1
|
Santovito LS, Bonanno S, Pasanisi MB, Gallone A, Ricci F, Tramacere I, Zanin R, Previtali SC, Maggi L. Natural history of skeletal muscle laminopathies: a 2-year prospective study. Neuromuscul Disord 2025; 47:105256. [PMID: 39657283 DOI: 10.1016/j.nmd.2024.105256] [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: 07/13/2024] [Revised: 11/05/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024]
Abstract
Skeletal muscle laminopathies (SMLs) are rare disorders characterized by skeletal muscle involvement caused by mutations in LMNA gene. To date, the natural history of SMLs has not been clearly elucidated. Through a 2-year prospective study, we aimed to describe the natural history of SMLs. We enrolled 26 SMLs patients, assessed with: North Star Ambulatory Assessment scale (NSAA), timed tests, manual muscle testing, joint range of motion, six-minutes walking test (6MWT); respiratory evaluation including forced vital capacity (FVC) and forced expiratory volume at 1 second (FEV1); individualized neuromuscular quality of life (INQoL). Muscular performance with the aforementioned tools significantly correlated with phenotypes at the baseline, showing the worse outcome in those with autosomal dominant Emery-Dreifuss muscular dystrophy as compared to limb girdle phenotype. NSAA score significantly (p = 0.0005) worsened during the 2-year follow-up. Moreover, the respiratory function through FVC and FEV1 significantly (p = 0.0086 and p = 0.0290, respectively) deteriorated over the follow-up period. 6MWT, INQoL and timed tests did not significantly change, as well as ankle, knee, and elbow contractures. This study showed a slow progression of motor and respiratory function in SMLs patients over a period of 2 years.
Collapse
Affiliation(s)
- Luca Spiro Santovito
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, USA; Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Silvia Bonanno
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Maria Barbara Pasanisi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy; Multiple Sclerosis Center, IRCCS Fondazione don Carlo Gnocchi ONLUS, Milan, Italy
| | - Annamaria Gallone
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Federica Ricci
- Department of Public Health and Pediatric Sciences, Section of Child and Adolescent Neuropsychiatry, University of Turin, Italy
| | - Irene Tramacere
- Department of Research and Clinical Development, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Riccardo Zanin
- Developmental Neurology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Stefano Carlo Previtali
- InSpe and Division of Neuroscience, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Maggi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| |
Collapse
|
2
|
de Laat ECM, Houwen-van Opstal SLS, Bouman K, van Doorn JLM, Cameron D, van Alfen N, Dittrich ATM, Kamsteeg EJ, Smeets HJM, Groothuis JT, Erasmus CE, Voermans NC. A 5-year natural history study in LAMA2-related muscular dystrophy and SELENON-related myopathy: the Extended LAST STRONG study. BMC Neurol 2024; 24:409. [PMID: 39443859 PMCID: PMC11515704 DOI: 10.1186/s12883-024-03852-4] [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: 08/16/2024] [Accepted: 09/03/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND SELENON-related myopathy (SELENON-RM) is a rare congenital myopathy characterized by slowly progressive axial muscle weakness, rigidity of the spine, scoliosis, and respiratory insufficiency. Laminin-a2-related muscular dystrophy (LAMA2-MD) has a similar clinical phenotype, which ranges from severe, early-onset congenital muscular dystrophy type 1A (MDC1A) to milder forms presenting as childhood- or adult-onset limb-girdle type muscular dystrophy. The first 1.5-year natural history follow-up showed that 90% of the patients had low bone quality, respiratory impairments were found in all SELENON-RM and most of the LAMA2-MD patients, and many had cardiac risk factors. However, further extensive knowledge on long-term natural history data, and clinical and functional outcome measures is needed to reach trial readiness. Therefore, we extended the natural history study with 3- and 5-year follow-up visits (Extended LAST STRONG). METHODS The Extended LAST STRONG is a long-term natural history study in Dutch-speaking patients of all ages diagnosed with genetically confirmed SELENON-RM or LAMA2-MD, starting in September 2023. Patients visit our hospital twice over a period of 2 years to complete a 5-year follow up from the initial LAST-STRONG study. At both visits, they undergo standardized neurological examination, hand-held dynamometry (age ≥ 5 years), functional measurements, muscle ultrasound, respiratory assessments (spirometry, maximal inspiratory and expiratory pressure, sniff nasal inspiratory pressure; age ≥ 5 years), Dual-energy X-ray absorptiometry (DEXA-)scan (age ≥ 2 years), X-ray of the left hand (age ≤ 17 years), lower extremity MRI (age ≥ 10 years), accelerometry for 8 days (age ≥ 2 years), and questionnaires (patient report and/or parent proxy; age ≥ 2 years). All examinations are adapted to the patient's age and functional abilities. Disease progression between all subsequent visits and relationships between outcome measures will be assessed. DISCUSSION This study will provide valuable insights into the 5-year natural history of patients with SELENON-RM and LAMA2-MD and contribute to further selecting relevant and sensitive to change clinical and functional outcome measures. Furthermore, this data will help optimize natural history data collection in clinical care and help develop clinical care guidelines. TRIAL REGISTRATION This study protocol including the patient information and consent forms has been approved by medical ethical reviewing committee ('METC Oost-Nederland'; https://www.ccmo.nl/metcs/erkende-metcs/metc-oost-nederland , file number: 2023-16401). It is registered at ClinicalTrials.gov (NCT06132750; study registration date: 2023-10-05; study first passed date: 2023-11-15).
Collapse
Affiliation(s)
- E C M de Laat
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - S L S Houwen-van Opstal
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - K Bouman
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Pediatric Neurology, Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J L M van Doorn
- Department of Neurology, Clinical Neuromuscular Imaging Group, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - D Cameron
- Department of Radiology, Clinical Neuromuscular Imaging Group, Radboud University Medical Center, Nijmegen, The Netherlands
| | - N van Alfen
- Department of Neurology, Clinical Neuromuscular Imaging Group, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A T M Dittrich
- Department of Pediatrics, Radboud University Medical Center, Radboud Institute for Health Sciences, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - E J Kamsteeg
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - H J M Smeets
- Department of Toxicogenomics, Research Institutes MHeNS and GROW, Maastricht University, Maastricht, The Netherlands
| | - J T Groothuis
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - C E Erasmus
- Department of Pediatric Neurology, Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nicol C Voermans
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
3
|
Bello L, Sansone V, Masson R, Bruno C. The Epigenetic Rescue of Dystrophin Dysfunction study of givinostat in ambulatory Duchenne muscular dystrophy patients. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2024; 43:114-115. [PMID: 39468968 PMCID: PMC11537717 DOI: 10.36185/2532-1900-637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/30/2024]
Affiliation(s)
- Luca Bello
- Department of Neurosciences DNS, University of Padua, Padua, Italy
| | - Valeria Sansone
- The NeMO Clinical Center in Milan, Neurorehabilitation Unit, University of Milan, Milan, Italy
| | - Riccardo Masson
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Claudio Bruno
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Genova, Italy, Department of Neuroscience, Rehabilitation, Ophthalmology Genetics, Maternal and Child Health, DiNOGMI, University of Genova, Genova, Italy
| |
Collapse
|
4
|
Sobierajska-Rek A, Jabłońska-Brudło J, Dąbrowska A, Wojnicz W, Meyer-Szary J, Wierzba J. Timed rolling and rising tests in Duchenne muscular dystrophy ambulant boys: a feasibility study. Minerva Pediatr (Torino) 2024; 76:208-216. [PMID: 38639735 DOI: 10.23736/s2724-5276.21.05977-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
BACKGROUND Functional activities are extensively used in motor assessments of patients with Duchenne muscular dystrophy. The role of timed items has been reported as an early prognostic factor for disease progression. However, there are two functional activities that are not widely assessed in clinical practice among Duchenne muscular dystrophy patients: rolling and bed rising. This study aimed to investigate whether the 360-degree roll (roll) and supine to sit-to-edge (bed rise) measurements are feasible tools reflecting the functional status of ambulatory DMD children by establishing possible correlations between validated measures: the Vignos Scale (VS), timed rise from floor and the 6-Minute Walk Test (6MWT). METHODS A total of 32 ambulant boys with DMD were assessed using timed items, the 6MWT and VS. RESULTS The roll and bed rise are correlated with each other. The 6MWT, the floor rise and VS are correlated with the roll and with the bed rise. CONCLUSIONS Findings offer preliminary empirical evidence addressing feasibility and safety of roll and bed rise measurements. There is a potential clinical utility of these tests in assessing functional status of DMD ambulant patients.
Collapse
Affiliation(s)
- Agnieszka Sobierajska-Rek
- Department of Rehabilitation Medicine, Faculty of Health Sciences with Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdansk, Poland -
| | - Joanna Jabłońska-Brudło
- Department of Rehabilitation Medicine, Faculty of Health Sciences with Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Aneta Dąbrowska
- Department of Rehabilitation Medicine, Faculty of Health Sciences with Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Wiktoria Wojnicz
- Faculty of Mechanical Engineering and Ship Technology, Gdansk University of Technology, Gdansk, Poland
| | - Jarosław Meyer-Szary
- Department of Pediatric Cardiology and Congenital Heart Defects, Medical University of Gdansk, Gdansk, Poland
| | - Jolanta Wierzba
- Department of Pediatric and Internal Nursing, Institute of Nursing and Midwifery, Faculty of Health Sciences with Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
5
|
Bouman K, Groothuis JT, Doorduin J, van Alfen N, Udink Ten Cate FEA, van den Heuvel FMA, Nijveldt R, Kamsteeg EJ, Dittrich ATM, Draaisma JMT, Janssen MCH, van Engelen BGM, Erasmus CE, Voermans NC. LAMA2-Related Muscular Dystrophy Across the Life Span: A Cross-sectional Study. Neurol Genet 2023; 9:e200089. [PMID: 37476021 PMCID: PMC10356133 DOI: 10.1212/nxg.0000000000200089] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 05/31/2023] [Indexed: 07/22/2023]
Abstract
Background and Objectives LAMA2-related muscular dystrophy (LAMA2-MD) is a rare neuromuscular disease characterized by proximal and axial muscle weakness, rigidity of the spine, scoliosis, and respiratory impairment. No curative treatment options exist, yet promising preclinical studies are ongoing. Currently, there is a paucity on natural history data, and appropriate clinical and functional outcome measures are needed. We aim for deep clinical phenotyping, establishment of a well-characterized baseline cohort for prospective follow-up and recruitment for future clinical trials, improvement of clinical care, and selection of outcome measures for reaching trial readiness. Methods We performed a cross-sectional, single-center, observational study. This study included neurologic examination and functional measurements among others the Motor Function Measure 20/32 (MFM-20/32) as primary outcome measure, accelerometry, questionnaires, muscle ultrasound, respiratory function tests, electrocardiography and echocardiography, and dual-energy X-ray absorptiometry. Results Twenty-seven patients with genetically confirmed LAMA2-MD were included (21 ± 13 years; M = 9; ambulant = 7). Axial and proximal muscle weakness was most pronounced. The mean MFM-20/32 score was 42.0% ± 29.4%, with domain 1 (standing and transfers) being severely affected and domain 3 (distal muscle function) relatively spared. Physical activity as measured through accelerometry showed very strong correlations to MFM-20/32 (Pearson correlation, -0.928, p < 0.01). Muscle ultrasound showed symmetrically increased echogenicity, with the sternocleidomastoid muscle most affected. Respiratory function was impaired in 85% of patients without prominent diaphragm dysfunction and was independent of age. Ten patients (37%) needed (non)invasive ventilatory support. Cardiac assessment revealed QRS fragmentation in 62%, abnormal left ventricular global longitudinal strain in 25%, and decreased left ventricular ejection fraction in 14% of patients. Decreased bone quality leading to fragility fractures was seen in most of the patients. Discussion LAMA2-MD has a widely variable phenotype. Based on the results of this cross-sectional study and current standards of care for congenital muscular dystrophies, we advise routine cardiorespiratory follow-up and optimization of bone quality. We propose MFM-20/32, accelerometry, and muscle ultrasound for assessing disease severity and progression. For definitive clinical recommendations and outcome measures, natural history data are needed. Clinical Trials Registration This study was registered at clinicaltrials.gov (NCT04478981, 21 July 2020). The first patient was enrolled in September 2020.
Collapse
Affiliation(s)
- Karlijn Bouman
- From the Department of Neurology (K.B., J.D., N.A., B.G.M.E., N.C.V.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Neurology (K.B., C.E.E.), Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital; Department of Rehabilitation (J.T.G.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Cardiology (F.E.A.U.C.), Amalia Children's Hospital; Department of Cardiology (F.M.A.H., R.N.); Department of Human Genetics (E.-J.K.); Department of Pediatrics (A.T.M.D., J.M.T.D.), Radboud Institute for Health Sciences, Amalia Children's Hospital; and Department of Internal Medicine (M.C.H.J.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan T Groothuis
- From the Department of Neurology (K.B., J.D., N.A., B.G.M.E., N.C.V.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Neurology (K.B., C.E.E.), Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital; Department of Rehabilitation (J.T.G.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Cardiology (F.E.A.U.C.), Amalia Children's Hospital; Department of Cardiology (F.M.A.H., R.N.); Department of Human Genetics (E.-J.K.); Department of Pediatrics (A.T.M.D., J.M.T.D.), Radboud Institute for Health Sciences, Amalia Children's Hospital; and Department of Internal Medicine (M.C.H.J.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jonne Doorduin
- From the Department of Neurology (K.B., J.D., N.A., B.G.M.E., N.C.V.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Neurology (K.B., C.E.E.), Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital; Department of Rehabilitation (J.T.G.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Cardiology (F.E.A.U.C.), Amalia Children's Hospital; Department of Cardiology (F.M.A.H., R.N.); Department of Human Genetics (E.-J.K.); Department of Pediatrics (A.T.M.D., J.M.T.D.), Radboud Institute for Health Sciences, Amalia Children's Hospital; and Department of Internal Medicine (M.C.H.J.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nens van Alfen
- From the Department of Neurology (K.B., J.D., N.A., B.G.M.E., N.C.V.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Neurology (K.B., C.E.E.), Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital; Department of Rehabilitation (J.T.G.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Cardiology (F.E.A.U.C.), Amalia Children's Hospital; Department of Cardiology (F.M.A.H., R.N.); Department of Human Genetics (E.-J.K.); Department of Pediatrics (A.T.M.D., J.M.T.D.), Radboud Institute for Health Sciences, Amalia Children's Hospital; and Department of Internal Medicine (M.C.H.J.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Floris E A Udink Ten Cate
- From the Department of Neurology (K.B., J.D., N.A., B.G.M.E., N.C.V.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Neurology (K.B., C.E.E.), Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital; Department of Rehabilitation (J.T.G.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Cardiology (F.E.A.U.C.), Amalia Children's Hospital; Department of Cardiology (F.M.A.H., R.N.); Department of Human Genetics (E.-J.K.); Department of Pediatrics (A.T.M.D., J.M.T.D.), Radboud Institute for Health Sciences, Amalia Children's Hospital; and Department of Internal Medicine (M.C.H.J.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frederik M A van den Heuvel
- From the Department of Neurology (K.B., J.D., N.A., B.G.M.E., N.C.V.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Neurology (K.B., C.E.E.), Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital; Department of Rehabilitation (J.T.G.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Cardiology (F.E.A.U.C.), Amalia Children's Hospital; Department of Cardiology (F.M.A.H., R.N.); Department of Human Genetics (E.-J.K.); Department of Pediatrics (A.T.M.D., J.M.T.D.), Radboud Institute for Health Sciences, Amalia Children's Hospital; and Department of Internal Medicine (M.C.H.J.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Robin Nijveldt
- From the Department of Neurology (K.B., J.D., N.A., B.G.M.E., N.C.V.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Neurology (K.B., C.E.E.), Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital; Department of Rehabilitation (J.T.G.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Cardiology (F.E.A.U.C.), Amalia Children's Hospital; Department of Cardiology (F.M.A.H., R.N.); Department of Human Genetics (E.-J.K.); Department of Pediatrics (A.T.M.D., J.M.T.D.), Radboud Institute for Health Sciences, Amalia Children's Hospital; and Department of Internal Medicine (M.C.H.J.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erik-Jan Kamsteeg
- From the Department of Neurology (K.B., J.D., N.A., B.G.M.E., N.C.V.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Neurology (K.B., C.E.E.), Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital; Department of Rehabilitation (J.T.G.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Cardiology (F.E.A.U.C.), Amalia Children's Hospital; Department of Cardiology (F.M.A.H., R.N.); Department of Human Genetics (E.-J.K.); Department of Pediatrics (A.T.M.D., J.M.T.D.), Radboud Institute for Health Sciences, Amalia Children's Hospital; and Department of Internal Medicine (M.C.H.J.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anne T M Dittrich
- From the Department of Neurology (K.B., J.D., N.A., B.G.M.E., N.C.V.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Neurology (K.B., C.E.E.), Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital; Department of Rehabilitation (J.T.G.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Cardiology (F.E.A.U.C.), Amalia Children's Hospital; Department of Cardiology (F.M.A.H., R.N.); Department of Human Genetics (E.-J.K.); Department of Pediatrics (A.T.M.D., J.M.T.D.), Radboud Institute for Health Sciences, Amalia Children's Hospital; and Department of Internal Medicine (M.C.H.J.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jos M T Draaisma
- From the Department of Neurology (K.B., J.D., N.A., B.G.M.E., N.C.V.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Neurology (K.B., C.E.E.), Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital; Department of Rehabilitation (J.T.G.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Cardiology (F.E.A.U.C.), Amalia Children's Hospital; Department of Cardiology (F.M.A.H., R.N.); Department of Human Genetics (E.-J.K.); Department of Pediatrics (A.T.M.D., J.M.T.D.), Radboud Institute for Health Sciences, Amalia Children's Hospital; and Department of Internal Medicine (M.C.H.J.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mirian C H Janssen
- From the Department of Neurology (K.B., J.D., N.A., B.G.M.E., N.C.V.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Neurology (K.B., C.E.E.), Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital; Department of Rehabilitation (J.T.G.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Cardiology (F.E.A.U.C.), Amalia Children's Hospital; Department of Cardiology (F.M.A.H., R.N.); Department of Human Genetics (E.-J.K.); Department of Pediatrics (A.T.M.D., J.M.T.D.), Radboud Institute for Health Sciences, Amalia Children's Hospital; and Department of Internal Medicine (M.C.H.J.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Baziel G M van Engelen
- From the Department of Neurology (K.B., J.D., N.A., B.G.M.E., N.C.V.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Neurology (K.B., C.E.E.), Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital; Department of Rehabilitation (J.T.G.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Cardiology (F.E.A.U.C.), Amalia Children's Hospital; Department of Cardiology (F.M.A.H., R.N.); Department of Human Genetics (E.-J.K.); Department of Pediatrics (A.T.M.D., J.M.T.D.), Radboud Institute for Health Sciences, Amalia Children's Hospital; and Department of Internal Medicine (M.C.H.J.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Corrie E Erasmus
- From the Department of Neurology (K.B., J.D., N.A., B.G.M.E., N.C.V.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Neurology (K.B., C.E.E.), Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital; Department of Rehabilitation (J.T.G.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Cardiology (F.E.A.U.C.), Amalia Children's Hospital; Department of Cardiology (F.M.A.H., R.N.); Department of Human Genetics (E.-J.K.); Department of Pediatrics (A.T.M.D., J.M.T.D.), Radboud Institute for Health Sciences, Amalia Children's Hospital; and Department of Internal Medicine (M.C.H.J.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nicol C Voermans
- From the Department of Neurology (K.B., J.D., N.A., B.G.M.E., N.C.V.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Neurology (K.B., C.E.E.), Donders Institute for Brain, Cognition and Behaviour, Amalia Children's Hospital; Department of Rehabilitation (J.T.G.), Donders Institute for Brain, Cognition and Behaviour; Department of Pediatric Cardiology (F.E.A.U.C.), Amalia Children's Hospital; Department of Cardiology (F.M.A.H., R.N.); Department of Human Genetics (E.-J.K.); Department of Pediatrics (A.T.M.D., J.M.T.D.), Radboud Institute for Health Sciences, Amalia Children's Hospital; and Department of Internal Medicine (M.C.H.J.), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
6
|
Bello L, Hoffman EP, Pegoraro E. Is it time for genetic modifiers to predict prognosis in Duchenne muscular dystrophy? Nat Rev Neurol 2023; 19:410-423. [PMID: 37308617 DOI: 10.1038/s41582-023-00823-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/14/2023]
Abstract
Patients with Duchenne muscular dystrophy (DMD) show clinically relevant phenotypic variability, despite sharing the same primary biochemical defect (dystrophin deficiency). Factors contributing to this clinical variability include allelic heterogeneity (specific DMD mutations), genetic modifiers (trans-acting genetic polymorphisms) and variations in clinical care. Recently, a series of genetic modifiers have been identified, mostly involving genes and/or proteins that regulate inflammation and fibrosis - processes increasingly recognized as being causally linked with physical disability. This article reviews genetic modifier studies in DMD to date and discusses the effect of genetic modifiers on predicting disease trajectories (prognosis), clinical trial design and interpretation (inclusion of genotype-stratified subgroup analyses) and therapeutic approaches. The genetic modifiers identified to date underscore the importance of progressive fibrosis, downstream of dystrophin deficiency, in driving the disease process. As such, genetic modifiers have shown the importance of therapies aimed at slowing this fibrotic process and might point to key drug targets.
Collapse
Affiliation(s)
- Luca Bello
- Department of Neurosciences (DNS), University of Padova, Padova, Italy
| | - Eric P Hoffman
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University (State University of New York), Binghamton, NY, USA
| | - Elena Pegoraro
- Department of Neurosciences (DNS), University of Padova, Padova, Italy.
| |
Collapse
|
7
|
Bouman K, Groothuis JT, Doorduin J, van Alfen N, Udink ten Cate FE, van den Heuvel FM, Nijveldt R, Kamsteeg EJ, Dittrich AT, Draaisma JM, Janssen MC, van Engelen BG, Erasmus CE, Voermans NC. SELENON-Related Myopathy Across the Life Span, a Cross-Sectional Study for Preparing Trial Readiness. J Neuromuscul Dis 2023; 10:1055-1074. [PMID: 37807786 PMCID: PMC10657684 DOI: 10.3233/jnd-221673] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND SELENON(SEPN1)-related myopathy (SELENON-RM) is a rare congenital neuromuscular disease characterized by proximal and axial muscle weakness, spinal rigidity, scoliosis and respiratory impairment. No curative treatment options exist, but promising preclinical studies are ongoing. Currently, natural history data are lacking, while selection of appropriate clinical and functional outcome measures is needed to reach trial readiness. OBJECTIVE We aim to identify all Dutch and Dutch-speaking Belgian SELENON-RM patients, deep clinical phenotyping, trial readiness and optimization of clinical care. METHODS This cross-sectional, single-center, observational study comprised neurological examination, functional measurements including Motor Function Measurement 20/32 (MFM-20/32) and accelerometry, questionnaires, muscle ultrasound, respiratory function tests, electro- and echocardiography, and dual-energy X-ray absorptiometry. RESULTS Eleven patients with genetically confirmed SELENON-RM were included (20±13 (3-42) years, 73% male). Axial and proximal muscle weakness were most pronounced. The mean MFM-20/32 score was 71.2±15.1%, with domain 1 (standing and transfers) being most severely affected. Accelerometry showed a strong correlation with MFM-20/32. Questionnaires revealed impaired quality of life, pain and problematic fatigue. Muscle ultrasound showed symmetrically increased echogenicity in all muscles. Respiratory function, and particularly diaphragm function, was impaired in all patients, irrespective of the age. Cardiac assessment showed normal left ventricular systolic function in all patients but abnormal left ventricular global longitudinal strain in 43% of patients and QRS fragmentation in 80%. Further, 80% of patients showed decreased bone mineral density on dual-energy X-ray absorptiometry scan and 55% of patients retrospectively experienced fragility long bone fractures. CONCLUSIONS We recommend cardiorespiratory follow-up as a part of routine clinical care in all patients. Furthermore, we advise vitamin D supplementation and optimization of calcium intake to improve bone quality. We recommend management interventions to reduce pain and fatigue. For future clinical trials, we propose MFM-20/32, accelerometry and muscle ultrasound to capture disease severity and possibly disease progression.
Collapse
Affiliation(s)
- Karlijn Bouman
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, The Netherlands
- Department of Pediatric Neurology, Donders Institute for Brain, Cognition and Behaviour, Amalia Children’s Hospital, Radboud university medical center, Nijmegen, The Netherlands
| | - Jan T. Groothuis
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, The Netherlands
| | - Jonne Doorduin
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, The Netherlands
| | - Nens van Alfen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, The Netherlands
| | - Floris E.A. Udink ten Cate
- Department of Pediatric cardiology, Amalia Children’s Hospital, Radboud university medical center, Nijmegen, The Netherlands
| | | | - Robin Nijveldt
- Department of Cardiology, Radboud university medical center, Nijmegen, The Netherlands
| | - Erik-Jan Kamsteeg
- Department of Human Genetics, Radboud university medical center, Nijmegen, The Netherlands
| | - Anne T.M. Dittrich
- Department of Pediatrics, Radboud Institute for Health Sciences, Amalia Children’s Hospital, Radboud university medical center, Nijmegen, The Netherlands
| | - Jos M.T. Draaisma
- Department of Pediatrics, Radboud Institute for Health Sciences, Amalia Children’s Hospital, Radboud university medical center, Nijmegen, The Netherlands
| | - Mirian C.H. Janssen
- Department of Internal Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Baziel G.M. van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, The Netherlands
| | - Corrie E. Erasmus
- Department of Pediatric Neurology, Donders Institute for Brain, Cognition and Behaviour, Amalia Children’s Hospital, Radboud university medical center, Nijmegen, The Netherlands
| | - Nicol C. Voermans
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
8
|
Zambon AA, Ayyar Gupta V, Ridout D, Manzur A, Baranello G, Trucco F, Muntoni F, Douglas M, McFetridge J, Parasuraman D, Alhaswani Z, McMurchie H, Rabb R, Majumdar A, Vijayakumar K, Amin S, Mason F, Frimpong‐Ansah C, Gibbon F, Parson B, Naismith K, Burslem J, Baxter A, Eadie C, Horrocks I, Di Marco M, Childs A, Pallant L, Spinty S, Shillington A, Gregson S, Cheshman L, Wraige E, Gowda V, Jungbluth H, Sheehan J, Hughes I, Warner S, Straub V, Guglieri M, Mayhew A, Chow G, Williamson S, Willis T, Kulshrestha R, Emery N, Ramdas S, Ramjattan H, de Goede C, Selley A, Ong M, White K, Illingworth M, Geary M, Palmer J, White C, Greenfield K, Hewawitharana G, Julien Y, Stephens E, Tewnion J, Ambegaonkar G, Krishnakumar D, Taylor J, Ward C, Willis T, Wright E, Rylance C. Peak functional ability and age at loss of ambulation in Duchenne muscular dystrophy. Dev Med Child Neurol 2022; 64:979-988. [PMID: 35385138 PMCID: PMC9303180 DOI: 10.1111/dmcn.15176] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 12/04/2022]
Abstract
AIM To correlate the North Star Ambulatory Assessment (NSAA) and timed rise from floor (TRF) recorded at age of expected peak with age at loss of ambulation (LOA) in Duchenne muscular dystrophy (DMD). METHOD Male children with DMD enrolled in the UK North Start Network database were included according to the following criteria: follow-up longer than 3 years, one NSAA record between 6 years and 7 years 6 months (baseline), at least one visit when older than 8 years. Data about corticosteroid treatment, LOA, genotype, NSAA, and TRF were analysed. Age at LOA among the different groups based on NSAA and TRF was determined by log-rank tests. Cox proportional hazard models were used for multivariable analysis. RESULTS A total of 293 patients from 13 different centres were included. Mean (SD) age at first and last visit was 5 years 6 months (1 year 2 months) and 12 years 8 months (2 years 11 months) (median follow-up 7 years 4 months). Higher NSAA and lower TRF at baseline were associated with older age at LOA (p<0.001). Patients scoring NSAA 32 to 34 had a probability of 0.61 of being ambulant when older than 13 years compared with 0.34 for those scoring 26 to 31. In multivariable analysis, NSAA, TRF, and corticosteroid daily regimen (vs intermittent) were all independently associated with outcome (p=0.01). INTERPRETATION Higher functional abilities at peak are associated with older age at LOA in DMD. This information is important for counselling families. These baseline measures should also be considered when designing clinical trials.
Collapse
Affiliation(s)
- Alberto A. Zambon
- Dubowitz Neuromuscular CentreUCL Great Ormond Street Institute of Child Health & Great Ormond Street HospitalLondonUK,Neuromuscular Repair UnitInstitute of Experimental Neurology (InSpe)Division of NeuroscienceIRCCS Ospedale San RaffaeleMilanItaly
| | - Vandana Ayyar Gupta
- Dubowitz Neuromuscular CentreUCL Great Ormond Street Institute of Child Health & Great Ormond Street HospitalLondonUK
| | - Deborah Ridout
- Population, Policy and Practice Research and Teaching DepartmentUCL Great Ormond Street Institute of Child HealthLondonUK,NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | - Adnan Y. Manzur
- Dubowitz Neuromuscular CentreUCL Great Ormond Street Institute of Child Health & Great Ormond Street HospitalLondonUK,NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | - Giovanni Baranello
- Dubowitz Neuromuscular CentreUCL Great Ormond Street Institute of Child Health & Great Ormond Street HospitalLondonUK
| | - Federica Trucco
- Dubowitz Neuromuscular CentreUCL Great Ormond Street Institute of Child Health & Great Ormond Street HospitalLondonUK,Children’s Sleep MedicineEvelina Children Hospital ‐ Paediatric Respiratory Department Royal Brompton HospitalGuy’s and St Thomas’ TrustLondonUK
| | - Francesco Muntoni
- Dubowitz Neuromuscular CentreUCL Great Ormond Street Institute of Child Health & Great Ormond Street HospitalLondonUK,NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Brogna C, Cristiano L, Verdolotti T, Norcia G, Ficociello L, Ruiz R, Coratti G, Fanelli L, Forcina N, Petracca G, Chieppa F, Tartaglione T, Colosimo C, Pane M, Mercuri E. Longitudinal Motor Functional Outcomes and Magnetic Resonance Imaging Patterns of Muscle Involvement in Upper Limbs in Duchenne Muscular Dystrophy. Medicina (B Aires) 2021; 57:medicina57111267. [PMID: 34833484 PMCID: PMC8624281 DOI: 10.3390/medicina57111267] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 11/25/2022] Open
Abstract
Background and Objectives: The aim of this study was to evaluate longitudinal changes using both upper limb muscle Magnetic Resonance Imaging (MRI) at shoulder, arm and forearm levels and Performance of upper limb (PUL) in ambulant and non-ambulant Duchenne Muscular Dystrophy (DMD) patients. We also wished to define whether baseline muscle MRI could help to predict functional changes after one year. Materials and Methods: Twenty-seven patients had both baseline and 12month muscle MRI and PUL assessments one year later. Results: Ten were ambulant (age range 5–16 years), and 17 non ambulant (age range 10–30 years). Increased abnormalities equal or more than 1.5 point on muscle MRI at follow up were found on all domains: at shoulder level 12/27 patients (44%), at arm level 4/27 (15%) and at forearm level 6/27 (22%). Lower follow up PUL score were found in 8/27 patients (30%) at shoulder level, in 9/27 patients (33%) at mid-level whereas no functional changes were found at distal level. There was no constant association between baseline MRI scores and follow up PUL scores at arm and forearm levels but at shoulder level patients with moderate impairment on the baseline MRI scores between 16 and 34 had the highest risk of decreased function on PUL over a year. Conclusions: Our results confirmed that the integrated use of functional scales and imaging can help to monitor functional and MRI changes over time.
Collapse
Affiliation(s)
- Claudia Brogna
- Pediatric Neurology Unit, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy;
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (G.N.); (G.C.); (L.F.); (N.F.); (M.P.)
| | - Lara Cristiano
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.C.); (R.R.); (G.P.); (F.C.)
| | - Tommaso Verdolotti
- Institute of Radiology, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (T.V.); (L.F.); (C.C.)
| | - Giulia Norcia
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (G.N.); (G.C.); (L.F.); (N.F.); (M.P.)
| | - Luana Ficociello
- Institute of Radiology, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (T.V.); (L.F.); (C.C.)
| | - Roberta Ruiz
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.C.); (R.R.); (G.P.); (F.C.)
| | - Giorgia Coratti
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (G.N.); (G.C.); (L.F.); (N.F.); (M.P.)
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.C.); (R.R.); (G.P.); (F.C.)
| | - Lavinia Fanelli
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (G.N.); (G.C.); (L.F.); (N.F.); (M.P.)
| | - Nicola Forcina
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (G.N.); (G.C.); (L.F.); (N.F.); (M.P.)
| | - Giorgia Petracca
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.C.); (R.R.); (G.P.); (F.C.)
| | - Fabrizia Chieppa
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.C.); (R.R.); (G.P.); (F.C.)
| | - Tommaso Tartaglione
- Department of Radiology, Istituto Dermatologico Italiano, IRCCS, 00167 Rome, Italy;
| | - Cesare Colosimo
- Institute of Radiology, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (T.V.); (L.F.); (C.C.)
- Institute of Radiology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marika Pane
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (G.N.); (G.C.); (L.F.); (N.F.); (M.P.)
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.C.); (R.R.); (G.P.); (F.C.)
| | - Eugenio Mercuri
- Pediatric Neurology Unit, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy;
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (G.N.); (G.C.); (L.F.); (N.F.); (M.P.)
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.C.); (R.R.); (G.P.); (F.C.)
- Correspondence: ; Tel.: +39-06-30155340; Fax: +39-06-30154363
| |
Collapse
|
10
|
Markati T, De Waele L, Schara-Schmidt U, Servais L. Lessons Learned from Discontinued Clinical Developments in Duchenne Muscular Dystrophy. Front Pharmacol 2021; 12:735912. [PMID: 34790118 PMCID: PMC8591262 DOI: 10.3389/fphar.2021.735912] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 10/12/2021] [Indexed: 02/04/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked condition caused by a deficiency of functional dystrophin protein. Patients experience progressive muscle weakness, cardiomyopathy and have a decreased life expectancy. Standards of care, including treatment with steroids, and multidisciplinary approaches have extended the life expectancy and improved the quality of life of patients. In the last 30 years, several compounds have been assessed in preclinical and clinical studies for their ability to restore functional dystrophin levels or to modify pathways involved in DMD pathophysiology. However, there is still an unmet need with regards to a disease-modifying treatment for DMD and the attrition rate between early-phase and late-phase clinical development remains high. Currently, there are 40 compounds in clinical development for DMD, including gene therapy and antisense oligonucleotides for exon skipping. Only five of them have received conditional approval in one jurisdiction subject to further proof of efficacy. In this review, we present data of another 16 compounds that failed to complete clinical development, despite positive results in early phases of development in some cases. We examine the reasons for the high attrition rate and we suggest solutions to avoid similar mistakes in the future.
Collapse
Affiliation(s)
- Theodora Markati
- MDUK Oxford Neuromuscular Center, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Liesbeth De Waele
- KU Leuven Department of Development and Regeneration, Leuven, Belgium
- Department of Paediatric Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Urlike Schara-Schmidt
- Department of Pediatric Neurology, Center for Neuromuscular Diseases, Center for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| | - Laurent Servais
- MDUK Oxford Neuromuscular Center, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Division of Child Neurology, Reference Center for Neuromuscular Disease, Centre Hospitalier Régional de Références des Maladies Neuromusculaires, Department of Pediatrics, University Hospital Liège, Liège, Belgium
| |
Collapse
|
11
|
Ambrosini A, Baldessari D, Pozzi S, Battaglia M, Beltrami E, Merico AM, Rasconi M, Monaco L. Fondazione Telethon and Unione Italiana Lotta alla Distrofia Muscolare, a successful partnership for neuromuscular healthcare research of value for patients. Orphanet J Rare Dis 2021; 16:408. [PMID: 34600567 PMCID: PMC8487484 DOI: 10.1186/s13023-021-02047-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 09/19/2021] [Indexed: 11/10/2022] Open
Abstract
In 2001, Fondazione Telethon and the Italian muscular dystrophy patient organisation Unione Italiana Lotta alla Distrofia Muscolare joined their efforts to design and launch a call for grant applications specifically dedicated to clinical projects in the field of neuromuscular disorders. This strategic initiative, run regularly over the years and still ongoing, aims at supporting research with impact on the daily life of people with a neuromuscular condition and is centred on macro-priorities identified by the patient organisation. It is investigator-driven, and all proposals are peer-reviewed for quality and feasibility. Over the years, this funding program contributed to strengthening the activities of the Italian neuromuscular clinical network, reaching many achievements in healthcare research. Moreover, it has been an enabling factor for innovative therapy experimentation at international level and prepared the clinical ground to make therapies available to Italian patients. The ultimate scope of healthcare research is to ameliorate the delivery of care. In this paper, the achievements of the funded studies are analysed also from this viewpoint, to ascertain to which extent they have fulfilled the original goals established by the patient organisation. The evidence presented indicates that this has been a highly fruitful program. Factors that contributed to its success, lessons learned, challenges, and issues that remain to be addressed are discussed to provide practical examples of an experience that could inspire also other organizations active in the field of rare disease research.
Collapse
Affiliation(s)
| | | | - Silvia Pozzi
- Fondazione Telethon, Via Poerio 14, Milan, Italy
- B.E.A. Consulting, Milan, Italy
| | | | | | | | - Marco Rasconi
- UILDM, Unione Italiana Lotta alla Distrofia Muscolare, Padua, Italy
| | - Lucia Monaco
- Fondazione Telethon, Via Poerio 14, Milan, Italy
| |
Collapse
|
12
|
Verwaaijen EJ, van Hulst A, Fiocco M, Hartman A, Grootenhuis M, Pluijm S, Pieters R, van den Akker E, van den Heuvel-Eibrink MM. Dexamethasone-induced sarcopenia and physical frailty in children with acute lymphoblastic leukemia: Protocol for a prospective cohort study (Preprint). JMIR Res Protoc 2021; 11:e33517. [PMID: 35403603 PMCID: PMC9039819 DOI: 10.2196/33517] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/24/2022] [Accepted: 02/07/2022] [Indexed: 12/24/2022] Open
Abstract
Background During treatment for pediatric acute lymphoblastic leukemia (ALL), children receive high doses of dexamethasone for its apoptotic effect on leukemia cells; however, muscle atrophy is a well-known serious side effect. Muscle atrophy (loss of muscle mass) accompanied by a decreased muscle strength may lead to a generalized impaired skeletal muscle state called sarcopenia. Loss of muscle mass is also an indicator of physical frailty, which is defined as a state of increased vulnerability that is characterized by co-occurrence of low muscle mass, muscle weakness, fatigue, slow walking speed, and low physical activity. Both sarcopenia and physical frailty are related to an increased risk of infections, hospitalizations, and decreased survival in children with chronic diseases. Objective This study aims to (1) estimate the occurrence of sarcopenia and physical frailty in children during ALL maintenance therapy, (2) evaluate the effect of administering dexamethasone, and (3) explore determinants associated with these outcomes. Methods This prospective study is being pursued within the framework of the DexaDays-2 study: a randomized controlled trial on neurobehavioral side effects in pediatric patients with ALL. A total of 105 children (3-18 years) undergoing ALL maintenance treatment at the Princess Máxima Center for Pediatric Oncology are included in this study. Sarcopenia/frailty assessments are performed before and just after a 5-day dexamethasone course. A subset of 50 children participating in the DexaDays-2 trial because of severe dexamethasone-induced neurobehavioral problems were assessed at 3 additional timepoints. The sarcopenia/frailty assessment consists of bioimpedance analysis (skeletal muscle mass [SMM]), handheld dynamometry (handgrip strength), Pediatric Quality of Life Inventory Multidimensional Fatigue Scale (fatigue), Timed Up and Go Test (TUG; walking speed), and physical activity questionnaires. To evaluate potential change in sarcopenia/frailty components after a 5-day dexamethasone administration, a paired Student t test or Mann-Whitney U test will be used. Because of the presence of repeated measurements, generalized linear mixed models will be used to estimate the effect of dexamethasone on sarcopenia and frailty outcomes. Multivariable regression models will be estimated to investigate associations between the assessment scores and patient and treatment-related factors. Results Patient accrual started in 2018 and was finalized in spring 2021. From autumn 2021 onward final data analyses will be performed. Conclusions This first study combining parameters of sarcopenia and physical frailty is of importance because these conditions can seriously complicate continuation of ALL therapy, independence in physical functioning, reaching motor milestones, and participating in daily life activities. The results will provide knowledge about these complications, the association between dexamethasone treatment and muscle loss and other components of frailty, and therefore insights into the severity of this side effect. By exploring potential determinants that may be associated with sarcopenia and physical frailty, we may be able to identify children at risk at an earlier stage and provide timely interventions. International Registered Report Identifier (IRRID) DERR1-10.2196/33517
Collapse
Affiliation(s)
| | | | - Marta Fiocco
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Annelies Hartman
- Department of Pediatric Physiotherapy, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, Netherlands
| | | | - Saskia Pluijm
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Erica van den Akker
- Department of Endocrinology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, Netherlands
| | | |
Collapse
|
13
|
North Star Ambulatory Assessment changes in ambulant Duchenne boys amenable to skip exons 44, 45, 51, and 53: A 3 year follow up. PLoS One 2021; 16:e0253882. [PMID: 34170974 PMCID: PMC8232423 DOI: 10.1371/journal.pone.0253882] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/14/2021] [Indexed: 11/19/2022] Open
Abstract
Introduction The aim of this study was to report 36-month longitudinal changes using the North Star Ambulatory Assessment (NSAA) in ambulant patients affected by Duchenne muscular dystrophy amenable to skip exons 44, 45, 51 or 53. Materials and methods We included 101 patients, 34 had deletions amenable to skip exon 44, 25 exon 45, 19 exon 51, and 28 exon 53, not recruited in any ongoing clinical trials. Five patients were counted to skip exon 51 and 53 since they had a single deletion of exon 52. Results The difference between subgroups (skip 44, 45, 51 and 53) was significant at 12 (p = 0.043), 24 (p = 0.005) and 36 months (p≤0.001). Discussion Mutations amenable to skip exons 53 and 51 had lower baseline values and more negative changes than the other subgroups while those amenable to skip exon 44 had higher scores both at baseline and at follow up. Conclusion Our results confirm different progression of disease in subgroups of patients with deletions amenable to skip different exons. This information is relevant as current long term clinical trials are using the NSAA in these subgroups of mutations.
Collapse
|
14
|
Davidson ZE, Hughes I, Ryan MM, Kornberg AJ, Cairns AG, Jones K, Hutchence M, Sampaio H, Morrison M, Truby H. Effect of a multicomponent nutritional supplement on functional outcomes for Duchenne muscular dystrophy: A randomized controlled trial. Clin Nutr 2021; 40:4702-4711. [PMID: 34237697 DOI: 10.1016/j.clnu.2021.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/15/2021] [Accepted: 06/04/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND & AIMS Duchenne muscular dystrophy (DMD) is an X-linked neuromuscular condition causing progressive muscle weakness and premature death. Whilst effective treatments such as gene therapy are developed, families often seek complementary therapies such as nutrition supplements to help their son maintain function; however, there is limited evidence supporting the use of nutritional supplements in DMD. This study aimed to compare the effect of a Standard nutritional supplement with an Enhanced nutritional supplement combining three nutriceuticals on functional outcomes in ambulatory boys with Duchenne muscular dystrophy (DMD). DESIGN A 50-week double blinded, randomized, controlled crossover trial was conducted in four Australian neuromuscular centres. Primary outcome measures were 6-min walk distance (6MWD) and community ambulation (StepWatch™ Activity Monitoring). Secondary outcome measures included body composition and quality of life. Serum 25-hydroxyvitamin D was measured. RESULTS Twenty-seven boys completed the intervention. Traditional crossover analysis demonstrated the Enhanced supplement compared to the Standard supplement was associated with a difference of +12 (95% CI: -16, 40) metres in 6MWD, +0.5 (95% CI: -53, 54) inactive minutes per day and -95 (95% CI: -887, 696) steps per day. A mixed effect model indicated a potentially clinically important effect of the Enhanced supplement on the 6MWD of +31 (95% CI: -19, 81) metres. Mean serum 25 hydroxyvitamin D levels at week 50 was 94 (95% CI: 84, 104) nmol/L. There was no observable effect of either supplement regime on body composition or quality of life. CONCLUSIONS Whilst a positive effect of the Enhanced supplement on functional outcomes was observed, this finding was inconclusive due to the small sample size. The results do not support the use of combined nutritional supplements to improve body composition or quality of life in DMD. A dose of 2000 IU vitamin D was an adequate dose to raise serum 25-hydroxyvitamin D over 50 weeks. CLINICAL TRIAL REGISTRY Registry #: ACTRN12610000462088, http://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?ACTRN=12610000462088.
Collapse
Affiliation(s)
- Zoe E Davidson
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; Murdoch Children's Research Institute, Parkville, Victoria, Australia; Neurology Department, Royal Children's Hospital, Parkville, Victoria, Australia.
| | - Ian Hughes
- Office for Research Governance and Development, Gold Coast University Hospital, Southport, Queensland, Australia
| | - Monique M Ryan
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; Murdoch Children's Research Institute, Parkville, Victoria, Australia; Neurology Department, Royal Children's Hospital, Parkville, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Parkville, Australia
| | - Andrew J Kornberg
- Murdoch Children's Research Institute, Parkville, Victoria, Australia; Neurology Department, Royal Children's Hospital, Parkville, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Parkville, Australia
| | - Anita G Cairns
- Neurosciences Department, Queensland's Children Hospital, South Brisbane, Queensland, Australia
| | - Kristi Jones
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Westmead, New South Wales, Australia; Discipline of Paediatrics, University of Sydney, Sydney, Australia
| | - Meghan Hutchence
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Hugo Sampaio
- Department of Neurology, Sydney Children's Hospital, Randwick, New South Wales, Australia; School of Women's and Children's Health, University of New South Wales, Australia
| | - Margot Morrison
- Department of Neurology, Sydney Children's Hospital, Randwick, New South Wales, Australia
| | - Helen Truby
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; School of Human Movement and Nutrition Sciences, University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
15
|
Filiz MB, Toraman NF, Kutluk MG, Filiz S, Doğan ŞK, Çakır T, Yaman A. Effects of lumbar lordosis increment on gait deteriorations in ambulant boys with Duchenne Muscular Dystrophy: A cross-sectional study. Braz J Phys Ther 2021; 25:749-755. [PMID: 34119442 DOI: 10.1016/j.bjpt.2021.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/24/2021] [Accepted: 05/06/2021] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Increment of lumbar lordosis, a frequent spinal finding in Duchenne Muscular Dystrophy (DMD), is a compensatory mechanism secondary to muscle weakness. However, excessive lumbar lordosis may change the position of the center of mass, and lead to balance and walking difficulties. OBJECTIVE To study the relationship between factors that may influence ambulatory function in boys with DMD and to investigate the effects of lumbar lordosis increment on gait and balance perturbations. METHODS Twenty-one ambulant patients with DMD and 10 healthy boys were included. Lumbar lordosis and thoracic kyphosis angles, dynamic and static balance tests, ambulatory function, muscle strength, and disease severity were assessed. Usage of steroids and orthotic devices were recorded. Scoliosis was assessed on radiographs. Receiver operator characteristic curves were formed and area under curve (AUC) measurements were performed to assess the ability of the tests to discriminate ambulatory status and optimal cut-off values were established according to the Youden index. RESULTS The amount of lumbar lordosis correlated strongly and negatively with quality of ambulation (r = -0.710) and moderately with performance on balance tests. The strength of both upper limbs and lower limbs muscles were not associated with any of the variables. According to the AUC analysis, patients with a lumbar lordosis higher than 36° had worse scores on gait and dynamic balance tests. CONCLUSION Ambulation and dynamic balance are negatively affected by the increment of lumbar lordosis with a cut-off point of 36°in boys with DMD.
Collapse
Affiliation(s)
- Meral Bilgilisoy Filiz
- Antalya Training and Research Hospital, Department of Physical Medicine and Rehabilitation, Health Sciences University, Antalya, Turkey; Antalya Training and Research Hospital, Neuromuscular Disease Center, Health Sciences University, Antalya, Turkey.
| | - Naciye Füsun Toraman
- Antalya Training and Research Hospital, Department of Physical Medicine and Rehabilitation, Health Sciences University, Antalya, Turkey; Antalya Training and Research Hospital, Neuromuscular Disease Center, Health Sciences University, Antalya, Turkey
| | - Muhammet Gültekin Kutluk
- Antalya Training and Research Hospital, Department of Pediatric Neurology, Health Sciences University, Antalya, Turkey; Antalya Training and Research Hospital, Neuromuscular Disease Center, Health Sciences University, Antalya, Turkey
| | - Serkan Filiz
- Antalya Training and Research Hospital, Department of Pediatric Allergy and Immunology, Health Sciences University, Antalya, Turkey; Antalya Training and Research Hospital, Neuromuscular Disease Center, Health Sciences University, Antalya, Turkey
| | - Şebnem Koldaş Doğan
- Antalya Training and Research Hospital, Department of Physical Medicine and Rehabilitation, Health Sciences University, Antalya, Turkey
| | - Tuncay Çakır
- Department of Physical Medicine and Rehabilitation, Private ASV Life Hospital, Antalya, Turkey
| | - Aylin Yaman
- Antalya Training and Research Hospital, Department of Neurology, Health Sciences University, Antalya, Turkey; Antalya Training and Research Hospital, Neuromuscular Disease Center, Health Sciences University, Antalya, Turkey
| |
Collapse
|
16
|
Cheng D, Ayyagari R, Signorovitch J. The statistical performance of matching-adjusted indirect comparisons: Estimating treatment effects with aggregate external control data. Ann Appl Stat 2020. [DOI: 10.1214/20-aoas1359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
17
|
Can simple and low-cost motor function assessments help in the diagnostic suspicion of Duchenne muscular dystrophy? JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2020. [DOI: 10.1016/j.jpedp.2019.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
18
|
Pereira AC, Araújo APDQC, Ribeiro MG. Can simple and low-cost motor function assessments help in the diagnostic suspicion of Duchenne muscular dystrophy? J Pediatr (Rio J) 2020; 96:503-510. [PMID: 31009620 PMCID: PMC9432257 DOI: 10.1016/j.jped.2019.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 01/25/2019] [Accepted: 02/18/2019] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE Duchenne muscular dystrophy, an X-linked genetic disease, leads to progressive muscle weakness mainly in the lower limbs. Motor function tests help to monitor disease progression. Can low-cost, simple assessments help in the diagnostic suspicion of Duchenne muscular dystrophy? The authors aim to define the sensitivity of time to rise from the floor, time to walk 10meters, and time to run 10meters, evaluating them as eventual diagnostic screening tools. METHODS This is an analytical, observational, retrospective (1998-2015), and prospective study (2015-2018). Cases were recruited from the database of the pediatric neurology department and the healthy, from child care consultations, with normal gait development (up to 15 months) and without other comorbidities (neuromuscular, pulmonary, heart diseases) from the same university hospital. RESULTS 128 Duchenne muscular dystrophy patients and 344 healthy children were analyzed, equally distributed in age groups. In Duchenne muscular dystrophy, there is a progressive increase in the means of the times to perform the motor tests according to the age group, which accelerates very abruptly after 7 years of age. Healthy children acquire maximum motor capacity at 6 years and stabilize their times. The time to rise showed a p-value <0.05 and a strong association (effect size [ES] >0.8) in all age groups (except at 12 years), with time to walk 10 meters from 9 years, and with time to run 10 meters , from 5 years. The 100% sensitivity points were defined as follows: time to rise, at 2s; time to walk 10 meters, 5s; time to run 10 meters, 4s. CONCLUSIONS Time to rise is a useful and simple tool in the screening of neuromuscular disorders such as Duchenne muscular dystrophy, a previously incurable disease with new perspectives for treatment.
Collapse
Affiliation(s)
- Aline Chacon Pereira
- Universidade Federal do Rio de Janeiro (UFRJ), Departamento de Pediatria, Neuropediatria, Rio de Janeiro, RJ, Brazil.
| | | | - Márcia Gonçalves Ribeiro
- Universidade Federal do Rio de Janeiro (UFRJ), Departamento de Pediatria, Genética Clínica, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
19
|
Goemans N, Wong B, Van den Hauwe M, Signorovitch J, Sajeev G, Cox D, Landry J, Jenkins M, Dieye I, Yao Z, Hossain I, Ward SJ. Prognostic factors for changes in the timed 4-stair climb in patients with Duchenne muscular dystrophy, and implications for measuring drug efficacy: A multi-institutional collaboration. PLoS One 2020; 15:e0232870. [PMID: 32555695 PMCID: PMC7302444 DOI: 10.1371/journal.pone.0232870] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 04/22/2020] [Indexed: 12/25/2022] Open
Abstract
The timed 4-stair climb (4SC) assessment has been used to measure function in Duchenne muscular dystrophy (DMD) practice and research. We sought to identify prognostic factors for changes in 4SC, assess their consistency across data sources, and the extent to which prognostic scores could be useful in DMD clinical trial design and analysis. Data from patients with DMD in the placebo arm of a phase 3 trial (Tadalafil DMD trial) and two real-world sources (Universitaire Ziekenhuizen, Leuven, Belgium [Leuven] and Cincinnati Children's Hospital Medical Center [CCHMC]) were analyzed. One-year changes in 4SC completion time and velocity (stairs/second) were analyzed. Prognostic models included age, height, weight, steroid use, and multiple timed function tests and were developed using multivariable regression, separately in each data source. Simulations were used to quantify impacts on trial sample size requirements. Data on 1-year changes in 4SC were available from the Tadalafil DMD trial (n = 92) Leuven (n = 67), and CCHMC (n = 212). Models incorporating multiple timed function tests, height, and weight significantly improved prognostic accuracy for 1-year change in 4SC (R2: 29%-36% for 4SC velocity, and 29%-34% for 4SC time) compared to models including only age, baseline 4SC and steroid duration (R2:8%-17% for 4SC velocity and 2%-13% for 4SC time). Measures of walking and rising ability contributed important prognostic information for changes in 4SC. In a randomized trial with equal allocation to treatment and placebo, adjustment for such a prognostic score would enable detection (at 80% power) of a treatment effect of 0.25 stairs/second with 100–120 patients, compared to 170–190 patients without prognostic score adjustment. Combining measures of ambulatory function doubled prognostic accuracy for 1-year changes in 4SC completion time and velocity. Randomized clinical trials incorporating a validated prognostic score could reduce sample size requirements by approximately 40%. Knowledge of important prognostic factors can also inform adjusted comparisons to external controls.
Collapse
Affiliation(s)
- Nathalie Goemans
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
- * E-mail:
| | - Brenda Wong
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, United States of America
| | | | - James Signorovitch
- Analysis Group Inc., Boston, Massachusetts, United States of America
- The Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
| | - Gautam Sajeev
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - David Cox
- Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - John Landry
- Eli Lilly and Company, Toronto, Ontario, Canada
| | | | - Ibrahima Dieye
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Zhiwen Yao
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Intekhab Hossain
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Susan J. Ward
- The Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
| | | |
Collapse
|
20
|
Hafner P, Bonati U, Klein A, Rubino D, Gocheva V, Schmidt S, Schroeder J, Bernert G, Laugel V, Steinlin M, Capone A, Gloor M, Bieri O, Hemkens LG, Speich B, Zumbrunn T, Gueven N, Fischer D. Effect of Combination l-Citrulline and Metformin Treatment on Motor Function in Patients With Duchenne Muscular Dystrophy: A Randomized Clinical Trial. JAMA Netw Open 2019; 2:e1914171. [PMID: 31664444 PMCID: PMC6824222 DOI: 10.1001/jamanetworkopen.2019.14171] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
IMPORTANCE Nitric oxide precursors, such as the amino acid l-arginine and the biguanide antidiabetic drug metformin, have been associated with metabolism and muscle function in patients with Duchenne muscular dystrophy (DMD). The treatment of DMD remains an unmet medical need. OBJECTIVE To evaluate the benefits and harms of a combination of l-citrulline and metformin treatment among patients with DMD. DESIGN, SETTING, AND PARTICIPANTS A single-center randomized double-blind placebo-controlled parallel-group clinical trial was conducted between December 12, 2013, and March 30, 2016, at the University Children's Hospital Basel in Switzerland. A total of 47 ambulant male patients aged 6.5 to 10 years with genetically confirmed DMD were recruited locally and from the patient registries of Switzerland, Germany, Austria, and France. Data were analyzed from April 6, 2016, to September 5, 2019. INTERVENTIONS Patients in the treatment group received 2500 mg of l-citrulline and 250 mg of metformin (combination therapy) 3 times a day for 26 weeks compared with patients in the control group, who received placebo. MAIN OUTCOMES AND MEASURES The primary end point was the change in transfer and standing posture, as assessed by the first dimension of the Motor Function Measure, version 32, from baseline to week 26. Secondary end points included assessments of timed function, quantitative muscle force, biomarkers for muscle necrosis, and adverse events. The 2 prespecified subgroups comprised patients who were able to walk 350 m or more in 6 minutes (stable subgroup) and patients who were not able to walk 350 m in 6 minutes (unstable subgroup) at baseline. RESULTS Among 49 ambulant male children with DMD who were screened for eligibility, 47 patients with a mean (SD) age of 8.2 (1.1) years were randomized to a treatment group receiving combination therapy (n = 23) or a control group receiving placebo (n = 24), and 45 patients completed the study. No significant differences between groups were found in the results of timed function and muscle force tests for overall, proximal and axial, and distal motor function. Among patients receiving combination therapy, the Motor Function Measure first dimension subscore decrease was 5.5% greater than that of patients receiving placebo (95% CI, -1.0% to 12.1%; P = .09). The administration of combination therapy had significantly favorable effects on the first dimension subscore decrease among the 29 patients in the stable subgroup (6.7%; 95% CI, 0.9%-12.6%; P = .03) but not among the 15 patients in the unstable subgroup (3.9%; 95% CI, -13.2% to 20.9%; P = .63). Overall, the treatment was well tolerated with only mild adverse effects. CONCLUSIONS AND RELEVANCE Treatment with combination therapy was not associated with an overall reduction in motor function decline among ambulant patients with DMD; however, a reduction in motor function decline was observed among the stable subgroup of patients treated with combination therapy. The statistically nonsignificant difference of distal motor function in favor of combination therapy and the reduced degeneration of muscle tissue appear to support the treatment concept, but the study may have lacked sufficient statistical power. Further research exploring this treatment option with a greater number of patients is warranted. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT01995032.
Collapse
Affiliation(s)
- Patricia Hafner
- Division of Pediatric Neurology, University Children's Hospital Basel, Basel, Switzerland
- Division of Neurology, Medical University Clinic, Kantonsspital Baselland, Bruderholz, Switzerland
| | - Ulrike Bonati
- Division of Pediatric Neurology, University Children's Hospital Basel, Basel, Switzerland
| | - Andrea Klein
- Division of Pediatric Neurology, University Children's Hospital Basel, Basel, Switzerland
- Division of Pediatric Neurology, University of Berne Hospital, Berne, Switzerland
- Division of Pediatric Neurology, Lausanne University Hospital, Lausanne, Switzerland
| | - Daniela Rubino
- Division of Pediatric Neurology, University Children's Hospital Basel, Basel, Switzerland
| | - Vanya Gocheva
- Division of Pediatric Neurology, University Children's Hospital Basel, Basel, Switzerland
| | - Simone Schmidt
- Division of Pediatric Neurology, University Children's Hospital Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Basel, Switzerland
| | - Jonas Schroeder
- Division of Pediatric Neurology, University Children's Hospital Basel, Basel, Switzerland
| | - Günther Bernert
- Department of Pediatrics, Kaiser Franz Josef Hospital, Vienna, Austria
| | - Vincent Laugel
- Department of Pediatric Neurology, Strasbourg University Hospital, Strasbourg, France
| | - Maja Steinlin
- Division of Pediatric Neurology, University of Berne Hospital, Berne, Switzerland
| | - Andrea Capone
- Division of Pediatric Neurology, Children's Hospital, Aarau, Switzerland
| | - Monika Gloor
- Division of Radiological Physics, Department of Radiology, University Hospital Basel, Basel, Switzerland
| | - Oliver Bieri
- Division of Radiological Physics, Department of Radiology, University Hospital Basel, Basel, Switzerland
| | - Lars G. Hemkens
- Basel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Benjamin Speich
- Basel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Thomas Zumbrunn
- Clinical Trial Unit, Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Nuri Gueven
- Pharmacy, School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Dirk Fischer
- Division of Pediatric Neurology, University Children's Hospital Basel, Basel, Switzerland
- Division of Neurology, Medical University Clinic, Kantonsspital Baselland, Bruderholz, Switzerland
- Department of Neurology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
21
|
Varsanik JS, Kimmel ZM, Laforet GA, Ricotti V, Sajeev G, Signorovitch J, Quiroz JA, Chevalier TW. Validation of an ambient measurement system (AMS) for physical activities in a paediatric population. J Med Eng Technol 2019; 43:182-189. [PMID: 31305192 DOI: 10.1080/03091902.2019.1640308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Ambient measurement systems (AMSs) can enable continuous assessment of functional performance at home, increasing the availability of data for monitoring of neuromuscular disease. An AMS passively measures movement whenever someone is in range of the sensor, without the need for any wearable sensors. The current study evaluates the performance of an AMS for three metrics associated with functional assessments in Duchenne muscular dystrophy (DMD): ambulation speed, rise-to-stand speed and arm-raise speed. Healthy paediatric subjects performed a series of functional tasks and were graded by both a human rater and an AMS. Linear mixed-effect models were fit to calculate agreement between the two measurement methods. For all activities, the AMS and human rater supplied similar measurements of average speed, with correlation coefficients of 0.76-0.92 and systematic differences ranging in magnitude from 0 to 0.48 m per second. The largest systematic difference was for the 10-m run, which was likely due to human rater reaction time. Systematic differences in arm-raise measurements were due to incomplete execution of movements by test participants. These results are consistent with previous studies comparing automated and manual measurements of movement. This study demonstrates that an AMS device is able to measure ambulation speed, rise-to-stand speed and arm-raise speed in a paediatric population in a controlled setting without the need for complicated installation, calibration or worn sensors.
Collapse
|
22
|
Brogna C, Coratti G, Pane M, Ricotti V, Messina S, D’Amico A, Bruno C, Vita G, Berardinelli A, Mazzone E, Magri F, Ricci F, Mongini T, Battini R, Bello L, Pegoraro E, Baranello G, Previtali SC, Politano L, Comi GP, Sansone VA, Donati A, Bertini E, Muntoni F, Goemans N, Mercuri E. Long-term natural history data in Duchenne muscular dystrophy ambulant patients with mutations amenable to skip exons 44, 45, 51 and 53. PLoS One 2019; 14:e0218683. [PMID: 31237898 PMCID: PMC6592545 DOI: 10.1371/journal.pone.0218683] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/06/2019] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION The aim of this international collaborative effort was to report 36-month longitudinal changes using the 6MWT in ambulant patients affected by Duchenne muscular dystrophy amenable to skip exons 44, 45, 51 or 53. MATERIALS AND METHODS Of the 92 patients included in the study, 24 had deletions amenable to skip exon 44, 27 exon 45, 18 exon 51, and 28 exon 53. Five patients with a single deletion of exon 52 were counted in both subgroups skipping exon 51 and 53. RESULTS The difference between subgroups amenable to skip different exons was not significant at 12 months but became significant at both 24 (p≤0.05) and 36 months (p≤0.01). DISCUSSION Mutations amenable to skip exon 53 had lower baseline values and more negative changes than the other subgroups while those amenable to skip exon 44 had better results both at baseline and at follow up. Deletions amenable to skip exon 45 were associated with a more variable pattern of progression. Single exon deletions were more often associated with less drastic changes but this was not always true in individual cases. CONCLUSION Our results confirm that the progression of disease can differ between patients with different deletions, although the changes only become significant from 24 months onwards. This information is relevant because there are current clinical trials specifically targeting patients with these subgroups of mutations.
Collapse
Affiliation(s)
- Claudia Brogna
- Pediatric Neurology, Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giorgia Coratti
- Pediatric Neurology, Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Marika Pane
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Valeria Ricotti
- Dubowitz Neuromuscular Centre, UCL & Great Ormond Street Hospital, London, United Kingdom
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom
| | - Sonia Messina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
- Nemo SUD Clinical Centre, University Hospital “G. Martino”, Messina, Italy
| | - Adele D’Amico
- Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Claudio Bruno
- Center of Myology and Neurodegenerative Disorders, Istituto Giannina Gaslini, Genoa, Italy
| | - Gianluca Vita
- Nemo SUD Clinical Centre, University Hospital “G. Martino”, Messina, Italy
| | - Angela Berardinelli
- Child Neurology and Psychiatry Unit, ‘‘Casimiro Mondino” Foundation, Pavia, Italy
| | - Elena Mazzone
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesca Magri
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Dino Ferrari Centre, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Federica Ricci
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Torino, Torino, Italy
| | - Tiziana Mongini
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Torino, Torino, Italy
| | - Roberta Battini
- Department of Developmental Neuroscience, Stella Maris Institute, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luca Bello
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padua, Padua, Italy
| | | | | | - Luisa Politano
- Dipartimento di Medicina Sperimentale, Seconda Università di Napoli, Napoli, Italy
| | - Giacomo P. Comi
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Dino Ferrari Centre, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Valeria A. Sansone
- The NEMO Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, Milan, Italy
| | - Alice Donati
- Metabolic Unit, A. Meyer Children's Hospital, Florence, Italy
| | - Enrico Bertini
- Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL & Great Ormond Street Hospital, London, United Kingdom
| | - Nathalie Goemans
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Eugenio Mercuri
- Pediatric Neurology, Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | |
Collapse
|
23
|
Landfeldt E, Sejersen T, Tulinius M. A mini-review and implementation model for using ataluren to treat nonsense mutation Duchenne muscular dystrophy. Acta Paediatr 2019; 108:224-230. [PMID: 30188594 DOI: 10.1111/apa.14568] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/18/2018] [Accepted: 09/04/2018] [Indexed: 02/05/2023]
Abstract
AIM Ataluren has been approved for treating nonsense mutation Duchenne muscular dystrophy (nmDMD), and there are currently discussions concerning drug access and applications beyond the development programme. This study provides an overview of nmDMD and ataluren, stipulates clinical rules for treatment initiation and discontinuation and proposes a model for the implementation of orphan drugs in clinical practice in Sweden. METHODS This was a targeted mini-review of the literature from 1995 to 2018, which included cohort studies, guidelines, randomised clinical trials, clinical commentaries and reviews. The review covered the pathophysiology, epidemiology and burden of nmDMD and the clinical programme for ataluren. RESULTS Based on the current evidence, and our experiences, we recommend that patients with nmDMD should be given ataluren as soon as possible after diagnosis and this treatment should continue until they reach a forced vital capacity of <30%, and, or, a score of at least six on the Brooke upper extremity scale. We propose an implementation model that comprises a coordinating specialist physician and a national expert committee responsible for providing clinical intelligence to ensure appropriate use. CONCLUSION Our clinical recommendations and proposed implementation model will inform the optimum medical management of nmDMD in Sweden and help ensure timely, equal access to ataluren and similar orphan drugs.
Collapse
Affiliation(s)
- Erik Landfeldt
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
- ICON plc; Stockholm Sweden
| | - Thomas Sejersen
- The Department of Women's and Children's Health, Paediatric Neurology; Karolinska University Hospital; Astrid Lindgren Children's Hospital; Karolinska Institutet; Stockholm Sweden
| | - Már Tulinius
- Department of Pediatrics; Queen Silvia Children's Hospital; University of Gothenburg; Gothenburg Sweden
| |
Collapse
|
24
|
Mohassel P, Landon-Cardinal O, Foley AR, Donkervoort S, Pak KS, Wahl C, Shebert RT, Harper A, Fequiere P, Meriggioli M, Toro C, Drachman D, Allenbach Y, Benveniste O, Béhin A, Eymard B, Lafôret P, Stojkovic T, Mammen AL, Bönnemann CG. Anti-HMGCR myopathy may resemble limb-girdle muscular dystrophy. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2018; 6:e523. [PMID: 30588482 PMCID: PMC6292490 DOI: 10.1212/nxi.0000000000000523] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/16/2018] [Indexed: 12/12/2022]
Abstract
Objective To determine the prevalence and clinical features of anti-HMGCR myopathy among patients with presumed limb-girdle muscular dystrophy (LGMD) in whom genetic testing has failed to elucidate causative mutations. Methods Patients with presumed LGMD and unrevealing genetic testing were selected based on a few clinico-pathologic features and tested for anti-HMGCR autoantibodies (n = 11). These clinico-pathologic features are peak creatine kinase (CK) greater than 1,000 IU/L and at least 3 of the following features: (1) limb-girdle pattern of weakness, (2) selective involvement of posterior thigh on clinical examination or muscle imaging, (3) dystrophic changes on muscle biopsy, and (4) no family history of muscular dystrophy. Results Six patients tested positive for anti-HMGCR autoantibodies. In 4, there was a presymptomatic phase, lasting as long as 10 years, characterized by elevated CK levels without weakness. Muscle biopsies revealed variable degrees of a dystrophic pathology without prominent inflammation. In an independent cohort of patients with anti-HMGCR myopathy, 17 of 51 (∼33%) patients were initially presumed to have a form of LGMD based on clinico-pathologic features but were ultimately found to have anti-HMGCR myopathy. Most of these patients responded favorably to immunomodulatory therapies, evidenced by reduction of CK levels and improved strength. Conclusions Anti-HMGCR myopathy can resemble LGMD. Diagnosis of patients with a LGMD-like presentation of anti-HMGCR myopathy is critical because these patients may respond favorably to immunotherapy, especially those with shorter disease duration.
Collapse
Affiliation(s)
- Payam Mohassel
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Océane Landon-Cardinal
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - A Reghan Foley
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Sandra Donkervoort
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Katherine S Pak
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Colleen Wahl
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Robert T Shebert
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Amy Harper
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Pierre Fequiere
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Matthew Meriggioli
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Camilo Toro
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Daniel Drachman
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Yves Allenbach
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Olivier Benveniste
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Anthony Béhin
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Bruno Eymard
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Pascal Lafôret
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Tanya Stojkovic
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Andrew L Mammen
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| | - Carsten G Bönnemann
- National Institutes of Health (P.M., A.R.F., S.D., C.G.B.), NINDS, NNDCS, Bethesda, MD; Department of Internal Medicine and Clinical Immunology (O.L.-C., Y.A., O.B.), Sorbonne Universités, University Pierre et Marie et Curie, APHP, Hôpital Pitié-Salpêtrière, Paris, France; National Institutes of Health (K.P., A.L.M.), NIAMS; National Institutes of Health (C.W., C.T.), NHGRI, UDP, Bethesda, MD; Department of Neurology (R.T.S.), University of Miami, Miami, FL; Department of Neurology (A.H.), Virginia Commonwealth University, Richmond, VA; Division of Pediatric Neurology (P.F.), Department of Pediatrics, University of Alabama, Birmingham; Department of Neurological Sciences (M.M.), Rush University Medical Center, Chicago, IL; Department of Neurology (D.D., A.L.M.), Department of Medicine (A.L.M.), Johns Hopkins University, Baltimore, MD; AP-HP (A.B., B.E., T.S.), G-H Pitié-Salpêtrière, Institut de Myologie, Paris; and Neurology Department (P.L.), Raymond Poincaré Hospital, Garches, APHP and INSERM U1179, END-ICAP, Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux, France
| |
Collapse
|
25
|
Ambrosini A, Calabrese D, Avato FM, Catania F, Cavaletti G, Pera MC, Toscano A, Vita G, Monaco L, Pareyson D. The Italian neuromuscular registry: a coordinated platform where patient organizations and clinicians collaborate for data collection and multiple usage. Orphanet J Rare Dis 2018; 13:176. [PMID: 30286784 PMCID: PMC6172847 DOI: 10.1186/s13023-018-0918-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/21/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The worldwide landscape of patient registries in the neuromuscular disease (NMD) field has significantly changed in the last 10 years, with the international TREAT-NMD network acting as strong driver. At the same time, the European Medicines Agency and the large federations of rare disease patient organizations (POs), such as EURORDIS, contributed to a great cultural change, by promoting a paradigm shift from product-registries to patient-centred registries. In Italy, several NMD POs and Fondazione Telethon undertook the development of a TREAT-NMD linked patient registry in 2009, with the referring clinical network providing input and support to this initiative through the years. This article describes the outcome of this joint effort and shares the experience gained. METHODS The Italian NMD registry is based on an informatics technology platform, structured according to the most rigorous legal national and European requirements for management of patient sensitive data. A user-friendly web interface allows both direct patients and clinicians' participation. The platform's design permits expansion to incorporate new modules and new registries, and is suitable of interoperability with other international efforts. RESULTS When the Italian NMD Registry was initiated, an ad hoc legal entity (NMD Registry Association) was devised to manage registries' data. Currently, several disease-specific databases are hosted on the platform. They collect molecular and clinical details of individuals affected by Duchenne or Becker muscular dystrophy, Charcot-Marie-Tooth disease, transthyretin type-familial amyloidotic polyneuropathy, muscle glycogen storage disorders, spinal and bulbar muscular atrophy, and spinal muscular atrophy. These disease-specific registries are at different stage of development, and the NMD Registry itself has gone through several implementation steps to fulfil different technical and governance needs. The new governance model is based on the agreement between the NMD Registry Association and the professional societies representing the Italian NMD clinical network. Overall, up to now the NMD registry has collected data on more than 2000 individuals living with a NMD condition. CONCLUSIONS The Italian NMD Registry is a flexible platform that manages several condition-specific databases and is suitable to upgrade. All stakeholders participate in its management, with clear roles and responsibilities. This governance model has been key to its success. In fact, it favored patient empowerment and their direct participation in research, while also engaging the expert clinicians of the Italian network in the collection of accurate clinical data according to the best clinical practices.
Collapse
Affiliation(s)
| | - Daniela Calabrese
- UOC Malattie neurodegenerative e neurometaboliche rare, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | | | - Guido Cavaletti
- School of Medicine and Surgery and Experimental Neurology Unit, University of Milano-Bicocca, Monza, Italy
| | - Maria Carmela Pera
- Paediatric Neurology and Centro Clinico Nemo, Catholic University and Policlinico Gemelli, Rome, Italy
| | - Antonio Toscano
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Giuseppe Vita
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Lucia Monaco
- Fondazione Telethon, Via Poerio 14, 20129 Milan, Italy
| | - Davide Pareyson
- UOC Malattie neurodegenerative e neurometaboliche rare, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
26
|
Araujo APQC, Nardes F, Fortes CPDD, Pereira JA, Rebel MF, Dias CM, Barbosa RDCGA, Lopes MVR, Langer AL, Neves FR, Reis EF. Brazilian consensus on Duchenne muscular dystrophy. Part 2: rehabilitation and systemic care. ARQUIVOS DE NEURO-PSIQUIATRIA 2018; 76:481-489. [DOI: 10.1590/0004-282x20180062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 04/30/2018] [Indexed: 12/19/2022]
Abstract
ABSTRACT Significant advances in the understanding and management of Duchenne muscular dystrophy (DMD) have occurred since the publication of international guidelines for DMD care in 2010. Our objective was to provide an evidence-based national consensus statement for multidisciplinary care of DMD in Brazil. A combination of the Delphi technique with a systematic review of studies from 2010 to 2016 was employed to classify evidence levels and grade of recommendations for the guideline. Our recommendations were divided in two parts. Guideline methodology and overall disease concept descriptions are found in Part 1. Here we present Part 2, where we provide the results and recommendations on rehabilitation and systemic care for DMD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Edna Fátima Reis
- Associação Brasileira de Distrofia Muscular Viver Bem sem Limite, Brasil; Associação de Assistência à Criança Deficiente, Brasil
| |
Collapse
|
27
|
McDonald CM, Henricson EK, Abresch RT, Duong T, Joyce NC, Hu F, Clemens PR, Hoffman EP, Cnaan A, Gordish-Dressman H. Long-term effects of glucocorticoids on function, quality of life, and survival in patients with Duchenne muscular dystrophy: a prospective cohort study. Lancet 2018; 391:451-461. [PMID: 29174484 DOI: 10.1016/s0140-6736(17)32160-8] [Citation(s) in RCA: 317] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/30/2017] [Accepted: 07/25/2017] [Indexed: 01/14/2023]
Abstract
BACKGROUND Glucocorticoid treatment is recommended as a standard of care in Duchenne muscular dystrophy; however, few studies have assessed the long-term benefits of this treatment. We examined the long-term effects of glucocorticoids on milestone-related disease progression across the lifespan and survival in patients with Duchenne muscular dystrophy. METHODS For this prospective cohort study, we enrolled male patients aged 2-28 years with Duchenne muscular dystrophy at 20 centres in nine countries. Patients were followed up for 10 years. We compared no glucocorticoid treatment or cumulative treatment duration of less than 1 month versus treatment of 1 year or longer with regard to progression of nine disease-related and clinically meaningful mobility and upper limb milestones. We used Kaplan-Meier analyses to compare glucocorticoid treatment groups for time to stand from supine of 5 s or longer and 10 s or longer, and loss of stand from supine, four-stair climb, ambulation, full overhead reach, hand-to-mouth function, and hand function. Risk of death was also assessed. This study is registered with ClinicalTrials.gov, number NCT00468832. FINDINGS 440 patients were enrolled during two recruitment periods (2006-09 and 2012-16). Time to all disease progression milestone events was significantly longer in patients treated with glucocorticoids for 1 year or longer than in patients treated for less than 1 month or never treated (log-rank p<0·0001). Glucocorticoid treatment for 1 year or longer was associated with increased median age at loss of mobility milestones by 2·1-4·4 years and upper limb milestones by 2·8-8·0 years compared with treatment for less than 1 month. Deflazacort was associated with increased median age at loss of three milestones by 2·1-2·7 years in comparison with prednisone or prednisolone (log-rank p<0·012). 45 patients died during the 10-year follow-up. 39 (87%) of these deaths were attributable to Duchenne-related causes in patients with known duration of glucocorticoids usage. 28 (9%) deaths occurred in 311 patients treated with glucocorticoids for 1 year or longer compared with 11 (19%) deaths in 58 patients with no history of glucocorticoid use (odds ratio 0·47, 95% CI 0·22-1·00; p=0·0501). INTERPRETATION In patients with Duchenne muscular dystrophy, glucocorticoid treatment is associated with reduced risk of losing clinically meaningful mobility and upper limb disease progression milestones across the lifespan as well as reduced risk of death. FUNDING US Department of Education/National Institute on Disability and Rehabilitation Research; US Department of Defense; National Institutes of Health/National Institute of Arthritis and Musculoskeletal and Skin Diseases; and Parent Project Muscular Dystrophy.
Collapse
Affiliation(s)
- Craig M McDonald
- University of California Davis School of Medicine, Sacramento, CA, USA.
| | - Erik K Henricson
- University of California Davis School of Medicine, Sacramento, CA, USA
| | - Richard T Abresch
- University of California Davis School of Medicine, Sacramento, CA, USA
| | | | - Nanette C Joyce
- University of California Davis School of Medicine, Sacramento, CA, USA
| | - Fengming Hu
- Center for Genetic Medicine, Children's National Health System and the George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | | | - Eric P Hoffman
- Binghamton University's School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Avital Cnaan
- Center for Genetic Medicine, Children's National Health System and the George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Heather Gordish-Dressman
- Center for Genetic Medicine, Children's National Health System and the George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
28
|
Identification of plasma interleukins as biomarkers for deflazacort and omega-3 based Duchenne muscular dystrophy therapy. Cytokine 2018; 102:55-61. [DOI: 10.1016/j.cyto.2017.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 01/23/2023]
|
29
|
Goemans N, Mercuri E, Belousova E, Komaki H, Dubrovsky A, McDonald CM, Kraus JE, Lourbakos A, Lin Z, Campion G, Wang SX, Campbell C. A randomized placebo-controlled phase 3 trial of an antisense oligonucleotide, drisapersen, in Duchenne muscular dystrophy. Neuromuscul Disord 2017; 28:4-15. [PMID: 29203355 DOI: 10.1016/j.nmd.2017.10.004] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 09/23/2017] [Accepted: 10/17/2017] [Indexed: 01/16/2023]
Abstract
This 48-week, randomized, placebo-controlled phase 3 study (DMD114044; NCT01254019) evaluated efficacy and safety of subcutaneous drisapersen 6 mg/kg/week in 186 ambulant boys aged ≥5 years, with Duchenne muscular dystrophy (DMD) resulting from an exon 51 skipping amenable mutation. Drisapersen was generally well tolerated, with injection-site reactions and renal events as most commonly reported adverse events. A nonsignificant treatment difference (P = 0.415) in the change from baseline in six-minute walk distance (6MWD; primary efficacy endpoint) of 10.3 meters in favor of drisapersen was observed at week 48. Key secondary efficacy endpoints (North Star Ambulatory Assessment, 4-stair climb ascent velocity, and 10-meter walk/run velocity) gave consistent findings. Lack of statistical significance was thought to be largely due to greater data variability and subgroup heterogeneity. The increased standard deviation alone, due to less stringent inclusion/exclusion criteria, reduced the statistical power from pre-specified 90% to actual 53%. Therefore, a post-hoc analysis was performed in 80 subjects with a baseline 6MWD 300-400 meters and ability to rise from floor. A statistically significant improvement in 6MWD of 35.4 meters (P = 0.039) in favor of drisapersen was observed in this subpopulation. Results suggest that drisapersen could have benefit in a less impaired population of DMD subjects.
Collapse
Affiliation(s)
- Nathalie Goemans
- Department of Pediatrics and Child Neurology, University Hospitals Leuven, Leuven, Belgium.
| | | | - Elena Belousova
- Research and Clinical Institute of Pediatrics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Hirofumi Komaki
- Department of Child Neurology, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Alberto Dubrovsky
- Fundacion Cenit, Instituto de Neurociencias, Fundación Favaloro, Buenos Aires, Argentina
| | - Craig M McDonald
- School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - John E Kraus
- GlaxoSmithKline, Research Triangle Park, NC, USA
| | | | | | | | | | - Craig Campbell
- Paediatric Neurology, Schulich School of Medicine, Western University, London, Canada
| | | |
Collapse
|
30
|
Larkindale J, Porter JD. Seeking a better landscape for therapy development in neuromuscular disorders. Muscle Nerve 2017; 57:16-19. [PMID: 28881009 DOI: 10.1002/mus.25961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2017] [Indexed: 11/10/2022]
Abstract
Although the neuromuscular field has seen accelerated approval of a drug for Duchenne muscular dystrophy (DMD) and full approval of one for spinal muscular atrophy, these experiences have shown that objective data and an adequate level of effect are essential for drug approval and reimbursement. The appropriateness and validity of biomarkers and clinically meaningful endpoints and an understanding of disease progression rates all played essential roles in the levels of evidence for these drugs. Such tools are best developed through integration of clinical data. The siloing of clinical data for rare neuromuscular diseases represents a considerable barrier to achieving better care and novel therapies for patients living with neuromuscular diseases. We discuss a data-sharing model implemented for DMD and urge cultural changes in the ways natural history and clinical trial data are collected and shared across all neuromuscular diseases in order to benefit the primary stakeholder, the patient. Muscle Nerve 57: 16-19, 2018.
Collapse
Affiliation(s)
- Jane Larkindale
- Duchenne Regulatory Science Consortium, Critical Path Institute, 1730 East River Road, Tucson, Arizona, 85718, USA
| | - John D Porter
- Myotonic Dystrophy Foundation, San Francisco, California, USA
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW To summarize current literature describing the respiratory complications of neuromuscular disease (NMD) and the effect of respiratory interventions and to explore new gene therapies for patients with NMD. RECENT FINDINGS Measurements of respiratory function focus on vital capacity and maximal inspiratory and expiratory pressure and show decline over time. Management of respiratory complications includes lung volume recruitment, mechanical insufflation-exsufflation, chest physiotherapy and assisted ventilation. Lung volume recruitment can slow the progression of lung restriction. New gene-specific therapies for Duchenne muscular dystrophy and spinal muscular atrophy have the potential to preserve respiratory function longitudinally. However, the long-term therapeutic benefit remains unknown. SUMMARY Although NMDs are heterogeneous, many lead to progressive muscle weakness that compromises the function of the respiratory system including upper airway tone, cough and secretion clearance and chest wall support. Respiratory therapies augment or support the normal function of these components of the respiratory system. From a respiratory perspective, the new mutation and gene-specific therapies for NMD are likely to confer long-term therapeutic benefit. More sensitive and standard tools to assess respiratory function longitudinally are needed to monitor respiratory complications in children with NMD, particularly the youngest patients.
Collapse
|
32
|
Guiraud S, Edwards B, Squire SE, Babbs A, Shah N, Berg A, Chen H, Davies KE. Identification of serum protein biomarkers for utrophin based DMD therapy. Sci Rep 2017; 7:43697. [PMID: 28252048 PMCID: PMC5333102 DOI: 10.1038/srep43697] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/30/2017] [Indexed: 12/18/2022] Open
Abstract
Despite promising therapeutic avenues, there is currently no effective treatment for Duchenne muscular dystrophy (DMD), a lethal monogenic disorder caused by the loss of the large cytoskeletal protein, dystrophin. A highly promising approach to therapy, applicable to all DMD patients irrespective to their genetic defect, is to modulate utrophin, a functional paralogue of dystrophin, able to compensate for the primary defects of DMD restoring sarcolemmal stability. One of the major difficulties in assessing the effectiveness of therapeutic strategies is to define appropriate outcome measures. In the present study, we utilised an aptamer based proteomics approach to profile 1,310 proteins in plasma of wild-type, mdx and Fiona (mdx overexpressing utrophin) mice. Comparison of the C57 and mdx sera revealed 83 proteins with statistically significant >2 fold changes in dystrophic serum abundance. A large majority of previously described biomarkers (ANP32B, THBS4, CAMK2A/B/D, CYCS, CAPNI) were normalised towards wild-type levels in Fiona animals. This work also identified potential mdx markers specific to increased utrophin (DUS3, TPI1) and highlights novel mdx biomarkers (GITR, MYBPC1, HSP60, SIRT2, SMAD3, CNTN1). We define a panel of putative protein mdx biomarkers to evaluate utrophin based strategies which may help to accelerate their translation to the clinic.
Collapse
Affiliation(s)
- Simon Guiraud
- Medical Research Council Functional Genomics Unit at the University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, OX1 3PT, United Kingdom
| | - Benjamin Edwards
- Medical Research Council Functional Genomics Unit at the University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, OX1 3PT, United Kingdom
| | - Sarah E Squire
- Medical Research Council Functional Genomics Unit at the University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, OX1 3PT, United Kingdom
| | - Arran Babbs
- Medical Research Council Functional Genomics Unit at the University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, OX1 3PT, United Kingdom
| | - Nandini Shah
- Medical Research Council Functional Genomics Unit at the University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, OX1 3PT, United Kingdom
| | - Adam Berg
- Medical Research Council Functional Genomics Unit at the University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, OX1 3PT, United Kingdom
| | - Huijia Chen
- Medical Research Council Functional Genomics Unit at the University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, OX1 3PT, United Kingdom
| | - Kay E Davies
- Medical Research Council Functional Genomics Unit at the University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, OX1 3PT, United Kingdom
| |
Collapse
|