1
|
İnci A, Ezgü FS, Tümer L. Advances in Immune Tolerance Induction in Enzyme Replacement Therapy. Paediatr Drugs 2024; 26:287-308. [PMID: 38664313 PMCID: PMC11074017 DOI: 10.1007/s40272-024-00627-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2024] [Indexed: 05/07/2024]
Abstract
Inborn errors of metabolism (IEMs) are a group of genetic diseases that occur due to the either deficiency of an enzyme involved in a metabolic/biochemical pathway or other disturbances in the metabolic pathway including transport protein or activator protein deficiencies, cofactor deficiencies, organelle biogenesis, maturation or trafficking problems. These disorders are collectively significant due to their substantial impact on both the well-being and survival of affected individuals. In the quest for effective treatments, enzyme replacement therapy (ERT) has emerged as a viable strategy for patients with many of the lysosomal storage disorders (LSD) and enzyme substitution therapy in the rare form of the other inborn errors of metabolism including phenylketonuria and hypophosphatasia. However, a major challenge associated with enzyme infusion in patients with these disorders, mainly LSD, is the development of high antibody titres. Strategies focusing on immunomodulation have shown promise in inducing immune tolerance to ERT, leading to improved overall survival rates. The implementation of immunomodulation concurrent with ERT administration has also resulted in a decreased occurrence of IgG antibody development compared with cases treated solely with ERT. By incorporating the knowledge gained from current approaches and analysing the outcomes of immune tolerance induction (ITI) modalities from clinical and preclinical trials have demonstrated significant improvement in the efficacy of ERT. In this comprehensive review, the progress in ITI modalities is assessed, drawing insights from both clinical and preclinical trials. The focus is on evaluating the advancements in ITI within the context of IEM, specifically addressing LSDs managed through ERT.
Collapse
Affiliation(s)
- Aslı İnci
- Department of Paediatric Metabolism and Nutrition, Gazi University School of Medicine, Emniyet Street, Yenimahalle, Ankara, Turkey.
| | - Fatih Süheyl Ezgü
- Department of Paediatric Metabolism and Nutrition, Gazi University School of Medicine, Emniyet Street, Yenimahalle, Ankara, Turkey
- Department of Paediatric Genetic, Gazi University School of Medicine, Ankara, Turkey
| | - Leyla Tümer
- Department of Paediatric Metabolism and Nutrition, Gazi University School of Medicine, Emniyet Street, Yenimahalle, Ankara, Turkey
| |
Collapse
|
2
|
Watanabe R, Tsuji D, Tanaka H, Uno MS, Ohnishi Y, Kitaguchi S, Matsugu T, Nakae R, Teramoto H, Yamamoto K, Shinohara Y, Hirokawa T, Okino N, Ito M, Itoh K. Lysoglycosphingolipids have the ability to induce cell death through direct PI3K inhibition. J Neurochem 2023; 167:753-765. [PMID: 37975558 DOI: 10.1111/jnc.16012] [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] [Received: 06/16/2022] [Revised: 08/04/2023] [Accepted: 10/31/2023] [Indexed: 11/19/2023]
Abstract
Sphingolipidoses are inherited metabolic disorders associated with glycosphingolipids accumulation, neurodegeneration, and neuroinflammation leading to severe neurological symptoms. Lysoglycosphingolipids (lysoGSLs), also known to accumulate in the tissues of sphingolipidosis patients, exhibit cytotoxicity. LysoGSLs are the possible pathogenic cause, but the mechanisms are still unknown in detail. Here, we first show that lysoGSLs are potential inhibitors of phosphoinositide 3-kinase (PI3K) to reduce cell survival signaling. We found that phosphorylated Akt was commonly reduced in fibroblasts from patients with sphingolipidoses, including GM1/GM2 gangliosidoses and Gaucher's disease, suggesting the contribution of lysoGSLs to the pathogenesis. LysoGSLs caused cell death and decreased the level of phosphorylated Akt as in the patient fibroblasts. Extracellularly administered lysoGM1 permeated the cell membrane to diffusely distribute in the cytoplasm. LysoGM1 and lysoGM2 also inhibited the production of phosphatidylinositol-(3,4,5)-triphosphate and the translocation of Akt from the cytoplasm to the plasma membrane. We also predicted that lysoGSLs could directly bind to the catalytic domain of PI3K by in silico docking study, suggesting that lysoGSLs could inhibit PI3K by directly interacting with PI3K in the cytoplasm. Furthermore, we revealed that the increment of lysoGSLs amounts in the brain of sphingolipidosis model mice correlated with the neurodegenerative progression. Our findings suggest that the down-regulation of PI3K/Akt signaling by direct interaction of lysoGSLs with PI3K in the brains is a neurodegenerative mechanism in sphingolipidoses. Moreover, we could propose the intracellular PI3K activation or inhibition of lysoGSLs biosynthesis as novel therapeutic approaches for sphingolipidoses because lysoGSLs should be cell death mediators by directly inhibiting PI3K, especially in neurons.
Collapse
Affiliation(s)
- Ryosuke Watanabe
- Department of Medicinal Biotechnology, Graduate school of Pharmaceutical Science, Tokushima University, Tokushima, Japan
| | - Daisuke Tsuji
- Department of Medicinal Biotechnology, Graduate school of Pharmaceutical Science, Tokushima University, Tokushima, Japan
- Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan
| | - Hiroki Tanaka
- Department of Medicinal Biotechnology, Graduate school of Pharmaceutical Science, Tokushima University, Tokushima, Japan
| | - Michael Shintaro Uno
- Department of Medicinal Biotechnology, Graduate school of Pharmaceutical Science, Tokushima University, Tokushima, Japan
| | - Yukiya Ohnishi
- Department of Medicinal Biotechnology, Graduate school of Pharmaceutical Science, Tokushima University, Tokushima, Japan
| | - Shindai Kitaguchi
- Department of Medicinal Biotechnology, Graduate school of Pharmaceutical Science, Tokushima University, Tokushima, Japan
| | - Tsuyoshi Matsugu
- Department of Medicinal Biotechnology, Graduate school of Pharmaceutical Science, Tokushima University, Tokushima, Japan
| | - Ryuto Nakae
- Department of Medicinal Biotechnology, Graduate school of Pharmaceutical Science, Tokushima University, Tokushima, Japan
| | - Hiromi Teramoto
- Department of Medicinal Biotechnology, Graduate school of Pharmaceutical Science, Tokushima University, Tokushima, Japan
| | - Kei Yamamoto
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Yasuo Shinohara
- Institute for Genome Research, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
- Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan
| | - Takatsugu Hirokawa
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Nozomu Okino
- Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Makoto Ito
- Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Kohji Itoh
- Department of Medicinal Biotechnology, Graduate school of Pharmaceutical Science, Tokushima University, Tokushima, Japan
| |
Collapse
|
3
|
Tucci F, Consiglieri G, Cossutta M, Bernardo ME. Current and Future Perspective in Hematopoietic Stem Progenitor Cell-gene Therapy for Inborn Errors of Metabolism. Hemasphere 2023; 7:e953. [PMID: 37711990 PMCID: PMC10499111 DOI: 10.1097/hs9.0000000000000953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 09/16/2023] Open
Affiliation(s)
- Francesca Tucci
- Pediatric Immunohematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan, Italy
| | - Giulia Consiglieri
- Pediatric Immunohematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan, Italy
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Italy
| | - Matilde Cossutta
- Pediatric Immunohematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
- University of Rome Tor Vergata, Italy
| | - Maria Ester Bernardo
- Pediatric Immunohematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan, Italy
- “Vita-Salute” San Raffaele University, Milan, Italy
| |
Collapse
|
4
|
Ditters IAM, van Kooten HA, van der Beek NAME, Hardon JF, Ismailova G, Brusse E, Kruijshaar ME, van der Ploeg AT, van den Hout JMP, Huidekoper HH. Home-Based Infusion of Alglucosidase Alfa Can Safely be Implemented in Adults with Late-Onset Pompe Disease: Lessons Learned from 18,380 Infusions. BioDrugs 2023; 37:685-698. [PMID: 37326923 PMCID: PMC10432339 DOI: 10.1007/s40259-023-00609-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Enzyme replacement therapy (ERT) with alglucosidase alfa is the treatment for patients with Pompe disease, a hereditary metabolic myopathy. Home-based ERT is unavailable in many countries because of the boxed warning alglucosidase alfa received due to the risk of infusion-associated reactions (IARs). Since 2008, home infusions have been provided in The Netherlands. OBJECTIVES This study aimed to provide an overview of our experience with home-based infusions with alglucosidase alfa in adult Pompe patients, focusing on safety, including management of IARs. METHOD We analysed infusion data and IARs from adult patients starting ERT between 1999 and 2018. ERT was initially given in the hospital during the first year. Patients were eligible for home treatment if they were without IARs for multiple consecutive infusions and if a trained home nurse, with on-call back-up by a doctor, was available. The healthcare providers graded IARs. RESULTS We analysed data on 18,380 infusions with alglucosidase alfa in 121 adult patients; 4961 infusions (27.0%) were given in hospital and 13,419 (73.0%) were given at home. IARs occurred in 144 (2.9%) hospital infusions and 113 (0.8%) home infusions; 115 (79.9% of 144) IARs in hospital and 104 (92.0% of 113) IARs at home were mild, 25 IARs (17.4%) in hospital and 8 IARs (7.1%) at home were moderate, and very few severe IARs occurred (4 IARs in hospital [2.8%] and 1 IAR at home [0.9%]). Only one IAR in the home situation required immediate clinical evaluation in the hospital. CONCLUSION Given the small numbers of IARs that occurred with the home infusions, of which only one was severe, we conclude that alglucosidase alfa can be administered safely in the home situation, provided the appropriate infrastructure is present.
Collapse
Affiliation(s)
- Imke A M Ditters
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Sophia Children's Hospital, PO Box 2060, 3000 CB, Rotterdam, The Netherlands
| | - Harmke A van Kooten
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Nadine A M E van der Beek
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Jacqueline F Hardon
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Sophia Children's Hospital, PO Box 2060, 3000 CB, Rotterdam, The Netherlands
| | - Gamida Ismailova
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Esther Brusse
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Michelle E Kruijshaar
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Sophia Children's Hospital, PO Box 2060, 3000 CB, Rotterdam, The Netherlands
| | - Ans T van der Ploeg
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Sophia Children's Hospital, PO Box 2060, 3000 CB, Rotterdam, The Netherlands.
| | - Johanna M P van den Hout
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Sophia Children's Hospital, PO Box 2060, 3000 CB, Rotterdam, The Netherlands
| | - Hidde H Huidekoper
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Sophia Children's Hospital, PO Box 2060, 3000 CB, Rotterdam, The Netherlands
| |
Collapse
|
5
|
Lopes N, Maia ML, Pereira CS, Mondragão-Rodrigues I, Martins E, Ribeiro R, Gaspar A, Aguiar P, Garcia P, Cardoso MT, Rodrigues E, Leão-Teles E, Giugliani R, Coutinho MF, Alves S, Macedo MF. Leukocyte Imbalances in Mucopolysaccharidoses Patients. Biomedicines 2023; 11:1699. [PMID: 37371793 DOI: 10.3390/biomedicines11061699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Mucopolysaccharidoses (MPSs) are rare inherited lysosomal storage diseases (LSDs) caused by deficient activity in one of the enzymes responsible for glycosaminoglycans lysosomal degradation. MPS II is caused by pathogenic mutations in the IDS gene, leading to deficient activity of the enzyme iduronate-2-sulfatase, which causes dermatan and heparan sulfate storage in the lysosomes. In MPS VI, there is dermatan sulfate lysosomal accumulation due to pathogenic mutations in the ARSB gene, leading to arylsulfatase B deficiency. Alterations in the immune system of MPS mouse models have already been described, but data concerning MPSs patients is still scarce. Herein, we study different leukocyte populations in MPS II and VI disease patients. MPS VI, but not MPS II patients, have a decrease percentage of natural killer (NK) cells and monocytes when compared with controls. No alterations were identified in the percentage of T, invariant NKT, and B cells in both groups of MPS disease patients. However, we discovered alterations in the naïve versus memory status of both helper and cytotoxic T cells in MPS VI disease patients compared to control group. Indeed, MPS VI disease patients have a higher frequency of naïve T cells and, consequently, lower memory T cell frequency than control subjects. Altogether, these results reveal MPS VI disease-specific alterations in some leukocyte populations, suggesting that the type of substrate accumulated and/or enzyme deficiency in the lysosome may have a particular effect on the normal cellular composition of the immune system.
Collapse
Affiliation(s)
- Nuno Lopes
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal
| | - Maria L Maia
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal
| | - Cátia S Pereira
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal
- Cell Activation & Gene Expression (CAGE), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Inês Mondragão-Rodrigues
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal
- Cell Activation & Gene Expression (CAGE), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Departamento de Ciências Médicas, Universidade de Aveiro, Campus Universitário de Santiago, Agra do Crasto, Edifício 30, 3810-193 Aveiro, Portugal
| | - Esmeralda Martins
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar Universitário de Santo António, 4099-001 Porto, Portugal
| | - Rosa Ribeiro
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar Universitário de Santo António, 4099-001 Porto, Portugal
| | - Ana Gaspar
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar e Universitário Lisboa Norte (CHULN), 1649-035 Lisbon, Portugal
| | - Patrício Aguiar
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar e Universitário Lisboa Norte (CHULN), 1649-035 Lisbon, Portugal
- Faculdade de Medicina da Universidade de Lisboa, Universidade de Lisboa, 1649-190 Lisbon, Portugal
| | - Paula Garcia
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar e Universitário de Coimbra, Centro de Desenvolvimento da Criança, 3000-075 Coimbra, Portugal
| | - Maria Teresa Cardoso
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar Universitário de São João (CHUSJ), 4200-319 Porto, Portugal
| | - Esmeralda Rodrigues
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar Universitário de São João (CHUSJ), 4200-319 Porto, Portugal
| | - Elisa Leão-Teles
- Centro de Referência de Doenças Hereditárias do Metabolismo (DHM), Centro Hospitalar Universitário de São João (CHUSJ), 4200-319 Porto, Portugal
| | - Roberto Giugliani
- Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, DASA e Casa dos Raros, Porto Alegre 90610-150, Brazil
| | - Maria F Coutinho
- Research and Development Unit, Department of Genetics, INSA, 4000-055 Porto, Portugal
| | - Sandra Alves
- Research and Development Unit, Department of Genetics, INSA, 4000-055 Porto, Portugal
| | - M Fátima Macedo
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal
- Cell Activation & Gene Expression (CAGE), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Departamento de Ciências Médicas, Universidade de Aveiro, Campus Universitário de Santiago, Agra do Crasto, Edifício 30, 3810-193 Aveiro, Portugal
| |
Collapse
|
6
|
Durán A, Priestman DA, Las Heras M, Rebolledo-Jaramillo B, Olguín V, Calderón JF, Zanlungo S, Gutiérrez J, Platt FM, Klein AD. A Mouse Systems Genetics Approach Reveals Common and Uncommon Genetic Modifiers of Hepatic Lysosomal Enzyme Activities and Glycosphingolipids. Int J Mol Sci 2023; 24:4915. [PMID: 36902345 PMCID: PMC10002577 DOI: 10.3390/ijms24054915] [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] [Received: 01/07/2023] [Revised: 02/07/2023] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
Identification of genetic modulators of lysosomal enzyme activities and glycosphingolipids (GSLs) may facilitate the development of therapeutics for diseases in which they participate, including Lysosomal Storage Disorders (LSDs). To this end, we used a systems genetics approach: we measured 11 hepatic lysosomal enzymes and many of their natural substrates (GSLs), followed by modifier gene mapping by GWAS and transcriptomics associations in a panel of inbred strains. Unexpectedly, most GSLs showed no association between their levels and the enzyme activity that catabolizes them. Genomic mapping identified 30 shared predicted modifier genes between the enzymes and GSLs, which are clustered in three pathways and are associated with other diseases. Surprisingly, they are regulated by ten common transcription factors, and their majority by miRNA-340p. In conclusion, we have identified novel regulators of GSL metabolism, which may serve as therapeutic targets for LSDs and may suggest the involvement of GSL metabolism in other pathologies.
Collapse
Affiliation(s)
- Anyelo Durán
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | | | - Macarena Las Heras
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Boris Rebolledo-Jaramillo
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Valeria Olguín
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Juan F. Calderón
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago 7610658, Chile
| | - Silvana Zanlungo
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330033, Chile
| | - Jaime Gutiérrez
- Cellular Signaling and Differentiation Laboratory, School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago 7510602, Chile
| | - Frances M. Platt
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Andrés D. Klein
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| |
Collapse
|
7
|
[Application of adeno-associated virus-mediated gene therapy in lysosomal storage diseases]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2022; 24:1281-1287. [PMID: 36398557 PMCID: PMC9678058 DOI: 10.7499/j.issn.1008-8830.2207055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Lysosomal storage disorders (LSDs) are a group of single-gene inherited metabolic diseases caused by defects in lysosomal enzymes or function-related proteins. Enzyme replacement therapy is the main treatment method in clinical practice, but it has a poor effect in patients with neurological symptoms. With the rapid development of multi-omics, sequencing technology, and bioengineering, gene therapy has been applied in patients with LSDs. As one of the vectors of gene therapy, adeno-associated virus (AAV) has good prospects in the treatment of genetic and metabolic diseases. More and more studies have shown that AAV-mediated gene therapy is effective in LSDs. This article reviews the application of AAV-mediated gene therapy in LSDs.
Collapse
|
8
|
Treatment of Neuronopathic Mucopolysaccharidoses with Blood-Brain Barrier-Crossing Enzymes: Clinical Application of Receptor-Mediated Transcytosis. Pharmaceutics 2022; 14:pharmaceutics14061240. [PMID: 35745811 PMCID: PMC9229961 DOI: 10.3390/pharmaceutics14061240] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 02/04/2023] Open
Abstract
Enzyme replacement therapy (ERT) has paved the way for treating the somatic symptoms of lysosomal storage diseases (LSDs), but the inability of intravenously administered enzymes to cross the blood-brain barrier (BBB) has left the central nervous system (CNS)-related symptoms of LSDs largely impervious to the therapeutic benefits of ERT, although ERT via intrathecal and intracerebroventricular routes can be used for some neuronopathic LSDs (in particular, mucopolysaccharidoses). However, the considerable practical issues involved make these routes unsuitable for long-term treatment. Efforts have been made to modify enzymes (e.g., by fusing them with antibodies against innate receptors on the cerebrovascular endothelium) so that they can cross the BBB via receptor-mediated transcytosis (RMT) and address neuronopathy in the CNS. This review summarizes the various scientific and technological challenges of applying RMT to the development of safe and effective enzyme therapeutics for neuronopathic mucopolysaccharidoses; it then discusses the translational and methodological issues surrounding preclinical and clinical evaluation to establish RMT-applied ERT.
Collapse
|
9
|
Nagree MS, Felizardo TC, Faber ML, Rybova J, Rupar CA, Foley SR, Fuller M, Fowler DH, Medin JA. Autologous, lentivirus-modified, T-rapa cell "micropharmacies" for lysosomal storage disorders. EMBO Mol Med 2022; 14:e14297. [PMID: 35298086 PMCID: PMC8988206 DOI: 10.15252/emmm.202114297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 11/11/2022] Open
Abstract
T cells are the current choice for many cell therapy applications. They are relatively easy to access, expand in culture, and genetically modify. Rapamycin-conditioning ex vivo reprograms T cells, increasing their memory properties and capacity for survival, while reducing inflammatory potential and the amount of preparative conditioning required for engraftment. Rapamycin-conditioned T cells have been tested in patients and deemed to be safe to administer in numerous settings, with reduced occurrence of infusion-related adverse events. We demonstrate that ex vivo lentivirus-modified, rapamycin-conditioned CD4+ T cells can also act as next-generation cellular delivery vehicles-that is, "micropharmacies"-to disseminate corrective enzymes for multiple lysosomal storage disorders. We evaluated the therapeutic potential of this treatment platform for Fabry, Gaucher, Farber, and Pompe diseases in vitro and in vivo. For example, such micropharmacies expressing α-galactosidase A for treatment of Fabry disease were transplanted in mice where they provided functional enzyme in key affected tissues such as kidney and heart, facilitating clearance of pathogenic substrate after a single administration.
Collapse
Affiliation(s)
- Murtaza S Nagree
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
- Department of PediatricsMedical College of WisconsinMilwaukeeWIUSA
| | | | - Mary L Faber
- Department of PediatricsMedical College of WisconsinMilwaukeeWIUSA
| | - Jitka Rybova
- Department of PediatricsMedical College of WisconsinMilwaukeeWIUSA
| | - C Anthony Rupar
- Department of Pathology and Laboratory MedicineWestern UniversityLondonONCanada
| | - S Ronan Foley
- Juravinski Hospital and Cancer CentreMcMaster UniversityHamiltonONCanada
| | - Maria Fuller
- Genetics and Molecular PathologySA Pathology at Women's and Children's HospitalNorth AdelaideSAAustralia
| | | | - Jeffrey A Medin
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
- Department of PediatricsMedical College of WisconsinMilwaukeeWIUSA
- Department of BiochemistryMedical College of WisconsinMilwaukeeWIUSA
| |
Collapse
|
10
|
van Kooten HA, Ditters IAM, Hoogeveen-Westerveld M, Jacobs EH, van den Hout JMP, van Doorn PA, Pijnappel WWMP, van der Ploeg AT, van der Beek NAME. Antibodies against recombinant human alpha-glucosidase do not seem to affect clinical outcome in childhood onset Pompe disease. Orphanet J Rare Dis 2022; 17:31. [PMID: 35109913 PMCID: PMC8812154 DOI: 10.1186/s13023-022-02175-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/16/2022] [Indexed: 01/16/2023] Open
Abstract
Background Enzyme replacement therapy (ERT) with recombinant human alpha-glucosidase (rhGAA, alglucosidase alfa) has improved survival, motor outcomes, daily life activity and quality of life in Pompe patients. However, ERT in Pompe disease often induces formation of antibodies, which may reduce the efficacy of treatment and can lead to adverse events. In this study antibody formation and their effect on clinical outcome in patients with childhood onset Pompe disease treated with enzyme replacement therapy (ERT) with recombinant human alpha-glucosidase (rhGAA) are analyzed. Methods Enzyme-linked immunosorbent assay (ELISA) was used to determine anti-rhGAA antibody titers at predefined time points. The effect of antibodies on rhGAA activity (neutralizing effects) was measured in vitro. Clinical effects were evaluated by assessing muscle strength (MRC score) and function (QMFT-score), pulmonary function and infusion associated reactions (IARs). Results Twenty-two patients were included (age at start ERT 1.1–16.4 years, median treatment duration 12.4 years). Peak antibody titers were low (< 1:1250) in 9%, intermediate (1:1250–1:31,250) in 68% and high (≥ 1:31250) in 23% of patients; three patients (14%) had more than one titer of ≥ 1:31,250. Four patients (18%) experienced IARs; two patients from the high titer group had 86% of all IARs. Inhibition of intracellular GAA activity (58%) in vitro was found in one sample. The clinical course did not appear to be influenced by antibody titers. Conclusions Ninety-one percent of childhood onset Pompe patients developed anti-rhGAA antibodies (above background level), a minority of whom had high antibody titers at repeated time points, which do not seem to interfere with clinical outcome. High antibody titers may be associated with the occurrence of IARs. Although the majority of patients does not develop high titers; antibody titers should be determined in case of clinical deterioration. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02175-2.
Collapse
Affiliation(s)
- Harmke A van Kooten
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Imke A M Ditters
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marianne Hoogeveen-Westerveld
- Department of Pediatrics, Department of Clinical Genetics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Edwin H Jacobs
- Department of Pediatrics, Department of Clinical Genetics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Johanna M P van den Hout
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Pieter A van Doorn
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - W W M Pim Pijnappel
- Department of Pediatrics, Department of Clinical Genetics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Ans T van der Ploeg
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nadine A M E van der Beek
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands. .,Department of Neurology, Erasmus University Medical Center, Mailbox 2040, 3000 CA, Rotterdam, the Netherlands.
| |
Collapse
|
11
|
Massaro G, Geard AF, Liu W, Coombe-Tennant O, Waddington SN, Baruteau J, Gissen P, Rahim AA. Gene Therapy for Lysosomal Storage Disorders: Ongoing Studies and Clinical Development. Biomolecules 2021; 11:611. [PMID: 33924076 PMCID: PMC8074255 DOI: 10.3390/biom11040611] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Rare monogenic disorders such as lysosomal diseases have been at the forefront in the development of novel treatments where therapeutic options are either limited or unavailable. The increasing number of successful pre-clinical and clinical studies in the last decade demonstrates that gene therapy represents a feasible option to address the unmet medical need of these patients. This article provides a comprehensive overview of the current state of the field, reviewing the most used viral gene delivery vectors in the context of lysosomal storage disorders, a selection of relevant pre-clinical studies and ongoing clinical trials within recent years.
Collapse
Affiliation(s)
- Giulia Massaro
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| | - Amy F. Geard
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa;
| | - Wenfei Liu
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| | - Oliver Coombe-Tennant
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| | - Simon N. Waddington
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa;
- Gene Transfer Technology Group, EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK
| | - Julien Baruteau
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1EH, UK;
- Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, National Institute of Health Research, University College London, London WC1N 1EH, UK;
| | - Paul Gissen
- Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, National Institute of Health Research, University College London, London WC1N 1EH, UK;
| | - Ahad A. Rahim
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| |
Collapse
|
12
|
Ghosh A, Liao A, O’Leary C, Mercer J, Tylee K, Goenka A, Holley R, Jones SA, Bigger BW. Strategies for the Induction of Immune Tolerance to Enzyme Replacement Therapy in Mucopolysaccharidosis Type I. Mol Ther Methods Clin Dev 2019; 13:321-333. [PMID: 30976609 PMCID: PMC6441787 DOI: 10.1016/j.omtm.2019.02.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/24/2019] [Indexed: 01/16/2023]
Abstract
Enzyme replacement therapy with laronidase is an established treatment for Mucopolysaccharidosis type I (MPS I), but its efficacy may be limited by the development of anti-drug antibodies, which inhibit cellular uptake of the enzyme. In a related disorder, infantile Pompe disease, immune tolerance induction with low-dose, short-course methotrexate appears to reduce antibody formation. We investigated a similar regimen using oral methotrexate in three MPS I patients. All patients developed anti-laronidase immunoglobulin G (IgG) and immunoglobulin M (IgM) antibodies, and they had clinically relevant levels of cellular uptake inhibition. We then explored several immune tolerance induction strategies in MPS I mice: (1) methotrexate, (2) combination of non-depleting anti-CD4 and anti-CD8 monoclonal antibodies, (3) methotrexate with anti-CD4 and anti-CD8 monoclonals, (4) anti-CD4 monoclonal, and (5) anti-CD8 monoclonal. Treated mice received 10 weekly laronidase injections, and laronidase was delivered with adjuvant on day 49 to further challenge the immune system. Most regimens were only partially effective at reducing antibody responses, but two courses of non-depleting anti-CD4 monoclonal antibody (mAb) ablated immune responses to laronidase in seven of eight MPS I mice (87.5%), even after adjuvant stimulation. Immune tolerance induction with methotrexate does not appear to be effective in MPS I patients, but use of non-depleting anti-CD4 monoclonal is a promising strategy.
Collapse
Affiliation(s)
- Arunabha Ghosh
- Stem Cell and Neurotherapies, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
- Willink Biochemical Genetics Unit, Manchester Centre for Genomic Medicine, St. Mary’s Hospital, Manchester, UK
| | - Aiyin Liao
- Stem Cell and Neurotherapies, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| | - Claire O’Leary
- Stem Cell and Neurotherapies, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| | - Jean Mercer
- Willink Biochemical Genetics Unit, Manchester Centre for Genomic Medicine, St. Mary’s Hospital, Manchester, UK
| | - Karen Tylee
- Willink Biochemical Genetics Unit, Manchester Centre for Genomic Medicine, St. Mary’s Hospital, Manchester, UK
| | - Anu Goenka
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| | - Rebecca Holley
- Stem Cell and Neurotherapies, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| | - Simon A. Jones
- Willink Biochemical Genetics Unit, Manchester Centre for Genomic Medicine, St. Mary’s Hospital, Manchester, UK
| | - Brian W. Bigger
- Stem Cell and Neurotherapies, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
13
|
Nagree MS, Scalia S, McKillop WM, Medin JA. An update on gene therapy for lysosomal storage disorders. Expert Opin Biol Ther 2019; 19:655-670. [DOI: 10.1080/14712598.2019.1607837] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Murtaza S. Nagree
- Department of Medical Biophysics, University of Toronto, Toronto,
Ontario, Canada
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee,
WI, USA
| | - Simone Scalia
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee,
WI, USA
| | | | - Jeffrey A. Medin
- Department of Medical Biophysics, University of Toronto, Toronto,
Ontario, Canada
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee,
WI, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee,
WI, USA
| |
Collapse
|
14
|
Owens P, Wong M, Bhattacharya K, Ellaway C. Infantile-onset Pompe disease: A case series highlighting early clinical features, spectrum of disease severity and treatment response. J Paediatr Child Health 2018; 54:1255-1261. [PMID: 29889338 DOI: 10.1111/jpc.14070] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/26/2018] [Accepted: 05/02/2018] [Indexed: 02/03/2023]
Abstract
AIM Pompe disease is a rare, autosomal, recessive disorder. Alterations in the gene encoding lysosomal acid alpha-glucosidase cause impaired glycogen degradation and resultant lysosomal glycogen accumulation. Classic infantile-onset Pompe disease (IPD) manifests soon after birth, severe cases have complete/near complete enzyme deficiency. IPD is associated with a broad spectrum of non-specific clinical features, and diagnostic delays are common. Without treatment, death typically occurs within the first 2 years of life. We present case experiences to help expand paediatricians' understanding of factors contributing to diagnostic delay, clinical decline and to highlight the need for timely therapy. METHODS Data were extracted from IPD cases managed at our hospital. Key aspects of clinical presentation, diagnosis, genetic variations, management and overall outcomes were collated then compared with what is already known in the literature. RESULTS We report four IPD cases (three female). Two patients were cross-reactive immunological material negative. Age at symptom onset was 3-9 months, presenting clinical features were varied, and confirmatory diagnosis was significantly delayed in one patient. In concert with the literature, cardiomegaly, ventricular hypertrophy and delayed developmental milestones were seen in all four cases. Our cases demonstrate a range of disease severity, response to enzyme replacement therapy and antibody development. Significant immune responses were seen in two cases (one cross-reactive immunological material positive); despite immunomodulation therapy, both were associated with fatal outcomes. CONCLUSION Timely diagnosis and initiation of enzyme replacement therapy is critical to patient outcomes as IPD progresses rapidly and irreversible changes in clinical status may occur during the delay.
Collapse
Affiliation(s)
- Penny Owens
- Genetic Metabolic Disorders Service, Western Sydney Genetics Program, Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Melanie Wong
- Department of Immunology, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Child and Adolescent Health, University of Sydney, Sydney, New South Wales, Australia
| | - Kaustuv Bhattacharya
- Genetic Metabolic Disorders Service, Western Sydney Genetics Program, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Child and Adolescent Health, University of Sydney, Sydney, New South Wales, Australia.,Discipline of Genetic Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Carolyn Ellaway
- Genetic Metabolic Disorders Service, Western Sydney Genetics Program, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Child and Adolescent Health, University of Sydney, Sydney, New South Wales, Australia.,Discipline of Genetic Medicine, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
15
|
Platt FM, d'Azzo A, Davidson BL, Neufeld EF, Tifft CJ. Lysosomal storage diseases. Nat Rev Dis Primers 2018. [PMID: 30275469 DOI: 10.1038/s41572-018-0025-4]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lysosomal storage diseases (LSDs) are a group of over 70 diseases that are characterized by lysosomal dysfunction, most of which are inherited as autosomal recessive traits. These disorders are individually rare but collectively affect 1 in 5,000 live births. LSDs typically present in infancy and childhood, although adult-onset forms also occur. Most LSDs have a progressive neurodegenerative clinical course, although symptoms in other organ systems are frequent. LSD-associated genes encode different lysosomal proteins, including lysosomal enzymes and lysosomal membrane proteins. The lysosome is the key cellular hub for macromolecule catabolism, recycling and signalling, and defects that impair any of these functions cause the accumulation of undigested or partially digested macromolecules in lysosomes (that is, 'storage') or impair the transport of molecules, which can result in cellular damage. Consequently, the cellular pathogenesis of these diseases is complex and is currently incompletely understood. Several LSDs can be treated with approved, disease-specific therapies that are mostly based on enzyme replacement. However, small-molecule therapies, including substrate reduction and chaperone therapies, have also been developed and are approved for some LSDs, whereas gene therapy and genome editing are at advanced preclinical stages and, for a few disorders, have already progressed to the clinic.
Collapse
Affiliation(s)
- Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, UK.
| | - Alessandra d'Azzo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Beverly L Davidson
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth F Neufeld
- Department of Biological Chemistry, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | - Cynthia J Tifft
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
16
|
Abstract
Lysosomal storage diseases (LSDs) are a group of over 70 diseases that are characterized by lysosomal dysfunction, most of which are inherited as autosomal recessive traits. These disorders are individually rare but collectively affect 1 in 5,000 live births. LSDs typically present in infancy and childhood, although adult-onset forms also occur. Most LSDs have a progressive neurodegenerative clinical course, although symptoms in other organ systems are frequent. LSD-associated genes encode different lysosomal proteins, including lysosomal enzymes and lysosomal membrane proteins. The lysosome is the key cellular hub for macromolecule catabolism, recycling and signalling, and defects that impair any of these functions cause the accumulation of undigested or partially digested macromolecules in lysosomes (that is, 'storage') or impair the transport of molecules, which can result in cellular damage. Consequently, the cellular pathogenesis of these diseases is complex and is currently incompletely understood. Several LSDs can be treated with approved, disease-specific therapies that are mostly based on enzyme replacement. However, small-molecule therapies, including substrate reduction and chaperone therapies, have also been developed and are approved for some LSDs, whereas gene therapy and genome editing are at advanced preclinical stages and, for a few disorders, have already progressed to the clinic.
Collapse
|
17
|
Le SQ, Kan SH, Clarke D, Sanghez V, Egeland M, Vondrak KN, Doherty TM, Vera MU, Iacovino M, Cooper JD, Sands MS, Dickson PI. A Humoral Immune Response Alters the Distribution of Enzyme Replacement Therapy in Murine Mucopolysaccharidosis Type I. Mol Ther Methods Clin Dev 2018; 8:42-51. [PMID: 29159202 PMCID: PMC5684429 DOI: 10.1016/j.omtm.2017.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/30/2017] [Indexed: 11/28/2022]
Abstract
Antibodies against recombinant proteins can significantly reduce their effectiveness in unanticipated ways. We evaluated the humoral response of mice with the lysosomal storage disease mucopolysaccharidosis type I treated with weekly intravenous recombinant human alpha-l-iduronidase (rhIDU). Unlike patients, the majority of whom develop antibodies to recombinant human alpha-l-iduronidase, only approximately half of the treated mice developed antibodies against recombinant human alpha-l-iduronidase and levels were low. Serum from antibody-positive mice inhibited uptake of recombinant human alpha-l-iduronidase into human fibroblasts by partial inhibition compared to control serum. Tissue and cellular distributions of rhIDU were altered in antibody-positive mice compared to either antibody-negative or naive mice, with significantly less recombinant human alpha-l-iduronidase activity in the heart and kidney in antibody-positive mice. In the liver, recombinant human alpha-l-iduronidase was preferentially found in sinusoidal cells rather than in hepatocytes in antibody-positive mice. Antibodies against recombinant human alpha-l-iduronidase enhanced uptake of recombinant human alpha-l-iduronidase into macrophages obtained from MPS I mice. Collectively, these results imply that a humoral immune response against a therapeutic protein can shift its distribution preferentially into macrophage-lineage cells, causing decreased availability of the protein to the cells that are its therapeutic targets.
Collapse
Affiliation(s)
- Steven Q. Le
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Shih-hsin Kan
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Don Clarke
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Valentina Sanghez
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Martin Egeland
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Kristen N. Vondrak
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Terence M. Doherty
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Moin U. Vera
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Michelina Iacovino
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Jonathan D. Cooper
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Mark S. Sands
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Patricia I. Dickson
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| |
Collapse
|
18
|
Nelson BC, Hashem SI, Adler ED. Human-Induced Pluripotent Stem Cell-Based Modeling of Cardiac Storage Disorders. Curr Cardiol Rep 2017; 19:26. [PMID: 28251514 DOI: 10.1007/s11886-017-0829-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW The aim of this study is to review the published human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) models of cardiac storage disorders and to evaluate the limitations and future applications of this technology. RECENT FINDINGS Several cardiac storage disorders (CSDs) have been modeled using patient-specific hiPSC-CMs, including Anderson-Fabry disease, Danon disease, and Pompe disease. These models have shown that patient-specific hiPSC-CMs faithfully recapitulate key phenotypic features of CSDs and respond predictably to pharmacologic manipulation. hiPSC-CMs generated from patients with CSDs are representative models of the patient disease state and can be used as an in vitro system for the study of human cardiomyocytes. While these models suffer from several limitations, they are likely to play an important role in future mechanistic studies of cardiac storage disorders and the development of targeted therapeutics for these diseases.
Collapse
Affiliation(s)
- Bradley C Nelson
- Department of Medicine, Division of Cardiology, University of California San Diego, 9500 Gilman Drive, Biomedical Research Facility, Room 1217 AA, La Jolla, CA, 92093, USA
| | - Sherin I Hashem
- Department of Medicine, Division of Cardiology, University of California San Diego, 9500 Gilman Drive, Biomedical Research Facility, Room 1217 AA, La Jolla, CA, 92093, USA
| | - Eric D Adler
- Department of Medicine, Division of Cardiology, University of California San Diego, 9500 Gilman Drive, Biomedical Research Facility, Room 1217 AA, La Jolla, CA, 92093, USA.
| |
Collapse
|
19
|
Gatto F, Rossi B, Tarallo A, Polishchuk E, Polishchuk R, Carrella A, Nusco E, Alvino FG, Iacobellis F, De Leonibus E, Auricchio A, Diez-Roux G, Ballabio A, Parenti G. AAV-mediated transcription factor EB (TFEB) gene delivery ameliorates muscle pathology and function in the murine model of Pompe Disease. Sci Rep 2017; 7:15089. [PMID: 29118420 PMCID: PMC5678083 DOI: 10.1038/s41598-017-15352-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 10/24/2017] [Indexed: 01/15/2023] Open
Abstract
Pompe disease (PD) is a metabolic myopathy due to acid alpha-glucosidase deficiency and characterized by extensive glycogen storage and impaired autophagy. We previously showed that modulation of autophagy and lysosomal exocytosis by overexpression of the transcription factor EB (TFEB) gene was effective in improving muscle pathology in PD mice injected intramuscularly with an AAV-TFEB vector. Here we have evaluated the effects of TFEB systemic delivery on muscle pathology and on functional performance, a primary measure of efficacy in a disorder like PD. We treated 1-month-old PD mice with an AAV2.9-MCK-TFEB vector. An animal cohort was analyzed at 3 months for muscle and heart pathology. A second cohort was followed at different timepoints for functional analysis. In muscles from TFEB-treated mice we observed reduced PAS staining and improved ultrastructure, with reduced number and increased translucency of lysosomes, while total glycogen content remained unchanged. We also observed statistically significant improvements in rotarod performance in treated animals compared to AAV2.9-MCK-eGFP-treated mice at 5 and 8 months. Cardiac echography showed significant reduction in left-ventricular diameters. These results show that TFEB overexpression and modulation of autophagy result in improvements of muscle pathology and of functional performance in the PD murine model, with delayed disease progression.
Collapse
Affiliation(s)
- Francesca Gatto
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Barbara Rossi
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | | | | | | | | | - Edoardo Nusco
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | | | | | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Institute of Genetics and Biophysics, CNR, Naples, Italy
| | - Alberto Auricchio
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | | | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Translational Medical Sciences, Federico II University, Naples, Italy.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Giancarlo Parenti
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy. .,Department of Translational Medical Sciences, Federico II University, Naples, Italy.
| |
Collapse
|
20
|
|
21
|
Coutinho MF, Santos JI, Alves S. Less Is More: Substrate Reduction Therapy for Lysosomal Storage Disorders. Int J Mol Sci 2016; 17:ijms17071065. [PMID: 27384562 PMCID: PMC4964441 DOI: 10.3390/ijms17071065] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 06/24/2016] [Accepted: 06/27/2016] [Indexed: 12/11/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are a group of rare, life-threatening genetic disorders, usually caused by a dysfunction in one of the many enzymes responsible for intralysosomal digestion. Even though no cure is available for any LSD, a few treatment strategies do exist. Traditionally, efforts have been mainly targeting the functional loss of the enzyme, by injection of a recombinant formulation, in a process called enzyme replacement therapy (ERT), with no impact on neuropathology. This ineffectiveness, together with its high cost and lifelong dependence is amongst the main reasons why additional therapeutic approaches are being (and have to be) investigated: chaperone therapy; gene enhancement; gene therapy; and, alternatively, substrate reduction therapy (SRT), whose aim is to prevent storage not by correcting the original enzymatic defect but, instead, by decreasing the levels of biosynthesis of the accumulating substrate(s). Here we review the concept of substrate reduction, highlighting the major breakthroughs in the field and discussing the future of SRT, not only as a monotherapy but also, especially, as complementary approach for LSDs.
Collapse
Affiliation(s)
- Maria Francisca Coutinho
- Department of Human Genetics, Research and Development Unit, National Health Institute Doutor Ricardo Jorge, Rua Alexandre Herculano, 321 4000-055 Porto, Portugal.
| | - Juliana Inês Santos
- Department of Human Genetics, Research and Development Unit, National Health Institute Doutor Ricardo Jorge, Rua Alexandre Herculano, 321 4000-055 Porto, Portugal.
| | - Sandra Alves
- Department of Human Genetics, Research and Development Unit, National Health Institute Doutor Ricardo Jorge, Rua Alexandre Herculano, 321 4000-055 Porto, Portugal.
| |
Collapse
|