 25822196 PMCID: PMC4379093 DOI: 10.1371/journal.pone.0121977] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/09/2015] [Indexed: 12/17/2022] Open
Abstract
Purpose Inorganic catalytic nanoceria or cerium oxide nanoparticles (CeNPs) are bona fide antioxidants that possess regenerative radical scavenging activities in vitro. Previously, we demonstrated that CeNPs had neuroprotective and anti-angiogenic properties in rodent retinal degeneration and neovascularization models. However, the cellular mechanisms and duration of the catalytic activity of CeNPs in preventing photoreceptor cell loss are still unknown. In this study, we sought to answer these questions using the P23H-1 rat, an autosomal dominant retinitis pigmentosa (adRP) model. Methods A single dose of either saline or CeNPs was delivered intravitreally into the eyes of P23H-1 rats at 2–3 weeks of age. Retinal functions were examined at 3 to 7 weeks post injection. We quantified retinal proteins by Western blot analyses and counted the number of apoptotic (TUNEL+) profiles in the outer nuclear layer (ONL) of retinal sections. We measured free 8-isoprostanes to quantify lipid peroxidation in retinal tissues. Results We observed increased rod and cone cell functions up to three weeks post injection. Apoptotic cells were reduced by 46%, 56%, 21%, and 24% at 3, 7, 14, 21 days, respectively, after CeNPs injection compared to saline. Additionally, reduction of lipid peroxidation in the retinas of CeNPs-treated vs saline-treated animals was detected 14 days post injection. Conclusions We validated that CeNPs were effective in delaying loss of photoreceptor cell function in an adRP rat model. This represents the fourth rodent retinal disease model that shows delay in disease progression after a single application of CeNPs. We further demonstrated that CeNPs slowed the rate of photoreceptor cell death. We deduced that the catalytic activity of CeNPs in vivo in this rat model to be undiminished for at least 7 days and then declined over the next 14 days after CeNPs administration.
Collapse
Affiliation(s)
- Lily L. Wong
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, College of Medicine, and Dean McGee Eye Institute, Oklahoma City, Oklahoma, United States of America
- * E-mail: (LLW); (JFM)
| | - Quentin N. Pye
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, College of Medicine, and Dean McGee Eye Institute, Oklahoma City, Oklahoma, United States of America
| | - Lijuan Chen
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, College of Medicine, and Dean McGee Eye Institute, Oklahoma City, Oklahoma, United States of America
| | - Sudipta Seal
- Advanced Materials Processing Analysis Center, Materials Science and Engineering, Nanoscience and Technology Center, College of Medicine, University of Central Florida, Orlando, Florida, United States of America
| | - James F. McGinnis
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, College of Medicine, and Dean McGee Eye Institute, Oklahoma City, Oklahoma, United States of America
- Department of Cell Biology and Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Graduate College, Oklahoma City, Oklahoma, United States of America
- * E-mail: (LLW); (JFM)
| |
Collapse
|
21
|
Fernandez-San Jose P, Blanco-Kelly F, Corton M, Trujillo-Tiebas MJ, Gimenez A, Avila-Fernandez A, Garcia-Sandoval B, Lopez-Molina MI, Hernan I, Carballo M, Riveiro-Alvarez R, Ayuso C. Prevalence of Rhodopsin mutations in autosomal dominant Retinitis Pigmentosa in Spain: clinical and analytical review in 200 families. Acta Ophthalmol 2015; 93:e38-44. [PMID: 25408095 DOI: 10.1111/aos.12486] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 05/24/2014] [Indexed: 01/31/2023]
Abstract
PURPOSE We aimed to determine the prevalence of mutations in the RHO gene in Spanish families with autosomal dominant Retinitis Pigmentosa (adRP), to assess genotype-phenotype correlations and to establish an accurate diagnostic algorithm after 23 years of data collection. PATIENTS AND METHODS Two hundred patients were analysed through a combination of denaturing gradient gel electrophoresis, single-strand conformation polymorphism, genotyping microarray and Sanger sequencing of the RHO gene. RESULTS Overall, 42 of 200 Spanish adRP families were mutated for RHO (21.0%). Twenty-seven different RHO mutations were detected; seven of them were novel. A genotype-phenotype correlation was established with clinical data from 107 patients. The most prevalent p.Pro347Leu mutation, responsible for 4.5% (9/200) of all mutated adRP families, was associated with a phenotype of early onset and severe course diffuse RP. CONCLUSIONS This retrospective study provides a wide spectrum of mutations in the RHO gene in Spanish patients with adRP. Also, the prevalence of mutations is similar to that reported in European population. Genotyping microarray followed by RHO sequencing is proposed as a first step in molecular diagnosis of adRP Spanish families. An increasing understanding of causal RHO alleles in adRP facilitates disease diagnosis and prognosis, especially for the prevalent p.Pro347Leu mutation.
Collapse
Affiliation(s)
- Patricia Fernandez-San Jose
- Department of Genetics; Health Research Institute Fundacion Jimenez Diaz; University Hospital (IIS-FJD, UAM); Madrid Spain
- Centre for Biomedical Network Research on Rare Diseases CIBERER; ISCIII; Madrid Spain
| | - Fiona Blanco-Kelly
- Department of Genetics; Health Research Institute Fundacion Jimenez Diaz; University Hospital (IIS-FJD, UAM); Madrid Spain
- Centre for Biomedical Network Research on Rare Diseases CIBERER; ISCIII; Madrid Spain
| | - Marta Corton
- Department of Genetics; Health Research Institute Fundacion Jimenez Diaz; University Hospital (IIS-FJD, UAM); Madrid Spain
- Centre for Biomedical Network Research on Rare Diseases CIBERER; ISCIII; Madrid Spain
| | - Maria-Jose Trujillo-Tiebas
- Department of Genetics; Health Research Institute Fundacion Jimenez Diaz; University Hospital (IIS-FJD, UAM); Madrid Spain
- Centre for Biomedical Network Research on Rare Diseases CIBERER; ISCIII; Madrid Spain
| | - Ascension Gimenez
- Department of Genetics; Health Research Institute Fundacion Jimenez Diaz; University Hospital (IIS-FJD, UAM); Madrid Spain
- Centre for Biomedical Network Research on Rare Diseases CIBERER; ISCIII; Madrid Spain
| | - Almudena Avila-Fernandez
- Department of Genetics; Health Research Institute Fundacion Jimenez Diaz; University Hospital (IIS-FJD, UAM); Madrid Spain
- Centre for Biomedical Network Research on Rare Diseases CIBERER; ISCIII; Madrid Spain
| | - Blanca Garcia-Sandoval
- Centre for Biomedical Network Research on Rare Diseases CIBERER; ISCIII; Madrid Spain
- Department of Ophthalmology; Health Research Institute Fundacion Jimenez Diaz; University Hospital (IIS-FJD, UAM); Madrid Spain
| | - Maria-Isabel Lopez-Molina
- Centre for Biomedical Network Research on Rare Diseases CIBERER; ISCIII; Madrid Spain
- Department of Ophthalmology; Health Research Institute Fundacion Jimenez Diaz; University Hospital (IIS-FJD, UAM); Madrid Spain
| | - Inma Hernan
- Molecular Genetics Unit; Hospital de Terrassa; Terrassa Barcelona Spain
| | - Miguel Carballo
- Molecular Genetics Unit; Hospital de Terrassa; Terrassa Barcelona Spain
| | - Rosa Riveiro-Alvarez
- Department of Genetics; Health Research Institute Fundacion Jimenez Diaz; University Hospital (IIS-FJD, UAM); Madrid Spain
- Centre for Biomedical Network Research on Rare Diseases CIBERER; ISCIII; Madrid Spain
| | - Carmen Ayuso
- Department of Genetics; Health Research Institute Fundacion Jimenez Diaz; University Hospital (IIS-FJD, UAM); Madrid Spain
- Centre for Biomedical Network Research on Rare Diseases CIBERER; ISCIII; Madrid Spain
| |
Collapse
|
22
|
Abstract
Adaptive optics is a relatively new tool that is available to ophthalmologists for study of cellular level details. In addition to the axial resolution provided by the spectral-domain optical coherence tomography, adaptive optics provides an excellent lateral resolution, enabling visualization of the photoreceptors, blood vessels and details of the optic nerve head. We attempt a mini review of the current role of adaptive optics in retinal imaging. PubMed search was performed with key words Adaptive optics OR Retina OR Retinal imaging. Conference abstracts were searched from the Association for Research in Vision and Ophthalmology (ARVO) and American Academy of Ophthalmology (AAO) meetings. In total, 261 relevant publications and 389 conference abstracts were identified.
Collapse
Affiliation(s)
- Rajani Battu
- Narayana Nethralaya, Bangalore, Karnataka, India
| | | | | | | | | |
Collapse
|
23
|
Abstract
Significant advances have been made over the last decade or two in the elucidation of the molecular pathogenesis of inherited ocular disorders. In particular, remarkable successes have been achieved in exploration of gene-based medicines for these conditions, both in preclinical and in clinical studies. Progress in the development of gene therapies targeted toward correcting the primary genetic defect or focused on modulating secondary effects associated with retinal pathologies are discussed in the review. Likewise, the recent utilization of genes encoding light-sensing molecules to provide new functions to residual retinal cells in the degenerating retina is discussed. While a great deal has been learned over the last two decades, the next decade should result in an increasing number of preclinical studies progressing to human clinical trial, an exciting prospect for patients, those active in research and development and bystanders alike.
Collapse
|
24
|
Jastrzebska B. GPCR: G protein complexes--the fundamental signaling assembly. Amino Acids 2013; 45:1303-14. [PMID: 24052187 DOI: 10.1007/s00726-013-1593-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 09/10/2013] [Indexed: 12/15/2022]
Abstract
G protein coupled receptors (GPCR) constitute the largest group of cell surface receptors that transmit various signals across biological membranes through the binding and activation of heterotrimeric G proteins, which amplify the signal and activate downstream effectors leading to the biological responses. Thus, the first critical step in this signaling cascade is the interaction between receptor and its cognate G protein. Understanding this critical event at the molecular level is of high importance because abnormal function of GPCRs is associated with many diseases. Thus, these receptors are targets for drug development.
Collapse
Affiliation(s)
- Beata Jastrzebska
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106-4965, USA,
| |
Collapse
|
25
|
Carroll J, Kay DB, Scoles D, Dubra A, Lombardo M. Adaptive optics retinal imaging--clinical opportunities and challenges. Curr Eye Res 2013; 38:709-21. [PMID: 23621343 PMCID: PMC4031042 DOI: 10.3109/02713683.2013.784792] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The array of therapeutic options available to clinicians for treating retinal disease is expanding. With these advances comes the need for better understanding of the etiology of these diseases on a cellular level as well as improved non-invasive tools for identifying the best candidates for given therapies and monitoring the efficacy of those therapies. While spectral domain optical coherence tomography offers a widely available tool for clinicians to assay the living retina, it suffers from poor lateral resolution due to the eye's monochromatic aberrations. Ophthalmic adaptive optics (AO) is a technique to compensate for the eye's aberrations and provide nearly diffraction-limited resolution. The result is the ability to visualize the living retina with cellular resolution. While AO is unquestionably a powerful research tool, many clinicians remain undecided on the clinical potential of AO imaging - putting many at a crossroads with respect to adoption of this technology. This review will briefly summarize the current state of AO retinal imaging, discuss current as well as future clinical applications of AO retinal imaging, and finally provide some discussion of research needs to facilitate more widespread clinical use.
Collapse
Affiliation(s)
- Joseph Carroll
- Department of Ophthalmology, Medical College of Wisconsin, Milwaukee, WI, USA.
| | | | | | | | | |
Collapse
|
26
|
Abstract
Sight-restoring therapy for the visually impaired and blind is a major unmet medical need. Ocular gene therapy is a rational choice for restoring vision or preventing the loss of vision because most blinding diseases originate in cellular components of the eye, a compartment that is optimally suited for the delivery of genes, and many of these diseases have a genetic origin or genetic component. In recent years we have witnessed major advances in the field of ocular gene therapy, and proof-of-concept studies are under way to evaluate the safety and efficacy of human gene therapies. Here we discuss the concepts and recent advances in gene therapy in the retina. Our review discusses traditional approaches such as gene replacement and neuroprotection and also new avenues such as optogenetic therapies. We conjecture that advances in gene therapy in the retina will pave the way for gene therapies in other parts of the brain.
Collapse
Affiliation(s)
- José-Alain Sahel
- INSERM UMR_S 968, UPMC, University of Paris 06, Institut de la Vision, Paris, France.
| | | |
Collapse
|
27
|
Simonato M, Bennett J, Boulis NM, Castro MG, Fink DJ, Goins WF, Gray SJ, Lowenstein PR, Vandenberghe LH, Wilson TJ, Wolfe JH, Glorioso JC. Progress in gene therapy for neurological disorders. Nat Rev Neurol 2013; 9:277-91. [PMID: 23609618 PMCID: PMC3908892 DOI: 10.1038/nrneurol.2013.56] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diseases of the nervous system have devastating effects and are widely distributed among the population, being especially prevalent in the elderly. These diseases are often caused by inherited genetic mutations that result in abnormal nervous system development, neurodegeneration, or impaired neuronal function. Other causes of neurological diseases include genetic and epigenetic changes induced by environmental insults, injury, disease-related events or inflammatory processes. Standard medical and surgical practice has not proved effective in curing or treating these diseases, and appropriate pharmaceuticals do not exist or are insufficient to slow disease progression. Gene therapy is emerging as a powerful approach with potential to treat and even cure some of the most common diseases of the nervous system. Gene therapy for neurological diseases has been made possible through progress in understanding the underlying disease mechanisms, particularly those involving sensory neurons, and also by improvement of gene vector design, therapeutic gene selection, and methods of delivery. Progress in the field has renewed our optimism for gene therapy as a treatment modality that can be used by neurologists, ophthalmologists and neurosurgeons. In this Review, we describe the promising gene therapy strategies that have the potential to treat patients with neurological diseases and discuss prospects for future development of gene therapy.
Collapse
Affiliation(s)
- Michele Simonato
- Section of Pharmacology, Department of Medical Sciences, University of Ferrara, Fossato di Mortara 17-19, 44121 Ferrara, Italy. michele.simonato@ unife.it
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
|
29
|
Abstract
Clinical trials are currently underway using gene therapy to treat retinal disease such as Leber congenital amaurosis (LCA). Viral vectors that have been utilized to target retinal cells include adenoviruses, lentiviruses, and recombinant adeno-associated viruses (rAAV). Of the three classes, rAAV vectors show the greatest promise for retinal gene therapy. Recent developments in virus technology such as the development of hybrid and capsid mutant rAAV vectors mean that specific retinal cells can be targeted and faster stronger transgene expression is now possible compared to that achieved with the first generation of vectors. Gene therapy trials in dogs have been very important in the development of therapy for RPE65 LCA which is currently in phase I/II clinical trials in humans. Recent successes in using gene therapy to treat canine achromatopsia, X-linked progressive retinal atrophy (PRA) and the more severe rapid degenerations such as rod-cone dysplasia type 3 may lead also to the translation to human clinical trials. Dogs have played and continue to play an important role as animal models for proof-of-concept studies of retinal gene therapy. As modifications and improvements in gene therapy protocols are made from experience gathered from human clinical trials perhaps gene therapy for the treatment of canine clinical patients will become available to veterinary ophthalmologists.
Collapse
Affiliation(s)
- Simon M Petersen-Jones
- Veterinary Medical Center, Michigan State University, 736 Wilson Road, D-208, East Lansing, MI 48824, USA.
| |
Collapse
|
30
|
Makino CL, Wen XH, Michaud NA, Covington HI, DiBenedetto E, Hamm HE, Lem J, Caruso G. Rhodopsin expression level affects rod outer segment morphology and photoresponse kinetics. PLoS One 2012; 7:e37832. [PMID: 22662234 PMCID: PMC3360601 DOI: 10.1371/journal.pone.0037832] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 04/26/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The retinal rod outer segment is a sensory cilium that is specialized for the conversion of light into an electrical signal. Within the cilium, up to several thousand membranous disks contain as many as a billion copies of rhodopsin for efficient photon capture. Disks are continually turned over, requiring the daily synthesis of a prodigious amount of rhodopsin. To promote axial diffusion in the aqueous cytoplasm, the disks have one or more incisures. Across vertebrates, the range of disk diameters spans an order of magnitude, and the number and length of the incisures vary considerably, but the mechanisms controlling disk architecture are not well understood. The finding that transgenic mice overexpressing rhodopsin have enlarged disks lacking an incisure prompted us to test whether lowered rhodopsin levels constrain disk assembly. METHODOLOGY/PRINCIPAL FINDINGS The structure and function of rods from hemizygous rhodopsin knockout (R+/-) mice with decreased rhodopsin expression were analyzed by transmission electron microscopy and single cell recording. R+/- rods were structurally altered in three ways: disk shape changed from circular to elliptical, disk surface area decreased, and the single incisure lengthened to divide the disk into two sections. Photocurrent responses to flashes recovered more rapidly than normal. A spatially resolved model of phototransduction indicated that changes in the packing densities of rhodopsin and other transduction proteins were responsible. The decrease in aqueous outer segment volume and the lengthened incisure had only minor effects on photon response amplitude and kinetics. CONCLUSIONS/SIGNIFICANCE Rhodopsin availability limits disk assembly and outer segment girth in normal rods. The incisure may buffer the supply of structural proteins needed to form larger disks. Decreased rhodopsin level accelerated photoresponse kinetics by increasing the rates of molecular collisions on the membrane. Faster responses, together with fewer rhodopsins, combine to lower overall sensitivity of R+/- rods to light.
Collapse
Affiliation(s)
- Clint L Makino
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|