1
|
Haldrup SH, Fabian-Jessing BK, Jakobsen TS, Lindholm AB, Adsersen RL, Aagaard L, Bek T, Askou AL, Corydon TJ. Subretinal AAV delivery of RNAi-therapeutics targeting VEGFA reduces choroidal neovascularization in a large animal model. Mol Ther Methods Clin Dev 2024; 32:101242. [PMID: 38605811 PMCID: PMC11007540 DOI: 10.1016/j.omtm.2024.101242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/21/2024] [Indexed: 04/13/2024]
Abstract
Neovascular age-related macular degeneration (nAMD) is a frequent cause of vision loss among the elderly in the Western world. Current disease management with repeated injections of anti-VEGF agents accumulates the risk for adverse events and constitutes a burden for society and the individual patient. Sustained suppression of VEGF using gene therapy is an attractive alternative, which we explored using adeno-associated virus (AAV)-based delivery of novel RNA interference (RNAi) effectors in a porcine model of choroidal neovascularization (CNV). The potency of VEGFA-targeting, Ago2-dependent short hairpin RNAs placed in pri-microRNA scaffolds (miR-agshRNA) was established in vitro and in vivo in mice. Subsequently, AAV serotype 8 (AAV2.8) vectors encoding VEGFA-targeting or irrelevant miR-agshRNAs under the control of a tissue-specific promotor were delivered to the porcine retina via subretinal injection before CNV induction by laser. Notably, VEGFA-targeting miR-agshRNAs resulted in a significant and sizable reduction of CNV compared with the non-targeting control. We also demonstrated that single-stranded and self-complementary AAV2.8 vectors efficiently transduce porcine retinal pigment epithelium cells but differ in their transduction characteristics and retinal safety. Collectively, our data demonstrated a robust anti-angiogenic effect of VEGFA-targeting miR-aghsRNAs in a large translational animal model, thereby suggesting AAV-based delivery of anti-VEGFA RNAi therapeutics as a valuable tool for the management of nAMD.
Collapse
Affiliation(s)
- Silja Hansen Haldrup
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Bjørn K. Fabian-Jessing
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 167, 8200 Aarhus N, Denmark
| | - Thomas Stax Jakobsen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 167, 8200 Aarhus N, Denmark
| | - Anna Bøgh Lindholm
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Rikke L. Adsersen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Lars Aagaard
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Toke Bek
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 167, 8200 Aarhus N, Denmark
| | - Anne Louise Askou
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 167, 8200 Aarhus N, Denmark
| | - Thomas J. Corydon
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 167, 8200 Aarhus N, Denmark
| |
Collapse
|
2
|
She K, Su J, Wang Q, Liu Y, Zhong X, Jin X, Zhao Q, Xiao J, Li R, Deng H, Lu F, Yang Y, Wei Y. Delivery of nVEGFi using AAV8 for the treatment of neovascular age-related macular degeneration. Mol Ther Methods Clin Dev 2022; 24:210-221. [PMID: 35141350 PMCID: PMC8800040 DOI: 10.1016/j.omtm.2022.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 01/05/2022] [Indexed: 11/01/2022]
Abstract
Inhibition of vascular endothelial growth factor (VEGF) is the standard therapy for neovascular age-related macular degeneration (nAMD). However, anti-VEGF agents used in the clinic require repeated injections, causing adverse effects. Gene therapy could provide sustained anti-VEGF levels after a single injection, thereby drastically decreasing the treatment burden and improving visual outcomes. In this study, we developed a novel VEGF Trap, nVEGFi, containing domains 1 and 2 of VEGFR1 and domain 3 of VEGFR2 fused to the Fc portion of human IgG. The nVEGFi had a higher expression level than aflibercept under the same expression cassettes of adeno-associated virus (AAV)8 in vitro and in vivo. nVEGFi was found to be noninferior to aflibercept in binding and blocking VEGF in vitro. AAV8-mediated expression of nVEGFi was maintained for at least 12 weeks by subretinal delivery in C57BL/6J mice. In a mouse laser-induced choroidal neovascularization (CNV) model, 4 × 108 genome copies of AAV8-nVEGFi exhibited a significantly increased reduction in the CNV area compared with AAV8-aflibercept (78.1% vs. 63.9%, p < 0.05), while causing no structural or functional changes to the retina. In conclusion, this preclinical study showed that subretinal injection of AAV8-nVEGFi was long lasting, well tolerated, and effective for nAMD treatment, supporting future translation to the clinic.
Collapse
Affiliation(s)
- Kaiqin She
- Department of Ophthalmology, West China Hospital, Sichuan University, No.37, Guoxue Xiang, Chengdu, Sichuan 610041, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 1, Ke-yuan Road 4, Gao-peng Street, Chengdu, Sichuan 610041, China
| | - Jing Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 1, Ke-yuan Road 4, Gao-peng Street, Chengdu, Sichuan 610041, China
| | - Qingnan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 1, Ke-yuan Road 4, Gao-peng Street, Chengdu, Sichuan 610041, China
| | - Yi Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 1, Ke-yuan Road 4, Gao-peng Street, Chengdu, Sichuan 610041, China
| | - Xiaomei Zhong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 1, Ke-yuan Road 4, Gao-peng Street, Chengdu, Sichuan 610041, China
| | - Xiu Jin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 1, Ke-yuan Road 4, Gao-peng Street, Chengdu, Sichuan 610041, China
| | - Qinyu Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 1, Ke-yuan Road 4, Gao-peng Street, Chengdu, Sichuan 610041, China
| | - Jianlu Xiao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 1, Ke-yuan Road 4, Gao-peng Street, Chengdu, Sichuan 610041, China
| | - Ruiting Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 1, Ke-yuan Road 4, Gao-peng Street, Chengdu, Sichuan 610041, China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 1, Ke-yuan Road 4, Gao-peng Street, Chengdu, Sichuan 610041, China
| | - Fang Lu
- Department of Ophthalmology, West China Hospital, Sichuan University, No.37, Guoxue Xiang, Chengdu, Sichuan 610041, China
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 1, Ke-yuan Road 4, Gao-peng Street, Chengdu, Sichuan 610041, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 1, Ke-yuan Road 4, Gao-peng Street, Chengdu, Sichuan 610041, China
| |
Collapse
|
3
|
Tan TE, Fenner BJ, Barathi VA, Tun SBB, Wey YS, Tsai ASH, Su X, Lee SY, Cheung CMG, Wong TY, Mehta JS, Teo KYC. Gene-Based Therapeutics for Acquired Retinal Disease: Opportunities and Progress. Front Genet 2021; 12:795010. [PMID: 34950193 PMCID: PMC8688942 DOI: 10.3389/fgene.2021.795010] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/16/2021] [Indexed: 12/22/2022] Open
Abstract
Acquired retinal diseases such as age-related macular degeneration and diabetic retinopathy rank among the leading causes of blindness and visual loss worldwide. Effective treatments for these conditions are available, but often have a high treatment burden, and poor compliance can lead to disappointing real-world outcomes. Development of new treatment strategies that provide more durable treatment effects could help to address some of these unmet needs. Gene-based therapeutics, pioneered for the treatment of monogenic inherited retinal disease, are being actively investigated as new treatments for acquired retinal disease. There are significant advantages to the application of gene-based therapeutics in acquired retinal disease, including the presence of established therapeutic targets and common pathophysiologic pathways between diseases, the lack of genotype-specificity required, and the larger potential treatment population per therapy. Different gene-based therapeutic strategies have been attempted, including gene augmentation therapy to induce in vivo expression of therapeutic molecules, and gene editing to knock down genes encoding specific mediators in disease pathways. We highlight the opportunities and unmet clinical needs in acquired retinal disease, review the progress made thus far with current therapeutic strategies and surgical delivery techniques, and discuss limitations and future directions in the field.
Collapse
Affiliation(s)
- Tien-En Tan
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Beau James Fenner
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Veluchamy Amutha Barathi
- Singapore Eye Research Institute, Singapore, Singapore.,Duke-National University of Singapore Medical School, Singapore, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sai Bo Bo Tun
- Singapore Eye Research Institute, Singapore, Singapore
| | - Yeo Sia Wey
- Singapore Eye Research Institute, Singapore, Singapore
| | - Andrew Shih Hsiang Tsai
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Xinyi Su
- Singapore Eye Research Institute, Singapore, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore.,Department of Ophthalmology, National University Hospital, Singapore, Singapore
| | - Shu Yen Lee
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Chui Ming Gemmy Cheung
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Tien Yin Wong
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Jodhbir Singh Mehta
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Kelvin Yi Chong Teo
- Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-National University of Singapore Medical School, Singapore, Singapore
| |
Collapse
|
4
|
Hansen S, Askou AL, la Cour M, Corydon TJ, Bek T. Subretinal Saline Protects the Neuroretina From Thermic Damage During Laser Induction of Experimental Choroidal Neovascularization in Pigs. Transl Vis Sci Technol 2021; 10:29. [PMID: 34185056 PMCID: PMC8254010 DOI: 10.1167/tvst.10.7.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to develop a porcine model for photocoagulation induced choroidal neovascularization (CNV) with high success rate and minimal thermic damage to the neuroretina. Methods Experimental CNV was induced by laser photocoagulation in both eyes of 16 domestic pigs. In the left eyes, photocoagulation was preceded by subretinal injection of saline to protect the neuroretina from thermic damage, whereas the right eyes were treated with photocoagulation only. The development of the CNV after 3, 7, 14, 28, and 42 days was evaluated by optical coherence tomography (OCT) scanning, fluorescein angiography, and OCT angiography, and by histology after enucleation. Results From day 7 after the photocoagulation, OCT showed subretinal density in all lesions of 14 alive animals, and either fluorescein or OCT angiography confirmed CNV formation in 11 of 14 of the eyes that had received photocoagulation alone and those in which photocoagulation had been preceded by subretinal injection of saline. In all cases pretreated with subretinal saline, the neuroretina was protected from immediate thermic damage. The formation of CNVs were confirmed by histology. For both groups, the largest lesions were observed within 14 days after photocoagulation. Conclusions Injection of subretinal saline can protect the retina from thermic damage induced by retinal photocoagulation without reducing the success rate in producing experimental CNV. The effect of interventional studies aimed at reducing photocoagulation induced experimental CNV in pigs can be evaluated within 2 weeks after photocoagulation. Translational Relevance This model provides a fundament to develop and evaluate novel treatment methods for neovascular retinal diseases.
Collapse
Affiliation(s)
- Silja Hansen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Morten la Cour
- Department of Ophthalmology, Rigshospitalet, Copenhagen, Denmark
| | - Thomas J Corydon
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
| | - Toke Bek
- Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
5
|
The Alter Retina: Alternative Splicing of Retinal Genes in Health and Disease. Int J Mol Sci 2021; 22:ijms22041855. [PMID: 33673358 PMCID: PMC7917623 DOI: 10.3390/ijms22041855] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
Alternative splicing of mRNA is an essential mechanism to regulate and increase the diversity of the transcriptome and proteome. Alternative splicing frequently occurs in a tissue- or time-specific manner, contributing to differential gene expression between cell types during development. Neural tissues present extremely complex splicing programs and display the highest number of alternative splicing events. As an extension of the central nervous system, the retina constitutes an excellent system to illustrate the high diversity of neural transcripts. The retina expresses retinal specific splicing factors and produces a large number of alternative transcripts, including exclusive tissue-specific exons, which require an exquisite regulation. In fact, a current challenge in the genetic diagnosis of inherited retinal diseases stems from the lack of information regarding alternative splicing of retinal genes, as a considerable percentage of mutations alter splicing or the relative production of alternative transcripts. Modulation of alternative splicing in the retina is also instrumental in the design of novel therapeutic approaches for retinal dystrophies, since it enables precision medicine for specific mutations.
Collapse
|
6
|
Holmgaard AB, Askou AL, Jensen EG, Alsing S, Bak RO, Mikkelsen JG, Corydon TJ. Targeted Knockout of the Vegfa Gene in the Retina by Subretinal Injection of RNP Complexes Containing Cas9 Protein and Modified sgRNAs. Mol Ther 2021; 29:191-207. [PMID: 33022212 PMCID: PMC7791085 DOI: 10.1016/j.ymthe.2020.09.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/20/2020] [Accepted: 09/20/2020] [Indexed: 12/21/2022] Open
Abstract
The therapeutic effect of retinal gene therapy using CRISPR/Cas9-mediated genome editing and knockout applications is dependent on efficient and safe delivery of gene-modifying tool kits. Recently, transient administration of single guide RNAs (sgRNAs) and SpCas9 proteins delivered as ribonucleoproteins (RNPs) has provided potent gene knockout in vitro. To improve efficacy of CRISPR-based gene therapy, we delivered RNPs containing SpCas9 protein complexed to chemically modified sgRNAs (msgRNAs). In K562 cells, msgRNAs significantly increased the insertion/deletion (indel) frequency (25%) compared with unmodified counterparts leading to robust knockout of the VEGFA gene encoding vascular endothelial growth factor A (96% indels). Likewise, in HEK293 cells, lipoplexes containing varying amounts of RNP and EGFP mRNA showed efficient VEGFA knockout (43% indels) and strong EGFP expression, indicative of efficacious functional knockout using small amounts of RNP. In mice, subretinal injections of equivalent lipoplexes yielded 6% indels in Vegfa of isolated EGFP-positive RPE cells. However, signs of toxicity following delivery of lipoplexes containing high amounts of RNP were observed. Although the mechanism resulting in the varying efficacy remains to be elucidated, our data suggest that a single subretinal injection of RNPs carrying msgRNAs and SpCas9 induces targeted retinal indel formation, thus providing a clinically relevant strategy relying on nonviral delivery of short-lived nuclease activity.
Collapse
Affiliation(s)
| | - Anne Louise Askou
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | | | - Sidsel Alsing
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Rasmus O Bak
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark; Aarhus Institute of Advanced Studies (AIAS), Aarhus University, 8000 Aarhus C, Denmark
| | | | - Thomas J Corydon
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark; Department of Ophthalmology, Aarhus University Hospital, 8200 Aarhus N, Denmark.
| |
Collapse
|
7
|
Abstract
INTRODUCTION In the retina, noncoding RNA (ncRNA) plays an integral role in regulating apoptosis, inflammatory responses, visual perception, and photo-transduction, with altered levels reported in diseased states. AREAS COVERED MicroRNA (miRNA), a class of ncRNA, regulates post-transcription gene expression through the binding of complementary sites of target messenger RNA (mRNA) with resulting translational repression. Small-interfering RNA (siRNA) is a double-stranded RNA (dsRNA) that regulates gene expression, leading to selective silencing of genes through a process called RNA interference (RNAi). Another form of RNAi involves short hairpin RNA (shRNA). In age-related macular degeneration (AMD) and diabetic retinopathy (DR), miRNA has been implicated in the regulation of angiogenesis, oxidative stress, immune response, and inflammation. EXPERT OPINION Many RNA-based therapies in development are conveniently administered intravitreally, with the potential for pan-retinal effect. The majority of these RNA therapeutics are synthetic ncRNA's and hold promise for the treatment of AMD, DR, and inherited retinal diseases (IRDs). These RNA-based therapies include siRNA therapy with its high specificity, shRNA to 'knock down' autosomal dominant toxic gain of function-mutated genes, antisense oligonucleotides (ASOs), which can restore splicing defects, and translational read-through inducing drugs (TRIDs) to increase expression of full-length protein from genes with premature stop codons.
Collapse
Affiliation(s)
- Michael C Gemayel
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, IN, USA
| | - Ashay D Bhatwadekar
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, IN, USA
| | - Thomas Ciulla
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, IN, USA.,Preclinical and Clinical Development, Clearside Biomedical, Inc, Alpharetta, GA, USA.,Midwest Eye Institute, Indianapolis, IN, USA
| |
Collapse
|
8
|
Lin FL, Wang PY, Chuang YF, Wang JH, Wong VHY, Bui BV, Liu GS. Gene Therapy Intervention in Neovascular Eye Disease: A Recent Update. Mol Ther 2020; 28:2120-2138. [PMID: 32649860 PMCID: PMC7544979 DOI: 10.1016/j.ymthe.2020.06.029] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/15/2020] [Accepted: 06/26/2020] [Indexed: 12/13/2022] Open
Abstract
Aberrant growth of blood vessels (neovascularization) is a key feature of severe eye diseases that can cause legal blindness, including neovascular age-related macular degeneration (nAMD) and diabetic retinopathy (DR). The development of anti-vascular endothelial growth factor (VEGF) agents has revolutionized the treatment of ocular neovascularization. Novel proangiogenic targets, such as angiopoietin and platelet-derived growth factor (PDGF), are under development for patients who respond poorly to anti-VEGF therapy and to reduce adverse effects from long-term VEGF inhibition. A rapidly advancing area is gene therapy, which may provide significant therapeutic benefits. Viral vector-mediated transgene delivery provides the potential for continuous production of antiangiogenic proteins, which would avoid the need for repeated anti-VEGF injections. Gene silencing with RNA interference to target ocular angiogenesis has been investigated in clinical trials. Proof-of-concept gene therapy studies using gene-editing tools such as CRISPR-Cas have already been shown to be effective in suppressing neovascularization in animal models, highlighting the therapeutic potential of the system for treatment of aberrant ocular angiogenesis. This review provides updates on the development of anti-VEGF agents and novel antiangiogenic targets. We also summarize current gene therapy strategies already in clinical trials and those with the latest approaches utilizing CRISPR-Cas gene editing against aberrant ocular neovascularization.
Collapse
Affiliation(s)
- Fan-Li Lin
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Peng-Yuan Wang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, VIC 3122, Australia.
| | - Yu-Fan Chuang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Jiang-Hui Wang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Vickie H Y Wong
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC 3010, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC 3010, Australia
| | - Guei-Sheung Liu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia; Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC 3002, Australia.
| |
Collapse
|
9
|
Ameri H, Murat C, Arbabi A, Jiang W, Janga SR, Qin PZ, Hamm-Alvarez SF. Reduced Expression of VEGF-A in Human Retinal Pigment Epithelial Cells and Human Muller Cells Following CRISPR-Cas9 Ribonucleoprotein-Mediated Gene Disruption. Transl Vis Sci Technol 2020; 9:23. [PMID: 32855870 PMCID: PMC7422915 DOI: 10.1167/tvst.9.8.23] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 05/22/2020] [Indexed: 11/30/2022] Open
Abstract
Purpose To evaluate the effects of vascular endothelial growth factor-A (VEGF-A) gene editing in human retinal pigment epithelial (RPE) cells and human Muller cells, which are the main VEGF-A producing cells in the eye. Methods CRISPR-Cas9 ribonucleoprotein was used to target exon 1 in VEGF-A gene. Lipofectamine CRISPRMAX was used as a vehicle. In vitro gene editing efficiency was assessed on oligonucleotides and genomic DNAs. Sanger sequencing was performed to detect indels. VEGF-A messenger RNA and protein expressions were assessed using quantitative polymerase chain reaction and enzyme-linked immunosorbent assay. Results In vitro cleavage assay on a 60-nucleotide DNA duplex showed 88% cleavage of the precursor. The cleavage efficiency was 40% in RPE cells and 32% in Muller cells. Sanger sequencing in the CRISPR-Cas9 treated RPE and Muller cells showed indels at the predicted cut site in both cells. After the VEGF-A gene disruption, VEGF-A protein levels decreased 43% in RPE cells (P < 0.0001) and 38% in Muller cells (P < 0.0001). Conclusions CRISPR-Cas9–mediated gene disruption resulted in a significant decrease in the VEGF-A gene protein expression in human RPE and Muller cells. CRISPR-Cas9 ribonucleoprotein may allow simultaneous targeting of multiple VEGF-A producing cells. Translational Relevance VEGF-A gene disruption using CRISPR-Cas9 ribonucleoprotein has a potential in treating retinal vascular diseases.
Collapse
Affiliation(s)
- Hossein Ameri
- USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Christopher Murat
- USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Amirmohsen Arbabi
- USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Wei Jiang
- USC Dornsife College of Letters, Arts and Sciences, Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Srikanth R Janga
- USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Peter Zhifeng Qin
- USC Dornsife College of Letters, Arts and Sciences, Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Sarah F Hamm-Alvarez
- USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
10
|
Guimaraes TACD, Georgiou M, Bainbridge JWB, Michaelides M. Gene therapy for neovascular age-related macular degeneration: rationale, clinical trials and future directions. Br J Ophthalmol 2020; 105:151-157. [PMID: 32269060 PMCID: PMC7848059 DOI: 10.1136/bjophthalmol-2020-316195] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/18/2020] [Accepted: 03/20/2020] [Indexed: 01/19/2023]
Abstract
Age-related macular degeneration (AMD) is one of the leading causes of irreversible blindness in the developed world. Antivascular endothelial growth factor therapy has transformed the management and outcome of neovascular AMD (nAMD), although the need for repeated intravitreal injections—even lifelong—and the related complications, high drug costs, frequent clinic visits and repeated imaging have resulted in an enormous burden both to healthcare systems and patients. The application of gene therapy approaches for sustained delivery of a range of antiangiogenic proteins has the promise of helping to address these aforementioned challenges. A number of early phase clinical trials of gene therapy in nAMD have provided encouraging results, with many more ongoing or anticipated. There remain significant areas of controversy, including regarding the optimal treatment targets, routes of administration and potential safety concerns. In this review we aim to provide an update of the current status of gene therapy for nAMD and briefly discuss future prospects.
Collapse
Affiliation(s)
| | - Michalis Georgiou
- UCL Institute of Ophthalmology, University College London, London, UK.,Moorfields Eye Hospital, London, UK
| | - James W B Bainbridge
- UCL Institute of Ophthalmology, University College London, London, UK.,Moorfields Eye Hospital, London, UK
| | - Michel Michaelides
- UCL Institute of Ophthalmology, University College London, London, UK .,Moorfields Eye Hospital, London, UK
| |
Collapse
|
11
|
Hughes CP, O’Flynn NM, Gatherer M, McClements ME, Scott JA, MacLaren RE, Goverdhan S, Glennie MJ, Lotery AJ. AAV2/8 Anti-angiogenic Gene Therapy Using Single-Chain Antibodies Inhibits Murine Choroidal Neovascularization. Mol Ther Methods Clin Dev 2019; 13:86-98. [PMID: 30719487 PMCID: PMC6350388 DOI: 10.1016/j.omtm.2018.11.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 11/16/2018] [Indexed: 12/15/2022]
Abstract
While anti-angiogenic therapies for wet age-related macular degeneration (AMD) are effective for many patients, they require multiple injections and are expensive and prone to complications. Gene therapy could be an elegant solution for this problem by providing a long-term source of anti-angiogenic proteins after a single administration. Another potential issue with current therapeutic proteins containing a fragment crystallizable (Fc) domain (such as whole antibodies like bevacizumab) is the induction of an unwanted immune response. In wet AMD, a low level of inflammation is already present, so to avoid exacerbation of disease by the therapeutic protein, we propose single-chain fragment variable (scFv) antibodies, which lack the Fc domain, as a safer alternative. To investigate the feasibility of this, anti-vascular endothelial growth factor (VEGF)-blocking antibodies in two formats were produced and tested in vitro and in vivo. The scFv transgene was then cloned into an adeno-associated virus (AAV) vector. A therapeutic effect in a mouse model of choroidal neovascularization (CNV) was demonstrated with antibodies in both scFv and immunoglobulin G1 (IgG1) formats (p < 0.04). Importantly, the scFv anti-VEGF antibody expressed from an AAV vector also had a significant beneficial effect (p = 0.02), providing valuable preclinical data for future translation to the clinic.
Collapse
Affiliation(s)
- Chris P. Hughes
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Neil M.J. O’Flynn
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Maureen Gatherer
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Michelle E. McClements
- Oxford Eye Hospital and Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jennifer A. Scott
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Robert E. MacLaren
- Oxford Eye Hospital and Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Srinivas Goverdhan
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Martin J. Glennie
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Andrew J. Lotery
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
12
|
Lee SH, Yang JY, Madrakhimov S, Park HY, Park K, Park TK. Adeno-Associated Viral Vector 2 and 9 Transduction Is Enhanced in Streptozotocin-Induced Diabetic Mouse Retina. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 13:55-66. [PMID: 30666309 PMCID: PMC6330514 DOI: 10.1016/j.omtm.2018.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 11/25/2018] [Indexed: 12/19/2022]
Abstract
Adeno-associated viruses (AAVs) are currently the most popular vector platform technology for ocular gene therapy. While transduction efficiency and tropism of intravitreally administered AAV has been fairly well established in various retinal conditions, its transduction pattern in diabetic retinas has not previously been characterized. Here, we describe the transduction efficiencies of four different AAV serotypes, AAV2, 5, 8, and 9, in streptozotocin (STZ)-induced diabetic mouse retinas after intravitreal injections, which differed according to the duration of diabetic induction. STZ was intraperitoneally injected into C57/B6 diabetic mice subjected to unilateral intravitreal injection of AAV2, AAV5, AAV8, and AAV9 packaged with EGFP. Significantly enhanced AAV2 and AAV9 transduction was observed in 2-month-old diabetic mouse retinas compared to the 2-week-old diabetic mouse retinas and nondiabetic, vector uninjected or injected retinas. Intravitreal injection of AAV5 or AAV8 serotype in 2-month- and 2-week-old diabetic mouse retinas did not show any significant vector transduction enhancement compared to the nondiabetic control retinas. The tropism of AAV2 and AAV9 in diabetic mouse retinas differed. AAV2 was transduced into various retinal cells, including Müller cells, microglia, retinal ganglion cells (RGCs), bipolar cells, horizontal cells, and amacrine cells, whereas AAV9 was effectively transduced only into RGC and horizontal cells. The expression levels of receptors and co-receptors for AAV2 and AAV9 were significantly increased in 2-month-old diabetic mouse retinas. The results of our study demonstrated that AAV2 and AAV9 may be the vector of choice in treating diabetic retinopathy (DR) with gene therapy, and DR-related retinal changes may improve AAV vector transduction efficiency.
Collapse
Affiliation(s)
- Si Hyung Lee
- Department of Ophthalmology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea.,Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon 14584, Republic of Korea
| | - Jin Young Yang
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon 14584, Republic of Korea.,Department of Biomedical Science, Graduate School, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Sanjar Madrakhimov
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon 14584, Republic of Korea.,Department of Biomedical Science, Graduate School, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Ha Yan Park
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon 14584, Republic of Korea
| | - Keerang Park
- Department of Biopharmacy, Chungbuk Health & Science University, Cheongju, Chungbuk 28150, Republic of Korea
| | - Tae Kwann Park
- Department of Ophthalmology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea.,Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon 14584, Republic of Korea
| |
Collapse
|
13
|
Wang JK, Huang TL, Chang PY, Chen YT, Chang CW, Chen FT, Hsu YR, Chen YJ. Intravitreal aflibercept versus bevacizumab for treatment of myopic choroidal neovascularization. Sci Rep 2018; 8:14389. [PMID: 30258077 PMCID: PMC6158246 DOI: 10.1038/s41598-018-32761-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/13/2018] [Indexed: 12/13/2022] Open
Abstract
The authors performed a retrospective and comparative study to compare the efficacy of intravitreal aflibercept and bevacizumab for patients with myopic choroidal neovascularization (mCNV). The patients with treatment-naïve mCNV received 1 + PRN intravitreal bevacizumab from March 2008 to February 2013, while from March 2013 to July 2016 patients were treated by 1 + PRN intravitreal aflibercept, all with monthly follow-up for 12 months. Primary outcome measures included change in central foveal thickness (CFT) in 1 mm by spectral-domain optic coherence tomography, and best corrected visual acuity (BCVA) at month 12. Complications after injections were recorded. The intra-group changes in CFT and BCVA were compared with Wilcoxon signed rank test, the between-group difference compared with Wilcoxon rank sum test. Fisher's exact test was used for categorical comparison between groups. Seventy-eight eyes of 78 patients were collected. There were 42 eyes in bevacizumab group, with mean age of 53.2 ± 5.4 years and 27 female patients of them. The mean BCVA significantly improved from baseline 0.56 ± 0.35 logMAR to 0.35 ± 0.35 logMAR at Month 12 after bevacizumab treatment (p < 0.001). The mean CFT significantly decreased from baseline 315.3 ± 25.6 μm to 253.7 ± 24.4 μm at Month 12 following intravitreal bevacizumab (p < 0.001). There were 36 eyes in aflibercept group, with mean age of 52.8 ± 6.8 years and 24 female patients of them. The mean BCVA significantly improved from baseline 0.61 ± 0.47 logMAR to 0.38 ± 0.41 logMAR at Month 12 after aflibercept treatment (p < 0.001). The mean CFT significantly decreased from baseline 328.2 ± 19.8 μm to 241.8 ± 27.2 μm at Month 12 following intravitreal aflibercept (p < 0.001). The baseline demographics, lens status, axial length, refractive errors, duration of symptoms, BCVA, and CFT did not differ significantly between groups (p > 0.05). There was no significant difference between bevacizumab and aflibercept groups in BCVA and CFT from Month 1 to Month 12 (p > 0.05). Injection number of aflibercept was 2.11 ± 0.41, less than that of bevacizumab (3.23 ± 0.38) during 12-month period (p = 0.01). There were no systemic thromboembolic event, elevated intraocular pressure, retinal detachment, or infectious endophthalmitis following injections in both groups. We concluded that both aflibercept and bevacizumab can effectively treat choroidal neovascularization in high myopes. Intravitreal aflibercept had similar efficacy but less treatment number than bevacizumab for mCNV during 12-month period.
Collapse
Affiliation(s)
- Jia-Kang Wang
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan. .,Department of electrical engineering, Yuan Ze University, Taoyuan City, Taiwan. .,Department of Medicine, National Yang Ming University, Taipei City, Taiwan. .,Department of Healthcare Administration and Department of Nursing, Oriental Institute of Technology, New Taipei City, Taiwan. .,Department of Medicine, National Taiwan University, Taipei City, Taiwan.
| | - Tzu-Lun Huang
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan.,Department of electrical engineering, Yuan Ze University, Taoyuan City, Taiwan
| | - Pei-Yao Chang
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan.,Department of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Yen-Ting Chen
- Department of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Chin-Wei Chang
- Department of Medicine, National Yang Ming University, Taipei City, Taiwan
| | - Fang-Ting Chen
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan.,Department of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Yung-Ray Hsu
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan.,Department of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Yun-Ju Chen
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan.,Department of Medicine, National Taiwan University, Taipei City, Taiwan
| |
Collapse
|
14
|
Askou AL, Alsing S, Holmgaard A, Bek T, Corydon TJ. Dissecting microRNA dysregulation in age-related macular degeneration: new targets for eye gene therapy. Acta Ophthalmol 2018; 96:9-23. [PMID: 28271607 DOI: 10.1111/aos.13407] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/02/2017] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) are key regulators of gene expression in humans. Overexpression or depletion of individual miRNAs is associated with human disease. Current knowledge suggests that the retina is influenced by miRNAs and that dysregulation of miRNAs as well as alterations in components of the miRNA biogenesis machinery are involved in retinal diseases, including age-related macular degeneration (AMD). Furthermore, recent studies have indicated that the vitreous has a specific panel of circulating miRNAs and that this panel varies according to the specific pathological stress experienced by the retinal cells. MicroRNA (miRNA) profiling indicates subtype-specific miRNA profiles for late-stage AMD highlighting the importance of proper miRNA regulation in AMD. This review will describe the function of important miRNAs involved in inflammation, oxidative stress and pathological neovascularization, the key molecular mechanisms leading to AMD, and focus on dysregulated miRNAs as potential therapeutic targets in AMD.
Collapse
Affiliation(s)
| | - Sidsel Alsing
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| | | | - Toke Bek
- Department of Ophthalmology; Aarhus University Hospital; Aarhus C Denmark
| | | |
Collapse
|
15
|
Transduction Patterns of Adeno-associated Viral Vectors in a Laser-Induced Choroidal Neovascularization Mouse Model. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 9:90-98. [PMID: 29766021 PMCID: PMC5948198 DOI: 10.1016/j.omtm.2018.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/16/2018] [Indexed: 12/17/2022]
Abstract
Adeno-associated virus (AAV) vector is a promising platform technology for ocular gene therapy. Recently clinical successes to treat choroidal neovascularization (CNV) in wet type age-related macular degeneration have been reported. However, because pathologic conditions of the retina may alter the tropism of viral vectors, it is necessary to evaluate the transduction efficiency of different serotypes of AAV vectors in the retinas with CNVs. Here, we show the patterns and efficacy of transduction of AAV2, -5, and -8 vectors in a laser-induced CNV mouse model. C57BL/6J mice were subjected to unilateral laser photocoagulation on the right eye to induce CNV 5 days prior to intravitreal injection of AAV2, -5, and -8 capsids expressing EGFP. Transduction was increased around CNV lesions for all AAV capsid types, and AAV2 resulted in the highest transduction efficiency. In the absence of CNV, the AAV2 vector transduced ganglion and inner nuclear layer (INL) cells, and AAV5 and AAV8 transduced only a small proportion of cells in the retinal ganglion cell layer. CNV increased AAV2 vector expression throughout the retina and in and around CNVs; the transduced cells included retinal ganglion cells, Müller cells, cells from the INL and outer nuclear layer (ONL), photoreceptors, and retinal pigment epithelium (RPE) cells. Inflammatory cells and endothelial cells in CNVs were also transduced by AAV2. AAV5 and AAV8 were transduced in retinal ganglion, Müller, INL, ONL, and RPE cells in a localized pattern, and only endothelial cells at the surface of CNV lesions showed EGFP expression. Taken together, CNV formation resulted in enhanced transduction of AAV2, -5, and -8, and AAV2 exhibited the highest transduction efficiency in cells in CNV lesions.
Collapse
|
16
|
Askou AL, Benckendorff JNE, Holmgaard A, Storm T, Aagaard L, Bek T, Mikkelsen JG, Corydon TJ. Suppression of Choroidal Neovascularization in Mice by Subretinal Delivery of Multigenic Lentiviral Vectors Encoding Anti-Angiogenic MicroRNAs. Hum Gene Ther Methods 2018; 28:222-233. [PMID: 28817343 DOI: 10.1089/hgtb.2017.079] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Lentivirus-based vectors have been used for the development of potent gene therapies. Here, application of a multigenic lentiviral vector (LV) producing multiple anti-angiogenic microRNAs following subretinal delivery in a laser-induced choroidal neovascularization (CNV) mouse model is presented. This versatile LV, carrying back-to-back RNApolII-driven expression cassettes, enables combined expression of microRNAs targeting vascular endothelial growth factor A (Vegfa) mRNA and fluorescent reporters. In addition, by including a vitelliform macular dystrophy 2 (VMD2) promoter, expression of microRNAs is restricted to the retinal pigment epithelial (RPE) cells. Six days post injection (PI), robust and widespread fluorescent signals of eGFP are already observed in the retina by funduscopy. The eGFP expression peaks at day 21 PI and persists with stable expression for at least 9 months. In parallel, prominent AsRED co-expression, encoded from the VMD2-driven microRNA expression cassette, is evident in retinal sections and flat-mounts, revealing RPE-specific expression of microRNAs. Furthermore, LV-delivered microRNAs targeting the Vegfa gene in RPE cells reduced the size of laser-induced CNV in mice 28 days PI, as a consequence of diminished VEGF levels, suggesting that LVs delivered locally are powerful tools in the development of gene therapy-based strategies for treatment of age-related macular degeneration.
Collapse
Affiliation(s)
| | | | | | - Tina Storm
- 1 Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Lars Aagaard
- 1 Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Toke Bek
- 2 Department of Ophthalmology, Aarhus University Hospital , Aarhus, Denmark
| | | | - Thomas Juhl Corydon
- 1 Department of Biomedicine, Aarhus University, Aarhus, Denmark .,2 Department of Ophthalmology, Aarhus University Hospital , Aarhus, Denmark
| |
Collapse
|
17
|
Development of Multigenic Lentiviral Vectors for Cell-Specific Expression of Antiangiogenic miRNAs and Protein Factors. Methods Mol Biol 2018; 1715:47-60. [PMID: 29188505 DOI: 10.1007/978-1-4939-7522-8_4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Generation of lentivirus (LV)-based vectors holding multiple gene cassettes for coexpression of several therapeutic factors provides potent tools in both gene delivery studies as well as in gene therapy. Here we describe the development of such multigenic LV gene delivery vectors enabling cell-specific coexpression of antiangiogenic microRNA (miRNA) and protein factors and, if preferred, a fluorescent reporter, from RNApol(II)-driven expression cassettes orientated in a back-to-back fashion. This configuration may contribute to the development of new combination therapies for amelioration of diseases involving intraocular neovascularization including exudative age-related macular degeneration (AMD).
Collapse
|
18
|
Askou AL, Benckendorff JNE, Holmgaard A, Storm T, Aagaard L, Bek T, Mikkelsen JG, Corydon TJ. Suppression of choroidal neovascularization in mice by subretinal delivery of multigenic lentiviral vectors encoding anti-angiogenic microRNAs. Hum Gene Ther Methods 2017. [DOI: 10.1089/hum.2017.079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
| | | | | | - Tina Storm
- Aarhus University, Department of Biomedicine, Aarhus C, Denmark
| | - Lars Aagaard
- Aarhus University, Department of Biomedicine, Aarhus C, Denmark
| | - Toke Bek
- Aarhus University Hospital, Department of Ophthalmology, Aarhus C, Denmark
| | | | - Thomas Juhl Corydon
- Aarhus University, Department of Biomedicine, Wilhelm Meyers Alle, Aarhus C, Denmark, 8000
| |
Collapse
|
19
|
Moore NA, Bracha P, Hussain RM, Morral N, Ciulla TA. Gene therapy for age-related macular degeneration. Expert Opin Biol Ther 2017; 17:1235-1244. [PMID: 28726562 DOI: 10.1080/14712598.2017.1356817] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION In neovascular age related macular degeneration (nAMD), gene therapy to chronically express anti-vascular endothelial growth factor (VEGF) proteins could ameliorate the treatment burden of chronic intravitreal therapy and improve limited visual outcomes associated with 'real world' undertreatment. Areas covered: In this review, the authors assess the evolution of gene therapy for AMD. Adeno-associated virus (AAV) vectors can transduce retinal pigment epithelium; one such early application was a phase I trial of AAV2-delivered pigment epithelium derived factor gene in advanced nAMD. Subsequently, gene therapy for AMD shifted to the investigation of soluble fms-like tyrosine kinase-1 (sFLT-1), an endogenously expressed VEGF inhibitor, binding and neutralizing VEGF-A. After some disappointing results, research has centered on novel vectors, including optimized AAV2, AAV8 and lentivirus, as well as genes encoding other anti-angiogenic proteins, including ranibizumab, aflibercept, angiostatin and endostatin. Also, gene therapy targeting the complement system is being investigated for geographic atrophy due to non-neovascular AMD. Expert opinion: The success of gene therapy for AMD will depend on the selection of the most appropriate therapeutic protein and its level of chronic expression. Future investigations will center on optimizing vector, promoter and delivery methods, and evaluating the risks of the chronic expression of anti-angiogenic or anti-complement proteins.
Collapse
Affiliation(s)
- Nicholas A Moore
- a Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Peter Bracha
- a Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Rehan M Hussain
- a Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Nuria Morral
- c Department of Medical and Molecular Genetics , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Thomas A Ciulla
- a Department of Ophthalmology , Indiana University School of Medicine , Indianapolis , IN , USA.,b Retina Service , Midwest Eye Institute , Indianapolis , IN , USA
| |
Collapse
|
20
|
Adeno-Associated Viral Vector-Mediated mTOR Inhibition by Short Hairpin RNA Suppresses Laser-Induced Choroidal Neovascularization. MOLECULAR THERAPY-NUCLEIC ACIDS 2017; 8:26-35. [PMID: 28918027 PMCID: PMC5477068 DOI: 10.1016/j.omtn.2017.05.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/29/2017] [Accepted: 05/29/2017] [Indexed: 12/20/2022]
Abstract
Choroidal neovascularization (CNV) is the defining characteristic feature of the wet subtype of age-related macular degeneration (AMD) and may result in irreversible blindness. Based on anti-vascular endothelial growth factor (anti-VEGF), the current therapeutic approaches to CNV are fraught with difficulties, and mammalian target of rapamycin (mTOR) has recently been proposed as a possible therapeutic target, although few studies have been conducted. Here, we show that a recombinant adeno-associated virus-delivered mTOR-inhibiting short hairpin RNA (rAAV-mTOR shRNA), which blocks the activity of both mTOR complex 1 and 2, represents a promising therapeutic approach for the treatment of CNV. Eight-week-old male C57/B6 mice were treated with the short hairpin RNA (shRNA) after generating CNV lesions in the eyes via laser photocoagulation. The recombinant adeno-associated virus (rAAV) delivery vehicle was able to effectively transduce cells in the inner retina, and significantly fewer inflammatory cells and less extensive CNV were observed in the animals treated with rAAV-mTOR shRNA when compared with control- and rAAV-scrambled shRNA-treated groups. Presumably related to the reduction of CNV, increased autophagy was detected in CNV lesions treated with rAAV-mTOR shRNA, whereas significantly fewer apoptotic cells detected in the outer nuclear layer around the CNV indicate that mTOR inhibition may also have neuroprotective effects. Taken together, these results demonstrate the therapeutic potential of mTOR inhibition, resulting from rAAV-mTOR shRNA activity, in the treatment of AMD-related CNV.
Collapse
|
21
|
Yiu G, Tieu E, Nguyen AT, Wong B, Smit-McBride Z. Genomic Disruption of VEGF-A Expression in Human Retinal Pigment Epithelial Cells Using CRISPR-Cas9 Endonuclease. Invest Ophthalmol Vis Sci 2016; 57:5490-5497. [PMID: 27768202 PMCID: PMC5072539 DOI: 10.1167/iovs.16-20296] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/18/2016] [Indexed: 12/26/2022] Open
Abstract
PURPOSE To employ type II clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 endonuclease to suppress ocular angiogenesis by genomic disruption of VEGF-A in human RPE cells. METHODS CRISPR sequences targeting exon 1 of human VEGF-A were computationally identified based on predicted Cas9 on- and off-target probabilities. Single guide RNA (gRNA) cassettes with these target sequences were cloned into lentiviral vectors encoding the Streptococcuspyogenes Cas9 endonuclease (SpCas9) gene. The lentiviral vectors were used to infect ARPE-19 cells, a human RPE cell line. Frequency of insertion or deletion (indel) mutations was assessed by T7 endonuclease 1 mismatch detection assay; mRNA levels were assessed with quantitative real-time PCR; and VEGF-A protein levels were determined by ELISA. In vitro angiogenesis was measured using an endothelial cell tube formation assay. RESULTS Five gRNAs targeting VEGF-A were selected based on the highest predicted on-target probabilities, lowest off-target probabilities, or combined average of both scores. Lentiviral delivery of the top-scoring gRNAs with SpCas9 resulted in indel formation in the VEGF-A gene at frequencies up to 37.0% ± 4.0% with corresponding decreases in secreted VEGF-A protein up to 41.2% ± 7.4% (P < 0.001), and reduction of endothelial tube formation up to 39.4% ± 9.8% (P = 0.02). No significant indel formation in the top three putative off-target sites tested was detected. CONCLUSIONS The CRISPR-Cas9 endonuclease system may reduce VEGF-A secretion from human RPE cells and suppress angiogenesis, supporting the possibility of employing gene editing for antiangiogenesis therapy in ocular diseases.
Collapse
Affiliation(s)
- Glenn Yiu
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California, United States
| | - Eric Tieu
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California, United States
| | - Anthony T. Nguyen
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California, United States
| | - Brittany Wong
- Duke University, Durham, North Carolina, United States
| | - Zeljka Smit-McBride
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, California, United States
| |
Collapse
|
22
|
Ishihara Y, Tsuno S, Kuwamoto S, Yamashita T, Endo Y, Miura K, Miura Y, Sato T, Hasegawa J, Miura N. Tumor-suppressive effects of atelocollagen-conjugated hsa-miR-520d-5p on un-differentiated cancer cells in a mouse xenograft model. BMC Cancer 2016; 16:415. [PMID: 27388711 PMCID: PMC4936056 DOI: 10.1186/s12885-016-2467-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 06/29/2016] [Indexed: 12/31/2022] Open
Abstract
Background We previously demonstrated that hsa-miR-520d-5p can convert cancer cells into induced pluripotent stem cells (iPSCs) or mesenchymal stem cells (MSCs) via a demethylation process and p53 upregulation in vivo. Additionally, we have reported the non-tumorigenic effect of miR-520d-5p on normal human cells, including fibroblasts. Methods We used atelocollagen-conjugated miR-520d-5p (520d/atelocollagen) to confirm the possibility of a therapeutic effect on cancer cells. We traced the size and signal intensity of GFP-expressing tumors in mice each week, beginning 4 weeks after subcutaneous inoculation. Results 520d/atelocollagen treatment suppressed tumor growth by greater than 80 % each week relative to controls and resulted in an approximately 30 % disappearance of tumors. In mice whose tumors disappeared, the existence of human genomic material at the injection site was examined by quantitative Alu-PCR, and we confirmed the co-existence of both species-derived cells. In every site where a tumor disappeared in immunodeficient mice, GFP protein was expressed in the connective tissues, and approximately 0.1 % of the extracted DNA contained human genomic material. We could not identify any adverse effects in vivo. Conclusions This is the first report to confirm an inhibitory effect of 520d/atelocollagen on cancer cells in vivo. The development of optimized modifications of this carrier is expected to enhance the efficiency of entry into tumor cells and the induction of its inhibitory effect. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2467-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yoshitaka Ishihara
- Division of Pharmacotherapeutics, Department of Pathophysiological & Therapeutic Science, Faculty of Medicine, Tottori University, 86 Nishicho, Yonago, Tottori, 683-8503, Japan
| | - Satoshi Tsuno
- Division of Pharmacotherapeutics, Department of Pathophysiological & Therapeutic Science, Faculty of Medicine, Tottori University, 86 Nishicho, Yonago, Tottori, 683-8503, Japan
| | - Satoshi Kuwamoto
- Division of Molecular Pathology, Faculty of Medicine, Tottori University, 86 Nishicho, Yonago, Tottori, 683-8503, Japan
| | - Taro Yamashita
- Department of Gastroenterology, Tottori University Hospital, 86 Nishicho, Yonago, Tottori, 683-8504, Japan
| | - Yusuke Endo
- Division of Pharmacotherapeutics, Department of Pathophysiological & Therapeutic Science, Faculty of Medicine, Tottori University, 86 Nishicho, Yonago, Tottori, 683-8503, Japan
| | - Keigo Miura
- PEZY-Pharma, 86 Nishicho, Yonago, Tottori, 683-8503, Japan
| | - Yugo Miura
- Orthopedic Surgery, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Takemasa Sato
- Division of Neurobiology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishicho, Yonago, Tottori, 683-8503, Japan
| | - Junichi Hasegawa
- Division of Pharmacotherapeutics, Department of Pathophysiological & Therapeutic Science, Faculty of Medicine, Tottori University, 86 Nishicho, Yonago, Tottori, 683-8503, Japan
| | - Norimasa Miura
- Division of Pharmacotherapeutics, Department of Pathophysiological & Therapeutic Science, Faculty of Medicine, Tottori University, 86 Nishicho, Yonago, Tottori, 683-8503, Japan.
| |
Collapse
|
23
|
Abstract
The idea of treating disease in humans with genetic material was conceived over two decades ago and with that a promising journey involving development and efficacy studies in cells and animals of a large number of novel therapeutic reagents unfolded. In the footsteps of this process, successful gene therapy treatment of genetic conditions in humans has shown clear signs of efficacy. Notably, significant advancements using gene supplementation and silencing strategies have been made in the field of ocular gene therapy, thereby pinpointing ocular gene therapy as one of the compelling "actors" bringing gene therapy to the clinic. Most of all, this success has been facilitated because of (1) the fact that the eye is an effortlessly accessible, exceedingly compartmentalized, and immune-privileged organ offering a unique advantage as a gene therapy target, and (2) significant progress toward efficient, sustained transduction of cells within the retina having been achieved using nonintegrating vectors based on recombinant adeno-associated virus and nonintegrating lentivirus vectors. The results from in vivo experiments and trials suggest that treatment of inherited retinal dystrophies, ocular angiogenesis, and inflammation with gene therapy can be both safe and effective. Here, the progress of ocular gene therapy is examined with special emphasis on the potential use of RNAi- and protein-based antiangiogenic gene therapy to treat exudative age-related macular degeneration.
Collapse
Affiliation(s)
- Thomas J Corydon
- Department of Biomedicine, Aarhus University , Aarhus C, Denmark
| |
Collapse
|
24
|
Retinal gene delivery by adeno-associated virus (AAV) vectors: Strategies and applications. Eur J Pharm Biopharm 2015; 95:343-52. [DOI: 10.1016/j.ejpb.2015.01.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 01/10/2015] [Accepted: 01/12/2015] [Indexed: 11/20/2022]
|
25
|
Pattnaik BR, Shahi PK, Marino MJ, Liu X, York N, Brar S, Chiang J, Pillers DAM, Traboulsi EI. A Novel KCNJ13 Nonsense Mutation and Loss of Kir7.1 Channel Function Causes Leber Congenital Amaurosis (LCA16). Hum Mutat 2015; 36:720-7. [PMID: 25921210 DOI: 10.1002/humu.22807] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/20/2015] [Indexed: 11/08/2022]
Abstract
Mutations in the KCNJ13 gene that encodes the inwardly rectifying potassium channel Kir7.1 cause snowflake vitreoretinal degeneration (SVD) and leber congenital amaurosis (LCA). Kir7.1 controls the microenvironment between the photoreceptors and the retinal pigment epithelium (RPE) and also contributes to the function of other organs such as uterus and brain. Heterologous expressions of the mutant channel have suggested a dominant-negative loss of Kir7.1 function in SVD, but parallel studies in LCA16 have been lacking. Herein, we report the identification of a novel nonsense mutation in the second exon of the KCNJ13 gene that leads to a premature stop codon in association with LCA16. We have determined that the mutation results in a severe truncation of the Kir7.1 C-terminus, alters protein localization, and disrupts potassium currents. Coexpression of the mutant and wild-type channel has no negative influence on the wild-type channel function, consistent with the normal clinical phenotype of carrier individuals. By suppressing Kir7.1 function in mice, we were able to reproduce the severe LCA electroretinogram phenotype. Thus, we have extended the observation that Kir7.1 mutations are associated with vision disorders to include novel insights into the molecular mechanism of disease pathobiology in LCA16.
Collapse
Affiliation(s)
- Bikash R Pattnaik
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin.,Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, Wisconsin.,McPherson Eye Research Institute, University of Wisconsin, Madison, Wisconsin
| | - Pawan K Shahi
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin
| | - Meghan J Marino
- Center for Genetic Eye Diseases and Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Xinying Liu
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin
| | - Nathaniel York
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin
| | - Simran Brar
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin
| | - John Chiang
- Casey Molecular Diagnostic Laboratory, Oregon Health & Science University, Portland, Oregon
| | - De-Ann M Pillers
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin.,McPherson Eye Research Institute, University of Wisconsin, Madison, Wisconsin
| | - Elias I Traboulsi
- Center for Genetic Eye Diseases and Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| |
Collapse
|
26
|
Askou AL, Aagaard L, Kostic C, Arsenijevic Y, Hollensen AK, Bek T, Jensen TG, Mikkelsen JG, Corydon TJ. Multigenic lentiviral vectors for combined and tissue-specific expression of miRNA- and protein-based antiangiogenic factors. Mol Ther Methods Clin Dev 2015; 2:14064. [PMID: 26052532 PMCID: PMC4449022 DOI: 10.1038/mtm.2014.64] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/14/2014] [Accepted: 12/10/2014] [Indexed: 01/22/2023]
Abstract
Lentivirus-based gene delivery vectors carrying multiple gene cassettes are powerful tools in gene transfer studies and gene therapy, allowing coexpression of multiple therapeutic factors and, if desired, fluorescent reporters. Current strategies to express transgenes and microRNA (miRNA) clusters from a single vector have certain limitations that affect transgene expression levels and/or vector titers. In this study, we describe a novel vector design that facilitates combined expression of therapeutic RNA- and protein-based antiangiogenic factors as well as a fluorescent reporter from back-to-back RNApolII-driven expression cassettes. This configuration allows effective production of intron-embedded miRNAs that are released upon transduction of target cells. Exploiting such multigenic lentiviral vectors, we demonstrate robust miRNA-directed downregulation of vascular endothelial growth factor (VEGF) expression, leading to reduced angiogenesis, and parallel impairment of angiogenic pathways by codelivering the gene encoding pigment epithelium-derived factor (PEDF). Notably, subretinal injections of lentiviral vectors reveal efficient retinal pigment epithelium-specific gene expression driven by the VMD2 promoter, verifying that multigenic lentiviral vectors can be produced with high titers sufficient for in vivo applications. Altogether, our results suggest the potential applicability of combined miRNA- and protein-encoding lentiviral vectors in antiangiogenic gene therapy, including new combination therapies for amelioration of age-related macular degeneration.
Collapse
Affiliation(s)
| | - Lars Aagaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Corinne Kostic
- Department of Ophthalmology, Unit of Gene Therapy and Stem Cell Biology, University of Lausanne, Jules-Gonin Eye Hospital, Lausanne, Switzerland
| | - Yvan Arsenijevic
- Department of Ophthalmology, Unit of Gene Therapy and Stem Cell Biology, University of Lausanne, Jules-Gonin Eye Hospital, Lausanne, Switzerland
| | | | - Toke Bek
- Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | |
Collapse
|
27
|
Cho YW, Han YS, Chung IY, Kim SJ, Seo SW, Yoo JM, Park JM. Suppression of laser-induced choroidal neovascularization by intravitreal injection of tristetraprolin. Int J Ophthalmol 2014; 7:952-8. [PMID: 25540745 DOI: 10.3980/j.issn.2222-3959.2014.06.07] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 08/25/2014] [Indexed: 11/02/2022] Open
Abstract
AIM To examine the effect of intravitreal adenoviral vector-mediated tristetraprolin (Ad-TTP) on VEGF mRNA expression in a rat model of laser-induced choroidal neovascularization. METHODS Ad-TTP was prepared using a commercial kit. Retinal laser-induced photocoagulation (10 spots per eye) was performed on rats in this experimental choroidal neovascularization (CNV) model. Rats were divided into four groups: control (single intravitreal injection of balanced salt solution, n=10), laser-induced CNV (photocoagulation only, n=20), laser-induced CNV plus Ad-TTP injection (photocoagulation plus a single intravitreal Ad-TTP injection, n=20) and Ad-TTP injection only (n=10). Changes in choroidal morphology were evaluated in ten rats in the laser only and the laser plus Ad-TTP groups. Two weeks after laser injury, the size of CNV was calculated by perfusion with high-molecular-weight fluorescein isothiocyanate (FITC)-dextran. VEGF mRNA expression in retina-choroid tissue from ten rats in each group was measured by reverse transcription polymerase chain reaction (RT-PCR). RESULTS Two weeks after treatment, the area of laser-induced CNV was reduced by approximately 60% in the rats given the Ad-TTP injection compared with that in the laser-only group. There was a tendency toward decreased VEGF mRNA expression in the Ad-TTP injection groups. CONCLUSION A single intravitreal injection of Ad-TTP significantly suppressed CNV size in this experimental laser-induced CNV model. Ad-TTP injection also decreased VEGF mRNA expression compared with that in the laser-induced CNV group. The present study is meaningful as the first study to investigate the effect of tristetraprolin delivered via intravitreal injection.
Collapse
Affiliation(s)
- Yong Wun Cho
- Department of Ophthalmology, Gyeongsang National University School of Medicine, Jinju 660-702, Korea
| | - Yong Seop Han
- Department of Ophthalmology, Gyeongsang National University School of Medicine, Jinju 660-702, Korea ; Gyeongsang Institute of Health Science, Gyeongsang National University, Jinju 660-702, Korea
| | - In Young Chung
- Department of Ophthalmology, Gyeongsang National University School of Medicine, Jinju 660-702, Korea ; Gyeongsang Institute of Health Science, Gyeongsang National University, Jinju 660-702, Korea
| | - Seong Jae Kim
- Department of Ophthalmology, Gyeongsang National University School of Medicine, Jinju 660-702, Korea
| | - Seong Wook Seo
- Department of Ophthalmology, Gyeongsang National University School of Medicine, Jinju 660-702, Korea ; Gyeongsang Institute of Health Science, Gyeongsang National University, Jinju 660-702, Korea
| | - Ji Myong Yoo
- Department of Ophthalmology, Gyeongsang National University School of Medicine, Jinju 660-702, Korea ; Gyeongsang Institute of Health Science, Gyeongsang National University, Jinju 660-702, Korea
| | - Jong Moon Park
- Department of Ophthalmology, Gyeongsang National University School of Medicine, Jinju 660-702, Korea ; Gyeongsang Institute of Health Science, Gyeongsang National University, Jinju 660-702, Korea
| |
Collapse
|
28
|
Pechan P, Wadsworth S, Scaria A. Gene Therapies for Neovascular Age-Related Macular Degeneration. Cold Spring Harb Perspect Med 2014; 5:a017335. [PMID: 25524721 DOI: 10.1101/cshperspect.a017335] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pathological neovascularization is a key component of the neovascular form (also known as the wet form) of age-related macular degeneration (AMD) and proliferative diabetic retinopathy. Several preclinical studies have shown that antiangiogenesis strategies are effective for treating neovascular AMD in animal models. Vascular endothelial growth factor (VEGF) is one of the main inducers of ocular neovascularization, and several clinical trials have shown the benefits of neutralizing VEGF in patients with neovascular AMD or diabetic macular edema. In this review, we summarize several preclinical and early-stage clinical trials with intraocular gene therapies, which have the potential to reduce or eliminate the repeated intravitreal injections that are currently required for the treatment of neovascular AMD.
Collapse
Affiliation(s)
- Peter Pechan
- Gene Therapy, Sanofi-Genzyme R&D Center, Framingham, Massachusetts 01701
| | - Samuel Wadsworth
- Gene Therapy, Sanofi-Genzyme R&D Center, Framingham, Massachusetts 01701
| | - Abraham Scaria
- Gene Therapy, Sanofi-Genzyme R&D Center, Framingham, Massachusetts 01701
| |
Collapse
|
29
|
Li H, Wan C, Du L, Li F. Enhanced downregulation of transforming growth factor‑β2 in rat retinal pigment epithelium cells by adeno‑associated virus‑mediated ribonucleic acid interference combined with ultrasound or microbubbles. Mol Med Rep 2014; 11:1099-104. [PMID: 25370502 DOI: 10.3892/mmr.2014.2845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 10/06/2014] [Indexed: 11/06/2022] Open
Abstract
The present study was designed to determine the efficiency and safety of ultrasound (US) and/or US contrast agent microbubbles (MBs) in the delivery of type 2 recombinant adeno-associated virus‑delivered transforming growth factor‑β2 short hairpin ribonucleic acid encoding the enhanced green fluorescent protein gene (rAAV2‑TGFβ2 shRNA‑EGFP) and the downregulation of TGFβ2 in rat retinal pigment epithelium (RPE‑J) cells. The effects of US and/or MBs on the delivery of rAAV2‑EGFP and rAAV2‑TGFβ2 shRNA‑EGFP were evaluated by fluorescence microscopy and flow cytometry. The potential toxicity of cell viability under various US or MB conditions was assessed by CellTiter 96® AQueous One solution cell proliferation assay. The level of TGFβ2 mRNA in RPE‑J cells under various conditions was estimated by reverse transcription‑quantitative polymerase chain reaction analysis. The results obtained demonstrated that low-intensity US (0.5 W/cm2 and 30 sec) or SonoVue (MB:cell ratio, 40:1) increased the delivery efficiency of rAAV2‑EGFP and rAAV2‑TGFβ2 shRNA‑EGFP to RPE‑J cells, whereas the combination of US with MBs did not further increase but instead decreased rAAV transfection. Under the optimal conditions of rAAV delivery, enhanced TGFβ2 gene silencing with a combination of US or SonoVue with rAAV2‑TGFβ2 shRNA resulted in a significant decrease in mRNA levels compared with rAAV2‑TGFβ2 shRNA alone. US or SonoVue was used safely to enhance the delivery of rAAV2‑TGFβ2 shRNA to RPE‑J cells. A combination of the biological (rAAV2‑TGFβ2 shRNA) and physical (US or SonoVue) approaches downregulated the mRNA level of TGFβ2 more effectively.
Collapse
Affiliation(s)
- Hongli Li
- Department of Ultrasound, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Caifeng Wan
- Department of Ultrasound, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Lianfang Du
- Department of Ultrasound, Shanghai First People's Hospital, Shanghai 200080, P.R. China
| | - Fenghua Li
- Department of Ultrasound, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| |
Collapse
|
30
|
Biswal MR, Prentice HM, Dorey CK, Blanks JC. A hypoxia-responsive glial cell-specific gene therapy vector for targeting retinal neovascularization. Invest Ophthalmol Vis Sci 2014; 55:8044-53. [PMID: 25377223 DOI: 10.1167/iovs.14-13932] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Müller cells, the major glial cell in the retina, play a significant role in retinal neovascularization in response to tissue hypoxia. We previously designed and tested a vector using a hypoxia-responsive domain and a glial fibrillary acidic protein (GFAP) promoter to drive green fluorescent protein (GFP) expression in Müller cells in the murine model of oxygen-induced retinopathy (OIR). This study compares the efficacy of regulated and unregulated Müller cell delivery of endostatin in preventing neovascularization in the OIR model. METHODS Endostatin cDNA was cloned into plasmids with hypoxia-regulated GFAP or unregulated GFAP promoters, and packaged into self-complementary adeno-associated virus serotype 2 vectors (scAAV2). Before placement in hyperoxia on postnatal day (P)7, mice were given intravitreal injections of regulated or unregulated scAAV2, capsid, or PBS. Five days after return to room air, on P17, neovascular and avascular areas, as well as expression of the transgene and vascular endothelial growth factor (VEGF), were compared in OIR animals treated with a vector, capsid, or PBS. RESULTS The hypoxia-regulated, glial-specific, vector-expressing endostatin reduced neovascularization by 93% and reduced the central vaso-obliteration area by 90%, matching the results with the unregulated GFAP-Endo vector. Retinas treated with the regulated endostatin vector expressed substantial amounts of endostatin protein, and significantly reduced VEGF protein. Endostatin production from the regulated vector was undetectable in retinas with undamaged vasculature. CONCLUSIONS These findings suggest that the hypoxia-regulated, glial cell-specific vector expressing endostatin may be useful for treatment of neovascularization in proliferative diabetic retinopathy.
Collapse
Affiliation(s)
- Manas R Biswal
- Integrative Biology PhD Program, Florida Atlantic University, Boca Raton, Florida, United States
| | - Howard M Prentice
- Center for Complex Systems and Brain Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, Florida, United States Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, United States
| | - C Kathleen Dorey
- Virginia Tech Carilion School of Medicine, Roanoke, Virginia, United States
| | - Janet C Blanks
- Center for Complex Systems and Brain Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, Florida, United States
| |
Collapse
|
31
|
Askou AL. Development of gene therapy for treatment of age-related macular degeneration. Acta Ophthalmol 2014. [DOI: 10.1111/aos.12569] [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]
|
32
|
Zhang X, Das SK, Passi SF, Uehara H, Bohner A, Chen M, Tiem M, Archer B, Ambati BK. AAV2 delivery of Flt23k intraceptors inhibits murine choroidal neovascularization. Mol Ther 2014; 23:226-34. [PMID: 25306972 DOI: 10.1038/mt.2014.199] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 10/02/2014] [Indexed: 01/09/2023] Open
Abstract
Long-term inhibition of extracellular vascular endothelial growth factor (VEGF) in the treatment of age-related macular degeneration (AMD) may induce retinal neuronal toxicity and risk other side effects. We developed a novel strategy which inhibits retinal pigment epithelium (RPE)-derived VEGF, sparing other highly sensitive retinal tissues. Flt23k, an intraceptor inhibitor of VEGF, was able to inhibit VEGF in vitro. Adeno-associated virus type 2 (AAV2)-mediated expression of Flt23k was maintained for up to 6 months postsubretinal injection in mice. Flt23k was able to effectively inhibit laser-induced murine choroidal neovascularization (CNV). VEGF levels in the RPE/choroid complex decreased significantly in AAV2.Flt23k treated eyes. Neither retinal structure detected by Heidelberg Spectralis nor function measured by electroretinography (ERG) was adversely affected by treatment with AAV2.Flt23k. Hence AAV2.Flt23k can effectively maintain long-term expression and inhibit laser-induced CNV in mice through downregulation of VEGF while maintaining a sound retinal safety profile. These findings suggest a promising novel approach for the treatment of CNV.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Moran Eye Center, University of Utah, Salt Lake City, Utah, USA
| | - Subrata K Das
- Moran Eye Center, University of Utah, Salt Lake City, Utah, USA
| | - Samuel F Passi
- Moran Eye Center, University of Utah, Salt Lake City, Utah, USA
| | - Hironori Uehara
- Moran Eye Center, University of Utah, Salt Lake City, Utah, USA
| | - Austin Bohner
- Moran Eye Center, University of Utah, Salt Lake City, Utah, USA
| | - Marcus Chen
- Moran Eye Center, University of Utah, Salt Lake City, Utah, USA
| | - Michelle Tiem
- Moran Eye Center, University of Utah, Salt Lake City, Utah, USA
| | - Bonnie Archer
- Moran Eye Center, University of Utah, Salt Lake City, Utah, USA
| | | |
Collapse
|
33
|
Conley SM, Naash MI. Gene therapy for PRPH2-associated ocular disease: challenges and prospects. Cold Spring Harb Perspect Med 2014; 4:a017376. [PMID: 25167981 DOI: 10.1101/cshperspect.a017376] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The peripherin-2 (PRPH2) gene encodes a photoreceptor-specific tetraspanin protein called peripherin-2/retinal degeneration slow (RDS), which is critical for the formation and maintenance of rod and cone outer segments. Over 90 different disease-causing mutations in PRPH2 have been identified, which cause a variety of forms of retinitis pigmentosa and macular degeneration. Given the disease burden associated with PRPH2 mutations, the gene has long been a focus for preclinical gene therapy studies. Adeno-associated viruses and compacted DNA nanoparticles carrying PRPH2 have been successfully used to mediate improvement in the rds(-/-) and rds(+/-) mouse models. However, complexities in the pathogenic mechanism for PRPH2-associated macular disease coupled with the need for a precise dose of peripherin-2 to combat a severe haploinsufficiency phenotype have delayed the development of clinically viable genetic treatments. Here we discuss the progress and prospects for PRPH2-associated gene therapy.
Collapse
Affiliation(s)
- Shannon M Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Muna I Naash
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| |
Collapse
|
34
|
Cuenca N, Fernández-Sánchez L, Campello L, Maneu V, De la Villa P, Lax P, Pinilla I. Cellular responses following retinal injuries and therapeutic approaches for neurodegenerative diseases. Prog Retin Eye Res 2014; 43:17-75. [PMID: 25038518 DOI: 10.1016/j.preteyeres.2014.07.001] [Citation(s) in RCA: 302] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/03/2014] [Accepted: 07/07/2014] [Indexed: 01/17/2023]
Abstract
Retinal neurodegenerative diseases like age-related macular degeneration, glaucoma, diabetic retinopathy and retinitis pigmentosa each have a different etiology and pathogenesis. However, at the cellular and molecular level, the response to retinal injury is similar in all of them, and results in morphological and functional impairment of retinal cells. This retinal degeneration may be triggered by gene defects, increased intraocular pressure, high levels of blood glucose, other types of stress or aging, but they all frequently induce a set of cell signals that lead to well-established and similar morphological and functional changes, including controlled cell death and retinal remodeling. Interestingly, an inflammatory response, oxidative stress and activation of apoptotic pathways are common features in all these diseases. Furthermore, it is important to note the relevant role of glial cells, including astrocytes, Müller cells and microglia, because their response to injury is decisive for maintaining the health of the retina or its degeneration. Several therapeutic approaches have been developed to preserve retinal function or restore eyesight in pathological conditions. In this context, neuroprotective compounds, gene therapy, cell transplantation or artificial devices should be applied at the appropriate stage of retinal degeneration to obtain successful results. This review provides an overview of the common and distinctive features of retinal neurodegenerative diseases, including the molecular, anatomical and functional changes caused by the cellular response to damage, in order to establish appropriate treatments for these pathologies.
Collapse
Affiliation(s)
- Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain; Multidisciplinary Institute for Environmental Studies "Ramon Margalef", University of Alicante, Alicante, Spain.
| | - Laura Fernández-Sánchez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Laura Campello
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, Alicante, Spain
| | - Pedro De la Villa
- Department of Systems Biology, University of Alcalá, Alcalá de Henares, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Isabel Pinilla
- Department of Ophthalmology, Lozano Blesa University Hospital, Aragon Institute of Health Sciences, Zaragoza, Spain
| |
Collapse
|
35
|
RNA interference-based therapy for spinocerebellar ataxia type 7 retinal degeneration. PLoS One 2014; 9:e95362. [PMID: 24759684 PMCID: PMC3997397 DOI: 10.1371/journal.pone.0095362] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 03/26/2014] [Indexed: 11/21/2022] Open
Abstract
Spinocerebellar ataxia type 7 (SCA7) is an autosomal dominant neurodegenerative disease characterized by loss of motor coordination and retinal degeneration with no current therapies in the clinic. The causative mutation is an expanded CAG repeat in the ataxin-7 gene whose mutant protein product causes cerebellar and brainstem degeneration and retinal cone-rod dystrophy. Here, we reduced the expression of both mutant and wildtype ataxin-7 in the SCA7 mouse retina by RNA interference and evaluated retinal function 23 weeks post injection. We observed a preservation of normal retinal function and no adverse toxicity with ≥50% reduction of mutant and wildtype ataxin-7 alleles. These studies address an important safety concern regarding non-allele specific silencing of ataxin-7 for SCA7 retinal therapy.
Collapse
|
36
|
Birke MT, Lipo E, Adhi M, Birke K, Kumar-Singh R. AAV-mediated expression of human PRELP inhibits complement activation, choroidal neovascularization and deposition of membrane attack complex in mice. Gene Ther 2014; 21:507-13. [PMID: 24670995 DOI: 10.1038/gt.2014.24] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 02/08/2014] [Accepted: 02/11/2014] [Indexed: 01/17/2023]
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness among the elderly. Approximately 50% of AMD patients have a polymorphism in the negative regulator of complement known as Factor H. Individuals homozygous for a Y402H polymorphism in Factor H have elevated levels of membrane attack complex (MAC) in their choroid and retinal pigment epithelium relative to individuals homozygous for the wild-type allele. An inability to form MAC due to a polymorphism in C9 is protective against the formation of choroidal neovascularization (CNV) in AMD patients. Hence, blocking MAC in AMD patients may be protective against CNV. Here we investigate the potential of human proline/arginine-rich end leucine-rich repeat protein (PRELP) as an inhibitor of complement-mediated damage when delivered via the subretinal route using an AAV2/8 vector. In a fluorescence-activated cell sorting (FACS) lysis assay, PRELP inhibited normal human serum-mediated lysis of Hepa-1c1c7 cells by 18.7%. Unexpectedly, PRELP enhanced the formation of tubes by human umbilical vein endothelial cells (HUVECs) by approximately 240%, but, when delivered via an AAV vector to the retina of mice, PRELP inhibited laser-induced CNV by 60%. PRELP reduced deposition of MAC in vivo by 25.5%. Our results have implications for the development of complement inhibitors as a therapy for AMD.
Collapse
Affiliation(s)
- M T Birke
- Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA
| | - E Lipo
- Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA
| | - M Adhi
- Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA
| | - K Birke
- Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA
| | - R Kumar-Singh
- Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
37
|
Cui J, Liu Y, Zhang J, Yan H. An experimental study on choroidal neovascularization induced by Krypton laser in rat model. Photomed Laser Surg 2013; 32:30-6. [PMID: 24328846 DOI: 10.1089/pho.2013.3588] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE The purpose of this work was to study the efficacy and safety of choroidal neovascularization (CNV) formation induced by Krypton laser in Brown Norway (BN) rats, and observe the trend of the change of CNV after laser photocoagulation. METHODS Twenty-five male BN rats were involved in this study. Two eyes of one rat without any laser photocoagulation were randomly selected as the control group, and the other 48 eyes of 24 rats were selected as the experimental group. Eight eyes of four rats were randomly selected to receive the examinations of fundus fluorescein angiography (FFA), histopathology, and transmission electron microscopy 3, 7, 14, 21, 28, and 56 days after laser photocoagulation. RESULTS After laser photocoagulation, the leakage appeared in burns on day 7 (59%), reached the peak on day 21 (84%), (p<0.01), and remained stable after day 21, proven by FFA and histopathology (p>0.05). The thickness of CNV increased from day 7 to day 21 (p<0.01), and remained stable after day 21 (p>0.05). CONCLUSIONS The experimental model of CNV can be successfully induced by Krypton laser in rats with a stable, long-lasting, and high success rate. After laser photocoagulation, the leakages appear on day 7, reach the peak on day 21, and remain stable after day 21.
Collapse
Affiliation(s)
- Jing Cui
- 1 Department of Ophthalmology, Tianjin Medical University General Hospital , Tianjin, China
| | | | | | | |
Collapse
|