1
|
Jolly H, Aartsma-Rus A, Bertini E, De Waele L, Haberlova J, Klein A, Niks E, Servais L. Gene therapy approval for Duchenne muscular dystrophy: a European perspective. Lancet 2025; 405:1572-1573. [PMID: 40318867 DOI: 10.1016/s0140-6736(25)00717-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 05/07/2025]
Affiliation(s)
- Hannah Jolly
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 9DU, UK
| | | | - Enrico Bertini
- Research Unit of Neuromuscular and Neurodegenerative Disease, Bambino Gesù Children's Research Hospital, IRCCS, Rome, Italy
| | - Liesbeth De Waele
- Department of Paediatrics, University Hospitals Leuven and Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Jana Haberlova
- Department of Pediatric Neurology, Neuromuscular Reference Centre, University Hospital Motol, Prague, Czech Republic
| | - Andrea Klein
- Division of Neuropaediatrics, Development and Rehabilitation, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Erik Niks
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
| | - Laurent Servais
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 9DU, UK; Department of Paediatrics, Neuromuscular Reference Centre, University Hospital of Liège, Liège, Belgium.
| |
Collapse
|
2
|
Applebaum V, Baker E, Kim T, Stimpson G, Challenor P, Wedgwood KCA, Anderson M, Bamsey I, Baranello G, Manzur A, Muntoni F, Tsaneva-Atanasova K. Fully personalized modelling of Duchenne Muscular Dystrophy ambulation. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2025; 383:20240218. [PMID: 40172561 DOI: 10.1098/rsta.2024.0218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/20/2024] [Accepted: 01/14/2025] [Indexed: 04/04/2025]
Abstract
Duchenne Muscular Dystrophy is a progressive neuromuscular disorder characterized by the gradual weakening and deterioration of muscles, leading to loss of ambulation in affected individuals. This decline in mobility can be effectively assessed using the North Star Ambulatory Assessment (NSAA) scores, along with measures such as the 10-m walk time and the time taken to rise from the floor. We propose a dynamic linear model to predict the trajectories of these clinical outcomes, with a primary focus on NSAA scores. Our model aims to assist clinicians in forecasting the progression of the disease, thereby enabling more informed and personalized treatment plans for their patients. We also evaluate the effectiveness of our models in generating synthetic NSAA score datasets. We assess the performance of our modelling approach and compare the results with those of a previous study. We show that the most robust model demonstrates narrower prediction intervals and improved quantile coverage, indicating superior predictive accuracy and reliability.This article is part of the theme issue 'Uncertainty quantification for healthcare and biological systems (Part 2)'.
Collapse
Affiliation(s)
- Victor Applebaum
- Department of Mathematics and Statistics and EPSRC Hub for Quantitative Modelling in Healthcare, University of Exeter, Exeter, UK
| | - Evan Baker
- Department of Mathematics and Statistics and EPSRC Hub for Quantitative Modelling in Healthcare, University of Exeter, Exeter, UK
| | - Thomas Kim
- Certus Technology Associates Ltd, Exeter, UK
| | - Georgia Stimpson
- 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, UK
| | - Peter Challenor
- Department of Mathematics and Statistics and EPSRC Hub for Quantitative Modelling in Healthcare, University of Exeter, Exeter, UK
| | - Kyle Carlton Abesser Wedgwood
- Department of Mathematics and Statistics and EPSRC Hub for Quantitative Modelling in Healthcare, University of Exeter, Exeter, UK
| | | | - Ian Bamsey
- Certus Technology Associates Ltd, Exeter, UK
| | - Giovanni Baranello
- 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, UK
| | - 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, UK
| | - 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, UK
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics and Statistics and EPSRC Hub for Quantitative Modelling in Healthcare, University of Exeter, Exeter, UK
| |
Collapse
|
3
|
Mayhew AG, Signorovitch J, Johnson M, Frean M, Ward SJ, Posner N, Merla V, Mahn M, Stimpson G, Guglieri M, Straub V, Muni-Lofra R, Manzur A, Baranello G, Muntoni F. Visualizing ambulatory performance by age and rates of decline among patients with Duchenne muscular dystrophy. J Neuromuscul Dis 2025:22143602241313116. [PMID: 40097910 DOI: 10.1177/22143602241313116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
In Duchenne muscular dystrophy (DMD), age at symptom onset and rate of decline thereafter vary considerably. This study contrasted disease progression over time using the North Star Ambulatory Assessment (NSAA) in an overall sample of patients with DMD (mean age 7.1 years; baseline total NSAA score 22.2) with that of a centrally representative subgroup (mean age 6.9 years; NSAA score 24.0) defined according to median age at loss of ambulation. The average disease trajectory in the overall sample understated the more rapid rates of decline experienced by patients in the centrally representative subgroup.
Collapse
Affiliation(s)
- Anna G Mayhew
- The John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle upon Tyne, UK
| | - James Signorovitch
- Analysis Group, Inc., Boston, MA, USA
- Collaborative Trajectory Analysis Project (cTAP), Cambridge, MA, USA
| | | | | | - Susan J Ward
- Collaborative Trajectory Analysis Project (cTAP), Cambridge, MA, USA
| | | | | | | | - Georgia Stimpson
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Michela Guglieri
- The John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle upon Tyne, UK
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle upon Tyne, UK
| | - Robert Muni-Lofra
- The John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle upon Tyne, UK
| | - Adnan Manzur
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Giovanni Baranello
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
4
|
Chesshyre M, Ridout D, Stimpson G, Ricotti V, De Lucia S, Niks EH, Straub V, Servais L, Hogrel JY, Baranello G, Manzur A, Muntoni F. Dystrophin isoform deficiency and upper-limb and respiratory function in Duchenne muscular dystrophy. Dev Med Child Neurol 2025. [PMID: 40084496 DOI: 10.1111/dmcn.16282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 03/16/2025]
Abstract
AIM To investigate the associations between mutations expected to differentially affect Dp140 expression and long-term trajectories of respiratory and upper-limb motor outcomes in Duchenne muscular dystrophy (DMD). METHOD In a retrospective analysis of population-based longitudinal data from three real-world and natural history data sources, individuals with DMD aged 5 years to 18 years were subdivided according to the predicted effects of the participants' DMD mutation on dystrophin isoform expression (group 1, Dp427 absent, Dp140/Dp71 present; group 2, Dp427/Dp140 absent, Dp71 present). RESULTS A total of 459 participants were studied, with upper-limb outcomes assessed in 71 (27 in group 1 and 44 in group 2) and forced vital capacity percentage predicted (%pred) assessed in 434 (224 in group 1 and 210 in group 2). Mean grip strength %pred was on average 7.1 percentage points lower in group 2 than in group 1 (p = 0.03). Mean pinch strength %pred was on average 9.2 percentage points lower in group 2 than in group 1 (p = 0.04). Mean forced vital capacity %pred was on average 4.3 percentage points lower in group 2 than in group 1 (p = 0.01). INTERPRETATION In individuals with DMD, DMD mutations predicted to affect Dp140 expression were associated with more severe trajectories of respiratory and upper-limb motor outcomes.
Collapse
Affiliation(s)
- Mary Chesshyre
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Deborah Ridout
- Population, Policy and Practice Research and Teaching Department, UCL GOS Institute of Child Health, London, UK
| | - Georgia Stimpson
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Valeria Ricotti
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, UCL, London, UK
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Silvana De Lucia
- AP-HP, Sorbonne University, Institute of Myology, AFM-Telethon, I-Motion Pediatric Clinical Trials Platform, Armand Trousseau Hospital, Paris, France
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, University of Newcastle, Newcastle upon Tyne, UK
| | - Laurent Servais
- Centre de Référence Des Maladies Neuromusculaires, CHU de Liège, University of Liège, Liège, Belgium
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre, Oxford, UK
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | | | - Giovanni Baranello
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Adnan Manzur
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, UCL, London, UK
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
5
|
Mendell JR, Muntoni F, McDonald CM, Mercuri EM, Ciafaloni E, Komaki H, Leon-Astudillo C, Nascimento A, Proud C, Schara-Schmidt U, Veerapandiyan A, Zaidman CM, Guridi M, Murphy AP, Reid C, Wandel C, Asher DR, Darton E, Mason S, Potter RA, Singh T, Zhang W, Fontoura P, Elkins JS, Rodino-Klapac LR. AAV gene therapy for Duchenne muscular dystrophy: the EMBARK phase 3 randomized trial. Nat Med 2025; 31:332-341. [PMID: 39385046 PMCID: PMC11750718 DOI: 10.1038/s41591-024-03304-z] [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: 03/22/2024] [Accepted: 09/17/2024] [Indexed: 10/11/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a rare, X-linked neuromuscular disease caused by pathogenic variants in the DMD gene that result in the absence of functional dystrophin, beginning at birth and leading to progressive impaired motor function, loss of ambulation and life-threatening cardiorespiratory complications. Delandistrogene moxeparvovec, an adeno-associated rh74-viral vector-based gene therapy, addresses absent functional dystrophin in DMD. Here the phase 3 EMBARK study aimed to assess the efficacy and safety of delandistrogene moxeparvovec in patients with DMD. Ambulatory males with DMD, ≥4 years to <8 years of age, were randomized and stratified by age group and North Star Ambulatory Assessment (NSAA) score to single-administration intravenous delandistrogene moxeparvovec (1.33 × 1014 vector genomes per kilogram; n = 63) or placebo (n = 62). At week 52, the primary endpoint, change from baseline in NSAA score, was not met (least squares mean 2.57 (delandistrogene moxeparvovec) versus 1.92 (placebo) points; between-group difference, 0.65; 95% confidence interval (CI), -0.45, 1.74; P = 0.2441). Secondary efficacy endpoints included mean micro-dystrophin expression at week 12: 34.29% (treated) versus 0.00% (placebo). Other secondary efficacy endpoints at week 52 (between-group differences (95% CI)) included: Time to Rise (-0.64 (-1.06, -0.23)), 10-meter Walk/Run (-0.42 (-0.71, -0.13)), stride velocity 95th centile (0.10 (0.00, 0.19)), 100-meter Walk/Run (-3.29 (-8.28, 1.70)), time to ascend 4 steps (-0.36 (-0.71, -0.01)), PROMIS Mobility and Upper Extremity (0.05 (-0.08, 0.19); -0.04 (-0.24, 0.17)) and number of NSAA skills gained/improved (0.19 (-0.67, 1.06)). In total, 674 adverse events were recorded with delandistrogene moxeparvovec and 514 with placebo. There were no deaths, discontinuations or clinically significant complement-mediated adverse events; 7 patients (11.1%) experienced 10 treatment-related serious adverse events. Delandistrogene moxeparvovec did not lead to a significant improvement in NSAA score at week 52. Some of the secondary endpoints numerically favored treatment, although no statistical significance can be claimed. Safety was manageable and consistent with previous delandistrogene moxeparvovec trials. ClinicalTrials.gov: NCT05096221.
Collapse
Affiliation(s)
- Jerry R Mendell
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA.
- The Ohio State University, Columbus, OH, USA.
- Sarepta Therapeutics, Inc., Cambridge, MA, USA.
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health and Institute of Neurology, University College London and Great Ormond Street Hospital Trust, London, UK
| | | | - Eugenio M Mercuri
- Pediatric Neurology Institute, Catholic University and Nemo Pediatrico, Fondazione Policlinico Gemelli IRCCS, Rome, Italy
| | - Emma Ciafaloni
- University of Rochester Medical Center, Rochester, NY, USA
| | - Hirofumi Komaki
- Translational Medical Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | | | - Andrés Nascimento
- Neuromuscular Unit, Neuropaediatrics Department, Hospital Sant Joan de Déu, Fundacion Sant Joan de Déu, CIBERER - ISC III, Barcelona, Spain
| | - Crystal Proud
- Children's Hospital of the King's Daughters, Norfolk, VA, USA
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology, Center for Neuromuscular Disorders in Children and Adolescents, University Clinic Essen, University of Duisburg-Essen, Essen, Germany
| | - Aravindhan Veerapandiyan
- Department of Pediatrics, Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock, AR, USA
| | - Craig M Zaidman
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | | | | | - Carol Reid
- Roche Products, Ltd., Welwyn Garden City, UK
| | | | | | | | | | | | - Teji Singh
- Sarepta Therapeutics, Inc., Cambridge, MA, USA
| | | | | | | | | |
Collapse
|
6
|
Lee J, Park SE, Kim M, Kim H, Kwon JY, Jeon HB, Chang JW, Lee J. Safety and Tolerability of Wharton's Jelly-Derived Mesenchymal Stem Cells for Patients With Duchenne Muscular Dystrophy: A Phase 1 Clinical Study. J Clin Neurol 2025; 21:40-52. [PMID: 39778566 PMCID: PMC11711273 DOI: 10.3988/jcn.2024.0299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/15/2024] [Accepted: 10/31/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND AND PURPOSE This study was an open-label, dose-escalation, phase 1 clinical trial to determine the safety and dose of EN001 for patients with Duchenne muscular dystrophy (DMD). EN001, developed by ENCell, are allogeneic early-passage Wharton's jelly-derived mesenchymal stem cells that originate at the umbilical cord, with preclinical studies demonstrating their high therapeutic efficacy for DMD. METHODS This phase 1 clinical trial explored the safety and tolerability of EN001 as a potential treatment option for patients with DMD. Six pediatric participants with DMD were divided into two subgroups of equal size: low-dose EN001 (5.0×10⁵ cells/kg) and high-dose EN001 (2.5×10⁶ cells/kg). All participants were monitored for 12 weeks after EN001 administration to assess its safety. Dose-limiting toxicity (DLT) was evaluated across 2 weeks post administration. Exploratory efficacy was evaluated by measuring serum creatine kinase levels, and functional evaluations-including spirometry, myometry, the North Star Ambulatory Assessment, and the 6-minute walk test-were conducted at week 12 and compared with the baseline values. RESULTS No participants experienced serious adverse events related to EN001 injection during the 12-week follow-up period. Mild adverse events included injection-related local erythema, edema, parosmia, and headache, but DLT was not observed. Functional evaluations at week 12 revealed no significant changes from baseline. CONCLUSIONS These results demonstrated that EN001 are safe and well tolerated for patients with DMD, and did not cause serious adverse events. The efficacy of EN001 could be confirmed through larger-scale future studies that incorporate repeated dosing and have a randomized controlled trial design.
Collapse
Affiliation(s)
- Jiwon Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang Eon Park
- Cell and Gene Therapy Research Institute, ENCell Co. Ltd., Seoul, Korea
- Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Mira Kim
- Clinical Development Department, ENCell Co. Ltd., Seoul, Korea
| | - Hyeongseop Kim
- Cell and Gene Therapy Research Institute, ENCell Co. Ltd., Seoul, Korea
- Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Jeong-Yi Kwon
- Department of Physical and Rehabilitation Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hong Bae Jeon
- Cell and Gene Therapy Research Institute, ENCell Co. Ltd., Seoul, Korea
| | - Jong Wook Chang
- Cell and Gene Therapy Research Institute, ENCell Co. Ltd., Seoul, Korea
- Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Jeehun Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea.
| |
Collapse
|
7
|
Stimpson G, James MK, Guglieri M, Wolfe A, Manzur A, Sarkozy A, Baranello G, Muntoni F, Mayhew A. Understanding North Star Ambulatory Assessment total scores and their implications for standards of care using observational data. Eur J Paediatr Neurol 2024; 53:123-130. [PMID: 39500128 DOI: 10.1016/j.ejpn.2024.09.004] [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: 04/26/2024] [Revised: 08/28/2024] [Accepted: 09/11/2024] [Indexed: 12/01/2024]
Abstract
NorthStar Ambulatory Assessment (NSAA) total score (TS) is an ordinal scale to evaluate disease progression and treatment response in ambulatory Duchenne Muscular Dystrophy individuals. Clinical management according to standard of care could be enhanced by understanding how changes in the TS could inform standards of care. Here we describe the associated item performance patterns in the NorthStar Database for ranges of NSAA TS and its timed tests (10 m walk/run and rise from floor). We then compare these patterns depending on whether a participant is on an improving/stable (≤2-point loss in the prior year) or declining (>2-point loss in the prior year) trend. These TS and trends are subsequently linked and referenced to therapy standards of care. We included 761 participants from the UK NorthStar observational clinical database between 5 and 16 years, who were on steroids. Differences and trends in item ability, compensations, and times can suggest specific disease complications and lead towards anticipatory therapy recommendations. Families and therapists can benefit from using the TS and trend to guide therapy management.
Collapse
Affiliation(s)
- Georgia Stimpson
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Meredith K James
- Newcastle University and Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Michela Guglieri
- Newcastle University and Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Amy Wolfe
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK; Evelina London Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Adnan Manzur
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Anna Sarkozy
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Giovanni Baranello
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Anna Mayhew
- Newcastle University and Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
8
|
Baranello G, Muntoni F. AAV gene therapy for Duchenne Muscular Dystrophy: lessons learned from a phase 3 trial. Gene Ther 2024; 31:541-543. [PMID: 39443734 DOI: 10.1038/s41434-024-00494-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/29/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Affiliation(s)
- Giovanni Baranello
- The Dubowitz Neuromuscular Centre, Developmental Neuroscience Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre & Great Ormond Street Hospital NHS Foundation Trust, 30 Guilford Street, London, UK.
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, Developmental Neuroscience Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre & Great Ormond Street Hospital NHS Foundation Trust, 30 Guilford Street, London, UK.
- The Genetic Therapy Accelerator, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK.
| |
Collapse
|
9
|
Coratti G, Civitello M, Rohwer A, Albamonte E, Montes J, Glanzman AM, Pasternak A, De Sanctis R, Young SD, Duong T, Mizzoni I, Milev E, Sframeli M, Morando S, D'Amico A, Catteruccia M, Brolatti N, Pane M, Scoto M, Messina S, Hirano M, Zolkipli-Cunningham Z, Darras BT, Bertini E, Bruno C, Sansone VA, Salmin F, Day J, Baranello G, Pera MC, Muntoni F, Finkel RS, Mercuri E. Upper limb function changes over 12 months in untreated SMA II and III individuals: an item-level analysis using the Revised Upper Limb Module. Neuromuscul Disord 2024; 44:104449. [PMID: 39299818 DOI: 10.1016/j.nmd.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024]
Abstract
The Revised upper limb module (RULM) has been increasingly used in clinical trials and in clinical settings. The aim of this study was to use the 'shift analysis' to assess the patterns of lost or gained abilities for each item on the RULM in an untreated cohort, stratified by SMA type, age, SMN2 copy number, and motor functional status. The analysis was performed on 222 12-month paired assessments from 129 individuals (115 assessment from type II and 107 from type III) who had at least two assessments at yearly intervals. There was a loss of one or more activities in 54% in type II and in 29% type III. A gain of one or more activities was found in 42% type II and in 22% type III. There were concomitant gains and losses in 27% in SMA II and in 9% in SMA III. Our results, measuring the number of abilities that are lost or gained, provide an additional method of detecting changes that can be used for the interpretation of the longitudinal data collected in treated SMA individuals.
Collapse
Affiliation(s)
- Giorgia Coratti
- Pediatric Neurology Unit, Catholic University, 00135 Rome, Italy.; Centro Clinico Nemo, U.O.C. Neuropsichiatria Infantile Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Matthew Civitello
- Center for Experimental Neurotherapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Annemarie Rohwer
- Department of Developmental Neuroscience, Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.; Department of Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Emilio Albamonte
- The NEMO Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, 20162 Milan, Italy
| | | | - Allan M Glanzman
- Division of Physical TherapyRehabilitation, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Amy Pasternak
- Neuromuscular Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Roberto De Sanctis
- Pediatric Neurology Unit, Catholic University, 00135 Rome, Italy.; Centro Clinico Nemo, U.O.C. Neuropsichiatria Infantile Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Sally Dunaway Young
- Departments of Neurology and Clinical Neurosciences, Stanford University, Palo Alto, CA 94305, USA
| | - Tina Duong
- Departments of Neurology and Clinical Neurosciences, Stanford University, Palo Alto, CA 94305, USA
| | - Irene Mizzoni
- Unit of Neuromuscular and Neurodegenerative Disorders, Translational Paediatrics and Clinical Genetics, Bambino Gesù Children's Hospital, IRCCS 00165 Rome, Italy
| | - Evelin Milev
- Department of Developmental Neuroscience, Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.; Department of Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Maria Sframeli
- Unit of Neurodegenerative Disorders, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy
| | - Simone Morando
- 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
| | - Adele D'Amico
- Unit of Neuromuscular and Neurodegenerative Disorders, Translational Paediatrics and Clinical Genetics, Bambino Gesù Children's Hospital, IRCCS 00165 Rome, Italy
| | - Michela Catteruccia
- Unit of Neuromuscular and Neurodegenerative Disorders, Translational Paediatrics and Clinical Genetics, Bambino Gesù Children's Hospital, IRCCS 00165 Rome, Italy
| | - Noemi Brolatti
- 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
| | - Marika Pane
- Pediatric Neurology Unit, Catholic University, 00135 Rome, Italy.; Centro Clinico Nemo, U.O.C. Neuropsichiatria Infantile Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Mariacristina Scoto
- Department of Developmental Neuroscience, Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.; Department of Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Sonia Messina
- Unit of Neurodegenerative Disorders, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy
| | - Michio Hirano
- Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Zarazuela Zolkipli-Cunningham
- Division of Human Genetics, Children's Hospital of Philadelphia, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Basil T Darras
- Neuromuscular Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Translational Paediatrics and Clinical Genetics, Bambino Gesù Children's Hospital, IRCCS 00165 Rome, 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
| | - Valeria A Sansone
- The NEMO Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, 20162 Milan, Italy
| | - Francesca Salmin
- The NEMO Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, 20162 Milan, Italy
| | - John Day
- Departments of Neurology and Clinical Neurosciences, Stanford University, Palo Alto, CA 94305, USA
| | - Giovanni Baranello
- Department of Developmental Neuroscience, Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.; Department of Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Maria Carmela Pera
- Pediatric Neurology Unit, Catholic University, 00135 Rome, Italy.; Centro Clinico Nemo, U.O.C. Neuropsichiatria Infantile Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Muntoni
- Department of Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.; School of Health and Sports Sciences, University of Suffolk, Ipswich IP4 1QJ, UK
| | - Richard S Finkel
- Center for Experimental Neurotherapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Eugenio Mercuri
- Pediatric Neurology Unit, Catholic University, 00135 Rome, Italy.; Centro Clinico Nemo, U.O.C. Neuropsichiatria Infantile Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy..
| |
Collapse
|
10
|
Gooch KL, Audhya I, Ricchetti-Masterson K, Szabo SM. Current Challenges of Using Patient-Level Claims and Electronic Health Record Data for the Longitudinal Evaluation of Duchenne Muscular Dystrophy Outcomes. Adv Ther 2024; 41:3615-3632. [PMID: 39080221 DOI: 10.1007/s12325-024-02897-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/09/2024] [Indexed: 08/28/2024]
Abstract
INTRODUCTION Insurance claims data and electronic health records (EHRs) have been used to characterize Duchenne muscular dystrophy (DMD) in real-world populations. The ability to assess patient-level DMD disease progression within insurance claims or EHR data infrastructures is unknown. Insurance claims and EHR data were comprehensively examined for availability and reliability of DMD outcomes that describe functional status and disease progression at the individual patient level over time. METHODS MarketScan Commercial and Medicaid claims, and EHR-linked Clarivate open claims datasets were examined for data measuring 54 previously identified DMD-relevant outcomes in patients with DMD. Each outcome was assigned to one of five categories: functional and clinical events, clinical measures, biomarkers, functional measures, or patient-reported outcomes (PROs). Patients were identified using published coding algorithms. Annual 5-year attrition and data availability for each outcome was determined. The ability to distinguish disease severity and identify test results was also considered where applicable. RESULTS A total of 1964 (MarketScan Commercial), 2007 (MarketScan Medicaid), and 10,639 (Clarivate) patients were identified. At 5 years, 31.7%, 35.1%, and 59.1% of patients remained in MarketScan Commercial, MarketScan Medicaid, and Clarivate, respectively. Claims were available for five of six functional and clinical events, with 45.5% (MarketScan Commercial), 48.0% (Clarivate), and 48.5% (MarketScan Medicaid) of patients with ≥ 1 claim for the most frequently identified clinical event (cardiomyopathy diagnosis). No data were available to describe frequency of wheelchair use or loss of ambulation. Very limited EHR data (≤ 2% of patients) were available to indicate tests were ordered for clinical measures, biomarkers, or functional assessments. No PRO notes or scores were observed. Data existed for inferring disease severity (e.g., hospitalization for cardiomyopathy); however, it was not apparent whether these events were incident. CONCLUSION Insurance claims and EHR-linked open claims data are of limited utility for holistically evaluating the progression and burden of DMD in individual patients.
Collapse
Affiliation(s)
- Katherine L Gooch
- Global Health Economics, Patient Centered Outcomes and RWE, Sarepta Therapeutics, Inc., 215 First St, Cambridge, MA, 02142, USA.
| | - Ivana Audhya
- Global Health Economics, Patient Centered Outcomes and RWE, Sarepta Therapeutics, Inc., 215 First St, Cambridge, MA, 02142, USA
| | - Kristen Ricchetti-Masterson
- Global Health Economics, Patient Centered Outcomes and RWE, Sarepta Therapeutics, Inc., 215 First St, Cambridge, MA, 02142, USA
| | - Shelagh M Szabo
- Pharmacoepidemiology and RWE, Broadstreet HEOR, 343 Railway St, Vancouver, BC, Canada
| |
Collapse
|
11
|
Hoskens J, Paulussen S, Goemans N, Feys H, De Waele L, Klingels K. Early motor, cognitive, language, behavioural and social emotional development in infants and young boys with Duchenne Muscular Dystrophy- A systematic review. Eur J Paediatr Neurol 2024; 52:29-51. [PMID: 39003996 DOI: 10.1016/j.ejpn.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/11/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024]
Abstract
Duchenne Muscular Dystrophy (DMD) is an X-linked recessive disorder caused by mutations in the dystrophin gene. Deficiency of the dystrophin protein causes not only motor, but also cognitive, language, behavioural and social emotional problems. This is the first systematic review investigating five early developmental domains in boys with DMD between 0 and 6 years old. Interactions between different domains and links with mutation types and sites were explored. A systematic search was performed in PubMed, Web of Science and Scopus. An adapted version of the Scottish Intercollegiate Guidelines Network (SIGN) Checklists for case-control and cohort studies was used to evaluate quality. Fifty-five studies of high or acceptable quality were included. One was an RCT of level 1b; 50 were cohort studies of level 2b; and four were an aggregation of case-control and cohort studies receiving levels 2b and 3b. We found that young boys with DMD experienced problems in all five developmental domains, with significant interactions between these. Several studies also showed relationships between mutation sites and outcomes. We conclude that DMD is not only characterised by motor problems but by a more global developmental delay with a large variability between boys. Our results emphasise the need for harmonisation in evaluation and follow-up of young boys with DMD. More high-quality research is needed on the different early developmental domains in young DMD to facilitate early detection of difficulties and identification of associated early intervention strategies.
Collapse
Affiliation(s)
- Jasmine Hoskens
- Faculty of Rehabilitation Sciences, Rehabilitation Research Centre (REVAL), UHasselt, Campus Diepenbeek, Agoralaan, 3590, Diepenbeek, Hasselt, Belgium; Department of Rehabilitation Sciences, Research Group for Neurorehabilitation (eNRGy), KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Silke Paulussen
- Faculty of Rehabilitation Sciences, Rehabilitation Research Centre (REVAL), UHasselt, Campus Diepenbeek, Agoralaan, 3590, Diepenbeek, Hasselt, Belgium
| | - Nathalie Goemans
- Department of Child Neurology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Hilde Feys
- Department of Rehabilitation Sciences, Research Group for Neurorehabilitation (eNRGy), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Liesbeth De Waele
- Department of Child Neurology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium; Department of Development and Regeneration, KU Leuven, 3000, Leuven, Belgium
| | - Katrijn Klingels
- Faculty of Rehabilitation Sciences, Rehabilitation Research Centre (REVAL), UHasselt, Campus Diepenbeek, Agoralaan, 3590, Diepenbeek, Hasselt, Belgium; Department of Rehabilitation Sciences, Research Group for Neurorehabilitation (eNRGy), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| |
Collapse
|
12
|
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
|
13
|
Coratti G, Civitello M, Rohwer A, Salmin F, Glanzman AM, Montes J, Pasternak A, De Sanctis R, Young SD, Duong T, Mizzoni I, Milev E, Sframeli M, Morando S, Albamonte E, D'Amico A, Brolatti N, Pane M, Scoto M, Messina S, Hirano M, Zolkipli-Cunningham Z, Darras BT, Bertini E, Bruno C, Sansone VA, Day J, Baranello G, Pera MC, Muntoni F, Finkel R, Mercuri E. Changes in abilities over the initial 12 months of nusinersen treatment for type II SMA. Neuromuscul Disord 2024; 41:42-50. [PMID: 38936290 DOI: 10.1016/j.nmd.2024.05.003] [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: 02/21/2024] [Revised: 04/12/2024] [Accepted: 05/08/2024] [Indexed: 06/29/2024]
Abstract
Several studies have shown the efficacy of new disease-modifying therapies in slowing down type II SMA progression using the Hammersmith Functional Motor Scale Expanded (HFMSE). This research aims to enhance understanding of activity changes across age groups post-nusinersen treatment using shift analysis, compared with untreated individuals. Retrospective data from the, international SMA consortium (iSMAc) dataset were analyzed, assessing individual item changes over 12 months. Shift analysis was used to determine the gain or loss of abilities, defining "gain" as a positive change between scores from 0 to either 1 or 2 and "loss" as a negative change from either 2 or 1 to 0. The cohort included 130 SMA II patients who underwent 12-month assessments from their first nusinersen dose, with age range between 0.6 and 49.6 years. One-third of the entire cohort experienced at least a loss in one activity, while 60% experienced a gain, particularly notable in children aged 2.5 to 5 years and 5 to 13 years. Overall, the study demonstrates a positive impact of nusinersen treatment on SMA II patients, showing a trend of increased activity gains and decreased probability of ability loss across different age groups.
Collapse
Affiliation(s)
- Giorgia Coratti
- Pediatric Neurology Unit, Catholic University, 00135 Rome, Italy; Centro Clinico Nemo, U.O.C. Neuropsichiatria Infantile Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | | | - Annemarie Rohwer
- Department of Developmental Neuroscience, Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK; Department of Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Francesca Salmin
- The NEMO Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, 20162 Milan, Italy
| | - Allan M Glanzman
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jaqueline Montes
- Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Amy Pasternak
- Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Roberto De Sanctis
- Pediatric Neurology Unit, Catholic University, 00135 Rome, Italy; Centro Clinico Nemo, U.O.C. Neuropsichiatria Infantile Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | | | - Tina Duong
- Departments of Neurology TD, Stanford University, Palo Alto, CA 94305, USA
| | - Irene Mizzoni
- Unit of Neuromuscular and Neurodegenerative Disorders, Translational Paediatrics and Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Evelin Milev
- Department of Developmental Neuroscience, Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK; Department of Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Maria Sframeli
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Simone Morando
- 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
| | - Emilio Albamonte
- The NEMO Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, 20162 Milan, Italy
| | - Adele D'Amico
- Unit of Neuromuscular and Neurodegenerative Disorders, Translational Paediatrics and Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Noemi Brolatti
- 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
| | - Marika Pane
- Pediatric Neurology Unit, Catholic University, 00135 Rome, Italy; Centro Clinico Nemo, U.O.C. Neuropsichiatria Infantile Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Mariacristina Scoto
- Department of Developmental Neuroscience, Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK; Department of Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Sonia Messina
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Michio Hirano
- Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Zarazuela Zolkipli-Cunningham
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Basil T Darras
- Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Translational Paediatrics and Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, 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
| | - Valeria A Sansone
- The NEMO Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, 20162 Milan, Italy
| | - John Day
- Departments of Neurology TD, Stanford University, Palo Alto, CA 94305, USA
| | - Giovanni Baranello
- Department of Developmental Neuroscience, Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK; Department of Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Maria Carmela Pera
- Pediatric Neurology Unit, Catholic University, 00135 Rome, Italy; Centro Clinico Nemo, U.O.C. Neuropsichiatria Infantile Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Muntoni
- Department of Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK; School of Health and Sports Sciences, University of Suffolk, Ipswich IP4 1QJ, UK
| | - Richard Finkel
- St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Eugenio Mercuri
- Pediatric Neurology Unit, Catholic University, 00135 Rome, Italy; Centro Clinico Nemo, U.O.C. Neuropsichiatria Infantile Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy.
| |
Collapse
|
14
|
McDonald CM, Signorovitch J, Mercuri E, Niks EH, Wong B, Fillbrunn M, Sajeev G, Yim E, Dieye I, Miller D, Ward SJ, Goemans N, Investigators from the PRO-DMD-01 Study, Collaborative Trajectory Analysis Project (cTAP). 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
|
15
|
Baranello G. Interpreting motor function outcomes in young males with Duchenne muscular dystrophy. Dev Med Child Neurol 2024; 66:557-558. [PMID: 38297454 DOI: 10.1111/dmcn.15858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 02/02/2024]
Abstract
This commentary is on the original article by Hoskens et al. on pages 644–653 of this issue.
Collapse
Affiliation(s)
- Giovanni Baranello
- University College London - The Dubowitz Neuromuscular Centre, UCL NIHR GOSH Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
16
|
Hoskens J, Schiava M, Goemans N, Feys H, McDermott MP, Martens WB, Mayhew A, Griggs RC, Klingels K, Guglieri M. Reference curves of motor function outcomes in young steroid-naïve males with Duchenne muscular dystrophy. Dev Med Child Neurol 2024; 66:644-653. [PMID: 37885269 DOI: 10.1111/dmcn.15788] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/12/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023]
Abstract
AIM To investigate functional motor performance in a large cohort of young steroid-naïve males with Duchenne muscular dystrophy (DMD) and typically developing males, and to develop specific reference curves for both groups. Also, to describe associations between anthropometric values and functional motor outcomes. METHOD Cross-sectional data of 196 steroid-naïve males with DMD aged 4 to 8 years and 497 typically developing males aged 2 years 6 months to 8 years were included. Both groups were evaluated with the time to rise from the floor test, 10-metre walk/run test, 6-minute walk test, and North Star Ambulatory Assessment. Reference curves with centiles 5%, 10%, 25%, 50%, 75%, 90%, and 95% were estimated using quantile regression. RESULTS Males with DMD scored significantly worse on all functional motor outcomes than age-matched typically developing males (p < 0.001): 89% to 95% of the males with DMD scored below the 5th centile of the typically developing males. No or weak correlations exist between anthropometric values and functional motor outcomes. INTERPRETATION The estimated reference curves can support consultation with families of young males with DMD and can support the evaluation of treatment for reaching motor skills and functional motor outcomes compared with typically developing males.
Collapse
Affiliation(s)
- Jasmine Hoskens
- Faculty of Rehabilitation Sciences, Rehabilitation Research Center (REVAL), UHasselt, Leuven, Belgium
- Department of Rehabilitation Sciences, Research Group for Neurorehabilitation (eNRGy), KU Leuven, Leuven, Belgium
| | - Marianela Schiava
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle upon Tyne, UK
| | - Nathalie Goemans
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Hilde Feys
- Department of Rehabilitation Sciences, Research Group for Neurorehabilitation (eNRGy), KU Leuven, Leuven, Belgium
| | - Michael P McDermott
- Department of Neurology, University of Rochester Medical Centre, Rochester, NY, USA
- Department of Biostatistics and Computational Biology, University of Rochester Medical Centre, Rochester, NY, USA
| | - William B Martens
- Department of Neurology, University of Rochester Medical Centre, Rochester, NY, USA
| | - Anna Mayhew
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle upon Tyne, UK
| | - Robert C Griggs
- Department of Neurology, University of Rochester Medical Centre, Rochester, NY, USA
| | - Katrijn Klingels
- Faculty of Rehabilitation Sciences, Rehabilitation Research Center (REVAL), UHasselt, Leuven, Belgium
- Department of Rehabilitation Sciences, Research Group for Neurorehabilitation (eNRGy), KU Leuven, Leuven, Belgium
| | - Michela Guglieri
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle upon Tyne, UK
| |
Collapse
|
17
|
Boehler JF, Brown KJ, Ricotti V, Morris CA. N-terminal titin fragment: a non-invasive, pharmacodynamic biomarker for microdystrophin efficacy. Skelet Muscle 2024; 14:2. [PMID: 38229112 PMCID: PMC10790446 DOI: 10.1186/s13395-023-00334-y] [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: 11/13/2023] [Accepted: 12/29/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Multiple clinical trials to assess the efficacy of AAV-directed gene transfer in participants with Duchenne muscular dystrophy (DMD) are ongoing. The success of these trials currently relies on standard functional outcome measures that may exhibit variability within and between participants, rendering their use as sole measures of drug efficacy challenging. Given this, supportive objective biomarkers may be useful in enhancing observed clinical results. Creatine kinase (CK) is traditionally used as a diagnostic biomarker of DMD, but its potential as a robust pharmacodynamic (PD) biomarker is difficult due to the wide variability seen within the same participant over time. Thus, there is a need for the discovery and validation of novel PD biomarkers to further support and bolster traditional outcome measures of efficacy in DMD. METHOD Potential PD biomarkers in DMD participant urine were examined using a proteomic approach on the Somalogic platform. Findings were confirmed in both mdx mice and Golden Retriever muscular dystrophy (GRMD) dog plasma samples. RESULTS Changes in the N-terminal fragment of titin, a well-known, previously characterized biomarker of DMD, were correlated with the expression of microdystrophin protein in mice, dogs, and humans. Further, titin levels were sensitive to lower levels of expressed microdystrophin when compared to CK. CONCLUSION The measurement of objective PD biomarkers such as titin may provide additional confidence in the assessment of the mechanism of action and efficacy in gene therapy clinical trials of DMD. TRIAL REGISTRATION ClinicalTrials.gov NCT03368742.
Collapse
Affiliation(s)
- Jessica F Boehler
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA, 02129, USA.
| | - Kristy J Brown
- Rejuvenate Bio, 11425 Sorrento Valley Road, San Diego, CA, 92121, USA
| | - Valeria Ricotti
- National Institute for Health and Care Research Great Ormond Street Hospital Biomedical Research Centre/University College London Great Ormond Street Institute of Child Health, London, UK
| | - Carl A Morris
- PHDL Consulting LLC, 43 Sylvanus Wood Lane, Woburn, MA, 01801, USA
| |
Collapse
|
18
|
Coratti G, Pane M, Brogna C, D'Amico A, Pegoraro E, Bello L, Sansone VA, Albamonte E, Ferraroli E, Mazzone ES, Fanelli L, Messina S, Sframeli M, Catteruccia M, Cicala G, Capasso A, Ricci M, Frosini S, De Luca G, Rolle E, De Sanctis R, Forcina N, Norcia G, Passamano L, Scutifero M, Gardani A, Pini A, Monaco G, D'Angelo MG, Leone D, Zanin R, Vita GL, Panicucci C, Bruno C, Mongini T, Ricci F, Berardinelli A, Battini R, Masson R, Baranello G, Dosi C, Bertini E, Nigro V, Politano L, Mercuri E. Gain and loss of upper limb abilities in Duchenne muscular dystrophy patients: A 24-month study. Neuromuscul Disord 2024; 34:75-82. [PMID: 38157655 DOI: 10.1016/j.nmd.2023.11.011] [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: 08/18/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a neuromuscular condition characterized by muscle weakness. The Performance of upper limb (PUL) test is designed to evaluate upper limb function in DMD patients across three domains. The aim of this study is to identify frequently lost or gained PUL 2.0 abilities at distinct functional stages in DMD patients. This retrospective study analyzed prospectively collected data on 24-month PUL 2.0 changes related to ambulatory function. Ambulant patients were categorized based on initial 6MWT distance, non-ambulant patients by time since ambulation loss. Each PUL 2.0 item was classified as shift up, no change, or shift down. The study's cohort incuded 274 patients, with 626 paired evaluations at the 24-month mark. Among these, 55.1 % had activity loss, while 29.1 % had gains. Ambulant patients showed the lowest loss rates, mainly in the shoulder domain. The highest loss rate was in the shoulder domain in the transitioning subgroup and in elbow and distal domains in the non-ambulant patients. Younger ambulant patients demonstrated multiple gains, whereas in the other functional subgroups there were fewer gains, mostly tied to singular activities. Our findings highlight divergent upper limb domain progression, partly linked to functional status and baseline function.
Collapse
Affiliation(s)
- Giorgia Coratti
- Centro Clinico Nemo, IRCCS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy; Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marika Pane
- Centro Clinico Nemo, IRCCS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy; Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudia Brogna
- Centro Clinico Nemo, IRCCS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Adele D'Amico
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Luca Bello
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Valeria A Sansone
- The NEMO Clinical Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, Milan, Italy
| | - Emilio Albamonte
- The NEMO Clinical Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, Milan, Italy
| | | | | | - Lavinia Fanelli
- Centro Clinico Nemo, IRCCS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Sonia Messina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Maria Sframeli
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Michela Catteruccia
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Gianpaolo Cicala
- Centro Clinico Nemo, IRCCS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy; Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Capasso
- Centro Clinico Nemo, IRCCS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy; Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Martina Ricci
- Centro Clinico Nemo, IRCCS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy; Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Silvia Frosini
- Department of Developmental Neuroscience, IRCCS Stella Maris, Pisa, Italy
| | - Giacomo De Luca
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Enrica Rolle
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Torino, Turin, Italy
| | - Roberto De Sanctis
- Centro Clinico Nemo, IRCCS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy; Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Nicola Forcina
- Centro Clinico Nemo, IRCCS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Giulia Norcia
- Centro Clinico Nemo, IRCCS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Luigia Passamano
- Cardiomiology and Medical Genetics, Luigi Vanvitelli University Hospital, Naples, Italy
| | - Marianna Scutifero
- Cardiomiology and Medical Genetics, Luigi Vanvitelli University Hospital, Naples, Italy
| | - Alice Gardani
- Child and Adolescence Neurological Unit, National Neurological Institute Casimiro Mondino Foundation, IRCCS, IRCCS Mondino Foundation, Pavia, Italy
| | - Antonella Pini
- Child Neurology and Psychiatry Unit, IRCCS Institute of Neurological Sciences, Bellaria Hospital, Bologna, Italy
| | - Giulia Monaco
- Child Neurology and Psychiatry Unit, IRCCS Institute of Neurological Sciences, Bellaria Hospital, Bologna, Italy
| | | | - Daniela Leone
- Centro Clinico Nemo, IRCCS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Riccardo Zanin
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Gian Luca Vita
- Unit of Neurology, IRCCS Centro Neurolesi Bonino-Pulejo - P.O. Piemonte, Messina, Italy
| | - Chiara Panicucci
- Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health-DINOGMI, Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini IRCCS, University of Genova, Genova, Italy
| | - Claudio Bruno
- Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health-DINOGMI, Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini IRCCS, University of Genova, Genova, Italy
| | - Tiziana Mongini
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Torino, Turin, Italy
| | - Federica Ricci
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Torino, Turin, Italy
| | - Angela Berardinelli
- Child and Adolescence Neurological Unit, National Neurological Institute Casimiro Mondino Foundation, IRCCS, IRCCS Mondino Foundation, Pavia, Italy
| | - Roberta Battini
- Department of Developmental Neuroscience, IRCCS Stella Maris, Pisa, Italy; Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Riccardo Masson
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giovanni Baranello
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Claudia Dosi
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Vincenzo Nigro
- Department of Precision Medicine, Luigi Vanvitelli and Telethon Institute of Genetics and Medicine, University of Campania, Italy
| | - Luisa Politano
- Cardiomiology and Medical Genetics, Luigi Vanvitelli University Hospital, Naples, Italy
| | - Eugenio Mercuri
- Centro Clinico Nemo, IRCCS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy; Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
19
|
Mendell JR, Sahenk Z, Lehman KJ, Lowes LP, Reash NF, Iammarino MA, Alfano LN, Lewis S, Church K, Shell R, Potter RA, Griffin DA, Hogan M, Wang S, Mason S, Darton E, Rodino-Klapac LR. Long-term safety and functional outcomes of delandistrogene moxeparvovec gene therapy in patients with Duchenne muscular dystrophy: A phase 1/2a nonrandomized trial. Muscle Nerve 2024; 69:93-98. [PMID: 37577753 DOI: 10.1002/mus.27955] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 08/15/2023]
Abstract
INTRODUCTION/AIMS Delandistrogene moxeparvovec is indicated in the United States for the treatment of ambulatory pediatric patients aged 4 through 5 years with Duchenne muscular dystrophy (DMD) with a confirmed mutation in the DMD gene. Long-term delandistrogene moxeparvovec microdystrophin protein (a shortened dystrophin that retains key functional domains of the wild-type protein) expression may positively alter disease progression in patients with DMD. We evaluated long-term safety and functional outcomes of delandistrogene moxeparvovec in patients with DMD. METHODS An open-label, phase 1/2a, nonrandomized controlled trial (Study 101; NCT03375164) enrolled ambulatory males, ≥4 to <8 years old, with DMD. Patients received a single intravenous infusion (2.0 × 1014 vg/kg by supercoiled quantitative polymerase chain reaction) of delandistrogene moxeparvovec and prednisone (1 mg/kg/day) 1 day before to 30 days after treatment. The primary endpoint was safety. Functional outcomes were change from baseline in North Star Ambulatory Assessment (NSAA) and timed function tests. RESULTS Four patients (mean age, 5.1 years) were enrolled. There were 18 treatment-related adverse events; all occurred within 70 days posttreatment and resolved. Mean NSAA total score increased from 20.5 to 27.5, baseline to year 4, with a mean (standard deviation) change of +7.0 (2.9). Post hoc analysis demonstrated a statistically significant and clinically meaningful 9-point difference in NSAA score, relative to a propensity-score-weighted external control cohort (least-squares mean [standard error] = 9.4 [3.4]; P = .0125). DISCUSSION Gene transfer therapy with delandistrogene moxeparvovec treatment is well tolerated, with a favorable safety profile. Functional improvements are sustained through 4 years, suggesting delandistrogene moxeparvovec may positively alter disease progression.
Collapse
Affiliation(s)
- Jerry R Mendell
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Zarife Sahenk
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
- Departments of Pathology and Neurology, The Ohio State University, Columbus, Ohio, USA
| | - Kelly J Lehman
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Linda P Lowes
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Natalie F Reash
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Megan A Iammarino
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Lindsay N Alfano
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Sarah Lewis
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Kathleen Church
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Richard Shell
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, Ohio, USA
| | | | | | - Mark Hogan
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Shufang Wang
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Stefanie Mason
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Eddie Darton
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | | |
Collapse
|
20
|
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
|
21
|
Servais L, Eggenspieler D, Poleur M, Grelet M, Muntoni F, Strijbos P, Annoussamy M. First regulatory qualification of a digital primary endpoint to measure treatment efficacy in DMD. Nat Med 2023; 29:2391-2392. [PMID: 37814063 DOI: 10.1038/s41591-023-02459-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Affiliation(s)
- Laurent Servais
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.
- Division of Child Neurology, Department of Pediatrics, Centre de Référence des Maladies Neuromusculaires, University Hospital Liège and University of Liège, Liège, Belgium.
| | | | - Margaux Poleur
- Department of Neurology, Centre de Référence des Maladies Neuromusculaires, University Hospital Liège and University of Liège, Liège, Belgium
| | | | - 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
| | | | | |
Collapse
|
22
|
Xu T, Xu K, Song Y, Zhou Z, Fu H, Xu R, Cai X, Guo Y, Ye P, Xu H. High-Speed T 2 -Corrected Multiecho Magnetic Resonance Spectroscopy for Quantitatively Detecting Skeletal Muscle Fatty Infiltration and Predicting the Loss of Ambulation in Patients With Duchenne Muscular Dystrophy. J Magn Reson Imaging 2023; 58:1270-1278. [PMID: 36773028 DOI: 10.1002/jmri.28641] [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: 11/23/2022] [Revised: 01/26/2023] [Accepted: 01/26/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND High-speed T2 -corrected multiecho MRS (HISTO-MRS) is emerging as a quantitative modality for detecting muscle fat infiltration (MFF). However, the predictive value of HISTO-MRS for the loss of ambulation (LoA) in Duchenne muscular dystrophy (DMD) is unknown. PURPOSE To determine the feasibility of HISTO-MRS for assessing MFF in DMD and further identify the predictive value of HISTO-MRS for the LoA. STUDY TYPE Prospective. SUBJECTS A total of 134 DMD boys (9.20 ± 2.43 years old) and 21 healthy boys (9.25 ± 2.10 years old). FIELD STRENGTH/SEQUENCE A 3 T, fast spin echo T1 -weighted imaging (T1 WI), two-point-Dixon gradient echo sequence (2-pt-Dixon) and HISTO-MRS. ASSESSMENT Subjective T1 WI fat grades by three radiologists, ROI analysis for MFF on 2 pt-Dixon (Dixon MFF) and MFF on HISTO-MRS (HISTO MFF) by two radiologists. Clinical motor function: North Star Ambulatory Assessment, 10-m run/walk time, Gowers maneuver, and time to four-stairs climb and descend. STATISTICAL TESTS Spearman rank correlation was used to assess the relation of fat filtration assessments and motor ability. Bland-Altman plots was performed to determine the agreement of HISTO MFF and Dixon MFF. Receiver operating characteristic (ROC) curves were analyzed to determine the discriminating ability of above MRI modalities for ambulatory and nonambulatory DMD. Logistic regression was used to identify the predictor of LoA. Variables with P < 0.05 in univariate logistic regression analysis were entered into the multivariate logistic regression model. RESULTS HISTO MFF was significantly correlated with Dixon MFF. Bland-Altman plots show good agreement of HISTO MFF and Dixon MFF. ROC curves indicated that HISTO MFF show similar discrimination of LoA for DMD with Dixon MFF but better value than T1WI fat grades. Logistic regression showed that HISTO MFF was an independent predictor for LoA. DATA CONCLUSION HISTO-MRS is a potential quantitative method for assessing fat infiltration and shows predictive value for LoA in DMD patients. LEVEL OF EVIDENCE: 1 TECHNICAL EFFICACY Stage 5.
Collapse
Affiliation(s)
- Ting Xu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Wuhou District, Chengdu, China
| | - Ke Xu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Wuhou District, Chengdu, China
| | - Yu Song
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Wuhou District, Chengdu, China
| | - Ziqi Zhou
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Wuhou District, Chengdu, China
| | - Hang Fu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Wuhou District, Chengdu, China
| | - Rong Xu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Wuhou District, Chengdu, China
| | - Xiaotang Cai
- Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, Wuhou District, Chengdu, China
| | - Yingkun Guo
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Wuhou District, Chengdu, China
| | - Pengfei Ye
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Wuhou District, Chengdu, China
| | - Huayan Xu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Wuhou District, Chengdu, China
| |
Collapse
|
23
|
Abstract
Delandistrogene moxeparvovec (delandistrogene moxeparvovec-rokl; ELEVIDYS®) is an adeno-associated virus (AAV) vector-based gene therapy designed to deliver a gene encoding a micro-dystrophin protein [i.e. a shortened (138 kDa) version of the dystrophin protein expressed in normal muscle cells (427 kDa)] to all muscles involved in the pathology of Duchenne muscular dystrophy (DMD). Developed by Sarepta Therapeutics, it is the first gene therapy to be approved (in June 2023 under the Accelerated Approval pathway) for the treatment of DMD in the USA, where it is indicated for ambulatory paediatric patients aged 4 through 5 years with DMD and a confirmed mutation in the dystrophin (DMD) gene. The recommended dose of delandistrogene moxeparvovec is 1.33 × 1014 vector genomes per kg of body weight or 10 mL/kg body weight, administered as a single intravenous infusion. Delandistrogene moxeparvovec is undergoing clinical development in several countries/regions, including the EU and Japan. This article summarizes the milestones in the development of delandistrogene moxeparvovec leading to this first approval in the USA for the treatment of ambulatory paediatric patients aged 4 through 5 years with DMD and a confirmed mutation in the DMD gene.
Collapse
Affiliation(s)
- Sheridan M Hoy
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
24
|
Roberts EK, Elliott MR, Taylor JMG. Solutions for surrogacy validation with longitudinal outcomes for a gene therapy. Biometrics 2023; 79:1840-1852. [PMID: 35833874 DOI: 10.1111/biom.13720] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 07/01/2022] [Indexed: 11/30/2022]
Abstract
Valid surrogate endpoints S can be used as a substitute for a true outcome of interest T to measure treatment efficacy in a clinical trial. We propose a causal inference approach to validate a surrogate by incorporating longitudinal measurements of the true outcomes using a mixed modeling approach, and we define models and quantities for validation that may vary across the study period using principal surrogacy criteria. We consider a surrogate-dependent treatment efficacy curve that allows us to validate the surrogate at different time points. We extend these methods to accommodate a delayed-start treatment design where all patients eventually receive the treatment. Not all parameters are identified in the general setting. We apply a Bayesian approach for estimation and inference, utilizing more informative prior distributions for selected parameters. We consider the sensitivity of these prior assumptions as well as assumptions of independence among certain counterfactual quantities conditional on pretreatment covariates to improve identifiability. We examine the frequentist properties (bias of point and variance estimates, credible interval coverage) of a Bayesian imputation method. Our work is motivated by a clinical trial of a gene therapy where the functional outcomes are measured repeatedly throughout the trial.
Collapse
Affiliation(s)
- Emily K Roberts
- Department of Biostatistics, University Michigan, Ann Arbor, Michigan, USA
| | - Michael R Elliott
- Department of Biostatistics, University Michigan, Ann Arbor, Michigan, USA
- Survey Methodology Program, Institute for Social Research, Ann Arbor, Michigan, USA
| | - Jeremy M G Taylor
- Department of Biostatistics, University Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
25
|
Wijekoon N, Gonawala L, Ratnayake P, Amaratunga D, Hathout Y, Mohan C, Steinbusch HWM, Dalal A, Hoffman EP, de Silva KRD. Duchenne Muscular Dystrophy from Brain to Muscle: The Role of Brain Dystrophin Isoforms in Motor Functions. J Clin Med 2023; 12:5637. [PMID: 37685704 PMCID: PMC10488491 DOI: 10.3390/jcm12175637] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/26/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023] Open
Abstract
Brain function and its effect on motor performance in Duchenne muscular dystrophy (DMD) is an emerging concept. The present study explored how cumulative dystrophin isoform loss, age, and a corticosteroid treatment affect DMD motor outcomes. A total of 133 genetically confirmed DMD patients from Sri Lanka were divided into two groups based on whether their shorter dystrophin isoforms (Dp140, Dp116, and Dp71) were affected: Group 1, containing patients with Dp140, Dp116, and Dp71 affected (n = 98), and Group 2, containing unaffected patients (n = 35). A subset of 52 patients (Group 1, n = 38; Group 2, n = 14) was followed for up to three follow-ups performed in an average of 28-month intervals. The effect of the cumulative loss of shorter dystrophin isoforms on the natural history of DMD was analyzed. A total of 74/133 (56%) patients encountered developmental delays, with 66/74 (89%) being in Group 1 and 8/74 (11%) being in Group 2 (p < 0.001). Motor developmental delays were predominant. The hip and knee muscular strength, according to the Medical Research Council (MRC) scale and the North Star Ambulatory Assessment (NSAA) activities, "standing on one leg R", "standing on one leg L", and "walk", declined rapidly in Group 1 (p < 0.001 In the follow-up analysis, Group 1 patients became wheelchair-bound at a younger age than those of Group 2 (p = 0.004). DMD motor dysfunction is linked to DMD mutations that affect shorter dystrophin isoforms. When stratifying individuals for clinical trials, considering the DMD mutation site and its impact on a shorter dystrophin isoform is crucial.
Collapse
Affiliation(s)
- Nalaka Wijekoon
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda 10250, Sri Lanka; (N.W.); (L.G.)
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6200 Maastricht, The Netherlands;
| | - Lakmal Gonawala
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda 10250, Sri Lanka; (N.W.); (L.G.)
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6200 Maastricht, The Netherlands;
| | | | | | - Yetrib Hathout
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13902, USA; (Y.H.); (E.P.H.)
| | - Chandra Mohan
- Department of Bioengineering, University of Houston, Houston, TX 77204, USA;
| | - Harry W. M. Steinbusch
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6200 Maastricht, The Netherlands;
| | - Ashwin Dalal
- Diagnostics Division, Center for DNA Fingerprinting and Diagnostics, Hyderabad 500039, India;
| | - Eric P. Hoffman
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13902, USA; (Y.H.); (E.P.H.)
| | - K. Ranil D. de Silva
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda 10250, Sri Lanka; (N.W.); (L.G.)
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6200 Maastricht, The Netherlands;
- Institute for Combinatorial Advanced Research and Education (KDU-CARE), General Sir John Kotelawala Defence University, Ratmalana 10390, Sri Lanka
| |
Collapse
|
26
|
Audhya I, Rogula B, Szabo SM, Feeny D, Bolatova T, Gooch K. Exploring the relationship between North Star Ambulatory Assessment and Health Utilities Index scores in Duchenne muscular dystrophy. Health Qual Life Outcomes 2023; 21:76. [PMID: 37468890 PMCID: PMC10355009 DOI: 10.1186/s12955-023-02160-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND The North Star Ambulatory Assessment (NSAA) documents motor performance in ambulatory individuals with Duchenne muscular dystrophy (DMD). Health Utilities Index (HUI) scores, reflecting preferences for health-related quality-of-life (HRQoL) implications of health states, are commonly estimated within trials. This study sought to characterize the relationship between the NSAA score and utility in DMD. METHODS Family members serving as proxy respondents for placebo-treated ambulatory individuals with DMD (NCT01254019; BioMarin Pharmaceuticals Inc) completed the HUI and the NSAA (score range, 0-34). Mean change over time on these measures was estimated, and the correlation between changes in NSAA score and a) HUI utility; b) HUI3 ambulation and HUI2 mobility attribute scores, over 48 weeks was calculated. RESULTS Baseline mean (range) age was 8.0 years (5-16; n = 61) and mean (standard deviation [SD]) scores were 0.87 (0.13; HUI2), 0.82 (0.19; HUI3), and 21.0 (8.1; NSAA). Mean (SD) change over 48 weeks was -0.05 (0.14; HUI2), -0.06 (0.19; HUI3), and -2.9 (4.7; NSAA). Weak positive correlations were observed between baseline NSAA score and HUI utility (HUI2: r = 0.29; HUI3: r = 0.17) and for change over 48 weeks (HUI2: r = 0.16; HUI3: r = 0.15). Stronger correlations were observed between change in NSAA score and the HUI3 ambulation (r = 0.41) and HUI2 mobility (r = 0.41) attributes. CONCLUSIONS Among ambulatory individuals with DMD, NSAA score is weakly correlated with HUI utility, suggesting that motor performance alone does not fully explain HRQoL. Stronger relationships were observed between HUI ambulation and mobility attributes, and NSAA. Although unidimensional measures like the NSAA are informative for documenting disease-specific health impacts, they may not correlate well with measures of overall health status; requiring use in conjunction with other patient-reported and preference-based outcomes.
Collapse
Affiliation(s)
| | - Basia Rogula
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, V6A 1A6, Canada
| | - Shelagh M Szabo
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, V6A 1A6, Canada.
| | - David Feeny
- McMaster University and Health Utilities Inc, Hamilton, ON, Canada
| | - Talshyn Bolatova
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, V6A 1A6, Canada
| | | |
Collapse
|
27
|
Mendell JR, Shieh PB, McDonald CM, Sahenk Z, Lehman KJ, Lowes LP, Reash NF, Iammarino MA, Alfano LN, Sabo B, Woods JD, Skura CL, Mao HC, Staudt LA, Griffin DA, Lewis S, Wang S, Potter RA, Singh T, Rodino-Klapac LR. Expression of SRP-9001 dystrophin and stabilization of motor function up to 2 years post-treatment with delandistrogene moxeparvovec gene therapy in individuals with Duchenne muscular dystrophy. Front Cell Dev Biol 2023; 11:1167762. [PMID: 37497476 PMCID: PMC10366687 DOI: 10.3389/fcell.2023.1167762] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/09/2023] [Indexed: 07/28/2023] Open
Abstract
Introduction: Delandistrogene moxeparvovec (SRP-9001) is an investigational gene transfer therapy designed for targeted expression of SRP-9001 dystrophin protein, a shortened dystrophin retaining key functional domains of the wild-type protein. Methods: This Phase 2, double-blind, two-part (48 weeks per part) crossover study (SRP-9001-102 [Study 102]; NCT03769116) evaluated delandistrogene moxeparvovec in patients, aged ≥4 to <8 years with Duchenne muscular dystrophy. Primary endpoints (Part 1) were change from baseline (CFBL) in SRP-9001 dystrophin expression (Week 12), by Western blot, and in North Star Ambulatory Assessment (NSAA) score (Week 48). Safety assessments included treatment-related adverse events (TRAEs). Patients were randomized and stratified by age to placebo (n = 21) or delandistrogene moxeparvovec (n = 20) and crossed over for Part 2. Results: SRP-9001 dystrophin expression was achieved in all patients: mean CFBL to Week 12 was 23.82% and 39.64% normal in Parts 1 and 2, respectively. In Part 1, CFBL to Week 48 in NSAA score (least-squares mean, LSM [standard error]) was +1.7 (0.6) with treatment versus +0.9 (0.6) for placebo; p = 0.37. Disparity in baseline motor function between groups likely confounded these results. In 4- to 5-year-olds with matched baseline motor function, CFBL to Week 48 in NSAA scores was significantly different (+2.5 points; p = 0.0172), but not significantly different in 6-to-7-year-olds with imbalanced baseline motor function (-0.7 points; p = 0.5384). For patients treated with delandistrogene moxeparvovec in Part 2, CFBL to Week 48 in NSAA score was +1.3 (2.7), whereas for those treated in Part 1, NSAA scores were maintained. As all patients in Part 2 were exposed to treatment, results were compared with a propensity-score-weighted external control (EC) cohort. The LSM difference in NSAA score between the Part 2 treated group and EC cohort was statistically significant (+2.0 points; p = 0.0009). The most common TRAEs were vomiting, decreased appetite, and nausea. Most occurred within the first 90 days and all resolved. Discussion: Results indicate robust expression of SRP-9001 dystrophin and overall stabilization in NSAA up to 2 years post-treatment. Differences in NSAA between groups in Part 1 were not significant for the overall population, likely because cohorts were stratified only by age, and other critical prognostic factors were not well matched at baseline.
Collapse
Affiliation(s)
- Jerry R. Mendell
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
- Department of Neurology, The Ohio State University, Columbus, OH, United States
| | | | - Craig M. McDonald
- Departments of Physical Medicine and Rehabilitation and Pediatrics, Lawrence J. Ellison Ambulatory Care Center, UC Davis Health, Sacramento, CA, United States
| | - Zarife Sahenk
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
- Department of Neurology, The Ohio State University, Columbus, OH, United States
| | - Kelly J. Lehman
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Linda P. Lowes
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| | - Natalie F. Reash
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Megan A. Iammarino
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Lindsay N. Alfano
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Brenna Sabo
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
| | | | | | | | | | | | - Sarah Lewis
- Sarepta Therapeutics Inc, Cambridge, MA, United States
| | - Shufang Wang
- Sarepta Therapeutics Inc, Cambridge, MA, United States
| | | | - Teji Singh
- Sarepta Therapeutics Inc, Cambridge, MA, United States
| | | |
Collapse
|
28
|
Klimchak AC, Sedita LE, Rodino-Klapac LR, Mendell JR, McDonald CM, Gooch KL, Malone DC. Assessing the value of delandistrogene moxeparvovec (SRP-9001) gene therapy in patients with Duchenne muscular dystrophy in the United States. JOURNAL OF MARKET ACCESS & HEALTH POLICY 2023; 11:2216518. [PMID: 37261034 PMCID: PMC10228300 DOI: 10.1080/20016689.2023.2216518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/02/2023]
Abstract
Background: Delandistrogene moxeparvovec (SRP-9001) is an investigational gene therapy that may delay progression of Duchenne muscular dystrophy (DMD), a severe, rare neuromuscular disease caused by DMD gene mutations. Early cost-effectiveness analyses are important to help contextualize the value of gene therapies for reimbursement decision making. Objective: To determine the potential value of delandistrogene moxeparvovec using a cost-effectiveness analysis. Study design: A simulation calculated lifetime costs and equal value of life years gained (evLYG). Inputs included extrapolated clinical trial results and published utilities/costs. As a market price for delandistrogene moxeparvovec has not been established, threshold analyses established maximum treatment costs as they align with value, including varying willingness-to-pay up to $500,000, accounting for severity/rarity. Setting: USA, healthcare system perspective Patients: Boys with DMD Intervention: Delandistrogene moxeparvovec plus standard of care (SoC; corticosteroids) versus SoC alone Main outcome measure: Maximum treatment costs at a given willingness-to-pay threshold Results: Delandistrogene moxeparvovec added 10.30 discounted (26.40 undiscounted) evLYs. The maximum treatment cost was approximately $5 M, assuming $500,000/evLYG. Varying the benefit discount rate to account for the single administration increased the estimated value to #$5M, assuming $500,000/evLYG. Conclusion: In this early economic model, delandistrogene moxeparvovec increases evLYs versus SoC and begins to inform its potential value from a healthcare perspective.
Collapse
Affiliation(s)
- Alexa C. Klimchak
- Global HEOR, RWE & Analytics, Sarepta Therapeutics, Inc, Cambridge, MA, USA
| | - Lauren E. Sedita
- Global HEOR, RWE & Analytics, Sarepta Therapeutics, Inc, Cambridge, MA, USA
| | | | - Jerry R. Mendell
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics and Neurology, The Ohio State University, Columbus, OH, USA
| | - Craig M. McDonald
- Department of Pediatrics, University of California Davis School of Medicine, Davis, CA, USA
| | | | - Daniel C. Malone
- College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
29
|
Signorelli M, Tsonaka R, Aartsma-Rus A, Spitali P. Multiomic characterization of disease progression in mice lacking dystrophin. PLoS One 2023; 18:e0283869. [PMID: 37000843 PMCID: PMC10065259 DOI: 10.1371/journal.pone.0283869] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/19/2023] [Indexed: 04/03/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by genetic mutations leading to lack of dystrophin in skeletal muscle. A better understanding of how objective biomarkers for DMD vary across subjects and over time is needed to model disease progression and response to therapy more effectively, both in pre-clinical and clinical research. We present an in-depth characterization of disease progression in 3 murine models of DMD by multiomic analysis of longitudinal trajectories between 6 and 30 weeks of age. Integration of RNA-seq, mass spectrometry-based metabolomic and lipidomic data obtained in muscle and blood samples by Multi-Omics Factor Analysis (MOFA) led to the identification of 8 latent factors that explained 78.8% of the variance in the multiomic dataset. Latent factors could discriminate dystrophic and healthy mice, as well as different time-points. MOFA enabled to connect the gene expression signature in dystrophic muscles, characterized by pro-fibrotic and energy metabolism alterations, to inflammation and lipid signatures in blood. Our results show that omic observations in blood can be directly related to skeletal muscle pathology in dystrophic muscle.
Collapse
Affiliation(s)
- Mirko Signorelli
- Mathematical Institute, Leiden University, Leiden, The Netherlands
| | - Roula Tsonaka
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Pietro Spitali
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
30
|
Hibma JE, Jayachandran P, Neelakantan S, Harnisch LO. Disease progression modeling of the North Star Ambulatory Assessment for Duchenne Muscular Dystrophy. CPT Pharmacometrics Syst Pharmacol 2023; 12:375-386. [PMID: 36718719 PMCID: PMC10014057 DOI: 10.1002/psp4.12921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 02/01/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a rare genetic disorder caused by decreased or absent dystrophin gene leading to progressive muscle degeneration and weakness in young boys. Disease progression models for the North Star Ambulatory Assessment (NSAA), a functional measurement widely used to assess outcomes in clinical trials, were developed using a longitudinal population modeling approach. The relationship between NSAA total score over time, loss of ambulation, and potential covariates that may influence disease progression were evaluated. Data included individual participant observations from an internal placebo-controlled phase II clinical trial and from the external natural history database for male patients with DMD obtained through the Cooperative International Neuromuscular Research Group (CINRG). A modified indirect response model for NSAA joined to a loss of ambulation (LOA) time-to-event model described the data well. Age was used as the independent variable because ambulatory function is known to vary with age. The NSAA and LOA models were linked using the dissipation rate constant parameter from the NSAA model by including the parameter as a covariate on the hazard equation for LOA. No covariates were identified. The model was then used as a simulation tool to explore various clinical trial design scenarios. This model contributes to the quantitative understanding of disease progression in DMD and may guide model-informed drug development decisions for ongoing and future DMD clinical trials.
Collapse
Affiliation(s)
- Jennifer E Hibma
- Global Product Development, Pfizer Inc., La Jolla, California, USA
| | | | | | | |
Collapse
|
31
|
Peng F, Xu H, Song Y, Xu K, Li S, Cai X, Guo Y, Gong L. Longitudinal study of multi-parameter quantitative magnetic resonance imaging in Duchenne muscular dystrophy: hyperresponsiveness of gluteus maximus and detection of subclinical disease progression in functionally stable patients. J Neurol 2023; 270:1439-1451. [PMID: 36385201 DOI: 10.1007/s00415-022-11470-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To describe the disease progression of Duchenne muscular dystrophy (DMD) in the pelvic and thigh muscles over 1-year using multiple-parameter quantitative magnetic resonance imaging (qMRI), and to determine the most responsive muscle and predict subclinical disease progression in functionally stable patients. METHODS Fifty-four DMD patients (mean age 8.9 ± 2.5, range 5-15 years) completed baseline and 1-year follow-up qMRI examinations/biomarkers [3-point Dixon/fat fraction (FF); T1 mapping/T1; T2 mapping/T2]. Meanwhile, clinical assessments [NorthStar ambulatory assessment (NSAA) score] and timed function tests were performed in DMD patients. Twenty-four healthy male controls (range 5-15 years) accomplished baseline qMRI examinations. Group differences were compared using the Wilcoxon test. The standardized response mean (SRM) was taken as the responsiveness to the disease progression index. RESULTS FF, T1, and T2 in all DMD age subgroups changed significantly over 1-year (P < 0.05). Even in functionally stable patients (NSAA score increased, unchanged, or decreased by 1-point) over 1-year, significant increases in FF and T2 and decreases in T1 were observed in gluteus maximus (GMa), gluteus medius, vastus lateralis, and adductor magnus (P < 0.05). Overall, the SRM of FF, T1, and T2 was all the highest in GMa, which were 1.25, - 0.92, and 0.93, respectively. CONCLUSIONS qMRI biomarkers are responsive to disease progression and can also detect subclinical disease progression in functionally stable DMD patients over 1-year. GMa is the most responsive to disease progression of all the muscles analyzed. TRIAL REGISTRATION Chinese Clinical Trial Registry ( http://www.chictr.org.cn/index.aspx ) ChiCTR1800018340, 09/12/2018, prospectively registered.
Collapse
Affiliation(s)
- Fei Peng
- Department of Medical Imaging Center, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi Province, China
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Huayan Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Yu Song
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Ke Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Shuhao Li
- Department of Medical Imaging Center, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi Province, China
| | - Xiaotang Cai
- Department of Pediatrics Neurology, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China.
| | - Yingkun Guo
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China.
| | - Lianggeng Gong
- Department of Medical Imaging Center, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
32
|
Fang Y, McDonald CM, Clemens PR, Gordish HD, Illei K, Hoffman EP, Dang UJ. Modeling Early Heterogeneous Rates of Progression in Boys with Duchenne Muscular Dystrophy. J Neuromuscul Dis 2023; 10:349-364. [PMID: 36806514 DOI: 10.3233/jnd-221527] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) exhibits substantial variability in rates of disease progression and response to treatment. This has hindered treatment development and complicated interpretation of drug effects in clinical trials. OBJECTIVE We hypothesized that a multivariate combination of early-age clinical outcome measurements can explain differential disease progression. METHODS Data on boys with DMD (ages 4-<10 years), both treated with steroidal anti-inflammatories and untreated, were obtained from CINRG Duchenne Natural History Study (n = 209) and vamorolone VBP15-002/003/LTE (n = 46) studies. Velocities from three timed function tests (TFTs; stand from supine, run/walk 10 meters, and climb 4 stairs) were simultaneously modeled in a longitudinal latent class analysis. RESULTS Three classes of differentially progressing early age DMD motor trajectories were identified. Quicker decline/progression was associated with lower baseline TFT velocities, earlier loss of ability to finish a TFT, and lower predicted velocities. Earlier substantial steroid exposure was associated with greater TFT velocities while the moderate progression class was observed to have the largest difference in performance between boys treated early with steroids vs. not. Sample size calculations with the class showing the largest treatment response showed a large reduction in required sample size as compared to using summaries from all participants. Gene mutations were also investigated in post-hoc analyses, with mutations near the beginning of the DMD gene (Dp427 absent and Dp140/Dp71 present) found to be enriched in the slowest progressing class. CONCLUSIONS This study provides insight into the variation in DMD progression through a latent class analysis. Our findings show class-related trajectories of motor outcomes and pharmacological response to corticosteroids, and suggest that enrichment strategies and/or subgroup analyses could be considered further in design of therapeutic interventions in DMD.
Collapse
Affiliation(s)
- Yuan Fang
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Craig M McDonald
- University of California Davis School of Medicine, Sacramento, CA, USA
| | - Paula R Clemens
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Veteran Affairs Medical Center, Pittsburgh, PA, USA
| | | | | | - Eric P Hoffman
- ReveraGen BioPharma, Rockville, MD, USA.,Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University-SUNY, Binghamton, NY, USA
| | | | - Utkarsh J Dang
- Department of Health Sciences, Carleton University, Ottawa, Canada
| |
Collapse
|
33
|
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
|
34
|
Muntoni F, Guglieri M, Mah JK, Wagner KR, Brandsema JF, Butterfield RJ, McDonald CM, Mayhew AG, Palmer JP, Marraffino S, Charnas L, Mercuri E. Novel approaches to analysis of the North Star Ambulatory Assessment (NSAA) in Duchenne muscular dystrophy (DMD): Observations from a phase 2 trial. PLoS One 2022; 17:e0272858. [PMID: 35998119 PMCID: PMC9397979 DOI: 10.1371/journal.pone.0272858] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 07/22/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction The North Star Ambulatory Assessment (NSAA) tool is a key instrument for measuring clinical outcomes in patients with Duchenne muscular dystrophy (DMD). To gain a better understanding of the longitudinal utility of the NSAA, we evaluated NSAA data from a phase II trial of 120 patients with DMD treated with domagrozumab or placebo. Methods The NSAA exploratory analyses included assessment of individual skills gained/lost, total skills gained/lost, cumulative loss of function, and the impact of transient loss of function due to a temporary disability on NSAA total score (temporary zero score). Results There was no significant difference in the total number of NSAA skills gained (mean 1.41 and 1.04, respectively; p = 0.3314) or lost (3.90 vs. 5.0; p = 0.0998) between domagrozumab- vs. placebo-treated patients at week 49. However, domagrozumab-treated patients were less likely to lose the ability to perform a NSAA item (hazard ratio 0.80, 95% confidence interval [CI]: 0.65–0.98, p = 0.029) over 48-weeks vs. placebo-treated patients. When temporary zero scores were changed to “not obtainable” (8 values from 7 patients), domagrozumab-treated patients scored higher on the NSAA total score versus placebo-treated patients (difference at week 49: 2.0, 95% CI: 0.1–3.9, p = 0.0359). Conclusions These exploratory analyses reveal additional approaches to interpreting the NSAA data beyond just change in NSAA total score. These observations also highlight the importance of reporting items as “not obtainable” for a patient with a temporary/transient physical disability that impacts their ability to perform the NSAA test. ClinicalTrials.gov identifier NCT02310763.
Collapse
Affiliation(s)
- Francesco Muntoni
- 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
- * E-mail:
| | - Michela Guglieri
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Jean K. Mah
- Cumming School of Medicine, University of Calgary, Alberta Children’s Hospital, Calgary, Alberta, Canada
| | - Kathryn R. Wagner
- Center for Genetic Muscle Disorders, and Departments of Neurology and Neuroscience, Kennedy Krieger Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - John F. Brandsema
- Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | | | - Craig M. McDonald
- Lawson Health Research Institute, Children’s Hospital, London, Ontario, Canada
| | - Anna G. Mayhew
- Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom
| | | | | | | | - Eugenio Mercuri
- Paediatric Neurology, Catholic University, and Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
35
|
Zambon AA, Trucco F, Laverty A, Riley M, Ridout D, Manzur AY, Abel F, Muntoni F. Respiratory Function and Sleep Disordered Breathing in Pediatric Duchenne Muscular Dystrophy. Neurology 2022; 99:e1216-e1226. [PMID: 35953292 DOI: 10.1212/wnl.0000000000200932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 05/19/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The decline of respiratory function in Duchenne muscular dystrophy (DMD) is associated with sleep disordered breathing (SDB) and alteration of nocturnal gas exchange, first manifesting as nocturnal hypoventilation (NH). However, the correlation between pulmonary function measured by spirometry (PFT) and the onset of SDB with or without NH is unclear. AIM To identify the prevalence and features of SDB and to investigate the relationship between lung function determined by forced vital capacity (FVC) and sleep abnormalities in a large paediatric DMD population. METHODS Retrospective, single-center cohort study. FVC% predicted (FVC%) was calculated using predicted equations from the Global Lung Function Initiative. NH was defined by transcutaneous (tc) CO2 >50 mmHg for >25% of total sleep time (TST); borderline NH by a mean tcCO2 between 45-50mmHg or tcCO2>50mmHg for ≤25% of TST; Clinically meaningful obstructive sleep apnea (OSA) by obstructive Apnea-Hypopnea Index >5. The sensitivity, specificity, positive and negative predictive value of FVC<50% to indicate presence of nocturnal hypoventilation were calculated. RESULTS 134 patients underwent 284 sleep studies and 1222 PFT. Mean (SD) age at first and last sleep study was 12.9 (2.7) and 14.3 (2.6) years. Borderline NH (n=31) was detected in both ambulant and early-non ambulant subjects, while 100% of NH cases (n=14) were non-ambulant. NH was detected in 4/14 patients despite an FVC>50%. Seventeen/26 patients with OSA presented with concomitant NH or borderline NH. FVC<50% was associated with NH indicating a sensitivity and specificity of 73% and 86%, respectively. Positive and negative predictive value were 32% and 97% respectively. PFT showed a non-linear, sudden FVC% decline in 18% of cases. CONCLUSIONS FVC% <50 was associated with NH in close to a third of patients. CO2 elevation can be associated with obstructive/pseudo-obstructive events and was also observed in early non-ambulant cases or in the presence of FVC>50%. These results are relevant for the clinical management of SDB.
Collapse
Affiliation(s)
- Alberto A Zambon
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK.,Neuromuscular Repair Unit, Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Federica Trucco
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK.,Children's Sleep Medicine, Evelina Children Hospital - Paediatric Respiratory Department Royal Brompton Hospital, Guy's and St Thomas' Trust, London, UK
| | - Aidan Laverty
- Department of Paediatric Respiratory Medicine, Great Ormond Street Hospital & UCL Institute of Child Health, London, UK
| | - Mollie Riley
- Department of Paediatric Respiratory Medicine, Great Ormond Street Hospital & UCL Institute of Child Health, London, UK
| | - Deborah Ridout
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK.,Population, Policy and Practice Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Adnan Y Manzur
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Francois Abel
- Children's Sleep Medicine, Evelina Children Hospital - Paediatric Respiratory Department Royal Brompton Hospital, Guy's and St Thomas' Trust, London, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK .,NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| |
Collapse
|
36
|
Zambon AA, Ayyar Gupta V, Ridout D, Manzur A, Baranello G, Trucco F, Muntoni F, The UK Northstar Clinical Network TirupathSandyaDouglasMelanieMcFetridgeJaciParasuramanDeepakAlhaswaniZoyaMcMurchieHeatherRabbRosannaMajumdarAnirbanVijayakumarKayalAminSamMasonFayeFrimpong‐AnsahClaireGibbonFrancesParsonBethanNaismithKarenBurslemJulieBaxterAlexEadieClareHorrocksIainDi MarcoMarinaChildsAnne‐MariePallantLindseySpintyStefanShillingtonAlisonGregsonSarahCheshmanLauraWraigeElizabethGowdaVasanthaJungbluthHeinzSheehanJennieHughesImeldaWarnerSineadStraubVolkerGuglieriMichelaMayhewAnnaChowGabbyWilliamsonSarahWillisTraceyKulshresthaRichaEmeryNicholasRamdasSitharaRamjattanHayleyde GoedeChristianSelleyAndreaOngMinWhiteKayIllingworthMarjorieGearyMichellePalmerJenniWhiteCathyGreenfieldKateHewawitharanaGemunuJulienYvonneStephensElmaTewnionJaneAmbegaonkarGautamKrishnakumarDeepaTaylorJacquiWardCatherineWillisTraceyWrightElizabethRylanceClaire, 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
|
37
|
Coratti G, Lenkowicz J, Norcia G, Lucibello S, Ferraroli E, d’Amico A, Bello L, Pegoraro E, Messina S, Ricci F, Mongini T, Berardinelli A, Masson R, Previtali SC, D’angelo G, Magri F, Comi GP, Politano L, Passamano L, Vita G, Sansone VA, Albamonte E, Panicucci C, Bruno C, Pini A, Bertini E, Patarnello S, Pane M, Mercuri E, for the italian DMD study group. Age, corticosteroid treatment and site of mutations affect motor functional changes in young boys with Duchenne Muscular Dystrophy. PLoS One 2022; 17:e0271681. [PMID: 35905042 PMCID: PMC9337636 DOI: 10.1371/journal.pone.0271681] [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: 02/02/2022] [Accepted: 07/05/2022] [Indexed: 11/18/2022] Open
Abstract
The aim of this study was to establish the possible effect of age, corticosteroid treatment and brain dystrophin involvement on motor function in young boys affected by Duchenne Muscular Dystrophy who were assessed using the North Star Ambulatory Assessment between the age of 4 and 7 years. The study includes 951 North Star assessments from 226 patients. Patients were subdivided according to age, to the site of mutation and therefore to the involvement of different brain dystrophin isoforms and to corticosteroids duration. There was a difference in the maximum North Star score achieved among patients with different brain dystrophin isoforms (p = 0.007). Patients with the involvement of Dp427, Dp140 and Dp71, had lower maximum NSAA scores when compared to those with involvement of Dp427 and Dp140 or of Dp427 only. The difference in the age when the maximum score was achieved in the different subgroups did not reach statistical significance. Using a linear regression model on all assessments we found that each of the three variables, age, site of mutation and corticosteroid treatment had an influence on the NSAA values and their progression over time. A second analysis, looking at 12-month changes showed that within this time interval the magnitude of changes was related to corticosteroid treatment but not to site of mutation. Our findings suggest that each of the considered variables appear to play a role in the progression of North Star scores in patients between the age of 4 and 7 years and that these should be carefully considered in the trial design of boys in this age range.
Collapse
Affiliation(s)
- 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
| | - Jacopo Lenkowicz
- Fondazione Policlinico Universitario A.Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giulia Norcia
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Simona Lucibello
- 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
| | - Elisabetta Ferraroli
- 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
| | - Adele d’Amico
- Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Luca Bello
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Sonia Messina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Federica Ricci
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Turin, Torino, Italy
| | - Tiziana Mongini
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Turin, Torino, Italy
| | | | - Riccardo Masson
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | | | - Francesca Magri
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Giacomo P. Comi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Luisa Politano
- Cardiomyology and Medical Genetics, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Luigia Passamano
- Cardiomyology and Medical Genetics, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gianluca Vita
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Valeria A. Sansone
- The NEMO Center in Milan, Neurorehabilitation Unit, ASST Niguarda Hospital, University of Milan, Milan, Italy
| | - Emilio Albamonte
- The NEMO Center in Milan, Neurorehabilitation Unit, ASST Niguarda Hospital, University of Milan, Milan, Italy
| | - Chiara Panicucci
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, and Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health—DINOGMI, University of Genova, Genoa, Italy
| | - Claudio Bruno
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, and Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health—DINOGMI, University of Genova, Genoa, Italy
| | - Antonella Pini
- Neuromuscular Pediatric Unit, UOC di Neuropsichiatria dell’età pediatrica, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Enrico Bertini
- Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Stefano Patarnello
- Fondazione Policlinico Universitario A.Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marika Pane
- 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
| | - 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
- * E-mail:
| | | |
Collapse
|
38
|
Peng F, Xu H, Song Y, Xu K, Li S, Cai X, Guo Y, Gong L. Utilization of T1-Mapping for the pelvic and thigh muscles in Duchenne Muscular Dystrophy: a quantitative biomarker for disease involvement and correlation with clinical assessments. BMC Musculoskelet Disord 2022; 23:681. [PMID: 35842609 PMCID: PMC9288085 DOI: 10.1186/s12891-022-05640-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 07/08/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Little is known about the disease distribution and severity detected by T1-mapping in Duchenne muscular dystrophy (DMD). Furthermore, the correlation between skeletal muscle T1-values and clinical assessments is less studied. Hence, the purposes of our study are to investigate quantitative T1-mapping in detecting the degree of disease involvement by detailed analyzing the hip and thigh muscle, future exploring the predicting value of T1-mapping for the clinical status of DMD. METHODS Ninety-two DMD patients were included. Grading fat infiltration and measuring the T1-values of 19 pelvic and thigh muscles (right side) in axial T1-weighted images (T1WI) and T1-maps, respectively, the disease distribution and severity were evaluated and compared. Clinical assessments included age, height, weight, BMI, wheelchair use, timed functional tests, NorthStar ambulatory assessment (NSAA) score, serum creatine kinase (CK) level. Correlation analysis were performed between the muscle T1-value and clinical assessments. Multiple linear regression analysis was conducted for the independent association of T1-value and motor function. RESULTS The gluteus maximus had the lowest T1-value, and the gracilis had the highest T1-value. T1-value decreased as the grade of fat infiltration increased scored by T1WI (P < 0.001). The decreasing of T1-values was correlated with the increase of age, height, weight, wheelchair use, and timed functional tests (P < 0.05). T1-value correlated with NSAA (r = 0.232-0.721, P < 0.05) and CK (r = 0.208-0.491, P < 0.05) positively. T1-value of gluteus maximus, tensor fascia, vastus lateralis, vastus intermedius, vastus medialis, and adductor magnus was independently associated with the clinical motor function tests (P < 0.05). Interclass correlation coefficient (ICC) analysis and Bland-Altman plots showed excellent inter-rater reliability of T1-value region of interest (ROI) measurements. CONCLUSION T1-mapping can be used as a quantitative biomarker for disease involvement, further assessing the disease severity and predicting motor function in DMD.
Collapse
Affiliation(s)
- Fei Peng
- Department of Medical Imaging center, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi Province, China
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Huayan Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Yu Song
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Ke Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Shuhao Li
- Department of Medical Imaging center, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi Province, China
| | - Xiaotang Cai
- Department of Pediatrics Neurology, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China.
| | - Yingkun Guo
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China.
| | - Lianggeng Gong
- Department of Medical Imaging center, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
39
|
Emerging therapies for Duchenne muscular dystrophy. Lancet Neurol 2022; 21:814-829. [DOI: 10.1016/s1474-4422(22)00125-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 02/21/2022] [Accepted: 03/18/2022] [Indexed: 12/11/2022]
|
40
|
Iff J, Zhong Y, Gupta D, Paul X, Tuttle E, Henricson E, Schrader R. Disease Progression Stages and Burden in Patients with Duchenne Muscular Dystrophy Using Administrative Claims Supplemented by Electronic Medical Records. Adv Ther 2022; 39:2906-2919. [PMID: 35460510 DOI: 10.1007/s12325-022-02117-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/07/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION This study aims to identify stages of Duchenne muscular dystrophy (DMD) and assess the disease burden by progression stage using real-world administrative claims supplemented by relevant electronic medical record (EMR) data. METHODS Claims and EMR data from the Decision Resources Group's Real World Data Repository (2011-2020) were used to identify patients with DMD by diagnosis code and to stratify them into four disease stages by diagnosis and procedure markers reflective of DMD progression. Clinical and medical history data from the Cooperative International Neuromuscular Research Group (CINRG) were used to validate the developed claims-based staging algorithm. The distribution and drivers by disease stage, as well as disease burden, were examined. RESULTS A total of 938 (94%) of patients with DMD identified in claims/EMR data had sufficient information for stage classification. Patients were classified by stage based on patient characteristics and the presence or absence of progression markers such as genetic testing, wheelchair usage, scoliosis treatment, or ventilation assistance. Average ages at stages 1-4 are 7, 13, 18, and 23 years, respectively. Using natural history data, the claims-based staging algorithm was validated with high sensitivity and specificity rates. Both healthcare resource utilization and medical charges increased by stage. For example, the average annualized total charges were $17,688 (stage 1), $36,868 (stage 2), $72,801 (stage 3), and $167,285 (stage 4). CONCLUSIONS Large-scale claims data supplemented by EMR data can be used to characterize DMD progression and evaluate disease burden which may inform the design of future real-world studies about DMD.
Collapse
Affiliation(s)
- Joel Iff
- Sarepta Therapeutics, Inc, Cambridge, MA, USA
| | - Yi Zhong
- Analysis Group, Inc., 1010 El Camino Real #310, Menlo Park, CA, 94025, USA.
| | - Deepshekhar Gupta
- Analysis Group, Inc., 1010 El Camino Real #310, Menlo Park, CA, 94025, USA
| | - Xander Paul
- Analysis Group, Inc., 1010 El Camino Real #310, Menlo Park, CA, 94025, USA
| | - Edward Tuttle
- Analysis Group, Inc., 1010 El Camino Real #310, Menlo Park, CA, 94025, USA
| | | | | |
Collapse
|
41
|
Zamani G, Hosseinpour S, Ashrafi MR, Mohammadi M, Badv RS, Tavasoli AR, Akbari MG, Bereshneh AH, Malamiri RA, Heidari M. Characteristics of disease progression and genetic correlation in ambulatory Iranian boys with Duchenne muscular dystrophy. BMC Neurol 2022; 22:162. [PMID: 35501714 PMCID: PMC9059913 DOI: 10.1186/s12883-022-02687-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 04/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy in the pediatric population. The manifestations of this disease include progressive muscle weakness, gait dysfunction, and motor impairment, leading to a loss of ambulation by the age of 13 years. Molecular diagnosis is the standard diagnostic tool for DMD. This study aimed to investigate disease progression and genetic patterns in Iranian ambulant boys and to find the correlation between genotypes and motor function phenotypes. METHODS This study was performed on 152 DMD patients. Clinical history, including the disease phenotype, steroid therapy, and the North Star Ambulatory Assessment (NSAA) score, was taken for all the patients. Molecular diagnoses were confirmed by multiplex ligation-dependent probe amplification and next-generation sequencing tests. RESULTS A total of 152 Iranian DMD patients were examined in this study. The mean age at the time of disease onset was 4.04 ± 2.00 years, and the mean age at diagnosis was 5.05 ± 2.08 years. The mean age of ambulation loss was 10.9 years. Contracture was reported in 38.9% of cases. In terms of age, the mean total NSAA score showed a peak at 4 years of age, with a mean NSAA score of 24. Annual changes in the NSAA score were determined for all cases, based on the mutation type and exon site. Deletion mutation was found in 79.1% of cases, duplication in 6.8%, nonsense in 12.8%, and splice site in 1.4%. The most common single exon deletion was exon 44 (5.3%), and the most common multiexon deletions were attributed to exons 45-50 and exons 45-52 (4.6%). The results did not indicate any correlation between the mutation type and age at the time of disease onset, loss of ambulation age, and wheelchair dependence; however, a significant association was found between contracture and mutation type. The results showed a significant difference in the NSAA score between the deletion and nonsense groups at the age of 3 years (P = 0.04). No significant correlation was found between the phenotype and exon site. Overall, 91.1% of the study population had a history of corticosteroid use, and 54.1% showed compliance with rehabilitation therapy. CONCLUSION This study demonstrated the phenotypes and mutational features of Iranian DMD boys and provided information regarding the natural motor history of the disease, disease progression, diagnosis, and status of DMD management in Iran. The present findings can promote the development of clinical trials and future advanced molecular therapies in Iran.
Collapse
Affiliation(s)
- Gholamreza Zamani
- Pediatrics Center of Excellence, Department of Pediatric Neurology, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sareh Hosseinpour
- Department of Pediatric Neurology, Vali-e-Asr Hospital, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Reza Ashrafi
- Pediatrics Center of Excellence, Department of Pediatric Neurology, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Mohammadi
- Pediatrics Center of Excellence, Department of Pediatric Neurology, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Shervin Badv
- Pediatrics Center of Excellence, Department of Pediatric Neurology, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Reza Tavasoli
- Pediatrics Center of Excellence, Department of Pediatric Neurology, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Masood Ghahvechi Akbari
- Physical Medicine and Rehabilitation Department, Children's Medical Center , Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Hosseini Bereshneh
- Prenatal Diagnosis and Genetic Research Center, Dastgheib Hospital, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Reza Azizi Malamiri
- Department of Pediatric Neurology, Golestan Medical, Educational, and Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Morteza Heidari
- Pediatrics Center of Excellence, Department of Pediatric Neurology, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
42
|
Chesshyre M, Ridout D, Hashimoto Y, Ookubo Y, Torelli S, Maresh K, Ricotti V, Abbott L, Gupta VA, Main M, Ferrari G, Kowala A, Lin YY, Tedesco FS, Scoto M, Baranello G, Manzur A, Aoki Y, Muntoni F. Investigating the role of dystrophin isoform deficiency in motor function in Duchenne muscular dystrophy. J Cachexia Sarcopenia Muscle 2022; 13:1360-1372. [PMID: 35083887 PMCID: PMC8977977 DOI: 10.1002/jcsm.12914] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 11/03/2021] [Accepted: 12/06/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is caused by DMD mutations leading to dystrophin loss. Full-length Dp427 is the primary dystrophin isoform expressed in muscle and is also expressed in the central nervous system (CNS). Two shorter isoforms, Dp140 and Dp71, are highly expressed in the CNS. While a role for Dp140 and Dp71 on DMD CNS comorbidities is well known, relationships between mutations expected to disrupt Dp140 and Dp71 and motor outcomes are not. METHODS Functional outcome data from 387 DMD boys aged 4-15 years were subdivided by DMD mutation expected effects on dystrophin isoform expression; Group 1 (Dp427 absent, Dp140/Dp71 present, n = 201); Group 2 (Dp427/Dp140 absent, Dp71 present, n = 152); and Group 3 (Dp427/Dp140/Dp71 absent, n = 34). Relationships between isoform group and North Star ambulatory assessment (NSAA) scores, 10 m walk/run velocities and rise time velocities were explored using regression analysis. Western blot analysis was used to study Dp427, Dp140 and Dp71 production in myogenic cells (control and DMD human), control skeletal muscle, DMD skeletal muscle from the three isoform groups and cerebral cortex from mice (wild-type and DMD models). Grip strength and rotarod running test were studied in wild-type mice and DMD mouse models. DMD mouse models were mdx (Dp427 absent, Dp140/Dp71 present), mdx52 (Dp427/Dp140 absent, Dp71 present) and DMD-null (lacking all isoforms). RESULTS In DMD boys, mean NSAA scores at 5 years of age were 6.1 points lower in Group 3 than Group 1 (P < 0.01) and 4.9 points lower in Group 3 than Group 2 (P = 0.05). Mean peak NSAA scores were 4.0 points lower in Group 3 than Group 1 (P < 0.01) and 1.6 points lower in Group 2 than Group 1 (P = 0.04). Mean four-limb grip strength was 1.5 g/g lower in mdx52 than mdx mice (P = 0.003) and 1.5 g/g lower in DMD-null than mdx mice (P = 0.002). Dp71 was produced in myogenic cells (control and DMD human) and skeletal muscle from humans in Groups 1 and 2 and mdx mice, but not skeletal muscle from human controls, myogenic cells and skeletal muscle from humans in Group 3 or skeletal muscle from wild-type, mdx52 or DMD-null mice. CONCLUSIONS Our results highlight the importance of considering expected effects of DMD mutations on dystrophin isoform production when considering patterns of DMD motor impairment and the implications for clinical practice and clinical trials. Our results suggest a complex relationship between dystrophin isoforms expressed in the brain and DMD motor function.
Collapse
Affiliation(s)
- Mary Chesshyre
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Deborah Ridout
- Population, Policy and Practice Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Yasumasa Hashimoto
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Japan
| | - Yoko Ookubo
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Japan
| | - Silvia Torelli
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Kate Maresh
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Valeria Ricotti
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Lianne Abbott
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Vandana Ayyar Gupta
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Marion Main
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Giulia Ferrari
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Anna Kowala
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Yung-Yao Lin
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Francesco Saverio Tedesco
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Cell and Developmental Biology, University College London, London, UK.,The Francis Crick Institute, London, UK
| | - Mariacristina Scoto
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Giovanni Baranello
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Adnan Manzur
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Japan
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
43
|
Abdulhady H, Sakr HM, Elsayed NS, El-Sobky TA, Fahmy N, Saadawy AM, Elsedfy H. Ambulatory Duchenne muscular dystrophy children: cross-sectional correlation between function, quantitative muscle ultrasound and MRI. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2022; 41:1-14. [PMID: 35465338 PMCID: PMC9004336 DOI: 10.36185/2532-1900-063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/11/2022] [Indexed: 01/24/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a progressive genetic muscle disease. Quantitative muscle ultrasound (US), muscle MRI, and functional tools are important to delineate characteristics of muscle involvement. We aimed to establish correlations between clinical/functional and above-named imaging tools respecting their diagnostic and prognostic role in DMD children. A cross-sectional retrospective study of 27 steroid-naive, ambulant male children/adolescents with genetically-confirmed DMD (mean age, 8.8 ± 3.3 years). Functional performance was assessed using motor function measure (MFM) which assess standing/transfer (D1), proximal (D2) and distal (D3) motor function, and six-minute walk test (6MWT). Imaging evaluation included quantitative muscle MRI which measured muscle fat content in a specific location of right rectus femoris by mDixon sequence. Quantitative muscle US measured right rectus femoris muscle brightness in standardized US image as an indicator of muscle fat content. We found a highly significant positive correlation between the mean MFM total score and 6MWT (R = 0.537, p = 0.007), and a highly significant negative correlation between fat content by muscle US and MFM total score (R = -0.603, p = 0.006) and its D1 subscore (R =-0.712, p = 0.001), and a significant negative correlation between fat content by US and 6MWT (R = -0.529, p = 0.02), and a significant positive correlation between muscle fat content by mDixon MRI and patient's age (R = 0.617, p = 0.01). Quantitative muscle US correlates significantly with clinical/functional assessment tools as MFM and 6MWT, and augments their role in disease-tracking of DMD. Quantitative muscle US has the potential to act as a substitute to functional assessment tools.
Collapse
Affiliation(s)
- Hala Abdulhady
- Department of Physical Medicine, Rheumatology and Rehabilitation, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hossam M. Sakr
- Department of Diagnostic and Interventional Radiology and Molecular Imaging, Faculty of Medicine, Ain Shams University, Cairo, Egypt,Correspondence Hossam M. Sakr Department of Diagnostic and Interventional Radiology and Molecular Imaging Faculty of Medicine, Ain Shams University, Abbassia square, 11381 Cairo, Egypt. E-mail:
| | - Nermine S. Elsayed
- Department of Medical Genetics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Tamer A. El-Sobky
- Division of Pediatric Orthopedics, Department of Orthopedic Surgery, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nagia Fahmy
- Neuromuscular Unit, Department of Neuropsychiatry, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Amr M. Saadawy
- Department of Diagnostic and Interventional Radiology and Molecular Imaging, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Heba Elsedfy
- Department of Medical Genetics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
44
|
Zambon AA, Waldrop MA, Alles R, Weiss RB, Conroy S, Moore-Clingenpeel M, Previtali S, Flanigan KM. Phenotypic Spectrum of Dystrophinopathy Due to Duchenne Muscular Dystrophy Exon 2 Duplications. Neurology 2022; 98:e730-e738. [PMID: 34937785 PMCID: PMC8865888 DOI: 10.1212/wnl.0000000000013246] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 12/13/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES To describe the phenotypic spectrum of dystrophinopathy in a large cohort of individuals with DMD exon 2 duplications (Dup2), who may be particularly amenable to therapies directed at restoring expression of either full-length dystrophin or nearly full-length dystrophin through utilization of the DMD exon 5 internal ribosome entry site (IRES). METHODS In this retrospective observational study, we analyzed data from large genotype-phenotype databases (the United Dystrophinopathy Project [UDP] and the Italian DMD network) and classified participants into Duchenne muscular dystrophy (DMD), intermediate muscular dystrophy (IMD), or Becker muscular dystrophy (BMD) phenotypes. Log-rank tests for time-to-event variables were used to compare age at loss of ambulation (LOA) in participants with Dup2 vs controls without Dup2 in the UDP database and for comparisons between steroid-treated vs steroid-naive participants with Dup2. RESULTS Among 66 participants with Dup2 (UDP = 40, Italy = 26), 61% were classified as DMD, 9% as IMD, and 30% as BMD. Median age at last observation was 15.4 years (interquartile range 8.79-26.0) and 75% had been on corticosteroids for at least 6 months. Age at LOA differed significantly between participants with Dup2 DMD and historical controls without Dup2 DMD (p < 0.001). Valid spirometry was limited but suggested a delay in the typical age-related decline in forced vital capacity and 24 of 55 participants with adequate cardiac data had cardiomyopathy. DISCUSSION Some patients with Dup2 display a milder disease course than controls without Dup2 DMD, and prolonged ambulation with corticosteroids suggests the potential of IRES activation as a molecular mechanism. As Dup2-targeted therapies reach clinical applications, this information is critical to aid in the interpretation of the efficacy of new treatments.
Collapse
Affiliation(s)
- Alberto A Zambon
- From Inspe and Division of Neuroscience (A.A.Z., S.P.), IRCCS Ospedale San Raffaele, Milan, Italy; The Center for Gene Therapy, Abigail Wexner Research Institute (M.A.W., R.A., K.M.F.), and Biostatistics Research Core (S.C., M.M.-C.), Nationwide Children's Hospital, Columbus, OH; Departments of Pediatrics and Neurology (M.A.W., K.M.F.), Ohio State University Medical Center, Columbus; and Department of Human Genetics (R.B.W.), University of Utah, Salt Lake City
| | - Megan A Waldrop
- From Inspe and Division of Neuroscience (A.A.Z., S.P.), IRCCS Ospedale San Raffaele, Milan, Italy; The Center for Gene Therapy, Abigail Wexner Research Institute (M.A.W., R.A., K.M.F.), and Biostatistics Research Core (S.C., M.M.-C.), Nationwide Children's Hospital, Columbus, OH; Departments of Pediatrics and Neurology (M.A.W., K.M.F.), Ohio State University Medical Center, Columbus; and Department of Human Genetics (R.B.W.), University of Utah, Salt Lake City
| | - Roxane Alles
- From Inspe and Division of Neuroscience (A.A.Z., S.P.), IRCCS Ospedale San Raffaele, Milan, Italy; The Center for Gene Therapy, Abigail Wexner Research Institute (M.A.W., R.A., K.M.F.), and Biostatistics Research Core (S.C., M.M.-C.), Nationwide Children's Hospital, Columbus, OH; Departments of Pediatrics and Neurology (M.A.W., K.M.F.), Ohio State University Medical Center, Columbus; and Department of Human Genetics (R.B.W.), University of Utah, Salt Lake City
| | - Robert B Weiss
- From Inspe and Division of Neuroscience (A.A.Z., S.P.), IRCCS Ospedale San Raffaele, Milan, Italy; The Center for Gene Therapy, Abigail Wexner Research Institute (M.A.W., R.A., K.M.F.), and Biostatistics Research Core (S.C., M.M.-C.), Nationwide Children's Hospital, Columbus, OH; Departments of Pediatrics and Neurology (M.A.W., K.M.F.), Ohio State University Medical Center, Columbus; and Department of Human Genetics (R.B.W.), University of Utah, Salt Lake City
| | - Sara Conroy
- From Inspe and Division of Neuroscience (A.A.Z., S.P.), IRCCS Ospedale San Raffaele, Milan, Italy; The Center for Gene Therapy, Abigail Wexner Research Institute (M.A.W., R.A., K.M.F.), and Biostatistics Research Core (S.C., M.M.-C.), Nationwide Children's Hospital, Columbus, OH; Departments of Pediatrics and Neurology (M.A.W., K.M.F.), Ohio State University Medical Center, Columbus; and Department of Human Genetics (R.B.W.), University of Utah, Salt Lake City
| | - Melissa Moore-Clingenpeel
- From Inspe and Division of Neuroscience (A.A.Z., S.P.), IRCCS Ospedale San Raffaele, Milan, Italy; The Center for Gene Therapy, Abigail Wexner Research Institute (M.A.W., R.A., K.M.F.), and Biostatistics Research Core (S.C., M.M.-C.), Nationwide Children's Hospital, Columbus, OH; Departments of Pediatrics and Neurology (M.A.W., K.M.F.), Ohio State University Medical Center, Columbus; and Department of Human Genetics (R.B.W.), University of Utah, Salt Lake City
| | - Stefano Previtali
- From Inspe and Division of Neuroscience (A.A.Z., S.P.), IRCCS Ospedale San Raffaele, Milan, Italy; The Center for Gene Therapy, Abigail Wexner Research Institute (M.A.W., R.A., K.M.F.), and Biostatistics Research Core (S.C., M.M.-C.), Nationwide Children's Hospital, Columbus, OH; Departments of Pediatrics and Neurology (M.A.W., K.M.F.), Ohio State University Medical Center, Columbus; and Department of Human Genetics (R.B.W.), University of Utah, Salt Lake City
| | - Kevin M Flanigan
- From Inspe and Division of Neuroscience (A.A.Z., S.P.), IRCCS Ospedale San Raffaele, Milan, Italy; The Center for Gene Therapy, Abigail Wexner Research Institute (M.A.W., R.A., K.M.F.), and Biostatistics Research Core (S.C., M.M.-C.), Nationwide Children's Hospital, Columbus, OH; Departments of Pediatrics and Neurology (M.A.W., K.M.F.), Ohio State University Medical Center, Columbus; and Department of Human Genetics (R.B.W.), University of Utah, Salt Lake City.
| |
Collapse
|
45
|
Woof AL, Selby K, Harris SR. Ankle contractures and functional motor decline in Duchenne muscular dystrophy. Brain Dev 2022; 44:105-113. [PMID: 34629214 DOI: 10.1016/j.braindev.2021.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 11/16/2022]
Abstract
INTRODUCTION This prospective, correlational pilot study investigated the relationship between ankle plantar flexion contractures and motor function in boys with Duchenne muscular dystrophy in British Columbia (BC), Canada. PARTICIPANTS Ambulatory boys with Duchenne muscular dystrophy were recruited from BC Children's Hospital, which follows everyone with Duchenne muscular dystrophy in BC ≤ 18 years of age (n = 14). METHODS Spearman and Pearson correlation coefficients were estimated to examine the association between the degree of ankle dorsiflexion range of motion and North Star Ambulatory Assessment scores and the degree of ankle dorsiflexion range and six-minute walk test distances. RESULTS Our analysis showed a moderate correlation between the degree of ankle dorsiflexion range and North Star Ambulatory Assessment scores [rho (14) = 0.50; p = 0.070] and a weak correlation between ankle dorsiflexion range of motion and six-minute walk test distances [rho (13) = 0.08; p = 0.747], however neither result was statistically significant. DISCUSSION Although a significant relationship between ankle dorsiflexion range of motion and motor function was not found, the variability of ankle dorsiflexion range suggests challenges with preventing ankle contracture. This reinforces the importance of assessing ankle range of motion in boys with Duchenne muscular dystrophy with sufficient frequency to identify a need for additional interventions.
Collapse
Affiliation(s)
- Angelina L Woof
- Rehabilitation Science Online Programs, Faculty of Medicine, University of British Columbia, T325-2211 Wesbrook Mall, Vancouver, British Columbia V6T 2B5, Canada; BC Centre for Ability, 2805 Kingsway, Vancouver, British Columbia V5R 5H9, Canada
| | - Kathryn Selby
- British Columbia Children's Hospital, University of British Columbia, 4500 Oak St., Vancouver, British Columbia V6H 3N1, Canada
| | - Susan R Harris
- Department of Physical Therapy, Faculty of Medicine, University of British Columbia, 212-2177 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3, Canada.
| |
Collapse
|
46
|
Mayhew AG, Moat D, McDermott MP, Eagle M, Griggs RC, James MK, Muni-Lofra R, Shillington A, Gregson S, Pallant L, Skura C, Staudt LA, Eichinger K, McMurchie H, Rabb R, Marco MD, Brown S, Zanin R, Arnoldi MT, McIntyre M, Wilson A, Alfano LN, Lowes LP, Blomgren C, Milev E, Iodice M, Pasternak A, Chiu A, Lehnert I, Claus N, Dieruf KA, Rolle E, Nicorici A, Andres B, Hobbiebrunken E, Roetmann G, Kern V, Civitello M, Vogt S, Hayes MJ, Scholtes C, Lacroix C, Gunn T, Warner S, Newman J, Barp A, Kundrat K, Kovelman S, Powers PJ, Guglieri M. Functional outcome measures in young, steroid-naïve boys with Duchenne muscular dystrophy. Neuromuscul Disord 2022; 32:460-467. [DOI: 10.1016/j.nmd.2022.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/25/2022] [Accepted: 02/23/2022] [Indexed: 10/19/2022]
|
47
|
Crabtree NJ, Roper H, Shaw NJ. Cessation of ambulation results in a dramatic loss of trabecular bone density in boys with Duchenne muscular dystrophy (DMD). Bone 2022; 154:116248. [PMID: 34718220 DOI: 10.1016/j.bone.2021.116248] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/12/2021] [Accepted: 10/26/2021] [Indexed: 11/02/2022]
Abstract
Glucocorticoids are currently used to improve muscle strength and prolong ambulation in boys with DMD although the effect on bone health is still unclear. The aim of this study was to compare bone strength in healthy children and boys with DMD and investigate the interaction between diminished muscle function, loss of ambulation and high dose oral steroids, over a two year time frame. Fifty children were studied, 14 healthy boys (HB), 13 boys with DMD who remained ambulant (DMD-RA) and 23 boys with DMD who lost ambulation (DMD-LA). All boys with DMD had taken oral glucocorticoids. Peripheral quantitative computed tomography was used to measure bone geometry, density, strength and muscle mass of the non-dominant tibia and radius. Measurements were made at baseline, 12 and 24 months at the distal metaphysis and mid diaphysis sites. Differences between the three groups were evaluated using ANOVA and a repeated measures model. There were no significant differences in age between the groups: mean age was 9.4, 8.7 and 8.8 years for HB, DMD-RA and DMD-LA, respectively. There was no significant difference in steroid exposure between the DMD groups. However, boys who lost ambulation had significantly lower muscle function at baseline (North Star Ambulatory Assessment DMD-RA 23.6 vs. DMD-LA 18.8; p < 0.05). At baseline, healthy boys had significantly greater trabecular bone density at the distal radius /ulna (23%/27%) and distal tibia/fibula (30%/46%) than boys with DMD (p < 0.05). They also had significantly larger diaphyseal tibiae/fibulae (74%/36%) and radii/ulnae (49%/31%) with thicker corticies and consequently greater bone strength. In contrast, boys with DMD had greater cortical density (4%). Over time, there were small significant differences in the rate of change of both muscle and bone parameters between healthy boys and boys with DMD. For both ambulant and non-ambulant boys with DMD the greatest changes in cortical bone were evident at the tibia. After two years boys with DMD had on average, 63% less bone strength than healthy boys. However, the most strikingly significant difference was in trabecular bone density for boys who became non-ambulant. By 2 years non-ambulant DMD boys had 53% less trabecular bone density at distal tibia than their healthy age matched peers compared with boys who remained ambulant who had 27% less trabecular bone density. In conclusion, bone and muscle strength is reduced for all boys with DMD even while they remain ambulant. However, tibia trabecular bone density loss is significantly accelerated in DMD boys who lose independent ambulation compared to DMD boys who remain ambulant despite equivalent levels of corticosteroid exposure.
Collapse
Affiliation(s)
- N J Crabtree
- Department of Endocrinology and Diabetes, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK; Department of Paediatrics, Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
| | - H Roper
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - N J Shaw
- Department of Endocrinology and Diabetes, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK; Department of Paediatrics, Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
48
|
Buckon CE, Sienko SE, Fowler EG, Bagley AM, Staudt LA, Sison-Williamson M, Heberer KR, McDonald CM, Sussman MD. A Longitudinal Study of Quantitative Muscle Strength and Functional Motor Ability in Ambulatory Boys with Duchenne Muscular Dystrophy. J Neuromuscul Dis 2021; 9:321-334. [PMID: 34924398 DOI: 10.3233/jnd-210704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is an X-linked recessive genetic disorder, that is characterized by progressive muscle degeneration and loss of ambulation between 7-13 years of age. Novel pharmacological agents targeting the genetic defects and disease mechanisms are becoming available; however, corticosteroid (CS) therapy remains the standard of care. OBJECTIVE The purpose of this longitudinal study was to elucidate the effect of CS therapy on the rate of muscle strength and gross motor skill decline in boys with DMD and assess the sensitivity of selected outcome measures. METHODS Eighty-four ambulatory boys with DMD (49-180 months), 70 on CS, 14 corticosteroid naïve (NCS), participated in this 8-year multi-site study. Outcomes included; isokinetic dynamometry, the Standing (STD) and Walking/Running/jumping (WRJ) dimensions of the Gross Motor Function Measure (GMFM), and Timed Function Tests (TFTs). Nonlinear mixed modeling procedures determined the rate of change with age and the influence of steroids. RESULTS Despite CS therapy the rate of decline in strength with age was significant in all muscle groups assessed. CS therapy significantly slowed decline in knee extensor strength, as the NCS group declined at 3x the rate of the CS group. Concurrently, WRJ skills declined in the NCS group at twice the rate of the CS group. 4-stair climb and 10 meter walk/run performance was superior in the boys on CS therapy. CONCLUSION CS therapy slowed the rate of muscle strength decline and afforded longer retention of select gross motor skills in boys on CS compared to boys who were NCS. Isokinetic dynamometry, Walk/Run/Jump skills, and select TFTs may prove informative in assessing the efficacy of new therapeutics in ambulatory boys with DMD.
Collapse
Affiliation(s)
| | | | - Eileen G Fowler
- Department of Orthopaedics, University of California, Los Angeles, CA, California
| | - Anita M Bagley
- Shriners Hospitals for Children, Northern California, CA, California
| | - Loretta A Staudt
- Department of Orthopaedics, University of California, Los Angeles, CA, California
| | | | - Kent R Heberer
- Department of Orthopaedics, University of California, Los Angeles, CA, California
| | - Craig M McDonald
- Department of Physical Medicine, University of California Davis Medical Center, Sacramento, CA, California
| | | |
Collapse
|
49
|
Stimpson G, Chesshyre M, Baranello G, Muntoni F. Lessons Learned From Translational Research in Neuromuscular Diseases: Impact on Study Design, Outcome Measures and Managing Expectation. Front Genet 2021; 12:759994. [PMID: 36687260 PMCID: PMC9855753 DOI: 10.3389/fgene.2021.759994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/09/2021] [Indexed: 01/25/2023] Open
Abstract
Spinal Muscular Atrophy (SMA) and Duchenne Muscular Dystrophy (DMD), two of the most common, child onset, rare neuromuscular disorders, present a case study for the translation of preclinical research into clinical work. Over the past decade, well-designed clinical trials and innovative methods have led to the approval of several novel therapies for SMA and DMD, with many more in the pipeline. This review discusses several features that must be considered during trial design for neuromuscular diseases, as well as other rare diseases, to maximise the possibility of trial success using historic examples. These features include well-defined inclusion criteria, matching criteria, alternatives to placebo-controlled trials and the selection of trial endpoints. These features will be particularly important in the coming years as the investigation into innovative therapy approaches for neuromuscular diseases continues.
Collapse
Affiliation(s)
- Georgia Stimpson
- Developmental Neuroscience Research and Training Department, Dubowitz Neuromuscular Centre, Faculty of Population Health Sciences, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Mary Chesshyre
- Developmental Neuroscience Research and Training Department, Dubowitz Neuromuscular Centre, Faculty of Population Health Sciences, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom,NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Giovanni Baranello
- Developmental Neuroscience Research and Training Department, Dubowitz Neuromuscular Centre, Faculty of Population Health Sciences, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom,NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Francesco Muntoni
- Developmental Neuroscience Research and Training Department, Dubowitz Neuromuscular Centre, Faculty of Population Health Sciences, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom,NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom,*Correspondence: Francesco Muntoni,
| |
Collapse
|
50
|
Li X, Wang Y, Wang H, Wang Y. SARS-CoV-2-associated Guillain-Barré syndrome is a para-infectious disease. Autoimmun Rev 2021; 114:625-635. [PMID: 34043803 DOI: 10.1016/j.autrev.2021.102983] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been linked to the Guillain-Barré syndrome (GBS). The objective of the present study is to identify specific clinical features of cases of GBS reported in the literature associated with SARS-CoV-2 infection. We searched Pubmed, and included single case reports and case series with full text in English, reporting original data of patients with GBS and a confirmed recent SARS-CoV-2 infection. Clinical data were extracted. We identified 28 articles (22 single case reports and 6 case series), reporting on a total of 44 GBS patients with confirmed SARS-CoV-2 infection. SARS-CoV-2 infection was confirmed through serum reverse transcriptase-polymerase chain reaction in 72.7% of cases. A total of 40 patients (91%) had symptoms compatible with SARS-CoV-2 infection before the onset of the GBS. The median period between the onset of symptoms of SARS-CoV-2 infection and symptoms of the GBS was 11.2 days (range, 2-23). The most common clinical features were: leg weakness (61.4%), leg paresthesia (50%), arm weakness (50.4%), arm paresthesia (50.4%), hyporeflexia/areflexia (48%) and ataxia (22.7%). In total, 38.6% (n = 17) were found to have facial paralysis. Among 37 patients in whom nerve-conduction studies and electromyography were performed, of which 26 patients (59.1%) were consistent with the acute inflammatory demyelinating polyneuropathy subtype of the GBS. The present retrospective analysis support the role of the SARS-CoV-2 infection in the development of the GBS, may trigger GBS as para-infectious disease, and lead to SARS-CoV-2-associated GBS.
Collapse
Affiliation(s)
- X Li
- School of Clinical Medicine, Chifeng University, Chifeng 024005, PR China
| | - Y Wang
- Department of Neurology, The Affiliated Hospital of Chifeng University, Chifeng 024005, PR China
| | - H Wang
- Department of Neurology, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, PR China
| | - Y Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, PR China
| |
Collapse
|