1
|
Matuszek Z, Brown BL, Yrigollen CM, Keiser MS, Davidson BL. Current trends in gene therapy to treat inherited disorders of the brain. Mol Ther 2025; 33:1988-2014. [PMID: 40181540 DOI: 10.1016/j.ymthe.2025.03.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/05/2025] Open
Abstract
Gene therapy development, re-engineering, and application to patients hold promise to revolutionize medicine, including therapies for disorders of the brain. Advances in delivery modalities, expression regulation, and improving safety profiles are of critical importance. Additionally, each inherited disorder has its own unique characteristics as to regions and cell types impacted and the temporal dynamics of that impact that are essential for the design of therapeutic design strategies. Here, we review the current state of the art in gene therapies for inherited brain disorders, summarizing key considerations for vector delivery, gene addition, gene silencing, gene editing, and epigenetic editing. We provide examples from animal models, human cell lines, and, where possible, clinical trials. This review also highlights the various tools available to researchers for basic research questions and discusses our views on the current limitations in the field.
Collapse
Affiliation(s)
- Zaneta Matuszek
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Brandon L Brown
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Epilepsy and Neurodevelopmental Disorders (ENDD), Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Carolyn M Yrigollen
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Megan S Keiser
- Department of Neurological Surgery, The Ohio State Wexner Medical Center, Columbus, OH 43210, USA
| | - Beverly L Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Center for Epilepsy and Neurodevelopmental Disorders (ENDD), Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
2
|
Nisanov AM, Rivera de Jesús JA, Schaffer DV. Advances in AAV capsid engineering: Integrating rational design, directed evolution and machine learning. Mol Ther 2025; 33:1937-1945. [PMID: 40176349 DOI: 10.1016/j.ymthe.2025.03.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025] Open
Abstract
Adeno-associated virus (AAV) has emerged as a highly promising vector for human gene therapy due to its favorable safety profile, versatility, and ability to transduce a wide range of tissues. However, natural AAV serotypes have shortcomings, including suboptimal transduction efficiency, pre-existing immunity, and a lack of tissue specificity, that hinder their therapeutic potential. To address these challenges, significant efforts are being applied to engineer novel AAV capsids. Rational design leverages structural insights to enhance capsid properties, directed evolution enables unbiased selection of superior variants, and machine learning accelerates discovery by computational analysis of high-throughput screening results to enable predictive algorithms. These strategies have yielded novel capsids with improved transduction efficiency, reduced immunogenicity, and enhanced tissue targeting. Future advances that continue to integrate such multi-disciplinary approaches will further drive the clinical translation of AAV-based therapies.
Collapse
Affiliation(s)
- Alan M Nisanov
- Department of Chemistry, University of California, Berkeley, Berkeley CA 94720, USA
| | - Julio A Rivera de Jesús
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; Graduate Program in Bioengineering, University of California, Berkeley, San Francisco and University of California, Berkeley, CA 94720, USA; Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
3
|
Keeler AM, Zhan W, Ram S, Fitzgerald KA, Gao G. The curious case of AAV immunology. Mol Ther 2025; 33:1946-1965. [PMID: 40156190 DOI: 10.1016/j.ymthe.2025.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Immune responses to adeno-associated virus (AAV) have long been perplexing, from its first discovery to the latest clinical trials of recombinant AAV (rAAV) therapy. Wild-type AAV (wtAAV) does not cause any known disease, making it an ideal vector for gene therapy, as viral vectors retain virus-like properties. Although AAV stimulates only a mild immune response compared with other viruses, it is still recognized by the innate immune system and induces adaptive immune responses. B cell responses against both wtAAV and rAAV are robust and can hinder gene therapy applications and prevent redosing. T cell responses can clear transduced cells or establish tolerance against gene therapy. Immune responses to AAV gene therapy are influenced by many factors. Most clinical immunotoxicities that develop in response to gene therapies have emerged as higher doses of AAV vectors have been utilized and were not properly modeled in preclinical animal studies. Thus, several strategies have been undertaken to reduce or mitigate immune responses to AAV. While we have learned a considerable amount about how the immune system responds to AAV gene therapy since the discovery of AAV virus, it still remains a curious case that requires more investigation to fully understand.
Collapse
Affiliation(s)
- Allison M Keeler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; NeroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA; Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sanjay Ram
- Division of Infectious Diseases and Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Katherine A Fitzgerald
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
4
|
Suarez-Amaran L, Song L, Tretiakova AP, Mikhail SA, Samulski RJ. AAV vector development, back to the future. Mol Ther 2025; 33:1903-1936. [PMID: 40186350 DOI: 10.1016/j.ymthe.2025.03.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025] Open
Abstract
Adeno-associated virus (AAV) has become a pivotal tool in gene therapy, providing a safe and efficient platform for long-term transgene expression. This review presents a comprehensive analysis of AAV's historical development, from its initial identification as a "contaminant" to its current clinical applications. We examine the molecular evolution of AAV, detailing advancements in vector engineering, rational design, directed evolution platforms, and computational modeling, which have expanded its therapeutic potential across diverse disease areas. Additionally, we explore AAV genome regulation, with a particular focus on inverted terminal repeats (ITRs) and AAV capsid-genome interactions, which play a crucial role in vector transduction efficiency and host adaptation. An assessment of past and present clinical trials as well as future directions is provided to illustrate the field's trajectory. Finally, another unique milestone in AAV research is also reported; namely, a pool of AAV libraries has been successfully administered to human decedents and analyzed, representing a transformative step in AAV evolution and selection for human applications. These studies should pave the way for more refined AAV vector optimization, accelerating the development of next-generation gene therapies with enhanced clinical translatability, potentially accelerating the gene therapy revolution.
Collapse
Affiliation(s)
- Lester Suarez-Amaran
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA; Orthopaedic Research Center, C. Wayne McIlwraith Translational Medicine Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA
| | - Liujiang Song
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA
| | - Anna P Tretiakova
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA
| | - Sheila A Mikhail
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA
| | - Richard Jude Samulski
- M34, Inc., 870 Martin Luther King Jr. Boulevard, Chapel Hill, NC 27514-2600, USA; Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
5
|
Liu Z, Zhang H, Jia H, Wang H, Huang Z, Tang Y, Wang Z, Hu J, Zhao X, Li T, Sun X. The clinical safety landscape for ocular AAV gene therapies: A systematic review and meta-analysis. iScience 2025; 28:112265. [PMID: 40248125 PMCID: PMC12005934 DOI: 10.1016/j.isci.2025.112265] [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: 10/14/2024] [Revised: 01/23/2025] [Accepted: 03/18/2025] [Indexed: 04/19/2025] Open
Abstract
Adeno-associated virus (AAV) gene therapy is a promising approach for treating ocular monogenic or acquired diseases, though immunogenicity and safety remain critical considerations. We conducted a systematic review of 120 trials and 32 publications to assess immune responses across different delivery routes. Intravitreal administration was associated with higher rates of anterior uveitis (43.06% vs. 10.22%) and intermediate/posterior uveitis (40.36% vs. 6.18%) compared to subretinal delivery. Engineered AAV capsids, used exclusively in intravitreal studies, showed no significant difference in either type of uveitis incidence compared to natural serotypes. Prophylactic immunosuppression (PI) did not affect ocular or systemic immune responses in subretinal delivery, but significantly reduced systemic immune responses in intravitreal administration. These findings underscore the potential of PI to mitigate systemic immune responses in intravitreal AAV therapy. This review should help guide the choice of routes of administration and immunosuppression strategies, and highlights current trends in ocular AAV gene therapy.
Collapse
Affiliation(s)
- Zishi Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Ophthalmic Diseases, Shanghai, China
| | - Haoliang Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Ophthalmic Diseases, Shanghai, China
| | - Huixun Jia
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Ophthalmic Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
- Shanghai Gene Therapy Center, Shanghai, China
| | - Hong Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Ophthalmic Diseases, Shanghai, China
| | - Zhonghe Huang
- Qingdao University School of Mathematics and Statistics, Qingdao, China
| | - Yuhao Tang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Ophthalmic Diseases, Shanghai, China
| | - Zilin Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Ophthalmic Diseases, Shanghai, China
| | - Jing Hu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Ophthalmic Diseases, Shanghai, China
| | - Xiaohuan Zhao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Ophthalmic Diseases, Shanghai, China
| | - Tong Li
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Ophthalmic Diseases, Shanghai, China
- Shanghai Gene Therapy Center, Shanghai, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Ophthalmic Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
- Shanghai Gene Therapy Center, Shanghai, China
- Shanghai Key Laboratory of Fundus Diseases, Shanghai, China
| |
Collapse
|
6
|
Xia K, Liu S, Wu Z, Jiang JH. Research Status and Applications of Adeno-Associated Virus. Chembiochem 2025; 26:e202400856. [PMID: 39724465 DOI: 10.1002/cbic.202400856] [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: 10/15/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Adeno-associated virus (AAV) has emerged as a powerful and effective tool for the delivery of exogenous genes into various cells or tissues. To improve the gene delivery efficiency, as well as the safety and specificity of AAV's cell-targeting capabilities, extensive investigations have been conducted into its molecular biological characteristics, including capsid structure, cellular tropism, and the mechanisms underlying its entry, replication, DNA packaging, and capsid assembly. Significant differences exist between human and non-human primate AAVs regarding tissue targeting and transduction efficiency. These differences are primarily attributed to the amino acid sequences of AAV capsid proteins, the structural characteristics of these proteins, and the interactions of AAV with surface factors on host cells, such as cell surface receptors, signaling molecules, and associated proteins. This review primarily focuses on several key aspects of AAV, including its genome, coat proteins and their structures, genome replication, virus assembly, and the role of helper viruses. Additionally, it examines the utilization of recombinant adeno-associated viruses (rAAV), detailing their production methods, mechanisms of cell entry and trafficking, and various serotypes. The review further interprets the role of rAAV by analyzing its current applications in research and therapy.
Collapse
Affiliation(s)
- Ke Xia
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Affiliated Hospital of Hunan university, School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China
| | - Shuangling Liu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Affiliated Hospital of Hunan university, School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China
| | - Zhenkun Wu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Affiliated Hospital of Hunan university, School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China
| | - Jian-Hui Jiang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Affiliated Hospital of Hunan university, School of Biomedical Sciences, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
7
|
Tan F, Dong Y, Qi J, Yu W, Chai R. Artificial Intelligence-Based Approaches for AAV Vector Engineering. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411062. [PMID: 39932449 PMCID: PMC11884542 DOI: 10.1002/advs.202411062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/31/2024] [Indexed: 03/08/2025]
Abstract
Adeno-associated virus (AAV) has emerged as a leading vector for gene therapy due to its broad host range, low pathogenicity, and ability to facilitate long-term gene expression. However, AAV vectors face limitations, including immunogenicity and insufficient targeting specificity. To enhance the efficacy of gene therapy, researchers have been modifying the AAV vector using various methods. Traditional experimental approaches for optimizing AAV vector are often time-consuming, resource-intensive, and difficult to replicate. The advancement of artificial intelligence (AI), particularly machine learning, offers significant potential to accelerate capsid optimization while reducing development time and manufacturing costs. This review compares traditional and AI-based methods of AAV vector engineering and highlights recent research in AAV engineering using AI algorithms.
Collapse
Affiliation(s)
- Fangzhi Tan
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologySchool of MedicineAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjing210096China
| | - Yue Dong
- Immunowake, Inc.Shanghai201210China
| | - Jieyu Qi
- Department of NeurologyAerospace Center HospitalSchool of Life ScienceBeijing Institute of TechnologyBeijing100081China
- State Key Laboratory of Hearing and Balance ScienceBeijing Institute of TechnologyBeijing100081China
- School of Medical EngineeringAffiliated Zhuhai People's HospitalBeijing Institute of TechnologyZhuhai519088China
- Advanced Technology Research InstituteBeijing Institute of TechnologyJinan250300China
| | - Wenwu Yu
- School of MathematicsSoutheast UniversityNanjing210096China
| | - Renjie Chai
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologySchool of MedicineAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjing210096China
- Department of NeurologyAerospace Center HospitalSchool of Life ScienceBeijing Institute of TechnologyBeijing100081China
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantong226001China
- Department of Otolaryngology Head and Neck SurgerySichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaChengdu610072China
- Southeast University Shenzhen Research InstituteShenzhen518063China
| |
Collapse
|
8
|
Wang S, Xiao L. Progress in AAV-Mediated In Vivo Gene Therapy and Its Applications in Central Nervous System Diseases. Int J Mol Sci 2025; 26:2213. [PMID: 40076831 PMCID: PMC11899905 DOI: 10.3390/ijms26052213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
As the blood-brain barrier (BBB) prevents molecules from accessing the central nervous system (CNS), the traditional systemic delivery of chemical drugs limits the development of neurological drugs. However, in recent years, innovative therapeutic strategies have tried to bypass the restriction of traditional drug delivery methods. In vivo gene therapy refers to emerging biopharma vectors that carry the specific genes and target and infect specific tissues; these infected cells and tissues then undergo fundamental changes at the genetic level and produce therapeutic proteins or substances, thus providing therapeutic benefits. Clinical and preclinical trials mainly utilize adeno-associated viruses (AAVs), lentiviruses (LVs), and other viruses as gene vectors for disease investigation. Although LVs have a higher gene-carrying capacity, the vector of choice for many neurological diseases is the AAV vector due to its safety and long-term transgene expression in neurons. Here, we review the basic biology of AAVs and summarize some key issues in recombinant AAV (rAAV) engineering in gene therapy research; then, we summarize recent clinical trials using rAAV treatment for neurological diseases and provide translational perspectives and future challenges on target selection.
Collapse
Affiliation(s)
- Shuming Wang
- Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China;
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Lin Xiao
- Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China;
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
9
|
Hinsch VG, Boye SL, Boye SE. A Comprehensive Review of Clinically Applied Adeno-Associated Virus-Based Gene Therapies for Ocular Disease. Hum Gene Ther 2025. [PMID: 39989340 DOI: 10.1089/hum.2024.252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025] Open
Abstract
The eye is an ideal target for gene therapy due its accessibility, immune privilege, small size, and compartmentalization. Adeno-associated virus (AAV) is the gold standard vector for gene delivery and can be injected via multiple routes of administration to target different parts of this organ. The approval of Luxturna™, a subretinally delivered gene therapy for RPE65-associated Leber's congenital amaurosis, and the large number of successful proof of concept studies performed in animal models injected great momentum into the pursuit of additional AAV-based gene therapies for the treatment of retinal disease. This review provides a comprehensive summary of all subretinally, intravitreally, and suprachoroidally delivered AAV-based ocular gene therapies that have progressed to clinical stage. Attention is given to primary (safety) and secondary (efficacy) outcomes, or lack thereof. Lessons learned and future directions are addressed, both of which point to optimism that the ocular gene therapy field is poised for continued momentum and additional regulatory approvals.
Collapse
Affiliation(s)
- Valerie G Hinsch
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Sanford L Boye
- Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, Florida, USA
| | - Shannon E Boye
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
10
|
Aliev TI, Yudkin DV. AAV-based vectors for human diseases modeling in laboratory animals. Front Med (Lausanne) 2025; 11:1499605. [PMID: 40007819 PMCID: PMC11859266 DOI: 10.3389/fmed.2024.1499605] [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: 09/26/2024] [Accepted: 11/26/2024] [Indexed: 02/27/2025] Open
Abstract
The development of therapeutic drugs and vaccines requires the availability of appropriate model animals that replicate the pathogenesis of human diseases. Both native and transgenic animals can be utilized as models. The advantage of transgenic animals lies in their ability to simulate specific properties desired by researchers. However, there is often a need for the rapid production of transgenic animal models, especially in situations like a pandemic, as was evident during COVID-19. An important tool for transgenesis is the adeno-associated virus. The genome of adeno-associated virus serves as a convenient expression cassette for delivering various DNA constructs into cells, and this method has proven effective in practice. This review analyzes the features of the adeno-associated virus genome that make it an advantageous vector for transgenesis. Additionally, examples of utilizing adeno-associated viral vectors to create animal models for hereditary, oncological, and viral human diseases are provided.
Collapse
Affiliation(s)
- Timur I. Aliev
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Dmitry V. Yudkin
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| |
Collapse
|
11
|
Wei L, Yu P, Wang H, Liu J. Adeno-associated viral vectors deliver gene vaccines. Eur J Med Chem 2025; 281:117010. [PMID: 39488197 DOI: 10.1016/j.ejmech.2024.117010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
Adeno-associated viruses (AAVs) are leading platforms for in vivo delivery of gene therapies, with six licensed AAV-based therapeutics attributed to their non-pathogenic nature, low immunogenicity, and high efficiency. In the realm of gene-based vaccines, one of the most vital therapeutic areas, AAVs are also emerging as promising delivery tools. We scrutinized AAVs, focusing on their virological properties, as well as bioengineering and chemical modifications to demonstrate their significant potential in gene vaccine delivery, and detailing the preparation of AAV particles. Additionally, we summarized the use of AAV vectors in vaccines for both infectious and non-infectious diseases, such as influenza, COVID-19, Alzheimer's disease, and cancer. Furthermore, this review, along with the latest clinical trial updates, provides a comprehensive overview of studies on the potential of using AAV vectors for gene vaccine delivery. It aims to deepen our understanding of the challenges and limitations in nucleic acid delivery and pave the way for future clinical success.
Collapse
Affiliation(s)
- Lai Wei
- College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Peng Yu
- College of Biotechnology, Tianjin University of Science & Technology, 300457 Tianjin, China
| | - Haomeng Wang
- CanSino (Shanghai) Biological Research Co., Ltd, 201208, Shanghai, China.
| | - Jiang Liu
- Rosalind Franklin Institute, Harwell Campus, OX11 0QS, Oxford, United Kingdom; Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT, Oxford, United Kingdom.
| |
Collapse
|
12
|
Sorroza-Martinez L, Pelletier M, Guay D, Gaillet B. Recent Advances in Therapeutics and Manufacturing Processes of Recombinant Adeno-Associated Virus for the Treatment of Lung Diseases. Curr Gene Ther 2025; 25:237-256. [PMID: 39225214 DOI: 10.2174/0115665232294935240826061311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/01/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Developing delivery vectors capable of transducing genetic material across the lung epithelia and mucus barrier is a major challenge and of great interest to enable gene therapies to treat pulmonary diseases. Recombinant Adeno-associated Viruses (rAAVs) have emerged as attractive candidates among viral and non-viral vectors due to their broad tissue tropism, ability to transduce dividing and quiescent cells, and their safety profile in current human applications. While rAAVs have demonstrated safety in earlier clinical trials for lung disease applications, there are still some limitations regarding rAAV-transgene delivery in pulmonary cells. Thus, further improvements in rAAV engineering are needed to enhance the effectiveness of rAAV-based therapies for lung diseases. Such therapies could benefit patients with chronic lung diseases, such as asthma, chronic obstructive pulmonary disease, pulmonary hypertension, and cystic fibrosis, among others, by regulating hereditary gene mutations or acquired gene deregulations causing these conditions. Alongside therapeutic development, advances in the rAAV production process are essential to meet increasing production demands, while reducing manufacturing costs. This review discusses current challenges and recent advances in the field of rAAV engineering and manufacturing to encourage the clinical development of new pulmonary gene therapy treatments.
Collapse
Affiliation(s)
- Luis Sorroza-Martinez
- Département de génie chimique, Université Laval, Room #3570, 1065 avenue de la Médecine, Pavillon Adrien- Pouliot, Québec, QC, G1V 0A6, Canada
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC, G1P 4S6, Canada
| | - Mia Pelletier
- Département de génie chimique, Université Laval, Room #3570, 1065 avenue de la Médecine, Pavillon Adrien- Pouliot, Québec, QC, G1V 0A6, Canada
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC, G1P 4S6, Canada
| | - David Guay
- Département de génie chimique, Université Laval, Room #3570, 1065 avenue de la Médecine, Pavillon Adrien- Pouliot, Québec, QC, G1V 0A6, Canada
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC, G1P 4S6, Canada
| | - Bruno Gaillet
- Département de génie chimique, Université Laval, Room #3570, 1065 avenue de la Médecine, Pavillon Adrien- Pouliot, Québec, QC, G1V 0A6, Canada
| |
Collapse
|
13
|
Lim Y, Campochiaro PA, Green JJ. Suprachoroidal Delivery of Viral and Nonviral Vectors for Treatment of Retinal and Choroidal Vascular Diseases. Am J Ophthalmol 2024:S0002-9394(24)00571-3. [PMID: 39716546 DOI: 10.1016/j.ajo.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/25/2024]
Abstract
PURPOSE Current treatments for retinal and choroidal neovascular diseases suffer from insufficient durability, including anti-vascular endothelial growth factor-A agents. It is, therefore, of interest to explore alternative methods that could allow for robust improvement in visual acuity with fewer injections required. DESIGN Literature review. RESULTS Among various preclinical and clinical studies in the literature, a promising approach is the use of suprachoroidal injection with viral and nonviral gene delivery vectors. Compared with other ocular injection methods, suprachoroidal injection has demonstrated wide biodistribution of injected agents and safety as an outpatient procedure. In terms of viral vectors, suprachoroidal injection of an adeno-associated virus 8 vector expressing an anti-vascular endothelial growth factor-A antibody fragment has shown an excellent safety profile and evidence of biological activity. In terms of nonviral vectors, lipid nanoparticles and polymeric nanoparticles both demonstrate strong promise for ocular gene therapy in large animal models. In particular, biodegradable poly(β-amino ester) nanoparticles show excellent biodistribution, safety, and efficacy for gene therapy via the suprachoroidal route. Nonviral nanoparticle approaches can have notable advantages over viral vectors in terms of carrying capacity, redosability, and manufacturing costs. An advantage of gene therapy is that once a delivery vector has been optimized, genetic cargos can be readily tailored without changing the safety, efficacy, and pharmacokinetic properties of the delivery vector. CONCLUSIONS This review highlights recent progress that has been made and compares viral and nonviral suprachoroidal gene delivery for the treatment of retinal and choroidal vascular diseases. Suprachoroidal gene therapy is an emerging biotechnology that holds substantial potential to make a translational impact in treating these diseases.
Collapse
Affiliation(s)
- Yeongseo Lim
- From the Department of Biomedical Engineering (Y.L., J.J.G.), Johns Hopkins University, Baltimore, Maryland, USA; Translational Tissue Engineering Center (Y.L., J.J.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Johns Hopkins Translational ImmunoEngineering Center (Y.L., J.J.G.), Johns Hopkins University, Baltimore, Maryland, USA
| | - Peter A Campochiaro
- Department of Ophthalmology (P.A.C., J.J.G.), Johns Hopkins University, Baltimore, Maryland, USA; Department of Neuroscience (P.A.C.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Jordan J Green
- From the Department of Biomedical Engineering (Y.L., J.J.G.), Johns Hopkins University, Baltimore, Maryland, USA; Translational Tissue Engineering Center (Y.L., J.J.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Johns Hopkins Translational ImmunoEngineering Center (Y.L., J.J.G.), Johns Hopkins University, Baltimore, Maryland, USA; Department of Ophthalmology (P.A.C., J.J.G.), Johns Hopkins University, Baltimore, Maryland, USA; Departments of Chemical & Biomolecular Engineering and Materials Science & Engineering (J.J.G.), Johns Hopkins University, Baltimore, Maryland, USA; Departments of Neurosurgery and Oncology (J.J.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Institute for Nanobiotechnology, Johns Hopkins University (J.J.G.), Baltimore, Maryland, USA..
| |
Collapse
|
14
|
Sandoval IM, Kelley CM, Bernal-Conde LD, Steece-Collier K, Marmion DJ, Davidsson M, Crosson SM, Boye SL, Boye SE, Manfredsson FP. Engineered AAV capsid transport mutants overcome transduction deficiencies in the aged CNS. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102332. [PMID: 39445231 PMCID: PMC11497394 DOI: 10.1016/j.omtn.2024.102332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 09/05/2024] [Indexed: 10/25/2024]
Abstract
Adeno-associated virus (AAV)-based gene therapy has enjoyed great successes over the past decade, with Food and Drug Administration-approved therapeutics and a robust clinical pipeline. Nonetheless, barriers to successful translation remain. For example, advanced age is associated with impaired brain transduction, with the diminution of infectivity depending on anatomical region and capsid. Given that CNS gene transfer is often associated with neurodegenerative diseases where age is the chief risk factor, we sought to better understand the causes of this impediment. We assessed two AAV variants hypothesized to overcome factors negatively impacting transduction in the aged brain; specifically, changes in extracellular and cell-surface glycans, and intracellular transport. We evaluated a heparin sulfate proteoglycan null variant with or without mutations enhancing intracellular transport. Vectors were injected into the striatum of young adult or aged rats to address whether improving extracellular diffusion, removing glycan receptor dependence, or improving intracellular transport are important factors in transducing the aged brain. We found that, regardless of the viral capsid, there was a reduction in many of our metrics of transduction in the aged brain. However, the transport mutant was less sensitive to age, suggesting that changes in the cellular transport of AAV capsids are a key factor in age-related transduction deficiency.
Collapse
Affiliation(s)
- Ivette M. Sandoval
- Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Christy M. Kelley
- Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Luis Daniel Bernal-Conde
- Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, Michigan State University College of Human Medicine, Grand Rapids, MI 49506, USA
| | - David J. Marmion
- Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Marcus Davidsson
- Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Sean M. Crosson
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | - Sanford L. Boye
- Powell Gene Therapy Center, Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | - Shannon E. Boye
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | - Fredric P. Manfredsson
- Parkinson’s Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| |
Collapse
|
15
|
Carneiro AD, Schaffer DV. Engineering novel adeno-associated viruses (AAVs) for improved delivery in the nervous system. Curr Opin Chem Biol 2024; 83:102532. [PMID: 39342684 DOI: 10.1016/j.cbpa.2024.102532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024]
Abstract
Harnessing adeno-associated virus (AAV) vectors for therapeutic gene delivery has emerged as a progressively promising strategy to treat disorders of both the central nervous system (CNS) and peripheral nervous system (PNS), and there are many ongoing clinical trials. However, unique physiological and molecular characteristics of the CNS and PNS pose obstacles to efficient vector delivery, ranging from the blood-brain barrier to the diverse nature of nervous system disorders. Engineering novel AAV capsids may help overcome these ongoing challenges and maximize therapeutic transgene delivery. This article discusses strategies for innovative AAV capsid development, highlighting recent advances. Notably, advances in next generation sequencing and machine learning have sparked new approaches for capsid investigation and engineering. Furthermore, we outline future directions and additional challenges in AAV-mediated gene therapy in the CNS and PNS.
Collapse
Affiliation(s)
- Ana D Carneiro
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA; Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA; California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
16
|
Huang W, Bates R, Appana B, Mohammed T, Cao L. Development of an adipose-tropic AAV capsid ablating liver tropism. iScience 2024; 27:110930. [PMID: 39398244 PMCID: PMC11467673 DOI: 10.1016/j.isci.2024.110930] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/08/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
AAV vectors are mainstream delivery platforms in gene therapy, yet AAV-mediated gene transfer to adipose tissue is underdeveloped due to low efficiency of natural AAVs. We previously demonstrated that an engineered capsid Rec2 displayed improved adipo-tropism but with the caveat of liver transduction. To generate highly adipo-tropic capsid, we modified Rec2 capsid by site-specific mutagenesis and found the variant V7 with F503Y, Y708D and K709I substitution to harbor highly selective adipo-tropism while diminishing liver transduction. Intraperitoneal injection favored transduction to visceral fat while intravenous administration favored subcutaneous fat. Intraperitoneal administration of V7 vector harboring human leptin and adiponectin as single transcript normalized the metabolic dysfunction of ob/ob mice at a low dose. Moreover, introducing the same mutagenesis to AAV8 capsid diminished liver transduction suggesting F503, Y708 and K709 critical for liver transduction. The Rec2.V7 vector may provide a powerful tool for basic research and potent vehicle for adipose-targeting gene therapy.
Collapse
Affiliation(s)
- Wei Huang
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Rhiannon Bates
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Bhavya Appana
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Tawfiq Mohammed
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Lei Cao
- Department of Cancer Biology & Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
17
|
He B, Wilson B, Chen SH, Sharma K, Scappini E, Cook M, Petrovich R, Martin NP. Molecular Engineering of Virus Tropism. Int J Mol Sci 2024; 25:11094. [PMID: 39456875 PMCID: PMC11508178 DOI: 10.3390/ijms252011094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Engineered viral vectors designed to deliver genetic material to specific targets offer significant potential for disease treatment, safer vaccine development, and the creation of novel biochemical research tools. Viral tropism, the specificity of a virus for infecting a particular host, is often modified in recombinant viruses to achieve precise delivery, minimize off-target effects, enhance transduction efficiency, and improve safety. Key factors influencing tropism include surface protein interactions between the virus and host-cell, the availability of host-cell machinery for viral replication, and the host immune response. This review explores current strategies for modifying the tropism of recombinant viruses by altering their surface proteins. We provide an overview of recent advancements in targeting non-enveloped viruses (adenovirus and adeno-associated virus) and enveloped viruses (retro/lentivirus, Rabies, Vesicular Stomatitis Virus, and Herpesvirus) to specific cell types. Additionally, we discuss approaches, such as rational design, directed evolution, and in silico and machine learning-based methods, for generating novel AAV variants with the desired tropism and the use of chimeric envelope proteins for pseudotyping enveloped viruses. Finally, we highlight the applications of these advancements and discuss the challenges and future directions in engineering viral tropism.
Collapse
Affiliation(s)
- Bo He
- Viral Vector Core, Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (B.H.); (B.W.); (S.-H.C.)
| | - Belinda Wilson
- Viral Vector Core, Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (B.H.); (B.W.); (S.-H.C.)
| | - Shih-Heng Chen
- Viral Vector Core, Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (B.H.); (B.W.); (S.-H.C.)
| | - Kedar Sharma
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (K.S.); (M.C.); (R.P.)
| | - Erica Scappini
- Fluorescent Microscopy and Imaging Center, Molecular and Cellular Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA;
| | - Molly Cook
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (K.S.); (M.C.); (R.P.)
| | - Robert Petrovich
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (K.S.); (M.C.); (R.P.)
| | - Negin P. Martin
- Viral Vector Core, Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; (B.H.); (B.W.); (S.-H.C.)
| |
Collapse
|
18
|
Demircan MB, Zinser LJ, Michels A, Guaza-Lasheras M, John F, Gorol JM, Theuerkauf SA, Günther DM, Grimm D, Greten FR, Chlanda P, Thalheimer FB, Buchholz CJ. T-cell specific in vivo gene delivery with DART-AAVs targeted to CD8. Mol Ther 2024; 32:3470-3484. [PMID: 39113357 PMCID: PMC11489536 DOI: 10.1016/j.ymthe.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/12/2024] [Accepted: 08/02/2024] [Indexed: 08/26/2024] Open
Abstract
One of the biggest challenges for in vivo gene therapy are vectors mediating highly selective gene transfer into a defined population of therapy-relevant cells. Here we present DARPin-targeted AAVs (DART-AAVs) displaying DARPins specific for human and murine CD8. Insertion of DARPins into the GH2/GH3 loop of the capsid protein 1 (VP1) of AAV2 and AAV6 resulted in high selectivity for CD8-positive T cells with unimpaired gene delivery activity. Remarkably, the capsid core structure was unaltered with protruding DARPins detectable. In complex primary cell mixtures, including donor blood or systemic injections into mice, the CD8-targeted AAVs were by far superior to unmodified AAV2 and AAV6 in terms of selectivity, target cell viability, and gene transfer rates. In vivo, up to 80% of activated CD8+ T cells were hit upon a single vector injection into conditioned humanized or immunocompetent mice. While gene transfer rates decreased significantly under non-activated conditions, genomic modification selectively in CD8+ T cells was still detectable upon Cre delivery into indicator mice. In both mouse models, selectivity for CD8+ T cells was close to absolute with exceptional detargeting from liver. The CD8-AAVs described here expand strategies for immunological research and in vivo gene therapy options.
Collapse
Affiliation(s)
| | - Luca J Zinser
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany
| | - Alexander Michels
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany
| | - Mar Guaza-Lasheras
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany
| | - Fabian John
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany; Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt, Germany
| | - Johanna M Gorol
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany; Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt, Germany; Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt, Germany
| | - Samuel A Theuerkauf
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany
| | - Dorothee M Günther
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany; Ernst Strüngmann Institute for Neuroscience in Cooperation with Max Planck Society, 60528 Frankfurt, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty and Faculty of Engineering Sciences, Heidelberg University, BioQuant, 69120 Heidelberg, Germany
| | - Florian R Greten
- Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt, Germany; Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt, Germany
| | - Petr Chlanda
- Schaller Research Groups, Department of Infectious Diseases/Virology, Medical Faculty, Heidelberg University, BioQuant, 69120 Heidelberg, Germany
| | - Frederic B Thalheimer
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany; HZG Hematology, Cell and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany
| | - Christian J Buchholz
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany; Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt, Germany.
| |
Collapse
|
19
|
Wang H, Li R, Sadekar S, Kamath AV, Shen BQ. A novel approach to quantitate biodistribution and transduction of adeno-associated virus gene therapy using radiolabeled AAV vectors in mice. Mol Ther Methods Clin Dev 2024; 32:101326. [PMID: 39286334 PMCID: PMC11404148 DOI: 10.1016/j.omtm.2024.101326] [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: 03/21/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024]
Abstract
An understanding of recombinant adeno-associated virus (AAV) biodistribution profiles is an important element of a preclinical development program. Here, we have developed a radiolabeling strategy utilizing the co-delivery of 125I (non-residualizing) and 111In (residualizing) radionuclide-conjugated AAVs to provide a detailed distribution quantification at tissue level delineating between the cellular internalized AAV (degraded, 111In-125I) and AAV remaining in the extracellular matrix (intact, 125I). This labeling method has been successfully applied to AAV9 and AAV-PHP.eB as tool molecules without altering the physical properties and biological activities of the AAVs. Upon labeling with either of the radioactive probes, these molecules were systemically injected into C57BL/6 mice. The biodistribution results indicate that AAVs, with a fast distribution profile, were mainly located in the extracellular matrix of highly perfused organs such as liver and spleen at early time points, leading to a difference between capsid quantification and vector genome quantification. The results suggest that the 125I-AAV/111In-AAV co-delivery approach offers a robust and efficient analytical strategy to investigate the detailed tissue distribution of AAV vectors, including both vector genome and protein capsids. This novel method has the potential to be applied to capsid optimization, selection, and lead candidate development.
Collapse
Affiliation(s)
- Hongzhi Wang
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ran Li
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Shraddha Sadekar
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Amrita V Kamath
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ben-Quan Shen
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
20
|
Araujo AE, Bentler M, Perez Garmendia X, Kaleem A, Fabian C, Morgan M, Hacker UT, Büning H. Adeno-Associated Virus Vectors-a Target of Cellular and Humoral Immunity-are Expanding Their Reach Toward Hematopoietic Stem Cell Modification and Immunotherapies. Hum Gene Ther 2024; 35:586-603. [PMID: 39193633 DOI: 10.1089/hum.2024.114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024] Open
Abstract
All current market-approved gene therapy medical products for in vivo gene therapy of monogenic diseases rely on adeno-associated virus (AAV) vectors. Advances in gene editing technologies and vector engineering have expanded the spectrum of target cells and, thus, diseases that can be addressed. Consequently, AAV vectors are now being explored to modify cells of the hematopoietic system, including hematopoietic stem and progenitor cells (HSPCs), to develop novel strategies to treat monogenic diseases, but also to generate cell- and vaccine-based immunotherapies. However, the cell types that represent important new targets for the AAV vector system are centrally involved in immune responses against the vector and its transgene product as discussed briefly in the first part of this review. In the second part, studies exploring AAV vectors for genetic engineering of HSPCs, T and B lymphocytes, and beyond are presented.
Collapse
Affiliation(s)
- Angela E Araujo
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Martin Bentler
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | | - Asma Kaleem
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Claire Fabian
- Laboratory for Vector based immunotherapy, Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
- Medical Department II, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Cancer Center Central Germany, Leipzig, Germany
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Ulrich T Hacker
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Laboratory for Vector based immunotherapy, Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
- Medical Department II, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Cancer Center Central Germany, Leipzig, Germany
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| |
Collapse
|
21
|
Pham Q, Glicksman J, Chatterjee A. Chemical approaches to probe and engineer AAV vectors. NANOSCALE 2024; 16:13820-13833. [PMID: 38978480 PMCID: PMC11271820 DOI: 10.1039/d4nr01300j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/14/2024] [Indexed: 07/10/2024]
Abstract
Adeno-associated virus (AAV) has emerged as the most promising vector for in vivo human gene therapy, with several therapeutic approvals in the last few years and countless more under development. Underlying this remarkable success are several attractive features that AAV offers, including lack of pathogenicity, low immunogenicity, long-term gene expression without genomic integration, the ability to infect both dividing and non-dividing cells, etc. However, the commonly used wild-type AAV capsids in therapeutic development present significant challenges, including inadequate tissue specificity and the need for large doses to attain therapeutic effectiveness, raising safety concerns. Additionally, significant preexisting adaptive immunity against most natural capsids, and the development of such anti-capsid immunity after the first treatment, represent major challenges. Strategies to engineer the AAV capsid are critically needed to address these challenges and unlock the full promise of AAV gene therapy. Chemical modification of the AAV capsid has recently emerged as a powerful new approach to engineer its properties. Unlike genetic strategies, which can be more disruptive to the delicate capsid assembly and packaging processes, "late-stage" chemical modification of the assembled capsid-whether at natural amino acid residues or site-specifically installed noncanonical amino acid residues-often enables a versatile approach to introducing new properties to the capsid. This review summarizes the significant recent progress in AAV capsid engineering strategies, with a particular focus on chemical modifications in advancing the next generation of AAV-based gene therapies.
Collapse
Affiliation(s)
- Quan Pham
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA 02467, USA.
| | - Jake Glicksman
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA 02467, USA.
| | - Abhishek Chatterjee
- Department of Chemistry, Boston College, 2609 Beacon Street, Chestnut Hill, MA 02467, USA.
| |
Collapse
|
22
|
Ling C, Yu C, Wang C, Yang M, Yang H, Yang K, He Y, Shen Y, Tang S, Yu X, Zhou Z, Zhou S, Zhou J, Zhu L, Li J. rAAV capsid mutants eliminate leaky expression from DNA donor template for homologous recombination. Nucleic Acids Res 2024; 52:6518-6531. [PMID: 38783157 PMCID: PMC11194064 DOI: 10.1093/nar/gkae401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 04/28/2024] [Accepted: 05/04/2024] [Indexed: 05/25/2024] Open
Abstract
Precise genomic editing through the combination of CRISPR/Cas systems and recombinant adeno-associated virus (rAAV)-delivered homology directed repair (HDR) donor templates represents a powerful approach. However, the challenge of effectively suppressing leaky transcription from the rAAV vector, a phenomenon associated to cytotoxicity, persists. In this study, we demonstrated substantial promoter activities of various homology arms and inverted terminal repeats (ITR). To address this issue, we identified a novel rAAV variant, Y704T, which not only yields high-vector quantities but also effectively suppresses in cis mRNA transcription driven by a robust promoter. The Y704T variant maintains normal functionality in receptor interaction, intracellular trafficking, nuclear entry, uncoating, and second-strand synthesis, while specifically exhibiting defects in transcription. Importantly, this inhibitory effect is found to be independent of ITR, promoter types, and RNA polymerases. Mechanistic studies unveiled the involvement of Valosin Containing Protein (VCP/p97) in capsid-mediated transcription repression. Remarkably, the Y704T variant delivers HDR donor templates without compromising DNA replication ability and homologous recombination efficiency. In summary, our findings enhance the understanding of capsid-regulated transcription and introduce novel avenues for the application of the rAAV-CRISPR/Cas9 system in human gene therapy.
Collapse
Affiliation(s)
- Chen Ling
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Chenghui Yu
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Cong Wang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Ming Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Hengbin Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Keying Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Yun He
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Yajie Shen
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Huashan Hospital, Shanghai Engineering Research Center of Industrial Microorganisms, MOE Engineering Research Center of Gene Technology, Fudan University, Shanghai 200438, China
| | - Shiyi Tang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiaomin Yu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zhengjun Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shaolai Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Liqing Zhu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Department of Clinical Laboratory, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Jixi Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Huashan Hospital, Shanghai Engineering Research Center of Industrial Microorganisms, MOE Engineering Research Center of Gene Technology, Fudan University, Shanghai 200438, China
| |
Collapse
|
23
|
Wang YC, Mao XT, Sun C, Wang YH, Zheng YZ, Xiong SH, Liu MY, Mao SH, Wang QW, Ma GX, Wu D, Li ZS, Chen JM, Zou WB, Liao Z. Pancreas-directed AAV8 -hSPINK1 gene therapy safely and effectively protects against pancreatitis in mice. Gut 2024; 73:1142-1155. [PMID: 38553043 DOI: 10.1136/gutjnl-2023-330788] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 03/19/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVE Currently, there is no cure for chronic pancreatitis (CP). Germline loss-of-function variants in SPINK1 (encoding trypsin inhibitor) are common in patients with CP and are associated with acute attacks and progression of the disease. This preclinical study was conducted to explore the potential of adeno-associated virus type 8 (AAV8)-mediated overexpression of human SPINK1 (hSPINK1) for pancreatitis therapy in mice. DESIGN A capsid-optimised AAV8-mediated hSPINK1 expression vector (AAV8-hSPINK1) to target the pancreas was constructed. Mice were treated with AAV8-hSPINK1 by intraperitoneal injection. Pancreatic transduction efficiency and safety of AAV8-hSPINK1 were dynamically evaluated in infected mice. The effectiveness of AAV8-hSPINK1 on pancreatitis prevention and treatment was studied in three mouse models (caerulein-induced pancreatitis, pancreatic duct ligation and Spink1 c.194+2T>C mouse models). RESULTS The constructed AAV8-hSPINK1 vector specifically and safely targeted the pancreas, had low organ tropism for the heart, lungs, spleen, liver and kidneys and had a high transduction efficiency (the optimal expression dose was 2×1011 vg/animal). The expression and efficacy of hSPINK1 peaked at 4 weeks after injection and remained at significant level for up to at least 8 weeks. In all three mouse models, a single dose of AAV8-hSPINK1 before disease onset significantly alleviated the severity of pancreatitis, reduced the progression of fibrosis, decreased the levels of apoptosis and autophagy in the pancreas and accelerated the pancreatitis recovery process. CONCLUSION One-time injection of AAV8-hSPINK1 safely targets the pancreas with high transduction efficiency and effectively ameliorates pancreatitis phenotypes in mice. This approach is promising for the prevention and treatment of CP.
Collapse
Affiliation(s)
- Yuan-Chen Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China
| | - Xiao-Tong Mao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Chang Sun
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Ya-Hui Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Yi-Zhou Zheng
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Si-Huai Xiong
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Mu-Yun Liu
- Department of Gastroenterology, No. 905 Hospital of PLA Navy Affiliated to Naval Medical University, Shanghai, China
| | - Sheng-Han Mao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Qi-Wen Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Guo-Xiu Ma
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Di Wu
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China
| | - Jian-Min Chen
- Univ Brest, Inserm, EFS, UMR 1078, GGB, Brest, France
| | - Wen-Bin Zou
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China
| | - Zhuan Liao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
- National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China
| |
Collapse
|
24
|
Wang H, Kwak SE, Zheng A, Arias EB, Pan X, Duan D, Cartee GD. Phosphorylation of AS160-serine 704 is not essential for exercise-increase in insulin-stimulated glucose uptake by skeletal muscles from female or male rats. Am J Physiol Endocrinol Metab 2024; 326:E807-E818. [PMID: 38656130 PMCID: PMC11376492 DOI: 10.1152/ajpendo.00010.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/18/2024] [Accepted: 04/07/2024] [Indexed: 04/26/2024]
Abstract
One exercise session can increase subsequent insulin-stimulated glucose uptake (ISGU) by skeletal muscle from rodents and humans of both sexes. We recently found that concurrent mutation of three key sites to prevent their phosphorylation (Ser588, Thr642, and Ser704) on Akt substrate of 160 kDa (AS160; also known as TBC1D4) reduced the magnitude of the enhancement of postexercise ISGU (PEX-ISGU) by muscle from male, but not female rats. However, we did not test the role of individual phosphorylation sites on PEX-ISGU. Accordingly, our current aim was to test whether AS160 Ser704 phosphorylation (pSer704) is required for elevated PEX-ISGU by muscle. AS160-knockout (AS160-KO) rats (female and male) were studied when either in sedentary or 3 h after acute exercise. Adeno-associated virus (AAV) vectors were used to enable muscle expression of wild-type AS160 (AAV-WT-AS160) or AS160 mutated Ser704 to alanine to prevent phosphorylation (AAV-1P-AS160). Paired epitrochlearis muscles from each rat were injected with AAV-WT-AS160 or AAV-1P-AS160. We discovered that regardless of sex 1) AS160 abundance in AS160-KO rats was similar in paired muscles expressing WT-AS160 versus 1P-AS160; 2) muscles from exercised versus sedentary rats had greater ISGU, and PEX-ISGU was slightly greater for muscles expressing 1P-AS160 versus contralateral muscles expressing WT-AS160; and 3) pAS160Thr642 was lower in muscles expressing 1P-AS160 versus paired muscles expressing WT-AS160. These results indicate that pAS160Ser704 was not essential for elevated PEX-ISGU by skeletal muscle from rats of either sex. Furthermore, elimination of the postexercise increase in pAS160Thr642 did not lessen the postexercise effect on ISGU.NEW & NOTEWORTHY The current study evaluated the role of Akt substrate of 160 kDa (AS160) phosphorylation on Ser704 in increased insulin-stimulated glucose uptake by skeletal muscle after exercise. Adeno-associated virus vectors were engineered to express either wild-type-AS160 or AS160 mutated so that it could not be phosphorylated on Ser704 in paired muscles from AS160-knockout rats. The results demonstrated that AS160 phosphorylation on Ser704 was not essential for exercise-induced elevation in insulin-stimulated glucose uptake by rats of either sex.
Collapse
Affiliation(s)
- Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Seong Eun Kwak
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Amy Zheng
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Edward B Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, United States
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, United States
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Chemical and Biomedical Engineering, College of Engineering, University of Missouri, Columbia, Missouri, United States
| | - Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
25
|
Wang JH, Gessler DJ, Zhan W, Gallagher TL, Gao G. Adeno-associated virus as a delivery vector for gene therapy of human diseases. Signal Transduct Target Ther 2024; 9:78. [PMID: 38565561 PMCID: PMC10987683 DOI: 10.1038/s41392-024-01780-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
Adeno-associated virus (AAV) has emerged as a pivotal delivery tool in clinical gene therapy owing to its minimal pathogenicity and ability to establish long-term gene expression in different tissues. Recombinant AAV (rAAV) has been engineered for enhanced specificity and developed as a tool for treating various diseases. However, as rAAV is being more widely used as a therapy, the increased demand has created challenges for the existing manufacturing methods. Seven rAAV-based gene therapy products have received regulatory approval, but there continue to be concerns about safely using high-dose viral therapies in humans, including immune responses and adverse effects such as genotoxicity, hepatotoxicity, thrombotic microangiopathy, and neurotoxicity. In this review, we explore AAV biology with an emphasis on current vector engineering strategies and manufacturing technologies. We discuss how rAAVs are being employed in ongoing clinical trials for ocular, neurological, metabolic, hematological, neuromuscular, and cardiovascular diseases as well as cancers. We outline immune responses triggered by rAAV, address associated side effects, and discuss strategies to mitigate these reactions. We hope that discussing recent advancements and current challenges in the field will be a helpful guide for researchers and clinicians navigating the ever-evolving landscape of rAAV-based gene therapy.
Collapse
Affiliation(s)
- Jiang-Hui Wang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, 3002, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, 3002, Australia
| | - Dominic J Gessler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Neurological Surgery, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Thomas L Gallagher
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
26
|
Li W, Feng SL, Herrschaft L, Samulski RJ, Li C. Rationally engineered novel AAV capsids for intra-articular gene delivery. Mol Ther Methods Clin Dev 2024; 32:101211. [PMID: 38435130 PMCID: PMC10907215 DOI: 10.1016/j.omtm.2024.101211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/12/2024] [Indexed: 03/05/2024]
Abstract
Intra-articular adeno-associated virus (AAV) gene therapy has been explored as a potential strategy for joint diseases. However, concerns of low transduction efficacy, off-target expression, and neutralizing antibodies (Nabs) still need to be addressed. In this study, we demonstrated that AAV6 was the best serotype to transduce joints after screening serotypes 1 to 9. To develop a more effective AAV vector, a set of novel AAV capsids were rationally engineered. The mutant AAV62 created by swapping variable region I (VRI) of AAV2 into AAV6 induced a higher transduction efficiency per AAV genome copy number. To further investigate the roles of specific amino acids in the transduction of AAV62 and AAV6, we found out that AAV6D with the deletion of threonine at residue 265 induced a 2-fold higher transduction than AAV6, while the transduction efficiency from AAV6M with the mutation of alanine to glutamine at residue 263 was 10-fold lower. AAV6D efficiently transduced both synoviocytes and chondrocytes with low AAV genome copy numbers in other tissues and less Nab formation. This study demonstrates that novel AAV mutants with rational engineering may enhance joint transduction after intra-articular administration in mice, with the potential to evade AAV Nabs and minimize off-target effects in the liver.
Collapse
Affiliation(s)
- Wenjun Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Division of Oral and Craniofacial Biomedicine, University of North Carolina Adams School of Dentistry, Chapel Hill, NC, USA
| | - Susi Liu Feng
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lizette Herrschaft
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - R. Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC 27510, USA
| |
Collapse
|
27
|
Mével M, Pichard V, Bouzelha M, Alvarez-Dorta D, Lalys PA, Provost N, Allais M, Mendes A, Landagaray E, Ducloyer JB, Toublanc E, Galy A, Brument N, Lefevre GM, Gouin SG, Isiegas C, Le Meur G, Cronin T, Le Guiner C, Weber M, Moullier P, Ayuso E, Deniaud D, Adjali O. Mannose-coupled AAV2: A second-generation AAV vector for increased retinal gene therapy efficiency. Mol Ther Methods Clin Dev 2024; 32:101187. [PMID: 38327809 PMCID: PMC10847035 DOI: 10.1016/j.omtm.2024.101187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
Inherited retinal diseases are a leading and untreatable cause of blindness and are therefore candidate diseases for gene therapy. Recombinant vectors derived from adeno-associated virus (rAAV) are currently the most promising vehicles for in vivo therapeutic gene delivery to the retina. However, there is a need for novel AAV-based vectors with greater efficacy for ophthalmic applications, as underscored by recent reports of dose-related inflammatory responses in clinical trials of rAAV-based ocular gene therapies. Improved therapeutic efficacy of vectors would allow for decreases in the dose delivered, with consequent reductions in inflammatory reactions. Here, we describe the development of new rAAV vectors using bioconjugation chemistry to modify the rAAV capsid, thereby improving the therapeutic index. Covalent coupling of a mannose ligand, via the formation of a thiourea bond, to the amino groups of the rAAV capsid significantly increases vector transduction efficiency of both rat and nonhuman primate retinas. These optimized rAAV vectors have important implications for the treatment of a wide range of retinal diseases.
Collapse
Affiliation(s)
- Mathieu Mével
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | - Virginie Pichard
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | - Mohammed Bouzelha
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | | | | | - Nathalie Provost
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | - Marine Allais
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | - Alexandra Mendes
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | | | - Jean-Baptiste Ducloyer
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | - Estelle Toublanc
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | - Anne Galy
- Coave Therapeutics (formerly, Horama), 75012 Paris, France
| | - Nicole Brument
- Coave Therapeutics (formerly, Horama), 75012 Paris, France
| | | | | | - Carolina Isiegas
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | - Guylène Le Meur
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | - Thérèse Cronin
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | - Caroline Le Guiner
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | - Michel Weber
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | - Philippe Moullier
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | - Eduard Ayuso
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| | - David Deniaud
- Nantes Université, CNRS, CEISAM UMR 6230, 44000 Nantes, France
| | - Oumeya Adjali
- Nantes Université, CHU de Nantes, INSERM UMR 1089, TaRGeT-Translational Research in Gene Therapy Laboratory, 44200 Nantes, France
| |
Collapse
|
28
|
Lopez-Gordo E, Chamberlain K, Riyad JM, Kohlbrenner E, Weber T. Natural Adeno-Associated Virus Serotypes and Engineered Adeno-Associated Virus Capsid Variants: Tropism Differences and Mechanistic Insights. Viruses 2024; 16:442. [PMID: 38543807 PMCID: PMC10975205 DOI: 10.3390/v16030442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 05/23/2024] Open
Abstract
Today, adeno-associated virus (AAV)-based vectors are arguably the most promising in vivo gene delivery vehicles for durable therapeutic gene expression. Advances in molecular engineering, high-throughput screening platforms, and computational techniques have resulted in a toolbox of capsid variants with enhanced performance over parental serotypes. Despite their considerable promise and emerging clinical success, there are still obstacles hindering their broader use, including limited transduction capabilities, tissue/cell type-specific tropism and penetration into tissues through anatomical barriers, off-target tissue biodistribution, intracellular degradation, immune recognition, and a lack of translatability from preclinical models to clinical settings. Here, we first describe the transduction mechanisms of natural AAV serotypes and explore the current understanding of the systemic and cellular hurdles to efficient transduction. We then outline progress in developing designer AAV capsid variants, highlighting the seminal discoveries of variants which can transduce the central nervous system upon systemic administration, and, to a lesser extent, discuss the targeting of the peripheral nervous system, eye, ear, lung, liver, heart, and skeletal muscle, emphasizing their tissue and cell specificity and translational promise. In particular, we dive deeper into the molecular mechanisms behind their enhanced properties, with a focus on their engagement with host cell receptors previously inaccessible to natural AAV serotypes. Finally, we summarize the main findings of our review and discuss future directions.
Collapse
|
29
|
Biber J, Jabri Y, Glänzer S, Dort A, Hoffelner P, Schmidt CQ, Bludau O, Pauly D, Grosche A. Gliosis-dependent expression of complement factor H truncated variants attenuates retinal neurodegeneration following ischemic injury. J Neuroinflammation 2024; 21:56. [PMID: 38388518 PMCID: PMC10885619 DOI: 10.1186/s12974-024-03045-3] [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: 10/11/2023] [Accepted: 02/14/2024] [Indexed: 02/24/2024] Open
Abstract
Inherited, age-related, and acute retinal diseases are often exacerbated by an aberrant or excessive activity of the complement system. Consequently, cells not directly affected by an acute event or genetic variants may degenerate, resulting in enhanced visual impairment. The therapeutic potential of supplementation of complement factor H (FH), a key regulator of the complement cascade, is therefore particularly promising in the context of retinal diseases caused by complement activation. In this study, we engineered adeno-associated viruses (AAVs) containing sequences of two truncated human FH variants. The expression of these variants was regulated by the glial fibrillary acidic protein (GFAP) promoter, which is selectively active in gliotic Müller cells. Both FH variants consisted of FH domains 19-20, which were connected to domains 1-4 and 1-7, respectively, by a polyglycine linker. These AAVs were intravitreally injected following ischemic injury of C57BL/6J mouse retinas. We observed transgene expression in gliotic Müller cells and to some extent in astrocytes. The expression correlated directly with damage severity. Interventions resulted in decreased complement activation, accelerated normalization of microglia activity and morphological improvements. Reduced levels of C3 transcripts and C3d protein in conjunction with higher transcript levels of inhibitory regulators like Cfi and Cfh, hinted at attenuated complement activity. This study demonstrates the great potential of complement regulatory gene addition therapy. With further in vivo testing it could be applied to treat a wide range of retinal diseases where no causative therapies are available.
Collapse
Affiliation(s)
- Josef Biber
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Yassin Jabri
- Department of Ophthalmology, University Hospital Regensburg, Regensburg, Germany
| | - Sarah Glänzer
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Aaron Dort
- Experimental Ophthalmology, University of Marburg, Marburg, Germany
| | - Patricia Hoffelner
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Christoph Q Schmidt
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Center, Ulm, Germany
- Institute of Pharmacy, Biochemical Pharmacy Group, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Oliver Bludau
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Diana Pauly
- Experimental Ophthalmology, University of Marburg, Marburg, Germany.
| | - Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| |
Collapse
|
30
|
Hassan S, Hsu Y, Thompson JM, Kalmanek E, VandeLune JA, Stanley S, Drack AV. The dose-response relationship of subretinal gene therapy with rAAV2tYF-CB-h RS1 in a mouse model of X-linked retinoschisis. Front Med (Lausanne) 2024; 11:1304819. [PMID: 38414621 PMCID: PMC10898246 DOI: 10.3389/fmed.2024.1304819] [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: 09/30/2023] [Accepted: 01/16/2024] [Indexed: 02/29/2024] Open
Abstract
Purpose X-linked retinoschisis (XLRS), due to loss-of-function mutations in the retinoschisin (RS1) gene, is characterized by a modest to severe decrease in visual acuity. Clinical trials for XLRS utilizing intravitreal (IVT) gene therapy showed ocular inflammation. We conducted a subretinal dose-response preclinical study using rAAV2tYF-CB-hRS1 utilizing the Rs1 knockout (Rs1-KO) mouse to investigate short- and long-term retinal rescue after subretinal gene delivery. Methods Rs1-KO mice were subretinally injected with 2 μL of rAAV2tYF-CB-hRS1 vector with 8E9 viral genomes (vg)/eye, 8E8 vg/eye, 8E7 vg/eye, or sham injection, and compared to untreated eyes. Reconstitution of human RS1 protein was detected using western blotting. Analysis of retinal function by electroretinography (ERG) and structural analysis by optical coherence tomography (OCT) were performed at 1, 2, 3, 5, 7, and 12 months post injection (MPI). Immunohistochemistry (IHC) was performed to evaluate cone rescue on the cellular level. Functional vision was evaluated using a visually guided swim assay (VGSA). Results Western blotting analysis showed human RS1 protein expression in a dose-dependent manner. Quantification of western blotting showed that the RS1 protein expression in mice treated with the 8E8 vg dose was near the wild-type (WT) expression levels. ERG demonstrated dose-dependent effects: At 1 MPI the 8E8 vg dose treated eyes had higher light-adapted (LA) ERG amplitudes in 3.0 flash and 5 Hz flicker compared to untreated (p < 0.0001) and sham-treated eyes (p < 0.0001) which persisted until the 12 MPI endpoint, consistent with improved cone function. ERG b-wave amplitudes were higher in response to dark-adapted (DA) 0.01 dim flash and 3.0 standard combined response (SCR) compared to sham-treated (p < 0.01) and untreated eyes (p < 0.001) which persisted until 3 MPI, suggesting short-term improvement of the rod photoreceptors. All injections, including sham-treated, resulted in a cyst severity score of 1 (no cavities), with significant reductions compared to untreated eyes up to 3 MPI (p < 0.05). The high and low dose groups showed inconsistent ERG improvements, despite reduced cyst severity, emphasizing the dose-dependent nature of gene augmentation's efficacy and the tenuous connection between cyst reduction and ERG improvement. IHC data showed a significant cone rescue in eyes treated with the 8E8 vg dose compared to sham-treated and untreated eyes. VGSA showed better functional vision in 8E8 vg dose treated mice. Eyes treated with the highest dose showed occasional localized degeneration in the outer nuclear layer. Conclusion Our data suggest that a dose of 8E8 vg/eye subretinally improves retinal function and structure in the Rs1-KO mouse. It improves cone function, rod function, and reduces cyst severity. Sham treatment resolves schisis cysts, but 8E8 vg/eye is needed for optimal retinal electrical function rescue. These findings offer a promising path for clinical translation to human trials.
Collapse
Affiliation(s)
- Salma Hassan
- Department of Ophthalmology and Visual Sciences, Institute for Vision Research, and Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Biomedical Science-Cell and Developmental Biology Graduate Program, Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, United States
| | - Ying Hsu
- Department of Ophthalmology and Visual Sciences, Institute for Vision Research, and Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Jacob M Thompson
- Department of Ophthalmology and Visual Sciences, Institute for Vision Research, and Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, United States
| | - Emily Kalmanek
- Department of Ophthalmology and Visual Sciences, Institute for Vision Research, and Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Joel A VandeLune
- Department of Ophthalmology and Visual Sciences, Institute for Vision Research, and Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Sarah Stanley
- Department of Ophthalmology and Visual Sciences, Institute for Vision Research, and Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Arlene V Drack
- Department of Ophthalmology and Visual Sciences, Institute for Vision Research, and Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Biomedical Science-Cell and Developmental Biology Graduate Program, Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, United States
- Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
31
|
Leray A, Lalys PA, Varin J, Bouzelha M, Bourdon A, Alvarez-Dorta D, Pavageau K, Depienne S, Marchand M, Mellet A, Demilly J, Ducloyer JB, Girard T, Fraysse B, Ledevin M, Guilbaud M, Gouin SG, Ayuso E, Adjali O, Larcher T, Cronin T, Le Guiner C, Deniaud D, Mével M. Novel chemical tyrosine functionalization of adeno-associated virus improves gene transfer efficiency in liver and retina. Biomed Pharmacother 2024; 171:116148. [PMID: 38232661 DOI: 10.1016/j.biopha.2024.116148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 01/19/2024] Open
Abstract
Decades of biological and clinical research have led to important advances in recombinant adeno-associated viruses rAAV-based gene therapy gene therapy. However, several challenges must be overcome to fully exploit the potential of rAAV vectors. Innovative approaches to modify viral genome and capsid elements have been used to overcome issues such as unwanted immune responses and off-targeting. While often successful, genetic modification of capsids can drastically reduce vector yield and often fails to produce vectors with properties that translate across different animal species, such as rodents, non-human primates, and humans. Here, we describe a chemical bioconjugation strategy to modify tyrosine residues on AAV capsids using specific ligands, thereby circumventing the need to genetically engineer the capsid sequence. Aromatic electrophilic substitution of the phenol ring of tyrosine residues on AAV capsids improved the in vivo transduction efficiency of rAAV2 vectors in both liver and retinal targets. This tyrosine bioconjugation strategy represents an innovative technology for the engineering of rAAV vectors for human gene therapy.
Collapse
Affiliation(s)
- Aurélien Leray
- Nantes Université, CNRS, CEISAM, UMR 6230, F-44000 Nantes, France
| | | | - Juliette Varin
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | - Mohammed Bouzelha
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | - Audrey Bourdon
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | | | - Karine Pavageau
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | | | - Maia Marchand
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | - Anthony Mellet
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | - Joanna Demilly
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | - Jean-Baptiste Ducloyer
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | - Tiphaine Girard
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | - Bodvaël Fraysse
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | | | - Mickaël Guilbaud
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | | | - Eduard Ayuso
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | - Oumeya Adjali
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | | | - Thérèse Cronin
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | - Caroline Le Guiner
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France
| | - David Deniaud
- Nantes Université, CNRS, CEISAM, UMR 6230, F-44000 Nantes, France.
| | - Mathieu Mével
- Nantes Université, TaRGeT, Translational Research for Gene Therapies, CHU Nantes, INSERM, UMR 1089, F-44000 Nantes, France.
| |
Collapse
|
32
|
Huang X, Wang X, Sun Y, Li L, Li A, Xu W, Xie X, Diao Y. Bleomycin promotes rAAV2 transduction via DNA-PKcs/Artemis-mediated DNA break repair pathways. Virology 2024; 590:109959. [PMID: 38100984 DOI: 10.1016/j.virol.2023.109959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/17/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
Because it is safe and has a simple genome, recombinant adeno-associated virus (rAAV) is an extremely appealing vector for delivery in in vivo gene therapy. However, its low transduction efficiency for some cells, limits its further application in the field of gene therapy. Bleomycin is a chemotherapeutic agent approved by the FDA whose effect on rAAV transduction has not been studied. In this study, we systematically investigated the effect of Bleomycin on the second-strand synthesis and used CRISPR/CAS9 and RNAi methods to understand the effects of Bleomycin on rAAV vector transduction, particularly the effect of DNA repair enzymes. The results showed that Bleomycin could promote rAAV2 transduction both in vivo and in vitro. Increased transduction was discovered to be a direct result of decreased cytoplasmic rAAV particle degradation and increased second-strand synthesis. TDP1, PNKP, and SETMAR are required to repair the DNA damage gap caused by Bleomycin, TDP1, PNKP, and SETMAR promote rAAV second-strand synthesis. Bleomycin induced DNA-PKcs phosphorylation and phosphorylated DNA-PKcs and Artemis promoted second-strand synthesis. The current study identifies an effective method for increasing the capability and scope of in-vivo and in-vitro rAAV applications, which can amplify cell transduction at Bleomycin concentrations. It also supplies information on combining tumor gene therapy with chemotherapy.
Collapse
Affiliation(s)
- Xiaoping Huang
- College of Chemical Engineering and Materials Sciences, Quanzhou Normal University, Quanzhou, China; Institute of Molecular Medicine, Huaqiao University, Quanzhou, China
| | - Xiao Wang
- Institute of Molecular Medicine, Huaqiao University, Quanzhou, China
| | - Yaqi Sun
- College of Chemical Engineering and Materials Sciences, Quanzhou Normal University, Quanzhou, China
| | - Ling Li
- Institute of Molecular Medicine, Huaqiao University, Quanzhou, China
| | - Anna Li
- Institute of Molecular Medicine, Huaqiao University, Quanzhou, China
| | - Wentao Xu
- College of Chemical Engineering and Materials Sciences, Quanzhou Normal University, Quanzhou, China
| | - Xiaolan Xie
- College of Chemical Engineering and Materials Sciences, Quanzhou Normal University, Quanzhou, China.
| | - Yong Diao
- Institute of Molecular Medicine, Huaqiao University, Quanzhou, China.
| |
Collapse
|
33
|
Smith J, Carillo S, Kulkarni A, Redman E, Yu K, Bones J. Rapid characterization of adeno-associated virus (AAV) capsid proteins using microchip ZipChip CE-MS. Anal Bioanal Chem 2024; 416:1069-1084. [PMID: 38102410 PMCID: PMC10800304 DOI: 10.1007/s00216-023-05097-5] [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: 11/08/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023]
Abstract
Adeno-associated viruses (AAVs) are viral vectors used as delivery systems for gene therapies. Intact protein characterization of AAV viral capsid proteins (VPs) and their post-translational modifications is critical to ensuring product quality. In this study, microchip-based ZipChip capillary electrophoresis-mass spectrometry (CE-MS) was applied for the rapid characterization of AAV intact VPs, specifically full and empty viral capsids of serotypes AAV6, AAV8 and AAV9, which was accomplished using 5 min of analysis time. Low levels of dimethyl sulfoxide (4%) in the background electrolyte (BGE) improved MS signal quality and component detection. A sensitivity evaluation revealed consistent detection of VP proteoforms when as little as 2.64 × 106 viral particles (≈26.4 picograms) were injected. Besides the traditional VP proteoforms used for serotype identification, multiple VP3 variants were detected, including truncated VP3 variants most likely generated by leaky scanning as well as unacetylated and un-cleaved VP3 proteoforms. Phosphorylation, known to impact AAV transduction efficiency, was also seen in all serotypes analysed. Additionally, low abundant fragments originating from either N- or C-terminus truncation were detected. As the aforementioned VP components can impact product quality and efficacy, the ZipChip's ability to rapidly characterize them illustrates its strength in monitoring product quality during AAV production.
Collapse
Affiliation(s)
- Josh Smith
- Characterisation and Comparability Laboratory, The National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, A94 X099, Co. Dublin, Ireland
| | - Sara Carillo
- Characterisation and Comparability Laboratory, The National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, A94 X099, Co. Dublin, Ireland
| | - Aditya Kulkarni
- 908 Devices Inc., 645 Summer Street #201, Boston, MA, 02210, USA
| | - Erin Redman
- 908 Devices Inc., 511 Davis Dr Suite 450, Morrisville, NC, 27560, USA
| | - Kate Yu
- 908 Devices Inc., 645 Summer Street #201, Boston, MA, 02210, USA
| | - Jonathan Bones
- Characterisation and Comparability Laboratory, The National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, A94 X099, Co. Dublin, Ireland.
- School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland.
| |
Collapse
|
34
|
Zhu D, Brookes DH, Busia A, Carneiro A, Fannjiang C, Popova G, Shin D, Donohue KC, Lin LF, Miller ZM, Williams ER, Chang EF, Nowakowski TJ, Listgarten J, Schaffer DV. Optimal trade-off control in machine learning-based library design, with application to adeno-associated virus (AAV) for gene therapy. SCIENCE ADVANCES 2024; 10:eadj3786. [PMID: 38266077 PMCID: PMC10807795 DOI: 10.1126/sciadv.adj3786] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024]
Abstract
Adeno-associated viruses (AAVs) hold tremendous promise as delivery vectors for gene therapies. AAVs have been successfully engineered-for instance, for more efficient and/or cell-specific delivery to numerous tissues-by creating large, diverse starting libraries and selecting for desired properties. However, these starting libraries often contain a high proportion of variants unable to assemble or package their genomes, a prerequisite for any gene delivery goal. Here, we present and showcase a machine learning (ML) method for designing AAV peptide insertion libraries that achieve fivefold higher packaging fitness than the standard NNK library with negligible reduction in diversity. To demonstrate our ML-designed library's utility for downstream engineering goals, we show that it yields approximately 10-fold more successful variants than the NNK library after selection for infection of human brain tissue, leading to a promising glial-specific variant. Moreover, our design approach can be applied to other types of libraries for AAV and beyond.
Collapse
Affiliation(s)
- Danqing Zhu
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - David H. Brookes
- Biophysics Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Akosua Busia
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ana Carneiro
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Galina Popova
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Psychiatry and Behavioural Sciences, University of California San Francisco, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
| | - David Shin
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Psychiatry and Behavioural Sciences, University of California San Francisco, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kevin C. Donohue
- Department of Psychiatry and Behavioural Sciences, University of California San Francisco, San Francisco, CA 94143, USA
- School of Medicine, University of California San Francisco, San Francisco, CA, USA. 94143
- Kavli Institute of Fundamental Neuroscience, University of California San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Li F. Lin
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Zachary M. Miller
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Evan R. Williams
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Edward F. Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Tomasz J. Nowakowski
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Psychiatry and Behavioural Sciences, University of California San Francisco, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jennifer Listgarten
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94720, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - David V. Schaffer
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Institute (IGI), University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
35
|
Castro BFM, Steel JC, Layton CJ. AAV-Based Strategies for Treatment of Retinal and Choroidal Vascular Diseases: Advances in Age-Related Macular Degeneration and Diabetic Retinopathy Therapies. BioDrugs 2024; 38:73-93. [PMID: 37878215 PMCID: PMC10789843 DOI: 10.1007/s40259-023-00629-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/26/2023]
Abstract
Age-related macular degeneration (AMD) and diabetic retinopathy (DR) are vascular diseases with high prevalence, ranking among the leading causes of blindness and vision loss worldwide. Despite being effective, current treatments for AMD and DR are burdensome for patients and clinicians, resulting in suboptimal compliance and real risk of vision loss. Thus, there is an unmet need for long-lasting alternatives with improved safety and efficacy. Adeno-associated virus (AAV) is the leading vector for ocular gene delivery, given its ability to enable long-term expression while eliciting relatively mild immune responses. Progress has been made in AAV-based gene therapies for not only inherited retinal diseases but also acquired conditions with preclinical and clinical studies of AMD and DR showing promising results. These studies have explored several pathways involved in the disease pathogenesis, as well as different strategies to optimise gene delivery. These include engineered capsids with enhanced tropism to particular cell types, and expression cassettes incorporating elements for a targeted and controlled expression. Multiple-acting constructs have also been investigated, in addition to gene silencing and editing. Here, we provide an overview of strategies employing AAV-mediated gene delivery to treat AMD and DR. We discuss preclinical efficacy studies and present the latest data from clinical trials for both diseases.
Collapse
Affiliation(s)
- Brenda F M Castro
- LVF Ophthalmology Research Centre, Translational Research Institute, Brisbane, QLD, 4102, Australia.
- Greenslopes Clinical School, University of Queensland School of Medicine, Brisbane, QLD, Australia.
| | - Jason C Steel
- LVF Ophthalmology Research Centre, Translational Research Institute, Brisbane, QLD, 4102, Australia
- Greenslopes Clinical School, University of Queensland School of Medicine, Brisbane, QLD, Australia
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD, Australia
| | - Christopher J Layton
- LVF Ophthalmology Research Centre, Translational Research Institute, Brisbane, QLD, 4102, Australia.
- Greenslopes Clinical School, University of Queensland School of Medicine, Brisbane, QLD, Australia.
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD, Australia.
| |
Collapse
|
36
|
Luo LL, Xu J, Wang BQ, Chen C, Chen X, Hu QM, Wang YQ, Zhang WY, Jiang WX, Li XT, Zhou H, Xiao X, Zhao K, Lin S. A novel capsid-XL32-derived adeno-associated virus serotype prompts retinal tropism and ameliorates choroidal neovascularization. Biomaterials 2024; 304:122403. [PMID: 38016335 DOI: 10.1016/j.biomaterials.2023.122403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/24/2023] [Accepted: 11/14/2023] [Indexed: 11/30/2023]
Abstract
Gene therapy has been adapted, from the laboratory to the clinic, to treat retinopathies. In contrast to subretinal route, intravitreal delivery of AAV vectors displays the advantage of bypassing surgical injuries, but the viral particles are more prone to be nullified by the host neutralizing factors. To minimize such suppression of therapeutic effect, especially in terms of AAV2 and its derivatives, we introduced three serine-to-glycine mutations, based on the phosphorylation sites identified by mass spectrum analysis, to the XL32 capsid to generate a novel serotype named AAVYC5. Via intravitreal administration, AAVYC5 was transduced more effectively into multiple retinal layers compared with AAV2 and XL32. AAVYC5 also enabled successful delivery of anti-angiogenic molecules to rescue laser-induced choroidal neovascularization and astrogliosis in mice and non-human primates. Furthermore, we detected fewer neutralizing antibodies and binding IgG in human sera against AAVYC5 than those specific for AAV2 and XL32. Our results thus implicate this capsid-optimized AAVYC5 as a promising vector suitable for a wide population, particularly those with undesirable AAV2 seroreactivity.
Collapse
Affiliation(s)
- Lin-Lin Luo
- Department of Ophthalmology, Army Medical Center of PLA, Army Medical University, Chongqing, 400042, China
| | - Jie Xu
- Department of Ophthalmology, Army Medical Center of PLA, Army Medical University, Chongqing, 400042, China
| | - Bing-Qiao Wang
- Department of Neurology, The Second Affiliated Hospital, Army Medical University, Chongqing, 400042, China
| | - Chen Chen
- School of Bioengineering, East China University of Science and Technology, Shanghai, 200237, China; Belief BioMed Co., Ltd, Shanghai, China
| | - Xi Chen
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, 400064, China
| | - Qiu-Mei Hu
- Department of Ophthalmology, Army Medical Center of PLA, Army Medical University, Chongqing, 400042, China
| | - Yu-Qiu Wang
- School of Bioengineering, East China University of Science and Technology, Shanghai, 200237, China; Analytical Research Center for Organic and Biological Molecules, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wan-Yun Zhang
- Department of Neurology, The Second Affiliated Hospital, Army Medical University, Chongqing, 400042, China
| | - Wan-Xiang Jiang
- Sichuan Greentech Bioscience Co,. Ltd, Bencao Avenue, New Economic Development Zone, Meishan, Sichuan, 620010, China
| | - Xin-Ting Li
- School of Bioengineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Hu Zhou
- Analytical Research Center for Organic and Biological Molecules, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao Xiao
- School of Bioengineering, East China University of Science and Technology, Shanghai, 200237, China; Belief BioMed Co., Ltd, Shanghai, China.
| | - Kai Zhao
- School of Bioengineering, East China University of Science and Technology, Shanghai, 200237, China; Belief BioMed Co., Ltd, Shanghai, China.
| | - Sen Lin
- Department of Neurology, The Second Affiliated Hospital, Army Medical University, Chongqing, 400042, China; Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, 400064, China.
| |
Collapse
|
37
|
Barker RA, Buttery PC. Disease-specific interventions: The use of cell and gene therapies for Parkinson disease. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:171-191. [PMID: 39341654 DOI: 10.1016/b978-0-323-90120-8.00003-4] [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: 10/01/2024]
Abstract
Approaches to repair the brain around the loss of the nigrostriatal dopaminergic pathways in Parkinson disease (PD) are not new and have been attempted over many years. However, of late, the situation has moved forward in two main ways. In the case of cell therapies, the ability to make large numbers of authentic midbrain dopaminergic neuroblasts from human pluripotent stem cell sources has turned what was an interesting avenue of research into a major area of investment and trialing, by academics in conjunction with Pharma. In the case of gene therapies, their use around dopamine replacement has waned, as the interest in using them for disease modification targeting PD-specific pathways has grown. In this chapter, we discuss all these developments and the current status of cell and gene therapies for PD.
Collapse
Affiliation(s)
- Roger A Barker
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.
| | - Philip C Buttery
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
38
|
Smith J, Guapo F, Strasser L, Millán-Martín S, Milian SG, Snyder RO, Bones J. Development of a Rapid Adeno-Associated Virus (AAV) Identity Testing Platform through Comprehensive Intact Mass Analysis of Full-Length AAV Capsid Proteins. J Proteome Res 2023; 23:161-174. [PMID: 38123456 PMCID: PMC10775144 DOI: 10.1021/acs.jproteome.3c00513] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023]
Abstract
Adeno-associated viruses (AAVs) are commonly used as vectors for the delivery of gene therapy targets. Characterization of AAV capsid proteins (VPs) and their post-translational modifications (PTMs) have become a critical attribute monitored to evaluate product quality. Liquid chromatography-mass spectrometry (LC-MS) analysis of intact AAV VPs provides both quick and reliable serotype identification as well as proteoform information on each VP. Incorporating these analytical strategies into rapid good manufacturing practice (GMP)-compliant workflows containing robust, but simplified, data processing methods is necessary to ensure effective product quality control (QC) during production. Here, we present a GMP-compliant LC-MS workflow for the rapid identification and in-depth characterization of AAVs. Hydrophilic interaction liquid chromatography (HILIC) MS with difluoroacetic acid as a mobile phase modifier is utilized to achieve the intact separation and identification of AAV VPs and their potential proteoforms. Peptide mapping is performed to confirm PTMs identified during intact VP analysis and for in-depth PTM characterization. The intact separations platform is then incorporated into a data processing workflow developed using GMP-compliant software capable of rapid AAV serotype identification and, if desired, specific serotype PTM monitoring and characterization. Such a platform provides product QC capabilities that are easily accessible in a regulatory setting.
Collapse
Affiliation(s)
- Josh Smith
- Characterisation
and Comparability Laboratory, The National
Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Dublin A94 X099, Ireland
| | - Felipe Guapo
- Characterisation
and Comparability Laboratory, The National
Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Dublin A94 X099, Ireland
| | - Lisa Strasser
- Characterisation
and Comparability Laboratory, The National
Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Dublin A94 X099, Ireland
| | - Silvia Millán-Martín
- Characterisation
and Comparability Laboratory, The National
Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Dublin A94 X099, Ireland
| | - Steven G. Milian
- Patheon
Viral Vector Services, 13859 Progress Blvd, Alachua, Florida 32615, United States
| | - Richard O. Snyder
- Patheon
Viral Vector Services, 13859 Progress Blvd, Alachua, Florida 32615, United States
| | - Jonathan Bones
- Characterisation
and Comparability Laboratory, The National
Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Dublin A94 X099, Ireland
- School
of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin D04 V1W8.F, Ireland
| |
Collapse
|
39
|
Jacobs R, Dogbey MD, Mnyandu N, Neves K, Barth S, Arbuthnot P, Maepa MB. AAV Immunotoxicity: Implications in Anti-HBV Gene Therapy. Microorganisms 2023; 11:2985. [PMID: 38138129 PMCID: PMC10745739 DOI: 10.3390/microorganisms11122985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/30/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Hepatitis B virus (HBV) has afflicted humankind for decades and there is still no treatment that can clear the infection. The development of recombinant adeno-associated virus (rAAV)-based gene therapy for HBV infection has become important in recent years and research has made exciting leaps. Initial studies, mainly using mouse models, showed that rAAVs are non-toxic and induce minimal immune responses. However, several later studies demonstrated rAAV toxicity, which is inextricably associated with immunogenicity. This is a major setback for the progression of rAAV-based therapies toward clinical application. Research aimed at understanding the mechanisms behind rAAV immunity and toxicity has contributed significantly to the inception of approaches to overcoming these challenges. The target tissue, the features of the vector, and the vector dose are some of the determinants of AAV toxicity, with the latter being associated with the most severe adverse events. This review discusses our current understanding of rAAV immunogenicity, toxicity, and approaches to overcoming these hurdles. How this information and current knowledge about HBV biology and immunity can be harnessed in the efforts to design safe and effective anti-HBV rAAVs is discussed.
Collapse
Affiliation(s)
- Ridhwaanah Jacobs
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Infectious Diseases and Oncology Research Institute (IDORI), Faculty of Health Sciences, University of the Witwatersrand, Parktown 2193, South Africa
| | - Makafui Dennis Dogbey
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa; (M.D.D.)
| | - Njabulo Mnyandu
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Infectious Diseases and Oncology Research Institute (IDORI), Faculty of Health Sciences, University of the Witwatersrand, Parktown 2193, South Africa
| | - Keila Neves
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Infectious Diseases and Oncology Research Institute (IDORI), Faculty of Health Sciences, University of the Witwatersrand, Parktown 2193, South Africa
| | - Stefan Barth
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa; (M.D.D.)
- South African Research Chair in Cancer Biotechnology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Patrick Arbuthnot
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Infectious Diseases and Oncology Research Institute (IDORI), Faculty of Health Sciences, University of the Witwatersrand, Parktown 2193, South Africa
| | - Mohube Betty Maepa
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Infectious Diseases and Oncology Research Institute (IDORI), Faculty of Health Sciences, University of the Witwatersrand, Parktown 2193, South Africa
| |
Collapse
|
40
|
Shoti J, Qing K, Keeler GD, Duan D, Byrne BJ, Srivastava A. Development of capsid- and genome-modified optimized AAVrh74 vectors for muscle gene therapy. Mol Ther Methods Clin Dev 2023; 31:101147. [PMID: 38046199 PMCID: PMC10690633 DOI: 10.1016/j.omtm.2023.101147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 10/31/2023] [Indexed: 12/05/2023]
Abstract
The first generation of adeno-associated virus (AAV) vectors composed of the naturally occurring capsids and genomes, although effective in some instances, are unlikely to be optimal for gene therapy in humans. The use of the first generation of two different AAV serotype vectors (AAV9 and AAVrh74) in four separate clinical trials failed to be effective in patients with Duchenne muscular dystrophy, although some efficacy was observed in a subset of patients with AAVrh74 vectors leading to US Food and Drug Administration approval (Elevidys). In two trials with the first generation of AAV9 vectors, several serious adverse events were observed, including the death of a patient in one trial, and more recently, in the death of a second patient in an N-of-1 clinical trial. In a fourth trial with the first generation of AAVrh74 vectors, myositis and myocarditis were also observed. Here, we report that capsid- and genome-modified optimized AAVrh74 vectors are significantly more efficient in transducing primary human skeletal muscle cells in vitro and in all major muscle tissues in vivo following systemic administration in a murine model. The availability of optimized AAVrh74 vectors promises to be safe and effective in the potential gene therapy of muscle diseases in humans.
Collapse
Affiliation(s)
- Jakob Shoti
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Keyun Qing
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Geoffrey D. Keeler
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Dongsheng Duan
- Departments of Microbiology and Immunology, Neurology, Biomedical Sciences, and Chemical and Biomedical Engineering, University of Missouri, Columbia, MO, USA
| | - Barry J. Byrne
- Child Health Research Institute, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| | - Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
41
|
Kellish PC, Marsic D, Crosson SM, Choudhury S, Scalabrino ML, Strang CE, Hill J, McCullough KT, Peterson JJ, Fajardo D, Gupte S, Makal V, Kondratov O, Kondratova L, Iyer S, Witherspoon CD, Gamlin PD, Zolotukhin S, Boye SL, Boye SE. Intravitreal injection of a rationally designed AAV capsid library in non-human primate identifies variants with enhanced retinal transduction and neutralizing antibody evasion. Mol Ther 2023; 31:3441-3456. [PMID: 37814449 PMCID: PMC10727955 DOI: 10.1016/j.ymthe.2023.10.001] [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: 08/23/2023] [Revised: 09/25/2023] [Accepted: 10/04/2023] [Indexed: 10/11/2023] Open
Abstract
Adeno-associated virus (AAV) continues to be the gold standard vector for therapeutic gene delivery and has proven especially useful for treating ocular disease. Intravitreal injection (IVtI) is a promising delivery route because it increases accessibility of gene therapies to larger patient populations. However, data from clinical and non-human primate (NHP) studies utilizing currently available capsids indicate that anatomical barriers to AAV and pre-existing neutralizing antibodies can restrict gene expression to levels that are "sub-therapeutic" in a substantial proportion of patients. Here, we performed a combination of directed evolution in NHPs of an AAV2-based capsid library with simultaneous mutations across six surface-exposed variable regions and rational design to identify novel capsid variants with improved retinal transduction following IVtI. Following two rounds of screening in NHP, enriched variants were characterized in intravitreally injected mice and NHPs and shown to have increased transduction relative to AAV2. Lead capsid variant, P2-V1, demonstrated an increased ability to evade neutralizing antibodies in human vitreous samples relative to AAV2 and AAV2.7m8. Taken together, this study further contributed to our understanding of the selective pressures associated with retinal transduction via the vitreous and identified promising novel AAV capsid variants for clinical consideration.
Collapse
Affiliation(s)
- Patrick C Kellish
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
| | - Damien Marsic
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
| | - Sean M Crosson
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
| | - Shreyasi Choudhury
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
| | - Miranda L Scalabrino
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
| | - Christianne E Strang
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham AL 35294, USA
| | - Julie Hill
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham AL 35294, USA
| | - K Tyler McCullough
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
| | - James J Peterson
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
| | - Diego Fajardo
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
| | - Siddhant Gupte
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
| | - Victoria Makal
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
| | - Oleksandr Kondratov
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
| | - Liudmyla Kondratova
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
| | - Siva Iyer
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - C Douglas Witherspoon
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham AL 35294, USA
| | - Paul D Gamlin
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham AL 35294, USA
| | - Sergei Zolotukhin
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
| | - Sanford L Boye
- Powell Gene Therapy Center, Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | - Shannon E Boye
- Division of Cellular and Molecular Therapy, Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
42
|
Cooney AL, Brommel CM, Traore S, Newby GA, Liu DR, McCray PB, Sinn PL. Reciprocal mutations of lung-tropic AAV capsids lead to improved transduction properties. Front Genome Ed 2023; 5:1271813. [PMID: 38077224 PMCID: PMC10702583 DOI: 10.3389/fgeed.2023.1271813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/17/2023] [Indexed: 12/21/2023] Open
Abstract
Considerable effort has been devoted to developing adeno-associated virus (AAV)-based vectors for gene therapy in cystic fibrosis (CF). As a result of directed evolution and capsid shuffling technology, AAV capsids are available with widespread tropism for airway epithelial cells. For example, AAV2.5T and AAV6.2 are two evolved capsids with improved airway epithelial cell transduction properties over their parental serotypes. However, limited research has been focused on identifying their specific cellular tropism. Restoring cystic fibrosis transmembrane conductance regulator (CFTR) expression in surface columnar epithelial cells is necessary for the correction of the CF airway phenotype. Basal cells are a progenitor population of the conducting airways responsible for replenishing surface epithelial cells (including secretory cells and ionocytes), making correction of this cell population vital for a long-lived gene therapy strategy. In this study, we investigate the tropism of AAV capsids for three cell types in primary cultures of well-differentiated human airway epithelial (HAE) cells and primary human airway basal cells. We observed that AAV2.5T transduced surface epithelial cells better than AAV6.2, while AAV6.2 transduced airway basal cells better than AAV2.5T. We also investigated a recently developed capsid, AAV6.2FF, which has two surface tyrosines converted to phenylalanines. Next, we incorporated reciprocal mutations to create AAV capsids with further improved surface and basal cell transduction characteristics. Lastly, we successfully employed a split-intein approach using AAV to deliver an adenine base editor (ABE) to repair the CFTR R553X mutation. Our results suggest that rational incorporation of AAV capsid mutations improves AAV transduction of the airway surface and progenitor cells and may ultimately lead to improved pulmonary function in people with CF.
Collapse
Affiliation(s)
- Ashley L. Cooney
- University of Iowa, Stead Family Department of Pediatrics, Iowa City, IA, United States
- Pappajohn Biomedical Institute, Iowa City, IA, United States
- Center for Cystic Fibrosis Gene Therapy, University of Iowa, Iowa City, IA, United States
| | - Christian M. Brommel
- University of Iowa, Stead Family Department of Pediatrics, Iowa City, IA, United States
- Pappajohn Biomedical Institute, Iowa City, IA, United States
- Center for Cystic Fibrosis Gene Therapy, University of Iowa, Iowa City, IA, United States
| | - Soumba Traore
- University of Iowa, Stead Family Department of Pediatrics, Iowa City, IA, United States
- Pappajohn Biomedical Institute, Iowa City, IA, United States
- Center for Cystic Fibrosis Gene Therapy, University of Iowa, Iowa City, IA, United States
| | - Gregory A. Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, United States
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, United States
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, United States
| | - David R. Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, United States
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, United States
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, United States
| | - Paul B. McCray
- University of Iowa, Stead Family Department of Pediatrics, Iowa City, IA, United States
- Pappajohn Biomedical Institute, Iowa City, IA, United States
- Center for Cystic Fibrosis Gene Therapy, University of Iowa, Iowa City, IA, United States
| | - Patrick L. Sinn
- University of Iowa, Stead Family Department of Pediatrics, Iowa City, IA, United States
- Pappajohn Biomedical Institute, Iowa City, IA, United States
- Center for Cystic Fibrosis Gene Therapy, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
43
|
Pacak CA, Suzuki-Hatano S, Khadir F, Daugherty AL, Sriramvenugopal M, Gosiker BJ, Kang PB, Cade WT. One episode of low intensity aerobic exercise prior to systemic AAV9 administration augments transgene delivery to the heart and skeletal muscle. J Transl Med 2023; 21:748. [PMID: 37875924 PMCID: PMC10598899 DOI: 10.1186/s12967-023-04626-1] [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: 08/11/2023] [Accepted: 10/13/2023] [Indexed: 10/26/2023] Open
Abstract
INTRODUCTION The promising potential of adeno-associated virus (AAV) gene delivery strategies to treat genetic disorders continues to grow with an additional three AAV-based therapies recently approved by the Food and Drug Administration and dozens of others currently under evaluation in clinical trials. With these developments, it has become increasingly apparent that the high doses currently needed for efficacy carry risks of toxicity and entail enormous manufacturing costs, especially for clinical grade products. Strategies to increase the therapeutic efficacy of AAV-mediated gene delivery and reduce the minimal effective dose would have a substantial impact on this field. We hypothesized that an exercise-induced redistribution of tissue perfusion in the body to favor specific target organs via acute aerobic exercise prior to systemic intravenous (IV) AAV administration could increase efficacy. BACKGROUND Aerobic exercise triggers an array of downstream physiological effects including increased perfusion of heart and skeletal muscle, which we expected could enhance AAV transduction. Prior preclinical studies have shown promising results for a gene therapy approach to treat Barth syndrome (BTHS), a rare monogenic cardioskeletal myopathy, and clinical studies have shown the benefit of low intensity exercise in these patients, making this a suitable disease in which to test the ability of aerobic exercise to enhance AAV transduction. METHODS Wild-type (WT) and BTHS mice were either systemically administered AAV9 or completed one episode of low intensity treadmill exercise immediately prior to systemic administration of AAV9. RESULTS We demonstrate that a single episode of acute low intensity aerobic exercise immediately prior to IV AAV9 administration improves marker transgene delivery in WT mice as compared to mice injected without the exercise pre-treatment. In BTHS mice, prior exercise improved transgene delivery and additionally increased improvement in mitochondrial gene transcription levels and mitochondrial function in the heart and gastrocnemius muscles as compared to mice treated without exercise. CONCLUSIONS Our findings suggest that one episode of acute low intensity aerobic exercise improves AAV9 transduction of heart and skeletal muscle. This low-risk, cost effective intervention could be implemented in clinical trials of individuals with inherited cardioskeletal disease as a potential means of improving patient safety for human gene therapy.
Collapse
Affiliation(s)
- Christina A Pacak
- Paul and Sheila Wellstone Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, 420 Delaware St SE, Minneapolis, MN, 55455, USA.
| | - Silveli Suzuki-Hatano
- College of Medicine, Department of Pediatrics, University of Florida, Gainesville, USA
| | - Fatemeh Khadir
- Paul and Sheila Wellstone Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, 420 Delaware St SE, Minneapolis, MN, 55455, USA
| | - Audrey L Daugherty
- Paul and Sheila Wellstone Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, 420 Delaware St SE, Minneapolis, MN, 55455, USA
| | | | - Bennett J Gosiker
- College of Medicine, Department of Pediatrics, University of Florida, Gainesville, USA
| | - Peter B Kang
- Paul and Sheila Wellstone Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, 420 Delaware St SE, Minneapolis, MN, 55455, USA
| | - William Todd Cade
- Physical Therapy Division, Department of Orthopaedic Surgery, Duke University School of Medicine, 311 Trent Drive, Durham, NC, 27710, USA.
| |
Collapse
|
44
|
Noda M, Koshu R, Shimada Dias M, Saito C, Takino N, Ito M, Yoshimura H, Ito M, Muramatsu SI. Enhanced Cochlear Transduction by AAV9 with High-Concentration Sucrose. Hum Gene Ther 2023; 34:1064-1071. [PMID: 37642269 DOI: 10.1089/hum.2023.111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023] Open
Abstract
The inner ear is a primary lesion in sensorineural hearing loss and has been a target in gene therapy. The efficacy of gene therapy depends on achieving sufficient levels of transduction at a safe vector dose. Vectors derived from various adeno-associated viruses (AAVs) are predominantly used to deliver therapeutic genes to inner ear cells. AAV9 and its variants vector are attractive candidates for clinical applications since they can cross the mesothelial cell layer and transduce inner hair cells (IHCs), although this requires relatively high doses. In this study, we investigated the effects of sucrose on the transduction of a variant of the AAV9 vector for gene transfer in the inner ear. We found that high concentrations of sucrose increased gene transduction in House Ear Institute-Organ of Corti 1 (HEI-OC1) cells in vitro. In addition, we demonstrated that simultaneous administration of sucrose enhanced the transduction of mouse IHCs and spiral ligament cells using an AAV9 variant vector. The procedure did not increase the thresholds in the auditory brainstem response, suggesting that sucrose had no adverse effect on auditory function. This versatile method may be valuable in the development of novel gene therapies for adult-onset sensorineural hearing loss.
Collapse
Affiliation(s)
- Masao Noda
- Department of Otolaryngology-Head and Neck Surgery, Jichi Medical University, Tochigi, Japan
- Division of Neurological Gene Therapy, Jichi Medical University, Tochigi, Japan
| | - Ryota Koshu
- Department of Otolaryngology-Head and Neck Surgery, Jichi Medical University, Tochigi, Japan
| | - Mari Shimada Dias
- Department of Otolaryngology-Head and Neck Surgery, Jichi Medical University, Tochigi, Japan
| | - Chizu Saito
- Department of Otolaryngology-Head and Neck Surgery, Jichi Medical University, Tochigi, Japan
| | - Naomi Takino
- Division of Neurological Gene Therapy, Jichi Medical University, Tochigi, Japan
| | - Mika Ito
- Division of Neurological Gene Therapy, Jichi Medical University, Tochigi, Japan
| | - Hidekane Yoshimura
- Department of Otolaryngology-Head and Neck Surgery, Shinshu University, Japan
| | - Makoto Ito
- Department of Otolaryngology-Head and Neck Surgery, Jichi Medical University, Tochigi, Japan
| | - Shin-Ichi Muramatsu
- Division of Neurological Gene Therapy, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
45
|
Jo A, Deniz S, Xu J, Duvoisin RM, DeVries SH, Zhu Y. A sign-inverted receptive field of inhibitory interneurons provides a pathway for ON-OFF interactions in the retina. Nat Commun 2023; 14:5937. [PMID: 37741839 PMCID: PMC10517963 DOI: 10.1038/s41467-023-41638-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 09/11/2023] [Indexed: 09/25/2023] Open
Abstract
A fundamental organizing plan of the retina is that visual information is divided into ON and OFF streams that are processed in separate layers. This functional dichotomy originates in the ON and OFF bipolar cells, which then make excitatory glutamatergic synapses onto amacrine and ganglion cells in the inner plexiform layer. We have identified an amacrine cell (AC), the sign-inverting (SI) AC, that challenges this fundamental plan. The glycinergic, ON-stratifying SI-AC has OFF light responses. In opposition to the classical wiring diagrams, it receives inhibitory inputs from glutamatergic ON bipolar cells at mGluR8 synapses, and excitatory inputs from an OFF wide-field AC at electrical synapses. This "inhibitory ON center - excitatory OFF surround" receptive-field of the SI-AC allows it to use monostratified dendrites to conduct crossover inhibition and push-pull activation to enhance light detection by ACs and RGCs in the dark and feature discrimination in the light.
Collapse
Affiliation(s)
- Andrew Jo
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Sercan Deniz
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jian Xu
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Robert M Duvoisin
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Steven H DeVries
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| | - Yongling Zhu
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
46
|
Huang X, Wang X, Li L, Wang Q, Xu W, Wu W, Xie X, Diao Y. MiR133b-mediated inhibition of EGFR-PTK pathway promotes rAAV2 transduction by facilitating intracellular trafficking and augmenting second-strand synthesis. J Cell Mol Med 2023; 27:2714-2729. [PMID: 37469226 PMCID: PMC10494303 DOI: 10.1111/jcmm.17858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) is an extremely attractive vector in the in vivo delivery of gene therapy as it is safe and its genome is simple. However, challenges including low permissiveness to specific cells and restricted tissue specificity have hindered its clinical application. Based on the previous studies, epidermal growth factor receptor-protein tyrosine kinase (EGFR-PTK) negatively regulated rAAV transduction, and EGFR-positive cells were hardly permissive to rAAV transduction. We constructed a novel rAAV-miRNA133b vector, which co-expressed miRNA133b and transgene, and investigated its in vivo and in vitro transduction efficiency. Confocal microscopy, live-cell imaging, pharmacological reagents and labelled virion tracking were used to analyse the effect of miRNA133b on rAAV2 transduction and the underlying mechanisms. The results demonstrated that miRNA133b could promote rAAV2 transduction and the effects were limited to EGFR-positive cells. The increased transduction was found to be a direct result of decreased rAAV particles degradation in the cytoplasm and enhanced second-strand synthesis. ss-rAAV2-miRNA133b vector specifically increased rAAV2 transduction in EGFR-positive cells or tissues, while ss-rAAV2-Fluc-miRNA133b exerted an antitumor effect. rAAV-miRNA133b vector might emerge as a promising platform for delivering various transgene to treat EGFR-positive cell-related diseases, such as non-small-cell lung cancer.
Collapse
Affiliation(s)
- Xiaoping Huang
- College of Chemical Engineering and Materials SciencesQuanzhou Normal UniversityQuanzhouChina
- School of MedicineHuaqiao UniversityQuanzhouChina
| | - Xiao Wang
- School of MedicineHuaqiao UniversityQuanzhouChina
| | - Ling Li
- School of MedicineHuaqiao UniversityQuanzhouChina
| | - Qizhao Wang
- School of MedicineHuaqiao UniversityQuanzhouChina
| | - Wentao Xu
- College of Chemical Engineering and Materials SciencesQuanzhou Normal UniversityQuanzhouChina
| | - Wenlin Wu
- College of Chemical Engineering and Materials SciencesQuanzhou Normal UniversityQuanzhouChina
| | - Xiaolan Xie
- College of Chemical Engineering and Materials SciencesQuanzhou Normal UniversityQuanzhouChina
| | - Yong Diao
- School of MedicineHuaqiao UniversityQuanzhouChina
| |
Collapse
|
47
|
Wang H, Zheng A, Arias EB, Kwak SE, Pan X, Duan D, Cartee GD. AS160 expression, but not AS160 Serine-588, Threonine-642, and Serine-704 phosphorylation, is essential for elevated insulin-stimulated glucose uptake by skeletal muscle from female rats after acute exercise. FASEB J 2023; 37:e23021. [PMID: 37289137 DOI: 10.1096/fj.202300282rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/09/2023]
Abstract
One exercise session can increase subsequent insulin-stimulated glucose uptake (ISGU) by skeletal muscle in both sexes. We recently found that muscle expression and phosphorylation of key sites of Akt substrate of 160 kDa (AS160; also called TBC1D4) are essential for the full-exercise effect on postexercise-ISGU (PEX-ISGU) in male rats. In striking contrast, AS160's role in increased PEX-ISGU has not been rigorously tested in females. Our rationale was to address this major knowledge gap. Wild-type (WT) and AS160-knockout (KO) rats were either sedentary or acutely exercised. Adeno-associated virus (AAV) vectors were engineered to express either WT-AS160 or AS160 mutated on key serine and threonine residues (Ser588, Thr642, and Ser704) to alanine to prevent their phosphorylation. AAV vectors were delivered to the muscle of AS160-KO rats to determine if WT-AS160 or phosphorylation-inactivated AS160 would influence PEX-ISGU. AS160-KO rats have lower skeletal muscle abundance of the GLUT4 glucose transporter protein. This GLUT4 deficit was rescued using AAV delivery of GLUT4 to determine if eliminating muscle GLUT4 deficiency would normalize PEX-ISGU. The novel results were as follows: (1) AS160 expression was required for greater PEX-ISGU; (2) rescuing muscle AS160 expression in AS160-KO rats restored elevated PEX-ISGU; (3) AS160's essential role for the postexercise increase in ISGU was not attributable to reduced muscle GLUT4 content; and (4) AS160 phosphorylation on Ser588, Thr642, and Ser704 was not essential for greater PEX-ISGU. In conclusion, these novel findings revealed that three phosphosites widely proposed to influence PEX-ISGU are not required for this important outcome in female rats.
Collapse
Affiliation(s)
- Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Amy Zheng
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Edward B Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Seong Eun Kwak
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, Missouri, USA
| | - Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
48
|
Graves LE, Horton A, Alexander IE, Srinivasan S. Gene Therapy for Paediatric Homozygous Familial Hypercholesterolaemia. Heart Lung Circ 2023; 32:769-779. [PMID: 37012174 DOI: 10.1016/j.hlc.2023.01.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/26/2022] [Accepted: 01/04/2023] [Indexed: 04/03/2023]
Abstract
The clinical outcome for children and adolescents with homozygous familial hypercholesterolaemia (HoFH) can be devastating, and treatment options are limited in the presence of a null variant. In HoFH, atherosclerotic risk accumulates from birth. Gene therapy is an appealing treatment option as restoration of low-density lipoprotein receptor (LDLR) gene function could provide a cure for HoFH. A clinical trial using a recombinant adeno-associated vector (rAAV) to deliver LDLR DNA to adult patients with HoFH was recently completed; results have not yet been reported. However, this treatment strategy may face challenges when translating to the paediatric population. The paediatric liver undergoes substantial growth which is significant as rAAV vector DNA persists primarily as episomes (extra-chromosomal DNA) and are not replicated during cell division. Therefore, rAAV-based gene addition treatment administered in childhood would likely only have a transient effect. With over 2,000 unique variants in LDLR, a goal of genomic editing-based therapy development would be to treat most (if not all) mutations with a single set of reagents. For a robust, durable effect, LDLR must be repaired in the genome of hepatocytes, which could be achieved using genomic editing technology such as clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 and a DNA repair strategy such as homology-independent targeted integration. This review discusses this issue in the context of the paediatric patient group with severe compound heterozygous or homozygous null variants which are associated with aggressive early-onset atherosclerosis and myocardial infarction, together with the important pre-clinical studies that use genomic editing strategies to treat HoFH in place of apheresis and liver transplantation.
Collapse
Affiliation(s)
- Lara E Graves
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, NSW, Australia; Children's Hospital at Westmead Clinical School, University of Sydney, Sydney, NSW, Australia; Gene Therapy Research Unit, Children's Medical Research Institute, Sydney, NSW, Australia.
| | - Ari Horton
- Monash Heart and Monash Children's Hospital, Monash Health, Melbourne, Vic, Australia; Monash Cardiovascular Research Centre, Victorian Heart Institute, Melbourne, Vic, Australia; Monash Genetics, Monash Health, Melbourne, Vic, Australia; Department of Genomic Medicine, The Royal Melbourne Hospital, Parkville, Vic, Australia; Department of Paediatrics, Monash University Clayton, Vic, Australia
| | - Ian E Alexander
- Children's Hospital at Westmead Clinical School, University of Sydney, Sydney, NSW, Australia; Gene Therapy Research Unit, Children's Medical Research Institute, Sydney, NSW, Australia
| | - Shubha Srinivasan
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, NSW, Australia; Children's Hospital at Westmead Clinical School, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
49
|
Srivastava A. Rationale and strategies for the development of safe and effective optimized AAV vectors for human gene therapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:949-959. [PMID: 37293185 PMCID: PMC10244667 DOI: 10.1016/j.omtn.2023.05.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Recombinant adeno-associated virus (AAV) vectors have been, or are currently in use, in 332 phase I/II/III clinical trials in a number of human diseases, and in some cases, remarkable clinical efficacy has also been achieved. There are now three US Food and Drug Administration (FDA)-approved AAV "drugs," but it has become increasingly clear that the first generation of AAV vectors are not optimal. In addition, relatively large vector doses are needed to achieve clinical efficacy, which has been shown to provoke host immune responses culminating in serious adverse events and, more recently, in the deaths of 10 patients to date. Thus, there is an urgent need for the development of the next generation of AAV vectors that are (1) safe, (2) effective, and (3) human tropic. This review describes the strategies to potentially overcome each of the limitations of the first generation of AAV vectors and the rationale and approaches for the development of the next generation of AAV serotype vectors. These vectors promise to be efficacious at significant reduced doses, likely to achieve clinical efficacy, thereby increasing the safety as well as reducing vector production costs, ensuring translation to the clinic with higher probability of success, without the need for the use of immune suppression, for gene therapy of a wide variety of diseases in humans.
Collapse
Affiliation(s)
- Arun Srivastava
- Division of Cellular and Molecular Therapy, Departments of Pediatrics, Molecular Genetics and Microbiology, Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
50
|
Periasamy R, Patel DD, Boye SL, Boye SE, Lipinski DM. Improving retinal vascular endothelial cell tropism through rational rAAV capsid design. PLoS One 2023; 18:e0285370. [PMID: 37167304 PMCID: PMC10174500 DOI: 10.1371/journal.pone.0285370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/21/2023] [Indexed: 05/13/2023] Open
Abstract
Vascular endothelial cells (VEC) are essential for retinal homeostasis and their dysfunction underlies pathogenesis in diabetic retinopathy (DR) and exudative age-related macular degeneration (AMD). Studies have shown that recombinant adeno-associated virus (rAAV) vectors are effective at delivering new genetic material to neural and glial cells within the retina, but targeting VECs remains challenging. To overcome this limitation, herein we developed rAAV capsid mutant vectors with improved tropism towards retinal VEC. rAAV2/2, 2/2[QuadYF-TV], and rAAV2/9 serotype vectors (n = 9, capsid mutants per serotype) expressing GFP were generated by inserting heptameric peptides (7AA) designed to increase endothelial targeting at positions 588 (2/2 and 2/2[QuadYF-TV] or 589 (2/9) of the virus protein (VP 1-3). The packaging and transduction efficiency of the vectors were assessed in HEK293T and bovine VECs using Fluorescence microscopy and flow cytometry, leading to the identification of one mutant, termed EC5, that showed improved endothelial tropism when inserted into all three capsid serotypes. Intra-ocular and intravenous administration of EC5 mutants in C57Bl/6j mice demonstrated moderately improved transduction of the retinal vasculature, particularly surrounding the optic nerve head, and evidence of sinusoidal endothelial cell transduction in the liver. Most notably, intravenous administration of the rAAV2/2[QuadYF-TV] EC5 mutant led to a dramatic and unexpected increase in cardiac muscle transduction.
Collapse
Affiliation(s)
- Ramesh Periasamy
- Department of Ophthalmology and Visual Science, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Dwani D. Patel
- Department of Ophthalmology and Visual Science, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Cell Biology, Neurobiology, Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Sanford L. Boye
- Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL, United States of America
| | - Shannon E. Boye
- Department of Pediatrics, Division of Cellular and Molecular Biology, University of Florida, Gainesville, FL, United States of America
| | - Daniel M. Lipinski
- Department of Ophthalmology and Visual Science, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Cell Biology, Neurobiology, Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| |
Collapse
|