1
|
Ning K, Zhang X, Feng Z, Hao S, Kuz CA, Cheng F, Park SY, McFarlin S, Engelhardt JF, Yan Z, Qiu J. Inhibition of DNA-dependent protein kinase catalytic subunit boosts rAAV transduction of polarized human airway epithelium. Mol Ther Methods Clin Dev 2023; 31:101115. [PMID: 37841417 PMCID: PMC10568418 DOI: 10.1016/j.omtm.2023.101115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023]
Abstract
Adeno-associated virus 2.5T (AAV2.5T) was selected from the directed evolution of AAV capsid library in human airway epithelia. This study found that recombinant AAV2.5T (rAAV2.5T) transduction of well-differentiated primary human airway epithelia induced a DNA damage response (DDR) characterized by the phosphorylation of replication protein A32 (RPA32), histone variant H2AX (H2A histone family member X), and all three phosphatidylinositol 3-kinase-related kinases: ataxia telangiectasia mutated kinase, ataxia telangiectasia and Rad3-related kinase (ATR), and DNA-dependent protein kinase catalytic subunit (DNA-PKcs). While suppressing the expression of ATR by a specific pharmacological inhibitor or targeted gene silencing inhibited rAAV2.5T transduction, DNA-PKcs inhibition or targeted gene silencing significantly increased rAAV2.5T transgene expression. Notably, DNA-PKcs inhibitors worked as a "booster" to further increase rAAV2.5T transgene expression after treatment with doxorubicin and did not compromise epithelial integrity. Thus, our study provides evidence that DDR is associated with rAAV transduction in well-differentiated human airway epithelia, and DNA-PKcs inhibition has the potential to boost rAAV transduction. These findings highlight that the application of DDR inhibition-associated pharmacological interventions has the potential to increase rAAV transduction and thus to reduce the required vector dose.
Collapse
Affiliation(s)
- Kang Ning
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xiujuan Zhang
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Zehua Feng
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Siyuan Hao
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Cagla Aksu Kuz
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Fang Cheng
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Soo Yuen Park
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Shane McFarlin
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Ziying Yan
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
2
|
MacLaren RE, Fischer MD, Gow JA, Lam BL, Sankila EMK, Girach A, Panda S, Yoon D, Zhao G, Pennesi ME. Subretinal timrepigene emparvovec in adult men with choroideremia: a randomized phase 3 trial. Nat Med 2023; 29:2464-2472. [PMID: 37814062 PMCID: PMC10579095 DOI: 10.1038/s41591-023-02520-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 07/28/2023] [Indexed: 10/11/2023]
Abstract
Choroideremia is a rare, X-linked retinal degeneration resulting in progressive vision loss. A randomized, masked, phase 3 clinical trial evaluated the safety and efficacy over 12 months of follow-up in adult males with choroideremia randomized to receive a high-dose (1.0 × 1011 vector genomes (vg); n = 69) or low-dose (1.0 × 1010 vg; n = 34) subretinal injection of the AAV2-vector-based gene therapy timrepigene emparvovec versus non-treated control (n = 66). Most treatment-emergent adverse events were mild or moderate. The trial did not meet its primary endpoint of best-corrected visual acuity (BCVA) improvement. In the primary endpoint analysis, three of 65 participants (5%) in the high-dose group, one of 34 (3%) participants in the low-dose group and zero of 62 (0%) participants in the control group had ≥15-letter Early Treatment Diabetic Retinopathy Study (ETDRS) improvement from baseline BCVA at 12 months (high dose, P = 0.245 versus control; low dose, P = 0.354 versus control). As the primary endpoint was not met, key secondary endpoints were not tested for significance. In a key secondary endpoint, nine of 65 (14%), six of 35 (18%) and one of 62 (2%) participants in the high-dose, low-dose and control groups, respectively, experienced ≥10-letter ETDRS improvement from baseline BCVA at 12 months. Potential opportunities to enhance future gene therapy studies for choroideremia include optimization of entry criteria (more preserved retinal area), surgical techniques and clinical endpoints. EudraCT registration: 2015-003958-41 .
Collapse
Affiliation(s)
- Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
- Oxford University Hospitals NIHR Biomedical Research Centre, Oxford, UK.
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - M Dominik Fischer
- University Eye Hospital Tübingen, Center for Ophthalmology, Tübingen, Germany
| | | | - Byron L Lam
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Aniz Girach
- Formerly of Nightstar Therapeutics, London, UK
| | | | | | | | - Mark E Pennesi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
3
|
Nikonov S, Aravand P, Lyubarsky A, Nikonov R, Luo AJ, Wei Z, Maguire AM, Phelps NT, Shpylchak I, Willett K, Aleman TS, Huckfeldt RM, Ramachandran PS, Bennett J. Restoration of Vision and Retinal Responses After Adeno-Associated Virus-Mediated Optogenetic Therapy in Blind Dogs. Transl Vis Sci Technol 2022; 11:24. [PMID: 35604672 PMCID: PMC9145127 DOI: 10.1167/tvst.11.5.24] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 11/17/2021] [Indexed: 12/02/2022] Open
Abstract
Purpose Optogenetic gene therapy to render remaining retinal cells light-sensitive in end-stage retinal degeneration is a promising strategy for treatment of individuals blind because of a variety of different inherited retinal degenerations. The clinical trials currently in progress focus on delivery of optogenetic genes to ganglion cells. Delivery of optogenetic molecules to cells in the outer neural retina is predicted to be even more advantageous because it harnesses more of the retinal circuitry. However, this approach has not yet been tested in large animal models. For this reason, we evaluated the safety and efficacy of optogenetic therapy targeting remaining diseased cone photoreceptors in the Rcd1 dog model of retinitis pigmentosa. Methods Imaging and measures of retinal function and functional vision were carried out, as well as terminal studies evaluating multi-electrode array recordings and histology. Results Animals remained healthy and active throughout the study and showed improved retinal and visual function as assessed by electroretinography and visual-evoked potentials, improved navigational vision, and improved function of cone photoreceptors and the downstream retinal circuitry. Conclusions The findings demonstrate that an optogenetic approach targeting the outer retina in a blind large animal model can partially restore vision. Translational Relevance This work has translational relevance because the approach could potentially be extrapolated to treat humans who are totally blind because of retinal degenerative disease.
Collapse
Affiliation(s)
- Sergei Nikonov
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Puya Aravand
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Arkady Lyubarsky
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Roman Nikonov
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Angela J. Luo
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Zhangyong Wei
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Albert M. Maguire
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Nicholas T. Phelps
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Ivan Shpylchak
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Keirnan Willett
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Tomas S. Aleman
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Rachel M. Huckfeldt
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Pavitra S. Ramachandran
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Jean Bennett
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology Scheie Eye Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
4
|
Shoshina II, Hovis JK, Felisberti FM, Santos NA, Adreeva A, Butler PD, Fernandes TP. Visual processing and BDNF levels in first-episode schizophrenia. Psychiatry Res 2021; 305:114200. [PMID: 34653830 DOI: 10.1016/j.psychres.2021.114200] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/13/2022]
Abstract
Previous studies have shown that patients diagnosed with schizophrenia (SCZ) have deficits in early visual processing, namely contrast processing. The brain-derived neurotropic factor (BDNF) is an important measure to investigate neuroplasticity in some visual functions like visual perception. In this study, we investigated the relationship between visual processing and BDNF levels in first-episode SCZ patients. Thirty-nine healthy controls and 43 first-episode SCZ patients were enrolled. Contrast sensitivity measurements were conducted using low, mid- and high spatial frequencies. First-episode SCZ patients had higher contrast sensitivity than healthy controls for all frequencies, except for the middle spatial frequency. Negative correlations were found between BDNF, contrast sensitivity and clinical variables, mostly for middle and high spatial frequencies among females. Our results provide support for (i) the association of SCZ with alterations of magno- and parvocellular pathway functioning and (ii) decreased BDNF levels in first-episode SCZ patients. This study highlights the importance of using biomarkers along with other measures to investigate visual processing in SCZ and other disorders.
Collapse
Affiliation(s)
- Irina I Shoshina
- Pavlov Institute of Physiology, Russian Academy of Science, Department of Visual Physiology, Saint-Petersburg, Russian Federation; St. Petersburg State University, Department of Liberal Arts and Sciences, Saint-Petersburg, Russian Federation
| | - Jeffery K Hovis
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| | - Fatima M Felisberti
- Department of Psychology, Kingston University London, London, United Kingdom
| | - Natanael A Santos
- Department of Psychology, Federal University of Paraiba, Joao Pessoa, Brazil; Perception, Neuroscience and Behaviour Laboratory, Federal University of Paraiba, Brazil
| | - Anna Adreeva
- St. Petersburg State University, Department of Liberal Arts and Sciences, Saint-Petersburg, Russian Federation
| | - Pamela D Butler
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States; Department of Psychiatry, New York University School of Medicine, NY, USA
| | - Thiago P Fernandes
- Department of Psychology, Federal University of Paraiba, Joao Pessoa, Brazil; Perception, Neuroscience and Behaviour Laboratory, Federal University of Paraiba, Brazil.
| |
Collapse
|
5
|
Liu D, Deng Q, Lei X, Lu W, Zhao Q, Shen Y. Overexpression of BMP4 protects retinal ganglion cells in a mouse model of experimental glaucoma. Exp Eye Res 2021; 210:108728. [PMID: 34390734 DOI: 10.1016/j.exer.2021.108728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 08/02/2021] [Accepted: 08/10/2021] [Indexed: 01/10/2023]
Abstract
PURPOSE Activation of bone morphogenetic protein (BMP) 4 signaling promotes the survival of retinal ganglion cell (RGC) after acute injury. Chordin-like 1 (CHRDL1) is an endogenous BMP antagonist. In this study, we researched whether CHRDL1 was involved in BMP4 signaling and regulation of RGC degeneration in a mouse model of glaucoma. METHODS Magnetic microbeads were intracameral injected to induce experimental glaucoma in a mouse model. A recombinant adeno-associated virus (rAAV) system was designed for overexpression of BMP4 or CHRDL1 in mouse retina. Immunohistochemistry and hematoxylin-eosin (HE) stains were performed to identify changes in retinal morphology. Electroretinogram (ERG) recordings were used to assess changes in visual function. RESULTS The mRNA expression levels of Bmp4 and its downstream BMPRIa, small mothers against decapentaplegic 1 (Smad1), were significantly upregulated in retinas with glaucoma. RGC survival was significantly enhanced in the beads + AAV-BMP4 group and significantly reduced in the beads + AAV-CHRDL1 group, compared with the beads + AAV-EGFP group. Similar results were observed in retinal explant culture in vitro. Consistent with these findings, the photopic negative response (PhNR)responses in ERG, which indicate RGC function, were restored in mice overexpressing BMP4, whereas a-wave and b-wave responses were not. Activation of CHRLD1 inhibited Smad1/5/8 phosphorylation and exacerbated RGC damage. The expression of Glial fibrillary acidic protein (GFAP) was decreased significantly in beads + AAV-BMP4 group. CONCLUSIONS BMP4 promoted RGC survival and visual function in an experimental glaucoma model. Activation of CHRDL1 exaggerated RGC degeneration by inhibiting the BMP4/Smad1/5/8 pathway. The mechanism of BMP4/Smad1/5/8 pathway may be related to the inhibition of glial cell activation. Our studies suggested that BMP4 and CHRLD1 might serve as therapeutic targets in glaucoma.
Collapse
Affiliation(s)
- Dongmei Liu
- Eye Institute, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei, China
| | - Qinqin Deng
- Eye Institute, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei, China
| | - Xinlan Lei
- Eye Institute, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei, China
| | - Wei Lu
- Eye Institute, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei, China
| | - Qingqing Zhao
- Eye Institute, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei, China
| | - Yin Shen
- Eye Institute, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei, China; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
6
|
Fuller-Carter PI, Basiri H, Harvey AR, Carvalho LS. Focused Update on AAV-Based Gene Therapy Clinical Trials for Inherited Retinal Degeneration. BioDrugs 2021; 34:763-781. [PMID: 33136237 DOI: 10.1007/s40259-020-00453-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inherited retinal diseases (IRDs) comprise a clinically and genetically heterogeneous group of disorders that can ultimately result in photoreceptor dysfunction/death and vision loss. With over 270 genes known to be involved in IRDs, translation of treatment strategies into clinical applications has been historically difficult. However, in recent years there have been significant advances in basic research findings as well as translational studies, culminating in an increasing number of clinical trials with the ultimate goal of reducing vision loss and associated morbidities. The recent approval of Luxturna® (voretigene neparvovec-rzyl) for Leber congenital amaurosis type 2 (LCA2) prompts a review of the current clinical trials for IRDs, with a particular focus on the importance of adeno-associated virus (AAV)-based gene therapies. The present article reviews the current state of AAV use in gene therapy clinical trials for IRDs, with a brief background on AAV and the reasons behind its dominance in ocular gene therapy. It will also discuss pre-clinical progress in AAV-based therapies aimed at treating other ocular conditions that can have hereditable links, and what alternative technologies are progressing in the same therapeutic space.
Collapse
Affiliation(s)
- Paula I Fuller-Carter
- Centre for Ophthalmology and Visual Sciences (Incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia
| | - Hamed Basiri
- Centre for Ophthalmology and Visual Sciences (Incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia
| | - Alan R Harvey
- School of Human Sciences, The University of Western Australia, Crawley, WA, Australia.,Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
| | - Livia S Carvalho
- Centre for Ophthalmology and Visual Sciences (Incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia.
| |
Collapse
|
7
|
Maguire AM, Bennett J, Aleman EM, Leroy BP, Aleman TS. Clinical Perspective: Treating RPE65-Associated Retinal Dystrophy. Mol Ther 2021; 29:442-463. [PMID: 33278565 PMCID: PMC7854308 DOI: 10.1016/j.ymthe.2020.11.029] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/16/2020] [Accepted: 11/25/2020] [Indexed: 12/25/2022] Open
Abstract
Until recently, there was no approved treatment for a retinal degenerative disease. Subretinal injection of a recombinant adeno-associated virus (AAV) delivering the normal copy of the human RPE65 cDNA led to reversal of blindness first in animal models and then in humans. This led to the first US Food and Drug Administration (FDA)-approved gene therapy product for a genetic disease, voretigene neparvovec-rzyl (Luxturna). Luxturna was then approved by the European Medicines Association and is now available in the US through Spark Therapeutics and worldwide through Novartis. Not only has treatment with Luxturna changed the lives of people previously destined to live a life of blindness, but it has fueled interest in developing additional gene therapy reagents targeting numerous other genetic forms of inherited retinal disease. This review describes many of the considerations for administration of Luxturna and describes how lessons from experience with Luxturna could lead to additional gene-based treatments of blindness.
Collapse
Affiliation(s)
- Albert M Maguire
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Jean Bennett
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Elena M Aleman
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bart P Leroy
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Ophthalmology and Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Tomas S Aleman
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA.
| |
Collapse
|
8
|
Fujita K, Nishiguchi KM, Sato K, Nakagawa Y, Nakazawa T. In vivo imaging of the light response in mouse retinal ganglion cells based on a neuronal activity-dependent promoter. Biochem Biophys Res Commun 2020; 521:471-477. [PMID: 31672273 DOI: 10.1016/j.bbrc.2019.10.155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/22/2019] [Indexed: 11/19/2022]
Abstract
Diseases of the retinal ganglion cells (RGCs) are an important cause of blindness, yet the light response of individual RGCs is difficult to assess in vivo, particularly in mammals, due to a lack of effective methods. We report a simple in vivo platform for imaging the light response of mouse RGCs based on a fluorescent reporter-tagged enhanced synaptic activity-responsive element (E-SARE) that mediates neuronal activity-dependent gene transcription. When E-SARE-driven d2Venus, packaged into an AAV vector, was injected intravitreally, light-responsive retinal neurons expressing d2Venus were visible at single-cell resolution using confocal ophthalmoscopy. Immunohistological assessment identified the majority of these cells as RGCs. In a murine model of RGC injury, the number of d2Venus-positive cells was correlated with the amplitude of light-induced responses and with visual acuity, measured electrophysiologically at the visual cortex, indicating that the vector can be used as a tool to assess visual function in RGCs. The platform described herein allows a simple in vivo assessment of RGC function, which should help basic research into the mechanisms of RGC death and the development of treatments for diseases involving the RGCs.
Collapse
Affiliation(s)
- Kosuke Fujita
- Department of Ophthalmic Imaging and Information Analytics, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan; Department of Ophthalmology, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan
| | - Koji M Nishiguchi
- Department of Advanced Ophthalmic Medicine, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan
| | - Kota Sato
- Collaborative Program of Ophthalmic Drug Discovery, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan; Department of Ophthalmology, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan
| | - Yurika Nakagawa
- Department of Ophthalmology, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan
| | - Toru Nakazawa
- Department of Ophthalmic Imaging and Information Analytics, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan; Department of Advanced Ophthalmic Medicine, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan; Collaborative Program of Ophthalmic Drug Discovery, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan; Department of Ophthalmology, Graduate School of Medicine, Tohoku University, Sendai, 980-8574, Japan.
| |
Collapse
|
9
|
Weed L, Ammar MJ, Zhou S, Wei Z, Serrano LW, Sun J, Lee V, Maguire AM, Bennett J, Aleman TS. Safety of Same-Eye Subretinal Sequential Readministration of AAV2-hRPE65v2 in Non-human Primates. Mol Ther Methods Clin Dev 2019; 15:133-148. [PMID: 31660416 PMCID: PMC6807311 DOI: 10.1016/j.omtm.2019.08.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 08/26/2019] [Indexed: 01/28/2023]
Abstract
We have demonstrated safe and effective subretinal readministration of recombinant adeno-associated virus serotype (rAAV) to the contralateral eye in large animals and humans even in the setting of preexisting neutralizing antibodies (NAbs). Readministration of AAV to the same retina may be desirable in order to treat additional areas of the retina not targeted initially or to boost transgene expression levels at a later time point. To better understand the immune and structural consequences of subretinal rAAV readministration to the same eye, we administered bilateral subretinal injections of rAAV2-hRPE65v2 to three unaffected non-human primates (NHPs) and repeated the injections in those same eyes 2 months later. Ophthalmic exams and retinal imaging were performed after the first and second injections. Peripheral blood monocytes, serum, and intraocular fluids were collected at baseline and post-injection time points to characterize the cellular and humoral immune responses. Histopathologic and immunohistochemical studies were carried out on the treated retinas. Ipsilateral readministration of AAV2-hRPE65v2 in NHPs did not threaten the ocular or systemic health through the time span of the study. The repeat injections were immunologically and structurally well tolerated, even in the setting of preexisting serum NAbs. Localized structural abnormalities confined to the outer retina and retinal pigmented epithelium (RPE) after readministration of the treatment do not differ from those observed after single or contralateral administration of an AAV carrying a non-therapeutic transgene in NHPs and were not observed in a patient treated with the nearly identical, FDA-approved, AAV2-hRPE65v2 vector (voretigene neparvovec-rzyl), suggesting NHP-specific abnormalities.
Collapse
Affiliation(s)
- Lindsey Weed
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael J. Ammar
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shangzhen Zhou
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zhangyong Wei
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Leona W. Serrano
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Junwei Sun
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Vivian Lee
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Albert M. Maguire
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jean Bennett
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Tomas S. Aleman
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| |
Collapse
|
10
|
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: 28] [Impact Index Per Article: 4.7] [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
|
11
|
McGill TJ, Stoddard J, Renner LM, Messaoudi I, Bharti K, Mitalipov S, Lauer A, Wilson DJ, Neuringer M. Allogeneic iPSC-Derived RPE Cell Graft Failure Following Transplantation Into the Subretinal Space in Nonhuman Primates. Invest Ophthalmol Vis Sci 2018; 59:1374-1383. [PMID: 29625461 PMCID: PMC5846443 DOI: 10.1167/iovs.17-22467] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose To characterize the intraocular immune response following transplantation of iPS-derived allogeneic RPE cells into the subretinal space of non–immune-suppressed rhesus macaques. Methods GFP-labeled allogeneic iPS-derived RPE cells were transplanted into the subretinal space of one eye (n = 6), and into the contralateral eye 1 day to 4 weeks later, using a two-stage transretinal and transscleral approach. Retinas were examined pre- and post-surgery by color fundus photography, fundus autofluorescence, and optical coherence tomography (OCT) imaging. Animals were euthanized between 2 hours and 7 weeks following transplantation. T-cell (CD3), B-cell (CD20), and microglial (Iba1) responses were assessed immunohistochemically. Results Cells were delivered into the subretinal space in all eyes without leakage into the vitreous. Transplanted RPE cells were clearly visible at 4 days after surgery but were no longer detectable by 3 weeks. In localized areas within the bleb containing transplanted cells, T- and B-cell infiltrates and microglia were observed in the subretinal space and underlying choroid. A T-cell response predominated at 4 days, but converted to a B-cell response at 3 weeks. By 7 weeks, few infiltrates or microglia remained. Host RPE and choroid were disrupted in the immediate vicinity of the graft, with fibrosis in the subretinal space. Conclusions Engraftment of allogeneic RPE cells failed following transplantation into the subretinal space of rhesus macaques, likely due to rejection by the immune system. These data underscore the need for autologous cell sources and/or confirmation of adequate immune suppression to ensure survival of transplanted RPE cells.
Collapse
Affiliation(s)
- Trevor J McGill
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States.,Department of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| | - Jonathan Stoddard
- Department of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| | - Lauren M Renner
- Department of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California, United States
| | - Kapil Bharti
- Unit on Ocular and Stem Cell Translational Research, National Eye Institute/National Institutes of Health, Bethesda, Maryland, United States
| | - Shoukhrat Mitalipov
- Department of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| | - Andreas Lauer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - David J Wilson
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Martha Neuringer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States.,Department of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| |
Collapse
|
12
|
Day TP, Byrne LC, Flannery JG, Schaffer DV. Screening for Neutralizing Antibodies Against Natural and Engineered AAV Capsids in Nonhuman Primate Retinas. Methods Mol Biol 2018; 1715:239-249. [PMID: 29188518 DOI: 10.1007/978-1-4939-7522-8_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Adeno-associated virus (AAV) has shown promise as a therapeutic gene delivery vector for inherited retinal degenerations in both preclinical disease models and human clinical trials. The retinas of nonhuman primates (NHPs) share many anatomical similarities to humans and are an important model for evaluating AAV gene delivery. Recent evidence has shown that preexisting immunity in the form of neutralizing antibodies (NABs) in NHPs strongly correlates with weak or lack of AAV transduction in the retina when administered intravitreally, work with translational implications. This necessitates prescreening of NHPs before intravitreal delivery of AAV. In this chapter, we describe a method for screening NHP serum for preexisting NABs.
Collapse
Affiliation(s)
- Timothy P Day
- The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Leah C Byrne
- The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John G Flannery
- The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Vision Science Graduate Group, School of Optometry, University of California, Berkeley, CA, USA
| | - David V Schaffer
- The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA.
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
- Department of Bioengineering, University of California, Berkeley, CA, USA.
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA.
| |
Collapse
|
13
|
Sullivan JA, Stanek LM, Lukason MJ, Bu J, Osmond SR, Barry EA, O'Riordan CR, Shihabuddin LS, Cheng SH, Scaria A. Rationally designed AAV2 and AAVrh8R capsids provide improved transduction in the retina and brain. Gene Ther 2018; 25:205-219. [PMID: 29785047 DOI: 10.1038/s41434-018-0017-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 03/25/2018] [Accepted: 03/27/2018] [Indexed: 01/18/2023]
Abstract
The successful application of adeno-associated virus (AAV) gene delivery vectors as a therapeutic paradigm will require efficient gene delivery to the appropriate cells in affected organs. In this study, we utilized a rational design approach to introduce modifications to the AAV2 and AAVrh8R capsids and the resulting variants were evaluated for transduction activity in the retina and brain. The modifications disrupted either capsid/receptor binding or altered capsid surface charge. Specifically, we mutated AAV2 amino acids R585A and R588A, which are required for binding to its receptor, heparan sulfate proteoglycans, to generate a variant referred to as AAV2-HBKO. In contrast to parental AAV2, the AAV2-HBKO vector displayed low-transduction activity following intravitreal delivery to the mouse eye; however, following its subretinal delivery, AAV2-HBKO resulted in significantly greater photoreceptor transduction. Intrastriatal delivery of AAV2-HBKO to mice facilitated widespread striatal and cortical expression, in contrast to the restricted transduction pattern of the parental AAV2 vector. Furthermore, we found that altering the surface charge on the AAVrh8R capsid by modifying the number of arginine residues on the capsid surface had a profound impact on subretinal transduction. The data further validate the potential of capsid engineering to improve AAV gene therapy vectors for clinical applications.
Collapse
Affiliation(s)
| | - Lisa M Stanek
- Sanofi, 49 New York Avenue, Framingham, MA, 01701-9322, USA
| | | | - Jie Bu
- Sanofi, 49 New York Avenue, Framingham, MA, 01701-9322, USA
| | | | | | | | | | - Seng H Cheng
- Sanofi, 49 New York Avenue, Framingham, MA, 01701-9322, USA
| | - Abraham Scaria
- Sanofi, 49 New York Avenue, Framingham, MA, 01701-9322, USA
| |
Collapse
|
14
|
Petit L, Ma S, Cheng SY, Gao G, Punzo C. Rod Outer Segment Development Influences AAV-Mediated Photoreceptor Transduction After Subretinal Injection. Hum Gene Ther 2018; 28:464-481. [PMID: 28510482 PMCID: PMC5488363 DOI: 10.1089/hum.2017.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Vectors based on the adeno-associated virus (AAV) are currently the preferred tools for delivering genes to photoreceptors (PR) in small and large animals. AAVs have been applied successfully in various models of PR dystrophies. However, unknown barriers still limit AAV's efficient application in several forms of severe PR degenerations due to insufficient transgene expression and/or treated cells at the time of injection. Optimizations of PR gene therapy strategies will likely benefit from the identification of the cellular factors that influence PR transduction. Interestingly, recent studies have shown that the AAV transduction profile of PRs differs significantly between neonatal and adult mouse retinas after subretinal injection. This phenomenon may provide clues to identify host factors that influence the efficiency of AAV-mediated PR transduction. This study demonstrates that rod outer segments are critical modulators of efficient AAV-mediated rod transduction. During retinal development, rod transduction correlated temporally and spatially with the differentiation order of PRs when vectors were introduced subretinally but not when introduced intravitreally. All subretinally injected vectors had an initial preference to transduce cones in the absence of formed rod outer segments and then displayed a preference for rods as the cells matured, independently of the expression cassette or AAV serotype. Consistent with this observation, altered development of rod outer segments was associated with a strong reduction of rod transduction and an increase in the percentage of transduced cones by 2- to 2.8-fold. A similar increase of cone transduction was observed in the adult retinal degeneration 1 (rd1) retina compared to wild-type mice. These results suggest that the loss of rod outer segments in diseased retinas could markedly affect gene transfer efficiency of AAV vectors by limiting the ability of AAVs to infect dying rods efficiently. This information could be exploited for the development of more efficient AAV-based PR gene delivery procedures.
Collapse
Affiliation(s)
- Lolita Petit
- 1 Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Shan Ma
- 1 Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Shun-Yun Cheng
- 1 Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Guangping Gao
- 3 Department of Microbiology and Physiological Systems and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Claudio Punzo
- 1 Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Neurobiology, University of Massachusetts Medical School , Worcester, Massachusetts
| |
Collapse
|
15
|
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
|
16
|
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: 5.9] [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
|
17
|
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
|
18
|
Evaluation of tolerance to lentiviral LV-RPE65 gene therapy vector after subretinal delivery in non-human primates. Transl Res 2017; 188:40-57.e4. [PMID: 28754419 DOI: 10.1016/j.trsl.2017.06.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 04/30/2017] [Accepted: 06/30/2017] [Indexed: 12/17/2022]
Abstract
Several approaches have been developed for gene therapy in RPE65-related Leber congenital amaurosis. To date, strategies that have reached the clinical stages rely on adeno-associated viral vectors and two of them documented limited long-term effect. We have developed a lentiviral-based strategy of RPE65 gene transfer that efficiently restored protein expression and cone function in RPE65-deficient mice. In this study, we evaluated the ocular and systemic tolerances of this lentiviral-based therapy (LV-RPE65) on healthy nonhuman primates (NHPs), without adjuvant systemic anti-inflammatory prophylaxis. For the first time, we describe the early kinetics of retinal detachment at 2, 4, and 7 days after subretinal injection using multimodal imaging in 5 NHPs. We revealed prolonged reattachment times in LV-RPE65-injected eyes compared to vehicle-injected eyes. Low- (n = 2) and high-dose (n = 2) LV-RPE65-injected eyes presented a reduction of the outer nuclear and photoreceptor outer segment layer thickness in the macula, that was more pronounced than in vehicle-injected eyes (n = 4). All LV-RPE65-injected eyes showed an initial perivascular reaction that resolved spontaneously within 14 days. Despite foveal structural changes, full-field electroretinography indicated that the overall retinal function was preserved over time and immunohistochemistry identified no difference in glial, microglial, or leucocyte ocular activation between low-dose, high-dose, and vehicle-injected eyes. Moreover, LV-RPE65-injected animals did not show signs of vector shedding or extraocular targeting, confirming the safe ocular restriction of the vector. Our results evidence a limited ocular tolerance to LV-RPE65 after subretinal injection without adjuvant anti-inflammatory prophylaxis, with complications linked to this route of administration necessitating to block this transient inflammatory event.
Collapse
|
19
|
Tshilenge KT, Ameline B, Weber M, Mendes-Madeira A, Nedellec S, Biget M, Provost N, Libeau L, Blouin V, Deschamps JY, Le Meur G, Colle MA, Moullier P, Pichard V, Rolling F. Vitrectomy Before Intravitreal Injection of AAV2/2 Vector Promotes Efficient Transduction of Retinal Ganglion Cells in Dogs and Nonhuman Primates. Hum Gene Ther Methods 2017; 27:122-34. [PMID: 27229628 DOI: 10.1089/hgtb.2016.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Recombinant adeno-associated virus (AAV) has emerged as a promising vector for retinal gene delivery to restore visual function in certain forms of inherited retinal dystrophies. Several studies in rodent models have shown that intravitreal injection of the AAV2/2 vector is the optimal route for efficient retinal ganglion cell (RGC) transduction. However, translation of these findings to larger species, including humans, is complicated by anatomical differences in the eye, a key difference being the comparatively smaller volume of the vitreous chamber in rodents. Here, we address the role of the vitreous body as a potential barrier to AAV2/2 diffusion and transduction in the RGCs of dogs and macaques, two of the most relevant preclinical models. We intravitreally administered the AAV2/2 vector carrying the CMV-eGFP reporter cassette in dog and macaque eyes, either directly into the vitreous chamber or after complete vitrectomy, a surgical procedure that removes the vitreous body. Our findings suggest that the vitreous body appears to trap the injected vector, thus impairing the diffusion and transduction of AAV2/2 to inner retinal neurons. We show that vitrectomy before intravitreal vector injection is an effective means of overcoming this physical barrier, improving the transduction of RGCs in dog and macaque retinas. These findings support the use of vitrectomy in clinical trials of intravitreal gene transfer techniques targeting inner retinal neurons.
Collapse
Affiliation(s)
| | - Baptiste Ameline
- 1 Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Michel Weber
- 2 CHU de Nantes, Service d'Ophtalmologie, Nantes, France
| | | | - Steven Nedellec
- 3 Cellular and Tissular Imaging Core Facility of Nantes University, SFR Santé Francois Bonamy INSERM UMS016/CNRS UMS3556, Nantes, France
| | - Marine Biget
- 1 Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Nathalie Provost
- 1 Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Lyse Libeau
- 1 Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Véronique Blouin
- 1 Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Jack-Yves Deschamps
- 4 Emergency and Critical Care Unit, ONIRIS, Nantes-Atlantic College of Veterinary Medicine Food Science and Engineering, Nantes, France
| | | | - Marie-Anne Colle
- 5 UMR 703 PAnTher INRA/ONIRIS, Nantes-Atlantic College of Veterinary Medicine Food Science and Engineering, Nantes, France
| | - Philippe Moullier
- 1 Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France.,6 Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida , Gainesville, Florida
| | - Virginie Pichard
- 1 Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| | - Fabienne Rolling
- 1 Atlantic Gene Therapies, INSERM UMR 1089, Université de Nantes, CHU de Nantes, Nantes, France
| |
Collapse
|
20
|
Correction of Monogenic and Common Retinal Disorders with Gene Therapy. Genes (Basel) 2017; 8:genes8020053. [PMID: 28134823 PMCID: PMC5333042 DOI: 10.3390/genes8020053] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/19/2017] [Indexed: 11/16/2022] Open
Abstract
The past decade has seen major advances in gene-based therapies, many of which show promise for translation to human disease. At the forefront of research in this field is ocular disease, as the eye lends itself to gene-based interventions due to its accessibility, relatively immune-privileged status, and ability to be non-invasively monitored. A landmark study in 2001 demonstrating successful gene therapy in a large-animal model for Leber congenital amaurosis set the stage for translation of these strategies from the bench to the bedside. Multiple clinical trials have since initiated for various retinal diseases, and further improvements in gene therapy techniques have engendered optimism for alleviating inherited blinding disorders. This article provides an overview of gene-based strategies for retinal disease, current clinical trials that engage these strategies, and the latest techniques in genome engineering, which could serve as the next frontline of therapeutic interventions.
Collapse
|
21
|
Becirovic E, Böhm S, Nguyen ONP, Riedmayr LM, Hammelmann V, Schön C, Butz ES, Wahl-Schott C, Biel M, Michalakis S. AAV Vectors for FRET-Based Analysis of Protein-Protein Interactions in Photoreceptor Outer Segments. Front Neurosci 2016; 10:356. [PMID: 27516733 PMCID: PMC4963399 DOI: 10.3389/fnins.2016.00356] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/14/2016] [Indexed: 11/18/2022] Open
Abstract
Fluorescence resonance energy transfer (FRET) is a powerful method for the detection and quantification of stationary and dynamic protein-protein interactions. Technical limitations have hampered systematic in vivo FRET experiments to study protein-protein interactions in their native environment. Here, we describe a rapid and robust protocol that combines adeno-associated virus (AAV) vector-mediated in vivo delivery of genetically encoded FRET partners with ex vivo FRET measurements. The method was established on acutely isolated outer segments of murine rod and cone photoreceptors and relies on the high co-transduction efficiency of retinal photoreceptors by co-delivered AAV vectors. The procedure can be used for the systematic analysis of protein-protein interactions of wild type or mutant outer segment proteins in their native environment. Conclusively, our protocol can help to characterize the physiological and pathophysiological relevance of photoreceptor specific proteins and, in principle, should also be transferable to other cell types.
Collapse
Affiliation(s)
- Elvir Becirovic
- Department of Pharmacy - Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität MünchenMunich, Germany; Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität MünchenMunich, Germany
| | - Sybille Böhm
- Department of Pharmacy - Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität MünchenMunich, Germany; Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität MünchenMunich, Germany
| | - Ong N P Nguyen
- Department of Pharmacy - Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität MünchenMunich, Germany; Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität MünchenMunich, Germany
| | - Lisa M Riedmayr
- Department of Pharmacy - Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität MünchenMunich, Germany; Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität MünchenMunich, Germany
| | - Verena Hammelmann
- Department of Pharmacy - Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität MünchenMunich, Germany; Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität MünchenMunich, Germany
| | - Christian Schön
- Department of Pharmacy - Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität MünchenMunich, Germany; Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität MünchenMunich, Germany
| | - Elisabeth S Butz
- Department of Pharmacy - Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität MünchenMunich, Germany; Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität MünchenMunich, Germany
| | - Christian Wahl-Schott
- Department of Pharmacy - Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität MünchenMunich, Germany; Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität MünchenMunich, Germany
| | - Martin Biel
- Department of Pharmacy - Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität MünchenMunich, Germany; Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität MünchenMunich, Germany
| | - Stylianos Michalakis
- Department of Pharmacy - Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität MünchenMunich, Germany; Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität MünchenMunich, Germany
| |
Collapse
|
22
|
Pensado A, Diaz-Corrales FJ, De la Cerda B, Valdés-Sánchez L, Del Boz AA, Rodriguez-Martinez D, García-Delgado AB, Seijo B, Bhattacharya SS, Sanchez A. Span poly-L-arginine nanoparticles are efficient non-viral vectors for PRPF31 gene delivery: An approach of gene therapy to treat retinitis pigmentosa. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:2251-2260. [PMID: 27381066 DOI: 10.1016/j.nano.2016.06.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/31/2016] [Accepted: 06/16/2016] [Indexed: 12/21/2022]
Abstract
Retinitis pigmentosa (RP) is the most common cause of inherited blindness in adults. Mutations in the PRPF31 gene produce autosomal dominant RP (adRP). To date there are no effective treatments for this disease. The purpose of this study was to design an efficient non-viral vector for human PRPF31 gene delivery as an approach to treat this form of adRP. Span based nanoparticles were developed to mediate gene transfer in the subretinal space of a mouse model of adRP carrying a point mutation (A216P) in the Prpf31 gene. Funduscopic examination, electroretinogram, optomotor test and optical coherence tomography were conducted to further in vivo evaluate the safety and efficacy of the nanosystems developed. Span-polyarginine (SP-PA) nanoparticles were able to efficiently transfect the GFP and PRPF31 plasmid in mice retinas. Statistically significant improvement in visual acuity and retinal thickness were found in Prpf31A216P/+ mice treated with the SP-PA-PRPF31 nanomedicine.
Collapse
Affiliation(s)
- Andrea Pensado
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidade de Santiago de Compostela, Campus Vida, Santiago de Compostela, Spain
| | - Francisco J Diaz-Corrales
- Department of Cell Therapy and Regenerative Medicine, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Seville, Spain
| | - Berta De la Cerda
- Department of Cell Therapy and Regenerative Medicine, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Seville, Spain
| | - Lourdes Valdés-Sánchez
- Department of Cell Therapy and Regenerative Medicine, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Seville, Spain
| | - Ana Aramburu Del Boz
- Department of Cell Therapy and Regenerative Medicine, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Seville, Spain
| | - Daniel Rodriguez-Martinez
- Department of Cell Therapy and Regenerative Medicine, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Seville, Spain
| | - Ana B García-Delgado
- Department of Cell Therapy and Regenerative Medicine, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Seville, Spain
| | - Begoña Seijo
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidade de Santiago de Compostela, Campus Vida, Santiago de Compostela, Spain; Molecular Image Group, Health Research Institute-University Clinical Hospital of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Shomi S Bhattacharya
- Department of Cell Therapy and Regenerative Medicine, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Seville, Spain
| | - Alejandro Sanchez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidade de Santiago de Compostela, Campus Vida, Santiago de Compostela, Spain; Molecular Image Group, Health Research Institute-University Clinical Hospital of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.
| |
Collapse
|
23
|
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: 1.6] [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
|
24
|
Characterization of the Adeno-Associated Virus 1 and 6 Sialic Acid Binding Site. J Virol 2016; 90:5219-5230. [PMID: 26962225 DOI: 10.1128/jvi.00161-16] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 03/04/2016] [Indexed: 12/29/2022] Open
Abstract
UNLABELLED The adeno-associated viruses (AAVs), which are being developed as gene delivery vectors, display differential cell surface glycan binding and subsequent tissue tropisms. For AAV serotype 1 (AAV1), the first viral vector approved as a gene therapy treatment, and its closely related AAV6, sialic acid (SIA) serves as their primary cellular surface receptor. Toward characterizing the SIA binding site(s), the structure of the AAV1-SIA complex was determined by X-ray crystallography to 3.0 Å. Density consistent with SIA was observed in a pocket located at the base of capsid protrusions surrounding icosahedral 3-fold axes. Site-directed mutagenesis substitution of the amino acids forming this pocket with structurally equivalent residues from AAV2, a heparan sulfate binding serotype, followed by cell binding and transduction assays, further mapped the critical residues conferring SIA binding to AAV1 and AAV6. For both viruses five of the six binding pocket residues mutated (N447S, V473D, N500E, T502S, and W503A) abolished SIA binding, whereas S472R increased binding. All six mutations abolished or decreased transduction by at least 50% in AAV1. Surprisingly, the T502S substitution did not affect transduction efficiency of wild-type AAV6. Furthermore, three of the AAV1 SIA binding site mutants-S472R, V473D, and N500E-escaped recognition by the anti-AAV1 capsid antibody ADK1a. These observations demonstrate that common key capsid surface residues dictate both virus binding and entry processes, as well as antigenic reactivity. This study identifies an important functional capsid surface "hot spot" dictating receptor attachment, transduction efficiency, and antigenicity which could prove useful for vector engineering. IMPORTANCE The adeno-associated virus (AAV) vector gene delivery system has shown promise in several clinical trials and an AAV1-based vector has been approved as the first gene therapy treatment. However, limitations still exist with respect to transduction efficiency and the detrimental effects of preexisting host antibodies. This study aimed to identify key capsid regions which can be engineered to overcome these limitations. A sialic glycan receptor recognition pocket was identified in AAV1 and its closely related AAV6, using X-ray crystallography. The site was confirmed by mutagenesis followed by cell binding and transduction assays. Significantly, residues controlling gene expression efficiency, as well as antibody escape variants, were also identified. This study thus provides, at the amino acid level, information for rational structural engineering of AAV vectors with improved therapeutic efficacy.
Collapse
|
25
|
Sondhi D, Crystal RG, Kaminsky SM. Gene Therapy for Inborn Errors of Metabolism: Batten Disease. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
26
|
In vivo cellular imaging of various stress/response pathways using AAV following axonal injury in mice. Sci Rep 2015; 5:18141. [PMID: 26670005 PMCID: PMC4680972 DOI: 10.1038/srep18141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/10/2015] [Indexed: 11/30/2022] Open
Abstract
Glaucoma, a leading cause of blindness worldwide, is instigated by various factors, including axonal injury, which eventually leads to a progressive loss of retinal ganglion cells (RGCs). To study various pathways reportedly involved in the pathogenesis of RGC death caused by axonal injury, seven pathways were investigated. Pathway-specific fluorescent protein-coded reporters were each packaged into an adeno-associated virus (AAV). After producing axonal injury in the eye, injected with AAV to induce RGC death, the temporal activity of each stress-related pathway was monitored in vivo through the detection of fluorescent RGCs using confocal ophthalmoscopy. We identified the activation of ATF6 and MCP-1 pathways involved in endoplasmic reticulum stress and macrophage recruitment, respectively, as early markers of RGC stress that precede neuronal death. Conversely, inflammatory responses probed by NF-κB and cell-death-related pathway p53 were most prominent in the later phases, when RGC death was already ongoing. AAV-mediated delivery of stress/response reporters followed by in vivo cellular imaging is a powerful strategy to characterize the temporal aspects of complex molecular pathways involved in retinal diseases. The identification of promoter elements that are activated before the death of RGCs enables the development of pre-emptive gene therapy, exclusively targeting the early phases of diseased cells.
Collapse
|
27
|
Abstract
Choroideremia is a complex and rare disease that is frequently misdiagnosed due to its similar appearance to classic retinitis pigmentosa. Recent advances in genetic testing have identified specific genetic mutations in many retinal dystrophies, and the identification of the mutation of the CHM gene on the X chromosome 25 years ago has paved the way for gene replacement therapy with the first human trials now underway. This article reviews the epidemiological and pathological features of choroideremia and new prospects in imaging to monitor disease progression, as well as potential treatment approaches for choroideremia.
Collapse
Affiliation(s)
- Martin S Zinkernagel
- Department of Ophthalmology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland ; Department of Clinical Research, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, University of Oxford and Oxford Eye Hospital, Oxford University NHS Trust NIHR Biomedical Research Centre, Oxford, UK ; Moorfields Eye Hospital NIHR Biomedical Research Centre, London, UK
| |
Collapse
|
28
|
Dimopoulos IS, Chan S, MacLaren RE, MacDonald IM. Pathogenic mechanisms and the prospect of gene therapy for choroideremia. Expert Opin Orphan Drugs 2015; 3:787-798. [PMID: 26251765 PMCID: PMC4522943 DOI: 10.1517/21678707.2015.1046434] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Choroideremia is a rare, X-linked disorder recognized by its specific ocular phenotype as a progressive degenerative retinopathy resulting in blindness. New therapeutic approaches, primarily based on genetic mechanisms, have emerged that aim to prevent the progressive vision loss. AREAS COVERED This article will review the research that has progressed incrementally over the past two decades from mapping to gene discovery, uncovering the presumed mechanisms triggering the retinopathy to preclinical testing of potential therapies. EXPERT OPINION While still in an evaluative phase, the introduction of gene replacement as a potential therapy has been greeted with great enthusiasm by patients, advocacy groups and the medical community.
Collapse
Affiliation(s)
- Ioannis S Dimopoulos
- University of Alberta, Department of Ophthalmology and Visual Sciences, Edmonton, Alberta, Canada
| | - Stephanie Chan
- University of Alberta, Department of Ophthalmology and Visual Sciences, Edmonton, Alberta, Canada
| | - Robert E MacLaren
- Oxford Eye Hospital and Nuffield Laboratory of Ophthalmology, John Radcliffe Hospital, Oxford, UK
- Moorfields Eye Hospital Foundation Trust, NIHR Ophthalmology Biomedical Research Centre, London, UK
| | - Ian M MacDonald
- University of Alberta, Department of Ophthalmology and Visual Sciences, Edmonton, Alberta, Canada
| |
Collapse
|
29
|
Khabou H, Dalkara D. [Developments in gene delivery vectors for ocular gene therapy]. Med Sci (Paris) 2015; 31:529-37. [PMID: 26059304 DOI: 10.1051/medsci/20153105015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Gene therapy is quickly becoming a reality applicable in the clinic for inherited retinal diseases. Its remarkable success in safety and efficacy, in clinical trials for Leber's congenital amaurosis (LCA) type II generated significant interest and opened up possibilities for a new era of retinal gene therapies. Success in these clinical trials was mainly due to the favorable characteristics of the retina as a target organ. The eye offers several advantages as it is readily accessible and has some degree of immune privilege making it suitable for application of viral vectors. The viral vectors most frequently used for retinal gene delivery are lentivirus, adenovirus and adeno-associated virus (AAV). Here we will discuss the use of these viral vectors in retinal gene delivery with a strong focus on favorable properties of AAV. Thanks to its small size, AAV diffuses well in the inter-neural matrix making it suitable for applications in neural retina. Building on this initial clinical success with LCA II, we have now many opportunities to extend this proof-of-concept to other retinal diseases using AAV as a vector. This article will discuss what are some of the most imminent cellular targets for such therapies and the AAV toolkit that has been built to target these cells successfully. We will also discuss some of the challenges that we face in translating AAV-based gene therapies to the clinic.
Collapse
Affiliation(s)
- Hanen Khabou
- Inserm UMR S968, Institut de la vision, 17, rue Moreau, 75012 Paris, France - Sorbonne universités, UPMC université Paris 6, UMR S968, 75012 Paris, France - CNRS, UMR 7210, 75012 Paris, France
| | - Deniz Dalkara
- Inserm UMR S968, Institut de la vision, 17, rue Moreau, 75012 Paris, France - Sorbonne universités, UPMC université Paris 6, UMR S968, 75012 Paris, France - CNRS, UMR 7210, 75012 Paris, France
| |
Collapse
|
30
|
Bennett J. My career path for developing gene therapy for blinding diseases: the importance of mentors, collaborators, and opportunities. Hum Gene Ther 2015; 25:663-70. [PMID: 25136912 DOI: 10.1089/hum.2014.2529] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Jean Bennett
- Department of Ophthalmology and Center for Advanced Retinal and Ophthalmic Therapeutics, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA 19104
| |
Collapse
|
31
|
Wilkinson GS, Breden F, Mank JE, Ritchie MG, Higginson AD, Radwan J, Jaquiery J, Salzburger W, Arriero E, Barribeau SM, Phillips PC, Renn SCP, Rowe L. The locus of sexual selection: moving sexual selection studies into the post-genomics era. J Evol Biol 2015; 28:739-55. [PMID: 25789690 DOI: 10.1111/jeb.12621] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/04/2015] [Accepted: 03/06/2015] [Indexed: 02/07/2023]
Abstract
Sexual selection drives fundamental evolutionary processes such as trait elaboration and speciation. Despite this importance, there are surprisingly few examples of genes unequivocally responsible for variation in sexually selected phenotypes. This lack of information inhibits our ability to predict phenotypic change due to universal behaviours, such as fighting over mates and mate choice. Here, we discuss reasons for this apparent gap and provide recommendations for how it can be overcome by adopting contemporary genomic methods, exploiting underutilized taxa that may be ideal for detecting the effects of sexual selection and adopting appropriate experimental paradigms. Identifying genes that determine variation in sexually selected traits has the potential to improve theoretical models and reveal whether the genetic changes underlying phenotypic novelty utilize common or unique molecular mechanisms. Such a genomic approach to sexual selection will help answer questions in the evolution of sexually selected phenotypes that were first asked by Darwin and can furthermore serve as a model for the application of genomics in all areas of evolutionary biology.
Collapse
Affiliation(s)
- G S Wilkinson
- Department of Biology, University of Maryland, College Park, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Treatment of ocular disorders by gene therapy. Eur J Pharm Biopharm 2014; 95:331-42. [PMID: 25536112 DOI: 10.1016/j.ejpb.2014.12.022] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/08/2014] [Accepted: 12/15/2014] [Indexed: 12/27/2022]
Abstract
Gene therapy to treat ocular disorders is still starting, and current therapies are primarily experimental, with most human clinical trials still in research state, although beginning to show encouraging results. Currently 33 clinical trials have been approved, are in progress, or have been completed. The most promising results have been obtained in clinical trials of ocular gene therapy for Leber Congenital Amaurosis, which have prompted the study of several ocular diseases that are good candidates to be treated with gene therapy: glaucoma, age-related macular degeneration, retinitis pigmentosa, or choroideremia. The success of gene therapy relies on the efficient delivery of the genetic material to target cells, achieving optimum long-term gene expression. Although viral vectors have been widely used, their potential risk associated mainly with immunogenicity and mutagenesis has promoted the design of non-viral vectors. In this review, the main administration routes and the most studied delivery systems, viral and non-viral, for ocular gene therapy are presented. The primary ocular disease candidates to be treated with gene therapy have been also reviewed, including the genetic basis and the most relevant preclinical and clinical studies.
Collapse
|
33
|
Barnard AR, Groppe M, MacLaren RE. Gene therapy for choroideremia using an adeno-associated viral (AAV) vector. Cold Spring Harb Perspect Med 2014; 5:a017293. [PMID: 25359548 DOI: 10.1101/cshperspect.a017293] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Choroideremia is an outer retinal degeneration with a characteristic clinical appearance that was first described in the nineteenth century. The disorder begins with reduction of night vision and gradually progresses to blindness by middle age. The appearance of the fundus in sufferers is recognizable by the characteristic pale color caused by the loss of the outer retina, retinal-pigmented epithelium, and choroidal vessels, leading to exposure of the underlying sclera. Choroideremia shows X-linked recessive inheritance and the choroideremia gene (CHM) was one of the first to be identified by positional cloning in 1990. Subsequent identification and characterization of the CHM gene, which encodes Rab escort protein 1 (REP1), has led to better comprehension of the disease and enabled advances in genetic diagnosis. Despite several decades of work to understand the exact pathogenesis, no established treatments currently exist to stop or even slow the progression of retinal degeneration in choroideremia. Encouragingly, several specific molecular and clinical features make choroideremia an ideal candidate for treatment with gene therapy. This work describes the considerations and challenges in the development of a new clinical trial using adeno-associated virus (AAV) encoding the CHM gene.
Collapse
Affiliation(s)
- Alun R Barnard
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences and Oxford Biomedical Research Centre, University of Oxford, The John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
| | - Markus Groppe
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences and Oxford Biomedical Research Centre, University of Oxford, The John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom Moorfields Eye Hospital and NIHR Biomedical Research Centre for Ophthalmology, London EC1V 2PD, United Kingdom
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences and Oxford Biomedical Research Centre, University of Oxford, The John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom Moorfields Eye Hospital and NIHR Biomedical Research Centre for Ophthalmology, London EC1V 2PD, United Kingdom
| |
Collapse
|
34
|
Morgan JIW, Han G, Klinman E, Maguire WM, Chung DC, Maguire AM, Bennett J. High-resolution adaptive optics retinal imaging of cellular structure in choroideremia. Invest Ophthalmol Vis Sci 2014; 55:6381-97. [PMID: 25190651 DOI: 10.1167/iovs.13-13454] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE We characterized retinal structure in patients and carriers of choroideremia using adaptive optics and other high resolution modalities. METHODS A total of 57 patients and 18 carriers of choroideremia were imaged using adaptive optics scanning light ophthalmoscopy (AOSLO), optical coherence tomography (OCT), autofluorescence (AF), and scanning light ophthalmoscopy (SLO). Cone density was measured in 59 eyes of 34 patients where the full cone mosaic was observed. RESULTS The SLO imaging revealed scalloped edges of RPE atrophy and large choroidal vessels. The AF imaging showed hypo-AF in areas of degeneration, while central AF remained present. OCT images showed outer retinal tubulations and thinned RPE/interdigitation layers. The AOSLO imaging revealed the cone mosaic in central relatively intact retina, and cone density was either reduced or normal at 0.5 mm eccentricity. The border of RPE atrophy showed abrupt loss of the cone mosaic at the same location. The AF imaging in comparison with AOSLO showed RPE health may be compromised before cone degeneration. Other disease features, including visualization of choroidal vessels, hyper-reflective clumps of cones, and unique retinal findings, were tabulated to show the frequency of occurrence and model disease progression. CONCLUSIONS The data support the RPE being one primary site of degeneration in patients with choroideremia. Photoreceptors also may degenerate independently. High resolution imaging, particularly AOSLO in combination with OCT, allows single cell analysis of disease in choroideremia. These modalities promise to be useful in monitoring disease progression, and in documenting the efficacy of gene and cell-based therapies for choroideremia and other diseases as these therapies emerge. (ClinicalTrials.gov number, NCT01866371.).
Collapse
Affiliation(s)
- Jessica I W Morgan
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Grace Han
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Eva Klinman
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - William M Maguire
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Daniel C Chung
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Albert M Maguire
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Jean Bennett
- Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
35
|
Petrs-Silva H, Linden R. Advances in gene therapy technologies to treat retinitis pigmentosa. Clin Ophthalmol 2013; 8:127-36. [PMID: 24391438 PMCID: PMC3878960 DOI: 10.2147/opth.s38041] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Retinitis pigmentosa (RP) is a class of diseases that leads to progressive degeneration of the retina. Experimental approaches to gene therapy for the treatment of inherited retinal dystrophies have advanced in recent years, inclusive of the safe delivery of genes to the human retina. This review is focused on the development of gene therapy for RP using recombinant adenoassociated viral vectors, which show a positive safety record and have so far been successful in several clinical trials for congenital retinal disease. Gene therapy for RP is under development in a variety of animal models, and the results raise expectations of future clinical application. Nonetheless, the translation of such strategies to the bedside requires further understanding of the mutations and mechanisms that cause visual defects, as well as thorough examination of potential adverse effects.
Collapse
Affiliation(s)
- Hilda Petrs-Silva
- Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael Linden
- Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
36
|
Manfredi A, Marrocco E, Puppo A, Cesi G, Sommella A, Della Corte M, Rossi S, Giunti M, Craft CM, Bacci ML, Simonelli F, Surace EM, Auricchio A. Combined rod and cone transduction by adeno-associated virus 2/8. Hum Gene Ther 2013; 24:982-92. [PMID: 24067103 DOI: 10.1089/hum.2013.154] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Gene transfer to both cone and rod photoreceptors (PRs) is essential for gene therapy of inherited retinal degenerations that are caused by mutations in genes expressed in both PR types. Vectors based on the adeno-associated virus (AAV) efficiently transduce PRs of different species. However, these are predominantly rods and little is known about the ability of the AAV to transduce cones in combination with rods. Here we show that AAV2/8 transduces pig cones to levels that are similar to AAV2/9, and the outer nuclear layer (mainly rods) to levels that are on average higher, although not statistically significant, than both AAV2/5 and AAV2/9. We additionally found that the ubiquitous cytomegalovirus (CMV), but not the PR-specific GRK1 promoter, transduced pig cones efficiently, presumably because GRK1 is not expressed in pig cones as observed in mice and humans. Indeed, the GRK1 and CMV promoters transduce a similar percentage of murine cones with the CMV reaching the highest expression levels. Consistent with this, the AAV2/8 vectors with either the CMV or the GRK1 promoter restore cone function in a mouse model of Leber congenital amaurosis type 1 (LCA1), supporting the use of AAV2/8 for gene therapy of LCA1 as well as of other retinal diseases requiring gene transfer to both PR types.
Collapse
Affiliation(s)
- Anna Manfredi
- 1 Telethon Institute of Genetics and Medicine , Naples 80131, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Functional expression of Rab escort protein 1 following AAV2-mediated gene delivery in the retina of choroideremia mice and human cells ex vivo. J Mol Med (Berl) 2013; 91:825-37. [PMID: 23756766 PMCID: PMC3695676 DOI: 10.1007/s00109-013-1006-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 01/23/2013] [Accepted: 01/31/2013] [Indexed: 01/10/2023]
Abstract
Choroideremia (CHM) is an X-linked retinal degeneration of photoreceptors, the retinal pigment epithelium (RPE) and choroid caused by loss of function mutations in the CHM/REP1 gene that encodes Rab escort protein 1. As a slowly progressing monogenic retinal degeneration with a clearly identifiable phenotype and a reliable diagnosis, CHM is an ideal candidate for gene therapy. We developed a serotype 2 adeno-associated viral vector AAV2/2-CBA-REP1, which expresses REP1 under control of CMV-enhanced chicken β-actin promoter (CBA) augmented by a Woodchuck hepatitis virus post-transcriptional regulatory element. We show that the AAV2/2-CBA-REP1 vector provides strong and functional transgene expression in the D17 dog osteosarcoma cell line, CHM patient fibroblasts and CHM mouse RPE cells in vitro and in vivo. The ability to transduce human photoreceptors highly effectively with this expression cassette was confirmed in AAV2/2-CBA-GFP transduced human retinal explants ex vivo. Electroretinogram (ERG) analysis of AAV2/2-CBA-REP1 and AAV2/2-CBA-GFP-injected wild-type mouse eyes did not show toxic effects resulting from REP1 overexpression. Subretinal injections of AAV2/2-CBA-REP1 into CHM mouse retinas led to a significant increase in a- and b-wave of ERG responses in comparison to sham-injected eyes confirming that AAV2/2-CBA-REP1 is a promising vector suitable for choroideremia gene therapy in human clinical trials.
Collapse
|
38
|
McClements ME, MacLaren RE. Gene therapy for retinal disease. Transl Res 2013; 161:241-54. [PMID: 23305707 PMCID: PMC3831157 DOI: 10.1016/j.trsl.2012.12.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 12/12/2012] [Accepted: 12/13/2012] [Indexed: 01/16/2023]
Abstract
Gene therapy strategies for the treatment of inherited retinal diseases have made major advances in recent years. This review focuses on adeno-associated viral (AAV) vector approaches to treat retinal degeneration and, thus, prevent or delay the onset of blindness. Data from human clinical trials of gene therapy for retinal disease show encouraging signs of safety and efficacy from AAV vectors. Recent progress in enhancing cell-specific targeting and transduction efficiency of the various retinal layers plus the use of AAV-delivered growth factors to augment the therapeutic effect and limit cell death suggest even greater success in future human trials is possible.
Collapse
Affiliation(s)
- Michelle E McClements
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | |
Collapse
|
39
|
Hsu PH, Wei KC, Huang CY, Wen CJ, Yen TC, Liu CL, Lin YT, Chen JC, Shen CR, Liu HL. Noninvasive and targeted gene delivery into the brain using microbubble-facilitated focused ultrasound. PLoS One 2013; 8:e57682. [PMID: 23460893 PMCID: PMC3584045 DOI: 10.1371/journal.pone.0057682] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 01/28/2013] [Indexed: 11/19/2022] Open
Abstract
Recombinant adeno-associated viral (rAAV) vectors are potentially powerful tools for gene therapy of CNS diseases, but their penetration into brain parenchyma is severely limited by the blood-brain barrier (BBB) and current delivery relies on invasive stereotactic injection. Here we evaluate the local, targeted delivery of rAAV vectors into the brains of mice by noninvasive, reversible, microbubble-facilitated focused ultrasound (FUS), resulting in BBB opening that can be monitored and controlled by magnetic resonance imaging (MRI). Using this method, we found that IV-administered AAV2-GFP (green fluorescence protein) with a low viral vector titer (1×10(9) vg/g) can successfully penetrate the BBB-opened brain regions to express GFP. We show that MRI monitoring of BBB-opening could serve as an indicator of the scale and distribution of AAV transduction. Transduction peaked at 3 weeks and neurons and astrocytes were affected. This novel, noninvasive delivery approach could significantly broaden the application of AAV-viral-vector-based genes for treatment of CNS diseases.
Collapse
Affiliation(s)
- Po-Hung Hsu
- Department of Electrical Engineering, Chang-Gung University, Taoyuan, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Chang-Gung University and Memorial Hospital, Taoyuan, Taiwan
| | - Chiung-Yin Huang
- Department of Neurosurgery, Chang-Gung University and Memorial Hospital, Taoyuan, Taiwan
| | - Chih-Jen Wen
- Molecular Imaging Center, Chang-Gung University and Memorial Hospital, Taoyuan, Taiwan
| | - Tzu-Chen Yen
- Molecular Imaging Center, Chang-Gung University and Memorial Hospital, Taoyuan, Taiwan
- Department of Nuclear Medicine, Chang-Gung University and Memorial Hospital, Taoyuan, Taiwan
| | - Chao-Lin Liu
- Department of Chemical Engineering, Min-Chi University of Technology, Taipei, Taiwan
| | - Ya-Tin Lin
- Graduate Institute of Biomedical Sciences, Chang-Gung University, Taoyuan, Taiwan
| | - Jin-Chung Chen
- Graduate Institute of Biomedical Sciences, Chang-Gung University, Taoyuan, Taiwan
| | - Chia-Rui Shen
- Department of Medical Biotechnology and Laboratory Science, Medical College, Chang-Gung University, Taoyuan, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, Chang-Gung University, Taoyuan, Taiwan
| |
Collapse
|
40
|
Vandenberghe LH, Bell P, Maguire AM, Xiao R, Hopkins TB, Grant R, Bennett J, Wilson JM. AAV9 targets cone photoreceptors in the nonhuman primate retina. PLoS One 2013; 8:e53463. [PMID: 23382846 PMCID: PMC3559681 DOI: 10.1371/journal.pone.0053463] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 11/22/2012] [Indexed: 12/31/2022] Open
Abstract
Transduction of retinal pigment epithelial cells with an adeno-associated viral vector (AAV) based on serotype 2 has partially corrected retinal blindness in Leber congenital amaurosis type 2. However, many applications of gene therapy for retinal blindness rely on the efficient transduction of rod and cone photoreceptor which is difficult to achieve with first generation vector technology. To address this translational need, we evaluated rod and cone photoreceptor targeting of 4 novel AAV capsids (AAV7, AAV9, rh.64R1 and rh.8R) versus AAV2 and AAV8 in a foveated retina. Eyes of 20 nonhuman primates were injected subretinally in the proximity of the fovea. While numerous vectors efficiently transduced rods, only AAV9 targeted cones both centrally and peripherally efficiently at low doses, likely due to the abundance of galactosylated glycans, the primary receptor for AAV9, on cone photoreceptors. We conclude AAV9 is an ideal candidate for strategies that require restoration of cone photoreceptor function.
Collapse
Affiliation(s)
- Luk H. Vandenberghe
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (JMW); (LHV)
| | - Peter Bell
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Albert M. Maguire
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ru Xiao
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Tim B. Hopkins
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Rebecca Grant
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jean Bennett
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - James M. Wilson
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (JMW); (LHV)
| |
Collapse
|
41
|
Lipinski DM, Thake M, MacLaren RE. Clinical applications of retinal gene therapy. Prog Retin Eye Res 2013; 32:22-47. [DOI: 10.1016/j.preteyeres.2012.09.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 09/04/2012] [Accepted: 09/04/2012] [Indexed: 02/08/2023]
|
42
|
Bennett J, Maguire AM. Gene Therapy for Retinal Disease. Retina 2013. [DOI: 10.1016/b978-1-4557-0737-9.00034-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
43
|
Kay CN, Ryals RC, Aslanidi GV, Min SH, Ruan Q, Sun J, Dyka FM, Kasuga D, Ayala AE, Van Vliet K, Agbandje-McKenna M, Hauswirth WW, Boye SL, Boye SE. Targeting photoreceptors via intravitreal delivery using novel, capsid-mutated AAV vectors. PLoS One 2013; 8:e62097. [PMID: 23637972 PMCID: PMC3637363 DOI: 10.1371/journal.pone.0062097] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 03/16/2013] [Indexed: 12/23/2022] Open
Abstract
Development of viral vectors capable of transducing photoreceptors by less invasive methods than subretinal injection would provide a major advancement in retinal gene therapy. We sought to develop novel AAV vectors optimized for photoreceptor transduction following intravitreal delivery and to develop methodology for quantifying this transduction in vivo. Surface exposed tyrosine (Y) and threonine (T) residues on the capsids of AAV2, AAV5 and AAV8 were changed to phenylalanine (F) and valine (V), respectively. Transduction efficiencies of self-complimentary, capsid-mutant and unmodified AAV vectors containing the smCBA promoter and mCherry cDNA were initially scored in vitro using a cone photoreceptor cell line. Capsid mutants exhibiting the highest transduction efficiencies relative to unmodified vectors were then injected intravitreally into transgenic mice constitutively expressing a Rhodopsin-GFP fusion protein in rod photoreceptors (Rho-GFP mice). Photoreceptor transduction was quantified by fluorescent activated cell sorting (FACS) by counting cells positive for both GFP and mCherry. To explore the utility of the capsid mutants, standard, (non-self-complementary) AAV vectors containing the human rhodopsin kinase promoter (hGRK1) were made. Vectors were intravitreally injected in wildtype mice to assess whether efficient expression exclusive to photoreceptors was achievable. To restrict off-target expression in cells of the inner and middle retina, subsequent vectors incorporated multiple target sequences for miR181, an miRNA endogenously expressed in the inner and middle retina. Results showed that AAV2 containing four Y to F mutations combined with a single T to V mutation (quadY-F+T-V) transduced photoreceptors most efficiently. Robust photoreceptor expression was mediated by AAV2(quadY-F+T-V) -hGRK1-GFP. Observed off-target expression was reduced by incorporating target sequence for a miRNA highly expressed in inner/middle retina, miR181c. Thus we have identified a novel AAV vector capable of transducing photoreceptors following intravitreal delivery to mouse. Furthermore, we describe a robust methodology for quantifying photoreceptor transduction from intravitreally delivered AAV vectors.
Collapse
Affiliation(s)
- Christine N. Kay
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Renee C. Ryals
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - George V. Aslanidi
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Seok Hong Min
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Qing Ruan
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Jingfen Sun
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Frank M. Dyka
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Daniel Kasuga
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Andrea E. Ayala
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Kim Van Vliet
- Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, Florida, United States of America
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, Florida, United States of America
| | - William W. Hauswirth
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Sanford L. Boye
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Shannon E. Boye
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
44
|
Yan H, Cui J, Wang Y, Yu Y. Comparison of the effects between intravitreal and periocular injections of adenoviral vectored pigment epithelium-derived factor on suppressing choroidal neovascularization in rats. Ophthalmic Res 2012; 49:81-9. [PMID: 23257710 DOI: 10.1159/000342979] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 08/03/2012] [Indexed: 11/19/2022]
Abstract
PURPOSE To compare the effects between intravitreal and periocular injections of adenoviral vectored pigment epithelium-derived factor (AdPEDF) on suppressing established choroidal neovascularization (CNV) in rats. METHODS Sixty-eight female BN rats (136 eyes) aged 6-8 weeks were involved in this study. The CNV animal models were created with laser photocoagulation. After CNV, 16 rats underwent intravitreal injection with AdPEDF 1 µl (group A), 16 rats received intravitreal injection with control vector (AdNull) 1 µl (group B), 16 rats had periocular injection with AdPEDF 1 µl (group C), and 16 rats had periocular injection with AdNull 1 µl (group D). The effects between intravitreal and periocular AdPEDF injections on suppressing established CNV in rats were compared and evaluated by fundus fluorescein angiography (FFA), maximal thickness of CNV, histopathology and transferase-mediated uridine nick end labeling (TUNEL) staining 3, 7, 14 and 28 days after treatment. RESULTS (1) There were no significant changes in leakage in groups A and C 3 days after injection compared with that before injection seen by FFA (p > 0.05). The leakages in groups A and C decreased significantly 7 days after injection compared with that before injection (p < 0.05). (2) There was no significant difference in the incidence of CNV between groups A and B, as well as groups C and D 3 days after injection (p > 0.05). The incidence of CNV decreased significantly in group A compared with that in group B 7, 14 and 28 days after injection (p < 0.01). CNV retained fibrovascular proliferation in groups B and D 7 days after injection. (3) The maximal thickness of CNV in groups A and C diminished significantly compared with that in the control group after injection, and it still diminished with time (p < 0.05). There was no significant difference in maximal thickness of CNV between 14 and 28 days after injection of AdPEDF (p > 0.05). The maximal thickness of CNV in group A was larger than that in group C 3 days after injection (p < 0.05), yet it was smaller than that in group C 14 and 28 days after injection (p < 0.05). (4) Histopathologically, a great deal of CNV was shown 3 days after injection of AdPEDF or AdNull. CNV decreased significantly with lumen diminution 14 days after injection of AdPEDF. (5) TUNEL cells appeared in groups A and C 7, 14 and 28 days after injection, and there were no TUNEL cells in groups B and D. TUNEL cells were not seen in choroidal inherent vascular endothelial cells in all groups. CONCLUSION Compared with the effect of periocular AdPEDF injection on suppressing established CNV in rats, the effect of intravitreal injection started slowly, but lasted longer. The effect appeared on day 7, reached the peak on day 14 and remained stable on day 28 after the treatment.
Collapse
Affiliation(s)
- Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China.
| | | | | | | |
Collapse
|
45
|
Abstract
Gene therapy holds promise for the treatment of many inherited and acquired diseases of the eye. Successful ocular gene therapy interventions depend on efficient gene transfer to targeted cells with minimal toxicity. A major challenge is to overcome both intracellular and extracellular barriers associated with ocular gene delivery. Numerous viral and nonviral vectors were explored to improve transfection efficiency. Among nonviral delivery systems, polymeric vectors have gained significant attention in recent years owing to their nontoxic and non-immunogenic nature. Polyplexes or nanoparticles can be prepared by interaction of cationic polymers with DNA, which facilitate cellular uptake, endolysosomal escape and nuclear entry through active mechanisms. Chemical modification of these polymers allows for the generation of flexible delivery vectors with desirable properties. In this article several synthetic and natural polymeric systems utilized for ocular gene delivery are discussed.
Collapse
|
46
|
Bennett J, Ashtari M, Wellman J, Marshall KA, Cyckowski LL, Chung DC, McCague S, Pierce EA, Chen Y, Bennicelli JL, Zhu X, Ying GS, Sun J, Wright JF, Auricchio A, Simonelli F, Shindler KS, Mingozzi F, High KA, Maguire AM. AAV2 gene therapy readministration in three adults with congenital blindness. Sci Transl Med 2012; 4:120ra15. [PMID: 22323828 PMCID: PMC4169122 DOI: 10.1126/scitranslmed.3002865] [Citation(s) in RCA: 298] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Demonstration of safe and stable reversal of blindness after a single unilateral subretinal injection of a recombinant adeno-associated virus (AAV) carrying the RPE65 gene (AAV2-hRPE65v2) prompted us to determine whether it was possible to obtain additional benefit through a second administration of the AAV vector to the contralateral eye. Readministration of vector to the second eye was carried out in three adults with Leber congenital amaurosis due to mutations in the RPE65 gene 1.7 to 3.3 years after they had received their initial subretinal injection of AAV2-hRPE65v2. Results (through 6 months) including evaluations of immune response, retinal and visual function testing, and functional magnetic resonance imaging indicate that readministration is both safe and efficacious after previous exposure to AAV2-hRPE65v2.
Collapse
Affiliation(s)
- Jean Bennett
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, 309 Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA 19104, USA
- Center for Cellular and Molecular Therapeutics at The Children’s Hospital of Philadelphia, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19014, USA
| | - Manzar Ashtari
- Department of Radiology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19014, USA
| | - Jennifer Wellman
- Center for Cellular and Molecular Therapeutics at The Children’s Hospital of Philadelphia, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19014, USA
| | - Kathleen A. Marshall
- Center for Cellular and Molecular Therapeutics at The Children’s Hospital of Philadelphia, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19014, USA
| | - Laura L. Cyckowski
- Department of Radiology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19014, USA
| | - Daniel C. Chung
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, 309 Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA 19104, USA
- Center for Cellular and Molecular Therapeutics at The Children’s Hospital of Philadelphia, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19014, USA
| | - Sarah McCague
- Center for Cellular and Molecular Therapeutics at The Children’s Hospital of Philadelphia, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19014, USA
| | - Eric A. Pierce
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, 309 Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA 19104, USA
- Department of Ophthalmology, Children’s Hospital of Philadelphia, Philadelphia, PA 19014, USA
| | - Yifeng Chen
- Center for Cellular and Molecular Therapeutics at The Children’s Hospital of Philadelphia, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19014, USA
| | - Jeannette L. Bennicelli
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, 309 Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA 19104, USA
| | - Xiaosong Zhu
- Department of Ophthalmology, Children’s Hospital of Philadelphia, Philadelphia, PA 19014, USA
| | - Gui-shuang Ying
- Scheie Eye Institute, Center for Preventive Ophthalmology and Biostatistics, Department of Ophthalmology, University of Pennsylvania, 3535 Market Street, Suite 700, Philadelphia, PA 19104, USA
| | - Junwei Sun
- Center for Cellular and Molecular Therapeutics at The Children’s Hospital of Philadelphia, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19014, USA
| | - J. Fraser Wright
- Center for Cellular and Molecular Therapeutics at The Children’s Hospital of Philadelphia, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19014, USA
| | - Alberto Auricchio
- Telethon Institute of Genetics and Medicine, Via P. Castellino 111, 80131 Naples, Italy
- Medical Genetics, Department of Pediatrics, “Federico II” University, Via S. Pansini 5, 80131 Naples, Italy
| | - Francesca Simonelli
- Telethon Institute of Genetics and Medicine, Via P. Castellino 111, 80131 Naples, Italy
- Department of Ophthalmology, Seconda Università degli Studi di Napoli, Via S. Pansini 5, 80131 Naples, Italy
| | - Kenneth S. Shindler
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, 309 Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA 19104, USA
| | - Federico Mingozzi
- Center for Cellular and Molecular Therapeutics at The Children’s Hospital of Philadelphia, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19014, USA
| | - Katherine A. High
- Center for Cellular and Molecular Therapeutics at The Children’s Hospital of Philadelphia, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19014, USA
- Howard Hughes Medical Institute, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Albert M. Maguire
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, 309 Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA 19104, USA
- Center for Cellular and Molecular Therapeutics at The Children’s Hospital of Philadelphia, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19014, USA
- Department of Ophthalmology, Children’s Hospital of Philadelphia, Philadelphia, PA 19014, USA
| |
Collapse
|
47
|
Abstract
Twelve AAV serotypes have been described so far in human and nonhuman primate (NHP) populations while surprisingly high diversity of AAV sequences is detected in tissue biopsies. The analysis of these novel AAV sequences has indicated a rapid evolution of the viral genome both by accumulation of mutations and recombination. This chapter describes how this rich resource of naturally evolved sequences is used to derive gene transfer vectors with a wide array of activities depending on the nature of the cap gene used in the packaging system. AAV2-based recombinant genomes have been packaged in dozens of different capsid types, resulting in a wide array of "pseudotyped vectors" that constitute a rich resource for the development of gene therapy clinical trials. We describe a polymerase chain reaction-based molecular rescue method for novel AAV isolation that uses primers designed to recognize the highly conserved regions in known AAV isolates and generate amplicons across the hypervariable regions of novel AAV genomes present in the analyzed sample.
Collapse
|
48
|
Abstract
With the recent progress in identifying disease-causing genes in humans and in animal models, there are more and more opportunities for using retinal gene transfer to learn more about retinal physiology and also to develop therapies for blinding disorders. Success in preclinical studies for one form of inherited blindness have led to testing in human clinical trials. This paves the way to consider a number of other retinal diseases as ultimate gene therapy targets in human studies. The information presented here is designed to assist scientists and clinicians to use gene transfer to probe the biology of the retina and/or to move appropriate gene-based treatment studies from the bench to the clinic.
Collapse
Affiliation(s)
- Jean Bennett
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
49
|
Abstract
Retinal gene therapy holds great promise for the treatment of inherited and noninherited blinding diseases such as retinitis pigmentosa and age-related macular degeneration. The most widely used vectors for ocular gene delivery are based on adeno-associated virus (AAV) because it mediates long-term transgene expression in a variety of retinal cell types and elicits minimal immune responses. Inherited retinal diseases are nonlethal and have a wide level of genetic heterogeneity. Many of the genes have now been identified and their function elucidated, providing a major step towards the development of gene-based treatments. Extensive preclinical evaluation of gene transfer strategies in small and large animal models is key to the development of successful gene-based therapies for the retina. These preclinical studies have already allowed the field to reach the point where gene therapy to treat inherited blindness has been brought to clinical trial.In this chapter, we focus on AAV-mediated specific gene therapy for inherited retinal degenerative diseases, describing the disease targets, the preclinical studies in animal models and the recent success of the LCA-RPE65 clinical trials.
Collapse
|
50
|
Goldenberg-Cohen N, Avraham-Lubin BCR, Sadikov T, Goldstein RS, Askenasy N. Primitive stem cells derived from bone marrow express glial and neuronal markers and support revascularization in injured retina exposed to ischemic and mechanical damage. Stem Cells Dev 2011; 21:1488-500. [PMID: 21905921 DOI: 10.1089/scd.2011.0366] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Ischemic or mechanical injury to the optic nerve is an irreversible cause of vision loss, associated with limited regeneration and poor response to neuroprotective agents. The aim of this study was to assess the capacity of adult bone marrow cells to participate in retinal regeneration following the induction of anterior ischemic optic neuropathy (AION) and optic nerve crush (ONC) in a rodent model. The small-sized subset of cells isolated by elutriation and lineage depletion (Fr25lin(-)) was found to be negative for the neuroglial markers nestin and glial fibrillary acidic protein (GFAP). Syngeneic donor cells, identified by genomic marker in sex-mismatched transplants and green fluorescent protein, incorporated into the injured retina (AION and ONC) at a frequency of 0.35%-0.45% after intravenous infusion and 1.8%-2% after intravitreous implantation. Perivascular cells with astrocytic morphology expressing GFAP and vimentin were of the predominant lineage that engrafted after AION injury; 10%-18% of the donor cells incorporated in the retinal ganglion cell layer and expressed NeuN, Thy-1, neurofilament, and beta-tubulin III. The Fr25lin(-) cells displayed an excellent capacity to migrate to sites of tissue disruption and developed coordinated site-specific morphological and phenotypic neural and glial markers. In addition to cellular reconstitution of the injured retinal layers, these cells contributed to endothelial revascularization and apparently supported remodeling by secretion of insulin-like growth factor-1. These results suggest that elutriated autologous adult bone marrow-derived stem cells may serve as an accessible source for cellular reconstitution of the retina following injury.
Collapse
Affiliation(s)
- Nitza Goldenberg-Cohen
- Krieger Eye Research Laboratory, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | | | | | | | | |
Collapse
|