1
|
Zhang W, Huang Y, Su X, Zhao X, Sheng H, Liang C, Jiang M, Zeng C, Cai Y, Lin Y, Shao Y, Liu S, Jiang H, Liu L. Allogeneic hematopoietic stem cell transplantation for mucopolysaccharidosis patients: a single-center experience and assessment of quality of life. Ital J Pediatr 2025; 51:83. [PMID: 40102994 PMCID: PMC11921649 DOI: 10.1186/s13052-025-01919-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/22/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Allogeneic hematopoietic stem cell transplantation (HSCT) has proven to be a viable treatment option for patients with mucopolysaccharidoses (MPS). We investigate the efficacy and improvements in the quality of life of HSCT in pediatric patients with MPS. METHODS A retrospective analysis of transplantation data from 46 cases of MPS from a single institution in China was conducted. RESULTS The cohort of 46 patients included 9 cases of MPS I, 16 cases of MPS II, 15 cases of MPS IVA and 6 cases of MPS VI. The median age at diagnosis was 2.59 years. The median age at transplantation was 3.80 years. The median follow-up time was 3.1 years (range, 0.8-8.1 years) and 43 patients were alive. The incidence of grades II to IV aGVHD was 17.4%, wherein the incidence of grades III and IV aGVHD was 4.3%. The incidence of moderate-to-severe cGVHD was 6.5%. GAGs urinary excretion decreased and enzyme activity levels reached normal. After HSCT, multiple bone dysplasia, upper-airway obstruction and recurrent otitis media were significantly improved; vision, corneal clouding, cardiovascular disease, hepatosplenomegaly and hydrocephalus were improved or remained stable; neurological symptoms were improved or remained stable in most patients but progressed in others; the patients with MPS IH/S and MPS II reached nearly normal growth rate of height and weight. Meanwhile, the patients with MPS IH, MPS IVA and MPS VI remained poor growth after HSCT. The Activities of Daily Living (ADL) scores were improved in most patients with MPS. ADL scores in patients with severe phenotypes were lower than health control subjects and patients with attenuated phenotypes. CONCLUSIONS HSCT is a good therapeutic option for MPS and improves the quality of life of patients. MPS patients with attenuated phenotypes provide a better outcome in ADL after HSCT.
Collapse
Affiliation(s)
- Wen Zhang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China
| | - Yonglan Huang
- Department of Guangzhou Newborn Screening Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xueying Su
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China
| | - Xiaoyuan Zhao
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China
| | - Huiying Sheng
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China
| | - Cuili Liang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China
| | - Minyan Jiang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China
| | - Chunhua Zeng
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China
| | - Yanna Cai
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China
| | - Yunting Lin
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China
| | - Yongxian Shao
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China
| | - Sha Liu
- Department of Hematology and Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Hua Jiang
- Department of Hematology and Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| | - Li Liu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, Guangzhou, 510623, Guangdong, China.
| |
Collapse
|
2
|
Abelleyra Lastoria DA, Keynes S, Hughes D. Current and Emerging Therapies for Lysosomal Storage Disorders. Drugs 2025; 85:171-192. [PMID: 39826077 DOI: 10.1007/s40265-025-02145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2025] [Indexed: 01/20/2025]
Abstract
Lysosomal storage disorders (LSDs) are rare inherited metabolic disorders characterized by defects in the function of specific enzymes responsible for breaking down substrates within cellular organelles (lysosomes) essential for the processing of macromolecules. Undigested substrate accumulates within lysosomes, leading to cellular dysfunction, tissue damage, and clinical manifestations. Clinical features vary depending on the degree and type of enzyme deficiency, the type and extent of substrate accumulated, and the tissues affected. The heterogeneous nature of LSDs results in a variety of treatment approaches, which must be tailored to patient presentation and characteristics. The treatment landscape for LSDs is rapidly evolving. An up-to-date discussion of current evidence is required to provide clinicians with an appropriate overview of treatment options. Therefore, we aimed to review current and ongoing trials pertaining to the treatment of common LSDs.
Collapse
Affiliation(s)
| | - Sophie Keynes
- Institute for Medical and Biomedical Education, St George's, University of London, London, SW17 0RE, UK
| | - Derralynn Hughes
- Lysosomal Storage Disorders Unit, Royal Free London NHS Foundation Trust, University College London, London, NW3 2QG, UK.
| |
Collapse
|
3
|
Yalcin K, Uygun V, Ozturk Hismi B, Celen S, Ozturkmen S, Zhumatayev S, Daloglu H, Karasu G, Yesilipek A. Hematopoietic stem cell transplantation in children with mucopolysaccharidosis IVA: single center experience. Bone Marrow Transplant 2025; 60:47-51. [PMID: 39402187 DOI: 10.1038/s41409-024-02439-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 01/12/2025]
Abstract
Mucopolysaccharidosis IVA (MPS IVA; Morquio syndrome) is a lysosomal storage disorder and features systemic skeletal dysplasia that is caused by defective Nacetylgalactosamine-6-sulfate sulfatase (GALNS). Although there are convincing data for hematopoietic stem cell transplantation (HSCT) in certain types of MPS, the studies are limited for MPS IVA and more data is still pending to show the efficacy/safety of HSCT. This study included 3 girls and 7 boys, with a median age of 75,5 months (35-186 months), who underwent allogeneic HSCT for severe MPS IVA between February 12, 2021, and March 10, 2023. Enzyme levels, height growth, the most involved organs (ear, eye, and heart), and the activities of daily living (ADL) scoring system were monitored to assess the benefit of HSCT. In a median follow-up of 20 months (9-34 months), there is no severe transplant-related adverse event was observed. In all cases, normal enzyme levels were reached after HSCT. During the short follow-up period, our cases showed an increase in stature and improvement in daily activity functions. Here we present the data of our HSCT experience in MPS IVA with promising results regarding both safety and efficacy. Although there are signs of amelioration with HSCT, we need more data and long-term follow-up to comment properly on the benefits of HSCT in MPS IVA.
Collapse
Affiliation(s)
- Koray Yalcin
- Bahcesehir University, Medicalpark Goztepe Hospital, Pediatric Bone Marrow Transplantation Unit, Istanbul, Türkiye.
- Acibadem University, Institute of Health Science, Department of Medical Biotechnology, Istanbul, Türkiye.
| | - Vedat Uygun
- Istinye University, Medicalpark Antalya Hospital, Pediatric Bone Marrow Transplantation Unit, Antalya, Türkiye
| | - Burcu Ozturk Hismi
- Marmara University, Department of Pediatrics, Division of Inherited Metabolic Diseases, Istanbul, Türkiye
| | - Suna Celen
- Bahcesehir University, Medicalpark Goztepe Hospital, Pediatric Bone Marrow Transplantation Unit, Istanbul, Türkiye
| | - Seda Ozturkmen
- Medicalpark Antalya Hospital, Pediatric Bone Marrow Transplantation Unit, Antalya, Türkiye
| | - Suleimen Zhumatayev
- Medicalpark Goztepe Hospital, Pediatric Bone Marrow Transplantation Unit, Istanbul, Türkiye
| | - Hayriye Daloglu
- Antalya Bilim University, Medicalpark Antalya Hospital, Pediatric Bone Marrow Transplantation Unit, Antalya, Türkiye
| | - Gülsün Karasu
- Medicalpark Antalya Hospital, Pediatric Bone Marrow Transplantation Unit, Antalya, Türkiye
- Medicalpark Goztepe Hospital, Pediatric Bone Marrow Transplantation Unit, Istanbul, Türkiye
| | - Akif Yesilipek
- Medicalpark Antalya Hospital, Pediatric Bone Marrow Transplantation Unit, Antalya, Türkiye
- Medicalpark Goztepe Hospital, Pediatric Bone Marrow Transplantation Unit, Istanbul, Türkiye
| |
Collapse
|
4
|
Jubert C, De Berranger E, Castelle M, Dalle JH, Ouachee-Chardin M, Sevin C, Yakoub-Agha I, Brassier A. [Inborn error of metabolism and allogenic hematopoietic cell transplantation: Guidelines from the SFGM-TC]. Bull Cancer 2023; 110:S1-S12. [PMID: 36244825 DOI: 10.1016/j.bulcan.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/07/2022]
Abstract
Inherited Metabolic Diseases (IMD) are rare genetic diseases, including both lysosomal and peroxisomal diseases. Lysosomal diseases are related to the deficiency of one or more lysosomal enzymes or transporter. Lysosomal diseases are progressive and involve several tissues with most often neurological damage. Among peroxisomal diseases, X-linked adrenoleukodystrophy (ALD) is a neurodegenerative disease combining neurological and adrenal damage. For these diseases, enzyme replacement therapy (ERT), allogeneic hematopoietic cell transplantation (allo-HCT) and gene therapy represent various possible treatment options, used alone or in combination. The purpose of this workshop is to describe the indications, modalities, and follow-up of allo-HCT as well as the use of ERT peri-transplant. All indications for transplant in these rare diseases are associated with comorbidities and are subject to criteria that must be discussed in a dedicated national multidisciplinary consultation meeting. There are some consensual indications in type I-H mucopolysaccharidosis (MPS-IH) and in the cerebral form of ALD. For other IMDs, no clear benefit from the transplant has been demonstrated. The ideal donor is a non-heterozygous HLA-identical sibling. The recommended conditioning is myeloablative combining fludarabine and busulfan. In MPS-IH, ERT has to be started at diagnosis and continued until complete chimerism and normal enzyme assay are achieved. The pre-transplant assessment and post-transplant follow-up are made according to the published recommendations (PNDS). Standard follow-up is carried out jointly by the transplant and referral teams.
Collapse
Affiliation(s)
- Charlotte Jubert
- CHU de Bordeaux, groupe hospitalier Pellegrin, unité d'hématologie oncologie pédiatrique, place Améli-Raba-Léon, 33076 Bordeaux cedex, France.
| | - Eva De Berranger
- CHRU de Lille, service d'hématologie pédiatrique, avenue Eugène-Avinée, 59037 Lille, France
| | - Martin Castelle
- CHU de Necker-Enfants Malades, unité d'immuno-hématologie et rhumatologie pédiatrique, 149, rue de Sèvres, 75015 Paris, France
| | - Jean-Hugues Dalle
- Hôpital Robert-Debré, GHU Nord-Université de Paris, service d'immuno-hématologie pédiatrique, 48, boulevard Serurier, 75019 Paris, France
| | - Marie Ouachee-Chardin
- Institut d'hématologie et d'oncologie pédiatrique, 1, place Joseph-Renault, 69008 Lyon, France
| | - Caroline Sevin
- CHU de Kremlin-Bicêtre, neurologie pédiatrique, 78, rue du General-Leclerc, 94275 Le Kremlin-Bicêtre, France; ICM, 47, boulevard de l'Hôpital, 75013 Paris, France
| | - Ibrahim Yakoub-Agha
- Université de Lille, CHRU de Lille, Infinite, Inserm U1286, 59000 Lille, France
| | - Anais Brassier
- CHU de Necker, centre de référence des maladies héréditaires du métabolisme, 149, rue de Sèvres, 75015 Paris, France
| |
Collapse
|
5
|
Ream MA, Lam WKK, Grosse SD, Ojodu J, Jones E, Prosser LA, Rosé AM, Comeau AM, Tanksley S, Powell CM, Kemper AR. Evidence and recommendation for mucopolysaccharidosis type II newborn screening in the United States. Genet Med 2023; 25:100330. [PMID: 36445366 PMCID: PMC9905270 DOI: 10.1016/j.gim.2022.10.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/24/2022] [Accepted: 10/24/2022] [Indexed: 12/03/2022] Open
Abstract
Mucopolysaccharidosis type II (MPS II), also known as Hunter syndrome, is an X-linked condition caused by pathogenic variants in the iduronate-2-sulfatase gene. The resulting reduced activity of the enzyme iduronate-2-sulfatase leads to accumulation of glycosaminoglycans that can progressively affect multiple organ systems and impair neurologic development. In 2006, the US Food and Drug Administration approved idursulfase for intravenous enzyme replacement therapy for MPS II. After the data suggesting that early treatment is beneficial became available, 2 states, Illinois and Missouri, implemented MPS II newborn screening. Following a recommendation of the Advisory Committee on Heritable Disorders in Newborns and Children in February 2022, in August 2022, the US Secretary of Health and Human Services added MPS II to the Recommended Uniform Screening Panel, a list of conditions recommended for newborn screening. MPS II was added to the Recommended Uniform Screening Panel after a systematic evidence review reported the accuracy of screening, the benefit of presymptomatic treatment compared with usual case detection, and the feasibility of implementing MPS II newborn screening. This manuscript summarizes the findings of the evidence review that informed the Advisory Committee's decision.
Collapse
Affiliation(s)
- Margie A Ream
- Division of Child Neurology, Nationwide Children's Hospital, Columbus, OH.
| | - Wendy K K Lam
- Duke Clinical and Translational Science Institute, Duke University School of Medicine, Durham, NC
| | - Scott D Grosse
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA
| | - Jelili Ojodu
- Association of Public Health Laboratories, Silver Spring, MD
| | - Elizabeth Jones
- Association of Public Health Laboratories, Silver Spring, MD
| | - Lisa A Prosser
- Susan B. Meister Child Health Evaluation and Research Center, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Angela M Rosé
- Susan B. Meister Child Health Evaluation and Research Center, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Anne Marie Comeau
- New England Newborn Screening Program, Department of Pediatrics, UMass Chan School of Medicine, Worcester, MA
| | - Susan Tanksley
- Laboratory Services Section, Texas Department of State Health Services, Austin, TX
| | - Cynthia M Powell
- Division of Genetics and Metabolism, Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Alex R Kemper
- Department of Pediatrics, Nationwide Children's Hospital, Columbus, OH
| |
Collapse
|
6
|
Penon-Portmann M, Blair DR, Harmatz P. Current and new therapies for mucopolysaccharidoses. Pediatr Neonatol 2023; 64 Suppl 1:S10-S17. [PMID: 36464587 DOI: 10.1016/j.pedneo.2022.10.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/03/2022] [Indexed: 12/05/2022] Open
Abstract
The mucopolysaccharidoses (MPSs) are a subset of lysosomal storage diseases caused by deficiencies in the enzymes required to metabolize glycosaminoglycans (GAGs), a group of extracellular heteropolysaccharides that play diverse roles in human physiology. As a result, GAGs accumulate in multiple tissues, and affected patients typically develop progressive, multi-systemic symptoms in early childhood. Over the last 30 years, the treatments available for the MPSs have evolved tremendously. There are now multiple therapies that delay the progression of these debilitating disorders, although their effectiveness varies according to MPS sub-type. In this review, we discuss the basic principle underlying MPS treatment (enzymatic "cross correction"), and we review the three general modalities currently available: hematopoietic stem cell transplantation, enzymatic replacement, and gene therapy. For each treatment type, we discuss its effectiveness across the MPS subtypes, its inherent risks, and future directions. Long term, we suspect that treatment for the MPSs will continue to evolve, and through a combination of early diagnosis and effective management, these patients will continue to live longer lives with improved outcomes for quality of life.
Collapse
Affiliation(s)
- Monica Penon-Portmann
- UCSF Benioff Children's Hospital Oakland, Oakland, CA, USA; Seattle Children's Hospital, Seattle, WA, USA.
| | - David R Blair
- UCSF Benioff Children's Hospital Oakland, Oakland, CA, USA; Division of Medical Genetics and Genomics, Department of Pediatrics, UCSF, San Francisco, CA, USA
| | - Paul Harmatz
- UCSF Benioff Children's Hospital Oakland, Oakland, CA, USA
| |
Collapse
|
7
|
A novel preclinical model of mucopolysaccharidosis type II for developing human hematopoietic stem cell gene therapy. Gene Ther 2022; 30:288-296. [PMID: 35835952 DOI: 10.1038/s41434-022-00357-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 11/08/2022]
Abstract
A hematopoietic stem cell (HSC) gene therapy (GT) using lentiviral vectors has attracted interest as a promising treatment approach for neuropathic lysosomal storage diseases. To proceed with the clinical development of HSC-GT, evaluation of the therapeutic potential of gene-transduced human CD34+ (hCD34+) cells in vivo is one of the key issues before human trials. Here, we established an immunodeficient murine model of mucopolysaccharidosis type II (MPS II), which are transplantable human cells, and demonstrated the application of those mice in evaluating the therapeutic efficacy of gene-modified hCD34+ cells. NOG/MPS II mice, which were generated using CRISPR/Cas9, exhibited a reduction of disease-causing enzyme iduronate-2-sulfatatase (IDS) activity and the accumulation of glycosaminoglycans in their tissues. When we transplanted hCD34+ cells transduced with a lentiviral vector carrying the IDS gene into NOG/MPS II mice, a significant amelioration of biochemical pathophenotypes was observed in the visceral and neuronal tissues of those mice. In addition, grafted cells in the NOG/MPS II mice showed the oligoclonal integration pattern of the vector, but no obvious clonal dominance was detected in the mice. Our findings indicate the promising application of NOG/MPS II mice to preclinical study of HSC-GT for MPS II using human cells.
Collapse
|
8
|
Sreekantam S, Smith L, Stewart C, Kearney S, Lawson S, Raiman J, Vijay S, Santra S. Efficacy of early haematopoietic stem cell transplantation versus enzyme replacement therapy on neurological progression in severe Hunter syndrome: Case report of siblings and literature review. Mol Genet Metab Rep 2022; 32:100881. [PMID: 35663302 PMCID: PMC9160838 DOI: 10.1016/j.ymgmr.2022.100881] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 11/30/2022] Open
Abstract
Hunter syndrome is a neurodegenerative lysosomal storage disorder with limited treatment options to halt the progressive neurocognitive decline. Whilst Intravenous enzyme replacement therapy (ERT) does not cross the blood brain barrier; Intrathecal ERT, in clinical studies, did not demonstrate significant effect on cognition, despite having better CNS delivery. Hematopoietic stem cell transplantation (HSCT) has the potential to treat CNS disease. We reviewed the literature and outline our experience of treating two siblings with severe Hunter syndrome: ‘Sibling A' with intravenous and intrathecal ERT and ‘Sibling B' with Early HSCT. A literature review identified 8 articles reporting on the comparative efficacy of both treatments. Our clinical outcomes indicate that Sibling B performed better than Sibling A in relation to early developmental milestones as well as neurocognition, activities of daily living, quality of life and neurophysiological outcomes in mid childhood. Sibling A's developmental trajectory fell within the extremely low range and Sibling B's development trajectory fell within the low-average to average range. This suggests HSCT had a disease modifying effect and highlights the efficacy of early HSCT in moderating the CNS progression in Hunter syndrome. Long term follow up is needed to elucidate the efficacy of HSCT on neurological progression.
Collapse
Affiliation(s)
- Srividya Sreekantam
- Department of Inherited Metabolic Disorders, Birmingham Women's And Children's Hospital NHS Trust, UK
| | - Laura Smith
- Department of Clinical Psychology, Birmingham Women's And Children's Hospital NHS Trust, UK
| | - Catherine Stewart
- Department of Inherited Metabolic Disorders, Birmingham Women's And Children's Hospital NHS Trust, UK
| | - Shauna Kearney
- Department of Clinical Psychology, Birmingham Women's And Children's Hospital NHS Trust, UK
| | - Sarah Lawson
- Department of Haematology, Birmingham Women's And Children's Hospital NHS Trust, UK
| | - Julian Raiman
- Department of Inherited Metabolic Disorders, Birmingham Women's And Children's Hospital NHS Trust, UK
| | - Suresh Vijay
- Department of Inherited Metabolic Disorders, Birmingham Women's And Children's Hospital NHS Trust, UK
| | - Saikat Santra
- Department of Inherited Metabolic Disorders, Birmingham Women's And Children's Hospital NHS Trust, UK
| |
Collapse
|
9
|
Rossini L, Durante C, Marzollo A, Biffi A. New Indications for Hematopoietic Stem Cell Gene Therapy in Lysosomal Storage Disorders. Front Oncol 2022; 12:885639. [PMID: 35646708 PMCID: PMC9136164 DOI: 10.3389/fonc.2022.885639] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/11/2022] [Indexed: 12/04/2022] Open
Abstract
Lysosomal storage disorders (LSDs) are a heterogenous group of disorders due to genetically determined deficits of lysosomal enzymes. The specific molecular mechanism and disease phenotype depends on the type of storage material. Several disorders affect the brain resulting in severe clinical manifestations that substantially impact the expectancy and quality of life. Current treatment modalities for LSDs include enzyme replacement therapy (ERT) and hematopoietic cell transplantation (HCT) from allogeneic healthy donors, but are available for a limited number of disorders and lack efficacy on several clinical manifestations. Hematopoietic stem cell gene therapy (HSC GT) based on integrating lentiviral vectors resulted in robust clinical benefit when administered to patients affected by Metachromatic Leukodystrophy, for whom it is now available as a registered medicinal product. More recently, HSC GT has also shown promising results in Hurler syndrome patients. Here, we discuss possible novel HSC GT indications that are currently under development. If these novel drugs will prove effective, they might represent a new standard of care for these disorders, but several challenges will need to be addresses, including defining and possibly expanding the patient population for whom HSC GT could be efficacious.
Collapse
Affiliation(s)
- Linda Rossini
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
| | - Caterina Durante
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
| | - Antonio Marzollo
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
- Fondazione Citta’ della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
| | - Alessandra Biffi
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
- Maternal and Child Health Department, Padua University, Padua, Italy
- *Correspondence: Alessandra Biffi,
| |
Collapse
|
10
|
Kimura M, Azuma Y, Taguchi S, Takagi M, Mori H, Shimomura Y, Niwa JI, Doyu M, Okumura A. Subcortical infarction in a young adult with Hunter syndrome. Brain Dev 2022; 44:343-346. [PMID: 35125232 DOI: 10.1016/j.braindev.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/16/2021] [Accepted: 01/16/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Hunter syndrome (mucopolysaccharidosis type II, MPS II) is an X-linked lysosomal storage disease caused by deficiency of iduronate-2-sulfatase. Recently, stroke caused by embolization with Hunter syndrome has been reported. Here, we report the case of a 23-year-old Japanese man with Hunter syndrome who developed subcortical infarction by the mechanism similar to branch atheromatous disease (BAD). CASE PRESENTATION He had been treated with idursulfase supplementation. He presented with left-sided weakness and conjugate eye deviation to the right, and was diagnosed with branch atheromatous disease affecting the right corona radiata, based on MRI findings. The patient was treated with argatroban and aspirin. Magnetic resonance angiography demonstrated no evidence of luminal narrowing of the cerebral arteries. T1-sampling perfection with application-optimized contrasts by using different flip angle evolutions (SPACE) imaging revealed thickened middle cerebral artery. The patient had markedly low flow-mediated vasodilation, suggesting impaired vasodilation in response to nitric monoxide. CONCLUSION The arterial wall thickening and impaired vasodilation in the cerebral arteries related to subcortical infarction. We should clarify the mechanism of cerebral infarction in Hunter syndrome patients.
Collapse
Affiliation(s)
- Motoya Kimura
- Department of Pediatrics, Aichi Medical University, Nagakute, Japan; Department of Neurology, Aichi Medical University, Nagakute, Japan.
| | - Yoshiteru Azuma
- Department of Pediatrics, Aichi Medical University, Nagakute, Japan
| | - Soutarou Taguchi
- Department of Neurology, Aichi Medical University, Nagakute, Japan; Parkinson's Disease Advanced Therapy Center, Aichi Medical University Hospital, Nagakute, Japan
| | - Mizuki Takagi
- Department of Pediatrics, Aichi Medical University, Nagakute, Japan
| | - Hiromitsu Mori
- Department of Pediatrics, Aichi Medical University, Nagakute, Japan
| | - Yasuto Shimomura
- Department of Pediatrics, Aichi Medical University, Nagakute, Japan
| | - Jun-Ichi Niwa
- Department of Neurology, Aichi Medical University, Nagakute, Japan
| | - Manabu Doyu
- Department of Neurology, Aichi Medical University, Nagakute, Japan
| | - Akihisa Okumura
- Department of Pediatrics, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
11
|
Horgan C, Jones SA, Bigger BW, Wynn R. Current and Future Treatment of Mucopolysaccharidosis (MPS) Type II: Is Brain-Targeted Stem Cell Gene Therapy the Solution for This Devastating Disorder? Int J Mol Sci 2022; 23:4854. [PMID: 35563245 PMCID: PMC9105950 DOI: 10.3390/ijms23094854] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 02/04/2023] Open
Abstract
Mucopolysaccharidosis type II (Hunter Syndrome) is a rare, x-linked recessive, progressive, multi-system, lysosomal storage disease caused by the deficiency of iduronate-2-sulfatase (IDS), which leads to the pathological storage of glycosaminoglycans in nearly all cell types, tissues and organs. The condition is clinically heterogeneous, and most patients present with a progressive, multi-system disease in their early years. This article outlines the pathology of the disorder and current treatment strategies, including a detailed review of haematopoietic stem cell transplant outcomes for MPSII. We then discuss haematopoietic stem cell gene therapy and how this can be employed for treatment of the disorder. We consider how preclinical innovations, including novel brain-targeted techniques, can be incorporated into stem cell gene therapy approaches to mitigate the neuropathological consequences of the condition.
Collapse
Affiliation(s)
- Claire Horgan
- Blood and Marrow Transplant Unit, Department of Paediatric Haematology, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK;
| | - Simon A. Jones
- Willink Unit, Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK;
| | - Brian W. Bigger
- Stem Cell and Neuropathies, Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester M13 9PT, UK;
| | - Robert Wynn
- Blood and Marrow Transplant Unit, Department of Paediatric Haematology, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK;
| |
Collapse
|
12
|
Fang X, Zhu C, Zhu X, Feng Y, Jiao Z, Duan H, Kong X, Liu N. Molecular analysis and novel variation identification of Chinese pedigrees with mucopolysaccharidosis using targeted next-generation sequencing. Clin Chim Acta 2022; 524:194-200. [PMID: 34813777 DOI: 10.1016/j.cca.2021.11.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 01/15/2023]
Abstract
BACKGROUND Mucopolysaccharidosis (MPS) refers to a group of lysosomal storage disorders for which seven types and 11 subtypes are currently recognized. Targeted next-generation sequencing (NGS) offers an important method of disease typing, diagnosis, prenatal diagnosis, and treatment. METHODS Gene variations in 48 Chinese MPS patients were evaluated using NGS, and the pathogenicity of the DNA alterations was evaluated using PolyPhen2, SIFT, and Mutation Taster. The effect of amino acid substitution on protein structure was also assessed. RESULTS Four pedigrees with MPS I (8.3%), 28 with MPS II (58.3%), two with MPS IIIA (4.2%), two with MPS IIIB (4.2%), six with MPS IVA (12.5%), one with MPS IVB (2.1%), and five with MPS VI (10.4%) were identified. Of the 69 variations identified, 11 were novel variants (three in IDUA, five in IDS, and three in GALNS), all of which were predicted to be disease-causing except for one, and were associated with impaired protein structure and function. CONCLUSIONS Targeted NGS technology is effective for the gene-based testing of MPS disorders, which show high allelic heterogeneity. MPS II was the predominant form in Chinese. Our study expands the existing variation spectrum of MPS, which is important for disease management and genetic counseling.
Collapse
Affiliation(s)
- Xiaohua Fang
- Obstetrics and Gynecology Department, Genetics and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, 450052 PR China
| | - Chaofeng Zhu
- Obstetrics and Gynecology Department, Genetics and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, 450052 PR China
| | - Xiaofan Zhu
- Obstetrics and Gynecology Department, Genetics and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, 450052 PR China
| | - Yin Feng
- Obstetrics and Gynecology Department, Genetics and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, 450052 PR China
| | - Zhihui Jiao
- Obstetrics and Gynecology Department, Genetics and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, 450052 PR China
| | - Huikun Duan
- Obstetrics and Gynecology Department, Genetics and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, 450052 PR China
| | - Xiangdong Kong
- Obstetrics and Gynecology Department, Genetics and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, 450052 PR China
| | - Ning Liu
- Obstetrics and Gynecology Department, Genetics and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, 450052 PR China.
| |
Collapse
|
13
|
Tomita K, Okamoto S, Seto T, Hamazaki T. Real world long-term outcomes in patients with mucopolysaccharidosis type II: A retrospective cohort study. Mol Genet Metab Rep 2021; 29:100816. [PMID: 34745890 PMCID: PMC8554623 DOI: 10.1016/j.ymgmr.2021.100816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 11/27/2022] Open
Abstract
We investigated the decline of activities of daily living with symptomatic progression in patients with mucopolysaccharidosis type II (MPS II) and investigated the associated factors. Clinical data were retrospectively collected from the medical records of 28 patients with MPS II who visited our hospital between October 2007 and August 2019. Activities of daily living were assessed over time using a 5-point scale (from stage 1, indicating independent, to stage 5, indicating total assistance + medical care); the relationships of the interval years from stage 2 (mild symptoms) to stage 4 (total assistance) with therapeutic intervention, anti-drug antibodies (ADA), urinary glycosaminoglycans (uGAG), and genotypes were analyzed. Eight are attenuated types, and 20 are severe types. Further, 20 underwent enzyme replacement therapy (ERT) alone, 5 underwent hematopoietic stem cell transplantation (HSCT) alone, and 3 underwent both therapy. The mean interval years (standard deviation) from stage 2 to 4 was 3.5 (0.7) and 7.3 (3.3) in patients who started undergoing ERT (n = 6) and HSCT (n = 3) at stage 2, respectively, whereas it was 3.1 (1.5) in patients who received no treatment until they reached stage 4 (n = 8). The study findings revealed the process of changes in the activities of daily living over a long duration in patients with MPS II undergoing different treatments. In severe type, the activity deteriorated regardless of the stage at which ERT was initiated. The activity declined slower in patients who received HSCT at an early stage.
Collapse
Affiliation(s)
- Kazuyoshi Tomita
- Corresponding author at: 1-4-3 Asahi-machi, Abeno-ward Osaka City, Osaka Pref 545-8585, Japan.
| | | | | | | |
Collapse
|
14
|
Chen H, Khan S, Celik B, Suzuki Y, Ago Y, Tomatsu S. Activity of daily living in mucopolysaccharidosis IVA patients: Evaluation of therapeutic efficacy. Mol Genet Genomic Med 2021; 9:e1806. [PMID: 34623762 PMCID: PMC8606213 DOI: 10.1002/mgg3.1806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 01/16/2023] Open
Abstract
Background Mucopolysaccharidosis IVA (MPS IVA, also called Morquio A syndrome) is caused by a deficiency of N‐acetylglucosamine‐6‐sulfate sulfatase (GALNS) and results in skeletal dysplasia symptoms such as short stature and abnormal gait. Treatments include enzyme replacement therapy (ERT) and hematopoietic stem cell transplantation (HSCT), but the effects are limited depending on the age of initiation and clinical phenotype. Thus, this study aims to assess the effects of treatments on MPS IVA patients compared to untreated MPS IVA patients and an age‐matched control group. Methods We used activity of daily living (ADL) survey with 4 sections: “movement,” “movement with cognition,” “cognition,” and “other MPS symptoms.” Lower scores indicate more assistance required. This study included 161 patients, 270 total surveys, and 70 patients with longitudinal data. Results We describe 134 severe patients and 25 attenuated patients. ERT and HSCT treatment improved only the “other MPS symptoms” section in severe patients. There were no differences between ERT and HSCT severe patient scores. A 19‐year‐old male patient, who had robust physical training, provided a significant increase in “movement” without treatment, suggesting the importance of exercise. Conclusion Overall, this ADL questionnaire has demonstrated validation and reliability in assessing the MPS IVA patients and therapeutic efficacy.
Collapse
Affiliation(s)
- Hui Chen
- University of Delaware, Newark, DE, USA.,Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Shaukat Khan
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Betul Celik
- University of Delaware, Newark, DE, USA.,Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Yasuyuki Suzuki
- Medical Education Development Center, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Yasuhiko Ago
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- University of Delaware, Newark, DE, USA.,Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA.,Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan.,Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
15
|
Differences in MPS I and MPS II Disease Manifestations. Int J Mol Sci 2021; 22:ijms22157888. [PMID: 34360653 PMCID: PMC8345985 DOI: 10.3390/ijms22157888] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
Mucopolysaccharidosis (MPS) type I and II are two closely related lysosomal storage diseases associated with disrupted glycosaminoglycan catabolism. In MPS II, the first step of degradation of heparan sulfate (HS) and dermatan sulfate (DS) is blocked by a deficiency in the lysosomal enzyme iduronate 2-sulfatase (IDS), while, in MPS I, blockage of the second step is caused by a deficiency in iduronidase (IDUA). The subsequent accumulation of HS and DS causes lysosomal hypertrophy and an increase in the number of lysosomes in cells, and impacts cellular functions, like cell adhesion, endocytosis, intracellular trafficking of different molecules, intracellular ionic balance, and inflammation. Characteristic phenotypical manifestations of both MPS I and II include skeletal disease, reflected in short stature, inguinal and umbilical hernias, hydrocephalus, hearing loss, coarse facial features, protruded abdomen with hepatosplenomegaly, and neurological involvement with varying functional concerns. However, a few manifestations are disease-specific, including corneal clouding in MPS I, epidermal manifestations in MPS II, and differences in the severity and nature of behavioral concerns. These phenotypic differences appear to be related to different ratios between DS and HS, and their sulfation levels. MPS I is characterized by higher DS/HS levels and lower sulfation levels, while HS levels dominate over DS levels in MPS II and sulfation levels are higher. The high presence of DS in the cornea and its involvement in the arrangement of collagen fibrils potentially causes corneal clouding to be prevalent in MPS I, but not in MPS II. The differences in neurological involvement may be due to the increased HS levels in MPS II, because of the involvement of HS in neuronal development. Current treatment options for patients with MPS II are often restricted to enzyme replacement therapy (ERT). While ERT has beneficial effects on respiratory and cardiopulmonary function and extends the lifespan of the patients, it does not significantly affect CNS manifestations, probably because the enzyme cannot pass the blood-brain barrier at sufficient levels. Many experimental therapies, therefore, aim at delivery of IDS to the CNS in an attempt to prevent neurocognitive decline in the patients.
Collapse
|
16
|
Therapy-type related long-term outcomes in mucopolysaccaridosis type II (Hunter syndrome) - Case series. Mol Genet Metab Rep 2021; 28:100779. [PMID: 34258227 PMCID: PMC8251508 DOI: 10.1016/j.ymgmr.2021.100779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 11/22/2022] Open
Abstract
Mucopolysaccharidosis type II (MPS II, Hunter syndrome) is a rare, X-linked recessive multisystem lysosomal storage disease due to iduronate-2-sulfatase enzyme deficiency. We presented three unrelated Slovenian patients with the severe form of MPS II that received three different management approaches: natural course of the disease without received specific treatment, enzyme replacement therapy (ERT), and hematopoietic stem cell transplantation (HSCT). The decision on the management depended on disease severity, degree of cognitive impairment, and parent's informed decision. The current benefits of MPS II treatments are limited. The lifelong costly intravenous ERT brings significant benefits but the patients with severe phenotypes and neurological involvement progress to cognitive decline and disability regardless of ERT, as demonstrated in published reviews and our case series. The patient after HSCT was the only one of the three cases reported to show a slowly progressing cognitive development. The type of information from the case series is insufficient for generalized conclusions, but with advanced myeloablative conditioning, HSCT may be a preferred treatment option in early diagnosed MPS II patients with the severe form of the disease and low disease burden at the time of presentation.
Collapse
|
17
|
McBride KL, Flanigan KM. Update in the Mucopolysaccharidoses. Semin Pediatr Neurol 2021; 37:100874. [PMID: 33892850 DOI: 10.1016/j.spen.2021.100874] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 12/15/2022]
Abstract
The mucopolysaccharidoses (MPS) are a genetically heterogenous group of enzyme deficiencies marked by accumulation of glycosaminoglycans in lysosomes leading to multisystem disease. Although significant therapeutic advances have been made for the MPS disorders, including recombinant enzyme replacement approaches, the neuronopathic features of MPS lack adequate treatment. Gene therapies, including adeno-associated virus vectors targeting the central nervous system, hold significant promise for this group of disorders. Optimal outcomes of all therapies will require early disease identification and treatment, ideally by newborn screening.
Collapse
Affiliation(s)
- Kim L McBride
- The Center for Cardiovascular Research and the Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital; and the Ohio State University, Columbus, OH; Department of Pediatrics, the Ohio State University, Columbus, OH.
| | - Kevin M Flanigan
- Department of Pediatrics, the Ohio State University, Columbus, OH; Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital; and the Ohio State University, Columbus, OH.
| |
Collapse
|
18
|
Kubaski F, Vairo F, Baldo G, de Oliveira Poswar F, Corte AD, Giugliani R. Therapeutic Options for Mucopolysaccharidosis II (Hunter Disease). Curr Pharm Des 2020; 26:5100-5109. [PMID: 33138761 DOI: 10.2174/1381612826666200724161504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/17/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Mucopolysaccharidosis type II (Hunter syndrome, or MPS II) is an X-linked lysosomal disorder caused by the deficiency of iduronate-2-sulfatase, which leads to the accumulation of glycosaminoglycans (GAGs) in a variety of tissues, resulting in a multisystemic disease that can also impair the central nervous system (CNS). OBJECTIVE This review focuses on providing the latest information and expert opinion about the therapies available and under development for MPS II. METHODS We have comprehensively revised the latest studies about hematopoietic stem cell transplantation (HSCT), enzyme replacement therapy (ERT - intravenous, intrathecal, intracerebroventricular, and intravenous with fusion proteins), small molecules, gene therapy/genome editing, and supportive management. RESULTS AND DISCUSSION Intravenous ERT is a well-established specific therapy, which ameliorates the somatic features but not the CNS manifestations. Intrathecal or intracerebroventricular ERT and intravenous ERT with fusion proteins, presently under development, seem to be able to reduce the levels of GAGs in the CNS and have the potential of reducing the impact of the neurological burden of the disease. Gene therapy and/or genome editing have shown promising results in preclinical studies, bringing hope for a "one-time therapy" soon. Results with HSCT in MPS II are controversial, and small molecules could potentially address some disease manifestations. In addition to the specific therapeutic options, supportive care plays a major role in the management of these patients. CONCLUSION At this time, the treatment of individuals with MPS II is mainly based on intravenous ERT, whereas HSCT can be a potential alternative in specific cases. In the coming years, several new therapy options that target the neurological phenotype of MPS II should be available.
Collapse
Affiliation(s)
- Francyne Kubaski
- Postgraduate Program in Genetics and Molecular Biology, UFRGS, Porto Alegre, Brazil
| | - Filippo Vairo
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
| | - Guilherme Baldo
- Postgraduate Program in Genetics and Molecular Biology, UFRGS, Porto Alegre, Brazil
| | | | - Amauri Dalla Corte
- Postgraduation Program in Medicine: Medical Sciences, UFRGS, Porto Alegre, Brazil
| | - Roberto Giugliani
- Postgraduate Program in Genetics and Molecular Biology, UFRGS, Porto Alegre, Brazil
| |
Collapse
|
19
|
Hogan MJ, Stephens K, Smith E, Jalazo ER, Hendriksz CJ, Edwards LJ, Bjoraker KJ. Toileting Abilities Survey as a surrogate outcome measure for cognitive function: Findings from neuronopathic mucopolysaccharidosis II patients treated with idursulfase and intrathecal idursulfase. Mol Genet Metab Rep 2020; 25:100669. [PMID: 33101985 PMCID: PMC7578548 DOI: 10.1016/j.ymgmr.2020.100669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 10/12/2020] [Indexed: 10/31/2022] Open
Abstract
An outcome measure of toileting skills, the Toileting Abilities Survey or TAS, with sensitivity to detect change in a neurodegenerative disorder such as MPS II, was developed. The TAS was used in a research study of patients (n = 86) with the neuronopathic form of MPS II to measure treatment benefit of intrathecal idursulfase. Treatment with idursulfase and intrathecal idursulfase is associated with significantly higher individual and overall toileting skills versus treatment with idursulfase alone.
Collapse
Affiliation(s)
| | | | - Erin Smith
- Backpack Health, a Konica Minolta Service, Boston, MA, USA
| | - Elizabeth R Jalazo
- Division of Genetics and Metabolism, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christian J Hendriksz
- Department of Paediatrics and Child Health at the Steve Biko Academic Unit, University of Pretoria, South Africa
| | - Lloyd J Edwards
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | | |
Collapse
|
20
|
Miwa S, Watabe AM, Shimada Y, Higuchi T, Kobayashi H, Fukuda T, Kato F, Ida H, Ohashi T. Efficient engraftment of genetically modified cells is necessary to ameliorate central nervous system involvement of murine model of mucopolysaccharidosis type II by hematopoietic stem cell targeted gene therapy. Mol Genet Metab 2020; 130:262-273. [PMID: 32631737 DOI: 10.1016/j.ymgme.2020.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/12/2020] [Accepted: 06/12/2020] [Indexed: 10/24/2022]
Abstract
Mucopolysaccharidosis type II (MPS II) is a lysosomal storage disease (LSD) caused by a deficiency of the iduronate-2-sulfatase (IDS) that catabolizes glycosaminoglycans (GAGs). Abnormal accumulations of GAGs in somatic cells lead to various manifestations including central nervous system (CNS) disease. Enzyme replacement therapy (ERT) and hematopoietic stem cell transplantation (HSCT) are the currently available therapy for MPS II, but both therapies fail to improve CNS manifestations. We previously showed that hematopoietic stem cell targeted gene therapy (HSC-GT) with lethal irradiation improved CNS involvement in a murine model of MPS II which lacks the gene coding for IDS. However, the strong preconditioning, with lethal irradiation, would cause a high rate of morbidity and mortality. Therefore, we tested milder preconditioning procedures with either low dose irradiation or low dose irradiation plus an anti c-kit monoclonal antibody (ACK2) to assess CNS effects in mice with MPS II after HSC-GT. Mice from all the HSC-GT groups displayed super-physiological levels of IDS enzyme activity and robust reduction of abnormally accumulated GAGs to the wild type mice levels in peripheral organs. However, only the mice treated with lethal irradiation showed significant cognitive function improvement as well as IDS elevation and GAG reduction in the brain. These results suggest that an efficient engraftment of genetically modified cells for HSC-GT requires strong preconditioning to ameliorate CNS involvement in cases with MPS II.
Collapse
Affiliation(s)
- Saori Miwa
- Division of Gene Therapy, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan; Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Ayako M Watabe
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Yohta Shimada
- Division of Gene Therapy, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Higuchi
- Division of Gene Therapy, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroshi Kobayashi
- Division of Gene Therapy, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan; Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Takahiro Fukuda
- Division of Neuropathology, Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Fusao Kato
- Division of Neuroscience, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroyuki Ida
- Division of Gene Therapy, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan; Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Toya Ohashi
- Division of Gene Therapy, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan; Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
21
|
Parini R, Deodato F. Intravenous Enzyme Replacement Therapy in Mucopolysaccharidoses: Clinical Effectiveness and Limitations. Int J Mol Sci 2020; 21:E2975. [PMID: 32340185 PMCID: PMC7215308 DOI: 10.3390/ijms21082975] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/13/2020] [Accepted: 04/21/2020] [Indexed: 12/21/2022] Open
Abstract
The aim of this review is to summarize the evidence on efficacy, effectiveness and safety of intravenous enzyme replacement therapy (ERT) available for mucopolysaccharidoses (MPSs) I, II, IVA, VI and VII, gained in phase III clinical trials and in observational post-approval studies. Post-marketing data are sometimes conflicting or controversial, possibly depending on disease severity, differently involved organs, age at starting treatment, and development of anti-drug antibodies (ADAs). There is general agreement that ERT is effective in reducing urinary glycosaminoglycans and liver and spleen volume, while heart and joints outcomes are variable in different studies. Effectiveness on cardiac valves, trachea and bronchi, hearing and eyes is definitely poor, probably due to limited penetration in the specific tissues. ERT does not cross the blood-brain barrier, with the consequence that the central nervous system is not cured by intravenously injected ERT. All patients develop ADAs but their role in ERT tolerance and effectiveness has not been well defined yet. Lack of reliable biomarkers contributes to the uncertainties about effectiveness. The data obtained from affected siblings strongly indicates the need of neonatal screening for treatable MPSs. Currently, other treatments are under evaluation and will surely help improve the prognosis of MPS patients.
Collapse
Affiliation(s)
- Rossella Parini
- UOS Malattie Metaboliche Rare, Clinica Pediatrica dell’Università Milano Bicocca, Fondazione MBBM, ATS Monza e Brianza, 20900 Monza, Italy
| | - Federica Deodato
- Division of Metabolic Disease, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| |
Collapse
|
22
|
Assessment of Activity of Daily Life in Mucopolysaccharidosis Type II Patients with Hematopoietic Stem Cell Transplantation. Diagnostics (Basel) 2020; 10:diagnostics10010046. [PMID: 31963134 PMCID: PMC7168225 DOI: 10.3390/diagnostics10010046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/03/2020] [Accepted: 01/08/2020] [Indexed: 12/04/2022] Open
Abstract
The effectiveness of hematopoietic stem cell transplantation (HSCT) for mucopolysaccharidosis type II (MPS II, Hunter disease) remains controversial although recent studies have shown HSCT provides more clinical impact. This study aims to evaluate the long-term effectiveness of HSCT using the activity of daily living (ADL) scores in patients with MPS II. Sixty-nine severely affected MPS II patients (19 patients who received HSCT and 50 untreated patients) and 40 attenuated affected patients (five with HSCT and 35 untreated) were investigated by a simplified ADL questionnaire. The frequency of clinical findings and the scores of ADL (verbal, gross motor, and the level of care) were analyzed statistically. The mean age of onset of 19 severely affected patients who received HSCT was 1.40 years ± 1.06, which is not statistically different from that of 50 untreated patients (p = 0.11). Macroglossia, frequent airway infection, hepatosplenomegaly, joint contracture, and sleep apnea were less frequent in the HSCT-treated group of severe MPS II patients. The severe phenotype HSCT treated group reported a statistically significant higher score of verbal function and gross motor function between the ages of 10 and 15 years and a higher level of care score between 10 and 20 years. Patients with the attenuated phenotype showed high ADL scores, and all of five HSCT treated patients reported a lower frequency of frequent airway infection, coarse skin, umbilical/inguinal hernia, hepatosplenomegaly, heart valve disorders, and carpal tunnel. In conclusion, HSCT is effective, resulting in improvements in clinical features and ADL in patients with MPS II. HSCT should be re-reviewed as a therapeutic option for MPS II patients.
Collapse
|
23
|
Safety Study of Sodium Pentosan Polysulfate for Adult Patients with Mucopolysaccharidosis Type II. Diagnostics (Basel) 2019; 9:diagnostics9040226. [PMID: 31861164 PMCID: PMC6963688 DOI: 10.3390/diagnostics9040226] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/07/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022] Open
Abstract
Current therapies for the mucopolysaccharidoses (MPS) do not effectively address skeletal and neurological manifestations. Pentosan polysulfate (PPS) is an alternative treatment strategy that has been shown to improve bone architecture, mobility, and neuroinflammation in MPS animals. The aims of this study were to a) primarily establish the safety of weekly PPS injections in attenuated MPS II, b) assess the efficacy of treatment on MPS pathology, and c) define appropriate clinical endpoints and biomarkers for future clinical trials. Subcutaneous injections were administered to three male Japanese patients for 12 weeks. Enzyme replacement therapy was continued in two of the patients while they received PPS and halted for two months in one patient before starting PPS. During treatment, one patient experienced an elevation of alanine transaminase, and another patient experienced convulsions; however, these incidences were non-cumulative and unrelated to PPS administration, respectively. Overall, the drug was well-tolerated in all patients, and no serious drug-related adverse events were noted. Generally, PPS treatment led to an increase in several parameters of shoulder range of motion and decrease of the inflammatory cytokines, MIF and TNF-α, which are potential clinical endpoints and biomarkers, respectively. Changes in urine and serum glycosaminoglycans were inconclusive. Overall, this study demonstrates the safety of using PPS in adults with MPS II and suggests the efficacy of PPS on MPS pathology with the identification of potential clinical endpoints and biomarkers.
Collapse
|
24
|
Dubot P, Sabourdy F, Plat G, Jubert C, Cancès C, Broué P, Touati G, Levade T. First Report of a Patient with MPS Type VII, Due to Novel Mutations in GUSB, Who Underwent Enzyme Replacement and Then Hematopoietic Stem Cell Transplantation. Int J Mol Sci 2019; 20:ijms20215345. [PMID: 31661765 PMCID: PMC6861985 DOI: 10.3390/ijms20215345] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 11/25/2022] Open
Abstract
We report the case of a boy who was diagnosed with mucopolysaccharidosis (MPS) VII at two weeks of age. He harbored three missense β-glucuronidase (GUSB) variations in exon 3: two novel, c.422A>C and c.424C>T, inherited from his mother, and the rather common c.526C>T, inherited from his father. Expression of these variations in transfected HEK293T cells demonstrated that the double mutation c.422A>C;424C>T reduces β-glucuronidase enzyme activity. Enzyme replacement therapy (ERT), using UX003 (vestronidase alfa), was started at four months of age, followed by a hematopoietic stem cell allograft transplantation (HSCT) at 13 months of age. ERT was well tolerated and attenuated visceromegaly and skin infiltration. After a severe skin and gut graft-versus-host disease, ERT was stopped six months after HSCT. The last follow-up examination (at the age of four years) revealed a normal psychomotor development, stabilized growth curve, no hepatosplenomegaly, and no other organ involvement. Intriguingly, enzyme activity had normalized in leukocytes but remained low in plasma. This case report illustrates: (i) The need for an early diagnosis of MPS, and (ii) the possible benefit of a very early enzymatic and/or cellular therapy in this rare form of lysosomal storage disease.
Collapse
Affiliation(s)
- Patricia Dubot
- Laboratoire de Biochimie Métabolique, Centre de Référence en Maladies Héréditaires du Métabolisme, Institut Fédératif de Biologie, CHU de Toulouse, 31059 Toulouse cedex 9, France.
- INSERM UMR1037, CRCT (Cancer Research Center of Toulouse), Université Paul Sabatier, 31037 Toulouse, France.
| | - Frédérique Sabourdy
- Laboratoire de Biochimie Métabolique, Centre de Référence en Maladies Héréditaires du Métabolisme, Institut Fédératif de Biologie, CHU de Toulouse, 31059 Toulouse cedex 9, France.
- INSERM UMR1037, CRCT (Cancer Research Center of Toulouse), Université Paul Sabatier, 31037 Toulouse, France.
| | - Geneviève Plat
- Service d'Hématologie Pédiatrique, CHU de Toulouse, 31058 Toulouse, France.
| | - Charlotte Jubert
- Service d'Hématologie Pédiatrique, CHU de Bordeaux, 33076 Bordeaux, France.
| | - Claude Cancès
- Hôpital des Enfants, Centre de Référence en Maladies Héréditaires du Métabolisme, CHU de Toulouse, 31059 Toulouse, France.
| | - Pierre Broué
- Hôpital des Enfants, Centre de Référence en Maladies Héréditaires du Métabolisme, CHU de Toulouse, 31059 Toulouse, France.
| | - Guy Touati
- Hôpital des Enfants, Centre de Référence en Maladies Héréditaires du Métabolisme, CHU de Toulouse, 31059 Toulouse, France.
| | - Thierry Levade
- Laboratoire de Biochimie Métabolique, Centre de Référence en Maladies Héréditaires du Métabolisme, Institut Fédératif de Biologie, CHU de Toulouse, 31059 Toulouse cedex 9, France.
- INSERM UMR1037, CRCT (Cancer Research Center of Toulouse), Université Paul Sabatier, 31037 Toulouse, France.
| |
Collapse
|
25
|
Taylor M, Khan S, Stapleton M, Wang J, Chen J, Wynn R, Yabe H, Chinen Y, Boelens JJ, Mason RW, Kubaski F, Horovitz DDG, Barth AL, Serafini M, Bernardo ME, Kobayashi H, Orii KE, Suzuki Y, Orii T, Tomatsu S. Hematopoietic Stem Cell Transplantation for Mucopolysaccharidoses: Past, Present, and Future. Biol Blood Marrow Transplant 2019; 25:e226-e246. [PMID: 30772512 PMCID: PMC6615945 DOI: 10.1016/j.bbmt.2019.02.012] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/11/2019] [Indexed: 12/16/2022]
Abstract
Allogenic hematopoietic stem cell transplantation (HSCT) has proven to be a viable treatment option for a selected group of patients with mucopolysaccharidoses (MPS), including those with MPS types I, II, IVA, VI, and VII. Early diagnosis and timely referral to an expert in MPS are critical, followed by a complete examination and evaluation by a multidisciplinary team, including a transplantation physician. Treatment recommendations for MPS are based on multiple biological, sociological, and financial factors, including type of MPS, clinical severity, prognosis, present clinical signs and symptoms (disease stage), age at onset, rate of progression, family factors and expectations, financial burden, feasibility, availability, risks and benefits of available therapies such as HSCT, enzyme replacement therapy (ERT), surgical interventions, and other supportive care. International collaboration and data review are critical to evaluating the therapeutic efficacy and adverse effects of HSCT for MPS. Collaborative efforts to assess HSCT for MPS have been ongoing since the first attempt at HSCT in a patient with MPS reported in 1981. The accumulation of data since then has made it possible to identify early outcomes (ie, transplantation outcomes) and long-term disease-specific outcomes resulting from HSCT. The recent identification of predictive factors and the development of innovative regimens have significantly improved the outcomes of both engraftment failure and transplantation-related mortality. Assessment of long-term outcomes has considered a variety of factors, including type of MPS, type of graft, age at transplantation, and stage of disease progression, among others. Studies on long-term outcomes are considered a key factor in the use of HSCT in patients with MPS. These studies have shown the effects and limitations of HSCT on improving disease manifestations and quality of life. In this review, we summarize the efficacy, side effects, risks, and cost of HSCT for each type of MPS.
Collapse
Affiliation(s)
- Madeleine Taylor
- Department of Biomedical, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware; Deparment of Biological Science, University of Delaware, Newark, Delaware
| | - Shaukat Khan
- Department of Biomedical, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Molly Stapleton
- Department of Biomedical, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware; Deparment of Biological Science, University of Delaware, Newark, Delaware
| | - Jianmin Wang
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Chen
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Robert Wynn
- Department of Paediatric Haematology and Cell Therapy, University of Manchester, Manchester, United Kingdom
| | - Hiromasa Yabe
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Yasutsugu Chinen
- Department of Pediatrics, Faculty of Medicine, University of the Ryukyus, Nishihara, Japan
| | - Jaap Jan Boelens
- Stem Cell Transplantation and Cellular Therapies, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert W Mason
- Department of Biomedical, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware; Deparment of Biological Science, University of Delaware, Newark, Delaware
| | - Francyne Kubaski
- Medical Genetics Service, Hospital de ClÃnicas de Porto Alegre (HCPA), Department of Genetics and Molecular Biology- Program Partnership Graduate in Genetics and Molecular Biology (PPGBM), Federal University of Rio Grande do Sul (UFRGS), and National Institute of Populational Medical Genetics (INAGEMP), Porto Alegre, Brazil
| | - Dafne D G Horovitz
- Medical Genetics Department, National Institute of Women, Children, and Adolescent Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Anneliese L Barth
- Medical Genetics Department, National Institute of Women, Children, and Adolescent Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Marta Serafini
- Department of Pediatrics, Dulbecco Telethon Institute, University of Milano-Bicocca, Monza, Italy
| | - Maria Ester Bernardo
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele-Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Hironori Kobayashi
- Department of Pediatrics, Shimane University Faculty of Medicine, Shimane, Japan
| | - Kenji E Orii
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Yasuyuki Suzuki
- Medical Education Development Center, Gifu University, Gifu, Japan
| | - Tadao Orii
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Department of Biomedical, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware; Department of Pediatrics, Shimane University Faculty of Medicine, Shimane, Japan; Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan; Department of Pediatrics, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
26
|
Stapleton M, Arunkumar N, Kubaski F, Mason RW, Tadao O, Tomatsu S. Clinical presentation and diagnosis of mucopolysaccharidoses. Mol Genet Metab 2018; 125:4-17. [PMID: 30057281 DOI: 10.1016/j.ymgme.2018.01.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 01/04/2018] [Indexed: 01/09/2023]
Abstract
Mucopolysaccharidoses (MPS) are estimated to affect1 in 25,000 live births although specific rates vary between the ethnic origin and country. MPS are a group of lysosomal storage disorders, which cause the buildup of GAG(s) due to insufficient or absent GAG-degrading enzymes. With seven types of MPS disorders and eleven subtypes, the MPS family presents unique challenges for early clinical diagnosis due to the molecular and clinical heterogeneity between groups and patients. Novel methods of early identification, particularly newborn screening through mass spectrometry, can change the flow of diagnosis, allowing enzyme and GAG quantification before the presentation of clinical symptoms improving outcomes. Genetic testing of patients and their families can also be conducted preemptively. This testing enables families to make informed decisions about family planning, leading to prenatal diagnosis. In this review, we discuss the clinical symptoms of each MPS type as they initially appear in patients, biochemical and molecular diagnostic methods, and the future of newborn screening for this group of disorders.
Collapse
Affiliation(s)
- Molly Stapleton
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States; Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Nivethitha Arunkumar
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States; Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Francyne Kubaski
- Department of Molecular Biology and Genetics, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Robert W Mason
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States; Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Orii Tadao
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States; Department of Biological Sciences, University of Delaware, Newark, DE, United States; Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan; Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pediatrics, Shimane University, Shimane, Japan.
| |
Collapse
|
27
|
Barth AL, Horovitz DDG. Hematopoietic Stem Cell Transplantation in Mucopolysaccharidosis Type II. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2018. [DOI: 10.1177/2326409818779097] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Anneliese L. Barth
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira/Fiocruz – Rio de Janeiro, Brazil
| | - Dafne D. G. Horovitz
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira/Fiocruz – Rio de Janeiro, Brazil
| |
Collapse
|
28
|
Chuang CK, Lin HY, Wang TJ, Huang YH, Chan MJ, Liao HC, Lo YT, Wang LY, Tu RY, Fang YY, Chen TL, Ho HC, Chiang CC, Lin SP. Status of newborn screening and follow up investigations for Mucopolysaccharidoses I and II in Taiwan. Orphanet J Rare Dis 2018; 13:84. [PMID: 29801497 PMCID: PMC5970538 DOI: 10.1186/s13023-018-0816-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/26/2018] [Indexed: 11/24/2022] Open
Abstract
Background Mucopolysaccharidoses (MPS) are lysosomal storage diseases in which mutations of genes encoding for lysosomal enzymes cause defects in the degradation of glycosaminoglycans (GAGs). The accumulation of GAGs in lysosomes results in cellular dysfunction and clinical abnormalities. The early initiation of enzyme replacement therapy (ERT) can slow or prevent the development of severe clinical manifestations. MPS I and II newborn screening has been available in Taiwan since August 2015. Infants who failed the recheck at recall were referred to MacKay Memorial Hospital for a detailed confirmatory diagnosis. Methods From August 2015 to November 2017, 294,196 and 153,032 infants were screened using tandem mass spectrometry for MPS I and MPS II, respectively. Of these infants, 84 suspected cases (eight for MPS I; 76 for MPS II) were referred for confirmation. Urinary first-line biochemistry examinations were performed first, including urinary GAG quantification, two-dimensional electrophoresis, and tandem mass spectrometry assay for predominant disaccharides derived from GAGs. If the results were positive, a confirmative diagnosis was made according to the results of leukocyte enzymatic assay and molecular DNA analysis. Leukocyte pellets were isolated from EDTA blood and used for fluorescent α-iduronidase (IDUA) or iduronate-2-sulfatase (IDS) enzymatic assay. DNA sequencing analysis was also performed. Results Normal IDS and IDUA enzyme activities were found in most of the referred cases except for four who were strongly suspected of having MPS I and three who were strongly suspected of having MPS II. Of these infants, three with novel mutations of the IDS gene (c.817C > T, c.1025A > G, and c.311A > T) and four with two missense mutations of the IDUA gene (C.300-3C > G, c.1874A > C; c.1037 T > G, c.1091C > T) showed significant deficiencies in IDS and IDUA enzyme activities (< 5% of mean normal activity), respectively. Urinary dermatan sulfate and heparan sulfate quantitative analyses by tandem mass spectrometry also demonstrated significant elevations. The prevalence rates of MPS I and MPS II in Taiwan were 1.35 and 1.96 per 100,000 live births, respectively. Conclusions The early initiation of ERT for MPS can result in better clinical outcomes. An early confirmatory diagnosis increases the probability of receiving appropriate medical care such as ERT quickly enough to avoid irreversible manifestations. All high risk infants identified in this study so far remain asymptomatic and are presumed to be affected with the attenuated disease variants.
Collapse
Affiliation(s)
- Chih-Kuang Chuang
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.,College of Medicine, Fu-Jen Catholic University, Taipei, Taiwan
| | - Hsiang-Yu Lin
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan.,The Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Early Childhood Care and Education, Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan.,Department of Medicine, MacKay Medical College, Taipei, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Tuan-Jen Wang
- Department of Laboratory Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - You-Hsin Huang
- The Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
| | - Min-Ju Chan
- The Chinese Foundation of Health, Neonatal Screening Center, Taipei, Taiwan
| | - Hsuan-Chieh Liao
- The Chinese Foundation of Health, Neonatal Screening Center, Taipei, Taiwan
| | - Yun-Ting Lo
- Department of Laboratory Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Li-Yun Wang
- Taipei Institute of Pathology, Neonatal Screening Center, Taipei, Taiwan
| | - Ru-Yi Tu
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yi-Ya Fang
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Tzu-Lin Chen
- Department of Laboratory Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Hui-Chen Ho
- Taipei Institute of Pathology, Neonatal Screening Center, Taipei, Taiwan
| | - Chuan-Chi Chiang
- The Chinese Foundation of Health, Neonatal Screening Center, Taipei, Taiwan.
| | - Shuan-Pei Lin
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan. .,Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan. .,The Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan. .,Department of Early Childhood Care and Education, Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan. .,Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan. .,Departments of Pediatrics and Medical Research, MacKay Memorial Hospital, No. 92, Sec. 2, Chung-Shan N. Rd, Taipei, 10449, Taiwan.
| |
Collapse
|
29
|
Selvanathan A, Ellaway C, Wilson C, Owens P, Shaw PJ, Bhattacharya K. Effectiveness of Early Hematopoietic Stem Cell Transplantation in Preventing Neurocognitive Decline in Mucopolysaccharidosis Type II: A Case Series. JIMD Rep 2018; 41:81-89. [PMID: 29671225 DOI: 10.1007/8904_2018_104] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 03/18/2018] [Accepted: 03/19/2018] [Indexed: 12/02/2022] Open
Abstract
The early progressive form of the X-linked disorder, Hunter syndrome or mucopolysaccharidosis type II (MPS II) (OMIM #309900), is characterized by cognitive decline, and pulmonary and cardiac complications that often cause death before 20 years of age. Deficiency of the lysosomal enzyme, iduronate-2-sulfatase (EC 3.1.6.13) results in deposition of the glycosaminoglycans, dermatan, and heparan sulfate in various tissues. In recent years, enzyme replacement therapy (ERT) has become the mainstay of treatment, but is expensive and ineffective in arresting cognitive decline. Hematopoietic stem cell transplantation (HSCT) also provides enzyme replacement, and may be effective in stabilizing neurocognitive function if initiated early, though data are limited. We present a case series of four patients who demonstrated neurocognitive stabilization with early HSCT. HSCT is a potentially underutilized treatment strategy for select groups of MPS II patients.
Collapse
Affiliation(s)
- A Selvanathan
- Genetic Metabolic Disorders Service, The Children's Hospital at Westmead, Westmead, NSW, Australia.
- Discipline of Child and Adolescent Health, The University of Sydney, Camperdown, NSW, Australia.
| | - C Ellaway
- Genetic Metabolic Disorders Service, The Children's Hospital at Westmead, Westmead, NSW, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Camperdown, NSW, Australia
| | - C Wilson
- Starship Paediatric Metabolic Service, Starship Children's Hospital, Auckland, New Zealand
| | - P Owens
- Genetic Metabolic Disorders Service, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - P J Shaw
- Discipline of Child and Adolescent Health, The University of Sydney, Camperdown, NSW, Australia
- Blood and Marrow Transplant Service, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - K Bhattacharya
- Genetic Metabolic Disorders Service, The Children's Hospital at Westmead, Westmead, NSW, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
30
|
Nagao K, Morlet T, Haley E, Padilla J, Nemith J, Mason RW, Tomatsu S. Neurophysiology of hearing in patients with mucopolysaccharidosis type IV. Mol Genet Metab 2018; 123:472-478. [PMID: 29472067 PMCID: PMC5891367 DOI: 10.1016/j.ymgme.2018.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 02/05/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Hearing impairment is a common problem in patients with mucopolysaccharidosis IV (MPS IV) throughout their life. Many of the adult patients with MPS IV exhibit permanent or severe hearing loss. However, there has been no systematic review of detailed audiological test results in MPS IV. MATERIALS AND METHODS Fourteen individuals with MPS IV (13 MPS IVA and 1 MPS IVB; aged between 12 and 38 years old) participated in the current study. We obtained auditory neurophysiological responses (auditory brainstem responses and otoacoustic emissions test) in addition to pure-tone audiometry and middle ear function tests (tympanometry and acoustic reflexes). RESULTS The results indicated various levels and types of hearing loss with abnormal neurophysiological responses even in those patients with MPS IVA with normal pure tone thresholds. We also found a strong relationship between height (short stature is an indicator of skeletal severity) and hearing sensitivity as well as a strong relationship between height and outer hair cell function in the inner ear (measured by otoacoustic emissions) among MPS IVA patients. CONCLUSION The strong correlation between reduced height and hearing loss indicates that patients with severe skeletal dysplasia may be at higher risk of developing more severe hearing loss. More importantly, the spectrum of hearing disorders indicates that MPS IV patients should have annual neurophysiological hearing tests in addition to audiometric testing from an early age regardless of their skeletal severity to more carefully monitor disease progression.
Collapse
Affiliation(s)
- Kyoko Nagao
- Department of Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE, United States; Department of Linguistics and Cognitive Science, University of Delaware, Newark, DE, United States; College of Health Sciences, University of Delaware, Newark, DE, United States.
| | - Thierry Morlet
- Department of Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE, United States; Department of Linguistics and Cognitive Science, University of Delaware, Newark, DE, United States; Georges Osborne College of Audiology, Salus University, Elkins Park, PA, United States
| | - Elizabeth Haley
- Department of Communication Science & Disorders, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Julianne Nemith
- College of Health Sciences, University of Delaware, Newark, DE, United States
| | - Robert W Mason
- Department of Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Shunji Tomatsu
- Department of Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE, United States; Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
31
|
Chuang CK, Lin HY, Wang TJ, Huang SF, Lin SP. Bio-Plex immunoassay measuring the quantity of lysosomal N-acetylgalactosamine-6-sulfatase protein in dried blood spots for the screening of mucopolysaccharidosis IVA in newborn: a pilot study. BMJ Open 2017; 7:e014410. [PMID: 28710204 PMCID: PMC5734244 DOI: 10.1136/bmjopen-2016-014410] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Mucopolysaccharidosis (MPS) IVA (Morquio syndrome A) is an autosomal-recessive lysosomal storage disorder caused by the deficiency of N-acetylgalactosamine-6-sulfatase (GALNS) resulting in excessive lysosomal storage of keratan sulfate. Treatments for MPS IVA have recently become available with optimal outcomes associated with early diagnosis and treatment which can be achieved by newborn screening. DESIGN Newborn screening programme for MPS IVA pilot study. SETTING MacKay Memorial Hospital (MMH), Taipei and another three branch hospitals in Taiwan. PARTICIPANTS A total of 7415 newborns were born in four branch hospitals of MMH and had joined the MPS IVA newborn screening programme. Written informed consents were obtained from parents prior to the screening process (12MMHIS188 approved by MacKay Memorial Hospital Institutional Review Board). OUTCOME MEASURES An alternative newborn screening method for MPS IVA has been performed. Screening involved measuring the quantity of GALNS in dried blood spot (DBS) from newborn infants using the Bio-Plex immunoassay. The amount of fluorescence sorting detected by yttrium aluminium garnet laser was proportional to the quantity of GALNS protein. RESULTS Of the 7415 neonates analysed, eight infants whose GALNS levels were below the cut-off value of 8.30 µg/L had been recalled for a second DBS collection. The reference values were 8.30-27.43 µg/L. In patients with confirmed MPS IVA (n=11), the GALNS quantities were far below 5% of the normal population. CONCLUSION The Bio-Plex immunoassay is a validated method used for measuring GALNS protein in DBS and has the potential to be adopted for MPS IVA newborn screening study design.
Collapse
Affiliation(s)
- Chih-Kuang Chuang
- Division of Genetics and Metabolism, Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
- College of Medicine, Fu-Jen Catholic University, Taipei, Taiwan
- Departmentof Chemical Engineering and Biotechnology, Institute of Chemical Engineering. National Taipei University of Technology, Taipei, Taiwan., Taipei, Taiwan
| | - Hsiang-Yu Lin
- Division of Genetics and Metabolism, Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
- Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan
- Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Tuan-Jen Wang
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Sung-Fa Huang
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Shuan-Pei Lin
- Division of Genetics and Metabolism, Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
- Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Department of Infant and Child Care, National Taipei University of Nursing and Health Science, Taipei, Taiwan
| |
Collapse
|
32
|
Kubaski F, Yabe H, Suzuki Y, Seto T, Hamazaki T, Mason RW, Xie L, Onsten TGH, Leistner-Segal S, Giugliani R, Dũng VC, Ngoc CTB, Yamaguchi S, Montaño AM, Orii KE, Fukao T, Shintaku H, Orii T, Tomatsu S. Hematopoietic Stem Cell Transplantation for Patients with Mucopolysaccharidosis II. Biol Blood Marrow Transplant 2017; 23:1795-1803. [PMID: 28673849 DOI: 10.1016/j.bbmt.2017.06.020] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/26/2017] [Indexed: 10/19/2022]
Abstract
There is limited information regarding the long-term outcomes of hematopoietic stem cell transplantation (HSCT) for mucopolysaccharidosis II (MPS II). In this study, clinical, biochemical, and radiologic findings were assessed in patients who underwent HSCT and/or enzyme replacement therapy (ERT). Demographic data for 146 HSCT patients were collected from 27 new cases and 119 published cases and were compared with 51 ERT and 15 untreated cases. Glycosaminoglycan (GAG) levels were analyzed by liquid chromatography tandem mass spectrometry in blood samples from HSCT, ERT, and untreated patients as well as age-matched controls. Long-term magnetic resonance imaging (MRI) findings were investigated in 13 treated patients (6 ERT and 7 HSCT). Mean age at HSCT was 5.5 years (range, 2 to 21.4 years) in new patients and 5.5 years (range, 10 months to 19.8 years) in published cases. None of the 27 new patients died as a direct result of the HSCT procedure. Graft-versus-host disease occurred in 8 (9%) out of 85 published cases, and 9 (8%) patients died from transplantation-associated complications. Most HSCT patients showed greater improvement in somatic features, joint movements, and activity of daily living than the ERT patients. GAG levels in blood were significantly reduced by ERT and levels were even lower after HSCT. HSCT patients showed either improvement or no progression of abnormal findings in brain MRI while abnormal findings became more extensive after ERT. HSCT seems to be more effective than ERT for MPS II in a wide range of disease manifestations and could be considered as a treatment option for this condition.
Collapse
Affiliation(s)
- Francyne Kubaski
- Department of Research, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware; Department of Biological Sciences, University of Delaware, Newark, Delaware; Instituto Nacional de Ciência e Tecnologia de Genética Médica Populacional, Porto Alegre, Brazil
| | - Hiromasa Yabe
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Yasuyuki Suzuki
- Medical Education Development Center, Gifu University, Gifu, Japan
| | - Toshiyuki Seto
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Takashi Hamazaki
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Robert W Mason
- Department of Research, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware; Department of Biological Sciences, University of Delaware, Newark, Delaware
| | - Li Xie
- Department of Research, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Tor Gunnar Hugo Onsten
- Haemotherapy Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Department of Internal Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Sandra Leistner-Segal
- Instituto Nacional de Ciência e Tecnologia de Genética Médica Populacional, Porto Alegre, Brazil; Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Postgraduate Program in Medicine, Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Roberto Giugliani
- Instituto Nacional de Ciência e Tecnologia de Genética Médica Populacional, Porto Alegre, Brazil; Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Postgraduate Program in Medicine, Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Department of Genetics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Vũ Chí Dũng
- Vietnam National Children's Hospital, Department of Medical Genetics, Metabolism and Endocrinology, Hanoi, Vietnam
| | - Can Thi Bich Ngoc
- Vietnam National Children's Hospital, Department of Medical Genetics, Metabolism and Endocrinology, Hanoi, Vietnam
| | - Seiji Yamaguchi
- Department of Pediatrics, Shimane University, Shimane, Japan
| | - Adriana M Montaño
- Department of Pediatrics, Saint Louis University, St. Louis, Missouri; Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, Missouri
| | - Kenji E Orii
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Toshiyuki Fukao
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Haruo Shintaku
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tadao Orii
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Department of Research, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware; Department of Pediatrics, Shimane University, Shimane, Japan; Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan; Department of Pediatrics, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
33
|
Barth AL, de Magalhães TSPC, Reis ABR, de Oliveira ML, Scalco FB, Cavalcanti NC, Silva DSE, Torres DA, Costa AAP, Bonfim C, Giugliani R, Llerena JC, Horovitz DDG. Early hematopoietic stem cell transplantation in a patient with severe mucopolysaccharidosis II: A 7 years follow-up. Mol Genet Metab Rep 2017. [PMID: 28649514 PMCID: PMC5470531 DOI: 10.1016/j.ymgmr.2017.05.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mucopolysaccharidosis type II (MPS II - Hunter syndrome) is an X-linked lysosomal storage disorder caused by a deficiency in the enzyme iduronate-2 sulfatase (I2S), leading to the accumulation of the glycosaminoglycans, affecting multiple organs and systems. Enzyme replacement therapy does not cross the blood brain barrier, limiting results in neurological forms of the disease. Another option of treatment for severe MPS, hematopoietic stem cell transplantation (HSCT) has become the treatment of choice for the severe form of MPS type I, since it can preserve neurocognition when performed early in the course of the disease. To date, only few studies have examined the long-term outcomes of HSCT in patients with MPS II. We describe the seven-year follow-up of a prenatally diagnosed MPS II boy with positive family history of severe MPS form, submitted to HSCT with umbilical cord blood cells at 70 days of age. Engraftment after 30 days revealed mixed chimerism with 79% donor cells; after 7 years engraftment remains at 80%. I2S activity 30 days post-transplant was low in plasma and normal in leukocytes and the same pattern is observed to date. At age 7 years growth charts are normal and he is very healthy, although mild signs of dysostosis multiplex are present, as well as hearing loss. The neuropsychological evaluation (Wechsler Intelligence Scale for Children - Fourth Edition - WISC-IV), disclosed an IQ of 47. Despite this low measured IQ, the patient continues to show improvements in cognitive, language and motor skills, being quite functional. We believe that HSCT is a therapeutic option for MPS II patients with the severe phenotype, as it could preserve neurocognition or even halt neurodegeneration, provided strict selection criteria are followed.
Collapse
Affiliation(s)
- Anneliese L Barth
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira - Fiocruz, Rio de Janeiro, Brazil
| | - Tatiana S P C de Magalhães
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira - Fiocruz, Rio de Janeiro, Brazil
| | - Ana Beatriz R Reis
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira - Fiocruz, Rio de Janeiro, Brazil
| | - Maria Lucia de Oliveira
- Laboratório de Erros Inatos do Metabolismo, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda B Scalco
- Laboratório de Erros Inatos do Metabolismo, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nicolette C Cavalcanti
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira - Fiocruz, Rio de Janeiro, Brazil
| | - Daniel S E Silva
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira - Fiocruz, Rio de Janeiro, Brazil
| | - Danielle A Torres
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira - Fiocruz, Rio de Janeiro, Brazil
| | - Alessandra A P Costa
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira - Fiocruz, Rio de Janeiro, Brazil
| | - Carmem Bonfim
- Bone Marrow Transplantation Unit, Federal University of Paraná, Curitiba, Brazil
| | - Roberto Giugliani
- Medical Genetics Service, Hospital de Clinicas de Alegre, Porto Alegre, Brazil
| | - Juan C Llerena
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira - Fiocruz, Rio de Janeiro, Brazil
| | - Dafne D G Horovitz
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira - Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
34
|
Bradley LA, Haddow HRM, Palomaki GE. Treatment of mucopolysaccharidosis type II (Hunter syndrome): results from a systematic evidence review. Genet Med 2017. [PMID: 28640238 DOI: 10.1038/gim.2017.30] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
PurposeA pilot systematic evidence review to establish methodology utility in rare genetic diseases, support clinical recommendations, and identify important knowledge gaps.MethodsBroad-based published/gray-literature searches through December 2015 for studies of males with confirmed mucopolysaccharidosis type II (any age, phenotype, genotype, family history) treated with enzyme replacement therapy or hematopoietic stem cell transplantation. Preset inclusion criteria employed for abstract and full document selection, and standardized methods for data extraction and assessment of quality and strength of evidence.ResultsTwelve outcomes reported included benefits of urinary glycosaminoglycan and liver/spleen volume reductions and harms of immunoglobulin G/neutralizing antibody development (moderate strength of evidence). Less clear were benefits of improved 6-minute walk tests, height, early treatment, and harms of other adverse reactions (low strength of evidence). Benefits and harms of other outcomes were unclear (insufficient strength of evidence). Current benefits and harms of hematopoietic stem cell transplantation are unclear, based on dated, low-quality studies. A critical knowledge gap is long-term outcomes. Consensus on selection of critical outcomes and measures is needed to definitively evaluate treatment safety and effectiveness.ConclusionMinor methodology modifications and a focus on critical evidence can reduce review time and resources. Summarized evidence was sufficient to support guidance development and highlight important knowledge gaps.
Collapse
Affiliation(s)
- Linda A Bradley
- Department of Pathology and Laboratory Medicine, Women & Infants Hospital/Warren Alpert, Medical School at Brown University, Providence, Rhode Island, USA
| | | | - Glenn E Palomaki
- Department of Pathology and Laboratory Medicine, Women & Infants Hospital/Warren Alpert, Medical School at Brown University, Providence, Rhode Island, USA
| |
Collapse
|
35
|
Stapleton M, Kubaski F, Mason RW, Yabe H, Suzuki Y, Orii KE, Orii T, Tomatsu S. Presentation and Treatments for Mucopolysaccharidosis Type II (MPS II; Hunter Syndrome). Expert Opin Orphan Drugs 2017; 5:295-307. [PMID: 29158997 PMCID: PMC5693349 DOI: 10.1080/21678707.2017.1296761] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/15/2017] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Mucopolysaccharidosis Type II (MPS II; Hunter syndrome) is an X- linked lysosomal storage disorder caused by a deficiency of iduronate-2-sulfatase (IDS). IDS deficiency leads to primary accumulation of dermatan sulfate (DS) and heparan sulfate (HS). MPS II is both multi-systemic and progressive. Phenotypes are classified as either attenuated or severe (based on absence or presence of central nervous system impairment, respectively). AREAS COVERED Current treatments available are intravenous enzyme replacement therapy (ERT), hematopoietic stem cell transplantation (HSCT), anti-inflammatory treatment, and palliative care with symptomatic surgeries. Clinical trials are being conducted for intrathecal ERT and gene therapy is under pre-clinical investigation. Treatment approaches differ based on age, clinical severity, prognosis, availability and feasibility of therapy, and health insurance.This review provides a historical account of MPS II treatment as well as treatment development with insights into benefits and/or limitations of each specific treatment. EXPERT OPINION Conventional ERT and HSCT coupled with surgical intervention and palliative therapy are currently the treatment options available to MPS II patients. Intrathecal ERT and gene therapy are currently under investigation as future therapies. These investigative treatments are critical to address the limitations in treatment of the central nervous system (CNS).
Collapse
Affiliation(s)
- Molly Stapleton
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Francyne Kubaski
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Robert W. Mason
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Hiromasa Yabe
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Yasuyuki Suzuki
- Medical Education Development Center, Gifu University, Gifu, Japan
| | - Kenji E. Orii
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Tadao Orii
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| |
Collapse
|
36
|
Sawamoto K, Suzuki Y, Mackenzie WG, Theroux MC, Pizarro C, Yabe H, Orii KE, Mason RW, Orii T, Tomatsu S. Current therapies for Morquio A syndrome and their clinical outcomes. Expert Opin Orphan Drugs 2016; 4:941-951. [PMID: 28217429 PMCID: PMC5312776 DOI: 10.1080/21678707.2016.1214572] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 07/15/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Morquio A syndrome is characterized by a unique skeletal dysplasia, leading to short neck and trunk, pectus carinatum, laxity of joints, kyphoscoliosis, and tracheal obstruction. Cervical spinal cord compression/inability, a restrictive and obstructive airway, and/or bone deformity and imbalance of growth, are life-threatening to Morquio A patients, leading to a high morbidity and mortality. It is critical to review the current therapeutic approaches with respect to their efficacy and limitations. AREAS COVERED Patients with progressive skeletal dysplasia often need to undergo orthopedic surgical interventions in the first two decades of life. Recently, we have treated four patients with a new surgery to correct progressive tracheal obstruction. Enzyme replacement therapy (ERT) has been approved clinically. Cell-based therapies such as hematopoietic stem cell therapy (HSCT) and gene therapy are typically one-time, permanent treatments for enzyme deficiencies. We report here on four Morquio A patients treated with HSCT approved in Japan and followed for at least ten years after treatment. Gene therapy is under investigation on mouse models but not yet available as a therapeutic option. EXPERT OPINION ERT and HSCT in combination with surgical intervention(s) are a therapeutic option for Morquio A; however, the approach for bone and cartilage lesion remains an unmet challenge.
Collapse
Affiliation(s)
- Kazuki Sawamoto
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Yasuyuki Suzuki
- Medical Education Development Center, Gifu University, Gifu, Japan
| | | | - Mary C. Theroux
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | | | - Hiromasa Yabe
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Kenji E. Orii
- Division of Neonatal Intensive Care Unit, Gifu University Hospital, Gifu, Japan
| | - Robert W. Mason
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Tadao Orii
- Department of Pediatrics, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
- Department of Pediatrics, Gifu University, Gifu, Japan
| |
Collapse
|
37
|
Schuh RS, Baldo G, Teixeira HF. Nanotechnology applied to treatment of mucopolysaccharidoses. Expert Opin Drug Deliv 2016; 13:1709-1718. [DOI: 10.1080/17425247.2016.1202235] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Roselena S. Schuh
- Programa de Pós-Graduação em Ciências Farmacêuticas da UFRGS, Faculdade de Farmácia, Porto Alegre, RS, Brazil
| | - Guilherme Baldo
- Programa de Pós-Graduação em Genética e Biologia Molecular da UFRGS, Departamento de Fisiologia, Porto Alegre, RS, Brazil
| | - Helder F. Teixeira
- Programa de Pós-Graduação em Ciências Farmacêuticas da UFRGS, Faculdade de Farmácia, Porto Alegre, RS, Brazil
| |
Collapse
|
38
|
Yasuda E, Suzuki Y, Shimada T, Sawamoto K, Mackenzie WG, Theroux MC, Pizarro C, Xie L, Miller F, Rahman T, Kecskemethy HH, Nagao K, Morlet T, Shaffer TH, Chinen Y, Yabe H, Tanaka A, Shintaku H, Orii KE, Orii KO, Mason RW, Montaño AM, Fukao T, Orii T, Tomatsu S. Activity of daily living for Morquio A syndrome. Mol Genet Metab 2016; 118:111-22. [PMID: 27161890 PMCID: PMC5016714 DOI: 10.1016/j.ymgme.2016.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/07/2016] [Indexed: 01/26/2023]
Abstract
The aim of this study was to evaluate the activity of daily living (ADL) and surgical interventions in patients with mucopolysaccharidosis IVA (MPS IVA). The factor(s) that affect ADL are age, clinical phenotypes, surgical interventions, therapeutic effect, and body mass index. The ADL questionnaire comprises three domains: "Movement," "Movement with cognition," and "Cognition." Each domain has four subcategories rated on a 5-point scale based on the level of assistance. The questionnaire was collected from 145 healthy controls and 82 patients with MPS IVA. The patient cohort consisted of 63 severe and 17 attenuated phenotypes (2 were undefined); 4 patients treated with hematopoietic stem cell transplantation (HSCT), 33 patients treated with enzyme replacement therapy (ERT) for more than a year, and 45 untreated patients. MPS IVA patients show a decline in ADL scores after 10years of age. Patients with a severe phenotype have a lower ADL score than healthy control subjects, and lower scores than patients with an attenuated phenotype in domains of "Movement" and "Movement with cognition." Patients, who underwent HSCT and were followed up for over 10years, had higher ADL scores and fewer surgical interventions than untreated patients. ADL scores for ERT patients (2.5years follow-up on average) were similar with the-age-matched controls below 10years of age, but declined in older patients. Surgical frequency was higher for severe phenotypic patients than attenuated ones. Surgical frequency for patients treated with ERT was not decreased compared to untreated patients. In conclusion, we have shown the utility of the proposed ADL questionnaire and frequency of surgical interventions in patients with MPS IVA to evaluate the clinical severity and therapeutic efficacy compared with age-matched controls.
Collapse
Affiliation(s)
- Eriko Yasuda
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Medical Informatics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yasuyuki Suzuki
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Japan
| | - Tsutomu Shimada
- Medical Education Development Center, Gifu University, Gifu, Japan
| | - Kazuki Sawamoto
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | | | - Mary C Theroux
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | | | - Li Xie
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Freeman Miller
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Tariq Rahman
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | | | - Kyoko Nagao
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Thierry Morlet
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Thomas H Shaffer
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Yasutsugu Chinen
- Department of Pediatrics, Faculty of Medicine, University of the Ryukyus, Ryukyu, Japan
| | - Hiromasa Yabe
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Akemi Tanaka
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Haruo Shintaku
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kenji E Orii
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Koji O Orii
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Robert W Mason
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Adriana M Montaño
- Department of Pediatrics, Saint Louis University, St. Louis, MO, USA; Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, MO, USA
| | - Toshiyuki Fukao
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Tadao Orii
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan.
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan.
| |
Collapse
|
39
|
Yabe H, Tanaka A, Chinen Y, Kato S, Sawamoto K, Yasuda E, Shintaku H, Suzuki Y, Orii T, Tomatsu S. Hematopoietic stem cell transplantation for Morquio A syndrome. Mol Genet Metab 2016; 117:84-94. [PMID: 26452513 PMCID: PMC5016080 DOI: 10.1016/j.ymgme.2015.09.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 09/29/2015] [Accepted: 09/29/2015] [Indexed: 12/15/2022]
Abstract
Morquio A syndrome features systemic skeletal dysplasia. To date, there has been no curative therapy for this skeletal dysplasia. No systemic report on a long-term effect of hematopoietic stem cell transplantation (HSCT) for Morquio A has been described. We conducted HSCT for 4 cases with Morquio A (age at HSCT: 4-15years, mean 10.5years) and followed them at least 10years (range 11-28years; mean 19years). Current age ranged between 25 and 36years of age (mean 29.5years). All cases had a successful full engraftment of allogeneic bone marrow transplantation without serious GVHD. Transplanted bone marrow derived from HLA-identical siblings (three cases) or HLA-identical unrelated donor. The levels of the enzyme activity in the recipient's lymphocytes reached the levels of donors' enzyme activities within two years after HSCT. For the successive over 10years post-BMT, GALNS activity in lymphocytes was maintained at the same level as the donors. Except one case who had osteotomy in both legs one year later post BMT, other three cases had no orthopedic surgical intervention. All cases remained ambulatory, and three of them could walk over 400m. Activity of daily living (ADL) in patients with HSCT was better than untreated patients. The patient who underwent HSCT at four years of age showed the best ADL score. In conclusion, the long-term study of HSCT has demonstrated therapeutic effect in amelioration of progression of the disease in respiratory function, ADL, and biochemical findings, suggesting that HSCT is a therapeutic option for patients with Morquio A.
Collapse
Affiliation(s)
- Hiromasa Yabe
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan.
| | - Akemi Tanaka
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan.
| | - Yasutsugu Chinen
- Department of Pediatrics, Faculty of Medicine, University of the Ryukyus, Japan.
| | - Shunichi Kato
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Kazuki Sawamoto
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Eriko Yasuda
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Haruo Shintaku
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yasuyuki Suzuki
- Medical Education Development Center, Gifu University, Japan
| | - Tadao Orii
- Department of Pediatrics, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Department of Pediatrics, Gifu University, Gifu, Japan; Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States.
| |
Collapse
|
40
|
Tylki-Szymańska A, Jurecka A. Prospective therapies for mucopolysaccharidoses. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1089167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
41
|
Tomatsu S, Sawamoto K, Shimada T, Bober MB, Kubaski F, Yasuda E, Mason RW, Khan S, Alméciga-Díaz CJ, Barrera LA, Mackenzie WG, Orii T. Enzyme replacement therapy for treating mucopolysaccharidosis type IVA (Morquio A syndrome): effect and limitations. Expert Opin Orphan Drugs 2015; 3:1279-1290. [PMID: 26973801 PMCID: PMC4788508 DOI: 10.1517/21678707.2015.1086640] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Following a Phase III, randomized, double-blind, placebo (PBO)-controlled, multinational study in subjects with mucopolysaccharidosis IVA (MPS IVA), enzyme replacement therapy (ERT) of elosulfase alfa has been approved in several countries. The study was designed to evaluate safety and efficacy of elosulfase alfa in patients with MPS IVA aged 5 years and older. AREAS COVERED Outcomes of clinical trials for MPS IVA have been described. Subjects received either 2.0 mg/kg/week, 2.0 mg/kg/every other week, or PBO, for 24 weeks. The primary endpoint was the change from baseline 6-min walk test (6MWT) distance compared to PBO. The 6MWT results improved in patients receiving 2 mg/kg weekly compared to PBO. The every other week regimen resulted in walk distances comparable to PBO. There was no change from baseline in the 3 Min Stair Climb Test in both treatment groups. Following completion of the initial study, patients, who continued to receive elosulfase alfa 2 mg/kg weekly (QW) for another 48 weeks (for a total of up to 72-week exposure), did not show additional improvement on 6MWT. EXPERT OPINION We suggest that ERT is a therapeutic option for MPS IVA, providing a modest effect and the majority of the effects are seen in the soft tissues.
Collapse
Affiliation(s)
- Shunji Tomatsu
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Rd., Wilmington, DE 19899-0269, USA
- Department of Pediatrics, Gifu University, Gifu, Japan, Japan
| | - Kazuki Sawamoto
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Rd., Wilmington, DE 19899-0269, USA
| | - Tsutomu Shimada
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Rd., Wilmington, DE 19899-0269, USA
- Department of Medical Informatics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Michael B. Bober
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Rd., Wilmington, DE 19899-0269, USA
| | - Francyne Kubaski
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Rd., Wilmington, DE 19899-0269, USA
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Eriko Yasuda
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Rd., Wilmington, DE 19899-0269, USA
- Department of Medical Informatics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Robert W. Mason
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Rd., Wilmington, DE 19899-0269, USA
| | - Shaukat Khan
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Rd., Wilmington, DE 19899-0269, USA
| | - Carlos J. Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, School of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Luis A. Barrera
- Institute for the Study of Inborn Errors of Metabolism, School of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - William G. Mackenzie
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Rd., Wilmington, DE 19899-0269, USA
| | - Tadao Orii
- Department of Pediatrics, Gifu University, Gifu, Japan, Japan
| |
Collapse
|
42
|
Tomatsu S, Sawamoto K, Alméciga-Díaz CJ, Shimada T, Bober MB, Chinen Y, Yabe H, Montaño AM, Giugliani R, Kubaski F, Yasuda E, Rodríguez-López A, Espejo-Mojica AJ, Sánchez OF, Mason RW, Barrera LA, Mackenzie WG, Orii T. Impact of enzyme replacement therapy and hematopoietic stem cell transplantation in patients with Morquio A syndrome. Drug Des Devel Ther 2015; 9:1937-53. [PMID: 25897204 PMCID: PMC4389814 DOI: 10.2147/dddt.s68562] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Patients with mucopolysaccharidosis IVA (MPS IVA) can present with systemic skeletal dysplasia, leading to a need for multiple orthopedic surgical procedures, and often become wheelchair bound in their teenage years. Studies on patients with MPS IVA treated by enzyme replacement therapy (ERT) showed a sharp reduction on urinary keratan sulfate, but only modest improvement based on a 6-minute walk test and no significant improvement on a 3-minute climb-up test and lung function test compared with the placebo group, at least in the short-term. Surgical remnants from ERT-treated patients did not show reduction of storage materials in chondrocytes. The impact of ERT on bone lesions in patients with MPS IVA remains limited. ERT seems to be enhanced in a mouse model of MPS IVA by a novel form of the enzyme tagged with a bone-targeting moiety. The tagged enzyme remained in the circulation much longer than untagged native enzyme and was delivered to and retained in bone. Three-month-old MPS IVA mice treated with 23 weekly infusions of tagged enzyme showed marked clearance of the storage materials in bone, bone marrow, and heart valves. When treatment was initiated at birth, reduction of storage materials in tissues was even greater. These findings indicate that specific targeting of the enzyme to bone at an early stage may improve efficacy of ERT for MPS IVA. Recombinant N-acetylgalactosamine-6-sulfate sulfatase (GALNS) in Escherichia coli BL21 (DE3) (erGALNS) and in the methylotrophic yeast Pichia pastoris (prGALNS) has been produced as an alternative to the conventional production in Chinese hamster ovary cells. Recombinant GALNS produced in microorganisms may help to reduce the high cost of ERT and the introduction of modifications to enhance targeting. Although only a limited number of patients with MPS IVA have been treated with hematopoietic stem cell transplantation (HSCT), beneficial effects have been reported. A wheelchair-bound patient with a severe form of MPS IVA was treated with HSCT at 15 years of age and followed up for 10 years. Radiographs showed that the figures of major and minor trochanter appeared. Loud snoring and apnea disappeared. In all, 1 year after bone marrow transplantation, bone mineral density at L2-L4 was increased from 0.372 g/cm(2) to 0.548 g/cm(2) and was maintained at a level of 0.48±0.054 for the following 9 years. Pulmonary vital capacity increased approximately 20% from a baseline of 1.08 L to around 1.31 L over the first 2 years and was maintained thereafter. Activity of daily living was improved similar to the normal control group. After bilateral osteotomies, a patient can walk over 400 m using hip-knee-ankle-foot orthoses. This long-term observation of a patient shows that this treatment can produce clinical improvements although bone deformity remained unchanged. In conclusion, ERT is a therapeutic option for MPS IVA patients, and there are some indications that HSCT may be an alternative to treat this disease. However, as neither seems to be a curative therapy, at least for the skeletal dysplasia in MPS IVA patients, new approaches are investigated to enhance efficacy and reduce costs to benefit MPS IVA patients.
Collapse
Affiliation(s)
- Shunji Tomatsu
- Nemours/Alfred I duPont Hospital for Children, Wilmington, DE, USA
- Department of Pediatrics, Gifu University, Gifu, Japan
| | - Kazuki Sawamoto
- Nemours/Alfred I duPont Hospital for Children, Wilmington, DE, USA
| | - Carlos J Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, School of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Tsutomu Shimada
- Nemours/Alfred I duPont Hospital for Children, Wilmington, DE, USA
| | - Michael B Bober
- Nemours/Alfred I duPont Hospital for Children, Wilmington, DE, USA
| | - Yasutsugu Chinen
- Department of Pediatrics, Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Hiromasa Yabe
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| | | | - Roberto Giugliani
- Medical Genetics Service/HCPA and Department of Genetics/UFRGS, Porto Alegre, Brazil
| | - Francyne Kubaski
- Nemours/Alfred I duPont Hospital for Children, Wilmington, DE, USA
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Eriko Yasuda
- Nemours/Alfred I duPont Hospital for Children, Wilmington, DE, USA
| | - Alexander Rodríguez-López
- Institute for the Study of Inborn Errors of Metabolism, School of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Angela J Espejo-Mojica
- Institute for the Study of Inborn Errors of Metabolism, School of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Oscar F Sánchez
- School of Chemical Engineering, Purdue University, West Lafayette, IN, USA
| | - Robert W Mason
- Nemours/Alfred I duPont Hospital for Children, Wilmington, DE, USA
| | - Luis A Barrera
- Institute for the Study of Inborn Errors of Metabolism, School of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Tadao Orii
- Department of Pediatrics, Gifu University, Gifu, Japan
| |
Collapse
|
43
|
Tomatsu S, Alméciga-Díaz CJ, Montaño AM, Yabe H, Tanaka A, Dung VC, Giugliani R, Kubaski F, Mason RW, Yasuda E, Sawamoto K, Mackenzie W, Suzuki Y, Orii KE, Barrera LA, Sly WS, Orii T. Therapies for the bone in mucopolysaccharidoses. Mol Genet Metab 2015; 114:94-109. [PMID: 25537451 PMCID: PMC4312706 DOI: 10.1016/j.ymgme.2014.12.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 11/30/2014] [Accepted: 12/01/2014] [Indexed: 12/24/2022]
Abstract
Patients with mucopolysaccharidoses (MPS) have accumulation of glycosaminoglycans in multiple tissues which may cause coarse facial features, mental retardation, recurrent ear and nose infections, inguinal and umbilical hernias, hepatosplenomegaly, and skeletal deformities. Clinical features related to bone lesions may include marked short stature, cervical stenosis, pectus carinatum, small lungs, joint rigidity (but laxity for MPS IV), kyphoscoliosis, lumbar gibbus, and genu valgum. Patients with MPS are often wheelchair-bound and physical handicaps increase with age as a result of progressive skeletal dysplasia, abnormal joint mobility, and osteoarthritis, leading to 1) stenosis of the upper cervical region, 2) restrictive small lung, 3) hip dysplasia, 4) restriction of joint movement, and 5) surgical complications. Patients often need multiple orthopedic procedures including cervical decompression and fusion, carpal tunnel release, hip reconstruction and replacement, and femoral or tibial osteotomy through their lifetime. Current measures to intervene in bone disease progression are not perfect and palliative, and improved therapies are urgently required. Enzyme replacement therapy (ERT), hematopoietic stem cell transplantation (HSCT), and gene therapy are available or in development for some types of MPS. Delivery of sufficient enzyme to bone, especially avascular cartilage, to prevent or ameliorate the devastating skeletal dysplasias remains an unmet challenge. The use of an anti-inflammatory drug is also under clinical study. Therapies should start at a very early stage prior to irreversible bone lesion, and damage since the severity of skeletal dysplasia is associated with level of activity during daily life. This review illustrates a current overview of therapies and their impact for bone lesions in MPS including ERT, HSCT, gene therapy, and anti-inflammatory drugs.
Collapse
Affiliation(s)
- Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Medical Genetics Service/HCPA and Department of Genetics/UFRGS, Porto Alegre, Brazil.
| | - Carlos J Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá, D.C., Colombia
| | - Adriana M Montaño
- Department of Pediatrics, Saint Louis University, St. Louis, MO, USA
| | - Hiromasa Yabe
- Department of Cell Transplantation, Tokai University School of Medicine, Isehara, Japan
| | - Akemi Tanaka
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Vu Chi Dung
- Department of Endocrinology, Metabolism & Genetics, Vietnam National Hospital of Pediatrics, Hanoi, Viet Nam
| | - Roberto Giugliani
- Medical Genetics Service/HCPA and Department of Genetics/UFRGS, Porto Alegre, Brazil
| | - Francyne Kubaski
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Robert W Mason
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Eriko Yasuda
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Kazuki Sawamoto
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | | | - Yasuyuki Suzuki
- Medical Education Development Center, Gifu University, Japan
| | - Kenji E Orii
- Department of Pediatrics, Gifu University, Gifu, Japan
| | - Luis A Barrera
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá, D.C., Colombia
| | - William S Sly
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, MO, USA
| | - Tadao Orii
- Department of Pediatrics, Gifu University, Gifu, Japan.
| |
Collapse
|