1
|
MPSI Manifestations and Treatment Outcome: Skeletal Focus. Int J Mol Sci 2022; 23:ijms231911168. [PMID: 36232472 PMCID: PMC9569890 DOI: 10.3390/ijms231911168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/16/2022] [Accepted: 09/17/2022] [Indexed: 11/23/2022] Open
Abstract
Mucopolysaccharidosis type I (MPSI) (OMIM #252800) is an autosomal recessive disorder caused by pathogenic variants in the IDUA gene encoding for the lysosomal alpha-L-iduronidase enzyme. The deficiency of this enzyme causes systemic accumulation of glycosaminoglycans (GAGs). Although disease manifestations are typically not apparent at birth, they can present early in life, are progressive, and include a wide spectrum of phenotypic findings. Among these, the storage of GAGs within the lysosomes disrupts cell function and metabolism in the cartilage, thus impairing normal bone development and ossification. Skeletal manifestations of MPSI are often refractory to treatment and severely affect patients’ quality of life. This review discusses the pathological and molecular processes leading to impaired endochondral ossification in MPSI patients and the limitations of current therapeutic approaches. Understanding the underlying mechanisms responsible for the skeletal phenotype in MPSI patients is crucial, as it could lead to the development of new therapeutic strategies targeting the skeletal abnormalities of MPSI in the early stages of the disease.
Collapse
|
2
|
Stepien KM, Bentley A, Chen C, Dhemech MW, Gee E, Orton P, Pringle C, Rajan J, Saxena A, Tol G, Gadepalli C. Non-cardiac Manifestations in Adult Patients With Mucopolysaccharidosis. Front Cardiovasc Med 2022; 9:839391. [PMID: 35321113 PMCID: PMC8935042 DOI: 10.3389/fcvm.2022.839391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/10/2022] [Indexed: 12/12/2022] Open
Abstract
Mucopolysaccharidoses (MPS) are a heterogeneous group of disorders that results in the absence or deficiency of lysosomal enzymes, leading to an inappropriate storage of glycosaminoglycans (GAGs) in various tissues of the body such as bones, cartilage, heart valves, arteries, upper airways, cornea, teeth, liver and nervous system. Clinical manifestations can become progressively exacerbated with age and affect their quality of life. Developments in advanced supportive treatment options such as enzyme replacement therapy (ERT), hematopoietic stem cell transplantation (HSCT) may have improved patients' life span. Adult MPS patients require specialist clinical surveillance long-term. In many cases, in addition to the MPS-related health problems, they may develop age-related complications. Considering the complexity of their clinical manifestations and lack of guidelines on the management of adult MPS disorders, multispecialty and multidisciplinary teams' care is essential to diagnose and treat health problems that are likely to be encountered. This review presents non-cardiac clinical manifestations, their pathophysiology, management and long-term outcomes in adult MPS patients.
Collapse
Affiliation(s)
- Karolina M. Stepien
- Adult Inherited Metabolic Diseases, Salford Royal National Health Service Foundation Trust, Salford, United Kingdom
| | - Andrew Bentley
- Northwest Ventilation Unit and Sleep Department, Wythenshawe Hospital, Manchester University National Health Service Foundation Trust, Manchester, United Kingdom
- Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
- Intensive Care & Respiratory Medicine, Manchester University National Health Service Foundation Trust, Manchester, United Kingdom
| | - Cliff Chen
- Clinical Neuropsychology, Salford Royal National Health Service Foundation Trust, Salford, United Kingdom
| | - M. Wahab Dhemech
- Northwest Ventilation Unit and Sleep Department, Wythenshawe Hospital, Manchester University National Health Service Foundation Trust, Manchester, United Kingdom
| | - Edward Gee
- Trauma and Orthopaedic Surgery, Salford Royal National Health Service Foundation Trust, Salford, United Kingdom
| | - Peter Orton
- Trauma and Orthopaedic Surgery, Salford Royal National Health Service Foundation Trust, Salford, United Kingdom
| | - Catherine Pringle
- Neurosurgery, Salford Royal National Health Service Foundation Trust, Salford, United Kingdom
| | - Jonathan Rajan
- Manchester and Salford Pain Centre, Salford Royal National Health Service Foundation Trust, Salford, United Kingdom
| | - Ankur Saxena
- Neurosurgery, Salford Royal National Health Service Foundation Trust, Salford, United Kingdom
| | - Govind Tol
- Anaesthetics Department, Salford Royal National Health Service Foundation Trust, Salford, United Kingdom
| | - Chaitanya Gadepalli
- Ear, Nose and Throat, Salford Royal National Health Service Foundation Trust, Salford, United Kingdom
| |
Collapse
|
3
|
Eisengart JB, Esler AN, Ellinwood NM, Hudock RL, King KE, Klein TL, Lee C, Morton J, Stephens K, Ziegler R, O'Neill C. Issues of COVID-19-related distance learning for children with neuronopathic mucopolysaccharidoses. Mol Genet Metab 2021; 134:68-76. [PMID: 34247933 PMCID: PMC8553302 DOI: 10.1016/j.ymgme.2021.06.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/30/2021] [Accepted: 06/30/2021] [Indexed: 12/25/2022]
Abstract
The COVID-19 pandemic has impacted the education of children around the world, forcing a large proportion of teaching to be carried out remotely. The implications of this disruption have yet to be fully elucidated, but initial assessments suggest that COVID-19-related school closures and reliance on virtual learning may have a long-term negative impact on educational attainment and future earnings as well as life expectancy of children in the United States. Among children with neurodegenerative disorders, such as neuronopathic mucopolysaccharidoses (MPS disorders), the effects of the pandemic are likely to be even greater. We aim to shine a spotlight on the impact of COVID-19 on the education, treatment and general wellbeing of children and families affected by MPS disorders by highlighting the important role that educators and therapists play in supporting the neurocognitive function and quality of life of children with neuronopathic MPS disorders. This article will serve as a resource that caregivers, educators, clinicians and therapists can use when considering how best to advocate for children with neuronopathic MPS disorders in circumstances where in-school teaching or in-clinic treatment is compromised or not possible. Given that the current pandemic is likely to have a prolonged course and impact and that similar epidemics and pandemics are a near certainty in the future, it is essential that steps are taken to support the learning and care of children with neuronopathic MPS disorders. We must prioritize strategies to safely resume this fragile community's access to in-person education and supportive care, and to address gaps that have emerged during prolonged pauses in access, whenever possible.
Collapse
Affiliation(s)
- Julie B Eisengart
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.
| | - Amy N Esler
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | | | - Rebekah L Hudock
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Kelly E King
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | | | - Chimei Lee
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Richard Ziegler
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | | |
Collapse
|
4
|
Hampe CS, Wesley J, Lund TC, Orchard PJ, Polgreen LE, Eisengart JB, McLoon LK, Cureoglu S, Schachern P, McIvor RS. Mucopolysaccharidosis Type I: Current Treatments, Limitations, and Prospects for Improvement. Biomolecules 2021; 11:189. [PMID: 33572941 PMCID: PMC7911293 DOI: 10.3390/biom11020189] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/16/2022] Open
Abstract
Mucopolysaccharidosis type I (MPS I) is a lysosomal disease, caused by a deficiency of the enzyme alpha-L-iduronidase (IDUA). IDUA catalyzes the degradation of the glycosaminoglycans dermatan and heparan sulfate (DS and HS, respectively). Lack of the enzyme leads to pathologic accumulation of undegraded HS and DS with subsequent disease manifestations in multiple organs. The disease can be divided into severe (Hurler syndrome) and attenuated (Hurler-Scheie, Scheie) forms. Currently approved treatments consist of enzyme replacement therapy (ERT) and/or hematopoietic stem cell transplantation (HSCT). Patients with attenuated disease are often treated with ERT alone, while the recommended therapy for patients with Hurler syndrome consists of HSCT. While these treatments significantly improve disease manifestations and prolong life, a considerable burden of disease remains. Notably, treatment can partially prevent, but not significantly improve, clinical manifestations, necessitating early diagnosis of disease and commencement of treatment. This review discusses these standard therapies and their impact on common disease manifestations in patients with MPS I. Where relevant, results of animal models of MPS I will be included. Finally, we highlight alternative and emerging treatments for the most common disease manifestations.
Collapse
Affiliation(s)
| | | | - Troy C. Lund
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; (T.C.L.); (P.J.O.); (J.B.E.)
| | - Paul J. Orchard
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; (T.C.L.); (P.J.O.); (J.B.E.)
| | - Lynda E. Polgreen
- The Lundquist Institute at Harbor, UCLA Medical Center, Torrance, CA 90502, USA;
| | - Julie B. Eisengart
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA; (T.C.L.); (P.J.O.); (J.B.E.)
| | - Linda K. McLoon
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Sebahattin Cureoglu
- Department of Otolaryngology, Head and Neck Surgery, University of Minnesota, Minneapolis, MN 55455, USA; (S.C.); (P.S.)
| | - Patricia Schachern
- Department of Otolaryngology, Head and Neck Surgery, University of Minnesota, Minneapolis, MN 55455, USA; (S.C.); (P.S.)
| | - R. Scott McIvor
- Immusoft Corp, Minneapolis, MN 55413, USA;
- Department of Genetics, Cell Biology and Development and Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
5
|
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: 60] [Impact Index Per Article: 15.0] [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
|
6
|
Kuiper GA, Nijmeijer SCM, Roelofs MJM, van der Lee JH, Hollak CEM, Bosch AM. Limited data to evaluate real-world effectiveness of enzyme replacement therapy for mucopolysaccharidosis type I. J Inherit Metab Dis 2019; 42:762-775. [PMID: 31020996 DOI: 10.1002/jimd.12103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/28/2019] [Accepted: 04/24/2019] [Indexed: 12/22/2022]
Abstract
Orphan medicinal products (OMPs) are often authorized based on pivotal phase II and III trials that do not always meet high quality criteria. Laronidase is an example of an OMP used for treatment of mucopolysaccharidosis I (MPS I). One randomized controlled trial demonstrated efficacy on several somatic symptoms. However, effectiveness in the real-world setting remains to be determined. We performed a systematic review to evaluate the effectiveness of enzyme replacement therapy (ERT) on clinically relevant outcomes in MPS I. A search in OVID MEDLINE and OVID EMBASE was performed. Postmarketing studies including MPS I patients treated with ERT and reporting data on any of 19 predefined clinical outcome measures obtained before the start of ERT and at follow-up were eligible. Three scenarios were used to define effectiveness of ERT. The first scenario (A) assumes that improvement is essential, while the second scenario (B) also includes stabilization of signs and symptoms. The third scenario (C) defines failure of therapy. Twenty case series were included. The criteria indicating effectiveness (A), were met for four of 19 outcome measures while the criteria, indicating unclear effectiveness (B) were met for five of 19. For one of 19 nonverifiable data were reported and for nine of 19 no overall conclusions could be drawn (ambiguous results). Real-world effectiveness of laronidase is extremely difficult to assess, 15 years after marketing authorization. This is partially due to insufficient natural history data. We recommend the conduct of rigorous and independent postmarketing studies including core outcome sets for OMPs, enforced by marketing and/or reimbursing authorities aiming to demonstrate real-world effectiveness within a reasonable time frame.
Collapse
Affiliation(s)
- Gé-Ann Kuiper
- Amsterdam UMC, University of Amsterdam, Pediatric Metabolic Diseases, Emma Children's Hospital and Amsterdam Lysosome Center "Sphinx", Meibergdreef 9, Amsterdam, Netherlands
| | - Stephanie C M Nijmeijer
- Amsterdam UMC, University of Amsterdam, Pediatric Metabolic Diseases, Emma Children's Hospital and Amsterdam Lysosome Center "Sphinx", Meibergdreef 9, Amsterdam, Netherlands
| | - Manouck J M Roelofs
- Amsterdam UMC, University of Amsterdam, Pediatric Metabolic Diseases, Emma Children's Hospital, Meibergdreef 9, Amsterdam, Netherlands
| | - Johanneke H van der Lee
- Amsterdam UMC, University of Amsterdam, Pediatric Clinical Research Office, Meibergdreef 9, Amsterdam, Netherlands
| | - Carla E M Hollak
- Amsterdam UMC, University of Amsterdam, Endocrinology and Metabolism, Amsterdam Lysosome Center "Sphinx", Meibergdreef 9, Amsterdam, Netherlands
| | - Annet M Bosch
- Amsterdam UMC, University of Amsterdam, Pediatric Metabolic Diseases, Emma Children's Hospital, Meibergdreef 9, Amsterdam, Netherlands
| |
Collapse
|
7
|
Davari M, Nabizadeh A, Kadivar M, Abdollahi Asl A. Healthcare Resource Utilization and the Cost of Care for Mucopolysaccharidosis I Patients in Iran. Value Health Reg Issues 2019; 18:165-169. [PMID: 31082797 DOI: 10.1016/j.vhri.2019.01.009] [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: 04/10/2018] [Revised: 12/09/2018] [Accepted: 01/31/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND Mucopolysaccharidosis I (MPS-I) is one of the most common types of MPS and lysosomal storage diseases, which impose considerable amount of economic burden on society. OBJECTIVES The aim of this study was to examine the cost drivers in the treatment of MPS-I patients in Iran. METHODS This is a cost-analysis study. The prevalence approach was used to evaluate costs from the healthcare payer's perspective. The number of patients found to have α-L-iduronidase deficiency was identified using the national registry database of the Ministry of Health (MOH). The direct medical costs of the patients were evaluated. Prescriptions; medical interventions; inpatient, outpatient, and diagnostic services, and also their costs were extracted from the patient's profiles in Iran Food and Drug Administration (IFDA). The prices of the medical services were taken out from Iranian medical tariff book 2014-15. Data extraction was performed from January 2017 to March 2018. RESULTS Sixty-six patients were registered as MPS-I in MOH databases. The average annual healthcare cost for every patient was $87 971.99, 96.9% of which was allocated to medication therapy. Therapeutic and diagnostic services costs (2.4% and 0.7% correspondingly) were ranked second and third, respectively, but with huge differences in medication cost. CONCLUSIONS The average annual cost of treatment for MPS-I patients is as high as 16.2 times the GDP per capita in Iran. The highest share of the cost belongs to medication. Selecting appropriate strategies for reducing the birth of a child with MPS could support allocative efficiency of the limited resources effectively.
Collapse
Affiliation(s)
- Majid Davari
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Azita Nabizadeh
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran.
| | - Maliheh Kadivar
- Division of Neonatology, Department of Pediatrics, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Akbar Abdollahi Asl
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
8
|
Abstract
Enzyme replacement therapy (ERT) is available for mucopolysaccharidosis (MPS) I, MPS II, MPS VI, and MPS IVA. The efficacy of ERT has been evaluated in clinical trials and in many post-marketing studies with a long-term follow-up for MPS I, MPS II, and MPS VI. While ERT is effective in reducing urinary glycosaminoglycans (GAGs) and liver and spleen volume, cartilaginous organs such as the trachea and bronchi, bones and eyes are poorly impacted by ERT probably due to limited penetration in the specific tissue. ERT in the present formulations also does not cross the blood–brain barrier, with the consequence that the central nervous system is not cured by ERT. This is particularly important for severe forms of MPS I and MPS II characterized by cognitive decline. For severe MPS I patients (Hurler), early haematopoietic stem cell transplantation is the gold standard, while still controversial is the role of stem cell transplantation in MPS II. The use of ERT in patients with severe cognitive decline is the subject of debate; the current position of the scientific community is that ERT must be started in all patients who do not have a more effective treatment. Neonatal screening is widely suggested for treatable MPS, and many pilot studies are ongoing. The rationale is that early, possibly pre-symptomatic treatment can improve prognosis. All patients develop anti-ERT antibodies but only a few have drug-related adverse reactions. It has not yet been definitely clarified if high-titre antibodies may, at least in some cases, reduce the efficacy of ERT.
Collapse
Affiliation(s)
- Daniela Concolino
- Department of Medical and Surgical Science, Pediatric Unit, University "Magna Graecia", Catanzaro, Italy
| | - Federica Deodato
- Division of Metabolic Disease, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Rossella Parini
- UOS Malattie Metaboliche Rare, Clinica Pediatrica dell'Università Milano Bicocca, Fondazione MBBM, ATS Monza e Brianza, Via Pergolesi 33, 20900, Monza, Italy. .,San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
9
|
Pain in Mucopolysaccharidoses: Analysis of the Problem and Possible Treatments. Int J Mol Sci 2018; 19:ijms19103063. [PMID: 30297617 PMCID: PMC6213542 DOI: 10.3390/ijms19103063] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 09/30/2018] [Accepted: 10/02/2018] [Indexed: 01/01/2023] Open
Abstract
Mucopolysaccharidosis (MPS) are a group of lysosomal storage disorders that are caused by the deficiency of enzymes involving in the catabolism of glycosaminoglycan (GAGs). GAGs incompletely degraded accumulate in many sites, damaging tissues and cells, leading to a variety of clinical manifestations. Many of these manifestations are painful, but few data are available in the literature concerning the prevalence, etiology, and pathogenesis of pain in children with MPS. This review, through the analysis of the data available the in literature, underscores the relevant prevalence of pain in MPSs’ children, provides the instruments to discern the etiopathogenesis of the disease and of pain, illustrates the available molecules for the management of pain and the possible advantages of non-pharmacological pain therapy in MPSs’ patients.
Collapse
|
10
|
Wittke TC, Schmidtke J, Grigull L. Rare diseases and sports: A pilot project to improve physical activity in patients with mucopolysaccharidosis. TRANSLATIONAL SPORTS MEDICINE 2018. [DOI: 10.1002/tsm2.35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | - Jörg Schmidtke
- Institute of Human Genetics; Hannover Medical School; Hannover Germany
| | - Lorenz Grigull
- Department of Pediatric Hematology and Oncology; Hannover Medical School; Hannover Germany
| |
Collapse
|
11
|
Abstract
Inborn errors of metabolism encompass a wide spectrum of disorders, frequently affecting bone. The most important metabolic disorders that primarily influence calcium or phosphate balance, resulting in skeletal pathology, are hypophosphatemic rickets and hypophosphatasia. Conditions involving bone marrow or affecting skeletal growth and development are mainly the lysosomal storage disorders, in particular the mucopolysaccharidoses. In these disorders skeletal abnormalities are often the presenting symptom and early recognition and intervention improves outcome in many of these diseases. Many disorders of intermediary metabolism may impact bone health as well, resulting in higher frequencies of osteopenia and osteoporosis. In these conditions factors contributing to the reduced bone mineralization can be the disorder itself, the strict dietary treatment, reduced physical activity or sunlight exposure and/or early ovarian failure. Awareness of these primary or secondary bone problems amongst physicians treating patients with inborn errors of metabolism is of importance for optimization bone health and recognition of skeletal complications.
Collapse
Affiliation(s)
- M Langeveld
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.
| | - C E M Hollak
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| |
Collapse
|
12
|
Dornelles AD, Artigalás O, da Silva AA, Ardila DLV, Alegra T, Pereira TV, Vairo FPE, Schwartz IVD. Efficacy and safety of intravenous laronidase for mucopolysaccharidosis type I: A systematic review and meta-analysis. PLoS One 2017; 12:e0184065. [PMID: 28859139 PMCID: PMC5578671 DOI: 10.1371/journal.pone.0184065] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/17/2017] [Indexed: 12/19/2022] Open
Abstract
Mucopolysaccharidosis type I (MPS I) is an autosomal recessive disease caused by deficient activity of alpha-L-iduronidase. Intravenous (IV) enzyme replacement therapy (ERT) with laronidase is currently used for treating patients with MPS I.
Collapse
Affiliation(s)
- Alícia Dorneles Dornelles
- Postgraduate Program in Genetics Applied to Medicine, Department of Pediatrics, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- * E-mail:
| | - Osvaldo Artigalás
- Hospital Materno-Infantil Presidente Vargas, Porto Alegre, RS, Brazil
- Clinical Genetics Unit, Children's Hospital, Grupo Hospitalar Conceição, Porto Alegre, RS, Brazil
| | | | | | - Taciane Alegra
- Nutrition, Biomarkers and Health Research Group, University College Dublin, Dublin, Ireland
| | - Tiago Veiga Pereira
- Instituto de Educação e Ciências em Saúde, Hospital Alemão Osvaldo Cruz, São Paulo, SP, Brazil
| | - Filippo Pinto e Vairo
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Ida Vanessa Doederlein Schwartz
- Postgraduate Program in Genetics Applied to Medicine, Department of Pediatrics, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of 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
- BRAIN Laboratory, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| |
Collapse
|
13
|
Laraway S, Mercer J, Jameson E, Ashworth J, Hensman P, Jones SA. Outcomes of Long-Term Treatment with Laronidase in Patients with Mucopolysaccharidosis Type I. J Pediatr 2016; 178:219-226.e1. [PMID: 27788836 DOI: 10.1016/j.jpeds.2016.08.033] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 06/20/2016] [Accepted: 08/09/2016] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To evaluate long-term outcomes of laronidase enzyme replacement therapy in patients with attenuated mucopolysaccharidosis type I. STUDY DESIGN Retrospective analyses of case notes, laboratory results, and data from clinical trials were used to evaluate urinary glycosaminoglycans, forced vital capacity (FVC), 6-minute walk test (6MWT), height-for-age Z score, cardiac valve function, corneal clouding, and visual acuity in 35 patients with attenuated mucopolysaccharidosis type I (Hurler-Scheie and Scheie syndromes) for up to 10 years following the initiation of laronidase therapy. RESULTS Statistically significant (P < .001) reductions in mean urinary glycosaminoglycan levels relative to baseline were observed 6 months after treatment initiation and were sustained throughout follow-up. Disease remained stable after treatment initiation with no statistically significant changes in mean FVC, 6MWT, or height-for-age Z score. At last assessments, mitral and aortic valve function remained stable in 65% (22/34) of patients; corneal clouding remained stable in 78% (18/23); visual acuity remained stable in 33% (8/24) and improved in 42% (10/24) of patients. Younger patients (<10 years at treatment initiation) maintained disease measures closer to norms for age for FVC, 6MWT, and height and showed fewer deteriorations in mitral and aortic valve disease and corneal clouding compared with patients aged ≥10 years at treatment initiation. CONCLUSION Laronidase treatment resulted in disease stabilization in the majority of patients with a mean follow-up of 6.1 years. Data suggest that early treatment may result in better outcomes.
Collapse
Affiliation(s)
- Sarah Laraway
- Willink Biochemical Genetic Unit, Manchester Centre for Genomic Medicine, Saint Mary's Hospital, Manchester, UK.
| | - Jean Mercer
- Willink Biochemical Genetic Unit, Manchester Centre for Genomic Medicine, Saint Mary's Hospital, Manchester, UK
| | - Elisabeth Jameson
- Willink Biochemical Genetic Unit, Manchester Centre for Genomic Medicine, Saint Mary's Hospital, Manchester, UK
| | - Jane Ashworth
- Manchester Royal Eye Hospital, Central Manchester University Hospitals National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK; Centre for Ophthalmology and Vision Sciences, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| | - Pauline Hensman
- Willink Biochemical Genetic Unit, Manchester Centre for Genomic Medicine, Saint Mary's Hospital, Manchester, UK
| | - Simon A Jones
- Willink Biochemical Genetic Unit, Manchester Centre for Genomic Medicine, Saint Mary's Hospital, Manchester, UK
| |
Collapse
|
14
|
Hendriksz CJ, Berger KI, Lampe C, Kircher SG, Orchard PJ, Southall R, Long S, Sande S, Gold JI. Health-related quality of life in mucopolysaccharidosis: looking beyond biomedical issues. Orphanet J Rare Dis 2016; 11:119. [PMID: 27561270 PMCID: PMC5000418 DOI: 10.1186/s13023-016-0503-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/17/2016] [Indexed: 11/28/2022] Open
Abstract
The mucopolysaccharidoses (MPS) comprise a heterogeneous family of rare, genetic lysosomal storage disorders that result in severe morbidity and reduced life expectancy. Emerging treatments for several of these disorders have triggered the search for clinically relevant biomarkers and clinical markers associated with treatment efficacy in populations and individuals. However, biomedical measures do not tell the whole story when characterizing a complex chronic disorder such as MPS. Health-related quality of life (HRQoL) tools that utilize patient reported outcomes to address patient parameters such as symptoms (pain, fatigue, psychological health), functioning (activity and limitations), or quality of life, have been used to supplement traditional biomedical endpoints. Many of these HRQoL tools have demonstrated that quality of life is negatively impacted in patients with MPS. There is both the opportunity and need to formally standardize and validate HRQoL tools for the different MPS disorders.
Collapse
Affiliation(s)
- Christian J Hendriksz
- Adult Inherited Metabolic Disorders, Consultant Transitional Metabolic Medicine, The Mark Holland Metabolic Unit, Salford Royal NHS Foundation Trust, Ladywell NW2- 2nd Floor Room 112, Salford, Manchester, M6 8HD, UK. .,Paediatrics and Child Health, University of Pretoria, Steve Biko Academic Unit, Pretoria, South Africa.
| | - Kenneth I Berger
- Division of Pulmonary, Critical Care and Sleep Medicine, New York University School of Medicine and André Cournand Pulmonary Physiology Laboratory, Bellevue Hospital, New York, USA
| | - Christina Lampe
- Centre for Rare Diseases, Clinic for children and adolescents, Helios Dr. Horst Schmidt Kliniken, Wiesbaden, Germany
| | - Susanne G Kircher
- Institute of Medical Chemistry and Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Paul J Orchard
- Department of Pediatrics, Division of Blood & Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | | | - Sarah Long
- School of Sociology and Social Policy, University of Bath, Bath, UK
| | | | - Jeffrey I Gold
- Keck School of Medicine, Departments of Anesthesiology, Pediatrics, and Psychiatry & Behavioral Sciences, Children's Hospital Los Angeles, Anesthesiology Critical Care Medicine, Pediatric Pain Management Clinic, University of Southern California, California, USA
| |
Collapse
|
15
|
Guarany NR, Vanz AP, Wilke MVMB, Bender DD, Borges MD, Giugliani R, Schwartz IVD. Mucopolysaccharidosis. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2015. [DOI: 10.1177/2326409815613804] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Nicole Ruas Guarany
- Graduate Program in Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Occupational Therapy, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Ana Paula Vanz
- Graduate Program in Children and Adolescent Health, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Daniele Dorneles Bender
- Undergraduate of School Occupational Therapy, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Mariana Dumer Borges
- Undergraduate of School Occupational Therapy, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Roberto Giugliani
- Service of Medical Genetics, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Department of Genetics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ida Vanessa Doederlein Schwartz
- Graduate Program in Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Service of Medical Genetics, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Department of Genetics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
16
|
Lampe C, Jain M, Olaye A, Meesen B, Decker C, Mengel E. Relationship Between Patient-Reported Outcomes and Clinical Outcomes in Patients With Morquio A Syndrome. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2015. [DOI: 10.1177/2326409815576188] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Christina Lampe
- Centre for Rare Diseases, Clinic for children and adolescents, Dr Horst Schmidt Klinik, Wiesbaden, Germany
| | - Mohit Jain
- BioMarin Europe Ltd, London, United Kingdom
| | | | | | | | - Eugen Mengel
- Department of Pediatric and Adolescent Medicine, Villa Metabolica, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
17
|
Langereis EJ, van Vlies N, Wijburg FA. Diagnosis, classification and treatment of mucopolysaccharidosis type I. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1016908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
18
|
|
19
|
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: 8.1] [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
|
20
|
Johnson BA, Dajnoki A, Bodamer OA. Diagnosing lysosomal storage disorders: mucopolysaccharidosis type I. ACTA ACUST UNITED AC 2015; 84:17.17.1-17.17.8. [PMID: 25599668 DOI: 10.1002/0471142905.hg1717s84] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Mucopolysaccharidosis type I (MPS I) is a lysosomal storage disorder due to deficiency of alpha iduronidase (IDUA). Progressive storage of dermatan and heparan sulfate throughout the body lead to a multiorgan presentation including short stature, dysostosis multiplex, corneal clouding, hearing loss, coarse facies, hepatosplenomegaly, and intellectual disability. Diagnosis of MPS I is based on IDUA enzyme analysis in leukocytes or dried blood spots (DBS) followed by molecular confirmation of the IDUA gene mutations in individuals with low enzyme activity. The advent of mass spectrometry methods for enzyme analysis in DBS has enabled high-throughput screening for MPS I in symptomatic individuals and newborn infants. The following unit provides the detailed analytical protocol for measurement of IDUA activity in DBS using tandem mass spectrometry.
Collapse
Affiliation(s)
- Britt A Johnson
- Division of Clinical and Translational Genetics, Dr. John T. MacDonald Foundation, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida
| | - Angela Dajnoki
- Division of Clinical and Translational Genetics, Dr. John T. MacDonald Foundation, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida
| | - Olaf A Bodamer
- Division of Clinical and Translational Genetics, Dr. John T. MacDonald Foundation, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
21
|
Yang JS, Min HK, Oh HJ, Woo HI, Lee SY, Kim JW, Song J, Park HD. A simple and rapid method based on liquid chromatography-tandem mass spectrometry for the measurement of α-L-iduronidase activity in dried blood spots: an application to mucopolysaccharidosis I (Hurler) screening. Ann Lab Med 2014; 35:41-9. [PMID: 25553279 PMCID: PMC4272964 DOI: 10.3343/alm.2015.35.1.41] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 02/17/2014] [Accepted: 10/02/2014] [Indexed: 12/02/2022] Open
Abstract
Background We developed an analytical method to measure α-L-iduronidase (IDUA) activity in dried blood spots. This was achieved by using liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) with electrospray ionization in the positive ion mode. Methods Chromatographic separation was completed using mobile phase involving water-formic acid and acetonitrile-formic acid over 2.8 min of run time on a column with a Kinetex XB-C18 (Phenomenex, USA). The detection of column effluent was performed using a Xevo TQ-S triple quadrupole mass spectrometer (Waters, USA) in the multiple-reaction monitoring mode. This method was verified with blank and control samples at four activity levels: base, low, medium, and high. Control materials were provided from Centers for Disease Control and Prevention (CDC). Results Intra- and inter-day precisions were between 2.6% and 16.5% and between 7.9% and 17.0%, respectively. A correlative regression study on the IDUA activity in CDC-control samples performed to assess the validity of the developed method showed a highly significant linear association (r2=0.9976) between the calculated and CDC-reported values and an obvious difference in activity among the four levels. This reliable analytical method was applied to mucopolysaccharidosis I (Hurler) screening of patients under treatment (n=4) and in normal controls (n=129). IDUA activity ranged from 8.98 to 77.12 µmol/hr/L) in normal controls, and patients undergoing medical treatment showed low IDUA activity. Conclusions This method had advantages of simplicity, rapid sample preparation, and liquid chromatographic separation, which efficiently inhibited ionization suppression induced by matrix effects in mass spectrometric detection.
Collapse
Affiliation(s)
- Jeong Soo Yang
- Clinical Trial Center, Clinical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Hye Kyeong Min
- Clinical Trial Center, Clinical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Hyeon Ju Oh
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Seoul, Korea
| | - Hye In Woo
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Seoul, Korea. ; Department of Laboratory Medicine and Genetics, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo-Youn Lee
- Clinical Trial Center, Clinical Research Institute, Samsung Medical Center, Seoul, Korea. ; Department of Laboratory Medicine and Genetics, Samsung Medical Center, Seoul, Korea. ; Department of Laboratory Medicine and Genetics, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong-Won Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Seoul, Korea. ; Department of Laboratory Medicine and Genetics, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Junghan Song
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hyung-Doo Park
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Seoul, Korea. ; Department of Laboratory Medicine and Genetics, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
22
|
Pasqualim G, Ribeiro MG, da Fonseca GGG, Szlago M, Schenone A, Lemes A, Rojas MVM, Matte U, Giugliani R. p.L18P: a novel IDUA mutation that causes a distinct attenuated phenotype in mucopolysaccharidosis type I patients. Clin Genet 2014; 88:376-80. [PMID: 25256405 DOI: 10.1111/cge.12507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/17/2014] [Accepted: 09/19/2014] [Indexed: 10/24/2022]
Abstract
Mucopolysaccharidosis type I is a rare autosomal recessive disorder caused by deficiency of α-l-iduronidase (IDUA) which leads to a wide spectrum of clinical severity. Here, we describe the case of four male patients who present the previously undescribed p.L18P mutation. Patient 1 (p.L18P/p.L18P) presents, despite multiple joint contractures, an attenuated phenotype. Patient 2 (p.L18P/p.W402X) was diagnosed at 4 years of age with bone dysplasia, coarse facies, limited mobility, claw hands and underwent bilateral carpal tunnel surgery at 6 years of age. Patients 3 and 4 (both p.L18P/p.L18P) are brothers. Patient 3 was diagnosed at 4 years of age, when presented claw hands, lower limb and shoulder pain, restricted articular movement and bilateral carpal tunnel syndrome. Patient 4 was diagnosed at 17 months of age when presented lower limb pain at night, respiratory allergy and repeated upper airways infections. Bioinformatics analysis indicates that p.L18P mutation reduces the signal peptide to 25 amino acids and alters its secondary structure. In conclusion, we report a new IDUA variant that alters the structure of the signal peptide, which likely impairs transport to lysosomes. Moreover, it leads to a distinct attenuated phenotype with mainly bone and cartilage symptoms, without visceromegalies, heart disease, or cognitive impairment.
Collapse
Affiliation(s)
- G Pasqualim
- Post-Graduate Program on Genetics and Molecular Biology, UFRGS, Porto Alegre, Brazil.,Gene Therapy Center, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - M G Ribeiro
- Instituto de Puericultura e Pediatria Martagão Gesteira, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro, Brazil
| | - G G G da Fonseca
- Instituto de Puericultura e Pediatria Martagão Gesteira, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro, Brazil
| | - M Szlago
- Fundación para el Estudio de las Enfermedades metabólicas (FESEN), Buenos Aires, Argentina
| | - A Schenone
- Fundación para el Estudio de las Enfermedades metabólicas (FESEN), Buenos Aires, Argentina
| | - A Lemes
- Instituto de Genética Médica, Hospital Italiano, Montevideo, Uruguay
| | - M V M Rojas
- Genzyme, Genzyme Latin America - A Sanofi Company, Rio de Janeiro, Brazil
| | - U Matte
- Post-Graduate Program on Genetics and Molecular Biology, UFRGS, Porto Alegre, Brazil.,Gene Therapy Center, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Department of Genetics, UFRGS, Porto Alegre, Brazil.,INAGEMP, Porto Alegre, Brazil
| | - R Giugliani
- Post-Graduate Program on Genetics and Molecular Biology, UFRGS, Porto Alegre, Brazil.,Gene Therapy Center, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Department of Genetics, UFRGS, Porto Alegre, Brazil.,INAGEMP, Porto Alegre, Brazil.,Medical Genetics Service, Hospital de Clinicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
23
|
Boulay C, Lemoine M, Chabrol B. Prise en charge globale fonctionnelle des mucopolysaccharidoses. Arch Pediatr 2014; 21 Suppl 1:S46-9. [DOI: 10.1016/s0929-693x(14)72259-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
24
|
Muenzer J. Early initiation of enzyme replacement therapy for the mucopolysaccharidoses. Mol Genet Metab 2014; 111:63-72. [PMID: 24388732 DOI: 10.1016/j.ymgme.2013.11.015] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 11/29/2013] [Accepted: 11/29/2013] [Indexed: 10/25/2022]
Abstract
The mucopolysaccharidoses (MPS), a group of rare genetic disorders caused by defects in glycosaminoglycan (GAG) catabolism, are progressive, multi-systemic diseases with a high burden of morbidity. Enzyme replacement therapy (ERT) is available for MPS I, II, and VI, and may improve walking ability, endurance, and pulmonary function as evidenced by data from pivotal trials and extension studies. Despite these demonstrable benefits, cardiac valve disease, joint disease, and skeletal disease, all of which cause significant morbidity, do not generally improve with ERT if pathological changes are already established. Airway disease improves, but usually does not normalize. These limitations can be well understood by considering the varied functions of GAG in the body. Disruption of GAG catabolism has far-reaching effects due to the triggering of secondary pathogenic cascades. It appears that many of the consequences of these secondary pathogenic events, while they may improve on treatment, cannot be fully corrected even with long-term exposure to enzyme, thereby supporting the treatment of patients with MPS before the onset of clinical disease. This review examines the data from clinical trials and other studies in human patients to explore the limits of ERT as currently used, then discusses the pathophysiology, fetal tissue studies, animal studies, and sibling reports to explore the question of how early to treat an MPS patient with a firm diagnosis. The review is followed by an expert opinion on the rationale for and the benefits of early treatment.
Collapse
Affiliation(s)
- Joseph Muenzer
- Division of Genetics and Metabolism, Department of Pediatrics, CB 7487, Medical School Wing E Room 117, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7487, USA.
| |
Collapse
|
25
|
Opoka-Winiarska V, Jurecka A, Emeryk A, Tylki-Szymańska A. Osteoimmunology in mucopolysaccharidoses type I, II, VI and VII. Immunological regulation of the osteoarticular system in the course of metabolic inflammation. Osteoarthritis Cartilage 2013; 21:1813-23. [PMID: 23954699 DOI: 10.1016/j.joca.2013.08.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 07/27/2013] [Accepted: 08/03/2013] [Indexed: 02/02/2023]
Abstract
BACKGROUND Mucopolysaccharidoses (MPSs) are rare genetic diseases caused by a deficient activity of one of the lysosomal enzymes involved in the glycosaminoglycan (GAG) breakdown pathway. These metabolic blocks lead to the accumulation of GAGs in various organs and tissues, resulting in a multisystemic clinical picture. The pathological GAG accumulation begins a cascade of interrelated responses: metabolic, inflammatory and immunological with systemic effects. Metabolic inflammation, secondary to GAG storage, is a significant cause of osteoarticular symptoms in MPS disorders. OBJECTIVE AND METHOD The aim of this review is to present recent progress in the understanding of the role of inflammatory and immune processes in the pathophysiology of osteoarticular symptoms in MPS disorders and potential therapeutic interventions based on published reports in MPS patients and studies in animal models. RESULTS AND CONCLUSIONS The immune and skeletal systems have a number of shared regulatory molecules and many relationships between bone disorders and aberrant immune responses in MPS can be explained by osteoimmunology. The treatment options currently available are not sufficiently effective in the prevention, inhibition and treatment of osteoarticular symptoms in MPS disease. A lot can be learnt from interactions between skeletal and immune systems in autoimmune diseases such as rheumatoid arthritis (RA) and similarities between RA and MPS point to the possibility of using the experience with RA in the treatment of MPS in the future. The use of different anti-inflammatory drugs requires further study, but it seems to be an important direction for new therapeutic options for MPS patients.
Collapse
Affiliation(s)
- V Opoka-Winiarska
- Department of Paediatric Pulmonology and Rheumatology, Medical University of Lublin, Lublin, Poland.
| | | | | | | |
Collapse
|
26
|
de Oliveira PG, Baldo G, Mayer FQ, Martinelli B, Meurer L, Giugliani R, Matte U, Xavier RM. Characterization of joint disease in mucopolysaccharidosis type I mice. Int J Exp Pathol 2013; 94:305-11. [PMID: 23786352 PMCID: PMC3781776 DOI: 10.1111/iep.12033] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 05/11/2013] [Indexed: 11/30/2022] Open
Abstract
Mucopolysaccharidoses (MPS) are lysosomal storage disorders characterized by mutations in enzymes that degrade glycosaminoglycans (GAGs). Joint disease is present in most forms of MPS, including MPS I. This work aimed to describe the joint disease progression in the murine model of MPS I. Normal (wild-type) and MPS I mice were sacrificed at different time points (from 2 to 12 months). The knee joints were collected, and haematoxylin-eosin staining was used to evaluate the articular architecture. Safranin-O and Sirius Red staining was used to analyse the proteoglycan and collagen content. Additionally, we analysed the expression of the matrix-degrading metalloproteinases (MMPs), MMP-2 and MMP-9, using immunohistochemistry. We observed progressive joint alterations from 6 months, including the presence of synovial inflammatory infiltrate, the destruction and thickening of the cartilage extracellular matrix, as well as proteoglycan and collagen depletion. Furthermore, we observed an increase in the expression of MMP-2 and MMP-9, which could conceivably explain the degenerative changes. Our results suggest that the joint disease in MPS I mice may be caused by a degenerative process due to increase in proteases expression, leading to loss of collagen and proteoglycans. These results may guide the development of ancillary therapies for joint disease in MPS I.
Collapse
Affiliation(s)
- Patricia G de Oliveira
- Programa de pós-graduação em medicina: ciências medicas, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Tomatsu S, Alméciga-Díaz CJ, Barbosa H, Montaño AM, Barrera LA, Shimada T, Yasuda E, Mackenzie WG, Mason RW, Suzuki Y, Orii KE, Orii T. Therapies of mucopolysaccharidosis IVA (Morquio A syndrome). Expert Opin Orphan Drugs 2013; 1:805-818. [PMID: 25419501 DOI: 10.1517/21678707.2013.846853] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Morquio A syndrome (mucopolysaccharidosis type IVA, MPS IVA) is one of the lysosomal storage diseases and is caused by the deficiency of N-acetylgalactosamine-6-sulfate sulfatase (GALNS). Deficiency of this enzyme leads to accumulation of glycosaminoglycans (GAGs), keratan sulfate (KS) and chondroitin-6-sulfate (C6S). The majority of KS is produced by chondrocytes, and therefore, the undegraded substrates accumulate mainly in cells and extracelluar matrix (ECM) of cartilage. This has a direct impact on cartilage and bone development, leading to systemic skeletal dysplasia. In patients with Morquio A, cartilage cells are vacuolated, and this results in abnormal chondrogenesis and/or endochondral ossification. AREAS COVERED This article describes the advanced therapies of Morquio A, focused on enzyme replacement therapy (ERT) and gene therapy to deliver the drug to avascular bone lesions. ERT and gene therapies for other types of MPS are also discussed, which provide therapeutic efficacy to bone lesions. EXPERT OPINION ERT, gene therapy and hematopietic stem therapy are clinically and/or experimentally conducted. However, there is no effective curative therapy for bone lesion to date. One of the limitations for Morquio A therapy is that targeting avascular cartilage tissues remains an unmet challenge. ERT or gene therapy with bone-targeting system will improve the bone pathology and skeletal manifestations more efficiently.
Collapse
Affiliation(s)
- Shunji Tomatsu
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA ; Nemours/Alfred I. duPont Hospital for Children, Skeletal Dysplasia Center, Nemours Biomedical Research, 1600 Rockland Rd., Wilmington, DE 19803, USA
| | - Carlos J Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Hector Barbosa
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Adriana M Montaño
- Saint Louis University, Department of Pediatrics, St. Louis, MO, USA
| | - Luis A Barrera
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Tsutomu Shimada
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Eriko Yasuda
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - William G Mackenzie
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Robert W Mason
- Professor and Director, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Yasuyuki Suzuki
- Gifu University, Medical Education Development Center, Gifu, Japan
| | - Kenji E Orii
- Gifu University, Department of Pediatrics, Gifu, Japan
| | - Tadao Orii
- Gifu University, Department of Pediatrics, Gifu, Japan
| |
Collapse
|
28
|
Baldo G, Mayer FQ, Martinelli BZ, de Carvalho TG, Meyer FS, de Oliveira PG, Meurer L, Tavares A, Matte U, Giugliani R. Enzyme replacement therapy started at birth improves outcome in difficult-to-treat organs in mucopolysaccharidosis I mice. Mol Genet Metab 2013; 109:33-40. [PMID: 23562162 DOI: 10.1016/j.ymgme.2013.03.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 03/09/2013] [Accepted: 03/09/2013] [Indexed: 11/25/2022]
Abstract
Since we previously observed that in patients with mucopolysaccharidosis (MPS) the storage of undegraded glycosaminoglycans (GAG) occurs from birth, in the present study we aimed to compare normal, untreated MPS I mice (knockout for alpha-l-iduronidase-IDUA), and MPS I mice treated with enzyme replacement therapy (ERT, Laronidase, 1.2mg/kg every 2 weeks) started from birth (ERT-neo) or from 2 months of age (ERT-ad). All mice were sacrificed at 6 months. Both treatments were equally effective in normalizing GAG levels in the viscera but had no detectable effect on the joint. Heart function was also improved with both treatments. On the other hand, mice treated from birth presented better outcomes in the difficult-to-treat aortas and heart valves. Surprisingly, both groups had improvements in behavior tests, and normalization of GAG levels in the brain and IDUA injection resulted in detectable levels of enzyme in the brain tissue 1h after administration. ERT-ad mice developed significantly more anti-IDUA-IgG antibodies, and mice that didn't develop antibodies had better performances in behavior tests, indicating that development of antibodies may reduce enzyme bioavailability. Our results suggest that ERT started from birth leads to better outcomes in the aorta and heart valves, as well as a reduction in antibody levels. Some poor vascularized organs, such as the joints, had partial or no benefit and ancillary therapies might be needed for patients. The results presented here support the idea that ERT started from birth leads to better treatment outcomes and should be considered whenever possible, a observation that gains relevance as newborn screening programs are being considered for MPS and other treatable lysosomal storage disorders.
Collapse
Affiliation(s)
- Guilherme Baldo
- Centro de Terapia Gênica, Hospital de Clinicas de Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Lagranha VL, de Carvalho TG, Giugliani R, Matte U. Treatment of MPS I mice with microencapsulated cells overexpressing IDUA: effect of the prednisolone administration. J Microencapsul 2013; 30:383-9. [DOI: 10.3109/02652048.2012.746745] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
30
|
Jurecka A, Marucha J, Jurkiewicz E, Różdżyńska-Świątkowska A, Tylki-Szymańska A. Enzyme replacement therapy in an attenuated case of mucopolysaccharidosis type I (Scheie syndrome): a 6.5-year detailed follow-up. Pediatr Neurol 2012; 47:461-5. [PMID: 23127271 DOI: 10.1016/j.pediatrneurol.2012.08.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 08/13/2012] [Indexed: 10/27/2022]
Abstract
We present the 6.5-year follow-up of a boy with Scheie syndrome whose therapy was initiated at age 2.5 years. Detailed anthropometric features, echocardiography, ophthalmologic and audiologic examinations, psychologic tests, joint range of motion, skeletal radiographs, ultrasound studies of liver and spleen volumes, urinary glycosaminoglycans, and antibodies were documented. After 6.5 years of enzyme replacement therapy, the patient experienced a decline in overall status, and the disease progressed significantly despite treatment. The patient's height at age 9 was equal to that at age 6. The patient developed heart insufficiency and a deterioration of airway flow. The patient's intelligence quotient remained unchanged, i.e., at the average level of 86 on the Terman-Merill Scale. Magnetic resonance imaging revealed cervical spinal canal stenosis and marked spinal cord compression with myelopathy. A worsening of carpal tunnel syndrome was also evident. Ophthalmologic evaluation revealed increased central corneal thickness and intraocular pressure. In audiologic assessment, the patient's results were similar to those after 3 years of treatment. The only benefit involved temporarily improved passive and active shoulder flexion. Overall, the benefit of enzyme replacement therapy with laronidase on Scheie syndrome appeared minimal.
Collapse
Affiliation(s)
- Agnieszka Jurecka
- Department of Metabolic Diseases, Children's Memorial Health Institute, Warsaw, Poland.
| | | | | | | | | |
Collapse
|
31
|
Macsai CE, Derrick-Roberts ALK, Ding X, Zarrinkalam KH, McIntyre C, Anderson PH, Anson DS, Byers S. Skeletal response to lentiviral mediated gene therapy in a mouse model of MPS VII. Mol Genet Metab 2012; 106:202-13. [PMID: 22525091 DOI: 10.1016/j.ymgme.2012.03.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 03/30/2012] [Accepted: 03/30/2012] [Indexed: 12/01/2022]
Abstract
Mucopolysaccharidosis VII (MPS VII) is an autosomal recessive, lysosomal storage disorder caused by β-glucuronidase (GUSB) deficiency, resulting in the accumulation of glycosaminoglycans (GAGs), in a variety of cell types. Severe, progressive skeletal pathology, termed dysostosis multiplex, is a prominent clinical feature of MPS VII. We have evaluated a gene therapy protocol for its efficacy in preventing the development and progression of bone pathology in MPS VII mice treated with a lentiviral vector at birth or at 7 weeks. Two weeks after injections, high levels of vector expression were observed in liver, spleen and bone marrow and to a lesser extent in kidney, lung and heart. Widespread clearance of GAG storage was observed in somatic tissues of both groups and some clearance of neuronal storage was observed in mice treated from birth. Micro-CT analysis demonstrated a significant decrease in vertebral and femoral bone mineral volume, trabecular number, bone surface density and cortical bone thickness in both treatment groups. Lumbar and femoral bone lengths were significantly decreased in untreated MPS VII mice, while growth plate heights were increased and these parameters did not change upon treatment. Small improvements in performance in the open field and rotarod behaviour tests were noted. Overall, systemic lentiviral-mediated gene therapy results in a measurable improvement in parameters of bone mass and architecture as well as biochemical and enzymatic correction. Conversely, growth plate chondrocytes were not responsive to treatment, as evidenced by the lack of improvement in vertebral and femoral bone length and growth plate height.
Collapse
Affiliation(s)
- Carmen E Macsai
- Genetics and Molecular Pathology, SA Pathology (CYWHS Site), Adelaide, SA 5006, Australia
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Marucha J, Jurecka A, Syczewska M, Różdżyńska-Świątkowska A, Tylki-Szymańska A. Restricted joint range of motion in patients with MPS II: correlation with height, age and functional status. Acta Paediatr 2012; 101:e183-8. [PMID: 22077147 DOI: 10.1111/j.1651-2227.2011.02522.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM The aims of the study were to assess shoulder range of motion (ROM) in patients with mucopolysaccharidosis type II (MPS II) and to correlate joint mobility with patients' height, age and functional status. METHODS Passive ROM and Z-score of height were followed in 29 patients with MPS II (mean age 11.5 years, range 2-29 years) between the years 2005 and 2010. Passive ROM was measured by a goniometer, and height, by a stadiometer. Functional status was assessed by an age-appropriate health assessment questionnaire (HAQ). RESULTS (i) A strong correlation was observed between patients' age and Z-score of patients' height (R = 0.78, p < 0.001). (ii) A medium correlation was observed between Z-score of patients' height and passive shoulder flexion and abduction (R = 0.697, p < 0.001 and R = 0.63, p < 0.001, respectively). The progression of restriction was slower in attenuated patients. (iii) Restrictions in shoulder flexion and abduction were already observed before the second year of life. (iv) ROM limitations intensified and became more severe with age. (v) Activities of daily living depended on cognitive impairment of patients with MPS II. CONCLUSION Range of motion limitations in patients with MPS II correlate with patients' height, increase with patients' age and are more pronounced in a severe form of MPS II.
Collapse
Affiliation(s)
- Jolanta Marucha
- Metabolic Diseases Clinic, The Children's Memorial Health Institute, Warsaw, Poland
| | | | | | | | | |
Collapse
|
33
|
Musculoskeletal manifestations of mucopolysaccharidosis type VI and effects of enzyme replacement therapy. Open Med (Wars) 2012. [DOI: 10.2478/s11536-012-0003-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe aim of this study was to describe the musculoskeletal manifestations of mucopolysaccharidosis VI and to assess the effectiveness of enzyme replacement therapy (ERT) with recombinant human arylsulfatase B on the bone and joint involvement in a patient with a severe phenotype of the disease. Before the initiation of ERT, the patient presented with significant range of motion (ROM) limitations at multiple joints. Flexion contractures were noticeable in all joints. After 48 weeks of ERT, improvement in active and passive shoulder flexion, as well as passive elbow and wrist flexion, was noticed. ROM improvements were reflected in patient’s enhanced self-care.
Collapse
|
34
|
Abstract
A decade has passed since the initial report that parenteral use of recombinant human α-L-iduronidase results in amelioration of symptoms in patients with mucopolysaccharidosis type I (MPS I). As a result, MPS I became the first mucopolysaccharide storage disorder to benefit from enzyme replacement therapy (ERT); subsequent ERTs have been approved for MPS II and VI. The ability of lysosomal storage disorders to respond to ERT is unique among genetic disorders and relates to the capability of cells to take up recombinant lysosomal enzymes through cell surface receptors and deliver them to the lysosome, a processed coined as 'cross-correction'. Although the concept of ERT is straightforward, the evaluation of its efficacy in disorders like MPS I is challenging. This article reviews the use of laronidase in the management of MPS I, with a particular emphasis on the unique issues inherent in the evaluation of therapeutics for such a rare, complex and progressive disorder.
Collapse
Affiliation(s)
- Lorne A Clarke
- a Department of Medical Genetics, University of British Columbia, Child and Family Research Institute, 4500 Oak Street, RM C234, Vancouver, British Columbia, V6H-3N1, Canada.
| |
Collapse
|
35
|
Glycosaminoglycan storage disorders: a review. Biochem Res Int 2011; 2012:471325. [PMID: 22013531 PMCID: PMC3195295 DOI: 10.1155/2012/471325] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Accepted: 08/09/2011] [Indexed: 12/21/2022] Open
Abstract
Impaired degradation of glycosaminoglycans (GAGs) with consequent intralysosomal accumulation of undegraded products causes a group of lysosomal storage disorders known as mucopolysaccharidoses (MPSs). Characteristically, MPSs are recognized by increased excretion in urine of partially degraded GAGs which ultimately result in progressive cell, tissue, and organ dysfunction. There are eleven different enzymes involved in the stepwise degradation of GAGs. Deficiencies in each of those enzymes result in seven different MPSs, all sharing a series of clinical features, though in variable degrees. Usually MPS are characterized by a chronic and progressive course, with different degrees of severity. Typical symptoms include organomegaly, dysostosis multiplex, and coarse facies. Central nervous system, hearing, vision, and cardiovascular function may also be affected. Here, we provide an overview of the molecular basis, enzymatic defects, clinical manifestations, and diagnosis of each MPS, focusing also on the available animal models and describing potential perspectives of therapy for each one.
Collapse
|
36
|
Matte U, Lagranha VL, de Carvalho TG, Mayer FQ, Giugliani R. Cell microencapsulation: a potential tool for the treatment of neuronopathic lysosomal storage diseases. J Inherit Metab Dis 2011; 34:983-90. [PMID: 21614584 DOI: 10.1007/s10545-011-9350-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 04/17/2011] [Accepted: 05/04/2011] [Indexed: 02/03/2023]
Abstract
Lysosomal storage disorders (LSD) are monogenic diseases caused by the deficiency of different lysosomal enzymes that degrade complex substrates such as glycosaminoglycans, sphingolipids, and others. As a consequence there is multisystemic storage of these substrates. Most treatments for these disorders are based in the fact that most of these enzymes are soluble and can be internalized by adjacent cells via mannose-6-phosphate receptor. In that sense, these disorders are good candidates to be treated by somatic gene therapy based on cell microencapsulation. Here, we review the existing data about this approach focused on the LSD treatments, the advantages and limitations faced by these studies.
Collapse
Affiliation(s)
- Ursula Matte
- Gene Therapy Center, Experimental Research Center, Hospital de Clínicas, Porto Alegre, RS, Brazil
| | | | | | | | | |
Collapse
|
37
|
de Ru MH, Boelens JJ, Das AM, Jones SA, van der Lee JH, Mahlaoui N, Mengel E, Offringa M, O'Meara A, Parini R, Rovelli A, Sykora KW, Valayannopoulos V, Vellodi A, Wynn RF, Wijburg FA. Enzyme replacement therapy and/or hematopoietic stem cell transplantation at diagnosis in patients with mucopolysaccharidosis type I: results of a European consensus procedure. Orphanet J Rare Dis 2011; 6:55. [PMID: 21831279 PMCID: PMC3170181 DOI: 10.1186/1750-1172-6-55] [Citation(s) in RCA: 161] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 08/10/2011] [Indexed: 11/10/2022] Open
Abstract
Background Mucopolysaccharidosis type I (MPS I) is a lysosomal storage disorder that results in the accumulation of glycosaminoglycans causing progressive multi-organ dysfunction. Its clinical spectrum is very broad and varies from the severe Hurler phenotype (MPS I-H) which is characterized by early and progressive central nervous system (CNS) involvement to the attenuated Scheie phenotype (MPS I-S) with no CNS involvement. Indication, optimal timing, safety and efficacy of the two available treatment options for MPS I, enzyme replacement therapy (ERT) and hematopoietic stem cell transplantation (HSCT), are subject to continuing debate. A European consensus procedure was organized to reach consensus about the use of these two treatment strategies. Methods A panel of specialists, including 8 specialists for metabolic disorders and 7 bone marrow transplant physicians, all with acknowledged expertise in MPS I, participated in a modified Delphi process to develop consensus-based statements on MPS I treatment. Fifteen MPS I case histories were used to initiate the discussion and to anchor decisions around either treatment mode. Before and at the meeting all experts gave their opinion on the cases (YES/NO transplantation) and reasons for their decisions were collected. A set of draft statements on MPS I treatment options composed by a planning committee were discussed and revised during the meeting until full consensus. Results Full consensus was reached on several important issues, including the following: 1) The preferred treatment for patients with MPS I-H diagnosed before age 2.5 yrs is HSCT; 2) In individual patients with an intermediate phenotype HSCT may be considered if there is a suitable donor. However, there are no data on efficacy of HSCT in patients with this phenotype; 3) All MPS I patients including those who have not been transplanted or whose graft has failed may benefit significantly from ERT; 4) ERT should be started at diagnosis and may be of value in patients awaiting HSCT. Conclusions This multidisciplinary consensus procedure yielded consensus on the main issues related to therapeutic choices and research for MPS I. This is an important step towards an international, collaborative approach, the only way to obtain useful evidence in rare diseases.
Collapse
Affiliation(s)
- Minke H de Ru
- Department of Pediatrics and Amsterdam Lysosome Center 'Sphinx', Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Marucha J, Tylki-Szymańska A, Jakóbkiewicz-Banecka J, Piotrowska E, Kloska A, Czartoryska B, Węgrzyn G. Improvement in the range of joint motion in seven patients with mucopolysaccharidosis type II during experimental gene expression-targeted isoflavone therapy (GET IT). Am J Med Genet A 2011; 155A:2257-62. [PMID: 21834048 DOI: 10.1002/ajmg.a.34146] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 04/23/2011] [Indexed: 11/11/2022]
Abstract
Mucopolysaccharidosis type II (MPS II, Hunter disease) is an X chromosome-linked inherited metabolic disease caused by mutations resulting in deficiency of activity of iduronate-2-sulfatase (IDS) and accumulation of undegraded glycosaminoglycans (GAGs), heparan sulfate, and dermatan sulfate. Previous experiments with cell cultures and studies on animal model of MPS II suggested that gene expression-targeted isoflavone therapy (GET IT), based on genistein-mediated reduction of efficiency of GAG synthesis, might be a suitable therapy for this disease. In this report, we demonstrate efficacy of GET IT in connective tissue elasticity, particularly in improving the range of joint motion in seven patients with MPS II after 26 weeks of treatment with an isoflavone extract at the dose corresponding to 5 mg/kg/day of genistein.
Collapse
Affiliation(s)
- Jolanta Marucha
- Department of Metabolic Diseases, The Children's Memorial Health Institute, Warsaw, Poland
| | | | | | | | | | | | | |
Collapse
|