1
|
Mirza MB. Endodontic management of taurodontism in a patient with Morquio syndrome: Case report of a 16-year-old girl. SPECIAL CARE IN DENTISTRY 2024; 44:1581-1588. [PMID: 39039767 DOI: 10.1111/scd.13047] [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: 02/26/2024] [Revised: 04/29/2024] [Accepted: 07/13/2024] [Indexed: 07/24/2024]
Abstract
BACKGROUND AND AIM Mucopolysaccharidosis (MPS) type IV, referred to as Morquio A syndrome (MAS), is a rare genetic disorder characterized by an insufficient level in the storage of glycosaminoglycans within lysosomes. Diagnosis generally depends on clinical examination, skeletal radiographs, and histochemical tests. The condition is characterized by prominent skeletal deformities, limited joint mobility, significant growth impairment, abnormalities in tooth alignment, and defects in tooth enamel. The present clinical case report aims to provide details of a MAS case with peculiar dental findings, including multiple taurodonts where emergency dental treatment was rendered. CASE REPRESENTATION A 16-year-old girl with short stature and prominent facial characteristics reported severe pain in the right maxillary back region. Clinical examination revealed multiple areas of alveolar bone loss, decayed #36, and temporary restoration on #16. Radiographic examination indicated multiple posterior teeth with taurodontism. Past medical history was confirmative of Morquio syndrome. Treating taurodonts endodontically is challenging due to the enlarged pulp chamber, shortened roots, and constricted root canal anatomy. Motorized canal preparation and the use of bioceramic sealer with single cone obturation were done to achieve the best results. CONCLUSION The present case report encountered difficulty locating and negotiating the root canals and establishing the glide path. Using a dental operating microscope and stiff hand files for glide path and short-length Niti rotary files helped achieve the desired results. Although MPS IV is not frequently seen in dental practice, persons with this syndrome can continue to have good oral and overall health if their dental and medical condition if appropriately managed.
Collapse
Affiliation(s)
- Mubashir Baig Mirza
- Conservative Dental Sciences, College of Dentistry, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| |
Collapse
|
2
|
AlSayed M, Arafa D, Al-Khawajha H, Afqi M, Al-Sanna'a N, Sunbul R, Faden M. Consensus-based expert recommendations on the management of MPS IVa and VI in Saudi Arabia. Orphanet J Rare Dis 2024; 19:269. [PMID: 39020431 PMCID: PMC11253461 DOI: 10.1186/s13023-024-03237-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/28/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Mucopolysaccharidosis type IVa (Morquio A syndrome) and mucopolysaccharidosis type VI (Maroteaux-Lamy syndrome) are rare inherited lysosomal storage diseases associated with significant functional impairment and a wide spectrum of debilitating clinical manifestations. These conditions are thought to have higher-than-average prevalence rates in Saudi Arabia due to high rates of consanguineous marriage in the country. There are several unmet needs associated with the management of these diseases in Saudi Arabia. MAIN BODY The aim of this manuscript is to contextualize unmet management needs and provide recommendations to optimize diagnosis, multidisciplinary care delivery, and local data generation in this disease area. An expert panel was assembled comprising seven consultant geneticists from across Saudi Arabia. The Delphi methodology was used to obtain a consensus on statements relating to several aspects of mucopolysaccharidosis types IVa and VI. A consensus was reached for all statements by means of an online, anonymized voting system. The consensus statements pertain to screening and diagnosis, management approaches, including recommendations pertaining to enzyme replacement therapy, and local data generation. CONCLUSION The consensus statements presented provide specific recommendations to improve diagnostic and treatment approaches, promote multidisciplinary care and data sharing, and optimize the overall management of these rare inherited diseases in Saudi Arabia.
Collapse
Affiliation(s)
- Moeenaldeen AlSayed
- Department of Medical Genomics, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia.
| | - Dia Arafa
- Consultant Pediatrician and Medical Genetics, Maternity and Children Hospital, Makkah, Saudi Arabia
| | - Huda Al-Khawajha
- Consultant Pediatrician & Medical Genetics, Maternity and Children Hospital, Al-Ahsa, Saudi Arabia
| | - Manal Afqi
- Clinical Genetics and Metabolic Disorders, Consultant Pediatrician, Maternity and Children Hospital, Madinah, Saudi Arabia
| | - Nouriya Al-Sanna'a
- Clinical Geneticist, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
| | - Rawda Sunbul
- Consultant Pediatrician and Medical Genetics, Qatif Central Hospital, Qatif, Saudi Arabia
| | - Maha Faden
- Genetic Unit, Maternity and Children Hospital, Consultant Pediatrician, Clinical Genetics - Metabolic and Skeletal Dysplasia, King Saud Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
3
|
Ellison S, Parker H, Bigger B. Advances in therapies for neurological lysosomal storage disorders. J Inherit Metab Dis 2023; 46:874-905. [PMID: 37078180 DOI: 10.1002/jimd.12615] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 04/21/2023]
Abstract
Lysosomal Storage Disorders (LSDs) are a diverse group of inherited, monogenic diseases caused by functional defects in specific lysosomal proteins. The lysosome is a cellular organelle that plays a critical role in catabolism of waste products and recycling of macromolecules in the body. Disruption to the normal function of the lysosome can result in the toxic accumulation of storage products, often leading to irreparable cellular damage and organ dysfunction followed by premature death. The majority of LSDs have no curative treatment, with many clinical subtypes presenting in early infancy and childhood. Over two-thirds of LSDs present with progressive neurodegeneration, often in combination with other debilitating peripheral symptoms. Consequently, there is a pressing unmet clinical need to develop new therapeutic interventions to treat these conditions. The blood-brain barrier is a crucial hurdle that needs to be overcome in order to effectively treat the central nervous system (CNS), adding considerable complexity to therapeutic design and delivery. Enzyme replacement therapy (ERT) treatments aimed at either direct injection into the brain, or using blood-brain barrier constructs are discussed, alongside more conventional substrate reduction and other drug-related therapies. Other promising strategies developed in recent years, include gene therapy technologies specifically tailored for more effectively targeting treatment to the CNS. Here, we discuss the most recent advances in CNS-targeted treatments for neurological LSDs with a particular emphasis on gene therapy-based modalities, such as Adeno-Associated Virus and haematopoietic stem cell gene therapy approaches that encouragingly, at the time of writing are being evaluated in LSD clinical trials in increasing numbers. If safety, efficacy and improved quality of life can be demonstrated, these therapies have the potential to be the new standard of care treatments for LSD patients.
Collapse
Affiliation(s)
- S Ellison
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, United Kingdom
| | - H Parker
- Division of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, United Kingdom
| | - B Bigger
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
4
|
Mitchell JJ, Burton BK, Bober MB, Campeau PM, Cohen S, Dosenovic S, Ellaway C, Bhattacharya K, Guffon N, Hinds D, Lail A, Lin SP, Magner M, Raiman J, Schwartz-Sagi L, Stepien KM. Findings from the Morquio A Registry Study (MARS) after 6 years: Long-term outcomes of MPS IVA patients treated with elosulfase alfa. Mol Genet Metab 2022; 137:164-172. [PMID: 36087504 DOI: 10.1016/j.ymgme.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/16/2022] [Accepted: 08/24/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND The Morquio A Registry Study (MARS) is an ongoing, multinational, observational study of patients with MPS IVA. Key objectives of MARS are to characterize the heterogeneity and natural history of disease and to evaluate long-term effectiveness and safety of elosulfase alfa enzyme replacement therapy (ERT). Enrollment began in September 2014; data on medical history, clinical outcomes, and safety assessments are collected as part of routine care. RESULTS As of February 2021, 381 subjects from 17 countries had enrolled in MARS: 58 ERT-naïve subjects and 323 ERT-treated subjects (≥1 infusion), with a mean ERT exposure of 5.5 years (SD 2.8) and median age at first ERT treatment of 9.8 years. ERT-treated subjects were younger at diagnosis (median 3.4 vs 6.5 years) relative to ERT-naïve subjects. Among ERT-treated subjects, urinary keratan sulfate (uKS) levels declined from pre-ERT baseline to last follow-up on treatment (mean % change [95% confidence interval]: -52.5% [-57.5%, -47.4%]; n = 115) and 6-min walk test distance remained stable (mean change: -6.1 [-27.6, 15.5] m; n = 131) over a mean follow-up of 5.5 years. Forced expiratory volume in 1 s (FEV1) and forced vital capacity (FVC) increased in subjects who were < 18 years of age at ERT initiation (mean change: +0.3 [0.1, 0.4] L and + 0.4 [0.3, 0.5] L; mean follow-up: ∼6 years; n = 82) and were stable in subjects ≥18 years (mean change: 0.0 [-0.0, 0.1] L and 0.0 [-0.1, 0.1] L; mean follow-up: 4.6 years; n = 38). Overall, 148 (47.1%) ERT-treated subjects experienced ≥1 adverse event (AE) and 110 subjects (35%) reported ≥1 serious AE. Drug-related AEs were reported in 39 (12.4%) subjects; the most common were hypersensitivity (9 subjects [2.9%]), urticaria (8 subjects [2.5%]), and pyrexia (7 subjects [2.2%]). CONCLUSIONS MARS is the longest and largest observational study of MPS IVA patients to date, with a heterogenous population that is representative of the MPS IVA population overall. Data collected over the first 6 years of MARS provide real-world evidence for long-term stabilization of endurance and respiratory function among ERT-treated patients, with no new safety concerns identified.
Collapse
Affiliation(s)
| | - Barbara K Burton
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.
| | - Michael B Bober
- Nemours/Alfred I. DuPont Hospital for Children, Wilmington, DE, USA.
| | | | | | | | | | | | - Nathalie Guffon
- Reference Centre of Inherited Metabolic Disease, HCL Hospital, Lyon, France.
| | - David Hinds
- BioMarin Pharmaceutical Inc., Novato, CA, USA.
| | - Alice Lail
- BioMarin Pharmaceutical Inc., Novato, CA, USA.
| | | | - Martin Magner
- Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | | | |
Collapse
|
5
|
Grabowski GA, Mistry PK. Therapies for lysosomal storage diseases: Principles, practice, and prospects for refinements based on evolving science. Mol Genet Metab 2022; 137:81-91. [PMID: 35933791 DOI: 10.1016/j.ymgme.2022.07.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 12/25/2022]
Affiliation(s)
- Gregory A Grabowski
- University of Cincinnati College of Medicine, Department of Pediatrics, Department of Molecular Genetics, Biochemistry and Microbiology, United States of America; Division of Human Genetics, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, United States of America.
| | - Pramod K Mistry
- Yale School of Medicine, Department of Medicine, Department of Pediatrics, Department of Cellular & Molecular Physiology, New Haven, CT, United States of America
| |
Collapse
|
6
|
Lee CL, Chuang CK, Chiu HC, Tu RY, Lo YT, Chang YH, Lin SP, Lin HY. Clinical Utility of Elosulfase Alfa in the Treatment of Morquio A Syndrome. Drug Des Devel Ther 2022; 16:143-154. [PMID: 35046639 PMCID: PMC8759989 DOI: 10.2147/dddt.s219433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/07/2021] [Indexed: 11/11/2022] Open
Abstract
Mucopolysaccharidosis type IVA (MPS IVA or Morquio A) is an autosomal recessive disorder and is one of the lysosomal storage diseases. Patients with MPS IVA have a striking skeletal phenotype but normal intellect. The phenotypic continuum of MPS IVA ranges from severe and rapid progress to mild and slow progress. The diagnosis of MPS IVA is usually suspected based on abnormal bone findings and dysplasia on physical examination and radiographic investigation in the preschool years. In the past, only supportive care was available. Due to the early and severe skeletal abnormalities, the orthopedic specialist was usually the main care provider. However, patients need aggressive monitoring and management of their systemic disease. Therefore, they need an interdisciplinary team for their care, comprising medical geneticists, cardiologists, pulmonary specialists, gastroenterologists, otolaryngologists, audiologists, and ophthalmologists. After the US Food and Drug Administration approved elosulfase alfa in 2014, patients older than 5 years could benefit from this treatment. Clinical trials showed clinically meaningful improvements with once-a-week intravenous dosing (2.0 mg/kg per week), significantly improving the 6min walk test, the 3min stair climb test, and respiratory function when compared with placebo. Elosulfase alfa is well-tolerated, and there is a good response indicated by decreasing urine glycosaminoglycans.
Collapse
Affiliation(s)
- Chung-Lin Lee
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan,Institute of Clinical Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan,MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan,Department of Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
| | - 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
| | - Huei-Ching Chiu
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ru-Yi Tu
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yun-Ting Lo
- Department of Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ya-Hui Chang
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan,Department of Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
| | - Shuan-Pei Lin
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan,Department of Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan,Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan,Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan,Correspondence: Shuan-Pei Lin; Hsiang-Yu Lin Department of Pediatrics, MacKay Memorial Hospital, No. 92, Sec. 2, Chung-Shan North Road, Taipei, 10449, TaiwanTel +886-2-2543-3535 ext. 3090; +886-2-2543-3535 ext. 3089Fax +886-2-2543-3642 Email ;
| | - Hsiang-Yu Lin
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan,MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan,Department of Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan,Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
7
|
Stevens B, Kenny T, Thomas S, Morrison A, Jarrett J, Jain M. Elosulfase alfa in the treatment of mucopolysaccharidosis type IVA: insights from the first managed access agreement. Orphanet J Rare Dis 2021; 16:394. [PMID: 34563214 PMCID: PMC8467187 DOI: 10.1186/s13023-021-01876-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 05/20/2021] [Indexed: 11/10/2022] Open
Abstract
Managed access agreements provide a crucial mechanism whereby real-world data can be collected systematically to reduce uncertainty around available clinical and economic data, whilst providing the opportunity to identify patient sub-populations who are most likely to benefit from a new treatment. This manuscript aims to share learnings from the first managed access agreement, which was initiated following positive conditional approval in 2015 from the National Institute for Health and Care Excellence (NICE) for elosulfase alfa, an enzyme replacement therapy for the treatment of mucopolysaccharidosis type IVA (MPS IVA). This managed access agreement enabled the collection of comprehensive real-world data for patients with MPS IVA, with results demonstrating that patients starting elosulfase alfa treatment showed gains similar to those seen in the pivotal trial for outcomes including endurance, respiratory and cardiac function, pain, quality of life measures and urinary keratan sulfate levels. In addition, former trial patients continued to see benefits in both clinical assessments and quality of life/activities of daily living nine years after beginning treatment. Key strengths of the process included recruitment of a high proportion of MPS IVA patients treated in England (72/89 known eligible patients) with a wide range of ages (2-58 years). Participation of a patient organisation (the MPS society) ensured that the patient voice was present throughout the process, whilst a contract research organisation (Rare Disease Research Partners) ensured that patients were represented when interpreting agreement criteria and during patient assessment meetings. Longer-term follow-up will be required for several MPS IVA outcomes (e.g. skeletal measures) to further reduce uncertainty, and continued follow-up of patients who had stopped treatment was found to be challenging. The burden associated with this managed access agreement was found to be high for patients, physicians, patient organisations, NHS England and the manufacturer, therefore costs and benefits of future agreements should be considered carefully before initiation. Through evaluation of the strengths and limitations of this process, it is hoped that learnings from this managed access agreement can be used to inform future agreements.
Collapse
Affiliation(s)
- Bob Stevens
- The MPS Society, Amersham, Buckinghamshire, UK
| | - Tom Kenny
- Rare Disease Research Partners, Amersham, Buckinghamshire, UK
| | | | | | | | - Mohit Jain
- BioMarin Europe Ltd., 10 Bloomsbury Way, London, WC1A 2SL, UK.
| |
Collapse
|
8
|
Frigeni M, Rodriguez-Buritica DF, Saavedra H, Gunther KA, Hillman PR, Balaguru D, Northrup H. The youngest pair of siblings with Mucopolysaccharidosis type IVA to receive enzyme replacement therapy to date: A case report. Am J Med Genet A 2021; 185:3510-3516. [PMID: 34472180 DOI: 10.1002/ajmg.a.62469] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 07/26/2021] [Accepted: 08/05/2021] [Indexed: 11/11/2022]
Abstract
Mucopolysaccharidosis type IVA (OMIM 253000) is an autosomal recessive disorder caused by defective activity of the N-acetylgalactosamine 6-sulfatase (GALNS) enzyme. In 2014, enzyme replacement therapy (ERT) using recombinant human GALNS became available for affected patients. There is a limited number of studies to date that have explored the effect of ERT in infancy and there is also a lack of data assessing the effect of ERT in systems other than the skeletal. Here, we report on the effect of ERT in the youngest pair of siblings treated to date: Patient A, currently 4 years old, who started treatment at the age of 5 months; and Patient B, currently 3 years old, who started treatment at 58 days of life. Moreover, we investigate the effect of early ERT on the cardiovascular system. Our results show that, even when ERT is started before 2 months of age, it cannot fully prevent disease progression. As for the effect of ERT on the cardiovascular system, our preliminary results suggest that early treatment might play a role in preserving a normal left ventricular mass index in affected patients at least up to 1 year, but further observation over time will be required. Overall, this report shows that early diagnosis remains crucial and that prompt initiation of ERT has limited effect in slowing progression of the skeletal phenotype, thus confirming the need for new therapeutic approaches that target the skeletal system in affected patients.
Collapse
Affiliation(s)
- Marta Frigeni
- Division of Medical Genetics, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - David F Rodriguez-Buritica
- Division of Medical Genetics, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Heather Saavedra
- Division of Medical Genetics, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kathryn A Gunther
- Division of Medical Genetics, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Paul R Hillman
- Division of Medical Genetics, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Duraisamy Balaguru
- Division of Pediatric and Congenital Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Division of Cardiology, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Hope Northrup
- Division of Medical Genetics, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
9
|
Kılavuz S, Basaran S, Kor D, Bulut FD, Erdem S, Ballı HT, Dağkıran M, Bisgin A, Mungan HNÖ. Morquio A syndrome and effect of enzyme replacement therapy in different age groups of Turkish patients: a case series. Orphanet J Rare Dis 2021; 16:144. [PMID: 33752727 PMCID: PMC7983100 DOI: 10.1186/s13023-021-01761-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/25/2021] [Indexed: 12/28/2022] Open
Abstract
Background This case series includes longitudinal clinical data of ten patients with Morquio A syndrome from south and southeastern parts of Turkey, which were retrospectively collected from medical records. All patients received enzyme replacement therapy (ERT). Clinical data collected included physical appearance, anthropometric data, neurological and psychological examinations, cardiovascular evaluation, pulmonary function tests, eye and ear-nose-throat examinations, endurance in the 6-min walk test and/or 3-min stair climb test, joint range of motion, and skeletal investigations (X-rays, bone mineral density). Results At the time of ERT initiation, two patients were infants (1.8 and 2.1 years), five were children (3.4–7.1 years), and three were adults (16.5–39.5 years). Patients had up to 4 years follow-up. Most patients had classical Morquio A, based on genotypic and phenotypic data. Endurance was considerably reduced in all patients, but remained relatively stable or increased over time in most cases after treatment initiation. Length/height fell below normal growth curves, except in the two infants who started ERT at ≤ 2.1 years of age. All patients had skeletal and/or joint abnormalities when ERT was started. Follow-up data did not suggest improvements in skeletal abnormalities, except in one of the younger infants. Nine patients had corneal clouding, which resolved after treatment initiation in the two infants, but not in the other patients. Hepatomegaly was reported in seven patients and resolved with treatment in five of them. Other frequent findings at treatment initiation were coarse facial features (N = 9), hearing loss (N = 6), and cardiac abnormalities (N = 6). Cardiac disease deteriorated over time in three patients, but did not progress in the others. Conclusions Overall, this case series with Morquio A patients confirms clinical trial data showing long-term stabilization of endurance after treatment initiation across ages and suggest that very early initiation of ERT optimizes growth outcomes. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-01761-0.
Collapse
Affiliation(s)
- Sebile Kılavuz
- Division of Pediatric Metabolism and Nutrition, Department of Pediatrics, Faculty of Medicine, Çukurova University,, Adana, Turkey
| | - Sibel Basaran
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Deniz Kor
- Division of Pediatric Metabolism and Nutrition, Department of Pediatrics, Faculty of Medicine, Çukurova University,, Adana, Turkey
| | - Fatma Derya Bulut
- Division of Pediatric Metabolism and Nutrition, Department of Pediatrics, Faculty of Medicine, Çukurova University,, Adana, Turkey
| | - Sevcan Erdem
- Division of Pediatric Cardiology, Department of Pediatrics, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Hüseyin Tuğsan Ballı
- Department of Radiology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Muhammed Dağkıran
- Department of Ear, Nose and Throat Diseases, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Atil Bisgin
- Medical Genetics Department of Medical Faculty, Cukurova University AGENTEM (Adana Genetic Diseases Diagnosis and Treatment Center), Adana, Turkey
| | - Halise Neslihan Önenli Mungan
- Division of Pediatric Metabolism and Nutrition, Department of Pediatrics, Faculty of Medicine, Çukurova University, 01130, Sarıçam, Adana, Turkey.
| |
Collapse
|
10
|
Ficicioglu C, Matalon DR, Luongo N, Menello C, Kornafel T, Degnan AJ. Diagnostic journey and impact of enzyme replacement therapy for mucopolysaccharidosis IVA: a sibling control study. Orphanet J Rare Dis 2020; 15:336. [PMID: 33256811 PMCID: PMC7706253 DOI: 10.1186/s13023-020-01618-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Background Mucopolysaccharidosis (MPS) IVA, also known as Morquio A syndrome, is a rare autosomal recessive lysosomal storage disorder caused by a deficiency in the enzyme N-acetylgalactosamine-6-sulfatase. Early recognition, diagnosis, and treatment of this progressive, multisystem disease by enzyme replacement therapy (ERT) can lead to improved outcomes and reduced mortality. Methods This report documents the diagnostic journey and treatment with ERT of three siblings with MPS IVA. Clinical outcome measures included growth, endurance, imaging, cardiac, respiratory, ophthalmology, and laboratory evaluations. Results Three siblings, diagnosed at 14.7, 10.1, and 3.2 years of age, demonstrated clinical improvement with weekly infusions of 2.0 mg/kg elosulfase alfa (Vimizim®, BioMarin Pharmaceutical, Novato, CA, USA). Patient 1 (oldest sibling) and Patient 2 (middle sibling) experienced a diagnostic delay of 8 years 7 months and 4 years after symptom onset, respectively. All three patients demonstrated improvements in growth, 6-min walk distance, joint range of motion, and respiratory function after 30 months of ERT. The treatment was well tolerated without any adverse events. Conclusions This case series highlights the importance of early recognition of the clinical and imaging findings that are initially subtle in MPS IVA. Early treatment with ERT is necessary to slow irreversible disease progression and improve patient outcomes. The oldest sibling experienced improvements in mobility despite severe symptoms resulting from a late diagnosis. When evaluating patients with skeletal anomalies, imaging multiple body regions is recommended. When findings such as anterior beaking of vertebrae or bilateral femoral head dysplasia are present, MPS IVA should be included in the differential diagnosis. Newborn screening must be considered for early detection, accurate diagnosis, and initiation of treatment to reduce morbidity.
Collapse
Affiliation(s)
- Can Ficicioglu
- Division of Human Genetics/Metabolism, Lysosomal Storage Diseases Program, The Children's Hospital of Philadelphia, Perelman School of Medicine, The University of Pennsylvania, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA.
| | - Dena R Matalon
- Stanford University, Lucile Packard Children's Hospital, Palo Alto, CA, USA
| | - Nicole Luongo
- Division of Human Genetics/Metabolism, Lysosomal Storage Diseases Program, The Children's Hospital of Philadelphia, Perelman School of Medicine, The University of Pennsylvania, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Caitlin Menello
- Division of Human Genetics/Metabolism, Lysosomal Storage Diseases Program, The Children's Hospital of Philadelphia, Perelman School of Medicine, The University of Pennsylvania, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Tracy Kornafel
- Division of Human Genetics/Metabolism, Lysosomal Storage Diseases Program, The Children's Hospital of Philadelphia, Perelman School of Medicine, The University of Pennsylvania, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | | |
Collapse
|
11
|
Sosa AC, Kariuki B, Gan Q, Knutsen AP, Bellone CJ, Guzmán MA, Barrera LA, Tomatsu S, Chauhan AK, Armbrecht E, Montaño AM. Oral immunotherapy tolerizes mice to enzyme replacement therapy for Morquio A syndrome. J Clin Invest 2020; 130:1288-1300. [PMID: 31743109 DOI: 10.1172/jci125607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 11/13/2019] [Indexed: 12/14/2022] Open
Abstract
Immune response to therapeutic enzymes poses a detriment to patient safety and treatment outcome. Enzyme replacement therapy (ERT) is a standard therapeutic option for some types of mucopolysaccharidoses, including Morquio A syndrome caused by N-acetylgalactosamine-6-sulfate sulfatase (GALNS) deficiency. Current protocols tolerize patients using cytotoxic immunosuppressives, which can cause adverse effects. Here we show development of tolerance in Morquio A mice via oral delivery of peptide or GALNS for 10 days prior to ERT. Our results show that using an immunodominant peptide (I10) or the complete GALNS enzyme to orally induce tolerance to GALNS prior to ERT resulted in several improvements to ERT in mice: (a) decreased splenocyte proliferation after in vitro GALNS stimulation, (b) modulation of the cytokine secretion profile, (c) decrease in GALNS-specific IgG or IgE in plasma, (d) decreased GAG storage in liver, and (e) fewer circulating immune complexes in plasma. This model could be extrapolated to other lysosomal storage disorders in which immune response hinders ERT.
Collapse
Affiliation(s)
- Angela C Sosa
- Department of Pediatrics, Division of Medical Genetics, School of Medicine, Saint Louis University, St. Louis, Missouri, USA.,Instituto de Errores Innatos del Metabolismo, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Barbara Kariuki
- Department of Pediatrics, Division of Allergy and Immunology
| | - Qi Gan
- Department of Pediatrics, Division of Medical Genetics, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Alan P Knutsen
- Department of Pediatrics, Division of Allergy and Immunology
| | | | - Miguel A Guzmán
- Department of Pathology, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Luis A Barrera
- Instituto de Errores Innatos del Metabolismo, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware, USA
| | - Anil K Chauhan
- Department of Internal Medicine, Division of Rheumatology, School of Medicine
| | | | - Adriana M Montaño
- Department of Pediatrics, Division of Medical Genetics, School of Medicine, Saint Louis University, St. Louis, Missouri, USA.,Department of Biochemistry and Molecular Biology, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| |
Collapse
|
12
|
van der Lee JH, Morton J, Adams HR, Clarke L, Eisengart JB, Escolar ML, Giugliani R, Harmatz P, Hogan M, Kearney S, Muenzer J, Muschol N, Rust S, Saville BR, Semrud-Clikeman M, Wang R, Shapiro E. Therapy development for the mucopolysaccharidoses: Updated consensus recommendations for neuropsychological endpoints. Mol Genet Metab 2020; 131:181-196. [PMID: 32917509 DOI: 10.1016/j.ymgme.2020.08.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 08/24/2020] [Indexed: 01/11/2023]
Abstract
Neurological dysfunction represents a significant clinical component of many of the mucopolysaccharidoses (also known as MPS disorders). The accurate and consistent assessment of neuropsychological function is essential to gain a greater understanding of the precise natural history of these conditions and to design effective clinical trials to evaluate the impact of therapies on the brain. In 2017, an International MPS Consensus Panel published recommendations for best practice in the design and conduct of clinical studies investigating the effects of therapies on cognitive function and adaptive behavior in patients with neuronopathic mucopolysaccharidoses. Based on an International MPS Consensus Conference held in February 2020, this article provides updated consensus recommendations and expands the objectives to include approaches for assessing behavioral and social-emotional state, caregiver burden and quality of life in patients with all mucopolysaccharidoses.
Collapse
Affiliation(s)
- Johanna H van der Lee
- Knowledge Institute of the Dutch Association of Medical Specialists, Utrecht, Netherlands; Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Pediatric Clinical Research Office, Amsterdam, Netherlands
| | | | - Heather R Adams
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Lorne Clarke
- Department of Medical Genetics, Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Julie B Eisengart
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Maria L Escolar
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Roberto Giugliani
- Department of Genetics, UFRGS, and Medical Genetics Service, HPCA, Porto Alegre, Brazil
| | - Paul Harmatz
- UCSF Benioff Children's Hospital Oakland, Oakland, CA, USA
| | | | - Shauna Kearney
- Clinical Paediatric Psychology, Birmingham Children's Hospital NHS Foundation Trust, Birmingham, UK
| | - Joseph Muenzer
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nicole Muschol
- Department of Pediatric, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Stewart Rust
- Paediatric Psychosocial Department, Royal Manchester Children's Hospital, Manchester, UK
| | - Benjamin R Saville
- Berry Consultants LLC, Austin, TX, USA; Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Margaret Semrud-Clikeman
- Department of Medical Genetics, Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Raymond Wang
- Division of Metabolic Disorders, Children's Hospital of Orange County, Orange, CA, USA
| | - Elsa Shapiro
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Shapiro Neuropsychology Consulting LLC, Portland, OR, USA.
| |
Collapse
|
13
|
Long-Term Outcomes of Early Enzyme Replacement Therapy for Mucopolysaccharidosis IV: Clinical Case Studies of Two Siblings. Diagnostics (Basel) 2020; 10:diagnostics10020108. [PMID: 32079294 PMCID: PMC7168314 DOI: 10.3390/diagnostics10020108] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/15/2022] Open
Abstract
Enzyme replacement therapy (ERT) is one of the available therapies for mucopolysaccharidosis (MPS). This study presents a follow-up of two siblings with MPS IVA (Morquio A disease) that received ERT. Both siblings received weekly intravenous infusions of elosulfase alfa for 4.5 years. One sibling (patient 1, P1; male) started therapy at 54 months of age, and the other sibling (patient 2, P2; female) started at 11 months of age. ERT was well-tolerated. In comparison to P1, P2’s growth curves deviated less from the norm. The orthopedic deformities of P1 were more severe than those of P2 and required several surgical corrections. P1’s sleep test at 48 months revealed obstructive sleep apnea, while by the age of 102 months, parameters were normal. P2 never had sleep apnea. Only P1 demonstrated ear, nose, and throat clinical illnesses. In comparison to P1, P2’s physical function was better maintained. In conclusion, ERT was safe in both patients during a 4.5-year follow-up. Although the typical characteristics of this disease were similar in both patients, P1 had a complex clinical course in comparison to P2, which influenced function and quality of life. Therefore, in order to make the most of ERT, it may be more beneficial when initiated at a relatively young age.
Collapse
|
14
|
A Case Report of a Japanese Boy with Morquio A Syndrome: Effects of Enzyme Replacement Therapy Initiated at the Age of 24 Months. Int J Mol Sci 2020; 21:ijms21030989. [PMID: 32024277 PMCID: PMC7037301 DOI: 10.3390/ijms21030989] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Morquio A syndrome, mucopolysaccharidosis type IVA (MPS IVA), is a lysosomal storage disorder caused by the deficient activity of N-acetylgalactosamine-6-sulfatase (GalNac6S), due to alterations in the GALNS gene. This disorder results in marked abnormalities in bones and connective tissues, and affects multiple organs. Here, we describe the clinical course of a Japanese boy with MPS IVA who began enzyme replacement therapy (ERT) at the age of 24 months. Patient: the patient presented for kyphosis treatment at 22 months of age. An X-ray examination revealed dysostosis multiplex. Uronic acids were elevated in the urine and the keratan sulfate (KS) fraction was predominant. The leukocyte GalNac6S enzyme activity was extremely low. The patient exhibited the c.463G > A (p.Gly155Arg) mutation in GALNS. Based on these findings, his disease was diagnosed as classical (severe) Morquio A syndrome. An elosulfase alfa infusion was initiated at the age of 24 months. The patient’s body height improved from −2.5 standard deviation (SD) to −2 SD and his physical activity increased during the first 9 months on ERT. However, he gradually developed paralysis in the lower legs with declining growth velocity, which required cervical decompression surgery in the second year of the ERT. The mild mitral regurgitation, serous otitis media, and mild hearing loss did not progress during treatment. Conclusion: early initiation of the elosulfase alfa to our patient showed good effects on the visceral system and muscle strength, while its effect on bones appeared limited. Careful observation is necessary to ensure timely surgical intervention for skeletal disorders associated with neurological symptoms. Centralized and multidisciplinary management is essential to improve the prognosis of pediatric patients with MPS IVA.
Collapse
|
15
|
Bhattacharya K, Balasubramaniam S, Murray K, Peters H, Ketteridge D, Inwood A, Lee J, Ellaway C, Owens P, Wong M, Ly C, McGill J. Safety and Efficacy of Elosulfase Alfa in Australian Patients with Morquio a Syndrome: A Phase 3b Study. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2020. [DOI: 10.1590/2326-4594-jiems-2020-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
| | | | | | - Heidi Peters
- The Royal Children's Hospital Melbourne, Australia
| | | | | | - Joy Lee
- The Royal Children's Hospital Melbourne, Australia
| | - Carolyn Ellaway
- Sydney Children's Hospitals Network, Australia; University of Sydney, Australia
| | - Penny Owens
- Sydney Children's Hospitals Network, Australia
| | - Melanie Wong
- University of Sydney, Australia; Children's Hospital at Westmead, Australia
| | | | - Jim McGill
- Queensland Children's Hospital, Australia
| |
Collapse
|
16
|
Enzyme replacement therapy for mucopolysaccharidoses; past, present, and future. J Hum Genet 2019; 64:1153-1171. [PMID: 31455839 DOI: 10.1038/s10038-019-0662-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 11/08/2022]
Abstract
Mucopolysaccharidoses (MPS) are a group of lysosomal storage disorders, which lack an enzyme corresponding to the specific type of MPS. Enzyme replacement therapy (ERT) has been the standard therapeutic option for some types of MPS because of the ability to start immediate treatment with feasibility and safety and to improve prognosis. There are several disadvantages for current ERT, such as limited impact to the brain and avascular cartilage, weekly or biweekly infusions lasting 4-5 h, the immune response against the infused enzyme, a short half-life, and the high cost. Clinical studies of ERT have shown limited efficacy in preventing or resolving progression in neurological, cardiovascular, and skeletal diseases. One focus is to penetrate the avascular cartilage area to at least stabilize, if not reverse, musculoskeletal diseases. Although early intervention in some types of MPS has shown improvements in the severity of skeletal dysplasia and stunted growth, this limits the desired effect of ameliorating musculoskeletal disease progression to young MPS patients. Novel ERT strategies are under development to reach the brain: (1) utilizing a fusion protein with monoclonal antibody to target a receptor on the BBB, (2) using a protein complex from plant lectin, glycan, or insulin-like growth factor 2, and (3) direct infusion across the BBB. As for MPS IVA and VI, bone-targeting ERT will be an alternative to improve therapeutic efficacy in bone and cartilage. This review summarizes the effect and limitations on current ERT for MPS and describes the new technology to overcome the obstacles of conventional ERT.
Collapse
|
17
|
Akyol MU, Alden TD, Amartino H, Ashworth J, Belani K, Berger KI, Borgo A, Braunlin E, Eto Y, Gold JI, Jester A, Jones SA, Karsli C, Mackenzie W, Marinho DR, McFadyen A, McGill J, Mitchell JJ, Muenzer J, Okuyama T, Orchard PJ, Stevens B, Thomas S, Walker R, Wynn R, Giugliani R, Harmatz P, Hendriksz C, Scarpa M. Recommendations for the management of MPS IVA: systematic evidence- and consensus-based guidance. Orphanet J Rare Dis 2019; 14:137. [PMID: 31196221 PMCID: PMC6567385 DOI: 10.1186/s13023-019-1074-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/17/2019] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Mucopolysaccharidosis (MPS) IVA or Morquio A syndrome is an autosomal recessive lysosomal storage disorder (LSD) caused by deficiency of the N-acetylgalactosamine-6-sulfatase (GALNS) enzyme, which impairs lysosomal degradation of keratan sulphate and chondroitin-6-sulphate. The multiple clinical manifestations of MPS IVA present numerous challenges for management and necessitate the need for individualised treatment. Although treatment guidelines are available, the methodology used to develop this guidance has come under increased scrutiny. This programme was conducted to provide evidence-based, expert-agreed recommendations to optimise management of MPS IVA. METHODS Twenty six international healthcare professionals across multiple disciplines, with expertise in managing MPS IVA, and three patient advocates formed the Steering Committee (SC) and contributed to the development of this guidance. Representatives from six Patient Advocacy Groups (PAGs) were interviewed to gain insights on patient perspectives. A modified-Delphi methodology was used to demonstrate consensus among a wider group of healthcare professionals with experience managing patients with MPS IVA and the manuscript was evaluated against the validated Appraisal of Guidelines for Research and Evaluation (AGREE II) instrument by three independent reviewers. RESULTS A total of 87 guidance statements were developed covering five domains: (1) general management principles; (2) recommended routine monitoring and assessments; (3) disease-modifying interventions (enzyme replacement therapy [ERT] and haematopoietic stem cell transplantation [HSCT]); (4) interventions to support respiratory and sleep disorders; (5) anaesthetics and surgical interventions (including spinal, limb, ophthalmic, cardio-thoracic and ear-nose-throat [ENT] surgeries). Consensus was reached on all statements after two rounds of voting. The overall guideline AGREE II assessment score obtained for the development of the guidance was 5.3/7 (where 1 represents the lowest quality and 7 represents the highest quality of guidance). CONCLUSION This manuscript provides evidence- and consensus-based recommendations for the management of patients with MPS IVA and is for use by healthcare professionals that manage the holistic care of patients with the intention to improve clinical- and patient-reported outcomes and enhance patient quality of life. It is recognised that the guidance provided represents a point in time and further research is required to address current knowledge and evidence gaps.
Collapse
Affiliation(s)
| | - Tord D. Alden
- Department of Neurosurgery, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Hernan Amartino
- Child Neurology Department, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Jane Ashworth
- Department of Paediatric Ophthalmology, Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Kumar Belani
- Department of Anesthesiology, University of Minnesota, Minneapolis, MN USA
| | - Kenneth I. Berger
- Departments of Medicine and Neuroscience and Physiology, New York University School of Medicine, André Cournand Pulmonary Physiology Laboratory, Bellevue Hospital, New York, NY USA
| | - Andrea Borgo
- Orthopaedics Clinic, Padova University Hospital, Padova, Italy
| | - Elizabeth Braunlin
- Division of Pediatric Cardiology, University of Minnesota, Minneapolis, MN USA
| | - Yoshikatsu Eto
- Advanced Clinical Research Centre, Institute of Neurological Disorders, Kanagawa, Japan and Department of Paediatrics/Gene Therapy, Tokyo Jikei University School of Medicine, Tokyo, Japan
| | - Jeffrey I. Gold
- Keck School of Medicine, Departments of Anesthesiology, Pediatrics, and Psychiatry & Behavioural Sciences, Children’s Hospital Los Angeles, Department of Anesthesiology Critical Care Medicine, 4650 Sunset Boulevard, Los Angeles, CA USA
| | - Andrea Jester
- Hand and Upper Limb Service, Department of Plastic Surgery, Birmingham Women’s and Children’s Hospital, Birmingham, UK
| | - Simon A. Jones
- Willink Biochemical Genetic Unit, Manchester Centre for Genomic Medicine, St Mary’s Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Cengiz Karsli
- Department of Anesthesiology and Pain Medicine, The Hospital for Sick Children, Toronto, Canada
| | - William Mackenzie
- Department of Orthopedics, Nemours/Alfred I, Dupont Hospital for Children, Wilmington, DE USA
| | - Diane Ruschel Marinho
- Department of Ophthalmology, UFRGS, and Ophthalmology Service, HCPA, Porto Alegre, Brazil
| | | | - Jim McGill
- Department of Metabolic Medicine, Queensland Children’s Hospital, Brisbane, Australia
| | - John J. Mitchell
- Division of Pediatric Endocrinology, Montreal Children’s Hospital, Montreal, QC Canada
| | - Joseph Muenzer
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Torayuki Okuyama
- Department of Clinical Laboratory Medicine, National Centre for Child Health and Development, Tokyo, Japan
| | - Paul J. Orchard
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN USA
| | | | | | - Robert Walker
- Department of Paediatric Anaesthesia, Royal Manchester Children’s Hospital, Manchester, UK
| | - Robert Wynn
- Department of Paediatric Haematology, Royal Manchester Children’s Hospital, Manchester, UK
| | - Roberto Giugliani
- Department of Genetics, UFRGS, and Medical Genetics Service, HCPA, Porto Alegre, Brazil
| | - Paul Harmatz
- UCSF Benioff Children’s Hospital Oakland, Oakland, CA USA
| | - Christian Hendriksz
- Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
| | - Maurizio Scarpa
- Center for Rare Diseases at Host Schmidt Kliniken, Wiesbaden, Germany and Department of Paediatrics University of Padova, Padova, Italy
| |
Collapse
|
18
|
Doherty C, Stapleton M, Piechnik M, Mason RW, Mackenzie WG, Yamaguchi S, Kobayashi H, Suzuki Y, Tomatsu S. Effect of enzyme replacement therapy on the growth of patients with Morquio A. J Hum Genet 2019; 64:625-635. [PMID: 31019230 DOI: 10.1038/s10038-019-0604-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/22/2019] [Accepted: 04/08/2019] [Indexed: 01/07/2023]
Abstract
Mucopolysaccharidosis IVA (MPS IVA) is a degenerative systemic skeletal dysplasia, in which children exhibit marked short stature and become physically handicapped. This study evaluated the growth patterns of patients treated with enzyme replacement therapy (ERT), compared with those of untreated patients. Cross-sectional and longitudinal data of heights and weights were collected from 128 MPS IVA patients and compared with the growth charts of MPS IVA. Twelve patients (six males, six females) starting ERT before 5 years old were treated for at least 2 years. Six out of 12 patients (50%) with ERT over 2 years stopped growing between 94 and 98 cm (mean height of 95.1 ± 2.2 cm) from 5.0 years to 9.0 years of age (mean age of 6.2 ± 1.6 years). The other patients, except one attenuated case, exhibited a marked slow growth velocity from 3.6 years to 7.7 years. Treated and untreated patients with severe phenotype reached their final heights by ~10 years of age. Patients treated with ERT exhibited a reduced pubertal growth spurt analogous to their untreated counterparts, which contributes to the marked short stature associated with MPS IVA. Compared with the growth charts for untreated patients, patients treated with ERT did not show any significant increase in growth in any age group. Overall, ERT-treated patients do not experience growth improvement and continue to exhibit poor growth despite early ERT intervention before 5 years of age. These findings indicate that current intravenous ERT is ineffective at correcting abnormal growth in MPS IVA.
Collapse
Affiliation(s)
- Caitlin Doherty
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA.,University of Delaware, Newark, DE, USA
| | - Molly Stapleton
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA.,University of Delaware, Newark, DE, USA
| | - Matthew Piechnik
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA.,University of Delaware, Newark, DE, USA
| | - Robert W Mason
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA.,University of Delaware, Newark, DE, USA
| | | | - Seiji Yamaguchi
- Department of Pediatrics, Shimane University, Shimane, Japan
| | | | - Yasuyuki Suzuki
- Medical Education Development Center, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA. .,Department of Pediatrics, Shimane University, Shimane, Japan. .,Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
19
|
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
|
20
|
Shemesh E, Deroma L, Hendriksz CJ, Hollak C, Krishan A. Enzyme replacement therapy for mucopolysaccharidosis type IV (Morquio syndrome). Hippokratia 2018. [DOI: 10.1002/14651858.cd012961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Elad Shemesh
- Institute of Child Health, University of Liverpool; c/o Cochrane Cystic Fibrosis & Genetic Disorders Review Group; Alder Hey Children's NHS Foundation Trust Eaton Road Liverpool UK L12 2AP
| | - Laura Deroma
- University Hospital "Santa Maria della Misericordia"; Regional Coordinator Centre for Rare Diseases; Piazzale Santa Maria della Misericordia 15 Udine Italy 33100
| | - Christian J Hendriksz
- Salford Royal NHS Foundation Trust; Adult Inherited Metabolic Disorders, The Mark Holland Metabolic Unit; Stott Lane Salford UK M6 8HD
| | - Carla Hollak
- Academic Medical Center; Department of Internal Medicine, Division of Endocrinology & Metabolism; Postbus 22660 Amsterdam Netherlands 1100 DD
| | - Ashma Krishan
- University of Edinburgh; Edinburgh Clinical Trials Unit; Outpatients Building, 2nd Floor Western General Hospital, Crewe Road South Edinburgh UK EH4 2XU
| |
Collapse
|
21
|
Puckett Y, Mulinder H, Montaño AM. Enzyme Replacement Therapy with Elosulfase alfa for Mucopolysaccharidosis IVA (Morquio A Syndrome): Milestones and Challenges. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2017.1366900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Yana Puckett
- Department of General Surgery, Texas Tech University, School of Medicine, Lubbock, TX, USA
| | - Holly Mulinder
- Department of General Surgery, Texas Tech University, School of Medicine, Lubbock, TX, USA
| | - Adriana M. Montaño
- Department of Pediatrics, Saint Louis University, School of Medicine, Edward A. Doisy Research Center, St. Louis, MO, USA
- Department of Biochemistry and Molecular Biology, Saint Louis University, School of Medicine, Edward A. Doisy Research Center, St. Louis, MO, USA
| |
Collapse
|
22
|
Khan S, Alméciga-Díaz CJ, Sawamoto K, Mackenzie WG, Theroux MC, Pizarro C, Mason RW, Orii T, Tomatsu S. Mucopolysaccharidosis IVA and glycosaminoglycans. Mol Genet Metab 2017; 120:78-95. [PMID: 27979613 PMCID: PMC5293636 DOI: 10.1016/j.ymgme.2016.11.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/16/2016] [Accepted: 11/16/2016] [Indexed: 12/21/2022]
Abstract
Mucopolysaccharidosis IVA (MPS IVA; Morquio A: OMIM 253000) is a lysosomal storage disease with an autosomal recessive trait caused by the deficiency of N-acetylgalactosamine-6-sulfate sulfatase. Deficiency of this enzyme leads to accumulation of specific glycosaminoglycans (GAGs): chondroitin-6-sulfate (C6S) and keratan sulfate (KS). C6S and KS are mainly produced in the cartilage. Therefore, the undegraded substrates are stored primarily in cartilage and in its extracellular matrix (ECM), leading to a direct impact on cartilage and bone development, and successive systemic skeletal dysplasia. Chondrogenesis, the earliest phase of skeletal formation, is maintained by cellular interactions with the ECM, growth and differentiation factors, signaling pathways, and transcription factors in a temporal-spatial manner. In patients with MPS IVA, the cartilage is disrupted at birth as a consequence of abnormal chondrogenesis and/or endochondral ossification. The unique skeletal features are distinguished by a disproportional short stature, odontoid hypoplasia, spinal cord compression, tracheal obstruction, pectus carinatum, kyphoscoliosis, platyspondyly, coxa valga, genu valgum, waddling gait, and laxity of joints. In spite of many descriptions of these unique clinical features, delay of diagnosis still happens. The pathogenesis and treatment of systemic skeletal dysplasia in MPS IVA remains an unmet challenge. In this review article, we comprehensively describe historical aspect, property of GAGs, diagnosis, screening, pathogenesis, and current and future therapies of MPS IVA.
Collapse
Affiliation(s)
- Shaukat Khan
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Carlos J Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| | - Kazuki Sawamoto
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - William G Mackenzie
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Mary C Theroux
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Christian Pizarro
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Robert W Mason
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Tadao Orii
- Department of Pediatrics, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States; Department of Pediatrics, Gifu University, Gifu, Japan; Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
23
|
Hendriksz CJ. Elosulfase alfa (BMN 110) for the treatment of mucopolysaccharidosis IVA (Morquio A Syndrome). Expert Rev Clin Pharmacol 2016; 9:1521-1532. [PMID: 27855521 DOI: 10.1080/17512433.2017.1260000] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Morquio A syndrome is a rare, autosomal recessive, lysosomal storage disorder caused by a deficiency in the enzyme N-acetylgalactosamine-6-sulfatase (GALNS). In 2014, the use of recombinant human GALNS, elosulfase alfa, was approved in the European Union, Canada, the United States, Australia, and Brazil for the treatment of Morquio A syndrome. Elosulfase alfa is administered intravenously once-weekly at a dose of 2.0 mg/kg. Areas covered: This is a review of the efficacy, safety and tolerability, pharmacokinetics and pharmacodynamics, and other outcomes of elosulfase alfa treatment of patients with Morquio A. A discussion of other treatment considerations, limitations, and future directions in the use of elosulfase alfa is provided. Expert commentary: Pharmacokinetic studies outside of clinical trials and in 'real-world' clinical settings need to be performed. We cannot currently predict which patient is going to respond well to enzyme replacement therapy; thus, all patients should be given the option to receive treatment for at least 12 months. Additionally, accurate biomarkers for evaluating disease state and drug responsiveness would greatly aid in the treatment of patients with Morquio A. In addition, improved and innovative daily lifestyle measures are greatly needed to adequately measure clinical response and true impact on quality of life.
Collapse
Affiliation(s)
- Christian J Hendriksz
- a Paediatrics and Child Health , University of Pretoria , Steve Biko Academic Unit, Pretoria , South Africa
| |
Collapse
|
24
|
Do Cao J, Wiedemann A, Quinaux T, Battaglia-Hsu SF, Mainard L, Froissart R, Bonnemains C, Ragot S, Leheup B, Journeau P, Feillet F. 30 months follow-up of an early enzyme replacement therapy in a severe Morquio A patient: About one case. Mol Genet Metab Rep 2016; 9:42-45. [PMID: 27761411 PMCID: PMC5065040 DOI: 10.1016/j.ymgmr.2016.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/01/2016] [Accepted: 10/01/2016] [Indexed: 11/25/2022] Open
Abstract
Patients under 5 years were not evaluated in the phase-3 study for enzyme replacement therapy (ERT) in MPS IV A. Here we describe the evolution of a severe Morquio A pediatric patient who was diagnosed at 19 months old and treated by ERT at 21 months old for the next 30 months. Applying the standard ERT protocol on this very young patient appeared to reduce his urinary excretion of glycosaminoglycans (GAGs); the improvements in both the 6 minute-walk test (6MWT) and the stair climb test, however, were no different than those reported in the nature history study. Additionally, this young patient experienced many ERT-associated side effects, and as a result a specific corticosteroid protocol (1 mg/kg of betamethasone the day before and 1 h before the ERT infusion) was given to avoid adverse events. Under these treatments, the height of this patient increased during the first year of the ERT although no more height gain was observed thereafter for 18 months. However, despite of ERT, his bone deformities (including severe pectus carinatum) actually worsened and his medullar cervical spine compression showed no improvement (thus needed decompression surgery). CONCLUSION early ERT treatment did not improve the bone outcome in this severe MPS IV A patient after the 30 months-long treatment. A longer term follow up is required to further assess the efficacy of ERT on both the motor and the respiratory function of the patient.
Collapse
Affiliation(s)
- J Do Cao
- Reference Center for Inborn Errors of Metabolism, University Children's Hospital, 5 rue du Morvan, 54511 Vandoeuvre-les-Nancy, France
| | - A Wiedemann
- Reference Center for Inborn Errors of Metabolism, University Children's Hospital, 5 rue du Morvan, 54511 Vandoeuvre-les-Nancy, France
| | - T Quinaux
- Reference Center for Inborn Errors of Metabolism, University Children's Hospital, 5 rue du Morvan, 54511 Vandoeuvre-les-Nancy, France
| | - S F Battaglia-Hsu
- Reference Center for Inborn Errors of Metabolism, University Children's Hospital, 5 rue du Morvan, 54511 Vandoeuvre-les-Nancy, France
| | - L Mainard
- Radiology Department, University Children's Hospital, Vandoeuvre-les-Nancy, France
| | - R Froissart
- Biochemistry and Molecular Biology, University Hospital, Lyon, France
| | - C Bonnemains
- Reference Center for Inborn Errors of Metabolism, University Children's Hospital, 5 rue du Morvan, 54511 Vandoeuvre-les-Nancy, France
| | - S Ragot
- Rehabilitation center, University Children's Hospital, Vandoeuvre-les-Nancy, France
| | - B Leheup
- Genetic Department, University Children's Hospital, Vandoeuvre-les-Nancy, France
| | - P Journeau
- Pediatric Orthopedic surgery department, University Children's Hospital, Vandoeuvre-les-Nancy, France
| | - F Feillet
- Reference Center for Inborn Errors of Metabolism, University Children's Hospital, 5 rue du Morvan, 54511 Vandoeuvre-les-Nancy, France
| |
Collapse
|
25
|
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: 3.9] [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
|
26
|
Regier DS, Tanpaiboon P. Role of elosulfase alfa in mucopolysaccharidosis IVA. APPLICATION OF CLINICAL GENETICS 2016; 9:67-74. [PMID: 27366102 PMCID: PMC4913537 DOI: 10.2147/tacg.s69080] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Mucopolysaccharidosis type IVA (MPS IVA or Morquio A) is an autosomal recessive lysosomal storage disease which results in a striking skeletal phenotype, but does not negatively impact the intellect of the patient. MPS IVA has a phenotypic continuum that ranges from a severe and rapidly progressing form to a slowly progressive form. The clinical diagnosis is often made in the preschool years based on abnormal bone findings on physical examination and dysplasia on radiographic imaging. Supportive care has been the mainstay in caring for patients. Orthopedic physicians often form the core of the care team due to the early and severe skeletal abnormalities; however, systemic disease is common and requires aggressive monitoring and management. Interdisciplinary care teams often consist of medical geneticists, cardiologists, pulmonary specialists, gastroenterologists, otolaryngologists, audiologists, and ophthalmologists. With the US Food and Drug Administration's approval of elosulfase alfa, patients >5 years of age now have access to this medication from the time of diagnosis. The clinical trial with once weekly intravenous dosing (2.0 mg/kg per week) showed improvement in the 6-minute walk test. The composite end point analysis to evaluate the combining changes from baseline in 6-minute walk test, 3-minute stair climb test, and respiratory function showed that at a dose of 2.0 mg/kg per week, subjects performed better when compared to placebo. This indication was clinically meaningful in the treatment group. The treatment was generally well tolerated, and the uncommon infusion reactions responded well to traditional enzyme replacement therapy infusion reaction management algorithms. Currently, clinical trials are underway to determine the efficacy and safety in MPS IVA patients <5 years of age.
Collapse
Affiliation(s)
- Debra S Regier
- Division of Genetics and Metabolism, Children's National Medical Center, Washington, DC, USA
| | - Pranoot Tanpaiboon
- Division of Genetics and Metabolism, Children's National Medical Center, Washington, DC, USA
| |
Collapse
|
27
|
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
|
28
|
Lin HY, Chuang CK, Chen MR, Lin SM, Hung CL, Chang CY, Chiu PC, Tsai WH, Niu DM, Tsai FJ, Lin SJ, Hwu WL, Lin JL, Lin SP. Cardiac structure and function and effects of enzyme replacement therapy in patients with mucopolysaccharidoses I, II, IVA and VI. Mol Genet Metab 2016; 117:431-7. [PMID: 26899310 DOI: 10.1016/j.ymgme.2016.02.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 02/15/2016] [Accepted: 02/15/2016] [Indexed: 11/26/2022]
Abstract
BACKGROUND While enzyme replacement therapy (ERT) has been shown to improve endurance and joint mobility for patients with mucopolysaccharidoses (MPS) I, II, IVA and VI, the impact of ERT on cardiac abnormalities remains uncertain. METHODS Medical records and echocardiograms of 28 Taiwanese MPS patients (9 with MPS I, 7 with MPS II, 7 with MPS IVA, and 5 with MPS VI) treated with ERT for 1-10.8years were retrospectively reviewed. RESULTS At start of ERT, z scores>2 were identified in 46% and 75% for left ventricular mass index (LVMI) and interventricular septum thickness in diastole (IVSd) in these patients, respectively. Twenty-four patients (86%) had valvular heart disease. After ERT, the mean IVSd z score of all patients decreased significantly from 3.87 to 2.57 (p=0.016). For 11 patients starting ERT before 12years of age, z scores for both LVMI and IVSd decreased significantly (p<0.01) after ERT. However, the condition of valve regurgitation or stenosis did not show improvement despite ERT. CONCLUSIONS ERT was shown to be an effective therapy for reducing cardiac hypertrophy, with best results seen when ERT was started at an early age. ERT, however, had little impact on valvular heart disease.
Collapse
Affiliation(s)
- Hsiang-Yu Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan; Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan; Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan; Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Kuang Chuang
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan; Medical College, Fu-Jen Catholic University, Taipei, Taiwan; Institute of Biotechnology, National Taipei University of Technology, Taipei, Taiwan
| | - Ming-Ren Chen
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan; Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan; Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Shan-Miao Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan; Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan; Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Chung-Lieh Hung
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Division of Cardiology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chia-Ying Chang
- Department of Pediatrics, Mackay Memorial Hospital, Hsinchu, Taiwan
| | - Pao Chin Chiu
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wen-Hui Tsai
- Department of Pediatrics, Chi Mei Medical Center, Tainan, Taiwan
| | - Dau-Ming Niu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Fuu-Jen Tsai
- Department of Pediatrics, China Medical University Hospital, Taichung, Taiwan
| | - Shio Jean Lin
- Department of Pediatrics, Chi Mei Medical Center, Tainan, Taiwan
| | - Wuh-Liang Hwu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Ju-Li Lin
- Division of Genetics and Endocrinology, Department of Pediatrics, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Shuan-Pei Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan; Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan; Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan; Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan.
| |
Collapse
|
29
|
Burton BK, Berger KI, Lewis GD, Tarnopolsky M, Treadwell M, Mitchell JJ, Muschol N, Jones SA, Sutton VR, Pastores GM, Lau H, Sparkes R, Genter F, Shaywitz AJ, Harmatz P. Safety and physiological effects of two different doses of elosulfase alfa in patients with morquio a syndrome: A randomized, double-blind, pilot study. Am J Med Genet A 2015; 167A:2272-81. [PMID: 26069231 PMCID: PMC4744659 DOI: 10.1002/ajmg.a.37172] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 05/07/2015] [Indexed: 11/25/2022]
Abstract
The primary treatment outcomes of a phase 2, randomized, double‐blind, pilot study evaluating safety, physiological, and pharmacological effects of elosulfase alfa in patients with Morquio A syndrome are herewith presented. Patients aged ≥7 years and able to walk ≥200 m in the 6‐min walk test (6MWT) were randomized to elosulfase alfa 2.0 or 4.0 mg/kg/week for 27 weeks. The primary objective was to evaluate the safety of both doses. Secondary objectives were to evaluate effects on endurance (6MWT and 3‐min stair climb test [3MSCT]), exercise capacity (cardio‐pulmonary exercise test [CPET]), respiratory function, muscle strength, cardiac function, pain, and urine keratan sulfate (uKS) levels, and to determine pharmacokinetic parameters. Twenty‐five patients were enrolled (15 randomized to 2.0 mg/kg/week and 10 to 4.0 mg/kg/week). No new or unexpected safety signals were observed. After 24 weeks, there were no improvements versus baseline in the 6MWT, yet numerical improvements were seen in the 3MSCT with 4.0 mg/kg/week. uKS and pharmacokinetic data suggested no linear relationship over the 2.0–4.0 mg/kg dose range. Overall, an abnormal exercise capacity (evaluated in 10 and 5 patients in the 2.0 and 4.0 mg/kg/week groups, respectively), impaired muscle strength, and considerable pain were observed at baseline, and there were trends towards improvements in all domains after treatment. In conclusion, preliminary data of this small study in a Morquio A population with relatively good endurance confirmed the acceptable safety profile of elosulfase alfa and showed a trend of increased exercise capacity and muscle strength and decreased pain. © 2015 The Authors. American Journal of Medical Genetics Part A Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Barbara K Burton
- Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois.,Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | | | | | | | | | - Nicole Muschol
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon A Jones
- Willink Unit, St. Mary's Hospital, CMFT, MAHSC, University of Manchester, Manchester, United Kingdom
| | - V Reid Sutton
- Texas Children's Hospital, Houston, Texas.,Baylor College of Medicine, Houston, Texas
| | - Gregory M Pastores
- New York University School of Medicine, New York, New York.,Mater Misericordiae University Hospital, Dublin, Ireland
| | - Heather Lau
- New York University School of Medicine, New York, New York
| | | | - Fred Genter
- BioMarin Pharmaceutical Inc., Novato, California
| | | | - Paul Harmatz
- UCSF Benioff Children's Hospital Oakland, Oakland, California
| |
Collapse
|