1
|
Jenkins BM, Dixon LD, Kokesh KJ, Zingariello CD, Vandenborne K, Walter GA, Barnard AM. Skeletal muscle symptoms and quantitative MRI in females with dystrophinopathy. Muscle Nerve 2024; 70:988-999. [PMID: 39221574 PMCID: PMC11493146 DOI: 10.1002/mus.28235] [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: 12/01/2023] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION/AIMS The dystrophinopathies primarily affect males; however, female carriers of pathogenic dystrophin variants can develop skeletal muscle symptoms. This study aimed to evaluate muscle involvement and symptoms in females with dystrophinopathy using quantitative magnetic resonance imaging (MRI), functional assessments, and patient-reported outcomes. METHODS Controls and females with dystrophinopathy with muscle symptoms of pain, weakness, fatigue, or excessive tightness were enrolled in this cross-sectional study. Participants underwent lower extremity MRI to quantify muscle inflammation, replacement by fat, and disease asymmetry. Cardiac MRI, functional ability, muscle symptoms, and serum creatine kinase levels were also evaluated. RESULTS Six pediatric females with dystrophinopathy (mean age: 11.7 years), 11 adult females with dystrophinopathy (mean age: 41.3 years), and seven controls enrolled. The mean fat fraction was increased in females with dystrophinopathy compared to controls in the soleus (0.11 vs. 0.03, p = .0272) and vastus lateralis (0.16 vs. 0.03, p = .004). Magnetic resonance spectroscopy water T2, indicative of muscle inflammation, was elevated in the soleus and/or vastus lateralis in 11 of 17 individuals. North Star Ambulatory Assessment score was lower in the dystrophinopathy group compared to controls (29 vs. 34 points, p = .0428). From cardiac MRI, left ventricle T1 relaxation times were elevated in females with dystrophinopathy compared to controls (1311 ± 55 vs. 1263 ± 25 ms, p < .05), but ejection fraction and circumferential strain did not differ. DISCUSSION Symptomatic females with dystrophinopathy quantitatively demonstrate muscle replacement by fat and inflammation, along with impairments in functional ability and cardiac function. Additional research is needed to evaluate how symptoms and muscle involvement change longitudinally.
Collapse
Affiliation(s)
| | | | - Kevin J Kokesh
- Department of Pediatrics, Division of Pulmonology; University of Florida
| | - Carla D Zingariello
- Department of Pediatrics, Division of Pediatric Neurology; University of Florida
| | | | - Glenn A Walter
- Department of Physiology and Aging; University of Florida
| | | |
Collapse
|
2
|
Willis AB, Zelikovich AS, Sufit R, Ajroud-Driss S, Vandenborne K, Demonbreun AR, Batra A, Walter GA, McNally EM. Serum protein and imaging biomarkers after intermittent steroid treatment in muscular dystrophy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.14.24308858. [PMID: 38947030 PMCID: PMC11213068 DOI: 10.1101/2024.06.14.24308858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background Weekly Steroids in Muscular Dystrophy (WSiMD) was a pilot study to evaluate once weekly prednisone in patients with Limb Girdle and Becker muscular dystrophy (LGMD and BMD, respectively). At study endpoint, there were trends towards increased lean mass, reduced fat mass, reduced creatine kinase and improved motor function. The investigation was motivated by studies in mouse muscular dystrophy models in which once weekly glucocorticoid exposure enhanced muscle strength and reduced fibrosis. Methods WSiMD participants provided blood samples for aptamer serum profiling at baseline and after 6 months of weekly steroids. A subset completed magnetic resonance (MR) evaluation of muscle at study onset and endpoint. Results/Conclusions At baseline compared to age and sex-matched healthy controls, the aggregate serum protein profile in the WSiMD cohort was dominated by muscle proteins, reflecting leak of muscle proteins into serum. Disease status produced more proteins differentially present in serum compared to steroid-treatment effect. Nonetheless, a response to prednisone was discernable in the WSiMD cohort, even at this low dose. Glucocorticoids downregulated muscle proteins and upregulated certain immune process- and matrix-associated proteins. Muscle MR fat fraction showed trends with functional status. The prednisone-responsive markers could be used in larger trial of prednisone efficacy.
Collapse
Affiliation(s)
- Alexander B. Willis
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Aaron S. Zelikovich
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Robert Sufit
- Dept of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Senda Ajroud-Driss
- Dept of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Alexis R. Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Abhinandan Batra
- Department of Physical Therapy, University of Louisiana at Monroe, Monroe, LA
| | - Glenn A. Walter
- Department of Physiology and Aging, University of Florida, Gainesville, FL
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
3
|
Schiava M, McDermott MP, Broomfield J, Abrams KR, Mayhew AG, McDonald CM, Martens WB, Gregory SJ, Griggs RC, Guglieri M. Factors Associated With Early Motor Function Trajectories in DMD After Glucocorticoid Initiation: Post Hoc Analysis of the FOR-DMD Trial. Neurology 2024; 102:e209206. [PMID: 38710006 DOI: 10.1212/wnl.0000000000209206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Clinical trials in Duchenne muscular dystrophy (DMD) require 3-6 months of stable glucocorticoids, and the primary outcome is explored at 48-52 weeks. The factors that influence the clinical outcome assessment (COA) trajectories soon after glucocorticoid initiation are relevant for the design and analysis of clinical trials of novel drugs. We describe early COA trajectories, associated factors, and the time from glucocorticoid initiation to COA peak. METHODS This was a prospective 18-month analysis of the Finding the Optimum Corticosteroid Regimen for Duchenne Muscular Dystrophy study. Four COAs were investigated: rise from supine velocity (RFV), 10-meter walk/run velocity (10MWRV), North Star Ambulatory Assessment (NSAA) total score, and 6-minute walk test distance (6MWT). The relationships of baseline age (4-5 vs 6-7 years), COA baseline performance, genotype, and glucocorticoid regimen (daily vs intermittent) with the COA trajectories were evaluated using linear mixed-effects models. RESULTS One hundred ninety-six glucocorticoid-naïve boys with DMD aged 4-7 years were enrolled. The mean age at baseline was 5.9 ± 1.0 years, 66% (n = 130) were on daily regimens, 55% (n = 107) showed a 6MWT distance >330 metres; 41% (n = 78) showed RFV >0.2 rise/s; 76% (n = 149) showed 10MWRV >0.142 10m/s, and 41.0% (n = 79) showed NSAA total score >22 points. Mean COA trajectories differed by age at glucocorticoid initiation (p < 0.01 for RFV, 10MWRV, and NSAA; p < 0.05 for 6MWT) and regimen (p < 0.01 for RFV, 10MWRV, and NSAA). Boys younger than 6 years reached their peak performance 12-18 months after glucocorticoid initiation. Boys aged 6 years or older on a daily regimen peaked between months 9 and 12 and those on an intermittent regimen by 9 months. The baseline COA performance was associated with the NSAA (p < 0.01) and the 6MWT trajectory in boys younger than 6 years on a daily regimen (p < 0.01). Differences in the mean trajectories by genotype were not significant. DISCUSSION Glucocorticoid regimen, age, duration of glucocorticoid exposure, and baseline COA performance need to be considered in the design and analysis of clinical trials in young boys with DMD.
Collapse
Affiliation(s)
- Marianela Schiava
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Michael P McDermott
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Jonathan Broomfield
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Keith R Abrams
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Anna G Mayhew
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Craig M McDonald
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - William B Martens
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Stephanie J Gregory
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Robert C Griggs
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Michela Guglieri
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| |
Collapse
|
4
|
Nava S, Conte G, Triulzi FM, Comi GP, Magri F, Velardo D, Cinnante CM. Diffusion tensor imaging reveals subclinical alterations in muscles of patients with Becker muscular dystrophy. Br J Radiol 2024; 97:947-953. [PMID: 38574384 DOI: 10.1093/bjr/tqae070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 11/21/2023] [Accepted: 03/27/2024] [Indexed: 04/06/2024] Open
Abstract
OBJECTIVES Becker muscular dystrophy (BMD) is a relatively less investigated neuromuscular disease, partially overlapping the phenotype of Duchenne dystrophy (DMD). Physiopathological and anatomical patterns are still not comprehensively known, despite recent effort in the search of early biomarkers. Aim of this study was to selectively compare normal appearing muscles of BMD with healthy controls. METHODS Among a pool of 40 BMD patients and 20 healthy controls, Sartorius and gracilis muscles were selected on the basis of a blinded clinical quantitative/qualitative evaluation, if classified as normal (0 or 1 on Mercuri scale) and subsequently segmented on diffusion tensor MRI scans with a tractographic approach. Diffusion derived parameters were extracted. RESULTS Non-parametric testing revealed significant differences between normal and normal appearing BMD derived parameters in both muscles, the difference being more evident in sartorius. Bonferroni-corrected P-values (<.05) of Mann-Whitney test could discriminate between BMD and controls for standard deviation of all diffusion parameters (mean diffusivity, fractional anisotropy, axial and radial diffusivity) in both sartorius and gracilis, while in sartorius the significant difference was found also in the average values of the same parameters (with exception of RD). CONCLUSIONS This method could identify microstructural alterations in BMD normal appearing sartorius and gracilis. ADVANCES IN KNOWLEDGE Diffusion based MRI could be able to identify possible early or subclinical microstructural alterations in dystrophic patients with BMD.
Collapse
Affiliation(s)
- Simone Nava
- Neuroradiology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| | - Giorgio Conte
- Neuroradiology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| | - Fabio M Triulzi
- Neuroradiology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| | - Giacomo P Comi
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, via Francesco Sforza 35, 20122 MilanItaly
| | - Francesca Magri
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, via Francesco Sforza 35, 20122 MilanItaly
| | - Daniele Velardo
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, via Francesco Sforza 35, 20122 MilanItaly
| | - Claudia M Cinnante
- Radiology Department, Istituto Auxologico Italiano IRCCS, Piazzale Brescia 20, 20149 Milan, Italy
| |
Collapse
|
5
|
Mercuri E, Vilchez JJ, Boespflug-Tanguy O, Zaidman CM, Mah JK, Goemans N, Müller-Felber W, Niks EH, Schara-Schmidt U, Bertini E, Comi GP, Mathews KD, Servais L, Vandenborne K, Johannsen J, Messina S, Spinty S, McAdam L, Selby K, Byrne B, Laverty CG, Carroll K, Zardi G, Cazzaniga S, Coceani N, Bettica P, McDonald CM. Safety and efficacy of givinostat in boys with Duchenne muscular dystrophy (EPIDYS): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Neurol 2024; 23:393-403. [PMID: 38508835 DOI: 10.1016/s1474-4422(24)00036-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Duchenne muscular dystrophy, the most common childhood muscular dystrophy, is caused by dystrophin deficiency. Preclinical and phase 2 study data have suggested that givinostat, a histone deacetylase inhibitor, might help to counteract the effects of this deficiency. We aimed to evaluate the safety and efficacy of givinostat in the treatment of Duchenne muscular dystrophy. METHODS This multicentre, randomised, double-blind, placebo-controlled, phase 3 trial was done at 41 tertiary care sites in 11 countries. Eligible participants were ambulant, male, and aged at least 6 years, had a genetically confirmed diagnosis of Duchenne muscular dystrophy, completed two four-stair climb assessments with a mean of 8 s or less (≤1 s variance), had a time-to-rise of at least 3 s but less than 10 s, and had received systemic corticosteroids for at least 6 months. Participating boys were randomly assigned (2:1, allocated according to a list generated by the interactive response technology provider) to receive either oral givinostat or matching placebo twice a day for 72 weeks, stratified by concomitant steroid use. Boys, investigators, and site and sponsor staff were masked to treatment assignment. The dose was flexible, based on weight, and was reduced if not tolerated. Boys were divided into two groups on the basis of their baseline vastus lateralis fat fraction (VLFF; measured by magnetic resonance spectroscopy): group A comprised boys with a VLFF of more than 5% but no more than 30%, whereas group B comprised boys with a VLFF of 5% or less, or more than 30%. The primary endpoint compared the effects of givinostat and placebo on the change in results of the four-stair climb assessment between baseline and 72 weeks, in the intention-to-treat, group A population. Safety was assessed in all randomly assigned boys who received at least one dose of study drug. When the first 50 boys in group A completed 12 months of treatment, an interim futility assessment was conducted, after which the sample size was adapted using masked data from the four-stair climb assessments. Furthermore, the starting dose of givinostat was reduced following a protocol amendment. This trial is registered with ClinicalTrials.gov, NCT02851797, and is complete. FINDINGS Between June 6, 2017, and Feb 22, 2022, 359 boys were assessed for eligibility. Of these, 179 were enrolled into the study (median age 9·8 years [IQR 8·1-11·0]), all of whom were randomly assigned (118 to receive givinostat and 61 to receive placebo); 170 (95%) boys completed the study. Of the 179 boys enrolled, 120 (67%) were in group A (81 givinostat and 39 placebo); of these, 114 (95%) completed the study. For participants in group A, comparing the results of the four-stair climb assessment at 72 weeks and baseline, the geometric least squares mean ratio was 1·27 (95% CI 1·17-1·37) for boys receiving givinostat and 1·48 (1·32-1·66) for those receiving placebo (ratio 0·86, 95% CI 0·745-0·989; p=0·035). The most common adverse events in the givinostat group were diarrhoea (43 [36%] of 118 boys vs 11 [18%] of 61 receiving placebo) and vomiting (34 [29%] vs 8 [13%]); no treatment-related deaths occurred. INTERPRETATION Among ambulant boys with Duchenne muscular dystrophy, results of the four-stair climb assessment worsened in both groups over the study period; however, the decline was significantly smaller with givinostat than with placebo. The dose of givinostat was reduced after an interim safety analysis, but no new safety signals were reported. An ongoing extension study is evaluating the long-term safety and efficacy of givinostat in patients with Duchenne muscular dystrophy. FUNDING Italfarmaco.
Collapse
Affiliation(s)
- Eugenio Mercuri
- Pediatric Neurology, Department of Woman and Child Health and Public Health, Child Health Area, Universita Cattolica del Sacro Cuore, Rome, Italy; Centro Clinico Nemo Fondazione Policlinico Gemelli IRCCS, Rome, Italy.
| | - Juan J Vilchez
- Servicio de Neurología, Neuromuscular Unit, CIBERER, EURO-RN-NMD, Hospital Universitario y Politécnico La Fe Valencia, Valencia, Spain
| | - Odile Boespflug-Tanguy
- I-Motion, Institut de Myologie, Hôpital Armand-Trousseau, APHP, Sorbonne Université, Paris, France; Université Paris Cité UMR INSERM 1141, Hôpital Robert Debré, Paris, France
| | | | - Jean K Mah
- Division of Pediatric Neurology, Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nathalie Goemans
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Wolfgang Müller-Felber
- LMU Munich, University Hospital, Hauner Children's Hospital, Pediatric Neurology and Developmental Medicine, Munich, Germany
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands; Duchenne Center Netherlands, Netherlands
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology, Children's University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Enrico Bertini
- Research Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giacomo P Comi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Milan, Italy; Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Katherine D Mathews
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Laurent Servais
- MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; Neuromuscular Reference Center, Department of Paediatrics, University and University Hospital of Liege, Belgium
| | - Krista Vandenborne
- ImagingDMD, University of Florida, Gainesville, FL, USA; Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Jessika Johannsen
- University Medical Center Hamburg-Eppendorf, Department of Pediatrics, Hamburg, Germany
| | - Sonia Messina
- Department of Clinical and Experimental Medicine, Unit of Neurodegenerative Diseases, AOU Policlinico G Martino, University of Mesina, Messina, Italy
| | - Stefan Spinty
- Department of Paediatric Neurology, Alder Hey Children's Hospital NHS Trust, Liverpool, UK
| | - Laura McAdam
- Holland Bloorview Kids Rehabilitation Hospital, Bloorview Research Institute, Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Kathryn Selby
- The University of British Columbia, Children's and Women's Health Centre, Vancouver, BC, Canada
| | - Barry Byrne
- Child Health Research Institute, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Chamindra G Laverty
- Department of Neuroscience, University of California, San Diego, San Diego, CA, USA
| | | | | | | | | | | | | |
Collapse
|
6
|
Dabaj I, Ducatez F, Marret S, Bekri S, Tebani A. Neuromuscular disorders in the omics era. Clin Chim Acta 2024; 553:117691. [PMID: 38081447 DOI: 10.1016/j.cca.2023.117691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023]
Abstract
Neuromuscular disorders encompass a spectrum of conditions characterized by primary lesions within the peripheral nervous system, which include the anterior horn cell, peripheral nerve, neuromuscular junction, and muscle. In pediatrics, most of these disorders are linked to genetic causes. Despite the considerable progress, the diagnosis of these disorders remains a challenging due to wide clinical presentation, disease heterogeneity and rarity. It is noteworthy that certain neuromuscular disorders, once deemed untreatable, can now be effectively managed through novel therapies. Biomarkers emerge as indispensable tools, serving as objective measures that not only refine diagnostic accuracy but also provide guidance for therapeutic decision-making and the ongoing monitoring of long-term outcomes. Herein a comprehensive review of biomarkers in neuromuscular disorders is provided. We highlight the role of omics-based technologies that further characterize neuromuscular pathophysiology as well as identify potential therapeutic targets to guide treatment strategies.
Collapse
Affiliation(s)
- Ivana Dabaj
- Normandie Univ, UNIROUEN, INSERM U1245, Nord/Est/Ile de France Neuromuscular Reference Center CHU Rouen, Department of Neonatalogy, Pediatric Intensive Care, and Neuropediatrics, F-76000 Rouen, France.
| | - Franklin Ducatez
- Normandie Univ, UNIROUEN, INSERM U1245, Nord/Est/Ile de France Neuromuscular Reference Center CHU Rouen, Department of Neonatalogy, Pediatric Intensive Care, and Neuropediatrics, F-76000 Rouen, France
| | - Stéphane Marret
- Normandie Univ, UNIROUEN, INSERM U1245, Nord/Est/Ile de France Neuromuscular Reference Center CHU Rouen, Department of Neonatalogy, Pediatric Intensive Care, and Neuropediatrics, F-76000 Rouen, France
| | - Soumeya Bekri
- Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, Department of Metabolic Biochemistry, F-76000 Rouen, France
| | - Abdellah Tebani
- Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, Department of Metabolic Biochemistry, F-76000 Rouen, France
| |
Collapse
|
7
|
Willcocks RJ, Barnard AM, Daniels MJ, Forbes SC, Triplett WT, Brandsema JF, Finanger EL, Rooney WD, Kim S, Wang D, Lott DJ, Senesac CR, Walter GA, Sweeney HL, Vandenborne K. Clinical importance of changes in magnetic resonance biomarkers for Duchenne muscular dystrophy. Ann Clin Transl Neurol 2024; 11:67-78. [PMID: 37932907 PMCID: PMC10791017 DOI: 10.1002/acn3.51933] [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: 04/17/2023] [Revised: 10/03/2023] [Accepted: 10/08/2023] [Indexed: 11/08/2023] Open
Abstract
OBJECTIVE Magnetic resonance (MR) measures of muscle quality are highly sensitive to disease progression and predictive of meaningful functional milestones in Duchenne muscular dystrophy (DMD). This investigation aimed to establish the reproducibility, responsiveness to disease progression, and minimum clinically important difference (MCID) for multiple MR biomarkers at different disease stages in DMD using a large natural history dataset. METHODS Longitudinal MR imaging and spectroscopy outcomes and ambulatory function were measured in 180 individuals with DMD at three sites, including repeated measurements on two separate days (within 1 week) in 111 participants. These data were used to calculate day-to-day reproducibility, responsiveness (standardized response mean, SRM), minimum detectable change, and MCID. A survey of experts was also performed. RESULTS MR spectroscopy fat fraction (FF), as well as MR imaging transverse relaxation time (MRI-T2 ), measures performed in multiple leg muscles, and had high reproducibility (Pearson's R > 0.95). Responsiveness to disease progression varied by disease stage across muscles. The average FF from upper and lower leg muscles was highly responsive (SRM > 0.9) in both ambulatory and nonambulatory individuals. MCID estimated from the distribution of scores, by anchoring to function, and via expert opinion was between 0.01 and 0.05 for FF and between 0.8 and 3.7 ms for MRI-T2 . INTERPRETATION MR measures of FF and MRI T2 are reliable and highly responsive to disease progression. The MCID for MR measures is less than or equal to the typical annualized change. These results confirm the suitability of these measures for use in DMD and potentially other muscular dystrophies.
Collapse
Affiliation(s)
- Rebecca J. Willcocks
- Department of Physical Therapy, College of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
| | - Alison M. Barnard
- Department of Physical Therapy, College of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
| | | | - Sean C. Forbes
- Department of Physical Therapy, College of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
| | - William T. Triplett
- Department of Physical Therapy, College of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
| | - John F. Brandsema
- Division of NeurologyThe Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Erika L. Finanger
- Department of Pediatrics and NeurologyOregon Health & Science UniversityPortlandOregonUSA
| | - William D. Rooney
- Advanced Imaging Research CenterOregon Health & Science UniversityPortlandOregonUSA
| | - Sarah Kim
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Dah‐Jyuu Wang
- Department of RadiologyChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Donovan J. Lott
- Department of Physical Therapy, College of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
| | - Claudia R. Senesac
- Department of Physical Therapy, College of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
| | - Glenn A. Walter
- Department of Physiology and Functional Genomics, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - H. Lee Sweeney
- Department of Pharmacology and Therapeutics, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Krista Vandenborne
- Department of Physical Therapy, College of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
8
|
McDonald C, Camino E, Escandon R, Finkel RS, Fischer R, Flanigan K, Furlong P, Juhasz R, Martin AS, Villa C, Sweeney HL. Draft Guidance for Industry Duchenne Muscular Dystrophy, Becker Muscular Dystrophy, and Related Dystrophinopathies - Developing Potential Treatments for the Entire Spectrum of Disease. J Neuromuscul Dis 2024; 11:499-523. [PMID: 38363616 DOI: 10.3233/jnd-230219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Background Duchenne muscular dystrophy (DMD) and related dystrophinopathies are neuromuscular conditions with great unmet medical needs that require the development of effective medical treatments. Objective To aid sponsors in clinical development of drugs and therapeutic biological products for treating DMD across the disease spectrum by integrating advancements, patient registries, natural history studies, and more into a comprehensive guidance. Methods This guidance emerged from collaboration between the FDA, the Duchenne community, and industry stakeholders. It entailed a structured approach, involving multiple committees and boards. From its inception in 2014, the guidance underwent revisions incorporating insights from gene therapy studies, cardiac function research, and innovative clinical trial designs. Results The guidance provides a deeper understanding of DMD and its variants, focusing on patient engagement, diagnostic criteria, natural history, biomarkers, and clinical trials. It underscores patient-focused drug development, the significance of dystrophin as a biomarker, and the pivotal role of magnetic resonance imaging in assessing disease progression. Additionally, the guidance addresses cardiomyopathy's prominence in DMD and the burgeoning field of gene therapy. Conclusions The updated guidance offers a comprehensive understanding of DMD, emphasizing patient-centric approaches, innovative trial designs, and the importance of biomarkers. The focus on cardiomyopathy and gene therapy signifies the evolving realm of DMD research. It acts as a crucial roadmap for sponsors, potentially leading to improved treatments for DMD.
Collapse
Affiliation(s)
| | - Eric Camino
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Rafael Escandon
- DGBI Consulting, LLC, Bainbridge Island, Washington, DC, USA
| | | | - Ryan Fischer
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Kevin Flanigan
- Center for Experimental Neurotherapeutics, Department of Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Pat Furlong
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Rose Juhasz
- Nationwide Children's Hospital, Columbus, OH, USA
| | - Ann S Martin
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Chet Villa
- Trinity Health Michigan, Grand Rapids, MI, USA
| | - H Lee Sweeney
- Cincinnati Children's Hospital Medical Center within the UC Department of Pediatrics, Cincinnati, OH, USA
| |
Collapse
|
9
|
Song Y, Xu K, Xu HY, Guo YK, Xu R, Fu H, Yuan WF, Zhou ZQ, Xu T, Chen XJ, Wang YL, Fu C, Zhou H, Cai XT, Li XS. Longitudinal changes in magnetic resonance imaging biomarkers of the gluteal muscle groups and functional ability in Duchenne muscular dystrophy: a 12-month cohort study. Pediatr Radiol 2023; 53:2672-2682. [PMID: 37889296 PMCID: PMC10697878 DOI: 10.1007/s00247-023-05791-7] [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: 07/01/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Quantitative magnetic resonance imaging (MRI) is considered an objective biomarker of Duchenne muscular dystrophy (DMD), but the longitudinal progression of MRI biomarkers in gluteal muscle groups and their predictive value for future motor function have not been described. OBJECTIVE To explore MRI biomarkers of the gluteal muscle groups as predictors of motor function decline in DMD by characterizing the progression over 12 months. MATERIALS AND METHODS A total of 112 participants with DMD were enrolled and underwent MRI examination of the gluteal muscles to determine fat fraction and longitudinal relaxation time (T1). Investigations were based on gluteal muscle groups including flexors, extensors, adductors, and abductors. The North Star Ambulatory Assessment and timed functional tests were performed. All participants returned for follow-up at an average of 12 months and were divided into two subgroups (functional stability/decline groups) based on changes in timed functional tests. Univariable and multivariable logistic regression methods were used to explore the risk factors associated with future motor function decline. RESULTS For the functional decline group, all T1 values decreased, while fat fraction values increased significantly over 12 months (P<0.05). For the functional stability group, only the fat fraction of the flexors and abductors increased significantly over 12 months (P<0.05). The baseline T1 value was positively correlated with North Star Ambulatory Assessment and negatively correlated with timed functional tests at the 12-month follow-up (P<0.001), while the baseline fat fraction value was negatively correlated with North Star Ambulatory Assessment and positively correlated with timed functional tests at the 12-month follow-up (P<0.001). Multivariate regression showed that increased fat fraction of the abductors was associated with future motor function decline (model 1: odds ratio [OR]=1.104, 95% confidence interval [CI]: 1.026~1.187, P=0.008; model 2: OR=1.085, 95% CI: 1.013~1.161, P=0.019), with an area under the curve of 0.874. CONCLUSION Fat fraction of the abductors is a powerful predictor of future motor functional decline in DMD patients at 12 months, underscoring the importance of focusing early on this parameter in patients with DMD.
Collapse
Affiliation(s)
- Yu Song
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ke Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hua-Yan Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying-Kun Guo
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Rong Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hang Fu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei-Feng Yuan
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Zi-Qi Zhou
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xi-Jian Chen
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Yi-Lei Wang
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuan Fu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hui Zhou
- Department of Rehabilitation Medicine, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiao-Tang Cai
- Department of Rehabilitation Medicine, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xue-Sheng Li
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Cardone N, Taglietti V, Baratto S, Kefi K, Periou B, Gitiaux C, Barnerias C, Lafuste P, Pharm FL, Pharm JN, Panicucci C, Desguerre I, Bruno C, Authier FJ, Fiorillo C, Relaix F, Malfatti E. Myopathologic trajectory in Duchenne muscular dystrophy (DMD) reveals lack of regeneration due to senescence in satellite cells. Acta Neuropathol Commun 2023; 11:167. [PMID: 37858263 PMCID: PMC10585739 DOI: 10.1186/s40478-023-01657-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/19/2023] [Indexed: 10/21/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating X-linked muscular disease, caused by mutations in the DMD gene encoding Dystrophin and affecting 1:5000 boys worldwide. Lack of Dystrophin leads to progressive muscle wasting and degeneration resulting in cardiorespiratory failure. Despite the absence of a definitive cure, innovative therapeutic avenues are emerging. Myopathologic studies are important to further understand the biological mechanisms of the disease and to identify histopathologic benchmarks for clinical evaluations. We conducted a myopathologic analysis on twenty-four muscle biopsies from DMD patients, with particular emphasis on regeneration, fibro-adipogenic progenitors and muscle stem cells behavior. We describe an increase in content of fibro-adipogenic progenitors, central orchestrators of fibrotic progression and lipid deposition, concurrently with a decline in muscle regenerative capacity. This regenerative impairment strongly correlates with compromised activation and expansion of muscle stem cells. Furthermore, our study uncovers an early acquisition of a senescence phenotype by DMD-afflicted muscle stem cells. Here we describe the myopathologic trajectory intrinsic to DMD and establish muscle stem cell senescence as a pivotal readout for future therapeutic interventions.
Collapse
Affiliation(s)
| | | | - Serena Baratto
- Centre of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Kaouthar Kefi
- Univ Paris Est Creteil, INSERM, IMRB, 94010, Creteil, France
| | - Baptiste Periou
- Univ Paris Est Creteil, INSERM, IMRB, 94010, Creteil, France
- APHP, Filnemus, EuroNMD, Centre de Référence de Pathologie Neuromusculaire Nord-Est-Ile-de-France, Henri Mondor Hospital, Paris, France
| | - Ciryl Gitiaux
- Neurophysiologie clinique pédiatrique, Centre de référence des maladies neuromusculaires Hôpital universitaire Necker-Enfants Malades-Paris, Centre de Référence de Pathologie Neuromusculaire Nord-Est-Ile-de-France, Henri Mondor Hospital, Université Paris Est, U955 INSERM, IMRB, APHP, Creteil, France
- Reference Center for Neuromuscular Disorders, Filnemus, EuroNMD, Assistance Publique-Hôpitaux de Paris (APHP) Necker Enfants Malades Hospital, Paris, France
| | - Christine Barnerias
- Reference Center for Neuromuscular Disorders, Filnemus, EuroNMD, Assistance Publique-Hôpitaux de Paris (APHP) Necker Enfants Malades Hospital, Paris, France
| | - Peggy Lafuste
- Univ Paris Est Creteil, INSERM, IMRB, 94010, Creteil, France
| | - France Leturcq Pharm
- Service de Médecine Génomique, Maladies de Système et d'Organe - Fédération de Génétique et de Médecine Génomique, DMU BioPhyGen, APHP Centre-Université Paris Cité - Hôpital Cochin, Paris, France
| | - Juliette Nectoux Pharm
- Service de Médecine Génomique, Maladies de Système et d'Organe - Fédération de Génétique et de Médecine Génomique, DMU BioPhyGen, APHP Centre-Université Paris Cité - Hôpital Cochin, Paris, France
| | - Chiara Panicucci
- Centre of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Isabelle Desguerre
- Reference Center for Neuromuscular Disorders, Filnemus, EuroNMD, Assistance Publique-Hôpitaux de Paris (APHP) Necker Enfants Malades Hospital, Paris, France
| | - Claudio Bruno
- Centre of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health-DINOGMI, University of Genova, Genoa, Italy
| | - François-Jerome Authier
- Univ Paris Est Creteil, INSERM, IMRB, 94010, Creteil, France
- APHP, Filnemus, EuroNMD, Centre de Référence de Pathologie Neuromusculaire Nord-Est-Ile-de-France, Henri Mondor Hospital, Paris, France
| | - Chiara Fiorillo
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health-DINOGMI, University of Genova, Genoa, Italy
- Child Neuropsychiatry, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Frederic Relaix
- Univ Paris Est Creteil, INSERM, IMRB, 94010, Creteil, France.
| | - Edoardo Malfatti
- Univ Paris Est Creteil, INSERM, IMRB, 94010, Creteil, France.
- APHP, Filnemus, EuroNMD, Centre de Référence de Pathologie Neuromusculaire Nord-Est-Ile-de-France, Henri Mondor Hospital, Paris, France.
| |
Collapse
|
11
|
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: 0] [Impact Index Per Article: 0] [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
|
12
|
Stephenson D, Belfiore-Oshan R, Karten Y, Keavney J, Kwok DK, Martinez T, Montminy J, Müller MLTM, Romero K, Sivakumaran S. Transforming Drug Development for Neurological Disorders: Proceedings from a Multidisease Area Workshop. Neurotherapeutics 2023; 20:1682-1691. [PMID: 37823970 PMCID: PMC10684834 DOI: 10.1007/s13311-023-01440-x] [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] [Accepted: 09/06/2023] [Indexed: 10/13/2023] Open
Abstract
Neurological disorders represent some of the most challenging therapeutic areas for successful drug approvals. The escalating global burden of death and disability for such diseases represents a significant worldwide public health challenge, and the rate of failure of new therapies for chronic progressive disorders of the nervous system is higher relative to other non-neurological conditions. However, progress is emerging rapidly in advancing the drug development landscape in both rare and common neurodegenerative diseases. In October 2022, the Critical Path Institute (C-Path) and the US Food and Drug Administration (FDA) organized a Neuroscience Annual Workshop convening representatives from the drug development industry, academia, the patient community, government agencies, and regulatory agencies regarding the future development of tools and therapies for neurological disorders. This workshop focused on five chronic progressive diseases: Alzheimer's disease, Parkinson's disease, Huntington's disease, Duchenne muscular dystrophy, and inherited ataxias. This special conference report reviews the key points discussed during the three-day dynamic workshop, including shared learnings, and recommendations that promise to catalyze future advancement of novel therapies and drug development tools.
Collapse
|
13
|
Engelke K, Chaudry O, Gast L, Eldib MAB, Wang L, Laredo JD, Schett G, Nagel AM. Magnetic resonance imaging techniques for the quantitative analysis of skeletal muscle: State of the art. J Orthop Translat 2023; 42:57-72. [PMID: 37654433 PMCID: PMC10465967 DOI: 10.1016/j.jot.2023.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/04/2023] [Accepted: 07/19/2023] [Indexed: 09/02/2023] Open
Abstract
Background Magnetic resonance imaging (MRI) is the dominant 3D imaging modality to quantify muscle properties in skeletal muscle disorders, in inherited and acquired muscle diseases, and in sarcopenia, in cachexia and frailty. Methods This review covers T1 weighted and Dixon sequences, introduces T2 mapping, diffusion tensor imaging (DTI) and non-proton MRI. Technical concepts, strengths, limitations and translational aspects of these techniques are discussed in detail. Examples of clinical applications are outlined. For comparison 31P-and 13C-MR Spectroscopy are also addressed. Results MRI technology provides a rich toolset to assess muscle deterioration. In addition to classical measures such as muscle atrophy using T1 weighted imaging and fat infiltration using Dixon sequences, parameters characterizing inflammation from T2 maps, tissue sodium using non-proton MRI techniques or concentration or fiber architecture using diffusion tensor imaging may be useful for an even earlier diagnosis of the impairment of muscle quality. Conclusion Quantitative MRI provides new options for muscle research and clinical applications. Current limitations that also impair its more widespread use in clinical trials are lack of standardization, ambiguity of image segmentation and analysis approaches, a multitude of outcome parameters without a clear strategy which ones to use and the lack of normal data.
Collapse
Affiliation(s)
- Klaus Engelke
- Department of Medicine III, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
- Institute of Medical Physics (IMP), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Henkestr. 91, 91052, Erlangen, Germany
- Clario Inc, Germany
| | - Oliver Chaudry
- Department of Medicine III, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
| | - Lena Gast
- Institute of Radiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Maximiliansplatz 3, 91054, Erlangen, Germany
| | | | - Ling Wang
- Department of Radiology, Beijing Jishuitan Hospital, Beijing, China
| | - Jean-Denis Laredo
- Service d’Imagerie Médicale, Institut Mutualiste Montsouris & B3OA, UMR CNRS 7052, Inserm U1271 Université de Paris-Cité, Paris, France
| | - Georg Schett
- Department of Medicine III, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
| | - Armin M. Nagel
- Institute of Radiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Maximiliansplatz 3, 91054, Erlangen, Germany
- Division of Medical Physics in Radiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| |
Collapse
|
14
|
Song Y, Xu HY, Xu K, Guo YK, Xie LJ, Peng F, Xu R, Fu H, Yuan WF, Zhou ZQ, Cheng BC, Fu C, Zhou H, Cai XT, Li XS. Clinical utilisation of multimodal quantitative magnetic resonance imaging in investigating muscular damage in Duchenne muscular dystrophy: a study on the association between gluteal muscle groups and motor function. Pediatr Radiol 2023; 53:1648-1658. [PMID: 36892624 PMCID: PMC10359373 DOI: 10.1007/s00247-023-05632-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 03/10/2023]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a neuromuscular disease characterised by progressive muscular weakness and atrophy. Currently, studies on DMD muscle function mostly focus on individual muscles; little is known regarding the effect of gluteal muscle group damage on motor function. OBJECTIVE To explore potential imaging biomarkers of hip and pelvic muscle groups for measuring muscular fat replacement and inflammatory oedema in DMD with multimodal quantitative magnetic resonance imaging (MRI). MATERIALS AND METHODS One hundred fifty-nine DMD boys and 32 healthy male controls were prospectively included. All subjects underwent MRI examination of the hip and pelvic muscles with T1 mapping, T2 mapping and Dixon sequences. Quantitatively measured parameters included longitudinal relaxation time (T1), transverse relaxation time (T2) and fat fraction. Investigations were all based on hip and pelvic muscle groups covering flexors, extensors, adductors and abductors. The North Star Ambulatory Assessment and stair climbing tests were used to measure motor function in DMD. RESULTS T1 of the extensors (r = 0.720, P < 0.01), flexors (r = 0.558, P < 0.01) and abductors (r = 0.697, P < 0.001) were positively correlated with the North Star Ambulatory Assessment score. In contrast, T2 of the adductors (r = -0.711, P < 0.01) and fat fraction of the extensors (r = -0.753, P < 0.01) were negatively correlated with the North Star Ambulatory Assessment score. Among them, T1 of the abductors (b = 0.013, t = 2.052, P = 0.042), T2 of the adductors (b = -0.234, t = -2.554, P = 0.012) and fat fraction of the extensors (b = -0.637, t = - 4.096, P < 0.001) significantly affected the North Star Ambulatory Assessment score. Moreover, T1 of the abductors was highly predictive for identifying motor dysfunction in DMD, with an area under the curve of 0.925. CONCLUSION Magnetic resonance biomarkers of hip and pelvic muscle groups (particularly T1 values of the abductor muscles) have the potential to be used as independent risk factors for motor dysfunction in DMD.
Collapse
Affiliation(s)
- Yu Song
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hua-Yan Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ke Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying-Kun Guo
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin-Jun Xie
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Fei Peng
- Department of Radiology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rong Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hang Fu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei-Feng Yuan
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Zi-Qi Zhou
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Bo-Chao Cheng
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuan Fu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hui Zhou
- Department of Rehabilitation, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiao-Tang Cai
- Department of Rehabilitation, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xue-Sheng Li
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
15
|
Norris AM, Appu AB, Johnson CD, Zhou LY, McKellar DW, Renault MA, Hammers D, Cosgrove BD, Kopinke D. Hedgehog signaling via its ligand DHH acts as cell fate determinant during skeletal muscle regeneration. Nat Commun 2023; 14:3766. [PMID: 37355632 PMCID: PMC10290686 DOI: 10.1038/s41467-023-39506-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 06/16/2023] [Indexed: 06/26/2023] Open
Abstract
Successful muscle regeneration relies on the interplay of multiple cell populations. However, the signals required for this coordinated intercellular crosstalk remain largely unknown. Here, we describe how the Hedgehog (Hh) signaling pathway controls the fate of fibro/adipogenic progenitors (FAPs), the cellular origin of intramuscular fat (IMAT) and fibrotic scar tissue. Using conditional mutagenesis and pharmacological Hh modulators in vivo and in vitro, we identify DHH as the key ligand that acts as a potent adipogenic brake by preventing the adipogenic differentiation of FAPs. Hh signaling also impacts muscle regeneration, albeit indirectly through induction of myogenic factors in FAPs. Our results also indicate that ectopic and sustained Hh activation forces FAPs to adopt a fibrogenic fate resulting in widespread fibrosis. In this work, we reveal crucial post-developmental functions of Hh signaling in balancing tissue regeneration and fatty fibrosis. Moreover, they provide the exciting possibility that mis-regulation of the Hh pathway with age and disease could be a major driver of pathological IMAT formation.
Collapse
Affiliation(s)
- Alessandra M Norris
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Ambili Bai Appu
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Connor D Johnson
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Lylybell Y Zhou
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - David W McKellar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Marie-Ange Renault
- Biology of Cardiovascular Diseases, INSERM, University of Bordeaux, Pessac, France
| | - David Hammers
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Benjamin D Cosgrove
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
16
|
Peng F, Xu H, Song Y, Xu K, Li S, Cai X, Guo Y, Gong L. Longitudinal study of multi-parameter quantitative magnetic resonance imaging in Duchenne muscular dystrophy: hyperresponsiveness of gluteus maximus and detection of subclinical disease progression in functionally stable patients. J Neurol 2023; 270:1439-1451. [PMID: 36385201 DOI: 10.1007/s00415-022-11470-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To describe the disease progression of Duchenne muscular dystrophy (DMD) in the pelvic and thigh muscles over 1-year using multiple-parameter quantitative magnetic resonance imaging (qMRI), and to determine the most responsive muscle and predict subclinical disease progression in functionally stable patients. METHODS Fifty-four DMD patients (mean age 8.9 ± 2.5, range 5-15 years) completed baseline and 1-year follow-up qMRI examinations/biomarkers [3-point Dixon/fat fraction (FF); T1 mapping/T1; T2 mapping/T2]. Meanwhile, clinical assessments [NorthStar ambulatory assessment (NSAA) score] and timed function tests were performed in DMD patients. Twenty-four healthy male controls (range 5-15 years) accomplished baseline qMRI examinations. Group differences were compared using the Wilcoxon test. The standardized response mean (SRM) was taken as the responsiveness to the disease progression index. RESULTS FF, T1, and T2 in all DMD age subgroups changed significantly over 1-year (P < 0.05). Even in functionally stable patients (NSAA score increased, unchanged, or decreased by 1-point) over 1-year, significant increases in FF and T2 and decreases in T1 were observed in gluteus maximus (GMa), gluteus medius, vastus lateralis, and adductor magnus (P < 0.05). Overall, the SRM of FF, T1, and T2 was all the highest in GMa, which were 1.25, - 0.92, and 0.93, respectively. CONCLUSIONS qMRI biomarkers are responsive to disease progression and can also detect subclinical disease progression in functionally stable DMD patients over 1-year. GMa is the most responsive to disease progression of all the muscles analyzed. TRIAL REGISTRATION Chinese Clinical Trial Registry ( http://www.chictr.org.cn/index.aspx ) ChiCTR1800018340, 09/12/2018, prospectively registered.
Collapse
Affiliation(s)
- Fei Peng
- Department of Medical Imaging Center, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi Province, China
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Huayan Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Yu Song
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Ke Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Shuhao Li
- Department of Medical Imaging Center, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi Province, China
| | - Xiaotang Cai
- Department of Pediatrics Neurology, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China.
| | - Yingkun Guo
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China.
| | - Lianggeng Gong
- Department of Medical Imaging Center, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
17
|
van de Velde NM, Koeks Z, Signorelli M, Verwey N, Overzier M, Bakker JA, Sajeev G, Signorovitch J, Ricotti V, Verschuuren J, Brown K, Spitali P, Niks EH. Longitudinal Assessment of Creatine Kinase, Creatine/Creatinine ratio, and Myostatin as Monitoring Biomarkers in Becker Muscular Dystrophy. Neurology 2023; 100:e975-e984. [PMID: 36849458 PMCID: PMC9990441 DOI: 10.1212/wnl.0000000000201609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 10/11/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The slow and variable disease progression of Becker muscular dystrophy (BMD) urges the development of biomarkers to facilitate clinical trials. We explored changes in 3 muscle-enriched biomarkers in serum of patients with BMD over 4-year time and studied associations with disease severity, disease progression, and dystrophin levels in BMD. METHODS We quantitatively measured creatine kinase (CK) using the International Federation of Clinical Chemistry reference method, creatine/creatinineratio (Cr/Crn) using liquid chromatography-tandem mass spectrometry, and myostatin with ELISA in serum and assessed functional performance using the North Star Ambulatory Assessment (NSAA), 10-meter run velocity (TMRv), 6-Minute Walking Test (6MWT), and forced vital capacity in a 4-year prospective natural history study. Dystrophin levels were quantified in the tibialis anterior muscle using capillary Western immunoassay. The correlation between biomarkers, age, functional performance, mean annual change, and prediction of concurrent functional performance was analyzed using linear mixed models. RESULTS Thirty-four patients with 106 visits were included. Eight patients were nonambulant at baseline. Cr/Crn and myostatin were highly patient specific (intraclass correlation coefficient for both = 0.960). Cr/Crn was strongly negatively correlated, whereas myostatin was strongly positively correlated with the NSAA, TMRv, and 6MWT (Cr/Crn rho = -0.869 to -0.801 and myostatin rho = 0.792 to 0.842, all p < 0.001). CK showed a negative association with age (p = 0.0002) but was not associated with patients' performance. Cr/Crn and myostatin correlated moderately with the average annual change of the 6MWT (rho = -0.532 and 0.555, p = 0.02). Dystrophin levels did not correlate with the selected biomarkers nor with performance. Cr/Crn, myostatin, and age could explain up to 75% of the variance of concurrent functional performance of the NSAA, TMRv, and 6MWT. DISCUSSION Both Cr/Crn and myostatin could potentially serve as monitoring biomarkers in BMD, as higher Cr/Crn and lower myostatin were associated with lower motor performance and predictive of concurrent functional performance when combined with age. Future studies are needed to more precisely determine the context of use of these biomarkers.
Collapse
Affiliation(s)
- Nienke M van de Velde
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Zaïda Koeks
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Mirko Signorelli
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Nisha Verwey
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Maurice Overzier
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Jaap A Bakker
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Gautam Sajeev
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - James Signorovitch
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Valeria Ricotti
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Jan Verschuuren
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Kristy Brown
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Pietro Spitali
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Erik H Niks
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom.
| |
Collapse
|
18
|
Monforte M, Attarian S, Vissing J, Diaz-Manera J, Tasca G. 265th ENMC International Workshop: Muscle imaging in Facioscapulohumeral Muscular Dystrophy (FSHD): relevance for clinical trials. 22-24 April 2022, Hoofddorp, The Netherlands. Neuromuscul Disord 2023; 33:65-75. [PMID: 36369218 DOI: 10.1016/j.nmd.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/10/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Mauro Monforte
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Shahram Attarian
- Reference Center for Neuromuscular Disorders and ALS, CHU La Timone Aix-Marseille Hospital University Marseille, France
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jordi Diaz-Manera
- John Walton Muscular Dystrophy Research Center, University of Newcastle, Newcastle upon Tyne, United Kingdom
| | - Giorgio Tasca
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, Rome 00168, Italy.
| |
Collapse
|
19
|
Barnard AM, Hammers DW, Triplett WT, Kim S, Forbes SC, Willcocks RJ, Daniels MJ, Senesac CR, Lott DJ, Arpan I, Rooney WD, Wang RT, Nelson SF, Sweeney HL, Vandenborne K, Walter GA. Evaluating Genetic Modifiers of Duchenne Muscular Dystrophy Disease Progression Using Modeling and MRI. Neurology 2022; 99:e2406-e2416. [PMID: 36240102 PMCID: PMC9687406 DOI: 10.1212/wnl.0000000000201163] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/11/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Duchenne muscular dystrophy (DMD) is a progressive muscle degenerative disorder with a well-characterized disease phenotype but considerable interindividual heterogeneity that is not well understood. The aim of this study was to evaluate the effects of dystrophin variations and genetic modifiers of DMD on rate and age of muscle replacement by fat. METHODS One hundred seventy-five corticosteroid treated participants from the ImagingDMD natural history study underwent repeated magnetic resonance spectroscopy (MRS) of the vastus lateralis (VL) and soleus (SOL) to determine muscle fat fraction (FF). MRS was performed annually in most instances; however, some individuals had additional visits at 3 or 6 monthss intervals. FF changes over time were modeled using nonlinear mixed effects to estimate disease trajectories based on the age that the VL or SOL reached half-maximum change in FF (mu) and the time required for FF change (sigma). Computed mu and sigma values were evaluated for dystrophin variations that have demonstrated the ability to lead to a mild phenotype as well as compared between different genetic polymorphism groups. RESULTS Participants with dystrophin gene deletions amenable to exon 8 skipping (n = 4) had minimal increases in SOL FF and had an increase in VL mu value by 4.4 years compared with a reference cohort (p = 0.039). Participants with nonsense variations within exons that may produce milder phenotypes (n = 11) also had minimal increases in SOL and VL FFs. No differences in estimated FF trajectories were seen for individuals amenable to exon 44 skipping (n = 10). Modeling of the SPP1, LTBP4, and thrombospondin-1 (THBS1) genetic modifiers did not result in significant differences in muscle FF trajectories between genotype groups (p > 0.05); however, trends were noted for the polymorphisms associated with long-range regulation of LTBP4 and THBS1 that deserve further follow-up. DISCUSSION The results of this study link the historically mild phenotypes seen in individuals amenable to exon 8 skipping and with certain nonsense variations with alterations in trajectories of lower extremity muscle replacement by fat.
Collapse
Affiliation(s)
- Alison M Barnard
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - David W Hammers
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - William T Triplett
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Sarah Kim
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Sean C Forbes
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Rebecca J Willcocks
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Michael J Daniels
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Claudia R Senesac
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Donovan J Lott
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Ishu Arpan
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - William D Rooney
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Richard T Wang
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Stanley F Nelson
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - H Lee Sweeney
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Krista Vandenborne
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Glenn A Walter
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville.
| |
Collapse
|
20
|
Sherlock SP, Palmer J, Wagner KR, Abdel-Hamid HZ, Tian C, Mah JK, Muntoni F, Guglieri M, Butterfield RJ, Charnas L, Marraffino S. Dual-energy X-ray absorptiometry measures of lean body mass as a biomarker for progression in boys with Duchenne muscular dystrophy. Sci Rep 2022; 12:18762. [PMID: 36335191 PMCID: PMC9637094 DOI: 10.1038/s41598-022-23072-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
We evaluated whether whole-body dual-energy X-ray absorptiometry (DXA) measures of lean body mass can be used as biomarkers for disease progression and treatment effects in patients with Duchenne muscular dystrophy. This post hoc analysis utilized data from a randomized, 2-period study of domagrozumab versus placebo in 120 ambulatory boys with DMD. DXA measures of lean body mass were obtained from the whole body (excluding head), arms, legs and appendicular skeleton at baseline and every 16 weeks. Treatment effects on DXA measures for domagrozumab versus placebo were assessed at Week 49. At Week 49, domagrozumab statistically significantly increased lean body mass versus placebo in the appendicular skeleton (p = 0.050) and arms (p < 0.001). The relationship between lean body mass at Week 49 and functional endpoints at Week 97 was evaluated. Changes in lean body mass at Week 49 in all regions except arms were significantly correlated with percent change from baseline in 4-stair climb (4SC) at Week 97. DXA-derived percent lean mass at Week 49 also correlated with 4SC and North Star Ambulatory Assessment at Week 97. These data indicate that whole-body DXA measures can be used as biomarkers for treatment effects and disease progression in patients with DMD, and warrant further investigation.Trial registration: ClinicalTrials.gov, NCT02310763; registered 8 December 2014.
Collapse
Affiliation(s)
| | | | - Kathryn R Wagner
- Kennedy Krieger Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hoda Z Abdel-Hamid
- Division of Child Neurology, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cuixia Tian
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Jean K Mah
- Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Michela Guglieri
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle, UK
| | | | | | | |
Collapse
|
21
|
Hammers DW. NOX4 inhibition promotes the remodeling of dystrophic muscle. JCI Insight 2022; 7:158316. [PMID: 36278481 PMCID: PMC9714779 DOI: 10.1172/jci.insight.158316] [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: 01/10/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
The muscular dystrophies (MDs) are genetic muscle diseases that result in progressive muscle degeneration followed by the fibrotic replacement of affected muscles as regenerative processes fail. Therapeutics that specifically address the fibrosis and failed regeneration associated with MDs represent a major unmet clinical need for MD patients, particularly those with advanced-stage disease progression. The current study investigated targeting NAD(P)H oxidase 4 (NOX4) as a potential strategy to reduce fibrosis and promote regeneration in disease-burdened muscle that models Duchenne muscular dystrophy (DMD). NOX4 was elevated in the muscles of dystrophic mice and DMD patients, localizing primarily to interstitial cells located between muscle fibers. Genetic and pharmacological targeting of NOX4 significantly reduced fibrosis in dystrophic respiratory and limb muscles. Mechanistically, NOX4 targeting decreased the number of fibrosis-depositing cells (myofibroblasts) and restored the number of muscle-specific stem cells (satellite cells) localized to their physiological niche, thereby rejuvenating muscle regeneration. Furthermore, acute inhibition of NOX4 was sufficient to induce apoptotic clearing of myofibroblasts within dystrophic muscle. These data indicate that targeting NOX4 is an effective strategy to promote the beneficial remodeling of disease-burdened muscle representative of DMD and, potentially, other MDs and muscle pathologies.
Collapse
Affiliation(s)
- David W. Hammers
- Department of Pharmacology & Therapeutics and
- Myology Institute, University of Florida College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
22
|
Veeger TTJ, van de Velde NM, Keene KR, Niks EH, Hooijmans MT, Webb AG, de Groot JH, Kan HE. Baseline fat fraction is a strong predictor of disease progression in Becker muscular dystrophy. NMR IN BIOMEDICINE 2022; 35:e4691. [PMID: 35032073 PMCID: PMC9286612 DOI: 10.1002/nbm.4691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/24/2021] [Accepted: 01/10/2022] [Indexed: 06/14/2023]
Abstract
In Becker muscular dystrophy (BMD), muscle weakness progresses relatively slowly, with a highly variable rate among patients. This complicates clinical trials, as clinically relevant changes are difficult to capture within the typical duration of a trial. Therefore, predictors for disease progression are needed. We assessed if temporal increase of fat fraction (FF) in BMD follows a sigmoidal trajectory and whether fat fraction at baseline (FFbase) could therefore predict FF increase after 2 years (ΔFF). Thereafter, for two different MR-based parameters, we tested the additional predictive value to FFbase. We used 3-T Dixon data from the upper and lower leg, and multiecho spin-echo MRI and 7-T 31 P MRS datasets from the lower leg, acquired in 24 BMD patients (age: 41.4 [SD 12.8] years). We assessed the pattern of increase in FF using mixed-effects modelling. Subsequently, we tested if indicators of muscle damage like standard deviation in water T2 (stdT2 ) and the phosphodiester (PDE) over ATP ratio at baseline had additional value to FFbase for predicting ∆FF. The association between FFbase and ΔFF was described by the derivative of a sigmoid function and resulted in a peak ΔFF around 0.45 FFbase (fourth-order polynomial term: t = 3.7, p < .001). StdT2 and PDE/ATP were not significantly associated with ∆FF if FFbase was included in the model. The relationship between FFbase and ∆FF suggests a sigmoidal trajectory of the increase in FF over time in BMD, similar to that described for Duchenne muscular dystrophy. Our results can be used to identify muscles (or patients) that are in the fast progressing stage of the disease, thereby facilitating the conduct of clinical trials.
Collapse
Affiliation(s)
- Thom T. J. Veeger
- C. J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical Center (LUMC)LeidenThe Netherlands
| | - Nienke M. van de Velde
- Department of Neurology, Leiden University Medical Center (LUMC)LeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| | - Kevin R. Keene
- Department of Neurology, Leiden University Medical Center (LUMC)LeidenThe Netherlands
| | - Erik H. Niks
- Department of Neurology, Leiden University Medical Center (LUMC)LeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| | - Melissa T. Hooijmans
- Department of Radiology & Nuclear MedicineAmsterdam University Medical CentersAmsterdamThe Netherlands
| | - Andrew G. Webb
- C. J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical Center (LUMC)LeidenThe Netherlands
| | - Jurriaan H. de Groot
- Department of Rehabilitation Medicine, Leiden University Medical Center (LUMC)LeidenThe Netherlands
| | - Hermien E. Kan
- C. J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical Center (LUMC)LeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| |
Collapse
|
23
|
Gallone A, Mazzi F, Bonanno S, Zanin R, Moscatelli M, Aquino D, Maggi L. Muscle quantitative MRI in adult SMA patients on nusinersen treatment: a longitudinal study. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2022; 41:76-83. [PMID: 35832502 PMCID: PMC9237750 DOI: 10.36185/2532-1900-074] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
Abstract
The recent approval of disease-modifying therapies for spinal muscular atrophy (SMA) raised the need of alternative outcome measures to evaluate treatment efficacy. In this study, we investigated the potential of muscle quantitative MRI (qMRI) as a biomarker of disease progression in adult SMA3 patients during nusinersen treatment. Six adult SMA3 patients (age ranging from 19 to 65 years) underwent 2-point Dixon muscle qMRI at beginning of nusinersen treatment (T0) and after 14 months (T14) to evaluate the muscle fat fraction (FF) at thigh and leg levels; patients were clinically assessed at T0 and T14 with the Hammersmith Functional Rating Scale Expanded (HFMSE), the Revised Upper Limb Module (RULM) and the 6-minute walk test (6MWT). At T0, vastus lateralis muscle displayed the highest mean FF (67.5%), while tibialis anterior was the most preserved one (mean FF = 35.2%). At T0, a slightly significant correlation of FF with HFMSE (p = 0.042) and disease duration (p = 0.042) at thigh level and only with HFMSE (p = 0.042) at leg level was found. At T14, no significant change of mean FF values at thigh and leg muscles was found compared to T0. Conversely, a statistically significant (p = 0.042) improvement of HFMSE was reported at T14. We observed no significant change of FF in thigh and leg muscles after 14 months of nusinersen therapy despite a significant clinical improvement of HFMSE. Further studies with longer follow-up and larger cohorts are needed to better investigate the role of qMRI as marker of disease progression in SMA patients.
Collapse
Affiliation(s)
- Annamaria Gallone
- Neuroimmunology and Neuromuscular Diseases Unit, Foundation IRCCS “Carlo Besta” Neurological Institute, Milan, Italy
| | - Federica Mazzi
- Neuroradiology Unit, Foundation IRCCS “Carlo Besta” Neurological Institute, Milan, Italy
| | - Silvia Bonanno
- Neuroimmunology and Neuromuscular Diseases Unit, Foundation IRCCS “Carlo Besta” Neurological Institute, Milan, Italy
| | - Riccardo Zanin
- Developmental Neurology, Foundation IRCCS “Carlo Besta” Neurological Institute, Milan, Italy
| | - Marco Moscatelli
- Neuroradiology Unit, Foundation IRCCS “Carlo Besta” Neurological Institute, Milan, Italy, Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Domenico Aquino
- Neuroradiology Unit, Foundation IRCCS “Carlo Besta” Neurological Institute, Milan, Italy
| | - Lorenzo Maggi
- Neuroimmunology and Neuromuscular Diseases Unit, Foundation IRCCS “Carlo Besta” Neurological Institute, Milan, Italy,Correspondence Lorenzo Maggi Neuroimmunology and Neuromuscular Diseases; Foundation IRCCS “Carlo Besta” Neurological Institute, via Celoria 11, 20133 Milan, Italy. E-mail:
| |
Collapse
|
24
|
Johnson CD, Zhou LY, Kopinke D. A Guide to Examining Intramuscular Fat Formation and its Cellular Origin in Skeletal Muscle. JOURNAL OF VISUALIZED EXPERIMENTS : JOVE 2022:10.3791/63996. [PMID: 35695517 PMCID: PMC9741761 DOI: 10.3791/63996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fibro-adipogenic progenitors (FAPs) are mesenchymal stromal cells that play a crucial role during skeletal muscle homeostasis and regeneration. FAPs build and maintain the extracellular matrix that acts as a molecular myofiber scaffold. In addition, FAPs are indispensable for myofiber regeneration as they secrete a multitude of beneficial factors sensed by the muscle stem cells (MuSCs). In diseased states, however, FAPs are the cellular origin of intramuscular fat and fibrotic scar tissue. This fatty fibrosis is a hallmark of sarcopenia and neuromuscular diseases, such as Duchenne Muscular Dystrophy. One significant barrier in determining why and how FAPs differentiate into intramuscular fat is effective preservation and subsequent visualization of adipocytes, especially in frozen tissue sections. Conventional methods of skeletal muscle tissue processing, such as snap-freezing, do not properly preserve the morphology of individual adipocytes, thereby preventing accurate visualization and quantification. To overcome this hurdle, a rigorous protocol was developed that preserves adipocyte morphology in skeletal muscle sections allowing visualization, imaging, and quantification of intramuscular fat. The protocol also outlines how to process a portion of muscle tissue for RT-qPCR, enabling users to confirm observed changes in fat formation by viewing differences in the expression of adipogenic genes. Additionally, it can be adapted to visualize adipocytes by whole-mount immunofluorescence of muscle samples. Finally, this protocol outlines how to perform genetic lineage tracing of Pdgfrα-expressing FAPs to study the adipogenic conversion of FAPs. This protocol consistently yields high-resolution and morphologically accurate immunofluorescent images of adipocytes, along with confirmation by RT-qPCR, allowing for robust, rigorous, and reproducible visualization and quantification of intramuscular fat. Together, the analysis pipeline described here is the first step to improving our understanding of how FAPs differentiate into intramuscular fat, and provides a framework to validate novel interventions to prevent fat formation.
Collapse
Affiliation(s)
- Connor D. Johnson
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine
| | - Lylybell Y. Zhou
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine
| | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine,Myology Institute, University of Florida College of Medicine
| |
Collapse
|
25
|
Widholm P, Ahlgren A, Karlsson M, Romu T, Tawil R, Wagner KR, Statland JM, Wang LH, Shieh PB, van Engelen BGM, Cadavid D, Ronco L, Odueyungbo AO, Jiang JG, Mellion ML, Dahlqvist Leinhard O. Quantitative muscle analysis in facioscapulohumeral muscular dystrophy using whole-body fat-referenced MRI: Protocol development, multicenter feasibility, and repeatability. Muscle Nerve 2022; 66:183-192. [PMID: 35585766 DOI: 10.1002/mus.27638] [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] [Received: 09/23/2021] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 11/07/2022]
Abstract
INTRODUCTION/AIMS Functional performance tests are the gold standard to assess disease progression and treatment effects in neuromuscular disorders. These tests can be confounded by motivation, pain, fatigue, and learning effects, increasing variability and decreasing sensitivity to disease progression, limiting efficacy assessment in clinical trials with small sample sizes. We aimed to develop and validate a quantitative and objective method to measure skeletal muscle volume and fat content based on whole-body fat-referenced magnetic resonance imaging (MRI) for use in multisite clinical trials. METHODS Subjects aged 18 to 65 years, genetically confirmed facioscapulohumeral muscular dystrophy 1 (FSHD1), clinical severity 2 to 4 (Ricci's scale, range 0-5), were enrolled at six sites and imaged twice 4-12 weeks apart with T1-weighted two-point Dixon MRI covering the torso and upper and lower extremities. Thirty-six muscles were volumetrically segmented using semi-automatic multi-atlas-based segmentation. Muscle fat fraction (MFF), muscle fat infiltration (MFI), and lean muscle volume (LMV) were quantified for each muscle using fat-referenced quantification. RESULTS Seventeen patients (mean age ± SD, 49.4 years ±13.02; 12 men) were enrolled. Within-patient SD ranged from 1.00% to 3.51% for MFF and 0.40% to 1.48% for MFI in individual muscles. For LMV, coefficients of variation ranged from 2.7% to 11.7%. For the composite score average of all muscles, observed SDs were 0.70% and 0.32% for MFF and MFI, respectively; composite LMV coefficient of variation was 2.0%. DISCUSSION We developed and validated a method for measuring skeletal muscle volume and fat content for use in multisite clinical trials of neuromuscular disorders.
Collapse
Affiliation(s)
- Per Widholm
- AMRA Medical AB, Linköping, Sweden.,Department of Radiology, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.,Centre for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | | | | | | | - Rabi Tawil
- University of Rochester Medical Center, Rochester, New York, USA
| | - Kathryn R Wagner
- Kennedy Krieger Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | - Leo H Wang
- University of Washington, Seattle, Washington, USA
| | - Perry B Shieh
- University of California, Los Angeles, California, USA
| | - Baziel G M van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | | | - John G Jiang
- Fulcrum Therapeutics, Cambridge, Massachusetts, USA
| | | | - Olof Dahlqvist Leinhard
- AMRA Medical AB, Linköping, Sweden.,Centre for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden.,Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW This review highlights the key studies investigating various types of biomarkers in Duchenne muscular dystrophy (DMD). RECENT FINDINGS Several proteomic and metabolomic studies have been undertaken in both human DMD patients and animal models of DMD that have identified potential biomarkers in DMD. Although there have been a number of proteomic and metabolomic studies that have identified various potential biomarkers in DMD, more definitive studies still need to be undertaken in DMD patients to firmly correlate these biomarkers with diagnosis, disease progression, and monitoring the effects of novel treatment strategies being developed.
Collapse
Affiliation(s)
- Theo Lee-Gannon
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Xuan Jiang
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tara C Tassin
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Pradeep P A Mammen
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
- Heart Failure, Ventricular Assist Device & Heart Transplant Program, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
- Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
27
|
Suslov V, Suslova G, Lytaev S. MRI Assessment of Motor Capabilities in Patients with Duchenne Muscular Dystrophy According to the Motor Function Measure Scale. Tomography 2022; 8:948-960. [PMID: 35448710 PMCID: PMC9025497 DOI: 10.3390/tomography8020076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022] Open
Abstract
The research was aimed on the study of motor capabilities on the Motor Function Measure (MFM) scale in ambulant and non-ambulant patients with Duchenne muscular dystrophy, and to conduct a correlation analysis between the results of the MFM scale and Magnetic Resonance Imaging (MRI) data. A total of 46 boys who had genetically confirmed Duchenne muscular dystrophy (age from 2.1 to 16.7 years) and were in clinical rehabilitation were investigated. An assessment was performed according to the Motor Function Measure scale (subsections D1, D2, D3, and the total score), an MRI obtaining T1-VI of the muscles of the pelvic girdle was conducted, and the thighs and lower legs were further assessed in terms of the severity of fibrous-fat degeneration according to the Mercuri scale. In ambulant patients, the ability to stand up and move (D1) was 74.4%, axial and proximal motor functions (D2)—97.6%, distal motor functions (D3)—96.2%, and total score was 87.9%. In non-ambulant patients, the ability to stand up and move (D1) was 1.7%, axial and proximal motor functions (D2)—47%, distal motor functions (D3)—67.5%, and the total score—33.1%. A high inverse correlation (r = −0.7, p < 0.05) of the MRI data of the pelvic girdle and thighs with tasks D1, as well as a noticeable inverse correlation with tasks D2 (r = −0.6, p < 0.05) of the scale MFM, were revealed in the ambulant group of patients. In the non-ambulant group of patients, the MRI data of the lower legs muscles were characterized by a high inverse correlation (r = −0.7, p < 0.05) with tasks D3 and a noticeable inverse correlation (r = −0.6, p < 0.05) with tasks D1 of the MFM scale. Conclusion: The Motor Function Measure scale allows effective assessment of the motor capabilities of patients with Duchenne muscular dystrophy at different stages of the disease, which is confirmed by visualization of fibro-fatty muscle replacement.
Collapse
Affiliation(s)
- Vasily Suslov
- Department of Rehabilitation, Saint Petersburg State Pediatric Medical University, 194100 Saint Petersburg, Russia;
- Correspondence: ; Tel.: +7-911-2297049
| | - Galina Suslova
- Department of Rehabilitation, Saint Petersburg State Pediatric Medical University, 194100 Saint Petersburg, Russia;
| | - Sergey Lytaev
- Department of Normal Physiology, Saint Petersburg State Pediatric Medical University, 194100 Saint Petersburg, Russia;
| |
Collapse
|
28
|
Cristiano L, Brogna C, Tasca G, Verdolotti T, Pane M, Mercuri E. Muscle-MRI and Functional Levels for the Evaluation of Upper Limbs in Duchenne Muscular Dystrophy: A Critical Review of the Literature. Medicina (B Aires) 2022; 58:medicina58030440. [PMID: 35334617 PMCID: PMC8954550 DOI: 10.3390/medicina58030440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/02/2022] Open
Abstract
Many qualitative and quantitative Magnetic Resonance Imaging (MRI) techniques have been applied to evaluate muscle fat degeneration in Duchenne muscular dystrophy (DMD) subjects, but only few studies have focused on the upper limbs. We reviewed the literature in order to evaluate the association between muscle MRI findings and motor function levels in the upper limbs of DMD patients. Ten studies with upper limb muscle MRI data were available. Four explored all upper limb segments, while six explored only the forearm. Functional assessments were performed in nine of the ten studies. All of the studies showed a significant correlation between muscle MRI changes and motor function levels in both ambulant and non-ambulant DMD patients.
Collapse
Affiliation(s)
- Lara Cristiano
- Pediatric Neurology Unit, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (L.C.); (M.P.); (E.M.)
| | - Claudia Brogna
- Pediatric Neurology Unit, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (L.C.); (M.P.); (E.M.)
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy
- Correspondence: ; Tel.: +39-06-30155340; Fax: +39-06-30154363
| | - Giorgio Tasca
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy;
| | - Tommaso Verdolotti
- Institute of Radiology, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy;
| | - Marika Pane
- Pediatric Neurology Unit, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (L.C.); (M.P.); (E.M.)
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy
| | - Eugenio Mercuri
- Pediatric Neurology Unit, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (L.C.); (M.P.); (E.M.)
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
29
|
Mensch A, Nägel S, Zierz S, Kraya T, Stoevesandt D. Bildgebung der Muskulatur bei Neuromuskulären Erkrankungen
– von der Initialdiagnostik bis zur Verlaufsbeurteilung. KLIN NEUROPHYSIOL 2022. [DOI: 10.1055/a-1738-5356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
ZusammenfassungDie bildgebende Diagnostik hat sich zu einem integralen Element der Betreuung von
PatientInnen mit neuromuskulären Erkrankungen entwickelt. Als
wesentliches Diagnostikum ist hierbei die Magnetresonanztomografie als breit
verfügbares und vergleichsweise standardisiertes Untersuchungsverfahren
etabliert, wobei die Sonografie der Muskulatur bei hinreichend erfahrenem
Untersucher ebenfalls geeignet ist, wertvolle diagnostische Informationen zu
liefern. Das CT hingegen spielt eine untergeordnete Rolle und sollte nur bei
Kontraindikationen für eine MRT in Erwägung gezogen werden.
Zunächst wurde die Bildgebung bei Muskelerkrankungen primär in
der Initialdiagnostik unter vielfältigen Fragestellungen eingesetzt. Das
Aufkommen innovativer Therapiekonzepte bei verschiedenen neuromuskulären
Erkrankungen machen neben einer möglichst frühzeitigen
Diagnosestellung insbesondere auch eine multimodale Verlaufsbeurteilung zur
Evaluation des Therapieansprechens notwendig. Auch hier wird die Bildgebung der
Muskulatur als objektiver Parameter des Therapieerfolges intensiv diskutiert und
in Forschung wie Praxis zunehmend verwendet.
Collapse
Affiliation(s)
- Alexander Mensch
- Universitätsklinik und Poliklinik für Neurologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
| | - Steffen Nägel
- Universitätsklinik und Poliklinik für Neurologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
| | - Stephan Zierz
- Universitätsklinik und Poliklinik für Neurologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
| | - Torsten Kraya
- Universitätsklinik und Poliklinik für Neurologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
- Klinik für Neurologie, Klinikum St. Georg,
Leipzig
| | - Dietrich Stoevesandt
- Universitätsklinik und Poliklinik für Radiologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
| |
Collapse
|
30
|
Rebecca JW, Alison MB, Ryan JW, Claudia RS, Donovan JL, Ann TH, Kirsten LZ, Sean CF, William DR, Dah-Jyuu W, Erika LF, Gihan IT, Michael JD, William TT, Glenn AW, Krista V. Development of Contractures in DMD in Relation to MRI-Determined Muscle Quality and Ambulatory Function. J Neuromuscul Dis 2022; 9:289-302. [PMID: 35124659 DOI: 10.3233/jnd-210731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Joint contractures are common in boys and men with Duchenne muscular dystrophy (DMD), and management of contractures is an important part of care. The optimal methods to prevent and treat contractures are controversial, and the natural history of contracture development is understudied in glucocorticoid treated individuals at joints beyond the ankle. OBJECTIVE To describe the development of contractures over time in a large cohort of individuals with DMD in relation to ambulatory ability, functional performance, and muscle quality measured using magnetic resonance imaging (MRI) and spectroscopy (MRS). METHODS In this longitudinal study, range of motion (ROM) was measured annually at the hip, knee, and ankle, and at the elbow, forearm, and wrist at a subset of visits. Ambulatory function (10 meter walk/run and 6 minute walk test) and MR-determined muscle quality (transverse relaxation time (T2) and fat fraction) were measured at each visit. RESULTS In 178 boys with DMD, contracture prevalence and severity increased with age. Among ambulatory participants, more severe contractures (defined as greater loss of ROM) were significantly associated with worse ambulatory function, and across all participants, more severe contractures significantly associated with higher MRI T2 or MRS FF (ρ: 0.40-0.61 in the lower extremity; 0.20-0.47 in the upper extremity). Agonist/antagonist differences in MRI T2 were not strong predictors of ROM. CONCLUSIONS Contracture severity increases with disease progression (increasing age and muscle involvement and decreasing functional ability), but is only moderately predicted by muscle fatty infiltration and MRI T2, suggesting that other changes in the muscle, tendon, or joint contribute meaningfully to contracture formation in DMD.
Collapse
Affiliation(s)
| | | | - J Wortman Ryan
- Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | - T Harrington Ann
- Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Arcadia University, Glennside, PA, USA
| | - L Zilke Kirsten
- Shriners Hospitals for Children -Portland, OR, USA.,Oregon Health and Science University, Portland, OR, USA
| | | | | | - Wang Dah-Jyuu
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Nair KS, Lott DJ, Forbes SC, Barnard AM, Willcocks RJ, Senesac CR, Daniels MJ, Harrington AT, Tennekoon GI, Zilke K, Finanger EL, Finkel RS, Rooney WD, Walter GA, Vandenborne K. Step Activity Monitoring in Boys with Duchenne Muscular Dystrophy and its Correlation with Magnetic Resonance Measures and Functional Performance. J Neuromuscul Dis 2022; 9:423-436. [PMID: 35466946 PMCID: PMC9257666 DOI: 10.3233/jnd-210746] [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] [Indexed: 01/03/2023]
Abstract
BACKGROUND Muscles of boys with Duchenne muscular dystrophy (DMD) are progressively replaced by fatty fibrous tissues, and weakness leads to loss of ambulation (LoA). Step activity (SA) monitoring is a quantitative measure of real-world ambulatory function. The relationship between quality of muscle health and SA is unknown in DMD. OBJECTIVE To determine SA in steroid treated boys with DMD across various age groups, and to evaluate the association of SA with quality of muscle health and ambulatory function. METHODS Quality of muscle health was measured by magnetic resonance (MR) imaging transverse magnetization relaxation time constant (MRI-T2) and MR spectroscopy fat fraction (MRS-FF). SA was assessed via accelerometry, and functional abilities were assessed through clinical walking tests. Correlations between SA, MR, and functional measures were determined. A threshold value of SA was determined to predict the future LoA. RESULTS The greatest reduction in SA was observed in the 9- < 11years age group. SA correlated with all functional and MR measures.10m walk/run test had the highest correlation with SA. An increase in muscle MRI-T2 and MRS-FF was associated with a decline in SA. Two years prior to LoA, SA in boys with DMD was 32% lower than age matched boys with DMD who maintained ambulation for more than two-year period. SA monitoring can predict subsequent LoA in Duchenne, as a daily step count of 3200 at baseline was associated with LoA over the next two-years. CONCLUSION SA monitoring is a feasible and accessible tool to measure functional capacity in the real-world environment.
Collapse
Affiliation(s)
- Kavya S. Nair
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Donovan J. Lott
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Sean C. Forbes
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Alison M. Barnard
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Rebecca J. Willcocks
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Claudia R. Senesac
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Michael J. Daniels
- Department of Statistics, University of Florida, Gainesville, Florida, USA
| | - Ann T. Harrington
- Center for Rehabilitation, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Gihan I. Tennekoon
- Department of Neurology and Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kirsten Zilke
- Department of Pediatrics and Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | - Erika L. Finanger
- Department of Pediatrics and Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | - Richard S. Finkel
- Center for Experimental Neurotherapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - William D. Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Glenn A. Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
32
|
Karlsson A, Peolsson A, Romu T, Dahlqvist Leinhard O, Spetz Holm AC, Thorell S, West J, Borga M. The effect on precision and T1 bias comparing two flip angles when estimating muscle fat infiltration using fat-referenced chemical shift-encoded imaging. NMR IN BIOMEDICINE 2021; 34:e4581. [PMID: 34232549 DOI: 10.1002/nbm.4581] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 05/26/2021] [Accepted: 06/15/2021] [Indexed: 06/13/2023]
Abstract
Investigation of the effect on accuracy and precision of different parameter settings is important for quantitative MRI. The purpose of this study was to investigate T1 bias and precision for muscle fat infiltration (MFI) measurements using fat-referenced chemical shift MFI measurements at flip angles of 5° and 10°. The fat-referenced measurements were compared with fat fractions, which is a more commonly used measure of MFI. This retrospective study was performed on data from a clinical intervention study including 40 postmenopausal women. Test and retest images were acquired with a 3-T scanner using four-point 3D spoiled gradient multiecho acquisition. Postprocessing included T2* correction and fat-referenced calibration, where the fat signal was calibrated using adipose tissue as reference. The mean MFI was calculated in six different muscle regions using both the fat-referenced fat signal and the fat fraction, defined as the fat signal divided by the sum of the fat and water signals. Both methods used the same fat and water images as input. The variance of the difference between mean MFI from test and retest was used as the measure of precision. The signal-to-noise ratio (SNR) characteristics were analyzed by measuring the full width at half maximum (FWHM) of the fat signal distribution. There was no difference in the mean MFI at different flip angles for the fat-referenced technique (p = 0.66), while the measured fat fractions were 3.3 percentage points larger for 10° compared with 5° (p < 0.001). No significant difference in the precision was found in any of the muscles analyzed. However, the FWHM of the fat signal distribution was significantly (p = 0.01) lower at 10°. This strenghtens the hypothesis that fat-referenced MFI is insensitive to flip angle-induced T1 bias in CSE-MRI, enabling usage of a higher and more SNR-effective flip angle. The lower FWHM in fat-referenced MFI at 10° indicates that high flip angle acquisition is advantageous even although no significant differences in precision were observed comparing 5° and 10°.
Collapse
Affiliation(s)
- Anette Karlsson
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
- Center for Medical Image Sciences and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Anneli Peolsson
- Center for Medical Image Sciences and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Health, Medicine and Caring Sciences, unit of Physiotherapy, Linköping University, Linköping, Sweden
| | | | - Olof Dahlqvist Leinhard
- Center for Medical Image Sciences and Visualization (CMIV), Linköping University, Linköping, Sweden
- AMRA Medical AB, Linköping, Sweden
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Anna-Clara Spetz Holm
- Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Sofia Thorell
- Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Janne West
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
- Center for Medical Image Sciences and Visualization (CMIV), Linköping University, Linköping, Sweden
- AMRA Medical AB, Linköping, Sweden
| | - Magnus Borga
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
- Center for Medical Image Sciences and Visualization (CMIV), Linköping University, Linköping, Sweden
- AMRA Medical AB, Linköping, Sweden
| |
Collapse
|
33
|
Duong T, Canbek J, Fernandez-Fernandez A, Henricson E, Birkmeier M, Siener C, Rocha CT, McDonald C, Gordish-Dressman H. Knee Strength and Ankle Range of Motion Impacts on Timed Function Tests in Duchenne Muscular Dystrophy: In the Era of Glucocorticoids. J Neuromuscul Dis 2021; 9:147-159. [PMID: 34719507 DOI: 10.3233/jnd-210724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Duchenne Muscular Dystrophy (DMD) is a neuromuscular disorder that presents in childhood and is characterized by slowly progressive proximal weakness and lower extremity contractures that limit ambulatory ability [1, 2]. Contractures develop in the ankles, knees, and hips due to muscle imbalances, fibrotic changes, loss of strength, and static positioning [2, 5]. Currently, standards of care guidelines emphasize the importance of maintaining good musculoskeletal alignment through stretching, bracing, and glucocorticoid (GC) therapy to preserve strength and function. METHODS This is a retrospective analysis of prospectively collected data through the CINRG Duchenne Natural history study (DNHS). The objectives of this analysis are to understand the progression of ankle contractures for individuals with DMD and to investigate the relationship between progressive lower limb contractures, knee strength, and Timed Function Tests.A collection of TFTs including supine to stand (STS), 10 meter walk test (10MWT), and timed stair climbing (4SC) have been used to monitor disease progression and are predictive of loss of ambulation in these patients [4]. Multiple factors contribute to loss of ambulation, including progressive loss of strength and contracture development that leads to changing biomechanical demands for ambulation. A better understanding of the changes in strength and range of motion (ROM) that contribute to loss of function is important in a more individualized rehabilitation management plan. In this longitudinal study, we measured strength using quantitative muscle testing (QMT) with the CINRG Quantitative Measurement System (CQMS)), ROM was measuresed with a goniometer and TFTs were measured using a standard stopwatch and methodology. RESULTS We enrolled 440 participants; mean baseline age was 8.9 (2.1, 28.0) years with 1321 observations used for analysis. GC use was stratified based on duration on drug with 18.7%at < 6 months or naïve; 4.3%<1 year; 58.0%1 < 10 years; and 19.3%between 10-25 years of GC use. Ankle ROM was better for those on GC compared to GC naive but did not significantly influence long-term progression rates. QMT, ROM, age and GCs contribute to speed of TFTs. Knee extension (KE) strength and Dorsiflexion (DF) ROM are significant predictors of speed for all TFTs (p < 0.001). Of the variables used in this analysis, KE strength is the primary predictor of walking speed, estimating that every pound increase in KE results in a 0.042 m/s improvement in 10MWT, and a smaller similar increase of 0.009 m/s with every degree of ankle DF ROM. CONCLUSION GC use provides an improvement in strength and ROM but does not affect rate of change. Knee strength has a greater influence on speed of TFTs than DF ROM, although both are statistically significant predictors of speed. Results show that retaining knee strength [1, 2], along with joint flexibility, may be important factors in the ability to perform walking, climbing and supine to stand activities.
Collapse
Affiliation(s)
- Tina Duong
- Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Rehabilitation, Stanford Healthcare, Stanford, CA, USA
| | - Jennifer Canbek
- Physical Therapy Department, Nova Southeastern University, Fort Lauderdale, FL, USA
| | | | - Erik Henricson
- University of California, Davis, Department of Neurology, Sacramento, CA USA
| | - Marisa Birkmeier
- Department of Health, Human Function, and Rehabilitation Sciences, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Catherine Siener
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Carolina Tesi Rocha
- Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA
| | - Craig McDonald
- University of California, Davis, Department of Neurology, Sacramento, CA USA
| | | | | |
Collapse
|
34
|
Lilien C, Reyngoudt H, Seferian AM, Gidaro T, Annoussamy M, Chê V, Decostre V, Ledoux I, Le Louër J, Guemas E, Muntoni F, Hogrel JY, Carlier PG, Servais L. Upper limb disease evolution in exon 53 skipping eligible patients with Duchenne muscular dystrophy. Ann Clin Transl Neurol 2021; 8:1938-1950. [PMID: 34453498 PMCID: PMC8528463 DOI: 10.1002/acn3.51417] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/24/2021] [Accepted: 06/07/2021] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE To understand the natural disease upper limb progression over 3 years of ambulatory and non-ambulatory patients with Duchenne muscular dystrophy (DMD) using functional assessments and quantitative magnetic resonance imaging (MRI) and to exploratively identify prognostic factors. METHODS Forty boys with DMD (22 non-ambulatory and 18 ambulatory) with deletions in dystrophin that make them eligible for exon 53-skipping therapy were included. Clinical assessments, including Brooke score, motor function measure (MFM), hand grip and key pinch strength, and upper limb distal coordination and endurance (MoviPlate), were performed every 6 months and quantitative MRI of fat fraction (FF) and lean muscle cross sectional area (flexor and extensor muscles) were performed yearly. RESULTS In the whole population, there were strong nonlinear correlations between outcome measures. In non-ambulatory patients, annual changes over the course of 3 years were detected with high sensitivity standard response mean (|SRM| ≥0.8) for quantitative MRI-based FF, hand grip and key pinch, and MFM. Boys who presented with a FF<20% and a grip strength >27% were able to bring a glass to their mouth and retained this ability in the following 3 years. Ambulatory patients with grip strength >35% of predicted value and FF <10% retained ambulation 3 years later. INTERPRETATION We demonstrate that continuous decline in upper limb strength, function, and MRI measured muscle structure can be reliably measured in ambulatory and non-ambulatory boys with DMD with high SRM and strong correlations between outcomes. Our results suggest that a combination of grip strength and FF can be used to predict important motor milestones.
Collapse
Affiliation(s)
- Charlotte Lilien
- Institut de Myologie, Paris, France.,Department of Paediatrics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, United Kingdom
| | - Harmen Reyngoudt
- Institut de Myologie, Paris, France.,CEA/DRF/IBFJ/MIRCen, Paris, France
| | | | | | | | | | | | | | - Julien Le Louër
- Institut de Myologie, Paris, France.,CEA/DRF/IBFJ/MIRCen, Paris, France
| | | | - 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
| | | | - Pierre Georges Carlier
- Institut de Myologie, Paris, France.,Université Paris-Saclay, CEA, DRF, Service Hospitalier Frederic Joliot, Orsay, France
| | - Laurent Servais
- Institut de Myologie, Paris, France.,Department of Paediatrics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, United Kingdom.,Division of Child Neurology Reference Center for Neuromuscular Disease, Centre Hospitalier Régional de Références des Maladies Neuromusculaires, Department of Paediatrics, University Hospital Liège & University of La Citadelle, Liège, Belgium
| | | |
Collapse
|
35
|
Batra A, Lott DJ, Willcocks R, Forbes SC, Triplett W, Dastgir J, Yun P, Reghan Foley A, Bönnemann CG, Vandenborne K, Walter GA. Lower Extremity Muscle Involvement in the Intermediate and Bethlem Myopathy Forms of COL6-Related Dystrophy and Duchenne Muscular Dystrophy: A Cross-Sectional Study. J Neuromuscul Dis 2021; 7:407-417. [PMID: 32538860 DOI: 10.3233/jnd-190457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Collagen VI-related dystrophies (COL6-RDs) and Duchenne muscular dystrophy (DMD) cause progressive muscle weakness and disability. COL6-RDs are caused by mutations in the COL6 genes (COL6A1, COL6A2 and COL6A3) encoding the extracellular matrix protein collagen VI, and DMD is caused by mutations in the DMD gene encoding the cytoplasmic protein dystrophin. Both COL6-RDs and DMD are characterized by infiltration of the muscles by fatty and fibrotic tissue. This study examined the effect of disease pathology on skeletal muscles in lower extremity muscles of COL6-RDs using timed functional tests, strength measures and qualitative/ quantitative magnetic resonance imaging/spectroscopy measures (MRI/MRS) in comparison to unaffected (control) individuals. Patients with COL6-RD were also compared to age and gender matched patients with DMD.Patients with COL6-RD presented with a typical pattern of fatty infiltration of the muscle giving rise to an apparent halo effect around the muscle, while patients with DMD had evidence of fatty infiltration throughout the muscle areas imaged. Quantitatively, fat fraction, and transverse relaxation time (T2) were elevated in both COL6-RD and DMD patients compared to unaffected (control) individuals. Patients with COL6-RD had widespread muscle atrophy, likely contributing to weakness. In contrast, patients with DMD revealed force deficits even in muscle groups with increased contractile areas.
Collapse
Affiliation(s)
- Abhinandan Batra
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Donovan J Lott
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Rebecca Willcocks
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Sean C Forbes
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - William Triplett
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Jahannaz Dastgir
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Pomi Yun
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Glenn A Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
36
|
Abstract
Direct-type cavus foot deformities are most commonly encountered and are primarily sagittal plane deformities. Direct deformities should be delineated from rarer triplane pes cavovarus deformities. The lateral weight-bearing radiograph is the cornerstone of imaging evaluation of direct pes cavus foot deformity. The apex of Meary talo-first metatarsal angle on the lateral radiograph represents the pinnacle of the cavus deformity and assists in subclassification of the deformity. With routine application, ancillary radiographic imaging techniques, such as the modified Saltzman view or the modified Coleman block test, can give valuable insight into deformity assessment and surgical planning.
Collapse
Affiliation(s)
- Lawrence Osher
- Radiology, Division of Podiatric and General Medicine, Kent State University College of Podiatric Medicine, 6000 Rockside Woods Blvd. N, Independence, OH 44131, USA.
| | - Jeffrey E Shook
- Adjunct Faculty, St. Vincent Charity Medical Center, Cleveland, OH, USA
| |
Collapse
|
37
|
Lopez C, Taivassalo T, Berru MG, Saavedra A, Rasmussen HC, Batra A, Arora H, Roetzheim AM, Walter GA, Vandenborne K, Forbes SC. Postcontractile blood oxygenation level-dependent (BOLD) response in Duchenne muscular dystrophy. J Appl Physiol (1985) 2021; 131:83-94. [PMID: 34013753 PMCID: PMC8325615 DOI: 10.1152/japplphysiol.00634.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 04/28/2021] [Accepted: 05/13/2021] [Indexed: 11/22/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by a progressive replacement of muscle by fat and fibrous tissue, muscle weakness, and loss of functional abilities. Impaired vasodilatory and blood flow responses to muscle activation have also been observed in DMD and associated with mislocalization of neuronal nitric oxide synthase mu (nNOSμ) from the sarcolemma. The objective of this study was to determine whether the postcontractile blood oxygen level-dependent (BOLD) MRI response is impaired in DMD and correlated with established markers of disease severity in DMD, including MRI muscle fat fraction (FF) and clinical functional measures. Young boys with DMD (n = 16, 5-14 yr) and unaffected controls (n = 16, 5-14 yr) were evaluated using postcontractile BOLD, FF, and functional assessments. The BOLD response was measured following five brief (2 s) maximal voluntary dorsiflexion contractions, each separated by 1 min of rest. FFs from the anterior compartment lower leg muscles were quantified via chemical shift-encoded imaging. Functional abilities were assessed using the 10 m walk/run and the 6-min walk distance (6MWD). The peak BOLD responses in the tibialis anterior and extensor digitorum longus were reduced (P < 0.001) in DMD compared with controls. Furthermore, the anterior compartment peak BOLD response correlated with function (6MWD ρ = 0.87, P < 0.0001; 10 m walk/run time ρ = -0.78, P < 0.001) and FF (ρ = -0.52, P = 0.05). The reduced postcontractile BOLD response in DMD may reflect impaired microvascular function. The relationship observed between the postcontractile peak BOLD response and functional measures and FF suggests that the BOLD response is altered with disease severity in DMD.NEW & NOTEWORTHY This study examined the postcontractile blood oxygen level-dependent (BOLD) response in boys with Duchenne muscular dystrophy (DMD) and unaffected controls, and correlated this measure to markers of disease severity. Our findings indicate that the postcontractile BOLD response is impaired in DMD after brief muscle contractions, is correlated to disease severity, and may be valuable to implement in future studies to evaluate treatments targeting microvascular function in DMD.
Collapse
Affiliation(s)
- Christopher Lopez
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Tanja Taivassalo
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Maria G Berru
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Andres Saavedra
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Hannah C Rasmussen
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Abhinandan Batra
- Department of Physical Therapy, University of Florida, Gainesville, Florida
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Harneet Arora
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Alex M Roetzheim
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Glenn A Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Sean C Forbes
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| |
Collapse
|
38
|
van de Velde NM, Hooijmans MT, Sardjoe Mishre ASD, Keene KR, Koeks Z, Veeger TTJ, Alleman I, van Zwet EW, Beenakker JWM, Verschuuren JJGM, Kan HE, Niks EH. Selection Approach to Identify the Optimal Biomarker Using Quantitative Muscle MRI and Functional Assessments in Becker Muscular Dystrophy. Neurology 2021; 97:e513-e522. [PMID: 34162720 PMCID: PMC8356376 DOI: 10.1212/wnl.0000000000012233] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/29/2021] [Indexed: 12/30/2022] Open
Abstract
Objective To identify the best quantitative fat–water MRI biomarker for disease progression of leg muscles in Becker muscular dystrophy (BMD) by applying a stepwise approach based on standardized response mean (SRM) over 24 months, correlations with baseline ambulatory tests, and reproducibility. Methods Dixon fat–water imaging was performed at baseline (n = 24) and 24 months (n = 20). Fat fractions (FF) were calculated for 3 center slices and the whole muscles for 19 muscles and 6 muscle groups. Contractile cross-sectional area (cCSA) was obtained from the center slice. Functional assessments included knee extension and flexion force and 3 ambulatory tests (North Star Ambulatory Assessment [NSAA], 10-meter run, 6-minute walking test). MRI measures were selected using SRM (≥0.8) and correlation with all ambulatory tests (ρ ≤ −0.8). Measures were evaluated based on intraclass correlation coefficient (ICC) and SD of the difference. Sample sizes were calculated assuming 50% reduction in disease progression over 24 months in a clinical trial with 1:1 randomization. Results Median whole muscle FF increased between 0.2% and 2.6% without consistent cCSA changes. High SRMs and strong functional correlations were found for 8 FF but no cCSA measures. All measures showed excellent ICC (≥0.999) and similar SD of the interrater difference. Whole thigh 3 center slices FF was the best biomarker (SRM 1.04, correlations ρ ≤ −0.81, ICC 1.00, SD 0.23%, sample size 59) based on low SD and acquisition and analysis time. Conclusion In BMD, median FF of all muscles increased over 24 months. Whole thigh 3 center slices FF reduced the sample size by approximately 40% compared to NSAA.
Collapse
Affiliation(s)
- Nienke M van de Velde
- From the Department of Neurology (N.M.v.d.V., K.R.K., Z.K., J.J.G.M.V., E.H.N.), C.J. Gorter Center for High-Field MRI, Department of Radiology (M.T.H., A.S.D.S.M., K.R.K., T.T.J.V., J.-W.M.B., H.E.K.), Department of Orthopaedics, Rehabilitation and Physical Therapy (I.A.), Department of Biomedical Data Sciences (E.W.v.Z.), and Department of Ophthalmology (J.-W.M.B.), Leiden University Medical Center, the Netherlands; and Duchenne Center Netherlands (N.M.v.d.V., J.J.G.M.V., H.E.K., E.H.N.), the Netherlands
| | - Melissa T Hooijmans
- From the Department of Neurology (N.M.v.d.V., K.R.K., Z.K., J.J.G.M.V., E.H.N.), C.J. Gorter Center for High-Field MRI, Department of Radiology (M.T.H., A.S.D.S.M., K.R.K., T.T.J.V., J.-W.M.B., H.E.K.), Department of Orthopaedics, Rehabilitation and Physical Therapy (I.A.), Department of Biomedical Data Sciences (E.W.v.Z.), and Department of Ophthalmology (J.-W.M.B.), Leiden University Medical Center, the Netherlands; and Duchenne Center Netherlands (N.M.v.d.V., J.J.G.M.V., H.E.K., E.H.N.), the Netherlands
| | - Aashley S D Sardjoe Mishre
- From the Department of Neurology (N.M.v.d.V., K.R.K., Z.K., J.J.G.M.V., E.H.N.), C.J. Gorter Center for High-Field MRI, Department of Radiology (M.T.H., A.S.D.S.M., K.R.K., T.T.J.V., J.-W.M.B., H.E.K.), Department of Orthopaedics, Rehabilitation and Physical Therapy (I.A.), Department of Biomedical Data Sciences (E.W.v.Z.), and Department of Ophthalmology (J.-W.M.B.), Leiden University Medical Center, the Netherlands; and Duchenne Center Netherlands (N.M.v.d.V., J.J.G.M.V., H.E.K., E.H.N.), the Netherlands
| | - Kevin R Keene
- From the Department of Neurology (N.M.v.d.V., K.R.K., Z.K., J.J.G.M.V., E.H.N.), C.J. Gorter Center for High-Field MRI, Department of Radiology (M.T.H., A.S.D.S.M., K.R.K., T.T.J.V., J.-W.M.B., H.E.K.), Department of Orthopaedics, Rehabilitation and Physical Therapy (I.A.), Department of Biomedical Data Sciences (E.W.v.Z.), and Department of Ophthalmology (J.-W.M.B.), Leiden University Medical Center, the Netherlands; and Duchenne Center Netherlands (N.M.v.d.V., J.J.G.M.V., H.E.K., E.H.N.), the Netherlands
| | - Zaïda Koeks
- From the Department of Neurology (N.M.v.d.V., K.R.K., Z.K., J.J.G.M.V., E.H.N.), C.J. Gorter Center for High-Field MRI, Department of Radiology (M.T.H., A.S.D.S.M., K.R.K., T.T.J.V., J.-W.M.B., H.E.K.), Department of Orthopaedics, Rehabilitation and Physical Therapy (I.A.), Department of Biomedical Data Sciences (E.W.v.Z.), and Department of Ophthalmology (J.-W.M.B.), Leiden University Medical Center, the Netherlands; and Duchenne Center Netherlands (N.M.v.d.V., J.J.G.M.V., H.E.K., E.H.N.), the Netherlands
| | - Thom T J Veeger
- From the Department of Neurology (N.M.v.d.V., K.R.K., Z.K., J.J.G.M.V., E.H.N.), C.J. Gorter Center for High-Field MRI, Department of Radiology (M.T.H., A.S.D.S.M., K.R.K., T.T.J.V., J.-W.M.B., H.E.K.), Department of Orthopaedics, Rehabilitation and Physical Therapy (I.A.), Department of Biomedical Data Sciences (E.W.v.Z.), and Department of Ophthalmology (J.-W.M.B.), Leiden University Medical Center, the Netherlands; and Duchenne Center Netherlands (N.M.v.d.V., J.J.G.M.V., H.E.K., E.H.N.), the Netherlands
| | - Iris Alleman
- From the Department of Neurology (N.M.v.d.V., K.R.K., Z.K., J.J.G.M.V., E.H.N.), C.J. Gorter Center for High-Field MRI, Department of Radiology (M.T.H., A.S.D.S.M., K.R.K., T.T.J.V., J.-W.M.B., H.E.K.), Department of Orthopaedics, Rehabilitation and Physical Therapy (I.A.), Department of Biomedical Data Sciences (E.W.v.Z.), and Department of Ophthalmology (J.-W.M.B.), Leiden University Medical Center, the Netherlands; and Duchenne Center Netherlands (N.M.v.d.V., J.J.G.M.V., H.E.K., E.H.N.), the Netherlands
| | - Erik W van Zwet
- From the Department of Neurology (N.M.v.d.V., K.R.K., Z.K., J.J.G.M.V., E.H.N.), C.J. Gorter Center for High-Field MRI, Department of Radiology (M.T.H., A.S.D.S.M., K.R.K., T.T.J.V., J.-W.M.B., H.E.K.), Department of Orthopaedics, Rehabilitation and Physical Therapy (I.A.), Department of Biomedical Data Sciences (E.W.v.Z.), and Department of Ophthalmology (J.-W.M.B.), Leiden University Medical Center, the Netherlands; and Duchenne Center Netherlands (N.M.v.d.V., J.J.G.M.V., H.E.K., E.H.N.), the Netherlands
| | - Jan-Willem M Beenakker
- From the Department of Neurology (N.M.v.d.V., K.R.K., Z.K., J.J.G.M.V., E.H.N.), C.J. Gorter Center for High-Field MRI, Department of Radiology (M.T.H., A.S.D.S.M., K.R.K., T.T.J.V., J.-W.M.B., H.E.K.), Department of Orthopaedics, Rehabilitation and Physical Therapy (I.A.), Department of Biomedical Data Sciences (E.W.v.Z.), and Department of Ophthalmology (J.-W.M.B.), Leiden University Medical Center, the Netherlands; and Duchenne Center Netherlands (N.M.v.d.V., J.J.G.M.V., H.E.K., E.H.N.), the Netherlands
| | - Jan J G M Verschuuren
- From the Department of Neurology (N.M.v.d.V., K.R.K., Z.K., J.J.G.M.V., E.H.N.), C.J. Gorter Center for High-Field MRI, Department of Radiology (M.T.H., A.S.D.S.M., K.R.K., T.T.J.V., J.-W.M.B., H.E.K.), Department of Orthopaedics, Rehabilitation and Physical Therapy (I.A.), Department of Biomedical Data Sciences (E.W.v.Z.), and Department of Ophthalmology (J.-W.M.B.), Leiden University Medical Center, the Netherlands; and Duchenne Center Netherlands (N.M.v.d.V., J.J.G.M.V., H.E.K., E.H.N.), the Netherlands
| | - Hermien E Kan
- From the Department of Neurology (N.M.v.d.V., K.R.K., Z.K., J.J.G.M.V., E.H.N.), C.J. Gorter Center for High-Field MRI, Department of Radiology (M.T.H., A.S.D.S.M., K.R.K., T.T.J.V., J.-W.M.B., H.E.K.), Department of Orthopaedics, Rehabilitation and Physical Therapy (I.A.), Department of Biomedical Data Sciences (E.W.v.Z.), and Department of Ophthalmology (J.-W.M.B.), Leiden University Medical Center, the Netherlands; and Duchenne Center Netherlands (N.M.v.d.V., J.J.G.M.V., H.E.K., E.H.N.), the Netherlands
| | - Erik H Niks
- From the Department of Neurology (N.M.v.d.V., K.R.K., Z.K., J.J.G.M.V., E.H.N.), C.J. Gorter Center for High-Field MRI, Department of Radiology (M.T.H., A.S.D.S.M., K.R.K., T.T.J.V., J.-W.M.B., H.E.K.), Department of Orthopaedics, Rehabilitation and Physical Therapy (I.A.), Department of Biomedical Data Sciences (E.W.v.Z.), and Department of Ophthalmology (J.-W.M.B.), Leiden University Medical Center, the Netherlands; and Duchenne Center Netherlands (N.M.v.d.V., J.J.G.M.V., H.E.K., E.H.N.), the Netherlands.
| |
Collapse
|
39
|
Murphy AP, Greally E, O'Hogain D, Blamire A, Caravan P, Straub V. Use of EP3533-Enhanced Magnetic Resonance Imaging as a Measure of Disease Progression in Skeletal Muscle of mdx Mice. Front Neurol 2021; 12:636719. [PMID: 34220666 PMCID: PMC8248789 DOI: 10.3389/fneur.2021.636719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 05/12/2021] [Indexed: 11/13/2022] Open
Abstract
As putative treatments are developed for Duchenne muscular dystrophy (DMD), sensitive, non-invasive measures are increasingly important to quantify disease progression. Fibrosis is one of the histological hallmarks of muscular dystrophy and has been directly linked to prognosis. EP3533 is a novel contrast agent with an affinity to collagen 1 that has demonstrated a significant and high correlation to ex vivo fibrosis quantification. Halofuginone is an established anti-fibrotic compound shown to reduce collagen skeletal muscle fibrosis in murine models of DMD. This experiment explored whether EP3533 could be used to detect signal change in skeletal muscle of mdx mice before and after a 12 week course of halofuginone compared to controls. Four age-matched groups of treated and untreated mice were evaluated: 2 groups of mdx (n = 8 and n = 13, respectively), and 2 groups of BL10 mice (n = 5 and n = 3, respectively). Treated mice received an intraperitoneal injection with halofuginone three times per week for 12 weeks, with the remaining mice being given vehicle. Both mdx groups and the untreated BL10 were scanned at baseline, then all groups were scanned on week 13. All subjects were scanned using a 7T Varian scanner before and after administration of EP3533 using a T1 mapping technique. Mice underwent grip testing in week 13 prior to dissection. Skeletal muscle was used for Masson's trichrome quantification, hydroxyproline assay, and immunofluorescent antibody staining. Untreated mdx mice demonstrated a significant increase in R1 signal from pre- to post-treatment scan in three out of four muscles (gastrocnemius p = 0.04, hamstrings p = 0.009, and tibialis anterior p = 0.01), which was not seen in either the treated mdx or the BL10 groups. Histological quantification of fibrosis also demonstrated significantly higher levels in the untreated mdx mice with significant correlation seen between histology and EP3533 signal change. Forelimb weight adjusted-grip strength was significantly lower in the untreated mdx group, compared to the treated group. EP3533 can be used over time as an outcome measure to quantify treatment effect of an established anti-fibrotic drug. Further studies are needed to evaluate the use of this contrast agent in humans.
Collapse
Affiliation(s)
- Alexander Peter Murphy
- The Institute of Cancer and Genomics, Birmingham University, Birmingham, United Kingdom.,The John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Elizabeth Greally
- The John Walton Muscular Dystrophy Research Centre, Institute of Translational and Clinical Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Dara O'Hogain
- Newcastle Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrew Blamire
- Newcastle Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Peter Caravan
- Department of Radiology, Martinos Center for Biomedical Imaging, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| | - Volker Straub
- The Institute of Cancer and Genomics, Birmingham University, Birmingham, United Kingdom.,Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
40
|
Waisman A, Norris AM, Elías Costa M, Kopinke D. Automatic and unbiased segmentation and quantification of myofibers in skeletal muscle. Sci Rep 2021; 11:11793. [PMID: 34083673 PMCID: PMC8175575 DOI: 10.1038/s41598-021-91191-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/24/2021] [Indexed: 12/05/2022] Open
Abstract
Skeletal muscle has the remarkable ability to regenerate. However, with age and disease muscle strength and function decline. Myofiber size, which is affected by injury and disease, is a critical measurement to assess muscle health. Here, we test and apply Cellpose, a recently developed deep learning algorithm, to automatically segment myofibers within murine skeletal muscle. We first show that tissue fixation is necessary to preserve cellular structures such as primary cilia, small cellular antennae, and adipocyte lipid droplets. However, fixation generates heterogeneous myofiber labeling, which impedes intensity-based segmentation. We demonstrate that Cellpose efficiently delineates thousands of individual myofibers outlined by a variety of markers, even within fixed tissue with highly uneven myofiber staining. We created a novel ImageJ plugin (LabelsToRois) that allows processing of multiple Cellpose segmentation images in batch. The plugin also contains a semi-automatic erosion function to correct for the area bias introduced by the different stainings, thereby identifying myofibers as accurately as human experts. We successfully applied our segmentation pipeline to uncover myofiber regeneration differences between two different muscle injury models, cardiotoxin and glycerol. Thus, Cellpose combined with LabelsToRois allows for fast, unbiased, and reproducible myofiber quantification for a variety of staining and fixation conditions.
Collapse
Affiliation(s)
- Ariel Waisman
- CONICET - Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Laboratorio de Investigación Aplicada a Neurociencias (LIAN), Buenos Aires, Argentina.
| | - Alessandra Marie Norris
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, 32610, FL, USA.,Myology Institute, University of Florida College of Medicine, Gainesville, FL, USA
| | | | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, 32610, FL, USA. .,Myology Institute, University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
41
|
Musculoskeletal magnetic resonance imaging in the DE50-MD dog model of Duchenne muscular dystrophy. Neuromuscul Disord 2021; 31:736-751. [PMID: 34384671 PMCID: PMC8449064 DOI: 10.1016/j.nmd.2021.05.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 11/23/2022]
Abstract
Normalized muscle volumes distinguish between wild type and DE50-MD dogs. Global muscle T2 signal intensities discriminate between wild type and DE50-MD dogs. Musculoskeletal changes detected by MRI reflect those seen in human DMD patients. Musculoskeletal MRI in this model will be useful to assess treatment efficacy.
The DE50-MD canine model of Duchenne muscular dystrophy (DMD) has a dystrophin gene splice site mutation causing deletion of exon 50, an out-of-frame transcript and absence of dystrophin expression in striated muscles. We hypothesized that the musculoskeletal phenotype of DE50-MD dogs could be detected using Magnetic Resonance Imaging (MRI), that it would progress with age and that it would reflect those in other canine models and DMD patients. 15 DE50-MD and 10 age-matched littermate wild type (WT) male dogs underwent MRI every 3 months from 3 to 18 months of age. Normalized muscle volumes, global muscle T2 and ratio of post- to pre-gadolinium T1-weighted SI were evaluated in 7 pelvic limb and 4 lumbar muscles bilaterally. DE50-MD dogs, compared to WT, had smaller volumes in all muscles, except the cranial sartorius; global muscle T2 was significantly higher in DE50-MD dogs compared to WT. Muscle volumes plateaued and global muscle T2 decreased with age. Normalized muscle volumes and global muscle T2 revealed significant differences between groups longitudinally and should be useful to determine efficacy of therapeutics in this model with suitable power and low sample sizes. Musculoskeletal changes reflect those of DMD patients and other dog models.
Collapse
|
42
|
Sherlock SP, Zhang Y, Binks M, Marraffino S. Quantitative muscle MRI biomarkers in Duchenne muscular dystrophy: cross-sectional correlations with age and functional tests. Biomark Med 2021; 15:761-773. [PMID: 34155911 PMCID: PMC8253163 DOI: 10.2217/bmm-2020-0801] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/23/2021] [Indexed: 01/07/2023] Open
Abstract
Aim: Using baseline data from a clinical trial of domagrozumab in Duchenne muscular dystrophy, we evaluated the correlation between functional measures and quantitative MRI assessments of thigh muscle. Patients & methods: Analysis included timed functional tests, knee extension/strength and North Star Ambulatory Assessment. Patients (n = 120) underwent examinations of one thigh, with MRI sequences to enable measurements of muscle volume (MV), MV index, mean T2 relaxation time via T2-mapping and fat fraction. Results: MV was moderately correlated with strength assessments. MV index, fat fraction and T2-mapping measures had moderate correlations (r ∼ 0.5) to all functional tests, North Star Ambulatory Assessment and age. Conclusion: The moderate correlation between functional tests, age and baseline MRI measures supports MRI as a biomarker in Duchenne muscular dystrophy clinical trials. Trial registration: ClinicalTrials.gov, NCT02310763; registered 4 November 2014.
Collapse
Affiliation(s)
| | - Yao Zhang
- Pfizer Inc, Cambridge, MA 02139, USA
| | | | | |
Collapse
|
43
|
Greve T, Burian E, Zoffl A, Feuerriegel G, Schlaeger S, Dieckmeyer M, Sollmann N, Klupp E, Weidlich D, Inhuber S, Löffler M, Montagnese F, Deschauer M, Schoser B, Bublitz S, Zimmer C, Karampinos DC, Kirschke JS, Baum T. Regional variation of thigh muscle fat infiltration in patients with neuromuscular diseases compared to healthy controls. Quant Imaging Med Surg 2021; 11:2610-2621. [PMID: 34079727 DOI: 10.21037/qims-20-1098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Chemical shift encoding-based water-fat magnetic resonance imaging (CSE-MRI) measures a quantitative biomarker: the proton density fat fraction (PDFF). The aim was to assess regional and proximo-distal PDFF variations at the thigh in patients with myotonic dystrophy type 2 (DM2), limb-girdle muscular dystrophy type 2A (LGMD2A), and late-onset Pompe disease (LOPD) as compared to healthy controls. Methods Seven patients (n=2 DM2, n=2 LGMD2A, n=3 LOPD) and 20 controls were recruited. A 3D-spoiled gradient echo sequence was used to scan the thigh musculature. Muscles were manually segmented to generate mean muscle PDFF. Results In all three disease entities, there was an increase in muscle fat replacement compared to healthy controls. However, within each disease group, there were patients with a shorter time since symptom onset that only showed mild PDFF elevation (range, 10% to 20%) compared to controls (P≤0.05), whereas patients with a longer period since symptom onset showed a more severe grade of fat replacement with a range of 50% to 70% (P<0.01). Increased PDFF of around 5% was observed for vastus medialis, semimembranosus and gracilis muscles in advanced compared to early DM2. LGMD2A_1 showed an early disease stage with predominantly mild PDFF elevations over all muscles and levels (10.9%±7.1%) compared to controls. The quadriceps, gracilis and biceps femoris muscles showed the highest difference between LGMD2A_1 with 5 years since symptom onset (average PDFF 11.1%±6.9%) compared to LGMD2A_2 with 32 years since symptom onset (average PDFF 66.3%±6.3%). For LOPD patients, overall PDFF elevations were observed in all major hip flexors and extensors (range, 25.8% to 30.8%) compared to controls (range, 1.7% to 2.3%, P<0.05). Proximal-to-distal PDFF highly varied within and between diseases and within controls. The intra-reader reliability was high (reproducibility coefficient ≤2.19%). Conclusions By quantitatively measuring muscle fat infiltration at the thigh, we identified candidate muscles for disease monitoring due to their gradual PDFF elevation with longer disease duration. Regional variation between proximal, central, and distal muscle PDFF was high and is important to consider when performing longitudinal MRI follow-ups in the clinical setting or in longitudinal studies.
Collapse
Affiliation(s)
- Tobias Greve
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| | - Egon Burian
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Agnes Zoffl
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Georg Feuerriegel
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Sarah Schlaeger
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Michael Dieckmeyer
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Nico Sollmann
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Elisabeth Klupp
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Dominik Weidlich
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stephanie Inhuber
- Department of Sports and Health Sciences, Technical University of Munich, Munich, Germany
| | - Maximilian Löffler
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Federica Montagnese
- Friedrich Baur Institute at the Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | - Marcus Deschauer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Benedikt Schoser
- Friedrich Baur Institute at the Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | - Sarah Bublitz
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Claus Zimmer
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Dimitrios C Karampinos
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jan S Kirschke
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Thomas Baum
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
44
|
The increasing role of muscle MRI to monitor changes over time in untreated and treated muscle diseases. Curr Opin Neurol 2021; 33:611-620. [PMID: 32796278 DOI: 10.1097/wco.0000000000000851] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW This review aims to discuss the recent results of studies published applying quantitative MRI sequences to large cohorts of patients with neuromuscular diseases. RECENT FINDINGS Quantitative MRI sequences are now available to identify and quantify changes in muscle water and fat content. These two components have been associated with acute and chronic injuries, respectively. Studies show that the increase in muscle water is not only reversible if therapies are applied successfully but can also predict fat replacement in neurodegenerative diseases. Muscle fat fraction correlates with muscle function tests and increases gradually over time in parallel with the functional decline of patients with neuromuscular diseases. There are new spectrometry-based sequences to quantify other components, such as glycogen, electrolytes or the pH of the muscle fibre, extending the applicability of MRI to the study of several processes in neuromuscular diseases. SUMMARY The latest results obtained from the study of long cohorts of patients with various neuromuscular diseases open the door to the use of this technology in clinical trials, which would make it possible to obtain a new measure for assessing the effectiveness of new treatments. The challenge is currently the popularization of these studies and their application to the monitoring of patients in the daily clinic.
Collapse
|
45
|
Clark BC, Rutkove S, Lupton EC, Padilla CJ, Arnold WD. Potential Utility of Electrical Impedance Myography in Evaluating Age-Related Skeletal Muscle Function Deficits. Front Physiol 2021; 12:666964. [PMID: 34025454 PMCID: PMC8138591 DOI: 10.3389/fphys.2021.666964] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/13/2021] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle function deficits associated with advancing age are due to several physiological and morphological changes including loss of muscle size and quality (conceptualized as a reduction in the intrinsic force-generating capacity of a muscle when adjusted for muscle size). Several factors can contribute to loss of muscle quality, including denervation, excitation-contraction uncoupling, increased fibrosis, and myosteatosis (excessive levels of inter- and intramuscular adipose tissue and intramyocellular lipids). These factors also adversely affect metabolic function. There is a major unmet need for tools to rapidly and easily assess muscle mass and quality in clinical settings with minimal patient and provider burden. Herein, we discuss the potential for electrical impedance myography (EIM) as a tool to evaluate muscle mass and quality in older adults. EIM applies weak, non-detectible (e.g., 400 μA), mutifrequency (e.g., 1 kHz–1 MHz) electrical currents to a muscle (or muscle group) through two excitation electrodes, and resulting voltages are measured via two sense electrodes. Measurements are fast (~5 s/muscle), simple to perform, and unaffected by factors such as hydration that may affect other simple measures of muscle status. After nearly 2 decades of study, EIM has been shown to reflect muscle health status, including the presence of atrophy, fibrosis, and fatty infiltration, in a variety of conditions (e.g., developmental growth and maturation, conditioning/deconditioning, and obesity) and neuromuscular diseases states [e.g., amyotrophic lateral sclerosis (ALS) and muscular dystrophies]. In this article, we describe prior work and current evidence of EIM’s potential utility as a measure of muscle health in aging and geriatric medicine.
Collapse
Affiliation(s)
- Brian C Clark
- Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, Athens, OH, United States.,Department of Biomedical Sciences, Ohio University, Athens, OH, United States.,Division of Geriatric Medicine, Ohio University, Athens, OH, United States
| | - Seward Rutkove
- Department of Neurology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | | | - Carlos J Padilla
- Department of Neurology, Ohio State University, Columbus, OH, United States
| | - W David Arnold
- Department of Neurology, Ohio State University, Columbus, OH, United States
| |
Collapse
|
46
|
Finkel RS, Finanger E, Vandenborne K, Sweeney HL, Tennekoon G, Shieh PB, Willcocks R, Walter G, Rooney WD, Forbes SC, Triplett WT, Yum SW, Mancini M, MacDougall J, Fretzen A, Bista P, Nichols A, Donovan JM. Disease-modifying effects of edasalonexent, an NF-κB inhibitor, in young boys with Duchenne muscular dystrophy: Results of the MoveDMD phase 2 and open label extension trial. Neuromuscul Disord 2021; 31:385-396. [PMID: 33678513 DOI: 10.1016/j.nmd.2021.02.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/12/2020] [Accepted: 02/01/2021] [Indexed: 12/18/2022]
Abstract
Chronic activation of NF-κB is a key driver of muscle degeneration and suppression of muscle regeneration in Duchenne muscular dystrophy. Edasalonexent (CAT-1004) is an orally-administered novel small molecule that covalently links two bioactive compounds (salicylic acid and docosahexaenoic acid) that inhibit NF-κB. This placebo-controlled, proof-of-concept phase 2 study with open-label extension in boys ≥4-<8 years old with any dystrophin mutation examined the effect of edasalonexent (67 or 100 mg/kg/day) compared to placebo or off-treatment control. Endpoints were safety/tolerability, change from baseline in MRI T2 relaxation time of lower leg muscles and functional assessment, as well as pharmacodynamics and biomarkers. Treatment was well-tolerated and the majority of adverse events were mild, and most commonly of the gastrointestinal system (primarily diarrhea). There were no serious adverse events in the edasalonexent groups. Edasalonexent 100 mg/kg was associated with slowing of disease progression and preservation of muscle function compared to an off-treatment control period, with decrease in levels of NF-κB-regulated genes and improvements in biomarkers of muscle health and inflammation. These results support investigating edasalonexent in future trials and have informed the design of the edasalonexent phase 3 clinical trial in boys with Duchenne.
Collapse
Affiliation(s)
- Richard S Finkel
- St. Jude Children's Research Hospital, Memphis, TN and Nemours Children's Hospital, Orlando, FL, United States.
| | - Erika Finanger
- Oregon Health & Science University, Portland, OR, United States
| | | | - H Lee Sweeney
- University of Florida Health, Gainesville, FL, United States
| | - Gihan Tennekoon
- The Children's Hospital of Philadelphia, and the University of Pennsylvania, Philadelphia, PA, United States
| | - Perry B Shieh
- University of California, Los Angeles, Los Angeles, CA, United States
| | | | - Glenn Walter
- University of Florida Health, Gainesville, FL, United States
| | | | - Sean C Forbes
- University of Florida Health, Gainesville, FL, United States
| | | | - Sabrina W Yum
- The Children's Hospital of Philadelphia, and the University of Pennsylvania, Philadelphia, PA, United States
| | - Maria Mancini
- Catabasis Pharmaceuticals, Inc., Boston, MA, United States
| | | | | | - Pradeep Bista
- Catabasis Pharmaceuticals, Inc., Boston, MA, United States
| | - Andrew Nichols
- Catabasis Pharmaceuticals, Inc., Boston, MA, United States
| | | |
Collapse
|
47
|
Otto LA, Froeling M, van Eijk RP, Asselman F, Wadman R, Cuppen I, Hendrikse J, van der Pol W. Quantification of disease progression in spinal muscular atrophy with muscle MRI-a pilot study. NMR IN BIOMEDICINE 2021; 34:e4473. [PMID: 33480130 PMCID: PMC7988555 DOI: 10.1002/nbm.4473] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/30/2020] [Indexed: 05/02/2023]
Abstract
OBJECTIVES Quantitative MRI (qMRI) of muscles is a promising tool to measure disease progression or to assess therapeutic effects in neuromuscular diseases. Longitudinal imaging studies are needed to show sensitivity of qMRI in detecting disease progression in spinal muscular atrophy (SMA). In this pilot study we therefore studied one-year changes in quantitative MR parameters in relation to clinical scores. METHODS We repeated quantitative 3 T MR analysis of thigh muscles and clinical testing one year after baseline in 10 treatment-naïve patients with SMA, 5 with Type 2 (21.6 ± 7.0 years) and 5 with Type 3 (33.4 ± 11.9 years). MR protocol consisted of Dixon, T2 mapping and diffusion tensor imaging (DTI). The temporal relation of parameters was examined with a mixed model. RESULTS We detected a significant increase in fat fraction (baseline, 38.2% SE 0.6; follow-up, 39.5% SE 0.6; +1.3%, p = 0.001) in all muscles. Muscles with moderate to high fat infiltration at baseline show a larger increase over time (+1.6%, p < 0.001). We did not find any changes in DTI parameters except for low fat-infiltration muscles (m. adductor longus and m. biceps femoris (short head)). The T2 of muscles decreased from 28.2 ms to 28.0 ms (p = 0.07). Muscle strength and motor function scores were not significantly different between follow-up and baseline. CONCLUSION Longitudinal imaging data show slow disease progression in skeletal muscle of the thigh of (young-) adult patients with SMA despite stable strength and motor function scores. Quantitative muscle imaging demonstrates potential as a biomarker for disease activity and monitoring of therapy response.
Collapse
Affiliation(s)
- Louise A.M. Otto
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Martijn Froeling
- Department of RadiologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Ruben P.A. van Eijk
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
- Biostatistics & Research Support, Julius Center for Health Sciences and Primary CareUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Fay‐Lynn Asselman
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Renske Wadman
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Inge Cuppen
- Department of Neurology and Child Neurology, UMC Utrecht Brain CenterUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Jeroen Hendrikse
- Department of RadiologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - W‐Ludo van der Pol
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
48
|
Lawal TA, Patankar A, Todd JJ, Razaqyar MS, Chrismer IC, Zhang X, Waite MR, Jain MS, Emile-Backer M, Witherspoon JW, Liu CY, Grunseich C, Meilleur KG. Ryanodine Receptor 1-Related Myopathies: Quantification of Intramuscular Fatty Infiltration from T1-Weighted MRI. J Neuromuscul Dis 2021; 8:657-668. [PMID: 33646171 PMCID: PMC8385519 DOI: 10.3233/jnd-200549] [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] [Indexed: 01/14/2023]
Abstract
Background: Ryanodine receptor 1-related myopathy (RYR1-RM) can present with a selective pattern and gradient of intramuscular fatty infiltration (IMFI) on magnetic resonance imaging (MRI). Objective: To demonstrate an automated protocol for quantification of IMFI in the lower extremity muscles of individuals with RYR1-RM using T1-weighted MRI and to examine the relationships of IMFI with motor function and clinical severity. Methods: Axial images of the lower extremity muscles were acquired by T1-weighted fast spin-echo and short tau inversion recovery (STIR) sequences. A modified ImageJ-based program was used for quantification. IMFI data was analyzed by mode of inheritance, motor function, and clinical severity. Results: Upper and lower leg IMFI from 36 genetically confirmed and ambulatory RYR1-RM affected individuals (26 dominant and 10 recessive) were analyzed using Grey-scale quantification. There was no statistically significant difference in IMFI between dominant and recessive cases in upper or lower legs. IMFI in both upper and lower legs was inversely correlated with participant performance on the motor function measure (MFM-32) total score (upper leg: p < 0.001; lower leg: p = 0.003) and the six-minute walk test (6MWT) distance (upper leg: p < 0.001; lower leg: p = 0.010). There was no significant difference in mean IMFI between participants with mild versus severe clinical phenotypes (p = 0.257). Conclusion: A modified ImageJ-based algorithm was able to select and quantify fatty infiltration in a cohort of heterogeneously affected individuals with RYR1-RM. IMFI was not predictive of mode of inheritance but showed strong correlation with motor function and capacity tests including MFM-32 and 6MWT, respectively.
Collapse
Affiliation(s)
- Tokunbor A Lawal
- Tissue Injury Branch, National Institute of Nursing Research (NIH), Bethesda, MD, USA
| | - Aneesh Patankar
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke (NIH), Bethesda, MD, USA
| | - Joshua J Todd
- Tissue Injury Branch, National Institute of Nursing Research (NIH), Bethesda, MD, USA
| | - Muslima S Razaqyar
- Tissue Injury Branch, National Institute of Nursing Research (NIH), Bethesda, MD, USA
| | - Irene C Chrismer
- Tissue Injury Branch, National Institute of Nursing Research (NIH), Bethesda, MD, USA
| | - Xuemin Zhang
- Tissue Injury Branch, National Institute of Nursing Research (NIH), Bethesda, MD, USA
| | - Melissa R Waite
- Mark O. Hatfield Clinical Research Center, NIH, Bethesda, MD, USA
| | - Minal S Jain
- Mark O. Hatfield Clinical Research Center, NIH, Bethesda, MD, USA
| | - Magalie Emile-Backer
- Tissue Injury Branch, National Institute of Nursing Research (NIH), Bethesda, MD, USA
| | - Jessica W Witherspoon
- Tissue Injury Branch, National Institute of Nursing Research (NIH), Bethesda, MD, USA
| | - Chia-Ying Liu
- Division of Cardiology, Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Christopher Grunseich
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke (NIH), Bethesda, MD, USA
| | - Katherine G Meilleur
- Tissue Injury Branch, National Institute of Nursing Research (NIH), Bethesda, MD, USA
| |
Collapse
|
49
|
Alic L, Griffin JF, Eresen A, Kornegay JN, Ji JX. Using MRI to quantify skeletal muscle pathology in Duchenne muscular dystrophy: A systematic mapping review. Muscle Nerve 2021; 64:8-22. [PMID: 33269474 PMCID: PMC8247996 DOI: 10.1002/mus.27133] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022]
Abstract
There is a great demand for accurate non‐invasive measures to better define the natural history of disease progression or treatment outcome in Duchenne muscular dystrophy (DMD) and to facilitate the inclusion of a large range of participants in DMD clinical trials. This review aims to investigate which MRI sequences and analysis methods have been used and to identify future needs. Medline, Embase, Scopus, Web of Science, Inspec, and Compendex databases were searched up to 2 November 2019, using keywords “magnetic resonance imaging” and “Duchenne muscular dystrophy.” The review showed the trend of using T1w and T2w MRI images for semi‐qualitative inspection of structural alterations of DMD muscle using a diversity of grading scales, with increasing use of T2map, Dixon, and MR spectroscopy (MRS). High‐field (>3T) MRI dominated the studies with animal models. The quantitative MRI techniques have allowed a more precise estimation of local or generalized disease severity. Longitudinal studies assessing the effect of an intervention have also become more prominent, in both clinical and animal model subjects. Quality assessment of the included longitudinal studies was performed using the Newcastle‐Ottawa Quality Assessment Scale adapted to comprise bias in selection, comparability, exposure, and outcome. Additional large clinical trials are needed to consolidate research using MRI as a biomarker in DMD and to validate findings against established gold standards. This future work should use a multiparametric and quantitative MRI acquisition protocol, assess the repeatability of measurements, and correlate findings to histologic parameters.
Collapse
Affiliation(s)
- Lejla Alic
- Department of Electrical & Computer Engineering, Texas A&M University, Doha, Qatar.,Magnetic Detection and Imaging group, Technical Medical Centre, University of Twente, The Netherlands
| | - John F Griffin
- College of Vet. Med. & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Aydin Eresen
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Department of Electrical & Computer Engineering, Texas A&M University, College Station, Texas, USA
| | - Joe N Kornegay
- College of Vet. Med. & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Jim X Ji
- Department of Electrical & Computer Engineering, Texas A&M University, Doha, Qatar.,Department of Electrical & Computer Engineering, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
50
|
Willcocks RJ, Forbes SC, Walter GA, Sweeney L, Rodino-Klapac LR, Mendell JR, Vandenborne K. Assessment of rAAVrh.74.MHCK7.micro-dystrophin Gene Therapy Using Magnetic Resonance Imaging in Children With Duchenne Muscular Dystrophy. JAMA Netw Open 2021; 4:e2031851. [PMID: 33394000 PMCID: PMC7783546 DOI: 10.1001/jamanetworkopen.2020.31851] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
This case-control study uses magnetic resonance imaging and spectroscopy to evaluate the association between treatment with recombinant adeno-associated virus serotype rh74 (rAAVrh74) and muscle quality in children with Duchenne muscular dystrophy.
Collapse
Affiliation(s)
| | - Sean C. Forbes
- Department of Physical Therapy, University of Florida, Gainesville
| | - Glenn A. Walter
- Department of Physical Therapy, University of Florida, Gainesville
| | - Lee Sweeney
- Department of Physical Therapy, University of Florida, Gainesville
| | | | - Jerry R. Mendell
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | | |
Collapse
|