1
|
Moyo B, Brown LBC, Khondaker II, Bao G. Engineering adeno-associated viral vectors for CRISPR/Cas based in vivo therapeutic genome editing. Biomaterials 2025; 321:123314. [PMID: 40203649 DOI: 10.1016/j.biomaterials.2025.123314] [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: 12/23/2024] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
The recent approval of the first gene editing therapy for sickle cell disease and transfusion-dependent beta-thalassemia by the U.S. Food and Drug Administration (FDA) demonstrates the immense potential of CRISPR (clustered regularly interspaced short palindromic repeats) technologies to treat patients with genetic disorders that were previously considered incurable. While significant advancements have been made with ex vivo gene editing approaches, the development of in vivo CRISPR/Cas gene editing therapies has not progressed as rapidly due to significant challenges in achieving highly efficient and specific in vivo delivery. Adeno-associated viral (AAV) vectors have shown great promise in clinical trials as vehicles for delivering therapeutic transgenes and other cargos but currently face multiple limitations for effective delivery of gene editing machineries. This review elucidates these challenges and highlights the latest engineering strategies aimed at improving the efficiency, specificity, and safety profiles of AAV-packaged CRISPR/Cas systems (AAV-CRISPR) to enhance their clinical utility.
Collapse
Affiliation(s)
- Buhle Moyo
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Lucas B C Brown
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA; Graduate Program in Systems, Synthetic, and Physical Biology, Rice University, Houston, TX, 77030, USA
| | - Ishika I Khondaker
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
Vo K, Shila S, Sharma Y, Pei GJ, Rosales CY, Dahiya V, Fields PE, Rumi MAK. Detection of mRNA Transcript Variants. Genes (Basel) 2025; 16:343. [PMID: 40149494 PMCID: PMC11942493 DOI: 10.3390/genes16030343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Most eukaryotic genes express more than one mature mRNA, defined as transcript variants. This complex phenomenon arises from various mechanisms, such as using alternative transcription start sites and alternative post-transcriptional processing events. The resulting transcript variants can lead to synthesizing proteins that possess distinct functional domains or may even generate noncoding RNAs, each with unique roles in cellular processes. The generation of these transcript variants is not merely a random occurrence; it is cell-type specific and varies with developmental stages, aging processes, or pathogenesis of diseases. This highlights the biological significance of transcript variants in regulating gene expression and their potential impact on cellular functionality. Despite the biological importance, investigating transcript variants has been hampered by challenges associated with detecting their expression. This review article addresses the advancements in molecular techniques in detecting transcript variants. Traditional methods such as RT-PCR and RT-qPCR can easily detect known transcript variants using primers that target unique exons associated with the variants. Other techniques like RACE-PCR and hybridization-based methods, including Northern blotting, RNase protection assays, and microarrays, have also been utilized to detect transcript variants. Nevertheless, RNA sequencing (RNA-Seq) has emerged as a powerful technique for identifying transcript variants, especially those with previously unknown sequences. The effectiveness of RNA sequencing in transcript variant detection depends on the specific sequencing approach and the precision of data analysis. By understanding the strengths and weaknesses of each laboratory technique, researchers can develop more effective strategies for detecting mRNA transcript variants. This ability will be crucial for our comprehensive understanding of gene regulation and the implications of transcript diversity in various biological contexts.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - M. A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (K.V.); (S.S.); (Y.S.); (G.J.P.); (C.Y.R.); (V.D.); (P.E.F.)
| |
Collapse
|
3
|
Holzwarth D, Calaminus G, Friese J, Sejersen T, Büning H, John-Neek P, Bastone AL, Rothe M, Mansfield K, Libertini S, Dubost V, Kuzmiski B, Alecu I, Labik I, Kirschner J. Pilocytic astrocytoma in a child with spinal muscular atrophy treated with onasemnogene abeparvovec. Mol Ther 2025:S1525-0016(25)00115-7. [PMID: 39955617 DOI: 10.1016/j.ymthe.2025.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/21/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025] Open
Abstract
Spinal muscular atrophy (SMA) is a severe neuromuscular disease, leading to progressive muscle weakness and potentially early mortality if untreated. Onasemnogene abeparvovec is a recombinant adeno-associated virus serotype 9 (rAAV9)-based gene therapy that has demonstrated improvements in survival and motor function for SMA patients. Here, we present a case of a patient diagnosed with a grade 1 pilocytic astrocytoma at the age of 2 years, approximately 8 months after onasemnogene abeparvovec treatment. Although vector genomes delivered by rAAVs persist primarily as episomes, rare integration events have been linked to tumor formation in neonate murine models. Therefore, we investigated the presence and possible integration of onasemnogene abeparvovec in formalin-fixed paraffin embedded (FFPE) and frozen tumor samples. In situ hybridization demonstrated variable transduction levels in individual tumor cells, while droplet digital PCR measured an average vector copy number ranging from 0.7 to 4.9 vector genomes/diploid genome. Integration site analysis identified a low number of integration sites that were not conserved between technical replicates, nor between FFPE and frozen samples, indicating that cells hosting integrating vector genomes represented a minority in the overall cell population. Thus, molecular analysis of the tumor tissue suggests that tumorigenesis was causally independent of the administration of onasemnogene abeparvovec.
Collapse
Affiliation(s)
| | | | | | - Thomas Sejersen
- Pediatric Neurology, Karolinska University Hospital, 171 77 Stockholm, Sweden; Department of Women's and Children's Health, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Philipp John-Neek
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Michael Rothe
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | | | | | | | | | - Iulian Alecu
- Novartis Pharmaceuticals, 4056 Basel, Switzerland
| | - Ivan Labik
- ProtaGene CGT GmbH, 69120 Heidelberg, Germany
| | - Janbernd Kirschner
- Department for Neuropediatrics and Muscle Disease, Medical Center - University of Freiburg, Faculty of Medicine, 79106 Freiburg, Germany
| |
Collapse
|
4
|
De Giorgi M, Park SH, Castoreno A, Cao M, Hurley A, Saxena L, Chuecos MA, Walkey CJ, Doerfler AM, Furgurson MN, Ljungberg MC, Patel KR, Hyde S, Chickering T, Lefebvre S, Wassarman K, Miller P, Qin J, Schlegel MK, Zlatev I, Han J, Beeton C, Li RG, Kim J, Martin JF, Bissig KD, Jadhav V, Bao G, Lagor WR. In vivo expansion of gene-targeted hepatocytes through transient inhibition of an essential gene. Sci Transl Med 2025; 17:eadk3920. [PMID: 39937884 DOI: 10.1126/scitranslmed.adk3920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 07/29/2024] [Accepted: 01/17/2025] [Indexed: 02/14/2025]
Abstract
Homology-directed repair (HDR)-based genome editing is an approach that could permanently correct a broad range of genetic diseases. However, its utility is limited by inefficient and imprecise DNA repair mechanisms in terminally differentiated tissues. Here, we tested Repair Drive, a platform technology for selectively expanding HDR-corrected hepatocytes in adult mice in vivo. Repair Drive involves transient conditioning of the liver by knocking down an essential gene, fumarylacetoacetate hydrolase (Fah), and delivering an untargetable version of the essential gene in cis with a therapeutic transgene. We show that Repair Drive increased the percentage of correctly targeted hepatocytes in healthy wild-type mice up to 25%, which resulted in a fivefold increased expression of a therapeutic transgene, human factor IX (FIX). Repair Drive was well tolerated and did not induce toxicity or tumorigenesis during a 1-year follow-up. This approach may broaden the range of liver diseases that can be treated with somatic genome editing.
Collapse
Affiliation(s)
- Marco De Giorgi
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - So Hyun Park
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | | | - Mingming Cao
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Ayrea Hurley
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lavanya Saxena
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Marcel A Chuecos
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Christopher J Walkey
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexandria M Doerfler
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mia N Furgurson
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - M Cecilia Ljungberg
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Kalyani R Patel
- Department of Pathology, Texas Children's Hospital, Houston, TX 77030, USA
| | - Sarah Hyde
- Alnylam Pharmaceuticals Inc., Cambridge, MA 02142, USA
| | | | | | | | | | - June Qin
- Alnylam Pharmaceuticals Inc., Cambridge, MA 02142, USA
| | | | - Ivan Zlatev
- Alnylam Pharmaceuticals Inc., Cambridge, MA 02142, USA
| | - Jun Han
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
- UVic-GBC Proteomics Centre, Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Christine Beeton
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rich Gang Li
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Heart Institute, Houston, TX 77030, USA
| | - Jong Kim
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Heart Institute, Houston, TX 77030, USA
| | - James F Martin
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Heart Institute, Houston, TX 77030, USA
| | - Karl-Dimiter Bissig
- Department of Pediatrics, Alice and Y. T. Chen Center for Genetics and Genomics, Division of Medical Genetics, Duke University, Durham, NC 27710, USA
| | - Vasant Jadhav
- Alnylam Pharmaceuticals Inc., Cambridge, MA 02142, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - William R Lagor
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Zwi-Dantsis L, Mohamed S, Massaro G, Moeendarbary E. Adeno-Associated Virus Vectors: Principles, Practices, and Prospects in Gene Therapy. Viruses 2025; 17:239. [PMID: 40006994 PMCID: PMC11861813 DOI: 10.3390/v17020239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Gene therapy offers promising potential as an efficacious and long-lasting therapeutic option for genetic conditions, by correcting defective mutations using engineered vectors to deliver genetic material to host cells. Among these vectors, adeno-associated viruses (AAVs) stand out for their efficiency, versatility, and safety, making them one of the leading platforms in gene therapy. The enormous potential of AAVs has been demonstrated through their use in over 225 clinical trials and the FDA's approval of six AAV-based gene therapy products, positioning these vectors at the forefront of the field. This review highlights the evolution and current applications of AAVs in gene therapy, focusing on their clinical successes, ongoing developments, and the manufacturing processes required for the rapid commercial growth anticipated in the AAV therapy market. It also discusses the broader implications of these advancements for future therapeutic strategies targeting more complex and multi-systemic conditions and biological processes such as aging. Finally, we explore some of the major challenges currently confronting the field.
Collapse
Affiliation(s)
- Limor Zwi-Dantsis
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| | - Saira Mohamed
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| | - Giulia Massaro
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Emad Moeendarbary
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| |
Collapse
|
6
|
Jaiswal R, Braud B, Hernandez-Ramirez K, Santosh V, Washington A, Escalante C. Cryo-EM structure of AAV2 Rep68 bound to integration site AAVS1: insights into the mechanism of DNA melting. Nucleic Acids Res 2025; 53:gkaf033. [PMID: 39883011 PMCID: PMC11780844 DOI: 10.1093/nar/gkaf033] [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: 06/13/2024] [Revised: 12/18/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025] Open
Abstract
The Rep68 protein from Adeno-Associated Virus (AAV) is a multifunctional SF3 helicase that performs most of the DNA transactions necessary for the viral life cycle. During AAV DNA replication, Rep68 assembles at the origin of replication, catalyzing the DNA melting and nicking reactions during the hairpin rolling replication process to complete the second-strand synthesis of the AAV genome. We report the cryo-electron microscopy structures of Rep68 bound to the adeno-associated virus integration site 1 in different nucleotide-bound states. In the nucleotide-free state, Rep68 forms a heptameric complex around DNA, with three origin-binding domains (OBDs) bound to the Rep-binding element sequence, while three remaining OBDs form transient dimers with them. The AAA+ domains form an open ring without interactions between subunits and DNA. We hypothesize that the heptameric structure is crucial for loading Rep68 onto double-stranded DNA. The ATPγS complex shows that only three subunits associate with the nucleotide, leading to a conformational change that promotes the formation of both intersubunit and DNA interactions. Moreover, three phenylalanine residues in the AAA+ domain induce a steric distortion in the DNA. Our study provides insights into how an SF3 helicase assembles on DNA and provides insights into the DNA melting process.
Collapse
Affiliation(s)
- Rahul Jaiswal
- Department of Physiology and Biophysics, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, United States
| | - Brandon Braud
- Department of Physiology and Biophysics, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, United States
| | - Karen C Hernandez-Ramirez
- Department of Physiology and Biophysics, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, United States
| | - Vishaka Santosh
- Department of Physiology and Biophysics, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, United States
| | - Alexander Washington
- Department of Physiology and Biophysics, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, United States
| | - Carlos R Escalante
- Department of Physiology and Biophysics, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, United States
| |
Collapse
|
7
|
Stevens CS, Carmichael JC, Watkinson R, Kowdle S, Reis RA, Hamane K, Jang J, Park A, Pernet O, Khamaikawin W, Hong P, Thibault P, Gowlikar A, An DS, Lee B. A temperature-sensitive and less immunogenic Sendai virus for efficient gene editing. J Virol 2024; 98:e0083224. [PMID: 39494910 PMCID: PMC11650993 DOI: 10.1128/jvi.00832-24] [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: 05/09/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024] Open
Abstract
The therapeutic potential of gene editing technologies hinges on the development of safe and effective delivery methods. In this study, we developed a temperature-sensitive and less immunogenic Sendai virus (ts SeV) as a novel delivery vector for CRISPR-Cas9 and for efficient gene editing in sensitive human cell types with limited induction of an innate immune response. ts SeV demonstrates high transduction efficiency in human CD34+ hematopoietic stem and progenitor cells (HSPCs) including transduction of the CD34+/CD38-/CD45RA-/CD90+(Thy1+)/CD49fhigh stem cell enriched subpopulation. The frequency of CCR5 editing exceeded 90% and bi-allelic CCR5 editing exceeded 70% resulting in significant inhibition of HIV-1 infection in primary human CD14+ monocytes. These results demonstrate the potential of the ts SeV platform as a safe, efficient, and flexible addition to the current gene-editing tool delivery methods, which may help further expand the possibilities in personalized medicine and the treatment of genetic disorders. IMPORTANCE Gene editing has the potential to be a powerful tool for the treatment of human diseases including HIV, β-thalassemias, and sickle cell disease. Recent advances have begun to overcome one of the major limiting factors of this technology, namely delivery of the CRISPR-Cas9 gene editing machinery, by utilizing viral vectors. However, gene editing therapies have yet to be implemented due to inherent risks associated with the DNA viral vectors typically used for delivery. As an alternative strategy, we have developed an RNA-based Sendai virus CRISPR-Cas9 delivery vector that does not integrate into the genome, is temperature sensitive, and does not induce a significant host interferon response. This recombinant SeV successfully delivered CRISPR-Cas9 in primary human CD14+ monocytes ex vivo resulting in a high level of CCR5 editing and inhibition of HIV infection.
Collapse
Affiliation(s)
- Christian S. Stevens
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jillian C. Carmichael
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ruth Watkinson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shreyas Kowdle
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rebecca A. Reis
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kory Hamane
- UCLA School of Nursing, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Jason Jang
- UCLA School of Nursing, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Arnold Park
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Olivier Pernet
- UCLA School of Nursing, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Wannisa Khamaikawin
- UCLA School of Nursing, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Patrick Hong
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Patricia Thibault
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aditya Gowlikar
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dong Sung An
- UCLA School of Nursing, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
8
|
Sheehan M, Kumpf SW, Qian J, Rubitski DM, Oziolor E, Lanz TA. Comparison and cross-validation of long-read and short-read target-enrichment sequencing methods to assess AAV vector integration into host genome. Mol Ther Methods Clin Dev 2024; 32:101352. [PMID: 39506980 PMCID: PMC11539135 DOI: 10.1016/j.omtm.2024.101352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024]
Abstract
Evaluation of host integration profiles by adeno-associated virus (AAV) is an important component of de-risking novel AAV gene therapies. Targeted enrichment sequencing (TES) is a cost-effective and comprehensive method for assessing integration. Most published TES datasets have been generated using short-read sequencing, which enables quantitation of integration sites (ISs) and identifies patterns such as hotspots or clonal expansion. Characteristics such as IS length and recombination require longer reads to measure. The present study compared short-read to long-read TES using samples from monkeys treated with AAV and used in vitro lentiviral-treated samples, a stable cell line, and an engineered spike-in as controls. Both methods showed stochastic integration by both AAV and lentivirus, with most vector domains identified in ISs. More ISs were identified by short-read TES, as deeper coverage per base was achieved from a single sequencing run. AAV-treated samples showed minimal evidence of clonal expansion, in contrast to in vitro treated and stably transduced lentiviral cell line samples. Long-read TES revealed vector rearrangement in 4%-40% of ISs in AAV-treated animals. In summary, both methods yielded similar conclusions about relative numbers of ISs and overall patterns. Long-read TES identified fewer ISs but enabled measurement of IS length and recombination patterns.
Collapse
Affiliation(s)
- Mark Sheehan
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc, Groton, CT 06340, USA
| | - Steven W. Kumpf
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc, Groton, CT 06340, USA
| | - Jessie Qian
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc, Groton, CT 06340, USA
| | - David M. Rubitski
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc, Groton, CT 06340, USA
| | - Elias Oziolor
- Computational Safety Sciences, Pfizer Inc, Groton, CT 06340, USA
| | - Thomas A. Lanz
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc, Groton, CT 06340, USA
| |
Collapse
|
9
|
Wang JH, Zhan W, Gallagher TL, Gao G. Recombinant adeno-associated virus as a delivery platform for ocular gene therapy: A comprehensive review. Mol Ther 2024; 32:4185-4207. [PMID: 39489915 PMCID: PMC11638839 DOI: 10.1016/j.ymthe.2024.10.017] [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: 05/10/2024] [Revised: 09/18/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
Adeno-associated virus (AAV) has emerged as a leading platform for in vivo gene therapy, particularly in ocular diseases. AAV-based therapies are characterized by low pathogenicity and broad tissue tropism and have demonstrated clinical success, as exemplified by voretigene neparvovec-rzyl (Luxturna) being the first gene therapy to be approved by the U.S. Food and Drug Administration to treat RPE65-associated Leber congenital amaurosis (LCA). However, several challenges remain in the development of AAV-based gene therapies, including immune responses, limited cargo capacity, and the need for enhanced transduction efficiency, especially for intravitreal delivery to photoreceptors and retinal pigment epithelium cells. This review explores the biology of AAVs in the context of gene therapy, innovations in capsid engineering, and clinical advancements in AAV-based ocular gene therapy. We highlight ongoing clinical trials targeting inherited retinal diseases and acquired conditions, discuss immune-related limitations, and examine novel strategies for enhancing AAV vector performance to address current barriers.
Collapse
Affiliation(s)
- Jiang-Hui Wang
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia; Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC 3002, Australia
| | - Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Thomas L Gallagher
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
10
|
Yudaeva A, Kostyusheva A, Kachanov A, Brezgin S, Ponomareva N, Parodi A, Pokrovsky VS, Lukashev A, Chulanov V, Kostyushev D. Clinical and Translational Landscape of Viral Gene Therapies. Cells 2024; 13:1916. [PMID: 39594663 PMCID: PMC11592828 DOI: 10.3390/cells13221916] [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: 09/23/2024] [Revised: 10/29/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Gene therapies hold significant promise for treating previously incurable diseases. A number of gene therapies have already been approved for clinical use. Currently, gene therapies are mostly limited to the use of adeno-associated viruses and the herpes virus. Viral vectors, particularly those derived from human viruses, play a critical role in this therapeutic approach due to their ability to efficiently deliver genetic material to target cells. Despite their advantages, such as stable gene expression and efficient transduction, viral vectors face numerous limitations that hinder their broad application. These limitations include small cloning capacities, immune and inflammatory responses, and risks of insertional mutagenesis. This review explores the current landscape of viral vectors used in gene therapy, discussing the different types of DNA- and RNA-based viral vectors, their characteristics, limitations, and current medical and potential clinical applications. The review also highlights strategies to overcome existing challenges, including optimizing vector design, improving safety profiles, and enhancing transgene expression both using molecular techniques and nanotechnologies, as well as by approved drug formulations.
Collapse
Affiliation(s)
- Alexandra Yudaeva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
| | - Anastasiya Kostyusheva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
| | - Artyom Kachanov
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
| | - Sergey Brezgin
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
| | - Natalia Ponomareva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
- Department of Pharmaceutical and Toxicological Chemistry, Sechenov First Moscow State Medical University, 119146 Moscow, Russia
| | - Alessandro Parodi
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
| | - Vadim S. Pokrovsky
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
- Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia
- Department of Biochemistry, People’s Friendship University, 117198 Moscow, Russia
| | - Alexander Lukashev
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
- Research Institute for Systems Biology and Medicine, 117246 Moscow, Russia
| | - Vladimir Chulanov
- Department of Infectious Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia;
| | - Dmitry Kostyushev
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.Y.); (A.K.); (A.K.); (S.B.); (N.P.); (A.L.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.P.); (V.S.P.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
11
|
Maritato R, Medugno A, D'Andretta E, De Riso G, Lupo M, Botta S, Marrocco E, Renda M, Sofia M, Mussolino C, Bacci ML, Surace EM. A DNA base-specific sequence interposed between CRX and NRL contributes to RHODOPSIN expression. Sci Rep 2024; 14:26313. [PMID: 39487168 PMCID: PMC11530525 DOI: 10.1038/s41598-024-76664-8] [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: 07/05/2024] [Accepted: 10/15/2024] [Indexed: 11/04/2024] Open
Abstract
Gene expression emerges from DNA sequences through the interaction of transcription factors (TFs) with DNA cis-regulatory sequences. In eukaryotes, TFs bind to transcription factor binding sites (TFBSs) with differential affinities, enabling cell-specific gene expression. In this view, DNA enables TF binding along a continuum ranging from low to high affinity depending on its sequence composition; however, it is not known whether evolution has entailed a further level of entanglement between DNA-protein interaction. Here we found that the composition and length (22 bp) of the DNA sequence interposed between the CRX and NRL retinal TFs in the proximal promoter of RHODOPSIN (RHO) largely controls the expression levels of RHO. Mutagenesis of CRX-NRL DNA linking sequences (here termed "DNA-linker") results in uncorrelated gene expression variation. In contrast, mutual exchange of naturally occurring divergent human and mouse Rho cis-regulatory elements conferred similar yet species-specific Rho expression levels. Two orthogonal DNA-binding proteins targeted to the DNA-linker either activate or repress the expression of Rho depending on the DNA-linker orientation relative to the CRX and NRL binding sites. These results argue that, in this instance, DNA itself contributes to CRX and NRL activities through a code based on specific base sequences of a defined length, ultimately determining optimal RHO expression levels.
Collapse
Affiliation(s)
- Rosa Maritato
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy
| | - Alessia Medugno
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy
| | - Emanuela D'Andretta
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy
| | - Giulia De Riso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
- AOU Federico II, Naples, Italy
| | - Mariangela Lupo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Salvatore Botta
- Department of Translational Medical Science, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Elena Marrocco
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Mario Renda
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Martina Sofia
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | | | - Maria Laura Bacci
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Enrico Maria Surace
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
12
|
Libertini S, Jadlowsky JK, Lanz TA, Mihalcik LM, Pizzurro DM. Genotoxicity evaluation of gene therapies: A report from the International Workshop on Genotoxicity Testing (IWGT) 2022. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2024. [PMID: 39301812 DOI: 10.1002/em.22633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/22/2024]
Abstract
At the 8th International Workshop on Genotoxicity Testing meeting in Ottawa, in August 2022, a plenary session was dedicated to the genotoxicity risk evaluation of gene therapies, including insertional oncogenesis and off-target genome editing. This brief communication summarizes the topics of discussion and the main insights from the speakers. Common themes included recommendations to conduct tailored risk assessments based on a weight-of-evidence approach, to promote data sharing, transparency, and cooperation between stakeholders, and to develop state-of-the-art validated tests relevant to clinical scenarios.
Collapse
Affiliation(s)
- S Libertini
- Novartis Biomedical Research, Basel, Switzerland
| | - J K Jadlowsky
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - T A Lanz
- Pfizer Drug Safety Research & Development, Groton, Connecticut, USA
| | - L M Mihalcik
- Aclairo Pharmaceutical Development Group, Sterling, Virginia, USA
| | | |
Collapse
|
13
|
Rallabandi R, Sharp B, Majerus S, Royster A, Hoffer S, Ikeda M, Devaux P. Engineering single-cycle MeV vector for CRISPR-Cas9 gene editing. Mol Ther Methods Clin Dev 2024; 32:101290. [PMID: 39070290 PMCID: PMC11283025 DOI: 10.1016/j.omtm.2024.101290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/21/2024] [Indexed: 07/30/2024]
Abstract
CRISPR-Cas9-mediated gene editing has vast applications in basic and clinical research and is a promising tool for several disorders. Our lab previously developed a non-integrating RNA virus, measles virus (MeV), as a single-cycle reprogramming vector by replacing the viral attachment protein with the reprogramming factors for induced pluripotent stem cell generation. Encouraged by the MeV reprogramming vector efficiency, in this study, we develop a single-cycle MeV vector to deliver the gRNA(s) and Cas9 nuclease to human cells for efficient gene editing. We show that the MeV vector achieved on-target gene editing of the reporter (mCherry) and endogenous genes (HBB and FANCD1) in human cells. Additionally, the MeV vector achieved precise knock-in via homology-directed repair using a single-stranded oligonucleotide donor. The MeV vector is a new and flexible platform for gene knock-out and knock-in modifications in human cells, capable of incorporating new technologies as they are developed.
Collapse
Affiliation(s)
- Ramya Rallabandi
- Mayo Clinic Graduate School of Biomedical Sciences, Virology and Gene Therapy Graduate Track, Mayo Clinic, Rochester, MN 55905, USA
| | - Brenna Sharp
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Spencer Majerus
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Austin Royster
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Sarrianna Hoffer
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Mia Ikeda
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Patricia Devaux
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Virology and Gene Therapy Graduate Track, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
14
|
Agarwal S, Sandza K, Obrochta Moss K, Vora M, Bowen A, Bunch B, Holcomb J, Robinson TM, Jayaram K, Russell CB, Zoog S, Vettermann C, Henshaw J. Blood biodistribution and vector shedding of valoctocogene roxaparvovec in people with severe hemophilia A. Blood Adv 2024; 8:4606-4615. [PMID: 39024543 PMCID: PMC11401231 DOI: 10.1182/bloodadvances.2024013150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 07/20/2024] Open
Abstract
ABSTRACT Following systemically administered adeno-associated virus gene therapy, vector particles are widely distributed, raising concerns about horizontal or germline vector transmission. Characterization of biodistribution and kinetics of vector DNA in body fluids can address these concerns and provide insights into vector behavior in accessible samples. We investigated biodistribution and vector shedding profile of valoctocogene roxaparvovec in men with severe hemophilia A enrolled in the phase 3 GENEr8-1 trial. Participants (n = 134) received a single 6 × 1013 vector genome (vg)/kg infusion and were assessed over 3 years. Vector DNA was measured using 4 different assays. Total vector DNA was evaluated in blood, saliva, stool, semen, and urine by quantitative polymerase chain reaction (qPCR). Encapsidated vector DNA was measured in plasma and semen with immunocapture-based qPCR. Contiguity of vgs and assembly of inverted terminal repeat fusions were measured in whole blood and peripheral blood mononuclear cells (PBMCs) using multicolor digital PCR. Median peak vector DNA levels observed 1 to 8 days after dosing were highest in blood, followed by saliva, semen, stool, and urine. Concentrations declined steadily. Encapsidated vector DNA cleared faster than total vector DNA, achieving clearance by ≤12 weeks in plasma and semen. Predominant vector genome forms transitioned from noncontiguous to full-length over time in whole blood and PBMCs, indicating formation of stable circularized episomes within nucleated cells. The replication-incompetent nature of valoctocogene roxaparvovec, coupled with steady clearance of total and encapsidated vector DNA from shedding matrices, indicates transmission risk is low. This trial was registered at www.ClinicalTrials.gov as #NCT03370913.
Collapse
Affiliation(s)
- Suresh Agarwal
- Translational Sciences, BioMarin Pharmaceutical Inc, Novato, CA
| | - Krystal Sandza
- Translational Sciences, BioMarin Pharmaceutical Inc, Novato, CA
| | | | - Monica Vora
- Translational Sciences, BioMarin Pharmaceutical Inc, Novato, CA
| | - Alisa Bowen
- Translational Sciences, BioMarin Pharmaceutical Inc, Novato, CA
| | - Brenna Bunch
- Translational Sciences, BioMarin Pharmaceutical Inc, Novato, CA
| | | | - Tara M. Robinson
- Late Clinical Development, BioMarin Pharmaceutical Inc, Novato, CA
| | - Kala Jayaram
- Pharmacovigilance, BioMarin Pharmaceutical Inc, Novato, CA
| | | | - Stephen Zoog
- Translational Sciences, BioMarin Pharmaceutical Inc, Novato, CA
| | | | - Joshua Henshaw
- Translational Sciences, BioMarin Pharmaceutical Inc, Novato, CA
| |
Collapse
|
15
|
Huang Y, Mei H, Deng C, Wang W, Yuan C, Nie Y, Li JD, Liu J. EXTL3 and NPC1 are mammalian host factors for Autographa californica multiple nucleopolyhedrovirus infection. Nat Commun 2024; 15:7711. [PMID: 39231976 PMCID: PMC11374996 DOI: 10.1038/s41467-024-52193-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024] Open
Abstract
Baculovirus is an obligate parasitic virus of the phylum Arthropoda. Baculovirus including Autographa californica multiple nucleopolyhedrovirus (AcMNPV) has been widely used in the laboratory and industrial preparation of proteins or protein complexes. Due to its large packaging capacity and non-replicative and non-integrative natures in mammals, baculovirus has been proposed as a gene therapy vector for transgene delivery. However, the mechanism of baculovirus transduction in mammalian cells has not been fully illustrated. Here, we employed a cell surface protein-focused CRISPR screen to identify host dependency factors for baculovirus transduction in mammalian cells. The screening experiment uncovered a series of baculovirus host factors in human cells, including exostosin-like glycosyltransferase 3 (EXTL3) and NPC intracellular cholesterol transporter 1 (NPC1). Further investigation illustrated that EXTL3 affected baculovirus attachment and entry by participating in heparan sulfate biosynthesis. In addition, NPC1 promoted baculovirus transduction by mediating membrane fusion and endosomal escape. Moreover, in vivo, baculovirus transduction in Npc1-/+ mice showed that disruption of Npc1 gene significantly reduced baculovirus transduction in mouse liver. In summary, our study revealed the functions of EXTL3 and NPC1 in baculovirus attachment, entry, and endosomal escape in mammalian cells, which is useful for understanding baculovirus transduction in human cells.
Collapse
Affiliation(s)
- Yuege Huang
- Furong Laboratory, Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Hong Mei
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China.
| | - Chunchen Deng
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Wei Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Chao Yuan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yan Nie
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Jia-Da Li
- Furong Laboratory, Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Animal Models for Human Diseases, Changsha, Hunan, China.
| | - Jia Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Shanghai Clinical Research and Trial Center, Shanghai, China.
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China.
- Shanghai Asiflyerbio Biotechnology, Shanghai, China.
| |
Collapse
|
16
|
Liu JQ, Jabbari A, Lin CH, Akkanapally V, Frankel WL, Basu S, He K, Zheng P, Liu Y, Bai XF. IL-27 Gene Therapy Ameliorates IPEX Syndrome Caused by Germline Mutation of Foxp3 Gene: A Major Role for Induction of IL-10. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:559-566. [PMID: 38975727 PMCID: PMC11333164 DOI: 10.4049/jimmunol.2400056] [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: 02/05/2024] [Accepted: 06/13/2024] [Indexed: 07/09/2024]
Abstract
Inactivating mutations of Foxp3, the master regulator of regulatory T cell development and function, lead to immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome in mice and humans. IPEX is a fatal autoimmune disease, with allogeneic stem cell transplant being the only available therapy. In this study, we report that a single dose of adeno-associated virus (AAV)-IL-27 to young mice with naturally occurring Foxp3 mutation (Scurfy mice) substantially ameliorates clinical symptoms, including growth retardation and early fatality. Correspondingly, AAV-IL-27 gene therapy significantly prevented naive T cell activation, as manifested by downregulation of CD62L and upregulation of CD44, and immunopathology typical of IPEX. Because IL-27 is known to induce IL-10, a key effector molecule of regulatory T cells, we evaluated the contribution of IL-10 induction by crossing IL-10-null allele to Scurfy mice. Although IL-10 deficiency does not affect the survival of Scurfy mice, it largely abrogated the therapeutic effect of AAV-IL-27. Our study revealed a major role for IL-10 in AAV-IL-27 gene therapy and demonstrated that IPEX is amenable to gene therapy.
Collapse
MESH Headings
- Animals
- Forkhead Transcription Factors/genetics
- Mice
- Interleukin-10/genetics
- Interleukin-10/immunology
- Genetic Therapy/methods
- Germ-Line Mutation
- T-Lymphocytes, Regulatory/immunology
- Genetic Diseases, X-Linked/therapy
- Genetic Diseases, X-Linked/immunology
- Genetic Diseases, X-Linked/genetics
- Interleukins/immunology
- Interleukins/genetics
- Diarrhea/genetics
- Diarrhea/therapy
- Diarrhea/immunology
- Intestinal Diseases/immunology
- Intestinal Diseases/genetics
- Intestinal Diseases/therapy
- Dependovirus/genetics
- Mice, Inbred C57BL
- Immune System Diseases/immunology
- Immune System Diseases/therapy
- Immune System Diseases/genetics
- Immune System Diseases/congenital
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/therapy
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/congenital
- Mice, Knockout
- Lymphocyte Activation/immunology
- Humans
- Interleukin-27/genetics
Collapse
Affiliation(s)
- Jin-Qing Liu
- Department of Pathology and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Ali Jabbari
- Department of Dermatology, University of Iowa, College of Medicine, Iowa City, Iowa, USA
| | - Cho-Hao Lin
- Department of Pathology and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Venu Akkanapally
- Department of Pathology and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Wendy L. Frankel
- Department of Pathology and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Sujit Basu
- Department of Pathology and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Kai He
- Division of Medical Oncology, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Pan Zheng
- OncoC4, Inc., 640 Medical Center Drive, Rockville, MD, USA
| | - Yang Liu
- OncoC4, Inc., 640 Medical Center Drive, Rockville, MD, USA
| | - Xue-Feng Bai
- Department of Pathology and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
17
|
Mukalel AJ, Hamilton AG, Billingsley MM, Li J, Thatte AS, Han X, Safford HC, Padilla MS, Papp T, Parhiz H, Weissman D, Mitchell MJ. Oxidized mRNA Lipid Nanoparticles for In Situ Chimeric Antigen Receptor Monocyte Engineering. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2312038. [PMID: 39628840 PMCID: PMC11611297 DOI: 10.1002/adfm.202312038] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Indexed: 12/06/2024]
Abstract
Chimeric antigen receptor (CAR) monocyte and macrophage therapies are promising solid tumor immunotherapies that can overcome the challenges facing conventional CAR T cell therapy. mRNA lipid nanoparticles (mRNA-LNPs) offer a viable platform for in situ engineering of CAR monocytes with transient and tunable CAR expression to reduce off-tumor toxicity and streamline cell manufacturing. However, identifying LNPs with monocyte tropism and intracellular delivery potency is difficult using traditional screening techniques. Here, ionizable lipid design and high-throughput in vivo screening are utilized to identify a new class of oxidized LNPs with innate tropism and mRNA delivery to monocytes. A library of oxidized (oLNPs) and unoxidized LNPs (uLNPs) is synthesized to evaluate mRNA delivery to immune cells. oLNPs demonstrate notable differences in morphology, ionization energy, and pKa, therefore enhancing delivery to human macrophages, but not T cells. Subsequently, in vivo library screening with DNA barcodes identifies an oLNP formulation, C14-O2, with innate tropism to monocytes. In a proof-of-concept study, the C14-O2 LNP is used to engineer functional CD19-CAR monocytes in situ for robust B cell aplasia (45%) in healthy mice. This work highlights the utility of oxidized LNPs as a promising platform for engineering CAR macrophages/monocytes for solid tumor CAR monocyte therapy.
Collapse
Affiliation(s)
- Alvin J. Mukalel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Alex G. Hamilton
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Margaret M. Billingsley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jacqueline Li
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Ajay S. Thatte
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Xuexiang Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Hannah C. Safford
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Marshall S. Padilla
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Tyler Papp
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Hamideh Parhiz
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
18
|
Catalán-Tatjer D, Tzimou K, Nielsen LK, Lavado-García J. Unravelling the essential elements for recombinant adeno-associated virus (rAAV) production in animal cell-based platforms. Biotechnol Adv 2024; 73:108370. [PMID: 38692443 DOI: 10.1016/j.biotechadv.2024.108370] [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: 01/16/2024] [Revised: 04/05/2024] [Accepted: 04/27/2024] [Indexed: 05/03/2024]
Abstract
Recombinant adeno-associated viruses (rAAVs) stand at the forefront of gene therapy applications, holding immense significance for their safe and efficient gene delivery capabilities. The constantly increasing and unmet demand for rAAVs underscores the need for a more comprehensive understanding of AAV biology and its impact on rAAV production. In this literature review, we delved into AAV biology and rAAV manufacturing bioprocesses, unravelling the functions and essentiality of proteins involved in rAAV production. We discuss the interconnections between these proteins and how they affect the choice of rAAV production platform. By addressing existing inconsistencies, literature gaps and limitations, this review aims to define a minimal set of genes that are essential for rAAV production, providing the potential to advance rAAV biomanufacturing, with a focus on minimizing the genetic load within rAAV-producing cells.
Collapse
Affiliation(s)
- David Catalán-Tatjer
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark
| | - Konstantina Tzimou
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark
| | - Lars K Nielsen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark; Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Australia
| | - Jesús Lavado-García
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Denmark.
| |
Collapse
|
19
|
Mohanty S, Batabyal S, Ayyagari A, Sharif NA. Safety of intravitreally delivered AAV2 vector-mediated multi-characteristic opsin genetic construct in wild type beagle dogs. J Gene Med 2024; 26:e3720. [PMID: 39041639 DOI: 10.1002/jgm.3720] [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: 03/09/2024] [Revised: 06/19/2024] [Accepted: 06/28/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND A novel adeno-associated virus 2 (AAV2)-carried multi-characteristic opsin (MCO) (MCO-010) is undergoing several clinical trials as a novel therapeutic modality for the treatment of degenerative retinal diseases including retinitis pigmentosa and Stargardt disease. The present study aimed to determine the ocular and systemic safety of MCO-010 and the AAV2 vehicle in adult Beagle dogs following intravitreal (IVT) injection. METHODS The current safety/toxicology studies spanning 13 weeks described here utilized well-documented techniques to assess the effects of IVT injection of MCO-010 up to 2.2 × 1011 genome copies (gc) per eye, or the AAV2 capsid (vehicle control) on gross behavioral and immunogenic changes, alterations in body weights, blood biochemistry, hematology, blood coagulation, gross necropsy lesions, organ weight changes and histopathology in the dogs (n = 4 per group; two males and two females per group). Immunohistochemical and functional electroretinogram studies were also conducted to determine MCO expression in the retina and determine any retinal toxicity associated with MCO-010. RESULTS There were no significant deleterious effects of the MCO-010 (or the AAV2 at the tested doses) on any of the examined parameters, including the absence of any severe ocular or systemic adverse events. However, as expected, inflammation after IVT delivery of AAV2 and MCO-010 was observed in the conjunctivae of all groups of animals, although this self-resolved within 1 week post-injection. Quantitative immunohistochemical analyses of MCO-010-associated mCherry revealed successful delivery of the gene therapy within the inner retina. CONCLUSIONS In summary, MCO-010 demonstrated a favorable safety profile when administered to the eyes of adult Beagle dogs of both sexes at dose levels up to 2.2 × 1011 gc per eye, with no adverse effects observed. This dose was identified as the No Observed Adverse Effect Level (i.e. NOAEL) and guided selection of safe doses for human clinical trials.
Collapse
Affiliation(s)
- Samarendra Mohanty
- Nanoscope Technologies LLC, Bedford, TX, USA
- Nanoscope Therapeutics Inc., Dallas, TX, USA
| | | | | | | |
Collapse
|
20
|
Chung M, Imanaka K, Huang Z, Watarai A, Wang MY, Tao K, Ejima H, Aida T, Feng G, Okuyama T. Conditional knockout of Shank3 in the ventral CA1 by quantitative in vivo genome-editing impairs social memory in mice. Nat Commun 2024; 15:4531. [PMID: 38866749 PMCID: PMC11169449 DOI: 10.1038/s41467-024-48430-x] [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: 12/30/2022] [Accepted: 04/26/2024] [Indexed: 06/14/2024] Open
Abstract
Individuals with autism spectrum disorder (ASD) have a higher prevalence of social memory impairment. A series of our previous studies revealed that hippocampal ventral CA1 (vCA1) neurons possess social memory engram and that the neurophysiological representation of social memory in the vCA1 neurons is disrupted in ASD-associated Shank3 knockout mice. However, whether the dysfunction of Shank3 in vCA1 causes the social memory impairment observed in ASD remains unclear. In this study, we found that vCA1-specific Shank3 conditional knockout (cKO) by the adeno-associated virus (AAV)- or specialized extracellular vesicle (EV)- mediated in vivo gene editing was sufficient to recapitulate the social memory impairment in male mice. Furthermore, the utilization of EV-mediated Shank3-cKO allowed us to quantitatively examine the role of Shank3 in social memory. Our results suggested that there is a certain threshold for the proportion of Shank3-cKO neurons required for social memory disruption. Thus, our study provides insight into the population coding of social memory in vCA1, as well as the pathological mechanisms underlying social memory impairment in ASD.
Collapse
Affiliation(s)
- Myung Chung
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Katsutoshi Imanaka
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ziyan Huang
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akiyuki Watarai
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Mu-Yun Wang
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Kentaro Tao
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Hirotaka Ejima
- Department of Materials Engineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Tomomi Aida
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Teruhiro Okuyama
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan.
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
21
|
Christoffers S, Seiler L, Wiebe E, Blume C. Possibilities and efficiency of MSC co-transfection for gene therapy. Stem Cell Res Ther 2024; 15:150. [PMID: 38783353 PMCID: PMC11119386 DOI: 10.1186/s13287-024-03757-6] [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: 02/12/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are not only capable of self-renewal, trans-differentiation, homing to damaged tissue sites and immunomodulation by secretion of trophic factors but are also easy to isolate and expand. Because of these characteristics, they are used in numerous clinical trials for cell therapy including immune and neurological disorders, diabetes, bone and cartilage diseases and myocardial infarction. However, not all trials have successful outcomes, due to unfavourable microenvironmental factors and the heterogenous nature of MSCs. Therefore, genetic manipulation of MSCs can increase their prospect. Currently, most studies focus on single transfection with one gene. Even though the introduction of more than one gene increases the complexity, it also increases the effectivity as different mechanism are triggered, leading to a synergistic effect. In this review we focus on the methodology and efficiency of co-transfection, as well as the opportunities and pitfalls of these genetically engineered cells for therapy.
Collapse
Affiliation(s)
- Sina Christoffers
- Institute for Technical Chemistry, Leibniz University Hannover, Callinstr. 3-5, 30167, Hannover, Germany.
- Cluster of Excellence Hearing4all, Hannover, Germany.
| | - Lisa Seiler
- Institute for Technical Chemistry, Leibniz University Hannover, Callinstr. 3-5, 30167, Hannover, Germany
| | - Elena Wiebe
- Institute for Technical Chemistry, Leibniz University Hannover, Callinstr. 3-5, 30167, Hannover, Germany
- Cluster of Excellence Hearing4all, Hannover, Germany
| | - Cornelia Blume
- Institute for Technical Chemistry, Leibniz University Hannover, Callinstr. 3-5, 30167, Hannover, Germany
- Cluster of Excellence Hearing4all, Hannover, Germany
| |
Collapse
|
22
|
Stevens CS, Carmichael J, Watkinson R, Kowdle S, Reis RA, Hamane K, Jang J, Park A, Pernet O, Khamaikawin W, Hong P, Thibault P, Gowlikar A, An DS, Lee B. A temperature-sensitive and interferon-silent Sendai virus vector for CRISPR-Cas9 delivery and gene editing in primary human cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592383. [PMID: 38746439 PMCID: PMC11092779 DOI: 10.1101/2024.05.03.592383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The transformative potential of gene editing technologies hinges on the development of safe and effective delivery methods. In this study, we developed a temperature-sensitive and interferon-silent Sendai virus (ts SeV) as a novel delivery vector for CRISPR-Cas9 and for efficient gene editing in sensitive human cell types without inducing IFN responses. ts SeV demonstrates unprecedented transduction efficiency in human CD34+ hematopoietic stem and progenitor cells (HSPCs) including transduction of the CD34+/CD38-/CD45RA-/CD90+(Thy1+)/CD49fhigh stem cell enriched subpopulation. The frequency of CCR5 editing exceeded 90% and bi-allelic CCR5 editing exceeded 70% resulting in significant inhibition of HIV-1 infection in primary human CD14+ monocytes. These results demonstrate the potential of the ts SeV platform as a safe, efficient, and flexible addition to the current gene-editing tool delivery methods, which may help to further expand the possibilities in personalized medicine and the treatment of genetic disorders.
Collapse
Affiliation(s)
- Christian S Stevens
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Jillian Carmichael
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ruth Watkinson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Shreyas Kowdle
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Rebecca A Reis
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Kory Hamane
- UCLA School of Nursing, Los Angeles, California, 90095
- UCLA AIDS Institute, Los Angeles, California, 90095
| | - Jason Jang
- UCLA School of Nursing, Los Angeles, California, 90095
- UCLA AIDS Institute, Los Angeles, California, 90095
| | - Arnold Park
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Olivier Pernet
- UCLA School of Nursing, Los Angeles, California, 90095
- UCLA AIDS Institute, Los Angeles, California, 90095
| | - Wannisa Khamaikawin
- UCLA School of Nursing, Los Angeles, California, 90095
- UCLA AIDS Institute, Los Angeles, California, 90095
| | - Patrick Hong
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Patricia Thibault
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Aditya Gowlikar
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Dong Sung An
- UCLA School of Nursing, Los Angeles, California, 90095
- UCLA AIDS Institute, Los Angeles, California, 90095
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
23
|
Anguela XM, High KA. Hemophilia B and gene therapy: a new chapter with etranacogene dezaparvovec. Blood Adv 2024; 8:1796-1803. [PMID: 38592711 PMCID: PMC11006816 DOI: 10.1182/bloodadvances.2023010511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/01/2024] [Indexed: 04/10/2024] Open
Abstract
ABSTRACT The US Food and Drug Administration (FDA)'s authorization of etranacogene dezaparvovec (Hemgenix) is a significant milestone, constituting not only the first FDA approval of a gene therapy for hemophilia but also the first approval of a liver-targeted adeno-associated virus vector gene therapy. This review summarizes the nonclinical studies and clinical development that supported regulatory clearance. Similar to other gene therapies for single gene disorders, both the short-term safety and the phenotypic improvement were unequivocal, justifying the modest-sized safety and efficacy database, which included 57 participants across the phase 2b (3 participants) and phase 3 (54 participants) studies. The most common adverse reactions included liver enzyme elevation, headache, flu-like symptoms, infusion-related reactions, creatine kinase elevation, malaise, and fatigue; these were mostly transient. One participant had hepatocellular carcinoma on a study-mandated liver ultrasound conducted 1 year after vector infusion; molecular analysis of the resected tumor showed no evidence of vector-related insertional mutagenesis as the etiology. A remarkable 96% of participants in the phase 3 trial were able to stop factor IX (FIX) prophylaxis, with the study demonstrating noninferiority to FIX prophylaxis in terms of the primary end point, annualized bleeding rate. Key secondary end points such as the annualized infusion rate, which declined by 97%, and the plasma FIX activity level at 18 months after infusion, with least squares mean increase of 34.3 percentage points compared with baseline, were both clinically and statistically significant. The FDA's landmark approval of Hemgenix as a pioneering treatment for hemophilia stands on the shoulders of >20 years of gene therapy clinical research and heralds a promising future for genomic medicines.
Collapse
Affiliation(s)
| | - Katherine A. High
- Rockefeller University, New York, NY
- Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
24
|
Henriques C, Lopes MM, Silva AC, Lobo DD, Badin RA, Hantraye P, Pereira de Almeida L, Nobre RJ. Viral-based animal models in polyglutamine disorders. Brain 2024; 147:1166-1189. [PMID: 38284949 DOI: 10.1093/brain/awae012] [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: 07/09/2023] [Revised: 11/26/2023] [Accepted: 12/30/2023] [Indexed: 01/30/2024] Open
Abstract
Polyglutamine disorders are a complex group of incurable neurodegenerative disorders caused by an abnormal expansion in the trinucleotide cytosine-adenine-guanine tract of the affected gene. To better understand these disorders, our dependence on animal models persists, primarily relying on transgenic models. In an effort to complement and deepen our knowledge, researchers have also developed animal models of polyglutamine disorders employing viral vectors. Viral vectors have been extensively used to deliver genes to the brain, not only for therapeutic purposes but also for the development of animal models, given their remarkable flexibility. In a time- and cost-effective manner, it is possible to use different transgenes, at varying doses, in diverse targeted tissues, at different ages, and in different species, to recreate polyglutamine pathology. This paper aims to showcase the utility of viral vectors in disease modelling, share essential considerations for developing animal models with viral vectors, and provide a comprehensive review of existing viral-based animal models for polyglutamine disorders.
Collapse
Affiliation(s)
- Carina Henriques
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Miguel M Lopes
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research (III), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Ana C Silva
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research (III), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Diana D Lobo
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research (III), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Romina Aron Badin
- CEA, DRF, Institute of Biology François Jacob, Molecular Imaging Research Center (MIRCen), 92265 Fontenay-aux-Roses, France
- CNRS, CEA, Paris-Sud University, Université Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), 92265 Fontenay-aux-Roses, France
| | - Philippe Hantraye
- CEA, DRF, Institute of Biology François Jacob, Molecular Imaging Research Center (MIRCen), 92265 Fontenay-aux-Roses, France
- CNRS, CEA, Paris-Sud University, Université Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), 92265 Fontenay-aux-Roses, France
| | - Luís Pereira de Almeida
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Rui Jorge Nobre
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research (III), University of Coimbra, 3030-789 Coimbra, Portugal
| |
Collapse
|
25
|
Wang JH, Gessler DJ, Zhan W, Gallagher TL, Gao G. Adeno-associated virus as a delivery vector for gene therapy of human diseases. Signal Transduct Target Ther 2024; 9:78. [PMID: 38565561 PMCID: PMC10987683 DOI: 10.1038/s41392-024-01780-w] [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: 07/05/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
Adeno-associated virus (AAV) has emerged as a pivotal delivery tool in clinical gene therapy owing to its minimal pathogenicity and ability to establish long-term gene expression in different tissues. Recombinant AAV (rAAV) has been engineered for enhanced specificity and developed as a tool for treating various diseases. However, as rAAV is being more widely used as a therapy, the increased demand has created challenges for the existing manufacturing methods. Seven rAAV-based gene therapy products have received regulatory approval, but there continue to be concerns about safely using high-dose viral therapies in humans, including immune responses and adverse effects such as genotoxicity, hepatotoxicity, thrombotic microangiopathy, and neurotoxicity. In this review, we explore AAV biology with an emphasis on current vector engineering strategies and manufacturing technologies. We discuss how rAAVs are being employed in ongoing clinical trials for ocular, neurological, metabolic, hematological, neuromuscular, and cardiovascular diseases as well as cancers. We outline immune responses triggered by rAAV, address associated side effects, and discuss strategies to mitigate these reactions. We hope that discussing recent advancements and current challenges in the field will be a helpful guide for researchers and clinicians navigating the ever-evolving landscape of rAAV-based gene therapy.
Collapse
Affiliation(s)
- Jiang-Hui Wang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, 3002, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, 3002, Australia
| | - Dominic J Gessler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Neurological Surgery, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Thomas L Gallagher
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
26
|
Pille M, Avila JM, Park SH, Le CQ, Xue H, Haerynck F, Saxena L, Lee C, Shpall EJ, Bao G, Vandekerckhove B, Davis BR. Gene editing-based targeted integration for correction of Wiskott-Aldrich syndrome. Mol Ther Methods Clin Dev 2024; 32:101208. [PMID: 38414825 PMCID: PMC10897892 DOI: 10.1016/j.omtm.2024.101208] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 02/02/2024] [Indexed: 02/29/2024]
Abstract
Wiskott-Aldrich syndrome (WAS) is a severe X-linked primary immunodeficiency resulting from a diversity of mutations distributed across all 12 exons of the WAS gene. WAS encodes a hematopoietic-specific and developmentally regulated cytoplasmic protein (WASp). The objective of this study was to develop a gene correction strategy potentially applicable to most WAS patients by employing nuclease-mediated, site-specific integration of a corrective WAS gene sequence into the endogenous WAS chromosomal locus. In this study, we demonstrate the ability to target the integration of WAS2-12-containing constructs into intron 1 of the endogenous WAS gene of primary CD34+ hematopoietic stem and progenitor cells (HSPCs), as well as WASp-deficient B cell lines and WASp-deficient primary T cells. This intron 1 targeted integration (TI) approach proved to be quite efficient and restored WASp expression in treated cells. Furthermore, TI restored WASp-dependent function to WAS patient T cells. Edited CD34+ HSPCs exhibited the capacity for multipotent differentiation to various hematopoietic lineages in vitro and in transplanted immunodeficient mice. This methodology offers a potential editing approach for treatment of WAS using patient's CD34+ cells.
Collapse
Affiliation(s)
- Melissa Pille
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium
| | - John M. Avila
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - So Hyun Park
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Cuong Q. Le
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Haipeng Xue
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Filomeen Haerynck
- Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Lavanya Saxena
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Ciaran Lee
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | - Brian R. Davis
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
27
|
He X, Fu Y, Ma L, Yao Y, Ge S, Yang Z, Fan X. AAV for Gene Therapy in Ocular Diseases: Progress and Prospects. RESEARCH (WASHINGTON, D.C.) 2023; 6:0291. [PMID: 38188726 PMCID: PMC10768554 DOI: 10.34133/research.0291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 11/27/2023] [Indexed: 01/09/2024]
Abstract
Owing to the promising therapeutic effect and one-time treatment advantage, gene therapy may completely change the management of eye diseases, especially retinal diseases. Adeno-associated virus (AAV) is considered one of the most promising viral gene delivery tools because it can infect various types of tissues and is considered as a relatively safe gene delivery vector. The eye is one of the most popular organs for gene therapy, since its limited volume is suitable for small doses of AAV stably transduction. Recently, an increasing number of clinical trials of AAV-mediated gene therapy are underway. This review summarizes the biological functions of AAV and its application in the treatment of various ocular diseases, as well as the characteristics of different AAV delivery routes in clinical applications. Here, the latest research progresses in AAV-mediated gene editing and silencing strategies to modify that the genetic ocular diseases are systematically outlined, especially by base editing and prime editing. We discuss the progress of AAV in ocular optogenetic therapy. We also summarize the application of AAV-mediated gene therapy in animal models and the difficulties in its clinical transformation.
Collapse
Affiliation(s)
- Xiaoyu He
- Department of Ophthalmology, Ninth People’s Hospital,
Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yidian Fu
- Department of Ophthalmology, Ninth People’s Hospital,
Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Liang Ma
- Department of Ophthalmology, Ninth People’s Hospital,
Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yizheng Yao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University; Clinical Research Center of Neurological Disease,
The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People’s Hospital,
Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Zhi Yang
- Department of Ophthalmology, Ninth People’s Hospital,
Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People’s Hospital,
Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
28
|
Klinkovskij A, Shepelev M, Isaakyan Y, Aniskin D, Ulasov I. Advances of Genome Editing with CRISPR/Cas9 in Neurodegeneration: The Right Path towards Therapy. Biomedicines 2023; 11:3333. [PMID: 38137554 PMCID: PMC10741756 DOI: 10.3390/biomedicines11123333] [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: 11/08/2023] [Revised: 12/06/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
The rate of neurodegenerative disorders (NDDs) is rising rapidly as the world's population ages. Conditions such as Alzheimer's disease (AD), Parkinson's disease (PD), and dementia are becoming more prevalent and are now the fourth leading cause of death, following heart disease, cancer, and stroke. Although modern diagnostic techniques for detecting NDDs are varied, scientists are continuously seeking new and improved methods to enable early and precise detection. In addition to that, the present treatment options are limited to symptomatic therapy, which is effective in reducing the progression of neurodegeneration but lacks the ability to target the root cause-progressive loss of neuronal functioning. As a result, medical researchers continue to explore new treatments for these conditions. Here, we present a comprehensive summary of the key features of NDDs and an overview of the underlying mechanisms of neuroimmune dysfunction. Additionally, we dive into the cutting-edge treatment options that gene therapy provides in the quest to treat these disorders.
Collapse
Affiliation(s)
- Aleksandr Klinkovskij
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia; (A.K.); (D.A.)
| | - Mikhail Shepelev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilova Str., Moscow 119334, Russia
| | - Yuri Isaakyan
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 8 Trubetskaya Str., Moscow 119991, Russia;
| | - Denis Aniskin
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia; (A.K.); (D.A.)
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia; (A.K.); (D.A.)
| |
Collapse
|
29
|
Rostami MR, Leopold PL, Vasquez JM, de Mulder Rougvie M, Al Shakaki A, Hssain AA, Robay A, Hackett NR, Mezey JG, Crystal RG. Predicted deleterious variants in the human genome relevant to gene therapy with adeno-associated virus vectors. Mol Ther Methods Clin Dev 2023; 31:101136. [PMID: 38089635 PMCID: PMC10711236 DOI: 10.1016/j.omtm.2023.101136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 10/11/2023] [Indexed: 01/02/2025]
Abstract
Based on the observation that humans have variable responses of gene expression with the same dose of an adeno-associated vector, we hypothesized that there are deleterious variants in genes coding for processes required for adeno-associated virus (AAV)-mediated gene transfer/expression that may hamper or enhance the effectiveness of AAV-mediated gene therapy. To assess this hypothesis, we evaluated 69,442 whole genome sequences from three populations (European, African/African American, and Qatari) for predicted deleterious variants in 62 genes known to play a role in AAV-mediated gene transfer/expression. The analysis identified 5,564 potentially deleterious mutations of which 27 were classified as common based on an allele frequency ≥1% in at least one population studied. Many of these deleterious variants are predicated to prevent while others enhance effective AAV gene transfer/expression, and several are linked to known hereditary disorders. The data support the hypothesis that, like other drugs, human genetic variability contributes to the person-to-person effectiveness of AAV gene therapy and the screening for genetic variability should be considered as part of future clinical trials.
Collapse
Affiliation(s)
| | - Philip L. Leopold
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jenifer M. Vasquez
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Alya Al Shakaki
- Department of Genetic Medicine, Weill Cornell Medicine - Qatar, Doha, Qatar
| | | | - Amal Robay
- Department of Genetic Medicine, Weill Cornell Medicine - Qatar, Doha, Qatar
| | - Neil R. Hackett
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jason G. Mezey
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY, USA
| | - Ronald G. Crystal
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
30
|
Argirò A, Ding J, Adler E. Gene therapy for heart failure and cardiomyopathies. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2023; 76:1042-1054. [PMID: 37506969 DOI: 10.1016/j.rec.2023.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023]
Abstract
Gene therapy strategies encompass a range of approaches, including gene replacement and gene editing. Gene replacement involves providing a functional copy of a modified gene, while gene editing allows for the correction of existing genetic mutations. Gene therapy has already received approval for treating genetic disorders like Leber's congenital amaurosis and spinal muscular atrophy. Currently, research is being conducted to explore its potential use in cardiology. This review aims to summarize the mechanisms behind different gene therapy strategies, the available delivery systems, the primary risks associated with gene therapy, ongoing clinical trials, and future targets, with a particular emphasis on cardiomyopathies.
Collapse
Affiliation(s)
- Alessia Argirò
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy.
| | - Jeffrey Ding
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Eric Adler
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
31
|
Retson L, Tiwari N, Vaughn J, Bernes S, Adelson PD, Mansfield K, Libertini S, Kuzmiski B, Alecu I, Gabriel R, Mangum R. Epithelioid neoplasm of the spinal cord in a child with spinal muscular atrophy treated with onasemnogene abeparvovec. Mol Ther 2023; 31:2991-2998. [PMID: 37598295 PMCID: PMC10556221 DOI: 10.1016/j.ymthe.2023.08.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/02/2023] [Accepted: 08/17/2023] [Indexed: 08/21/2023] Open
Abstract
Spinal muscular atrophy is an autosomal recessive disease resulting in motor neuron degeneration and progressive life-limiting motor deficits when untreated. Onasemnogene abeparvovec is an adeno-associated virus serotype 9-based gene therapy that improves survival, motor function, and motor milestone achievement in symptomatic and presymptomatic patients. Although the adeno-associated virus genome is maintained as an episome, theoretical risk of tumorigenicity persists should genomic insertion occur. We present the case of a 16-month-old male with spinal muscular atrophy who was diagnosed with an epithelioid neoplasm of the spinal cord approximately 14 months after receiving onasemnogene abeparvovec. In situ hybridization analysis detected an onasemnogene abeparvovec nucleic acid signal broadly distributed in many but not all tumor cells. Integration site analysis on patient formalin-fixed, paraffin-embedded tumor samples failed to detect high-confidence integration sites of onasemnogene abeparvovec. The finding was considered inconclusive because of limited remaining tissue/DNA input. The improved life expectancy resulting from innovative spinal muscular atrophy therapies, including onasemnogene abeparvovec, has created an opportunity to analyze the long-term adverse events and durability of these therapies as well as identify potential disease associations that were previously unrecognized because of the premature death of these patients.
Collapse
Affiliation(s)
- Laura Retson
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ 85016, USA
| | - Nishant Tiwari
- Pathology & Laboratory Medicine, Phoenix Children's Hospital, Phoenix, AZ 85016, USA
| | - Jennifer Vaughn
- Department of Radiology, Phoenix Children's Hospital, Phoenix, AZ 85016, USA
| | - Saunder Bernes
- Department of Neurology, Phoenix Children's Hospital, Phoenix, AZ 85016, USA
| | - P David Adelson
- Department of Neurosurgery, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Keith Mansfield
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Silvana Libertini
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Brent Kuzmiski
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Iulian Alecu
- Novartis Pharmaceuticals, 4056 Basel, Switzerland
| | | | - Ross Mangum
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ 85016, USA; Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, USA; Creighton University School of Medicine, Phoenix, AZ 85012, USA; Mayo Clinic Alix School of Medicine, Phoenix, AZ 85054, USA.
| |
Collapse
|
32
|
Moço PD, Farnós O, Sharon D, Kamen AA. Targeted Delivery of Chimeric Antigen Receptor into T Cells via CRISPR-Mediated Homology-Directed Repair with a Dual-AAV6 Transduction System. Curr Issues Mol Biol 2023; 45:7705-7720. [PMID: 37886930 PMCID: PMC10605174 DOI: 10.3390/cimb45100486] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023] Open
Abstract
CAR-T cell therapy involves genetically engineering T cells to recognize and attack tumour cells by adding a chimeric antigen receptor (CAR) to their surface. In this study, we have used dual transduction with AAV serotype 6 (AAV6) to integrate an anti-CD19 CAR into human T cells at a known genomic location. The first viral vector expresses the Cas9 endonuclease and a guide RNA (gRNA) targeting the T cell receptor alpha constant locus, while the second vector carries the DNA template for homology-mediated CAR insertion. We evaluated three gRNA candidates and determined their efficiency in generating indels. The AAV6 successfully delivered the CRISPR/Cas9 machinery in vitro, and molecular analysis of the dual transduction showed the integration of the CAR transgene into the desired location. In contrast to the random integration methods typically used to generate CAR-T cells, targeted integration into a known genomic locus can potentially lower the risk of insertional mutagenesis and provide more stable levels of CAR expression. Critically, this method also results in the knockout of the endogenous T cell receptor, allowing target cells to be derived from allogeneic donors. This raises the exciting possibility of "off-the-shelf" universal immunotherapies that would greatly simplify the production and administration of CAR-T cells.
Collapse
Affiliation(s)
| | | | | | - Amine A. Kamen
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada; (P.D.M.)
| |
Collapse
|
33
|
Li S, Mereby SA, Rothstein M, Johnson MR, Brack BJ, Mallarino R. TIGER: Single-step in vivo genome editing in a non-traditional rodent. Cell Rep 2023; 42:112980. [PMID: 37573509 PMCID: PMC10528174 DOI: 10.1016/j.celrep.2023.112980] [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: 06/02/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023] Open
Abstract
Rodents are taxonomically diverse and have evolved a variety of traits. A mechanistic understanding of such traits has remained elusive, however, largely because genome editing in non-traditional model species remains challenging. Here, using the African striped mouse (Rhabdomys pumilio), we describe TIGER (targeted in vivo genome editing in rodents), a method that relies on a simple intraoviductal injecting technique and uses recombinant adeno-associated viruses (rAAVs) as the sole vehicle to deliver reagents into pregnant females. We demonstrate that TIGER generates knockout and knockin (up to 3 kb) lines with high efficiency. Moreover, we engineer a double-cleaving repair rAAV template and find that it significantly increases knockin frequency and germline transmission rates. Lastly, we show that an oversized double-cleaving rAAV template leads to an insertion of 3.8 kb. Thus, TIGER constitutes an attractive alternative to traditional ex vivo genome-editing methods and has the potential to be extended to a broad range of species.
Collapse
Affiliation(s)
- Sha Li
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | - Sarah A Mereby
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | - Megan Rothstein
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | - Matthew R Johnson
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | - Benjamin J Brack
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | - Ricardo Mallarino
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA.
| |
Collapse
|
34
|
De Giorgi M, Park SH, Castoreno A, Cao M, Hurley A, Saxena L, Chuecos MA, Walkey CJ, Doerfler AM, Furgurson MN, Ljungberg MC, Patel KR, Hyde S, Chickering T, Lefebvre S, Wassarman K, Miller P, Qin J, Schlegel MK, Zlatev I, Li RG, Kim J, Martin JF, Bissig KD, Jadhav V, Bao G, Lagor WR. In vivo expansion of gene-targeted hepatocytes through transient inhibition of an essential gene. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.26.550728. [PMID: 37546995 PMCID: PMC10402145 DOI: 10.1101/2023.07.26.550728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Homology Directed Repair (HDR)-based genome editing is an approach that could permanently correct a broad range of genetic diseases. However, its utility is limited by inefficient and imprecise DNA repair mechanisms in terminally differentiated tissues. Here, we tested "Repair Drive", a novel method for improving targeted gene insertion in the liver by selectively expanding correctly repaired hepatocytes in vivo. Our system consists of transient conditioning of the liver by knocking down an essential gene, and delivery of an untargetable version of the essential gene in cis with a therapeutic transgene. We show that Repair Drive dramatically increases the percentage of correctly targeted hepatocytes, up to 25%. This resulted in a five-fold increased expression of a therapeutic transgene. Repair Drive was well-tolerated and did not induce toxicity or tumorigenesis in long term follow up. This approach will broaden the range of liver diseases that can be treated with somatic genome editing.
Collapse
Affiliation(s)
- Marco De Giorgi
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - So Hyun Park
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Adam Castoreno
- Alnylam Pharmaceuticals Inc, 675 W Kendall St, Cambridge, MA 02142, USA
| | - Mingming Cao
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Ayrea Hurley
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lavanya Saxena
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Marcel A. Chuecos
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Christopher J. Walkey
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexandria M. Doerfler
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mia N. Furgurson
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - M. Cecilia Ljungberg
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Kalyani R. Patel
- Department of Pathology, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Sarah Hyde
- Alnylam Pharmaceuticals Inc, 675 W Kendall St, Cambridge, MA 02142, USA
| | - Tyler Chickering
- Alnylam Pharmaceuticals Inc, 675 W Kendall St, Cambridge, MA 02142, USA
| | | | - Kelly Wassarman
- Alnylam Pharmaceuticals Inc, 675 W Kendall St, Cambridge, MA 02142, USA
| | - Patrick Miller
- Alnylam Pharmaceuticals Inc, 675 W Kendall St, Cambridge, MA 02142, USA
| | - June Qin
- Alnylam Pharmaceuticals Inc, 675 W Kendall St, Cambridge, MA 02142, USA
| | - Mark K. Schlegel
- Alnylam Pharmaceuticals Inc, 675 W Kendall St, Cambridge, MA 02142, USA
| | - Ivan Zlatev
- Alnylam Pharmaceuticals Inc, 675 W Kendall St, Cambridge, MA 02142, USA
| | - Rich Gang Li
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Heart Institute, Houston, TX 77030, USA
| | - Jong Kim
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Heart Institute, Houston, TX 77030, USA
| | - James F. Martin
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Heart Institute, Houston, TX 77030, USA
| | - Karl-Dimiter Bissig
- Department of Pediatrics, Alice and Y. T. Chen Center for Genetics and Genomics, Division of Medical Genetics, Duke University, Durham, NC 27710, USA
| | - Vasant Jadhav
- Alnylam Pharmaceuticals Inc, 675 W Kendall St, Cambridge, MA 02142, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - William R. Lagor
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
35
|
Oziolor EM, Kumpf SW, Qian J, Gosink M, Sheehan M, Rubitski DM, Newman L, Whiteley LO, Lanz TA. Comparing molecular and computational approaches for detecting viral integration of AAV gene therapy constructs. Mol Ther Methods Clin Dev 2023; 29:395-405. [PMID: 37251978 PMCID: PMC10209688 DOI: 10.1016/j.omtm.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 04/28/2023] [Indexed: 05/31/2023]
Abstract
Many current gene therapy targets use recombinant adeno-associated virus (AAV). The majority of delivered AAV therapeutics persist as episomes, separate from host DNA, yet some viral DNA can integrate into host DNA in different proportions and at genomic locations. The potential for viral integration leading to oncogenic transformation has led regulatory agencies to require investigation into AAV integration events following gene therapy in preclinical species. In the present study, tissues were collected from cynomolgus monkeys and mice 6 and 8 weeks, respectively, following administration of an AAV vector delivering transgene cargo. We compared three different next-generation sequencing approaches (shearing extension primer tag selection ligation-mediated PCR, targeted enrichment sequencing [TES], and whole-genome sequencing) to contrast the specificity, scope, and frequency of integration detected by each method. All three methods detected dose-dependent insertions with a limited number of hotspots and expanded clones. While the functional outcome was similar for all three methods, TES was the most cost-effective and comprehensive method of detecting viral integration. Our findings aim to inform the direction of molecular efforts to ensure a thorough hazard assessment of AAV viral integration in our preclinical gene therapy studies.
Collapse
Affiliation(s)
- Elias M. Oziolor
- Global Computational Safety Sciences, Pfizer Inc., Groton, CT 06340, USA
| | - Steven W. Kumpf
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc., Groton, CT 06340, USA
| | - Jessie Qian
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc., Groton, CT 06340, USA
| | - Mark Gosink
- Global Computational Safety Sciences, Pfizer Inc., Groton, CT 06340, USA
| | - Mark Sheehan
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc., Groton, CT 06340, USA
| | - David M. Rubitski
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc., Groton, CT 06340, USA
| | - Leah Newman
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc., Groton, CT 06340, USA
| | | | - Thomas A. Lanz
- Global Discovery, Investigative and Translational Sciences, Pfizer Inc., Groton, CT 06340, USA
| |
Collapse
|
36
|
Deschenes NM, Cheng C, Ryckman AE, Quinville BM, Khanal P, Mitchell M, Chen Z, Sangrar W, Gray SJ, Walia JS. Biochemical Correction of GM2 Ganglioside Accumulation in AB-Variant GM2 Gangliosidosis. Int J Mol Sci 2023; 24:ijms24119217. [PMID: 37298170 DOI: 10.3390/ijms24119217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/09/2023] [Accepted: 05/15/2023] [Indexed: 06/12/2023] Open
Abstract
GM2 gangliosidosis is a group of genetic disorders that result in the accumulation of GM2 ganglioside (GM2) in brain cells, leading to progressive central nervous system (CNS) atrophy and premature death in patients. AB-variant GM2 gangliosidosis (ABGM2) arises from loss-of-function mutations in the GM2 activator protein (GM2AP), which is essential for the breakdown of GM2 in a key catabolic pathway required for CNS lipid homeostasis. In this study, we show that intrathecal delivery of self-complementary adeno-associated virus serotype-9 (scAAV9) harbouring a functional human GM2A transgene (scAAV9.hGM2A) can prevent GM2 accumulation in in GM2AP-deficient mice (Gm2a-/- mice). Additionally, scAAV9.hGM2A efficiently distributes to all tested regions of the CNS within 14 weeks post-injection and remains detectable for the lifespan of these animals (up to 104 weeks). Remarkably, GM2AP expression from the transgene scales with increasing doses of scAAV9.hGM2A (0.5, 1.0 and 2.0 × 1011 vector genomes (vg) per mouse), and this correlates with dose-dependent correction of GM2 accumulation in the brain. No severe adverse events were observed, and comorbidities in treated mice were comparable to those in disease-free cohorts. Lastly, all doses yielded corrective outcomes. These data indicate that scAAV9.hGM2A treatment is relatively non-toxic and tolerable, and biochemically corrects GM2 accumulation in the CNS-the main cause of morbidity and mortality in patients with ABGM2. Importantly, these results constitute proof-of-principle for treating ABGM2 with scAAV9.hGM2A by means of a single intrathecal administration and establish a foundation for future preclinical research.
Collapse
Affiliation(s)
- Natalie M Deschenes
- Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Camilyn Cheng
- Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Alex E Ryckman
- Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Brianna M Quinville
- Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Prem Khanal
- Department of Pediatrics, Queen's University, Kingston, ON K7L 2V7, Canada
| | - Melissa Mitchell
- Department of Pediatrics, Queen's University, Kingston, ON K7L 2V7, Canada
| | - Zhilin Chen
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Waheed Sangrar
- Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Steven J Gray
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jagdeep S Walia
- Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
- Department of Pediatrics, Queen's University, Kingston, ON K7L 2V7, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| |
Collapse
|
37
|
Major L, McClements ME, MacLaren RE. A Review of CRISPR Tools for Treating Usher Syndrome: Applicability, Safety, Efficiency, and In Vivo Delivery. Int J Mol Sci 2023; 24:ijms24087603. [PMID: 37108761 PMCID: PMC10146473 DOI: 10.3390/ijms24087603] [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: 03/10/2023] [Revised: 04/12/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
This review considers research into the treatment of Usher syndrome, a deaf-blindness syndrome inherited in an autosomal recessive manner. Usher syndrome mutations are markedly heterogeneous, involving many different genes, and research grants are limited due to minimal patient populations. Furthermore, gene augmentation therapies are impossible in all but three Usher syndromes as the cDNA sequence exceeds the 4.7 kb AAV packaging limit. It is, therefore, vital to focus research efforts on alternative tools with the broadest applicability. The CRISPR field took off in recent years following the discovery of the DNA editing activity of Cas9 in 2012. New generations of CRISPR tools have succeeded the original CRISPR/Cas9 model to enable more sophisticated genomic amendments such as epigenetic modification and precise sequence alterations. This review will evaluate the most popular CRISPR tools to date: CRISPR/Cas9, base editing, and prime editing. It will consider these tools in terms of applicability (in relation to the ten most prevalent USH2A mutations), safety, efficiency, and in vivo delivery potential with the intention of guiding future research investment.
Collapse
Affiliation(s)
- Lauren Major
- Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Michelle E McClements
- Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Robert E MacLaren
- Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| |
Collapse
|
38
|
Issa SS, Shaimardanova AA, Solovyeva VV, Rizvanov AA. Various AAV Serotypes and Their Applications in Gene Therapy: An Overview. Cells 2023; 12:785. [PMID: 36899921 PMCID: PMC10000783 DOI: 10.3390/cells12050785] [Citation(s) in RCA: 130] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/22/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Despite scientific discoveries in the field of gene and cell therapy, some diseases still have no effective treatment. Advances in genetic engineering methods have enabled the development of effective gene therapy methods for various diseases based on adeno-associated viruses (AAVs). Today, many AAV-based gene therapy medications are being investigated in preclinical and clinical trials, and new ones are appearing on the market. In this article, we present a review of AAV discovery, properties, different serotypes, and tropism, and a following detailed explanation of their uses in gene therapy for disease of different organs and systems.
Collapse
Affiliation(s)
- Shaza S. Issa
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Alisa A. Shaimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| |
Collapse
|
39
|
Heller G, Bradbury AM, Sands MS, Bongarzone ER. Preclinical studies in Krabbe disease: A model for the investigation of novel combination therapies for lysosomal storage diseases. Mol Ther 2023; 31:7-23. [PMID: 36196048 PMCID: PMC9840155 DOI: 10.1016/j.ymthe.2022.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 08/16/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022] Open
Abstract
Krabbe disease (KD) is a lysosomal storage disease (LSD) caused by mutations in the galc gene. There are over 50 monogenetic LSDs, which largely impede the normal development of children and often lead to premature death. At present, there are no cures for LSDs and the available treatments are generally insufficient, short acting, and not without co-morbidities or long-term side effects. The last 30 years have seen significant advances in our understanding of LSD pathology as well as treatment options. Two gene therapy-based clinical trials, NCT04693598 and NCT04771416, for KD were recently started based on those advances. This review will discuss how our knowledge of KD got to where it is today, focusing on preclinical investigations, and how what was discovered may prove beneficial for the treatment of other LSDs.
Collapse
Affiliation(s)
- Gregory Heller
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, 808 S. Wood St M/C 512, Chicago, IL, USA.
| | - Allison M Bradbury
- Center for Gene Therapy, Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Abigail Wexner Research Institute Nationwide Children's Hospital Department of Pediatrics, The Ohio State University, Wexner Medical Center, Columbus, OH 43205, USA.
| | - Mark S Sands
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue Box 8007, St. Louis, MO, USA; Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue Box 8007, St. Louis, MO, USA.
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, 808 S. Wood St M/C 512, Chicago, IL, USA.
| |
Collapse
|
40
|
Zheng Z, Ye J, Leng M, Gan C, Tang N, Li W, Valencia CA, Dong B, Chow HY. Enhanced sensitivity of neutralizing antibody detection for different AAV serotypes using HeLa cells with overexpressed AAVR. Front Pharmacol 2023; 14:1188290. [PMID: 37188274 PMCID: PMC10176094 DOI: 10.3389/fphar.2023.1188290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
A cell-based transduction inhibition assay (TI) is widely used in clinical trials to detect neutralizing antibody (NAb) titers against recombinant adeno-associated virus (rAAV), one of the most important criteria to exclude patients in gene therapy. Different cell lines are used in cell-based TI because the rAAV transduction efficiencies vary largely among serotypes. A cell line suitable for TI for most serotypes is highly desirable, especially for those with very low transduction efficiencies in vitro such as rAAV8 and rAAV9. Herein, we report an AAVR-HeLa, a stable cell line with overexpressed AAVR, a newly identified receptor for rAAVs, was established for cell-based TIs. The AAVR expression level in AAVR-HeLa cells was approximately 10-fold higher than in HeLa cells, and was stably transfected after twenty three passages. For all AAV serotypes (AAV1-10), except for AAV4, the transduction efficiencies increased significantly in AAVR-HeLa cells. It was demonstrated that the AAVR enhancement of transduction efficiency was only for rAAV and not for lentiviral and adenoviral vectors. According to the minimal multiplicity of infection (MOIs) for the assay, the NAb detection sensitivity increased at least 10 and 20 fold for AAV8 and AAV9, respectively. The seroprevalence of NAbs were investigated at the 1:30 level as a cutoff value using AAVR-HeLa cells. It was shown that the seropositive rate for AAV2 was 87% in serum samples from 99 adults, followed by lower seropositive rates for AAV5 (7%), AAV8 (7%) and AAV9 (1%). Venn diagram analysis showed the presence of cross-reactivity of NAbs to two or three serotypes in 13 samples (13.1%). However, no patient was found to possess NAbs for all the four serotypes. These results demonstrated that the AAVR-HeLa cell line may be utilized to detect the NAbs through cell-based TI assays for most of AAV serotypes.
Collapse
Affiliation(s)
- Zhaoyue Zheng
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jingya Ye
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Mi Leng
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chunmei Gan
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Na Tang
- Sichuan Real and Best Biotech Co., Ltd., Chengdu, China
| | - Wei Li
- Department of Dermatovenereology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - C. Alexander Valencia
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Biao Dong
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Real and Best Biotech Co., Ltd., Chengdu, China
- *Correspondence: Hoi Yee Chow, ; Biao Dong,
| | - Hoi Yee Chow
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Hoi Yee Chow, ; Biao Dong,
| |
Collapse
|
41
|
Ebner LJA, Grimm C. AAV Serotypes and Their Suitability for Retinal Gene Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:131-134. [PMID: 37440025 DOI: 10.1007/978-3-031-27681-1_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Throughout the last 25 years, exceptional progress in retinal gene therapy was achieved. The major breakthrough was realized in 2017 when the FDA approved the adeno-associated virus (AAV)-based gene therapy for treatment of the monogenetic disorder Leber congenital amaurosis type 2 (LCA2). Since then, many therapies for inherited retinal diseases (IRD) reached phase I/II clinical trials, targeting diseases like achromatopsia, choroideremia, retinitis pigmentosa, Stargardt disease, and many more (reviewed in (Trapani and Auricchio, Trends Mol Med 24:669-681, 2018)). Advanced vector and capsid design technologies as well as improved gene transfer and gene editing methods may lead to refined therapies for various eye diseases. Many research departments worldwide focus on optimizing transgene expression by designing novel AAV serotypes. Besides serotype tropism, the method of injection (intravitreal, subretinal, or suprachoroidal) (Han et al., Hum Gene Ther 31:1288-1299, 2020) defines the efficiency outcome along with the use of tissue-specific promotors which play a critical role for cell targeting.
Collapse
Affiliation(s)
- Lynn J A Ebner
- Lab for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland.
| | - Christian Grimm
- Lab for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren, Switzerland
| |
Collapse
|
42
|
A link between severe hepatitis in children and adenovirus 41 and adeno-associated virus 2 infections. J Gen Virol 2022; 103. [DOI: 10.1099/jgv.0.001783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Over the past few months there have been reports of severe acute hepatitis in several hundred, otherwise healthy, immunocompetent young children. Several deaths have been recorded and a relatively large proportion of the patients have needed liver transplants. Most of the cases, so far, have been seen in the UK and in North America, but it has also been reported in many other European countries, the Middle East and Asia. Most common viruses have been ruled out as a causative agent; hepatitis A virus (HAV), hepatitis B virus (HBV) and hepatitis C virus (HCV) were not detected, nor were Epstein–Barr virus (EBV), cytomegalovirus (CMV) and human immunodeficiency virus (HIV) in many cases. A small proportion of the children had been infected with SARS-CoV-2 but these seem to be in a minority; similarly, almost none of the children had been vaccinated against COVID-19. Significantly, many of the patients were infected with adenovirus 41 (HAdV-F41). Previously, HAdV-41 had not been linked to hepatitis and is usually considered to cause gastroenteritis in both immunocompetent and immunocompromised patients. In two most recent studies, adeno-associated virus 2 (AAV2) was detected in almost all patients, together with species C and F HAdVs and human herpesvirus 6B (HHV6B). Here, I discuss the possibility that a change in tropism of HAdV-41 and changes in AAV2 may be responsible for their links to acute hepatitis.
Collapse
|
43
|
Sant’Anna TB, Araujo NM. Adeno-associated virus infection and its impact in human health: an overview. Virol J 2022; 19:173. [PMID: 36316711 PMCID: PMC9623951 DOI: 10.1186/s12985-022-01900-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 01/24/2023] Open
Abstract
Discovered as a contaminant of adenovirus stocks in the 1960s, adeno-associated virus (AAV) is a mono-stranded DNA virus that depends on helper factors to replicate. Even though AAV is endemic in the human population (35-80%), it is remarkable that many issues concerning the natural infection by this virus remain unanswered. In this study, we reflect on the main basic aspects of AAV biology and provide an overview of the studies exploring the impact of AAV infection on human health, focusing on three major research areas including, (i) cervical and (ii) liver cancer, and (iii) reproductive system disorders. Conflicting results have been obtained into the association of AAV infection with the occurrence of adverse reproductive outcomes, such as placental complications, spontaneous abortion, and fertility disorders, or with a protective role in HPV-related cervical carcinogenesis. Noteworthy, recent reports have identified AAV insertional mutagenesis as a novel risk factor for the development of hepatocellular carcinoma. This latest finding raises concern regarding the widespread usage of AAV vectors in liver-targeted gene therapy.
Collapse
Affiliation(s)
- Thaís B Sant’Anna
- grid.418068.30000 0001 0723 0931Laboratory of Molecular Virology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Natalia M Araujo
- grid.418068.30000 0001 0723 0931Laboratory of Molecular Virology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| |
Collapse
|
44
|
Ferrari S, Jacob A, Cesana D, Laugel M, Beretta S, Varesi A, Unali G, Conti A, Canarutto D, Albano L, Calabria A, Vavassori V, Cipriani C, Castiello MC, Esposito S, Brombin C, Cugnata F, Adjali O, Ayuso E, Merelli I, Villa A, Di Micco R, Kajaste-Rudnitski A, Montini E, Penaud-Budloo M, Naldini L. Choice of template delivery mitigates the genotoxic risk and adverse impact of editing in human hematopoietic stem cells. Cell Stem Cell 2022; 29:1428-1444.e9. [PMID: 36206730 PMCID: PMC9550218 DOI: 10.1016/j.stem.2022.09.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 07/18/2022] [Accepted: 09/06/2022] [Indexed: 12/14/2022]
Abstract
Long-range gene editing by homology-directed repair (HDR) in hematopoietic stem/progenitor cells (HSPCs) often relies on viral transduction with recombinant adeno-associated viral vector (AAV) for template delivery. Here, we uncover unexpected load and prolonged persistence of AAV genomes and their fragments, which trigger sustained p53-mediated DNA damage response (DDR) upon recruiting the MRE11-RAD50-NBS1 (MRN) complex on the AAV inverted terminal repeats (ITRs). Accrual of viral DNA in cell-cycle-arrested HSPCs led to its frequent integration, predominantly in the form of transcriptionally competent ITRs, at nuclease on- and off-target sites. Optimized delivery of integrase-defective lentiviral vector (IDLV) induced lower DNA load and less persistent DDR, improving clonogenic capacity and editing efficiency in long-term repopulating HSPCs. Because insertions of viral DNA fragments are less frequent with IDLV, its choice for template delivery mitigates the adverse impact and genotoxic burden of HDR editing and should facilitate its clinical translation in HSPC gene therapy.
Collapse
Affiliation(s)
- Samuele Ferrari
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy,Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Aurelien Jacob
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Daniela Cesana
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Marianne Laugel
- INSERM UMR 1089, University of Nantes, CHU of Nantes, Nantes 44200, France
| | - Stefano Beretta
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Angelica Varesi
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Giulia Unali
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Anastasia Conti
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Daniele Canarutto
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy,Vita-Salute San Raffaele University, Milan 20132, Italy,Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Luisa Albano
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Andrea Calabria
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Valentina Vavassori
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Carlo Cipriani
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Maria Carmina Castiello
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy,Institute for Genetic and Biomedical Research (UOS Milan Unit), National Research Council, Milan 20132, Italy
| | - Simona Esposito
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Chiara Brombin
- University Center for Statistics in the Biomedical Sciences, Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Federica Cugnata
- University Center for Statistics in the Biomedical Sciences, Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Oumeya Adjali
- INSERM UMR 1089, University of Nantes, CHU of Nantes, Nantes 44200, France
| | - Eduard Ayuso
- INSERM UMR 1089, University of Nantes, CHU of Nantes, Nantes 44200, France
| | - Ivan Merelli
- Institute for Biomedical Technologies, National Research Council, Segrate 20090, Italy
| | - Anna Villa
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy,Institute for Genetic and Biomedical Research (UOS Milan Unit), National Research Council, Milan 20132, Italy
| | - Raffaella Di Micco
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Anna Kajaste-Rudnitski
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Eugenio Montini
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | | | - Luigi Naldini
- San Rafaelle Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy,Vita-Salute San Raffaele University, Milan 20132, Italy,Corresponding author
| |
Collapse
|
45
|
Ramamurthy RM, Atala A, Porada CD, Almeida-Porada G. Organoids and microphysiological systems: Promising models for accelerating AAV gene therapy studies. Front Immunol 2022; 13:1011143. [PMID: 36225917 PMCID: PMC9549755 DOI: 10.3389/fimmu.2022.1011143] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/01/2022] [Indexed: 11/24/2022] Open
Abstract
The FDA has predicted that at least 10-20 gene therapy products will be approved by 2025. The surge in the development of such therapies can be attributed to the advent of safe and effective gene delivery vectors such as adeno-associated virus (AAV). The enormous potential of AAV has been demonstrated by its use in over 100 clinical trials and the FDA’s approval of two AAV-based gene therapy products. Despite its demonstrated success in some clinical settings, AAV-based gene therapy is still plagued by issues related to host immunity, and recent studies have suggested that AAV vectors may actually integrate into the host cell genome, raising concerns over the potential for genotoxicity. To better understand these issues and develop means to overcome them, preclinical model systems that accurately recapitulate human physiology are needed. The objective of this review is to provide a brief overview of AAV gene therapy and its current hurdles, to discuss how 3D organoids, microphysiological systems, and body-on-a-chip platforms could serve as powerful models that could be adopted in the preclinical stage, and to provide some examples of the successful application of these models to answer critical questions regarding AAV biology and toxicity that could not have been answered using current animal models. Finally, technical considerations while adopting these models to study AAV gene therapy are also discussed.
Collapse
|
46
|
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
|
47
|
Zhang H, Zhan Q, Huang B, Wang Y, Wang X. AAV-mediated gene therapy: Advancing cardiovascular disease treatment. Front Cardiovasc Med 2022; 9:952755. [PMID: 36061546 PMCID: PMC9437345 DOI: 10.3389/fcvm.2022.952755] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Gene therapy has revolutionized the field of medicine, offering new hope for those with common and rare diseases. For nearly three decades, adeno-associated virus (AAV) has shown significant therapeutic benefits in multiple clinical trials, mainly due to its unique replication defects and non-pathogenicity in humans. In the field of cardiovascular disease (CVD), compared with non-viral vectors, lentiviruses, poxviruses, and adenovirus vectors, AAV possesses several advantages, including high security, low immunogenicity, sustainable and stable exogenous gene expression etc., which makes AAV one of the most promising candidates for the treatment of many genetic disorders and hereditary diseases. In this review, we evaluate the current information on the immune responses, transport pathways, and mechanisms of action associated with AAV-based CVD gene therapies and further explore potential optimization strategies to improve the efficiency of AAV transduction for the improved safety and efficiency of CVD treatment. In conclusion, AAV-mediated gene therapy has great potential for development in the cardiovascular system.
Collapse
Affiliation(s)
- Huili Zhang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
- Oncology Department, Zhejiang Xiaoshan HospitaI, Hangzhou, China
| | - Qi Zhan
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yigang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
- Yigang Wang
| | - Xiaoyan Wang
- Oncology Department, Zhejiang Xiaoshan HospitaI, Hangzhou, China
- *Correspondence: Xiaoyan Wang
| |
Collapse
|
48
|
Sabatino DE, Bushman FD, Chandler RJ, Crystal RG, Davidson BL, Dolmetsch R, Eggan KC, Gao G, Gil-Farina I, Kay MA, McCarty DM, Montini E, Ndu A, Yuan J. Evaluating the state of the science for adeno-associated virus integration: An integrated perspective. Mol Ther 2022; 30:2646-2663. [PMID: 35690906 PMCID: PMC9372310 DOI: 10.1016/j.ymthe.2022.06.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 12/12/2022] Open
Abstract
On August 18, 2021, the American Society of Gene and Cell Therapy (ASGCT) hosted a virtual roundtable on adeno-associated virus (AAV) integration, featuring leading experts in preclinical and clinical AAV gene therapy, to further contextualize and understand this phenomenon. Recombinant AAV (rAAV) vectors are used to develop therapies for many conditions given their ability to transduce multiple cell types, resulting in long-term expression of transgenes. Although most rAAV DNA typically remains episomal, some rAAV DNA becomes integrated into genomic DNA at a low frequency, and rAAV insertional mutagenesis has been shown to lead to tumorigenesis in neonatal mice. Currently, the risk of rAAV-mediated oncogenesis in humans is theoretical because no confirmed genotoxic events have been reported to date. However, because insertional mutagenesis has been reported in a small number of murine studies, there is a need to characterize this genotoxicity to inform research, regulatory needs, and patient care. The purpose of this white paper is to review the evidence of rAAV-related host genome integration in animal models and possible risks of insertional mutagenesis in patients. In addition, technical considerations, regulatory guidance, and bioethics are discussed.
Collapse
Affiliation(s)
- Denise E Sabatino
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Hematology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Randy J Chandler
- National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, NY, USA
| | - Beverly L Davidson
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Mark A Kay
- Departments of Pediatrics and Genetics, Stanford University, Stanford, CA, USA
| | | | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Adora Ndu
- BridgeBio Pharma, Inc., Palo Alto, CA, USA
| | - Jing Yuan
- Drug Safety Research and Development, Pfizer Inc., Cambridge, MA, USA
| |
Collapse
|
49
|
Majeau N, Fortin-Archambault A, Gérard C, Rousseau J, Yamégo P, Tremblay JP. SERUM EXTRACELLULAR VESICLES FOR DELIVERY OF CRISPR-CAS9 RIBONUCLEOPROTEINS TO MODIFY THE DYSTROPHIN GENE. Mol Ther 2022; 30:2429-2442. [PMID: 35619556 DOI: 10.1016/j.ymthe.2022.05.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 05/19/2022] [Accepted: 05/21/2022] [Indexed: 01/19/2023] Open
Abstract
Extracellular vesicles (EVs) mediate intercellular biomolecule exchanges in the body, making them promising delivery vehicles for therapeutic cargo. Genetic engineering by CRISPR system is an interesting therapeutic avenue for genetic diseases such as Duchenne Muscular Dystrophy (DMD). We developed a simple method for loading EVs with CRISPR ribonucleoproteins (RNPs) consisting of SpCas9 proteins and guide RNAs (gRNAs). EVs were first purified from human or mouse serum using ultrafiltration and size-exclusion chromatography. Using protein transfectant to load RNPs into serum EVs, we showed that EVs are good carriers of RNPs in vitro and restored the expression of the tdTomato fluorescent protein in muscle fibers of Ai9 mice. EVs carrying RNPs targeting introns 22 and 24 of the DMD gene were also injected into muscles of mdx mice having a non-sense mutation in exon 23. Up to 19% of the cDNA extracted from treated mdx mice had the intended deletion of exons 23 and 24, allowing dystrophin expression in muscle fibers. RNPs alone, without EVs, were inefficient in generating detectable deletions in mouse muscles. This method opens new opportunities for rapid and safe delivery of CRISPR components to treat DMD.
Collapse
Affiliation(s)
- Nathalie Majeau
- Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval Québec, Québec, QC, Canada
| | - Annabelle Fortin-Archambault
- Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval Québec, Québec, QC, Canada
| | - Catherine Gérard
- Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval Québec, Québec, QC, Canada
| | - Joël Rousseau
- Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval Québec, Québec, QC, Canada
| | - Pouiré Yamégo
- Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval Québec, Québec, QC, Canada
| | - Jacques P Tremblay
- Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval Québec, Québec, QC, Canada.
| |
Collapse
|
50
|
Collin V, Flamand L. [The importance of telomeres in human herpesvirus-6A/B infections]. Med Sci (Paris) 2022; 38:168-176. [PMID: 35179471 DOI: 10.1051/medsci/2022008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Herpesviruses are undisputed masters of disguise. The ability to become invisible to the immune system effectors is a complex process resting on a variety of stealth approaches. Among these, human herpesviruses-6A and -6B (HHV-6A/B) have developed the unique ability to integrate their genome within the ends of chromosomes allowing viral persistence in the absence of viral protein expression. This aptitude, unique to HHV-6A/B among human herpesviruses, requires close interactions between the telomeric regions of chromosomes and the viral genome. In this review article, the biology of telomeres and the mechanisms responsible for viral integration are discussed. In closing, the possible biological consequences of HHV-6A/B integration into chromosomal DNA are discussed.
Collapse
Affiliation(s)
- Vanessa Collin
- Axe des maladies infectieuses et immunitaires, Centre de recherche du CHU de Québec - Université Laval, Québec, 2705 boulevard Laurier, Québec, Canada
| | - Louis Flamand
- Axe des maladies infectieuses et immunitaires, Centre de recherche du CHU de Québec - Université Laval, Québec, 2705 boulevard Laurier, Québec, Canada - Département de microbiologie, maladies infectieuses et immunologie, Faculté de médecine, Université Laval, Québec, 2705 boulevard Laurier, Québec, Canada
| |
Collapse
|