2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Prevalence and Relevance of Pre-Existing Anti-Adeno-Associated Virus Immunity in the Context of Gene Therapy for Crigler–Najjar Syndrome

      research-article

      Read this article at

          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Adeno-associated virus (AAV) vector-mediated gene therapy is currently evaluated as a potential treatment for Crigler–Najjar syndrome (CN) (NCT03466463). Pre-existing immunity to AAV is known to hinder gene transfer efficacy, restricting enrollment of seropositive subjects in ongoing clinical trials. We assessed the prevalence of anti-AAV serotype 8 (AAV8) neutralizing antibodies (NAbs) in subjects affected by CN and investigated the impact of low NAb titers (<1:5) on liver gene transfer efficacy in an in vivo passive immunization model. A total of 49 subjects with a confirmed molecular diagnosis of CN were included in an international multicenter study (NCT02302690). Pre-existing NAbs against AAV8 were detected in 30.6% (15/49) of screened patients and, in the majority of positive cases, cross-reactivity to AAV2 and AAV5 was detected. To investigate the impact of low NAbs on AAV vector-mediated liver transduction efficiency, adult wild-type C57BL/6 mice were passively immunized with pooled human donor-derived immunoglobulins to achieve titers of up to 1:3.16. After immunization, animals were injected with different AAV8 vector preparations. Hepatic vector gene copy number was unaffected by low anti-AAV8 NAb titers when column-purified AAV vector batches containing both full and empty capsids were used. In summary, although pre-existing anti-AAV8 immunity can be found in about a third of subjects affected by CN, low anti-AAV8 NAb titers are less likely to affect liver transduction efficiency when using AAV vector preparations manufactured to contain both full and empty capsids. These findings have implications for the design of liver gene transfer clinical trials and for the definition of inclusion criteria related to seropositivity of potential participants.

          Related collections

          Most cited references27

          • Record: found
          • Abstract: found
          • Article: not found

          Overcoming preexisting humoral immunity to AAV using capsid decoys.

          Adeno-associated virus (AAV) vectors delivered through the systemic circulation successfully transduce various target tissues in animal models. However, similar attempts in humans have been hampered by the high prevalence of neutralizing antibodies to AAV, which completely block vector transduction. We show in both mouse and nonhuman primate models that addition of empty capsid to the final vector formulation can, in a dose-dependent manner, adsorb these antibodies, even at high titers, thus overcoming their inhibitory effect. To further enhance the safety of the approach, we mutated the receptor binding site of AAV2 to generate an empty capsid mutant that can adsorb antibodies but cannot enter a target cell. Our work suggests that optimizing the ratio of full/empty capsids in the final formulation of vector, based on a patient's anti-AAV titers, will maximize the efficacy of gene transfer after systemic vector delivery.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Antigen-selective modulation of AAV immunogenicity with tolerogenic rapamycin nanoparticles enables successful vector re-administration

            Gene therapy mediated by recombinant adeno-associated virus (AAV) vectors is a promising treatment for systemic monogenic diseases. However, vector immunogenicity represents a major limitation to gene transfer with AAV vectors, particularly for vector re-administration. Here, we demonstrate that synthetic vaccine particles encapsulating rapamycin (SVP[Rapa]), co-administered with AAV vectors, prevents the induction of anti-capsid humoral and cell-mediated responses. This allows successful vector re-administration in mice and nonhuman primates. SVP[Rapa] dosed with AAV vectors reduces B and T cell activation in an antigen-selective manner, inhibits CD8+ T cell infiltration in the liver, and efficiently blocks memory T cell responses. SVP[Rapa] immunomodulatory effects can be transferred from treated to naive mice by adoptive transfer of splenocytes, and is inhibited by depletion of CD25+ T cells, suggesting a role for regulatory T cells. Co-administration of SVP[Rapa] with AAV vector represents a powerful strategy to modulate vector immunogenicity and enable effective vector re-administration.
              Bookmark
              • Record: found
              • Abstract: not found
              • Article: not found

              Bilirubin-induced neurologic damage--mechanisms and management approaches.

                Bookmark

                Author and article information

                Journal
                Hum Gene Ther
                Hum. Gene Ther
                hum
                Human Gene Therapy
                Mary Ann Liebert, Inc., publishers (140 Huguenot Street, 3rd FloorNew Rochelle, NY 10801USA )
                1043-0342
                1557-7422
                01 October 2019
                24 September 2019
                24 September 2019
                : 30
                : 10
                : 1297-1305
                Affiliations
                [ 1 ]Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
                [ 2 ]Genethon, Evry, France
                [ 3 ]Department of Hereditary Diseases of Hepatic Metabolism, Hôpital Antoine Béclère, Clamart, France
                [ 4 ]Department of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam, The Netherlands
                [ 5 ]Department of Paediatric Gastroenterology and Hepatology, Hannover Medical School, Hannover, Germany
                [ 6 ]Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
                [ 7 ]Department of Paediatric Hepatology, Gastroenterology and Transplantation, Papa Giovanni XXIII Hospital, Bergamo, Italy
                [ 8 ]Telethon Institute of Genetics & Medicine (TIGEM), Pozzuoli, Italy
                [ 9 ]Department of Translational Medicine, Federico II University of Naples, Naples, Italy.
                Author notes
                [*] [ * ]Correspondence: Dr. Federico Mingozzi, Généthon, 1 bis rue de l'Internationale, Evry 91000, France fmingozzi@ 123456genethon.fr
                Article
                10.1089/hum.2019.143
                10.1089/hum.2019.143
                6763963
                31502485
                3737fb6c-b893-475b-8967-7511ef4d2f5d
                © Sem J. Aronson et al., 2019; Published by Mary Ann Liebert, Inc.

                This Open Access article is distributed under the terms of the Creative Commons Attribution Noncommercial License ( http://creativecommons.org/licenses/by-nc/4.0/) that permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and the source are cited.

                History
                : 17 June 2019
                : 17 July 2019
                Page count
                Figures: 4, Tables: 2, References: 45, Pages: 9
                Categories
                Research Articles

                crigler–najjar syndrome,ugt1a1,unconjugated hyperbilirubinemia,aav gene therapy,anti-aav neutralizing antibodies,pre-existing immunity

                Comments

                Comment on this article