1
|
Chen T, Chen Q, Ye J, Wu Y, Liu T, Zhang Y. Postmarketing adverse events associated with onasemnogene abeparvovec: a real-world pharmacovigilance study. Orphanet J Rare Dis 2025; 20:215. [PMID: 40329332 PMCID: PMC12057001 DOI: 10.1186/s13023-025-03715-2] [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: 10/15/2023] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Onasemnogene abeparvovec (OA) is an adeno-associated virus vector-based gene therapy indicated for the treatment of paediatric patients with spinal muscular atrophy(SMA) with biallelic mutations in the survival motor neuron 1 (SMN1) gene. This study focused on analysis of the postmarketing adverse events(AEs) of onasemnogene abeparvovec (OA) reported in the US Food and Drug Administration public data open project (openFDA) database to assess the safety of OA in the real world and to provide a reference for the rational use of this drug in the clinic. RESULTS In total, 1,959 AEs were reported with "onasemnogene abeparvovec" as the primary suspected drug. The top 5 most frequent AEs were pyrexia (461 cases), vomiting (434 cases), aspartate aminotransferase increase (284 cases), alanine aminotransferase increase (260 cases), and hepatic enzyme increase (237 cases). A total of 77 alert signals were generated, 60 of which were not included in the drug label. The top 5 signals included troponin I increase ( ROR of 895.21, 95% CI: 734.43-1091.18), troponin T increase ( ROR of 313.30, 95% CI:220.85-444.44), rhinovirus infection ( ROR of 175.80, 95% CI:130.86-236.17), troponin increase ( ROR of 143.49, 95% CI:114.96-179.10), and increased bronchial secretion ( ROR of 142.71, 95% CI:96.63-210.77). Further analysis of AEs associated with gender and age differences identified 14 high-risk signals related to gender and 10 high-risk signals related to age. Female patients should be vigilant for vomiting, thrombotic microangiopathy, increased troponin T, proteinuria, haematuria, haemolytic anaemia, urinary tract infection, generalised oedema, and atypical haemolytic uraemic syndrome. Male patients should be alert to increased hepatic enzyme, increased bronchial secretion, respiratory tract infection, pallor, and increased blood creatine phosphokinase MB. Patients under 2 years of age should be vigilant for lethargy, increased monocyte count, decreased blood creatinine, and decreased neutrophil count. Patients over 2 years of age should be alert to hypertension, haematuria, rhinovirus infection, increased blood creatine phosphokinase, headache, and malaise. CONCLUSIONS Mining of OA alert signals using the openFDA database provides supplementary information on AEs not included in the drug label. Clinical attention should be focused on common, strong-signal, and label-unmentioned AEs to optimise medication regimens and control risks in clinical use.
Collapse
Affiliation(s)
| | - Qiying Chen
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| | | | - Yuzhu Wu
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Ting Liu
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | | |
Collapse
|
2
|
Zhang W, Yin Y, Yang D, Liu M, Ye C, Yan R, Li R. Comprehensive analysis of adverse events associated with onasemnogene abeparvovec (Zolgensma) in spinal muscular atrophy patients: insights from FAERS database. Front Pharmacol 2025; 15:1475884. [PMID: 39840097 PMCID: PMC11747325 DOI: 10.3389/fphar.2024.1475884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025] Open
Abstract
Onasemnogene Abeparvovec (Zolgensma) is a gene therapy for the treatment of Spinal Muscular Atrophy (SMA) with improved motor neuron function and the potential for a singular treatment. Information on its adverse drug reactions is mainly from clinical trials and real-world studies with extensive sample sizes are lacking. In this study, we analyzed the U.S. Food and Drug Administration's Adverse Event Reporting System (FAERS) database to assess the drug safety profile of Zolgensma. A total of 1951 adverse event reports associated with onasemnogene abeparvovec (Zolgensma), containing 778 import important medical event (IME) signals, were identified from the FAERS database, and multiple disproportionate analysis algorithms were used to determine the significance of these adverse events. This study identified 281 onasemnogene abeparvovec-related adverse events (AEs), including some significant adverse events not mentioned in the product labelling. Elevated liver enzymes, fever, vomiting, and thrombocytopenia were the most common adverse reactions. Most adverse events manifested within the initial month of onasemnogene abeparvovec use, especially the first 8 days, but some may still occur after 1 year of treatment. Sex-specific scrutiny revealed differing risk levels for adverse events among women and men. Thrombocytopenia and thrombotic microangiopathy are more common in patients weighing ≥8.5 kg, and changes in renal function need to be closely monitored if thrombotic microangiopathy occurs. The above findings provide valuable insights into optimizing the utilization of onasemnogene abeparvovec, improving its effectiveness, and minimizing potential side effects, thereby greatly facilitating its practical application in clinical settings.
Collapse
Affiliation(s)
| | | | | | | | | | - Ruiling Yan
- Department of Fetal Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ruiman Li
- Department of Fetal Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
3
|
Emami MR, Brimble MA, Espinoza A, Owens J, Whiteley LO, Casinghino S, Lanz TA, Farahat PK, Pellegrini M, Young CS, Thomas PG, McNally EM, Villalta SA, Schattgen SA, Spencer MJ. Single cell and TCR analysis of immune cells from AAV gene therapy-dosed Duchenne muscular dystrophy patients. Mol Ther Methods Clin Dev 2024; 32:101349. [PMID: 39524974 PMCID: PMC11546459 DOI: 10.1016/j.omtm.2024.101349] [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: 02/09/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024]
Abstract
Clinical trials for Duchenne muscular dystrophy (DMD) are assessing the therapeutic efficacy of systemically delivered adeno-associated virus (AAV) carrying a modified DMD transgene. High vector doses (>1E14 vg/kg) are needed to globally transduce skeletal muscles; however, such doses trigger immune-related adverse events. Mitigating these immune responses is crucial for widespread application of AAV-based therapies. We used single-cell RNA sequencing and T cell receptor (TCR) sequencing on peripheral blood mononuclear cells from five participants prior to, and after, dosing. One subject in the high-dose cohort experienced thrombotic microangiopathy (TMA). Few changes in cell frequencies occurred after treatment; however, differential gene expression demonstrated induction of interferon response genes in most T cell types. T cell clonotype and clumping analysis showed the expansion or appearance of groups of related TCR sequences in the post-treatment samples. Three of these expanded clumps could be assigned to prior human herpesvirus infections, two of which were present in the participant that exhibited TMA. These data provide insight on the mechanistic basis of human immune-AAV interactions and lay a foundation for improved understanding of why TMA arises in some patients and not others.
Collapse
Affiliation(s)
- Michael R. Emami
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- MyoGene Bio, San Diego, CA 92121, USA
| | - Mark A. Brimble
- Department of Host-Microbe Interactions, St Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Alejandro Espinoza
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jane Owens
- Pfizer Inc., Rare Disease Research Unit, Cambridge, MA 02139, USA
| | | | - Sandra Casinghino
- Pfizer Inc., Drug Safety Research & Development, Groton, CT 06340, USA
| | - Thomas A. Lanz
- Pfizer Inc., Drug Safety Research & Development, Groton, CT 06340, USA
| | - Philip K. Farahat
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, and the Institute for Quantitative and Computational Biosciences – The Collaboratory, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | - Paul G. Thomas
- Department of Host-Microbe Interactions, St Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - S. Armando Villalta
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Stefan A. Schattgen
- Department of Host-Microbe Interactions, St Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Melissa J. Spencer
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
4
|
Vrellaku B, Sethw Hassan I, Howitt R, Webster CP, Harriss E, McBlane F, Betts C, Schettini J, Lion M, Mindur JE, Duerr M, Shaw PJ, Kirby J, Azzouz M, Servais L. A systematic review of immunosuppressive protocols used in AAV gene therapy for monogenic disorders. Mol Ther 2024; 32:3220-3259. [PMID: 39044426 PMCID: PMC11489562 DOI: 10.1016/j.ymthe.2024.07.016] [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: 02/06/2024] [Revised: 05/24/2024] [Accepted: 07/18/2024] [Indexed: 07/25/2024] Open
Abstract
The emergence of adeno-associated virus (AAV)-based gene therapy has brought hope to patients with severe monogenic disorders. However, immune responses to AAV vectors and transgene products present challenges that require effective immunosuppressive strategies. This systematic review focuses on the immunosuppressive protocols used in 38 clinical trials and 35 real-world studies, considering a range of monogenic diseases, AAV serotypes, and administration routes. The review underscores the need for a deeper understanding of immunosuppressive regimens to enhance the safety and effectiveness of AAV-based gene therapy. Characterizing the immunological responses associated with various gene therapy treatments is crucial for optimizing treatment protocols and ensuring the safety and efficacy of forthcoming gene therapy interventions. Further research and understanding of the impact of immunosuppression on disease, therapy, and route of administration will contribute to the development of more effective and safer gene therapy approaches in the future.
Collapse
Affiliation(s)
- Besarte Vrellaku
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Ilda Sethw Hassan
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | | | - Christopher P Webster
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Eli Harriss
- Bodleian Health Care Libraries, University of Oxford, Oxford, UK
| | | | - Corinne Betts
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Jorge Schettini
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Mattia Lion
- Takeda Pharmaceuticals USA, Inc, Cambridge, MA, USA
| | | | - Michael Duerr
- Bayer Aktiengesellschaft, CGT&Rare Diseases, Leverkusen, Deutschland
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Janine Kirby
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Mimoun Azzouz
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, School of Medicine and Population Health, University of Sheffield, Sheffield, UK; Gene Therapy Innovation & Manufacturing Centre (GTIMC), University of Sheffield, Sheffield, UK.
| | - Laurent Servais
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; Division of Child Neurology, Department of Paediatrics, Centre de Référence des Maladies Neuromusculaires, University Hospital Liège and University of Liège, Liège, Belgium.
| |
Collapse
|
5
|
Mendonça RH, Ortega AB, Matsui C, van der Linden V, Kerstenetzky M, Grossklauss LF, Silveira-Lucas EL, Polido GJ, Zanoteli E. Gene replacement therapy for spinal muscular atrophy: safety and preliminary efficacy in a Brazilian cohort. Gene Ther 2024; 31:391-399. [PMID: 38839888 DOI: 10.1038/s41434-024-00456-y] [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: 01/30/2023] [Revised: 04/30/2024] [Accepted: 05/09/2024] [Indexed: 06/07/2024]
Abstract
Spinal muscular atrophy (SMA) is a motor neuron disease associated with progressive muscle weakness, ventilatory failure, and reduced survival. Onasemnogene abeparvovec is the first gene replacement therapy (GT) approved to treat this condition. An observational retrospective study was conducted to assess adverse events and efficacy of GT in SMA patients. Forty-one patients with SMA (58.5% females and 80.1% SMA type 1) were included. The mean age at GT dosing was 18 (±6.4) months. Thirty-six patients (87.8%) were under previous treatment with nusinersen, and 10 (24.4%) continued nusinersen after GT. Mean CHOP-INTEND increased 13 points after 6 months and this finding did not differ between groups according to nusinersen maintenance after GT (p = 0.949). Among SMA type 1 patients, 14 (46.6%) reached the ability to sit alone. Liver transaminases elevation at least two times higher than the upper limit of normal value occurred in 29 (70.7%) patients. Thrombocytopenia occurred in 13 (31.7%) patients, and one presented thrombotic microangiopathy. Older age (>2 years) was associated with more prolonged use of corticosteroids (p = 0.021). GT is effective in SMA patients, combined nusinersen after GT did not appear to add gain in motor function and older age is associated with prolonged corticosteroid use.
Collapse
Affiliation(s)
- Rodrigo Holanda Mendonça
- Department of Neurology, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, Brazil.
- Hospital Samaritano Higienópolis, São Paulo, Brazil.
| | | | - Ciro Matsui
- Department of Neurology, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, Brazil
- Hospital Samaritano Higienópolis, São Paulo, Brazil
| | | | | | | | | | - Graziela Jorge Polido
- Department of Neurology, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Edmar Zanoteli
- Department of Neurology, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, Brazil
| |
Collapse
|
6
|
Schwotzer N, El Sissy C, Desguerre I, Frémeaux-Bacchi V, Servais L, Fakhouri F. Thrombotic Microangiopathy as an Emerging Complication of Viral Vector-Based Gene Therapy. Kidney Int Rep 2024; 9:1995-2005. [PMID: 39081755 PMCID: PMC11284364 DOI: 10.1016/j.ekir.2024.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/08/2024] [Indexed: 08/02/2024] Open
Abstract
Gene therapy has brought tremendous hope for patients with severe life-threatening monogenic diseases. Although studies have shown the efficacy of gene therapy, serious adverse events have also emerged, including thrombotic microangiopathy (TMA) following viral vector-based gene therapy. In this review, we briefly summarize the concept of gene therapy, and the immune response triggered by viral vectors. We also discuss the incidence, presentation, and potential underlying mechanisms, including complement activation, of gene therapy-associated TMA. Further studies are needed to better define the pathogenesis of this severe complication of gene therapy, and the optimal measures to prevent it.
Collapse
Affiliation(s)
- Nora Schwotzer
- Service of Nephrology and Hypertension, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Carine El Sissy
- Department of Immunology, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
- Paris University, Paris, France
| | - Isabelle Desguerre
- Paediatric Neurology Department, Necker Hospital, APHP Centre, Université Paris Cité, Paris, France
| | - Véronique Frémeaux-Bacchi
- Department of Immunology, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
- Paris University, Paris, France
| | - Laurent Servais
- MDUK Oxford Neuromuscular Center and NIHR Oxford Biomedical Research Center, University of Oxford, Oxford, UK
- Neuromuscular Center, Department of Pediatrics, University of Liege and University Hospital of Liege, Belgium
| | - Fadi Fakhouri
- Service of Nephrology and Hypertension, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
7
|
Salabarria SM, Corti M, Coleman KE, Wichman MB, Berthy JA, D’Souza P, Tifft CJ, Herzog RW, Elder ME, Shoemaker LR, Leon-Astudillo C, Tavakkoli F, Kirn DH, Schwartz JD, Byrne BJ. Thrombotic microangiopathy following systemic AAV administration is dependent on anti-capsid antibodies. J Clin Invest 2024; 134:e173510. [PMID: 37988172 PMCID: PMC10760971 DOI: 10.1172/jci173510] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/25/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUNDSystemic administration of adeno-associated virus (AAV) can trigger life-threatening inflammatory responses, including thrombotic microangiopathy (TMA), acute kidney injury due to atypical hemolytic uremic syndrome-like complement activation, immune-mediated myocardial inflammation, and hepatic toxicity.METHODSWe describe the kinetics of immune activation following systemic AAV serotype 9 (AAV9) administration in 38 individuals following 2 distinct prophylactic immunomodulation regimens. Group 1 received corticosteroids and Group 2 received rituximab plus sirolimus in addition to steroids to prevent anti-AAV antibody formation.RESULTSGroup 1 participants had a rapid increase in immunoglobulin M (IgM) and IgG. Increase in D-dimer, decline in platelet count, and complement activation are indicative of TMA. All Group 1 participants demonstrated activation of both classical and alternative complement pathways, as indicated by depleted C4 and elevated soluble C5b-9, Ba, and Bb antigens. Group 2 patients did not have a significant change in IgM or IgG and had minimal complement activation.CONCLUSIONSThis study demonstrates that TMA in the setting of AAV gene therapy is antibody dependent (classical pathway) and amplified by the alternative complement pathway. Critical time points and interventions are identified to allow for management of immune-mediated events that impact the safety and efficacy of systemic gene therapy.
Collapse
Affiliation(s)
| | - Manuela Corti
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Kirsten E. Coleman
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Megan B. Wichman
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Julie A. Berthy
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Precilla D’Souza
- National Human Genome Research Institute, NIH, Bethesda, Maryland, USA
| | - Cynthia J. Tifft
- National Human Genome Research Institute, NIH, Bethesda, Maryland, USA
| | | | - Melissa E. Elder
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | | | | | | | - David H. Kirn
- 4D Molecular Therapeutics, Emeryville, California, USA
| | | | - Barry J. Byrne
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
8
|
Kohn DB, Chen YY, Spencer MJ. Successes and challenges in clinical gene therapy. Gene Ther 2023; 30:738-746. [PMID: 37935854 PMCID: PMC10678346 DOI: 10.1038/s41434-023-00390-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/20/2023] [Accepted: 02/07/2023] [Indexed: 11/09/2023]
Abstract
Despite the ups and downs in the field over three decades, the science of gene therapy has continued to advance and provide enduring treatments for increasing number of diseases. There are active clinical trials approaching a variety of inherited and acquired disorders of different organ systems. Approaches include ex vivo modification of hematologic stem cells (HSC), T lymphocytes and other immune cells, as well as in vivo delivery of genes or gene editing reagents to the relevant target cells by either local or systemic administration. In this article, we highlight success and ongoing challenges in three areas of high activity in gene therapy: inherited blood cell diseases by targeting hematopoietic stem cells, malignant disorders using immune effector cells genetically modified with chimeric antigen receptors, and ophthalmologic, neurologic, and coagulation disorders using in vivo administration of adeno-associated virus (AAV) vectors. In recent years, there have been true cures for many of these diseases, with sustained clinical benefit that exceed those from other medical approaches. Each of these treatments faces ongoing challenges, namely their high one-time costs and the complexity of manufacturing the therapeutic agents, which are biological viruses and cell products, at pharmacologic standards of quality and consistency. New models of reimbursement are needed to make these innovative treatments widely available to patients in need.
Collapse
Affiliation(s)
- Donald B Kohn
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Yvonne Y Chen
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Chemical and Biomolecular Engineering, Henry Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA, USA
- Parker Institute for Cancer Immunotherapy Center at UCLA, University of California, Los Angeles, Los Angeles, CA, USA
| | - Melissa J Spencer
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
9
|
Mendonça RH, Zanoteli E. Gene therapy in neuromuscular disorders. ARQUIVOS DE NEURO-PSIQUIATRIA 2022; 80:249-256. [PMID: 35976325 PMCID: PMC9491441 DOI: 10.1590/0004-282x-anp-2022-s135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/29/2022] [Indexed: 06/15/2023]
Abstract
Monogenic neuromuscular disorders are potentially treatable through gene therapy. Using viral vectors, a therapeutic transgene aims to restore normal levels of a protein not produced by the defective gene, or to silence a gene whose expression leads to toxic effects. Spinal Muscular Atrophy (SMA) is a good example of a monogenic disease that currently has an AAV9-based vector gene therapy as a therapeutic option. In this review, we intend to discuss the viral vectors and their mechanisms of action, in addition to reviewing the clinical trials that supported the approval of gene therapy (AVXS-101) for SMA as well as neuromuscular diseases that are potentially treatable with gene replacement therapy.
Collapse
Affiliation(s)
- Rodrigo Holanda Mendonça
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, São Paulo, SP, Brazil
| | - Edmar Zanoteli
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, São Paulo, SP, Brazil
| |
Collapse
|