Evaluation of Readministration of a Recombinant Adeno-Associated Virus Vector Expressing Acid Alpha-Glucosidase in Pompe Disease: Preclinical to Clinical Planning.

A recombinant serotype 9 adeno-associated virus (rAAV9) vector carrying a transgene that expresses codon-optimized human acid alpha-glucosidase (hGAA, or GAA) driven by a human desmin (DES) promoter (i.e., rAAV9-DES-hGAA) has been generated as a clinical candidate vector for Pompe disease. The rAAV9-DES-hGAA vector is being developed as a treatment for both early- and late-onset Pompe disease, in which patients lack sufficient lysosomal alpha-glucosidase leading to glycogen accumulation. In young patients, the therapy may need to be readministered after a period of time to maintain therapeutic levels of GAA. Administration of AAV-based gene therapies is commonly associated with the production of neutralizing antibodies that may reduce the effectiveness of the vector, especially if readministration is required. Previous studies have demonstrated the ability of rAAV9-DES-hGAA to correct cardiac and skeletal muscle pathology in Gaa(-/-) mice, an animal model of Pompe disease. This article describes the IND-enabling preclinical studies supporting the program for a phase I/II clinical trial in adult patients with Pompe. These studies were designed to evaluate the toxicology, biodistribution, and potential for readministration of rAAV9-DES-hGAA injected intramuscularly into the tibialis anterior muscle using an immune modulation strategy developed for this study. In the proposed clinical study, six adult participants with late-onset Pompe disease will be enrolled. The goal of the immune modulation strategy is to ablate B-cells before the initial exposure of the study agent in one leg and the subsequent exposure of the same vector to the contralateral leg four months after initial dosing. The dosing of the active agent is accompanied by a control injection of excipient dosing in the contralateral leg to allow for blinding and randomization of dosing, which may also strengthen the evidence generated from gene therapy studies in the future. Patients will act as their own controls. Repeated measures, at baseline and during the three months following each dosing will assess the safety, biochemical, and functional impact of the vector.

[1]  B. Byrne,et al.  Altered activation of the tibialis anterior in individuals with Pompe disease: Implications for motor unit dysfunction , 2015, Muscle & nerve.

[2]  B. Byrne,et al.  Peripheral nerve and neuromuscular junction pathology in Pompe disease. , 2015, Human molecular genetics.

[3]  T. Conlon,et al.  B-cell depletion is protective against anti-AAV capsid immune response: a human subject case study , 2014, Molecular therapy. Methods & clinical development.

[4]  D. Gaudet,et al.  Immune responses to intramuscular administration of alipogene tiparvovec (AAV1-LPL(S447X)) in a phase II clinical trial of lipoprotein lipase deficiency gene therapy. , 2014, Human gene therapy.

[5]  A. Ballabio,et al.  Intracerebral administration of adeno-associated viral vector serotype rh.10 carrying human SGSH and SUMF1 cDNAs in children with mucopolysaccharidosis type IIIA disease: results of a phase I/II trial. , 2014 .

[6]  B. Byrne,et al.  The respiratory neuromuscular system in Pompe disease , 2013, Respiratory Physiology & Neurobiology.

[7]  James M. Wilson,et al.  Humoral Immune Response to AAV , 2013, Front. Immunol..

[8]  K. High,et al.  Immune responses to AAV vectors: overcoming barriers to successful gene therapy. , 2013, Blood.

[9]  Stephan Wolf,et al.  A largely random AAV integration profile after LPLD gene therapy , 2013, Nature Medicine.

[10]  F. Mingozzi,et al.  Humoral immunity to AAV vectors in gene therapy: challenges and potential solutions. , 2013, Discovery medicine.

[11]  T. Conlon,et al.  Phase I/II trial of adeno-associated virus-mediated alpha-glucosidase gene therapy to the diaphragm for chronic respiratory failure in Pompe disease: initial safety and ventilatory outcomes. , 2013, Human gene therapy.

[12]  William J Feuer,et al.  Gene therapy for leber congenital amaurosis caused by RPE65 mutations: safety and efficacy in 15 children and adults followed up to 3 years. , 2012, Archives of ophthalmology.

[13]  Pratima Chowdary,et al.  Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. , 2011, The New England journal of medicine.

[14]  B. Byrne,et al.  Hypoglossal Neuropathology and Respiratory Activity in Pompe Mice , 2011, Front. Physio..

[15]  B. Byrne,et al.  Sustained alpha‐sarcoglycan gene expression after gene transfer in limb‐girdle muscular dystrophy, type 2D , 2010, Annals of neurology.

[16]  K. Foust,et al.  Over the barrier and through the blood: To CNS delivery we go , 2009, Cell cycle.

[17]  K. Foust,et al.  AAV as a gene transfer vector for the treatment of neurological disorders: novel treatment thoughts for ALS. , 2009, Current gene therapy.

[18]  B. Byrne,et al.  Limb‐girdle muscular dystrophy type 2D gene therapy restores α‐sarcoglycan and associated proteins , 2009, Annals of neurology.

[19]  A. J. Roman,et al.  Human RPE65 gene therapy for Leber congenital amaurosis: persistence of early visual improvements and safety at 1 year. , 2009, Human gene therapy.

[20]  B. Byrne,et al.  Neural deficits contribute to respiratory insufficiency in Pompe disease , 2009, Proceedings of the National Academy of Sciences.

[21]  A. Pestronk,et al.  Clinical features of late‐onset Pompe disease: A prospective cohort study , 2008, Muscle & nerve.

[22]  B. Byrne,et al.  Tissue specific promoters improve specificity of AAV9 mediated transgene expression following intra-vascular gene delivery in neonatal mice , 2008, Genetic vaccines and therapy.

[23]  J. Mendell,et al.  Lack of toxicity of alpha-sarcoglycan overexpression supports clinical gene transfer trial in LGMD2D , 2008, Neurology.

[24]  Eduardo Soriano,et al.  Mechanism suppressing glycogen synthesis in neurons and its demise in progressive myoclonus epilepsy , 2007, Nature Neuroscience.

[25]  Sihong Song,et al.  Phase I trial of intramuscular injection of a recombinant adeno-associated virus serotype 2 alphal-antitrypsin (AAT) vector in AAT-deficient adults. , 2006, Human gene therapy.

[26]  B. Thurberg,et al.  Characterization of pre- and post-treatment pathology after enzyme replacement therapy for pompe disease , 2006, Laboratory Investigation.

[27]  J. Rasko,et al.  Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response , 2006, Nature Medicine.

[28]  Samuel R Ward,et al.  Density and hydration of fresh and fixed human skeletal muscle. , 2005, Journal of biomechanics.

[29]  Shiu-Feng Huang,et al.  Infantile-onset glycogen storage disease type II (Pompe disease): report of a case with genetic diagnosis and pathological findings. , 2004, Chang Gung medical journal.

[30]  B. Bembi,et al.  Intractable fever and cortical neuronal glycogen storage in glycogenosis type 2 , 2001, Neurology.

[31]  Aryeh Routtenberg,et al.  GAP-43: an intrinsic determinant of neuronal development and plasticity , 1997, Trends in Neurosciences.

[32]  L. Rakić,et al.  GAP-43 mRNA expression in early development of human nervous system. , 1996, Brain research. Molecular brain research.

[33]  Bertorini Te,et al.  RESPIRATORY INSUFFICIENCY IN ADULT-TYPE ACID MALTASE DEFICIENCY , 1993 .

[34]  J. Martín,et al.  Pompe's disease: An inborn lysosomal disorder with storage of glycogen , 1973, Acta Neuropathologica.

[35]  S. Dimauro,et al.  NERVOUS SYSTEM IN POMPE'S DISEASE: ULTRASTRUCTURE AND BIOCHEMISTRY , 1971, Journal of neuropathology and experimental neurology.

[36]  R. G. Berry,et al.  POMPE'S DISEASE (DIFFUSE GLYCOGENOSIS) WITH NEURONAL STORAGE. , 1965, Journal of neuropathology and experimental neurology.