1
|
Bengtsson NE, Tasfaout H, Chamberlain JS. The road toward AAV-mediated gene therapy of Duchenne muscular dystrophy. Mol Ther 2025; 33:2035-2051. [PMID: 40181545 DOI: 10.1016/j.ymthe.2025.03.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/31/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025] Open
Abstract
Forty years after the dystrophin gene was cloned, significant progress has been made in developing gene therapy approaches for Duchenne muscular dystrophy (DMD). The disorder has presented numerous challenges, including the enormous size of the gene (2.2 MB), the need to target muscles body wide, and immunogenic issues against both vectors and dystrophin. Among human genetic disorders, DMD is relatively common, and the genetics are complicated since one-third of all cases arise from a spontaneous new mutation, resulting in thousands of independent lesions throughout the locus. Many approaches have been pursued in the goal of finding an effective therapy, including exon skipping, nonsense codon suppression, upregulation of surrogate genes, gene replacement, and gene editing. Here, we focus specifically on methods using AAV vectors, as these approaches have been tested in numerous clinical trials and are able to target muscles systemically. We discuss early advances to understand the structure of dystrophin, which are crucial for the design of effective DMD gene therapies. Included is a summary of efforts to deliver micro-, mini-, and full-length dystrophins to muscles. Finally, we review current approaches to adapt gene editing to the enormous DMD gene with prospects for improved therapies using all these methods.
Collapse
Affiliation(s)
- Niclas E Bengtsson
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98109, USA; Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA 98109, USA.
| | - Hichem Tasfaout
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98109, USA; Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA 98109, USA.
| | - Jeffrey S Chamberlain
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98109, USA; Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Biochemistry, University of Washington School of Medicine, Seattle, WA 98109, USA.
| |
Collapse
|
2
|
Suarez-Amaran L, Song L, Tretiakova AP, Mikhail SA, Samulski RJ. AAV vector development, back to the future. Mol Ther 2025; 33:1903-1936. [PMID: 40186350 DOI: 10.1016/j.ymthe.2025.03.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025] Open
Abstract
Adeno-associated virus (AAV) has become a pivotal tool in gene therapy, providing a safe and efficient platform for long-term transgene expression. This review presents a comprehensive analysis of AAV's historical development, from its initial identification as a "contaminant" to its current clinical applications. We examine the molecular evolution of AAV, detailing advancements in vector engineering, rational design, directed evolution platforms, and computational modeling, which have expanded its therapeutic potential across diverse disease areas. Additionally, we explore AAV genome regulation, with a particular focus on inverted terminal repeats (ITRs) and AAV capsid-genome interactions, which play a crucial role in vector transduction efficiency and host adaptation. An assessment of past and present clinical trials as well as future directions is provided to illustrate the field's trajectory. Finally, another unique milestone in AAV research is also reported; namely, a pool of AAV libraries has been successfully administered to human decedents and analyzed, representing a transformative step in AAV evolution and selection for human applications. These studies should pave the way for more refined AAV vector optimization, accelerating the development of next-generation gene therapies with enhanced clinical translatability, potentially accelerating the gene therapy revolution.
Collapse
Affiliation(s)
- Lester Suarez-Amaran
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA; Orthopaedic Research Center, C. Wayne McIlwraith Translational Medicine Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA
| | - Liujiang Song
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA
| | - Anna P Tretiakova
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA
| | - Sheila A Mikhail
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA
| | - Richard Jude Samulski
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA; Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
3
|
McMahon M, Maquat LE. Exploring the therapeutic potential of modulating nonsense-mediated mRNA decay. RNA (NEW YORK, N.Y.) 2025; 31:333-348. [PMID: 39667907 PMCID: PMC11874985 DOI: 10.1261/rna.080334.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024]
Abstract
Discovered more than four decades ago, nonsense-mediated mRNA decay (NMD) plays a fundamental role in the regulation of gene expression and is a major contributor to numerous diseases. With advanced technologies, several novel approaches aim to directly circumvent the effects of disease-causing frameshift and nonsense mutations. Additional therapeutics aim to globally dampen the NMD pathway in diseases associated with pathway hyperactivation, one example being Fragile X syndrome. In other cases, therapeutics have been designed to hijack or inhibit the cellular NMD machinery to either activate or obviate transcript-specific NMD by modulating pre-mRNA splicing. Here, we discuss promising approaches employed to regulate NMD for therapeutic purposes and highlight potential challenges in future clinical development. We are optimistic that the future of developing target-specific and global modulators of NMD (inhibitors as well as activators) is bright and will revolutionize the treatment of many genetic disorders, especially those with high unmet medical need.
Collapse
Affiliation(s)
- Mary McMahon
- ReviR Therapeutics, Brisbane, California 94005, USA
| | - Lynne E Maquat
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA
- Center for RNA Biology, University of Rochester, Rochester, New York 14642, USA
| |
Collapse
|
4
|
Maurer AC, Benyamini B, Whitney ON, Fan VB, Cattoglio C, Kissiov DU, Dailey GM, Darzacq X, Weitzman MD, Tjian R. Double-strand break repair pathways differentially affect processing and transduction by dual AAV vectors. Nat Commun 2025; 16:1532. [PMID: 39934106 PMCID: PMC11814140 DOI: 10.1038/s41467-025-56738-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/29/2025] [Indexed: 02/13/2025] Open
Abstract
Recombinant adeno-associated viral vectors (rAAV) are a powerful tool for gene delivery but have a limited DNA carrying capacity. Efforts to expand this genetic payload have focused on engineering the vector components, such as dual trans-splicing vectors which double the delivery size by exploiting the natural concatenation of rAAV genomes in host nuclei. We hypothesized that inefficient dual vector transduction could be improved by modulating host factors which affect concatenation. Since factors mediating concatenation are not well defined, we performed a genome-wide screen to identify host cell regulators. We discover that Homologous Recombination (HR) is inhibitory to dual vector transduction. We demonstrate that depletion or inhibition of HR factors BRCA1 and Rad51 significantly increase reconstitution of a large split transgene by increasing both concatenation and expression from rAAVs. Our results define roles for DNA damage repair in rAAV transduction and highlight the potential for pharmacological intervention to increase genetic payload of rAAV vectors.
Collapse
Affiliation(s)
- Anna C Maurer
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
- CIRM Center of Excellence, University of California, Berkeley, CA, USA.
| | - Brian Benyamini
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Oscar N Whitney
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Vinson B Fan
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Claudia Cattoglio
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- CIRM Center of Excellence, University of California, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
| | - Djem U Kissiov
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
| | - Gina M Dailey
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Li Ka Shing Center for Biomedical & Health Sciences, University of California, Berkeley, CA, USA
| | - Xavier Darzacq
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Li Ka Shing Center for Biomedical & Health Sciences, University of California, Berkeley, CA, USA
| | - Matthew D Weitzman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine and the Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Robert Tjian
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
- Li Ka Shing Center for Biomedical & Health Sciences, University of California, Berkeley, CA, USA
| |
Collapse
|
5
|
Ivanchenko MV, Hathaway DM, Mulhall EM, Booth KT, Wang M, Peters CW, Klein AJ, Chen X, Li Y, György B, Corey DP. PCDH15 Dual-AAV Gene Therapy for Deafness and Blindness in Usher Syndrome Type 1F Models. J Clin Invest 2024; 134:e177700. [PMID: 39441757 PMCID: PMC11601915 DOI: 10.1172/jci177700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 10/03/2024] [Indexed: 10/25/2024] Open
Abstract
Usher syndrome type 1F (USH1F), resulting from mutations in the protocadherin-15 (PCDH15) gene, is characterized by congenital lack of hearing and balance, and progressive blindness in the form of retinitis pigmentosa. In this study, we explore an approach for USH1F gene therapy, exceeding the single AAV packaging limit by employing a dual adeno-associated virus (AAV) strategy to deliver the full-length PCDH15 coding sequence. We demonstrate the efficacy of this strategy in mouse USH1F models, effectively restoring hearing and balance in these mice. Importantly, our approach also proves successful in expressing PCDH15 protein in clinically relevant retinal models, including human retinal organoids and non-human primate retina, showing efficient targeting of photoreceptors and proper protein expression in the calyceal processes. This research represents a major step toward advancing gene therapy for USH1F and the multiple challenges of hearing, balance, and vision impairment.
Collapse
Affiliation(s)
| | - Daniel M. Hathaway
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Eric M. Mulhall
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Kevin T.A. Booth
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Mantian Wang
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Cole W. Peters
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Alex J. Klein
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Xinlan Chen
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Yaqiao Li
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Bence György
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - David P. Corey
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Palmieri L, Ferrand M, Vu Hong A, Richard I, Albini S. In Silico Structural Prediction for the Generation of Novel Performant Midi-Dystrophins Based on Intein-Mediated Dual AAV Approach. Int J Mol Sci 2024; 25:10444. [PMID: 39408775 PMCID: PMC11476470 DOI: 10.3390/ijms251910444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is a pediatric disorder characterized by progressive muscle degeneration and premature death, and has no current cure. The current, most promising therapeutic avenue is based on gene replacement mediated by adeno-associated viruses (AAVs) using a shortened, but still functional, version of dystrophin, known as micro-dystrophin (µDys), to fit AAV capacity. The limited improvements observed in clinical trials suggest a sub-optimal performance of µDys in the human context that could be due to the lack of key domains in the protein. Therefore, expressing larger dystrophin proteins may be necessary for a more complete correction of the disease phenotype. In this study, we developed three novel midi-dystrophin constructs using a dual-AAV approach, leveraging split-intein-based protein trans-splicing. The midi-dystrophins include additional domains compared to µDys, such as the central cytoskeleton-binding domain, nNOS and Par1b interacting domains, and a complete C-terminal region. Given the limited capacity of each AAV vector, we strategically partially reduced hinge regions while ensuring that the structural stability of the protein remains intact. We predicted the interactions between the two halves of the split midi-Dys proteins thanks to the deep learning algorithm AphaFold3. We observed strong associations between the N- and C-termini in midi-Dys 1 and 2, while a weaker interaction in midi-Dys 3 was revealed. Our subsequent experiments confirmed the efficient protein trans-splicing both in vitro and in vivo in DBA2/mdx mice of the midi-Dys 1 and 2 and not in midi-Dys 3 as expected from the structural prediction. Additionally, we demonstrated that midi-Dys 1 and 2 exhibit significant therapeutic efficacy in DBA2/mdx mice, highlighting their potential as therapeutic agents for DMD. Overall, these findings highlight the potential of deep learning-based structural modeling for the generation of intein-based dystrophin versions and pose the basis for further investigation of these new midi-dystrophins versions for clinical studies.
Collapse
Affiliation(s)
- Laura Palmieri
- Genethon, 91000 Evry, France; (L.P.); (M.F.); (A.V.H.); (I.R.)
- INTEGRARE Research Unit UMR_S951 (INSERM, Université Paris-Saclay, Univ Evry), 91000 Evry, France
| | - Maxime Ferrand
- Genethon, 91000 Evry, France; (L.P.); (M.F.); (A.V.H.); (I.R.)
- INTEGRARE Research Unit UMR_S951 (INSERM, Université Paris-Saclay, Univ Evry), 91000 Evry, France
| | - Ai Vu Hong
- Genethon, 91000 Evry, France; (L.P.); (M.F.); (A.V.H.); (I.R.)
- INTEGRARE Research Unit UMR_S951 (INSERM, Université Paris-Saclay, Univ Evry), 91000 Evry, France
| | - Isabelle Richard
- Genethon, 91000 Evry, France; (L.P.); (M.F.); (A.V.H.); (I.R.)
- INTEGRARE Research Unit UMR_S951 (INSERM, Université Paris-Saclay, Univ Evry), 91000 Evry, France
- Atamyo Therapeutics, 1, Bis Rue de l’Internationale, 91000 Evry, France
| | - Sonia Albini
- Genethon, 91000 Evry, France; (L.P.); (M.F.); (A.V.H.); (I.R.)
- INTEGRARE Research Unit UMR_S951 (INSERM, Université Paris-Saclay, Univ Evry), 91000 Evry, France
| |
Collapse
|
7
|
Zhou Y, Zhang C, Xiao W, Herzog RW, Han R. Systemic delivery of full-length dystrophin in Duchenne muscular dystrophy mice. Nat Commun 2024; 15:6141. [PMID: 39034316 PMCID: PMC11271493 DOI: 10.1038/s41467-024-50569-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
Current gene therapy for Duchenne muscular dystrophy (DMD) utilizes adeno-associated virus (AAV) to deliver micro-dystrophin (µDys), which does not provide full protection for striated muscles as it lacks many important functional domains of full-length (FL) dystrophin. Here we develop a triple vector system to deliver FL-dystrophin into skeletal and cardiac muscles. We split FL-dystrophin into three fragments linked to two orthogonal pairs of split intein, allowing efficient assembly of FL-dystrophin. The three fragments packaged in myotropic AAV (MyoAAV4A) restore FL-dystrophin expression in both skeletal and cardiac muscles in male mdx4cv mice. Dystrophin-glycoprotein complex components are also restored at the sarcolemma of dystrophic muscles. MyoAAV4A-delivered FL-dystrophin significantly improves muscle histopathology, contractility, and overall strength comparable to µDys, but unlike µDys, it also restores defective cavin 4 localization and associated signaling in mdx4cv heart. Therefore, our data support the feasibility of a mutation-independent FL-dystrophin gene therapy for DMD, warranting further clinical development.
Collapse
Affiliation(s)
- Yuan Zhou
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Chen Zhang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Weidong Xiao
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Roland W Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Renzhi Han
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
8
|
Meng X, Jia R, Zhao X, Zhang F, Chen S, Yu S, Liu X, Dou H, Feng X, Zhang J, Wang N, Xu B, Yang L. In vivo genome editing via CRISPR/Cas9-mediated homology-independent targeted integration for Bietti crystalline corneoretinal dystrophy treatment. Nat Commun 2024; 15:3773. [PMID: 38710738 DOI: 10.1038/s41467-024-48092-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 04/22/2024] [Indexed: 05/08/2024] Open
Abstract
Bietti crystalline corneoretinal dystrophy (BCD) is an autosomal recessive chorioretinal degenerative disease without approved therapeutic drugs. It is caused by mutations in CYP4V2 gene, and about 80% of BCD patients carry mutations in exon 7 to 11. Here, we apply CRISPR/Cas9 mediated homology-independent targeted integration (HITI)-based gene editing therapy in HEK293T cells, BCD patient derived iPSCs, and humanized Cyp4v3 mouse model (h-Cyp4v3mut/mut) using two rAAV2/8 vectors via sub-retinal administration. We find that sgRNA-guided Cas9 generates double-strand cleavage on intron 6 of the CYP4V2 gene, and the HITI donor inserts the carried sequence, part of intron 6, exon 7-11, and a stop codon into the DNA break, achieving precise integration, effective transcription and translation both in vitro and in vivo. HITI-based editing restores the viability of iPSC-RPE cells from BCD patient, improves the morphology, number and metabolism of RPE and photoreceptors in h-Cyp4v3mut/mut mice. These results suggest that HITI-based editing could be a promising therapeutic strategy for those BCD patients carrying mutations in exon 7 to 11, and one injection will achieve lifelong effectiveness.
Collapse
Affiliation(s)
- Xiang Meng
- Department of Ophthalmology, Third Hospital, Peking University, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Ruixuan Jia
- Department of Ophthalmology, Third Hospital, Peking University, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | | | - Fan Zhang
- Beijing Chinagene Co., LTD, Beijing, China
| | | | - Shicheng Yu
- Department of Ophthalmology, Third Hospital, Peking University, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Xiaozhen Liu
- Department of Ophthalmology, Third Hospital, Peking University, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Hongliang Dou
- Department of Ophthalmology, Third Hospital, Peking University, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Xuefeng Feng
- Department of Ophthalmology, Third Hospital, Peking University, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | | | - Ni Wang
- Beijing Chinagene Co., LTD, Beijing, China
| | - Boling Xu
- Department of Ophthalmology, Third Hospital, Peking University, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Liping Yang
- Department of Ophthalmology, Third Hospital, Peking University, Beijing, China.
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
9
|
Han R, Zhou Y, Zhang C, Xiao W, Herzog R. Systemic Delivery of Full-Length Dystrophin in DMD Mice. RESEARCH SQUARE 2024:rs.3.rs-3867299. [PMID: 38746161 PMCID: PMC11092816 DOI: 10.21203/rs.3.rs-3867299/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Current gene therapy for Duchenne muscular dystrophy (DMD) utilizes adeno-associated virus (AAV) to deliver miniaturized dystrophin (micro-dystrophin or µDys), which does not provide full protection for striated muscles as it lacks many important functional domains within full-length (FL) dystrophin. Here we develop a triple vector system to deliver FL-dystrophin into skeletal and cardiac muscles. We rationally split FL-dystrophin into three fragments (N, M, and C) linked to two orthogonal pairs of split intein, allowing efficient, unidirectional assembly of FL-dystrophin. The three fragments packaged in myotropic AAV (MyoAAV4A) restore FL-dystrophin expression in both skeletal and cardiac muscles in male mdx 4cv mice. Dystrophin-glycoprotein complex components are also restored in the sarcolemma of dystrophic muscles. MyoAAV4A-delivered FL-dystrophin significantly improves muscle histopathology, contractility, and overall strength comparable to µDys, but unlike µDys, it also restores defective ERK signaling in heart. The FL-dystrophin gene therapy therefore promises to offer superior protection for DMD.
Collapse
|
10
|
Kolesnik VV, Nurtdinov RF, Oloruntimehin ES, Karabelsky AV, Malogolovkin AS. Optimization strategies and advances in the research and development of AAV-based gene therapy to deliver large transgenes. Clin Transl Med 2024; 14:e1607. [PMID: 38488469 PMCID: PMC10941601 DOI: 10.1002/ctm2.1607] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 02/07/2024] [Accepted: 02/15/2024] [Indexed: 03/18/2024] Open
Abstract
Adeno-associated virus (AAV)-based therapies are recognized as one of the most potent next-generation treatments for inherited and genetic diseases. However, several biological and technological aspects of AAV vectors remain a critical issue for their widespread clinical application. Among them, the limited capacity of the AAV genome significantly hinders the development of AAV-based gene therapy. In this context, genetically modified transgenes compatible with AAV are opening up new opportunities for unlimited gene therapies for many genetic disorders. Recent advances in de novo protein design and remodelling are paving the way for new, more efficient and targeted gene therapeutics. Using computational and genetic tools, AAV expression cassette and transgenic DNA can be split, miniaturized, shuffled or created from scratch to mediate efficient gene transfer into targeted cells. In this review, we highlight recent advances in AAV-based gene therapy with a focus on its use in translational research. We summarize recent research and development in gene therapy, with an emphasis on large transgenes (>4.8 kb) and optimizing strategies applied by biomedical companies in the research pipeline. We critically discuss the prospects for AAV-based treatment and some emerging challenges. We anticipate that the continued development of novel computational tools will lead to rapid advances in basic gene therapy research and translational studies.
Collapse
Affiliation(s)
- Valeria V. Kolesnik
- Martsinovsky Institute of Medical ParasitologyTropical and Vector‐Borne Diseases, Sechenov UniversityMoscowRussia
| | - Ruslan F. Nurtdinov
- Martsinovsky Institute of Medical ParasitologyTropical and Vector‐Borne Diseases, Sechenov UniversityMoscowRussia
| | - Ezekiel Sola Oloruntimehin
- Martsinovsky Institute of Medical ParasitologyTropical and Vector‐Borne Diseases, Sechenov UniversityMoscowRussia
| | | | - Alexander S. Malogolovkin
- Martsinovsky Institute of Medical ParasitologyTropical and Vector‐Borne Diseases, Sechenov UniversityMoscowRussia
- Center for Translational MedicineSirius University of Science and TechnologySochiRussia
| |
Collapse
|
11
|
McDonald A, Wijnholds J. Retinal Ciliopathies and Potential Gene Therapies: A Focus on Human iPSC-Derived Organoid Models. Int J Mol Sci 2024; 25:2887. [PMID: 38474133 PMCID: PMC10932180 DOI: 10.3390/ijms25052887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
The human photoreceptor function is dependent on a highly specialised cilium. Perturbation of cilial function can often lead to death of the photoreceptor and loss of vision. Retinal ciliopathies are a genetically diverse range of inherited retinal disorders affecting aspects of the photoreceptor cilium. Despite advances in the understanding of retinal ciliopathies utilising animal disease models, they can often lack the ability to accurately mimic the observed patient phenotype, possibly due to structural and functional deviations from the human retina. Human-induced pluripotent stem cells (hiPSCs) can be utilised to generate an alternative disease model, the 3D retinal organoid, which contains all major retinal cell types including photoreceptors complete with cilial structures. These retinal organoids facilitate the study of disease mechanisms and potential therapies in a human-derived system. Three-dimensional retinal organoids are still a developing technology, and despite impressive progress, several limitations remain. This review will discuss the state of hiPSC-derived retinal organoid technology for accurately modelling prominent retinal ciliopathies related to genes, including RPGR, CEP290, MYO7A, and USH2A. Additionally, we will discuss the development of novel gene therapy approaches targeting retinal ciliopathies, including the delivery of large genes and gene-editing techniques.
Collapse
Affiliation(s)
- Andrew McDonald
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZC Leiden, The Netherlands;
| | - Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZC Leiden, The Netherlands;
- Netherlands Institute of Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands
| |
Collapse
|
12
|
Li L, Shen T, Liu S, Qi J, Zhao Y. Advancements and future prospects of adeno-associated virus-mediated gene therapy for sensorineural hearing loss. Front Neurosci 2024; 18:1272786. [PMID: 38327848 PMCID: PMC10847333 DOI: 10.3389/fnins.2024.1272786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/12/2024] [Indexed: 02/09/2024] Open
Abstract
Sensorineural hearing loss (SNHL), a highly prevalent sensory impairment, results from a multifaceted interaction of genetic and environmental factors. As we continually gain insights into the molecular basis of auditory development and the growing compendium of deafness genes identified, research on gene therapy for SNHL has significantly deepened. Adeno-associated virus (AAV), considered a relatively secure vector for gene therapy in clinical trials, can deliver various transgenes based on gene therapy strategies such as gene replacement, gene silencing, gene editing, or gene addition to alleviate diverse types of SNHL. This review delved into the preclinical advances in AAV-based gene therapy for SNHL, spanning hereditary and acquired types. Particular focus is placed on the dual-AAV construction method and its application, the vector delivery route of mouse inner ear models (local, systemic, fetal, and cerebrospinal fluid administration), and the significant considerations in transforming from AAV-based animal model inner ear gene therapy to clinical implementation.
Collapse
Affiliation(s)
- Linke Li
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Shen
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shixi Liu
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jieyu Qi
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Yu Zhao
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Ning K, Zhang X, Feng Z, Hao S, Kuz CA, Cheng F, Park SY, McFarlin S, Engelhardt JF, Yan Z, Qiu J. Inhibition of DNA-dependent protein kinase catalytic subunit boosts rAAV transduction of polarized human airway epithelium. Mol Ther Methods Clin Dev 2023; 31:101115. [PMID: 37841417 PMCID: PMC10568418 DOI: 10.1016/j.omtm.2023.101115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023]
Abstract
Adeno-associated virus 2.5T (AAV2.5T) was selected from the directed evolution of AAV capsid library in human airway epithelia. This study found that recombinant AAV2.5T (rAAV2.5T) transduction of well-differentiated primary human airway epithelia induced a DNA damage response (DDR) characterized by the phosphorylation of replication protein A32 (RPA32), histone variant H2AX (H2A histone family member X), and all three phosphatidylinositol 3-kinase-related kinases: ataxia telangiectasia mutated kinase, ataxia telangiectasia and Rad3-related kinase (ATR), and DNA-dependent protein kinase catalytic subunit (DNA-PKcs). While suppressing the expression of ATR by a specific pharmacological inhibitor or targeted gene silencing inhibited rAAV2.5T transduction, DNA-PKcs inhibition or targeted gene silencing significantly increased rAAV2.5T transgene expression. Notably, DNA-PKcs inhibitors worked as a "booster" to further increase rAAV2.5T transgene expression after treatment with doxorubicin and did not compromise epithelial integrity. Thus, our study provides evidence that DDR is associated with rAAV transduction in well-differentiated human airway epithelia, and DNA-PKcs inhibition has the potential to boost rAAV transduction. These findings highlight that the application of DDR inhibition-associated pharmacological interventions has the potential to increase rAAV transduction and thus to reduce the required vector dose.
Collapse
Affiliation(s)
- Kang Ning
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Xiujuan Zhang
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Zehua Feng
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Siyuan Hao
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Cagla Aksu Kuz
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Fang Cheng
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Soo Yuen Park
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Shane McFarlin
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Ziying Yan
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
14
|
Zhang L, Wang H, Xun M, Tang H, Wang J, Lv J, Zhu B, Chen Y, Wang D, Hu S, Gao Z, Liu J, Chen ZY, Chen B, Li H, Shu Y. Preclinical evaluation of the efficacy and safety of AAV1-hOTOF in mice and nonhuman primates. Mol Ther Methods Clin Dev 2023; 31:101154. [PMID: 38027066 PMCID: PMC10679773 DOI: 10.1016/j.omtm.2023.101154] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023]
Abstract
Pathogenic mutations in the OTOF gene cause autosomal recessive hearing loss (DFNB9), one of the most common forms of auditory neuropathy. There is no biological treatment for DFNB9. Here, we designed an OTOF gene therapy agent by dual-adeno-associated virus 1 (AAV1) carrying human OTOF coding sequences with the expression driven by the hair cell-specific promoter Myo15, AAV1-hOTOF. To develop a clinical application of AAV1-hOTOF gene therapy, we evaluated its efficacy and safety in animal models using pharmacodynamics, behavior, and histopathology. AAV1-hOTOF inner ear delivery significantly improved hearing in Otof-/- mice without affecting normal hearing in wild-type mice. AAV1 was predominately distributed to the cochlea, although it was detected in other organs such as the CNS and the liver, and no obvious toxic effects of AAV1-hOTOF were observed in mice. To further evaluate the safety of Myo15 promoter-driven AAV1-transgene, AAV1-GFP was delivered into the inner ear of Macaca fascicularis via the round window membrane. AAV1-GFP transduced 60%-94% of the inner hair cells along the cochlear turns. AAV1-GFP was detected in isolated organs and no significant adverse effects were detected. These results suggest that AAV1-hOTOF is well tolerated and effective in animals, providing critical support for its clinical translation.
Collapse
Affiliation(s)
- Longlong Zhang
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Hui Wang
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Mengzhao Xun
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Honghai Tang
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Jinghan Wang
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Jun Lv
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Biyun Zhu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Yuxin Chen
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Daqi Wang
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Shaowei Hu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Ziwen Gao
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Jianping Liu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Zheng-Yi Chen
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, 243 Charles Street, Boston, MA 02114, USA
| | - Bing Chen
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Yilai Shu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| |
Collapse
|
15
|
Riaz S, Sethna S, Duncan T, Naeem MA, Redmond TM, Riazuddin S, Riazuddin S, Carvalho LS, Ahmed ZM. Dual AAV-based PCDH15 gene therapy achieves sustained rescue of visual function in a mouse model of Usher syndrome 1F. Mol Ther 2023; 31:3490-3501. [PMID: 37864333 PMCID: PMC10727994 DOI: 10.1016/j.ymthe.2023.10.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/20/2023] [Accepted: 10/18/2023] [Indexed: 10/22/2023] Open
Abstract
Mutations in the PCDH15 gene, encoding protocadherin-15, are among the leading causes of Usher syndrome type 1 (USH1F), and account for up to 12% USH1 cases worldwide. A founder truncating variant of PCDH15 has a ∼2% carrier frequency in Ashkenazi Jews accounting for nearly 60% of their USH1 cases. Although cochlear implants can restore hearing perception in USH1 patients, presently there are no effective treatments for the vision loss due to retinitis pigmentosa. We established a founder allele-specific Pcdh15 knockin mouse model as a platform to ascertain therapeutic strategies. Using a dual-vector approach to circumvent the size limitation of adeno-associated virus, we observed robust expression of exogenous PCDH15 in the retinae of Pcdh15KI mice, sustained recovery of electroretinogram amplitudes and key retinoid oxime, substantially improved light-dependent translocation of phototransduction proteins, and enhanced levels of retinal pigment epithelium-derived enzymes. Thus, our data raise hope and pave the way for future gene therapy trials in USH1F subjects.
Collapse
Affiliation(s)
- Sehar Riaz
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; National Centre of Excellence in Molecular Biology, University of the Punjab, Lahore 54500, Pakistan
| | - Saumil Sethna
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Todd Duncan
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Muhammad A Naeem
- National Centre of Excellence in Molecular Biology, University of the Punjab, Lahore 54500, Pakistan
| | - T Michael Redmond
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sheikh Riazuddin
- Jinnah Burn and Reconstructive Surgery Centre, Allama Iqbal Medical Research, University of Health Sciences, Lahore 54500, Pakistan
| | - Saima Riazuddin
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Molecular Biology and Biochemistry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Livia S Carvalho
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA 6009, Australia; Retinal Genomics and Therapy Group, Lions Eye Institute Ltd, Nedlands, WA 6009, Australia
| | - Zubair M Ahmed
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Molecular Biology and Biochemistry, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
16
|
Ivanchenko MV, Hathaway DM, Mulhall EM, Booth KT, Wang M, Peters CW, Klein AJ, Chen X, Li Y, György B, Corey DP. PCDH15 Dual-AAV Gene Therapy for Deafness and Blindness in Usher Syndrome Type 1F. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.09.566447. [PMID: 38014037 PMCID: PMC10680673 DOI: 10.1101/2023.11.09.566447] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Usher syndrome type 1F (USH1F), resulting from mutations in the protocadherin-15 (PCDH15) gene, is characterized by congenital lack of hearing and balance, and progressive blindness in the form of retinitis pigmentosa. In this study, we explore a novel approach for USH1F gene therapy, exceeding the single AAV packaging limit by employing a dual adeno-associated virus (AAV) strategy to deliver the full-length PCDH15 coding sequence. We demonstrate the efficacy of this strategy in mouse USH1F models, effectively restoring hearing and balance in these mice. Importantly, our approach also proves successful in expressing PCDH15 in clinically relevant retinal models, including human retinal organoids and non-human primate retina, showing efficient targeting of photoreceptors and proper protein expression in the calyceal processes. This research represents a major step toward advancing gene therapy for USH1F and the multiple challenges of hearing, balance, and vision impairment.
Collapse
|
17
|
Riedmayr LM, Hinrichsmeyer KS, Thalhammer SB, Mittas DM, Karguth N, Otify DY, Böhm S, Weber VJ, Bartoschek MD, Splith V, Brümmer M, Ferreira R, Boon N, Wögenstein GM, Grimm C, Wijnholds J, Mehlfeld V, Michalakis S, Fenske S, Biel M, Becirovic E. mRNA trans-splicing dual AAV vectors for (epi)genome editing and gene therapy. Nat Commun 2023; 14:6578. [PMID: 37852949 PMCID: PMC10584818 DOI: 10.1038/s41467-023-42386-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/10/2023] [Indexed: 10/20/2023] Open
Abstract
Large genes including several CRISPR-Cas modules like gene activators (CRISPRa) require dual adeno-associated viral (AAV) vectors for an efficient in vivo delivery and expression. Current dual AAV vector approaches have important limitations, e.g., low reconstitution efficiency, production of alien proteins, or low flexibility in split site selection. Here, we present a dual AAV vector technology based on reconstitution via mRNA trans-splicing (REVeRT). REVeRT is flexible in split site selection and can efficiently reconstitute different split genes in numerous in vitro models, in human organoids, and in vivo. Furthermore, REVeRT can functionally reconstitute a CRISPRa module targeting genes in various mouse tissues and organs in single or multiplexed approaches upon different routes of administration. Finally, REVeRT enabled the reconstitution of full-length ABCA4 after intravitreal injection in a mouse model of Stargardt disease. Due to its flexibility and efficiency REVeRT harbors great potential for basic research and clinical applications.
Collapse
Affiliation(s)
- Lisa Maria Riedmayr
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
| | | | | | - David Manuel Mittas
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
| | - Nina Karguth
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
| | - Dina Yehia Otify
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
| | | | - Valentin Johannes Weber
- Laboratory for Retinal Gene Therapy, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren, 8952, Switzerland
| | | | | | - Manuela Brümmer
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
| | - Raphael Ferreira
- Genetics Department, Harvard Medical School, Boston, MA, 02115, USA
| | - Nanda Boon
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA, Leiden, Netherlands
| | - Gabriele Maria Wögenstein
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren, 8952, Switzerland
| | - Christian Grimm
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren, 8952, Switzerland
| | - Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA, Leiden, Netherlands
- Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA, Amsterdam, Netherlands
| | - Verena Mehlfeld
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
| | - Stylianos Michalakis
- Department of Ophthalmology, University Hospital, LMU Munich, 80336, Munich, Germany
| | - Stefanie Fenske
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, 81377, Germany
| | - Martin Biel
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
| | - Elvir Becirovic
- Laboratory for Retinal Gene Therapy, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren, 8952, Switzerland.
| |
Collapse
|
18
|
Li L, Vasan L, Kartono B, Clifford K, Attarpour A, Sharma R, Mandrozos M, Kim A, Zhao W, Belotserkovsky A, Verkuyl C, Schmitt-Ulms G. Advances in Recombinant Adeno-Associated Virus Vectors for Neurodegenerative Diseases. Biomedicines 2023; 11:2725. [PMID: 37893099 PMCID: PMC10603849 DOI: 10.3390/biomedicines11102725] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) vectors are gene therapy delivery tools that offer a promising platform for the treatment of neurodegenerative diseases. Keeping up with developments in this fast-moving area of research is a challenge. This review was thus written with the intention to introduce this field of study to those who are new to it and direct others who are struggling to stay abreast of the literature towards notable recent studies. In ten sections, we briefly highlight early milestones within this field and its first clinical success stories. We showcase current clinical trials, which focus on gene replacement, gene augmentation, or gene suppression strategies. Next, we discuss ongoing efforts to improve the tropism of rAAV vectors for brain applications and introduce pre-clinical research directed toward harnessing rAAV vectors for gene editing applications. Subsequently, we present common genetic elements coded by the single-stranded DNA of rAAV vectors, their so-called payloads. Our focus is on recent advances that are bound to increase treatment efficacies. As needed, we included studies outside the neurodegenerative disease field that showcased improved pre-clinical designs of all-in-one rAAV vectors for gene editing applications. Finally, we discuss risks associated with off-target effects and inadvertent immunogenicity that these technologies harbor as well as the mitigation strategies available to date to make their application safer.
Collapse
Affiliation(s)
- Leyao Li
- Department of Biochemistry, University of Toronto, Medical Sciences Building, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
| | - Lakshmy Vasan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Bryan Kartono
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Kevan Clifford
- Institute of Medical Science, University of Toronto, Medical Sciences Building, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Centre for Addiction and Mental Health (CAMH), 250 College St., Toronto, ON M5T 1R8, Canada
| | - Ahmadreza Attarpour
- Department of Medical Biophysics, University of Toronto, 101 College St., Toronto, ON M5G 1L7, Canada
| | - Raghav Sharma
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Matthew Mandrozos
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Ain Kim
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Wenda Zhao
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Ari Belotserkovsky
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Claire Verkuyl
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Centre, 6th Floor, 60 Leonard Avenue, Toronto, ON M5T 0S8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Medical Sciences Building, 6th Floor, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
19
|
Maurer AC, Benyamini B, Fan VB, Whitney ON, Dailey GM, Darzacq X, Weitzman MD, Tjian R. Double-Strand Break Repair Pathways Differentially Affect Processing and Transduction by Dual AAV Vectors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.19.558438. [PMID: 37790316 PMCID: PMC10542147 DOI: 10.1101/2023.09.19.558438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Recombinant adeno-associated viral vectors (rAAV) are a powerful tool for gene delivery but have a limited DNA carrying capacity. Efforts to expand this genetic payload have focused on engineering the vector components, such as dual trans-splicing vectors which double the delivery size by exploiting the natural concatenation of rAAV genomes in host nuclei. We hypothesized that inefficient dual vector transduction could be improved by modulating host factors which affect concatenation. Since factors mediating concatenation are not well defined, we performed a genome-wide screen to identify host cell regulators. We discovered that Homologous Recombination (HR) is inhibitory to dual vector transduction. We demonstrate that depletion or inhibition of HR factors BRCA1 and Rad51 significantly increase reconstitution of a large split transgene by increasing both concatenation and expression from rAAVs. Our results define new roles for DNA damage repair in rAAV transduction and highlight the potential for pharmacological intervention to increase genetic payload of rAAV vectors.
Collapse
Affiliation(s)
- Anna C. Maurer
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- CIRM Center of Excellence, University of California, Berkeley, CA
| | - Brian Benyamini
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Vinson B. Fan
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Oscar N. Whitney
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Gina M. Dailey
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Li Ka Shing Center for Biomedical & Health Sciences, University of California, Berkeley, CA, USA
| | - Xavier Darzacq
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Li Ka Shing Center for Biomedical & Health Sciences, University of California, Berkeley, CA, USA
| | - Matthew D. Weitzman
- University of Pennsylvania Perelman School of Medicine and the Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Robert Tjian
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Li Ka Shing Center for Biomedical & Health Sciences, University of California, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
| |
Collapse
|
20
|
Albini S, Palmieri L, Dubois A, Bourg N, Lostal W, Richard I. Assessment of Therapeutic Potential of a Dual AAV Approach for Duchenne Muscular Dystrophy. Int J Mol Sci 2023; 24:11421. [PMID: 37511179 PMCID: PMC10380683 DOI: 10.3390/ijms241411421] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a yet incurable rare genetic disease that affects the skeletal and cardiac muscles, leading to progressive muscle wasting and premature death. DMD is caused by the lack of dystrophin, a muscle protein essential for the biochemical support and integrity of muscle fibers. Gene replacement strategies for Duchenne muscular dystrophy (DMD) employing the adeno-associated virus (AAV) face the challenge imposed by the limited packaging capacity of AAV, only allowing the accommodation of a short version of dystrophin (µDys) that is still far removed from correcting human disease. The need to develop strategies leading to the expression of a best performing dystrophin variant led to only few studies reporting on the use of dual vectors, but none reported on a method to assess in vivo transgene reconstitution efficiency, the degree of which directly affects the use of safe AAV dosing. We report here on the generation of a dual AAV vector approach for the expression of a larger dystrophin version (quasidystrophin) based on homologous recombination, and the development of a methodology employing a strategic droplet digital PCR design, to determine the recombination efficiency as well as the occurrence of unwanted concatemerization events or aberrant expression from the single vectors. We demonstrated that, upon systemic delivery in the dystrophic D2.B10-Dmdmdx/J (DBA2mdx) mice, our dual AAV approach led to high transgene reconstitution efficiency and negligible Inverted Terminal Repeats (ITR)-dependent concatemerization, with consequent remarkable protein restoration in muscles and improvement of muscle pathology. This evidence supports the suitability of our system for gene therapy application and the potential of this methodology to assess and improve the feasibility for therapeutic translation of multiple vector approaches.
Collapse
Affiliation(s)
- Sonia Albini
- Genethon, 91100 Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Laura Palmieri
- Genethon, 91100 Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Auriane Dubois
- Genethon, 91100 Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Nathalie Bourg
- Genethon, 91100 Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - William Lostal
- Genethon, 91100 Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Isabelle Richard
- Genethon, 91100 Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| |
Collapse
|
21
|
Barbon E, Kawecki C, Marmier S, Sakkal A, Collaud F, Charles S, Ronzitti G, Casari C, Christophe OD, Denis CV, Lenting PJ, Mingozzi F. Development of a dual hybrid AAV vector for endothelial-targeted expression of von Willebrand factor. Gene Ther 2023; 30:245-254. [PMID: 33456057 PMCID: PMC10113149 DOI: 10.1038/s41434-020-00218-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 11/06/2020] [Accepted: 12/30/2020] [Indexed: 12/30/2022]
Abstract
Von Willebrand disease (VWD), the most common inherited bleeding disorder in humans, is caused by quantitative or qualitative defects in von Willebrand factor (VWF). VWD represents a potential target for gene therapy applications, as a single treatment could potentially result in a long-term correction of the disease. In recent years, several liver-directed gene therapy approaches have been exploited for VWD, but their efficacy was generally limited by the large size of the VWF transgene and the reduced hemostatic activity of the protein produced from hepatocytes. In this context, we aimed at developing a gene therapy strategy for gene delivery into endothelial cells, the natural site of biosynthesis of VWF. We optimized an endothelial-specific dual hybrid AAV vector, in which the large VWF cDNA was put under the control of an endothelial promoter and correctly reconstituted upon cell transduction by a combination of trans-splicing and homologous recombination mechanisms. In addition, we modified the AAV vector capsid by introducing an endothelial-targeting peptide to improve the efficiency for endothelial-directed gene transfer. This vector platform allowed the reconstitution of full-length VWF transgene both in vitro in human umbilical vein endothelial cells and in vivo in VWD mice, resulting in long-term expression of VWF.
Collapse
Affiliation(s)
- Elena Barbon
- Généthon, 91000, Evry, France
- Université Paris-Saclay, Université Evry, INSERM, Généthon, Integrare research unit UMR_S951, 91000, Evry, France
| | - Charlotte Kawecki
- Laboratory of Hemostasis, Inflammation and Thrombosis, Institut National de la Santé et de la Recherche Médicale UMR_1176, Université Paris-Saclay, 94276, Le Kremlin-Bicêtre, France
| | - Solenne Marmier
- Généthon, 91000, Evry, France
- Université Paris-Saclay, Université Evry, INSERM, Généthon, Integrare research unit UMR_S951, 91000, Evry, France
| | - Aboud Sakkal
- Généthon, 91000, Evry, France
- Université Paris-Saclay, Université Evry, INSERM, Généthon, Integrare research unit UMR_S951, 91000, Evry, France
| | - Fanny Collaud
- Généthon, 91000, Evry, France
- Université Paris-Saclay, Université Evry, INSERM, Généthon, Integrare research unit UMR_S951, 91000, Evry, France
| | - Severine Charles
- Généthon, 91000, Evry, France
- Université Paris-Saclay, Université Evry, INSERM, Généthon, Integrare research unit UMR_S951, 91000, Evry, France
| | - Giuseppe Ronzitti
- Généthon, 91000, Evry, France
- Université Paris-Saclay, Université Evry, INSERM, Généthon, Integrare research unit UMR_S951, 91000, Evry, France
| | - Caterina Casari
- Laboratory of Hemostasis, Inflammation and Thrombosis, Institut National de la Santé et de la Recherche Médicale UMR_1176, Université Paris-Saclay, 94276, Le Kremlin-Bicêtre, France
| | - Olivier D Christophe
- Laboratory of Hemostasis, Inflammation and Thrombosis, Institut National de la Santé et de la Recherche Médicale UMR_1176, Université Paris-Saclay, 94276, Le Kremlin-Bicêtre, France
| | - Cécile V Denis
- Laboratory of Hemostasis, Inflammation and Thrombosis, Institut National de la Santé et de la Recherche Médicale UMR_1176, Université Paris-Saclay, 94276, Le Kremlin-Bicêtre, France
| | - Peter J Lenting
- Laboratory of Hemostasis, Inflammation and Thrombosis, Institut National de la Santé et de la Recherche Médicale UMR_1176, Université Paris-Saclay, 94276, Le Kremlin-Bicêtre, France
| | - Federico Mingozzi
- Généthon, 91000, Evry, France.
- Université Paris-Saclay, Université Evry, INSERM, Généthon, Integrare research unit UMR_S951, 91000, Evry, France.
- Spark Therapeutics, Philadelphia, PA, 19103, USA.
| |
Collapse
|
22
|
Shitik EM, Shalik IK, Yudkin DV. AAV- based vector improvements unrelated to capsid protein modification. Front Med (Lausanne) 2023; 10:1106085. [PMID: 36817775 PMCID: PMC9935841 DOI: 10.3389/fmed.2023.1106085] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) is the leading platform for delivering genetic constructs in vivo. To date, three AAV-based gene therapeutic agents have been approved by the FDA and are used in clinical practice. Despite the distinct advantages of gene therapy development, it is clear that AAV vectors need to be improved. Enhancements in viral vectors are mainly associated with capsid protein modifications. However, there are other structures that significantly affect the AAV life cycle and transduction. The Rep proteins, in combination with inverted terminal repeats (ITRs), determine viral genome replication, encapsidation, etc. Moreover, transgene cassette expression in recombinant variants is directly related to AAV production and transduction efficiency. This review discusses the ways to improve AAV vectors by modifying ITRs, a transgene cassette, and the Rep proteins.
Collapse
|
23
|
Hansen S, McClements ME, Corydon TJ, MacLaren RE. Future Perspectives of Prime Editing for the Treatment of Inherited Retinal Diseases. Cells 2023; 12:440. [PMID: 36766782 PMCID: PMC9913839 DOI: 10.3390/cells12030440] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Inherited retinal diseases (IRD) are a clinically and genetically heterogenous group of diseases and a leading cause of blindness in the working-age population. Even though gene augmentation therapies have shown promising results, they are only feasible to treat a small number of autosomal recessive IRDs, because the size of the gene is limited by the vector used. DNA editing however could potentially correct errors regardless of the overall size of the gene and might also be used to correct dominant mutations. Prime editing is a novel CRISPR/Cas9 based gene editing tool that enables precise correction of point mutations, insertions, and deletions without causing double strand DNA breaks. Due to its versatility and precision this technology may be a potential treatment option for virtually all genetic causes of IRD. Since its initial description, the prime editing technology has been further improved, resulting in higher efficacy and a larger target scope. Additionally, progress has been achieved concerning the size-related delivery issue of the prime editor components. This review aims to give an overview of these recent advancements and discusses prime editing as a potential treatment for IRDs.
Collapse
Affiliation(s)
- Silja Hansen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX3 9DU, UK
| | - Michelle E. McClements
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Thomas J. Corydon
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
- Department of Ophthalmology, Aarhus University Hospital, 8000 Aarhus, Denmark
| | - Robert E. MacLaren
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| |
Collapse
|
24
|
Pupo A, Fernández A, Low SH, François A, Suárez-Amarán L, Samulski RJ. AAV vectors: The Rubik's cube of human gene therapy. Mol Ther 2022; 30:3515-3541. [PMID: 36203359 PMCID: PMC9734031 DOI: 10.1016/j.ymthe.2022.09.015] [Citation(s) in RCA: 174] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 12/12/2022] Open
Abstract
Defective genes account for ∼80% of the total of more than 7,000 diseases known to date. Gene therapy brings the promise of a one-time treatment option that will fix the errors in patient genetic coding. Recombinant viruses are highly efficient vehicles for in vivo gene delivery. Adeno-associated virus (AAV) vectors offer unique advantages, such as tissue tropism, specificity in transduction, eliciting of a relatively low immune responses, no incorporation into the host chromosome, and long-lasting delivered gene expression, making them the most popular viral gene delivery system in clinical trials, with three AAV-based gene therapy drugs already approved by the US Food and Drug Administration (FDA) or European Medicines Agency (EMA). Despite the success of AAV vectors, their usage in particular scenarios is still limited due to remaining challenges, such as poor transduction efficiency in certain tissues, low organ specificity, pre-existing humoral immunity to AAV capsids, and vector dose-dependent toxicity in patients. In the present review, we address the different approaches to improve AAV vectors for gene therapy with a focus on AAV capsid selection and engineering, strategies to overcome anti-AAV immune response, and vector genome design, ending with a glimpse at vector production methods and the current state of recombinant AAV (rAAV) at the clinical level.
Collapse
Affiliation(s)
- Amaury Pupo
- R&D Department, Asklepios BioPharmaceutical, Inc. (AskBio), 20 T.W. Alexander, Suite 110 RTP, Durham, NC 27709, USA
| | - Audry Fernández
- R&D Department, Asklepios BioPharmaceutical, Inc. (AskBio), 20 T.W. Alexander, Suite 110 RTP, Durham, NC 27709, USA
| | - Siew Hui Low
- R&D Department, Asklepios BioPharmaceutical, Inc. (AskBio), 20 T.W. Alexander, Suite 110 RTP, Durham, NC 27709, USA
| | - Achille François
- Viralgen. Parque Tecnológico de Guipuzkoa, Edificio Kuatro, Paseo Mikeletegui, 83, 20009 San Sebastián, Spain
| | - Lester Suárez-Amarán
- R&D Department, Asklepios BioPharmaceutical, Inc. (AskBio), 20 T.W. Alexander, Suite 110 RTP, Durham, NC 27709, USA
| | - Richard Jude Samulski
- R&D Department, Asklepios BioPharmaceutical, Inc. (AskBio), 20 T.W. Alexander, Suite 110 RTP, Durham, NC 27709, USA,Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA,Corresponding author: Richard Jude Samulski, R&D Department, Asklepios BioPharmaceutical, Inc. (AskBio), 20 T.W. Alexander, Suite 110 RTP, NC 27709, USA.
| |
Collapse
|
25
|
Sarli SL, Watts JK. Harnessing nucleic acid technologies for human health on earth and in space. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:113-126. [PMID: 36336357 PMCID: PMC11845088 DOI: 10.1016/j.lssr.2022.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/01/2022] [Accepted: 08/18/2022] [Indexed: 06/16/2023]
Abstract
Nucleic acid therapeutics are a versatile class of sequence-programmable drugs that offer a robust and clinically viable strategy to modulate expression or correct genetic defects contributing to disease. The majority of drugs currently on the market target proteins; however, proteins only represent a subset of possible disease targets. Nucleic acid therapeutics allow intuitive engagement with genome sequences providing a more direct way to target many diseases at their genetic root cause. Their clinical success depends on platform technologies which can support durable and well tolerated pharmacological activity in a given tissue. Nucleic acid drugs possess a potent combination of target specificity and adaptability required to advance drug development for many diseases. As these therapeutic technologies mature, their clinical applications can also expand access to personalized therapies for patients with rare or solo genetic diseases. Spaceflight crew members exposed to the unique hazards of spaceflight, especially those related to galactic cosmic radiation (GCR) exposure, represent another patient subset who may also benefit from nucleic acid drugs as countermeasures. In this review, we will discuss the various classes of RNA- and DNA-targeted nucleic acid drugs, provide an overview of their present-day clinical applications, and describe major strategies to improve their delivery, safety, and overall efficacy.
Collapse
Affiliation(s)
- Samantha L Sarli
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jonathan K Watts
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
26
|
Yee T, Wert KJ. Base and Prime Editing in the Retina-From Preclinical Research toward Human Clinical Trials. Int J Mol Sci 2022; 23:12375. [PMID: 36293232 PMCID: PMC9604474 DOI: 10.3390/ijms232012375] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022] Open
Abstract
Inherited retinal diseases (IRDs) are a clinically and genetically heterogeneous group of diseases that are one of the leading causes of vision loss in young and aged individuals. IRDs are mainly caused by a loss of the post-mitotic photoreceptor neurons of the retina, or by the degeneration of the retinal pigment epithelium. Unfortunately, once these cells are damaged, it is irreversible and leads to permanent vision impairment. Thought to be previously incurable, gene therapy has been rapidly evolving to be a potential treatment to prevent further degeneration of the retina and preserve visual function. The development of clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) base and prime editors have increased the capabilities of the genome editing toolbox in recent years. Both base and prime editors evade the creation of double-stranded breaks in deoxyribonucleic acid (DNA) and the requirement of donor template of DNA for repair, which make them advantageous methods in developing clinical therapies. In addition, establishing a permanent edit within the genome could be better suited for patients with progressive degeneration. In this review, we will summarize published uses of successful base and prime editing in treating IRDs.
Collapse
Affiliation(s)
- Tiffany Yee
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Katherine J. Wert
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
27
|
Burdett T, Nuseibeh S. Changing trends in the development of AAV-based gene therapies: a meta-analysis of past and present therapies. Gene Ther 2022; 30:323-335. [PMID: 36089633 DOI: 10.1038/s41434-022-00363-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 08/01/2022] [Accepted: 08/26/2022] [Indexed: 11/09/2022]
Abstract
Gene therapy has seen a transformation from a proof-of-concept approach to a clinical reality over the past several decades, with adeno-associated virus (AAV)-mediated gene therapy emerging as the leading platform for in vivo gene transfer. A systematic review of AAV-based gene therapies in clinical development was conducted herein to determine why only a handful of AAV-based gene therapy products have achieved market approval. The indication to be treated, route of administration and vector design were investigated as critical factors and assessed for their impact on clinical safety and efficacy. A shift in recent years towards high-dose systemic administration for the treatment of metabolic, neurological and haematological diseases was identified, with intravenous administration demonstrating the highest efficacy and safety risks in clinical trials. Recent years have seen a decline in favour of traditional AAV serotypes and promoters, accompanied by an increase in favour and higher clinical success rate for novel capsids and tissue-specific promoters. Furthermore, a meta-analysis was performed to identify factors that may inhibit the translation of therapeutic efficacy from preclinical large animal studies to first-in-human clinical trials and a detrimental effect on clinical efficacy was associated with alterations to administration routes.
Collapse
|
28
|
Rovai A, Chung B, Hu Q, Hook S, Yuan Q, Kempf T, Schmidt F, Grimm D, Talbot SR, Steinbrück L, Götting J, Bohne J, Krooss SA, Ott M. In vivo adenine base editing reverts C282Y and improves iron metabolism in hemochromatosis mice. Nat Commun 2022; 13:5215. [PMID: 36064805 PMCID: PMC9445023 DOI: 10.1038/s41467-022-32906-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/24/2022] [Indexed: 11/22/2022] Open
Abstract
Hemochromatosis is one of the most common inherited metabolic diseases among white populations and predominantly originates from a homozygous C282Y mutation in the HFE gene. The G > A transition at position c.845 of the gene causes misfolding of the HFE protein, ultimately resulting in its absence at the cell membrane. Consequently, the lack of interaction with the transferrin receptors 1 and 2 leads to systemic iron overload. We screened potential gRNAs in a highly precise cell culture assay and applied an AAV8 split-vector expressing the adenine base editor ABE7.10 and our candidate gRNA in 129-Hfetm.1.1Nca mice. Here we show that a single injection of our therapeutic vector leads to a gene correction rate of >10% and improved iron metabolism in the liver. Our study presents a proof-of-concept for a targeted gene correction therapy for one of the most frequent hereditary diseases affecting humans.
Collapse
Affiliation(s)
- Alice Rovai
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- Twincore Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - BoMee Chung
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Qingluan Hu
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- Twincore Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Sebastian Hook
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- Twincore Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Qinggong Yuan
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- Twincore Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Tibor Kempf
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Florian Schmidt
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Cluster of Excellence CellNetworks, BioQuant BQ0030, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
- BioQuant, Center for Integrative Infectious Diseases Research (CIID), University of Heidelberg, Heidelberg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Cluster of Excellence CellNetworks, BioQuant BQ0030, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
- BioQuant, Center for Integrative Infectious Diseases Research (CIID), University of Heidelberg, Heidelberg, Germany
- German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Heidelberg, Germany
| | - Steven R Talbot
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Lars Steinbrück
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Jasper Götting
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Jens Bohne
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Simon A Krooss
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.
- Twincore Centre for Experimental and Clinical Infection Research, Hannover, Germany.
- Institute of Virology, Hannover Medical School, Hannover, Germany.
| | - Michael Ott
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.
- Twincore Centre for Experimental and Clinical Infection Research, Hannover, Germany.
| |
Collapse
|
29
|
Molecular analysis of AAV5-hFVIII-SQ vector-genome-processing kinetics in transduced mouse and nonhuman primate livers. Mol Ther Methods Clin Dev 2022; 24:142-153. [PMID: 35036471 PMCID: PMC8749450 DOI: 10.1016/j.omtm.2021.12.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/17/2021] [Indexed: 12/18/2022]
Abstract
Valoctocogene roxaparvovec (AAV5-hFVIII-SQ) is an adeno-associated virus serotype 5 (AAV5)-based gene therapy vector containing a B-domain-deleted human coagulation factor VIII (hFVIII) gene controlled by a liver-selective promoter. AAV5-hFVIII-SQ is currently under clinical investigation as a treatment for severe hemophilia A. The full-length AAV5-hFVIII-SQ is >4.9 kb, which is over the optimal packaging limit of AAV5. Following administration, the vector must undergo a number of genome-processing, assembly, and repair steps to form full-length circularized episomes that mediate long-term FVIII expression in target tissues. To understand the processing kinetics of the oversized AAV5-hFVIII-SQ vector genome into circular episomes, we characterized the various molecular forms of the AAV5-hFVIII-SQ genome at multiple time points up to 6 months postdose in the liver of murine and non-human primate models. Full-length circular episomes were detected in liver tissue beginning 1 week postdosing. Over 6 months, quantities of circular episomes (in a predominantly head-to-tail configuration) increased, while DNA species lacking inverted terminal repeats were preferentially degraded. Levels of duplex, circular, full-length genomes significantly correlated with levels of hFVIII-SQ RNA transcripts in mice and non-human primates dosed with AAV5-hFVIII-SQ. Altogether, we show that formation of full-length circular episomes in the liver following AAV5-hFVIII-SQ transduction was associated with long-term FVIII expression.
Collapse
|
30
|
Fakhiri J, Grimm D. Best of most possible worlds: Hybrid gene therapy vectors based on parvoviruses and heterologous viruses. Mol Ther 2021; 29:3359-3382. [PMID: 33831556 PMCID: PMC8636155 DOI: 10.1016/j.ymthe.2021.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 01/12/2021] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Parvoviruses and especially the adeno-associated virus (AAV) species provide an exciting and versatile platform for the rational design or molecular evolution of human gene-therapy vectors, documented by literature from over half a century, hundreds of clinical trials, and the recent commercialization of multiple AAV gene therapeutics. For the last three decades, the power of these vectors has been further potentiated through various types of hybrid vectors created by intra- or inter-genus juxtaposition of viral DNA and protein cis elements or by synergistic complementation of parvoviral features with those of heterologous, prokaryotic, or eukaryotic viruses. Here, we provide an overview of the history and promise of this rapidly expanding field of hybrid parvoviral gene-therapy vectors, starting with early generations of chimeric particles composed of a recombinant AAV genome encapsidated in shells of synthetic AAVs or of adeno-, herpes-, baculo-, or protoparvoviruses. We then dedicate our attention to two newer, highly promising types of hybrid vectors created via (1) pseudotyping of AAV genomes with bocaviral serotypes and capsid mutants or (2) packaging of AAV DNA into, or tethering of entire vector particles to, bacteriophages. Finally, we conclude with an outlook summarizing critical requirements and improvements toward clinical translation of these original concepts.
Collapse
Affiliation(s)
- Julia Fakhiri
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Heidelberg, Germany; BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Heidelberg, Germany; BioQuant, University of Heidelberg, Heidelberg, Germany; German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), Partner site Heidelberg, Heidelberg, Germany.
| |
Collapse
|
31
|
Wang Q, Liu J, Janssen JM, Tasca F, Mei H, Gonçalves MAFV. Broadening the reach and investigating the potential of prime editors through fully viral gene-deleted adenoviral vector delivery. Nucleic Acids Res 2021; 49:11986-12001. [PMID: 34669958 PMCID: PMC8599732 DOI: 10.1093/nar/gkab938] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022] Open
Abstract
Prime editing is a recent precision genome editing modality whose versatility offers the prospect for a wide range of applications, including the development of targeted genetic therapies. Yet, an outstanding bottleneck for its optimization and use concerns the difficulty in delivering large prime editing complexes into cells. Here, we demonstrate that packaging prime editing constructs in adenoviral capsids overcomes this constrain resulting in robust genome editing in both transformed and non-transformed human cells with up to 90% efficiencies. Using this cell cycle-independent delivery platform, we found a direct correlation between prime editing activity and cellular replication and disclose that the proportions between accurate prime editing events and unwanted byproducts can be influenced by the target-cell context. Hence, adenovector particles permit the efficacious delivery and testing of prime editing reagents in human cells independently of their transformation and replication statuses. The herein integrated gene delivery and gene editing technologies are expected to aid investigating the potential and limitations of prime editing in numerous experimental settings and, eventually, in ex vivo or in vivo therapeutic contexts.
Collapse
Affiliation(s)
- Qian Wang
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Jin Liu
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Josephine M Janssen
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Francesca Tasca
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Hailiang Mei
- Department of Biomedical Data Sciences, Sequencing Analysis Support Core, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Manuel A F V Gonçalves
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| |
Collapse
|
32
|
Martinez Velazquez LA, Ballios BG. The Next Generation of Molecular and Cellular Therapeutics for Inherited Retinal Disease. Int J Mol Sci 2021; 22:ijms222111542. [PMID: 34768969 PMCID: PMC8583900 DOI: 10.3390/ijms222111542] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/26/2022] Open
Abstract
Inherited retinal degenerations (IRDs) are a diverse group of conditions that are often characterized by the loss of photoreceptors and blindness. Recent innovations in molecular biology and genomics have allowed us to identify the causative defects behind these dystrophies and to design therapeutics that target specific mechanisms of retinal disease. Recently, the FDA approved the first in vivo gene therapy for one of these hereditary blinding conditions. Current clinical trials are exploring new therapies that could provide treatment for a growing number of retinal dystrophies. While the field has had early success with gene augmentation strategies for treating retinal disease based on loss-of-function mutations, many novel approaches hold the promise of offering therapies that span the full spectrum of causative mutations and mechanisms. Here, we provide a comprehensive review of the approaches currently in development including a discussion of retinal neuroprotection, gene therapies (gene augmentation, gene editing, RNA modification, optogenetics), and regenerative stem or precursor cell-based therapies. Our review focuses on technologies that are being developed for clinical translation or are in active clinical trials and discusses the advantages and limitations for each approach.
Collapse
Affiliation(s)
| | - Brian G. Ballios
- Department of Ophthalmology and Vision Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 3A9, Canada
- Correspondence:
| |
Collapse
|
33
|
Huang D, Heath Jeffery RC, Aung-Htut MT, McLenachan S, Fletcher S, Wilton SD, Chen FK. Stargardt disease and progress in therapeutic strategies. Ophthalmic Genet 2021; 43:1-26. [PMID: 34455905 DOI: 10.1080/13816810.2021.1966053] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: Stargardt disease (STGD1) is an autosomal recessive retinal dystrophy due to mutations in ABCA4, characterized by subretinal deposition of lipofuscin-like substances and bilateral centrifugal vision loss. Despite the tremendous progress made in the understanding of STGD1, there are no approved treatments to date. This review examines the challenges in the development of an effective STGD1 therapy.Materials and Methods: A literature review was performed through to June 2021 summarizing the spectrum of retinal phenotypes in STGD1, the molecular biology of ABCA4 protein, the in vivo and in vitro models used to investigate the mechanisms of ABCA4 mutations and current clinical trials.Results: STGD1 phenotypic variability remains an challenge for clinical trial design and patient selection. Pre-clinical development of therapeutic options has been limited by the lack of animal models reflecting the diverse phenotypic spectrum of STDG1. Patient-derived cell lines have facilitated the characterization of splice mutations but the clinical presentation is not always predicted by the effect of specific mutations on retinoid metabolism in cellular models. Current therapies primarily aim to delay vision loss whilst strategies to restore vision are less well developed.Conclusions: STGD1 therapy development can be accelerated by a deeper understanding of genotype-phenotype correlations.
Collapse
Affiliation(s)
- Di Huang
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Western Australia, Australia.,Centre for Ophthalmology and Visual Science (Incorporating Lions Eye Institute), the University of Western Australia, Nedlands, Western Australia, Australia.,Perron Institute for Neurological and Translational Science & the University of Western Australia, Nedlands, Western Australia, Australia
| | - Rachael C Heath Jeffery
- Centre for Ophthalmology and Visual Science (Incorporating Lions Eye Institute), the University of Western Australia, Nedlands, Western Australia, Australia
| | - May Thandar Aung-Htut
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Western Australia, Australia.,Perron Institute for Neurological and Translational Science & the University of Western Australia, Nedlands, Western Australia, Australia
| | - Samuel McLenachan
- Centre for Ophthalmology and Visual Science (Incorporating Lions Eye Institute), the University of Western Australia, Nedlands, Western Australia, Australia
| | - Sue Fletcher
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Western Australia, Australia.,Perron Institute for Neurological and Translational Science & the University of Western Australia, Nedlands, Western Australia, Australia
| | - Steve D Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Western Australia, Australia.,Perron Institute for Neurological and Translational Science & the University of Western Australia, Nedlands, Western Australia, Australia
| | - Fred K Chen
- Centre for Ophthalmology and Visual Science (Incorporating Lions Eye Institute), the University of Western Australia, Nedlands, Western Australia, Australia.,Australian Inherited Retinal Disease Registry and DNA Bank, Department of Medical Technology and Physics, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Department of Ophthalmology, Royal Perth Hospital, Perth, Western Australia, Australia.,Department of Ophthalmology, Perth Children's Hospital, Nedlands, Western Australia, Australia
| |
Collapse
|
34
|
Li YT, Wu HL, Liu CJ. Molecular Mechanisms and Animal Models of HBV-Related Hepatocellular Carcinoma: With Emphasis on Metastatic Tumor Antigen 1. Int J Mol Sci 2021; 22:9380. [PMID: 34502289 PMCID: PMC8431721 DOI: 10.3390/ijms22179380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/22/2021] [Accepted: 08/27/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is an important cause of cancer death worldwide, and hepatitis B virus (HBV) infection is a major etiology, particularly in the Asia-Pacific region. Lack of sensitive biomarkers for early diagnosis of HCC and lack of effective therapeutics for patients with advanced HCC are the main reasons for high HCC mortality; these clinical needs are linked to the molecular heterogeneity of hepatocarcinogenesis. Animal models are the basis of preclinical and translational research in HBV-related HCC (HBV-HCC). Recent advances in methodology have allowed the development of several animal models to address various aspects of chronic liver disease, including HCC, which HBV causes in humans. Currently, multiple HBV-HCC animal models, including conventional, hydrodynamics-transfection-based, viral vector-mediated transgenic, and xenograft mice models, as well as the hepadnavirus-infected tree shrew and woodchuck models, are available. This review provides an overview of molecular mechanisms and animal models of HBV-HCC. Additionally, the metastatic tumor antigen 1 (MTA1), a cancer-promoting molecule, was introduced as an example to address the importance of a suitable animal model for studying HBV-related hepatocarcinogenesis.
Collapse
Affiliation(s)
- Yung-Tsung Li
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Hepatitis Research Center, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Hui-Lin Wu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Hepatitis Research Center, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chun-Jen Liu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Hepatitis Research Center, National Taiwan University Hospital, Taipei 100, Taiwan
| |
Collapse
|
35
|
Bisgin A, Sanlioglu AD, Eksi YE, Griffith TS, Sanlioglu S. Current Update on Severe Acute Respiratory Syndrome Coronavirus 2 Vaccine Development with a Special Emphasis on Gene Therapy Viral Vector Design and Construction for Vaccination. Hum Gene Ther 2021; 32:541-562. [PMID: 33858231 DOI: 10.1089/hum.2021.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Severe acute respiratory syndrome (SARS) is a newly emerging infectious disease (COVID-19) caused by the novel coronavirus SARS-coronavirus 2 (CoV-2). To combat the devastating spread of SARS-CoV-2, extraordinary efforts from numerous laboratories have focused on the development of effective and safe vaccines. Traditional live-attenuated or inactivated viral vaccines are not recommended for immunocompromised patients as the attenuated virus can still cause disease via phenotypic or genotypic reversion. Subunit vaccines require repeated dosing and adjuvant use to be effective, and DNA vaccines exhibit lower immune responses. mRNA vaccines can be highly unstable under physiological conditions. On the contrary, naturally antigenic viral vectors with well-characterized structure and safety profile serve as among the most effective gene carriers to provoke immune response via heterologous gene transfer. Viral vector-based vaccines induce both an effective cellular immune response and a humoral immune response owing to their natural adjuvant properties via transduction of immune cells. Consequently, viral vectored vaccines carrying the SARS-CoV-2 spike protein have recently been generated and successfully used to activate cytotoxic T cells and develop a neutralizing antibody response. Recent progress in SARS-CoV-2 vaccines, with an emphasis on gene therapy viral vector-based vaccine development, is discussed in this review.
Collapse
Affiliation(s)
- Atil Bisgin
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
- Department of Medical Genetics, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Ahter D Sanlioglu
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Yunus Emre Eksi
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Thomas S Griffith
- The Department of Urology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Salih Sanlioglu
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
36
|
Trapani I, Tornabene P, Auricchio A. Large gene delivery to the retina with AAV vectors: are we there yet? Gene Ther 2021; 28:220-222. [PMID: 32661283 DOI: 10.1038/s41434-020-0174-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/23/2020] [Accepted: 07/01/2020] [Indexed: 12/26/2022]
Affiliation(s)
- Ivana Trapani
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Medical Genetics, Department of Translational Medicine, Federico II University, Naples, Italy
| | - Patrizia Tornabene
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Medical Genetics, Department of Translational Medicine, Federico II University, Naples, Italy
| | - Alberto Auricchio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.
- Department of Advanced Biomedicine, Federico II University, Naples, Italy.
| |
Collapse
|
37
|
Ku CA, Pennesi ME. The new landscape of retinal gene therapy. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2020; 184:846-859. [PMID: 32888388 DOI: 10.1002/ajmg.c.31842] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 12/15/2022]
Abstract
Novel therapeutics for inherited retinal dystrophies (IRDs) have rapidly evolved since groundbreaking clinical trials for LCA due to RPE65 mutations led to the first FDA-approved in vivo gene therapy. Since then, advancements in viral vectors have led to more efficient AAV transduction and developed other viral vectors for gene augmentation therapy of large gene targets. Furthermore, significant developments in gene editing and RNA modulation technologies have introduced novel capabilities for treatment of autosomal dominant diseases, intronic mutations, and/or large genes otherwise unable to be treated with current viral vectors. We highlight strategies currently being evaluated in gene therapy clinical trials and promising preclinical developments for IRDs.
Collapse
Affiliation(s)
- Cristy A Ku
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Mark E Pennesi
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
38
|
Brommel CM, Cooney AL, Sinn PL. Adeno-Associated Virus-Based Gene Therapy for Lifelong Correction of Genetic Disease. Hum Gene Ther 2020; 31:985-995. [PMID: 32718227 PMCID: PMC7495917 DOI: 10.1089/hum.2020.138] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/27/2020] [Indexed: 12/27/2022] Open
Abstract
The list of successful gene therapy trials using adeno-associated virus (AAV)-based vectors continues to grow and includes a wide range of monogenic diseases. Replication incompetent AAV genomes typically remain episomal and expression dilutes as cells divide and die. Consequently, long-term transgene expression from AAV is best suited for quiescent cell types, such as retinal cells, myocytes, or neurons. For genetic diseases that involve cells with steady turnover, AAV-conferred correction may require routine readministration, where every dose carries the risk of developing an adaptive immune response that renders treatment ineffective. Here, we discuss innovative approaches to permanently modify the host genome using AAV-based platforms, thus potentially requiring only a single dose. Such approaches include using AAV delivery of DNA transposons, homologous recombination templates into safe harbors, and nucleases for targeting integration. In tissues with continual cell turnover, genetic modification of progenitor cell populations will help ensure persistent therapeutic outcomes. Combining the safety profile of AAV-based gene therapy vectors with the ability to integrate a therapeutic transgene creates novel solutions to the challenge of lifelong curative treatments for human genetic diseases.
Collapse
Affiliation(s)
| | - Ashley L. Cooney
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| | - Patrick L. Sinn
- Program in Molecular Medicine, University of Iowa, Iowa City, Iowa, USA
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
39
|
Reisinger E. Dual-AAV delivery of large gene sequences to the inner ear. Hear Res 2020; 394:107857. [DOI: 10.1016/j.heares.2019.107857] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/22/2019] [Indexed: 01/06/2023]
|
40
|
Akil O. Dual and triple AAV delivery of large therapeutic gene sequences into the inner ear. Hear Res 2020; 394:107912. [DOI: 10.1016/j.heares.2020.107912] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/04/2020] [Accepted: 02/07/2020] [Indexed: 12/17/2022]
|
41
|
Kariyawasam D, Alexander IE, Kurian M, Farrar MA. Great expectations: virus-mediated gene therapy in neurological disorders. J Neurol Neurosurg Psychiatry 2020; 91:849-860. [PMID: 32503884 DOI: 10.1136/jnnp-2019-322327] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 12/25/2022]
Abstract
Gene therapy (GT) has tremendous potential for the treatment of neurological disorders to transform patient care. The successful application of virus-mediated GT to treat spinal muscular atrophy is a significant milestone, serving to accelerate similar progress in a spectrum of neurological conditions, with more than 50 clinical trials currently underway, across neurodevelopmental, neurodegenerative, muscular dystrophy, epilepsy, chronic pain and neoplastic diseases. This review provides an overview of the key features of virus-mediated GT, paradigms of delivery and dosing, potential risks and highlights ongoing research to optimise safe and effective delivery of vectors into the nervous system. Examples of the application of GT in various neurological diseases alongside clinical development challenges will be presented. As the development and translation of GTs gain pace, success can only ultimately be realised for patients following implementation in the health system. The challenges and controversies of daunting costs, ethics, early diagnosis and health system readiness will require innovative pricing schemes, regulatory policies, education and organisation of a skilled workforce to deliver of high-quality care in clinical practice as we prepare for advanced therapeutics in neurology.
Collapse
Affiliation(s)
- Didu Kariyawasam
- Neurology, Sydney Children's Hospital Randwick, Randwick, New South Wales, Australia.,School of Women's and Children's Health, UNSW Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Ian E Alexander
- Discipline of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Gene Therapy Unit, Children's Medical Research Institute, Westmead, New South Wales, Australia
| | - Manju Kurian
- Neurosciences Unit, Institute of Child Health, University College London, London, UK.,Neurology, Great Ormond Street Hospital for Children, London, UK
| | - Michelle Anne Farrar
- Neurology, Sydney Children's Hospital Randwick, Randwick, New South Wales, Australia .,School of Women's and Children's Health, UNSW Medicine, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
42
|
French LS, Mellough CB, Chen FK, Carvalho LS. A Review of Gene, Drug and Cell-Based Therapies for Usher Syndrome. Front Cell Neurosci 2020; 14:183. [PMID: 32733204 PMCID: PMC7363968 DOI: 10.3389/fncel.2020.00183] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022] Open
Abstract
Usher syndrome is a genetic disorder causing neurosensory hearing loss and blindness from retinitis pigmentosa (RP). Adaptive techniques such as braille, digital and optical magnifiers, mobility training, cochlear implants, or other assistive listening devices are indispensable for reducing disability. However, there is currently no treatment to reduce or arrest sensory cell degeneration. There are several classes of treatments for Usher syndrome being investigated. The present article reviews the progress this research has made towards delivering commercial options for patients with Usher syndrome.
Collapse
Affiliation(s)
- Lucy S French
- Centre for Ophthalmology and Visual Sciences (incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia
| | - Carla B Mellough
- Centre for Ophthalmology and Visual Sciences (incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia
| | - Fred K Chen
- Centre for Ophthalmology and Visual Sciences (incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia.,Department of Ophthalmology, Royal Perth Hospital, Perth, WA, Australia.,Department of Ophthalmology, Perth Children's Hospital, Nedlands, WA, Australia
| | - Livia S Carvalho
- Centre for Ophthalmology and Visual Sciences (incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
43
|
Tornabene P, Trapani I, Minopoli R, Centrulo M, Lupo M, de Simone S, Tiberi P, Dell'Aquila F, Marrocco E, Iodice C, Iuliano A, Gesualdo C, Rossi S, Giaquinto L, Albert S, Hoyng CB, Polishchuk E, Cremers FPM, Surace EM, Simonelli F, De Matteis MA, Polishchuk R, Auricchio A. Intein-mediated protein trans-splicing expands adeno-associated virus transfer capacity in the retina. Sci Transl Med 2020; 11:11/492/eaav4523. [PMID: 31092694 DOI: 10.1126/scitranslmed.aav4523] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/21/2018] [Accepted: 04/04/2019] [Indexed: 12/26/2022]
Abstract
Retinal gene therapy with adeno-associated viral (AAV) vectors holds promises for treating inherited and noninherited diseases of the eye. Although clinical data suggest that retinal gene therapy is safe and effective, delivery of large genes is hindered by the limited AAV cargo capacity. Protein trans-splicing mediated by split inteins is used by single-cell organisms to reconstitute proteins. Here, we show that delivery of multiple AAV vectors each encoding one of the fragments of target proteins flanked by short split inteins results in protein trans-splicing and full-length protein reconstitution in the retina of mice and pigs and in human retinal organoids. The reconstitution of large therapeutic proteins using this approach improved the phenotype of two mouse models of inherited retinal diseases. Our data support the use of split intein-mediated protein trans-splicing in combination with AAV subretinal delivery for gene therapy of inherited blindness due to mutations in large genes.
Collapse
Affiliation(s)
- Patrizia Tornabene
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Ivana Trapani
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy.,Medical Genetics, Department of Translational Medicine, Federico II University, 80131 Naples, Italy
| | - Renato Minopoli
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Miriam Centrulo
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Mariangela Lupo
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Sonia de Simone
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Paola Tiberi
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Fabio Dell'Aquila
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Elena Marrocco
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Carolina Iodice
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Antonella Iuliano
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Carlo Gesualdo
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy
| | - Settimio Rossi
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy
| | - Laura Giaquinto
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Silvia Albert
- Department of Human Genetics and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 Nijmegen, Netherlands
| | - Carel B Hoyng
- Department of Ophthalmology and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 Nijmegen, Netherlands
| | - Elena Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Frans P M Cremers
- Department of Ophthalmology and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 Nijmegen, Netherlands
| | - Enrico M Surace
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy.,Medical Genetics, Department of Translational Medicine, Federico II University, 80131 Naples, Italy
| | - Francesca Simonelli
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy
| | - Maria A De Matteis
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy.,Department of Molecular Medicine and Medical Biotechnology, Federico II University, 80131 Naples, Italy
| | - Roman Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Alberto Auricchio
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy. .,Department of Advanced Biomedicine, Federico II University, 80131 Naples, Italy
| |
Collapse
|
44
|
Hair Cell Transduction Efficiency of Single- and Dual-AAV Serotypes in Adult Murine Cochleae. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:1167-1177. [PMID: 32518805 PMCID: PMC7270144 DOI: 10.1016/j.omtm.2020.05.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/07/2020] [Indexed: 01/03/2023]
Abstract
Gene delivery is a key component for the treatment of genetic hearing loss. To date, a myriad of adeno-associated virus (AAV) serotypes and surgical approaches have been employed to deliver transgenes to cochlear hair cells, but the efficacy of dual transduction remains unclear. Herein, we investigated cellular tropism of single injections of AAV serotype 1 (AAV1), AAV2, AAV8, AAV9, and Anc80L65, and quantitated dual-vector co-transduction rates following co-injection of AAV2 and AAV9 vectors in adult murine cochlea. We used the combined round window membrane and canal fenestration (RWM+CF) injection technique for vector delivery. Single AAV2 injections were most robust and transduced 96.7% ± 1.1% of inner hair cells (IHCs) and 83.9% ± 2.0% of outer hair cells (OHCs) throughout the cochlea without causing hearing impairment or hair cell loss. Dual AAV2 injection co-transduced 96.9% ± 1.7% of IHCs and 65.6% ± 8.95% of OHCs. Together, RWM+CF-injected single or dual AAV2 provides the highest auditory hair cell transduction efficiency of the AAV serotypes we studied. These findings broaden the application of cochlear gene therapy targeting hair cells.
Collapse
|
45
|
Chemello F, Bassel-Duby R, Olson EN. Correction of muscular dystrophies by CRISPR gene editing. J Clin Invest 2020; 130:2766-2776. [PMID: 32478678 PMCID: PMC7259998 DOI: 10.1172/jci136873] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Muscular dystrophies are debilitating disorders that result in progressive weakness and degeneration of skeletal muscle. Although the genetic mutations and clinical abnormalities of a variety of neuromuscular diseases are well known, no curative therapies have been developed to date. The advent of genome editing technology provides new opportunities to correct the underlying mutations responsible for many monogenic neuromuscular diseases. For example, Duchenne muscular dystrophy, which is caused by mutations in the dystrophin gene, has been successfully corrected in mice, dogs, and human cells through CRISPR/Cas9 editing. In this Review, we focus on the potential for, and challenges of, correcting muscular dystrophies by editing disease-causing mutations at the genomic level. Ideally, because muscle tissues are extremely long-lived, CRISPR technology could offer a one-time treatment for muscular dystrophies by correcting the culprit genomic mutations and enabling normal expression of the repaired gene.
Collapse
|
46
|
Maurer AC, Weitzman MD. Adeno-Associated Virus Genome Interactions Important for Vector Production and Transduction. Hum Gene Ther 2020; 31:499-511. [PMID: 32303138 PMCID: PMC7232694 DOI: 10.1089/hum.2020.069] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022] Open
Abstract
Recombinant adeno-associated virus has emerged as one of the most promising gene therapy delivery vectors. Development of these vectors took advantage of key features of the wild-type adeno-associated virus (AAV), enabled by basic studies of the underlying biology and requirements for transcription, replication, and packaging of the viral genome. Each step in generating and utilizing viral vectors involves numerous molecular interactions that together determine the efficiency of vector production and gene delivery. Once delivered into the cell, interactions with host proteins will determine the fate of the viral genome, and these will impact the intended goal of gene delivery. Here, we provide an overview of known interactions of the AAV genome with viral and cellular proteins involved in its amplification, packaging, and expression. Further appreciation of how the AAV genome interacts with host factors will enhance how this simple virus can be harnessed for an array of vector purposes that benefit human health.
Collapse
Affiliation(s)
- Anna C. Maurer
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | - Matthew D. Weitzman
- Division of Protective Immunity, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
47
|
Maguire CA, Corey DP. Viral vectors for gene delivery to the inner ear. Hear Res 2020; 394:107927. [PMID: 32199720 DOI: 10.1016/j.heares.2020.107927] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 02/04/2023]
Abstract
Gene therapy using virus vectors to treat hereditary diseases has made remarkable progress in the past decade. There are FDA-approved products for ex-vivo gene therapy for diseases such as immunodeficiencies (e.g., SCID), and in vivo gene therapy for a rare blindness and neuro-muscular disease. Gene therapy for hereditary hearing loss has picked up pace in the past five years due to progress in understanding disease gene function as well as the development of better technologies such as adeno-associated virus (AAV) vectors, to deliver nucleic acid to target cells in the inner ear. This review has two major goals. One is to review the state of the art for investigators already working in preclinical cochlear gene therapy. The other is to present the language of vectorology and important considerations for designing and using AAV vectors to inner ear neurobiologists who might use AAV vectors in the cochlea for either therapeutic or basic biological applications.
Collapse
Affiliation(s)
- Casey A Maguire
- Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, 149 13th Street, Charlestown, MA, 02114, USA; Department of Neurology, Harvard Medical School, Boston, MA, 02115, USA.
| | - David P Corey
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
48
|
Tornabene P, Trapani I. Can Adeno-Associated Viral Vectors Deliver Effectively Large Genes? Hum Gene Ther 2020; 31:47-56. [DOI: 10.1089/hum.2019.220] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Patrizia Tornabene
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Medical Genetics, Department of Translational Medicine, Federico II University, Naples, Italy
| | - Ivana Trapani
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Medical Genetics, Department of Translational Medicine, Federico II University, Naples, Italy
| |
Collapse
|
49
|
Wilmott P, Lisowski L, Alexander IE, Logan GJ. A User's Guide to the Inverted Terminal Repeats of Adeno-Associated Virus. Hum Gene Ther Methods 2019; 30:206-213. [DOI: 10.1089/hgtb.2019.276] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Patrick Wilmott
- Translational Vectorology Group, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Leszek Lisowski
- Translational Vectorology Group, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
- Military Institute of Hygiene and Epidemiology, The Biological Threats Identification and Countermeasure Centre, Puławy, Poland
- Vector and Genome Engineering Facility; Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Ian E. Alexander
- Discipline of Child and Adolescent Health, University of Sydney, Westmead, Australia
- Gene Therapy Research Unit, Children's Medical Research Institute and Sydney Children's Hospitals Network, University of Sydney, Westmead, Australia
| | - Grant J. Logan
- Gene Therapy Research Unit, Children's Medical Research Institute and Sydney Children's Hospitals Network, University of Sydney, Westmead, Australia
| |
Collapse
|
50
|
Haggerty DL, Grecco GG, Reeves KC, Atwood B. Adeno-Associated Viral Vectors in Neuroscience Research. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 17:69-82. [PMID: 31890742 PMCID: PMC6931098 DOI: 10.1016/j.omtm.2019.11.012] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adeno-associated viral vectors (AAVs) are increasingly useful preclinical tools in neuroscience research studies for interrogating cellular and neurocircuit functions and mapping brain connectivity. Clinically, AAVs are showing increasing promise as viable candidates for treating multiple neurological diseases. Here, we briefly review the utility of AAVs in mapping neurocircuits, manipulating neuronal function and gene expression, and activity labeling in preclinical research studies as well as AAV-based gene therapies for diseases of the nervous system. This review highlights the vast potential that AAVs have for transformative research and therapeutics in the neurosciences.
Collapse
Affiliation(s)
- David L. Haggerty
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gregory G. Grecco
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kaitlin C. Reeves
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brady Atwood
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
- Corresponding author: Brady Atwood, PhD, Department of Pharmacology & Toxicology, Indiana University School of Medicine, 320 West 15th Street, NB-400C, Indianapolis, IN 46202, USA.
| |
Collapse
|