11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Assessment of Systemic Delivery of rAAVrh74.MHCK7.micro-dystrophin in Children With Duchenne Muscular Dystrophy : A Nonrandomized Controlled Trial

      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

          This nonrandomized controlled trial analyzes safety, biological, and functional outcomes associated with the infusion of rAAVrh74.MHCK7.micro-dystrophin gene transfer in a small group of patients with Duchenne muscular dystrophy.

          Key Points

          Question

          Is rAAVrh74.MHCK7.micro-dystrophin gene transfer safe and well tolerated in patients with Duchenne muscular dystrophy?

          Findings

          In this nonrandomized controlled trial of 4 young patients with Duchenne muscular dystrophy, rAAVrh74.MHCK7.micro-dystrophin gene transfer was well tolerated, with minimal adverse events, and was associated with robust micro-dystrophin expression, reduced serum creatine kinase levels, and functional improvement as measured by the North Star Ambulatory Assessment.

          Meaning

          These results indicated the safe systemic delivery of micro-dystrophin transgene and targeted expression of functional micro-dystrophin protein product, suggesting the potential for rAAVrh74.MHCK7.micro-dystrophin to provide clinically meaningful functional improvement that is greater than the standard of care.

          Abstract

          Importance

          Micro-dystrophin gene transfer shows promise for treating patients with Duchenne muscular dystrophy (DMD) using recombinant adeno-associated virus serotype rh74 (rAAVrh74) and codon-optimized human micro-dystrophin driven by a skeletal and cardiac muscle-specific promoter with enhanced cardiac expression (MHCK7).

          Objective

          To identify the 1-year safety and tolerability of intravenous rAAVrh74.MHCK7.micro-dystrophin in patients with DMD.

          Design, Setting, and Participants

          This open-label, phase 1/2a nonrandomized controlled trial was conducted at the Nationwide Children’s Hospital in Columbus, Ohio. It began on November 2, 2017, with a planned duration of follow-up of 3 years, ending in March 2021. The first 4 patients who met eligibility criteria were enrolled, consisting of ambulatory male children with DMD without preexisting AAVrh74 antibodies and a stable corticosteroid dose (≥12 weeks).

          Interventions

          A single dose of 2.0 × 10 14 vg/kg rAAVrh74.MHCK7.micro-dystrophin was infused through a peripheral limb vein. Daily prednisolone, 1 mg/kg, started 1 day before gene delivery (30-day taper after infusion).

          Main Outcomes and Measures

          Safety was the primary outcome. Secondary outcomes included micro-dystrophin expression by Western blot and immunohistochemistry. Functional outcomes measured by North Star Ambulatory Assessment (NSAA) and serum creatine kinase were exploratory outcomes.

          Results

          Four patients were included (mean [SD] age at enrollment, 4.8 [1.0] years). All adverse events (n = 53) were considered mild (33 [62%]) or moderate (20 [38%]), and no serious adverse events occurred. Eighteen adverse events were considered treatment related, the most common of which was vomiting (9 of 18 events [50%]). Three patients had transiently elevated γ-glutamyltransferase, which resolved with corticosteroids. At 12 weeks, immunohistochemistry of gastrocnemius muscle biopsy specimens revealed robust transgene expression in all patients, with a mean of 81.2% of muscle fibers expressing micro-dystrophin with a mean intensity of 96% at the sarcolemma. Western blot showed a mean expression of 74.3% without fat or fibrosis adjustment and 95.8% with adjustment. All patients had confirmed vector transduction and showed functional improvement of NSAA scores and reduced creatine kinase levels (posttreatment vs baseline) that were maintained for 1 year.

          Conclusions and Relevance

          This trial showed rAAVrh74.MHCK7.micro-dystrophin to be well tolerated and have minimal adverse events; the safe delivery of micro-dystrophin transgene; the robust expression and correct localization of micro-dystrophin protein; and improvements in creatine kinase levels and NSAA scores. These findings suggest that rAAVrh74.MHCK7.micro-dystrophin can provide functional improvement that is greater than that observed under standard of care.

          Trial Registration

          ClinicalTrials.gov Identifier: NCT03375164

          Related collections

          Most cited references45

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

          Single-Dose Gene-Replacement Therapy for Spinal Muscular Atrophy

          Spinal muscular atrophy type 1 (SMA1) is a progressive, monogenic motor neuron disease with an onset during infancy that results in failure to achieve motor milestones and in death or the need for mechanical ventilation by 2 years of age. We studied functional replacement of the mutated gene encoding survival motor neuron 1 (SMN1) in this disease.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Evidence-based path to newborn screening for Duchenne muscular dystrophy.

            Creatine kinase (CK) levels are increased on dried blood spots in newborns related to the birthing process. As a marker for newborn screening, CK in Duchenne muscular dystrophy (DMD) results in false-positive testing. In this report, we introduce a 2-tier system using the dried blood spot to first assess CK with follow-up DMD gene testing. A fluorometric assay based upon the enzymatic transphosphorylation of adenosine diphosphate to adenosine triphosphate was used to measure CK activity. Preliminary studies established a population-based range of CK in newborns using 30,547 deidentified anonymous dried blood spot samples. Mutation analysis used genomic DNA extracted from the dried blood spot followed by whole genome amplification with assessment of single-/multiexon deletions/duplications in the DMD gene using multiplex ligation-dependent probe amplification. DMD gene mutations (all exonic deletions) were found in 6 of 37,649 newborn male subjects, all of whom had CK levels>2,000U/l. In 3 newborns with CK>2,000U/l in whom DMD gene abnormalities were not found, we identified limb-girdle muscular dystrophy gene mutations affecting DYSF, SGCB, and FKRP. A 2-tier system of analysis for newborn screening for DMD has been established. This path for newborn screening fits our health care system, minimizes false-positive testing, and uses predetermined levels of CK on dried blood spots to predict DMD gene mutations. Copyright © 2012 American Neurological Association.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response.

              We have previously shown that a single portal vein infusion of a recombinant adeno-associated viral vector (rAAV) expressing canine Factor IX (F.IX) resulted in long-term expression of therapeutic levels of F.IX in dogs with severe hemophilia B. We carried out a phase 1/2 dose-escalation clinical study to extend this approach to humans with severe hemophilia B. rAAV-2 vector expressing human F.IX was infused through the hepatic artery into seven subjects. The data show that: (i) vector infusion at doses up to 2 x 10(12) vg/kg was not associated with acute or long-lasting toxicity; (ii) therapeutic levels of F.IX were achieved at the highest dose tested; (iii) duration of expression at therapeutic levels was limited to a period of approximately 8 weeks; (iv) a gradual decline in F.IX was accompanied by a transient asymptomatic elevation of liver transaminases that resolved without treatment. Further studies suggested that destruction of transduced hepatocytes by cell-mediated immunity targeting antigens of the AAV capsid caused both the decline in F.IX and the transient transaminitis. We conclude that rAAV-2 vectors can transduce human hepatocytes in vivo to result in therapeutically relevant levels of F.IX, but that future studies in humans may require immunomodulation to achieve long-term expression.
                Bookmark

                Author and article information

                Journal
                JAMA Neurol
                JAMA Neurol
                JAMA Neurol
                JAMA Neurology
                American Medical Association
                2168-6149
                2168-6157
                September 2020
                15 June 2020
                15 June 2020
                : 77
                : 9
                : 1-10
                Affiliations
                [1 ]Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
                [2 ]Department of Pediatrics, The Ohio State University, Columbus
                [3 ]Department of Neurology, The Ohio State University, Columbus
                [4 ]Sarepta Therapeutics Inc, Cambridge, Massachusetts
                [5 ]Department of Radiology, Vascular and Interventional Radiology, Nationwide Children's Hospital, Columbus, Ohio
                Author notes
                Article Information
                Accepted for Publication: February 27, 2020.
                Published Online: June 15, 2020. doi:10.1001/jamaneurol.2020.1484
                Open Access: This is an open access article distributed under the terms of the CC-BY-NC-ND License. © 2020 Mendell JR et al. JAMA Neurology.
                Corresponding Authors: Jerry R. Mendell, MD, Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, 700 Children's Dr, Columbus, OH 43205 ( Jerry.Mendell@ 123456nationwidechildrens.org ); Louise R. Rodino-Klapac, PhD, Sarepta Therapeutics Inc, 215 First St, Cambridge, MA 02142 ( LRodinoKlapac@ 123456Sarepta.com ).
                Author Contributions: Dr Mendell had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.
                Concept and design: Mendell, Lehman, Lowes, Miller, Alfano, Al-Zaidy, Church, Shell, Rodino-Klapac.
                Acquisition, analysis, or interpretation of data: Mendell, Sahenk, Lehman, Nease, Lowes, Miller, Iammarino, Alfano, Nicholl, Al-Zaidy, Lewis, Cripe, Potter, Griffin, Pozsgai, Dugar, Hogan, Rodino-Klapac.
                Drafting of the manuscript: Mendell, Lehman, Miller, Iammarino, Potter, Dugar, Rodino-Klapac.
                Critical revision of the manuscript for important intellectual content: All authors.
                Statistical analysis: Mendell, Alfano.
                Obtained funding: Mendell, Rodino-Klapac.
                Administrative, technical, or material support: Mendell, Sahenk, Lehman, Nicholl, Al-Zaidy, Lewis, Church, Shell, Cripe, Potter, Griffin, Pozsgai, Dugar, Hogan, Rodino-Klapac.
                Supervision: Mendell, Rodino-Klapac.
                Other—data collection: Lowes.
                Conflict of Interest Disclosures: Dr Mendell reported receiving grants from Parent Project Muscular Dystrophy; receiving personal fees from Sarepta Therapeutics Inc., and Nationwide Children's Hospital outside the submitted work; and holding a pending patent to micro-dystrophin cassette for gene therapy and an issued patent to rAAV.SGCA delivery isolated limb infusion. Dr Sahenk reported receiving grants from Sarepta Therapeutics Inc during the conduct of the study. Ms Lehman reported receiving grants from Sarepta Therapeutics Inc. during the conduct of the study. Ms Nease reported receiving grants from Nationwide Children's Hospital during the conduct of the study. Ms Nicholl reported receiving grants from Nationwide Children's Hospital during the conduct of the study. Dr Al-Zaidy reported receiving consulting fees from AveXis Inc outside the submitted work. Dr Lewis reported being an employee of Sarepta Therapeutics Inc. during the conduct of the study. Dr Potter reported receiving other from Sarepta Therapeutics Inc. during the conduct of the . Ms Griffin reported receiving other from Sarepta Therapeutics Inc. during the conduct of the study. Dr Pozsgai reported receiving other from Sarepta Therapeutics Inc. during the conduct of the study and outside the submitted work. Dr Dugar reported being an employee of Sarepta Therapeutics Inc. Dr Rodino-Klapac reported receiving personal fees from Myonexus Therapeutics outside the submitted work, holding a patent (pending, licensed, and with royalties paid) to adeno-associated virus delivery of muscle-specific micro-dystrophin to treat patients with muscular dystrophy, and being an employee of Sarepta Therapeutics Inc. No other disclosures were reported.
                Funding/Support: This study was funded by Sarepta Therapeutics Inc., Nationwide Children’s Foundation, and Parent Project Muscular Dystrophy.
                Role of the Funder/Sponsor: The funders had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; approval of the manuscript; and decision to submit the manuscript for publication.
                Additional Contributions: We thank the Clinical Research Services at Nationwide Children’s Hospital and its director Grace Wentzel. Yanning Liu, Ping Zheng, MS, and Alex Yang, MS, Sarepta Therapeutics Inc., provided critical review of the data. Navid Khan, PhD, Damon Asher, PhD, and Sachi Dharia, PharmD, Sarepta Therapeutics Inc., provided critical review of the manuscript. Aaron Novack, Senior Director and Head of Medical Communications at Sarepta Therapeutics Inc., provided administrative support in responding to questions and comments from the Journal. These individuals received no additional compensation, outside of their usual salary, for their contributions. Khampaseuth Thapa, PhD, Sarepta Therapeutics Inc, and Purvi Kobawala Smith, MS, MPH, Health & Wellness Partners, LLC, provided medical writing and editorial support; they received payment from Sarepta Therapeutics Inc.
                Article
                PMC7296461 PMC7296461 7296461 noi200033
                10.1001/jamaneurol.2020.1484
                7296461
                32539076
                4193cc90-2f8c-41bc-b8f3-0dedc7304498
                Copyright 2020 Mendell JR et al. JAMA Neurology.

                This is an open access article distributed under the terms of the CC-BY-NC-ND License.

                History
                : 14 November 2019
                : 27 February 2020
                Categories
                Research
                Research
                Original Investigation
                Online First
                Comments

                Comments

                Comment on this article