Adeno Associated Virus

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 360 Experts worldwide ranked by ideXlab platform

James M Wilson - One of the best experts on this subject based on the ideXlab platform.

  • deamidation of amino acids on the surface of Adeno Associated Virus capsids leads to charge heterogeneity and altered vector function
    Molecular Therapy, 2018
    Co-Authors: April R Giles, Kevin B Turner, Mauricio R Alvira, Martin Lock, Joshua J Sims, Lakshmanan Govindasamy, James M Wilson
    Abstract:

    Post-translational modification of the Adeno-Associated Virus capsids is a poorly understood factor in the development of these viral vectors into pharmaceutical products. Here we report the extensive capsid deamidation of Adeno-Associated Virus serotype 8 and seven other diverse Adeno-Associated Virus serotypes, with supporting evidence from structural, biochemical, and mass spectrometry approaches. The extent of deamidation at each site depended on the vector’s age and multiple primary-sequence and three-dimensional structural factors. However, the extent of deamidation was largely independent of the vector recovery and purification conditions. We demonstrate the potential for deamidation to impact transduction activity and, moreover, correlate an early time point loss in vector activity to rapidly progressing spontaneous deamidation at several Adeno-Associated Virus 8 asparagines. We explore mutational strategies that stabilize side-chain amides, improving vector transduction and reducing the lot-to-lot molecular variability that presents a key concern in biologics manufacturing. This study illuminates a previously unknown aspect of Adeno-Associated Virus capsid heterogeneity and highlights its importance in the development of these vectors for gene therapy.

  • toxicology study of intra cisterna magna Adeno Associated Virus 9 expressing iduronate 2 sulfatase in rhesus macaques
    Molecular therapy. Methods & clinical development, 2018
    Co-Authors: Juliette Hordeaux, Elizabeth L. Buza, Tamara Goode, Laura K. Richman, Christian Hinderer, Peter Bell, Roberto Calcedo, James M Wilson
    Abstract:

    Hunter syndrome is an X-linked recessive disease caused by deficiency of the lysosomal enzyme iduronate-2-sulfatase. The severe form of this progressive, systemic, and neurodegenerative disease results in loss of cognitive skills and early death. Several clinical trials are evaluating Adeno-Associated Virus 9 for the treatment of neurodegenerative diseases using systemic or intrathecal lumbar administration. In large animals, administration via suboccipital puncture gives better brain transduction than lumbar administration. Here, we conducted a good laboratory practice-compliant investigational new drug-enabling study to determine the safety of suboccipital Adeno-Associated Virus 9 gene transfer of human iduronate-2-sulfatase into nonhuman primates. Thirteen rhesus macaques received vehicle or one of two doses of vector with or without immunosuppression. We assessed in-life safety and immune responses. Animals were euthanized 90 days post-administration and sampled for histopathology and biodistribution. The procedure was well tolerated in all animals. Minimal mononuclear cerebrospinal fluid pleocytosis occurred in some animals. Asymptomatic minimal-to-moderate toxicity to some dorsal root ganglia sensory neurons and their Associated axons occurred in all vector-treated animals. This study supports the clinical development of suboccipital Adeno-Associated Virus 9 delivery for severe Hunter syndrome and highlights a potential toxicity that warrants monitoring in first-in-human studies.

  • Toxicology Study of Intra-Cisterna Magna Adeno-Associated Virus 9 Expressing Iduronate-2-Sulfatase in Rhesus Macaques
    Elsevier, 2018
    Co-Authors: Juliette Hordeaux, Elizabeth L. Buza, Tamara Goode, Laura K. Richman, Christian Hinderer, Peter Bell, Roberto Calcedo, James M Wilson
    Abstract:

    Hunter syndrome is an X-linked recessive disease caused by deficiency of the lysosomal enzyme iduronate-2-sulfatase. The severe form of this progressive, systemic, and neurodegenerative disease results in loss of cognitive skills and early death. Several clinical trials are evaluating Adeno-Associated Virus 9 for the treatment of neurodegenerative diseases using systemic or intrathecal lumbar administration. In large animals, administration via suboccipital puncture gives better brain transduction than lumbar administration. Here, we conducted a good laboratory practice-compliant investigational new drug-enabling study to determine the safety of suboccipital Adeno-Associated Virus 9 gene transfer of human iduronate-2-sulfatase into nonhuman primates. Thirteen rhesus macaques received vehicle or one of two doses of vector with or without immunosuppression. We assessed in-life safety and immune responses. Animals were euthanized 90 days post-administration and sampled for histopathology and biodistribution. The procedure was well tolerated in all animals. Minimal mononuclear cerebrospinal fluid pleocytosis occurred in some animals. Asymptomatic minimal-to-moderate toxicity to some dorsal root ganglia sensory neurons and their Associated axons occurred in all vector-treated animals. This study supports the clinical development of suboccipital Adeno-Associated Virus 9 delivery for severe Hunter syndrome and highlights a potential toxicity that warrants monitoring in first-in-human studies. Keywords: AAV9, intrathecal, MPS I

  • cpg depleted Adeno Associated Virus vectors evade immune detection
    Journal of Clinical Investigation, 2013
    Co-Authors: Susan M Faust, Peter Bell, Benjamin J Cutler, Scott Ashley, Yanqing Zhu, Joseph E Rabinowitz, James M Wilson
    Abstract:

    Due to their efficient transduction potential, Adeno-Associated Virus (AAV) vectors are leading candidates for gene therapy in skeletal muscle diseases. However, immune responses toward the vector or transgene product have been observed in preclinical and clinical studies. TLR9 has been implicated in promoting AAV-directed immune responses, but vectors have not been developed to circumvent this barrier. To assess the requirement of TLR9 in promoting immunity toward AAV-Associated antigens following skeletal muscle gene transfer in mice, we compared immunological responses in WT and Tlr9-deficient mice that received an AAV vector with an immunogenic capsid, AAVrh32.33. In Tlr9-deficient mice, IFN-γ T cell responses toward capsid and transgene antigen were suppressed, resulting in minimal cellular infiltrate and stable transgene expression in target muscles. These findings suggest that AAV-directed immune responses may be circumvented by depleting the ligand for TLR9 (CpG sequences) from the vector genome. Indeed, we found that CpG-depleted AAVrh32.33 vectors could establish persistent transgene expression, evade immunity, and minimize infiltration of effector cells. Thus, CpG-depleted AAV vectors could improve outcome of clinical trials of gene therapy for skeletal muscle disease.

  • Adeno Associated Virus antibody profiles in newborns children and adolescents
    Clinical and Vaccine Immunology, 2011
    Co-Authors: Roberto Calcedo, Hiroki Morizono, Lili Wang, Robert Mccarter, David Jones, Mark L Batshaw, James M Wilson
    Abstract:

    Neutralizing antibodies (NAb) to an Adeno-Associated Virus (AAV) vector due to previous natural infection with wild-type AAV can significantly limit gene transfer. NAb titers to AAV serotype 2 (AAV2) and AAV8 in human subjects (0 to 18 years) were studied. NAb prevalence is moderate at birth, decreases markedly from 7 to 11 months, and then progressively increases through childhood and adolescence.

Jude R Samulski - One of the best experts on this subject based on the ideXlab platform.

  • Adeno Associated Virus capsid promoter interactions in the brain translate from rat to the nonhuman primate
    Human Gene Therapy, 2020
    Co-Authors: Martin O Bohlen, Thomas J. Mccown, Sara K Powell, Hala G Elnahal, Tierney Daw, Michele A Basso, Marc A Sommer, Jude R Samulski
    Abstract:

    Recently, we established an Adeno-Associated Virus (AAV9) capsid-promoter interaction that directly determined cell-specific gene expression across two synthetic promoters, Cbh and CBA, in the rat ...

  • engineering Adeno Associated Virus vectors for gene therapy
    Nature Reviews Genetics, 2020
    Co-Authors: Jude R Samulski
    Abstract:

    Adeno-Associated Virus (AAV) vector-mediated gene delivery was recently approved for the treatment of inherited blindness and spinal muscular atrophy, and long-term therapeutic effects have been achieved for other rare diseases, including haemophilia and Duchenne muscular dystrophy. However, current research indicates that the genetic modification of AAV vectors may further facilitate the success of AAV gene therapy. Vector engineering can increase AAV transduction efficiency (by optimizing the transgene cassette), vector tropism (using capsid engineering) and the ability of the capsid and transgene to avoid the host immune response (by genetically modifying these components), as well as optimize the large-scale production of AAV.

  • Adeno Associated Virus serotype specific inverted terminal repeat sequence role in vector transgene expression
    Human Gene Therapy, 2020
    Co-Authors: Lauriel F Earley, Laura Conatser, Amanda Lee Dobbins, Matthew L Hirsch, Chengwen Li, Jude R Samulski
    Abstract:

    Adeno-Associated viral vectors have been successfully used in laboratory and clinical settings for efficient gene delivery. In these vectors, 96% of the Adeno-Associated Virus (AAV) genome is repla...

  • Adeno Associated Virus vectorology manufacturing and clinical applications
    Methods in Enzymology, 2012
    Co-Authors: Joshua C Grieger, Jude R Samulski
    Abstract:

    Adeno-Associated Virus (AAV) has emerged as an attractive vector for gene therapy. The benefits of using AAV for gene therapy include long-term gene expression, the inability to autonomously replicate without a helper Virus, transduction of dividing and nondividing cells, and the lack of pathogenicity from wild-type infections. A number of Phase I and Phase II clinical trials utilizing AAV have been carried out worldwide (Aucoin et al., 2008; Mueller and Flotte, 2008). A number of challenges have been identified based upon data generated from these clinical trials. These challenges include (1) large scale manufacturing technologies in accordance with current Good Manufacturing Practices (cGMP), (2) tissue specific tropism of AAV vectors, (3) high-quality/high potency recombinant AAV vectors (rAAV), and (4) immune response to AAV capsids and transgene. In this chapter, we will provide an overview of AAV biology, AAV vectorology, rAAV manufacturing, and the current status on the latest rAAV clinical trials.

  • inducible Adeno Associated Virus mediated il 2 gene therapy prevents autoimmune diabetes
    Journal of Immunology, 2011
    Co-Authors: Kevin Goudy, Jude R Samulski, Chengwen Li, Mark C Johnson, Alaina L Garland, Bo Wang, Roland Tisch
    Abstract:

    IL-2 and TGF-β1 play key roles in the immunobiology of Foxp3-expressing CD25+CD4+ T cells (Foxp3+Treg). Administration of these cytokines offers an appealing approach to manipulate the Foxp3+Treg pool and treat T cell-mediated autoimmunity such as type 1 diabetes. However, efficacy of cytokine treatment is dependent on the mode of application, and the potent pleiotropic effects of cytokines like IL-2 may lead to severe side effects. In the current study, we used a gene therapy-based approach to assess the efficacy of recombinant Adeno-Associated Virus vectors expressing inducible IL-2 or TGF-β1 transgenes to suppress ongoing β cell autoimmunity in NOD mice. Intramuscular vaccination of recombinant Adeno-Associated Virus to 10-wk-old NOD female mice and a subsequent 3 wk induction of IL-2 was sufficient to prevent diabetes and block the progression of insulitis. Protection correlated with an increased frequency of Foxp3+Treg in the periphery as well as in the draining pancreatic lymph nodes and islets. IL-2 induced a shift in the ratio favoring Foxp3+Treg versus IFN-γ–expressing T cells infiltrating the islets. Induction of IL-2 had no systemic effect on the frequency or activational status of T cells and NK cells. Induction of TGF-β1 had no effect on the Foxp3+Treg pool or the progression of β cell autoimmunity despite induced systemic levels of activated TGF-β1 that were comparable to IL-2. These results demonstrate that inducible IL-2 gene therapy is an effective and safe approach to manipulate Foxp3+Treg and suppress T cell-mediated autoimmunity and that under the conditions employed, IL-2 is more potent than TGF-β1.

Nicholas Muzyczka - One of the best experts on this subject based on the ideXlab platform.

  • Structure of Adeno-Associated Virus Type 4
    Journal of virology, 2005
    Co-Authors: Eric Padron, Lakshmanan Govindasamy, John A. Chiorini, Valorie D. Bowman, Nikola Kaludov, Hazel C. Levy, Phillip Nick, Nicholas Muzyczka, Timothy S. Baker
    Abstract:

    Adeno-Associated Virus (AAV) is a member of the Parvoviridae, belonging to the DependoVirus genus. Currently, several distinct isolates of AAV are in development for use in human gene therapy applications due to their ability to transduce different target cells. The need to manipulate AAV capsids for specific tissue delivery has generated interest in understanding their capsid structures. The structure of AAV type 4 (AAV4), one of the most antigenically distinct serotypes, was determined to 13-A resolution by cryo-electron microscopy and image reconstruction. A pseudoatomic model was built for the AAV4 capsid by use of a structure-based sequence alignment of its major capsid protein, VP3, with that of AAV2, to which AAV4 is 58% identical and constrained by its reconstructed density envelope. The model showed variations in the surface loops that may account for the differences in receptor binding and antigenicity between AAV2 and AAV4. The AAV4 capsid surface topology also shows an unpredicted structural similarity to that of Aleutian mink disease Virus and human parvoVirus B19, autonomous members of the genus, despite limited sequence homology.

  • Structure of Adeno-Associated Virus Type 4
    2004
    Co-Authors: Eric Padron, Lakshmanan Govindasamy, John A. Chiorini, Nikola Kaludov, Phillip Nick, Nicholas Muzyczka, Valorie Bowman, Hazel Levy, Timothy S. Baker
    Abstract:

    Adeno-Associated Virus (AAV) is a member of the Parvoviridae, belonging to the DependoVirus genus. Currently, several distinct isolates of AAV are in development for use in human gene therapy applications due to their ability to transduce different target cells. The need to manipulate AAV capsids for specific tissue delivery has generated interest in understanding their capsid structures. The structure of AAV type 4 (AAV4), one of the most antigenically distinct serotypes, was determined to 13-Å resolution by cryo-electron microscopy and image reconstruction. A pseudoatomic model was built for the AAV4 capsid by use of a structure-based sequence alignment of its major capsid protein, VP3, with that of AAV2, to which AAV4 is 58 % identical and constrained by its reconstructed density envelope. The model showed variations in the surface loops that may account for the differences in receptor binding and antigenicity between AAV2 and AAV4. The AAV4 capsid surface topology also shows an unpredicted structural similarity to that of Aleutian mink disease Virus and human parvoVirus B19, autonomous members of the genus, despite limited sequence homology. Adeno-Associated Virus (AAV) is a member of the Parvoviridae family (45). AAV virions have a T�1 icosahedral capsid consisting of 60 copies of three related proteins, VP1, VP2, and VP3, at an estimated ratio of 1:1:8, which surrounds a singlestrande

  • Adeno Associated Virus vector mediated gene transfer to somatic cells in the central nervous system
    Advances in Virus Research, 2000
    Co-Authors: Ronald L Klein, Ronald J Mandel, Nicholas Muzyczka
    Abstract:

    Publisher Summary This chapter focuses on Adeno-Associated Virus vector-mediated gene transfer to somatic cells in the central nervous system (CNS). The recombinant Adeno-Associated Virus (rAAV) vector system incorporates features that make it a good choice for in vivo research. These features include efficient and long-term gene expression in a broad range of organs that appears to be well tolerated by laboratory animals. The brain is a, particularly good target for viral vector approaches because of the topic maps of neuroanatomical organization. Wild-type AAV infection commonly occurs early in life, and up to 80% of the human population is seropositive for AAV. AAV is a dependoVirus requiring a helper Virus to complete its life cycle. Apart from gene therapy applications, in vivo rAAV somatic cell gene transfer is likely to be useful for disease modeling. The pathology of Parkinson's disease is characterized by movement disorders concomitant with the degeneration of dopaminergic neurons in the substantia nigra and their axons in the striatum. Alzheimer's disease is, in part, a cholinergic deficit supported by the role of the neurotransmitter acetylcholine in memory function.

  • recombinant Adeno Associated Virus purification using novel methods improves infectious titer and yield
    Gene Therapy, 1999
    Co-Authors: Sergei Zolotukhin, Irene Zolotukhin, Barry J Byrne, Richard J Samulski, E Mason, Mark R Potter, Kye Chesnut, C Summerford, Nicholas Muzyczka
    Abstract:

    Recombinant Adeno-Associated Virus purification using novel methods improves infectious titer and yield

Lili Wang - One of the best experts on this subject based on the ideXlab platform.

Jerry R Mendell - One of the best experts on this subject based on the ideXlab platform.

  • Adeno Associated Virus serotype 9 antibodies in patients screened for treatment with onasemnogene abeparvovec
    Molecular therapy. Methods & clinical development, 2021
    Co-Authors: John W Day, R Finkel, Eugenio Mercuri, Kathryn J Swoboda, Melissa Menier, Rudolf Van Olden, Sitra Tauscherwisniewski, Jerry R Mendell
    Abstract:

    Spinal muscular atrophy is a progressive, recessively inherited monogenic neurologic disease, the genetic root cause of which is the absence of a functional survival motor neuron 1 gene. Onasemnogene abeparvovec (formerly AVXS-101) is an Adeno-Associated Virus serotype 9 vector-based gene therapy that delivers a fully functional copy of the human survival motor neuron gene. We report anti–Adeno-Associated Virus serotype 9 antibody titers for patients with spinal muscular atrophy when they were screened for eligibility in the onasemnogene abeparvovec clinical trials (intravenous and intrathecal administration) and managed access programs (intravenous). Through December 31, 2019, 196 patients and 155 biologic mothers were screened for anti–Adeno-Associated Virus serotype 9 binding antibodies with an enzyme-linked immunosorbent assay. Of these, 15 patients (7.7%) and 23 biologic mothers (14.8%) had titers >1:50 on their initial screening tests. Eleven patients (5.6%) had elevated titers on their final screening tests. The low percentage of patients with exclusionary antibody titers indicates that most infants with spinal muscular atrophy type 1 should be able to receive onasemnogene abeparvovec. Retesting may identify patients whose antibody titers later decrease to below the threshold for treatment, and retesting should be considered for patients with anti–Adeno-Associated Virus serotype 9 antibody titers >1:50.