1
|
Bomma M, Lott D, Forbes S, Shih R, Coppola JA, Christle JW, Duong T, Russo J, Pant A, Leon-Astudillo C, Berthy J, Cousins C, Corti M, Byrne B, May J, Xue W, Taivassalo T. Cardiopulmonary exercise testing as an integrative approach to explore physiological limitations in Duchenne muscular dystrophy. J Neuromuscul Dis 2025:22143602251319170. [PMID: 40033993 DOI: 10.1177/22143602251319170] [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: 03/05/2025]
Abstract
BACKGROUND Cardiopulmonary exercise testing (CPET) is the gold-standard for quantification of peak oxygen uptake (VO2) and cardiorespiratory and muscle responses to exercise. Its application to Duchenne muscular dystrophy (DMD) has been scarce due to the notion that muscle weakness inherent to disease restricts the cardiorespiratory system from reaching maximal capacity. OBJECTIVE To investigate the utility of CPET in DMD by 1) establishing whether patients can perform maximal-effort exercise for valid VO2 peak assessment; 2) quantifying VO2 peak repeatability; 3) characterizing muscle and cardiorespiratory responses; 4) comparing VO2 peak to 6-min walk distance (6MWD). METHODS Twenty-seven DMD and eight healthy boys (6 years and older) underwent CPET using an incremental work-rate protocol for leg (ambulatory) or arm (non-ambulatory) cycling with measurement of heart rate (HR) and gas-exchange variables from rest to maximal-effort. The oxygen cost of work (ΔVO2/Δwork-rate) was calculated, and peak exercise parameters (VO2, HR, O2 pulse, ventilation (VE) and ventilatory threshold (VT)) were considered valid if the respiratory exchange ratio ≥1.01. RESULTS VO2 peak was valid (81.5% of patients), repeatable (intraclass correlation coefficient = 0.998) and low in ambulatory and non-ambulatory DMD compared to controls (19.0 ± 6.0; 10.7 ± 2; 35.2 ± 4.5 mL/kg/min respectively). VT was low (30.8 ± 10.7; 19.4 ± 3.0; 61.2 ± 6.9% VO2 peak) reflecting significant muscle metabolic impairment. Peak HR in ambulatory-DMD (172 ± 14 bpm) was similar to controls (183 ± 8.3 bpm), but O2 pulse was low (3.4 ± 1.0; 6.5 ± 1.1 mL/beat). Peak VE/VO2 (ambulatory = 42.1 ± 6.8; non-ambulatory = 42.2 ± 7.8; controls = 34.3 ± 4.6) and ΔVO2/Δwork-rate were elevated (ambulatory = 12.4 ± 4.9; non-ambulatory = 19.0 ± 9.7; controls = 10.1 ± 0.8) revealing ventilatory and mechanical inefficiency. Despite strong correlation between VO2 peak and 6MWD, severity of impairment was discordant. CONCLUSION Valid CPET is feasible in DMD, revealing low VO2 peak due to abnormal muscle metabolic and cardiorespiratory responses during dynamic exercise. CPET reveals cardiorespiratory limitations in DMD boys with unremarkable 6MWD, and should be considered an integrative approach in clinical care and assessment of emerging therapeutics.
Collapse
Affiliation(s)
- Meghana Bomma
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - Donovan Lott
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Sean Forbes
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Renata Shih
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - John-Anthony Coppola
- Department of Pediatrics, Congenital Heart Center, University of Florida, Gainesville, FL, USA
| | - Jeffrey W Christle
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Tina Duong
- Division of Neurology and Neurological Sciences, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Joseph Russo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Aditi Pant
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | | | - Julie Berthy
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Christina Cousins
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Manuela Corti
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Barry Byrne
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - James May
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - W Xue
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Tanja Taivassalo
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| |
Collapse
|
2
|
Hernández Rodríguez MY, Biswas DD, Slyne AD, Lee J, Scarrow E, Abdelbarr SM, Daniels H, O’Halloran KD, Ferreira LF, Gersbach CA, ElMallah MK. Respiratory pathology in the mdx/utrn -/- mouse: A murine model for Duchenne Muscular Dystrophy (DMD). PLoS One 2025; 20:e0316295. [PMID: 39919154 PMCID: PMC11805407 DOI: 10.1371/journal.pone.0316295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/09/2024] [Indexed: 02/09/2025] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked devastating disease caused by a lack of dystrophin which results in progressive muscle weakness. As muscle weakness progresses, respiratory insufficiency and hypoventilation result in significant morbidity and mortality. The most studied DMD mouse model- the mdx mouse- has a milder respiratory phenotype compared to humans, likely due to compensatory overexpression of utrophin. mdx/utrn-/- mice lack both dystrophin and utrophin proteins. These mice have an early onset of muscular dystrophy, severe muscle weakness, and premature death, but the respiratory pathophysiology is unclear. The objective of this study is to characterize the respiratory pathophysiology and histopathology using whole body plethysmography to measure breathing and metabolism, diaphragm muscle functional analysis, histology, and immunohistochemistry. The mdx/utrn-/- mice have significant respiratory and metabolic deficits with respiratory insufficiency and hypoventilation when exposed to hypoxia and hypercarbia as early as 6 weeks of age. They also have significant diaphragmatic weakness and disrupted diaphragmatic structural pathology. The mdx/utrn-/- mice display respiratory dysfunction that mimics the DMD phenotype and therefore can provide a useful model to study the impact of novel therapies on respiratory function for DMD.
Collapse
Affiliation(s)
- Marán Y. Hernández Rodríguez
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Debolina D. Biswas
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Aoife D. Slyne
- Department of Physiology, University College Cork, Cork, Ireland
| | - Jane Lee
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Evelyn Scarrow
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Sarra M. Abdelbarr
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Heather Daniels
- Center for Advanced Genomic Technologies, Duke University, Durham, North Carolina, United States of America
| | | | - Leonardo F. Ferreira
- Department of Orthopedic Surgery, Duke University, Durham, North Carolina, United States of America
| | - Charles A. Gersbach
- Center for Advanced Genomic Technologies, Duke University, Durham, North Carolina, United States of America
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Department of Surgery, Duke University Hospital, Durham, North Carolina, United States of America
| | - Mai K. ElMallah
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
- Department of Neurobiology, Duke University, Durham, North Carolina, United States of America
- Department of Cell Biology, Duke University, Durham, North Carolina, United States of America
| |
Collapse
|
3
|
Benemei S, Gatto F, Boni L, Pane M. "If you cannot measure it, you cannot improve it". Outcome measures in Duchenne Muscular Dystrophy: current and future perspectives. Acta Neurol Belg 2025; 125:1-12. [PMID: 39080230 PMCID: PMC11876273 DOI: 10.1007/s13760-024-02600-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/11/2024] [Indexed: 09/04/2024]
Abstract
Duchenne Muscular Dystrophy (DMD) is an X-linked recessive neuromuscular disorder primarily affecting males, caused by mutations in the dystrophin gene. The absence of dystrophin protein leads to progressive skeletal muscle degeneration. Recent advances in the therapeutic landscape underscore the need to identify appropriate outcome measures to assess treatment efficacy in ambulant and non-ambulant DMD patients, across clinical and research settings. This is essential for accurately evaluating new treatments and attributing therapeutic benefits.It is crucial to establish a robust correlation between outcome scores and disease progression patterns. This task is challenging since functional test performance may be influenced by different patient's characteristics, including the physiological evolution of the neurodevelopment together with the disease progression. While widely used DMD outcomes such as the North Star Ambulatory Assessment, the 6-Minute Walking Test, the 4 stairs climbed, and the Performance of the Upper Limb exhibit reliability and validity, their clinical significance is influenced by the wide phenotype and progression variability of the disease.We present and discuss the features (relevance, quantifiability, validity, objectivity, reliability, sensitivity, specificity, precision) of available DMD outcome measures, including new potential measures that may be provided by digital tools and artificial intelligence.
Collapse
Affiliation(s)
| | | | - Luca Boni
- U.O. Epidemiologia Clinica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Marika Pane
- Nemo Clinical Centre, Fondazione Policlinico Universitario A. Gemelli IRCSS, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
4
|
Yoon DY, Daniels MJ, Willcocks RJ, Triplett WT, Morales JF, Walter GA, Rooney WD, Vandenborne K, Kim S. Five multivariate Duchenne muscular dystrophy progression models bridging six-minute walk distance and MRI relaxometry of leg muscles. J Pharmacokinet Pharmacodyn 2024; 51:671-683. [PMID: 38609673 PMCID: PMC11470134 DOI: 10.1007/s10928-024-09910-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/15/2024] [Indexed: 04/14/2024]
Abstract
The study aimed to provide quantitative information on the utilization of MRI transverse relaxation time constant (MRI-T2) of leg muscles in DMD clinical trials by developing multivariate disease progression models of Duchenne muscular dystrophy (DMD) using 6-min walk distance (6MWD) and MRI-T2. Clinical data were collected from the prospective and longitudinal ImagingNMD study. Disease progression models were developed by a nonlinear mixed-effect modeling approach. Univariate models of 6MWD and MRI-T2 of five muscles were developed separately. Age at assessment was the time metric. Multivariate models were developed by estimating the correlation of 6MWD and MRI-T2 model variables. Full model estimation approach for covariate analysis and five-fold cross validation were conducted. Simulations were performed to compare the models and predict the covariate effects on the trajectories of 6MWD and MRI-T2. Sigmoid Imax and Emax models best captured the profiles of 6MWD and MRI-T2 over age. Steroid use, baseline 6MWD, and baseline MRI-T2 were significant covariates. The median age at which 6MWD is half of its maximum decrease in the five models was similar, while the median age at which MRI-T2 is half of its maximum increase varied depending on the type of muscle. The models connecting 6MWD and MRI-T2 successfully quantified how individual characteristics alter disease trajectories. The models demonstrate a plausible correlation between 6MWD and MRI-T2, supporting the use of MRI-T2. The developed models will guide drug developers in using the MRI-T2 to most efficient use in DMD clinical trials.
Collapse
Affiliation(s)
- Deok Yong Yoon
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Michael J Daniels
- Department of Statistics, University of Florida, Gainesville, FL, USA
| | | | - William T Triplett
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Juan Francisco Morales
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Glenn A Walter
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - William D Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Sarah Kim
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA.
| |
Collapse
|
5
|
Hoskens J, Vandekerckhove I, De Waele L, Feys H, Goemans N, Klingels K. How do fine and gross motor skills develop in preschool boys with Duchenne Muscular Dystrophy? RESEARCH IN DEVELOPMENTAL DISABILITIES 2024; 154:104845. [PMID: 39340934 DOI: 10.1016/j.ridd.2024.104845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 09/16/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Boys with Duchenne Muscular Dystrophy (DMD) experience both fine and gross motor problems. Nowadays, early intervention focuses almost exclusively on gross motor skills. AIMS We aimed to explore early motor development in preschool boys with DMD and investigate the influence of cognition. METHODS AND PROCEDURES Seventeen boys with DMD (11 months- 6 years) were compared to typically developing (TD) peers and followed-up with the Bayley Scales of Infant and Toddler Development (Bayley-III); Peabody developmental motor scales (PDMS-II) and Motor Function Measure (MFM-20). The longitudinal evolution of fine and gross motor skills was investigated using linear mixed effect models (LMM). Cognition was added to the LMM as a covariate. OUTCOMES AND RESULTS Preschool boys with DMD scored lower compared to TD peers on both fine and gross motor skills (p<0.001). The evolution of motor development was subscale-dependent. A significant influence of cognition was found on different subscales (p= 0.002-0.04). CONCLUSIONS AND IMPLICATIONS Preschool boys with DMD do not achieve the same functioning level as TD boys. Cognition plays a crucial role in the evolution of motor skills. Our results suggest a shift to a broader psychomotor approach including both fine and gross motor skills, also considering the impact of cognition. WHAT THIS PAPER ADDS?: Our study provides a detailed mapping of early fine and gross motor development in preschool boys with Duchenne Muscular Dystrophy (DMD) and describes the influence of cognition on both fine and gross motor skills. Preschool boys with DMD do not achieve the same functioning level compared to typically developing boys. They score significantly lower on both fine and gross motor skills. The evolution of fine and gross motor development was subscale-dependent e.g. a negative-positive evolution was seen for grasping skills, with a tipping point around the age of four; stationary scaled scores decreased followed by a stabilization around the age four to five and locomotion scaled scores remained stable over time. Finally, we also found that cognition plays a crucial role in the evolution of both fine and gross motor skills. These new insights in the evolution of early motor development could be of added value for future clinical trials in young boys with DMD. Subsequently, increased alertness to early symptoms, e.g. developmental delay, may advance the age of diagnosis, as well as associated early intervention.
Collapse
Affiliation(s)
- Jasmine Hoskens
- KU Leuven, Department of Rehabilitation Sciences, Research Group for Neurorehabilitation (eNRGy), Herestraat 49, Leuven 3000, Belgium; UHasselt, Faculty of Rehabilitation Sciences, Rehabilitation Research Centre (REVAL), Campus Diepenbeek, Agoralaan, Diepenbeek, Hasselt 3590, Belgium.
| | - Ines Vandekerckhove
- KU Leuven, Department of Rehabilitation Sciences, Research Group for Neurorehabilitation (eNRGy), Herestraat 49, Leuven 3000, Belgium.
| | - Liesbeth De Waele
- University Hospitals Leuven, Department of Child Neurology, Herestraat 49, Leuven 3000, Belgium; KU Leuven, Department of Development and Regeneration, Herestraat 49, Leuven 3000, Belgium.
| | - Hilde Feys
- KU Leuven, Department of Rehabilitation Sciences, Research Group for Neurorehabilitation (eNRGy), Herestraat 49, Leuven 3000, Belgium.
| | - Nathalie Goemans
- University Hospitals Leuven, Department of Child Neurology, Herestraat 49, Leuven 3000, Belgium.
| | - Katrijn Klingels
- UHasselt, Faculty of Rehabilitation Sciences, Rehabilitation Research Centre (REVAL), Campus Diepenbeek, Agoralaan, Diepenbeek, Hasselt 3590, Belgium.
| |
Collapse
|
6
|
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
|
7
|
Iff J, Done N, Tuttle E, Zhong Y, Wei F, Darras BT, McDonald CM, Mercuri E, Muntoni F. Survival among patients receiving eteplirsen for up to 8 years for the treatment of Duchenne muscular dystrophy and contextualization with natural history controls. Muscle Nerve 2024; 70:60-70. [PMID: 38482981 DOI: 10.1002/mus.28075] [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: 06/02/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 06/06/2024]
Abstract
INTRODUCTION/AIMS Eteplirsen, approved in the US for patients with Duchenne muscular dystrophy (DMD) with exon 51 skip-amenable variants, is associated with attenuated ambulatory/pulmonary decline versus DMD natural history (NH). We report overall survival in a US cohort receiving eteplirsen and contextualize these outcomes versus DMD NH. METHODS US patients with DMD receiving eteplirsen were followed through a patient support program, with data collected on ages at eteplirsen initiation and death/end of follow-up. Individual DMD NH data were extracted by digitizing Kaplan-Meier (KM) curves from published systematic and targeted literature reviews. Overall survival age was analyzed using KM curves and contextualized with DMD NH survival curves; subanalyses considered age groups and duration of eteplirsen exposure. Overall survival time from treatment initiation was also evaluated. RESULTS A total of 579 eteplirsen-treated patients were included. During a total follow-up of 2119 person-years, median survival age was 32.8 years. DMD NH survival curves extracted from four publications (follow-up for 1224 DMD NH controls) showed overall pooled median survival age of 27.4 years. Eteplirsen-treated patients had significantly longer survival from treatment initiation versus age-matched controls (age-adjusted hazard ratio [HR], 0.65; 95% confidence interval [CI], 0.44-0.98; p < .05). Longer treatment exposure was associated with improved survival (HR, 0.15; 95% CI, 0.05-0.41; p < .001). Comparisons using different DMD NH cohorts to address common risks of bias yielded consistent findings. DISCUSSION Data suggest eteplirsen may prolong survival in patients with DMD across a wide age range. As more data become available, the impact of eteplirsen on survival will be further elucidated.
Collapse
Affiliation(s)
- Joel Iff
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Nicolae Done
- Analysis Group, Inc., Boston, Massachusetts, USA
| | | | - Yi Zhong
- Analysis Group, Inc., Boston, Massachusetts, USA
| | - Fangzhou Wei
- Analysis Group, Inc., Boston, Massachusetts, USA
| | - Basil T Darras
- Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Craig M McDonald
- University of California Davis Health System, Sacramento, California, USA
| | - Eugenio Mercuri
- Paediatric Neurology and Centro Clinico NeMO, Catholic University and Policlinico Gemelli, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, Rome, Italy
- Centro Clinico NeMO, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, Rome, Italy
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health and Great Ormond Street Hospital for Children, London, UK
- Great Ormond Street Hospital, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| |
Collapse
|
8
|
Deng J, Liu F, Feng Z, Liu Z. Population longitudinal analysis of Gait Profile Score and North Star Ambulatory Assessment in children with Duchenne muscular dystrophy. CPT Pharmacometrics Syst Pharmacol 2024; 13:891-903. [PMID: 38539027 PMCID: PMC11098163 DOI: 10.1002/psp4.13126] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 05/18/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a rare X-linked recessive disorder characterized by loss-of-function mutations in the gene encoding dystrophin. These mutations lead to progressive functional deterioration including muscle weakness, respiratory insufficiency, and musculoskeletal deformities. Three-dimensional gait analysis (3DGA) has been used as a tool to analyze gait pathology through the quantification of altered joint kinematics, kinetics, and muscle activity patterns. Among 3DGA indices, the Gait Profile Score (GPS), has been used as a sensitive overall measure to detect clinically relevant changes in gait patterns in children with DMD. To enhance our understanding of the clinical translation of 3DGA, we report here the development of a population nonlinear mixed-effect model that jointly describes the disease progression of the 3DGA index, GPS, and the functional endpoint, North Star Ambulatory Assessment (NSAA). The final model consists of a quadratic structure for GPS progression and a linear structure for GPS-NSAA correlation. Our model was able to capture the improvement in function in GPS and NSAA in younger subjects, as well as the decline of function in older subjects. Furthermore, the model predicted NSAA (CFB) at 1 year reasonably well for DMD subjects ≤7 years old at baseline. The model tended to slightly underpredict the decline in NSAA after 1 year for those >7 years old at baseline, but the prediction summary statistics were well maintained within the standard deviation of observed data. Quantitative models such as this may help answer clinically relevant questions to facilitate the development of novel therapies in DMD.
Collapse
Affiliation(s)
- Jiexin Deng
- School of Nursing and HealthHenan UniversityKaifengChina
| | - Fangli Liu
- School of Nursing and HealthHenan UniversityKaifengChina
| | - Zhifen Feng
- School of Nursing and HealthHenan UniversityKaifengChina
| | - Zhigang Liu
- Department of OrthopedicsFirst Affiliated Hospital of Henan UniversityKaifengChina
| |
Collapse
|
9
|
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
|
10
|
Stimpson G, Ridout D, Wolfe A, Milev E, O’Reilly E, Manzur A, Sarkozy A, Muntoni F, Cole TJ, Baranello G, on behalf of the NorthStar Network. Quantifying Variability in Motor Function in Duchenne Muscular Dystrophy: UK Centiles for the NorthStar Ambulatory Assessment, 10 m Walk Run Velocity and Rise from Floor Velocity in GC Treated Boys. J Neuromuscul Dis 2024; 11:153-166. [PMID: 37980680 PMCID: PMC10789350 DOI: 10.3233/jnd-230159] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2023] [Indexed: 11/21/2023]
Abstract
Background Boys with Duchenne Muscular Dystrophy (DMD) display heterogeneous motor function trajectory in clinics, which represents a significant obstacle to monitoring. OBJECTIVE In this paper, we present the UK centiles for the North Star Ambulatory Assessment (NSAA), the 10 m walk/run time (10MWR) and velocity (10MWRV), and the rise from floor time (RFF) and velocity (RFFV) created from a cohort of glucocorticoid treated DMD boys between the age of 5 and 16 years. METHODS Participants were included from the UK NorthStar registry if they had initiated steroids (primarily deflazacorts/prednisolone, intermittent/daily) and were not enrolled in an interventional trial. Assessments were included if the participant had a complete NSAA, the timed tests had been completed or the corresponding items were 0, or the participant was recorded as non-ambulant, in which case the NSAA was assumed 0. RESULTS We analysed 3987 assessments of the NSAA collected from 826 participants. Of these, 1080, 1849 and 1199 were imputed as 0 for the NSAA, RFFV and 10MWRV respectively. The 10th, 25th, 50th, 75th and 90th centiles were presented. The NSAA centiles showed a peak score of 14, 20, 26, 30 and 32 respectively, with loss of ambulation at 10.7, 12.2 and 14.3 years for the 25th, 50th and 75th centiles, respectively. The centiles showed loss of rise from floor at 8.6, 10.1 and 11.9 years and a loss of 10MWR of 0 at 8.9, 10.3 and 13.8 years for the 25th, 50th and 75th centiles, respectively. The centiles were pairwise less correlated than the raw scores, suggesting an increased ability to detect variability in the DMD cohort. CONCLUSIONS The NSAA, 10MWR and RFF centiles may provide insights for clinical monitoring of DMD boys, particularly in late ambulatory participants who are uniformly declining. Future work will validate the centiles in national and international natural history cohorts.
Collapse
Affiliation(s)
- Georgia Stimpson
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Deborah Ridout
- Population, Policy & Practice Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Amy Wolfe
- 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
| | - Evelin Milev
- 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
| | - Emer O’Reilly
- 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
| | - Anna Sarkozy
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - 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
| | - Tim J. Cole
- Population, Policy & Practice Department, 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
| | - on behalf of the NorthStar Network
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
- Population, Policy & Practice 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
| |
Collapse
|
11
|
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
|
12
|
Wong BL, Summer S, Horn PS, Rutter MM, Rybalsky I, Tian C, Shellenbarger KC, Kalkwarf HJ. Appendicular lean mass index changes in patients with Duchenne muscular dystrophy and Becker muscular dystrophy. J Cachexia Sarcopenia Muscle 2023; 14:2804-2812. [PMID: 37878526 PMCID: PMC10751441 DOI: 10.1002/jcsm.13357] [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: 03/27/2023] [Revised: 09/05/2023] [Accepted: 09/18/2023] [Indexed: 10/27/2023] Open
Abstract
INTRODUCTION Mutations in the 79 exons of the dystrophin gene result in muscle wasting and weakness of varying clinical severity, ranging from severe/typical Duchenne muscular dystrophy (DMD) to intermediate DMD and mild Becker muscular dystrophy (BMD), depending on the frameshift of the mutation. We previously reported that males with DMD have progressively declining appendicular lean mass (ALM) and ALM index (ALMI) with age and worsening functional motor ability compared with healthy controls. These indices have not been studied in patients with intermediate DMD and BMD phenotypes and across DMD genotypes. In this study, we compared age-related trajectories of ALM and ALMI of patients who had (1) BMD without functional mobility deficits with patients who had DMD at different stages of disease and healthy controls; (2) a DMD intermediate phenotype with patients who had a typical DMD phenotype; and (3) DMD categorized by genotype. METHODS We conducted a retrospective review of ALM and ALMI data from 499 patients (ages 5-23 years) with DMD (466 typical and 33 intermediate) and 46 patients (ages 5-21 years) with BMD (without functional mobility deficits and functional mobility score of 1). Patients were grouped according to age reflecting disease stage (ages 5 to <7, 7 to <10, 10 to <14, and 14 to <20 years) and genotype (mutations in exons 1-30, 31-44, 45-62, and 63-79). RESULTS ALM and ALMI trajectories of patients with BMD paralleled those of healthy controls until adolescence, in contrast to patients with DMD. ALMI Z-scores of patients with BMD remained within ±2 SD without decline while those of patients with DMD fell below -2 SD around age 12 years. Patients with BMD had increasing ALM and ALMI with age, with peak accrual between ages 10 to <14 years. ALMI declined after age 14 years for those with intermediate DMD compared with 10 years for patients with typical DMD. Patients with mutations in exons 63-79 had a greater decline in ALMI as compared with those with other genotypes after age 10 years. CONCLUSIONS Age-related changes in ALMI in patients with BMD and intermediate DMD differ from those with typical DMD, reflecting their clinical phenotypes. ALM and ALMI should be further studied in patients with BMD and DMD subtypes for their potential value as surrogate markers to characterize the severity of BMD and DMD and inform clinical care decisions and clinical trial designs.
Collapse
Affiliation(s)
- Brenda L. Wong
- Department of Pediatrics and Neurology, DMD ProgramUniversity of Massachusetts Chan Medical SchoolWorcesterMAUSA
| | - Suzanne Summer
- Clinical Translational Research CenterCincinnati Children's Hospital Medical CenterCincinnatiOHUSA
| | - Paul S. Horn
- Division of NeurologyCincinnati Children's Hospital Medical CenterCincinnatiOHUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOHUSA
| | - Meilan M. Rutter
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOHUSA
- Division of EndocrinologyCincinnati Children's Hospital Medical CenterCincinnatiOHUSA
| | - Irina Rybalsky
- Division of NeurologyCincinnati Children's Hospital Medical CenterCincinnatiOHUSA
| | - Cuixia Tian
- Division of NeurologyCincinnati Children's Hospital Medical CenterCincinnatiOHUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOHUSA
| | - Karen C. Shellenbarger
- Department of Pediatrics and Neurology, DMD ProgramUniversity of Massachusetts Chan Medical SchoolWorcesterMAUSA
| | - Heidi J. Kalkwarf
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOHUSA
- Division of Gastroenterology, Hepatology and NutritionCincinnati Children's Hospital Medical CenterCincinnatiOHUSA
| |
Collapse
|
13
|
Kim S, Willcocks RJ, Daniels MJ, Morales JF, Yoon DY, Triplett WT, Barnard AM, Conrado DJ, Aggarwal V, Belfiore‐Oshan R, Martinez TN, Walter GA, Rooney WD, Vandenborne K. Multivariate modeling of magnetic resonance biomarkers and clinical outcome measures for Duchenne muscular dystrophy clinical trials. CPT Pharmacometrics Syst Pharmacol 2023; 12:1437-1449. [PMID: 37534782 PMCID: PMC10583249 DOI: 10.1002/psp4.13021] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/08/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023] Open
Abstract
Although regulatory agencies encourage inclusion of imaging biomarkers in clinical trials for Duchenne muscular dystrophy (DMD), industry receives minimal guidance on how to use these biomarkers most beneficially in trials. This study aims to identify the optimal use of muscle fat fraction biomarkers in DMD clinical trials through a quantitative disease-drug-trial modeling and simulation approach. We simultaneously developed two multivariate models quantifying the longitudinal associations between 6-minute walk distance (6MWD) and fat fraction measures from vastus lateralis and soleus muscles. We leveraged the longitudinal individual-level data collected for 10 years through the ImagingDMD study. Age of the individuals at assessment was chosen as the time metric. After the longitudinal dynamic of each measure was modeled separately, the selected univariate models were combined using correlation parameters. Covariates, including baseline scores of the measures and steroid use, were assessed using the full model approach. The nonlinear mixed-effects modeling was performed in Monolix. The final models showed reasonable precision of the parameter estimates. Simulation-based diagnostics and fivefold cross-validation further showed the model's adequacy. The multivariate models will guide drug developers on using fat fraction assessment most efficiently using available data, including the widely used 6MWD. The models will provide valuable information about how individual characteristics alter disease trajectories. We will extend the multivariate models to incorporate trial design parameters and hypothetical drug effects to inform better clinical trial designs through simulation, which will facilitate the design of clinical trials that are both more inclusive and more conclusive using fat fraction biomarkers.
Collapse
Affiliation(s)
- Sarah Kim
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of PharmacyUniversity of FloridaOrlandoFloridaUSA
| | | | | | - Juan Francisco Morales
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of PharmacyUniversity of FloridaOrlandoFloridaUSA
| | - Deok Yong Yoon
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of PharmacyUniversity of FloridaOrlandoFloridaUSA
| | | | - Alison M. Barnard
- Department of Physical TherapyUniversity of FloridaGainesvilleFloridaUSA
| | | | | | | | | | - Glenn A. Walter
- Department of Physiology and AgingUniversity of FloridaGainesvilleFloridaUSA
| | - William D. Rooney
- Advanced Imaging Research CenterOregon Health & Science UniversityPortlandOregonUSA
| | - Krista Vandenborne
- Department of Physical TherapyUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
14
|
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
|
15
|
Bello L, Hoffman EP, Pegoraro E. Is it time for genetic modifiers to predict prognosis in Duchenne muscular dystrophy? Nat Rev Neurol 2023; 19:410-423. [PMID: 37308617 DOI: 10.1038/s41582-023-00823-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/14/2023]
Abstract
Patients with Duchenne muscular dystrophy (DMD) show clinically relevant phenotypic variability, despite sharing the same primary biochemical defect (dystrophin deficiency). Factors contributing to this clinical variability include allelic heterogeneity (specific DMD mutations), genetic modifiers (trans-acting genetic polymorphisms) and variations in clinical care. Recently, a series of genetic modifiers have been identified, mostly involving genes and/or proteins that regulate inflammation and fibrosis - processes increasingly recognized as being causally linked with physical disability. This article reviews genetic modifier studies in DMD to date and discusses the effect of genetic modifiers on predicting disease trajectories (prognosis), clinical trial design and interpretation (inclusion of genotype-stratified subgroup analyses) and therapeutic approaches. The genetic modifiers identified to date underscore the importance of progressive fibrosis, downstream of dystrophin deficiency, in driving the disease process. As such, genetic modifiers have shown the importance of therapies aimed at slowing this fibrotic process and might point to key drug targets.
Collapse
Affiliation(s)
- Luca Bello
- Department of Neurosciences (DNS), University of Padova, Padova, Italy
| | - Eric P Hoffman
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University (State University of New York), Binghamton, NY, USA
| | - Elena Pegoraro
- Department of Neurosciences (DNS), University of Padova, Padova, Italy.
| |
Collapse
|
16
|
Brogna C, Pane M, Coratti G, D'Amico A, Pegoraro E, Bello L, Sansone VAM, Albamonte E, Messina S, Pini A, D'Angelo MG, Bruno C, Mongini T, Ricci FS, Berardinelli A, Battini R, Masson R, Bertini ES, Politano L, Mercuri E. Upper Limb Changes in DMD Patients Amenable to Skipping Exons 44, 45, 51 and 53: A 24-Month Study. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10040746. [PMID: 37189996 DOI: 10.3390/children10040746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION The Performance of Upper Limb version 2.0 (PUL 2.0) is increasingly used in Duchenne Muscular Dystrophy (DMD) to study longitudinal functional changes of motor upper limb function in ambulant and non-ambulant patients. The aim of this study was to evaluate changes in upper limb functions in patients carrying mutations amenable to skipping exons 44, 45, 51 and 53. METHODS All DMD patients were assessed using the PUL 2.0 for at least 2 years, focusing on 24-month paired visits in those with mutations eligible for skipping exons 44, 45, 51 and 53. RESULTS 285 paired assessments were available. The mean total PUL 2.0 12-month change was -0.67 (2.80), -1.15 (3.98), -1.46 (3.37) and -1.95 (4.04) in patients carrying mutations amenable to skipping exon 44, 45, 51 and 53, respectively. The mean total PUL 2.0 24-month change was -1.47 (3.73), -2.78 (5.86), -2.95 (4.56) and -4.53 (6.13) in patients amenable to skipping exon 44, 45, 51 and 53, respectively. The difference in PUL 2.0 mean changes among the type of exon skip class for the total score was not significant at 12 months but was significant at 24 months for the total score (p < 0.001), the shoulder (p = 0.01) and the elbow domain (p < 0.001), with patients amenable to skipping exon 44 having smaller changes compared to those amenable to skipping exon 53. There was no difference within ambulant or non-ambulant cohorts when subdivided by exon skip class for the total and subdomains score (p > 0.05). CONCLUSIONS Our results expand the information on upper limb function changes detected by the PUL 2.0 in a relatively large group of DMD patients with distinct exon-skipping classes. This information can be of help when designing clinical trials or in the interpretation of the real world data including non-ambulant patients.
Collapse
Affiliation(s)
- Claudia Brogna
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Marika Pane
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Giorgia Coratti
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Adele D'Amico
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padua, 35128 Padua, Italy
| | - Luca Bello
- Department of Neurosciences, University of Padua, 35128 Padua, Italy
| | - Valeria Ada Maria Sansone
- The NEMO Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, 20162 Milan, Italy
| | - Emilio Albamonte
- The NEMO Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, 20162 Milan, Italy
| | - Sonia Messina
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Antonella Pini
- Neuromuscular Pediatric Unit, IRRCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | | | - Claudio Bruno
- Center of Translational and Experimental Myology and Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, IRCCS Istituto Giannina Gaslini and University of Genoa, 16132 Genoa, Italy
| | - Tiziana Mongini
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Torino, 10100 Turin, Italy
| | - Federica Silvia Ricci
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Torino, 10100 Turin, Italy
| | - Angela Berardinelli
- National Neurological Institute C. Mondino Foundation, IRCCS, 27100 Pavia, Italy
| | - Roberta Battini
- Department of Developmental Neuroscience, IRCCS Stella Maris, 56018 Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Riccardo Masson
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Enrico Silvio Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Luisa Politano
- Cardiomiology and Medical Genetics, Department of Experimental Medicine, Università della Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Eugenio Mercuri
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
17
|
Pane M, Coratti G, Brogna C, Bovis F, D'Amico A, Pegoraro E, Bello L, Sansone V, Albamonte E, Ferraroli E, Mazzone ES, Fanelli L, Messina S, Catteruccia M, Cicala G, Ricci M, Frosini S, De Luca G, Rolle E, De Sanctis R, Forcina N, Norcia G, Passamano L, Gardani A, Pini A, Monaco G, D'Angelo MG, Capasso A, 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, Politano L, Mercuri E. Longitudinal Analysis of PUL 2.0 Domains in Ambulant and Non-Ambulant Duchenne Muscular Dystrophy Patients: How do they Change in Relation to Functional Ability? J Neuromuscul Dis 2023:JND221556. [PMID: 37066919 DOI: 10.3233/jnd-221556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
BACKGROUND The performance of upper limb 2.0 (PUL) is widely used to assess upper limb function in DMD patients. The aim of the study was to assess 24 month PUL changes in a large cohort of DMD patients and to establish whether domains changes occur more frequently in specific functional subgroups. METHODS The PUL was performed in 311 patients who had at least one pair of assessments at 24 months, for a total of 808 paired assessments. Ambulant patients were subdivided according to the ability to walk: >350, 250-350, ≤250 meters. Non ambulant patients were subdivided according to the time since they lost ambulation: <1, 1-2, 2-5 or >5 years. RESULTS At 12 months, the mean PUL 2.0 change on all the paired assessments was -1.30 (-1.51--1.05) for the total score, -0.5 (-0.66--0.39) for the shoulder domain, -0.6 (-0.74--0.5) for the elbow domain and -0.1 (-0.20--0.06) for the distal domain.At 24 months, the mean PUL 2.0 change on all the paired assessments was -2.9 (-3.29--2.60) for the total score, -1.30 (-1.47--1.09) for the shoulder domain, -1.30 (-1.45--1.11) for the elbow domain and -0.4 (-1.48--1.29) for the distal domain.Changes at 12 and 24 months were statistically significant between subgroups with different functional abilities for the total score and each domain (p < 0.001). CONCLUSION There were different patterns of changes among the functional subgroups in the individual domains. The time of transition, including the year before and after loss of ambulation, show the peak of negative changes in PUL total scores that reflect not only loss of shoulder but also of elbow activities. These results suggest that patterns of changes should be considered at the time of designing clinical trials.
Collapse
Affiliation(s)
- Marika Pane
- Pediatric Neurology, Università Cattolica delSacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Giorgia Coratti
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Claudia Brogna
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Francesca Bovis
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
| | - Adele D'Amico
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children'sHospital, Rome, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Luca Bello
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Valeria 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, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Sonia Messina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Michela Catteruccia
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children'sHospital, Rome, Italy
| | - Gianpaolo Cicala
- Pediatric Neurology, Università Cattolica delSacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Martina Ricci
- Pediatric Neurology, Università Cattolica delSacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Silvia Frosini
- Department of Developmental Neuroscience, IRCCS Stella Maris, Pisa, Italy
| | - Giacomo De Luca
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children'sHospital, Rome, Italy
| | - Enrica Rolle
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Torino, Turin, Italy
| | - Roberto De Sanctis
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Nicola Forcina
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Giulia Norcia
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Luigia Passamano
- Department of Experimental Medicine, Cardiomiology and Medical Genetics, Second University of Naples, Naples, Italy
| | - Alice Gardani
- Child and Adolescence NeurologicalUnit, National Neurological Institute Casimiro MondinoFoundation, IRCCS, Pavia, Italy
| | - Antonella Pini
- Child Neurologyand Psychiatry Unit, IRCCS Institute of Neurological Sciences, Bellaria Hospital, Bologna, Italy
| | - Giulia Monaco
- Child Neurologyand Psychiatry Unit, IRCCS Institute of Neurological Sciences, Bellaria Hospital, Bologna, Italy
| | | | - Anna Capasso
- Pediatric Neurology, Università Cattolica delSacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Daniela Leone
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - 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
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, and Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and ChildHealth-DINOGMI, University of Genova, Genova, Italy
| | - Claudio Bruno
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, and Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and ChildHealth-DINOGMI, 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 NeurologicalUnit, National Neurological Institute Casimiro MondinoFoundation, IRCCS, 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'sHospital, Rome, Italy
| | - Luisa Politano
- Cardiomyology and Medical Genetics Unit, Università degli Studi della CampaniaLuigi Vanvitelli Scuola di Medicina e Chirurgia, Napoli, Italy
| | - Eugenio Mercuri
- Pediatric Neurology, Università Cattolica delSacro Cuore, Rome, Italy
| |
Collapse
|
18
|
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
|
19
|
Soto ME, Pérez-Torres I, Rubio-Ruiz ME, Cano-Martínez A, Manzano-Pech L, Guarner-Lans V. Frailty and the Interactions between Skeletal Muscle, Bone, and Adipose Tissue-Impact on Cardiovascular Disease and Possible Therapeutic Measures. Int J Mol Sci 2023; 24:ijms24054534. [PMID: 36901968 PMCID: PMC10003713 DOI: 10.3390/ijms24054534] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/18/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Frailty is a global health problem that impacts clinical practice. It is complex, having a physical and a cognitive component, and it is the result of many contributing factors. Frail patients have oxidative stress and elevated proinflammatory cytokines. Frailty impairs many systems and results in a reduced physiological reserve and increased vulnerability to stress. It is related to aging and to cardiovascular diseases (CVD). There are few studies on the genetic factors of frailty, but epigenetic clocks determine age and frailty. In contrast, there is genetic overlap of frailty with cardiovascular disease and its risk factors. Frailty is not yet considered a risk factor for CVD. It is accompanied by a loss and/or poor functioning of muscle mass, which depends on fiber protein content, resulting from the balance between protein breakdown and synthesis. Bone fragility is also implied, and there is a crosstalk between adipocytes, myocytes, and bone. The identification and assessment of frailty is difficult, without there being a standard instrument to identify or treat it. Measures to prevent its progression include exercises, as well as supplementing the diet with vitamin D and K, calcium, and testosterone. In conclusion, more research is needed to better understand frailty and to avoid complications in CVD.
Collapse
Affiliation(s)
- María Elena Soto
- Department of Endocrinology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Israel Pérez-Torres
- Department of Cardiovascular Biomedicine, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - María Esther Rubio-Ruiz
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Agustina Cano-Martínez
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Linaloe Manzano-Pech
- Department of Cardiovascular Biomedicine, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Verónica Guarner-Lans
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
- Correspondence:
| |
Collapse
|
20
|
Kennedy RA, de Valle K, Adams J, Ryan MM, Fitzgerald AK, Carroll K. Characterising gait in paediatric neuromuscular disorders: an observational study of spatio-temporal gait in a clinical cohort. Disabil Rehabil 2022; 44:7023-7029. [PMID: 34546805 DOI: 10.1080/09638288.2021.1977399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
AIM Few studies have characterised the relationship between disease and gait function in children and young people with rare neuromuscular disorders (NMDs). This study aimed to describe the relationship between disease and gait in a large paediatric cohort from a neuromuscular outpatient clinic. METHODS A prospective, cross-sectional study of gait in independently ambulant children and young people aged 4-21 years with a clinical or genetically confirmed NMD. Participants traversed an electronic walkway barefoot and in footwear at self-selected and fast pace. Analysis of disease included a typically developing (TD) reference group. RESULTS A sample of 113 participants with NMD, mean age 9.5 years (SD 3.1), 28% female, grouped into nine diagnostic subgroups. Eighty percent reported limitations to functional mobility. Children with NMD walked slower, with a shorter and wider step compared to a TD reference group, with moderate to large effect sizes for each of these gait parameters indicative of the clinical significance of these gait deviations. Children with Duchenne muscular dystrophy (DMD) walked slowest with a markedly wide gait pattern. Footwear had little overall effect on gait in children with NMDs. All children could accelerate over short distances. CONCLUSIONS Gait, notably speed, step length, and width are clinically significant biomarkers of disease in paediatric NMDs, affording objective functional measures in clinical settings and research.Implications for rehabilitationGait should be considered a functional biomarker of disease in children and young people with neuromuscular disorders (NMDs).Comparison of gait in a paediatric neuromuscular cohort indicates that children with Duchenne muscular dystrophy (DMD) walk slowest with a shorter step length and a wider step width which increases with age and disease progression.Measurement of gait speed is a simple, pragmatic tool to monitor disease progression in the outpatient clinical environment and relates to everyday function.In clinical research, gait can be measured as a functional outcome to demonstrate change from disease-modifying interventions and treatments in NMDs.
Collapse
Affiliation(s)
- Rachel A Kennedy
- Clinical Sciences Group, Murdoch Children's Research Institute, Parkville, Australia.,Department of Neurology, The Royal Children's Hospital, Parkville, Australia
| | - Katy de Valle
- Clinical Sciences Group, Murdoch Children's Research Institute, Parkville, Australia.,Department of Neurology, The Royal Children's Hospital, Parkville, Australia
| | - Justine Adams
- Clinical Sciences Group, Murdoch Children's Research Institute, Parkville, Australia
| | - Monique M Ryan
- Clinical Sciences Group, Murdoch Children's Research Institute, Parkville, Australia.,Department of Neurology, The Royal Children's Hospital, Parkville, Australia
| | - Alisha K Fitzgerald
- Physiotherapy Department, Donvale Rehabilitation Hospital, Donvale, Australia
| | - Kate Carroll
- Clinical Sciences Group, Murdoch Children's Research Institute, Parkville, Australia.,Department of Neurology, The Royal Children's Hospital, Parkville, Australia
| |
Collapse
|
21
|
Almeida-Becerril T, Rodríguez-Cruz M, Hernández-Cruz SY, Ruiz-Cruz ED, Mendoza CRS, Cárdenas-Conejo A, Escobar-Cedillo RE, Ávila-Moreno F, Aquino-Jarquin G. Natural history of circulating miRNAs in Duchenne disease: Association with muscle injury and metabolic parameters. Acta Neurol Scand 2022; 146:512-524. [PMID: 36000352 DOI: 10.1111/ane.13673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/24/2022] [Accepted: 07/05/2022] [Indexed: 12/28/2022]
Abstract
OBJECTIVES This study aimed to evaluate whether the expression of circulating dystromiRs and a group of oxidative stress-related (OS-R) miRNAs is associated with muscle injury and circulating metabolic parameters in Duchenne muscular dystrophy (DMD) patients. METHODS Twenty-four DMD patients were included in this cross-sectional study. Clinical scales to evaluate muscle injury (Vignos, GMFCS, Brooke, and Medical Research Council), enzymatic muscle injury parameters (CPK, ALT, and AST), anthropometry, metabolic indicators, physical activity, serum dystromiRs (miR-1-3p, miR-133a-3p, and miR-206), and OS-R miRNAs (miR-21-5p, miR-31-5p, miR-128-3p, and miR-144-3p) levels were measured in ambulatory and non-ambulatory DMD patients. RESULTS DystromiRs (except miR-1-3p) and miRNAs OS-R levels were lower (p-value <.05) in the non-ambulatory group than the ambulatory group. The expression of those miRNAs correlated with Vignos scale score (For instance, rho = -0.567, p-value <0.05 for miR-21-5p) and with other scales scores of muscle function and strength. CPK, AST, and ALT concentration correlated with expression of all miRNAs (For instance, rho = 0.741, p-value <.05 between miR-206 level and AST concentration). MiR-21-5p level correlated with glucose concentration (rho = -0.369, p-value = .038), and the miR-1-3p level correlated with insulin concentration (rho = 0.343, p-value = .05). CONCLUSIONS Non-ambulatory DMD patients have lower circulating dystromiRs and OS-R miRNAs levels than ambulatory DMD patients. The progressive muscle injury is associated with a decrease in the expression of those miRNAs, evidencing DMD progress. These findings add new information about the natural history of DMD.
Collapse
Affiliation(s)
- Tomas Almeida-Becerril
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad Hospital de Pediatría "Dr. Silvestre Frenk Freund, Centro Médico Nacional Siglo XXI (CMN-SXXI), Instituto Mexicano del Seguro Social (IMSS), Mexico City (CDMX), Mexico.,Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), CDMX, Mexico
| | - Maricela Rodríguez-Cruz
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad Hospital de Pediatría "Dr. Silvestre Frenk Freund, Centro Médico Nacional Siglo XXI (CMN-SXXI), Instituto Mexicano del Seguro Social (IMSS), Mexico City (CDMX), Mexico
| | - Sthephanie Yannín Hernández-Cruz
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad Hospital de Pediatría "Dr. Silvestre Frenk Freund, Centro Médico Nacional Siglo XXI (CMN-SXXI), Instituto Mexicano del Seguro Social (IMSS), Mexico City (CDMX), Mexico
| | - Eugenia Dolores Ruiz-Cruz
- Departamento de Genética, Unidad Médica de Alta Especialidad Hospital General "Dr. Gaudencio González Garza", Centro Médico Nacional La Raza, IMSS, CDMX, Mexico
| | - Christian Ricardo Sánchez Mendoza
- Departamento de Genética, Unidad Médica de Alta Especialidad Hospital General "Dr. Gaudencio González Garza", Centro Médico Nacional La Raza, IMSS, CDMX, Mexico
| | - Alan Cárdenas-Conejo
- Departamento de Genética Médica, Hospital de Pediatría "Dr. Silvestre Frenk Freund", CMN-Siglo XXI, IMSS, CDMX, Mexico
| | | | - Federico Ávila-Moreno
- Lung Diseases Laboratory 12, Biomedicine Research Unit (UBIMED), Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - Guillermo Aquino-Jarquin
- Laboratorio de Investigación en Genómica, Genética y Bioinformática, Hospital Infantil de México "Federico Gómez", CDMX, Mexico
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Gagliardi D, Rizzuti M, Brusa R, Ripolone M, Zanotti S, Minuti E, Parente V, Dioni L, Cazzaniga S, Bettica P, Bresolin N, Comi GP, Corti S, Magri F, Velardo D. MicroRNAs as serum biomarkers in Becker muscular dystrophy. J Cell Mol Med 2022; 26:4678-4685. [PMID: 35880500 PMCID: PMC9443944 DOI: 10.1111/jcmm.17462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/19/2022] [Accepted: 06/07/2022] [Indexed: 11/30/2022] Open
Abstract
Becker muscular dystrophy (BMD) is an X‐linked neuromuscular disorder due to mutation in the DMD gene, encoding dystrophin. Despite a wide clinical variability, BMD is characterized by progressive muscle degeneration and proximal muscle weakness. Interestingly, a dysregulated expression of muscle‐specific microRNAs (miRNAs), called myomirs, has been found in patients affected with muscular dystrophies, although few studies have been conducted in BMD. We analysed the serum expression levels of a subset of myomirs in a cohort of 29 ambulant individuals affected by BMD and further classified according to the degree of alterations at muscle biopsy and in 11 age‐matched healthy controls. We found a significant upregulation of serum miR‐1, miR‐133a, miR‐133b and miR‐206 in our cohort of BMD patients, supporting the role of these miRNAs in the pathophysiology of the disease, and we identified serum cut‐off levels discriminating patients from healthy controls, confiming the potential of circulating miRNAs as promising noninvasive biomarkers. Moreover, serum levels of miR‐133b were found to be associated with fibrosis at muscle biopsy and with patients' motor performances, suggesting that miR‐133b might be a useful prognostic marker for BMD patients. Taken together, our data showed that these serum myomirs may represent an effective tool that may support stratification of BMD patients, providing the opportunity of both monitoring disease progression and assessing the treatment efficacy in the context of clinical trials.
Collapse
Affiliation(s)
- Delia Gagliardi
- Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, Dino Ferrari Center, University of Milan, Milan, Italy
| | - Mafalda Rizzuti
- Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Roberta Brusa
- Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Michela Ripolone
- Neuromuscular and Rare Diseases Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Simona Zanotti
- Neuromuscular and Rare Diseases Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Minuti
- Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valeria Parente
- Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Laura Dioni
- EPIGET Lab, Unit of Occupational Medicine, Department of Clinical Sciences and Community Health, IRCCS Ca' Granda Foundation Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | | | | | - Nereo Bresolin
- Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, Dino Ferrari Center, University of Milan, Milan, Italy
| | - Giacomo Pietro Comi
- Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, Dino Ferrari Center, University of Milan, Milan, Italy.,Neuromuscular and Rare Diseases Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, Dino Ferrari Center, University of Milan, Milan, Italy
| | - Francesca Magri
- Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Neuromuscular and Rare Diseases Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniele Velardo
- Neuromuscular and Rare Diseases Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
25
|
Szabo SM, Audhya IF, Rogula B, Feeny D, Gooch KL. Factors associated with the health-related quality of life among people with Duchenne muscular dystrophy: a study using the Health Utilities Index (HUI). Health Qual Life Outcomes 2022; 20:93. [PMID: 35690783 PMCID: PMC9188127 DOI: 10.1186/s12955-022-02001-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
Background Data on health state utility in Duchenne muscular dystrophy (DMD) are few. This study estimated mean utility values by age, ambulatory status and over time, and investigated which aspects of health-related quality-of-life (HRQoL) are most strongly associated with utility in DMD. Methods Data from placebo-treated ambulant boys with DMD with exon 51 skip amenable mutations, (NCT01254019), were included. Ambulatory function assessments were conducted at baseline and every 12 weeks for the trial duration. Family member proxies completed the Health Utility Index (HUI) at baseline, 24 and 48 weeks; and HUI3 and HUI2 utility values were summarized. Changes in HUI attribute level over time, and predictors of changes in utility, were explored. Results Sixty-one boys (mean [range] age of 8.0 [5–16] years) were included in the analysis. Mean baseline utilities were 0.82 (HUI3) and 0.87 (HUI2); and utilities were 0.35 (HUI3) and 0.55 (HUI2) after loss of ambulation (LOA, where applicable). Over the follow-up period mean utility declined more among the older versus younger boys. Pain accounted for the highest proportion of variability (42%) in change in HUI3 utility from baseline to week 48, while for HUI2, self-care (39%) did. After LOA, change in ambulation levels explained 88% of the decline in mean HUI3 utility and change in mobility levels explained 66% of the decline in mean HUI2 utility. Conclusions Utility values among this sample were higher than previously published estimates. In younger boys utility remained relatively stable, but older boys and those losing ambulation experienced important declines over follow-up.
Supplementary Information The online version contains supplementary material available at 10.1186/s12955-022-02001-0.
Collapse
Affiliation(s)
- Shelagh M Szabo
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, Canada.
| | - Ivana F Audhya
- Sarepta Therapeutics, 215 First St, Cambridge, MA, 02142, USA
| | - Basia Rogula
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, Canada
| | - David Feeny
- McMaster University and Health Utilities Inc., Hamilton, ON, Canada
| | | |
Collapse
|
26
|
Emery N, Strachan K, Kulshrestha R, Kuiper JH, Willis T. Evaluating the Feasibility and Reliability of Remotely Delivering and Scoring the North Star Ambulatory Assessment in Ambulant Patients with Duchenne Muscular Dystrophy. CHILDREN (BASEL, SWITZERLAND) 2022; 9:728. [PMID: 35626905 PMCID: PMC9139300 DOI: 10.3390/children9050728] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE The North Star Ambulatory Assessment (NSAA) is a validated 17-item functional rating scale and widely used to assess motor function in boys with Duchenne muscular dystrophy (DMD). The SARS-CoV-2 pandemic and subsequent Government 'lockdown' resulted in no face-to-face clinic visits hence the motor abilities were not monitored. The aim was to investigate whether the NSAA was feasible and reliable by video assessment. METHOD Ten ambulant DMD boys were selected from the electronic hospital records. Two physiotherapists scored the boys' NSAA independently and the intraclass correlation coefficient was used to assess agreement. The video scores were compared to two previous NSAA in-clinic scores. RESULTS Mean scores (SD) for clinic visit one were 22.6 (4.19) and clinic visit two 21.8 (5.3). The two physiotherapists video mean scores were 20.6 (5.66) for physiotherapist 1 and 20.6 (6.53) for physiotherapist 2. The intraclass correlation coefficient was 0.98 (95% CI 0.93-1.00) for the total NSAA and 1.00 (95% CI 1.00 to 1.00) for the rise time. The mean decline in score from clinic visit one (-12 months) to video assessment was 2.0 (2.8SD). CONCLUSION The results from the study suggest that video NSAA is partially feasible and reliable.
Collapse
Affiliation(s)
- Nicholas Emery
- Neuromuscular Service, TORCH Building, Robert Jones and Agnes Hunt Orthopaedic Hopsital, Shropshire SY10 7AG, UK; (N.E.); (K.S.); (J.H.K.); (T.W.)
| | - Kate Strachan
- Neuromuscular Service, TORCH Building, Robert Jones and Agnes Hunt Orthopaedic Hopsital, Shropshire SY10 7AG, UK; (N.E.); (K.S.); (J.H.K.); (T.W.)
| | - Richa Kulshrestha
- Neuromuscular Service, TORCH Building, Robert Jones and Agnes Hunt Orthopaedic Hopsital, Shropshire SY10 7AG, UK; (N.E.); (K.S.); (J.H.K.); (T.W.)
| | - Jan Herman Kuiper
- Neuromuscular Service, TORCH Building, Robert Jones and Agnes Hunt Orthopaedic Hopsital, Shropshire SY10 7AG, UK; (N.E.); (K.S.); (J.H.K.); (T.W.)
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK
| | - Tracey Willis
- Neuromuscular Service, TORCH Building, Robert Jones and Agnes Hunt Orthopaedic Hopsital, Shropshire SY10 7AG, UK; (N.E.); (K.S.); (J.H.K.); (T.W.)
| |
Collapse
|
27
|
Krosschell KJ, Townsend EL, Kiefer M, Simeone SD, Zumpf K, Welty L, Swoboda KJ. Natural history of 10-meter walk/run test performance in spinal muscular atrophy: A longitudinal analysis. Neuromuscul Disord 2022; 32:125-134. [PMID: 35063329 PMCID: PMC8908436 DOI: 10.1016/j.nmd.2021.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/20/2021] [Accepted: 08/17/2021] [Indexed: 02/03/2023]
Abstract
As trials and treatments for spinal muscular atrophy (SMA) rapidly evolve, understanding the natural history and potential utility of the 10-meter walk/run test (10MWRT) in ambulant individuals is critical. Study aims were to: 1) establish change over time and across age for 10MWRT time in an untreated natural history cohort of young, ambulatory participants with SMA and 2) identify relations between 10MWRT time and age, SMA type, SMN2 copy number and anthropometrics. Untreated individuals (n = 56) age 2 to 21 years who were enrolled in a long-term natural history study between 2005 and 2014 and met inclusion criteria were included. Linear mixed effects models were used to assess changes in 10MWRT time with age and associations with SMA type, SMN2 copy number, and body mass. SMA type 3b (versus 3a), SMN2 copy number 4 (versus 3) and lower body mass were associated with faster 10MWRT. 10MWRT performance improved between 3 and 8 years of age, was stable between 9 and 10, and gradually declined from 11 to 18. Findings provide the first longitudinal natural history report of 10MWRT time in young individuals with SMA and offer a critical foundation for interpreting childhood change in short distance walking speed with pharmacologic treatment.
Collapse
Affiliation(s)
- Kristin J. Krosschell
- Department of Physical Therapy & Human Movement Sciences and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elise L. Townsend
- MGH Institute of Health Professions, Boston, MA, USA,Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Sarah D. Simeone
- Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Katelyn Zumpf
- Biostatistics Collaboration Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Leah Welty
- Biostatistics Collaboration Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kathryn J. Swoboda
- Department of Neurology and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
28
|
Ricci G, Bello L, Torri F, Schirinzi E, Pegoraro E, Siciliano G. Therapeutic opportunities and clinical outcome measures in Duchenne muscular dystrophy. Neurol Sci 2022; 43:625-633. [PMID: 35608735 PMCID: PMC9126754 DOI: 10.1007/s10072-022-06085-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/14/2022] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is a devastatingly severe genetic muscle disease characterized by childhood-onset muscle weakness, leading to loss of motor function and premature death due to respiratory and cardiac insufficiency. DISCUSSION In the following three and half decades, DMD kept its paradigmatic role in the field of muscle diseases, with first systematic description of disease progression with ad hoc outcome measures and the first attempts at correcting the disease-causing gene defect by several molecular targets. Clinical trials are critical for developing and evaluating new treatments for DMD. CONCLUSIONS In the last 20 years, research efforts converged in characterization of the disease mechanism and development of therapeutic strategies. Same effort needs to be dedicated to the development of outcome measures able to capture clinical benefit in clinical trials.
Collapse
Affiliation(s)
- Giulia Ricci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luca Bello
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Francesca Torri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Erika Schirinzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
29
|
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
|
30
|
Servais L, Mercuri E, Straub V, Guglieri M, Seferian AM, Scoto M, Leone D, Koenig E, Khan N, Dugar A, Wang X, Han B, Wang D, Muntoni F. Long-Term Safety and Efficacy Data of Golodirsen in Ambulatory Patients with Duchenne Muscular Dystrophy Amenable to Exon 53 Skipping: A First-in-human, Multicenter, Two-Part, Open-Label, Phase 1/2 Trial. Nucleic Acid Ther 2021; 32:29-39. [PMID: 34788571 PMCID: PMC8817703 DOI: 10.1089/nat.2021.0043] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The aim of this Phase 1/2, 2-part, multicenter trial was to report clinical safety and efficacy of long-term golodirsen treatment among ambulatory patients with exon 53 skip-amenable Duchenne muscular dystrophy (DMD). Part 1 was a 12-week, randomized, double-blind, placebo-controlled, dose-titration study followed by 9-week safety review. Part 2 was a 168-week, open-label evaluation of golodirsen 30 mg/kg. Part 1 primary endpoint was safety. Part 2 primary endpoints were dystrophin protein expression and 6-minute walk test (6MWT); secondary endpoints were percent predicted forced vital capacity (FVC%p) and safety. Post hoc ambulation analyses used mutation-matched external natural history controls. All patients from Part 1 (golodirsen, n = 8; placebo, n = 4) plus 13 additional patients entered Part 2; 23 completed the study. Adverse events were generally mild, nonserious, and unrelated to golodirsen, with no safety-related discontinuations or deaths. Golodirsen increased dystrophin protein (16.0-fold; P < 0.001) and exon skipping (28.9-fold; P < 0.001). At 3 years, 6MWT change from baseline was −99.0 m for golodirsen-treated patients versus −181.4 m for external controls (P = 0.067), and loss of ambulation occurred in 9% versus 26% (P = 0.21). FVC%p declined 8.4% over 3 years in golodirsen-treated patients, comparing favorably with literature-reported rates. This study provides evidence for golodirsen biologic activity and long-term safety in a declining DMD population and suggests functional benefit versus external controls. Clinical Trial Registration number: NCT02310906.
Collapse
Affiliation(s)
- Laurent Servais
- I-Motion Institute, Hôpital Armand Trousseau, Paris, France.,Division of Child Neurology, Centre de Références des Maladies Neuromusculaires, Department of Pediatrics, University Hospital Liège & University of Liège, Liège, Belgium.,MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, United Kingdom
| | - Eugenio Mercuri
- Pediatric Neurology Unit, Università Cattolica del Sacro Cuore Roma, Rome, Italy.,Nemo Clinical Centre, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Michela Guglieri
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | | | - Mariacristina Scoto
- Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London, United Kingdom.,National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom
| | - Daniela Leone
- Nemo Clinical Centre, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
| | - Erica Koenig
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Navid Khan
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Ashish Dugar
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Xiaodong Wang
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Baoguang Han
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Dan Wang
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London, United Kingdom.,National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom
| | | |
Collapse
|
31
|
Clinical outcome assessments in Duchenne muscular dystrophy and spinal muscular atrophy: past, present and future. Neuromuscul Disord 2021; 31:1028-1037. [PMID: 34412961 DOI: 10.1016/j.nmd.2021.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 11/23/2022]
Abstract
Scores and scales used in pediatric motor development for neuromuscular disorders have evolved greatly since the beginning of their development. In this review we provide a brief history of scales used in pediatric patients with neuromuscular disorders and an update regarding the advancement of the scales commonly used in patients with spinal muscular atrophy and Duchenne muscular dystrophy. We focus on the collaborative effort that has led to the development of outcomes and speak to the possible future of Clinical Outcome Assessments.
Collapse
|
32
|
Mitelman O, Abdel-Hamid HZ, Byrne BJ, Connolly AM, Heydemann P, Proud C, Shieh PB, Wagner KR, Dugar A, Santra S, Signorovitch J, Goemans N, McDonald CM, Mercuri E, Mendell JR. A Combined Prospective and Retrospective Comparison of Long-Term Functional Outcomes Suggests Delayed Loss of Ambulation and Pulmonary Decline with Long-Term Eteplirsen Treatment. J Neuromuscul Dis 2021; 9:39-52. [PMID: 34420980 PMCID: PMC8842766 DOI: 10.3233/jnd-210665] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background: Studies 4658-201/202 (201/202) evaluated treatment effects of eteplirsen over 4 years in patients with Duchenne muscular dystrophy and confirmed exon-51 amenable genetic mutations. Chart review Study 4658-405 (405) further followed these patients while receiving eteplirsen during usual clinical care. Objective: To compare long-term clinical outcomes of eteplirsen-treated patients from Studies 201/202/405 with those of external controls. Methods: Median total follow-up time was approximately 6 years of eteplirsen treatment. Outcomes included loss of ambulation (LOA) and percent-predicted forced vital capacity (FVC%p). Time to LOA was compared between eteplirsen-treated patients and standard of care (SOC) external controls and was measured from eteplirsen initiation in 201/202 or, in the SOC group, from the first study visit. Comparisons were conducted using univariate Kaplan-Meier analyses and log-rank tests, and multivariate Cox proportional hazards models with regression adjustment for baseline characteristics. Annual change in FVC%p was compared between eteplirsen-treated patients and natural history study patients using linear mixed models with repeated measures. Results: Data were included from all 12 patients in Studies 201/202 and the 10 patients with available data from 405. Median age at LOA was 15.16 years. Eteplirsen-treated patients experienced a statistically significant longer median time to LOA by 2.09 years (5.09 vs. 3.00 years, p < 0.01) and significantly attenuated rates of pulmonary decline vs. natural history patients (FVC%p change: –3.3 vs. –6.0 percentage points annually, p < 0.0001). Conclusions: Study 405 highlights the functional benefits of eteplirsen on ambulatory and pulmonary function outcomes up to 7 years of follow-up in comparison to external controls.
Collapse
Affiliation(s)
| | | | | | - Anne M Connolly
- Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA
| | | | - Crystal Proud
- Children's Hospital of The King's Daughters, Norfolk, VA, USA
| | - Perry B Shieh
- University of California Los Angeles, Los Angeles, CA, USA
| | - Kathryn R Wagner
- Kennedy Krieger Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | - Craig M McDonald
- University of California Davis Health System, Sacramento, CA, USA
| | | | | | | | - Jerry R Mendell
- Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
33
|
Thangarajh M, Bello L, Gordish-Dressman H. Longitudinal motor function in proximal versus distal DMD pathogenic variants. Muscle Nerve 2021; 64:467-473. [PMID: 34255858 DOI: 10.1002/mus.27371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 07/02/2021] [Accepted: 07/09/2021] [Indexed: 11/08/2022]
Abstract
INTRODUCTION/AIMS There is considerable heterogenicity in clinical outcomes in Duchenne muscular dystrophy (DMD). The aim of this study was to assess whether dystrophin gene (DMD) pathogenic variant location influences upper or lower extremity motor function outcomes in a large prospective cohort. METHODS We used longitudinal timed and quantitative motor function measurements obtained from 154 boys with DMD over a 10-y period by the Cooperative International Neuromuscular Research Group Duchenne Natural History Study (CINRG-DNHS) to understand how the trajectories of motor function differ based on proximal versus distal DMD pathogenic variants. Proximal variants were defined as located proximal to 5' DMD intron 44, and distal variants as those including nucleotides 3' DMD including intron 44. Distal DMD variants are predicted to alter the expression of short dystrophin isoforms (Dp140, Dp116, and Dp71). We compared various upper extremity and lower extremity motor function measures in these two groups, after adjusting for total lifetime corticosteroid use. RESULTS The time to loss-of-ambulation and timed motor function measurements of both upper and lower limbs over a 10-y period were comparable between boys with proximal (n = 53) and distal (n = 101) DMD pathogenic variants. Age had a significant effect on several motor function outcomes. Boys younger than 7 y of age (n = 49) showed gain in function whereas boys 7 y and older (n = 71) declined, regardless of dystrophin pathogenic variant location. DISCUSSION The longitudinal decline in upper and lower motor function is independent of proximal versus distal location of DMD pathogenic variants.
Collapse
Affiliation(s)
- Mathula Thangarajh
- Department of Neurology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Luca Bello
- Department of Neurosciences DNS, University of Padova, Padova, Italy
| | - Heather Gordish-Dressman
- Center for Genetic Medicine, Children's Research Institute, Children's National Health System, Washington, District of Columbia, USA
| | | |
Collapse
|
34
|
Finkel RS, McDonald CM, Lee Sweeney H, Finanger E, Neil Knierbein E, Wagner KR, Mathews KD, Marks W, Statland J, Nance J, McMillan HJ, McCullagh G, Tian C, Ryan MM, O'Rourke D, Müller-Felber W, Tulinius M, Bryan Burnette W, Nguyen CT, Vijayakumar K, Johannsen J, Phan HC, Eagle M, MacDougall J, Mancini M, Donovan JM. A Randomized, Double-Blind, Placebo-Controlled, Global Phase 3 Study of Edasalonexent in Pediatric Patients with Duchenne Muscular Dystrophy: Results of the PolarisDMD Trial. J Neuromuscul Dis 2021; 8:769-784. [PMID: 34120912 PMCID: PMC8543277 DOI: 10.3233/jnd-210689] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background: Edasalonexent (CAT-1004) is an orally-administered novel small molecule drug designed to inhibit NF-κB and potentially reduce inflammation and fibrosis to improve muscle function and thereby slow disease progression and muscle decline in Duchenne muscular dystrophy (DMD). Objective: This international, randomized 2 : 1, placebo-controlled, phase 3 study in patients ≥4 – < 8 years old with DMD due to any dystrophin mutation examined the effect of edasalonexent (100 mg/kg/day) compared to placebo over 52 weeks. Methods: Endpoints were changes in the North Star Ambulatory Assessment (NSAA; primary) and timed function tests (TFTs; secondary). Assessment of health-related function used the Pediatric Outcomes Data Collection tool (PODCI). Results: One hundred thirty one patients received edasalonexent (n = 88) and placebo (n = 43). At week 52, differences between edasalonexent and placebo for NSAA total score and TFTs were not statistically significant, although there were consistently less functional declines in the edasalonexent group. A pre-specified analysis by age demonstrated that younger patients (≤6.0 years) showed more robust and statistically significant differences between edasalonexent and placebo for some assessments. Treatment was well-tolerated and the majority of adverse events were mild, and most commonly involved the gastrointestinal system (primarily diarrhea). Conclusions: Edasalonexent was generally well-tolerated with a manageable safety profile at the dose of 100 mg/kg/day. Although edasalonexent did not achieve statistical significance for improvement in primary and secondary functional endpoints for assessment of DMD, subgroup analysis suggested that edasalonexent may slow disease progression if initiated before 6 years of age. (NCT03703882)
Collapse
Affiliation(s)
- Richard S Finkel
- St. Jude Children's Research Hospital, Memphis, TN and Nemours Children's Hospital, Orlando, FL
| | | | - H Lee Sweeney
- University of Florida College of Medicine, Gainesville, FL
| | | | | | - Kathryn R Wagner
- Kennedy Krieger Institute, The Johns Hopkins School of Medicine, Baltimore, MD
| | | | | | | | | | | | | | - Cuixia Tian
- Cincinnati Children's Hospital & University of Cincinnati, Cincinnati, OH
| | | | | | | | - Mar Tulinius
- Queen Silvia Children's Hospital, Gothenburg, Sweden
| | | | | | | | | | - Han C Phan
- Rare Disease Research, LLC, Atlanta GA, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Mendell JR, Sahenk Z, Lehman K, Nease C, Lowes LP, Miller NF, Iammarino MA, Alfano LN, Nicholl A, Al-Zaidy S, Lewis S, Church K, Shell R, Cripe LH, Potter RA, Griffin DA, Pozsgai E, Dugar A, Hogan M, Rodino-Klapac LR. Assessment of Systemic Delivery of rAAVrh74.MHCK7.micro-dystrophin in Children With Duchenne Muscular Dystrophy: A Nonrandomized Controlled Trial. JAMA Neurol 2021; 77:1122-1131. [PMID: 32539076 PMCID: PMC7296461 DOI: 10.1001/jamaneurol.2020.1484] [Citation(s) in RCA: 254] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Question Is rAAVrh74.MHCK7.micro-dystrophin gene transfer safe and well tolerated in patients with Duchenne muscular dystrophy? Findings In this nonrandomized controlled trial of 4 young patients with Duchenne muscular dystrophy, rAAVrh74.MHCK7.micro-dystrophin gene transfer was well tolerated, with minimal adverse events, and was associated with robust micro-dystrophin expression, reduced serum creatine kinase levels, and functional improvement as measured by the North Star Ambulatory Assessment. Meaning These results indicated the safe systemic delivery of micro-dystrophin transgene and targeted expression of functional micro-dystrophin protein product, suggesting the potential for rAAVrh74.MHCK7.micro-dystrophin to provide clinically meaningful functional improvement that is greater than the standard of care. Importance Micro-dystrophin gene transfer shows promise for treating patients with Duchenne muscular dystrophy (DMD) using recombinant adeno-associated virus serotype rh74 (rAAVrh74) and codon-optimized human micro-dystrophin driven by a skeletal and cardiac muscle-specific promoter with enhanced cardiac expression (MHCK7). Objective To identify the 1-year safety and tolerability of intravenous rAAVrh74.MHCK7.micro-dystrophin in patients with DMD. Design, Setting, and Participants This open-label, phase 1/2a nonrandomized controlled trial was conducted at the Nationwide Children’s Hospital in Columbus, Ohio. It began on November 2, 2017, with a planned duration of follow-up of 3 years, ending in March 2021. The first 4 patients who met eligibility criteria were enrolled, consisting of ambulatory male children with DMD without preexisting AAVrh74 antibodies and a stable corticosteroid dose (≥12 weeks). Interventions A single dose of 2.0 × 1014 vg/kg rAAVrh74.MHCK7.micro-dystrophin was infused through a peripheral limb vein. Daily prednisolone, 1 mg/kg, started 1 day before gene delivery (30-day taper after infusion). Main Outcomes and Measures Safety was the primary outcome. Secondary outcomes included micro-dystrophin expression by Western blot and immunohistochemistry. Functional outcomes measured by North Star Ambulatory Assessment (NSAA) and serum creatine kinase were exploratory outcomes. Results Four patients were included (mean [SD] age at enrollment, 4.8 [1.0] years). All adverse events (n = 53) were considered mild (33 [62%]) or moderate (20 [38%]), and no serious adverse events occurred. Eighteen adverse events were considered treatment related, the most common of which was vomiting (9 of 18 events [50%]). Three patients had transiently elevated γ-glutamyltransferase, which resolved with corticosteroids. At 12 weeks, immunohistochemistry of gastrocnemius muscle biopsy specimens revealed robust transgene expression in all patients, with a mean of 81.2% of muscle fibers expressing micro-dystrophin with a mean intensity of 96% at the sarcolemma. Western blot showed a mean expression of 74.3% without fat or fibrosis adjustment and 95.8% with adjustment. All patients had confirmed vector transduction and showed functional improvement of NSAA scores and reduced creatine kinase levels (posttreatment vs baseline) that were maintained for 1 year. Conclusions and Relevance This trial showed rAAVrh74.MHCK7.micro-dystrophin to be well tolerated and have minimal adverse events; the safe delivery of micro-dystrophin transgene; the robust expression and correct localization of micro-dystrophin protein; and improvements in creatine kinase levels and NSAA scores. These findings suggest that rAAVrh74.MHCK7.micro-dystrophin can provide functional improvement that is greater than that observed under standard of care. Trial Registration ClinicalTrials.gov Identifier: NCT03375164
Collapse
Affiliation(s)
- Jerry R Mendell
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus.,Department of Neurology, The Ohio State University, Columbus
| | - Zarife Sahenk
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus.,Department of Neurology, The Ohio State University, Columbus
| | - Kelly Lehman
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus
| | - Carrie Nease
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus
| | - Linda P Lowes
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus.,Department of Neurology, The Ohio State University, Columbus
| | - Natalie F Miller
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Megan A Iammarino
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Lindsay N Alfano
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Amanda Nicholl
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Samiah Al-Zaidy
- Department of Pediatrics, The Ohio State University, Columbus
| | - Sarah Lewis
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Sarepta Therapeutics Inc, Cambridge, Massachusetts
| | - Kathleen Church
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Richard Shell
- Department of Pediatrics, The Ohio State University, Columbus
| | - Linda H Cripe
- Department of Pediatrics, The Ohio State University, Columbus
| | - Rachael A Potter
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Sarepta Therapeutics Inc, Cambridge, Massachusetts
| | - Danielle A Griffin
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Sarepta Therapeutics Inc, Cambridge, Massachusetts
| | - Eric Pozsgai
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Sarepta Therapeutics Inc, Cambridge, Massachusetts
| | - Ashish Dugar
- Sarepta Therapeutics Inc, Cambridge, Massachusetts
| | - Mark Hogan
- Department of Radiology, Vascular and Interventional Radiology, Nationwide Children's Hospital, Columbus, Ohio
| | - Louise R Rodino-Klapac
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus.,Sarepta Therapeutics Inc, Cambridge, Massachusetts
| |
Collapse
|
36
|
Miller NF, Alfano LN, Iammarino MA, Connolly AM, Moore-Clingenpeel M, Powers BR, Tsao CY, Waldrop MA, Flanigan KM, Mendell JR, Lowes LP. Natural History of Steroid-Treated Young Boys With Duchenne Muscular Dystrophy Using the NSAA, 100m, and Timed Functional Tests. Pediatr Neurol 2020; 113:15-20. [PMID: 32979653 DOI: 10.1016/j.pediatrneurol.2020.08.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Clinical trials targeting younger cohorts of boys with Duchenne muscular dystrophy are necessary as earlier intervention may maximize treatment effect. Boys with Duchenne muscular dystrophy often have gross motor delays very early in life, and although they gain skills, they are on a lower trajectory than typical peers. Quantifying the natural rate of motor maturation in Duchenne muscular dystrophy from an early age permits identification of deviations from the expected trajectory related to treatment effects. METHODS The purpose of our study was to define the natural history in boys aged from ≥3 to <8 years using the North Star Ambulatory Assessment (NSAA), 100-meter timed test (100m), 10-meter walk/run (10m), time to rise (Rise), and 4-stair climb (4SC). Assessments were completed as standard of care during regularly scheduled clinic visits. RESULTS One hundred sixty-two boys with DMD aged 3.1 to 7.9 years on glucocorticoids were evaluated using one or more of the following tests as appropriate for age: NSAA (N = 158; 3.1-7.9 years), 100m (N = 131; 3.4-7.9 years), 10m (N = 162; 3.1-7.9 years), Rise (N = 160; 3.1-7.9 years), and 4SC (N = 153; 3.1-7.9 years). Longitudinal data are presented by age in a subcohort (N = 64). CONCLUSIONS Our study documents the baseline function of boys with DMD who are being treated with corticosteroids. These data will be useful to compare ongoing and future therapeutic intervention(s) for DMD.
Collapse
Affiliation(s)
- Natalie F Miller
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.
| | - Lindsay N Alfano
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Megan A Iammarino
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Anne M Connolly
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio; Department of Neurology, The College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Melissa Moore-Clingenpeel
- Biostatistics Resource, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Brenna R Powers
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Chang-Yong Tsao
- Department of Neurology, The College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Megan A Waldrop
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio; Department of Pediatrics, The College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Kevin M Flanigan
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio; Department of Pediatrics, The College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Jerry R Mendell
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio; Department of Pediatrics, The College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Linda P Lowes
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio; Department of Pediatrics, The College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
37
|
Jacques MF, Onambele-Pearson GL, Reeves ND, Stebbings GK, Dawson EA, Stockley RC, Edwards B, Morse CI. 12-Month changes of muscle strength, body composition and physical activity in adults with dystrophinopathies. Disabil Rehabil 2020; 44:1847-1854. [PMID: 32853037 DOI: 10.1080/09638288.2020.1808087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE Muscular dystrophy (MD) is an umbrella term for muscle wasting conditions, for which longitudinal changes in function and body composition are well established in children with Duchenne (DMD), however, changes in adults with DMD and Beckers (BMD), respectively, remain poorly reported. This study aims to assess 12-month changes in lower-limb strength, muscle size, body composition and physical activity in adults with Muscular Dystrophy (MD). METHODS Adult males with Duchenne MD (DMD; N = 15) and Beckers MD (BMD; N = 12) were assessed at baseline and 12-months for body composition (Body fat and lean body mass (LBM)), Isometric maximal voluntary contraction (Knee-Extension (KEMVC) and Plantar-Flexion (PFMVC)) and physical activity (tri-axial accelerometry). RESULTS 12-Month change in strength was found as -19% (PFMVC) and -14% (KEMVC) in DMD. 12-Month change in strength in BMD, although non-significant, was explained by physical activity (R2=0.532-0.585). Changes in LBM (DMD) and body fat (BMD) were both masked by non-significant changes in body mass. DISCUSSION 12-Month changes in adults with DMD appear consistent with paediatric populations. Physical activity appears important for muscle function maintenance. Specific monitoring of body composition, and potential co-morbidities, within adults with MD is highlighted.Implications for rehabilitationQuantitative muscle strength assessment shows progressive muscle weakness in adults with Duchenne Muscular Dystrophy is comparable to paediatric reports (-14 to -19%).Physical activity should be encouraged in adults with Beckers Muscular Dystrophy, anything appears better than nothing.Body composition, rather than body mass, should be monitored closely to identify any increase in body fat.
Collapse
Affiliation(s)
- Matthew F Jacques
- Faculty of Science and Engineering, School of Healthcare Science, Research Centre for Musculoskeletal Science & Sports Medicine, Manchester Metropolitan University, Manchester, United Kingdom
| | - Gladys L Onambele-Pearson
- Faculty of Science and Engineering, School of Healthcare Science, Research Centre for Musculoskeletal Science & Sports Medicine, Manchester Metropolitan University, Manchester, United Kingdom
| | - Neil D Reeves
- Faculty of Science and Engineering, School of Healthcare Science, Research Centre for Musculoskeletal Science & Sports Medicine, Manchester Metropolitan University, Manchester, United Kingdom
| | - Georgina K Stebbings
- Faculty of Science and Engineering, School of Healthcare Science, Research Centre for Musculoskeletal Science & Sports Medicine, Manchester Metropolitan University, Manchester, United Kingdom
| | - Ellen A Dawson
- Research Institute for Sport and Exercise Science, Liverpool John Moores University, Liverpool, United Kingdom
| | - Rachel C Stockley
- School of Nursing, University of Central Lancashire, Preston, United Kingdom
| | - Bryn Edwards
- The Neuromuscular Centre, Winsford, Cheshire, United Kingdom
| | - Christopher I Morse
- Faculty of Science and Engineering, School of Healthcare Science, Research Centre for Musculoskeletal Science & Sports Medicine, Manchester Metropolitan University, Manchester, United Kingdom
| |
Collapse
|
38
|
Gedlinske AM, Stephan CM, Mockler SRH, Laubscher KM, Laubenthal KS, Crockett CD, Zimmerman MB, Mathews KD. Motor outcome measures in patients with FKRP mutations: A longitudinal follow-up. Neurology 2020; 95:e2131-e2139. [PMID: 32764098 DOI: 10.1212/wnl.0000000000010604] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 05/06/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To test the hypothesis that we will be able to detect change in motor outcome measures over time in a cohort with mutations in FKRP. METHODS Individuals with documented FKRP mutations were evaluated annually with a battery of established motor outcome measures including limited quantitative myometry and timed function measures. Results were analyzed using random coefficient regression to determine annual change in each measure. Due to the nonlinear progression through the lifespan of the study participants, pediatric (<19 years) and adult (≥19 years) cohorts were analyzed separately. Effect of genotype was evaluated in each cohort. RESULTS Sixty-nine participants (30 pediatric, 44 adult) with at least 2 evaluations were included. There was a small but statistically significant decline in timed motor function measures in both pediatric and adult cohorts. Genotype significantly affected rate of decline in the pediatric but not the adult cohort. Some pediatric patients who are homozygous for the c.826C>A mutation showed improving motor performance in adolescence. Performance on the 10-meter walk/run was highly correlated with other timed function tests. CONCLUSIONS There is a slow annual decline in motor function in adults with FKRP mutations that can be detected with standard motor outcome measures, while the results in the pediatric population were more variable and affected by genotype. Overall, these analyses provide a framework for development of future clinical trials. The dystroglycanopathies natural history study (Clinical Trial Readiness for the Dystroglycanopathies) may be found on clinicaltrials.gov (NCT00313677).
Collapse
Affiliation(s)
- Amber M Gedlinske
- From the Department of Pediatrics (A.M.G., C.M.S., C.D.C., K.D.M.) and Center for Disabilities and Development (S.R.H.M., K.M.L., K.S.L.), University of Iowa Hospitals and Clinics; and Department of Biostatistics (M.B.Z.), University of Iowa College of Public Health, Iowa City. C.D.C. is now affiliated with Washington University, St. Louis, MO
| | - Carrie M Stephan
- From the Department of Pediatrics (A.M.G., C.M.S., C.D.C., K.D.M.) and Center for Disabilities and Development (S.R.H.M., K.M.L., K.S.L.), University of Iowa Hospitals and Clinics; and Department of Biostatistics (M.B.Z.), University of Iowa College of Public Health, Iowa City. C.D.C. is now affiliated with Washington University, St. Louis, MO
| | - Shelley R H Mockler
- From the Department of Pediatrics (A.M.G., C.M.S., C.D.C., K.D.M.) and Center for Disabilities and Development (S.R.H.M., K.M.L., K.S.L.), University of Iowa Hospitals and Clinics; and Department of Biostatistics (M.B.Z.), University of Iowa College of Public Health, Iowa City. C.D.C. is now affiliated with Washington University, St. Louis, MO
| | - Katie M Laubscher
- From the Department of Pediatrics (A.M.G., C.M.S., C.D.C., K.D.M.) and Center for Disabilities and Development (S.R.H.M., K.M.L., K.S.L.), University of Iowa Hospitals and Clinics; and Department of Biostatistics (M.B.Z.), University of Iowa College of Public Health, Iowa City. C.D.C. is now affiliated with Washington University, St. Louis, MO
| | - Karla S Laubenthal
- From the Department of Pediatrics (A.M.G., C.M.S., C.D.C., K.D.M.) and Center for Disabilities and Development (S.R.H.M., K.M.L., K.S.L.), University of Iowa Hospitals and Clinics; and Department of Biostatistics (M.B.Z.), University of Iowa College of Public Health, Iowa City. C.D.C. is now affiliated with Washington University, St. Louis, MO
| | - Cameron D Crockett
- From the Department of Pediatrics (A.M.G., C.M.S., C.D.C., K.D.M.) and Center for Disabilities and Development (S.R.H.M., K.M.L., K.S.L.), University of Iowa Hospitals and Clinics; and Department of Biostatistics (M.B.Z.), University of Iowa College of Public Health, Iowa City. C.D.C. is now affiliated with Washington University, St. Louis, MO
| | - M Bridget Zimmerman
- From the Department of Pediatrics (A.M.G., C.M.S., C.D.C., K.D.M.) and Center for Disabilities and Development (S.R.H.M., K.M.L., K.S.L.), University of Iowa Hospitals and Clinics; and Department of Biostatistics (M.B.Z.), University of Iowa College of Public Health, Iowa City. C.D.C. is now affiliated with Washington University, St. Louis, MO
| | - Katherine D Mathews
- From the Department of Pediatrics (A.M.G., C.M.S., C.D.C., K.D.M.) and Center for Disabilities and Development (S.R.H.M., K.M.L., K.S.L.), University of Iowa Hospitals and Clinics; and Department of Biostatistics (M.B.Z.), University of Iowa College of Public Health, Iowa City. C.D.C. is now affiliated with Washington University, St. Louis, MO.
| |
Collapse
|
39
|
Can simple and low-cost motor function assessments help in the diagnostic suspicion of Duchenne muscular dystrophy? JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2020. [DOI: 10.1016/j.jpedp.2019.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
40
|
Pereira AC, Araújo APDQC, Ribeiro MG. Can simple and low-cost motor function assessments help in the diagnostic suspicion of Duchenne muscular dystrophy? J Pediatr (Rio J) 2020; 96:503-510. [PMID: 31009620 PMCID: PMC9432257 DOI: 10.1016/j.jped.2019.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 01/25/2019] [Accepted: 02/18/2019] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE Duchenne muscular dystrophy, an X-linked genetic disease, leads to progressive muscle weakness mainly in the lower limbs. Motor function tests help to monitor disease progression. Can low-cost, simple assessments help in the diagnostic suspicion of Duchenne muscular dystrophy? The authors aim to define the sensitivity of time to rise from the floor, time to walk 10meters, and time to run 10meters, evaluating them as eventual diagnostic screening tools. METHODS This is an analytical, observational, retrospective (1998-2015), and prospective study (2015-2018). Cases were recruited from the database of the pediatric neurology department and the healthy, from child care consultations, with normal gait development (up to 15 months) and without other comorbidities (neuromuscular, pulmonary, heart diseases) from the same university hospital. RESULTS 128 Duchenne muscular dystrophy patients and 344 healthy children were analyzed, equally distributed in age groups. In Duchenne muscular dystrophy, there is a progressive increase in the means of the times to perform the motor tests according to the age group, which accelerates very abruptly after 7 years of age. Healthy children acquire maximum motor capacity at 6 years and stabilize their times. The time to rise showed a p-value <0.05 and a strong association (effect size [ES] >0.8) in all age groups (except at 12 years), with time to walk 10 meters from 9 years, and with time to run 10 meters , from 5 years. The 100% sensitivity points were defined as follows: time to rise, at 2s; time to walk 10 meters, 5s; time to run 10 meters, 4s. CONCLUSIONS Time to rise is a useful and simple tool in the screening of neuromuscular disorders such as Duchenne muscular dystrophy, a previously incurable disease with new perspectives for treatment.
Collapse
Affiliation(s)
- Aline Chacon Pereira
- Universidade Federal do Rio de Janeiro (UFRJ), Departamento de Pediatria, Neuropediatria, Rio de Janeiro, RJ, Brazil.
| | | | - Márcia Gonçalves Ribeiro
- Universidade Federal do Rio de Janeiro (UFRJ), Departamento de Pediatria, Genética Clínica, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
41
|
Capitanio D, Moriggi M, Torretta E, Barbacini P, De Palma S, Viganò A, Lochmüller H, Muntoni F, Ferlini A, Mora M, Gelfi C. Comparative proteomic analyses of Duchenne muscular dystrophy and Becker muscular dystrophy muscles: changes contributing to preserve muscle function in Becker muscular dystrophy patients. J Cachexia Sarcopenia Muscle 2020; 11:547-563. [PMID: 31991054 PMCID: PMC7113522 DOI: 10.1002/jcsm.12527] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 11/08/2019] [Accepted: 11/24/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are characterized by muscle wasting leading to loss of ambulation in the first or third decade, respectively. In DMD, the lack of dystrophin hampers connections between intracellular cytoskeleton and cell membrane leading to repeated cycles of necrosis and regeneration associated with inflammation and loss of muscle ordered structure. BMD has a similar muscle phenotype but milder. Here, we address the question whether proteins at variance in BMD compared with DMD contribute to the milder phenotype in BMD, thus identifying a specific signature to be targeted for DMD treatment. METHODS Proteins extracted from skeletal muscle from DMD/BMD patients and young healthy subjects were either reduced and solubilized prior two-dimensional difference in gel electrophoresis/mass spectrometry differential analysis or tryptic digested prior label-free liquid chromatography with tandem mass spectrometry. Statistical analyses of proteins and peptides were performed by DeCyder and Perseus software and protein validation and verification by immunoblotting. RESULTS Proteomic results indicate minor changes in the extracellular matrix (ECM) protein composition in BMD muscles with retention of mechanotransduction signalling, reduced changes in cytoskeletal and contractile proteins. Conversely, in DMD patients, increased levels of several ECM cytoskeletal and contractile proteins were observed whereas some proteins of fast fibres and of Z-disc decreased. Detyrosinated alpha-tubulin was unchanged in BMD and increased in DMD although neuronal nitric oxide synthase was unchanged in BMD and greatly reduced in DMD. Metabolically, the tissue is characterized by a decrement of anaerobic metabolism both in DMD and BMD compared with controls, with increased levels of the glycogen metabolic pathway in BMD. Oxidative metabolism is severely compromised in DMD with impairment of malate shuttle; conversely, it is active in BMD supporting the tricarboxylic acid cycle and respiratory chain. Adipogenesis characterizes DMD, whereas proteins involved in fatty acids beta-oxidation are increased in BMD. Proteins involved in protein/amino acid metabolism, cell development, calcium handling, endoplasmic reticulum/sarcoplasmic reticulum stress response, and inflammation/immune response were increased in DMD. Both disorders are characterized by the impairment of N-linked protein glycosylation in the endoplasmic reticulum. Authophagy was decreased in DMD whereas it was retained in BMD. CONCLUSIONS The mechanosensing and metabolic disruption are central nodes of DMD/BMD phenotypes. The ECM proteome composition and the metabolic rewiring in BMD lead to preservation of energy levels supporting autophagy and cell renewal, thus promoting the retention of muscle function. Conversely, DMD patients are characterized by extracellular and cytoskeletal protein dysregulation and by metabolic restriction at the level of α-ketoglutarate leading to shortage of glutamate-derived molecules that over time triggers lipogenesis and lipotoxicity.
Collapse
Affiliation(s)
- Daniele Capitanio
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy.,IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Manuela Moriggi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Enrica Torretta
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Pietro Barbacini
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Sara De Palma
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Agnese Viganò
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Hanns Lochmüller
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany.,Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain.,Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Canada.,Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, Canada
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, University College London, Institute of Child Health, London, UK.,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
| | - Alessandra Ferlini
- Dubowitz Neuromuscular Centre, University College London, Institute of Child Health, London, UK.,Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Marina Mora
- Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy.,IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| |
Collapse
|
42
|
Kennedy RA, Carroll K, McGinley JL, Paterson KL. Walking and weakness in children: a narrative review of gait and functional ambulation in paediatric neuromuscular disease. J Foot Ankle Res 2020; 13:10. [PMID: 32122377 PMCID: PMC7052968 DOI: 10.1186/s13047-020-0378-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 02/18/2020] [Indexed: 12/18/2022] Open
Abstract
Background Weakness is the primary impairment in paediatric neuromuscular diseases, impacting gait and gait-related functional activities in ambulant children affected by these rare and often degenerative diseases. Gait speed is an indicator of health and disability, yet gait is a complex, multi-faceted activity. Using the International Classification of Function, Health and Disability (ICF) model, assessment of gait and functional ambulation should consider the impairments, activity limitations and participation restrictions due to disease, and factors related to the environment and the individual person. Methods This narrative review involved a literature search of databases including Medline, Embase and Pubmed from 1946 to October 2019. Inclusion criteria included assessments of gait, endurance and ambulatory function in paediatric (0–18 years) neuromuscular diseases. Results Fifty-two papers were identified reporting assessments of gait speed, timed function, endurance and ambulatory capacity, gait-related balance and qualitative descriptive assessments of gait function and effect of disease on gait and gait-related activities. Gait speed is an indicator of disability and children with neuromuscular disease walk slower than typically developing peers. Increasing disease severity and age were associated with slower walking in children with Duchenne muscular dystrophy and Charcot-Marie-Tooth disease. The six-minute walk test is used widely as a test of endurance and ambulatory capacity; six-minute walk distance was substantially reduced across all paediatric neuromuscular diseases. Endurance and ambulatory capacity was more limited in children with spinal muscular atrophy type 3, congenital muscular dystrophy and older boys with Duchenne muscular dystrophy. Only a few papers considered normalisation of gait parameters accounting for the effect on gait of height in heterogeneous groups of children and linear growth in longitudinal studies. Balance related to gait was considered in five papers, mainly in children with Charcot-Marie-Tooth disease. There was limited investigation of factors including distance requirements and terrain in children’s typical environments and personal factors related to self-perception of disease effect on gait and gait-related function. Conclusion Assessments of gait and functional ambulation are important considerations in documenting disease progression and treatment efficacy in the clinical setting; and in clinical trials of disease-modifying agents and physiotherapeutic interventions in paediatric neuromuscular diseases. There is a need for expert consensus on core gait and functional ambulation assessments for use in clinical and research settings.
Collapse
Affiliation(s)
- Rachel A Kennedy
- Department of Neurology, The Royal Children's Hospital, Parkville, Vic, Australia. .,Murdoch Children's Research Institute, Parkville, Vic, Australia. .,Department of Physiotherapy, The University of Melbourne, Parkville, Vic, Australia.
| | - Kate Carroll
- Department of Neurology, The Royal Children's Hospital, Parkville, Vic, Australia.,Murdoch Children's Research Institute, Parkville, Vic, Australia.,Department of Physiotherapy, The University of Melbourne, Parkville, Vic, Australia
| | - Jennifer L McGinley
- Murdoch Children's Research Institute, Parkville, Vic, Australia.,Department of Physiotherapy, The University of Melbourne, Parkville, Vic, Australia
| | - Kade L Paterson
- Department of Physiotherapy, The University of Melbourne, Parkville, Vic, Australia
| |
Collapse
|
43
|
Lennie JL, Mondick JT, Gastonguay MR. Latent process model of the 6-minute walk test in Duchenne muscular dystrophy : A Bayesian approach to quantifying rare disease progression. J Pharmacokinet Pharmacodyn 2020; 47:91-104. [PMID: 31960231 DOI: 10.1007/s10928-020-09671-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/05/2020] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a rare X-linked genetic pediatric disease characterized by a lack of functional dystrophin production in the body, resulting in muscle deterioration. Lower body muscle weakness progresses to non-ambulation typically by early teenage years, followed by upper body muscle deterioration and ultimately death by the late twenties. The objective of this study was to enhance the quantitative understanding of DMD disease progression through nonlinear mixed effects modeling of the population mean and variability of the 6-min walk test (6MWT) clinical endpoint. An indirect response model with a latent process was fit to digitized literature data using full Bayesian estimation. The modeling data set consisted of 22 healthy controls and 218 DMD patients from one interventional and four observational trials. The model reasonably described the central tendency and population variability of the 6MWT in healthy subjects and DMD patients. An exploratory categorical covariate analysis indicated that there was no apparent effect of corticosteroid administration on DMD disease progression. The population predicted 6MWT began to rise at 1.32 years of age, plateauing at 654 meters (m) at 17.2 years of age for the healthy population. The DMD trajectory reached a maximum of 411 m at 8.90 years before declining and falling below 1 m at age 18.0. The model has potential to be used as a Bayesian estimation and posterior simulation tool to make informed model-based drug development decisions that incorporate prior knowledge with new data.
Collapse
Affiliation(s)
- Janelle L Lennie
- Metrum Research Group, Tariffville, CT, 06081, USA.
- University of Connecticut, Storrs, CT, 06268, USA.
| | | | - Marc R Gastonguay
- Metrum Research Group, Tariffville, CT, 06081, USA
- University of Connecticut, Storrs, CT, 06268, USA
| |
Collapse
|
44
|
Senesac CR, Lott DJ, Willcocks RJ, Duong T, Smith BK. Lower Extremity Functional Outcome Measures in Duchenne Muscular Dystrophy-A Delphi Survey. J Neuromuscul Dis 2020; 6:75-83. [PMID: 30562905 PMCID: PMC6698889 DOI: 10.3233/jnd-180337] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a progressive neuromuscular disease characterized by progressive muscle weakness, multiple system involvement and premature mortality. Effective treatments for DMD through clinical trials and natural history studies are currently underway. Clinical trials in DMD typically include several outcome measures of motor function. Research sites and studies have been found to have slightly different operational definitions for a given functional outcome resulting in different procedures and protocols for these measurements. OBJECTIVE The goal of this study is to establish agreement among experts in the field around best practices in collecting functional outcome data in DMD providing researchers and clinicians with guidance on best practices. METHODS A group of 30 experts in Duchenne Muscular Dystrophy (DMD) with experience in the development and/ or execution of lower extremity outcome measures for this population met face to face to identify incongruences in the collection of this data. This effort was based in the United States (US) and sponsored by Parent Project Muscular Dystrophy. Several discrepancies were categorized for each outcome which included: 6-minute walk test, 10-meter walk/run, supine to stand, ascend 4 stairs, sit to stand, and the NorthStar Ambulatory Assessment. Following this meeting an additional 32 experts in DMD (28 from the United States and 11 international participants) consented to participate in a Delphi Survey to reach consensus on the protocols and execution of lower extremity outcomes. RESULTS Round one: 70 operationally defined questions were surveyed with 45 (64%) reaching >70% consensus. Round two: 27 questions were operational, with 20 (74%) reaching >70% consensus. Those questions that did not reach consensus appear minor. CONCLUSION With minor modifications in the collection of data across sites, outcomes could potentially be normalized across research studies. This would reduce excessive training for evaluators in trials and produce minimal differences between protocols. Consistency in protocols will promote more efficient study start up, less errors between administration of items across studies, and ultimately improve quality and reliability of the functional outcomes. The authors strongly advocate for the establishment of a "research network library" that could be utilized by all those performing clinical assessments and trials in DMD.
Collapse
Affiliation(s)
- Claudia R. Senesac
- Department of Physical Therapy, College of PHHP, University of Florida Gainesville, FL, USA
| | - Donovan J. Lott
- Department of Physical Therapy, College of PHHP, University of Florida Gainesville, FL, USA
| | - Rebecca J. Willcocks
- Department of Physical Therapy, College of PHHP, University of Florida Gainesville, FL, USA
| | - Tina Duong
- Research Physical Therapist, Stanford Children’s Health, Stanford University School of Medicine, Palo Alto, CA
| | - Barbara K. Smith
- Department of Physical Therapy, College of PHHP, University of Florida Gainesville, FL, USA
| |
Collapse
|
45
|
Spitali P, Zaharieva I, Bohringer S, Hiller M, Chaouch A, Roos A, Scotton C, Claustres M, Bello L, McDonald CM, Hoffman EP, Koeks Z, Eka Suchiman H, Cirak S, Scoto M, Reza M, 't Hoen PAC, Niks EH, Tuffery-Giraud S, Lochmüller H, Ferlini A, Muntoni F, Aartsma-Rus A. TCTEX1D1 is a genetic modifier of disease progression in Duchenne muscular dystrophy. Eur J Hum Genet 2020; 28:815-825. [PMID: 31896777 PMCID: PMC7253478 DOI: 10.1038/s41431-019-0563-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 11/08/2019] [Accepted: 12/03/2019] [Indexed: 11/29/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by pathogenic variants in the DMD gene leading to the lack of dystrophin. Variability in the disease course suggests that other factors influence disease progression. With this study we aimed to identify genetic factors that may account for some of the variability in the clinical presentation. We compared whole-exome sequencing (WES) data in 27 DMD patients with extreme phenotypes to identify candidate variants that could affect disease progression. Validation of the candidate SNPs was performed in two independent cohorts including 301 (BIO-NMD cohort) and 109 (CINRG cohort of European ancestry) DMD patients, respectively. Variants in the Tctex1 domain containing 1 (TCTEX1D1) gene on chromosome 1 were associated with age of ambulation loss. The minor alleles of two independent variants, known to affect TCTEX1D1 coding sequence and induce skipping of its exon 4, were associated with earlier loss of ambulation. Our data show that disease progression of DMD is affected by a new locus on chromosome 1 and demonstrate the possibility to identify genetic modifiers in rare diseases by studying WES data in patients with extreme phenotypes followed by multiple layers of validation.
Collapse
Affiliation(s)
- Pietro Spitali
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| | - Irina Zaharieva
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health, London, UK.
| | - Stefan Bohringer
- Department of Medical Statistics, Leiden University Medical Center, Leiden, The Netherlands
| | - Monika Hiller
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Amina Chaouch
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle upon Tyne, UK.,Greater Manchester Neuroscience Centre, Salford Royal Foundation Trust, Salford, UK
| | - Andreas Roos
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle upon Tyne, UK
| | - Chiara Scotton
- Department of Medical Sciences, Section of Microbiology and Medical Genetics, University of Ferrara, Ferrara, Italy
| | - Mireille Claustres
- Laboratory of Genetics of Rare Diseases (LGMR - EA7402), University of Montpellier, Montpellier, France
| | - Luca Bello
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA.,Department of Neuroscience, University of Padova, Padova, Italy
| | - Craig M McDonald
- University of California Davis Medical Center, Sacramento, CA, USA
| | - Eric P Hoffman
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA
| | | | - Zaida Koeks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - H Eka Suchiman
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sebahattin Cirak
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health, London, UK.,Department of Pediatrics, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Mariacristina Scoto
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Mojgan Reza
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle upon Tyne, UK
| | - Peter A C 't Hoen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sylvie Tuffery-Giraud
- Laboratory of Genetics of Rare Diseases (LGMR - EA7402), University of Montpellier, Montpellier, France
| | - Hanns Lochmüller
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle upon Tyne, UK.,Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, Canada.,Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | - Alessandra Ferlini
- Department of Medical Sciences, Section of Microbiology and Medical Genetics, University of Ferrara, Ferrara, Italy
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health, London, UK.,National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.,John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
46
|
Chrzanowski SM, Darras BT, Rutkove SB. The Value of Imaging and Composition-Based Biomarkers in Duchenne Muscular Dystrophy Clinical Trials. Neurotherapeutics 2020; 17:142-152. [PMID: 31879850 PMCID: PMC7007477 DOI: 10.1007/s13311-019-00825-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
As the drug development pipeline for Duchenne muscular dystrophy (DMD) rapidly advances, clinical trial outcomes need to be optimized. Effective assessment of disease burden, natural history progression, and response to therapy in clinical trials for Duchenne muscular dystrophy are critical factors for clinical trial success. By choosing optimal biomarkers to better assess therapeutic efficacy, study costs and sample size requirements can be reduced. Currently, functional measures continue to serve as the primary outcome for the majority of DMD clinical trials. Quantitative measures of muscle health, including magnetic resonance imaging and spectroscopy, electrical impedance myography, and ultrasound, sensitively identify diseased muscle, disease progression, and response to a therapeutic intervention. Furthermore, such non-invasive techniques have the potential to identify disease pathology prior to onset of clinical symptoms. Despite robust supportive evidence, non-invasive quantitative techniques are still not frequently utilized in clinical trials for Duchenne muscular dystrophy. Non-invasive quantitative techniques have demonstrated the ability to quantify disease progression and potential response to therapeutic intervention, and should be used as a supplement to current standard functional measures. Such methods have the potential to significantly accelerate the development and approval of therapies for DMD.
Collapse
Affiliation(s)
- Stephen M Chrzanowski
- Department of Medicine, Boston Children's Hospital, 300 Longwood Ave., Boston, MA, 02115, USA.
| | - Basil T Darras
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Seward B Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
47
|
Ropars J, Gravot F, Ben Salem D, Rousseau F, Brochard S, Pons C. Muscle MRI: A biomarker of disease severity in Duchenne muscular dystrophy? A systematic review. Neurology 2019; 94:117-133. [PMID: 31892637 DOI: 10.1212/wnl.0000000000008811] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/29/2019] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE To assess the evidence of a relationship between muscle MRI and disease severity in Duchenne muscular dystrophy (DMD). METHODS We conducted a systematic review of studies that analyzed correlations between MRI measurements and motor function in patients with DMD. PubMed, Cochrane, Scopus, and Web of Science were searched using relevant keywords and inclusion/exclusion criteria (January 1, 1990-January 31, 2019). We evaluated article quality using the Joanna Briggs Institute scale. Information regarding the samples included, muscles evaluated, MRI protocols and motor function tests used was collected from each article. Correlations between MRI measurements and motor function were reported exhaustively. RESULTS Seventeen of 1,629 studies identified were included. Most patients included were ambulant with a mean age of 8.9 years. Most studies evaluated lower limb muscles. Moderate to excellent correlations were found between MRI measurements and motor function. The strongest correlations were found for quantitative MRI measurements such as fat fraction or mean T2. Correlations were stronger for lower leg muscles such as soleus. One longitudinal study reported that changes in soleus mean T2 were highly correlated with changes in motor function. CONCLUSION The findings of this systematic review showed that MRI measurements can be used as biomarkers of disease severity in ambulant patients with DMD. Guidelines are proposed to help clinicians choose the most appropriate MRI measurements and muscles to evaluate. Studies exploring upper limb muscles, other stages of the disease, and sensitivity of measurements to change are needed.
Collapse
Affiliation(s)
- Juliette Ropars
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France.
| | - France Gravot
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| | - Douraied Ben Salem
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| | - François Rousseau
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| | - Sylvain Brochard
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| | - Christelle Pons
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| |
Collapse
|
48
|
Romano A, Favetta M, Schirinzi T, Summa S, Minosse S, D'Amico A, Catteruccia M, Petrarca M, Castelli E, Bertini E, Vasco G. Evaluation of gait in Duchenne Muscular Dystrophy: Relation of 3D gait analysis to clinical assessment. Neuromuscul Disord 2019; 29:920-929. [PMID: 31791868 DOI: 10.1016/j.nmd.2019.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 08/26/2019] [Accepted: 10/28/2019] [Indexed: 10/25/2022]
Abstract
Walking ability in Duchenne Muscular Dystrophy (DMD) deteriorates progressively until complete loss of the function. Interventions aimed at maintaining ambulatory ability relies on accurate clinical-based scores and evaluations of walking. This kind of assessment has intrinsic limitations. A 3D optoelectronic system could provide elements useful for the functional evaluation of patients with DMD. Nineteen boys with DMD were evaluated using the 6-Minutes Walking Test, North Star Ambulatory Assessment and 3D gait analysis. Participants' gait parameters were compared to those of an age-matched control group and correlated with standard clinical scores. Seventeen kinematic variables differed between DMD and control groups. Strong correlations with North Star Ambulatory Assessment were found for stride width, gait velocity and ankle angles on the sagittal plane. The 6-Minutes Walking test did not correlate with investigated kinematic variables but showed a correlation with North Star Ambulatory Assessment. Our data support the reported DMD gait pattern characterized by increased anterior pelvic tilt and ankle plantar flexion. The stride width and ankle kinematics emerged as the main representative gait parameters of DMD global ambulatory status. Although preliminary, our findings suggest that 3D gait analysis may provide useful objective and accurate parameters reflecting the functional ability of individuals with DMD.
Collapse
Affiliation(s)
- Alberto Romano
- Department of Neuroscience, Unit of Neurorehabilitation, Movement Analysis and Robotics Laboratory, Bambino Gesù Children's Hospital, Via della torre di Palidoro, snc, Fiumicino, Rome, Italy
| | - Martina Favetta
- Department of Neuroscience, Unit of Neurorehabilitation, Movement Analysis and Robotics Laboratory, Bambino Gesù Children's Hospital, Via della torre di Palidoro, snc, Fiumicino, Rome, Italy
| | - Tommaso Schirinzi
- Department of Neuroscience, Unit of Neurorehabilitation, Movement Analysis and Robotics Laboratory, Bambino Gesù Children's Hospital, Via della torre di Palidoro, snc, Fiumicino, Rome, Italy; Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Susanna Summa
- Department of Neuroscience, Unit of Neurorehabilitation, Movement Analysis and Robotics Laboratory, Bambino Gesù Children's Hospital, Via della torre di Palidoro, snc, Fiumicino, Rome, Italy
| | - Silvia Minosse
- Department of Neuroscience, Unit of Neurorehabilitation, Movement Analysis and Robotics Laboratory, Bambino Gesù Children's Hospital, Via della torre di Palidoro, snc, Fiumicino, Rome, Italy
| | - Adele D'Amico
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Disease, Bambino Gesù Children's Hospital, Rome, Italy
| | - Michela Catteruccia
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Disease, Bambino Gesù Children's Hospital, Rome, Italy
| | - Maurizio Petrarca
- Department of Neuroscience, Unit of Neurorehabilitation, Movement Analysis and Robotics Laboratory, Bambino Gesù Children's Hospital, Via della torre di Palidoro, snc, Fiumicino, Rome, Italy.
| | - Enrico Castelli
- Department of Neuroscience, Unit of Neurorehabilitation, Movement Analysis and Robotics Laboratory, Bambino Gesù Children's Hospital, Via della torre di Palidoro, snc, Fiumicino, Rome, Italy
| | - Enrico Bertini
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Disease, Bambino Gesù Children's Hospital, Rome, Italy
| | - Gessica Vasco
- Department of Neuroscience, Unit of Neurorehabilitation, Movement Analysis and Robotics Laboratory, Bambino Gesù Children's Hospital, Via della torre di Palidoro, snc, Fiumicino, Rome, Italy
| |
Collapse
|
49
|
Longitudinal natural history in young boys with Duchenne muscular dystrophy. Neuromuscul Disord 2019; 29:857-862. [PMID: 31629611 DOI: 10.1016/j.nmd.2019.09.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 11/23/2022]
Abstract
The aim of this prospective multicentric study was to document disease progression in young boys affected by Duchenne muscular dystrophy (DMD) between age 3 and 6 years (±3 months) using the North Star Ambulatory Assessment scale. One hundred fifty-three DMD boys (573 assessments) younger than 6 years (mean: 4.68, SD: 0.84) with a genetically proven DMD diagnoses were included. Our results showed North Star Ambulatory Assessment scores progressively increased with age. The largest increase was observed between age 3 and 4 years but further increase was steadily observed until age of 6 years. Using a multiple linear regression analysis, we found that both the use of corticosteroids and the site of mutation significantly contributed to the North Star Ambulatory Assessment changes (p < 0.001). At each age point, boys on corticosteroid treatment had higher scores than corticosteroid naïve ones (p < 0.001). Similarly, patients with mutations downstream exon 44, had lower baseline scores and lower magnitude of changes compared to those with mutations located at the 5' end of the gene (p < 0,001). Very few boys achieved the age appropriate maximum score. These results provide useful information for the assessment and counselling of young DMD boys and for the design of clinical trials in this age group.
Collapse
|
50
|
Muntoni F, Domingos J, Manzur AY, Mayhew A, Guglieri M, The UK NorthStar Network, Sajeev G, Signorovitch J, Ward SJ. Categorising trajectories and individual item changes of the North Star Ambulatory Assessment in patients with Duchenne muscular dystrophy. PLoS One 2019; 14:e0221097. [PMID: 31479456 PMCID: PMC6719875 DOI: 10.1371/journal.pone.0221097] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 07/30/2019] [Indexed: 01/16/2023] Open
Abstract
Functional variability among boys with Duchenne muscular dystrophy (DMD) is well recognised and complicates interpretation of clinical studies. We hypothesised that boys with DMD could be clustered into groups sharing similar trajectories of ambulatory function over time, as measured by the North Star Ambulatory Assessment (NSAA) total score. We also explored associations with other variables such as age, functional abilities, and genotype. Using the NorthStar Clinical Network database, 395 patients with >1 NSAA assessment were identified. We utilised latent class trajectory analysis of longitudinal NSAA scores, which produced evidence for at least four clusters of boys sharing similar trajectories versus age in decreasing order of clinical severity: 25% of the boys were in cluster 1 (NSAA falling to ≤ 5 at age ~10y), 35% were in cluster 2 (NSAA ≤ 5 ~12y), 21% in were cluster 3 (NSAA≤ 5 ~14y), and 19% in cluster 4 (NSAA > 5 up to 15y). Mean ages at diagnosis of DMD were similar across clusters (4.2, 3.9, 4.3, and 4.8y, respectively). However, at the first NSAA assessment, a significant (p<0.05) association was observed between earlier declining clusters and younger age, worse NSAA, slower rise from supine, slower 10 metre walk/run times, and younger age of steroid initiation. In order to assess the probability of observing complete loss of function for individual NSAA items, we examined the proportion of patients who shifted from a score of 1 or 2 at baseline to a score of 0. We also assessed the probability of gain of function using the inverse assessment and stratified the probability of deterioration, improvement-or static behavior-by age ranges and using baseline functional status. Using this tool, our study provides a comprehensive assessment of the NSAA in a large population of patients with DMD and, for the first time, describes discrete clusters of disease progression; this will be invaluable for future DMD clinical trial design and interpretation of findings.
Collapse
Affiliation(s)
- Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, United Kingdom
- National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- * E-mail:
| | - Joana Domingos
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, United Kingdom
| | - Adnan Y. Manzur
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, United Kingdom
| | - Anna Mayhew
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle, United Kingdom
| | - Michela Guglieri
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle, United Kingdom
| | | | - Gautam Sajeev
- Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - James Signorovitch
- Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Susan J. Ward
- Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
| |
Collapse
|