1
|
Tarallo A, Damiano C, Strollo S, Minopoli N, Indrieri A, Polishchuk E, Zappa F, Nusco E, Fecarotta S, Porto C, Coletta M, Iacono R, Moracci M, Polishchuk R, Medina DL, Imbimbo P, Monti DM, De Matteis MA, Parenti G. Correction of oxidative stress enhances enzyme replacement therapy in Pompe disease. EMBO Mol Med 2021; 13:e14434. [PMID: 34606154 PMCID: PMC8573602 DOI: 10.15252/emmm.202114434] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pompe disease is a metabolic myopathy due to acid alpha-glucosidase deficiency. In addition to glycogen storage, secondary dysregulation of cellular functions, such as autophagy and oxidative stress, contributes to the disease pathophysiology. We have tested whether oxidative stress impacts on enzyme replacement therapy with recombinant human alpha-glucosidase (rhGAA), currently the standard of care for Pompe disease patients, and whether correction of oxidative stress may be beneficial for rhGAA therapy. We found elevated oxidative stress levels in tissues from the Pompe disease murine model and in patients' cells. In cells, stress levels inversely correlated with the ability of rhGAA to correct the enzymatic deficiency. Antioxidants (N-acetylcysteine, idebenone, resveratrol, edaravone) improved alpha-glucosidase activity in rhGAA-treated cells, enhanced enzyme processing, and improved mannose-6-phosphate receptor localization. When co-administered with rhGAA, antioxidants improved alpha-glucosidase activity in tissues from the Pompe disease mouse model. These results indicate that oxidative stress impacts on the efficacy of enzyme replacement therapy in Pompe disease and that manipulation of secondary abnormalities may represent a strategy to improve the efficacy of therapies for this disorder.
Collapse
Affiliation(s)
- Antonietta Tarallo
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Carla Damiano
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Sandra Strollo
- Telethon Institute of Genetics and MedicinePozzuoliItaly
| | - Nadia Minopoli
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Alessia Indrieri
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Institute for Genetic and Biomedical Research (IRGB)National Research Council (CNR)MilanItaly
| | | | - Francesca Zappa
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Present address:
Department of Molecular, Cellular, and Developmental BiologyUniversity of CaliforniaSanta BarbaraCAUSA
| | - Edoardo Nusco
- Telethon Institute of Genetics and MedicinePozzuoliItaly
| | - Simona Fecarotta
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Caterina Porto
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Marcella Coletta
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
- Present address:
IInd Division of NeurologyMultiple Sclerosis CenterUniversity of Campania "Luigi Vanvitelli"NaplesItaly
| | - Roberta Iacono
- Department of BiologyUniversity of Naples "Federico II", Complesso Universitario di Monte S. AngeloNaplesItaly
- Institute of Biosciences and BioResources ‐ National Research Council of ItalyNaplesItaly
| | - Marco Moracci
- Department of BiologyUniversity of Naples "Federico II", Complesso Universitario di Monte S. AngeloNaplesItaly
- Institute of Biosciences and BioResources ‐ National Research Council of ItalyNaplesItaly
| | | | - Diego Luis Medina
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Paola Imbimbo
- Department of Chemical SciencesFederico II UniversityNaplesItaly
| | | | - Maria Antonietta De Matteis
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Molecular Medicine and Medical BiotechnologiesFederico II UniversityNaplesItaly
| | - Giancarlo Parenti
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| |
Collapse
|
2
|
Abstract
Phenylketonuria (PKU; also known as phenylalanine hydroxylase (PAH) deficiency) is an autosomal recessive disorder of phenylalanine metabolism, in which especially high phenylalanine concentrations cause brain dysfunction. If untreated, this brain dysfunction results in severe intellectual disability, epilepsy and behavioural problems. The prevalence varies worldwide, with an average of about 1:10,000 newborns. Early diagnosis is based on newborn screening, and if treatment is started early and continued, intelligence is within normal limits with, on average, some suboptimal neurocognitive function. Dietary restriction of phenylalanine has been the mainstay of treatment for over 60 years and has been highly successful, although outcomes are still suboptimal and patients can find the treatment difficult to adhere to. Pharmacological treatments are available, such as tetrahydrobiopterin, which is effective in only a minority of patients (usually those with milder PKU), and pegylated phenylalanine ammonia lyase, which requires daily subcutaneous injections and causes adverse immune responses. Given the drawbacks of these approaches, other treatments are in development, such as mRNA and gene therapy. Even though PAH deficiency is the most common defect of amino acid metabolism in humans, brain dysfunction in individuals with PKU is still not well understood and further research is needed to facilitate development of pathophysiology-driven treatments.
Collapse
Affiliation(s)
- Francjan J van Spronsen
- Beatrix Children's Hospital, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands.
| | - Nenad Blau
- University Children's Hospital in Zurich, Zurich, Switzerland
| | - Cary Harding
- Department of Molecular and Medical Genetics and Department of Pediatrics, Oregon Health & Science University, Oregon, USA
| | | | - Nicola Longo
- Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| | - Annet M Bosch
- University of Amsterdam, Department of Pediatrics, Division of Metabolic Disorders, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
3
|
Xu F, Liu X, Zhang D, Zhao F, Fan Z, Hu S, Mei S, Huang Y, Sun H, Wei L, Guo L, Wang J, Cen S, Liang C, Guo F. The Engineered MARCH8-Resistant Vesicular Stomatitis Virus Glycoprotein Enhances Lentiviral Vector Transduction. Hum Gene Ther 2021; 32:936-948. [PMID: 33678011 DOI: 10.1089/hum.2020.292] [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] [Indexed: 11/12/2022] Open
Abstract
Lentiviral vectors are one of the most commonly used viral delivery systems for gene therapy. Vesicular stomatitis virus-G envelope glycoprotein (VSV G)-pseudotyped lentiviral vectors have been widely used in clinical studies for treatment of virus infections and genetic deficient diseases. However, the efficiency of lentiviral vector transduction has been long recognized as a limiting factor in clinical gene therapy application, especially in transducing hematopoietic stem cells. MARCH8 (membrane-associated RING-CH 8), an E3 ubiquitin ligase, has been reported to target and downregulate VSV G. Results in this study show that MARCH8 induces ubiquitination and lysosome degradation of VSV G, and knockout of MARCH8 in virus-producing cells increases lentiviral vector transduction by elevating the level of VSV G protein. We then engineered VSV G mutant that has the lysine residues in the cytoplasmic domain substituted for arginine, and showed that this G mutant resists degradation by MARCH8, and allows the enhancement of transduction efficiency of lentiviral vector particles than the parental VSV G protein. This engineered VSV G mutant thus further advances the lentiviral vector system as a powerful tool in gene therapy.
Collapse
Affiliation(s)
- Fengwen Xu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Xiaoman Liu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Di Zhang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Fei Zhao
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Zhangling Fan
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Siqi Hu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Shan Mei
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Yu Huang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Hong Sun
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Liang Wei
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Li Guo
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Jianwei Wang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Shan Cen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Chen Liang
- McGill University AIDS Centre, Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada
| | - Fei Guo
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for AIDS Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| |
Collapse
|
4
|
Evaluation of HIV-1 derived lentiviral vectors as transductors of Mucopolysaccharidosis type IV a fibroblasts. Gene 2021; 780:145527. [PMID: 33636292 DOI: 10.1016/j.gene.2021.145527] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 01/15/2021] [Accepted: 02/09/2021] [Indexed: 11/23/2022]
Abstract
Mucopolysaccharidosis type IVA (MPS IVA) is a lysosomal storage disease produced by the deficiency of the N-acetylgalactosamine-6-sulfate sulfatase (GALNS) enzyme, leading to glycosaminoglycans (GAGs) accumulation. Since currently available treatments remain limited and unspecific, novel therapeutic approaches are essential for the disease treatment. In an attempt to reduce treatment limitations, gene therapy rises as a more effective and specific alternative. We present in this study the delivery assessment of GALNS and sulfatase-modifying factor 1 (SUMF1) genes via HIV-1 derived lentiviral vectors into fibroblasts from MPS IVA patients. After transduction, we determined GALNS enzymatic activity, lysosomal mass change, and autophagy pathway impairment. Additionally, we computationally assessed the effect of mutations over the enzyme-substrate interaction and phenotypic effects. The results showed that the co-transduction of MPS IVA fibroblasts with GALNS and SUMF1 cDNAs led to a significant increase in GALNS enzyme activity and a reduction of lysosomal mass. We show that patient-specific differences in cellular response are directly associated with the set of mutations on each patient. Lastly, we present new evidence supporting autophagy impairment in MPS IVA due to the presence and changes in autophagy proteins in treated MPS IVA fibroblasts. Our results offer new evidence that demonstrate the potential of lentiviral vectors as a strategy to correct GALNS deficiency.
Collapse
|
5
|
Abstract
Pompe disease (PD) is caused by the deficiency of the lysosomal enzyme acid α-glucosidase (GAA), resulting in systemic pathological glycogen accumulation. PD can present with cardiac, skeletal muscle, and central nervous system manifestations, as a continuum of phenotypes among two main forms: classical infantile-onset PD (IOPD) and late-onset PD (LOPD). IOPD is caused by severe GAA deficiency and presents at birth with cardiac hypertrophy, muscle hypotonia, and severe respiratory impairment, leading to premature death, if not treated. LOPD is characterized by levels of residual GAA activity up to ∼20% of normal and presents both in children and adults with a varied severity of muscle weakness and motor and respiratory deficit. Enzyme replacement therapy (ERT), based on repeated intravenous (i.v.) infusions of recombinant human GAA (rhGAA), represents the only available treatment for PD. Upon more than 10 years from its launch, it is becoming evident that ERT can extend the life span of IOPD and stabilize disease progression in LOPD; however, it does not represent a cure for PD. The limited uptake of the enzyme in key affected tissues and the high immunogenicity of rhGAA are some of the hurdles that limit ERT efficacy. GAA gene transfer with adeno-associated virus (AAV) vectors has been shown to reduce glycogen storage and improve the PD phenotype in preclinical studies following different approaches. Here, we present an overview of the different gene therapy approaches for PD, focusing on in vivo gene transfer with AAV vectors and discussing the potential opportunities and challenges in developing safe and effective gene therapies for the disease. Based on emerging safety and efficacy data from clinical trials for other protein deficiencies, in vivo gene therapy with AAV vectors appears to have the potential to provide a therapeutically relevant, stable source of GAA enzyme, which could be highly beneficial in PD.
Collapse
Affiliation(s)
- Pasqualina Colella
- Genethon, Evry, France.,Department of Pediatrics, Stanford University, Stanford, California
| | - Federico Mingozzi
- Genethon, Evry, France.,Spark Therapeutics, Philadelphia, Pennsylvania
| |
Collapse
|
6
|
Yadak R, Cabrera-Pérez R, Torres-Torronteras J, Bugiani M, Haeck JC, Huston MW, Bogaerts E, Goffart S, Jacobs EH, Stok M, Leonardelli L, Biasco L, Verdijk RM, Bernsen MR, Ruijter G, Martí R, Wagemaker G, van Til NP, de Coo IF. Preclinical Efficacy and Safety Evaluation of Hematopoietic Stem Cell Gene Therapy in a Mouse Model of MNGIE. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 8:152-165. [PMID: 29687034 PMCID: PMC5908387 DOI: 10.1016/j.omtm.2018.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/02/2018] [Indexed: 12/15/2022]
Abstract
Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is an autosomal recessive disorder caused by thymidine phosphorylase (TP) deficiency resulting in systemic accumulation of thymidine (d-Thd) and deoxyuridine (d-Urd) and characterized by early-onset neurological and gastrointestinal symptoms. Long-term effective and safe treatment is not available. Allogeneic bone marrow transplantation may improve clinical manifestations but carries disease and transplant-related risks. In this study, lentiviral vector-based hematopoietic stem cell gene therapy (HSCGT) was performed in Tymp−/−Upp1−/− mice with the human phosphoglycerate kinase (PGK) promoter driving TYMP. Supranormal blood TP activity reduced intestinal nucleoside levels significantly at low vector copy number (median, 1.3; range, 0.2–3.6). Furthermore, we covered two major issues not addressed before. First, we demonstrate aberrant morphology of brain astrocytes in areas of spongy degeneration, which was reversed by HSCGT. Second, long-term follow-up and vector integration site analysis were performed to assess safety of the therapeutic LV vectors in depth. This report confirms and supplements previous work on the efficacy of HSCGT in reducing the toxic metabolites in Tymp−/−Upp1−/− mice, using a clinically applicable gene transfer vector and a highly efficient gene transfer method, and importantly demonstrates phenotypic correction with a favorable risk profile, warranting further development toward clinical implementation.
Collapse
Affiliation(s)
- Rana Yadak
- Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Raquel Cabrera-Pérez
- Research Group on Neuromuscular and Mitochondrial Diseases, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, and Biomedical Network Research Centre on Rare Diseases (CIBERER), Barcelona, Catalonia, Spain
| | - Javier Torres-Torronteras
- Research Group on Neuromuscular and Mitochondrial Diseases, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, and Biomedical Network Research Centre on Rare Diseases (CIBERER), Barcelona, Catalonia, Spain
| | - Marianna Bugiani
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Joost C. Haeck
- Department of Radiology & Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marshall W. Huston
- Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Elly Bogaerts
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Steffi Goffart
- Department of Biology, University of Eastern Finland, Joensuu, Finland
| | - Edwin H. Jacobs
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Merel Stok
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Lorena Leonardelli
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Milan, Italy
| | - Luca Biasco
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Milan, Italy
- Gene Therapy Program, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA, USA
- University College of London (UCL), Great Ormond Street Institute of Child Health (ICH), London, UK
| | - Robert M. Verdijk
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Monique R. Bernsen
- Department of Radiology & Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - George Ruijter
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ramon Martí
- Research Group on Neuromuscular and Mitochondrial Diseases, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, and Biomedical Network Research Centre on Rare Diseases (CIBERER), Barcelona, Catalonia, Spain
| | - Gerard Wagemaker
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Hacettepe University, Stem Cell Research and Development Center, Ankara, Turkey
- Raisa Gorbacheva Memorial Research Institute for Pediatric Oncology and Hematology, Saint Petersburg, Russia
| | - Niek P. van Til
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Irenaeus F.M. de Coo
- Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Corresponding author: Irenaeus F.M. de Coo, Department of Neurology, Erasmus University Medical Center, PO Box 2060, 3000 CB Rotterdam, the Netherlands.
| |
Collapse
|
7
|
CRYSTAL RONALDG, PAGOVICH ODELYAE. THE JEREMIAH METZGER LECTURE NOVEL THERAPEUTIC STRATEGIES OF ALLERGIC AND IMMUNOLOGIC DISORDERS. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2018; 129:250-265. [PMID: 30166721 PMCID: PMC6116601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Advances in understanding the immunological basis and mechanisms underlying allergic and immunologic disorders have led to effective but costly long-term and repetitive biologic therapies. Gene therapy is a rapidly advancing technology, in which a single administration of an adeno-associated virus encoding the therapeutic protein or monoclonal antibody may provide effective long-term therapy for allergic and immunologic disorders. In this review, we summarize the recent studies from our laboratory developing gene therapy strategies to treat hereditary angioedema and peanut allergy. The unraveling of the pathogenesis of immune-based disorders, including hereditary deficiencies of components of the immune system and allergic disorders, has led to the development of therapies using parenteral administration of recombinant proteins or monoclonal antibodies (1). While many of these therapies are highly effective, they are limited by the half-life of the therapeutic protein or antibody, requiring repetitive administration of days to weeks (2-15). The focus of recent work in our laboratory has been to solve this problem by substituting protein/monoclonal antibody administration with gene therapy, where current technology allows for a single administration of the gene coding for a protein or antibody to provide persistent expression of effective levels of the therapeutic protein or antibody. Gene therapy is a drug delivery platform which uses genetic material, usually in the form of coding exons of the therapeutic gene, to correct, compensate for, or prevent the development of an abnormal phenotype (16). Originally conceptualized as a strategy to treat rare hereditary disorders, gene therapy is being developed for a wide range of human disorders, including common acquired conditions (17-20). In this review, we will describe how we have adopted gene therapy technology to develop therapies for immune-related disorders, using as examples hereditary angioedema, an inherited autosomal dominant disorder, and peanut allergy, a common acquired allergic disorder.
Collapse
|
8
|
Yadak R, Sillevis Smitt P, van Gisbergen MW, van Til NP, de Coo IFM. Mitochondrial Neurogastrointestinal Encephalomyopathy Caused by Thymidine Phosphorylase Enzyme Deficiency: From Pathogenesis to Emerging Therapeutic Options. Front Cell Neurosci 2017; 11:31. [PMID: 28261062 PMCID: PMC5309216 DOI: 10.3389/fncel.2017.00031] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 02/01/2017] [Indexed: 01/05/2023] Open
Abstract
Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is a progressive metabolic disorder caused by thymidine phosphorylase (TP) enzyme deficiency. The lack of TP results in systemic accumulation of deoxyribonucleosides thymidine (dThd) and deoxyuridine (dUrd). In these patients, clinical features include mental regression, ophthalmoplegia, and fatal gastrointestinal complications. The accumulation of nucleosides also causes imbalances in mitochondrial DNA (mtDNA) deoxyribonucleoside triphosphates (dNTPs), which may play a direct or indirect role in the mtDNA depletion/deletion abnormalities, although the exact underlying mechanism remains unknown. The available therapeutic approaches include dialysis and enzyme replacement therapy, both can only transiently reverse the biochemical imbalance. Allogeneic hematopoietic stem cell transplantation is shown to be able to restore normal enzyme activity and improve clinical manifestations in MNGIE patients. However, transplant related complications and disease progression result in a high mortality rate. New therapeutic approaches, such as adeno-associated viral vector and hematopoietic stem cell gene therapy have been tested in Tymp-/-Upp1-/- mice, a murine model for MNGIE. This review provides background information on disease manifestations of MNGIE with a focus on current management and treatment options. It also outlines the pre-clinical approaches toward future treatment of the disease.
Collapse
Affiliation(s)
- Rana Yadak
- Department of Neurology, Erasmus University Medical Center Rotterdam, Netherlands
| | - Peter Sillevis Smitt
- Department of Neurology, Erasmus University Medical Center Rotterdam, Netherlands
| | - Marike W van Gisbergen
- Department of Radiation Oncology (MaastRO-Lab), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre Maastricht, Netherlands
| | - Niek P van Til
- Laboratory of Translational Immunology, University Medical Center Utrecht Utrecht, Netherlands
| | - Irenaeus F M de Coo
- Department of Neurology, Erasmus University Medical Center Rotterdam, Netherlands
| |
Collapse
|
9
|
Doerfler PA, Nayak S, Corti M, Morel L, Herzog RW, Byrne BJ. Targeted approaches to induce immune tolerance for Pompe disease therapy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:15053. [PMID: 26858964 PMCID: PMC4729315 DOI: 10.1038/mtm.2015.53] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/04/2015] [Accepted: 11/28/2015] [Indexed: 12/31/2022]
Abstract
Enzyme and gene replacement strategies have developed into viable therapeutic approaches for the treatment of Pompe disease (acid α-glucosidase (GAA) deficiency). Unfortunately, the introduction of GAA and viral vectors encoding the enzyme can lead to detrimental immune responses that attenuate treatment benefits and can impact patient safety. Preclinical and clinical experience in addressing humoral responses toward enzyme and gene therapy for Pompe disease have provided greater understanding of the immunological consequences of the provided therapy. B- and T-cell modulation has been shown to be effective in preventing infusion-associated reactions during enzyme replacement therapy in patients and has shown similar success in the context of gene therapy. Additional techniques to induce humoral tolerance for Pompe disease have been the targeted expression or delivery of GAA to discrete cell types or tissues such as the gut-associated lymphoid tissues, red blood cells, hematopoietic stem cells, and the liver. Research into overcoming preexisting immunity through immunomodulation and gene transfer are becoming increasingly important to achieve long-term efficacy. This review highlights the advances in therapies as well as the improved understanding of the molecular mechanisms involved in the humoral immune response with emphasis on methods employed to overcome responses associated with enzyme and gene therapies for Pompe disease.
Collapse
Affiliation(s)
- Phillip A Doerfler
- Department of Pediatrics, University of Florida , Gainesville, Florida, USA
| | - Sushrusha Nayak
- Department of Medicine, Karolinska Institute , Stockholm, Sweden
| | - Manuela Corti
- Department of Pediatrics, University of Florida , Gainesville, Florida, USA
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida , Gainesville, Florida, USA
| | - Roland W Herzog
- Department of Pediatrics, University of Florida , Gainesville, Florida, USA
| | - Barry J Byrne
- Department of Pediatrics, University of Florida , Gainesville, Florida, USA
| |
Collapse
|
10
|
Abstract
Gene transfer vectors based on retroviridae are increasingly becoming a tool of choice for biomedical research and for the development of biotherapies in rare diseases or cancers. To meet the challenges of preclinical and clinical production, different steps of the production process of self-inactivating γ-retroviral (RVs) and lentiviral vectors (LVs) have been improved (e.g., transfection, media optimization, cell culture conditions). However, the increasing need for mass production of such vectors is still a challenge and could hamper their availability for therapeutic use. Recently, we observed that the use of a neutral pH during vector production is not optimal. The use of mildly acidic pH conditions (pH 6) can increase by two- to threefold the production of RVs and LVs pseudotyped with the vesicular stomatitis virus G (VSV-G) or gibbon ape leukemia virus (GALV) glycoproteins. Here, we describe the production protocol in mildly acidic pH conditions of GALVTR- and VSV-G-pseudotyped LVs using the transient transfection of HEK293T cells and the production protocol of GALV-pseudotyped RVs produced from a murine producer cell line. These protocols should help to achieve higher titers of vectors, thereby facilitating experimental research and therapeutic applications.
Collapse
Affiliation(s)
- Nathalie Holic
- Généthon, 91002, Evry, France.
- INSERM, UMR_S951, Généthon, 1bis, rue de l'Internationale-BP60, 91002, Evry, France.
- Université Evry Val d'Essonne, UMR_S951, 91002, Evry, France.
| | - David Fenard
- Généthon, 91002, Evry, France.
- INSERM, UMR_S951, Généthon, 1bis, rue de l'Internationale-BP60, 91002, Evry, France.
- Université Evry Val d'Essonne, UMR_S951, 91002, Evry, France.
| |
Collapse
|
11
|
Hu S, Mohan Kumar D, Sax C, Schuler C, Akkina R. Pseudotyping of lentiviral vector with novel vesiculovirus envelope glycoproteins derived from Chandipura and Piry viruses. Virology 2015; 488:162-8. [PMID: 26650691 DOI: 10.1016/j.virol.2015.11.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 11/13/2015] [Accepted: 11/14/2015] [Indexed: 12/22/2022]
Abstract
While the envelope glycoprotein of vesicular stomatitis virus (VSV-G) is widely used for pseudotyping of lentiviral vectors, sub-optimal gene transfer into certain cell types and its sensitivity to inactivation by human complement hinders its broader applications. To find alternative candidates, here we evaluated two serologically distinct novel viral envelopes derived from Chandipura (CNV-G) and Piry (PRV-G) vesiculoviruses. Both permitted generation of high titer psuedotyped lentiviral vectors with a capacity for high efficiency gene transfer into various cell types from different species. In human lymphoid and hematopoietic stem cells, their transduction efficiency was significantly lower than that of VSV-G. However, both novel envelopes were found to be more resistant to inactivation by human serum complement compared to VSV-G. Thus CNV-G and PRV-G envelopes can be harnessed for multiple uses in the future based on the cell type that needs to be gene transduced and possibly for in vivo gene transfer.
Collapse
Affiliation(s)
- Shuang Hu
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Dipu Mohan Kumar
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Chelsea Sax
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Clayton Schuler
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Ramesh Akkina
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
12
|
Chu Y, Oum YH, Carrico IS. Surface modification via strain-promoted click reaction facilitates targeted lentiviral transduction. Virology 2015; 487:95-103. [PMID: 26499046 DOI: 10.1016/j.virol.2015.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/19/2015] [Accepted: 09/21/2015] [Indexed: 11/29/2022]
Abstract
As a result of their ability to integrate into the genome of both dividing and non-dividing cells, lentiviruses have emerged as a promising vector for gene delivery. Targeted gene transduction of specific cells and tissues by lentiviral vectors has been a major goal, which has proven difficult to achieve. We report a novel targeting protocol that relies on the chemoselective attachment of cancer specific ligands to unnatural glycans on lentiviral surfaces. This strategy exhibits minimal perturbation on virus physiology and demonstrates remarkable flexibility. It allows for targeting but can be more broadly useful with applications such as vector purification and immunomodulation.
Collapse
Affiliation(s)
- Yanjie Chu
- Department of Chemistry, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA
| | - Yoon Hyeun Oum
- Department of Chemistry, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA
| | - Isaac S Carrico
- Department of Chemistry, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA; Institute of Chemical Biology and Drug Discovery, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA.
| |
Collapse
|
13
|
Wakabayashi T, Shimada Y, Akiyama K, Higuchi T, Fukuda T, Kobayashi H, Eto Y, Ida H, Ohashi T. Hematopoietic Stem Cell Gene Therapy Corrects Neuropathic Phenotype in Murine Model of Mucopolysaccharidosis Type II. Hum Gene Ther 2015; 26:357-66. [PMID: 25761450 DOI: 10.1089/hum.2014.158] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mucopolysaccharidosis type II (MPS II) is a neuropathic lysosomal storage disorder caused by a deficiency of iduronate-2-sulfatase (IDS), which leads to the accumulation of glycosaminoglycans (GAGs). We demonstrated that biochemical alterations in the brains of MPS II mice are not corrected by bone marrow transplantation (BMT) or enzyme replacement therapy, although BMT has been shown to be effective for other neurodegenerative MPSs, such as Hurler syndrome. In this study, we demonstrated that lentiviral isogeneic hematopoietic stem cell (HSC) gene therapy corrected neuronal manifestations by ameliorating lysosomal storage and autophagic dysfunction in the brains of MPS II mice. IDS-transduced HSCs increased enzyme activity both in various visceral organs and the CNS. Decreased levels of GAGs were observed in many organs, including cerebra, after transplantation of IDS-transduced HSCs. In addition, lentiviral HSC gene therapy normalized the secondary accumulation of autophagic substrates, such as p62 and ubiquitin-protein conjugates, in cerebra. Furthermore, in contrast to naive MPS II mice, there was no deterioration of neuronal function observed in transplant recipients. These results indicated that lentiviral HSC gene therapy is a promising approach for the treatment of CNS lesions in MPS II.
Collapse
Affiliation(s)
- Taichi Wakabayashi
- 1 Department of Pediatrics, Jikei University School of Medicine , Tokyo 105-8461, Japan .,2 Division of Gene Therapy, Research Center for Medical Sciences, Jikei University School of Medicine , Tokyo 105-8461, Japan
| | - Yohta Shimada
- 2 Division of Gene Therapy, Research Center for Medical Sciences, Jikei University School of Medicine , Tokyo 105-8461, Japan
| | - Kazumasa Akiyama
- 3 Department of Pediatrics, Kitasato University School of Medicine , Kanagawa 252-0374, Japan
| | - Takashi Higuchi
- 2 Division of Gene Therapy, Research Center for Medical Sciences, Jikei University School of Medicine , Tokyo 105-8461, Japan
| | - Takahiro Fukuda
- 4 Division of Neuropathology, Department of Pathology, Jikei University School of Medicine , Tokyo 105-8461, Japan
| | - Hiroshi Kobayashi
- 1 Department of Pediatrics, Jikei University School of Medicine , Tokyo 105-8461, Japan .,2 Division of Gene Therapy, Research Center for Medical Sciences, Jikei University School of Medicine , Tokyo 105-8461, Japan
| | - Yoshikatsu Eto
- 5 Advanced Clinical Research Center, Institute of Neurological Disorders , Kanagawa 215-0026, Japan
| | - Hiroyuki Ida
- 1 Department of Pediatrics, Jikei University School of Medicine , Tokyo 105-8461, Japan .,2 Division of Gene Therapy, Research Center for Medical Sciences, Jikei University School of Medicine , Tokyo 105-8461, Japan
| | - Toya Ohashi
- 1 Department of Pediatrics, Jikei University School of Medicine , Tokyo 105-8461, Japan .,2 Division of Gene Therapy, Research Center for Medical Sciences, Jikei University School of Medicine , Tokyo 105-8461, Japan
| |
Collapse
|