101
|
Provenzano F, Nyberg S, Giunti D, Torazza C, Parodi B, Bonifacino T, Usai C, Kerlero de Rosbo N, Milanese M, Uccelli A, Shaw PJ, Ferraiuolo L, Bonanno G. Micro-RNAs Shuttled by Extracellular Vesicles Secreted from Mesenchymal Stem Cells Dampen Astrocyte Pathological Activation and Support Neuroprotection in In-Vitro Models of ALS. Cells 2022; 11:cells11233923. [PMID: 36497181 PMCID: PMC9741322 DOI: 10.3390/cells11233923] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease with no effective cure. Astrocytes display a toxic phenotype in ALS and contribute to motoneuron (MN) degeneration. Modulating astrocytes' neurotoxicity can reduce MN death. Our previous studies showed the beneficial effect of mesenchymal stem cell (MSC) administration in SOD1G93A ALS mice, but the mechanisms are still unclear. We postulated that the effects could be mediated by extracellular vesicles (EVs) secreted by MSCs. We investigated, by immunohistochemical, molecular, and in vitro functional analyses, the activity of MSC-derived EVs on the pathological phenotype and neurotoxicity of astrocytes isolated from the spinal cord of symptomatic SOD1G93A mice and human astrocytes (iAstrocytes) differentiated from inducible neural progenitor cells (iNPCs) of ALS patients. In vitro EV exposure rescued mouse and human ALS astrocytes' neurotoxicity towards MNs. EVs significantly dampened the pathological phenotype and neuroinflammation in SOD1G93A astrocytes. In iAstrocytes, exposure to EVs increased the antioxidant factor Nrf2 and reduced reactive oxygen species. We previously found nine miRNAs upregulated in MSC-derived EVs. Here, the transfection of SOD1G93A astrocytes with single miRNA mimics reduced astrocytes' activation and the expression of neuroinflammatory factors. Moreover, miR-466q and miR-467f mimics downregulate Mapk11, while miR-466m-5p and miR-466i-3p mimics promote the nuclear translocation of Nrf2. In iAstrocytes, transfection with miR-29b-3p mimic upregulated NQO1 antioxidant activity and reduced neurotoxicity towards MNs. MSC-derived EVs modulate astrocytes' reactive phenotype and neurotoxicity through anti-inflammatory and antioxidant-shuttled miRNAs, thus representing a therapeutic strategy in ALS.
Collapse
Affiliation(s)
- Francesca Provenzano
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
| | - Sophie Nyberg
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Debora Giunti
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Largo Paolo Daneo, 316132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Carola Torazza
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
| | - Benedetta Parodi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Largo Paolo Daneo, 316132 Genoa, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| | - Cesare Usai
- Institute of Biophysics, National Research Council (CNR), Via De Marini 6, 16149 Genoa, Italy
| | - Nicole Kerlero de Rosbo
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
- TomaLab, Institute of Nanotechnology, National Research Council (CNR), Piazzale Aldo Moro 5, 0018 Rome, Italy
| | - Marco Milanese
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
- Correspondence: (M.M.); (L.F.); Tel.: +39-01-0335-2046 (M.M.); +44-(0)114-222-2257 (L.F.)
| | - Antonio Uccelli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Largo Paolo Daneo, 316132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
- Correspondence: (M.M.); (L.F.); Tel.: +39-01-0335-2046 (M.M.); +44-(0)114-222-2257 (L.F.)
| | - Giambattista Bonanno
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| |
Collapse
|
102
|
Horton RH, Saade D, Markati T, Harriss E, Bönnemann CG, Muntoni F, Servais L. A systematic review of adeno-associated virus gene therapies in neurology: the need for consistent safety monitoring of a promising treatment. J Neurol Neurosurg Psychiatry 2022; 93:1276-1288. [PMID: 36190933 DOI: 10.1136/jnnp-2022-329431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
Abstract
Adeno-associated virus (AAV) gene therapies are generating much excitement in the rare disease field, particularly for previously untreatable neurological conditions. Efficacy has been claimed for several gene therapy products and the number of trials is rapidly increasing. However, reports of severe treatment-related adverse reactions are emerging, including death. There is still insufficient knowledge about their aetiology, prevention and treatment. We therefore undertook to systematically review publicly available data on AAV gene therapies in order to collate existing information on both safety and efficacy. Here, we review emerging efficacy reports of these novel therapies, many of which show promise. We also collate an increasing number of adverse reactions. Overwhelmingly, these results make a case for unified reporting of adverse events. This is likely to be critical for improving the safety of these promising treatments.
Collapse
Affiliation(s)
| | - Dimah Saade
- Division of Neurology, University of Iowa, Iowa, USA
| | | | - Elinor Harriss
- Bodleian Health Care Libraries, University of Oxford, Oxford, UK
| | - Carsten G Bönnemann
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Francesco Muntoni
- Dubowtiz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, UK, London, UK
| | - Laurent Servais
- Department of Paediatrics, University of Oxford, Oxford, UK .,Neuromuscular Reference Center, Department of Paediatrics, CHU of Liège, Liège, Belgium
| |
Collapse
|
103
|
Gallagher T. Meeting Summary: ESGCT 2022 AAV Safety Plenary Session. Hum Gene Ther 2022; 33:1217-1220. [DOI: 10.1089/hum.2022.29230.tga] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Thomas Gallagher
- Managing Editor, Human Gene Therapy, Mary Ann Liebert, Inc. New Rochelle, New York, USA
| |
Collapse
|
104
|
Sun K, Liao MZ. Clinical Pharmacology Considerations on Recombinant Adeno‐Associated Virus–Based Gene Therapy. J Clin Pharmacol 2022; 62 Suppl 2:S79-S94. [DOI: 10.1002/jcph.2141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/16/2022] [Indexed: 12/04/2022]
Affiliation(s)
- Kefeng Sun
- Takeda Development Center Americas Cambridge Massachusetts USA
| | - Michael Z. Liao
- Clinical Pharmacology, Genentech Inc. South San Francisco California USA
| |
Collapse
|
105
|
Shen W, Liu S, Ou L. rAAV immunogenicity, toxicity, and durability in 255 clinical trials: A meta-analysis. Front Immunol 2022; 13:1001263. [PMID: 36389770 PMCID: PMC9647052 DOI: 10.3389/fimmu.2022.1001263] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/18/2022] [Indexed: 11/23/2022] Open
Abstract
Recombinant Adeno-associated virus (rAAV) is one of the main delivery vectors for gene therapy. To assess immunogenicity, toxicity, and features of AAV gene therapy in clinical settings, a meta-analysis of 255 clinical trials was performed. A total of 7,289 patients are planned to be dosed. AAV2 was the most dominantly used serotype (29.8%, n=72), and 8.3% (n=20) of trials used engineered capsids. 38.7% (n=91) of trials employed neutralizing antibody assays for patient enrollment, while 15.3% (n=36) used ELISA-based total antibody assays. However, there was high variability in the eligibility criteria with cut-off tiers ranging from 1:1 to 1:1,600. To address potential immunogenicity, 46.3% (n=118) of trials applied immunosuppressants (prophylactic or reactive), while 32.7% (n=18) of CNS and 37.5% (n=24) of ocular-directed trials employed immunosuppressants, possibly due to the immune-privileged status of CNS and retina. There were a total of 11 patient deaths across 8 trials, and 18 out of 30 clinical holds were due to toxicity findings in clinical studies. 30.6% (n=78) of trials had treatment-emergent serious adverse events (TESAEs), with hepatotoxicity and thrombotic microangiopathy (systemic delivery) and neurotoxicity (CNS delivery) being the most prominent. Additionally, the durability of gene therapy may be impacted by two distinct decline mechanisms: 1) rapid decline presumably due to immune responses; or 2) gradual decline due to vector dilution. The durability varied significantly depending on disease indication, dose, serotypes, and patient individuals. Most CNS (90.0%) and muscle trials (73.3%) achieved durable transgene expression, while only 43.6% of ocular trials had sustained clinical outcomes. The rAAV production system can affect rAAV quality and thus immunogenicity and toxicity. Out of 186 trials that have disclosed production system information, 63.0% (n=126) of trials used the transient transfection of the HEK293/HEK293T system, while 18.0% (n=36) applied the baculovirus/Sf9 (rBac/Sf9) system. There were no significant differences in TESAEs and durability between AAV generated by rBac/Sf9 and HEK293/HEK293T systems. In summary, rAAV immunogenicity and toxicity poses significant challenges for clinical development of rAAV gene therapies, and it warrants collaborative efforts to standardize monitoring/measurement methods, design novel strategies to overcome immune responses, and openly share relevant information.
Collapse
Affiliation(s)
| | | | - Li Ou
- 3Genemagic Biosciences, Wallingford, PA, United States,4Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States,*Correspondence: Li Ou,
| |
Collapse
|
106
|
Yang YS, Kim JM, Xie J, Chaugule S, Lin C, Ma H, Hsiao E, Hong J, Chun H, Shore EM, Kaplan FS, Gao G, Shim JH. Suppression of heterotopic ossification in fibrodysplasia ossificans progressiva using AAV gene delivery. Nat Commun 2022; 13:6175. [PMID: 36258013 PMCID: PMC9579182 DOI: 10.1038/s41467-022-33956-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 10/04/2022] [Indexed: 12/24/2022] Open
Abstract
Heterotopic ossification is the most disabling feature of fibrodysplasia ossificans progressiva, an ultra-rare genetic disorder for which there is currently no prevention or treatment. Most patients with this disease harbor a heterozygous activating mutation (c.617 G > A;p.R206H) in ACVR1. Here, we identify recombinant AAV9 as the most effective serotype for transduction of the major cells-of-origin of heterotopic ossification. We use AAV9 delivery for gene replacement by expression of codon-optimized human ACVR1, ACVR1R206H allele-specific silencing by AAV-compatible artificial miRNA and a combination of gene replacement and silencing. In mouse skeletal cells harboring a conditional knock-in allele of human mutant ACVR1 and in patient-derived induced pluripotent stem cells, AAV gene therapy ablated aberrant Activin A signaling and chondrogenic and osteogenic differentiation. In Acvr1(R206H) knock-in mice treated locally in early adulthood or systemically at birth, trauma-induced endochondral bone formation was markedly reduced, while inflammation and fibroproliferative responses remained largely intact in the injured muscle. Remarkably, spontaneous heterotopic ossification also substantially decreased in in Acvr1(R206H) knock-in mice treated systemically at birth or in early adulthood. Collectively, we develop promising gene therapeutics that can prevent disabling heterotopic ossification in mice, supporting clinical translation to patients with fibrodysplasia ossificans progressiva.
Collapse
Affiliation(s)
- Yeon-Suk Yang
- Department of Medicine/Division of Rheumatology, UMass Chan Medical School, Worcester, MA, USA
| | - Jung-Min Kim
- Department of Medicine/Division of Rheumatology, UMass Chan Medical School, Worcester, MA, USA
| | - Jun Xie
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA, USA
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Viral Vector Core, UMass Chan Medical School, Worcester, MA, USA
| | - Sachin Chaugule
- Department of Medicine/Division of Rheumatology, UMass Chan Medical School, Worcester, MA, USA
| | - Chujiao Lin
- Department of Medicine/Division of Rheumatology, UMass Chan Medical School, Worcester, MA, USA
| | - Hong Ma
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA, USA
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Viral Vector Core, UMass Chan Medical School, Worcester, MA, USA
| | - Edward Hsiao
- Division of Endocrinology and Metabolism, Department of Medicine; the Institute for Human Genetics; the Program in Craniofacial Biology; and the Eli and Edyth Broad Institute of Regeneration Medicine, University of California-San Francisco, San Francisco, CA, USA
| | - Jaehyoung Hong
- Department of Mathematical Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Hyonho Chun
- Department of Mathematical Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Eileen M Shore
- Department of Orthopaedic Surgery, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- The Center for Research in FOP and Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Frederick S Kaplan
- Department of Orthopaedic Surgery, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- The Center for Research in FOP and Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Guangping Gao
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA, USA.
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA.
- Viral Vector Core, UMass Chan Medical School, Worcester, MA, USA.
- Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA, USA.
| | - Jae-Hyuck Shim
- Department of Medicine/Division of Rheumatology, UMass Chan Medical School, Worcester, MA, USA.
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA, USA.
- Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
107
|
Ding Y, Zhang Y, Liu X. Combinational treatments of RNA interference and extracellular vesicles in the spinocerebellar ataxia. Front Mol Neurosci 2022; 15:1043947. [PMID: 36311034 PMCID: PMC9606576 DOI: 10.3389/fnmol.2022.1043947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
Spinocerebellar ataxia (SCA) is an autosomal dominant neurodegenerative disease (ND) with a high mortality rate. Symptomatic treatment is the only clinically adopted treatment. However, it has poor effect and serious complications. Traditional diagnostic methods [such as magnetic resonance imaging (MRI)] have drawbacks. Presently, the superiority of RNA interference (RNAi) and extracellular vesicles (EVs) in improving SCA has attracted extensive attention. Both can serve as the potential biomarkers for the diagnosing and monitoring disease progression. Herein, we analyzed the basis and prospect of therapies for SCA. Meanwhile, we elaborated the development and application of miRNAs, siRNAs, shRNAs, and EVs in the diagnosis and treatment of SCA. We propose the combination of RNAi and EVs to avoid the adverse factors of their respective treatment and maximize the benefits of treatment through the technology of EVs loaded with RNA. Obviously, the combinational therapy of RNAi and EVs may more accurately diagnose and cure SCA.
Collapse
Affiliation(s)
- Yingying Ding
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, Zhejiang, China
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Yong Zhang
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, Zhejiang, China
| | - Xuehong Liu
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, Zhejiang, China
- *Correspondence: Xuehong Liu,
| |
Collapse
|
108
|
Gray-Edwards H, McElroy A, Sena-Esteves M, Arjomandnejad M, Keeler-Klunk AM. AAV Gene Therapy Redosing in the CNS. Hum Gene Ther 2022; 33:889-892. [PMID: 36074937 DOI: 10.1089/hum.2022.170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Heather Gray-Edwards
- University of Massachusetts Medical School , Department of Radiology, Horae Gene Therapy Center, Horae Gene Therapy Center, 368 Plantation Street, ASC6-2041, Worcester, Massachusetts, United States, 01605;
| | - Abigail McElroy
- UMass Chan Medical School, Worcester, Massachusetts, United States;
| | - Miguel Sena-Esteves
- University of Massachusetts Medical School, Neurology and Gene Therapy Center, 381 Plantation Street, Suite 250, Worcester, Massachusetts, United States, 01605;
| | - Motahareh Arjomandnejad
- University of Massachusetts Medical School, Horae Gene Therapy Center, 55 Lake Avenue, Worcester, Massachusetts, United States, 01605.,umass medical scool, Worcester, United States;
| | - Allison May Keeler-Klunk
- University of Massachusetts Medical School, Gene Therapy Center, 55 Lake Avenue North, Worcester, Massachusetts, United States, 01604;
| |
Collapse
|
109
|
Transplantation of human neural progenitor cells secreting GDNF into the spinal cord of patients with ALS: a phase 1/2a trial. Nat Med 2022; 28:1813-1822. [PMID: 36064599 PMCID: PMC9499868 DOI: 10.1038/s41591-022-01956-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/18/2022] [Indexed: 11/08/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) involves progressive motor neuron loss, leading to paralysis and death typically within 3–5 years of diagnosis. Dysfunctional astrocytes may contribute to disease and glial cell line-derived neurotrophic factor (GDNF) can be protective. Here we show that human neural progenitor cells transduced with GDNF (CNS10-NPC-GDNF) differentiated to astrocytes protected spinal motor neurons and were safe in animal models. CNS10-NPC-GDNF were transplanted unilaterally into the lumbar spinal cord of 18 ALS participants in a phase 1/2a study (NCT02943850). The primary endpoint of safety at 1 year was met, with no negative effect of the transplant on motor function in the treated leg compared with the untreated leg. Tissue analysis of 13 participants who died of disease progression showed graft survival and GDNF production. Benign neuromas near delivery sites were common incidental findings at post-mortem. This study shows that one administration of engineered neural progenitors can provide new support cells and GDNF delivery to the ALS patient spinal cord for up to 42 months post-transplantation. A phase 1/2a study shows that human neural progenitor cells modified to release the growth factor GDNF are safely transplanted into the spinal cord of patients with ALS, with cell survival and GDNF production for over 3 years.
Collapse
|
110
|
Li X, Wei X, Lin J, Ou L. A versatile toolkit for overcoming AAV immunity. Front Immunol 2022; 13:991832. [PMID: 36119036 PMCID: PMC9479010 DOI: 10.3389/fimmu.2022.991832] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/17/2022] [Indexed: 12/12/2022] Open
Abstract
Recombinant adeno-associated virus (AAV) is a promising delivery vehicle for in vivo gene therapy and has been widely used in >200 clinical trials globally. There are already several approved gene therapy products, e.g., Luxturna and Zolgensma, highlighting the remarkable potential of AAV delivery. In the past, AAV has been seen as a relatively non-immunogenic vector associated with low risk of toxicity. However, an increasing number of recent studies indicate that immune responses against AAV and transgene products could be the bottleneck of AAV gene therapy. In clinical studies, pre-existing antibodies against AAV capsids exclude many patients from receiving the treatment as there is high prevalence of antibodies among humans. Moreover, immune response could lead to loss of efficacy over time and severe toxicity, manifested as liver enzyme elevations, kidney injury, and thrombocytopenia, resulting in deaths of non-human primates and patients. Therefore, extensive efforts have been attempted to address these issues, including capsid engineering, plasmapheresis, IgG proteases, CpG depletion, empty capsid decoy, exosome encapsulation, capsid variant switch, induction of regulatory T cells, and immunosuppressants. This review will discuss these methods in detail and highlight important milestones along the way.
Collapse
Affiliation(s)
- Xuefeng Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital; State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Shenzhen Luohu People’s Hospital, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiaoli Wei
- Guangzhou Dezheng Biotechnology Co., Ltd., Guangzhou, China
| | - Jinduan Lin
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital; State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Li Ou
- Genemagic Biosciences, Philadelphia, PA, United States
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
- *Correspondence: Li Ou,
| |
Collapse
|
111
|
Johnson SA, Fang T, De Marchi F, Neel D, Van Weehaeghe D, Berry JD, Paganoni S. Pharmacotherapy for Amyotrophic Lateral Sclerosis: A Review of Approved and Upcoming Agents. Drugs 2022; 82:1367-1388. [PMID: 36121612 DOI: 10.1007/s40265-022-01769-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2022] [Indexed: 11/03/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disorder involving loss of upper and lower motor neurons, with most cases ending in death within 3-5 years of onset. Several molecular and cellular pathways have been identified to cause ALS; however, treatments to stop or reverse disease progression are yet to be found. Riluzole, a neuroprotective agent offering only a modest survival benefit, has long been the sole disease-modifying therapy for ALS. Edaravone, which demonstrated statistically significant slowing of ALS disease progression, is gaining approval in an increasing number of countries since its first approval in 2015. Sodium phenylbutyrate and taurursodiol (PB-TURSO) was conditionally approved in Canada in 2022, having shown significant slowing of disease progression and prolonged survival. Most clinical trials have focused on testing small molecules affecting common cellular pathways in ALS: targeting glutamatergic, apoptotic, inflammatory, and oxidative stress mechanisms among others. More recently, clinical trials utilizing stem cell transplantation and other biologics have emerged. This rich and ever-growing pipeline of investigational products, along with innovative clinical trial designs, collaborative trial networks, and an engaged ALS community', provide renewed hope to finding a cure for ALS. This article reviews existing ALS therapies and the current clinical drug development pipeline.
Collapse
Affiliation(s)
- Stephen A Johnson
- Neurological Clinical Research Institute (NCRI), Healey & AMG Center for ALS, Massachusetts General Hospital, 165 Cambridge St, Suite 600, Boston, MA, 02114, USA
| | - Ton Fang
- University of Massachusetts Medical School, Worcester, MA, USA
| | - Fabiola De Marchi
- Department of Neurology, ALS Centre, Maggiore della Carità Hospital, Novara, Italy
- Department of Translational Medicine, University of Piemonte Orientale, 28100, Novara, Italy
| | | | - Donatienne Van Weehaeghe
- Nuclear Medicine Subdivision, Department of Imaging and Pathology, University Hospital Leuven, Leuven, Belgium
| | - James D Berry
- Neurological Clinical Research Institute (NCRI), Healey & AMG Center for ALS, Massachusetts General Hospital, 165 Cambridge St, Suite 600, Boston, MA, 02114, USA
| | - Sabrina Paganoni
- Neurological Clinical Research Institute (NCRI), Healey & AMG Center for ALS, Massachusetts General Hospital, 165 Cambridge St, Suite 600, Boston, MA, 02114, USA.
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, MA, USA.
| |
Collapse
|
112
|
Yang TY, Braun M, Lembke W, McBlane F, Kamerud J, DeWall S, Tarcsa E, Fang X, Hofer L, Kavita U, Upreti VV, Gupta S, Loo L, Johnson AJ, Chandode RK, Stubenrauch KG, Vinzing M, Xia CQ, Jawa V. Immunogenicity assessment of AAV-based gene therapies: An IQ consortium industry white paper. Mol Ther Methods Clin Dev 2022; 26:471-494. [PMID: 36092368 PMCID: PMC9418752 DOI: 10.1016/j.omtm.2022.07.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Immunogenicity has imposed a challenge to efficacy and safety evaluation of adeno-associated virus (AAV) vector-based gene therapies. Mild to severe adverse events observed in clinical development have been implicated with host immune responses against AAV gene therapies, resulting in comprehensive evaluation of immunogenicity during nonclinical and clinical studies mandated by health authorities. Immunogenicity of AAV gene therapies is complex due to the number of risk factors associated with product components and pre-existing immunity in human subjects. Different clinical mitigation strategies have been employed to alleviate treatment-induced or -boosted immunogenicity in order to achieve desired efficacy, reduce toxicity, or treat more patients who are seropositive to AAV vectors. In this review, the immunogenicity risk assessment, manifestation of immunogenicity and its impact in nonclinical and clinical studies, and various clinical mitigation strategies are summarized. Last, we present bioanalytical strategies, methodologies, and assay validation applied to appropriately monitor immunogenicity in AAV gene therapy-treated subjects.
Collapse
|
113
|
Abstract
Gene transfer using adeno-associated viral (AAV) vectors has made tremendous progress in the last decade and has achieved cures of debilitating diseases such as hemophilia A and B. Nevertheless, progress is still being hampered by immune responses against the AAV capsid antigens or the transgene products. Immunosuppression designed to blunt T cell responses has shown success in some patients but failed in others especially if they received very high AAV vectors doses. Although it was initially thought that AAV vectors induce only marginal innate responses below the threshold of systemic symptoms recent trials have shown that complement activation can results in serious adverse events. Dorsal root ganglia toxicity has also been identified as a complication of high vector doses as has severe hepatotoxicity. Most of the critical complications occur in patients who are treated with very high vector doses indicating that the use of more efficient AAV vectors to allow for dose sparing or giving smaller doses repeatedly, the latter in conjunction with antibody or B cell depleting measures, should be explored.
Collapse
Affiliation(s)
- Hildegund C. J. Ertl
- Ertl Laboratory, Vaccine Center, The Wistar Institute, Philadelphia, PA, United States
| |
Collapse
|
114
|
Lewin AS, Smith WC. Gene Therapy for Rhodopsin Mutations. Cold Spring Harb Perspect Med 2022; 12:a041283. [PMID: 35940643 PMCID: PMC9435570 DOI: 10.1101/cshperspect.a041283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Mutations in RHO, the gene for rhodopsin, account for a large fraction of autosomal-dominant retinitis pigmentosa (adRP). Patients fall into two clinical classes, those with early onset, pan retinal photoreceptor degeneration, and those who experience slowly progressive disease. The latter class of patients are candidates for photoreceptor-directed gene therapy, while former may be candidates for delivery of light-responsive proteins to interneurons or retinal ganglion cells. Gene therapy for RHO adRP may be targeted to the mutant gene at the DNA or RNA level, while other therapies preserve the viability of photoreceptors without addressing the underlying mutation. Correcting the RHO gene and replacing the mutant RNA show promise in animal models, while sustaining viable photoreceptors has the potential to delay the loss of central vision and may preserve photoreceptors for gene-directed treatments.
Collapse
Affiliation(s)
- Alfred S Lewin
- Departments of Molecular Genetics and Microbiology and Ophthalmology, University of Florida College of Medicine, Gainesville, Florida 32610, USA
| | - W Clay Smith
- Departments of Molecular Genetics and Microbiology and Ophthalmology, University of Florida College of Medicine, Gainesville, Florida 32610, USA
| |
Collapse
|
115
|
Epigenetic genes and epilepsy - emerging mechanisms and clinical applications. Nat Rev Neurol 2022; 18:530-543. [PMID: 35859062 DOI: 10.1038/s41582-022-00693-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2022] [Indexed: 12/21/2022]
Abstract
An increasing number of epilepsies are being attributed to variants in genes with epigenetic functions. The products of these genes include factors that regulate the structure and function of chromatin and the placing, reading and removal of epigenetic marks, as well as other epigenetic processes. In this Review, we provide an overview of the various epigenetic processes, structuring our discussion around five function-based categories: DNA methylation, histone modifications, histone-DNA crosstalk, non-coding RNAs and chromatin remodelling. We provide background information on each category, describing the general mechanism by which each process leads to altered gene expression. We also highlight key clinical and mechanistic aspects, providing examples of genes that strongly associate with epilepsy within each class. We consider the practical applications of these findings, including tissue-based and biofluid-based diagnostics and precision medicine-based treatments. We conclude that variants in epigenetic genes are increasingly found to be causally involved in the epilepsies, with implications for disease mechanisms, treatments and diagnostics.
Collapse
|
116
|
Gene-based therapeutics for rare genetic neurodevelopmental psychiatric disorders. Mol Ther 2022; 30:2416-2428. [PMID: 35585789 PMCID: PMC9263284 DOI: 10.1016/j.ymthe.2022.05.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 11/23/2022] Open
Abstract
We are in an emerging era of gene-based therapeutics with significant promise for rare genetic disorders. The potential is particularly significant for genetic central nervous system disorders that have begun to achieve Food and Drug Administration approval for select patient populations. This review summarizes the discussions and presentations of the National Institute of Mental Health-sponsored workshop "Gene-Based Therapeutics for Rare Genetic Neurodevelopmental Psychiatric Disorders," which was held in January 2021. Here, we distill the points raised regarding various precision medicine approaches related to neurodevelopmental and psychiatric disorders that may be amenable to gene-based therapies.
Collapse
|
117
|
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating motor neuron disease for which there is currently no robust therapy. Recent progress in understanding ALS disease mechanisms and genetics in combination with innovations in gene modulation strategies creates promising new options for the development of ALS therapies. In recent years, six gene modulation therapies have been tested in ALS patients. These target gain-of-function pathology of the most common ALS genes, SOD1, C9ORF72, FUS, and ATXN2, using adeno-associated virus (AAV)-mediated microRNAs and antisense oligonucleotides (ASOs). Here, we review the latest clinical and preclinical advances in gene modulation approaches for ALS, including gene silencing, gene correction, and gene augmentation. These techniques have the potential to positively impact the direction of future research trials and transform ALS treatments for this grave disease.
Collapse
Affiliation(s)
- Katharina E Meijboom
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Robert H Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
118
|
Demarest TG, Biferi MG. Translation of gene therapy strategies for amyotrophic lateral sclerosis. Trends Mol Med 2022; 28:795-796. [DOI: 10.1016/j.molmed.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/29/2022] [Accepted: 07/06/2022] [Indexed: 10/16/2022]
|
119
|
Gene Therapy in Amyotrophic Lateral Sclerosis. Cells 2022; 11:cells11132066. [PMID: 35805149 PMCID: PMC9265980 DOI: 10.3390/cells11132066] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 12/30/2022] Open
Abstract
Since the discovery of Cu/Zn superoxide dismutase (SOD1) gene mutation, in 1993, as the first genetic abnormality in amyotrophic lateral sclerosis (ALS), over 50 genes have been identified as either cause or modifier in ALS and ALS/frontotemporal dementia (FTD) spectrum disease. Mutations in C9orf72, SOD1, TAR DNA binding protein 43 (TARDBP), and fused in sarcoma (FUS) genes are the four most common ones. During the last three decades, tremendous effort has been made worldwide to reveal biological pathways underlying the pathogenesis of these gene mutations in ALS/FTD. Accordingly, targeting etiologic genes (i.e., gene therapies) to suppress their toxic effects have been investigated widely. It includes four major strategies: (i) removal or inhibition of abnormal transcribed RNA using microRNA or antisense oligonucleotides (ASOs), (ii) degradation of abnormal mRNA using RNA interference (RNAi), (iii) decrease or inhibition of mutant proteins (e.g., using antibodies against misfolded proteins), and (iv) DNA genome editing with methods such as clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein (CRISPR/Cas). The promising results of these studies have led to the application of some of these strategies into ALS clinical trials, especially for C9orf72 and SOD1. In this paper, we will overview advances in gene therapy in ALS/FTD, focusing on C9orf72, SOD1, TARDBP, and FUS genes.
Collapse
|
120
|
Alarcan H, Al Ojaimi Y, Lanznaster D, Escoffre JM, Corcia P, Vourc'h P, Andres CR, Veyrat-Durebex C, Blasco H. Taking Advantages of Blood–Brain or Spinal Cord Barrier Alterations or Restoring Them to Optimize Therapy in ALS? J Pers Med 2022; 12:jpm12071071. [PMID: 35887567 PMCID: PMC9319288 DOI: 10.3390/jpm12071071] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder that still lacks an efficient therapy. The barriers between the central nervous system (CNS) and the blood represent a major limiting factor to the development of drugs for CNS diseases, including ALS. Alterations of the blood–brain barrier (BBB) or blood–spinal cord barrier (BSCB) have been reported in this disease but still require further investigations. Interestingly, these alterations might be involved in the complex etiology and pathogenesis of ALS. Moreover, they can have potential consequences on the diffusion of candidate drugs across the brain. The development of techniques to bypass these barriers is continuously evolving and might open the door for personalized medical approaches. Therefore, identifying robust and non-invasive markers of BBB and BSCB alterations can help distinguish different subgroups of patients, such as those in whom barrier disruption can negatively affect the delivery of drugs to their CNS targets. The restoration of CNS barriers using innovative therapies could consequently present the advantage of both alleviating the disease progression and optimizing the safety and efficiency of ALS-specific therapies.
Collapse
Affiliation(s)
- Hugo Alarcan
- Laboratoire de Biochimie et Biologie Moleculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Yara Al Ojaimi
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Debora Lanznaster
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Jean-Michel Escoffre
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Philippe Corcia
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
- Service de Neurologie, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
| | - Patrick Vourc'h
- Laboratoire de Biochimie et Biologie Moleculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Christian R Andres
- Laboratoire de Biochimie et Biologie Moleculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Charlotte Veyrat-Durebex
- Laboratoire de Biochimie et Biologie Moleculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| | - Hélène Blasco
- Laboratoire de Biochimie et Biologie Moleculaire, CHRU Bretonneau, 2 Boulevard Tonnellé, 37000 Tours, France
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37000 Tours, France
| |
Collapse
|
121
|
Paul A, Collins MG, Lee HY. Gene Therapy: The Next-Generation Therapeutics and Their Delivery Approaches for Neurological Disorders. Front Genome Ed 2022; 4:899209. [PMID: 35832929 PMCID: PMC9272754 DOI: 10.3389/fgeed.2022.899209] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/05/2022] [Indexed: 12/14/2022] Open
Abstract
Neurological conditions like neurodevelopmental disorders and neurodegenerative diseases are quite complex and often exceedingly difficult for patients. Most of these conditions are due to a mutation in a critical gene. There is no cure for the majority of these neurological conditions and the availability of disease-modifying therapeutics is quite rare. The lion's share of the treatments that are available only provide symptomatic relief, as such, we are in desperate need of an effective therapeutic strategy for these conditions. Considering the current drug development landscape, gene therapy is giving us hope as one such effective therapeutic strategy. Consistent efforts have been made to develop gene therapy strategies using viral and non-viral vectors of gene delivery. Here, we have discussed both of these delivery methods and their properties. We have summarized the relative advantages and drawbacks of viral and non-viral vectors from the perspectives of safety, efficiency, and productivity. Recent developments such as clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-mediated gene editing and its use in vivo have been described here as well. Given recent advancements, gene therapy shows great promise to emerge as a next-generation therapeutic for many of the neurodevelopmental and neurodegenerative conditions.
Collapse
|
122
|
Farrar MA, Groen E, Alves CR. Circulating neurofilaments to track dorsal root ganglion toxicity risks with AAV-mediated gene therapy. Mol Ther Methods Clin Dev 2022; 26:96-97. [PMID: 35795776 PMCID: PMC9234248 DOI: 10.1016/j.omtm.2022.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Identifying non-invasive biomarkers is critical to evaluate the long-term safety of adeno-associated virus (AAV)-mediated therapies. Plasma neurofilament light chain (Nf-L) levels are associated with dorsal root ganglia toxicity in rats and monkeys, suggesting that circulating Nf-L is a promising tool to be included in clinical trials and practice.
Collapse
Affiliation(s)
- Michelle A. Farrar
- Department of Neurology, Sydney Children’s Hospital Network, Level 8, Bright Alliance Building, Avoca Street, Randwick, NSW 2031, Australia,Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia,Corresponding author: Michelle A. Farrar, Level 8, Bright Alliance Building, Avoca Street, Department of Neurology, Sydney Children’s Hospital Network, Randwick, NSW 2031, Australia.
| | - Ewout Groen
- Department of Neurology, UMC Utrecht Brain Center, 3584 CX Utrecht, the Netherlands
| | - Christiano R.R. Alves
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Center for Genomic Medicine, Simches Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
123
|
Onasemnogene abeparvovec for presymptomatic infants with two copies of SMN2 at risk for spinal muscular atrophy type 1: the Phase III SPR1NT trial. Nat Med 2022; 28:1381-1389. [PMID: 35715566 PMCID: PMC9205281 DOI: 10.1038/s41591-022-01866-4] [Citation(s) in RCA: 166] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/10/2022] [Indexed: 12/19/2022]
Abstract
SPR1NT ( NCT03505099 ) was a Phase III, multicenter, single-arm study to investigate the efficacy and safety of onasemnogene abeparvovec for presymptomatic children with biallelic SMN1 mutations treated at ≤6 weeks of life. Here, we report final results for 14 children with two copies of SMN2, expected to develop spinal muscular atrophy (SMA) type 1. Efficacy was compared with a matched Pediatric Neuromuscular Clinical Research natural-history cohort (n = 23). All 14 enrolled infants sat independently for ≥30 seconds at any visit ≤18 months (Bayley-III item #26; P < 0.001; 11 within the normal developmental window). All survived without permanent ventilation at 14 months as per protocol; 13 maintained body weight (≥3rd WHO percentile) through 18 months. No child used nutritional or respiratory support. No serious adverse events were considered related to treatment by the investigator. Onasemnogene abeparvovec was effective and well-tolerated for children expected to develop SMA type 1, highlighting the urgency for universal newborn screening.
Collapse
|
124
|
Fader KA, Pardo ID, Kovi RC, Somps CJ, Wang HH, Vaidya VS, Ramaiah SK, Sirivelu MP. Circulating neurofilament light chain as a promising biomarker of AAV-induced dorsal root ganglia toxicity in nonclinical toxicology species. Mol Ther Methods Clin Dev 2022; 25:264-277. [PMID: 35505662 PMCID: PMC9024379 DOI: 10.1016/j.omtm.2022.03.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/27/2022] [Indexed: 12/14/2022]
Abstract
Adeno-associated virus (AAV)-induced dorsal root ganglia (DRG) toxicity has been observed in several nonclinical species, where lesions are characterized by neuronal degeneration/necrosis, nerve fiber degeneration, and mononuclear cell infiltration. As AAV vectors become an increasingly common platform for novel therapeutics, non-invasive biomarkers are needed to better characterize and manage the risk of DRG neurotoxicity in both nonclinical and clinical studies. Based on biological relevance, reagent availability, antibody cross-reactivity, DRG protein expression, and assay performance, neurofilament light chain (NF-L) emerged as a promising biomarker candidate. Dose- and time-dependent changes in NF-L were evaluated in male Wistar Han rats and cynomolgus monkeys following intravenous or intrathecal AAV injection, respectively. NF-L profiles were then compared against microscopic DRG lesions on day 29 post-dosing. In animals exhibiting DRG toxicity, plasma/serum NF-L was strongly associated with the severity of neuronal degeneration/necrosis and nerve fiber degeneration, with elevations beginning as early as day 8 in rats (≥5 × 1013 vg/kg) and day 14 in monkeys (≥3.3 × 1013 vg/dose). Consistent with the unique positioning of DRGs outside the blood-brain barrier, NF-L in cerebrospinal fluid was only weakly associated with DRG findings. In summary, circulating NF-L is a promising biomarker of AAV-induced DRG toxicity in nonclinical species.
Collapse
Affiliation(s)
- Kelly A Fader
- Pfizer Worldwide Research, Development and Medical, Drug Safety Research and Development, Groton, CT 06340, USA
| | | | - Ramesh C Kovi
- Pfizer Worldwide Research, Development and Medical, Drug Safety Research and Development, Pfizer Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Christopher J Somps
- Pfizer Worldwide Research, Development and Medical, Drug Safety Research and Development, Groton, CT 06340, USA
| | - Helen Hong Wang
- Pfizer Worldwide Research, Development and Medical, Drug Safety Research and Development, Pfizer Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Vishal S Vaidya
- Pfizer Worldwide Research, Development and Medical, Drug Safety Research and Development, Pfizer Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Shashi K Ramaiah
- Pfizer Worldwide Research, Development and Medical, Drug Safety Research and Development, Pfizer Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Madhu P Sirivelu
- Pfizer Worldwide Research, Development and Medical, Drug Safety Research and Development, Pfizer Inc., 300 Technology Square, Cambridge, MA 02139, USA
| |
Collapse
|
125
|
Hur SK, Hunter M, Dominique MA, Farag M, Cotton-Samuel D, Khan T, Trojanowski JQ, Spiller KJ, Lee VMY. Slow motor neurons resist pathological TDP-43 and mediate motor recovery in the rNLS8 model of amyotrophic lateral sclerosis. Acta Neuropathol Commun 2022; 10:75. [PMID: 35568882 PMCID: PMC9107273 DOI: 10.1186/s40478-022-01373-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 01/22/2023] Open
Abstract
In the intermediate stages of amyotrophic lateral sclerosis (ALS), surviving motor neurons (MNs) that show intrinsic resistance to TDP-43 proteinopathy can partially compensate for the loss of their more disease-susceptible counterparts. Elucidating the mechanisms of this compensation may reveal approaches for attenuating motor impairment in ALS patients. In the rNLS8 mouse model of ALS-like pathology driven by doxycycline-regulated neuronal expression of human TDP-43 lacking a nuclear localization signal (hTDP-43ΔNLS), slow MNs are more resistant to disease than fast-fatigable (FF) MNs and can mediate recovery following transgene suppression. In the present study, we used a viral tracing strategy to show that these disease-resistant slow MNs sprout to reinnervate motor endplates of adjacent muscle fibers vacated by degenerated FF MNs. Moreover, we found that neuromuscular junctions within fast-twitch skeletal muscle (tibialis anterior, TA) reinnervated by SK3-positive slow MNs acquire resistance to axonal dieback when challenged with a second course of hTDP-43ΔNLS pathology. The selective resistance of reinnervated neuromuscular junctions was specifically induced by the unique pattern of reinnervation following TDP-43-induced neurodegeneration, as recovery from unilateral sciatic nerve crush did not produce motor units resistant to subsequent hTDP-43ΔNLS. Using cross-reinnervation and self-reinnervation surgery in which motor axons are disconnected from their target muscle and reconnected to a new muscle, we show that FF MNs remain hTDP-43ΔNLS-susceptible and slow MNs remain resistant, regardless of which muscle fibers they control. Collectively, these findings demonstrate that MN identity dictates the susceptibility of neuromuscular junctions to TDP-43 pathology and slow MNs can drive recovery of motor systems due to their remarkable resilience to TDP-43-driven degeneration. This study highlights a potential pathway for regaining motor function with ALS pathology in the advent of therapies that halt the underlying neurodegenerative process.
Collapse
Affiliation(s)
- Seong Kwon Hur
- grid.25879.310000 0004 1936 8972Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Maloney Building, 3rd Floor, 3600 Spruce Street, Philadelphia, PA 19104-2676 USA ,grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
| | - Mandana Hunter
- grid.25879.310000 0004 1936 8972Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Maloney Building, 3rd Floor, 3600 Spruce Street, Philadelphia, PA 19104-2676 USA ,grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
| | - Myrna A. Dominique
- grid.25879.310000 0004 1936 8972Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Maloney Building, 3rd Floor, 3600 Spruce Street, Philadelphia, PA 19104-2676 USA ,grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
| | - Madona Farag
- grid.25879.310000 0004 1936 8972Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Maloney Building, 3rd Floor, 3600 Spruce Street, Philadelphia, PA 19104-2676 USA ,grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
| | - Dejania Cotton-Samuel
- grid.25879.310000 0004 1936 8972Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Maloney Building, 3rd Floor, 3600 Spruce Street, Philadelphia, PA 19104-2676 USA ,grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
| | - Tahiyana Khan
- grid.25879.310000 0004 1936 8972Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Maloney Building, 3rd Floor, 3600 Spruce Street, Philadelphia, PA 19104-2676 USA ,grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
| | - John Q. Trojanowski
- grid.25879.310000 0004 1936 8972Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Maloney Building, 3rd Floor, 3600 Spruce Street, Philadelphia, PA 19104-2676 USA ,grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA ,grid.25879.310000 0004 1936 8972Alzheimer’s Disease Research Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
| | - Krista J. Spiller
- grid.497530.c0000 0004 0389 4927Janssen Research and Development, Neuroscience Therapeutic Area, 1400 McKean Rd, Spring House, PA 19002 USA
| | - Virginia M.-Y. Lee
- grid.25879.310000 0004 1936 8972Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Maloney Building, 3rd Floor, 3600 Spruce Street, Philadelphia, PA 19104-2676 USA ,grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA ,grid.25879.310000 0004 1936 8972Alzheimer’s Disease Research Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
| |
Collapse
|
126
|
Kaczmarek R. Gene therapy - are we ready now? Haemophilia 2022; 28 Suppl 4:35-43. [PMID: 35521736 PMCID: PMC9325484 DOI: 10.1111/hae.14530] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 01/19/2023]
Abstract
Introduction Haemophilia therapy has evolved from rudimentary transfusion‐based approaches to an unprecedented level of innovation with glimmers of functional cure brought by gene therapy. After decades of misfires, gene therapy has normalized factor (F)VIII and factor (F)IX levels in some individuals in the long term. Several clinical programmes testing adeno‐associated viral (AAV) vector gene therapy are approaching completion with imminent regulatory approvals. Discussion Phase 3 studies along with multiyear follow‐up in earlier phase investigations raised questions about efficacy as well as short‐ and long‐term safety, prompting a reappraisal of AAV vector gene therapy. Liver toxicities, albeit mostly low‐grade, occur in the first year in at least some individuals in all haemophilia A and B trials and are poorly understood. Extreme variability and unpredictability of outcome, as well as a slow decline in factor expression (seemingly unique to FVIII gene therapy), are vexing because immune responses to AAV vectors preclude repeat dosing, which could increase suboptimal or restore declining expression, while overexpression may result in phenotoxicity. The long‐term safety will need lifelong monitoring because AAV vectors, contrary to conventional wisdom, integrate into chromosomes at the rate that calls for vigilance. Conclusions AAV transduction and transgene expression engage the host immune system, cellular DNA processing, transcription and translation machineries in ways that have been only cursorily studied in the clinic. Delineating those mechanisms will be key to finding mitigants and solutions to the remaining problems, and including individuals who cannot avail of gene therapy at this time.
Collapse
Affiliation(s)
- Radoslaw Kaczmarek
- Coagulation Products Safety Supply and Access Committee, World Federation of Hemophilia, Montreal, Quebec, Canada.,Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
127
|
Pipe SW, Reddy KR, Chowdary P. Gene therapy: Practical aspects of implementation. Haemophilia 2022; 28 Suppl 4:44-52. [PMID: 35521727 PMCID: PMC9324089 DOI: 10.1111/hae.14545] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 12/17/2022]
Abstract
The first wave of gene therapies for haemophilia submitted for regulatory review utilize a liver-directed approach in which a functional gene copy of factor VIII (FVIII) or factor IX (FIX) is packaged inside a recombinant adeno-associated viral vector (rAAV). Following a single treatment event, these particles are taken up into liver cells, where the rAAV uncoats and delivers the DNA to the nucleus of the cell, where genetic elements that accompany the gene allow for efficient expression and secretion of FVIII or FIX protein into the plasma. An immune response to the vector capsid has been manifest by elevations in common liver enzymes that must be diligently followed postinfusion for weeks and months afterward and if signs of toxicity appear, will trigger a course of immunosuppression. Despite this, the studies have shown that this works in the great majority of individuals and the immunosuppression course is either avoided or short-lived for many. Optimal outcomes in the haemophilia population will be dependent on proper screening assessment and maintenance of liver health prior to consideration of gene therapy, close short-term follow up and implementation of immunomodulatory strategies to identify and manage liver toxicity and preserve durable transgene expression. This review proposes best practices to assist clinical teams with overcoming the challenges this platform of therapy poses to the traditional clinical care models and infrastructure within the haemophilia treatment centres (HTCs) who will be coordinating the patient's journey through this potentially transformative therapy.
Collapse
Affiliation(s)
- Steven W Pipe
- Departments of Paediatrics and Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - K Rajender Reddy
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Pratima Chowdary
- Katharine Dormandy Haemophilia and Thrombosis Centre, Royal Free Hospital, London, UK
| |
Collapse
|
128
|
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
|
129
|
Advanced Gene-Targeting Therapies for Motor Neuron Diseases and Muscular Dystrophies. Int J Mol Sci 2022; 23:ijms23094824. [PMID: 35563214 PMCID: PMC9101723 DOI: 10.3390/ijms23094824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 12/19/2022] Open
Abstract
Gene therapy is a revolutionary, cutting-edge approach to permanently ameliorate or amend many neuromuscular diseases by targeting their genetic origins. Motor neuron diseases and muscular dystrophies, whose genetic causes are well known, are the frontiers of this research revolution. Several genetic treatments, with diverse mechanisms of action and delivery methods, have been approved during the past decade and have demonstrated remarkable results. However, despite the high number of genetic treatments studied preclinically, those that have been advanced to clinical trials are significantly fewer. The most clinically advanced treatments include adeno-associated virus gene replacement therapy, antisense oligonucleotides, and RNA interference. This review provides a comprehensive overview of the advanced gene therapies for motor neuron diseases (i.e., amyotrophic lateral sclerosis and spinal muscular atrophy) and muscular dystrophies (i.e., Duchenne muscular dystrophy, limb-girdle muscular dystrophy, and myotonic dystrophy) tested in clinical trials. Emphasis has been placed on those methods that are a few steps away from their authoritative approval.
Collapse
|
130
|
McMillan HJ, Proud CM, Farrar MA, Alexander IE, Muntoni F, Servais L. Onasemnogene abeparvovec for the treatment of spinal muscular atrophy. Expert Opin Biol Ther 2022; 22:1075-1090. [PMID: 35437095 DOI: 10.1080/14712598.2022.2066471] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Gene therapy for spinal muscular atrophy (SMA) represents a significant milestone in the treatment of neurologic diseases. SMA is a neurodegenerative disease that results in motor neuron loss because of mutations of the survival motor neuron 1 gene, which directs survival motor neuron (SMN) protein production. Onasemnogene abeparvovec, a one-time gene replacement therapy, delivers a functional transgene to restore SMN protein expression. Onasemnogene abeparvovec has demonstrated improved survival and motor milestone achievements for presymptomatic infants and patients with SMA type 1. AREAS COVERED This expert review describes the current state of gene therapy for SMA, reviews the mechanism of and clinical experience with onasemnogene abeparvovec, explains future efforts to expand applications of gene therapy for SMA, and provides context for developing gene therapy for other conditions. EXPERT OPINION Onasemnogene abeparvovec has demonstrated efficacy in clinical trials and, because of this, is a valuable treatment option for patients with symptomatic infantile SMA and those identified by newborn screening. Gene therapy is still in its infancy, and challenges and uncertainties associated with transgene delivery must be addressed. With ongoing development of vector technology, more specific tissue tropism, reduced "off-target" effects, and an enhanced safety profile will continue to evolve.
Collapse
Affiliation(s)
- Hugh J McMillan
- Departments of Pediatrics, Neurology & Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, Montreal, Canada
| | - Crystal M Proud
- Children's Hospital of The King's Daughters, Norfolk, VA, United States
| | - Michelle A Farrar
- School of Women's and Children's Health, UNSW Medicine, UNSW Sydney.,Sydney Children's Hospital Network, Sydney, Australia
| | - Ian E Alexander
- Gene Therapy Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney and Sydney Children's Hospitals Network, Westmead, NSW, Australia.,Discipline of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, London, UK
| | - Laurent Servais
- Department of Pediatrics, Centre Hospitalier Universitaire de Liège & Université de Liège, Liège, Belgium.,MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| |
Collapse
|
131
|
Mirian A, Moszczynski A, Soleimani S, Aubert I, Zinman L, Abrahao A. Breached Barriers: A Scoping Review of Blood-Central Nervous System Barrier Pathology in Amyotrophic Lateral Sclerosis. Front Cell Neurosci 2022; 16:851563. [PMID: 35431812 PMCID: PMC9009245 DOI: 10.3389/fncel.2022.851563] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Recent studies have implicated changes in the blood-central nervous system barriers (BCNSB) in amyotrophic lateral sclerosis (ALS). The objective of this scoping review is to synthesize the current evidence for BCNSB structure and functional abnormalities in ALS studies and propose how BCNSB pathology may impact therapeutic development. Methods A literature search was conducted using Ovid Medline, EMBASE, and Web of Science, from inception to November 2021 and limited to entries in English language. Simplified search strategy included the terms ALS/motor neuron disease and [BCNSB or blood-brain barrier (BBB) or blood-spinal cord barrier (BSCB)]. Henceforth, BCNSB is used as a term that is inclusive of the BBB and BSCB. Four independent reviewers conducted a title and abstract screening, hand-searched the reference lists of review papers, and performed a full text review of eligible studies. Included studies were original peer-reviewed full text publications, evaluating the structure and function of the BCNSB in preclinical models of ALS, clinical ALS, or postmortem human ALS tissue. There was no restriction on study design. The four reviewers independently extracted the data. Results The search retrieved 2,221 non-duplicated articles and 48 original studies were included in the synthesis. There was evidence that the integrity of the BCNSB is disrupted throughout the course of the disease in rodent models, beginning prior to symptom onset and detectable neurodegeneration. Increased permeability, pharmacoresistance with upregulated efflux transporters, and morphological changes in the supporting cells of the BCNSB, including pericytes, astrocytes, and endothelial cells were observed in animal models. BCNSB abnormalities were also demonstrated in postmortem studies of ALS patients. Therapeutic interventions targeting BCNSB dysfunction were associated with improved motor neuron survival in animal models of ALS. Conclusion BCNSB structural and functional abnormalities are likely implicated in ALS pathophysiology and may occur upstream to neurodegeneration. Promising therapeutic strategies targeting BCNSB dysfunction have been tested in animals and can be translated into ALS clinical trials.
Collapse
Affiliation(s)
- Ario Mirian
- Clinical Neurological Sciences, Western University, London Health Sciences, London, ON, Canada
| | | | - Serena Soleimani
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States
| | - Isabelle Aubert
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Lorne Zinman
- Division of Neurology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Neurology, Department of Medicine, Sunnybrook Health Science Centre, Toronto, ON, Canada
- Evaluative Clinical Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Agessandro Abrahao
- Division of Neurology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Neurology, Department of Medicine, Sunnybrook Health Science Centre, Toronto, ON, Canada
- Evaluative Clinical Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
132
|
Tukov FF, Mansfield K, Milton M, Meseck E, Penraat K, Chand D, Hartmann A. Single-Dose Intrathecal Dorsal Root Ganglia Toxicity of Onasemnogene Abeparvovec in Cynomolgus Monkeys. Hum Gene Ther 2022; 33:740-756. [PMID: 35331006 PMCID: PMC9347375 DOI: 10.1089/hum.2021.255] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Intravenous onasemnogene abeparvovec is approved for the treatment of spinal muscular atrophy in children < 2 years. For later-onset patients, intrathecal onasemnogene abeparvovec may be advantageous over intravenous administration. Recently, microscopic dorsal root ganglion (DRG) changes were observed in nonhuman primates (NHPs) following intrathecal onasemnogene abeparvovec administration. To characterize these DRG findings, two NHP studies evaluating intrathecal onasemnogene abeparvovec administration were conducted: a 12-month study with a 6-week interim cohort and a 13-week study with a 2-week interim cohort. The latter investigated the potential impact of prednisolone or rituximab plus everolimus on DRG toxicity. An additional 6-month, single-dose, intravenous NHP study conducted in parallel evaluated onasemnogene abeparvovec safety (including DRG toxicity) with or without prednisolone coadministration. Intrathecal onasemnogene abeparvovec administration was well tolerated and not associated with clinical observations. Microscopic onasemnogene abeparvovec-related changes were observed in the DRG and trigeminal ganglion (TG) and included mononuclear cell inflammation and/or neuronal degeneration, which was colocalized with high vector transcript expression at 6 weeks postdose. Incidence and severity of DRG changes were generally decreased after 52 weeks compared with 6 weeks postdose. Other onasemnogene abeparvovec-related microscopic findings of axonal degeneration, mononuclear cell infiltrates and/or gliosis in the spinal cord, dorsal spinal nerve root/spinal nerves, and/or peripheral nerves were absent or found at decreased incidences and/or severities after 52 weeks. DRG and/or TG microscopic findings following intravenous onasemnogene abeparvovec dosing included minimal to slight neuronal degeneration and mononuclear cell inflammation at 6 weeks and 6 months postdose. Nervous system microscopic findings following intrathecal onasemnogene abeparvovec (≥1.2 × 1013 vg/animal) trended toward resolution after 52 weeks, supporting nonprogression of changes, including in the DRG. Onasemnogene abeparvovec-related DRG findings were not associated with electrophysiology changes and were not ameliorated by prednisolone or rituximab plus everolimus coadministration. The pathogenesis is possibly a consequence of increased vector genome transduction and/or transgene expression.
Collapse
Affiliation(s)
| | - Keith Mansfield
- Novartis Institutes for BioMedical Research Inc, 33413, Cambridge, Massachusetts, United States;
| | - Mark Milton
- Novartis Institutes for BioMedical Research Inc, 33413, Cambridge, Massachusetts, United States;
| | - Emily Meseck
- Novartis Pharmaceuticals Corp, 33412, East Hanover, New Jersey, United States;
| | - Kelley Penraat
- Novartis Institutes for BioMedical Research Inc, 33413, Cambridge, Massachusetts, United States;
| | - Deepa Chand
- Novartis Gene Therapies, Inc., Bannockburn, United States.,Washington University School of Medicine in Saint Louis, 12275, St Louis, Missouri, United States;
| | | |
Collapse
|
133
|
Lin TJ, Cheng KC, Wu LY, Lai WY, Ling TY, Kuo YC, Huang YH. Potential of Cellular Therapy for ALS: Current Strategies and Future Prospects. Front Cell Dev Biol 2022; 10:851613. [PMID: 35372346 PMCID: PMC8966507 DOI: 10.3389/fcell.2022.851613] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/15/2022] [Indexed: 12/15/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by progressive upper and lower motor neuron (MN) degeneration with unclear pathology. The worldwide prevalence of ALS is approximately 4.42 per 100,000 populations, and death occurs within 3-5 years after diagnosis. However, no effective therapeutic modality for ALS is currently available. In recent years, cellular therapy has shown considerable therapeutic potential because it exerts immunomodulatory effects and protects the MN circuit. However, the safety and efficacy of cellular therapy in ALS are still under debate. In this review, we summarize the current progress in cellular therapy for ALS. The underlying mechanism, current clinical trials, and the pros and cons of cellular therapy using different types of cell are discussed. In addition, clinical studies of mesenchymal stem cells (MSCs) in ALS are highlighted. The summarized findings of this review can facilitate the future clinical application of precision medicine using cellular therapy in ALS.
Collapse
Affiliation(s)
- Ting-Jung Lin
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuang-Chao Cheng
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Luo-Yun Wu
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Yu Lai
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Yung-Che Kuo
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yen-Hua Huang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Comprehensive Cancer Center of Taipei Medical University, Taipei, Taiwan
- PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
134
|
Buss N, Lanigan L, Zeller J, Cissell D, Metea M, Adams E, Higgins M, Kim KH, Budzynski E, Yang L, Liu Y, Butt M, Danos O, Fiscella M. Characterization of AAV-mediated dorsal root ganglionopathy. Mol Ther Methods Clin Dev 2022; 24:342-354. [PMID: 35229008 PMCID: PMC8851102 DOI: 10.1016/j.omtm.2022.01.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/27/2022] [Indexed: 12/12/2022]
Abstract
Recent studies in non-human primates administered recombinant adeno-associated viruses (rAAVs) have shown lesions in the dorsal root ganglia (DRG) of unknown pathogenesis. In this study, rAAV9s manufactured using different purification methods alongside a non-expressing Null AAV9 vector was administered to groups of cynomolgus monkeys followed by neuropathological evaluation after 4 weeks. Lesions, including neuronal degeneration, increased cellularity, and nerve fiber degeneration, were observed in the DRG, regardless of purification methods. Animals did not develop any neurological signs throughout the study, and there was no loss of function observed in neuro-electrophysiological endpoints or clear effects on intraepidermal nerve fiber density. However, magnetic resonance imaging (MRI) of animals with axonopathy showed an increase in short tau inversion recovery (STIR) intensity and decrease in fractional anisotropy. In animals administered the Null AAV9 vector, DRG lesions were not observed despite vector DNA being detected in the DRG at levels equivalent to or greater than rAAV9-treated animals. This study further supports that DRG toxicity is associated with transgene overexpression in DRGs, with particular sensitivity at the lumbar and lumbosacral level. The data from this study also showed that the nerve fiber degeneration did not correlate with any functional effect on nerve conduction but was detectable by MRI.
Collapse
Affiliation(s)
| | | | | | | | - Monica Metea
- Preclinical Electrophysiology Consulting, Mattapoisett, MA 02739, USA
| | | | | | | | | | - Lin Yang
- REGENXBIO, Rockville, MD 20850, USA
| | - Ye Liu
- REGENXBIO, Rockville, MD 20850, USA
| | | | | | | |
Collapse
|
135
|
Chavda V, Patel C, Modh D, Ertas YN, Sonak SS, Munshi NK, Anand K, Soni A, Pandey S. Therapeutic Approaches to Amyotrophic Lateral Sclerosis from the Lab to the Clinic. Curr Drug Metab 2022; 23:200-222. [PMID: 35272595 DOI: 10.2174/1389200223666220310113110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 01/07/2022] [Accepted: 02/02/2022] [Indexed: 11/22/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a terminal neuro-degenerative disorder that is clinically recognized as a gradual degeneration of the upper and lower motor neurons, with an average duration of 3 to 5 years from initiation of symptoms to death. The mechanisms underlying the pathogenesis and progression of the disease are multifactorial. Therefore, to find effective treatments, it is necessary to understand this heterogeneity underlying the progression of ALS. Recent developments in gene therapy have opened a new avenue to treat this condition, especially for the characterized genetic types. Gene therapy methods have been studied in a variety of pre-clinical settings and clinical trials, and they may be a promising path for developing an effective and safe ALS cure. A growing body of evidence demonstrates abnormalities in energy metabolism at the cellular and whole-body level in animal models and in people living with ALS. The use and incorporation of high-throughput "omics" methods has radically transformed our thought about ALS, strengthening our understanding of the disease's dynamic molecular architecture, differentiating distinct patient subtypes, and creating a reasonable basis for the identification of biomarkers and novel individualised treatments. Future clinical and laboratory trials would also focus on the diverse relationships between metabolism and ALS to address the issue of whether targeting deficient metabolism in ALS is an effective way to change disease progression. In this review, we focus on the detailed pathogenesis of ALS and highlight principal genes, i.e., SOD1, TDP-43, C9orf72, and FUS, targeted therapeutic approaches of ALS. An attempt is made to provide up-to-date information on clinical outcomes, including various biomarkers which are thought to be important players in early ALS detection.
Collapse
Affiliation(s)
- Vivek Chavda
- Department of Pharmaceutic, L M College of Pharmacy, Ahmedabad - 380009 (India)
| | - Chirag Patel
- Department of Pharmacology, L M College of Pharmacy, Ahmedabad - 380009 (India)
| | - Dharti Modh
- Department of pharmaceutical chemistry, Poona college of pharmacy, Bharti vidhyapith, Pune - 411030 (India)
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering at Erciyes University, Kayseri, Turkey
- ERNAM - Nanotechnology Research and Application Center, Erciyes University, Kayseri 38039, Turkey
| | - Shreya S Sonak
- Department of pharmaceutical chemistry, Poona college of pharmacy, Bharti vidhyapith, Pune - 411030 (India)
| | - Nafisa K Munshi
- Department of pharmaceutical chemistry, Poona college of pharmacy, Bharti vidhyapith, Pune - 411030 (India)
| | - Krishna Anand
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences and National Health Laboratory Service, University of the Free State, Bloemfontein 9300, South Africa
| | - Arun Soni
- Department of Pharmacology, SSR College of Pharmacy, Silvassa, Dadra and Nagar Haveli - 396230(India)
| | - Sonal Pandey
- Research and Development, Meril Diagnostic Pvt. Ltd, Vapi - 396191 (India)
| |
Collapse
|
136
|
Masrori P, Ospitalieri S, Forsberg K, Moens TG, Poesen K, Race V, Brännström T, Andersen PM, Thal DR, Van Damme P. Respiratory onset of amyotrophic lateral sclerosis in a pregnant woman with a novel SOD1 mutation. Eur J Neurol 2022; 29:1279-1283. [PMID: 35253968 PMCID: PMC9311846 DOI: 10.1111/ene.15224] [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: 09/28/2021] [Revised: 11/29/2021] [Accepted: 12/14/2021] [Indexed: 12/01/2022]
Abstract
Background and purpose With the advent of gene therapies for amyotrophic lateral sclerosis (ALS), the importance of gene testing in ALS is increasing. This will likely lead to the identification of new variants for which the pathogenicity is not established. We aimed to study the pathogenicity of a newly identified variant in superoxide dismutase 1 (SOD1). Methods Gene testing was performed using Sanger sequencing. SOD1 activity in erythrocytes was measured using spectrophotometry. Postmortem brain and spinal cord sections were stained with antibodies against phospho‐TDP‐43 and SOD1. Results We identified a novel c.416G>T (p.Gly139Val) mutation in SOD1, which caused a rapidly progressive respiratory onset form of ALS. The mutation resulted in a 50% drop of SOD1 activity. Postmortem examination confirmed the absence of TDP‐43 pathology and displayed typical SOD1 inclusions in remaining motor neurons, confirming the pathogenic nature of the mutation. Conclusions Novel variants of unknown pathogenicity will be identified as a result of a surge in gene testing in people with ALS. An in‐depth study of a newly identified p.Gly139Val mutation in SOD1 confirmed the pathogenicity of this mutation. Future patients with this particular mutation should qualify for SOD1 silencing or editing therapies.
Collapse
Affiliation(s)
- Pegah Masrori
- Department of Neurology, Neuromuscular Reference Center, University Hospitals Leuven, Leuven, Belgium.,Department of Neurosciences, Experimental Neurology, Leuven Brain Institute, KU Leuven-University of Leuven, Leuven, Belgium.,Laboratory of Neurobiology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| | - Simona Ospitalieri
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Karin Forsberg
- Department of Clinical Sciences, Neurosciences, Umeå University, Umeå, Sweden.,Pathology Unit, Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | - Thomas G Moens
- Department of Neurosciences, Experimental Neurology, Leuven Brain Institute, KU Leuven-University of Leuven, Leuven, Belgium.,Laboratory of Neurobiology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| | - Koen Poesen
- Laboratory for Molecular Neurobiomarker Research, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium.,Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Valerie Race
- Laboratory for Molecular Diagnosis, University Hospitals Leuven, Leuven, Belgium
| | - Thomas Brännström
- Department of Clinical Sciences, Neurosciences, Umeå University, Umeå, Sweden
| | - Peter M Andersen
- Department of Clinical Sciences, Neurosciences, Umeå University, Umeå, Sweden
| | - Dietmar R Thal
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Philip Van Damme
- Department of Neurology, Neuromuscular Reference Center, University Hospitals Leuven, Leuven, Belgium.,Department of Neurosciences, Experimental Neurology, Leuven Brain Institute, KU Leuven-University of Leuven, Leuven, Belgium.,Laboratory of Neurobiology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| |
Collapse
|
137
|
The Biogenesis of miRNAs and Their Role in the Development of Amyotrophic Lateral Sclerosis. Cells 2022; 11:cells11030572. [PMID: 35159383 PMCID: PMC8833997 DOI: 10.3390/cells11030572] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/02/2022] [Accepted: 02/05/2022] [Indexed: 02/07/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that affects upper and lower motor neurons. As there is no effective treatment for ALS, it is particularly important to screen key gene therapy targets. The identifications of microRNAs (miRNAs) have completely changed the traditional view of gene regulation. miRNAs are small noncoding single-stranded RNA molecules involved in the regulation of post-transcriptional gene expression. Recent advances also indicate that miRNAs are biomarkers in many diseases, including neurodegenerative diseases. In this review, we summarize recent advances regarding the mechanisms underlying the role of miRNAs in ALS pathogenesis and its application to gene therapy for ALS. The potential of miRNAs to target diverse pathways opens a new avenue for ALS therapy.
Collapse
|
138
|
Flotte TR, Cataltepe O, Puri A, Batista AR, Moser R, McKenna-Yasek D, Douthwright C, Gernoux G, Blackwood M, Mueller C, Tai PWL, Jiang X, Bateman S, Spanakis SG, Parzych J, Keeler AM, Abayazeed A, Rohatgi S, Gibson L, Finberg R, Barton BA, Vardar Z, Shazeeb MS, Gounis M, Tifft CJ, Eichler FS, Brown RH, Martin DR, Gray-Edwards HL, Sena-Esteves M. AAV gene therapy for Tay-Sachs disease. Nat Med 2022; 28:251-259. [PMID: 35145305 PMCID: PMC10786171 DOI: 10.1038/s41591-021-01664-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 12/17/2021] [Indexed: 12/17/2022]
Abstract
Tay-Sachs disease (TSD) is an inherited neurological disorder caused by deficiency of hexosaminidase A (HexA). Here, we describe an adeno-associated virus (AAV) gene therapy expanded-access trial in two patients with infantile TSD (IND 18225) with safety as the primary endpoint and no secondary endpoints. Patient TSD-001 was treated at 30 months with an equimolar mix of AAVrh8-HEXA and AAVrh8-HEXB administered intrathecally (i.t.), with 75% of the total dose (1 × 1014 vector genomes (vg)) in the cisterna magna and 25% at the thoracolumbar junction. Patient TSD-002 was treated at 7 months by combined bilateral thalamic (1.5 × 1012 vg per thalamus) and i.t. infusion (3.9 × 1013 vg). Both patients were immunosuppressed. Injection procedures were well tolerated, with no vector-related adverse events (AEs) to date. Cerebrospinal fluid (CSF) HexA activity increased from baseline and remained stable in both patients. TSD-002 showed disease stabilization by 3 months after injection with ongoing myelination, a temporary deviation from the natural history of infantile TSD, but disease progression was evident at 6 months after treatment. TSD-001 remains seizure-free at 5 years of age on the same anticonvulsant therapy as before therapy. TSD-002 developed anticonvulsant-responsive seizures at 2 years of age. This study provides early safety and proof-of-concept data in humans for treatment of patients with TSD by AAV gene therapy.
Collapse
Affiliation(s)
- Terence R Flotte
- Department of Pediatrics, UMass Chan Medical School, Worcester, MA, USA.
- Horae Gene Therapy Center and The Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA, USA.
| | - Oguz Cataltepe
- Department of Pediatrics, UMass Chan Medical School, Worcester, MA, USA
- Department of Neurosurgery, UMass Chan Medical School, Worcester, MA, USA
| | - Ajit Puri
- Department of Radiology, UMass Chan Medical School, Worcester, MA, USA
| | - Ana Rita Batista
- Horae Gene Therapy Center and The Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA, USA
- Department of Neurology, UMass Chan Medical School, Worcester, MA, USA
| | - Richard Moser
- Department of Neurosurgery, UMass Chan Medical School, Worcester, MA, USA
| | | | | | - Gwladys Gernoux
- Department of Pediatrics, UMass Chan Medical School, Worcester, MA, USA
- Horae Gene Therapy Center and The Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA, USA
| | - Meghan Blackwood
- Department of Pediatrics, UMass Chan Medical School, Worcester, MA, USA
- Horae Gene Therapy Center and The Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA, USA
| | - Christian Mueller
- Department of Pediatrics, UMass Chan Medical School, Worcester, MA, USA
- Horae Gene Therapy Center and The Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA, USA
| | - Phillip W L Tai
- Horae Gene Therapy Center and The Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA, USA
| | - Xuntian Jiang
- Department of Medicine and Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Scot Bateman
- Department of Pediatrics, UMass Chan Medical School, Worcester, MA, USA
| | - Spiro G Spanakis
- Departments of Anesthesiology and Perioperative Medicine, UMass Chan Medical School, Worcester, MA, USA
| | - Julia Parzych
- Departments of Anesthesiology and Perioperative Medicine, UMass Chan Medical School, Worcester, MA, USA
| | - Allison M Keeler
- Department of Pediatrics, UMass Chan Medical School, Worcester, MA, USA
- Horae Gene Therapy Center and The Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA, USA
| | - Aly Abayazeed
- Department of Radiology, UMass Chan Medical School, Worcester, MA, USA
| | - Saurabh Rohatgi
- Department of Radiology, UMass Chan Medical School, Worcester, MA, USA
| | - Laura Gibson
- Department of Pediatrics, UMass Chan Medical School, Worcester, MA, USA
- Department of Internal Medicine, UMass Chan Medical School, Worcester, MA, USA
| | - Robert Finberg
- Department of Internal Medicine, UMass Chan Medical School, Worcester, MA, USA
| | - Bruce A Barton
- Population and Quantitative Health Sciences, UMass Chan Medical School, Worcester, MA, USA
| | - Zeynep Vardar
- Department of Radiology, UMass Chan Medical School, Worcester, MA, USA
| | | | - Matthew Gounis
- Department of Radiology, UMass Chan Medical School, Worcester, MA, USA
| | - Cynthia J Tifft
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Florian S Eichler
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Robert H Brown
- Horae Gene Therapy Center and The Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA, USA
- Department of Neurology, UMass Chan Medical School, Worcester, MA, USA
| | - Douglas R Martin
- Scott-Ritchey Research Center, Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Heather L Gray-Edwards
- Horae Gene Therapy Center and The Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA, USA
- Department of Radiology, UMass Chan Medical School, Worcester, MA, USA
| | - Miguel Sena-Esteves
- Horae Gene Therapy Center and The Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA, USA.
- Department of Neurology, UMass Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
139
|
Flotte TR, Gessler DJ. Gene Therapy for Rare Neurological Disorders. Clin Pharmacol Ther 2022; 111:743-757. [PMID: 35102556 DOI: 10.1002/cpt.2543] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 01/23/2022] [Indexed: 11/10/2022]
Abstract
There are over 7 000 diseases that are individually rare, but collectively affect missions of people worldwide. They are very commonly neurologic single-gene disorders. Recent advances in recombinant adeno-associated virus (rAAV) vectors have enabled breakthroughs, including FDA-approved gene therapies for Inherited Retinal Dystrophy due to RPE65 mutation and spinal muscular atrophy. A range of other gene therapies for rare neurologic diseases are at various stages of development. Future development of gene editing technologies promises further to broaden the potential for more patients with these disorders to benefit from innovative therapies.
Collapse
Affiliation(s)
| | - Dominic J Gessler
- University of Massachusetts Chan Medical School.,University of Minnesota
| |
Collapse
|
140
|
Tarantino N, Canfora I, Camerino GM, Pierno S. Therapeutic Targets in Amyotrophic Lateral Sclerosis: Focus on Ion Channels and Skeletal Muscle. Cells 2022; 11:cells11030415. [PMID: 35159225 PMCID: PMC8834084 DOI: 10.3390/cells11030415] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 02/04/2023] Open
Abstract
Amyotrophic Lateral Sclerosis is a neurodegenerative disease caused by progressive loss of motor neurons, which severely compromises skeletal muscle function. Evidence shows that muscle may act as a molecular powerhouse, whose final signals generate in patients a progressive loss of voluntary muscle function and weakness leading to paralysis. This pathology is the result of a complex cascade of events that involves a crosstalk among motor neurons, glia, and muscles, and evolves through the action of converging toxic mechanisms. In fact, mitochondrial dysfunction, which leads to oxidative stress, is one of the mechanisms causing cell death. It is a common denominator for the two existing forms of the disease: sporadic and familial. Other factors include excitotoxicity, inflammation, and protein aggregation. Currently, there are limited cures. The only approved drug for therapy is riluzole, that modestly prolongs survival, with edaravone now waiting for new clinical trial aimed to clarify its efficacy. Thus, there is a need of effective treatments to reverse the damage in this devastating pathology. Many drugs have been already tested in clinical trials and are currently under investigation. This review summarizes the already tested drugs aimed at restoring muscle-nerve cross-talk and on new treatment options targeting this tissue.
Collapse
|
141
|
Benatti HR, Gray-Edwards HL. Adeno-Associated Virus Delivery Limitations for Neurological Indications. Hum Gene Ther 2022; 33:1-7. [PMID: 35049369 DOI: 10.1089/hum.2022.29196.hrb] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Hector Ribeiro Benatti
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Heather L Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA.,Department of Radiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
142
|
Powell JE, Lim CKW, Krishnan R, McCallister TX, Saporito-Magriña C, Zeballos MA, McPheron GD, Gaj T. Targeted gene silencing in the nervous system with CRISPR-Cas13. SCIENCE ADVANCES 2022; 8:eabk2485. [PMID: 35044815 PMCID: PMC8769545 DOI: 10.1126/sciadv.abk2485] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 11/24/2021] [Indexed: 05/14/2023]
Abstract
Cas13 nucleases are a class of programmable RNA-targeting CRISPR effector proteins that are capable of silencing target gene expression in mammalian cells. Here, we demonstrate that RfxCas13d, a Cas13 ortholog with favorable characteristics to other family members, can be delivered to the mouse spinal cord and brain to silence neurodegeneration-associated genes. Intrathecally delivering an adeno-associated virus vector encoding an RfxCas13d variant programmed to target superoxide dismutase 1 (SOD1), a protein whose mutation can cause amyotrophic lateral sclerosis, reduced SOD1 mRNA and protein in the spinal cord by >50% and improved outcomes in a mouse model of the disorder. We further show that intrastriatally delivering an RfxCas13d variant programmed to target huntingtin (HTT), a protein whose mutation is causative for Huntington’s disease, led to a ~50% reduction in HTT protein in the mouse brain. Our results establish RfxCas13d as a versatile platform for knocking down gene expression in the nervous system.
Collapse
Affiliation(s)
- Jackson E. Powell
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA
| | - Colin K. W. Lim
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA
| | - Ramya Krishnan
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA
| | | | | | - Maria A. Zeballos
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA
| | | | - Thomas Gaj
- Department of Bioengineering, University of Illinois, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
143
|
Miccio A, Antoniou P, Ciura S, Kabashi E. Novel genome-editing-based approaches to treat motor neuron diseases: Promises and challenges. Mol Ther 2022; 30:47-53. [PMID: 33823304 PMCID: PMC8753272 DOI: 10.1016/j.ymthe.2021.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 01/07/2023] Open
Abstract
Motor neuron diseases are untreatable with common pharmacological approaches. Spinal muscular atrophy (SMA) is caused by SMN1 gene mutations leading to lowered SMN expression. Symptoms are alleviated in infants with a higher copy number of the SMN2 gene, which, however, displays a splicing defect resulting in low SMN levels. Amyotrophic lateral sclerosis (ALS) is caused by a number of mutations, with C9orf72 repeat expansions the most common genetic cause and SOD1 gain-of-function mutations the first genetic cause identified for this disease. Genetic therapies based on oligonucleotides that enhance SMN2 splicing and SMN production or lower SOD1 expression have shown promise in initial clinical trials for individuals with SMA and ALS harboring SOD1 mutations, respectively. Gene addition/silencing approaches using adeno-associated viruses (AAVs) are also currently under clinical investigation in trials for SMA and ALS. Here we provide a brief overview of these efforts and their advantages and challenges. We also review genome editing approaches aimed at correcting the disease-causing mutations or modulating the expression of genetic modifiers, e.g., by repairing SOD1 mutations or the SMN2 splicing defect or deleting C9orf72 expanded repeats. These studies have shown promising results to approach therapeutic trials that should significantly lower the progression of these deadly disorders.
Collapse
Affiliation(s)
- Annarita Miccio
- Laboratory of Chromatin and Gene Regulation during Development, Imagine Institute, Université de Paris, INSERM UMR 1163, 75015, Paris, France,Corresponding author: Annarita Miccio, Laboratory of Chromatin and Gene Regulation during Development, Imagine Institute, Université de Paris, INSERM UMR 1163, 75015, Paris, France.
| | - Panagiotis Antoniou
- Laboratory of Chromatin and Gene Regulation during Development, Imagine Institute, Université de Paris, INSERM UMR 1163, 75015, Paris, France
| | - Sorana Ciura
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, Université de Paris, INSERM UMR 1163, 75015, Paris, France
| | - Edor Kabashi
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, Université de Paris, INSERM UMR 1163, 75015, Paris, France,Corresponding author: Edor Kabashi, Laboratory of Translational Research for Neurological Disorders, Imagine Institute, Université de Paris, INSERM UMR 1163, 75015, Paris, France.
| |
Collapse
|
144
|
Hirunagi T, Sahashi K, Meilleur KG, Katsuno M. Nucleic Acid-Based Therapeutic Approach for Spinal and Bulbar Muscular Atrophy and Related Neurological Disorders. Genes (Basel) 2022; 13:genes13010109. [PMID: 35052449 PMCID: PMC8775157 DOI: 10.3390/genes13010109] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/01/2022] [Accepted: 01/03/2022] [Indexed: 12/14/2022] Open
Abstract
The recent advances in nucleic acid therapeutics demonstrate the potential to treat hereditary neurological disorders by targeting their causative genes. Spinal and bulbar muscular atrophy (SBMA) is an X-linked and adult-onset neurodegenerative disorder caused by the expansion of trinucleotide cytosine-adenine-guanine repeats, which encodes a polyglutamine tract in the androgen receptor gene. SBMA belongs to the family of polyglutamine diseases, in which the use of nucleic acids for silencing a disease-causing gene, such as antisense oligonucleotides and small interfering RNAs, has been intensively studied in animal models and clinical trials. A unique feature of SBMA is that both motor neuron and skeletal muscle pathology contribute to disease manifestations, including progressive muscle weakness and atrophy. As both motor neurons and skeletal muscles can be therapeutic targets in SBMA, nucleic acid-based approaches for other motor neuron diseases and myopathies may further lead to the development of a treatment for SBMA. Here, we review studies of nucleic acid-based therapeutic approaches in SBMA and related neurological disorders and discuss current limitations and perspectives to apply these approaches to patients with SBMA.
Collapse
Affiliation(s)
- Tomoki Hirunagi
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Syowa-ku, Nagoya 466-8550, Japan; (T.H.); (K.S.)
| | - Kentaro Sahashi
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Syowa-ku, Nagoya 466-8550, Japan; (T.H.); (K.S.)
| | - Katherine G. Meilleur
- Research and Clinical Development, Neuromuscular Development Unit, Biogen, 300, Binney Street, Cambridge, MA 02142, USA;
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Syowa-ku, Nagoya 466-8550, Japan; (T.H.); (K.S.)
- Department of Clinical Research Education, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Syowa-ku, Nagoya 466-8550, Japan
- Correspondence:
| |
Collapse
|
145
|
Jablonka S, Hennlein L, Sendtner M. Therapy development for spinal muscular atrophy: perspectives for muscular dystrophies and neurodegenerative disorders. Neurol Res Pract 2022; 4:2. [PMID: 34983696 PMCID: PMC8725368 DOI: 10.1186/s42466-021-00162-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/21/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Major efforts have been made in the last decade to develop and improve therapies for proximal spinal muscular atrophy (SMA). The introduction of Nusinersen/Spinraza™ as an antisense oligonucleotide therapy, Onasemnogene abeparvovec/Zolgensma™ as an AAV9-based gene therapy and Risdiplam/Evrysdi™ as a small molecule modifier of pre-mRNA splicing have set new standards for interference with neurodegeneration. MAIN BODY Therapies for SMA are designed to interfere with the cellular basis of the disease by modifying pre-mRNA splicing and enhancing expression of the Survival Motor Neuron (SMN) protein, which is only expressed at low levels in this disorder. The corresponding strategies also can be applied to other disease mechanisms caused by loss of function or toxic gain of function mutations. The development of therapies for SMA was based on the use of cell culture systems and mouse models, as well as innovative clinical trials that included readouts that had originally been introduced and optimized in preclinical studies. This is summarized in the first part of this review. The second part discusses current developments and perspectives for amyotrophic lateral sclerosis, muscular dystrophies, Parkinson's and Alzheimer's disease, as well as the obstacles that need to be overcome to introduce RNA-based therapies and gene therapies for these disorders. CONCLUSION RNA-based therapies offer chances for therapy development of complex neurodegenerative disorders such as amyotrophic lateral sclerosis, muscular dystrophies, Parkinson's and Alzheimer's disease. The experiences made with these new drugs for SMA, and also the experiences in AAV gene therapies could help to broaden the spectrum of current approaches to interfere with pathophysiological mechanisms in neurodegeneration.
Collapse
Affiliation(s)
- Sibylle Jablonka
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Versbacher Str. 5, 97078, Wuerzburg, Germany.
| | - Luisa Hennlein
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Versbacher Str. 5, 97078, Wuerzburg, Germany
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Versbacher Str. 5, 97078, Wuerzburg, Germany.
| |
Collapse
|
146
|
Muhuri M, Gao G. Membranous Bubbles: High-Purity and High-Titer Exosomes as the Potential Solution for Adeno-Associated Viruses to Evade Neutralization? Hum Gene Ther 2021; 32:1427-1429. [PMID: 34935455 DOI: 10.1089/hum.2021.29189.mmu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Manish Muhuri
- Horae Gene Therapy Center.,Department of Microbiology and Physiological Systems.,VIDE Program, and
| | - Guangping Gao
- Horae Gene Therapy Center.,Department of Microbiology and Physiological Systems.,Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
147
|
Taghian T, Batista AR, Kamper S, Caldwell M, Lilley L, Li H, Rodriguez P, Mesa K, Zheng S, King RM, Gounis MJ, Todeasa S, Maguire A, Martin DR, Sena-Esteves M, Meade TJ, Gray-Edwards HL. Real-time MR tracking of AAV gene therapy with βgal-responsive MR probe in a murine model of GM1-gangliosidosis. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 23:128-134. [PMID: 34703836 PMCID: PMC8517204 DOI: 10.1016/j.omtm.2021.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/16/2021] [Indexed: 11/16/2022]
Abstract
Transformative results of adeno-associated virus (AAV) gene therapy in patients with spinal muscular atrophy and Leber's congenital amaurosis led to approval of the first two AAV products in the United States to treat these diseases. These extraordinary results led to a dramatic increase in the number and type of AAV gene-therapy programs. However, the field lacks non-invasive means to assess levels and duration of therapeutic protein function in patients. Here, we describe a new magnetic resonance imaging (MRI) technology for real-time reporting of gene-therapy products in the living animal in the form of an MRI probe that is activated in the presence of therapeutic protein expression. For the first time, we show reliable tracking of enzyme expression after a now in-human clinical trial AAV gene therapy (ClinicalTrials.gov: NTC03952637) encoding lysosomal acid beta-galactosidase (βgal) using a self-immolative βgal-responsive MRI probe. MRI enhancement in AAV-treated enzyme-deficient mice (GLB-1-/-) correlates with βgal activity in central nervous system and peripheral organs after intracranial or intravenous AAV gene therapy, respectively. With >1,800 gene therapies in phase I/II clinical trials (ClinicalTrials.gov), development of a non-invasive method to track gene expression over time in patients is crucial to the future of the gene-therapy field.
Collapse
Affiliation(s)
- Toloo Taghian
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA.,Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Ana Rita Batista
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA.,Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Sarah Kamper
- Departments of Chemistry, Molecular Biosciences, Neurobiology and Radiology, Northwestern University, Evanston, IL 60208, USA
| | - Michael Caldwell
- Departments of Chemistry, Molecular Biosciences, Neurobiology and Radiology, Northwestern University, Evanston, IL 60208, USA
| | - Laura Lilley
- Departments of Chemistry, Molecular Biosciences, Neurobiology and Radiology, Northwestern University, Evanston, IL 60208, USA
| | - Hao Li
- Departments of Chemistry, Molecular Biosciences, Neurobiology and Radiology, Northwestern University, Evanston, IL 60208, USA
| | - Paola Rodriguez
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA.,Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Katerina Mesa
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA.,Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Shaokuan Zheng
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Robert M King
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA.,Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Matthew J Gounis
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Sophia Todeasa
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA.,Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Anne Maguire
- Scott-Ritchey Research Center, Auburn University, Auburn, AL 36849, USA
| | - Douglas R Martin
- Scott-Ritchey Research Center, Auburn University, Auburn, AL 36849, USA
| | - Miguel Sena-Esteves
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA.,Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Thomas J Meade
- Departments of Chemistry, Molecular Biosciences, Neurobiology and Radiology, Northwestern University, Evanston, IL 60208, USA
| | - Heather L Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA.,Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
148
|
George LA. Hemophilia gene therapy: ushering in a new treatment paradigm? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2021; 2021:226-233. [PMID: 34889378 PMCID: PMC8877054 DOI: 10.1182/hematology.2021000254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
After 3 decades of clinical trials, repeated proof-of-concept success has now been demonstrated in hemophilia A and B gene therapy. Current clinical hemophilia gene therapy efforts are largely focused on the use of systemically administered recombinant adeno-associated viral (rAAV) vectors for F8 or F9 gene addition. With multiple ongoing trials, including licensing studies in hemophilia A and B, many are cautiously optimistic that the first AAV vectors will obtain regulatory approval within approximately 1 year. While supported optimism suggests that the goal of gene therapy to alter the paradigm of hemophilia care may soon be realized, a number of outstanding questions have emerged from clinical trial that are in need of answers to harness the full potential of gene therapy for hemophilia patients. This article reviews the use of AAV vector gene addition approaches for hemophilia A and B, focusing specifically on information to review in the process of obtaining informed consent for hemophilia patients prior to clinical trial enrollment or administering a licensed AAV vector.
Collapse
Affiliation(s)
- Lindsey A. George
- Correspondence Lindsey A. George, University of Pennsylvania School of Medicine, Colket Translational Research Bldg, Rm 5016, 3501 Civic Center Blvd, Philadelphia, PA 19104; e-mail:
| |
Collapse
|
149
|
Chipika RH, Siah WF, McKenna MC, Li Hi Shing S, Hardiman O, Bede P. The presymptomatic phase of amyotrophic lateral sclerosis: are we merely scratching the surface? J Neurol 2021; 268:4607-4629. [PMID: 33130950 DOI: 10.1007/s00415-020-10289-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023]
Abstract
Presymptomatic studies in ALS have consistently captured considerable disease burden long before symptom manifestation and contributed important academic insights. With the emergence of genotype-specific therapies, however, there is a pressing need to address practical objectives such as the estimation of age of symptom onset, phenotypic prediction, informing the optimal timing of pharmacological intervention, and identifying a core panel of biomarkers which may detect response to therapy. Existing presymptomatic studies in ALS have adopted striking different study designs, relied on a variety of control groups, used divergent imaging and electrophysiology methods, and focused on different genotypes and demographic groups. We have performed a systematic review of existing presymptomatic studies in ALS to identify common themes, stereotyped shortcomings, and key learning points for future studies. Existing presymptomatic studies in ALS often suffer from sample size limitations, lack of disease controls and rarely follow their cohort until symptom manifestation. As the characterisation of presymptomatic processes in ALS serves a multitude of academic and clinical purposes, the careful review of existing studies offers important lessons for future initiatives.
Collapse
Affiliation(s)
- Rangariroyashe H Chipika
- Computational Neuroimaging Group (CNG), Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin, Ireland
| | - We Fong Siah
- Computational Neuroimaging Group (CNG), Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin, Ireland
| | - Mary Clare McKenna
- Computational Neuroimaging Group (CNG), Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin, Ireland
| | - Stacey Li Hi Shing
- Computational Neuroimaging Group (CNG), Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin, Ireland
| | - Orla Hardiman
- Computational Neuroimaging Group (CNG), Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin, Ireland
| | - Peter Bede
- Computational Neuroimaging Group (CNG), Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin, Ireland.
| |
Collapse
|
150
|
Wang W, Zhou P, Wang X, Chen F, Christensen E, Thompson J, Ren X, Kells A, Stanek L, Carter T, Hou J, Sah D. Efficient and Precise Processing of the Optimized Pri-amiRNA in a Huntingtin-Lowering AAV Gene Therapy in Vitro and in Mice and Nonhuman Primates. Hum Gene Ther 2021; 33:37-60. [PMID: 34806402 PMCID: PMC10112875 DOI: 10.1089/hum.2021.221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Huntington's Disease is a fatal neurodegenerative disorder caused by an inherited mutation in the huntingtin gene (HTT) comprising an expanded cytosine-adenine-guanine (CAG) trinucleotide repeat sequence that results in a pathogenic huntingtin protein. AAV gene therapy containing a primary artificial microRNA (pri-amiRNA) specifically targeting HTT mRNA has the potential to provide long-lasting therapeutic benefit, via durable reduction of mutant HTT expression after a single administration. The efficiency and precision of processing of the pri-amiRNA precursor to the mature guide strand by transduced cells is critical for specific and potent HTT lowering. The selection of the optimized pri-amiRNA comprised a series of in vitro studies followed by in vivo studies in small and then large mammals. Our studies demonstrate the predictivity of certain cell culture systems and rodent models for nonhuman primates (NHP) with respect to some, but not all key features of pri-amiRNA processing. In addition, our results show that the processing of pri-amiRNAs to the mature guide strand can differ greatly across different scaffolds and sequences while providing the same levels of target lowering. Importantly, our data demonstrate that there is a combinatorial effect of guide and passenger strand sequences, together with the scaffold, on pri-amiRNA processing, with different guide and passenger strand sequences within the same scaffold dramatically altering pri-amiRNA processing. Taken together, our results highlight the importance of optimizing not only target lowering, but also the efficiency and precision of pri-amiRNA processing in vitro, in rodents and in large mammals to identify the most potent and selective AAV gene therapy that harnesses the endogenous miRNA biogenesis pathway for target lowering without perturbing the endogenous cellular miRNA profile. The optimized pri-amiRNA was selected with this focus on efficiency and precision of pri-amiRNA processing in addition to its pharmacological activity on HTT lowering, and general tolerability in vivo.
Collapse
Affiliation(s)
- Wei Wang
- Voyager Therapeutics Inc, 461444, Cambridge, Massachusetts, United States;
| | - Pengcheng Zhou
- Voyager Therapeutics Inc, 461444, Cambridge, Massachusetts, United States;
| | - Xin Wang
- Voyager Therapeutics Inc, 461444, Cambridge, Massachusetts, United States;
| | - Fen Chen
- Voyager Therapeutics Inc, 461444, Cambridge, Massachusetts, United States;
| | - Emily Christensen
- Voyager Therapeutics Inc, 461444, Cambridge, Massachusetts, United States;
| | - Jeffrey Thompson
- Voyager Therapeutics Inc, 461444, Cambridge, Massachusetts, United States;
| | - Xiaoqin Ren
- Voyager Therapeutics Inc, 461444, Cambridge, Massachusetts, United States;
| | - Adrian Kells
- Voyager Therapeutics Inc, 461444, Cambridge, Massachusetts, United States;
| | - Lisa Stanek
- Sanofi Genzyme, 2194, Cambridge, Massachusetts, United States;
| | - Todd Carter
- Voyager Therapeutics Inc, 461444, Cambridge, Massachusetts, United States;
| | - Jay Hou
- Voyager Therapeutics Inc, 461444, Cambridge, Massachusetts, United States;
| | - Dinah Sah
- Voyager Therapeutics Inc, 461444, 75 Sidney Street, Cambridge, Massachusetts, United States, 02139;
| |
Collapse
|