1
|
Fang K, Yang X, Liu Y, Xia J, Wu R, Yang F, Feng C, Liu X, Shi L, Geng G, Yang H. A comprehensive study of AAV tropism across C57BL/6 mice, BALB/c mice, and crab-eating macaques. Mol Ther Methods Clin Dev 2025; 33:101434. [PMID: 40104150 PMCID: PMC11919325 DOI: 10.1016/j.omtm.2025.101434] [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: 04/28/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025]
Abstract
Recombinant adeno-associated viruses (AAVs) have been widely used for gene delivery and gene therapy. However, certain AAV serotypes exhibited distinct transduction patterns among different mouse strains or between mice and non-human primates (NHPs). These variations prompted us to investigate the AAV tropism of 21 capsid variants using barcoded AAV libraries among different tissues in C57BL/6 and BALB/c mice, as well as in crab-eating macaques. Our study unveiled that AAV tropisms varied significantly among different mouse strains and species, particularly in capsid variants such as AAV4, AAV9, PHP.B, and CAP-B10. Notably, AAV4 exhibited liver-detargeting properties in both mice and NHPs, and was remarkably efficient in transducing the lung, glomerulus, and pancreatic islet. These findings furnish crucial insights into the variations of AAV tropism among different mouse strains and species and facilitate the selection of appropriate AAV capsids for target tissues among different mouse strains and in NHPs.
Collapse
Affiliation(s)
- Kailun Fang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiali Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuanhua Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Junhui Xia
- Huidagene Therapeutics Inc., Shanghai 200131, China
| | - Ruoxi Wu
- Huidagene Therapeutics Inc., Shanghai 200131, China
| | - Fan Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Canbin Feng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinyu Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Linyu Shi
- Huidagene Therapeutics Inc., Shanghai 200131, China
| | - Guannan Geng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Hui Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired Intelligence, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- Huidagene Therapeutics Inc., Shanghai 200131, China
| |
Collapse
|
2
|
Sun B, Li R, Ji N, Liu H, Wang H, Chen C, Bai L, Su J, Chen J. Brain-targeting drug delivery systems: The state of the art in treatment of glioblastoma. Mater Today Bio 2025; 30:101443. [PMID: 39866779 PMCID: PMC11759563 DOI: 10.1016/j.mtbio.2025.101443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Glioblastoma (GBM) is the most prevalent primary malignant brain tumor, characterized by a high mortality rate and a poor prognosis. The blood-brain barrier (BBB) and the blood-tumor barrier (BTB) present significant obstacles to the efficacy of tumor-targeted pharmacotherapy, thereby impeding the therapeutic potential of numerous candidate drugs. Targeting delivery of adequate doses of drug across the BBB to treat GBM has become a prominent research area in recent years. This emphasis has driven the exploration and evaluation of diverse technologies for GBM pharmacotherapy, with some already undergoing clinical trials. This review provides a thorough overview of recent advancements and challenges in targeted drug delivery for GBM treatment. It specifically emphasizes systemic drug administration strategies to assess their potential and limitations in GBM treatment. Furthermore, this review highlights promising future research directions in the development of intelligent drug delivery systems aimed at overcoming current challenges and enhancing therapeutic efficacy against GBM. These advancements not only support foundational research on targeted drug delivery systems for GBM but also offer methodological approaches for future clinical applications.
Collapse
Affiliation(s)
- Bo Sun
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Rong Li
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ning Ji
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Hongxiang Wang
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chao Chen
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Juxiang Chen
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
3
|
Bijlani S, Pang KM, Bugga LV, Rangasamy S, Narayanan V, Chatterjee S. Nuclease-free precise genome editing corrects MECP2 mutations associated with Rett syndrome. Front Genome Ed 2024; 6:1346781. [PMID: 38495533 PMCID: PMC10940404 DOI: 10.3389/fgeed.2024.1346781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
Rett syndrome is an acquired progressive neurodevelopmental disorder caused by de novo mutations in the X-linked MECP2 gene which encodes a pleiotropic protein that functions as a global transcriptional regulator and a chromatin modifier. Rett syndrome predominantly affects heterozygous females while affected male hemizygotes rarely survive. Gene therapy of Rett syndrome has proven challenging due to a requirement for stringent regulation of expression with either over- or under-expression being toxic. Ectopic expression of MECP2 in conjunction with regulatory miRNA target sequences has achieved some success, but the durability of this approach remains unknown. Here we evaluated a nuclease-free homologous recombination (HR)-based genome editing strategy to correct mutations in the MECP2 gene. The stem cell-derived AAVHSCs have previously been shown to mediate seamless and precise HR-based genome editing. We tested the ability of HR-based genome editing to correct pathogenic mutations in Exons 3 and 4 of the MECP2 gene and restore the wild type sequence while preserving all native genomic regulatory elements associated with MECP2 expression, thus potentially addressing a significant issue in gene therapy for Rett syndrome. Moreover, since the mutations are edited directly at the level of the genome, the corrections are expected to be durable with progeny cells inheriting the edited gene. The AAVHSC MECP2 editing vector was designed to be fully homologous to the target MECP2 region and to insert a promoterless Venus reporter at the end of Exon 4. Evaluation of AAVHSC editing in a panel of Rett cell lines bearing mutations in Exons 3 and 4 demonstrated successful correction and rescue of expression of the edited MECP2 gene. Sequence analysis of edited Rett cells revealed successful and accurate correction of mutations in both Exons 3 and 4 and permitted mapping of HR crossover events. Successful correction was observed only when the mutations were flanked at both the 5' and 3' ends by crossover events, but not when both crossovers occurred either exclusively upstream or downstream of the mutation. Importantly, we concluded that pathogenic mutations were successfully corrected in every Rett line analyzed, demonstrating the therapeutic potential of HR-based genome editing.
Collapse
Affiliation(s)
- Swati Bijlani
- Department of Surgery, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Ka Ming Pang
- Department of Surgery, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Lakshmi V. Bugga
- Department of Surgery, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Sampath Rangasamy
- Center for Rare Childhood Disorders (C4RCD), Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, United States
| | - Vinodh Narayanan
- Center for Rare Childhood Disorders (C4RCD), Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, United States
| | - Saswati Chatterjee
- Department of Surgery, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| |
Collapse
|
4
|
Yao Y, Bei F. Adeno-associated Virus-Mediated Gene Delivery Across the Blood-Brain Barrier. ADVANCES IN NEUROBIOLOGY 2024; 41:91-112. [PMID: 39589711 DOI: 10.1007/978-3-031-69188-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Recombinant adeno-associated viruses (AAVs) have emerged as a popular tool for gene therapy in the central nervous system (CNS). Given the dense vasculature in the CNS, systemic administration is an appealing approach for achieving a broad distribution of AAV vectors across the CNS. However, the blood-brain barrier (BBB) is a major obstacle that blocks the entry of AAV vectors into the brain and spinal cord. Thus, there is a great need to develop novel AAV vector technology with enhanced BBB penetration. In this chapter, we briefly summarize AAV biology, possible mechanisms for AAV vectors to overcome the BBB and further engineering strategies, and current clinical trials using systemic AAV gene therapy for CNS diseases.
Collapse
Affiliation(s)
- Yizheng Yao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Fengfeng Bei
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
van Lieshout LP, Rubin M, Costa-Grant K, Ota S, Golebiowski D, Panico T, Wiberg E, Szymczak K, Gilmore R, Stanvick M, Burnham B, Gagnon J, Iwuchukwu I, Yang G, Ghazi I, Meola A, Dickerson R, Thiers T, Mustich L, Hayes A, Rivas I, Lotterhand J, Avila N, McGivney J, Yin J, Kelly T. A novel dual-plasmid platform provides scalable transfection yielding improved productivity and packaging across multiple AAV serotypes and genomes. Mol Ther Methods Clin Dev 2023; 29:426-436. [PMID: 37273900 PMCID: PMC10238442 DOI: 10.1016/j.omtm.2023.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 05/04/2023] [Indexed: 06/06/2023]
Abstract
Transient transfection of mammalian cells using plasmid DNA is a standard method to produce adeno-associated virus (AAV) vectors allowing for flexible and scalable manufacture. Typically, three plasmids are used to encode the necessary components to facilitate vector production; however, a dual-plasmid system, termed pDG, was introduced over 2 decades ago demonstrating two components could be combined resulting in comparable productivity to triple transfection. We have developed a novel dual-plasmid system, pOXB, with an alternative arrangement of sequences that results in significantly increased AAV vector productivity and percentage of full capsids packaged in comparison to the pDG dual design and triple transfection. Here, we demonstrate the reproducibility of these findings across seven recombinant AAV genomes and multiple capsid serotypes as well as the scalability of the pOXB dual-plasmid transfection at 50-L bioreactor scale. Purified drug substance showed a consistent product quality profile in line with triple-transfected vectors, except for a substantial improvement in intact genomes packaged using the pOXB dual- transfection system. Furthermore, pOXB dual- and triple-transfection-based vectors performed consistently in vivo. The pOXB dual plasmid represents an innovation in AAV manufacturing resulting in significant process gains while maintaining the flexibility of a transient transfection platform.
Collapse
Affiliation(s)
| | - Miranda Rubin
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | | | - Stacy Ota
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Diane Golebiowski
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Troy Panico
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Eli Wiberg
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Klaudia Szymczak
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Richard Gilmore
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Marissa Stanvick
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Brenda Burnham
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Jeff Gagnon
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | | | - Guang Yang
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Iraj Ghazi
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Alex Meola
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Ryan Dickerson
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Thomas Thiers
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Luke Mustich
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - April Hayes
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - Israel Rivas
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - Jason Lotterhand
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - Nancy Avila
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - James McGivney
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Jin Yin
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| | - Tim Kelly
- Oxford Biomedica Solutions LLC, 1 Patriots Park, Bedford, MA 01730, USA
| |
Collapse
|
6
|
St Martin T, Seabrook TA, Gall K, Newman J, Avila N, Hayes A, Kivaa M, Lotterhand J, Mercaldi M, Patel K, Rivas IJ, Woodcock S, Wright TL, Seymour AB, Francone OL, Gingras J. Single Systemic Administration of a Gene Therapy Leading to Disease Treatment in Metachromatic Leukodystrophy Arsa Knock-Out Mice. J Neurosci 2023; 43:3567-3581. [PMID: 36977578 PMCID: PMC10184740 DOI: 10.1523/jneurosci.1829-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/20/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) is a rare, inherited, demyelinating lysosomal storage disorder caused by mutations in the arylsulfatase-A gene (ARSA). In patients, levels of functional ARSA enzyme are diminished and lead to deleterious accumulation of sulfatides. Herein, we demonstrate that intravenous administration of HSC15/ARSA restored the endogenous murine biodistribution of the corresponding enzyme, and overexpression of ARSA corrected disease biomarkers and ameliorated motor deficits in Arsa KO mice of either sex. In treated Arsa KO mice, when compared with intravenously administered AAV9/ARSA, significant increases in brain ARSA activity, transcript levels, and vector genomes were observed with HSC15/ARSA Durability of transgene expression was established in neonate and adult mice out to 12 and 52 weeks, respectively. Levels and correlation between changes in biomarkers and ARSA activity required to achieve functional motor benefit was also defined. Finally, we demonstrated blood-nerve, blood-spinal and blood-brain barrier crossing as well as the presence of circulating ARSA enzyme activity in the serum of healthy nonhuman primates of either sex. Together, these findings support the use of intravenous delivery of HSC15/ARSA-mediated gene therapy for the treatment of MLD.SIGNIFICANCE STATEMENT Herein, we describe the method of gene therapy adeno-associated virus (AAV) capsid and route of administration selection leading to an efficacious gene therapy in a mouse model of metachromatic leukodystrophy. We demonstrate the therapeutic outcome of a new naturally derived clade F AAV capsid (AAVHSC15) in a disease model and the importance of triangulating multiple end points to increase the translation into higher species via ARSA enzyme activity and biodistribution profile (with a focus on the CNS) with that of a key clinically relevant biomarker.
Collapse
Affiliation(s)
| | | | | | - Jenn Newman
- Homology Medicines, Bedford, Massachusetts 01730
| | - Nancy Avila
- Homology Medicines, Bedford, Massachusetts 01730
| | - April Hayes
- Homology Medicines, Bedford, Massachusetts 01730
| | | | | | | | - Kruti Patel
- Homology Medicines, Bedford, Massachusetts 01730
| | | | | | | | | | | | | |
Collapse
|
7
|
Zhou K, Han J, Wang Y, Zhang Y, Zhu C. Routes of administration for adeno-associated viruses carrying gene therapies for brain diseases. Front Mol Neurosci 2022; 15:988914. [PMID: 36385771 PMCID: PMC9643316 DOI: 10.3389/fnmol.2022.988914] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/03/2022] [Indexed: 08/27/2023] Open
Abstract
Gene therapy is a powerful tool to treat various central nervous system (CNS) diseases ranging from monogenetic diseases to neurodegenerative disorders. Adeno-associated viruses (AAVs) have been widely used as the delivery vehicles for CNS gene therapies due to their safety, CNS tropism, and long-term therapeutic effect. However, several factors, including their ability to cross the blood-brain barrier, the efficiency of transduction, their immunotoxicity, loading capacity, the choice of serotype, and peripheral off-target effects should be carefully considered when designing an optimal AAV delivery strategy for a specific disease. In addition, distinct routes of administration may affect the efficiency and safety of AAV-delivered gene therapies. In this review, we summarize different administration routes of gene therapies delivered by AAVs to the brain in mice and rats. Updated knowledge regarding AAV-delivered gene therapies may facilitate the selection from various administration routes for specific disease models in future research.
Collapse
Affiliation(s)
- Kai Zhou
- Henan Neurodevelopment Engineering Research Center for Children, Zhengzhou Key Laboratory of Pediatric Neurobehavior, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Jinming Han
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yafeng Wang
- Henan Neurodevelopment Engineering Research Center for Children, Zhengzhou Key Laboratory of Pediatric Neurobehavior, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Department of Hematology and Oncology, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Yaodong Zhang
- Henan Neurodevelopment Engineering Research Center for Children, Zhengzhou Key Laboratory of Pediatric Neurobehavior, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, The Third Affiliated Hospital and Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Centre for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
8
|
Variants of the adeno-associated virus serotype 9 with enhanced penetration of the blood-brain barrier in rodents and primates. Nat Biomed Eng 2022; 6:1257-1271. [PMID: 36217021 DOI: 10.1038/s41551-022-00938-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/13/2022] [Indexed: 11/08/2022]
Abstract
The development of gene therapies for the treatment of diseases of the central nervous system has been hindered by the limited availability of adeno-associated viruses (AAVs) that efficiently traverse the blood-brain barrier (BBB). Here, we report the rational design of AAV9 variants displaying cell-penetrating peptides on the viral capsid and the identification of two variants, AAV.CPP.16 and AAV.CPP.21, with improved transduction efficiencies of cells of the central nervous system on systemic delivery (6- to 249-fold across 4 mouse strains and 5-fold in cynomolgus macaques, with respect to the AAV9 parent vector). We also show that the neurotropism of AAV.CPP.16 is retained in young and adult macaques, that this variant displays enhanced transcytosis at the BBB as well as increased efficiency of cellular transduction relative to AAV9, and that it can be used to deliver antitumour payloads in a mouse model of glioblastoma. AAV capsids that can efficiently penetrate the BBB will facilitate the clinical translation of gene therapies aimed at the central nervous system.
Collapse
|
9
|
Smith LJ, Schulman LA, Smith S, Van Lieshout L, Barnes CM, Behmoiras L, Scarpitti M, Kivaa M, Duong KL, Benard LO, Ellsworth JL, Avila N, Faulkner D, Hayes A, Lotterhand J, Rivas JI, Sengooba AV, Tzianabos A, Seymour AB, Francone OL. Natural variations in AAVHSC16 significantly reduce liver tropism and maintain broad distribution to periphery and CNS. Mol Ther Methods Clin Dev 2022; 26:224-238. [PMID: 35859693 PMCID: PMC9287613 DOI: 10.1016/j.omtm.2022.06.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/27/2022] [Indexed: 12/19/2022]
Abstract
Adeno-associated viruses derived from human hematopoietic stem cells (AAVHSCs) are naturally occurring AAVs. Fifteen AAVHSCs have demonstrated broad biodistribution while displaying differences in transduction. We examine the structure-function relationships of these natural amino acid variations on cellular binding. We demonstrate that AAVHSC16 is the only AAVHSC that does not preferentially bind to terminal galactose. AAVHSC16 contains two unique amino acids, 501I and 706C, compared with other AAVHSCs. Through mutagenesis, we determined that residue 501 contributes to the lack of galactose binding. Structural analysis revealed that residue 501 is in proximity to the galactose binding pocket, hence confirming its functional role in galactose binding. Biodistribution analysis of AAVHSC16 indicated significantly less liver tropism in mice and non-human primates compared with other clade F members, likely associated with overall binding differences observed in vitro. AAVHSC16 maintained robust tropism to other key tissues in the peripheral and central nervous systems after intravenous injection, including to the brain, heart, and gastrocnemius. Importantly, AAVHSC16 did not induce elevated liver enzyme levels in non-human primates after intravenous injection at high doses. The unique glycan binding and tropism of AAVHSC16 makes this naturally occurring capsid an attractive candidate for therapies requiring less liver tropism while maintaining broad biodistribution.
Collapse
Affiliation(s)
- Laura J Smith
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | | | - Samantha Smith
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | | | - Carmen M Barnes
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - Liana Behmoiras
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - Meghan Scarpitti
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - Monicah Kivaa
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - Khanh L Duong
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - Ludo O Benard
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - Jeff L Ellsworth
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - Nancy Avila
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - Deiby Faulkner
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - April Hayes
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - Jason Lotterhand
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | | | | | - Alec Tzianabos
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - Albert B Seymour
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| | - Omar L Francone
- Homology Medicines, Inc., 1 Patriots Park, Bedford, MA 01730, USA
| |
Collapse
|
10
|
Challis RC, Ravindra Kumar S, Chen X, Goertsen D, Coughlin GM, Hori AM, Chuapoco MR, Otis TS, Miles TF, Gradinaru V. Adeno-Associated Virus Toolkit to Target Diverse Brain Cells. Annu Rev Neurosci 2022; 45:447-469. [PMID: 35440143 DOI: 10.1146/annurev-neuro-111020-100834] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recombinant adeno-associated viruses (AAVs) are commonly used gene delivery vehicles for neuroscience research. They have two engineerable features: the capsid (outer protein shell) and cargo (encapsulated genome). These features can be modified to enhance cell type or tissue tropism and control transgene expression, respectively. Several engineered AAV capsids with unique tropisms have been identified, including variants with enhanced central nervous system transduction, cell type specificity, and retrograde transport in neurons. Pairing these AAVs with modern gene regulatory elements and state-of-the-art reporter, sensor, and effector cargo enables highly specific transgene expression for anatomical and functional analyses of brain cells and circuits. Here, we discuss recent advances that provide a comprehensive (capsid and cargo) AAV toolkit for genetic access to molecularly defined brain cell types. Expected final online publication date for the Annual Review of Neuroscience, Volume 45 is July 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Rosemary C Challis
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Sripriya Ravindra Kumar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Xinhong Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - David Goertsen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Gerard M Coughlin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Acacia M Hori
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Miguel R Chuapoco
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Thomas S Otis
- Sainsbury Wellcome Centre, University College London, London, United Kingdom
| | - Timothy F Miles
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| |
Collapse
|
11
|
Pietersz KL, Pouw S, Klima J, Ellederova Z, Bohuslavova B, Chrastina J, Liscak R, Urgosik D, Starek Z, Crha M, Lewis O, Wooley M, Johnson D, Brouwers CC, Evers M, Motlik J, Martens GJM, Konstantinova PS, Blits B. Transduction profiles in minipig following MRI guided delivery of AAV-5 into thalamic and corona radiata areas. J Neurosci Methods 2022; 365:109382. [PMID: 34637809 DOI: 10.1016/j.jneumeth.2021.109382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND As a step towards clinical use of AAV-mediated gene therapy, brains of large animals are used to settle delivery parameters as most brain connections, and relative sizes in large animals and primates, are reasonably common. Prior to application in the clinic, approaches that have shown to be successful in rodent models are tested in larger animal species, such as dogs, non-human primates, and in this case, minipigs. NEW METHOD We evaluated alternate delivery routes to target the basal ganglia by injections into the more superficial corona radiata, and, deeper into the brain, the thalamus. Anatomically known connections can be used to predict the expression of the transgene following infusion of AAV5. For optimal control over delivery of the vector with regards to anatomical location in the brain and spread in the tissue, we have used magnetic resonance image-guided convection-enhanced diffusion delivery. RESULTS While the transduction of the cortex was observed, only partial transduction of the basal ganglia was achieved via the corona radiata. Thalamic administration, on the other hand, resulted in widespread transduction from the midbrain to the frontal cortex COMPARISON WITH EXISTING METHODS: Compared to other methods, such as delivery directly to the striatum, thalamic injection may provide an alternative when for instance, injection into the basal ganglia directly is not feasible. CONCLUSIONS The study results suggest that thalamic administration of AAV5 has significant potential for indications where the transduction of specific areas of the brain is required.
Collapse
Affiliation(s)
- K L Pietersz
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, The Netherlands; Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - S Pouw
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, The Netherlands
| | - J Klima
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Z Ellederova
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - B Bohuslavova
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - J Chrastina
- Department of Neurosurgery, St. Anne's University Hospital, Brno, Czech Republic
| | - R Liscak
- Department of Stereotactic Radioneurosurgery, Na Homolce Hospital, Prague, Czech Republic
| | - D Urgosik
- Department of Stereotactic Radioneurosurgery, Na Homolce Hospital, Prague, Czech Republic
| | - Z Starek
- Interventional Cardiac Electrophysiology, St.' Anne's University Hospital, Brno, Czech Republic
| | - M Crha
- Small Animal Clinic, Veterinary and Pharmaceutical University, Brno, Czech Republic
| | - O Lewis
- Renishaw Neuro Solutions (RNS) ltd, Renishaw plc, Gloucestershire, UK
| | - M Wooley
- Renishaw Neuro Solutions (RNS) ltd, Renishaw plc, Gloucestershire, UK
| | - D Johnson
- Renishaw Neuro Solutions (RNS) ltd, Renishaw plc, Gloucestershire, UK
| | - C C Brouwers
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, The Netherlands
| | - M Evers
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, The Netherlands
| | - J Motlik
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - G J M Martens
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - P S Konstantinova
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, The Netherlands
| | - B Blits
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, The Netherlands.
| |
Collapse
|
12
|
Jensen TL, Gøtzsche CR, Woldbye DPD. Current and Future Prospects for Gene Therapy for Rare Genetic Diseases Affecting the Brain and Spinal Cord. Front Mol Neurosci 2021; 14:695937. [PMID: 34690692 PMCID: PMC8527017 DOI: 10.3389/fnmol.2021.695937] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, gene therapy has been raising hopes toward viable treatment strategies for rare genetic diseases for which there has been almost exclusively supportive treatment. We here review this progress at the pre-clinical and clinical trial levels as well as market approvals within diseases that specifically affect the brain and spinal cord, including degenerative, developmental, lysosomal storage, and metabolic disorders. The field reached an unprecedented milestone when Zolgensma® (onasemnogene abeparvovec) was approved by the FDA and EMA for in vivo adeno-associated virus-mediated gene replacement therapy for spinal muscular atrophy. Shortly after EMA approved Libmeldy®, an ex vivo gene therapy with lentivirus vector-transduced autologous CD34-positive stem cells, for treatment of metachromatic leukodystrophy. These successes could be the first of many more new gene therapies in development that mostly target loss-of-function mutation diseases with gene replacement (e.g., Batten disease, mucopolysaccharidoses, gangliosidoses) or, less frequently, gain-of-toxic-function mutation diseases by gene therapeutic silencing of pathologic genes (e.g., amyotrophic lateral sclerosis, Huntington's disease). In addition, the use of genome editing as a gene therapy is being explored for some diseases, but this has so far only reached clinical testing in the treatment of mucopolysaccharidoses. Based on the large number of planned, ongoing, and completed clinical trials for rare genetic central nervous system diseases, it can be expected that several novel gene therapies will be approved and become available within the near future. Essential for this to happen is the in depth characterization of short- and long-term effects, safety aspects, and pharmacodynamics of the applied gene therapy platforms.
Collapse
Affiliation(s)
- Thomas Leth Jensen
- Department of Neurology, Rigshospitalet University Hospital, Copenhagen, Denmark
| | | | | |
Collapse
|
13
|
Bell RD. Considerations When Developing Blood-Brain Barrier Crossing Drug Delivery Technology. Handb Exp Pharmacol 2021; 273:83-95. [PMID: 34463850 DOI: 10.1007/164_2021_453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Efficient therapeutic transport across the neurovasculature remains a challenge for developing medicine to treat central nervous system (CNS) disorders (Bell and Ehlers, Neuron 81:1-3, 2014). This chapter is meant to provide some insight and key considerations for developing and evaluating various technologies and approaches to CNS drug delivery. First, a brief review of various biological barriers, including the immune system, cellular and protein components of the blood-brain barrier (BBB), and clearance mechanisms in peripheral organs is provided. Next, a few examples and learnings from existing BBB-crossing modalities will be reviewed. Insight from "BBBomic" databases and thoughts on basic requirements for successful in vivo validation studies are discussed. Finally, an additional engineering barrier, namely manufacturing and product scalability, is highlighted as it relates to clinical translation and feasibility for developing BBB-crossing delivery technologies. A goal of this chapter is to provide an overview of the many barriers to the successful delivery of medicines into the brain. An emphasis will be placed on biotherapeutic and gene therapy applications for the treatment of neurological and neurodegenerative disorders.
Collapse
Affiliation(s)
- Robert D Bell
- Rare Disease Research Unit, Pfizer Worldwide Research, Development and Medicine, Cambridge, MA, USA.
| |
Collapse
|
14
|
Chatterjee S, Sivanandam V, Wong KKM. Adeno-Associated Virus and Hematopoietic Stem Cells: The Potential of Adeno-Associated Virus Hematopoietic Stem Cells in Genetic Medicines. Hum Gene Ther 2021; 31:542-552. [PMID: 32253938 DOI: 10.1089/hum.2020.049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adeno-associated virus (AAV)-based vectors have transformed into powerful elements of genetic medicine with proven therapeutic efficacy and a good safety profile. Over the years, efforts to transduce hematopoietic stem cells (HSCs) with AAV2 vectors have, however, been challenging. While there was evidence that AAV2 delivered vector genomes to primitive, quiescent, multipotential, self-renewing, in vivo engrafting HSCs, transgene expression was elusive. In this study, we review the evolution of AAV transduction of HSC, starting with AAV2 vectors leading to the isolation of a family of naturally occurring AAVs from human CD34+ HSC, the AAVHSC. The stem cell-derived AAVHSCs have turned out to have remarkable potentials for genetic therapies well beyond the hematopoietic system. AAVHSCs have tropism for a wide variety of peripheral tissues, including the liver, muscle, and the retina. They cross the blood-brain barrier and transduce cells of the central nervous system. Preclinical gene therapy studies underway using AAVHSC vectors are discussed. We review the notable ability of AAVHSCs to mediate efficient, seamless homologous recombination in the absence of exogenous nuclease activity and discuss the therapeutic implications. We also discuss early results from an AAVHSC-based clinical gene therapy trial that is underway for the treatment of phenylketonuria. Thus, the stem cell-derived AAVHSC, offer a multifaceted platform for in vivo gene therapy and genome editing for the treatment of inherited diseases.
Collapse
Affiliation(s)
- Saswati Chatterjee
- Department of Surgery, Beckman Research Institute of City of Hope Medical Center, Duarte, California, USA
| | - Venkatesh Sivanandam
- Department of Surgery, Beckman Research Institute of City of Hope Medical Center, Duarte, California, USA
| | - Kamehameha Kai-Min Wong
- Department of Hematology and Stem Cell Transplantation, City of Hope Medical Center, Duarte, California, USA
| |
Collapse
|
15
|
Croze RH, Kotterman M, Burns CH, Schmitt CE, Quezada M, Schaffer D, Kirn D, Francis P. Viral Vector Technologies and Strategies: Improving on Nature. Int Ophthalmol Clin 2021; 61:59-89. [PMID: 34196318 PMCID: PMC8253506 DOI: 10.1097/iio.0000000000000361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
16
|
von Jonquieres G, Rae CD, Housley GD. Emerging Concepts in Vector Development for Glial Gene Therapy: Implications for Leukodystrophies. Front Cell Neurosci 2021; 15:661857. [PMID: 34239416 PMCID: PMC8258421 DOI: 10.3389/fncel.2021.661857] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Central Nervous System (CNS) homeostasis and function rely on intercellular synchronization of metabolic pathways. Developmental and neurochemical imbalances arising from mutations are frequently associated with devastating and often intractable neurological dysfunction. In the absence of pharmacological treatment options, but with knowledge of the genetic cause underlying the pathophysiology, gene therapy holds promise for disease control. Consideration of leukodystrophies provide a case in point; we review cell type – specific expression pattern of the disease – causing genes and reflect on genetic and cellular treatment approaches including ex vivo hematopoietic stem cell gene therapies and in vivo approaches using adeno-associated virus (AAV) vectors. We link recent advances in vectorology to glial targeting directed towards gene therapies for specific leukodystrophies and related developmental or neurometabolic disorders affecting the CNS white matter and frame strategies for therapy development in future.
Collapse
Affiliation(s)
- Georg von Jonquieres
- Translational Neuroscience Facility, Department of Physiology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Caroline D Rae
- Neuroscience Research Australia, Randwick, NSW, Australia
| | - Gary D Housley
- Translational Neuroscience Facility, Department of Physiology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
17
|
Zhu D, Schieferecke AJ, Lopez PA, Schaffer DV. Adeno-Associated Virus Vector for Central Nervous System Gene Therapy. Trends Mol Med 2021; 27:524-537. [PMID: 33895085 DOI: 10.1016/j.molmed.2021.03.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/15/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022]
Abstract
The past several years have witnessed significant advances in the development of therapeutic gene delivery for neurological disorders of the central nervous system (CNS). In particular, genome-wide sequencing analysis has deepened our understanding of mutations that underlie many monogenic disorders, which in turn has contributed to clinical advances involving adeno-associated virus (AAV) vector delivery of replacement genes to treat recessive disorders. Moreover, gene therapy has been further bolstered with advances in genome editing tools that allow researchers to silence, repair, and amend endogenous genes. However, despite strong preclinical and clinical progress, challenges remain, including delivery and safety. Here, we discuss advances in AAV engineering, recent developments in cargo design, and translation of these technologies towards clinical progress.
Collapse
Affiliation(s)
- Danqing Zhu
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA, 94720, USA
| | - Adam J Schieferecke
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Paola A Lopez
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
| | - David V Schaffer
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA, 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA; Department of Bioengineering, University of California, Berkeley, CA, 94720, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA.
| |
Collapse
|
18
|
Nonnenmacher M, Wang W, Child MA, Ren XQ, Huang C, Ren AZ, Tocci J, Chen Q, Bittner K, Tyson K, Pande N, Chung CHY, Paul SM, Hou J. Rapid evolution of blood-brain-barrier-penetrating AAV capsids by RNA-driven biopanning. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 20:366-378. [PMID: 33553485 PMCID: PMC7841218 DOI: 10.1016/j.omtm.2020.12.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/16/2020] [Indexed: 12/26/2022]
Abstract
Therapeutic payload delivery to the central nervous system (CNS) remains a major challenge in gene therapy. Recent studies using function-driven evolution of adeno-associated virus (AAV) vectors have successfully identified engineered capsids with improved blood-brain barrier (BBB) penetration and CNS tropism in mouse. However, these strategies require transgenic animals and thus are limited to rodents. To address this issue, we developed a directed evolution approach based on recovery of capsid library RNA transcribed from CNS-restricted promoters. This RNA-driven screen platform, termed TRACER (Tropism Redirection of AAV by Cell-type-specific Expression of RNA), was tested in the mouse with AAV9 peptide display libraries and showed rapid emergence of dominant sequences. Ten individual variants were characterized and showed up to 400-fold higher brain transduction over AAV9 following systemic administration. Our results demonstrate that the TRACER platform allows rapid selection of AAV capsids with robust BBB penetration and CNS tropism in non-transgenic animals.
Collapse
Affiliation(s)
| | - Wei Wang
- Voyager Therapeutics, Cambridge, MA 02139, USA
| | | | | | - Carol Huang
- Voyager Therapeutics, Cambridge, MA 02139, USA
| | | | - Jenna Tocci
- Voyager Therapeutics, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | - Jay Hou
- Voyager Therapeutics, Cambridge, MA 02139, USA
| |
Collapse
|
19
|
Winkelsas AM, Fischbeck KH. Nucleic acid therapeutics in neurodevelopmental disease. Curr Opin Genet Dev 2020; 65:112-116. [PMID: 32623324 PMCID: PMC11441424 DOI: 10.1016/j.gde.2020.05.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/22/2020] [Indexed: 12/16/2022]
Abstract
Nucleic acid therapeutics allow sequence-based targeting of mutation-harboring genes. They can be used to increase the expression and function of disease genes or to decrease the expression of toxic gene products. Antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), and gene-replacement therapies have received FDA approval, and in vivo gene editing applications are currently under development. Special consideration should be given to target engagement in neurons and amelioration of neurological phenotypes. Here we discuss the uses and limitations of different nucleic acid therapeutics, highlighting examples in the clinical and pre-clinical application of these modalities for the treatment of neurodevelopmental diseases.
Collapse
Affiliation(s)
- Audrey M Winkelsas
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States; Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Kenneth H Fischbeck
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
20
|
Shaimardanova AA, Chulpanova DS, Solovyeva VV, Mullagulova AI, Kitaeva KV, Allegrucci C, Rizvanov AA. Metachromatic Leukodystrophy: Diagnosis, Modeling, and Treatment Approaches. Front Med (Lausanne) 2020; 7:576221. [PMID: 33195324 PMCID: PMC7606900 DOI: 10.3389/fmed.2020.576221] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/18/2020] [Indexed: 12/31/2022] Open
Abstract
Metachromatic leukodystrophy is a lysosomal storage disease, which is characterized by damage of the myelin sheath that covers most of nerve fibers of the central and peripheral nervous systems. The disease occurs due to a deficiency of the lysosomal enzyme arylsulfatase A (ARSA) or its sphingolipid activator protein B (SapB) and it clinically manifests as progressive motor and cognitive deficiency. ARSA and SapB protein deficiency are caused by mutations in the ARSA and PSAP genes, respectively. The severity of clinical course in metachromatic leukodystrophy is determined by the residual ARSA activity, depending on the type of mutation. Currently, there is no effective treatment for this disease. Clinical cases of bone marrow or cord blood transplantation have been reported, however the therapeutic effectiveness of these methods remains insufficient to prevent aggravation of neurological disorders. Encouraging results have been obtained using gene therapy for delivering the wild-type ARSA gene using vectors based on various serotypes of adeno-associated viruses, as well as using mesenchymal stem cells and combined gene-cell therapy. This review discusses therapeutic strategies for the treatment of metachromatic leukodystrophy, as well as diagnostic methods and modeling of this pathology in animals to evaluate the effectiveness of new therapies.
Collapse
Affiliation(s)
- Alisa A Shaimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Daria S Chulpanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, The Russian Academy of Sciences, Moscow, Russia
| | - Valeriya V Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, The Russian Academy of Sciences, Moscow, Russia
| | - Aysilu I Mullagulova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Kristina V Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Cinzia Allegrucci
- School of Veterinary Medicine and Science (SVMS) and Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
21
|
Chen HM, Resendes R, Ghodssi A, Sookiasian D, Tian M, Dollive S, Adamson-Small L, Avila N, Tazearslan C, Thompson JF, Ellsworth JL, Francone O, Seymour A, Wright JB. Molecular characterization of precise in vivo targeted gene integration in human cells using AAVHSC15. PLoS One 2020; 15:e0233373. [PMID: 32453743 PMCID: PMC7250422 DOI: 10.1371/journal.pone.0233373] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/04/2020] [Indexed: 02/08/2023] Open
Abstract
Targeted gene integration via precise homologous recombination (HR)-based gene editing has the potential to correct genetic diseases. AAV (adeno-associated virus) can mediate nuclease-free gene integration at a disease-causing locus. Therapeutic application of AAV gene integration requires quantitative molecular characterization of the edited sequence that overcome technical obstacles such as excess episomal vector genomes and lengthy homology arms. Here we describe a novel molecular methodology that utilizes quantitative next-generation sequencing to characterize AAV-mediated targeted insertion and detects the presence of unintended mutations. The methods described here quantify targeted insertion and query the entirety of the target locus for the presence of insertions, deletions, single nucleotide variants (SNVs) and integration of viral components such as inverted terminal repeats (ITR). Using a humanized liver murine model, we demonstrate that hematopoietic stem-cell derived AAVHSC15 mediates in vivo targeted gene integration into human chromosome 12 at the PAH (phenylalanine hydroxylase) locus at 6% frequency, with no sign of co-incident random mutations at or above a lower limit of detection of 0.5% and no ITR sequences at the integration sites. Furthermore, analysis of heterozygous variants across the targeted locus using the methods described shows a pattern of strand cross-over, supportive of an HR mechanism of gene integration with similar efficiencies across two different haplotypes. Rapid advances in the application of AAV-mediated nuclease-free target integration, or gene editing, as a new therapeutic modality requires precise understanding of the efficiency and the nature of the changes being introduced to the target genome at the molecular level. This work provides a framework to be applied to homologous recombination gene editing platforms for assessment of introduced and natural sequence variation across a target site.
Collapse
Affiliation(s)
- Huei-Mei Chen
- Homology Medicines Inc., Bedford, Massachusetts, United States of America
| | - Rachel Resendes
- Homology Medicines Inc., Bedford, Massachusetts, United States of America
| | - Azita Ghodssi
- Homology Medicines Inc., Bedford, Massachusetts, United States of America
| | | | - Michael Tian
- Homology Medicines Inc., Bedford, Massachusetts, United States of America
| | - Serena Dollive
- Homology Medicines Inc., Bedford, Massachusetts, United States of America
| | | | - Nancy Avila
- Homology Medicines Inc., Bedford, Massachusetts, United States of America
| | - Cagdas Tazearslan
- Homology Medicines Inc., Bedford, Massachusetts, United States of America
| | - John F. Thompson
- Homology Medicines Inc., Bedford, Massachusetts, United States of America
| | - Jeff L. Ellsworth
- Homology Medicines Inc., Bedford, Massachusetts, United States of America
| | - Omar Francone
- Homology Medicines Inc., Bedford, Massachusetts, United States of America
| | - Albert Seymour
- Homology Medicines Inc., Bedford, Massachusetts, United States of America
| | - Jason B. Wright
- Homology Medicines Inc., Bedford, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|