1
|
Kuo YS, Chiang PC, Kuo CY, Huang CG, Kuo ML, Chiu YF. Inhibition of influenza A virus proliferation in mice via universal RNA interference. Antiviral Res 2025; 238:106149. [PMID: 40147537 DOI: 10.1016/j.antiviral.2025.106149] [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: 10/24/2024] [Revised: 03/01/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Influenza A virus (IAV) is a respiratory pathogen that causes seasonal outbreaks and periodic pandemics. As frequent mutations in the IAV viral genome often render vaccines ineffective or inefficient in preventing the latest outbreak, there is a need to explore other preventive strategies to control the disease. This study sought to investigate the use of antiviral short hairpin RNA (shRNA), delivered by a recombinant adeno-associated virus (AAV), for the prevention of IAV infections. Conserved regions with less than 10 % of variation were identified from IAV genome sequences deposited in the National Center for Biotechnology Information (NCBI) database between 2000 and 2023. The shRNA targeting these conserved sequences was transcribed from the human RNA polymerase III U6 promoter in an AAV system. This study demonstrates that AAV delivery of shRNA against IAV genes encoding two of the viral RNA-dependent RNA polymerase subunits, PB1 and PB2, inhibits the replication of IAV H1N1 and H3N2 viruses in Madin-Darby canine kidney (MDCK) cells. Delivery of shPB1 to lung tissue in mice through AAV also provided effective protection against IAV infection. These results offer support for a shRNA-based strategy of influenza prevention.
Collapse
Affiliation(s)
- Yu-Shen Kuo
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Pei-Chuan Chiang
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Chieh-Ying Kuo
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Chung-Guei Huang
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ming-Ling Kuo
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Ya-Fang Chiu
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan; Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan; Division of Infectious Diseases, Department of Medicine, New Taipei Municipal Tucheng Hospital (Built and Operated By Chang Gung Medical Foundation), New Taipei City, Taiwan; Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
2
|
Powers TW, Sloan CD, Stano D, Evans B, Liu K, Mariani S, Campbell JA, Lerch TF, Mo JJ. Implementing a robust platform analytical procedure for measuring adeno-associated virus vector genome titer. Mol Ther Methods Clin Dev 2024; 32:101381. [PMID: 39670179 PMCID: PMC11634990 DOI: 10.1016/j.omtm.2024.101381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/12/2024] [Indexed: 12/14/2024]
Abstract
The vector genome (vg) titer measurement, which is used to control patient dosing and ensure control over drug product manufacturing, is essential for the development of recombinant adeno-associated virus (AAV) gene therapy products. While qPCR and droplet digital PCR technologies are commonly implemented for measuring vg titer, chromatographic techniques with UV detectors represent promising future approaches, in line with traditional biotherapeutics. Here, we introduce a novel vg titer measurement approach using size-exclusion high-performance liquid chromatography with UV detection, which achieves excellent method precision (<2% relative SD), demonstrates linearity across a range of concentrations and varied particle content, is stability indicating, and can be bridged with existing vg titer methods. As there is no bias between this procedure and existing vg titer procedures, such as qPCR, this method can be implemented even at late stages during pharmaceutical development. The procedure was demonstrated to be applicable across serotypes and transgenes, enabling the approach to be used as a platform method for AAV. Given the method performance and criticality of vg titer measurements for AAV, this approach represents a beneficial technology for AAV therapeutics.
Collapse
Affiliation(s)
- Thomas W. Powers
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Courtney D.K. Sloan
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Don Stano
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Brad Evans
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Kang Liu
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Shawn Mariani
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Jessica A. Campbell
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Thomas F. Lerch
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| | - Jim J. Mo
- Pfizer, Analytical Research and Development, 875 Chesterfield Pkwy. West, Chesterfield, MO 63017, USA
| |
Collapse
|
3
|
Ertl HCJ. Circumventing B Cell Responses to Allow for Redosing of Adeno-Associated Virus Vectors. Hum Gene Ther 2024; 35:416-424. [PMID: 37861281 DOI: 10.1089/hum.2023.162] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
Adeno-associated virus (AAV)-mediated gene therapy has made significant progress in the last few decades. Nevertheless, challenges imposed by the immune system remain. The very high doses of AAV vectors used for some disorders have resulted in serious adverse events (SAEs) or even deaths, demonstrating that AAV vector doses that can safely be injected into patients are limited and for some indications below the therapeutic dose. Currently used immunosuppressive drugs have not prevented the SAEs, indicating that it may be prudent to treat patients with repeated transfer of moderate doses rather than a single injection of high doses of AAV vectors. The former approach has been avoided as AAV vectors elicit neutralizing antibodies that prevent successful reapplication of serologically crossreactive vectors. Immunosuppressive regimens that block B cell responses to AAV vectors or treatments that remove AAV neutralizing antibodies thus need to be developed to allow for a shift from toxic single-dose injections of AAV vectors to repeated treatments with more moderate and safe doses. Preventing or blocking antibody responses would also allow for redosing of patients with declining transgene product expression, or for effective AAV-mediated gene transfer into patients with the pre-existing neutralizing antibodies.
Collapse
Affiliation(s)
- Hildegund C J Ertl
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Muczynski V, Nathwani AC. AAV mediated gene therapy for haemophilia B: From the early attempts to modern trials. Thromb Res 2024; 236:242-249. [PMID: 38383218 DOI: 10.1016/j.thromres.2020.12.033] [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: 05/13/2020] [Revised: 12/14/2020] [Accepted: 12/23/2020] [Indexed: 02/23/2024]
Abstract
Early gene therapy clinical trials for the treatment of Haemophilia B have been instrumental to our global understanding of gene therapy and have significantly contributed to the rapid expansion of the field. The use of adeno-associated viruses (AAVs) as vectors for gene transfer has successfully led to therapeutic expression of coagulation factor IX (FIX) in severe haemophilia B patients. Expression of FIX has remained stable following a single administration of vector for up to 8 years at levels that are clinically relevant to reduce the incidence of spontaneous bleeds and have permitted a significant change in the disease management with reduction or elimination of the need for coagulation factor concentrates. These trials have also shed light on several concerns around AAV-mediated gene transfer such as the high prevalence of pre-existing immunity against the vector capsid as well as the elevation of liver transaminases that is associated with a loss of FIX transgene expression in some patients. However, this field is advancing very rapidly with the development of increasingly more efficient strategies to overcome some of these obstacles and importantly raise the possibility of a functional cure, which has been long sought after. This review overviews the evolution of gene therapy for haemophilia B over the last two decades.
Collapse
Affiliation(s)
- Vincent Muczynski
- Department of Haematology, University College London - Cancer Institute, United Kingdom of Great Britain and Northern Ireland
| | - Amit C Nathwani
- Department of Haematology, University College London - Cancer Institute, United Kingdom of Great Britain and Northern Ireland; Katharine Dormandy Haemophilia and Thrombosis Unit, Royal Free London NHS Foundation Trust, United Kingdom of Great Britain and Northern Ireland; Freeline Therapeutics Ltd., United Kingdom of Great Britain and Northern Ireland.
| |
Collapse
|
5
|
Widner H. Immunology of cell and gene therapy approaches for neurologic diseases. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:135-144. [PMID: 39341650 DOI: 10.1016/b978-0-323-90120-8.00018-6] [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
Repair and replacement strategies using cell replacement or viral gene transfer for neurologic diseases are becoming increasingly efficacious with clinically meaningful benefits in several conditions. An increased understanding of disease processes opens up opportunities for genetic therapies and precision medicine methods aiming at disease modification or repair of lesioned neurologic structures. However, such therapeutic effects may be limited or rendered ineffective by immune responses against gene products or cells used for the intended treatments. When introducing therapeutic agents into the nervous system, a set of biologic responses are inevitably triggered, which may lead to host responses that limit the intended therapeutic goals. Factors of importance include the type of vector used and origin of cells, the mode of introduction, the degree of host immunization, and any prior exposure to the agents used. It is possible to apply specific treatments that interfere with many of these steps and factors in order to limit host immunization and to reduce or eliminate host effector reactions against the therapeutic agents. This includes immune-evading design measures of the advanced therapeutic medicinal products and various immunosuppressive processes. Limited duration of specific immune modulations may be possible under carefully monitored programs.
Collapse
Affiliation(s)
- Håkan Widner
- Department of Neurology, Skåne University Hospital, Lund, Sweden; Section for Neurology, Department of Clinical Sciences, Lund University, Lund, Sweden.
| |
Collapse
|
6
|
Blay E, Hardyman E, Morovic W. PCR-based analytics of gene therapies using adeno-associated virus vectors: Considerations for cGMP method development. Mol Ther Methods Clin Dev 2023; 31:101132. [PMID: 37964893 PMCID: PMC10641278 DOI: 10.1016/j.omtm.2023.101132] [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] [Indexed: 11/16/2023]
Abstract
The field of gene therapy has evolved and improved so that today the treatment of thousands of genetic diseases is now possible. An integral aspect of the drug development process is generating analytical methods to be used throughout clinical and commercial manufacturing. Enumeration and identification assays using genetic testing are critical to ensure the safety, efficacy, and stability of many active pharmaceutical ingredients. While nucleic acid-based methods are already reliable and rapid, there are unique biological, technological, and regulatory aspects in gene therapies that must be considered. This review surveys aspects of method development and validation using nucleic acid-based testing of gene therapies by focusing on adeno-associated virus (AAV) vectors and their co-transfection factors. Key differences between quantitative PCR and droplet digital technologies are discussed to show how improvements can be made while still adhering to regulatory guidance. Example validation parameters for AAV genome titers are described to demonstrate the scope of analytical development. Finally, several areas for improving analytical testing are presented to inspire future innovation, including next-generation sequencing and artificial intelligence. Reviewing the broad characteristics of gene therapy assessment serves as an introduction for new researchers, while clarifying processes for professionals already involved in pharmaceutical manufacturing.
Collapse
Affiliation(s)
- Emmanuel Blay
- Gene & Cell Therapy, PPD GMP Laboratories, Part of ThermoFisher Scientific, Middleton, WI, USA
| | - Elaine Hardyman
- Gene & Cell Therapy, PPD GMP Laboratories, Part of ThermoFisher Scientific, Middleton, WI, USA
| | - Wesley Morovic
- Gene & Cell Therapy, PPD GMP Laboratories, Part of ThermoFisher Scientific, Middleton, WI, USA
| |
Collapse
|
7
|
Chen M, Kim B, Jarvis MI, Fleury S, Deng S, Nouraein S, Butler S, Lee S, Chambers C, Hodges HC, Szablowski JO, Suh J, Veiseh O. Immune profiling of adeno-associated virus response identifies B cell-specific targets that enable vector re-administration in mice. Gene Ther 2023; 30:429-442. [PMID: 36372846 PMCID: PMC10183056 DOI: 10.1038/s41434-022-00371-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 09/11/2022] [Accepted: 10/04/2022] [Indexed: 11/15/2022]
Abstract
Adeno-associated virus (AAV) vector-based gene therapies can be applied to a wide range of diseases. AAV expression can last for months to years, but vector re-administration may be necessary to achieve life-long treatment. Unfortunately, immune responses against these vectors are potentiated after the first administration, preventing the clinical use of repeated administration of AAVs. Reducing the immune response against AAVs while minimizing broad immunosuppression would improve gene delivery efficiency and long-term safety. In this study, we quantified the contributions of multiple immune system components of the anti-AAV response in mice. We identified B-cell-mediated immunity as a critical component preventing vector re-administration. Additionally, we found that IgG depletion alone was insufficient to enable re-administration, suggesting IgM antibodies play an important role in the immune response against AAV. Further, we found that AAV-mediated transduction is improved in µMT mice that lack functional IgM heavy chains and cannot form mature B-cells relative to wild-type mice. Combined, our results suggest that B-cells, including non-class switched B-cells, are a potential target for therapeutics enabling AAV re-administration. Our results also suggest that the µMT mice are a potentially useful experimental model for gene delivery studies since they allow repeated dosing for more efficient gene delivery from AAVs.
Collapse
Affiliation(s)
- Maria Chen
- Department of Bioengineering, Rice University, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Boram Kim
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Maria I Jarvis
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Samantha Fleury
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Shuyun Deng
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Shirin Nouraein
- Department of Bioengineering, Rice University, Houston, TX, USA
- Rice Neuroengineering Initiative, George R. Brown School of Engineering, Rice University, Houston, TX, USA
| | - Susan Butler
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Sangsin Lee
- Department of Bioengineering, Rice University, Houston, TX, USA
- Rice Neuroengineering Initiative, George R. Brown School of Engineering, Rice University, Houston, TX, USA
| | - Courtney Chambers
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - H Courtney Hodges
- Department of Bioengineering, Rice University, Houston, TX, USA
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Jerzy O Szablowski
- Department of Bioengineering, Rice University, Houston, TX, USA
- Rice Neuroengineering Initiative, George R. Brown School of Engineering, Rice University, Houston, TX, USA
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, TX, USA
| | - Junghae Suh
- Department of Bioengineering, Rice University, Houston, TX, USA.
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, TX, USA.
- Department of BioSciences, Rice University, Houston, TX, USA.
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
8
|
Arjomandnejad M, Dasgupta I, Flotte TR, Keeler AM. Immunogenicity of Recombinant Adeno-Associated Virus (AAV) Vectors for Gene Transfer. BioDrugs 2023; 37:311-329. [PMID: 36862289 PMCID: PMC9979149 DOI: 10.1007/s40259-023-00585-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 03/03/2023]
Abstract
Recombinant adeno-associated viruses (AAVs) have emerged as promising gene delivery vehicles resulting in three US Food and Drug Administration (FDA) and one European Medicines Agency (EMA)-approved AAV-based gene therapies. Despite being a leading platform for therapeutic gene transfer in several clinical trials, host immune responses against the AAV vector and transgene have hampered their widespread application. Multiple factors, including vector design, dose, and route of administration, contribute to the overall immunogenicity of AAVs. The immune responses against the AAV capsid and transgene involve an initial innate sensing. The innate immune response subsequently triggers an adaptive immune response to elicit a robust and specific response against the AAV vector. AAV gene therapy clinical trials and preclinical studies provide important information about the immune-mediated toxicities associated with AAV, yet studies suggest preclinical models fail to precisely predict the outcome of gene delivery in humans. This review discusses the contribution of the innate and adaptive immune response against AAVs, highlighting the challenges and potential strategies to mitigate these responses, thereby enhancing the therapeutic potential of AAV gene therapy.
Collapse
Affiliation(s)
- Motahareh Arjomandnejad
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, 386 Plantation Street, Worcester, MA, 01605, USA
| | - Ishani Dasgupta
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, 386 Plantation Street, Worcester, MA, 01605, USA
| | - Terence R Flotte
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, 386 Plantation Street, Worcester, MA, 01605, USA
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Allison M Keeler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, 386 Plantation Street, Worcester, MA, 01605, USA.
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, USA.
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
9
|
Bobo TA, Samowitz PN, Robinson MI, Montes LI, Forsberg LJ, Feng R, Nicely NI, Fu H. IgG-cleavage protein allows therapeutic AAV gene delivery in passively immunized MPS IIIA mice. Gene Ther 2023; 30:377-385. [PMID: 36253453 DOI: 10.1038/s41434-022-00368-9] [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: 05/02/2022] [Revised: 07/21/2022] [Accepted: 09/27/2022] [Indexed: 11/08/2022]
Abstract
The widespread pre-existing αAAV-Abs in humans pose a critical challenge in translation of AAV gene therapy. The IgG degrading enzyme of Streptococci (IdeS) is demonstrated to specifically cleave IgG of humans and other species (not mouse). This study developed a modified new modified IdeS protein product (IdeSop). When incubated in vitro, IdeSop was shown to completely cleave human and rabbit IgGs within 6 h. To test IdeSop in a disease setting, we established a rabbitized αAAV9-Ab+ mouse by an IV infusion of purified acute αAAV9-Ab+ rabbit IgG into MPS IIIA mice, resulting in serum αAAV9-IgG at 1:6,400 and αAAV9-nAbs at 1:800. IdeSop-Ab-cleavage was shown to be dose-dependent. An IV IdeSop infusion at the effective doses resulted in rapid IgG depletion and clearance of pre-existing αAAV9-IgG and αAAV9-nAbs in rabbitized αAAV9-Abs+ MPS IIIA mice. Importantly, an IV injection of a high dose AAV9-hSGSHop vector (5 × 1013vg/kg) at 24 h post IdeSop treatment led to transduction as effective in αAAV9-Abs+ MPS IIIA mice, as in αAAV9-Abs-negative controls. We believe that transient IdeSop administration may offer a great tool to address the pre-existing-αAAV-Abs for the translation of rAAV gene therapy to treat diseases in humans, making effective rAAV gene therapy available to all patients in need.
Collapse
Affiliation(s)
- Tierra A Bobo
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Genetics and Metabolism, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Preston N Samowitz
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael I Robinson
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Laura I Montes
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lawrence J Forsberg
- Protein Production & Purification Core, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Richard Feng
- Protein Production & Purification Core, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nathan I Nicely
- Protein Production & Purification Core, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Haiyan Fu
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Division of Genetics and Metabolism, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
10
|
Li X, Wei X, Lin J, Ou L. A versatile toolkit for overcoming AAV immunity. Front Immunol 2022; 13:991832. [PMID: 36119036 PMCID: PMC9479010 DOI: 10.3389/fimmu.2022.991832] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/17/2022] [Indexed: 12/12/2022] Open
Abstract
Recombinant adeno-associated virus (AAV) is a promising delivery vehicle for in vivo gene therapy and has been widely used in >200 clinical trials globally. There are already several approved gene therapy products, e.g., Luxturna and Zolgensma, highlighting the remarkable potential of AAV delivery. In the past, AAV has been seen as a relatively non-immunogenic vector associated with low risk of toxicity. However, an increasing number of recent studies indicate that immune responses against AAV and transgene products could be the bottleneck of AAV gene therapy. In clinical studies, pre-existing antibodies against AAV capsids exclude many patients from receiving the treatment as there is high prevalence of antibodies among humans. Moreover, immune response could lead to loss of efficacy over time and severe toxicity, manifested as liver enzyme elevations, kidney injury, and thrombocytopenia, resulting in deaths of non-human primates and patients. Therefore, extensive efforts have been attempted to address these issues, including capsid engineering, plasmapheresis, IgG proteases, CpG depletion, empty capsid decoy, exosome encapsulation, capsid variant switch, induction of regulatory T cells, and immunosuppressants. This review will discuss these methods in detail and highlight important milestones along the way.
Collapse
Affiliation(s)
- Xuefeng Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital; State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Shenzhen Luohu People’s Hospital, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiaoli Wei
- Guangzhou Dezheng Biotechnology Co., Ltd., Guangzhou, China
| | - Jinduan Lin
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital; State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Li Ou
- Genemagic Biosciences, Philadelphia, PA, United States
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
- *Correspondence: Li Ou,
| |
Collapse
|
11
|
Gross DA, Tedesco N, Leborgne C, Ronzitti G. Overcoming the Challenges Imposed by Humoral Immunity to AAV Vectors to Achieve Safe and Efficient Gene Transfer in Seropositive Patients. Front Immunol 2022; 13:857276. [PMID: 35464422 PMCID: PMC9022790 DOI: 10.3389/fimmu.2022.857276] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/16/2022] [Indexed: 11/23/2022] Open
Abstract
One of the major goals of in vivo gene transfer is to achieve long-term expression of therapeutic transgenes in terminally differentiated cells. The extensive clinical experience and the recent approval of Luxturna® (Spark Therapeutics, now Roche) and Zolgensma® (AveXis, now Novartis) place vectors derived from adeno-associated viruses (AAV) among the best options for gene transfer in multiple tissues. Despite these successes, limitations remain to the application of this therapeutic modality in a wider population. AAV was originally identified as a promising virus to derive gene therapy vectors because, despite infecting humans, it was not associated with any evident disease. Thee large proportion of AAV infections in the human population is now revealing as a limitation because after exposure to wild-type AAV, anti-AAV antibodies develops and may neutralize the vectors derived from the virus. Injection of AAV in humans is generally well-tolerated although the immune system can activate after the recognition of AAV vectors capsid and genome. The formation of high-titer neutralizing antibodies to AAV after the first injection precludes vector re-administration. Thus, both pre-existing and post-treatment humoral responses to AAV vectors greatly limit a wider application of this gene transfer modality. Different methods were suggested to overcome this limitation. The extensive preclinical data available and the large clinical experience in the control of AAV vectors immunogenicity are key to clinical translation and to demonstrate the safety and efficacy of these methods and ultimately bring a curative treatment to patients.
Collapse
Affiliation(s)
- David-Alexandre Gross
- Genethon, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951, Evry, France
| | - Novella Tedesco
- Genethon, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951, Evry, France
| | - Christian Leborgne
- Genethon, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951, Evry, France
| | - Giuseppe Ronzitti
- Genethon, Evry, France
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951, Evry, France
| |
Collapse
|
12
|
Muhuri M, Maeda Y, Ma H, Ram S, Fitzgerald KA, Tai PW, Gao G. Overcoming innate immune barriers that impede AAV gene therapy vectors. J Clin Invest 2021; 131:143780. [PMID: 33393506 DOI: 10.1172/jci143780] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The field of gene therapy has made considerable progress over the past several years. Adeno-associated virus (AAV) vectors have emerged as promising and attractive tools for in vivo gene therapy. Despite the recent clinical successes achieved with recombinant AAVs (rAAVs) for therapeutics, host immune responses against the vector and transgene product have been observed in numerous preclinical and clinical studies. These outcomes have hampered the advancement of AAV gene therapies, preventing them from becoming fully viable and safe medicines. The human immune system is multidimensional and complex. Both the innate and adaptive arms of the immune system seem to play a concerted role in the response against rAAVs. While most efforts have been focused on the role of adaptive immunity and developing ways to overcome it, the innate immune system has also been found to have a critical function. Innate immunity not only mediates the initial response to the vector, but also primes the adaptive immune system to launch a more deleterious attack against the foreign vector. This Review highlights what is known about innate immune responses against rAAVs and discusses potential strategies to circumvent these pathways.
Collapse
Affiliation(s)
- Manish Muhuri
- Horae Gene Therapy Center.,Department of Microbiology and Physiological Systems.,VIDE Program
| | - Yukiko Maeda
- Horae Gene Therapy Center.,VIDE Program.,Department of Medicine
| | | | - Sanjay Ram
- Division of Infectious Diseases and Immunology
| | | | - Phillip Wl Tai
- Horae Gene Therapy Center.,Department of Microbiology and Physiological Systems.,VIDE Program
| | - Guangping Gao
- Horae Gene Therapy Center.,Department of Microbiology and Physiological Systems.,Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
13
|
Zhong C, Jiang W, Wang Y, Sun J, Wu X, Zhuang Y, Xiao X. Repeated systemic dosing of AAV vectors in immunocompetent mice after blockade of T-cell costimulatory pathways. Hum Gene Ther 2021; 33:290-300. [PMID: 34486389 DOI: 10.1089/hum.2021.129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Neutralizing antibodies (NAbs) strongly limit adeno-associated virus (AAV) vector transduction and repeated administration. Previous studies have shown that NAbs induced by AAVs are associated with T and B cell activation and that the B7/CD28 and CD40/CD40L costimulation signaling pathways are involved. CTLA4 and CD40 are vital molecules that participate in the costimulatory pathway. In this study, we evaluated CTLA4-Ig and CD40-Ig immunosuppressive efficacies through AAV and investigated their effects on the feasibility for multiple systemic administrations of AAV vectors. The results showed that a single administration of AAV vector carrying either CTLA4-Ig alone or with CD40-Ig could greatly reduce the level of NAbs. An AAV serotype-specific immune tolerance could be successfully established, which enabled repeated, i.e., second and third, systemic administration of AAV vectors in the same mice. A combination of CTLA4-Ig and CD40-Ig delivered via AAV vectors significantly inhibited T and B cell activations without affecting immune response to the total immunoglobulin G (IgG) production and cytokines. Interestingly, exogenous gene expression significantly improved after multiple administrations of AAV vector in vivo. Our study generates a reliable and effective method for repeated dosing of AAV vectors that is needed on gene therapy.
Collapse
Affiliation(s)
- Chen Zhong
- East China University of Science and Technology, 47860, State Key Laboratory of Bioreactor Engineering, School of Biotechnology, shanghai, China;
| | - Wei Jiang
- East China University of Science and Technology, 47860, Shanghai, Shanghai, China;
| | - Yefan Wang
- East China University of Science and Technology, 47860, Shanghai, Shanghai, China;
| | - Junjiang Sun
- The University of North Carolina at Chapel Hill, 2331, Gene Therapy Center, Chapel Hill, North Carolina, United States.,University of North Carolina at Chapel Hill Eshelman School of Pharmacy, 15521, Division of Molecular Pharmaceutics, Chapel Hill, North Carolina, United States;
| | - Xia Wu
- East China University of Science and Technology, 47860, School of Pharmacy, Shanghai, Shanghai, China;
| | - Yingping Zhuang
- East China University of Science and Technology, 47860, State Key Laboratory of Bioreactor Engineering, School of Biotechnology, Shanghai, Shanghai, China;
| | - Xiao Xiao
- East China University of Science and Technology, 47860, School of Pharmacy, Shanghai, Shanghai, China;
| |
Collapse
|
14
|
Ertl HCJ. T Cell-Mediated Immune Responses to AAV and AAV Vectors. Front Immunol 2021; 12:666666. [PMID: 33927727 PMCID: PMC8076552 DOI: 10.3389/fimmu.2021.666666] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/22/2021] [Indexed: 12/18/2022] Open
Abstract
Adeno-associated virus (AAV)-mediated gene transfer has benefited patients with inherited diseases, such as hemophilia B, by achieving long-term expression of the therapeutic transgene. Nevertheless, challenges remain due to rejection of AAV-transduced cells, which in some, but not all, patients can be prevented by immunosuppression. It is assumed that CD8+ T cells induced by natural infections with AAVs are recalled by the AAV vector's capsid and upon activation eliminate cells expressing the degraded capsid antigens. Alternatively, it is feasible that AAV vectors, especially if given at high doses, induce de novo capsid- or transgene product-specific T cell responses. This chapter discusses CD8+ T cell responses to AAV infections and AAV gene transfer and avenues to prevent their activation or block their effector functions.
Collapse
|
15
|
Whitehead M, Osborne A, Yu-Wai-Man P, Martin K. Humoral immune responses to AAV gene therapy in the ocular compartment. Biol Rev Camb Philos Soc 2021; 96:1616-1644. [PMID: 33837614 DOI: 10.1111/brv.12718] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 12/11/2022]
Abstract
Viral vectors can be utilised to deliver therapeutic genes to diseased cells. Adeno-associated virus (AAV) is a commonly used viral vector that is favoured for its ability to infect a wide range of tissues whilst displaying limited toxicity and immunogenicity. Most humans harbour anti-AAV neutralising antibodies (NAbs) due to subclinical infections by wild-type virus during infancy and these pre-existing NAbs can limit the efficiency of gene transfer depending on the target cell type, route of administration and choice of serotype. Vector administration can also result in de novo NAb synthesis that could limit the opportunity for repeated gene transfer to diseased sites. A number of strategies have been described in preclinical models that could circumvent NAb responses in humans, however, the successful translation of these innovations into the clinical arena has been limited. Here, we provide a comprehensive review of the humoral immune response to AAV gene therapy in the ocular compartment. We cover basic AAV biology and clinical application, the role of pre-existing and induced NAbs, and possible approaches to overcoming antibody responses. We conclude with a framework for a comprehensive strategy for circumventing humoral immune responses to AAV in the future.
Collapse
Affiliation(s)
- Michael Whitehead
- John Van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge, U.K
| | - Andrew Osborne
- John Van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge, U.K
| | - Patrick Yu-Wai-Man
- John Van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge, U.K.,MRC Mitochondrial Biology Unit, Department of Clinical Neurosciences, University of Cambridge, Cambridge, U.K.,NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, U.K
| | - Keith Martin
- John Van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge, U.K.,Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, U.K.,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC, Australia.,Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
16
|
Chu WS, Ng J. Immunomodulation in Administration of rAAV: Preclinical and Clinical Adjuvant Pharmacotherapies. Front Immunol 2021; 12:658038. [PMID: 33868303 PMCID: PMC8049138 DOI: 10.3389/fimmu.2021.658038] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/05/2021] [Indexed: 12/26/2022] Open
Abstract
Recombinant adeno-associated virus (rAAV) has attracted a significant research focus for delivering genetic therapies to target cells. This non-enveloped virus has been trialed in many clinical-stage therapeutic strategies but important obstacle in clinical translation is the activation of both innate and adaptive immune response to the protein capsid, vector genome and transgene product. In addition, the normal population has pre-existing neutralizing antibodies against wild-type AAV, and cross-reactivity is observed between different rAAV serotypes. While extent of response can be influenced by dosing, administration route and target organ(s), these pose concerns over reduction or complete loss of efficacy, options for re-administration, and other unwanted immunological sequalae such as local tissue damage. To reduce said immunological risks, patients are excluded if they harbor anti-AAV antibodies or have received gene therapy previously. Studies have incorporated immunomodulating or suppressive regimens to block cellular and humoral immune responses such as systemic corticosteroids pre- and post-administration of Luxturna® and Zolgensma®, the two rAAV products with licensed regulatory approval in Europe and the United States. In this review, we will introduce the current pharmacological strategies to immunosuppress or immunomodulate the host immune response to rAAV gene therapy.
Collapse
Affiliation(s)
- Wing Sum Chu
- Pharmacy Department, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Joanne Ng
- Gene Transfer Technology Group, Department of Maternal and Fetal Medicine, EGA Institute for Women's Health, University College London, London, United Kingdom
| |
Collapse
|
17
|
Comprehensive characterization and quantification of adeno associated vectors by size exclusion chromatography and multi angle light scattering. Sci Rep 2021; 11:3012. [PMID: 33542328 PMCID: PMC7862616 DOI: 10.1038/s41598-021-82599-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 01/22/2021] [Indexed: 01/10/2023] Open
Abstract
Adeno associated virus (AAV) capsids are a leading modality for in vivo gene delivery. Complete and precise characterization of capsid particles, including capsid and vector genome concentration, is necessary to safely and efficaciously dose patients. Size exclusion chromatography (SEC) coupled to multiangle light scattering (MALS) offers a straightforward approach to comprehensively characterize AAV capsids. The current study demonstrates that this method provides detailed AAV characterization information, including but not limited to aggregation profile, size-distribution, capsid content, capsid molar mass, encapsidated DNA molar mass, and total capsid and vector genome titer. Currently, multiple techniques are required to generate this information, with varying accuracy and precision. In the current study, a new series of equations for SEC-MALS are used in tandem with intrinsic properties of the capsids and encapsidated DNA to quantify multiple physical AAV attributes in one 20-min run with minimal sample manipulation, high accuracy, and high precision. These novel applications designate this well-established method as a powerful tool for product development and process analytics in future gene therapy programs.
Collapse
|
18
|
Matagne V, Borloz E, Ehinger Y, Saidi L, Villard L, Roux JC. Severe offtarget effects following intravenous delivery of AAV9-MECP2 in a female mouse model of Rett syndrome. Neurobiol Dis 2020; 149:105235. [PMID: 33383186 DOI: 10.1016/j.nbd.2020.105235] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 01/06/2023] Open
Abstract
Rett syndrome (RTT) is a severe X-linked neurodevelopmental disorder that is primarily caused by mutations in the methyl CpG binding protein 2 gene (MECP2). RTT is the second most prevalent genetic cause of intellectual disability in girls, and there is currently no cure for the disease. We have previously shown that gene therapy using a self-complementary AAV9 viral vector expressing a codon-optimized Mecp2 version (AAV9-MCO) significantly improved symptoms and increased survival in male Mecp2-deficient mice. Here, we pursued our studies and investigated the safety and efficacy of long-term gene therapy in the genetically relevant RTT mouse model: the heterozygous (HET) Mecp2 deficient female mouse. These mice were injected with the AAV9-MCO vector through the tail vein and an array of behavioral tests was performed. At 16- and 30-weeks post-injection, this treatment was able to rescue apneas and improved the spontaneous locomotor deficits and circadian locomotor activity in Mecp2 HET mice treated with AAV9-MCO at a dose of 5 × 1011 vg/mouse. To examine whether a higher dose of vector could result in increased improvements, we injected Mecp2 HET mice with a higher MCO vector dose (1012 vg/mouse), which resulted in some severe, sometimes lethal, side effects. In order to confirm these effects, a new cohort of Mecp2 HET mice were administered increasing doses of MCO vector (1011, 5 × 1011 and 1012 vg/mouse). Again, two weeks after vector administration, some Mecp2 HET mice were found dead while others displayed severe side effects and had to be euthanized. These deleterious effects were not observed in Mecp2 HET mice injected with a high dose of AAV9-GFP and were directly proportionate to vector dosage (0, 23 or 54% mortality at an AAV9-MCO dose of 1011, 5 × 1011, 1012 vg/mouse, respectively), and no such lethality was observed in wild-type (WT) mice. In the Mecp2 HET mice treated with the high and medium AAV9-MCO doses, blood chemistry analysis and post-mortem histology showed liver damage with drastically elevated levels of liver transaminases and disorganized liver architecture. Apoptosis was confirmed by the presence of TUNEL- and cleaved-caspase 3-positive cells in the Mecp2 HET mice treated with the higher doses of AAV9-MCO. We then studied the involvement of the unfolded protein response (UPR) in triggering apoptosis since it can be activated by AAV vectors. Increased expression of the C/EBP homologous protein (CHOP), one of UPR downstream effectors, was confirmed in Mecp2 HET mice after vector administration. The toxic reaction seen in some treated mice indicates that, although gene therapy for RTT improved breathing deficits observed in Mecp2 HET mice, further studies are needed to better understand the underlying mechanisms and caution must be exercised before similar attempts are undertaken in female Rett patients.
Collapse
Affiliation(s)
- Valerie Matagne
- Aix Marseille Univ, INSERM, MMG, U1251, Faculté de médecine Timone, 13385 Marseille, France
| | - Emilie Borloz
- Aix Marseille Univ, INSERM, MMG, U1251, Faculté de médecine Timone, 13385 Marseille, France
| | - Yann Ehinger
- Aix Marseille Univ, INSERM, MMG, U1251, Faculté de médecine Timone, 13385 Marseille, France
| | - Lydia Saidi
- Aix Marseille Univ, INSERM, MMG, U1251, Faculté de médecine Timone, 13385 Marseille, France
| | - Laurent Villard
- Aix Marseille Univ, INSERM, MMG, U1251, Faculté de médecine Timone, 13385 Marseille, France
| | - Jean-Christophe Roux
- Aix Marseille Univ, INSERM, MMG, U1251, Faculté de médecine Timone, 13385 Marseille, France.
| |
Collapse
|
19
|
Frentsch M, Japp AS, Dingeldey M, Matzmohr N, Thiel A, Scheiflinger F, Reipert BM, de la Rosa M. Blockade of the costimulatory CD28-B7 family signal axis enables repeated application of AAV8 gene vectors. J Thromb Haemost 2020; 18:1075-1080. [PMID: 32011092 PMCID: PMC7318590 DOI: 10.1111/jth.14757] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 12/31/2019] [Accepted: 01/27/2020] [Indexed: 12/30/2022]
Abstract
Adeno-associated virus serotype 8 (AAV8) gene therapy has shown efficacy in several clinical trials and is considered a highly promising technology to treat monogenic diseases such as hemophilia A and B. However, a major drawback of AAV8 gene therapy is that it can be applied only once because anti-AAV8 immunity develops after the first treatment. Readministration may be required in patients who are expected to need redosing, eg, due to organ growth, or to boost suboptimal expression levels, but no redosing protocol has been established. We have developed a preventive immune-suppressive protocol for a human factor IX (FIX) vector with an intended dose of ~5 × 1011 vg/kg that inhibits the development of anti-AAV8 neutralizing-antibody (NAb) responses and anti-AAV8 T-cell responses using CTLA4-IgG (abatacept). In a preclinical model, transient treatment with abatacept during initial human FIX gene therapy efficiently inhibited the generation of AAV8-specific cellular and humoral responses, and thus permitted redosing of FIX. Furthermore, our data suggest that by suppression of anti-AAV8 NAb responses after the second higher dose (4 × 1012 vg/kg) this protocol can be used to enable redosing up to such high doses. An additional advantage of CTLA4-IgG blocking CD28-mediated signals is its potential suppression of AAV8-specific cytotoxic CD8 T-cell responses, which are believed to kill transduced hepatocytes and might interfere with a successful readministration. Redosing protocols using approved drugs would be beneficial for patients because they could effortlessly be applied in clinical trials and enable safe and efficient treatment options for patients undergoing AAV8 gene therapy.
Collapse
Affiliation(s)
- Marco Frentsch
- Berlin‐Brandenburg Center for Regenerative TherapiesCharité‐UniversitätsmedizinBerlinGermany
| | - Alberto Sada Japp
- Berlin‐Brandenburg Center for Regenerative TherapiesCharité‐UniversitätsmedizinBerlinGermany
| | - Manuela Dingeldey
- Berlin‐Brandenburg Center for Regenerative TherapiesCharité‐UniversitätsmedizinBerlinGermany
| | - Nadine Matzmohr
- Berlin‐Brandenburg Center for Regenerative TherapiesCharité‐UniversitätsmedizinBerlinGermany
| | - Andreas Thiel
- Berlin‐Brandenburg Center for Regenerative TherapiesCharité‐UniversitätsmedizinBerlinGermany
| | | | - Birgit M. Reipert
- Baxalta Innovations GmbHA Member of the Takeda Group of CompaniesViennaAustria
| | - Maurus de la Rosa
- Baxalta Innovations GmbHA Member of the Takeda Group of CompaniesViennaAustria
| |
Collapse
|
20
|
Immune Response Mechanisms against AAV Vectors in Animal Models. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 17:198-208. [PMID: 31970198 PMCID: PMC6965504 DOI: 10.1016/j.omtm.2019.12.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Early preclinical studies in rodents and other species did not reveal that vector or transgene immunity would present a significant hurdle for sustained gene expression. While there was early evidence of mild immune responses to adeno-associated virus (AAV) in preclinical studies, it was generally believed that these responses were too weak and transient to negatively impact sustained transduction. However, translation of the cumulative success in treating hemophilia B in rodents and dogs with an AAV2-F9 vector to human studies was not as successful. Despite significant progress in recent clinical trials for hemophilia, new immunotoxicities to AAV and transgene are emerging in humans that require better animal models to assess and overcome these responses. The animal models designed to address these immune complications have provided critical information to assess how vector dose, vector capsid processing, vector genome, difference in serotypes, and variations in vector delivery route can impact immunity and to develop approaches for overcoming pre-existing immunity. Additionally, a comprehensive dissection of innate, adaptive, and regulatory responses to AAV vectors in preclinical studies has provided a framework that can be utilized for development of immunomodulatory therapies to overcome or bypass immune responses and for developing strategic approaches toward engineering stealth AAV vectors that can circumvent immunity.
Collapse
|
21
|
Pouzolles M, Machado A, Guilbaud M, Irla M, Gailhac S, Barennes P, Cesana D, Calabria A, Benedicenti F, Sergé A, Raman I, Li QZ, Montini E, Klatzmann D, Adjali O, Taylor N, Zimmermann VS. Intrathymic adeno-associated virus gene transfer rapidly restores thymic function and long-term persistence of gene-corrected T cells. J Allergy Clin Immunol 2019; 145:679-697.e5. [PMID: 31513879 DOI: 10.1016/j.jaci.2019.08.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 07/28/2019] [Accepted: 08/05/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Patients with T-cell immunodeficiencies are generally treated with allogeneic hematopoietic stem cell transplantation, but alternatives are needed for patients without matched donors. An innovative intrathymic gene therapy approach that directly targets the thymus might improve outcomes. OBJECTIVE We sought to determine the efficacy of intrathymic adeno-associated virus (AAV) serotypes to transduce thymocyte subsets and correct the T-cell immunodeficiency in a zeta-associated protein of 70 kDa (ZAP-70)-deficient murine model. METHODS AAV serotypes were injected intrathymically into wild-type mice, and gene transfer efficiency was monitored. ZAP-70-/- mice were intrathymically injected with an AAV8 vector harboring the ZAP70 gene. Thymus structure, immunophenotyping, T-cell receptor clonotypes, T-cell function, immune responses to transgenes and autoantibodies, vector copy number, and integration were evaluated. RESULTS AAV8, AAV9, and AAV10 serotypes all transduced thymocyte subsets after in situ gene transfer, with transduction of up to 5% of cells. Intrathymic injection of an AAV8-ZAP-70 vector into ZAP-70-/- mice resulted in a rapid thymocyte differentiation associated with the development of a thymic medulla. Strikingly, medullary thymic epithelial cells expressing the autoimmune regulator were detected within 10 days of gene transfer, correlating with the presence of functional effector and regulatory T-cell subsets with diverse T-cell receptor clonotypes in the periphery. Although thymocyte reconstitution was transient, gene-corrected peripheral T cells harboring approximately 1 AAV genome per cell persisted for more than 40 weeks, and AAV vector integration was detected. CONCLUSIONS Intrathymic AAV-transduced progenitors promote a rapid restoration of the thymic architecture, with a single wave of thymopoiesis generating long-term peripheral T-cell function.
Collapse
Affiliation(s)
- Marie Pouzolles
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Alice Machado
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Mickaël Guilbaud
- INSERM UMR1089, Université de Nantes, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Magali Irla
- Center of Immunology Marseille-Luminy (CIML), INSERM U1104, CNRS UMR7280, Aix-Marseille Université UM2, Marseille, France
| | - Sarah Gailhac
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Pierre Barennes
- Sorbonne Université, INSERM, Immunology-Immunopathology-Immunotherapy (i3), Paris, France
| | - Daniela Cesana
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Calabria
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Fabrizio Benedicenti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Arnauld Sergé
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Indu Raman
- Microarray Core Facility, University of Texas Southwestern Medical Center, Dallas, Tex
| | - Quan-Zhen Li
- Microarray Core Facility, University of Texas Southwestern Medical Center, Dallas, Tex; Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Tex
| | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - David Klatzmann
- Sorbonne Université, INSERM, Immunology-Immunopathology-Immunotherapy (i3), Paris, France; AP-HP, Hôpital Pitié-Salpêtrière, Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), Paris, France
| | - Oumeya Adjali
- INSERM UMR1089, Université de Nantes, Centre Hospitalier Universitaire de Nantes, Nantes, France.
| | - Naomi Taylor
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France; Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md.
| | - Valérie S Zimmermann
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
22
|
Immune-orthogonal orthologues of AAV capsids and of Cas9 circumvent the immune response to the administration of gene therapy. Nat Biomed Eng 2019; 3:806-816. [PMID: 31332341 PMCID: PMC6783354 DOI: 10.1038/s41551-019-0431-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 06/16/2019] [Indexed: 12/19/2022]
Abstract
Protein-based therapeutics can activate the adaptive immune system and lead to the production of neutralizing antibodies and to cytotoxic-T-cell-mediated clearance of the treated cells. Here, we show that the sequential use of immune-orthogonal orthologues of the CRISPR-associated protein 9 (Cas9) and of adeno-associated viruses (AAVs) eludes adaptive immune responses and enables effective gene editing from repeated dosing. We compared total sequence similarities and predicted binding strengths to class-I and class-II major-histocompatibility-complex proteins for 284 DNA-targeting and 84 RNA-targeting CRISPR effectors, and for 167 AAV VP1-capsid-protein orthologues. We predict the absence of cross-reactive immune responses for 79% of the DNA-targeting Cas orthologs, which we validate for three Cas9 orthologs in mice, yet anticipate broad immune cross-reactivity among the AAV serotypes. We also show that efficacious in vivo gene editing is uncompromised when using multiple dosing with orthologues of AAVs and Cas9 in mice previously immunized against the AAV vector and the Cas9 payload. Multiple dosing with protein orthologues may allow for sequential regimens of protein therapeutics that circumvent pre-existing immunity or induced immunity.
Collapse
|
23
|
Recino A, Gan SU, Sia KC, Sawyer Y, Trendell J, Kay R, Gribble FM, Reimann F, Foale R, Notaridou M, Holmes N, Lever A, Lee KO, Nathwani A, Cooke A, Calne R, Wallberg M. Immunosuppression overcomes insulin- and vector-specific immune responses that limit efficacy of AAV2/8-mediated insulin gene therapy in NOD mice. Gene Ther 2019; 26:40-56. [PMID: 30514969 PMCID: PMC6514884 DOI: 10.1038/s41434-018-0052-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/09/2018] [Accepted: 11/06/2018] [Indexed: 12/23/2022]
Abstract
We report the restoration of euglycaemia in chemically induced diabetic C57BL/6 mice and spontaneously diabetic Non Obese Diabetic (NOD) mice by intravenous systemic administration of a single-stranded adeno-associated virus (ssAAV2/8) codon optimised (co) vector encoding furin cleavable human proinsulin under a liver-specific promoter. There were no immunological barriers to efficacy of insulin gene therapy in chemically induced C57BL/6 mice, which enjoyed long-lasting correction of hyperglycaemia after therapy, up to 250 days. Euglycaemia was also restored in spontaneously diabetic NOD mice, although these mice required a 7-10-fold higher dose of vector to achieve similar efficacy as the C57BL/6 mice and the immunodeficient NODscid mice. We detected CD8+ T cell reactivity to insulin and mild inflammatory infiltration in the livers of gene therapy recipient NOD mice, neither of which were observed in the treated C57BL/6 mice. Efficacy of the gene therapy in NOD mice was partially improved by targeting the immune system with anti-CD4 antibody treatment, while transfer of NOD mouse AAV2/8-reactive serum to recipients prevented successful restoration of euglycaemia in AAV2/8-HLP-hINSco-treated NODscid mice. Our data indicate that both immune cells and antibodies form a barrier to successful restoration of euglycaemia in autoimmune diabetic recipient mice with insulin gene therapy, but that this barrier can be overcome by increasing the dose of vector and by suppressing immune responses.
Collapse
Affiliation(s)
- Asha Recino
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK.
| | - Shu Uin Gan
- Department of Surgery, National University of Singapore, Singapore, Singapore
| | - Kian Chuan Sia
- Department of Surgery, National University of Singapore, Singapore, Singapore
| | - Yvonne Sawyer
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Jenny Trendell
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Richard Kay
- Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Fiona M Gribble
- Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Frank Reimann
- Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Rob Foale
- Dick White Referrals, Station Farm, Six Mile Bottom, Suffolk, UK
| | | | - Nick Holmes
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Andrew Lever
- Department of Medicine, University of Cambridge, Cambridge, UK
- Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Kok Onn Lee
- Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Amit Nathwani
- Department of Haematology, UCL Cancer Institute, London, UK
| | - Anne Cooke
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Roy Calne
- Department of Surgery, National University of Singapore, Singapore, Singapore
- Department of Medicine, National University of Singapore, Singapore, Singapore
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Maja Wallberg
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK.
| |
Collapse
|
24
|
Ellsworth JL, O'Callaghan M, Rubin H, Seymour A. Low Seroprevalence of Neutralizing Antibodies Targeting Two Clade F AAV in Humans. HUM GENE THER CL DEV 2019; 29:60-67. [PMID: 29624457 DOI: 10.1089/humc.2017.239] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
To assess the therapeutic utility of AAVHSC15 and AAVHSC17, two recently described Clade F adeno-associated viruses (AAVs), the seroprevalence of neutralizing antibodies (NAbs) to these AAVs was assessed in a representative human population and compared to that of AAV9. NAb levels were measured in 100 unique human sera of different races (34, Black, 33 Caucasian, and 33 Hispanic) and sex (49% female, 51% male) collected within the United States. Fifty-six sera were tested in Huh7 cells and 44 sera were tested in 2V6.11 cells with vectors packaged with either a CMV-promoter upstream of LacZ or a CBA-promoter upstream of Firefly Luciferase, respectively. For AAVHSC15, AAVHSC17, and AAV9, 24/100 (24%), 21/100 (21%), and 17/100 (17%), respectively, of all sera tested were seropositive for NAbs using 50% inhibition of cellular transduction at a 1/16 dilution of serum as cutoff for seropositivity. Only 6% of positive sera had titers of 1/150 to 1/340, indicating that the majority of positive sera were of low titer. Significant cross-reactivity of NAbs across all three AAV serotypes was observed. These data show that approximately 80% of humans evaluated were seronegative for pre-existing NAbs to the AAV serotypes tested, suggesting that the vast majority of human subjects would be amenable to therapeutic intervention with Clade F AAVs.
Collapse
|
25
|
Palaschak B, Herzog RW, Markusic DM. AAV-Mediated Gene Delivery to the Liver: Overview of Current Technologies and Methods. Methods Mol Biol 2019; 1950:333-360. [PMID: 30783984 DOI: 10.1007/978-1-4939-9139-6_20] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adeno-associated virus (AAV) vectors to treat liver-specific genetic diseases are the focus of several ongoing clinical trials. The ability to give a peripheral injection of virus that will successfully target the liver is one of many attractive features of this technology. Although initial studies of AAV liver gene transfer revealed some limitations, extensive animal modeling and further clinical development have helped solve some of these issues, resulting in several successful clinical trials that have reached curative levels of clotting factor expression in hemophilia. Looking beyond gene replacement, recent technologies offer the possibility for AAV liver gene transfer to directly repair deficient genes and potentially treat autoimmune disease.
Collapse
Affiliation(s)
- Brett Palaschak
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Roland W Herzog
- Department of Pediatrics, University of Florida, Gainesville, FL, USA.,Department of Pediatrics, Indiana University, Indianapolis, IN, USA
| | - David M Markusic
- Department of Pediatrics, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
26
|
Ellsworth JL, OCallaghan M, Rubin H, Seymour A. Low Seroprevalence of Neutralizing Antibodies Targeting Two Clade F AAV in Humans. HUM GENE THER CL DEV 2018. [DOI: 10.1089/hum.2017.239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Jeff L Ellsworth
- Homology Medicines, Inc., 45 Wiggins Ave, Bedford, Massachusetts, United States, 01730,
| | | | - Hillard Rubin
- Homology Medicines, Inc., Bedford, Massachusetts, United States,
| | - Albert Seymour
- Homology Medicines, Inc., Bedford, Massachusetts, United States,
| |
Collapse
|
27
|
Liu Y, Fan Z, Li K, Deng F, Xiong Y, Liang M, Ge J. An optimized gene transfection system in WERI-Rb1 cells. Int J Mol Med 2017; 40:801-813. [PMID: 28713896 PMCID: PMC5547939 DOI: 10.3892/ijmm.2017.3058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 06/30/2017] [Indexed: 11/24/2022] Open
Abstract
The pathogenesis of Rb1 gene inactivation indicates that gene therapy could be a promising treatment for retinoblastoma. An appropriate gene transfer system is the basis for successful gene therapy; however, little attention has been given to an effective gene transfer system for retinoblastoma therapy in previous studies. This study was designed to provide an optimized transgene system for WERI-Rb1 cells (W-RBCs). Green fluorescent protein (GFP) was adopted as a reporter. Four classic viral vectors based on retroviruses, recombinant adeno-associated viruses (rAAV2, rAAV2/1), lentiviruses (LVs) and a novel non-viral vector X-treme HP reagent were adopted for W-RBC gene transfection. The efficacy and cytotoxicity were comprehensively compared among the different vectors through GFP expression and the trypan blue exclusion test. Furthermore, the serum and cell culture status were also optimized for better transfection. Cells transfected by rAAV2/1 expressed more GFP protein and exhibited less staining with trypan blue, compared to the rAAV2 counterpart. However, in comparison to the retroviral group, both the rAAV2/1 and LV groups had considerably less GFP+ cells. Interestingly, the X-treme HP presented a similar GFP transfection capacity to the retroviral vector, but with a much lower cytotoxicity. Furthermore, there were more GFP+ cells in a suspended condition than that in an adherent culture. Moreover, cells in a serum-positive system expressed more GFP, while cells in a serum-free system showed lower GFP expression and higher cytotoxicity. In conclusion, the retroviral vector and the X-treme HP are effective for W-RBC gene transfection, while the X-treme HP is more preferable due to its lower cytotoxicity. Moreover, the suspended cell culture system is superior to the adherent system, and the serum protects cell viability and facilitates the gene transfection of W-RBCs. This study presents an effective, convenient, and low toxic transfection system for gene delivery in W-RBCs and provides a promising system for further gene therapy of retinoblastoma.
Collapse
Affiliation(s)
- Ying Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Zhigang Fan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Kang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Fei Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Yunfan Xiong
- The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Meixin Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
28
|
Baruteau J, Waddington SN, Alexander IE, Gissen P. Gene therapy for monogenic liver diseases: clinical successes, current challenges and future prospects. J Inherit Metab Dis 2017; 40:497-517. [PMID: 28567541 PMCID: PMC5500673 DOI: 10.1007/s10545-017-0053-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 02/08/2023]
Abstract
Over the last decade, pioneering liver-directed gene therapy trials for haemophilia B have achieved sustained clinical improvement after a single systemic injection of adeno-associated virus (AAV) derived vectors encoding the human factor IX cDNA. These trials demonstrate the potential of AAV technology to provide long-lasting clinical benefit in the treatment of monogenic liver disorders. Indeed, with more than ten ongoing or planned clinical trials for haemophilia A and B and dozens of trials planned for other inherited genetic/metabolic liver diseases, clinical translation is expanding rapidly. Gene therapy is likely to become an option for routine care of a subset of severe inherited genetic/metabolic liver diseases in the relatively near term. In this review, we aim to summarise the milestones in the development of gene therapy, present the different vector tools and their clinical applications for liver-directed gene therapy. AAV-derived vectors are emerging as the leading candidates for clinical translation of gene delivery to the liver. Therefore, we focus on clinical applications of AAV vectors in providing the most recent update on clinical outcomes of completed and ongoing gene therapy trials and comment on the current challenges that the field is facing for large-scale clinical translation. There is clearly an urgent need for more efficient therapies in many severe monogenic liver disorders, which will require careful risk-benefit analysis for each indication, especially in paediatrics.
Collapse
Affiliation(s)
- Julien Baruteau
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, London, UK.
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
- Gene Transfer Technology Group, Institute for Women's Health, University College London, London, UK.
| | - Simon N Waddington
- Gene Transfer Technology Group, Institute for Women's Health, University College London, London, UK
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ian E Alexander
- Gene Therapy Research Unit, The Children's Hospital at Westmead and Children's Medical Research Institute, Westmead, Australia
- Discipline of Child and Adolescent Health, University of Sydney, Sydney, Australia
| | - Paul Gissen
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, London, UK
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| |
Collapse
|
29
|
Choudhury SR, Hudry E, Maguire CA, Sena-Esteves M, Breakefield XO, Grandi P. Viral vectors for therapy of neurologic diseases. Neuropharmacology 2017; 120:63-80. [PMID: 26905292 PMCID: PMC5929167 DOI: 10.1016/j.neuropharm.2016.02.013] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 02/07/2016] [Accepted: 02/15/2016] [Indexed: 12/21/2022]
Abstract
Neurological disorders - disorders of the brain, spine and associated nerves - are a leading contributor to global disease burden with a shockingly large associated economic cost. Various treatment approaches - pharmaceutical medication, device-based therapy, physiotherapy, surgical intervention, among others - have been explored to alleviate the resulting extent of human suffering. In recent years, gene therapy using viral vectors - encoding a therapeutic gene or inhibitory RNA into a "gutted" viral capsid and supplying it to the nervous system - has emerged as a clinically viable option for therapy of brain disorders. In this Review, we provide an overview of the current state and advances in the field of viral vector-mediated gene therapy for neurological disorders. Vector tools and delivery methods have evolved considerably over recent years, with the goal of providing greater and safer genetic access to the central nervous system. Better etiological understanding of brain disorders has concurrently led to identification of improved therapeutic targets. We focus on the vector technology, as well as preclinical and clinical progress made thus far for brain cancer and various neurodegenerative and neurometabolic disorders, and point out the challenges and limitations that accompany this new medical modality. Finally, we explore the directions that neurological gene therapy is likely to evolve towards in the future. This article is part of the Special Issue entitled "Beyond small molecules for neurological disorders".
Collapse
Affiliation(s)
- Sourav R Choudhury
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Eloise Hudry
- Alzheimer's Disease Research Unit, Harvard Medical School & Massachusetts General Hospital, Charlestown, MA 02129, USA.
| | - Casey A Maguire
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA.
| | - Miguel Sena-Esteves
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Xandra O Breakefield
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA.
| | - Paola Grandi
- Department of Neurological Surgery, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15219, USA.
| |
Collapse
|
30
|
Successful Repeated Hepatic Gene Delivery in Mice and Non-human Primates Achieved by Sequential Administration of AAV5 ch and AAV1. Mol Ther 2017; 25:1831-1842. [PMID: 28596114 DOI: 10.1016/j.ymthe.2017.05.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 05/02/2017] [Accepted: 05/02/2017] [Indexed: 12/31/2022] Open
Abstract
In the gene therapy field, re-administration of adeno-associated virus (AAV) is an important topic because a decrease in therapeutic protein expression might occur over time. However, an efficient re-administration with the same AAV serotype is impossible due to serotype-specific, anti-AAV neutralizing antibodies (NABs) that are produced after initial AAV treatment. To address this issue, we explored the feasibility of using chimeric AAV serotype 5 (AAV5ch) and AAV1 for repeated liver-targeted gene delivery. To develop a relevant model, we immunized animals with a high dose of AAV5ch-human secreted embryonic alkaline phosphatase (hSEAP) that generates high levels of anti-AAV5ch NAB. Secondary liver transduction with the same dose of AAV1-human factor IX (hFIX) in the presence of high levels of anti-AAV5ch NAB proved to be successful because expression/activity of both reporter transgenes was observed. This is the first time that two different transgenes are shown to be produced by non-human primate (NHP) liver after sequential administration of clinically relevant doses of both AAV5ch and AAV1. The levels of transgene proteins achieved after delivery with AAV5ch and AAV1 illustrate the possibility of both serotypes for liver targeting. Furthermore, transgene DNA and RNA biodistribution patterns provided insight into the potential cause of decrease or loss of transgene protein expression over time in NHPs.
Collapse
|
31
|
Nienhuis AW, Nathwani AC, Davidoff AM. Gene Therapy for Hemophilia. Mol Ther 2017; 25:1163-1167. [PMID: 28411016 PMCID: PMC5417837 DOI: 10.1016/j.ymthe.2017.03.033] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/28/2017] [Accepted: 03/28/2017] [Indexed: 12/11/2022] Open
Abstract
The X-linked bleeding disorder hemophilia causes frequent and exaggerated bleeding that can be life-threatening if untreated. Conventional therapy requires frequent intravenous infusions of the missing coagulation protein (factor VIII [FVIII] for hemophilia A and factor IX [FIX] for hemophilia B). However, a lasting cure through gene therapy has long been sought. After a series of successes in small and large animal models, this goal has finally been achieved in humans by in vivo gene transfer to the liver using adeno-associated viral (AAV) vectors. In fact, multiple recent clinical trials have shown therapeutic, and in some cases curative, expression. At the same time, cellular immune responses against the virus have emerged as an obstacle in humans, potentially resulting in loss of expression. Transient immune suppression protocols have been developed to blunt these responses. Here, we provide an overview of the clinical development of AAV gene transfer for hemophilia, as well as an outlook on future directions.
Collapse
Affiliation(s)
- Arthur W Nienhuis
- Division of Experimental Hematology, Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Amit C Nathwani
- Department of Haematology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6BT, UK
| | - Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
32
|
Kotin RM, Snyder RO. Manufacturing Clinical Grade Recombinant Adeno-Associated Virus Using Invertebrate Cell Lines. Hum Gene Ther 2017; 28:350-360. [DOI: 10.1089/hum.2017.042] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Robert M. Kotin
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
| | | |
Collapse
|
33
|
Effective Depletion of Pre-existing Anti-AAV Antibodies Requires Broad Immune Targeting. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 4:159-168. [PMID: 28345001 PMCID: PMC5363314 DOI: 10.1016/j.omtm.2017.01.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/04/2017] [Indexed: 11/22/2022]
Abstract
Pre-existing antibodies (Abs) to AAV pose a critical challenge for the translation of gene therapies. No effective approach is available to overcome pre-existing Abs. Given the complexity of Ab production, overcoming pre-existing Abs will require broad immune targeting. We generated a mouse model of pre-existing AAV9 Abs to test multiple immunosuppressants, including bortezomib, rapamycin, and prednisolone, individually or in combination. We identified an effective approach combining rapamycin and prednisolone, reducing serum AAV9 Abs by 70%–80% at 4 weeks and 85%–93% at 8 weeks of treatment. The rapamycin plus prednisolone treatment resulted in significant decreases in the frequency of B cells, plasma cells, and IgG-secreting and AAV9-specific Ab-producing plasma cells in bone marrow. The rapamycin plus prednisolone treatment also significantly reduced frequencies of IgD−IgG+ class-switched/FAS+CL7+ germinal center B cells, and of activated CD4+ T cells expressing PD1 and GL7, in spleen. These data suggest that rapamycin plus prednisolone has selective inhibitory effects on both T helper type 2 support of B cell activation in spleen and on bone marrow plasma cell survival, leading to effective AAV9 Abs depletion. This promising immunomodulation approach is highly translatable, and it poses minimal risk in the context of therapeutic benefits promised by gene therapy for severe monogenetic diseases, with a single or possibly a few treatments over a lifetime.
Collapse
|
34
|
Transient CD4+ T Cell Depletion Results in Delayed Development of Functional Vaccine-Elicited Antibody Responses. J Virol 2016; 90:4278-4288. [PMID: 26865713 PMCID: PMC4836333 DOI: 10.1128/jvi.00039-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 02/05/2016] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED We have recently demonstrated that CD4(+)T cell help is required at the time of adenovirus (Ad) vector immunization for the development of functional CD8(+)T cell responses, but the temporal requirement for CD4(+)T cell help for the induction of antibody responses remains unclear. Here we demonstrate that induction of antibody responses in C57BL/6 mice can occur at a time displaced from the time of Ad vector immunization by depletion of CD4(+)T cells. Transient depletion of CD4(+)T cells at the time of immunization delays the development of antigen-specific antibody responses but does not permanently impair their development or induce tolerance against the transgene. Upon CD4(+)T cell recovery, transgene-specific serum IgG antibody titers develop and reach a concentration equivalent to that in undepleted control animals. These delayed antibody responses exhibit no functional defects with regard to isotype, functional avidity, expansion after boosting immunization, or the capacity to neutralize a simian immunodeficiency virus (SIV) Env-expressing pseudovirus. The development of this delayed transgene-specific antibody response is temporally linked to the expansion of de novo antigen-specific CD4(+)T cell responses, which develop after transient depletion of CD4(+)T cells. These data demonstrate that functional vaccine-elicited antibody responses can be induced even if CD4(+)T cell help is provided at a time markedly separated from the time of vaccination. IMPORTANCE CD4(+)T cells have a critical role in providing positive help signals to B cells, which promote robust antibody responses. The paradigm is that helper signals must be provided immediately upon antigen exposure, and their absence results in tolerance against the antigen. Here we demonstrate that, in contrast to the current model that the absence of CD4(+)T cell help at priming results in long-term antibody nonresponsiveness, antibody responses can be induced by adenovirus vector immunization or alum-adjuvanted protein immunization even if CD4(+)T cell help is not provided until >1 month after immunization. These data demonstrate that the time when CD4(+)T cell help signals must be provided is more dynamic and flexible than previously appreciated. These data suggest that augmentation of CD4(+)T cell helper function even after the time of vaccination can enhance vaccine-elicited antibody responses and thereby potentially enhance the immunogenicity of vaccines in immunocompromised individuals.
Collapse
|
35
|
Abstract
Adeno-associated viral vectors have been developed for the treatment of hemophilia A and B. Derivation of vector particles is achieved after multiplasmid transfection of cells that package the vector genome to yield vector particles. To date, three clinical trials have been performed for hemophilia B. The results of these trials are described. The trial that we conducted with our collaborators has yielded evidence of clinical efficacy for hemophilia B. A vector for treating hemophilia A has been developed and a clinical trial is planned.
Collapse
Affiliation(s)
- Arthur W Nienhuis
- 1 Division of Experimental Hematology, Department of Hematology, St. Jude Children's Research Hospital , Memphis, Tennessee
| | - Amit C Nathwani
- 2 Department of Haematology, University College London Cancer Institute , London, United Kingdom
| | - Andrew M Davidoff
- 3 Department of Surgery, St. Jude Children's Research Hospital , Memphis, Tennessee
| |
Collapse
|
36
|
Tse LV, Moller-Tank S, Asokan A. Strategies to circumvent humoral immunity to adeno-associated viral vectors. Expert Opin Biol Ther 2015; 15:845-55. [PMID: 25985812 DOI: 10.1517/14712598.2015.1035645] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Recent success in gene therapy of certain monogenic diseases in the clinic has infused enthusiasm into the continued development of recombinant adeno-associated viral (AAV) vectors as next-generation biologics. However, progress in clinical trials has also highlighted the challenges posed by the host humoral immune response to AAV vectors. Specifically, while pre-existing neutralizing antibodies (NAbs) limit the cohort of eligible patients, NAb generation following treatment prevents vector re-dosing. AREAS COVERED In this review, we discuss a spectrum of complementary strategies that can help circumvent the host humoral immune response to AAV. EXPERT OPINION Specifically, we present a dual perspective, that is, vector versus host, and highlight the clinical attributes, potential caveats and limitations as well as complementarity associated with the various approaches.
Collapse
Affiliation(s)
- Longping V Tse
- University of North Carolina, Gene Therapy Center , CB#7352, Thurston Building, Chapel Hill, NC 27599 , USA
| | | | | |
Collapse
|
37
|
Hareendran S, Ramakrishna B, Jayandharan GR. Synergistic inhibition of PARP-1 and NF-κB signaling downregulates immune response against recombinant AAV2 vectors during hepatic gene therapy. Eur J Immunol 2015; 46:154-66. [PMID: 26443873 DOI: 10.1002/eji.201545867] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 09/04/2015] [Accepted: 09/30/2015] [Indexed: 02/02/2023]
Abstract
Host immune response remains a key obstacle to widespread application of adeno-associated virus (AAV) based gene therapy. Thus, targeted inhibition of the signaling pathways that trigger such immune responses will be beneficial. Previous studies have reported that DNA damage response proteins such as poly(ADP-ribose) polymerase-1 (PARP-1) negatively affect the integration of AAV in the host genome. However, the role of PARP-1 in regulating AAV transduction and the immune response against these vectors has not been elucidated. In this study, we demonstrate that repression of PARP-1 improves the transduction of single-stranded AAV vectors both in vitro (∼174%) and in vivo (two- to 3.4-fold). Inhibition of PARP-1, also significantly downregulated the expression of several proinflammatory and cytokine markers such as TLRs, ILs, NF-κB subunit proteins associated with the host innate response against self-complementary AAV2 vectors. The suppression of the inflammatory response targeted against these vectors was more effective upon combined inhibition of PARP-1 and NF-κB signaling. This strategy also effectively attenuated the AAV capsid-specific cytotoxic T-cell response, with minimal effect on vector transduction, as demonstrated in normal C57BL/6 and hemophilia B mice. These data suggest that targeting specific host cellular proteins could be useful to attenuate the immune barriers to AAV-mediated gene therapy.
Collapse
Affiliation(s)
- Sangeetha Hareendran
- Centre for Stem Cell Research, Christian Medical College, Vellore, Tamil Nadu, India
| | - Banumathi Ramakrishna
- Department of General Pathology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Giridhara R Jayandharan
- Centre for Stem Cell Research, Christian Medical College, Vellore, Tamil Nadu, India.,Department of Hematology, Christian Medical College, Vellore, Tamil Nadu, India.,Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh, India
| |
Collapse
|
38
|
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is a relatively common inherited disorder caused by defective expression of the protein dystrophin. The most direct approach to treating this disease would be to restore dystrophin production in muscle. Recent progress has greatly increased the prospects for successful gene therapy of DMD, and here we summarize the most promising developments. AREAS COVERED Gene transfer using vectors derived from adeno-associated virus (AAV) has emerged as a promising method to restore dystrophin production in muscles bodywide, and represents a treatment option applicable to all DMD patients. Using information gleaned from PubMed searches of the literature, attendance at scientific conferences and results from our own lab, we provide an overview of the potential for gene therapy of DMD using AAV vectors including a summary of promising developments and issues that need to be resolved prior to large-scale therapeutic implementation. EXPERT OPINION Of the many approaches being pursued to treat DMD and BMD, gene therapy based on AAV-mediated delivery of microdystrophin is the most direct and promising method to treat the cause of the disorder. The major challenges to this approach are ensuring that microdystrophin can be delivered safely and efficiently without eliciting an immune response.
Collapse
Affiliation(s)
- Julian Ramos
- University of Washington, Wellstone Muscular Dystrophy Research Center, Department of Neurology, Seattle, WA, 98195-7720, USA
| | - Jeffrey S Chamberlain
- University of Washington, Wellstone Muscular Dystrophy Research Center, Department of Neurology, Seattle, WA, 98195-7720, USA
| |
Collapse
|
39
|
Han SO, Li S, Brooks ED, Masat E, Leborgne C, Banugaria S, Bird A, Mingozzi F, Waldmann H, Koeberl D. Enhanced efficacy from gene therapy in Pompe disease using coreceptor blockade. Hum Gene Ther 2015; 26:26-35. [PMID: 25382056 DOI: 10.1089/hum.2014.115] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Enzyme replacement therapy (ERT) is the standard-of-care treatment of Pompe disease, a lysosomal storage disorder caused by deficiency of acid α-glucosidase (GAA). One limitation of ERT with recombinant human (rh) GAA is antibody formation against GAA. Similarly, in adeno-associated virus (AAV) vector-mediated gene transfer for Pompe disease, development of antibodies against the GAA transgene product and the AAV vector prevents therapeutic efficacy and vector readministration, respectively. Here a nondepleting anti-CD4 monoclonal antibody (mAb) was administrated intravenously prior to administration of an AAV2/9 vector encoding GAA to suppress anti-GAA responses, leading to a substantial reduction of anti-GAA immunoglobulins, including IgG1, IgG2a, IgG2b, IgG2c, and IgG3. Transduction efficiency in liver with a subsequent AAV2/8 vector was massively improved by the administration of anti-CD4 mAb with the initial AAV2/9 vector, indicating a spread of benefit derived from control of the immune response to the first AAV2/9 vector. Anti-CD4 mAb along with AAV2/9-CBhGAApA significantly increased GAA activity in heart and skeletal muscles along with a significant reduction of glycogen accumulation. Taken together, these data demonstrated that the addition of nondepleting anti-CD4 mAb with gene therapy controls humoral immune responses to both vector and transgene, resulting in clear therapeutic benefit in mice with Pompe disease.
Collapse
Affiliation(s)
- Sang-oh Han
- 1 Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center , Durham, NC 27710
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang Z, Zourelias L, Wu C, Edwards PC, Trombetta M, Passineau MJ. Ultrasound-assisted nonviral gene transfer of AQP1 to the irradiated minipig parotid gland restores fluid secretion. Gene Ther 2015; 22:739-49. [PMID: 25871828 PMCID: PMC4560616 DOI: 10.1038/gt.2015.36] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 03/17/2015] [Accepted: 03/31/2015] [Indexed: 12/16/2022]
Abstract
Rationale Xerostomia is a common side effect of ionizing radiation used to treat head and neck cancer. A groundbreaking Phase I human clinical trial utilizing Adenoviral gene transfer of Aquaporin-1 (AQP1) to a single salivary gland of individuals suffering from radiation-induced xerostomia has recently been reported. Unfortunately, the limitations of the Adenoviral vector system utilized in this pioneering trial preclude its advancement to a Phase II trial and we have thus undertaken to evaluate the therapeutic potential of ultrasound-assisted non-viral gene transfer (UAGT) as an alternative means of delivering AQP1 gene therapy to the salivary gland by comparing head-to-head with the canonical Adenoviral vector in a swine model. Findings Swine irradiated unilaterally with a 10Gy electron beam targeted at the parotid gland suffered from significant, sustained hyposalivation that was bilateral, despite irradiation being confined to the targeted gland. Unilateral AQP1 gene therapy with UAGT resulted in bilateral restoration of stimulated salivary flow at 48 hours and one week post-treatment (1.62+/−0.48ml, 1.87+/−0.45ml) to pre-injury levels (1.34+/−0.14ml) in a manner comparable to Adenoviral delivery (2.32+/−0.6ml, 1.33+/−0.97ml). Conclusions UAGT can replace the Adenoviral vector as a means of delivering AQP1 gene therapy in the irradiated swine model and is a candidate for advancement to a Phase I human clinical trial.
Collapse
Affiliation(s)
- Z Wang
- Gene Therapy Program, Division of Cardiovascular Medicine, Department of Medicine, Allegheny Health Network, Pittsburgh, PA, USA
| | - L Zourelias
- Gene Therapy Program, Division of Cardiovascular Medicine, Department of Medicine, Allegheny Health Network, Pittsburgh, PA, USA
| | - C Wu
- Gene Therapy Program, Division of Cardiovascular Medicine, Department of Medicine, Allegheny Health Network, Pittsburgh, PA, USA
| | - P C Edwards
- Department of Oral Pathology, Medicine and Radiology, University of Indiana School of Dentistry, Indianapolis, IN, USA
| | - M Trombetta
- Division of Radiation Oncology, Department of Oncology, Allegheny Health Network, Pittsburgh, PA, USA
| | - M J Passineau
- Gene Therapy Program, Division of Cardiovascular Medicine, Department of Medicine, Allegheny Health Network, Pittsburgh, PA, USA
| |
Collapse
|
41
|
Rogers GL, Suzuki M, Zolotukhin I, Markusic DM, Morel LM, Lee B, Ertl HCJ, Herzog RW. Unique Roles of TLR9- and MyD88-Dependent and -Independent Pathways in Adaptive Immune Responses to AAV-Mediated Gene Transfer. J Innate Immun 2015; 7:302-14. [PMID: 25612611 DOI: 10.1159/000369273] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 10/22/2014] [Indexed: 12/12/2022] Open
Abstract
The immune system represents a significant barrier to successful gene therapy with adeno-associated viral (AAV) vectors. In particular, adaptive immune responses to the viral capsid or the transgene product are of concern. The sensing of AAV by toll-like receptors (TLRs) TLR2 and TLR9 has been suggested to play a role in innate immunity to the virus and may also shape subsequent adaptive immune responses. Here, we investigated the functions of TLR2, TLR9 and the downstream signaling adaptor MyD88 in antibody and CD8+ T-cell responses. Antibody formation against the transgene product occurred largely independently of TLR signaling following gene transfer with AAV1 or AAV2 vectors, whereas loss of signaling through the TLR9-MyD88 pathway substantially reduced CD8+ T-cell responses. In contrast, MyD88 (but neither of the TLRs) regulated antibody responses to capsid. B cell-intrinsic MyD88 was required for the formation of anti-capsid IgG2c independently of vector serotype or route of administration. However, MyD88(-/-) mice instead produced anti-capsid IgG1 that emerged with delayed kinetics but nonetheless completely prevented in vivo readministration. We conclude that there are distinct roles for TLR9 and MyD88 in promoting adaptive immune responses to AAV-mediated gene transfer and that there are redundant MyD88-dependent and MyD88-independent mechanisms that stimulate neutralizing antibody formation against AAV.
Collapse
Affiliation(s)
- Geoffrey L Rogers
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, Fla., USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Boisgérault F, Mingozzi F. The Skeletal Muscle Environment and Its Role in Immunity and Tolerance to AAV Vector-Mediated Gene Transfer. Curr Gene Ther 2015; 15:381-94. [PMID: 26122097 PMCID: PMC4515578 DOI: 10.2174/1566523215666150630121750] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 06/15/2015] [Accepted: 06/19/2015] [Indexed: 02/08/2023]
Abstract
Since the early days of gene therapy, muscle has been one the most studied tissue targets for the correction of enzyme deficiencies and myopathies. Several preclinical and clinical studies have been conducted using adeno-associated virus (AAV) vectors. Exciting progress has been made in the gene delivery technologies, from the identification of novel AAV serotypes to the development of novel vector delivery techniques. In parallel, significant knowledge has been generated on the host immune system and its interaction with both the vector and the transgene at the muscle level. In particular, the role of underlying muscle inflammation, characteristic of several diseases affecting the muscle, has been defined in terms of its potential detrimental impact on gene transfer with AAV vectors. At the same time, feedback immunomodulatory mechanisms peculiar of skeletal muscle involving resident regulatory T cells have been identified, which seem to play an important role in maintaining, at least to some extent, muscle homeostasis during inflammation and regenerative processes. Devising strategies to tip this balance towards unresponsiveness may represent an avenue to improve the safety and efficacy of muscle gene transfer with AAV vectors.
Collapse
Affiliation(s)
| | - Federico Mingozzi
- Genethon, Evry, France
- University Pierre and Marie Curie, Paris, France
| |
Collapse
|
43
|
Abstract
Hemophilia is an X-linked inherited bleeding disorder consisting of two classifications, hemophilia A and hemophilia B, depending on the underlying mutation. Although the disease is currently treatable with intravenous delivery of replacement recombinant clotting factor, this approach represents a significant cost both monetarily and in terms of quality of life. Gene therapy is an attractive alternative approach to the treatment of hemophilia that would ideally provide life-long correction of clotting activity with a single injection. In this review, we will discuss the multitude of approaches that have been explored for the treatment of both hemophilia A and B, including both in vivo and ex vivo approaches with viral and nonviral delivery vectors.
Collapse
Affiliation(s)
- Geoffrey L Rogers
- University of Florida, Department of Pediatrics, Division of Cellular and Molecular Therapy, Gainesville, FL 32610
| | - Roland W Herzog
- University of Florida, Department of Pediatrics, Division of Cellular and Molecular Therapy, Gainesville, FL 32610
| |
Collapse
|
44
|
Rocca CJ, Ur SN, Harrison F, Cherqui S. rAAV9 combined with renal vein injection is optimal for kidney-targeted gene delivery: conclusion of a comparative study. Gene Ther 2014; 21:618-28. [PMID: 24784447 PMCID: PMC4047163 DOI: 10.1038/gt.2014.35] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 03/03/2014] [Accepted: 03/14/2014] [Indexed: 12/19/2022]
Abstract
Effective gene therapy strategies for the treatment of kidney disorders remain elusive. We report an optimized kidney-targeted gene delivery strategy using recombinant adeno-associated virus (rAAV) administered via retrograde renal vein injection in mice. Renal vein injection of rAAV consistently resulted in superior kidney transduction compared with tail vein injection using as little as half the tail vein dose. We compared rAAV5, 6, 8 and 9, containing either green fluorescent protein (GFP) or luciferase reporter genes driven by the Cytomegalovirus promoter. We demonstrated that although rAAV6 and 8 injected via renal vein transduced the kidney, transgene expression was mainly restricted to the medulla. Transgene expression was systematically low after rAAV5 injection, attributed to T-cell immune response, which could be overcome by transient immunosuppression. However, rAAV9 was the only serotype that permitted high-transduction efficiency of both the cortex and medulla. Moreover, both the glomeruli and tubules were targeted, with a higher efficiency within the glomeruli. To improve the specificity of kidney-targeted gene delivery with rAAV9, we used the parathyroid hormone receptor 'kidney-specific' promoter. We obtained a more efficient transgene expression within the kidney, and a significant reduction in other tissues. Our work represents the first comprehensive and clinically relevant study for kidney gene delivery.
Collapse
Affiliation(s)
- Céline J. Rocca
- Department of Pediatrics, Division of Genetics, University of California, San Diego, 9500 Gilman drive, MC 0734, La Jolla, California 92093-0734, USA
| | - Sarah N. Ur
- Department of Pediatrics, Division of Genetics, University of California, San Diego, 9500 Gilman drive, MC 0734, La Jolla, California 92093-0734, USA
| | - Frank Harrison
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Stephanie Cherqui
- Department of Pediatrics, Division of Genetics, University of California, San Diego, 9500 Gilman drive, MC 0734, La Jolla, California 92093-0734, USA
| |
Collapse
|
45
|
Influence of immune responses in gene/stem cell therapies for muscular dystrophies. BIOMED RESEARCH INTERNATIONAL 2014; 2014:818107. [PMID: 24959590 PMCID: PMC4052166 DOI: 10.1155/2014/818107] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/07/2014] [Accepted: 04/30/2014] [Indexed: 02/06/2023]
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of diseases, caused by mutations in different components of sarcolemma, extracellular matrix, or enzymes. Inflammation and innate or adaptive immune response activation are prominent features of MDs. Various therapies under development are directed toward rescuing the dystrophic muscle damage using gene transfer or cell therapy. Here we discussed current knowledge about involvement of immune system responses to experimental therapies in MDs.
Collapse
|
46
|
Basner-Tschakarjan E, Bijjiga E, Martino AT. Pre-Clinical Assessment of Immune Responses to Adeno-Associated Virus (AAV) Vectors. Front Immunol 2014; 5:28. [PMID: 24570676 PMCID: PMC3916775 DOI: 10.3389/fimmu.2014.00028] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 01/17/2014] [Indexed: 12/21/2022] Open
Abstract
Transitioning to human trials from pre-clinical models resulted in the emergence of inhibitory AAV vector immune responses which has become a hurdle for sustained correction. Early animal studies did not predict the full range of host immunity to the AAV vector in human studies. While pre-existing antibody titers against AAV vectors has been a lingering concern, cytotoxic T-cell (CTL) responses against the input capsid can prevent long-term therapy in humans. These discoveries spawned more thorough profiling of immune response to rAAV in pre-clinical models, which have assessed both innate and adaptive immunity and explored methods for bypassing these responses. Many efforts toward measuring innate immunity have utilized Toll-like receptor deficient models and have focused on differential responses to viral capsid and genome. From adaptive studies, it is clear that humoral responses are relevant for initial vector transduction efficiency while cellular responses impact long-term outcomes of gene transfer. Measuring humoral responses to AAV vectors has utilized in vitro neutralizing antibody assays and transfer of seropositive serum to immunodeficient mice. Overcoming antibodies using CD20 inhibitors, plasmapheresis, altering route of delivery and using different capsids have been explored. CTL responses were measured using in vitro and in vivo models. In in vitro assays expansion of antigen-specific T-cells as well as cytotoxicity toward AAV transduced cells can be shown. Many groups have successfully mimicked antigen-specific T-cell proliferation, but actual transgene level reduction and parameters of cytotoxicity toward transduced target cells have only been shown in one model. The model utilized adoptive transfer of capsid-specific in vitro expanded T-cells isolated from immunized mice with LPS as an adjuvant. Finally, the development of immune tolerance to AAV vectors by enriching regulatory T-cells as well as modulating the response pharmacologically has also been explored.
Collapse
Affiliation(s)
| | - Enoch Bijjiga
- Department of Pharmaceutical Sciences, St. John's University , Queens, NY , USA
| | - Ashley T Martino
- Department of Pharmaceutical Sciences, St. John's University , Queens, NY , USA
| |
Collapse
|
47
|
Hareendran S, Balakrishnan B, Sen D, Kumar S, Srivastava A, Jayandharan GR. Adeno-associated virus (AAV) vectors in gene therapy: immune challenges and strategies to circumvent them. Rev Med Virol 2013; 23:399-413. [DOI: 10.1002/rmv.1762] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 08/08/2013] [Accepted: 08/09/2013] [Indexed: 12/12/2022]
Affiliation(s)
- Sangeetha Hareendran
- Centre for Stem Cell Research; Christian Medical College; Vellore Tamil Nadu India
| | - Balaji Balakrishnan
- Department of Hematology; Christian Medical College; Vellore Tamil Nadu India
| | - Dwaipayan Sen
- Department of Hematology; Christian Medical College; Vellore Tamil Nadu India
| | - Sanjay Kumar
- Centre for Stem Cell Research; Christian Medical College; Vellore Tamil Nadu India
| | - Alok Srivastava
- Centre for Stem Cell Research; Christian Medical College; Vellore Tamil Nadu India
- Department of Hematology; Christian Medical College; Vellore Tamil Nadu India
| | - Giridhara R. Jayandharan
- Centre for Stem Cell Research; Christian Medical College; Vellore Tamil Nadu India
- Department of Hematology; Christian Medical College; Vellore Tamil Nadu India
| |
Collapse
|
48
|
Wang F, Cui X, Wang M, Xiao W, Xu R. A reliable and feasible qPCR strategy for titrating AAV vectors. Med Sci Monit Basic Res 2013; 19:187-93. [PMID: 23828206 PMCID: PMC3706409 DOI: 10.12659/msmbr.883968] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Previous studies have revealed that traditional real-time quantitative PCR (qPCR) underestimates adeno-associated virus (AAV) titer. Because the inverted terminal repeat (ITR) exists in all AAV vectors, the only remaining element from the wild genome could form special configurations to interfere with qPCR titration. To solve this problem, a modified and universal qPCR method was tested and established. MATERIAL AND METHODS In this work, there was a great variation in titration of ssAAV2-EGFP (Enhanced Green Fluorescence Protein) and scAAV2-EGFP genome by traditional qPCR. For ssAAV2-EGFP, the highest titer was found by using the targeting EGFP primers and the lowest titer was measured by those targeting bovine growth hormone polyA element (pBGH) primers. RESULTS Experimental data were reverse for ssAAV2-EGFP and scAAV2-EGFP. Here we report an improved and universal SmaI qPCR method, based on cleaving all ITRs in AAV2 genome by SmaI with several advantages: (1) impact of all ITRs in ssAAV2 and scAAV2 was dismissed; (2) titers increased remarkably, up to 7-fold, especially for scAAV2; (3) the variation of titers was reduced when different primers were applied. A similar phenomenon was also observed in other ssAAV2 and scAAV2 products when the range of titration was at 3×107 to 7×109 V.G/µl in this study. CONCLUSIONS This modified qPCR strategy can increase rAAV' titer and reduce titration variance, possibly become a universal method for titrating AAV vectors.
Collapse
Affiliation(s)
- Feng Wang
- School of Biomedical Science and Institute of Molecular Medicine, Huaqiao University and Engineering Research Center of Molecular Medicine, Ministry of Education, Quanzhou, Fujian, China
| | | | | | | | | |
Collapse
|
49
|
Pre-immunization with an intramuscular injection of AAV9-human erythropoietin vectors reduces the vector-mediated transduction following re-administration in rat brain. PLoS One 2013; 8:e63876. [PMID: 23667683 PMCID: PMC3648480 DOI: 10.1371/journal.pone.0063876] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 04/08/2013] [Indexed: 12/22/2022] Open
Abstract
We have recently demonstrated that adeno-associated virus serotype 9 (AAV9)-mediated human erythropoietin (hEPO) gene delivery into the brain protects dopaminergic (DA) neurons in the substantia nigra in a rat model of Parkinson's disease. In the present study, we examined whether pre-exposure to AAV9-hEPO vectors with an intramuscular or intrastriatal injection would reduce AAV9-mediated hEPO transduction in rat brain. We first characterized transgene expression and immune responses against AAV9-hEPO vectors in rat striatum at 4 days, 3 weeks and 6 months, and with doses ranging from 1011 to 1013 viral genomes. To sensitize immune system, rats received an injection of AAV9-hEPO into either the muscle or the left striatum, and then sequentially an injection of AAV9-hEPO into the right striatum 3 weeks later. We observed that transgene expression exhibited in a time course and dose dependent manner, and inflammatory and immune responses displayed in a time course manner. Intramuscular, but not intrastriatal injections of AAV9-hEPO resulted in reduced levels of hEPO transduction and increased levels of the major histocompatibility complex (MHC) class I and class II antigen expression in the striatum following AAV9-hEPO re-administration. There were infiltration of the cluster of differentiation 4 (CD4)-and CD8-lymphacytes, and accumulation of activated microglial cells and astrocytes in the virally injected striatum. In addition, the sera from the rats with intramuscular injections of AAV9-hEPO contained greater levels of antibodies against both AAV9 capsid protein and hEPO protein than the other treatment groups. hEPO gene expression was negatively correlated with the levels of circulating antibodies against AAV9 capsid protein. Intramuscular and intrastriatal re-administration of AAV9-hEPO led to increased numbers of red blood cells in peripheral blood. Our results suggest that pre-immunization with an intramuscular injection can lead to the reduction of transgene expression in the striatal re-administration.
Collapse
|
50
|
Markusic DM, Herzog RW. Liver-Directed Adeno-Associated Viral Gene Therapy for Hemophilia. ACTA ACUST UNITED AC 2013; 1:1-9. [PMID: 23565343 DOI: 10.4172/2157-7412.s1-009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hemophilia A and B are monogenic bleeding disorders resulting from loss of functional coagulation factors VIII or IX, respectively. Prophylactic treatment requires frequent intravenous injections of exogenous factor VIII (F.VIII) or factor IX (F.IX), due to the short half-life of both factors. Hemophilia patients are at risk of developing neutralizing antibodies to F.VIII (~25-30%) or F.IX (~2-4%), which require the use of expensive bypass agents and immune tolerance induction protocols. Viral vector mediated liver gene transfer of F.VIII or F.IX offers an alternative treatment for hemophilia with easily defined clinical endpoints and no need for strict regulation of coagulation factor expression, as both proteins circulate as inactive zymogens. Adeno-associated viral (AAV) vectors are derived from a non-pathogenic human virus that efficiently transduce non-dividing cells, such as hepatocytes, and provide stable transgene expression. In vivo liver gene transfer of AAV-F.VIII and -F.IX vectors has restored hemostasis in murine and canine hemophilia models long-term, and has also been shown to induce immune tolerance. Consequently, two Phase I/II clinical trials have been conducted, based on hepatic AAV-FIX gene transfer to patients with severe hemophilia B. The first trial, utilizing serotype 2, demonstrated transient correction, which was limited by a cellular immune response against the viral capsid. However, sustained therapeutic expression has been achieved in a second trial, using AAV8 for expression of a codon-optimized F.IX transgene. Translation of F.VIII gene transfer studies into the clinic may require additional optimization of gene transfer and vector to effectively express the larger cDNA of F.VIII.
Collapse
Affiliation(s)
- David M Markusic
- Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | | |
Collapse
|