1
|
Liu D, Li T, Liu L, Che X, Li X, Liu C, Wu G. Adeno-associated virus therapies: Pioneering solutions for human genetic diseases. Cytokine Growth Factor Rev 2024:S1359-6101(24)00078-9. [PMID: 39322487 DOI: 10.1016/j.cytogfr.2024.09.003] [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: 09/03/2024] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024]
Abstract
Adeno-associated virus (AAV) has emerged as a fundamental component in the gene therapy landscape, widely acknowledged for its effectiveness in therapeutic gene delivery. The success of AAV-based therapies, such as Luxturna and Zolgensma, underscores their potential as a leading vector in gene therapy. This article provides an in-depth review of the development and mechanisms of AAV vector-based therapies, offering a comprehensive analysis of the latest clinical trial outcomes in central nervous system (CNS) diseases, ocular conditions, and hemophilia, where AAV therapies have shown promising results. Additionally, we discusse the selection of administration methods and serotypes tailored to specific diseases. Our objective is to showcase the innovative applications and future potential of AAV-based gene therapy, laying the groundwork for continued clinical advancements.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Lei Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaorui Li
- Department of oncology, Cancer Hospital of Dalian University of Technology, Shenyang 110042, China.
| | - Chang Liu
- Department of thoracic surgery, Shenyang Tenth People's Hospital, Shenyang 110042, China.
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
2
|
Castro BFM, Steel JC, Layton CJ. AAV-Based Strategies for Treatment of Retinal and Choroidal Vascular Diseases: Advances in Age-Related Macular Degeneration and Diabetic Retinopathy Therapies. BioDrugs 2024; 38:73-93. [PMID: 37878215 PMCID: PMC10789843 DOI: 10.1007/s40259-023-00629-y] [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] [Accepted: 09/20/2023] [Indexed: 10/26/2023]
Abstract
Age-related macular degeneration (AMD) and diabetic retinopathy (DR) are vascular diseases with high prevalence, ranking among the leading causes of blindness and vision loss worldwide. Despite being effective, current treatments for AMD and DR are burdensome for patients and clinicians, resulting in suboptimal compliance and real risk of vision loss. Thus, there is an unmet need for long-lasting alternatives with improved safety and efficacy. Adeno-associated virus (AAV) is the leading vector for ocular gene delivery, given its ability to enable long-term expression while eliciting relatively mild immune responses. Progress has been made in AAV-based gene therapies for not only inherited retinal diseases but also acquired conditions with preclinical and clinical studies of AMD and DR showing promising results. These studies have explored several pathways involved in the disease pathogenesis, as well as different strategies to optimise gene delivery. These include engineered capsids with enhanced tropism to particular cell types, and expression cassettes incorporating elements for a targeted and controlled expression. Multiple-acting constructs have also been investigated, in addition to gene silencing and editing. Here, we provide an overview of strategies employing AAV-mediated gene delivery to treat AMD and DR. We discuss preclinical efficacy studies and present the latest data from clinical trials for both diseases.
Collapse
Affiliation(s)
- Brenda F M Castro
- LVF Ophthalmology Research Centre, Translational Research Institute, Brisbane, QLD, 4102, Australia.
- Greenslopes Clinical School, University of Queensland School of Medicine, Brisbane, QLD, Australia.
| | - Jason C Steel
- LVF Ophthalmology Research Centre, Translational Research Institute, Brisbane, QLD, 4102, Australia
- Greenslopes Clinical School, University of Queensland School of Medicine, Brisbane, QLD, Australia
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD, Australia
| | - Christopher J Layton
- LVF Ophthalmology Research Centre, Translational Research Institute, Brisbane, QLD, 4102, Australia.
- Greenslopes Clinical School, University of Queensland School of Medicine, Brisbane, QLD, Australia.
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD, Australia.
| |
Collapse
|
3
|
Diabetic Macular Edema: Current Understanding, Molecular Mechanisms and Therapeutic Implications. Cells 2022; 11:cells11213362. [PMID: 36359761 PMCID: PMC9655436 DOI: 10.3390/cells11213362] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 11/24/2022] Open
Abstract
Diabetic retinopathy (DR), with increasing incidence, is the major cause of vision loss and blindness worldwide in working-age adults. Diabetic macular edema (DME) remains the main cause of vision impairment in diabetic patients, with its pathogenesis still not completely elucidated. Vascular endothelial growth factor (VEGF) plays a pivotal role in the pathogenesis of DR and DME. Currently, intravitreal injection of anti-VEGF agents remains as the first-line therapy in DME treatment due to the superior anatomic and functional outcomes. However, some patients do not respond satisfactorily to anti-VEGF injections. More than 30% patients still exist with persistent DME even after regular intravitreal injection for at least 4 injections within 24 weeks, suggesting other pathogenic factors, beyond VEGF, might contribute to the pathogenesis of DME. Recent advances showed nearly all the retinal cells are involved in DR and DME, including breakdown of blood-retinal barrier (BRB), drainage dysfunction of Müller glia and retinal pigment epithelium (RPE), involvement of inflammation, oxidative stress, and neurodegeneration, all complicating the pathogenesis of DME. The profound understanding of the changes in proteomics and metabolomics helps improve the elucidation of the pathogenesis of DR and DME and leads to the identification of novel targets, biomarkers and potential therapeutic strategies for DME treatment. The present review aimed to summarize the current understanding of DME, the involved molecular mechanisms, and the changes in proteomics and metabolomics, thus to propose the potential therapeutic recommendations for personalized treatment of DME.
Collapse
|
4
|
Sarkar A, Junnuthula V, Dyawanapelly S. Ocular Therapeutics and Molecular Delivery Strategies for Neovascular Age-Related Macular Degeneration (nAMD). Int J Mol Sci 2021; 22:10594. [PMID: 34638935 PMCID: PMC8508687 DOI: 10.3390/ijms221910594] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/26/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of vision loss in geriatric population. Intravitreal (IVT) injections are popular clinical option. Biologics and small molecules offer efficacy but relatively shorter half-life after intravitreal injections. To address these challenges, numerous technologies and therapies are under development. Most of these strategies aim to reduce the frequency of injections, thereby increasing patient compliance and reducing patient-associated burden. Unlike IVT frequent injections, molecular therapies such as cell therapy and gene therapy offer restoration ability hence gained a lot of traction. The recent approval of ocular gene therapy for inherited disease offers new hope in this direction. However, until such breakthrough therapies are available to the majority of patients, antibody therapeutics will be on the shelf, continuing to provide therapeutic benefits. The present review aims to highlight the status of pre-clinical and clinical studies of neovascular AMD treatment modalities including Anti-VEGF therapy, upcoming bispecific antibodies, small molecules, port delivery systems, photodynamic therapy, radiation therapy, gene therapy, cell therapy, and combination therapies.
Collapse
Affiliation(s)
- Aira Sarkar
- Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA;
| | | | - Sathish Dyawanapelly
- Department of Pharmaceutical Sciences & Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Mumbai 400019, India
| |
Collapse
|
5
|
Chung SH, Frick SL, Yiu G. Targeting vascular endothelial growth factor using retinal gene therapy. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1277. [PMID: 34532414 PMCID: PMC8421957 DOI: 10.21037/atm-20-4417] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022]
Abstract
Pharmacotherapies targeting vascular endothelial growth factor (VEGF) have revolutionized the management for neovascular retinal disorders including diabetic retinopathy and neovascular age-related macular degeneration. However, the burden of frequent injections, high cost, and treatment resistance in some patients remain unresolved. To overcome these challenges, newer generations of anti-angiogenic biological therapies, engineered proteins, implantable delivery systems, and biopolymers are currently being developed to enable more sustained, longer-lasting treatments. The use of gene therapies for pathologic angiogenesis has garnered renewed interests since the first FDA-approval of a gene therapy to treat inherited retinal diseases associated with biallelic RPE65 mutations. Newer generations of viral vectors and novel methods of intraocular injections helped overcome ocular barriers, improving the efficiency of transduction as well as safety profile. In addition, unlike current anti-VEGF gene therapy strategies which employ a biofactory approach to mimic existing pharmacotherapies, novel genome editing strategies that target pro-angiogenic factors at the DNA level offer a unique and distinct mechanistic approach that can potentially be more precise and lead to a permanent cure. Here, we review current anti-VEGF therapies and newer pharmacologic agents under development, examine technologies and progress in adapting anti-VEGF gene therapies, and explore the future application of CRISPR-Cas9 technology to suppress ocular angiogenesis.
Collapse
Affiliation(s)
- Sook H Chung
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, CA, USA
| | - Sonia L Frick
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, CA, USA
| | - Glenn Yiu
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
6
|
Ruan Y, Jiang S, Gericke A. Age-Related Macular Degeneration: Role of Oxidative Stress and Blood Vessels. Int J Mol Sci 2021; 22:ijms22031296. [PMID: 33525498 PMCID: PMC7866075 DOI: 10.3390/ijms22031296] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Age-related macular degeneration (AMD) is a common irreversible ocular disease characterized by vision impairment among older people. Many risk factors are related to AMD and interact with each other in its pathogenesis. Notably, oxidative stress and choroidal vascular dysfunction were suggested to be critically involved in AMD pathogenesis. In this review, we give an overview on the factors contributing to the pathophysiology of this multifactorial disease and discuss the role of reactive oxygen species and vascular function in more detail. Moreover, we give an overview on therapeutic strategies for patients suffering from AMD.
Collapse
Affiliation(s)
- Yue Ruan
- Correspondence: (Y.R.); (A.G.); Tel.: +49-6131-178-276 (Y.R. & A.G.)
| | | | - Adrian Gericke
- Correspondence: (Y.R.); (A.G.); Tel.: +49-6131-178-276 (Y.R. & A.G.)
| |
Collapse
|
7
|
Chung SH, Sin TN, Ngo T, Yiu G. CRISPR Technology for Ocular Angiogenesis. Front Genome Ed 2020; 2:594984. [PMID: 34713223 PMCID: PMC8525361 DOI: 10.3389/fgeed.2020.594984] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/01/2020] [Indexed: 12/24/2022] Open
Abstract
Among genome engineering tools, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-based approaches have been widely adopted for translational studies due to their robustness, precision, and ease of use. When delivered to diseased tissues with a viral vector such as adeno-associated virus, direct genome editing can be efficiently achieved in vivo to treat different ophthalmic conditions. While CRISPR has been actively explored as a strategy for treating inherited retinal diseases, with the first human trial recently initiated, its applications for complex, multifactorial conditions such as ocular angiogenesis has been relatively limited. Currently, neovascular retinal diseases such as retinopathy of prematurity, proliferative diabetic retinopathy, and neovascular age-related macular degeneration, which together constitute the majority of blindness in developed countries, are managed with frequent and costly injections of anti-vascular endothelial growth factor (anti-VEGF) agents that are short-lived and burdensome for patients. By contrast, CRISPR technology has the potential to suppress angiogenesis permanently, with the added benefit of targeting intracellular signals or regulatory elements, cell-specific delivery, and multiplexing to disrupt different pro-angiogenic factors simultaneously. However, the prospect of permanently suppressing physiologic pathways, the unpredictability of gene editing efficacy, and concerns for off-target effects have limited enthusiasm for these approaches. Here, we review the evolution of gene therapy and advances in adapting CRISPR platforms to suppress retinal angiogenesis. We discuss different Cas9 orthologs, delivery strategies, and different genomic targets including VEGF, VEGF receptor, and HIF-1α, as well as the advantages and disadvantages of genome editing vs. conventional gene therapies for multifactorial disease processes as compared to inherited monogenic retinal disorders. Lastly, we describe barriers that must be overcome to enable effective adoption of CRISPR-based strategies for the management of ocular angiogenesis.
Collapse
Affiliation(s)
| | | | | | - Glenn Yiu
- Department of Ophthalmology and Vision Science, University of California, Davis, Sacramento, CA, United States
| |
Collapse
|
8
|
Lin FL, Wang PY, Chuang YF, Wang JH, Wong VHY, Bui BV, Liu GS. Gene Therapy Intervention in Neovascular Eye Disease: A Recent Update. Mol Ther 2020; 28:2120-2138. [PMID: 32649860 PMCID: PMC7544979 DOI: 10.1016/j.ymthe.2020.06.029] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/15/2020] [Accepted: 06/26/2020] [Indexed: 12/13/2022] Open
Abstract
Aberrant growth of blood vessels (neovascularization) is a key feature of severe eye diseases that can cause legal blindness, including neovascular age-related macular degeneration (nAMD) and diabetic retinopathy (DR). The development of anti-vascular endothelial growth factor (VEGF) agents has revolutionized the treatment of ocular neovascularization. Novel proangiogenic targets, such as angiopoietin and platelet-derived growth factor (PDGF), are under development for patients who respond poorly to anti-VEGF therapy and to reduce adverse effects from long-term VEGF inhibition. A rapidly advancing area is gene therapy, which may provide significant therapeutic benefits. Viral vector-mediated transgene delivery provides the potential for continuous production of antiangiogenic proteins, which would avoid the need for repeated anti-VEGF injections. Gene silencing with RNA interference to target ocular angiogenesis has been investigated in clinical trials. Proof-of-concept gene therapy studies using gene-editing tools such as CRISPR-Cas have already been shown to be effective in suppressing neovascularization in animal models, highlighting the therapeutic potential of the system for treatment of aberrant ocular angiogenesis. This review provides updates on the development of anti-VEGF agents and novel antiangiogenic targets. We also summarize current gene therapy strategies already in clinical trials and those with the latest approaches utilizing CRISPR-Cas gene editing against aberrant ocular neovascularization.
Collapse
Affiliation(s)
- Fan-Li Lin
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Peng-Yuan Wang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, VIC 3122, Australia.
| | - Yu-Fan Chuang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Jiang-Hui Wang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Vickie H Y Wong
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC 3010, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC 3010, Australia
| | - Guei-Sheung Liu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia; Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC 3002, Australia.
| |
Collapse
|
9
|
Xiao M, Liu Y, Wang L, Liang J, Wang T, Zhai Y, Wang Y, Liu S, Liu W, Luo X, Wang F, Sun X. Intraocular VEGF deprivation induces degeneration and fibrogenic response in retina. FASEB J 2019; 33:13920-13934. [DOI: 10.1096/fj.201901283rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Meichun Xiao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingyu Wang
- GloriousMed Technology Company, Limited, Shanghai, China
| | - Jian Liang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Tianjun Wang
- School of Life Science, University of Liverpool, Liverpool, United Kingdom
| | - Yuanqi Zhai
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Yafang Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjia Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueting Luo
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Fenghua Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| |
Collapse
|
10
|
Al-Khersan H, Hussain RM, Ciulla TA, Dugel PU. Innovative therapies for neovascular age-related macular degeneration. Expert Opin Pharmacother 2019; 20:1879-1891. [PMID: 31298960 DOI: 10.1080/14656566.2019.1636031] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Investigational anti-VEGF treatments for neovascular age-related macular degeneration (nAMD) aim to improve visual outcomes and reduce treatment burden; these include long-acting agents, combination strategies, topical agents, sustained-release, and genetic therapies. Areas covered: The authors provide a comprehensive review of investigational therapies for nAMD, focusing on therapies currently in clinical trial. Expert opinion: Long-acting anti-VEGF agents have demonstrated promising results in phase 3 studies, and include Brolucizumab, a single-chain antibody fragment, and Abicipar, a designed ankyrin repeat protein (DARPin). Other unique anti-VEGF agents in current trials include Conbercept - a fusion protein of the VEGF receptor domains, KSI-301 - an anti-VEGF antibody biopolymer conjugate, and OPT-302 - an inhibitor of VEGF-C/D. Strategies to activate the Tie-2 receptor, some in combination with VEGF inhibition, are of interest, with recent trials of Faricimab, ARP-1536, and nesvacumab. Topical anti-VEGF ± anti-PDGF agents, such as pazopanib, squalamine lactate, regorafenib, and LHA510 have shown limited efficacy and/or have not been advanced, although PAN-90806 continues to advance with promising initial results. Sustained-release anti-VEGF treatments, to address treatment burden, include the ranibizumab Port Delivery System, GB-102, NT-503, hydrogel depot, Durasert, and ENV1305. Similarly, genetic therapies, including RGX-314 and ADVM-022, aim to provide sustained anti-VEGF expression from the retina.
Collapse
Affiliation(s)
- Hasenin Al-Khersan
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine , Miami , FL , USA
| | - Rehan M Hussain
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine , Miami , FL , USA
| | - Thomas A Ciulla
- Department of Ophthalmology, Indiana University School of Medicine , Indianapolis , IN , USA.,Retina Service, Midwest Eye Institute , Indianapolis , IN , USA.,Clearside Biomedical , Alpharetta , GA , USA
| | - Pravin U Dugel
- Retinal Consultants of Arizona , Phoenix , Arizona.,USC Roski Eye Institute, Keck School of Medicine, University of Southern California , Los Angeles , CA , USA
| |
Collapse
|
11
|
Quinn PM, Mulder AA, Henrique Alves C, Desrosiers M, de Vries SI, Klooster J, Dalkara D, Koster AJ, Jost CR, Wijnholds J. Loss of CRB2 in Müller glial cells modifies a CRB1-associated retinitis pigmentosa phenotype into a Leber congenital amaurosis phenotype. Hum Mol Genet 2019; 28:105-123. [PMID: 30239717 DOI: 10.1093/hmg/ddy337] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/17/2018] [Indexed: 11/14/2022] Open
Abstract
Variations in the human Crumbs homolog-1 (CRB1) gene lead to an array of retinal dystrophies including early onset of retinitis pigmentosa (RP) and Leber congenital amaurosis (LCA) in children. To investigate the physiological roles of CRB1 and CRB2 in retinal Müller glial cells (MGCs), we analysed mouse retinas lacking both proteins in MGC. The peripheral retina showed a faster progression of dystrophy than the central retina. The central retina showed retinal folds, disruptions at the outer limiting membrane, protrusion of photoreceptor nuclei into the inner and outer segment layers and ingression of photoreceptor nuclei into the photoreceptor synaptic layer. The peripheral retina showed a complete loss of the photoreceptor synapse layer, intermingling of photoreceptor nuclei within the inner nuclear layer and ectopic photoreceptor cells in the ganglion cell layer. Electroretinography showed severe attenuation of the scotopic a-wave at 1 month of age with responses below detection levels at 3 months of age. The double knockout mouse retinas mimicked a phenotype equivalent to a clinical LCA phenotype due to loss of CRB1. Localization of CRB1 and CRB2 in non-human primate (NHP) retinas was analyzed at the ultrastructural level. We found that NHP CRB1 and CRB2 proteins localized to the subapical region adjacent to adherens junctions at the outer limiting membrane in MGC and photoreceptors. Our data suggest that loss of CRB2 in MGC aggravates the CRB1-associated RP-like phenotype towards an LCA-like phenotype.
Collapse
Affiliation(s)
- Peter M Quinn
- Department of Ophthalmology, Leiden University Medical Center, RC Leiden, The Netherlands
| | - Aat A Mulder
- Department of Cell & Chemical Biology, Leiden University Medical Center (LUMC), RC Leiden, The Netherlands
| | - C Henrique Alves
- Department of Ophthalmology, Leiden University Medical Center, RC Leiden, The Netherlands
| | - Mélissa Desrosiers
- Department of Therapeutics, Institut de la Vision, Sorbonne Universités, UPMC Univ Paris, UMR_S INSERM, CNRS, UMR, Paris, France
| | - Sharon I de Vries
- Department of Axonal Signaling, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), BA Amsterdam, The Netherlands
| | - Jan Klooster
- Department of Retina Signal Processing, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), BA Amsterdam, The Netherlands
| | - Deniz Dalkara
- Department of Therapeutics, Institut de la Vision, Sorbonne Universités, UPMC Univ Paris, UMR_S INSERM, CNRS, UMR, Paris, France
| | - Abraham J Koster
- Department of Cell & Chemical Biology, Leiden University Medical Center (LUMC), RC Leiden, The Netherlands
| | - Carolina R Jost
- Department of Cell & Chemical Biology, Leiden University Medical Center (LUMC), RC Leiden, The Netherlands
| | - Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center, RC Leiden, The Netherlands.,The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, BA Amsterdam, The Netherlands
| |
Collapse
|
12
|
Fu X, Huu VAN, Duan Y, Kermany DS, Valentim CCS, Zhang R, Zhu J, Zhang CL, Sun X, Zhang K. Clinical applications of retinal gene therapies. PRECISION CLINICAL MEDICINE 2018; 1:5-20. [PMID: 35694125 PMCID: PMC8982485 DOI: 10.1093/pcmedi/pby004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/27/2018] [Accepted: 04/03/2018] [Indexed: 02/05/2023] Open
Abstract
Retinal degenerative diseases are a major cause of blindness. Retinal gene therapy is a
trail-blazer in the human gene therapy field, leading to the first FDA approved gene
therapy product for a human genetic disease. The application of Clustered Regularly
Interspaced Short Palindromic Repeat/Cas9 (CRISPR/Cas9)-mediated gene editing technology
is transforming the delivery of gene therapy. We review the history, present, and future
prospects of retinal gene therapy.
Collapse
Affiliation(s)
- Xin Fu
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomic Medicine, University of California, San Diego, La Jolla, California, USA
| | - Viet Anh Nguyen Huu
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomic Medicine, University of California, San Diego, La Jolla, California, USA
| | - Yaou Duan
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomic Medicine, University of California, San Diego, La Jolla, California, USA
| | - Daniel S Kermany
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomic Medicine, University of California, San Diego, La Jolla, California, USA
| | - Carolina C S Valentim
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomic Medicine, University of California, San Diego, La Jolla, California, USA
| | - Runze Zhang
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomic Medicine, University of California, San Diego, La Jolla, California, USA
| | - Jie Zhu
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomic Medicine, University of California, San Diego, La Jolla, California, USA
| | - Charlotte L Zhang
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomic Medicine, University of California, San Diego, La Jolla, California, USA
| | - Xiaodong Sun
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai General Hospital, Shanghai Jiaodong University, Shanghai, China
| | - Kang Zhang
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomic Medicine, University of California, San Diego, La Jolla, California, USA
- Molecular Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Tu L, Wang JH, Barathi VA, Prea SM, He Z, Lee JH, Bender J, King AE, Logan GJ, Alexander IE, Bee YS, Tai MH, Dusting GJ, Bui BV, Zhong J, Liu GS. AAV-mediated gene delivery of the calreticulin anti-angiogenic domain inhibits ocular neovascularization. Angiogenesis 2018; 21:95-109. [PMID: 29318471 DOI: 10.1007/s10456-017-9591-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 12/23/2017] [Indexed: 12/20/2022]
Abstract
Ocular neovascularization is a common pathological feature in diabetic retinopathy and neovascular age-related macular degeneration that can lead to severe vision loss. We evaluated the therapeutic efficacy of a novel endogenous inhibitor of angiogenesis, the calreticulin anti-angiogenic domain (CAD180), and its functional 112-residue fragment, CAD-like peptide 112 (CAD112), delivered using a self-complementary adeno-associated virus serotype 2 (scAAV2) in rodent models of oxygen-induced retinopathy and laser-induced choroidal neovascularization. The expression of CAD180 and CAD112 was elevated in human umbilical vein endothelial cells transduced with scAAV2-CAD180 or scAAV2-CAD112, respectively, and both inhibited angiogenic activity in vitro. Intravitreal gene delivery of scAAV2-CAD180 or scAAV2-CAD112 significantly inhibited ischemia-induced retinal neovascularization in rat eyes (CAD180: 52.7% reduction; CAD112: 49.2% reduction) compared to scAAV2-mCherry, as measured in retinal flatmounts stained with isolectin B4. Moreover, the retinal structure and function were unaffected by scAAV2-CAD180 or scAAV2-CAD112, as measured by optical coherence tomography and electroretinography. Moreover, subretinal delivery of scAAV2-CAD180 or scAAV2-CAD112 significantly attenuated laser-induced choroidal neovascularization in mouse eyes compared to scAAV2-mCherry, as measured by fundus fluorescein angiography (CAD180: 62.4% reduction; CAD112: 57.5% reduction) and choroidal flatmounts (CAD180: 40.21% reduction; CAD112: 43.03% reduction). Gene delivery using scAAV2-CAD180 or scAAV2-CAD112 has significant potential as a therapeutic option for the management of ocular neovascularization.
Collapse
Affiliation(s)
- Leilei Tu
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
| | - Jiang-Hui Wang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC, Australia
| | - Veluchamy A Barathi
- Translational Pre-clinical Model Platform, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, DUKE-NUS Graduate Medical School, Singapore, Singapore
| | - Selwyn M Prea
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Zheng He
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Jia Hui Lee
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
| | - James Bender
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, TAS, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, TAS, Australia
| | - Grant J Logan
- Gene Therapy Research Unit, Children's Medical Research Institute and Sydney Children's Hospitals Network, University of Sydney, Sydney, NSW, Australia
| | - Ian E Alexander
- Gene Therapy Research Unit, Children's Medical Research Institute and Sydney Children's Hospitals Network, University of Sydney, Sydney, NSW, Australia
- Discipline of Child and Adolescent Health, University of Sydney, Westmead, NSW, Australia
| | - Youn-Shen Bee
- Department of Ophthalmology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Ming-Hong Tai
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Gregory J Dusting
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Jingxiang Zhong
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| | - Guei-Sheung Liu
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia.
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC, Australia.
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.
- , Liverpool St, Hobart, TAS, 7000, Australia.
| |
Collapse
|
14
|
Hartman RR, Kompella UB. Intravitreal, Subretinal, and Suprachoroidal Injections: Evolution of Microneedles for Drug Delivery. J Ocul Pharmacol Ther 2017; 34:141-153. [PMID: 29206556 PMCID: PMC5963636 DOI: 10.1089/jop.2017.0121] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 01/04/2023] Open
Abstract
Even though the very thought of an injection into the eye may be frightening, an estimated 6 million intravitreal (IVT) injections were made in the USA during 2016. With the introduction of new therapeutic agents, this number is expected to increase. In addition, drug products that are injectable in ocular compartments other than the vitreous humor are expected to enter the back of the eye market in the not so distant future. Besides the IVT route, some of the most actively investigated routes of invasive administration to the eye include periocular, subretinal, and suprachoroidal (SC) routes. While clinical efficacy is the driving force behind new injectable drug product development for the eye, safety is also being improved with time. In the case of IVT injections, the procedural guidelines have evolved over the years to improve patient comfort and reduce injection-related injury and infection. Similar advances are anticipated for other routes of administration of injectable products to the eye. In addition to procedural improvements, the design of needles, particularly those with smaller diameters, length, and controlled bevel angles are expected to improve overall safety and acceptance of injected ophthalmic drug products. A key development in this area is the introduction of microneedles of a length less than a millimeter that can target the SC space. In the future, needles with smaller diameters and lengths, potentially approaching nanodimensions, are expected to revolutionize ophthalmic disease management.
Collapse
Affiliation(s)
- Rachel R. Hartman
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Uday B. Kompella
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
15
|
Hussain RM, Ciulla TA. Emerging vascular endothelial growth factor antagonists to treat neovascular age-related macular degeneration. Expert Opin Emerg Drugs 2017; 22:235-246. [DOI: 10.1080/14728214.2017.1362390] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Rehan M Hussain
- Department of Ophthalmology, 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
| |
Collapse
|
16
|
Moore NA, Bracha P, Hussain RM, Morral N, Ciulla TA. Gene therapy for age-related macular degeneration. Expert Opin Biol Ther 2017; 17:1235-1244. [PMID: 28726562 DOI: 10.1080/14712598.2017.1356817] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION In neovascular age related macular degeneration (nAMD), gene therapy to chronically express anti-vascular endothelial growth factor (VEGF) proteins could ameliorate the treatment burden of chronic intravitreal therapy and improve limited visual outcomes associated with 'real world' undertreatment. Areas covered: In this review, the authors assess the evolution of gene therapy for AMD. Adeno-associated virus (AAV) vectors can transduce retinal pigment epithelium; one such early application was a phase I trial of AAV2-delivered pigment epithelium derived factor gene in advanced nAMD. Subsequently, gene therapy for AMD shifted to the investigation of soluble fms-like tyrosine kinase-1 (sFLT-1), an endogenously expressed VEGF inhibitor, binding and neutralizing VEGF-A. After some disappointing results, research has centered on novel vectors, including optimized AAV2, AAV8 and lentivirus, as well as genes encoding other anti-angiogenic proteins, including ranibizumab, aflibercept, angiostatin and endostatin. Also, gene therapy targeting the complement system is being investigated for geographic atrophy due to non-neovascular AMD. Expert opinion: The success of gene therapy for AMD will depend on the selection of the most appropriate therapeutic protein and its level of chronic expression. Future investigations will center on optimizing vector, promoter and delivery methods, and evaluating the risks of the chronic expression of anti-angiogenic or anti-complement proteins.
Collapse
Affiliation(s)
- Nicholas A Moore
- a Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Peter Bracha
- a Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Rehan M Hussain
- a Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Nuria Morral
- c Department of Medical and Molecular Genetics , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Thomas A Ciulla
- a Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA.,b Retina Service , Midwest Eye Institute , Indianapolis , IN , USA
| |
Collapse
|
17
|
Affiliation(s)
- Elizabeth P Rakoczy
- Centre for Ophthalmology and Visual Sciences, University of Western Australia, Perth, WA 6009, Australia.
| |
Collapse
|
18
|
Constable IJ, Lai CM, Magno AL, French MA, Barone SB, Schwartz SD, Blumenkranz MS, Degli-Esposti MA, Rakoczy EP. Gene Therapy in Neovascular Age-related Macular Degeneration: Three-Year Follow-up of a Phase 1 Randomized Dose Escalation Trial. Am J Ophthalmol 2017; 177:150-158. [PMID: 28245970 DOI: 10.1016/j.ajo.2017.02.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/16/2017] [Accepted: 02/17/2017] [Indexed: 01/23/2023]
Abstract
PURPOSE To assess the safety of rAAV.sFlt-1 subretinal injection in neovascular age-related macular degeneration (wet AMD) over 36 months. DESIGN Phase 1 dose escalation trial. METHODS Eight subjects with advanced, treatment-experienced wet AMD were randomly assigned (3:1) to treatment and non-gene therapy control groups. Eligible subjects were ≥65 years, had wet AMD, and had best-corrected visual acuity (BCVA) 10/200 to 20/80 in the study eye and 20/200 or better in the other eye. Three of the treatment group subjects received low-dose (1 × 1010 vector genomes) and 3 high-dose (1 × 1011 vector genomes) rAAV.sFLT-1 via subretinal injection. Study monitoring was monthly to the primary endpoint at month 12 and then protocol-driven follow-up study visits were conducted at months 18 and 36. All subjects received intravitreal ranibizumab at baseline and at week 4, and retreatment injections at subsequent visits based on prespecified criteria for active wet AMD. The primary endpoint was ocular and systemic safety, but exploratory data including BCVA, retinal center point thickness, and the number of ranibizumab retreatments at and between study visits were also analyzed. RESULTS Six of the 8 subjects completed the 36-month study. Subretinal injection with pars plana vitrectomy was well tolerated in this cohort. No ocular or systemic safety signals were observed during the long-term follow-up period. Exploratory data analysis suggests stability of wet AMD over the 36-month period. CONCLUSIONS Subretinal delivery of rAAV.sFLT-1 was well tolerated and demonstrated a favourable safety profile through month 36. Thus, rAAV.sFLT-1 could be safely considered for future evaluation in the treatment of wet AMD.
Collapse
Affiliation(s)
- Ian J Constable
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, Western Australia, Australia; Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Chooi-May Lai
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, Western Australia, Australia
| | - Aaron L Magno
- Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Martyn A French
- School of Pathology and Laboratory Medicine, The University of Western Australia, Crawley, Western Australia, Australia; Department of Clinical Immunology, Royal Perth Hospital and PathWest Laboratory Medicine, Perth, Australia
| | | | | | - Mark S Blumenkranz
- Byers Eye Institute, Stanford University School of Medicine, Palo Alto, California
| | - Mariapia A Degli-Esposti
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, Western Australia, Australia
| | - Elizabeth P Rakoczy
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, Western Australia, Australia.
| |
Collapse
|
19
|
Zhang X, Bohner A, Bhuvanagiri S, Uehara H, Upadhyay AK, Emerson LL, Bondalapati S, Muddana SK, Fang D, Li M, Sandhu Z, Hussain A, Carroll LS, Tiem M, Archer B, Kompella U, Patil R, Ambati BK. Targeted Intraceptor Nanoparticle for Neovascular Macular Degeneration: Preclinical Dose Optimization and Toxicology Assessment. Mol Ther 2017; 25:1606-1615. [PMID: 28236576 DOI: 10.1016/j.ymthe.2017.01.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 01/13/2017] [Accepted: 01/14/2017] [Indexed: 10/20/2022] Open
Abstract
Neovascular age-related macular degeneration (AMD) is treated with anti-VEGF intravitreal injections, which can cause geographic atrophy, infection, and retinal fibrosis. To minimize these toxicities, we developed a nanoparticle delivery system for recombinant Flt23k intraceptor plasmid (RGD.Flt23k.NP) to suppress VEGF intracellularly within choroidal neovascular (CNV) lesions in a laser-induced CNV mouse model through intravenous administration. In the current study, we examined the efficacy and safety of RGD.Flt23k.NP in mice. The effect of various doses was determined using fluorescein angiography and optical coherence tomography to evaluate CNV leakage and volume. Efficacy was determined by the rate of inhibition of CNV volume at 2 weeks post-treatment. RGD.Flt23k.NP had peak efficacy at a dose range of 30-60 μg pFlt23k/mouse. Using the lower dose (30 μg pFlt23k/mouse), RGD.Flt23k.NP safety was determined both in single-dose groups and in repeat-dose (three times) groups by measuring body weight, organ weight, hemoglobin levels, complement C3 levels, and histological changes in vital organs. Neither toxicity nor inflammation from RGD.Flt23k.NP was detected. No side effect was detected on visual function. Thus, systemic RGD.Flt23k.NP may be an alternative to standard intravitreal anti-VEGF therapy for the treatment of neovascular AMD.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Moran Eye Center, University of Utah, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA
| | - Austin Bohner
- Moran Eye Center, University of Utah, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA
| | - Sai Bhuvanagiri
- Moran Eye Center, University of Utah, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA
| | - Hironori Uehara
- Moran Eye Center, University of Utah, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA
| | - Arun Kumar Upadhyay
- University of Colorado School of Pharmacy, Mail Stop C238, 12850 E. Montview Blvd. V20-4129, Aurora, CO 80045, USA
| | - Lyska L Emerson
- Department of Pathology, University of Utah and Associated Regional and University Pathologists (ARUP) Laboratories, 500 Chipeta Way, Salt Lake City, UT 84108, USA
| | - Sailaja Bondalapati
- Moran Eye Center, University of Utah, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA
| | - Santosh Kumar Muddana
- Moran Eye Center, University of Utah, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA
| | - Daniel Fang
- Moran Eye Center, University of Utah, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA
| | - Miaoling Li
- Moran Eye Center, University of Utah, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Zoya Sandhu
- Moran Eye Center, University of Utah, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA
| | - Alya Hussain
- Moran Eye Center, University of Utah, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA
| | - Lara S Carroll
- Moran Eye Center, University of Utah, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA
| | - Michelle Tiem
- Moran Eye Center, University of Utah, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA
| | - Bonnie Archer
- Moran Eye Center, University of Utah, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA
| | - Uday Kompella
- University of Colorado School of Pharmacy, Mail Stop C238, 12850 E. Montview Blvd. V20-4129, Aurora, CO 80045, USA
| | - Rajkumar Patil
- Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Balamurali K Ambati
- Moran Eye Center, University of Utah, 65 Mario Capecchi Dr., Salt Lake City, UT 84132, USA.
| |
Collapse
|
20
|
Constable IJ, Pierce CM, Lai CM, Magno AL, Degli-Esposti MA, French MA, McAllister IL, Butler S, Barone SB, Schwartz SD, Blumenkranz MS, Rakoczy EP. Phase 2a Randomized Clinical Trial: Safety and Post Hoc Analysis of Subretinal rAAV.sFLT-1 for Wet Age-related Macular Degeneration. EBioMedicine 2016; 14:168-175. [PMID: 27865764 PMCID: PMC5161436 DOI: 10.1016/j.ebiom.2016.11.016] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND We present the results of a Phase 2a randomized controlled trial investigating the safety, and secondary endpoints of subretinal rAAV.sFLT-1 gene therapy in patients with active wet age-related macular degeneration (wAMD). METHODS All patients (n=32), (ClinicalTrials.gov; NCT01494805), received ranibizumab injections at baseline and week 4, and thereafter according to prespecified criteria. Patients in the gene therapy group (n=21) received rAAV.sFLT-1 (1×1011vg). All patients were assessed every 4weeks to the week 52 primary endpoint. FINDINGS Ocular adverse events (AEs) in the rAAV.sFLT-1 group were mainly procedure related and self-resolved. All 11 phakic patients in the rAAV.sFLT-1 group showed progression of cataract following vitrectomy. No systemic safety signals were observed and none of the serious AEs were associated with rAAV.sFLT-1. AAV2 capsid was not detected and rAAV.sFLT-1 DNA was detected transiently in the tears of 13 patients. ELISPOT analysis did not identify any notable changes in T-cell response. In the rAAV.sFLT-1 group 12 patients had neutralizing antibodies (nAb) to AAV2. There was no change in sFLT-1 levels in bodily fluids. In the rAAV.sFLT-1 group, Best Corrected Visual Acuity (BCVA) improved by a median of 1.0 (IQR: -3.0 to 9.0) Early Treatment Diabetic Retinopathy Study (ETDRS) letters from baseline compared to a median of -5.0 (IQR: -17.5 to 1.0) ETDRS letters change in the control group. Twelve (57%) patients in the rAAV.sFLT-1 group maintained or improved vision compared to 4 (36%) in the control group. The median number of ranibizumab retreatments was 2.0 (IQR: 1.0 to 6.0) for the gene therapy group compared to 4.0 (IQR: 3.5 to 4.0) for the control group. Interpretation rAAV.sFLT-1 combined with the option for co-treatment appears to be a safe and promising approach to the treatment of wAMD. FUNDING National Health and Medical Research Council of Australia (AP1010405), Lions Eye Institute, Perth Australia, Avalanche Biotechnologies, Menlo Pk, CA, USA.
Collapse
Affiliation(s)
- Ian J Constable
- Lions Eye Institute, Nedlands, WA, Australia; Sir Charles Gairdner Hospital, Nedlands, WA, Australia; Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA, Australia
| | | | - Chooi-May Lai
- Lions Eye Institute, Nedlands, WA, Australia; Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA, Australia
| | | | - Mariapia A Degli-Esposti
- Lions Eye Institute, Nedlands, WA, Australia; Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA, Australia
| | - Martyn A French
- School of Pathology and Laboratory Medicine, The University of Western Australia, Crawley, WA, Australia; Department of Clinical Immunology, The University of Western Australia, Crawley, WA, Australia
| | - Ian L McAllister
- Lions Eye Institute, Nedlands, WA, Australia; Sir Charles Gairdner Hospital, Nedlands, WA, Australia; Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA, Australia
| | - Steve Butler
- Avalanche Biotechnologies, Inc., Menlo Park, CA, USA
| | | | | | - Mark S Blumenkranz
- Byers Eye Institute, Stanford Department of Ophthalmology, Palo Alto, CA, USA
| | - Elizabeth P Rakoczy
- Lions Eye Institute, Nedlands, WA, Australia; Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA, Australia.
| |
Collapse
|
21
|
Constable IJ, Blumenkranz MS, Schwartz SD, Barone S, Lai CM, Rakoczy EP. Gene Therapy for Age-Related Macular Degeneration. Asia Pac J Ophthalmol (Phila) 2016; 5:300-3. [PMID: 27488071 DOI: 10.1097/apo.0000000000000222] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The purpose of this article was to evaluate safety and signals of efficacy of gene therapy with subretinal rAAV.sFlt-1 for wet age-related macular degeneration (wet AMD). A phase 1 dose-escalating single-center controlled unmasked human clinical trial was followed up by extension of the protocol to a phase 2A single-center trial. rAAV.sFlt-1 vector was used to deliver a naturally occurring anti-vascular endothelial growth factor agent, sFlt-1, into the subretinal space. In phase 1, step 1 randomized 3 subjects to low-dose rAAV.sFlt-1 (1 × 10 vector genomes) and 1 subject to the control arm; step 2 randomized an additional 3 subjects to treatment with high-dose rAAV.sFlt-1 (1 × 10 vector genomes) and 1 subject to the control arm. Follow-up studies demonstrated that rAAV.sFlt-1 was well tolerated with a favorable safety profile in these elderly subjects with wet AMD. Subretinal injection was highly reproducible, and no drug-related adverse events were reported. Procedure-related adverse events were mild and self-resolving. Two phakic patients developed cataract and underwent cataract surgery. Four of the 6 patients responded better than the small control group in this study and historical controls in terms of maintaining vision and a relatively dry retina with zero ranibizumab retreatments per annum. Two patients required 1 ranibizumab injection over the 52-week follow-up period. rAAV.sFlt-1 gene therapy may prove to be a potential adjunct or alternative to conventional intravitreal injection for patients with wet AMD by providing extended delivery of a naturally occurring antiangiogenic protein.
Collapse
Affiliation(s)
- Ian Jeffery Constable
- From the *Lions Eye Institute; †Sir Charles Gairdner Hospital, Nedlands; ‡Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, Western Australia, Australia; §University of California Los Angeles, Los Angeles; ¶Byers Eye Institute at Stanford, Palo Alto; and ‖Avalanche Biotechnologies, Inc, Menlo Park, CA
| | | | | | | | | | | |
Collapse
|
22
|
Lambert NG, Zhang X, Rai RR, Uehara H, Choi S, Carroll LS, Das SK, Cahoon JM, Kirk BH, Bentley BM, Ambati BK. Subretinal AAV2.COMP-Ang1 suppresses choroidal neovascularization and vascular endothelial growth factor in a murine model of age-related macular degeneration. Exp Eye Res 2016; 145:248-257. [PMID: 26775053 PMCID: PMC5862038 DOI: 10.1016/j.exer.2016.01.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 12/21/2015] [Accepted: 01/12/2016] [Indexed: 11/17/2022]
Abstract
To assess whether Tie2-mediated vascular stabilization ameliorates neovascular age-related macular degeneration (AMD), we investigated the impact of adeno-associated virus-mediated gene therapy with cartilage oligomeric matrix protein angiopoietin-1 (AAV2.COMP-Ang1) on choroidal neovascularization (CNV), vascular endothelial growth factor (VEGF), and hypoxia-inducible factor (HIF) in a mouse model of the disease. We treated mice with subretinal injections of AAV2.COMP-Ang1 or control (AAV2.AcGFP, AAV2.LacZ, and phosphate-buffered saline). Subretinal AAV2 localization and plasmid protein expression was verified in the retinal pigment epithelium (RPE)/choroid of mice treated with all AAV2 constructs. Laser-assisted simulation of neovascular AMD was performed and followed by quantification of HIF, VEGF, and CNV in each experimental group. We found that AAV2.COMP-Ang1 was associated with a significant reduction in VEGF levels (29-33%, p < 0.01) and CNV volume (60-70%, p < 0.01), without a concomitant decrease in HIF1-α, compared to all controls. We concluded that a) AAV2 is a viable vector for delivering COMP-Ang1 to subretinal tissues, b) subretinal COMP-Ang1 holds promise as a prospective treatment for neovascular AMD, and c) although VEGF suppression in the RPE/choroid may be one mechanism by which AAV2.COMP-Ang1 reduces CNV, this therapeutic effect may be hypoxia-independent. Taken together, these findings suggest that AAV2.COMP-Ang1 has potential to serve as an alternative or complementary option to anti-VEGF agents for the long-term amelioration of neovascular AMD.
Collapse
Affiliation(s)
| | - Xiaohui Zhang
- Ambati Lab, John A. Moran Eye Center, Salt Lake City, UT, USA; Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT, USA
| | - Ruju R Rai
- Ambati Lab, John A. Moran Eye Center, Salt Lake City, UT, USA; Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT, USA
| | - Hironori Uehara
- Ambati Lab, John A. Moran Eye Center, Salt Lake City, UT, USA; Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT, USA
| | - Susie Choi
- Ambati Lab, John A. Moran Eye Center, Salt Lake City, UT, USA; Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT, USA
| | - Lara S Carroll
- Ambati Lab, John A. Moran Eye Center, Salt Lake City, UT, USA; Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT, USA
| | - Subrata K Das
- Ambati Lab, John A. Moran Eye Center, Salt Lake City, UT, USA; Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT, USA
| | - Judd M Cahoon
- Ambati Lab, John A. Moran Eye Center, Salt Lake City, UT, USA
| | - Brian H Kirk
- Ambati Lab, John A. Moran Eye Center, Salt Lake City, UT, USA
| | | | - Balamurali K Ambati
- Ambati Lab, John A. Moran Eye Center, Salt Lake City, UT, USA; Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
23
|
Agarwal A, Ingham SA, Harkins KA, Do DV, Nguyen QD. The role of pharmacogenetics and advances in gene therapy in the treatment of diabetic retinopathy. Pharmacogenomics 2016; 17:309-20. [PMID: 26807609 DOI: 10.2217/pgs.15.173] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Diabetic retinopathy (DR) and its complications such as diabetic macular edema continue to remain a major cause for legal blindness in the developed world. While the introduction of anti-tVEGF agents has significantly improved visual outcomes of patients with DR, unpredictable response, largely due to genetic polymorphisms, appears to be a challenge with this therapy. With advances in identification of various genetic biomarkers, novel therapeutic strategies consisting of gene transfer are being developed and tested for patients with DR. Application of pharmacogenetic principles appears to be a promising futuristic strategy to attenuate diabetes-mediated retinal vasculopathy. In this comprehensive review, data from recent studies in the field of pharmacogenomics for the treatment of DR have been provided.
Collapse
Affiliation(s)
- Aniruddha Agarwal
- Ocular Imaging Research & Reading Center (OIRRC), Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, South 42nd Street & Emile St, Omaha, NE 68198, USA
| | - Sally A Ingham
- College of Medicine, University of Nebraska Medical Center, South 42nd Street & Emile St, Omaha, NE 68198, USA
| | - Keegan A Harkins
- Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, South 42nd Street & Emile St, Omaha, NE 68198, USA
| | - Diana V Do
- Ocular Imaging Research & Reading Center (OIRRC), Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, South 42nd Street & Emile St, Omaha, NE 68198, USA.,Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, South 42nd Street & Emile St, Omaha, NE 68198, USA
| | - Quan Dong Nguyen
- Ocular Imaging Research & Reading Center (OIRRC), Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, South 42nd Street & Emile St, Omaha, NE 68198, USA.,Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, South 42nd Street & Emile St, Omaha, NE 68198, USA
| |
Collapse
|
24
|
Rakoczy EP, Lai CM, Magno AL, Wikstrom ME, French MA, Pierce CM, Schwartz SD, Blumenkranz MS, Chalberg TW, Degli-Esposti MA, Constable IJ. Gene therapy with recombinant adeno-associated vectors for neovascular age-related macular degeneration: 1 year follow-up of a phase 1 randomised clinical trial. Lancet 2015; 386:2395-403. [PMID: 26431823 DOI: 10.1016/s0140-6736(15)00345-1] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Neovascular, or wet, age-related macular degeneration causes central vision loss and represents a major health problem in elderly people, and is currently treated with frequent intraocular injections of anti-VEGF protein. Gene therapy might enable long-term anti-VEGF therapy from a single treatment. We tested the safety of rAAV.sFLT-1 in treatment of wet age-related macular degeneration with a single subretinal injection. METHODS In this single-centre, phase 1, randomised controlled trial, we enrolled patients with wet age-related macular degeneration at the Lions Eye Institute and the Sir Charles Gairdner Hospital (Nedlands, WA, Australia). Eligible patients had to be aged 65 years or older, have age-related macular degeneration secondary to active subfoveal choroidal neovascularisation, with best corrected visual acuity (BCVA) of 3/60-6/24 and 6/60 or better in the other eye. Patients were randomly assigned (3:1) to receive either 1 × 10(10) vector genomes (vg; low-dose rAAV.sFLT-1 group) or 1 × 10(11) vg (high-dose rAAV.sFLT-1 group), or no gene-therapy treatment (control group). Randomisation was done by sequential group assignment. All patients and investigators were unmasked. Staff doing the assessments were masked to the study group at study visits. All patients received ranibizumab at baseline and week 4, and rescue treatment during follow-up based on prespecified criteria including BCVA measured on the Early Treatment Diabetic Retinopathy Study (EDTRS) scale, optical coherence tomography, and fluorescein angiography. The primary endpoint was ocular and systemic safety. This trial is registered with ClinicalTrials.gov, number NCT01494805. FINDINGS From Dec 16, 2011, to April 5, 2012, we enrolled nine patients of whom eight were randomly assigned to receive either intervention (three patients in the low-dose rAAV.sFLT-1 group and three patients in the high-dose rAAV.sFLT-1 group) or no treatment (two patients in the control group). Subretinal injection of rAAV.sFLT-1 was highly reproducible. No drug-related adverse events were noted; procedure-related adverse events (subconjunctival or subretinal haemorrhage and mild cell debris in the anterior vitreous) were generally mild and self-resolving. There was no evidence of chorioretinal atrophy. Clinical laboratory assessments generally remained unchanged from baseline. Four (67%) of six patients in the treatment group required zero rescue injections, and the other two (33%) required only one rescue injection each. INTERPRETATION rAAV.sFLT-1 was safe and well tolerated. These results support ocular gene therapy as a potential long-term treatment option for wet age-related macular degeneration. FUNDING National Health and Medical Research Council of Australia, Richard Pearce Bequest, Lions Save Sight Foundation, Brian King Fellowship, and Avalanche Biotechnologies, Inc.
Collapse
Affiliation(s)
- Elizabeth P Rakoczy
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA, Australia; Lions Eye Institute, Nedlands, WA, Australia.
| | - Chooi-May Lai
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA, Australia; Lions Eye Institute, Nedlands, WA, Australia
| | | | - Matthew E Wikstrom
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA, Australia; Lions Eye Institute, Nedlands, WA, Australia
| | - Martyn A French
- School of Pathology and Laboratory Medicine, The University of Western Australia, Crawley, WA, Australia
| | | | | | | | | | - Mariapia A Degli-Esposti
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA, Australia; Lions Eye Institute, Nedlands, WA, Australia
| | - Ian J Constable
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA, Australia; Lions Eye Institute, Nedlands, WA, Australia; Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| |
Collapse
|
25
|
Affiliation(s)
- Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK; Oxford University Eye Hospital, Oxford, UK; Moorfields Eye Hospital NHS Foundation Trust, London, UK.
| |
Collapse
|
26
|
Zhang JX, Wang NL, Lu QJ. Development of gene and stem cell therapy for ocular neurodegeneration. Int J Ophthalmol 2015; 8:622-30. [PMID: 26086019 DOI: 10.3980/j.issn.2222-3959.2015.03.33] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 11/12/2014] [Indexed: 12/21/2022] Open
Abstract
Retinal degenerative diseases pose a serious threat to eye health, but there is currently no effective treatment available. Recent years have witnessed rapid development of several cutting-edge technologies, such as gene therapy, stem cell therapy, and tissue engineering. Due to the special features of ocular structure, some of these technologies have been translated into ophthalmological clinic practice with fruitful achievements, setting a good example for other fields. This paper reviews the development of the gene and stem cell therapies in ophthalmology.
Collapse
Affiliation(s)
- Jing-Xue Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, China
| | - Ning-Li Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, China
| | - Qing-Jun Lu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, China
| |
Collapse
|
27
|
Agarwal A, Rhoades WR, Hanout M, Soliman MK, Sarwar S, Sadiq MA, Sepah YJ, Do DV, Nguyen QD. Management of neovascular age-related macular degeneration: current state-of-the-art care for optimizing visual outcomes and therapies in development. Clin Ophthalmol 2015; 9:1001-15. [PMID: 26089632 PMCID: PMC4467654 DOI: 10.2147/opth.s74959] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Contemporary management of neovascular age-related macular degeneration (AMD) has evolved significantly over the last few years. The goal of treatment is shifting from merely salvaging vision to maintaining a high quality of life. There have been significant breakthroughs in the identification of viable drug targets and gene therapies. Imaging tools with near-histological precision have enhanced our knowledge about pathophysiological mechanisms that play a role in vision loss due to AMD. Visual, social, and vocational rehabilitation are all important treatment goals. In this review, evidence from landmark clinical trials is summarized to elucidate the optimum modern-day management of neovascular AMD. Therapeutic strategies currently under development, such as gene therapy and personalized medicine, are also described.
Collapse
Affiliation(s)
- Aniruddha Agarwal
- Stanley M Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, USA
| | - William R Rhoades
- Stanley M Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mostafa Hanout
- Stanley M Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mohamed Kamel Soliman
- Stanley M Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, USA
| | - Salman Sarwar
- Stanley M Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mohammad Ali Sadiq
- Stanley M Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yasir Jamal Sepah
- Stanley M Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, USA
| | - Diana V Do
- Stanley M Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, USA
| | - Quan Dong Nguyen
- Stanley M Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
28
|
Wu Z, Hiriyanna S, Qian H, Mookherjee S, Campos MM, Gao C, Fariss R, Sieving PA, Li T, Colosi P, Swaroop A. A long-term efficacy study of gene replacement therapy for RPGR-associated retinal degeneration. Hum Mol Genet 2015; 24:3956-70. [PMID: 25877300 DOI: 10.1093/hmg/ddv134] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 04/13/2015] [Indexed: 11/14/2022] Open
Abstract
Mutations in the retinitis pigmentosa GTPase regulator (RPGR) gene account for >70% of X-linked retinitis pigmentosa (XLRP) and 15-20% of all inherited retinal degeneration. Gene replacement therapy for RPGR-XLRP was hampered by the relatively slow disease progression in mouse models and by difficulties in cloning the full-length RPGR-ORF15 cDNA that includes a purine-rich 3'-coding region; however, its effectiveness has recently been demonstrated in four dogs with RPGR mutations. To advance the therapy to clinical stage, we generated new stable vectors in AAV8 or AAV9 carrying mouse and human full-length RPGR-ORF15-coding sequence and conducted a comprehensive long-term dose-efficacy study in Rpgr-knockout mice. After validating their ability to produce full-length proteins that localize to photoreceptor connecting cilia, we evaluated various vector doses in mice during a 2-year study. We demonstrate that eyes treated with a single injection of mouse or human RPGR-ORF15 vector at an optimal dose maintained the expression of RPGR-ORF15 throughout the study duration and exhibited higher electroretinogram amplitude, thicker photoreceptor layer and better targeting of opsins to outer segments compared with sham-treated eyes. Furthermore, mice that received treatment at an advanced age also showed remarkable preservation of retinal structure and function. Retinal toxicity was observed at high vector doses, highlighting the importance of careful dose optimization in future clinical experiments. Our long-term dose-efficacy study should facilitate the design of human trials with human RPGR-ORF15 vector as a clinical candidate.
Collapse
Affiliation(s)
- Zhijian Wu
- National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Suja Hiriyanna
- National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haohua Qian
- National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Maria M Campos
- National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chun Gao
- National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert Fariss
- National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paul A Sieving
- National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tiansen Li
- National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter Colosi
- National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anand Swaroop
- National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
29
|
Abstract
The field of gene therapy for retinal blinding disorders is experiencing incredible momentum, justified by hopeful results in early stage clinical trials for inherited retinal degenerations. The premise of the use of the gene as a drug has come a long way, and may have found its niche in the treatment of retinal disease. Indeed, with only limited treatment options available for retinal indications, gene therapy has been proven feasible, safe, and effective and may lead to durable effects following a single injection. Here, we aim at putting into context the promise and potential, the technical, clinical, and economic boundaries limiting its application and development, and speculate on a future in which gene therapy is an integral component of ophthalmic clinical care.
Collapse
Affiliation(s)
- Luk H Vandenberghe
- Ocular Genomics Institute, Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts 02114
| |
Collapse
|
30
|
Wimmer T, Lorenz B, Stieger K. Functional Characterization of AAV-Expressed Recombinant Anti-VEGF Single-Chain Variable Fragments In Vitro. J Ocul Pharmacol Ther 2015; 31:269-76. [PMID: 25867736 DOI: 10.1089/jop.2014.0125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Most retinal neovascular disorders are caused by upregulation of vascular endothelial growth factor (VEGF) expression. These disorders are treated with repeated injections of anti-VEGF molecules, which may have severe side effects. The expression of anti-VEGF molecules by the retina itself in a controlled manner following adeno-associated viral (AAV) gene transfer could be a replacement of this therapy. METHODS The open reading frames (orf) of the light and the heavy chain of ranibizumab were cloned into an expression plasmid separated by an internal ribosomal entry site (IRES). The construct was mutated to generate ranibizumab single-chain variable fragments (scFv). Expression was verified by western blotting and the concentrations were measured with a custom-made ranibizumab ELISA. Biological activity, VEGF-binding properties, and the doxycycline-dependent induction of anti-VEGF expression were tested. An AAV2/5 vector was generated containing the optimal variant Ra02. RESULTS Ra01-Ra05 molecules were detected in the cell culture medium. While the VEGF-binding affinity was significantly lower for Ra01 and Ra02 compared to Lucentis(®), the inhibition of cell migration was comparable and the maximum inhibition of Ra01 and Ra02 was reached at lower doses. The expression of Ra01 and Ra02 was shown to be regulable with the TetOn-system(®) as plasmid (Ra01, Ra02) and AAV vector construct (Ra02). CONCLUSION Ra01 and Ra02 can be produced in eukaryotic cells after AAV-mediated gene transfer in a regulable manner in vitro and display comparable biological activity as Lucentis. These results are the basis for in vivo studies in human VEGF-overexpressing mice, a model for human neovascular disorders.
Collapse
Affiliation(s)
- Tobias Wimmer
- Department of Ophthalmology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Birgit Lorenz
- Department of Ophthalmology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Knut Stieger
- Department of Ophthalmology, Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
31
|
Ronquillo CC, Passi SF, Ambati BK. Restoring Physiologic Barriers Against Neovascular Invasion. ESSENTIALS IN OPHTHALMOLOGY 2015. [DOI: 10.1007/978-3-662-45188-5_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
32
|
Pechan P, Wadsworth S, Scaria A. Gene Therapies for Neovascular Age-Related Macular Degeneration. Cold Spring Harb Perspect Med 2014; 5:a017335. [PMID: 25524721 DOI: 10.1101/cshperspect.a017335] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pathological neovascularization is a key component of the neovascular form (also known as the wet form) of age-related macular degeneration (AMD) and proliferative diabetic retinopathy. Several preclinical studies have shown that antiangiogenesis strategies are effective for treating neovascular AMD in animal models. Vascular endothelial growth factor (VEGF) is one of the main inducers of ocular neovascularization, and several clinical trials have shown the benefits of neutralizing VEGF in patients with neovascular AMD or diabetic macular edema. In this review, we summarize several preclinical and early-stage clinical trials with intraocular gene therapies, which have the potential to reduce or eliminate the repeated intravitreal injections that are currently required for the treatment of neovascular AMD.
Collapse
Affiliation(s)
- Peter Pechan
- Gene Therapy, Sanofi-Genzyme R&D Center, Framingham, Massachusetts 01701
| | - Samuel Wadsworth
- Gene Therapy, Sanofi-Genzyme R&D Center, Framingham, Massachusetts 01701
| | - Abraham Scaria
- Gene Therapy, Sanofi-Genzyme R&D Center, Framingham, Massachusetts 01701
| |
Collapse
|
33
|
Askou AL. Development of gene therapy for treatment of age-related macular degeneration. Acta Ophthalmol 2014; 92 Thesis3:1-38. [PMID: 24953666 DOI: 10.1111/aos.12452] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Intraocular neovascular diseases are the leading cause of blindness in the Western world in individuals over the age of 50. Age-related macular degeneration (AMD) is one of these diseases. Exudative AMD, the late-stage form, is characterized by abnormal neovessel development, sprouting from the choroid into the avascular subretinal space, where it can suddenly cause irreversible damage to the vulnerable photoreceptor (PR) cells essential for our high-resolution, central vision. The molecular basis of AMD is not well understood, but several growth factors have been implicated including vascular endothelial growth factor (VEGF), and the advent of anti-VEGF therapy has markedly changed the outcome of treatment. However, common to all current therapies for exudative AMD are the complications of repeated monthly intravitreal injections, which must be continued throughout one's lifetime to maintain visual benefits. Additionally, some patients do not benefit from established treatments. Strategies providing long-term suppression of inappropriate ocular angiogenesis are therefore needed, and gene therapy offers a potential powerful technique. This study aimed to develop a strategy based on RNA interference (RNAi) for the sustained attenuation of VEGF. We designed a panel of anti-VEGF short hairpin RNAs (shRNA), and based on the most potent shRNAs, microRNA (miRNA)-mimicked hairpins were developed. We demonstrated an additive VEGF silencing effect when we combined the miRNAs in a tricistronic miRNA cluster. To meet the requirements for development of medical treatments for AMD with long-term effects, the shRNA/miRNA is expressed from vectors based on adeno-associated virus (AAV) or lentivirus (LV). Both vector systems have been found superior in terms of transduction efficiency and persistence in gene expression in retinal cells. The capacity of AAV-encoded RNAi effector molecules to silence endogenous VEGF gene expression was evaluated in mouse models, including the model of laser-induced choroidal neovascularization (CNV), and we found that subretinal administration of self-complementary (sc)-AAV2/8 encoding anti-VEGF shRNAs can impair vessel formation. In parallel, a significant reduction of endogenous VEGF was demonstrated following injection of scAAV2/8 vectors expressing multiple anti-VEGF miRNAs into murine hind limb muscles. Furthermore, in an ongoing project we have designed versatile, multigenic LV vectors with combined expression of multiple miRNAs and proteins, including pigment epithelium-derived factor (PEDF), a multifunctional, secreted protein that has anti-angiogenic and neurotrophic functions. Co-expression of miRNAs and proteins from a single viral vector increases safety by minimizing the viral load necessary to obtain a therapeutic effect and thereby reduces the risk of insertional mutagenesis as well as the immune response against viral proteins. Our results show co-expression of functional anti-VEGF-miRNAs and PEDF in cell studies, and in vivo studies reveal an efficient retinal pigment epithelium (RPE)-specific gene expression following the incorporation of the vitelliform macular dystrophy 2 (VMD2) promoter, demonstrating the potential applicability of our multigenic LV vectors in ocular anti-VEGF gene therapy, including combination therapy for treatment of exudative AMD. In conclusion, these highly promising data clearly demonstrate that viral-encoded RNAi effector molecules can be used for the inhibition of neovascularization and will, in combination with the growing interest of applying DNA- or RNA-based technologies in the clinic, undoubtedly contribute to the development of efficacious long-term gene therapy treatment of intraocular neovascular diseases.
Collapse
|
34
|
Willett K, Bennett J. Immunology of AAV-Mediated Gene Transfer in the Eye. Front Immunol 2013; 4:261. [PMID: 24009613 PMCID: PMC3757345 DOI: 10.3389/fimmu.2013.00261] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 08/16/2013] [Indexed: 12/20/2022] Open
Abstract
The eye has been at the forefront of translational gene therapy largely owing to suitable disease targets, anatomic accessibility, and well-studied immunologic privilege. These advantages have fostered research culminating in several clinical trials and adeno-associated virus (AAV) has emerged as the vector of choice for many ocular therapies. Pre-clinical and clinical investigations have assessed the humoral and cellular immune responses to a variety of naturally occurring and engineered AAV serotypes as well as their delivered transgenes and these data have been correlated to potential clinical sequelae. Encouragingly, AAV appears safe and effective with clinical follow-up surpassing 5 years in some studies. As disease targets continue to expand for AAV in the eye, thorough and deliberate assessment of immunologic safety is critical. With careful study, the development of these technologies should concurrently inform the biology of the ocular immune response.
Collapse
Affiliation(s)
- Keirnan Willett
- Department of Ophthalmology, Scheie Eye Institute, F.M. Kirby Center for Molecular Ophthalmology, University of Pennsylvania , Philadelphia, PA , USA
| | | |
Collapse
|
35
|
Boye SE, Boye SL, Lewin AS, Hauswirth WW. A comprehensive review of retinal gene therapy. Mol Ther 2013; 21:509-19. [PMID: 23358189 DOI: 10.1038/mt.2012.280] [Citation(s) in RCA: 204] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Blindness, although not life threatening, is a debilitating disorder for which few, if any treatments exist. Ocular gene therapies have the potential to profoundly improve the quality of life in patients with inherited retinal disease. As such, tremendous focus has been given to develop such therapies. Several factors make the eye an ideal organ for gene-replacement therapy including its accessibility, immune privilege, small size, compartmentalization, and the existence of a contralateral control. This review will provide a comprehensive summary of (i) existing gene therapy clinical trials for several genetic forms of blindness and (ii) preclinical efficacy and safety studies in a variety of animal models of retinal disease which demonstrate strong potential for clinical application. To be as comprehensive as possible, we include additional proof of concept studies using gene replacement, neurotrophic/neuroprotective, optogenetic, antiangiogenic, or antioxidative stress strategies as well as a description of the current challenges and future directions in the ocular gene therapy field to this review as a supplement.
Collapse
Affiliation(s)
- Shannon E Boye
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA.
| | | | | | | |
Collapse
|
36
|
Miller JW. Age-related macular degeneration revisited--piecing the puzzle: the LXIX Edward Jackson memorial lecture. Am J Ophthalmol 2013; 155:1-35.e13. [PMID: 23245386 DOI: 10.1016/j.ajo.2012.10.018] [Citation(s) in RCA: 182] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 10/19/2012] [Accepted: 10/23/2012] [Indexed: 12/11/2022]
Abstract
PURPOSE To present the current understanding of age-related macular degeneration (AMD) pathogenesis, based on clinical evidence, epidemiologic data, histopathologic examination, and genetic data; to provide an update on current and emerging therapies; and to propose an integrated model of the pathogenesis of AMD. DESIGN Review of published clinical and experimental studies. METHODS Analysis and synthesis of clinical and experimental data. RESULTS We are closer to a complete understanding of the pathogenesis of AMD, having progressed from clinical observations to epidemiologic observations and clinical pathologic correlation. More recently, modern genetic and genomic studies have facilitated the exploration of molecular pathways. It seems that AMD is a complex disease that results from the interaction of genetic susceptibility with aging and environmental factors. Disease progression also seems to be driven by a combination of genetic and environmental factors. CONCLUSIONS Therapies based on pathophysiologic features have changed the paradigm for treating neovascular AMD. With improved understanding of the underlying genetic susceptibility, we can identify targets to halt early disease and to prevent progression and vision loss.
Collapse
|