1
|
Weber JA, Lang JF, Carrell EM, Alameh MG, Davidson BL. Temporal restriction of Cas9 expression improves CRISPR-mediated deletion efficacy and fidelity. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102172. [PMID: 38978694 PMCID: PMC11229411 DOI: 10.1016/j.omtn.2024.102172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 03/08/2024] [Indexed: 07/10/2024]
Abstract
Clinical application of CRISPR-Cas9 technology for large deletions of somatic mutations is inefficient, and methods to improve utility suffer from our inability to rapidly assess mono- vs. biallelic deletions. Here we establish a model system for investigating allelic heterogeneity at the single-cell level and identify indel scarring from non-simultaneous nuclease activity at gRNA cut sites as a major barrier to CRISPR-del efficacy both in vitro and in vivo. We show that non-simultaneous nuclease activity is partially prevented via restriction of CRISPR-Cas9 expression via inducible adeno-associated viruses (AAVs) or lipid nanoparticles (LNPs). Inducible AAV-based expression of CRISPR-del machinery significantly improved mono- and biallelic deletion frequency in vivo, supporting the use of the Xon cassette over traditional constitutively expressing AAV approaches. These data depicting improvements to deletions and insight into allelic heterogeneity after CRISPR-del will inform therapeutic approaches for phenotypes that require either large mono- or biallelic deletions, such as autosomal recessive diseases or where mutant allele-specific gRNAs are not readily available, or in situations where the targeted sequence for excision is located multiple times in a genome.
Collapse
Affiliation(s)
- Jesse A Weber
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Cell and Molecular Biology Graduate Group, Biomedical Graduate Studies, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan F Lang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Cell and Molecular Biology Graduate Group, Biomedical Graduate Studies, University of Pennsylvania, Philadelphia, PA, USA
| | - Ellie M Carrell
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mohamad-Gabriel Alameh
- Penn Institute for RNA Innovation, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Beverly L Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
2
|
Wang Y, Lifshitz L, Silverstein NJ, Mintzer E, Luk K, StLouis P, Brehm MA, Wolfe SA, Deeks SG, Luban J. Transcriptional and chromatin profiling of human blood innate lymphoid cell subsets sheds light on HIV-1 pathogenesis. EMBO J 2023; 42:e114153. [PMID: 37382276 PMCID: PMC10425848 DOI: 10.15252/embj.2023114153] [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: 03/30/2023] [Revised: 05/30/2023] [Accepted: 06/12/2023] [Indexed: 06/30/2023] Open
Abstract
Innate lymphoid cells (ILCs) are a diverse population of cells that include NK cells and contribute to tissue homeostasis and repair, inflammation, and provide protection from infection. The interplay between human blood ILCs, as well as their responses to HIV-1 infection, remains poorly understood. This study used transcriptional and chromatin profiling to explore these questions. Transcriptional profiling and flow cytometry analysis support that there are four main ILC subsets found in human blood. Unlike in mice, human NK cells expressed the tissue repair protein amphiregulin (AREG). AREG production was induced by TCF7/WNT, IL-2, and IL-15, and inhibited by TGFB1, a cytokine increased in people living with HIV-1. In HIV-1 infection, the percentage of AREG+ NK cells correlated positively with the numbers of ILCs and CD4+ T cells but negatively with the concentration of inflammatory cytokine IL-6. NK-cell knockout of the TGFB1-stimulated WNT antagonist RUNX3 increased AREG production. Antiviral gene expression was increased in all ILC subsets from HIV-1 viremic people, and anti-inflammatory gene MYDGF was increased in an NK-cell subset from HIV-1-infected people whose viral load was undetectable in the absence of antiretroviral therapy. The percentage of defective NK cells in people living with HIV-1 correlated inversely with ILC percentage and CD4+ T-cell counts. CD4+ T cells and their production of IL-2 prevented the loss of NK-cell function by activating mTOR. These studies clarify how ILC subsets are interrelated and provide insight into how HIV-1 infection disrupts NK cells, including an uncharacterized homeostatic function in NK cells.
Collapse
Affiliation(s)
- Yetao Wang
- Hospital for Skin Diseases (Institute of Dermatology)Chinese Academy of Medical Sciences and Peking Union Medical CollegeNanjingChina
- Key Laboratory of Basic and Translational Research on Immune‐Mediated Skin DiseasesChinese Academy of Medical SciencesNanjingChina
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of DermatologyChinese Academy of Medical Sciences and Peking Union Medical CollegeNanjingChina
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Lawrence Lifshitz
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Noah J Silverstein
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Esther Mintzer
- Department of Molecular, Cell and Cancer BiologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Kevin Luk
- Department of Molecular, Cell and Cancer BiologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Pamela StLouis
- Diabetes Center of ExcellenceUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Michael A Brehm
- Diabetes Center of ExcellenceUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Scot A Wolfe
- Department of Molecular, Cell and Cancer BiologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Steven G Deeks
- Department of MedicineUniversity of CaliforniaSan FranciscoCAUSA
| | - Jeremy Luban
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
- Department of Biochemistry and Molecular BiotechnologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
- Broad Institute of MIT and HarvardCambridgeMAUSA
- Ragon Institute of MGH, MIT, and HarvardCambridgeMAUSA
- Massachusetts Consortium on Pathogen ReadinessBostonMAUSA
| |
Collapse
|
3
|
Rapti K, Grimm D. Adeno-Associated Viruses (AAV) and Host Immunity - A Race Between the Hare and the Hedgehog. Front Immunol 2021; 12:753467. [PMID: 34777364 PMCID: PMC8586419 DOI: 10.3389/fimmu.2021.753467] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
Adeno-associated viruses (AAV) have emerged as the lead vector in clinical trials and form the basis for several approved gene therapies for human diseases, mainly owing to their ability to sustain robust and long-term in vivo transgene expression, their amenability to genetic engineering of cargo and capsid, as well as their moderate toxicity and immunogenicity. Still, recent reports of fatalities in a clinical trial for a neuromuscular disease, although linked to an exceptionally high vector dose, have raised new caution about the safety of recombinant AAVs. Moreover, concerns linger about the presence of pre-existing anti-AAV antibodies in the human population, which precludes a significant percentage of patients from receiving, and benefitting from, AAV gene therapies. These concerns are exacerbated by observations of cellular immune responses and other adverse events, including detrimental off-target transgene expression in dorsal root ganglia. Here, we provide an update on our knowledge of the immunological and molecular race between AAV (the “hedgehog”) and its human host (the “hare”), together with a compendium of state-of-the-art technologies which provide an advantage to AAV and which, thus, promise safer and more broadly applicable AAV gene therapies in the future.
Collapse
Affiliation(s)
- Kleopatra Rapti
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Heidelberg, Germany.,BioQuant Center, BQ0030, University of Heidelberg, Heidelberg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Heidelberg, Germany.,BioQuant Center, BQ0030, University of Heidelberg, Heidelberg, Germany.,German Center for Infection Research Deutsches Zentrum für Infektionsforschung (DZIF) and German Center for Cardiovascular Research Deutsches Zentrum für Herz-Kreislauf-Erkrankungen (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| |
Collapse
|
4
|
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: 75] [Impact Index Per Article: 25.0] [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
|
5
|
Chowdhury EA, Meno-Tetang G, Chang HY, Wu S, Huang HW, Jamier T, Chandran J, Shah DK. Current progress and limitations of AAV mediated delivery of protein therapeutic genes and the importance of developing quantitative pharmacokinetic/pharmacodynamic (PK/PD) models. Adv Drug Deliv Rev 2021; 170:214-237. [PMID: 33486008 DOI: 10.1016/j.addr.2021.01.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/17/2022]
Abstract
While protein therapeutics are one of the most successful class of drug molecules, they are expensive and not suited for treating chronic disorders that require long-term dosing. Adeno-associated virus (AAV) mediated in vivo gene therapy represents a viable alternative, which can deliver the genes of protein therapeutics to produce long-term expression of proteins in target tissues. Ongoing clinical trials and recent regulatory approvals demonstrate great interest in these therapeutics, however, there is a lack of understanding regarding their cellular disposition, whole-body disposition, dose-exposure relationship, exposure-response relationship, and how product quality and immunogenicity affects these important properties. In addition, there is a lack of quantitative studies to support the development of pharmacokinetic-pharmacodynamic models, which can support the discovery, development, and clinical translation of this delivery system. In this review, we have provided a state-of-the-art overview of current progress and limitations related to AAV mediated delivery of protein therapeutic genes, along with our perspective on the steps that need to be taken to improve clinical translation of this therapeutic modality.
Collapse
|
6
|
Engineering adeno-associated virus vectors for gene therapy. Nat Rev Genet 2020; 21:255-272. [DOI: 10.1038/s41576-019-0205-4] [Citation(s) in RCA: 342] [Impact Index Per Article: 85.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2019] [Indexed: 02/06/2023]
|
7
|
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: 50] [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
|
8
|
Nidetz NF, McGee MC, Tse LV, Li C, Cong L, Li Y, Huang W. Adeno-associated viral vector-mediated immune responses: Understanding barriers to gene delivery. Pharmacol Ther 2019; 207:107453. [PMID: 31836454 DOI: 10.1016/j.pharmthera.2019.107453] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 11/26/2019] [Indexed: 02/06/2023]
Abstract
Adeno-associated viral (AAV) vectors have emerged as the leading gene delivery platform for gene therapy and vaccination. Three AAV-based gene therapy drugs, Glybera, LUXTURNA, and ZOLGENSMA were approved between 2012 and 2019 by the European Medicines Agency and the United States Food and Drug Administration as treatments for genetic diseases hereditary lipoprotein lipase deficiency (LPLD), inherited retinal disease (IRD), and spinal muscular atrophy (SMA), respectively. Despite these therapeutic successes, clinical trials have demonstrated that host anti-viral immune responses can prevent the long-term gene expression of AAV vector-encoded genes. Therefore, it is critical that we understand the complex relationship between AAV vectors and the host immune response. This knowledge could allow for the rational design of optimized gene transfer vectors capable of either subverting host immune responses in the context of gene therapy applications, or stimulating desirable immune responses that generate protective immunity in vaccine applications to AAV vector-encoded antigens. This review provides an overview of our current understanding of the AAV-induced immune response and discusses potential strategies by which these responses can be manipulated to improve AAV vector-mediated gene transfer.
Collapse
Affiliation(s)
- Natalie F Nidetz
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Michael C McGee
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Longping V Tse
- Department of Epidemiology, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Le Cong
- Department of Pathology and Department of Genetics, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Yunxing Li
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Weishan Huang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
9
|
Chai Z, Zhang X, Dobbins AL, Rigsbee KM, Wang B, Samulski RJ, Li C. Optimization of Dexamethasone Administration for Maintaining Global Transduction Efficacy of Adeno-Associated Virus Serotype 9. Hum Gene Ther 2019; 30:829-840. [PMID: 30700148 DOI: 10.1089/hum.2018.233] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoids have been commonly used in clinic for their anti-inflammatory and immunosuppressive effects, and it has been proposed that they be used to prevent liver toxicity when systemic administration of adeno-associated virus (AAV) vectors is needed in patients with central nervous system diseases and muscular disorders. Glucocorticoids also enable modulation of vascular permeability. First, this study investigated the impact of dexamethasone on AAV vascular permeability after systemic injection. When a low dose of AAV9 was injected into mice treated with dexamethasone, global transduction and vector biodistribution were not significantly different in most tissues, other than the liver and the heart, when compared to control mice. When AAV9 vectors were used at a high dose, both the transgene expression and the AAV vector genome copy number were significantly decreased in the majority of murine tissues. However, no effect on global transduction was observed when dexamethasone was administered 2 h after AAV vector injection. The study on the kinetics of AAV virus clearance demonstrated that dexamethasone slowed down the clearance of AAV9 in the blood after systemic application. The mechanism study showed that dexamethasone inhibited the enhancement of AAV9 vascular permeability mediated by serum proteins. The findings indicate that dexamethasone is able to inhibit the vascular permeability of AAV and compromise the therapeutic effect after systemic administration of AAV vector. In conclusion, this study provides valuable information for the design of future clinical studies when glucocorticoids are needed to be compatible with the systemic administration of AAV vectors in patients with central nervous system and muscular diseases.
Collapse
Affiliation(s)
- Zheng Chai
- 1Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Xintao Zhang
- 1Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Amanda Lee Dobbins
- 1Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kelly Michelle Rigsbee
- 1Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Bing Wang
- 2Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Richard Jude Samulski
- 1Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,3Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Chengwen Li
- 1Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,4Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,5Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
10
|
Brehm MA, Kenney LL, Wiles MV, Low BE, Tisch RM, Burzenski L, Mueller C, Greiner DL, Shultz LD. Lack of acute xenogeneic graft- versus-host disease, but retention of T-cell function following engraftment of human peripheral blood mononuclear cells in NSG mice deficient in MHC class I and II expression. FASEB J 2019; 33:3137-3151. [PMID: 30383447 PMCID: PMC6404556 DOI: 10.1096/fj.201800636r] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 10/01/2018] [Indexed: 12/19/2022]
Abstract
Immunodeficient mice engrafted with human peripheral blood mononuclear cells (PBMCs) support preclinical studies of human pathogens, allograft rejection, and human T-cell function. However, a major limitation of PBMC engraftment is development of acute xenogeneic graft- versus-host disease (GVHD) due to human T-cell recognition of murine major histocompatibility complex (MHC). To address this, we created 2 NOD- scid IL-2 receptor subunit γ ( IL2rg) null (NSG) strains that lack murine MHC class I and II [NSG-β-2-microglobulin ( B2M) null ( IA IE)null and NSG -( Kb Db) null ( IAnull)]. We observed rapid human IgG clearance in NSG- B2Mnull ( IA IE) null mice whereas clearance in NSG -( Kb Db) null ( IAnull) mice and NSG mice was comparable. Injection of human PBMCs into both strains enabled long-term engraftment of human CD4+ and CD8+ T cells without acute GVHD. Engrafted human T-cell function was documented by rejection of human islet allografts. Administration of human IL-2 to NSG -( Kb Db) null ( IAnull) mice via adeno-associated virus vector increased human CD45+ cell engraftment, including an increase in human regulatory T cells. However, high IL-2 levels also induced the development of GVHD. These data document that NSG mice deficient in murine MHC support studies of human immunity in the absence of acute GVHD and enable evaluation of human antibody therapeutics targeting human T cells.-Brehm, M. A., Kenney, L. L., Wiles, M. V., Low, B. E., Tisch, R. M., Burzenski, L., Mueller, C., Greiner, D. L., Shultz, L. D. Lack of acute xenogeneic graft- versus-host disease, but retention of T-cell function following engraftment of human peripheral blood mononuclear cells in NSG mice deficient in MHC class I and II expression.
Collapse
Affiliation(s)
- Michael A. Brehm
- Diabetes Center of Excellence University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Laurie L. Kenney
- Diabetes Center of Excellence University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | | | - Roland M. Tisch
- Department of Immunology and Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; and
| | | | - Christian Mueller
- Department of Pediatrics and Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Dale L. Greiner
- Diabetes Center of Excellence University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | |
Collapse
|
11
|
Shao W, Earley LF, Chai Z, Chen X, Sun J, He T, Deng M, Hirsch ML, Ting J, Samulski RJ, Li C. Double-stranded RNA innate immune response activation from long-term adeno-associated virus vector transduction. JCI Insight 2018; 3:120474. [PMID: 29925692 DOI: 10.1172/jci.insight.120474] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/17/2018] [Indexed: 12/16/2022] Open
Abstract
Data from clinical trials for hemophilia B using adeno-associated virus (AAV) vectors have demonstrated decreased transgenic coagulation factor IX (hFIX) expression 6-10 weeks after administration of a high vector dose. While it is likely that capsid-specific cytotoxic T lymphocytes eliminate vector-transduced hepatocytes, thereby resulting in decreased hFIX, this observation is not intuitively consistent with restored hFIX levels following prednisone application. Although the innate immune response is immediately activated following AAV vector infection via TLR pathways, no studies exist regarding the role of the innate immune response at later time points after AAV vector transduction. Herein, activation of the innate immune response in cell lines, primary human hepatocytes, and hepatocytes in a human chimeric mouse model was observed at later time points following AAV vector transduction. Mechanistic analysis demonstrated that the double-stranded RNA (dsRNA) sensor MDA5 was necessary for innate immune response activation and that transient knockdown of MDA5, or MAVS, decreased IFN-β expression while increasing transgene production in AAV-transduced cells. These results both highlight the role of the dsRNA-triggered innate immune response in therapeutic transgene expression at later time points following AAV transduction and facilitate the execution of effective strategies to block the dsRNA innate immune response in future clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Junjiang Sun
- Gene Therapy Center.,Division of Pharmacoengineering and Molecular Pharmaceutics, School of Pharmacy
| | | | - Meng Deng
- Lineberger Comprehensive Cancer Center
| | | | | | | | - Chengwen Li
- Gene Therapy Center.,Department of Pediatrics, School of Medicine, and.,Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
12
|
Zhang X, He T, Chai Z, Samulski RJ, Li C. Blood-brain barrier shuttle peptides enhance AAV transduction in the brain after systemic administration. Biomaterials 2018; 176:71-83. [PMID: 29860139 DOI: 10.1016/j.biomaterials.2018.05.041] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 12/20/2022]
Abstract
The adeno-associated virus (AAV) vector has been used in preclinical and clinical trials of gene therapy for central nervous system (CNS) diseases. One of the biggest challenges of effectively delivering AAV to the brain is to surmount the blood-brain barrier (BBB). Herein, we identified several potential BBB shuttle peptides that significantly enhanced AAV8 transduction in the brain after a systemic administration, the best of which was the THR peptide. The enhancement of AAV8 brain transduction by THR is dose-dependent, and neurons are the primary THR targets. Mechanism studies revealed that THR directly bound to the AAV8 virion, increasing its ability to cross the endothelial cell barrier. Further experiments showed that binding of THR to the AAV virion did not interfere with AAV8 infection biology, and that THR competitively blocked transferrin from binding to AAV8. Taken together, our results demonstrate, for the first time, that BBB shuttle peptides are able to directly interact with AAV and increase the ability of the AAV vectors to cross the BBB for transduction enhancement in the brain. These results will shed important light on the potential applications of BBB shuttle peptides for enhancing brain transduction with systemic administration of AAV vectors.
Collapse
Affiliation(s)
- Xintao Zhang
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ting He
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zheng Chai
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27510, USA.
| |
Collapse
|
13
|
Pei X, He T, Hall NE, Gerber D, Samulski RJ, Li C. AAV8 virions hijack serum proteins to increase hepatocyte binding for transduction enhancement. Virology 2018; 518:95-102. [PMID: 29455066 PMCID: PMC5911186 DOI: 10.1016/j.virol.2018.02.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/02/2018] [Accepted: 02/05/2018] [Indexed: 12/19/2022]
Abstract
It has been demonstrated that human serum albumin (HSA) directly interacts with AAV virions and enhances AAV transduction. Several other proteins have also been identified a potential for enhancing AAV8 liver transduction. In our study, LDL or transferrin could enhance transduction in vitro and in vivo. We also found that any combination of two or three of these proteins (HSA, LDL, and transferrin) increased AAV8 transduction in Huh7 cells and in mice liver, which was similar to albumin alone. Pre-incubation of HSA with AAV8 virions prevented AAV8 virions from binding to other proteins. Furthermore, these serum protein receptors didn't impact AAV8 transduction but blocked the transduction enhancement from AAV8-serum protein complexes. These results indicate that serum proteins are hijacked by AAV8 vectors to increase hepatocyte binding, which shares same binding site. Importantly, the results could help us design an optimal formulation for effective AAV vector delivery in future clinical trials.
Collapse
Affiliation(s)
- Xiaolei Pei
- Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China; Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Ting He
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Nikita E Hall
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - David Gerber
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
14
|
Pei X, Earley LF, He Y, Chen X, Hall NE, Samulski RJ, Li C. Efficient Capsid Antigen Presentation From Adeno-Associated Virus Empty Virions In Vivo. Front Immunol 2018; 9:844. [PMID: 29725339 PMCID: PMC5916967 DOI: 10.3389/fimmu.2018.00844] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 04/05/2018] [Indexed: 12/03/2022] Open
Abstract
Adeno-associated virus (AAV) vectors have been successfully applied in clinical trials for hemophilic patients. Although promising, the clinical results suggest that the capsid-specific CD8+T cell response has a negative effect on therapeutic success. In an in vitro analysis using an engineered AAV virus carrying immune-dominant SIINFEKL peptide in the capsid backbone, we have previously demonstrated that capsid antigen presentation from full (genome containing) AAV capsids requires endosome escape and is proteasome dependent and that no capsid antigen presentation is induced from empty virions. In the present study, we examined capsid antigen presentation from administration of empty virions in animal models. In wild-type mice, similar to AAV full particles, capsid antigen presentation from AAV empty virion infection was dose dependent, and the kinetics studies showed that antigen presentation was detected from 2 to 40 days after AAV empty virion administration. In the transporter associated with antigen processing 1 deficient (TAP−/−) mice, capsid antigen presentation was inhibited from both AAV full and empty virions, but higher inhibition was achieved from AAV full particle administration than that from empty virions. This indicates that the pathway of capsid antigen presentation from AAV transduction is dependent on proteasome-mediated degradation of AAV capsids (mainly for full particles) and that the endosomal pathway may also play a role in antigen presentation from empty particles but not full virions. The capsid antigen presentation efficiency from AAV preparations was positively correlated with the amount of empty virions contaminated with full particles. Collectively, the results indicate that contamination of AAV empty virions induces efficient antigen presentation in vivo and the mechanism of capsid antigen presentation from empty virions involves both endosomal and proteasomal pathways. The elucidation of capsid antigen presentation from AAV empty virions may allow us to rationally design effective strategies to prevent elimination of AAV transduced target cells by capsid specific CD8+ T cells.
Collapse
Affiliation(s)
- Xiaolei Pei
- Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Hematology and Blood Diseases Hospital, Tianjin, China.,Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lauriel Freya Earley
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yi He
- Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Hematology and Blood Diseases Hospital, Tianjin, China.,Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Xiaojing Chen
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Nikita Elexa Hall
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Richard Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
15
|
Durost PA, Aryee KE, Manzoor F, Tisch RM, Mueller C, Jurczyk A, Shultz LD, Brehm MA. Gene Therapy with an Adeno-Associated Viral Vector Expressing Human Interleukin-2 Alters Immune System Homeostasis in Humanized Mice. Hum Gene Ther 2018; 29:352-365. [DOI: 10.1089/hum.2017.072] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Philip A. Durost
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Ken-Edwin Aryee
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Fatima Manzoor
- Department of Immunology and Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Roland M. Tisch
- Department of Immunology and Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Christian Mueller
- Department of Pediatrics and Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Agata Jurczyk
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | | | - Michael A. Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
16
|
Chai Z, Sun J, Rigsbee KM, Wang M, Samulski RJ, Li C. Application of polyploid adeno-associated virus vectors for transduction enhancement and neutralizing antibody evasion. J Control Release 2017; 262:348-356. [PMID: 28789965 PMCID: PMC5819605 DOI: 10.1016/j.jconrel.2017.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/18/2017] [Accepted: 08/02/2017] [Indexed: 01/13/2023]
Abstract
Adeno-associated virus (AAV) vectors have been used successfully in clinical trials for patients with hemophilia or blindness, but pre-existing neutralizing antibodies (Nab) are common in the general population and exclude many patients from clinical trials. Exploration of effective strategies to enhance AAV transduction and escape from Nab activity is still imperative. Previous studies have shown the compatibility of capsids from AAV serotypes and homology of recognition sites of AAV Nab located on different capsid subunits from one virion. In this study, we co-transfected AAV2 and AAV8 helper plasmids at different ratios (3:1, 1:1 and 1:3) to assemble haploid capsids and study both their transduction efficiency and Nab escape activity. After muscular injection, all of the haploid viruses induced higher transduction than their parental AAV vectors (2- to 9-fold over AAV2), with the highest of these being the haploid vector AAV2/8 3:1. After systemic administration, a 4-fold higher transduction in the liver was observed with haploid AAV2/8 1:3 than that with AAV8 alone. We then packaged the therapeutic factor IX cassette into haploid AAV2/8 1:3 capsids and injected them into FIX knockout mice via the tail vein. Higher FIX expression and improved phenotypic correction were achieved with the haploid AAV2/8 1:3 virus vector when compared to that of AAV8. Additionally, the haploid virus AAV2/8 1:3 was able to escape AAV2 neutralization and did not increase capsid antigen presentation capacity when compared to AAV8. To improve the Nab evasion ability of the haploid virus, we produced the triploid vector AAV2/8/9 by co-transfecting AAV2, AAV8 and AAV9 helper plasmids at a ratio of 1:1:1. After systemic administration, a 2-fold higher transduction in the liver was observed with the triploid vector AAV2/8/9 than that with AAV8. Nab analysis demonstrated that the triploid AAV2/8/9 vector was able to escape Nab activity from mouse sera immunized with parental serotypes. These results indicate that polyploid viruses might potentially acquire advantages from parental serotypes for enhancement of AAV transduction and evasion of Nab recognition without increasing capsid antigen presentation in target cells. Polyploid AAV vectors can be generated from any AAV serotype, whether natural, rational, library derived or a combination thereof, providing a novel strategy that should be explored in future clinical trials in patients with neutralizing antibodies.
Collapse
Affiliation(s)
- Zheng Chai
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Junjiang Sun
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Kelly Michelle Rigsbee
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Mei Wang
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
17
|
Li C, Wu S, Albright B, Hirsch M, Li W, Tseng YS, Agbandje-McKenna M, McPhee S, Asokan A, Samulski RJ. Development of Patient-specific AAV Vectors After Neutralizing Antibody Selection for Enhanced Muscle Gene Transfer. Mol Ther 2016; 24:53-65. [PMID: 26220272 PMCID: PMC4754536 DOI: 10.1038/mt.2015.134] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 07/18/2015] [Indexed: 01/07/2023] Open
Abstract
A major hindrance in gene therapy trials with adeno-associated virus (AAV) vectors is the presence of neutralizing antibodies (NAbs) that inhibit AAV transduction. In this study, we used directed evolution techniques in vitro and in mouse muscle to select novel NAb escape AAV chimeric capsid mutants in the presence of individual patient serum. AAV mutants isolated in vitro escaped broad patient-specific NAb activity but had poor transduction ability in vivo. AAV mutants isolated in vivo had enhanced NAb evasion from cognate serum and had high muscle transduction ability. More importantly, structural modeling identified a 100 amino acid motif from AAV6 in variable region (VR) III that confers this enhanced muscle tropism. In addition, a predominantly AAV8 capsid beta barrel template with a specific preference for AAV1/AAV9 in VR VII located at threefold symmetry axis facilitates NAb escape. Our data strongly support that chimeric AAV capsids composed of modular and nonoverlapping domains from various serotypes are capable of evading patient-specific NAbs and have enhanced muscle transduction.
Collapse
Affiliation(s)
- Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Shuqing Wu
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- China National Academy of Nanotechnology & Engineering, Tianjin, China
| | - Blake Albright
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Matthew Hirsch
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wuping Li
- Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Beijing, China
| | - Yu-Shan Tseng
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Scott McPhee
- Asklepios BioPharmaceutical Inc., Chapel Hill, North Carolina, USA
| | - Aravind Asokan
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
18
|
Nance ME, Duan D. Perspective on Adeno-Associated Virus Capsid Modification for Duchenne Muscular Dystrophy Gene Therapy. Hum Gene Ther 2015; 26:786-800. [PMID: 26414293 PMCID: PMC4692109 DOI: 10.1089/hum.2015.107] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/01/2015] [Indexed: 12/19/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a X-linked, progressive childhood myopathy caused by mutations in the dystrophin gene, one of the largest genes in the genome. It is characterized by skeletal and cardiac muscle degeneration and dysfunction leading to cardiac and/or respiratory failure. Adeno-associated virus (AAV) is a highly promising gene therapy vector. AAV gene therapy has resulted in unprecedented clinical success for treating several inherited diseases. However, AAV gene therapy for DMD remains a significant challenge. Hurdles for AAV-mediated DMD gene therapy include the difficulty to package the full-length dystrophin coding sequence in an AAV vector, the necessity for whole-body gene delivery, the immune response to dystrophin and AAV capsid, and the species-specific barriers to translate from animal models to human patients. Capsid engineering aims at improving viral vector properties by rational design and/or forced evolution. In this review, we discuss how to use the state-of-the-art AAV capsid engineering technologies to overcome hurdles in AAV-based DMD gene therapy.
Collapse
MESH Headings
- Animals
- Capsid/chemistry
- Capsid/metabolism
- Capsid Proteins/genetics
- Capsid Proteins/metabolism
- Dependovirus/genetics
- Dependovirus/metabolism
- Dystrophin/deficiency
- Dystrophin/genetics
- Gene Expression
- Genetic Therapy/methods
- Genetic Vectors/chemistry
- Genetic Vectors/metabolism
- Humans
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/metabolism
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Animal/therapy
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/therapy
- Mutation
- Protein Engineering
- Species Specificity
Collapse
Affiliation(s)
- Michael E. Nance
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
19
|
Assmann JC, Körbelin J, Schwaninger M. Genetic manipulation of brain endothelial cells in vivo. Biochim Biophys Acta Mol Basis Dis 2015; 1862:381-94. [PMID: 26454206 DOI: 10.1016/j.bbadis.2015.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/30/2015] [Accepted: 10/05/2015] [Indexed: 12/19/2022]
Affiliation(s)
- Julian C Assmann
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Jakob Körbelin
- University Medical Center Hamburg-Eppendorf, Hubertus Wald Cancer Center, Department of Oncology and Hematology, Martinistr. 52, 20246 Hamburg, Germany
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany.
| |
Collapse
|
20
|
Weinberg MS, Nicolson S, Bhatt AP, McLendon M, Li C, Samulski RJ. Recombinant adeno-associated virus utilizes cell-specific infectious entry mechanisms. J Virol 2014; 88:12472-84. [PMID: 25142580 PMCID: PMC4248914 DOI: 10.1128/jvi.01971-14] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 08/07/2014] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Understanding the entry and trafficking mechanism(s) of recombinant adeno-associated virus (rAAV) into host cells can lead to evolution in capsid and vector design and delivery methods, resulting in enhanced transduction and therapeutic gene expression. Variability of findings regarding the early entry pathway of rAAV supports the possibility that rAAV, like other viruses, can utilize more than one infectious entry pathway. We tested whether inhibition of macropinocytosis impacted rAAV transduction of HeLa cells compared to hepatocellular carcinoma cell lines. We found that macropinocytosis inhibitor cytochalasin D blocked rAAV transduction of HeLa cells (>2-fold) but enhanced (10-fold) transduction in HepG2 and Huh7 lines. Similar results were obtained with another macropinocytosis inhibitor, 5-(N-ethyl-N-isopropyl) amiloride (EIPA). The augmented transduction was due to neither viral binding nor promoter activity, affected multiple rAAV serotypes (rAAV2, rAAV2-R585E, and rAAV8), and influenced single-stranded and self-complementary virions to comparable extents. Follow-up studies using CDC42 inhibitor ML141 and p21-activated kinase 1 (PAK1) siRNA knockdown also resulted in enhanced HepG2 transduction. Microscopy revealed that macropinocytosis inhibition correlated with expedited nuclear entry of the rAAV virions into HepG2 cells. Enhancement of hepatocellular rAAV transduction extended to the mouse liver in vivo (4-fold enhancement) but inversely blocked heart tissue transduction (13-fold). This evidence of host cell-specific rAAV entry pathways confers a potent means for controlling and enhancing vector delivery and could help unify the divergent accounts of rAAV cellular entry mechanisms. IMPORTANCE There is a recognized need for improved rAAV vector targeting strategies that result in delivery of fewer total particles, averting untoward toxicity and/or an immune response against the vector. A critical step in rAAV transduction is entry and early trafficking through the host cellular machinery, the mechanisms of which are under continued study. However, should the early entry and trafficking mechanisms of rAAV differ across virus serotype or be dependent on host cell environment, this could expand our ability to target particular cells and tissue for selective transduction. Thus, the observation that inhibiting macropinocytosis leads to cell-specific enhancement or inhibition of rAAV transduction that extends to the organismic level exposes a new means of modulating vector targeting.
Collapse
Affiliation(s)
- Marc S Weinberg
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Sarah Nicolson
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Aadra P Bhatt
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Michael McLendon
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina, USA Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
21
|
Basner-Tschakarjan E, Mingozzi F. Cell-Mediated Immunity to AAV Vectors, Evolving Concepts and Potential Solutions. Front Immunol 2014; 5:350. [PMID: 25101090 PMCID: PMC4107954 DOI: 10.3389/fimmu.2014.00350] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 07/08/2014] [Indexed: 11/13/2022] Open
Abstract
Adeno-associated virus (AAV) vectors are one of the most efficient in vivo gene delivery platforms. Over the past decade, clinical trials of AAV vector-mediated gene transfer led to some of the most exciting results in the field of gene therapy and, recently, to the market approval of an AAV-based drug in Europe. With clinical development, however, it became obvious that the host immune system represents an important obstacle to successful gene transfer with AAV vectors. In this review article, we will discuss the issue of cytotoxic T cell responses directed against the AAV capsid encountered on human studies. While over the past several years the field has acquired a tremendous amount of information on the interactions of AAV vectors with the immune system, a lot of questions are still unanswered. Novel concepts are emerging, such as the relationship between the total capsid dose and the T cell-mediated clearance of transduced cells, the potential role of innate immunity in vector immunogenicity highlighted in preclinical studies, and the cross talk between regulatory and effector T cells in the determination of the outcome of gene transfer. There is still a lot to learn about immune responses in AAV gene transfer, for example, it is not well understood what are the determinants of the kinetics of activation of T cells in response to vector administration, why not all subjects develop detrimental T cell responses following gene transfer, and whether the intervention strategies currently in use to block T cell-mediated clearance of transduced cells will be safe and effective for all gene therapy indications. Results from novel preclinical models and clinical studies will help to address these points and to reach the important goal of developing safe and effective gene therapy protocols to treat human diseases.
Collapse
Affiliation(s)
| | - Federico Mingozzi
- University Pierre and Marie Curie , Paris , France ; Genethon , Evry , France
| |
Collapse
|
22
|
Gao K, Li M, Zhong L, Su Q, Li J, Li S, He R, Zhang Y, Hendricks G, Wang J, Gao G. Empty Virions In AAV8 Vector Preparations Reduce Transduction Efficiency And May Cause Total Viral Particle Dose-Limiting Side-Effects. Mol Ther Methods Clin Dev 2014; 1:20139. [PMID: 25485285 PMCID: PMC4255953 DOI: 10.1038/mtm.2013.9] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 11/12/2013] [Indexed: 02/05/2023]
Abstract
Empty virions are inadvertent by-products of recombinant adeno-associated virus (rAAV) packaging process, resulting in vector lots with mixtures of full and empty virions at variable ratios. Impact of empty virions on the efficiency and side-effects of rAAV transduction has not been well characterized. Here, we generated partially and completely empty AAV8 virions, fully packaged rAAV8 lots as well as mixtures of empty and fully packaged virions with variable ratios of empty virions (REVs). The aforementioned dosing formulations of rAAV8 expressing either cellular (EGFP or nuclear-targeted (n) LacZ) or secreted (human α1-antitrypsin, hA1AT) reporter genes were intravenously injected into two different mouse strains, followed by analyses of transgene expressions and serum alanine aminotransferase (ALT) levels at different time points. We found that addition of empty particles to the fixed doses of rAAV8 preparations repressed liver transduction up to 64% (serum hA1AT) and 44% (nLacZ) in C57BL/6 mice, respectively. The similar trend in inhibiting EGFP expression together with concurrent elevations of serum ATL levels were observed in the BALB/c mice, indicating that empty particles may also exacerbate side-effects of rAAV8EGFP transduction. Our results suggest that removal of empty particles from rAAV preparations may improve efficacy and safety of AAV in clinical applications.
Collapse
Affiliation(s)
- Kai Gao
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- National Institutes for Food and Drug Control, Beijing, China
| | - Mengxin Li
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Microbiology and Physiology Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Li Zhong
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Division of Hematology/Oncology, Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Qin Su
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jia Li
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Microbiology and Physiology Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Shaoyong Li
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Microbiology and Physiology Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Ran He
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Yu Zhang
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Oncology, Guizhou People’s Hospital, Guiyang, Guizhou, China
| | - Gregory Hendricks
- Department of Cell and Developmental Biology, Electron Microscopy Core Facility, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Junzhi Wang
- National Institutes for Food and Drug Control, Beijing, China
| | - Guangping Gao
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Microbiology and Physiology Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|