1
|
Klinger L, Siegert ASM, Holzinger R, Trofimova L, Ada S, Drdla-Schutting R. An evaluation of distinct adeno-associated virus vector strategies for driving transgene expression in spinal inhibitory neurons of the rat. Front Neurosci 2025; 19:1558581. [PMID: 40376611 PMCID: PMC12078132 DOI: 10.3389/fnins.2025.1558581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/21/2025] [Indexed: 05/18/2025] Open
Abstract
The spinal cord dorsal horn (DH) is essential for processing and transmitting nociceptive information. Its neuronal subpopulations exhibit significant heterogeneity in morphology and intrinsic properties, forming complex circuits that remain only partially understood. Under physiological and pathological conditions, inhibitory interneurons in the DH are of particular interest. These neurons modulate and refine pain-related signals entering the central nervous system. The ability to selectively target these inhibitory interneurons is key to investigating the underlying circuitry and mechanisms of pain processing, as well as to understand the specific role of inhibitory signaling within these processes. We employed a viral vector approach to deliver a fluorescent reporter protein specifically to inhibitory interneurons in the rat spinal cord. Using adeno-associated virus (AAV) vectors designed to express enhanced green fluorescent protein (EGFP) under the control of various promoters, we targeted distinct subtypes of spinal inhibitory interneurons. Through immunostaining, in situ hybridization, and confocal imaging, we evaluated the specificity and efficacy of these promoters. Our findings revealed that the promoter/vector combinations used did not achieve the desired specificity for targeting distinct interneuron populations in the DH. Despite these limitations, this work provides valuable insights into the potential and challenges of designing AAV-based approaches for selective neuronal targeting. These results emphasize the need for further refinement of promoter designs to achieve precise and reliable expression in specific spinal interneuron subtypes. Addressing these challenges will be crucial for advancing our understanding of spinal nociceptive circuits and developing targeted therapeutic approaches for pain syndromes.
Collapse
Affiliation(s)
| | | | | | | | | | - Ruth Drdla-Schutting
- Division of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Sevigny J, Uspenskaya O, Heckman LD, Wong LC, Hatch DA, Tewari A, Vandenberghe R, Irwin DJ, Saracino D, Le Ber I, Ahmed R, Rohrer JD, Boxer AL, Boland S, Sheehan P, Brandes A, Burstein SR, Shykind BM, Kamalakaran S, Daniels CW, David Litwack E, Mahoney E, Velaga J, McNamara I, Sondergaard P, Sajjad SA, Kobayashi YM, Abeliovich A, Hefti F. Progranulin AAV gene therapy for frontotemporal dementia: translational studies and phase 1/2 trial interim results. Nat Med 2024; 30:1406-1415. [PMID: 38745011 PMCID: PMC11108785 DOI: 10.1038/s41591-024-02973-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 04/03/2024] [Indexed: 05/16/2024]
Abstract
GRN mutations cause progranulin haploinsufficiency, which eventually leads to frontotemporal dementia (FTD-GRN). PR006 is an investigational gene therapy delivering the granulin gene (GRN) using an adeno-associated virus serotype 9 (AAV9) vector. In non-clinical studies, PR006 transduced neurons derived from induced pluripotent stem cells of patients with FTD-GRN, resulted in progranulin expression and improvement of lipofuscin, lysosomal and neuroinflammation pathologies in Grn-knockout mice, and was well tolerated except for minimal, asymptomatic dorsal root ganglionopathy in non-human primates. We initiated a first-in-human phase 1/2 open-label trial. Here we report results of a pre-specified interim analysis triggered with the last treated patient of the low-dose cohort (n = 6) reaching the 12-month follow-up timepoint. We also include preliminary data from the mid-dose cohort (n = 7). Primary endpoints were safety, immunogenicity and change in progranulin levels in cerebrospinal fluid (CSF) and blood. Secondary endpoints were Clinical Dementia Rating (CDR) plus National Alzheimer's Disease Coordinating Center (NACC) Frontotemporal Lobar Degeneration (FTLD) rating scale and levels of neurofilament light chain (NfL). One-time administration of PR006 into the cisterna magna was generally safe and well tolerated. All patients developed treatment-emergent anti-AAV9 antibodies in the CSF, but none developed anti-progranulin antibodies. CSF pleocytosis was the most common PR006-related adverse event. Twelve serious adverse events occurred, mostly unrelated to PR006. Deep vein thrombosis developed in three patients. There was one death (unrelated) occurring 18 months after treatment. CSF progranulin increased after PR006 treatment in all patients; blood progranulin increased in most patients but only transiently. NfL levels transiently increased after PR006 treatment, likely reflecting dorsal root ganglia toxicity. Progression rates, based on the CDR scale, were within the broad ranges reported for patients with FTD. These data provide preliminary insights into the safety and bioactivity of PR006. Longer follow-up and additional studies are needed to confirm the safety and potential efficacy of PR006. ClinicalTrials.gov identifier: NCT04408625 .
Collapse
Affiliation(s)
- Jeffrey Sevigny
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA.
| | - Olga Uspenskaya
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Laura Dean Heckman
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Li Chin Wong
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Daniel A Hatch
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Ambika Tewari
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Rik Vandenberghe
- Neurology Service, University Hospitals Leuven, Leuven, Belgium and Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - David J Irwin
- Department of Neurology, Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Dario Saracino
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau, ICM, Inserm, CNRS UMR 7225 APHP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau, ICM, Inserm, CNRS UMR 7225 APHP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Rebekah Ahmed
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Jonathan D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Center, UCL Queen Square Institute of Neurology, London, UK
| | - Adam L Boxer
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Sebastian Boland
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Patricia Sheehan
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Alissa Brandes
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Suzanne R Burstein
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Benjamin M Shykind
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Sitharthan Kamalakaran
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Carter W Daniels
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - E David Litwack
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Erin Mahoney
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Jenny Velaga
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Ilan McNamara
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Patricia Sondergaard
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Syed A Sajjad
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Yvonne M Kobayashi
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Asa Abeliovich
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| | - Franz Hefti
- Prevail Therapeutics, a wholly owned subsidiary of Eli Lilly and Company, New York, NY, USA
| |
Collapse
|
3
|
Rosenberg JB, Fung EK, Dyke JP, De BP, Lou H, Kelly JM, Reejhsinghani L, Ricart Arbona RJ, Sondhi D, Kaminsky SM, Cartier N, Hinderer C, Hordeaux J, Wilson JM, Ballon DJ, Crystal RG. Positron Emission Tomography Quantitative Assessment of Off-Target Whole-Body Biodistribution of I-124-Labeled Adeno-Associated Virus Capsids Administered to Cerebral Spinal Fluid. Hum Gene Ther 2023; 34:1095-1106. [PMID: 37624734 PMCID: PMC10659018 DOI: 10.1089/hum.2023.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/09/2023] [Indexed: 08/27/2023] Open
Abstract
Based on studies in experimental animals demonstrating that administration of adeno-associated virus (AAV) vectors to the cerebrospinal fluid (CSF) is an effective route to transfer genes to the nervous system, there are increasing number of clinical trials using the CSF route to treat nervous system disorders. With the knowledge that the CSF turns over four to five times daily, and evidence in experimental animals that at least some of CSF administered AAV vectors are distributed to systemic organs, we asked: with AAV administration to the CSF, what fraction of the total dose remains in the nervous system and what fraction goes off target and is delivered systemically? To quantify the biodistribution of AAV capsids immediately after administration, we covalently labeled AAV capsids with iodine 124 (I-124), a cyclotron generated positron emitter, enabling quantitative positron emission tomography scanning of capsid distribution for up to 96 h after AAV vector administration. We assessed the biodistribution to nonhuman primates of I-124-labeled capsids from different AAV clades, including 9 (clade F), rh.10 (E), PHP.eB (F), hu68 (F), and rh91(A). The analysis demonstrated that 60-90% of AAV vectors administered to the CSF through either the intracisternal or intrathecal (lumbar) routes distributed systemically to major organs. These observations have potentially significant clinical implications regarding accuracy of AAV vector dosing to the nervous system, evoking systemic immunity at levels similar to that with systemic administration, and potential toxicity of genes designed to treat nervous system disorders being expressed in non-nervous system organs. Based on these data, individuals in clinical trials using AAV vectors administered to the CSF should be monitored for systemic as well as nervous system adverse events and CNS dosing considerations should account for a significant AAV systemic distribution.
Collapse
Affiliation(s)
| | - Edward K. Fung
- Department of Radiology, Citigroup Biomedical Imaging Center; Weill Cornell Medicine, New York, New York, USA
| | - Jonathan P. Dyke
- Department of Radiology, Citigroup Biomedical Imaging Center; Weill Cornell Medicine, New York, New York, USA
| | | | | | - James M. Kelly
- Department of Radiology, Citigroup Biomedical Imaging Center; Weill Cornell Medicine, New York, New York, USA
| | - Layla Reejhsinghani
- Department of Radiology, Citigroup Biomedical Imaging Center; Weill Cornell Medicine, New York, New York, USA
| | - Rodolfo J. Ricart Arbona
- Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, New York, USA
| | | | | | - Nathalie Cartier
- Neurogencell INSERM U1127 Paris Brain Institute, Paris Sorbonne University, Paris, France; and
| | - Christian Hinderer
- Gene Therapy Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Juliette Hordeaux
- Gene Therapy Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - James M. Wilson
- Gene Therapy Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Douglas J. Ballon
- Department of Genetic Medicine
- Department of Radiology, Citigroup Biomedical Imaging Center; Weill Cornell Medicine, New York, New York, USA
| | | |
Collapse
|
4
|
De BP, Rosenberg JB, Selvan N, Wilson I, Yusufzai N, Greco A, Kaminsky SM, Heier LA, Ricart Arbona RJ, Miranda IC, Monette S, Nair A, Khanna R, Crystal RG, Sondhi D. Assessment of Safety and Biodistribution of AAVrh.10hCLN2 Following Intracisternal Administration in Nonhuman Primates for the Treatment of CLN2 Batten Disease. Hum Gene Ther 2023; 34:905-916. [PMID: 37624739 PMCID: PMC10517331 DOI: 10.1089/hum.2023.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/10/2023] [Indexed: 08/27/2023] Open
Abstract
CLN2 disease is a fatal, childhood autosomal recessive disorder caused by mutations in ceroid lipofuscinosis type 2 (CLN2) gene, encoding tripeptidyl peptidase 1 (TPP-1). Loss of TPP-1 activity leads to accumulation of storage material in lysosomes and resultant neuronal cell death with neurodegeneration. Genotype/phenotype comparisons suggest that the phenotype should be ameliorated with increase of TPP-1 levels to 5-10% of normal with wide central nervous system (CNS) distribution. Our previous clinical study showed that intraparenchymal (IPC) administration of AAVrh.10hCLN2, an adeno-associated vector serotype rh.10 encoding human CLN2, slowed, but did not stop disease progression, suggesting that this may be insufficient to distribute the therapy throughout the CNS (Sondhi 2020). In this study, we assessed whether the less invasive intracisternal delivery route would be safe and provide a wider distribution of TPP-1. A study was conducted in nonhuman primates (NHPs) with intracisternal delivery to cerebrospinal fluid (CSF) of AAVrh.10hCLN2 (5 × 1013 genome copies) or phosphate buffered saline (PBS). No abnormal behavior was noted. CNS magnetic resonance imaging and clinical chemistry data were all unremarkable. Histopathology of major organs had no abnormal finding attributable to the intervention or the vector, except that in one out of two animals treated with AAVrh.10hCLN2, dorsal root ganglia showed mild-to-moderate mononuclear cell infiltrates and neuronal degeneration. In contrast to our previous NHP study (Sondhi 2012) with IPC administration where TPP-1 activity was >2 × above controls in 30% of treated brains, in the two intracisternal treated NHPs, the TPP-1 activity was >2 × above controls in 50% and 41% of treated brains, and 52% and 84% of brain had >1,000 vector genomes/μg DNA, compared to 0% in the two PBS NHP. CSF TPP1 levels in treated animals were 43-62% of normal human levels. Collectively, these data indicate that AAVrh.10hCLN2 delivered by intracisternal route is safe and widely distributes TPP-1 in brain and CSF at levels that are potentially therapeutic. Clinical Trial Registration: NCT02893826, NCT04669535, NCT04273269, NCT03580083, NCT04408625, NCT04127578, and NCT04792944.
Collapse
Affiliation(s)
- Bishnu P. De
- Department of Genetic Medicine, New York, New York, USA
| | | | | | | | | | | | | | - Linda A. Heier
- Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - Rodolfo J. Ricart Arbona
- Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Ileana C. Miranda
- Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, The Rockefeller University, Weill Cornell Medicine, New York, New York, USA
| | - Sebastien Monette
- Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, The Rockefeller University, Weill Cornell Medicine, New York, New York, USA
| | - Anju Nair
- LEXEO Therapeutics, New York, New York, USA
| | | | | | - Dolan Sondhi
- Department of Genetic Medicine, New York, New York, USA
| |
Collapse
|
5
|
Eftekharpour E, Shcholok T. Cre-recombinase systems for induction of neuron-specific knockout models: a guide for biomedical researchers. Neural Regen Res 2023; 18:273-279. [PMID: 35900402 PMCID: PMC9396489 DOI: 10.4103/1673-5374.346541] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Gene deletion has been a valuable tool for unraveling the mysteries of molecular biology. Early approaches included gene trapping and gene targetting to disrupt or delete a gene randomly or at a specific location, respectively. Using these technologies in mouse embryos led to the generation of mouse knockout models and many scientific discoveries. The efficacy and specificity of these approaches have significantly increased with the advent of new technology such as clustered regularly interspaced short palindromic repeats for targetted gene deletion. However, several limitations including unwanted off-target gene deletion have hindered their widespread use in the field. Cre-recombinase technology has provided additional capacity for cell-specific gene deletion. In this review, we provide a summary of currently available literature on the application of this system for targetted deletion of neuronal genes. This article has been constructed to provide some background information for the new trainees on the mechanism and to provide necessary information for the design, and application of the Cre-recombinase system through reviewing the most frequent promoters that are currently available for genetic manipulation of neurons. We additionally will provide a summary of the latest technological developments that can be used for targeting neurons. This may also serve as a general guide for the selection of appropriate models for biomedical research.
Collapse
|
6
|
Hordeaux J, Jeffrey BA, Jian J, Choudhury GR, Michalson K, Mitchell TW, Buza EL, Chichester J, Dyer C, Bagel J, Vite CH, Bradbury AM, Wilson JM. Efficacy and Safety of a Krabbe Disease Gene Therapy. Hum Gene Ther 2022; 33:499-517. [PMID: 35333110 PMCID: PMC9142772 DOI: 10.1089/hum.2021.245] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Krabbe disease is a lysosomal storage disease caused by mutations in the gene that encodes galactosylceramidase, in which galactosylsphingosine (psychosine) accumulation drives demyelination in the central and peripheral nervous systems, ultimately progressing to death in early childhood. Gene therapy, alone or in combination with transplant, has been developed for almost two decades in mouse models, with increasing therapeutic benefit paralleling the improvement of next-generation adeno-associated virus (AAV) vectors. This effort has recently shown remarkable efficacy in the canine model of the disease by two different groups that used either systemic or cerebrospinal fluid (CSF) administration of AAVrh10 or AAV9. Building on our experience developing CSF-delivered, AAV-based drug products for a variety of neurodegenerative disorders, we conducted efficacy, pharmacology, and safety studies of AAVhu68 delivered to the CSF in two relevant natural Krabbe animal models, and in nonhuman primates. In newborn Twitcher mice, the highest dose (1 × 1011 genome copies [GC]) of AAVhu68.hGALC injected into the lateral ventricle led to a median survival of 130 days compared to 40.5 days in vehicle-treated mice. When this dose was administered intravenously, the median survival was 49 days. A single intracisterna magna injection of AAVhu68.cGALC at 3 × 1013 GC into presymptomatic Krabbe dogs increased survival for up to 85 weeks compared to 12 weeks in controls. It prevented psychosine accumulation in the CSF, preserved peripheral nerve myelination, ambulation, and decreased brain neuroinflammation and demyelination, although some regions remained abnormal. In a Good Laboratory Practice-compliant toxicology study, we administered the clinical candidate into the cisterna magna of 18 juvenile rhesus macaques at 3 doses that displayed efficacy in mice. We observed no dose-limiting toxicity and sporadic minimal degeneration of dorsal root ganglia (DRG) neurons. Our studies demonstrate the efficacy, scalability, and safety of a single cisterna magna AAVhu68 administration to treat Krabbe disease. ClinicalTrials.Gov ID: NCT04771416.
Collapse
Affiliation(s)
- Juliette Hordeaux
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brianne A Jeffrey
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jinlong Jian
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gourav R Choudhury
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kristofer Michalson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Thomas W Mitchell
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth L Buza
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jessica Chichester
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cecilia Dyer
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jessica Bagel
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Charles H Vite
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Allison M Bradbury
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Ozgür-Günes Y, Chedik M, LE Stunff C, Fovet CM, Bougneres P. Long-term disease prevention with a gene therapy targeting oligodendrocytes in a mouse model of adrenomyeloneuropathy. Hum Gene Ther 2022; 33:936-949. [PMID: 35166123 DOI: 10.1089/hum.2021.293] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Adrenomyeloneuropathy (AMN) is a late-onset axonopathy of spinal cord tracts caused by mutations of the ABCD1 gene that encodes ALDP, a peroxisomal transporter of very long chain fatty acids (VLCFA). Disturbed metabolic interaction between oligodendrocytes (OL) and axons is suspected to play a major role in AMN axonopathy. To develop a vector targeting OL, the human ABCD1 gene driven by a short 0.3 kb part of the human myelin-associated glycoprotein (MAG) promoter was packaged into an adeno-associated viral serotype 9 (rAAV9). An intravenous injection of this vector at postnatal day 10 (P10) in Abcd1-/- mice, a model of AMN, allowed a near normal motor performance to persist for 24 months, while age-matched untreated mice developed major defects of balance and motricity. Three weeks post vector, 50-54% of spinal cord white matter OL were expressing ALDP at the cervical level, and only 6-7% after 24 months. In addition, 29-32% of cervical spinal cord astrocytes at 3 weeks and 16-19% at 24 months also expressed ALDP. C26:0-lysoPC, a sensitive VLCFA marker of AMN, was lower by 41% and 50%, respectively in the spinal cord and brain of vector-treated compared with untreated mice. In a non-human primate (NHP), the intrathecal injection of the rAAV9-MAG vector induced abundant ALDP expression at 3 weeks in spinal cord OL (43%, 29%, 26% at cervical, thoracic and lumbar levels) and cerebellum OL (35%). In addition, 33-41 % of spinal cord astrocytes expressed hALDP, and 27% of cerebellar astrocytes. To our knowledge, OL targeting had not been obtained before in primates with other vectors or promoters. The current results thus provide a robust proof-of-concept not only for the gene therapy of AMN but for other CNS diseases where the targeting of OL with the rAAV9-MAG vector may be of interest.
Collapse
Affiliation(s)
| | - Malha Chedik
- INSERM, 27102, Le Kremlin-Bicêtre, Île-de-France, France;
| | | | | | - Pierre Bougneres
- INSERM, 27102, 80 rue du Général Leclercc, Le Kremlin Bicêtre, France, 94276;
| |
Collapse
|
8
|
Fischell JM, Fishman PS. A Multifaceted Approach to Optimizing AAV Delivery to the Brain for the Treatment of Neurodegenerative Diseases. Front Neurosci 2021; 15:747726. [PMID: 34630029 PMCID: PMC8497810 DOI: 10.3389/fnins.2021.747726] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022] Open
Abstract
Despite major advancements in gene therapy technologies, there are no approved gene therapies for diseases which predominantly effect the brain. Adeno-associated virus (AAV) vectors have emerged as the most effective delivery vector for gene therapy owing to their simplicity, wide spread transduction and low immunogenicity. Unfortunately, the blood-brain barrier (BBB) makes IV delivery of AAVs, to the brain highly inefficient. At IV doses capable of widespread expression in the brain, there is a significant risk of severe immune-mediated toxicity. Direct intracerebral injection of vectors is being attempted. However, this method is invasive, and only provides localized delivery for diseases known to afflict the brain globally. More advanced methods for AAV delivery will likely be required for safe and effective gene therapy to the brain. Each step in AAV delivery, including delivery route, BBB transduction, cellular tropism and transgene expression provide opportunities for innovative solutions to optimize delivery efficiency. Intra-arterial delivery with mannitol, focused ultrasound, optimized AAV capsid evolution with machine learning algorithms, synthetic promotors are all examples of advanced strategies which have been developed in pre-clinical models, yet none are being investigated in clinical trials. This manuscript seeks to review these technological advancements, and others, to improve AAV delivery to the brain, and to propose novel strategies to build upon this research. Ultimately, it is hoped that the optimization of AAV delivery will allow for the human translation of many gene therapies for neurodegenerative and other neurologic diseases.
Collapse
Affiliation(s)
- Jonathan M Fischell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Paul S Fishman
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
9
|
Gross AL, Gray-Edwards HL, Bebout CN, Ta NL, Nielsen K, Brunson BL, Mercado KRL, Osterhoudt DE, Batista AR, Maitland S, Seyfried TN, Sena-Esteves M, Martin DR. Intravenous delivery of adeno-associated viral gene therapy in feline GM1 gangliosidosis. Brain 2021; 145:655-669. [PMID: 34410345 DOI: 10.1093/brain/awab309] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/12/2021] [Accepted: 07/28/2021] [Indexed: 11/14/2022] Open
Abstract
GM1 gangliosidosis is a fatal neurodegenerative disease caused by a deficiency of lysosomal β-galactosidase. In its most severe form, GM1 gangliosidosis causes death by 4 years of age, and no effective treatments exist. Previous work has shown that injection of the brain parenchyma with an adeno-associated viral vector provides pronounced therapeutic benefit in a feline GM1 model. To develop a less invasive treatment for the brain and increase systemic biodistribution, intravenous injection of AAV9 was evaluated. AAV9 expressing feline β-galactosidase was intravenously administered at 1.5x1013 vector genomes/kilogram body weight to six GM1 cats at approximately 1 month of age. The animals were divided into two cohorts: 1) a long-term group, which was followed to humane endpoint, and 2) a short-term group, which was analyzed 16-weeks post treatment. Clinical assessments included neurological exams, cerebrospinal fluid and urine biomarkers, and 7-Telsa magnetic resonance imaging and spectroscopy. Postmortem analysis included β-galactosidase and virus distribution, histological analysis, and ganglioside content. Untreated GM1 animals survived 8.0 ± 0.6 months while intravenous treatment increased survival to an average of 3.5 years (n = 2) with substantial improvements in quality of life and neurologic function. Neurological abnormalities, which in untreated animals progress to the inability to stand and debilitating neurological disease by 8 months of age, were mild in all treated animals. Cerebrospinal fluid biomarkers were normalized, indicating decreased central nervous system cell damage in the treated animals. Urinary glycosaminoglycans decreased to normal levels in the long-term cohort. Magnetic resonance imaging and spectroscopy showed partial preservation of the brain in treated animals, which was supported by postmortem histological evaluation. β-galactosidase activity was increased throughout the central nervous system, reaching carrier levels in much of the cerebrum and normal levels in the cerebellum, spinal cord and cerebrospinal fluid. Ganglioside accumulation was significantly reduced by treatment. Peripheral tissues such as heart, skeletal muscle, and sciatic nerve also had normal β-galactosidase activity in treated GM1 cats. GM1 histopathology was largely corrected with treatment. There was no evidence of tumorigenesis or toxicity. Restoration of β-galactosidase activity in the central nervous system and peripheral organs by intravenous gene therapy led to profound increases in lifespan and quality of life in GM1 cats. This data supports the promise of intravenous gene therapy as a safe, effective treatment for GM1 gangliosidosis.
Collapse
Affiliation(s)
- Amanda L Gross
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849 USA.,Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, AL 36849 USA
| | - Heather L Gray-Edwards
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849 USA
| | - Cassie N Bebout
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849 USA
| | - Nathan L Ta
- Biology Department, Boston College, Chestnut Hill, MA 02467 USA
| | - Kayly Nielsen
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849 USA
| | - Brandon L Brunson
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, AL 36849 USA
| | - Kalajan R Lopez Mercado
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849 USA
| | - Devin E Osterhoudt
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849 USA
| | - Ana Rita Batista
- Department of Neurology, University of Massachusetts Medical School, Worcester MA 01605 USA.,Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester MA 01605 USA
| | - Stacy Maitland
- Department of Neurology, University of Massachusetts Medical School, Worcester MA 01605 USA.,Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester MA 01605 USA
| | | | - Miguel Sena-Esteves
- Department of Neurology, University of Massachusetts Medical School, Worcester MA 01605 USA.,Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester MA 01605 USA
| | - Douglas R Martin
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849 USA.,Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, AL 36849 USA
| |
Collapse
|
10
|
Piguet F, de Saint Denis T, Audouard E, Beccaria K, André A, Wurtz G, Schatz R, Alves S, Sevin C, Zerah M, Cartier N. The Challenge of Gene Therapy for Neurological Diseases: Strategies and Tools to Achieve Efficient Delivery to the Central Nervous System. Hum Gene Ther 2021; 32:349-374. [PMID: 33167739 DOI: 10.1089/hum.2020.105] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
For more than 10 years, gene therapy for neurological diseases has experienced intensive research growth and more recently therapeutic interventions for multiple indications. Beneficial results in several phase 1/2 clinical studies, together with improved vector technology have advanced gene therapy for the central nervous system (CNS) in a new era of development. Although most initial strategies have focused on orphan genetic diseases, such as lysosomal storage diseases, more complex and widespread conditions like Alzheimer's disease, Parkinson's disease, epilepsy, or chronic pain are increasingly targeted for gene therapy. Increasing numbers of applications and patients to be treated will require improvement and simplification of gene therapy protocols to make them accessible to the largest number of affected people. Although vectors and manufacturing are a major field of academic research and industrial development, there is a growing need to improve, standardize, and simplify delivery methods. Delivery is the major issue for CNS therapies in general, and particularly for gene therapy. The blood-brain barrier restricts the passage of vectors; strategies to bypass this obstacle are a central focus of research. In this study, we present the different ways that can be used to deliver gene therapy products to the CNS. We focus on results obtained in large animals that have allowed the transfer of protocols to human patients and have resulted in the generation of clinical data. We discuss the different routes of administration, their advantages, and their limitations. We describe techniques, equipment, and protocols and how they should be selected for safe delivery and improved efficiency for the next generation of gene therapy trials for CNS diseases.
Collapse
Affiliation(s)
- Françoise Piguet
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Timothée de Saint Denis
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Pediatric Neurosurgery, Hôpital Necker-Enfants Malades, APHP Centre. Université de Paris, Paris, France
| | - Emilie Audouard
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Kevin Beccaria
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Pediatric Neurosurgery, Hôpital Necker-Enfants Malades, APHP Centre. Université de Paris, Paris, France
| | - Arthur André
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Neurosurgery, Hôpitaux Universitaires La Pitié-Salpêtrière, Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | - Guillaume Wurtz
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Raphael Schatz
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Sandro Alves
- BrainVectis-Askbio France, iPeps Paris Brain Institute, Paris, France
| | - Caroline Sevin
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,BrainVectis-Askbio France, iPeps Paris Brain Institute, Paris, France.,APHP, Department of Neurology, Hopital le Kremlin Bicetre, Paris, France
| | - Michel Zerah
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Pediatric Neurosurgery, Hôpital Necker-Enfants Malades, APHP Centre. Université de Paris, Paris, France
| | - Nathalie Cartier
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
11
|
O'Carroll SJ, Cook WH, Young D. AAV Targeting of Glial Cell Types in the Central and Peripheral Nervous System and Relevance to Human Gene Therapy. Front Mol Neurosci 2021; 13:618020. [PMID: 33505247 PMCID: PMC7829478 DOI: 10.3389/fnmol.2020.618020] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Different glial cell types are found throughout the central (CNS) and peripheral nervous system (PNS), where they have important functions. These cell types are also involved in nervous system pathology, playing roles in neurodegenerative disease and following trauma in the brain and spinal cord (astrocytes, microglia, oligodendrocytes), nerve degeneration and development of pain in peripheral nerves (Schwann cells, satellite cells), retinal diseases (Müller glia) and gut dysbiosis (enteric glia). These cell type have all been proposed as potential targets for treating these conditions. One approach to target these cell types is the use of gene therapy to modify gene expression. Adeno-associated virus (AAV) vectors have been shown to be safe and effective in targeting cells in the nervous system and have been used in a number of clinical trials. To date, a number of studies have tested the use of different AAV serotypes and cell-specific promoters to increase glial cell tropism and expression. However, true glial-cell specific targeting for a particular glial cell type remains elusive. This review provides an overview of research into developing glial specific gene therapy and discusses some of the issues that still need to be addressed to make glial cell gene therapy a clinical reality.
Collapse
Affiliation(s)
- Simon J O'Carroll
- Spinal Cord Injury Research Group, Department of Anatomy and Medical Imaging, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - William H Cook
- Molecular Neurotherapeutics Group, Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Deborah Young
- Molecular Neurotherapeutics Group, Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
12
|
Ballon DJ, Rosenberg JB, Fung EK, Nikolopoulou A, Kothari P, De BP, He B, Chen A, Heier LA, Sondhi D, Kaminsky SM, Mozley PD, Babich JW, Crystal RG. Quantitative Whole-Body Imaging of I-124-Labeled Adeno-Associated Viral Vector Biodistribution in Nonhuman Primates. Hum Gene Ther 2020; 31:1237-1259. [PMID: 33233962 PMCID: PMC7769048 DOI: 10.1089/hum.2020.116] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/03/2020] [Indexed: 12/19/2022] Open
Abstract
A method is presented for quantitative analysis of the biodistribution of adeno-associated virus (AAV) gene transfer vectors following in vivo administration. We used iodine-124 (I-124) radiolabeling of the AAV capsid and positron emission tomography combined with compartmental modeling to quantify whole-body and organ-specific biodistribution of AAV capsids from 1 to 72 h following administration. Using intravenous (IV) and intracisternal (IC) routes of administration of AAVrh.10 and AAV9 vectors to nonhuman primates in the absence or presence of anticapsid immunity, we have identified novel insights into initial capsid biodistribution and organ-specific capsid half-life. Neither I-124-labeled AAVrh.10 nor AAV9 administered intravenously was detected at significant levels in the brain relative to the administered vector dose. Approximately 50% of the intravenously administered labeled capsids were dispersed throughout the body, independent of the liver, heart, and spleen. When administered by the IC route, the labeled capsid had a half-life of ∼10 h in the cerebral spinal fluid (CSF), suggesting that by this route, the CSF serves as a source with slow diffusion into the brain. For both IV and IC administration, there was significant influence of pre-existing anticapsid immunity on I-124-capsid biodistribution. The methodology facilitates quantitative in vivo viral vector dosimetry, which can serve as a technique for evaluation of both on- and off-target organ biodistribution, and potentially accelerate gene therapy development through rapid prototyping of novel vector designs.
Collapse
Affiliation(s)
- Douglas J. Ballon
- Department of Radiology, Citigroup Biomedical Imaging Center
- Department of Genetic Medicine
| | | | - Edward K. Fung
- Department of Radiology, Citigroup Biomedical Imaging Center
| | | | - Paresh Kothari
- Department of Radiology, Citigroup Biomedical Imaging Center
| | | | - Bin He
- Department of Radiology, Citigroup Biomedical Imaging Center
| | | | - Linda A. Heier
- Department of Radiology; Weill Cornell Medical College, New York, New York, USA
| | | | | | | | - John W. Babich
- Department of Radiology, Citigroup Biomedical Imaging Center
| | | |
Collapse
|
13
|
Nieuwenhuis B, Haenzi B, Hilton S, Carnicer-Lombarte A, Hobo B, Verhaagen J, Fawcett JW. Optimization of adeno-associated viral vector-mediated transduction of the corticospinal tract: comparison of four promoters. Gene Ther 2020; 28:56-74. [PMID: 32576975 PMCID: PMC7902269 DOI: 10.1038/s41434-020-0169-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/01/2020] [Accepted: 06/11/2020] [Indexed: 12/22/2022]
Abstract
Adeno-associated viral vectors are widely used as vehicles for gene transfer to the nervous system. The promoter and viral vector serotype are two key factors that determine the expression dynamics of the transgene. A previous comparative study has demonstrated that AAV1 displays efficient transduction of layer V corticospinal neurons, but the optimal promoter for transgene expression in corticospinal neurons has not been determined yet. In this paper, we report a side-by-side comparison between four commonly used promoters: the short CMV early enhancer/chicken β actin (sCAG), human cytomegalovirus (hCMV), mouse phosphoglycerate kinase (mPGK) and human synapsin (hSYN) promoter. Reporter constructs with each of these promoters were packaged in AAV1, and were injected in the sensorimotor cortex of rats and mice in order to transduce the corticospinal tract. Transgene expression levels and the cellular transduction profile were examined after 6 weeks. The AAV1 vectors harbouring the hCMV and sCAG promoters resulted in transgene expression in neurons, astrocytes and oligodendrocytes. The mPGK and hSYN promoters directed the strongest transgene expression. The mPGK promoter did drive expression in cortical neurons and oligodendrocytes, while transduction with AAV harbouring the hSYN promoter resulted in neuron-specific expression, including perineuronal net expressing interneurons and layer V corticospinal neurons. This promoter comparison study contributes to improve transgene delivery into the brain and spinal cord. The optimized transduction of the corticospinal tract will be beneficial for spinal cord injury research.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK. .,Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands.
| | - Barbara Haenzi
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
| | - Sam Hilton
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
| | - Alejandro Carnicer-Lombarte
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
| | - Barbara Hobo
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands.,Centre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| | - James W Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK.,Centre of Reconstructive Neuroscience, Institute of Experimental Medicine, Vídeňská 1083, 142 20, Prague 4, Czech Republic
| |
Collapse
|
14
|
Adachi K, Dissen GA, Lomniczi A, Xie Q, Ojeda SR, Nakai H. Adeno-associated virus-binding antibodies detected in cats living in the Northeastern United States lack neutralizing activity. Sci Rep 2020; 10:10073. [PMID: 32572045 PMCID: PMC7308316 DOI: 10.1038/s41598-020-66596-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/20/2020] [Indexed: 12/26/2022] Open
Abstract
Cats are a critical pre-clinical model for studying adeno-associated virus (AAV) vector-mediated gene therapies. A recent study has described the high prevalence of anti-AAV neutralizing antibodies among domestic cats in Switzerland. However, our knowledge of pre-existing humoral immunity against various AAV serotypes in cats is still limited. Here, we show that, although antibodies binding known AAV serotypes (AAV1 to AAV11) are prevalent in cats living in the Northeastern United States, these antibodies do not necessarily neutralize AAV infectivity. We analyzed sera from 35 client-owned, 20 feral, and 30 specific pathogen-free (SPF) cats for pre-existing AAV-binding antibodies against the 11 serotypes. Antibody prevalence was 7 to 90% with an overall median of 50%. The AAV-binding antibodies showed broad reactivities with other serotypes. Of 44 selected antibodies binding AAV2, AAV6 or AAV9, none exhibited appreciable neutralizing activities. Instead, AAV6 or AAV9-binding antibodies showed a transduction-enhancing effect. AAV6-binding antibodies were highly prevalent in SPF cats (83%), but this was primarily due to cross-reactivity with preventive vaccine-induced anti-feline panleukopenia virus antibodies. These results indicate that prevalent pre-existing immunity in cats is not necessarily inhibitory to AAV and highlight a substantial difference in the nature of AAV-binding antibodies in cats living in geographically different regions.
Collapse
Affiliation(s)
- Kei Adachi
- Department of Molecular & Medical Genetics, Oregon Health & Science University School of Medicine, Portland, Oregon, 97239, USA
| | - Gregory A Dissen
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, 97006, United States of America.,Molecular Virology Core, Oregon National Primate Research Center, Beaverton, Oregon, 97006, United States of America
| | - Alejandro Lomniczi
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, 97006, United States of America
| | - Qing Xie
- Department of Molecular & Medical Genetics, Oregon Health & Science University School of Medicine, Portland, Oregon, 97239, USA
| | - Sergio R Ojeda
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, 97006, United States of America
| | - Hiroyuki Nakai
- Department of Molecular & Medical Genetics, Oregon Health & Science University School of Medicine, Portland, Oregon, 97239, USA. .,Department of Molecular Microbiology & Immunology, Oregon Health & Science University School of Medicine, Portland, Oregon, 97239, USA. .,Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, 97006, United States of America.
| |
Collapse
|
15
|
O'Connor DM, Lutomski C, Jarrold MF, Boulis NM, Donsante A. Lot-to-Lot Variation in Adeno-Associated Virus Serotype 9 (AAV9) Preparations. Hum Gene Ther Methods 2019; 30:214-225. [PMID: 31752530 PMCID: PMC6919242 DOI: 10.1089/hgtb.2019.105] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Viral vectors are complex drugs that pose a particular challenge for manufacturing. Previous studies have shown that, unlike small-molecule drugs, vector preparations do not yield a collection of identical particles. Instead, a mixture of particles that vary in capsid stoichiometry and impurities is created, which may differ from lot to lot. The consequences of this are unclear, but conflicting reports regarding the biological properties of vectors, including transduction patterns, suggest that this variability may have an effect. However, other variables, including differences in animal strains and techniques, make it difficult to identify a cause. Here, we report lot-to-lot variation in spinal cord gray matter transduction following intrathecal delivery of self-complementary adeno-associated virus serotype 9 vectors. Eleven lots of vector were evaluated from six vector cores, including one preclinical/Good Laboratory Practice lot. Eight of the lots, including the preclinical lot, failed to transduce the gray matter, whereas the other three provided robust transduction. The cause for this variation is unknown, but it did not correlate with vector titer, buffer, or purification method. These results highlight the need to identify the cause of this variation and to develop improved production and quality control methods to ensure lot-to-lot consistency of vector potency.
Collapse
Affiliation(s)
- Deirdre M. O'Connor
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia
| | | | | | - Nicholas M. Boulis
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia
| | - Anthony Donsante
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia;,Correspondence: Dr. Anthony Donsante, Department of Neurosurgery, Emory University School of Medicine, 101 Woodruff Circle, Room 6339, Atlanta, GA 30322.
| |
Collapse
|
16
|
Giorgio E, Lorenzati M, Rivetti di Val Cervo P, Brussino A, Cernigoj M, Della Sala E, Bartoletti Stella A, Ferrero M, Caiazzo M, Capellari S, Cortelli P, Conti L, Cattaneo E, Buffo A, Brusco A. Allele-specific silencing as treatment for gene duplication disorders: proof-of-principle in autosomal dominant leukodystrophy. Brain 2019; 142:1905-1920. [PMID: 31143934 DOI: 10.1093/brain/awz139] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 01/16/2019] [Accepted: 03/31/2019] [Indexed: 11/14/2022] Open
Abstract
Allele-specific silencing by RNA interference (ASP-siRNA) holds promise as a therapeutic strategy for downregulating a single mutant allele with minimal suppression of the corresponding wild-type allele. This approach has been effectively used to target autosomal dominant mutations and single nucleotide polymorphisms linked with aberrantly expanded trinucleotide repeats. Here, we propose ASP-siRNA as a preferable choice to target duplicated disease genes, avoiding potentially harmful excessive downregulation. As a proof-of-concept, we studied autosomal dominant adult-onset demyelinating leukodystrophy (ADLD) due to lamin B1 (LMNB1) duplication, a hereditary, progressive and fatal disorder affecting myelin in the CNS. Using a reporter system, we screened the most efficient ASP-siRNAs preferentially targeting one of the alleles at rs1051644 (average minor allele frequency: 0.45) located in the 3' untranslated region of the gene. We identified four siRNAs with a high efficacy and allele-specificity, which were tested in ADLD patient-derived fibroblasts. Three of the small interfering RNAs were highly selective for the target allele and restored both LMNB1 mRNA and protein levels close to control levels. Furthermore, small interfering RNA treatment abrogates the ADLD-specific phenotypes in fibroblasts and in two disease-relevant cellular models: murine oligodendrocytes overexpressing human LMNB1, and neurons directly reprogrammed from patients' fibroblasts. In conclusion, we demonstrated that ASP-silencing by RNA interference is a suitable and promising therapeutic option for ADLD. Moreover, our results have a broad translational value extending to several pathological conditions linked to gene-gain in copy number variations.
Collapse
Affiliation(s)
- Elisa Giorgio
- University of Torino, Department of Medical Sciences, Torino, Italy
| | - Martina Lorenzati
- University of Torino, Department of Neuroscience Rita Levi Montalcini and Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy
| | - Pia Rivetti di Val Cervo
- University of Milan, Department of Biosciences, Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Milan, Italy
| | | | - Manuel Cernigoj
- University of Milan, Department of Biosciences, Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Milan, Italy
| | | | | | - Marta Ferrero
- University of Torino, Department of Medical Sciences, Torino, Italy
| | - Massimiliano Caiazzo
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, CG, Utrecht, The Netherlands
- Department of Molecular Medicine and Medical Biotechnology, University of Naples 'Federico II', Naples, Italy
| | - Sabina Capellari
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
- University of Bologna, Department of Biomedical and Neuromotor Sciences, Bologna, Italy
| | - Pietro Cortelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
- University of Bologna, Department of Biomedical and Neuromotor Sciences, Bologna, Italy
| | - Luciano Conti
- University of Trento, Centre for Integrative Biology (CIBIO), Laboratory of Computational Oncology, Trento, Italy
| | - Elena Cattaneo
- University of Milan, Department of Biosciences, Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Milan, Italy
- National Institute of Molecular Genetics (INGM) Romeo and Enrica Invernizzi, Milano, Italy
| | - Annalisa Buffo
- University of Torino, Department of Neuroscience Rita Levi Montalcini and Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy
| | - Alfredo Brusco
- University of Torino, Department of Medical Sciences, Torino, Italy
- Città della Salute e della Scienza University Hospital, Medical Genetics Unit, Torino, Italy
| |
Collapse
|
17
|
Anderson HE, Schaller KL, Caldwell JH, Weir RFF. Intravascular injections of adenoassociated viral vector serotypes rh10 and PHP.B transduce murine sciatic nerve axons. Neurosci Lett 2019; 706:51-55. [PMID: 31078676 DOI: 10.1016/j.neulet.2019.05.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 02/03/2023]
Abstract
Adenoassociated viral vectors provide a safe and robust method for expression of transgenes in nondividing cells such as neurons. Intravenous injections of these vectors provide a means of transducing motoneurons of peripheral nerves. Previous research has demonstrated that serotypes 1, rh10 and PHP.B can transduce motor neuron cell bodies in the spinal cord, but has not quantified expression in the peripheral nerve axon. Axonal labeling is crucial for optogenetic stimulation and detection of action potentials in peripheral nerve. Therefore, in this study, serotypes 1, PHP.B, and rh10 were tested for their ability to label axons of the murine sciatic and tibial nerve following intravenous injection. Serotype rh10 elicits expression in 10% of acetylcholine transferase positive axons of the sciatic nerve in immunohistochemically-stained sections. Serotype rh10 transduces a variety of axon diameters from <1-12 μm, while PHP.B transduces larger axons of diameter (4-16 μm). Expression was not seen with serotype 1. These results show the potential of serotypes PHP.B and rh10 delivery of transgenic products to axons of the peripheral nerve.
Collapse
Affiliation(s)
- Hans E Anderson
- Department of Bioengineering, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA.
| | - Kristin L Schaller
- Department of Neurology, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - John H Caldwell
- Department of Cell and Developmental Biology, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Richard F Ff Weir
- Department of Bioengineering, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
18
|
Castle MJ, Cheng Y, Asokan A, Tuszynski MH. Physical positioning markedly enhances brain transduction after intrathecal AAV9 infusion. SCIENCE ADVANCES 2018; 4:eaau9859. [PMID: 30443600 PMCID: PMC6235539 DOI: 10.1126/sciadv.aau9859] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/15/2018] [Indexed: 05/10/2023]
Abstract
Several neurological disorders may benefit from gene therapy. However, even when using the lead vector candidate for intrathecal administration, adeno-associated virus serotype 9 (AAV9), the strength and distribution of gene transfer to the brain are inconsistent. On the basis of preliminary observations that standard intrathecal AAV9 infusions predominantly drive reporter gene expression in brain regions where gravity might cause cerebrospinal fluid to settle, we tested the hypothesis that counteracting vector "settling" through animal positioning would enhance vector delivery to the brain. When rats are either inverted in the Trendelenburg position or continuously rotated after intrathecal AAV9 infusion, we find (i) a significant 15-fold increase in the number of transduced neurons, (ii) a marked increase in gene delivery to cortical regions, and (iii) superior animal-to-animal consistency of gene expression. Entorhinal, prefrontal, frontal, parietal, hippocampal, limbic, and basal forebrain neurons are extensively transduced: 95% of transduced cells are neurons, and greater than 70% are excitatory. These findings provide a novel and simple method for broad gene delivery to the cortex and are of substantial relevance to translational programs for neurological disorders, including Alzheimer's disease and related dementias, stroke, and traumatic brain injury.
Collapse
Affiliation(s)
- Michael J. Castle
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yuhsiang Cheng
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Aravind Asokan
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark H. Tuszynski
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
- Veterans Administration Medical Center, San Diego, CA 92161, USA
| |
Collapse
|
19
|
Wang D, Li J, Tran K, Burt DR, Zhong L, Gao G. Slow Infusion of Recombinant Adeno-Associated Viruses into the Mouse Cerebrospinal Fluid Space. Hum Gene Ther Methods 2018; 29:75-85. [PMID: 29596011 DOI: 10.1089/hgtb.2017.250] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Recombinant adeno-associated viruses (rAAVs) are the leading in vivo gene delivery platform, and have been extensively studied in gene therapy targeting various tissues, including the central nervous system (CNS). A single-bolus rAAV injection to the cerebrospinal fluid (CSF) space has been widely used to target the CNS, but it suffers from several drawbacks, such as leakage to peripheral tissues. Here, a protocol is described using an osmotic pump to infuse rAAV slowly into the mouse CSF space. Compared to the single-bolus injection technique, pump infusion can lead to higher CNS transduction and lower transduction in the peripheral tissues.
Collapse
Affiliation(s)
- Dan Wang
- 1 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School , Worcester, Massachusetts.,3 Department of Microbiology and Physiological Systems, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Jia Li
- 1 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Karen Tran
- 1 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Daniel R Burt
- 1 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Li Zhong
- 1 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School , Worcester, Massachusetts.,3 Department of Microbiology and Physiological Systems, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Guangping Gao
- 1 Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School , Worcester, Massachusetts.,3 Department of Microbiology and Physiological Systems, University of Massachusetts Medical School , Worcester, Massachusetts.,4 Viral Vector Core, University of Massachusetts Medical School , Worcester, Massachusetts.,5 West China Hospital, Sichuan University , Chengdu, China
| |
Collapse
|
20
|
Rosenberg JB, Kaplitt MG, De BP, Chen A, Flagiello T, Salami C, Pey E, Zhao L, Ricart Arbona RJ, Monette S, Dyke JP, Ballon DJ, Kaminsky SM, Sondhi D, Petsko GA, Paul SM, Crystal RG. AAVrh.10-Mediated APOE2 Central Nervous System Gene Therapy for APOE4-Associated Alzheimer's Disease. HUM GENE THER CL DEV 2018; 29:24-47. [PMID: 29409358 DOI: 10.1089/humc.2017.231] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive degenerative neurological disorder affecting nearly one in nine elderly people in the United States. Population studies have shown that an inheritance of the apolipoprotein E (APOE) variant APOE4 allele increases the risk of developing AD, whereas APOE2 homozygotes are protected from late-onset AD. It was hypothesized that expression of the "protective" APOE2 variant by genetic modification of the central nervous system (CNS) of APOE4 homozygotes could reverse or prevent progressive neurologic damage. To assess the CNS distribution and safety of APOE2 gene therapy for AD in a large-animal model, intraparenchymal, intracisternal, and intraventricular routes of delivery to the CNS of nonhuman primates of AAVrh.10hAPOE2-HA, an AAVrh.10 serotype coding for an HA-tagged human APOE2 cDNA sequence, were evaluated. To evaluate the route of delivery that achieves the widest extent of APOE2 expression in the CNS, the expression of APOE2 in the CNS was evaluated 2 months following vector administration for APOE2 DNA, mRNA, and protein. Finally, using conventional toxicology assays, the safety of the best route of delivery was assessed. The data demonstrated that while all three routes are capable of mediating ApoE2 expression in AD relevant regions, intracisternal delivery of AAVrh.10hAPOE2-HA safely mediated wide distribution of ApoE2 with the least invasive surgical intervention, thus providing the optimal strategy to deliver vector-mediated human APOE2 to the CNS.
Collapse
Affiliation(s)
- Jonathan B Rosenberg
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Michael G Kaplitt
- 2 Department of Neurosurgery, Weill Cornell Medical College , New York, New York
| | - Bishnu P De
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Alvin Chen
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Thomas Flagiello
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Christiana Salami
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Eduard Pey
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Lingzhi Zhao
- 3 Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College , New York, New York
| | - Rodolfo J Ricart Arbona
- Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sebastien Monette
- Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, The Rockefeller University , Weill Cornell Medical College, New York, New York
| | - Jonathan P Dyke
- 6 Department of Radiology, Weill Cornell Medical College , New York, New York
| | - Douglas J Ballon
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York.,6 Department of Radiology, Weill Cornell Medical College , New York, New York
| | - Stephen M Kaminsky
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Dolan Sondhi
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Gregory A Petsko
- 3 Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College , New York, New York
| | - Steven M Paul
- 7 Voyager Therapeutics, Inc. , Cambridge, Massachusetts
| | - Ronald G Crystal
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| |
Collapse
|
21
|
Sawamoto K, Chen HH, Alméciga-Díaz CJ, Mason RW, Tomatsu S. Gene therapy for Mucopolysaccharidoses. Mol Genet Metab 2018; 123:59-68. [PMID: 29295764 PMCID: PMC5986190 DOI: 10.1016/j.ymgme.2017.12.434] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 12/19/2022]
Abstract
Mucopolysaccharidoses (MPS) are a group of lysosomal storage disorders (LSDs) caused by a deficiency of lysosomal enzymes, leading to a wide range of various clinical symptoms depending upon the type of MPS or its severity. Enzyme replacement therapy (ERT), hematopoietic stem cell transplantation (HSCT), substrate reduction therapy (SRT), and various surgical procedures are currently available for patients with MPS. However, there is no curative treatment for this group of disorders. Gene therapy should be a one-time permanent therapy, repairing the cause of enzyme deficiency. Preclinical studies of gene therapy for MPS have been developed over the past three decades. Currently, clinical trials of gene therapy for some types of MPS are ongoing in the United States, some European countries, and Australia. Here, in this review, we summarize the development of gene therapy for MPS in preclinical and clinical trials.
Collapse
Affiliation(s)
- Kazuki Sawamoto
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Hui-Hsuan Chen
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States; Department of Medical Laboratory Sciences, University of Delaware, Newark, DE, United States
| | - Carlos J Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Robert W Mason
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States; Department of Pediatrics, Gifu University, Gifu, Japan; Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
22
|
Hardcastle N, Boulis NM, Federici T. AAV gene delivery to the spinal cord: serotypes, methods, candidate diseases, and clinical trials. Expert Opin Biol Ther 2017; 18:293-307. [PMID: 29249183 DOI: 10.1080/14712598.2018.1416089] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Adeno-associated viral (AAV) vector-mediated gene delivery to the spinal cord has finally entered the pathway towards regulatory approval. Phase 1 clinical trials using AAV gene therapy for pediatric disorders - spinal muscular atrophy (SMA) and giant axonal neuropathy (GAN) - are now underway. AREAS COVERED This review addresses the latest progress in the field of AAV gene delivery to the spinal cord, particularly focusing on the most prominent AAV serotypes and delivery methodologies to the spinal cord. Candidate diseases and scaling up experiments in large animals are also discussed. EXPERT OPINION Intravenous (IV) and intrathecal (IT) deliveries seem to undoubtedly be the preferred routes of administration for diffuse spinal cord delivery of therapeutic AAV vectors that can cross the blood-brain barrier (BBB) and correct inherited genetic disorders. Conversely, intraparenchymal delivery is still an undervalued but very viable approach for segmental therapy in afflictions such as ALS or Pompe Disease as a means to prevent respiratory dysfunction.
Collapse
Affiliation(s)
- Nathan Hardcastle
- a Department of Neurosurgery , Emory University , Atlanta , GA , USA
| | - Nicholas M Boulis
- a Department of Neurosurgery , Emory University , Atlanta , GA , USA
| | - Thais Federici
- a Department of Neurosurgery , Emory University , Atlanta , GA , USA
| |
Collapse
|
23
|
Hinderer C, Bell P, Katz N, Vite CH, Louboutin JP, Bote E, Yu H, Zhu Y, Casal ML, Bagel J, O'Donnell P, Wang P, Haskins ME, Goode T, Wilson JM. Evaluation of Intrathecal Routes of Administration for Adeno-Associated Viral Vectors in Large Animals. Hum Gene Ther 2017; 29:15-24. [PMID: 28806897 DOI: 10.1089/hum.2017.026] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Delivery of adeno-associated viral (AAV) vectors into the cerebrospinal fluid (CSF) can achieve gene transfer to cells throughout the brain and spinal cord, potentially making many neurological diseases tractable gene therapy targets. Identifying the optimal route of CSF access for intrathecal AAV delivery will be a critical step in translating this approach to clinical practice. We previously demonstrated that vector injection into the cisterna magna is a safe and effective method for intrathecal AAV delivery in nonhuman primates; however, this procedure is not commonly used in clinical practice. More routine methods of administration into the CSF are now being explored, including intracerebroventricular (ICV) injection and injection through a lumbar puncture. In this study, we compared ICV and intracisternal (IC) AAV administration in dogs. We also evaluated vector administration via lumbar puncture in nonhuman primates, with some animals placed in the Trendelenburg position after injection, a maneuver that has been suggested to improve cranial distribution of vector. In the dog study, ICV and IC vector administration resulted in similarly efficient transduction throughout the brain and spinal cord. However, animals in the ICV cohort developed encephalitis associated with a T-cell response to the transgene product, a phenomenon that was not observed in the IC cohort. In the nonhuman primate study, transduction efficiency was not improved by placing animals in the Trendelenburg position after injection. These findings illustrate important limitations of commonly used methods for CSF access in the context of AAV delivery, and will be important for informing the selection of a route of administration for first-in-human studies.
Collapse
Affiliation(s)
- Christian Hinderer
- 1 Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Peter Bell
- 1 Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nathan Katz
- 1 Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Charles H Vite
- 2 Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jean-Pierre Louboutin
- 3 Section of Anatomy, Department of Basic Medical Sciences, University of the West Indies , Kingston, Jamaica
| | - Erin Bote
- 1 Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hongwei Yu
- 1 Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yanqing Zhu
- 1 Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Margret L Casal
- 2 Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jessica Bagel
- 2 Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Patricia O'Donnell
- 2 Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ping Wang
- 2 Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mark E Haskins
- 2 Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tamara Goode
- 1 Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - James M Wilson
- 1 Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
24
|
Hordeaux J, Dubreil L, Robveille C, Deniaud J, Pascal Q, Dequéant B, Pailloux J, Lagalice L, Ledevin M, Babarit C, Costiou P, Jamme F, Fusellier M, Mallem Y, Ciron C, Huchet C, Caillaud C, Colle MA. Long-term neurologic and cardiac correction by intrathecal gene therapy in Pompe disease. Acta Neuropathol Commun 2017; 5:66. [PMID: 28874182 PMCID: PMC5585940 DOI: 10.1186/s40478-017-0464-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/07/2017] [Indexed: 11/10/2022] Open
Abstract
Pompe disease is a lysosomal storage disorder caused by acid-α-glucosidase (GAA) deficiency, leading to glycogen storage. The disease manifests as a fatal cardiomyopathy in infantile form. Enzyme replacement therapy (ERT) has recently prolonged the lifespan of these patients, revealing a new natural history. The neurologic phenotype and the persistence of selective muscular weakness in some patients could be attributed to the central nervous system (CNS) storage uncorrected by ERT. GAA-KO 6neo/6neo mice were treated with a single intrathecal administration of adeno-associated recombinant vector (AAV) mediated gene transfer of human GAA at 1 month and their neurologic, neuromuscular, and cardiac function was assessed for 1 year. We demonstrate a significant functional neurologic correction in treated animals from 4 months onward, a neuromuscular improvement from 9 months onward, and a correction of the hypertrophic cardiomyopathy at 12 months. The regions most affected by the disease i.e. the brainstem, spinal cord, and the left cardiac ventricular wall all show enzymatic, biochemical and histological correction. Muscle glycogen storage is not affected by the treatment, thus suggesting that the restoration of muscle functionality is directly related to the CNS correction. This unprecedented global and long-term CNS and cardiac cure offer new perspectives for the management of patients.
Collapse
|
25
|
Christie-Brown V, Mitchell J, Talbot K. The SMA Trust: the role of a disease-focused research charity in developing treatments for SMA. Gene Ther 2017; 24:544-546. [PMID: 28561814 DOI: 10.1038/gt.2017.47] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 02/04/2023]
Abstract
SMA is a rare hereditary neuromuscular disease that causes weakness and muscle wasting as a result of the loss of spinal motor neurons. In its most severe form, SMA is the commonest genetic cause of death in infants, and children with less severe forms of SMA face the prospect of lifelong disability from progressive muscle wasting, loss of mobility and limb weakness. The initial discovery of the defective gene has been followed by major advances in our understanding of the genetic, cellular and molecular basis of SMA, providing the foundation for a range of approaches to treatment, including gene therapy, antisense oligonucleotide treatments and more traditional drug-based approaches to slow or halt disease progression. The approval by the US Food and Drug Administration (FDA) of Spinraza (nusinersen), the first targeted treatment for spinal muscular atrophy (SMA), is a historic moment. Disease-focused research charities, such as The SMA Trust (UK), continue to have a crucial role in promoting the development of additional treatments for SMA, both by funding translational research and by promoting links between researchers, people living with SMA and other stakeholders, including pharmaceutical companies and healthcare providers.
Collapse
|
26
|
Bey K, Ciron C, Dubreil L, Deniaud J, Ledevin M, Cristini J, Blouin V, Aubourg P, Colle MA. Efficient CNS targeting in adult mice by intrathecal infusion of single-stranded AAV9-GFP for gene therapy of neurological disorders. Gene Ther 2017; 24:325-332. [PMID: 28425480 DOI: 10.1038/gt.2017.18] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/17/2017] [Accepted: 02/28/2017] [Indexed: 12/19/2022]
Abstract
Adeno-associated virus (AAV) gene therapy constitutes a powerful tool for the treatment of neurodegenerative diseases. While AAVs are generally administered systemically to newborns in preclinical studies of neurological disorders, in adults the maturity of the blood-brain barrier (BBB) must be considered when selecting the route of administration. Delivery of AAVs into the cerebrospinal fluid (CSF) represents an attractive approach to target the central nervous system (CNS) and bypass the BBB. In this study, we investigated the efficacy of intra-CSF delivery of a single-stranded (ss) AAV9-CAG-GFP vector in adult mice via intracisternal (iCist) or intralumbar (it-Lumb) administration. It-Lumb ssAAV9 delivery resulted in greater diffusion throughout the entire spinal cord and green fluorescent protein (GFP) expression mainly in the cerebellum, cortex and olfactory bulb. By contrast, iCist delivery led to strong GFP expression throughout the entire brain. Comparison of the transduction efficiency of ssAAV9-CAG-GFP versus ssAAV9-SYN1-GFP following it-Lumb administration revealed widespread and specific GFP expression in neurons and motoneurons of the spinal cord and brain when the neuron-specific synapsin 1 (SYN1) promoter was used. Our findings demonstrate that it-Lumb ssAAV9 delivery is a safe and highly efficient means of targeting the CNS in adult mice.
Collapse
Affiliation(s)
- K Bey
- INRA/ONIRIS UMR U703, Animal Pathophysiology and Biotherapy for Muscle and Nervous System Diseases, Nantes, France.,Atlantic Gene Therapies, Nantes, France.,ONIRIS, CS 40706, Nantes-Atlantic National College of Veterinary Medicine, Food Science and Engineering, Bretagne Loire University (UBL), Nantes, France
| | - C Ciron
- INRA/ONIRIS UMR U703, Animal Pathophysiology and Biotherapy for Muscle and Nervous System Diseases, Nantes, France.,Atlantic Gene Therapies, Nantes, France.,ONIRIS, CS 40706, Nantes-Atlantic National College of Veterinary Medicine, Food Science and Engineering, Bretagne Loire University (UBL), Nantes, France
| | - L Dubreil
- INRA/ONIRIS UMR U703, Animal Pathophysiology and Biotherapy for Muscle and Nervous System Diseases, Nantes, France.,Atlantic Gene Therapies, Nantes, France.,ONIRIS, CS 40706, Nantes-Atlantic National College of Veterinary Medicine, Food Science and Engineering, Bretagne Loire University (UBL), Nantes, France
| | - J Deniaud
- INRA/ONIRIS UMR U703, Animal Pathophysiology and Biotherapy for Muscle and Nervous System Diseases, Nantes, France.,Atlantic Gene Therapies, Nantes, France.,ONIRIS, CS 40706, Nantes-Atlantic National College of Veterinary Medicine, Food Science and Engineering, Bretagne Loire University (UBL), Nantes, France
| | - M Ledevin
- INRA/ONIRIS UMR U703, Animal Pathophysiology and Biotherapy for Muscle and Nervous System Diseases, Nantes, France.,Atlantic Gene Therapies, Nantes, France.,ONIRIS, CS 40706, Nantes-Atlantic National College of Veterinary Medicine, Food Science and Engineering, Bretagne Loire University (UBL), Nantes, France
| | - J Cristini
- Department of Neurosurgery, Nantes Hospital, Nantes, France
| | - V Blouin
- INSERM UMR 1089, Atlantic Gene Therapies, Nantes, France
| | - P Aubourg
- INSERM U1169, Thérapie Génique, Génétique, Epigénétique en Neurologie, Endocrinologie et Développement de l'Enfant, Université Paris Sud, CEA, Le Kremlin Bicêtre, France
| | - M-A Colle
- INRA/ONIRIS UMR U703, Animal Pathophysiology and Biotherapy for Muscle and Nervous System Diseases, Nantes, France.,Atlantic Gene Therapies, Nantes, France.,ONIRIS, CS 40706, Nantes-Atlantic National College of Veterinary Medicine, Food Science and Engineering, Bretagne Loire University (UBL), Nantes, France
| |
Collapse
|
27
|
Saraiva J, Nobre RJ, Pereira de Almeida L. Gene therapy for the CNS using AAVs: The impact of systemic delivery by AAV9. J Control Release 2016; 241:94-109. [PMID: 27637390 DOI: 10.1016/j.jconrel.2016.09.011] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 09/09/2016] [Accepted: 09/12/2016] [Indexed: 12/15/2022]
Abstract
Several attempts have been made to discover the ideal vector for gene therapy in central nervous system (CNS). Adeno-associated viruses (AAVs) are currently the preferred vehicle since they exhibit stable transgene expression in post-mitotic cells, neuronal tropism, low risk of insertional mutagenesis and diminished immune responses. Additionally, the discovery that a particular serotype, AAV9, bypasses the blood-brain barrier has raised the possibility of intravascular administration as a non-invasive delivery route to achieve widespread CNS gene expression. AAV9 intravenous delivery has already shown promising results for several diseases in animal models, including lysosomal storage disorders and motor neuron diseases, opening the way to the first clinical trial in the field. This review presents an overview of clinical trials for CNS disorders using AAVs and will focus on preclinical studies based on the systemic gene delivery using AAV9.
Collapse
Affiliation(s)
- Joana Saraiva
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Rui Jorge Nobre
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Portugal
| | - Luis Pereira de Almeida
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Portugal.
| |
Collapse
|
28
|
Motas S, Haurigot V, Garcia M, Marcó S, Ribera A, Roca C, Sánchez X, Sánchez V, Molas M, Bertolin J, Maggioni L, León X, Ruberte J, Bosch F. CNS-directed gene therapy for the treatment of neurologic and somatic mucopolysaccharidosis type II (Hunter syndrome). JCI Insight 2016; 1:e86696. [PMID: 27699273 DOI: 10.1172/jci.insight.86696] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mucopolysaccharidosis type II (MPSII) is an X-linked lysosomal storage disease characterized by severe neurologic and somatic disease caused by deficiency of iduronate-2-sulfatase (IDS), an enzyme that catabolizes the glycosaminoglycans heparan and dermatan sulphate. Intravenous enzyme replacement therapy (ERT) currently constitutes the only approved therapeutic option for MPSII. However, the inability of recombinant IDS to efficiently cross the blood-brain barrier (BBB) limits ERT efficacy in treating neurological symptoms. Here, we report a gene therapy approach for MPSII through direct delivery of vectors to the CNS. Through a minimally invasive procedure, we administered adeno-associated virus vectors encoding IDS (AAV9-Ids) to the cerebrospinal fluid of MPSII mice with already established disease. Treated mice showed a significant increase in IDS activity throughout the encephalon, with full resolution of lysosomal storage lesions, reversal of lysosomal dysfunction, normalization of brain transcriptomic signature, and disappearance of neuroinflammation. Moreover, our vector also transduced the liver, providing a peripheral source of therapeutic protein that corrected storage pathology in visceral organs, with evidence of cross-correction of nontransduced organs by circulating enzyme. Importantly, AAV9-Ids-treated MPSII mice showed normalization of behavioral deficits and considerably prolonged survival. These results provide a strong proof of concept for the clinical translation of our approach for the treatment of Hunter syndrome patients with cognitive impairment.
Collapse
Affiliation(s)
- Sandra Motas
- Center of Animal Biotechnology and Gene Therapy and.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Virginia Haurigot
- Center of Animal Biotechnology and Gene Therapy and.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Miguel Garcia
- Center of Animal Biotechnology and Gene Therapy and.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Sara Marcó
- Center of Animal Biotechnology and Gene Therapy and.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Albert Ribera
- Center of Animal Biotechnology and Gene Therapy and.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Carles Roca
- Center of Animal Biotechnology and Gene Therapy and.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Xavier Sánchez
- Center of Animal Biotechnology and Gene Therapy and.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Víctor Sánchez
- Center of Animal Biotechnology and Gene Therapy and.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Maria Molas
- Center of Animal Biotechnology and Gene Therapy and.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Joan Bertolin
- Center of Animal Biotechnology and Gene Therapy and.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Luca Maggioni
- Center of Animal Biotechnology and Gene Therapy and.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Xavier León
- Center of Animal Biotechnology and Gene Therapy and.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| | - Jesús Ruberte
- Center of Animal Biotechnology and Gene Therapy and.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain.,Department of Animal Health and Anatomy, School of Veterinary Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy and.,Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Madrid, Spain
| |
Collapse
|
29
|
Gurda BL, De Guilhem De Lataillade A, Bell P, Zhu Y, Yu H, Wang P, Bagel J, Vite CH, Sikora T, Hinderer C, Calcedo R, Yox AD, Steet RA, Ruane T, O'Donnell P, Gao G, Wilson JM, Casal M, Ponder KP, Haskins ME. Evaluation of AAV-mediated Gene Therapy for Central Nervous System Disease in Canine Mucopolysaccharidosis VII. Mol Ther 2016; 24:206-216. [PMID: 26447927 PMCID: PMC4817811 DOI: 10.1038/mt.2015.189] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/22/2015] [Indexed: 12/11/2022] Open
Abstract
Mucopolysaccharidosis VII (MPS VII) is a lysosomal storage disease arising from mutations in β-d-glucuronidase (GUSB), which results in glycosaminoglycan (GAG) accumulation and a variety of clinical manifestations including neurological disease. Herein, MPS VII dogs were injected intravenously (i.v.) and/or intrathecally (i.t.) via the cisterna magna with AAV9 or AAVrh10 vectors carrying the canine GUSB cDNA. Although i.v. injection alone at 3 days of age resulted in normal cerebrospinal fluid (CSF) GUSB activity, brain tissue homogenates had only ~1 to 6% normal GUSB activity and continued to have elevated GAG storage. In contrast, i.t. injection at 3 weeks of age resulted in CSF GUSB activity 44-fold normal while brain tissue homogenates had >100% normal GUSB activity and reduced GAGs compared with untreated dogs. Markers for secondary storage and inflammation were eliminated in i.t.-treated dogs and reduced in i.v.-treated dogs compared with untreated dogs. Given that i.t.-treated dogs expressed higher levels of GUSB in the CNS tissues compared to those treated i.v., we conclude that i.t. injection of AAV9 or AAVrh10 vectors is more effective than i.v. injection alone in the large animal model of MPS VII.
Collapse
Affiliation(s)
- Brittney L Gurda
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | | | - Peter Bell
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yanqing Zhu
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Hongwei Yu
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ping Wang
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jessica Bagel
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Charles H Vite
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tracey Sikora
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Medicine, Division of Translational Medicine and Human Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Christian Hinderer
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Roberto Calcedo
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Alexander D Yox
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Richard A Steet
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Therese Ruane
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Patricia O'Donnell
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Guangping Gao
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA; Department of Microbiology and Physiology Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - James M Wilson
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Margret Casal
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katherine P Ponder
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mark E Haskins
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
30
|
Yoon SY, Bagel JH, O'Donnell PA, Vite CH, Wolfe JH. Clinical Improvement of Alpha-mannosidosis Cat Following a Single Cisterna Magna Infusion of AAV1. Mol Ther 2015; 24:26-33. [PMID: 26354342 DOI: 10.1038/mt.2015.168] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/01/2015] [Indexed: 11/09/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are debilitating neurometabolic disorders for most of which long-term effective therapies have not been developed. Gene therapy is a potential treatment but a critical barrier to treating the brain is the need for global correction. We tested the efficacy of cisterna magna infusion of adeno-associated virus type 1 (AAV1) expressing feline alpha-mannosidase gene in the postsymptomatic alpha-mannosidosis (AMD) cat, a homologue of the human disease. Lysosomal alpha-mannosidase (MANB) activity in the cerebrospinal fluid (CSF) and serum were increased above the control values in untreated AMD cats. Clinical neurological signs were delayed in onset and reduced in severity. The lifespan of the treated cats was significantly extended. Postmortem histopathology showed resolution of lysosomal storage lesions throughout the brain. MANB activity in brain tissue was significantly above the levels of untreated tissues. The results demonstrate that a single cisterna magna injection of AAV1 into the CSF can mediate widespread neuronal transduction of the brain and meaningful clinical improvement. Thus, cisterna magna gene delivery by AAV1 appears to be a viable strategy for treatment of the whole brain in AMD and should be applicable to many of the neurotropic LSDs as well as other neurogenetic disorders.
Collapse
Affiliation(s)
- Sea Young Yoon
- Research Institute of the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jessica H Bagel
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Patricia A O'Donnell
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Charles H Vite
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John H Wolfe
- Research Institute of the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
31
|
Chen H. Adeno-associated virus vectors for human gene therapy. World J Med Genet 2015; 5:28-45. [DOI: 10.5496/wjmg.v5.i3.28] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 04/08/2015] [Accepted: 05/18/2015] [Indexed: 02/06/2023] Open
Abstract
Adeno-associated virus (AAV) is a small, non-enveloped virus that contains a single-stranded DNA genome. It was the first gene therapy drug approved in the Western world in November 2012 to treat patients with lipoprotein lipase deficiency. AAV made history and put human gene therapy in the forefront again. More than four decades of research on AAV vector biology and human gene therapy has generated a huge amount of valuable information. Over 100 AAV serotypes and variants have been isolated and at least partially characterized. A number of them have been used for preclinical studies in a variety of animal models. Several AAV vector production platforms, especially the baculovirus-based system have been established for commercial-scale AAV vector production. AAV purification technologies such as density gradient centrifugation, column chromatography, or a combination, have been well developed. More than 117 clinical trials have been conducted with AAV vectors. Although there are still challenges down the road, such as cross-species variation in vector tissue tropism and gene transfer efficiency, pre-existing humoral immunity to AAV capsids and vector dose-dependent toxicity in patients, the gene therapy community is forging ahead with cautious optimism. In this review I will focus on the properties and applications of commonly used AAV serotypes and variants, and the technologies for AAV vector production and purification. I will also discuss the advancement of several promising gene therapy clinical trials.
Collapse
|
32
|
Tanguy Y, Biferi MG, Besse A, Astord S, Cohen-Tannoudji M, Marais T, Barkats M. Systemic AAVrh10 provides higher transgene expression than AAV9 in the brain and the spinal cord of neonatal mice. Front Mol Neurosci 2015; 8:36. [PMID: 26283910 PMCID: PMC4516891 DOI: 10.3389/fnmol.2015.00036] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 07/06/2015] [Indexed: 12/14/2022] Open
Abstract
Systemic delivery of self-complementary (sc) adeno-associated-virus vector of serotype 9 (AAV9) was recently shown to provide robust and widespread gene transfer to the central nervous system (CNS), opening new avenues for practical, and non-invasive gene therapy of neurological diseases. More recently, AAV of serotype rh10 (AAVrh10) was also found highly efficient to mediate CNS transduction after intravenous administration in mice. However, only a few studies compared AAV9 and AAVrh10 efficiencies, particularly in the spinal cord. In this study, we compared the transduction capabilities of AAV9 and AAVrh10 in the brain, the spinal cord, and the peripheral nervous system (PNS) after intravenous delivery in neonatal mice. As reported in previous studies, AAVrh10 achieved either similar or higher transduction than AAV9 in all the examined brain regions. The superiority of AAVrh10 over AAV9 appeared statistically significant only in the medulla and the cerebellum, but a clear trend was also observed in other structures like the hippocampus or the cortex. In contrast to previous studies, we found that AAVrh10 was more efficient than AAV9 for transduction of the dorsal spinal cord and the lower motor neurons (MNs). However, differences between the two serotypes appeared mainly significant at low dose, and surprisingly, increasing the dose did not improve AAVrh10 distribution in the spinal cord, in contrary to AAV9. Similar dose-related differences between transduction efficiency of the two serotypes were also observed in the sciatic nerve. These findings suggest differences in the transduction mechanisms of these two serotypes, which both hold great promise for gene therapy of neurological diseases.
Collapse
Affiliation(s)
- Yannick Tanguy
- Center of Research on Myology, FRE 3617 Centre National de la Recherche Scientifique, UMRS 974 INSERM, French Institute of Myology, Pierre and Marie Curie University Paris, France
| | - Maria G Biferi
- Center of Research on Myology, FRE 3617 Centre National de la Recherche Scientifique, UMRS 974 INSERM, French Institute of Myology, Pierre and Marie Curie University Paris, France
| | - Aurore Besse
- Center of Research on Myology, FRE 3617 Centre National de la Recherche Scientifique, UMRS 974 INSERM, French Institute of Myology, Pierre and Marie Curie University Paris, France
| | - Stephanie Astord
- Center of Research on Myology, FRE 3617 Centre National de la Recherche Scientifique, UMRS 974 INSERM, French Institute of Myology, Pierre and Marie Curie University Paris, France
| | - Mathilde Cohen-Tannoudji
- Center of Research on Myology, FRE 3617 Centre National de la Recherche Scientifique, UMRS 974 INSERM, French Institute of Myology, Pierre and Marie Curie University Paris, France
| | - Thibaut Marais
- Center of Research on Myology, FRE 3617 Centre National de la Recherche Scientifique, UMRS 974 INSERM, French Institute of Myology, Pierre and Marie Curie University Paris, France
| | - Martine Barkats
- Center of Research on Myology, FRE 3617 Centre National de la Recherche Scientifique, UMRS 974 INSERM, French Institute of Myology, Pierre and Marie Curie University Paris, France
| |
Collapse
|
33
|
Aronovich EL, Hackett PB. Lysosomal storage disease: gene therapy on both sides of the blood-brain barrier. Mol Genet Metab 2015; 114:83-93. [PMID: 25410058 PMCID: PMC4312729 DOI: 10.1016/j.ymgme.2014.09.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 09/29/2014] [Accepted: 09/29/2014] [Indexed: 12/17/2022]
Abstract
Most lysosomal storage disorders affect the nervous system as well as other tissues and organs of the body. Previously, the complexities of these diseases, particularly in treating neurologic abnormalities, were too great to surmount. However, based on recent developments there are realistic expectations that effective therapies are coming soon. Gene therapy offers the possibility of affordable, comprehensive treatment associated with these diseases currently not provided by standards of care. With a focus on correction of neurologic disease by systemic gene therapy of mucopolysaccharidoses types I and IIIA, we review some of the major recent advances in viral and non-viral vectors, methods of their delivery and strategies leading to correction of both the nervous and somatic tissues as well as evaluation of functional correction of neurologic manifestations in animal models. We discuss two questions: what systemic gene therapy strategies work best for correction of both somatic and neurologic abnormalities in a lysosomal storage disorder and is there evidence that targeting peripheral tissues (e.g., in the liver) has a future for ameliorating neurologic disease in patients?
Collapse
Affiliation(s)
- Elena L Aronovich
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, United States; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, United States.
| | - Perry B Hackett
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, United States; Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, United States
| |
Collapse
|
34
|
Intrathecal gene therapy corrects CNS pathology in a feline model of mucopolysaccharidosis I. Mol Ther 2014; 22:2018-2027. [PMID: 25027660 DOI: 10.1038/mt.2014.135] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 07/09/2014] [Indexed: 02/03/2023] Open
Abstract
Enzyme replacement therapy has revolutionized the treatment of the somatic manifestations of lysosomal storage diseases (LSD), although it has been ineffective in treating central nervous system (CNS) manifestations of these disorders. The development of neurotrophic vectors based on novel serotypes of adeno-associated viruses (AAV) such as AAV9 provides a potential platform for stable and efficient delivery of enzymes to the CNS. We evaluated the safety and efficacy of intrathecal delivery of AAV9 expressing α-l-iduronidase (IDUA) in a previously described feline model of mucopolysaccharidosis I (MPS I). A neurological phenotype has not been defined in these animals, so our analysis focused on the biochemical and histological CNS abnormalities characteristic of MPS I. Five MPS I cats were dosed with AAV9 vector at 4-7 months of age and followed for 6 months. Treated animals demonstrated virtually complete correction of biochemical and histological manifestations of the disease throughout the CNS. There was a range of antibody responses against IDUA in this cohort which reduced detectable enzyme without substantially reducing efficacy; there was no evidence of toxicity. This first demonstration of the efficacy of intrathecal gene therapy in a large animal model of a LSD should pave the way for translation into the clinic.
Collapse
|