1
|
Kiefer M, Townsend E, Goncalves C, Shellenbarger KC, Gochyyev P, Wong BL. Appendicular lean mass index and motor function in ambulatory patients with Duchenne muscular dystrophy. Muscle Nerve 2024; 70:226-231. [PMID: 38837739 DOI: 10.1002/mus.28173] [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: 08/16/2023] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
INTRODUCTION/AIMS Appendicular lean mass index (ALMI) has been linked to motor function in patients with Duchenne muscular dystrophy (DMD). However, quantification of the relationship between ALMI and disease-specific clinical outcome assessment trajectories is needed. The purpose of this study was to determine associations between dual-energy x-ray absorptiometry (DXA) derived estimates of ALMI and motor function in ambulatory patients with DMD. METHODS A retrospective analysis of longitudinal clinical visit data from 137 glucocorticoid-treated patients with DMD collected via structured motor assessment protocol evaluated associations between ALMI and motor function indexed by the North Star Ambulatory Assessment (NSAA) and 10 Meter Walk/run Test (10MWT). Body composition was assessed using DXA. ALMI was calculated by dividing arm and leg lean mass by height in m2; fat mass index (FMI) was calculated by dividing whole body fat mass by height in m2. Linear mixed-effects models were used to estimate associations between ALMI and motor function, controlling for age and FMI. RESULTS The full prediction model (age, age,2 ALMI, and FMI) explained 57% of the variance in NSAA scores and 63% of the variance in 10MWT speed. A 1 kg/m2 higher ALMI value predicted a 5.4-point higher NSAA score (p < .001) and 0.45 m/s faster 10MWT speed (p < .001). A 1 kg/m2 higher FMI value predicted a 1.5-point lower NSAA score (p < .001) and 0.14 meters/second slower 10MWT speed (p < .001). DISCUSSION DXA-derived estimates of ALMI and FMI are associated with motor function in DMD and may explain variation in DMD disease progression.
Collapse
Affiliation(s)
- Michael Kiefer
- School of Health and Rehabilitation Sciences, MGH Institute of Health Professions, Boston, Massachusetts, USA
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Physical Therapy, College of Health Professions, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Elise Townsend
- School of Health and Rehabilitation Sciences, MGH Institute of Health Professions, Boston, Massachusetts, USA
| | - Celina Goncalves
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - K Courtney Shellenbarger
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Perman Gochyyev
- School of Health and Rehabilitation Sciences, MGH Institute of Health Professions, Boston, Massachusetts, USA
| | - Brenda L Wong
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
2
|
Muntoni F, Signorovitch J, Sajeev G, Done N, Yao Z, Goemans N, McDonald C, Mercuri E, Niks EH, Wong B, Vandenborne K, Straub V, de Groot IJM, Tian C, Manzur A, Dieye I, Lane H, Ward SJ, Servais L. Meaningful changes in motor function in Duchenne muscular dystrophy (DMD): A multi-center study. PLoS One 2024; 19:e0304984. [PMID: 38985784 PMCID: PMC11236155 DOI: 10.1371/journal.pone.0304984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 05/20/2024] [Indexed: 07/12/2024] Open
Abstract
Evaluations of treatment efficacy in Duchenne muscular dystrophy (DMD), a rare genetic disease that results in progressive muscle wasting, require an understanding of the 'meaningfulness' of changes in functional measures. We estimated the minimal detectable change (MDC) for selected motor function measures in ambulatory DMD, i.e., the minimal degree of measured change needed to be confident that true underlying change has occurred rather than transient variation or measurement error. MDC estimates were compared across multiple data sources, representing >1000 DMD patients in clinical trials and real-world clinical practice settings. Included patients were ambulatory, aged ≥4 to <18 years and receiving steroids. Minimal clinically important differences (MCIDs) for worsening were also estimated. Estimated MDC thresholds for >80% confidence in true change were 2.8 units for the North Star Ambulatory Assessment (NSAA) total score, 1.3 seconds for the 4-stair climb (4SC) completion time, 0.36 stairs/second for 4SC velocity and 36.3 meters for the 6-minute walk distance (6MWD). MDC estimates were similar across clinical trial and real-world data sources, and tended to be slightly larger than MCIDs for these measures. The identified thresholds can be used to inform endpoint definitions, or as benchmarks for monitoring individual changes in motor function in ambulatory DMD.
Collapse
Affiliation(s)
- Francesco Muntoni
- Dubowitz Neuromuscular Centre, NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, London, United Kingdom
| | - 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
| | - Nicolae Done
- Analysis Group, Inc., Boston, Massachusetts, United States of America
| | - Zhiwen Yao
- Analysis Group, Inc., Boston, Massachusetts, United States of America
| | | | - Craig McDonald
- Department of Physical Medicine and Rehabilitation and Pediatrics, University of California, Davis, Sacramento, California, United States of America
| | - Eugenio Mercuri
- Department of Pediatric Neurology, Fondazione Policlinico Gemelli IRCCS, Catholic University, Rome, Italy
| | - Erik H. Niks
- Department of Neurology, Leiden University Medical Centre, Leiden, Netherlands
| | - Brenda Wong
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, Florida, United States of America
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Imelda J. M. de Groot
- Department of Rehabilitation, Donders Centre of Neuroscience, Radboud University Nijmegen Medical Center, Nijmegen, Netherlands
| | - Cuixia Tian
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio & College of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Adnan Manzur
- Dubowitz Neuromuscular Centre, NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, London, United Kingdom
| | - Ibrahima Dieye
- Analysis Group, Inc., Boston, Massachusetts, United States of America
| | - Henry Lane
- Analysis Group, Inc., Boston, Massachusetts, United States of America
| | - Susan J. Ward
- The collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
| | - Laurent Servais
- Department of Paediatrics, MDUK Oxford Neuromuscular Center, University of Oxford, Oxford, United Kingdom
- Neuromuscular Center of Liège, Division of Paediatrics, CHU and University of Liège, Liège, Belgium
| | | | | | | | | | | |
Collapse
|
3
|
McDonald CM, Signorovitch J, Mercuri E, Niks EH, Wong B, Fillbrunn M, Sajeev G, Yim E, Dieye I, Miller D, Ward SJ, Goemans N. Functional trajectories before and after loss of ambulation in Duchenne muscular dystrophy and implications for clinical trials. PLoS One 2024; 19:e0304099. [PMID: 38829874 PMCID: PMC11146704 DOI: 10.1371/journal.pone.0304099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 04/24/2024] [Indexed: 06/05/2024] Open
Abstract
This study examined functional trajectories of subjects during the transition phase between ambulatory and non-ambulatory Duchenne muscular dystrophy (DMD) to inform clinical trial designs for new therapeutics. Ambulatory, pulmonary, and upper limb function leading up to loss of ambulation (LoA) and non-ambulatory measures following LoA were quantified; time ordering of pulmonary and upper limb milestones relative to LoA were determined; and the 10-second time threshold for 10-meter walk/run (10MWR) as a marker of approaching LOA was explored. Included in this analysis were 51 subjects aged between 7 and 18 years who experienced LoA during follow-up in the PRO-DMD-01 natural history study. Mean age at LoA was 12.7 (7.1-18.6) years. Mean annual rates of decline in forced vital capacity (FVC) <80%-predicted and performance of upper limb (PUL) 1.2 total score were smaller before than after LoA, but not significantly (FVC %-predicted: 5.6% vs. 10.1%, p = 0.21; PUL 1.2 total score: 2.3 vs. 3.8 units, p = 0.20). More than half of patients experienced clinically significant deficits in FVC %-predicted and PUL 1.2 before experiencing LoA. Among subjects with baseline 10MWR >10 s, those with <1 year to LoA had similar mean ages but significantly worse mean ambulatory function at baseline compared to those with ≥1 year to LoA. Enriching DMD clinical trials for patients with declining pulmonary or upper limb function is achievable without restricting enrollment to non-ambulatory patients. The sequencing of LoA and initial deficits in pulmonary and upper limb function varied across patients and highlights the potential for composite outcomes or multi-outcome trial designs to assess disease-modifying therapies more comprehensively.
Collapse
Affiliation(s)
- Craig M. McDonald
- Department of Physical Medicine and Rehabilitation and Department of Pediatrics, University of California Davis Health System, Sacramento, California, United States of America
| | - James Signorovitch
- Analysis Group Inc., Boston, Massachusetts, United States of America
- Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
| | - Eugenio Mercuri
- Child Neurology Unit e Centro Nemo, IRCCS Fondazione Policlinico Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Erik H. Niks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Brenda Wong
- Department of Pediatrics and Neurology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Mirko Fillbrunn
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Gautam Sajeev
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Erica Yim
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Ibrahima Dieye
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Debra Miller
- CureDuchenne, Newport Beach, California, United States of America
| | - Susan J. Ward
- Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
| | | | | |
Collapse
|
4
|
Vandekerckhove I, Papageorgiou E, Hanssen B, De Beukelaer N, Van den Hauwe M, Goemans N, Van Campenhout A, De Waele L, De Groote F, Desloovere K. Gait classification for growing children with Duchenne muscular dystrophy. Sci Rep 2024; 14:10828. [PMID: 38734731 PMCID: PMC11088636 DOI: 10.1038/s41598-024-61231-y] [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: 08/31/2023] [Accepted: 05/02/2024] [Indexed: 05/13/2024] Open
Abstract
Classifying gait patterns into homogeneous groups could enhance communication among healthcare providers, clinical decision making and clinical trial designs in boys with Duchenne muscular dystrophy (DMD). Sutherland's classification has been developed 40 years ago. Ever since, the state-of-the-art medical care has improved and boys with DMD are now longer ambulatory. Therefore, the gait classification requires an update. The overall aim was to develop an up-to-date, valid DMD gait classification. A total of 137 three-dimensional gait analysis sessions were collected in 30 boys with DMD, aged 4.6-17 years. Three classes were distinguished, which only partly aligned with increasing severity of gait deviations. Apart from the mildly affected pattern, two more severely affected gait patterns were found, namely the tiptoeing pattern and the flexion pattern with distinct anterior pelvic tilt and posterior trunk leaning, which showed most severe deviations at the ankle or at the proximal segments/joints, respectively. The agreement between Sutherland's and the current classification was low, suggesting that gait pathology with the current state-of-the-art medical care has changed. However, overlap between classes, especially between the two more affected classes, highlights the complexity of the continuous gait changes. Therefore, caution is required when classifying individual boys with DMD into classes.
Collapse
Affiliation(s)
| | | | - Britta Hanssen
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
| | - Nathalie De Beukelaer
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
- Department of Surgery, University of Geneva, Geneva, Switzerland
| | - Marleen Van den Hauwe
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
- Department of Child Neurology, University Hospital Leuven, Leuven, Belgium
| | - Nathalie Goemans
- Department of Child Neurology, University Hospital Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Anja Van Campenhout
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Orthopedics, University Hospital Leuven, Leuven, Belgium
| | - Liesbeth De Waele
- Department of Child Neurology, University Hospital Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | | | - Kaat Desloovere
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
- Clinical Motion Analysis Laboratory, University Hospital Leuven, Pellenberg, Belgium
| |
Collapse
|
5
|
Mercuri E, Vilchez JJ, Boespflug-Tanguy O, Zaidman CM, Mah JK, Goemans N, Müller-Felber W, Niks EH, Schara-Schmidt U, Bertini E, Comi GP, Mathews KD, Servais L, Vandenborne K, Johannsen J, Messina S, Spinty S, McAdam L, Selby K, Byrne B, Laverty CG, Carroll K, Zardi G, Cazzaniga S, Coceani N, Bettica P, McDonald CM. Safety and efficacy of givinostat in boys with Duchenne muscular dystrophy (EPIDYS): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Neurol 2024; 23:393-403. [PMID: 38508835 DOI: 10.1016/s1474-4422(24)00036-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Duchenne muscular dystrophy, the most common childhood muscular dystrophy, is caused by dystrophin deficiency. Preclinical and phase 2 study data have suggested that givinostat, a histone deacetylase inhibitor, might help to counteract the effects of this deficiency. We aimed to evaluate the safety and efficacy of givinostat in the treatment of Duchenne muscular dystrophy. METHODS This multicentre, randomised, double-blind, placebo-controlled, phase 3 trial was done at 41 tertiary care sites in 11 countries. Eligible participants were ambulant, male, and aged at least 6 years, had a genetically confirmed diagnosis of Duchenne muscular dystrophy, completed two four-stair climb assessments with a mean of 8 s or less (≤1 s variance), had a time-to-rise of at least 3 s but less than 10 s, and had received systemic corticosteroids for at least 6 months. Participating boys were randomly assigned (2:1, allocated according to a list generated by the interactive response technology provider) to receive either oral givinostat or matching placebo twice a day for 72 weeks, stratified by concomitant steroid use. Boys, investigators, and site and sponsor staff were masked to treatment assignment. The dose was flexible, based on weight, and was reduced if not tolerated. Boys were divided into two groups on the basis of their baseline vastus lateralis fat fraction (VLFF; measured by magnetic resonance spectroscopy): group A comprised boys with a VLFF of more than 5% but no more than 30%, whereas group B comprised boys with a VLFF of 5% or less, or more than 30%. The primary endpoint compared the effects of givinostat and placebo on the change in results of the four-stair climb assessment between baseline and 72 weeks, in the intention-to-treat, group A population. Safety was assessed in all randomly assigned boys who received at least one dose of study drug. When the first 50 boys in group A completed 12 months of treatment, an interim futility assessment was conducted, after which the sample size was adapted using masked data from the four-stair climb assessments. Furthermore, the starting dose of givinostat was reduced following a protocol amendment. This trial is registered with ClinicalTrials.gov, NCT02851797, and is complete. FINDINGS Between June 6, 2017, and Feb 22, 2022, 359 boys were assessed for eligibility. Of these, 179 were enrolled into the study (median age 9·8 years [IQR 8·1-11·0]), all of whom were randomly assigned (118 to receive givinostat and 61 to receive placebo); 170 (95%) boys completed the study. Of the 179 boys enrolled, 120 (67%) were in group A (81 givinostat and 39 placebo); of these, 114 (95%) completed the study. For participants in group A, comparing the results of the four-stair climb assessment at 72 weeks and baseline, the geometric least squares mean ratio was 1·27 (95% CI 1·17-1·37) for boys receiving givinostat and 1·48 (1·32-1·66) for those receiving placebo (ratio 0·86, 95% CI 0·745-0·989; p=0·035). The most common adverse events in the givinostat group were diarrhoea (43 [36%] of 118 boys vs 11 [18%] of 61 receiving placebo) and vomiting (34 [29%] vs 8 [13%]); no treatment-related deaths occurred. INTERPRETATION Among ambulant boys with Duchenne muscular dystrophy, results of the four-stair climb assessment worsened in both groups over the study period; however, the decline was significantly smaller with givinostat than with placebo. The dose of givinostat was reduced after an interim safety analysis, but no new safety signals were reported. An ongoing extension study is evaluating the long-term safety and efficacy of givinostat in patients with Duchenne muscular dystrophy. FUNDING Italfarmaco.
Collapse
Affiliation(s)
- Eugenio Mercuri
- Pediatric Neurology, Department of Woman and Child Health and Public Health, Child Health Area, Universita Cattolica del Sacro Cuore, Rome, Italy; Centro Clinico Nemo Fondazione Policlinico Gemelli IRCCS, Rome, Italy.
| | - Juan J Vilchez
- Servicio de Neurología, Neuromuscular Unit, CIBERER, EURO-RN-NMD, Hospital Universitario y Politécnico La Fe Valencia, Valencia, Spain
| | - Odile Boespflug-Tanguy
- I-Motion, Institut de Myologie, Hôpital Armand-Trousseau, APHP, Sorbonne Université, Paris, France; Université Paris Cité UMR INSERM 1141, Hôpital Robert Debré, Paris, France
| | | | - Jean K Mah
- Division of Pediatric Neurology, Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nathalie Goemans
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Wolfgang Müller-Felber
- LMU Munich, University Hospital, Hauner Children's Hospital, Pediatric Neurology and Developmental Medicine, Munich, Germany
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands; Duchenne Center Netherlands, Netherlands
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology, Children's University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Enrico Bertini
- Research Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giacomo P Comi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Milan, Italy; Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Katherine D Mathews
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Laurent Servais
- MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; Neuromuscular Reference Center, Department of Paediatrics, University and University Hospital of Liege, Belgium
| | - Krista Vandenborne
- ImagingDMD, University of Florida, Gainesville, FL, USA; Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Jessika Johannsen
- University Medical Center Hamburg-Eppendorf, Department of Pediatrics, Hamburg, Germany
| | - Sonia Messina
- Department of Clinical and Experimental Medicine, Unit of Neurodegenerative Diseases, AOU Policlinico G Martino, University of Mesina, Messina, Italy
| | - Stefan Spinty
- Department of Paediatric Neurology, Alder Hey Children's Hospital NHS Trust, Liverpool, UK
| | - Laura McAdam
- Holland Bloorview Kids Rehabilitation Hospital, Bloorview Research Institute, Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Kathryn Selby
- The University of British Columbia, Children's and Women's Health Centre, Vancouver, BC, Canada
| | - Barry Byrne
- Child Health Research Institute, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Chamindra G Laverty
- Department of Neuroscience, University of California, San Diego, San Diego, CA, USA
| | | | | | | | | | | | | |
Collapse
|
6
|
Billich N, Bray P, Truby H, Evans M, Ryan MM, Carroll K, de Valle K, Villano D, Kornberg A, Sowerby B, Farrar MA, Menezes MP, Holland S, Lindeback R, Cairns A, Davidson ZE. Exploring caregivers' attitudes and beliefs about nutrition and weight management for young people with Duchenne muscular dystrophy. Muscle Nerve 2024; 69:448-458. [PMID: 38353293 DOI: 10.1002/mus.28062] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION/AIMS Obesity disproportionately affects children and adolescents with Duchenne muscular dystrophy (DMD) and with adverse consequences for disease progression. This study aims to: explore barriers, enablers, attitudes, and beliefs about nutrition and weight management; and to obtain caregiver preferences for the design of a weight management program for DMD. METHODS We surveyed caregivers of young people with DMD from four Australian pediatric neuromuscular clinics. Survey questions were informed by the Theoretical Domains Framework and purposefully designed to explore barriers and enablers to food and weight management. Caregivers were asked to identify their preferred features in a weight management program for families living with DMD. RESULTS Fifty-three caregivers completed the survey. Almost half (48%) perceived their son as above healthy weight. Consequences for those children were perceived to be self-consciousness (71%), a negative impact on self-esteem (64%) and movement (57%). Preventing weight gain was a common reason for providing healthy food and healthy eating was a high priority for families. Barriers to that intention included: time constraints, selective food preferences, and insufficient nutrition information. Caregivers preferred an intensive six-week weight management program addressing appetite management and screen time. DISCUSSION Managing weight is an important issue for caregivers of sons with DMD; yet several barriers exist. Individualized 6 week programs are preferred by caregivers to improve weight management for DMD.
Collapse
Affiliation(s)
- Natassja Billich
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Department of Neurology, The Royal Children's Hospital, Melbourne, Australia
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Australia
| | - Paula Bray
- Children's Hospital, Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Department of Neurology, The Children's Hospital at Westmead, Sydney, Australia
| | - Helen Truby
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Australia
- School of Primary and Allied Health Care Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Australia
| | - Maureen Evans
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Department of Metabolic Medicine, The Royal Children's Hospital, Melbourne, Australia
| | - Monique M Ryan
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Department of Neurology, The Royal Children's Hospital, Melbourne, Australia
- Neurosciences Group, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, Melbourne University, Melbourne, Australia
| | - Kate Carroll
- Department of Neurology, The Royal Children's Hospital, Melbourne, Australia
- Neurosciences Group, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Physiotherapy, School of Primary and Allied Health Care, Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Australia
| | - Katy de Valle
- Department of Neurology, The Royal Children's Hospital, Melbourne, Australia
- Neurosciences Group, Murdoch Children's Research Institute, Melbourne, Australia
| | - Daniella Villano
- Department of Neurology, The Royal Children's Hospital, Melbourne, Australia
- Neurosciences Group, Murdoch Children's Research Institute, Melbourne, Australia
| | - Andrew Kornberg
- Department of Neurology, The Royal Children's Hospital, Melbourne, Australia
- Neurosciences Group, Murdoch Children's Research Institute, Melbourne, Australia
| | - Bianca Sowerby
- Department of Neurology, The Royal Children's Hospital, Melbourne, Australia
| | - Michelle A Farrar
- Department of Neurology, Sydney Children's Hospital, Sydney, Australia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Sydney, Australia
| | - Manoj P Menezes
- Children's Hospital, Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Department of Neurology, The Children's Hospital at Westmead, Sydney, Australia
| | - Sandra Holland
- Department of Neurology, Sydney Children's Hospital, Sydney, Australia
| | - Rachel Lindeback
- Department of Neurology, Sydney Children's Hospital, Sydney, Australia
| | - Anita Cairns
- Department of Neurology, Queensland Children's Hospital, Brisbane, Australia
| | - Zoe E Davidson
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Department of Neurology, The Royal Children's Hospital, Melbourne, Australia
- Neurosciences Group, Murdoch Children's Research Institute, Melbourne, Australia
| |
Collapse
|
7
|
Hernández-Sánchez A, Parra-Sánchez L, Montolio M, Rueda-Ruzafa L, Ortiz-Comino L, Sánchez-Joya MDM. Family Involvement and at-Home Physical Therapy on Duchenne Muscular Dystrophy: A Randomized Controlled Trial. Pediatr Neurol 2024; 152:34-40. [PMID: 38184986 DOI: 10.1016/j.pediatrneurol.2023.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 11/27/2023] [Accepted: 12/18/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a genetic condition that causes muscle weakness and begins in early childhood. To treat its complications, the rehabilitation program includes physical therapy, mainly on the musculoskeletal and the respiratory complications that appear on the evolution of the disease. This study aims to explore the effects of physical therapy with or without an at-home program on motor function among children with DMD. METHODS A randomized controlled trial was carried out for one year (one group with at-home and conventional physical therapy and another with conventional physical therapy). Motor function was measured using the Motor Function Measure (MFM) scale, the Vignos and Brooke scales, the Timed-up-and-Go test, and the six-minute walk distance test. RESULTS Twenty-seven participants with DMD participated in this study. In the at-home and conventional physical therapy group, better motor function at the distal and global level was maintained, per the results of the MFM scale (P < 0.05). The rest of the variables did not achieve statistically significant changes. CONCLUSIONS Our results suggest that complementing conventional treatment with at-home treatment in which the family is involved maintains better motor function, in participants with DMD.
Collapse
Affiliation(s)
- Andrea Hernández-Sánchez
- Faculty of Health Sciences, Department of Nursing, Physical Therapy and Medicine, University of Almería, Almería, Spain
| | | | - Marisol Montolio
- Duchenne Parent Project España, Madrid, Spain; Faculty of Biology, Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
| | - Lola Rueda-Ruzafa
- Faculty of Health Sciences, Department of Nursing, Physical Therapy and Medicine, University of Almería, Almería, Spain
| | - Lucía Ortiz-Comino
- Faculty of Health Sciences (Melilla), Department of Physical Therapy, University of Granada, Melilla, Spain; Sport and Health Research Center (IMUDs), Granada, Spain; Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain.
| | - María Del Mar Sánchez-Joya
- Faculty of Health Sciences, Department of Nursing, Physical Therapy and Medicine, University of Almería, Almería, Spain
| |
Collapse
|
8
|
McDonald C, Camino E, Escandon R, Finkel RS, Fischer R, Flanigan K, Furlong P, Juhasz R, Martin AS, Villa C, Sweeney HL. Draft Guidance for Industry Duchenne Muscular Dystrophy, Becker Muscular Dystrophy, and Related Dystrophinopathies - Developing Potential Treatments for the Entire Spectrum of Disease. J Neuromuscul Dis 2024; 11:499-523. [PMID: 38363616 DOI: 10.3233/jnd-230219] [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] [Indexed: 02/17/2024]
Abstract
Background Duchenne muscular dystrophy (DMD) and related dystrophinopathies are neuromuscular conditions with great unmet medical needs that require the development of effective medical treatments. Objective To aid sponsors in clinical development of drugs and therapeutic biological products for treating DMD across the disease spectrum by integrating advancements, patient registries, natural history studies, and more into a comprehensive guidance. Methods This guidance emerged from collaboration between the FDA, the Duchenne community, and industry stakeholders. It entailed a structured approach, involving multiple committees and boards. From its inception in 2014, the guidance underwent revisions incorporating insights from gene therapy studies, cardiac function research, and innovative clinical trial designs. Results The guidance provides a deeper understanding of DMD and its variants, focusing on patient engagement, diagnostic criteria, natural history, biomarkers, and clinical trials. It underscores patient-focused drug development, the significance of dystrophin as a biomarker, and the pivotal role of magnetic resonance imaging in assessing disease progression. Additionally, the guidance addresses cardiomyopathy's prominence in DMD and the burgeoning field of gene therapy. Conclusions The updated guidance offers a comprehensive understanding of DMD, emphasizing patient-centric approaches, innovative trial designs, and the importance of biomarkers. The focus on cardiomyopathy and gene therapy signifies the evolving realm of DMD research. It acts as a crucial roadmap for sponsors, potentially leading to improved treatments for DMD.
Collapse
Affiliation(s)
| | - Eric Camino
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Rafael Escandon
- DGBI Consulting, LLC, Bainbridge Island, Washington, DC, USA
| | | | - Ryan Fischer
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Kevin Flanigan
- Center for Experimental Neurotherapeutics, Department of Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Pat Furlong
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Rose Juhasz
- Nationwide Children's Hospital, Columbus, OH, USA
| | - Ann S Martin
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Chet Villa
- Trinity Health Michigan, Grand Rapids, MI, USA
| | - H Lee Sweeney
- Cincinnati Children's Hospital Medical Center within the UC Department of Pediatrics, Cincinnati, OH, USA
| |
Collapse
|
9
|
Zaidman CM, Proud CM, McDonald CM, Lehman KJ, Goedeker NL, Mason S, Murphy AP, Guridi M, Wang S, Reid C, Darton E, Wandel C, Lewis S, Malhotra J, Griffin DA, Potter RA, Rodino-Klapac LR, Mendell JR. Delandistrogene Moxeparvovec Gene Therapy in Ambulatory Patients (Aged ≥4 to <8 Years) with Duchenne Muscular Dystrophy: 1-Year Interim Results from Study SRP-9001-103 (ENDEAVOR). Ann Neurol 2023; 94:955-968. [PMID: 37539981 DOI: 10.1002/ana.26755] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/05/2023]
Abstract
OBJECTIVE Delandistrogene moxeparvovec is approved in the USA for the treatment of ambulatory patients (4-5 years) with Duchenne muscular dystrophy. ENDEAVOR (SRP-9001-103; NCT04626674) is a single-arm, open-label study to evaluate delandistrogene moxeparvovec micro-dystrophin expression, safety, and functional outcomes following administration of commercial process delandistrogene moxeparvovec. METHODS In cohort 1 of ENDEAVOR (N = 20), eligible ambulatory males, aged ≥4 to <8 years, received a single intravenous infusion of delandistrogene moxeparvovec (1.33 × 1014 vg/kg). The primary endpoint was change from baseline (CFBL) to week 12 in delandistrogene moxeparvovec micro-dystrophin by western blot. Additional endpoints evaluated included: safety; vector genome copies; CFBL to week 12 in muscle fiber-localized micro-dystrophin by immunofluorescence; and functional assessments, including North Star Ambulatory Assessment, with comparison with a propensity score-weighted external natural history control. RESULTS The 1-year safety profile of commercial process delandistrogene moxeparvovec in ENDEAVOR was consistent with safety data reported in other delandistrogene moxeparvovec trials (NCT03375164 and NCT03769116). Delandistrogene moxeparvovec micro-dystrophin expression was robust, with sarcolemmal localization at week 12; mean (SD) CFBL in western blot, 54.2% (42.6); p < 0.0001. At 1 year, patients demonstrated stabilized or improved North Star Ambulatory Assessment total scores; mean (SD) CFBL, +4.0 (3.5). Treatment versus a propensity score-weighted external natural history control demonstrated a statistically significant difference in least squares mean (standard error) CFBL in North Star Ambulatory Assessment, +3.2 (0.6) points; p < 0.0001. INTERPRETATION Results confirm efficient transduction of muscle by delandistrogene moxeparvovec. One-year post-treatment, delandistrogene moxeparvovec was well tolerated, and demonstrated stabilized or improved motor function, suggesting a clinical benefit for patients with Duchenne muscular dystrophy. ANN NEUROL 2023;94:955-968.
Collapse
Affiliation(s)
- Craig M Zaidman
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Crystal M Proud
- Children's Hospital of the King's Daughters, Norfolk, VA, USA
| | | | - Kelly J Lehman
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA
| | - Natalie L Goedeker
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | | | | | | | | | - Carol Reid
- Roche Products Ltd, Welwyn Garden City, UK
| | | | | | - Sarah Lewis
- Sarepta Therapeutics, Inc., Cambridge, MA, USA
| | | | | | | | | | - Jerry R Mendell
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA
- The Ohio State University, Columbus, OH, USA
| |
Collapse
|
10
|
Naarding KJ, Stimpson G, Ward SJ, Goemans N, McDonald C, Mercuri E, Muntoni F. 269th ENMC international workshop: 10 years of clinical trials in Duchenne muscular dystrophy - What have we learned? 9-11 December 2022, Hoofddorp, The Netherlands. Neuromuscul Disord 2023; 33:897-910. [PMID: 37926638 DOI: 10.1016/j.nmd.2023.10.003] [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: 09/30/2023] [Accepted: 10/10/2023] [Indexed: 11/07/2023]
Abstract
There are multiple avenues for therapeutic development in Duchenne muscular dystrophy (DMD), which are highlighted in the first section of this report for the "10 years of Clinical trials in DMD - What have we learned?" workshop. This report then provides an overview of the presentations made at the workshop grouped into the following core themes: trial outcomes, disease heterogeneity, meaningfulness of outcomes and the utility of real-world data in trials. Finally, we present the consensus that was achieved at the workshop on the learning points from 10 years of clinical trials in DMD, and possible action points from these. This includes further work in expanding the scope and range of trial outcomes and assessing the efficacy of new trial structures for DMD. We also highlight several points which should be addressed during future interactions with regulators, such as clinical meaningfulness and the use of real-world data.
Collapse
Affiliation(s)
- Karin J Naarding
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands; Duchenne Center Netherlands, the Netherlands
| | - Georgia Stimpson
- UCL Great Ormond Street Institute of Child Health, Dubowitz Neuromuscular Centre, London, UK
| | - Susan J Ward
- Collaborative Trajectory Analysis Project (cTAP), United States
| | - Nathalie Goemans
- University Hospitals Leuven, Dept of Child Neurology, Leuven, Belgium
| | - Craig McDonald
- Department of Physical Medicine and Rehabilitation in Sacramento, University of California, Davis, CA, United States
| | - Eugenio Mercuri
- Pediatric Neurology Unit, Catholic University, Rome, Italy; Centro Clinico Nemo, U.O.C. Neuropsichiatria Infantile Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesco Muntoni
- UCL Great Ormond Street Institute of Child Health, Dubowitz Neuromuscular Centre, London, UK; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
11
|
Brogna C, Pane M, Coratti G, D'Amico A, Pegoraro E, Bello L, Sansone VAM, Albamonte E, Messina S, Pini A, D'Angelo MG, Bruno C, Mongini T, Ricci FS, Berardinelli A, Battini R, Masson R, Bertini ES, Politano L, Mercuri E. Upper Limb Changes in DMD Patients Amenable to Skipping Exons 44, 45, 51 and 53: A 24-Month Study. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10040746. [PMID: 37189996 DOI: 10.3390/children10040746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION The Performance of Upper Limb version 2.0 (PUL 2.0) is increasingly used in Duchenne Muscular Dystrophy (DMD) to study longitudinal functional changes of motor upper limb function in ambulant and non-ambulant patients. The aim of this study was to evaluate changes in upper limb functions in patients carrying mutations amenable to skipping exons 44, 45, 51 and 53. METHODS All DMD patients were assessed using the PUL 2.0 for at least 2 years, focusing on 24-month paired visits in those with mutations eligible for skipping exons 44, 45, 51 and 53. RESULTS 285 paired assessments were available. The mean total PUL 2.0 12-month change was -0.67 (2.80), -1.15 (3.98), -1.46 (3.37) and -1.95 (4.04) in patients carrying mutations amenable to skipping exon 44, 45, 51 and 53, respectively. The mean total PUL 2.0 24-month change was -1.47 (3.73), -2.78 (5.86), -2.95 (4.56) and -4.53 (6.13) in patients amenable to skipping exon 44, 45, 51 and 53, respectively. The difference in PUL 2.0 mean changes among the type of exon skip class for the total score was not significant at 12 months but was significant at 24 months for the total score (p < 0.001), the shoulder (p = 0.01) and the elbow domain (p < 0.001), with patients amenable to skipping exon 44 having smaller changes compared to those amenable to skipping exon 53. There was no difference within ambulant or non-ambulant cohorts when subdivided by exon skip class for the total and subdomains score (p > 0.05). CONCLUSIONS Our results expand the information on upper limb function changes detected by the PUL 2.0 in a relatively large group of DMD patients with distinct exon-skipping classes. This information can be of help when designing clinical trials or in the interpretation of the real world data including non-ambulant patients.
Collapse
Affiliation(s)
- Claudia Brogna
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Marika Pane
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Giorgia Coratti
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Adele D'Amico
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padua, 35128 Padua, Italy
| | - Luca Bello
- Department of Neurosciences, University of Padua, 35128 Padua, Italy
| | - Valeria Ada Maria Sansone
- The NEMO Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, 20162 Milan, Italy
| | - Emilio Albamonte
- The NEMO Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, 20162 Milan, Italy
| | - Sonia Messina
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Antonella Pini
- Neuromuscular Pediatric Unit, IRRCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | | | - Claudio Bruno
- Center of Translational and Experimental Myology and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, IRCCS Istituto Giannina Gaslini and University of Genoa, 16132 Genoa, Italy
| | - Tiziana Mongini
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Torino, 10100 Turin, Italy
| | - Federica Silvia Ricci
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Torino, 10100 Turin, Italy
| | - Angela Berardinelli
- National Neurological Institute C. Mondino Foundation, IRCCS, 27100 Pavia, Italy
| | - Roberta Battini
- Department of Developmental Neuroscience, IRCCS Stella Maris, 56018 Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Riccardo Masson
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Enrico Silvio Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Luisa Politano
- Cardiomiology and Medical Genetics, Department of Experimental Medicine, Università della Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Eugenio Mercuri
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
12
|
van de Velde NM, Koeks Z, Signorelli M, Verwey N, Overzier M, Bakker JA, Sajeev G, Signorovitch J, Ricotti V, Verschuuren J, Brown K, Spitali P, Niks EH. Longitudinal Assessment of Creatine Kinase, Creatine/Creatinine ratio, and Myostatin as Monitoring Biomarkers in Becker Muscular Dystrophy. Neurology 2023; 100:e975-e984. [PMID: 36849458 PMCID: PMC9990441 DOI: 10.1212/wnl.0000000000201609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 10/11/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The slow and variable disease progression of Becker muscular dystrophy (BMD) urges the development of biomarkers to facilitate clinical trials. We explored changes in 3 muscle-enriched biomarkers in serum of patients with BMD over 4-year time and studied associations with disease severity, disease progression, and dystrophin levels in BMD. METHODS We quantitatively measured creatine kinase (CK) using the International Federation of Clinical Chemistry reference method, creatine/creatinineratio (Cr/Crn) using liquid chromatography-tandem mass spectrometry, and myostatin with ELISA in serum and assessed functional performance using the North Star Ambulatory Assessment (NSAA), 10-meter run velocity (TMRv), 6-Minute Walking Test (6MWT), and forced vital capacity in a 4-year prospective natural history study. Dystrophin levels were quantified in the tibialis anterior muscle using capillary Western immunoassay. The correlation between biomarkers, age, functional performance, mean annual change, and prediction of concurrent functional performance was analyzed using linear mixed models. RESULTS Thirty-four patients with 106 visits were included. Eight patients were nonambulant at baseline. Cr/Crn and myostatin were highly patient specific (intraclass correlation coefficient for both = 0.960). Cr/Crn was strongly negatively correlated, whereas myostatin was strongly positively correlated with the NSAA, TMRv, and 6MWT (Cr/Crn rho = -0.869 to -0.801 and myostatin rho = 0.792 to 0.842, all p < 0.001). CK showed a negative association with age (p = 0.0002) but was not associated with patients' performance. Cr/Crn and myostatin correlated moderately with the average annual change of the 6MWT (rho = -0.532 and 0.555, p = 0.02). Dystrophin levels did not correlate with the selected biomarkers nor with performance. Cr/Crn, myostatin, and age could explain up to 75% of the variance of concurrent functional performance of the NSAA, TMRv, and 6MWT. DISCUSSION Both Cr/Crn and myostatin could potentially serve as monitoring biomarkers in BMD, as higher Cr/Crn and lower myostatin were associated with lower motor performance and predictive of concurrent functional performance when combined with age. Future studies are needed to more precisely determine the context of use of these biomarkers.
Collapse
Affiliation(s)
- Nienke M van de Velde
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Zaïda Koeks
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Mirko Signorelli
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Nisha Verwey
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Maurice Overzier
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Jaap A Bakker
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Gautam Sajeev
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - James Signorovitch
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Valeria Ricotti
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Jan Verschuuren
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Kristy Brown
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Pietro Spitali
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Erik H Niks
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom.
| |
Collapse
|
13
|
Muntoni F, Straub V, Servais L, Mercuri E. Letter to the Editor: In response to P.R. Clemens et al., Efficacy and Safety of Viltolarsen in Boys with Duchenne Muscular Dystrophy: Results From the Phase 2, Open-Label, 4-Year Extension Study, and Long-Term Functional Efficacy and Safety of Viltolarsen in Patients with Duchenne Muscular Dystrophy. J Neuromuscul Dis 2023; 10:1151-1153. [PMID: 37955093 PMCID: PMC10657664 DOI: 10.3233/jnd-239004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Affiliation(s)
- Francesco Muntoni
- Dubowitz Neuromuscular Centre and MRC Centre for Neuromuscular Diseases, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital for Children Foundation Trust, London, UK
| | - Volker Straub
- Institute of Genetic Medicine, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| | | | | |
Collapse
|
14
|
Verreydt I, Vandekerckhove I, Stoop E, Peeters N, van Tittelboom V, Van de Walle P, Van den Hauwe M, Goemans N, De Waele L, Van Campenhout A, Hanssen B, Desloovere K. Instrumented strength assessment in typically developing children and children with a neural or neuromuscular disorder: A reliability, validity and responsiveness study. Front Physiol 2022; 13:855222. [PMID: 36338500 PMCID: PMC9627606 DOI: 10.3389/fphys.2022.855222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/27/2022] [Indexed: 01/11/2023] Open
Abstract
The aim of this study was to determine the clinimetric properties, i.e., reliability, validity and responsiveness of an instrumented strength assessment in typically developing (TD) children and children with cerebral palsy (CP) and Duchenne muscular dystrophy (DMD). Force (N), torque (Nm) and normalized torque (Nm/kg) were defined for maximal voluntary isometric contractions (MVICs) of the lower limb muscles using a pre-established protocol. Intraclass correlation coefficient (ICC), standard error of measurement (SEM) and minimal detectable change (MDC) of TD children (n = 14), children with CP (n = 11) and DMD (n = 11) were used to evaluate intra-rater reliability for the three cohorts and the inter-rater intersession as well as inter-rater intrasession reliability for TD children. Construct validity was assessed by comparing MVICs in TD children (n = 28) to children with CP (n = 26) and to children with DMD (n = 30), using the Kruskal Wallis and post-hoc Mann-Whitney U tests. Responsiveness was investigated by assessing changes in MVICs following a strength intervention in CP (n = 26) and a 1 and 2 year follow-up study in DMD (n = 13 and n = 6, respectively), using the Wilcoxon Signed-Rank test. The overall intra-rater reliability, was classified as good to excellent for 65.1%, moderate for 27.0% and poor for 7.9% of the measures (47.6%, 76.2%, and 66.7% good-excellent; 28.6%, 23.8%, and 33.7% moderate; 23.8%, 0%, and 0% poor in TD, CP, and DMD, respectively), while ICC values for TD children were slightly lower for inter-rater intrasession reliability (38.1% good-excellent, 33.3% moderate and 26.6% poor) and for inter-rater intersession reliability (47.6% good-excellent, 23.8% moderate and 28.6% poor). Children with CP and DMD were significantly weaker than TD children (p < 0.001) and the majority of these strength differences exceeded the MDC. Children with CP significantly improved strength after training, with changes that exceeded the SEMs, whereas only limited strength decreases over time were observed in the DMD cohort. In conclusion, the investigated instrumented strength assessment was sufficiently reliable to confirm known-group validity for both cohorts and could detect the responsiveness of children with CP after a strength intervention. However, more research is necessary to determine the responsiveness of this assessment in children with DMD regarding their natural decline.
Collapse
Affiliation(s)
- Ineke Verreydt
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
| | | | - Elze Stoop
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
- Clinical Motion Analysis Laboratory, University Hospitals Leuven, Pellenberg, Belgium
| | - Nicky Peeters
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
- Department of Rehabilitation Sciences, Ghent University, Ghent, Belgium
| | | | - Patricia Van de Walle
- Research Group MOVANT, Department of Rehabilitation Sciences and Physiotherapy (REVAKI), University of Antwerp, Wilrijk, Belgium
- Multidisciplinary Motor Centre Antwerp (M2OCEAN), University of Antwerp, Antwerpen, Belgium
- Heder, Laboratory of Clinical Movement Analysis Antwerp, Antwerpen, Belgium
| | - Marleen Van den Hauwe
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Nathalie Goemans
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Liesbeth De Waele
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Anja Van Campenhout
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Pediatric Orthopedics, Department of Orthopedics, University Hospitals, Leuven, Leuven, Belgium
| | - Britta Hanssen
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
- Department of Rehabilitation Sciences, Ghent University, Ghent, Belgium
| | - Kaat Desloovere
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
- Clinical Motion Analysis Laboratory, University Hospitals Leuven, Pellenberg, Belgium
| |
Collapse
|
15
|
Billich N, Adams J, Carroll K, Truby H, Evans M, Ryan MM, Davidson ZE. The Relationship between Obesity and Clinical Outcomes in Young People with Duchenne Muscular Dystrophy. Nutrients 2022; 14:nu14163304. [PMID: 36014811 PMCID: PMC9412587 DOI: 10.3390/nu14163304] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Duchenne muscular dystrophy (DMD) is a severe X-linked neuromuscular disorder. Young people with DMD have high rates of obesity. There is emerging evidence that a higher BMI may negatively affect clinical outcomes in DMD. This study aimed to explore the relationship between obesity and clinical outcomes in DMD. Methods: This was a retrospective clinical audit of young people (two–21 years) with DMD. Height and weight were collected to calculate BMI z-scores to classify obesity, overweight and no overweight or obesity (reference category). Cox proportional hazards models determined the impact of obesity at five to nine years on clinical milestones including time to: loss of ambulation, timed function test cut-offs, obstructive sleep apnoea (OSA) diagnosis and first fracture. Results: 158 young people with DMD were included; most (89.9%) were steroid-treated. Mean follow-up was 8.7 ± 4.7 years. Obesity prevalence increased from age five (16.7%) to 11 years (50.6%). Boys with obesity at nine years sustained a fracture earlier (hazard ratio, HR: 2.050; 95% CI: 1.038–4.046). Boys with obesity at six to nine years were diagnosed with OSA earlier (e.g., obesity nine years HR: 2.883; 95% CI: 1.481–5.612). Obesity at eight years was associated with a 10 m walk/run in 7–10 s occurring at an older age (HR: 0.428; 95% CI: 0.207–0.887), but did not impact other physical function milestones. Conclusions: Although 50% of boys with DMD developed early obesity, the impact of obesity on physical function remains unclear. Obesity puts boys with DMD at risk of OSA and fractures at a younger age. Early weight management interventions are therefore important.
Collapse
Affiliation(s)
- Natassja Billich
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University Melbourne, Victoria 3168, Australia
- Neurology Department, The Royal Children’s Hospital Melbourne, Victoria 3052, Australia
- School of Human Movement and Nutrition Sciences, The University of Queensland Brisbane, Queensland 4072, Australia
| | - Justine Adams
- Neurology Department, The Royal Children’s Hospital Melbourne, Victoria 3052, Australia
- Murdoch Children’s Research Institute Melbourne, Victoria 3052, Australia
| | - Kate Carroll
- Neurology Department, The Royal Children’s Hospital Melbourne, Victoria 3052, Australia
- Murdoch Children’s Research Institute Melbourne, Victoria 3052, Australia
- Department of Physiotherapy, School of Primary and Allied Health Care, Faculty of Medicine, Nursing and Health Sciences, Monash University Melbourne, Victoria 3168, Australia
| | - Helen Truby
- School of Human Movement and Nutrition Sciences, The University of Queensland Brisbane, Queensland 4072, Australia
- School of Primary and Allied Health Care, Faculty of Medicine, Nursing and Health Sciences, Monash University Melbourne, Victoria 3168, Australia
| | - Maureen Evans
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University Melbourne, Victoria 3168, Australia
- Metabolic Medicine, The Royal Children’s Hospital Melbourne, Victoria 3052, Australia
| | - Monique M. Ryan
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University Melbourne, Victoria 3168, Australia
- Neurology Department, The Royal Children’s Hospital Melbourne, Victoria 3052, Australia
- Murdoch Children’s Research Institute Melbourne, Victoria 3052, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, Melbourne University, Victoria 3010, Australia
| | - Zoe E. Davidson
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University Melbourne, Victoria 3168, Australia
- Neurology Department, The Royal Children’s Hospital Melbourne, Victoria 3052, Australia
- Murdoch Children’s Research Institute Melbourne, Victoria 3052, Australia
- Correspondence:
| |
Collapse
|
16
|
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
|
17
|
Sherlock SP, Palmer J, Wagner KR, Abdel-Hamid HZ, Bertini E, Tian C, Mah JK, Kostera-Pruszczyk A, Muntoni F, Guglieri M, Brandsema JF, Mercuri E, Butterfield RJ, McDonald CM, Charnas L, Marraffino S. Quantitative magnetic resonance imaging measures as biomarkers of disease progression in boys with Duchenne muscular dystrophy: a phase 2 trial of domagrozumab. J Neurol 2022; 269:4421-4435. [PMID: 35396602 PMCID: PMC9294028 DOI: 10.1007/s00415-022-11084-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 01/14/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a progressive, neuromuscular disorder caused by mutations in the DMD gene that results in a lack of functional dystrophin protein. Herein, we report the use of quantitative magnetic resonance imaging (MRI) measures as biomarkers in the context of a multicenter phase 2, randomized, placebo-controlled clinical trial evaluating the myostatin inhibitor domagrozumab in ambulatory boys with DMD (n = 120 aged 6 to < 16 years). MRI scans of the thigh to measure muscle volume, muscle volume index (MVI), fat fraction, and T2 relaxation time were obtained at baseline and at weeks 17, 33, 49, and 97 as per protocol. These quantitative MRI measurements appeared to be sensitive and objective biomarkers for evaluating disease progression, with significant changes observed in muscle volume, MVI, and T2 mapping measures over time. To further explore the utility of quantitative MRI measures as biomarkers to inform longer term functional changes in this cohort, a regression analysis was performed and demonstrated that muscle volume, MVI, T2 mapping measures, and fat fraction assessment were significantly correlated with longer term changes in four-stair climb times and North Star Ambulatory Assessment functional scores. Finally, less favorable baseline measures of MVI, fat fraction of the muscle bundle, and fat fraction of lean muscle were significant risk factors for loss of ambulation over a 2-year monitoring period. These analyses suggest that MRI can be a valuable tool for use in clinical trials and may help inform future functional changes in DMD.Trial registration: ClinicalTrials.gov identifier, NCT02310763; registered December 2014.
Collapse
Affiliation(s)
| | | | - Kathryn R Wagner
- Kennedy Krieger Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hoda Z Abdel-Hamid
- Division of Child Neurology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Enrico Bertini
- Unit of Neuromuscular Disease, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Cuixia Tian
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Jean K Mah
- Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Michela Guglieri
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle, UK
| | | | - Eugenio Mercuri
- Pediatric Neurology, Catholic University, Rome, Italy
- Centro Nemo, Fondazione Policlinico Gemelli IRCCS, Rome, Italy
| | | | | | | | | |
Collapse
|
18
|
Deng J, Zhang J, Shi K, Liu Z. Drug development progress in duchenne muscular dystrophy. Front Pharmacol 2022; 13:950651. [PMID: 35935842 PMCID: PMC9353054 DOI: 10.3389/fphar.2022.950651] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/28/2022] [Indexed: 12/22/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe, progressive, and incurable X-linked disorder caused by mutations in the dystrophin gene. Patients with DMD have an absence of functional dystrophin protein, which results in chronic damage of muscle fibers during contraction, thus leading to deterioration of muscle quality and loss of muscle mass over time. Although there is currently no cure for DMD, improvements in treatment care and management could delay disease progression and improve quality of life, thereby prolonging life expectancy for these patients. Furthermore, active research efforts are ongoing to develop therapeutic strategies that target dystrophin deficiency, such as gene replacement therapies, exon skipping, and readthrough therapy, as well as strategies that target secondary pathology of DMD, such as novel anti-inflammatory compounds, myostatin inhibitors, and cardioprotective compounds. Furthermore, longitudinal modeling approaches have been used to characterize the progression of MRI and functional endpoints for predictive purposes to inform Go/No Go decisions in drug development. This review showcases approved drugs or drug candidates along their development paths and also provides information on primary endpoints and enrollment size of Ph2/3 and Ph3 trials in the DMD space.
Collapse
Affiliation(s)
- Jiexin Deng
- School of Nursing and Health, Henan University, Kaifeng, China
- *Correspondence: Jiexin Deng, ; Zhigang Liu,
| | - Junshi Zhang
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Keli Shi
- School of Medicine, Henan University, Kaifeng, China
| | - Zhigang Liu
- Department of Orthopedics, First Affiliated Hospital of Henan University, Kaifeng, China
- *Correspondence: Jiexin Deng, ; Zhigang Liu,
| |
Collapse
|
19
|
Bernardo Figueirêdo B, Reinaux C, Fuzari H, Sarmento A, Fernandes J, Dornelas de Andrade A. Chest wall volumes, diaphragmatic mobility, and functional capacity in patients with mucopolysaccharidoses. Disabil Rehabil 2022:1-10. [PMID: 35695376 DOI: 10.1080/09638288.2022.2084777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE We investigated respiratory muscle strength, diaphragm mobility, lung function, functional capacity, quality of life, body composition, breathing pattern, and chest wall (VT,CW) and compartmental volumes of Mucopolysaccharidosis (MPS) patients and compared these variables with matched healthy individuals. METHODS A cross-sectional study with data analyzed separately according to age group. A total of 68 individuals (34 MPS and 34 matched-healthy subjects) were included. Six-minute walking test assessed functional capacity and ultrasound assessed diaphragm mobility during quiet spontaneous breathing (QB). Optoelectronic plethysmography assessed VT,CW and breathing pattern during QB in two different positions: seated and supine (45° trunk inclination). RESULTS Body composition, lung function, respiratory muscle strength, and functional capacity were reduced in MPS (all p < 0.01). Diaphragm mobility was only reduced in adolescents (p = 0.01) and correlated with body composition and breathing pattern. Upper chest wall compartmental volumes were significantly lower in MPS, while abdominal volume only differed significantly in adolescents. Percentage contribution (%) of upper ribcage compartments to tidal volume was reduced in MPS children, whereas %AB was significantly increased compared with healthy subjects. CONCLUSION Lung function, respiratory muscle strength, functional capacity, diaphragm mobility, and quality of life are reduced in MPS compared with matched healthy subjects. VT,CW was mainly reduced due to pulmonary and abdominal ribcage impairment. Implications for RehabilitationReduction in respiratory muscle strength, functional capacity, diaphragm excursion and low lung volumes were found in individuals with Mucopolysaccharidoses (MPS).Chest wall volumes and the upper chest wall compartmental volumes during quiet spontaneous breathing are reduced in MPS.Assessment and monitoring of the respiratory system for individuals with MPS should be performed periodically through standardized assessments to enable identification of changes and early intervention by rehabilitation protocols.This study may provide the necessary basis for carrying out respiratoty rehabilitation protocols that can improving chest wall mechanics with breathing exercise in this group.
Collapse
Affiliation(s)
- Bárbara Bernardo Figueirêdo
- Department of Physical Therapy, Universidade Federal de Pernambuco, Recife, Brazil.,Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco, Recife, Brazil
| | - Cyda Reinaux
- Department of Physical Therapy, Universidade Federal de Pernambuco, Recife, Brazil
| | - Helen Fuzari
- Department of Physical Therapy, Universidade Federal de Pernambuco, Recife, Brazil
| | - António Sarmento
- Department of Physical Therapy, Universidade Federal de Pernambuco, Recife, Brazil
| | - Juliana Fernandes
- Department of Physical Therapy, Universidade Federal de Pernambuco, Recife, Brazil
| | - Armèle Dornelas de Andrade
- Department of Physical Therapy, Universidade Federal de Pernambuco, Recife, Brazil.,Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco, Recife, Brazil
| |
Collapse
|
20
|
Coratti G, Lenkowicz J, Patarnello S, Gullì C, Pera MC, Masciocchi C, Rinaldi R, Lovato V, Leone A, Cesario A, Mercuri E. Predictive models in SMA II natural history trajectories using machine learning: A proof of concept study. PLoS One 2022; 17:e0267930. [PMID: 35511762 PMCID: PMC9070873 DOI: 10.1371/journal.pone.0267930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/20/2022] [Indexed: 11/25/2022] Open
Abstract
It is known from previous literature that type II Spinal Muscular Atrophy (SMA) patients generally, after the age of 5 years, presents a steep deterioration until puberty followed by a relative stability, as most abilities have been lost. Although it is possible to identify points of slope indicating early improvement, steep decline and relative stabilizations, there is still a lot of variability within each age group and it's not always possible to predict individual trajectories of progression from age only. The aim of the study was to develop a predictive model based on machine learning using an XGBoost algorithm for regression and report, explore and quantify, in a single centre longitudinal natural history study, the influence of clinical variables on the 6/12-months Hammersmith Motor Functional Scale Expanded score prediction (HFMSE). This study represents the first approach to artificial intelligence and trained models for the prediction of individualized trajectories of HFMSE disease progression using individual characteristics of the patient. The application of this method to larger cohorts may allow to identify different classes of progression, a crucial information at the time of the new commercially available therapies.
Collapse
Affiliation(s)
- Giorgia Coratti
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Jacopo Lenkowicz
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Patarnello
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Consolato Gullì
- Department of Radiological and Hematological Sciences Fondazione, Policlinico Universitario A. Gemelli, IRCCS Università Cattolica del Sacro Cuore, Largo A. Gemelli, Rome, Italy
| | - Maria Carmela Pera
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Carlotta Masciocchi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Riccardo Rinaldi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Antonio Leone
- Department of Radiological and Hematological Sciences Fondazione, Policlinico Universitario A. Gemelli, IRCCS Università Cattolica del Sacro Cuore, Largo A. Gemelli, Rome, Italy
| | - Alfredo Cesario
- Open Innovation Manager, Scientific Directorate, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eugenio Mercuri
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
21
|
Reddy C, Patil AN, Suthar R, Sankhyan N, Sirari T, Kumar A, Bhattacharjee S, Saxena S, Saini AG, Sahu JK. Deflazacort dose optimization and safety evaluation in Duchenne muscular dystrophy (DOSE): A randomized, double-blind non-inferiority trial. Eur J Paediatr Neurol 2022; 38:77-84. [PMID: 35500465 DOI: 10.1016/j.ejpn.2022.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND US food and drug administration has recently approved deflazacort for Duchenne muscular dystrophy (DMD) and recommended the dosage of 0.9 mg/kg/d for patients aged ≥5years. However, data assessing the minimal efficacious dose and need of dose-titration based on age or disease severity is limited. OBJECTIVE To determine whether deflazacort 0.45 mg/kg/d (proposed lower dosage) is non-inferior to 0.9 mg/kg/d among newly diagnosed patients with DMD. METHOD A double-blinded, non-inferiority, randomized trial, conducted between December 2018 and July 2020. Newly diagnosed patient aged 5-15 years with genetic or muscle biopsy confirmed DMD and baseline 6-min walk distance (6MWD) > 150 m were screened. Patients were randomly assigned (1:1), stratified to prespecified subgroups by age (≤7years and >7years), and baseline 6MWD (≤350 m and >350 m), to receive either 0.45 mg/kg/d or 0.9 mg/kg/d regimens. The primary endpoint was the change in 6MWD, from baseline to week-24 of intervention. The trial was powered with a predefined, non-inferiority margin of 30 m. The analyses were by modified intention-to-treat (mITT). RESULT A total of 97 patients were enrolled, 40 receiving 0.45 mg/kg/d and 45 receiving 0.9 mg/kg/d deflazacort comprised of mITT population. For primary endpoint analysis the mean (SD) change in 6MWD from baseline to week-24 was 9.7 m (41.5) in deflazacort 0.45 mg/kg/d, and 34.7 m (43.5) for 0.9 mg/kg/d. The mean difference in change in 6MWD across the group was 24.8 m (95% CI 6.7 to 43, p value 0.008). The mean difference in change in 6MWD in the subgroups of boys ≤7 years of age was 21.8 m (95% CI -0.82, 44.5, p = 0.059), with baseline 6MWD of >350 m was 19.9 m (95% CI -2.4, 42.4; p = 0.08). The incidence of combined moderate to severe treatment-related adverse events was significant in the 0.9 mg/kg/d group by week 24 (odds ratio 0.36 [95% CI, 0.14 to 0.89], p = 0.03). DISCUSSION The efficacy of proposed low dose deflazacort in comparison to the standard dose did not meet the prespecified criteria for non-inferiority. The low dose deflazacort was non-inferior in subgroup of patients with age ≤7 years and baseline 6MWD of >350 m. TRIAL REGISTRATION Clinical Trial Registry-India Identifier: CTRI/2019/02/017388.
Collapse
Affiliation(s)
- Chaithanya Reddy
- Pediatric Neurology Unit, Department of Pediatrics, Advanced Pediatrics Centre, PGIMER, Chandigarh, 160012, India
| | - Amol N Patil
- Department of Pharmacology, PGIMER, Chandigarh, 160012, India
| | - Renu Suthar
- Pediatric Neurology Unit, Department of Pediatrics, Advanced Pediatrics Centre, PGIMER, Chandigarh, 160012, India.
| | - Naveen Sankhyan
- Pediatric Neurology Unit, Department of Pediatrics, Advanced Pediatrics Centre, PGIMER, Chandigarh, 160012, India
| | - Titiksha Sirari
- Pediatric Neurology Unit, Department of Pediatrics, Advanced Pediatrics Centre, PGIMER, Chandigarh, 160012, India
| | - Ankit Kumar
- Department of Pharmacology, PGIMER, Chandigarh, 160012, India
| | | | - Somya Saxena
- Department of Physical Medicine and Rehabilitation, PGIMER, Chandigarh, 160012, India
| | - Arushi G Saini
- Pediatric Neurology Unit, Department of Pediatrics, Advanced Pediatrics Centre, PGIMER, Chandigarh, 160012, India
| | - Jitendra K Sahu
- Pediatric Neurology Unit, Department of Pediatrics, Advanced Pediatrics Centre, PGIMER, Chandigarh, 160012, India; Department of Management, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
22
|
Ferizovic N, Summers J, de Zárate IBO, Werner C, Jiang J, Landfeldt E, Buesch K. Prognostic indicators of disease progression in Duchenne muscular dystrophy: A literature review and evidence synthesis. PLoS One 2022; 17:e0265879. [PMID: 35333888 PMCID: PMC8956179 DOI: 10.1371/journal.pone.0265879] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 03/09/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a rare, severely debilitating, and fatal neuromuscular disease characterized by progressive muscle degeneration. Like in many orphan diseases, randomized controlled trials are uncommon in DMD, resulting in the need to indirectly compare treatment effects, for example by pooling individual patient-level data from multiple sources. However, to derive reliable estimates, it is necessary to ensure that the samples considered are comparable with respect to factors significantly affecting the clinical progression of the disease. To help inform such analyses, the objective of this study was to review and synthesise published evidence of prognostic indicators of disease progression in DMD. We searched MEDLINE (via Ovid), Embase (via Ovid) and the Cochrane Library (via Wiley) for records published from inception up until April 23 2021, reporting evidence of prognostic indicators of disease progression in DMD. Risk of bias was established with the grading system of the Centre for Evidence-Based Medicine (CEBM). RESULTS Our search included 135 studies involving 25,610 patients from 18 countries across six continents (Africa, Asia, Australia, Europe, North America and South America). We identified a total of 23 prognostic indicators of disease progression in DMD, namely age at diagnosis, age at onset of symptoms, ataluren treatment, ATL1102, BMI, cardiac medication, DMD genetic modifiers, DMD mutation type, drisapersen, edasalonexent, eteplirsen, glucocorticoid exposure, height, idebenone, lower limb surgery, orthoses, oxandrolone, spinal surgery, TAS-205, vamorolone, vitlolarsen, ventilation support, and weight. Of these, cardiac medication, DMD genetic modifiers, DMD mutation type, and glucocorticoid exposure were designated core prognostic indicators, each supported by a high level of evidence and significantly affecting a wide range of clinical outcomes. CONCLUSION This study provides a current summary of prognostic indicators of disease progression in DMD, which will help inform the design of comparative analyses and future data collection initiatives in this patient population.
Collapse
Affiliation(s)
- Nermina Ferizovic
- MAP BioPharma Ltd, Cambridge, England, United Kingdom
- BresMed Health Solutions, Sheffield, England, United Kingdom
| | | | | | | | - Joel Jiang
- PTC Therapeutics, South Plainfield, New Jersey, United States of America
| | | | | |
Collapse
|
23
|
Krosschell KJ, Townsend EL, Kiefer M, Simeone SD, Zumpf K, Welty L, Swoboda KJ. Natural history of 10-meter walk/run test performance in spinal muscular atrophy: A longitudinal analysis. Neuromuscul Disord 2022; 32:125-134. [PMID: 35063329 PMCID: PMC8908436 DOI: 10.1016/j.nmd.2021.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/20/2021] [Accepted: 08/17/2021] [Indexed: 02/03/2023]
Abstract
As trials and treatments for spinal muscular atrophy (SMA) rapidly evolve, understanding the natural history and potential utility of the 10-meter walk/run test (10MWRT) in ambulant individuals is critical. Study aims were to: 1) establish change over time and across age for 10MWRT time in an untreated natural history cohort of young, ambulatory participants with SMA and 2) identify relations between 10MWRT time and age, SMA type, SMN2 copy number and anthropometrics. Untreated individuals (n = 56) age 2 to 21 years who were enrolled in a long-term natural history study between 2005 and 2014 and met inclusion criteria were included. Linear mixed effects models were used to assess changes in 10MWRT time with age and associations with SMA type, SMN2 copy number, and body mass. SMA type 3b (versus 3a), SMN2 copy number 4 (versus 3) and lower body mass were associated with faster 10MWRT. 10MWRT performance improved between 3 and 8 years of age, was stable between 9 and 10, and gradually declined from 11 to 18. Findings provide the first longitudinal natural history report of 10MWRT time in young individuals with SMA and offer a critical foundation for interpreting childhood change in short distance walking speed with pharmacologic treatment.
Collapse
Affiliation(s)
- Kristin J. Krosschell
- Department of Physical Therapy & Human Movement Sciences and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elise L. Townsend
- MGH Institute of Health Professions, Boston, MA, USA,Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Sarah D. Simeone
- Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Katelyn Zumpf
- Biostatistics Collaboration Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Leah Welty
- Biostatistics Collaboration Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kathryn J. Swoboda
- Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
24
|
Muntoni F, Signorovitch J, Sajeev G, Goemans N, Wong B, Tian C, Mercuri E, Done N, Wong H, Moss J, Yao Z, Ward SJ, Manzur A, Servais L, Niks EH, Straub V, de Groot IJM, McDonald C. Real-world and natural history data for drug evaluation in Duchenne muscular dystrophy: suitability of the North Star Ambulatory Assessment for comparisons with external controls. Neuromuscul Disord 2022; 32:271-283. [DOI: 10.1016/j.nmd.2022.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 10/19/2022]
|
25
|
Servais L, Yen K, Guridi M, Lukawy J, Vissière D, Strijbos P. Stride Velocity 95th Centile: Insights into Gaining Regulatory Qualification of the First Wearable-Derived Digital Endpoint for use in Duchenne Muscular Dystrophy Trials. J Neuromuscul Dis 2022; 9:335-346. [PMID: 34958044 PMCID: PMC9028650 DOI: 10.3233/jnd-210743] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In 2019, stride velocity 95th centile (SV95C) became the first wearable-derived digital clinical outcome assessment (COA) qualified by the European Medicines Agency (EMA) for use as a secondary endpoint in trials for Duchenne muscular dystrophy. SV95C was approved via the EMA's qualification pathway for novel methodologies for medicine development, which is a voluntary procedure for assessing the regulatory acceptability of innovative methods used in pharmaceutical research and development. SV95C is an objective, real-world digital ambulation measure of peak performance, representing the speed of the fastest strides taken by the wearer over a recording period of 180 hours. SV95C is correlated with traditional clinic-based assessments of motor function and has greater sensitivity to clinical change over 6 months than other wearable-derived stride variables, for example, median stride length or velocity. SV95C overcomes many limitations of episodic, clinic-based motor function testing, allowing the assessment of ambulation ability between clinic visits and under free-living conditions. Here we highlight considerations and challenges in developing SV95C using evidence generated by a high-performance wearable sensor. We also provide a commentary of the device's technical capabilities, which were a determining factor in the regulatory approval of SV95C. This article aims to provide insights into the methods employed, and the challenges faced, during the regulatory approval process for researchers developing new digital tools for patients with diseases that affect motor function.
Collapse
Affiliation(s)
- Laurent Servais
- Division of Child Neurology, Centre de Références des Maladies Neuromusculaires, Department of Pediatrics, University Hospital Liège and University of Liège, Liège, Belgium
- Muscular Dystrophy UK Oxford Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Karl Yen
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | | | | | | |
Collapse
|
26
|
Billich N, Evans M, Truby H, Ryan MM, Davidson ZE. The association between dietary factors and body weight and composition in boys with Duchenne muscular dystrophy. J Hum Nutr Diet 2021; 35:804-815. [PMID: 34936149 DOI: 10.1111/jhn.12987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/28/2021] [Accepted: 12/13/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a X-linked neuromuscular disorder. Boys with DMD have high rates of obesity, but little is known about dietary factors that may contribute to weight gain in this population. This study aimedto explore the relationship between dietary factors, body mass index (BMI) z-score, body composition and motor function and to describe dietary intake in boys with DMD. METHODOLOGY A cross-sectional analysis of 3-day food diaries from ambulant and steroid treated boys with DMD aged 5-13 years was conducted. Correlation analysis explored the relationship between dietary factors, BMI z-score, fat mass % (FM%) and lean mass (LM%). RESULTS Themedian agewas 8.5 years [interquartile range (IQR) 7.2, 10.5]. Median energy/kg/day in those within a healthy weight range (n=11) was316 kJ/kg/day [IQR 276, 355] and greater than estimated requirements; and forthose above a healthy weight (n=26) energy intake was 185kJ/kg/day [IQR 143, 214] and lower than estimated requirements. Energy/kg/day was negatively associatedwith BMI z-score (r=-0. 650) and FM% (r=-0.817)but positively associated with LM% (r=0.805, all analyses p =<0.01). Younger age was associated (r=-0.609 p=<0.01) with a higher energy/kg/day. For all participants vegetable, grains, meat/alternatives and dairy intake was sub-optimal. PRINCIPAL CONCLUSIONS Younger boys with DMD within a healthy weight range are overconsuming energy dense nutrient poor foods. A focus on improving diet quality during early childhood may prove a useful strategy to reduce excess weight gain and support healthier eating habits in this vulnerable clinical population. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Natassja Billich
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Australia.,The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Maureen Evans
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Australia.,The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Helen Truby
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia.,School of Primary and Allied Health Care, Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Australia
| | - Monique M Ryan
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Australia.,Department of Neurology, The Royal Children's Hospital, Melbourne, Victoria, Australia.,Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics,Faculty of Medicine, Dentistry and Health Sciences, Melbourne University, Victoria, Australia
| | - Zoe E Davidson
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Australia.,Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Neurology Department, The Royal Children's Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
27
|
Naarding KJ, Doorenweerd N, Koeks Z, Hendriksen RGF, Chotkan KA, Krom YD, de Groot IJM, Straathof CS, Niks EH, Kan HE. Decision-Making And Selection Bias in Four Observational Studies on Duchenne and Becker Muscular Dystrophy. J Neuromuscul Dis 2021; 7:433-442. [PMID: 32925089 PMCID: PMC7902964 DOI: 10.3233/jnd-200541] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Natural history data are essential for trial design in Duchenne (DMD) and Becker muscular dystrophy (BMD), but recruitment for observational studies can be challenging. OBJECTIVE We reviewed reasons why patients or caregivers declined participation, and compared characteristics of participants and non-participants to assess possible selection bias in four observational studies, three on DMD and one on BMD. METHODS Three pediatric DMD studies focused on cross-sectional cognitive function and brain MRI (DMDbrain, n = 35 and DMDperfusion, n = 12), and on longitudinal upper extremity function and muscle MRI (DMDarm, n = 22). One adult BMD study assessed longitudinal functioning (n = 36). Considerations for non-participation were retrospectively reviewed from screening logs. Age, travel-time, DMD gene mutations and age at loss of ambulation (DMDarm and BMD study only), of participants and non-participants were derived from the Dutch Dystrophinopathy Database and compared using nonparametric tests (p < 0.05). RESULTS The perceived burden of the protocol (38.2%), use of MRI (30.4%), and travel-time to the study site (19.1%) were the most frequently reported considerations for non-participation. Only few patients reported lack of personal gain (0.0- 5.9%). Overall, participating patients were representative for the studied sub-populations, except for a younger age of DMDarm study participants and a complete lack of participants with a mutation beyond exon 63. CONCLUSION Optimizing patient involvement in protocol design, improving MRI experiences, and integrating research into clinics are important factors to decrease burden and facilitate participation. Nationwide registries are essential to compare participants and non-participants and ensure representative observational research. Specific effort is needed to include patients with distal mutations in cognitive studies.
Collapse
Affiliation(s)
- Karin J Naarding
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center Netherlands
| | - Nathalie Doorenweerd
- C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands.,John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Zaïda Koeks
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Ruben G F Hendriksen
- C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands
| | - Kinita A Chotkan
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Yvonne D Krom
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center Netherlands
| | - Imelda J M de Groot
- Duchenne Center Netherlands.,Department of Rehabilitation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Chiara S Straathof
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center Netherlands
| | - Hermien E Kan
- Duchenne Center Netherlands.,C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands
| |
Collapse
|
28
|
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: 2] [Impact Index Per Article: 0.5] [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
|
29
|
Lake SL, Quintana MA, Broglio K, Panagoulias J, Berry SM, Panzara MA. Bayesian adaptive design for clinical trials in Duchenne muscular dystrophy. Stat Med 2021; 40:4167-4184. [PMID: 33960507 DOI: 10.1002/sim.9021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 01/13/2023]
Abstract
A Bayesian adaptive design is proposed for a clinical trial in Duchenne muscular dystrophy. The trial was designed to demonstrate treatment efficacy on an ambulatory-based clinical endpoint and to identify early success on a biomarker (dystrophin protein levels) that can serve as a basis for accelerated approval in the United States. The trial incorporates placebo augmentation using placebo data from past clinical trials. A thorough simulation study was conducted to understand the operating characteristics of the trial. This trial design was selected for the US FDA Complex Innovative Trial Design Pilot Meeting Program and the experience in that program is summarized.
Collapse
|
30
|
Brogna C, Coratti G, Rossi R, Neri M, Messina S, Amico AD, Bruno C, Lucibello S, Vita G, Berardinelli A, Magri F, Ricci F, Pedemonte M, Mongini T, Battini R, Bello L, Pegoraro E, Baranello G, Politano L, Comi GP, Sansone VA, Albamonte E, Donati A, Bertini E, Goemans N, Previtali S, Bovis F, Pane M, Ferlini A, Mercuri E. The nonsense mutation stop+4 model correlates with motor changes in Duchenne muscular dystrophy. Neuromuscul Disord 2021; 31:479-488. [PMID: 33773883 DOI: 10.1016/j.nmd.2021.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 10/22/2022]
Abstract
The aim was to assess 3-year longitudinal data using 6MWT in 26 ambulant boys affected by DMD carrying nonsense mutations and to compare their results to other small mutations. We also wished to establish, within the nonsense mutations group, patterns of change according to several variables. Patients with nonsense mutations were categorized according to the stop codon type newly created by the mutation and also including the adjacent 5' (upstream) and 3' (downstream) nucleotides. No significant difference was found between nonsense mutations and other small mutations (p > 0.05) on the 6MWT. Within the nonsense mutations group, there was no difference in 6MWT when the patients were subdivided according to: Type of stop codon, frame status of exons involved, protein domain affected. In contrast, there was a difference when the stop codon together with the 3' adjacent nucleotide ("stop+4 model") was considered (p < 0.05) with patients with stop codon TGA and 3' adjacent nucleotide G (TGAG) having a more rapid decline. Our finding suggest that the stop+4 model may help in predicting functional changes. This data will be useful at the time of interpreting the long term follow up of patients treated with Ataluren that are becoming increasingly available.
Collapse
Affiliation(s)
- Claudia Brogna
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy; Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Rome 00152, Italy
| | - Giorgia Coratti
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy; Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Rome 00152, Italy
| | - Rachele Rossi
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Marcella Neri
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Sonia Messina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy; Nemo SUD Clinical Center, University Hospital "G. Martino", Messina, Italy
| | - Adele D' Amico
- Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Claudio Bruno
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Simona Lucibello
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy; Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Rome 00152, Italy
| | - Gianluca Vita
- Nemo SUD Clinical Center, University Hospital "G. Martino", Messina, Italy
| | - Angela Berardinelli
- Child Neurology and Psychiatry Unit, ''Casimiro Mondino'' Foundation, Pavia, Italy
| | - Francesca Magri
- Department of Pathophysiology and Transplantation, Fondazione IRCCS Ca' Grande Ospedale Maggiore Policlinico, Dino Ferrari Center, , University of Milan, Milan, Italy
| | - Federica Ricci
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Torino, Italy
| | - Marina Pedemonte
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Tiziana Mongini
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of 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
- Cardiomiologia e Genetica Medica, Dipartimento di Medicina Sperimentale, Università della Campania Luigi Vanvitelli, Napoli, Italy
| | - Giacomo P Comi
- Department of Pathophysiology and Transplantation, Fondazione IRCCS Ca' Grande Ospedale Maggiore Policlinico, Dino Ferrari Center, , University of Milan, Milan, Italy
| | - Valeria A Sansone
- The NEMO Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, Milan, Italy
| | - Emilio Albamonte
- 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, IRCCS, Rome, Italy
| | - Nathalie Goemans
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Stefano Previtali
- Neuromuscular Repair Unit, Inspe and Division of Neuroscience, IRCSS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Bovis
- Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| | - Marika Pane
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy; Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Rome 00152, Italy
| | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Eugenio Mercuri
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy; Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Rome 00152, Italy.
| | | |
Collapse
|
31
|
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]
|
32
|
Coratti G, Messina S, Lucibello S, Pera MC, Montes J, Pasternak A, Bovis F, Exposito Escudero J, Mazzone ES, Mayhew A, Glanzman AM, Young SD, Salazar R, Duong T, Muni Lofra R, De Sanctis R, Carnicella S, Milev E, Civitello M, Pane M, Scoto M, Bettolo CM, Antonaci L, Frongia A, Sframeli M, Vita GL, D'Amico A, Van Den Hauwe M, Albamonte E, Goemans N, Darras BT, Bertini E, Sansone V, Day J, Nascimento Osorio A, Bruno C, Muntoni F, De Vivo DC, Finkel RS, Mercuri E. Clinical Variability in Spinal Muscular Atrophy Type
III. Ann Neurol 2020; 88:1109-1117. [DOI: 10.1002/ana.25900] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 09/01/2020] [Accepted: 09/06/2020] [Indexed: 01/02/2023]
Affiliation(s)
- Giorgia Coratti
- Pediatric Neurology Università Cattolica del Sacro Cuore Rome Italy
- Centro Clinico Nemo Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy
| | - Sonia Messina
- Department of Clinical and Experimental Medicine and Centro Clinico Nemo Sud University of Messina Messina Italy
| | - Simona Lucibello
- Pediatric Neurology Università Cattolica del Sacro Cuore Rome Italy
- Centro Clinico Nemo Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy
| | - Maria Carmela Pera
- Pediatric Neurology Università Cattolica del Sacro Cuore Rome Italy
- Centro Clinico Nemo Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy
| | - Jacqueline Montes
- Departments of Rehabilitation and Regenerative Medicine and Neurology Columbia University Irving Medical Center New York NY
| | - Amy Pasternak
- Departments of Neurology Boston Children's Hospital, Harvard Medical School Boston MA
| | - Francesca Bovis
- Department of Health Sciences (DISSAL) University of Genova Genoa Italy
| | - Jessica Exposito Escudero
- Neuromuscular Unit, Neuropaediatrics Department Institut de Recerca Hospital Universitari Sant Joan de Déu, ISCIII, CIBERER Barcelona Spain
| | | | - Anna Mayhew
- The John Walton Muscular Dystrophy Research Centre Newcastle University and Newcastle Hospitals NHS Foundation Trust Newcastle upon Tyne UK
| | - Allan M. Glanzman
- Department of Physical Therapy Children's Hospital of Philadelphia Philadelphia PA
| | | | - Rachel Salazar
- Department of Clinical and Experimental Medicine and Centro Clinico Nemo Sud University of Messina Messina Italy
| | - Tina Duong
- Department of Neurology Stanford University Stanford CA
| | - Robert Muni Lofra
- The John Walton Muscular Dystrophy Research Centre Newcastle University and Newcastle Hospitals NHS Foundation Trust Newcastle upon Tyne UK
| | - Roberto De Sanctis
- Centro Clinico Nemo Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy
| | - Sara Carnicella
- Centro Clinico Nemo Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy
| | - Evelin Milev
- Dubowitz Neuromuscular Centre UCL Institute of Child Health & Great Ormond Street Hospital London UK
| | - Matthew Civitello
- Center for Experimental Neurotherapeutics St. Jude Children's Research Hospital Memphis TN
| | - Marika Pane
- Centro Clinico Nemo Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy
| | - Mariacristina Scoto
- Dubowitz Neuromuscular Centre UCL Institute of Child Health & Great Ormond Street Hospital London UK
| | - Chiara Marini Bettolo
- The John Walton Muscular Dystrophy Research Centre Newcastle University and Newcastle Hospitals NHS Foundation Trust Newcastle upon Tyne UK
| | - Laura Antonaci
- Pediatric Neurology Università Cattolica del Sacro Cuore Rome Italy
- Centro Clinico Nemo Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy
| | - Annalia Frongia
- Pediatric Neurology Università Cattolica del Sacro Cuore Rome Italy
- Centro Clinico Nemo Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy
| | - Maria Sframeli
- Department of Clinical and Experimental Medicine and Centro Clinico Nemo Sud University of Messina Messina Italy
| | - Gian Luca Vita
- Department of Clinical and Experimental Medicine and Centro Clinico Nemo Sud University of Messina Messina Italy
| | - Adele D'Amico
- Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences IRCCS Bambino Gesù Children's Hospital Rome Italy
| | | | - Emilio Albamonte
- Neurorehabilitation Unit University of Milan, Neuromuscular Omnicentre Clinical Center, Niguarda Hospital Milan Italy
| | - Nathalie Goemans
- Department of Child Neurology University Hospitals Leuven Leuven Belgium
| | - Basil T. Darras
- Departments of Neurology Boston Children's Hospital, Harvard Medical School Boston MA
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences IRCCS Bambino Gesù Children's Hospital Rome Italy
| | - Valeria Sansone
- Neurorehabilitation Unit University of Milan, Neuromuscular Omnicentre Clinical Center, Niguarda Hospital Milan Italy
| | - John Day
- Department of Neurology Stanford University Stanford CA
| | - Andres Nascimento Osorio
- Neuromuscular Unit, Neuropaediatrics Department Institut de Recerca Hospital Universitari Sant Joan de Déu, ISCIII, CIBERER Barcelona Spain
| | - Claudio Bruno
- Center of Experimental and Translational Myology IRCCS Istituto Giannina Gaslini Genoa Italy
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre UCL Institute of Child Health & Great Ormond Street Hospital London UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre London UK
| | - Darryl C. De Vivo
- Department of Clinical and Experimental Medicine and Centro Clinico Nemo Sud University of Messina Messina Italy
| | - Richard S. Finkel
- Center for Experimental Neurotherapeutics St. Jude Children's Research Hospital Memphis TN
| | - Eugenio Mercuri
- Pediatric Neurology Università Cattolica del Sacro Cuore Rome Italy
- Centro Clinico Nemo Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy
| |
Collapse
|
33
|
Campbell C, Barohn RJ, Bertini E, Chabrol B, Comi GP, Darras BT, Finkel RS, Flanigan KM, Goemans N, Iannaccone ST, Jones KJ, Kirschner J, Mah JK, Mathews KD, McDonald CM, Mercuri E, Nevo Y, Péréon Y, Renfroe JB, Ryan MM, Sampson JB, Schara U, Sejersen T, Selby K, Tulinius M, Vílchez JJ, Voit T, Wei LJ, Wong BL, Elfring G, Souza M, McIntosh J, Trifillis P, Peltz SW, Muntoni F. Meta-analyses of ataluren randomized controlled trials in nonsense mutation Duchenne muscular dystrophy. J Comp Eff Res 2020; 9:973-984. [DOI: 10.2217/cer-2020-0095] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Assess the totality of efficacy evidence for ataluren in patients with nonsense mutation Duchenne muscular dystrophy (nmDMD). Materials & methods: Data from the two completed randomized controlled trials (ClinicalTrials.gov: NCT00592553; NCT01826487) of ataluren in nmDMD were combined to examine the intent-to-treat (ITT) populations and two patient subgroups (baseline 6-min walk distance [6MWD] ≥300–<400 or <400 m). Meta-analyses examined 6MWD change from baseline to week 48. Results: Statistically significant differences in 6MWD change with ataluren versus placebo were observed across all three meta-analyses. Least-squares mean difference (95% CI): ITT (n = 342), +17.2 (0.2–34.1) m, p = 0.0473; ≥300–<400 m (n = 143), +43.9 (18.2–69.6) m, p = 0.0008; <400 m (n = 216), +27.7 (6.4–49.0) m, p = 0.0109. Conclusion: These meta-analyses support previous evidence for ataluren in slowing disease progression versus placebo in patients with nmDMD over 48 weeks. Treatment benefit was most evident in patients with a baseline 6MWD ≥300–<400 m (the ambulatory transition phase), thereby informing future trial design.
Collapse
Affiliation(s)
- Craig Campbell
- Schulich School of Medicine & Dentistry, Western University, London, ON, N6A 5C1, Canada
| | | | - Enrico Bertini
- Bambino Gesù Children’s Research Hospital, Rome, 00146, Italy
| | - Brigitte Chabrol
- Hôpital de la Timone, Unité de Médecine Infantile, Marseille, 13385, France
| | - Giacomo Pietro Comi
- IRCCS Fondazione Ca'Granda Ospedale Maggiore Policlinico, Dino Ferrari Centre, Department of Pathophysiology & Transplantation, University of Milan, Milan, 20122, Italy
| | - Basil T Darras
- Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Richard S Finkel
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | | | | | | | - Kristi J Jones
- Kids Neuroscience, The Children’s Hospital at Westmead, Westmead, NSW, 2145, Australia
| | - Janbernd Kirschner
- Department of Neuropediatrics & Muscle Disorders, Medical Center, University of Freiburg, Freiburg 79110, Germany
| | - Jean K Mah
- Department of Pediatrics, Division of Pediatric Neurology, Alberta Children’s Hospital, University of Calgary, Calgary, AB T3B 6A8, Canada
| | - Katherine D Mathews
- Departments of Pediatrics & Neurology, Division of Pediatric Neurology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Craig M McDonald
- University of California Davis Health, Sacramento, CA 95817, USA
| | - Eugenio Mercuri
- Department of Pediatric Neurology, Catholic University, Rome, 00168, Italy
| | - Yoram Nevo
- Schneider Children’s Medical Center, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Yann Péréon
- Reference Centre for Neuromuscular Disorders AOC, Hôtel-Dieu, Nantes, 44000, France
| | - J Ben Renfroe
- Child Neurology Center of Northwest Florida, Gulf Breeze, FL 32561, USA
| | - Monique M Ryan
- The Royal Children’s Hospital, Parkville, Victoria, 3052, Australia
| | - Jacinda B Sampson
- Stanford University Medical Center, Department of Neurology & Neurological Sciences, Stanford, CA 94305, USA
| | - Ulrike Schara
- Department of Pediatric Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, 45122, Germany
| | - Thomas Sejersen
- Karolinska University Hospital, Karolinska Institutet, Stockholm, 171 76, Sweden
| | - Kathryn Selby
- Division of Neurology, British Columbia Children’s Hospital, Vancouver, BC, V6H 3N1, Canada
| | - Már Tulinius
- Gothenburg University, Queen Silvia Children’s Hospital, Gothenburg, 416 85, Sweden
| | - Juan J Vílchez
- Hospital Universitario y Politécnico La Fe, CIBERER, Valencia, 46026, Spain
| | - Thomas Voit
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London & UCL Great Ormond Street Institute of Child Health, Great Ormond Street Hospital Trust, London, WC1N 1EH, UK
| | - Lee-Jen Wei
- Harvard TH Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - Brenda L Wong
- University of Massachusetts Medical School, UMass, Worcester, MA 01655, USA
| | - Gary Elfring
- PTC Therapeutics Inc., South Plainfield, NJ 07080, USA
| | - Marcio Souza
- PTC Therapeutics Inc., South Plainfield, NJ 07080, USA
| | | | | | | | - Francesco Muntoni
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London & UCL Great Ormond Street Institute of Child Health, Great Ormond Street Hospital Trust, London, WC1N 1EH, UK
| | | | | | | |
Collapse
|
34
|
Coratti G, Pera MC, Lucibello S, Montes J, Pasternak A, Mayhew A, Glanzman AM, Young SD, Pane M, Scoto M, Messina S, Goemans N, Osorio AN, Pedemonte M, Sansone V, Bertini E, De Vivo DC, Finkel R, Muntoni F, Mercuri E. Age and baseline values predict 12 and 24-month functional changes in type 2 SMA. Neuromuscul Disord 2020; 30:756-764. [PMID: 32900576 DOI: 10.1016/j.nmd.2020.07.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 11/27/2022]
Abstract
The aim of this retrospective study was to establish the range of functional changes at 12 and 24-month in 267 type 2 Spinal Muscular Atrophy (SMA) patients with multiple assessments. We included 652 Hammersmith Functional Motor Scale Expanded (HFMSE) assessments at 12 month- and 305 at 24 month- intervals. The cohort was subdivided by functional level, Survival of Motor Neuron copy number and age. Stable scores (± 2 points) were found in 68% at 12 months and in 55% at 24 months. A decrease ≥2 points was found in 21% at 12 months and in 35% at 24 months. An increase ≥2 points was found in 11% at 12 months and 9.5% at 24 months. The risk of losing ≥2 points increased with age and HFMSE score at baseline both at 12 and 24-month. For each additional HFMSE point at baseline, the relative risk of a >2 point decline at 12 months increases by 5% before age 5 years (p = 0.023), by 8% between 5 and 13 (p<0.001) and by 26% after 13 years (p = 0.003). The combination of age and HFMSE scores at baseline increased the ability to predict progression in type 2 SMA.
Collapse
Affiliation(s)
- Giorgia Coratti
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy; Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria C Pera
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy; Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Simona Lucibello
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy; Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Jacqueline Montes
- Departments of Neurology and Pediatrics, Columbia University Irving Medical Center, New York, United States; Departments of Rehabilitation and Regenerative Medicine and Neurology, Columbia University Irving Medical Center, New York, United States
| | - Amy Pasternak
- Departments of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Anna Mayhew
- The John Walton Muscular Dystrophy Research Centre, Newcastle University, Integrated Laboratory Medicine Directorate, Institute of Genetic Medicine, Newcastle Upon Tyne NHS Foundation Trust, Newcastle Upon Tyne, United Kingdom
| | - Allan M Glanzman
- Department of Physical Therapy, The Children's Hospital of Philadelphia, Philadelphia
| | | | - Marika Pane
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Mariacristina Scoto
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health & Great Ormond Street Hospital, London
| | - Sonia Messina
- Department of Clinical and Experimental Medicine and Centro Clinico Nemo Sud, University of Messina, Messina, Italy
| | - Nathalie Goemans
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Andres Nascimiento Osorio
- Neuromuscular Unit, Neuropaediatrics Department, Institut de Recerca Hospital Universitari Sant Joan de Deu, Barcelona, Spain
| | - Marina Pedemonte
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Valeria Sansone
- Neurorehabilitation Unit, University of Milan, The NEMO Clinical Center in Milan, Italy
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences, IRCCS Bambino GesùChildren's Hospital, Rome, Italy
| | - Darryl C De Vivo
- Departments of Neurology and Pediatrics, Columbia University Irving Medical Center, New York, United States
| | - Richard Finkel
- Nemours Children's Hospital, University of Central Florida College of Medicine, Orlando, United States
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health & Great Ormond Street Hospital, London; NIHR Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom
| | - Eugenio Mercuri
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy; Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
| | | |
Collapse
|
35
|
Goemans N, Signorovitch J, Sajeev G, Yao Z, Gordish-Dressman H, McDonald CM, Vandenborne K, Miller D, Ward SJ, Mercuri E. Suitability of external controls for drug evaluation in Duchenne muscular dystrophy. Neurology 2020; 95:e1381-e1391. [PMID: 32611643 PMCID: PMC7538219 DOI: 10.1212/wnl.0000000000010170] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 03/06/2020] [Indexed: 12/19/2022] Open
Abstract
Objective To evaluate the suitability of real-world data (RWD) and natural history data (NHD) for use as external controls in drug evaluations for ambulatory Duchenne muscular dystrophy (DMD). Methods The consistency of changes in the 6-minute walk distance (Δ6MWD) was assessed across multiple clinical trial placebo arms and sources of NHD/RWD. Six placebo arms reporting 48-week Δ6MWD were identified via literature review and represented 4 sets of inclusion/exclusion criteria (n = 383 patients in total). Five sources of RWD/NHD were contributed by Universitaire Ziekenhuizen Leuven, DMD Italian Group, The Cooperative International Neuromuscular Research Group, ImagingDMD, and the PRO-DMD-01 study (n = 430 patients, in total). Mean Δ6MWD was compared between each placebo arm and RWD/NHD source after subjecting the latter to the inclusion/exclusion criteria of the trial for baseline age, ambulatory function, and steroid use. Baseline covariate adjustment was investigated in a subset of patients with available data. Results Analyses included ∼1,200 patient-years of follow-up. Differences in mean Δ6MWD between trial placebo arms and RWD/NHD cohorts ranged from −19.4 m (i.e., better outcomes in RWD/NHD) to 19.5 m (i.e., worse outcomes in RWD/NHD) and were not statistically significant before or after covariate adjustment. Conclusions We found that Δ6MWD was consistent between placebo arms and RWD/NHD subjected to equivalent inclusion/exclusion criteria. No evidence for systematic bias was detected. These findings are encouraging for the use of RWD/NHD to augment, or possibly replace, placebo controls in DMD trials. Multi-institution collaboration through the Collaborative Trajectory Analysis Project rendered this study feasible.
Collapse
Affiliation(s)
- Nathalie Goemans
- From the University Hospitals Leuven (N.G.), Child Neurology, Belgium; Analysis Group, Inc (J.S., G.S., Z.Y.), Boston; Collaborative Trajectory Analysis Project (J.S., S.J.W.), Cambridge, MA; Children's National Medical Center (H.G.-D.), Research Center for Genetic Medicine, Washington, DC; Department of Physical Medicine and Rehabilitation and Pediatrics (C.M.M.), University of California, Davis, Sacramento; Department of Physical Therapy (K.V.), University of Florida, Gainesville; CureDuchenne (D.M.), Newport Beach, CA; and Department of Pediatric Neurology (E.M.), Fondazione Policlinico Gemelli IRCCS, Catholic University, Rome, Italy
| | - James Signorovitch
- From the University Hospitals Leuven (N.G.), Child Neurology, Belgium; Analysis Group, Inc (J.S., G.S., Z.Y.), Boston; Collaborative Trajectory Analysis Project (J.S., S.J.W.), Cambridge, MA; Children's National Medical Center (H.G.-D.), Research Center for Genetic Medicine, Washington, DC; Department of Physical Medicine and Rehabilitation and Pediatrics (C.M.M.), University of California, Davis, Sacramento; Department of Physical Therapy (K.V.), University of Florida, Gainesville; CureDuchenne (D.M.), Newport Beach, CA; and Department of Pediatric Neurology (E.M.), Fondazione Policlinico Gemelli IRCCS, Catholic University, Rome, Italy
| | - Gautam Sajeev
- From the University Hospitals Leuven (N.G.), Child Neurology, Belgium; Analysis Group, Inc (J.S., G.S., Z.Y.), Boston; Collaborative Trajectory Analysis Project (J.S., S.J.W.), Cambridge, MA; Children's National Medical Center (H.G.-D.), Research Center for Genetic Medicine, Washington, DC; Department of Physical Medicine and Rehabilitation and Pediatrics (C.M.M.), University of California, Davis, Sacramento; Department of Physical Therapy (K.V.), University of Florida, Gainesville; CureDuchenne (D.M.), Newport Beach, CA; and Department of Pediatric Neurology (E.M.), Fondazione Policlinico Gemelli IRCCS, Catholic University, Rome, Italy
| | - Zhiwen Yao
- From the University Hospitals Leuven (N.G.), Child Neurology, Belgium; Analysis Group, Inc (J.S., G.S., Z.Y.), Boston; Collaborative Trajectory Analysis Project (J.S., S.J.W.), Cambridge, MA; Children's National Medical Center (H.G.-D.), Research Center for Genetic Medicine, Washington, DC; Department of Physical Medicine and Rehabilitation and Pediatrics (C.M.M.), University of California, Davis, Sacramento; Department of Physical Therapy (K.V.), University of Florida, Gainesville; CureDuchenne (D.M.), Newport Beach, CA; and Department of Pediatric Neurology (E.M.), Fondazione Policlinico Gemelli IRCCS, Catholic University, Rome, Italy
| | - Heather Gordish-Dressman
- From the University Hospitals Leuven (N.G.), Child Neurology, Belgium; Analysis Group, Inc (J.S., G.S., Z.Y.), Boston; Collaborative Trajectory Analysis Project (J.S., S.J.W.), Cambridge, MA; Children's National Medical Center (H.G.-D.), Research Center for Genetic Medicine, Washington, DC; Department of Physical Medicine and Rehabilitation and Pediatrics (C.M.M.), University of California, Davis, Sacramento; Department of Physical Therapy (K.V.), University of Florida, Gainesville; CureDuchenne (D.M.), Newport Beach, CA; and Department of Pediatric Neurology (E.M.), Fondazione Policlinico Gemelli IRCCS, Catholic University, Rome, Italy
| | - Craig M McDonald
- From the University Hospitals Leuven (N.G.), Child Neurology, Belgium; Analysis Group, Inc (J.S., G.S., Z.Y.), Boston; Collaborative Trajectory Analysis Project (J.S., S.J.W.), Cambridge, MA; Children's National Medical Center (H.G.-D.), Research Center for Genetic Medicine, Washington, DC; Department of Physical Medicine and Rehabilitation and Pediatrics (C.M.M.), University of California, Davis, Sacramento; Department of Physical Therapy (K.V.), University of Florida, Gainesville; CureDuchenne (D.M.), Newport Beach, CA; and Department of Pediatric Neurology (E.M.), Fondazione Policlinico Gemelli IRCCS, Catholic University, Rome, Italy
| | - Krista Vandenborne
- From the University Hospitals Leuven (N.G.), Child Neurology, Belgium; Analysis Group, Inc (J.S., G.S., Z.Y.), Boston; Collaborative Trajectory Analysis Project (J.S., S.J.W.), Cambridge, MA; Children's National Medical Center (H.G.-D.), Research Center for Genetic Medicine, Washington, DC; Department of Physical Medicine and Rehabilitation and Pediatrics (C.M.M.), University of California, Davis, Sacramento; Department of Physical Therapy (K.V.), University of Florida, Gainesville; CureDuchenne (D.M.), Newport Beach, CA; and Department of Pediatric Neurology (E.M.), Fondazione Policlinico Gemelli IRCCS, Catholic University, Rome, Italy
| | - Debra Miller
- From the University Hospitals Leuven (N.G.), Child Neurology, Belgium; Analysis Group, Inc (J.S., G.S., Z.Y.), Boston; Collaborative Trajectory Analysis Project (J.S., S.J.W.), Cambridge, MA; Children's National Medical Center (H.G.-D.), Research Center for Genetic Medicine, Washington, DC; Department of Physical Medicine and Rehabilitation and Pediatrics (C.M.M.), University of California, Davis, Sacramento; Department of Physical Therapy (K.V.), University of Florida, Gainesville; CureDuchenne (D.M.), Newport Beach, CA; and Department of Pediatric Neurology (E.M.), Fondazione Policlinico Gemelli IRCCS, Catholic University, Rome, Italy
| | - Susan J Ward
- From the University Hospitals Leuven (N.G.), Child Neurology, Belgium; Analysis Group, Inc (J.S., G.S., Z.Y.), Boston; Collaborative Trajectory Analysis Project (J.S., S.J.W.), Cambridge, MA; Children's National Medical Center (H.G.-D.), Research Center for Genetic Medicine, Washington, DC; Department of Physical Medicine and Rehabilitation and Pediatrics (C.M.M.), University of California, Davis, Sacramento; Department of Physical Therapy (K.V.), University of Florida, Gainesville; CureDuchenne (D.M.), Newport Beach, CA; and Department of Pediatric Neurology (E.M.), Fondazione Policlinico Gemelli IRCCS, Catholic University, Rome, Italy
| | - Eugenio Mercuri
- From the University Hospitals Leuven (N.G.), Child Neurology, Belgium; Analysis Group, Inc (J.S., G.S., Z.Y.), Boston; Collaborative Trajectory Analysis Project (J.S., S.J.W.), Cambridge, MA; Children's National Medical Center (H.G.-D.), Research Center for Genetic Medicine, Washington, DC; Department of Physical Medicine and Rehabilitation and Pediatrics (C.M.M.), University of California, Davis, Sacramento; Department of Physical Therapy (K.V.), University of Florida, Gainesville; CureDuchenne (D.M.), Newport Beach, CA; and Department of Pediatric Neurology (E.M.), Fondazione Policlinico Gemelli IRCCS, Catholic University, Rome, Italy.
| | | |
Collapse
|
36
|
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
|
37
|
Naarding KJ, Reyngoudt H, van Zwet EW, Hooijmans MT, Tian C, Rybalsky I, Shellenbarger KC, Le Louër J, Wong BL, Carlier PG, Kan HE, Niks EH. MRI vastus lateralis fat fraction predicts loss of ambulation in Duchenne muscular dystrophy. Neurology 2020; 94:e1386-e1394. [PMID: 31937624 PMCID: PMC7274919 DOI: 10.1212/wnl.0000000000008939] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 10/08/2019] [Indexed: 02/04/2023] Open
Abstract
Objective We studied the potential of quantitative MRI (qMRI) as a surrogate endpoint in Duchenne muscular dystrophy by assessing the additive predictive value of vastus lateralis (VL) fat fraction (FF) to age on loss of ambulation (LoA). Methods VL FFs were determined on longitudinal Dixon MRI scans from 2 natural history studies in Leiden University Medical Center (LUMC) and Cincinnati Children's Hospital Medical Center (CCHMC). CCHMC included ambulant patients, while LUMC included a mixed ambulant and nonambulant population. We fitted longitudinal VL FF values to a sigmoidal curve using a mixed model with random slope to predict individual trajectories. The additive value of VL FF over age to predict LoA was calculated from a Cox model, yielding a hazard ratio. Results Eighty-nine MRIs of 19 LUMC and 15 CCHMC patients were included. At similar age, 6-minute walking test distances were smaller and VL FFs were correspondingly higher in LUMC compared to CCHMC patients. Hazard ratio of a percent-point increase in VL FF for the time to LoA was 1.15 for LUMC (95% confidence interval [CI] 1.05–1.26; p = 0.003) and 0.96 for CCHMC (95% CI 0.84–1.10; p = 0.569). Conclusions The hazard ratio of 1.15 corresponds to a 4.11-fold increase of the instantaneous risk of LoA in patients with a 10% higher VL FF at any age. Although results should be confirmed in a larger cohort with prospective determination of the clinical endpoint, this added predictive value of VL FF to age on LoA supports the use of qMRI FF as an endpoint or stratification tool in clinical trials.
Collapse
Affiliation(s)
- Karin J Naarding
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH.
| | - Harmen Reyngoudt
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Erik W van Zwet
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Melissa T Hooijmans
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Cuixia Tian
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Irina Rybalsky
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Karen C Shellenbarger
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Julien Le Louër
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Brenda L Wong
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Pierre G Carlier
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Hermien E Kan
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Erik H Niks
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| |
Collapse
|
38
|
Chrzanowski SM, Darras BT, Rutkove SB. The Value of Imaging and Composition-Based Biomarkers in Duchenne Muscular Dystrophy Clinical Trials. Neurotherapeutics 2020; 17:142-152. [PMID: 31879850 PMCID: PMC7007477 DOI: 10.1007/s13311-019-00825-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
As the drug development pipeline for Duchenne muscular dystrophy (DMD) rapidly advances, clinical trial outcomes need to be optimized. Effective assessment of disease burden, natural history progression, and response to therapy in clinical trials for Duchenne muscular dystrophy are critical factors for clinical trial success. By choosing optimal biomarkers to better assess therapeutic efficacy, study costs and sample size requirements can be reduced. Currently, functional measures continue to serve as the primary outcome for the majority of DMD clinical trials. Quantitative measures of muscle health, including magnetic resonance imaging and spectroscopy, electrical impedance myography, and ultrasound, sensitively identify diseased muscle, disease progression, and response to a therapeutic intervention. Furthermore, such non-invasive techniques have the potential to identify disease pathology prior to onset of clinical symptoms. Despite robust supportive evidence, non-invasive quantitative techniques are still not frequently utilized in clinical trials for Duchenne muscular dystrophy. Non-invasive quantitative techniques have demonstrated the ability to quantify disease progression and potential response to therapeutic intervention, and should be used as a supplement to current standard functional measures. Such methods have the potential to significantly accelerate the development and approval of therapies for DMD.
Collapse
Affiliation(s)
- Stephen M Chrzanowski
- Department of Medicine, Boston Children's Hospital, 300 Longwood Ave., Boston, MA, 02115, USA.
| | - Basil T Darras
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Seward B Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
39
|
van der Meulen MH, Boer SD, du Marchie Sarvaas GJ, Blom NA, ten Harkel ADJ, Breur HMPJ, Rammeloo LAJ, Tanke R, Helbing WA, Boersma E, Dalinghaus M. Does Repeated Measurement of a 6-Min Walk Test Contribute to Risk Prediction in Children with Dilated Cardiomyopathy? Pediatr Cardiol 2020; 41:223-229. [PMID: 31713652 PMCID: PMC7072046 DOI: 10.1007/s00246-019-02244-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/31/2019] [Indexed: 11/26/2022]
Abstract
A single 6-min walk test (6MWT) can be used to identify children with dilated cardiomyopathy (DCM) with a high risk of death or heart transplantation. To determine if repeated 6MWT has added value in addition to a single 6MWT in predicting death or heart transplantation in children with DCM. Prospective multicenter cohort study including ambulatory DCM patients ≥ 6 years. A 6MWT was performed 1 to 4 times per year. The distance walked was expressed as percentage of predicted (6MWD%). We compared the temporal evolution of 6MWD% in patients with and without the study endpoint (SE: all-cause death or heart transplantation), using a linear mixed effects model. In 57 patients, we obtained a median of 4 (IQR 2-6) 6MWTs per patient during a median of 3.0 years of observation (IQR 1.5-5.1). Fourteen patients reached a SE (3 deaths, 11 heart transplantations). At any time during follow-up, the average estimate of 6MWD% was significantly lower in patients with a SE compared to patients without a SE. In both patients groups, 6MWD% remained constant over time. An absolute 1% lower 6MWD% was associated with an 11% higher risk (hazard) of the SE (HR 0.90, 95% CI 0.86-0.95 p < 0.001). Children with DCM who died or underwent heart transplantation had systematically reduced 6MWD%. The performance of all patients was stable over time, so repeated measurement of 6MWT within this time frame had little added value over a single test.
Collapse
Affiliation(s)
- Marijke H. van der Meulen
- Department of Pediatric Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Susanna den Boer
- Department of Pediatric Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Gideon J. du Marchie Sarvaas
- Department of Pediatric Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nico A. Blom
- Department of Pediatric Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Pediatric Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Arend D. J. ten Harkel
- Department of Pediatric Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans M. P. J. Breur
- Department of Pediatric Cardiology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lukas A. J. Rammeloo
- Department of Pediatric Cardiology, Free University Medical Center, Amsterdam, The Netherlands
| | - Ronald Tanke
- Department of Pediatric Cardiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Willem A. Helbing
- Department of Pediatric Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pediatric Cardiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Eric Boersma
- Department of Biostatistics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Michiel Dalinghaus
- Department of Pediatric Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pediatric Cardiology, Erasmus University Medical Center, Dr. Molewaterplein 60, P.O. Box 2060, 3000 CB Rotterdam, The Netherlands
| |
Collapse
|
40
|
Lima AARD, Cordeiro L. Fisioterapia aquática em indivíduos com distrofia muscular: uma revisão sistemática do tipo escopo. FISIOTERAPIA E PESQUISA 2020. [DOI: 10.1590/1809-2950/18031327012020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
RESUMO O objetivo deste estudo foi mapear o uso da fisioterapia aquática em indivíduos com distrofias musculares, de forma a caracterizar as intervenções no meio aquático e identificar componentes mensurados (variáveis estudadas e instrumentos utilizados nos estudos). A revisão sistemática do tipo de escopo incluiu estudos experimentais, descritivos e observacionais (em inglês, português e espanhol). As buscas foram realizadas nas plataformas Medline (PubMed), CINAHL, Embase, PEDro, Lilacs, ERIC, Scopus, Web of Science e Google Scholar. Os dados extraídos foram alocados em três categorias: (1) caracterização dos registros, (2) informações referentes a fisioterapia aquática e (3) componentes mensurados. Foram encontrados 556 registros e, destes, selecionados 20. As amostras dos estudos selecionados incluíram, na maioria, indivíduos com distrofia muscular de Duchenne, com idade entre 5 e 22 anos, que fizeram fisioterapia aquática com duração média de 45 minutos uma ou duas vezes por semana, por 21 semanas. Essas características corroboram estudos feitos em diferentes populações. A maioria dos estudos investigou alterações pulmonares e controle postural/desempenho funcional, poucos avaliaram os efeitos no sistema cardíaco. Recomenda-se usar a Egen Klassifikation, a North Star Ambulatory Assessment e fazer o teste de caminhada de seis minutos.
Collapse
|
41
|
Mercuri E, Bönnemann CG, Muntoni F. Muscular dystrophies. Lancet 2019; 394:2025-2038. [PMID: 31789220 DOI: 10.1016/s0140-6736(19)32910-1] [Citation(s) in RCA: 269] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 09/02/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022]
Abstract
Muscular dystrophies are primary diseases of muscle due to mutations in more than 40 genes, which result in dystrophic changes on muscle biopsy. Now that most of the genes responsible for these conditions have been identified, it is possible to accurately diagnose them and implement subtype-specific anticipatory care, as complications such as cardiac and respiratory muscle involvement vary greatly. This development and advances in the field of supportive medicine have changed the standard of care, with an overall improvement in the clinical course, survival, and quality of life of affected individuals. The improved understanding of the pathogenesis of these diseases is being used for the development of novel therapies. In the most common form, Duchenne muscular dystrophy, a few personalised therapies have recently achieved conditional approval and many more are at advanced stages of clinical development. In this Seminar, we concentrate on clinical manifestations, molecular pathogenesis, diagnostic strategy, and therapeutic developments for this group of conditions.
Collapse
Affiliation(s)
- Eugenio Mercuri
- Pediatric Neurology Unit, Università Cattolica del Sacro Cuore Roma, Rome, Italy; Nemo Clinical Centre, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London, UK; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
42
|
Janssen MJ, Nieskens TTG, Steevels TAM, Caetano-Pinto P, den Braanker D, Mulder M, Ponstein Y, Jones S, Masereeuw R, den Besten C, Wilmer MJ. Therapy with 2'-O-Me Phosphorothioate Antisense Oligonucleotides Causes Reversible Proteinuria by Inhibiting Renal Protein Reabsorption. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 18:298-307. [PMID: 31610379 PMCID: PMC6796739 DOI: 10.1016/j.omtn.2019.08.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 08/28/2019] [Indexed: 11/18/2022]
Abstract
Antisense oligonucleotide therapy has been reported to be associated with renal injury. Here, the mechanism of reversible proteinuria was investigated by combining clinical, pre-clinical, and in vitro data. Urine samples were obtained from Duchenne muscular dystrophy (DMD) patients treated with drisapersen, a modified 2′O-methyl phosphorothioate antisense oligonucleotide (6 mg/kg). Urine and kidney tissue samples were collected from cynomolgus monkeys (Macaca fascicularis) dosed with drisapersen (39 weeks). Cell viability and protein uptake were evaluated in vitro using human conditionally immortalized proximal tubule epithelial cells (ciPTECs). Oligonucleotide treatment in DMD patients was associated with an increase in urinary alpha-1-microglobulin (A1M), which returned to baseline following treatment interruptions. In monkeys, increased urinary A1M correlated with dose-dependent accumulation of oligonucleotide in kidney tissue without evidence of tubular damage. Furthermore, oligonucleotides accumulated in the lysosomes of ciPTECs and reduced the absorption of A1M, albumin, and receptor-associated protein, but did not affect cell viability when incubated for up to 7 days. In conclusion, phosphorothioate oligonucleotides appear to directly compete for receptor-mediated endocytosis in proximal tubules. We postulate that oligonucleotide-induced low molecular weight proteinuria in patients is therefore a transient functional change and not indicative of tubular damage.
Collapse
Affiliation(s)
- Manoe J Janssen
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, the Netherlands.
| | - Tom T G Nieskens
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | | | - Pedro Caetano-Pinto
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, the Netherlands
| | - Dirk den Braanker
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | | | | | | | - Rosalinde Masereeuw
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, the Netherlands
| | | | - Martijn J Wilmer
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| |
Collapse
|
43
|
Muntoni F, Domingos J, Manzur AY, Mayhew A, Guglieri M, Sajeev G, Signorovitch J, Ward SJ. Categorising trajectories and individual item changes of the North Star Ambulatory Assessment in patients with Duchenne muscular dystrophy. PLoS One 2019; 14:e0221097. [PMID: 31479456 PMCID: PMC6719875 DOI: 10.1371/journal.pone.0221097] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 07/30/2019] [Indexed: 01/16/2023] Open
Abstract
Functional variability among boys with Duchenne muscular dystrophy (DMD) is well recognised and complicates interpretation of clinical studies. We hypothesised that boys with DMD could be clustered into groups sharing similar trajectories of ambulatory function over time, as measured by the North Star Ambulatory Assessment (NSAA) total score. We also explored associations with other variables such as age, functional abilities, and genotype. Using the NorthStar Clinical Network database, 395 patients with >1 NSAA assessment were identified. We utilised latent class trajectory analysis of longitudinal NSAA scores, which produced evidence for at least four clusters of boys sharing similar trajectories versus age in decreasing order of clinical severity: 25% of the boys were in cluster 1 (NSAA falling to ≤ 5 at age ~10y), 35% were in cluster 2 (NSAA ≤ 5 ~12y), 21% in were cluster 3 (NSAA≤ 5 ~14y), and 19% in cluster 4 (NSAA > 5 up to 15y). Mean ages at diagnosis of DMD were similar across clusters (4.2, 3.9, 4.3, and 4.8y, respectively). However, at the first NSAA assessment, a significant (p<0.05) association was observed between earlier declining clusters and younger age, worse NSAA, slower rise from supine, slower 10 metre walk/run times, and younger age of steroid initiation. In order to assess the probability of observing complete loss of function for individual NSAA items, we examined the proportion of patients who shifted from a score of 1 or 2 at baseline to a score of 0. We also assessed the probability of gain of function using the inverse assessment and stratified the probability of deterioration, improvement-or static behavior-by age ranges and using baseline functional status. Using this tool, our study provides a comprehensive assessment of the NSAA in a large population of patients with DMD and, for the first time, describes discrete clusters of disease progression; this will be invaluable for future DMD clinical trial design and interpretation of findings.
Collapse
Affiliation(s)
- Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, United Kingdom
- National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- * E-mail:
| | - Joana Domingos
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, United Kingdom
| | - Adnan Y. Manzur
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, United Kingdom
| | - Anna Mayhew
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle, United Kingdom
| | - Michela Guglieri
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle, United Kingdom
| | | | - Gautam Sajeev
- Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - James Signorovitch
- Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Susan J. Ward
- Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
| |
Collapse
|
44
|
Mercuri E, Lucibello S, Pera MC, Carnicella S, Coratti G, de Sanctis R, Messina S, Mazzone E, Forcina N, Fanelli L, Norcia G, Antonaci L, Frongia AL, Pane M. Long-term progression in type II spinal muscular atrophy: A retrospective observational study. Neurology 2019; 93:e1241-e1247. [PMID: 31451515 DOI: 10.1212/wnl.0000000000008166] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/30/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To report the long-term progression in a cohort of patients with type II spinal muscular atrophy (SMA) assessed with the Hammersmith Functional Motor Scale-Expanded. METHODS Seventy-three patients (age 2.6-25 years) were included in the study. Twenty-eight of the 73 were first assessed before the age of 5 years and had been followed up for ≈5 years or longer. We observed an overall progression that was not linear. A piecewise regression analysis showed an improvement of scores in the younger patients with a point of slope change at ≈5 years of age, a decline between 5 and 13 years of age, and stability/slower decline after that. RESULTS Patients with the lowest scores at baseline had the earliest onset of scoliosis and a higher need for noninvasive ventilation compared to those with higher scores. Our results confirm that on the long-term follow-up all patients with type II SMA show a clear and progressive decline. CONCLUSION The severity of functional impairment at baseline can help to predict the magnitude of changes over time and the overall progression, including onset of scoliosis and need for noninvasive ventilation.
Collapse
Affiliation(s)
- Eugenio Mercuri
- From Pediatric Neurology (E.M., S.L., M.C.P., G.C., L.A., A.L.F.), Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore; Centro Clinico Nemo (E.M., S.L., M.C.P., S.C., G.C., R.d.S., E.M., N.F., L.F., G.N., L.A., A.L.F., M.P.), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome; and Department of Neurosciences, Psychiatry and Anaesthesiology (S.M.), University of Messina, Italy.
| | - Simona Lucibello
- From Pediatric Neurology (E.M., S.L., M.C.P., G.C., L.A., A.L.F.), Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore; Centro Clinico Nemo (E.M., S.L., M.C.P., S.C., G.C., R.d.S., E.M., N.F., L.F., G.N., L.A., A.L.F., M.P.), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome; and Department of Neurosciences, Psychiatry and Anaesthesiology (S.M.), University of Messina, Italy
| | - Maria Carmela Pera
- From Pediatric Neurology (E.M., S.L., M.C.P., G.C., L.A., A.L.F.), Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore; Centro Clinico Nemo (E.M., S.L., M.C.P., S.C., G.C., R.d.S., E.M., N.F., L.F., G.N., L.A., A.L.F., M.P.), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome; and Department of Neurosciences, Psychiatry and Anaesthesiology (S.M.), University of Messina, Italy
| | - Sara Carnicella
- From Pediatric Neurology (E.M., S.L., M.C.P., G.C., L.A., A.L.F.), Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore; Centro Clinico Nemo (E.M., S.L., M.C.P., S.C., G.C., R.d.S., E.M., N.F., L.F., G.N., L.A., A.L.F., M.P.), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome; and Department of Neurosciences, Psychiatry and Anaesthesiology (S.M.), University of Messina, Italy
| | - Giorgia Coratti
- From Pediatric Neurology (E.M., S.L., M.C.P., G.C., L.A., A.L.F.), Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore; Centro Clinico Nemo (E.M., S.L., M.C.P., S.C., G.C., R.d.S., E.M., N.F., L.F., G.N., L.A., A.L.F., M.P.), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome; and Department of Neurosciences, Psychiatry and Anaesthesiology (S.M.), University of Messina, Italy
| | - Roberto de Sanctis
- From Pediatric Neurology (E.M., S.L., M.C.P., G.C., L.A., A.L.F.), Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore; Centro Clinico Nemo (E.M., S.L., M.C.P., S.C., G.C., R.d.S., E.M., N.F., L.F., G.N., L.A., A.L.F., M.P.), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome; and Department of Neurosciences, Psychiatry and Anaesthesiology (S.M.), University of Messina, Italy
| | - Sonia Messina
- From Pediatric Neurology (E.M., S.L., M.C.P., G.C., L.A., A.L.F.), Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore; Centro Clinico Nemo (E.M., S.L., M.C.P., S.C., G.C., R.d.S., E.M., N.F., L.F., G.N., L.A., A.L.F., M.P.), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome; and Department of Neurosciences, Psychiatry and Anaesthesiology (S.M.), University of Messina, Italy
| | - Elena Mazzone
- From Pediatric Neurology (E.M., S.L., M.C.P., G.C., L.A., A.L.F.), Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore; Centro Clinico Nemo (E.M., S.L., M.C.P., S.C., G.C., R.d.S., E.M., N.F., L.F., G.N., L.A., A.L.F., M.P.), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome; and Department of Neurosciences, Psychiatry and Anaesthesiology (S.M.), University of Messina, Italy
| | - Nicola Forcina
- From Pediatric Neurology (E.M., S.L., M.C.P., G.C., L.A., A.L.F.), Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore; Centro Clinico Nemo (E.M., S.L., M.C.P., S.C., G.C., R.d.S., E.M., N.F., L.F., G.N., L.A., A.L.F., M.P.), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome; and Department of Neurosciences, Psychiatry and Anaesthesiology (S.M.), University of Messina, Italy
| | - Lavinia Fanelli
- From Pediatric Neurology (E.M., S.L., M.C.P., G.C., L.A., A.L.F.), Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore; Centro Clinico Nemo (E.M., S.L., M.C.P., S.C., G.C., R.d.S., E.M., N.F., L.F., G.N., L.A., A.L.F., M.P.), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome; and Department of Neurosciences, Psychiatry and Anaesthesiology (S.M.), University of Messina, Italy
| | - Giulia Norcia
- From Pediatric Neurology (E.M., S.L., M.C.P., G.C., L.A., A.L.F.), Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore; Centro Clinico Nemo (E.M., S.L., M.C.P., S.C., G.C., R.d.S., E.M., N.F., L.F., G.N., L.A., A.L.F., M.P.), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome; and Department of Neurosciences, Psychiatry and Anaesthesiology (S.M.), University of Messina, Italy
| | - Laura Antonaci
- From Pediatric Neurology (E.M., S.L., M.C.P., G.C., L.A., A.L.F.), Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore; Centro Clinico Nemo (E.M., S.L., M.C.P., S.C., G.C., R.d.S., E.M., N.F., L.F., G.N., L.A., A.L.F., M.P.), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome; and Department of Neurosciences, Psychiatry and Anaesthesiology (S.M.), University of Messina, Italy
| | - Anna Lia Frongia
- From Pediatric Neurology (E.M., S.L., M.C.P., G.C., L.A., A.L.F.), Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore; Centro Clinico Nemo (E.M., S.L., M.C.P., S.C., G.C., R.d.S., E.M., N.F., L.F., G.N., L.A., A.L.F., M.P.), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome; and Department of Neurosciences, Psychiatry and Anaesthesiology (S.M.), University of Messina, Italy
| | - Marika Pane
- From Pediatric Neurology (E.M., S.L., M.C.P., G.C., L.A., A.L.F.), Department of Woman and Child Health and Public Health, Child Health Area, Università Cattolica del Sacro Cuore; Centro Clinico Nemo (E.M., S.L., M.C.P., S.C., G.C., R.d.S., E.M., N.F., L.F., G.N., L.A., A.L.F., M.P.), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome; and Department of Neurosciences, Psychiatry and Anaesthesiology (S.M.), University of Messina, Italy
| |
Collapse
|
45
|
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: 41] [Impact Index Per Article: 6.8] [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
|
46
|
Pera MC, Coratti G, Mazzone ES, Montes J, Scoto M, De Sanctis R, Main M, Mayhew A, Muni Lofra R, Dunaway Young S, Glanzman AM, Duong T, Pasternak A, Ramsey D, Darras B, Day JW, Finkel RS, De Vivo DC, Sormani MP, Bovis F, Straub V, Muntoni F, Pane M, Mercuri E. Revised upper limb module for spinal muscular atrophy: 12 month changes. Muscle Nerve 2019; 59:426-430. [DOI: 10.1002/mus.26419] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 01/15/2019] [Accepted: 01/19/2019] [Indexed: 11/12/2022]
Affiliation(s)
- Maria Carmela Pera
- Paediatric Neurology and Centro Clinico Nemo; Catholic University and Policlinico Gemelli, Fondazione Policlinico Universitario Agostino Gemelli IRCSS; Rome Italy
| | - Giorgia Coratti
- Paediatric Neurology and Centro Clinico Nemo; Catholic University and Policlinico Gemelli, Fondazione Policlinico Universitario Agostino Gemelli IRCSS; Rome Italy
| | - Elena S. Mazzone
- Paediatric Neurology and Centro Clinico Nemo; Catholic University and Policlinico Gemelli, Fondazione Policlinico Universitario Agostino Gemelli IRCSS; Rome Italy
| | - Jacqueline Montes
- Departments of Rehabilitation and Regenerative Medicine; Columbia University Medical Center; New York USA
- Departments of Neurology; Columbia University Medical Center; New York USA
| | - Mariacristina Scoto
- Dubowitz Neuromuscular Centre and NIHR Great Ormond Street Hospital Biomedical Research Centre; Institute of Child Health & Great Ormond Street Hospital; London UK
| | - Roberto De Sanctis
- Paediatric Neurology and Centro Clinico Nemo; Catholic University and Policlinico Gemelli, Fondazione Policlinico Universitario Agostino Gemelli IRCSS; Rome Italy
| | - Marion Main
- Dubowitz Neuromuscular Centre and NIHR Great Ormond Street Hospital Biomedical Research Centre; Institute of Child Health & Great Ormond Street Hospital; London UK
| | - Anna Mayhew
- John Walton Muscular Dystrophy Research Centre; Newcastle University; UK
| | - Robert Muni Lofra
- John Walton Muscular Dystrophy Research Centre; Newcastle University; UK
| | - Sally Dunaway Young
- Departments of Rehabilitation and Regenerative Medicine; Columbia University Medical Center; New York USA
- Departments of Neurology; Columbia University Medical Center; New York USA
| | - Allan M. Glanzman
- Department of Physical Therapy; The Children's Hospital of Philadelphia; Philadelphia
| | - Tina Duong
- Department of Neurology; Stanford University; Stanford California USA
| | - Amy Pasternak
- Department of Neurology; Boston Children's Hospital, Harvard Medical School; Boston MA USA
- Department of Physical Therapy and Occupational Therapy; Boston Children's Hospital; Boston MA USA
| | - Danielle Ramsey
- Dubowitz Neuromuscular Centre and NIHR Great Ormond Street Hospital Biomedical Research Centre; Institute of Child Health & Great Ormond Street Hospital; London UK
| | - Basil Darras
- Department of Neurology; Boston Children's Hospital, Harvard Medical School; Boston MA USA
| | - John W. Day
- Department of Neurology; Stanford University; Stanford California USA
| | - Richard S. Finkel
- Nemours Children's Hospital; University of Central Florida College of Medicine; Orlando USA
| | - Darryl C. De Vivo
- Departments of Neurology; Columbia University Medical Center; New York USA
| | - Maria Pia Sormani
- Department of Health Sciences (DISSAL); Section of Biostatistics, University of Genova; Genova Italy
| | - Francesca Bovis
- Department of Health Sciences (DISSAL); Section of Biostatistics, University of Genova; Genova Italy
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre; Newcastle University; UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre and NIHR Great Ormond Street Hospital Biomedical Research Centre; Institute of Child Health & Great Ormond Street Hospital; London UK
| | - Marika Pane
- Paediatric Neurology and Centro Clinico Nemo; Catholic University and Policlinico Gemelli, Fondazione Policlinico Universitario Agostino Gemelli IRCSS; Rome Italy
| | - Eugenio Mercuri
- Paediatric Neurology and Centro Clinico Nemo; Catholic University and Policlinico Gemelli, Fondazione Policlinico Universitario Agostino Gemelli IRCSS; Rome Italy
| | | |
Collapse
|
47
|
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
|
48
|
Straub V, Mercuri E. Report on the workshop: Meaningful outcome measures for Duchenne muscular dystrophy, London, UK, 30-31 January 2017. Neuromuscul Disord 2018; 28:690-701. [PMID: 30033203 DOI: 10.1016/j.nmd.2018.05.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Volker Straub
- Institute of Genetic Medicine, Newcastle University John Walton Muscular Dystrophy Research Centre, Newcastle, UK
| | - Eugenio Mercuri
- Pediatric Neurology Unit, Fondazione Policlinico Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy.
| | | |
Collapse
|
49
|
Birnkrant DJ, Bushby K, Bann CM, Apkon SD, Blackwell A, Brumbaugh D, Case LE, Clemens PR, Hadjiyannakis S, Pandya S, Street N, Tomezsko J, Wagner KR, Ward LM, Weber DR. Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and neuromuscular, rehabilitation, endocrine, and gastrointestinal and nutritional management. Lancet Neurol 2018; 17:251-267. [PMID: 29395989 PMCID: PMC5869704 DOI: 10.1016/s1474-4422(18)30024-3] [Citation(s) in RCA: 730] [Impact Index Per Article: 104.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 10/03/2017] [Accepted: 11/20/2017] [Indexed: 12/12/2022]
Abstract
Since the publication of the Duchenne muscular dystrophy (DMD) care considerations in 2010, multidisciplinary care of this severe, progressive neuromuscular disease has evolved. In conjunction with improved patient survival, a shift to more anticipatory diagnostic and therapeutic strategies has occurred, with a renewed focus on patient quality of life. In 2014, a steering committee of experts from a wide range of disciplines was established to update the 2010 DMD care considerations, with the goal of improving patient care. The new care considerations aim to address the needs of patients with prolonged survival, to provide guidance on advances in assessments and interventions, and to consider the implications of emerging genetic and molecular therapies for DMD. The committee identified 11 topics to be included in the update, eight of which were addressed in the original care considerations. The three new topics are primary care and emergency management, endocrine management, and transitions of care across the lifespan. In part 1 of this three-part update, we present care considerations for diagnosis of DMD and neuromuscular, rehabilitation, endocrine (growth, puberty, and adrenal insufficiency), and gastrointestinal (including nutrition and dysphagia) management.
Collapse
Affiliation(s)
- David J Birnkrant
- Department of Pediatrics, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH, USA.
| | - Katharine Bushby
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Carla M Bann
- RTI International, Research Triangle Park, NC, USA
| | - Susan D Apkon
- Department of Rehabilitation Medicine, Seattle Children's Hospital, Seattle, WA, USA
| | | | - David Brumbaugh
- Section of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Colorado, Aurora, CO, USA
| | - Laura E Case
- Doctor of Physical Therapy Division, Department of Orthopaedics, Duke University School of Medicine, Durham, NC, USA
| | - Paula R Clemens
- Department of Neurology, University of Pittsburgh School of Medicine, and Neurology Service, Department of Veterans Affairs Medical Center, Pittsburgh, PA, USA
| | - Stasia Hadjiyannakis
- Division of Endocrinology and Metabolism, Children's Hospital of Eastern Ontario, and University of Ottawa, Ottawa, ON, Canada
| | - Shree Pandya
- School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Natalie Street
- Rare Disorders and Health Outcomes Team, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jean Tomezsko
- Medical Nutrition Consulting of Media LLC, and Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kathryn R Wagner
- Center for Genetic Muscle Disorders, Kennedy Krieger Institute, and Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Leanne M Ward
- Division of Endocrinology and Metabolism, Children's Hospital of Eastern Ontario, and University of Ottawa, Ottawa, ON, Canada
| | - David R Weber
- Division of Endocrinology and Diabetes, Golisano Children's Hospital, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
50
|
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: 304] [Impact Index Per Article: 43.4] [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
|